18.06.2014 Views

Abstract Book - PDF - San Antonio Breast Cancer Symposium

Abstract Book - PDF - San Antonio Breast Cancer Symposium

Abstract Book - PDF - San Antonio Breast Cancer Symposium

SHOW MORE
SHOW LESS

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

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

<strong>Cancer</strong><br />

Research<br />

December 15, 2012 • Volume 72 • Number 24 • Supplement to <strong>Cancer</strong> Research • Pages 1s–608s<br />

Driving Innovation to<br />

Prevent and Cure <strong>Cancer</strong><br />

www.aacrjournals.org


Publishing<br />

Information<br />

<strong>Cancer</strong><br />

Research<br />

Print ISSN: 0008-5472 • Online ISSN: 1538-7445<br />

AACR Officers<br />

Frank McCormick, President<br />

Charles L. Sawyers, President-Elect<br />

Judy E. Garber, Past President<br />

William H. Hait, Treasurer<br />

Margaret Foti, Chief Executive Officer<br />

Publications Committee<br />

Michael E. Berens<br />

Gideon M. Bollag<br />

Michael A. Caligiuri, Chairperson<br />

Richard M. Caprioli<br />

Arul M. Chinnaiyan<br />

Joseph F. Costello<br />

Chi Van Dang<br />

Jessica Clague DeHart<br />

Lisa Diller<br />

Levi A. Garraway<br />

John D. Groopman<br />

Ernest T. Hawk<br />

<strong>San</strong>dra J. Horning<br />

Nancy E. Hynes<br />

Pasi A. Jänne<br />

Emma M. Lees<br />

Guillermina Lozano<br />

Richard M. Marais<br />

William G. Nelson<br />

David R. Parkinson<br />

David Piwnica-Worms<br />

Robert H. Vonderheide<br />

Publishing Staff<br />

Publisher<br />

Diane Scott-Lichter<br />

Director, Editorial<br />

Helen Barsky Atkins<br />

Asst. Director, Editorial Systems<br />

& Managing Editor<br />

Kelly A. Hadsell<br />

Senior Associate Editor<br />

Arthur M. Buchberg<br />

Editorial Staff<br />

Crystal Cheepudom<br />

Amir Kalili<br />

Naima D. Stone<br />

Timothy Rinehart<br />

Patricia Russo<br />

Associate Director, Production<br />

Charlene Squibb<br />

Production Staff<br />

Brenda Roberson<br />

Jeremy Rosenberg<br />

Sue Russell<br />

Electronic Publishing Staff<br />

Shira Carroll<br />

Mary Beth Cunney<br />

Diane M. Marinelli<br />

Associate Director,<br />

Sales and Marketing<br />

Nicola L. Hill<br />

Assistant Director,<br />

Circulation and Fulfillment<br />

Karola Rac<br />

Marketing and<br />

Creative Services<br />

Director<br />

Paul Driscoll<br />

Design Staff<br />

Susan Moore<br />

Publishing Information: <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Co-Directors<br />

Carlos L. Arteaga<br />

C. Kent Osborne<br />

Peter M. Ravdin<br />

Executive Committee<br />

Carlos L. Arteaga<br />

Gary C. Chamness<br />

Margaret Foti<br />

Susan G. Hilsenbeck<br />

Ismail Jatoi<br />

C. Kent Osborne<br />

Peter M. Ravdin<br />

Andrea Richardson<br />

Ian M. Thompson, Jr.<br />

Symposia Director<br />

Rich Markow<br />

Meeting Profile<br />

For 35 years, the mission of the <strong>San</strong><br />

<strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong> has<br />

been to provide state-of-the-art information<br />

on breast cancer research. The<br />

symposium aims to achieve a balance<br />

of clinical, translational, and basic<br />

research, providing a forum for interaction,<br />

communication, and education<br />

<strong>Cancer</strong> Research [ISSN 0008-5472;<br />

CNREA8] is published twice a month,<br />

one volume per year, by the American<br />

Association for <strong>Cancer</strong> Research, Inc.<br />

Postage paid at Philadelphia, PA, and<br />

additional mailing offices. POSTMAS-<br />

TER: Send address changes to Member<br />

Services, AACR, 615 Chestnut Street,<br />

17th Floor, Philadelphia, PA 19106-<br />

4404 or E-mail: membership@aacr.<br />

org. Copyright 2012 by the American<br />

Association for <strong>Cancer</strong> Research,<br />

Inc. Printed on acid free paper in the<br />

United States of America.<br />

Subscription Information<br />

AACR Members. Subscription options<br />

are print with online access or see<br />

http://www.aacr.org and go to the membership<br />

page for further information.<br />

Institutions and Nonmembers.<br />

E-mail contacts listed below are for<br />

single-site online access only (limited<br />

to library) and optional print subscriptions.<br />

USA, Canada, and Mexico:<br />

turpinna@turpin-distribution.com<br />

Japan:<br />

e-support@maruzen.co.jp; info@<br />

jp.swets.com; japan@ebsco.com;<br />

journal@kinokuniya.co.jp; or usaco@<br />

usaco.co.jp<br />

Rest of World:<br />

custserv@turpin-distribution.com<br />

Site Licenses. Send site license (online<br />

access open to multiple buildings, locations,<br />

remote and proxy) inquiries to<br />

sitelicense@aacr.org.<br />

for a broad spectrum of researchers,<br />

health professionals, and those with a<br />

special interest in breast cancer.<br />

In 2007, the <strong>Cancer</strong> Therapy & Research<br />

Center (CTRC) at UT Health<br />

Science Center <strong>San</strong> <strong>Antonio</strong> and the<br />

American Association for <strong>Cancer</strong><br />

Research (AACR) announced a collaboration<br />

for the future of the <strong>San</strong><br />

<strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong>.<br />

The symposium has been renamed<br />

the CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong><br />

<strong>Cancer</strong> <strong>Symposium</strong>. Complementing<br />

the clinical strengths of the highly<br />

regarded annual <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong><br />

<strong>Cancer</strong> <strong>Symposium</strong>, the AACR’s scientific<br />

prestige in basic, translational,<br />

and clinical cancer research will<br />

create a unique and comprehensive<br />

scientific meeting that will advance<br />

breast cancer research for the benefit<br />

of patients.<br />

In 2005, Baylor College of Medicine became<br />

a joint sponsor of the symposium<br />

and will remain a part the CTRC-AACR<br />

collaboration.<br />

Publishing Information: <strong>Cancer</strong> Research<br />

Change of Address. Send change<br />

of address notifications 60 days in<br />

advance and include both old and<br />

new addresses. Member subscribers<br />

should contact AACR Member<br />

Services, 615 Chestnut St., 17th Floor,<br />

Philadelphia, PA 19106-4404; e-mail<br />

membership@aacr.org. Nonmember<br />

subscribers contact Turpin Distribution,<br />

The Bleachery, 143 West<br />

Street, New Milford, CT 06776; e-mail<br />

turpinna@turpin-distribution.com,<br />

or Turpin Distribution, Pegasus Drive,<br />

Stratton Business Park, Biggleswade,<br />

Bedfordshire SG18 8TQ, United<br />

Kingdom; e-mail custserv@turpindistribution.com.<br />

Claims. Claims for missing issues<br />

made within 90 days of the issue’s<br />

publication date will be honored while<br />

supplies last. Member subscribers<br />

should contact membership@aacr.org,<br />

phone 215-440-9300, fax 215-440-9412.<br />

Institutions and nonmember subscribers<br />

should direct claims to Turpin<br />

Distribution at the appropriate address<br />

above or by e-mail turpinna@turpindistribution.com.<br />

Single Issue Sales. Single issues<br />

and back stock of the journal may be<br />

ordered from the AACR main office<br />

by calling 866-423-3965 (toll-free) or<br />

215-440-9300. Prices are available upon<br />

request.<br />

Copyright and Permissions<br />

As a not-for-profit organization<br />

incorporated in the United States,<br />

AACR adheres to U.S. copyright law<br />

(PL 94-553), which became effective<br />

Future Meeting Dates<br />

December 10–14, 2013<br />

December 9–13, 2014<br />

December 8–12, 2015<br />

Copyright and Permissions<br />

As a collaborating partner, the AACR<br />

is publishing the abstracts of the<br />

<strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

in this Supplement to <strong>Cancer</strong> Research<br />

on behalf of the <strong>Symposium</strong> organizers.<br />

Permission to reprint or re-use any<br />

abstract in this Supplement must be<br />

obtained from the copyright holder.<br />

For more information, contact the<br />

Symposia Director (below).<br />

<strong>Symposium</strong> Contact<br />

Mr. Rich Markow, Symposia Director<br />

<strong>Cancer</strong> Therapy & Research Center<br />

UTHSCSA<br />

7979 Wurzbach Road, MC 8224<br />

<strong>San</strong> <strong>Antonio</strong>, TX 78229<br />

Phone: 210-450-1550<br />

Fax: 210-450-1560<br />

E-mail: sabcs@uthscsa.edu<br />

Website Address: www.sabcs.org<br />

January 1, 1978. The law stipulates<br />

that copyright for works is vested<br />

in the author from the moment of<br />

creation and remains the property of<br />

the author until legally transferred.<br />

Authors who wish to publish articles<br />

and other material in AACR journals<br />

must formally transfer copyright to<br />

AACR. The copyright transfer form<br />

must be signed by all authors before<br />

AACR can proceed with publication.<br />

Appropriate forms for transfer of<br />

copyright will be requested from all<br />

authors after a manuscript has been<br />

deemed potentially acceptable.<br />

To read the complete version of<br />

AACR’s Copyright and Permissions<br />

Policy, please visit http://www.aacr.<br />

org/home/scientists/publications-ofthe-aacr/copyright-and-permissionspolicy.aspx.<br />

This policy includes further<br />

information on authors’ rights of<br />

usage, permission requests from third<br />

parties, free access to AACR journals,<br />

funding agency requirements, and the<br />

National Institutes of Health Public<br />

Access Policy.<br />

<strong>Cancer</strong> Research is abstracted or indexed<br />

in Biological <strong>Abstract</strong>s, Biochemistry<br />

& Biophysics Citation Index, Basic<br />

BIOSIS, BIOSIS Previews (R) Database,<br />

Chemical <strong>Abstract</strong>s, Index Medicus,<br />

MEDLINE, Current Contents/Clinical<br />

Medicine, Reference Update, and Science<br />

Citation Index (expanded).<br />

No responsibility is accepted by the<br />

Editors or by AACR, Inc. for the opinions<br />

expressed by contributors or for<br />

the content of the advertisements.<br />

<strong>Cancer</strong> Research Supplement


Editor-in-Chief<br />

George C. Prendergast<br />

<strong>Cancer</strong><br />

Research<br />

Special Advisors to the<br />

Editor-in-Chief<br />

Olivera J. Finn<br />

Giorgio Trinchieri<br />

Deputy Editor for Meeting<br />

Reports<br />

Mariano Barbacid<br />

Deputy Editor for<br />

Breaking Advances<br />

William A. Weiss<br />

Breaking Advances Editors<br />

Kenneth D. Aldape<br />

Frances Balkwill<br />

Paul B. Fisher<br />

Davide Ruggero<br />

Deputy Editor for Reviews<br />

Danny R. Welch<br />

Reviews Editors<br />

Suzanne A. Eccles<br />

Jeremy R. Graff<br />

Martine F. Roussel<br />

Senior Editors<br />

Clinical Studies<br />

Theodore S. Lawrence<br />

Integrated Systems & Technologies<br />

Martin W. Brechbiel<br />

Arul M. Chinnaiyan<br />

Trachette L. Jackson<br />

Sofia D. Merajver<br />

David W. Speicher<br />

Microenvironment & Immunology<br />

Hellmut G. Augustin<br />

Mario P. Colombo<br />

Yves A. DeClerck<br />

Suzanne Ostrand-Rosenberg<br />

Kornelia Polyak<br />

Mark J. Smyth<br />

Laurence Zitvogel<br />

Molecular & Cellular Pathobiology<br />

John D. Carpten<br />

Junjie Chen<br />

Kathleen R. Cho<br />

Mark W. Dewhirst<br />

Karen E. Knudsen<br />

Hiroyuki Mano<br />

Jeffrey E. Settleman<br />

Prevention & Epidemiology<br />

Saraswati Sukumar<br />

Stephen M. Schwartz<br />

Tumor & Stem Cell Biology<br />

Laura E. Benjamin<br />

Maria A. Blasco<br />

Myles A. Brown<br />

Lewis Chodosh<br />

Philip W. Hinds<br />

Nancy E. Hynes<br />

Rakesh Kumar<br />

Toshikazu Ushijima<br />

Jane E. Visvader<br />

Max Wicha<br />

Therapeutics, Targets & Chemical<br />

Biology<br />

Robert Clarke<br />

<strong>Antonio</strong> T. Fojo<br />

Janet A. Houghton<br />

William G. Nelson<br />

Patrick M. O’Connor<br />

Yves Pommier<br />

David B. Solit<br />

Kenneth D. Tew<br />

Editorial Board<br />

Cory A. Abate-Shen<br />

James L. Abbruzzese<br />

Gregory P. Adams<br />

Natalie G. Ahn<br />

Arthur S. Alberts<br />

Adriana Albini<br />

Alexander R.A. Anderson<br />

Andrew E. Aplin<br />

Leonard H. Augenlicht<br />

Albert S. Baldwin<br />

Glen N. Barber<br />

Menashe Bar-Eli<br />

Per H. Basse<br />

Paula J. Bates<br />

Stephen B. Baylin<br />

David G. Beer<br />

Doris M. Benbrook<br />

Gabriele Bergers<br />

Jay A. Berzofsky<br />

Darell D. Bigner<br />

Diane F. Birt<br />

Mikhail V. Blagosklonny<br />

Carl P. Blobel<br />

Melissa Bondy<br />

Rolf A. Brekken<br />

Angela M. H. Brodie<br />

Lisa H. Butterfield<br />

Bruno Calabretta<br />

Anthony J. Capobianco<br />

Giuseppe Carruba<br />

Graham Casey<br />

Esteban Celis<br />

James R. Cerhan<br />

Timothy C. Chambers<br />

Jia Chen<br />

Cheng-Yang Chou<br />

David C. Christiani<br />

Robert J. Coffey<br />

Michael Cole<br />

David R. Corey<br />

Vittorio Cristini<br />

Tom Curran<br />

Rajvir Dahiya<br />

William S. Dalton<br />

Mary B. Daly<br />

Chi Van Dang<br />

Ramana V. Davuluri<br />

Michael J. Detmar<br />

Ivan Dikic<br />

Julie Y. Djeu<br />

Ethan Dmitrovsky<br />

Zigang Dong<br />

Martin Eilers<br />

Wafik S. El-Deiry<br />

Lee M. Ellis<br />

Manel Esteller<br />

Stephen P. Ethier<br />

Jia Fan<br />

Eric R. Fearon<br />

Dean W. Felsher<br />

Napoleone Ferrara<br />

Soldano Ferrone<br />

Susan M. Fischer<br />

Richard Fishel<br />

James M. Ford<br />

Albert J. Fornace<br />

Steven M. Frisch<br />

Simone Fulda<br />

Amy M. Fulton<br />

Dmitry I. Gabrilovich<br />

Thomas F. Gajewski<br />

Joel R. Garbow<br />

Robert A. Gatenby<br />

Richard B. Gaynor<br />

Adi F. Gazdar<br />

Edward P. Gelmann<br />

Amato J. Giaccia<br />

Susan K. Gilmour<br />

David Gius<br />

Candece L. Gladson<br />

Andrew K. Godwin<br />

Marc T. Goodman<br />

Steven Grant<br />

Mark Greene<br />

Andrei V. Gudkov<br />

David H. Gutmann<br />

William C. Hahn<br />

Susan E. Hankinson<br />

Stephen S. Hecht<br />

Kristian Helin<br />

Mary J.C. Hendrix<br />

Roy S. Herbst<br />

Peter J. Houghton<br />

Suyun Huang<br />

Tim H-M Huang<br />

Mien-Chie Hung<br />

Kent W. Hunter<br />

John T. Isaacs<br />

William B. Isaacs<br />

Jean-Pierre Issa<br />

Kuan-Teh Jeang<br />

Daniel E. Johnson<br />

Randall S. Johnson<br />

Peter A. Jones<br />

Carl H. June<br />

Scott E. Kern<br />

Khandan Keyomarsi<br />

Roya Khosravi-Far<br />

Timothy J. Kinsella<br />

Erik Knudsen<br />

H. Phillip Koeffler<br />

Guido Kroemer<br />

Jonathan M. Kurie<br />

Natasha Kyprianou<br />

Lucia R. Languino<br />

Edmund C. Lattime<br />

Gustavo W. Leone<br />

Jason S. Lewis<br />

Linheng Li<br />

Michael P. Lisanti<br />

A. Thomas Look<br />

Philip Maini<br />

Linda H. Malkas<br />

Sridhar Mani<br />

James A. McCubrey<br />

Rene H. Medema<br />

Giovanni Melillo<br />

Andrew Mellor<br />

Stephen J. Meltzer<br />

Alea A. Mills<br />

Debabrata Mukhopadhyay<br />

Hasan Mukhtar<br />

Alexander Muller<br />

David H. Munn<br />

Maureen E. Murphy<br />

Akira Nakagawara<br />

Keiichi Nakayama<br />

Steven Narod<br />

Peter S. Nelson<br />

Tetsuo Noda<br />

Jeffrey A. Norton<br />

Giuseppina Nucifora<br />

Olufunmilayo Olopade<br />

Andrew F. Olshan<br />

Torben F. Orntoft<br />

Donald M. O’Rourke<br />

Alan B. Packard<br />

Renata Pasqualini<br />

Linda Z. Penn<br />

Russell O. Pieper<br />

Christoph Plass<br />

Elizabeth A. Platz<br />

Eckhard R. Podack<br />

Vito Quaranta<br />

Andrew A. Quong<br />

Nancy Raab-Traub<br />

Ayyappan Rajasekaran<br />

Ratna Ray<br />

Melvin Reichman<br />

Tannishtha Reya<br />

Ulrich Rodeck<br />

Eric K. Rowinsky<br />

Anil K. Rustgi<br />

Marianne D. Sadar<br />

Edward A. Sausville<br />

Janet A. Sawicki<br />

Jeffrey Schlom<br />

Hans-Joachim Schmoll<br />

Michael J. Seckl<br />

Gregg L. Semenza<br />

Charles L. Sentman<br />

Julian M. Simon<br />

Frank J. Slack<br />

Michael B. Sporn<br />

M. Sharon Stack<br />

Patricia S. Steeg<br />

Joann B. Sweasy<br />

Bin T. Teh<br />

Rajeshwar R. Tekmal<br />

Joseph R. Testa<br />

Andrew M. Thorburn<br />

Donald J. Tindall<br />

Nicholas Tonks<br />

David A. Tuveson<br />

Cornelia M. Ulrich<br />

Alejandra C. Ventura<br />

Paula M. Vertino<br />

Robert H. Vonderheide<br />

Kristiina Vuori<br />

Cheryl L. Walker<br />

Jean Y.J. Wang<br />

Liwei Wang<br />

Jeffrey S. Weber<br />

Robert J. Wechsler-Reya<br />

Louis M. Weiner<br />

Cynthia Wetmore<br />

T.C. Wu<br />

Xifeng Wu<br />

Keping Xie<br />

Ningzhi Xu<br />

Xiang-Xi Xu<br />

Minoru Yoshida<br />

Ming You<br />

Dihua Yu<br />

Ming-Ming Zhou<br />

Mary M. Zutter<br />

<strong>Cancer</strong> Research Supplement


<strong>Cancer</strong> Research<br />

A Journal of the American Association for <strong>Cancer</strong> Research<br />

Volume 72 • Number 24 • Supplement 3<br />

December 15, 2012 • Pages 1s–608s<br />

2012 <strong>Abstract</strong>s<br />

35th Annual <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

December 4–8, 2012<br />

<strong>San</strong> <strong>Antonio</strong>, Texas, USA<br />

www.sabcs.org<br />

Program Schedule<br />

Invited <strong>Abstract</strong>s<br />

1s<br />

78s<br />

<strong>Abstract</strong>s<br />

General Sessions [S1-1 – S6-7]<br />

Poster Discussion Sessions [PD01-01 – PD10-09]<br />

Poster Session 1 [P1-01-01– P1-15-12]<br />

Poster Session 2 [P2-01-01– P2-16-04]<br />

Poster Session 3 [P3-01-01– P3-14-06]<br />

Poster Session 4 [P4-01-01– P4-17-09]<br />

Poster Session 5 [P5-01-01– P5-21-04]<br />

Poster Session 6 [P6-01-01– P6-14-05]<br />

Poster Session Ongoing Trial 1 [OT1-1-01– OT1-4-06]<br />

Poster Session Ongoing Trial 2 [OT2-1-01– OT2-3-11]<br />

Poster Session Ongoing Trial 3 [OT3-1-01– OT3-4-06]<br />

89s<br />

110s<br />

147s<br />

214s<br />

285s<br />

351s<br />

420s<br />

486s<br />

556s<br />

566s<br />

576s<br />

Author Index for <strong>Abstract</strong>s<br />

587s<br />

Note Regarding the Citation of <strong>Abstract</strong>s<br />

<strong>Abstract</strong>s published in this Supplement can be cited as shown in the following example:<br />

Impact of breast cancer CME: Physician practice pattern, knowledge, and competence assessments. Haas M, Heintz A, Stacy T.<br />

<strong>Cancer</strong> Res 2012;72(24 Suppl.):<strong>Abstract</strong> nr P6-14-05.


December 4–8, 2012<br />

Program Schedule<br />

DECEMBER 4–8, 2012<br />

35TH ANNUAL<br />

SAN ANTONIO BREAST CANCER SYMPOSIUM<br />

SAN ANTONIO, TEXAS, USA<br />

WWW.SABCS.ORG<br />

www.aacrjournals.org 1s <strong>Cancer</strong> Res; 72(24Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

We are proud to acknowledge the following for their contributions to<br />

and generous support of our program (at press time).<br />

Premier<br />

Genentech, A Member of the Roche Group<br />

Angel<br />

Amgen<br />

Celgene Corporation<br />

Eisai, Inc.<br />

Genomic Health, Inc.<br />

Novartis Oncology<br />

Patron<br />

AlphaMed Press/The Oncologist<br />

Eli Lilly and Company<br />

Major Supporter<br />

AstraZeneca<br />

Celltrion Healthcare Co., Ltd.<br />

GE Healthcare<br />

Nektar Therapeutics<br />

Contributor<br />

Agendia, Inc.<br />

Bristol-Myers Squibb<br />

Fresenius Kabi Deutschland GMBH<br />

Merck<br />

Myriad Genetic Laboratories, Inc.<br />

NanoString Technologies, Inc.<br />

Pfizer<br />

DONOR<br />

Almac<br />

Ambry Genetics<br />

BioMed Central<br />

Boehringer Ingelheim Pharmaceuticals, Inc.<br />

Caris Life Sciences<br />

Carl Zeiss Meditec, Inc.<br />

DARA Biosciences<br />

Dilon Diagnostics<br />

Dune Medical Devices<br />

Elekta, Inc.<br />

Endomagnetics Ltd.<br />

Faxitron Bioptics<br />

Genoptix, Inc.<br />

GlaxoSmithKline (Commercial)<br />

GlaxoSmithKline (R&D)<br />

HistoRx, Inc.<br />

Inspire<br />

Leica Microsystems<br />

Mammotome<br />

ProStrakan<br />

Scion Medical Technologies<br />

Varian Medical Systems<br />

Veridex, LLC<br />

Xentech<br />

CONFERENCE GRANT<br />

American <strong>Cancer</strong> Society, Texas Division<br />

National <strong>Cancer</strong> Institute<br />

SABCS gratefully acknowledges<br />

Susan G. Komen for the Cure® for generous support of<br />

AACR Outstanding Investigator Award<br />

for <strong>Breast</strong> <strong>Cancer</strong> Research,<br />

CTCR-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Educational Sessions,<br />

AACR Scholar-in-Training Awards<br />

SABCS wishes to thank Avon Foundation for support of the<br />

Avon Foundation-AACR International<br />

Scholar-In-Training Grants<br />

Future <strong>Symposium</strong> Meeting Dates<br />

36th Annual SABCS December 10–14, 2013<br />

37th Annual SABCS December 9–13, 2014<br />

38th Annual SABCS December 8–12, 2015<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012 2s <strong>Cancer</strong> Research


December 4–8, 2012<br />

Program Schedule<br />

2012 CTRC-AACR<br />

<strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong><br />

<strong>Symposium</strong><br />

Program Schedule<br />

(AT PRESS TIME)<br />

Refer to www.sabcs.org for the most current information.<br />

Room Locations<br />

Exhibit Halls A, B, C & D: Street Level<br />

Ballrooms A & B: Street Level<br />

Bridge Hall: Street Level<br />

Room 206 A-B: Concourse Level<br />

Tuesday, December 4, 2012<br />

8:00 am–7:30 pm<br />

REGISTRATION<br />

Bridge Hall<br />

Pre-registered attendees can obtain materials. Those who<br />

have not yet registered may do so.<br />

12:30 pm–2:00 pm<br />

Career Development Forum: A Networking Session for<br />

Young Investigators<br />

Room 206 A–B<br />

Networking and career development opportunities for early career<br />

scientists. The session is open to early-career scientists, defined as<br />

graduate students, postdoctoral or clinical fellows, or medical students<br />

and residents, who are registered attendees of the SABCS.<br />

Space in the workshop is limited to 200 participants; registrations will be<br />

accepted on a first-come, first-served basis and is free of charge.<br />

Discussion topics & mentors*<br />

Balancing Research and Clinical Practice I<br />

Judy E. Garber, Dana-Farber <strong>Cancer</strong> Institute<br />

Ann H. Partridge, Dana-Farber <strong>Cancer</strong> Institute<br />

Balancing Research and Clinical Practice II<br />

Clifford A. Hudis, Memorial Sloan-Kettering <strong>Cancer</strong> Center<br />

Eric P. Winer, Dana-Farber <strong>Cancer</strong> Institute<br />

Careers in Industry<br />

Steven Shak, Genomic Health, Inc.<br />

Careers in Industry<br />

Mika Derynck, Genentech, Inc.<br />

Careers in Translational Research<br />

Carlos L. Arteaga, Vanderbilt-Ingram <strong>Cancer</strong> Center<br />

Grant Writing - Basic/Translational<br />

Yi Li, Baylor College of Medicine <strong>Cancer</strong> Center<br />

Grant Writing - Clinical/Translational<br />

Virginia F. Borges, University of Colorado Denver<br />

Matthew J. Ellis, Washington University Siteman <strong>Cancer</strong> Center<br />

How to Become a Clinical Trialist<br />

John R. Yarnold, The Royal Marsden Hospital<br />

Nancy U. Lin, Dana-Farber <strong>Cancer</strong> Institute<br />

How to Get the Most Out of Your Fellowship Years<br />

Ian Elliott Krop, Dana-Farber <strong>Cancer</strong> Institute<br />

How to Get the Most Out of Your Fellowship Years<br />

Rachel Schiff, Baylor College of Medicine<br />

How to Get the Most Out of Your Predoctoral Experience<br />

Rong Li, UT Health Science Center at <strong>San</strong> <strong>Antonio</strong><br />

How to Make the Most Out of Being A Cooperative Group Member<br />

Kathy D. Miller, Indiana University School of Medicine<br />

Hope S. Rugo, UCSF Helen Diller Family Comprehensive <strong>Cancer</strong> Center<br />

Making the Transition From Fellowship to Faculty<br />

Pepper Jo Schedin, University of Colorado Anschutz Medical Campus<br />

Bryan P. Schneider, Indiana University Melvin and Bren Simon <strong>Cancer</strong><br />

Center<br />

Mentoring and Supervising<br />

Cathrin Brisken, Ecole Polytechnique Fédérale de Lausanne<br />

Jason S. Carroll, <strong>Cancer</strong> Research UK Cambridge Research Institute<br />

Negotiating a Job Offer or Promotion<br />

Melissa L. Bondy, Baylor College of Medicine <strong>Cancer</strong> Center<br />

Oral Presentation Skills<br />

Laura J. van ‘t Veer, UCSF Helen Diller Family Comprehensive<br />

<strong>Cancer</strong> Center<br />

Douglas Yee, Masonic <strong>Cancer</strong> Center, University of Minnesota<br />

Publication Strategies<br />

Harold J. Burstein, Dana-Farber <strong>Cancer</strong> Institute<br />

Searching for a Job and Interviewing<br />

Michael T. Lewis, Baylor College of Medicine <strong>Cancer</strong> Center<br />

Setting Up and Managing a Laboratory<br />

Jeffrey M. Rosen, Baylor College of Medicine<br />

Tenure and Promotion<br />

Suzanne A.W. Fuqua, Baylor College of Medicine <strong>Cancer</strong> Center<br />

Sharon H. Giordano, The University of Texas MD Anderson<br />

<strong>Cancer</strong> Center<br />

The Path Leading to Clinical Trials<br />

Massimo Cristofanilli, Fox Chase <strong>Cancer</strong> Center<br />

* Topics and mentors are subject to change<br />

2:00 pm–7:30 pm<br />

Educational Sessions<br />

Ballrooms A & B and Exhibit Hall D<br />

Supported by an educational grant from<br />

Susan G. Komen for the Cure®<br />

An update on advances in the technologies available for translational<br />

research. Sessions are to provide people with a better understanding<br />

of the topics of special current interest they hear using the techniques<br />

described. These presentations also provide researchers with a guide to<br />

the techniques they should be considering for their studies.<br />

2:00 pm–3:30 pm<br />

The Practical Use of Molecular Profiling<br />

Ballroom A<br />

Moderator: Peter M. Ravdin, MD, PhD<br />

UT Health Science Center <strong>San</strong> <strong>Antonio</strong><br />

<strong>San</strong> <strong>Antonio</strong>, TX<br />

Molecular profiling for in situ carcinoma of the breast<br />

Lawrence J. Solin, MD, FACR, FASTRO<br />

Albert Einstein Medical Center<br />

Philadelphia, PA<br />

The practical use of molecular profiling: The view of a<br />

medical oncologist<br />

<strong>Antonio</strong> C. Wolff, MD<br />

Johns Hopkins Kimmel <strong>Cancer</strong> Center<br />

Baltimore, MD<br />

Use of genomic tests in routine practice and clinical<br />

research in metastatic breast cancer<br />

Lajos Pusztai, MD, DPhil<br />

Yale <strong>Cancer</strong> Center<br />

New Haven, CT<br />

www.aacrjournals.org<br />

3s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Biology of Triple-Negative <strong>Breast</strong> <strong>Cancer</strong><br />

Ballroom B<br />

Moderator: Nicholas Turner, MD<br />

Royal Marsden Hospital<br />

London, UNITED KINGDOM<br />

New vulnerabilities for TNBC – from genetics to<br />

therapeutics<br />

Thomas Westbrook, PhD<br />

Baylor College of Medicine<br />

Houston, TX<br />

Triple negative breast cancer: Subtypes, molecular targets,<br />

and therapeutic approaches<br />

Jennifer A. Pietenpol, PhD<br />

Vanderbilt University School of Medicine<br />

Nashville, TN<br />

The clonal and mutational evolution of primary triple<br />

negative breast cancers<br />

Samuel Aparicio, PhD<br />

University of British Columbia<br />

Vancouver, CANADA<br />

Clinical Trial Designs for New Therapies<br />

Exhibit Hall D<br />

Moderator: Susan G. Hilsenbeck, PhD<br />

Baylor College of Medicine<br />

Houston, TX<br />

Early phase trials - What are they for?<br />

Susan G. Hilsenbeck, PhD<br />

Baylor College of Medicine<br />

Houston, TX<br />

What agents should go into trials?<br />

Angela DeMichele, MD, MSCE<br />

University of Pennsylvania Perelman School of Medicine<br />

Philadelphia, PA<br />

What endpoints should we use?<br />

Clifford A. Hudis, MD<br />

Memorial Sloan-Kettering <strong>Cancer</strong> Center<br />

New York, NY<br />

Randomized clinical trial designs: Still important<br />

Sally Hunsberger, PhD<br />

National <strong>Cancer</strong> Institute<br />

Rockville, MD<br />

What have we learned?<br />

Susan G. Hilsenbeck, PhD<br />

Baylor College of Medicine<br />

Houston, TX<br />

4:00 pm–5:30 pm<br />

Navigating the Obstacles and Risks of Survivorship<br />

Ballroom A<br />

Moderator: Susan W. Rafte<br />

Pink Ribbons Project<br />

Houston, TX<br />

Emerging sexual pharmacology for the breast cancer<br />

survivor<br />

Michael Krychman, MD<br />

The Southern California Center for Sexual Health and Survivorship<br />

Newport Beach, CA<br />

Cognitive changes and breast cancer treatments<br />

Patricia A. Ganz, MD<br />

University of California, Los Angeles<br />

Los Angeles, CA<br />

Risk of second primary breast cancer and other serious late<br />

complications issues among survivors of breast cancer<br />

Jonine L. Bernstein, PhD<br />

Memorial Sloan-Kettering <strong>Cancer</strong> Center<br />

New York, NY<br />

Mammary Cell Lineages and <strong>Breast</strong> <strong>Cancer</strong> Subtypes<br />

Ballroom B<br />

Moderator: Michael T. Lewis, PhD<br />

Baylor College of Medicine<br />

Houston, TX<br />

Primitive normal human mammary cells – the forerunners<br />

of malignant populations<br />

Connie Eaves, PhD<br />

University of British Columbia<br />

Vancouver, CANADA<br />

Deciphering the cellular hierarchy of the mammary gland<br />

and its implication for breast cancer<br />

Marielle Ousset, PhD<br />

Université Libre de Bruxelles<br />

Bruxelles, BELGIUM<br />

Understanding human cancer with single cell genetics<br />

Nicholas E. Navin, PhD<br />

UT MD Anderson <strong>Cancer</strong> Center<br />

Houston, TX<br />

Controversies in the Surgical Management of <strong>Breast</strong> <strong>Cancer</strong><br />

Exhibit Hall D<br />

Moderator: Ismail Jatoi, MD, PhD, FACS<br />

UT Health Science Center <strong>San</strong> <strong>Antonio</strong><br />

<strong>San</strong> <strong>Antonio</strong>, TX<br />

Sentinel node biopsy in breast cancer: Before or after<br />

neoadjuvant chemotherapy<br />

Eleftherios Mamounas, MD<br />

Aultman Hospital<br />

Canton, OH<br />

Evolving trends in implant breast reconstruction<br />

Andrea L. Pusic, MD, MHS, FRCSC<br />

Memorial Sloan-Kettering <strong>Cancer</strong> Center<br />

New York, NY<br />

The role of MRI in management of primary breast cancer<br />

Ismail Jatoi, MD, PhD, FACS<br />

UT Health Science Center <strong>San</strong> <strong>Antonio</strong><br />

<strong>San</strong> <strong>Antonio</strong>, TX<br />

6:00 pm–7:30 pm<br />

Tumor Dormancy and Late Recurrences<br />

Ballroom A<br />

Moderator: George W. Sledge, Jr, MD<br />

Indiana University Simon <strong>Cancer</strong> Center<br />

Indianapolis, IN<br />

Mechanisms of breast cancer dormancy and recurrence<br />

Lewis A. Chodosh, MD, PhD<br />

Perelman School of Medicine<br />

University of Pennsylvania<br />

Philadelphia, PA<br />

Tumor dormancy from an experimental biologist’s<br />

perspective<br />

Ann F. Chambers, PhD<br />

London Regional <strong>Cancer</strong> Program<br />

London, CANADA<br />

Attacking tumor dormancy in the clinic: How do we design<br />

the trials?<br />

George W. Sledge, Jr, MD<br />

Indiana University Simon <strong>Cancer</strong> Center<br />

Indianapolis, IN<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012 4s <strong>Cancer</strong> Research


December 4–8, 2012<br />

Program Schedule<br />

Inflammation and <strong>Breast</strong> <strong>Cancer</strong><br />

Ballroom B<br />

Moderator: Charlotte Kuperwasser, PhD<br />

Tufts University School of Medicine<br />

Boston, MA<br />

Mechanism-based insights into prognosis and treatment<br />

of breast cancer derived from high resolution multiphoton<br />

imaging<br />

John Condeelis, PhD<br />

Albert Einstein College of Medicine<br />

Bronx, NY<br />

<strong>Breast</strong> cancer stroma: a predictor of clinical outcome and<br />

tumour heterogeneity<br />

Morag Park, PhD<br />

McGill University<br />

Montreal, CANADA<br />

Links between inflammation, breast cancer and obesity<br />

Charlotte Kuperwasser, PhD<br />

Tufts University School of Medicine<br />

Boston, MA<br />

Her2-Positive <strong>Breast</strong> <strong>Cancer</strong><br />

Exhibit Hall D<br />

Moderator: Eric P. Winer, MD<br />

Dana-Farber <strong>Cancer</strong> Institute<br />

Boston, MA<br />

Adjuvant therapy of HER2 positive breast cancer – the next<br />

installment<br />

Karen Gelmon, MD<br />

BC <strong>Cancer</strong> Agency<br />

Vancouver, CANADA<br />

Advances in HER2+ breast cancer<br />

Nancy U. Lin, MD<br />

Dana-Farber <strong>Cancer</strong> Institute<br />

Boston, MA<br />

Neoadjuvant approach in HER2 over-expressing breast<br />

cancer: Therapeutic implications and biomarker discovery<br />

Mothaffar Rimawi, MD<br />

Baylor College of Medicine<br />

Houston, TX<br />

Wednesday, December 5, 2012<br />

7:00 am–5:15 pm<br />

Registration<br />

Bridge Hall<br />

7:00 am–8:30 am<br />

Continental Breakfast<br />

Exhibit Hall C<br />

7:45 am–8:00 am<br />

WELCOME AND Opening Remarks<br />

Exhibit Hall D<br />

8:00 am–8:30 am<br />

PLENARY LECTURE 1<br />

Exhibit Hall D<br />

Hormones and the <strong>Breast</strong>: Local and Environmental Interactions<br />

Cathrin Brisken, MD, PhD<br />

Ecole Polytechnique Fédérale de Lausanne<br />

Lausanne, SWITZERLAND<br />

8:30 am–11:15 am<br />

GENERAL SESSION 1<br />

Exhibit Hall D<br />

Moderator: Kathy S. Albain, MD, FACP<br />

Loyola University Chicago Stritch School of Medicine<br />

Maywood, IL<br />

8:30 S1-1. Relative effectiveness of letrozole compared with tamoxifen<br />

for patients with lobular carcinoma in the BIG 1-98 trial<br />

Metzger O, Giobbie-Hurder A, Mallon E, Viale G, Winer E, Thürlimann B,<br />

Gelber RD, Colleoni M, Ejlertsen B, Bonnefoi H, Coates AS, Goldhirsch A,<br />

Gusterson B. BIG 1-98 Collaborative Group, International <strong>Breast</strong> <strong>Cancer</strong><br />

Study Group.<br />

8:45 S1-2. ATLAS – 10 v 5 years of adjuvant tamoxifen (TAM) in ER+<br />

disease: Effects on outcome in the first and in the second decade<br />

after diagnosis<br />

Davies C, Pan H, Godwin J, Gray R, Peto R, on Behalf of ATLAS<br />

Collaborators Worldwide. University of Oxford, Oxford, United Kingdom;<br />

Glasgow Caledonian University, Glasgow, United Kingdom.<br />

9:00 S1-3. Discussion<br />

William E. Barlow, PhD, Fred Hutchinson <strong>Cancer</strong> Research Center,<br />

Seattle, WA<br />

9:15 S1-4. Final analysis of overall survival for the Phase III CONFIRM<br />

trial: fulvestrant 500 mg versus 250 mg<br />

Di Leo A, Jerusalem G, Petruzelka L, Torres R, Bondarenko IN, Khasanov<br />

R, Verhoeven D, Pedrini JL, Smirnova I, Lichinitser MR, Pendergrass K,<br />

Garnett S, Rukazenkov Y, Martin M. Hospital of Prato, Prato, Italy; Centre<br />

Hospitalier Universitaire Sart Tilman, Liège, Belgium; First Faculty of<br />

Medicine of Charles University, Prague, Czech Republic; Instituto Nacional<br />

del Cáncer, <strong>San</strong>tiago, Chile; Dnipropetrovsk Municipal Clinical Hospital,<br />

Dnipropetrovsk, Ukraine; Republican Clinical Oncological Center, Kazan,<br />

Russian Federation; AZ Klina, Brasschaat, Belgium; Hospital Nossa<br />

Senhora da Conceição, Porto Alegre, Brazil; Medical Radiological Science<br />

Center, Obninsk, Russian Federation; Russian <strong>Cancer</strong> Research Centre,<br />

Moscow, Russian Federation; Kansas City <strong>Cancer</strong> Center, Kansas City;<br />

AstraZeneca Pharmaceuticals, Macclesfield, United Kingdom; Hospital<br />

Universitario Gregorio Maranon, Madrid, Spain.<br />

9:30 S1-5. PIK3CA mutations are linked to PgR expression: A Tamoxifen<br />

Exemestane Adjuvant Multinational (TEAM) pathology study<br />

Sabine VS, Crozier C, Drake C, Piper T, van de Velde CJH, Hasenburg A,<br />

Kieback DG, Markopoulos C, Dirix L, Seynaeve C, Rea D, Bartlett JMS.<br />

Ontario Institute for <strong>Cancer</strong> Research, Toronto, ON, Canada; University<br />

of Edinburgh <strong>Cancer</strong> Research Centre, Institute of Genetics & Molecular<br />

Medicine, Edinburgh, Scotland, United Kingdom; Leiden University<br />

Medical Center, Leiden, Netherlands; University Hospital, Freiburg,<br />

Germany; Elblandklinikum, Riesa, Germany; Athens University Medical<br />

School, Athens, Greece; St. Augustinus Hospital, Antwerp, Belgium;<br />

Erasmus Medical Center-Daniel den Hoed, Rotterdam, United Kingdom;<br />

University of Birmingham, Birmingham, United Kingdom.<br />

9:45 S1-6. Results of a randomized phase 2 study of PD 0332991, a<br />

cyclin-dependent kinase (cdk) 4/6 inhibitor, in combination with<br />

letrozole vs letrozole alone for first-line treatment of ER+/HER2-<br />

advanced breast cancer (BC)<br />

Finn RS, Crown JP, Lang I, Boer K, Bondarenko IM, Kulyk SO, Ettl J, Patel<br />

R, Pinter T, Schmidt M, Shparyk Y, Thummala AR, Voytko NL, Breazna A,<br />

Kim ST, Randolph S, Slamon DJ. University of California, Los Angeles, CA;<br />

Irish Cooperative Oncology Research Group, Dublin, Ireland; Orszagos<br />

Onkologiai Intezet, Budapest, Hungary; Szent Margit Korhaz, Budapest,<br />

Hungary; Dnipropetrovsk City Multiple-Discipline Clinical Hospital,<br />

Ukraine; Municipal Treatment-and-Prophylactic Institution “Donetsk<br />

City Oncological Dispensary”, Ukraine; Technical University of Munich,<br />

Germany; Comprehensive Blood and <strong>Cancer</strong> Center, Bakersfield, CA; Petz<br />

Aladar Megyei Oktato Korhaz, Gyor, Hungary; University Hospital Mainz,<br />

Germany; Lviv State Oncologic Regional Treatment and Diagnostic<br />

Center, Ukraine; Comprehensive <strong>Cancer</strong> Centers of Nevada, Henderson,<br />

NV; Kyiv City Clinical Oncology Center, Ukraine; Pfizer Oncology, New<br />

York, NY; Pfizer Oncology, <strong>San</strong> Diego, CA.<br />

www.aacrjournals.org<br />

5s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

10:00 S1-7. Phase III trial evaluating the addition of bevacizumab to<br />

endocrine therapy as first-line treatment for advanced breast<br />

cancer - First efficacy results from the LEA study<br />

Martin M, Loibl S, von Minckwitz G, Morales S, Crespo C, Anton A,<br />

Guerrero A, Aktas B, Schoenegg W, Muñoz M, Garcia-Saenz JA, Gil M,<br />

Ramos M, Carrasco E, Liedtke C, Wachsmann G, Mehta K, De la Haba<br />

JR, On behalf of GEICAM (Spanish <strong>Breast</strong> <strong>Cancer</strong> Research Group), GBG<br />

(German <strong>Breast</strong> Group). Instituto de Investigacion <strong>San</strong>itaria Gregorio<br />

Marañon, Madrid, Spain; GBG (German <strong>Breast</strong> Group), Neu-Isenburg,<br />

Germany; University Women´s Hospital Essen, Germany; Medical<br />

Practice Berlin, Germany; University Women´s Hospital Muenster,<br />

Germany; Klinikum Boeblingen, Germany; H. Arnau Vilanova de Lerida,<br />

Spain; Hospital U. Ramon y Cajal, Spain; Hospital Universitario Miguel<br />

Servet, Spain; Instituto Valenciano de Oncologia, Spain; Hospital Clinic i<br />

Provincial, Spain; Hospital Clinico U. <strong>San</strong> Carlos, Spain; Instituto Catala d’<br />

Oncologia Hospitalet, Spain; GEICAM (Spanish <strong>Breast</strong> <strong>Cancer</strong> Research<br />

Group), Spain; Hospital U. Reina Sofia, Spain; Centro Oncologico de<br />

Galicia, Spain.<br />

10:15 S1-8. Prolactin-humanized mice: an improved animal recipient<br />

for therapy response-testing of patient-derived breast cancer<br />

xenotransplants<br />

Rui H, Utama FE, Yanac AF, Xia G, Peck AR, Liu C, Rosenberg AL, Wagner<br />

K-U, Yang N. Thomas Jefferson University, Philadelphia, PA; Eppley<br />

Institute for Research in <strong>Cancer</strong> and Allied Diseases, Omaha, NE.<br />

10:30 S1-9. Comparative performance of breast cancer Index (BCI) vs.<br />

Oncotype Dx and IHC4 in the prediction of late recurrence in<br />

hormonal receptor-positive lymph node-negative breast cancer<br />

patients: A TransATAC Study<br />

Sgroi DC, Sestak I, Cuzick J, Zhang Y, Schnabel CA, Erlander MG, Goss PE,<br />

Dowsett M. Massachusetts General Hospital, Harvard Medical School,<br />

Boston, MA; Centre for <strong>Cancer</strong> Prevention, Queen Mary University,<br />

London, United Kingdom; bioTheranostics Inc., <strong>San</strong> Diego, CA;<br />

Breakthrough <strong>Breast</strong> <strong>Cancer</strong> Centre, Royal Marsden Hospital, London,<br />

United Kingdom.<br />

10:45 S1-10. Association between the 21-gene recurrence score (RS)<br />

and benefit from adjuvant paclitaxel (Pac) in node-positive (N+),<br />

ER-positive breast cancer patients (pts): Results from NSABP B-28<br />

Mamounas EP, Tang G, Paik S, Baehner FL, Liu Q, Jeong J-H, Kim S-R,<br />

Butler SM, Jamshidian F, Cherbavaz DB, Sing AP, Shak S, Julian TB,<br />

Lembersky BC, Wickerham DL, Costantino JP, Wolmark N. National<br />

Surgical Adjuvant <strong>Breast</strong> and Bowel Project Operations and Biostatistical<br />

Centers; Aultman Hospital; Graduate School of Public Health, University<br />

of Pittsburgh; Genomic Health, Inc.; Allegheny <strong>Cancer</strong> Center at<br />

Allegheny General Hospital; University of Pittsburgh <strong>Cancer</strong> Institute.<br />

11:00 S1-11. Meta-analysis Results from the Collaborative Trials in<br />

Neoadjuvant <strong>Breast</strong> <strong>Cancer</strong> (CTNeoBC)<br />

Cortazar P, Zhang L, Untch M, Mehta K, Costantino J, Wolmark N,<br />

Bonnefoi H, Cameron D, Gianni L, Valagussa P, Zujewski JA, Justice R,<br />

Loibl S, Wickerham L, Bogaerts J, Baselga J, Perou C, Blumenthal G,<br />

Blohmer J, Mamounas E, Bergh J, Semiglazov V, Prowell T, Eidtmann<br />

H, Paik S, Piccart M, Sridhara R, Fasching P, Swain SM, Slaets L, Tang<br />

S, Gerber B, Geyer C, Pazdur R, Ditsch N, Rastogi P, Eiermann W,<br />

vonMincwitz G. FDA; HELIOS Klinikum Berlin-Buch, Berlin, Germany;<br />

NSABP, Pittsburgh, PA; University of Pittsburgh; Institut Bergonié , INSERM<br />

U916, and Université Bordeaux Segalen, Bordeaux, France; University of<br />

Edinburgh, Edinburgh, Scotland, United Kingdom; <strong>San</strong> Raffaele Scientific<br />

Insitute, Milan, Italy; Fondazione Michelangelo, Milan, Italy; NCI, Bethesda,<br />

MD; Lineberger Comprehensive <strong>Cancer</strong> Center, Chapel Hill, NC; GBG<br />

Forschungs GmbH, Germany; Medstar Washington Hospital Center,<br />

Washington, DC; St. Gertrauden Hospital, Berlin, Germany; University<br />

Women’s Hospital, Kiel, Germany; University Womens Hospital, Erlangen,<br />

Germany; University Women’s Hospital, Rostock, Germany; University<br />

Women’s Hospital, Munich, Germany; Private Practice, Munich, Germany;<br />

Karolinska Institutet and University Hospital, Stockholm, Sweden; EORTC<br />

Headquarters, Brussels, Belgium; Memorial Sloan Kettering <strong>Cancer</strong><br />

Center, NY; NN Petrov Res. Inst. Of Oncology, St.-Petersburg, Russian<br />

Federation; Jules Bordet Institute, Brussels, Belgium.<br />

11:15 am–12:00 pm<br />

William L. McGuire Memorial Lecture<br />

Exhibit Hall D<br />

Neoadjuvant Systemic Therapy: Promising Experimental Model, or<br />

Improved Standard of Care?<br />

Gabriel N. Hortobagyi, MD<br />

UT MD Anderson <strong>Cancer</strong> Center<br />

Houston, TX<br />

12:00 pm–1:35 pm<br />

Lunch<br />

12:30 pm–1:35 pm<br />

Clinical Science Forum<br />

Treatment on the Edges: Discordance Between Stage and Biology<br />

Ballroom A<br />

Moderator: Kathy S. Albain, MD, FACP<br />

Loyola University Chicago Stritch School of Medicine<br />

Maywood, IL<br />

Bad stage, but good biology<br />

Kathy S. Albain, MD, FACP<br />

Loyola University Chicago Stritch School of Medicine<br />

Maywood, IL<br />

Low stage…but adverse biology<br />

Martine J. Piccart, MD, PhD<br />

Jules Bordet Institute<br />

Brussels, BELGIUM<br />

12:30 pm–1:35 pm<br />

Basic Science Forum<br />

Metastasis – Niches<br />

Ballroom B<br />

Moderator: Yibin Kang, PhD<br />

Princeton University<br />

Princeton, NJ<br />

<strong>Cancer</strong> stem cells interactions with their niche determine<br />

formation of breast cancer metastasis<br />

Joerg Huelsken, PhD<br />

Federal Technical University Lausanne<br />

Lausanne, SWITZERLAND<br />

Tumor-stromal interactions in bone metastasis<br />

Yibin Kang, PhD<br />

Princeton University<br />

Princeton, NJ<br />

1:45 pm–3:15 pm<br />

Mini-<strong>Symposium</strong> 1<br />

The mTOR Pathway: Role in Metabolism and as a<br />

Therapeutic Target<br />

Exhibit Hall D<br />

Moderator: Carlos L. Arteaga, MD<br />

Vanderbilt-Ingram <strong>Cancer</strong> Center<br />

Vanderbilt University<br />

Nashville, TN<br />

Targeting PI3K<br />

Lewis Cantley, PhD<br />

Beth Israel Deaconess Medical Center<br />

Boston, MA<br />

Translating regulation of mTOR and protein synthesis to the<br />

clinic for advanced breast cancer<br />

Robert Schneider, PhD<br />

NYU School of Medicine<br />

New York, NY<br />

Clinical development of mTOR inhibitors<br />

Fabrice André, MD, PhD<br />

Gustave Roussy Institute<br />

Villejuif, FRANCE<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012 6s <strong>Cancer</strong> Research


December 4–8, 2012<br />

Program Schedule<br />

3:15 pm–4:00 pm<br />

GENERAL SESSION 2<br />

Exhibit Hall D<br />

Moderator: Anthony Lucci, Jr., MD, FACS<br />

UT MD Anderson <strong>Cancer</strong> Center<br />

Houston, TX<br />

3:15 S2-1. The role of sentinel lymph node surgery in patients<br />

presenting with node positive breast cancer (T0-T4, N1-2) who<br />

receive neoadjuvant chemotherapy - results from the ACOSOG<br />

Z1071 trial<br />

Boughey JC, Suman VJ, Mittendorf EA, Ahrendt GM, Wilke LG, Taback B,<br />

Leitch AM, Flippo-Morton TS, Byrd DR, Ollila DW, Julian TB, McLaughlin<br />

SA, McCall L, Symmans WF, Le-Petross HT, Haffty BG, Buchholz TA, Hunt<br />

KK. Mayo Clinic, Rochester, MN; MD Anderson <strong>Cancer</strong> Center, Houston,<br />

TX; Magee-Womens Surgical Associates, Pittsburgh, PA; University<br />

of Wisconsin-Madison, WI; Columbia University Medical Center, New<br />

York, NY; University of Texas Southwestern Medical Center, Dallas,<br />

TX; Carolinas Medical Center, Charlotte, NC; University of Washington<br />

Medical Center, Seattle, WA; University of North Carolina - Chapel Hill,<br />

NC; Allegheny General Hospital, Pittsburgh, PA; Mayo Clinic, Jacksonville,<br />

FL; Duke University Medical Center, Durham, NC; The <strong>Cancer</strong> Institute of<br />

New Jersey, New Brunswick, NJ.<br />

3:30 S2-2. Sentinel lymph node biopsy before or after neoadjuvant<br />

chemotherapy - final results from the prospective German,<br />

multiinstitutional SENTINA-trial<br />

Kuehn T, Bauerfeind IGP, Fehm T, Fleige B, Helms G, Lebeau A, Liedtke C,<br />

von Minckwitz G, Nekljudova V, Schrenk P, Staebler A, Untch M. Klinikum<br />

Esslingen, Esslingen, Baden Wuerttemberg, Germany; Klinikum Landshut,<br />

Landshut, Bayern, Germany; Universitaetsfrauenklinik Tuebingen, Baden<br />

Wuerttemberg, Germany; Helios Klinikum Berlin-Buch, Berlin, Germany;<br />

University Medical Center Hamburg-Eppendorf, Hamburg, Germany;<br />

University Medical Center, Muenster, Nordrhein-Westfalen, Germany;<br />

German <strong>Breast</strong> Group, Neu- Isenburg, Hessen, Germany; AKH-LFKK, Linz,<br />

Austria; University Medical Center, Tuebingen, Baden-Wuerttemberg,<br />

Germany.<br />

3:45 S2-3. Disparities in the utilization of axillary sentinel lymph node<br />

biopsy among black and white patients with node-negative breast<br />

cancer from 2002-2007<br />

Black DM, Jiang J, Kuerer HM, Buchholz TA, Smith BD. MD Anderson<br />

<strong>Cancer</strong> Center, Houston, TX.<br />

4:00 pm–5:00 pm<br />

SUSAN G. KOMEN FOR THE CURE® BRINKER AWARDS FOR<br />

SCIENTIFIC DISTINCTION LECTURES<br />

Exhibit Hall D<br />

The Basic Science Award is presented to a researcher whose scientific<br />

discoveries or novel technologies have added substantively to our<br />

understanding of the basic biology of breast cancer and the intrinsic<br />

molecular processes that drive the disease, and/or whose work has<br />

bridged the gap between basic research and patient care. This year the<br />

award is being presented to:<br />

Yosef Yarden, PhD<br />

Department of Biological Regulation<br />

Weizmann Institute of Science<br />

Rehovot, ISRAEL<br />

How Does HER2 Contribute to <strong>Breast</strong> <strong>Cancer</strong> Progression?<br />

The Clinical Research Award is presented to a clinical or translational<br />

researcher who has advanced the identification of new prevention,<br />

detection or treatment approaches for breast cancer and promoted their<br />

incorporation into clinical care. This year the award is being presented to:<br />

Hyman B. Muss, MD<br />

Lineberger Comprehensive <strong>Cancer</strong> Center<br />

The University of North Carolina, Chapel Hill<br />

Chapel Hill, NC<br />

Older Women and <strong>Breast</strong> <strong>Cancer</strong>: Challenges and Opportunities<br />

5:00 pm–7:00 pm<br />

Poster Discussion 1: Endocrine Resistance<br />

Ballroom A<br />

Viewing 5:00 pm<br />

Discussion 5:15 pm<br />

Rachel Schiff, PhD, Chair<br />

Baylor College of Medicine<br />

Houston, TX<br />

Steffi Oesterreich, PhD, Discussant<br />

University of Pittsburgh <strong>Cancer</strong> Institute<br />

Pittsburgh, PA<br />

and<br />

Suleiman Massarweh, MD, Discussant<br />

University of Kentucky and Markey <strong>Cancer</strong> Center<br />

Lexington, KY<br />

PD01-01 Overcoming endocrine therapy resistance related to PTEN loss<br />

by strategic combinations with mTOR, AKT, or MEK inhibitors<br />

Fu X, Kumar V, Shea M, Biswal NC, Nanda S, Chayanam S, Mitchell<br />

T, Hergenroeder G, Meerbrey KL, Joshi A, Westbrook TF, Mills GB,<br />

Creighton CJ, Hilsenbeck SG, Osborne CK, Schiff R. Lester & Sue<br />

Smith <strong>Breast</strong> Center, Baylor College of Medicine, Houston, TX; Dan<br />

L Duncan <strong>Cancer</strong> Center, Baylor College of Medicine, Houston, TX;<br />

Baylor College of Medicine, Houston, TX; M.D. Anderson <strong>Cancer</strong><br />

Center, Houston, TX.<br />

PD01-02 Increased expression of phosphorylated mTOR in metastatic<br />

breast tumors compared to primary tumors in patients who<br />

received adjuvant endocrine therapy<br />

Hoefnagel LDC, Beelen KJ, Opdam M, Vincent A, Linn SC, vanDiest<br />

PJ. University Medical Center, Utrecht, Netherlands; The Netherlands<br />

<strong>Cancer</strong> Institute, Amsterdam, Netherlands.<br />

PD01-03 Phosphorylated p-70S6K predicts tamoxifen resistance in<br />

postmenopausal breast cancer patients randomized between<br />

adjuvant tamoxifen versus no systemic treatment<br />

Beelen KJ, Opdam M, Severson TM, Koornstra RHT, Vincent A,<br />

Wesseling J, Muris JJ, Berns EMJJ, Vermorken JB, vanDiest PJ, Linn<br />

SC. The Netherlands <strong>Cancer</strong> Institute, Amsterdam, Netherlands;<br />

Erasmus University Medical Center-Daniel den Hoed <strong>Cancer</strong> Center,<br />

Rotterdam, Netherlands; Antwerp University Hospital, Edegem,<br />

Belgium; University Medical Center, Utrecht, Netherlands.<br />

PD01-04 Deep kinome sequencing identifies a novel D189Y mutation<br />

in the Src family kinase LYN as a possible mediator of<br />

antiestrogen resistance in ER+ breast cancer<br />

Fox EM, Balko JM, Arteaga CL. Vanderbilt-Ingram <strong>Cancer</strong> Center,<br />

Vanderbilt University, Nashville, TN.<br />

PD01-05 Analysis of patients with ER-positive breast tumors treated<br />

with neoadjuvant aromatase inhibition identifies chemokine<br />

receptors as potential modulators of endocrine resistance<br />

Ribas R, Ghazoui Z, Dowsett M, Martin L-A. The Institute of <strong>Cancer</strong><br />

Research, London, United Kingdom; Royal Marsden Hospital,<br />

London, United Kingdom.<br />

PD01-06 Inhibition of the Ret receptor tyrosine kinase in combination<br />

with endocrine therapy impacts on migration and metastatic<br />

potential of estrogen receptor positive breast cancer models<br />

Hynes NE, Gattelli A, Nalvarte I, Roloff T. Friedrich Miescher Institute<br />

for Biomedical Research, Basel, Switzerland.<br />

PD01-07<br />

High throughput sequencing following cross-linked immuneprecipitation<br />

(HITS-CLIP) of Argonaute protein reveals novel<br />

miRNA regulatory pathways of estrogen receptor in breast<br />

cancer<br />

Kabos P, Kline E, Brown J, Flory K, Sartorius C, Hesselberth J, Pillai<br />

MM. University of Colorado Denver, Aurora, CO.<br />

www.aacrjournals.org<br />

7s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

PD01-08 Differences in estrogen receptor signaling in non-malignant<br />

primary ER-positive breast epithelial cells and breast cancer<br />

Lim E, He HH, Chi D, Yeung TY, Schnitt S, Liu SX, Garber J, Richardson<br />

A, Brown M. Dana-Farber <strong>Cancer</strong> Institute, Boston, MA; Harvard<br />

School of Public Health, Boston, MA; Harvard Medical School, Boston,<br />

MA; Beth Israel Deaconess Medical Center, Boston, MA; Brigham and<br />

Women’s Hospital, Boston, MA.<br />

5:00 pm–7:00 pm<br />

Poster Discussion 2: HER2<br />

Ballroom B<br />

Viewing 5:00 pm<br />

Discussion 5:15 pm<br />

Andrea Richardson, MD, PhD, Chair<br />

Dana-Farber <strong>Cancer</strong> Institute<br />

Boston, MA<br />

Marc Van de Vijver, MD, PhD, Discussant<br />

Academic Medical Center<br />

Amsterdam, NETHERLANDS<br />

and<br />

<strong>Antonio</strong> C. Wolff, MD, Discussant<br />

The Sidney Kimmel Comprehensive <strong>Cancer</strong> Center at<br />

Johns Hopkins<br />

Baltimore, MD<br />

PD02-01 Her2 expression measured by AQUA analysis on BCIRG-005<br />

and BCIRG-006 predicts the benefit of Herceptin therapy<br />

Christiansen J, Barakat N, Murphy D, Rimm DL, Dabbas B, Nerenberg<br />

M, Beruti S, Quinaux E, Hall J, Press M, Slamon D. Genoptix Medical<br />

Laboratory; Yale University; IDDI; University of Southern California;<br />

University of California, Los Angeles.<br />

PD02-02 The effect of HER2 expression on luminal A breast tumors<br />

Ellsworth RE, Valente AL, Shriver CD. Henry M. Jackson Foundation,<br />

Windber, PA; Windber Research Institute, Windber, PA; Walter Reed<br />

National Military Medical Center, Bethesda, MD.<br />

PD02-03 Added value of HER-2 amplification testing by multiplex<br />

ligation-dependent probe amplification (MLPA)<br />

van Slooten H-J, Kuijpers CC, Moelans CB, Horstman A, Al-Janabi<br />

S, Hinrichs JW, van Diest PJ, Jiwa M. Symbiant Pathology Expert<br />

Centre, Alkmaar, Netherlands; University Medical Centre Utrecht,<br />

Netherlands.<br />

PD02-04 Automated quantitative RNA in situ hybridization for<br />

resolution of equivocal and heterogeneous ERBB2 (HER2)<br />

status in invasive breast carcinoma<br />

Wang Z, Portier BP, Gruver AM, Bui S, Wang H, Su N, Vo H-T, Ma X-J,<br />

Luo Y, Budd GT, Tubbs RR. Cleveland Clinic, Cleveland, OH; Advanced<br />

Cell Diagnostics, Hayward, CA.<br />

PD02-05 Comparison of fluorescence in situ hybridization (FISH) and<br />

dual-ISH (D-ISH) in the determination of HER2 status<br />

Mansfield AS, Sakai Y, Walsh FJ, Wiktor AE, Sukov WR, Dogan A,<br />

Jenkins RB. Mayo Clinic, Rochester, MN.<br />

PD02-06 Concordance between immunohistochemistry and FISH<br />

(fluorescence in situ hybridization) & SISH(silver in situ<br />

hybridization) for assessment of the HER2<br />

Lee M-R, Cho S-H, Kim D-C, Lee K-C, Lee J-H, Kwon H-C, Lee H-W,<br />

Lee S-e. Dong-A University Medical Center, Busan, Korea.<br />

PD02-07 Next-generation sequencing of FFPE breast cancers<br />

demonstrates high concordance with FISH in calling<br />

HER2 amplifications and commonly detects other clinically<br />

relevant genomic alterations<br />

Lipson D, He J, Yelensky R, Miller V, Sheehan C, Brennan K, Jarosz M,<br />

Stephens P, Cronin M, Ross J. Foundation Medicine, Inc, Cambridge,<br />

MA; Albany Medical College, Albany, NY.<br />

5:00 pm–7:00 pm<br />

POSTER SESSION 1 & RECEPTION<br />

Exhibit Halls A-B<br />

Detection/Diagnosis: Axillary Staging and Sentinel Nodes<br />

P1-01-01 Fluorescence mapping with indocyanine green for sentinel<br />

lymph node detection in early breast cancer – results of the<br />

ICG-10 study<br />

Benson JR, Loh S-W, Jones LA, Wishart GC. Addenbrooke’s Hospital,<br />

Cambridge, Cambridgeshire, United Kingdom.<br />

P1-01-02 Withdrawn<br />

P1-01-03 Comparison of sentinel lymph node biopsy guided by the<br />

multi-modal method of indocyanine green fluorescence,<br />

radioisotope and blue dye versus the radioisotope in breast<br />

cancer; A randomized phase II trial<br />

Jung S-Y, Kim S-K, Kim SW, Lee S, Lee J, Shin I-s, Kwon Y, Lee E.<br />

National <strong>Cancer</strong> Center, Goyang, Korea.<br />

P1-01-04 Tc 99m Tilmanocept and Sulfur Colloid Injection: A<br />

comparison of preoperative imaging and intraoperative<br />

lymphatic mapping of breast cancer patients – Localization<br />

rate and degree of localization<br />

Cope FO, Metz WL, Hartman RD, Joy MT, Potter BM, Abbruzzese<br />

BC, Shuping JL, Blue MS, Christman LA, King DW. Navidea<br />

Biopharmaceuticals, Dublin, OH; STATKING Consulting, Inc.,<br />

Fairfield, OH.<br />

P1-01-05 The Efficacy of Arm Node Preserving Surgery Using Axillary<br />

Reverse Mapping for Preventing Lymphedema in Patients<br />

with <strong>Breast</strong> <strong>Cancer</strong>: The results from a 2-year follow-up<br />

Lee J-H, Son G-T, Choi J-E, Kang S-H, Lee S-J, Bae Y-K. Yeungnam<br />

University college of medicine, Daegu, Republic of Korea.<br />

P1-01-06 Evaluation of the stage IB designation of the 7th edition of the<br />

AJCC staging system: Biologic factors are more important<br />

Mittendorf EA, Ballman KV, McCall LM, Hansen N, Lucci A, Gabram<br />

S, Urist M, Crow J, Hurd T, Hunt KK, Giuliano AE. The University of<br />

Texas MD Anderson <strong>Cancer</strong> Center; Mayo Clinic; American College<br />

of Surgeons Oncology Group; Northwestern University; Emory<br />

University; University of Alabama Birmingham; University of Texas<br />

Health Science Center <strong>San</strong> <strong>Antonio</strong>; Cedars-Sinai Medical Center.<br />

P1-01-07 The Institut Curie Nomogram including HER2 status predicts<br />

additional axillary metastasis in breast cancer patients with a<br />

positive sentinel node biopsy: a multicentric validation<br />

Ngo C, De Rycke Y, Belichard C, Guilhen N, Doridot V, Rouzier R,<br />

Coutant C, Hudry D, Fritel X, Fourchotte V, Feron J-G, Pierga J-Y,<br />

Salomon A, Alran S. Institut Curie, Paris, France; Poitiers University<br />

Hospital, Poitiers, France; Centre de <strong>San</strong>té République, Clermont-<br />

Ferrand, France; Hopital Tenon, Paris, France; Centre Georges<br />

François Leclerc, Dijon, France.<br />

P1-01-08 A Nomogram for predicting two or less axillary lymph node<br />

involvement for breast cancer<br />

Ahn SK, Kim JS, Kim Mk, Lee JW, Kim T, Kim JY, Moon HG, Han W,<br />

Noh D-Y. Seoul National University College of Medicine, Seoul, Korea.<br />

P1-01-09 Which nomograms may be the best for predicting<br />

nonsentinel lymph node metastasis in breast cancer patients:<br />

a meta-analysis<br />

Chen K, Jin L, Zhu L, Shan Q, Su F. Sun Yat-sen Memorial Hopsital,<br />

Guangzhou, Guangdong, China.<br />

P1-01-10 Comparison of sentinel lymph node positivity rates pre and<br />

post introduction of OSNA molecular analysis<br />

Rusby J, Agabiti E, Waheed S, Barry P, Roche N, Allum W, Gui G,<br />

MacNeill F, Christaki G, Osin P, Nerurkar A. Royal Marsden NHS<br />

Foundation Trust, London, United Kingdom.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012 8s <strong>Cancer</strong> Research


December 4–8, 2012<br />

Program Schedule<br />

P1-01-11<br />

P1-01-12<br />

P1-01-13<br />

P1-01-14<br />

P1-01-15<br />

P1-01-16<br />

P1-01-17<br />

P1-01-18<br />

P1-01-19<br />

P1-01-20<br />

Is OSNA mRNA copy number in sentinel lymph node biopsy<br />

predictive of further disease in the axilla?<br />

Rusby JE, Agabiti E, Waheed S, Barry P, Roche N, Allum W, Gui G,<br />

MacNeill F, Christaki G, Osin P, Nerurkar A. Royal Marsden NHS<br />

Foundation Trust, London, United Kingdom.<br />

The Performance of the One Step Nucleic Acid Amplification<br />

(OSNA) Assay in <strong>Breast</strong> <strong>Cancer</strong> Patients with Receiving<br />

Preoperative Systemic Therapy<br />

Yagata H, Yamauchi H, Horii R, Osako T, Iwase T, Akiyama F, Kinoshita<br />

T, Tsuda H, Tsugawa K, Nakamura S. St. Luke’s International Hospital,<br />

Tokyo, Japan; <strong>Cancer</strong> Institute Hospital of the Japanese Foundation<br />

for <strong>Cancer</strong> Research, Tokyo, Japan; National <strong>Cancer</strong> Center Hospital,<br />

Tokyo, Japan; St. Marianna University School of Medicine, Kanagawa,<br />

Japan; Showa University School of Medicine, Tokyo, Japan.<br />

Patterns of definitive axillary management in the era prior to<br />

reporting ACOSOG Z0011: comparison between NCCN Centers<br />

and hospitals in Michigan<br />

Breslin T, Hwang S, Mamet R, Hughes M, Otteson R, Edge S, Moy B,<br />

Rugo H, Wong Y-N, Wilson J, Laronga C, Weeks J, Silver S, Marcom<br />

P. University of Michigan, Ann Arbor, MI; Duke University; City of<br />

Hope; Dana-Farber/Brigham and Women’s <strong>Cancer</strong> Center; Roswell<br />

Park <strong>Cancer</strong> Institute; University of California <strong>San</strong> Fransisco; Fox<br />

Chase <strong>Cancer</strong> Center; Ohio State University; Moffitt <strong>Cancer</strong> Center;<br />

Massachusetts General Hospital <strong>Cancer</strong> Center.<br />

Effects of axillary lymph node dissection on survival of<br />

patients with sentinel lymph node metastasis of breast cancer<br />

in the Surveillance, Epidemiology and End Results (SEER)<br />

database using a propensity score matching analysis<br />

Bendifallah S, Chereau E, Bezu C, Coutant C, Rouzier R. Tenon, Paris,<br />

France; Centre Leclerc, Dijon, France.<br />

Accuracy of sentinel lymph node in determining the<br />

requirement for second axillary surgeries in early breast<br />

cancer with retrospective application of the Z0011 criteria<br />

Waters PS, Fennessy PJ, Alazawi D, Sweeney KJ, Kerin MJ. National<br />

University of Ireland, Galway, Ireland.<br />

Intraoperatively-palpable “non-sentinel” nodes: should they<br />

be removed?<br />

Crivello ML, Ruth K, Sigurdson ER, Egleston BL, Boraas M, Bleicher RJ.<br />

Fox Chase <strong>Cancer</strong> Center, Philadelphia, PA.<br />

The impact of triple negativity on lymph node positivity in<br />

breast cancer<br />

Weber JJ, Bellin LS, Milbourn DE, Wong J. East Carolina University,<br />

Brody School of Medicine, Greenville, NC; University of North<br />

Carolina, Lineberger Comprehensive <strong>Cancer</strong> Center, Chapel Hill, NC.<br />

The results of 55 consecutive cases of intra-operative sentinel<br />

node analysis using real time polymerase chain reaction<br />

detection of cytokeratin 19 and mammoglobin expression<br />

(Metasin) to direct immediate axillary clearance<br />

Nadi K, Sharaiha Y, Sai-Girdhar P, Huws A, Khawaja S, Holt S. Prince<br />

Philip Hospital, Llanelli, United Kingdom.<br />

Prediction of axillary lymph node status in male breast<br />

carcinoma<br />

Vaysse C, Sroussi J, Mallon P, Feron J-G, Rivain A-L, Ngo C, Belichard<br />

C, Lasry S, Pierga J-Y, Couturaud B, Fitoussi A, Laki F, Fourchotte V,<br />

Alran S, Kirova YM, Vincent-Salomon A, Sastre-Garau X, Sigal-Zafrani<br />

B, Rouzier R, Reyal F. Institut Curie, Paris, France.<br />

Clinicopathlogic Analysis of Invasive <strong>Breast</strong> Carcinoma with<br />

Micropapillary Component<br />

Yamanaka T, Suganuma N, Yamanaka A, Kojima I, Nishiyama<br />

S, Mukaibashi T, Nakayama H, Matsuura H, Matsuzu K, Chiba A,<br />

Inaba M, Rino Y, Yoshida A, Shimizu S, Masuda M. Yokohama City<br />

University, Yokohama-City, Kanagawa, Japan; Kanagawa <strong>Cancer</strong><br />

Center, Yokohama-City, Kanagawa, Japan; Ito Hospital, Tokyo, Japan.<br />

P1-01-21<br />

P1-01-22<br />

P1-01-23<br />

P1-01-24<br />

P1-01-25<br />

P1-01-26<br />

P1-01-27<br />

P1-01-28<br />

Sentinel Lymph Node detection after previous breast tumour<br />

surgical resection: identification rate and false negative rate<br />

through a prospective multi institutional study<br />

Classe J-M, Andrieux N, Tunon de Lara C, Charitansky H, Lecuru F,<br />

Houpeau J-L, Faure C, De Blaye P, Houvenaeghel G, Kere D, Marchal<br />

F, Raro P, Lefebvre C, Dupré P-F, Rodier J-F. Institut de <strong>Cancer</strong>ologie<br />

de l’Ouest, Nantes Saint Herblain, France; Institut Bergonié, Bordeaux,<br />

France; Institut Claudius Regaud, Toulouse, France; Centre Hospitalier<br />

Georges Pompidou, Paris, France; Centre Oscar Lambret, Lille, France;<br />

Centre léon Bérard, Lyon, France; Centre Hospitalier Les Oudairies,<br />

La Roche sur Yon, France; Institut Paoli Calmettes, Marseille, France;<br />

Institut Jean Godinot, Reims, France; Centre Alexis Vautrin, Nancy,<br />

France; Centre Hospitalier Universitaire, Angers, France; Centre<br />

Hospitalier Morvan, Brest, France; Centre Paul Strauss, Strasbourg,<br />

France.<br />

The utility of axillary ultrasound and sentinel lymph node<br />

biopsy in the management of metaplastic breast carcinoma<br />

Hieken TJ, Fazzio RT, Reynolds C, Jones KN, Ghosh K, Glazebrook KN.<br />

Mayo Clinic, Rochester, MN.<br />

Increased Diagnostic Performance of Sentinel Lymph Node<br />

Biopsy Combined with Radiologic-pathologic Factors After<br />

Neoadjuvant Chemotherapy in <strong>Breast</strong> <strong>Cancer</strong> Patients with<br />

Cytologically Proven Node Metastasis at Diagnosis<br />

Park S, Koo JS, Kim MJ, Park JM, Cho JH, Hwang H, Park HS, Kim E-K,<br />

Kim SI, Park B-W. Yonsei University College of Medicine, Seoul, Korea.<br />

Which combinations are helpful to predict axillary<br />

lymph node metastasis in T1 breast cancer with<br />

ultrasonography and contrast-enhanced MRI and contrastenhanced<br />

18 F-FDG PET-CT?<br />

Hwang SO, Park HY, Jung JH, Kim WW, Lee YH, Lee JJ, Choi HH,<br />

Hwangbo SM. Kyungpook National University School of Medicine,<br />

Daegu, Korea; Hyosung Hospital, Daegu, Korea.<br />

Sentinel lymph node biopsy in breast cancer: The approach<br />

in day surgery under local anaesthesia for quality-of-life and<br />

significant cost reduction<br />

Ricci F, Capuano LG, Saralli E, Di Legge P, Violante A, Polistena A,<br />

Scala T, Pacchiarotti A, Cannas P, Cianni R, Fanelli G, Bellardini P, De<br />

Masi C. S.M. Goretti Hospital, Latina, Italy; LILT, Latina, Italy.<br />

Procoagulant biomarkers may direct axillary nodal surgery<br />

Shaker H, Bundred NJ, Glassey E, Kirwan CC. University Hospital<br />

of South Manchester, Manchester, United Kingdom; University of<br />

Manchester, United Kingdom.<br />

Novel diagnostic procedure of metastasis to the sentinel<br />

lymph node of breast cancer using a semi-dry dot-blot<br />

method<br />

Otsubo R, Oikawa M, Shibata K, Hirakawa H, Yano H, Matsumoto<br />

M, Hatachi T, Nakao K, Hayashi T, Abe K, Kinoshita N, Nakashima M,<br />

Taniguchi H, Omagari T, Itoyanagi N, Nagayasu T. Nagasaki University<br />

Hospital, Nagasaki, Japan; The Japanese Red Cross Nagasaki Genbaku<br />

Hospital, Nagasaki, Japan; Aiyuukai Memorial Hospital, Chiba, Japan;<br />

Nagasaki University Atomic Bomb Disease Institute, Nagasaki, Japan;<br />

St. Francis Hospital, Nagasaki, Japan.<br />

What is the actual impact of micrometastases in sentinel<br />

lymph nodes in regards to global survival and disease free<br />

survival<br />

Barbosa EM, Araujo Neto JT, Filho EC, Infante KP, Felzener MM,<br />

Matielle CR, Modesto MG, Basso R, Goes JCS. Instituto Brasileiro de<br />

Controle do <strong>Cancer</strong> - IBCC, Sao Paulo, Brazil.<br />

www.aacrjournals.org<br />

9s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P1-01-29 Intraoperative molecular analysis of sentinel lymph node as a<br />

new predictor of axillary status in early breast cancer patients<br />

Peg V, Espinosa-Bravo M, Vieites B, Vilardell F, Antúnez JR, <strong>San</strong>cho<br />

de Salas M, <strong>San</strong>sano I, Delgado Sánchez JJ, Pinto W, Gozalbo F,<br />

Petit A, Rubio I. Hospital Universitario Vall d’Hebron, Barcelona,<br />

Spain; Hospital Universitario Virgen del Rocío, Sevilla, Spain; Hospital<br />

Universitari Arnau de Vilanova de Lleida, Lleida, Spain; Complejo<br />

hospitalario Universitario <strong>San</strong>tiago de Compostela, <strong>San</strong>tiago de<br />

Compostela, Spain; Hospital Universitario de Salamanca, Salamanca,<br />

Spain; Hospital Universitario 12 de Octubre, Madrid, Spain; Hospital<br />

Universitario e Gran Canaria Doctor Negrín, Las Palmas de Gran<br />

Canaria, Spain; Instituto Valenciano de Oncología (IVO), Valencia,<br />

Spain; Hospital Universitari de Bellvitge, Hospitalet de Llobregat,<br />

Spain.<br />

Detection/Diagnosis: Biopsy Techniques<br />

P1-02-01 Flat epithelial atypia diagnosed on breast core biopsy:<br />

what next?<br />

Plichta JK, Lapetino S, Rumas N, Rajan P, Godellas C, Perez C. Loyola<br />

University Medical Center, Maywood, IL.<br />

P1-02-02 Receptor discordance in breast cancer recurrence: Is re-biopsy<br />

a necessity?<br />

Fujita T, Sawaki M, Hattori M, Kondo N, Horio A, Ushio A, Gondo N,<br />

Hiroji I. Aichi <strong>Cancer</strong> Center Hospital, Nagoya, Japan.<br />

Tumor Cell and Molecular Biology: Etiology/Carcinogenesis<br />

P1-03-01 Evidence for the Warburg effect in mammary atypia from<br />

high-risk African American women<br />

Seewaldt V, Hoffman A, Ibarra-Drendall C. Duke University,<br />

Durham, NC.<br />

P1-03-02 ‘Normal’ tissue adjacent to breast cancer is not normal<br />

Clare SE, Pardo I, Mathieson T, Lillemoe HA, Blosser RJ, Choi M,<br />

Sauder CAM, Doxey DK, Badve S, Storniolo AMV, Atale R, Radovich<br />

M. Indiana University School of Medicine, Indianapolis, IN; Susan<br />

G. Komen for the Cure Tissue Bank at the IU Simon <strong>Cancer</strong> Center,<br />

Indiana University School of Medicine, Indianapolis, IN.<br />

P1-03-03 Milk deposition in women’s mammary duct has been a<br />

potential risk factor of breast tumor<br />

Xu Z, Gu Y, Gong G, Zhang Y. <strong>Breast</strong> Disease Institution of Jilin<br />

Province, Changchun, Jilin, China; Spinal Disease Institution of Jilin<br />

Province, Changchun, Jilin, China.<br />

Tumor Cell and Molecular Biology: Oncogenes/Tumor Suppressor Genes<br />

P1-04-01 Loss of Notch4 reduces mammary tumorigenesis by MYC and<br />

activated KRAS<br />

Rodriguez EM, Bishop JM. G. W. Hooper Research Foundation,<br />

University of California, <strong>San</strong> Francisco, CA.<br />

P1-04-02 The role of the mTORC1/S6K1 signaling pathway in ER-positive<br />

breast cancer<br />

Holz MK, Unger HA, Sedletcaia A. Stern College for Women of<br />

Yeshiva University; Albert Einstein College of Medicine.<br />

P1-04-03 Knocking down Suppressor of Cytokine Signaling 7 in<br />

breast cancer: The role in Insulin-like Growth Factor - I /<br />

Phospholipase Cγ-1 signaling<br />

Sasi W, Ye L, Jiang WG, Mokbel K, Sharma A. St George’s Hospital<br />

Medical School, University of London, United Kingdom; Cardiff<br />

University School of Medicine, Cardiff, Wales, United Kingdom; The<br />

London <strong>Breast</strong> Institute, The Princess Grace Hospital, London, United<br />

Kingdom.<br />

P1-04-04 KSR1 is involved in functional interaction between p53 and<br />

BRCA1 and is an independent predictor of overall survival in<br />

breast cancer<br />

Zhang H, Lombardo Y, Filipovic A, Periyasamy M, Coombes RC,<br />

Stebbing J, Giamas G. Imperial College London, United Kingdom.<br />

P1-04-05 Role of the Rb and p53 Tumor Suppressor Pathways in<br />

Mammary Tumorigenesis<br />

Jones RA, Liu JC, Zhe J, Schimmer AA, Eldad Z. University Health<br />

Network, Toronto, ON, Canada.<br />

P1-04-06 Insertional mutagenesis identifies HACE1 as a HER2/Neu<br />

Cooperating <strong>Breast</strong> <strong>Cancer</strong> Tumor Suppressor Gene<br />

Goka E, Miller P, Baker K, Stark G, Lippman ME. University of Miami<br />

Miller School of Medicine, Miami, FL; Cleveland Clinic, Cleveland, OH;<br />

University of Miami, FL.<br />

P1-04-07 The mRNA Expression of DAP1 in Human <strong>Breast</strong> <strong>Cancer</strong>:<br />

Correlation with Clinicopathological Parameters<br />

Wazir U, Jiang WG, Sharma AK, Mokbel K. St Georges’ Healthcare<br />

NHS Trust, London, United Kingdom; Cardiff University-Peking<br />

University Oncology Joint Institute, Cardiff, United Kingdom; The<br />

London <strong>Breast</strong> Institute, The Princess Grace Hospital, London, United<br />

Kingdom.<br />

P1-04-08 Evidence for anti-apoptosis function of GNB1 in human<br />

breast cancer<br />

Wazir U, Kasem A, Sharma AK, Jiang W, Mokbel K. The London <strong>Breast</strong><br />

Institute, The Princess Grace Hospital, London, United Kingdom;<br />

Cardiff University-Peking University Oncology Joint Institute, Cardiff,<br />

United Kingdom; St Georges’ Healthcare NHS Trust, London, United<br />

Kingdom.<br />

P1-04-09 mTORC1 and Rictor expression in human breast cancer:<br />

correlations with clinicopathological parameters and disease<br />

outcome<br />

Wazir U, Kasem A, Sharma AK, Jiang WG, Mokbel K. The London<br />

<strong>Breast</strong> Institute, The Princess Grace Hospital, London, United<br />

Kingdom; Cardiff University-Peking University Oncology Joint<br />

Institute, Cardiff, United Kingdom; St Georges’ Healthcare NHS Trust,<br />

London, United Kingdom.<br />

P1-04-10 PDGFRA signaling in Inflammatory breast cancer<br />

Joglekar M, van Golen K. University of Delaware, Newark, DE.<br />

P1-04-11 EZH2 Expands <strong>Breast</strong> Stem Cells via NOTCH Signaling, Acting<br />

to Accelerate <strong>Breast</strong> <strong>Cancer</strong> Initiation<br />

Kleer CG, Li X, Moore HM, Toy KA, Gonzalez ME. University of<br />

Michigan, Ann Arbor, MI.<br />

Tumor Cell and Molecular Biology: Tumor Progression, Invasion, and<br />

Metastasis<br />

P1-05-01 Chemokine-mediated nuclear translocation and novel nuclear<br />

role for LIM and SH3 Protein-1(LASP-1) in breast cancer<br />

Raman D, Duvall-Noelle NL, Richmond A. Vanderbilt University<br />

Medical Center, Nashville, TN.<br />

P1-05-02 Epithelial-to-epithelial transition precedes collective breast<br />

cancer invasion<br />

Cheung KJ, Ewald AJ. Johns Hopkins School of Medicine,<br />

Baltimore, MD.<br />

P1-05-03 A requirement for neural precursor cell-expressed<br />

developmentally downregulated gene 9 during the initiation<br />

of mammary tumorigenesis in MMTV-neu mice<br />

Serzhanova VA, Little JL, Izumchenko E, Seo S, Kurokawa M,<br />

Egleston B, Klein-Szanto AA, Golemis EA. Fox Chase <strong>Cancer</strong> Center,<br />

Philadelphia, PA; Ben Gurion University of the Negev, Beer Sheva,<br />

Israel; University of Tokyo, Japan.<br />

P1-05-04 Expression of Quiescin Sulfhydryl Oxidase 1 is associated<br />

with a highly invasive phenotype and correlates with a poor<br />

prognosis in Luminal B breast cancer<br />

Katchman BA, Ocal T, Hostetter G, Cunliffe HE, Wantanabe A, Lake<br />

DF. Arizona State University, Tempe, AZ; Mayo Clinic Arizona,<br />

Scottsdale, AZ; Translational Genomic Research Institute,<br />

Phoenix, AZ.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012 10s <strong>Cancer</strong> Research


December 4–8, 2012<br />

Program Schedule<br />

P1-05-05<br />

Podocalyxin is a key regulator of breast cancer progression<br />

and metastasis<br />

Snyder KA, Hughes MR, Graves M, Roskelly C, McNagny KM.<br />

University of British Columbia, Vancouver, BC, Canada.<br />

A novel mutation in the tyrosine kinase domain of ErbB2:<br />

molecular and proteomic investigation of its role in breast<br />

cancer invasion<br />

O’Hara J, Kast J. University of British Columbia, Vancouver, BC,<br />

Canada.<br />

Prognostic relevance of Claudin-2 expression in metastatic<br />

breast cancer<br />

Hedenfalk I, Kimbung S, Kovacs A, Skoog L, Einbeigi Z, Walz T,<br />

Malmberg M, Loman N, Fernö M, Hatschek T, TEX Study Group.<br />

Lund University, Lund, Sweden; CREATE Health Strategic Center<br />

for Translational <strong>Cancer</strong> Research, Lund University, Lund, Sweden;<br />

Sahlgrenska University Hospital, Gothenburg, Sweden; Karolinska<br />

University Hospital, Solna, Sweden; Linköping University Hospital,<br />

Linköping, Sweden; Helsingborg General Hospital, Helsingborg,<br />

Sweden.<br />

Targeting integrin signaling suppresses invasive recurrence<br />

in a three-dimensional model of radiation treated ductal<br />

carcinoma in situ<br />

Nam J-M, Ahmed KM, Costes S, Zhang H, Sabe H, Shirato H, Park<br />

CC. Hokkaido University Graduate School of Medicine, Sapporo,<br />

Hokkaido, Japan; Ernest Orlando Lawrence Berkeley National<br />

Laboratory, Berkeley, CA; UCSF, <strong>San</strong> Francisco, CA.<br />

FH535 inhibited migration and growth of breast cancer cells<br />

Dorchak JA, Iida J, Clancy R, Luo C, Chen Y, Hu H, Mural RJ, Shriver CD.<br />

Windber Research Institute, Windber, PA; Walter Reed National Military<br />

Medical Center, Bethesda, MD.<br />

Targeting breast cancer metastasis through disruption of<br />

novel PELP1-G9a complex<br />

Mann M, Chakravarty D, Kim CA, Vadlamudi RK. University of Texas<br />

Health Science Center at <strong>San</strong> <strong>Antonio</strong>, TX; Weill Cornell Medical<br />

College, New York, NY.<br />

Biological characterization of tumor-associated leukocytes in<br />

positive and negative lymph node breast cancer patients<br />

ElGhonaimy EA, El-Shinawi M, Abd-El-Tawab R, El Mamlouk T, Sloane<br />

BF, Mohamed MM. Faculty of Science, Cairo University, Giza, Cairo,<br />

Egypt; Faculty of Medicine, Ain Shams University, Cairo, Egypt;<br />

Wayne State University, Detroit, MI.<br />

Metastatic xenograft models of human estrogen receptor<br />

negative breast cancer primary cultures are driven by the<br />

recruitment of myeloid-derived suppressor cells<br />

Drews-Elger K, Iorns E, Brinkman JA, Berry DL, Lippman ME, El-Ashry<br />

D. Sylvester Comprehensive <strong>Cancer</strong> Center, Braman Family <strong>Breast</strong><br />

<strong>Cancer</strong> Institute, University of Miami, FL; Science Exchange, Palo Alto,<br />

CA; Georgetown University Medical Center, Washington, DC.<br />

CLIC3 is associated with invasive behaviour and poorer<br />

prognosis in estrogen receptor-negative breast cancer<br />

Macpherson IR, Dozynkiewicz M, Kalna G, Speirs C, Chaudhary S,<br />

Edwards J, Timpson P, Norman J. University of Glasgow, United<br />

Kingdom; Beatson Institute for <strong>Cancer</strong> Research, Glasgow, United<br />

Kingdom.<br />

Multiscale computational modeling of breast cancer invasion:<br />

Towards a predictive patient-based tool<br />

Trucu D, Thompson A, Chaplain MAJ. University of Dundee, United<br />

Kingdom; Ninewells Hospital, University of Dundee, United Kingdom.<br />

P1-05-15<br />

Identification of a novel Hypoxia Inducible Factor-1 regulated<br />

gene involved in breast cancer growth and metastasis<br />

Peacock DL, Schwab LP, Seagroves TN. University of Tennessee<br />

Health Science Center, Memphis, TN.<br />

Sushi Domain Containing 2 (SUSD2): a plasma membrane<br />

protein that increases immune evasion in breast<br />

tumorigenesis<br />

Watson AP, Davis EM, Egland KA. <strong>San</strong>ford Research/USD,<br />

Sioux Falls, SD.<br />

S100A7/RAGE axis enhances breast cancer growth by<br />

activating Stat3 signaling<br />

Nasser MW, Wani NA, Qamri Z, Ahirwar D, Powell CA, Ganju RK. The<br />

Ohio State University, Columbus, OH.<br />

Determining the molecular mechanism of the breast cancerinduced<br />

brain metastasis and a role of a novel pan-TGF-β<br />

inhibitor as a potential therapy for brain metastasis in a<br />

mouse xenograft model<br />

De Mukhopadhyay K, Elkahloun AG, Hinck AP, Yoon K, Cornell JE,<br />

Shu L, Yang J, Sun L. University of Texas Health Science Center,<br />

<strong>San</strong> <strong>Antonio</strong>, TX; National Human Genome Research Institute-NIH,<br />

Bethesda, MD.<br />

Modeling <strong>Cancer</strong> Recurrence and the Therapeutic Effect of<br />

Adjuvant Systemic Therapy<br />

Ganesan S, Bhanot G, Boemo M, Lee JH. <strong>Cancer</strong> Institute of New<br />

Jersey- UMDNJ, New Brunswick, NJ; Rutgers University,<br />

VPiscataway, NJ.<br />

Fluorescent Hyaluronan Probes Distinguish Heterogeneous<br />

<strong>Breast</strong> <strong>Cancer</strong> Cell Subsets and Predict their Invasive Behavior<br />

Veiseh M, Kwon DH, Borowsky AD, Toelg C, Leong H, Lewis J, Turley<br />

EA, Bissell MJ. Lawrence Berkeley National Laboratories, Berkeley,<br />

CA; University of California Davis, Davis, CA; London Health Sciences<br />

Centre-London Regional <strong>Cancer</strong> Program; University of Western<br />

Ontario, London, ON, Canada.<br />

The role of epsin in promoting Epithelial-Mesenchymal<br />

Transition and metastasis by activating NF-kB signaling in<br />

breast cancer<br />

Cai X, Brophy ML, Hahn S, McManus J, Chang B, Pasula S, Chen<br />

H. Oklahoma Medical Research Foundation, Oklahoma City, OK;<br />

University of Oklahoma Health Science Center, Oklahoma City, OK.<br />

<strong>Breast</strong> cancer cells that undergo an Epithelial-to-Mesenchymal<br />

transition co-opt LPP, a regulator of mesenchymal cell<br />

migration and invasion<br />

Ngan E, Northey JJ, Ursini-Siegel J, Siegel PM. McGill University,<br />

Montreal, QC, Canada; Lady Davis Institute for Medical Research,<br />

Montreal, QC, Canada.<br />

Blockade of mTORC1 decreases CXCR4-mediated migration<br />

and metastasis<br />

Dillenburg Pilla P, Patel V, Dorsam RT, Masedunskas A,<br />

Amornphimoltham P, Weigert R, Molinolo AA, Gutkind JS. NIH,<br />

Bethesda, MD.<br />

Pharmacologic reversion of epigenetic silencing of the PRKD1<br />

promoter blocks breast tumor cell invasion and metastasis<br />

Borges S, Doppler H, Andorfer CA, Perez EA, Sun Z, Anastasiadis PZ,<br />

Thompson AE, Geiger XJ, Storz P. Mayo Clinic, Jacksonville, FL; Mayo<br />

Clinic, Rochester, MN.<br />

P1-05-06<br />

P1-05-16<br />

P1-05-07<br />

P1-05-08<br />

P1-05-09<br />

P1-05-10<br />

P1-05-11<br />

P1-05-12<br />

P1-05-13<br />

P1-05-14<br />

P1-05-17<br />

P1-05-18<br />

P1-05-19<br />

P1-05-20<br />

P1-05-21<br />

P1-05-22<br />

P1-05-23<br />

P1-05-24<br />

www.aacrjournals.org<br />

11s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Tumor Cell and Molecular Biology: Angiogenesis<br />

P1-06-01 Upregulation of metabolism as a potential resistance<br />

mechanism to bevacizumab in primary breast cancer<br />

Mehta S, Hughes NP, Adams RF, Li SP, Han C, Kaur K, Taylor NJ,<br />

Padhani AR, Makris A, Buffa FM, Harris AL. Weatherall Institute of<br />

Molecular Medicine, Oxford, United Kingdom; Stanford University,<br />

Stanford, CA; Oxford University Hospitals NHS Trust, Oxford, United<br />

Kingdom; Mount Vernon <strong>Cancer</strong> Centre, Northwood, Middlesex,<br />

United Kingdom; <strong>Cancer</strong> and Haematology Centre, Churchill<br />

Hospital, Oxford, United Kingdom; Paul Strickland Scanner Centre,<br />

Mount Vernon Hospital, Northwood, Middlesex, United Kingdom.<br />

P1-06-02 A newly angiogenic biomarker Vasohibin-1 expression in<br />

ductal carcinoma in situ of the breast<br />

Tamaki K, Tamaki N, Kamada Y, Uehara K, Miyashita M, Ishida T,<br />

Ohuchi N, Sasano H. Nahanishi Clinic Okinawa, Naha, Okinawa,<br />

Japan; Tohoku University Graduate School of Medicine, Sendai,<br />

Miyagi, Japan; Tohoku University Hospital, Sendai, Miyagi, Japan.<br />

P1-06-03 Microvessel density as determined by computerized image<br />

analysis of CD34 and CD105 expression correlates with poor<br />

outcome in triple-negative breast cancer<br />

De Brot M, Rocha RM, Soares FA, Gobbi H. Universidade Federal de<br />

Minas Gerais, Belo Horizonte, MG, Brazil; Hospital A.C. Camargo, Sao<br />

Paulo, SP, Brazil.<br />

Prognostic and Predictive Factors: Biomarkers - Methods<br />

P1-07-01 Attainment of extremely high concordance rates between<br />

fluorescence in situ hybridization and immunohistochemistry<br />

in testing for human epidermal growth factor receptor 2<br />

(HER2) in breast cancer using a normalized scoring system for<br />

immunohistochemistry: A four year experience with over<br />

9000 cases<br />

Gown AM, Goldstein LC, Tse CH, Hwang HC, Kandalaft PL. PhenoPath<br />

Laboratories, Seattle, WA.<br />

P1-07-02 Withdrawn<br />

P1-07-03 Quantification of HER2 expression at the single cell level<br />

and HER2 intratumoral heterogeneity of breast cancer tissue<br />

samples using automated image analysis<br />

Geretti E, Paragas V, Onsum M, Kudla A, Moulis S, Luus L, Wickham T,<br />

McDonagh C, MacBeath G, Hendriks B. Merrimack Pharmaceuticals,<br />

Cambridge, MA.<br />

P1-07-04 Comparison of HER2 expression by immunohistochemistry<br />

(IHC) using automated imaging system and fluorescence in<br />

situ hybridization (FISH). A retrospective analysis of<br />

2853 cases<br />

Collins R, Xiang D, Christie A, Leitch M, Euhus D, Rao R, Haley B,<br />

Sarode V. University of Texas Southwestern Medical Center,<br />

Dallas, TX.<br />

P1-07-05 Evaluation of tissue processing factors affecting HER2 IHC<br />

staining intensity in breast cancer cell lines<br />

Jensen K, Erickson J, Webster S, Pedersen HC. Dako Denmark,<br />

Glostrup, Denmark; Dako North America, Carpinteria, CA.<br />

P1-07-06 Effect of biospecimen variables on proteomic biomarker<br />

assessment in breast cancer<br />

Meric-Bernstam F, Akcakanat A, Chen H, Sahin A, Tarco E, Carkaci<br />

S, Adrada B, Singh G, Anh-Do K, Garces Z, Mittendorf EA, Babiera G,<br />

Wagner J, Bedrosian I, Krishnamurthy S, Symmans WF, Gonzalez-<br />

Angulo AM, Mills G. UT MD Anderson <strong>Cancer</strong> Center, Houston, TX;<br />

Ohio State University, Columbus, OH.<br />

P1-07-07 Inflammatory gene expression variations in the interval<br />

between core needle biopsy and excisional biopsy in early<br />

breast cancer<br />

Jeselsohn RM, Regan MM, Werner L, Fatima A, He HH, Brown M,<br />

Iglehart JD, Richardson AL, Come S. Beth Israel Deaconess Medical<br />

Center, Boston, MA; Dana Farber <strong>Cancer</strong> Institute, Boston, MA;<br />

Brigham and Women’s Hospital, Boston, MA.<br />

P1-07-08 Effect of sample preservation method and transportation<br />

duration on tumor gene expression profiling in breast cancer<br />

Fumagalli D, Jose V, Salgado R, Majjaj S, Singhal S, Vincent D,<br />

Maetens M, Larsimont D, Symmans F, Dinh P, Piccart M, Michiels<br />

S, Sotiriou C, Loi S. Institut Jules Bordet, Brussels, Belgium; <strong>Breast</strong><br />

International Group (BIG), Brussels, Belgium; The University of Texas<br />

MD Anderson <strong>Cancer</strong> Center, Houston, TX.<br />

P1-07-09 Estrogen receptor positivity: 10% or 1%?<br />

Yi M, Huo L, Koenig KB, Mittendorf EA, Meric-Bernstam F, Kuerer HM,<br />

Bedrosian I, Symmans WF, Hortobagyi GN, Crow JR, Shah RR, Hunt<br />

KK. University of Texas MD Anderson <strong>Cancer</strong> Center, Houston, TX.<br />

P1-07-10 Comparison of three commercial ER/PR assays on a single<br />

clinical outcome series<br />

Kornaga EN, Klimowicz AC, Konno M, Guggisberg N, Ogilvie T,<br />

Cartun RW, Morris DG, Webster MA, Magliocco AM. Alberta Health<br />

Services, Calgary, AB, Canada; Calgary Laboratory Services, Calgary,<br />

AB, Canada; Hartford Hospital, Hartford, CT; University of Calgary, AB,<br />

Canada; H. Lee Moffitt <strong>Cancer</strong> Center & Research Institute, Tampa, FL.<br />

P1-07-11 Characterization of progesterone receptor biomarker for<br />

predicting antiprogestin activity in human cancers<br />

Bonneterre J, Bosq J, Lange C, Gilles E. Centre Oscar Lambret, France;<br />

Institut Gustave Roussy, France; University of Minnesota; Invivis<br />

Pharmaceuticals.<br />

P1-07-12 Using Natural Language Processing to Identify and Extract<br />

HER2 Value from a large EMR system<br />

Zheng C, Avila C, Haque R. Kaiser Permanente Southern California,<br />

Pasadena, CA.<br />

P1-07-13 Prognostic relevance of statistically standardized estrogen<br />

receptor (ER), progesterone receptor (PR), and human<br />

epidermal growth factor receptor 2 (HER2) in tamoxifen(TAM)-<br />

treated NCIC CTG MA.14 patients<br />

Chapman J-AW, Sgroi D, Goss PE, Richardson E, Binns SN, Zhang Y,<br />

Schnabel CA, Erlander MG, Pritchard KI, Han L, Shepherd LE, Pollak<br />

MN. NCIC Clinical Trials Group, Queen’s University, Kingston, ON,<br />

Canada; Harvard University, Boston, MA; bioTheranostics, Inc., <strong>San</strong><br />

Diego, CA; Sunnybrook Odette <strong>Cancer</strong> Centre, University of Toronto,<br />

ON, Canada; Jewish General Hospital, McGill University, Montreal,<br />

QC, Canada.<br />

P1-07-14 Enabling biomarker validation in breast cancer molecular<br />

subtypes: sensitivity and specificity of array-based subtype<br />

classification in 983 patients<br />

Györffy B, Lanczky A. Semmelweis University.<br />

P1-07-15 Revisiting chromosome 17q copy number aberrations in early<br />

high-risk breast cancer<br />

Kotoula V, Bobos M, Eleftheraki AG, Timotheadou E, Razis E, Goussia<br />

A, Levva S, Kalogeras KT, Pectasides D, Fountzilas G. Hellenic<br />

Cooperative Oncology Group (HeCOG), Athens, Greece.<br />

P1-07-16 Liver derived epithelial cells as source of false positive<br />

circulating tumor cells in early breast cancer<br />

Habets L, Körber W, Frenken B, Danaei M, Kusche M, Peisker U, Kroll<br />

T, Pachmann K. Metares.e.V, Aachen, NRW, Germany; Brustzentrum<br />

Aachen Kreis Heinsberg, Aachen, NRW, Germany; Medizinische<br />

Universitätsklinik Jena, Thueringen, Germany.<br />

P1-07-17 V Array: A novel tool for constructing virtual tissue<br />

microarrays (TMAs), an evaluation of its use in optimizing TMA<br />

construction for Ductal Carcinoma in Situ (DCIS)<br />

Quintayo MA, Starczynski J, Yan FJ, Bartlett JMS, Benko L, Hanna W,<br />

Nofech-Mozes S, Rakovitch E. Ontario Institute of <strong>Cancer</strong> Research,<br />

Toronto, ON, Canada; Sunnybrook Health Sciences Centre, Toronto,<br />

ON, Canada; Leica Microsystems, Buffallo Grove, IL.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012 12s <strong>Cancer</strong> Research


December 4–8, 2012<br />

Program Schedule<br />

P1-07-18 Association between Bone Turnover Markers in patients<br />

with breast cancer and bone metastases on treatment with<br />

bisphosphonates (ZOMAR study)<br />

Tusquets I, De la Piedra C, Manso L, Crespo C, Gómez P, Calvo L,<br />

Ruiz M, Martínez P, Perelló A, Antón A, Codes M, Margelí M, Murias<br />

A, Salvador J, Seguí MA, De Juán A, Gavilá J, Luque M, Pérez D,<br />

Zamora P, Arizcum A, Chacón JI, Heras L, Barnadas A. Hospital<br />

del Mar, Barcelona, Spain; Instituto de Investigación <strong>San</strong>itaria<br />

Fundación Jiménez Díaz, Madrid, Spain; Hospital 12 de Octubre,<br />

Madrid, Spain; Hospital Universitario Ramón y Cajal, Madrid, Spain;<br />

Hospital Universitario Vall d’Hebrón, Barcelona, Spain; Hospital<br />

Universitario A Coruña Juan Canalejo, A Coruña, Spain; Hospital<br />

Universitario Virgen del Rocío, Sevilla, Spain; Hospital de Basurto,<br />

Vizcaya, Spain; Hospital Son Dureta, Palma de Mallorca, Spain;<br />

Hospital Miguel Servet, Zaragoza, Spain; Hospital Virgen Macarena,<br />

Sevilla, Spain; H. Universitario Trias y Pujol, Barcelona, Spain;<br />

Hospital Universitario Insular de Gran Canaria, Gran Canaria, Spain;<br />

Hospital Nuestra Señora de Valme, Sevilla, Spain; Hospital Parc Taulí<br />

Sabadell, Barcelona, Spain; Hospital Marqués Valdecilla, <strong>San</strong>tander,<br />

Spain; Instituto Valenciano de Oncología, Valencia, Spain; Hospital<br />

General de Asturias, Oviedo, Spain; Hospital Costa del Sol, Málaga,<br />

Spain; Hospital La Paz, Madrid, Spain; Hospital Palencia Río Carrión,<br />

Palencia, Spain; Hospital Virgen de la Salud, Toledo, Spain; Hospital<br />

Cruz Roja Hospitalet del Llobregat, Barcelona, Spain; Hospital <strong>San</strong>ta<br />

Creu i <strong>San</strong>t Pau, Barcelona, Spain.<br />

P1-07-19 Mass Spectrometry Based Quantitative Analysis of the HER<br />

Family receptors in FFPE <strong>Breast</strong> <strong>Cancer</strong> Tissue<br />

Hembrough TA, Scaltriti M, Serra V, Jimenez J, Perez J, Liao W-L,<br />

Thyparambil S, Cortes J, Baselga J, Burrows J. OncoPlex Diagnostics,<br />

Inc., Rockville, MD; Vall d’Hebron Istitute of Oncology, Barcelona,<br />

Spain; Massachusetts General Hospital, Boston, MA.<br />

Epidemiology, Risk, and Prevention: Prevention - Preclinical Studies and<br />

Model Systems<br />

P1-08-01 Effects Of An Allosteric AKT Inhibitor (MK2206)<br />

Administered With Or Without The Aromatase Inhibitor<br />

Vorozole In An ER + Rat Mammary <strong>Cancer</strong> Model: Preventive<br />

And Therapeutic Effects<br />

Lubet RA, Ellis MJ, Grubbs CJ. National <strong>Cancer</strong> Institutes, Bethesda,<br />

MD; University of Washington at Saint Louis, MO; University of<br />

Alabama at Birmingham, AL.<br />

P1-08-02 Gene expression changes in methylnitrosourea (MNU)-induced<br />

ER + mammary cancers following short-term treatment of rats<br />

with SERMs (Tamoxifen and Arzoxifene)<br />

Lubet R, Vedell P, Grubbs C, Bernard P, You M. National <strong>Cancer</strong><br />

Institute, Bethesda, MD; Medical School of Wisconsin, Milwaukee, WI;<br />

Hunstman <strong>Cancer</strong> Center, Salt Lake City, UT; University of Alabama at<br />

Birmingham, AL.<br />

Epidemiology, Risk, and Prevention: Prevention - Clinical Trials<br />

P1-09-01 Long-term effect of tamoxifen use on the risk of contralateral<br />

breast cancer<br />

Mellemkjær L, Steding-Jessen M, Frederiksen K, Andersson M, Olsen<br />

JH. Danish <strong>Cancer</strong> Society Research Center, Copenhagen, Denmark;<br />

Copenhagen University Hospital, Rigshospitalet, Copenhagen,<br />

Denmark.<br />

P1-09-02 Pilot study of a 1-year intervention of high-dose vitamin D in<br />

women at high risk for breast cancer<br />

Sivasubramanian PS, Hershman DL, Maurer M, Kalinsky K, Feldman<br />

S, Brafman L, Refice S, Kranwinkle G, Crew KD. Columbia University,<br />

New York, NY.<br />

P1-09-03 Chemoprevention in patients with newly diagnosed<br />

breast cancers<br />

Refinetti AP, Chun J, Schnabel F, Guth A, Axelrod D. NYU Langone<br />

Medical Center, New York, NY.<br />

P1-09-04 Down-regulation of trefoil protein 1(TFF1) in normal breast<br />

tissue of postmenopausal women at increased risk for breast<br />

cancer on exemestane<br />

Gatti-Mays M, Kallakury BVS, Makariou E, Venzon D, Permaul<br />

E, Isaacs C, Cohen P, Warren R, Gallagher A, Eng-Wong J.<br />

Georgetown University Hospital, Washington, DC; National <strong>Cancer</strong><br />

Institute, National Institutes of Health, Bethesda, MD; Lombardi<br />

Comprehensive <strong>Cancer</strong> Center, Georgetown University Medical<br />

Center, Washington, DC.<br />

P1-09-05 The RAZOR trial: a phase II prevention trial of screening<br />

plus goserilin and raloxifene versus screening alone in premenopausal<br />

women at increased risk of breast cancer<br />

Motion J, Ashcroft L, Dowsett M, Cuzick J, Hickman J, Evans G,<br />

Eccles D, Eeles R, Greenhalgh R, Affen J, Bundred S, Boggis C,<br />

Sergeant J, Fallowfield L, Adams J, Howell A. University Hospital<br />

South Manchester, Manchester, United Kingdom; The Christie NHS<br />

Foundation Trust, Manchester, United Kingdom; Royal Marsden<br />

Hospital, London, United Kingdom; Queen Mary, University of<br />

London, United Kingdom; Princess Anne Hospital, Southampton,<br />

United Kingdom; The Institute of <strong>Cancer</strong> Research, Royal Marsden<br />

Hospital, London, United Kingdom; University of Sussex - (SHORE-C),<br />

Brighton, United Kingdom; University of Manchester, United<br />

Kingdom; Manchester Royal Infirmary, Manchester, United Kingdom.<br />

P1-09-06 Biological Effects of Green Tea Capsule Supplementation in<br />

Pre-surgery <strong>Breast</strong> <strong>Cancer</strong> Patients<br />

Yu S, Spicer D, Hawes D, Wu A. University of Southern California, Los<br />

Angeles, CA.<br />

P1-09-07 Topical 4-OHT trial in women with DCIS of the breast: Vreport<br />

of plasma and breast tissue concentration of tamoxifen<br />

metabolites<br />

Lee O, Chatterton RT, Muzzio M, Page K, Jovanovic B, Helenowski I,<br />

Dunn B, Heckman-Stoddard B, Foster K, Shklovskaya J, Skripkauskas<br />

S, Bergan R, Khan SA. Northwestern University, Chicago, IL; IIT<br />

Research Institute, Chicago, IL; National <strong>Cancer</strong> Institute,<br />

Bethesda, MD.<br />

Epidemiology, Risk, and Prevention: Prevention - Nutritional Studies<br />

P1-10-01 Curcumin suppresses MMP-9 expression via inhibition of<br />

PKCα/MAPKs and NF-κB/AP-1 activation in MCF-7 cells<br />

Kim SK, Kim YW, Youn HJ, Jung SH. Chonbuk National University<br />

Medical School, Jeonju, Jeollabukdo, Republic of Korea.<br />

P1-10-02 Comparative Preventive Efficacy of Select Chinese Herbs in<br />

<strong>Breast</strong> Carcinoma Derived Isogenic Cells with Modulated<br />

Estrogen Receptor Functions<br />

Telang NT, Li G, Katdare M, Sepkovic DW, Bradlow HL, Wong GYC.<br />

Palindrome Liaisons, Montvale, NJ; American Foundation for Chinese<br />

Medicine, New York, NY; Skin of Color Research Institute, Leroy T.<br />

Canoles Jr. <strong>Cancer</strong> Research Center, Hampton University, Eastern<br />

Virginia Medical School, Hampton, VA; David & Alice Jurist Institute<br />

for Research, Hackensack University Medical Center, Hackensack, NJ;<br />

David & Alice Jurist Institute for Research, Hackensack, NJ; American<br />

Foundation for Chinese Medicine, Beth Israel Medical Center,<br />

New York, NY.<br />

Epidemiology, Risk, and Prevention: Prevention - Behavioral Interventions<br />

P1-11-01 Physical Activity Reduces the Risk of <strong>Breast</strong> <strong>Cancer</strong><br />

Hardefeldt PJ, Edirimanne S, Eslick GD. University of Sydney, NSW,<br />

Australia; Nepean Hospital, University of Sydney, Penrith, NSW,<br />

Australia.<br />

www.aacrjournals.org<br />

13s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Treatment: Chemotherapy - Advanced Disease<br />

P1-12-01 Evaluation on efficacy and safety of capecitabine plus<br />

docetaxel versus docetaxel monotherapy in metastatic breast<br />

cancer patients pretreated with anthracycline: Results from a<br />

randomized phase III study (JO21095)<br />

Sato N, Yamamoto D, Rai Y, Iwase H, Saito M, Iwata H, Masuda N,<br />

Oura S, Watanabe J, Kuroi K. Niigata <strong>Cancer</strong> Center Hospital, Niigata,<br />

Japan; Kansai Medical University Hirakata Hospital, Hirakata, Osaka,<br />

Japan; Sagara Hospital, Kagoshima, Japan; Kumamoto University<br />

Hospital, Kumamoto, Japan; Juntendo University Hospital, Bunkyo,<br />

Tokyo, Japan; Aichi <strong>Cancer</strong> Center Hospital, Nagoya, Aichi, Japan;<br />

National Hospital Organization Osaka National Hospital, Osaka,<br />

Japan; Wakayama Medical University, Wakayama, Wakayama, Japan;<br />

Shizuoka <strong>Cancer</strong> Center, Nagaizumi-cho, Suntou-gun, Shizuoka,<br />

Japan; Tokyo Metropolitan <strong>Cancer</strong> and Infectious Diseases Center<br />

Komagome Hospital, Bunkyo, Tokyo, Japan.<br />

P1-12-02 Results of a phase 2, multicenter, single-arm study of<br />

eribulin mesylate as first-line therapy for locally recurrent or<br />

metastatic HER2-negative breast cancer<br />

Vahdat L, Schwartzberg L, Glück S, Rege J, Liao J, Cox D,<br />

O’Shaughnessy J. Weill Cornell Medical College, New York, NY; The<br />

West Clinic, Memphis, TN; Sylvester Comprehensive <strong>Cancer</strong> Center,<br />

Miami, FL; Eisai Inc, Woodcliff Lake, NJ; Texas Oncology-Baylor<br />

Charles A. Sammons <strong>Cancer</strong> Center, Dallas, TX.<br />

P1-12-03 Low-dose capecitabine monotherapy in HER-2 negative<br />

metastatic breast cancer: a retrospective study<br />

Ambros T, Zeichner SB, Zaravinos J, Montero AJ, Ahn E, Mani A,<br />

Kronish L, Mahtani RL, Vogel CL. University of Miami, FL; Mount Sinai<br />

Medical Center, Miami Beach, FL; Memorial Hospital West, Pembroke<br />

Pines, FL; SUNY Downstate, Brooklyn, NY.<br />

P1-12-04 Carboplatin, nab-paclitaxel and bevacizumab as first-line<br />

treatment for metastatic breast cancer<br />

Lo SS, Guo R, Czaplicki KL, Robinson PA, Gaynor E, Barhamand FB,<br />

Schulz WC, Kash JJ, Horvath LE, Bayer RA, Petrowsky C, De la Torre<br />

R, Park JH, Albain KS. Loyola University Medical Center, Maywood,<br />

IL; Hematology Oncology Consultants Ltd., Naperville, IL; Swedish<br />

Americal Regional <strong>Cancer</strong> Center, Rockford, IL; Edward <strong>Cancer</strong><br />

Center, Naperville, IL; Central Dupage <strong>Cancer</strong> Center, Winfield, IL;<br />

CDPG Oncology at Delnor, Delnor, IL.<br />

P1-12-05 First-line chemotherapy with pegylated liposomal doxorubicin<br />

versus capecitabine in elderly patients with metastatic breast<br />

cancer: results of the phase III OMEGA study of the Dutch<br />

<strong>Breast</strong> <strong>Cancer</strong> Trialists’ Group (BOOG)<br />

Smorenburg CH, Seynaeve C, Wymenga MANM, Maartense E,<br />

de Graaf H, de Jongh FE, Braun HJ, Los M, Schrama JG, Portielje<br />

JEA, Hamaker M, van Tinteren H, de Groot SM, van Leeuwen-Stok<br />

EAE, Nortier HWR. Medical Center Alkmaar, Alkmaar, Netherlands;<br />

Erasmus Medical Center-Daniel den Hoed <strong>Cancer</strong> Center,<br />

Rotterdam, Netherlands; Medisch Spectrum Twente, Enschede,<br />

Netherlands; Reinier de Graaf Hospital, Delft, Netherlands; Medical<br />

Center Leeuwarden, Leeuwarden, Netherlands; Ikazia Hospital,<br />

Rotterdam, Netherlands; Vlietland Hospital, Schiedam, Netherlands;<br />

St. Antonius Hospital, Nieuwegein, Netherlands; Spaarne Hospital,<br />

Hoofddorp, Netherlands; Haga Hospital, The Hague, Netherlands;<br />

Diaconessehuis, Utrecht, Netherlands; Antoni van Leeuwenhoek<br />

Hospital/Netherlands <strong>Cancer</strong> Institute, Amsterdam, Netherlands;<br />

Comprehensive <strong>Cancer</strong> Centre the Netherlands, Amsterdam,<br />

Netherlands; Dutch <strong>Breast</strong> <strong>Cancer</strong> Trialists’ Group BOOG, Amsterdam,<br />

Netherlands; Leiden University Medical Center, Leiden, Netherlands.<br />

P1-12-06 N0937 (Alliance): Preliminary results of a phase II clinical trial<br />

of cisplatin and the novel agent brostallicin in patients with<br />

metastatic triple negative breast cancer (mTNBC)<br />

Moreno-Aspitia A, Rowland, Jr. KM, Allred JB, Liu H, Stella PJ, Gross<br />

HM, Soori GS, Karlin NJ, Perez EA. Mayo Clinic, Jacksonville, FL; Carle<br />

Foundation - Carle <strong>Cancer</strong> Center, Urbana, IL; Mayo Clinic, Rochester,<br />

MN; St. Joseph Mercy Health System, Ann Arbor, MI; Hematology<br />

& Oncology of Dayton, Inc., Dayton, OH; Missouri Valley <strong>Cancer</strong><br />

Consortium CCOP, Omaha, NE; Mayo Clinic, Scottsdale, AZ.<br />

P1-12-07 A Retrospective Analysis of nab-Paclitaxel as First-Line<br />

Therapy for Metastatic <strong>Breast</strong> <strong>Cancer</strong> Patients with Poor<br />

Prognostic Factors<br />

O’Shaughnessy J, Gradishar W, Bhar P, Iglesias J. Baylor Sammons<br />

<strong>Cancer</strong> Center, Texas Oncology and US Oncology, Dallas, TX;<br />

Maggie Daley Center for Women’s <strong>Cancer</strong> Care, Robert H. Lurie<br />

Comprehensive <strong>Cancer</strong> Center, Northwestern University Feinberg<br />

School of Medicine, Northwestern Memorial Hospital, Chicago,<br />

IL; Celgene Corporation, Durham, NC; Celgene Corporation,<br />

Mississauga, ON, Canada.<br />

P1-12-08 Withdrawn<br />

Treatment: Chemotherapy - Adjuvant<br />

P1-13-01 A randomized trial of CEF versus dose dense EC followed by<br />

paclitaxel versus AC followed by paclitaxel in women with<br />

node positive or high risk node negative breast cancer, NCIC<br />

CTG MA.21: Results of the final relapse free survival analysis<br />

Burnell MJ, Shepherd L, Gelmon K, Bramwell V, Walley B, Vandenberg<br />

E, Chalchal H, Pritchard K, Whelan T, Albain K, Perez E, Rugo H,<br />

O’Brien P, Chapman J, Levine M. NCIC Clinical Trials Group, Queen’s<br />

University, Kingston, ON, Canada; British Columbia <strong>Cancer</strong> Agency<br />

(BCCA), Vancouver, BC, Canada; University of Calgary, AB, Canada;<br />

Saint John Regional Hospital, Saint John, NB, Canada; London<br />

Regional <strong>Cancer</strong> Centre, London, ON, Canada; Sunnybrook <strong>Cancer</strong><br />

Centre, Toronto, ON, Canada; Juravinski <strong>Cancer</strong> Centre, Hamilton<br />

Health Sciences Centre, Hamilton, ON, Canada; Loyola University<br />

Medical Center, Maywood, IL; Mayo Clinic Jacksonville, FL; University<br />

of California, <strong>San</strong> Francisco, CA; McMaster University, Hamilton, ON,<br />

Canada; Allan Blair <strong>Cancer</strong> Centre, Regina, SK, Canada.<br />

P1-13-02 Withdrawn<br />

P1-13-03 Mature analysis of UK Taxotere as Adjuvant Chemotherapy<br />

(TACT) trial (CRUK 01/001); effects of treatment and<br />

characterisation of patterns of breast cancer relapse<br />

Bliss JM, Ellis P, Kilburn L, Bartlett J, Bloomfield D, Cameron D, Canney<br />

P, Coleman RE, Dowsett M, Earl H, Verril M, Wardley A, Yarnold J,<br />

Ahern R, Atkins N, Fletcher M, McLinden M, Barrett-Lee P. Institute<br />

of <strong>Cancer</strong> Research, Sutton, Surrey, United Kingdom; Guy’s Hospital,<br />

Kings Health Partners AHSC, London, United Kingdom; Velindre NHS<br />

Trust <strong>Cancer</strong> Centre, Cardiff, United Kingdom; Edinburgh <strong>Cancer</strong><br />

Research Centre, University of Edinburgh, United Kingdom; The<br />

Christie Hospital, Manchester, United Kingdom; Northern Centre<br />

for <strong>Cancer</strong> Care, Newcastle upon Tyne, United Kingdom; Brighton<br />

& Sussex University Hospitals, Brighton, United Kingdom; Ontario<br />

Institute for <strong>Cancer</strong> Research, Toronto, Canada; Beatson West of<br />

Scotland <strong>Cancer</strong> Centre, Glasgow, United Kingdom; Leeds Institute of<br />

Molecular Medicine, University of Leeds, Leeds, United Kingdom; ICR<br />

and Royal Marsden NHS Trust, London, United Kingdom; University<br />

of Cambridge, University of Cambridge and NIHR Cambridge<br />

Biomedical Research Centre, Cambridge, United Kingdom; Weston<br />

Park Hospital, Sheffield, United Kingdom; NHS National Services<br />

Scotland, Edinburgh, United Kingdom.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012 14s <strong>Cancer</strong> Research


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P1-13-04<br />

P1-13-05<br />

P1-13-06<br />

P1-13-07<br />

P1-13-08<br />

P1-13-09<br />

P1-13-10<br />

Optimal duration of adjuvant chemotherapy for high risk<br />

node negative breast cancer patients: 6-year results of the<br />

prospective randomized phase III trial PACS 05<br />

Kerbrat P, Coudert B, Asselain B, Levy C, Lortholary A, Marre A, Delva<br />

R, Rios M, Viens P, Brain E, Serin D, Edel M, Mauriac L, Campone<br />

M, Mouret-Reynier M-A, Bachelot T, Foucher-Goudier M-J, Roca L,<br />

Martin A-L, Roche H. Centre Eugene Marquis, Rennes, France; Centre<br />

Georges François Leclerc, Dijon, France; Institut Curie, Paris, France;<br />

Centre Francois Baclesse, Caen, France; Centre Catherine de Sienne,<br />

Nantes, France; Centre Hospitalier, Rodez, France; ICO Centre Paul<br />

Papin, Angers, France; Centre Alexis Vautrin, Vandoeuvre-les-Nancy,<br />

France; Institut Paoli Calmettes, Marseille, France; Institut Curie,<br />

Saint Cloud, France; Institut Sainte-Catherine, Avignon, France;<br />

Centre Hospitalier Emile Muller, Mulhouse, France; Institut Bergonié,<br />

Bordeaux, France; ICO Centre René Gauducheau, Saint Herblain,<br />

France; Centre Jean Perrin, Clermont-Ferrand, France; Centre Léon<br />

Berard, Lyon, France; Centre Hospitalier Bretagne-Sud, Lorient,<br />

France; Centre Val d’Aurelle, Montpellier, France; R&D Unicancer,<br />

Paris, France; Institut Claudius Regaud, Toulouse, France.<br />

The association between timing in adjuvant chemotherapy<br />

administration and overall survival for women with breast<br />

cancer within the National Comprehensive <strong>Cancer</strong> Network<br />

(NCCN)<br />

Vandergrift JL, Breslin TM, Niland JC, Edge SB, Wolff AC, Marcom<br />

PK, Rugo HS, Moy B, Wilson JL, Ottesen RA, Weeks JC, Wong Y-N.<br />

National Comprehensive <strong>Cancer</strong> Network, Fort Washington, PA;<br />

University of Michigan Comprehensive <strong>Cancer</strong> Center, Ann Arbor,<br />

MI; City of Hope Comprehensive <strong>Cancer</strong> Center, Duarte, CA; Dana-<br />

Farber/Brigham Women’s <strong>Cancer</strong> Center, Boston, MA; Roswell Park<br />

<strong>Cancer</strong> Institute, Buffalo, NY; The Sidney Kimmel Comprehensive<br />

<strong>Cancer</strong> Center at Johns Hopkins, Baltimore, MD; Duke <strong>Cancer</strong><br />

Institute, Durham, NC; UCSF Helen Diller Family Comprehensive<br />

<strong>Cancer</strong> Center, <strong>San</strong> Francisco, CA; The Ohio State University<br />

Comprehensive <strong>Cancer</strong> Center and James <strong>Cancer</strong> Hospital and<br />

Solove Research Institute, Columbus, OH; Massachusetts General<br />

Hospital <strong>Cancer</strong> Center, Boston, MA; Fox Chase <strong>Cancer</strong> Center,<br />

Philadelphia, PA.<br />

Withdrawn<br />

Association between Delayed Initiation of Adjuvant<br />

Chemotherapy and Survival in <strong>Breast</strong> <strong>Cancer</strong>: A Single-<br />

Institution Study and a Systematic Review and Meta-analysis<br />

Yu K-D, Fan L, Yang C, Shao Z-M. <strong>Cancer</strong> Center and <strong>Cancer</strong> Institute,<br />

Shanghai Medical College, Fudan University, Shanghai, China.<br />

Chemotherapy for <strong>Breast</strong> <strong>Cancer</strong> Causes Sustained Alteration<br />

in Number and Type of B Lymphocytes<br />

Verma R, Smalle N, McCurtin RE, Horgan K, Carter CRD, Hughes TA.<br />

Leeds General Infirmary, Leeds, West Yorkshire, United Kingdom; St<br />

James’s University Hospital, Leeds, West Yorkshire, United Kingdom;<br />

University of Leeds, West Yorkshire, United Kingdom.<br />

A multicenter randomized study comparing the dose dense<br />

G-CSF-supported sequential administration of FEC followed<br />

by docetaxel versus paclitaxel as adjuvant chemotherapy in<br />

women with axillary lymph node positive breast cancer<br />

Mavroudis D, Malamos N, Boukovinas I, Kakolyris S, Kourousis C,<br />

Athanasiadis A, Ziras N, Makrantonakis P, Polyzos A, Christophylakis<br />

C, Georgoulias V. Hellenic Oncology Research Group (HORG),<br />

Athens, Greece.<br />

Efficacy, toxicity and quality of life in older patients with earlystage<br />

breast cancer treated with oral Tegafur-uracil or classical<br />

CMF (cyclophosphamide, methotrexate, and fluorouracil): an<br />

exploratory analysis of National Surgical Adjuvant Study for<br />

<strong>Breast</strong> <strong>Cancer</strong> (N-SAS BC) 01 Trial<br />

Hara F, Watanabe T, Shimozuma K, Ohashi Y. NHO Shikoku <strong>Cancer</strong><br />

Center, Matsuyama, Ehime, Japan; Hamamatsu Oncology Center,<br />

Hamamatsu, Shizuoka, Japan; College of Life Sciences, Ritsumeikan<br />

University, Kusatsu, Shiga, Japan; School of Public Health, University<br />

of Tokyo, Bunkyo-ku, Tokyo, Japan.<br />

P1-13-11 Adjuvant treatment of early-stage breast cancer with<br />

eribulin mesylate following dose-dense doxorubicin and<br />

cyclophosphamide: preliminary results from a phase 2,<br />

single-arm feasibility study<br />

Traina TA, Hudis C, Fornier M, Lake D, Lehman R, Berkowitz AP,<br />

Rege J, Liao J, Cox D, Seidman AD. Memorial Sloan-Kettering <strong>Cancer</strong><br />

Center, New York, NY; Eisai Inc, Woodcliff Lake, NJ.<br />

P1-13-12 Withdrawn<br />

P1-13-13 First planned efficacy analysis of the NNBC 3-Europe trial:<br />

Addition of docetaxel to anthracycline containing adjuvant<br />

chemotherapy in high risk node-negative breast cancer<br />

patients<br />

Thomssen C, Kantelhardt EJ, Meisner C, Vetter M, Schmidt M, Martin<br />

P-M, Veyret C, Augustin D, Hanf V, Paepke D, Meinerz W, Hoffmann<br />

G, Wiest W, Sweep FCGJ, Schmitt M, Jaenicke F, von Minckwitz G,<br />

Harbeck N, On Behalf of the NNBC 3-Europe Study Group. Martin-<br />

Luther University Halle-Wittenberg, Halle an der Saale, Germany;<br />

Eberhard-Karls-University Tuebingen, Tuebingen, Germany;<br />

Johannes Gutenberg-Universität Mainz, Mainz, Germany; Medical<br />

Faculty Marseille, Marseille, France; Henri Becquerel Center, Rouen,<br />

France; Klinikum Deggendorf, Deggendorf, Germany; Klinikum Fürth,<br />

Fürth, Germany; Technical University Munich, München, Germany;<br />

St. Vincenz-Hospital, Paderborn, Germany; St. Josefs-Hospital,<br />

Wiesbaden, Germany; Katholisches Klinikum, Mainz, Germany;<br />

Radbourd University Nijmegen, Nijmegen, Netherlands; University<br />

Hamburg, Hamburg, Germany; German <strong>Breast</strong> Group, Neu-Isenburg,<br />

Germany; Ludwig-Maximilian-University, Munich, Germany.<br />

Treatment: Neoadjuvant Chemotherapy<br />

P1-14-01 Adding capecitabine and trastuzumab to neoadjuvant breast<br />

cancer chemotherapy - first survival analysis of the GBG/AGO<br />

intergroup-study GeparQuattro<br />

von Minckwitz G, Rezai M, Loibl S, Fasching PA, Huober J, Tesch H,<br />

Bauerfeind I, Hilfrich J, Eidtmann H, Gerber B, Hanusch C, Blohmer<br />

J-U, Costa S-D, Jackisch C, Paepke S, Schneeweiss A, Kuemmel S,<br />

Denkert C, Mehta K, Untch M. German <strong>Breast</strong> Group, Neu-Isenburg;<br />

Louisenkrankenhaus Düsseldorf; University Erlangen; University<br />

Duesseldorf; Bethanien-Kankenhaus Frankfurt; Klinikum Landshut;<br />

Eilenriedeklinik Duesseldorf; University Kiel; University Rostock;<br />

Roteskreuzklinikum Muenchen; <strong>San</strong>kt Gertrauden Berlin; University<br />

Magdeburg; Klinikum Offenbach; Frauenklinik München; University<br />

Heidelberg; Kliniken Essen Mitte; Charite Berlin; Helios Kliniken Berlin.<br />

P1-14-02 Preoperative docetaxel (T) with or without capecitabine (X)<br />

following epirubicin, 5-fluorouracil and cyclophosphamide<br />

(FEC) in patients with operable breast cancer (OOTR N003):<br />

Results of comparative study and predictive marker analysis<br />

Toi M, Ohno S, Sato N, Masuda N, Sasano H, Takahashi F, Bando<br />

H, Iwata H, Morimoto T, Kamigaki S, Nakayama T, Murakami S,<br />

Nakamura S, Kuroi K, Aogi K, Kashiwaba M, Yamashita H, Hisamatsu<br />

K, Ito Y, Yamamoto Y, Ueno T, Fakhrejahani E, Yoshida N, Chow LWC.<br />

Organisation for Oncology and Translational Research (OOTR),<br />

Kyoto, Japan.<br />

P1-14-03 Overall survival results of a multicenter randomized phase II<br />

study in locally advanced breast cancer patients treated with<br />

or without neoadjuvant Trastuzumab for HER2 positive tumor<br />

(Remagus 02 trial)<br />

Giacchetti S, Pierga J-Y, Asselain B, Delaloge S, Brain E, Espié M,<br />

Mathieu M-C, Bertheau p, de Cremoux P, Tembo O, Marty M. Hôpital<br />

Saint Louis, Assitance Publique-Hôpitaux, Paris, France; Institut Curie,<br />

Paris, France; Institut Gustave Roussy, Villejuif, France; Institut Curie-<br />

Saint Cloud, Saint Cloud, France.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012 15s <strong>Cancer</strong> Research


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P1-14-04<br />

P1-14-05<br />

P1-14-06<br />

P1-14-07<br />

P1-14-08<br />

Long- term outcome after neoadjuvant radiochemotherapy<br />

in locally advanced non-inflammatory breast cancer and<br />

predictive factors for a pathologic complete remission; results<br />

of a multi-variate analysis<br />

Matuschek C, Boelke E, Roth S, Bojar H, Audretsch W, Janni JW,<br />

Nestle-Kraemling C, Sauer R, Speer V, Budach W. Heinrich Heine<br />

University, Düsseldorf, Germany; European Institute for Molecular<br />

Oncology, Düsseldorf, Germany; Marien Hospital, Düsseldorf,<br />

Germany; Krankenhaus Gerresheim, Düsseldorf; University of<br />

Erlangen, Germany.<br />

Phase I/II Trial of primary chemotherapy with non-pegylated<br />

liposomal doxorubicin, paclitaxel and lapatinib in patients<br />

with HER2-positive, early stage breast cancer<br />

Aktas B, Kümmel S, Krocker J, Elling D, Lantzsch T, Bischoff J, Fersis<br />

N, Böhme M, Belau AK, Lampe D, Schmid P. University Hospital<br />

of Essen, Germany; Kliniken Essen Mitte, Essen, Germany; <strong>San</strong>a<br />

Klinikum Lichtenberg, Berlin, Germany; St. Barbara Krabkenhaus,<br />

Halle, Germany; University Hospital of Magdeburg, Germany;<br />

Klinikum Chemnitz, Germany; Klinik St. Marienstift, Magdeburg,<br />

Germany; University Hospital of Greifswald, Germany; Asklepios Klinik<br />

Weißenfels, Weißenfels, Germany; University of Sussex,<br />

United Kingdom.<br />

Significance of examining biomarkers of residual tumors<br />

after neoadjuvant chemotherapy using trastuzumab in<br />

combination with anthracycline and taxane in patients with<br />

primary HER2-positive breast cancer<br />

Kurozumi S, Takei H, Inoue K, Matsumoto H, Hayashi Y, Ninomiya J,<br />

Kubo K, Tsuboi M, Nagai S, Ookubo F, Oba H, Kurosumi M, Horiguchi<br />

J, Takeyoshi I. Saitama <strong>Cancer</strong> Center, Saitama, Japan; Gunma<br />

University Graduate School of Medicine, Gunma, Japan.<br />

Neoadjuvant chemotherapy and pathologic complete<br />

response in relation to the clinical response, results from a<br />

phase III study (INTENS) of the Dutch <strong>Breast</strong> <strong>Cancer</strong> Trialists’<br />

Group (BOOG)<br />

Tjan-Heijnen VCG, Vriens BE, de Vries B, van Gastel SM, Wals J, Smilde<br />

TJ, van Warmerdam LJ, van Laarhoven HW, van Spronsen DJ, Borm<br />

GF. Maastricht University Medical Centre, Maastricht, Netherlands;<br />

Comprehensive <strong>Cancer</strong> Centre, Nijmegen, Netherlands; Atrium<br />

Medical Centre, Heerlen, Netherlands; Jeroen Bosch Hospital,<br />

‘s Hertogenbosch, Netherlands; Catharina Hospital, Eindhoven,<br />

Netherlands; Radboud University Nijmegen Medical Centre,<br />

Nijmegen, Netherlands; Canisius-Wilhelmina Hospital, Nijmegen,<br />

Netherlands.<br />

A prospective multicenter randomized phase II neo-adjuvant<br />

study of 5-fluorouracil, epirubicin and cyclophosphamide<br />

(FEC) followed by docetaxel, cyclophosphamide and<br />

trastuzumab (TCH) versus TCH followed by FEC versus TCH<br />

alone, in patients (pts) with operable HER2 positive breast<br />

cancer: JBCRG-10 study<br />

Masuda N, Sato N, Higaki K, Kashiwaba M, Matsunami N, Takano T,<br />

Yamamura J, Kaneko K, Takahashi M, Ohno S, Fujisawa T, Tsuyuki<br />

S, Miyoshi Y, Ohtani S, Yamamoto Y, Bando H, Onoda T, Kawabata<br />

H, Morita S, Ueno T, Toi M. NHO Osaka National Hospital, Osaka,<br />

Japan; Niigata <strong>Cancer</strong> Center Hospital, Niigata, Japan; Hiroshima<br />

City Hospital, Hiroshima, Japan; Iwate Medical University, Morioka,<br />

Japan; Osaka Rosai Hospital, Sakai, Japan; Toranomon Hospital,<br />

Tokyo, Japan; Hokkaido <strong>Cancer</strong> Center, Sapporo, Japan; National<br />

Kyushu <strong>Cancer</strong> Center, Fukuoka, Japan; Gunma Prefectural <strong>Cancer</strong><br />

Center, Ohta, Japan; Osaka Red Cross Hospital, Osaka, Japan; Hyogo<br />

College of Medicine, Nishinomiya, Japan; Kumamoto University<br />

Hospital, Kumamoto, Japan; University of Tsukuba, Faculty of<br />

Medicine, Tsukuba, Japan; Yokohama Asahi Central General Hospital,<br />

Yokohama, Japan; Yokohama City University Graduate School of<br />

Medicine and Medical Center, Yokohama, Japan; Kyoto University,<br />

Kyoto, Japan.<br />

P1-14-09<br />

P1-14-10<br />

P1-14-11<br />

P1-14-12<br />

P1-14-13<br />

P1-14-14<br />

P1-14-15<br />

P1-14-16<br />

P1-14-17<br />

Immunohistochemical classification of intrinsic subtypes as<br />

a predictive biomarker of pathological complete response<br />

in breast cancer patients treated with preoperative<br />

chemotherapy<br />

Nagayama A, Jinno H, Takahashi M, Hayashida T, Hirose S, Kitagawa<br />

Y. School of Medicine, Keio University, Shinjuku, Tokyo, Japan.<br />

Final results of neoadjuvant trial of bevacizumab (B) and<br />

trastuzumab (T) in combination with weekly paclitaxel (P)<br />

as neoadjuvant treatment in HER2-positive breast cancer: A<br />

phase II trial (AVANTHER)<br />

Fernandez M, Calvo I, Martinez N, Herrero M, Quijano Y, Duran H,<br />

Garcia-Aranda M, Suarez A, Lopez-Rios F, Perez D, Perea S, Hidalgo<br />

M, Garcia-Estevez L. Centro Integral Oncológico Clara Campal,<br />

Madrid, Spain; Hospital Ramón y Cajal, Madrid, Spain; Hospital Costa<br />

del Sol, Marbella, Spain.<br />

Assessing Prognosis and Therapy Response in Primary<br />

Systemic Therapy of <strong>Breast</strong> <strong>Cancer</strong> with Magnetic Resonance<br />

Spectroscopy<br />

Bolan PJ, Wey A, Eberly LE, Nelson MT, Haddad TC, Yee D, Garwood<br />

M. University of Minnesota, Minneapolis, MN; Masonic <strong>Cancer</strong><br />

Center, University of Minnesota, Minneapolis, MN.<br />

Response to neoadjuvant chemotherapy and prognosis of<br />

primary breast cancer according to intrinsic subtype<br />

Kochi M, Ito M, Ohtani S, Higaki K. Hiroshima City Hospital,<br />

Hiroshima, Japan.<br />

Increased Pathologic Complete Response Rate and Reduced<br />

Tumour RNA Levels Upon Treatment of Locally Advanced<br />

<strong>Breast</strong> <strong>Cancer</strong> with Neoadjuvant Concurrent Chemotherapy<br />

and Radiation<br />

Brackstone M, Chambers A, Guo B, Vandenberg T, Potvin K, Perea F,<br />

Parissenti A. London Health Sciences Centre, London, ON, Canada;<br />

Health Sciences North, Sudbury, ON, Canada; RNA Diagnostics, Inc.,<br />

Sudbury, ON, Canada.<br />

Neoadjuvant Sunitinib (S) Administered with Weekly<br />

Paclitaxel (P)/Carboplatin(C) in Patients (Pts) with Locally<br />

Advanced Triple-Negative <strong>Breast</strong> <strong>Cancer</strong> (TNBC):<br />

Preliminary Results from a Phase I/II Trial of the Sarah<br />

Cannon Research Institute<br />

Yardley DA, Barton J, Hendricks C, Webb C, Priego V, Nimeh N,<br />

Gravenor D, Shastry M, Chirwa T, Burris HA. Sarah Cannon Research<br />

Institute, Nashville, TN; Tennessee Oncology, PLLC, Nashville, TN;<br />

National Capital Clinical Research Consortium, Bethesda, MD; Baptist<br />

Hospital East, Louisville, KY; Center for <strong>Cancer</strong> and Blood Disorders,<br />

Bethesda, MD; <strong>Cancer</strong> Center of Southwest Oklahoma Research,<br />

Lawton, OK; Family <strong>Cancer</strong> Center Foundation, Inc., Memphis, TN.<br />

Recent experience of neoadjuvant chemotherapy according<br />

to breast cancer subtype: experience from a large United<br />

Kindgom teaching hospital<br />

Rattay T, Kaushik M, Ahmed S, Shokuhi S. University Hospitals of<br />

Leicester, United Kingdom.<br />

Young age: predicts poor response rate after neoadjuvant<br />

chemotherapy in endocrine-responsive breast cancer<br />

Kim MK, Noh D-Y, Han W, Moon H-G, Ahn S-K, Kim JS, Kim T, Kim JY,<br />

Lee JW. Seoul National University Hospital, Seoul, Korea.<br />

Study of breast cancer shrinkage modes after neoadjuvant<br />

chemotherapy with whole-mount serial sections and threedimensional<br />

pathological and MRI reconstruction<br />

Wang Y-S, Zhang Z-P, Liu G, Mu D-B, Sun X-Y. Shandong <strong>Cancer</strong><br />

Hospital & Institute, Jinan, Shandong, China.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012 16s <strong>Cancer</strong> Research


December 4–8, 2012<br />

Program Schedule<br />

P1-14-18 Overall survival results of a multicenter randomized<br />

phase II study in locally advanced breast cancer patients<br />

treated with or without celecoxib for HER2 negative tumor<br />

(Remagus 02 trial)<br />

Giacchetti S, Pierga J-Y, Delaloge S, Asselain B, Brain E, Guinebretière<br />

JM, Che-Lehman J, Mathieu M-C, Sigal B, Marty M. Hôpital Saint<br />

Louis, Assitance Publique-Hôpitaux, Paris, France; Institut Curie, Paris,<br />

France; Institut Gustave Roussy, Villejuif, France; Institut Curie-Saint<br />

Cloud, Saint Cloud, France.<br />

P1-14-19 <strong>Breast</strong>-Conserving Surgery after Neoadjuvant Chemotherapy<br />

Is Oncologically Safe for Stage III <strong>Breast</strong> <strong>Cancer</strong> Patients<br />

Shin H-C, Han W, Moon H-G, Im S-A, Park S-J, Noh D-Y. Chung-<br />

Ang University Hospital, Seoul, Republic of Korea; Seoul National<br />

University Hospital, Seoul, Republic of Korea.<br />

P1-14-20 Withdrawn<br />

P1-14-21 High antitumoral activity of neoadjuvant chemotherapy<br />

(NCT) with weekly paclitaxel + capecitabine and trastuzumab<br />

in patients with locally advanced HER2+ breast cancer (HER2+<br />

LABC): Preliminary results<br />

Augereau P, Raro P, Verriele V, Delva R, Abadie-Lacourtoisie S,<br />

Ceban T, Paillocher N, Valo I, Maillart P, Soulie P. ICO Paul Papin,<br />

Angers, France.<br />

Treatment: Toxicities - Management<br />

P1-15-01 Patterns of granulocyte colony stimulating factor (G-CSF)<br />

use in elderly breast cancer (BC) patients receiving<br />

myelosuppressive chemotherapy<br />

Blaes AH, Chia V, Solid C, Page J, Barron RL, Choi MR, Arneson TJ.<br />

University of Minnesota, Minneapolis, MN; Amgen, Inc., Thousand<br />

Oaks, CA; Minneapolis Medical Research Foundation,<br />

Minneapolis, MN.<br />

P1-15-02 Febrile neutropenia (FN) risk assessment and granulocyte<br />

colony-stimulating factor (G-CSF) guideline adherence<br />

in patients with breast cancer – results from a German<br />

prospective multicentre observational study (PROTECT)<br />

Steffens C-C, Eschenburg H, Kurbacher C, Goehler T, Schmidt M,<br />

Eustermann H, Schaffrik M, Otremba B. MVZ für Hämatologie/<br />

Onkologie, Klinik Dr. Hancken; Praxis für Hämatologie und<br />

internistische Onkologie, Güstrow; Praxis für Gynäkologie und<br />

gynäkologische Onkologie, Medizinisches Zentrum Bonn; Praxis für<br />

Hämatologie und internistische Onkologie, Dresden; Gynäkologische<br />

Onkologie, Universitätsfrauenklinik Mainz; WiSP Wissenschaftlicher<br />

Service Pharma GmbH, Langenfeld; Amgen GmbH, München;<br />

Onkologische Praxis Oldenburg/Delmenhorst.<br />

P1-15-03 Withdrawn<br />

P1-15-04 The relationship of relative dose intensity and supportive care<br />

to febrile neutropenia rates in patients with early stage breast<br />

cancer receiving chemotherapy: a prospective cohort study of<br />

chemotherapy-associated toxicity<br />

Culakova E, Poniewierski MS, Wogu AF, Kuderer NM, Crawford J,<br />

Dale DC, Lyman GH. Duke University, Durham, NC; University of<br />

Washington, Seattle, WA.<br />

P1-15-05 GSTP1 polymorphism is associated with chemotherapy<br />

induced neuropathy<br />

Miltenburg NC, Opdam M, Winter M, van Geer M, Oosterkamp HM,<br />

Boogerd W, Linn SC. The Netherlands <strong>Cancer</strong> Institute, Amsterdam,<br />

Netherlands.<br />

P1-15-06 The impact of musculoskeletal toxicity on adherence to<br />

endocrine therapy in women with early stage breast cancer–<br />

observations in a non-trial setting<br />

Dent SF, Campbell MM, Crawley FL, Clemons MJ. The Ottawa<br />

Hospital Regional <strong>Cancer</strong> Center, Ottawa, ON, Canada.<br />

P1-15-07 Ixabepilone-associated peripheral neuropathy in<br />

metastatic breast cancer patients and its effects on the<br />

ultrastructure of neurons<br />

Jain S, Carlson K, Chuang E, Cigler T, Moore A, Donovan D, Lam C,<br />

Cobham MV, Schneider S, Ramnarain A, Carey B, Ward M, Lane M,<br />

Strickland S, Vahdat L. Weill Cornell Medical College; Rockefeller<br />

University.<br />

P1-15-08 Higher toxicity of docetaxel for obese women with early<br />

breast cancer: lean body mass is a significant predictor of<br />

chemotherapy dose intensity reduction<br />

Gouerant S, Clatot F, Modzlewski R, Chaker M, Rigal O, Veyret<br />

C, Leheurteur M. Henri Becquerel Center, Rouen, France; Rouen<br />

University Hospital, Rouen, France.<br />

P1-15-09 Multi-Institutional Evaluation of Bioimpedance<br />

Spectroscopy (BIS) in the Early Detection of <strong>Breast</strong> <strong>Cancer</strong><br />

Related Lymphedema<br />

Vicini F, Arthur D, Shah C, Anglin BV, Curcio L, Laidley AL, Beitsch<br />

P, Whitworth P, Lyden M. Michigan Healthcare Professionals/21st<br />

Century Oncology; Virginia Commonwealth University; Beaumont<br />

Health System; The Medical Center of Plano; Advanced <strong>Breast</strong> Care<br />

Specialists of Orange County; Texas <strong>Breast</strong> Specialists; Dallas Surgical<br />

Group; Nashville <strong>Breast</strong> Center; Biostat Inc.<br />

P1-15-10 Chemotherapy-Induced Neutropenia in <strong>Breast</strong> <strong>Cancer</strong><br />

Patients Receiving Sequential Anthracycline and Taxane<br />

Chemotherapy: An Institutional Experience<br />

Staudigl C, Seifert M, Tea M-K, Pfeiler G, Fink-Retter A, Fritzer N,<br />

Singer CF. Medical University of Vienna, Austria; Alps-Adria University<br />

Klagenfurt, Klagenfurt, Austria.<br />

P1-15-11 The Kampo medicine Goshajinkigan prevents docetaxelrelated<br />

peripheral neuropathy in breast cancer patients<br />

Abe H, Mori T, Kawai Y, Itoi N, Tomida K, Cho H, Kubota Y, Umeda<br />

T, Tani T. Shiga University of Medical Science Hospital, Otsu, Shiga,<br />

Japan; Shiga University of Medical Science, Otsu, Shiga, Japan.<br />

P1-15-12 Single Nucleotide Polymorphism (SNP) Bayesian networks<br />

(BNs) Predict Risk of Chemotherapy-Induced Side Effects<br />

in Patients with <strong>Breast</strong> <strong>Cancer</strong> Receiving Dose Dense (DD)<br />

Doxorubicin/Cyclophosphamide Plus Paclitaxel (AC+T)<br />

Schwartzberg LS, Sonis ST, Walker MS, Weidner SM, Alterovitz G. The<br />

West Clinic, Memphis, TN; Brigham and Women’s Hospital, Boston,<br />

MA; ACORN CRO, Memphis, TN; Inform Genomics, Boston, MA;<br />

Harvard Medical School, Boston, MA.<br />

Ongoing Trials 1: Her2<br />

OT1-1-01 A phase II study of neoadjuvant epirubicin/cyclophosphamide<br />

(EC) followed by weekly nanoparticle albumin-bound<br />

paclitaxel with or without trastuzumab for node-positive<br />

breast cancer<br />

Hirano A, Hattori A, Kamimura M, Ogura K, Kim N, Setoguchi Y,<br />

Okubo F, Inoue H, Jibiki N, Miyamoto R, Kinoshita J, Kimura K,<br />

Fujibayashi M, Shimizu T. Tokyo Women’s Medical University,<br />

Medical Center East, Tokyo, Japan; Tokyo Women’s Medical<br />

University, Yachiyo Medical Center, Yachiyo, Japan.<br />

OT1-1-02 A single-arm phase IIIb study of pertuzumab and trastuzumab<br />

with a taxane as first-line therapy for patients with HER2-<br />

positive advanced breast cancer (PERUSE)<br />

Bachelot T, Ciruelos E, Peretz-Yablonski T, Schneeweiss A, Puglisi<br />

F, Mitchell L, Dünne A, Miles D. Centre Leon Bérard, Lyon, France;<br />

Hospital Universitario 12 Octubre, Madrid, Spain; Hadassah- Hebrew<br />

University Medical Center, Jerusalem, Israel; National Center for<br />

Tumor Diseases, University-Hospital Heidelberg, Germany; University<br />

Hospital of Udine, Italy; F. Hoffmann-La Roche, Basel, Switzerland;<br />

Mount Vernon <strong>Cancer</strong> Centre, Mount Vernon Hospital, Middlesex,<br />

United Kingdom.<br />

www.aacrjournals.org<br />

17s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

OT1-1-03<br />

OT1-1-04<br />

PERSEPHONE: Duration of Trastuzumab with Chemotherapy<br />

in women with HER2 positive early breast cancer<br />

Earl HM, Cameron DA, Miles D, Wardley AM, Ogburn ERM, Vallier<br />

A-L, Loi S, Higgins HB, Hiller L, Dunn JA. University of Cambridge,<br />

Cambridge, United Kingdom; NIHR Cambridge Biomedical Research<br />

Centre, Cambridge, United Kingdom; Edinburgh University,<br />

Edinburgh, United Kingdom; Mount Vernon <strong>Cancer</strong> Centre,<br />

Middlesex, United Kingdom; The Christie Hospital, Manchester,<br />

United Kingdom; Warwick Clinical Trials Unit, University of Warwick,<br />

Coventry, United Kingdom; Cambridge <strong>Cancer</strong> Trials Centre,<br />

Cambridge.<br />

ALTERNATIVE: safety and efficacy of lapatinib (L), trastuzumab<br />

(T), or both in combination with an aromatase inhibitor<br />

(AI) for the treatment of hormone receptor-positive (HR+),<br />

human epidermal growth factor receptor 2 positive (HER2+)<br />

metastatic breast cancer<br />

Johnston S, Wroblewski S, Huang Y, Harvey C, Nagi F, Franklin N,<br />

Gradishar W. Royal Marsden NHS Foundation Trust and Institute<br />

of <strong>Cancer</strong> Research, London, United Kingdom; GlaxoSmithKline,<br />

Collegeville, PA; GlaxoSmithKline, Stockley Park, United Kingdom;<br />

Northwestern University, Chicago, IL.<br />

A Phase I pharmacokinetics trial comparing PF-05280014 and<br />

trastuzumab in healthy volunteers (REFLECTIONS B327-01)<br />

Ricart AD, Zacharchuk C, Reich SD, Meng X, Barker KB, Taylor CT,<br />

Hansson AG. Pfizer Inc., <strong>San</strong> Diego, CA; Pfizer Inc., Cambridge, MA;<br />

Pfizer Inc., New Haven, CT.<br />

A phase III randomized study of Paclitaxel and Trastuzumab<br />

versus Paclitaxel, Trastuzumab and Lapatinib in first line<br />

treatment of HER2 positive metastatic breast cancer<br />

Crown JP, Moulton B, O’Donovan N. St Vincent’s University Hospital,<br />

Elm Park, Dublin, Ireland; ICORG (All Ireland Cooperative Oncology<br />

Research Group), Dublin 4, Ireland; National Institute for Cellular<br />

Biotechnology, Dublin City University, Dublin 9, Ireland.<br />

Human epidermal growth factor receptor 2 (HER2)<br />

suppression with the addition of lapatinib to trastuzumab<br />

in HER2-positive metastatic breast cancer (LTP112515)<br />

Lin N, Danso MA, David AK, Muscato J, Rayson D, Houck, III WA,<br />

Ellis C, DeSilvio M, Garofalo A, Nagarwala Y, Winer E. Dana-Farber<br />

<strong>Cancer</strong> Institute, Boston, MA; Virginia Oncology Associates, Norfolk,<br />

VA; Augusta Oncology Associates, Augusta, GA; Missouri <strong>Cancer</strong><br />

Associates, Columbia, MO; QEII Health Sciences Centre, Halifax,<br />

NS, Canada; Virginia <strong>Cancer</strong> Specialists, PC, Winchester, VA;<br />

GlaxoSmithKline Oncology, Collegeville, PA.<br />

Clinical outcomes among ErbB2+ MBC patients treated with<br />

lapatinib-capecitabine after trastuzumab progression:<br />

Role of early switch to lapatinib (TYCO study)<br />

Abulkhair O, Uslu R, Sezgin C, Büyükberber S, Darwish T, Isikdogan<br />

A, Gumus M, Dane F, Sevinc A, Halawani H, Uncu D, Marrero N,<br />

Tobler J, Soares C, Landis S, Moraes E, Gidekel R, <strong>San</strong>tillana S,<br />

Nunez P, Cagnolati S, Rodriguez JG. Ege University Medical Faculty,<br />

Izmir, Turkey; Gazi University Medical Faculty, Ankara, Turkey; King<br />

Abdulaziz Medical City National Guard Health Affairs, Riyadh, Saudi<br />

Arabia; King Abdullah Medical City - Oncology Centre, Jeddah, Saudi<br />

Arabia; Dicle University Medical Faculty, Diyarbakir, Turkey; Kartal<br />

Training and Research Hospital, Istanbul, Turkey; Marmara University<br />

Training and Research Hospital, Istanbul, Turkey; Gaziantep<br />

University Medical Faculty, Gaziantep, Turkey; King Fahad Specialist<br />

Hospital, Dammam, Saudi Arabia; Numune Training and Research<br />

Hospital, Ankara, Turkey; Instituto Docente de Urología, Valencia,<br />

Venezuela; GlaxoSmithKline, Rio de Janeiro, Brazil; GlaxoSmithKline,<br />

Buenos Aires, Argentina; GlaxoSmithKline, Stockley Park, United<br />

Kingdom; Hospital Oncológico Padre Machado, Caracas, Venezuela;<br />

Centro Oncologico-FIDES-La Plata, Buenos Aires, Argentina.<br />

OT1-1-09<br />

OT1-1-10<br />

Opti-HER HEART: A prospective, multicenter, single-arm,<br />

phase II study to evaluate the safety of neoadjuvant liposomal<br />

doxorubicin plus paclitaxel, trastuzumab, and pertuzumab in<br />

patients with operable HER2-positive breast cancer<br />

Gavilá J, Llombart A, Guerrero A, Ruiz A, Guillem V. Fundación<br />

Instituto Valenciano de Oncología, Valencia, Spain; Hospital Arnau<br />

de Vilanova, Valencia, Spain; SOLTI <strong>Breast</strong> <strong>Cancer</strong> Research Group,<br />

Barcelona, Spain.<br />

DETECT III - A multicenter, randomized, phase III study to<br />

compare standard therapy alone versus standard therapy plus<br />

lapatinib in patients with initially HER2-negative metastatic<br />

breast cancer but with HER2-positive circulating tumor cells<br />

Melcher CA, Janni JW, Schneeweiss A, Fasching PA, Hagenbeck CD,<br />

Aktas B, Pantel K, Solomayer EF, Ortmann U, Jaeger BAS, Mueller<br />

V, Rack BK, Fehm TN. University Hospital Duesseldorf, Germany;<br />

National Center for Tumor Diseases, Heidelberg, Germany; University<br />

Hospital Erlangen; University Hospital Essen, Germany; University<br />

Medical Center Hamburg-Eppendorf, Hamburg, Germany; University<br />

Hospital Homburg, Germany; Ludwig-Maximilians-University<br />

Munich, Munich, Germany; University Hospital Hamburg-Eppendorf,<br />

Germany; University Hospital Tuebingen, Germany.<br />

TBCRC 022: Phase II Trial of Neratinib for Patients with Human<br />

Epidermal Growth Factor Receptor 2 (HER2)-Positive <strong>Breast</strong><br />

<strong>Cancer</strong> and Brain Metastases<br />

Freedman RA, Gelman RS, Wefel JS, Krop IE, Melisko ME, Ly A, Agar<br />

NYR, Connolly RM, Blackwell KL, Nabell LM, Ingle JN, Van Poznak<br />

CH, Puhalla SL, Niravath PA, Ryabin N, Wolff AC, Winer EP, Lin N.<br />

Dana-Farber <strong>Cancer</strong> Institute, Boston, MA; The University of Texas<br />

MD Anderson <strong>Cancer</strong> Center, Houston, TX; University of California,<br />

<strong>San</strong> Francisco, CA; Brigham and Women’s Hospital, Boston, MA;<br />

Johns Hopkins University, Baltimore, MD; Duke University, Durham,<br />

NC; University of Alabama, Birmingham, AL; Mayo Clinic, Rochester,<br />

MN; University of Michigan, Ann Arbor, MI; Univerity of Pittsburgh,<br />

Pittsburgh, PA; Baylor, Houston, TX,<br />

NSABP FB-7 Trial: A Phase II Randomized Clinical Trial<br />

Evaluating Neoadjuvant Therapy Regimens with Weekly<br />

Paclitaxel and Neratinib or Trastuzumab or Neratinib and<br />

Trastuzumab Followed by Doxorubicin and Cyclophosphamide<br />

with Postoperative Trastuzumab in Women with Locally<br />

Advanced HER2-Positive <strong>Breast</strong> <strong>Cancer</strong><br />

Lu J, Jacobs SA, Buyse ME, Paik S, Wolmark N. State University of New<br />

York at Stony Brook, Stony Brook, NY; National Surgical Adjuvant<br />

<strong>Breast</strong> and Bowel Project, Pittsburgh, PA.<br />

Dual blockade with Afatinib and Trastuzumab as neoadjuvant<br />

treatment for patients with locally advanced or operable<br />

breast cancer receiving taxane-anthracycline containing<br />

chemotherapy (DAFNE)-GBG70<br />

Hanusch C, Schneeweiss A, Untch M, Paepke S, Kuemmel S,<br />

Jackisch C, Huober J, Hilfrich J, Gerber B, Eidtmann H, Denkert C,<br />

Costa S-D, Blohmer J-U, Loibl S, Nekljudova V, von Minckwitz G.<br />

Rotkreuzklinikum Muenchen; University Heidelberg; Helios Klinik<br />

Berlin; Frauenklinik Muenchen; Kliniken Essen Mitte; Klinikum<br />

offenbach; University Duesseldorf; Eilenriedeklinik Düsseldorf;<br />

University Rostock; University Kiel; Charite Berlin; University<br />

Magdeburg; <strong>San</strong>kt Gertrauden Berlin; German <strong>Breast</strong> Group, Neu-<br />

Isenburg.<br />

Open-label, Phase II trial of afatinib, with or without<br />

vinorelbine, for the treatment of HER2-overexpressing<br />

inflammatory breast cancer (IBC)*<br />

Swanton C, Cromer J, On behalf of the 1200.89 trial group. CR-UK<br />

London Research Institute, London, United Kingdom; Boehringer<br />

Ingelheim, North Ryde, Australia.<br />

OT1-1-05<br />

OT1-1-06<br />

OT1-1-07<br />

OT1-1-08<br />

OT1-1-11<br />

OT1-1-12<br />

OT1-1-13<br />

OT1-1-14<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012 18s <strong>Cancer</strong> Research


December 4–8, 2012<br />

Program Schedule<br />

OT1-1-15 LUX-<strong>Breast</strong> 3: Randomized Phase II trial of afatinib (BIBW<br />

2992) alone or with vinorelbine versus investigator’s choice of<br />

treatment in patients (pts) with HER2-positive breast cancer<br />

(BC) with progressive brain metastases after trastuzumab and/<br />

or lapatinib-based therapy*<br />

Joensuu H, Ould-Kaci M, On behalf of the 1200.67 trial group.<br />

Helsinki University Central Hospital, Helsinki, Finland; Boehringer<br />

Ingelheim, Paris, France.<br />

OT1-1-16 LUX-<strong>Breast</strong> 1: Randomized, Phase III trial of afatinib (BIBW<br />

2992) and vinorelbine vs. trastuzumab and vinorelbine in<br />

patients with HER2-overexpressing metastatic breast cancer<br />

(MBC) failing one prior trastuzumab treatment*<br />

Xu B, Im S-A, Huang C-S, Im Y-H, Ro J, Zhang Q, Arora R, Mehta<br />

A, Jung K, Yeh D-C, Lee S, Jassem J, Wojtukiewicz M, Chen S-C,<br />

Lahogue A, Uttenreuther-Fischer M, Hurvitz S-A, Harbeck N, Piccart-<br />

Gebhart M, On behalf of the LUX-<strong>Breast</strong> 1 study group. Chinese<br />

Academy of Medical Sciences, Beijing, China; Seoul National<br />

University Hospital, Seoul, Korea; National Taiwan University Hospital,<br />

Taipei, Taiwan; Samsung Medical Center, Seoul, Korea; National<br />

<strong>Cancer</strong> Center, Goyang, Korea; Third Affiliated Hospital of Harbin<br />

Medical University, Heilongjiang Province, China; Sujan Surgical<br />

<strong>Cancer</strong> Hospital & Amravati <strong>Cancer</strong> Foundation, Amravati, India;<br />

Central India <strong>Cancer</strong> Research Institute, Nagpur, India; Asan Medical<br />

Center, Seoul, Korea; Taichung Veterans General Hospital, Taichung,<br />

Taiwan; Severance Hospital, Seoul, Korea; Medical University, Gdansk,<br />

Poland; Medical University, Bialystok, Poland; Chang Gung Memorial<br />

Hospital - Linkou Branch, Taoyuan County, Taiwan; SCS Boehringer-<br />

Ingelheim Comm.V, Brussels, Belgium; Boehringer Ingelheim<br />

Pharma GmbH & Co. KG, Biberach, Germany; UCLA, Los Angeles,<br />

CA; University of Munich, Munich, Germany; Institut Jules Bordet,<br />

Brussels, Belgium.<br />

OT1-1-17 LUX-<strong>Breast</strong> 2: Phase II, open-label study of oral afatinib in<br />

HER2-overexpressing metastatic breast cancer (MBC) patients<br />

(pts) who progressed on prior trastuzumab and/or lapatinib*<br />

Hickish T, Mehta A, Jain M, Huang C-S, Kovalenko N, Udovitsa D,<br />

Pemberton K, Uttenreuther-Fischer M, Tseng L-M, On behalf of the<br />

LUX-<strong>Breast</strong> 2 study group. Bournemouth Hospital, Bournemouth<br />

University, Dorset, United Kingdom; Central India <strong>Cancer</strong> Research<br />

Institute, Maharashtra, India; Ruby Hall Clinic, Maharashtra, India;<br />

National Taiwan University Hospital, Taipei, Taiwan; Regional<br />

Clinical Oncology Dispensary, Stavropol, Russian Federation; GUZ<br />

Oncological Dispesary #2, Sochi, Russian Federation; Boehringer<br />

Ingelheim Limited, Bracknell, United Kingdom; Boehringer Ingelheim<br />

Pharma GmbH & Co. KG, Biberach, Germany; Taipei Veterans General<br />

Hospital, National Yang Ming University, Taipei, Taiwan.<br />

Ongoing Trials 1: Radiation Therapy<br />

OT1-2-01 NSABP B-43: A phase III clinical trial to compare trastuzumab<br />

(T) given concurrently with radiation therapy (RT) to RT alone<br />

for women with HER2+ DCIS resected by lumpectomy (Lx)<br />

Cobleigh MA, Anderson SJ, Julian TB, Siziopikou KP, Arthur DW,<br />

Rabinovitch R, Zheng P, Mamounas EP, Wolmark N. National Surgical<br />

Adjuvant <strong>Breast</strong> and Bowel Project, Pittsburgh, PA; Rush University<br />

Medical Center, Chicago, IL; University of Pittsburgh Graduate<br />

School of Public Health, Pittsburgh, PA; Allegheny General Hospital,<br />

Pittsburgh, PA; Northwestern University Feinberg School<br />

of Medicine, Chicago, IL; Virginia Commonwealth University,<br />

Richmond, VA; University of Colorado, Aurora, CO; Aultman Hospital,<br />

Canton, OH.<br />

OT1-2-02 SHARE. A French multicenter phase III trial comparing<br />

accelerated partial irradiation (APBI) versus standard or<br />

hypofractionated whole breast irradiation in low risk of local<br />

recurrence breast cancer<br />

Belkacemi Y, Bourgier C, Kramar A, Lemonier J, Auzac G, Dumas<br />

I, Lacornerie T, Mijonnet S, Mege J-P, Lartigau E. APHP; GH Henry<br />

Mondor and UPEC, Paris, Créteil, France; Institut Gustave Roussy,<br />

Villejuif, France; Oscar Lambret Center, Lille, France; UNICANCER,<br />

Paris, France.<br />

7:30 pm–9:30 pm<br />

OPEN SATELLITE EVENT presented by Research to Practice<br />

Marriott Rivercenter<br />

ONE YEAR LATER: The Practical Application of Research Advances in the<br />

Management of Early and Advanced <strong>Breast</strong> cancer<br />

Website: http://www.researchtopractice.com/Meetings/SA2012<br />

Thursday, December 6, 2012<br />

6:45 am–5:15 pm<br />

Registration<br />

Bridge Hall<br />

7:00 am–9:00 am<br />

Poster Discussion 3: Metformin/Statins<br />

Ballroom A<br />

Viewing 7:00 am<br />

Discussion 7:45 am<br />

Adrian Lee, PhD, Chair<br />

University of Pittsburgh <strong>Cancer</strong> Institute<br />

Pittsburgh, PA<br />

Michael Pollak, MD, Discussant<br />

McGill University<br />

Montreal, CANADA<br />

and<br />

Vered Stearns, MD, Discussant<br />

The Sidney Kimmel Comprehensive <strong>Cancer</strong> Center at<br />

Johns Hopkins<br />

Baltimore, MD<br />

PD03-01 Effect of metformin on apoptosis in a presurgical trial in nondiabetic<br />

patients with breast cancer<br />

Cazzaniga M, DeCensi A, Pruneri G, Puntoni M, Guerrieri-Gonzaga<br />

A, Dell’Orto P, Gentilini OD, Vingiani A, Pagani G, Puccio A, Bonanni<br />

B. European Institute of Oncology (EIO), Milan; Ospaedali Galliera,<br />

Genova; University of Milan.<br />

PD03-02 Evidence for the anti-cancer action of metformin mediated<br />

via tumor AMPK, Akt and Ki67, in a preoperative window of<br />

opportunity trial<br />

Hadad SM, Dowling RJO, Chang MC, Done SJ, Purdie CA, Jordan<br />

LB, Dewar J, Goodwin PJ, Stambolic V, Thompson AM. University<br />

of Sheffield, United Kingdom; University Health Network, Toronto,<br />

Canada; Mount Sinai Hospital, Toronto, Canada; University of<br />

Dundee, United Kingdom; Princess Margaret Hospital and Mount<br />

Sinai Hospital, Toronto, Canada.<br />

PD03-03 Pre-surgical trial of metformin in overweight and obese, multiethnic<br />

patients with newly diagnosed breast cancer<br />

Kalinsky K, Crew KD, Refice S, Wang A, Feldman SM, Taback B,<br />

Hibshoosh H, Maurer M, Hershman DL. Columbia University Medical<br />

Center, New York, NY.<br />

PD03-04 Discovery of metformin derivatives with potent antitumor<br />

activity in triple-negative breast cancer<br />

Márquez-Garbán DC, Deng G, Anderson N, Aivazyan L, Kazmi N, Hamilton<br />

N, Jung ME, Pietras RJ. UCLA, Los Angeles, CA.<br />

PD03-05 Analysis of tumour cell signaling in response to neoadjuvant<br />

metformin in women with early stage breast cancer<br />

Dowling RJO, Niraula S, Chang MC, Done SJ, Ennis M, Hood N,<br />

McCready DR, Leong W, Escallon JM, Reedijk M, Goodwin PJ,<br />

Stambolic V. Ontario <strong>Cancer</strong> Institute, University Health Network,<br />

Toronto, ON, Canada; University Health Network, Toronto, ON,<br />

Canada; Mt. Sinai Hospital, Toronto, ON, Canada; Campbell Family<br />

Institute for <strong>Breast</strong> <strong>Cancer</strong> Research and Laboratory Medicine<br />

Program, University Health Network, Toronto, ON; Campbell Family<br />

Institute for <strong>Breast</strong> <strong>Cancer</strong> Research, Princess Margaret Hospital,<br />

Toronto, ON, Canada; Applied Statistician, Markham, ON, Canada.<br />

www.aacrjournals.org<br />

19s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

PD03-06 Simvastatin radiosensitizes differentiated and stem-like breast<br />

cancer cell lines and is associated with improved local control<br />

in inflammatory breast cancer patients treated with postmastectomy<br />

radiation<br />

Lacerda L, Reddy J, Liu D, Larson R, Masuda H, Brewer T, Debeb B, Xu<br />

W, Hortobagyi GN, Buchholz TA, Ueno NT, Woodward WA. Morgan<br />

Welch Inflammatory <strong>Breast</strong> <strong>Cancer</strong> Research Program and Clinic, The<br />

University of Texas MD Anderson <strong>Cancer</strong> Center, Houston, TX.<br />

PD03-07 Statin-induced decrease in proliferation depends on HMG-CoA<br />

reductase expression in breast cancer<br />

Bjarnadottir O, Romero Q, Bendahl P-O, Rydén L, Rose C, Loman N,<br />

Uhlén M, Jirström K, Grabau D, Borgquist S. Clinical Sciences, Lund<br />

University, Lund, Sweden; School of Biotechnology, KTH - Royal<br />

Institute of Technology, Stockholm, Sweden.<br />

PD03-08 Statin use and improved survival outcome in primary<br />

inflammatory breast cancer: retrospective cohort study<br />

Brewer TM, Masuda H, Iwamoto T, Liu P, Kai K, Barnett CM,<br />

Woodward WA, Reuben JM, Yang P, Hortobagyi GN, Ueno NT. The<br />

University of Texas M.D. Anderson <strong>Cancer</strong> Center, Houston, TX;<br />

Eastern Virginia Medical School, Norfolk, VA; Okayama University<br />

Hospital, Okayama, Japan.<br />

PD03-09 Statins and breast cancer risk: A follow-up analysis of the<br />

Women’s Health Initiative Cohort<br />

Desai P, Jay A, Wu C, Cauley JA, Manson J, Peters U, Agalliu I, Abdul-<br />

Hussein M, Bock C, Budrys N, Chlebowski R, Cote M, Lane D, Luo J,<br />

Martin L, Park H, Petrucelli N, Rosenberg CA, Thomas F, Wactawski-<br />

Wende J, Simon MS. Providence Hospital Medical Center, Southfield,<br />

MI; Wayne State University, Detroit, MI; Fred Hutchinson <strong>Cancer</strong><br />

Research Center, Seattle, WA; University of Pittsburgh, PA; Harvard<br />

School of Medicine, Boston, MA; Albert Einstein College of Medicine,<br />

Bronx, NY; Lakeland Regional Medical Center, MI; Karmanos <strong>Cancer</strong><br />

Institute, Wayne State University, Detroit, MI; University of Texas<br />

Health Science Center <strong>San</strong> <strong>Antonio</strong>, TX; Los Angeles Biomedical<br />

Research Institute at Harbor-UCLA Medical Center, Torrance, CA;<br />

Stony Brook University Medical Center, Stony Brook, NY; West<br />

Virginia University, Morgantown, WV; George Washington University,<br />

Washington, DC; University of California, Irvine, CA; NorthShore<br />

University Health System, Evanston, IL; University of Tennessee<br />

Health Science Center, Memphis, TN; University at Buffalo, NY.<br />

7:00 am–9:00 am<br />

Poster Discussion 4: Local THERAPY Decision Making<br />

and DCIS<br />

Ballroom B<br />

Viewing 7:00 am<br />

Discussion 7:45 am<br />

Richard Crownover, MD, PhD, Chair<br />

UT Health Science Center<br />

<strong>San</strong> <strong>Antonio</strong>, TX<br />

John Benson, MD, PHD, MA, DM, FRCS, Discussant<br />

Addenbrooke’s Hospital<br />

Cambridge, UNITED KINGDOM<br />

and<br />

I-Tien Yeh, MD, Discussant<br />

Virginia Hospital Center<br />

Arlington, VA<br />

PD04-01 Intra-operative ultrasound in breast-conserving surgery for<br />

palpable breast cancer significantly improves both surgical<br />

accuracy and cosmetic outcome while saving costs<br />

Haloua M, Krekel N, Coupe V, van den Tol P, Meijer S. VU Medical<br />

Center, Amsterdam, Nederland, Netherlands; Alkmaar Medical<br />

Center, Alkmaar, Noord-Holland, Netherlands.<br />

PD04-02 Accelerated hypofractionated versus conventional whole<br />

breast radiotherapy for localised left-sided breast cancer: the<br />

effect on long-term cardiac morbidity<br />

Chan EK, Woods R, Virani S, Speers C, Wai ES, Nichol A, McBride ML,<br />

Tyldesley S. British Columbia <strong>Cancer</strong> Agency, Vancouver Centre;<br />

Vancouver General Hospital; British Columbia <strong>Cancer</strong> Agency,<br />

Vancouver Island Centre; British Columbia <strong>Cancer</strong> Agency.<br />

PD04-03 Is breast conservation therapy an option for young women<br />

with operable breast cancer? Local recurrence rates in young<br />

women following surgery: a single centre experience<br />

Lewis CR, Smith R, Matthews A, Choo E, Lee C. Prince of Wales<br />

<strong>Cancer</strong> Centre (POWCC), Randwick, NSW, Australia; Prince of Wales<br />

Hospital, Randwick, NSW, Australia; NHMRC Clinical Trials Unit,<br />

University of Sydney, NSW, Australia.<br />

PD04-04 What is influencing breast conservation rates in the United<br />

States? Data from the National Accreditation Program of<br />

<strong>Breast</strong> Centers<br />

Chagpar AB, Kaufman CS, Connolly J, Burgin C, Granville T,<br />

Winchester D. Yale University School of Medicine, New Haven,<br />

CT; Bellingham <strong>Breast</strong> Center; Beth Israel Deaconess, Boston, MA;<br />

National Accreditation Center for <strong>Breast</strong> Centers; Pat Nolan Center<br />

for <strong>Breast</strong> Health, Glenview, IL.<br />

PD04-05 Molecular predictors for type of recurrence following<br />

conservative treatment for DCIS<br />

Sakr RA, Andrade VP, Chandarlapaty S, Giovanni C, Giri D, Heguy A,<br />

De Brot M, Olvera N, Muhsen S, Koslow S, Van Zee KJ, Morrow M,<br />

Rosen N, King TA. Memorial Sloan-Kettering <strong>Cancer</strong> Center,<br />

New York.<br />

PD04-06 Molecular phenotypes of DCIS predict invasive and DCIS<br />

recurrence<br />

Williams KE, Barnes NLP, Cheema K, Dimopoulos N, Bundred NJ,<br />

Landberg G. The University of Manchester, Manchester, Greater<br />

Manchester, United Kingdom.<br />

PD04-07 The Ki-67 labeling index predicts the risk of recurrence of<br />

DIN patients treated with radiotherapy following breast<br />

conserving surgery<br />

Pruneri G, Lazzeroni M, Guerrieri-Gonzaga A, Botteri E, Leonardi<br />

MC, Rotmensz N, Serrano D, Varricchio C, Disalvatore L, Del Castillo<br />

A, Viale G, Bonanni B. European Institute of Oncology, Milan, Italy;<br />

European Institute of Oncology, Milan; University of Milan, School<br />

of Medicine.<br />

PD04-08 Cell cycle algorithm correlates with grade of DCIS and p53<br />

status, allows elimination of ‘intermediate grade’ disease and<br />

gives clinically meaningful information<br />

Sainsbury R, Loddo M, Proctor I, Stoeber K, Williams G, Thorat M,<br />

Cuzick J. University College London, United Kingdom; Wolfson<br />

Institute of Preventative Medicine, Queen Mary College, London,<br />

United Kingdom.<br />

7:00 am–9:00 am<br />

POSTER SESSION 2 & CONTINENTAL BREAKFAST<br />

Exhibit Halls A-B<br />

P2-01-01 Establishment and validation of circulating tumor<br />

cell-based prognostic nomograms in 497 first-line metastatic<br />

breast cancer patients<br />

Giordano A, Reuben JM, Egleston BL, Hajage D, Hortobagyi GN,<br />

Cristofanilli M, Pierga J-Y, Bidard F-C. The University of Texas MD<br />

Anderson <strong>Cancer</strong> Center, Houston, TX; Fox Chase <strong>Cancer</strong> Center,<br />

Philadelphia, PA; Institut Curie, Paris, France.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012 20s <strong>Cancer</strong> Research


December 4–8, 2012<br />

Program Schedule<br />

P2-01-02<br />

P2-01-03<br />

P2-01-04<br />

P2-01-05<br />

P2-01-06<br />

Circulating Tumor Cells (CTC) may Express HER2/neu in<br />

Patients With Early HER2/neu Negative <strong>Breast</strong> <strong>Cancer</strong> – Results<br />

of the German SUCCESS C Trial<br />

Jaeger BAS, Rack BK, Andergassen U, Neugebauer JK, Melcher CA,<br />

Scholz C, Hagenbeck C, Schueller K, Lorenz R, Decker T, Heinrich<br />

G, Fehm T, Schneeweiss A, Lichtenegger W, Beckmann MW,<br />

Pantel K, Sommer HL, Friese K, Janni W. Klinikum der Ludwig-<br />

Maximilians-Universitaet, Munich, Germany; Heinrich Heine<br />

University, Duesseldorf, Germany; Stat-up Statistische Beratung<br />

und Dienstleistung, Munich, Germany; Gemeinschaftspraxis Dr.<br />

Lorenz/Hecker/Wesche, Braunschweig, Germany; Studienzentrum<br />

Onkologie Ravensburg, Ravensburg, Germany; Praxis Dr. Heinrich,<br />

Fuerstenwalde, Germany; Eberhard Karls Universitaet Tuebingen,<br />

Tuebingen, Germany; National Center for Tumor Disease and<br />

Department of Gynecology and Obstetrics, University Hospital<br />

Heidelberg, Germany; Charité Medical University, Berlin, Germany;<br />

Universitaet Erlangen, Germany; University Medical Center Hamburg-<br />

Eppendorf, Hamburg, Germany.<br />

Truncated HER2 receptor in circulating tumor cells (CTCs) of<br />

early and metastatic breast cancer patients<br />

Kallergi G, Nasias D, Papadaki M, Mavroudis D, Georgoulias V, Agelaki<br />

S. University of Crete, Heraklion, Crete, Greece; University General<br />

Hospital of Heraklion, Crete, Greece.<br />

Long term independent prognostic impact of circulating<br />

tumor cells detected before neoadjuvant chemotherapy<br />

in non-metastatic breast cancer: 70 months analysis of the<br />

REMAGUS02 study<br />

Bidard F-C, Delaloge S, Giacchetti S, Brain E, de Cremoux P, Vincent-<br />

Salomon A, Marty M, Pierga J-Y. Institut Curie, France; Institut<br />

Gustave Roussy, France; Hopital Saint Louis, France.<br />

Parallel DNA and RNA profiling of EpCAM-positive cells in<br />

blood of metastatic breast cancer (MBC) patients confirm their<br />

malignant nature<br />

Magbanua MJM, Hauranieh L, Sosa EV, Pendyala P, Scott JH, Rugo<br />

HS, Park JW. University of California, <strong>San</strong> Francisco, CA.<br />

Association between Circulating Tumor Cells and Bone<br />

Turnover Markers in patients with breast cancer and bone<br />

metastases on treatment with bisphosphonates<br />

(ZOMAR study)<br />

Manso L, Barnadas A, Tusquets I, Crespo C, Gómez P, Calvo L, Ruiz<br />

M, Martínez P, Perelló A, Antón A, Codes M, Margelí M, Murias A,<br />

Salvador J, Seguí MA, De Juán A, Gavilá J, Luque M, Pérez D, Zamora<br />

P, Arizcum A, Chacón JI, Heras L, De la Piedra C. Hospital 12 de<br />

Octubre, Madrid, Spain; Hospital <strong>San</strong>ta Creu i <strong>San</strong>t Pau, Barcelona,<br />

Spain; Hospital del Mar, Barcelona, Spain; Hospital Universitario<br />

Ramón y Cajal, Madrid, Spain; Hospital Universitario Vall d’Hebrón,<br />

Barcelona, Spain; Hospital Universitario A Coruña Juan Canalejo, A<br />

Coruña, Spain; Hospital Universitario Virgen del Rocío, Sevilla, Spain;<br />

Hospital Basurto, Vizcaya, Spain; Hospital Son Dureta, Palma de<br />

Mallorca, Spain; Hospital Miguel Servet, Zaragoza, Spain; Hospital<br />

Virgen Macarena, Sevilla, Spain; Hospital Universitario Trias y Pujol,<br />

Barcelona, Spain; Hospital Universitario Insular de Gran Canaria, Gran<br />

Canaria, Spain; Hospital Nuestra Señora de Valme, Sevilla, Spain;<br />

Hospital Parc Taulí Sabadell, Barcelona, Spain; Hospital Marqués<br />

Valdecilla, <strong>San</strong>tander, Spain; Instituto Valenciano de Oncología,<br />

Valencia, Spain; Hospital General de Asturias, Oviedo, Spain; Hospital<br />

Costa del Sol, Málaga, Spain; Hospital La Paz, Madrid, Spain; Hospital<br />

Palencia Río Carrión, Palencia, Spain; Hospital Virgen de la Salud,<br />

Toledo, Spain; Hospital Cruz Roja Hospitalet del Llobregat, Barcelona,<br />

Spain; Instituto de Investigación <strong>San</strong>itaria Fundación Jiménez Díaz,<br />

Madrid, Spain<br />

P2-01-07<br />

P2-01-08<br />

P2-01-09<br />

P2-01-10<br />

P2-01-11<br />

P2-01-12<br />

P2-01-13<br />

P2-01-14<br />

Effect of first-line chemotherapy in the expression of<br />

stemness and epithelial-to-mesenchymal transition markers<br />

in circulating tumor cells of patients with metastatic<br />

breast cancer<br />

Papadaki MA, Kallergi G, Mavroudis D, Georgoulias V,<br />

Theodoropoulos PA, Agelaki S. University of Crete, Heraklion, Crete,<br />

Greece; University General Hospital of Heraklion, Crete, Greece.<br />

Different numbers and prognostic significance of circulating<br />

tumour cells in patients with metastatic breast cancer<br />

according to immunohistochemical subtypes<br />

Peeters DJ, van Dam P-J, Wuyts H, Van den Eynden GG, Jeuris K,<br />

Prové A, Rutten A, Peeters M, Pauwels P, Van Laere SJ, Hauspy J, van<br />

Dam PA, Vermeulen PB, Dirix LY. GZA Hospitals Sint-Augustinus,<br />

Antwerp, Belgium; University of Antwerp, Belgium; Catholic<br />

University of Leuven, Leuven, Vlaams-Brabant, Belgium.<br />

Tumor cell emboli in the lung and transcriptional profiles<br />

of circulating tumor cells derived from different vascular<br />

compartments in patients with metastatic breast cancer<br />

Peeters DJ, Van den Eynden GG, Rutten A, Onstenk W, Sieuwerts<br />

AM, De Laere B, van Dam PA, Peeters M, Pauwels P, Van Laere SJ,<br />

Vermeulen PB, Dirix LY. GZA Hospitals Sint-Augustinus, Antwerp,<br />

Belgium; University of Antwerp, Belgium; Erasmus University Medical<br />

Center and <strong>Cancer</strong> Genomics Center, Rotterdam, South Holland,<br />

Netherlands; Catholic University of Leuven, Leuven, Vlaams-Brabant,<br />

Belgium.<br />

Immunocytochemistry staining for estrogen and progesterone<br />

receptor in circulating tumor cells: Concordance between<br />

primary and metastatic tumors<br />

Bischoff FZ, Pham T, Wong KL, Villarin E, Xu X, Kalinsky K, Mayer JA.<br />

Biocept, Inc., <strong>San</strong> Diego, CA; Columbia University Medical Center,<br />

New York, NY.<br />

Detection and characterization of circulating and<br />

disseminated tumour cells in blood and bone marrow of<br />

breast cancer patients by two different biochemical methods<br />

Andergassen U, Rack BJ, Zebisch M, Kölbl AC, Schindlbeck C,<br />

Neugebauer J, Liesche F, Hiller Ral, Friese K, Jeschke U. Ludwig -<br />

Maximilians-University, Munich, Germany; Klinikum Traunstein,<br />

Traunstein, Germany.<br />

Circulating tumor cells and Epithelial Mesenchymal Transition<br />

in primary breast cancer patients<br />

Mego M, Karaba M, Minarik G, Benca J, Sedlackova T, Manasova D,<br />

Sieberova G, Gronesova P, Pechan J, Mardiak J, Reuben JM. Faculty of<br />

Medicine, Comenius University, Bratislava, Slovakia (Slovak Republic);<br />

National <strong>Cancer</strong> Institute, Bratislava, Slovakia (Slovak Republic);<br />

<strong>Cancer</strong> Research Institute, Bratislava, Slovakia (Slovak Republic);<br />

University of Texas, MD Anderson <strong>Cancer</strong> Center, Houston, TX.<br />

Prognostic value of Circulating Tumor Cells count at<br />

progressive disease after first line chemotherapy metastatic<br />

breast patients in a large prospective multicenter trial<br />

including serum tumor markers (IC 2006-04 study)<br />

Pierga J-Y, Hajage D, Bachelot T, Delaloge S, Brain E, Campone M,<br />

Asselain B, Cottu PH, Dieras V, Bidard F-C. Institut Curie, Paris; Centre<br />

Léon Bérard, Lyon; Institut Gustave Roussy, Villejuif; Institut de<br />

Cancérologie de L’Ouest.<br />

Circulating tumor cells in breast cancer exhibit dynamic<br />

changes in epithelial and mesenchymal cell composition<br />

Yu M, Bardia A, Wittner BS, Stott SL, Smas ME, Ting DT, Isakoff<br />

SJ, Ciciliano JC, Wells MN, Shah AM, Concannon KF, Sequist LV,<br />

Brachtel E, Sgroi D, Baselga J, Ramaswamy S, Toner M, Haber DA,<br />

Maheswaran S. Massachusetts General Hospital, Harvard Medical<br />

School; Howard Hughes Medical Institute.<br />

www.aacrjournals.org<br />

21s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Detection/Diagnosis: Circulating Markers<br />

P2-02-01 Accurate identification of metastatic breast cancer using<br />

methylated gene markers in circulating free DNA in<br />

peripheral blood<br />

Sukumar S, Fackler MJ, Lopez-Bujanda Z, Teo WW, Jeter S, Umbricht<br />

C, Visvanathan K, Wolff AC. Johns Hopkins University School of<br />

Medicine, Baltimore, MD.<br />

P2-02-02 Prognostic significance of the ratio of absolute neutrophil<br />

to lymphocyte counts for breast cancer patients in<br />

neoadjuvant setting<br />

Koh YW, Lee HJ, Ahn J-H, Lee JW, Gong G. University of Ulsan<br />

College of Medicine, Asan Medical Center, Seoul, Korea; Seoul<br />

National University Bundang Hospital, Seongnam, Bundang-Gu,<br />

Kyeonggi-Do, Korea.<br />

P2-02-03 The Collagen Receptor Endo180: A Metastatic Plasma Marker<br />

in <strong>Breast</strong> <strong>Cancer</strong> Modulated by Bisphosphonate Treatment<br />

Sturge J, Caley MP, Purshouse K, Fonseca A-V, Rodriguez-Teja M,<br />

Kogianni G, Waxman J, Palmieri C. Imperial College London, United<br />

Kingdom; The University of Hull, Hull, United Kingdom.<br />

P2-02-04 The interaction between menopausal status and<br />

zoledronic acid can differentially affect serum levels of the<br />

TGFβ superfamily<br />

Wilson C, Winter MC, Holen I, Freeman JV, Evans AC, Coleman RE.<br />

University of Sheffield, United Kingdom; Sheffield Teaching Hospitals<br />

NHS Trust, Sheffield, United Kingdom.<br />

Detection/Diagnosis: Micrometastases<br />

P2-03-01 Identification of cancer stem cells (CD44+CD24-/lo) in bone<br />

marrow as a prognostic factor in early breast cancer patients<br />

Giordano A, Gao H, Cohen EN, Anfossi S, Hess KR, Krishnamurthy S,<br />

Tin S, Cristofanilli M, Hortobagyi GN, Woodward WA, Ueno NT, Lucci<br />

A, Reuben JM. The University of Texas MD Anderson <strong>Cancer</strong> Center,<br />

Houston, TX; The University of Texas MD Anderson <strong>Cancer</strong> Center;<br />

Fox Chase <strong>Cancer</strong> Center.<br />

Tumor Cell and Molecular Biology: Animal Models<br />

P2-04-01 Development of mouse breast cancer models based on<br />

induced cancer stem cells (iCSC)<br />

Takamoto Y, Onishi N, Kai K, Saya H. Institute for Advanced Medical<br />

Research (IAMR), School of Medicine, Keio university, Shinjyuku-ku,<br />

Tokyo, Japan; The University of Texas MD Anderson <strong>Cancer</strong> Center,<br />

Houston, TX.<br />

P2-04-02 Identification of genes associated with breast cancer<br />

micrometastatic disease in bone marrow using a<br />

human-in-mouse xenograft system<br />

Aft R, Li S, Mudalagiriyappa C, Dasgupta N, Watson M, Fleming T, Ellis<br />

M, Pillai S. Washington University, St. Louis, MO.<br />

P2-04-03 A robust transgenic mouse model to study male breast cancer<br />

Krause S, Lurvey HL, Tobin H, Ingber DE. Boston’s Children’s Hospital,<br />

Boston, MA; Wyss Institute of Biologically Inspired Engineering,<br />

Boston, MA.<br />

P2-04-04 Prolactin cooperates with loss of p53 to promote<br />

mammary tumorigenesis<br />

O’Leary KA, Sullivan R, Rugowski DE, Schuler LA. University of<br />

Wisconsin, Madison, WI.<br />

P2-04-05 Prolonged targeted overexpression of Aurora-A in mammary<br />

epithelium promotes mammary adenocarcinoma with<br />

genomic instability<br />

Treekitkarnmongkol W, Katayama H, Sen S. University of Texas M.D.<br />

Anderson <strong>Cancer</strong> Center, Houston, TX.<br />

P2-04-06 Transgenic expression of a breast-cancer specific GATA3<br />

mutant leads to mammary hyperplasia<br />

Kenny PA, Chandiramani N. Albert Einstein College of Medicine,<br />

Bronx, NY.<br />

P2-04-07 Modeling orthotopic and metastatic progression of<br />

mammary tumors to evaluate the efficacy of TGF-β inhibitors<br />

in a pre-clinical setting<br />

Biswas T, Yang J, Zhao L, Sun L. UTHSCSA, <strong>San</strong> <strong>Antonio</strong>, TX;<br />

UCSD, CA.<br />

P2-04-08 Targeted Expression of the Human Chaperone BAG3 to the<br />

Murine Mammary Gland Dysregulates Mammary Gland<br />

Development and Differentiation By Unrestricted Expansion of<br />

Luminal Cells<br />

Virador VM, Casagrange G, Raafat A, Callahan R, Kohn E. National<br />

<strong>Cancer</strong> Institute, Bethesda, MD.<br />

Tumor Cell and Molecular Biology: Biomarkers<br />

P2-05-01 Gene expression changes associated with response to<br />

neoadjuvant chemotherapy are observed early in treatment:<br />

results from the I-SPY 1 TRIAL (CALGB 150007/150012;<br />

ACRIN 6657)<br />

Wolf DM, Yau C, Magbanua M, Boudreau A, Davis S, Haqq C, Park J,<br />

I-SPY TRIAL-1 Investigators, Esserman L, van’t Veer L. University of<br />

California, <strong>San</strong> Francisco, CA; I-SPY 1 TRIAL Institutions.<br />

P2-05-02 Clinical significance of microRNA regulator Lin28 expression in<br />

patient with early breast cancer<br />

Park S, Shin YK, Song BJ, Chae BJ, Jung SS, Choi Y-L. Seoul St. Mary’s<br />

Hospital, Catholic University School of Medicine, Seoul, Korea; Seoul<br />

National University College of Pharmacy, Seoul, Korea; Samsung<br />

Medical Center, Sungkyunkwan University School of Medicine,<br />

Seoul, Korea.<br />

P2-05-03 Mouse double minute 2 nuclear expression as a prognostic<br />

marker in patients with breast cancer<br />

Park HS, Park S, Koo JS, Cho JH, Park JM, Kim S-I, Park B-W. Yonsei<br />

University College of Medicine, Seodaemoon-gu, Seoul, Korea.<br />

P2-05-04 Differential potency of TORC1/C2 (INK/MLN-128) and pan-PI3K<br />

(GDC0941) inhibitors on breast cancer polysome composition<br />

and phosphoprotein response biomarkers<br />

Wilson-Edell KA, Yevtushenko M, Hanson I, Rogers AN, Scott GK,<br />

Benz CC. Buck Institute for Research on Aging, Novato, CA.<br />

P2-05-05 Circulating HER2 extracellular domain (ECD) predicts a poor<br />

prognosis for metastatic breast cancer patients<br />

Wang X-j, Shao X-Y, Xu X-H, Chen Z-H, Wang S, Cai J-F, Huang J,<br />

Huang P, Lou C-J, Ling Z-Q, Han MX. Zhejiang <strong>Cancer</strong> Hospital,<br />

Hangzhou, Zhejiang, China; Hangzhou Polar Gene Company,<br />

Hangzhou, Zhejiang, China.<br />

P2-05-06 Quantitative measurement of HER2 expression in breast<br />

cancers: comparison with “real world” HER2 testing in a<br />

multi-center Collaborative Biomarker Study (CBS) and<br />

correlation with clinicopathological features<br />

Yardley DA, Kaufman PA, Adams JW, Krekow L, Savin M, Lawler WE,<br />

Zrada S, Starr A, Einhorn H, Schwartzberg LS, Huang W, Weidler J, Lie<br />

Y, Paquet A, Haddad M, Anderson S, Brigino M, Bosserman L. Sarah<br />

Cannon Research Institute, Nashville, TN; Tennessee Oncology PLLC,<br />

Nashville, TN; Dartmouth Hitchcock Medical Center, Lebanon, NH;<br />

Arlington <strong>Cancer</strong> Center, Arlington, TX; Texas Oncology Bedford,<br />

Bedford, TX; Texas Oncology at Medical City Dallas 2, Dallas, TX; St.<br />

Jude Heritage Medical Group, Fullerton, CA; The Center for <strong>Cancer</strong><br />

and Hematologic Disease, Cherry Hill, NJ; Monroe Medical Associates,<br />

Harvey, IL; Swedish American Regional <strong>Cancer</strong> Center, Rockford,<br />

IL; The West Clinic, Memphis, TN; Monogram Biosciences, Inc., So.<br />

<strong>San</strong> Francisco, CA; Center for Molecular Biology and Pathology,<br />

Laboratory Corporation of America, Inc., Research Triangle Park, NC;<br />

Wilshire Oncology Medical Group, Rancho Cucamonga, CA.<br />

P2-05-07 Comparison of hormone receptor and HER-2 expression in<br />

primary breast cancers and sentinel lymph node metastases<br />

Rokutanda N, Horiguchi J, Takata D, Nagaoka R, Sato A, Tokiniwa H,<br />

Tozuka K, Uchida S, Takeyoshi I. Gunma University Graduate School<br />

of Medicine, Maebashi, Gunma, Japan.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012 22s <strong>Cancer</strong> Research


December 4–8, 2012<br />

Program Schedule<br />

P2-05-08<br />

P2-05-09<br />

P2-05-10<br />

P2-05-11<br />

P2-05-12<br />

P2-05-13<br />

P2-05-14<br />

P2-05-15<br />

P2-05-16<br />

P2-05-17<br />

Determination of HER2 amplification by dual-color in situ<br />

hybridization before and after neoadjuvant chemotherapy<br />

Niikura N, Kumaki N, Iwamoto T, Tsuda B, Okamura T, Yuki S, Suzuki<br />

Y, Tokuda Y. Tokai University School of Medicine, Isehara, Kanagawa,<br />

Japan; Okayama University Hospital, Okayama, Japan.<br />

Identification and validation of a miRNA signature associated<br />

with breast cancer progression<br />

Waters PS, Dwyer RM, Kerin MJ. National University of Ireland,<br />

Galway, Ireland.<br />

64<br />

Cu-DOTA-trastuzumab positron emission tomography<br />

imaging of HER2 in women with advanced breast cancer<br />

Mortimer JE, Conti P, Shan T, Carroll M, Kofi P, Colcher D, Raubitschek<br />

AA, Bading JR, Miles J. City of Hope <strong>Cancer</strong> Center/Beckman<br />

Research Institute, Duarte, CA; USC, Los Angeles, CA.<br />

Proteomic identification and in-silico verification of subtypespecific<br />

signatures in breast cancer<br />

Pavlou MP, Dimitromanolakis A, Diamandis EP. University of Toronto,<br />

ON, Canada; Mount Sinai Hospital, Toronto, ON, Canada; University<br />

Health Network, Toronto, ON, Canada.<br />

Effects of de-escalated bisphosphonate therapy on bone<br />

turnover or metastasis markers and their correlation with<br />

risk of skeletal related events – A biomarker analysis in<br />

conjunction with the REFORM study<br />

Addison CL, Zhao H, Mazzarello S, Mallick R, Amir E, Tannock I,<br />

Clemons M. Ottawa Hospital Research Institute, Ottawa, ON, Canada;<br />

Princess Margaret Hospital, Toronto, ON, Canada; The Ottawa<br />

Hospital <strong>Cancer</strong> Centre, Ottawa, ON, Canada.<br />

Correlation of conventional versus experimental biomarkers<br />

of bone turnover and metastasis behaviour with skeletal<br />

related events – A biomarker analysis in conjunction with the<br />

TRIUMPH study<br />

Addison CL, Kuchuk I, Bouganim N, Zhao H, Mazzarello S,<br />

Vandermeer L, Mallick R, Goss GD, Clemons M. Ottawa Hospital<br />

Research Institute, Ottawa, ON, Canada; The Ottawa Hospital <strong>Cancer</strong><br />

Centre, Ottawa, ON, Canada; McGill University Health Centre,<br />

Montreal, QC, Canada.<br />

Clinical significance of Lysyl oxidase-like 2 (LOXL 2) in<br />

breast cancer<br />

Ahn SG, Lee HM, Hwang SH, Lee SA, Jeong J, Lee H-E. Gangnam<br />

Severance Hospital, Yonsei University Medical College, Seoul,<br />

Republic of Korea.<br />

Assessment of Notch Signaling Pathway Components as<br />

Biomarkers for Triple Negative <strong>Breast</strong> <strong>Cancer</strong>: Comparison of<br />

Triple Negative <strong>Breast</strong> <strong>Cancer</strong> Cell Lines and Human <strong>Breast</strong><br />

<strong>Cancer</strong> Samples<br />

Zlobin A, Olsauskas-Kuprys R, Hodge S, O’Toole M, Ersahin C, Osipo<br />

C. Loyola University Chicago, Cardinal Bernardin <strong>Cancer</strong> Center,<br />

Maywood, IL.<br />

Expression of β-III tubulin, foxo 3 protein and deoxythymidine<br />

kinase in breast cancer patients receiving neoadjuvant<br />

chemotherapy<br />

Chow LWC, Loo WTY, Yip AYS, Ong EYY, Ng W-K. UNIMED Medical<br />

Institute, Hong Kong; Organisation for Oncology and Translational<br />

Research, Hong Kong; Central Hospital, Hong Kong.<br />

Correlation between cyclin D1, estrogen and progesterone<br />

receptors in invasive breast cancer after short-term treatment<br />

with tamoxifen or anastrozole<br />

Millen EC, Mattar A, Logullo AF, Nonogaki S, Soares FA, Gebrim LH.<br />

Federal University of Sao Paulo, Sao Paulo, SP, Brazil; Perola Byington<br />

Hospital, Sao Paulo, SP, Brazil; Federal University of Sao Paulo, Sao<br />

Paulo, SP; Arnaldo Vieira de Carvalho, Sao Paulo, SP, Brazil.<br />

P2-05-18 Withdrawn<br />

P2-05-19 Pathway-based analysis of breast cancer<br />

Song D, Cui M, Fan Z, Yang Y, Xue L, Zhang DY, Ye F. The First<br />

Hospital of Jilin University, Changchun, Jilin, China; The Mount Sinai<br />

Medical Center, New York, NY; Nanfang Hospital Southern Medical<br />

University, Guangzhou, China.<br />

P2-05-20 Validation and comparison of CS-IHC4 score with a nomogram<br />

based on Ki67 index, Adjuvant! Online, and St. Gallen risk<br />

stratification to predict recurrence in early Hormone Receptor<br />

(HR)-positive breast cancers<br />

Park YH, Im S-A, Cho EY, Ahn JH, Woo SY, Kim S, Keam B, Lee JE,<br />

Han W, Nam SJ, Park IA, Noh D-Y, Yang JH, Ahn JS, Im Y-H. Samsung<br />

Medical Center; Seoul National University College of Medicine.<br />

P2-05-21 Molecular subtypes of invasive breast cancers show<br />

differential expression of the proliferation marker Aurora<br />

Kinase A (AURKA)<br />

Kovatich AJ, Luo C, Chen Y, Hooke JA, Kvecher L, Rui H, Shriver CD,<br />

Mural RJ, Hu H. Windber Research Institute, Windber, PA; Walter<br />

Reed National Military Medical Center, Bethesda, MD; MDR, Global<br />

Systems LLC, Windber, PA; Thomas Jefferson University,<br />

Philadelphia, PA.<br />

P2-05-22 Preferential Activation of BP1 and c-Myc in <strong>Breast</strong> <strong>Cancer</strong> of<br />

African American Women Compared with Caucasian<br />

American Women<br />

Berg PE, Ghimbovschi S, Grizzle WE, Nguyen TH. George Washington<br />

University Medical Center, Washington, DC; Children’s National<br />

Medical Center, Washington, DC; University of Alabama at<br />

Birmingham, AL.<br />

Tumor Cell and Molecular Biology: Genomics<br />

P2-06-01 <strong>Breast</strong>-to-breast metastasis can cause hormone-receptor<br />

positive / triple negative bilateral synchronous tumors<br />

Schwab RB, Bao L, Pu M, Crain B, Dai Y, Nazareth LV, Matsui H,<br />

Wallace AM, Hasteh F, Harismendy O, Frazer KA, Parker BA, Messer<br />

K. University of California, <strong>San</strong> Diego, La Jolla, CA; Rady Children’s<br />

Hospital, Division of Genome Information Sciences, University of<br />

California, <strong>San</strong> Diego, La Jolla, CA.<br />

P2-06-02 Genomic evolution from primary breast cancers to distant<br />

metastases<br />

Hoefnagel LDC, Moelans CB, van der Groep P, van der Wall E, van<br />

Diest PJ. University Medical Center Utrecht, Netherlands.<br />

P2-06-03 Specific Transcriptional Response of Four Blockers of Estrogen<br />

Receptors on Estrogen-Modulated Genes in ZR-75-1 <strong>Breast</strong><br />

<strong>Cancer</strong> Xenografts<br />

Calvo E, Luu-The V, Martel C, Labrie F. Laval University Hospital<br />

Research Center (CRCHUL), Quebec City, QC, Canada; Laval<br />

University, Quebec City, QC, Canada; EndoCeutics Inc., Quebec City,<br />

QC, Canada.<br />

P2-06-04 Impact of genomic testing on chemotherapy utilization<br />

Riley L, Nakijima H, Balinger K, Anderson D. St. Luke’s University<br />

Health Network, Bethlehem, PA.<br />

P2-06-05 Single-cell RNA sequencing of paclitaxel-treated breast cancer<br />

cell lines to find individual cell response<br />

Vaske CJ, Lee W, Benz SC, <strong>San</strong>born JZ, Emerson BM, Pourmand N,<br />

Lopez Diaz F. Five3 Genomics, LLC, <strong>San</strong>ta Cruz, CA; University of<br />

California, <strong>San</strong>ta Cruz, CA; Salk Institute, La Jolla, CA.<br />

P2-06-06 Integrating multiple molecular profiles with pathways to learn<br />

sub-type specific interactions with PARADIGM<br />

Sedgewick AJ, Vaske CJ, Benz SC. Five3 Genomics, <strong>San</strong>ta Cruz, CA;<br />

University of Pittsburgh, PA.<br />

www.aacrjournals.org<br />

23s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P2-06-07 Exome sequencing identifies somatic mutations in basal-like<br />

breast cancer before and after neoadjuvant chemotherapy<br />

Jiang Y-Z, Yu K-D, Shao Z-M. Shanghai <strong>Cancer</strong> Center, Shanghai,<br />

Shanghai, China.<br />

Tumor Cell and Molecular Biology: Genetics - Germline Changes<br />

P2-07-01 BRCA1 regulates RHAMM function in breast cancer<br />

Fleisch MC, Yang Y, Mei Q, Sadat F, Brandi L, Iwaniuk KM, Honisch E,<br />

Maxwell CA, Niederacher D. Heinrich Heine University, Duesseldorf,<br />

Germany; University of British Columbia, Vancouver, BC, Canada.<br />

P6-07-31 Assessing germline Homologous Recombination pathway<br />

deficiency in BRCA1 mutation carriers using Single Cell<br />

Network Profiling<br />

Rosen DB, Leung LY, Louie B, Evensen E, Fields SZ, Cesano A, Shapira<br />

I, Hawtin RE. Nodality Inc, South <strong>San</strong> Francisco, CA; Monter <strong>Cancer</strong><br />

Center, North Shore Long Island Jewish Medical School, Lake<br />

Success, NY.<br />

Tumor Cell and Molecular Biology: Genetics - Somatic Changes<br />

P2-08-01 PIK3CA mutations associate with decreased Ki67 in early<br />

stage breast cancer (BC) and better outcomes in patients even<br />

among those with low Ki67 tumors<br />

Datko FM, Patil S, Kalinsky K, Asher M, Wen YH, Hedvat C, Moynahan<br />

ME. Memorial Sloan-Kettering <strong>Cancer</strong> Center; Columbia University<br />

Medical Center.<br />

P2-08-02 The large scale structure of the HER-2/neu amplicon in<br />

breast cancer<br />

Pauletti G, Anderson L, Rong H-M, Yu J, Slamon DJ. David Geffen<br />

School of Medicine, University of California, Los Angeles, CA.<br />

P2-08-03 Phosphatidylinositol-3-kinase mutations are common in<br />

lobular neoplasia<br />

Troxell ML, Ang D, Warrick A, Beadling C, Corless CL. Oregon Health<br />

& Science University, Portland, OR.<br />

P2-08-04 HER2 gene status change after taxane based chemotherapy:<br />

be aware of mis-interpretation of polyploidization! Impact for<br />

patient management<br />

Valent A, Penault-Llorca F, Cayre A, Kroemer G. Institut Gustave<br />

Roussy, Villejuif, France; Centre Jean Perrin, Clermont-Ferrand,<br />

France.<br />

Tumor Cell and Molecular Biology: Epigenetics<br />

P2-09-01 Reactivation of epigenetically silenced retinoic acid receptorbeta<br />

for therapy of breast cancer- from molecular mechanism<br />

to potential clinical applications<br />

Ordentlich P, Nguyen N, Jin K, Sadik H, Han L, Sukumar S. Syndax<br />

Pharmaceuticals; Sidney Kimmel <strong>Cancer</strong> Center, Johns Hopkins<br />

University School of Medicine.<br />

P2-09-02 Epigenetic Regulation of histone variants - a role in hormone<br />

therapy resistant breast cancer?<br />

Nayak SR, Oesterreich S, Pathiraja T. Magee Women’s Research<br />

Institute, University of Pittsburgh Medical Center, Pittsburgh, PA;<br />

Lester & Sue Smith <strong>Breast</strong> Center, Baylor College of Medicine,<br />

Houston, TX.<br />

P2-09-03 Identifying a landscape of DNA methylation-driven genes in<br />

breast cancer using MethylMix<br />

Gevaert O, Plevritis S. Stanford University.<br />

P2-09-04 DNA methylation landscapes in ER-negative breast cancer<br />

Worsham MJ, Chen KM, Chitale D, Divine G. Henry Ford Health<br />

System, Detroit, MI.<br />

P2-09-05 Screening of significantly hypermethylated genes in breast<br />

cancer using MIRA-based microarray and identifying their<br />

expression levels<br />

Lian Z-q, Wang Q, Li W-p, Zhang A-q, Wu L. <strong>Breast</strong> Disease Center,<br />

Guangdong Women and Children Hospital of Guangzhou<br />

Medical College.<br />

P2-09-06 The structure design and biological activities of inhibitory<br />

peptides, which block the interactions among polycomb<br />

repressive complex 2<br />

LI KK, Luo L, Kong X, Li L, Luo C. Rui Jin Hospital Affiliated with the<br />

Shanghai JiaoTong University School of Medicine, Shanghai, China;<br />

Shanghai Institute of Materia Medica, Chinese Academy of Sciences,<br />

Shanghai, China.<br />

Prognostic and Predictive Factors: Prognostic Factors - Clinical<br />

P2-10-01 PAM50 gene signature is prognostic for breast cancer patients<br />

treated with adjuvant anthracycline and taxane based<br />

chemotherapy<br />

Liu MC, Pitcher BN, Mardis ER, Davies SR, Snider JE, Vickery T, Reed JP,<br />

DeSchryver K, Singh B, Friedman PN, Gradishar WJ, Perez EA, Martino<br />

S, Citron ML, Norton L, Winer EP, Hudis CA, Perou CM, Ellis MJ,<br />

Barry WT. Georgetown University Lombardi Comprehensive <strong>Cancer</strong><br />

Center, Washington, DC; Duke University Medical Center, Durham,<br />

NC; Washington University, St. Louis, MO; Washington University<br />

School of Medicine, St. Louis, MO; New York University Medical<br />

Center, New York, NY; University of Chicago, IL; Robert H. Lurie<br />

Comprehensive <strong>Cancer</strong> Center, Northwestern University Feinberg<br />

School of Medicine, Chicago, IL; Mayo Clinic, Jacksonville, FL; The<br />

Angeles Clinic and Research Institute, <strong>San</strong>ta Monica, CA; Hofstra<br />

North Shore-LIJ School of Medicine, ProHEALTH Care Associates,<br />

Lake Success, NY; Memorial Sloan-Kettering <strong>Cancer</strong> Center, New<br />

York, NY; Dana Farber <strong>Cancer</strong> Institute, Harvard Medical School,<br />

Boston, MA; Lineberger Comprehensive <strong>Cancer</strong> Center, University<br />

of North Carolina at Chapel Hill, NC; Siteman <strong>Cancer</strong> Center,<br />

Washington University School of Medicine, St. Louis, MO.<br />

P2-10-02 Clinical validation of the PAM50 risk of recurrence (ROR) score<br />

for predicting residual risk of distant-recurrence (DR) after<br />

endocrine therapy in postmenopausal women with ER+ early<br />

breast cancer (EBC): An ABCSG study<br />

Gnant M, Filipits M, Mlineritsch B, Dubsky P, Jakesz R, Kwasny<br />

W, Fitzal F, Rudas M, Knauer M, Singer C, Greil R, Ferree S,<br />

Storhoff J, Cowens JW, Schaper C, Liu S, Nielsen T, On behalf of<br />

the ABCSG. Medical University of Vienna, Austria; Medizinische<br />

Universität Salzburg, Austria; A.ö. KH Wr. Neustadt, Wr. Neustadt,<br />

Austria; Hospital of the Sisters of Mercy, Linz, Austria; Nanostring<br />

Technologies, Seattle, WA; Vancouver Hospital, Vancouver, BC,<br />

Canada; MyRAQA Inc, Redwood Shores, CA; Vancouver Coastal<br />

Health, Vancouver, BC, Canada.<br />

P2-10-03 A cross-platform comparison of genomic signatures and<br />

OncotypeDx score to discover potential prognostic/predictive<br />

genes and pathways<br />

Kuderer NM, Barry WT, Geradts J, Ginsburg GS, Lyman GH, Datto<br />

M, Liotcheva V, Isner P, Veldman T, Agarwal P, Hwang S, Ready N,<br />

Marcom PK. Duke University Medical Center, Durham, NC.<br />

P2-10-04 The Mammostrat Test Is an Effective Tool To Stratify Patient<br />

Samples Previously Characterized as Intermediate by the<br />

Oncotype Dx Test<br />

Bloom KJ, Kyshtoobayeva A, Hilaire L, Gutekunst K. Clarient<br />

Diagnostic Services, Aliso Viejo, CA.<br />

P2-10-05 Continuous association of a 200-gene prognostic risk score<br />

with probability of neoadjuvant chemotherapy response and<br />

translation from fresh frozen to FFPE tissue for clinical use<br />

van Laar R, DiPaola J, Carbaugh H, Murphy T, DiRamio A, Flinchum R,<br />

Brown N, Albino T. Signal Genetics, New York, NY.<br />

P2-10-06 Multicenter I-SPY 1 TRIAL (CALGB 150007/150012): <strong>Breast</strong><br />

cancer stem cells are associated with intrinsic chemoresistance<br />

and worse survival<br />

Landis MD, Yau C, Neumeister V, Rimm DL, I-SPY 1 TRIAL<br />

Investigators, Esserman L, Chang JC. The Methodist Hospital,<br />

Houston, TX; University of California, <strong>San</strong> Francisco, CA; Yale<br />

University School of Medicine, New Haven, CT.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012 24s <strong>Cancer</strong> Research


December 4–8, 2012<br />

Program Schedule<br />

P2-10-07<br />

P2-10-08<br />

P2-10-09<br />

P2-10-10<br />

P2-10-11<br />

P2-10-12<br />

Stem cell marker aldehyde dehydrogenase 1 in stromal cells<br />

strongly correlates with prognosis in breast cancer<br />

Sjöström M, Hartman L, Holmdahl J, Grabau D, Malmström P, Fernö<br />

M, Niméus-Malmström E. Lund University, Lund, Sweden.<br />

Prospective comparison of Recurrence Score and different<br />

definitions of luminal subtypes by central pathology<br />

assessment of single markers in early breast cancer: results<br />

from the phase III WSG-planB Trial<br />

Gluz O, Kreipe HH, Kates RE, Christgen M, Liedtke C, Shak S,<br />

Clemens M, Salem M, Liedtke B, Aktas B, Markmann S, Uleer C,<br />

Augustin D, Thomssen C, Nitz U, Harbeck N. West German Study<br />

Group, Moenchengladbach, NRW, Germany; Ev. Bethesda Hospital,<br />

Moenchengladbach, NRW, Germany; Medizinische Hochschule<br />

Hannover, Niedersachsen, Germany; University of Muenster, Muenster,<br />

Germany; Genomic Health, Inc., Germany; Clinics Mutterhaus, Trier,<br />

Germany; University of Cologne, Germany; Ev. Hospital, Bergisch<br />

Gladbach, Germany; University of Essen, Germany; Clinics Südstadt,<br />

Rostock, Germany; Gemeinschaftspraxis Hildesheim, Hildesheim,<br />

Germany; Clinics Deggendorf, Deggendorf, Germany; University of<br />

Halle, Germany; University of Munich, Munich, Bavaria, Germany.<br />

The incidence of false negative of HER2/Neu status in primary<br />

breast cancer in the era of standardized testing: a Canadian<br />

prospective study<br />

Hanna W, Barnes PJ, Chang MC, Gilks B, Magliocco A, Rees H,<br />

Robertson S, SenGupta SK, Nofech-Mozes S. Sunnybrook Health<br />

Sciences Centre; Capital Health District Authority; Mount Sinai<br />

Hospital; Vancouver General Hospital; Tom Baker <strong>Cancer</strong> Centre;<br />

Saskatoon City Hospital; Ottawa General Hospital;<br />

Kingston General Hospital.<br />

Clinical implications of molecular heterogeneity in highly<br />

proliferative, ER-positive, HER2-negative breast cancer<br />

Bianchini G, Pusztai L, Kelly CM, Iwamoto T, Callari M, Symmans WF,<br />

Gianni L. Ospedale <strong>San</strong> Raffaele, Milan, Italy; MD Anderson <strong>Cancer</strong><br />

Center, Houston, TX; Mater Misericordiae University Hospital, Dublin,<br />

Ireland; Okayama University Hospital, Okayama, Japan; Fondazione<br />

IRCCS Istituto Nazionale dei Tumori, Milan, Italy.<br />

Prognostic performance of the EndoPredict score in nodepositive<br />

chemotherapy-treated ER+/HER2- breast cancer<br />

patients: results from the GEICAM/9906 trial<br />

Martin M, Brase JC, Ruiz-Borrego M, Krappmann K, Munarriz B, Fisch<br />

K, Ruiz A, Weber KE, Crespo C, Petry C, Rodriguez CA, Kronenwett R,<br />

Calvo L, Alba E, Carrasco E, Casas M, Caballero R, Rodriguez-Lescure<br />

A. Hospital General Universitario Gregorio Marañon, Madrid, Spain;<br />

Sividon Diagnostics GmbH, Cologne, Germany; Hospital Universitario<br />

Virgen del Rocio, Sevilla, Spain; Hospital Universitario La Fe,<br />

Valencia, Spain; Instituto Valenciano de Oncologia, Valencia, Spain;<br />

Hospital Universitario Ramon y Cajal, Madrid, Spain; Hospital Clinico<br />

Universitario de Salamanca, Salamanca, Spain; Complejo Hospitalario<br />

Universitario A Coruña, Coruña, Spain; Hospital Universitario Virgen<br />

de la Victoria, Malaga, Spain; GEICAM (Spanish <strong>Breast</strong> <strong>Cancer</strong><br />

Research Group), Madrid, Spain; Hospital General Universitario de<br />

Elche, Alicante, Spain.<br />

How well predict the 2011 St Gallen early breast cancer<br />

surrogate phenotypes metastatic survival?<br />

Vanderstichele A, Brouckaert O, Tuyls S, Amant F, Leunen K, Smeets<br />

A, Berteloot P, Van Limbergen E, Weltens C, Peeters S, Moerman<br />

P, Floris G, Paridaens R, Wildiers H, Vergote I, Christiaens M-R, Van<br />

Calster B, Neven P. University Hospitals, Leuven, Belgium.<br />

CD4 positive tumor-infiltrating lymphocytes are associated<br />

with improved prognosis in node-negative breast cancer<br />

Schmidt M, van de <strong>San</strong>dt L, Boehm D, Sicking I, Battista M, Lebrecht<br />

A, Solbach C, Koelbl H, Gehrmann M, Rahnenführer J, Hengstler JG.<br />

University Hospital, Mainz, Germany; Technical University, Dortmund,<br />

Germany; Bayer, Leverkusen, Germany; Leibniz Research Centre<br />

for Working Environment and Human Factors (IfADo), Dortmund,<br />

Germany.<br />

P2-10-14<br />

P2-10-15<br />

P2-10-16<br />

P2-10-17<br />

P2-10-18<br />

P2-10-19<br />

Triple Negative <strong>Breast</strong> <strong>Cancer</strong>: prognosis of triple-negative<br />

breast cancers and non-triple-negative breast cancers in a<br />

large registry of certified breast units<br />

Kern P, Rezai M. <strong>Breast</strong> Unit Düsseldorf Louis Hospital, Düsseldorf,<br />

Northrhine-Westfalia, Germany.<br />

Evaluation of Prognostic and Predictive Performance of <strong>Breast</strong><br />

<strong>Cancer</strong> Index and Its Components in Hormonal Receptor-<br />

Positive <strong>Breast</strong> <strong>Cancer</strong> Patients: A TransATAC Study<br />

Sgroi DC, Sestak I, Zhang Y, Erlander MG, Schnabel CA, Goss PE,<br />

Cuzick J, Dowsett M. Massachusetts General Hospital and Harvard<br />

Medical School, Boston, MA; Queen Mary University, London,<br />

United Kingdom; bioTheranostics Inc, <strong>San</strong> Diego, CA; Royal Marsden<br />

Hospital, London, United Kingdom.<br />

Quantitative HER3 protein expression and PIK3CA mutation<br />

status in matched samples from primary and metastatic breast<br />

cancer tissues and correlation with time to recurrence<br />

Sperinde J, Lara J, Michaelson R, Sun X, Conte P, Guarneri V, Barbieri<br />

E, Ali S, Leitzel K, Weidler J, Lie Y, Cook J, Haddad M, Paquet A,<br />

Winslow J, Howitt J, Hurley L, Eisenberg M, Petropoulos C, Huang<br />

W, Lipton A. Monogram Biosciences/Integrated Oncology/LabCorp,<br />

South <strong>San</strong> Francisco, CA; Saint Barnabas Medical Center, Livingston,<br />

NJ; University of Modena, Modena, Italy; Penn State/Hershey Medical<br />

Center, Hershey, PA; Lebanon VA Medical Center, Lebanon, PA;<br />

Laboratory Corporation of America, Research Triangle Park, NC.<br />

SET index predicts response to endocrine therapy rather than<br />

prognosis independently of other genomic signatures in a<br />

blinded validation study<br />

Karn T, Hatzis C, Symmans F, Pusztai L, Ruckhäberle E, Schmidt M,<br />

Müller V, Hanker L, Heinrich T, Holtrich U, Kaufmann M, Rody A.<br />

Goethe-University; Nuvera Biosciences; The University of Texas MD<br />

Anderson <strong>Cancer</strong> Center; Gutenberg-University Mainz; University<br />

Hospital Hamburg-Eppendorf; Saarland-University, Homburg.<br />

Cell Cycle Profiling – risk score (C2P-RS) based on the specific<br />

activity of CDK1 and CDK2 predicts relapse in node-negative,<br />

hormone receptor-positive breast cancer treated with<br />

endocrine therapy<br />

Kim SJ, Masuda N, Tsukamoto F, Inaji H, Akiyama F, Sonoo H,<br />

Kurebayashi J, Yoshidome K, Tsujimoto M, Takei H, Masuda S,<br />

Nakamura S, Noguchi S. Graduate School of Medicine, Osaka<br />

University, Suita, Osaka, Japan; National Hospital Organization<br />

Osaka National Hospital, Osaka, Japan; Osaka Koseinenkin Hospital,<br />

Osaka, Japan; Osaka Medical Center for <strong>Cancer</strong> & Cardiovascular<br />

Diseases, Osaka, Japan; The <strong>Cancer</strong> Institute of Japan Foundation for<br />

<strong>Cancer</strong> Research, Tokyo, Japan; Kawasaki Medical School, Kurashiki,<br />

Okayama, Japan; Osaka Police Hospital, Osaka, Japan; Saitama <strong>Cancer</strong><br />

Center, Kita-Adachi, Saitama, Japan; Nihon University School of<br />

Medicine, Tokyo, Japan; Showa University School of Medicine,<br />

Tokyo, Japan.<br />

Are the mitotic factors Mitotic Activity Index (MAI) and<br />

Phosphohistone 3 (PPH3) stronger prognostic proliferation<br />

factors than Ki67 in node-negative breast cancer?<br />

Klintman M, Strand C, Gudlaugsson E, Janssen E, Skaland I,<br />

Malmström P, Baak J, Fernö M. Clinical Sciences, Lund University<br />

and Skåne University Hospital, Lund, Sweden; Stavanger University<br />

Hospital, Stavanger, Norway; Stavanger University Hospital and the<br />

Gade Institute, University of Bergen, Stavanger, Norway.<br />

P2-10-13<br />

www.aacrjournals.org<br />

25s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P2-10-20 A tumor DNA complexity index is an independent predictor<br />

of survival in a dataset of 1950 breast cancers; a METABRIC<br />

group study<br />

Vollan HKM, Rueda OM, Børresen-Dale A-L, Aparicio S, Caldas<br />

C. Institute for <strong>Cancer</strong> Research, Oslo University Hospital, Oslo,<br />

Norway; The K.G. Jebsen Center for <strong>Breast</strong> <strong>Cancer</strong> Research,<br />

Institute for Clinical Medicine, University of Oslo, Norway; Oslo<br />

University Hospital, Oslo, Norway; Cambridge Research Institute,<br />

Cambridge, United Kingdom; University of Cambridge, United<br />

Kingdom; University of British Colombia, Vancouver, BC, Canada;<br />

British Colombia <strong>Cancer</strong> Research Center, Vancouver, BC, Canada;<br />

Cambridge Experimental <strong>Cancer</strong> Medicine Centre, Cambridge,<br />

United Kingdom; Addenbrooke’s Hospital, Cambridge University<br />

Hospital, Cambridge, United Kingdom.<br />

P2-10-21 Prognostic value of estrogen receptor status in women with<br />

synchronous or metachronous breast cancers<br />

Huo D. University of Chicago, IL.<br />

P2-10-22 Phosphorylation of Steroid Receptor Coactivator 3 (SRC3)<br />

at Ser543 is a novel independent prognostic marker in<br />

breast cancer<br />

Palmieri C, Gojis O, Rudraraju B, Abdel-Fatah TMA, Moore D, Shaw J,<br />

Green A, Ellis IO, Coombes RC, Ali S. Imperial College London, United<br />

Kingdom; Nottingham University City Hospital, Nottingham, United<br />

Kingdom; University of Leicester, United Kingdom; Nottingham<br />

University Hospitals, City Hospital Campus, Nottingham, United<br />

Kingdom.<br />

P2-10-23 Lymphovascular Invasion (LVI) and Overall Survival in<br />

Node-negative and Node-positive <strong>Breast</strong> <strong>Cancer</strong> Patients:<br />

A Meta-analysis<br />

Sahebjam S, Diaz-Padilla I, Ocana A, Seruga B, Amir E. Princess<br />

Margaret Hospital; Institute of Oncology Ljubljana.<br />

P2-10-24 Independent validation of Recurrence Online using 1,638<br />

breast cancer microarray samples<br />

Györffy B, Weltz B, Benke Z, Timar J, Sztupinszki Z, Schaefer R.<br />

Hungarian Academy of Sciences; Semmelweis University; Charité.<br />

P2-10-25 Prognostic value of relative change in tumor marker CA 27.29<br />

in early stage breast cancer – The SUCCESS trial<br />

Hepp P, Tesch H, Forstbauer H, Rezai M, Beck T, Schrader I, Kleine-<br />

Tebbe A, Hucke J, Finas D, Soeling U, Zahm D-M, Weiss E, Beckmann<br />

MW, Janni W, Rack B. University Düsseldorf; Praxis Prof. Tesch<br />

Frankfurt; Gemeinschaftspraxis Dr. Forstbauer & Dr. Ziske Troisdorf;<br />

Luisenkrankenhaus Düsseldorf; Städtisches Klinikum Rosenheim;<br />

Henriettenstiftung Krankenhaus Hannover; DRK Kliniken Berlin<br />

Köpenick; Bethesda Krankenhaus Wuppertal; Universitätsklinikum<br />

Schleswig-Holstein, Campus Lübeck; Gemeinschaftspraxis Siehl &<br />

Söling; SRH Wald-Klinikum Gera; Klinikum Sindelfingen-Böblingen;<br />

Universitätsfrauenklinik Erlangen; Universitätsfrauenklinik Munich.<br />

P2-10-26 Association between circulating tumor cells and molecular<br />

breast cancer subtypes<br />

Gang N, Haibo W, Funian L, Chen L, Xiaoyi L, Xingang W, Zhidong L.<br />

Affiliated Hospital of Medical College, Qingdao University, Qingdao,<br />

Shandong, China.<br />

P2-10-27 No Discordant Receptor Status Results among 50 Paired<br />

<strong>Breast</strong> and Axillary Metastasis Core Biopsies when Pre-analytic<br />

Variation is Controlled<br />

Miller DV, Rosenthall RE, Avent JM, Carter CC, Hansen J, Hammond<br />

MEH. Intermountain Healthcare, Salt Lake City, UT.<br />

P2-10-28<br />

P2-10-29<br />

P2-10-30<br />

P2-10-31<br />

P2-10-32<br />

P2-10-33<br />

The Prognostic Index, KiGE, Combining Proliferation,<br />

Histological Grade and Estrogen Receptor Status Challenges<br />

Gene Profiling –<br />

A Study in 1,854 Chemo-Naïve Women with N0/N1 Primary<br />

<strong>Breast</strong> <strong>Cancer</strong><br />

Strand C, Bak M, Borgquist S, Chebil G, Falck A-K, Fjällskog M-L,<br />

Grabau D, Hedenfalk I, Jirström K, Klintman M, Malmtröm P, Olsson<br />

H, Rydén L, Stål O, Bendahl P-O, Fernö M. Lund University, Division<br />

of Oncology, Skåne University Hospital, Lund, Sweden; Odense<br />

University Hospital, Odense, Denmark; Mammography, Bergaliden,<br />

Helsingborg, Sweden; Helsingborg Hospital, Helsingborg, Sweden;<br />

Uppsala University Hospital, Uppsala, Sweden; Lund University,<br />

Skåne University Hospital, Lund, Sweden; Lund University, Division of<br />

Pathology, Lund, Sweden; Skåne University Hospital, Lund, Sweden;<br />

Linköping University, County Council of Östergötland, Linköping,<br />

Sweden; Lund University, Division of Surgery, Skåne University<br />

Hospital, Lund, Sweden.<br />

Time dependent breast cancer metastasis prediction using<br />

novel biological imaging, clinico-pathological and genomic<br />

data combined with Bayesian modeling to reduce over-fitting<br />

and improve on inter-cohort reproducibility<br />

Sheeba I, Kelleher M, Lawler K, Festy F, Barber P, Shamill E, Gargi P,<br />

Weitsman G, Barrett J, Fruhwirth G, Huang L, Tullis I, Woodman N,<br />

Pinder S, Ofo E, Fernandes L, Beutler M, Ameer-Beg S, Holmberg L,<br />

Purushotham A, Fraternali F, Condeelis J, Hanby A, Gillett C, Ellis P,<br />

Vojnovic B, Coolen A, Ng T. Kings College London, Guy’s Medical<br />

School Campus, London, England, United Kingdom; King’s College<br />

London, Strand Campus, London, England, United Kingdom; Guy’s<br />

and St Thomas Foundation Trust, London, England, United Kingdom;<br />

Gray Institute for Radiation Oncology & Biology, University of Oxford,<br />

England, United Kingdom; Leeds Institute of Molecular Medicine,<br />

Leeds, England, United Kingdom.<br />

Expression of lipid metabolism genes in contralateral<br />

unaffected breast associated with estrogen receptor status of<br />

breast cancer<br />

Wang J, Scholtens D, Holko M, Ivancic D, Lee O, Hu H, Chatterton RT,<br />

Khan SA. Northwestern university, Chicago, IL.<br />

Correlation of quantitative p95HER2 and total HER2 levels<br />

with clinical outcomes in a combined analysis of two cohorts<br />

of trastuzumab-treated metastatic breast cancer patients<br />

Duchnowska R, Sperinde J, Leitzel K, Szostakiewicz B, Paquet A, Ali<br />

SM, Jankowski T, Haddad M, Fuchs E-M, Arlukowicz-Czartoryska B,<br />

Winslow J, Singer C, Wysocki PJ, Lie Y, Horvat R, Foszczynska-Kloda<br />

M, Petropoulos C, Radecka B, Litwiniuk M, Debska S, Weidler J,<br />

Huang W, Biernat W, Köstler WJ, Jassem J, Lipton A. Military Institute<br />

of Medicine, Warsaw, Poland; Monogram Biosciences/Integrated<br />

Oncology/LabCorp, South <strong>San</strong> Francisco, CA; Penn State/Hershey<br />

Medical Center, Hershey, PA; Medical University of Gdansk, Poland;<br />

Lublin Oncology Center, Lublin, Poland; Medical University of Vienna,<br />

Austria; Bialystok Oncology Center, Bialystok, Poland; Greater Poland<br />

<strong>Cancer</strong> Center, Poznan, Poland; West Pomeranian Oncology Center,<br />

Szczecin, Poland; Opole Oncology Center, Opole, Poland; Poznan<br />

University of Medical Sciences, Poznan, Poland; Regional <strong>Cancer</strong><br />

Center, Lódz, Poland.<br />

Sialyl Lewis X and inflammatory mediators in breast cancer<br />

patients: biological correlations and prognostic value<br />

Lee B-N, Arun BK, Cohen EN, Tin S, Gutierrez-Barrera AM, Miura T,<br />

Kiyokawa I, Alvarez RH, Valero V, Ueno NT, Cristofanilli M, Reuben JM.<br />

University of Texas MD Anderson <strong>Cancer</strong> Center, Houston, TX; Nitto<br />

Boseki Co., Ltd, Tokyo, Japan; Fox Chase <strong>Cancer</strong> Center,<br />

Philadelphia, PA.<br />

Mitotic Component of Grade Can Distinguish <strong>Breast</strong> <strong>Cancer</strong><br />

Patients at Greatest Risk of Local Relapse<br />

Done SJ, Miller NA, Wei Shi W, Pintilie M, McCready DR, Liu F-F,<br />

Fyles A. University Health Network, Toronto, ON, Canada; Princess<br />

Margaret Hospital, University Health Network, Toronto, ON, Canada;<br />

University of Toronto, ON, Canada.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012 26s <strong>Cancer</strong> Research


December 4–8, 2012<br />

Program Schedule<br />

P2-10-34 Development and validation of ClinicoMolecular Triad<br />

Classification (CMTC), a platform for breast cancer (BC)<br />

prognostic and predictive gene signature portfolios<br />

Leong WL, Wang D-Y, Done S, McCready D. University Health<br />

Network, Toronto, ON, Canada.<br />

P2-10-35 The grade of accompanying DCIS in IDC as important<br />

prognostic factor<br />

Kim JY, Moon H-G, Han WS, Noh DY. Seoul National University<br />

Hospital, <strong>Breast</strong> <strong>Cancer</strong> Center, Seoul, Korea.<br />

P2-10-36 Analysis of biomarker expression and biological subtype<br />

in primary tumour, corresponding lymph node and distant<br />

metastasis with 5-year follow-up<br />

Falck A-K, Ferno M, Bendahl P-O, Chebil G, Olsson H, Rydén L. Clinical<br />

Sciences, Lund, Sweden.<br />

P2-10-37 Long-term prognosis of early breast cancer in a populationbased<br />

cohort with a known BRCA1/2 mutation status<br />

Nilsson MP, Werner Hartman L, Idvall I, Kristoffersson U, Olsson H,<br />

Johansson O, Borg Å, Loman N. Clinical Sciences, Lund University,<br />

Sweden; Lund University, Sweden; Landspitali University Hospital,<br />

Reykjavik, Iceland.<br />

P2-10-38 Prognostic factors uPA/PAI-1: measurement in core needle<br />

biopsies<br />

Vetter M, Landstorfer B, Lantzsch T, Buchmann J, Große R, Ruschke<br />

K, Holzhausen H-J, Thomssen C, Kantelhardt E-J. Martin-Luther<br />

University Halle-Wittenberg, Halle/Saale, Germany; St. Elisabeth & St.<br />

Barbara Hospital, Halle/Saale, Germany; Martha-Maria Hospital, Halle/<br />

Saale, Germany.<br />

P2-10-39 Does E-cadherin or N-cadherin or epithelial-mesenchymal<br />

transition have a probability of clinical implication of the<br />

prognostic marker in invasive ductal carcinoma?<br />

Lee J, Yang G, Paik SS, Chung MS. Hanyang University Medical<br />

Center, Seoul, Republic of Korea.<br />

P2-10-40 Correlation between expression of the prognostic marker<br />

Progranulin (GP88) with Oncotype Dx Recurrence Score in<br />

estrogen receptor positive breast tumors<br />

Serrero G, Koka M, Goicochea L, Tkaczuk KR, Fernandez KL, Logan<br />

LS, Tuttle K, Yue B, Ioffe OB. A&G Pharmaceutical Inc, Columbia, MD;<br />

University of Maryland Greenebaum <strong>Cancer</strong> Center, Baltimore, MD;<br />

Franklin Square Hospital, Baltimore, MD; Mercy Hospital,<br />

Baltimore, MD.<br />

P2-10-41 The prediction of invasion in ductal carcinoma in situ:<br />

developing prediction model and validation<br />

Lee SK, Kim M, Nam SJ, Lee JE, Yang J-H. Samsung Medical Center,<br />

Sungkyunkwan University School of Medicine; Konkuk University<br />

Medical Center.<br />

P2-10-42 Gene expression profiling to predict the risk of locoregional<br />

recurrence in breast cancer<br />

Drukker CA, Nijenhuis MV, Elias SG, Wesseling J, Russell NS, de<br />

Snoo F, van ‘t Veer LJ, Beitsch PD, Rutgers EJT. Netherlands<br />

<strong>Cancer</strong> Institute-Antoni van Leeuwenhoek Hospital, Amsterdam,<br />

Netherlands; University Medical Center Utrecht, Utrecht, Netherlands;<br />

Agendia Inc., Amsterdam, Netherlands; University of California <strong>San</strong><br />

Francisco, <strong>San</strong> Francisco; Dallas Surgical Group, Dallas.<br />

P2-10-43 The combination of immunohistochemistry for predicting<br />

TP53 mutation is useful prognostic marker in breast cancer<br />

Watanabe G, Ishida T, Takahasi S, Ishioka C, Watanabe M, Ohuchi N.<br />

Tohoku Univ., Sendai, Miyagi, Japan.<br />

Psychosocial, Quality of Life, and Educational Aspects: Survivorship<br />

Research<br />

P2-11-01 Effects of chemotherapy on the ovary: What you didn’t know<br />

Barton DL, Thompson SL, Senn-Reeves JN, Satele DV, Frost M. Mayo<br />

Clinic, Rochester, MN.<br />

P2-11-02<br />

P2-11-03<br />

P2-11-04<br />

P2-11-05<br />

P2-11-06<br />

P2-11-07<br />

P2-11-08<br />

P2-11-09<br />

P2-11-10<br />

P2-11-11<br />

P2-11-12<br />

Perception and practice of reproductive specialists towards<br />

fertility preservation of young breast cancer patients<br />

Shimizu C, Kato T, Tamura N, Asada Y, Hiroko B, Mizota Y, Yamamoto<br />

S, Fujiwara Y. National <strong>Cancer</strong> Center Hospital, Tokyo, Japan; Asada<br />

Lady’s Clinic, Nagoya, Aichi, Japan; University of Tsukuba, Ibaragi,<br />

Japan; National <strong>Cancer</strong> Center, Tokyo, Japan.<br />

Randomized, single blind trial comparing limited and<br />

intensive survivorship interventions following adjuvant<br />

therapy in a multiethnic cohort of breast cancer survivors<br />

Hershman DL, Greenlee H, Awad D, Kalinsky K, Maurer M, Kranwinkel<br />

G, Brooks-Brafman L, Fuentes D, Tsai W-Y, Crew KD. Columbia<br />

Univeristy Medical Center, New York, NY; Mailman School of Public<br />

Health, New York, NY.<br />

Chemotherapy Induces Neuroinflammation and Cognitive<br />

Deficits in Female Mice<br />

Jarrett BL, Lustberg MB, Hinzey A, Stuller KA, Shapiro CL, DeVries C.<br />

The Ohio State University Wexner Medical Center, Columbus, OH;<br />

The Ohio State University Comprehensive <strong>Cancer</strong> Center,<br />

Columbus, OH.<br />

Work status and financial stability during treatment for early<br />

breast cancer: a pilot study of an ethnically diverse sample<br />

Eberle CE, Min S-H, Jung S, Ramirez J, Patil S, Gany F, Blinder VS.<br />

Memorial Sloan-Kettering <strong>Cancer</strong> Center, New York, NY.<br />

Mortality among offspring of women diagnosed with breast<br />

cancer; a population based study<br />

Verkooijen HM, Ang X, Liu J, Czene K, Salim A, Hartman M.<br />

University Medical Center Utrecht, Netherlands; National University<br />

of Singapore, Singapore; Karolinska Institute, Stockholm, Sweden;<br />

National University Hospital, Singapore, Singapore.<br />

Endothelial progenitor cells as novel markers of anthracycline<br />

induced cardiac injury<br />

Lustberg MB, Ruppert AS, Carothers S, Bingman A, McCarthy B,<br />

Raman S, Das M, Kanji S, Lu J, Das H, Cinar-Akakin H, Gurcan MN,<br />

Berger MJ, Wesolowski R, Olson EM, Ramaswamy B, Mrozek E,<br />

Layman RM, Binkley P, Shapiro CL. The OSU <strong>Breast</strong> Program at<br />

Stefanie Spielman Comprehensive <strong>Breast</strong> Center; OSU.<br />

Feasibility of Enrollment into a Survivorship Care Plan Study at<br />

Initial Diagnosis<br />

Bulloch KJ, Irwin M, Chagpar A, <strong>San</strong>ft T. Yale University,<br />

New Haven, CT.<br />

Weight Change and Risk of Incident Diabetes after<br />

<strong>Breast</strong> <strong>Cancer</strong><br />

Erickson KD, Patterson RE, Natarajan L, Lindsay SP, Heath D, Caan BJ.<br />

Moores UC <strong>San</strong> Diego <strong>Cancer</strong> Center, University of California, <strong>San</strong><br />

Diego, La Jolla, CA; <strong>San</strong> Diego State University, <strong>San</strong> Diego, CA; Kaiser<br />

Permanente Northern California, Oakland, CA.<br />

Prospective memory impairment in early breast cancer<br />

survivors: Finally homing in on the real deficit?<br />

Verma S, Collins B, Song X, Bedard M, Paquet L. The Ottawa Hospital<br />

Regional <strong>Cancer</strong> Centre; The Ottawa Hospital; Carleton University.<br />

Patient Reported Outcomes after <strong>Breast</strong> <strong>Cancer</strong> Surgery and<br />

Adjuvant Therapy from a German <strong>Breast</strong> <strong>Cancer</strong> Centre<br />

Feiten S, Dünnebacke J, Heymanns J, Köppler H, Thomalla J, van<br />

Roye C, Wey D, Weide R. Institute for Health Care Research in<br />

Oncology, Koblenz, Germany; Catholic Clinical Centre Koblenz-<br />

Montabaur, Koblenz, Germany; Oncology Group Practice Koblenz,<br />

Germany.<br />

Proactive approach to risk reduction and early detection of<br />

breast cancer related lymphedema<br />

Fu MR, Guth A, Kleinman R, Cartwright F, Haber J, Axelrod D. New<br />

York University, New York, NY; NYU <strong>Cancer</strong> Center, New York, NY.<br />

www.aacrjournals.org<br />

27s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P2-11-13 The Effect of Positive Axillary Lymph Nodes on Symptoms,<br />

Physical Impairments, and Function<br />

Kesarwala AH, Pfalzer LA, O’Meara WP, Stout NL. National <strong>Cancer</strong><br />

Institute, Bethesda, MD; University of Michigan - Flint, MI; Lahey<br />

Clinic, Burlington, MA; Walter Reed National Military Medical Center,<br />

Bethesda, MD.<br />

P2-11-14 Symptoms, Physical Impairments, and Function in <strong>Breast</strong><br />

<strong>Cancer</strong> Patients with Negative Axillary Lymph Nodes<br />

Kesarwala AH, Pfalzer LA, O’Meara WP, Stout NL. National <strong>Cancer</strong><br />

Institute, Bethesda, MD; University of Michigan - Flint, MI; Lahey<br />

Clinic, Burlington, MA; Walter Reed National Military Medical Center,<br />

Bethesda, MD.<br />

P2-11-15 Development of a web-based survey tool to assess change<br />

in breast cancer (BrCa) survivor knowledge after receipt of<br />

cancer treatment summary and survivorship care plan (SCP)<br />

Custer JL, Rocque GB, Wisinski KB, Jones NR, Donohue S, Koehn TM,<br />

Champeny TL, Terhaar AR, Chen KB, Peck KA, Tun MT, Wiegmann<br />

DA, Sesto ME, Tevaarwerk AJ. University of Wisconsin, Madison, WI.<br />

P2-11-16 Cardiac Morbidity After Adjuvant Chemotherapy (CT) for Early<br />

<strong>Breast</strong> <strong>Cancer</strong> in the Community Setting<br />

Patt D, Espirito J, Turnwald B, Denduluri N, Wang Y, Lina A,<br />

Hoverman R, Neubauer M, Bosserman L, Busby L, Brooks B,<br />

Cartwright T, Sitarik M, Schnadig I, Winter W, Garey J, Ginsburg-<br />

Arlen A, Bergstrom K, Beveridge R. Pathways Task Force, US<br />

Oncology Network, McKesson Specialty Health, Austin, TX;<br />

Pathways Task Force, US Oncology Network, McKesson Specialty<br />

Health, The Woodlands, TX; Pathways Task Force, US Oncology<br />

Network, McKesson Specialty Health, Arlington, VA; US Oncology<br />

Network, McKesson Specialty Health, The Woodlands, TX; Kansas<br />

City <strong>Cancer</strong> Center, Pathways Task Force, US Oncology Network,<br />

McKesson Specialty Health, Overland Park, KS; Pathways Task<br />

Force, US Oncology Network, McKesson Specialty Health, Rancho<br />

Cucamonga, CA; Rocky Mountain <strong>Cancer</strong> Centers, Pathways Task<br />

Force, US Oncology Network, McKesson Specialty Health, Boulder,<br />

CO; Pathways Task Force, US Oncology Network, McKesson Specialty<br />

Health, Dallas, TX; Pathways Task Force, US Oncology Network,<br />

McKesson Specialty Health, Ocala, FL; Pathways Task Force, US<br />

Oncology Network, McKesson Specialty Health, Tualatin, OR;<br />

Pathways Task Force, US Oncology Network, McKesson Specialty<br />

Health, Portland, OR.<br />

P2-11-17 Pilot Study to Evaluate a Home-based Exercise and Weight<br />

Loss Intervention on Cardiopulmonary Fitness and Markers of<br />

<strong>Breast</strong> <strong>Cancer</strong> Risk in Postmenopausal <strong>Breast</strong> <strong>Cancer</strong> Survivors<br />

Burnett D, Klemp JR, Porter C, Schmitz KJ, Fabian CJ, Kluding P.<br />

University of Kansas Medical Center, Kansas City, KS; University<br />

of Kansas Hospital, Kansas City, KS; University of Pennsylvania,<br />

Philadelphia, PA.<br />

Psychosocial, Quality of Life, and Educational Aspects: Quality of Life -<br />

Supportive Care<br />

P2-12-01 Prospective Evaluation of Joint Symptoms in Postmenopausal<br />

Women Initiating Aromatase Inhibitors for Early Stage<br />

<strong>Breast</strong> <strong>Cancer</strong><br />

Crew KD, Chehayeb Makarem D, Awad D, Kalinsky K, Maurer M,<br />

Kranwinkel G, Brafman L, Fuentes D, Hershman DL. Columbia<br />

University Medical Center, New York, NY.<br />

P2-12-02 Withdrawn<br />

P2-12-03 A pilot study evaluating the benefits and feasibility of an<br />

exercise program for breast cancer patients receiving adjuvant<br />

chemotherapy<br />

Petrella TM, Laredo S, Oh P, Marzolini S, Warner E, Dent R, Verma S,<br />

Eisen A, Pritchard K, Trudeau M, Zhang L, Bjarnason G. Odette <strong>Cancer</strong><br />

Centre, Toronto, ON, Canada; Women’s College Hospital, Toronto,<br />

ON, Canada; Toronto Rehabilitation Institute, Toronto, ON, Canada;<br />

Macrostat Inc, Toronto, ON, Canada.<br />

P2-12-04 Music Therapy Reduces Radiotherapy-Induced<br />

Fatigue in Patients with <strong>Breast</strong> or Gynecological <strong>Cancer</strong>:<br />

A Randomized Trial<br />

Freitas NMA, Silva TRMA, Freitas-Junior R, Paula Junior W, Silva DJ,<br />

Machado GDP, Ribeiro MKA, Carneiro JP. Araujo Jorge Hospital/<br />

ACCG, Goiania, Goias, Brazil; Federal University of Goias, Goiania,<br />

Goias, Brazil; Instituto Integrado de Neurociencias/IINEURO, Goiania,<br />

Goias, Brazil.<br />

P2-12-05 Limited Absorption of Low Dose 10 µg Intravaginal 17-β<br />

Estradiol (Vagifem®) in Postmenopausal Women with <strong>Breast</strong><br />

<strong>Cancer</strong> on Aromatase Inhibitors<br />

Goldfarb SB, Dickler M, Dnistrian A, Patil S, Dunn L, Chang K,<br />

Berkowitz A, Tucker N, Carter J, Barakat R, Hudis C, Castiel M.<br />

Memorial Sloan-Kettering <strong>Cancer</strong> Center, New York, NY.<br />

P2-12-06 Ultra-low dose vaginal estriol and Lactobacillus acidophilus<br />

(Gynoflor®) in early breast cancer survivors on aromatase<br />

inhibitors: Pharmacokinetic, efficacy and safety results from<br />

a phase I study<br />

Neven P, Donders G, Mögele M, Lintermans A, Bellen G, Ortmann<br />

O, Buchholz S. University Hospital Gasthuisberg Leuven, Belgium;<br />

Femicare vzw, Clinical Research for Women, Tienen, Belgium;<br />

Universitätsklinikum Regensburg, Germany.<br />

P2-12-07 A review of clinical endpoints and use of quality-of-life<br />

outcomes in phase III metastatic breast cancer clinical trials<br />

Tatla R, Landaverde D, Victor C, Miles D, Verma S. University of<br />

Toronto, ON, Canada; Sunnybrook Odette <strong>Cancer</strong> Center, Toronto,<br />

ON, Canada; Dalla Lana School of Public Health, University of<br />

Toronto, ON, Canada; Mount Vernon <strong>Cancer</strong> Centre, United<br />

Kingdom.<br />

P2-12-08 Sorafenib for treatment of breast-cancer related lymphedema<br />

Zambetti M, Guidetti A, Carlo-Stella C, De Benedictis E, Tessari A,<br />

Balzarini A, Caraceni A, Gianni L, Gianni AM. IRCCS Ospedale <strong>San</strong><br />

Raffaele, Milan, Italy; Fondazione IRCCS Istituto Nazionale Tumori,<br />

Milan, Italy; Humanitas <strong>Cancer</strong> Center, IRCCS Istituto Clinico<br />

Humanitas, Rozzano, Italy.<br />

P2-12-09 A randomized controlled trial of support group intervention<br />

after breast cancer treatment: Results on sick leave, health<br />

care utilization and health economy<br />

Granstam Björneklett H, Rosenblad A, Lindemalm C, Ojutkangas<br />

M-L, Letocha H, Strang P, Bergkvist L. Centre for Clinical Research,<br />

Västerås, Västmanland, Sweden; <strong>Cancer</strong> Center Karolinska, Karolinska,<br />

Stockhoöm, Sweden; Karolinska Institutet, Stockholm, Sweden.<br />

P2-12-10 Psycho-spiritual therapy for improving the quality of life and<br />

spiritual well-being of women with breast cancer<br />

Loghmani A, Jafari N, Zamani A, Farajzadegan Z, Bahrami F, Emami<br />

H. Isfahan University of Medical Sciences, Isfahan, Islamic Republic of<br />

Iran; University of Isfahan, Islamic Republic of Iran.<br />

P2-12-11 Use of the DigniCap System To Prevent Hair Loss in Women<br />

Receiving Chemotherapy (CTX) for Stage I <strong>Breast</strong> <strong>Cancer</strong> (BC)<br />

Rugo HS, Serrurier KM, Melisko M, Glencer A, Hwang J, D’Agostino,<br />

Jr. R, Hutchens S, Esserman LJ, Melin S. University of California <strong>San</strong><br />

Francisco Helen Diller Comprehensive <strong>Cancer</strong> Center, <strong>San</strong> Francisco,<br />

CA; Wake Forest Baptist Health Medical Center, Winston-Salem, NC.<br />

P2-12-12 Efficacy and safety of scalp cooling (SC) treatment for alopecia<br />

prevention in women receiving chemotherapy (CTX) for breast<br />

cancer (BC)<br />

Serrurier KM, Melisko ME, Glencer A, Esserman LJ, Rugo HS. UCSF<br />

Helen Diller Comprehensive <strong>Cancer</strong> Center.<br />

P2-12-13 Results of randomised controlled phase II study (KBCSG02<br />

trial) of the efficiency of palonosetron, aprepitant, and<br />

dexamethasone for day 1 with or without dexamethasone on<br />

days 2 and 3<br />

Kosaka Y, Sengoku N, Kikuchi M, Nishimiya H, Enomoto T, Kuranami<br />

M, Watanabe M. Kitasato University School of Medicine, Sagamihara,<br />

Japan.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012 28s <strong>Cancer</strong> Research


December 4–8, 2012<br />

Program Schedule<br />

P2-12-14 Use of the MD Anderson Symptom Inventory To Screen for<br />

Depression in <strong>Breast</strong> <strong>Cancer</strong><br />

Kvale EA, Azuero CB, Azuero A, Fisch M, Ritchie C. Birmingham<br />

VA Medical Center, Birmingham, AL; University of Alabama at<br />

Birmingham, AL; University of Alabama, Tuscaloosa, AL; MD<br />

Anderson <strong>Cancer</strong> Center, Houston, TX; University of California, <strong>San</strong><br />

Francisco, CA.<br />

P2-12-15 Understanding the complex non face-to-face interventions<br />

delivered by the clinical nurse specialists in metastatic<br />

breast cancer<br />

Warren M, Mackie D, Leary A. Royal Marsden NHS Foundation Trust,<br />

Sutton, Surrey, United Kingdom; Royal Marsden NHS Foundation<br />

Trust, London, United Kingdom; Independent Healthcare Consultant<br />

and Research Analyst.<br />

Treatment: Endocrine Therapy - Adjuvant<br />

P2-13-01 Impact of Body Mass Index (BMI) on the efficacy of<br />

aromatase inhibitors to suppress estradiol serum levels<br />

in postmenopausal patients with early breast cancer: a<br />

prospective proof of principle<br />

Pfeiler G, Königsberg R, Hadji P, Fitzal F, Maroske M, Ban G, Zellinger<br />

J, Exner R, Seifert M, Singer C, Gnant M, Dubsky P. Medical University<br />

of Vienna, Austria; Applied <strong>Cancer</strong> Research – Institution for<br />

Translational Research Vienna (ACR-ITR VIEnna)/CEADDP, Austria;<br />

Universityhospital of Giessen and Marburg GmbH, Germany.<br />

P2-13-02 Effect of aspirin (ASP) or celecoxib (CC) use on outcomes<br />

in postmenopausal breast cancer patients randomized to<br />

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

Higgins MJ, Chapman J-AW, Ingle JN, Sledge G, Budd GT, Ellis MJ,<br />

Pritchard KI, Clemons M, Badovinac Crnjevic T, Han L, Gelmon K,<br />

Rabaglio M, Elliott C, Shepherd LE, Goss PE. Massachusetts General<br />

Hospital, Boston, MA; NCIC Clinical Trials Group, Queen’s University,<br />

Kingston, ON, Canada; Mayo Clinic, Rochester, MN; Ottawa Hospital<br />

and Faculty of Medicine, University of Ottawa, ON, Canada;<br />

Vancouver Centre, BCCA, Vancouver, BC, Canada; Sunnybrook Odette<br />

<strong>Cancer</strong> Centre, Toronto, ON, Cayman Islands; Indiana University,<br />

Indianapolis, IN; Cleveland Clinic, Cleveland, OH; Washington<br />

University in St. Louis, MO; University Hospital Berne, Switzerland.<br />

P2-13-03 Prevalence of non-metastatic breast cancer patients treated<br />

with aromatase inhibitors in the United States<br />

Liede A, Hernandez RK, Pirolli M, Quigley J, Quach D. Amgen Inc.,<br />

Thousand Oaks, CA; IMS Health, Plymouth Meeting, PA.<br />

P2-13-04 Superior efficacy of anastrozole to tamoxifen as adjuvant<br />

therapy for postmenopausal patients with hormoneresponsive<br />

breast cancer. Efficacy results of long-term<br />

follow-up data from N-SAS BC 03 trial<br />

Imoto S, Osumi S, Aogi K, Hozumi Y, Mukai H, Iwata H, Yokota I,<br />

Yamaguchi T, Ohashi Y, Watanabe T, Takatsuka Y, Aihara T. School<br />

of Medicine, Kyorin University, Tokyo, Japan; National Hospital<br />

Organization Shikoku <strong>Cancer</strong> Center, Ehime, Japan; Jichi Medical<br />

University, Tochigi, Japan; National <strong>Cancer</strong> Center Hospital East,<br />

Chiba, Japan; Aichi <strong>Cancer</strong> Center Hospital, Aichi, Japan; School of<br />

Public Health, University of Tokyo, Tokyo, Japan; Tohoku University<br />

School of Medicine, Miyagi, Japan; Hamamatsu Oncology Center,<br />

Shizuoka, Japan; Kansai Rosai Hospital, Hyogo, Japan; Aihara<br />

Hospital, Osaka, Japan.<br />

P2-13-05 A pilot prospective study of adherence to aromatase inhibitor<br />

adjuvant therapy in patients with stage 1-3 breast carcinoma<br />

Heiss B, Thompson J, Nightingale G, Tait N, Kesmodel S, Bellavance<br />

E, Chumsri S, Bao T, Goloubeva O, Feigenberg S, Tkaczuk K. Marlene<br />

and Stewart Greenebaum <strong>Cancer</strong> Center, University of Maryland,<br />

Baltimore, MD; Thomas Jefferson University, Philadelphia, PA.<br />

P2-13-06 Effect of letrozole on bone and joints in collagen-induced<br />

arthritis in mice<br />

Lintermans A, Verhaeghe J, Van Bree R, Vanderschueren D,<br />

Vanderhaegen J, Braem K, Lories R, Neven P. University Hospitals<br />

Leuven, KU Leuven, Belgium; KU Leuven, Leuven, Belgium; University<br />

Hospitals Leuven, Belgium.<br />

P2-13-07 Long-term follow-up data of the side effect profile of<br />

anastrozole compared with tamoxifen in Japanese women:<br />

findings from N-SAS BC03 trial<br />

Iwata H, Ohsumi S, Aogi K, Hozumi Y, Imoto S, Mukai H, Yokota I,<br />

Yamaguchi T, Ohashi Y, Watanabe T, Takatsuka Y, Aihara T. Aichi<br />

<strong>Cancer</strong> Center Hospital, Nagoya, Aichi, Japan; NHO Shikoku <strong>Cancer</strong><br />

Center, Matsuyama, Ehime, Japan; Jichi Medical University, Tochigi,<br />

Japan; School of Medicine, Kyorin University, Tokyo, Japan; National<br />

<strong>Cancer</strong> Center Hospital East, Chiba, Japan; School of Public Health,<br />

University of Tokyo, Tokyo, Japan; Tohoku University School of<br />

Medicine, Sendai, Japan; Hamamatsu Oncology Center, Hamamatsu,<br />

Japan; Kansai Rosai Hospital, Hyogo, Japan; Aihara Hospital,<br />

Osaka, Japan.<br />

P2-13-08 Comparison of Compliance to Anti Estrogen Therapy in<br />

Patients with Early <strong>Breast</strong> <strong>Cancer</strong> followed at Tertiary Centers<br />

versus Through Family Physicians and Primary Surgeons:<br />

A Practice Review<br />

Alkhayyat SS, Younus J, Mirza FN, Stitt L. The Scarborough Hospital,<br />

Scarborough, ON, Canada; The University of Western Ontario,<br />

London, ON, Canada; Harvard University, Cambridge, MA.<br />

P2-13-09 Pharmacological impact of endoxifen in a laboratory<br />

simulation of tamoxifen therapy in postmenopausal<br />

breast cancer patients<br />

Maximov PY, McDaniel RE, Bhatta P, Brauch H, Jordan VC. Lombardi<br />

Comprehensive <strong>Cancer</strong> Center, Georgetown University Medical<br />

Center, Washington, DC; Dr. Margarete Fischer-Bosch-Institute of<br />

Clinical Pharmacology, Stuttgart, Germany.<br />

P2-13-10 Prospective randomized and multicentric evaluation<br />

of cognition in menopausal breast cancer patients<br />

receiving adjuvant hormonotherapy: a phase III study<br />

(Preliminary results)<br />

Vanlemmens L, Delbeuck X, Servent V, Mailliez A, Vanlemmens L,<br />

Lefeuvre-Plesse C, Kerbrat P, Petit T, Fournier C, Vendel Y, Clisant S,<br />

Bonneterre J, Pasquier F, Le Rhun E. Centre Mémoire de Ressource<br />

et de Recherche - CHRU Lille, Lille, France; Centre Oscar Lambret,<br />

Université Lille Nord de France, Lille, France; Centre Eugène Marquis,<br />

Rennes, France; Centre Paul Strauss, Strasbourg, France; Centre Oscar<br />

Lambret, Lille, France; CHRU, Lille, France.<br />

Treatment: Endocrine Therapy - Advanced Disease<br />

P2-14-01 Fulvestrant vs exemestane for treatment of metastatic breast<br />

cancer in patients with acquired resistance to non-steroidal<br />

aromatase inhibitors – a meta-analysis of EFECT and SoFEA<br />

(CRUKE/03/021 & CRUK/09/007)<br />

Johnston SRD, Chia S, Kilburn LS, Gradishar WJ, Cameron D, Dodwell<br />

D, Ellis P, Howell A, Im Y-H, Coombes G, Piccart M, Dowsett M,<br />

Bliss J, On behalf of the SoFEA and EFECT Investigators. The Royal<br />

Marsden Hospital NHS Foundation Trust & The Institute of <strong>Cancer</strong><br />

Research, London, United Kingdom; British Columbia <strong>Cancer</strong><br />

Agency, University of British Columbia, Vancouver, BC, Canada;<br />

The Institute of <strong>Cancer</strong> Research, Sutton, Surrey, United Kingdom;<br />

Robert H. Lurie Comprehensive <strong>Cancer</strong> Center, Feinberg School of<br />

Medicine, Northwestern University, Chicago, IL; Christie Hospital NHS<br />

Trust, Manchester, United Kingdom; Edinburgh <strong>Cancer</strong> Research<br />

Centre, University of Edinburgh and NHS Lothian, Edinburgh,<br />

United Kingdom; Leeds Teaching Hospitals NHS Trust, St. James’s<br />

University Hospital, Leeds, United Kingdom; Guy’s and St Thomas’s<br />

NHS Foundation Trust, London, United Kingdom; Samsung Medical<br />

Center, Seoul, Korea; Jules Bordet Institute, Brussels, Belgium; The<br />

Royal Marsden NHS Foundation Trust, London, United Kingdom.<br />

www.aacrjournals.org<br />

29s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P2-14-02 Preclinical Evaluation of Enzalutamide in <strong>Breast</strong> <strong>Cancer</strong> Models<br />

Cochrane DR, Bernales S, Jacobsen BM, D’Amato NC, Guerrero J,<br />

Gómez F, Protter AA, Elias AD, Richer JK. University of Colorado<br />

Anschutz Medical Campus, Aurora, CO; Medivation Inc., <strong>San</strong><br />

Francisco, CA; Fundación Ciencia & Vida, <strong>San</strong>tiago, Chile.<br />

P2-14-03 “Ethinylestradiol” is beneficial for postmenopausal<br />

advanced breast cancer patients heavily pre-treated with<br />

endocrine agents<br />

Iwase H, Yamamoto Y, Murakami K-I, Yamamoto-Ibusuki M, Tomita S,<br />

Omoto Y. Kumamoto University, Kumamoto, Japan.<br />

P2-14-04 A Phase Ib Dose Escalation Trial of RO4929097 (A γ-secretase<br />

inhibitor) in Combination with Exemestane in Patients with<br />

ER + Metastatic <strong>Breast</strong> <strong>Cancer</strong><br />

Means-Powell JA, Minton SE, Mayer IA, Abramson VG, Ismail-Khan<br />

R, Arteaga CL, Ayers DA, <strong>San</strong>ders MS, Lush RM, Miele L. Vanderbilt-<br />

Ingram <strong>Cancer</strong> Center, Nashville, TN; Moffit <strong>Cancer</strong> Center, Tampa,<br />

FL; University of Mississippi Health Care <strong>Cancer</strong> Institute, Jackson, MS.<br />

P2-14-05 A phase II study of combined fulvestrant and RAD001<br />

(everolimus) in metastatic estrogen receptor (ER) positive<br />

breast cancer after aromatase inhibitor (AI) failure<br />

Croley JJ, Black EP, Romond E, Chambers M, Waynick S, Slone<br />

S, Waynick C, Stevens M, Weiss HL, Massarweh SA. University of<br />

Kentucky and Markey <strong>Cancer</strong> Center, Lexington, KY.<br />

P2-14-06 A phase II trial of low dose estradiol in postmenopausal<br />

women with advanced breast cancer and acquired resistance<br />

to an aromatase inhibitor<br />

Howell SJ, Seif MW, Armstrong AC, Cope J, Wilson G, Welch RS,<br />

Misra V, Ryder D, Blowers E, Palmieri C, Wardley AM. University of<br />

Manchester, United Kingdom; The Christie NHS Foundation Trust,<br />

Manchester, United Kingdom; Imperial College, London, United<br />

Kingdom.<br />

P2-14-07 Evaluation of Aromatase Inhibitor failure and total healthcare<br />

expenditures among post-menopausal women with<br />

metastatic ER+/HER2- breast cancer in the US<br />

Namjoshi M, Landsman-Blumberg P, Thomson E, Chu BC. Novartis<br />

Pharmaceuticals Corporation, East Hanover, NJ; Thomson Reuters,<br />

Washington, DC.<br />

P2-14-08 The Pilot Trial of Intermittent Exemestane Therapy for<br />

Metastatic <strong>Breast</strong> <strong>Cancer</strong><br />

Luu T, Frankel PH, Somlo G, Wong C, Leong L, Mortimer JE, Hurria<br />

A, Koczywas M, Chao J, Chow W, Chung C, Chen S. City of Hope,<br />

Duarte, CA; Angeles Clinic & Research Institute, <strong>San</strong>ta Monica, CA.<br />

Treatment: Patient Resources<br />

P2-15-01 An efficient resource to accelerate research into the cause and<br />

prevention of breast cancer: the Love/Avon Army of Women<br />

Love SM. Dr. Susan Love Research Foundation, <strong>San</strong>ta Monica, CA.<br />

P2-15-02 The Efficacy of Recruitment and Retention Strategies for<br />

Research Subjects in an Early Phase Investigator-Initiated<br />

<strong>Breast</strong> <strong>Cancer</strong> Trial<br />

Reichow J, Higgins D, Parker S, Childs J, Disis ML, Salazar LG.<br />

University of Washington, Seattle, WA.<br />

Treatment: Tissue and Data Banks<br />

P2-16-01 Prospective <strong>Breast</strong> <strong>Cancer</strong> Quality Evaluation Application<br />

Developed for Native Android Tablet and Web Browser for<br />

Improving Mutltidisciplinary <strong>Breast</strong> <strong>Cancer</strong> Care<br />

Grobmyer SR, Raum N, Sposato V. University of Florida, Gainesville,<br />

FL; University of Florida, Gaineville, FL.<br />

P2-16-02 A web-based data management system for a multicenter<br />

international breast cancer oriented blood, tissue, and data<br />

biobank<br />

Margossian AL, Saadjian H, Mira A, Contreras A, Rimawi MF,<br />

Margossian ML, Scheurer M, Osborne K, Gutierrez C. <strong>Breast</strong> Center,<br />

Buenos Aires, Argentina; Dan Duncan <strong>Cancer</strong> Center/Smith <strong>Breast</strong><br />

Center, Baylor College of Medicine, Houston, TX.<br />

P2-16-03 Creation of a ‘state-of-the-art’ breast cancer data and biobank<br />

in Argentina<br />

Margossian AL, Gutierrez C, Saadjian H, May M, Ohanessian R,<br />

Bacigalupo S, Baravalle S, Margossian J, Osborne KC, Scheurer ME.<br />

<strong>Breast</strong> Center, Buenos Aires, Argentina; Dan Duncan <strong>Cancer</strong> Center/<br />

Smith <strong>Breast</strong> Center, Baylor College of Medicine, Houston, TX.<br />

P2-16-04 Attitudes of metastatic breast cancer patients towards<br />

research biopsies<br />

Seah DS, Scott SM, Najita J, Openshaw T, Krag KJ, Frank E, Sohl J,<br />

Stadler ZK, Garrett M, Winer EP, Come S, Lin NU. Dana-Farber <strong>Cancer</strong><br />

Institute, Boston, MA; Beth Isreal Deaconness Medical Center, Boston,<br />

MA; <strong>Cancer</strong> Care of Maine, Brewer, ME; Mass General North Shore<br />

<strong>Cancer</strong> Center, Danvers, MA; Memorial Sloan-Kettering <strong>Cancer</strong><br />

Center, New York, NY.<br />

9:00 am–9:30 am<br />

PLENARY LECTURE 2<br />

Exhibit Hall D<br />

Estrogen Receptor Cistrome: Implications for<br />

<strong>Breast</strong> <strong>Cancer</strong><br />

Jason S. Carroll, PhD<br />

<strong>Cancer</strong> Research UK, University of Cambridge<br />

Cambridge, UNITED KINGDOM<br />

9:30 am–11:30 am<br />

GENERAL SESSION 3<br />

Exhibit Hall D<br />

Moderator: <strong>San</strong>dra M. Swain, MD<br />

Washington Hospital Center<br />

Washington, DC<br />

9:30 S3-1. Neoadjuvant chemotherapy in the very young 35 years of<br />

age or younger<br />

Loibl S, Jackisch C, Gade S, Untch M, Paepke S, Kuemmel S, Schneeweiss<br />

A, Jackisch C, Huober J, Hilfrich J, Hanusch C, Gerber B, Eidtmann<br />

H, Denkert C, Costa S-D, Blohmer J-U, Nekljudova V, Mehta K, von<br />

Minckwitz G. German <strong>Breast</strong> Group, Neu-Isenburg; Klinikum Offenbach;<br />

Helios Kliniken Berlin; University Muenchen; Kliniken Essen Mitte;<br />

University Heidelberg; Uni Duesseldorf; Eilenriedeklinik Duesseldorf;<br />

Rotkreuzklinikum Muenchen; University Rostock; University Kiel; Charite<br />

Berlin; University Magdeburg; <strong>San</strong>kt Gertrauden Berlin.<br />

9:45 S3-2. Chemotherapy prolongs survival for isolated local<br />

or regional recurrence of breast cancer: The CALOR trial<br />

(Chemotherapy as Adjuvant for Locally Recurrent breast cancer;<br />

IBCSG 27-02, NSABP B-37, BIG 1-02)<br />

Aebi S, Gelber S, Lang I, Anderson SJ, Robidoux A, Martin M, Nortier JWR,<br />

Mamounas EP, Geyer Jr. CE, Maibach R, Gelber RD, Wolmark N, Wapnir<br />

I. International <strong>Breast</strong> <strong>Cancer</strong> Study Group, Bern, Switzerland; National<br />

Surgical Adjuvant <strong>Breast</strong> and Bowel Project, Pittsburgh, PA; <strong>Breast</strong><br />

International Group, Brussels, Belgium.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012 30s <strong>Cancer</strong> Research


December 4–8, 2012<br />

Program Schedule<br />

10:00 S3-3. The UK TACT2 Trial: comparison of standard vs accelerated<br />

epirubicin in patients requiring chemotherapy for early breast<br />

cancer (EBC) (CRUK/05/019)<br />

Cameron D, Barrett-Lee P, Canney P, Banerji J, Bartlett J, Bloomfield<br />

D, Bowden S, Brunt M, Earl H, Ellis P, Fletcher M, Morden JP, Robinson<br />

A, Sergenson N, Stein R, Velikova G, Verrill M, Wardley A, Coleman R,<br />

Bliss JM. Edinburgh <strong>Cancer</strong> Research Centre, University of Edinburgh,<br />

United Kingdom; Velindre NHS Trust <strong>Cancer</strong> Centre, Cardiff, United<br />

Kingdom; Beatson West of Scotland <strong>Cancer</strong> Centre, Glasgow, United<br />

Kingdom; The Institute of <strong>Cancer</strong> Research, Sutton, United Kingdom;<br />

Ontario Institute for <strong>Cancer</strong> Research, Toronto, Canada; Brighton &<br />

Sussex University Hospitals, Brighton, United Kingdom; University of<br />

Birmingham, United Kingdom; University Hospital of North Staffordshire,<br />

Stoke-on-Trent, United Kingdom; University of Cambridge and NIHR<br />

Cambridge Biomedical Research Centre, Cambridge, United Kingdom;<br />

Guys Hospital & King College, London, United Kingdom; Leeds Institute<br />

of Molecular Medicine, Leeds, United Kingdom; Southend University<br />

Hospital, Southend, United Kingdom; Information Services Division NHS<br />

National Services Scotland, Edinburgh, United Kingdom; UCL Hospitals,<br />

London, United Kingdom; Northern Centre For <strong>Cancer</strong> Care, Newcastle<br />

upon Tyne, United Kingdom; The Christie Hospital, Manchester, United<br />

Kingdom; Weston Park Hospital, Sheffield, United Kingdom.<br />

10:15 S3-4. Ten year follow-up analysis of intense dose-dense adjuvant<br />

ETC (epirubicin (E), paclitaxel (T) and cyclophosphamide<br />

(C)) confirms superior DFS and OS benefit in comparison to<br />

conventional dosed chemotherapy in high-risk breast cancer<br />

patients with ≥ 4 positive lymph nodes<br />

Moebus V, Schneeweiss A, du Bois A, Lueck H-J, Eustermann H,<br />

Kuhn W, Kurbacher C, Nitz U, Kreienberg R, Jackisch C, Huober J,<br />

Thomssen C, Untch M. Klinikum Frankfurt Höchst, Frankfurt, Germany;<br />

University of Heidelberg, Germany; Klinikum Essen Mitte, Essen,<br />

Germany; Gynäkologisch-Onkologische Praxis, Hannover, Germany;<br />

WiSP Research Institute, Langenfeld, Germany; University of Bonn,<br />

Germany; Medizinisches Zentrum Bonn-Friedensplatz, Bonn, Germany;<br />

Ev. Krankenhaus Bethesda, Mönchengladbach, Germany; University of<br />

Ulm, Germany; Klinikum Offenbach, Offenbach, Germany; University<br />

of Duesseldorf, Germany; University of Halle, Germany; Helios Klinikum<br />

Berlin-Buch, Berlin, Germany.<br />

10:30 S3-5. Myelodysplatic syndrome and/or acute myelogenous<br />

leukemia (MDS and/or AML) after a breast cancer diagnosis: the<br />

National Comprehensive <strong>Cancer</strong> Network (NCCN) experience<br />

Karp JE, Blackford A, Visvanathan K, Rugo HS, Moy B, Goldstein LJ,<br />

Stockerl-Goldstein K, Neumayer L, Langbaum TS, Hughes ME, Weeks JC,<br />

Wolff AC. Johns Hopkins Kimmel <strong>Cancer</strong> Center; University of California<br />

<strong>San</strong> Francisco; Massachusetts General Hospital; Fox Chase <strong>Cancer</strong> Center;<br />

Washington University School of Medicine; University of Utah School of<br />

Medicine; Dana Farber <strong>Cancer</strong> Institute.<br />

10:45 S3-6. Profiling of triple-negative breast cancers after neoadjuvant<br />

chemotherapy identifies targetable molecular alterations in the<br />

treatment-refractory residual disease<br />

Balko JM, Wang K, <strong>San</strong>ders ME, Kuba MG, Pinto JA, Doimi F, Gomez H,<br />

Palmer G, Cronin MT, Miller VA, Yelensky R, Stephens PJ, Arteaga CL.<br />

Vanderbilt University, Nashville, TN; Foundation Medicine, Cambridge,<br />

MA; Oncosalud, Lima, Peru; Instituto Nacional de Enfermedades<br />

Neoplásicas, Lima, Peru.<br />

11:00 S3-7. Treatment with histone deacetylase inhibitors creates<br />

’BRCAness’ and sensitizes human triple negative breast cancer<br />

cells to PARP inhibitors and cisplatin<br />

Bhalla KN, Rao R, Sharma P, Das Gupta S, Chauhan L, Stecklein S, Fiskus S.<br />

University of Kansas <strong>Cancer</strong> Center, Kansas City, KS.<br />

11:15 S3-8. Identification of novel synthetic-lethal targets for MYCdriven<br />

triple-negative breast cancer<br />

Goga A, Samson S, Horiuchi D. UCSF, <strong>San</strong> Francisco, CA.<br />

11:30 am–12:00 pm<br />

AACR Outstanding Investigator Award for<br />

<strong>Breast</strong> <strong>Cancer</strong> Research, funded by Susan G. Komen for<br />

the Cure ®<br />

Exhibit Hall D<br />

<strong>Breast</strong> Tumor Evolution: Drivers and Clinical Relevance<br />

Kornelia Polyak, MD, PhD<br />

Dana-Farber <strong>Cancer</strong> Institute<br />

Boston, MA<br />

12:00 pm–1:35 pm<br />

LUNCH<br />

12:30 pm–1:35 pm<br />

CASE DISCUSSION 1<br />

Ballroom A<br />

Moderator: Mothaffar Rimawi, MD<br />

Baylor College of Medicine<br />

Houston, TX<br />

Panelists:<br />

Elisabeth Burgess<br />

<strong>Breast</strong> <strong>Cancer</strong> Aotearoa Coalition<br />

Auckland, NEW ZEALAND<br />

Michael Gnant, MD<br />

Medical University of Vienna<br />

Vienna, AUSTRIA<br />

Minetta Liu, MD<br />

Mayo Clinic<br />

Rochester, MN<br />

Eric P. Winer, MD<br />

Dana-Farber <strong>Cancer</strong> Institute<br />

Boston, MA<br />

Richard Zellars, MD<br />

Johns Hopkins University School of Medicine<br />

Baltimore, MD<br />

12:30 pm–1:35 pm<br />

Basic Science Forum<br />

Ballroom B<br />

Epigenetics in <strong>Breast</strong> <strong>Cancer</strong><br />

Moderator: Nancy E. Davidson, MD<br />

University of Pittsburgh <strong>Cancer</strong> Institute,<br />

Pittsburgh, PA<br />

Estrogen-mediated epigenetic gene silencing in luminal<br />

cancers<br />

Tim Hui-Ming Huang, PhD<br />

UT Health Science Center <strong>San</strong> <strong>Antonio</strong><br />

<strong>San</strong> <strong>Antonio</strong>, TX<br />

translational implications of epigenetic gene regulation<br />

Stephen B. Baylin, MD<br />

Johns Hopkins University School of Medicine<br />

Baltimore, MD<br />

1:45 pm–3:15 pm<br />

MINI-SYMPOSIUM 2<br />

Exhibit Hall D<br />

<strong>Breast</strong> Density: Mechanisms and Clinical Implications<br />

Moderator: Melissa Bondy, PhD<br />

Baylor College of Medicine<br />

Houston, TX<br />

Genetics and epidemiology<br />

Celine M. Vachon, PhD<br />

Mayo Clinic College of Medicine<br />

Rochester, MN<br />

www.aacrjournals.org<br />

31s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Clinical<br />

Norman Boyd, MD, DSC<br />

Ontario <strong>Cancer</strong> Institute<br />

Toronto, CANADA<br />

Biological basis of breast density and cancer risk<br />

Thea D. Tlsty, PhD<br />

University of California, <strong>San</strong> Francisco<br />

<strong>San</strong> Francisco, CA<br />

3:15 pm–5:00 pm<br />

GENERAL SESSION 4<br />

Exhibit Hall D<br />

Moderator: Matthew Goetz, MD<br />

Mayo Clinic College of Medicine<br />

Rochester, MN<br />

3:15 S4-1. The UK START (Standardisation of <strong>Breast</strong> Radiotherapy)<br />

Trials: 10-Year follow-up results<br />

Haviland JS, Agrawal R, Aird E, Barrett J, Barrett-Lee P, Brown J, Dewar J,<br />

Dobbs J, Hopwood P, Hoskin P, Lawton P, Magee B, Mills J, Morgan D,<br />

Owen R, Simmons S, Sydenham M, Venables K, Bliss JM, Yarnold JR, on<br />

behalf of the START Trialists. The Institute of <strong>Cancer</strong> Research, Sutton,<br />

United Kingdom; Shrewsbury and Telford Hospital NHS Trust, United<br />

Kingdom; Mount Vernon Hospital, Northwood, United Kingdom; Royal<br />

Berkshire NHS Foundation Trust, Reading, United Kingdom; Velindre<br />

Hospital NHS Trust, Cardiff, United Kingdom; previously University of<br />

Bristol, now Eli Lilly & Company, United Kingdom; formerly Ninewells<br />

Hospital, Dundee, United Kingdom; formerly Guys and St Thomas’<br />

NHS Trust, London, United Kingdom; Nottingham City Hospital, United<br />

Kingdom; formerly The Christie NHS Foundation Trust, Manchester,<br />

United Kingdom; formerly Nottingham University Hospital NHS Trust,<br />

United Kingdom; formerly Cheltenham General Hospital, United<br />

Kingdom; The Institute of <strong>Cancer</strong> Research and Royal Marsden NHS<br />

Foundation Trust, Sutton, United Kingdom.<br />

3:30 S4-2. Targeted intraoperative radiotherapy for early breast cancer:<br />

TARGIT-A trial- updated analysis of local recurrence and first<br />

analysis of survival<br />

Vaidya JS, Wenz F, Bulsara M, Joseph D, Tobias JS, Keshtgar M, Flyger<br />

H, Massarut S, Alvarado M, Saunders C, Eiermann W, Metaxas M,<br />

Sperk E, Sutterlin M, Brown D, Esserman L, Roncadin M, Thompson<br />

A, Dewar JA, Holtveg H, Pigorsch S, Falzon M, Harris E, Matthews A,<br />

Brew-Graves C, Potyka I, Corica T, Williams NR, Baum M. University<br />

College London, London, United Kingdom; University Medical Centre<br />

Mannheim, University of Heidelberg, Heidelberg, Germany; University of<br />

Notre Dame, Fremantle, Australia; Sir Charles Gairdner Hospital, Perth,<br />

Australia; University College Hospital, London, United Kingdom; Royal<br />

Free Hospital, London, United Kingdom; University of Copenhagen,<br />

Copenhagen, Denmark; Centro di Riferimento Oncologia, Aviano, Italy;<br />

University of California, <strong>San</strong> Francisco, CA; University of Western Australia,<br />

Perth, WA, Australia; Red Cross Hospital, Munich, Germany; Ninewells<br />

Hospital, Dundee, United Kingdom; Technical University of Munich,<br />

Munich, Germany; University College London Hospitals, London, United<br />

Kingdom; National <strong>Cancer</strong> Research Institute and Independent <strong>Cancer</strong><br />

Patient’s, Voice, United Kingdom; Moffit <strong>Cancer</strong> Centre , Tampa, FL.<br />

3:45 S4-3. The EndoPredict score identifies late distant metastases in<br />

ER+/HER2- breast cancer patients<br />

Dubsky P, Brase JC, Fisch K, Jakesz R, Singer CF, Greil R, Dietze O, Weber<br />

KE, Petry C, Kronenwett R, Rudas M, Knauer M, Gnant M, Filipits M.<br />

Medical University Vienna, Austria; Sividon Diagnostics GmbH, Cologne,<br />

Germany; Paracelsus Private Medical University, Salzburg, Austria; Sisters<br />

of Charity Hospital, Linz, Austria.<br />

4:00 S4-4. Independent validation of Genomic Grade in the<br />

BIG 1-98 study<br />

Sotiriou C, Ignatiadis M, Desmedt C, Azim Jr. HA, Veys I, Larsimont D,<br />

Lyng M, Viale G, Leyland-Jones B, Ditzel H, Giobbie-Hurder A, Regan M,<br />

Piccart M, Michiels S. Universite Libre de Bruxelles; Institut Jules Bordet;<br />

University of Southern Denmark; University of Milan; Edith <strong>San</strong>ford<br />

Research; Harvard Medical School.<br />

4:15 S4-5. Ki67 levels in pretherapeutic core biopsies as predictive and<br />

prognostic parameters in the neoadjuvant GeparTrio trial<br />

Denkert C. Blohmer JU, Müller BM, Eidtmann H, Eiermann W, Gerber<br />

B, Tesch H, Hilfrich J, Huober J, Fehm T, Barinoff J, Jackisch C, Prinzler<br />

J, Rüdiger T, Budczies J, Erbstößer E, Loibl S, von Minckwitz G. Charite<br />

University Hospital, Berlin, Germany; <strong>San</strong>kt Gertrauden Krankenhaus,<br />

Berlin, Germany; Christian-Albrechts Universität zu Kiel, Kiel, Germany;<br />

Rotkreuzklinikum, München, Germany; Universitätsfrauenklinik Rostock,<br />

Germany; Bethanien Krankenhaus, Frankfurt, Germany; Eilenriede Klinik,<br />

Hannover, Germany; University of Düsseldorf, Germany; Universitäts<br />

Frauenklinik, Tübingen, Germany; Kliniken Essen Mitte, Essen, Germany;<br />

Klinikum Offenbach, Offenbach, Germany; Klinikum Karlsruhe, Karlsruhe,<br />

Germany; Klinikum St. Salvator, Halberstadt, Germany; German <strong>Breast</strong><br />

Group, Neu-Isenburg, Germany.<br />

4:30 S4-6. An international Ki67 reproducibility study<br />

Nielsen TO, Polley M-YC, Leung SCY, Mastropasqua MG, Zabaglo LA,<br />

Bartlett JMS, Viale G, McShane LM, Hayes DF, Dowsett M, on behalf of<br />

the International Ki67 in <strong>Breast</strong> <strong>Cancer</strong> Working Group of the BIG-NABCG<br />

collaboration. University of British Columbia, Vancouver, BC, Canada;<br />

National <strong>Cancer</strong> Institute, Bethesda, MD; European Institute of Oncology,<br />

Milan, Italy; The Institute of <strong>Cancer</strong> Research, London, United Kingdom;<br />

Ontario Institute for <strong>Cancer</strong> Research, Toronto, ON, Canada; University of<br />

Michigan Comprehensive <strong>Cancer</strong> Center, Ann Arbor, MI; Royal Marsden<br />

Hospital, London, United Kingdom; <strong>Breast</strong> International Group-North<br />

American <strong>Breast</strong> <strong>Cancer</strong> Group Collaboration.<br />

4:45 S4-7. Discussion<br />

W. Fraser Symmans, MD, MD Anderson <strong>Cancer</strong> Center, Houston, TX<br />

5:00 pm–7:00 pm<br />

Poster Discussion 5: Clinical Sequencing<br />

Ballroom A<br />

Viewing 5:00 pm<br />

Discussion 5:15 pm<br />

Charles Perou, PhD, Chair<br />

University of North Carolina<br />

Chapel Hill, NC<br />

Katherine Hoadley, PhD, Discussant<br />

University of North Carolina<br />

Chapel Hill, NC<br />

and<br />

Christine Desmedt, PhD, Discussant<br />

Jules Bordet Institute<br />

Bruxelles, BELGIUM<br />

PD05-01 Next generation genomic sequencing (NGS) identifies<br />

molecular targets in inflammatory breast cancer (IBC)<br />

Cristofanilli M, Alpaugh KR, Ross J, Binghman C, Wu H, Stephens<br />

P, Lipson D, Palmer G. Fox Chase <strong>Cancer</strong> Center, Philadelphia, PA;<br />

Foundation Medicine, Cambridge, MA.<br />

PD05-02 Novel mutations in lobular carcinoma in situ (LCIS) as<br />

uncovered by targeted parallel sequencing<br />

De Brot M, Andrade VP, Morrogh M, Berger MF, Won HH, Koslow<br />

Mautner S, Qin L-X, Giri DD, Olvera N, Sakr RA, King TA. Memorial<br />

Sloan-Kettering <strong>Cancer</strong> Center, New York, NY.<br />

PD05-03 What is the appropriate sample (s) on which to perform<br />

sequencing for mutational analysis to guide the selection of<br />

targeted therapy?<br />

Alpaugh RK, Bingham C, Fittipaldi P, Banzi M, Palmer G, Cristofanilli<br />

M. Fox Chase <strong>Cancer</strong> Center, Philadelphia, PA; Silicon Biosystems,<br />

Spa, Bologna, Italy; Foundation Medicine, Cambridge, MA.<br />

PD05-04 Targeted resequencing in oncogenetics : developing a new<br />

approach for molecular diagnostics<br />

Sevenet N, Lafon D, Dupiot-Chiron J, Hubert C, Jones N, Debled M,<br />

Tunon de Lara C, Longy M, Bonnet F. Institut Bergonie, Bordeaux,<br />

France; Centre de Génomique Foncionnelle de Bordeaux, Bordeaux,<br />

France; Institut Bergonie & Universite Bordeaux Segalen, Bordeaux,<br />

France.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012 32s <strong>Cancer</strong> Research


December 4–8, 2012<br />

Program Schedule<br />

PD05-05 RNA-seq identifies unique transcriptomic changes after brief<br />

exposure to preoperative nab-paclitaxel (N), bevacizumab<br />

(B) or trastuzumab (T) and reveals down-regulation of TGF-β<br />

signaling associated with response to bevacizumab<br />

Varadan V, Kamalakaran S, Janevski A, Banerjee N, Lezon-Geyda<br />

K, Miskimen K, Bossuyt V, Abu-Khalaf M, Sikov W, Dimitrova N,<br />

Harris LN. Philips Research North America, Briarcliff Manor, NY; Yale<br />

University School of Medicine, New Haven, CT; Yale Comprehensive<br />

<strong>Cancer</strong> Center, New Haven, CT; Yale University School of Medicine,<br />

Yale Comprehensive <strong>Cancer</strong> Center, New Haven, CT; Warren Alpert<br />

Medical School of Brown University, New Haven, CT; Seidman <strong>Cancer</strong><br />

Center, Cleveland, OH.<br />

PD05-06 Next-generation RNA-sequencing of triple negative breast<br />

cancer compared to donated microdissected normal<br />

epithelium and adjacent normal tissues<br />

Radovich M, Atale R, Clare SE, Sledge GW, Schneider BP. Indiana<br />

University School of Medicine, Indianapolis, IN.<br />

PD05-07 Detection of fusion transcripts among 100 breast cancer<br />

samples by next generation sequencing<br />

Kim J, Han W, Moon H-G, Ahn SK, In Y-H, Kim S, Lee H-S, Lee JW,<br />

Kim JY, Kim T, Kim MK, Noh D-Y. Seoul National University Hospital,<br />

Seoul, Korea; <strong>Cancer</strong> Research institute, Seoul National University,<br />

Seoul, Korea; i-Pharm (Information Center for Bio-pharmacological<br />

Network) IBBI (Integrated Bioscience and Biotechnology Institute)<br />

Advanced Institute of Convergence Technology, Seoul National<br />

University, Seoul, Korea; Seoul National University, Seoul, Korea;<br />

Macrogen Inc., Seoul, Korea.<br />

PD05-08 Genomic characterisation of invasive breast cancers with<br />

heterogeneous HER2 gene amplification<br />

Ng CKY, Gauthier A, Mackay A, Lambros MBK, Rodrigues DN, Arnoud<br />

L, Lacroix-Triki M, Penault-Llorca F, Baranzelli MC, Sastre-Garau<br />

X, Lord CJ, Zvelebil M, Mitsopoulos C, Ashworth A, Natrajan R,<br />

Weigelt B, Delattre O, Cottu P, Reis-Filho JS, Vincent-Salomon A. The<br />

Breakthrough <strong>Breast</strong> <strong>Cancer</strong> Research Centre, The Institute of <strong>Cancer</strong><br />

Research, London, United Kingdom; Institut Curie, Paris, France; CRB<br />

Ferdinand Cabanne, Centre Georges François Leclerc, Dijon, France;<br />

Institut Claudius Regaud, Toulouse, France; Centre Jean Perrin,<br />

Clermont-Ferrand, France; Centre Oscar Lambret, Lille, France.<br />

PD05-09 Whole-genome progression of breast cancer from early<br />

neoplasia to invasive carcinoma<br />

West RB, Kashef-Haghighi D, Newburger D, Weng Z, Brunner A, Salari<br />

R, Guo X, Troxell M, Zhu S, Varma S, Sidow A, Batzoglou S. Stanford<br />

University, Stanford, CA; Oregon Health & Science University,<br />

Portland, OR.<br />

5:00 pm–7:00 pm<br />

Poster Discussion 6: Ki67<br />

Ballroom B<br />

Viewing 5:00 pm<br />

Discussion 5:15 pm<br />

Peter Ravdin, MD, Chair<br />

UT Health Science Center<br />

<strong>San</strong> <strong>Antonio</strong>, TX<br />

Rinat Yerushalmi, MD, Discussant<br />

Davidoff Center<br />

Petach Tikva, ISRAEL<br />

and<br />

Allen M. Gown, MD, Discussant<br />

PhenoPath Labs PLLC<br />

Seattle, WA<br />

PD06-01 Automated computational Ki67 scoring in the GeparTrio<br />

breast cancer study cohort<br />

Klauschen F, Wienert S, Blohmer J-U, Mueller BM, Eiermann W,<br />

Gerber B, Tesch H, Hilfrich J, Huober J, Fehm T, Barinoff J, Jackisch<br />

C, Erbstoesser E, Loibl S, Denkert C, von Minckwitz G. Charite<br />

Universitaetsmedizin Berlin, Berlin, Germany; <strong>San</strong>kt Getrauden<br />

Hospital, Berlin, Germany; University of Munich, Munich, Germany;<br />

Klinikum Suedstadt Rostock, Rostock, Germany; Onkologisches<br />

Zentrum am Bethanien-Kankenhaus, Frankfurt, Germany; Ellenriede<br />

Hospital, Hannover, Germany; University Duesseldorf, Germany;<br />

University of Tuebingen, Germany; Kliniken Essen Mitte, Essen,<br />

Germany; Kliniken Offenbach, Offenbach, Germany; Salvator Hospital,<br />

Germany; German <strong>Breast</strong> Group, Neu-Isenburg, Germany.<br />

PD06-02 Standardized assessment of Ki-67 using virtual slides and<br />

automated analyzer for breast cancer patients: Comparison of<br />

automated and central/local pathology assessment<br />

Mizuno Y, Yamada J, Abe H, Natori T, Sato K. Tokyo-West Tokushukai<br />

Hospital, Tokyo, Japan; SRL, Inc., Tokyo, Japan.<br />

PD06-03 Development of a highly reproducible clinical test for Ki67<br />

using AQUA technology<br />

Murphy DA, Pacia E, Beruti S, Lamoureux J, Dabbas B, Diver J,<br />

Christiansen J. Genoptix Medical Laboratory, Carlsbad, CA.<br />

PD06-04 St. Gallen 2011 clinicopathological subtyping of breast cancer:<br />

impact of different proliferation assessment methods<br />

Focke CM, Gläser D, Finsterbusch K, Decker T. Bonhoeffer Medical<br />

Center, Neubrandenburg, Germany.<br />

PD06-05 St. Gallen 2011 clinico-pathological subtypes of breast cancer:<br />

concordance of core biopsies and related surgical specimens<br />

Decker T, Focke CM. Bonhoeffer Medical Center, Neubrandenburg,<br />

Germany.<br />

PD06-06 Relationship between molecular subtype and change in Ki-67<br />

in the placebo arms of window of opportunity trials<br />

Pruneri G, Gandini S, Guerrieri-Gonzaga A, Serrano D, Cazzaniga<br />

M, Lazzeroni M, Puntoni M, Toesca A, Caldarella P, Johansson H,<br />

Bonanni B, DeCensi A. European Institute of Oncology, Milan,<br />

Italy; Galliera Hospital, Genoa, Italy; University of Milan, School of<br />

Medicine, Milan, Italy.<br />

PD06-07 Evaluation of an optimal cut-off point for the Ki-67 index<br />

as a prognostic factor in primary breast cancer<br />

Tashima R, Nishimur R. Kumamoto Municipal Hospital,<br />

Kumamoto, Japan.<br />

PD06-08 Strong prognostic concordance between Ki67 and binary, but<br />

not multi-level gene expression signatures<br />

Tobin NP, Lindström LS, Carlson JW, Bergh J, Wennmalm K.<br />

Karolinska Institutet and University Hospital, Stockholm, Sweden.<br />

5:00 pm–7:00 pm<br />

POSTER SESSION 3 & RECEPTION<br />

Exhibit Halls A–B<br />

Detection/Diagnosis: <strong>Breast</strong> Imaging - Mammography<br />

P3-01-01 Insulin, Insulin-like Growth Factor-1 and cycling estrogen<br />

predict premenopausal mammographic density<br />

Frydenberg H, Flote VG, Iversen A, Finstad SE, Furberg A-S, Fagerland<br />

M, Wist EA, Schlichting E, Ellison PT, McTiernan A, Ursin G, Thune<br />

I. Oslo University Hospital, Oslo, Norway; University of Tromsø,<br />

Norway; Harvard University, Cambridge, MA; Fred Hutchinson <strong>Cancer</strong><br />

Research Center, Seattle, WA; The Norwegian <strong>Cancer</strong> Registry,<br />

Oslo, Norway.<br />

P3-01-02 Correlation of Mammographic breast density and tumor<br />

characteristics in Korean breast cancer patients<br />

Cho JY, Ahn SH, Lee JW, Yu JH, Koh BS, Kim HJ, Lee JW, Son BH,<br />

Gong G-y, Kim HH. College of Medicine, University of Ulsan, Asan<br />

Medical Center, Seoul, Republic of Korea.<br />

www.aacrjournals.org<br />

33s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P3-01-03 <strong>Breast</strong> density profile in a prospective large breast cancer<br />

screening program in South Brazil<br />

Caleffi M, Zignani J, Clarissa A, Ademar BJ, Ribeiro R, Dakir DF. Núcleo<br />

Mama Porto Alegre, Hospital Moinhos de Vento, Porto Alegre,<br />

RS, Brazil.<br />

P3-01-04 Surgical management of mammographic microcalcification is<br />

improved by full field digital mammography (FFDM)<br />

Bundred SM, Zhou J, Hurley E, Wilson M, Morris J, Bundred NJ.<br />

University Hospital of South Manchester; University of Manchester.<br />

P3-01-05 The Role of Mammographic Calcification in the Neo-adjuvant<br />

Therapy of <strong>Breast</strong> <strong>Cancer</strong> Imaging Evaluation<br />

Li JJ, Chen CM, Di GH, Wu J, Lu JS, Shao Z-M. Shanghai <strong>Cancer</strong><br />

Center and <strong>Cancer</strong> Institute, Shanghai, China.<br />

P3-01-06 Ultrasound as single mode of imaging may miss significant<br />

pathology in symptomatic women aged 35-39<br />

Baker EL, Masudi T, Waterworth A. Calderdale and Huddersfield NHS<br />

Foundation Trust, Huddersfield, West Yorkshire, United Kingdom.<br />

P3-01-07 Estimated risk of radiation-induced breast cancer from<br />

mammographic screening<br />

Freitas-Junior R, Correa RS, Peixoto J-E, Ferreira RS, Tanaka RMN.<br />

Federal University of Goias, Goiania, Goias, Brazil; Comissão Nacional<br />

de Energia Nuclear, Goiania, Goias, Brazil; National <strong>Cancer</strong> Institute<br />

of Brazil, Rio de Janeiro, RJ, Brazil; Superintendência de Vigilância em<br />

Saúde do Estado de Goias, Goiania, Goias, Brazil.<br />

P3-01-08 Mammographic density and estrogen receptor α gene<br />

polymorphism in Javanese ethnic women<br />

Choridah L, Aryandono T, Faisal A, Sadewa AH, Purnomosari D.<br />

Faculty of Medicine, Universitas Gadjah Mada, Yogyakarta, DIY,<br />

Indonesia; Faculty of Medicine, Universitas Gadjah Mada.<br />

Detection/Diagnosis: Screening<br />

P3-02-01 Mammographic Screening: Good Prognosis Tumor Biology in<br />

Screen-detected <strong>Breast</strong> <strong>Cancer</strong>s<br />

Drukker CA, Schmidt MK, Rutgers EJT, Cardoso F, Kerlikowske<br />

K, Esserman LJ, Slaets L, Bogaerts J, van’t Veer LJ. Netherlands<br />

<strong>Cancer</strong> Institute-Antoni van Leeuwenhoek Hospital, Amsterdam,<br />

Netherlands; Champalimaud <strong>Cancer</strong> Centre, Lisbon, Portugal;<br />

University of California, <strong>San</strong> Francisco; EORTC, Brussels, Belgium;<br />

Agendia NV, Amsterdam, Netherlands.<br />

P3-02-02 Use of contrast-enhanced computed tomography in clinical<br />

staging of asymptomatic breast cancer patients to detect<br />

asymptomatic distant metastases<br />

Tanaka S, Sato N, Fujioka H, Takahashi Y, Kimura K, Iwamoto M,<br />

Uchiyama K. Osaka Medical College, Takatsuki City, Osaka, Japan.<br />

P3-02-03 Increased risk of breast cancer in women with falsepositive<br />

screening test: can it be entirely explained by<br />

misclassification?<br />

von Euler-Chelpin M, Kuchiki M, Vejborg I. University of Copenhagen,<br />

Denmark; University Hospital Copenhagen, Denmark.<br />

P3-02-04 Screen detected HER2 positive breast cancer within the<br />

West of London <strong>Breast</strong> Screening population:Incidence,<br />

Management and Outcome<br />

O’Cathail S, White A, Brindley JH, Hadjiminas D, Xynos Y, Cleator<br />

S, Hogben K, Palmieri C. Imperial College Healthcare NHS Trust,<br />

London, United Kingdom.<br />

P3-02-05 Subtypes of screen-detected invasive breast cancer and<br />

symptomatic invasive breast cancer and their impact on<br />

survival<br />

Kobayashi N, Hikichi M, Miyajima S, Utsumi T. Fujita Health University,<br />

Toyoake, Aichi, Japan.<br />

P3-02-06<br />

P3-02-07<br />

P3-02-08<br />

P3-02-09<br />

P3-02-10<br />

P3-02-11<br />

P3-02-12<br />

P3-02-13<br />

Survival impact of early detection of recurrence after surgery<br />

in early breast cancer patients<br />

Hojo T, Tamura K, Masuda N, Inoue K, Kinoshita T, Fujisawa T, Hara<br />

F, Saji S, Asaga S, Anan K, Yamamoto N, Wada N, Takahashi M,<br />

Nakagami K, Kuroi K, Iwata H. National <strong>Cancer</strong> Center Hospital,<br />

Tokyo, Japan; Osaka National Hospital, Osaka, Japan; Saitama <strong>Cancer</strong><br />

Center, Saitama, Japan; Gunma Prefectural <strong>Cancer</strong> Center, Gunma,<br />

Japan; Shikoku <strong>Cancer</strong> Center, Shikoku, Japan; Kyoto University<br />

Graduate School of Medicine, Kyoto, Japan; Kitakyushu Municipal<br />

Medical Center, Kitakyushu, Japan; Chiba <strong>Cancer</strong> Center, Chiba,<br />

Japan; National <strong>Cancer</strong> Center Hospital East, Chiba, Japan; Hokkaido<br />

<strong>Cancer</strong> Center, Hokkaido, Japan; Shizuoka General Hospital, Shizuoka,<br />

Japan; Tokyo Metropolitan <strong>Cancer</strong> and Infectious diseases Center<br />

Komagome Hospital, Tokyo, Japan; Aichi <strong>Cancer</strong> Center Hospital,<br />

Nagoya, Aichi, Japan.<br />

Rates of <strong>Cancer</strong> Detection and Abnormal Results among<br />

Clients Recruited for Mammography through Outreach and<br />

Education Supported by the Avon <strong>Breast</strong> Health Outreach<br />

Program<br />

Hurlbert M, Rose J, Opdyke KM, Gates-Ferris K. Cicatelli Associates<br />

Inc. (CAI), New York, NY; Avon Foundation for Women, New York, NY.<br />

Is repetition of the contralateral mammogram of patients<br />

referred from breast cancer screening for unilateral findings<br />

necessary?<br />

Castro C, Schipper R-J, van Roozendaal L, van Goethem M, Lobbes<br />

M, Smidt M. Maastricht University Medical Center, Maastricht,<br />

Netherlands; Antwerp University Hospital, Antwerp, Belgium.<br />

Cost-effectiveness of screening with additional MRI for<br />

women with familial risk for breast cancer without a genetic<br />

predisposition<br />

Saadatmand S, Heijnsdijk EA, Rutgers EJ, Hoogerbrugge N,<br />

Oosterwijk JC, Tollenaar RA, Hooning M, Obdeijn I-M, de Koning<br />

HJ, Tilanus-Linthorst MM. Erasmus Medical Center, Rotterdam,<br />

Netherlands; The Netherlands <strong>Cancer</strong> Institute, Antoni van<br />

Leeuwenhoek Hospital, Amsterdam, Netherlands; Radboud<br />

University Medical Center, Nijmegen, Netherlands; University Medical<br />

Center Groningen, Netherlands; Leiden University Medical Center,<br />

Leiden, Netherlands; Erasmus Medical Center, Netherlands.<br />

Implementation and uptake of a provincial, population-based,<br />

organized breast screening program for high risk women in<br />

Ontario: The Ontario breast screening program (OBSP) high<br />

risk program<br />

Eisen A, Carroll J, Chiarelli AM, Horgan M, Meschino W, Rabeneck L,<br />

Shumak R, Warner E. Sunnybrook Health Sciences Centre, Toronto,<br />

ON, Canada; University of Toronto, ON, Canada; Mount Sinai Hospital,<br />

Toronto, ON, Canada; <strong>Cancer</strong> Care Ontario, Toronto, ON, Canada;<br />

North York General Hospital, Toronto, ON, Canada.<br />

Screening Magnetic Resonance Imaging (MRI) of the breast<br />

in women at increased lifetime risk for breast cancer: A<br />

retrospective single institution study<br />

Ehsani S, Strigel R, Pettke E, Wilke L, Szalkucki L, Tevaarwerk AJ,<br />

Wisinski KB. University of Wisconsin Carbone <strong>Cancer</strong> Center,<br />

Madison, WI.<br />

The Cost of Mammography Screening in the United States by<br />

Screening Policy<br />

Thorsen CM, Eklund M, Ozanne EM, Esserman LJ. University of<br />

California, <strong>San</strong> Francisco, CA; Karolinska Institute, Stockholm,<br />

Sweden.<br />

Analysis of infrastructure for mammography screening in the<br />

State of Goias, Brazil, 2010<br />

Freitas-Junior R, Correa RS, Peixoto J-E, Rodrigues DCN, Rahal RMS.<br />

Federal University of Goias, Goiania, Goias, Brazil; Comissão Nacional<br />

de Energia Nuclear, Goiania, Goias, Brazil; National <strong>Cancer</strong> Institute of<br />

Brazil, Rio de Janeiro, RJ, Brazil.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012 34s <strong>Cancer</strong> Research


December 4–8, 2012<br />

Program Schedule<br />

P3-02-14 Integrated program for breast cancer control: Partial<br />

presentation of the results obtained in the first 24 months<br />

of operation<br />

Blanco EC, Borges MM, Pinotti M, Gennari MB, Ribeiro IM,<br />

Nascimento CCP, <strong>San</strong>tos LFG, Sahium RC. Oswaldo Cruz Alemão<br />

Hospital, São Paulo, Brazil.<br />

Detection/Diagnosis: Radiology - Tumor Monitoring<br />

P3-03-01 3D mapping of total choline in human breast cancer using<br />

high-speed MR spectroscopic imaging at 3T: initial experience<br />

during neoadjuvant therapy<br />

Posse S, Zhang T, Royce M, Dayao Z, Lopez S, Sillerud L, Casey L,<br />

Eberhardt S, Lomo L, Rajput A, Russell J, Lee S-j, Bolan P. University<br />

of New Mexico School of Medicine and UNM <strong>Cancer</strong> Center,<br />

Albuquerque, NM; New Mexico <strong>Cancer</strong> Center, Albuquerque, NM;<br />

University of Minnesota, Minneapolis, MN.<br />

P3-03-02 The metastatic rate of IMLN, when IMLN metastasis is<br />

suspected with PET CT<br />

Lee J-H, Son G-T, Choi J-E, Kang S-H, Lee S-J. Yeungnam University<br />

College of Medicine, Daegu, Republic of Korea.<br />

P3-03-03 A tri-modality imaging assessment algorithm to evaluate<br />

neoadjuvant therapy response in patients with operable<br />

breast cancer<br />

Umphrey H, Bernreuter W, Bland K, Carpenter J, Falkson C, Forero A,<br />

Keene K, Krontiras H, Meredith R, Urist M, De Los <strong>San</strong>tos J. University<br />

of Alabama at Birmingham, AL.<br />

Tumor Cell and Molecular Biology: Molecular Profiles<br />

P3-04-01 Protein pathway activation mapping of the I-SPY 1 biopsy<br />

specimens identifies new network focused drug targets for<br />

patients with triple negative tumors<br />

Wulfkuhle JD, Wolf D, Gallagher RI, Yau C, Calvert V, Espina V, Illi J,<br />

Wu Q, Boe M, Yan Y, I-SPY TRIAL-1 Investigators, Liotta LA, van’t Veer<br />

L, Esserman L, Petricoin EF. George Mason University, Manassas, VA;<br />

University of California, <strong>San</strong> Francisco, CA; Genentech, South <strong>San</strong><br />

Francisco, CA.<br />

P3-04-02 Protein pathway activation mapping of I-SPY 1 biopsy<br />

specimens identifies new network focused drug targets for<br />

patients with HR+/HER2- tumors<br />

Wulfkuhle JD, Yau C, Gallagher RI, Wolf D, Calvert V, Espina V, Illi J,<br />

Wu Q, Boe M, Yan Y, I-SPY TRIAL-1 Investigators, Liotta LA, van’t Veer<br />

L, Esserman L, Petricoin EF. George Mason University, Manassas, VA;<br />

University of California, <strong>San</strong> Francisco, CA; Genentech, South <strong>San</strong><br />

Francisco, CA.<br />

P3-04-03 A seven-gene profile predicting benefit of postmastectomy<br />

radiotherapy independently of nodal status in high risk breast<br />

cancer<br />

Tramm T, Mohammed H, Myhre S, Alsner J, Børresen-Dale A-L,<br />

Sørlie T, Frigessi A, Overgaard J. Aarhus University Hospital, Aarhus,<br />

Denmark; Oslo University Hospital, Radiumhospitalet, Oslo, Norway;<br />

University Oslo, Norway.<br />

P3-04-04 Identification of a ‘BRCAness’ signature in triple negative<br />

breast cancer by Comparative Genomic Hybridization<br />

Toffoli S, Bar I, Abdulsater F, Delrée P, Hilbert P, Cavallin F, Moreau F,<br />

Clark J, Lacroix-Triki M, Campone M, Martin A-L, Roché H, Machiels<br />

J-P, Carrasco J, Canon J-L. Institute of Pathology and Genetics,<br />

Gosselies, Belgium; MDxHealth, Liège, Belgium; Institut Claudius<br />

Regaud, Toulouse, France; Institut de Cancérologie de l’Ouest-René<br />

Gauducheau, Saint-Herblain - Nantes, France; UNICANCER, Paris,<br />

France; Cliniques Universitaires Saint-Luc, Brussels, Belgium; Grand<br />

Hôpital de Charleroi (GHdC), Charleroi, Belgium.<br />

P3-04-05 Kinomic and phospho-proteomic analysis of breast cancer<br />

stem-like cells<br />

Leth-Larsen R, Christensen AG, Ehmsen S, Moeller M, Palmisano<br />

G, Larsen MR, Ditzel HJ. University of Southern Denmark, Odense,<br />

Denmark; Odense University Hospital, Odense, Denmark.<br />

P3-04-06 Higher gene expression of CSPG4 in the basal-like subtype of<br />

invasive breast cancer and its negative association with lymph<br />

node metastasis<br />

Luo C, Iida J, Chen Y, Dorchak J, Kovatich AJ, Mural RJ, Hu H, Shriver<br />

CD. Windber Research Institute, Windber, PA; Walter Reed National<br />

Military Medical Center, Bethesda, MD; MDR, Global Systems LLC,<br />

Windber, PA.<br />

P3-04-07 Comparison of breast tumors with HER2 amplification and<br />

polysomy 17<br />

Field LA, Deyarmin B, Ellsworth RE, Shriver CD. Windber Research<br />

Institute, Windber, PA; Henry M. Jackson Foundation for the<br />

Advancement of Military Medicine, Windber, PA; Walter Reed<br />

National Military Medical Center, Bethesda, MD.<br />

P3-04-08 Expression of hormone-responsive genes in benign breast<br />

tissue varies with menstrual cycle phase and menopausal<br />

status<br />

Hu H, Wang J, Lee O, Shidfar A, Iyer S, Ivancic D, Chatterton RT,<br />

Stearns V, Sukumar S, Khan SA. Northwestern University, Chicago, IL;<br />

Johns Hopkins University School of Medicine.<br />

P3-04-09 Withdrawn<br />

P3-04-10 Comparison between RNA-Seq and Affymetrix gene<br />

expression data<br />

Fumagalli D, Haibe-Kains B, Michiels S, Brown DN, Gacquer D, Majjaj<br />

S, Salgado R, Larsimont D, Detour V, Piccart M, Sotiriou C, Desmedt<br />

C. Institut Jules Bordet, Brussels, Belgium; Institut de Recherches<br />

Cliniques de Montréal, Montréal, QC, Canada; Université Libre de<br />

Bruxelles, Campus Erasme, Brussels, Belgium.<br />

P3-04-11 Systemic treatment decision making for patients with stage I<br />

and II, hormone receptor positive, her2/neu negative breast<br />

cancer<br />

Zhu X, Graham N, Paquet L, Dent S, Song X. University of Ottawa, ON,<br />

Canada; The Ottawa Hospital <strong>Cancer</strong> Centre, Ottawa, ON, Canada;<br />

Carleton University, Ottawa, ON, Canada.<br />

P3-04-12 <strong>Breast</strong>Mark: An integrated approach to mining publically<br />

available Transcriptomic Datasets relating to <strong>Breast</strong> <strong>Cancer</strong><br />

Outcome<br />

Madden S, Gaule P, Clarke C, Aherne ST, O’Donovan N, Clynes M,<br />

Crown J, Gallagher WM. Molecular Therapeutics for <strong>Cancer</strong> Ireland,<br />

National Institute for Cellular Biotechnology, Dublin City University,<br />

Dublin 9, Ireland; University College Dublin, Dublin 9, Ireland.<br />

Tumor Cell and Molecular Biology: Tumor Heterogeneity/Molecular<br />

Subclassification<br />

P3-05-01 Molecular subtyping improves stratification of patients into<br />

diagnostically more meaningful risk groups<br />

Cristofanilli M, Turk M, Kaul K, Weaver J, Wesseling J, Stork-Sloots L,<br />

de Snoo F, Yao K. Fox Chase <strong>Cancer</strong> Center; NorthShore University<br />

HealthSystem; Netherlands <strong>Cancer</strong> Institute; Agendia NV.<br />

P3-05-02 Pathological assessment of discordant cases for molecular<br />

(BluePrint and MammaPrint) versus clinical subtypes for<br />

breast cancer among 621 patients from the EORTC 10041/BIG<br />

3-04 (MINDACT) trial<br />

Viale G, Slaets L, de Snoo F, van’t Veer L, Rutgers E, Piccart M,<br />

Bogaerts J, van den Akker J, Stork-Sloots L, Engelen K, Russo L,<br />

Dell’Orto P, Cardoso F. European Institute of Oncology; European<br />

Organisation for Research and Treatment of <strong>Cancer</strong>; Agendia NV;<br />

Netherlands <strong>Cancer</strong> Institute; Jules Bordet Institute; Champalimaud<br />

<strong>Cancer</strong> Center.<br />

www.aacrjournals.org<br />

35s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P3-05-03<br />

P3-05-04<br />

P3-05-05<br />

P3-05-06<br />

P3-05-07<br />

P3-05-08<br />

P3-05-09<br />

Characterization of PIK3CA mutations in lobular breast cancer<br />

Desmedt C, Metzger O, Fumagalli D, Brown D, Singhal S, Vincent<br />

D, Adnet P-Y, Smeets D, Bertucci F, Galant C, Salgado R, Veys I,<br />

Saini K, Pruneri G, Krop I, Winer E, Michiels S, Piccart M, Lambrechts<br />

D, Larsimont D, Viale G, Sotiriou C. Institut Jules Bordet, Brussels,<br />

Belgium; Dana-Farber <strong>Cancer</strong> Institute, Boston; Vesalius Research<br />

Centre, VIB, Leuven, Belgium; Institut Paoli Calmettes, Marseille,<br />

France; Université Catholique de Louvain, Brussels, Belgium;<br />

European Institute of Oncology, University of Milan, Milan, Italy.<br />

Intra-tumor heterogeneity as a predictor of therapy response<br />

in HER2 positive breast cancer<br />

Rye IH, Helland Å, Sætersdal A, Naume B, Almendro V, Polyak K,<br />

Børessen-Dale A-L, Russnes HG. Institute for <strong>Cancer</strong> Research, Oslo,<br />

Norway; Oslo University Hospital, Oslo, Norway; University of Oslo,<br />

Oslo, Norway; Dana-Farber <strong>Cancer</strong> Institute and Harvard Medical<br />

School, Boston, MA; Hospital Clínic, Barcelona, Spain.<br />

HER2 Expression and Gene copy analysis by<br />

Immunofluorescence and Fluorescence in situ Hybridization,<br />

on a Single formalin-fixed paraffin-embedded tissue section<br />

Ha T, Seppo A, Ginty F, Kenny K, Henderson D, Kyshtoobayeva A,<br />

Gerdes M, Larriera A, Liu X, Corwin A, Zingelewicz S, Lazare M, Jun<br />

N, Kyshtoobayeva A, Chow C, Al-Kofahi Y, Hollman D, Bloom K. GE<br />

Global Research, Niskayuna, NY; Clarient Diagnostics Services, Aliso<br />

Viejo, CA.<br />

Automated analysis of Her2 FISH using combined<br />

Immunofluorescence and FISH signals<br />

Seppo A, Al-Kofahi Y, Padfield D, Ha T, Jun N, Kyshtoobayeva A,<br />

Kaanumalle L, Corwin A, Henderson D, Kamath V, McCulloch C,<br />

Hollman D, Bloom KJ. GE Global Research, Niskayuna, NY; Clarient<br />

Diagnostics Services, Aliso Viejo, CA.<br />

Poor prognosis early breast cancer: pathological<br />

characteristics of the Unicancer-PACS08 trial including<br />

patients treated with docetaxel or ixabepilone in adjuvant<br />

setting<br />

Lacroix-Triki M, Delrée P, Filleron T, Penault-Llorca F, Bor C, Mery<br />

E, Maisongrosse V, Génin P, Jacquemier J, Reyre J, Caveriviere P,<br />

Quintyn-Ranty M-L, Escourrou G, Mesleard C, Lemonnier J, Martin<br />

A-L, Campone M. Institut Claudius Regaud, Toulouse, France; Institut<br />

de Pathologie et de Génétique, Gosselies, Belgium; Centre Jean<br />

Perrin, Clermont-Ferrand, France; Centre Francois Baclesse, Caen,<br />

France; Centre Alexis Vautrin, Vandoeuvre les Nancy, France; Institut<br />

Paoli Calmettes, Marseille, France; Laboratoire Anatomie et de<br />

Cytologie des Feuillants, Toulouse, France; CHU Rangueil, Toulouse,<br />

France; R&D Unicancer, Paris, France; ICO Centre René Gauducheau,<br />

Saint Herblain, France.<br />

Analysis of the expression of claudin-3, -4, -7 and E-cadherin<br />

in breast cancer: are they surrogates for the claudin-low<br />

subtype?<br />

Tokes A-M, Szasz AM, Kovács AK, Juhasz E, Nemeth Z, Baranyai Z,<br />

Madaras L, Kulka J. Semmelweis University, Budapest, Hungary;<br />

Uzsoki Memorial Hospital, Budapest, Hungary.<br />

The role of TGF-beta receptor type 3 in breast cancer<br />

progression<br />

Jovanovic B, Ashby WJ, Zijlstra A, Pietenpol JA, Moses HL. Vanderbilt<br />

University, Nashville, TN.<br />

Prognostic and Predictive Factors: Response Predictions - Biomarkers and<br />

Other Factors<br />

P3-06-01 Hotspot mutations in PIK3CA are predictive for treatment<br />

outcome on aromatase inhibitors but not for tamoxifen<br />

Ramirez Ardila DE, Helmijr JC, Lurkin I, Look M, Ruigrok-Ritstier K,<br />

Simon I, Van Laere S, Sweep F, Span P, Linn S, Foekens J, Sleijfer<br />

S, Berns EMJJ, Jansen MPHM. Erasmus MC - Daniel den Hoed,<br />

Rotterdam, Zuid Holland, Netherlands; Agendia BV, Amsterdam,<br />

Netherlands; Antwerp University/Oncology Centre, GZA Hospitals<br />

St-Augustinus, Antwerp, Belgium; Radboud University Nijmegen<br />

Medical Center, Nijmegen, Netherlands; Netherlands <strong>Cancer</strong><br />

Institute, Amsterdam, Netherlands.<br />

P3-06-02 Genetic polymorphisms from genome-wide association study<br />

associated with the metabolic and cell proliferation pathways<br />

affect the time to distant metastases of hormone receptorpositive<br />

and Her2-negative early breast cancer<br />

Huang C-S, Kuo S-H, Yang S-Y, Lien H-C, Lin C-H, Lu Y-S, Cheng A-L,<br />

Chang K-J. National Taiwan University Hospital and National Taiwan<br />

University College of Medicine, Taipei, Taiwan; College of Public<br />

Health, National Taiwan University, Taipei, Taiwan.<br />

P3-06-03 Association between PAM50 breast cancer intrinsic subtypes<br />

and effect of gemcitabine in advanced breast cancer patients<br />

Jørgensen CLT, Nielsen TO, Bjerre KD, Liu S, Wallden B, Balslev E,<br />

Nielsen DL, Ejlertsen B. Herlev University Hospital, Herlev, Denmark;<br />

Danish <strong>Breast</strong> <strong>Cancer</strong> Cooperative Group, Copenhagen, Denmark;<br />

University of British Columbia, Vancouver, BC, Canada; NanoString<br />

Technologies, Seattle, WA.<br />

P3-06-04 Role of pMAPkinase, pAKT, p27 & IGF-IR as predictive<br />

markers of response to trastuzumab in patients with HER2-<br />

positive invasive breast cancer treated with neoadjuvant<br />

chemotherapy ± trastuzumab in the REMAGUS02 trial<br />

Mathieu MC, Goubar A, Sigal B, Bertheau P, Guinebretière JM, André<br />

F, Pierga JY, Delaloge S, Giacchetti S, Brain E, Marty M. Institut<br />

Gustave Roussy - INSERM U981, Villejuif, France; Institut Curie, Paris,<br />

France; Hopital Saint-Louis, Paris, France.<br />

P3-06-05 Expression of SPARC in human breast cancer and its predictive<br />

value in the GeparTrio neoadjuvant trial<br />

Untch M, Prinzler J, Fasching P, Müller BM, Gade S, Meinhold-<br />

Heerlein I, Huober J, Karn T, Liedtke C, Loibl S, Müller V, Rack B,<br />

Schem C, Darb-Esfahani S, von Minckwitz G, Denkert C. Helios<br />

Klinikum Berlin-Buch, Germany; Charité Universitätsmedizin<br />

Berlin, Germany; Universitätsklinikum Erlangen, Germany; German<br />

<strong>Breast</strong> Group, Germany; Universitätsklinikum Aachen, Germany;<br />

Universitätsklinikum Düsseldorf, Germany; Universitätsklinikum<br />

Frankfurt am Main, Germany; Universitätsklinikum Münster,<br />

Germany; Universitätsklinikum Hamburg-Eppendorf, Germany;<br />

Universitätsklinikum München; Universitätsklinikum Schleswig-<br />

Holstein, Germany.<br />

P3-06-06 Lactate dehydrogenase B in breast cancer contributes to<br />

glycolytic phenotype and predicts response to neoadjuvant<br />

chemotherapy<br />

Dennison JB, Molina JR, Mitra S, Gonzalez-Angulo AM, Brown RE,<br />

Mills GB. MD Anderson <strong>Cancer</strong> Center, Houston, TX; University of<br />

Texas Health Science Center, Houston, TX.<br />

P3-06-07 Ki67 as a Predictive Marker of Response to Neoadjuvant<br />

Chemotherapy in Patients with Early-Stage <strong>Breast</strong> <strong>Cancer</strong><br />

(ESBC): A Systematic Review and Evidence Summary<br />

Lyman GH, Culakova E, Poniewierski MS, Wogu AF, Barry W, Ginsburg<br />

GS, Marcom PK, Ready N, Abernethy A, Geradts J, Hwang S, Kuderer<br />

NM. Duke University.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012 36s <strong>Cancer</strong> Research


December 4–8, 2012<br />

Program Schedule<br />

P3-06-08<br />

P3-06-09<br />

P3-06-10<br />

P3-06-11<br />

P3-06-12<br />

P3-06-13<br />

Ki-67 mRNA as a predictor for response to neoadjuvant<br />

chemotherapy in primary breast cancer<br />

Marme F, Schneeweiss A, Aigner J, Eidt S, Altevogt P, Sinn P,<br />

Wirtz RM. National Center for Tumor Diseases, University-Hospital<br />

Heidelberg, Germany; Institut of Pathologie at the St.-Elisabeth-<br />

Hospital, Cologne, Germany; German <strong>Cancer</strong> Research Center,<br />

Heidelberg, Germany; University of Heidelberg, Germany;<br />

STRATIFYER MolecularPathology GmbH, Cologne, Germany.<br />

Test of association between Ki67 index of early breast<br />

cancer and local relapse after adjuvant hypofractionated<br />

radiotherapy<br />

Rodrigues DN, Somaiah N, Daley F, Davies S, Rakha E, A’Hern R,<br />

Haviland J, Sydenham M, Owen R, Reis-Filho J, Yarnold JR. Institute<br />

of <strong>Cancer</strong> Research, London, United Kingdom; The Royal Marsden<br />

NHS Foundation Trust, Sutton, Surrey, United Kingdom; Institute<br />

of <strong>Cancer</strong> Research, Sutton, Surrey, United Kingdom; University<br />

of Nottingham and Nottingham University Hospitals NHS Trust,<br />

Nottingham, United Kingdom; Cheltenham General Hospital,<br />

Cheltenham, United Kingdom.<br />

Pretreatment Vitamin D Levels and Response to Neoadjuvant<br />

Chemotherapy in the I-SPY 1 TRIAL<br />

Clark AS, Chen J, Kapoor S, Esserman LJ, DeMichele A, I-SPY TRIAL-1<br />

Investigators. Abramson <strong>Cancer</strong> Center, Philadelphia, PA; Perelman<br />

School of Medicine at the University of Pennsylvania, Philadelphia,<br />

PA; Center for Clinical Epidemiology and Biostatistics, Philadelphia,<br />

PA; Helen Diller Family Comprehensive <strong>Cancer</strong> Center, <strong>San</strong> Francisco,<br />

CA; UCSF/Mount Zion Medical Center, <strong>San</strong> Francisco, CA; I-SPY1<br />

TRIAL Investigators.<br />

Response and long-term outcomes after neo-adjuvant<br />

chemotherapy: Pooled dataset of patients stratified by<br />

molecular subtyping using MammaPrint and BluePrint<br />

Glück S, Peeters J, Stork-Sloots L, Somlo G, van’t Veer L, de Snoo<br />

F. University of Miami, Sylvester Comprehensive <strong>Cancer</strong> Center;<br />

Agendia NV; City of Hope; University of California, <strong>San</strong> Francisco.<br />

Effect of TOP2A and cMYC gene copy number on outcome in<br />

a Phase II trial of adjuvant TC (Docetaxel/Cyclophosphamide)<br />

plus trastuzumab (HER TC) in HER2-positive early stage breast<br />

cancer<br />

Jones S, Collea R, Paul D, Sedlacek S, Favret A, Gore I, Lindquist<br />

DL, Holmes FA, Allison MAK, Steinberg MS, Stokoe C, Portillo<br />

RM, Crockett M, Wang Y, Lina A, Robert NJ, O’Shaughnessy J. US<br />

Oncology Research, McKesson Specialty Health, The Woodlands,<br />

TX; Texas Oncology, Baylor-Sammons <strong>Cancer</strong> Center, Dallas, TX;<br />

New York Oncology Hematology, Albany, NY; Rocky Mountain<br />

<strong>Cancer</strong> Center, Denver, CO; Virginia <strong>Cancer</strong> Specialists, Fairfax, VA;<br />

Birmingham Hematology and Oncology, Birmingham, AL; Arizona<br />

Oncology Associates, Sedona, AZ; Texas Oncology-Texas Memorial<br />

City, Houston, TX; Comprehensive <strong>Cancer</strong> Center, Henderson, NV;<br />

Virginia Oncology Associates, Virginia Beach, VA; Texas Oncology -<br />

Plano East, Plano, TX; Texas Oncology - El Paso West, El Paso, TX.<br />

Expression of Phosphorylated Activating Transcription factor<br />

2 (ATF2) is associated with sensitivity to endocrine therapy in<br />

breast cancer<br />

Palmieri C, Rudraraju B, Abdel-Fatah TMA, Moore DA, Shaw J, Green<br />

A, Ellis IO, Coombes RC, Simak A. Imperial College London, United<br />

Kingdom; Nottingham University City Hospital, Nottingham, United<br />

Kingdom; University of Leicester, United Kingdom.<br />

Identification of Prognosis-Relevant Subgroups in Patients<br />

with Chemoresistant Triple Negative <strong>Breast</strong> <strong>Cancer</strong><br />

Yu K-D, Zhu R, Zhan M, Shao Z-M, Yang W, Symmans WF, Rodriguez<br />

AA, Makris A, Wong ST, Chang JC. Shanghai <strong>Cancer</strong> Center and<br />

<strong>Cancer</strong> Institute of Fudan University, Shanghai, China; The Methodist<br />

Hospital, Houston, TX; The University of Texas MD Anderson <strong>Cancer</strong><br />

Center, Houston, TX; Mount Vernon <strong>Cancer</strong> Centre, United Kingdom;<br />

The Methodist Hospital Research Institute, Houston, TX.<br />

P3-06-15<br />

P3-06-16<br />

P3-06-17<br />

P3-06-18<br />

P3-06-19<br />

P3-06-20<br />

P3-06-21<br />

P3-06-22<br />

Baseline CD4/CD8 tumor infiltrating lymphocytes (TIL) ratio<br />

predicts pathologic response to neoadjuvant chemotherapy<br />

(NC) in breast cancer<br />

García-Martínez E, Luengo Gil G, Chaves Benito A, García García T,<br />

Vicente Conesa AM, Zafra Poves M, García Garre E, Vicente García V,<br />

Ayala de la Peña F. University Hospital Morales Meseguer, Murcia,<br />

Spain; Centro de Hemodonación Regional, Murcia, Spain.<br />

Predictive biomarker of pathologic complete response to<br />

neoadjuvant chemotherapy in triple negative breast cancer<br />

Kim T, Han W, Moon H-G, Noh D-Y. Seoul National University College<br />

of Medicine, Seoul, Korea.<br />

Withdrawn<br />

Increase of serum androgen and its metabolites in<br />

postmenopausal primary breast cancer patients with disease<br />

progression during neo-adjuvant exemestane treatment;<br />

JFMC 34-0601 TR<br />

Takada M, Saji S, Honma N, Masuda N, Yamamoto Y, Kuroi K,<br />

Yamashita H, Ohno S, Aogi K, Ueno T, Toi M. Graduate School<br />

of Medicine, Kyoto University, Kyoto, Japan; Tokyo Metropolitan<br />

Institute of Gerontology, Tokyo, Japan; Osaka National Hospital,<br />

Osaka, Japan; Kumamoto University Hospital, Kumamoto, Japan;<br />

Tokyo Metropolitan <strong>Cancer</strong> and Infectious Diseases Center,<br />

Komagome Hospital, Tokyo, Japan; Hokkaido University, Sapporo,<br />

Japan; National Hospital Organization Kyushu <strong>Cancer</strong> Center,<br />

Fukuoka, Japan; National Hospital Organization Shikoku <strong>Cancer</strong><br />

Center, Ehime, Japan.<br />

Ki-67 mRNA as a predictor for response to neoadjuvant<br />

chemotherapy in primary breast cancer<br />

Aigner J, Schneeweiss A, Marme F, Eidt S, Altevogt P, Sinn P, Wirtz R.<br />

National Center for Tumor Diseases, University-Hospital Heidelberg,<br />

Germany; Institut of Pathology at the St.-Elisabeth-Hospital, Cologne,<br />

Germany; German <strong>Cancer</strong> Research Center, Heidelberg, Germany;<br />

University of Heidelberg, Germany; STRATIFYER MolecularPathology<br />

GmbH, Cologne, Germany.<br />

Is it possible to predict the efficacy of a combination of<br />

Panitumumab plus FEC 100 followed by docetaxel (T) for<br />

patients with triple negative breast cancer (TNBC)? Final<br />

biomarker results from a phase II neoadjuvant trial<br />

Nabholtz J-M, Dauplat M-M, Abrial C, Weber B, Mouret-Reynier M-A,<br />

Gligorov J, Tredan O, Vanlemmens L, Petit T, Guiu S, Jouannaud C,<br />

Tubiana-Mathieu N, Kwiatkowski F, Cayre A, Uhrhammer N, Privat<br />

M, Desrichard A, Chollet P, Chalabi N, Penault-Llorca F. Centre Jean<br />

Perrin, Clermont-Ferrand, France; Centre Alexis Vautrin, Vandoeuvre<br />

les Nancy, France; Hôpital Tenon, Paris, France; Centre Leon Berard,<br />

Lyon, France; Centre Oscar Lambret, Lille, France; Centre Paul Strauss,<br />

Strasbourg, France; Centre Georges François Leclerc, Dijon, France;<br />

Institut Jean Godinot, Reims, France; CHU Dupuytren, Limoges,<br />

France.<br />

Unique Molecular Subtypes of Triple Negative <strong>Breast</strong><br />

Carcinomas by Routine IHC: Implications for Treatment and<br />

Prognosis<br />

Ashfaq R, Wright B, Russell K, Voss A. Caris Life Sciences, Phoenix, AZ.<br />

Mechanisms behind trastuzumab resistance as neoadjuvant<br />

therapy in HER2-positive operable breast cancer<br />

Jinno H, Sato T, Hayashida T, Takahashi M, Hirose S, Kitagawa Y. Keio<br />

University School of Medicine, Shinjuku, Tokyo, Japan.<br />

Predicting response to neoadjuvant letrozole<br />

Larionov A, Turnbull A, Sims A, Renshaw L, Kay C, Harrison D, Dixon<br />

JM. University of Edinburgh, Scotland, United Kingdom.<br />

Early activation of IFN/STAT signaling in tumor cells of patientderived<br />

triple negative breast cancer xenografts predicts<br />

tumor sensitivity to chemotherapy<br />

Legrier M-E, Yvonnet V, Beurdeley A, Stephant G, Le Ven E, Banis S,<br />

Lassalle M, Deas O, Cairo S, Judde J-G. Xentech, Evry, Ile de France,<br />

France.<br />

P3-06-14<br />

P3-06-23<br />

P3-06-24<br />

www.aacrjournals.org<br />

37s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P3-06-25<br />

P3-06-26<br />

P3-06-27<br />

P3-06-28<br />

P3-06-29<br />

P3-06-30<br />

P3-06-31<br />

P3-06-32<br />

P3-06-33<br />

P3-06-34<br />

PTEN mRNA positivity using in situ measurements is<br />

associated with better outcome in Tamoxifen treated breast<br />

cancer patients<br />

Schalper KA, Li K, Rimm DL. Yale School of Medicine, New Haven, CT.<br />

Serum anti-p53 antibody titers predict pathological response<br />

to preoperative chemotherapy in women with HER2 positive<br />

or triple negative breast cancer<br />

Yoshiyama T, Nakamura Y, Igawa A, Saruwatari A, Shigechi T, Ueda N,<br />

Kuba S, Ishida M, Nishimura S, Nishiyama K, Ohno S. National Kyushu<br />

<strong>Cancer</strong> Center, Fukuoka, Japan.<br />

Dynamic tomographic optical breast imaging (TOBI) to<br />

monitor response to neoadjuvant therapy in breast cancer<br />

Carp SA, Wanyo CM, Specht M, Schapira L, Moy B, Finkelstein DM,<br />

Boas DA, Isakoff SJ. Massachusetts General Hospital, Charlestown,<br />

MA; Massachusetts General Hospital, Boston, MA.<br />

Use of the MiCK drug-induced apoptosis assay improves<br />

clinical outcomes in recurrent breast cancer (BRCA)<br />

Bosserman L, Rogers K, Davidson D, Whitworth P, Karimi M,<br />

Upadhyaya G, Rutledge J, Hallquist A, Perree M, Presant C. Wilshire<br />

Oncology Medical Group-US Oncology; Nashville Oncology<br />

Associates; Tennessee Plateau Oncology; Nashville <strong>Breast</strong> Center;<br />

Data Vision; DiaTech Life Sciences.<br />

Change of circulating tumor cells before and after<br />

neoadjuvant chemotherapy in patients with primary<br />

breast cancer<br />

Horiguchi J, Takata D, Rokutanda N, Nagaoka R, Tokiniwa H, Tozuka<br />

K, Sato A, Kikuchi M, Oyama T, Takeyoshi I. Gunma University<br />

Hospital, Maebashi, Gunma, Japan.<br />

Predictors of Pathologic Complete Response to Chemotherapy<br />

and Antiangiotherapy in <strong>Breast</strong> <strong>Cancer</strong><br />

Makhoul I, Griffin RJ, Dhakal I, Raj V, Hennings L, Kadlubar SA.<br />

University of Arkansas for Medical Sciences, Little Rock, AR.<br />

Etirinotecan pegol in patients with metastatic breast cancer<br />

(mBC): Modeling CA27.29 response and its correlation with<br />

tumor response<br />

Chia YL, Hoch U, Hannah A, Eldon MA. Nektar Therapeutics, <strong>San</strong><br />

Francisco, CA.<br />

Topoisomerase 1 gene copy aberration is a frequent finding in<br />

clinical breast cancer samples<br />

Stenvang J, Smid M, Nielsen S, Balslev E, Timmermans M, Rømer M,<br />

Nygaard S, Christensen I, Nielsen D, Foekens J, Brünner N, Martens<br />

J. University of Copenhagen, Denmark; Erasmus University Medical<br />

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

Herlev University Hospital, Copenhagen, Denmark; Copenhagen<br />

Biocenter, Rigshospitalet, Copenhagen, Denmark.<br />

Effect of trastuzumab-based therapy on serum activin A levels<br />

in metastatic breast cancer<br />

Zubritsky LM, Ali SM, Leitzel K, Koestler W, Fuchs E-M, Costa L, Knight<br />

R, Laadem A, Sherman ML, Lipton A. Penn State Hershey Medical<br />

Center, Hershey, PA; Lebanon VA Medical Center, Lebanon, PA;<br />

Medical University of Vienna, Austria; <strong>San</strong>ta Maria Hospital, Lisbon,<br />

Portugal; Celgene Corp., Summit, NJ; Acceleron Pharma, Cambridge,<br />

MA.<br />

Plasma (p) VEGF-A and VEGFR-2 biomarker (BM) results from<br />

the BEATRICE phase III trial of bevacizumab (BEV) in triplenegative<br />

early breast cancer (BC)<br />

Carmeliet P, Pallaud C, Deurloo RJ, Bubuteishvili-Pacaud L, Henschel<br />

V, Dent R, Bell R, Mackey J, Scherer SJ, Cameron D. Vesalius<br />

Research Center, Leuven, Belgium; F. Hoffmann-La Roche Ltd, Basel,<br />

Switzerland; Genentech, Inc., South <strong>San</strong> Francisco; University of<br />

Edinburgh and <strong>Cancer</strong> Services, NHS Lothian, Edinburgh, United<br />

Kingdom; Sunnybrook Health Sciences Center and University of<br />

Toronto, Toronto, ON, Canada; National <strong>Cancer</strong> Center, Singapore,<br />

Singapore; Andrew Love <strong>Cancer</strong> Centre, Geelong, Australia; Cross<br />

Center Institute, Edmonton, Canada.<br />

P3-06-35 Topoisomerase 1 gene copy aberrations in 52 human breast<br />

cancer cell lines and association to gene expression<br />

Stenvang J, Smid M, Nielsen S, Timmermans M, Rømer M, Nielsen<br />

D, Foekens J, Brünner N, Martens J. University of Copenhagen,<br />

Denmark; Erasmus University Medical Center/Daniel den Hoed<br />

<strong>Cancer</strong> Center, Rotterdam, Netherlands; Herlev University Hospital,<br />

Copenhagen, Denmark.<br />

Epidemiology, Risk, and Prevention: Epidemiology - Population Studies<br />

P3-07-01 Withdrawn<br />

P3-07-02 Time-trends in survival in young women with breast cancer in<br />

a SEER population-based study<br />

Ademuyiwa FO, Groman A, Hong C-C, Kumar S, Levine EG, Miller A,<br />

Ambrosone CB. Roswell Park <strong>Cancer</strong> Institute.<br />

P3-07-03 The impact of Carcinoma in situ of the breast and family<br />

history on risk of subsequent breast cancer events and<br />

mortality-a population based study from Sweden<br />

Sackey H, Hui M, Czene K, Edgren G, Frisell J, Hartman M. Karolinska<br />

Institutet, Stockholm, Sweden; Saw Swee Hock School of Public<br />

Health, National University of Singapore, Singapore; Harvard School<br />

of Public Health, Boston; National University Hospital, Singapore.<br />

P3-07-04 Cigarette smoking and postmenopausal breast cancer risk:<br />

results from the NIH-AARP Diet and Health Study<br />

Nyante SJ, Gierach GL, Dallal CM, Park Y, Hollenbeck AR, Brinton LA.<br />

National <strong>Cancer</strong> Institute, Rockville, MD; AARP, Washington, DC.<br />

P3-07-05 Bisphosphonate use after primary breast cancer and risk of<br />

contralateral breast cancer using pharmacy data<br />

Kwan M, Habel L, Song J, Weltzien E, Chung J, Sun Y, Fletcher<br />

S, Haque R. Division of Research, Kaiser Permanente Northern<br />

California; Kaiser Permanente Southern California; Harvard Medical<br />

School.<br />

P3-07-06 Initial treatment and survival among elderly breast<br />

cancer patients with positive estrogen receptor status by<br />

progesterone receptor status and stage: an analysis of US<br />

national registry data 2000-2009<br />

Lang K, Huang H, Namjoshi M, Federico V, Menzin J. Boston Health<br />

Economics, Waltham, MA; Novartis Pharmaceuticals Corporation,<br />

East Hanover, NJ.<br />

P3-07-07 Incidence and survival among breast cancer patients in the<br />

United States by race and stage diagnosis: an analysis of<br />

national registry data 2000-2009<br />

Lang K, Huang H, Namjoshi M, Federico V, Menzin J. Boston Health<br />

Economics, Waltham, MA; Novartis Pharmaceuticals Corporation,<br />

East Hanover, NJ.<br />

P3-07-08 Recent trends in the characteristics of patients with distant<br />

stage breast cancer in the United States Surveillance,<br />

Epidemiology and End Stage Disease (SEER) program<br />

Li J, Dalvi T, Pawaskar M, Tsai K, Caspard H, Fryzek J. Medimmune,<br />

Gaithersburg, MD; Epistat Institute, Gaithersburg, MD.<br />

P3-07-09 Prevalence and effects of off-label use of chemotherapeutic<br />

agents in elderly breast cancer patients: estimates from<br />

Surveillance, Epidemiology and End Results-Medicare data<br />

Eaton AA, Sima CS, Panageas KS. Memorial Sloan-Kettering <strong>Cancer</strong><br />

Center, New York, NY.<br />

P3-07-10 Effect of lifestyle and single nucleotide polymorphisms on<br />

breast cancer risk: A case-control study in Japanese women<br />

Mizoo T, Taira N, Nishiyama K, Nogami T, Iwamoto T, Motoki T, Shien<br />

T, Matuoka J, Doihara H, Ishihara S, Kawai N, Kawasaki K, Ishibe<br />

Y, Ogasawara Y. Okayama Medical University, Okayama, Japan;<br />

Okayama Saiseikai General Hospital, Okayama, Japan; Okayama<br />

Rousai Hospital, Okayama, Japan; Kagawa Prefectural <strong>Cancer</strong><br />

Detection Center, Takamatsu, Kagawa, Japan; Mizushima Kyodo<br />

Hospital, Kurashiki, Okayama, Japan; Kagawa Prefectural Central<br />

Hospital, Takamatsu, Kagawa, Japan.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012 38s <strong>Cancer</strong> Research


December 4–8, 2012<br />

Program Schedule<br />

P3-07-11 Retrospective database review of outcomes in invasive lobular<br />

carcinoma and invasive ductal carcinoma of the breast<br />

Shih Y-C, Dillon P. University of Virginia, Charlottesville, VA.<br />

P3-07-12 Risk Factors for <strong>Breast</strong> <strong>Cancer</strong> in Women Served in Odete<br />

Valadares Maternity, Belo Horizonte-MG<br />

Sediyama CMNO, Peluzio MCG, Abranches MV. Universidade Federal<br />

de Viçosa, Viçosa, MG, Brazil.<br />

P3-07-13 Importance of socioeconomic status in relation to breast<br />

cancer risk and prognostic factors in Argentina<br />

Croce MV, Demichelis SO, Cermignani L, Zwenger A, Segal-Eiras<br />

A, Giacomi N. Faculty of Medical Sciences, UNLP, La Plata, Buenos<br />

Aires, Argentina.<br />

P3-07-14 Initial treatment and survival among elderly breast cancer<br />

patients in the United States by estrogen receptor status and<br />

cancer stage at diagnosis: an analysis of national registry data<br />

2000-2009<br />

Lang K, Huang H, Namjoshi M, Federico V, Menzin J. Boston Health<br />

Economics, Waltham, MA; Novartis Pharmaceuticals Corporation,<br />

East Hanover, NJ.<br />

Epidemiology, Risk, and Prevention: Epidemiology - Genetic and Molecular<br />

P3-08-01 The prospective risk of ovarian cancer in 1433 women in a<br />

breast cancer family history clinic: no increased risk in families<br />

testing negative for BRCA1 and BRCA2<br />

Evans DGR, Ingham SL, Sahin S, Buchan I, Warwick J, O’Hara C,<br />

Moran A, Howell A. St Mary’s Hospital, Manchester, United Kingdom;<br />

Wythenshawe Hospital, Manchester, United Kingdom; The University<br />

of Manchester, Manchester, United Kingdom; Imperial College<br />

London, United Kingdom; North West <strong>Cancer</strong> Intelligence Service,<br />

NHS Foundation Trust, Manchester, United Kingdom; Christie<br />

Hospital, Manchester, United Kingdom.<br />

P3-08-02 Common variants at 10p14 and 1p11.2 display heterogeneity<br />

in breast cancer associations by E-cadherin tumor tissue<br />

expression in two independent datasets<br />

Horne HN, Sherman ME, Garcia-Closas M, Pharoah PD, Blows FM,<br />

Yang XR, Lissowska J, Brinton LA, Chanock SJ, Figueroa JD. National<br />

<strong>Cancer</strong> Institute, Rockville, MD; The Institute of <strong>Cancer</strong> Research,<br />

Sutton, Surrey, United Kingdom; University of Cambridge, United<br />

Kingdom; M. Sklodowska-Curie Memorial <strong>Cancer</strong> Center and Institute<br />

of Oncology, Warsaw, Poland.<br />

P3-08-03 Urinary levels of prostaglandin E 2<br />

metabolite and<br />

postmenopausal breast cancer<br />

Kim S, Taylor JA, <strong>San</strong>dler DP. Georgia Health Sciences University,<br />

Augusta, GA; National Institute of Environmental Health Sciences,<br />

Research Triangle Park, NC.<br />

P3-08-04 Impact of CYP3A variation on estrone levels and breast<br />

cancer risk<br />

Ross GM, Johnson N, Orr N, Walker K, Gibson L, Folkerd E, Haynes B,<br />

Palles C, Coupland B, Shoemaker M, Jones M, Broderick P, Sawyer<br />

E, Kerin M, Tomlinson I, Zvelebil M, Chilcott-Burns S, Tomczyk<br />

K, Simpson G, Williamson J, Hillier S, Houlston R, Swerdlow A,<br />

Ashworth A, Dowsett M, Peto J, dos <strong>San</strong>tos I, Fletcher O. Royal<br />

Marsden Hospital, London, United Kingdom; Breakthrough <strong>Breast</strong><br />

<strong>Cancer</strong>, Institute of <strong>Cancer</strong> Research, London, United Kingdom;<br />

London School of Hygiene and Tropical Medicine, United Kingdom;<br />

Wellcome Trust Centre for Human Genetics, Oxford, United<br />

Kingdom; Biomedical Research Centre, Guys, United Kingdom;<br />

Edinburgh University, United Kingdom; University Hospital, Galway,<br />

Ireland.<br />

P3-08-05 Copy number variation and risk of chemotherapy-related<br />

infection<br />

Abraham JE, Rueda OM, Chin S-F, Guo Q, Harrington P, Earl HM,<br />

Pharoah PD, Caldas C. Strangeway’s Research Laboratory, University<br />

of Cambridge, Cambridgeshire, United Kingdom; University of<br />

Cambridge NHS Foundation Hospitals, Cambridge, Cambridgeshire,<br />

United Kingdom; <strong>Cancer</strong> Research UK Cambridge Research Institute,<br />

Li Ka Shing Centre, Cambridge, Cambridgeshire, United Kingdom.<br />

P3-08-06 Triple Negative <strong>Breast</strong> <strong>Cancer</strong> and BRCA Status: Implications<br />

for Genetic Counseling<br />

Haque R, Alvarado M, Ahmed SA, Shi JM, Chung JWL, Avila CC,<br />

Zheng CX, Tiller GE. Kaiser Permanente Southern California,<br />

Pasadena, CA.<br />

P3-08-07 HER2 positive breast carcinomas: trend in evolution between<br />

1998 and 2008 and relationship with clinico-pathological<br />

characteristics in a population based study<br />

Beltjens F, Bertaut A, Pigeonnat S, Pouget N, Guiu S, Poillot M-L,<br />

Charon-Barra C, Coudert B, Dabakuyo S, Fumoleau P, Arveux P,<br />

Arnould L. Centre GF Leclerc, Dijon, France.<br />

P3-08-08 Vitamin D status at breast cancer diagnosis: correlation with<br />

patient and tumor characteristics<br />

Berger N, Klein P, Boolbol SK, Gillego A, Estabrook A, Malamud<br />

S, Chadha M, Boachie-Adjei K, Shao T. Lutheran Medical Center,<br />

Brooklyn, NY; Beth Israel Medical Center, Continuum <strong>Cancer</strong> Centers<br />

of New York, New York, NY; St. Luke’s-Roosevelt Hospital center,<br />

Continuum <strong>Cancer</strong> Centers of New York, New York, NY.<br />

P3-08-09 Clinical and Pathological Characteristics of <strong>Breast</strong> <strong>Cancer</strong> in<br />

Women with Neurofibromatosis Type 1<br />

Yap Y-S, Wong J, Chan M, Yong W-S, Ong K-W, Lo S-K, Ngeow J,<br />

Tan B, Madhukumar P, Tan M-H, Ang P, Teh B-T, Tan P-H, Lee AS-G.<br />

National <strong>Cancer</strong> Centre Singapore; Singapore General Hospital.<br />

Epidemiology, Risk, and Prevention: Ethnic/Racial Aspects<br />

P3-09-01 Odds of the triple negative subtype and survival of stages 1-3<br />

breast cancer: Variation by race/ethnicity<br />

Parise C, Caggiano V. Sutter Institute for Medical Research,<br />

Sacramento, CA.<br />

P3-09-02 The HER2 –positive subtypes by stage and race/ethnicity<br />

Caggiano V, Parise C. Sutter Institute for Medical Research,<br />

Sacramento, CA.<br />

P3-09-03 <strong>Breast</strong> cancer subtype distribution among HapMap classified<br />

ethnic groups<br />

Luo C, Chen Y, Shriver CD, Hu H, Mural RJ. Windber Research<br />

Institute, Windber, PA; Walter Reed National Military Medical Center,<br />

Bethesda, MD.<br />

P3-09-04 The associations between body mass index and breast cancer<br />

intrinsic subtypes in Japanese women<br />

Kimura K, Tanaka S, Iwamoto M, Uchiyama K. Osaka Medical College,<br />

Takatsuki City, Osaka, Japan.<br />

P3-09-05 The role of breast size in detection of breast cancer in<br />

asian women<br />

Lee S, <strong>San</strong>dhu H, Zheng Y. Holy Name Medical Center.<br />

Treatment: Inflammatory <strong>Breast</strong> <strong>Cancer</strong><br />

P3-10-01 Epidemiological risk factors and normal breast tissue markers<br />

in inflammatory breast cancer<br />

Atkinson RL, Sexton KR, Ueno NT, El-Zein R, Brewster AM,<br />

Krishnamurthy SA, Woodward WA. University of Texas MD Anderson<br />

<strong>Cancer</strong> Center, Houston, TX; Dan L Duncan <strong>Cancer</strong> Center, Baylor<br />

College of Medicine, Houston, TX.<br />

P3-10-02 Modulation of mRNA and miRNAs by the novel histone<br />

deacetylase inhibitor, CG-1521 disrupts cytokinesis in<br />

SUM149PT inflammatory breast cancer cells<br />

Chatterjee N, Tenniswood MPR. University at Albany, Rensselaer, NY.<br />

www.aacrjournals.org<br />

39s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P3-10-03 Bartonella henselae Infection Detected in Patients with<br />

Inflammatory <strong>Breast</strong> <strong>Cancer</strong><br />

Fernandez SV, Aburto L, Maggi R, Breitschwerdt EB, Cristofanilli M.<br />

Fox Chase <strong>Cancer</strong> Center, Philadelphia, PA; North Caroline State<br />

University, Raleigh, NC.<br />

P3-10-04 Regulation of inflammatory breast cancer cell invasion<br />

through Akt1/PKBα phosphorylation of RhoC GTPase<br />

Lehman HL, Van Laere SJ, van Golen CM, Vermeulen PB, Dirix LY, van<br />

Golen KL. The University of Delaware, Newark, DE; Sint Augustine<br />

Hospital, Antwerp, Belgium; Catholic University, Leuven, Belgium;<br />

Delaware State University, Dover, DE.<br />

P3-10-05 Response to neoadjuvant systemic therapy (NST) in<br />

inflammatory breast cancer (IBC) according to estrogen<br />

receptor (ER) and HER2 expression<br />

Masuda H, Iwamoto T, Brewer T, Hsu L, Kai K, Woodward WA,<br />

Reuben JM, Valero V, Alvarez RH, Willey J, Hortobagyi GN, Ueno NT.<br />

Morgan Welch Inflammatory <strong>Breast</strong> <strong>Cancer</strong> Research Program and<br />

Clinic, The University of Texas MD Anderson <strong>Cancer</strong> Center, Houston,<br />

TX; The University of Texas MD Anderson <strong>Cancer</strong> Center, Houston,<br />

TX; Okayama University Hospital, Okayama, Japan.<br />

P3-10-06 Patterns of failure in patients with inflammatory breast<br />

cancer: the case for aggressive local/regional treatment<br />

Warren LE, Regan MM, Nakhlis F, Yeh ED, Jacene HA, Hirshfield-<br />

Bartek J, Overmoyer BA, Bellon JR. Harvard Medical School, Boston,<br />

MA; Dana Farber <strong>Cancer</strong> Institute, Harvard Medical School,<br />

Boston, MA.<br />

P3-10-07 Lymph node status and survival in inflammatory breast cancer<br />

Sieffert MR, Pedersen RC, Tereffe W, Cui H, Woods RR, Viscusi RK,<br />

LeBeau-Grasso L, Lang JE. University of Arizona College of Medicine,<br />

Tucson, AZ; University of Texas MD Anderson <strong>Cancer</strong> Center,<br />

Houston, TX; University of Arizona <strong>Cancer</strong> Center, Tucson, AZ;<br />

University of Arizona Health Sciences Center, Tucson, AZ; University<br />

of Southern California Norris Comprehensive <strong>Cancer</strong> Center, Los<br />

Angeles, CA.<br />

P3-10-08 A new in vitro method of growing and studying inflammatory<br />

breast cancer emboli<br />

Lehman HL, Daasner EJ, Vermeulen PB, Dirix LY, Van Laere S, van<br />

Golen KL. The University of Delaware, Newark, DE; Sint Augustine<br />

Hospital, Antwerp, Belgium; Catholic University, Leuven, Belgium.<br />

P3-10-09 Peptide-based molecular targeting of inflammatory<br />

breast cancer<br />

Eckhardt BL, Miao RY, Cao Y, Driessen WH, Krishnamurthy S,<br />

Arap W, Ueno N, Anderson RL, Pasqualini R. The University of Texas<br />

at MD Anderson <strong>Cancer</strong> Center, Houston, TX; Stanford University<br />

School of Medicine; Trescowthick Research Laboratories, Peter<br />

MacCallum <strong>Cancer</strong> Center; The University of Texas at MD Anderson<br />

<strong>Cancer</strong> Center.<br />

P3-10-10 Status of anaplastic lymphoma kinase (ALK) gene in<br />

inflammatory breast carcinoma<br />

Krishnamurthy S, Woodward W, Reuben JM, Tepperberg J, Ogura<br />

D, Niwa S, Ueno NT. MD Anderson <strong>Cancer</strong> Center, Houston, TX;<br />

LabCorp, Durham, NC; Link Genomics, Toyoka, Japan<br />

Treatment: Male <strong>Breast</strong> <strong>Cancer</strong><br />

P3-11-01 Matched-pair analysis of patients with male and female<br />

breast cancer<br />

Kim M, Lee SK, Choi M-Y, Kim S, Kim J, Jung SP, Bae SY, Kil WH, Lee<br />

JE, Nam SJ. Samsung Medical Center, Sungkyunkwan University<br />

School of Medicine, Seoul, Republic of Korea.<br />

P3-11-02 Male breast cancer: A comparison between BRCA mutation<br />

carriers and non-carriers in Hong Kong, Southern China<br />

Kwong A, Chau WW, Wong CHN, Law FBF, Ng EKO, Suen DTK, Kurian<br />

AW, West DW, Ford JM, Ma ESK. University of Hong Kong, Pokfulam,<br />

Hong Kong; Hong Kong Hereditary <strong>Breast</strong> <strong>Cancer</strong> Family Registry,<br />

Happy Valley, Hong Kong; Stanford University School of Medicine,<br />

Stanford, CA; Hong Kong <strong>San</strong>itorium & Hospital, Happy Valley,<br />

Hong Kong.<br />

P3-11-03 Treatment outcomes for early stage male breast cancer: a<br />

single centre retrospective case-control study<br />

Kwong A, Visram H, Graham N, Balchin K, Petrcich W, Dent S. The<br />

Ottawa Hospital, Ottawa, ON, Canada; Ottawa Hospital Research<br />

Institute, Ottawa, ON, Canada.<br />

P3-11-04 Male breast cancer: Overall survival in a single institution<br />

Bello MA, Bergmann A, Costa CRA, Pinto RR, Millen EC,<br />

Thuler LCS, Bender PFM. Brazilian National <strong>Cancer</strong> Institute,<br />

Rio de Janeiro, Brazil.<br />

Treatment: Brain Metastases<br />

P3-12-01 Serum biomarkers identification using quantitative<br />

proteomics in patients (pts) with untreated brain metastases<br />

from HER2-positive breast cancer receiving capecitabine (C)<br />

and lapatinib (L) (UNICANCER LANDsCAPE trial)<br />

Gonçalves A, Camoin L, Ben Younès I, Romieu G, Campone M, Diéras<br />

V, Cropet C, Mahier Aït-Oukhatar C, Dalenc F, Le Rhun E, Labbe-<br />

Devilliers C, Borg J-P, Bachelot T. Institut Paoli Calmettes, Marseille,<br />

France; Université de la Méditerranée, Marseille, France; Centre Val<br />

d’Aurelle, Montpellier, France; Institut de Cancérologie de l’Ouest,<br />

Nantes, France; Institut Curie, Paris, France; Centre Léon Bérard, Lyon,<br />

France; Unicancer, Paris, France; Institut Claudius Regaud, Toulouse,<br />

France; Centre Oscar Lambret, Lille, France.<br />

P3-12-02 The impact of estrogen receptor status on treatment<br />

outcomes following gamma knife radiosurgery for brain<br />

metastases of primary breast cancer<br />

Cupelo EC, Aridgides PD, Bogart JA, Hahn SS, Shapiro AJ. SUNY<br />

Upstate Medical University, Syracuse, NY.<br />

P3-12-03 Combined targeting of HER2 and VEGFR2 for effective<br />

treatment of HER2-amplified breast cancer brain metastases<br />

Kodack DP, Chung E, Yamashita H, Incio J, Peters A, Song Y, Ager E,<br />

Huang Y, Farrar C, Lussiez A, Goel S, Snuderl M, Kamoun W, Hiddingh<br />

L, Tannous BA, Fukumura D, Engelman JA, Jain RK. Massachusetts<br />

General Hospital, Boston, MA.<br />

P3-12-04 A phase 2, multi-center, open label study evaluating the<br />

efficacy and safety of GRN1005 alone or in combination with<br />

trastuzumab in patients with brain metastases from breast<br />

cancer<br />

Lin NU, Schwartzberg LS, Kesari S, Yardley DA, Verma S, Anders CK,<br />

Shih T, Shen Y, Miller K. UC <strong>San</strong> Diego, La Jolla, CA; Indiana University<br />

Melvin and Bren Simon <strong>Cancer</strong> Center, Indianapolis, IN; Sunnybrook<br />

Odette <strong>Cancer</strong> Centre, Toronto, ON, Canada; Dana-Farber <strong>Cancer</strong><br />

Institute, Boston, MA; The West Clinic, Memphis, TN; Geron, Menlo<br />

Park, CA; University of North Carolina at Chapel Hill, NC; Sarah<br />

Cannon Research Institute, Nashville, TN; Tennessee Oncology, PLLC.,<br />

Nashville, TN.<br />

P3-12-05 Clinical features and outcomes of leptomeningeal metastasis<br />

in patients with breast cancer: a single center experience<br />

Jo J-C, Kang MJ, Ahn J-H, Jung KH, Kim JE, Gong G, Kim HH, Ahn SD,<br />

Kim SS, Son BH, Ahn SH, Kim S-B. Asan Medical Center, University of<br />

Ulsan College of Medicine, Seoul, Korea.<br />

P3-12-06 Clinicopathological Analysis of <strong>Breast</strong> <strong>Cancer</strong> Patients with<br />

Brain Metastases<br />

Ushio A, Sawaki M, Fujita T, Hattori M, Kondo N, Horio A, Gondou N,<br />

Iwata H. Aichi <strong>Cancer</strong> Center, Japan.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012 40s <strong>Cancer</strong> Research


December 4–8, 2012<br />

Program Schedule<br />

P3-12-07 Voyagers and their aids: The role of interactions between<br />

tumor and endothelial cells in brain metastasis<br />

Shah KN, Faridi JS. University of the Pacific, Stockton, CA.<br />

P3-12-08 Incidence, predictive factors and outcome of brain metastases<br />

(BM) in a single institution cohort of breast cancer patients<br />

Perelló A, Alarcon J, Garcias C, Duran J, Colom B, Clemente G, Avella<br />

A, Canet R, Torrecabota J, Rifa J. Hospital Universitari Son Espases,<br />

Palma, Balearic Islands, Spain.<br />

P3-12-09 The risk of brain metastases according to expression<br />

of selected immunohistochemical markers in primary<br />

breast cancers<br />

Sosinska-Mielcarek K, Winczura P, Duchnowska R, Badzio A,<br />

Majewska H, Lakomy J, Peksa R, Pieczynska B, Radecka B, Debska<br />

S, Zok J, Rogowski W, Strzelecka M, Kulma-Kreft M, Blaszczyk<br />

P, Litwiniuk M, Jesien-Lewandowicz E, Rutkowski T, Jaworska-<br />

Jankowska M, Adamowicz K, Foszczynska-Kloda M, Biernat W,<br />

Jassem J. Regional Oncology Center, Gdansk, Poland; Medical<br />

University of Gdansk, Poland; Military Institute of Medicine, Warsaw,<br />

Poland; Regional Oncology Center in Opole, Opole, Poland; Medical<br />

University of Lódz, Poland; Warmia and Masuria Oncology Center,<br />

Olsztyn, Poland; PCK Marine Hospital, Gdynia, Poland; Oncology<br />

Center in Bydgoszcz, Poland; Medical University of Poznan, Poland;<br />

Maria Sklodowska-Curie Memorial <strong>Cancer</strong> Center and Institute of<br />

Oncology, Gliwice, Poland; Regional Hospital in Wroclaw, Wroclaw,<br />

Poland; Pomeranian Oncology Center, Szczecin, Poland.<br />

P3-12-10 Age, breast cancer subtype approximation and the risk of the<br />

development of brain metastasis in breast cancer patients<br />

Hung M-H, Liu C-Y, Chao T-C, Wang Y-L, Tsai Y-F, King K-L, Chiu J-H,<br />

Shiau C-Y, Shyr Y-M, Tzeng C-H, Tseng L-M. Taipei Veterans General<br />

Hospital, Taipei, Taiwan; National Yang-Ming University,<br />

Taipei, Taiwan.<br />

P3-12-11 Clinical outcome in patients with surgically resected brain<br />

metastases from breast cancer: prognostic considerations<br />

regarding molecular status and established prognostic<br />

classification systems<br />

Tabouret E, Metellus P, Tallet A, Figarella-Branger D, Charaffe-Jauffret<br />

E, Viens P, Gonçalves A. Institut Paoli Calmettes, Marseille, France;<br />

APHM, Marseille, France; Insitut Paoli-Calmettes, Marseille, France.<br />

P3-12-12 Chemotherapy and targeted therapy after whole-brain<br />

radiotherapy may improve survival in RPA class II/III patients<br />

with brain metastases from breast cancer<br />

Zhang Q, Chen J, Cai G, Yang Z, Chen J. Fudan University Shanghai<br />

<strong>Cancer</strong> Center, Shanghai, China.<br />

Treatment: Bone Metastases<br />

P3-13-01 Prospective study of treatment pattern, effectiveness, and<br />

safety of zoledronic acid (ZOL) therapy beyond 24 months:<br />

subgroup analysis of patients (pts) with metastatic bone<br />

disease (MBD) from breast cancer (BC)<br />

Van den Wyngaert T, Delforge M, Doyen C, Duck L, Wouters K,<br />

Delabaye I, Wouters C, Wildiers H. Antwerp University Hospital;<br />

University Hospitals Leuven; C.H.U. Mont – Godinne; Clinique<br />

St-Pierre; Novartis Pharmaceuticals.<br />

P3-13-02 Safety and Efficacy of Zoledronic Acid Beyond 24 Months in<br />

<strong>Breast</strong> <strong>Cancer</strong> Patients<br />

Suzuki Y, Saito Y, Ogiya R, Oshitanai R, Terao M, Terada M, Morioka T,<br />

Tsuda B, Niikura N, Okamura T, Tokuda Y. Tokai University School of<br />

Medicine, Isehara, Kanagawa, Japan.<br />

P3-13-03 Molecular factors associated with bone metastases in breast<br />

cancer patients<br />

Winczura P, Sosinska-Mielcarek K, Duchnowska R, Badzio A, Lakomy<br />

J, Majewska H, Peksa R, Pieczynska B, Radecka B, Debska S, Zok J,<br />

Rogowski W, Strzelecka M, Kulma-Kreft M, Blaszczyk P, Litwiniuk<br />

M, Jesien-Lewandowicz E, Rutkowski T, Jaworska-Jankowska M,<br />

Adamowicz K, Foszczynska-Kloda M, Biernat W, Jassem J. Medical<br />

University of Gdansk, Poland; Regional Oncology Center, Gdansk,<br />

Poland; Regional Oncology Center, Opole, Poland; Military Institute<br />

of Medicine, Warsaw, Poland; Medical University of Lódz, Poland;<br />

Warmia and Masuria Oncology Center, Olsztyn, Poland; Maritime<br />

Hospital, Gdynia, Poland; Oncology Center, Bydgoszcz, Poland;<br />

Maria Sklodowska-Curie Memorial <strong>Cancer</strong> Center and Institute of<br />

Oncology, Gliwice, Poland; Regional Hospital in Wroclaw, Poland;<br />

West Pomeranian Oncology Center, Szczecin, Poland.<br />

P3-13-04 Skeletal related Events and Bone Metastasis Patients with<br />

<strong>Breast</strong> <strong>Cancer</strong> in Japan. A Retrospective Study<br />

Yamashiro H, Takada M, Imai S, Yamauch A, Tsuyuki S, Inamoto<br />

T, Matsutani Y, Sakata S, Wada Y, Okamura R, Harada T, Tanaka F,<br />

Moriguchi Y, Kato H, Higashide S, Kan N, Yoshibayashi H, Suwa<br />

H, Okino T, Nakayama I, Ichinose Y, Yamagami K, Hashimoto T,<br />

Nakatani E, Nagata Y, Kudo Y, Toi M. National Hospital Organization<br />

KURE Medical Center, Kure, Hiroshima, Japan; Graduate School<br />

of Medicine, Kyoto University, Kyoto, Japan; Kurashiki Central<br />

Hospital, Kurashiki, Okayama, Japan; Kitano Hospital (The Tazuke<br />

Kofukai Medical Research Institute), Osaka, Japan; Osaka Red Cross<br />

Hospital, Osaka, Japan; Tenri Hospital, Tenri, Nara, Japan; National<br />

Hospital Organization Kyoto Medical Center, Kyoto, Japan; National<br />

Hospital Organization Himeji Medical Center, Himeji, Hyogo, Japan;<br />

Yamatotakada Municipal Hospital, Yamatotakada, Nara, Japan;<br />

Osaka Saiseikai Noe Hospital, Osaka, Japan; Fukui Red Cross Hospital,<br />

Fukui, Japan; Kyoto City Hospital, Kyoto, Japan; Nagahama City<br />

Hospital, Nagahama, Shiga, Japan; Kan Norimichi Clinic, Kyoto,<br />

Japan; Japanese Red Cross Society Wakayama Medical Center,<br />

Wakayama, Wakayama, Japan; Hyogo Prefectural Tsukaguchi<br />

Hospital, Amagasaki, Hyogo, Japan; Kouga Public Hospital, Kouga,<br />

Shiga, Japan; Kyoto Min-iren Chuo Hospital, Kyoto, Japan; Takatsuki<br />

Red Cross Hospital, Takatsuki, Osaka, Japan; Shinko Hospital, Kobe,<br />

Hyogo, Japan; Hashimoto Clinic, Kobe, Hyogo, Japan; Translational<br />

Research Informatics Center, Foundation for Biomedical Research<br />

and Innovation, Kobe, Hyogo, Japan; Kobe City Medical Center<br />

General Hospital, Kobe, Hyogo, Japan.<br />

P3-13-05 Evaluating efficacy of de-escalated bisphosphonate therapy<br />

in metastatic breast cancer patients at low-risk of skeletal<br />

related events. TRIUMPH: A pragmatic multicentre trial<br />

Bouganim N, Vandermeer L, Kuchuk I, Dent S, Hopkins S, Song X,<br />

Robbins D, Spencer P, Mazzarello S, Hilton JF, Amir E, Dranitsaris G,<br />

Addison C, Mallick R, Clemons MJ. McGill University Health Center,<br />

Montreal, QC, Canada; Ottawa Hospital <strong>Cancer</strong> Center, Ottawa,<br />

ON, Canada; Princess Margaret Hospital and University of Toronto,<br />

Toronto, ON, Canada; Health Economics and Biostatistics Consultant,<br />

Toronto, ON, Canada; The Ottawa Hospital Research Institute,<br />

Ottawa, ON.<br />

P3-13-06 Osteocytic Connexin 43 Hemichannels in Prevention of<br />

Bone Metastasis<br />

Zhou JZ, Jiang JX. University of Texas Health Science Center,<br />

<strong>San</strong> <strong>Antonio</strong>, TX.<br />

Treatment: DCIS/LCIS<br />

P3-14-01 Molecular definition of the transition of ductal carcinoma in<br />

situ (DCIS) to invasive ductal carcinoma (IDC)<br />

Colavito S, Stepansky A, Madan A, Harris LN, Hicks J, Bossuyt V, Rimm<br />

D, Lannin D, Stern DF. Yale University, New Haven, CT; Cold Spring<br />

Harbor Laboratory, Cold Spring Harbor, NY; Case Western Reserve<br />

University, Cleveland, OH.<br />

www.aacrjournals.org<br />

41s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P3-14-02<br />

Withdrawn<br />

P3-14-03 Differences in pathological and biological factors between<br />

DCIS, DCIS with microinvasion (DCIS-MI) and DCIS with<br />

concomitant invasive ductal carcinoma (DCIS-IDC)<br />

MacGrogan G, Baranzelli MC, Picquenot JM, Penault-Llorca F,<br />

Mathieu MC, Tas P, Fermeaux V, Mery E, Sagan C, Blanc-Fournier C,<br />

Brabencova E, Arnould L, Jacquemier J, Ettore F, Velasco V, Gonzalves<br />

B, Brouste V, Tunon de Lara C. Institut Bergonié, Bordeaux, France;<br />

Centre Oscar Lambret, Lille, France; Centre Henri Becquerel, Rouen,<br />

France; Centre Jean Perrin, Clermont-Ferrand, France; Institut<br />

Gustave Roussy, Villejuif, France; Centre Eugène Marquis, Rennes,<br />

France; CHU de Limoges, Limoges, France; Institut Claudius Regaud,<br />

Toulouse, France; Centre René Gauducheau, Saint Herblain, France;<br />

Centre François Baclesse, Caen, France; Institut Jean Godinot, Reims,<br />

France; Centre Georges François Leclerc, Dijon, France; Institut Paoli<br />

Calmettes, Marseille, France; Centre Antoigne Lacassagne, Nice,<br />

France.<br />

P3-14-04 Symptomatic DCIS is a risk factor for invasion and should be<br />

managed accordingly<br />

Dimopoulos N, Williams K, Kirwan CC, Johnson R, Howe M, Bundred<br />

NJ. University Hospital of South Manchester.<br />

P3-14-05 Recurrence in Patients Diagnosed with Ductal Carcinoma In<br />

Situ: Predictors and Prognostic Significance<br />

Sue GR, Killelea B, Horowitz NR, Lannin DR, Chagpar AB. Yale<br />

University School of Medicine, New Haven, CT.<br />

P3-14-06 The Utility of Margin Index To Predict Residual DCIS Following<br />

<strong>Breast</strong> Conserving Surgery<br />

Aneja S, Lannin DR, Killelea B, Horowitz NR, Chagpar AB. Yale<br />

University School of Medicine, New Haven, CT.<br />

Ongoing Trials 2: Surgery/Nodes<br />

OT2-1-01 Feasibility of sentinel node detection after neoadjuvant<br />

chemotherapy for patient with proved axillary lymph node<br />

involvement: the French prospective multiinstitutional GANEA<br />

2 ongoing trial<br />

Classe J-M, Bordes V, Gimbergues P, Tunon de Lara C, Faure C,<br />

Belichard C, Houpeau J-L, Raro P, Dupré P-F, Houvenaeghel G,<br />

Barranger E, Marchal F, Deblay P, Rouanet P, Lefebvre C, Bourcier C,<br />

Alran S. Institut de <strong>Cancer</strong>ologie de l’Ouest, Nantes Saint Herblain,<br />

France; Centre Jean Perrin, Clermont-Ferrand, France; Institut<br />

Bergonié, Bordeaux, France; Centre Leon Berard, Lyon, France;<br />

Centre Huguenin, Saint Cloud, France; Centre Oscar Lambret, Lille,<br />

France; Centre Hospitalier Universitaire Morvan, Brest, France; Paoli<br />

Calmettes, Marseille, France; CHU Lariboisière, Paris, France; Centre<br />

Alexis Vautrin, Nancy, France; Centre


December 4–8, 2012<br />

Program Schedule<br />

OT2-2-05 A prospective, randomised multi-centre phase II study<br />

evaluating the adjuvant, neoadjuvant or palliative treatment<br />

with tamoxifen +/- GnRH analogue versus aromatase inhibitor<br />

+ GnRH analogue in male breast cancer patients (GBG-54<br />

MALE)<br />

Linder M, von Minckwitz G, Kamischke A, Rudlowski C, Eggemann<br />

H, Nekljudova V, Loibl S. German <strong>Breast</strong> Group, Neu-Isenburg;<br />

Kinderwunschzentrum Münster, Germany; Universitätsfrauenklinik<br />

Bonn; Universitätsfrauenklinik Magdeburg.<br />

Ongoing Trials 2: Targeted Agents<br />

OT2-3-01 Phase Ib pilot study to evaluate reparixin in combination with<br />

chemotherapy with weekly paclitaxel in patients with HER-2<br />

negative metastatic breast cancer (MBC)<br />

Schott AF, Wicha M, Cristofanilli M, Ruffini P, McCanna S, Reuben<br />

JM, Goldstein LJ. University of Michigan, Ann Arbor, MI; Fox Chase<br />

<strong>Cancer</strong> Center, Philadelphia, PA; Dompé s.p.a., Milano, Italy; MD<br />

Anderson <strong>Cancer</strong> Center, Houston, TX.<br />

OT2-3-02 Phase Ib/II study of an oral PI3K/mTOR inhibitor plus letrozole<br />

compared with letrozole (L) in pre-operative setting in<br />

patients with Estrogen Receptor-positive, HER2-negative early<br />

breast cancer (BC): Phase Ib preliminary data<br />

Canon JL, Bergh J, Saura C, Oliveira M, Houk B, Millham R, Barton<br />

J, Dowsett M, Giorgetti C. Grand Hopital de Charleroi, Charleroi,<br />

Belgium; Karolinska Institutet and University Hospital, Stockholm,<br />

Sweden; Vall d’Hebron University Hospital, Vall d’Hebron Institute of<br />

Oncology (VHIO), Barcelona, Spain; Pfizer Oncology, La Jolla; Pfizer<br />

Oncology, Groton; Pfizer Biotechnology Unit & Oncology Clinical<br />

Research, <strong>San</strong> Diego; Royal Marsden Hospital, London, United<br />

Kingdom; Pfizer Oncology, Milan, Italy.<br />

OT2-3-03 Denosumab versus placebo as adjuvant treatment for<br />

women with early-stage breast cancer at high risk of<br />

disease recurrence (D-CARE): An international, randomized,<br />

double-blind, placebo-controlled phase 3 clinical trial<br />

Goss PE, Barrios CH, Chan A, Finkelstein DM, Iwata H, Martin M, Braun<br />

A, Ke C, Maniar T, Coleman RE. Massachusetts General Hospital,<br />

Boston, MA; PUCRS School of Medicine, Porto Alegre, Brazil; <strong>Breast</strong><br />

<strong>Cancer</strong> Research Centre, Perth, WA, Australia; Aichi <strong>Cancer</strong> Center,<br />

Nagoya, Chikusa-ku, Japan; Hospital Gregorio Maranon, Madrid,<br />

Spain; Amgen Inc., Thousand Oaks, CA; CR-UK/YCR Sheffield <strong>Cancer</strong><br />

Research Centre, Sheffield, United Kingdom.<br />

OT2-3-04 A pilot phase II study to evaluate the impact of denosumab<br />

on disseminated tumor cells (DTC) in patients with early stage<br />

breast cancer (ESBC)<br />

Li J, Rugo HS. University of California, <strong>San</strong> Francisco, CA.<br />

OT2-3-05 AVASTEM: a phase II randomized trial evaluating anticancer<br />

stem cell activity of pre-operative bevacizumab and<br />

chemotherapy in breast cancer<br />

Gonçalves A, Pierga J-Y, Brain E, Tarpin C, Cure H, Esterni B, Boyer-<br />

Chammard A, Bertucci F, Jalaguier A, Houvenaeghel G, Viens P,<br />

Charaffe-Jauffret E, Extra J-M. Institut Paoli Calmettes, Marseille,<br />

France; Institut Curie-Centre René Huguenin, Paris, France; Institut<br />

Jean Godinot, Reims, France.<br />

OT2-3-06 A phase II, non-randomized, multicenter, exploratory trial<br />

of single agent BKM120 in patients with triple-negative<br />

metastatic breast cancer<br />

Saura C, Lin N, Ciruelos E, Maurer M, Lluch A, Gavilé J, Winer E,<br />

Baselga J, Rodón J. Vall d’Hebron University Hospital, Barcelona,<br />

Spain; Dana-Farber <strong>Cancer</strong> Institute, Boston, MA; Hospital 12 de<br />

Octubre, Madrid, Spain; Columbia University Medical Center,<br />

New York; Hospital Clínico de Valencia, Valencia, Spain; Instituto<br />

Valenciano de Oncología, Valencia, Spain; Massachusetts General<br />

Hospital, Boston; SOLTI <strong>Breast</strong> <strong>Cancer</strong> Research Group,<br />

Barcelona, Spain.<br />

OT2-3-07<br />

OT2-3-08<br />

OT2-3-09<br />

OT2-3-10<br />

OT2-3-11<br />

A randomized, phase 2 study of the poly (ADP-ribose)<br />

polymerase (PARP) inhibitor veliparib (ABT-888) in<br />

combination with temozolomide (TMZ) or in combination<br />

with carboplatin (C) and paclitaxel (P) versus placebo plus C/P<br />

in subjects with BRCA1 or BRACA2 mutation and metastatic<br />

breast cancer<br />

Isakoff SJ, Pulhalla S, Shepherd SP, Falotico N, Kaufman B, Friedlander<br />

M, Robson M, Domchek S, Garber J, McKeegan E, Chyla B, Qian J,<br />

Giranda VL. Massachusetts General Hospital, Boston, MA; University<br />

of Pittsburgh, PA; Abbott, Abbott Park, IL, Virgin Islands, British;<br />

Sheba Medical Center, Israel; Prince of Wales Hospital, Sydney,<br />

Australia; Memorial Sloan-Kettering <strong>Cancer</strong> Center, New York, NY;<br />

University of Pennsylvania, Philadelphia, PA; Dana Farber <strong>Cancer</strong><br />

Institute, Boston, MA.<br />

Phase III randomized study of the oral pan-PI3K inhibitor<br />

BKM120 with fulvestrant in postmenopausal women with<br />

HR+/HER2– locally advanced or metastatic breast cancer,<br />

treated with aromatase inhibitor, and progressed on or after<br />

mTOR inhibitor-based treatment – BELLE-3<br />

Di Leo A, Germa C, Weber D, Di Tomaso E, Dharan B, Massacesi C,<br />

Hirawat S. <strong>San</strong>dro Pitigliani Hospital of Prato, Prato, Italy; Novartis<br />

Oncology, Paris, France; Novartis Institutes for BioMedical Research,<br />

Inc., Cambridge, MA; Novartis Pharmaceuticals Corporation, East<br />

Hanover, NJ.<br />

Phase III randomized study of the oral pan-PI3K inhibitor<br />

BKM120 with fulvestrant in postmenopausal women with<br />

HR+/HER2– locally advanced or metastatic breast cancer<br />

resistant to aromatase inhibitor – BELLE-2<br />

Baselga J, Campone M, Cortes J, Iwata H, de Laurentiis M, Jonat W,<br />

Di Tomaso E, Hachemi S, Goteti S, Germa C, Massacesi C, Arteaga<br />

C. Massachusetts General Hospital, Boston, MA; Centre René<br />

Gauducheau, Nantes, France; Vall d’Hebron Institute Oncology,<br />

Barcelona, Spain; Aichi <strong>Cancer</strong> Center Hospital, Nagoya, Aichi, Japan;<br />

Università degli Studi di Napoli Federico II, Naples, Italy; Christian-<br />

Albrechts-Universität zu Kiel, Kiel, Germany; Novartis Institutes for<br />

BioMedical Research, Inc., Cambridge, MA; Novartis Oncology, Paris,<br />

France; Novartis Pharmaceuticals Corporation, East Hanover, NJ;<br />

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

Phase II study of panitumumab, nab-paclitaxel, and<br />

carboplatin for patients with primary inflammatory breast<br />

cancer (IBC) without HER2 overexpression<br />

Willey JS, Alvarez RH, Valero V, Lara JM, Parker CA, Hortobagyi GN,<br />

Ueno NT. Morgan Welch Inflammatory <strong>Breast</strong> <strong>Cancer</strong> Research<br />

Program and Clinic, The University of Texas MD Anderson <strong>Cancer</strong><br />

Center, Houston, TX; University of TX, MD Anderson <strong>Cancer</strong> Center,<br />

Houston, TX.<br />

Tivozanib in combination with paclitaxel vs placebo with<br />

paclitaxel in patients with locally advanced or metastatic<br />

triple-negative breast cancer<br />

Mayer EL, Miller K, O’Shaughnessy J, Dickler M, Vogel C, Leyland-<br />

Jones B, Steelman L, Robinson M, Kuriyama N, Agarwal S. <strong>Breast</strong><br />

Oncology Center, Dana-Farber <strong>Cancer</strong> Institute, Boston, MA; Indiana<br />

University Melvin and Bren Simon <strong>Cancer</strong> Center, Indianapolis, IN;<br />

Baylor-Charles A. Sammons <strong>Cancer</strong> Center, Texas Oncology and US<br />

Oncology, Dallas, TX; Memorial Sloan-Kettering <strong>Cancer</strong> Center, New<br />

York, NY; Sylvester Comprehensive <strong>Cancer</strong> Center, Miami, FL; <strong>San</strong>ford<br />

Research/USD, Sioux Falls, SD; AVEO Oncology, Cambridge, MA.<br />

7:30 pm–10:00 pm<br />

OPEN SATELLITE EVENT presented by Clinical Care Options<br />

Marriott Rivercenter<br />

HER2-Positive <strong>Breast</strong> <strong>Cancer</strong>: Applying the Latest Developments to<br />

Clinical Practice<br />

WEBSITE: http://clinicaloptions.com/HER2Positive<br />

www.aacrjournals.org<br />

43s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

7:30 pm–10:00 pm<br />

OPEN SATELLITE EVENT presented by Clinical Care Options<br />

Marriott Rivercenter<br />

Practical Answers to Your Clinical Challenges: Optimal Use of Bone-<br />

Modifying Agents in <strong>Breast</strong> <strong>Cancer</strong><br />

WEBSITE: http://clinicaloptions.com/BoneHealth<strong>Breast</strong><strong>Cancer</strong><br />

Friday, December 7, 2012<br />

6:45 am–5:15 pm<br />

Registration<br />

Bridge Hall<br />

7:00 am–9:00 am<br />

Poster Discussion 7: Neoadjuvant Endocrine Therapy &<br />

Bisphosphonates<br />

Ballroom A<br />

Viewing 7:00 am<br />

Discussion 7:45 am<br />

Ruth O’Regan, MD, Chair and Discussant<br />

Emory University School of Medicine<br />

Atlanta, GA<br />

and<br />

Ingrid Mayer, MD, MSCI, Discussant<br />

Vanderbilt University Medical Center<br />

Nashville, TN<br />

PD07-01 Z1031B neoadjuvant aromatase inhibitor trial: A Phase 2 study<br />

of triage to chemotherapy based on 2 to 4 week Ki67 level<br />

>10%<br />

Ellis MJ, Suman V, McCall L, Luo R, Hoog J, Brink A, Watson M, Ma C,<br />

Unzeitig G, Pluard T, Whitworth P, Babiera G, Guenther M, Dayao Z,<br />

Leitch M, Ota D, Olson J, Hunt K, Allred C. Siteman <strong>Cancer</strong> Center,<br />

Washington University, St Louis, MO; Mayo Clinic; Duke University;<br />

Doctor’s Hospital of Laredo; Nashville <strong>Breast</strong> Center; MD Anderson<br />

<strong>Cancer</strong> Center; St. Elizabeth Med Ctr Southwest; Univ of New Mexico;<br />

UT Southwestern; University of Maryland Medical Center.<br />

PD07-02 Anticancer activity of letrozole plus zoledronic acid as<br />

neoadjuvant therapy for postmenopausal patients with breast<br />

cancer: FEMZONE trial results<br />

Fasching PA, Jud SM, Hauschild M, Kümmel S, Schütte M, Warm M,<br />

Hanf V, Muth M, Baier M, Schulz-Wendtland R, Beckmann MW, Lux<br />

MP. Erlangen University Hospital, Friedrich Alexander University of<br />

Erlangen Nuremberg, Erlangen, Germany; Frauenklinik Rheinfelden,<br />

Rheinfelden, Germany; Frauenklinik, Klinikum Essen-Mitte, Essen,<br />

Germany; Essen University Hospital, Essen, Germany; Cologne<br />

University Hospital, Cologne, Germany; Kliniken der Stadt Köln<br />

Holweide, Cologne, Germany; Klinikum Fuerth, Fuerth, Germany;<br />

Novartis Pharma GmbH, Nuremberg, Germany; Erlangen University<br />

Hospital, Friedrich Alexander University of Erlangen–Nuremberg,<br />

Erlangen, Germany.<br />

PD07-03 Increased pathologic complete response rate after a long term<br />

neoadjuvant letrozole treatment in postmenopausal estrogen<br />

and/or progesterone receptor-positive breast cancer<br />

Allevi G, Strina C, Andreis D, Zanoni V, Bazzola L, Bonardi S, Foroni C,<br />

Milani M, Cappelletti MR, Generali D, Berruti A, Bottini A. A.O. Istituti<br />

Ospitalieri di Cremona, Cremona, Italy; A.O.U. <strong>San</strong> Luigi Gonzaga di<br />

Orbassano, Orbassano, TO, Italy.<br />

PD07-04<br />

PD07-05<br />

PD07-06<br />

PD07-07<br />

PD07-08<br />

A randomized phase II neoadjuvant trial evaluating<br />

anastrozole and fulvestrant efficiency for post-menopausal<br />

ER-positive, HER2-negative breast cancer patients: first results<br />

of the UNICANCER CARMINA 02 French trial<br />

Lerebours F, Bourgier C, Alran S, Mouret-Reynier M-A, Venat Bouvet<br />

L, Kerbrat P, Salmon R, Mijonnet S, Becette V, Trassard M, Spyratos F,<br />

Lebas N, Martin A-L, Lemonnier J, Mouret-Fourme E. Institut Curie,<br />

Saint Cloud, France; Institut Gustave Roussy, Villejuif, France; Institut<br />

Curie, Paris, France; Centre Jean Perrin, Clermont-Ferrand, France;<br />

CHU, Limoges, France; Centre Eugene Marquis, Rennes, France; R&D<br />

Unicancer, Paris, France.<br />

A randomized controlled trial comparing zoledronic acid plus<br />

chemotherapy with chemotherapy alone as a neoadjuvant<br />

treatment in patients with HER2-negative primary breast<br />

cancer<br />

Hasegawa Y, Kohno N, Horiguchi J, Miura D, Ishikawa T, Hayashi<br />

M, Takao S, Kim SJ, Tanino H, Miyashita M, Konishi M, Shigeoka<br />

Y, Yamagami K, Akazawa K. Hirosaki Municipal Hospital, Hirosaki,<br />

Aomori, Japan; Tokyo Medical University Hospital, Tokyo, Japan;<br />

Gunma University Hospital, Maebashi, Gunma, Japan; Toranomon<br />

Hospital, Tokyo, Japan; Yokohama City University Medical Center,<br />

Yokohama, Kanagawa, Japan; Tokyo Medical University Hachioji<br />

Medical Center, Hachioji, Tokyo, Japan; Hyogo <strong>Cancer</strong> Center, Akashi,<br />

Hyogo, Japan; Osaka University Hospital, Suita, Osaka, Japan; Naga<br />

Municipal Hospital, Kinokawa, Wakayama, Japan; Konan Hospital,<br />

Kobe, Hyogo, Japan; Hyogo Prefectural Nishinomiya Hospital,<br />

Nishinomiya, Hyogo, Japan; Yodogawa Christian Hospital, Osaka,<br />

Japan; Shinko Hospital, Kobe, Hyogo, Japan; Niigata University<br />

Medical and Dental Hospital, Niigata, Japan.<br />

NEO-ZOTAC: Toxicity data of a phase III randomized trial with<br />

NEOadjuvant chemotherapy (TAC) with or without ZOledronic<br />

acid (ZA) for patients with HER2-negative large resectable or<br />

locally advanced breast cancer (BC)<br />

van de Ven S, Liefers G-j, Putter H, van Warmerdam LJ, Kessels LW,<br />

Dercksen W, Pepels MJ, Maartense E, van Laarhoven HWM, Vriens B,<br />

Smit VTHBM, Wasser MNJM, Meershoek-Klein Kranenbarg EM, van<br />

Leeuwen-Stok E, van de Velde CJH, Nortier JWR, Kroep JR. Leiden<br />

University Medical Center (LUMC), Leiden, Netherlands; Catharina<br />

Hospital, Eindhoven, Netherlands; Deventer Hospital, Deventer,<br />

Netherlands.<br />

Prediction of antiproliferative response to lapatinib by HER3 in<br />

an exploratory analysis of HER2-non-amplified (HER2-) breast<br />

cancer in the MAPLE presurgical study (CRUK E/06/039)<br />

Dowsett M, Leary A, Evans A, A’Hern R, Bliss J, Sahoo R, Detre S,<br />

Hills M, Haynes B, Harper-Wynne C, Bundred N, Coombes G, Smith<br />

IE, Johnston S. Royal Marsden Hospital, London, United Kingdom;<br />

Institut Gustave Roussy, Paris, France; Poole Hospital, Poole, Dorset,<br />

United Kingdom; Institute of <strong>Cancer</strong> Research, London, United<br />

Kingdom; Kent Oncology Centre, Maidstone, Kent, United Kingdom;<br />

University Hospital of South Manchester NHS Trust, Manchester,<br />

United Kingdom.<br />

Zoledronic acid specifically inhibits development of bone<br />

metastases in the post-menopausal setting – evidence from<br />

an in vivo breast cancer model<br />

Holen I, Wang N, Reeves KJ, Fowles AM, Croucher PI, Eaton CL,<br />

Ottewell PD. University of Sheffield, United Kingdom.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012 44s <strong>Cancer</strong> Research


December 4–8, 2012<br />

Program Schedule<br />

PD07-09 Zoledronate versus ibandronate comparative evaluation<br />

(ZICE) trial - first results of a UK NCRI 1,405 patient phase<br />

III trial comparing oral ibandronate versus intravenous<br />

zoledronate in the treatment of breast cancer patients with<br />

bone metastases<br />

Barrett-Lee PJ, Casbard A, Abraham J, Grieve R, Wheatley D,<br />

Simmons P, Coleman R, Hood K, Griffiths G, Murray N. Velindre<br />

NHS Trust, Cardiff, Wales, United Kingdom; Cardiff University School<br />

of Medicine, Cardiff, Wales, United Kingdom; University Hospital,<br />

Coventry, England, United Kingdom; Royal Cornwall Hospital,<br />

Truro, England, United Kingdom; University Hospital, Southampton,<br />

England, United Kingdom; Weston Park Hospital, Sheffield, England,<br />

United Kingdom; Royal Adelaide Hospital, Adelaid, South Australia,<br />

Australia.<br />

7:00 am–9:00 am<br />

Poster Discussion 8: Disparities<br />

Ballroom B<br />

Viewing 7:00 am<br />

Discussion 7:45 am<br />

Patricia Ganz, MD, Chair<br />

UCLA Jonsson Comprehensive <strong>Cancer</strong> Center<br />

Los Angeles, CA<br />

Dawn Hershman, MD, Discussant<br />

Columbia University Medical Center<br />

New York, NY<br />

and<br />

Amelie Ramirez, DRPH, MPH, DO, Discussant<br />

UT Health Science Center<br />

<strong>San</strong> <strong>Antonio</strong>, TX<br />

PD08-01 Utilization of Oncotype DX in an inner-city population:<br />

Race or place?<br />

Guth AA, Fineberg S, Fei K, Franco R, Bickell N. NYU School of<br />

Medicine, New York, NY; Montefiore Medical Center, Bronx, NY;<br />

Mount Sinai School of Medicine, New York, NY.<br />

PD08-02 Disparities in the utilization of reconstruction after<br />

mastectomy: The California Teachers Study<br />

Kruper L, Xu XX, Bernstein L, Henderson K. City of Hope <strong>Cancer</strong><br />

Center, Duarte, CA.<br />

PD08-03 Barriers to breast reconstructive surgery in an underprivileged<br />

community: Does income really matter?<br />

Zelek LH, Festa A, Barbeau E, Morere J-F. Assistance Publique<br />

Hôpitaux de Paris, CHU Avicenne, Bobigny, France; Oncologie 93,<br />

Bobigny, France.<br />

PD08-04 Factors which affect surgical management in an underinsured,<br />

county hospital population<br />

Komenaka IK, Olsen L, Klemens AE, Hsu C-H, Nodora J, Martinez ME,<br />

Thompson PA, Bouton M. Maricopa Medical Center, Phoenix, AZ;<br />

University of Arizona, Tucson, AZ; Moores <strong>Cancer</strong> Center, University<br />

of California, <strong>San</strong> Diego, CA.<br />

PD08-05 Spanning the continuum to assess, serve and navigate Latinas<br />

with breast cancer: A tale of six projects<br />

Ramirez AG, Holden AE, Gallion K, <strong>San</strong>Miguel SA, Munoz E, Penedo<br />

FJ, Perez-Stable EJ, Talavera GG, Carrillo JE, Fernandez ME. University<br />

of Texas Health Science Center at <strong>San</strong> <strong>Antonio</strong>, TX; Redes en Accion:<br />

The National Latino <strong>Cancer</strong> Research Network, The University of<br />

Texas Health Science Center at <strong>San</strong> <strong>Antonio</strong>, <strong>San</strong> <strong>Antonio</strong>, TX.<br />

PD08-06 Significant clinical impact of recurrent BRCA1<br />

and BRCA2 (BRCA) mutations in Mexico<br />

Villarreal-Garza C, Herrera LA, Herzog J, Port D, Mohar A, Perez-<br />

Plasencia C, Clague J, Alvarez RMa, <strong>San</strong>tibanez M, Blazer KR,<br />

Weitzel JN. Instituto Nacional de <strong>Cancer</strong>ologia, Mexico City, Mexico<br />

DF, Mexico; Instituto Nacional de <strong>Cancer</strong>ologia – Instituto de<br />

Investigaciones Biomedicas, UNAM, Mexico City, Mexico DF, Mexico;<br />

City of Hope, Duarte, CA.<br />

7:00 am–9:00 am<br />

POSTER SESSION 4 & Continental Breakfast<br />

Exhibit Halls A–B<br />

Detection/Diagnosis: <strong>Breast</strong> Imaging - MRI<br />

P4-01-01 Integrating dynamic magnetic resonance imaging and gene<br />

expression profiling reveals novel therapeutic targets in<br />

locally advanced breast cancer<br />

Hughes NP, Mehta S, Winchester L, Han C, Buffa FM, Adams RF,<br />

Harris AL. Stanford University, Stanford, CA; University of Oxford,<br />

United Kingdom; Churchill Hospital, Oxford, United Kingdom.<br />

P4-01-02 Association of DCE-MRI texture features with molecular<br />

phenotypes and neoadjuvant therapy response in<br />

breast cancer<br />

Banerjee N, Maity S, Varadan V, Janevski A, Kamalakaran S, Sikov W,<br />

Abu-Khalaf M, Bossuyt V, Lannin D, Harris L, Cornfeld D, Dimitrova N.<br />

Philips Research North America; Yale Comprehensive <strong>Cancer</strong> Center;<br />

Warren Alpert Medical School of Brown University; Yale-New Haven<br />

Hospital; Yale <strong>Breast</strong> <strong>Cancer</strong> Program; Seidman <strong>Cancer</strong> Center.<br />

P4-01-03 Quantitative DCE-MRI to predict the response of primary<br />

breast cancer to neoadjuvant therapy<br />

Li X, Arlinghaus LR, Chakravarthy AB, Abramson RG, Abramson VG,<br />

Farley J, Ayers GD, Mayer IA, Kelley MC, Meszoely IM, Means-Powell J,<br />

Grau AM, <strong>San</strong>ders ME, Yankeelov TE. Vanderbilt University.<br />

P4-01-04 MRI enhancement in stromal tissue surrounding breast<br />

tumors: Association with RFS following neoadjuvant<br />

chemotherapy<br />

Jones EF, Sinha SP, Newitt D, Klifa C, Kornak J, Park CC, Hylton NM.<br />

University of California, <strong>San</strong> Francisco, CA.<br />

P4-01-05 Utility of Preoperative Routine MRI and PET/CT in <strong>Breast</strong><br />

<strong>Cancer</strong> Staging vs. Surgical Staging<br />

Higaki K, Kochi M, Ito M, Otani S. Hiroshima City Hospital,<br />

Hiroshima, Japan.<br />

P4-01-06 Evaluation of 3D T2-weighted <strong>Breast</strong> MRI<br />

Moran CJ, Hargreaves BA, Saranathan M, Daniel BL. Stanford<br />

University, Stanford, CA.<br />

P4-01-07 The Relationship of <strong>Breast</strong> Density in Mammography and<br />

Magnetic Resonance (MR) Imaging in a High Risk Population<br />

Chun J, Refinetti AP, Schnabel F, Leite AP, Price A, Billig J, Schwartz S,<br />

Moy L. NYU Langone Medical Center, New York, NY.<br />

P4-01-08 Effect of bilateral salpingo-oophorectomy on breast MRI<br />

fibroglandular volume and background parenchymal<br />

enhancement for BRCA 1/2 mutation carriers<br />

DeLeo, III MJ, Domchek S, Kontos D, Conant E, Weinstein S. Hospital<br />

of the University of Pennsylvania, Philadelphia, PA.<br />

P4-01-09 Screening breast magnetic resonance imaging (MRI) in earlystage<br />

breast cancer survivors<br />

Dowton AA, Meyer ME, Ruddy KJ, Yeh ED, Partridge AH. Dana-Farber<br />

<strong>Cancer</strong> Institute, Boston, MA; Brigham & Women’s Hospital,<br />

Boston, MA.<br />

P4-01-10 High-resolution diffusion weighted imaging for the separation<br />

of benign from malignant BI-RADS 4/5 lesions found on breast<br />

MRI at 3 Tesla<br />

Wisner DJ, Rogers N, Deshpande VS, Newitt DN, Laub GA, Porter<br />

DA, Joe BN, Hylton NM. University of California, <strong>San</strong> Francisco, CA;<br />

Siemens Medical Solutions, USA, Inc, <strong>San</strong> Francisco, CA; Siemens<br />

Medical Solutions, Erlangen, Germany.<br />

P4-01-11 Clinical Findings and Outcomes from MRI Staging of <strong>Breast</strong><br />

<strong>Cancer</strong> in a Diverse Population<br />

Raghavendra A, Ji L, Ricker C, Tang S, Church TD, Larsen L, Sheth<br />

P, Sposto R, Sener S, Tripathy D. University of Southern California<br />

Keck School of Medicine, Los Angeles, CA; Los Angeles County and<br />

University of Southern California (LAC+USC) Healthcare Network,<br />

Los Angeles, CA; USC Norris Comprehensive <strong>Cancer</strong> Center,<br />

Los Angeles, CA.<br />

www.aacrjournals.org<br />

45s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P4-01-12 Compliance with Recommended Follow-Up after MRI-<br />

Guided Core Needle Biopsy of Suspicious <strong>Breast</strong> Lesions: A<br />

Retrospective Study<br />

Thompson MO, Lipson JA, Daniel BL, Harrigal CL, Mullarkey PJ, Ikeda<br />

DM. Stanford University School of Medicine, Stanford, CA.<br />

P4-01-13 Practice patterns of MRI utilization for breast cancer treatment<br />

within the University of California system as part of the<br />

Athena initiative<br />

Tokin CA, Ojeda H, Mayadev JS, Hylton NM, Fowble BL, Rugo HS,<br />

Hwang S, Hurvitz S, Wells C, Blair SL. University of California, <strong>San</strong><br />

Diego; University of California, Davis; University of California, <strong>San</strong><br />

Francisco; Duke University; University of California, Los Angeles.<br />

P4-01-14 Can MRI predict the response to neoadjuvant chemotherapy<br />

in breast cancer accurately?<br />

Fatayer H, Kim B, Dall B, Sharma N, Manuel D, Shabaan A, Perren<br />

T, Velikova G, Horgan K, Lansdown M. Leeds General Infirmary; St<br />

James‘s University Hospital.<br />

P4-01-15 Impact of Preoperative MRI on the Surgical Treatment of<br />

<strong>Breast</strong> <strong>Cancer</strong>: A SEER-Medicare Analysis<br />

Thorsen CM, Weiss JE, Kerlikowske K, Ozanne EM, Buist DS, Hubbard<br />

RA, Tosteson AN, Henderson LM, Virnig BA, Goodrich ME, Onega<br />

TL. University of California, <strong>San</strong> Francisco, CA; Dartmouth Medical<br />

School, Lebanon, NH; University of Washington, Seattle, WA;<br />

University of North Carolina, Chapel Hill, NC; University of Minnesota,<br />

Minneapolis, MN.<br />

P4-01-16 Withdrawn<br />

P4-01-17 Ductolobular breast carcinoma and the role of preoperative<br />

magnetic resonance imaging<br />

Postma EL, El Sharouni MA, Verkooijen HM, Witkamp AJ, van den<br />

Bosch MA, van Hillegersberg R, van Diest PJ. UMC Utrecht, Utrecht,<br />

Utrecht, Netherlands.<br />

Detection/Diagnosis: Molecular, Functional, and Novel Imaging<br />

P4-02-01 18F-FDG PET/CT for the assessment of locoregional lymph<br />

node involvement and radiotherapy indication in stage II-III<br />

breast cancer treated with neoadjuvant chemotherapy<br />

Koolen BB, Valdés Olmos RA, Vogel WV, Vrancken Peeters M-JTFD,<br />

Rodenhuis S, Rutgers EJT, Elkhuizen PHM. Netherlands <strong>Cancer</strong><br />

Institute - Antoni van Leeuwenhoek Hospital, Amsterdam,<br />

Netherlands.<br />

P4-02-02 Comparison of 18F-FDG PET-CT and 11C-MET PET-CT for<br />

assessment of response to neoadjuvant chemotherapy in<br />

locally advanced breast carcinoma<br />

Dinesh A, Ramteke VK, Chander J, Tripathi M, Mahajan S. Maulana<br />

Azad Medical College, New Delhi, Delhi, India; Institute of Nuclear<br />

Medicine & Allied Sciences, New Delhi, Delhi, India.<br />

P4-02-03 FDG PET/CT for early monitoring of response to neoadjuvant<br />

chemotherapy in breast cancer patients<br />

Andrade W, Soares F, Lima E, Maciel MdS, Toledo C, Iyeyasu H, Cruz<br />

M, Fanelli M. A. C. Camargo <strong>Cancer</strong> Hospital, São Paulo, SP, Brazil.<br />

P4-02-04 Tissue oxyhemoglobin dynamics measured with functional<br />

optical imaging immediately after starting chemotherapy<br />

correlates with markers of cellular proliferation and<br />

inflammation in a rat breast tumor model<br />

Ueda S, Roblyer D, Cerussi A, Saeki T, Tromberg B. Beckman Laser<br />

Institute, University of California, Irvine, Irvine, CA; Saitama Medical<br />

University, Hidaka, Saitama, Japan.<br />

P4-02-05 A novel 64Cu-liposomal PET agent (MM-DX-929) predicts<br />

response to liposomal chemotherapeutics in preclinical breast<br />

cancer models<br />

Lee H, Zheng J, Gaddy D, Kirpotin D, Dunne M, Drummond D, Allen<br />

C, Jaffray D, Hendriks B, Wickham T. Merrimack Pharmaceuticals,<br />

Boston, MA; Princess Margaret Hospital, University Health Network,<br />

Toronto, ON, Canada; Leslie Dan Faculty of Pharmacy, University of<br />

Toronto, ON, Canada.<br />

P4-02-06 Molecular imaging with trastuzumab and pertuzumab<br />

of HER2-positive breast cancer in mice: a step towards<br />

personalized medicine<br />

Collin B, Oudot A, Vrigneaud J-M, Moreau M, Raguin O, Duchamp<br />

O, Tizon X, Denat F, Varoqueaux N, Brunotte F, Fumoleau P.<br />

Centre Georges François Leclerc, Dijon, France; Institut de Chimie<br />

Moléculaire de l’université de Bourgogne, Dijon, France; Oncodesign,<br />

Dijon, France; Roche France, Boulogne Billancourt, France.<br />

P4-02-07 Early Optical Tomography Changes Predict <strong>Breast</strong> <strong>Cancer</strong><br />

Response to Neoadjuvant Chemotherapy<br />

Lim EA, Gunther JE, Flexman M, Kim HK, Hibshoosh H, Kalinsky K,<br />

Crew K, Maurer M, Taback B, Feldman S, Brown M, Refice S, Alvarez-<br />

Cid M, Hielscher A, Hershman DL. Columbia University Medical<br />

Center, New York, NY; Columbia University, New York, NY; Herbert<br />

Irving Comprehensive <strong>Cancer</strong> Center, New York, NY; Mailman School<br />

of Public Health, New York, NY.<br />

P4-02-08 Quantitative Characterization of 3D Vasculature Spatial<br />

Patterns Within Tumor Microenvironment of <strong>Breast</strong> <strong>Cancer</strong><br />

Stem Cells<br />

Zhan M, Li F, Zhu Y, Ma J, Landua J, Wei W, Vadakkan T, Zhang M,<br />

Dickinson M, Lewis M, Rosen J, Wong S. NCI Center for Modeling<br />

<strong>Cancer</strong> Development, The Methodist Hospital, Houston, TX; Baylor<br />

College of Medicine, Houston, TX.<br />

P4-02-09 Molecular <strong>Breast</strong> Imaging: the sensitivity of breastspecific<br />

gamma imaging (BSGI) as a diagnostic adjunct to<br />

mammography and ultrasound in a triple assessment protocol<br />

Weigert JM, Kieper DA, Stern LH, Böhm-Vélez M. Mandell and Blau<br />

MDs PC, New Britain, CT; Hampton University, Hampton, VA; Thomas<br />

Jefferson University, Methodist Division, Philadelphia, PA; Weinstein<br />

Imaging Associates, Pittsburgh, PA.<br />

P4-02-10 Molecular <strong>Breast</strong> Imaging: A multicenter clinical registry to<br />

compare breast-specific gamma imaging (BSGI) and breast<br />

MRI in the detection of breast carcinoma<br />

Weigert JM, Kieper DA, Stern LH, Böhm-Vélez M. Hampton<br />

University, Hampton, VA; Thomas Jefferson University, Methodist<br />

Division, Philadelphia, PA; Mandell and Blau MDs PC, New Britain, CT;<br />

Weinstein Imaging Associates, Pittsburgh, PA.<br />

Detection/Diagnosis: <strong>Breast</strong> Imaging - Other Methods<br />

P4-03-01 Distance of breast cancer from the skin influence axillary<br />

nodal metastasis<br />

Kim EJ, Chae BJ, Song BJ, Kwak HY, Chang EY, Kim SH, Jung SS. Seoul<br />

St. Mary’s Hospital, The Catholic University of Korea, Seoul, Republic<br />

of Korea.<br />

P4-03-02 Automatic BI-RADS Diagnosis of <strong>Breast</strong> Lesions by<br />

CAD(computer-aid diagnosis)<br />

Kuo W-H, Chuang S-C, Yang S-H, Chen C-N, Chen A, Chang K-J.<br />

National Taiwan University Hospital and National Taiwan University<br />

College of Medicine, Taipei, Taiwan; Graduate Institute of Industrial<br />

Engineering, National Taiwan University, Taipei, Taiwan.<br />

P4-03-03 Feasibility study of a new volume navigation system-guided<br />

breast biopsy method for incidental enhancing lesions<br />

detected by breast contrast-enhanced magnetic resonance<br />

imaging<br />

Takahashi M, Jinno H, Hayashida T, Nemoto M, Tanimoto A, Kitagawa<br />

Y. Keio University School of Medicine, Tokyo, Japan.<br />

P4-03-04 The potential use of Optical Coherence Tomography for<br />

intraoperative breast tumour margin width estimation<br />

Wilson BC, Akens MK, Niu CJ. University Health Network, Ontario<br />

<strong>Cancer</strong> Institute, Toronto, ON, Canada; Tornado Medical System,<br />

Toronto, ON, Canada.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012 46s <strong>Cancer</strong> Research


December 4–8, 2012<br />

Program Schedule<br />

P4-03-05 The Clinical Trial of New Optical Mammography<br />

Ogura H, Yoshimoto K, Nasu H, Hosokawa Y, Matsunuma R, Ide<br />

Y, Yamaki E, Yamashita D, Suzuki T, Oda M, Ueda Y, Yamashita Y,<br />

Sakahara H. Hamamatsu University School of Medicine, Hamamatsu,<br />

Shizuoka, Japan; Hamamatsu Photonics K.K., Hamamatsu,<br />

Shizuoka, Japan.<br />

P4-03-06 Non-invasive classification of microcalcifications by the use of<br />

X-ray phase contrast mammography as a novel tool in breast<br />

diagnostics<br />

Hauser N, Wang Z, Kubik-Huch RA, Singer G, Trippel M, Roessl E, Hohl<br />

MK, Stampanoni M. Interdisciplinary <strong>Breast</strong> Center Kantonsspital<br />

Baden, Baden, Switzerland; Paul Scherrer Institut, Villigen, Switzerland;<br />

Kantonsspital Baden, Baden, Switzerland; Philips Research<br />

Laboratories, Hamburg, Germany.<br />

P4-03-07 Angiogenic effect of bevacizumab and paclitaxel in<br />

metastatic breast cancer: evaluation by contrast-enhanced<br />

ultrasonography using Sonazoid®<br />

Ito T, Mizuno H, Iiboshi Y, Yamamura N, Fujii H, Hitora T, Fujii R,<br />

Ohashi T, Nakagawa T, Izukura M. Rinku General Medical Center,<br />

Izumisano, Osaka, Japan.<br />

P4-03-08 A new real-time image fusion technique, a coordinated<br />

sonography and MRI using magnetic position tracking system,<br />

improves the sonographic identification of enhancing lesions<br />

in breast MRI<br />

Nakano S, Fujii K, Yoshida M, Kousaka J, Mouri Y, Fukutomi T,<br />

Ishiguchi T. Aichi Medical University, Nakakute, Aichi, Japan.<br />

P4-03-09 A comparison of MRI, PET-CT, and ultrasonography for<br />

evaluation of tumor response to neoadjuvant chemo-therapy<br />

in patients with locally advanced breast cancer<br />

Boothe DL, Stessin A, Nagar H, Hayes MK. Weill Cornell Medical<br />

College, New York, NY.<br />

P4-03-10 Sonographic-pathological correlation in contrast-enhanced<br />

ultrasonography in the diagnosis of breast cancers<br />

Kato K, Miyamoto Y, Kamio M, Nogi H, Imawari Y, Mimoto R, Toriumi<br />

Y, Nakata N, Takeyama H, Uchida K. The Jikei University School of<br />

Medicine, Tokyo, Japan.<br />

P4-03-11 SUVmax of FDG-PET/CT Is Associated with Chemotherapy<br />

Response Assay Test Results and Prognostic Factors in <strong>Breast</strong><br />

<strong>Cancer</strong> Patients<br />

Lee A, Chang J, Bang B, Lee J, Han D, Min S, Yun C, Kim BS, Lim W,<br />

Paik N, Moon B-I. Ewha Womans University Hospital, Seoul, Republic<br />

of Korea.<br />

P4-03-12 Diagnostic value and clinical significance of integrin αvβ3 in<br />

breast mass<br />

Xu Z, Song Y, Sun L, Sun G, Ma Q, Gao S, Wang K. China-Japan<br />

Union Hospital of JiLin University, JiLin Province <strong>Breast</strong> Diseases<br />

Institute, Changchun, Jilin, China; China-Japan Union Hospital of JiLin<br />

University, Changchun, Jilin, China.<br />

Tumor Cell and Molecular Biology: Immunology and Preclinical<br />

Immunotherapy<br />

P4-04-01 Combination of intratumoral CpG with systemic anti-OX40 and<br />

anti-CTLA4 mAbs eradicates established triple negative breast<br />

tumors in mice<br />

Li J, Tandon V, Levy R, Esserman L, Campbell M. University of<br />

California, <strong>San</strong> Francisco, CA; Stanford University, Stanford, CA.<br />

P4-04-02 Tumor-initiated peripheral myeloid cell expansion is reversed<br />

by radiation therapy<br />

Gough MJ, Savage T, Bahjat KS, Redmond W, Bambina S, Kasiewicz M,<br />

Cottam B, Newell P, Crittenden MR. Earle A. Chiles Research Institute,<br />

Providence <strong>Cancer</strong> Center, Portland, OR; Providence <strong>Cancer</strong> Center,<br />

Portland, OR; The Oregon Clinic, Portland, OR.<br />

P4-04-03 Monocytic immature myeloid cells permit tumor immune<br />

evasion during postpartum involution<br />

Schedin P, Martinson H, Callihan E, Jindal S, Borges V. University of<br />

Colorado, Aurora, CO.<br />

P4-04-04 Immunohistochemical analysis of <strong>Cancer</strong> Testis antigens and<br />

Topoisomerase 2-alpha expression in triple negative breast<br />

carcinomas: a retrospective study<br />

Juretic A, Mrklic I, Spagnoli GC, Pogorelic Z, Tomic S. Zagreb<br />

University Hospital Centre, Zagreb, Croatia; Split University Hospital<br />

Centre, Split, Croatia; University of Basel, Switzerland.<br />

P4-04-05 Listeria monocytogenes-based bivalent Lm-LLO immunotherapy<br />

for the treatment of HER2/neu positive and triple negative<br />

breast cancer and its impact on immunosuppression<br />

Wallecha A, Ramos K, Malinina I, Singh R. Advaxis Inc, Princeton, NJ.<br />

P4-04-06 Young women’s breast cancer is characterized by increased<br />

immune suppression through circulating myeloid derived<br />

supressor cells<br />

Borges VF, Ramirez O, Borakove M, Manthey E, Diamond JR, Elias AD,<br />

Finlayson C, Kounalakis N, Jordan K. University of Colorado Denver,<br />

Aurora, CO; University of Colorado <strong>Cancer</strong> Center, Aurora, CO.<br />

P4-04-07 Tartrate-resistant Acid Phosphatase (TRAcP)-Expressed Tumor-<br />

Associated Macrophages Promote <strong>Breast</strong> <strong>Cancer</strong> Progression<br />

Dai M-S, Wu C-C, Chang P-Y, Ho C-L, Hsieh Y-F, Lo K-Y, Kao W-Y,<br />

Chao T-Y, Yu J-C. Tri-Service General Hospital, Taipei, Taiwan; Taipei<br />

Medical University, Taipei, Taiwan.<br />

P4-04-08 A Potential non-viral vector to transfect dendritic cell and<br />

thereby MHC-Class I antigen presentation might be a potential<br />

use in carcinoma<br />

Shaheen S, Akita H, Souichirou I, Miura N, Harashima H. Hokkaido<br />

University, Sapporo, Hokkaido, Japan.<br />

Tumor Cell and Molecular Biology: Gene Therapy<br />

P4-05-01 Oncolytic Herpes Simplex Virus Vector G47Δ Effectively Targets<br />

<strong>Breast</strong> <strong>Cancer</strong> Stem Cells<br />

Liu R, Zeng W, Hu P, Wu J, Li J, Wang J, Lei L. The Third Affiliated<br />

Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China;<br />

The Sichuan Province <strong>Cancer</strong> Hospital, Chengdu, Sichuan, China.<br />

Tumor Cell and Molecular Biology: Novel/Emerging Therapeutic Targets<br />

P4-06-01 JAK2/STAT3 activity in inflammatory breast cancer supports<br />

the investigation of JAK2 therapeutic targeting<br />

Overmoyer BA, Almendro V, Shu S, Peluffo G, Park SY, Nakhlis F,<br />

Bellon JR, Yeh ED, Jacene HA, Hirshfield-Bartek J, Polyak K. Dana<br />

Farber <strong>Cancer</strong> Institute, Harvard Medical School, Boston, MA; Seoul<br />

National University, Seoul, Korea.<br />

P4-06-02 Identification of novel G-protein coupled receptor targets in<br />

HER2-positive breast cancer<br />

Yadav P, Bhat RR, Chayanam S, Christiny PI, Nanda S, Hu H, Creighton<br />

C, Osborne CK, Schiff R, Trivedi MV. University of Houston, TX; Lester<br />

& Sue Smith <strong>Breast</strong> Center, Dan Duncan <strong>Cancer</strong> Center, Houston, TX;<br />

Baylor College of Medicine, Houston, TX.<br />

P4-06-03 Zinc Finger Nuclease Genome Engineering Reveals Multiple<br />

Functions of Secretory Leukocyte Peptidase Inhibitor in<br />

Regulating Pleuripotency of <strong>Cancer</strong> Stem Cells in Inflammatory<br />

<strong>Breast</strong> <strong>Cancer</strong><br />

Robertson FM, Hibbs S, Boley KM, Chu K, Ye Z, Wright MC, Liu H, Luo<br />

AZ, Cristofanilli M, Wemhoff G. The University of Texas MD Anderson<br />

<strong>Cancer</strong> Center, Houston, TX; Sigma-Aldrich, St. Louis, MO; Fox Chase<br />

<strong>Cancer</strong> Center, Philadelphia, PA.<br />

P4-06-04 Gamma-secretase inhibitors suppress the activation of NFκB<br />

and the expression of TNFα, IL-6 and IL-8 in triple negative<br />

breast cancer cells<br />

Gu J-W, King J, Makey KL, Chinchar E, Gibson J, Miele L. University of<br />

Mississippi Medical Center, Jackson, MS.<br />

www.aacrjournals.org<br />

47s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P4-06-05 NF-κB Upregulates β1-integrin via Increased Transcriptional<br />

Activity in Three-dimensional Culture: a Mechanism by which<br />

Malignant <strong>Breast</strong> Cells Acquire Radioresistance<br />

Ahmed KM, Zhang H, Park CC. Lawrence Berkeley National<br />

Laboratory, Berkeley, CA; University of California, <strong>San</strong> Francisco, CA.<br />

P4-06-06 RNAi screen of the breast cancer genome identifies KIF14 and<br />

TLN1 as genes that modulate chemosensitivity in breast cancer<br />

Singel SM, Cornelius C, Batten K, Fasciani G, Wright WE, Lum L, Shay<br />

JW. University of Texas Southwestern, Dallas, TX.<br />

P4-06-07 Moved to Poster Session 6, Saturday, December 8<br />

7:00 AM - 8:30 AM<br />

P4-06-08 Clinical utility of functional analysis of FGFR kinase family for<br />

selecting patients who may benefit from FGFR inhibitors<br />

Hoe N, Zhou J, Kuy C, Jin K, Srikrishnan R, Soundararajan A, Ma Y, Liu<br />

X, Singh S. Prometheus Labs, <strong>San</strong> Diego, CA.<br />

P4-06-09 Cell surface receptor CDCP1 as a potential marker of triple<br />

negative breast cancers progression<br />

Campiglio M, Sasso M, Bianchi F, De Cecco L, Turdo F, Triulzi T, Orlandi<br />

R, Morelli D, Aiello P, Ghirelli C, Agresti R, Tagliabue E. Fondazione<br />

IRCCS Istituto Nazionale dei Tumori, Milan, Italy; Fondazione IRCCS<br />

Istituto Nazionale dei Tumori.<br />

P4-06-10 Epigenetic silencing of glutamine synthetase (Glul) defines<br />

glutamine depletion therapy<br />

Cavicchioli F, Shia A, O’Leary K, Haley V, Crook TR, Thompson AM,<br />

Lackner M, Lo Nigro C, Schmid P. Brighton and Sussex Medical<br />

School, University of Sussex, Brighton, United Kingdom; Ninewells<br />

Hospital, University of Dundee, United Kingdom; Genentech, Inc., <strong>San</strong><br />

Francisco; S. Croce General Hospital, Cuneo, Italy.<br />

P4-06-11 Expression of genes spanning a breast cancer susceptibility<br />

locus on 6q25.1 is modulated by epigenetic modification<br />

Dunbier AK, White J, Van Huffel S. University of Otago, Dunedin,<br />

Otago, New Zealand.<br />

P4-06-12 Monoclonal antibodies against nicastrin for the treatment of<br />

breast cancer: in vitro and in vivo characterisation and function<br />

Filipovic A, Lombardo Y, Deonarain M, Giamas G, Cordingley H,<br />

Tralau-Stewart C, Coombes RC. Imperial College London, United<br />

Kingdom.<br />

P4-06-13 Effects of Statin on triple-negative breast cancer (TNBC) with<br />

Ets-1 overexpression<br />

Lee S, Jung HH, Park YH, Ahn JS, Im Y-H. Samsung Medical Center.<br />

P4-06-14 CD146-suppresses breast tumor invasion via a novel<br />

transcription target TIMPv<br />

Shanmuganathan S, AbdElmageed Z, Fernando A, Gaur R, Ramkumar<br />

A, Bhat S, Gupta I, Muzumdar S, Hakkim L., Ouhtit A. Sultan Qaboos<br />

University, Al-Khod, Seeb, Oman.<br />

P4-06-15 Mifepristone modifies the tumor microenvironment increasing<br />

the therapeutic efficiency of low doses of Doxorubicin<br />

liposomes or paclitaxel-albumin nanoparticles in a murine<br />

model of breast cancer<br />

Sequeira GR, Vanzulli SI, Lamb CA, Rojas PA, Lanari C. Instituto de<br />

Biología y Medicina Experimental, Ciudad Autónoma de Buenos Aires,<br />

Argentina; Academia Nacional de Medicina, Ciudad Autónoma de<br />

Buenos Aires, Argentina.<br />

P4-06-16 TGF-β2, A Novel Target of CD44-Promoted <strong>Breast</strong> <strong>Cancer</strong><br />

Invasion<br />

Gupta I, Madani S, Abdraboh M, Al Riyami H, Muzumdar S,<br />

AbdElmageed Z, Shanmuganathan S, Bhat S, Ramkumar A, Hakkim<br />

L, Ouhtit A. College of Medicine and Health Sciences, Sultan Qaboos<br />

University, Muscat, Al Khuwd, Oman.<br />

P4-06-17<br />

P4-06-18<br />

P4-06-19<br />

Antitumor activity of the novel mithramycin analog EC8042 in<br />

triple negative breast cancer<br />

Pandiella A, Montero JC, Cuenca D, Re-Louhau F, Nuñez LE, Moris<br />

F, Ocana A. Salamanca <strong>Cancer</strong> Research Center, Salamanca, Spain;<br />

Translational Research Unit, Albacete University Hospital, Albacete,<br />

Spain; Entrechem SL, Oviedo, Spain.<br />

Targeting basal-like breast cancer through downstream<br />

effectors of oncogene cooperation<br />

McMurray HR, Walters EA, Heyer DM, Candelaria PV, Wang S, Grose<br />

VA, Llop JR, Zhang Y. University of Rochester Medical Center,<br />

Rochester, NY.<br />

Astemizole and calcitriol: A novel targeted therapeutic strategy<br />

for breast cancer<br />

García J, García R, Villanueva O, <strong>San</strong>tos N, Barrera D, Avila E, Ordaz D,<br />

Halhali A, Camacho J, Larrea F, Díaz L. Instituto Nacional de Ciencias<br />

Médicas y Nutrición Salvador Zubirán, México, DF, Mexico; Centro de<br />

Investigación y de Estudios Avanzados del I.P.N., México, DF, Mexico.<br />

Tumor Cell and Molecular Biology: New Drugs and Mechanisms<br />

P4-07-01 The Class I Selective PI3K Inhibitor GDC-0941 Enhances the<br />

Efficacy of Docetaxel in Human <strong>Breast</strong> <strong>Cancer</strong> Models by<br />

Increasing the Rate of Apoptosis<br />

Wallin JJ, Guan J, Prior WW, Lee LB, Ross L, Belmont LD, Koeppen<br />

H, Belvin M, Sampath D, Friedman LS. Genentech, Inc., South <strong>San</strong><br />

Francisco, CA.<br />

P4-07-02 Withdrawn<br />

P4-07-03 Rapamycin and Dalotuzumab in combination inhibit parental<br />

and endocrine resistant breast cancer cells<br />

Beckwith H, Fettig-Anderson L, Yueh N, Douglas Y. University of<br />

Minnesota, Minneapolis, MN.<br />

P4-07-04 Methioninase cell-cycle synchronization potentiates<br />

chemotherapy for breast cancer<br />

Yano S, Li S, Han Q, Tan Y, Fujiwara T, Hoffman RM. Anti<strong>Cancer</strong> Inc.,<br />

<strong>San</strong> Diego, CA; Okayama University Graduate School of Medicine and<br />

Dentistry, Okayama, Japan; University of California, <strong>San</strong> Diego, CA.<br />

P4-07-05 MEDI3379, an antibody against HER3, is active in HER2-driven<br />

human breast tumor models<br />

Xiao Z, Rothstein R, Carrasco R, Wetzel L, Kinneer K, Chen H, Tice D,<br />

Hollingsworth R, Steiner P. MedImmune, LLC, Gaithersburg, MD.<br />

Tumor Cell and Molecular Biology: Drug Resistance<br />

P4-08-01 PI3K/mTOR inhibition overcomes in vitro and in<br />

vivo trastuzumab resistance independent of feedback<br />

activation of pAKT<br />

O’Brien NA, McDonald K, Tong L, Von Euw E, Conklin D, Kalous O,<br />

Di Tomaso E, Schnell C, Linnartz R, Hurvitz SA, Finn RS, Hirawat<br />

S, Slamon DJ. UCLA, Los Angeles, CA; Novartis Pharmaceuticals,<br />

Cambridge, MA; Novartis Pharmaceuticals, Basel, Switzerland.<br />

P4-08-02<br />

P4-08-03<br />

P4-08-04<br />

Reactivation of oncogenic signaling through mTOR inhibitorsinduced<br />

feedback adaptations<br />

Rodrik-Outmezguine V, Chandarlapaty S, Poulikakos P, Scaltriti M,<br />

Baselga J, Rosen N. MSKCC, New York, NY; MGH, Charlestown, MA.<br />

The impact of the heregulin-HER receptor signaling axis on<br />

response to HER tyrosine kinase inhibitors<br />

Gwin WR, Liu L, Zhao S, Xia W, Spector NL. Duke University, Durham,<br />

NC.<br />

Abcc10 status affects proliferation, metastases and tumor<br />

sensitivity<br />

Domanitskaya N, Paulose C, Jacobs J, Foster K, Hopper-Borge E. Fox<br />

Chase <strong>Cancer</strong> Center, Philadelphia, PA.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012 48s <strong>Cancer</strong> Research


December 4–8, 2012<br />

Program Schedule<br />

P4-08-05 Basement membrane localized tumor cells are protected from<br />

HER2-targeted therapy in vivo<br />

Zoeller JJ, Bronson RT, Gilmer TM, Selfors LM, Lu Y, Apple SK, Press<br />

MF, Hurvitz SA, Slamon DJ, Mills GB, Brugge JS. Harvard Medical<br />

School, Boston, MA; GlaxoSmithKline, Research Triangle Park, NC; UT<br />

MD Anderson <strong>Cancer</strong> Center, Houston, TX; University of California, Los<br />

Angeles, CA; University of Southern California, Los Angeles, CA.<br />

P4-08-06 Notch-dependent Regulation of Novel Genes Associated with<br />

Trastuzumab Resistance<br />

Osipo C, Baumgartner A, Zlobin A, O’Toole M. Loyola University<br />

Chicago, Maywood, IL.<br />

P4-08-07 Novel insight into the tumor “flare” phenomenon and lapatinib<br />

resistance<br />

Piede JA, Zhao S, Liu L, Lyerly HK, Osada T, Wang T, Xia W, Spector N.<br />

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

P4-08-08 HOXC10, a homeobox protein overexpressed in breast cancer,<br />

modulates the response to chemotherapy treatment<br />

Sadik H, Nguyen N, Panday H, Kumar R, Pandita T, Sukumar S. Sidney<br />

Kimmel Comprehensive <strong>Cancer</strong> Center at Johns Hopkins, Baltimore,<br />

MD; UT Southwestern Medical Center, Dallas, TX.<br />

P4-08-09 Targeting Thyroid Receptor β in Estrogen Receptor Negative<br />

<strong>Breast</strong> <strong>Cancer</strong><br />

Gu G, Covington K, Rechoum Y, O’Malley B, Mangelsdorf D, Minna<br />

J, Webb P, Fuqua S. Baylor College of Medicine, Houston, TX; UT<br />

Southwestern Medical Center; The Methodist Hospital Research<br />

Institute.<br />

P4-08-10 Systematic expression analysis of the genes related to drugresistance<br />

in isogenic docetaxel- and adriamycin-resistant<br />

breast cancer cell lines<br />

Tang J, Li W, Zhong S, Xu J, Zhao J. Jiangsu <strong>Cancer</strong> Hospital, Nanjing,<br />

Jiangsu, China; Jiangsu <strong>Cancer</strong> Hospital, Nanjing, Jiangsu, China.<br />

Prognostic and Predictive Factors: Prognostic Factors - Preclinical<br />

P4-09-01 Retrospective evaluation of precision of gene-expressionbased<br />

signatures of prognosis and tumor biology in replicate<br />

surgical biospecimens from patients with breast cancer<br />

Barry WT, Marcom PK, Geradts J, Datto MB. Duke University Medical<br />

Center, Durham, NC.<br />

P4-09-02 A robust signature of long-term clinical outcome in breast<br />

cancer<br />

Boudreau A, Elias SG, Yau C, Wolf DM, van’t Veer LJ. University of<br />

California, <strong>San</strong> Francisco; Netherlands <strong>Cancer</strong> Institute.<br />

P4-09-03 Withdrawn<br />

P4-09-04 Gene expression profile that predict outcome of Tamoxifentreated<br />

estrogen receptor-positive, high-risk, primary breast<br />

cancer patients: a DBCG study<br />

Lyng MB, Lænkholm A-V, Tan Q, Vach W, Gravgaard KH, Knoop<br />

A, Ditzel HJ. University of Southern Denmark, Odense, Denmark;<br />

Slagelse Hospital, Slagelse, Denmark; Odense University Hospital,<br />

Odense, Denmark; University Medical Center Freiburg, Germany.<br />

P4-09-05 Microarray anlyses of breast cancers identify CH25H, a<br />

cholesterol gene, as a potential marker and target for late<br />

metastatic reccurences<br />

Saghatchian M, Mittempergher L, Michiels S, Wolf D, Canisius<br />

SV, Dessen P, Delaloge S, Lazar V, Benz SC, Roepman P, Glas AM,<br />

Tursz T, Bernards R, van’t Veer LJ. The Netherlands <strong>Cancer</strong> Institute,<br />

Amsterdam, Netherlands; Institut Jules Bordet, Brussels, Belgium;<br />

UCSF Helen Diller Family Comprehensive <strong>Cancer</strong> Center, <strong>San</strong><br />

Francisco; Institut Gustave Roussy, Villejuif, France; University of<br />

California, <strong>San</strong>ta Cruz; Agendia, Amsterdam, Netherlands.<br />

P4-09-06 miR-187 is an independent prognostic factor in lymph nodepositive<br />

breast cancer patients<br />

O’Connor DP, Mulrane L, Brennan DJ, Madden S, Gremel G, McGee<br />

SF, McNally S, Martin FM, Crown JP, Jirstrom K, Higgins DG, Gallagher<br />

W. UCD Conway Institute, Dublin, Ireland; Molecular Therapeutics for<br />

<strong>Cancer</strong> Ireland, Dublin City University, Dublin, Ireland; St Vincent’s<br />

University Hospital, Dublin, Ireland; Lund University, Lund, Sweden.<br />

P4-09-07 ING1 Expression Measured by AQUA can be an Independent<br />

Prognostic Marker in <strong>Breast</strong> <strong>Cancer</strong><br />

Thakur S, Klimowicz A, Pohorelic B, Dean M, Konno M, Bose P,<br />

Magliocco A, Riabowol K. University of Calgary, AB, Canada; Tom Baker<br />

<strong>Cancer</strong> Center, Calgary, AB, Canada.<br />

P4-09-08 HOXB9, a gene promoting tumor angiogenesis and<br />

proliferation, is significantly associated with poor clincal<br />

outcomes in ER-positive breast cancer patients<br />

Seki H, Hayashida T, Jinno H, Takahashi M, Suzuki K, Kaneda M, Hara<br />

H, Osaku M, Asanuma F, Yamada Y, Mukai M, Kitagawa Y. Kitasato<br />

University Kitasato Institute Hospital, Tokyo, Japan; Keio University<br />

School of Medicine, Tokyo, Japan; Keio University School of Medicine,<br />

Japan.<br />

P4-09-09 Circulating HER2 extracellular domain (ECD) levels are<br />

associated with progression-free survival in metastatic breast<br />

cancer patients<br />

Wang X-j, Shao X-Y, Chen Z-H, Ye W-w, Qin J, Zhang Y-m, Zheng Y-b,<br />

Yu X-y, Lei L, Ling Z-Q, Feng J-g, Han MX. Zhejiang <strong>Cancer</strong> Hospital,<br />

Hangzhou, Zhejiang, China; Hangzhou Polar Gene Company,<br />

Hangzhou, Zhejiang, China.<br />

P4-09-10 Prospective Analysis of Fatty Acid Synthase (FASN) in <strong>Breast</strong><br />

<strong>Cancer</strong> Tissue of Early-Stage <strong>Breast</strong> <strong>Cancer</strong> Patients<br />

Puig T, Blancafort A, Casoliva G, Oliveras G, Casas M, Buxo M, Saiz E,<br />

Viñas G, Dorca J, Porta R. University of Girona and Girona Biomedical<br />

Research Institute (IDIBGi), Girona, Spain; Assistència <strong>San</strong>itària<br />

Institute, Girona, Spain; Catalan Institute of Oncology, Girona, Spain;<br />

Dr. Josep Trueta Hospital, Girona, Spain; Dr. Josep Trueta Hospital and<br />

Girona Biomedical Research Institute (IDIBGi), Girona, Spain; Health<br />

Inequalities Research Group - Employment Conditions Knowledge<br />

Network (GREDS-EMCONET), Universitat Pompeu Fabra, Barcelona,<br />

Spain; Dr. Josep Trueta Hospitaland Girona Biomedical Research<br />

Institute (IDIBGi), Girona, Spain.<br />

P4-09-11 Fatty Acid Synthase (FASN) expression in Triple-Negative<br />

<strong>Breast</strong> <strong>Cancer</strong><br />

Viñas G, Oliveras G, Perez-Bueno F, Giro A, Blancafort A, Puig-Vives<br />

M, Marcos-Gragera R, Dorca J, Brunet J, Puig T. Catalan Institute of<br />

Oncology and Girona Biomedical Research Institute (IDIBGi), Girona,<br />

Spain; University of Girona and Biomedical Research Institute (IDIBGi),<br />

Girona, Spain; Dr. Josep Trueta Hospital and Catalan Institute of<br />

Health (ICS), Girona, Spain; Catalan Institute of Oncology, Girona,<br />

Spain; University of Girona, Spain.<br />

P4-09-12 TIMP-4 – Prognostic Marker and Treatment Target for Triple-<br />

Negative <strong>Breast</strong> <strong>Cancer</strong>s<br />

Wallon UM, Sabol JL, Gilman PB, Zemba-Palko V, DuHadaway JB,<br />

Ciocca RM, Ali ZA, Carp NZ, Choy NS, Wojciechowski BS, Prendergast<br />

GC. Lankenau Medical Center, Wynnewood, PA.<br />

Epidemiology, Risk, and Prevention: Familial <strong>Breast</strong> <strong>Cancer</strong> - Molecular<br />

Genetics<br />

P4-10-01 Edgetic perturbation of BRCT-mediated interactions caused by<br />

the BRCA1 H1686Q sequence variant<br />

De Nicolo A, Pathania S, Parisini E, Joukov V. Dana-Farber <strong>Cancer</strong><br />

Institute, Boston, MA; Harvard Medical School, Boston, MA; Italian<br />

Institute of Technology, Milan, Italy.<br />

www.aacrjournals.org<br />

49s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P4-10-02 Targeted resequencing of BRCA1 and BRCA2 in inherited<br />

breast cancer<br />

Wong MW, Li S, Wilkins M, Avery-Kiejda KA, Bowden NA, Scott RJ.<br />

University of Newcastle, NSW, Australia; University of New South<br />

Wales, Sydney, NSW, Australia; Hunter Area Pathology Service (HAPS),<br />

Newcastle, NSW, Australia.<br />

Epidemiology, Risk, and Prevention: Familial <strong>Breast</strong> <strong>Cancer</strong> - Genetic<br />

Testing<br />

P4-11-01 Rapid genetic counseling and testing in newly diagnosed<br />

breast cancer patients, findings from an RCT<br />

Wevers MR, Ausems MG, Bleiker EM, Rutgers EJ, Witkamp AJ, Hahn<br />

DE, Brouwer T, Kuenen MA, van der <strong>San</strong>den-Melis J, van der Luijt RB,<br />

Hogervorst FB, van Dalen T, Theunissen EB, van Ooijen B, de Roos<br />

MA, Borgstein PJ, Vrouenraets BC, Huisman JJ, Bouma WH, Rijna H,<br />

Vente JP, Valdimarsdottir H, Verhoef S, Aaronson NK. Netherlands<br />

<strong>Cancer</strong> Institute - Antoni van Leeuwenhoek Hospital, Amsterdam,<br />

Netherlands; University Medical Center, Utrecht, Netherlands;<br />

Diakonessen Hospital, Utrecht, Netherlands; St. Antonius Hospital,<br />

Nieuwegein, Netherlands; Meander Medical Center, Amersfoort,<br />

Netherlands; Rivierenland Hospital, Tiel, Netherlands; Onze Lieve<br />

Vrouwe Gasthuis, Amsterdam, Netherlands; St. Lucas Andreas<br />

Hospital, Amsterdam, Netherlands; Tergooi Hospitals, Blaricum,<br />

Netherlands; Gelre Hospitals, Apeldoorn, Netherlands; Kennemer<br />

Gasthuis, Haarlem, Netherlands; Zuwe Hofpoort Hospital, Woerden,<br />

Netherlands; Mount Sinai School of Medicine, New York.<br />

P4-11-02 Novel BRCA1 and BRCA2 genomic rearrangements in Southern<br />

Chinese breast/ovarian cancer patients<br />

Kwong A, Ng EKO, Law FBF, Wa A, Wong CLP, Wong CHN, Kurian AW,<br />

West DW, Ford JM, Ma ESK. University of Hong Kong, Pokfulam, Hong<br />

Kong; Hong Kong <strong>San</strong>itorium & Hospital, Happy Valley, Hong Kong;<br />

Hong Kong Hereditary <strong>Breast</strong> <strong>Cancer</strong> Family Registry, Happy Valley,<br />

Hong Kong; Stanford University School of Medicine, Stanford, CA.<br />

P4-11-03 Single nucleotide polymorphism testing for breast cancer risk<br />

assessment: patient trust and willingness to pay<br />

Howe R, Omer Z, Hanoch Y, Miron-Shatz T, Thorsen C, Ozanne EM.<br />

Universiy of California <strong>San</strong> Francisco, CA; Massachusetts General<br />

Hospital, Boston, MA; Plymouth University, United Kingdom; Ono<br />

Academic College, Israel.<br />

P4-11-04 A Structured Genetic Risk Evaluation and Testing Program in<br />

the Community Oncology Practice Increases Identification of<br />

Individuals at Risk for BRCA Mutations<br />

Langer L, Clark L, Gress J, Patt D, Denduluri N, Wang Y, Andersen J,<br />

Solti M, Wheeler A, Delamelena T, Smith, II JW, <strong>San</strong>dbach J. Compass<br />

Oncology, Portland, OR; Texas Oncology, Austin, TX; Virginia <strong>Cancer</strong><br />

Specialists, Arlington, VA; McKesson Specialty Health/The US<br />

Oncology Network, The Woodlands, TX.<br />

P4-11-05 Optimal age to start preventive measures in women with<br />

BRCA1/2 mutations or high familial breast cancer risk<br />

Tilanus-Linthorst MMA, Lingsma H, Evans GD, Kaas R, Manders P,<br />

Hooning MJ, van Asperen CJ, Thompson D, Eeles R, Oosterwijk<br />

JC, Leach MO, Steyerberg EJ. Erasmus University MC, Rotterdam,<br />

Netherlands; Erasmus University MC, Netherlands; University of<br />

Manchester, United Kingdom; NKI/AVL, Amsterdam, Netherlands;<br />

UMC St Radboud, Netherlands; Leiden University Medical Centre,<br />

Netherlands; Centre for <strong>Cancer</strong> Genetic Epidemiology, Cambridge,<br />

United Kingdom; Royal Marsden NHS Foundation, United Kingdom;<br />

University Medical Centre Groningen, Netherlands; Institute of <strong>Cancer</strong><br />

Research, Royal Marsden, Sutton, United Kingdom.<br />

P4-11-06 Automatic referral to genetic counseling for identification<br />

of BRCA1/2 mutations: a pilot program at Norton <strong>Cancer</strong><br />

Institute, Louisville, KY<br />

Lewis AL, Alabek ML, Dreher C, Goldberg JM, Brooks SE. Norton<br />

Healthcare, Louisville, KY.<br />

Epidemiology, Risk, and Prevention: Risk Factors and Modeling<br />

P4-12-01 A nomogram based on clinical, imaging and histological data<br />

to predict the risk of upgrades to malignancy at surgery in<br />

biopsy-diagnosed premalignant lesions of the breast<br />

Uzan C, Mazouni C, Balleyguier C, Mathieu M-C, Ferchiou M, Delaloge<br />

S. Institut Gustave Roussy, Villejuif, France.<br />

P4-12-02 Serum 25-hydroxyvitamin D3 is associated with decreased risk<br />

of postmenopausal breast cancer in whites: the Multiethnic<br />

Cohort Study<br />

Kim Y, Franke AA, Shvetsov YB, Wilkens LR, Lurie G, Cooney RV,<br />

Maskarinec G, Hernandez BY, Le Marchand L, Henderson B, Kolonel<br />

LN, Goodman MT. University of Hawai’i <strong>Cancer</strong> Center; University of<br />

Hawaii; Keck School of Medicine, University of Southern California.<br />

P4-12-03 Towards a risk prediction model for breast cancer that utilizes<br />

breast tissue risk features<br />

Pankratz VS, Degnim AC, Visscher DW, Frank RD, Vierkant RA, Ghosh<br />

K, Aziza N, Vachon CM, Frost M, Radisky DC, Hartmann LC. Mayo<br />

Clinic, Rochester, MN; Mayo Clinic, Jacksonville, FL.<br />

P4-12-04 Association of single-strand breaks (SSBs) in normal breast<br />

DNA with estimates of breast cancer risk<br />

Chatterton RT, Sahadevan M, Heinz RE, Sukumar S, Stearns V, Fackler<br />

MJ, Lee O, Sivaraman I, Kenney K, Khan SA. Northwestern University<br />

Feinberg School of Medicine, Chicago, IL; Johns Hopkins School of<br />

Medicine, Baltimore, MD.<br />

P4-12-05 Enrichment of aldosterone synthase (CYP11B2) gene C-allele<br />

carriers in women at high risk in the OncoVue® polyfactorial<br />

risk model<br />

Jupe ER, Pugh TW, Knowlton NS, DeFreese DC. InterGenetics<br />

Incorporated, Oklahoma City, OK; NSK Statistical Solutions, Choctaw,<br />

OK.<br />

P4-12-06 A mammographic density prediction model using<br />

environmental factors, endogenous hormones and growth<br />

factors in Japanese women<br />

Yoshimoto N, Nishiyama T, Toyama T, Takahashi S, Shiraki N, Sugiura<br />

H, Endo Y, Iwasa M, Asano T, Fujii Y, Yamashita H. Nagoya City<br />

University Graduate School of Medical Sciences, Nagoya, Aichi, Japan.<br />

P4-12-07 Diabetes Increases the Risk of <strong>Breast</strong> <strong>Cancer</strong><br />

Hardefeldt PJ, Edirimanne S, Eslick GD. University of Sydney, NSW,<br />

Australia; Nepean Hospital, University of Sydney, Penrith, NSW,<br />

Australia.<br />

Epidemiology, Risk, and Prevention: Epidemiology, Risk, and Prevention -<br />

Other<br />

P4-13-01 Racial Disparities in the Incidence of Dose-limiting<br />

Chemotherapy Induced Peripheral Neuropathy<br />

Speck RM, Sammel MD, Farrar JT, Hennessy S, Mao JJ, Stineman<br />

MG, DeMichele A. Perelman School of Medicine, University of<br />

Pennsylvania.<br />

P4-13-02 Comparing data quality of client intake forms by interview<br />

mode: results of a pilot study on the use of audio computerassisted<br />

self-interview (ACASI) in the Avon <strong>Breast</strong> Health<br />

Outreach Program<br />

Hallum-Montes R, Senter L, D’Souza R, Hurlbert M, Gates-Ferris K,<br />

Anastario M. CAI Global, New York, NY; Avon Foundation for Women,<br />

New York, NY; New York University, New York, NY.<br />

P4-13-03 Hormone replacement therapy, is there an increased risk of in<br />

situ breast cancer? Data from a French cohort<br />

Coussy F, Giacchetti S, Hamy A-S, Porcher R, Cuvier C, Lalloum M,<br />

de Roquancourt A, Albiter M, Espié M. Hôpital St Louis, Paris, France;<br />

Assistance Publique-Hôpitaux de Paris, Hôpital St Louis, Paris, France.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012 50s <strong>Cancer</strong> Research


December 4–8, 2012<br />

Program Schedule<br />

P4-13-04 Estrogen and Avoidance of Invasive <strong>Breast</strong> <strong>Cancer</strong>, Coronary<br />

Heart Disease and All-cause Mortality. Public Health Impact of<br />

Estrogen Guidelines for Women entering Menopause<br />

Ragaz J, Wilson K, Shakeraneh S, Budlovsky J, Wong H. University of<br />

British Columbia, Vancouver, BC, Canada; British Columbia <strong>Cancer</strong><br />

Agency, Victoria, BC, Canada.<br />

P4-13-05 The gap between perceptions of risk and actual risk for<br />

breast cancer<br />

Kaplan CP, Lopez M, Tice J, Pasick R, Kerlikowske K, Chen A, Karliner L.<br />

University of California, <strong>San</strong> Francisco, CA.<br />

P4-13-06 Association of Age, Obesity and Incident <strong>Breast</strong> <strong>Cancer</strong><br />

Phenotypes<br />

Scheri R, Power S, Marks J, Seewaldt V, Marcom K, Hwang S. Duke<br />

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

P4-13-07 Meta-analysis of epidemiological studies of Insulin Glargine<br />

and <strong>Breast</strong> <strong>Cancer</strong> Risk<br />

Boyle P, Koechlin A, Boniol M, Bota M, Robertson C, Rosenstock J,<br />

Bolli GB. International Prevention Research Institute, Lyon, France;<br />

University of Strathclyde, Glasgow, United Kingdom; Dallas Diabetes<br />

and Endocrine Center, Dallas; University of Perugia, Italy.<br />

P4-13-08 Diabetes, Related Factors and <strong>Breast</strong> <strong>Cancer</strong> Risk<br />

Boyle P, Boniol M, Koechlin A, Bota M, Robertson C, Leroith D,<br />

Rosenstock J, Bolli GB, Autier P. International Prevention Research<br />

Institute, Lyon, France; University of Strathclyde, Glasgow, United<br />

Kingdom; Mt. Sinai, New York; Dallas Diabetes and Endocrine Center,<br />

Dallas; University of Perugia, Italy.<br />

P4-13-09 The effect of weight change on breast adipose and<br />

dense tissue<br />

Graffy HJ, Harvie MN, Warren RM, Boggis CR, Astley SM, Evans<br />

GD, Adams JE, Howell A. University Hospital of South Manchester,<br />

Manchester, United Kingdom; Addenbrooke’s Hospital, Cambridge,<br />

United Kingdom; University of Manchester, United Kingdom; Central<br />

Manchester University Hospitals NHS Foundation Trust, Manchester,<br />

United Kingdom.<br />

P4-13-10 Do BRCA1 and BRCA2 mutation carriers have an earlier natural<br />

menopause than their non-carrier relatives: A study from the<br />

Kathleen Cuningham Foundation Consortium For Research<br />

Into Familial <strong>Breast</strong> <strong>Cancer</strong> (kConFab)<br />

Collins IM, Milne RL, McLachlan SA, Friedlander M, Birch KE,<br />

Weideman PC, Hopper JL, Phillips K-A. Peter MacCallum <strong>Cancer</strong><br />

Centre, Melbourne, Australia; University of Melbourne, Australia; St<br />

Vincent’s Hospital, Melbourne, VIC, Australia; Prince of Wales <strong>Cancer</strong><br />

Centre, Sydney, Australia<br />

P4-13-11 Prognostic factors in young breast cancer patients over time –<br />

a 40 year longitudinal analysis<br />

Hagenbeck C, Muschler B, Jaeger BAS, Jueckstock J, Andergassen<br />

U, Katzorke N, Hepp P, Melcher CA, Janni JW, Rack BK. Heinrich<br />

Heine University, Duesseldorf, Germany; Freising Hospital, Freising,<br />

Germany; Ludwig-Maximilians-University, Munich, Germany.<br />

P4-13-12 Identifying women at increased risk for breast cancer using the<br />

electronic health record in an integrated health system<br />

Leader JB, Bengier A, Darer J, Stark A, Vogel VG. Geisinger Health<br />

System, Danville, PA; Geisinger Health System, Lewisburg, PA.<br />

P4-13-13 Risk Assessment and Personalized Decision Support: The<br />

University of California Athena <strong>Breast</strong> Health Network<br />

Ozanne EM, Crawford B, Petruse A, Madlensky L, Weiss L, Hogarth M,<br />

Wenger N, Goodman D, Park H, Anton-Culver H, Yasmeen S, Howell<br />

L, Ojeda H, Parker BA, Kaplan C, van’t Veer L, Esserman L, Naeim A.<br />

University of California, <strong>San</strong> Francisco, CA; University of California, Los<br />

Angeles, CA; University of California, <strong>San</strong> Diego, CA; Athena Program<br />

Management Office, <strong>San</strong> Francisco, CA; University of California, Davis,<br />

CA; University of California, Irvine, CA.<br />

P4-13-14 Lung cancer after treatment of breast cancer: retrospective<br />

study from Curie Institut<br />

Sebbagh S, Cosquer M, Kirova YM, Livartowski A. Institut Curie, Paris,<br />

France.<br />

Treatment: Surgery<br />

P4-14-01 Incidence rate of nipple areolar complex ischemia after nipple<br />

sparing mastectomy. Analysis of the American Society of<br />

<strong>Breast</strong> Surgeons Nipple Sparing Mastectomy Registry<br />

Mitchell SD, Willey SC, Feldman SM, Beitsch PD, Unzeitig GW,<br />

Manasseh DME, Neumayer LA, Laidley AL, Grutman SB, Habal N,<br />

Busch-Devereaux E, Dupont EL, Ashikari AY. White Plains Hospital<br />

Medical Center, White Plains, NY; Georgetown, Washington, DC;<br />

Columbia University College of Physicians & Surgeons, New York,<br />

NY; Dallas <strong>Breast</strong> Center, Dallas, TX; Laredo <strong>Breast</strong> Care, Laredo, TX;<br />

Maimonides Medical Center, Brooklyn, NY; University of Utah, Salt<br />

Lake City, UT; Texas <strong>Breast</strong> Specialists, Texas Oncology, Dallas, TX;<br />

American Society of <strong>Breast</strong> Surgeons, Columbia, MD; Carolina <strong>Breast</strong><br />

& Oncologic Surgery, Greenville, NC; <strong>Breast</strong> Surgery Associates,<br />

Greenlawn, NY; Watson Clinic, Lakeland, FL; Ashikari <strong>Breast</strong> Center,<br />

Dobbs Ferry, NY.<br />

P4-14-02 National Trends and Indications for Nipple-Sparing<br />

Mastectomy: An Analysis Using the Surveillance, Epidemiology,<br />

and End Results (SEER) Database<br />

Agarwal S, Agarwal S, Agarwal J. Wayne State University, Detroit, MI;<br />

University of Michigan, Ann Arbor, MI; University of Utah, Salt Lake<br />

City, UT.<br />

P4-14-03 Nipple-sparing mastectomy and intra-operative nipple biopsy:<br />

To freeze or not to freeze?<br />

Guth AA, Blechman K, Samra F, Shapiro R, Axelrod D, Choi M, Karp N,<br />

Alperovich M. NYU School of Medicine, New York, NY.<br />

P4-14-04 Total skin-sparing mastectomy in BRCA mutation carriers<br />

Warren Peled A, Hwang ES, Ewing CA, Alvarado M, Esserman LJ.<br />

University of California, <strong>San</strong> Francisco; Duke University Medical<br />

Center.<br />

P4-14-05 Skin sparing mastectomy – thorough breast tissue removal<br />

leads to a low local recurrence rate of breast cancer<br />

Paily AJ, Drabble EH. Derriford Hopsital, Plymouth, Devon, United<br />

Kingdom.<br />

P4-14-06 Withdrawn<br />

P4-14-07 Impact of preservation of the intercostobrachial nerve during<br />

axillary dissection on sensory change and health-related<br />

quality of life two years after breast cancer surgery<br />

Taira N, Shimozuma K, Ohsumi S, Kuroi K, Shiroiwa T, Watanabe T,<br />

Saito M. Okayama University Hospital, Okayama, Japan; Ritsumeikan<br />

University, Japan; National Shikoku <strong>Cancer</strong> Center, Japan; Tokyo<br />

Metropolitan <strong>Cancer</strong> and Infectious Diseases Center Komagome<br />

Hospital, Japan; Teikyo University, Japan; Sendai Medical Center,<br />

Japan; Juntendo University, Japan.<br />

P4-14-08 Intraoperative ultrasound guidance for excision of nonpalpable<br />

breast cancer<br />

Barentsz MW, van Dalen T, Gobardhan PD, Bongers V, Perre CI,<br />

Pijnappel RM, van den Bosch MA, Verkooijen HM. University Medical<br />

Center Utrecht, Netherlands; Diakonessenhuis, Utrecht, Netherlands;<br />

Amphia Hospital, Breda, Netherlands.<br />

P4-14-09 Feasibility of liposuction for treatment of arm lymphedema<br />

from breast cancer<br />

Doren EL, Smith PD, Sun W, Lacevic M, Fulp W, Reid R, Laronga C.<br />

University of South Florida, Tampa, FL; Moffitt <strong>Cancer</strong> Center,<br />

Tampa, FL.<br />

P4-14-10 Atypical Ductal Hyperplasia diagnosed on directional vacuumassisted<br />

biopsy: is surgical excision mandatory?<br />

Chauvet M-P, Rivaux G, Farre I, Houpeau J-L, Giard S, Ceugnart L.<br />

Centre Oscar Lambret, Lille, France.<br />

www.aacrjournals.org<br />

51s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P4-14-11 Risk reducing mastectomy for women with high personal<br />

breast cancer risk<br />

Onyekwelu O, Shetty G, Baildam A. Nightingale & Genesis <strong>Breast</strong><br />

<strong>Cancer</strong> Prevention Centre, University Hospital of South Manchester,<br />

Wythenshawe, Manchester, United Kingdom; St. Bartholomew’s<br />

Hospital (Bart’s), West Smithfield, London, United Kingdom.<br />

P4-14-12 Evaluation of the effect of pasireotide LAR administration in<br />

the lymphocele prevention after mastectomy with axillary<br />

lymph node dissection for breast cancer: results of a phase 2<br />

randomized study<br />

Chereau E, Uzan C, Zohar S, Bezu C, Mazouni C, Ballester M, Gouy<br />

S, Rimareix F, Garbay J-R, Darai E, Uzan S, Rouzier R. Tenon, APHP,<br />

UPMC - Paris 6, Paris, France; Institut Gustave Roussy, Villejuif, France;<br />

Hopital Saint-Louis, APHP, U444-INSERM, Paris 7, Paris, France.<br />

P4-14-13 Therapeutic mammaplasty does not cause a delay in the<br />

delivery of chemotherapy in high risk breast cancer patients<br />

Khan J, Barrett S, Stallard S, Forte C, Weiler-Mithoff E, Reid I, Winter<br />

A, Doughty J, Romics L. Victoria Infirmary, Glasgow, United Kingdom;<br />

Beatson West of Scotland <strong>Cancer</strong> Centre, Glasgow, United Kingdom;<br />

Western Infirmary, Glasgow, United Kingdom; Royal Infirmary,<br />

Glasgow, United Kingdom.<br />

P4-14-14 Phyllodes tumour of the breast: A retrospective analysis of<br />

87 cases<br />

Walter HS, Esmail F, Krupa J, Ahmed SI. Leicester Royal Infirmary,<br />

Leicester, United Kingdom; Glenfield Hospital, Leicester, United<br />

Kingdom.<br />

Treatment: <strong>Breast</strong> Conservation<br />

P4-15-01 Second conservative treatment for ipsilateral breast tumor<br />

recurrence: GEC-ESTRO <strong>Breast</strong> WG study<br />

Hannoun-Levi J-M, Resch A, Gal J, Niehoff P, Loessl K, Kovács G, Van<br />

Limbergen E, Polgar C. Antoine Lacassagne <strong>Cancer</strong> Center, University<br />

of Nice-Sophia, Nice, France; Medical University, General Hospital<br />

of Vienna, Austria; Antoine Lacassagne <strong>Cancer</strong> Center, Nice, France;<br />

University Erlangen-Nuremberg, Erlangen, Germany; University<br />

Hospital Bernes, Switzerland; University Hospital Schleswig-Holstein<br />

Campus Lübeck, Germany; University Hospital Gasthuisberg, Leuven,<br />

Belgium; National Institute of Oncology, Budapest, Hungary.<br />

P4-15-02 Timing of infectious complications following breast<br />

conserving therapy with catheter-based accelerated partial<br />

breast irradiation<br />

Haynes AB, Bloom ES, Bedrosian I, Kuerer HM, Hwang RF, Caudle AS,<br />

Hunt KK, Graviss L, Chemaly RF, Tereffe W, Shaitelman SF, Babiera GV.<br />

University of Texas MD Anderson <strong>Cancer</strong> Center, Houston, TX.<br />

P4-15-03 Patterns of relapse following re-irradiation of the breast using<br />

partial breast brachytherapy (PBB)<br />

Chadha M, Boachie-Adjei K, Boolbol SK, Kirstein L, Osborne MP, Tarter<br />

P, Harrison LB. Beth Israel Medical Center, New York, NY.<br />

P4-15-04 Impact of awake breast cancer surgery on postoperative<br />

lymphocyte responses<br />

Vanni G, De Felice V, Buonomo C, Esser A, Petrella G, Buonomo OC.<br />

Tor Vergata University, Rome, Italy.<br />

P4-15-05 Long-term outcome of breast cancer patients treated with<br />

radiofrequency ablation<br />

Earashi M, Noguchi M, Motoyoshi A, Komiya H, Fujii H. Yatsuo General<br />

Hospital, Toyama, Japan; Kanazawa Medical University Hospital,<br />

Uchinada-Daigaku, Ishikawa, Japan.<br />

Treatment: Radiotherapy<br />

P4-16-01 Accelerated hypofractionated whole breast radiotherapy<br />

for localized breast cancer: the effect of a boost on patient<br />

reported long-term cosmetic outcome<br />

Chan EK, Tabarsi N, Tyldesley S, Khan M, Woods R, Speers C, Weir L.<br />

British Columbia <strong>Cancer</strong> Agency, Vancouver Centre, Vancouver, BC,<br />

Canada; University of British Columbia, Vancouver, BC, Canada.<br />

P4-16-02<br />

P4-16-03<br />

P4-16-04<br />

P4-16-05<br />

P4-16-06<br />

P4-16-07<br />

P4-16-08<br />

P4-16-09<br />

P4-16-10<br />

A survival benefit from locoregional radiotherapy for<br />

node-positive and CMF treated breast cancer is most<br />

significant in Luminal A tumors<br />

Voduc D, Cheang MCU, Tyldesley S, Chia S, Gelmon K, Speers C,<br />

Nielsen TO. BC <strong>Cancer</strong> Agency, Vancouver, BC, Canada; University of<br />

British Columbia, Vancouver, BC, Canada.<br />

Patterns of failure after accelerated partial breast irradiation<br />

by consensus panel group: A pooled analysis of William<br />

Beaumont Hospital and the American Society of <strong>Breast</strong><br />

Surgeons Trial data<br />

Wilkinson JB, Beitsch PD, Arthur D, Shah C, Haffty BG, Wazer D, Keisch<br />

M, Shaitelman SF, Lyden M, Chen PY, Vicini FA. Oakland University<br />

William Beaumont School of Medicine, Royal Oak, MI; Dallas Surgical<br />

Group, Dallas, TX; Massey <strong>Cancer</strong> Center, Virginia Commonwealth<br />

University, Richmond, VA; Washington University School of Medicine,<br />

St. Louis, MO; <strong>Cancer</strong> Institute of New Jersey, Robert Wood Johnson<br />

Medical School, Camden, NJ; Tufts Medical Center and Rhode Island<br />

Hospital/Brown University, Boston, MA; <strong>Cancer</strong> Healthcare Associates,<br />

Miami, FL; University of Texas M.D. Anderson <strong>Cancer</strong> Center, Houston,<br />

TX; Biostat International, Inc., Tampa, FL; Michigan Healthcare<br />

Professionals/21st Century Oncology, Farmington Hills, MI.<br />

Outcome of stage II/III breast cancer treated with neoadjuvant<br />

versus adjuvant radiotherapy in British Columbia<br />

Lohmann AE, Voduc D, Speers C, Chia S. British Columbia <strong>Cancer</strong><br />

Agency, Vancouver, BC, Canada.<br />

Benefit from Postoperative Radiotherapy for N1 <strong>Breast</strong> <strong>Cancer</strong><br />

Tsai Y-CS, Cheng H-CS, Yu B-L, Horng C-F, Chen C-M, Jian J-MJ, Chu<br />

N-M, Tsou M-H, Liu M-C, Huang AT. Koo Foundation Sun Yat-Sen<br />

<strong>Cancer</strong> Center, Taipei, Taiwan; Duke University Medical Center,<br />

Durham, NC.<br />

Radiotherapy to the Primary Tumor Is Associated with<br />

Improved Survival in Stage IV <strong>Breast</strong> <strong>Cancer</strong><br />

Morgan SC, Caudrelier J-M, Clemons MJ. University of Ottawa, ON,<br />

Canada; The Ottawa Hospital <strong>Cancer</strong> Centre, Ottawa, ON, Canada.<br />

Selective use of post-mastectomy radiation therapy in the<br />

neoadjuvant setting<br />

Warren Peled A, Wang F, Stover AC, Rugo HS, Melisko ME, Park JW,<br />

Alvarado M, Ewing CA, Esserman LJ, Fowble B, Hwang ES. University<br />

of California, <strong>San</strong> Francisco; Duke University Medical Center.<br />

Intraoperative electron radiotherapy in early stage breast<br />

cancer. A single-institution experience<br />

Dall’Oglio S, Maluta S, Marciai N, Gabbani M, Franchini Z, Pietrarota P,<br />

Meliadò G, Guariglia S, Cavedon C. University Hospital, Verona, Italy.<br />

Dosimetric feasibility and acute toxicity in a prospective trial<br />

of ultra-short course accelerated partial breast irradiation<br />

(APBI) using a multi-lumen balloon brachytherapy device<br />

Khan AJ, Vicini FA, Brown S, Haffty BG, Kearney T, Dale R, Arthur<br />

DW. <strong>Cancer</strong> Institute of New Jersey, New Brunswick, NJ; Michigan<br />

Healthcare Professionals, Farmington Hills, MI; Wellstar Kennestone<br />

Hospital, Marietta, GA; Imperial College London, United Kingdom;<br />

Virginia Commonwealth University, Massey <strong>Cancer</strong> Center,<br />

Richmond, VA<br />

Positive sentinel lymph nodes (SLNs) without axillary<br />

dissection in breast cancer: the tangent fields of irradiation<br />

may not allow optimal coverage and dose distribution in<br />

level I and II of the axilla<br />

Qiong P, Khodari W, Bigorie V, Bosc R, Dao TH, Totobenazara J-L,<br />

Assaf E, Caillet P, Diana C, Lagrange J-L, Calitchi E, Belkacemi Y.<br />

APHP GH Henri Mondor et Université de Paris-Est, Créteil, France;<br />

APHP GH Henri Mondor, Créteil, France; Association of Radiotherapy<br />

and Oncology of the Mediterranean Area, France; French <strong>Breast</strong><br />

Intergroup, France.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012 52s <strong>Cancer</strong> Research


December 4–8, 2012<br />

Program Schedule<br />

P4-16-11 Impact of receptor status on prognosis among<br />

breast cancer patients with brain metastases treated with<br />

Cyberknife radiosurgery<br />

Fasola CE, Gibbs IC, Soltys SG, Horst KC. Stanford <strong>Cancer</strong> Center,<br />

Stanford, CA.<br />

P4-16-12 Outcomes of low-risk Ductal Carcinoma in situ in South East<br />

Asian women treated with breast conservation surgery<br />

Wong FY, Wang FQ, Chen JJ, Tan CH, Tan PH. National <strong>Cancer</strong> Centre<br />

Singapore; Singapore General Hospital, Singapore, Singapore.<br />

P4-16-13 Relationship between coverage of axillary lymph nodes, with<br />

tangential breast irradiation, and body mass index<br />

Inokuchi M, Furukawa H, Fujimura T, Ohta T, Ohashi S, Takanaka T.<br />

Kanazawa University Hospital, Kanazawa, Ishikawa, Japan.<br />

P4-16-14 Salvage Radiotherapy and Cisplatin for Triple Negative <strong>Breast</strong><br />

<strong>Cancer</strong>: A Multi-Centre Study<br />

Lee JW, Brackstone M, Gandhi S, Arce Salinas C, Dinniwell R.<br />

University of Toronto, ON, Canada; London Regional <strong>Cancer</strong> Program,<br />

London, ON, Canada; Princess Margaret Hospital, University of<br />

Toronto, ON, Canada.<br />

P4-16-15 Improvement of accuracy and consistency in delineating the<br />

breast lumpectomy cavity using surgical clips<br />

Atrchian S, Sadeghi P, Cwajna W, Helyer L, Rheaume D, Nolan<br />

M, Rutledge R, Calverley V, Bennett S, Ago T, Robar J. Dalhousie<br />

University, Halifax, NS, Canada.<br />

Treatment: Reconstruction<br />

P4-17-01 The effect of body mass index on breast reconstruction<br />

outcomes<br />

Lopez JJ, Laronga C, Doren EL, Sun W, Fulp WJ, Smith PD. University<br />

of South Florida, Tampa, FL; H. Lee Moffitt <strong>Cancer</strong> Center, Tampa, FL.<br />

P4-17-02 Radiation therapy and expander-implant breast<br />

reconstruction: an analysis of timing and comparison of<br />

complications<br />

Lentz RB, Higgins SA, Matthew MK, Kwei SL. Yale University School of<br />

Medicine, New Haven, CT.<br />

P4-17-03 Towards the standardisation of outcome reporting in<br />

reconstructive breast surgery: Initial results of the BRAVO<br />

(<strong>Breast</strong> Reconstruction and Valid Outcome) Study–A<br />

multicentre consensus process to develop a core outcome set<br />

for reconstructive breast surgery<br />

Potter S, Ward J, Cawthorn S, Holcombe C, Warr R, Wilson S, Tillett<br />

R, Weiler-Mithoff E, Winters Z, Barker J, Oates C, Harcourt D, Brookes<br />

S, Blazeby J. University of Bristol, United Kingdom; North Bristol NHS<br />

Trust, Bristol, United Kingdom; Linda McCartney <strong>Breast</strong> Centre, Royal<br />

Liverpool and Broadgreen University Hospitals NHS Trust, Liverpool,<br />

United Kingdom; NHS Greater Glasgow and Clyde, Glasgow, United<br />

Kingdom; University of the West of England, Bristol, United Kingdom.<br />

P4-17-04 BRECONDA: Development and acceptability of an interactive<br />

decisional support tool for women considering breast<br />

reconstruction<br />

Sherman KA, Harcourt D, Lam T, Boyages J. Macquarie University,<br />

Sydney, NSW, Australia; Westmead Hospital, University of Sydney,<br />

Westmead, NSW, Australia; University of the West of England, Bristol,<br />

United Kingdom.<br />

P4-17-05 Withdrawn<br />

P4-17-06 The use of specialist nurse-led clinics in preparation for<br />

reconstructive breast surgery<br />

Affan AM, Ali RA, Morton-Gittens JA, Perry A, Harry A, O’Donoghue<br />

JM, Rampaul R. St James Medical Complex, St James, POS, Trinidad<br />

and Tobago; Royal Victoria Infirmary, Newcastle-upon-Tyne, London,<br />

United Kingdom.<br />

P4-17-07 Reconstructive Outcomes of Nipple-Sparing Mastectomy: A<br />

Five Year Experience<br />

Guth AA, Blechman K, Samra F, Shapiro R, Axelrod D, Choi M, Karp N,<br />

Alperovich M. NYU School of Medicine, New York, NY.<br />

P4-17-08 Tissue Expander/Implant <strong>Breast</strong> Reconstruction with and<br />

without Postmastectomy Radiation: Predictive Factors for<br />

Complications<br />

Nguyen SKA, Oxley P, Rastegar R, Joffres M, Kwan W. British Columbia<br />

<strong>Cancer</strong> Agency, Fraser Valley <strong>Cancer</strong> Centre; University of British<br />

Columbia; Simon Fraser University.<br />

P4-17-09 Efficacy and safety of lipomodelling and adipose tissue derived<br />

degenerative stem cells(ADRC) for breast reconstruction –<br />

Medium term follow up<br />

Noor L, Bhaskar P, Hennessy C. University Hospital of North Tees,<br />

Cleveland, United Kingdom.<br />

9:00 am–9:30 am<br />

PLENARY LECTURE 3<br />

Exhibit Hall D<br />

<strong>Breast</strong> Radiotherapy: Fractionation and Other Fashions<br />

John Yarnold, MD<br />

Institute of <strong>Cancer</strong> Research<br />

Sutton, UNITED KINGDOM<br />

9:30 am–11:30 am<br />

GENERAL SESSION 5<br />

Exhibit Hall D<br />

Moderator: George W. Sledge, Jr., MD<br />

Indiana University Simon <strong>Cancer</strong> Center<br />

Indianapolis, IN<br />

9:30 S5-1. Biomarker analyses in CLEOPATRA: A phase III, placebocontrolled<br />

study of pertuzumab in HER2-positive, first-line<br />

metastatic breast cancer (MBC)<br />

Baselga J, Cortés J, Im S-A, Clark E, Kiermaier A, Ross G, Swain SM.<br />

Massachusetts General Hospital <strong>Cancer</strong> Center and Harvard Medical<br />

School, Boston, MA; Vall d’Hebron University Hospital, Barcelona,<br />

Spain; Seoul National University College of Medicine, Seoul, Korea; Roche<br />

Products Limited, Welwyn, United Kingdom; F. Hoffmann-La Roche<br />

Limited, Basel, Switzerland; MedStar Washington Hospital Center,<br />

Washington, DC.<br />

9:45 S5-2. HERA TRIAL: 2 years versus 1 year of trastuzumab after<br />

adjuvant chemotherapy in women with HER2-positive early breast<br />

cancer at 8 years of median follow up<br />

Goldhirsch A, Piccart-Gebhart MJ, Procter M, de Azambuja E, Weber HA,<br />

Untch M, Smith I, Gianni L, Jackisch C, Cameron D, Bell R, Dowsett M,<br />

Gelber RD, Leyland-Jones B, Baselga J, on behalf of the HERA Study Team<br />

NA. European Institute of Oncology, Milan, Italy; BrEAST Data Centre, Jules<br />

Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium; Frontier<br />

Science (Scotland) Ltd, Kincraig, Kingussie, United Kingdom; F Hoffmann-<br />

La Roche, Basel, Switzerland; Helios Klinikum Berlin-Buch, Akademisches<br />

LK der Universität Charité, Berlin, Germany; Royal Marsden Hospital and<br />

Institute of <strong>Cancer</strong> Research, London, United Kingdom; <strong>San</strong> Raffaele<br />

Institute, Milan, Italy; Klinikum Offenbach, Offenbach, Germany; University<br />

of Edinburgh, Western General Hospital, Edinburgh, United Kingdom;<br />

Geelong Hospital, Geelong, Australia; The Royal Marsden NHS Trust,<br />

London, United Kingdom; Dana-Farber <strong>Cancer</strong> Institute, Boston, MA;<br />

<strong>San</strong>ford Research, Sioux Falls, SD; Massachusetts General Hospital <strong>Cancer</strong><br />

Center, Boston, MA.<br />

www.aacrjournals.org<br />

53s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

10:00 S5-3. PHARE Trial results of subset analysis comparing 6 to 12<br />

months of trastuzumab in adjuvant early breast cancer<br />

Pivot X, Romieu G, Bonnefoi H, Pierga J-Y, Kerbrat P, Guastalla J-P,<br />

Lortholary A, Espié M, Fumoleau P, Khayat D, Pauporte I, Kramar A.<br />

University Hospital J. Minjoz, Besancon, France; Anti <strong>Cancer</strong> Center Val<br />

d’Aurelle, Montpellier, France; Anti <strong>Cancer</strong> Center Bergonie, Bordeaux,<br />

France; Curie Institute, Paris, France; Anti <strong>Cancer</strong> Center Eugene Marquis,<br />

Rennes, France; Anti <strong>Cancer</strong> Center Leon Berard, Lyon, France; Catherine<br />

de Sienne Center, Nantes, France; University Hospital St Louis, Paris,<br />

France; Anti <strong>Cancer</strong> Center Georges Francois Leclerc, Dijon, France;<br />

University Hospital Pitie Salpetriere, Paris, France; French National <strong>Cancer</strong><br />

Institut (INCa), France; Centre Oscar Lambret, Lille, France.<br />

10:15 S5-4. EGFR expression measured by quantitative<br />

immunofluorescense is associated with decreased benefit from<br />

trastuzumab in the adjuvant setting in the NCCTG (Alliance)<br />

N9831 trial<br />

Rimm D, Ballman KV, Cheng H, Vassilakopoulou M, Chen B, Gralow J,<br />

Hudis C, Davidson NE, Psyrri A, Fountzilas G, Perez EA. Yale University<br />

School of Medicine, New Haven, CT; Mayo Clinic, Rochester, MN; Seattle<br />

<strong>Cancer</strong> Care Alliance, Seattle, WA; Memorial Sloan-Kettering <strong>Cancer</strong><br />

Center, New York, NY; University of Pittsburgh <strong>Cancer</strong> Institute, Pittsburgh,<br />

PA; “Papageorgiou” Hospital, Aristotle University of Thessaloniki School<br />

of Medicine, Thessaloniki, Macedonia, Greece; Attikon University Hospital,<br />

Athens, Greece; Mayo Clinic, Jacksonville, FL.<br />

10:30 S5-5. Trastuzumab plus adjuvant chemotherapy for HER2-positive<br />

breast cancer: Final planned joint analysis of overall survival (OS)<br />

from NSABP B-31 and NCCTG N9831<br />

Romond E, Suman VJ, Jeong J-H, Sledge, Jr. GW, Geyer Jr. CE, Martino S,<br />

Rastogi P, Gralow J, Swain SM, Winer E, Colon-Otero G, Hudis C, Paik S,<br />

Davidson N, Mamounas EP, Zujewski JA, Wolmark N, Perez EA. National<br />

Surgical Adjuvant <strong>Breast</strong> and Bowel Project (NSABP) Operations and<br />

Biostatistical Centers; University of Kentucky; Mayo Clinic; University of<br />

Pittsburgh Graduate School of Public Health; IU Simon <strong>Cancer</strong> Center;<br />

University of Texas Southwestern Medical Center; The Angeles Clinic and<br />

Research Institute; University of Pittsburgh <strong>Cancer</strong> Institute; University of<br />

Washington; Medstar Washington Hospital Center; Dana-Farber <strong>Cancer</strong><br />

Institute; Memorial Sloan-Kettering <strong>Cancer</strong> Center; Aultman Hospital;<br />

<strong>Cancer</strong> Therapy Evaluation Program, National <strong>Cancer</strong> Institute, National<br />

Institutes of Health, D.H.H.S; Allegheny <strong>Cancer</strong> Center Allegheny General<br />

Hospital.<br />

10:45 S5-6. Activating HER2 mutations in HER2 gene amplification<br />

negative breast cancers<br />

Bose R, Kavuri SM, Searleman AC, Shen W, Shen D, Koboldt DC, Monsey<br />

J, Li S, Ding L, Mardis ER, Ellis MJ. Washington University School of<br />

Medicine, St. Louis, MO.<br />

11:00 S5-7. Combined blockade of PI3K/AKT and EGFR/HER3 enhances<br />

anti-tumor activity in triple negative breast cancer<br />

Tao J, Ip P, Auricchio N, Juric D, Yu M, Shyamala M, Kim P, Singh S,<br />

Hazra S, Haber D, Scaltriti M, Baselga J. Massachusetts General Hospital;<br />

Prometheus Lab.<br />

11:15 S5-8. Parallel upregulation of Bcl2 and estrogen receptor<br />

(ER) expression in HER2+ breast cancer patients treated with<br />

neoadjuvant lapatinib<br />

Giuliano M, Wang YC, Gutierrez C, Rimawi MF, Chang JC, Wang T,<br />

Hilsenbeck SG, Trivedi MV, Chamness GC, Osborne CK, Schiff R. Lester &<br />

Sue Smith <strong>Breast</strong> Center, Baylor College of Medicine, Houston, TX; Baylor<br />

College of Medicine, Houston, TX; The Methodist Hospital Research<br />

Institute, Houston, TX; University of Houston, Houston, TX.<br />

11:30 am–12:00 pm<br />

AACR Distinguished Lectureship in <strong>Breast</strong> <strong>Cancer</strong><br />

Research<br />

Exhibit Hall D<br />

Genes and the Microenvironment: The Twosome of Gene<br />

Expression and <strong>Breast</strong> <strong>Cancer</strong><br />

Mina J. Bissell, PhD<br />

Lawrence Berkeley National Laboratory<br />

Berkeley, CA<br />

12:00 pm–1:35 pm<br />

LUNCH<br />

12:30 pm–1:35 pm<br />

CASE DISCUSSION 2<br />

Ballroom A<br />

Moderator: Mothaffar Rimawi, MD<br />

Baylor College of Medicine<br />

Houston, TX<br />

Panelists:<br />

Dian Corneliussen-James<br />

METAvivor Research and Support<br />

Annapolis, MD<br />

Jennifer De Los <strong>San</strong>tos, MD<br />

University of Alabama at Birmingham<br />

Birmingham, AL<br />

Tari King, MD<br />

Memorial Sloan-Kettering <strong>Cancer</strong> Center<br />

New York, NY<br />

Hope Rugo, MD<br />

University of California <strong>San</strong> Francisco<br />

<strong>San</strong> Francisco, CA<br />

George W. Sledge, Jr., MD<br />

Indiana University Simon <strong>Cancer</strong> Center<br />

Indianapolis, IN<br />

12:30 pm–1:35 pm<br />

Basic Science Forum<br />

Ballroom B<br />

Molecular Imaging of <strong>Breast</strong> <strong>Cancer</strong>: Visualizing In Vivo <strong>Breast</strong><br />

<strong>Cancer</strong> Biology<br />

Moderator: David A. Mankoff, MD, PhD<br />

University of Pennsylvania Health System – PENN Medicine<br />

Philadelphia, PA<br />

Molecular imaging to characterize breast cancer models<br />

Lewis A. Chodosh, MD, PhD<br />

Perelman School of Medicine<br />

University of Pennsylvania<br />

Philadelphia, PA<br />

Molecular imaging for breast cancer patients<br />

David A. Mankoff, MD, PhD<br />

University of Pennsylvania Health System - PENN Medicine<br />

Philadelphia, PA<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012 54s <strong>Cancer</strong> Research


December 4–8, 2012<br />

Program Schedule<br />

1:45 pm–3:15 pm<br />

MINI-SYMPOSIUM 3<br />

Exhibit Hall D<br />

Snp’s - Germline Polymorphisms in <strong>Breast</strong> <strong>Cancer</strong> Susceptibility<br />

and Treatment Toxicity<br />

Moderator: Laura J. van ’t Veer, PhD<br />

University of California, <strong>San</strong> Francisco<br />

<strong>San</strong> Francisco, CA<br />

Germline polymorphisms and susceptibility to breast cancer<br />

Douglas Easton, PhD<br />

University of Cambridge<br />

Cambridge, UNITED KINGDOM<br />

Exploring germline variability as predictors for therapyinduced<br />

toxicity<br />

Bryan P. Schneider, MD, PhD<br />

Indiana University School of Medicine<br />

Indianapolis, IN<br />

Identifying the real promise of genomic medicine<br />

James P. Evans, MD, PhD<br />

University of North Carolina at Chapel Hill<br />

Chapel Hill, NC<br />

3:15 pm–5:00 pm<br />

GENERAL SESSION 6<br />

Exhibit Hall D<br />

Moderator: Suzanne Fuqua, PhD<br />

Baylor College of Medicine<br />

Houston, TX<br />

3:15 S6-1. Exploration of isoform switching and mutation expression in<br />

breast cancer by mRNA-sequencing analysis<br />

Hoadley KA, Parker JS, Wilkerson MD, Mose LE, Jefferys SR, Soloway MG,<br />

Turman YJ, Auman JT, Hayes DN, Perou CM. Lineberger Comprehensive<br />

<strong>Cancer</strong> Center, University of North Carolina at Chapel Hill, NC; University<br />

of North Carolina at Chapel Hill, Chapel Hill, NC.<br />

3:30 S6-2. Characterization of different foci of multifocal breast cancer<br />

using genomic, transcriptomic and epigenomic data<br />

Desmedt C, Nik-Zainal S, Fumagalli D, Rothé F, Singhal S, Majjaj S, Brown<br />

D, Dedeurwaerder S, Defrance M, Maetens M, Adnet P-Y, Vincent D,<br />

Salgado R, Fuks F, Piccart M, Larsimont D, Campbell P, Sotiriou C. Institut<br />

Jules Bordet, Brussels, Belgium; Wellcome Trust <strong>San</strong>ger Institute, Hinxton,<br />

United Kingdom; Université Libre de Bruxelles, Brussels, Belgium.<br />

3:45 S6-3. Neurocognitive impact in adjuvant chemotherapy for breast<br />

cancer linked to fatigue: A prospective functional MRI study<br />

Cimprich B, Hayes DF, Askren MK, Jung MS, Berman MG, Ossher L,<br />

Therrien B, Reuter-Lorenz PA, Zhang M, Peltier S, Noll DC. University of<br />

Michigan, Ann Arbor, MI; University of Washington, Seattle, WA; Rotman<br />

Research Institute at Baycrest, University of Toronto, Canada.<br />

4:00 S6-4. Vitamin D, but not bone turnover markers, predict relapse in<br />

women with early breast cancer: An AZURE translational study<br />

Coleman RE, Rathbone EJ, Marshall HC, Wilson C, Brown JE, Gossiel F,<br />

Gregory WM, Cameron D, Bell R. University of Leeds, United Kingdom;<br />

University of Sheffield, United Kingdom; University of Edinburgh, United<br />

Kingdom; Andrew Love <strong>Cancer</strong> Centre, Geelong, Australia.<br />

4:15 S6-5. Primary results of BEATRICE, a randomized phase III trial<br />

evaluating adjuvant bevacizumab-containing therapy in<br />

triple-negative breast cancer<br />

Cameron D, Brown J, Dent R, Jackisch C, Mackey J, Pivot X, Steger G, Suter<br />

T, Toi M, Parmar M, Bubuteishvili-Pacaud L, Henschel V, Laeufle R, Bell R.<br />

University of Edinburgh and <strong>Cancer</strong> Services, NHS Lothian, Edinburgh,<br />

United Kingdom; University of Leeds, United Kingdom; Sunnybrook<br />

Health Sciences Center and University of Toronto, Toronto, ON, Canada;<br />

Klinikum Offenbach, Offenbach, Germany; Cross Center Institute,<br />

Edmonton, Canada; University Hospital Jean Minjoz, Besançon, France;<br />

Medical University of Vienna, Austria; Bern University Hospital, Inselspital,<br />

Switzerland; Kyoto University, Kyoto, Japan; MRC Clinical Trials Unit,<br />

London, United Kingdom; F. Hoffmann-La Roche Ltd., Basel, Switzerland;<br />

Andrew Love <strong>Cancer</strong> Centre, Geelong, Australia; National <strong>Cancer</strong> Center,<br />

Singapore, Singapore.<br />

4:30 S6-6. A Phase III, open-label, randomized, multicenter study of<br />

eribulin mesylate versus capecitabine in patients with locally<br />

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

anthracyclines and taxanes<br />

Kaufman PA, Awada A, Twelves C, Yelle L, Perez EA, Wanders J, Olivo<br />

MS, He Y, Dutcus CE, Cortes. Norris Cotton <strong>Cancer</strong> Center, Dartmouth-<br />

Hitchcock Medical Center, Lebanon, NH; Jules Bordet Institute, Brussels,<br />

Belgium; Leeds Institute of Molecular Medicine and St James’s Institute<br />

of Oncology, Leeds, United Kingdom; University of Montreal, Montreal,<br />

Canada; Mayo Medical Clinic, Jacksonville, FL; Eisai Ltd., Hatfield, United<br />

Kingdom; Eisai Inc., Woodcliff Lake, NJ; Vall D’Hebron University Hospital,<br />

Barcelona, Spain.<br />

4:45 S6-7. Adaptive immune system and immune checkpoints are<br />

associated with response to pertuzumab (P) and trastuzumab (H)<br />

in the NeoSphere study<br />

Gianni L, Bianchini G, Valagussa P, Belousov A, Thomas M, Ross G, Pusztai<br />

L. <strong>San</strong> Raffaele Hospital - Scientific Institute, Milano, Italy; Fondazione<br />

Michelangelo, Milano, Italy; Roche Diagnostics GmbH, Penzberg,<br />

Germany; Roche Products Limited, Welwyn, United Kingdom; Yale School<br />

of Medicine, New Haven, CT.<br />

5:00 pm–7:00 pm<br />

Poster Discussion 9: DNA Repair<br />

Ballroom A<br />

Viewing 5:00 pm<br />

Discussion 5:15 pm<br />

Daniel Silver, MD, PhD, Chair and Discussant<br />

Dana-Farber <strong>Cancer</strong> Institute<br />

Boston, MA<br />

Shridar Ganesan, MD, PhD, Discussant<br />

The <strong>Cancer</strong> Institute of New Jersey<br />

New Brunswick, NJ<br />

PD09-01 BRCA1 inactivation induces NF-κB in human breast cancer cells<br />

and in murine and human mammary glands<br />

Sau A, Arnaout A, Pratt C. University of Ottawa, ON, Canada; Ottawa<br />

Hospital, Ottawa, ON, Canada.<br />

PD09-02 BRCA1 insufficiency is predictive of superior survival in<br />

patients with triple negative breast cancer treated with<br />

platinum based chemotherapy<br />

Sharma P, Stecklein S, Kimler BF, Klemp JR, Khan QJ, Fabian CJ, Tawfik<br />

OW, Connor CS, McGinness MK, Mammen JMW, Jensen RA. University<br />

of Kansas Medical Center, Westwood, KS; University of Kansas Medical<br />

Center, Kansas City, KS.<br />

www.aacrjournals.org<br />

55s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

PD09-03<br />

PD09-04<br />

PD09-05<br />

PD09-06<br />

PD09-07<br />

PD09-08<br />

PD09-09<br />

PD09-10<br />

Impact of BRCA1/2 mutation status in TBCRC009: A multicenter<br />

phase II study of cisplatin or carboplatin for metastatic triple<br />

negative breast cancer<br />

Isakoff SJ, Goss PE, Mayer EL, Traina T, Carey LA, Krag KJ, Liu MC,<br />

Rugo H, Stearns V, Come S, Finkelstein D, Hartman A-R, Garber JE,<br />

Ryan PD, Winer EP, Ellisen LW. Massachusetts General Hospital,<br />

Boston, MA; Dana-Farber <strong>Cancer</strong> Institute, Boston, MA; Memorial<br />

Sloan-Kettering <strong>Cancer</strong> Center, New York, NY; University of North<br />

Carolina Lineberger Comprehensive <strong>Cancer</strong> Center, Chapel Hill, NC;<br />

Mass General/North Shore <strong>Cancer</strong> Ctr, Danvers, MA; Georgetown<br />

Lombardi Comprehensive <strong>Cancer</strong> Center, Washington, DC; University<br />

of California, <strong>San</strong> Francisco Helen Diller Family Comprehensive<br />

<strong>Cancer</strong> Center, <strong>San</strong> Francisco, CA; Sidney Kimmel Comprehensive<br />

<strong>Cancer</strong> Center at Johns Hopkins University, Baltimore, MD; Beth Israel<br />

Deaconess Medical Center, Boston, MA; Myriad Genetics, Salt Lake<br />

City, UT; Fox Chase <strong>Cancer</strong> Center, Philadelphia, PA.<br />

Homologous Recombination Deficiency (HRD) score predicts<br />

pathologic response following neoadjuvant platinum-based<br />

therapy in triple-negative and BRCA1/2 mutation-associated<br />

breast cancer (BC)<br />

Telli ML, Jensen KC, Abkevich V, Hartman A-R, Vinayak S, Lanchbury<br />

J, Gutin A, Timms K, Ford JM. Stanford University School of Medicine,<br />

Stanford, CA; Myriad Genetics, Salt Lake City, UT.<br />

Single nucleotide polymorphism of XRCC1 which participates<br />

in DNA repair mechanism predicts clinical outcome in<br />

relapsed or metastatic breast cancer patients treated with<br />

S1 and oxaliplatin chemotherapy: Results from multicenter<br />

prospective study (TORCH_KCSG BR07-03)<br />

Im S-A, Oh D-Y, Keam B, Lee KS, Ahn J-H, Sohn J, Ahn JS, Kim JH,<br />

Lee MH, Lee KE, Kim HJ, Lee K-H, Han SW, Kim S-Y, Kim SB, Im Y-H,<br />

Ro J, Park H-S. Seoul National University Hospital, Seoul, Korea;<br />

National <strong>Cancer</strong> Center, Goyang, Korea; Asan Medical Center, Seoul,<br />

Korea; Yonsei University College of Medicine, Severance Hospital,<br />

Seoul, Korea; Samsung Medical Center, Seoul, Korea; Seoul National<br />

University Bundang Hospital, Seongnam, Korea; Inha University<br />

Hospital, Incheon, Korea; Ewha Womans University Medical Center,<br />

Seoul, Korea; Hallym University Sacred Heart Hospital, Anyang, Korea;<br />

Kyung-Hee University Hospital, Seoul, Korea; Soon Chun Hyang<br />

University Hospital, Seoul, Korea.<br />

Two phase I trials exploring different dosing schedules<br />

of carboplatin (C), paclitaxel (P), and the poly-ADP-ribose<br />

polymerase (PARP) inhibitor, veliparib (ABT-888) (V) with<br />

activity in triple negative breast cancer (TNBC)<br />

Puhalla SL, Appleman LJ, Beumer JH, Tawbi H, Stoller RG, Owonikoko<br />

TK, Ramalingam SS, Belani CP, Brufsky AM, Abraham J, Shepherd SP,<br />

Giranda V, Chen AP, Chu E. University of Pittsburgh <strong>Cancer</strong> Institute,<br />

Pittsburgh, PA; Winship <strong>Cancer</strong> Institute of Emory University, Atlanta,<br />

GA; West Virginia University <strong>Cancer</strong> Center, Morgantown, WV; Penn<br />

State Hershey <strong>Cancer</strong> Institute, Hershey, PA; National <strong>Cancer</strong> Institute,<br />

Bethesda, MD; Abbott Laboratories, Abbott Park, IL.<br />

Therapeutic potential of targeting ATM-PELP1-p53 axis in<br />

triple negative breast cancer<br />

Krishnan SR, Nair BC, Sareddy GR, Mann M, Roy SS, Vadlamudi RK.<br />

UTHSCSA, <strong>San</strong> <strong>Antonio</strong>, TX.<br />

The potential role of TLK2 as a therapeutic target in<br />

breast cancer<br />

Kim J-A, Cao X, Tan Y, Schiff R, Wang X. Lester & Sue Smith <strong>Breast</strong><br />

Center, Baylor College of Medicine, Houston, TX.<br />

The Akt inhibitor MK-2206 is an effective radio-sensitizer of<br />

p53 deficient triple negative breast cancer (TNBC) cells<br />

Connolly EP, Sun Y, Chao KC, Hei TK. Columbia University,<br />

New York, NY.<br />

DNA damage and repair in mammary gland development<br />

Kass EM, Helgadottir H, Moynahan ME, Jasin M. Memorial Sloan-<br />

Kettering <strong>Cancer</strong> Center, New York, NY.<br />

5:00 pm–7:00 pm<br />

Poster Discussion 10: Prognostic/Predictive<br />

Ballroom B<br />

Viewing 5:00 pm<br />

Discussion 5:15 pm<br />

Matthew Goetz, MD, Chair<br />

Mayo Clinic College of Medicine<br />

Rochester, MN<br />

Dennis C. Sgroi, MD, Discussant<br />

Massachusetts General Hospital<br />

Charlestown, MA<br />

and<br />

Hiltrud Brauch, PhD, Discussant<br />

Dr. Margarete Fischer-Bosch –<br />

Institute of Clinical Pharmacology<br />

Stuttgart, GERMANY<br />

PD10-01 Rescheduled as S1-10<br />

PD10-02 Metabolic syndrome and recurrence within the 21-gene<br />

recurrence score assay risk categories in lymph node negative<br />

breast cancer<br />

Lakhani A, Guo R, Duan X, Ersahin C, Gaynor ER, Godellas C, Kay C, Lo<br />

SS, Mai H, Perez C, Albain K, Robinson P. Loyola University Medical<br />

Center, Maywood, IL.<br />

PD10-03 Predictive value of a proliferation score (MS) in<br />

postmenopausal women with endocrine-responsive breast<br />

cancer: results from International <strong>Breast</strong> <strong>Cancer</strong> Study Group<br />

(IBCSG) Trial IX<br />

Sninsky J, Wang A, Gray K, Lagier R, Christopherson C, Rowland C,<br />

Chang M, Kammler R, Viale G, Kwok S, Regan M, Leyland-Jones B.<br />

Celera, Alameda, CA; Dana-Farber <strong>Cancer</strong> Institute, Boston, MA;<br />

IBCSG Coordinating Center, Berne, Switzerland; European Institute of<br />

Oncology, Milan, Italy; <strong>San</strong>ford Research, Sioux Falls, SD.<br />

PD10-04 Predictive genomic markers to chemotherapy and adjuvant<br />

trastuzumab via whole genome expression DASL profiling in the<br />

N9831 adjuvant study<br />

Perez EA, Eckel-Passow JE, Ballman KV, Anderson SK, Thompson<br />

EA, Asmann YW, Jen J, Dueck AC, Lingle WL, Sledge GW, Winer EP,<br />

Gralow J, Jenkins RB, Reinholz MM. Mayo Clinic, Jacksonville, FL; Mayo<br />

Clinic, Rochester, MN; Mayo Clinic, Scottsdale, AZ; Indiana University<br />

Simon <strong>Cancer</strong> Center, Indianapolis, IN; Dana Farber <strong>Cancer</strong> Institute,<br />

Boston, MA; Seattle <strong>Cancer</strong> Care Alliance, Seattle, WA; Ventana<br />

Medical Systems, Inc., Tuscon, AZ.<br />

PD10-05 HLA-DQA1*02:01/DRB1*07:01 as a biomarker for lapatinibinduced<br />

hepatotoxicity: prospective confirmation in a large<br />

randomised clinical trial (TEACH, EGF105485)<br />

Spraggs CF, Schaid DJ, Parham LR, McDonnell SK, Briley LP, King<br />

KS, Rappold E, Goss PE. GlaxoSmithKline, Stevenage, Hertfordshire,<br />

United Kingdom; GlaxoSmithKline, Raleigh, NC; Mayo Clinic,<br />

Rochester, MN; GlaxoSmithKline, Collegeville, PA; Massachussets<br />

General Hospital, Boston, MA.<br />

PD10-06 CXCL13 mRNA predicts docetaxel benefit in triple<br />

negative tumors<br />

Wirtz RM, Leinonen M, Bono P, Isola J, Kellokumpu-Lehtinen P-L,<br />

Kataja V, Turpeenniemi-Hujanen T, Jyrkkiö S, Eidt S, Schmidt M,<br />

Joensuu H. STRATIFYER Molecular Pathology GmbH, Cologne,<br />

Germany; Pharma, Turku, Finland; Helsinki University Central Hospital<br />

and University of Helsinki, Helsinki, Finland; University of Tampere and<br />

Tampere University Hospital, Finland; Tampere University Hospital,<br />

Tampere, Finland; Kuopio University Hospital, Kuopio, Finland; Oulu<br />

University Hospital, Oulu, Finland; Turku University Hospital, Turku,<br />

Finland; Institute of Pathology at the St-Elisabeth-Hospital, Cologne,<br />

Germany; University Hospital Mainz, Germany.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012 56s <strong>Cancer</strong> Research


December 4–8, 2012<br />

Program Schedule<br />

PD10-07 Patients carrying CYP2C8*3 are at increased risk of<br />

paclitaxel-induced neuropathy<br />

Hertz DL, Dees EC, Roy S, Motsinger-Reif AA, Drobish A, Clark LS,<br />

McLeod HL, Carey LA. University of North Carolina at Chapel Hill,<br />

NC; Lineberger Comprehensive <strong>Cancer</strong> Center, University of North<br />

Carolina at Chapel Hill, NC; North Carolina State University, Raleigh,<br />

NC; Gentris Corp., Morrisville, NC.<br />

PD10-08 Venlafaxine inhibits the CYP2D6 mediated metabolic<br />

activation of tamoxifen: Results of a prospective multicenter<br />

study: (NCT00667121)<br />

Goetz MP, Suman V, Henry NL, Reid J, Safgren S, Kosel M, Kuffel M,<br />

Sideras K, Flockhart D, Stearns V, Denduluri N, Irvin WJ, Ames M. Mayo<br />

Clinic, Rochester, MN; University of Michigan, Ann Arbor, MI; Indiana<br />

University, Indianapolis, IN; Johns Hopkins, Baltimore, MD; Fairfax-<br />

Northern Virginia Hematology-Oncology, Arlington, VA; University of<br />

North Carolina, Chapel Hill, NC.<br />

PD10-09 CYP2D6 and adjuvant tamoxifen: Impact on outcome in<br />

pre- but not postmenopausal breast cancer patients<br />

Margolin S, Lindh J, Thorén L, Xie H, Koukel L, Dahl M-L, Eliasson<br />

E. Karolinska Institutet, Stockholm, Sweden; Karolinska University<br />

Hospital, Stockholm, Sweden; Karolinska Institutet, Karolinska<br />

University Hospital Huddinge, Stockholm, Sweden.<br />

5:00 pm–7:00 pm<br />

POSTER SESSION 5 & RECEPTION<br />

Exhibit Halls A-B<br />

Detection/Diagnosis: Diagnostic Pathology<br />

P5-01-01 Predicting OncoDX Recurrence Scores with<br />

Immunohistochemical Markers<br />

Bradshaw SH, Gravel DH, Song X, Marginean EC, Robertson SJ. The<br />

Ottawa Hospital, Ottawa, ON, Canada.<br />

P5-01-02 Inter-observer concordance of Ki-67 labeling index in breast<br />

cancer: Japan <strong>Breast</strong> <strong>Cancer</strong> Research Group (JBCRG) Ki-67<br />

Ring Study<br />

Ueno T, Mikami Y, Yoshimura K, Tsuda H, Kurosumi M, Masuda S,<br />

Horii R, Toi M, Sasano H. Kyoto University Hospital, Kyoto, Japan;<br />

National <strong>Cancer</strong> Center Hospital, Tokyo, Japan; Saitama <strong>Cancer</strong><br />

Center, Saitama, Japan; Nihon University School of Medicine, Tokyo,<br />

Japan; The <strong>Cancer</strong> Institute Hospital of the Japanese Foundation for<br />

<strong>Cancer</strong> Research, Tokyo, Japan; Tohoku University School of Medicine,<br />

Sendai, Japan.<br />

P5-01-03 Discordant Estrogen and Progesterone Receptor Status in<br />

<strong>Breast</strong> <strong>Cancer</strong><br />

Hefti MM, Hu R, Knoblauch N, Collins L, Tamimi RM, Beck AH. Beth<br />

Israel Deaconess Medical Center, Boston, MA; Brigham and Women’s<br />

Hospital, Boston, MA.<br />

P5-01-04 Clinico-pathological features of low-grade triple negative early<br />

breast cancers<br />

Nourieh M, Furhmann L, Feron J-G, Caly M, Diéras V, Sastre-Garau X,<br />

Chavrier P, Vincent-Salomon A. Institut Curie, Paris, France.<br />

P5-01-05 Could C-myc amplification replace Cahan’s criteria to<br />

discriminate secondary from primary angiosarcoma<br />

of the breast?<br />

Laé M, Hamel F, Hadj-Hamou S, Malfoy B, Kirova YM. Institut Curie,<br />

Paris, France.<br />

P5-01-06 Differences in FGF1 and FGFR2 expression in BRCA1-<br />

associated, BRCA2-associated, and sporadic breast carcinomas<br />

Vos S, van der Wall E, van Diest PJ, van der Groep P. University<br />

Medical Center Utrecht, Netherlands.<br />

P5-01-07 Fibroadenomatoid changes are more prevalent in<br />

middle-aged women and have a positive association with<br />

invasive breast cancer<br />

Chen Y, Bekhash A, Kovatich AJ, Hooke JA, Kvecher L, Mitchell EP, Rui<br />

H, Mural RJ, Shriver CD, Hu H. Windber Research Institute, Windber,<br />

PA; Walter Reed National Military Medical Center, Bethesda, MD;<br />

MDR, Global Systems LLC, Windber, PA; Thomas Jefferson University,<br />

Philadelphia, PA.<br />

P5-01-08 Complex fibroadenoma is not an independent risk marker for<br />

breast cancer<br />

Nassar A, Visscher DW, Degnim AC, Frank RD, Vierkant RA, Hartmann<br />

LC, Frost MH, Ghosh K. Mayo Clinic, Rochester, MN.<br />

P5-01-09 Identification of Molecular Apocrine Triple Negative <strong>Breast</strong><br />

<strong>Cancer</strong> Using a Novel 2-Gene Assay and Comparison with<br />

Androgen Receptor Protein Expression and Gene Expression<br />

Profiling by DASL<br />

Alvarez J, Schaffer M, Karkera J, Martinez G, Gaffney D, Bell K, Sharp M,<br />

Wong J, Hertzog B, Ricci D, Platero S. Janssen Pharmaceuticals.<br />

P5-01-10 Clinical-pathological features and outcomes of Invasive lobular<br />

(ILC) vs Invasive ductal (IDC) breast cancer (BC): a monoinstitutional<br />

series<br />

Ferro A, Eccher C, Triolo R, Caldara A, Di Pasquale MC, Russo LM, De<br />

Carli NL, Cuorvo LV, Barbareschi M, Gasperetti F, Berlanda G, Pellegrini<br />

M, Moroso S, Galligioni E. S Chiara Hospital, Trento, Italy; FBK,<br />

Trento, Italy.<br />

P5-01-11 Same-day diagnosis’ of women suspected of breast cancer:<br />

success rate and impact on diagnostic quality and patients’<br />

anxiety levels<br />

Barentsz MW, Wessels H, van Diest PJ, Pijnappel RM, van der Pol CC,<br />

Witkamp AJ, van den Bosch MA, Verkooijen HM. University Medical<br />

Center Utrecht.<br />

P5-01-12 Over- and Undergrading of <strong>Breast</strong> <strong>Cancer</strong> on Core Biopsies in<br />

Comparison to Surgical Specimens<br />

Decker T, Focke CM, Gläser D, Finsterbusch K. Bonhoeffer Medical<br />

Center, Neubrandenburg, Germany.<br />

P5-01-13 Flat Epithelial Atypia: Management and outcome in three<br />

Dutch teaching hospitals<br />

Ghuijs PM, de Vries B, Strobbe LJA, van Deurzen CHM, Heuts<br />

EM, Keymeulen KBMI, Lobbes MBI, Wauters CAP, Van de Vijver<br />

KKBT, Smidt ML. Maastricht University Medical Centre, Maastricht,<br />

Netherlands; Canisius-Wilhelmina Hospital, Nijmegen, Netherlands;<br />

Erasmus Medical Centre, Rotterdam, Netherlands.<br />

P5-01-14 Discrepancy between routine and expert pathologists in<br />

histological assessment of early stage non palpable breast<br />

cancer and its impact on loco regional and systemic treatment<br />

Postma EL, Verkooijen HM, van Diest PJ, Willems SM, van den Bosch<br />

MA, van Hillegersberg R. UMC Utrecht.<br />

P5-01-15 Anti-ER monoclonal antibody SP1 seems more sensitive in the<br />

identification of ER alpha positive breast cancer cases than<br />

anti-ER monoclonal antibody 1D5<br />

Madeira KP, Daltoé RD, Sirtoli GM, Rezende LCD, Carvalho AA, Silva IV,<br />

Rangel LBA. Universidade Federal do Espírito <strong>San</strong>to, Vitória, ES, Brazil.<br />

P5-01-16 Value of ultrasonography appearance of breast mass in<br />

prediction of histologic type<br />

Xu Z, Sun L, Yang H, Song Y, Sun G. JiLin Province <strong>Breast</strong> Dieases<br />

Institue, Changchun, JiLin, China.<br />

Detection/Diagnosis: Detection/Diagnosis - Other<br />

P5-02-01 Discrepancy between CT and FDG-PET/CT in the staging of<br />

patients with inflammatory breast cancer: Implications for<br />

radiation therapy treatment planning<br />

Jacene H, DiPiro P, Bellon J, Nakhlis F, Hirshfield-Bartek J, Yeh E,<br />

Overmoyer B. Dana-Farber <strong>Cancer</strong> Institute, Harvard Medical School,<br />

Boston, MA.<br />

www.aacrjournals.org<br />

57s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P5-02-02 Factors influencing time to seeking medical advice and<br />

start of treatment in breast cancer (BC) patients – an<br />

International survey<br />

Jassem J, Ozmen V, Bacanu F, Drobniene M, Eglitis J, Kahan Z,<br />

Lakshmaiah K, Mardiak J, Pienkowski T, Semiglazova T, Stamatovic<br />

L, Timcheva C, Vasovics S, Vrbanec D, Zaborek P. Medical University<br />

of Gdansk, Poland; University of Istanbul, Turkey; Sf Maria Hospital,<br />

Bucharest, Romania; Institute of Oncology, Vilnius University, Vilnus,<br />

Lithuania; Riga East University Hospital, Riga, Latvia; University of<br />

Szeged, Hungary; Kidwai Memorial Institute of Oncology, Bangaluru,<br />

India; National <strong>Cancer</strong> Institute and Medical School of Comenius<br />

University, Bratislava, Slovakia (Slovak Republic); Medical Center<br />

of Postgraduate Education, ECZ, Otwock, Poland; Petrov Research<br />

Institute of Oncology, St. Petersburg, Russian Federation; Institute<br />

of Oncology and Radiology, Belgrade, Serbia; Chemiotherapy Clinic,<br />

Sofia, Bulgaria; Zagreb University Hospital Center, Zagreb, Croatia;<br />

Warsaw School of Economics, Warsaw, Poland.<br />

P5-02-03 Large-scale genomic instability consistently identifies BRCA1/2<br />

inactivation in breast cancers<br />

Stern M-H, Popova T, Manié E, Dubois T, Sigal-Zafrani B, Bollet M,<br />

Sastre-Garau X, Vincent-Salomon A, Houdayer C, Stoppa-Lyonnet D.<br />

Institut Curie, Paris, France.<br />

Tumor Cell and Molecular Biology: Stem/Progenitor Cells<br />

P5-03-01 <strong>Cancer</strong> stem cells predict engraftment and poor prognosis of<br />

primary breast tumors<br />

Charaffe-Jauffret E, Ginestier C, Bertucci F, Cabaud O, Wicinski J,<br />

Finetti p, Josselin E, Adelaide J, Nguyen T-T, Monville F, Jacquemier J,<br />

Thomassin-Piana J, Pinna G, Jalaguier A, Lambaudie E, Houvenaeghel<br />

G, Xerri l, Harel-bellan A, Chaffanet M, Viens P, Birnbaum D. CRCM-<br />

IPC; CEA.<br />

P5-03-02 Targeting mRNA Translation To Enhance the Radiosensitivity<br />

of Inflammatory <strong>Breast</strong> <strong>Cancer</strong> Stem Cells<br />

Silvera D, Connolly EP, Volta V, Arju R, Venuto T, Schneider RJ. NYU<br />

School of Medicine, New York, NY; NYU <strong>Cancer</strong> Institute, NYU School<br />

of Medicine, New York, NY; Columbia University College of Physicians<br />

and Surgeons, New York, NY.<br />

P5-03-03 Antitumor Activity and <strong>Cancer</strong> Stem Cells Effect of Cetuximab<br />

in Combination with Ixabepilone in Triple Negative <strong>Breast</strong><br />

<strong>Cancer</strong>s (TNBC)<br />

Tanei T, Rodriguez AA, Dobrolecki L, Choi DS, Landis M, Chang JC.<br />

The Methodist Hospital Research Institute and Weill Cornell Medical<br />

School, Houston, TX.<br />

P5-03-04 Effect of aging on the function and transformation of murine<br />

mammary stem cells<br />

Bandyopadhyay A, Dong Q, Wang D, Gao H, Wu A, Yeh I-T, Sun L.<br />

University of Texas Health Science Center, <strong>San</strong> <strong>Antonio</strong>, TX; Virginia<br />

Hospital Center, Arlington, VA.<br />

P5-03-05 Histone deacetylase (HDAC)-inhibitor mediated<br />

reprogramming drives cancer cells to the pentose phosphate<br />

metabolic pathway<br />

Debeb BG, Larson RA, Lacerda L, Xu W, Smith DL, Ueno NT, Reuben<br />

JM, Gilcrease M, Krishnamurthy S, Buchholz TA, Woodward WA. MD<br />

Anderson <strong>Cancer</strong> Center, Houston, TX.<br />

P5-03-06 Bisphenol A and mammary stem cells: implications in breast<br />

cancer susceptibility<br />

Dong Q, Wang D, Gao H, Bandyopadhyay A, Wu A, Yeh I-T, Huang<br />

C, Sun L. University of Texas Health Science Center at <strong>San</strong> <strong>Antonio</strong>,<br />

TX; Wenzhou Medical College, Wenzhou, Zhejiang, China; Virginia<br />

Hospital Center, Arlington, VA.<br />

P5-03-07 Targeting Hedgehog pathway to reverse chemoresistance in<br />

breast cancer stem cells<br />

Mañu A, Fresquet V, Mena M, Sánchez S, Espinós J, Fernandez<br />

Hidalgo OA, Fernández-Zapico M, Knutson KL, Martínez-Climent JA,<br />

<strong>San</strong>tisteban M. Clinic University of Navarra, Pamplona, Navarra, Spain;<br />

Foundation for Applied Medical Research, Pamplona, Navarra, Spain;<br />

Mayo Clinic, Rochester, MN.<br />

P5-03-08 A fluorescence STAT3 reporter preferentially expressed in<br />

human breast cancer tumor-initiating cells<br />

Wei W, Tweardy D, Zhang M, Roarty K, Rosen J, Lewis M. Lester and<br />

Sue Smith <strong>Breast</strong> Center, Baylor College of Medicine, Houston, TX.<br />

P5-03-09 Receptor Activator of Nuclear Factor Kappa B (RANK) as a<br />

potential therapeutic target in triple-negative breast cancer<br />

Reyes ME, Masuda H, Zhang D, Reuben JM, Woodward W, Darnay BG,<br />

Hortobagyi GN, Ueno NT. MD Anderson <strong>Cancer</strong> Center, Houston, TX.<br />

P5-03-10 HIF-1alpha knockout radiosensitizes select Inflammatory<br />

<strong>Breast</strong> <strong>Cancer</strong> cells through reduction of stem-like cancer cells<br />

Xu W, Debeb BG, Smith DL, Li JL, Ueno NT, Alvarez de Lacerda LC,<br />

Larson RA, Schwba LP, Seagroves TN, Woodward WA. MD Anderson<br />

<strong>Cancer</strong> Center, Houston, TX; U of Tennessee Health Science Center,<br />

Memphis, TN.<br />

P5-03-11 Possible role for cancer stem cells: results from a pilot<br />

neoadjuvant trial of HER-2 positive breast cancer patients<br />

treated with a combination of (Nab)-paclitaxel and lapatinib<br />

Siziopikou KP, Gradishar WJ, Kaklamani VG. Northwestern University<br />

Feinberg School of Medicine, Chicago, IL.<br />

P5-03-12 Targeting breast cancer stem cells using the autophagy<br />

inhibitor N-Acetyl cysteine<br />

Dave B, Granados S, Mitra S, Chang JC. Methodist Hostpital Research<br />

Institute, Houston, TX.<br />

P5-03-13 Detection of tumor initiating cells (TIC) among the peripherally<br />

circulating epithelial tumor cells from patients with breast<br />

cancer<br />

Pachmann K, Zimon D, Pizon M, Carl S, Rabenstein C, Camara O,<br />

Pachmann U. University Hospital Jena, Germany; Transfusion Center,<br />

Bayreuth, Germany.<br />

P5-03-14 Expression of ALDH1 in metastasizing axillary lymphnodes in<br />

breast cancer<br />

Shi A, Dong Y, Bi L, Xu N, Fan Z, Li S, Yang H, Li Y. First Hospital of<br />

Bethune Medical College, Jilin University, Changchun, Jilin, China;<br />

Lester & Sue Smith <strong>Breast</strong> Center, Baylor College of Medicine,<br />

Houston, TX.<br />

Tumor Cell and Molecular Biology: Epithelial-Mesenchymal Transition<br />

P5-04-01 Expression of epithelial-mesenchymal transition (EMT)-<br />

related markers in primary tumors and matched lymph node<br />

metastases in breast cancer patients<br />

Zaczek AJ, Ahrends T, Markiewicz A, Seroczynska B, Szade J, Welnicka-<br />

Jaskiewicz M, Jassem J. Intercollegiate Faculty of Biotechnology,<br />

University of Gdansk and Medical University of Gdansk, Gdansk,<br />

Poland; Medical University of Gdansk, Pomorskie, Poland.<br />

P5-04-02 DYRK2 regulates breast cancer invasion via Snail/E-cadherin<br />

pathway<br />

Mimoto R, Imawari Y, Kamio M, Kato K, Nogi H, Toriumi Y, Takeyama<br />

H, Yoshida K, Uchida K. The Jikei University School of Medicine,<br />

Tokyo, Japan.<br />

P5-04-03 Epithelial-mesenchymal transition is associated with in situ to<br />

invasive transition of basal-like breast cancer<br />

Park SY, Choi Y, Kim EJ, Lee HE, Lee HJ, Kang E, Kim S-W. Seoul<br />

National University College of Medicine, Seoul, Republic of Korea;<br />

Seoul National University Bundang Hospital, Seongnam,<br />

Republic of Korea.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012 58s <strong>Cancer</strong> Research


December 4–8, 2012<br />

Program Schedule<br />

P5-04-04 Significance of PELP1/HDAC2/microRNA-200 regulatory<br />

network in EMT and metastasis of breast cancer<br />

Roy SS, Gonugunta VK, Bandyopadhyay AM, Rao M, Goodall G, Sun<br />

L, Tekmal RR, Vadlamudi RK. UTHSCSA, <strong>San</strong> <strong>Antonio</strong>, TX; Centre for<br />

<strong>Cancer</strong> Biology, Adelaide, Australia.<br />

P5-04-05 E-Cadherin Is Required for In Vivo Growth of Inflammatory<br />

<strong>Breast</strong> <strong>Cancer</strong>: Importance of Mesenchymal-Epithelial<br />

Transition (MET) and Role of HIF1α<br />

Chu K, Boley KM, Cristofanilli M, Robertson FM. The University of<br />

Texas MD Anderson <strong>Cancer</strong> Center, Houston, TX; Fox Chase <strong>Cancer</strong><br />

Center, Philadelphia, PA.<br />

P5-04-06 Soluble factors from activated immune cells induce epithelial<br />

mesenchymal transition in inflammatory breast cancer cells<br />

Cohen EN, Gao H, Anfossi S, Giordano A, Tin S, Wu Q, Lee B-N, Luthra<br />

R, Krishnamurthy S, Hortobagyi GN, Ueno NT, Woodward WA, Reuben<br />

JM. The University of Texas MD Anderson <strong>Cancer</strong> Center, Houston, TX;<br />

The University of Texas at Houston Health Science Center, Houston,<br />

TX; The Morgan Welch Inflammatory <strong>Breast</strong> <strong>Cancer</strong> Research Program<br />

and Clinic, The University of Texas MD Anderson <strong>Cancer</strong> Center,<br />

Houston, TX.<br />

P5-04-07 Epithelial-mesenchymal transition phenotype in breast<br />

cancers is associated with clinicopathologic factors indicating<br />

aggressive biologic behavior and poor clinical outcome<br />

Bae YK, Kim A, Choi JE, Kang SH, Lee SJ. Yeungnam University College<br />

of Medicine, Daegu, Korea.<br />

P5-04-08 Identifying and characterizing human kinases for novel<br />

regulators of epithelial-mesenchymal transition in breast<br />

cancer cells<br />

Li L, Azizian N, Li W. Brown Foundation Institute of Molecular<br />

Medicine, University of Texas Health Science Center at Houston, TX.<br />

P5-04-09 Squamous Cell Carcinoma Antigen 1 (SCCA1) Upregulation<br />

Causes Epithelial to Mesenchymal Transition (EMT) and<br />

Promotes Mammary Tumorigenesis<br />

Sheshadri N, Ullman E, Catanzaro J, Chen E, Zong W-X. Stony Brook<br />

University, Stony Brook, NY.<br />

Tumor Cell and Molecular Biology: Systems Biology of <strong>Breast</strong> <strong>Cancer</strong><br />

P5-05-01 Catalogued phospho-sensing peptides identifying active,<br />

oncogenic kinase signatures<br />

Mori M, Chen Z, Boudreau A, van’t Veer L, Coppé J-P. University of<br />

California, <strong>San</strong> Francisco, CA; Kinogea Inc., Shanghai, China; Lawrence<br />

Berkeley National Laboratory, Berkeley, CA.<br />

P5-05-02 The role of cellular heterogeneity on the therapeutic response<br />

of breast cancer: Clinical insights from a hybrid multiscale<br />

computational model<br />

Powathil GG, Thompson A, Chaplain MAJ. University of Dundee,<br />

Scotland, United Kingdom; Dundee <strong>Cancer</strong> Centre, University of<br />

Dundee, Scotland, United Kingdom.<br />

P5-05-03 The 5p12 breast cancer susceptibility locus is associated with<br />

MRPS30 expression in estrogen receptor - positive tumors<br />

Quigley DA, Van Loo P, Alnæs GG, Tost J, Zelenika D, Balmain A,<br />

Børresen-Dale A-L, Kristensen VN. Institute for <strong>Cancer</strong> Research, Oslo<br />

University Hospital, Oslo, Norway; Wellcome Trust <strong>San</strong>ger Institute,<br />

Hinxton, United Kingdom; Helen Diller Family Comprehensive <strong>Cancer</strong><br />

Center, University of California, <strong>San</strong> Francisco, CA; CEA - Institut de<br />

Génomique, Evry, France.<br />

P5-05-04 Intraductal delivery of RNAi-based therapeutics to gene targets<br />

identified through computational systems modeling<br />

Brock A, Krause S, Li H, Kowalski M, Collins J, Ingber D. Wyss Institute,<br />

Harvard University, Boston, MA; Boston Children’s Hospital,<br />

Boston, MA.<br />

P5-05-05 Computational modeling of breast cancer growth and spread:<br />

the role of matrix degrading enzymes<br />

Deakin NE, Thompson AM, Chaplain MAJ. University of Dundee,<br />

United Kingdom; Dundee <strong>Cancer</strong> Centre, University of Dundee,<br />

United Kingdom.<br />

P5-05-06 Spatio-temporal computational modeling of the p53-Mdm2<br />

pathway<br />

Sturrock M, Thompson AM, Chaplain MAJ. University of Dundee,<br />

United Kingdom; Dundee <strong>Cancer</strong> Centre, University of Dundee,<br />

United Kingdom.<br />

Tumor Cell and Molecular Biology: Cell Cycle Regulation<br />

P5-06-01 Active pak6 induces aneuploidy in breast cancer cells<br />

Paul BA, Lu ML. University of Miami Miller School of Medicine, Miami,<br />

FL; Florida Atlantic University, Boca Raton, FL.<br />

P5-06-02 The Role of <strong>Breast</strong> Tumor Kinase (Brk) in Cell Cycle Control<br />

Nimnual AS, Chan E. Stony Brook University, Stony Brook, NY.<br />

Tumor Cell and Molecular Biology: Cellular Mechanisms<br />

P5-07-01 Defining the mechanism of breast cancer growth by<br />

chemokine receptor CXCR7 and Epidermal Growth Factor<br />

Receptor coupling interaction<br />

Salazar N, Muñoz D, Singh RK, Lokeshwar BL. University of Miami<br />

Miller School of Medicine, Miami, FL; Miami VA Medical Center,<br />

Miami, FL; Dow Corning Inc., Midland, MI.<br />

P5-07-02 Analysis of a novel synergistic relationship between the FK506<br />

binding protein FKBP52 and beta catenin in androgen receptor<br />

signaling pathways<br />

Storer CL, Olivares K, Fletterick RJ, Webb P, Cox MB. The University<br />

of Texas at El Paso, El Paso, TX; The University of California, <strong>San</strong><br />

Francisco, CA; The Methodist Hospital Research Institute, Houston, TX.<br />

P5-07-03 Gallotannins and Gallic acid From Mango Fruit (Mangifera<br />

Indica L) Suppress <strong>Breast</strong> <strong>Cancer</strong> Tumor Growth by Targeting<br />

Phosphatidylinositol3-kinase (PI3K)-Akt-NF-kB Pathway and<br />

Associated microRNAs<br />

Banerjee N, Kim H, Krenek K, Talcott S, Talcott SM. Texas A&M<br />

University, College Station, TX.<br />

P5-07-04 Aurora kinase regulates PKC-mediated MMP-9 expression and<br />

invasion in MCF-7 breast cancer cells<br />

Kim JS, Noh EM, Lee YR, Hwang B-M, Jung SH, Youn HJ, Lee SJ.<br />

Institute for Medical Sciences Chonbuk National University Medical<br />

School, Jeonju, Republic of Korea; Chonbuk National University<br />

Medical School, Jeonju, Republic of Korea; School of Dentistry,<br />

Wonkwang University, Iksan, Republic of Korea; College of Phamacy,<br />

Ewha Womans University, Seoul, Republic of Korea.<br />

P5-07-05 Insulin-like growth factor binding protein-3 is a key<br />

component of the breast cancer cell response to DNAdamaging<br />

therapy<br />

Baxter RC, Lin MZ, Marzec KA, Martin JL. University of Sydney,<br />

Sydney, NSW, Australia.<br />

P5-07-06 Triple negative receptor status is associated with low DNA<br />

repair capacity in women with breast cancer<br />

Matta J, Ortiz C, Vargas W, Echenique M, <strong>San</strong>chez F, Ramirez E,<br />

Torres A, Ortiz J, Bolaños G, Gonzalez J, Laboy J, Barnes R, <strong>San</strong>tiago S,<br />

Romero A, Martinez R, Alvarez-Garriga C, Bayona M. Ponce School<br />

of Medicine and Health Sciences, Ponce, Puerto Rico; Auxilio Mutuo<br />

Hospital, <strong>San</strong> Juan, Puerto Rico; Ponce School of Medicine and Health<br />

Sciences and Damas Hospital, Ponce, Puerto Rico; Private Office,<br />

Ponce, Puerto Rico; Ponce School of Medicine and Health Sciences<br />

and Private Practice, Ponce, Puerto Rico; Ponce School of Medicine<br />

and Health Sciences and <strong>San</strong> Lucas Hospital, Ponce, Puerto Rico;<br />

Ponce School of Medicine and Health Sciences, Ponce, Puerto Rico;<br />

Dr. Pila Hospital, Ponce, Puerto Rico; Food and Drug Administration,<br />

Silver Spring, MD.<br />

Tumor Cell and Molecular Biology: Telomeres/Telomerase<br />

P5-08-01 Investigating the role of BRCA1 in sensitivity to GRN163L, a<br />

telomerase template antagonist<br />

Phipps EA, Gryaznov SM, Herbert B-S. Indiana University School of<br />

Medicine, Indianapolis, IN; Geron Corporation, Menlo Park, CA.<br />

www.aacrjournals.org<br />

59s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Tumor Cell and Molecular Biology: Apoptosis and Senescence<br />

P5-09-01 Evaluation of BCL2 sequence variant in Iranian women patients<br />

with <strong>Breast</strong> cancer<br />

Motahari B, Ghaffarpour M, Javadi GH, Houshmand M. Science and<br />

Research branch, Islamic Azad University, Tehran, Islamic Republic<br />

of Iran; National Institute of Genetic Engineering and Biotechnology,<br />

Tehran, Islamic Republic of Iran; Iranian Research Organization for<br />

Science and Technology, Tehran, Islamic Republic of Iran; Special<br />

Medical Center, Tehran, Islamic Republic of Iran.<br />

Tumor Cell and Molecular Biology: MicroRNAs<br />

P5-10-01 MicroRNAs -181 and -135a modulate tumour<br />

infiltrating immune cell programs in distinct molecular breast<br />

cancer subtypes<br />

Paladini L, Truglia M, Arcangeli A, De Mattos Aruda L, Bianchini G,<br />

Iwamoto T, Bottai G, Pusztai L, Calin GA, Di Leo A, <strong>San</strong>tarpia L. Istituto<br />

Toscano Tumori, Prato, Italy; Istituto Toscano Tumori - Hospital of<br />

Prato, Prato, Italy; University of Florence, Italy; Vall d’Hebron Institute<br />

of Oncology, Vall d’Hebron University Hospital, Barcelona, Spain;<br />

Fondazione Centro <strong>San</strong> Raffaele del Monte Tabor, Milan, Italy;<br />

Okayama University Hospital, Okayama, Japan; The University of Texas<br />

M.D. Anderson <strong>Cancer</strong> Center, Houston.<br />

P5-10-02 High serum levels of miR-19a are associated with poor<br />

outcome in metastatic inflammatory breast cancer<br />

Anfossi S, Giordano A, Cohen EN, Gao H, Woodward W, Ueno NT,<br />

Valero V, Alvarez RH, Hortobagyi GN, Lee B-N, Cristofanilli M, Reuben<br />

JM. The University of Texas MD Anderson <strong>Cancer</strong> Center; Morgan<br />

Welch Inflammatory <strong>Breast</strong> <strong>Cancer</strong> Research Program and Clinic,<br />

The University of Texas MD Anderson <strong>Cancer</strong> Center; Fox Chase<br />

<strong>Cancer</strong> Center.<br />

P5-10-03 OncomiR-569 deregulates p53 pathway and initiate<br />

breast oncogenesis<br />

Chaluvally-Raghavan P, Zhang F, Pradeep S, Hee-Dong H, Lu Y,<br />

Borresen-Dale A-L, Flores ER, Sood AK, Mills GB. MD Anderson <strong>Cancer</strong><br />

Centre, Houston, TX; Institute for <strong>Cancer</strong> Research, Oslo University<br />

Hospital, Oslo, Oslo, Norway.<br />

P5-10-04 Metformin mediated upregulation of microRNA-193 triggers<br />

apoptosis by decreasing fatty acid synthase<br />

Cochrane DR, Wahdan-Alaswad RS, Edgerton SM, Terrell KL, Spoelstra<br />

NS, Thor AD, Anderson SM, Richer JK. University of Colorado Denver<br />

School of Medicine, Aurora, CO.<br />

P5-10-05 Novel Mechanisms of Metformin Action in TN <strong>Breast</strong> <strong>Cancer</strong>:<br />

Upregulation of miRNA 141 and 192, Are Associated with a<br />

Decrease in Targets GRB2 and MSN Involved in Signaling and<br />

Motility Respectively<br />

Edgerton SM, Richer JK, Fan Z, Spoelstra NS, Wahdan-Alsawad RS,<br />

Arnadottir SS, Thor AD. University of Colorado Denver School of<br />

Medicine, Aurora, CO; Aarhus University, Aarhus, Denmark.<br />

P5-10-06 A functional role for miR-150 in breast cancer<br />

D’Amato NC, Gu H, Lee M, Heinz R, Spoelstra NS, Jean A, Cochrane<br />

DR, Richer JK. University of Colorado Anschutz Medical Campus,<br />

Aurora, CO.<br />

P5-10-07 Luminal A breast cancer: identification of novel circulating<br />

miRNA biomarkers<br />

McDermott AM, Miller N, Ball G, Sweeney KJ, Kerin MJ. National<br />

University of Ireland, Galway, Galway, Ireland; Nottingham Trent<br />

University, Nottingham, United Kingdom.<br />

P5-10-08 Hyperactive MAPK signaling alters expression of miR-221/222<br />

and miR-30 families, which impacts multiple pathways and<br />

contributes to breast cancer aggressiveness and progression<br />

Miller P, El-Ashry D. University of Miami, FL.<br />

P5-10-09 Prediction of Trastuzumab treatment response for<br />

HER2-positive breast cancer by microRNA profiling<br />

Sato F, Wang Z, Ueno T, Myomoto A, Takizawa S, Masuda N, Mikami<br />

Y, Shimizu K, Tsujimoto G, Toi M. Graduate School of Medicine,<br />

Kyoto University, Kyoto, Japan; Toray Industry, Kamakura, Kanagawa,<br />

Japan; Kyoto University Hospital, Kyoto, Japan; Graduate School of<br />

Pharmaceutical Sciences, Kyoto University, Kyoto, Japan.<br />

P5-10-10 The miR-34a is down-regulated in breast cancer and breast<br />

stem cells and a potential to eradicating breast cancer via a<br />

systemic delivery of a VISA –miR-34a nanoparticle system<br />

Xie X, Li L, Xie X, Wei W, Kong Y, Wu M, Yang L, Gao J, Xiao X, Tang<br />

J, Xie Z, Wang X, Liu P, Li X, Guo J. Sun Yat-Sen University <strong>Cancer</strong><br />

Center, Guangzhou, China.<br />

P5-10-11 Clinical significance of microRNA expression as a prognostic<br />

factor in early N+ breast cancer (BC)<br />

Ciruelos EM, de Velasco GA, Castañeda C, Rodriguez-Peralto JL,<br />

Gamez A, Sepúlveda JM, Cortés-Funes H, Castellano DE, Fresno JA.<br />

Hospital Universitario 12 de Octubre, Madrid, Spain; Instituto de<br />

Enfermedades Neoplásicas, Lima, Peru; Hospital Universitario La Paz,<br />

Madrid, Spain.<br />

P5-10-12 Differences in microRNA expression patterns in breast cancer<br />

and triple negative tumors<br />

Romero-Cordoba SL, Rebollar-Vega RG, Quintanar-Jurado V,<br />

Rodriguez-Cuevas S, Bautista-Pina V, Maffuz-Aziz A, Hidalgo-Miranda<br />

A. National Institute of Genomic Medicine, Mexico; Instituto de<br />

Enfermedades de la Mama FUCAM, Mexico.<br />

P5-10-13 Pre-surgical plasma microRNA pattern defines a biologically<br />

distinct triple negative breast cancer (TNBC) occuring in black<br />

(B) compared to white (W) women<br />

Shapira I, Lee A, Oswald M, Taioli E, Bradley T, Barginear M, Mason C,<br />

Keogh M, Budman D. Monter <strong>Cancer</strong> Center, Hofstra North Shore LIJ<br />

School of Medicine, Lake Success, NY; Hofstra North Shore LIJ School<br />

of Medicine, Manhasset, NY.<br />

P5-10-14 MicroRNAs as biomarkers and therapeutic adjuvants for the<br />

prognosis and treatment of drug-resistant breast cancers<br />

Chang Y-F, Panneerdoss S, Zoghi B, Pertsemlidis A, Rao M. Greehey<br />

Children’s <strong>Cancer</strong> Research Institute, University of Texas Health<br />

Science Center at <strong>San</strong> <strong>Antonio</strong>, TX; UT Health Science Center at <strong>San</strong><br />

<strong>Antonio</strong>, TX.<br />

P5-10-15 Genetic variants located in beta2 adrenergic receptor gene<br />

(ADRB2) and miRNA let-7 binding site alter breast cancer<br />

susceptibility: a case control analysis<br />

Du Y, Lu J. Shanghai <strong>Cancer</strong> Center, Fudan University, Shanghai,<br />

China; Shanghai Medical College, Fudan University, Shanghai, China.<br />

P5-10-16 miRNAs as novel therapeutic adjuvants for the prognosis and<br />

treatment of triple negative breast cancers<br />

Zoghi B, Chang Y-F, Subbarayalu P, Plyler JR, Rao M. University of<br />

Texas Health Science Center at <strong>San</strong> <strong>Antonio</strong>, TX; Greehey Children’s<br />

<strong>Cancer</strong> Research Institute, University of Texas Health Science Center<br />

at <strong>San</strong> <strong>Antonio</strong>.<br />

P5-10-17 Radiotherapy-induced miR expression influences the<br />

formation of local recurrence in breast cancer<br />

Fabris L, Berton S, Massarut S, D’Andrea S, Roncadin M, Perin T,<br />

Calin G, Belletti B, Baldassarre G. CRO, National <strong>Cancer</strong> Institute; MD<br />

Anderson <strong>Cancer</strong> Center.<br />

Psychosocial, Quality of Life, and Educational Aspects: Advocacy<br />

P5-11-01 Extending breast conservation choices for women with<br />

breast cancer<br />

Egbeare DM, Chan HY. Cheltenham General Hospital, Cheltenham,<br />

Gloucestershire, United Kingdom.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012 60s <strong>Cancer</strong> Research


December 4–8, 2012<br />

Program Schedule<br />

P5-11-02 <strong>Breast</strong> <strong>Cancer</strong> Radiation Therapy and the Risk of Developing<br />

Bronchiolitis Obliterans Organizing Pneumonia (BOOP):<br />

Communication of BOOP Risk by <strong>Breast</strong> <strong>Cancer</strong> Information<br />

Websites and General Medical Information Websites<br />

Kelly EM. Kelly Consulting, Smithtown, NY.<br />

Psychosocial, Quality of Life, and Educational Aspects: Education<br />

P5-12-01 Are web-based resources the breast? An evaluation of the<br />

quality of online resources for breast cancer patients<br />

Nguyen S, Regehr G, Brar B, Lin J, Ingledew P. British Columbia<br />

<strong>Cancer</strong> Centre, Fraser Valley <strong>Cancer</strong> Centre, Surrey, BC, Canada;<br />

Centre for Health Education Scholarship, Vancouver, BC, Canada;<br />

UBC, Vancouver, BC, Canada.<br />

P5-12-02 Tangled in the <strong>Breast</strong> <strong>Cancer</strong> Web: An Evaluation of the<br />

Usage of Web-based Information Resources by <strong>Breast</strong><br />

<strong>Cancer</strong> Patients<br />

Nguyen SKA, Ingledew P. British Columbia <strong>Cancer</strong> Agency, FVCC,<br />

Surrey, BC, Canada.<br />

P5-12-03 Developing the ePromotora: Increasing Promotora’s Access to<br />

<strong>Breast</strong> <strong>Cancer</strong> Electronic Resources<br />

Lopez AM, Ryan J, Valencia A, El-Khayat Y, Nunez A. University of<br />

Arizona, Tucson, AZ.<br />

Psychosocial, Quality of Life, and Educational Aspects: Doctor-Patient<br />

Communication<br />

P5-13-01 Does empowering patients improve accrual to<br />

breast cancer trials?<br />

Arnaout A, Kuchuk I, Bouganim N, Verma S, Clemons M. Ottawa<br />

Hospital, Ottawa, ON, Canada; Mcgill University and Segal <strong>Cancer</strong><br />

Centre, Jewish General Hospital, Montreal, QC, Canada.<br />

P5-13-02 Decision making from multidisciplinary team meetings to<br />

bedside: factors predicting for physicians’ and breast cancer<br />

patients’ acceptance of clinical trials proposed by MTMs<br />

Deneuve J, Mazouni C, Arnedos M, Prenois F, Saghatchian M, André<br />

F, Bourrgier C, Delaloge S. Institut Gustave Roussy, Villejuif, France.<br />

P5-13-03 <strong>Breast</strong> cancer risk assessment for underserved minority<br />

women in primary care: patient and provider perspectives<br />

Hoskins KF, Anderson EE, Tejedo S, Stolley M, VandeWydeven K,<br />

Korah V, Moreno L, Rojas M, Carillo A, Caseras M, Awolala Y, Calhoun<br />

E, Campbell R, Warnecke R. The University of Illinois Hospital and<br />

Health Sciences System, Chicago, IL; The University of Illinois at<br />

Chicago School of Public Health, Chicago, IL; OSF Saint Anthony<br />

Medical Center, Rockford, IL; Loyola University Medical Center,<br />

Maywood, IL; Chicago Family Health Center, Chicago, IL.<br />

Psychosocial, Quality of Life, and Educational Aspects: Disparities and<br />

Barriers to Care<br />

P5-14-01 Withdrawn<br />

P5-14-02 Women’s responses to changes in US Preventive Services Task<br />

Force mammography screening guidelines: results from focus<br />

groups among ethnically diverse women<br />

Bluethmann SM, Allen JD, Hernandez C, Opdyke KM, Gates-Ferris K,<br />

Hurlbert M, Harden E. University of Texas School of Public Health;<br />

Dana Farber <strong>Cancer</strong> Institute; Avon Foundation for <strong>Breast</strong> <strong>Cancer</strong><br />

Crusade; Cicatelli and Associates.<br />

P5-14-03 <strong>Breast</strong> cancer screening and follow-up of abnormal<br />

mammogram results: A population-based study comparing<br />

results from an urban university cancer center to a national<br />

database<br />

Mitchell EP, Avery TP, Jaslow RC, Berger AC, Lee SY, Vaughan-Briggs<br />

C, Bonat J. Thomas Jefferson University, Philadelphia, PA.<br />

P5-14-04 Alleviate the pain in the system: Using process improvement<br />

and systems design tools to strive for a high quality breast<br />

healthcare system in the District of Columbia metro area<br />

Brousseau MK, Graham L, Kaye P, Nolan T, Moraras N. Primary Care<br />

Coalition of Montgomery County, Maryland, Silver Spring, MD;<br />

Regional Primary Care Coalition, Washington, DC; Associates in<br />

Process Improvement, Silver Spring, MD.<br />

P5-14-05 Compliance with radiation therapy in breast cancer patients in<br />

Southeastern Kentucky<br />

Elsoueidi R, Adane E, Dignan M. Appalachian Regional Healthcare,<br />

Hazard, KY; University of Kentucky, Lexington, KY.<br />

P5-14-06 Analysis of test-therapy concordance for biomarkers uPA and<br />

PAI-1 in primary breast cancer in clinical hospital routine:<br />

Results of a prospective multi-center study at Certified <strong>Breast</strong><br />

<strong>Cancer</strong>s in Germany<br />

Jacobs VR, Augustin D, Wischnik A, Kiechle M, Hoess C, Steinkohl O,<br />

Rack B, Kapitza T, Krase P. Paracelsus Medical University, Salzburg,<br />

Austria; Klinikum Deggendorf, Deggendorf; Klinikum Augsburg,<br />

Augsburg, Germany; Technical University Munich (TUM), Germany;<br />

Cooperative <strong>Breast</strong> Center Ebersberg-Rosenheim. Kreisklinikum<br />

Ebersberg, Ebersberg, Germany; Klinikum Dritter Orden, Munich,<br />

Germany; Ludwig-Maximilian-University (LMU), Munich, Germany;<br />

Top-Expertise, Germering/Munich, Germany; AOK Bayern, Munich.<br />

Psychosocial, Quality of Life, and Educational Aspects: Cost-Effectiveness<br />

P5-15-01 Cost-effectiveness of gene expression profiling for ductal<br />

carcinoma in-situ (Oncotype DCIS Score)<br />

Alvarado MD, Harrison BL, Solin LJ, Ozanne EM. University of<br />

California, <strong>San</strong> Francisco, CA; Albert Einstein Medical Center,<br />

Philadelphia, PA.<br />

P5-15-02 The benefit of targeted therapeutics in medical oncology since<br />

the development of trastuzumab<br />

Conter HJ, Conter D, Wolff RA, Valero V, Zwelling L. University of<br />

Texas M.D. Anderson <strong>Cancer</strong> Center, Houston, TX; Huron College,<br />

London, ON, Canada.<br />

P5-15-03 Cost-effectiveness of ‘radioguided occult lesion localization’<br />

(ROLL) versus ‘wire-guided localization’ (WGL) in breast<br />

conserving surgery for non-palpable breast cancer: results<br />

from a randomized controlled multicentre trial<br />

Postma EL, Koffijberg E, Verkooijen HM, Witkamp AJ, van den Bosch<br />

MA, van Hillegersberg R. UMC Utrecht; Julius Center Utrecht.<br />

P5-15-04 CTX and CTX-related direct medication costs saved by testing<br />

biomarkers uPA and PAI-1 in primary breast cancer: Results of<br />

a prospective multi-center study at Certified <strong>Breast</strong> Centers<br />

in Germany<br />

Jacobs VR, Augustin D, Wischnik A, Kiechle M, Hoess C, Steinkohl O,<br />

Rack B, Kapitza T, Krase P. Paracelsus Medical University, Salzburg,<br />

Austria; Klinikum Deggendorf, Deggendorf, Germany; Klinikum<br />

Augsburg, Augsburg, Germany; Technical University Munich<br />

(TUM), Germany; Cooperative <strong>Breast</strong> Center Ebersberg-Rosenheim.<br />

Kreisklinikum Ebersberg, Ebersberg, Germany; Klinikum Dritter<br />

Orden, Munich, Germany; Ludwig-Maximilian-University (LMU),<br />

Munich, Germany; Top Expertise, Germering/Munich, Germany; AOK<br />

Bayern, Munich, Germany.<br />

P5-15-05 Budget impact analysis of everolimus for estrogen receptor<br />

positive, human epidermal growth factor receptor-2 negative<br />

metastatic breast cancer patients in the United States<br />

Xie J, Diener M, De G, Yang H, Wu EQ, Namjoshi M. Analysis Group,<br />

Inc., New York, NY; Analysis Group, Inc., Boston, MA; Novartis<br />

Pharmaceuticals Corporation, East Hanover, NJ.<br />

www.aacrjournals.org<br />

61s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P5-15-06 Societal economics of the 21-gene Recurrence Score® in<br />

estrogen-receptor-positive early-stage breast cancer in Japan<br />

Yamauchi H, Nakagawa C, Yamashige S, Takei H, Yagata H, Yoshida<br />

A, Hayashi N, Hornberger J, Yu T, Chien R, Chao C, Yoshizawa C,<br />

Nakamura S. St. Luke’s International Hospital, Chu-o-ku, Tokyo,<br />

Japan; Hitotsubashi University, Tokyo, Japan; Saitama <strong>Cancer</strong> Center,<br />

Saitama, Japan; Cedar Associates LLC, Menlo Park, CA; Stanford<br />

University School of Medicine, Stanford, CA; Mailman School of<br />

Public Health, Columbia University, New York, NY; Genomic Health,<br />

Inc., Redwood City, CA; Showa University, Tokyo, Japan.<br />

P5-15-07 Time savings with trastuzumab subcutaneous (SC) injection<br />

vs. trastuzumab intravenous (IV) infusion: First results from a<br />

Time-and-Motion study (T&M)<br />

de Cock E, Tao S, Alexa U, Pivot X, Knoop A. United Biosource<br />

Corporation, Barcelona, Spain; United Biosource Corporation,<br />

Montreal, Canada; F Hoffmann-La Roche Ltd, Basel, Switzerland;<br />

CHU Jean Minjoz, Besancon, France; Odense University Hospital,<br />

Odense, Denmark.<br />

Treatment: Immunotherapy<br />

P5-16-01 Survival advantage in patients with metastatic breast cancer<br />

receiving endocrine therapy plus Sialyl Tn-KLH vaccine: post<br />

hoc analysis of a large randomized trial<br />

Ibrahim NK, Murray JL, Zhou D, Mittendorf EA, Sample D, Tautchin<br />

M, Miles D. University of Texas MD Anderson <strong>Cancer</strong> Center,<br />

Houston, TX; Biomira, Inc., AB, Canada; Mount Vernon <strong>Cancer</strong> Center,<br />

Northwood, Middlesex, United Kingdom.<br />

P5-16-02 Final Results of the Phase I/II Trials of the E75 Adjuvant<br />

<strong>Breast</strong> <strong>Cancer</strong> Vaccine<br />

Vreeland TJ, Clifton GT, Hale DF, Sears AK, Patil R, Holmes JP, Ponniah<br />

S, Mittendorf EA, Peoples GE. <strong>San</strong> <strong>Antonio</strong> Military Medical Center,<br />

<strong>San</strong> <strong>Antonio</strong>, TX; Joyce Murtha <strong>Breast</strong> Care Center, Windber, PA;<br />

Redwood Regional Medical Group, <strong>San</strong>ta Rosa Memorial Hospital,<br />

<strong>San</strong>ta Rosa, CA; Uniformed Services University of Health Sciences,<br />

Bethesda, MD; MD Anderson <strong>Cancer</strong> Center, Houston, TX.<br />

P5-16-03 Phase II study of autologous dendritic cell vaccination in<br />

patients with HER2 negative breast cancer combined with<br />

neoadjuvant chemotherapy<br />

Castillo A, Salgado E, Inoges S, Arevalo E, Castañon E, López Díaz<br />

de Cerio A, Sola JJ, Pina L, Nuñez J, Rodriguez-Spiteri N, Espinós<br />

J, Aramendia JM, Fernandez Hidalgo OA, <strong>San</strong>tisteban M. Clínica<br />

Universidad de Navarra, Pamplona, Navarra, Spain; Complejo<br />

Hospitalario de Navarra, Pamplona, Navarra, Spain; Universidad de<br />

Navarra, Pamplona, Navarra, Spain; Clínica Universidad de Navarra.<br />

P5-16-04 A phase I study of a DNA plasmid based vaccine encoding<br />

the HER-2/neu intracellular domain in subjects with HER2+<br />

breast cancer<br />

Salazar LG, Slota M, Higgens D, Coveler A, Dang Y, Childs J, Bates N,<br />

Guthrie K, Waisman J, Disis ML. University of Washington, Seattle,<br />

WA; BREASTLINK, Hawthorne, CA.<br />

P5-16-05 The combination of trastuzumab and HER2-directed peptide<br />

vaccines is safe in HER2-expressing breast cancer patients<br />

Hale DF, Vreeland TJ, Perez SA, Berry JS, Ardavanis A, Trappey<br />

AF, Tzonis P, Sears AK, Clifton GT, Shumway NM, Papamichail<br />

M, Ponniah S, Peoples GE, Mittendorf EA. Brooke Army Medical<br />

Center, <strong>San</strong> <strong>Antonio</strong>, TX; <strong>Cancer</strong> Immunology and Immunotherapy<br />

Center, Athens, Greece; MD Anderson <strong>Cancer</strong> Center, Houston, TX;<br />

Uniformed Services University of the Health Sciences, Bethesda, MD.<br />

P5-16-06 A phase 2 randomized trial of docetaxel (DOC) alone or<br />

in combination with therapeutic cancer vaccine, CEA-,<br />

MUC-1-TRICOM (PANVAC)<br />

Heery CR, Ibrahim NK, Mohebtash M, Madan RA, Arlen PM, Bilusic M,<br />

Kim JW, Singh NK, Hodge S, McMahon S, Steinberg SM, Hodge JW,<br />

Schlom J, Gulley JL. National <strong>Cancer</strong> Institute.<br />

Treatment: Antiangiogenic Therapy<br />

P5-17-01 Bevacizumab (B) in the adjuvant treatment of breast cancer -<br />

first toxicity results from Eastern Cooperative Oncology Group<br />

trial E5103<br />

Miller KD, O’Neill A, Dang C, Northfelt D, Gradishar W, Sledge GW.<br />

Indiana University Melvin and Bren Simon <strong>Cancer</strong> Center; Dana<br />

Farber <strong>Cancer</strong> Institute; Memorial Sloan Kettering <strong>Cancer</strong> Center;<br />

Mayo Clinic; Northwestern University.<br />

P5-17-02 Withdrawn<br />

P5-17-03 Quality of life (QoL) results from the TURANDOT trial<br />

comparing two bevacizumab (BEV)-containing regimens<br />

as first-line treatment for HER2-negative metastatic breast<br />

cancer (mBC)<br />

Láng I, Inbar MJ, Kahan Z, Greil R, Beslija S, Stemmer SM, Kaufman<br />

B, Ahlers S, Brodowicz T, Zielinski C. National Institute of Oncology,<br />

Budapest, Hungary; Tel Aviv Sourasky Medical Centre, Tel Aviv,<br />

Israel; University of Szeged, Hungary; Paracelsus Medical University,<br />

Salzburg, Austria; Institute of Oncology, Sarajevo, Bosnia and<br />

Herzegowina; Rabin Medical Center, Petah-Tikva, Israel; Sheba<br />

Medical Center, Ramat Gan, Israel; IST GmbH Mannheim, Mannheim,<br />

Germany; Medical University of Vienna and Central European<br />

Cooperative Oncology Group (CECOG), Vienna, Austria.<br />

P5-17-04 Management of Antiangiogenics’ Renovascular Safety in<br />

breast cancer. Subgroup and intermediate results of the<br />

MARS Study<br />

Launay-Vacher V, Janus N, Daniel C, Rey J-B, Ray-Coquard I, Gligorov<br />

J, Spano J-P, Thery J-C, Jouannaud C, Goldwasser F, Mir O, Morere<br />

J-F, Oudard S, Azizi M, Dorent R, Deray G, Beuzeboc P. Institut Curie,<br />

Paris, France; Hôpital de la Pitié-Salpêtrière, Paris, France; Institut Jean<br />

Godinot, Reims, France; Centre Léon Bérard, Lyon, France; Hôpital<br />

Tenon, Paris, France; Hôpital Cochin, Paris, France; Hôpital Avicenne,<br />

Bobigny, France; Hôpital Européen Georges Pompidou, Paris, France.<br />

P5-17-05 Sorafenib plus Ixabepilone as First-Line Treatment for<br />

Patients with HER2-Negative Metastatic <strong>Breast</strong> <strong>Cancer</strong>:<br />

Preliminary Results of the Phase II Trial of the Sarah Cannon<br />

Research Institute<br />

Yardley DA, Barton, Jr. J, Dickson N, Shipley D, Drosick DR, Hendricks<br />

C, Inhorn RC, Shastry M, Finney L, Burris HA. Tennessee Oncology,<br />

PLLC, Nashville, TN; Sarah Cannon Research Institute, Nashville,<br />

TN; National Capital Clinical Research Consortium, Bethesda, MD;<br />

Oncology Hematology Care, Cincinnati, OH; Mercy Hospital,<br />

Portland, ME.<br />

P5-17-06 The deficient eNOS c.894G>T genotype is not associated with<br />

increased severity of hypertension and proteinuria in breast<br />

cancer patients receiving bevacizumab<br />

Del Re M, Ferrarini I, Fontana A, <strong>San</strong>toro M, Bona E, Del Re I, Stasi<br />

I, Bertolini I, Laurà F, Landucci E, Salvadori B, Falcone A, Danesi R.<br />

University Hospital, Pisa, Italy.<br />

P5-17-07 Influence of bevacizumab on local-regional recurrence in<br />

triple negative breast cancer<br />

Prendergast BM, De Los <strong>San</strong>tos JF, Barrett OC, Forero A, Falkson<br />

CI, Urist MM, Krontiras H, Bland KI, Meredith RF, Keene KS, You Z,<br />

Carpenter JT. University of Alabama Birmingham, AL.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012 62s <strong>Cancer</strong> Research


December 4–8, 2012<br />

Program Schedule<br />

Treatment: HER2-Targeted Therapy<br />

P5-18-01 Pertuzumab (P) in combination with trastuzumab (T) and<br />

docetaxel (D) in elderly patients with HER2-positive metastatic<br />

breast cancer in the CLEOPATRA study<br />

Miles D, Baselga J, Amadori D, Sunpaweravong P, Semiglazov V,<br />

Knott A, Clark E, Ross G, Swain SM. Mount Vernon <strong>Cancer</strong> Centre,<br />

Middlesex, United Kingdom; Massachusetts General Hospital <strong>Cancer</strong><br />

Center and Harvard Medical School, Boston, MA; Istituto Scientifico<br />

Romagnolo per lo Studio e la Cura dei Tumori (IRST), Meldola, Italy;<br />

Songklanagarind Hospital, Prince of Songkla University, Hat Yai,<br />

Thailand; NN Petrov Research Institute of Oncology, St Petersburg,<br />

Russian Federation; Roche Products Limited, Welwyn, United<br />

Kingdom; MedStar Washington Hospital Center, Washington, DC.<br />

P5-18-02 Selective Crossover in Randomized Trials of Adjuvant<br />

Trastuzumab for <strong>Breast</strong> <strong>Cancer</strong>: Coping with Success<br />

Regan MM, Dafni U, Karlis D, Goldhirsch A, Untch M, Smith I, Gianni<br />

L, Jackisch C, de Azambuja E, Heinzmann D, Cameron D, Bell R,<br />

Dowsett M, Baselga J, Leyland-Jones B, Piccart-Gebhart MJ, Gelber<br />

RD, On behalf of the HERA Study Team. Dana-Farber <strong>Cancer</strong> Institute,<br />

Boston, MA; University of Athens and Frontier Science Foundation-<br />

Hellas, Athens, Greece; Institut Jules Bordet, Universite Libre de<br />

Bruxelles, Brussels, Belgium; Helios Klinikum Berlin Buch, Berlin,<br />

Germany; Royal Marsden Hospital and Institute of <strong>Cancer</strong> Research,<br />

London, United Kingdom; <strong>San</strong> Raffaele Institute, Milan, Italy; Western<br />

General Hospital and University of Edinburgh, United Kingdom;<br />

The Royal Marsden NHS Trust, London, United Kingdom; European<br />

Institute of Oncology, Milan, Italy; Athens University of Economics,<br />

Athens, Greece; Massachusetts General Hospital, Boston, MA; F.<br />

Hoffmann-La Roche, Basel, Switzerland; <strong>San</strong>ford Research, Sioux Falls,<br />

SD; The Andrew Love <strong>Cancer</strong> Centre, The Geelong Hospital, Geelong,<br />

Australia; Klinikum Offenbach, Offenbach, Germany.<br />

P5-18-03 Clinicopathological features among patients with HER2-<br />

positive breast cancer with prolonged response to<br />

trastuzumab based therapy<br />

Vaz-Luis I, Seah D, Olson E, Metzger O, Wagle N, Sohl J, Litsas G,<br />

Burstein H, Krop I, Winer E, Lin NU. Dana Farber <strong>Cancer</strong> Institute,<br />

Boston, MA; Ohio State University.<br />

P5-18-04 Tolerability and efficacy of targeting both mTOR and HER2<br />

signaling in trastuzumab-refractory HER2+ metastatic<br />

breast cancer<br />

Gajria D, King T, Pannu H, Sakr R, Modi S, Drullinsky P, Syldor A, Patil<br />

S, Seidman A, Norton L, Rosen N, Hudis C, Chandarlapaty S. Memorial<br />

Sloan-Kettering <strong>Cancer</strong> Center, New York, NY.<br />

P5-18-05 Interim Results from a Phase 1b/2a Study of Trastuzumab<br />

Emtansine and Docetaxel, With and Without Pertuzumab, in<br />

Patients With HER2-Positive Locally Advanced or Metastatic<br />

<strong>Breast</strong> <strong>Cancer</strong><br />

Martin M, García-Sáenz JÁ, Dewar JA, Albanell J, Limentani<br />

SA, Strasak A, Patre M, Branle F, Fumoleau P. Hospital General<br />

Universitario Gregorio Marañón; Hospital <strong>San</strong> Carlos; Ninewells<br />

Hospital; Hospital del Mar; Levine <strong>Cancer</strong> Institute; F. Hoffmann-La<br />

Roche Limited; Centre Georges François Leclerc.<br />

P5-18-06 Trastuzumab emtansine in HER2-positive metastatic breast<br />

cancer: pooled safety analysis from seven studies<br />

Diéras V, Harbeck N, Budd GT, Greenson JK, Guardino E, Samant<br />

M, Chernyukhin N, Smitt M, Krop IE. Institut Curie; <strong>Breast</strong> Center,<br />

University of Munich; Cleveland Clinic, Lerner College of Medicine;<br />

University of Michigan; Genentech, Inc.; Dana-Farber <strong>Cancer</strong> Institute.<br />

P5-18-07 Completed SN33 Trial: 60 month follow-up of Early Stage<br />

Node Positive HER2 Negative patients with NeuVax (E75)<br />

and sargramostim<br />

Mazanet R, Schwartz MW, Ramadan D, Lavin PT. Galena Biopharma,<br />

Lake Oswego, OR; Aptiv Solutions, Southborough, MA.<br />

P5-18-08<br />

P5-18-09<br />

P5-18-10<br />

P5-18-11<br />

P5-18-12<br />

P5-18-13<br />

P5-18-14<br />

P5-18-15<br />

P5-18-16<br />

Identification of ErbB2 function in the heart: implication for<br />

anti-ErbB2 therapy in breast cancer<br />

Perry M-C, Eichner LJ, Dufour CR, Muller WJ, Giguère V. Rosalind<br />

and Morris Goodman <strong>Cancer</strong> Research Centre, McGill University,<br />

Montréal, QC, Canada; McGill University, Montréal, QC, Canada.<br />

A Phase I Study of MM-302, a HER2-targeted Liposomal<br />

Doxorubicin, in Patients with Advanced, HER2- Positive<br />

<strong>Breast</strong> <strong>Cancer</strong><br />

Wickham T, Futch K. Merrimack Pharmaceuticals.<br />

Chemotherapy can enhance trastuzumab-mediated ADCC<br />

Tagliabue E, Sfondrini L, Regondi V, Casalini P, Pupa SM, Sommariva<br />

M, Balsari A, Triulzi T. Fondazione IRCCS Istituto Nazionale dei Tumori;<br />

Università degli Studi di Milano.<br />

Pharmacokinetics and exposure-efficacy relationship of<br />

trastuzumab emtansine in EMILIA, a phase 3 study of<br />

trastuzumab emtansine vs capecitabine and lapatinib in HER2-<br />

positive locally advanced or metastatic breast cancer<br />

Wang B, Jin J, Wada R, Fang L, Saad O, Olsen S, Althaus B, Swain<br />

S, Untch M, Girish S. Genentech, Inc.; Quantitative Solutions;<br />

Washington Hospital Center; HELIOS Hospital Berlin-Buch.<br />

Comparison of treatment patterns and outcomes in metastatic<br />

breast cancer patients initiated on trastuzumab vs. lapatinib; a<br />

retrospective analysis<br />

Gauthier G, Guérin A, Styles AL, Wu EQ, Masaquel A, Brammer MG,<br />

Lalla D. Analysis Group Inc., Boston, MA; Genentech, Inc., South <strong>San</strong><br />

Francisco, CA.<br />

Inhibiting telomerase to reverse trastuzumab (T) resistance in<br />

HER2+ breast cancer<br />

Miller KD, Steding CE, Prasad N, Rojas LA, Herbert B-S. Indiana<br />

University Melvin and Bren Simon <strong>Cancer</strong> Center.<br />

Cardiac monitoring during adjuvant trastuzumab therapy for<br />

breast cancer<br />

Ng D, Ferrusi I, Khong H, Earle C, Trudeau M, Marshall D, Leighl N.<br />

University of Toronto, ON, Canada; McMaster University, Hamilton,<br />

ON, Canada; University of Calgary, AB, Canada; Ontario Institute for<br />

<strong>Cancer</strong> Research, Toronto, ON, Canada.<br />

Molecular effects of lapatinib in HER2 positive ductal<br />

carcinoma in situ (DCIS)<br />

Estevez LG, Suarez A, Calvo I, Garcia E, Miro C, Durán H, Quijano<br />

Y, Perea S, Herrero M, Lopez-Rios F, Hidalgo M. Centro Integral<br />

Oncologico Clara Campal, Madrid, Spain.<br />

A Multicenter Phase 2 Study (JO22997) Evaluating the<br />

Efficacy and Safety of Trastuzumab Emtansine in Japanese<br />

Patients With Heavily Pretreated HER2-Positive Metastatic<br />

<strong>Breast</strong> <strong>Cancer</strong><br />

Masuda N, Ito Y, Takao S, Doihara H, Rai Y, Horiguchi J, Kohno N,<br />

Fujiwara Y, Tokuda Y, Watanabe J, Iwata H, Ishiguro H, Miyoshi Y,<br />

Matsubara M, Kashiwaba M. NHO Osaka National Hospital, Osaka,<br />

Japan; The <strong>Cancer</strong> Institute Hospital of JFCR, Tokyo, Japan; Hyogo<br />

<strong>Cancer</strong> Center, Akashi, Japan; Okayama University Hospital, Okayama,<br />

Japan; Sagara Hospital, Kagoshima, Japan; Gunma University,<br />

Maebashi, Japan; Tokyo Medical University, Tokyo, Japan; National<br />

<strong>Cancer</strong> Center Central Hospital, Tokyo, Japan; Tokai University,<br />

Isehara, Japan; Shizuoka <strong>Cancer</strong> Center, Sunto-gun, Japan; Aichi<br />

<strong>Cancer</strong> Center, Nagoya, Japan; Kyoto University, Kyoto, Japan; Hyogo<br />

College of Medicine, Nishinomiya, Japan; Chugai Pharmaceutical<br />

Co.,Ltd., Tokyo, Japan; Iwate Medical University, Morioka, Japan.<br />

www.aacrjournals.org<br />

63s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P5-18-17 Impact of compliance with National Comprehensive <strong>Cancer</strong><br />

Network guidelines on adjuvant trastuzumab administration<br />

for patients with HER2-positive breast cancer<br />

Mullins DN, Maly J, Abdel-Rasoul M, Shapiro CL, Olson EM.<br />

Comprehensive <strong>Cancer</strong> Center, The Ohio State University Wexner<br />

Medical Center, Arthur G. James <strong>Cancer</strong> Hospital and Richard J.<br />

Solove Research Institute, Columbus, OH; The Ohio State University<br />

Wexner Medical Center, Columbus, OH; The Ohio State University,<br />

Columbus, OH.<br />

P5-18-18 Lapatinib versus trastuzumab, or both, added to preoperative<br />

chemotherapy for breast cancer: A meta-analysis of<br />

randomized evidence<br />

Valachis A, Nearchou A, Lind P. University of Uppsala, Sweden;<br />

Mälarsjukhuset, Eskilstuna, Sweden; Karolinska Institute,<br />

Stockholm, Sweden.<br />

P5-18-19 The 2006 Adjuvant Trastuzumab Convention in Belgium: 5<br />

years later<br />

Vanderhaegen J, Paridaens R, Piccart M, Lalami Y, Machiels J-P, Louis<br />

E, Borms M, Mebis J, Dirix L, Lintermans A, Brouckaert O, Neven P.<br />

University Hospital Leuven, Belgium; University Hospital Leuven/<br />

Catholic University Leuven, Belgium; Institut Jules Bordet, Brussels,<br />

Belgium; Cliniques Universitaires St-Luc, Brussels, Belgium; CHU de<br />

Liège, Liege, Belgium; AZ Groeninge, Kortrijk, Belgium; Limburgs<br />

Oncologisch Centrum, Limburg, Belgium; St Augustinus Hospital,<br />

Wilrijk, Belgium.<br />

P5-18-20 Phase II study of pertuzumab, trastuzumab, and weekly<br />

paclitaxel in patients with metastatic HER2-overexpressing<br />

metastatic breast cancer<br />

Datko F, D’Andrea G, Dickler M, Theodoulou M, Goldfarb S, Lake D,<br />

Fornier M, Modi S, Sklarin N, Comen E, Fasano J, Gajria D, Drullinsky<br />

P, Gilewski T, Murphy C, Syldor A, Lau A, Hamilton N, Patil S, Liu J,<br />

Chandarlapaty S, Hudis C, Dang C. Memorial Sloan-Kettering <strong>Cancer</strong><br />

Center, New York, NY; Bon Secours Hospital, Cork, Ireland.<br />

P5-18-21 A Phase II randomized trial of lapatinib with either vinorelbine<br />

or capecitabine as first- and second-line therapy for ErbB2-<br />

overexpressing metastatic breast cancer (MBC)<br />

Janni W, Sarosiek T, Pikiel J, Karaszewska B, Staroslawska E, Salat<br />

C, Caglevic C, Potemski P, Brain E, Briggs K, de Silvio M, Sapunar F,<br />

Papadimitriou C. Klinikum der Heinrich-Heine-Universität Düsseldorf,<br />

Düsseldorf, Germany; Centrum Medyczne Ostrobramska, NZOZ<br />

Magodent, Warsaw, Poland; Wojewodzkie Centrum Onkologii,<br />

Centrum Badan Klinicznych, Gdansk, Poland; Przychodnia Lekarska<br />

NZOZ “KOMED”, Konin, Poland; Centrum Onkologii Ziemii Lubelskiej,<br />

Lublin, Poland; Hämato-Onkologische Gemeinschaftspraxis, München,<br />

Germany; Mariano <strong>San</strong>chez Fontecilla, Las Condes, Chile; Wojewodzki<br />

Szpital Specjalistyczny, Kopernika, Poland; Institut Curie - Hôpital René<br />

Huguenin, Saint-Cloud, France; GlaxoSmithKline Oncology, Uxbridge,<br />

Middlesex, United Kingdom; GlaxoSmithKline Oncology, Collegeville,<br />

PA; Therapeutic Clinic, General Hospital of Athens, Greece.<br />

P5-18-22 Long-term survival of patients with HER2 metastatic breast<br />

cancer treated by targeted therapies<br />

Fiteni F, Villanueva C, Bazan F, Chaigneau L, Cals L, Dobi E,<br />

Montcuquet P, Nerich V, Limat S, Pivot X. Besançon University<br />

Hospital, Besançon, Doubs, France.<br />

P5-18-23 The prognosis of T1a, T1b N0 M0, HER2+ patients in Korea<br />

Lee JW, Moon H-G, Han W, Noh D-Y. Seoul National University<br />

Hospital, Seoul, Korea.<br />

P5-18-24 Population pharmacokinetics of trastuzumab emtansine, a<br />

HER2-targeted antibody-drug conjugate, in patients with<br />

HER2-positive metastatic breast cancer: clinical implications of<br />

the effect of various covariates<br />

Lu D, Girish S, Gao Y, Wang B, Yi J-H, Guardino E, Samant M,<br />

Cobleigh M, Rimawi M, Conte P, Jin J. Genentech, Inc.; Quantitative<br />

Solutions; Rush University Medical Center; Baylor College of<br />

Medicine; University of Modena and Reggio Emilia.<br />

P5-18-25 Bispecific Fynomer-antibody fusion proteins targeting two<br />

epitopes on HER2<br />

Grabulovski D, Brack S, Toller I, Mourlane F, Bertschinger J. Covagen<br />

AG, Zurich-Schlieren, Switzerland.<br />

P5-18-26 Confirmatory overall survival (OS) analysis of CLEOPATRA: a<br />

randomized, double-blind, placebo-controlled Phase III study<br />

with pertuzumab (P), trastuzumab (T), and docetaxel (D) in<br />

patients (pts) with HER2-positive first-line (1L) metastatic<br />

breast cancer (MBC)<br />

Swain SM, Kim S-B, Cortes J, Ro J, Semiglazov V, Campone M,<br />

Ciruelos E, Ferrero J-M, Schneeweiss A, Knott A, Clark E, Ross G,<br />

Benyunes MC, Baselga J. MedStar Washington Hospital Center,<br />

Washington; Asan Medical Center, University of Ulsan College of<br />

Medicine, Seoul, Korea; Vall d’Hebron University Hospital, Barcelona,<br />

Spain; National <strong>Cancer</strong> Center, Goyang, Korea; NN Petrov Research<br />

Institute of Oncology, St Petersburg, Russian Federation; Centre<br />

René Gauducheau, Saint-Herblain (Nantes), France; Hospital 12 De<br />

Octubre, Madrid, Spain; Centre Antoine Lacassagne, Nice, France;<br />

University Hospital Heidelberg, Heidelberg, Germany; Roche<br />

Products Limited, Welwyn, United Kingdom; Genentech, South<br />

<strong>San</strong> Francisco; Massachusetts General Hospital <strong>Cancer</strong> Center and<br />

Harvard Medical School, Boston.<br />

Treatment: Signal Transduction Inhibitors<br />

P5-19-01 Targeting the HER3-phosphatidyl inositol-3 kinase pathway in<br />

breast cancers<br />

Cook RS, Morrison MM, Arteaga CL, Perou CM. Vanderbilt University,<br />

Nashville, TN; University of North Carolina.<br />

P5-19-02 Selective PI3K and dual PI3K/mTOR inhibitors enhance the<br />

efficacy of endocrine therapies in breast cancer models<br />

Friedman L, Ross LB, Wallin J, Guan J, Prior WW, Wu E, Nannini M,<br />

Sampath D. Genentech, Inc., South <strong>San</strong> Francisco, CA.<br />

P5-19-03 Olaparib plus carboplatin in combination with vandetanib<br />

inhibited the growth of BRCA-wt triple negative breast tumors<br />

in mice: Outside BRCA-box<br />

Dey N, Sun Y, De P, Leyland-Jones B. <strong>San</strong>ford Research/USD,<br />

Sioux Falls, SD.<br />

Treatment: Targeted Therapy - Advanced Disease<br />

P5-20-01 A randomized double-blind phase II study of the<br />

combination of oral WX-671 plus capecitabine vs.<br />

capecitabine monotherapy in first-line HER2- negative<br />

metastatic breast cancer (MBC)<br />

Goldstein LJ, Oliveria CT, Heinrich B, Stemmer SM, Mala C, Selder<br />

S, Bevan P, Harbeck N. Fox Chase <strong>Cancer</strong> Center, Philadelphia, PA;<br />

Instituto Brasilerio Controle <strong>Cancer</strong>, Sao Paulo, Brazil; Hamatologisch-<br />

Onkologische-Praxis Augsburg, Augsburg, Germany; Rabin Medical<br />

Center, Petah Tikva, Israel; Wilex, Munich, Germany; Univeristy of<br />

Munich, Munich, Germany.<br />

P5-20-02 Predictors of long-term survival in a large cohort of patients<br />

with HER2-positive (HER2+) metastatic breast cancer (MBC)<br />

Chavez Mac Gregor M, Lei X, Giordano SH, Valero V, Esteva F,<br />

Mittendorf EA, Gonzalez-Angulo AM, Hortobagyi GN. University of<br />

Texas MD Anderson <strong>Cancer</strong> Center, Houston, TX.<br />

P5-20-03 A phase 2, double-blind, randomized, placebo-controlled,<br />

dose-finding study of sotatercept for the treatment of<br />

patients with chemotherapy-induced anemia and metastatic<br />

breast cancer<br />

Auerbach M, Osborne CRC, Klesczewski K, Laadem A, Sherman<br />

ML, Bianca R. Auerbach Hematology/Oncology, Baltimore, MD;<br />

Texas Oncology, P.A., Sammons <strong>Cancer</strong> Center, Dallas, TX; Celgene<br />

Corporation, Summit, NJ; Acceleron Pharma, Cambridge, MA.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012 64s <strong>Cancer</strong> Research


December 4–8, 2012<br />

Program Schedule<br />

P5-20-04<br />

P5-20-05<br />

P5-20-06<br />

P5-20-07<br />

P5-20-08<br />

P5-20-09<br />

P5-20-10<br />

P5-20-11<br />

Eribulin mesylate + trastuzumab as first-line therapy for<br />

locally recurrent or metastatic HER2-positive breast cancer:<br />

results from a phase 2, multicenter, single-arm study<br />

Vahdat L, Schwartzberg L, Wilks S, Rege J, Liao J, Cox D,<br />

O’Shaughnessy J. Weill Cornell Medical College, New York, NY; The<br />

West Clinic, Memphis, TN; <strong>Cancer</strong> Care Centers of South Texas, <strong>San</strong><br />

<strong>Antonio</strong>, TX; Eisai Inc, Woodcliff Lake, NJ; Texas Oncology-Baylor<br />

Charles A. Sammons <strong>Cancer</strong> Center, Dallas, TX.<br />

A Phase 2 trial of RAD 001 and Carboplatin in patients with<br />

triple negative metastatic breast cancer<br />

Singh JC, Volm M, Novik Y, Speyer J, Adams S, Omene CO, Meyers<br />

M, Smith JA, Schneider R, Formenti S, Goldberg JD, Li X, Davis S,<br />

Beardslee B, Tiersten A. New York University, New York, NY.<br />

RAD001 (Everolimus) in combination with Letrozole in the<br />

treatment of postmenopausal women with estrogen receptor<br />

positive Metastatic <strong>Breast</strong> cancer after failure of hormonal<br />

therapy – a phase II study<br />

Safra T, Kaufman B, Ben Baruch N, Kadouri-Sonenfeld L, Nisenbaum<br />

B, Greenberg J, Ryvo L, Yerushalmi R, Evron E. Tel Aviv Sourasky MC,<br />

Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; Sheba<br />

Medical Center, Ramat Gan, Israel; Kaplan Medical Center, Rehovot,<br />

Israel; Hadassah Ein Karem Medical Center, Jerusalem, Israel; Meir<br />

Kfar Saba Medical Center, Kfar Saba, Israel; Rabin Medical Center<br />

Belinson Campus, Petach Tikva, Israel; Assaf Harofeh Medical Center,<br />

Assaf Harofeh, Israel.<br />

Phase II Trial of Dasatinib in Combination With Weekly<br />

Paclitaxel for Patients with Metastatic <strong>Breast</strong> Carcinoma<br />

Morris PG, Lake D, McArthur HL, Gilewski T, Dang C, Chaim J, Patl S,<br />

Lim K, Norton L, Hudis CA, Fornier MN. Memorial Sloan-Kettering<br />

<strong>Cancer</strong> Center, New York.<br />

Phase II trial of ixabepilone (Ixa) and dasatinib (D) for<br />

treatment of metastatic breast cancer (MBC)<br />

Schwartzberg LS, Tauer KW, Schnell FM, Hermann R, Rubin P,<br />

Christianson D, Weinstein P, Epperson A, Walker M. The West Clinic,<br />

Memphis, TN; Central Georgia <strong>Cancer</strong> Care, Macon, GA; Northwest<br />

Georgia Oncology Centers, Marietta, GA; Cone Health <strong>Cancer</strong> Center,<br />

Greensboro, NC; Hematology Oncology Centers of the Northern<br />

Rockies, Billings, MT; Hematology Oncology PC, Stamford, CT; ACORN<br />

Research, LLC, Memphis, TN.<br />

Tumor mutational analysis and therapy outcomes for<br />

patients (pts) with metastatic/unresectable locally advanced<br />

myoepithelial/metaplastic breast cancer treated with PI3K<br />

targeted therapy<br />

Moulder S, Wheler J, Albarracin C, Gilcrease M, Falchook G, Naing<br />

A, Hong D, Fu S, Piha-Paul S, Tsimberidou A, Janku F, Kurzrock R.<br />

University of Texas, MD Anderson <strong>Cancer</strong> Center.<br />

Panitumumab, Gemcitabine and Carboplatin in Triple-<br />

Negative Metastatic <strong>Breast</strong> <strong>Cancer</strong>: Preliminary Results of a<br />

Phase II Trial of the Sarah Cannon Research Institute<br />

Yardley DA, Ward P, Handricks C, Daniel B, Harwin W, Kannarkat<br />

G, Saez R, Shastry M, Chirwa T, Peacock N. Sarah Cannon Research<br />

Institute, Nashville, TN; Oncology Hematology Care, Cincinnati,<br />

OH; National Capital Clinical Research Consortium, Bethesda, MD;<br />

Chattanooga Oncology and Hematology Associates, Chattanooga,<br />

TN; Florida <strong>Cancer</strong> Specialists, Ft. Myers, FL; Peninsula <strong>Cancer</strong><br />

Institute, Newport News, VA; Texas Health Physician Group,<br />

Arlington, TX; Tennessee Oncolcogy, PLLC, Nashville, TN.<br />

Bevacizumab in metastatic breast cancer: a retrospective<br />

matched-pair analysis<br />

Gampenrieder SP, Romeder F, Muß C, Pircher M, Ressler S,<br />

Rinnerthaler G, Bartsch R, Sattlberger C, Mlineritsch B, Greil R.<br />

Paracelsus Medical University, Salzburg, Austria; Comprehensive<br />

<strong>Cancer</strong> Center Vienna, Medical University of Vienna, Austria; Hospital<br />

of Vöcklabruck, Vöcklabruck, Austria.<br />

P5-20-12 Aromatase inhibitor failure: predictors and time to first<br />

failure among women with metastatic ER+/HER2- breast<br />

cancer in the US<br />

Thomson E, Namjoshi M, Landsman-Blumberg P, Chu BC. Thomson<br />

Reuters, Washington, DC; Novartis Pharmaceuticals Corporation, East<br />

Hanover, NJ.<br />

P5-20-13 Preliminary report of a phase I/II study of entinostat (IND#NSC<br />

706995, /M275) and lapatinib (IND#NSC 727989) in patients<br />

with HER2-positive metastatic breast cancer in whom<br />

trastuzumab has failed<br />

Ueno NT, Jackson SA, Alvarez RH, Willey JS, Hortobagyi GN,<br />

Angulo-Gonzalez AM, Giordano SH, Booser DJ, Valero V. Morgan<br />

Welch Inflammatory <strong>Breast</strong> <strong>Cancer</strong> Research Program and Clinic,<br />

The University of Texas MD Anderson <strong>Cancer</strong> Center, Houston, TX;<br />

University of Texas, MD Anderson <strong>Cancer</strong> Center, Houston, TX.<br />

Treatment: Targeted Therapy - Adjuvant<br />

P5-21-01 pT1a,bpN0M0 breast cancer: clinicopathological<br />

characteristics and their impact on treatment decision. Central<br />

review of the prospective ODISSEE cohort<br />

Lacroix-Triki M, Radosevic-Robin N, Louis B, Roche-Comet I,<br />

Soubeyrand M-S, Bourgeois H, Chauvet M-P, Fourrier-Réglat A,<br />

Gligorov J, Peyrat J-P, Dalenc F, Belkacemi Y, Penault-Llorca F. Institut<br />

Claudius Regaud, Toulouse, France; Centre Jean Perrin, Clermont-<br />

Ferrand, France; Laboratoire d’Anatomie et Cytologie Pathologiques,<br />

Strasbourg, France; Institut Histo-Cyto-Pathologie, Le Bouscat,<br />

France; Cabinet CY-PATH, Villeurbanne, France; Clinique Victor Hugo,<br />

Le Mans, France; Centre Oscar Lambret, Lyon, France; Université<br />

Victor Segalen, Bordeaux, France; Hôpital Tenon, Paris, France; CHU<br />

Henri Mondor-UPEC, Créteil, France.<br />

P5-21-02 Hormone receptor status and endocrine therapy in a<br />

prospective observation study on trastuzumab (Herceptin®)<br />

in the adjuvant treatment of breast cancer<br />

Dall P, Friedrichs K, Petersen V, Hinke A, Brucker C, Schmidt P,<br />

von der Assen A, Jungberg P, Bohnsteen B. Städtisches Klinikum,<br />

Lüneburg, Germany; Krankenhaus Jerusalem, Mammazentrum,<br />

Hamburg, Germany; Praxis, Heidenheim, Germany; WiSP, Biostatistik,<br />

Langenfeld, Germany; Klinikum Nürnberg, Brustzentrum, Nürnberg,<br />

Germany; Praxis, Neunkirchen, Germany; Franziskus-Hospital<br />

Harderberg, Georgsmarienhütte, Germany; Praxis, Chemnitz,<br />

Germany; Praxis, Dessau-Rosslau, Germany.<br />

P5-21-03 Concurrent loco-regional radiotherapy and trastuzumab in<br />

early-stage breast cancer: Long term results of prospective<br />

single-institution study<br />

Jacob J, Belin L, Pierga J-Y, Vincent-Salomon A, Dendale R, Beuzeboc<br />

P, Cottu P-H, Campana F, Fourquet A, Kirova YM. Institut Curie,<br />

Paris, France.<br />

P5-21-04 Real-world use and effectiveness of adjuvant trastuzumab in<br />

2665 consecutive breast cancer patients<br />

Tjan-Heijnen VCG, Seferina SC, Lobbezoo DJA, Voogd AC, Dercksen<br />

MW, van den Berkmortel F, van Kampen RJW, van de Wouw<br />

AJ, Joore MA, Borm GF. Maastricht University Medical Centre,<br />

Maastricht, Limburg, Netherlands; GROW-School for Oncology<br />

and Developmental Biology, Maastricht University Medical Centre,<br />

Maastricht, Limburg, Netherlands; Máxima Medical Centre,<br />

Veldhoven, Brabant, Netherlands; Atrium Medical Centre Parkstad,<br />

Heerlen, Limburg, Netherlands; Orbis Medical Center, Sittard-Geleen,<br />

Limburg, Netherlands; VieCuri Medical Centre, Venlo, Limburg,<br />

Netherlands; Radboud University Medical Center, Nijmegen,<br />

Gelderland, Netherlands.<br />

www.aacrjournals.org<br />

65s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Ongoing Trials 3: Immunotherapy<br />

OT3-1-01 A pilot study of single-dose ipilimumab and/or cryoablation<br />

in women with early-stage breast cancer scheduled for<br />

mastectomy<br />

Diab A, McArthur HL, Solomon S, Comstock C, Maybody M, Sacchini<br />

V, Durack J, Gucalp A, Yuan J, Patil S, Thorne A, Sung J, Kotin A,<br />

Morris E, Brogi E, Morrow M, Wolchok J, Allison J, Hudis C, Norton L.<br />

Memorial Sloan-Kettering <strong>Cancer</strong> Center, New York, NY.<br />

OT3-1-02 Phase II randomized study of combination immunotherapy<br />

with or without Polysaccharide Krestin (PSK®) concurrently<br />

with a HER2 ICD peptide-based vaccine and trastuzumab in<br />

patients with stage IV breast cancer<br />

Childs JS, Higgins DM, Parker S, Reichow J, Lu H, Standish L, Disis ML,<br />

Salazar LG. University of Washington, Seattle, WA; Bastyr University,<br />

Kenmore, WA.<br />

Ongoing Trials 3: Survivorship<br />

OT3-2-01 Influence of strength and endurance training on selected<br />

physical and psychological parameters and on immune<br />

system, metabolism and circulating tumor cells during<br />

adjuvant chemotherapy<br />

Mundhenke C, Weisser B, Keller L, <strong>San</strong>ders L, Summa B, Duerkop J,<br />

Schmidt T, Jonat W. University of Kiel, SH, Germany; Institute of Sport<br />

Science, University of Kiel, SH, Germany; Comprehensive <strong>Cancer</strong><br />

Center North, Kiel, SH, Germany.<br />

OT3-2-02 The PREDICT Study ( Prospective, Randomized Early Detection<br />

and Intervention after <strong>Breast</strong> <strong>Cancer</strong>-Treatment, for women at<br />

risk of lymphedema)<br />

Taghian AG, Skolny MN, O’Toole J, Miller CL, Jammallo LS, Specht<br />

MC. Massachusetts General Hospital, Boston, MA.<br />

Ongoing Trials 3: Chemotherapy<br />

OT3-3-01 Eniluracil + 5-fluorouracil + leucovorin (EFL) vs. capecitabine<br />

phase 2 trial for metastatic breast cancer<br />

Rivera E, Chang JC, Semiglazov V, Gorbunova V, Manikhas A,<br />

Krasnozhon D, Kirby G, Spector T. Banner MD Anderson <strong>Cancer</strong><br />

Center, Gilbert, AZ; The Methodist Hospital Research Institute,<br />

Houston, TX; Road Clinical Hospital of the Russian Railways, St.<br />

Petersburg, Russian Federation; Russian Oncological Research<br />

Center n.s.Blokhin RAMS, Moscow, Russian Federation; City Clinical<br />

Oncology Center, St. Petersburg, Russian Federation; Institution<br />

Leningrad Regional Oncology Center, Leningrad Region, Russian<br />

Federation; Adherex Technologies, Inc., Research Triangle Park, NC.<br />

OT3-3-02 ADAPT - Adjuvant Dynamic marker-Adjusted Personalized<br />

Therapy trial optimizing risk assessment and therapy response<br />

prediction in early breast cancer<br />

Gluz O, Hofmann D, Kates RE, Harbeck N, Nitz U. West German Study<br />

Group, Moenchengladbach, NRW, Germany; Ev. Bethesda Hospital,<br />

Moenchengladbach, NRW, Germany; University Clinic Großhadern,<br />

Munich, Bavaria, Germany.<br />

OT3-3-03 Withdrawn<br />

OT3-3-04 ALOPREV : first cooling scalp trial for prevention of persisting<br />

alopecia after docetaxel for early breast cancer patients<br />

Bourgeois H, Soulié P, Lucas B, Mercier Blas A, Zannetti A, Delecroix<br />

V, L’haridon T, Blot E, Delaloge S, Grudé F. Clinique Victor Hugo, Le<br />

Mans, France; Observatoire dédié au <strong>Cancer</strong> Bretagne Pays de Loire,<br />

Angers, France; Institut de Cancérologie de l’Ouest, Angers Nantes,<br />

France; CHRU Morvan, Brest, France; CHP, Saint Grégoire, France;<br />

CHD, Cholet, France; Pôle Hospitalier Mutualiste, Saint Nazaire,<br />

France; CHD, La Roche sur Yon, France; Centre Hospitalier Bretagne<br />

Atlantique, Vannes, France; Clinique Océane, Centre Saint Yves,<br />

Vannes, France; Institut Gustave Roussy, Villejuif, France.<br />

OT3-3-05<br />

OT3-3-06<br />

OT3-3-07<br />

OT3-3-08<br />

OT3-3-09<br />

Neurotoxicity characterization phase II randomized study<br />

of nab-paclitaxel versus conventional paclitaxel as first-line<br />

therapy of metastatic HER2-negative breast cancer.<br />

An ONSOCUR Study Group<br />

Ciruelos E, Bueno C, Cantos B, Carrión R, Echarri M, Enrech S, García<br />

Saenz JA, Guerra JA, Lara MA, Martínez N, Rodríguez-Antona<br />

C, Domínguez-González C, <strong>San</strong>z JL, Baquero J, Cortés-Funes H,<br />

Sepúlveda JM. Hospital 12 de Octubre, Madrid, Spain; Hospital<br />

Universitario Ramón y Cajal, Madrid, Spain; Hospital Universitario<br />

del Sureste, Arganda del Rey (Madrid), Spain; Hospital Universitario<br />

Clínico <strong>San</strong> Carlos, Madrid, Spain; Hospital Universitario Severo<br />

Ochoa, Leganés (Madrid), Spain; Hospital Universitario Puerta<br />

de Hierro Majadahonda, Majadahonda (Madrid), Spain; Hospital<br />

Universitario Infanta Cristina, Parla (Madrid), Spain; Hospital<br />

Universitario Infanta Leonor, Madrid, Spain; Hospital Universitario de<br />

Getafe, Getafe (Madrid), Spain; Hospital Universitario de Fuenlabrada,<br />

Fuenlabrada (Madrid), Spain; APICES, Madrid, Spain; Celgene, Madrid,<br />

Spain; Centro Nacional de Investigaciones Oncologicas,<br />

Madrid, Spain.<br />

NeoEribulin: A Phase II, non-randomized, open-label, singlearm,<br />

multicenter, exploratory pharmacogenomic study of<br />

single agent eribulin as neoadjuvant treatment for operable<br />

Stage I-II HER2 non-overexpressing breast cancer<br />

Prat A, Llombart A, de la Peña L, Di Cosimo S, Ortega V, Rubio I,<br />

Muñoz E, Harbeck N, Cortés J. Vall d’Hebron University Hospital,<br />

Barcelona, Spain; Hospital Arnau de Vilanova, Valencia, Spain; SOLTI<br />

<strong>Breast</strong> <strong>Cancer</strong> Research Group, Barcelona, Spain; Istituto Nazionale<br />

dei Tumori, Milan, Italy; Brustzentrum am Klinikum der Universität<br />

München, Munich, Germany.<br />

Neoadjuvant bevacizumab with weekly nanoparticle<br />

albumin bound (nab)-paclitaxel plus carboplatin followed by<br />

doxorubicin plus cyclophosphamide (AC) for triple negative<br />

breast cancer<br />

Snider JN, Allen JW, Young R, Schwartzberg L, Javed Y, Jahanzeb<br />

M, Sachdev JC. University of Tennessee, Memphis, TN; The West<br />

Clinic, Memphis, TN; The Center for <strong>Cancer</strong> and Blood Disorders, Fort<br />

Worth, TX.<br />

Eribulin/Cyclophosphamide versus Docetaxel/<br />

Cyclophosphamide as Neoadjuvant Therapy in Locally<br />

Advanced HER2-Negative <strong>Breast</strong> <strong>Cancer</strong>: A Randomized Phase<br />

II Trial of the Sarah Cannon Research Institute<br />

Yardley DA, Hainsworth JD, Shastry M, Finney L, Burris HA. Tennessee<br />

Oncology, PLLC, Nashville, TN; Sarah Cannon Research Institute,<br />

Nashville, TN.<br />

ARTemis trial - randomised trial with neo-adjuvant<br />

chemotherapy for patients with early breast cancer<br />

Earl HM, Blenkinsop C, Grybowicz L, Vallier A-L, Cameron DA,<br />

Bartlett JMS, Murray N, Caldas C, Thomas J, Dunn JA, Higgins HB,<br />

Hiller L, Hayward L. University of Cambridge, United Kingdom;<br />

NIHR Cambridge Biomedical Research Centre, Cambridge, United<br />

Kingdom; University of Warwick, Coventry, United Kingdom;<br />

Cambridge University Hospitals NHS Foundation Trust, Cambridge,<br />

United Kingdom; Edinburgh University, Edinburgh, United Kingdom;<br />

University of Edinburgh, United Kingdom; Royal Adelaide Hospital,<br />

Adelaide, SA, Australia; <strong>Cancer</strong> Research UK Cambridge Research<br />

Institute, Cambridge, United Kingdom; Western General Hospital,<br />

Edinburgh, United Kingdom.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012 66s <strong>Cancer</strong> Research


December 4–8, 2012<br />

Program Schedule<br />

OT3-3-10 ASTER 70s: Benefit of adjuvant chemotherapy for oestrogen<br />

receptor-positive HER2-negative breast carcinoma in women<br />

over 70 according to Genomic Grade. A French UNICANCER<br />

Geriatric Oncology Group (GERICO) and <strong>Breast</strong> Group (UCBG)<br />

multicentre phase III trial<br />

Brain E, Baffert S, Bonnefoi HR, Cottu P, Girre V, Lacroix-Triki M,<br />

Latouche A, Leger-Falandry C, Peyro Saint Paul HP, Rollot F, Romieux<br />

G, Servent V, Orsini C, Bonnetain F. Institut Curie, Saint-Cloud, France;<br />

Institut Curie, Paris, France; Institut Bergonié, Bordeaux, France;<br />

Centre Hospitalier La Roche sur Yon, La Roche sur Yon, France;<br />

Institut Claudius Regaud, Toulouse, France; CNAM, Paris, France;<br />

Centre Hospitalier Lyon Sud, Lyon, France; Ipsogen SA, Marseille,<br />

France; Centre Val d’Aurelle, Montpellier, France; Centre Oscar<br />

Lambret, Lille, France; R&D Unicancer, Paris, France; Centre Georges<br />

François Leclerc, Dijon, France.<br />

OT3-3-11 A randomized phase III trial comparing nanoparticle-based<br />

paclitaxel with solvent-based paclitaxel as part of neoadjuvant<br />

chemotherapy for patients with early breast cancer<br />

(GeparSepto) GBG 69<br />

Untch M, Jackisch C, Blohmer J-U, Costa S-D, Denkert C, Eidtmann H,<br />

Gerber B, Hanusch C, Hilfrich J, Huober J, Kuemmel S, Schneeweiss<br />

A, Paepke S, Loibl S, Nekljudova V, von Minckwitz G. Helios Kliniken<br />

Berlin; Klinikum Offenbach; <strong>San</strong>kt-Gertrauden Berlin; University<br />

Magdeburg; Charite Berlin; University Kiel; University Rostock;<br />

Rotkreuzklinikum Muenchen; Eilenriedeklinik Dueseldorf; University<br />

Duesseldorf; Kliniken Essen Mitte; Frauenklinik Muenchen; German<br />

<strong>Breast</strong> Group, Neu-Isenburg.<br />

Ongoing Trials 3: Response Prediction<br />

OT3-4-01 PROMIS: Prospective Registry Of MammaPrint in breast cancer<br />

patients with an Intermediate recurrence Score<br />

Soliman H, Untch S, Stork-Sloots L. Moffitt <strong>Cancer</strong> Center; Agendia<br />

Inc; Agendia NV.<br />

OT3-4-02 MINT I: Multi-Institutional Neo-adjuvant Therapy,<br />

MammaPrint Project I<br />

Cox C, Blumencranz P, Reintgen D, Saez R, Howard N, Gibson J,<br />

Stork-Sloots L, Glück S. University of South Florida; Morton Plant<br />

Hospital; Florida Hospital North Pinellas; Plano <strong>Cancer</strong> Institute;<br />

Agendia Inc; Agendia NV; Miller School of Medicine,<br />

University of Miami.<br />

OT3-4-03 Prospective neo-adjuvant registry trial linking MammaPrint,<br />

subtyping and treatment response: Neo-adjuvant <strong>Breast</strong><br />

Registry – Symphony Trial (NBRST)<br />

Whitworth P, Gittleman M, Akbari S, Nguyen B, Baron P, Rotkiss M,<br />

Beatty J, Gibson J, Stork-Sloots L, de Snoo F, Beitsch P. Nashville<br />

<strong>Breast</strong> Center; <strong>Breast</strong> Care Specialists; Virginia Hospital Center;<br />

Todd <strong>Cancer</strong> Institute, Long Beach Memorial Medical Center;<br />

<strong>Cancer</strong> Specialists of Charleston; Northern Indiana <strong>Cancer</strong> Research<br />

Consortium; The <strong>Breast</strong> Place Charleston; Agendia Inc; Agendia NV;<br />

Dallas Surgical Group.<br />

OT3-4-04 InVite: an observational pilot study evaluating the feasibility<br />

of genome-wide association studies using self-reported data<br />

from patients with metastatic breast cancer treated with<br />

bevacizumab<br />

Leyland-Jones B, Faoro L, Barnholt K, Kiefer A, Yager S, Yi J, Turner B,<br />

Keane A, Wang L, Eriksson N, Milián ML, O’Neill V. Genentech, Inc.;<br />

23andMe; <strong>San</strong>ford Research/USD.<br />

OT3-4-05 Use of early CIRculating tumor CElls count changes to guide<br />

the use of chemotherapy in advanced metastatic breast cancer<br />

patients: the CirCe01 randomized trial<br />

Bidard F-C, Asselain B, Baffert S, Brain E, Campone M, Delaloge S,<br />

Bachelot T, Tubiana-Mathieu N, Pelissier S, Pierga J-Y. Institut Curie;<br />

Institut de Cancérologie de l’Ouest; Institut Gustave Roussy, Villejuif;<br />

Centre Léon Bérard, Lyon; CHU Limoges.<br />

OT3-4-06<br />

Circulating tumor cells to guide the choice between<br />

chemotherapy and hormone therapy as first line treatment for<br />

hormone receptors positive metastatic breast cancer patients:<br />

the STIC CTC METABREAST trial<br />

Bidard F-C, Baffert S, Hajage D, Brain E, Armanet S, Simondi C, Mignot<br />

L, Asselain B, Tresca P, Pierga J-Y. Institut Curie, France.<br />

Saturday, December 8, 2012<br />

6:45 am–9:00 am<br />

Registration<br />

Bridge Hall<br />

7:00 am–8:30 am<br />

POSTER SESSION 6 & continental breakfast<br />

Exhibit Hall C<br />

Tumor Cell and Molecular Biology: Metabolism and <strong>Breast</strong> <strong>Cancer</strong><br />

P6-01-01 Metastatic and non-metastatic isogenic breast cancer cells<br />

lines show different metabolic signatures in response to<br />

intermittent hypoxia and transient glutamine/glucose<br />

deprivation<br />

Simoes RV, Ackerstaff E, Serganova I, Kruchevsky N, Sukenick G,<br />

Blasberg R, Koutcher JA. Memorial Sloan-Kettering <strong>Cancer</strong> Center,<br />

New York, NY; Cornell University, New York, NY.<br />

P6-01-02 Adipose tissue from breast cancer patients with the metabolic<br />

syndrome promotes proliferation and invasion of tumor cells<br />

and influences expression of genes involved in carcinogenesis<br />

McGarrigle SA, Carroll PA, Healy LA, Boyle T, Pidgeon GP, Kennedy<br />

MJ, Connolly EM. Trinity College Dublin and St. James’s Hospital,<br />

Dublin, Ireland.<br />

P6-01-03 Decreasing the metastatic potential in Triple Negative <strong>Breast</strong><br />

<strong>Cancer</strong> through the miR-17 cluster<br />

Jin L, Simone B, <strong>San</strong>o Y, Lim M, Zhao S, Savage JE, Baserga<br />

R, Camphausen K, Simone NL. Thomas Jefferson University,<br />

Philadelphia, PA; National <strong>Cancer</strong> Institute, Bethesda, MD.<br />

P6-01-04 DDB2 a new regulator of metabolism and cell death in human<br />

breast tumor cells<br />

Klotz R, Besancenot V, Brunner E, Becuwe P, Grandemange S.<br />

Université de Lorraine, Vandoeuvre les Nancy, France.<br />

P6-01-05 Enhancement of 18 F-FDG uptake and glycolysis by epidermal<br />

growth factor via PI3K activation in T47D breast cancer cells<br />

Lee EJ, Park JW, Cung Q, Jung K-H, Lee JH, Paik J-Y, Lee K-H.<br />

Samsung Medical Center, Sungkyunkwan University School of<br />

Medicine, Seoul, Korea.<br />

Tumor Cell and Molecular Biology: Microenvironment - Stromal-Epithelial<br />

Interactions<br />

P6-02-01 Apoptotic cell clearance lies at the interface of post-lactational<br />

involution and breast cancer<br />

Cook RS, Stanford JC, Earp S. Vanderbilt University; University of<br />

North Carolina.<br />

P6-02-02 Altered matrix homeostasis regulates estrogen biosynthesis in<br />

adipose tissue<br />

Ghosh S, Ashcraft K, Li R. UT Health Science Center at<br />

<strong>San</strong> <strong>Antonio</strong>, TX.<br />

P6-02-03 Mouse Models Of <strong>Breast</strong> <strong>Cancer</strong> Identify Oncogene-Specific<br />

Stroma Associated With Human <strong>Breast</strong> <strong>Cancer</strong> Molecular<br />

Subtypes<br />

Saleh SM, Laferriere J, Cory S, Souleimanova M, Zacksenhaus E,<br />

Muller W, Hallett M, Park M. McGill University, Montreal, QC, Canada;<br />

Toronto General Hospital, Toronto, ON, Canada.<br />

www.aacrjournals.org<br />

67s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P6-02-04 TMEM (Tumor MicroEnvironment of Metastasis) in human<br />

breast cancer is a blood vessel associated intravasation<br />

microenvironment unrelated to lymphatics<br />

Ginter PS, Robinson BD, D’Alfonso TM, Oktay MH, Gertler FB, Rohan<br />

TE, Condeelis JS, Jones JG. Weill Cornell Medical College, New York,<br />

NY; Albert Einstein College of Medicine, Bronx, NY; Massachusetts<br />

Institute of Technology, Cambridge, MA.<br />

P6-02-05 A novel culturing 3-D model to evaluate the role of tumor<br />

microenvironment in IBC<br />

Dong X, Franco-Barraza J, Mu Z, Alpaugh RK, Cristofanilli M,<br />

Cukierman E. Fox Chase <strong>Cancer</strong> Center, Philadelphia, PA.<br />

P6-02-06 A 3D tri-culture model of normal mammary gland. A tool for<br />

breast cancer initiation studies<br />

Nash CE, Holliday DL, Mavria G, Tomlinson DC, Hanby AM, Speirs V.<br />

Leeds Institute of Molecular Medicine, The University of Leeds, Leeds,<br />

West Yorkshire, United Kingdom.<br />

P6-02-07 In vitro 3D Model of <strong>Breast</strong> Tumor Stroma<br />

Jaganathan H, Mitra S, Dave B, Godin B. The Methodist Hospital<br />

Research Institute, Houston, TX.<br />

P6-02-08 Molecular drivers of adipogenotoxicosis in breast tumorassociated<br />

adipose<br />

Ellsworth RE, Field LA, van Laar R, Deyarmin B, Hooke JA, Shriver CD.<br />

Windber Research Institute, Windber, PA; Signal Genetics, New York,<br />

NY; Walter Reed National Military Medical Center, Bethesda, MD;<br />

Henry M. Jackson Foundation, Windber, PA.<br />

P6-02-09 Role of HGF in obesity-associated tumorigenesis: C3(1)-Tag<br />

mice as a model for human basal-like breast cancer<br />

Sundaram S, Freemerman AJ, Hamilton J, McNaughton K, Galanko<br />

JA, Darr DB, Perou CM, Troester MA, Makowski L. University of North<br />

Carolina at Chapel Hill, Chapel Hill, NC.<br />

Tumor Cell and Molecular Biology: Mammary Development and<br />

Differentiation<br />

P6-03-01 Molecular Dissection of <strong>Breast</strong> Luminal Cell Transcription<br />

Factor Networks<br />

Bargiacchi FG, Earp HS, Perou CM. University of North Carolina<br />

Chapel Hill, NC.<br />

Tumor Cell and Molecular Biology: Endocrine Therapy and Resistance<br />

P6-04-01 Global analysis of breast cancer metastasis suggests<br />

cellular reprogramming is central to the endocrine resistant<br />

phenotype<br />

Bolger JC, McCartan D, Walsh CA, Hao Y, Hughes E, Byrne C, Hill ADK,<br />

O’Gaora P, Young LS. Royal College of Surgeons in Ireland, Dublin 2,<br />

Ireland; University College Dublin, Ireland.<br />

P6-04-02 Final progression-free survival analysis of BOLERO-2: a phase<br />

III trial of everolimus for postmenopausal women with<br />

advanced breast cancer<br />

Piccart M, Baselga J, Noguchi S, Burris H, Gnant M, Hortobagyi G,<br />

Mukhopadhyay P, Taran T, Sahmoud T, Rugo H. Institut Jules Bordet,<br />

Université Libre de Bruxelles, Brussels, Belgium; Massachusetts<br />

General Hospital <strong>Cancer</strong> Center and Harvard Medical School, Boston,<br />

MA; Osaka University, Osaka, Japan; Sarah Cannon Research Institute,<br />

Nashville, TN; Comprehensive <strong>Cancer</strong> Center, Medical University<br />

of Vienna, Vienna, Austria; The University of Texas MD Anderson<br />

<strong>Cancer</strong> Center, Houston, TX; Novartis Pharmaceuticals Corporation,<br />

East Hanover, NJ; University of California, <strong>San</strong> Francisco Helen Diller<br />

Family Comprehensive <strong>Cancer</strong> Center, UCSF, <strong>San</strong> Francisco, CA.<br />

P6-04-03<br />

P6-04-04<br />

P6-04-05<br />

P6-04-06<br />

P6-04-07<br />

P6-04-08<br />

P6-04-09<br />

P6-04-10<br />

P6-04-11<br />

Changes in breast tumor metabolism and estradiol<br />

binding as measured by FES PET in patients treated with<br />

the histone deacetylace inhibitor vorinostat and aromatase<br />

inhibitor therapy<br />

Linden HM, Kurland BF, Specht JM, Vijayakrishn GK, Gralow JR,<br />

Peterson LM, Schubert EK, Link JM, David MA, Eary JF, Krohn<br />

KA. University of Washington, Seattle, WA; Fred Hutchinson<br />

<strong>Cancer</strong> Research Center, Seattle, WA; University of Pennsylvania,<br />

Philadelphia, PA.<br />

Upregulation of the androgen agonist prosaposin in<br />

aromatase inhibitor resistant breast cancer; mediation by the<br />

developmental protein HOXC11<br />

McIlroy M, Hao Y, Bane FT, Young L, O’Gaora P. Royal College<br />

of Surgeons in Ireland, Dublin, Ireland; University College<br />

Dublin, Ireland.<br />

Tamoxifen dose escalation based on endoxifen level:<br />

a prospective trial with genotyping, phenotyping and<br />

pharmacokinetics over 4 months<br />

Zaman K, Dahmane E, Perey L, Bodmer A, Anchisi S, Wolfer A,<br />

Galmiche M, Stravodimou A, Buclin T, Eap C, Decosterd L, Csajka<br />

C, Leyvraz S. University Hospital CHUV, Lausanne, Switzerland;<br />

Ensemble Hospitalier de la Côte, Morges, Switzerland; University<br />

Hospital CHUV, University of Geneva, Lausanne, Switzerland;<br />

University Hospital, Geneva, Switzerland; Hôpital Cantonal, Sion,<br />

Switzerland.<br />

Inverse regulation of Neuregulin1 and HER-3 during<br />

treatment with aromatase inhibitors of estrogen receptorpositive<br />

breast cancer<br />

Flågeng MH, Larionov A, Geisler J, Dixon JM, Lønning PE, Mellgren<br />

G. Haukeland University Hospital, Bergen, Norway; University<br />

of Edinburgh, United Kingdom; Akershus University Hospital,<br />

Lørenskog, Norway; University of Bergen, Norway.<br />

Significance and therapeutic potential of PELP1-mTOR axis in<br />

breast cancer progression and therapy resistance<br />

Gonugunta VK, Cortez V, Sareddy GR, Roy SS, Zhang H, Tekmal<br />

RR, Vadlamudi RK. UTHSCSA, <strong>San</strong> <strong>Antonio</strong>, TX; Shantou University<br />

Medical College, Shantou, China.<br />

FOXA1 expression: regulated by EZH2 and associated with<br />

favorable outcome to tamoxifen in advanced breast cancer<br />

Reijm EA, Helmijr JCA, Soler CMJ, Beekman R, Gerritse FL, Pragervan<br />

der Smissen WJC, Ruigrok-Ritstier K, van IJcken WFJ, Stevens<br />

MG, Smid M, Look MP, Meijer ME, Sieuwerts AM, Sleijfer S, Foekens<br />

JA, Berns EMJJ, Jansen MPHM. Erasmus MC - Daniel den Hoed,<br />

Rotterdam, Netherlands; Erasmus MC, Rotterdam, Netherlands.<br />

Lack of response to aromatase inhibitors involves<br />

distinct mechanisms<br />

Turnbull AK, Larionov AA, Renshaw L, Kay C, Sims AH, Dixon JM.<br />

University of Edinburgh, United Kingdom.<br />

Comprehensive gene and protein assessment of the role<br />

of Her2 in the response to neoadjuvant Letrozole suggests<br />

patients without amplification may also benefit from anti-<br />

Her2 treatment<br />

Webber V, Turnbull AK, Larionov AA, Sims AH, Harrison D, Renshaw<br />

L, Dixon JM. Melville Trust for Care and Cure of <strong>Cancer</strong>, Edinburgh;<br />

Western General Hospital, Edinburgh.<br />

Combination of PI3K-AKT-mTOR and MEK-ERK pathway<br />

inhibitors overcome acquired resistance to letrozole in ER+<br />

breast cancer models<br />

De P, Sun Y, Friedman LS, Chen S, Dey N, Leyland-Jones B. <strong>San</strong>ford<br />

Research/USD, Sioux Falls, SD; Genentech, Inc., South <strong>San</strong> Francisco,<br />

CA; Beckman Research Institute of the City of Hope, Duarte, CA.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012 68s <strong>Cancer</strong> Research


December 4–8, 2012<br />

Program Schedule<br />

P6-04-12<br />

P6-04-13<br />

P6-04-14<br />

P6-04-15<br />

P6-04-16<br />

P6-04-17<br />

P6-04-18<br />

P6-04-19<br />

P6-04-20<br />

P6-04-21<br />

P6-04-22<br />

P6-04-23<br />

Novel selective estrogen receptors degraders regress tumors<br />

in pre-clinical models of endocrine-resistant breast cancer<br />

Hager JH, Darimont B, Joseph J, Govek S, Grillot K, Aparicio A,<br />

Bischoff E, Kahraman M, Kaufman J, Lai A, Lee K-J, Lu N, Nagasawa J,<br />

Prudente R, Qian J, Sensintaffar J, Shao G, Heyman R, Rix P, Smith ND.<br />

Aragon Pharmaceuticals, <strong>San</strong> Diego, CA.<br />

HOXB7 functions as a co-activator of estrogen receptor in<br />

the development of tamoxifen resistance<br />

Jin K, Teo WW, Yoshida T, Park S, Sukumar S. Johns Hopkins<br />

University School of Medicine, Baltimore, MD.<br />

Targeting the PI3K/mTOR pathway in patient-derived<br />

xenograft models of endocrine resistant luminal breast cancer<br />

Cottu PH, Bagarre T, Assayag F, Bièche I, Chateau-Joubert S, Fontaine<br />

J-J, Decaudin D, Slimane K, Vincent-Salomon A, Marangoni E.<br />

Institut Curie, Paris, France; Institut Curie, Saint-Cloud, France; Ecole<br />

Veterinaire d’Alfort, Maisons-Alfort, France; Novartis Pharma, Rueil<br />

Malmaison, France.<br />

The involvement of LMTK3 in endocrine resistance is mediated<br />

via multiple signaling pathways<br />

Giamas G, Xu Y, Filipovic A, Grothey A, Coombes CR, Stebbing J.<br />

Imperial College, London, United Kingdom.<br />

The role of RIP140 and FOXA1 in breast cancer endocrine<br />

sensitivity and resistance<br />

Harada-Shoji N, Coombes RC, Lam EW-F. Imperial College London,<br />

United Kingdom.<br />

The androgen metabolite-dependent growth in hormone<br />

receptor positive breast cancer as a novel aromatase inhibitorresistance<br />

mechanism<br />

Hanamura T, Niwa T, Nishikawa S, Konno H, Ghono T, Kobayashi Y,<br />

Kurosumi M, Takei H, Yamaguchi Y, Ito K-I, Hayashi S-I. Graduate<br />

School of Medicine, Tohoku University, Sendai, Japan; Shinshu<br />

University School of Medicine, Matsumoto, Japan; Saitama <strong>Cancer</strong><br />

Center, Saitama, Japan.<br />

Evaluation of the molecular mechanisms behind fulvestrant<br />

resistant breast cancer<br />

Kirkegaard T, Hansen SK, Reiter BE, Sorensen BS, Lykkesfeldt AE.<br />

Danish <strong>Cancer</strong> Society Research Center, Copenhagen, Denmark;<br />

Aarhus University Hospital, Aarhus, Denmark.<br />

Neutralizing antibody to human GP88 (progranulin) restores<br />

sensitivity to tamoxifen and inhibits breast tumor growth in<br />

mouse xenografts<br />

Serrero G, Dong J, Hayashi J. A&G Pharmaceutical Inc, Columbia, MD.<br />

Endocrine resistance in invasive lobular carcinoma cells<br />

parallels unique estrogen-mediated gene expression<br />

Sikora MJ, Luthra S, Chandran UR, Dabbs DJ, Welm AL, Oesterreich S.<br />

University of Pittsburgh <strong>Cancer</strong> Institute, Pittsburgh, PA; University of<br />

Pittsburgh, PA; Magee-Womens Hospital, Pittsburgh, PA; University<br />

of Utah Huntsman <strong>Cancer</strong> Institute, Salt Lake City, UT.<br />

AIB1 expression specifically predicts breast cancer patient<br />

response to aromatase inhibitor therapy<br />

Vareslija D, O’Hara J, Tibbitts P, McBryan J, Hao Y, Hill A, Young L.<br />

Royal College of Surgeons Ireland, Dublin, Ireland.<br />

Regulation of Notch localization by endocrine therapy<br />

in Estrogen Receptor positive breast cancer cells: Clinical<br />

implications for endocrine resistance<br />

Espinoza I, Caskey M, Baker RC, Miele L. University of Mississippi,<br />

Jackson, MS.<br />

Targeting of endocrine therapy resistance through combined<br />

mTOR and histone deacetylase inhibition<br />

Ordentlich P, Flechsig S, Hoffmann J. Syndax Pharmaceuticals,<br />

Waltham, MA; EPO-GmbH Berlin, Berlin, Germany.<br />

P6-04-24 Overexpression of protein kinase C alpha differentially<br />

activates transcription factors in T47D breast cancer cells<br />

in the presence of 17β-estradiol both in the 2D and 3D<br />

environments<br />

Pham TND, Asztalos S, Weiss MS, Shea LD, Tonetti DA. University of<br />

Illinois at Chicago, IL; Northwestern University, Evanston, IL.<br />

P6-04-25 Genomic Deregulation and Therapeutic Role of Nemo-like<br />

Kinase in Luminal B <strong>Breast</strong> <strong>Cancer</strong><br />

Cao X, Qin L, Kim J-A, Tan Y, Wang X, Schiff R. Lester & Sue Smith<br />

<strong>Breast</strong> Center, Baylor College of Medicine, Houston, TX.<br />

P6-04-26 Tamoxifen may block estrogen induced secretion of certain<br />

cytokines to interrupt tumor associated macrophage<br />

infiltration in breast cancer<br />

Ding J, Chen CM, Jin W, Shao Z, Wu J. <strong>Breast</strong> <strong>Cancer</strong> Institute, Fudan<br />

University Shanghai <strong>Cancer</strong> Center, Shanghai, China; Shanghai<br />

Medical College, Fudan University, Shanghai, China.<br />

P6-04-27 EBAG9 immunoreactivity is a potential prognostic factor<br />

for poor outcome of breast cancer patients with adjuvant<br />

tamoxifen therapy<br />

Shigekawa T, Ijichi N, Ikeda K, Miyazaki T, Horie-Inoue K, Shimizu C,<br />

Saji S, Aogi K, Tsuda H, Osaki A, Saeki T, Inoue S. Research Center for<br />

Genomic Medicine, Saitama Medical University; International Medical<br />

Center, Saitama Medical University; Tokyo Metropolitan <strong>Cancer</strong> and<br />

Infectious Disease Center, Komagome Hospital; National <strong>Cancer</strong><br />

Center Hospital; Graduate School of Medicine, Kyoto University;<br />

National Shikoku <strong>Cancer</strong> Center; Graduate School of Medicine, The<br />

University of Tokyo.<br />

P6-04-28 Changes in BAG-1 expression level affect the Tamoxifeninduced<br />

apoptosis and Gefitinib-induced apoptosis in<br />

breast cancer cells<br />

Lu S, Zhuang X, Liu H. Tianjin Medical University <strong>Cancer</strong> Institute<br />

and Hospital, Tianjin, China; Tianjin Medical University, Minstry of<br />

Education, Tianjin, China.<br />

P6-04-29 Vitamin D induces expression of estrogen receptor and<br />

restores endocrine therapy response in estrogen receptornegative<br />

breast cancer<br />

<strong>San</strong>tos N, Diaz L, Ordaz D, Garcia J, Barrera D, Avila E, Halhali A,<br />

Medina H, Camacho J, Larrea F, Garcia R. Instituto Nacional de<br />

Ciencias Médicas y Nutrición Salvador Zubirán, México, DF, Mexico;<br />

Centro de Investigación y de Estudios Avanzados del I.P.N., México,<br />

DF, Mexico.<br />

P6-04-30 COUP-TFII suppresses NFκB activation in endocrine-resistant<br />

breast cancer cells<br />

Litchfield LM, Klinge CM. University of Louisville School of Medicine,<br />

Louisville, KY.<br />

Tumor Cell and Molecular Biology: Hormonal Factors and Receptors<br />

P6-05-01 Evaluation of the prognostic significance of androgen<br />

receptor (AR) expression in relation to ER expression in<br />

breast cancer (BC)<br />

Gucalp A, Datko FM, Patil S, Hedvat CV, Kalinsky K, Hudis CA, Traina<br />

TA, Moynahan ME. Memorial Sloan-Kettering <strong>Cancer</strong> Center;<br />

Columbia University Medical Center.<br />

P6-05-02 Endocrine biomarkers in response to AR-inhibition with<br />

bicalutamide for the treatment of AR(+), ER/PR(-) metastatic<br />

breast cancer (MBC) (TBCRC011)<br />

Gucalp A, Tolaney S, Isakoff SJ, Ingle J, Liu MC, Carey L, Blackwell<br />

KL, Rugo H, Nabell L, Forero A, Stearns V, Momen L, Gonzalez J,<br />

Akhtar A, Giri DD, Patil S, Feigin KN, Hudis CA, Traina TA. Memorial<br />

Sloan-Kettering <strong>Cancer</strong> Center; Dana Farber/Harvard <strong>Cancer</strong> Center;<br />

Mayo Clinic <strong>Cancer</strong> Center; Lombardi Comprehensive <strong>Cancer</strong> Center<br />

at Georgetown University; University of North Carolina Lineberger<br />

<strong>Cancer</strong> Center; University of California <strong>San</strong> Francisco Comprehensive<br />

<strong>Cancer</strong> Center; Duke University Medical <strong>Cancer</strong> Center; University<br />

of Alabama Comprehensive <strong>Cancer</strong> Center; The Sidney Kimmel<br />

Comprehensive <strong>Cancer</strong> Center at Johns Hopkins University.<br />

www.aacrjournals.org<br />

69s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P6-05-03<br />

P6-05-04<br />

P6-05-05<br />

P6-05-06<br />

P6-05-07<br />

P6-05-08<br />

P6-05-09<br />

P6-05-10<br />

P6-05-11<br />

P6-05-12<br />

P6-05-13<br />

Targeted inhibition of recurrent PIK3CA mutations<br />

synergizes with bicalutamide in AR-expressing triple negative<br />

breast cancer<br />

Lehmann BD, Bauer JA, Schafer JM, Tang L, Pendleton CS, <strong>San</strong>ders<br />

ME, Pietenpol JA. Vanderbilt, Nashville, TN.<br />

Expression of the Androgen Receptor in triple negative<br />

tumors and its modulation by receptor tyrosine kinases and<br />

downstream pathways<br />

Cuenca D, Montero JC, Morales JC, Prat A, Pandiella A, Ocana A.<br />

Albacete University Hospital, Albacete, Spain; <strong>Cancer</strong> Research<br />

Center, CIC-CSIC, Salamanca, Spain; Lineberger <strong>Cancer</strong> Center,<br />

University of North Carolina.<br />

Triple receptor comparison between primary breast cancer<br />

and metachronous or synchronous liver metastasis<br />

Tuyls S, Brouckaert O, Vanderstichele A, Vanderhaegen J, Amant<br />

F, Leunen K, Smeets A, Berteloot P, Van Limbergen E, Weltens C,<br />

Peeters S, Vanbeckevoort D, Floris G, Moerman P, Paridaens R,<br />

Wildiers H, Vergote I, Christiaens M-R, Neven P. University Hospitals<br />

Leuven, Belgium.<br />

Serum estradiol levels in postmenopausal ER+/PR- breast<br />

cancer are lower than those in postmenopausal ER+/PR+<br />

Yamamoto Y, Ibusuki M, Goto H, Murakami K, Iwase H. Kumamoto<br />

University Hospital, Kumamoto, Japan.<br />

Amplified in breast cancer 1 and ankyrin repeat containing<br />

cofactor 1 mediate estrogen induced repression of the<br />

ErbB2 oncogene<br />

Garee JP, Riegel AT. Georgetown University, Washington, DC.<br />

Significant heterogeneity in ERalpha and PR receptor status in<br />

different distant breast cancer metastases of the same patient<br />

Hoefnagel LDC, van der Groep P, Broers JE, van der Wall E, van Diest<br />

PJ. UMC Utrecht, Utrecht, Netherlands.<br />

Unravelling the global effect of estrogen on breast cancer cell<br />

proteome using quantitative proteomics<br />

Pavlou MP, Drabovich AP, Dimitromanolakis A, Diamandis EP.<br />

University of Toronto, ON, Canada; Mount Sinai Hospital, Toronto,<br />

ON, Canada; University Health Network, Toronto, ON, Canada;<br />

Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto,<br />

ON, Canada.<br />

Progestins exert divergent growth effects and regulate<br />

tumor-unique gene cohorts in patient derived breast cancer<br />

xenografts<br />

Sartorius CA, Finlay-Schultz J, Li C, Rosen RB, Hendricks P, Wisell J,<br />

Finlayson C, Elias A, Kabos P. University of Colorado Denver Anschutz<br />

Medical Campus, Aurora, CO.<br />

Leptin and Leptin receptor expression in human breast cancer<br />

Wazir U, Al Sarakbi W, Jiang WG, Mokbel K. The London <strong>Breast</strong><br />

Institute, The Princess Grace Hospital, London, United Kingdom;<br />

Cardiff University-Peking University Oncology Joint Institute, Cardiff,<br />

United Kingdom.<br />

Lack of Frequent Estrogen Receptor Mutation in Primary<br />

<strong>Breast</strong> Tumors<br />

Oesterreich S, Kitchens C, Gavin P, Wu C-C, Riehle K, Coarfa C,<br />

Edwards D, Schiff R, Milosavljevic A, Lee A. University of Pittsburgh<br />

<strong>Cancer</strong> Institute, Pittsburgh, PA; Baylor College of Medicine, Houston,<br />

TX; NSABP, Pittsburgh, PA.<br />

ESR1 gene amplification in breast cancer: influence of RNAse<br />

treatment on FISH results<br />

Moelans CB, Holst F, Hellwinkel O, Simon R, van Diest PJ. University<br />

Medical Center Utrecht, Netherlands; University Medical Center<br />

Hamburg Eppendorf, Hamburg, Germany.<br />

P6-05-14 Estrogen-induced genes in ductal carcinoma in situ(DCIS):<br />

their comparison with invasive ductal carcinoma<br />

Ebata A, Suzuki T, Takagi K, Miki Y, Onodera Y, Nakamura Y, Fujishima<br />

F, Ishida K, Watanabe M, Tamaki K, Ishida T, Ohuchi N, Sasano H.<br />

Tohoku University Graduate School of Medicine, Sendai, Miyagi,<br />

Japan; Tohoku University Hospital, Sendai, Miyagi, Japan.<br />

P6-05-15 Glucocorticoids has diverse effect on the tumorigenicity<br />

in estrogen receptor positive and estrogen receptor<br />

negative cancer cells<br />

Gao M, Yeh L-C, Chang C-P, Cheng A-L, Lu Y-S. National Taiwan<br />

University Hospital, Taipei, Taiwan.<br />

Tumor Cell and Molecular Biology: Tumor Cell and<br />

Molecular Biology - Other<br />

P6-06-01 Lobular involution reduces breast cancer risk through<br />

downregulation of invasive and proliferative cellular<br />

processes<br />

Radisky DC, Visscher DW, Stallings-Mann ML, Frost MH, Allers TM,<br />

Degnim AC, Hartmann LC. Mayo Clinic, Jacksonville, FL; Mayo Clinic,<br />

Rochester, MN.<br />

P6-06-02 Characterization of an exosome-associated apoptosisinducing<br />

activity produced by triple negative<br />

breast cancer cells<br />

Georgoulia NE, Iliopoulos D, Mitchison TJ. Harvard Medical School,<br />

Boston, MA; Dana Farber <strong>Cancer</strong> Institute, Boston, MA.<br />

P6-06-03 Caveolin-1: A potential mediator of RhoC GTPase driven<br />

Inflammatory breast cancer cell invasion<br />

Joglekar M, van Golen K. University of Delaware, Newark, Delaware.<br />

P6-06-04 Withdrawn<br />

P6-06-05 Down-regulation of the circadian factor Period 2 by<br />

the oncogenic E3 ligase Mdm2: Relevance of circadian<br />

components for cell cycle progression<br />

Liu J, Gotoh T, Vila-Caballer M, <strong>San</strong>tos CS, Yang J, Finkielstein CV.<br />

Virginia Tech, Blacksburg, VA.<br />

P6-06-06 The therapeutic potential and hazard of exposure to Cerbera<br />

odollam leaf extracts<br />

Chung F, Chan K, Lim YY, Li B. Monash University Sunway Campus,<br />

Subang Jaya, Selangor, Malaysia; Taylor’s University, Subang Jaya,<br />

Selangor, Malaysia.<br />

Prognostic and Predictive Factors: Prognostic and<br />

Predictive Factors - Other<br />

P6-07-01 Withdrawn<br />

P6-07-02 Prediction of oncotype DX® recurrence score using pathology<br />

generated equations<br />

Bhargava R, Klein ME, Shuai Y, Brufsky AM, Puhalla SL, Jankowitz<br />

R, Dabbs DJ. Magee-Womens Hospital of UPMC, Pittsburgh, PA;<br />

University of Wisconsin-Madison School of Medicine and Public<br />

Health, Madison, WI; University of Pittsburgh <strong>Cancer</strong> Institute,<br />

Pittsburgh, PA.<br />

P6-07-03 Risk classification of Early Stage <strong>Breast</strong> <strong>Cancer</strong> as Assessed by<br />

MammaPrint and Oncotype DX Genomic Assays<br />

Clough KB, Poulet B, Jamshidian F, Butler S, Svedman C, Levy E.<br />

L’Institut du Sein-Paris <strong>Breast</strong> Centre, Paris, Ile de France, France;<br />

Genomic Health, Inc., Redwood City, CA.<br />

P6-07-04 Evaluation of Adjuvant! Online for primary operable grade 2<br />

breast cancers with 10-year follow-up<br />

Van Calster B, Brouckaert O, Wildiers H, Christiaens M-R,<br />

Weltens C, Paridaens R, Van Limbergen E, Neven P. On behalf of<br />

Multidisciplinary <strong>Breast</strong> Centre, UZ Leuven.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012 70s <strong>Cancer</strong> Research


December 4–8, 2012<br />

Program Schedule<br />

P6-07-05<br />

P6-07-06<br />

P6-07-07<br />

P6-07-08<br />

P6-07-09<br />

P6-07-10<br />

P6-07-11<br />

Prognosis of 368 women with primary breast cancer during<br />

pregnancy: results from an international collaborative trial<br />

Amant F, von Minckwitz G, Han SN, Bontenbal M, Ring A, Giermek<br />

J, Fehm T, Wildiers H, Linn SC, Schlehe B, Neven P, Westenend<br />

PJ, Müller V, Van Calsteren K, Rack B, Nekljudova V, Harbeck N,<br />

Lenhard M, Witteveen PO, Kaufmann M, Van Calster B, Loibl S.<br />

Leuven <strong>Cancer</strong> Institute, University Hospitals Leuven, KU Leuven,<br />

Belgium; German <strong>Breast</strong> Group, Neu-Isenburg, Germany; BOOG<br />

Study Center, Amsterdam, Netherlands; Royal Sussex County<br />

Hospital, Brighton, United Kingdom; Institute in Warsaw <strong>Breast</strong><br />

<strong>Cancer</strong> and Reconstructive Surgery Clinic, Warsaw, Poland; University<br />

Women Hospital, Tübingen, Germany; University Women Hospital<br />

Heidelberg, Germany; University Medical Center Hamburg-<br />

Eppendorf, Hamburg, Germany; University Hospitals Leuven, KU<br />

Leuven, Belgium; Ludwigs Maximilian University, Frauenklinik<br />

Innenstadt, München, Germany; University Women Hospital<br />

Cologne, Germany; Klinik und Poliklinik für Frauenheilkunde und<br />

Geburtshilfe Klinikum der Universität München, Grosshadern,<br />

Germany; University Medical Center Utrecht, Netherlands; J.W.<br />

Goethe University, Frankfurt, Germany.<br />

Patterns of locoregional failure in the CALOR (Chemotheraphy<br />

as Adjuvant for Locally Recurrent <strong>Breast</strong> <strong>Cancer</strong>) Trial<br />

Wapnir IL, Gelber S, Lang I, Anderson SJ, Robidoux A, Martin M,<br />

Nortier JWR, Mamounas EP, Geyer CE, Maibach R, Gelber RD,<br />

Wolmark N, Aebi S. National Surgical Adjuvant <strong>Breast</strong> and Bowel<br />

Project (NSABP) Operations and Biostatistical Centers; Stanford<br />

University Medical School of Medicine; International <strong>Breast</strong><br />

<strong>Cancer</strong> Study Group; National Institute of Oncology; University of<br />

Pittsburgh Graduate School of Public Health; Centre Hospitalier de<br />

l’Université de Montréal; Hospital General Universitario Gregorio<br />

Marañón; Leiden University Medical Center, Leiden, Netherlands;<br />

Aultman Hospital; University of Texas Southwestern Medical Center;<br />

Allegheny <strong>Cancer</strong> Center at Allegheny General Hospital.<br />

Withdrawn<br />

Associations between lifestyle parameters and prognostic<br />

factors used for stratifying for adjuvant treatment in<br />

breast cancer<br />

Guldberg TL, Christensen S, Ravnsbaek A, Zachariae B, Jensen AB.<br />

Aarhus University Hospital, Aarhus, Denmark.<br />

Identification of Trophinin associated protein (TROAP) as a<br />

novel biological marker in breast cancer (BC): Co-expression of<br />

TROAP and TOPO2A predicts response of anthracycline based<br />

chemotherapy (ATC-CT)<br />

Abdel-Fatah TMA, Balls G, Miles AK, Moseley P, Green A, Rees R, Ellis<br />

IO, Chan SYT. Nottinigham City Hospital NHS Trust; The Van Geest<br />

<strong>Cancer</strong> Research Center, Nottingham Trent University; University of<br />

Nottingham.<br />

Luminal A vs. Basal-like <strong>Breast</strong> <strong>Cancer</strong>: time dependent<br />

changes in the risk of relapse in the absence of treatment<br />

Cheang MCU, Parker J, DeSchryver K, Snider J, Walsh T, Davies S,<br />

Prat A, Vickery T, Reed J, Zehnbauer B, Leung S, Voduc D, Nielsen<br />

T, Mardis E, Bernard P, Perou C, Ellis M. University of North Carolina<br />

at Chapel Hill, NC; University of British Columbia, Vancouver, BC,<br />

Canada; Washington University School of Medicine;<br />

University of Utah.<br />

Is the prognosis of lymphotropic invasive micropapillary<br />

carcinoma worse than invasive ductal carcinoma?: A<br />

population-based study of 645 patients<br />

Chen AC, Paulino AC, Schwartz MR, Rodriguez AA, Bass BL, Chang<br />

JC, Teh BS. Baylor College of Medicine, Houston, TX; The Methodist<br />

Hospital, Houston, TX.<br />

P6-07-12<br />

P6-07-13<br />

P6-07-14<br />

P6-07-15<br />

P6-07-16<br />

P6-07-17<br />

P6-07-18<br />

Akt2 expression is associated with good long-term prognosis<br />

in estrogen receptor positive breast cancer<br />

Fohlin H, Pérez-Tenorio G, Fornander T, Skoog L, Nordenskjöld B,<br />

Carstensen J, Stål O. Regional <strong>Cancer</strong> Center, Southeast Sweden,<br />

Linköping, Sweden; Faculty of Health Sciences, Linköping University,<br />

Linköping, Sweden; Karolinska University Hospital, Karolinska<br />

Institute, Stockholm, Sweden; Linköping University, Linköping,<br />

Sweden.<br />

Local relapse and survival<br />

Harland R, Prathap P, Lionaki A, Mahmood N. Royal Albert Edward<br />

Infirmary, Wigan, United Kingdom; Euxton Hall Hospital, Chorley,<br />

United Kingdom.<br />

Mutational and transcriptomic characterization of breast<br />

cancer (BC) arising in young patients (pts) and during<br />

pregnancy and their associations with long-term outcome<br />

Azim Jr HA, Peccatori FA, Loi S, Lambrechts D, Majjaj S, Renne<br />

G, Desmedt C, Rotmensz N, Michiels S, Dell’Orto P, Ignatiadis M,<br />

Goldhirsch A, Piccart M, Viale G, Sotiriou C. Universite Libre de<br />

Bruxelles, Brussels, Belgium; Institut Jules Bordet, Brussels, Belgium;<br />

European Institute of Oncology, Milan, Italy; University of Leuven,<br />

Belgium.<br />

The effect of taxanes in ER+ early breast cancer is likely to be<br />

mitigated by chromosomal instability<br />

A’Hern RP, Bliss JM, Szallasi Z, Johnston S, Roylance R, Swanton C.<br />

The Institute of <strong>Cancer</strong> Research, Sutton, United Kingdom; Technical<br />

University of Denmark, Lyngby, Denmark; Barts and The Royal<br />

London Hospital, London, United Kingdom; <strong>Cancer</strong> Research UK<br />

London Research Institute, London, United Kingdom; Royal Marsden<br />

NHS Foundation Trust and The Institute of <strong>Cancer</strong> Research, London,<br />

United Kingdom.<br />

Evaluation of circulating tumor cell as a marker of prognosis<br />

and efficacy in a randomized phase III study in HER2 negative<br />

metastatic breast cancer patients treated with capecitabine<br />

and docetaxel: JO21095 study<br />

Masuda N, Yamamoto D, Sato N, Sagara Y, Yamamoto Y, Saito M,<br />

Iwata H, Oura S, Watanabe J, Kuroi K. National Hospital Organization<br />

Osaka National Hospital, Osaka, Japan; Kansai Medical University<br />

Hirakata Hospital, Hirakata, Osaka, Japan; Niigata <strong>Cancer</strong> Center<br />

Hospital, Niigata, Japan; Sagara Hospital, Kagoshima, Japan;<br />

Kumamoto University Hospital, Kumamoto, Japan; Juntendo<br />

University Hospital, Bunkyo, Tokyo, Japan; Aichi <strong>Cancer</strong> Center<br />

Hospital, Nagoya, Aichi, Japan; Wakayama Medical University,<br />

Wakayama, Wakayama, Japan; Shizuoka <strong>Cancer</strong> Center, Nagaizumicho,<br />

Suntou-gun, Shizuoka, Japan; Tokyo Metropolitan <strong>Cancer</strong> and<br />

Infectious Diseases Center Komagome Hospital, Bunkyo,<br />

Tokyo, Japan.<br />

Proteomic screening of FFPE tissue identifies FKBP4 as an<br />

independent prognostic factor in hormone receptor positive<br />

breast cancers<br />

Hu J, Pohorelic B, Konno M, Price JT, Morris D, Krizman D, Magliocco<br />

AM, Klimowicz AC. Alberta Health Services, Calgary, AB, Canada;<br />

University of Calgary, AB, Canada; Monash University, Clayton, VIC,<br />

Australia; OncoPlex Diagnostics, Rockville, MD; H. Lee Moffitt <strong>Cancer</strong><br />

Center & Research Institute, Tampa, FL.<br />

Identification of Sperm Associated Antigen 5 (SPAG5) as a<br />

novel biological and predictive biomarker in <strong>Breast</strong> cancer<br />

Abdel-Fatah TMA, Ball G, Miles AK, Moseley P, Green A, Rees B, Ellis<br />

IO, Chan SYT. Nottingham City Hospital NHS Trust, Nottingham;<br />

The John van Geest <strong>Cancer</strong> Research Centre, Nottingham Trent<br />

University; University of Nottingham.<br />

www.aacrjournals.org<br />

71s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P6-07-19<br />

P6-07-20<br />

P6-07-21<br />

P6-07-22<br />

P6-07-23<br />

P6-07-24<br />

P6-07-25<br />

P6-07-26<br />

P6-07-27<br />

P6-07-28<br />

Prognostic relevance of PR and detection mode in Luminal<br />

Her-2 negative breast cancer<br />

Brouckaert O, Van Calster B, Paridaens R, Wildiers H, Van Limbergen<br />

E, Weltens C, Christiaens M-R, Neven P. On behalf of Multidisciplinary<br />

<strong>Breast</strong> Centre.<br />

Variables measured at Central Nervous System (CNS) relapse,<br />

but not Immunophenotype, identify groups of breast cancer<br />

patients with shorter post CNS-relapse survival<br />

Gómez HL, Pinto JA, Schwarz JL, Vigil CE, Vallejos CS. Instituto<br />

Nacional de Enfermedades Neoplásicas, Lima, Peru; Oncosalud,<br />

Lima, Peru.<br />

Signs and symptoms at central nervous system (CNS) relapse<br />

in breast cancer patients with Leptomeningeal carcinomatosis<br />

related with shorter survival after recurrence<br />

Gomez HL, Schwarz LJ, Vásquez J, Neciosup SP, Pinto JA, Vidaurre<br />

T, Ferreyros G, Vallejos CS. Instituto Nacional de Enfermedades<br />

Neoplásicas, Peru; Oncosalud, Peru.<br />

Association between SPARC mRNA expression, prognosis and<br />

response to neoadjuvant chemotherapy in early breast cancer<br />

(BC): a pooled in-silico analysis<br />

Azim Jr HA, Singhal S, Michiels S, Ignatiadis M, Djazouli K, Fumagalli<br />

D, Desmedt C, Piccart M, Sotiriou C. Universite Libre de Bruxelles,<br />

Brussels, Belgium; Institut Jules Bordet, Brussels, Belgium;<br />

Celgene International.<br />

Proportion of invasive micropapillary carcinoma lesion and<br />

primary breast cancer prognosis<br />

Takei J, Nakayama K, Yagata H, Hayashi N, Yoshida A, Ohde S, Suzuki<br />

K, Nakamura S, Yamauchi H. St. Luke’s International Hospital, Tokyo,<br />

Japan; Showa University School of Medicine, Tokyo, Japan.<br />

Prognostic Tools in Early <strong>Breast</strong> <strong>Cancer</strong>: Predicting benefit of<br />

adjuvant chemotherapy<br />

Parkes EE, Davidson C, James CR, Hanna GG. Northern Ireland <strong>Cancer</strong><br />

Centre, Belfast City Hospital, Belfast, United Kingdom; Queen’s<br />

University of Belfast, United Kingdom.<br />

Annexin expression and survival outcomes in women with<br />

breast cancer<br />

Dawood S, Gonzalez-Angulo AM, Liu S, Chen H, Li Y, Albarracin CT.<br />

MD Anderson <strong>Cancer</strong> Center, Houston, TX; Dubai Hospital, United<br />

Arab Emirates.<br />

Prognostic significance of the maximal value of the baseline<br />

standardized uptake value on fluorine 18 fluorodeoxyglucose<br />

positron emission tomography /computed tomography for<br />

predicting pathologic malignancy of operable breast cancer<br />

with neoadjuvant chemotherapy<br />

Kadoya T, Akimoto E, Emi A, Shigematsu H, Masumoto N, Okada M.<br />

Hiroshima University, Hiroshima, Japan.<br />

Body mass index and survival after breast cancer diagnosis<br />

in Japanese women<br />

Kawai M, Minami Y, Nishino Y, Ohuchi N, Kakugawa Y. Tohoku<br />

University Graduate School of Medicine, Sendai, Miyagi, Japan;<br />

Miyagi <strong>Cancer</strong> Center Research Institute, Natori, Miyagi, Japan;<br />

Miyagi <strong>Cancer</strong> Center Hospital, Natori, Miyagi, Japan.<br />

Assessment of T stage in the multiple breast carcinomas<br />

Gong G, Jo J-H, Lee HJ, Kang J. University of Ulsan College of<br />

Medicine, Asan Medical Center, Republic of Korea; University of<br />

Ulsan College of Medicine, Gangneung Asan Hospital, Republic<br />

of Korea; Seoul National University Bundang Hospital, Republic of<br />

Korea; Haeundae Paik Hospital, Inje University College of Medicine,<br />

Republic of Korea.<br />

P6-07-29 Independent prognostic value of age depends on breast<br />

cancer subtype<br />

Brouckaert O, Salihi R, Laenen A, Vanderhaegen J, Amant F, Leunen<br />

K, Smeets A, Berteloot P, Van Limbergen E, Weltens C, Moerman<br />

P, Peeters S, Paridaens R, Floris G, Wildiers H, Vergote I, Christiaens<br />

M-R, Neven P. UZ Leuven; Interuniversity Centre for Biostatistics and<br />

Statistical Bioinformatics.<br />

P6-07-30 The clinical significance of pathologic complete response<br />

using different definitions after neoadjuvant chemotherapy in<br />

HER2 positive breast cancer patients according to hormonal<br />

receptor status<br />

Tanioka M, Hirokaga K, Kitao A, Matsumoto K, Yoshida S, Miki M,<br />

Maekawa Y, Takao S, Negoro S. Hyogo <strong>Cancer</strong> Center, Akashi, Japan.<br />

P6-07-31 Moved to Poster Session 2, Thursday, December 6 7:00 AM -<br />

9:00 AM<br />

P6-07-32 Prognosis of metastatic breast cancer subtypes: the hormone<br />

receptor/HER2 positive subtype is associated with the most<br />

favorable outcome<br />

Tjan-Heijnen VCG, Lobbezoo DJA, van Kampen RJW, Voogd AC,<br />

Dercksen MW, van den Berkmortel F, Smilde TJ, van de Wouw<br />

AJ, Peters FPJ, van Riel JMGH, Peters NAJB, Borm GF. Maastricht<br />

University Medical Center, Maastricht, Netherlands; Maxima<br />

Medical Center, Eindhoven, Netherlands; Atrium Medical Center<br />

Parkstad, Heerlen, Netherlands; Jeroen Bosch Hospital, Den Bosch,<br />

Netherlands; Viecuri Medical Center, Venlo, Netherlands; Orbis<br />

Medical Center, Sittard, Netherlands; St Elisabeth Hospital, Tilburg,<br />

Netherlands; St Jans Hospital, Weert, Netherlands; Radboud<br />

University Medical Center, Nijmegen, Netherlands.<br />

P6-07-33 A clinicopathologic analysis of 45 patients with metaplastic<br />

breast cancer<br />

Verma S, Cimino-Mathews A, Figueroa Magalhaes MC, Zhang Z,<br />

Stearns V, Connolly RM. Sidney Kimmel Comprehensive <strong>Cancer</strong><br />

Center at Johns Hopkins, Baltimore, MD; St. Agnes Hospital,<br />

Baltimore, MD; Johns Hopkins Hospital, Baltimore, MD.<br />

P6-07-34 Disease-related outcomes with adjuvant chemotherapy in<br />

HER2 positive or triple negative T1a/bN0 breast cancers<br />

Shao T, Olszewski AJ, Boolbol SK, Migdady Y, Boachie-Adjei K, Sakr<br />

BJ, Klein P, Sikov W. Beth Israel Medical Center, Continuum <strong>Cancer</strong><br />

Centers of New York, New York, NY; The Warren Alpert Medical<br />

School of Brown University, Providence, RI.<br />

P6-07-35 Dragonfly Effect or Ironic Paradox: Prognostic Implication of<br />

Postsurgical Drain in <strong>Breast</strong> <strong>Cancer</strong> Patients<br />

Yin W, Lin Y, Shen Z, Shao Z-M, Lu J. Fudan University Shanghai<br />

<strong>Cancer</strong> Center, Shanghai, China.<br />

P6-07-36 A Prognostic Model For Triple-Negative <strong>Breast</strong> <strong>Cancer</strong> Patients<br />

Based On Node Status, Cathpesin-D And Ki-67<br />

Huang L, Liu Yb, Chen S, Zhou Rj, Liu Y, Shao Z-M. Fudan University<br />

Shanghai <strong>Cancer</strong> Hospital, Shanghai, China.<br />

P6-07-37 Impact of body mass index on recurrence risk in patients with<br />

ductal carcinoma in situ<br />

Klepczyk LC, Meredith RF, De Los <strong>San</strong>tos JF, Li Y, Keene KS. University<br />

of Alabama at Birmingham, AL.<br />

P6-07-38 The Value of Progesterone Receptor in Predicting the<br />

Recurrence Score for Hormone-Receptor–Positive Invasive<br />

<strong>Breast</strong> <strong>Cancer</strong> Patients<br />

Onoda T, Yamauchi H, Yagata H, Hayashi N, Yoshida A, Nakamura<br />

S. St Luke’s International Hospital, Tokyo, Japan; Showa University,<br />

Tokyo, Japan; Yokohama Asahi Central General Hospital,<br />

Kanagawa, Japan.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012 72s <strong>Cancer</strong> Research


December 4–8, 2012<br />

Program Schedule<br />

P6-07-39 Prognostic Value of Body Mass Index in Japanese <strong>Breast</strong><br />

<strong>Cancer</strong> Patients: A Collaborative Study by the Kobe <strong>Breast</strong><br />

<strong>Cancer</strong> Oncology Group and Hokkaido <strong>Cancer</strong> Center<br />

Shigeoka Y, Watanabe K, Takahashi M, Hirokaga K, Takao S, Miyashita<br />

M, Wakita K, Miyoshi Y, Okuno T, Kohno S, Kishimoto M, Kokufu I.<br />

Yodogawa Christian Hospital, Osaka, Japan; Hokkaido <strong>Cancer</strong> Center,<br />

Sapporo, Japan; Hyogo <strong>Cancer</strong> Center, Akashi, Japan; Konan Hospital,<br />

Kobe, Japan; Chayamachi <strong>Breast</strong> Clinic, Osaka, Japan; Hyogo College<br />

of Medicine, Nishinomiya, Japan; Kobe Urban <strong>Breast</strong> Clinic, Kobe,<br />

Japan; Kobe University, Kobe, Japan; Meiwa Hospital, Nishinomiya,<br />

Japan; Kokufu <strong>Breast</strong> Clinic, Takarazuka, Japan.<br />

P6-07-40 Initial Neutrophil-to-Lymphocyte ratio in primary<br />

breast cancer patients: a simple and useful biomarker as<br />

prognostic factor<br />

Han A, Noh H, Lee J. Yonsei University Wonju Medical School, Wonju,<br />

Gangwon, Republic of Korea.<br />

P6-07-41 Sub classification of early stage hormone positive (HP) Her-2<br />

negative breast cancer (BC) by immunohistochemistry (IHC)<br />

into intrinsic subtypes (IS) and their correlation with Oncotype<br />

Recurrence Score (RS)<br />

Mahesh S, Lane J, Ravichandran P. Summa Health Systems,<br />

Akron, OH.<br />

P6-07-42 Patients with Nipple-areola Paget’s Disease and Underlying<br />

Invasive <strong>Breast</strong> Carcinoma had a Very Poor Survival: a Matched<br />

Cohort Study<br />

Ling H, Xu X-L, Liu Z-B, Shao Z-M. Fudan University Shanghai <strong>Cancer</strong><br />

Center, Shanghai, China.<br />

P6-07-43 The Maximum Standardized Uptake Value of 18 F-FDG to<br />

Prognosticate Prognosis of Hormone-Receptor Positive<br />

Metastatic <strong>Breast</strong> <strong>Cancer</strong><br />

Hu X-C, Zhang (co-first author) J, Jia Z, Ragaz J, Zhang Y-J, Zhou M,<br />

Zhang Y-P. Fudan University Shanghai <strong>Cancer</strong> Center, Shanghai,<br />

China; School of Population and Public Health; University of British<br />

Columbia, Vancouver, Canada.<br />

P6-07-44 Overall survival and the prognostic factors among women<br />

with infiltrating lobular carcinoma of the breast in National<br />

<strong>Cancer</strong> Institute of Brazil<br />

Bello MA, Monteiro SO, Lima DT, Gomes DO, Thuler LCS, Carmo PAO,<br />

Carvalho RM, Pinto RR, Bergmann A. Instituto Nacional de Câncer,<br />

Rio de Janeiro, RJ, Brazil.<br />

P6-07-45 Molecular Morphology Based Genomic Signatures of<br />

Moderate Complexity Predict Pathologic Complete Response<br />

in HER2 Molecular <strong>Breast</strong> Carcinoma Class Patients Treated<br />

with Trastuzumab-based Preoperative Therapy<br />

Tubbs RR, Portier BP, Morrison L, Wang Z, Minca E, Lanigan C,<br />

Budd T. Cleveland Clinic, Cleveland, OH; Ventana Medical Systems,<br />

Tucson, AZ; Cleveland Clinic Tausig <strong>Cancer</strong> Center, Cleveland, OH.<br />

Psychosocial, Quality of Life, and Educational Aspects:<br />

Psychosocial Aspects<br />

P6-08-01 Perceptions of breast cancer risk, psychological adjustment<br />

and behaviors in adolescent girls at high-risk and populationrisk<br />

for breast cancer<br />

Bradbury AR, Patrick-Miller L, Egleston B, Schwartz L, Tuchman L,<br />

Moore C, Rauch P, <strong>San</strong>ds C, Shorter R, Rowan B, Malhotra S, van<br />

Decker S, Schmidheiser H, Sicilia P, Bealin L, Daly M. University<br />

of Pennsylvania, Philadelphia, PA; University of Chicago, IL; Fox<br />

Chase <strong>Cancer</strong> Center, Philadelphia, PA; The Children’s Hospital of<br />

Philadelphia, Philadelphia, PA; Children’s National Medical Center,<br />

Washington, DC; Massachusetts General Hospital, Boston, MA.<br />

P6-08-02 Do personal, familial or counseling factors influence the<br />

choice for prophylactic mastectomy and/or bilateral salpingooöphorectomy<br />

of female BRCA mutation carriers?<br />

Caanen BA, Gielen M, GomezGarcia EB, Kruitwagen RF, Keymeulen<br />

KB, Ter Haar-van Eck SA, Schenk K, Helderman-van den Enden<br />

A. Maastricht University Medical Centre, GROW School for<br />

Oncology and Developmental Biology, Maastricht Medical Centre,<br />

Maastricht, Netherlands; NUTRIM School for Nutrition, Toxicology<br />

and Metaboloism, University of Birmingham, Birmingham, United<br />

Kingdom; Maxima Medical Centre, Eindhoven, Netherlands.<br />

P6-08-03 Informational needs and psychosocial assessment of patients<br />

in their first year after metastatic breast cancer diagnosis<br />

Seah DS, Lin NU, Curley C, Winer E, Partridge A. Dana-Farber <strong>Cancer</strong><br />

Institution, Boston, MA.<br />

P6-08-04 The Impact of a <strong>Breast</strong> <strong>Cancer</strong> Diagnosis in Young Women on<br />

their Relationship with their Mothers<br />

Ali A, Fergus K, Wright F, Pritchard K, Kiss A, Warner E. Sunnybrook<br />

Health Sciences Centre, Odette <strong>Cancer</strong> Centre, University of Toronto,<br />

Toronto.<br />

P6-08-05 Changes in Cognitive Function in Older Women With<br />

<strong>Breast</strong> <strong>Cancer</strong> Treated with Standard Chemotherapy and<br />

Capecitabine within CALGB 49907<br />

Freedman RA, Pitcher B, Keating NL, Barry WT, Ballman KV, Kornblith<br />

A, Mandelblatt J, Kimmick GG, Hurria A, Winer EP, Hudis CA, Cohen<br />

HJ, Muss HB. Dana-Farber <strong>Cancer</strong> Institute, Boston, MA; Duke<br />

University Medical Center, Durham, NC; Harvard Medical School,<br />

Boston, MA; Mayo Clinic, Rochester, MN; Georgetown University,<br />

Washington, DC; City of Hope, Duarte, CA; Memorial Sloan-Kettering,<br />

New York, NY; University of North Carolina, Chapel Hill, NC.<br />

P6-08-06 Use of an NIH PROMIS® instrument to identify predictors<br />

of fatigue in breast cancer patients receiving adjuvant<br />

chemotherapy<br />

Cohen J, Junghaenel DU, Schneider S, Mahler L, Stone A, Broderick J.<br />

Stony Brook University, Stony Brook, NY.<br />

P6-08-07 Quality of life of women with breast cancer: A Middle East<br />

perspective<br />

Jassim GA, Whitford DL. Royal College of Surgeons in Ireland-Medical<br />

University of Bahrain, Busaiteen, Bahrain.<br />

P6-08-08 Three months of adjuvant hormone therapy does not increase<br />

fatigue or cognitive failure: results of a prospective early stage<br />

breast cancer trial<br />

Kennedy D, Lower EE. Oncology Hematology Consultants,<br />

Cincinnati, OH.<br />

P6-08-09 Overcoming <strong>Breast</strong> <strong>Cancer</strong>: The Importance of Connecting<br />

with Fellow Survivors<br />

Geoghegan C, Whitman B. Y-ME National <strong>Breast</strong> <strong>Cancer</strong><br />

Organization, Chicago, IL; Whitman Insight Strategies, New York, NY.<br />

P6-08-10 <strong>Cancer</strong> as self: a novel assessment of patient identity as it<br />

relates to a cancer diagnosis<br />

Horst KC, Fero KE, Haimovitz K, Dweck CS. Stanford University,<br />

Stanford, CA.<br />

Psychosocial, Quality of Life and Educational Aspects: Psychosocial,<br />

QOL and Educational Aspects-Other<br />

P6-09-01 Investigating the effectiveness of a psycho-educational<br />

behavioral intervention for cancer-related cognitive<br />

dysfunction in women with breast and gynecological cancer:<br />

Knowledge, self-efficacy, and behavioral change<br />

Bernstein LJ, Dissanayake D, Tirona KM, Nyhof-Young JM, Catton<br />

PA. Princess Margaret Hospital, Toronto, ON, Canada; University of<br />

Toronto, ON, Canada.<br />

www.aacrjournals.org<br />

73s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P6-09-02<br />

P6-09-03<br />

P6-09-04<br />

P6-09-05<br />

P6-09-06<br />

P6-09-07<br />

P6-09-08<br />

P6-09-09<br />

P6-09-10<br />

P6-09-11<br />

Pre-treatment cognitive function (CF) in women with locally<br />

advanced breast cancer (LABC) and in healthy controls<br />

Bernstein LJ, Seruga B, Pond G, Tirona KM, Dodd A, Tannock IF.<br />

Princess Margaret Hospital, Toronto, ON, Canada; University of<br />

Toronto; Institute of Oncology Ljubljana, Slovenia; McMaster<br />

University, Hamilton, ON, Canada.<br />

Fatigue after breast cancer may be related to conditions other<br />

than the cancer. The impact of comorbidity is essential<br />

Reidunsdatter RJ, Hjermstad M, Oldervoll L, Lundgren S. HiST/NTNU,<br />

Trondheim, Norway; HIST, Trondheim, Norway; NTNU, Trondheim,<br />

Norway; Oslo University Hospital, Oslo, Norway; Røros Rehabilitation,<br />

Røros, Norway; St. Olav University Hospital, Trondheim, Norway.<br />

The Association of Low Level Arm Volume Increases<br />

with Lymphedema Symptoms Following Treatment for<br />

<strong>Breast</strong> <strong>Cancer</strong><br />

Skolny MN, Miller CL, Shenouda M, Jammallo LS, O’Toole J,<br />

Niemierko A, Taghian AG. Massachusetts General Hospital,<br />

Boston, MA.<br />

Women’s perceptions of lymphedema risk management:<br />

Psychological factors do matter<br />

Sherman KA, Roussi P, Miller SM. Macquarie University, Sydney, NSW,<br />

Australia; Aristotle University, Thessaloniki, Greece; Fox Chase <strong>Cancer</strong><br />

Center, Philadelphia, PA.<br />

Family Members’ Burden in Patients with Metastatic and Early<br />

Stage <strong>Breast</strong> <strong>Cancer</strong><br />

Wan Y, Gao X, Mehta S, Zhang Z, Faria C, Schwartzberg L. Pharmerit<br />

North America; Eisai, Inc.; The West Clinic.<br />

Impact of comprehensive geriatric assessment on treatment<br />

decision and follow-up in older breast cancer patients<br />

Kenis C, Decoster L, Bode H, Bastin J, Lobelle J-P, Milisen K, van<br />

Puyvelde K, Paridaens R, Neven P, Fontaine C, de Grève J, Flamaing J,<br />

Wildiers H. University Hospitals Leuven; University Hospitals Brussels;<br />

Consult in Statistics, Beernem; Catholic University Leuven.<br />

COMPliance and Arthralgia in Clinical Therapy:<br />

The COMPACT trial, assessing the incidence of arthralgia,<br />

therapy costs and compliance within the first year of adjuvant<br />

anastrozole therapy<br />

Harbeck N, Blettner M, Bolten WW, Hindenburg H-J, Jackisch C,<br />

Klein P, König K, Kreienberg R, Rief W, Wallwiener D, Zaun S, Hadji P.<br />

University of Munich, Germany; University of Mainz, Germany; Klaus-<br />

Miehlke-Hospital for Rheumatology, Wiesbaden, Germany; Head<br />

of BNGO e.V., Germany; Hospital for Gynecology and Obstetrics,<br />

Offenbach, Germany; d.s.h. Statistical Services, Rohrbach, Germany;<br />

Gynecological Society Germany e.V., Germany; University Women’s<br />

Hospital, Ulm, Germany; Outpatient Clinic for Psychotherapy,<br />

Philipps-University, Marburg, Germany; University Women’s Hospital,<br />

Tübingen, Germany; AstraZeneca GmbH, Wedel, Germany; Women’s<br />

Hospital Phillips-University, Marburg, Germany.<br />

Perceptions of marginalization in those affected by advanced<br />

breast cancer<br />

Ahmed I, Harvey A, Amsellem M. <strong>Cancer</strong> Support Community,<br />

Washington, DC.<br />

Informational needs among women considering breast<br />

reconstruction post-mastectomy<br />

Ahmed I, Harvey A, Amsellem M. <strong>Cancer</strong> Support Community,<br />

Washington, DC.<br />

Examining patient treatment choices involving efficacy,<br />

toxicity, and cost tradeoffs in the metastatic breast cancer<br />

setting<br />

White CB, Smith ML, Abidoye O, Lalla D. Carol B. White & Associates;<br />

Research Advocacy Network; Genentech, Inc.<br />

Treatment: Novel Targets and Targeted Agents<br />

P6-10-01 A randomized phase 2 study of the antibody-drug conjugate<br />

CDX-011 in advanced GPNMB-overexpressing breast cancer:<br />

The EMERGE study<br />

Yardley DA, Weaver R, Melisko ME, Saleh MN, Arena FP, Forero A,<br />

Cigler T, Stopeck A, Citron D, Oliff I, Bechhold R, Loutfi R, Garcia A,<br />

Crowley E, Green J, Yellin MJ, Davis TA, Vahdat LT. Florida <strong>Cancer</strong><br />

Specialists, Tampa, FL; University of California, <strong>San</strong> Francisco Helen<br />

Diller Family Comprehensive <strong>Cancer</strong> Center, <strong>San</strong> Francisco, CA; Sarah<br />

Cannon Research Institute/Tennessee Oncology, PLLC, Nashville,<br />

TN; Georgia <strong>Cancer</strong> Specialists PC, <strong>San</strong>dy Springs, GA; Arena Onc<br />

Assoc PC, Lake Success, NY; Bium Inc, Birmingham, AL; Weill Cornell<br />

Medical College, New York, NY; Celldex Therapeutics, Inc., Needham,<br />

MA; Arizona <strong>Cancer</strong> Center at the University of Arizona, Tucson,<br />

AZ; <strong>Cancer</strong> Treatment Centers of America/Midwestern Regional<br />

Medical Center, Zion, IL; Orchard Healthcare Research Inc., Skokie,<br />

IL; Oncology Hematology Care, Cincinnati, OH; Henry Ford Health<br />

System, Detroit, MI; USC/Norris Comprehensive <strong>Cancer</strong> Center, Los<br />

Angeles, CA.<br />

P6-10-02 MLN8237 (alisertib), an investigational Aurora A<br />

Kinase inhibitor, in patients with breast cancer:<br />

Emerging phase 2 results<br />

Alvarez RH, DeMichele A, Mailliez A, Benaim E, Fingert H,<br />

Schusterbauer C, Zhang B, Melichar B. The University of Texas<br />

MD Anderson <strong>Cancer</strong> Center, Houston, TX; Abramson <strong>Cancer</strong><br />

Center, Philadelphia, PA; Centre Oscar Lambret, Lille, Cedex 59,<br />

France; Millennium Pharmaceuticals, Inc., Cambridge, MA; Fakultní<br />

nemocnice Olomouc - Onkologická klinika, Olomouc, Czech<br />

Republic.<br />

p6-10-03 The PKC inhibitor PKC412 antagonizes breast cancer cell<br />

growth and enhances tamoxifen sensitivity<br />

Shou J, Chew SA, Mitsiades N, Kumar V, Fu X, Chamness G, Osborne<br />

K, Schiff R. Lester & Sue Smith <strong>Breast</strong> Center, Baylor College of<br />

Medicine, Houston, TX; Baylor College of Medicine, Houston, TX; Dan<br />

L Duncan <strong>Cancer</strong> Center, Baylor College of Medicine, Houston, TX.<br />

P6-10-04 The Presence of Anaplastic Lymphoma Kinase Recapitulates<br />

Formation of <strong>Breast</strong> Tumor Emboli with Encircling<br />

Lymphovasculogenesis<br />

Liu H, Chu K, Ochoa AE, Ye Z, Zhang X, Jin J, Wright MC, Barsky SH,<br />

Cristofanilli M, Robertson FM. The University of Texas MD Anderson<br />

<strong>Cancer</strong> Center, Houston, TX; University of Nevada School of<br />

Medicine, Reno, NV; Fox Chase <strong>Cancer</strong> Center, Philadelphia, PA.<br />

P6-10-05 SU2C Phase 1b Trial of Dual PI3K/ mTOR Inhibitor BEZ235 with<br />

Letrozole in ER+/HER2- Metastatic <strong>Breast</strong> <strong>Cancer</strong> (MBC)<br />

Mayer IA, Abramson VG, Balko JM, Isakoff SJ, Forero A, Kuba MG,<br />

<strong>San</strong>ders ME, Li Y, Winer E, Arteaga CL. Vanderbilt-Ingram <strong>Cancer</strong><br />

Center, Nashville, TN; Massachussets General Hospital, Boston, MD;<br />

University of Alabama at Birmingham, AL; MD Anderson <strong>Cancer</strong><br />

Center, Houston, TX; Dana-Farber <strong>Cancer</strong> Institute, Boston, MD.<br />

P6-10-06 Rational combination therapy against triple-negative<br />

breast cancer<br />

Al-Ejeh F, Miranda M, Simpson PT, Chenevix-Trench G, Lakhani SR,<br />

Khanna KK. Queensland Institute of Medical Research, Brisbane,<br />

QLD, Australia; The University of Queensland, Brisbane,<br />

QLD, Australia.<br />

P6-10-07 Phase I study of BYL719, an alpha-specific PI3K inhibitor,<br />

in patients with PIK3CA mutant advanced solid tumors:<br />

preliminary efficacy and safety in patients with PIK3CA mutant<br />

ER-positive (ER+) metastatic breast cancer (MBC)<br />

Juric D, Argiles G, Burris HA, Gonzalez-Angulo AM, Saura C, Quadt<br />

C, Douglas M, Demanse D, De Buck S, Baselga J. M.assachussetts<br />

General Hospital <strong>Cancer</strong> Center, Boston, MA; Vall d’Hebron University<br />

Hospital, Barcelona, Spain; Sarah Cannon Research Institute,<br />

Nashville, TN; M.D. Anderson <strong>Cancer</strong> Center, Houston, TX; Novartis<br />

Pharma Corporation, East Hanover, NJ; Novartis Pharma AG,<br />

Basel, Switzerland.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012 74s <strong>Cancer</strong> Research


December 4–8, 2012<br />

Program Schedule<br />

P4-06-07 Isoform specific antibodies against the fibroblast growth<br />

factor receptor 2 have predictive and therapeutic implications<br />

in the treatment of human breast cancers<br />

Ettyreddy AR, Somarelli J, Bitting R, Armstrong A, Garcia Blanco MA.<br />

Duke University, Durham, NC.<br />

Treatment: New Drugs and Treatment Strategies<br />

P6-11-01 Intermittent High Dose Proton Pump Inhibitor Improves<br />

Progression Free Survival as Compared to Standard<br />

Chemotherapy in the First Line Treatment of Patients with<br />

Metastatic <strong>Breast</strong> <strong>Cancer</strong><br />

Hu X, Wang B, Sun S, Chiesi A, Wang J, Zhang J, Fais S. Fudan<br />

University Shanghai <strong>Cancer</strong> Center, Shanghai, China; National<br />

Institute of Health, Roma, Italy.<br />

P6-11-02 Inhibition of mTOR and Fatty Acid Synthase (FASN) overcome<br />

acquired resistance to Trastuzumab, Lapatinib and both in<br />

HER2+ breast cancer<br />

Puig T, Blancafort A, Giro A, Viqueira A, Viñas G, Massaguer A,<br />

Carrion-Salip D, Bolos MV, Urruticoechea A, Oliveras G. University<br />

of Girona and Girona Biomedical Research Institute (IDIBGi), Girona,<br />

Spain; Catalan Institute of Oncology (ICO) and Girona Biomedical<br />

Research Institute (IDIBGi), Girona, Spain; Pfizer, Madrid, Spain;<br />

University of Girona, Spain; Catalan Institute of Oncology (ICO),<br />

Hospitalet de Llobregat, Barcelona, Spain.<br />

P6-11-03 Site-Specific, Concomitant Delivery of Rapamycin and<br />

Paclitaxel in <strong>Breast</strong> <strong>Cancer</strong>: Consequent Synergistic<br />

Efficacy Enhancement<br />

Blanco E, <strong>San</strong>gai T, Hsiao A, Ruiz-Esparza GU, Ferrari M, Meric-<br />

Bernstam F. The Methodist Hospital Research Institute, Houston, TX;<br />

The University of Texas MD Anderson <strong>Cancer</strong> Center, Houston, TX.<br />

P6-11-04 Targeting the tumor microenvironment: tetrathiomolybdate<br />

decreases circulating endothelial progenitor cells in women<br />

with breast cancer at high risk of relapse<br />

Jain S, Kornhauser N, Lam C, Ward MM, Chuang E, Cigler T, Moore<br />

A, Donovan D, Cobham MV, Schneider S, Hurtado Rua SM, Lane ME,<br />

Mittal V, Vahdat LT. Weill Cornell Medical College.<br />

P6-11-05 Novel sorafenib-derivatives induce apoptosis in breast cancer<br />

cells through STAT3 inhibition<br />

Liu C-Y, Tseng L-M, Chang K-C, Chu P-Y, Su J-C, Shiau C-W, Chen K-F.<br />

Taipei Veterans General Hospital, Taipei, Taiwan; National Yang-Ming<br />

University, Taipei, Taiwan; St. Martin De Porres Hospital, Chia-Yi,<br />

Taiwan; National Taiwan University Hospital, Taipei, Taiwan.<br />

P6-11-06 A phase Ib study of LCL161, an oral inhibitor of apoptosis (IAP)<br />

antagonist, in combination with weekly paclitaxel in patients<br />

with advanced solid tumors<br />

Dienstmann R, Vidal L, Dees EC, Chia S, Mayer EL, Porter D, Baney<br />

T, Dhuria S, Sen SK, Firestone B, Papoutsakis D, Cameron S, Infante<br />

JR. Vall d’Hebron University Hospital, Barcelona, Spain; Hospital<br />

Clinic i Provincial, Barcelona, Spain; University of North Carolina<br />

Lineberger Comprehensive <strong>Cancer</strong> Center, Chapel Hill, NC;<br />

British Columbia <strong>Cancer</strong> Agency, University of British Columbia,<br />

Vancouver, BC, Canada; Dana-Farber <strong>Cancer</strong> Institute, Boston, MA;<br />

Novartis Pharmaceuticals Corporation, Cambridge, MA; Novartis<br />

Pharmaceuticals Corporation, Florham Park, NJ; Sarah Canon<br />

Research Institute, Nashville, TN.<br />

P6-11-07 The <strong>Cancer</strong> Stem Cell-Targeting Wnt Inhibitor VS-507 Reduces<br />

<strong>Breast</strong> <strong>Cancer</strong> Growth and Metastasis<br />

Ring JE, Kolev VN, Padval MV, Keegan M, Vidal CM, Neill AA, Shapiro<br />

IM, Pachter JA, Xu Q. Verastem, Inc., Cambridge, MA.<br />

P6-11-08 A multicenter, open-label Ph IB/II study of BEZ235, an oral<br />

dual PI3K/mTOR inhibitor, in combination with paclitaxel in<br />

patients with HER2-negative, locally advanced or metastatic<br />

breast cancer<br />

Campone M, Fumoleau P, Gil-Martin M, Isambert N, Beck JT, Becerra<br />

C, Shtivelband M, Duval V, di Tomaso E, Roussou P, Urban P,<br />

Urruticoechea A. Centre René Gauducheau, Nantes, France; Centre<br />

Georges François Leclerc, Dijon, France; Institut Català d’Oncologia,<br />

Barcelona, Spain; Highlands Oncology Group, Fayetteville, AR; Baylor<br />

University Medical Center, Dallas, TX; Ironwood <strong>Cancer</strong> and Research<br />

Centers, Chandler, AZ; Novartis Pharma AG, Basel, Switzerland;<br />

Novartis Institutes for BioMedical Research, Inc., Cambridge, MA.<br />

P6-11-09 FAK Inhibitor VS-4718 Attenuates <strong>Breast</strong> <strong>Cancer</strong> Stem Cell<br />

Function In Vitro and In Vivo<br />

Kolev VN, Vidal CM, Shapiro IM, Pavdal M, Keegan M, Xu Q, Pachter<br />

JA. Verastem, Cambridge, MA.<br />

P6-11-10 IBL2001: Phase I/II study of a novel dose-dense schedule of<br />

oral indibulin for the treatment of metastastic breast cancer<br />

Traina TA, Hudis C, Seidman A, Gajria D, Gonzalez J, Anthony SP,<br />

Smith DA, Chandler JC, Jac J, Youssoufian H, Korth CC, Barrett JA,<br />

Sun L, Norton L. Memorial Sloan-Kettering <strong>Cancer</strong> Center, New<br />

York, NY; Evergreen Hematology and Oncology, Spokane, WA;<br />

Compass Oncology, Vancouver, WA; The West Clinic, Memphis, TN;<br />

US Oncology Research, Woodlands, TX; ZIOPHARM Oncology, Inc.,<br />

Boston, MA; Harmon Hill Consulting, New York, NY.<br />

P6-11-11 Multistage Delivery of Paclitaxel: Increased Drug Stability<br />

and Sustained Release Results in Enhanced Efficacy in<br />

<strong>Breast</strong> <strong>Cancer</strong><br />

Blanco E, <strong>San</strong>gai T, Hsiao A, Ferrati S, Bai L, Liu X, Meric-Bernstam F,<br />

Ferrari M. The Methodist Hospital Research Institute, Houston, TX;<br />

The University of Texas MD Anderson <strong>Cancer</strong> Center, Houston, TX.<br />

P6-11-12 Nanoparticle-enhanced chemotherapeutics delivery in<br />

drug-resistant triple-negative breast cancer<br />

van de Ven AL, Landis MD, Paskett LA, Meyn A, Frieboes HB, Chang<br />

JC, Ferrari M. The Methodist Hospital Research Institute, Houston, TX;<br />

University of Louisville, KY.<br />

P6-11-13 In vitro antineoplastic evaluation of rationally designed<br />

naphthoquinone-derived drugs in triple-negative<br />

breast cancer cell line<br />

Madeira KP, Daltoé RD, Herlinger AL, Guimarães IS, Allochio Filho JF,<br />

Teixeira SF, Valadão IC, Greco S, Rangel LBA. Federal University of<br />

Espirito <strong>San</strong>to, Brazil.<br />

P6-11-14 Post-hoc safety and tolerability assessment in patients<br />

receiving palliative radiation during treatment with eribulin<br />

mesylate for metastatic breast cancer<br />

Yardley DA, Vahdat L, Rege J, Cortés J, Wanders J, Twelves C. Sarah<br />

Cannon Research Institute, Nashville, TN; Weill Cornell Medical<br />

College, New York, NY; Eisai Inc, Woodcliff Lake, NJ; Vall d’Hebron<br />

University Hospital, Barcelona, Spain; European Knowledge Center,<br />

Eisai Ltd., Hatfield, Hertfordshire, United Kingdom; St James<br />

University Hospital, Leeds, United Kingdom.<br />

Treatment: Adjuvant Therapy - Other<br />

P6-12-01 Adjuvant treatment in breast cancer patients aged 70<br />

years or older during three years. A systematic review of<br />

patients charts<br />

Nätterdal T, Klint L, Holmberg E, Gunnarsdottír KA, Linderholm BK.<br />

Institution of Medical Sciences, Sahlgrenska University Hospital,<br />

Gothenburg, Sweden; Sahlgrenska University Hospital, Gothenburg,<br />

Sweden; Karolinska Institute Science Park, Stockholm, Sweden.<br />

P6-12-02 Use of cytochrome P450 interacting medications in the setting<br />

of adjuvant therapy for breast cancer<br />

Njiaju UO, Kolesar JM, Johnston SA, Eickhoff JC, Osterby KR, Poggi<br />

LE, Tevaarwerk AJ, Millholland RJ, Oliver KA, Heideman JL, Wisinski<br />

KB. University of Wisconsin-Madison, WI; University of Wisconsin-<br />

Madison; University of Wisconsin Hospitals and Clinics.<br />

www.aacrjournals.org<br />

75s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P6-12-03 Effects of high dose of bisphosphonate therapy on bone<br />

microarchitecture of the peripheral skeleton in women with<br />

early stage breast cancer<br />

Shao T, Shane ES, McMahon D, Crew KD, Kalinsky K, Maurer M,<br />

Brown M, Gralow JR, Hershman DL. Beth Israel Medical Center,<br />

Continuum <strong>Cancer</strong> Centers of New York, New York, NY; Columbia<br />

University Medical Center, New York, NY; University of Washington/<br />

Seattle <strong>Cancer</strong> Care Alliance, Seattle, WA.<br />

Treatment: Therapy for Advanced Disease - Other<br />

P6-13-01 Lyso-thermosensitive liposomal doxorubicin + local<br />

hyperthermia for radiation-pretreated chest wall recurrence<br />

Formenti S, Rugo H, Myerson R, Diederich C, Straube W, O’Conner B,<br />

Matzkowitz AJ, Goodman RL, Muggia F. New York University <strong>Cancer</strong><br />

Institute, New York, NY; UC <strong>San</strong> Francisco, <strong>San</strong> Francisco, CA; Siteman<br />

<strong>Cancer</strong> Center, Saint Louis, MO; Rhode Island Hospital, Providence,<br />

RI; New Hope <strong>Cancer</strong> Center, Hudson, FL; Saint Barnabas Medical<br />

Center, Livingston, NJ.<br />

P6-13-02 Overcoming therapy resistance of metastatic breast cancer by<br />

enhanced tumor delivery of polymeric doxorubicin<br />

Shen H, Xu R, Mai J, Huang Y, Ferrari M. The Methodist Hospital<br />

Research Institute, Houston, TX.<br />

P6-13-03 Symptomatic bone marrow involvement (BMinv) in breast<br />

cancer (BC): Clinical presentation, treatment and prognosis<br />

according to BC subtype and Zoledronic acid (ZA) use.<br />

A single institution review<br />

Torrejon-Castro D, Zamora E, <strong>San</strong>chez-Olle G, Balmaña J, Gomez<br />

P, Saura C, Perez-Garcia J, Muñoz-Cousuelo E, Vidal M, Ortega V,<br />

Oliveira M, De Mattos L, Cortes J, Bellet M. Vall d´Hebron University<br />

Hospital, Barcelona, Spain.<br />

Treatment: Treatment - Other<br />

P6-14-01 Estrogen/progestogen use after <strong>Breast</strong> <strong>Cancer</strong> – a long-term<br />

follow-up of the Stockholm randomized trial<br />

Fahlén MC, Fornander T, Johansson H, Rutqvist L-E, Wilking N, von<br />

Schoultz E. Capio St Görans Hospital, Stockholm, Sweden; Karolinska<br />

Institutet, Stockholm, Sweden.<br />

P6-14-02 Withdrawn<br />

P6-14-03 Statin and aspirin use is not associated with a reduced risk of<br />

VTE’s in breast cancer patients<br />

Shai A, Rennert HS, Ballan Haj M, Lavie O, Steiner M, Rennert G. Lin<br />

Medical Center, Haifa, Israel; Carmel Lady Davis Medical Center,<br />

Haifa, Israel.<br />

P6-14-04 Rosehip extracts prevent triple negative breast cancer<br />

cell proliferation by regulating the phosphorylation of<br />

p70S6 Kinase<br />

Coburn TL, Cagle PD, Shofoluwe AI, Martin PM. North Carolina<br />

Agricultural and Technical State University, Greensboro, NC.<br />

P6-14-05 Impact of breast cancer CME: Physician practice pattern,<br />

knowledge, and competence assessments<br />

Haas M, Heintz A, Stacy T. Educational Concepts Group, LLC,<br />

Atlanta, GA.<br />

8:30 am–9:00 am<br />

PLENARY LECTURE 4<br />

Exhibit Hall D<br />

1st International Consensus Guidelines Conference for Advanced<br />

<strong>Breast</strong> <strong>Cancer</strong> – ABC1<br />

Fatima Cardoso, MD<br />

Champalimaud <strong>Cancer</strong> Centre<br />

Lisbon, PORTUGAL<br />

9:00 am–11:00 am<br />

THE YEAR IN REVIEW<br />

Exhibit Hall D<br />

Moderator: C. Kent Osborne, MD<br />

Baylor College of Medicine<br />

Houston, TX<br />

Advances in basic breast cancer research<br />

Jeffrey M. Rosen, PhD<br />

Baylor College of Medicine<br />

Houston, TX<br />

Discoveries in translational breast cancer research<br />

in 2012<br />

Douglas Yee, MD<br />

University of Minnesota<br />

Minneapolis, MN<br />

Adjuvant and neoadjuvant<br />

Kathy D. Miller, MD<br />

Indiana University School of Medicine<br />

Indianapolis, IN<br />

New therapies in 2012 for advanced disease –<br />

where do they all fit in<br />

Stephen Johnston, MA, PhD, FRCP<br />

Royal Marsden Hospital<br />

London, UNITED KINGDOM<br />

11:00 am<br />

ADJOURNMENT<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012 76s <strong>Cancer</strong> Research


December 4–8, 2012<br />

Program Schedule<br />

www.aacrjournals.org 77s <strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

ES1-1<br />

Molecular Profiling for In Situ Carcinoma of the <strong>Breast</strong><br />

Solin LJ. Albert Einstein Medical Center, Philadelphia, PA<br />

The management of in situ carcinoma of the breast represents a clinical<br />

dilemma. Such a non-invasive lesion places the patient at risk for<br />

subsequent development of an invasive lesion, and various treatments<br />

have been shown to reduce this risk. However, most patients are<br />

asymptomatic at initial presentation, and standard treatments are<br />

associated with the risk of side effects. It can be difficult for the<br />

individual patient to find the most appropriate balance between<br />

the risks and benefits of adding treatment. Although many patients<br />

elect for all treatments to minimize risk of progression regardless<br />

of potential side effects, other patients elect against treatment, often<br />

because of the potential risk of associated side effects.<br />

Standard approaches to managing the patient with in situ disease have<br />

employed various clinical and pathologic characteristics to assess<br />

and stratify risk. Models for determining risk have used a number<br />

of clinical and pathologic characteristics, including patient age,<br />

lesion size, and grade. Ductal carcinoma in situ (DCIS) and lobular<br />

carcinoma in situ (LCIS) have different risk profiles. For example,<br />

LCIS is well known to have subsequent risk that is more frequently<br />

bilateral than DCIS.<br />

Prospective randomized clinical trials for DCIS have demonstrated<br />

a substantial reduction in the risk of recurrence associated with<br />

adding radiation treatment and tamoxifen after lumpectomy. Five<br />

randomized DCIS trials have shown that adding radiation treatment<br />

after lumpectomy lowers the risk of local recurrence and invasive<br />

local recurrence in the ipsilateral breast by approximately 50%. Two<br />

randomized DCIS trials have demonstrated that adding tamoxifen<br />

reduces the risk of all breast cancer events (combined ipsilateral plus<br />

contralateral) by approximately 30-40%. No survival benefit has been<br />

shown for adding either radiation or tamoxifen. For LCIS, hormonal<br />

treatment has been shown to reduce subsequent risk.<br />

More recent efforts have focused on the value of molecular markers<br />

to individualize and tailor risk profiling for the individual patient.<br />

Using such molecular risk profiling, adding treatment can be selected<br />

for higher risk patients, and omitting treatment can be chosen for<br />

lower risk patients. To be clinically useful, molecular profiling<br />

should demonstrate value beyond that seen using standard clinical<br />

and pathologic factors. While current efforts have focused on using<br />

molecular profiling as a prognostic factor, future efforts will evaluate<br />

molecular profiling as a predictive factor.<br />

ES1-2<br />

The Practical Use of Molecular Profiling: The View of a Medical<br />

Oncologist<br />

Wolff AC. Johns Hopkins Kimmel Comprehensive <strong>Cancer</strong> Center<br />

Adjuvant therapy changed the natural of early stage breast cancer.<br />

Starting in the 1970s, randomized trials have shown a survival<br />

benefit from adjuvant chemotherapy in operable, node-pos disease.<br />

Since 1985, the Oxford Overview led the rapid adoption of adjuvant<br />

chemotherapy (for ER-neg disease) and tamoxifen (for ER-pos<br />

disease), and this trend accelerated after the 1988 NCI Clinical Alert<br />

on adjuvant chemotherapy in node-neg disease. However, this was<br />

not without controversy. In 1990, an NIH Consensus Development<br />

Conference concluded that “… some degree of improvement (from<br />

adjuvant therapy) may be so small that they are outweighed by the<br />

disadvantages of therapy.” Soon, adjuvant chemotherapy also became<br />

a standard in ER-pos, node-neg disease.<br />

Starting in the mid 1990s, data began to suggest that not all patients<br />

with ER-pos disease benefitted from chemotherapy. Decision<br />

algorithms based on routine clinicopathologic factors (eg, TNM,<br />

grade, ER, and HER2) proved quite useful for decision-making for<br />

the average patient. But by the early 2000s, new molecular tools to<br />

stratify recurrence risk (prognosis) and likelihood of benefit from<br />

chemotherapy (prediction) became available in clinical practice to<br />

further individualize clinical decisions. Prospective-retrospective<br />

studies tested the prognostic/predictive utility of Oncotype DX.<br />

Soon, it became clear that it and routine clinicopathologic parameters<br />

offered independent prognostic utility in ER-pos disease. The TailoRx<br />

(node-neg) and RxPONDER (node-pos) trials are now prospectively<br />

testing Oncotype DX to guide the decision to add chemotherapy to<br />

endocrine therapy in patients with a lower/intermediate recurrence<br />

score. Mammaprint became the first FDA-cleared IVDMIA assay, and<br />

the MINDACT study directly addresses this predictive utility question<br />

by having randomized patients with a discordant result (compared to<br />

Adjuvant! Online) also receive chemotherapy or not.<br />

Patients with HER2-pos disease are offered HER2-directed therapy<br />

plus chemotherapy. Consequently, a clinical decision gap exists in<br />

triple-neg disease (TNBC), which affects 50,000 women just in the<br />

US each year. Decision algorithms for them are exclusively based on<br />

clinicopathological factors like TNM, grade, and HER2, as molecular<br />

assays like Oncotype DX and MammaPrint have no role in this setting.<br />

New molecular assays based on DNA methylation, immune markers,<br />

or other gene expression signatures are under development. In the<br />

meantime, adjuvant chemotherapy is routinely offered to all TNBC<br />

patients with at least T1b tumors, despite the fact that most patients<br />

with ER-neg, node-neg disease remain disease-free long-term when<br />

treated with just locoregional therapy.<br />

Various studies examined clinical decisions based on standard<br />

clinicopathologic parameters alone or with knowledge of molecular<br />

assay results. However, data beyond costs of care are needed to<br />

assess their independent and complementary role to improve the<br />

most important clinical outcome to patients (i.e., survival). Until then,<br />

access to accurate routine clinicopathologic markers for all patients<br />

worldwide remains critical to ensure the most optimal outcome to all<br />

patients in high and low income countries.<br />

ES2-2<br />

Triple Negative <strong>Breast</strong> <strong>Cancer</strong>: Subtypes, Molecular Targets, and<br />

Therapeutic Approaches<br />

Pietenpol JA. Vanderbilt-Ingram <strong>Cancer</strong> Center; Vanderbilt<br />

University School of Medicine, Nashville, TN<br />

The term “triple negative breast cancer” (TNBC) is used to classify<br />

10%-20% of all breast cancers that lack estrogen receptor (ER)<br />

and progesterone receptor expression as well as amplification of<br />

the human epidermal growth factor receptor 2 (HER2) 1 . Disease<br />

heterogeneity and the absence of well-defined molecular targets<br />

have made treatment of TNBC challenging. To make significant<br />

clinical advances for TNBC patients, integrative and comprehensive<br />

genomic and molecular analyses of TNBC are required to understand<br />

the complexity of the disease as well as to allow identification<br />

of homogeneous subsets and ‘driver pathways’ that can then be<br />

therapeutically targeted. In response to this need, we compiled<br />

an extensive number of TNBC gene expression (GE) profiles and<br />

initiated molecular subtyping of the disease 2 . We identified two basallike<br />

TNBC subtypes with cell cycle and DDR GE signatures (BL1<br />

and BL2); two mesenchymal subtypes with high expression of genes<br />

involved in differentiation and growth factor pathways (M and MSL);<br />

an immunomodulatory (IM) type; and a luminal subtype driven by<br />

androgen signaling (LAR). Differential GE was used to designate 25<br />

TNBC cell line models representative of these subtypes. Predicted<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

78s<br />

<strong>Cancer</strong> Research


December 4-8, 2012<br />

<strong>Abstract</strong>s: Invited Speakers<br />

‘driver’ signaling pathways were pharmacologically targeted in these<br />

preclinical models as proof of concept that analysis of distinct GE<br />

signatures can inform therapy selection. Representative BL1 and BL2<br />

subtype cell lines preferentially respond to cisplatin. Mesenchymal,<br />

basal, and luminal subtype lines with aberrations in PI3K signaling<br />

have the greatest sensitivity, in general, to phosphatidylinositol<br />

3-kinase (PI3K) inhibitors. The LAR subtype cell lines express<br />

AR and are uniquely sensitive to bicalutamide (AR antagonist).<br />

We have also developed “TNBCtype,” a web-based subtyping tool<br />

for candidate TNBC tumor samples using our GE metadata and<br />

classification methods 3 . The approaches used and data generated<br />

have value for biomarker selection, drug discovery, and clinical trial<br />

design that will enable alignment of TNBC patients to appropriate<br />

targeted therapies.<br />

1. Dent, R., Trudeau, M., Pritchard, K.I., Hanna, W.M., Kahn, H.K.,<br />

Sawka, C.A., Lickley, L.A., Rawlinson, E., Sun, P., and Narod, S.A.<br />

(2007). Triple-negative breast cancer: clinical features and patterns<br />

of recurrence. Clin <strong>Cancer</strong> Res 13, 4429-4434.<br />

2. Lehmann, B.D., Bauer, J.A., Chen, X., <strong>San</strong>ders, M.E., Chakravarthy,<br />

A.B., Shyr, Y., and Pietenpol, J.A. (2011). Identification of human<br />

triple-negative breast cancer subtypes and preclinical models for<br />

selection of targeted therapies. J Clin Invest. 121: 2750-67.<br />

3. Chen, X., Li, J., Gray, W.H., Lehmann, B.D., Bauer, J.A., Shyr, Y.,<br />

and Pietenpol, J.A. (2012). TNBCtype: A Subtyping Tool for Triple-<br />

Negative <strong>Breast</strong> <strong>Cancer</strong>. <strong>Cancer</strong> informatics 11, 147-156.<br />

ES2-3<br />

The Clonal and Mutational Composition of Triple Negative<br />

<strong>Breast</strong> <strong>Cancer</strong>s<br />

Aparicio S. University of British Columbia<br />

The notion that cancers are mosaics, composed of clones of cells<br />

has been fundamental to cancer biology for several decades.<br />

Clonal evolution may underpin clinical features of cancer, such as<br />

emergence of treatment resistance, the heterogeneous responses of<br />

metastatic disease within individual patients and divergence between<br />

primary and metastatic disease. Spatial and temporal variation<br />

in clonal composition poses significant challenges for diagnostic<br />

sampling and disease monitoring. We have recently developed next<br />

generation sequencing approaches to these questions and applied<br />

them to the understanding of mutational and clonal composition<br />

of primary triple negative breast cancers (TNBC). The notion that<br />

triple negative breast cancers form a distinct disease entity is already<br />

strongly challenged by expression analysis and small scale mutational<br />

analysis. Our results show that both the mutational composition and<br />

clonal structure of primary triple negative breast cancers is in fact a<br />

complete spectrum, exploding the notion of TNBC (currently defined),<br />

as a distinct disease. Clonal analysis suggests a means by which the<br />

genetic complexity might be reduced by following patient evolution<br />

over time and space. I will discuss the specific implications of the<br />

mutational and transcriptome landscapes of TNBC in relation to<br />

possible disease biologies.<br />

ES3-1<br />

Early Phase Trials – What Are They For?<br />

Hilsenbeck SG. Baylor College of Medicine, Houston, TX<br />

Early in drug development, there are lots of things we need to learn<br />

about new agents or new combinations of agents in order to make<br />

‘good’ decisions about whether and how to proceed. The landscape<br />

of clinical trials is changing, with greater focus on targeted therapies<br />

and biomarker defined patient populations. Trial design, as we will<br />

hear in this session, must adapt to these new challenges. However,<br />

fundamentals still apply. We still need to demonstrate that drugs are<br />

safe and effective. As an introduction to the session, we will review<br />

the basic steps in drug development, especially early development,<br />

and the basic questions that must be answered at each step, in order<br />

to develop evidence for safety and efficacy.<br />

ES3-2<br />

Clinical Trial Designs for New Therapies: What Agents Should<br />

We Study?<br />

DeMichele A. University of Pennsylvania Perelman School of<br />

Medicine, Philadelphia, PA<br />

Biologically-targeted, novel agents hold great promise in breast<br />

cancer, but require adjustments to traditional study design, conduct<br />

and patient selection to be tested safely, effectively and economically.<br />

Key questions to be address in this talk include: Is the drug safe?<br />

Is it hitting the target? Can we identify the “right” patients to test?<br />

In what setting do we test it? Toxicities of these agents move wellbeyond<br />

traditional myelosuppression and fatigue, encompassing<br />

almost every organ system and requiring changes in monitoring<br />

and management to assure safety. Dosing and assessment strategies<br />

must be aimed at optimally modulating target and assessing whether<br />

hitting the target has the desired effect on the natural history of the<br />

disease. The concurrent development of predictive biomarkers is<br />

necessary to select appropriate study subjects, which may be limited<br />

to a relatively small subset of patients with the disease. Finally,<br />

selecting the best therapeutic setting (treatment naïve, refractory,<br />

acquired resistance) can greatly impact the performance of the agent<br />

and requires knowledge of cancer biological evolution through the<br />

metastatic process.<br />

ES3-3<br />

Clinical Trial Designs for New Therapies: What Endpoints<br />

Should We Use?<br />

Hudis CA. Memorial Sloan-Kettering <strong>Cancer</strong> Center, New York, NY;<br />

Weill Cornell Medical College, New York, NY<br />

The selection of endpoints is critical to the interpretation of clinical<br />

trials results. In early phase clinical trials investigators seek evidence<br />

of activity, then predictors of clinical benefit, and in most randomized<br />

trials, accepted surrogates of meaningful benefit. At one extreme we<br />

may seek improvements in overall survival while in another we may<br />

be satisfied simply with evidence of tumor stabilization or changes in<br />

circulating biomarkers. In between these extremes are intermediate<br />

and composite endpoints including “partial responses”, “clinical<br />

benefit rate”, and those that are time-dependent endpoints such as<br />

disease-free or progression free survival. Many of the discussions and<br />

disagreements we hear in the context of drug development, benefit,<br />

and approval can be linked to a fundamental lack of agreement on the<br />

value and meaning of many of these endpoints. Activity of a given<br />

novel agent may, or may not, persuade some investigators, clinicians,<br />

and patients, that it is a worthwhile addition to our treatment options.<br />

On the other hand, a small but statistically significant improvement<br />

in overall survival in advanced cancer may, or may not, convince this<br />

same audience that the tested treatment is worthy of approval and use.<br />

Again, the intermediate endpoints may be unreliable or only weak<br />

predictors of more profound benefits. We will review and explore<br />

these issues considering not only the endpoints themselves but also<br />

their meaning in specific clinical settings and the impact of endpoint<br />

selection on further drug development.<br />

www.aacrjournals.org 79s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

ES3-4<br />

Clinical Trial Designs for New Therapies: When Should We<br />

Randomize?<br />

Hunsberger S. National <strong>Cancer</strong> Institute<br />

There is a tension in clinical trial study design between the desire to<br />

keep the number of patients in a study small (so that as few patients<br />

as possible are exposed to potentially harmful agents, resources are<br />

minimized, and an answer is obtained as early as possible) and how<br />

much certainty is needed in the conclusion of the study. Different<br />

phases of drug development require different levels of certainty.<br />

Single arm studies require fewer patients than randomized studies<br />

but an observed improvement in the study defined endpoint in<br />

a single arm study may be observed for reasons unrelated to the<br />

experimental treatment. In randomized studies factors (known and<br />

unknown) that influence the endpoint of interest should be balanced<br />

between groups therefore any improvement in the endpoint of interest<br />

can be attributed to the experimental agent. In this talk I discuss<br />

circumstances that would be appropriate to perform single arms<br />

studies and circumstances that require randomized studies.<br />

ES4-1<br />

Navigating the Obstacles and Risks of Survivorship: <strong>Breast</strong><br />

<strong>Cancer</strong> Sexuality: Issues and Answers<br />

Krychman M. The Southern California Center for Sexual Health and<br />

Survivorship, Newport Beach, CA; University of California Irvine,<br />

Irvine, CA; University of Southern California, Los Angeles, CA<br />

The sexual consequences of breast cancer and its treatments are well<br />

known. Sexual difficulties include alteration in hormonal levels,<br />

changes in sexual organs and anatomy, vaginal and vulvar dryness,<br />

changes in libido, increased latency to orgasm and decreased intensity<br />

of orgasmic response. Changes in body image, sexual self-esteem and<br />

relationship dynamic tension may present as survivorship challenges<br />

for the breast cancer patient and her partner. Severe vulvo vaginal<br />

and clitoral atrophy as a result of chemotherapy and/or adjuvant<br />

hormone therapy, and loss of libido secondary to dyspareunia are<br />

widespread. Cytostatic medications that are prescribed for long-term<br />

use (aromatase inhibitors) can impact bone health, sexual functioning/<br />

satisfaction and overall quality of life. Many women on aromatase<br />

inhibitors have severe complaints of vulvar and vaginal atrophy.<br />

Sexual dysfunction is common for breast cancer population and<br />

limited acceptable treatments are available. Minimally absorbed local<br />

vaginal estrogen use in this population remains controversial and a<br />

hotly debated controversial topic. More data should be forthcoming.<br />

There are multiple new innovative treatments in the research pipeline<br />

for female sexual dysfunction. DHEA intravaginal suppositories,<br />

Flibanserin and Bremelanotide (PT 141) remain promising although<br />

they have not been adequately studied in the breast cancer population.<br />

Ospemifene a novel estrogen agonist/ antagonist has demonstrated<br />

a strong estrogenic effect on the vaginal epithelium during a three<br />

month trial period and did not stimulate the endometrium; however,<br />

the medication did mildly aggravate hot flashes (this did not lead<br />

to drug discontinuation). Although not studied in breast cancer<br />

patients, pivotal Phase 3 data confirmed effectiveness of ospemifene<br />

as effective treatment for vulvovaginal atrophy. In a recent article<br />

in Menopause, Ospemifene and 4-hydroxyospemifene effectively<br />

prevented and treat breast cancer in Mtag.TG Transgenic Mouse.<br />

Testosterone use remains off label and is not FDA approved, yet it<br />

is widely used in the non cancer population. New emerging safety<br />

data will be presented. Vaginal moisturizers, lubricants and dilators in<br />

association with genito pelvic floor therapy and intravaginal valium<br />

suppositories can be considered front line treatment for atrophy,<br />

dyspareunia and hypertonic vaginismus. New innovative data on non<br />

hormonal management of hot flashes is and important initial step in<br />

the overall management of the breast cancer patient who is suffering<br />

from sexual dysfunction.<br />

A multi model treatment paradigm will be presented which will<br />

include pharmacologic, nonpharmacologic, and psychosocial<br />

interventions.<br />

ES4-2<br />

Cognitive Changes and <strong>Breast</strong> <strong>Cancer</strong> Treatments<br />

Ganz PA. University of California, Los Angeles<br />

Cognitive difficulties after cancer treatment are among the most<br />

feared outcomes voiced by patients who are approaching the start of<br />

chemotherapy. This phenomenon, sometimes referred to as “chemo<br />

brain” or “chemo fog,” is quite common during the acute course of<br />

chemotherapy treatment. This may result from direct antieneoplastic<br />

drug toxicity, use of premedications and anxiolytics to prevent nausea<br />

and vomiting, as well as the expected anxiety, depression, and sleep<br />

deprivation that accompanies a new diagnosis of cancer and initiation<br />

of treatment. For most patients, this fog clears after the completion of<br />

acute treatments, and as with many physical symptoms, resolves in<br />

the following months. However, 15-25% of post-treatment patients,<br />

cognitive complaints persist long after treatment ends. Reports of<br />

difficulty concentrating, remembering recent events, multi-tasking,<br />

and paying attention are frequent in such patients. For those used to<br />

carrying on demanding executive function activities prior to cancer<br />

treatment, this can lead to substantial disruption in work and home<br />

life. Most of the studies conducted to date with early stage breast<br />

cancer have not accounted for the impact of adjuvant endocrine<br />

therapy alone or added to adjuvant chemotherapy. This presentation<br />

will review emerging data regarding self-reported cognitive<br />

complaints, neuropsychological testing, and brain imaging as they<br />

related to adjuvant therapies for breast cancer. In addition, some of<br />

the developing information about potential mechanisms associated<br />

with this late effect will be discussed.<br />

ES4-3<br />

Risk of Developing Second Primary <strong>Breast</strong> <strong>Cancer</strong> among<br />

Survivors of <strong>Breast</strong> <strong>Cancer</strong><br />

Bernstein JL. Memorial Sloan-Kettering <strong>Cancer</strong> Center, New York, NY<br />

Of the nearly 226,870 US women expected to develop breast cancer in<br />

2012 and the over 3 million breast cancer survivors currently residing<br />

in the US, five to ten percent will develop a subsequent primary in the<br />

contralateral breast (CBC) (www.cancer.org). This risk of developing<br />

CBC is higher than the overall risk of breast cancer in the general<br />

population. The rising incidence of breast cancer, coupled with an<br />

improved survival potential after cancer diagnosis has placed an<br />

increased number of women at risk for CBC, yet the epidemiology is<br />

poorly understood. Despite common genetics and exposures shared<br />

by both breasts, only a fraction of women who survive their first<br />

primary will develop a second primary in the contralateral breast.<br />

Epidemiologic studies have identified factors associated with an<br />

increased risk of developing CBC, including family history of breast<br />

cancer, early age at diagnosis, hormonal factors, reproductive history,<br />

body-mass index, and tumor characteristics of the first primary e.g.,<br />

lobular histology, stage and estrogen receptor (ER) status. The risk of<br />

developing CBC has also been associated with mutations of specific<br />

genes including BRCA1, BRCA2, and CHEK2. Importantly, the<br />

treatment a woman receives for her first primary breast cancer can<br />

also influence her risk of developing CBC; chemotherapy, hormonal<br />

therapies and tamoxifen significantly reduces the risk of developing<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

80s<br />

<strong>Cancer</strong> Research


December 4-8, 2012<br />

<strong>Abstract</strong>s: Invited Speakers<br />

CBC while the radiation received to the contralateral breast during<br />

radiotherapy increases the risk of CBC. Despite the number of<br />

established risk factors for increased CBC risk, their combined effect<br />

accounts for only a small portion of the CBCs that develop each year.<br />

It is unclear whether, and to what extent, genetic factors and radiation,<br />

individually or via interaction, contribute to the development of CBC.<br />

This talk will review the state of the field.<br />

ES5-3<br />

Investigating <strong>Breast</strong> <strong>Cancer</strong> with Single-Cell Sequencing<br />

Navin NE. UT MD Anderson <strong>Cancer</strong> Center, Houston, TX<br />

Tumors evolve from single cells. As they evolve, they acquire complex<br />

somatic mutations and diversify, forming distinct subpopulations of<br />

cells. This intratumor heterogeneity confounds basic research and<br />

clinical practice because tools do not exist to resolve it. Standard<br />

genomic methods are limited to reporting an average signal from a<br />

complex population of cells, because they require a large amount of<br />

input material. These bulk methods may mask population diversity<br />

and rare cells in the tumor - that may be the most malignant. To<br />

address this limitation my laboratory has developed single-cell<br />

sequencing tools to delineate intratumor heterogeneity and investigate<br />

mutational evolution in human cancers. We developed a method to<br />

profile genomic copy number in single cells and used this method to<br />

study genome evolution in triple-negative (ER-/PR-/Her2-) breast<br />

tumors (Navin et al., 2011 Nature) which revealed a punctuated<br />

model for copy number evolution. Recently, we have extended this<br />

method to obtain whole-genome, high-coverage sequencing data<br />

from single human tumor cells. From this data we can detect the full<br />

spectrum of genomic mutations, including point mutations, indels and<br />

microdeletions in single human cells. We applied this method to study<br />

clonal diversity in an estrogen-receptor-positive breast carcinoma<br />

and sequenced four single cells in addition to a population sample.<br />

In the population we detected only 36 coding mutations. However,<br />

our whole-genome single-cell sequencing data revealed hundreds of<br />

additional coding mutations, suggesting extensive clonal diversity<br />

in the tumor. Our data from this tumor support a mutator phenotype<br />

model for tumor progression, in which cancer is driven by elevating<br />

the rate of random mutations. Our studies show that a wealth of<br />

information can be obtained from studying single cancer cells, data<br />

that could not be obtained by analyzing the tumor en masse. We expect<br />

single-cell sequencing to have major clinical applications in early<br />

detection, analyzing scarce tissue samples and monitoring residual<br />

disease in patients with cancer.<br />

ES6-1<br />

Sentinel Node Biopsy in <strong>Breast</strong> <strong>Cancer</strong>: Before or after<br />

Neoadjuvant Chemotherapy?<br />

Mamounas EP. Aultman Hospital<br />

Neoadjuvant chemotherapy (NC) was originally used to render<br />

inoperable breast cancer (BC) patients to operable candidates. Its use<br />

subsequently expanded to operable BC patients requiring mastectomy<br />

to convert them to candidates for breast conserving surgery. NC has<br />

also been shown to downstage axillary lymph nodes in up to 40-50%<br />

of patients. This observation, coupled with the development and<br />

validation of sentinel node biopsy (SNB) in patients with operable BC<br />

provided yet another potential advantage for the use of neoadjuvant<br />

over adjuvant chemotherapy, i.e. the potential to decrease the extent<br />

and morbidity of axillary surgery.<br />

Several small, single-institution studies have examined the efficacy<br />

of lymphatic mapping and the accuracy of SNB after NC with<br />

significant variability in the rate of SN identification and in the rate<br />

of false negative SN. Nonetheless, when these studies are examined<br />

collectively or when larger, multi-center data sets are analyzed, SNB<br />

after NC appears to have somewha lower rate of SN identification but<br />

similar rate of false negative SN compared to SNB before systemic<br />

therapy. This also appears to be the case even in patients who have<br />

documented involvement of the axillary nodes before NC. Results<br />

of two prospective studies on this subject (ACOSOG 1071 and<br />

SENTINA) will be presented at the 2012 SABCS.<br />

Some have proposed that candidates for NC should have SNB<br />

performed before NC rather than after. They argue that this approach<br />

obtains information on the status of the SN without the potential<br />

confounding effects of NC and SN negative patients can avoid axillary<br />

dissection. Although this approach may be useful in patients who will<br />

not need adjuvant/neoadjuvant chemotherapy if the SN is negative,<br />

it is not generally useful for the majority of candidates for NC. SNB<br />

before NC commits patients to two surgical procedures whether the<br />

SN is negative or positive and this approach does not take advantage<br />

of the down-staging effect of NC on the axillary nodes.<br />

Knowledge of the pathologic SN status before NC does not seem to<br />

help in deciding which NC regimen to use or whether to use additional<br />

adjuvant chemotherapy after surgery. Furthermore, the prognostic<br />

significance of negative axillary nodes after NC and prior positive<br />

SNB is dubious since node negativity in this setting may reflect<br />

axillary nodal down-staging or just the prior removal of all positive<br />

nodes by SNB. Lastly, it has been argued that knowing whether the<br />

axillary nodes are involved before NC may provide useful information<br />

regarding the need for postoperative XRT to regional lymph nodes<br />

or the chest wall (after mastectomy). Recent results on the rates and<br />

patterns of loco-regional recurrence (LRR) from two NSABP NC<br />

trials (B-18 and B-27) have shown that achievement of pathologic<br />

complete response (pCR) in the breast with histologically negative<br />

axillary nodes at surgery is an independent predictor of LRR and that<br />

LRR rates are low for patients who are clinically node positive before<br />

NC but histologically node-negative after NC, particularly if they<br />

have pCR in the breast. Thus, based on this data the value of knowing<br />

the pathologic status of the SN(s) before NC is further diminished.<br />

ES6-2<br />

Evolving Trends in Implant <strong>Breast</strong> Reconstruction<br />

Pusic A. Memorial Sloan-Kettering <strong>Cancer</strong> Center, New York, NY<br />

For women undergoing mastectomy, implant breast reconstruction<br />

is increasingly common. Over the past 5 years, new trends in<br />

implant reconstruction have been evolving. The use of ‘nextgeneration’<br />

silicone implants, acellular dermal matrices and fat<br />

grafting have expanded the range of options available to patients.<br />

In addition, for women undergoing both skin-sparing and nipplesparing<br />

mastectomies, there is growing experience with single-stage<br />

implant reconstruction (i.e. implant placement directly at the time of<br />

mastectomy without use of a tissue expander). Finally, indications<br />

for post-mastectomy radiation continue to increase and this has<br />

important implications for long-term patient satisfaction with implant<br />

reconstruction.<br />

ES7-2<br />

Tumor Dormancy from an Experimental Biologist’s Perspective<br />

Chambers AF. London Regional <strong>Cancer</strong> Program, London, ON,<br />

Canada<br />

Survival from breast cancer has improved steadily over the past<br />

decades. However, most breast cancer deaths are due to metastasis,<br />

and considerably less progress has been made against metastatic<br />

disease. We still know remarkably little about preventing or delaying<br />

www.aacrjournals.org 81s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

metastatic recurrence or effectively treating the cancer if it does recur.<br />

Making breast cancer treatment even more challenging is the fact<br />

that breast cancer patients may appear to be cured of their disease,<br />

only to have it return later. These recurrences can happen even<br />

decades after apparently successful primary treatment. This creates<br />

years of uncertainty for breast cancer patients, who do not know if<br />

they have been cured or if the cancer will return, and makes clinical<br />

management difficult.<br />

We have used experimental models, both in vitro and in vivo, to<br />

try to understand the process of breast cancer metastasis. We have<br />

found that a population of cancer cells can remain as dormant<br />

cells, and have shown that these cells can resist being killed by<br />

cytotoxic chemotherapy that successfully kills actively diving<br />

cells. Experimental studies raise questions that must be addressed<br />

clinically. For example, what is the prevalence of dormant cells in<br />

breast cancer patients – are they common or rare? Are there factors<br />

inherent to the cancer cells that influence the likelihood of dormant<br />

cells persisting and subsequently re-awakening? Conversely, are there<br />

host microenvironmental factors, such as diet, exercise or stress, that<br />

can influence whether dormant cells persist and re-awaken? This<br />

information will be important in learning better how to prevent, delay<br />

and treat metastatic breast cancer.<br />

ES8-1<br />

Inflammation and <strong>Breast</strong> <strong>Cancer</strong><br />

Condeelis J. Albert Einstein College of Medicine<br />

Mechanism-based insights into prognosis and treatment of breast<br />

cancer derived from high resolution multiphoton imaging<br />

John Condeelis, Thomas Rohan, Maja Oktay - Einstein, New York;<br />

Tony Ng – King’s College London, UK;<br />

Seema Agarwal, David Rimm – Yale, New Haven;<br />

Brian Robinson, Joan Jones – Weill Cornell Medical Center, New York<br />

Frank Gertler MIT, Boston.<br />

Multi-photon microscopy allows the observation of tumor cell<br />

behavior at single cell resolution in vivo and has demonstrated<br />

that invasive and migratory tumor cells in mammary tumors form<br />

migratory streams and intravasate only when associated with<br />

macrophages. This analysis demonstrates that tumor cell streaming<br />

and intravasation are cell autonomous behaviors that are the direct<br />

result of the tumor cell -macrophage interaction.<br />

The in vivo invasion assay was developed to collect these streaming<br />

and intravasation competent tumor cells from primary tumors in vivo.<br />

Collection of these cells was coupled to expression profiling to reveal<br />

the genes expressed by tumor cells during streaming and intravasation<br />

in rat, mouse and human breast tumors (the Invasion Signature).<br />

The invasion, adhesion and motility pathways identified in the<br />

Invasion Signature converge on the RhoC/Cofilin/Mena pathway<br />

identifying it as a master regulator of chemotaxis, streaming and<br />

intravasation of breast tumor cells in vivo. Using prognostic markers<br />

derived from the RhoC/Cofilin/Mena pathway, anatomical landmarks<br />

have been developed for use with breast cancer patients. One of these,<br />

composed of the tumor cell-macrophage intravasation complex, is<br />

called TMEM (Tumor MicroEnvironment for Metastasis) in human<br />

breast tumors. TMEM density in histology sections of breast tumor<br />

tissue has been found to predict metastatic risk.<br />

The potential for tumor cell/macrophage pairing, steaming and TMEM<br />

assembly can be detected in breast tumors by the relative expression<br />

of isoforms of Mena-INV (enhancer) and Mena11a (suppressor of<br />

migration and dissemination, referred to as the MenaCalc test). Two<br />

retrospective studies with breast cancer tissue from patients with<br />

known recurrence and survival outcome have demonstrated the use,<br />

for prediction of metastatic risk, of the MenaCalc test and the TMEM<br />

test in resected breast tumor tissue.<br />

A CofilinxP-Cofilin test, also derived from the RhoC/Cofilin/Mena<br />

pathway of the human Invasion Signature, is showing promise as<br />

a predictor of disease-free survival. The Cofilin part of the RhoC/<br />

Mena/Cofilin pathway is associated with the overall activity of a<br />

biologically relevant sub-network of proteins (centered around four<br />

seed proteins ezrin/radixin/moesin/cofilin) in the cancer proteome<br />

that control membrane-cytoskeletal linkage and tumor cell motility.<br />

CofilinxP-Cofilin co-localization intensity was the most biologically<br />

significant variable measured among these others in predicting<br />

survival in a large retrospective trial of breast cancer patients. The<br />

value of this test derives from the fact that it is independent of antiestrogen<br />

(hormonal) treatment and chemotherapy.<br />

In summary, multi-photon imaging in deep tissue locations in live<br />

animals in real time has revealed molecular mechanisms associated<br />

with complex cancer cell behavior during metastatic dissemination<br />

leading directly to new markers of metastatic risk and survival.<br />

ES8-2<br />

<strong>Breast</strong> <strong>Cancer</strong> Stroma: A Predictor of Clinical Outcome and<br />

Tumour Heterogeneity<br />

Bertos N, Finak G, Lesurf R, Saleh SM, Zhao H, Souleimanova M,<br />

Meterrisian S, Omeroglu A, Hallett M, Park M. McGill University,<br />

Montreal, QC, Canada<br />

<strong>Breast</strong> cancer heterogeneity is one of the principal obstacles both<br />

to predicting outcome and to determining an effective course of<br />

treatment for this disease. Individual cases demonstrate heterogeneity<br />

at multiple levels, including those parameters assessed by studies of<br />

gene expression, chromosomal aberrations, and classical and immunopathology.<br />

Although genomic technologies have been used to gain a<br />

better understanding of the impact of gene expression heterogeneity<br />

on breast cancer outcome by identifying gene expression signatures<br />

associated with clinical outcome, histopathological breast cancer<br />

subtypes, and a variety of cancer--related pathways and processes,<br />

relatively little is known about the effects of heterogeneity in the<br />

tumor microenvironment. We have addressed changes in stroma by<br />

analyzing changes in gene expression in stromal tissue associated<br />

with breast tumors when compared to normal breast tissue. We have<br />

integrated gene expression data from laser capture microdissected<br />

breast tumor stroma with matched normal stroma. Using this approach<br />

we have identified that the microenvironment of a breast tumor can<br />

be classified into one of six distinct molecular phenotypes exhibiting<br />

distinct biological functions and carrying prognostic information<br />

independent of existing therapeutic biomarkers and tumor subtypes.<br />

A trained predictor of 23 genes was developed and contains new<br />

information to stratify breast cancer subtypes. This is independent of<br />

clinical parameters and published predictors of outcome and identifies<br />

patients with poor outcome in multiple breast cancer expression data<br />

generated using using whole tissue. The stromal predictor selects<br />

poor outcome patients from multiple clinical subtypes of breast<br />

cancer and contains genes representing distinct biological features,<br />

including differential immune response, angiogenic response, as<br />

well as a hypoxic response. Elements of this signature are present in<br />

murine models of breast cancer. These results highlight the complex<br />

relationship between the tumor and its microenvironment, and<br />

underline the role that the stroma plays in tumor progression.<br />

These results demonstrate an important role for the tumor<br />

microenvironment in defining breast cancer heterogeneity, with a<br />

consequent impact upon clinical outcome. Novel therapies could be<br />

targeted at the processes that define the stroma classes, suggesting new<br />

avenues for the development of individualized treatment regimens.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

82s<br />

<strong>Cancer</strong> Research


December 4-8, 2012<br />

<strong>Abstract</strong>s: Invited Speakers<br />

ES9-1<br />

Adjuvant Treatment of HER2 Positive <strong>Breast</strong> <strong>Cancer</strong>: The Next<br />

Installment<br />

Pietenpol JA. Vanderbilt-Ingram <strong>Cancer</strong> Center; Vanderbilt<br />

University School of Medicine, Nashville, TN<br />

The treatment of HER2 positive breast cancer radically changed in<br />

2005; at ASCO that year adjuvant trials reported that the addition of<br />

trastuzumab consistently showed a major improvement in progression<br />

free survival in the treatment of early breast cancer that was HER2<br />

positive. A benefit was seen when trastuzumab was given either<br />

concurrently with chemotherapy or sequentially. The trials generally<br />

studied one year of trastuzumab although data from Finland showed<br />

a benefit for 3 months of treatment. Additional, yet to be reported<br />

studies, have explored durations of 6 months and 2 years. Initially<br />

there was concern about cardio toxicity and the potential for significant<br />

long-term harm, but the rate of cardio toxicity remains low and<br />

usually is seen early in the treatment. To further improve outcomes,<br />

new agents have been studied in the neo and adjuvant settings. The<br />

large ALTTO study randomized patients to either chemotherapy with<br />

either a year of trastuzumab, a year of lapatinib (the TKI inhibitor<br />

with efficacy in the metastatic arena), the combination of both agents<br />

or sequential use of each agent. The lapatinib monotherapy arm was<br />

shown to be inferior in an interim analysis and discontinued. Final<br />

data is pending although extrapolating from the NEOALTTO study,<br />

one may expect heightened activity from dual blockade. The BETH<br />

study added bevacizumab to trastuzumab in a 2-arm study trying to<br />

combat angiogenesis with HER2 blockade; this study has not reported.<br />

Although the same combination studied in the AVEREL study in the<br />

advanced setting did not show a benefit, this may not be the case in<br />

earlier disease and we need to wait for data. Dual blockade with two<br />

antiHER2 agents has been beneficial in the advanced setting and this<br />

has led to the APHINITY study, which is comparing chemotherapy<br />

with standard trastuzumab or trastuzumab with another antiHER2<br />

antibody, pertuzumab. This study is accruing well and will provide<br />

evidence for whether dual blockade with two antibodies is of benefit.<br />

It will also provide some clinical trial data for small tumors, as these<br />

are eligible for enrollment in this trial. Other new agents, including<br />

TDM 1, are being taken into the neoadjuvant setting and will be in<br />

adjuvant trials soon. Subcutaneous trastuzumab may provide easier<br />

treatment for women over their year of adjuvant therapy. A big<br />

question is whether all HER2 tumors are the same or whether we can<br />

begin tailoring treatment according to molecular features. Are there<br />

some cancers that will be cured with a short course of chemotherapy<br />

and trastuzumab while others may need multiple antiHER2 agents<br />

combined to acquire the same result? With the collection of tumor<br />

samples we can hopefully begin to further understand this. Tailored<br />

treatment has the potential to decrease toxicity and improve quality of<br />

life issues, provide more rational use of costly resources, and finally<br />

develop more intensive therapy for resistant tumors thus improving<br />

outcomes overall.<br />

PL-1<br />

Hormones and the <strong>Breast</strong>: Local and Environmental Interactions<br />

Brisken C, Rajaram R, Ayyannan A, Beleut M, Yalcin O. Ecole<br />

Polytechnique Fédérale de Lausanne (EPFL); ISREC/SV; EPFL;<br />

Izmir Institute of Technology<br />

A woman’s reproductive history affects her risk to get breast cancer.<br />

In particular, the number of menstrual cycles she experiences<br />

correlates with risk. We use tissue recombination experiments and<br />

different mouse mutant models to discern the roles of the ovarian<br />

hormones estrogens and progesterone act in the mammary gland.<br />

While estrogens drive pubertal development, progesterone is the<br />

major driver of proliferation in the adult mammary epithelium. Both<br />

steroids rely strongly on paracrine signaling to elicit cell proliferation<br />

and other biological effects.<br />

Perinatal exposure to environmentally relevant concentrations of<br />

a high production volume chemical, bisphenol A, that is detected<br />

in human due to exposure through consumer products, results<br />

in persistent changes in the mammary gland. The cell number is<br />

increased in in adult females as is the response to progesterone.<br />

ML-1<br />

William l. McGuire Memorial Lecture: Neoadjuvant Systemic<br />

Therapy: Promising Experimental Model, or Improved Standard<br />

of Care?<br />

Hortobagyi GN. UT MD Anderson <strong>Cancer</strong> Center, Houston, TX<br />

Neoadjuvant chemotherapy (NACT) was developed to improve the<br />

management of largely inoperable breast cancers (BC), including<br />

inflammatory BC. In the early 1970s, despite heroic efforts combining<br />

radical surgical resection with high doses of radiotherapy, local failure<br />

rate remained between 20% and 50% and 5-year overall survival (OS)<br />

rates rarely approached 20%. With the introduction of anthracyclinecontaining<br />

combination chemotherapy regimens, most large primary<br />

tumors responded to chemotherapy, with greater than 50% reductions<br />

in almost 90% of patients. Clinical complete remissions (CR) were<br />

reported in about 10% of such large tumors. These results were<br />

obtained when adjuvant chemotherapy was in its earliest clinical<br />

evaluation. Our group at the MD Anderson <strong>Cancer</strong> Center and others<br />

pioneered multidisciplinary strategies consisting of NACT, followed<br />

by surgery and radiotherapy. Such strategies rapidly became the<br />

standard of care for locally advanced BC and inflammatory BC,<br />

improving local control rates to >80%, and 5-year OS rates to >30%.<br />

In the early 1980s, we described the concept of pathological CR (pCR)<br />

and its correlation with favorable long-term OS. We also described<br />

the clinical and pathological correlates of pCR: thus, most pCRs were<br />

observed in patients with estrogen receptor (ER)-negative, high grade<br />

and highly proliferative tumors. pCRs were rarely observed when<br />

tumors had the opposite characteristics. Achieving major clinical<br />

responses expanded breast conserving surgery to patients with large,<br />

even locally advanced BC. Additional advantages to these strategies<br />

included the ability to monitor response to NACT, both clinically<br />

and through serial biopsies, thus documenting the biological effects<br />

of systemic treatments. By 1990, the first neoadjuvant endocrine<br />

therapies were introduced in patients with ER-positive BC, who were<br />

largely refractory to NACT. With the initial steps in high throughput<br />

genomic assays, our group focused on the development of prognostic,<br />

and later predictive profiles based on gene expression. These research<br />

directions rapidly established the importance of molecular subtypes.<br />

The old paradigm, based on a monolithic concept of BC was rapidly<br />

displaced by the 4-5 main molecular (and biomarker defined) BC<br />

subtypes. In little over 10 years, selection of standard treatment<br />

became dependent on four clinical biomarkers, while novel, RNA,<br />

DNA and protein based profiles became rapidly established in our<br />

diagnostic toolbox. Our field is rapidly evolving toward baseline<br />

identification of molecular anomalies that might serve as targets for<br />

novel, targeted agents (antibodies, kinase inhibitors, etc.). The proof<br />

of concept was trastuzumab and other HER-2-directed agents. Drug<br />

development is rapidly moving from advanced, refractory metastatic<br />

BC to the neoadjuvant setting, where modest size clinical trials<br />

promise to accelerate clinical assessment of new drugs, delivering<br />

them to the patients most likely to benefit from them. Neoadjuvant<br />

systemic therapy is at least equivalent to adjuvant systemic therapy<br />

www.aacrjournals.org 83s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

and provides practical and scientific advantages that make it preferable<br />

in many subsets of BC. It is a new standard of care while continuing<br />

to serve as an excellent research tool.<br />

CS1-1<br />

Treatment on the Edges: Discordance between Stage and Biology<br />

Albain KS. Loyola University Chicago Stritch School of Medicine<br />

This Clinical Science Forum addresses one of the mort vexing issues<br />

that faces a clinician when guiding patients in adjuvant therapy<br />

decisions – what to do for treatment “on the edges”? That is, when<br />

there is discordance between stage/risk of relapse and biology, which<br />

should most influence the choice of therapy? The first presentation<br />

will address bad stage or high risk disease but “good” biology, and the<br />

second talk will consider good stage/traditionally lower risk disease<br />

but a “bad” biologic profile. This abstract provides an overview of<br />

the first scenario.<br />

Historically, in the era predating knowledge about biologic subtypes<br />

and without systemic adjuvant therapy, surgical series reported<br />

probable cures in patients with large and even locally advanced<br />

tumors, as well as in a subset with positive nodes. This data base<br />

serves as a backdrop for more modern series in which there is a wide<br />

range of long-term survival potential for patients with tumors that have<br />

adverse prognostic features. Currently, it is considered standard of<br />

care to offer systemic adjuvant chemotherapy, endocrine therapy and/<br />

or HER2 targeted treatments to all such patients – with the particular<br />

therapeutic choice based on tumor biology. In particular, it is not wise<br />

to withhold therapy in these higher risk (“bad stage”) scenarios when<br />

the disease is triple negative or HER2-positive.<br />

However, across the luminal subtypes, there is great variation in<br />

outcome even when tumors are large and/.or nodes are positive.<br />

Some of these patients will require only endocrine therapy despite<br />

the high risk status, whereas others need more creative therapeutic<br />

therapies to circumvent multi-pathway drivers of tumor growth<br />

and progression. Subsets defined by high levels of expression of<br />

the estrogen receptor and low proliferation have a better outcome<br />

and often are cured with endocrine therapy alone. Refinement of<br />

prediction of which groups may do very well with this approach<br />

alone has been possible with the advent of multigene assays such as<br />

the 21 gene Recurrence score or the 70 gene assay. In fact, it can be<br />

argued that many patients in the high risk (“bad stage”) breast cancer<br />

group are over-treated with chemotherapy. For example, in SWOG<br />

S8814, a study in postmenopausal, ER+, node positive disease, the<br />

10 year breast cancer specific survival was over 90% in those with<br />

a low 21 gene RS assay and this outcome did not vary whether the<br />

adjuvant treatment was tamoxifen alone or anthracycline-based<br />

therapy followed by tamoxifen. Similar findings were reported for<br />

higher risk, node-negative patients in NSABP B20. The current status<br />

of the three large prospective studies designed to validate and refine<br />

prediction of chemotherapy benefit will be updated (TAILORx,<br />

RxPONDER and MINDACT).<br />

Finally, there is the other group of patients with high risk (“bad stage”)<br />

ER-positive disease whose risk remains high with tumor biology<br />

relatively resistant to standard chemotherapy and insufficient efficacy<br />

of the endocrine therapy. In this group, optimizing inhibition of cross<br />

talk with other pathways that take over driving tumor growth - despite<br />

the endocrine therapy blockade - is mandatory. Only then can “bad<br />

stage” be turned into an excellent outcome.<br />

CS1-2<br />

Small Tumor – Aggressive Biology: When They Collide!<br />

Piccart MJ, Azim Jr HA. Institut Jules Bordet, Brussels, Belgium<br />

Historically, tumor stage was the main stay in determining the<br />

prognosis and the need for adjuvant therapy in patients with operable<br />

breast cancer (BC). Patients with a tumor size < 1cm (pT1a, b)<br />

and negative nodal involvement (pN0) were regarded to have very<br />

favorable prognosis. This in turn was reflected in the way these<br />

patients were managed. Since a decade or more, it became obvious<br />

that BC is not a single disease, but a group of molecularly distinct<br />

subtypes. The appreciation of the biological diversity of BC subtypes<br />

has urged the question on the relevance of tumor stage in the decisionmaking,<br />

nowadays. In other words, should patients with early-stage<br />

disease continue to receive “milder” or even no systemic therapy<br />

being historically known to have favorable prognosis, even if they<br />

are of aggressive nature?<br />

Evidence in this regards mainly rely on retrospective data as these<br />

patients are often excluded from randomized trials. Data based on<br />

analyzing untreated cohorts have shown that the 5-year relapse-free<br />

survival of patients with pT1a, b pN0 disease is rather favorable, in<br />

the range of 90%. However, obvious differences in outcome were<br />

observed according to BC subtype. Patients with ER-positive disease<br />

often have an even more favorable outcome reaching up to 95%<br />

unlike those with HER2 and t tumors in which 5-year RFS ranges<br />

around 75-80%. This calls for integrating biological information in<br />

managing these patients.<br />

Patients with ER-positive disease are composed of at least two<br />

subtypes, luminal-A and luminal-B.; the latter being more aggressive.<br />

It is unlikely that this differs in patients with pN0 disease. This was<br />

demonstrated by the Mammaprint®, which separate pT1a, b patients<br />

into two distinct groups with different outcome.<br />

Few data suggested that the addition of trastuzumab particularly<br />

in patients with pT1b is worthwhile. This is further endorsed by<br />

guidelines like NCCN and St Gallen. However, it is important to note<br />

that the exact magnitude of benefit of trastuzumab in these patients<br />

is unknown. It is plausible to assume that the relative risk reduction<br />

on relapse will be similar to that in patients with more advanced<br />

tumors, which is in the range of 50%. On the basis that these patients<br />

have an absolute risk of relapse in the range of 20-25%, it appears<br />

reasonable to consider trastuzumab, particularly in patients with pT1b,<br />

which were shown to experience more BC-related events compared<br />

to patients with pT1a.<br />

A similar argument exists on the added value of chemotherapy for<br />

patients with TN disease. Based on the recent EBCTCG overview,<br />

the relative risk reduction on the risk of relapse by administering<br />

chemotherapy does not exceed 30%. Acknowledging the absolute<br />

risk of relapse of the pT1a, b pN0 patients, the absolute benefit from<br />

offering chemotherapy appears to be modest; especially for those<br />

with pT1a.<br />

Hence, the appreciation of BC heterogeneity on the molecular level is<br />

vital and should be considered in managing patients with very small<br />

tumors. As these patients have “relatively” low risk of relapse, it is<br />

important to consider absolute risk reductions that are foreseen by the<br />

addition of chemotherapy ± trastuzumab. This would help refining<br />

management strategies and hence improve counseling of patients<br />

diagnosed with these relatively indolent, yet challenging tumors.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

84s<br />

<strong>Cancer</strong> Research


December 4-8, 2012<br />

<strong>Abstract</strong>s: Invited Speakers<br />

BS1-1<br />

Essential Role for Wnt Signaling in Metastatic Colonization<br />

Huelsken J. EPFL (Swiss Federal Institute of Technology Lausanne),<br />

Lausanne, Switzerland<br />

Metastatic growth in distant organs is the major cause of cancer<br />

mortality. Formation of metastasis is a multi-step process with several<br />

rate-limiting steps: while dissemination of tumour cells appears to<br />

be an early and frequent event, successful initiation of metastatic<br />

growth, a process termed metastatic colonization, is rather inefficient<br />

and accomplished only by a minority of cancer cells that reach distant<br />

sites. We now identify two key mechanisms essential for the initiation<br />

of metastasis: first, a small population of cancer stem cells that are<br />

critical for metastatic colonization, i.e. the initial expansion of cancer<br />

cells at the secondary site. Second, we find stromal niche signals to<br />

be crucial for this process. This niche is required to promote Wnt<br />

signaling in infiltrating cancer stem cells and thereby this pathway<br />

plays an essential role in metastatic colonization. We suggest that<br />

interfering with this stromal support may represent a novel treatment<br />

strategy against metastatic disease.<br />

BS1-2<br />

Reactive Stroma as Therapeutic Targets in <strong>Breast</strong> <strong>Cancer</strong> Bone<br />

Metastasis<br />

Kang Y. Princeton University, Princeton, NJ<br />

Metastasis represents the most devastating stage of cancer<br />

progression. Bone metastasis is a frequent occurrence in breast cancer,<br />

affecting more than 70% of late stage cancer patients. Discovering<br />

bone metastasis genes that are clinically relevant and functionally<br />

important are critical for the development of novel therapeutics<br />

for high-risk breast cancer patients. We apply a multidisciplinary<br />

approach to analyze the molecular basis of breast cancer bone<br />

metastasis, combining functional genomics tools with animal models<br />

and clinical analysis of cancer metastasis. Candidate bone metastasis<br />

genes were identified from gene expression profiling of highly bone<br />

metastatic cells derived after in vivo selection in animal models.<br />

Functional characterization of these genes revealed their novel role<br />

in mediating tumor-stromal interactions essential for the formation of<br />

osteolytic bone metastasis. We discovered a novel metalloproteaseinitiated,<br />

EGFR-dependent paracrine signaling cascade among tumor<br />

cells, osteoblasts and osteoclasts that promotes the maturation of<br />

bone-degrading osteoclasts and formation of osteolytic lesions. Using<br />

in vivo imaging technology, we showed that TGFbis released from<br />

bone matrix upon bone destruction, and signals to breast cancer to<br />

further enhance their malignancy in developing bone metastasis. We<br />

furthered identified Jagged1 as a TGFb target genes in tumor cells that<br />

engaged bone stromal cells through the activation of Notch signaling<br />

to provide a positive feedback to promote tumor growth and to activate<br />

osteoclast differentiation. More recently, we identified osteoclastderived<br />

miRNAs as biomarkers and therapeutic targets of osteolytic<br />

bone metastasis. Importantly, pharmaceutical inhibitors against<br />

EGFR, TGFb and Notch signaling pathways and miRNAs blocking<br />

osteoclast differentiation significantly reduce the development of<br />

bone metastasis, suggesting novel avenues for clinical management<br />

of skeletal complications of breast cancer.<br />

MS1-1<br />

Targeting PI3K<br />

Cantley L. Beth Israel Deaconess Medical Center<br />

Targeting PI3K<br />

Lewis C. Cantley<br />

Beth Israel Deaconess Medical Center and Harvard Medical School,<br />

Boston, MA USA<br />

Phosphoinositide 3-Kinase (PI3K) is a central enzyme in a signaling<br />

pathway that mediates cellular responses to growth factors. The<br />

signaling pathway downstream of PI3K is highly conserved from<br />

worms and flies to humans and genetic analysis of the pathway<br />

has revealed a conserved role in regulating glucose metabolism<br />

and cell growth. Mutational events that lead to hyperactivation of<br />

the PI3K pathway result in hamartoma syndromes and cancers.<br />

Activating mutations in PIK3CA, encoding the p110alpha catalytic<br />

subunit of PI3K (PIK3CA) or inactivating mutations in PTEN, a<br />

phosphoinositide 3-phosphatases that reverses the effects of PI3K,<br />

are among the most common events in solid tumors, especially breast<br />

cancers. Drugs that target PI3K are in clinical trials for a variety of<br />

cancers. It is likely that PI3K pathway inhibitors will need to be<br />

combined with other drugs to be broadly effective. We have employed<br />

genetically engineered mouse models that develop cancers due to<br />

mutations in genes in the PI3K pathway and are using these models<br />

to explore the efficacy of PI3K pathway inhibitors as single agents or<br />

in combination with other drugs. We are also interrogating the role of<br />

PI3K in the growth and survival of Brca1/p53 mutant breast cancers.<br />

The role of PI3K inhibitors for treating cancers in these mouse models<br />

and in human trials will be discussed.<br />

MS1-3<br />

Clinical Development of mTOR Inhibitors in <strong>Breast</strong> <strong>Cancer</strong><br />

André F. Gustave Roussy Institute<br />

mTOR is a serine/threonine kinase involved in cancer cell metabolism<br />

and protein synthesis. mTOR induces phosphorylation of estrogen<br />

receptor and resistance to endocrine therapy in preclinical models.<br />

Based on this rationale, clinical trials have evaluated whether first<br />

generation of mTOR inhibitors could reverse resistance to endocrine<br />

therapy. Two randomized trials, including one registration trial,<br />

have reported that everolimus improves progression-free survival<br />

in patients resistant to non-steroidal aromatase inhibitors. These<br />

data are consistent with a phase II randomized trial performed in<br />

the neoadjuvant setting. One large phase III trial performed with<br />

temsirolimus failed, mainly due to dose intensity of the drug and<br />

patient population. Overall, these finding suggest that rapalogs could<br />

reverse resistance to endocrine therapy. These data open several<br />

avenues in the field of breast cancer.<br />

First, two trials are starting that evaluate the efficacy of everolimus in<br />

the adjuvant setting. These trials evaluate everolimus either frontline<br />

or after 2-3 years of endocrine therapy.<br />

Second, rapalogs are being tested in other breast cancer subtypes<br />

including Her2-overexpressing breast cancers and triple negative<br />

breast cancers. Indeed, mTOR has been shown to be involved<br />

in trastuzumab resistance in preclinical studies. Early trials have<br />

suggested that mTOR inhibition could reverse trastuzumab resistance.<br />

mTOR inhibitors have also been shown to synergize with cisplatin in<br />

preclinical models, and this combination is currently being evaluated<br />

in a phase II randomized trial.<br />

Finally, several mechanisms of resistance to rapalogs have been<br />

identified. This includes positive feedback loops and lack of optimal<br />

bioactivity on p4EBP1. Rapalogs indeed mediate activating feedback<br />

loops that in turn induce a paradoxical activation of kinase pathways.<br />

www.aacrjournals.org 85s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Based on this observation, several trials have started that evaluate the<br />

combination between Rapalogs and IGF1R inhibitors. Preliminary<br />

results from phase I trial suggest that this combination is active in<br />

ER+/highly proliferative disease. Although they induce optimal<br />

inhibition of pS6K, several studies have reported that rapalogs could<br />

be suboptimal to inhibit p4EBP1, a protein downstream of mTORC1.<br />

4EBP1 is involved in protein synthesis. Second generation mTOR<br />

inhibitors (mTORC1/C2 inhibitors) present an increased bioactivity<br />

on 4EBP1, in addition to avoid mTORC2-mediated feedback loop.<br />

This second generation mTOR inhibitor is being tested in early<br />

phase trials.<br />

Overall, rapalogs have opened the path for the development of mTOR<br />

inhibitors in the field of breast cancer. This will further improve<br />

outcome by exploring new indications and by developing strategies<br />

to reverse resistance. Finally, the development of mTOR inhibitors<br />

will dramatically increase our knowledge on the role of metabolism<br />

and protein translation in cancer progression.<br />

BL-1<br />

Brinker Award Lecture: How Does HER2 Contribute to <strong>Breast</strong><br />

<strong>Cancer</strong> Progression?<br />

Yarden Y. Weizmann Institute, Rehovot, Israel<br />

HER2 (also called NEU and ERBB2), a receptor tyrosine kinase<br />

frequently overexpressed in breast cancer, is one of the most<br />

oncogenic kinases of the human kinome. Multiple mechanisms<br />

contribute to the transforming potential of HER2. Although HER2 is<br />

structurally similar to the receptors for EGF (EGFR) and neuregulins<br />

(ERBB3 and ERBB4), the cytoplasm-facing kinase domain of HER2<br />

is characterized by relatively high basal activity, and unlike the<br />

ectodomains of EGFR and ERBB3/ERBB4, HER2?s extracellular<br />

part cannot bind any known growth factor with high affinity.<br />

Nevertheless, HER2 acts as the preferred partner of the other three<br />

receptors, and the heterodimers it forms display not only high kinase<br />

activity but also increased binding affinity toward a broad spectrum of<br />

growth factors. In addition, HER2-containing heterodimers typically<br />

interact with a relatively wide set of cytoplasmic effector proteins,<br />

such as the phosphoinositol 3-kinase and SHC, which recruits the<br />

mitogen-activated protein kinase pathway. The collective outcome<br />

of these functional features translates to prolonged and enhanced<br />

signaling capacity.<br />

One way to comprehend HER2 is offered by the evolutionary<br />

history of the family of receptor tyrosine kinases. Accordingly, HER2<br />

and EGFR share a common precursor, but genome duplications<br />

denied a growth factor from HER2, and in parallel inactivated<br />

the catalytic function of ERBB3, to achieve a layered signaling<br />

network, whose autonomy and steady state are maintained by a<br />

dense web of feedback regulatory loops. In line with this scenario,<br />

HER2 cannot signal in isolation, but requires an activating growth<br />

factor receptor. Once recruited, HER2 enables evasion of negative<br />

feedback regulation, such as receptor ubiquitination and intracellular<br />

degradation: Unlike homodimers of EGFRs, which are targeted<br />

to lysosomes for degradation, HER2-containing heterodimers are<br />

targeted to a recycling pathway that replenishes the surface pool of<br />

HER2 molecules and further prolongs signaling.<br />

In-depth understanding of HER2?s oncogenic functions<br />

holds promise for therapy of a fraction of breast cancer patients. For<br />

example, the relatively high basal activation of the kinase domain<br />

means that HER2 relies on a chaperone called HSP90, hence blocking<br />

HSP90 might retard HER2-overexpressing tumors. Likewise, it is<br />

predictable that agents able to block either recycling of HER2 or its<br />

ability to form heterodimers, would curtail tumor growth.<br />

BL-2<br />

Brinker Award Lecture: Older Women and <strong>Breast</strong> <strong>Cancer</strong>:<br />

Challenges and Opportunities<br />

Muss HB. University of North Carolina, Chapel Hill, NC; , Chapel<br />

Hill, NC<br />

In the United States and other developed nations breast cancer<br />

incidence and mortality rates rise dramatically with increasing age.<br />

This fact, coupled with the aging of the U.S population is leading to<br />

a tsunami of older women with breast cancer. Most oncologists lack<br />

training in geriatrics and the frequent comorbidities in older patients<br />

along with a new diagnosis of breast cancer makes treatment selection<br />

challenging. Little data from clinical trials are available to guide<br />

physicians in treatment selection and a paucity of geriatricians makes<br />

it difficult to develop comprehensive treatment plans for many older<br />

patients. Newer tools that are validated, fast, and user-friendly are now<br />

available to help busy clinicians do meaningful geriatric assessments<br />

which are necessary for evaluating the medical, functional, cognitive,<br />

psychological, and social status of older patients. Such tools can<br />

be used to accurately estimate survival in older patients and more<br />

recently to predict chemotherapy related toxicity. For older patients,<br />

maintenance of independent living and cognitive function are their<br />

major goals and such preferences must be taken into consideration<br />

when planning treatment. Older women, like younger women, are<br />

interested in body image and when possible should be offered breast<br />

preservation with lumpectomy and radiation. In some older women<br />

treated with lumpectomy and adjuvant endocrine therapy for small<br />

tumors that are hormone-receptor positive and lower grade, breast<br />

radiation may be omitted with only a small increase in the risk of<br />

in-breast recurrence. The majority of older patients with early stage<br />

breast cancer have hormone receptor positive, HER-2 negative breast<br />

cancers and are excellent candidates for endocrine therapy. The<br />

major issue in these patients is defining the potential added value<br />

of chemotherapy. Elders with HER-2 positive tumors may be good<br />

candidates for chemotherapy and trastuzumab, while for many with<br />

“triple-negative” lesions, chemotherapy is the option of choice. Older<br />

patients with metastatic breast cancer whose tumors have progressed<br />

after endocrine therapy can be considered for palliative chemotherapy<br />

and many single agent regimens are well tolerated in this setting. In<br />

these patients, making sure they are offered “structured” palliative<br />

care as part of their treatment program is integral to maximizing<br />

quality of life and possibly even prolonging survival. Lastly, eligible<br />

elders should be offered clinical trials participation. Although getting<br />

elders on trials remains a challenge, it’s essential we work diligently<br />

on recruitment so as to obtain meaningful data on outcomes and<br />

toxicities of newer treatment regimens. Opportunities for trials of all<br />

types abound, not only for testing new treatments, but for defining<br />

interventions that minimize toxicity and loss of function. All of us<br />

in oncology will need to learn how to care for older cancer patients<br />

to provide them with optimal treatment.<br />

PL-2<br />

Estrogen Receptor Cistrome: Implications for <strong>Breast</strong> <strong>Cancer</strong><br />

Carroll JS. <strong>Cancer</strong> Research UK; University of Cambridge<br />

Estrogen Receptor (ER) is the defining feature of luminal breast<br />

cancers, where is functions as a transcription factor. The traditional<br />

view of ER getting recruited to promoters of target genes is too<br />

simplistic. The recent discovery of ER-DNA interaction regions from<br />

ER+ breast cancer cell lines has revealed that ER rarely associates<br />

with promoter regions of target genes and instead associates with<br />

enhancer elements significant distances from the target genes. The<br />

genomic mapping of ER binding events also revealed the enrichment<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

86s<br />

<strong>Cancer</strong> Research


December 4-8, 2012<br />

<strong>Abstract</strong>s: Invited Speakers<br />

of DNA motifs for Forkhead factors. The Forkhead protein FOXA1<br />

(HNF3α) was subsequently shown to bind to ∼half of the ER binding<br />

events in the genome and was required for ER to maintain interaction<br />

with DNA. We have extended on these findings to map ER binding<br />

events in primary breast cancers and distant metastases. We find<br />

context dependent ER cis-regulatory elements (cistromes) that give<br />

insight into underlying transcriptional networks. These differential<br />

ER binding profiles correlate with clinical response in ER+ breast<br />

cancers. We experimentally explore the binding dynamics between<br />

drug sensitive and resistant contexts and identify properties that<br />

govern ER binding differences. These data suggest that ER-DNA<br />

interactions are dynamic and can be modulated by changes in FOXA1.<br />

We are currently exploring mechanisms that mediate FOXA1-DNA<br />

interactions, in order to better understand ER transcriptional activity<br />

in breast cancer biology.<br />

MS2-1<br />

Genetics and Epidemiology of Mammographic <strong>Breast</strong> Density<br />

Vachon CMM. Mayo Clinic College of Medicine<br />

Differences in the relative amounts of fat, connective and epithelial<br />

tissues in the breast lead to variations in mammographic appearance.<br />

<strong>Breast</strong> density is a measure that reflects this variation in tissue<br />

composition and is most commonly assessed as the absolute amount<br />

or proportion of the mammogram comprised of fibroglandular or<br />

non-fatty tissue. <strong>Breast</strong> density has been characterized using both<br />

qualitative and quantitative estimates. The most widely used clinical<br />

density measure is the American College of Radiology’s <strong>Breast</strong><br />

Imaging Reporting and Data System (BI-RADS) four-category<br />

tissue composition measure. Quantitative estimates include userassisted<br />

thresholding methods, and more recently, automated area and<br />

volumetric density measures. Increased breast density is common,<br />

with 26-32% of women in the general population having dense tissue<br />

occupying over half of their breast. <strong>Breast</strong> density has been shown to<br />

be a strong risk factor for breast cancer at any age of mammography,<br />

in both Caucasian and non-Caucasian populations, and years prior to<br />

the breast cancer. Women with over 50% of their breast comprised of<br />

dense tissue have a 3-5 fold increased risk of breast cancer relative<br />

to those with low or no density. Further, up to one third of all breast<br />

cancers are found in women with density over 50%.<br />

<strong>Breast</strong> density decreases with age, with the greatest declines seen<br />

between ages 45-55. This decline has been hypothesized to reflect<br />

the reduction of hormones and involution of the breast tissue with<br />

onset of menopause. Several risk factors demonstrate associations<br />

with percentage breast density that are consistent with their breast<br />

cancer associations, suggesting these factors may potentially affect<br />

breast cancer risk through their influence on breast density. Positive<br />

associations are seen between breast density and nulliparity, late<br />

age at first birth, never having breast fed, postmenopausal hormone<br />

use (particularly combination therapy), and alcohol intake. Inverse<br />

associations are seen with tamoxifen use as well as body mass index<br />

(BMI), but BMI and breast density have been shown to be independent<br />

risk factors for breast cancer, and likely act through different pathways<br />

on risk. Taken together, the epidemiologic risk factors account for<br />

only 20 - 30% of the variation in breast density in the population.<br />

Genetics also contribute to the variability in breast density. Twin and<br />

family studies have shown that percentage breast density is highly<br />

heritable, and that inherited factors explain between 30-60% of the<br />

variance. Some of these genetic factors are shared between breast<br />

density and breast cancer. Of the established breast cancer loci, single<br />

nucleotide polymorphisms in LSP1, ZNF365, ESR1 and RAD51L1,<br />

are also associated with breast density. Additional genetic loci for<br />

breast density (including a variant at 12q24) have been validated<br />

in large consortia. However, the loci identified to date together<br />

account for less than 5% of the variance in breast density. Identifying<br />

additional genetic loci for breast density will not only help inform the<br />

biology of breast density but also contribute to understanding breast<br />

cancer and risk prediction efforts.<br />

MS2-2<br />

<strong>Breast</strong> Density: Clinical Implications<br />

Boyd NF. Ontario <strong>Cancer</strong> Institute, Toronto, ON, Canada<br />

Potential clinical applications of mammographic density include:<br />

1) the prediction of outcome of breast disease, 2) mammographic<br />

screening, 3) research and practice of cancer prevention, and 4)<br />

improved prediction of risk in individuals.<br />

1) <strong>Breast</strong> cancer characteristics and clinical outcomes. Extensive<br />

percent mammographic density (PMD) is associated with increased<br />

risks for the development of most of the histological non-obligate<br />

precursors of breast cancer, including ductal carcinoma in situ (DCIS),<br />

atypical hyperplasia, hyperplasia without atypia, and columnar cell<br />

lesions (CLLs). PMD is associated with an increased risk of both<br />

ER+ and ER- tumors, and with larger tumors of higher histological<br />

grade. Risk of contralateral second breast cancer is also increased by<br />

extensive PMD. However, among women with breast cancer, risk<br />

of death from the disease does not appear to be increased by PMD.<br />

2) Mammographic screening. Women with extensive PMD are<br />

both at higher risk of breast cancer and at greater risk that cancer<br />

will not be detected by mammography. This heterogeneity is not<br />

currently recognized in the design of screening programs. Women<br />

with extensive PMD, might benefit from screening more often than<br />

once every 2 to 3 years and with modalities such as MR or UST in<br />

addition to mammography. Conversely, for women with radiolucent<br />

breast tissue and a negative screening mammogram, in whom risk is<br />

lower and detection easier, re-screening less frequently than every<br />

2 to 3 years with mammography might be safe. Research into these<br />

issues is required.<br />

3) <strong>Breast</strong> cancer prevention trials. Unlike most other risk factors for<br />

breast cancer, PMD can be changed), suggesting that it might be used<br />

as a surrogate marker in clinical trials of breast cancer prevention.<br />

Clinical trials of breast cancer prevention require large numbers of<br />

subjects and long periods of observation and thus are expensive.<br />

Smaller, shorter, and less expensive trials of prevention strategies<br />

would be possible if there were a surrogate marker that allow the<br />

identification of interventions that would reduce breast cancer<br />

incidence. To be used as a surrogate for breast cancer, a biomarker<br />

such as PMD should meet the following criteria: (a) the marker should<br />

be associated with risk of breast cancer, (b) the marker should be<br />

changed by the intervention, and (c) the change in the marker should<br />

mediate the effect of the intervention on breast cancer risk. The extent<br />

to which these criteria can currently be met will be discussed.<br />

4) Improved risk prediction. PMD is inversely associated with age,<br />

while breast cancer incidence increases with age. We used data from<br />

3250 healthy women aged 15-80 to estimate cumulative breast density<br />

(CBD) in the population, which increases with age, and compared this<br />

with age-specific incidence of breast cancer in Canada. Log CBD and<br />

log breast cancer incidence had a linear association (r=0.98; p=6.0<br />

E-9) that was unchanged by adjustment for log age (r= 0.97; p=8.0 E<br />

-7). Log CBD alone was a better predictor of age-specific log breast<br />

cancer incidence (r2=0.97) than age alone (r2=0.88). The observed<br />

strong association between CBD and breast cancer incidence suggest<br />

that the factors responsible for CBD may be causally related to the<br />

age-specific incidence of breast cancer.<br />

www.aacrjournals.org 87s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

PL-3<br />

<strong>Breast</strong> Radiotherapy: Fractionation and Other Fashions<br />

Yarnold J. Institute of <strong>Cancer</strong> Research & Royal Marsden NHS<br />

Foundation Trust, Sutton, United Kingdom<br />

Hypofractionation, defined as the use of fraction sizes >2.0 Gy,<br />

requires a reduction in the total dose delivered in 1.8 - 2.0 Gy<br />

fractions that takes account of the greater effect per Gy of larger<br />

fractional doses. Generations of radiation oncologists have been<br />

taught that small (≤2.0 Gy) daily fractions are always gentler on the<br />

dose-limiting healthy tissues than on the cancer, thereby optimising<br />

clinical benefit. This relationship applies at many tumour sites, but<br />

breast cancer is different. Well-designed randomised trials involving<br />

>7000 women confirm that there are no advantages to using ≤2.0<br />

Gy fractions, which spare breast cancer as much as the normal<br />

tissues. The limits of hypofractionation for whole breast/chest wall<br />

radiotherapy are under evaluation in the UK FAST Forward trial,<br />

testing 2 dose levels of a 5-fraction regimen given in 1 week against<br />

the 3-week UK standard. Hypofractionation can also be employed to<br />

match dose intensity more effectively to the spatial pattern of tumour<br />

relapse by modulating fraction size across the breast in preference<br />

to fraction number, an approach under test in the UK IMPORT<br />

HIGH trial. In marked contrast to conservative attitudes towards<br />

whole breast hypofractionation, there has been a dash to accelerated<br />

hypofractionation schedules among practitioners of partial breast<br />

radiotherapy.<br />

The mean fractionation sensitivity of breast cancer, expressed as an<br />

α/β value, is approximately 3 Gy. The distribution around this point<br />

estimate could be wide or narrow; we don’t know. The fractionation<br />

sensitivities of early responding normal tissues and cancers are<br />

postulated to reflect the responses of a limited number of target<br />

stem cell populations. Late-responding normal tissues are more<br />

complex in that multiple parenchymal and stromal cell lineages and<br />

interactions need to be considered. Despite potential complexity, a<br />

strong association between the proliferative indices of normal tissue<br />

parenchymal cells and fractionation sensitivity support the notion that<br />

the latter has a cellular basis, justifying a focus on classical cellular<br />

recovery and DNA double strand break (DSB) repair. Differences<br />

between healthy and malignant tissues in how they handle DSB are<br />

but one of several potential modulators of fractionation sensitivity.<br />

The processes are likely to be strongly affected by mutational and<br />

epigenetic alternations in tumours, but they foretell a future when<br />

predictive biomarkers replace historical dogma in determining<br />

fractionation regimens for cancer patients.<br />

BS3-2<br />

Molecular Imaging of <strong>Breast</strong> <strong>Cancer</strong>: Visualizing In Vivo <strong>Breast</strong><br />

<strong>Cancer</strong> Biology<br />

Mankoff DA, Chodosh LA. Perelman School of Medicine, University<br />

of Pennsylvania, Philadelphia, PA<br />

The ability to measure biochemical and molecular processes underlies<br />

progress in breast cancer biology and treatment. These assays have<br />

traditionally been performed by analysis of cell culture or tissue<br />

samples. More recently, functional and molecular imaging allows<br />

in vivo assay of biochemistry and molecular biology that is highly<br />

complementary to tissue-based assay. Molecular imaging can serve<br />

and number of roles, from characterization of pre-clinical models, to<br />

testing the ability of novel drugs to interact with their intended targets,<br />

to characterizing entire burden of disease for patients with breast<br />

cancer, to helping direct therapeutic choices and assign their efficacy.<br />

This talk will review basic principals of molecular imaging, with an<br />

emphasis on those methods of greatest translational potential. The<br />

session will discuss the use of pre-clinical models of breast disease to<br />

help test and validate new molecular imaging methods, and in turn,<br />

the use of molecular imaging to characterize the disease process in<br />

the animal models. The session will then review the current state of<br />

molecular imaging in breast cancer patients, including methods in<br />

routine clinical use, those undergoing advanced clinical trials, and<br />

those in early-phase testing.<br />

The session is designed for a broad audience, ranging from breast<br />

cancer translational scientists to practicing physicians caring for<br />

breast cancer patients.<br />

References<br />

1. Vernon AE, Bakewell SJ, Chodosh LA. Deciphering the molecular<br />

basis of breast cancer metastasis with mouse models. Rev Endocr<br />

Metab Disord. 2007 Sep;8(3):199-213.<br />

2. Mankoff DA. Molecular imaging as a tool for translating breast<br />

cancer science. <strong>Breast</strong> <strong>Cancer</strong> Res. 2008;10 Suppl 1:S3.<br />

3. Specht JM, Mankoff DA. Advances in molecular imaging for breast<br />

cancer detection and characterization. <strong>Breast</strong> <strong>Cancer</strong> Res. 2012 Mar<br />

16;14(2):206.<br />

MS3-3<br />

Navigating the Genome: Quandaries and Approaches to Dealing<br />

with Genomic Information<br />

Evans JP. University of North Carolina at Chapel Hill, Chapel Hill,<br />

NC<br />

Recent technological advances have made large-scale DNA<br />

sequencing, including whole genome sequencing, a practical reality.<br />

The ability to sequence tumor genomes as well as the germ-line<br />

genome of individuals holds considerable promise for the practice<br />

of oncology. Broad assessment of cancer risk in individuals,<br />

identification of those at extreme risk for cancer who might benefit<br />

from specific interventions and pharmacogenomic guidance regarding<br />

therapy are possible benefits of assessing an individual’s genome.<br />

Likewise, analysis of genomic information in tumors is poised to<br />

begin helping to guide highly specific treatment of cancer in an<br />

increasingly individualized manner.<br />

However, as the field of oncology begins to use genomic queries to<br />

assess patient risk, characterize disease and guide treatment, many<br />

challenges must be addressed. Purely technical issues include the<br />

suboptimal accuracy of current platforms for massively parallel<br />

sequencing, tumor heterogeneity and difficulties interpreting genomic<br />

risk information in a clinically relevant manner. Systemic challenges<br />

include how to interpret, store and present massive amounts of<br />

genomic data in the currently highly fragmented (and limited)<br />

electronic medical record. Finally, in the process of genome-scale<br />

sequencing vast numbers of incidental results are inevitably generated,<br />

which may have important ramifications for patients and providers<br />

but may be unwelcome. Such ethical, legal and social implications<br />

of whole genome sequencing raise significant questions about patient<br />

autonomy, the physician’s duty to warn and the laboratory’s duty to<br />

report a wide variety of highly heterogeneous genomic findings. In<br />

this presentation some of these salient issues and possible solutions<br />

will be explored.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

88s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: General Session 1<br />

S1-1<br />

Relative effectiveness of letrozole compared with tamoxifen for<br />

patients with lobular carcinoma in the BIG 1-98 trial.<br />

Metzger O, Giobbie-Hurder A, Mallon E, Viale G, Winer E,<br />

Thürlimann B, Gelber RD, Colleoni M, Ejlertsen B, Bonnefoi H,<br />

Coates AS, Goldhirsch A, Gusterson B. BIG 1-98 Collaborative<br />

Group, International <strong>Breast</strong> <strong>Cancer</strong> Study Group<br />

Background: Invasive lobular carcinoma (ILC) is mostly hormone<br />

receptor-positive (ER+) and associated with higher risk of late<br />

relapse (> 5 years after diagnosis) when compared to invasive ductal<br />

carcinoma (IDC). In this analysis we investigate the magnitude of<br />

benefit of endocrine treatment (tamoxifen [tam] or letrozole [let])<br />

and the patterns of relapse over time in patients with ILC (classical<br />

subtype) or IDC enrolled in the monotherapy arms of BIG 1-98 study<br />

(12-year update).<br />

Material and methods: BIG 1-98 was a randomized, phase III study<br />

that compared five years of tam or let (monotherapy arms), or their<br />

sequences in post-menopausal women with ER+ early BC. There were<br />

4922 patients enrolled in the monotherapy arms of BIG 1-98. This<br />

analysis includes patients who had centrally-reviewed histology data<br />

(n = 4080) and classified as IDC and ILC (n = 3660). Centrally-defined<br />

ER, PgR, Ki-67 were used to defined ER+ subtypes (n = 2923). ER+<br />

subtypes were defined as Luminal A (ER+ and/or PR+ HER2- and<br />

Ki6799% TAM). With mean 7.1 woman-years<br />

follow-up (30,000 w-y in years 5-9, 16,000 in years 10-14, 2000 later),<br />

1328 recurrences were reported (900 in years 5-9, 379 in years 10-14).<br />

Recurrence was significantly lower with10 than 5 years TAM (Table:<br />

logrank 2p=0.002, rate ratio (RR)=0.90 se 0.06 in years 5-9 and 0.75<br />

se 0.08 in years 10+). So were both BCM (2p=0.01, RR=0.97 se 0.10<br />

in years 5-9 and 0.71 se 0.09 in years 10+) and all-cause mortality<br />

(2p=0.01, with no increase in non-BCM). Proportional risk reductions<br />

were homogeneous by country, age and stage. Kaplan-Meier risks<br />

in years 5-14 (K-M) were: recurrence 21.4 vs 25.1%, BCM 12.2 vs<br />

15.0%. Uterine cancer K-Ms in those randomized at age 50+ were:<br />

incidence 2.6 vs 1.6% (2p=0.08), mortality 0.2 vs 0.2%. In premenopausal<br />

women (where AIs are not an alternative to TAM) there<br />

was no apparent excess of uterine cancer.<br />

Discussion: Compared with just 5 years TAM, continuing TAM to<br />

year 10 safely protects further against recurrence and, particularly<br />

during the second decade, BCM. Combining results from ATLAS<br />

and the EBCTCG meta-analyses of 5 years TAM vs none (both had<br />

∼80% compliance), in a hypothetical trial of 10 vs 0 years TAM with<br />

∼80% compliance, 15-year BCM would be reduced by at least onethird.<br />

Hence, full compliance with 10 years TAM would yield even<br />

greater benefit (Table). Further follow-up of ATLAS will assess more<br />

reliably the apparently substantial mortality reduction in the second<br />

decade after diagnosis.<br />

ER+ disease: RR [95% CI] by time period from diagnosis for 5v0 years TAM in EBCTCG<br />

(n=10645, 80% complied); 10v5 years TAM in ATLAS (n=6846, ∼80% complied); and 10v0 years<br />

TAM (hypothetical)<br />

Time from<br />

diagnosis<br />

EBCTCG 5v0y<br />

TAM: ∼80%<br />

comply<br />

Product of trial RRs:<br />

ATLAS 10v5y TAM:<br />

effect of 10v0 y TAM<br />

∼80% comply<br />

if ∼80% comply<br />

RR for effect of<br />

10v0 y TAM if all<br />

comply**<br />

Recurrence<br />

0-4y 0.53[0.48-0.57]¢ 1 0.53[0.48-0.57]¢ 0.41<br />

5-9y 0.68[0.60-0.78]¢ 0.90[0.78-1.02] 0.61[0.51-0.73]¢ 0.52<br />

10+ 0.94[0.79-1.12] 0.75[0.62-0.90]* 0.70[0.54-0.91]* 0.63<br />

BCM<br />

0-4y 0.71[0.62-0.80]¢ 1 0.71 [0.62-0.81]¢ 0.64<br />

5-9y 0.66[0.58-0.75]¢ 0.97[0.79-1.18] 0.64 [0.50-0.82]† 0.55<br />

10+ 0.73[0.62-0.86]† 0.71[0.58-0.88]§ 0.52 [0.40-0.68]¢ 0.40<br />

**Estimated gain if all comply=1.25x gain if ∼80% comply. *2p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

S1-3<br />

DISCUSSANT<br />

William E. Barlow, PhD; SWOG Statistical Center; Fred Hutchinson<br />

<strong>Cancer</strong> Research Center; Seattle WA<br />

S1-4<br />

Final analysis of overall survival for the Phase III CONFIRM<br />

trial: fulvestrant 500 mg versus 250 mg<br />

Di Leo A, Jerusalem G, Petruzelka L, Torres R, Bondarenko IN,<br />

Khasanov R, Verhoeven D, Pedrini JL, Smirnova I, Lichinitser MR,<br />

Pendergrass K, Garnett S, Rukazenkov Y, Martin M. Hospital of<br />

Prato, Prato, Italy; Centre Hospitalier Universitaire Sart Tilman,<br />

Liège, Belgium; First Faculty of Medicine of Charles University,<br />

Prague, Czech Republic; Instituto Nacional del Cáncer, <strong>San</strong>tiago,<br />

Chile; Dnipropetrovsk Municipal Clinical Hospital, Dnipropetrovsk,<br />

Ukraine; Republican Clinical Oncological Center, Kazan, Russian<br />

Federation; AZ Klina, Brasschaat, Belgium; Hospital Nossa Senhora<br />

da Conceição, Porto Alegre, Brazil; Medical Radiological Science<br />

Center, Obninsk, Russian Federation; Russian <strong>Cancer</strong> Research<br />

Centre, Moscow, Russian Federation; Kansas City <strong>Cancer</strong> Center,<br />

Kansas City; AstraZeneca Pharmaceuticals, Macclesfield, United<br />

Kingdom; Hospital Universitario Gregorio Maranon, Madrid, Spain<br />

Background<br />

The COmparisoN of Faslodex In Recurrent or Metastatic breast<br />

cancer (CONFIRM) trial (NCT00099437) compared fulvestrant 500<br />

mg with fulvestrant 250 mg in postmenopausal women with locally<br />

advanced or metastatic estrogen receptor (ER)-positive breast cancer<br />

who had recurred or progressed following prior endocrine therapy.<br />

Fulvestrant 500 mg was associated with a statistically significant<br />

increase in progression-free survival compared with fulvestrant 250<br />

mg (Di Leo A et al. J Clin Oncol 2010; 28: 4594-4600). At the time of<br />

the primary analysis approximately 50% of patients had died. Median<br />

overall survival was 25.1 months and 22.8 months for fulvestrant 500<br />

mg and 250 mg, respectively (hazard ratio [HR] 0.84; 95% confidence<br />

interval [CI] 0.69, 1.03; p=0.091). A follow-up analysis of overall<br />

survival was subsequently planned for when 75% of patients had died<br />

and the results are presented here.<br />

Methods<br />

CONFIRM was a randomized, double-blind, parallel-group,<br />

multicenter, Phase III study. Patients were randomized 1:1 to either<br />

fulvestrant 500 mg (500 mg i.m. on Days 0, 14 and 28 and every 28<br />

days thereafter) or fulvestrant 250 mg (250 mg i.m. every 28 days).<br />

After the primary analysis, patients on fulvestrant 250 mg were<br />

permitted to switch to 500 mg and all patients were followed up<br />

for overall survival, regardless of treatment discontinuation, unless<br />

consent was withdrawn. Overall survival was analyzed using an<br />

unadjusted log-rank test. No adjustments were made for multiplicity.<br />

Serious adverse events (SAEs) were also reported.<br />

Results<br />

In total, 736 women (median age 61.0 years) were randomized<br />

between February 2005 and August 2007 from 128 centers in 17<br />

countries (fulvestrant 500 mg: n=362; fulvestrant 250 mg: n=374).<br />

At time of follow-up analysis, 63 (9.0%) patients were lost to followup,<br />

16 (2.2%) patients had withdrawn consent, 103 (14.0%) patients<br />

were still ongoing (21 [2.9%] on treatment and 82 [11.1%] not on<br />

treatment), and 554 (75.3%) patients had died. Eight (2.1%) patients<br />

crossed over from fulvestrant 250 mg to 500 mg. Median overall<br />

survival was 26.4 months for fulvestrant 500 mg and 22.3 months for<br />

fulvestrant 250 mg (HR 0.81; 95% CI, 0.69, 0.96; nominal p=0.016).<br />

During the treatment period, a total of 32 (8.9%) patients had at<br />

least one SAE in the fulvestrant 500 mg and 25 (6.7%) patients in<br />

the fulvestrant 250 mg groups, and SAEs that were causally related<br />

to study treatment were reported for 6 (1.7%) and 3 (0.8%) patients,<br />

respectively. SAEs with an outcome of death were reported for 5<br />

(1.4%) and 6 (1.6%) patients, respectively, during the treatment<br />

period. Overall, there were no clinically important differences in the<br />

profiles of SAEs between the treatment groups and no clustering of<br />

SAEs could be detected in either treatment group.<br />

Conclusions<br />

Overall survival data from CONFIRM demonstrate that, in patients<br />

with locally advanced or metastatic ER positive breast cancer,<br />

fulvestrant 500 mg is associated with a clinically relevant 4.1 month<br />

difference in median overall survival and 19% reduction in risk of<br />

death compared with fulvestrant 250 mg. There were no new safety<br />

concerns associated with the use of fulvestrant 500 mg.<br />

S1-5<br />

PIK3CA mutations are linked to PgR expression: a Tamoxifen<br />

Exemestane Adjuvant Multinational (TEAM) pathology study.<br />

Sabine VS, Crozier C, Drake C, Piper T, van de Velde CJH, Hasenburg<br />

A, Kieback DG, Markopoulos C, Dirix L, Seynaeve C, Rea D, Bartlett<br />

JMS. Ontario Institute for <strong>Cancer</strong> Research, Toronto, ON, Canada;<br />

University of Edinburgh <strong>Cancer</strong> Research Centre, Institute of Genetics<br />

& Molecular Medicine, Edinburgh, Scotland, United Kingdom;<br />

Leiden University Medical Center, Leiden, Netherlands; University<br />

Hospital, Freiburg, Germany; Elblandklinikum, Riesa, Germany;<br />

Athens University Medical School, Athens, Greece; St. Augustinus<br />

Hospital, Antwerp, Belgium; Erasmus Medical Center-Daniel den<br />

Hoed, Rotterdam, United Kingdom; University of Birmingham,<br />

Birmingham, United Kingdom<br />

Background: PIK3CA is mutated in about 26% of breast cancers<br />

(BC) and is the most frequently mutated gene in (BC). Almost 95% of<br />

mutations occur in exons 9 (E9) or 20 (E20). PIK3CA mutations may<br />

be associated with increased survival in endocrine-treated patients;<br />

however the impact of mutations in E9 vs E20 is not clear. We assessed<br />

10 common PIK3CA mutations (95% of all mutations), in ER-positive<br />

(+ve) samples from the TEAM pathology study (n=∼4500), and<br />

determined the impact of PIK3CA mutations on survival. We report<br />

an interim analysis of 1969 TEAM cases.<br />

Methods: DNA was extracted from formalin-fixed paraffin embedded<br />

sections. Mutational analyses were performed on 25 mutations<br />

in 6 genes (PIK3CAx10, Akt1x1, KRASx5, HRASx3, NRASx2 &<br />

BRAFx4), using Sequenom MassArray.<br />

Results: Mutations were found in PIK3CA: 37.5%; Akt1: 3.3%;<br />

KRAS: 0.3%; and BRAF: 0.1% of cases. No mutations were found<br />

in HRAS or NRAS (n=1969). 90% of PIK3CA mutations were<br />

located in E9 and E20. Outcome data was available for 1958/1969<br />

patients. Patients whose tumours contained any PIK3CA mutations<br />

(n=739) were at lower risk of distant metastasis, Hazard ratio=0.86<br />

(0.67 – 1.11), when compared to those without PIK3CA mutations<br />

(n=1219); although this difference was not statistically significant<br />

(Cox Regression; p=0.24). PIK3CA mutations were significantly<br />

more frequent in HER2-negative (-ve) (39%) than in HER2+ve<br />

samples (25%) (p=0.001), without evidence that PIK3CA mutations<br />

differentially impacted outcome in HER2+ve vs HER2-ve patients.<br />

A positive correlation was demonstrated between PIK3CA mutations<br />

and PgR Allred score (p=0.002) but not ER Allred score (p=0.37).<br />

With increasing PgR Allred score increase there is an increased<br />

frequency of mutations in E20 but not E9 (Table 1).<br />

Discussion: This study indicates a higher percentage of PIK3CA<br />

mutations in ER+ve BC samples than previously demonstrated, either<br />

for BC as a whole or for ER+ve cases, suggesting that in ER+ve early<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

90s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: General Session 1<br />

BC, PIK3CA mutations are more common than previously reported.<br />

Furthermore, increased PIK3CA mutation frequency is significantly<br />

associated with increasing PgR Allred score and this appears solely<br />

due to increased numbers of patients with E20 mutations further<br />

complicating the analysis of the impact of PIK3CA mutations in BC.<br />

This may explain current uncertainty regarding the impact of PIK3CA<br />

mutations in E9 vs E20 with respect to clinical outcome. Whilst we<br />

were unable to show a significant impact on outcome in patients<br />

whose tumours contained PIK3CA mutations, we believe the complex<br />

relationship between PgR expression (good prognosis indicator) and<br />

PI3K mutations requires further exploration in the full dataset using<br />

interaction techniques adjusting for the impact of PgR on outcome.<br />

Mutational analysis and correlation with clinical outcome data for<br />

the remaining ∼2500 DNA samples, along with the existing data for<br />

1969 patients, will be presented.<br />

PIK3CA mutation:<br />

PgR Allred<br />

score<br />

# cases All 10 Exon9 Exon20 Others<br />

0 - 4 503 30% 14% 13% 3%<br />

5 & 6 502 38% 16% 20% 2%<br />

7 & 8 878 42% 14% 22% 5%<br />

Table 1: Analysis of 10 common PIK3CA mutations in TEAM pathology study by PgR Allred score.<br />

S1-6<br />

Results of a randomized phase 2 study of PD 0332991, a cyclindependent<br />

kinase (CDK) 4/6 inhibitor, in combination with<br />

letrozole vs letrozole alone for first-line treatment of ER+/HER2-<br />

advanced breast cancer (BC)<br />

Finn RS, Crown JP, Lang I, Boer K, Bondarenko IM, Kulyk SO, Ettl<br />

J, Patel R, Pinter T, Schmidt M, Shparyk Y, Thummala AR, Voytko NL,<br />

Breazna A, Kim ST, Randolph S, Slamon DJ. University of California,<br />

Los Angeles, Los Angeles, CA; Irish Cooperative Oncology Research<br />

Group, Dublin, Ireland; Orszagos Onkologiai Intezet, Budapest,<br />

Hungary; Szent Margit Korhaz, Budapest, Hungary; Dnipropetrovsk<br />

City Multiple-Discipline Clinical Hospital, Ukraine; Municipal<br />

Treatment-and-Prophylactic Institution “Donetsk City Oncological<br />

Dispensary”, Ukraine; Technical University of Munich, Germany;<br />

Comprehensive Blood and <strong>Cancer</strong> Center, Bakersfield, CA; Petz<br />

Aladar Megyei Oktato Korhaz, Gyor, Hungary; University Hospital<br />

Mainz, Mainz, Germany; Lviv State Oncologic Regional Treatment<br />

and Diagnostic Center, Ukraine; Comprehensive <strong>Cancer</strong> Centers<br />

of Nevada, Henderson, NV; Kyiv City Clinical Oncology Center,<br />

Ukraine; Pfizer Oncology, New York, NY; Pfizer Oncology, <strong>San</strong><br />

Diego, CA<br />

Background: PD 0332991, a selective inhibitor of CDK 4/6, prevents<br />

cellular DNA synthesis by blocking cell cycle progression. Preclinical<br />

studies in a BC cell line panel identified the luminal ER subtype,<br />

elevated expression of cyclin D1 and Rb protein, and reduced p16<br />

expression as being associated with sensitivity to PD 0332991 (Finn<br />

et al. 2009). Synergistic activity was also observed in vitro when<br />

combined with tamoxifen. After determination of the recommended<br />

phase 2 dose in combination with letrozole (letrozole 2.5 mg QD<br />

plus PD 0332991 125 mg QD on Schedule 3/1), a randomized phase<br />

2 study comparing letrozole alone (L) to letrozole plus PD 0332991<br />

(L+P) was initiated.<br />

Methods: The phase 2 portion of the study was designed as a twopart<br />

study; Part 1 enrolled post- menopausal women with ER+/<br />

HER2- advanced BC; Part 2 in addition to ER+/HER2- as eligibility<br />

criteria, screened for CCND1 amplification and/or loss of p16 by<br />

FISH. The primary endpoint is progression-free survival (PFS);<br />

secondary endpoints include response rate, overall survival, safety,<br />

and correlative biomarker studies. In both parts, post-menopausal<br />

women with ER+/HER2- advanced BC were randomized 1:1 to<br />

receive letrozole either with or without PD 0332991. Pts continue<br />

on assigned study treatment until disease progression, unacceptable<br />

toxicity, or consent withdrawal, and are followed for tumor<br />

assessments every 2 months.<br />

Results: 66 pts were randomized in Part 1 and 99 pts in Part 2.<br />

Preliminary results from Part 1 of this study have been previously<br />

reported (IMPAKT <strong>Breast</strong> <strong>Cancer</strong> Conference, <strong>Abstract</strong> #292,<br />

Finn et al. May 2012) demonstrating a significant improvement in<br />

median PFS in the L+P vs. L arm (HR=0.35; 95% CI, 0.17 to 0.72;<br />

p=0.006). With the additional 99 pts randomized in Part 2 (N=165),<br />

the statistically significant improvement in median PFS (26.2 vs. 7.5<br />

months, respectively) continues to be observed with a HR=0.32 (95%<br />

CI, 0.19 to 0.56) with p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

The primary objective was to compare progression-free survival<br />

(PFS) between treatment arms. Secondary objectives were overall<br />

survival, time to treatment failure, overall response rate, response<br />

duration, clinical benefit rate, and safety. In total, 344 patients (172<br />

in each treatment arm) were needed to detect a hazard ratio of 0.69<br />

(corresponding to a median PFS of 9 months in the ET arm and 13<br />

months in the ET+B arm) with α=0.05 and β=0.2. With an expected<br />

drop-out rate of 10%, 378 patients were to be included. The efficacy<br />

analysis is pre-planned after 270 events.<br />

Results: From 11/2007 to 8/2011, 380 patients were randomized in<br />

Spain and Germany to ET (n= 189) or ET+B (n=191), 342 patients<br />

received letrozole and 38 fulvestrant. Baseline characteristics were<br />

well balanced. Median age was 65 years (38-86) and 72% of patients<br />

had ECOG PS 0. Twelve patients (4%) entered the trial with locally<br />

advanced disease, 65% had measurable lesions, 63% had bone<br />

and 45% visceral metastasis. 76% of patients had both hormone<br />

receptor positive. 44% had adjuvant chemotherapy and 51% adjuvant<br />

endocrine therapy from which 36% of patients received adjuvant AI.<br />

Full safety data will be presented at ESMO this year. The main side<br />

effects (any grade, per patient, ET+B vs ET) were anemia 76% vs<br />

44%, p


December 4-8, 2012 <strong>Abstract</strong>s: General Session 1<br />

χ2: 7.97, p=0.005) versus RS (LR-χ2: 0.51, p=0.5) and IHC4 (LR-χ2:<br />

1.63, p=0.2). Similar comparative results of BCI, RS and IHC4 for<br />

late recurrence were observed for the other clinical endpoints.<br />

Discussion: This is the first large-scale clinical study to compare<br />

several multi-gene signatures for their prognostic strength to quantify<br />

late residual risk of recurrence after endocrine therapy. BCI, unlike<br />

IHC4 and RS, is a significant prognostic factor for late recurrence and<br />

enables the assessment of individual recurrence risk for ER+ patients<br />

recurrence-free after 5 years of endocrine therapy.<br />

S1-10<br />

Association between the 21-gene recurrence score (RS) and benefit<br />

from adjuvant paclitaxel (Pac) in node-positive (N+), ER-positive<br />

breast cancer patients (pts): Results from NSABP B-28<br />

Mamounas EP, Tang G, Paik S, Baehner FL, Liu Q, Jeong J-H, Kim<br />

S-R, Butler SM, Jamshidian F, Cherbavaz DB, Sing AP, Shak S,<br />

Julian TB, Lembersky BC, Wickerham DL, Costantino JP, Wolmark<br />

N. National Surgical Adjuvant <strong>Breast</strong> and Bowel Project Operations<br />

and Biostatistical Centers; Aultman Hospital; Graduate School<br />

of Public Health, University of Pittsburgh; Genomic Health, Inc.;<br />

Allegheny <strong>Cancer</strong> Center at Allegheny General Hospital; University<br />

of Pittsburgh <strong>Cancer</strong> Institute<br />

Background: The RS result predicts outcome in N- and N+, ER+<br />

pts treated with adjuvant endocrine therapy. RS also predicts benefit<br />

from adjuvant chemotherapy (CT) and pts with a high RS result<br />

receive most of the benefit. We evaluated the association between<br />

RS and paclitaxel (Pac) benefit in N+, ER+ pts from NSABP B-28.<br />

Methods: B-28 compared 4 cycles of doxorubicin/cyclophosphamide<br />

(AC) vs. AC followed by Pac x 4 (AC→Pac). Pts ≥50 yrs and those


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

the breast and lymph nodes (ypT0ypN0 or ypT0/isypN0) was better<br />

associated with improved EFS and OS compared to eradication of<br />

tumor from the breast alone (ypT0/is). Patients who achieved a pCR<br />

(ypT0/isypN0) had an improved EFS (HR=0.48) and OS (HR=0.36)<br />

compared to those who did not. pCR was uncommon in patients with<br />

low-grade hormone receptor-positive (HR+) tumors (7%) and more<br />

common in the following tumor subtypes: high-grade HR+ (16%),<br />

triple negative (34%), HR+ /HER2+ (30%), and hormone receptornegative<br />

(HR-)/HER2+ (50%). Patients with more aggressive tumor<br />

subtypes who achieved pCR had greater EFS compared to patients<br />

who did not achieve pCR as follows: HR+ high grade (HR=0.27),<br />

HR+ /HER2+ (HR=0.58), HR- /HER2+ (HR=0.25), and triple<br />

negative (HR=0.24). A trial level analysis on the relationship between<br />

pCR effect size and EFS did not show a correlation. Conclusions:<br />

Individual patients who attain a pCR, defined as either ypT0ypN0 or<br />

ypT0/isypN0, have a more favorable long-term outcome. The data<br />

show comparable EFS or OS regardless of the presence or absence<br />

of DCIS. For consistency, a standard pCR definition (ypT0ypN0 or<br />

ypT0/isypN0) should be used in future trials. Impact of pCR effect<br />

is limited to patients with HR+/grade 3, HR-/HER2-, and HER2+<br />

tumors. This meta-analysis did not establish the magnitude of increase<br />

in pCR rate needed to predict the superiority of one regimen over<br />

another in terms of EFS or OS. This may be due to low pCR rates<br />

and the heterogeneity of the patient population included in this<br />

meta-analysis. The absolute magnitude of improvement in pCR<br />

rate needed to impact long-term outcome may be greater than the<br />

observed difference in these trials and may vary according to breast<br />

cancer subtype.<br />

S2-1<br />

The role of sentinel lymph node surgery in patients presenting<br />

with node positive breast cancer (T0-T4, N1-2) who receive<br />

neoadjuvant chemotherapy – results from the ACOSOG Z1071<br />

trial<br />

Boughey JC, Suman VJ, Mittendorf EA, Ahrendt GM, Wilke LG,<br />

Taback B, Leitch AM, Flippo-Morton TS, Byrd DR, Ollila DW,<br />

Julian TB, McLaughlin SA, McCall L, Symmans WF, Le-Petross HT,<br />

Haffty BG, Buchholz TA, Hunt KK. Mayo Clinic, Rochester, MN; MD<br />

Anderson <strong>Cancer</strong> Center, Houston, TX; Magee-Womens Surgical<br />

Associates, Pittsburgh, PA; University of Wisconsin-Madison, WI;<br />

Columbia University Medical Center, New York, NY; University<br />

of Texas Southwestern Medical Center, Dallas, TX; Carolinas<br />

Medical Center, Charlotte, NC; University of Washington Medical<br />

Center, Seattle, WA; University of North Carolina - Chapel Hill,<br />

NC; Allegheny General Hospital, Pittsburgh, PA; Mayo Clinic,<br />

Jacksonville, FL; Duke University Medical Center, Durham, NC;<br />

The <strong>Cancer</strong> Institute of New Jersey, New Brunswick, NJ<br />

Background:<br />

The utility of sentinel lymph node (SLN) surgery after neoadjuvant<br />

chemotherapy (NAC) in patients presenting with node-positive<br />

breast cancer has not been determined. The American College of<br />

Surgeons Oncology Group (ACOSOG) Z0171 trial was designed<br />

to evaluate SLN surgery after NAC in women presenting with node<br />

positive disease.<br />

Methods:<br />

ACOSOG Z1071 enrolled women with clinical T0-4, N1-2, M0 breast<br />

cancer receiving NAC. At the time of surgery, all patients were to<br />

undergo SLN surgery followed by axillary lymph node dissection<br />

(ALND). The primary endpoint was false negative rate (FNR) in<br />

women with cN1 disease with 2 or more SLNs reviewed. Positive<br />

SLNs were defined as metastases >0.2mm on H&E. The protocol<br />

encouraged dual tracer technique. A Bayesian study design with a<br />

non-informative prior was chosen to assess whether the probability<br />

that the SLN surgery FNR is greater than 10%.<br />

Results:<br />

From July 2009 to July 2011, 756 patients were enrolled from 136<br />

institutions. Fifteen women were ineligible and 33 withdrew. Of 708<br />

evaluable pts (668 cN1, 40 cN2), 643 had a SLN identified and an<br />

ALND (607 cN1, with indeterminate SLN results in 2); 52 pts (48<br />

cN1) had no SLN identified and had ALND; 11 underwent ALND<br />

only (all cN1), and 2 pts had SLN only (both cN1).<br />

In patients with SLN and ALND, the SLN identification rate was<br />

92.5% (92.7% in cN1, 90% in cN2). SLN correctly identified nodal<br />

status in 84% of the 695 pts [258 of pathologically node negative<br />

and 327 of pathologically node positive; cN1: 83.8% (549/655),<br />

cN2: 90.0% (36/40)].<br />

Of the 643 pts with a SLN identified there was a complete pathologic<br />

response in 40.3% (40.3 % for cN1 and 50% for cN2). Of the pts<br />

with a positive SLN, the SLN was the only site of disease in 40%.<br />

For pts with cN1 disease with 2+ SLNs identified with residual nodal<br />

disease, the SLN FNR was 12.8%. In pts with dual tracer technique<br />

the FNR was 11.1%. There were no FN results among pts with cN2<br />

disease with 2+ SLNs reviewed.<br />

Of the 40 pts with a false negative SLN of the 528 cN1 patients with<br />

2+ SLNs examined, the number of positive nodes at ALND was 1<br />

(50.0%); 2 (25%); 3 (10.0%) and 4-9 (15.0%).<br />

FN rates in pts with at least 2 SLNs examined<br />

N1 pts<br />

N1 & N2 pts<br />

FN rate 12.8% 12.2%<br />

Tracer<br />

Blue dye only 2/11 (18.8%) 2/13 (15.4%)<br />

Radiolabeled colloid only 10/50 (20.0%) 10/51 (19.6%)<br />

Both blue and radiolabeled<br />

colloid<br />

28/252 (11.1%) 28/263 (10.7%)<br />

Number of SLN reviewed<br />

2 19/91 (20.9%) 19/97 (19.6%)<br />

3 7/80 (8.8%) 7/84 (8.3%)<br />

4+ 14/142 (9.9%) 14/146 (9.6%)<br />

Clinical T stage<br />

T0/T1 6/40 (15.0%) 6/40 (15.0%)<br />

T2 27/187 (14.4%) 27/192 (14.1%)<br />

T3 6/75 (8.0%) 6/81(7.4%)<br />

T4 1/11 (9.1%) 1/14 (7.1%)<br />

Conclusions:<br />

NAC resulted in eradication of lymph node disease in 40% of node<br />

positive breast cancer patients. SLN surgery after NAC in node<br />

positive breast cancer pts correctly identified nodal status in 84% of<br />

all patients and was associated with a FNR of 12.8%.<br />

The FNR of SLN is higher than the prespecified study endpoint of<br />

10%. Further analysis of factors associated with FNR such as clinical<br />

response, histological findings and axillary ultrasound findings is<br />

warranted prior to widespread use of SLN in these patients.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

94s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: General Session 2<br />

S2-2<br />

Sentinel Lymph Node Biopsy Before or After Neoadjuvant<br />

Chemotherapy - Final Results from the Prospective German,<br />

multiinstitutional SENTINA-Trial -<br />

Kuehn T, Bauerfeind IGP, Fehm T, Fleige B, Helms G, Lebeau A,<br />

Liedtke C, von Minckwitz G, Nekljudova V, Schrenk P, Staebler A,<br />

Untch M. Klinikum Esslingen, Esslingen, Baden Wuerttemberg,<br />

Germany; Klinikum Landshut, Landshut, Bayern, Germany;<br />

Universitaetsfrauenklinik Tuebingen, Baden Wuerttemberg, Germany;<br />

Helios Klinikum Berlin-Buch, Berlin, Germany; University Medical<br />

Center Hamburg-Eppendorf, Hamburg, Germany; University<br />

Medical Center Muenster, Nordrhein-Westfalen, Germany; German<br />

<strong>Breast</strong> Group, Neu-Isenburg, Hessen, Germany; AKH-LFKK Linz,<br />

Austria; University Medical Center Tuebingen, Baden Wuerttemberg,<br />

Germany<br />

Background: The optimal timing for sentinel lymph node biopsy<br />

(SLNB) in the neoadjuvant setting is still unclear. Evidence for both,<br />

feasibility (detection rate, DR) and accuracy (false negative rate,<br />

FNR) of SLNB before and after primary systemic treatment (PST) is<br />

restricted to monocentric and/or retrospective trials. The success rates<br />

for SLNB are particularly unclear for patients who are downstaged<br />

from a cN1 to a ycN0 status.<br />

Material and Methods: The German SENTINA (SENTInel<br />

NeoAdjuvant) trial is a 4-arm prospective multicenter cohort<br />

study designed to evaluate a specific algorithm for the timing of a<br />

standardized SLNB procedure in patients, who undergo PST and<br />

to provide reliable data for the DR and FNR in different settings.<br />

Patients were categorized into four treatment arms according to the<br />

clinical axillary staging before and after chemotherapy. Patients with<br />

a cN0 status underwent SLNB prior to PST and were categorized<br />

as arm A and B: If the SLN was histologically negative no further<br />

axillary surgery was performed after PST (arm A), whereas in case<br />

of histologically positive SLN status, a second SLNB and axillary<br />

dissection (AD) was performed after PST (arm B). Patients with a<br />

cN1 status prior to PST underwent no axillary surgery prior to PST<br />

and were stratified as arm C and D: If patients converted to cN0 after<br />

PST, SLNB and AD were performed (arm C); patients presenting with<br />

cN1 status after PST underwent classical AD (arm D).<br />

Results: 1737 eligible patients from 103 institutions entered the trial.<br />

From these 662 patients were classified as arm A, 360 pts as arm B,<br />

592 pts as arm C and 123 pts as arm D.<br />

The DR for SLNB was 1013/1022 (99.1%) before PST (arms A and<br />

B), 474/592 (80.1%) in Arm C (after PST), and 219/360 (60.8%) in<br />

arm B (after prior SLNB and PST) (p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

S3-1<br />

Neoadjuvant Chemotherapy in the very young 35 years of age<br />

or younger<br />

Loibl S, Jackisch C, Gade S, Untch M, Paepke S, Kuemmel S,<br />

Schneeweiss A, Jackisch C, Huober J, Hilfrich J, Hanusch C, Gerber<br />

B, Eidtmann H, Denkert C, Costa S-D, Blohmer J-U, Nekljudova V,<br />

Mehta K, von Minckwitz G. German <strong>Breast</strong> Group, Neu-Isenburg;<br />

Klinikum Offenbach; Helios Kliniken Berlin; University Muenchen;<br />

Kliniken Essen Mitte; University Heidelberg; Uni Duesseldorf;<br />

Eilenriedeklinik Duessldorf; Rotkreuzklinikum Muenchen; University<br />

Rostock; University Kiel; Charite Berlin; University Magdeburg;<br />

<strong>San</strong>kt Gertrauden Berlin<br />

Background: In young women the course of breast cancer (BC) tends<br />

to be more aggressive. In several trials young age at diagnosis was<br />

an independent predictive factor for pathological complete response<br />

(pCR) after neoadjuvant chemotherapy. Here we investigate especially<br />

the rare entity of very young women at age 35 years or younger.<br />

Methods: 8949 patients from 8 neoadjuvant German studies with<br />

operable or locally advanced, non-metastatic breast cancer and<br />

follow-up were included (for details see von Minckwitz G et al,<br />

BCRT 2010 and NEJM 2012). A subgroup of 704 patients of age<br />

35 years or younger was analyzed. All patients with endocrine<br />

responsive disease received adjuvant endocrine therapy according to<br />

institutional standard. We compared pathological complete remission<br />

rate (pCR) defined as ypT0, ypN0 and disease free survival rate (DFS)<br />

of this very young group with older patients in total and in different<br />

histopathological subgroups (as defined previously by von Minckwitz<br />

J Clin Oncol 2012).<br />

Results: From 8949/6561 had known ER, PR, HER2 and grading.<br />

There were less Luminal A and more TNBC in the very young women<br />

compared to the one >35 years of age: Luminal A: 131 (21%) vs.<br />

2251 (27%); Luminal B HER2-: 50 (8%) vs. 783 (10%); Luminal B<br />

HER2+: 103 (17%) vs. 989 (14%); HER2+/HR-: 72 (11%) vs. 739<br />

(10%); TNBC: 164 (26%) vs. 1415 (19%).<br />

The pCR rate was significantly higher in the very young than in the<br />

group older than 35 years (23.6% vs. 15.7.%; p


December 4-8, 2012 <strong>Abstract</strong>s: General Session 3<br />

S3-3<br />

The UK TACT2 Trial: comparison of standard vs accelerated<br />

epirubicin in patients requiring chemotherapy for early breast<br />

cancer (EBC) (CRUK/05/019)<br />

Cameron D, Barrett-Lee P, Canney P, Banerji J, Bartlett J, Bloomfield<br />

D, Bowden S, Brunt M, Earl H, Ellis P, Fletcher M, Morden JP,<br />

Robinson A, Sergenson N, Stein R, Velikova G, Verrill M, Wardley<br />

A, Coleman R, Bliss JM. Edinburgh <strong>Cancer</strong> Research Centre,<br />

University of Edinburgh, United Kingdom; Velindre NHS Trust<br />

<strong>Cancer</strong> Centre, Cardiff, United Kingdom; Beatson West of Scotland<br />

<strong>Cancer</strong> Centre, Glasgow; The Institute of <strong>Cancer</strong> Research, Sutton,<br />

United Kingdom; Ontario Institute for <strong>Cancer</strong> Research, Toronto,<br />

Canada; Brighton & Sussex University Hospitals, Brighton, United<br />

Kingdom; University of Birmingham, United Kingdom; University<br />

Hospital of North Staffordshire, Stoke-on-Trent, United Kingdom;<br />

University of Cambridge and NIHR Cambridge Biomedical Research<br />

Centre, Cambridge, United Kingdom; Guys Hospital & King<br />

College, London, United Kingdom; Leeds Institute of Molecular<br />

Medicine, Leeds, United Kingdom; Southend University Hospital,<br />

Southend, United Kingdom; Information Services Division, NHS<br />

National Services Scotland, Edinburgh, United Kingdom; UCL<br />

Hospitals, London, United Kingdom; Northern Centre for <strong>Cancer</strong><br />

Care, Newcastle upon Tyne, United Kingdom; The Christie Hospital,<br />

Manchester, United Kingdom; Weston Park Hospital, Sheffield,<br />

United Kingdom<br />

Introduction: TACT2, a multicentre randomized phase III trial in<br />

patients with node +ve or high risk node -ve invasive EBC with<br />

E-CMF as control (Poole NEJM 2006), tests two hypotheses in a 2x2<br />

factorial design: A) accelerated epirubicin (aE) improves outcomes<br />

compared to standard epirubicin (E) (CALGB9741 Citron JCO 2003);<br />

and B) capecitabine (X) gives similar efficacy but preferential sideeffect<br />

profile to CMF. Here we focus on the impact of accelerating<br />

epirubicin treatment on patient outcome. Tolerability and toxicity of<br />

this regimen have already been presented (Cameron 2010, SABCS);<br />

33/4391 pts did not start chemotherapy. 4261 (97%) completed the<br />

initial 4 cycles (E 2143 (96%), aE 2118 (98%)) with RDI >85% for<br />

E 2061 (93%) and aE 1985 (91%). Serious CTCAE toxicity was less<br />

common with aE compared with E, however patients report poorer<br />

global QL, fatigue & role function which does not appear to be<br />

explained by impact on daily activities due to individual side-effects.<br />

Patients & Methods: Between December 2005 and December 2008,<br />

4391 patients (4371 women, 20 men) were randomized to receive<br />

either E (100mg/m 2 x 4) q3wk or aE (100mg/m 2 x 4 plus pegfilgrastim,<br />

6mg d2) q2wk followed by classical CMF q4wk x 4 or X (2500mg/<br />

m 2 /day x 14) q3wk x 4. Other adjuvant treatment was delivered<br />

according to local practice (HER2+ve – sequential trastuzumab<br />

for 1 year from June 2006; ER/PgR+ve - endocrine therapy for 5<br />

years). Tumor blocks were collected prospectively to enable central<br />

biomarker testing. Median age of patients was 52 years (IQR 46-59).<br />

53% patients had node +ve disease; 59% had tumors >2cm; 57% were<br />

grade 3. According to local assessment, of 4366 patients where both<br />

ER and HER2 status were known, 60% had ER+ve/HER2-ve tumors,<br />

19% HER2+ve, 21% ER-ve/HER2-ve. Primary endpoint was time to<br />

recurrence (TTR), defined as time to loco-regional or distant relapse,<br />

or breast cancer-death. Secondary endpoints included disease-free<br />

survival (DFS), overall survival (OS), tolerability of regimens and<br />

quality of life. TACT2 has over 90% power to detect a clinically<br />

meaningful 4% difference in TTR between standard and accelerated<br />

epirubicin schedules (from 80% to 84% at 5 years). Survival estimates<br />

will be compared using stratified log-rank tests and Cox regression.<br />

Results: At 1 st June 2012, median follow up was 49 months<br />

(interquartile range: 46-60) with 553 TTR events reported. The<br />

Independent Data Monitoring Committee consider these data<br />

sufficiently mature for presentation of analyses relating to the<br />

accelerated epirubicin hypothesis. A further data snapshot, after query<br />

resolution, for the main analysis will be taken in Q3 2012. Data to be<br />

presented, by treatment group, include TTR, DFS, OS, late toxicity<br />

and pre-planned subgroup analyses according to ER, PgR and HER2<br />

status and Ki67 levels.<br />

Discussion: TACT2 will provide a definitive analysis to confirm or<br />

refute benefits previously reported for accelerating anthracycline<br />

chemotherapy in early breast cancer including, via pre-planned<br />

subgroup analyses, exploration of the potential benefits in phenotypic<br />

subtypes.<br />

S3-4<br />

Ten year follow-up analysis of intense dose-dense adjuvant<br />

ETC (epirubicin (E), paclitaxel (T) and cyclophosphamide<br />

(C) ) confirms superior DFS and OS benefit in comparison to<br />

conventional dosed chemotherapy in high-risk breast cancer<br />

patients with ≥ 4 positive lymph nodes.<br />

Moebus V, Schneeweiss A, du Bois A, Lueck H-J, Eustermann H,<br />

Kuhn W, Kurbacher C, Nitz U, Kreienberg R, Jackisch C, Huober<br />

J, Thomssen C, Untch M. Klinikum Frankfurt Höchst, Frankfurt,<br />

Germany; University of Heidelberg, Germany; Klinikum Essen Mitte,<br />

Essen, Germany; Gynäkologisch-Onkologische Praxis, Hannover,<br />

Germany; WiSP Research Institute, Langenfeld, Germany; University<br />

of Bonn, Germany; Medizinisches Zentrum Bonn-Friedensplatz, Bonn,<br />

Germany; Ev. Krankenhaus Bethesda, Mönchengladbach, Germany;<br />

University of Ulm, Germany; Klinikum Offenbach, Offenbach,<br />

Germany; University of Duesseldorf, Germany; University of Halle,<br />

Germany; Helios Klinikum Berlin-Buch, Berlin, Germany<br />

Background: The 5-year analysis of adjuvant chemotherapy with<br />

intense dose-dense (IDD) ETC had shown a significant improved<br />

DFS (HR 0.72; p < 0.001) and OS (HR 0.76; p=0.29) in comparison<br />

with conventional dosed chemotherapy (J Clin Oncol 28: 2874-2880,<br />

2010). In contrast to other dose-dense trials the ETC regimen is dosedense<br />

and dose-intensified. Long-term results are essential to evaluate<br />

the impact of dose-dense chemotherapy in the adjuvant treatment of<br />

breast cancer patients (pts). We now report the final analysis of DFS,<br />

OS, and long-term safety including the application of epoetin alfa<br />

after 10 years of follow-up.<br />

Patients and Methods:<br />

A multi-center phase-III trial of the German AGO <strong>Breast</strong> Study<br />

Group recruited 1284 pts from 12/98 until 4/03. Pts below 65<br />

years of age were eligible if at least 4 axillary lymph nodes were<br />

infiltrated. In the experimental arm, pts were assigned to receive<br />

three courses each of epirubicin (150 mg/m 2 ), paclitaxel (225 mg/<br />

m 2 ) and cyclophosphamide (2500 mg/m 2 ) at 2-week intervals (q2w)<br />

(ETC) with G-CSF support (5µg/kg/SC day 3-10). In the standard<br />

arm 4 courses of conventional dosed epirubicin/cyclophosphamide<br />

(90/600 mg/m 2 ) followed by 4 courses of paclitaxel (175 mg/<br />

m 2 ) were given (EC→T). All cycles were administered in 3-week<br />

intervals without growth factor support. A second randomization ±<br />

epoetin alfa was performed in the IDD-ETC arm only (150IU/kg/sc<br />

three times weekly) to reduce the number of red blood cells (RBC’s)<br />

transfusion and to evaluate the impact of epoetin alfa on DFS and<br />

OS in the adjuvant setting.<br />

Results:<br />

58% and 42% of the pts presented with 4-9 and ≥ 10 positive nodes<br />

with a median number of 8 involved nodes. The median age was 51<br />

years and median follow-up was 122 months. We observed 604 DFS<br />

events (282 with IDD ETC; 322 with EC→T) (p=0.00014, one-sided;<br />

www.aacrjournals.org 97s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

HR 0.74; 95% CI, 0.63 to 0.87). IDD ETC improved DFS irrespective<br />

of nodal status, HER2 and ER status. 446 pts. have died (201 events<br />

in the IDD ETC arm vs. 245 events in the standard arm). 10 year OS<br />

rates were 69% with IDD ETC and 59% with EC→ T (p=0.0007;<br />

two-sided; HR, 0.72; 95% CI, 0.60-0.87). Nine cases of acute myeloid<br />

leukemia or myelodysplastic syndrome occurred in the IDD ETC arm<br />

vs. two cases in the standard arm. 28% of pts in the IDD ETC arm<br />

vs. 13% in the IDD ETC arm plus epoetin alfa (p


December 4-8, 2012 <strong>Abstract</strong>s: General Session 3<br />

common. Sporadic growth factor receptor amplifications occurred<br />

in EGFR, KIT, PDGFRA, PDGFRB, MET, FGFR1, FGFR2, and<br />

IGF1R. NGS identified 7 patients with ERBB2 gene amplification<br />

in the RD which was confirmed by FISH in both the pre- and posttreatment<br />

tissue, suggesting NGS could assist in the identification of<br />

ERBB2-overexpressing tumors misclassified at the time of diagnosis.<br />

In general, the gene amplifications identified by NGS corresponded<br />

to enhanced gene expression levels. Amplifications of MYC were<br />

independently associated with poor recurrence-free survival (RFS)<br />

and overall survival (OS). An interaction effect on survival was<br />

observed between MEK activation (assayed by a gene expression<br />

signature) and MYC amplification, suggesting cooperation between<br />

these pathways. Alterations in DNA repair also identified a subgroup<br />

with poor RFS and OS. In contrast, high post-NAC Ki67 score did<br />

not predict poor RFS or OS in this predominantly TNBC cohort.<br />

Conclusions: The diversity of lesions in residual TNBCs after NAC<br />

underscores the need for powerful and broad molecular approaches<br />

to identify actionable molecular alterations and, in turn, better inform<br />

personalized therapy of this aggressive disease. Incorporation of this<br />

platform into clinical studies and eventually standards of care should<br />

aid in the prioritization of patients with RD after NAC into rational<br />

adjuvant studies.<br />

S3-7<br />

Treatment with Histone Deacetylase Inhibitors Creates<br />

‘BRCAness’ and Sensitizes Human Triple Negative <strong>Breast</strong> <strong>Cancer</strong><br />

Cells to PARP Inhibitors and Cisplatin<br />

Bhalla KN, Rao R, Sharma P, Das Gupta S, Chauhan L, Stecklein<br />

S, Fiskus W. University of Kansas <strong>Cancer</strong> Center, Kansas City, KS<br />

DNA damage induces DNA damage response (DDR), which regulates<br />

cell cycle transit, DNA repair and apoptosis. During DDR, stability<br />

and activity of ATR and CHK1 is essential for the cell cycle arrest<br />

and subsequent DNA repair through homologous recombination<br />

(HR) in which BRCA1 protein is involved. We have previously<br />

reported that ATR, CHK1 and BRCA1 are hsp90 client proteins<br />

and treatment with hsp90 inhibitor sensitizes cancer cells to DNA<br />

damage. We have also previously shown that treatment with pan-<br />

HDAC inhibitors, including vorinostat (VS) and panobinostat (PS),<br />

induces hyperacetylation of hsp90, thereby inhibiting its chaperone<br />

function. In the present studies we determined that treatment with VS<br />

or PS induced hyper-acetylation and inhibition of chaperone function<br />

of nuclear hsp90, leading to proteasomal degradation and depletion<br />

of ATR, CHK1 and BRCA1. This led to inhibition of DDR and DNA<br />

repair following ionizing radiation (IR) through destabilization of<br />

ATR-CHK1 and BRCA1 proteins. Based on this, we hypothesized that<br />

treatment with VS or PS would create ‘BRCAness’ in breast cancer<br />

cells. Specific siRNA-mediated knockdown of HDAC3 but not of<br />

HDAC1 or HDAC2 also induced hyper-acetylation of the nuclear<br />

hsp90 and depletion of ATR and CHK1, indicating that among the<br />

class I HDACs, HDAC3 is the deacetylase for the nuclear hsp90. We<br />

next determined whether, by depleting DDR proteins and BRCA1<br />

and inducing ‘BRCAness’, treatment with VS would sensitize the<br />

triple negative breast cancer (TNBC) SUM59PT, MB-231 and<br />

HCC1937 cells to the PARP inhibitor ABT888 (veliparib). Indeed,<br />

combined treatment with VS or PS with ABT888 (10 to 20 µM) for<br />

48 hours synergistically induced apoptosis (Combination indices by<br />

isobologram analysis being < 1.0) of TNBC cells with (HCC1937)<br />

or without BRCA1 mutation (MB-231 and SUM159PT cells). As<br />

compared to treatment with each agent alone, co-treatment with VS<br />

and ABT888 also induced significantly more DNA strand breaks,<br />

as demonstrated by higher γ-H2AX levels. Combined treatment<br />

also induced markedly greater tail moment, as determined by the<br />

Comet assay that evaluates the SYBR green-stained DNA tails by<br />

fluorescent microscopy. Co-treatment with VS and ABT888 also<br />

induced more BH3 domain-only pro-death protein BIM, while<br />

knock-down of BIM by shRNA significantly reduced the apoptosis<br />

induced by co-treatment with VS and ABT888. It is noteworthy that<br />

treatment with VS also sensitized the TNBC cells to cisplatin (2.0<br />

to 10 µM)-induced apoptosis. Moreover, co-treatment with VS and<br />

ciplatin synergistically induced apoptosis of TNBC cells (CIs < 1.0).<br />

These findings indicate that treatment with pan-HDAC inhibitors VS<br />

or PS creates BRCAness, and in combination with a PARP inhibitor<br />

or cisplatin synergistically induces apoptosis in human TNBC cells.<br />

These findings support a compelling rationale to confirm whether<br />

the combination of VS or PS with ABT888 and cisplatin would be a<br />

highly active treatment in the in vivo models of human TNBC cells,<br />

irrespective of their expression of mutant BRCA1.<br />

S3-8<br />

Identification of Novel Synthetic-Lethal Targets for MYC-Driven<br />

Triple-Negative <strong>Breast</strong> <strong>Cancer</strong><br />

Goga A, Samson S, Horiuchi D. UCSF, <strong>San</strong> Francisco, CA; UCSF<br />

Currently, the greatest clinical challenge in treating breast cancer<br />

occurs in those patients whose tumors lack expression of the estrogen<br />

and progesterone receptors and the HER2 oncoprotein. No targeted<br />

agents currently exist against this aggressive type of receptor ‘triple<br />

negative’ breast cancer that disproportionately affects young African<br />

American women and also a substantial number of white American<br />

women. We have discovered that triple-negative tumors frequently<br />

express high levels of the MYC proto-oncogene. We sought to identify<br />

new “synthetic-lethal” strategies to selectively kill triple-negative<br />

breast tumors with MYC overexpression. Synthetic lethality arises<br />

when a combination of mutations in two or more genes leads to<br />

cell death, whereas a mutation in only one of these genes has little<br />

effect. Using this strategy, we can take advantage of the elevated<br />

MYC signaling in triple-negative tumors to selectively kill them,<br />

while sparing normal tissues in which MYC is expressed at much<br />

lower levels.<br />

RESULTS: We performed a shRNA synthetic-lethal screen to identify<br />

new molecules, such as cell cycle kinases, which when inhibited can<br />

preferentially kill triple-negative breast tumor cells. The unbiased<br />

shRNA-based screening approach was performed in human mammary<br />

epithelial cells (HMECs) that have inducible MYC over-expression.<br />

A high-throughput screen of ∼2000 shRNAs, that target the human<br />

kinome (∼ 600 kinases) was performed, and identified 13 kinases<br />

whose inhibition by >1 shRNAs gave rise to >50% inhibition of<br />

cell growth. Two of the kinases identified in our screen, ARK5 and<br />

GSK3A, were also recently identified by other studies and shown to<br />

have a synthetic-lethal interaction with MYC. These results validated<br />

our ability to identify synthetic-lethal targets. We are currently<br />

characterizing and validating the 11 novel targets identified in this<br />

screen, using human cancer cell lines as well as mouse cancer models<br />

to determine the impact of inhibiting these targets on triple-negative<br />

breast cancer development and proliferation. Additional syntheticlethal<br />

interactions with MYC over-expression in triple-negative breast<br />

cancers will be presented.<br />

www.aacrjournals.org 99s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

S4-1<br />

The UK START (Standardisation of <strong>Breast</strong> Radiotherapy) Trials:<br />

10-year follow-up results<br />

Haviland JS, Agrawal R, Aird E, Barrett J, Barrett-Lee P, Brown J,<br />

Dewar J, Dobbs J, Hopwood P, Hoskin P, Lawton P, Magee B, Mills<br />

J, Morgan D, Owen R, Simmons S, Sydenham M, Venables K, Bliss<br />

JM, Yarnold JR, On Behalf of the START Trialists. The Institute of<br />

<strong>Cancer</strong> Research, Sutton, United Kingdom; Shrewsbury and Telford<br />

Hospital NHS Trust, United Kingdom; Mount Vernon Hospital,<br />

Northwood, United Kingdom; Royal Berkshire NHS Foundation Trust,<br />

Reading, United Kingdom; Velindre Hospital NHS Trust, Cardiff,<br />

United Kingdom; previously University of Bristol, now Eli Lilly &<br />

Company, United Kingdom; Formerly Ninewells Hospital, Dundee,<br />

United Kingdom; Formerly Guys and St Thomas’ NHS Trust, London,<br />

United Kingdom; Nottingham City Hospital, United Kingdom;<br />

Formerly The Christie NHS Foundation Trust, Manchester, United<br />

Kingdom; Formerly Nottingham University Hospital NHS Trust,<br />

United Kingdom; Formerly Cheltenham General Hospital, United<br />

Kingdom; The Institute of <strong>Cancer</strong> Research and Royal Marsden NHS<br />

Foundation Trust, Sutton, United Kingdom<br />

Background International standard adjuvant radiotherapy regimens<br />

following primary surgery for early breast cancer have historically<br />

delivered a high total dose (50Gy) in 25 small daily doses (fractions)<br />

over 5 weeks, however randomised trials, including START, indicate<br />

that a lower total dose delivered in fewer, larger fractions (Fr) is likely<br />

to be at least as safe and effective (START Trialists’ Group, Lancet<br />

2008 & Lancet Oncol 2008). With patients remaining at risk of local<br />

relapse for many years, information on long-term outcomes is needed<br />

to provide confidence in clinical practice. Here, we report 10-year<br />

follow-up of the UK START Trials testing 13- and 15-Fr regimens<br />

in terms of local cancer control and late adverse effects.<br />

Methods Between 1999 and 2002, 4451 women with completely<br />

excised invasive breast cancer (T1-3, N0-1, M0) were randomised<br />

after primary surgery to comparisons of 50Gy in 25Fr over 5 weeks<br />

vs 41·6Gy or 39Gy in 13Fr over 5 weeks (START A), or 50Gy in<br />

25Fr over 5 weeks vs 40Gy in 15Fr over 3 weeks (START B). Women<br />

were eligible if aged over 18 years and did not have an immediate<br />

surgical reconstruction. Protocol-specified principal endpoints were<br />

local-regional (LR) tumour relapse and late normal tissue effects.<br />

Analysis was by intention to treat.<br />

Findings Median follow-up in survivors is now 9.3 years in START A<br />

and 9.9 years in START B, with 139 LR relapses in START A and 95<br />

in START B. In START A, the 10-year rate of LR relapse was 7.4%<br />

(95%CI 5.5-10.0) after 50Gy, 6.3% (95%CI 4.7-8.5) after 41·6Gy and<br />

8.8% (95%CI 6.7-11.4) after 39Gy. In START B, the 10-year rate of<br />

LR relapse was 5.5% (95%CI 4.2-7.2) after 50Gy and 4.3% (95%CI<br />

3.2-5.9) after 40Gy. Clinician assessments suggested lower 10-year<br />

rates of any moderate/marked late normal tissue effects after 39Gy<br />

(43.9%; 95%CI 39.3-48.7) and similar rates after 41.6Gy (49.5%;<br />

95%CI 44.9-54.3) compared with 50Gy (50.4%; 95%CI 45.8-55.3)<br />

in START A and lower rates after 40Gy in START B (37.9%; 95%CI<br />

34.5-41.5) compared with 50Gy (45.3%; 95%CI 41.7-49.0). From a<br />

planned meta-analysis of START A and the START pilot trial (Owen<br />

et al, Lancet Oncol 2006), the adjusted estimate of α/β value for<br />

tumour control was 3.5Gy (95% CI 1.2-5.7) and for late change in<br />

photographic breast appearance was 3.1Gy (95% CI 2.0-4.2).<br />

Interpretation Long-term follow-up confirms that breast cancer<br />

and the surrounding dose-limiting healthy tissues respond similarly<br />

to radiotherapy fraction size and thus that appropriately-dosed<br />

hypofractionated radiotherapy is safe and effective in treatment of<br />

patients with early breast cancer. 41·6Gy in 13Fr and 40Gy in 15Fr<br />

each appear comparable to 50Gy in 25Fr in terms of local-regional<br />

tumour control and late normal tissue effects. These results support<br />

the continued use of 40Gy in 15Fr as standard of care (UK NICE<br />

Guidance 2009) for women requiring adjuvant radiotherapy for early<br />

breast cancer.<br />

S4-2<br />

Targeted intraoperative radiotherapy for early breast cancer:<br />

TARGIT-A trial- updated analysis of local recurrence and first<br />

analysis of survival<br />

Vaidya JS, Wenz F, Bulsara M, Joseph D, Tobias JS, Keshtgar M,<br />

Flyger H, Massarut S, Alvarado M, Saunders C, Eiermann W,<br />

Metaxas M, Sperk E, Sutterlin M, Brown D, Esserman L, Roncadin<br />

M, Thompson A, Dewar JA, Holtveg H, Pigorsch S, Falzon M, Harris<br />

E, Matthews A, Brew-Graves C, Potyka I, Corica T, Williams NR,<br />

Baum M. University College London, London, United Kingdom;<br />

University Medical Centre Mannheim, University of Heidelberg,<br />

Heidelberg, Germany; University of Notre Dame, Fremantle,<br />

Australia; Sir Charles Gairdner Hospital, Perth, Australia; University<br />

College Hospital, London, United Kingdom; Royal Free Hospital,<br />

London, United Kingdom; University of Copenhagen, Copenhagen,<br />

Denmark; Centro di Riferimento Oncologia, Aviano, Italy; University<br />

of California, <strong>San</strong> Francisco, CA; University of Western Australia,<br />

Perth, WA, Australia; Red Cross Hospital, Munich, Germany;<br />

Ninewells Hospital, Dundee, United Kingdom; Technical University<br />

of Munich, Munich, Germany; University College London Hospitals,<br />

London, United Kingdom; National <strong>Cancer</strong> Research Institute and<br />

Independent <strong>Cancer</strong> Patients’ Voice, United Kingdom; Moffit <strong>Cancer</strong><br />

Centre<br />

Background TARGIT-A, an international phase 3 randomised trial<br />

(Lancet 2010;376:91-102) compared outcomes in patients undergoing<br />

breast conserving surgery followed by either whole breast external<br />

beam radiotherapy (EBRT) over several weeks, or a risk-adaptive<br />

approach using single dose targeted intra-operative radiotherapy<br />

(TARGIT). Risk-adaptive approach meant that if the final pathology<br />

report demonstrated unpredicted pre-specified adverse features, then<br />

EBRT was to be added to TARGIT.<br />

Method 3451 women aged 45 years or older with invasive ductal<br />

carcinoma were enrolled from 33 centres in 10 countries between<br />

2000 and 2012.<br />

Randomisation to TARGIT or EBRT arm was done either before<br />

lumpectomy (pre-pathology) or after lumpectomy (postpathology).<br />

If allocated to TARGIT, patients in the pre-pathology group received<br />

it immediately after surgical excision under the same anaesthesia;<br />

patients in the post-pathology group received it as a subsequent<br />

procedure. We pre-specified that analysis would be performed<br />

overall as the primary analysis and for these groups separately as<br />

a secondary analysis. The primary outcome was ipsilateral within<br />

breast recurrence (IBR) with an absolute non-inferiority margin of<br />

2.5% at 5 years and secondary outcome was survival. We performed<br />

exploratory analyses for loco-regional recurrence, ‘all recurrence’<br />

(ipsilateral or contralateral breast, axilla or distant), distant recurrence,<br />

and causes of death.<br />

Results 1721 patients were randomly allocated to receive TARGIT<br />

and 1730 to EBRT. 1010 patients have a minimum 4 years follow up<br />

and 611 patients have minimum 5 years follow up. Primary events<br />

have increased from 13 to 34 since 2010.<br />

For the primary outcome of ipsilateral breast recurrence, the absolute<br />

difference at 5-years was 2.0%, which was higher with TARGIT<br />

and reached the conventional levels of statistical significance<br />

(p=0.042), but was within the pre-specified non-inferiority margin;<br />

in prepathology the absolute difference in 5-year IBR was 1%; in<br />

postpathology it was 3.7%.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

100s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: General Session 4<br />

For the secondary outcome, there was a non-significant trend for<br />

improved overall survival with TARGIT (HR = 0.70(0.46-1.07)) due<br />

to fewer non-breast cancer deaths (17 vs. 35, HR 0.47 (0.26-0.84)).<br />

Cardiovascular deaths were 1 vs. 10 and deaths from cancers other<br />

than breast were 7 vs.16.<br />

Ipsilateral breast<br />

recurrenc(IBR)<br />

Events<br />

5-year cumulative risk (95%CI)<br />

TARGIT EBRT<br />

23 11<br />

HR (95% CI)<br />

3.3%(2.1-5.1) 1.3% (0.7-2.5) 2.07 (1.01-4.25)<br />

All recurrence (ipsilateral and<br />

contralateral breast, axilla 69 48<br />

and distant)<br />

8.2%(6.3-10.6) 5.7%(4.1-7.8) 1.44(0.99-2.08)<br />

Death 37 51<br />

3.9%(2.7-5.8) 5.3%(3.9-7.3) 0.70(0.46-1.07)<br />

Conclusion The risk-adapted approach using single dose TARGIT<br />

has a slightly higher local recurrence rate than EBRT for the<br />

primary endpoint of IBR, but was within the preset non-inferiority<br />

boundary, with the prepathology apparently performing better than<br />

the postpathology stratum. In addition there was a trend for improved<br />

overall survival in the TARGIT arm due to fewer non-breast cancer<br />

deaths.<br />

S4-3<br />

The EndoPredict score identifies late distant metastases in ER+/<br />

HER2- breast cancer patients<br />

Dubsky P, Brase JC, Fisch K, Jakesz R, Singer CF, Greil R, Dietze O,<br />

Weber KE, Petry C, Kronenwett R, Rudas M, Knauer M, Gnant M,<br />

Filipits M. Medical University Vienna, Austria; Sividon Diagnostics<br />

GmbH, Cologne, Germany; Paracelsus Private Medical University,<br />

Salzburg, Austria; Sisters of Charity Hospital, Linz, Austria; Medical<br />

University of Vienna, Austria<br />

Background:<br />

ER+/HER2- negative (ER+) breast cancers have a proclivity for late<br />

recurrence. A first step to an improved adjuvant treatment of late<br />

metastasis is to identify women at risk and understand the underlying<br />

biology. Several prognostic multigene tests have been developed for<br />

ER+ breast cancer patients. Some of these tests have been validated<br />

to predict early recurrence events. However, few gene expression<br />

assays have been shown to predict late metastases.<br />

Here, we assess whether the prognostic EndoPredict (EP) score,<br />

which incorporates both the expression levels of proliferative - and<br />

ESR1-related genes, can be used to identify late relapse events in<br />

ER+ breast cancer patients.<br />

Methods:<br />

Patients included in this study participated in the ABCSG-6<br />

(tamoxifen-only arm) or ABCSG-8 phase III adjuvant trial and<br />

received either tamoxifen for 5 years or tamoxifen for 2 years followed<br />

by anastrozole for 3 years. All 1702 ER+/HER2- breast cancer patients<br />

were retrospectively assigned into risk categories based on the EP and<br />

on common clinical parameters. The primary endpoint was distant<br />

metastasis. Ongoing collection of follow-up events allowed estimation<br />

of metastasis rates using the Kaplan–Meier method in an early and late<br />

time cohort: 0-5 years/early recurrence and 5-10 years/late recurrence.<br />

Results:<br />

49% of all patients were classified as low-risk according to the EP<br />

score. Kaplan Meier analysis demonstrated that the EP low-risk<br />

group had a significantly improved clinical outcome in the first (0-5<br />

years; p < 0.0001) and second time interval (5-10 years; p = 0.002).<br />

Nodal metastasis was also significantly associated with the clinical<br />

outcome in both time intervals, with node-positive tumors showing<br />

a considerably higher rate of late recurrence events. In contrast,<br />

Ki67 levels and grading were not significantly associated with late<br />

metastasis. Multivariate analysis showed that EP was an independent<br />

prognostic parameter after adjustment for age, grade, lymph node<br />

status, tumor size and Ki67 in both the first and second time interval.<br />

The EPclin – a combination of the EP and the clinical risk factors nodal<br />

status and tumor size – showed the best performance in predicting late<br />

relapse events. Exploratory analyses of proliferative - versus ESR-1<br />

related genes as contributors to early and late distant metastases show<br />

differential effects.<br />

Conclusions:<br />

The EndoPredict test identified a subgroup of patients that have a low<br />

likelihood of developing late metastases. The eight genes contained<br />

in the test provide complimentary prognostic information to clinicopathologic<br />

parameters. Both the expression of proliferative - and<br />

ESR1-related genes contributes to the underlying biology of late<br />

distant metastases.<br />

S4-4<br />

Independent validation of Genomic Grade in the BIG 1-98 study<br />

Sotiriou C, Ignatiadis M, Desmedt C, Azim Jr HA, Veys I, Larsimont<br />

D, Lyng M, Viale G, Leyland-Jones B, Ditzel H, Giobbie-Hurder<br />

A, Regan M, Piccart M, Michiels S. Universite Libre de Bruxelles;<br />

Institut Jules Bordet; University of Southern Denmark; University of<br />

Milan; Edith <strong>San</strong>ford Research; Harvard Medical School<br />

Background: We have reported that a 97-gene signature, the Genomic<br />

Grade Index (GGI) can separate histological grade (HG) 2 breast<br />

tumors into low vs high GGI tumors with different outcomes. We<br />

have also developed a quantitative reverse transcriptase polymerase<br />

chain reaction (qRT-PCR) tool including 6 reporter and 3 control<br />

genes that can reliably evaluate Genomic Grade (GG) in paraffin<br />

embedded tumors.<br />

Methods: We evaluated the qRT-PCR GG in women treated with either<br />

tamoxifen or letrozole monotherapy in the <strong>Breast</strong> International Group<br />

(BIG) 1-98 study with 8.1 year median follow-up. The association<br />

between continuous GG and distant recurrence-free interval (DRFI)<br />

was evaluated in Cox regression models stratified for the 2 vs 4<br />

arm randomization option, chemotherapy and hormonotherapy use,<br />

with and without adjustment for clinicopathological characteristics.<br />

Estrogen, progesterone receptor (ER, PR), Ki67 and HG were<br />

centrally reviewed. The clinicopathological model included age and<br />

log2 tumor size, ER and PR as continuous variables, nodal status (N0<br />

vs 1-3 vs >3), HG (1 vs 2 vs 3) and HER2 (negative vs positive).<br />

Similar analyses were performed for log2 Ki67 as continuous<br />

variable. Added prognostic value was assessed using the likelihood<br />

ratio statistic.<br />

Results: We obtained GG in 883 (62%) of 1428 samples. Non<br />

evaluable results were due to pre-analytical issues or technical failure.<br />

Among the 883 patients, 521 (59%) had N0 disease, 435 (49%) were<br />

treated with tamoxifen and 84 (10%) had distant recurrences. The<br />

GG classified 502 patients with HG2 tumors as either GG1 (N=202,<br />

40%), equivocal (N=220, 44%) or GG3 (N=80, 16%). In univariate<br />

analysis, increasing GG and Ki67 were significantly associated with<br />

lower DRFI (Table). When the 773 N0/1-3 patients were analyzed<br />

based on HG, the Kaplan-Meier estimate for 10-year DRFI was 98%<br />

(95% confidence intervals 96-100%) for the 123 HG1, 92% (88-95%)<br />

for the 456 HG2 and 84% (78-90%) for the 194 HG3 patients. In the<br />

N0/1-3 population, 10 year DRFI based on GG was 95% (96-100%)<br />

for the 290 GG1, 92% (89-95%) for the 309 GG equivocal and 84%<br />

(77-90%) for the 178 GG3 patients. Interestingly, when the 456 HG2<br />

and N0/1-3 patients were analyzed based on GG, the 10-year DRFI<br />

was 95% (92-100%) for the 185 HG2/GG1 patients, 92% (88-96%)<br />

for the 202 HG2/GG equivocal patients, 86% (76-96%) for the 69<br />

www.aacrjournals.org 101s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

HG2/GG3 patients. In all patients either GG or Ki67 as continuous<br />

variable significantly improved the clinicopathological model in<br />

predicting distant recurrence (Table).<br />

Conclusion: Either Genomic Grade or centrally reviewed Ki67<br />

provides independent prognostic information for risk of distant<br />

recurrence beyond clinicopathological characteristics in patients<br />

treated with endocrine therapy.<br />

Table: Univariate and multivariate analysis for DRFI including either GG or Ki67 alone or together<br />

with the clinicopathological model<br />

Degrees of<br />

Model Comparison Patients Chi-square<br />

Freedom P Concordance Index<br />

(95% CI)<br />

GG vs null 883 21.7 1 3.2E-06 0.66 (0.6-0.71)<br />

Ki67 vs null 862 16.9 1 4.0E-05 0.65 (0.6-0.71)<br />

Clinicopathological model + GG<br />

848<br />

vs Clinicopathological model<br />

4.6 1 0.03 0.76 (0.71-0.8)<br />

Clinicopathological model +<br />

Ki67 vs Clinicopathological 833 5.4 1 0.02 0.76 (0.71-0.81)<br />

model<br />

Concordance index of clinicopathological model alone is equal to 0.75 (0.7-0.8)<br />

S4-5<br />

Ki67 levels in pretherapeutic core biopsies as predictive and<br />

prognostic parameters in the neoadjuvant GeparTrio trial<br />

Denkert C, Blohmer JU, Müller BM, Eidtmann H, Eiermann W,<br />

Gerber B, Tesch H, Hilfrich J, Huober J, Fehm T, Barinoff J, Jackisch<br />

C, Prinzler J, Rüdiger T, Budczies J, Erbstoesser E, Loibl S, von<br />

Minckwitz G. Charite University Hospital, Berlin, Germany; <strong>San</strong>kt<br />

Gertrauden Krankenhaus, Berlin, Germany; Christian-Albrechts<br />

Universität zu Kiel, Kiel, Germany; Rotkreuzklinikum, München,<br />

Germany; Universitätsfrauenklinik Rostock, Germany; Bethanien<br />

Krankenhaus, Frankfurt, Germany; Eilenriede Klinik, Hannover,<br />

Germany; University of Düsseldorf, Germany; Universitäts<br />

Frauenklinik, Tübingen, Germany; Kliniken Essen Mitte, Essen,<br />

Germany; Klinikum Offenbach, Offenbach, Germany; Klinikum<br />

Karlsruhe, Karlsruhe, Germany; Klinikum St. Salvator, Halberstadt,<br />

Germany; German <strong>Breast</strong> Group, Neu-Isenburg, Germany<br />

Background: Ki67 has been suggested as a marker for definition of<br />

luminal A and luminal B tumors by the 2011 St. Gallen consensus<br />

panel. However, the cutoffs for Ki67 are still under debate. In<br />

particular, it is not clear if one single cutoff is useful for prognostic<br />

and predictive information in the different molecular subtypes. It is an<br />

advantage of the neoadjuvant approach that predictive and prognostic<br />

outcome measurements can be separated in the same cohort. In<br />

this study, we evaluated a large cohort of core biopsies from the<br />

neoadjuvant GeparTrio trial to investigate the impact of pretherapeutic<br />

Ki67 levels as a predictive marker for response to neoadjuvant<br />

chemotherapy as well as a prognostic marker for progression-free<br />

and overall survival. The analysis was stratified for hormone-receptor<br />

positive and negative tumors as well as HER2 status.<br />

Methods: A total of 1166 pretherapeutic core biopsies from<br />

the neoadjuvant Gepartrio trial were evaluated for Ki67 by<br />

immunohistochemistry, a total of 200 cells were counted in each<br />

sample. Ki67 cutoffs were evaluated using web-based software<br />

Cutoff Finder (http://molpath.charite.de/cutoff/). The details of the<br />

GeparTrio study design have been described before (von Minckwitz,<br />

JNCI 2008). We compared pCR rate as well as the overall and disease<br />

free survival in the complete cohort as well as subgroups of patients<br />

based on hormone receptor and HER2 expression.<br />

Results: Using Ki67 as a continuous parameter, a wide range of<br />

cutoffs between 10% and 80% for Ki67 were predictive for pCR.<br />

For DFS and OS, a wide range of cutoffs between 10% and 45% was<br />

significant. For further analysis, the three groups of Ki67 0-15% vs.<br />

Ki67 15.1%-35% vs. Ki67 >35 were defined and were compared for<br />

different outcome parameters. The pCR rates in these three groups<br />

of Ki67 expression were 4.2%, 12.9% and 29.0% (p35%) as a reasonable approach for further<br />

standardization of this marker.<br />

S4-6<br />

An international Ki67 reproducibility study<br />

Nielsen TO, Polley M-YC, Leung SCY, Mastropasqua MG, Zabaglo<br />

LA, Bartlett JMS, Viale G, McShane LM, Hayes DF, Dowsett M, On<br />

Behalf of the International Ki67 in <strong>Breast</strong> <strong>Cancer</strong> Working Group<br />

of the BIG-NABCG collaboration. University of British Columbia,<br />

Vancouver, BC, Canada; National <strong>Cancer</strong> Institute, Bethesda,<br />

MD; European Institute of Oncology, Milan, Italy; The Institute of<br />

<strong>Cancer</strong> Research, London, United Kingdom; Ontario Institute for<br />

<strong>Cancer</strong> Research, Toronto, ON, Canada; University of Michigan<br />

Comprehensive <strong>Cancer</strong> Center, Ann Arbor, MI; Royal Marsden<br />

Hospital, London, United Kingdom; <strong>Breast</strong> International Group-<br />

North American <strong>Breast</strong> <strong>Cancer</strong> Group Collaboration<br />

Introduction: In breast cancer, immunohistochemical assessment of<br />

the cell proliferation marker Ki67 is of interest for potential use in<br />

clinical management (e.g. prognosis, prediction of treatment response,<br />

monitoring the effect of neoadjuvant therapy). However, lack of<br />

consistency across labs has limited the value of Ki67. A working group<br />

was assembled to devise a strategy to harmonize Ki67 analysis and<br />

scoring and to identify procedures to improve concordance (Dowsett<br />

et al. JNCI,’11). As a result, we have conducted an international Ki67<br />

reproducibility study.<br />

Methods: Step 1: 100 breast cancer cases arranged into 1 mm core<br />

tissue microarrays (TMAs) were centrally stained using MIB-1<br />

antibody. Eight labs scored Ki67 as the percentage of positively<br />

stained invasive tumor cells using their own local method. Six labs<br />

repeated scoring of 50 cases on the same TMA 3 times. Ki67 data<br />

were log2-transformed to approximate a normal distribution. Pairwise<br />

intra-lab and inter-lab concordance was displayed using Bland-<br />

Altman plots. Sources of variation (e.g. patient and lab) were analyzed<br />

using two-way crossed random effects models with quantification of<br />

reproducibility via intraclass correlation coefficient (ICC; range of<br />

0-1, 1 = highest agreement).<br />

Step 2: A web-based scoring calibration interface was created to<br />

mitigate systematic differences in Ki67 interpretation observed in<br />

Step 1. Digital images of TMA cores from 9 “training” and 9 “test”<br />

cases representing a range of Ki67 values were assembled onto the<br />

web-based interface. An initial set of observers yielding consistent<br />

scores using a standardized scoring method served as reference labs.<br />

Detailed instructions for the standardized method were provided<br />

to 8 other labs, who were asked to score the cases and learn from<br />

discrepancies observed via the web tool. Statistical criteria for passing<br />

were pre-specified.<br />

Results: In Step 1, intra-lab reproducibility was high (ICC = 0.94).<br />

Inter-lab reproducibility was only moderate (ICC = 0.71). Absolute<br />

mean Ki67 values across the series ranged from 7% to 24%.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

102s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: General Session 5<br />

Contributing to inter-lab discordance were tumor region selection,<br />

formal counting of nuclei versus visual estimation, and subjective<br />

assessment of staining positivity. Labs using formal counting methods<br />

gave more consistent results than those using visual estimation. The<br />

calibration exercise (Step 2) helped to improve consistency: all 8 labs<br />

passed the training exercise, 4 at first attempt and 4 after 1-2 rounds<br />

of retraining. When the same 8 labs scored the 9 test calibration cases,<br />

3 passed on first attempt. Although overall concordance improved,<br />

discrepancies for cases with low Ki67 were largely responsible for<br />

the failure of the remaining 5 labs to pass the testing.<br />

Conclusions: Absolute values and cutpoints for Ki67, and associated<br />

clinical decisions, cannot be transferred between laboratories without<br />

careful standardization of scoring methodology. Our calibration<br />

training tool offers an initial process to improve inter-lab concordance,<br />

but further studies are required to establish what concordance can be<br />

achieved in practice. A large subsequent study will assess whether a<br />

pre-specified target of success (ICC = 0.9) can be achieved on glass<br />

slides after training with the web-based calibration tools developed<br />

as part of this study<br />

S4-7<br />

DISCUSSANT<br />

W. Fraser Symmans, MD, Department of Pathology, MD Anderson<br />

<strong>Cancer</strong> Center, Houston, TX<br />

S5-1<br />

Biomarker analyses in CLEOPATRA: A phase III, placebocontrolled<br />

study of pertuzumab in HER2-positive, first-line<br />

metastatic breast cancer (MBC)<br />

Baselga J, Cortés J, Im S-A, Clark E, Kiermaier A, Ross G, Swain SM.<br />

Massachusetts General Hospital <strong>Cancer</strong> Center and Harvard Medical<br />

School, Boston, MA; Vall d’Hebron University Hospital, Barcelona,<br />

Spain; Seoul National University College of Medicine, Seoul, Korea;<br />

Roche Products Limited, Welwyn, United Kingdom; F. Hoffmann-La<br />

Roche Limited, Basel, Switzerland; MedStar Washington Hospital<br />

Center, Washington, DC<br />

Background<br />

The anti-HER2 humanized monoclonal antibodies trastuzumab (T)<br />

and pertuzumab (P) have complementary mechanisms of action.<br />

CLEOPATRA showed that P+T+docetaxel (D) significantly improved<br />

progression-free survival (PFS) compared with placebo (Pla)+T+D<br />

in the first-line treatment of HER2-positive MBC (HR=0.62, 95%<br />

CI 0.51–0.75; p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Materials and Methods: HERA (BIG 01-01) is an international,<br />

multicenter, phase III randomized trial involving 5102 women<br />

with HER2-positive EBC. Pts were randomized, after completion<br />

of primary therapy [surgery, chemotherapy and radiotherapy as<br />

indicated], to T every 3 weeks for 1 yr, 2 years (yrs), or observation.<br />

This landmark efficacy analysis compares the outcome of pts<br />

randomized to either 2 yrs or 1 yr of T who were disease-free at 1 yr<br />

after randomization (N=1553 for 2 yrs, and N=1552 for 1 yr). The<br />

primary endpoint is DFS and secondary endpoints are OS and time to<br />

distant recurrence (TTDR). Updated efficacy analyses of the T arms<br />

vs. observation at 8-yrs of median follow-up (FU) are also presented.<br />

Results: On 12 April 2012 HERA reached the target number of 725<br />

DFS events needed for 80% power to detect a true hazard ratio (HR)<br />

of 0.80 for the comparison of 2 yrs vs. 1 yr of T. The unadjusted HR<br />

for an event in the 2-yr vs. 1-yr T arms was 0.99 (95% CI 0.85-1.14;<br />

P=0.86). OS in the two arms was comparable [HR=1.05 (95% CI<br />

0.86-1.28; P=0.63)]. TTDR results were similar. The primary cardiac<br />

endpoint* was comparable (1.0% vs. 0.8% for 2-yr and 1-yr arms,<br />

respectively), but the secondary cardiac endpoint** was higher in<br />

the 2-yr arm (7.2% vs. 4.1%). Importantly, the durable benefit in<br />

DFS and OS for both 1 yr and 2 yrs of T compared with observation<br />

remains stable at 8 yrs of median FU. Subgroup analyses including<br />

by hormone receptor status will be available for the meeting.<br />

Conclusions: These results confirm that 1 yr of adjuvant T remains<br />

the standard of care for HER2-positive EBC pts. It is also reassuring<br />

that the significant improvement in DFS and OS persists over time<br />

and that the incidence of cardiac endpoints remains low at a median<br />

FU of 8 yrs.<br />

* NYHA class III or IV, confirmed by a cardiologist, and LVEF <<br />

50% and ≥ 10% below baseline, OR cardiac death.<br />

** LVEF < 50% and ≥ 10% below baseline confirmed by repeat<br />

assessment, excluding patients with a primary cardiac endpoint.<br />

S5-3<br />

PHARE Trial results of subset analysis comparing 6 to 12 months<br />

of trastuzumab in adjuvant early breast cancer<br />

Pivot X, Romieu G, Bonnefoi H, Pierga J-Y, Kerbrat P, Guastalla J-P,<br />

Lortholary A, Espié M, Fumoleau P, Khayat D, Pauporte I, Kramar A.<br />

University Hospital J. Minjoz, Besancon, France; Anti <strong>Cancer</strong> Center<br />

Val d’Aurelle, Montpellier, France; Anti <strong>Cancer</strong> Center Bergonie,<br />

Bordeaux, France; Curie Institute, Paris, France; Anti <strong>Cancer</strong><br />

Center Eugene Marquis, Rennes, France; Anti <strong>Cancer</strong> Center Leon<br />

Berard, Lyon, France; Catherine de Sienne Center, Nantes, France;<br />

University Hospital St Louis, Paris, France; Anti <strong>Cancer</strong> Center<br />

Georges Francois Leclerc, Dijon, France; University Hospital Pitie<br />

Salpetriere, Paris, France; French National <strong>Cancer</strong> Institut (INCa),<br />

France; Centre Oscar Lambret, Lille, France<br />

Background: Since 2005, One year trastuzumab (T) treatment has<br />

been providing survival benefit to patients with early breast cancer<br />

and HER2 overexpression. However, the optimal duration of T<br />

has been debatable due to concerns for cardiac toxicity and results<br />

from the FinHer trial which showed that 9 weeks of T provided a<br />

similar magnitude of benefit than the 1-year treatment. The French<br />

National <strong>Cancer</strong> Institute (INCa) initiated an academic randomised<br />

non-inferiority trial aiming to compare a shorter T exposure of 6<br />

months versus the standard 12 months. This trial, named PHARE<br />

for ‘Protocol for Herceptin® as Adjuvant therapy with Reduced<br />

Exposure’ [NCT00381901] was approved byan independent ethics<br />

committee with regularly planned IDMC meetings.<br />

Patients and methods: Patients with HER2 + early breast cancer<br />

who received at least 4 cycles of (neo)-adjuvant chemotherapy<br />

were eligible. Randomization 1:1 using a minimisation algorithm<br />

was stratified on concomitant or sequential T administration with<br />

chemotherapy, oestrogen receptors (ER) status and center. The<br />

primary objective was to compare disease free survival (DFS). Overall<br />

survival (OS) and cardiac toxicity were investigated as secondary<br />

aims. An absolute loss of 2% in DFS in the experimental arm was<br />

defined as the non-inferiority margin (1.15 in relative terms) and<br />

required 3400 patients with alpha=0.05 and 80% power.<br />

Results: Between 5/2006 and 7/2010, 3382 patients were randomized<br />

to 6 or 12 months of T following the IDMC recommendation for<br />

accrual interruption and extended follow-up. Disease and treatment<br />

characteristics were well balanced between the 2 arms: median age<br />

55 years (range 21 - 86), median tumour size 20 mm (0 - 270), node<br />

involvement 45%, SBR grade III 56%, ER positive 58%, radiotherapy<br />

88%; concomitant T administration 58%, anthracyclin and taxane<br />

containing chemotherapy 73%. The data-base was locked on July<br />

30 th , 2012. The 95% HR confidence interval was ranged between 1.09<br />

- 1.66 for DFS between the 2 arms and it included the pre-specified<br />

non-inferiority margins of 1.15. Thus the results were inconclusive<br />

regarding the non-inferiority (p = 0.14). A subset analysis based on<br />

stratification factors will be presented at SABCS.<br />

S5-4<br />

EGFR expression measured by quantitative immunofluorescense<br />

is associated with decreased benefit from trastuzumab in the<br />

adjuvant setting in the NCCTG (Alliance) N9831 trial.<br />

Rimm D, Ballman KV, Cheng H, Vassilakopoulou M, Chen B,<br />

Gralow J, Hudis C, Davidson NE, Psyrri A, Fountzilas G, Perez<br />

EA. Yale University School of Medicine, New Haven, CT; Mayo<br />

Clinic, Rochester, MN; Seattle <strong>Cancer</strong> Care Alliance, Seattle, WA;<br />

Memorial Sloan-Kettering <strong>Cancer</strong> Center, New York, NY; University<br />

of Pittsburgh <strong>Cancer</strong> Institute, Pittsburgh, PA; “Papageorgiou”<br />

Hospital, Aristotle University of Thessaloniki School of Medicine,<br />

Thessaloniki, Macedonia, Greece; Attikon University Hospital,<br />

Athens, Greece; Mayo Clinic, Jacksonville, FL<br />

Background: Epidermal Growth Factor Receptor (EGFR, HER1) is<br />

known to heterodimerize with HER2 and may impact the effectiveness<br />

of trastuzumab. Historically, EGFR has been difficult to measure;<br />

thus there is no evidence regarding its effects on trastuzumab<br />

clinical therapy. Here we use a new, standardized, quantitative<br />

immunofluorescence assay and a novel EGFR antibody to evaluate<br />

the correlation between EGFR expression and clinical outcome in a<br />

cohort from the North Central <strong>Cancer</strong> Treatment Group (NCCTG,<br />

Alliance) N9831 trial.<br />

Methods: Of the 3505 women in the N9831 trial, tissue microarrays<br />

were constructed that allowed analysis of 1,444 patients randomly<br />

assigned to receive chemotherapy alone (Arm A), sequential<br />

trastuzumab after chemotherapy (Arm B) or concurrent trastuzumab<br />

with chemotherapy (Arm C). Fluorescence-based, standardized<br />

measurement of EGFR was done using the rabbit monoclonal EGFR<br />

antibody D38B1 on the AQUA platform (HistoRx). EGFR expression<br />

levels were analyzed as a continuous variable and dichotomized using<br />

recursive partitioning to define an optimal cut point. The cut point<br />

was validated on an independent retrospective cohort comprised<br />

of 130 patients with HER2-positive metastatic breast cancer who<br />

received trastuzumab.<br />

Results: EGFR assessed as a continuous variable shows an<br />

association with disease free survival (DFS) in Arm C (p=0.002) but<br />

not in Arms A or B. Dichotomizing EGFR expression shows that<br />

high level expression is associated with worse outcome (HR=2.2;<br />

95% CI 1.33-3.59, p=0.002) in arm C. Five year DFS within the low<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

104s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: General Session 5<br />

level EGFR expression group was 71%, 74% and 90% for Arms A,<br />

B and C, respectively (log-rank p-value=0.02) and within the high<br />

level EGFR expression (n= 213) group was 62%, 76% and 74%<br />

respectively (log-rank p-value 0.22). As validation, the same cut-point<br />

was used to define high EGFR expression in a retrospective metastatic<br />

breast cancer cohort. The median progression free survival difference<br />

between EGFR high group and EGFR low group was 6.1 months<br />

(p=0.0063) and the hazard ratio of high EGFR was 1.92 (p=0.0073).<br />

Conclusions: High expression of EGFR by the AQUA platform<br />

appears to be associated with decreased benefit from adjuvant<br />

concurrent (not sequential) trastuzumab and shorter PFS in the<br />

metastatic setting. The biological underpinning for these observations<br />

is being considered. Since there are other treatment options for HER2-<br />

driven tumors, this marker, once validated, could help select patients<br />

for alternative or additive therapy.<br />

S5-5<br />

Trastuzumab plus adjuvant chemotherapy for HER2-positive<br />

breast cancer: Final planned joint analysis of overall survival<br />

(OS) from NSABP B-31 and NCCTG N9831<br />

Romond E, Suman VJ, Jeong J-H, Sledge, Jr. GW, Geyer, Jr. CE,<br />

Martino S, Rastogi P, Gralow J, Swain SM, Winer E, Colon-Otero G,<br />

Hudis C, Paik S, Davidson N, Mamounas EP, Zujewski JA, Wolmark<br />

N, Perez EA. National Surgical Adjuvant <strong>Breast</strong> and Bowel Project<br />

(NSABP) Operations and Biostatistical Centers; University of<br />

Kentucky; Mayo Clinic; University of Pittsburgh Graduate School<br />

of Public Health; IU Simon <strong>Cancer</strong> Center; University of Texas<br />

Southwestern Medical Center; The Angeles Clinic and Research<br />

Institute; University of Pittsburgh <strong>Cancer</strong> Institute; University of<br />

Washington; Medstar Washington Hospital Center; Dana-Farber<br />

<strong>Cancer</strong> Institute; Memorial Sloan-Kettering <strong>Cancer</strong> Center; Aultman<br />

Hospital; <strong>Cancer</strong> Therapy Evaluation Program, National <strong>Cancer</strong><br />

Institute, National Institutes of Health, D.H.H.S; Allegheny <strong>Cancer</strong><br />

Center Allegheny General Hospital<br />

Background: Preplanned combined analysis of the National Surgical<br />

Adjuvant <strong>Breast</strong> and Bowel Project (NSABP) B-31 trial and North<br />

Central <strong>Cancer</strong> Treatment Group (NCCTG) (Alliance) N9831 Trial<br />

compared adjuvant chemotherapy with or without trastuzumab in<br />

women with HER2-positive breast cancer. The initial results were<br />

reported in 2005 and updated in early 2011<br />

Methods: 4045 patients with HER2-positive operable breast cancer<br />

were enrolled to receive doxorubicin plus cyclophosphamide followed<br />

by paclitaxel with or without trastuzumab in both trials. The definitive<br />

analysis for OS will occur when 710 events have been reported.<br />

Results: Median time on study is 8 years. The required number of<br />

events have occurred for the definitive analysis for OS to be conducted<br />

by the end of September 2012. Updated analyses of disease free<br />

survival and related subgroups will also be presented.<br />

Supported by NCI grants U10-CA-12027, -69651, -37377, -69974,<br />

-44066, U24-CA-114732, CA25224; <strong>Breast</strong> <strong>Cancer</strong> Research<br />

Foundation; Genentech 35-03<br />

S5-6<br />

Activating HER2 mutations in HER2 gene amplification negative<br />

breast cancers.<br />

Bose R, Kavuri SM, Searleman AC, Shen W, Shen D, Koboldt DC,<br />

Monsey J, Li S, Ding L, Mardis ER, Ellis MJ. Washington University<br />

School of Medicine, St. Louis, MO<br />

Background: <strong>Breast</strong> cancer genome sequencing projects, performed<br />

by the genome sequencing centers in the U.S., Canada, and the U.K.,<br />

are elucidating the somatic mutations and other genomic alterations<br />

that occur in human breast cancer. These studies recently identified<br />

somatic HER2 mutations in breast cancers lacking HER2 gene<br />

amplification.<br />

Results: Compilation of data from seven sequencing studies<br />

documented 22 patients with somatic HER2 mutations. These<br />

mutations clustered in three regions. The first cluster was at amino<br />

acid (aa) 309-310 (exon 8), located in the extracellular domain. These<br />

aa residues form part of the HER2 dimerization interface. The second<br />

cluster was at aa 755-781, located in the kinase domain (exons 19-20).<br />

This was the most common location for HER2 mutations, with 17 out<br />

of 22 patients having somatic mutations here. The third region was at<br />

aa 835-896, also in the kinase domain (exons 21-22). Using multiple<br />

experimental approaches (cell line experiments, in vitro kinase<br />

assays, protein structure modeling, and xenograft experiments), we<br />

tested seven of these HER2 mutations and showed that 4 of them are<br />

activating mutations that are sensitive to lapatinib and trastuzumab.<br />

Another 2 mutations were found to be lapatinib resistant and we<br />

determined their sensitivity to neratinib, canertinib, and gefitinib.<br />

Conclusions: These findings biologically validate somatic HER2<br />

mutations as good targets for breast cancer treatment, but the<br />

appropriate choice of targeted drug is dependent on the precise<br />

mutation present. This study is among the first to functionally<br />

characterize mutations identified by breast cancer genome sequencing.<br />

A prospective, multi-institutional clinical trial has been launched to<br />

screen for HER2 mutation positive patients and determine the clinical<br />

outcome of treatment with HER2 targeted drugs.<br />

S5-7<br />

Combined Blockade of PI3K/AKT and EGFR/HER3 Enhances<br />

Anti-Tumor Activity in Triple Negative <strong>Breast</strong> <strong>Cancer</strong><br />

Tao J, Ip P, Auricchio N, Juric D, Yu M, Shyamala M, Kim P, Singh<br />

S, Hazra S, Haber D, Scaltriti M, Baselga J. Massachusetts General<br />

Hospital; Prometheus Lab<br />

Background: Up to 60% of triple negative breast cancers (TNBCs)<br />

express high levels of EGFR (HER1). Moreover, TNBCs are<br />

associated with increased frequency of phosphatase and tension<br />

homologue (PTEN) loss of function, leading to hyperactivation of<br />

the phosphoinositide 3-kinase (PI3K)/AKT pathway. This provides<br />

the rationale for using PI3K/AKT inhibitors in this subset of patients.<br />

However, compensatory expression of receptor tyrosine kinases<br />

(RTKs) such as HER3 may limit efficacy of PI3K/AKT inhibitors.<br />

Therefore, we hypothesize that combined targeting of both HER<br />

family receptors and the PI3K/AKT pathway will result in superior<br />

antitumor activity compared to single agent blockade in TNBC.<br />

Materials and Methods: TNBC cell lines MDA-MB-231, MDA-<br />

MB-468, HCC70, HCC1143 and HCC1937 were treated with<br />

MEHD7945A, a dual-action antibody that targets both EGFR and<br />

HER3, AKT inhibitor GDC0068, and pan-PI3K inhibitor GDC0941.<br />

Cell viability was measured by CellTiter-Glo and Crystal Violet. Both<br />

cell line- and patient-derived xenograft models of TNBC were grown<br />

subcutaneously in Nu/Nu mice and then treated with MEHD7945A,<br />

GDC0068, GDC0941, or the combination of MEHD7945A with<br />

either GDC0068 or GDC0941. Tumor size and histology were<br />

examined. Circulating tumor cells (CTCs) were isolated from murine<br />

peripheral blood by Herring Bone CTC chip. Protein expression was<br />

measured by Western blot, Reverse Phase Protein Analysis (RPPA),<br />

and immunohistochemistry.<br />

Results: GDC0068 and GDC0941 treatment resulted in variable<br />

inhibition of cell viability, with IC50s ranging from 170 nM to<br />

>1 µM across all TNBC cell lines. Under full-serum (10% FBS)<br />

conditions, MEHD7945A led to a very mild decrease in cell viability<br />

www.aacrjournals.org 105s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

in vitro. Combination treatments were modestly more effective than<br />

monotherapy in vitro. In vivo, MEHD7945, GDC0941 and GDC0068<br />

showed variable delay in tumor growth whereas combination of<br />

MEHD7945A with either GDC0068 or GDC0941 was superior to<br />

single agent treatment. While untreated tumors increased over 14<br />

fold in size, tumor growth of MEHD7945, GDC0941 and GDC0068<br />

treated xenografts reached 6, 4 and 2.5 fold increase respectively.<br />

However, both combination arms prevented tumor growth, with<br />

complete responses achieved in 1/9 mice in each of the combination<br />

cohorts. Of note, all the treatments (up to 9 weeks of therapeutic<br />

exposure) were well tolerated. Analysis of treated tumors reveals<br />

potent inhibition of the PI3K/AKT pathway, with decreased levels<br />

phospho-PRAS40, and phospho-S6, as well as decreased expression<br />

of total EGFR and HER3. Circulating tumor cells are being used as<br />

surrogate markers of pathway inhibition.<br />

Conclusions: Combined therapy with MEHD7945A and either<br />

GDC0068 or GDC0941 was superior to monotherapy in preclinical<br />

models of TNBC. We observed significantly greater effect with the<br />

combination treatment in vivo compared to in vitro. We are currently<br />

exploring whether expression/activation of the HER receptors<br />

following PI3K/Akt blockade are accentuated in vivo. Additionally,<br />

this may be the first mouse model enabling CTC collection from<br />

subcutaneous xenografts, allowing for real-time pharmacodynamic<br />

investigation. These findings provide the rationale for combined<br />

targeting of PI3K/AKT and EGFR/HER3 in triple negative breast<br />

cancers.<br />

S5-8<br />

Parallel upregulation of Bcl2 and estrogen receptor (ER)<br />

expression in HER2+ breast cancer patients treated with<br />

neoadjuvant lapatinib.<br />

Giuliano M, Wang YC, Gutierrez C, Rimawi MF, Chang JC, Wang<br />

T, Hilsenbeck SG, Trivedi MV, Chamness GC, Osborne CK, Schiff<br />

R. Lester & Sue Smith <strong>Breast</strong> Center, Baylor College of Medicine,<br />

Houston, TX; Baylor College of Medicine, Houston, TX; The<br />

Methodist Hospital Research Institute, Houston, TX; University of<br />

Houston, TX<br />

Background: We previously showed in HER2+ models of breast<br />

cancer (BC) that potent inhibition of the HER receptor layer can<br />

lead to re-expression of estrogen receptor (ER) or activation of the<br />

ER pathway. Consequently, the anti-apoptotic ER gene product<br />

Bcl2 is upregulated, resulting in enhanced tumor cell survival and<br />

treatment resistance. In this study, we investigated whether Bcl2 and<br />

ER expression levels are simultaneously increased by neoadjuvant<br />

treatment with the dual HER1/2 tyrosine kinase inhibitor lapatinib<br />

in HER2+ BC patients.<br />

Methods: In a neoadjuvant phase II clinical trial 49 HER2+ BC<br />

patients were treated with lapatinib as a single agent for 6 weeks,<br />

followed by trastuzumab/docetaxel for 12 weeks before surgery.<br />

Tumor specimens were prospectively collected at different timepoints<br />

during lapatinib treatment (baseline, and weeks 2 and 4).<br />

Bcl2, ER, progesterone receptor (PR), total (t) and phosphorylated<br />

(p)-HER2, and Ki67 were assessed by immunohistochemistry.<br />

Spearman correlation was used to evaluate the association among the<br />

biomarkers at baseline, and the correlation of their changes over time.<br />

Fisher’s Exact test and non-parametric Wilcoxon rank sum test were<br />

used respectively to determine if the frequency and the magnitude<br />

of Bcl2 expression changes were associated with baseline ER status.<br />

Results: 35/49 HER2+ tumor specimens (71%) were available for<br />

baseline evaluation of Bcl2 and ER. Of those, 12 (34%) were ERpositive<br />

(Allred score ≥ 3) and 23 (66%) ER-negative. Baseline Bcl2<br />

expression correlated positively with ER (r=.75; p


December 4-8, 2012 <strong>Abstract</strong>s: General Session 6<br />

were performed using supervised and unsupervised methods. A<br />

bias in isoform quantitation was first identified that correlated with<br />

RNA Integrity Score (i.e. RIN). When this bias was accounted<br />

for, unsupervised methods identified 9 genes that demonstrated<br />

significant isoform switching that was independent of subtype.<br />

Supervised analysis of basal-like versus luminal tumors identified<br />

30 genes showing isoform switching including CD44, CTNND1 and<br />

RAD51L1; most of these unique isoform switches also correlated<br />

with the expression of many other genes that did not show isoform<br />

changes. Additional analyses are being performed including 1)<br />

identification of genes expressing more than one isoform, but which<br />

do not change dramatically in abundance across samples, and 2)<br />

correlations of genomic DNA somatic mutations with the mRNA-seq<br />

based mutation detection.<br />

Conclusions: Appropriate processing of mRNA-sequencing data<br />

yields unparalleled information in isoform level abundance and<br />

robust expression signatures. The modest isoform switching observed<br />

here segregated samples into distinct groups of common switching<br />

events, most of which correlated with subtype. The additional data<br />

types provided by mRNA-seq including isoform identification, fusion<br />

gene detection, and mutation information, extend the capabilities of<br />

expression analyses and may provide new data of clinical utility.<br />

S6-2<br />

Characterization of different foci of multifocal breast cancer using<br />

genomic, transcriptomic and epigenomic data<br />

Desmedt C, Nik-Zainal S, Fumagalli D, Rothé F, Singhal S, Majjaj<br />

S, Brown D, Dedeurwaerder S, Defrance M, Maetens M, Adnet P-Y,<br />

Vincent D, Salgado R, Fuks F, Piccart M, Larsimont D, Campbell<br />

P, Sotiriou C. Institut Jules Bordet, Brussels, Belgium; Wellcome<br />

Trust <strong>San</strong>ger Institute, Hinxton, United Kingdom; Université Libre<br />

de Bruxelles, Brussels, Belgium<br />

Background: A significant proportion of breast cancer (BC) patients<br />

(pts) develop multiple synchronous unilateral breast tumors, also<br />

referred to as multifocal BC (MBC), which represent a diagnostic<br />

and therapeutic challenge. Here, we aimed first to better define the<br />

incidence of MBCs and then to compare different foci from ductal<br />

MBCs in a global analysis using genomic, transcriptomic and<br />

epigenomic data.<br />

Methods: The incidence of MBCs was sought via a systematic query<br />

of all pathology reports between 2000 and 2010 within the Institut<br />

Bordet. The biological characterization of MBCs focused on 5 ductal<br />

MBCs for which the foci did not differ in terms of grade, hormonal<br />

receptors and HER2 status, since this is the case for the majority of<br />

MBCs. It involved the identification of somatic rearrangements (Rs),<br />

transcriptomic and epigenomic profiling in 2 foci from each pt, via<br />

low-coverage whole genome sequencing, HG133 Plus 2.0 Chips and<br />

Infinium Methylation 450K arrays respectively. Genomic Rs were<br />

validated using an orthogonal sequencing platform.<br />

Results: MBC is a frequent finding since it concerns 24% (1410/5811)<br />

of primary BCs. When investigating the genomics of the 5 MBCs,<br />

we observed that the number of Rs varied in each pt, with 3 pts<br />

having few ( 100Rs with a<br />

particular tandem-duplication phenotype. All pts had Rs common to<br />

both foci, suggesting a shared genetic background. Strikingly, in 4 pts,<br />

we observed a branched evolutionary pattern since private Rs were<br />

present in each focus. In contrast, we observed a linear evolutionary<br />

pattern in the remaining pt, since private Rs were only found in 1<br />

of the 2 foci.<br />

Two pts were genetically very similar with ≥75% common Rs.<br />

Although the transcriptomic profiles looked very similar between<br />

the 2 foci, significant epigenomic differences were observed in 1 of<br />

the 2 pts investigated. Two other pts had 50% common Rs. In one pt<br />

there were only minor differences in transcriptomic and methylation<br />

patterns between the 2 foci. Contrastingly, in the second pt, DNA<br />

methylation patterns were dramatically discordant between the foci,<br />

suggesting that genomic, transcriptomic and epigenomic patterns are<br />

not necessarily correlated. Only in 1 pt of the 5 investigated, the 2<br />

foci were extremely different, with only 14% of common Rs. These<br />

genetic differences were associated with dramatic differences in<br />

methylation and transcriptomic profiles. Interestingly, for 4 pts some<br />

of the identified rearrangements were found in parts of tumor-adjacent<br />

histologically normal breast tissue.<br />

Conclusions: Today, the College of American Pathologists<br />

recommends analysing more than 1 focus from MBC if they differ<br />

in histology and grading (Lester 2009). Here, we demonstrated<br />

for the first time that different lesions from MBC, which concerns<br />

¼ of the ductal BC population, can differ at the (epi)genetic and<br />

transcriptomic level even when the foci present similar histology,<br />

grading, hormonal and HER2 status. Since the number of genetic<br />

alterations with potential clinical utility is rapidly growing due to<br />

increasing numbers of targeted therapies, these findings suggest<br />

that interrogating only the largest lesion might not be sufficient for<br />

adequate management of MBCs.<br />

S6-3<br />

Neurocognitive impact in adjuvant chemotherapy for breast<br />

cancer linked to fatigue: A Prospective functional MRI study<br />

Cimprich B, Hayes DF, Askren MK, Jung MS, Berman MG, Ossher<br />

L, Therrien B, Reuter-Lorenz PA, Zhang M, Peltier S, Noll DC.<br />

University of Michigan, Ann Arbor, MI; University of Washington,<br />

Seattle, WA; Rotman Research Institute at Baycrest, University of<br />

Toronto, Canada<br />

Background: Our previous research showed evidence of compromised<br />

cognitive function prior to adjuvant chemotherapy for breast cancer,<br />

with fatigue as a contributory factor. Fatigue is a common symptom<br />

reported by women treated for breast cancer, yet its association with<br />

neurocognitive function has not been systematically examined. In this<br />

prospective study, we examined possible alterations in neurocognitive<br />

responses, namely, working memory, from pre- to post- adjuvant<br />

treatment during functional magnetic resonance imaging (fMRI) and<br />

further investigated whether early fatigue might be linked to cognitive<br />

alterations over time.<br />

Methods: Women treated with either adjuvant chemotherapy<br />

(anthracyline-based combination regimen, n=29) or radiotherapy<br />

(n=37) for localized breast cancer (Stages 0-IIIa) and age-matched<br />

healthy controls (n=32) were enrolled. Participants performed a verbal<br />

working memory task (VWMT) with varying levels of demand for<br />

cognitive control during fMRI scanning and provided self-reports of<br />

fatigue (FACT-F) at two time points coincident with pre- and onemonth<br />

post chemotherapy assessments. Imaging data were analyzed<br />

with general linear models using SPM5; comparative statistics were<br />

used to determine group differences, and correlational analyses<br />

addressed relationships of fatigue and neurocognitive measures.<br />

Findings: The chemotherapy group reported significantly greater<br />

severity of fatigue (p < .05) and performed less accurately on the<br />

VWMT both pre- and one-month post-treatment than the other groups.<br />

Greater fatigue was correlated with poorer performance on the VWMT<br />

at both time points across groups, with stronger correlation posttreatment<br />

(r = -.22, p = .03). A 2 time-point (pre- vs. post-treatment) x<br />

2 group (chemotherapy vs. controls) x 2 demand-level contrasts (high<br />

minus low vs. medium minus low) analytic model showed a significant<br />

www.aacrjournals.org 107s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

group x time interaction (p < .05), mainly due to lower pre-treatment<br />

activation in an area of the prefrontal cortex supporting working<br />

memory, the anatomical left inferior frontal gyrus (LiFG), at higher<br />

task demand in the chemotherapy group. The radiotherapy group<br />

scored between the other two groups with intermediate activation<br />

of those contrasts. Of interest, lower pre-treatment activation in the<br />

LiFG in the high-low demand contrast predicted severity of fatigue<br />

across all participants at the post-treatment assessment (r = -.27, p <<br />

.01), linking early compromise in neurocognitive performance with<br />

greater fatigue over time.<br />

Discussion: Neurocognitive alterations during a working memory task<br />

and greater fatigue were evident before any adjuvant chemotherapy<br />

for breast cancer. Notably, functional alterations in working memory<br />

processes were evident with fMRI before adjuvant chemotherapy and<br />

predicted severity of post-treatment fatigue. Importantly, across all<br />

participants, greater fatigue over time was correlated with reduced<br />

cognitive performance. Taken together, these findings indicate<br />

that pre-treatment neurocognitive compromise and fatigue are<br />

key contributors to the cognitive impact often attributed solely to<br />

chemotherapy. Early therapeutic interventions targeting fatigue may<br />

improve cognitive function and reduce the distress of “chemo brain”<br />

throughout the course of adjuvant treatment.<br />

S6-4<br />

Vitamin D, but not bone turnover markers, predict relapse in<br />

women with early breast cancer: an AZURE translational study.<br />

Coleman RE, Rathbone EJ, Marshall HC, Wilson C, Brown JE,<br />

Gossiel F, Gregory WM, Cameron D, Bell R. University of Leeds,<br />

United Kingdom; University of Sheffield, United Kingdom; University<br />

of Edinburgh, United Kingdom; Andrew Love <strong>Cancer</strong> Centre,<br />

Geelong, Australia<br />

Background: The AZURE trial evaluated the addition of zoledronic<br />

acid (ZOL) to standard adjuvant therapy on relapse rates and<br />

survival in pts with stage II/III breast cancer. While the overall<br />

analysis reported no benefit, a pre-planned subgroup analysis showed<br />

significant benefits in postmenopausal (PM) women. Here we report<br />

whether baseline measurements of 25-hydroxyvitamin D (25-OHD)<br />

and the serum bone turnover markers N-terminal propeptide type I<br />

procollagen (P1NP) and beta C-terminal telopeptide type I collagen<br />

(β-CTX) can identify pts at high risk of relapse and/or modify the<br />

treatment effects of ZOL.<br />

Methods: 872 AZURE pts consented for serum to be collected and<br />

stored for translational research projects (ZOL group n=431; control<br />

group n=441). Serum P1NP, β-CTX and 25-OHD levels were<br />

measured in 867, 863 and 856 pts respectively. These markers were<br />

analysed as categorical variables with determined cut-off points to<br />

predict bone as 1st site of distant recurrence (DR) or any distant<br />

relapse (P1NP (ng/ml), >70 versus ≤70; βCTX (ng/ml), >0.299 versus<br />

≤0.299; 25-OHD (ng/ml), ≤30 versus >30). Additionally, the markers<br />

were assessed for an interaction with ZOL. Analyses were adjusted<br />

for systemic therapy, lymph node and ER status. As in previous<br />

analyses of AZURE, the PM cohort was defined as ≥5 years since<br />

last menses; all other patients are included in a “not postmenopausal”<br />

(not-PM) group.<br />

Results: The baseline disease characteristics of the translational cohort<br />

were similar to those of the total AZURE population and showed a<br />

similar benefit of ZOL among the PM patients: hazard ratio (HR) for<br />

DR = 0.61 (95% CI 0.35, 1.07; p-value for interaction with not-PM<br />

women = 0.0173). At a median follow-up of 53.3 months, 150 pts<br />

have had a 1st DR event; 47 had bone-only relapse, 19 bone plus<br />

other distant site and 84 developed non-bone DR.<br />

Mean value of 25-OHD (ng/ml) was 18.30 (range, 3.16-54.82;<br />

SD=9.19) with 61.0% of pts 30.<br />

25-OHD levels >30 ng/ml were significantly associated with lower<br />

risk for bone relapse (HR 0.11, 95% CI 0.02, 0.76; p-value 0.0257).<br />

A similar trend was seen for any site of DR (HR 0.56, 95% CI 0.31,<br />

1.01; p-value = 0.0519). Additionally, 25-OHD levels were prognostic<br />

for bone relapse when analysed as a continuous value (HR 0.97, 95%<br />

CI 0.94, 1.00; p-value = 0.0474). Finally, 25-OHD levels >30 ng/<br />

ml predicted for benefit of treatment with ZOL in PM women with<br />

regards to DR (HR 0.09, 95% CI 0.01, 0.82; interaction p-value<br />

0.0747) but the trend was not seen in not-PM women.<br />

Mean values for P1NP and βCTX (range; SD) were 59.1 ng/ml (


December 4-8, 2012 <strong>Abstract</strong>s: General Session 6<br />

invasive disease-free survival (IDFS) [Hudis 2007] with adjuvant CT<br />

± 1 year of BEV. Secondary outcome measures are overall survival<br />

(OS), breast cancer-free interval, disease-free survival (DFS), distant<br />

DFS, and safety (NCI CTCAE v3.0). The sample size was calculated<br />

to provide 80% power for a HR=0.75 at α=0.05 assuming 5-year<br />

IDFS of 72.0% with CT vs 78.2% with CT + BEV with 388 events.<br />

BEATRICE also includes evaluation of potential predictive and<br />

prognostic biomarkers.<br />

Results: Between Dec 2007 and Mar 2010, 2591 patients were<br />

randomized. At data cut-off (Feb 29, 2012), median follow-up was<br />

32 months.<br />

Patient characteristics, % of patients CT (N=1290)<br />

CT + BEV (N=1301)<br />

Age, years<br />


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

docetaxel (T) (Gianni L, Lancet Oncol 2012) were characterized. We<br />

assessed the association of pre-selected adaptive immune functions<br />

and key immune regulatory genes with the likelihood of achieving a<br />

pathological complete response (pCR) in NeoSphere.<br />

Methods: Baseline core biopsies were collected from 387/417<br />

patients. Gene expression profiles (Affymetrix U133 Plus 2.0) were<br />

obtained for 367 samples (88% of all patients) that were evenly<br />

distributed among arms A (TH, n=90), B (THP, n=95), C (HP, n=98),<br />

and D (TP, n=84). ER status was defined by IHC. The primary endpoint<br />

was pCR in breast. We assessed metagenes (average expression of the<br />

associated genes) corresponding to: immunoglobulins (IgG), CD8A,<br />

MHC type I and type II (MHC1, MHC2), interferon inducible genes<br />

(I-IG) and STAT1-related genes. We also assessed the individual<br />

genes PD1, PD-L1, PD-L2, CTLA4, IFNG.<br />

Results: ER+ and ER- tumors had differential mRNA expression<br />

for: CD8A, IgG, PD-L2, PD1, IFNG (overexpressed in ER-), and<br />

I-IG (overexpressed in ER+). Positive correlations were observed<br />

between most of these biomarkers. Only PD1 was inversely correlated<br />

with STAT1, interferon and MHC2. In logistic univariate regression<br />

analyses some biomarkers showed moderate association with pCR or<br />

residual disease without consistent patterns between treatment arms.<br />

However, a multivariate logistic regression model constructed for<br />

all the selected biomarkers revealed that high expression of PD-L1<br />

was consistently associated with lower pCR rate in all chemotherapy<br />

containing arms (A, B and D). A similar trend was present also in Arm<br />

C, the arm with antibody treatment alone. In all arms, high expression<br />

of IFNG and/or STAT1 were associated with higher pCR rate. In<br />

multivariate analysis PD-L1, PD1 and STAT1 were associated with<br />

pCR irrespective of the ER status in combined arms A, C and D. In<br />

arm B, both CTLA4 and PD-L1 were independently associated with<br />

lower pCR in ER- tumors.<br />

Conclusions: The association between pCR and selected immune<br />

biomarkers in patients treated with pertuzumab, trastuzumab, or both,<br />

with or without chemotherapy, supports our understanding of the key<br />

role of the immune system in contributing to HER2-targeted antibody<br />

therapy on top of signaling inhibition. High PD-L1 expression<br />

emerged for its consistent association with lower pCR. These results<br />

provide a rationale for combining HER2-targeted treatments with<br />

immune-modulating agents and may allow the prediction of treatment<br />

benefit. Validation of these results with different assays is ongoing.<br />

PD01-01<br />

Overcoming endocrine therapy resistance related to PTEN loss<br />

by strategic combinations with mTOR, AKT, or MEK inhibitors<br />

Fu X, Kumar V, Shea M, Biswal NC, Nanda S, Chayanam S, Mitchell<br />

T, Hergenroeder G, Meerbrey KL, Joshi A, Westbrook TF, Mills GB,<br />

Creighton CJ, Hilsenbeck SG, Osborne CK, Schiff R. Lester & Sue<br />

Smith <strong>Breast</strong> Center, Baylor College of Medicine, Houston, TX; Dan<br />

L Duncan <strong>Cancer</strong> Center, Baylor College of Medicine, Houston, TX;<br />

Baylor College of Medicine, Houston, TX; M.D. Anderson <strong>Cancer</strong><br />

Center, Houston, TX<br />

Background: Hyperactive PI3K signaling is associated with a more<br />

aggressive subtype of estrogen receptor (ER) positive breast cancer<br />

(BC) and with endocrine resistance. Loss or downregulation of PI3K’s<br />

inhibitor PTEN is more common in basal and luminal B vs. luminal<br />

A BC. However, the role of PTEN in modulating response to various<br />

endocrine therapies is unclear. Here we investigated the effects of<br />

PTEN knockdown (KD) on endocrine sensitivity and the potential of<br />

multiple kinase inhibitors to restore and improve responses. Methods:<br />

Nude mice bearing ER+ BC xenograft tumors of MCF7 cells stably<br />

expressing a doxycycline (Dox)-inducible PTEN-shRNA were<br />

randomized to four endocrine treatment groups [continued estrogen<br />

(E2) supplementation, or E2-deprivation (ED) alone or in combination<br />

with tamoxifen (Tam) or fulvestrant (Ful)]; all -/+ Dox. The effects of<br />

single or combined kinase inhibitors on these endocrine treatments -/+<br />

Dox were studied in vitro using inhibitors (i) to mTOR (AZD2014,<br />

0.2 µM), AKT (AZD5363, 1 µM), or MEK (Selumetinib/ARRY-<br />

142886, 1 µM). Cell growth, apoptosis, and ER and progesterone<br />

receptor (PR) signaling were analyzed using cell cytometry, qRT/<br />

PCR, and Western blotting. Synergism tests were used to examine<br />

the growth effects of the most promising combinatorial therapy with<br />

multiple kinase inhibitors in different endocrine settings. Results: In<br />

wild-type (WT) PTEN xenograft tumors, endocrine therapies were<br />

very effective, inducing frequent tumor regression. In PTEN KD<br />

tumors endocrine therapies were less effective — PTEN KD delayed<br />

tumor regression in all endocrine regimens and accelerated tumor<br />

progression in the Tam treated group. Furthermore, at day 250, only<br />

1/8 and 0/7 tumors had developed resistance in the ED and the Ful<br />

(-Dox) groups, respectively, while with PTEN KD (+Dox), 7/15 and<br />

5/15 tumors developed resistance to ED and to Ful. In vitro PTEN<br />

KD also induced resistance to all endocrine therapies. mRNA and/<br />

or protein levels of ER and PR were suppressed by PTEN KD and<br />

restored by mTORi and AKTi. In cells with WT PTEN, mTORi was<br />

highly effective with or without endocrine therapy. However, AKTi<br />

and MEKi were more effective in combination with endocrine therapy.<br />

All three inhibitors were less effective upon PTEN KD. The mTORi<br />

plus AKTi combination resulted in a potent synergistic inhibition in<br />

PTEN KD cells in the presence of E2 or with ED. In contrast, in the<br />

presence of Tam, AKTi plus MEKi, independent of PTEN status, was<br />

the most effective combination at the doses chosen. Finally, these<br />

inhibitors and combinations were more effective in the presence of<br />

Ful than ED or Tam in WT PTEN cells. AKTi combined with Ful<br />

was still highly effective even in PTEN KD cells, but mTORi and<br />

MEKi were less effective. Conclusions: Our results suggest that PTEN<br />

loss renders endocrine therapy less effective in in vitro and in vivo<br />

experimental models. Single AKT/MEK kinase inhibitors are more<br />

potent in the presence of endocrine therapy. In PTEN KD cells, the<br />

activity of all three kinase inhibitors is largely diminished, except for<br />

AKTi in the presence of fulvestrant. Kinase inhibitor combinations<br />

are generally more effective, but the optimal combinations vary by<br />

PTEN status and type of endocrine therapy.<br />

PD01-02<br />

Increased expression of phosporylated mTOR in metastatic breast<br />

tumors compared to primary tumors in patients who received<br />

adjuvant endocrine therapy<br />

Hoefnagel LDC, Beelen KJ, Opdam M, Vincent A, Linn SC, vanDiest<br />

PJ. University Medical Center, Utrecht, Netherlands; The Netherlands<br />

<strong>Cancer</strong> Institute, Amsterdam, Netherlands<br />

BACKGROUND: Activation of the PI3K pathway as an adaptive<br />

change in response to estrogen depletion results in acquired endocrine<br />

therapy resistance in vitro. Metastatic breast cancer patients with<br />

previous exposure to endocrine therapy do derive substantial benefit<br />

from the addition of an mTOR inhibitor to endocrine therapy compared<br />

to endocrine therapy alone. This suggests that compensatory PI3K<br />

pathway activation might play a role in the development of clinical<br />

resistance to endocrine therapy. We evaluated the activation of the<br />

downstream PI3K pathway protein mTOR in primary breast tumors<br />

and matched metastatic lesions.<br />

METHODS: From a previously described series of breast cancer<br />

patients from whom both primary tumor tissue as well as metastatic<br />

tumor tissue was collected, we selected estrogen receptor α (ERα)<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

110s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Discussion 1<br />

positive patients who had received endocrine therapy (N=34) and<br />

who did not receive endocrine therapy (N=37). The difference in<br />

expression of phosphorylated mTOR (p-mTOR) between primary and<br />

metastatic tumor was assessed by immunohistochemistry, which was<br />

scored by using the proportion of tumor cells with sub-membranous<br />

p-mTOR expression. We assessed whether this p-mTOR difference<br />

between primary and metastatic tumor was associated with clinicopathological<br />

factors (location of metastasis, lymph node status,<br />

T-stage, grade, progesterone receptor status) or varied between<br />

patients who did and did not receive endocrine therapy, using Mann-<br />

Whitney tests. In addition we performed a linear regression model<br />

including the same clinico-pathological factors.<br />

RESULTS: In patients who had received endocrine therapy we<br />

observed an increase in p-mTOR expression in metastatic tumor<br />

lesions compared to the primary tumor (median difference 45%).<br />

This was significantly different from the mTOR changes observed<br />

in patients who did not receive endocrine therapy (median difference<br />

between metastatic and primary lesion 0 %) (p=0.003). The difference<br />

remained significant in the multivariate regression model (p=0.001).<br />

None of the other factors was significantly associated with a<br />

differential p-mTOR change.<br />

CONCLUSION: In patients who received previous adjuvant<br />

endocrine therapy, the increase in p-mTOR expression in metastatic<br />

tumor lesions compared to the primary tumor is significantly higher<br />

than in patients who did not receive endocrine therapy. Compensatory<br />

activation of the PI3K pathway might therefore be a clinically<br />

relevant resistance mechanism resulting in secondary endocrine<br />

therapy resistance.<br />

PD01-03<br />

Phosphorylated p-70S6K predicts tamoxifen resistance in<br />

postmenopausal breast cancer patients randomized between<br />

adjuvant tamoxifen versus no systemic treatment<br />

Beelen KJ, Opdam M, Severson TM, Koornstra RHT, Vincent A,<br />

Wesseling J, Muris JJ, Berns EMJJ, Vermorken JB, vanDiest PJ,<br />

Linn SC. The Netherlands <strong>Cancer</strong> Institute, Amsterdam, Netherlands;<br />

Erasmus University Medical Center-Daniel den Hoed <strong>Cancer</strong> Center,<br />

Rotterdam, Netherlands; Antwerp University hospital, Edegem,<br />

Belgium; University Medical Center, Utrecht, Netherlands<br />

BACKGROUND: Activation of the PI3K/MAPK pathways results<br />

in anti-estrogen resistance in vitro, however a biomarker that can<br />

predict clinical resistance has not yet been identified. Common<br />

drivers of these pathways are PIK3CA mutations, loss of PTEN<br />

and over-expression of HER2 and IGF-1R. We aimed to test the<br />

prognostic and predictive value of PI3K/MAPK pathway drivers<br />

as well as downstream activated proteins in postmenopausal breast<br />

cancer patients.<br />

METHODS: We collected primary tumor tissue from 563 ERα<br />

positive breast cancer patients who were randomized between<br />

tamoxifen (1-3 years) versus no adjuvant systemic therapy. PIK3CA<br />

hotspot mutations were assessed by Sequenom Mass Spectrometry.<br />

Immunohistochemistry was performed for expression of PTEN, IGF-<br />

1R and the downstream markers p-AKT(Thr308), p-AKT(Ser473),<br />

p-mTOR, p-p706SK and p-ERK1/2. Cox proportional hazard models<br />

for recurrence free interval were used to assess hazard ratios and<br />

interaction between these markers and treatment.<br />

RESULTS: No significant interaction between any of the tested PI3K/<br />

MAPK pathways drivers and tamoxifen was found. [/bold]However,<br />

interactions were identified between tamoxifen and the downstream<br />

activated proteins (p-AKT(Thr308), p- mTOR, p-p70S6K and<br />

p-ERK1/2). After correcting for multiple testing, p-p70S6K remained<br />

significantly associated with tamoxifen resistance. Patients whose<br />

tumor did not express p-p70S6K did derive significant benefit from<br />

tamoxifen (HR 0.24, 95 % CI= 0.12-0.47, p< 0.0001), while patients<br />

whose tumor did express p-p70S6K did not (multivariate HR=1.02,<br />

95% CI=0.48-2.21, p=0.95), p for interaction 0.003.<br />

CONCLUSION: We conclude that the downstream marker of PI3K/<br />

MAPK activation p-p70S6K predicts tamoxifen resistance in ERα<br />

positive postmenopausal breast cancer patients.<br />

PD01-04<br />

Deep kinome sequencing identifies a novel D189Y mutation in<br />

the Src family kinase LYN as a possible mediator of antiestrogen<br />

resistance in ER+ breast cancer<br />

Fox EM, Balko JM, Arteaga CL. Vanderbilt-Ingram <strong>Cancer</strong> Center,<br />

Vanderbilt University, Nashville, TN<br />

Estrogen receptor-positive (ER+) breast tumors adapt to hormone<br />

deprivation and acquire resistance to aromatase inhibitors (AIs). The<br />

proliferative rate of tumor cells measured by Ki67 after short-term<br />

treatment with an AI has been proposed as a surrogate endpoint<br />

of long term outcome. The purpose of this study was to identify<br />

kinase mutations associated with resistance to endocrine therapy<br />

as identified by a high Ki67 score after two weeks of pre-surgical<br />

therapy with letrozole. We performed deep-kinome sequencing on<br />

4 ER+/HER2– breast tumors that retained high Ki67 scores (14.8 to<br />

24.5%) following short-term letrozole treatment. Genomic DNA from<br />

the surgically excised tumors was deep sequenced using a capture<br />

approach with 120-bp biotinylated oligonucleotides hybridizing to<br />

612 genes, including 517 kinases with ≥300x coverage.<br />

All 4 tumors contained a ‘hot spot’ mutation in PIK3CA, the gene<br />

encoding the p110α catalytic subunit of PI3K. One tumor also<br />

contained a novel D189Y somatic mutation in LYN, a member of<br />

the Src family kinases (SFKs; variant frequency of 8%). D189Y<br />

(565G>T) is located in the LYN SH2 domain. Reverse-phase protein<br />

array (RPPA) analysis available in 10 tumors in this study revealed a<br />

significant correlation (p=0.006) between Y416 P-Src (which detects<br />

all SFKs) and a high post-letrozole Ki67 score. A siRNA screen<br />

targeting 779 kinases identified LYN as one of the top hits whose<br />

knockdown significantly reduced growth of MCF-7 breast cancer cells<br />

with acquired resistance to estrogen deprivation. We next analyzed<br />

the <strong>Cancer</strong> Genome Atlas (TCGA) SNP data to detect copy number<br />

changes for 444 tumors where corresponding gene expression data<br />

were available. Copy number increases (>0.8 log2 ratio over normal<br />

matched DNA) in LYN were present in approximately 10% of breast<br />

cancers, with the highest copy number gains observed in luminal B<br />

tumors. In contrast, other SFKs showed less frequent copy number<br />

increases (YES1


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

deprivation in a subset of ER+ breast cancers. Second, ER+ breast<br />

cancers harbor multiple molecular alterations capable of mediating<br />

hormone-independent growth.<br />

PD01-05<br />

Analysis of patients with ER-positive breast tumors treated with<br />

neoadjuvant aromatase inhibition identifies chemokine receptors<br />

as potential modulators of endocrine resistance<br />

Ribas R, Ghazoui Z, Dowsett M, Martin L-A. The Institute of <strong>Cancer</strong><br />

Research, London, United Kingdom; Royal Marsden Hospital,<br />

London, United Kingdom<br />

Background: Most breast cancers (BC) at primary diagnosis are<br />

estrogen receptor positive (ER+). Estrogen (E) mediates its effects<br />

by binding to the ER. Therapies targeting the estrogenic stimulation<br />

of tumor growth reduce mortality from ER+ BC. However, resistance<br />

remains a clinical problem. To identify molecular mechanisms<br />

associated with early resistance to E-deprivation, we overlaid global<br />

gene transcription data from patients treated with neoadjuvant<br />

anastrazole with those from MCF7 cells adapted to long-term<br />

E-deprivation (LTED).<br />

Methods: Global gene expression and Ki67 data were available<br />

from 81 postmenopausal women with ER+ early BC, at baseline and<br />

2-weeks post anastrazole treatment. Genes, that changed expression<br />

in response to anastrozole, were identified using multiple testing<br />

corrected multi-group analyses. In vitro global gene expression was<br />

available from MCF7 cells adapted to LTED. Functional consequence<br />

of target genes on proliferation, ER-mediated transcription and<br />

downstream cell signaling was assessed using siRNA.<br />

Results: Overlay of genes that predicted for a poor change in Ki67<br />

in patients treated with anastrazole (p


December 4-8, 2012 <strong>Abstract</strong>s: Poster Discussion 2<br />

PD01-07<br />

High throughput sequencing following cross-linked immuneprecipitation<br />

(HITS-CLIP) of Argonaute protein reveals novel<br />

miRNA regulatory pathways of Estrogen Receptor in breast<br />

cancer.<br />

Kabos P, Kline E, Brown J, Flory K, Sartorius C, Hesselberth J, Pillai<br />

MM. University of Colorado Denver, Aurora, CO<br />

Micro RNAs (miRNAs) are a class of small non-coding RNAs with<br />

well-known regulatory roles in normal physiological processes and<br />

cancer. Conventionally their study has been based on defining single<br />

miRNA-mRNA target interactions using a combination of miRNA<br />

expression arrays and bioinformatic predictions of binding to the 3’<br />

untranslated regions of genes, followed by miRNA over-expression<br />

in the relevant cell type. These approaches present circumstantial<br />

evidence for binding of a particular miRNA to its target but do not<br />

allow the study of global interactions nor provide direct evidence<br />

of binding. In order to study the genome wide impact of miRNA<br />

regulation in breast cancer we chose to use a recently described<br />

biochemical technique called Cross-Linked Immune-Precipitation of<br />

Argonaute (AGO) protein followed by high throughput sequencing<br />

(HITS-CLIP). Cross-linking of RNA to adjacent protein moieties by<br />

ultraviolet (UV) light allows for stringent isolation of the miRNAmRNA-AGO<br />

complexes by immune precipitation (IP); miRNAmRNA<br />

interaction within the RNA-induced silencing complex (RISC)<br />

is known to occur within the folds of AGO. The isolated miRNAmRNA-AGO<br />

complexes are then analyzed with next generation<br />

sequencing (NGS) to determine the miRNA-mRNA interactome.<br />

We performed HITS-CLIP on three well characterized breast cancer<br />

cell lines that represent ER+, Her2+ and triple negative (TN) disease<br />

(MCF7, BT474 and MDA231). To determine the role of miRNAs<br />

in coordinating the response to the estrogen receptor axis, MCF7<br />

and BT474 cells were analyzed with or without short term (24 hour)<br />

estrogen treatment.<br />

Analysis of sequencing data revealed several novel miRNA-mRNA<br />

interactions that target the ER pathway. For example, miR-9 directly<br />

regulates ER expression and miR-193a is involved in regulating<br />

expression of the ER co-activator NCOA3. We confirmed the<br />

biological relevance of these results using functional in vitro studies<br />

where manipulation of both miR-193a and miR-9 altered the<br />

responsiveness of breast cancer cells to tamoxifen. These results were<br />

further validated by quantitation of both transcript (RT-PCR) and<br />

protein (Western Blot) levels after transfection of miRNA precursors.<br />

Functional binding between these miRNAs and putative binding<br />

targets in the 3’ Untranslated Regions (3’UTRs) were also validated<br />

by luciferase-based reporter assays. Finally, we performed global<br />

analysis of miRNAs and their targets (as predicted by the HITS-CLIP<br />

datasets across all cell lines); this predicted regulation of core cellular<br />

processes such as cell proliferation, DNA repair and metabolism as<br />

being targeted by highly abundant miRNAs such as miR-27a and<br />

miR-21. In addition, our datasets confirmed previous reports of<br />

miRNA regulation of ER pathway such as miR-221 regulation of<br />

ER itself and miR-34a regulation of Myc. In summary, genome-wide<br />

biochemical approaches like HITS-CLIP allow for defining novel and<br />

clinically relevant miRNA-based regulatory pathways of endocrine<br />

responsiveness and resistance in breast cancer. In addition, breast<br />

cancer subtype specific biological pathways targeted by individual<br />

miRNAs can be predicted by this approach.<br />

PD01-08<br />

Differences in estrogen receptor signaling in non-malignant<br />

primary ER-positive breast epithelial cells and breast cancer.<br />

Lim E, He HH, Chi D, Yeung TY, Schnitt S, Liu SX, Garber J,<br />

Richardson A, Brown M. Dana-Farber <strong>Cancer</strong> Institute, Boston, MA;<br />

Harvard School of Public Health, Boston, MA; Harvard Medical<br />

School, Boston, MA; Beth Israel Deaconess Medical Center, Boston,<br />

MA; Brigham and Women’s Hospital, Boston, MA<br />

Background: The estrogen receptor (ER) is expressed in ∼70% of<br />

sporadic breast cancer and activates genes driving cell proliferation<br />

and tumorigenesis. We have previously performed genome-wide<br />

analysis of ER binding sites in MCF-7 breast cancer cells, and<br />

identified distinct mechanisms of ER signaling. We have also<br />

previously used EpCAM and CD49f as markers to enrich for viable<br />

ER-positive (ER+) cells obtained from non malignant breast tissue.<br />

Here, we seek to elucidate differences in ER signaling between nonmalignant<br />

and ER+ breast cancer cells.<br />

Methods: Primary breast epithelial cells were obtained from patients<br />

undergoing reduction mammoplasties and surgical excision of ER+<br />

breast cancer. After dissociation of breast reductions into a single-cell<br />

suspension, ER+ mature luminal (ML; EpCAM+CD49f-) and luminal<br />

progenitor (LP; EpCAM+CD49f+) subpopulations were obtained by<br />

flow cytometry. Following estrogen stimulation, RNA was extracted<br />

for gene microarray analysis. ER chromatin immunoprecipitation<br />

and DNA sequencing (ChIP-seq) was performed. These results were<br />

compared to MCF-7 breast cancer cells.<br />

Results: Reduction mammoplasty and ER+ breast cancer tissues were<br />

analyzed, and compared to MCF-7 cells. Gene expression profiles<br />

were different between non-malignant tissue and ER+ breast cancer<br />

cells following estrogen stimulation, with a 2-3 fold higher number<br />

of ER regulated genes in ER+ breast cancer compared to ER+ non<br />

malignant cells, and few overlapping estrogen regulated genes. Genes<br />

that promotes cell cycling and cell proliferation were downregulated<br />

in non-malignant tissue, but were upregulated in breast cancer cells<br />

(P < 10-5). CYP1A1, a major estradiol metabolizing enzyme, was<br />

upregulated in normal cells but downregulated in ER+ breast cancer<br />

cells. Motif analysis of ER ChIP-seq data in normal and ER+ breast<br />

cancer tissues demonstrated an enrichment of ER motifs in the<br />

overlapping sites and an enrichment of FOXA1 motifs in ER+ breast<br />

cancer cells and TCF12 motifs in non-malignant ER+ epithelial cells.<br />

Conclusions: There are contrasting differences in ER signaling<br />

between normal mammary and breast cancer cells, with estrogen<br />

having anti-proliferative effects in normal luminal cells compared to<br />

pro-proliferative effects in breast cancer. ER ChIP-Seq has identified<br />

TCF12 as a major co-factor in non-malignant breast tissue whilst<br />

FOXA1 is a major co-factor in ER+ breast cancer. Our data provides<br />

evidence for key alterations in ER-signaling during tumorigenesis, and<br />

identifies potential mechanisms to target cancer specific ER signaling.<br />

PD02-01<br />

Her2 expression measured by AQUA analysis on BCIRG-005 and<br />

BCIRG-006 predicts the benefit of Herceptin therapy<br />

Christiansen J, Barakat N, Murphy D, Rimm DL, Dabbas B,<br />

Nerenberg M, Beruti S, Quinaux E, Hall J, Press M, Slamon D.<br />

Genoptix Medical Laboratory; Yale University; IDDI; University of<br />

Southern California; University of California, Los Angeles<br />

Introduction: There have been disparate results reported in breast<br />

cancer testing for HER2 assessment as measured by protein<br />

expression or DNA amplification, yet both tests are routinely used<br />

to prescribe the drug Herceptin (trastuzumab, Genentech, So <strong>San</strong><br />

Francisco, CA). Typically, immunohistochemistry (IHC) staining<br />

www.aacrjournals.org 113s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

intensity of 3+ or FISH copy ratio of ≥2.0 are used to establish the<br />

cutoff between a negative and a positive result. However, it is unclear<br />

whether positivity is correlated with differential response to therapy.<br />

We used Automated Quantitative Analysis (AQUA) and a fluorescent<br />

immunohistochemical assay to measure HER2 expression in cases<br />

scored by central laboratory FISH and also receiving Herceptin<br />

therapy. The intentions of the study were two-fold: first, to provide<br />

further validation of the AQUA technology as applied to the clinical<br />

measurement of HER2 expression in breast cancer and second, to<br />

examine the potential of drug response stratification within those<br />

patients that are considered positive.<br />

Methods: AQUA fluorescence IHC staining was performed on a<br />

multi-cohort tissue microarray (TMA) set. The assay was constructed<br />

in the Genoptix CLIA laboratory per ASCO/CAP guidelines and<br />

with a cutpoint that was validated against IHC (with FISH reflex).<br />

The trial specimens tested were from the BCIRG-005/006 studies.<br />

BCIRG-005 had n=1544 cases all assessed as FISH- while the 006<br />

cohort had n=1477 cases all assessed as FISH+. Disease free survival<br />

(DFS) was used as the variable in subsequent modeling and analysis.<br />

Results: The BCIRG 005 and 006 cohorts, examined in aggregate,<br />

allowed for an initial examination of agreement relationships between<br />

HER2 levels as assessed by AQUA scoring and HER2 levels as<br />

assessed by central lab FISH. Results indicated a 77% negative<br />

agreement, a 97% positive agreement and an 87% overall concordance<br />

agreement for a total of n=3021 cases. Additional Cox modeling of<br />

the patients that were enrolled as FISH+ and stratified for those who<br />

did or did not receive Herceptin treatment demonstrated a significant<br />

overall hazard ratio (HR=0.75, CI=0.60,0.93) and when stratified for<br />

response to Herceptin, cases determined to be positive by AQUA<br />

showed significant benefit from treatment (HR=0.66, CI =0.52,0.85)<br />

in contrast to those who were scored as negative by AQUA that did<br />

demonstrate benefit from therapy (HR=1.19, CI=0.71,1.97).<br />

Conclusions: Analysis of the cases in this study originally determined<br />

to be HER2+ by FISH indicates that AQUA may improve predictions<br />

of which patients will benefit from Herceptin therapy.<br />

PD02-02<br />

The effect of HER2 expression on luminal A breast tumors<br />

Ellsworth RE, Valente AL, Shriver CD. Henry M. Jackson Foundation,<br />

Windber, PA; Windber Research Institute, Windber, PA; Walter Reed<br />

National Military Medical Center, Bethesda, MD<br />

Background: Luminal A (LumA) tumors are the most common<br />

subtype of breast cancer and have been associated with favorable<br />

prognosis. Although tumors can be considered LumA if they have<br />

>1% expression of ER, negative HER2 status ranges from 0+ (no<br />

expression) to 2+ (weak to moderate expression in >10% of cells)<br />

without concurrent genomic amplification. It is not clear how<br />

moderate levels of HER2 expression influence tumor biology in<br />

LumA tumors.<br />

Methods: The Clinical <strong>Breast</strong> Care Project database was queried<br />

to identify all hormone receptor positive cases with HER2 status<br />

defined as 0+ (n=89) or 2+/not amplified (n=154). Clinicopathological<br />

characteristics were compared between groups using chi-square<br />

analysis. Twenty-five 0+ and 25 2+ tumors were subjected to laser<br />

microdissection and hybridized to U133 2.0 microarrays (Affymetrix).<br />

Gene expression data was analyzed using Partek Genomics Suite.<br />

Results: Age at diagnosis did not differ between patients with no (58.5<br />

years) or moderate (59.3 years) HER2 expression. Patient groups did<br />

not differ by ethnicity, tumor stage, grade or size, or lymph node status.<br />

Although breast cancer mortality was low, all patients who died of<br />

disease (n=7) had moderate levels of HER2 expression (P=0.016). At<br />

the molecular level, four genes were differentially expressed (P2x-fold change): ESRRG expressed at significantly higher levels and<br />

DHRS2, SCUBE2 and SERPINA6 expressed at significantly lower<br />

levels in tumors with moderate HER2 expression.<br />

Discussion: LumA (2+) tumors are dissimilar from LumA (0+)<br />

tumors. Increased expression of ESRRG has been associated with<br />

increased cellular proliferation and tamoxifen resistance, while<br />

decreased expression of SCUBE2 and SERPINA6 have been<br />

associated with inhibition of growth and proliferation. Together,<br />

these molecular differences suggest that LumA tumors with moderate<br />

HER2 expression, although classified as HER2 negative, have more<br />

aggressive characteristics than those without HER2 expression<br />

which may contribute to the significantly higher mortality rate seen<br />

in patients with LumA tumors expressing HER2.<br />

PD02-03<br />

Added value of HER-2 amplification testing by multiplex ligationdependent<br />

probe amplification (MLPA)<br />

van Slooten H-J, Kuijpers CC, Moelans CB, Horstman A, Al-Janabi<br />

S, Hinrichs JW, van Diest PJ, Jiwa M. Symbiant Pathology Expert<br />

Centre, Alkmaar, Netherlands; University Medical Centre Utrecht,<br />

Netherlands<br />

Introduction<br />

Accurate determination of HER-2 status is critical for selection of<br />

breast cancer (BC) patients for trastuzumab treatment. According<br />

to the most commonly used testing protocol, patients with 3+<br />

immunohistochemistry (IHC) are considered for trastuzumab. In<br />

addition, 2+ IHC cases are tested for gene amplification and, when<br />

amplified, these patients are considered for trastuzumab treatment<br />

as well. Because in our experience IHC results have been proven to<br />

be sensitive to pre-analytical variation and inter- and intra-observer<br />

variation, we apply amplification testing not solely to 2+, but also to<br />

1+ and 3+ cases. At Symbiant Pathology Expert Centre, a validation<br />

study (n=99) comparing MLPA with chromogenic in situ hybridization<br />

(CISH) showed 100% concordance. Starting from April 2011, MLPA<br />

has been used as the method for HER-2 amplification testing, because<br />

in our hands this technique is more time- and cost-effective than<br />

CISH for analyzing large sample numbers. CISH was performed<br />

when MLPA was inconclusive, IHC score was heterogeneous or<br />

when invasive carcinoma could not be physically separated from<br />

DCIS. Here we report the results of a comparison between HER-2<br />

amplification tests and IHC in a cohort of 928 BC patients.<br />

Methods<br />

Pathology reports of 928 BC patients from 4 pathology laboratories<br />

were retrospectively reviewed. 728 patients were diagnosed between<br />

April 2011 and February 2012 in one of three laboratories of Symbiant<br />

Pathology Expert Centre. The other 200 patients were diagnosed<br />

in University Medical Centre Utrecht between January 2010 and<br />

December 2011. 18 patients had multiple tumors that were included<br />

as separate cases. In total, 949 tumors were reviewed.<br />

Results<br />

With IHC, major differences are observed between the 4 pathology<br />

laboratories concerning the percentage of cases scored as 0-1+ and 2+<br />

(Table 1). A considerable number of 3+ cases (6.4%) failed to show<br />

gene amplification by MLPA and/or CISH. A smaller percentage of<br />

0-1+ cases (1.8%) did show gene amplification (Table 2). 90 equivocal<br />

cases were double tested by CISH and MLPA. In 52/90 of these cases<br />

(58%) at least one test was inconclusive. The other 38 (42%) cases<br />

showed 100% concordance between MLPA and CISH.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

114s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Discussion 2<br />

Table 1. Comparison of HER-2 IHC scoring percentages between 4 pathology laboratories.<br />

Laboratory A (n=347) B (n=261) C (n=123) D (n=218) Total (n=949)<br />

0-1+ 58.2% 72.0% 70.7% 78.0% 68.2%<br />

2+ 30.5% 15.7% 16.3% 9.2% 19.7%<br />

3+ 11.2% 12.3% 13.0% 12.8% 12.1%<br />

Table 2. HER-2/neu gene amplification according to IHC score.<br />

No amplification Amplification Total<br />

0-1+ 484 (98.2%) 9 (1.8%) 493<br />

2+ 171 (91.9%) 15 (8.1%) 186<br />

3+ 7 (6.4%) 102 (93.6%) 109<br />

Total 662 (84.0%) 128 (16.0%) 788<br />

Conclusion<br />

MLPA is an accurate and cost-effective method for the determination<br />

of HER-2 status. We recommend the use of a gene amplification test<br />

to confirm HER-2 status in all IHC scores, not just the 2+ cases.<br />

However, it is still unclear whether the small subgroups of patients<br />

showing only HER-2 protein overexpression or only HER-2 gene<br />

amplification would benefit from trastuzumab. Follow-up data are<br />

therefore needed to determine the clinical value of HER-2 status<br />

assessed by each technique in order to improve the current standard<br />

of care for HER-2 testing.<br />

PD02-04<br />

Automated Quantitative RNA In Situ Hybridization for<br />

Resolution of Equivocal and Heterogeneous ERBB2 (HER2)<br />

Status in Invasive <strong>Breast</strong> Carcinoma<br />

Wang Z, Portier BP, Gruver AM, Bui S, Wang H, Su N, Vo H-T, Ma<br />

X-J, Luo Y, Budd GT, Tubbs RR. Cleveland Clinic, Cleveland, OH;<br />

Advanced Cell Diagnostics, Hayward, CA<br />

Background: <strong>Breast</strong> carcinomas that demonstrate a<br />

heterogeneous ERBB2 (HER2) status or equivocal results by both<br />

immunohistochemistry (IHC) and fluorescence in situ hybridization<br />

(FISH) present diagnostic challenges for which there is neither a<br />

standard methodology to achieve resolution in the clinical laboratory<br />

nor a uniform approach to management. We assessed the feasibility<br />

of using a novel automated and quantitative HER2 mRNA bright<br />

field in situ hybridization (ISH) assay capable of single molecule<br />

detection to determine HER2 status in invasive breast carcinomas that<br />

demonstrated significant tumor heterogeneity or failed to be resolved<br />

by standard IHC and FISH algorithmic testing.<br />

Design: Formalin-fixed, paraffin-embedded (FFPE) breast carcinomas<br />

from a non-consecutive series of 163 patients were analyzed for<br />

HER2 mRNA using a fully automated bright field RNA ISH assay<br />

(RNAscope, Advanced Cell Diagnostics, Hayward, CA). Cases were<br />

assigned into either a training set (n=34) or a validation set (n=129)<br />

and analyzed by both Q-RT-PCR and RNAscope. automated image<br />

analysis was used to numerate the punctate signal dots per cell in<br />

RNAscope-stained slides. A HER2 mRNA score based on single-cell<br />

quantification by RNAscope was developed and correlated to HER2<br />

FISH and HER2 mRNA Q-RT-PCR results. A simple cutoff value<br />

was derived using the training set and applied to the validation set.<br />

Results: Evaluable HER2 results were obtained for 154 cases (94.5%)<br />

by RNAscope and 163 cases (100%) by Q-RT-PCR. In the training<br />

set, both FISH/IHC positive and negative cases were definitively<br />

separated by both Q-RT-PCR and RNAscope. HER2 mRNA dots per<br />

cell correlated strongly to FISH (Spearman r=0.77) and Q-RT-PCR<br />

(r=0.81). Application of both methods to the validation set resulted<br />

in correct identification of 31/31 positive cases and 41/43 negative<br />

(overall concordance=97.3%) for both RNAscope and Q-RT-PCR.<br />

RNAscope showed a significant advantage over Q-RT-PCR in<br />

correctly identifying cases equivocal by FISH that were resolved<br />

by reflex IHC testing. RNAscope classified 7 of 26 (26.9%) FISH/<br />

IHC double equivocal cases as positives. In cases with HER2 protein<br />

heterogeneity, RNAscope showed a 100% concordance with FISH<br />

results, whereas Q-RT-PCR showed a 42.9% concordance.<br />

Conclusion: RNAscope analysis of HER2 mRNA is an effective<br />

means to resolve HER2 status in double equivocal cases and cases<br />

that demonstrate heterogeneity. Automation and image analysis-based<br />

quantification minimize analytical and post-analytical variability.<br />

Quantification of single RNA transcripts in situ at single-cell<br />

level demonstrates superiority over qRTPCR and great potential<br />

in predictive biomarker analysis. Further studies of larger cohorts<br />

correlating clinical response with HER2 mRNA expression in situ<br />

are warranted.<br />

PD02-05<br />

Comparison of fluorescence in situ hybridization (FISH) and<br />

dual-ISH (D-ISH) in the determination of HER2 status<br />

Mansfield AS, Sakai Y, Walsh FJ, Wiktor AE, Sukov WR, Dogan A,<br />

Jenkins RB. Mayo Clinic, Rochester, MN<br />

Introduction: The appropriate assessment of HER2 amplification<br />

status is critical to determining which patients should receive HER2-<br />

directed therapy. An alternative to FISH is D-ISH (Ventana Medical<br />

Systems Tucson, Arizona) which utilizes chromogenic HER2 and<br />

chromosome 17 probes. Both FISH and D-ISH utilize formalin-fixed,<br />

paraffin-embedded (FFPE) human breast cancer tissue specimens.<br />

D-ISH has two principle advantages over FISH: it can be visualized<br />

using light microscopy and after analysis the specimens can be<br />

archived.<br />

Methods: We tested 250 samples with FISH and with D-ISH. Fifty<br />

specimens were controls: 25 cases with no HER2 or CEP17anomaly<br />

and 25 cases with HER2 amplification. There were also 200 test<br />

subjects of 50 cases each of chromosome 17 aneusomy, HER2 deletion<br />

(HER2/CEP17 ratio


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

PD02-06<br />

Concordance between immunohistochemistry and<br />

FISH(fluorescence in sity hybridization) & SISH(silver in situ<br />

hybridization) for assessment of the HER2<br />

Lee M-R, Cho S-H, Kim D-C, Lee K-C, Lee J-H, Kwon H-C, Lee H-W,<br />

Lee S-e. Dong-A University Medical Center, Busan, Korea<br />

Goals: Accurate assessment of the human epidermal growth<br />

factor receptor 2 (HER2) of invasive breast cancer with<br />

Immunohistochemistry (IHC) and fluorescence in situ hybridization<br />

(FISH) & Silver in situ hybridization(SISH) is very important to<br />

decide treatment of targeted therapy with the humanized monoclonal<br />

antibody trastuzumab and the dual tyrosine kinase inhibitor lapatinib.<br />

The accuracy of these three testing methods can be adversely affected<br />

by various factors. We performed a retrospective analysis of the<br />

concordance of IHC and FISH & SISH analysis for HER2/neu at<br />

Donga-A Medical Center and reviewed possible causes of disparity<br />

between these two methods.<br />

Methods: We performed retrospective review of a total of 416<br />

invasive ductal carcinomas evaluated for HER2/neu at the Department<br />

of Surgery in Dong-A Medical Center between 2005 and 2011. We<br />

reviewed this data retrospectively and evaluated the concordance<br />

between IHC and FISH & SISH.<br />

Results: Comparison of the routinely assessed immunohistochemical<br />

HER2 status and FISH & SISH revealed that ten invasive carcinomas<br />

diagnosed immunohistochemically as highly overexpressed (score<br />

3+) did not show amplification of the HER2 gene by FISH n that<br />

these are false positives (SISH : false positive : 7 cases). Eight<br />

invasive carcinomas diagnosed immunohistochemically as Negatively<br />

expressed (score 0) showed amplification of the HER2 gene by SISH<br />

on that these are false negative. Fourteen tumor, which scored as 2+<br />

by IHC, was proven to be amplified on initial testing with FISH (SISH<br />

: 6 cases). Concordance between IHC and FISH analysis was good<br />

(171/181= 94.5%) (false positive : 5.5%) (SISH : 228/235=97.0%,<br />

false positive 3.4%, false negative 2.9%) and within the published<br />

range.<br />

Conclusion: IHC as first-line testing for HER2 may result in false<br />

positive and false negative results. The novel ASCO/CAP criteria<br />

for the immunohistochemical evaluation of the HER2 status may<br />

prevent some false positive results. The discordant cases between<br />

IHC and FISH & SISH represent a well-recognized subset of genetic<br />

heterogeneity in HER2/neu, which is known to contribute to positive<br />

IHC and negative FISH & SISH tests. The most powerful method for<br />

studying HER2 status is ISH. However, in heterogeneous carcinomas<br />

even FISH may not succeed in a correct evaluation of HER2.<br />

PD02-07<br />

Next-generation sequencing of FFPE breast cancers demonstrates<br />

high concordance with FISH in calling HER2 amplifications and<br />

commonly detects other clinically relevant genomic alterations<br />

Lipson D, He J, Yelensky R, Miller V, Sheehan C, Brennan K, Jarosz M,<br />

Stephens P, Cronin M, Ross J. Foundation Medicine, Inc, Cambridge,<br />

MA; Albany Medical College, Albany, NY<br />

Background: As more therapies targeting genomic alterations<br />

become available, next-generation sequencing (NGS) is increasingly<br />

performed in tumor types where mutational status may drive treatment<br />

choice. In addition to its ability to identify base substitutions,<br />

insertions and deletions across entire exons, NGS can detect relevant<br />

copy number changes such as amplification of HER2 in breast tumors.<br />

However, for NGS to be clinically applicable, it must reliably analyze<br />

FFPE tumor samples and show concordance with the best current<br />

diagnostic methods.<br />

Methods: To confirm a clinical role for NGS in detecting copy<br />

number alterations, we identified 35 FFPE invasive breast carcinomas<br />

previously tested for HER2 status by FISH, including 15 HER2<br />

positives (≥7 copies) and 20 HER2 negatives (900X uniquely-mapping reads was obtained. Sequence data were<br />

analyzed for HER2 copy number (blinded to FISH results) based on a<br />

statistical model using allele frequencies and coverage depth of HER2<br />

exons versus a process-matched normal control, classifying cases<br />

as HER2 positive (≥6 average copies), HER2 negative (27<br />

(Bonanni et al. JCO epub May 7, 2012). The objective of the current<br />

analysis was to determine whether metformin induced a modulation<br />

of apoptosis (TUNEL) overall and by HOMA index.<br />

TRIAL DESIGN: After tumor biopsy we randomly allocated 200<br />

non-diabetic women with operable breast cancer to either metformin<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

116s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Discussion 3<br />

(850 mg/bid) or placebo for 4 weeks prior to surgery. The primary<br />

outcome measure was the difference between arms in Ki-67 after 4<br />

weeks of treatment. Here we analyzed the apoptotic cell nuclei in 88<br />

consecutive core biopsies and their paired surgical samples from the<br />

initial 100 randomized subjects.<br />

RESULTS: Median TUNEL levels at surgery (Metformin = 10%, IQR,<br />

4-20, Placebo = 8%, IQR, 3-15) were significantly higher as compared<br />

with baseline (Metformin = 4%, IQR, 2-7, Placebo = 3%, IQR, 2-6,<br />

p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Serum Biomarker (unit) Pre-Metformin Mean (Range) Post-Metformin Mean (Range) P value<br />

Insulin (uIU/mL) 18.4 (4.5-72.0) 14.3 (4.1-34.4) 0.06<br />

Glucose (mg/dL) 93 (69 -121) 93 (72-127) 0.90<br />

HOMA 4.3 (0.7-18.8) 3.3 (0.9-8.8) 0.06<br />

Cholesterol (mg/dL) 203 (146-336) 186.6 (124-272)


December 4-8, 2012 <strong>Abstract</strong>s: Poster Discussion 3<br />

underway and will be reported. Integrated analysis of these factors<br />

combined with the physiological and molecular data described<br />

above will further enhance understanding of metformin action in<br />

the clinical setting.<br />

PD03-06<br />

Simvastatin radiosensitizes differentiated and stem-like breast<br />

cancer cell lines and is associated with improved local control<br />

in inflammatory breast cancer patients treated with postmastectomy<br />

radiation<br />

Lacerda L, Reddy J, Liu D, Larson R, Masuda H, Brewer T, Debeb<br />

B, Xu W, Hortobagyi GN, Buchholz TA, Ueno NT, Woodward WA.<br />

Morgan Welch Inflammatory <strong>Breast</strong> <strong>Cancer</strong> Research Program<br />

and Clinic, The University of Texas MD Anderson <strong>Cancer</strong> Center,<br />

Houston, TX<br />

Among women with non-inflammatory triple-negative and triplenegative<br />

inflammatory breast cancer (IBC), the 5-yr actuarial rates<br />

of local failure after radiation are 11%-35% and 45%, respectively,<br />

in part influenced by the contribution of radioresistant cancer stem<br />

cells to these cancers. Herein we explored the radiosensitization of<br />

breast cancer stem-like cells in vitro and examined the influence on<br />

local control after post-mastectomy radiation (PMRT) among IBC<br />

patients taking statins.<br />

SUM149, SUM159 and MCF-7 cells were cultured in standard<br />

monolayer cultures and stem cell enriching anchorage independent<br />

clonogenic cultures with simvastatin and treated with increasing<br />

concentrations of radiation. Survival curves were generated and<br />

t-test was used to compare surviving fraction (SF) of groups. p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Conclusions:<br />

HMGCR, the target of statin treatment, was up-regulated in breast<br />

cancer samples after atorvastatin treatment. This indicates HMGCR<br />

targeting within the tumor, which resulted in increased enzymatic<br />

activity. This study supports previous reports of statin-induced antiproliferative<br />

effects in breast cancer, and suggests that HMGCR could<br />

be used as a predictive marker for selection of breast cancer patients<br />

with beneficial anti-tumoral effects from statin treatment.<br />

PD03-08<br />

Statin use and improved survival outcome in primary<br />

inflammatory breast cancer: retrospective cohort study<br />

Brewer TM, Masuda H, Iwamoto T, Liu P, Kai K, Barnett CM,<br />

Woodward WA, Reuben JM, Yang P, Hortobagyi GN, Ueno NT. The<br />

University of Texas M.D. Anderson <strong>Cancer</strong> Center, Houston, TX;<br />

Eastern Virginia Medical School, Norfolk, VA; Okayama University<br />

Hospital, Okayama, Japan<br />

Background Inflammatory breast cancer (IBC) is the most aggressive<br />

type of breast cancer. HMG-CoA reductase inhibitors (statins)<br />

are cholesterol reducing agents with pleiotropic effects, including<br />

antitumorigenic and anti-inflammatory properties. We hypothesized<br />

that statins reduce the metastatic potential in primary IBC.<br />

Methods We retrospectively reviewed 993 patients diagnosed with<br />

and treated for IBC at The University of Texas MD Anderson <strong>Cancer</strong><br />

Center between Jan. 12, 1995 and Jan. 27, 2011. Patients with any<br />

records indicating statin use on the electronic medical record were<br />

compared with those without. We further compared clinical outcomes<br />

stratified by statin type (hydrophilic versus lipophilic). We used the<br />

Kaplan-Meier method to estimate the median disease-free survival<br />

(DFS) after surgery, overall survival (OS), and breast cancer-specific<br />

overall survival (BCOS), followed by Cox proportional hazards<br />

regression model to test the statistical significance of several potential<br />

prognostic factors.<br />

Results Primary IBC patients who used statins (n=109) were<br />

compared to those who did not (n=651). The median DFS was<br />

23.0 and 13.3 months (P


December 4-8, 2012 <strong>Abstract</strong>s: Poster Discussion 4<br />

Conclusion: Simvastatin was associated with a reduced risk of<br />

invasive breast cancer, and as a class, lipophilic statins were associated<br />

with a marginal benefit. This provides further evidence for possible<br />

class differences in statins with regard to chemo-preventive effects<br />

in breast cancer.<br />

PD04-01<br />

Intra-operative ultrasound in breast-conserving surgery for<br />

palpable breast cancer significantly improves both surgical<br />

accuracy and cosmetic outcome while saving costs.<br />

Haloua M, Krekel N, Coupe V, van den Tol P, Meijer S. VU Medical<br />

Center, Amsterdam, Nederland, Netherlands; Alkmaar Medical<br />

Center, Alkmaar, Noord-Holland, Netherlands<br />

Background: <strong>Breast</strong>-conserving surgery for palpable breast cancer is<br />

worldwide associated with a high rate of tumour-involved margins<br />

and excessive healthy tissue resection. This results in additional<br />

treatment and poor cosmetic outcome. Ultrasound-guided surgery<br />

(USS) may resolve both problems by improving surgical accuracy.<br />

A randomised controlled trial was initiated to compare USS with the<br />

standard palpation-guided surgery (PGS) for palpable breast cancer.<br />

Methods: A total of 134 eligible patients with palpable T1-T2 invasive<br />

breast cancer were randomised to either USS (n = 65) or PGS (n=69).<br />

Outcome measures included resection margin status, re-excision<br />

rates, mastectomy rates and additional radiotherapy. A calculated<br />

resection ratio (CRR) was derived from specimen volumes and tumour<br />

diameters, indicating healthy tissue resection. Secondary outcome<br />

measures were operative time, complications cosmetic outcome<br />

and cost-effectiveness. The costs of purchasing the US-system were<br />

included in the USS-group. The mean total costs per patient were<br />

calculated and compared between both study groups.<br />

Results: In the USS-group, 3.1% of margins were involved, compared<br />

with 17.4% in the PGS-group (P=0.009). The use of intra-operative<br />

US resulted in a significant reduction in additional therapies<br />

(P=0.015); in the PGS-group re-excisions were necessary in 3 patients<br />

(4.3%) and in 1 patient (1.5%) in the USS-group, mastectomies were<br />

performed in 5 patients (7.2%) of the PGS-group and in none of the<br />

patients of the USS-group, and additional radiotherapy boosts in 11<br />

patients (15.9%) of the PGS-group and 6 patients in the USS-group<br />

(9.2%). Excision volumes and CRR were both reduced with USS<br />

(38cc vs. 58cc and 1.0 vs. 1.7, respectively; both P23cm,<br />

so a propensity-score model was developed to balance cardiac and<br />

breast cancer risk factors between women who received AWBRT<br />

(42.5-44 Gy/16 fractions) versus CWBRT (45-50 Gy/25 fractions).<br />

The first event of a hospital admission for cardiac causes after RT<br />

was determined from hospitalization records. Cumulative incidence<br />

of cardiac morbidity was estimated at 10 years after RT for AWBRT<br />

versus CWBRT groups. Gray’s test was used to test for differences in<br />

the cumulative incidence curves between the AWBRT and CWBRT<br />

groups.<br />

RESULTS<br />

The median follow-up [range] was 13.2 [10-28] years. 485 women<br />

were treated with CWBRT and 2221 women were treated with<br />

AWBRT. The CWBRT group had a higher prevalence of diabetes,<br />

with a trend towards higher prevalence of HTN and other cardiac<br />

risk factors. Age, year of diagnosis, stage, grade, ER status, hormone<br />

and chemotherapy use were not statistically different between the RT<br />

groups. There was no statistically significant difference at 10 years<br />

in the incidence rate (as measured by first hospitalisation for cardiac<br />

morbidity) between the RT groups. The 10-year cumulative incidence<br />

of these hospitalization events for cardiac morbidity (95% CI) was<br />

20.8% (19.1-22.5) with AWBRT and 22.1% (18.5-25.9) with CWBRT.<br />

This difference was not significant between raw unadjusted values<br />

(p=0.07) or after propensity score adjustment with Gray’s test (p=0.2).<br />

Differences were also not statistically different at 15 years followup.<br />

These values are comparable with published long-term cardiac<br />

morbidity rates with CWBRT. Mortality due to cardiac cause was not<br />

statistically different between the two groups at 15 years follow-up.<br />

CONCLUSION<br />

Hospitalization for a primary or contributing cardiac cause at 15 years<br />

follow-up was not significantly different between women with leftsided<br />

early-stage breast cancer treated with AWBRT versus CWBRT<br />

even after correcting for pretreatment cardiac risk factors and events.<br />

www.aacrjournals.org 121s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

PD04-03<br />

Is breast conservation therapy an option for young women with<br />

operable breast cancer? Local recurrence rates in young women<br />

following surgery: a single centre experience.<br />

Lewis CR, Smith R, Matthews A, Choo E, Lee C. Prince of Wales<br />

<strong>Cancer</strong> Centre (POWCC), Randwick, NSW, Australia; Prince of<br />

Wales Hospital, Randwick, NSW, Australia; NHMRC Clinical Trials<br />

Unit, University of Sydney, NSW, Australia<br />

Background: <strong>Breast</strong> cancer (BC) is less common in young women<br />

(defined here as ≤ 40 years age), but is associated with more aggressive<br />

biological features, higher risk of local recurrence (LR) and poorer<br />

overall survival. This study examines and compares the incidence of<br />

LR following breast conservation therapy (BCT) versus mastectomy<br />

in women with operable BC treated at our centre.<br />

Methods: The POWCC breast cancer database was retrospectively<br />

reviewed for the period January 1995 to December 2008. 2250 eligible<br />

women with BC undergoing primary breast surgery were identified.<br />

LR rate was compared between young women and older women (age<br />

> 40 years), and according to type of surgery. Data were analysed<br />

using a competing risk Cox model to account for distant recurrence<br />

and death as competing events for local recurrence.<br />

Results: Median follow-up was 70 months. Of 2250 women, 246<br />

(11%) were young women, and the mastectomy rate was 49.2%. In<br />

older women (89%), mastectomy rate was 41.7%. LR occurred in<br />

17 (6.9%) and 57 (2.8%) in young and older women respectively<br />

(p=0.001). Amongst the young women, 12 (9.6%) and 5 (4.1%)<br />

patients recurred locally in BCT and mastectomy respectively<br />

(p=0.09). Amongst the older women, 43 (3.7%) and 14 (1.7%)<br />

patients recurred locally in BCT and mastectomy respectively<br />

(p=0.008). In univariate Cox analysis, significant risk factors for LR<br />

were BCT (p=0.003), positive surgical margins (p=0.03), age ≤ 40<br />

years (p=0.001), premenopausal status (p=0.003) and no adjuvant<br />

systemic therapy (0.02). Age remains a significant predictor of LR in<br />

multivariate Cox analysis (Table). There was no significant interaction<br />

between age and type of surgery on LR (p=0.72).<br />

Discussion: Our results demonstrate that young women who undergo<br />

BCT have the highest risk of early LR. Adjuvant systemic therapy<br />

is protective of early LR. This study is hypothesis-generating and a<br />

definitive prospective clinical trial is required to better determine the<br />

optimal type of breast surgery in young women.<br />

Multivariate Cox analysis: risk of local recurrence<br />

Characteristics HR 95% CI p-value<br />

Age > 40 years 0.48 0.25-0.92 0.03<br />

Mastectomy vs BCT 0.43 0.25-0.76 0.004<br />

Postmenopausal 0.55 0.32-0.94 0.03<br />

Systemic therapy (hormone<br />

+/- chemo) vs none<br />

0.53 0.33-0.84 0.007<br />

PD04-04<br />

What is influencing breast conservation rates in the United States?<br />

Data from the National Accreditation Program of <strong>Breast</strong> Centers<br />

Chagpar AB, Kaufman CS, Connolly J, Burgin C, Granville T,<br />

Winchester D. Yale University School of Medicine, New Haven, CT;<br />

Bellingham <strong>Breast</strong> Center; Beth Israel Deaconess, Boston, MA;<br />

National Accreditation Center for <strong>Breast</strong> Centers; Pat Nolan Center<br />

for <strong>Breast</strong> Health, Glenview, IL<br />

INTRODUCTION: <strong>Breast</strong> conservation (BC) rates have been utilized<br />

as a quality metric, yet mastectomy rates have been noted to be<br />

increasing nationwide. The purpose of this study was to evaluate the<br />

factors associated with BC in 437 US breast centers surveyed by the<br />

National Accreditation Program of <strong>Breast</strong> Centers (NAPBC).<br />

METHODS: From 2006 to 2010, 437 breast centers across the US<br />

were surveyed by the NAPBC. At each center, annual data regarding<br />

BC rates were reported in the Survey Application Record (SAR) for<br />

patients with Stage 0-II breast cancer. We evaluated characteristics<br />

of breast centers including geographic location, volume of patients<br />

treated, therapeutic modalities provided, and center organizational<br />

relationships. Non-parametric statistical analyses were performed to<br />

determine factors associated with BC rates in this cohort.<br />

RESULTS: Among the 437 breast centers surveyed from 2006-2011,<br />

data on 77,248 patients with Stage 0-II were reported. The median<br />

number of Stage 0-II breast cancer patients per center was 149 (range;<br />

10-891). The median proportion of these patients who underwent BC<br />

was 64.8% (range; 33-100%). No significant differences in median BC<br />

rate was noted over the six years of the study (range; 64.2% in 2006<br />

– 67.1% in 2011, p=0.788). However, significant regional variations<br />

were noted in median BC rates: 62.2% in the West (N=59 centers),<br />

63.8% in the South (N=145 centers), 64.1% in the Midwest (N=118<br />

centers), and 70.8% in the Northeast (N=104 centers), p


December 4-8, 2012 <strong>Abstract</strong>s: Poster Discussion 4<br />

was performed with the Nanostring nCounter® system. Data<br />

were normalized using the nCounter® digital analyzer software for<br />

analysis.<br />

Results: Among the 108 studied cases, 66 (61%) recurred as DCIS and<br />

42 (39%) recurred as invasive cancer. Median time to recurrence was<br />

40 months (range 7-156 mo) and did not differ by type of recurrence.<br />

Similarly, patient age, grade of DCIS, ER status, use of radiation or<br />

tamoxifen therapy did not differ. Unsupervised hierarchical clustering<br />

using all 32 genes demonstrated two predominant groups, one<br />

enriched for recurrent-as-invasive (RI) tumors, and the other enriched<br />

for recurrent-as-DCIS (RD) tumors. Refined analysis by selection<br />

of 14 genes with significant differential expression between the RI<br />

and the RD tumors identified four main groups. The first cluster was<br />

dominated by ERBB2 over-expression and is equally composed of RI<br />

and RD tumors. The second cluster was composed almost exclusively<br />

of RI tumors and showed high level of COX2 and CCND1 and low<br />

level of CDKN2A (p16). The clusters 3 and 4 were enriched for RD<br />

tumors, with the fourth cluster completely devoid of RI cases. This<br />

RD-only group showed the lowest level of CCND1, but highest level<br />

of AKT3, EGFR, CDKN2A, and MKI67, thus displaying molecular<br />

traits typical of basal-like tumors.<br />

Conclusion: In this cohort of patients who recurred following<br />

conservative treatment for DCIS, HER2 expression was not<br />

predictive, however gene expression analysis demonstrated that high<br />

COX2 and CCND1 and low CDKN2a (p16) levels were strongly<br />

associated with invasive recurrences (RI), whereas increased AKT3<br />

and MKI67 were associated with non-invasive recurrences (RD).<br />

Although the optimal combination of predictive markers requires<br />

validation, these data suggest that molecular alterations associated<br />

with RI differ from those associated with RD and warrant further<br />

investigation.<br />

PD04-06<br />

Molecular Phenotypes of DCIS predict Invasive and DCIS<br />

recurrence<br />

Williams KE, Barnes NLP, Cheema K, Dimopoulos N, Bundred NJ,<br />

Landberg G. The University of Manchester, Manchester, Greater<br />

Manchester, United Kingdom<br />

Introduction: Molecular phenotypes of invasive breast cancer predict<br />

early recurrence and survival. DCIS exhibits similar phenotypes<br />

but their frequency and clinical significance remain uncertain. To<br />

determine whether molecular phenotypes of DCIS predict recurrence,<br />

273 women (median age 57 years) with primary DCIS who were<br />

screened for or entered DCIS trials (Iressa/Lapatinib/ERISAC) in<br />

one unit from 1990-2010 were studied.<br />

Methods: HER2, oestrogen receptor (ER) and progesterone receptor<br />

(PR) expression within primary DCIS were established using<br />

immunohistochemistry within the trial protocols. HER2 was scored 0<br />

(absent) to 3 (maximum). Scores ≥2 were taken as positive if amplified<br />

on FISH testing. ER and PR scored positive if ≥5% of cells stained.<br />

64.2% patients were ER positive, 43.3% were HER2 positive and<br />

31.8% were high-grade lesions. 94 underwent mastectomy whilst<br />

185 had BCS.<br />

Results: There was an overall recurrence rate of 20.14% after a<br />

median follow-up period of 74 months (range 12-240). Of these<br />

recurrences, 36.4% were invasive. Conservation surgery (BCS)<br />

was used in 185 women who suffered 47 recurrences. Molecular<br />

phenotype predicted local recurrence by Log Rank analysis (P


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

respectively. The HR RT vs No RT for Triple Negative subtype was 0.40<br />

(95 % CI, 0.07-2.41) and was not adjusted because of the sparse<br />

number of events. Finally, we focused the analysis on DIN2 patients<br />

stratified by Ki-67 LI. Again, after adjustment for menopause, surgical<br />

margins, presence of necrosis, microcalcifications, and HT, RT was<br />

not effective in DIN2 patients with Ki-67 LI 500X) All classes of genomic alterations were detected.<br />

RESULTS: The site distribution and frequency of disease recurrence<br />

included: soft tissue (19) lung (4), bone and liver (3), and brain (2).<br />

The disease subtype classification based on the expression of ER,<br />

PR and HER-2 were as follow: ER-, PR-, HER-2-, Basal-like or TN<br />

(65%); ER+ and/or PR+, HER-2 -(grade3) or HER-2+, Luminal B<br />

(22%); ER-, PR-, HER2+ (13%). Genomic alterations were identified<br />

in all patients with various frequencies. Genetic mutations included:<br />

P53 (DNA-binding domain, splice site), PI3KCA (H1047R, R441N,<br />

E545K, K111E,); RB1; PTEN (D107Y); EGFR (L858R); ERBB2<br />

(V777L, S310F) in a patient with TN disease; ESR1 D538G. Gene<br />

amplifications included: MYC (26%); CCND1 (22%); ERBB2 (in<br />

a patient with TN disease), MLC-1, CCNE1, CK4, K-RAS. One<br />

patient had evidence of discordant genomic abnormalities in two<br />

simultaneous sites of recurrence (skin and pleura) suggesting disease<br />

heterogeneity. Three patients were effectively treated with genomic<br />

alterations-guided therapies.<br />

CONCLUSIONS: Genomic sequencing of advanced IBC is<br />

feasible, identifies potential therapeutic targets and can advance our<br />

understanding of the molecular alterations of this aggressive disease.<br />

PD05-02<br />

Novel mutations in lobular carcinoma in situ (LCIS) as uncovered<br />

by targeted parallel sequencing<br />

De Brot M, Andrade VP, Morrogh M, Berger MF, Won HH, Koslow<br />

Mautner S, Qin L-X, Giri DD, Olvera N, Sakr RA, King TA. Memorial<br />

Sloan-Kettering <strong>Cancer</strong> Center, New York, NY<br />

Background<br />

LCIS has traditionally been recognized as a marker of increased risk<br />

for the subsequent development of breast cancer, of either the lobular<br />

or ductal phenotype, yet due to the incidental nature of LCIS little is<br />

known about its underlying biology. Here we describe the first report<br />

of novel mutations in LCIS using targeted exome sequencing of fresh<br />

frozen tissue samples.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

124s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Discussion 5<br />

Methods<br />

Fresh frozen tissue samples from patients with a prior history of LCIS<br />

undergoing therapeutic or risk-reducing mastectomy from 2003-08<br />

were harvested and systematically reviewed to identify LCIS. Cells<br />

from individual LCIS lesions +/- associated cancers were collected<br />

by laser capture microdissection. For the purposes of this study,<br />

germline DNA (blood) and DNA from 12 unique LCIS lesions were<br />

subject to targeted parallel sequencing of exons corresponding to 230<br />

cancer genes using the Illumina HiSeq 2000 platform. DNA from<br />

an associated ductal carcinoma in situ (DCIS) and/or an invasive<br />

ductal (IDC) or lobular (ILC) lesion was also available for 7 of these<br />

cases resulting in 41 samples from 12 pts for mutational analysis.<br />

Normalized (RMA) Affymetrix U133A gene expression data were<br />

also available.<br />

Results<br />

DNA profiling reliably identified 7 somatic mutations in 5/12 LCIS<br />

samples (41.7%). Of these, 4/7 mutations were base substitutions<br />

(missense mutations); and the others included: 1 deletion; 1 silent<br />

and 1 splice-site mutation. Mutations in LCIS were identified in<br />

5/230 cancer genes analyzed, including: PIK3CA, CDH1, NOTCH4,<br />

PREX2 and ARAF. PIK3CA and CDH1 mutations were each<br />

identified in two samples, representing 4/7 (57.1%) mutations.<br />

Specific mutations found in LCIS and their frequencies are listed<br />

(table). Among 3 LCIS-ILC pairs, one shared the G914R mutation<br />

in PIK3CA, and 1/3 LCIS-IDC pairs exhibited an identical point<br />

mutation (R373W) in the NOTCH4 gene. No shared mutations were<br />

observed in 3 LCIS-DCIS pairs. Both CDH1 mutated cases were<br />

associated with decreased e-cadherin mRNA levels when compared<br />

to non-mutated cases (mean 9.88 vs 11.01), as was the NOTCH4<br />

mutation (mean 6.02 vs 6.47). Mutations in ARAF and PREX2 were<br />

associated with increased mRNA levels, mean 7.07 vs 6.52 and 4.82<br />

vs 4.22, respectively. The hotspot PIK3CA mutation (E545K) was<br />

also associated with increased gene expression (mean 5.15 vs 4.64)<br />

whereas the G914R mutant was associated with decreased expression<br />

(mean 4.13 vs 4.64).<br />

LCIS<br />

PIK3CA<br />

Sample<br />

CDH1 NOTCH4 PREX2 ARAF Shared Mutation<br />

1 - - - -<br />

Q167Q<br />

10.4% -<br />

2 - I178_splice 9.7% R373W 6.7% - - R373W IDC 10.7%<br />

3 - - - - - -<br />

4 E545K 7.2% - - - - -<br />

5 - - - - - -<br />

6 - - - - - -<br />

7 G914R 17.8% - - - - G914R ILC 29.6%<br />

8 - - - - - -<br />

9 - Indel 19.1% -<br />

G959D<br />

15.8%<br />

- -<br />

10 - - - - - -<br />

11 - - - - - -<br />

12 - - - - - -<br />

Conclusions<br />

This study represents the first targeted exon sequencing analysis of<br />

fresh frozen LCIS. Although LCIS has been regarded as a rather<br />

genetically stable lesion, somatic mutations were detected in 41.7%<br />

of lesions in this small cohort. While CDH1 mutations are expected<br />

in lobular neoplasia, this is the first report of mutations in ARAF,<br />

NOTCH4, PIK3CA and PREX2. Given the shared relevance of<br />

PIK3CA and PREX2 in the PI3K/AKT pathway, these findings<br />

suggest novel mechanisms for new chemoprevention strategies among<br />

women with LCIS.<br />

PD05-03<br />

What is the appropriate sample (s) on which to perform<br />

sequencing for mutational analysis to guide the selection of<br />

targeted therapy?<br />

Alpaugh RK, Bingham C, Fittipaldi P, Banzi M, Palmer G, Cristofanilli<br />

M. Fox Chase <strong>Cancer</strong> Center, Philadelphia, PA; Silicon Biosystems,<br />

Spa, Bologna, Italy; Foundation Medicine, Cambridge, MA<br />

BACKGROUND: Success of targeted therapy requires expression<br />

of the protein. Tumor tissue source can include diagnostic biopsy,<br />

surgical samples from initial or follow-up surgeries, fluids e.g. pleural<br />

or ascites and circulating tumor cells (CTC). The goal of using CTCs<br />

was 1. To determine whether CTC can be used as a “liquid” tumor<br />

biopsy and enable gene sequence information at the single cell level<br />

and 2. To determine the heterogeneity represented in the circulation<br />

compared to that seen in solid tumor by examining single cells (or a<br />

small cluster of cells) for the presence of a specific mutation which<br />

was detected in tissue tumor source.<br />

METHODS: We performed sequencing for mutational analysis on<br />

tissue(s) from patients with inflammatory breast cancer (IBC). Tumor<br />

sources varied from mastectomy tissue, metastatic site(s) e.g. liver or<br />

skin from chest wall disease, pleural fluid and CTC isolated into pure<br />

single cell populations (or groups of cells) using Silicon Biosystems<br />

DEPArray. Ampli1 WGA kit was used for CTC amplification. Of<br />

the 22 patients sequenced, mastectomy primary tumor was examined<br />

in 3, metastatic site skin chest wall disease in 15, other metastatic<br />

site in 4, pleural fluid in 2 and CTC collected to investigate p53<br />

mutations in 8.<br />

RESULTS: To date 22 patients have had mutational data performed,<br />

14/22 had mutations in p53, 4/22 in RB1, 2/22 in each PI3K and<br />

ERBB2, 1/22 in each of BRAC1, BRAC2, ATM, KRAS, Notch 1,<br />

MEN1 and ESR1. Numerous amplifications were noted including<br />

AKT1, RPTOR, MLC1, MYC, CCND1 and ERBB2. For one patient’s<br />

chest wall biopsy compared to two single CTCs and a cluster of 10<br />

CTCs the same TP53C229fs*10 mutation was detected revealing<br />

the same 2bp deletion. No 2bp deletion was found in single white<br />

blood cells. Whereas, another patient which showed a TP53 S215G<br />

mutation in her skin biopsy of chest wall disease, only amplifications<br />

of AURKA, CCND1, IGF1R, MDM2 and SRC in pleural tumor<br />

cells were detected and no mutations in three single CTC, two single<br />

pleural tumor cells and in single white blood cells were seen. Primary<br />

tumor tissue is being sort for both of these patients. Mutational data<br />

reviewed to date suggest that IBC is not one disease but a multiplicity<br />

of diseases, revealing a variety of target(s). Aberrations are not<br />

consistent across tissue source.<br />

CONCLUSIONS: Successful treatment outcomes using standard of<br />

care chemotherapy combined with target therapies will require not<br />

one, but a panel, of tissue sources for sequencing to guide the selection<br />

of appropriate targeted therapies.<br />

PD05-04<br />

Targeted resequencing in oncogenetics : developing a new<br />

approach for molecular diagnostics<br />

Sevenet N, Lafon D, Dupiot-Chiron J, Hubert C, Jones N, Debled M,<br />

Tunon de Lara C, Longy M, Bonnet F. Institut Bergonie, Bordeaux,<br />

France; Centre de Génomique Foncionnelle de Bordeaux, Bordeaux,<br />

France; Institut Bergonie & Universite Bordeaux Segalen, Bordeaux,<br />

France<br />

Introduction<br />

Next-generation sequencing (NGS) appears to be an accurate tool<br />

adapted for molecular oncogenetics. In contrast to certain somatic<br />

mutations, a lack of germline mutation hotspots in genes involved<br />

www.aacrjournals.org 125s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

in cancer predisposition syndromes, especially hereditary breast and<br />

ovarian cancer (HBOC), force the molecular biologist to screen the<br />

entire coding sequence.<br />

Material & methods<br />

In order to screen a panel of genes predominantly involved in HBOC<br />

& Cowden disease in a molecular diagnostic setting, we captured 408<br />

exons (and their surrounding intronic sequences) of 25 genes using<br />

the SureSelect oligonucleotide library capture (Agilent technologies).<br />

The bioinformatics resources Earray (Agilent technologies), Ensembl<br />

human genome v62 and UCSC repeat masker were used to create the<br />

capture design. This gave us 2899 unique 120-mers oligonucleotide<br />

probes which were duplicated 19 to 38 times depending on the<br />

GC content or the size of the targeted regions. Germline DNA<br />

of 190 patients (40 with previously identified mutations and 150<br />

without previous molecular genetic analysis) was sequenced using<br />

a MiSeq bench top next-generation sequencer (Illumina) and in<br />

parallel was analyzed using our current screening method (EMMA,<br />

Enhanced Mismatch Mutation Analysis, similar to high resolution<br />

melting). Samples were prepared according the manufacturer’s<br />

recommendations (Agilent technologies).<br />

Results<br />

All previously identified mutations along with 33 mutations in<br />

previously unscreened patients were detected. In addition, more than<br />

five hundred variants described as polymorphisms were identified.<br />

NGS seems to be more sensitive than our current screening method<br />

due to its detection and identification of homozygous variants. In<br />

order to limit the interpretation time, we restricted sequence analysis<br />

to the exon, -50 bp (including the acceptor splice site of the upstream<br />

intron) and +50 bp (including the donor splice site of the downstream<br />

intron). We have validated NGS as a technique adapted for germline<br />

mutation screening in oncogenetics. From the beginning of 2013, all<br />

germline DNA samples will be screened by NGS for oncogenetic<br />

molecular diagnostics. Sample preparation, technical organization,<br />

bioinformatics workflow and comprehensive results will be presented.<br />

PD05-05<br />

RNA-seq identifies unique transcriptomic changes after brief<br />

exposure to preoperative nab-paclitaxel (N), bevacizumab (B) or<br />

trastuzumab (T) and reveals down-regulation of TGF-β signaling<br />

associated with response to bevacizumab<br />

Varadan V, Kamalakaran S, Janevski A, Banerjee N, Lezon-Geyda K,<br />

Miskimen K, Bossuyt V, Abu-Khalaf M, Sikov W, Dimitrova N, Harris<br />

LN. Philips Research North America, Briarcliff Manor, NY; Yale<br />

University School of Medicine, New Haven, CT; Yale Comprehensive<br />

<strong>Cancer</strong> Center, New Haven, CT; Yale University School of Medicine,<br />

Yale Comprehensive <strong>Cancer</strong> Center, New Haven, CT; Warren Alpert<br />

Medical School of Brown University, New Haven, CT; Seidman<br />

<strong>Cancer</strong> Center, Cleveland, OH<br />

Background: Identification of differentially expressed transcripts<br />

upon brief exposure to preoperative therapy can help determine likely<br />

response markers. We quantify and compare differential transcript<br />

expression using RNA-seq on patient samples before and after one<br />

dose of T or B or N. We also evaluate correlation of brief-exposure<br />

transcriptomic changes with response to B.<br />

Methods: We sequenced transcriptomes of core biopsy RNA from<br />

50 pairs of breast tumors obtained from neoadjuvant clinical trials<br />

BrUOG 211A/211B. Patients were given a run-in dose of B or N or<br />

T, followed by combination biologic/chemotherapy (HER2- with B/<br />

carboplatin/N; HER2+ with T/carboplatin/N). We sequenced biopsy<br />

pairs obtained pre/post 10 day exposure to run-in monotherapy.<br />

Paired-end sequencing was done on Illumina GAII platform using<br />

amplified total RNA with 74bp read length, yielding expression data<br />

for 22,302 genes and 35,768 transcripts. We evaluated transcriptomic<br />

changes upon brief exposure to monotherapy assuming Poissondistributed<br />

read-counts, followed by multiple testing correction<br />

and enrichment analysis of 185 KEGG pathways. We investigated<br />

association of transcriptomic changes upon brief exposure and<br />

pathologic complete response (pCR) in the B arm. Differential<br />

expression of previously published signatures of tumor vasculature,<br />

TGF-β, β-catenin, MYC, E2F3 and RAF-MEK pathway activities<br />

were evaluated to identify associations with pCR.<br />

Results: PAM50-based clustering showed individual samples<br />

cluster together, demonstrating that tumor subtypes do not change<br />

over the 10-day treatment. We identified unique transcripts that<br />

were significantly differentially expressed in each therapy arm<br />

(p


December 4-8, 2012 <strong>Abstract</strong>s: Poster Discussion 5<br />

combined with RNA-seq data from an additional 77 TNBCs and 10<br />

adjacent normal tissues from the TCGA Data Portal. The merged<br />

gene expression values (RPKM) were quantile normalized, batch<br />

effect corrected, and submitted for differential gene expression<br />

analysis using Partek Genomics Suite. Differentially expressed<br />

genes were statistically determined with cutoffs of FDR±2. Pathway and network analyses utilized Ingenuity<br />

Pathway Analysis.<br />

Results: Principal components analysis (PCA) of all expressed<br />

protein coding genes revealed a close association of adjacent normal<br />

tissues to TNBCs whereas microdissected normal breast tissues were<br />

starkly separated. In comparing these 3 tissue types, 3841 genes were<br />

differentially expressed between TNBC vs. donated microdissected<br />

normal of which 1627 genes were also differentially expressed<br />

between adjacent normal vs. donated microdissected normal. The PCA<br />

along with the differential expression support the notion that adjacent<br />

normal gene expression is strongly influenced by the primary tumor,<br />

and in a significant set of genes, adjacent normal gene expression<br />

mimics the expression of primary tumors. The differential gene<br />

expression analysis also revealed 1240 genes that were restricted<br />

solely to the TNBC vs. donated microdissected normal that were not<br />

detected when TNBCs were compared to adjacent normal tissues.<br />

Examination of these genes reveals several potential kinase drug<br />

targets, genes involved in notch signaling, metabolism, cell cycle<br />

among other key cancer pathways. Preliminary analysis of the noncoding<br />

RNA component also reveals the same pattern of dysregulation<br />

in cancer associated microRNAs and lincRNAs.<br />

Conclusions: Gene expression within adjacent normal tissues<br />

is significantly impacted by the primary tumor and donated<br />

microdissected normal breast epithelium from healthy volunteers may<br />

be an optimal comparator for use in next-generation RNA sequencing<br />

breast cancer studies. Use of this comparator may identify therapeutic<br />

targets that would not otherwise be detected when using adjacent<br />

normal tissue as controls.<br />

PD05-07<br />

Detection of fusion transcripts among 100 breast cancer samples<br />

by next generation sequencing<br />

Kim J, Han W, Moon H-G, Ahn SK, In Y-H, Kim S, Lee H-S, Lee JW,<br />

Kim JY, Kim T, Kim MK, Noh D-Y. Seoul National University Hospital,<br />

Seoul, Korea; <strong>Cancer</strong> research institute, Seoul National University,<br />

Seoul, Korea; i-Pharm (Information Center for Bio-pharmacological<br />

Network) IBBI (Integrated Bioscience and Biotechnology Institute)<br />

Advanced Institute of Convergence Technology, Seoul National<br />

University, Seoul, Korea; Seoul National University, Seoul, Korea;<br />

Macrogen Inc., Seoul, Korea<br />

Introduction Fusions genes and it’s products are widely applied as<br />

biomarker and therapeutic target in hematopoietic cancers. It’s role<br />

as a “driver” of oncogenic pathway have also been proved in several<br />

epithelial solid cancers including prostate, lung and breast cancer.<br />

We performed high throughput next generation sequencing technique<br />

(RNA-Seq) to identify the novel fusion transcript(FT) in a large set<br />

of primary breast cancer samples.<br />

Materials and Method Total RNA was prepared using the Illumina®<br />

TrueSeq RNA sample Preparation Kit and TrueSeq mRNA library<br />

was constructed. Clustering was done with HiSeq 2000 and Solexa’s<br />

sequencing was performed. We used tissues extracted from previously<br />

collected 100 fresh-frozen primary breast cancer samples obtained<br />

after surgical resection. Among 100 samples, 1 failed to pass the<br />

process of RNA sequencing and no expression of fusion transcripts<br />

were observed in 7 samples resulting in 92 samples with mRNA-<br />

Seq data of fusion transcripts. Fourteen(15.2%) samples were cases<br />

with distant metastasis during follow up. Estrogen receptor(ER) was<br />

positive in 40(43.5%) samples and 23(25%) were triple negative<br />

breast cancer.<br />

Results and discussion We detected 958 fusion transcripts<br />

among 92 tumors and 58 were chosen for tumor-specific fusion<br />

transcript candidate (Fragments per kilo-base of exon per million<br />

fragments mapped, FPKM>500 and 3 times higher than average,<br />

probability>0.6). Most of the fusion transcripts were “private”,<br />

expressed only in one sample. Estrogen receptor(ER) positive<br />

breast cancer samples had 29 fusion transcripts and ER negative<br />

had 25 fusion transcripts among the 58 tumor-specific candidates.<br />

HER2 negative samples exhibited 29 fusion transcripts while 15<br />

were identified in HER2 negative samples. Triple negative sample<br />

showed 4 TF candidates. Among the 17 recurrence samples 190 fusion<br />

transcripts were identified. Of the 190 fusion transcripts in recurred<br />

samples, 24 candidates of recurrence-specific FTs were chosen by it’s<br />

high expression level (FPKM>300 and 2 times higher than average,<br />

probability>0.5). Eight were expressed both in recurred, non-recurred<br />

samples and 16 were exclusively expressed in recurred samples. No<br />

fusion transcript matched directly with the currently known actionable<br />

gene-list owing to the limited number while majority is unexploited.<br />

Further validation and functional pathway analysis is under progress<br />

to ascertain the role of highly suspected fusion transcripts with<br />

oncogenic property. Considering the high incidence of breast cancer<br />

world-wide, identification of TF expressed even in only a minor<br />

percentage among the whole breast cancer population, if druggable,<br />

it can be applicable to large number of patients.<br />

Conclusion We performed a whole-transcriptome sequencing with<br />

a large set of primary breast cancer samples and detected abundant<br />

fusion transcripts. The majority of breast cancer(92.9%, 92/99) had at<br />

least one fusion transcript suggesting it’s role during the tumorigenesis<br />

and progression process. With scarce evidence and confirmed data<br />

of actionable fusion gene and it’s product, data interpretation for it’s<br />

clinical utility is rather complicated. Accumulation of these large set<br />

data of fusion transcript must serve as a groundwork for future work.<br />

PD05-08<br />

Genomic characterisation of invasive breast cancers with<br />

heterogeneous HER2 gene amplification<br />

Ng CKY, Gauthier A, Mackay A, Lambros MBK, Rodrigues DN,<br />

Arnoud L, Lacroix-Triki M, Penault-Llorca F, Baranzelli MC, Sastre-<br />

Garau X, Lord CJ, Zvelebil M, Mitsopoulos C, Ashworth A, Natrajan<br />

R, Weigelt B, Delattre O, Cottu P, Reis-Filho JS, Vincent-Salomon A.<br />

The Breakthrough <strong>Breast</strong> <strong>Cancer</strong> Research Centre, The Institute of<br />

<strong>Cancer</strong> Research, London, United Kingdom; Institut Curie, Paris,<br />

France; CRB Ferdinand Cabanne, Centre Georges François Leclerc,<br />

Dijon, France; Institut Claudius Regaud, Toulouse, France; Centre<br />

Jean Perrin, Clermont-Ferrand, France; Centre Oscar Lambret,<br />

Lille, France<br />

Aims: HER2 gene amplification is observed in up to 15% of breast<br />

carcinomas. In a rare subset of breast cancers classified as HER2-<br />

positive by immunohistochemistry and in situ hybridisation, HER2<br />

overexpression and gene amplification are restricted to a subset of<br />

>30% but not all cancer cells. Here we sought to characterise the<br />

repertoire of gene copy number aberrations and somatic mutations<br />

in the HER2-positive and HER2-negative components of cases with<br />

heterogeneous HER2 overexpression and gene amplification.<br />

Material and methods: Cases diagnosed as HER2 positive but with<br />

>30% but


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

was re-assessed using immunohistochemistry and chromogenic and/<br />

or fluorescence in situ hybridisation. For cases with confirmed HER2<br />

gene amplification heterogeneity, HER2-positive and HER2-negative<br />

components were microdissected from tissue sections stained with the<br />

Herceptest antibody. DNA samples extracted from both components<br />

of each case were subjected to microarray-based comparative genomic<br />

hybridisation (aCGH), using a 32K BAC array platform with 50Kb<br />

resolution. The HER2-positive and HER2-negative components of<br />

cases with frozen material were also subjected to massively parallel<br />

targeted exome sequencing.<br />

Results: Twelve cases yielded sufficient DNA for aCGH analysis.<br />

Tumours were preferentially ER positive (83%) and of histological<br />

grade 3 (67%). The HER2-positive and HER2-negative components<br />

of all cases shared most of the copy number aberrations. A pairwise<br />

comparison of the genomic profiles of the two components from<br />

each case revealed that in ten of the twelve cases, copy number<br />

aberrations in addition to 17q12 amplification encompassing the<br />

HER2 gene locus were restricted to one of the two components.<br />

Exome sequencing of two cases suggested that the HER2-positive and<br />

HER2-negative components from each case harboured >30 somatic<br />

mutations in common, including identical TP53 somatic mutations<br />

in both components of each case. The HER2-negative component of<br />

one of the cases displayed a somatic mutation in NRG2, an ERBB<br />

receptor ligand, and the HER2-negative component of the other case<br />

harboured a mutation in PTTG1IP, a proto-oncogene with putative<br />

oestrogen receptor elements.<br />

Conclusions: Our results demonstrate that in HER2-positive breast<br />

cancers with heterogeneous HER2 gene amplification, the HER2-<br />

positive and HER2-negative components are clonally related. The<br />

distinct genomic profiles of HER2-positive and HER2-negative<br />

components, however, suggest that, at least in some of these cases,<br />

HER2 amplification may constitute a relatively late event in tumour<br />

evolution. Exome sequencing revealed mutations restricted to<br />

the HER2-negative components of HER2-positive tumours with<br />

heterogeneous HER2 overexpression/gene amplification, which may<br />

constitute potential drivers in the absence of HER2 overexpression/<br />

gene amplification.<br />

PD05-09<br />

Whole-genome progression of breast cancer from early neoplasia<br />

to invasive carcinoma<br />

West RB, Kashef-Haghighi D, Newburger D, Weng Z, Brunner A,<br />

Salari R, Guo X, Troxell M, Zhu S, Varma S, Sidow A, Batzoglou<br />

S. Stanford University, Stanford, CA; Oregon Health & Science<br />

University, Portland, OR<br />

<strong>Cancer</strong> evolution involves cycles of genomic damage, epigenetic<br />

deregulation, and increased cellular proliferation that eventually<br />

culminate in the carcinoma phenotype. Early breast neoplasias include<br />

usual ductal hyperplasia, columnar cell lesions, and flat epithelial<br />

atypia and some are thought to represent precursor stages. To elucidate<br />

their role in cancer evolution we performed comparative whole<br />

genome sequencing of early neoplasias, matched normal tissue, and<br />

carcinomas from six patients. The identified somatic mutations served<br />

as lineage markers to build trees that relate the tissue samples. On the<br />

basis of the lineage trees we inferred the order, timing, and rates of<br />

mutational events. We find that in a subset of cases, the last common<br />

ancestor of an early neoplasm and a carcinoma was hypermutated<br />

and had several aneuploidies, and that evolution further accelerated<br />

in the carcinoma lineage. In contrast to highly advanced tumors that<br />

are the focus of much of current cancer genome sequencing, the early<br />

neoplasia genomes, and the carcinomas as well, harbor a striking<br />

paucity of potentially functional somatic point mutations. The earliest<br />

significant events we could detect, those changes that occurred in<br />

common ancestors of neoplastic and tumor cells, are aneuploidies.<br />

Several aneuploidies are recurrent, suggesting that they are among<br />

the earliest genomic events that predispose breast tissue to eventual<br />

development of invasive carcinoma.<br />

PD06-01<br />

Automated computational Ki67 scoring in the GeparTrio breast<br />

cancer study cohort<br />

Klauschen F, Wienert S, Blohmer J-U, Mueller BM, Eiermann W,<br />

Gerber B, Tesch H, Hilfrich J, Huober J, Fehm T, Barinoff J, Jackisch<br />

C, Erbstoesser E, Loibl S, Denkert C, von Minckwitz G. Charite<br />

Universitaetsmedizin Berlin, Berlin, Germany; <strong>San</strong>kt Getrauden<br />

Hospital, Berlin, Germany; University of Munich, Munich, Germany;<br />

Klinikum Suedstadt Rostock, Rostock, Germany; Onkologisches<br />

Zentrum am Bethanien-Kankenhaus, Frankfurt, Germany; Ellenriede<br />

Hospital, Hannover, Germany; University Duesseldorf, Germany;<br />

University of Tuebingen, Germany; Kliniken Essen Mitte, Essen,<br />

Germany; Kliniken Offenbach, Offenbach, Germany; Salvator<br />

Hospital, Germany; German <strong>Breast</strong> Group, Neu-Isenburg, Germany<br />

Background: Scoring proliferation through Ki67-<br />

immunohistochemistry is an important component in predicting<br />

therapy response to chemotherapy in breast cancer patients. Therefore,<br />

an accurate and standardized Ki67-scoring is pivotal both in routine<br />

diagnostics and larger multi-center studies aiming at improving<br />

present or establishing new cut-off values for existing or novel<br />

therapy regimens. However, recent studies have cast some doubt on<br />

the reliability of “visual” Ki67 scoring by pathologists, especially<br />

within the lower - yet clinically important - proliferation range. Here,<br />

we present and apply a novel automated image analysis approach for<br />

Ki67-quantification in breast cancer tissue.<br />

Methods: We perform automated Ki67-scoring in 1219 breast cancer<br />

patients from the GeparTrio study cohort using a novel image analysis<br />

approach that avoids detection biases due to morphological variability<br />

by using a generic minimum-model approach. The method is capable<br />

of tumor-stroma-separation and may be used to process large data<br />

sets fully unsupervised in batch mode while allowing for efficient<br />

visual checks of the results. We compare these results with a different<br />

in-house-developed subtiling-based automated quantification method<br />

and moreover, gauge our approach with manual scoring performed<br />

by pathologists.<br />

Results: The results show deviations of 10% (automated method 1 vs.<br />

manual), 9% (automated method 2 vs. manual) and 3% (automated<br />

method 1 vs. automated method 2) on average. The Ki67 scores show<br />

Pearson correlations between automated and manual scoring of r>0.8<br />

(p0.95 (p


December 4-8, 2012 <strong>Abstract</strong>s: Poster Discussion 6<br />

PD06-02<br />

Standardized Assessment of Ki-67 Using Virtual Slides and<br />

Automated Analyzer for <strong>Breast</strong> <strong>Cancer</strong> Patients: Comparison of<br />

Automated and Central/Local Pathology Assessment<br />

Mizuno Y, Yamada J, Abe H, Natori T, Sato K. Tokyo-West Tokushukai<br />

Hospital, Tokyo, Japan; SRL, Inc., Tokyo, Japan<br />

Background: Ki-67 is useful in determining the effect of hormonal<br />

therapy and also as a prognostic factor. However, the method for its<br />

evaluation has not been standardized, and the results vary depending<br />

on the assessment by pathologists. We reported that the discordance<br />

rate of Ki-67 in preoperative core needle biopsy and the surgical<br />

specimens was higher than that of other biological markers, and<br />

this was considered to be due to the discrepancies in the evaluation<br />

methods (SABCS 2011). In this report, we reviewed the possibility<br />

of introducing an automated analyzer by comparing the diagnosis<br />

made by local pathologists and reassessment by central review to<br />

standardize the evaluation method for Ki-67.<br />

Subjects and Method: Of the 354 cases of primary breast cancer<br />

that had their first operation from October 2008 to March 2012,<br />

225 cases that had preoperative core needle biopsy at Tokyo-West<br />

Tokushukai Hospital were selected as the subjects of this study. Cases<br />

with ductal carcinoma in situ (DCIS) and cases with neoadjuvant<br />

chemotherapy were excluded. A cut-off value of 20% was used for<br />

Ki-67 positive criteria. The concordance rates of estrogen receptors<br />

(ER), progesterone receptor (PgR), human epidermal growth factor<br />

receptor 2 (HER2), and Ki-67 by local pathologists were reviewed,<br />

and in cases of non-matching Ki-67, tumor diameter (≈ heterogeneity)<br />

and operative method (≈ condition of formalin fixation) were studied.<br />

Next, non-matching cases (∼ to October 2011) were reassessed both<br />

by central review and by an automated analyzer (Ventana Digital<br />

Pathology) using virtual slides, and the results were studied and<br />

compared with the evaluation by local pathologists. The Wilcoxon-t<br />

test was used to review the p value for statistical analysis. Parameters<br />

such as tumor diameter, operative method were analyzed by χ 2<br />

analysis.<br />

Results: The concordance rate of Ki-67 in preoperative core needle<br />

biopsy and surgical specimens was 76.9% in 225 cases of local<br />

pathology assessment from October 2008 to March 2012. This rate<br />

was significantly lower (p < 0.001) than that of ER (95.6%), PgR<br />

(88.0%), and HER2 (91.5%). In non-matching cases, no difference<br />

was observed in tumor diameter and operative method. Reassessment<br />

of the 39 cases of non-matching Ki-67 to October 2011 using central<br />

review and an automated analyzer resulted in matching 27 cases<br />

(94.8%) and 32 (96.3%) out of 39 cases, respectively.<br />

Conclusion: The concordance rate of Ki-67 in preoperative core<br />

needle biopsy and surgical specimens was lower compared with<br />

other biological markers. However, they were nearly equal with<br />

reassessment using central review and an automated analyzer. Clinical<br />

application of Ki-67 and its standardization are challenging due to<br />

variable counting methods and measurement sites. Conducting central<br />

reviews in general clinical facilities is difficult, but introducing an<br />

automated analyzer with a Ki-67 labeling index is shown to achieve<br />

that goal.<br />

PD06-03<br />

Development of a highly reproducible clinical test for Ki67 using<br />

AQUA technology<br />

Murphy DA, Pacia E, Beruti S, Lamoureux J, Dabbas B, Diver J,<br />

Christiansen J. Genoptix Medical Laboratory, Carlsbad, CA<br />

Background: The measurement of Ki67 is showing promise in the<br />

management of breast cancer patients. It has shown potential for<br />

prognosis and as a chemotherapeutic response predictor, and also<br />

has a role in multivariate assessment of residual risk. However,<br />

the advancement in clinical testing has been arduous due to a lack<br />

of clinically validated scoring and/or cutpoints to guide clinicians.<br />

Standardization of Ki67 scoring is hindered by inter-laboratory<br />

variability in analysis of Ki67 in clinically relevant whole tissue<br />

sections, which are complicated not only by the subjective<br />

measurement required for IHC but also by heterogeneous expression<br />

due to biological variation and inconsistent fixation. In our diagnostic<br />

laboratory, we observed a significant amount of variability in the<br />

assessment of Ki67 using manual IHC scoring for percentage of<br />

positively stained nuclei. To attempt to reduce the variations in<br />

Ki67 assessment in clinically relevant specimens, our laboratory<br />

investigated the use of image analysis methods. Fluorescence<br />

immunohistochemistry (FIHC) coupled with the AQUA® technology<br />

has been established as a diagnostic test using image analysis and<br />

has been shown to eliminate much of the variation introduced in<br />

typical visual assessment. Materials and methods: FIHC, based on<br />

the SP6 antibody clone, was used with AQUA technology to assess<br />

Ki67 expression as measured by percentage of positively stained<br />

nuclei. Results were compared to manually scored IHC results<br />

generated using the commonly used anti-Ki67 MIB1 antibody. IHC<br />

interpretation was performed by three pathologists on the same slides.<br />

Results: The Ki67 assay using FIHC and AQUA technology was<br />

validated for use in a CLIA environment. Inter-reader variability by<br />

IHC was first demonstrated on whole tissues (Average difference<br />

in percent positive nuclei was 18% and ranged from 0-50%). This<br />

was further assessed on tissue microarrays, which mitigate the<br />

heterogeneity component of the tissue and resulted in the same<br />

high inter-reader variability (average percent difference was 18%).<br />

A significant correlation was observed for Ki67 percent positivity<br />

between AQUA and IHC in TMAs (Spearman: r=0.88, p< 0.0001).<br />

FIHC staining and scoring for Ki67 positivity by AQUA resulted<br />

in a 3-fold reduced variation compared to IHC and manual reads<br />

(6% variation). Using clinically relevant whole tissue sections, the<br />

accuracy of Ki67 measurement by AQUA was compared to IHC<br />

performed by an outside laboratory and resulted in a significant<br />

correlation between the two methods (Spearman: r=0.78, p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Instead, the consensus introduced clinicopathological BC subtypes<br />

(cpBCST) defined as sets of risk and predictive biomarkers which<br />

are established worlwide (Table 1).<br />

Table 1: St Gallen Clinico-pathological Subtype Definition<br />

Clinico-pathological Subtype<br />

Luminal A<br />

Biomarker<br />

ER/PR+<br />

Her2 neg<br />

Proliferation ⇓<br />

Luminal B Her2 neg ER/PR+<br />

Her2 neg<br />

Proliferation ⇑<br />

Her2+<br />

ER/PR+<br />

Her2+<br />

Her2 non luminal<br />

ER/PR neg<br />

Her2+<br />

Triple-negative<br />

ER/PR neg<br />

Her2 neg<br />

Special histological types endocrine responsive ER/PR+<br />

Her2 neg<br />

endocrine non-respnsive ER/PR neg<br />

Her2 neg<br />

To discriminate Luminal A vs. Her2 negative Luminal B BCs the<br />

St Gallen Consensus recommends Ki67 for defining proliferation.<br />

Although, because no standardized methodology or cut-off definition<br />

for Ki67 is available so far ‘some alternative measure of proliferation’<br />

was suggested. We analyzed the influence of different proliferation<br />

assessment methods on BC subtype distribution.<br />

Methods<br />

cpBCST assessment including proliferation measurement by different<br />

variants of Ki67-Labeling-Index (KI67LI) was performed on 225<br />

BCs. For comparison we used the Mitotic Activity Index (MAI) as<br />

the only prospectively evaluated and best reproducible proliferation<br />

measurement (Baak et al 2008). The Ki67-LI was based on counting<br />

any nuclear staining of distinctly defined target areas: (1) 100 tumor<br />

cells within the hot spot (Ki67-100), (2) 1020 tumor cells in 3 HPF<br />

in the tumor periphery including the hot spot (Ki67-1020periphery),<br />

and (3) 1020 tumor cells in 3 HPF including hot spot, cold spot, and<br />

an intermediate area (Ki67-1020spectrum). Staining quality and<br />

counting accuracy was tested as excellent by the 2011 circulation of<br />

the German Quality Initiative in Pathology. According to St Gallen,<br />

a cut-off


December 4-8, 2012 <strong>Abstract</strong>s: Poster Discussion 6<br />

PD06-06<br />

RELATIONSHIP BETWEEN MOLECULAR SUBTYPE AND<br />

CHANGE IN KI-67 IN THE PLACEBO ARMS OF WINDOW<br />

OF OPPORTUNITY TRIALS<br />

Pruneri G, Gandini S, Guerrieri-Gonzaga A, Serrano D, Cazzaniga<br />

M, Lazzeroni M, Puntoni M, Toesca A, Caldarella P, Johansson<br />

H, Bonanni B, DeCensi A. European Institute of Oncology, Milan,<br />

Italy; Galliera Hospital, Genoa, Italy; University of Milan, School<br />

of Medicine, Milan, Italy<br />

Background: Window-of-opportunity (WOP), presurgical models<br />

often use Ki-67 labeling index (LI) as the main surrogate biomarker<br />

for screening therapeutic activity of candidate agents and characterize<br />

their mechanism of action. Ki-67 LI in the placebo arm has been<br />

frequently reported to be higher at surgery than at baseline biopsy<br />

in WOP studies, a finding that has been ascribed to the higher<br />

proliferative activity in the tumor edge of surgical specimens that<br />

cannot be detected in the baseline core biopsy. Since this observation<br />

has important implications in sample size calculation and effect<br />

interpretation, we investigated which factors affected the changes<br />

of Ki-67 LI in a series of patients allocated to the placebo arms of a<br />

number of WOP trials carried out at IEO.<br />

Methods: Data from 181 patients with pT 1-2 invasive breast cancer<br />

from the placebo arms of three WOP randomized trials were pooled<br />

with those of 98 untreated patients, who had a diagnostic core biopsy<br />

preceding surgery but declined participation to the study or were<br />

ineligible for histological characteristics. Ki-67 LI was measured by<br />

evaluating the prevalence of neoplastic cells showing any definite<br />

nuclear immunostaining with the Mib-1 monoclonal antibody, in the<br />

whole invasive component of baseline core biopsies and in at least<br />

2000 invasive neoplastic cells of surgical samples, in accordance to<br />

the recently licensed international recommendations.<br />

Results: We collected data of 273 breast cancer patients with<br />

information on the changes in Ki-67 LI: 46 (17%) were Luminal A,<br />

85 (31%) Luminal B/HER2-, 24 (9%) Luminal B/HER2+, 38 (14%)<br />

HER2+ and 81 (30%) Triple Negative (TN). Median (IQR) Ki-67LI<br />

at baseline in each molecular subtype was 10% (7-11), 20% (16-<br />

29), 28% (22-35), 30% (25-45) and 50% (32-75) in the Luminal A,<br />

Luminal B/HER2-, Luminal B/HER2+, HER2+ and TN, respectively.<br />

Median (IQR) age was 50 years (44-60), median BMI was 24 (22-27),<br />

50% were post-menopausal. The median (range) time elapsed from<br />

biopsy to surgery was 41 days (33-48). Overall, the median change<br />

in Ki-67 LI between baseline biopsies and surgical samples was 0<br />

(IQR, -2, 5). Results from multivariate analysis showed that none of<br />

the factors investigated, including patient and tumor characteristics,<br />

time elapsed from biopsy to surgery and circulating biomarkers<br />

(IGF-I, SHBG and ultrasensitive CRP) were associated with the<br />

Ki-67 LI change except for the immunohistochemically defined<br />

molecular subtype, which explained most of the variability of the<br />

changes (P=0.004). As a matter of fact, we observed a 5%, significant<br />

increase of Ki-67 LI both in HER2+ and TN tumors, after adjustment<br />

for baseline values.<br />

LS means and 95%CI Ki-67 LI changes (surgery-baseline biopsy) in the placebo/untreated arms by<br />

molecular subtype<br />

Luminal A<br />

Luminal B/ Luminal B/<br />

HER2- HER2+<br />

HER2+ Triple negative<br />

n 41 90 26 34 88<br />

Mean -0.9 -0.4 1.8 5.0 5.3<br />

95% CI -3.9, +2.0 -2.4, +1.6 -1.8, +5.4 +2.2, +7.9 +2.8, +7.7<br />

Conclusions: We reported a significant increase in Ki-67 LI between<br />

baseline biopsy and endpoint surgery in the placebo arms of HER2+<br />

and TN tumors. This association suggests a real biological increase<br />

in proliferation rather than an analytical artifact, and should be taken<br />

into account in designing future WOP studies.<br />

PD06-07<br />

Evaluation of an optimal cut-off point for the Ki-67 index as a<br />

prognostic factor in primary breast cancer.<br />

Tashima R, Nishimur R. Kumamoto Municipal Hospital, Kumamoto,<br />

Japan<br />

Background: The proliferation biomarker Ki-67 is considered to<br />

be a prognostic factor for breast cancer and has been investigated in<br />

several studies. However, there is no standard cut-off point for the<br />

Ki-67 index. Therefore, we retrospectively investigated an optimal<br />

cut-off point in terms of the prognostic factor in primary breast cancer.<br />

Patients&Methods: Immunohistochemical (IHC) analysis of the<br />

proliferation marker Ki-67 index was performed on 4338 patients<br />

with primary breast cancer until March 2012 at Kumamoto City<br />

Hospital. Out of these patients, there were 2375 consecutive cases<br />

with ER and/or PgR positive and HER2 negative tumors since 1997.<br />

Items examined were ER, PgR, HER2, tumor size, nodal status,<br />

nuclear grade, and p53 overexpression. The Kaplan-Meier test was<br />

used to calculate prognosis (cumulative disease-free survival (DFS)<br />

and overall survival (OS)) and tested with the log-rank procedure.<br />

Cox’s proportional hazard model was used to perform a univariate<br />

and multivariate analyses of the factors related to DFS. In addition,<br />

an analysis was conducted to determine various cut-off values for<br />

the Ki-67 index.<br />

Results: The median Ki-67 value was 21%, with a mean of 26.2%.<br />

Univariate and multivariate analysis revealed that the Ki-67 index<br />

was an independent prognostic factor for DFS and OS. A multivariate<br />

analysis revealed the following hazard ratio (HR) and cut-off values<br />

for the Ki-67 index; HR was 1.92 in cases with a cut-off value of<br />

20%, 1.91 in 35%, 1.87 in 40%, 1.89 in 45% and 1.79 in 50%,<br />

respectively. These findings suggested that the optimal cut-off point<br />

was 20%. In the cases with luminal type (ER and/or PgR positive<br />

and HER2 negative) breast cancer, a higher Ki-67 value (20% ≤)<br />

correlated with larger tumor size, positive nodes, higher grade, and<br />

overexpression of p53. Moreover, patients with a higher Ki-67 value<br />

showed significantly lower DFS / OS rates. A multivariate analysis<br />

also revealed that the Ki-67 index is a significant prognostic factor<br />

in luminal type breast cancer.<br />

Conclusion: The optimal cut-off value for the Ki-67 index is 20%.<br />

Moreover, this proliferation marker is a significant prognostic factor<br />

in evaluating primary breast cancer.<br />

PD06-08<br />

Strong prognostic concordance between Ki67 and binary, but not<br />

multi-level gene expression signatures<br />

Tobin NP, Lindström LS, Carlson JW, Bergh J, Wennmalm K.<br />

Karolinska Institutet and University Hospital, Stockholm, Sweden<br />

Background: Several proliferation-related gene signatures have<br />

been proposed to aid breast cancer management by providing better<br />

prognostic and therapy predictive information on a patient-by-patient<br />

basis. It is unclear however, whether a less demanding assessment of<br />

cell division rate (as determined by Ki67 expression) can function in<br />

place of gene profiling.<br />

Methods: Agreement between literature-, distribution-based, as well<br />

as signature-derived threshold values for Ki67, relative to the genomic<br />

grade index (GGI), the 70-gene signature, the p53 signature, the<br />

recurrence score (RS), and the molecular subtype models of Sorlie,<br />

Hu, and Parker was investigated in a representative set of 253 breast<br />

cancers with 25 years follow up. Relevance for breast cancer specific<br />

survival was assessed for contrast groups, and in uni- and multivariate<br />

Cox models.<br />

www.aacrjournals.org 131s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Results: Our broad approach identified Ki67 cutoffs in the range of<br />

10%-20%. With optimum signature-reproducing cutoffs, similarity in<br />

classification of individual tumors was high for GGI (81%), the 70-<br />

gene (78%), and the p53 (82%) signatures, but lower for the recurrence<br />

score (68%), Sorlie (68%), Parker (67%) and Hu (68%) signatures.<br />

Consistent with strong agreement, no prognostic superiority was noted<br />

for Ki67 vs. the binary GGI, 70-gene and p53 signatures respectively,<br />

in multivariate analyses. In contrast, Ki67-independent prognostic<br />

capacity could be demonstrated for RS and molecular subtypes<br />

according to Sorlie, Parker and Hu (Table 1).<br />

Conclusions: The added prognostic value of binary proliferationrelated<br />

gene signatures appears limited for Ki67-assessed breast<br />

cancers. More complex, multi-level descriptions seem to have a<br />

greater potential in pinpointing the outcome heterogeneity in breast<br />

cancer. We aim to validate our findings in a second cohort of 159<br />

breast cancer patients with over 15 years of clinical follow up.<br />

Table 1. Cox uni- and bivariate analysis, hazard ratio (HR) and 95% conf. interval (CI)<br />

Covariate Separate models HRs (CI) Combined model HRs (CI)<br />

Genomic grade index 2.0 (1.27-3.3) 1.6 (0.85-2.9)<br />

Ki67 ≥ 15 1.9 (1.21-3.1) 1.5 (0.79-2.7)<br />

70-gene signature 2.0 (1.21-3.2) 1.5 (0.84-2.7)<br />

Ki67 ≥ 11 2.0 (1.24-3.3) 1.6 (0.89-2.9)<br />

P53 signature 1.8 (1.10-2.9) 1.2 (0.64-2.3)<br />

Ki67 ≥ 16 1.8 (1.15-3.0) 1.7 (0.90-3.1)<br />

Recurrence score* 2.5 (1.43-4.6) 2.1 (1.12-3.8)<br />

Ki67 ≥ 12 2.1 (1.28-3.4) 1.7 (1.00-2.8)<br />

Molecular subtype (Sorlie)† 2.4 (1.39-4.1) 2.0 (1.15-3.5)<br />

Ki67 ≥ 16 1.8 (1.15-3.0) 1.5 (0.89-2.4)<br />

Molecular subtype (Hu)‡ 2.5 (1.57-4.1) 2.2 (1.31-3.6)<br />

Ki67 ≥ 13 2.1 (1.30-3.4) 1.7 (1.00-2.8)<br />

Molecular subtype (Parker)‡ 2.8 (1.72-4.4) 2.2 (1.35-3.7)<br />

Ki67 ≥ 13 2.1 (1.30-3.4) 1.6 (0.97-2.7)<br />

*Hazard ratio for high or intermediate recurrence score; low as reference. †Hazard ratio for luminal<br />

B subtype; others as reference. ‡Hazard ratio for luminal B or HER2 subtype; others as reference.<br />

PD07-01<br />

Z1031B Neoadjuvant Aromatase Inhibitor Trial: A Phase 2 study<br />

of Triage to Chemotherapy Based on 2 to 4 week Ki67 level > 10%.<br />

Ellis MJ, Suman V, McCall L, Luo R, Hoog J, Brink A, Watson M,<br />

Ma C, Unzeitig G, Pluard T, Whitworth P, Babiera G, Guenther M,<br />

Dayao Z, Leitch M, Ota D, Olson J, Hunt K, Allred C. Siteman <strong>Cancer</strong><br />

Center, Washington University, St Louis, MO; Mayo Clinic; Duke<br />

University; Doctor’s Hospital of Laredo; Nashville <strong>Breast</strong> Center;<br />

MD Anderson <strong>Cancer</strong> Center; St. Elizabeth Med Ctr Southwest;<br />

Univ of New Mexico; UT Southwestern; University of Maryland<br />

Medical Center<br />

Background: Neoadjuvant aromatase inhibitor (AI) therapy would<br />

become a more acceptable alternative to chemotherapy if there was a<br />

way to identify AI-resistant cases early for triage to alternate systemic<br />

treatment. We therefore conducted a Phase 2 trial of post-menopausal<br />

patients with clinical stage 2 or 3 ER+ (Allred Score 6 to 8) breast<br />

cancer who were re-biopsied 2 to 4 weeks after initiating neoadjuvant<br />

AI therapy. If the tumor Ki67 level was > 10% (AI resistant) the<br />

patient was triaged to either chemotherapy or immediate surgery<br />

and if < 10% (AI sensitive) the patient completed 16 to 18 weeks of<br />

neoadjuvant AI treatment. Co-primary endpoints were: 1) to confirm<br />

that the pathological complete response (pCR) rate to standard NCCN<br />

chemotherapy in the AI resistant population is in the range of 20%<br />

(similar to ER- disease). This required the observation of least 4/35<br />

pCRs; 2) whether patients were willing to accept a recommendation<br />

of no chemotherapy when the preoperative endocrine prognostic<br />

index score was zero (PEPI-0) (pStage 1 or 2A, Ki675 yr postmenopausal subset), and ABCSG-12.<br />

In addition, combining ZOL with standard neoadjuvant therapy<br />

improved pathologic complete response (pCR) and reduced the need<br />

for mastectomy in the AZURE trial. The open-label, multicenter,<br />

phase 2, FEMZONE trial examined the effect of neoadjuvant letrozole<br />

(LET) ± ZOL on tumor response in postmenopausal patients with<br />

hormone receptor-positive (HR + ) primary BC. Here, we report the<br />

efficacy and safety after 6 mo of treatment.<br />

Methods: Postmenopausal women with HR + BC and ECOG<br />

performance status of 0-2 were randomized to LET (2.5 mg/d) ± ZOL<br />

(4 mg q4w). The primary efficacy endpoint was objective response<br />

rate (ORR; complete + partial response per RECIST criteria) at 6<br />

mo by central review. Secondary endpoints included ORR in the<br />

per-protocol and safety populations, best response, breast-conserving<br />

surgery, pCR, change in tumor size, overall survival, and safety.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

132s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Discussion 7<br />

Between-group differences in ORR were evaluated using Fisher’s<br />

exact test.<br />

Results: Among enrolled patients (N = 168; mean age 70.9 yr;<br />

mean treatment duration 6.5 mo), patients receiving LET + ZOL<br />

experienced numerically better ORR at 6 mo versus LET alone<br />

(69.2% vs 54.5%, respectively), with a between-group difference<br />

of 14.7% (P = .106). Although the primary endpoint did not reach<br />

statistical significance because of insufficient patient recruitment, the<br />

prespecified between-group difference of ≥10% was met. A similar<br />

trend in ORR at 6 mo favoring ZOL was also seen by local assessment<br />

(∆11%, P = .192). At 4 mo, higher ORR was observed for LET + ZOL<br />

versus LET alone both for the local (33.3% vs 24%, respectively)<br />

and central assessments (41.5% vs 30.3%, respectively). Numerically<br />

more patients receiving LET + ZOL experienced a partial response<br />

at 6 mo versus LET alone (66.2% vs 54.5%, respectively). At 6 mo,<br />

the mean tumor size had decreased from 3.34 ± 1.98 cm to 2.1 ± 1.76<br />

cm in the overall patient population, and the decrease was slightly<br />

larger with LET + ZOL compared with LET alone (–1.37 ± 0.96 cm<br />

vs –1.12 ± 0.92 cm, respectively). Overall, there was no difference<br />

in the number of patients requiring mastectomy between LET +<br />

ZOL and LET alone (15.6% vs 15.2%, respectively). Incidence of<br />

adverse events (AEs) was slightly higher among patients receiving<br />

LET + ZOL versus LET alone (92.1% vs 81%, respectively), and the<br />

types of AEs were consistent with the known safety profiles of these<br />

agents. No deaths or osteonecrosis of the jaw was reported during<br />

the course of the study.<br />

Conclusions: This study provides additional confirmation of the<br />

efficacy of LET as neoadjuvant therapy for early HR + BC, and<br />

supports previous reports of ZOL’s anticancer activity in this setting<br />

(as evidenced by the trend toward improved ORR with ZOL +<br />

LET versus LET alone). These data, together with other published<br />

ZOL studies, suggest that adding ZOL to neoadjuvant therapy may<br />

be considered for postmenopausal patients scheduled to receive<br />

neoadjuvant endocrine therapy for BC.<br />

PD07-03<br />

INCREASED PATHOLOGIC COMPLETE RESPONSE RATE<br />

AFTER A LONG TERM NEOADJUVANT LETROZOLE<br />

TREATMENT IN POSTMENOPAUSAL ESTROGEN AND/OR<br />

PROGESTERONE RECEPTOR-POSITIVE BREAST CANCER<br />

Allevi G, Strina C, Andreis D, Zanoni V, Bazzola L, Bonardi S, Foroni<br />

C, Milani M, Cappelletti MR, Generali D, Berruti A, Bottini A. A.O.<br />

Istituti Ospitalieri di Cremona, Cremona, Italy; A.O.U. <strong>San</strong> Luigi<br />

Gonzaga di Orbassano, Orbassano, TO, Italy<br />

Background:<br />

Neoadjuvant endocrine therapy has been increasingly employed in<br />

clinical practice to improve surgical options for postmenopausal<br />

women with bulky hormone receptor-positive breast cancer. Recent<br />

studies indicate that tumor response and in particular the pathologic<br />

complete response (CR) in this setting may predict long-term<br />

outcome. The letrozole approval in neoadjuvant setting is based on a<br />

study that included 4 months of letrozole treatment only. Prospective<br />

studies have not investigated treatment duration beyond 6 months,<br />

and retrospective studies suggest that useful responses can occur after<br />

this period. The purpose of this study was to determine the clinical<br />

and pathologic response of 2.5 mg daily letrozole used with different<br />

schedule in breast cancer patients (pts) unfit for chemotherapy.<br />

Patients and Methods:<br />

This single Institution retrospective study comprised 102<br />

postmenopausal women with primary breast cancer (clinical stage<br />

≥T2, N0-1), who were divided into 3 subgroups (34 each in single<br />

subgroup) according to the different duration of neoadjuvant letrozole<br />

treatment: 4 months (subgroup A), 8 months (subgroup B) or 12<br />

months (subgroup C) of treatment. The subgroups were comparable<br />

in terms of baseline characteristics.<br />

Results:<br />

Pts and tumor characteristics included: median age 77.3 years (range<br />

53.9-95.4); 100% estrogen receptor-positive, 81.4% progesterone<br />

receptor-positive; median Ki67 13.5% (range 2-90%). All pts were<br />

evaluable for clinical and pathologic response. A total of 77 pts<br />

(75.5%) achieved an objective response at individual end: 40 (39.2%)<br />

clinical CR and 37 (36.3%) partial responses (PR). The clinical CR<br />

were significantly confined to subgroups C (20/34 pts, 58.8%) and<br />

B (16/34 pts, 47.1%) than to subgroup A (4/34 pts, 11.8%) (p for<br />

trend =0.00001). Objective response rate was 46.1% (47/102 pts)<br />

at month 4, 83.8% (57/68 pts) at month 8, and 94.1% (32/34 pts) at<br />

month 12 compared with baseline. As expected, letrozole induced a<br />

significant reduction of Ki67 expression after treatment in any single<br />

subgroup, without significant differences between them. With very<br />

interest, the pathologic CR rate was significantly higher in subgroup<br />

C (7/34 pts, 20.6%) than in subgroups A (1/34 pt, 2.9%) and B (1/34<br />

pt, 2.9%) (p for trend


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Results<br />

Between Oct 2007 and Apr 2011, 59 pts were randomized to arm A<br />

and 57 to arm B. Main baseline characteristics were well-balanced<br />

between the 2 arms: Median age was 68 yrs-old (53-91) in arm A<br />

and 74 yrs-old (51-88) in arm B. Histological grades were EE I-II<br />

in 53 pts (89 %) and 49 pts (86%) and median clinical size before<br />

treatment was 41.5 mm and 42.3 mm in arm A and B respectively.<br />

Neither SAE nor grade 3/4 toxicity was reported. The most common<br />

treatment-related AEs were grade1/2 hot flushes (27% and 12% of<br />

pts in arm A and B respectively), and musculoskeletal symptoms<br />

(20% and 21%).<br />

35 pts in arm A and 29 pts in arm B continued assigned treatment up<br />

to 6 mo depending on the clinical response evaluated at 4 mo. Also,<br />

the clinical response rate was estimated at 4 mo orat 6 mo. 1 death<br />

post-surgery was reported in arm B with no proven relationship with<br />

treatment.<br />

Overall clinical response rates (CR + PR) at 4 or 6 mo were 62% (CI<br />

95% [49-75]) in arm A and 46% (CI 95% [32-59]) in arm B. Clinical<br />

response rate amelioration at 6 mo was observed among 15% of pts<br />

in each arm.<br />

BCS was performed in 59% of pts in arm A and 49% in arm B. (1 pt<br />

from arm B refused surgery).<br />

Pathological response according to Sataloff classification: TA and<br />

TB tumor responses were observed in 17/59 pts (29%) in arm A vs<br />

12/57 (21%) in arm B respectively.<br />

Conclusions<br />

Both anastrozole and fulvestrant show excellent efficacy and<br />

tolerability as neoadjuvant therapy in post-menopausal pts with<br />

endocrine-dependent, HER2-negative BC.<br />

Objective response rates and improvement in surgical outcome seem<br />

to be more frequent with anastrozole. However disease stabilization<br />

and tolerability are in favour of fulvestrant.<br />

Our data suggest that neo-adjuvant HT improves surgical options<br />

for HR+ post-menopausal women. Correlation between clinical &<br />

pathological responses and outcome as well as the identification of<br />

markers of sensitivity to both treatments will be also studied.<br />

PD07-05<br />

A randomized controlled trial comparing zoledronic acid plus<br />

chemotherapy with chemotherapy alone as a neoadjuvant<br />

treatment in patients with HER2-negative primary breast cancer<br />

Hasegawa Y, Kohno N, Horiguchi J, Miura D, Ishikawa T, Hayashi<br />

M, Takao S, Kim SJ, Tanino H, Miyashita M, Konishi M, Shigeoka<br />

Y, Yamagami K, Akazawa K. Hirosaki Municipal Hospital, Hirosaki,<br />

Aomori, Japan; Tokyo Medical University Hospital, Tokyo, Japan;<br />

Gunma University Hospital, Maebashi, Gunma, Japan; Toranomon<br />

Hospital, Tokyo, Japan; Yokohama City University Medical<br />

Center, Yokohama, Kanagawa, Japan; Tokyo Medical University<br />

Hachioji Medical Center, Hachioji, Tokyo, Japan; Hyogo <strong>Cancer</strong><br />

Center, Akashi, Hyogo, Japan; Osaka University Hospital, Suita,<br />

Osaka, Japan; Naga Municipal Hospital, Kinokawa, Wakayama,<br />

Japan; Konan Hospital, Kobe, Hyogo, Japan; Hyogo Prefectural<br />

Nishinomiya Hospital, Nishinomiya, Hyogo, Japan; Yodogawa<br />

Christian Hospital, Osaka, Japan; Shinko Hospital, Kobe, Hyogo,<br />

Japan; Niigata University Medical and Dental Hospital, Niigata,<br />

Japan<br />

Background:<br />

Zoledronic acid (ZOL) is a nitrogen-containing bisphosphonate,<br />

and it induces osteoclast apoptosis and inhibits bone resorption by<br />

inhibiting the mevalonate pathway. ZOL has also been found to have<br />

antitumor effects that include an angiogenesis inhibiting action, an<br />

inhibitory effect on tumor cell adhesion to bone, an inhibitory effect<br />

on invasion of the extracellular matrix, and activation of a gdT cells.<br />

In addition, it has been found to have a synergistic anti-proliferative<br />

effect when used in combination with antitumor drugs.<br />

In this study we investigated the pathological complete response<br />

(pCR) rate when ZOL is added to anthracycline followed by taxane to<br />

treat T2 and T3 breast cancer patients as a neoadjuvant chemotherapy.<br />

Methods:<br />

Women with resectable invasive StageIIA-IIIB (T ≥3 cm or T ≥2 cm<br />

and lymph node positive) breast cancer who are HER-2-negative,<br />

between 20 and 70 years of age, and ECOG PS 0-1 were eligible.<br />

Patients with distant metastasis, patients who have had received<br />

chemotherapy, hormone therapy, or radiotherapy for breast cancer,<br />

patients with serious complications, such as heart disease or an<br />

infection, patients with a complicating dental or jaw infection or<br />

traumatic condition of the teeth, and patients with a history of<br />

treatment with a bisphosphonate within the previous 12 months<br />

were excluded.<br />

A total of 4 courses of FEC100 were administered every 3 weeks<br />

followed by weekly paclitaxel for 12 courses. ZOL 4mg was<br />

administered every 3-4 weeks a total of 7 times.<br />

Patients were randomized 1:1 to chemotherapy + ZOL group or<br />

chemotherapy alone group, according to the presence or absence<br />

of lymph node metastasis, estrogen receptor (ER) status, and their<br />

menopausal status.<br />

The primary endpoint was the rate of pCR defined as absence of<br />

invasive disease in the breast at surgery. Secondary endpoints were<br />

tumor response rate (RECIST ver1.0), the breast-conserving surgery<br />

ratio, and disease-free survival (DFS).<br />

We calculated the sample size on the basis of a pCR rate of 18% in<br />

the chemotherapy alone group and 35% in the chemotherapy + ZOL<br />

group, at 5% significance level based on the one-sided chi-square<br />

test with the statistical power of 80%, and as a result we targeted a<br />

patient sample size of 180 patients.<br />

Results:<br />

188 patients were recruited between March 2010 and April 2012.<br />

Results of the primary analysis of efficacy and safety will be presented.<br />

PD07-06<br />

NEO-ZOTAC: Toxicity data of a phase III randomized trial with<br />

NEOadjuvant chemotherapy (TAC) with or without ZOledronic<br />

acid (ZA) for patients with HER2-negative large resectable or<br />

locally advanced breast cancer (BC)<br />

van de Ven S, Liefers G-j, Putter H, van Warmerdam LJ, Kessels LW,<br />

Dercksen W, Pepels MJ, Maartense E, van Laarhoven HWM, Vriens<br />

B, Smit VTHBM, Wasser MNJM, Meershoek-Klein Kranenbarg EM,<br />

van Leeuwen-Stok E, van de Velde CJH, Nortier JWR, Kroep JR.<br />

Leiden University Medical Center (LUMC), Leiden, Netherlands;<br />

Catharina Hospital, Eindhoven, Netherlands; Deventer Hospital,<br />

Deventer, Netherlands<br />

Background: The role of bisphosphonates (BP) when added to the<br />

(neo)adjuvant treatment of BC in enhancing the efficacy of therapy<br />

is still unknown. NEOZOTAC investigates the efficacy of ZA added<br />

to neoadjuvant chemotherapy in patients with HER2-negative BC.<br />

Trial design: NEOZOTAC is a Dutch multicenter study. Patients are<br />

1:1 randomized to 3-weekly TAC (docetaxel 75mg/m 2 , adriamycin 50<br />

mg/m 2 and cyclophosphamide 500 mg/m 2 i.v., day 1) chemotherapy<br />

supported by pegfilgrastim (6 mg sc), day 2 with or without ZA (4<br />

mg i.v. within 24 hr after chemotherapy) q3 weeks.<br />

Eligibility criteria: Main inclusion criteria: stage II or III,<br />

measurable, HER2-negative BC, age ≥18 years, WHO 0-2, adequate<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

134s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Discussion 7<br />

bone marrow-, renal-, and liver function, absence of prior BP usage<br />

and absence of active dental problems.<br />

Study endpoint: The primary endpoint is the pathologic complete<br />

response (pCR) rate. Secondary endpoints are toxicity, clinical<br />

response, tumor heterogeneity in core biopsy vs. operation specimen,<br />

and (disease free) survival. Optional side studies include fluorescent<br />

imaging (SoftScan®), changes in bone markers, single nucleotide<br />

polymorphisms and the insulin-like growth factor pathway, circulating<br />

tumor and endothelial cells and the false-negative rate of the sentinel<br />

node biopsy after neoadjuvant chemotherapy.<br />

Statistical Methods: Using a 5% significance level based on the twosided<br />

Fishers exact test with a power of 80%, 250 patients (125/arm)<br />

are needed to show an improvement of the pCR-rate from 17% to<br />

34% in the experimental arm. Randomization was done according to<br />

the Pococks minimisation technique stratified by cT, cN, and estrogen<br />

receptor status. Toxicity is analyzed using the Exact (2-sided) Chi-<br />

Square test.<br />

Results: From July 2010 to April 2012, 250 patients from 25<br />

participating sites were randomized. Toxicity data of 173 patients<br />

are currently available and data of all 250 patients will be presented<br />

at SABCS. Patient characteristics are presented in table 1.<br />

Table 1. Patient Characteristics<br />

TAC-ZA TAC Total<br />

Patient nr 90 83 173<br />

WHO 0 98% 98% 98%<br />

WHO 1 2% 2% 2%<br />

Median Age (range) 49 (29-67) 49 (29-70) 49 (29-70)<br />

T-stage cT2 56% 54% 55%<br />

cT3/T4 44% 46% 45%<br />

N-stage cN+ 56% 55% 56%<br />

ER-status ER+ 81% 86% 83%<br />

Menopausal status pre 56% 56% 56%<br />

peri 11% 15% 13%<br />

post 33% 30% 31%<br />

Hematological and non-hematological toxicities were not significantly<br />

different between both treatment arms. Main grade 3/4 NCI-CTCv4<br />

toxicities were neutropenia (8%), followed by febrile neutropenia<br />

(7%), fatigue (6%), diarrhea, hypertension, nausea (3%) and vomiting<br />

(1.2%). Bone pain, myalgia, and hypocalcemia occurred in one<br />

patient in the TAC-ZA arm (0.6%). Osteonecrosis of the jaw was<br />

not observed.<br />

Conclusions: Neoadjuvant TAC supported by pegfilgrastim plus ZA<br />

is feasible. No significant difference in toxicity are reported compared<br />

with the control arm.<br />

PD07-07<br />

Prediction of antiproliferative response to lapatinib by HER3 in<br />

an exploratory analysis of HER2-non-amplified (HER2-) breast<br />

cancer in the MAPLE presurgical study (CRUK E/06/039)<br />

Dowsett M, Leary A, Evans A, A’Hern R, Bliss J, Sahoo R, Detre S,<br />

Hills M, Haynes B, Harper-Wynne C, Bundred N, Coombes G, Smith<br />

IE, Johnston S. Royal Marsden Hospital, London, United Kingdom;<br />

Institut Gustave Roussy, Paris, France; Poole Hospital, Poole,<br />

Dorset, United Kingdom; Institute of <strong>Cancer</strong> Research, London,<br />

United Kingdom; Kent Oncology Centre, Maidstone, Kent, United<br />

Kingdom; University Hospital of South Manchester NHS Trust,<br />

Manchester, United Kingdom<br />

Aim: To identify pretreatment biomarker predictors of Ki67 response<br />

to lapatinib in women with HER2- primary breast cancer.<br />

Background: Lapatinib is an EGFR/HER2 inhibitor. Its clinical use is<br />

restricted to HER2 overexpressing disease. The MAPLE (Molecular<br />

Antiproliferative Predictors of Lapatinib’s Effects) presurgical<br />

window of opportunity study of lapatinib vs placebo was conducted<br />

in women with HER2-amplified (HER2+) or HER2- primary disease.<br />

Ki67 (primary end-point) was reduced by a geomean 46% (95%CI<br />

23-63%, p=0.002) and 27% (95%CI 8-42%, p=0.008) in HER2+<br />

and HER2- disease, respectively (Leary et al, AACR 2012). We have<br />

now assessed whether predictive biomarkers of the antiproliferative<br />

response in HER2- disease could be identified.<br />

Methods: 121 primary breast cancer patients were randomized (3:1)<br />

to 14 days of 1500mg/d lapatinib or placebo before surgery. Biopsies<br />

were taken before treatment and at surgery. Ki67 responders were<br />

defined as having a >/=50% reduction in Ki67 compared to baseline<br />

(Ellis,P et al, <strong>Breast</strong> <strong>Cancer</strong> Res Treat 1998, 48, 107). ER, PgR,<br />

HER2, EGFR, pAKT, pERK1/2 (nuclear and cytoplasmic), stathmin<br />

and apoptosis (TUNEL) were assessed by IHC (+FISH for HER2[all<br />

cases]) and scored visually by continuous methods. HER2, HER3,<br />

epiregulin (epir), amphiregulin (amphir) and neuregulin (neur) were<br />

assessed by qrtPCR.<br />

Results: Three of the 121 patients were excluded because of inadequate<br />

biopsy material. Ninety-one of the remaining 118 patients received<br />

lapatinib: 7/19 (37%) HER2+ cases and 10/72 (14%) HER2- cases<br />

were Ki67 responders. Thus while the proportion of Ki67 responders<br />

was higher for HER2+ disease there was a similar or higher absolute<br />

number of responders with HER2- disease. All of the following relates<br />

to patients with HER2- disease. None of the pretreatment levels<br />

of ER, PgR, pAKT, pERK1/2, EGFR, epir, amphir or neur were<br />

associated with Ki67 response (p>0.20). However, HER3 (p=0.01)<br />

and HER2 (p=0.06) mRNA levels were associated with greater Ki67<br />

response. There was a tendency for Ki67 response to be greater with<br />

lower baseline Ki67 (p=0.07). Multivariate analysis showed only<br />

HER3 mRNA levels to be independently significant. HER2 and<br />

HER3 mRNA levels were highly correlated (rho=0.67, p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

histomorphometry and measurement of serum bone markers. Presence<br />

of tumour cells in bone was detected by multiphoton microscopy.<br />

Results: OVX induced significant bone loss compared to sham<br />

within 2 weeks, which was completely inhibited by ZOL. Tumour<br />

growth was detected in the long bones of 29% of animals in the sham<br />

group on day 35 following tumour cell inoculation, and this was not<br />

affected by ZOL treatment. In contrast, 83% of the OVX animals<br />

had detectable tumours in the long bones, demonstrating that OVX<br />

mediated changes to the bone microenvironment that stimulated<br />

tumour growth. Crucially, this OVX-induced increase in tumour<br />

growth was completely prevented by weekly ZOL treatment, with<br />

only 17% of animals in the OVX+ZOL group having detectable bone<br />

tumours compared to 83% in the OVX+saline group. Multiphoton<br />

microscopy revealed that numerous individual tumour cells were<br />

present in long bones of animals in the OVX+ZOL group, supporting<br />

that ZOL acts by inhibiting an OVX-mediated trigger of tumour cell<br />

proliferation.<br />

Conclusions: Our data are in agreement with those reported from<br />

clinical trials of adjuvant zoledronic acid, showing an anti-cancer<br />

effect exclusively in the post-menopausal setting. This is the first<br />

demonstration that zoledronic acid prevents tumour progression in<br />

bone specifically through inhibition of OVX-induced proliferation<br />

of tumour cells resident in the bone marrow.<br />

PD07-09<br />

Zoledronate versus ibandronate comparative evaluation (ZICE)<br />

trial - first results of a UK NCRI 1,405 patient phase III trial<br />

comparing oral ibandronate versus intravenous zoledronate in<br />

the treatment of breast cancer patients with bone metastases.<br />

Barrett-Lee PJ, Casbard A, Abraham J, Grieve R, Wheatley D,<br />

Simmons P, Coleman R, Hood K, Griffiths G, Murray N. Velindre NHS<br />

Trust, Cardiff, Wales, United Kingdom; Cardiff University School<br />

of Medicine, Cardiff, Wales, United Kingdom; University Hospital,<br />

Coventry, England, United Kingdom; Royal Cornwall Hospital,<br />

Truro, England, United Kingdom; University Hospital, Southampton,<br />

England, United Kingdom; Weston Park Hospital, Sheffield, England,<br />

United Kingdom; Royal Adelaide Hospital, Adelaid, South Australia,<br />

Australia<br />

Introduction Bone metastases in patients with breast cancer have<br />

serious effects on health including pain, poor mobility, skeletal<br />

fractures, spinal cord compression and the need for radiotherapy/<br />

surgery. The introduction of intravenous (IV) bisphosphonates, such<br />

as zoledronic acid (Z) has significantly delayed the onset of skeletalrelated<br />

events (SRE). However, prolonged IV bisphosphonates place<br />

burdens upon patient and hospital, and can also cause renal and<br />

acute phase toxicities. Ibandronic acid (I), a third generation aminobisphosphonate<br />

in its oral form has previously been compared with<br />

placebo and was shown to be well tolerated and effective. Indirect<br />

comparisons with IV Z indicated similar efficacy in reducing bone<br />

events, but adverse events were overall comparable with placebo.<br />

One might therefore assume that oral ibandronate would be more<br />

acceptable to patients, and the ZICE Trial is the only large scale direct<br />

randomised comparison between IV Z and oral I to report. Methods<br />

Between January 2006 and October 2010, 1405 newly diagnosed<br />

metastatic breast cancer patients with proven bone metastases were<br />

randomised 1:1 to IV Z (4mg 15 min infusion every 3-4 weeks) or<br />

oral I (50mg per day) for up to 96 weeks. All patients were prescribed<br />

daily calcium & vitamin D supplementation, and patients with current<br />

active dental problems including infection were excluded. Patients<br />

also received chemotherapy, and or endocrine therapy as determined<br />

by their physician. The primary objective was to demonstrate noninferiority<br />

of oral I in comparison with IV Z in terms of the SRE rate,<br />

defined as the number of SREs reported per year (using multiple event<br />

analysis). Secondary endpoints included time to 1st SRE, proportion<br />

of patients with SRE, Pain Scores, side effect profiles including ONJ<br />

and renal toxicities, quality of life and Health resources and overall<br />

survival. The trial was run under the auspices of the NCRI, sponsored<br />

by Velindre NHS Trust, coordinated by the Wales <strong>Cancer</strong> Trials<br />

Unit, funded by an educational grant from Roche and peer reviewed/<br />

endorsed by <strong>Cancer</strong> Research UK (CRUKE/04/022). Results At the<br />

time of this analysis the last randomised patient had completed 96<br />

weeks of therapy, median follow up was 18.4 months and total number<br />

of SREs was 865 (468 in I and 397 in Z). For the primary objective,<br />

the SRE rate was 0.543 and 0.444 in I and Z groups respectively<br />

(Hazard ratio, 1.22; 95% CI, 1.04 to 1.45; P = .017). Ibandronate<br />

failed to meet the criteria for non-inferiority to Zoledronate, but was<br />

similar in delaying time to first SRE (hazard ratio, 1.11; 95% CI, 0.94<br />

to 1.31; P = .233). Overall survival (disease progression), was very<br />

similar between groups but renal AEs occurred more frequently with<br />

Z than I; Compliance with oral therapy was 82%. ONJ rate was very<br />

low in both arms (0.71%, I; 1.29%, Z; P = 0.28). Conclusion Oral I<br />

is inferior to Z in terms of the SRE rate in metastatic breast cancer<br />

patients with bone metastases, but is similar to Z in delaying time<br />

to first SRE. Both drugs had acceptable safety profiles, with adverse<br />

events consistent with those reported previously.<br />

PD08-01<br />

Utilization of Oncotype DX in an inner-city population: Race<br />

or Place?<br />

Guth AA, Fineberg S, Fei K, Franco R, Bickell N. NYU School of<br />

Medicine, New York, NY; Montefiore Medical Center, Bronx, NY;<br />

Mount Sinai School of Medicine, New York, NY<br />

Introduction: While women with early stage breast cancer frequently<br />

receive adjuvant chemotherapy to prevent recurrence, not all patients<br />

benefit from and require it. Oncotype DX(ODX) is a validated<br />

genomic predictor of outcome and response to chemotherapy in<br />

estrogen receptor (ER) positive, node-negative breast cancer. The<br />

assay has the potential to enable women to avoid unnecessary<br />

chemotherapy. There is little data available on use in minority/<br />

economically disadvantaged women.<br />

Methods: Women with an early-stage breast cancer surgically treated<br />

between 2006-2009 participated in a randomized controlled trial of<br />

patient assistance to reduce disparities in treatment.<br />

Results: Of 374 women, 225 had ER+ tumors stage 1B or greater<br />

eligible to undergo ODX testing. Chi-square/t tests and logistic models<br />

for multivariable comparisons. Race and municipal hospital were<br />

highly correlated, only 3 white women went to municipal hospitals.<br />

Because there was no racial difference in testing within tertiary<br />

centers, and only one woman in municipal hospitals got tested, we<br />

used hospital type rather than race in the final model. 23% (52/225)<br />

of women underwent ODX. Women tested had earlier stage cancer,<br />

higher income, were white and treated at tertiary referral centers.<br />

There was no racial difference in receipt of ODX among women<br />

treated at referral centers. Black and Hispanic women as compared<br />

to white women were more likely to be treated at municipal hospitals<br />

(29% vs 32% vs 3%; p


December 4-8, 2012 <strong>Abstract</strong>s: Poster Discussion 8<br />

19/52 (37%) had an intermediate score, and 9/52 (17%) had a high<br />

recurrence risk score, with no racial difference. All 9 patients at high<br />

risk (100%) were treated with chemotherapy; 58% at intermediate<br />

risk; only 4% at low risk were treated with chemotherapy.<br />

Discussion: The most important factors influencing ODX testing<br />

were tumor stage and care at a municipal hospital. Women treated<br />

at municipal hospitals were unlikely to get ODX. When Medicaid<br />

coverage became available for ODX, minority women at tertiary<br />

centers began to undergo testing but not municipal hospitals.<br />

This suggests adoption of ODX was slower in municipal than tertiary<br />

referral hospitals resulting in apparent racial disparity even after<br />

insurance coverage would have ensured equal access and saved<br />

unnecessary chemotherapy costs to patients and hospitals. Thus, the<br />

racial disparity in ODX testing reflected the site of care and not race<br />

since women were treated equally at the tertiary referral centers, with<br />

no racial disparity in those sites. These results reflect that diffusion<br />

of innovation lags at hospitals treating minority and underprivileged<br />

patients compared to tertiary referral centers.<br />

As Oncotype DX influences treatment and can reduce overuse of<br />

chemotherapy, it is imperative to implement innovations that improve<br />

quality care at safety net hospitals. Such approaches have the potential<br />

to reduce racial and socioeconomic disparities in cancer care.<br />

PD08-02<br />

Disparities in the Utilization of Reconstruction after Mastectomy:<br />

The California Teachers Study<br />

Kruper L, Xu XX, Bernstein L, Henderson K. City of Hope <strong>Cancer</strong><br />

Center, Duarte, CA<br />

Background: For breast cancer patients undergoing mastectomy,<br />

factors such as insurance status, race/ethnicity, age, and type of<br />

hospital influence whether post-mastectomy reconstruction (PMR)<br />

is performed. This study was undertaken to determine if additional<br />

patient variables and clinicopathologic features also influence the<br />

utilization of PMR using the California Teachers Study (CTS).<br />

Methods: Patients were identified from the CTS, a cohort of<br />

approximately 133,000 female public school teachers and<br />

administrators, followed prospectively from 1995 forward to<br />

investigate exposures associated with incident cancer and other<br />

outcomes. All in situ and invasive breast cancers were identified<br />

through linkage with the California <strong>Cancer</strong> Registry, as well<br />

as with the California Office of Statewide Health Planning and<br />

Development (OSHPD) hospital discharge database to determine<br />

the rates of mastectomy with and without reconstruction. Patterns<br />

in PMR rates were examined by calendar year, age, race/ethnicity,<br />

type of insurance, type of hospital, tumor stage, body mass index<br />

(BMI), family history of breast cancer (FH), smoking history,<br />

physical activity, and prior breast implant status using a chi-square<br />

test. Univariable and multivariable-adjusted odds ratios (OR) with<br />

95% confidence intervals (CI) were estimated for relative odds of<br />

immediate reconstruction vs. mastectomy only.<br />

Results: During follow-up, 1,253 CTS participants with incident<br />

breast cancer underwent mastectomy with (N=368) and without<br />

(N=885) reconstruction. In multivariable stepwise logistic regression<br />

analyses, calendar year, age, type of insurance, tumor stage, and prior<br />

breast implant were statistically significantly associated with use of<br />

reconstruction. The proportion of patients undergoing immediate<br />

PMR increased from 21.8% during 1995-1999 to 26.4% during 2005-<br />

2009. A statistically significant dose-response was apparent between<br />

older age at surgery and decreased likelihood of post-mastectomy<br />

reconstruction (Ptrend


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

(n=10, 27%) or feeling unprepared to BRS (n=9, 24%). Five patients<br />

(13%) considered BRS as “useless”: 4 of them were ≥65 and age was<br />

the main factor in their choice. Vulnerability was not correlated with<br />

the decisions about BRS, neither was marital status. Of note, for all<br />

patients but one financial difficulties were not regarded as a critical<br />

issue and all pts were aware that BRS can be reimbursed by the public<br />

health insurance; none of them, however, knew that extra fees were<br />

the rule when BRS is performed outside of public hospitals.<br />

CONCLUSIONS: In an universal healthcare system, only a minority<br />

of low-income or vulnerable patients choose BRS. Although BRS<br />

funding is usually not regarded as a problem, numerous barriers to<br />

BRS still exist, mainly related to irrational grounds or wrong beliefs.<br />

Whether these barriers can be overcome by improved patient-doctor<br />

communication or better information remains an issue.<br />

PD08-04<br />

Factors which affect surgical management in an underinsured,<br />

county hospital population<br />

Komenaka IK, Olsen L, Klemens AE, Hsu C-H, Nodora J, Martinez<br />

ME, Thompson PA, Bouton M. Maricopa Medical Center, Phoenix,<br />

AZ; University of Arizona, Tucson, AZ; Moores <strong>Cancer</strong> Center,<br />

University of California, <strong>San</strong> Diego, CA<br />

Background: Significant variation exists between institutions in the<br />

use of lumpectomy, mastectomy, and reconstruction. Much less is<br />

known about minorities and populations outside the large academic<br />

institutions. The current study was performed to evaluate variables<br />

that affect patient choice in surgical management in a county hospital<br />

population.<br />

Methods: A retrospective review of all patients seen at the county,<br />

safety net institution with breast cancer from January 2010 to May<br />

2012. Sociodemographic, clinical, and treatment variables were<br />

evaluated. Univariate analysis was performed to identify variables<br />

which were associated with type of operation. All of the variables with<br />

a p-value


December 4-8, 2012 <strong>Abstract</strong>s: Poster Discussion 9<br />

<strong>Antonio</strong>, Texas (P30-CA054174), Susan G. Komen for the Cure (POP<br />

2000 704), and the National <strong>Cancer</strong> Institute via Redes En Acción<br />

(U01-CA86117 and U54 CA153511-01).<br />

PD08-06<br />

Significant Clinical impact of recurrent BRCA1 and BRCA2<br />

(BRCA) mutations in Mexico<br />

Villarreal-Garza C, Herrera LA, Herzog J, Port D, Mohar A,<br />

Perez-Plasencia C, Clague J, Alvarez RMa, <strong>San</strong>tibanez M, Blazer<br />

KR, Weitzel JN. Instituto Nacional de <strong>Cancer</strong>ologia, Mexico City,<br />

Mexico DF, Mexico; Instituto Nacional de <strong>Cancer</strong>ologia – Instituto<br />

de Investigaciones Biomedicas, UNAM, Mexico City, Mexico DF,<br />

Mexico; City of Hope, Duarte, CA<br />

Background: <strong>Breast</strong> cancer (BC) is the most commonly diagnosed<br />

cancer and the leading cancer cause of death in Hispanic women.<br />

Although the incidence of breast cancer in Hispanics is less than<br />

that for non-Hispanic white women, our studies on the prevalence of<br />

deleterious mutations in the BRCA genes among high-risk Hispanics<br />

in the US suggest that BRCA mutations may account for a higher<br />

proportion of BC in this population. However, a lack of research<br />

on BRCA mutations in Hispanics has limited the implementation of<br />

prevention efforts and the scope of comparative studies of genetic<br />

factors that influence BC and ovarian cancer (OC) risk within Hispanic<br />

populations – this is especially true of Mexico where there has been<br />

little access to clinical BRCA gene analyses.<br />

Objective/Methods: We used an economic panel assay (HISPANEL)<br />

on a mass spectroscopy platform (Sequenom) to analyze DNA from<br />

189 cancer cases (92 unselected OC; 97 BC) from the National <strong>Cancer</strong><br />

Institute in Mexico City for recurrent BRCA mutations, including a<br />

large rearrangement (BRCA1 ex9-12del) that we hypothesize is a<br />

Mexican founder mutation.<br />

Results: Overall, 14% (27/189) harbored a germline BRCA mutation<br />

(25 BRCA1, 2 BRCA2) detected by the HISPANEL (17% in OC; 11%<br />

in BC). BRCA1 ex9-12del was detected in 9.8% (9/92) of unselected<br />

OC cases, representing 56% of the 16 BRCA mutations. It also<br />

represented 36% (4/11) of the BRCA mutations detected in the BC<br />

cases. Mean age at onset of BC for BRCA-associated cases (n=11)<br />

was 46 years old (range 31-63); 8/11 were triple negative BC (TNBC)<br />

– a BRCA mutation was detected in 24% (8/34) of all TNBC cases.<br />

Conclusions: The remarkable frequency of the BRCA1 ex9-12del<br />

mutation, accounting for 56% of BRCA mutations in the OC cases<br />

and 36% of BC cases, supports our hypothesis of regional origin<br />

of this Mexican founder mutation approximately 1,474 years ago.<br />

Similar to our experience in the Mexican American population, the<br />

HISPANEL, which includes recurrent BRCA mutations found in<br />

women of Hispanic ancestry, appears to have high sensitivity and<br />

thus is likely to have clinical utility while reducing overall genotyping<br />

cost among underserved women in Mexico. Both in the US Hispanic<br />

populations and in Mexico, the prevalence of the Mexican founder<br />

mutation make it a significant public health issue, and presents an<br />

opportunity for cost effective in BC and OC prevention.<br />

PD09-01<br />

BRCA1 inactivation induces NF-κB in human breast cancer cells<br />

and in murine and human mammary glands<br />

Sau A, Arnaout A, Pratt C. University of Ottawa, ON, Canada; Ottawa<br />

Hospital, Ottawa, ON, Canada<br />

Understanding the biological mechanisms underlying the initiation<br />

and progression of breast cancer it is an important step for its<br />

prevention and treatment. In 2011 in the United States, approximately<br />

230,000 women were diagnosed with breast cancer and 40,000<br />

died. Individuals with mutations in breast cancer-associated gene 1<br />

(BRCA1) have a lifetime risk of developing breast cancer up to 85%.<br />

It is well known that BRCA1 participates in DNA damage repair and<br />

cell cycle checkpoint control, serving as a tumor suppressor gene to<br />

maintain the global genomic stability. However, BRCA1 has also been<br />

shown to play a key role in maturation of mammary stem/progenitor<br />

cells, which are the targets for carcinogenesis in individuals who have<br />

undergone loss of heterozygosity (LOH) for BRCA1. Recently, it has<br />

also been shown that NF-κB activity is increased in both mammary<br />

carcinoma cell lines and primary human breast cancer tissue. Indeed,<br />

in a previous study it has been demonstrated that NF-κB inducible<br />

kinase (NIK), p100/p52 and RelB (all components of the alternative<br />

NF-κB pathways) were increased in BRCA1-mutated tumors.<br />

Here we show that BRCA1-loss or -mutation is responsible for<br />

activation of the alternative NF-κB pathway evidenced by NIK and<br />

IκB kinase-α (IKKα) phosphorylation, processing of p100 to p52<br />

and p52/RelB nuclear localization. Moreover, increased p52 was<br />

also observed after BRCA1 inhibition. A BRCA1-mutated human<br />

breast cancer cell line (HCC1937) was also used to understand<br />

the role played by NIK in NF-κB alternative pathway activation.<br />

Indeed, NIK inhibition in HCC1937 cell line resulted in a decrease<br />

in p52 formation. Moreover, a decrease in NIK mRNA level was also<br />

observed when wild-type BRCA1 was reconstituted in HCC1937<br />

cells. BRCA1 inactivation in MCF-7 cells also induced NIK<br />

phosphorylation and nuclear localization of RelB and p52. Overall,<br />

these data show that inactivation of BRCA1 increases NIK mRNA<br />

level, associated with induction of the NF-κB alternative pathway.<br />

Stem/progenitors cells sorted using the CD24/CD49f<br />

immunophenotype derived from BRCA1 knockout mouse mammary<br />

glands showed alternative NF-κB pathway activation. Inhibition of<br />

IKKα/β using BMS-345541 completely blocked mammary colony<br />

formation in a Matrigel assay. Moreover, increased p52 formation<br />

was found in mammary stem/progenitor cells and mammary gland<br />

paraffin sections obtained from BRCA1 knockout mice. Remarkably,<br />

RelB and p100/p52 were highly expressed in 20-50% of the lobular<br />

structures in histologically normal breast tissue obtained from human<br />

BRCA1 mutation carriers while no staining was evident in normal<br />

tissue from non-carrier mastectomy samples.<br />

Our data show that BRCA1 inactivation induces alternative NF-κB<br />

activation which ultimately promotes the expansion of the mammary<br />

progenitor population. These novel findings provide a new basis for<br />

functional classification of BRCA1 mutations and a potential method<br />

for predicting breast cancer in BRCA1 mutation carriers. Lastly our<br />

results suggest that targeting the alternative NF-κB pathway could<br />

be of benefit in the prevention of BRCA1-associated breast cancer<br />

by limiting progenitor cell expansion.<br />

PD09-02<br />

BRCA1 insufficiency is predictive of superior survival in patients<br />

with triple negative breast cancer treated with platinum based<br />

chemotherapy<br />

Sharma P, Stecklein S, Kimler BF, Klemp JR, Khan QJ, Fabian CJ,<br />

Tawfik OW, Connor CS, McGinness MK, Mammen JMW, Jensen RA.<br />

University of Kansas Medical Center, Westwood, KS; University of<br />

Kansas Medical Center, Kansas City, KS<br />

Introduction: Triple negative breast cancer (TNBC) and BRCA1-<br />

associated breast cancers share many histopathologic and molecular<br />

features. BRCA1 plays a crucial role in HR-dependent DNA repair<br />

and BRCA1-deficient cells are particularly susceptible to the DNA<br />

damaging agents like platinums. Increasing evidence suggests that<br />

in addition to germline BRCA defects, other mechanisms (like<br />

www.aacrjournals.org 139s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

epigenetic BRCA1 silencing) can lead to BRCA1 insufficiency in<br />

TNBC. However, the impact of BRCA1 insufficiency on the efficacy<br />

of DNA damaging agents in TNBC is not known.<br />

Aim: To investigate the impact of BRCA1 insufficiency on relapsefree<br />

survival (RFS) and overall survival (OS) in patients with stage<br />

II-III TNBC treated with neoadjuvant platinum-based chemotherapy.<br />

BRCA1 insufficiency (BRCA1 insuf ) state was defined as presence of<br />

germline BRCA1/2 mutation or BRCA1 promoter methylation (PM)<br />

and/or low BRCA1 expression (lowest quartile).<br />

Methods: Thirty patients with stage II/III TNBC received neoadjuvant<br />

chemotherapy (6 cycles of Carboplatin AUC 6, Docetaxel 75mg/m 2<br />

and Erlotinib 150 mg PO) on a phase II trial between 8/2007- 6/2010.<br />

All but one patient underwent comprehensive BRCA analysis (Myriad<br />

Genetic Laboratories). Pre-treatment tumor specimens were used<br />

for evaluation of BRCA1 PM and expression. Genomic DNA was<br />

isolated from FFPE samples, bisulfite converted and then subjected<br />

to methylation-specific PCR (MSP). RNA was isolated, reverse<br />

transcribed to cDNA and assayed by quantitative real-time PCR<br />

(qRT-PCR) for determination of BRCA1 mRNA transcript levels.<br />

RFS and OS were estimated according to the Kaplan-Meier method<br />

and compared among groups with log-rank statistic. Cox proportional<br />

hazards models were fit to determine the association of BRCA1 insuf<br />

with the risk of death after adjustment for other characteristics.<br />

Results: Median age: 51yrs, African American: 20%, Median<br />

tumor size: 3.3 cm, LN positive: 40%. Six of 30 patients (20%)<br />

harbored germline BRCA mutation (4 BRCA1, 2 BRCA2). Baseline<br />

tumor specimen was available for 26/30 patients. BRCA1 MSP was<br />

successful in 92% and BRCA1 qRT-PCR was successful in 84%<br />

of specimens. BRCA1 PM and low BRCA1 expression was present<br />

in 30% and 15% of subjects, respectively. There was evidence<br />

of BRCA1 insuf in 53% (16/30) of subjects. At a median time from<br />

diagnosis of 42 months (range, 23-59 months) there have been<br />

9(30%) recurrences and 7(23%) deaths. On univariate analysis node<br />

negativity, lower stage and presence of BRCA1 insuf were associated<br />

with better OS. At the median follow up, RFS is 81% for patients with<br />

BRCA1 insuf versus 54% for patients without BRCA1 insuf (p =0.16) ; OS<br />

is 83% for patients with BRCA1 insuf versus 46% for patients without<br />

BRCA1 insuf (p =0.021). After adjustment for clinical variables patients<br />

with BRCA1 insuf had a significantly better OS compared to patients<br />

without BRCA1 insuf (p=0.036).<br />

Conclusions: Germline BRCA testing plus tissue BRCA1 PM /<br />

expression can be used to identify a BRCA1 insuf sub-population within<br />

TNBC demonstrating a favorable outcome with platinum treatment.<br />

This BRCA1 insuf criteria can be easily used to select TNBC patients<br />

likely to benefit from DNA damaging agents like platinums and<br />

PARP inhibitors.<br />

PD09-03<br />

Impact of BRCA1/2 Mutation Status in TBCRC009: A multicenter<br />

phase II study of cisplatin or carboplatin for metastatic triple<br />

negative breast cancer.<br />

Isakoff SJ, Goss PE, Mayer EL, Traina T, Carey LA, Krag KJ, Liu<br />

MC, Rugo H, Stearns V, Come S, Finkelstein D, Hartman A-R, Garber<br />

JE, Ryan PD, Winer EP, Ellisen LW. Massachusetts General Hospital,<br />

Boston, MA; Dana-Farber <strong>Cancer</strong> Institute, Boston, MA; Memorial<br />

Sloan-Kettering <strong>Cancer</strong> Center, New York, NY; University of North<br />

Carolina Lineberger Comprehensive <strong>Cancer</strong> Center, Chapel Hill, NC;<br />

Mass General/North Shore <strong>Cancer</strong> Ctr, Danvers, MA; Georgetown<br />

Lombardi Comprehensive <strong>Cancer</strong> Center, Washington, DC; University<br />

of California, <strong>San</strong> Francisco Helen Diller Family Comprehensive<br />

<strong>Cancer</strong> Center, <strong>San</strong> Francisco, CA; Sidney Kimmel Comprehensive<br />

<strong>Cancer</strong> Center at Johns Hopkins University, Baltimore, MD; Beth<br />

Israel Deaconess Medical Center, Boston, MA; Myriad Genetics, Salt<br />

Lake City, UT; Fox Chase <strong>Cancer</strong> Center, Philadelphia, PA<br />

Background: Triple negative breast cancer (TNBC) has a poor<br />

prognosis compared to other BC subtypes and lacks identified specific<br />

therapeutic targets. TNBC is frequent among BRCA1-associated<br />

breast cancer. Platinum chemotherapy has activity in TNBC, and<br />

several studies have suggested particular efficacy of platinum agents<br />

in BRCA-associated breast cancer. TBCRC009 is a multicenter single<br />

arm phase II study of single agent platinum in metastatic TNBC.<br />

Here we sought to evaluate the impact of BRCA1/2 mutation status<br />

on outcomes in this study population.<br />

Methods: Eligible metastatic TNBC patients (pts) had measurable<br />

disease, available archival tumor, ≤ 1 prior metastatic therapy, and<br />

no prior platinum chemotherapy. Pts were assigned by physician<br />

choice (limited to equal enrollment in each arm) to cisplatin 75mg/<br />

m2 or carboplatin AUC=6 every 21 days with optional extension to<br />

28 days after cycle 1. Co-primary endpoints were: 1) Response Rate<br />

(RR) and 2) whether tumor p63/p73 expression predicts response.<br />

Secondary endpoints included determination of BRCA1/2 mutation<br />

status and exploratory analysis of correlation of BRCA1/2 status<br />

with outcomes. Baseline blood samples from all pts were analyzed<br />

by Myriad Genetics using BRCAnalysis and BART testing. Results<br />

of this exploratory analysis are presented here.<br />

Results: 86 TNBC pts enrolled between June 2007-Oct 2010. Results<br />

of primary endpoint 1 and patient characteristics were previously<br />

presented (ASCO 2011 Annual Meeting, J Clin Oncol 29: 2011 suppl;<br />

abstr 1025). In summary, median age=52 (30-78), 64% had ECOG<br />

PS=0, and 86% had adjuvant chemotherapy. Overall response rate<br />

(RR) was 30.2% (95% CI 20.8% - 41.1%), including 4 CR (4.7%),<br />

22 PR (25.6%). Among the 76 pts with BRCA1/2 status available,<br />

there were 11 (14.5%) BRCA1/2 carriers (9 BRCA1, 2 BRCA2)<br />

and 65 (85.5%) BRCA1/2 wild type (wt). Testing is ongoing for the<br />

remaining 10 pts. RR was 54.6% (5 PR, 1 CR) in BRCA1/2 carriers<br />

and 26.2% (14 PR, 3 CR) in BRCA1/2 wt (p=0.0506). There was no<br />

significant difference in PFS between BRCA1/2 carriers (median PFS<br />

99 days; 95% CI: 40-215) and BRCA1/2 wt (median PFS 86 days,<br />

95% CI: 46-146). Interestingly, 6 pts (7%), including 5 BRCA1/2 wt<br />

and 1 BRCA unknown, remain without progression for 53, 41, 33, 27,<br />

25, and 22 months as of June 2012. Of these 6 pts, 3 pts discontinued<br />

study drug due to toxicity and received no additional therapy and 3<br />

pts had major responses and had consolidation therapy with surgery,<br />

chemotherapy, or radiation.<br />

Conclusion: First or second line single agent platinum is effective<br />

in both BRCA1/2-associated and sporadic metastatic TNBC. In this<br />

exploratory analysis, we observed a higher RR in BRCA1/2 carriers<br />

than in BRCA wt patients, but we did not observe an improved PFS<br />

in this small cohort. We observed durable responses in 7% of pts<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

140s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Discussion 9<br />

which did not correlate with BRCA1/2 mutations. The lack of a<br />

clear relationship between the higher RR in patients with germline<br />

BRCA1/2 mutations and PFS is surprising and could be further<br />

elucidated by studies in a larger cohort, and our ongoing analyses of<br />

additional markers for platinum sensitivity.<br />

PD09-04<br />

Homologous Recombination Deficiency (HRD) score predicts<br />

pathologic response following neoadjuvant platinum-based<br />

therapy in triple-negative and BRCA1/2 mutation-associated<br />

breast cancer (BC)<br />

Telli ML, Jensen KC, Abkevich V, Hartman A-R, Vinayak S, Lanchbury<br />

J, Gutin A, Timms K, Ford JM. Stanford University School of<br />

Medicine, Stanford, CA; Myriad Genetics, Salt Lake City, UT<br />

Background: BC that arises in BRCA1/2 mutation carriers is<br />

characterized by defective homologous recombination DNA repair.<br />

Given the clinical potential for DNA repair-targeted therapeutics,<br />

Myriad Genetics has developed a tumor DNA-based assay that<br />

can identify underlying defects in the homologous recombination<br />

pathway, regardless of etiology, including identification of BRCA1/2<br />

mutation carriers with high sensitivity. This study was designed to<br />

assess ability of the HRD score to predict pathologic response to<br />

neoadjuvant platinum-based therapy in early-stage triple-negative<br />

and BRCA1/2 mutation-associated BC.<br />

Methods: The HRD score was assessed in a cohort of 55 fresh<br />

frozen tumors from pts with clinical stage I-IIIA triple-negative<br />

and BRCA1/2 mutation-associated BC enrolled onto a single-arm,<br />

phase II study of neoadjuvant gemcitabine, carboplatin and iniparib.<br />

Following neoadjuvant therapy (4 cycles=11; 6 cycles=44), pathologic<br />

response was assessed using the residual cancer burden (RCB) index<br />

by a central pathologist. Twenty-nine tumors were obtained from<br />

responders (RCB 0/1) and 26 tumors were from non-responders (RCB<br />

2/3). The HRD score was derived by count of the number of LOH<br />

regions (>15 Mb and < whole chromosome) observed in the tumor<br />

genome. Association of response to treatment and HRD score was<br />

performed by the Mann-Whitney U test.<br />

Results: Germline BRCA1/2 mutation status was known in all pts<br />

and 13/55 tumors were from carriers of deleterious BRCA1 (n=9),<br />

BRCA2 (n=3) or BRCA1&2 (n=1) mutations. Two mutation carriers<br />

had ER+/PR+(>5%) BC. The mean HRD score for responders is 16.5<br />

and no differences were noted between germline BRCA1/2 mutants<br />

vs. non-mutants. The mean HRD score for non-responders is 11.4.<br />

Among carriers of BRCA1/2 mutations, 12/13 achieved a favorable<br />

response. The one BRCA1 mutation carrier non-responder with<br />

TNBC had a low HRD score of 8. This pt was previously treated with<br />

chemotherapy twice 12 years prior for primary and locally recurrent<br />

contralateral TNBC. The p-value for association between response<br />

to treatment and HRD score by the Mann-Whitney U test is 0.004.<br />

If BRCA1/2 deficient samples are excluded, the association between<br />

response to treatment and HRD score remains significant (p=0.02).<br />

Multivariate analysis that includes HRD score, BRCA1/2 genotype,<br />

age, stage, tumor grade, PAM50 subtype, ER/PR status, and cycles<br />

of therapy will be presented together with data from an additional 35<br />

pts. Enrollment to the therapeutic trial is complete and final clinical<br />

results will be separately presented.<br />

Conclusion: The HRD score is significantly correlated with pathologic<br />

response to platinum-based chemotherapy in early-stage triplenegative<br />

and BRCA1/2 mutation-associated BC.<br />

PD09-05<br />

Single nucleotide polymorphism of XRCC1 which participates in<br />

DNA repair mechanism predicts clinical outcome in relapsed or<br />

metastatic breast cancer patients treated with S1 and oxaliplatin<br />

chemotherapy: Results from multicenter prospective study<br />

(TORCH_KCSG BR07-03)<br />

Im S-A, Oh D-Y, Keam B, Lee KS, Ahn J-H, Sohn J, Ahn JS, Kim JH,<br />

Lee MH, Lee KE, Kim HJ, Lee K-H, Han SW, Kim S-Y, Kim SB, Im Y-H,<br />

Ro J, Park H-S. Seoul National University Hospital, Seoul, Korea;<br />

National <strong>Cancer</strong> Center, Goyang, Korea; Asan Medical Center, Seoul,<br />

Korea; Yonsei University College of Medicine, Severance Hospital,<br />

Seoul, Korea; Samsung Medical Center, Seoul, Korea; Seoul National<br />

University Bundang Hospital, Seongnam, Korea; Inha University<br />

Hospital, Incheon, Korea; Ewha Womans University Medical Center,<br />

Seoul, Korea; Hallym University Sacred Heart Hospital, Anyang,<br />

Korea; Kyung-Hee University Hospital, Seoul, Korea; Soon Chun<br />

Hyang University Hospital, Seoul, Korea<br />

Background: S1 and oxaliplatin (SOX) combination chemotherapy is<br />

an effective regimen in anthracycline and taxane pretreated metastatic<br />

breast cancer (MBC) patients with manageable toxicities (KCSG<br />

BR07-03, SABCS 2011 #Abst P3-16-06). The aim of this study was<br />

to investigate the association of the single nucleotide polymorphisms<br />

(SNPs) and clinical outcome in MBC treated with SOX chemotherapy.<br />

Patients and Methods: A total of 87 MBC patients previously treated<br />

with or resistant to anthracycline and taxane chemotherapy were<br />

enrolled in this prospective multicenter trial. The patients received<br />

S-1 80mg/m 2 /day (day 1-14) and oxaliplatin 130 mg/m 2 (day 1)<br />

every 3 weeks till progression. Among the 87 patients, 77 patients<br />

were available for SNP analysis. Germline DNA from peripheral<br />

blood (PB) mononuclear cells was extracted. SNPs in 4 genes from<br />

pathways that may influence cellular sensitivity to S1 and oxaliplatin<br />

(TS, ERCC, XPD, and XRCC) were genotyped from PB sample using<br />

PCR-restriction fragment length polymorphism.<br />

Results: Overall response rate (RR) was 38.5% (95% CI: 27.7-49.3)<br />

and disease control rate was 67.9% (95% CI:57.5-78.3) to SOX.<br />

Median time-to-progression (TTP) and overall survival (OS) were<br />

6.0 mo (95% CI: 5.1-6.9 mo) and 19.4 mo (95% CI: not estimated),<br />

respectively. XRCC1 Arg194Trp SNP which participates in DNA<br />

repair mechanism showed correlation with the clinical outcome. RR<br />

was tend to higher in XRCC1 Arg194Trp CC genotype compared with<br />

CT or TT genotype (50.0 % vs 35.1% or 12.5%, P=0.121). TTP of<br />

patients with CC genotype in XRCC1 Arg194Trp was significantly<br />

longer than the TTP of patients with CT or TT genotype (median<br />

TTP: 6.4 mo in CC, 5.9 mo in CT, 3.0 mo in TT, P=0.007) as well as<br />

overall survival (OS) (median OS: not reached in CC, 13.9 mo in CT,<br />

7.1 mo in TT, P=0.006). After adjusting for hormone receptor status,<br />

performance status, and visceral involvement, prognostic value of<br />

XRCC1 Arg194Trp SNP remained significant (Hazard Ratio=1.322<br />

and 4.484, P=0.016). Other SNPs were not significantly associated<br />

with survival or toxicities.<br />

Conclusion: XRCC1 Arg194Trp SNP is associated with clinical<br />

outcome of MBC patients treated with SOX chemotherapy. Further<br />

studies of the relationship between germline polymorphisms in<br />

XRCC1 and functional mechanism researches are warranted.<br />

www.aacrjournals.org 141s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

PD09-06<br />

Two phase I trials exploring different dosing schedules of<br />

carboplatin (C), paclitaxel (P), and the poly-ADP-ribose<br />

polymerase (PARP) inhibitor, veliparib (ABT-888) (V) with<br />

activity in triple negative breast cancer (TNBC)<br />

Puhalla SL, Appleman LJ, Beumer JH, Tawbi H, Stoller RG,<br />

Owonikoko TK, Ramalingam SS, Belani CP, Brufsky AM, Abraham<br />

J, Shepherd SP, Giranda V, Chen AP, Chu E. University of Pittsburgh<br />

<strong>Cancer</strong> Institute, Pittsburgh, PA; Winship <strong>Cancer</strong> Institute of Emory<br />

University, Atlanta, GA; West Virginia University <strong>Cancer</strong> Center,<br />

Morgantown, WV; Penn State Hershey <strong>Cancer</strong> Institute, Hershey,<br />

PA; National <strong>Cancer</strong> Institute, Bethesda, MD; Abbott Laboratories,<br />

Abbott Park, IL<br />

Background:<br />

The histopathologic and molecular similarities between TNBC and<br />

BRCA-1 mutation related breast cancer provide rationale for the use<br />

of PARP inhibitors in treating sporadic TNBC. C and P are established<br />

drugs in the treatment of advanced breast cancer. In order to determine<br />

the recommend phase II dose (RP2D) of V in combination with C and<br />

P, two separate phase I trials evaluated the combination of V with C<br />

and P dosed weekly (q1wk) and every 3 weeks (q3wk).<br />

Methods:<br />

Both phase I trials were a standard 3+3 dose escalation design of C, P,<br />

and V open to patients (pts) with advanced solid tumors and enriched<br />

for pts with TNBC. In the q1wk trial, C was given weekly on day 3<br />

at an AUC of 2, P was given weekly on day 3 at a dose of 80 mg/m 2 ,<br />

and V was escalated from 50 mg twice daily (BID) to a maximum<br />

of 200 mg BID on days 1-5 every week over 4 dose levels. In the<br />

q3wk trial, C was given on day 3 every 21 days at an AUC of 6, P<br />

was given on day 3 every 21 days with the dose escalated from 150<br />

mg/m 2 to a dose of 200 mg/m 2 , and V was given on days 1-7 every 21<br />

days and escalated from 10 mg BID to a dose of 120 mg BID over 9<br />

dose levels. Toxicity was assessed by CTCAE v. 4 and response was<br />

determined using RECIST criteria.<br />

Results:<br />

The q1wk trial enrolled a total of 18 pts, 12 of whom had BC, and<br />

10 had TNBC. One TNBC pt was BRCA-1+ and the remainder were<br />

sporadic. The trial will be completed with the enrollment of one<br />

additional patient on dose level 4. Ten breast cancer pts are evaluable<br />

for response. DLT’s included thrombocytopenia on dose levels 3 (V at<br />

150 mg BID) and 4 (V at 200mg BID), and both occurred in TNBC<br />

pts. The range of cycles administered to TNBC pts was 1-11. The<br />

q3wk trial enrolled a total of 75 pts, 14 had BC (12 evaluable for<br />

response), and 9 had TNBC. One of these pts had a BRCA mutation;<br />

2 pts had a variant of undetermined significance. Three DLT’s were<br />

observed on this schedule: febrile neutropenia (C at AUC 6, P at 200<br />

mg/m 2 , V at 40 mg BID, 100 mg BID, and 120 mg BID) and grade<br />

3 hyponatremia. No DLT’s occurred in TNBC pts. The established<br />

RP2D for the q3wk schedule is C (AUC6), P (200mg/m2) and V<br />

(100mg bid) and additional pts are being accrued to an expansion<br />

cohort. The range of cycles administered to TNBC pts was 1-14.<br />

The clinical activity in TNBC is summarized in Table 1. Responses<br />

occurred in pts who had been previously treated both in the adjuvant<br />

and metastatic settings, as well as in those who received prior taxanes.<br />

Clinical Activity in TNBC pts<br />

Regimen Complete responses Partial responses Stable disease No Response<br />

Q1WK C/P/V (n=10) 2 3 3 2<br />

Q3WK C/P/V (n=9) 3 4 1 1<br />

Conclusion:<br />

There is evidence of promising clinical activity and acceptable<br />

tolerability with carboplatin, paclitaxel, and veliparib on both dosing<br />

schedules. An additional 12 pts with TNBC will be accrued to the<br />

q1wk trial with pre- and post-treatment biopsies to further evaluate<br />

underlying mechanisms of sensitivity and response with regards<br />

to DNA repair pathways. Based on our phase I experience, further<br />

randomized trials are warranted in breast cancer, particularly TNBC.<br />

PD09-07<br />

Therapeutic potential of targeting ATM-PELP1-p53 axis in triple<br />

negative breast cancer<br />

Krishnan SR, Nair BC, Sareddy GR, Mann M, Roy SS, Vadlamudi<br />

RK. UTHSCSA, <strong>San</strong> <strong>Antonio</strong>, TX<br />

Triple negative breast cancer (TNBC) comprises approximately 15%<br />

of all breast cancers, lacks expression of ER, PR, and HER2 and is<br />

clinically aggressive with shorter disease free survival. TNBC patients<br />

do not benefit from antiestrogen and herceptin-based therapies.<br />

Evolving evidence suggests that overexpression of mutant p53 is<br />

significantly associated with TNBC progression. DNA damage<br />

response (DDR) is critical for the maintenance of genome stability<br />

and serves as an anti-cancer barrier during tumorigenesis. However,<br />

the role of DDR in tumor progression and metastasis is less known.<br />

Recent studies suggest that the ATM kinase is hyperactive in late stage<br />

breast tumor tissues with lymph node metastasis. Our ongoing studies<br />

have identified proline, glutamic acid, leucine rich protein 1 (PELP1),<br />

a proto-oncogene overexpressed in breast cancer, as a novel substrate<br />

of ATM. The objective of this study is to determine the mechanism<br />

and significance of ATM mediated phosphorylation of PELP1 and<br />

its crosstalk with the p53 pathway in TNBC cells. To test this we<br />

have used three ER-negative mutant p53 breast cancer cell lines<br />

(MDA-MB-231, MDA-MB-468, BT20), with ER-positive (ZR-75<br />

and MCF7) cell lines as controls. Using PELP1 specific shRNAs, we<br />

generated model cells that have stable expression of either control or<br />

PELP1 shRNA. Our results with PELP1 knockdown models indicate<br />

that PELP1 promotes stability of p53 and functions as a co-regulator<br />

of p53. PELP1 has the potential to modulate CBP/p300 mediated<br />

acetylation of the Lysine 382 residue of p53. Immunoprecipitation<br />

and chromatin immunoprecipitation (ChIP) assays were performed to<br />

examine PELP1 interaction with p53 and its recruitment to p53 target<br />

genes respectively. ChIP assays revealed that PELP1 knockdown<br />

significantly reduces the recruitment of p53 to the target genes. The<br />

p53 regulatory role of PELP1 is mediated by the phosphorylation of<br />

PELP1 at the conserved SQ motif by ATM. We generated model cells<br />

that overexpress WT or mutant (S1033A) PELP1 and demonstrated<br />

the significance of ATM mediated phosphorylation in PELP1<br />

mediated p53 co-activation functions. Based on the sequence of the<br />

site of phosphorylation, we have developed a novel cell permeable<br />

peptide (TAT-1033PELP1 inhibitor) as well as a phospho-PELP1<br />

antibody that uniquely recognizes S1033 phosphorylated PELP1. We<br />

confirmed ATM mediated phosphorylation of PELP1 in vivo using<br />

phospho PELP1 antibody (1033p-PELP1). The TAT 1033 inhibitor<br />

peptide significantly reduced ATM mediated phosphorylation of<br />

endogenous PELP1. Treatment of ER- positive breast cancer cells<br />

with this peptide resulted in resistance to genotoxic stress compared<br />

to cells that are treated with control TAT peptide. However, in TNBC<br />

cells that has a mutant p53, PELP1 knockdown or treatment with<br />

TAT-PELP1 inhibitor resulted in significant loss of cell viability and<br />

increase in apoptosis in response to genotoxic stress. IHC analysis of<br />

tumor tissue array (n=100) revealed increased PELP1 phosphorylation<br />

in advanced ER-negative tumors and its status correlated with ATM.<br />

Collectively, our results suggest that hyperactive ATM-PELP1-p53<br />

pathway contributes to TNBC progression and that the TAT-<br />

1033PELP1 inhibitor represents a novel therapeutic to block PELP1<br />

oncogenic functions in TNBC.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

142s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Discussion 9<br />

PD09-08<br />

The potential role of TLK2 as a therapeutic target in breast<br />

cancer.<br />

Kim J-A, Cao X, Tan Y, Schiff R, Wang X. Lester & Sue Smith <strong>Breast</strong><br />

Center, Baylor College of Medicine, Houston, TX<br />

Invasive breast cancer (IBC) is a heterogeneous disease with distinct<br />

molecular and histological features as well as diverse biological<br />

behaviors. It is thus critical to find specific gene aberrations that can<br />

drive breast cancer initiation and progression as potential therapeutic<br />

targets. Our lab has established bioinformatics workflows to identify<br />

therapeutic oncogene targets. We have nominated Tousled-Like<br />

Kinase 2 (TLK2) as a candidate therapeutic target from 44,932<br />

human genes based on integrative copy number data from The<br />

<strong>Cancer</strong> Genome Atlas (TCGA). TLK2 is a nuclear serine/threonine<br />

kinase that serves as a cell cycle checkpoint regulating chromosome<br />

assembly, and is inactivated by phosphorylation through the ATM<br />

dependent manner in the response to the double strand breaks during<br />

the S-phase. Interestingly, we observed 12-14% of focal TLK2 gene<br />

amplifications and translocations in invasive breast cancers, which<br />

are more frequent in luminal B tumors. Chromosomal aberrations<br />

may leads the deregulation of TLK2 kinase activity and results in<br />

overcoming the cell cycle checkpoint upon DNA damage such as IR or<br />

chemotherapeutic drugs treatments in breast cancer, thus destabilizing<br />

the cancer genome. Indeed, our preliminary data show that TLK2<br />

knockdown substantially inhibits cell proliferation in breast cancer<br />

cells overexpressing TLK2. Further, this response is sustained in<br />

their derivative strains resistant to hormone or anti-Her2 therapies,<br />

suggesting the potential of TLK2 inhibition to sensitize these therapies<br />

as well. Overall, I expect that this study will provide compelling<br />

evidence regarding the role of TLK2 in breast cancer tumorigenesis<br />

and genomic instability, and establish its potential as a therapeutic<br />

target. The possibility of developing specific TLK2 inhibitors can<br />

be well demonstrated by the previous success of drugs against other<br />

cell cycle checkpoint kinases that are currently under clinical trials.<br />

In addition, molecular assays detecting TLK2 derangements may<br />

be applied in the clinics to identify patients who might benefit from<br />

future TLK2 inhibitor. If TLK2 endows resistance or sensitivity to<br />

other therapies, assays detecting TLK2 aberrations may also have<br />

important predictive value for personalization of these treatments.<br />

PD09-09<br />

The Akt inhibitor MK-2206 is an effective radio-sensitizer of p53<br />

deficient triple negative breast cancer (TNBC) cells.<br />

Connolly EP, Sun Y, Chao KC, Hei TK. Columbia University, New<br />

York, NY<br />

Purpose: Triple negative breast cancer (TNBC) is an aggressive<br />

tumor with a higher locoregional recurrence compared to estrogen<br />

receptor (ER) positive breast cancer. Mutations in the PI3K/Akt/<br />

mTOR pathway leading to up-regulation of Akt occur with high<br />

frequency in TNBC. Hyperactive Akt is associated with increase<br />

radiation resistance, mediated through inhibition of apoptosis and<br />

up-regulation of DNA damage-induced G2 arrest. p53 plays a key<br />

role in mediating G1 cell cycle arrest following genotoxic stress<br />

such as radiation; p53-deficient cells however must rely on alternate<br />

mechanisms for cell cycle arrest in S- and G2-phases. Inhibition of<br />

Akt therefore would be expected to act in a synthetic lethal fashion<br />

to radiosensitize p53 deficient TNBC cells by decreasing their ability<br />

to arrest in G2. Given that p53 mutations occur in 44% of TNBC this<br />

may be an effective strategy for radiosensitizing TNBC.<br />

Methods: Experiments were conducted using MDA-MB-468,<br />

MDA-MB-231 and SUM 149 cells, all well established models for<br />

p53-deficient TNBC. Cells were treated with MK-2206, a potent<br />

allosteric Akt inhibitor, alone or in combination with increasing doses<br />

of radiation from 0-8 Gy. In vitro studies preformed included; cell<br />

survival assays, MTT assay, immunoblot analysis of key proteins,<br />

FACS analysis for cell cycle, apoptosis, and gH2AX staining.<br />

Results: We found that the combination of IR and Akt inhibition by<br />

MK-2206 is synergistic. MK-2206 inhibited both endogenous and<br />

radiation-induced Akt activation and phosphorylation of its downstream<br />

targets. Decreased clonogenic survival was seen after 2 or 24<br />

hours pretreatment with MK-2206 as well as decreased viability by<br />

MTT assay. Cell cycle analysis demonstrates MK-2206 decreases<br />

the proportion of cells in G2/M arrest following irradiation, as well<br />

as induced a greater proportion of apoptosis. Evaluation of the DNA<br />

damage response pathway demonstrated decreased levels of total<br />

DNA-PK, as well as a delayed DNA damage response.<br />

Conclusions: These studies demonstrate that targeted inhibition<br />

of Akt effectively radiosensitizes p53 deficient TNBC cells lines,<br />

possibly through a synthetic lethal effect on the DNA damage<br />

response.<br />

PD09-10<br />

DNA Damage and Repair in Mammary Gland Development<br />

Kass EM, Helgadottir H, Moynahan ME, Jasin M. Memorial Sloan-<br />

Kettering <strong>Cancer</strong> Center, New York, NY<br />

Numerous factors are linked to breast cancer risk including<br />

reproductive history, early exposure to radiation, age and genetic<br />

background. Events that occur during specific stages of mammary<br />

development are believed to alter the lifetime risk of breast cancer.<br />

Epidemiological studies over the past 3 decades have suggested an<br />

association between parity and decreased lifetime breast cancer risk.<br />

Studies have also shown that breast cancer risk is significantly higher<br />

in women who received radiotherapy to the chest as adolescents<br />

compared to those who received chest radiation as adults. These<br />

associations implicate development specific changes in mammary<br />

epithelium that affect the molecular mechanisms whereby cells<br />

respond to DNA damage and, importantly, are at subsequent cancer<br />

risk.<br />

Because many of the genes linked to hereditary breast cancer<br />

are associated with DNA double-strand break (DSB) repair, we<br />

hypothesize that there are differences in overall DSB repair,<br />

homologous recombination (HR), and cell fate following damage<br />

in mammary epithelium at different stages of mammary gland<br />

development—puberty, pregnancy, lactation and post-involution. To<br />

assess these differences we are using mice containing the DR-GFP<br />

reporter targeted to chromosome 17 to study HR at chromosome<br />

breaks induced by the transient expression of I-SceI in mammary<br />

epithelial cells. DR-GFP is a widely used repair reporter in which<br />

direct repeats of two defective GFP genes are induced to recombine<br />

by I-SceI endonuclease cleavage of one of the repeats. We have<br />

observed that the efficiency of HR in primary mammary epithelial<br />

cells derived from 8-week old virgin mice is similar to that observed<br />

in other primary cells of somatic origin (approximately 0.65% of cells<br />

heterozygous for the DR-GFP reporter). Interestingly, the amount<br />

of HR is increased in mammary epithelium derived from pregnant<br />

mice compared to age-matched virgin controls (0.98 versus 0.65%).<br />

This 1.5 fold increase in HR suggests that in pregnancy, mammary<br />

epithelium is more proficient in the accurate repair of DNA DSBs.<br />

We are also assessing DNA damage signaling in different periods<br />

of mammary gland development. Preliminary analysis of histone<br />

H2AX phosphorylation following whole-body irradiation suggests<br />

more rapid kinetics of DNA damage signaling response in mammary<br />

www.aacrjournals.org 143s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

tissue derived from pregnant as compared to age-matched virgin mice.<br />

We have recently generated DR-GFP containing mice that express<br />

I-SceI from a randomly integrated I-SceI transgene under the control<br />

of a tetracycline-inducible promoter. We are currently using these mice<br />

to assess HR in an entirely in vivo system and are in the process of<br />

breeding this reporter line with mice carrying mutations in Brca1 or<br />

Brca2. This mouse model will be used to assess HR in vivo in the<br />

mammary tissue of these mice during different stages of mammary<br />

gland development.<br />

This project will provide valuable insight into how developmental<br />

changes unique to mammary gland epithelium affect cellular<br />

responses to DNA damage. We hope to consider therapeutic<br />

manipulation of identified stage-specific differences to decrease<br />

breast cancer susceptibility.<br />

PD10-01<br />

Withdrawn by Author<br />

PD10-02<br />

Metabolic syndrome and recurrence within the 21-gene<br />

recurrence score assay risk categories in lymph node negative<br />

breast cancer<br />

Lakhani A, Guo R, Duan X, Ersahin C, Gaynor ER, Godellas C, Kay<br />

C, Lo SS, Mai H, Perez C, Albain K, Robinson P. Loyola University<br />

Medical Center, Maywood, IL<br />

Background: The incidence of the metabolic syndrome (MS) has<br />

been increasing in the United States and elsewhere. The interaction of<br />

MS with breast cancer (BC) incidence, tumor biology and outcomes<br />

are under study. We hypothesized that the presence of MS would<br />

predict BC recurrence to a variable degree across the diverse BC<br />

biology as defined by the risk categories of the 21-gene recurrence<br />

score (RS) assay.<br />

Patients and Methods: We studied consecutive patients (pts) with<br />

newly diagnosed, estrogen receptor (ER) positive, lymph node (LN)<br />

negative BC treated in our institution between 2006-2011 who had<br />

a 21-gene RS assay done on their tumors. All pts were treated with<br />

standard systemic and local therapy. The electronic medical record<br />

was queried for key diagnoses including MS and its constituent parts.<br />

The WHO definition was used to categorize pts as having MS defined<br />

as diabetes mellitus (DM) or glucose intolerance, plus at least 2 of the<br />

following: hypertension (HTN), dyslipidemia (HL), central obesity<br />

and microalbuminemia. Tumor characteristics including Ki67 index,<br />

grade, tumor size, HER2/neu status; and pt characteristics including<br />

age, race, menopausal status, body mass index were recorded. The<br />

association of MS and the tumor and patient characteristics with the<br />

RS tertiles of low, intermediate and high risk was analyzed.<br />

Results: We identified 332 pts, median age 62 years, of whom 88<br />

(27%) had MS. There was no significant association between the<br />

MS and any of the patient or tumor variables including the 21-gene<br />

RS assay, except for race (p=0.004). Eleven of 21 (52%) African-<br />

American women had MS, 68 of 284 (24%) Caucasian women had<br />

MS, and 9 of 21 (43%) others including Hispanic and Asian women<br />

had MS. However, there was a significant association between<br />

recurrence and MS (p=0.0002) independent of other factors. Of the<br />

21 pts who recurred, 13 (61.9%) had MS. There was an association<br />

of recurrence and MS within RS tertiles. For pts with low risk scores,<br />

7/44 (15.9%) with MS vs. 1/126 (0.79%) without MS had recurrence<br />

(p=0.0003). For pts with intermediate risk scores, 5/30 (16.67%)<br />

with MS vs. 4/83 (4.82%) without MS had recurrence (p=0.05). For<br />

patients with high risk scores, 1/9 (11.11%) with MS vs. 2/15 (13.33%)<br />

without MS had recurrence (p=1).<br />

Conclusion: MS is an independent risk factor for BC recurrence<br />

among women with LN negative, ER positive BC treated with<br />

standard adjuvant therapy. There is a striking impact of MS on<br />

recurrence in pts with tumor biologies defined by low (and to a lesser<br />

degree) intermediate risk 21-gene RS assay scores. However, there<br />

is no difference in recurrence risk by MS among those pts with high<br />

RS. This implies that interventions directed at modifying MS in newly<br />

diagnosed pts with early BC may potentially favorably impact survival<br />

in those with specific tumor biologies as defined by multigene assays.<br />

Thus, long-term prospective studies should be conducted to further<br />

evaluate both the short and long term effects of MS on BC outcomes.<br />

PD10-03<br />

Predictive value of a proliferation score (MS) in postmenopausal<br />

women with endocrine-responsive breast cancer: results from<br />

International <strong>Breast</strong> <strong>Cancer</strong> Study Group (IBCSG) Trial IX<br />

Sninsky J, Wang A, Gray K, Lagier R, Christopherson C, Rowland C,<br />

Chang M, Kammler R, Viale G, Kwok S, Regan M, Leyland-Jones B.<br />

Celera, Alameda, CA; Dana-Farber <strong>Cancer</strong> Institute, Boston, MA;<br />

IBCSG Coordinating Center, Berne, Switzerland; European Institute<br />

of Oncology, Milan, Italy; <strong>San</strong>ford Research, Sioux Falls, SD<br />

Background<br />

While representing the largest fraction of women diagnosed with<br />

primary breast cancer, older postmenopausal women with ER+,<br />

HER2- tumors are less responsive to chemoendocrine therapy than<br />

younger women and have been underrepresented in molecular<br />

profiling of randomized trials. IBCSG Trial IX, a randomized<br />

controlled trial in postmenopausal women, median age 61y, with<br />

node negative disease, failed to demonstrate the benefit of preceding<br />

tamoxifen (T) by 3 cycles of CMF for ER+ tumors. We sought to<br />

determine if MS, a proliferation score, could identify a subset of<br />

women who differentially benefit from addition of chemotherapy<br />

to T in this trial.<br />

Methods<br />

From 1988-1999, 1669 eligible patients (1040 with ER+, HER2-<br />

tumors) were randomized to CMF→T vs T. Disease-free survival<br />

(DFS) was the primary trial endpoint; breast cancer-free interval<br />

(BCFI) which excludes second (non-breast) malignancies and censors<br />

deaths without prior cancer event was also evaluated. Analysis was<br />

limited to the first 7 years of follow-up. From 671 (ER+, HER2-)<br />

available subjects, 568 were successfully profiled by RT-PCR. The<br />

mRNA expression levels of 14 equally-weighted proliferation genes<br />

and 3 normalization genes were used to generate MS; predetermined<br />

binary categorization of MS was used. Analysis of this post hoc,<br />

pre-specified study used results from centralized laboratory IHC and<br />

Cox models to assess the predictive value of MS on DFS and BCFI,<br />

adjusting for traditional risk factors of local treatment, age, ER, PR,<br />

Ki67, tumor size and grade.<br />

Results<br />

Subgroups of MS (low, 169 samples (30%) and high, 399 samples<br />

(70%)) were identified. MS by treatment interaction was significant<br />

for DFS and BCFI (each p≤0.004). Among patients with low MS,<br />

CMF→T improved DFS (HR 0.19, 95% CI 0.06-0.59) and BCFI<br />

(HR 0.19, 95% CI 0.05-0.72) vs T; 7y DFS was 95% vs 83% with<br />

CMF→T vs T. Among patients with high MS, CMF→T did not<br />

improve DFS (HR 1.27, 95% CI 0.79-2.05) or BCFI (HR 1.37, 95%<br />

CI 0.80-2.33) and 7y DFS of 81% for CMF→T and T. Continuous MS<br />

was moderately correlated with log Ki67 (r=0.47) but not correlated<br />

with ER or PR. The MS by treatment interaction remained significant<br />

with Ki67 in the model.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

144s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Discussion 10<br />

Conclusions<br />

Low MS was associated with differential benefit favoring those women<br />

receiving CMF→T vs T alone for both DFS and BCFI in the first 7<br />

years. The effect was independent of traditional risk factors including<br />

Ki67. Hence this study, which is unconfounded by chemotherapyinduced<br />

ovarian ablation in younger women, identifies a subset of<br />

postmenopausal women with ER+, HER2- tumors that benefit from<br />

CMF chemotherapy. This seemingly incongruous observation is<br />

consistent with a) the prior observation that only the low-proliferation<br />

subgroup by PAM50 11-gene signature benefits from the addition of<br />

weekly paclitaxel to adjuvant FEC (GEICAM/9906), b) the ability<br />

of MS to identify a subset of women with tumors with disseminated<br />

luminal progenitor cells activated through the agonistic activity of<br />

tamoxifen, and c) the repetitive dosing of cyclophosphamide and taxol<br />

being hypothesized to act via tumor stroma/anti-angiogenesis. The<br />

relative contribution of these factors is under investigation.<br />

PD10-04<br />

Predictive genomic markers to chemotherapy and adjuvant<br />

trastuzumab via whole genome expression DASL profiling in the<br />

N9831 adjuvant study<br />

Perez EA, Eckel-Passow JE, Ballman KV, Anderson SK, Thompson<br />

EA, Asmann YW, Jen J, Dueck AC, Lingle WL, Sledge GW, Winer EP,<br />

Gralow J, Jenkins RB, Reinholz MM. Mayo Clinic, Jacksonville, FL;<br />

Mayo Clinic, Rochester, MN; Mayo Clinic, Scottsdale, AZ; Indiana<br />

University Simon <strong>Cancer</strong> Center, Indianapolis, IN; Dana Farber<br />

<strong>Cancer</strong> Institute, Boston, MA; Seattle <strong>Cancer</strong> Care Alliance, Seattle,<br />

WA; Ventana Medical Systems, Inc., Tuscon, AZ<br />

Background: Adding adjuvant trastuzumab to chemotherapy for<br />

patients (pts) with resected HER2+ breast cancer (BC) improves<br />

disease free and overall survival (Perez EA, et al. J Clin Oncol<br />

2011). Understanding which pts are most likely to benefit from these<br />

therapies is a goal of our team.<br />

Methods: Baseline 1639 annotated tumor specimens from pts enrolled<br />

in the phase III N9831 adjuvant trastuzumab trial (NCT00005970)<br />

were processed via the >29,000 gene probe DASL technology<br />

(Illumina) to identify genomic predictors of pt outcome to Arm A<br />

chemotherapy or Arm C chemotherapy plus concurrent trastuzumab.<br />

Samples were spotted in 96 well plates, with appropriate replicates.<br />

Extensive quality control and internal validation were performed.<br />

The association of each gene with recurrence-free survival (RFS) was<br />

determined for each treatment arm separately using CoxPH models<br />

adjusted for clinical and tumor risk factors. A p3x ULN) and bilirubin (>2x ULN) elevation, which represent<br />

possible Hy’s Law cases and a high risk of acute liver failure,<br />

among 1194 patients randomized to lapatinib treatment from whom<br />

pharmacogenetic data was available.<br />

This study prospectively validated prior reports of the association of<br />

the specified MHC variants with elevated ALT among women treated<br />

with lapatinib. The strongest effects were observed for carriers of<br />

the HLA alleles DQA1*02:01 and DRB1*07:01, with odds ratios of<br />

20 (95% CI: 8-40) between cases (n=34) and controls (n=807-808).<br />

These two HLA alleles are highly correlated, inherited together in<br />

most individuals and are consistent with a single genetic association.<br />

The overall risk of patients having an ALT (>3xULN) elevation<br />

was 3.0% and 0.7% during treatment with lapatinib and placebo<br />

respectively. Carriers of either HLA allele had a 12% chance (positive<br />

predictive value) of having an elevated ALT (>3xULN), in contrast<br />

to a 0.9% risk (negative predictive value, 99.1%) for non-carriers of<br />

the specified HLA alleles. These associations were maintained for<br />

higher ALT elevation thresholds and for cases of concurrent ALT<br />

and TBL elevation, consistent with possible Hy’s Law cases. These<br />

results strongly support the role of Class II HLA-modulated immune<br />

mechanisms in lapatinib-induced hepatotoxicity.<br />

Our results validate the large strength of association of the HLA alleles<br />

DRB1*07:01 and DQA1*02:01 with hepatotoxicity and provide the<br />

possibility of managing the risk of hepatotoxicity in women receiving<br />

lapatinib for early or late stage HER2 positive breast cancer. This<br />

association with immune mechanisms may have implications for<br />

toxicities with other TKIs in current use in cancer patients.<br />

www.aacrjournals.org 145s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

PD10-06<br />

CXCL13 mRNA predicts Docetaxel benefit in Triple Negative<br />

tumors<br />

Wirtz RM, Leinonen M, Bono P, Isola J, Kellokumpu-Lehtinen P-L,<br />

Kataja V, Turpeenniemi-Hujanen T, Jyrkkiö S, Eidt S, Schmidt M,<br />

Joensuu H. STRATIFYER Molecular Pathology GmbH, Cologne,<br />

Germany; Pharma, Turku, Finland; Helsinki University Central<br />

Hospital and University of Helsinki, Helsinki, Finland; University<br />

of Tampere and Tampere University Hospital, Finland; Tampere<br />

University Hospital, Tampere, Finland; Kuopio University Hospital,<br />

Kuopio, Finland; Oulu University Hospital, Oulu, Finland; Turku<br />

University Hospital, Turku, Finland; Institute of Pathology at the<br />

St-Elisabeth-Hospital, Cologne, Germany; University Hospital<br />

Mainz, Germany<br />

Background: Presence of immune cell infiltrates in tumor may<br />

influence outcome of patients with node negative breast cancer (BC)<br />

(Schmidt et al 2008). High expression of CXCL13 (B lymphocyte<br />

chemoattractant chemokine) was strongly associated with favorable<br />

survival in one recent series consisting of BC patients treated with<br />

adjuvant chemotherapy (Razis et al. 2012). Here we examined the<br />

clinical significance of breast tumor CXCL13 mRNA levels within<br />

the context of the FinHer trial, where patients were randomly assigned<br />

to receive 3 cycles of either docetaxel or vinorelbine followed by<br />

3 cycles of FEC. Patients with HER2-positive BC had a second<br />

randomization for trastuzumab vs. control. Since intratumoral B cells<br />

are of particular importance for obtaining response to chemotherapy<br />

in triple negative BC (TNBC) and of limited value in luminal BC<br />

(Schmidt et al. 2012), and trastuzumab treatment is a confounding<br />

factor in HER2+ BC, we focused on TNBC.<br />

Methods: RNA was extracted from FFPE tumor tissue of 917 (90.8%)<br />

out of the 1010 patients who participated in the FinHer trial. CXCL13<br />

mRNA expression was measured using RT-qPCR. The molecular<br />

subtypes (luminal, HER2-enriched and triple-negative) were<br />

determined using IHC and central chromogenic in situ hybridization<br />

(CISH) testing. Prognostic significance of factors was assessed using<br />

univariate and multivariate analyses. Distant disease-free survival<br />

(DDFS) between groups was compared using the log-rank test.<br />

Results: Tumor CXCL13 mRNA expression showed a bimodal<br />

distribution and correlated negatively with tumor ESR1 mRNA<br />

levels (r=-0,31; p


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 1<br />

PD10-08<br />

Venlafaxine inhibits the CYP2D6 mediated metabolic activation<br />

of tamoxifen: Results of a prospective multicenter study:<br />

(NCT00667121)<br />

Goetz MP, Suman V, Henry NL, Reid J, Safgren S, Kosel M, Kuffel<br />

M, Sideras K, Flockhart D, Stearns V, Denduluri N, Irvin WJ, Ames<br />

M. Mayo Clinic, Rochester, MN; University of Michigan, Ann Arbor,<br />

MI; Indiana University, Indianapolis, IN; Johns Hopkins, Baltimore,<br />

MD; Fairfax-Northern Virginia Hematology-Oncology, Arlington,<br />

VA; University of North Caroloina, Chapel Hill, NC<br />

Background: CYP2D6 is the rate limiting enzyme responsible for the<br />

metabolic activation of tamoxifen (tam) to endoxifen. Compared to<br />

CYP2D6 poor metabolizers (PM), tam-treated CYP2D6 extensive<br />

metabolizers (EM) have higher endoxifen concentrations, more<br />

vasomotor symptoms (Goetz, MP J Clin Oncol 2005), and are more<br />

likely to discontinue tam (Rae, JM 2009. Pharmacogenomics J).<br />

Additionally, higher endoxifen concentrations are associated with a<br />

stepwise increase in tam side-effects (Lorizio, W <strong>Breast</strong> <strong>Cancer</strong> Res<br />

Treat 2012). The data regarding CYP2D6 genotype and recurrence is<br />

mixed. Venlafaxine is a weak CYP2D6 inhibitor not known to alter<br />

tam pharmacokinetics (PK) and commonly recommended for tam<br />

induced hot flashes. We conducted a multicenter pharmacological<br />

study to determine whether venlafaxine altered the PK of tam and to<br />

determine the distribution of CYP2D6 genotypes in this population<br />

Methods: Women taking tam for at least 4 weeks and for whom<br />

venlafaxine was recommended for the treatment of hot flashes<br />

were eligible. Blood samples were collected prior to and 8-16<br />

weeks following initiation of venlafaxine for steady state tam and<br />

metabolites. Genotyping was performed for alleles associated with<br />

no (PM; *3, *4, *5,*6); reduced (intermediate, IM; *10, 17 and<br />

*41); and ultra-rapid (UM; *1X2) metabolism. Power calculations<br />

demonstrated that 17 patients with paired samples were required<br />

(two-sided alpha=0.05 t-test, 90% power) to detect a 25% change<br />

in endoxifen levels after at least 8 weeks of concurrent treatment.<br />

Results: 30 women (median age 48.5) initiated venlafaxine. CYP2D6<br />

genotypes were within Hardy Weinberg Equilibrium (HWE). CYP2D6<br />

UM allele frequency (6.7%) was higher while CYP2D6 *4 (13.3%)<br />

was lower than expected compared to an unselected population (0.5<br />

and 21% respectively; Sachse Am. J. Hum. Genet. 1997), resulting in<br />

the absence of CYP2D6 PM/PM. Mean (min/max) baseline endoxifen<br />

concentrations (8.73; 1.5-20.5 ng/ml) were correlated with CYP2D6<br />

phenotype as follows: intermediate (EM/PM, PM/IM): 6.8 (1.5-11.2);<br />

extensive (EM/EM, EM/IM): 9.4 (1.5-20.5) and ultra-rapid (UM/<br />

EM: 11.0; 7.8-14) (r 2 = 0.35 p=0.05). In patients with paired samples<br />

(n=20), venlafaxine resulted in a 23% decrease in endoxifen (-2.06<br />

ng/ml; 95% CI -0.69 to -3.04; p=0.004), but not tam, NDMT, or<br />

4HT concentrations. Following initiation of venlafaxine, CYP2D6<br />

genotype was no longer associated with endoxifen concentrations<br />

(r 2 = 0.28 p=0.23). For women with reduced CYP2D6 metabolism<br />

[EM/PM (n=9) or PM/IM (n=1)], venlafaxine lowered endoxifen<br />

concentrations (-2.98 ng/ml) to a level (5.41 ng/ml) reported to be<br />

associated with a higher risk of recurrence in adjuvant tam treated<br />

patients (Madlensky, L Clin Pharmacol Ther 2011).<br />

Conclusions: In this study, women with tam-induced vasomotor<br />

symptoms requiring venlafaxine were comprised predominantly<br />

of CYP2D6 EM and UM metabolizers. Venlafaxine significantly<br />

decreased endoxifen concentrations. Although the optimal<br />

concentration of endoxifen is unknown, given prior data linking low<br />

endoxifen concentrations with recurrence, venlafaxine should be used<br />

with caution in tam treated patients. (Supported by R01CA133049)<br />

PD10-09<br />

CYP2D6 and adjuvant tamoxifen: Impact on outcome in pre- but<br />

not postmenopausal breast cancer patients<br />

Margolin S, Lindh J, Thorén L, Xie H, Koukel L, Dahl M-L, Eliasson<br />

E. Karolinska Institutet, Stockholm, Sweden; Karolinska University<br />

Hospital, Stockholm, Sweden; Karolinska Institutet, Karolinska<br />

University Hospital Huddinge, Stockholm, Sweden<br />

Background<br />

The therapeutic effect of tamoxifen in adjuvant treatment of hormone<br />

receptor positive breast cancer might be impaired in patients with<br />

cytochrome P450 2D6 (CYP2D6) genotypes that predispose to<br />

decreased formation of potent anti-estrogenic tamoxifen metabolites.<br />

However, previous studies have shown inconsistent findings in this<br />

regard. In light of two recent studies failing to show an impact of<br />

CYP2D6 genotype on outcome in postmenopausal patients, we<br />

hypothesized that deficient CYP2D6 might be of greater importance<br />

in premenopausal patients with high levels of circulating estrogen.<br />

We therefore aimed to study the effect of CYP2D6 activity in both<br />

pre- and postmenopausal patients who were adherent to tamoxifen<br />

treatment for at least one year.<br />

Methods<br />

382 patients, from a population-based cohort of unselected breast<br />

cancer patients that were prescribed adjuvant tamoxifen for five<br />

years, constituted the base of the study. Information on menopausal<br />

status, tumor characteristics, treatment data including compliance<br />

and outcome was retrieved from medical records. Comprehensive<br />

CYP2D6 genotyping was performed and functionally translated into<br />

constitutive, metabolic activity.<br />

Results<br />

In patients adherent to tamoxifen for at least one year (n=313) there<br />

was an association of reduced CYP2D6 activity (≤50% of normal)<br />

to both recurrence (p=0.02) and breast cancer-specific mortality<br />

(p=0.03). In a multivariable analysis including CYP2D6 activity,<br />

age at diagnosis, tumor size, lymph node status, grade, adjuvant<br />

chemotherapy and concomitant use of CYP2D6 inhibitors, CYP2D6<br />

remained an independent predictor of recurrence (HR=0.39, 95% CI:<br />

0.18-0.85, p=0.02) and breast cancer specific survival (HR=0.33,<br />

95% CI 0.12-0.90, p=0.03). Gradually decreasing CYP2D6 activity<br />

paralleled increasing risk of recurrence and breast cancer related<br />

mortality. The effect of CYP2D6 derived mainly from premenopausal<br />

patients with an association to both recurrence (p=0.01) and breast<br />

cancer specific survival (p=0.04). There was no such association in<br />

the postmenopausal group.<br />

Conclusion<br />

In a prospectively collected cohort of tamoxifen-treated breast cancer<br />

patients, an association between CYP2D6 genotype and outcome was<br />

evident in patients that were adherent to tamoxifen treatment for at<br />

least a year. Importantly, this effect derived from premenopausal<br />

patients only.<br />

P1-01-01<br />

Fluorescence mapping with indocyanine green for sentinel lymph<br />

node detection in early breast cancer – results of the ICG-10 study<br />

Benson JR, Loh S-W, Jones LA, Wishart GC. Addenbrooke’s Hospital,<br />

Cambridge, Cambridgeshire, United Kingdom<br />

Background: Dual localization methods with blue dye and<br />

radioisotope are commonly employed for SLN identification but<br />

potential drawbacks include allergic reactions, radiation exposure<br />

and mandatory licencing. Identification rates exceeding 95% have<br />

been reported using the fluorescent tracer indocyanine green (ICG)<br />

as a navigation system. A feasibility study was undertaken to confirm<br />

www.aacrjournals.org 147s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

the sensitivity and safety of ICG as a tracer agent for sentinel lymph<br />

node (SLN) identification in early breast cancer.<br />

Methods: In an open, non-randomised study, 100 clinically node<br />

negative patients (95 unilateral; 5 bilateral) scheduled to undergo<br />

routine SLN biopsy for core-biopsy proven invasive breast cancer<br />

were identified at the multidisciplinary team meeting [53 screendetected;<br />

51 symptomatic]. All patients received triple localization<br />

with blue dye (2.5% Patent Blue), radiocolloid and ICG (0.5%). The<br />

number of sentinel nodes for each patient were recorded numerically<br />

and whether blue, radioactive or fluorescent. Subcutaneous lymphatics<br />

were visualized with a Photodynamic Eye camera and sensitivity of<br />

individual tracers alone and in combination calculated. Approval was<br />

granted by the Local Research Ethics Committee and the Medicines<br />

and Healthcare Products Regulatory Agency for use of ICG as a<br />

third tracer agent.<br />

Results: Final analysis was performed on 104 procedures in 99<br />

patients (1 exclusion - benign adenomyoepithelioma on definitive<br />

pathology). A total of 242 nodes were removed of which 201<br />

were defined as sentinel (blue and/or radioactive) with an average<br />

nodal retrieval of 2.33 per procedure. Amongst these, 25 contained<br />

either macrometastases (n=16) or micrometastases (n=9), yielding<br />

procedural and node specific positivity rates of 17.3% (18/104)<br />

and 12.4% (25/201) respectively. 100% of these ‘true’ SLNs were<br />

fluorescent (201/201), 95.0% (191/201) were both fluorescent and<br />

blue, 77.6% (156/201) were fluorescent and radioactive whilst 73.1%<br />

(147/201) were blue, radioactive and fluorescent. The procedural<br />

detection rates were 99% (103/104) for blue dye, 91.3% (95/104)<br />

for radioisotope and 100% for fluorescence. All 25 positive nodes<br />

were identified with all three tracers and all 18 node positive patients<br />

had tumour deposits in the first SLN excised. The majority of ‘nonsentinel’<br />

nodes were fluorescent (36/41) and no serious adverse<br />

reactions occurred.<br />

Conclusion: ICG fluorescence imaging permits real-time visualization<br />

of lymphatics and provides an additional dimension to SLN biopsy<br />

which is safe and effective. These results confirm high sensitivity for<br />

fluorescence in SLN identification and the combined nodal sensitivity<br />

for ICG and blue dye (95.0%) was higher than for blue dye and<br />

radioisotope (73.1%). With further refinements in the technique may<br />

it may be possible to eventually rely on ICG as a sole tracer agent.<br />

Moreover, a marginally higher nodal yield with fluorescent mapping<br />

may allow more confident omission of completion axillary dissection<br />

in selected SLN positive patients.<br />

P1-01-02<br />

Withdrawn by Author<br />

P1-01-03<br />

Comparison of sentinel lymph node biopsy guided by the multimodal<br />

method of indocyanine green fluorescence, radioisotope<br />

and blue dye versus the radioisotope in breast cancer; A<br />

randomized phase II trial<br />

Jung S-Y, Kim S-K, Kim SW, Lee S, Lee J, Shin I-s, Kwon Y, Lee E.<br />

National <strong>Cancer</strong> Center, Goyang, Korea<br />

Background: Sentinel lymph node (SLN) biopsy is a standard<br />

surgery for axillary staging of early breast cancer. This study aimed<br />

to evaluate the identification rate and surgery time of SLN biopsy<br />

with the multi-modal method using the mixture of indocyanine green<br />

(ICG) fluorescence, RI and blue dye as a new method, in comparison<br />

with the RI, in clinically node negative breast cancer patients.<br />

Materials and methods: In this phase-II randomized study, 86<br />

patients with clinically node negative breast cancer were randomized<br />

to receive periareolar injection of the mixture including ICG<br />

fluorescence, RI and blue dye or the only RI for SLN biopsy. We<br />

compared the identification rate, the number of SLN, and detections<br />

time of SLN biopsy between these two groups.<br />

Results: The mean age of the multimodal group and the only RI<br />

group was 48.2 years and 51.0 years (p=0.12), respectively. There<br />

were no differences of histopathologic factors including tumor size,<br />

node positivity, hormone receptor positivity and HER2 positivity<br />

between two groups. Sentinel lymph nodes were identified in all<br />

patients of both groups (100% in the multimodal group vs 100% in<br />

the RI group). The average number of SLN in the mixture group was<br />

more than that of the RI group (3.4 ±1.37 vs 2.3 ± 1.04; p


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 1<br />

Results: After inj. of Tilmanocept, 100% (22/22 immediate; 88/88<br />

overall) of pts demonstrated localization. After inj. of SCI, 90.1%<br />

(127/141) and 91.5% (129/141) of pts exhibited localization<br />

immediately and overall, respectively.<br />

Localization rate and degree of localization with the benchmark for<br />

SCI, H 0 , are provided in Table 1 (significance is considered


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

vs negative) or grade (grade 1 vs 2 vs 3), these biologic factors were<br />

able to significantly discriminate patients with respect to RFS, DSS<br />

and OS (table 2).<br />

Table 2. Survival outcomes for stage I patients by ER status and grade<br />

ER pos ER neg p-value Grade I Grade II Grade III p-value<br />

RFS - 5y 96.7% 89.6%


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 1<br />

P1-01-09<br />

Which nomograms may be the best for predicting nonsentinel<br />

lymph node metastasis in breast cancer patients: a meta-analysis.<br />

Chen K, Jin L, Zhu L, Shan Q, Su F. Sun Yat-sen Memorial Hopsital,<br />

Guangzhou, Guangdong, China<br />

Background: Axillary lymph node dissection (ALND) is the standard<br />

treatment for breast cancer patients with positive sentinel lymph<br />

nodes (SLNs). Several nomograms were developed to identify SLNpositive<br />

patients with low risk of nonsentinel lymph nodes (NSLNs)<br />

metastasis. These nomograms were validated in different populations<br />

and it is still unknown which is the best. This study is to present a<br />

systemic review and perform a meta-analysis to obtained the pooled<br />

AUC (Area Under the receiver-operator Curve) value of each models.<br />

Methods: This review focused on six models: Cambridge, MSKCC,<br />

Mayo, MDA, Tenon and Stanford models. A “Pubmed” search and<br />

“Web of science” search were conducted and 35 literatures were<br />

ultimately included. AUC and the number of patients with positive<br />

NSLNs were extracted. Publication bias and heterogeneity were<br />

analyzed. AUCs were converted to odds ratios (ORs) for combination.<br />

The combined ORs were converted back to AUCs to represent the<br />

integrated discriminative capabilities of each models.<br />

Findings: In total, the Cambridge, Mayo, MDA, MSKCC, Stanford<br />

and Tenon models were validated in 8, 6, 4, 39, 14 and 15 studies,<br />

with 2156, 2431, 843, 8143, 3700 and 3648 patients included,<br />

respectively. There were no publication bias or heterogeneity observed<br />

in the Cambridge, Mayo, MDA and Tenon models (Table 1). The<br />

combined ORs and the corresponding AUCs of each models were<br />

listed as follow: Cambridge (OR=3.86, AUC=0.71), Mayo (OR=3.71,<br />

AUC=0.71), MSKCC (OR=3.47, AUC=0.70), MDA(OR=3.44,<br />

AUC=0.70), Tenon (OR=3.46, AUC=0.70) and Stanford (OR=2.92,<br />

AUC=0.67). For each of the predictive models, both fixed and random<br />

effect models were used to calculate the combined OR. The presence<br />

of larger difference between the fixed and random effect analysis<br />

suggests small study effects, rendering the meta-analysis relatively<br />

less reliable. The combined ORs were identical when fixed and<br />

random effect models were used in the Cambridge and MDA models,<br />

suggesting that there was no small study effects in these two models.<br />

Table 1, Publication bias analysis, Heterogeneity analysis and Comined OR and converted AUC of<br />

each models.<br />

Test of<br />

publication Test of heterogeneity Fixed Effect Model† Random Effect Model†<br />

bias<br />

Begg’s Heterogeneity Combined Converted Combined Converted<br />

P<br />

method,P statistic<br />

OR AUC OR AUC<br />

Cambridge 0.051 4.85 0.563 3.86 0.71 3.86 0.71<br />

Mayo 0.624 6.39 0.172 3.71 0.71 4.28 0.73<br />

MSKCC


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P1-01-11<br />

Is OSNA mRNA copy number in sentinel lymph node biopsy<br />

predictive of further disease in the axilla?<br />

Rusby JE, Agabiti E, Waheed S, Barry P, Roche N, Allum W, Gui G,<br />

MacNeill F, Christaki G, Osin P, Nerurkar A. Royal Marsden NHS<br />

Foundation Trust, London, United Kingdom<br />

Introduction<br />

Intra-operative assessment of sentinel nodes (SLNs) allows immediate<br />

completion axillary dissection (cALND) in breast cancer patients.<br />

Molecular assessment such as one-step nucleic acid amplication<br />

(OSNA) promises greater sensitivity and provides a more accurate<br />

quantitative assessment than traditional methods.<br />

Our unit policy is to proceed to cALND in patients with macrometastases<br />

but not for micrometastases. However, evidence of upstaging has led<br />

us to seek to raise the threshold for proceeding to cALND. The CK19<br />

mRNA copy number is an expression of the metastatic burden in the<br />

SLN and may be related to the presence of additional disease in the<br />

cALND. Since the original copy number threshold between micro<br />

(250-5000 copies/microliter) and macrometastasis (>5000 copies/<br />

microliter) was based on few patients and serial pathological sections,<br />

we investigated the mRNA copy number in patients with and without<br />

additional disease in the cALND.<br />

Methods<br />

All patients in our unit undergo pre-operative axillary ultrasound<br />

with fine needle aspiration cytology of any suspicious nodes. Those<br />

with malignant cytology proceed directly to ALND. Radiologically<br />

and cytologically node negative patients undergo sentinel lymph<br />

node biopsy (SLNB) and OSNA. Electronic records of consecutive<br />

patients with invasive breast cancer undergoing SLNB with OSNA<br />

from August 2011 to March 2012 were retrospectively reviewed. Two<br />

parameters of mRNA copy number were examined: Copy number of<br />

the highest copy number SLN and the summed copy numbers of all<br />

positive SLNs. Their relationship to the presence of further disease<br />

in the axilla was examined using Student’s t test.<br />

Results<br />

Of 201 SLNBs, 45 (22%) had macrometastasis-positive OSNA and<br />

therefore underwent cALND (1 patient declined). Twenty patients<br />

(45%) had no further positive nodes (a negative cALND) with a total<br />

axillary metastatic burden of 1-2 in 11-27 nodes. Twenty four (55%)<br />

showed further disease (a positive cALND) with a burden of 2-20 in<br />

9-30 nodes, including the SLNs.<br />

There was no significant difference in tumour size or grade between<br />

patients with additional positive nodes in the cALND compared with<br />

those with no further disease.<br />

There was no significant difference in the copy number of the highest<br />

copy number positive SLN (p=0.44) or in the summed copy number<br />

of all positive SLNs (p=0.36) between the cALND positive and<br />

negative groups.<br />

Conclusion<br />

OSNA CK19 mRNA copy number does not correlate with the cALND<br />

metastatic burden. Therefore, raising the copy number threshold<br />

may be too simplistic as a method to better select patients with high<br />

probability of a positive cALND. A predictive model will be derived<br />

based on multivariate analysis of the larger patient population (>400<br />

patients) that will have undergone SLNB with OSNA by the time<br />

of SABCS.<br />

P1-01-12<br />

The Performance of the One Step Nucleic acid Amplification<br />

(OSNA) Assay in <strong>Breast</strong> <strong>Cancer</strong> Patients with Receiving<br />

Preoperative Systemic Therapy<br />

Yagata H, Yamauchi H, Horii R, Osako T, Iwase T, Akiyama<br />

F, Kinoshita T, Tsuda H, Tsugawa K, Nakamura S. St. Luke’s<br />

International Hospital, Tokyo, Japan; <strong>Cancer</strong> Institute Hospital of the<br />

Japanese Foundation for <strong>Cancer</strong> Research, Tokyo, Japan; National<br />

<strong>Cancer</strong> Center Hospital, Tokyo, Japan; St. Marianna University<br />

School of Medicine, Kanagawa, Japan; Showa University School of<br />

Medicine, Tokyo, Japan<br />

Background: The OSNA (One Step Nucleic acid Amplification) assay<br />

is a semi-automated lymph node examination method using molecular<br />

biological technique. The OSNA assay has been validated for breast<br />

cancer patients without receiving preoperative systemic therapy (PST)<br />

by several clinical studies and has currently become more popular as<br />

sentinel lymph node (SLN) examination method with the following<br />

two main advantages; 1) to allow examination of the whole portion of<br />

a node, 2) to allow intraoperative judgment of metastasis positive or<br />

negative. However, the feasibility of the OSNA assay in breast cancer<br />

patients treated by PST has never been confirmed. In this multi-central<br />

clinical study, we compared the judgments of the OSNA assay and of<br />

pathological examination on lymph nodes dissected after receiving<br />

PST to evaluate the performance of the OSNA assay.<br />

Material & Methods: Three hundred two nodes dissected from the 80<br />

breast cancer patients who received PST were examined. Each lymph<br />

node was divided at 2mm intervals and the slices were alternately<br />

applied to the OSNA assay and pathological examination with H&E<br />

staining and CK19 immunohistochemical staining of permanentsection.<br />

In pathological examination, judgments of metastasis positive<br />

or negative were determined by one central-review pathologist<br />

according to the criteria of AJCC 7 th edition (“positive” if >0.2mm<br />

metastases were detected).<br />

Result: The overall concordance rate between the OSNA assay and<br />

pathological examination was 91.1% (275/302) with sensitivity of<br />

88.3% (53/60) and specificity of 91.7% (222/242) (Table). These<br />

results are very similar to those of the Japanese clinical validation<br />

study in breast cancer patients without receiving PST which was<br />

conducted by the almost same protocol (Tamaki Y, et al. Clin <strong>Cancer</strong><br />

Res, 2009, 15: 2879-2884).<br />

Conclusion & Discussion: These results indicate the OSNA assay can<br />

be applicable for breast cancer patients after receiving PST as well<br />

as breast cancer patients without receiving PST. The OSNA assay<br />

will enable to examine the whole portion of nodes, leading to more<br />

detection of metastases (especially micrometastases) and more exact<br />

nodal staging for breast cancer patients treated by PST. Also, for the<br />

patients who receive sentinel lymph node biopsy after PST, the OSNA<br />

assay will be useful as intraoperative examination method of SLNs<br />

because it is expected to provide more correct judgments than current<br />

intraoperative methods such as frozen-section or touch-print cytology.<br />

Table<br />

N<br />

Overall concordance<br />

Sensitivity (95% C.I.) Specificity (95% C.I.)<br />

(95% C.I.)<br />

This study (after PST) 302 91.1% (87.3-94.0) 88.3% (77.4-95.2) 91.7% (87.5-94.9)<br />

The Japanese clinical<br />

validation study<br />

(without PST)<br />

450 92.2% (89.4-94.5) 86.1% (76.5-92.8) 93.5% (90.5-95.8)<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

152s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 1<br />

P1-01-13<br />

Patterns of definitive axillary management in the era prior to<br />

reporting ACOSOG Z0011: comparison between NCCN Centers<br />

and hospitals in Michigan<br />

Breslin T, Hwang S, Mamet R, Hughes M, Otteson R, Edge S, Moy B,<br />

Rugo H, Wong Y-N, Wilson J, Laronga C, Weeks J, Silver S, Marcom<br />

P. University of Michigan, Ann Arbor, MI; Duke University; City of<br />

Hope; Dana-Farber/Brigham and Women’s <strong>Cancer</strong> Center; Roswell<br />

Park <strong>Cancer</strong> Institute; University of California <strong>San</strong> Fransicso; Fox<br />

Chase <strong>Cancer</strong> Center; Ohio State University; Moffitt <strong>Cancer</strong> Center;<br />

Massachusetts General Hospital <strong>Cancer</strong> Center<br />

Background: The results of the ACOSOG- Z0011 trial have had<br />

potential practice changing implications for the management of<br />

patients with positive sentinel lymph node (SLN) undergoing<br />

lumpectomy and radiation for breast cancer. However, some evidence<br />

suggests a shift in axillary management even prior to the initial report<br />

of data supporting sentinel lymph node biopsy (SLNB) alone in<br />

mid-2010. We analyzed data in the National Comprehensive <strong>Cancer</strong><br />

Network (NCCN) outcomes database from NCCN centers and the<br />

Michigan <strong>Breast</strong> Oncology Quality Initiative (MiBOQI) hospitals<br />

to examine institutional practice patterns with respect to use of<br />

completion axillary dissection (CALND) for SLN positive breast<br />

cancer in the years leading up to publication of these trial results.<br />

We hypothesized that CALND would be omitted more frequently<br />

in women treated at NCCN centers compared to those treated at<br />

MiBOQI programs.<br />

Methods: We identified 2,172 women with clinical T1/T2 N0 breast<br />

cancer who underwent breast surgery and SLNB and had a positive<br />

SLN from 2007 through 2010 at one of 12 participating NCCN centers<br />

or 12 MiBOQI sites. Patient and tumor characteristics, definitive<br />

breast procedure, year of diagnosis, and institutional affiliation were<br />

analyzed as predictors of use of SLNB alone in univariate Chi-Square<br />

and multivariable logistic regression models.<br />

Results: CALND was omitted in 314 (14.5%) of the 2,172 patients.<br />

Over time, there was a dramatic increase in the use of SLNB<br />

alone (12% in 2007 to 23% in 2010). In the univariate analyses,<br />

increased patient age, later year of diagnosis, lower T stage, and<br />

lower pathologic N stage were significant predictors of use of SLNB<br />

alone (all p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Results<br />

The PSM was based on age, race, region of diagnosis, tumor type and<br />

grade, tumor size, nodal status (micrometastasis vs macrometastasis)<br />

and hormone receptor status. It generated a balanced, matched cohort<br />

(3229 patients in each group) in which baseline characteristics were<br />

not significantly different. Five-year overall survival was 96.9%<br />

(95% CI 96.1-97.6%) in the ALND group and 94.0 (95% CI 92.6-<br />

95.4%) in the SLN biopsy alone group. The benefit of complete<br />

lymphadenectomy was significant: p=0.028.<br />

Conclusion<br />

Using PSM analysis, our results show evidence of benefit for ALND<br />

in case of metastatic sentinel lymph node. The results of randomized<br />

trials demonstrating no benefit for ALND may not been generalizable.<br />

P1-01-15<br />

Accuracy of sentinel lymph node in determining the requirement<br />

for second axillary surgeries in early breast cancer with<br />

retrospective application of the Z0011 criteria.<br />

Waters PS, Fennessy PJ, Alazawi D, Sweeney KJ, Kerin MJ. National<br />

University of Ireland, Galway, Ireland<br />

Background: Lymph node status is the most important prognostic<br />

marker in breast cancer management. The ACOSOG Z0011 trial<br />

reported no difference in survival in patients undergoing sentinel<br />

lymph node biopsy(SLNB) alone versus axillary lymph node<br />

dissection(ALND) in T1-T2 tumours with 1 -2 positive lymph nodes.<br />

Aims: Whether sentinel lymph node biopsy was a true representative<br />

of axillary burden. We also addressed whether application of criteria<br />

from Z0011 trial would have prevented patients undergoing second<br />

axillary surgery.<br />

Methods: All patients with T1-T2 tumours undergoing sentinel<br />

node biopsy were included in our study (n=1019). Analysis of our<br />

prospectively updated breast cancer database was performed. Minitab<br />

version 16.0 was used for statistical analysis.<br />

Results: 1019 SLNB procedures for T1 & T2 tumours were performed<br />

over a 7 year period. 730 patients were reported as histologically<br />

negative and 289 were positive(1 node =95). Of<br />

the lymph node positive group, 223 patients progressed to axillary<br />

clearance. Staging of 149 patients remained unchanged with 74<br />

patients being upstaged with having >2 lymph nodes reported as<br />

positive. 72 patients from the SLNB negative group also had an<br />

axillary clearance. 5 of these patients had further axillary disease<br />

with 1 patient being upstaged having >2 positive lymph nodes. With<br />

application of Z0011 criteria 220 patients would have avoided second<br />

axillary surgery.<br />

Conclusions: The accuracy of sentinel lymph node biopsy in<br />

determining tumour burden has been demonstrated. Furthermore<br />

application of Z0011 will have major cost savings for the health<br />

service with decreased patient stay and morbidity.<br />

P1-01-16<br />

Intraoperatively-palpable “non-sentinel” nodes: should they be<br />

removed?<br />

Crivello ML, Ruth K, Sigurdson ER, Egleston BL, Boraas M, Bleicher<br />

RJ. Fox Chase <strong>Cancer</strong> Center, Philadelphia, PA<br />

Background:<br />

Sentinel lymphadenectomy is the standard of care for evaluation of the<br />

axilla in breast cancer. Not infrequently, despite a clinically negative<br />

axilla, axillary nodes are found and removed that are only palpable<br />

intraoperatively at the time of sentinel lymph node biopsy. There is<br />

limited data suggesting that their resection is beneficial, and there is<br />

some controversy over whether these non-hot, non-blue nodes should<br />

also be called “sentinel nodes” (SNs).<br />

Methods:<br />

A retrospective chart review was conducted of breast cancer patients<br />

who underwent sentinel lymph node biopsy from 2007 to 2011 at<br />

a single institution. Patients were only included if they had SNs<br />

removed (defined as blue and/or radioactive nodes), as well as<br />

additional non-sentinel nodes that were only palpable intraoperatively<br />

(pNSNs) and not part of an axillary dissection (ALND). Pathology<br />

of both SNs and pNSNs were reviewed. Histologic nodal evaluation<br />

was performed by H&E alone.<br />

Results:<br />

From 2007 to 2011, a total of 59 patients had both SNs and pNSNs<br />

removed. Fifty-five patients (93.2%) were female and the average<br />

age was 55 years old. Fifty-two patients (88.1%) had invasive ductal<br />

carcinoma while 7 had lobular carcinoma (11.9%). Average tumor<br />

size was 2.0 cm. Among the 59 patients, a total of 109 SNs (mean<br />

1.8, median 1, range 1-6) and 202 pNSNs (mean 3.4, median 2, range<br />

1-30) were removed at the same surgery. Twenty patients (33.9%)<br />

had metastases in either the SNs and/or pNSNs with 16 (80.0%) of<br />

these having metastases in the SNs alone. Sixteen patients proceeded<br />

to an ALND. There were 4 patients (6.8%, 95% CI 1.9-16.5%) who<br />

had SNs negative for tumor but pNSNs positive for tumor, and 4<br />

(6.8%, 95% CI 1.9-16.5%) separate patients who had extracapsular<br />

extension (ECE) in their pNSNs but no ECE in the SNs. The pNSNs<br />

thus provided information altering operative treatment by American<br />

College of Surgeons Oncology Group (ACOSOG) Z0011 trial criteria<br />

in 8 patients (13.6%). Two patients (3.4%, 95% CI 0.4-11.7%) had<br />

ECE present in ALND nodes, but not present in either their SNs or<br />

pNSNs.<br />

Conclusion:<br />

pNSNs provide information that changes the surgical plan by<br />

ACOSOG Z0011 criteria in 14% of cases. Whether their removal<br />

changes patient outcomes remains unclear, especially as SNs and<br />

pNSNs may, together, be falsely-negative for ECE present within<br />

the axilla. While we recommend their removal at this time based<br />

upon the added information they provide, further study is required to<br />

determine whether the changes resulting from pNSN dissection (i.e.<br />

need for ALND) provide any outcome benefit in this era of effective<br />

systemic therapy.<br />

P1-01-17<br />

The impact of triple negativity on lymph node positivity in breast<br />

cancer.<br />

Weber JJ, Bellin LS, Milbourn DE, Wong J. East Carolina University,<br />

Brody School of Medicine, Greenville, NC; University of North<br />

Carolina, Lineberger Comprehensive <strong>Cancer</strong> Center, Chapel Hill, NC<br />

Introduction<br />

Triple negative breast cancer (TNBC), defined as estrogen,<br />

progesterone, and HER2/Neu receptor-negative is biologically more<br />

aggressive than non-TNBC. Whether TNBC presents at a more<br />

advanced local/regional stage is controversial. We sought to determine<br />

whether TNBC had more frequent lymph node positivity.<br />

Methods<br />

ER and PR status was determined by routine immunohistochemical<br />

analysis. HER2/Neu status was determined by immunohistochemistry;<br />

if 2+ equivocal status was noted, fluorescence in situ hybridization<br />

was performed for confirmation. Patients with a clinically negative<br />

axilla underwent sentinel lymph node biopsy, and if the sentinel<br />

node was positive, underwent immediate completion axillary node<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

154s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 1<br />

dissection. Lymph node positivity was determined by hematoxlyneosin<br />

staining. The main outcome measures were lymph node status<br />

and pathologic tumor size. Statistical differences were compared by<br />

means of paired t-test and chi-squared test where appropriate.<br />

Results<br />

Between January 1, 2004 and December 31, 2010, 501 women<br />

with invasive breast cancer were treated at our institution. These<br />

patients ranged in age from 25 to 93 years (mean 57.6 yr), with<br />

53.1% of patients self-reported as non-Hispanic white and 43.7%<br />

non-Hispanic black. TNBC patients were younger (mean 55.0 yr vs.<br />

58.3 yr, p=0.005), and were more often non-Hispanic black (52.7%<br />

vs. 41.1%, p=0.03). The mean pathological tumor size of the entire<br />

population was 1.86 cm (range 0 to 18 cm). One hundred twelve<br />

patients (22.4%) were TNBC. The mean pathological tumor size<br />

for TNBC compared to non-TNBC patients was 2.07 cm (range 0<br />

to 8.7 cm) and 1.80 cm (range 0 to 18 cm) (p=0.068). There was<br />

a statistically significant difference in the T-stage at presentation<br />

between TNBC and non-TNBC tumors (T1, 58.9% TNBC vs. 72.0%<br />

non-TNBC; T2, 34.8% TNBC vs. 22.4% non-TNBC; T3/T4, 6.3%<br />

TNBC vs. 5.6% non-TNBC, p=0.02). One hundred sixty patients<br />

(31.9%) were node positive. The number of positive lymph nodes in<br />

the TNBC patients ranged from 1-18, and the non-TNBC ranged from<br />

1-35 (TNBC mean 2.94 vs. non-TNBC mean 3.94, p=0.09). TNBC<br />

patients were more often node positive than non-TNBC (39.3% vs.<br />

29.3%, p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P1-01-20<br />

Clinicopathlogic Analysis of Invasive <strong>Breast</strong> Carcinoma with<br />

Micropapillary Component<br />

Yamanaka T, Suganuma N, Yamanaka A, Kojima I, Nishiyama S,<br />

Mukaibashi T, Nakayama H, Matsuura H, Matsuzu K, Chiba A,<br />

Inaba M, Rino Y, Yoshida A, Shimizu S, Masuda M. Yokohama City<br />

University, Yokohama-City, Kanagawa, Japan; Kanagawa <strong>Cancer</strong><br />

Center, Yokohama-City, Kanagawa, Japan; Ito Hospital, Tokyo, Japan<br />

Background: Invasive micropapillary carcinoma(IMPC) is a rare<br />

variant of invasive breast cancer that is associated with a high<br />

incidence of lymph node(LN) metastasis. But, the meaning of focal<br />

micropapillary component has not been fully elucidated.<br />

Methods: We retrospectively reviewed 88 invasive breast carcinoma<br />

cases with IMPC and focal micropapillary component.<br />

Results: Sixty-four cases had micropapillary component only in<br />

operative specimens, 18 cases had only in core-needle specimens,<br />

and 6 cases had in both. Sixteen were diagnosed as IMPC, and 72<br />

cases had focal micropapillary component with other histological<br />

type of invasive breast carcinoma; 65 cases with invasive ductal<br />

carcinoma, 6 cases with mucinous carcinoma and one with metaplastic<br />

carcinoma. There was one male patient. Mean age was 56.5 years<br />

old. Median tumor size was 2.5 cm. Forty two (47.7%) cases were<br />

nuclear grade(NG)1, 16 (18.2%) were NG2 and 30 (34.1%) were<br />

NG3. Seventy eight cases (90%) were ERand/orPgR positive and<br />

18.5% were HER2 positive. Six cases had distant metastasis at<br />

initial diagnosis. LN metastases were observed in 63.6% of all<br />

cases, and this high rate of LN metastasis could be seen even in<br />

small tumor group; 45.5% in cases with tumor equal or less than 2<br />

cm. In the group of IMPC, 40.0% had LN metastasis, and 56.9% in<br />

cases with focal micropapillary component. There was no significant<br />

difference between IMPC and focal micropapillary component<br />

by univariate analysis(p=0.055). Multivariate analysis of all cases<br />

showed that tumor size(>2cm)(p=0.047), NG(1 vs 2&3, p=0.017),<br />

and lymphatic invasion(p=0.001) were significantly associated<br />

with LN metastasis. No significant difference between IMPC<br />

and focal micropapillary component was showed by multivariate<br />

analysis also (p=0.60). Hormone receptor status or HER2 status<br />

were also not correlated(p=0.92, p=0.27 respectively). Among<br />

female patients without neoadjuvant chemotherapy, sentinel lymph<br />

node biopsy(SLNB) were underwent in 38 cases, and 16 cases<br />

(42.1%) had positive sentinel lymph node(SLN). Fourteen patients<br />

underwent additional axillary dissection and 7 cases (50%) had<br />

non-SLN metastases. Among patients with SLNB, 12 patients had<br />

micropapillary component in needle biopsy specimens, and 6 (50%)<br />

cases had positive SLN.<br />

Conclusion: Our data showed very high rate of LN metastasis in<br />

patients with micropapillary component, and this tendency still exists<br />

in cases with only focal micropapillary pattern. High positivity of<br />

SLNB was observed in these groups, and our data suggests that the<br />

findings of micropapillary component in CNB specimen could be<br />

used as a predictor of SLN metastasis.<br />

P1-01-21<br />

Sentinel Lymph Node detection after previous breast tumour<br />

surgical resection: identification rate and false negative rate<br />

through a prospective multi institutional study.<br />

Classe J-M, Andrieux N, Tunon de Lara C, Charitansky H, Lecuru<br />

F, Houpeau J-L, Faure C, De Blaye P, Houvenaeghel G, Kere D,<br />

Marchal F, Raro P, Lefebvre C, Dupré P-F, Rodier J-F. Institut de<br />

<strong>Cancer</strong>ologie de l’Ouest, Nantes Saint Herblain, France; Institut<br />

Bergonié, Bordeaux, France; Institut Claudius Regaud, Toulouse,<br />

France; Centre Hospitalier Georges Pompidou, Paris, France; Centre<br />

Oscar Lambret, Lille, France; Centre léon Bérard, Lyon, France;<br />

Centre Hospitalier Les Oudairies, La Roche sur Yon, France; Institut<br />

Paoli Calmettes, Marseille, France; Institut Jean Godinot, Reims,<br />

France; Centre Alexis Vautrin, Nancy, France; Centre Hospitalier<br />

Universitaire, Angers, France; Centre Hospitalier Morvan, Brest,<br />

France; Centre Paul Strauss, Strasbourg, France<br />

Background: Large multi institutional studies have pointed that<br />

previous surgical resection of breast tumours before axillary sentinel<br />

node detection (ASLND) was the main criteria of failure of this<br />

technique. Screening campaigns provide small tumours and despite<br />

efforts to obtain a diagnosis of early breast cancer, this is not always<br />

obtained , due to small tumours or false negative results of micro<br />

biopsies. The aim of our series was to assess identification rates and<br />

false negative rates of ASLND after previous surgical resection of<br />

breast tumours.<br />

Material and Methods: In a prospective multi institutional setting<br />

(14 multidisciplinary teams), we have included patients with a<br />

previous breast tumour surgical resection for the diagnosis of<br />

infiltrative breast adenocarcinoma. Patients with only a core biopsy<br />

and no surgical removal of the tumor before axillary surgery were<br />

not included. Each patient underwent a secondary surgical procedure<br />

for ASLND and axillary lymphadenectomy, and sometimes a breast<br />

secondary surgical procedure for margins. ASLND was performed<br />

with the combined method, with blue dye and technetium. Pathology<br />

was performed with serial sectioning, eosin safron and immune histo<br />

chemistry (IHC).<br />

Results: From July 2006 to November 2011, 138 patients where<br />

included. The median tumor size was 9mm. Identification rate was<br />

86% (118/138). A macrometastasis was found in 11 cases, in a sentinel<br />

node (9), or in a non sentinel node(2). False negative rate was 9% (1<br />

false negative sentinel node with macrometastasis in non sentinel node<br />

from lymphadenectomy/11 cases with a macrometastasis in either a<br />

sentinel node or a non sentinel node). In 1 case a micrometastasis was<br />

found in a sentinel node through IHC, with a macrometastasis in a<br />

non sentinel node from lymphadenectomy. Without IHC or without<br />

the decision of performing a complementary lymphadenectomy in the<br />

case of micrometastasis, the false negative rate would have been 18%.<br />

Conclusions: After previous surgical resection of early breast cancer,<br />

ASLND remains feasible with a low identification rate of 86%, despite<br />

the use of the combined method. The False negative rate is acceptable<br />

with the use of IHC.<br />

P1-01-22<br />

The utility of axillary ultrasound and sentinel lymph node biopsy<br />

in the management of metaplastic breast carcinoma<br />

Hieken TJ, Fazzio RT, Reynolds C, Jones KN, Ghosh K, Glazebrook<br />

KN. Mayo Clinic, Rochester, MN<br />

Background: Metaplastic breast carcinoma (MBC) accounts for


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 1<br />

or osseous) elements. Patients typically present with large triple<br />

negative tumors and have a poor prognosis. The likelihood of lymph<br />

node (LN) involvement has been reported to be low in MBC patients.<br />

Due to the paucity of data, we undertook this study to explore the<br />

role of axillary ultrasound (US) and sentinel LN biopsy (SLNB) in<br />

the management of MBC.<br />

Methods: With IRB approval, we retrospectively identified patients<br />

diagnosed with MBC from 2001-2011 from the surgical pathology<br />

database. Histopathology and imaging were reviewed. Demographic,<br />

treatment and outcome data were obtained by clinical chart review.<br />

Data were analyzed using JMP 9.0 software.<br />

Results: We identified 41 women with MBC. Median age was 60 years<br />

(range 33-90). Histologic subtypes were spindle cell (46%), mixed<br />

adenocarcinoma and mesenchymal elements (20%), squamous cell<br />

(17%) chondroid/osseous (10%) and adenosquamous (7%). Tumor<br />

stage was T1 (24%), T2 (44%), T3 (12%) and T4 (20%). 26 patients<br />

(63%) were treated by mastectomy and 15 (37%) by wide excision.<br />

Of the 38 patients who underwent LN surgery (6 low-grade MBC, 32<br />

intermediate/high-grade MBC), 10 (26%) were LN positive. All lowgrade<br />

MBC patients were LN negative while 10 of 32 intermediate/<br />

high-grade MBC patients (31%) had LN metastasis. 22 patients had a<br />

preoperative axillary US. 14 patients had a negative axillary US and<br />

none had LN metastasis. 4 of 8 patients with suspicious axillary US<br />

findings had a preoperative axillary LN fine needle aspiration biopsy<br />

(FNAB) and 3 were positive for cancer. These patients proceeded<br />

directly to axillary LN dissection (ALND). 24 patients had a SLNB<br />

of whom one was SLN positive and underwent completion ALND.<br />

LN metastasis was associated with larger tumor size (p=0.003), higher<br />

tumor grade (p=0.04), angioinvasion (p=0.07) and abnormal axillary<br />

US (p=0.003). Surviving patients were followed for a mean (median)<br />

of 41 (30) months during which 13 (32%) recurred at a median of<br />

6 months (IQR 3-17 months) and 11 (27%) subsequently died of<br />

disease. One SLN negative patient developed an axillary recurrence<br />

at 8 months, was successfully treated by ALND and is disease-free at<br />

38 months. There were no axillary LN relapses after ALND.<br />

Conclusions: For clinically node-negative MBC patients, our<br />

contemporary data series suggests the incidence of occult LN<br />

metastasis is sufficiently high to warrant LN staging especially<br />

for patients with intermediate and high-grade tumors. This can<br />

be accomplished in a minimally invasive fashion with reasonable<br />

accuracy (∼97%) with axillary US, FNAB of sonographically<br />

suspicious LNs, and SLNB for patients with negative axillary US or<br />

FNAB. Axillary LND should be performed for patients with clinically<br />

and/or FNAB-positive LN for enhanced disease control.<br />

P1-01-23<br />

Increased Diagnostic Performance of Sentinel Lymph Node<br />

Biopsy Combined with Radiologic-pathologic Factors After<br />

Neoadjuvant Chemotherapy in <strong>Breast</strong> <strong>Cancer</strong> Patients with<br />

Cytologically Proven Node Metastasis at Diagnosis<br />

Park S, Koo JS, Kim MJ, Park JM, Cho JH, Hwang H, Park HS,<br />

Kim E-K, Kim SI, Park B-W. Yonsei University College of Medicine,<br />

Seoul, Korea<br />

Background: It is undetermined whether sentinel lymph node biopsy<br />

(SLNB) is feasible and accurate for predicting final nodal status after<br />

neoadjuvant chemotherapy (NCT) in breast cancer patients with<br />

cytologically proven node metastasis at the time of diagnosis, although<br />

currently completion node dissection is a standard surgical procedure<br />

for the management of axilla. The aim of this study was to investigate<br />

diagnostic performance of SLNB after NCT in this subgroup.<br />

Methods: Of 374 patients with T1-T3 breast cancer who received<br />

NCT, 178 had initially biopsy proven axillary/supraclavicular<br />

metastasis and subsequently underwent SLNB using radioisotope<br />

alone followed by completion node dissection between 2008 and<br />

2011. Detection rate, sensitivity, false negative rate (FNR), negative<br />

predictive value (NPV), and accuracy of SLNB were retrospectively<br />

analyzed and it was explored using regression analysis whether<br />

combination with clinicopathologic factors improved performance.<br />

Results: At initial presentation, 60.7% of patients were cT2 stage<br />

and 88.2% treated with concurrent or sequential anthracycline plus<br />

taxane preoperatively. SLNB was successfully performed in 169<br />

(94.9%) patients. The mean number of sentinel and regional nodes<br />

retrieved was 2.1 ± 1.6 and 12.8 ± 6.3, respectively. Tumoral nonresponder<br />

and extensive residual nodal disease were significantly<br />

associated with detection failure of SLNB. Conversion to nodenegative<br />

disease was noted in 69 (40.8%) patients. Sensitivity, FNR,<br />

NPV, and accuracy of SLNB were 78.0%, 22.0%, 75.8%, and 87.0%,<br />

respectively and diagnostic performance increased when ≥ 3 sentinel<br />

nodes were evaluated. By logistic regression model, tumoral and nodal<br />

responder, absent lymphovascular invasion (LVI), estrogen receptor<br />

(ER)-negativity, and HER2-positivity were significantly associated<br />

with node-negative disease after NCT. Area under the receiver<br />

operating characteristic curve increased from 0.890 to 0.949 when<br />

considering radiologic-pathologic factors and FNR was the lowest<br />

value of 14.3% in 46 patients with tumoral responder, absent LVI,<br />

and ER-negative tumor.<br />

Conclusions: SLNB was technically feasible but solely showed<br />

higher FNR in this study. Improved diagnostic performance of SLNB<br />

combined with radiologic-pathologic characteristics suggests possible<br />

clinical value of SLNB after NCT in highly selected patients with<br />

node metastasis at diagnosis.<br />

P1-01-24<br />

Which combinations are helpful to predict axillary lymph node<br />

metastasis in T1 breast cancer with ultrasonography and contrastenhanced<br />

MRI and contrast-enhanced 18F-FDG PET-CT?<br />

Hwang SO, Park HY, Jung JH, Kim WW, Lee YH, Lee JJ, Choi HH,<br />

Hwangbo SM. Kyungpook National University School of Medicine,<br />

Daegu, Korea; Hyosung Hospital, Daegu, Korea<br />

Backgrounds: Axillary lymph node(ALN) status has been important<br />

factor of treatment and prognosis for patients with breast cancer. Even<br />

though the better ultrasonographic instruments have been developed,<br />

it is still difficult to predict axillary lymh node metastasis (ALNM)<br />

with only ultrasonography(US) in T1 breast cancers which most of<br />

newly diagnosed breast cancers are recently since T1 breast cancers<br />

have low rate and less tumor burden of ALNM. This study evaluated<br />

the accuracy of prediction of ALNM in T1 breast cancer with US,<br />

contrast-enhanced MRI (cMRI) and contrast-enhanced 18F-FDG<br />

PET/CT (cPET/CT) and found out adequate combinations of these<br />

modalities.<br />

Method: Retrospectively, we reviewed 351 breast cancer patients with<br />

tumors(T1) ≤2cm in size between January 2008 and December 2011<br />

who were preoperatively examined with US, cMRI, and cPET/CT and<br />

underwent pathologic evaluation of axillary lymph nodes acquired<br />

by sentinel lymph node biopsy or axillary dissection.<br />

www.aacrjournals.org 157s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Clinical characteristics(n=351)<br />

Character-<br />

Character-<br />

Character-<br />

Charactern(%)<br />

n(%)<br />

n(%)<br />

n(%)<br />

istics<br />

istics<br />

istics<br />

istics<br />

Age(yrs) Surgery T stage Histology<br />

Mean(range) 51.3±10.2 <strong>Breast</strong><br />

(25∼79)<br />

1mi 10(2.8) IDC 319(90.9)<br />

≤40 52(14.8) BCS 231(65.8) 1a 17(4.8) ILC 15(4.3)<br />

>40 299(85.2) MRM 120(34.2) 1b 53(15.1) mucinous 8(2.3)<br />

Sex Axilla 1c 271(77.2) others 9(2.5)<br />

Female 349(99.4) SLNB 266(75.8) N stage Grade<br />

Male 2(0.6) SLNB+ALND 73(20.8) 0 257(73.2) 1 91(25.9)<br />

Palpability of ALN ALND 12(3.4) 1mi 8(2.3) 2 170(48.4)<br />

Yes 8(2.3) 1 68(19.4) 3 79(22.5)<br />

No 343(97.7) 2 15(4.3)<br />

3 3(0.9)<br />

ALN:axillary lymph node;BCS:breast conserving surgery;MRM:modified radical<br />

mastectomy;SLNB:sentinel lymph node biopsy;ALND:axillary lymph node dissection; IDC:invasive<br />

ductal carcinoma, NOS;ILC:invasive lobular carcinoma<br />

Prediction of Axillary lymph node metastasis by image studies<br />

Sensitivity Specificity PPV NPV Accuracy<br />

USG 0.457 0.887 0.597 0.817 0.772<br />

cMRI 0.489 0.887 0.613 0.826 0.780<br />

cPET/CT 0.447 0.942 0.737 0.823 0.809<br />

Used<br />

modalities<br />

≥1 0.563 0.809 0.520 0.835 0.744<br />

≥2 0.478 0.914 0.671 0.827 0.798<br />

≥3 0.351 0.992 0.943 0.807 0.821<br />

USG&cMRI 0.426 0.934 0.702 0.816 0.798<br />

USG&cPET/<br />

CT<br />

0.362 0.977 0.850 0.807 0.812<br />

cMRI&cPET/<br />

CT<br />

0.394 0.988 0.925 0.817 0.829<br />

PPV:positive predictive value;NPV:negative predictive value<br />

Results: 94(26.8%) patients of 351 had ALNM. The sensitivity,<br />

specificity, positive predictive value(PPV), negative predictive value<br />

(NPV), and accuracy of ALNM with US were 0.457, 0.887, 0.597,<br />

0.817, 0.772, respectively. cMRI had similar results with US. The<br />

sensitivity, specificity, positive predictive value (PPV), negative<br />

predictive value (NPV), and accuracy of ALNM with cPET/CT were<br />

0.447, 0.942, 0.737, 0.823, 0.809, respectively. The sensitivity if any<br />

one or more modalities were suspicious was 0.563. The specificity<br />

if all modalities were suspicious was 0.992. The PPV if cMRI and<br />

cPET/CT were suspicious was highest than if other combinations<br />

were suspicious.<br />

Conclusion: US, cMRI, and cPET/CT are helpful in prediction of<br />

ALNM of T1 breast cancers. However, there are no definite modality<br />

and combination of modalites to predict ALNM of T1 breast cancers.<br />

P1-01-25<br />

Sentinel lymph node biopsy in breast cancer: The approach in day<br />

surgery under local anaesthesia for quality-of-life and significant<br />

cost reduction<br />

Ricci F, Capuano LG, Saralli E, Di Legge P, Violante A, Polistena A,<br />

Scala T, Pacchiarotti A, Cannas P, Cianni R, Fanelli G, Bellardini P,<br />

De Masi C. S.M. Goretti Hospital, Latina, Italy; LILT, Latina, Italy;<br />

S.M. Goretti, Latina, Italy<br />

Background: Sentinel lymph node biospy (SLNB) is widely used in<br />

the management of breast cancer patients without axillary metastases<br />

or inflammatory breast cancer (IBC).<br />

Methods: From Jan. 1 st 2006 through Dic. 31 st 2011 we performed<br />

302 SLNB at St. M. Goretti Hospital. Mammary carcinoma was<br />

diagnosed as malignant by aspiration citology and/or biospy. In all<br />

cases with positive citology or biospy, we performed quadrantectomy<br />

and SLNB at the same time. All patients underwent pre-operative<br />

lymphoscintigraphy with intradermal pericicatritial and/or periareola<br />

injection of 12-15 MBq 99Tc colloidal albumin particles 50-80 nm<br />

size range, in 0,2 ml saline solution. We never used vital blue dye.<br />

All patients underwent surgical treatment 3-12 h. later. We performed<br />

SLNB and quadrantectomy in day surgery (DS) and local anaesthesia<br />

(LA) with 2% of Carbocaine. Axillary incision was 3-4 cm. This study<br />

was approved by an ethics committee, was discussed with all patients<br />

and informed consent was obtained.<br />

Purpose of the study is to investigate the approach in DS under LA<br />

for quality of life and significant cost reduction.<br />

Results: Six patients underwent pre-operative lymphoscintigraphy<br />

the radiotracer did not show any sentinel lymph node (SLN), in five<br />

cases we performed axillary dissection (AD). In one case of young<br />

patient who had previously been treated with chemotherapy for<br />

non-Hodgkin’s lymphoma, negative positron emission tomography<br />

(PET), we performed quadrantectomy without AD. In three cases<br />

the axilla was positive. In four cases of multifocal (MF) and two of<br />

multicentric (MC) invasive breast cancer, the SLN was identified<br />

in axilla and SLNB was perfomed. SLNB in MF and MC tumors<br />

was similar to unifocal cancers. Only one case of MC cancer the<br />

SLN was positive. Six patients classified T4b according to AJCC,<br />

were treated with neoadjuvant chemotherapy (NC). The axilla was<br />

negative to ultrasound (US), PET and citology. After completion of<br />

NC, lymphatic mapping was able to identify SLN and we performed<br />

SLNB. In these patients SLN was negative. In two cases of male<br />

cancer the axilla was negative to clinical examination, in both cases<br />

SLN was positive for macrometastases. Six cases showed axillary<br />

isolated tumor cells (ITC). Eighteen micrometastases. Thirty-two<br />

macrometastases. In two case of negative SLN there was a positive<br />

second palpable lymph node. One case showed a double SLN in<br />

the axilla and internal mammary chain, only the internal mammary<br />

lymph node was positive.<br />

The SLN identification rate was 99%. After surgery we distributed a<br />

questionnaire to the patients about the acceptability of this approach.<br />

P1-01-26<br />

Procoagulant biomarkers may direct axillary nodal surgery<br />

Shaker H, Bundred NJ, Glassey E, Kirwan CC. University Hospital<br />

of South Manchester, Manchester, United Kingdom; University of<br />

Manchester, United Kingdom<br />

Background<br />

Preoperative accurate staging of the axilla (through identifying<br />

invasive breast cancers with subclinical nodal metastases) will reduce<br />

the need for repeat axillary surgery.<br />

Tissue factor (TF) is overexpressed in cancer and is shed into the<br />

circulation leading to activation of the thrombin (extrinsic clotting)<br />

pathway. D-dimer is a fibrin degradation product and reliable systemic<br />

marker of thrombin pathway activation. TF and d-dimer are raised in<br />

breast cancer and may act as biomarkers to identify invasive cancer<br />

patients with undiagnosed lymph node (LN) metastases requiring<br />

axillary nodal clearance.<br />

Methods<br />

Plasma d-dimer and TF were measured pre-operatively using ELISA<br />

in patients undergoing curative surgery and sentinel node biopsy<br />

(SNB) for invasive breast cancer (n=125). D-dimer was considered<br />

to be raised if it was above the clinically used upper limit of normal<br />

of 500ng/ml and if TF was above 200pg/ml. D-dimer and TF were<br />

correlated with the presence or absence of axillary metastases at<br />

sentinel node biopsy.<br />

Results<br />

Median age was 61 years (range 24-84). Mean invasive tumour size<br />

was 16.7 mm (range 1.6-70). One hundred and eleven (79%) invasive<br />

cancers were oestrogen receptor positive, 93 (67%) were progesterone<br />

receptor positive and 17(12%) were HER2 receptor positive.<br />

Pre-operative d-dimer was higher in LN positive (mean 599 ng/<br />

ml,95% CI 415-864, n=32) versus LN negative (454ng/ml, 95% CI<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

158s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 1<br />

400-514, n=93, p=0.068) invasive cancer. TF was not significantly<br />

higher in LN positive cancer (vs LN negative), although TF did show<br />

weak correlation with d-dimer (r=0.2, p=0.03).<br />

Sixty eight patients (54%) had raised TF and 59 (47%) had raised<br />

d-dimer. On multivariate analysis (age, tumour size, invasive grade,<br />

ER status and presence of lymphovascular invasion as covariates) a<br />

raised pre-operative d-dimer combined with a raised pre-operative<br />

TF was a significant independent predictor of node positivity (odds<br />

ratio 6.43, CI 1.70 – 24.27, p=0.006). Markers of systemic thrombin<br />

pathway activity (i.e. a combination of plasma d-dimer and TF) had<br />

a sensitivity of 91%, specificity of 32%, positive predictive value of<br />

32% and negative predictive value of 92%.<br />

D-dimer >500ng/ml AND TF >200pg/ml<br />

No Yes<br />

Lymph node metastasis No 30 63 93<br />

Yes 3 29 32<br />

33 92 TOTAL<br />

Conclusion<br />

High levels of clotting activation are seen in breast cancer patients.<br />

Over 90% of patients with no axillary lymph node metastases had<br />

normal pre-operative levels of plasma TF and d-dimer. Combination<br />

of systemic markers of thrombin pathway activation may help predict<br />

axillary nodal involvement.<br />

The role of thrombin pathway activation in breast cancer metastasis<br />

is under ongoing investigation.<br />

P1-01-27<br />

Novel diagnostic procedure of metastasis to the sentinel lymph<br />

node of breast cancer using a semi-dry dot-blot method<br />

Otsubo R, Oikawa M, Shibata K, Hirakawa H, Yano H, Matsumoto<br />

M, Hatachi T, Nakao K, Hayashi T, Abe K, Kinoshita N, Nakashima<br />

M, Taniguchi H, Omagari T, Itoyanagi N, Nagayasu T. Nagasaki<br />

University Hospital, Nagasaki, Japan; The Japanese Red Cross<br />

Nagasaki Genbaku Hospital, Nagasaki, Japan; Aiyuukai Memorial<br />

Hospital, Chiba, Japan; Nagasaki University Atomic Bomb Disease<br />

Institute, Nagasaki, Japan; St. Francis Hospital, Nagasaki, Japan<br />

Background: Sentinel lymph node (SLN) biopsy is a common<br />

surgical procedure. However, novel diagnostic modalities are<br />

required because of a shortage of pathology specialists in Japan<br />

and discordance between the intraoperative and final pathological<br />

diagnosis of SLN metastasis. Efficient methods that use molecular<br />

markers for detecting SLN metastasis, such as one-step nucleic acid<br />

amplification, are commercially available, but special equipment<br />

(e.g., thermal cycler) is costly and the problem of false-negatives<br />

for CK19 non-expressing cells remains unresolved. Recently, we<br />

developed an easy, quick and cost-effective method for detection of<br />

cancer cells in lymph nodes by applying dot-blot analysis technology,<br />

called the “semi-dry dot-blot method (SDB method)”. The SDB<br />

method visualizes the presence of cancer cells with washing of<br />

sectioned lymph nodes by anti-pancytokeratin antibody (AE1/AE3)<br />

and chromogen on a dot-blot membrane. This method can detect 0.01<br />

mg/mL protein extracted from cancer tissue and 20 suspension cells<br />

(MCF-7) in approximately 20 minutes. The current study evaluated<br />

the efficacy of our SDB method in the diagnosis of SLN metastasis<br />

in breast cancer.<br />

Methods: (I) One hundred eighty dissected lymph nodes from 29<br />

cases, including breast, lung, gastric and colorectal cancer, were<br />

analyzed. Each lymph node was sliced at the maximum diameter<br />

and washed by phosphate-buffered saline (PBS), and this lavage<br />

fluid (PBS) was used for diagnosis of LN metastasis by the SDB<br />

method, and washed lymph node was sent to pathological section<br />

for pathological diagnosis. The sensitivity, specificity and accuracy<br />

of the SDB method were determined to compare with the final<br />

pathology report.<br />

(II) A multicenter prospective clinical trial was conducted, where 132<br />

SLN samples from 78 cases of clinically node-negative breast cancer<br />

were analyzed. Each SLN was sliced at 2-mm intervals and washed by<br />

PBS. Lavage fluid of sliced SLNs was used for the diagnosis of SLN<br />

metastasis by the SDB method in a blinded manner. The sensitivity,<br />

specificity, accuracy and the time required for the SDB method were<br />

determined and compared with the intra-operative pathology report.<br />

Results: (I) Lymph node metastasis was detected in 35 lymph nodes<br />

(19.4%). Comparison of the results between the final pathology and<br />

the SDB method showed complete concordance (accuracy: 100%).<br />

(II) Of the 132 lymph nodes eligible for analysis, 13 (10.0%) were<br />

assessed as positive and 114 as negative by the SDB method and intraoperative<br />

pathological examination, with an accuracy of 96.2%. All<br />

pathologically positive nodes, which included one micro metastasis,<br />

were also detected by the SDB method; (sensitivity: 100%). One<br />

hundred fourteen of the 119 pathologically negative nodes were<br />

assessed as negative with the SDB method (specificity: 95.8%). The<br />

mean required times of 16 cases for intra-operative pathological<br />

diagnosis and simultaneous SDB method were 43.5 and 42.2 minutes,<br />

respectively.<br />

Conclusions: The SDB method is simple, fast, accurate and costeffective<br />

for the intra-operative diagnosis of SLN metastasis.<br />

Because there is no loss of lymph node tissue, the SDB method and<br />

pathological investigation can be performed simultaneously.<br />

P1-01-28<br />

What is the actual impact of micrometastases in sentinel lymph<br />

nodes in regards to global survival and disease free survival<br />

Barbosa EM, Araujo Neto JT, Filho EC, Infante KP, Felzener MM,<br />

Matielle CR, Modesto MG, Basso R, Goes JCS. Instituto Brasileiro<br />

de Controle do <strong>Cancer</strong> - IBCC, Sao Paulo, Brazil<br />

OBJECTIVE: Assess the clinical relevance of micro metastasis<br />

in sentinel lymph nodes in the overall survival and disease free<br />

survival of 1,211 patients undergoing sentinel lymph node biopsy.<br />

METHODS: Retrospective cohort study including 1,211 patients<br />

undergoing consecutive SLB from 1998 to 2010. SLB were performed<br />

in patients with invasive carcinoma of the breast with clinically<br />

negative axilla. Age, tumor size, and histologic grades, estrogen and<br />

progesterone receptors expression, HER-2, lymphovascular invasion<br />

and size of metastases in lymph nodes were assessed. Tumor sizes<br />

were divided according to pathologic staging into pT1(< 2,0mm)<br />

and pT2(>2,0mm). Lymph nodes were classified as negative (pN0),<br />

positive with micro metastases with diameter ranging from 0,21mm<br />

to 2,0mm(pN1mi) and positive with macro metastases with diameter<br />

greater than 2,0mm(pN1). Statistical analyses were performed by<br />

the Likelihood ratio test or Chi-square test for insufficient samples<br />

between macro metastases and micro metastases in SLB. Time was<br />

considered in months and the overall follow-up time was 125 months<br />

and mean time was 65 months. Local regional and distant recurrences<br />

were considered as events for disease free survival and Kaplan-<br />

Meier survival functions were constructed, testing time between<br />

metastases sizes in SLB using the Log-rank test, with a significance<br />

level of 5%. RESULTS: Of the studied variables, lymphovascular<br />

invasion(p=0,068) and local regional and distant metastases(p=0,002)<br />

had a statistically significant association with overall survival. Tumor<br />

size(p=0,016); lymph vascular invasion(p=0,002) and metastases<br />

types in sentinel lymph node(p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

lymph node, the following were statistically significant: age(p=0,004),<br />

tumor size(p


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 1<br />

identified. While there may be a trend towards not excising some of<br />

these high-risk lesions, we believe that a core biopsy demonstrating<br />

FEA still warrants surgical excision.<br />

P1-02-02<br />

Receptor discordance in breast cancer recurrence: Is re-biopsy<br />

a necessity?<br />

Fujita T, Sawaki M, Hattori M, Kondo N, Horio A, Ushio A, Gondo<br />

N, Hiroji I. Aichi <strong>Cancer</strong> Center Hospital, Nagoya, Japan<br />

Background: Decision making on systematic treatment of patients<br />

with metastatic breast cancer is based on features like estrogen<br />

receptor (ER), progesterone receptor (PgR), and HER2 status assessed<br />

on the primary tumor. Recent prospective studies have investigated<br />

discordance in receptor status between the primary and metastatic<br />

tumor. We evaluated the discordance of receptor status between the<br />

primary and metastatic tumor and assessed the impact of re-biopsy<br />

on patient management.<br />

Methods: In breast cancer patients who underwent surgical resection<br />

of primary tumor and re-biopsy of metastatic tumor at Aichi <strong>Cancer</strong><br />

Center hospital, we examined the discordance of ER status, PgR<br />

status, and HER2 status between the primary and metastatic tumor. For<br />

sampling of metastases, core needle biopsy (CNB), trans-bronchial<br />

lung biopsy (TBLB), or surgical resection was performed. And fineneedle<br />

aspiration biopsy was not used.<br />

The ER and PgR status were assessed using Allreds scoring system<br />

by IHC. These statuses were categorized as positive when the total<br />

score was more than two. HER2 expression status was tested by IHC<br />

and FISH. HER2 3+ by IHC, or 2+ and FISH positive were judged<br />

as HER2 positive.<br />

Results: 48 patients underwent re-biopsy from 2003 to 2012. Twentyeight<br />

were loco-regional (local: 25, Supraclavicular: 2, Axilla: 1) and<br />

21 were distant (Lung: 14, Liver: 6, Bone: 1). Discordance in ER,<br />

PgR, or HER2 between the primary and metastatic tumor were 6.2%<br />

(3/48), 16.7% (8/48), and 4.2% (2/48), respectively. One patient had<br />

discordance on re-biopsy with initial triple negative primary tumor<br />

(1/5, 20%). 11/48 patients (22.9%) changed the molecular subtype<br />

(Group A), and 37/48 patients (77.1%) did not change (Group B).<br />

Time to recurrence (TTR) was significantly longer in Group A than<br />

Group B (82.3 months vs 51.2 months, p=0.04). No significant<br />

difference in re-biopsy techniques (CNB and TBLB vs surgical<br />

resection), metastatic lesions (loco-regional vs distant), and adjuvant<br />

therapy were observed between Group A and Group B.<br />

Conclusions: Our results show the low level of discordance for ER<br />

and HER2 between the primary and metastatic tumor. But re-biopsy<br />

should be considered in the patients with long TTR, since it is likely<br />

to impact treatment choice. Further study is needed to determine the<br />

value of re-biopsy.<br />

P1-03-01<br />

Evidence for the Warburg effect in mammary atypia from highrisk<br />

African American women.<br />

Seewaldt V, Hoffman A, Ibarra-Drendall C. Duke University, Durham,<br />

NC<br />

Background: Aggressive cancers are known to consume glucose<br />

avidly and produce lactic acid (rather than fully metabolize glucose<br />

via the Tricarboxylic Acid (TCA) cycle). This shift toward lactate<br />

production, even in the presence of adequate oxygen, is termed the<br />

Warburg effect. The Warburg effect is thought to be a late event in<br />

breast cancer, however, our studies in high-risk women provide<br />

evidence that the Warburg effect occurs during cancer initiation.<br />

This is an important observation as glucose-signaling can be readily<br />

targeted for breast cancer prevention with minimal toxicity. Here we<br />

investigated the role of the Warburg effect in breast cancer initiation<br />

in young high-risk women.<br />

Methods and Results: Similar to fluorodeoxyglucose, 2-(N-(7-<br />

nitrobenz-2-oxa-1,3-diazol-4-yl)amino)-2-deoxyglucose (2-NBDG)<br />

is a fluorescent glucose analog that can be used to track glucose uptake<br />

and glycolysis. 2-NBDG spectroscopy provides a means to track<br />

glucose metabolism in live mammary epithelial cells from high-risk<br />

women. We used 2-NBDG spectroscopy to measure glucose uptake<br />

in ER- breast cancer and live atypical mammary epithelial cells<br />

from high-risk premenopausal women. We observe that both triplenegative<br />

breast cancer and a subset of atypia exhibits accumulation<br />

of 2-NBDG.<br />

There is growing recognition that phosphoprotein signaling networks<br />

(rather than single genes) play a key role in breast cancer initiation<br />

and progression. Our team used Reverse Phase Proteomic Microarray<br />

(RPPM) profiling to test for activation of phosphoprotein signaling<br />

networks in atypical RPFNA cytology from high-risk premenopausal<br />

women in our cohort. RPFNA were obtained from two independent<br />

sets of 39 and 38 high-risk premenopausal women; 45% of these<br />

women were African American. The signaling network most highly<br />

expressed in precancerous cells contained activated signaling proteins<br />

associated with the Warburg effect (AKT/mTOR/PI3K), insulin<br />

signaling (pACC, IRS1) and epithelial to mesenchymal transition<br />

(EMT) IL6/Stat3/vimentin.<br />

Conclusions: This is the first evidence that abnormal glucose uptake<br />

and the Warburg effect occurs during breast cancer initiation in<br />

high-risk premenopausal women. These studies demonstrate our<br />

ability to identify abnormal glucose and activated signaling networks<br />

associated with the Warbug effect in atypical mammary cells from<br />

high-risk women.<br />

P1-03-02<br />

“Normal” tissue adjacent to breast cancer is not normal<br />

Clare SE, Pardo I, Mathieson T, Lillemoe HA, Blosser RJ, Choi M,<br />

Sauder CAM, Doxey DK, Badve S, Storniolo AMV, Atale R, Radovich<br />

M. Indiana University School of Medicine, Indianapolis, IN; Susan<br />

G. Komen for the Cure Tissue Bank at the IU Simon <strong>Cancer</strong> Center,<br />

Indiana University School of Medicine, Indianapolis, IN<br />

Background: Gene expression data from pancreatic cancer,<br />

histologically normal tissue adjacent to the cancer and normal<br />

pancreas reveals that adjacent normal has already acquired a number<br />

of transcriptional alterations and is not, therefore, an appropriate<br />

baseline for comparison with cancers. (Gadaleta et al., 2011) The<br />

purpose of this study was to determine if this is also the case for<br />

breast cancer and, if so, to identify the differences in gene expression<br />

between adjacent normal and normal breast.<br />

Methods: RNA-Seq data from breast cancer and adjacent normal was<br />

downloaded from the TCGA (The <strong>Cancer</strong> Genome Atlas) data portal.<br />

The epithelia from 20 frozen tissue cores from healthy premenopausal<br />

donors to the Susan G. Komen for the the Cure® Tissue Bank at the<br />

IU Simon <strong>Cancer</strong> Center were microdissected and the RNA isolated.<br />

RNA-seqeuncing was carried out using the Life Technologies SOLiD<br />

Platform. RPKM gene expression values from TCGA and sequencing<br />

of the Komen normal tissues were merged, quantile normalized, and<br />

batch effect corrected. Normalization and differential gene expression<br />

was performed using Partek Genomics Suite.<br />

Results: Principal component analysis (PCA) reveals complete<br />

separation between adjacent normal and healthy normal breast tissue.<br />

Setting a maximum FDR (false discover rate) of 5%, 2239 genes are<br />

www.aacrjournals.org 161s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

differentially expressed between adjacent normal and healthy normal.<br />

Ingenuity pathway analysis reveals that the Fos, Jun and TGFbeta<br />

pathways are active in the adjacent normal.<br />

Conclusions: Tissue adjacent to a primary breast cancer is not normal<br />

when using healthy breast tissue as a comparator. As RNA-Seq data<br />

is digital, it is possible to quantify the changes in gene expression<br />

starting from healthy normal to normal adjacent to tumor to tumor.<br />

Increasing and decreasing gene expression values provide clues to the<br />

fundamental molecular changes occurring in histologically normal<br />

appearing adjacent tissue. The differences in gene expression we have<br />

identified are some of the earliest changes in breast carcinogenesis<br />

and provide insight into the etiology of this disease and, potentially,<br />

its prevention.<br />

P1-03-03<br />

milk deposition in women’s mammary duct has been a potential<br />

risk factor of breast tumor<br />

Xu Z, Gu Y, Gong G, Zhang Y. <strong>Breast</strong> Disease Institution of Jilin<br />

Province, Changchun, Jilin, China; Spinal Disease Institution of<br />

Jilin Province, Changchun, Jilin, China<br />

Background: According to the pathology and general resection<br />

specimens, so many patients have breast tumors together with<br />

deposited milk. Therefore, we performed this retrospective study to<br />

determine the distribution and characteristics of breast diseases in<br />

women undergoing mammary ductoscopy and explore the clinical<br />

values and implications of mammary ductoscopy in the management<br />

of the breast diseases.<br />

Materials & Methods: Total of 10,082 women(20,082 breasts) with<br />

a mean age of 39.2 years old (ranging from 12 to 84 years old)<br />

underwent mammary ductoscopy from 2002 to 2011.We studied<br />

NSND in contrast with SND duct, specially focused on the situation<br />

of duct with intraductal mass. Comparing with the pathology, we<br />

studied the relationship between duct milk deposition and epithelial<br />

hyperplasia.<br />

Results:<br />

Table 1. disease distribution under mammary ductoscopy of 20,018 ductoscopic investigations<br />

(cases)<br />

FDS Reports NSND SND cases n(%)<br />

Intraductal mass 17 1414 1,431(7.15)<br />

Ductal milk deposition 3981 4604 8,585(42.89)<br />

Duct occlusion inflammation 6613 0 6,613(33.04)<br />

Duct inflammation with dilation 970 53 1,023(5.11)<br />

Duct dilation 454 1836 2,290(11.44)<br />

Negative results 76 0 76(0.4)<br />

From this study we concluded that duct milk deposition was a<br />

common disease in 25-59 years old women, it occupied 42.89%<br />

cases of the mammary ductal disease and almost 46% are NSND.<br />

The character of deposited milk became solid from liquid with the<br />

age growing up. Duct milk deposition 476/1431(33.26%)was mostly<br />

associated with intraductal masses(total 1431 cases), followed by<br />

duct dilatation 517/1431(36.13%). Pathology of patients diagnosed<br />

by FDS for duct milk deposition are epithelial hyperplasia(100%),<br />

adenosis(86.5%), chronic inflammation(8.8%), metaplasia(16.9),<br />

florid hyperplasia(10.8%), tumor-like hyperplasia (57.2%), intraductal<br />

tumor or fibroadenoma (27.8%), dysplasia (3.0%), breast cancer<br />

(11.7%).<br />

Discussion: Diagnosis of duct milk deposition, which may not<br />

necessarily presented by nipple discharge, is the most common finding<br />

and associated with proliferation and thus intraductal mass, etc. we<br />

should noticed 30-50 years old women groups,cause they have have<br />

stopped breast-feeding for years but still had yellow or brown, liquid<br />

or solid milk-like things in their lactiferous ducts. We predict duct<br />

milk deposition may be related with breast carcinogenesis. It is a kind<br />

of long time mechanical stimulation in the duct, both the physicochemical<br />

properties and the acid-alkaline environment had changes by<br />

years. Still, the deposited milk would be a best medium for microbial<br />

and virus , which would cause the genesis of hyperplasia even breast<br />

cancer. Therefore, we should go on the research of deposited milk<br />

and the changes of the microenvironment of the duct to find the<br />

pathogenesis of breast disease.<br />

P1-04-01<br />

Loss of Notch4 reduces mammary tumorigenesis by MYC and<br />

activated KRAS<br />

Rodriguez EM, Bishop JM. G. W. Hooper Research Foundation,<br />

University of California, <strong>San</strong> Francisco, CA<br />

Over-expression of Notch receptors and aberrant Notch signaling<br />

have been reported in both preinvasive ductal carcinoma in situ<br />

and invasive breast cancer. The Notch 4 receptor, in particular, is<br />

preferentially activated in human breast cancer stem cell-enriched<br />

populations, and inhibition of Notch 4 reduces tumor formation of<br />

human breast cancer cells in xenograft models (Harrison, 2010).<br />

Myc gene amplification has also been found in about 15% of breast<br />

tumors, and 22%-35% of tumors exhibit overexpression of Myc at the<br />

transcriptional level. Transgenic mice over-expressing MYC develop<br />

invasive adenocarcinomas and preferentially harbor secondary<br />

activating mutations in KRas. We examined whether Notch receptors<br />

were activated in mouse mammary tumors over-expressing MYC and<br />

activated KRAS (KRASV12) and found Notch 4 to be significantly<br />

activated in these tumors compared to normal mammary tissue.<br />

Furthermore, Notch 4 was localized to the nucleus in the tumors<br />

over-expressing MYC and KRASV12, but not in mammary glands<br />

over-expressing MYC or activated KRASV12 alone. To examine<br />

the requirement of Notch4 in mammary tumorigenesis by MYC and<br />

KRASV12, we transplanted mammary epithelial cells isolated from<br />

Notch4 knockout mice and wild-type mice that have been transduced<br />

with MYC and KRASV12 into the mammary glands of syngeneic<br />

mice. There was a significant reduction in the onset of tumorigenesis<br />

in mammary glands that had been transplanted with mammary<br />

epithelial cells lacking Notch4. These findings suggest that Notch4<br />

is important in mammary tumorigenesis induced by cooperation of<br />

MYC and activated RAS. We are currently examining the relevance<br />

of Nocth4 activation in human breast cancers that over-express MYC<br />

and the mechanism by which Notch4 promotes tumorigenesis by<br />

Myc and activated RAS.<br />

P1-04-02<br />

The role of the mTORC1/S6K1 signaling pathway in ER-positive<br />

breast cancer.<br />

Holz MK, Unger HA, Sedletcaia A. Stern College for Women of<br />

Yeshiva University; Albert Einstein College of Medicine<br />

The mammalian target of rapamycin complex 1 (mTORC1) is a central<br />

signaling node in mediating cellular responses to mitogens, hormones<br />

and nutrients. The 40S ribosomal S6 kinase 1 (S6K1) is a conserved<br />

serine/threonine protein kinase that belongs to the AGC family of<br />

protein kinases, which also includes Akt, RSK and many others.<br />

S6K1 is the principal kinase effector downstream mTORC1. S6K1 is<br />

sensitive to a wide range of signaling inputs, including growth factors,<br />

amino acids, energy levels, and hypoxia. S6K1 relays these signals to<br />

regulate a growing list of substrates and interacting proteins in control<br />

of oncogenic processes, such as cell growth and proliferation, cell<br />

survival and apoptosis, and cell migration and invasion.<br />

Growing evidence indicates that there exists a close interaction<br />

between the mTORC1/S6K1 pathway and estrogen receptor (ER)<br />

signaling. Notably, endocrine resistance is often associated with<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

162s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 1<br />

ligand-independent activation of ERα signaling due to hyperactivation<br />

of the mTORC1 signaling pathway, and can be reversed by the<br />

mTORC1 inhibitor everolimus in vitro.<br />

Several lines of evidence suggest an important role for S6K1 in<br />

ER-positive breast cancer. We demonstrated that S6K1 directly<br />

phosphorylates ERα on Ser167, leading to activation of its<br />

transcriptional activity. While the MAPK-regulated p90 RSK also<br />

contributes to phosphorylation of this site, its contribution is early and<br />

transient, while the phosphorylation of Ser167 by S6K1 is prolonged<br />

and sustained. Intriguingly, we also found that S6K1 expression is<br />

estrogenically regulated. This regulation involves the transcription<br />

factor GATA-3 and others. Thus it appears that S6K1 expression<br />

is maintained in two modes: a basal level of expression, typical of<br />

normal cells, and an estrogen/ERα-dependent specific upregulation.<br />

Therefore, there exists a feed-forward regulatory loop whereby S6K1<br />

activates ERα transcriptional activity, leading to increased expression<br />

of S6K1. This finding is of great interest since it not only sheds light<br />

on the mode of transcriptional regulation of S6K1 expression, but<br />

may help understand the role of S6K1 overexpression and activation<br />

in the development and progression of breast cancer.<br />

Therefore, hyperactivation of mTORC1/S6K1 signaling may be<br />

closely related to ER-positive status in breast cancer, and may be<br />

utilized as a marker for prognosis and a therapeutic target.<br />

P1-04-03<br />

Knocking down Suppressor of Cytokine Signaling 7 in breast<br />

cancer: The role in Insulin-like Growth Factor - I / Phospholipase<br />

Cγ-1 signaling<br />

Sasi W, Ye L, Jiang WG, Mokbel K, Sharma A. St George’s Hospital<br />

Medical School, University of London, United Kingdom; Cardiff<br />

University School of Medicine, Cardiff, Wales, United Kingdom;<br />

The London <strong>Breast</strong> Institute, The Princess Grace Hospital, London,<br />

United Kingdom<br />

BACKGROUND:<br />

Suppressor of Cytokine Signaling 7 (SOCS7) is a member of the<br />

SOCS family and is known to interact with Phospholipase Cγ-1<br />

(PLCγ-1), one of the Insulin like Growth Factor - I (IGF-I) receptor<br />

downstream molecules. In the present study, we sought to examine the<br />

effect of knocking down SOCS7 gene on breast cancer cells growth<br />

and migration and to elucidate whether this has involved IGF-I —<br />

PLCγ-1 signaling using the PLCγ-1 blocker U73122.<br />

METHODS:<br />

Suitable breast cancer cells (MCF7 and MDA-MB-231) were<br />

transfected with anti-SOCS7 ribozymal transgene, to create sublines<br />

with SOCS7 knockdown that was verified by RT-PCR. The growth<br />

and migration of the cells were evaluated in the presence or absence of<br />

IGF-I and PLCγ-1 inhibitor using in vitro growth assay and Electrical<br />

Cell Impedance Sensing (ECIS) migration assay.<br />

RESULTS:<br />

MCF7 ∆SOCS7 and MDA-MB-231 ∆SOCS7 (SOCS7 knockdown) were<br />

constructed. Both sublines showed a higher rate of growth compared<br />

to control cells with and without IGF-I stimulation. U73122 treatment<br />

did not appear to change this growth outcome. Using ECIS migration<br />

assay, it was shown that knocking down SOCS7 had a significant<br />

positive effect on the migration of MCF7 and MDA-MB-231<br />

cells, and that both IGF-I treatment and SOCS7 knockdown had a<br />

synergistic positive influence on their migration (p < 0.05). We further<br />

demonstrated that the impact of U73122 on the IGF-I migratory<br />

effect was dependent upon SOCS7 knockdown as it has significantly<br />

blocked the stimulatory effect of IGF-I on MCF7 ∆SOCS7 and MDA-MB-<br />

231 ∆SOCS7 migration but not that of the control cells. While SOCS7 has<br />

acted to control the IGF-I effect, it appeared to cancel the inhibitory<br />

function of U73122, indicating a specific anti - PLCγ-1 role for SOCS7<br />

in IGF-I induced breast cancer cellular migration.<br />

CONCLUSION<br />

SOCS7 loss resulted in increased growth and migration of breast<br />

cancer cells and this had a synergistic effect on their response to IGF-I.<br />

This role could be related to its interaction with PLCγ-1 during the<br />

cellular migration but not the growth. It may be possible that SOCS7<br />

acts through different mechanism to control the cellular growth.<br />

P1-04-04<br />

KSR1 is involved in functional interaction between p53 and<br />

BRCA1 and is an independent predictor of overall survival in<br />

breast cancer<br />

Zhang H, Lombardo Y, Filipovic A, Periyasamy M, Coombes RC,<br />

Stebbing J, Giamas G. Imperial College London, United Kingdom<br />

Background: Our recent human kinome screening to identify new<br />

regulators of estrogen receptor α (ERα) suggested an important role<br />

of kinase suppressor of ras 1 (KSR1) in breast cancer. KSR1 was<br />

originally identified as a positive regulator interacting with both RAS<br />

and RAF in the mitogen-activated protein kinase (MAPK) pathway.<br />

Recent studies have suggested that KSR1 may have dual functions as<br />

a scaffolding protein and also a protein kinase. Although KSR1 has<br />

been implicated in Ras-dependent cancers, its clinical significance<br />

and function in breast cancer have not been elucidated.<br />

Methods: The clinical significance of KSR1 was assessed by<br />

analyzing its expression in breast cancer tissue microarrays (TMAs,<br />

n=1000) by immunohistochemistry. Pearson chi 2 and Fisher’s Exact<br />

Test were used to correlate the expression levels with various tumor<br />

biomarkers; Kaplan Meier analyses were used to investigate impact<br />

on disease-free (DFS) and overall survival (OS). Multivariate Coxproportional<br />

hazards analysis was also performed to evaluate the<br />

significant of KSR1 as an independent factor in breast cancer-specific<br />

survival (BCSS) and disease-free interval (DFI). The expression levels<br />

of KSR1 in breast cancer cell lines were measured by RT-qPCR and<br />

western blotting. A KSR1 stable over-expression breast cancer cell<br />

line (MCF7-KSR1) was generated to study its effect on growth and<br />

colony formation by 3D matrigel assay and soft agar assay in vitro.<br />

Furthermore, MCF7-KSR1 stable cells were also used in nude mice<br />

xenograft model in vivo. Luciferase reporter assay was employed to<br />

examine the effect of KSR1 on p53 transcriptional activity. Further<br />

immunofluorescence staining, western blotting and reporter assays<br />

were performed to elucidate the interaction between KSR1, BRCA1<br />

and p53.<br />

Results: KSR1 was expressed with low (60%) and high levels (40%)<br />

in breast cancer patients. High expression correlated significantly<br />

with longer DFS (p=0.014) and longer OS (p=0.012) in more than<br />

20 yrs follow-up. Interestingly, KSR1 was positively associated with<br />

BRCA1 (p=0.002) and reversely with p53 (p=0.038). High KSR1<br />

levels were a significant predictor of longer breast cancer-specific<br />

survival BCSS (p = 0.001) and longer disease free interval DFI (p =<br />

0.002) independent of tumour stage, grade and size. RT-qPCR and<br />

western blotting demonstrated that KSR1 is ubiquitously expressed<br />

in breast cancer cell lines. In vitro 3D matrigel and soft agar assay<br />

showed that MCF7-KSR1 cells had a significant decreased number<br />

of colonies and formed smaller size colonies compared to MCF7-<br />

vector cells. Furthermore, over-expression of KSR1 (MCF7-KSR1)<br />

significantly suppressed the growth of breast cancer xenografts in<br />

nude mice. Finally, KSR1 was involved in the functional interaction<br />

between BRCA1 and p53 and elevated KSR1 potentially resulted<br />

www.aacrjournals.org 163s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

in up-regulated BRCA1. KSR1 also inhibited p53-dependent<br />

transcriptional activity through suppression of p53 acetylation and<br />

promotion of p53 neddylation.<br />

Conclusion: KSR1 is an independent prognostic factor in breast<br />

cancer and high KSR1 expression correlates with longer DFS and<br />

OS. Furthermore, it is potential that KSR1 is important in tumour<br />

suppression by its involvement in functional feedback regulation of<br />

BRCA1 and p53.<br />

P1-04-05<br />

Role of the Rb and p53 Tumor Suppressor Pathways in Mammary<br />

Tumorigenesis<br />

Jones RA, Liu JC, Zhe J, Schimmer AA, Eldad Z. University Health<br />

Network, Toronto, ON, Canada<br />

The retinoblastoma (Rb) and p53 tumor suppressor pathways are<br />

frequently altered in human malignancies including breast cancer. To<br />

investigate the role of these pathways in mammary tumorigenesis, we<br />

crossed mice expressing Cre recombinase in the mammary epithelium<br />

(MMTV-Cre) with mice harboring Rb and p53 floxed alleles to<br />

generate MMTV-Cre:Rb f/f :p53 f/f mutant mice. Combined somatic loss<br />

of Rb and p53 led to the formation of aggressive triple-negative tumors<br />

(ER - , PR - , HER2 - ) that often displayed features of an epithelial to<br />

mesenchymal transition (EMT) including sarcomatoid differentiation<br />

and high expression of the mesenchymal marker N-Cadherin.<br />

Molecular profiling revealed Rb/p53 deficient tumors shared similar<br />

gene expression profiles with mouse and human claudin-low breast<br />

cancers. Interstingly, limiting dilution transplantation analysis also<br />

suggests that Rb/p53 deficient claudin-low tumors are enriched for<br />

tumor-initiating cells (TICs).<br />

To investigate the cellular origin of claudin-low tumors, the Rb f/f and<br />

p53 f/f alleles were deleted within the luminal and basal compartments<br />

of the mammary gland using a Cre-expressing adenovirus (Ad-Cre)<br />

and CD24-CD49f-based FACS analysis. The different cellular<br />

fractions were then transplanted into the cleared mammary fat pad<br />

of recipient mice which are currently being monitored for tumor<br />

development.<br />

Finally, primary cell lines isolated from Rb/p53 deficient mammary<br />

tumors were used in conjunction with high-throughput chemical and<br />

genetic screens to identify new therapeutic targets. A primary screen<br />

of 260 kinase inhibitors and 312 FDA-approved off-patent drugs has<br />

identified novel candidates and a genome wide negative selection<br />

(‘drop-out’) screen is currently in progress.<br />

Ultimately, the results of this research will provide important insight<br />

into the biology of triple-negative breast cancer and will lead to the<br />

identification of novel therapeutic targets for the treatment of patients<br />

living with this disease.<br />

P1-04-06<br />

Insertional mutagenesis identifies HACE1 as a HER2/Neu<br />

Cooperating <strong>Breast</strong> <strong>Cancer</strong> Tumor Suppressor Gene<br />

Goka E, Miller P, Baker K, Stark G, Lippman ME. University of Miami<br />

Miller School of Medicine, Miami, FL; Cleveland Clinic, Cleveland,<br />

OH; University of Miami, FL<br />

The development of human breast cancer is a multi-step process<br />

leading from normal epithelium to fully transformed breast cancer.<br />

Early genetic changes result in hyperplasias which evolves into<br />

ductal carcinoma in situ (DCIS) and culminates as invasive ductal<br />

carcinoma. The transition from DCIS to invasive disease has been<br />

implicated as the key transition in breast cancer progression. Specific<br />

genomic, transcriptomic, and proteomic alterations responsible for<br />

this transition process have yet to been elucidated.<br />

HER2/Neu, a member of the EGFR family of receptor tyrosine<br />

kinases, is associated with poor clinical outcome. HER2/Neu<br />

overexpression/amplification is commonly seen (50-60%) in noninvasive<br />

lesions but is significantly less common (20-30%) in invasive<br />

and metastatic breast carcinoma. This indicates that HER2/Neu gives<br />

normal epithelium a proliferative advantage but additional genetic<br />

alterations are required for full malignant transformation. This idea<br />

is supported by the fact that transgenic mice that overexpress HER2/<br />

Neu develop spontaneous mammary tumors but only after a prolonged<br />

latency period.<br />

Recent forward genetic approaches use insertional mutagenesis to<br />

randomly integrate a strong promoter into the genome, activating<br />

downstream gene transcription that can lead to either gain or lossof-function<br />

mutations based on the integration site. We used this<br />

insertional mutagenesis approach to induce anchorage-independent<br />

growth of isolated murine HER2/Neu over-expressing mammary<br />

epithelial cells. We identified HECT domain and ankyrin repeat<br />

containing E3 ubiquitin protein ligase 1 (HACE1) as a putative<br />

breast tumor suppressor protein whose loss leads to the formation of<br />

anchorage-independent colonies.<br />

Loss of HACE1 expression is commonly seen in breast cancer patients<br />

[as well as other tumor types] supporting the role of HACE1 as a<br />

breast tumor suppressor gene. Knockdown of HACE1 in human<br />

mammary epithelial cells (HMECs) results in the acquisition of<br />

anchorage-independent growth. Moreover, knockdown of HACE1<br />

in established breast cancer cell lines that express moderate levels<br />

of HACE1 results in enhanced cloniginicity.<br />

Therefore, we propose that the loss of HACE1 can potentially be<br />

used as a predictive marker for breast cancer progression. Additional<br />

studies investigating the mechanism of HACE1 action may also<br />

identify therapeutic targets that counteract HACE1 loss.<br />

P1-04-07<br />

The mRNA Expression of DAP1 in Human <strong>Breast</strong> <strong>Cancer</strong>:<br />

Correlation with Clinicopathological Parameters<br />

Wazir U, Jiang WG, Sharma AK, Mokbel K. St Georges’ Healthcare<br />

NHS Trust, London, United Kingdom; Cardiff University-Peking<br />

University Oncology Joint Institute, Cardiff, United Kingdom; The<br />

London <strong>Breast</strong> Institute, The Princess Grace Hospital, London,<br />

United Kingdom<br />

BACKGROUND: This pilot study is the first to focus on the potential<br />

role for death-associated protein 1 (DAP1) in human breast cancer.<br />

MATERIAL AND METHODS: mRNA expression of DAP1 in breast<br />

cancer tissues (n= 127) and normal background tissues (n=33) were<br />

determined using quantitative polymerase chain reaction (qPCR) and<br />

correlated with clinicopathological data accumulated over a 10-year<br />

follow-up period. Furthermore the effects of DAP1 knockout in breast<br />

cancer cell lines were investigated.<br />

RESULTS: The expression of DAP1 mRNA was demonstrated to<br />

decrease with increasing Nottingham Prognostic Index (NPI2 vs.<br />

NPI3, p = 0.0026), and TNM stage (TNM1 vs. 4, p=0.0039). Lower<br />

DAP1 expression levels were significantly associated with local<br />

recurrence (p=0.02) and distant metastasis (p=0.001).<br />

The knockout of the DAP1 gene in MCF-7 cell lines resulted in a<br />

significant decrease in the mRNA expression of P-21 and DELE<br />

compared with controls, p=0.04). However, the knockout of DAP1<br />

had no effect on the expression of caspase 8 and 9.<br />

CONCLUSIONS: This study demonstrates an inverse association<br />

between DAP1 mRNA levels and tumour stage and clinical<br />

outcome in breast cancer. This may be suggestive of a relationship<br />

between oncogenesis and the autophagy pathway meriting further<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

164s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 1<br />

investigation. These results also indicate that the pro-apoptotic<br />

function seems to be estrogen-dependent but independent of caspase<br />

8 and 9. The DAP1 pro-apoptotic pathway may involve P-21 and<br />

DELE proteins.<br />

P1-04-08<br />

Evidence for anti-apoptosis function of GNB1 in human breast<br />

cancer<br />

Wazir U, Kasem A, Sharma AK, Jiang W, Mokbel K. The London<br />

<strong>Breast</strong> Institute, The Princess Grace Hospital, London, United<br />

Kingdom; Cardiff University-Peking University Oncology Joint<br />

Institute, Cardiff, United Kingdom; St Georges’ Healthcare NHS<br />

Trust, London, United Kingdom<br />

INTRODUCTION: Guanine nucleotide binding protein beta<br />

polypeptide 1 (GNB1) integrates signals between receptors and<br />

effector proteins and regulates certain signal transduction receptors<br />

and effectors. We have hypothesised that GNB1 is involved in the<br />

anti-apoptosis pathway mediated by mTor. Therefore, this protein<br />

may play role in human carcinogenesis, however, there has been no<br />

investigations of this potential role published in the literature. The<br />

aim of the study was to investigate the mRNA expression of GNB1<br />

in human breast cancer and examine the relationship between its<br />

expression and the tumour characteristics and disease outcome.<br />

Furthermore, the correlation between GNB1 and mTORC1 was also<br />

investigated.<br />

METHODS: Specimens of breast cancer (BC) tissues (N=136)<br />

and normal tissues (N=30) underwent RNA extraction and reverse<br />

transcription. GNB1 transcript levels were determined using realtime<br />

quantitative PCR. Expression levels were analysed against<br />

clinicopathological data accrued over a 10 year follow-up period.<br />

RESULTS: Significantly higher mRNA transcript levels were found<br />

in the breast cancer specimens compared to normal glandular tissue<br />

in paired samples (p=0.0029). The expression of GNB1 mRNA was<br />

demonstrated to increase with increasing TNM stage (from 0.01 to<br />

15.9) and this reached statistical significance when comparing TNM1<br />

vs. TNM2/3/4 (p=0.036). Furthermore, the expression levels increased<br />

with increasing tumour grade and this reached statistical significance<br />

when comparing grade 2 vs. grade 3 (p=0.006). GNB1 expression<br />

was found to be higher in ductal tumours compared with non-ductal<br />

tumours (p=0.0081). The patients who developed recurrent disease<br />

or died from breast cancer had higher expression levels than those<br />

who had been disease-free after a median follow-up period of 10<br />

years (p=0.017). Those who died from breast cancer had significantly<br />

higher levels than those who have remained disease-free (33.9 vs.<br />

0.01, p=0.0009). GNB1 showed a significantly positive correlation<br />

with mTORC1 mRNA levels (r= 0.57, p < 0.000001).<br />

CONCLUSION: GNB1 expression was found to be significantly<br />

higher in BC specimens compared to normal breast tissue. Higher<br />

transcript levels were significantly associated with unfavourable<br />

pathological parameters and adverse clinical outcomes. These<br />

observations in addition to the positive correlation with mTORC1<br />

levels support the hypothesis that GNB1 is an important upstream<br />

component of the anti-apoptosis pathway and, therefore, can be a<br />

potential target for therapeutic intervention in human breast cancer.<br />

P1-04-09<br />

mTORC1 and Rictor expression in human breast cancer:<br />

correlations with clinicopathological parameters and disease<br />

outcome<br />

Wazir U, Kasem A, Sharma AK, Jiang WG, Mokbel K. The London<br />

<strong>Breast</strong> Institute, The Princess Grace Hospital, London, United<br />

Kingdom; Cardiff University-Peking University Oncology Joint<br />

Institute, Cardiff, United Kingdom; St Georges’ Healthcare NHS<br />

Trust, London, United Kingdom<br />

BACKGROUND:mTOR (the mammalian target of rapamycin)<br />

plays a key role in regulation of cellular metabolism, growth, and<br />

proliferation. It forms two multi-protein complexes known as<br />

complex 1 (mTORC1) and 2 (mTORC2). Raptor and Rictor are the<br />

core proteins for mTORC1 and mTORC2 respectively. There is a<br />

growing body of evidence that mTORC1 is up-regulated in many<br />

cancers and plays a role in carcinogenesis. The aim of the study was<br />

to investigate the mRNA expression of mTOR and Rictor in human<br />

breast cancer and examine the relationship between their expression<br />

and clinicopathological parameters.<br />

METHODS: Specimens of breast cancer (BC) tissues (N=150)<br />

and normal tissues (N=31) underwent RNA extraction and reverse<br />

transcription. mTOR and Rictor transcript levels were determined<br />

using real-time quantitative PCR. Expression levels were analyzed<br />

against tumor size, grade, nodal involvement, TNM stage, and clinical<br />

outcome over a 10 year follow-up period.<br />

RESULTS: Significantly higher mRNA transcript levels of mTOR<br />

were found in the breast cancer specimens compared to normal<br />

glandular tissue (p=0.0018). The expression of mTOR mRNA was<br />

demonstrated to increase with increasing NPI (53 for NPI1 to 219 for<br />

NPI3) and tumor grade (37 for grade 2 vs. 159 for grade 3, p=0.047).<br />

mTOR expression was found to be higher in ductal tumors compared<br />

with non-ductal tumors (p=0.0014). The patients who developed<br />

recurrent disease or died from breast cancer had higher expression<br />

levels than those who had been disease-free after a median followup<br />

period of 10 years (p=0.17). Higher expression levels were<br />

significantly associated with worse overall survival (p=0.01).<br />

In contrast, higher levels of Rictor mRNA expression were found<br />

in normal breast tissue, lower NPI stage (NPI1 vs. 2: p= 0.03) and<br />

lower tumor grade (grade 1 vs. grade 3: p=0.01). Patients with higher<br />

Rictor expression had a significantly better overall (p=0.037) and<br />

disease-free (p=0.048) survival.<br />

CONCLUSIONS: mTOR expression was found to be significantly<br />

higher in BC specimens compared to normal breast tissue. Higher<br />

transcript levels were significantly associated with unfavorable<br />

pathological parameters and adverse clinical outcomes. The core<br />

protein of the less predominant mTORC2 was associated with<br />

favorable pathological parameters and clinical outcome. Taken<br />

together these observations are consistent with mTORC1 role in<br />

the anti-apoptosis pathway and suggest that selective inhibitors of<br />

mTORC1 are likely to be a more effective therapeutic strategy than<br />

dual inhibitors in human breast cancer.<br />

P1-04-10<br />

PDGFRA signaling in Inflammatory breast cancer.<br />

Joglekar M, van Golen K. University of Delaware, Newark, DE<br />

Inflammatory breast cancer is the most lethal form of locally advanced<br />

breast cancer. It is clear from the recent research advancements in IBC<br />

that it is entirely a different entity, not only in its clinical presentation<br />

but also in its molecular expression profile. Therefore, it is essential<br />

www.aacrjournals.org 165s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

to carry out molecular expression studies in IBC compared to non-<br />

IBC as well as to pursue these differences to study their effects on<br />

IBC phenotype.<br />

In collaboration with the Translational <strong>Cancer</strong> Research group in<br />

Antwerp Belgium, our lab has recently found that PDGFRA is<br />

significantly over expressed in IBC patient samples (between 15-<br />

36%) compared to non-IBC patient samples. This project focuses on<br />

identifying the PDGFRA activation signature in SUM149 IBC cells.<br />

Upon ligand stimulation, PDGFR can either homo or heterodimerize<br />

(AA, AB, BB) and produce an array of signaling events responsible<br />

for cell growth, cell proliferation, cell differentiation in normal cells as<br />

well as in several cancer cells. SUM149 and SUM190 cells lack ER,<br />

PR and HER-2 expression and hence are named triple negative IBC<br />

cells. Triple negative breast cancers are highly aggressive in nature<br />

and therefore suffer from poor prognosis. We seek to determine if<br />

signaling via over expressed PDGFRA could be a potential surviving<br />

factor in producing such fast growing and highly invasive IBC tumors.<br />

Preliminary data on immunostaining of PDGFRA with and without<br />

permeabilization suggests intracellular localization of PDGFRA.<br />

We also know that SUM149s and SUM190s express all four PDGF<br />

ligands (A, B, C, D). This unusual expression of all the PDGF lignads<br />

suggests the self-sufficiency of these cells for PDGFRA signaling. In<br />

other words, these cells maintain constitutively active PDGFRA via<br />

intracrine/autocrine loop. Protein expression of active PDGFRA was<br />

studied by using two different phospho specific PDGFRA antibodies<br />

(Tyr 1018 and Tyr 849) without any prior stimulation by PDGF<br />

ligands. Moreover our data shows that PDGFR- activation in IBC,<br />

unlike in n-IBC, is PDGF-C driven.<br />

Possibility of such intracrine/autocrine loop could significantly<br />

increase the overall PDGFRA signaling in IBC cells and hence<br />

can produce fast growing aggressive IBC tumors. Thus targeting<br />

PDGFRA, an important biomarker in IBC could stand as a novel and<br />

systematic therapeutic approach.<br />

P1-04-11<br />

EZH2 Expands <strong>Breast</strong> Stem Cells via NOTCH Signaling, Acting<br />

to Accelerate <strong>Breast</strong> <strong>Cancer</strong> Initiation<br />

Kleer CG, Li X, Moore HM, Toy KA, Gonzalez ME. University of<br />

Michigan, Ann Arbor, MI<br />

Introduction: EZH2 is a critical epigenetic regulator of cellular<br />

memory and has oncogenic functions in breast cancer. EZH2<br />

upregulation signals increased risk for breast cancer when detected<br />

in benign breast biopsies. We hypothesized that EZH2 may promote<br />

breast cancer initiation through regulation of mammary stem cells.<br />

Methods: EZH2 was overexpressed in nontumorigenic breast<br />

cells using a Dox-regulated or an adenoviral system. EZH2 was<br />

downregulated in breast cancer cell lines and cells derived from<br />

fresh patients’ tumors. The effect of EZH2 levels on mammary<br />

stem cells was investigated using the mammosphere assay and flow<br />

cytometry (ALDH1+ and CD44+/CD24- assays). PCR-based stem<br />

cell microarray was used to discover EZH2 regulated genes. To<br />

investigate the effect of EZH2 on NOTCH1 transcriptional activity<br />

and promoter binding we employed reporter and ChIP assays. The<br />

effect of EZH2 in tumor initiation was investigated using xenografts<br />

and genetic mammary-specific EZH2 overexpression in MMTV-neu<br />

mice.<br />

Results: EZH2 overexpression (EZH2-OV) increased, while EZH2<br />

knockdown (KD) decreased the number of mammospheres and the<br />

percentage of ALDH1+ and CD44+/CD24- cells in nontumorigenic<br />

and in breast cancer cells, respectively. NOTCH1 was one of the<br />

most significantly downregulated genes and pathways upon EZH2<br />

KD in breast cancer cells. In nontumorigenic breast cells, EZH2-OV<br />

led to NOTCH1 mRNA and protein upregulation, and an increase in<br />

NOTCH1 transcriptional activity. We found that EZH2 binds to the<br />

NOTCH1 promoter (at -1.2kb). This function requires the HII domain<br />

of EZH2, is independent of the SET domain, and occurs in association<br />

with an increased H3K4me3 activating mark and RNA polymerase<br />

binding. Genetic mammary-specific EZH2 overexpression in MMTVneu<br />

mice led to accelerated tumor initiation (log rank p


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 1<br />

microscopy and cytokine antibody microarray respectively along<br />

with additional biochemical methods and gene expression analysis<br />

by oligo microarray.<br />

Conclusions/Significance<br />

We demonstrate here for the first time that LASP-1 translocates to<br />

the nucleus through activation of i) GPCRs -chemokine receptors<br />

CXCR2, CXCR4 and ii) Receptor tyrosine kinases - EGF receptor<br />

and HER2. Shuttling of LASP1 to the nucleus through the activation<br />

of a variety of receptors present in the tumor cells, macrophages,<br />

neutrophils and endothelial cells of the tumor microenvironment is of<br />

high significance. Silencing of LASP-1 in breast cancer cells revealed<br />

an altered profile of the 3D-clusters and the secreted cytokines.<br />

P1-05-02<br />

Epithelial-to-epithelial transition precedes collective breast<br />

cancer invasion<br />

Cheung KJ, Ewald AJ. Johns Hopkins School of Medicine, Baltimore,<br />

MD<br />

BACKGROUND: Invasion represents a fundamental step in tumor<br />

progression and is a driving force for metastasis and cancer-related<br />

mortality. Invasion has been studied largely in the context of single<br />

cells, but in vivo, most tumor cells are not found in isolation, but<br />

rather are found as ensembles of cells with substantial molecular<br />

heterogeneity. It remains poorly understood in this context which<br />

cells are invasive, how these cells arise or how these cells interact<br />

collectively to accomplish invasion.<br />

RESULTS: We developed an ex-vivo 3D culture assay enabling us<br />

to distinguish invasive from non-invasive cells and to interrogate<br />

their molecular phenotype. In two mouse models of luminal breast<br />

cancer, we find that invasive cells express markers of basal epithelium<br />

including cytokeratin K14, p63 and P-cadherin. Invasive K14+<br />

cells were also molecularly and functionally distinct from normal<br />

myoepithelial cells and co-expressed luminal K8 and E-cadherin.<br />

In vivo, K14+ cells were localized to tumor-stromal interfaces in<br />

the primary tumor and in lung micro and macro-metastases. Using<br />

a molecular biosensor and time-lapse microscopy, we observed that<br />

K14+ invasive cells were derived from K14-negative luminal tumor<br />

cells by direct phenotypic conversion. Interestingly, the capacity of<br />

luminal tumor cells to acquire basal markers varied by model, with<br />

few K14+ cells in MMTV-Her2 and many K14+ cells in MMTV-<br />

PyMT mice, correlating with the known in vivo kinetics of tumor<br />

progression in these models. To block this epithelial to epithelial<br />

transition, we tested the activity of an inhibitor of ribosome S6 kinase<br />

(RSK), which has been shown previously to inhibit myoepithelial<br />

differentiation in normal mammary epithelium. RSK inhibition caused<br />

relative depletion of K14+ tumor cells in 3D culture and abrogation<br />

of invasion. Finally, our assay revealed that in fresh primary tissue<br />

from human ER+ luminal breast cancers, invasive cells also expressed<br />

basal epithelial markers.<br />

CONCLUSIONS: Taken together, these data suggest that an epithelialto-epithelial<br />

transition precedes collective invasion in luminal breast<br />

cancers. Because we observe K14+ leader cells independent of tumor<br />

estrogen receptor status, these data may be generally applicable to<br />

breast cancer invasion across subtypes. Because many epithelial<br />

tissues have basal populations, K14+ tumor cells may also lead<br />

invasion in other cancers.<br />

P1-05-03<br />

A requirement for neural precursor cell-expressed developmentally<br />

downregulated gene 9 during the initiation of mammary<br />

tumorigenesis in MMTV-neu mice<br />

Serzhanova VA, Little JL, Izumchenko E, Seo S, Kurokawa M,<br />

Egleston B, Klein-Szanto AA, Golemis EA. Fox Chase <strong>Cancer</strong> Center,<br />

Philadelphia, PA; Ben Gurion University of the Negev, Beer Sheva,<br />

Israel; University of Tokyo, Japan<br />

The Neural precursor cell-Expressed Developmentally Downregulated<br />

gene 9 (NEDD9; also HEF1, CAS-L) scaffolding protein regulates<br />

many signaling pathways associated with mitosis, survival, migration,<br />

and ciliary integrity. Within the last few years, elevated NEDD9<br />

expression has been implicated in progression and metastasis of<br />

several types of cancer, including those of the lung, skin, and brain. We<br />

have investigated the consequences of introducing a Nedd9 genotype<br />

into the MMTV-neu mouse mammary tumor model, which in many<br />

respects recapitulates features of human HER2+ breast cancer. 80% of<br />

wild-type MMTV-Neu; Nedd9+/+ animals developed tumors with an<br />

average latency of 339 days, but only 18% of MMTV-Neu;Nedd9-/-<br />

animals developed tumors with an average latency of 416 days. This<br />

highly significant difference indicates that the Nedd9-/- genotype<br />

significantly prevents neu-dependent mammary tumor formation.<br />

HER2-positive human breast tumors and MMTV-neu-induced tumors<br />

originate from mammary luminal epithelial progenitor cells. We<br />

evaluated mammary progenitor cell populations from non-tumor<br />

bearing MMTV-neu;Nedd9-/- versus MMTV-neu;Nedd9+/+ 4-month<br />

old mice (2 months prior to appearance of tumors). Flow cytometry<br />

analysis indicated a significantly reduced CD24high;CD49flow<br />

luminal progenitor subpopulation in MMTV-neu;Nedd9-/- and<br />

Nedd9-/- mammary glands, that could be contributed to the<br />

tumorigenesis resistance in the MMTV-neu;Nedd9-/- model. The<br />

MMTV-neu;Nedd9-/-genotype negatively affected the Matrigel<br />

mammosphere colony-forming potential of luminal progenitor cells<br />

compared to MMTV-neu; Nedd9+/+cells, causing formation of<br />

fewer colonies with aberrant size and morphology. Quantification<br />

of mitotic division planes of MMTV-neu;Nedd9-/- mammospheres<br />

revealed no contribution to the observed defects. However, MMTVneu;Nedd9-/-<br />

mammospheres had defective expression of signaling<br />

proteins governing cell attachment, with reduced levels of FAK and<br />

more cytoplasmic localization of SRC.<br />

The results reported above, together with other data, support three<br />

main conclusions. First, they revealed a very substantial requirement<br />

for Nedd9 during early stages of HER2/neu-dependent tumor<br />

formation. Second, these results are the first to demonstrate a role<br />

for Nedd9 in supporting the abundance and colony-forming potential<br />

of mammary luminal progenitor cells. Third, they indicate that the<br />

defects in mammosphere growth likely involve perturbation of crucial<br />

cellular attachment signaling pathways involving FAK and SRC. In<br />

sum, these data provide a strong justification for future analysis of<br />

NEDD9 in the defective signaling of transformed mammary epithelial<br />

progenitor cell populations that initiate human breast cancer.<br />

www.aacrjournals.org 167s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P1-05-04<br />

Expression of Quiescin Sulfhydryl Oxidase 1 is associated with a<br />

highly invasive phenotype and correlates with a poor prognosis<br />

in Luminal B breast cancer.<br />

Katchman BA, Ocal T, Hostetter G, Cunliffe HE, Wantanabe A, Lake<br />

DF. Arizona State University, Tempe, AZ; Mayo Clinic Arizona,<br />

Scottsdale, AZ; Translational Genomic Research Institute, Phoenix,<br />

AZ<br />

Quiescin sulfhydryl oxidase 1 (QSOX1) oxidizes sulfhydryl groups<br />

to form disulfide bonds in proteins. Informatic analysis using the<br />

newly available “Gene Expression Based Outcome for <strong>Breast</strong> <strong>Cancer</strong><br />

Online” (GOBO) tool indicated high levels of QSOX1 expression<br />

in Estrogen Receptor positive (ER+) subtypes of breast cancer. We<br />

confirmed this finding by evaluation of QSOX1 protein expression in<br />

breast tumors and in a panel of breast cancer cell lines. In addition,<br />

Kaplan Meyer analyses revealed QSOX1 as a significant predictive<br />

marker for both relapse and poor overall survival in Luminal B tumors,<br />

but not in other intrinsic subtypes. To investigate malignant cell<br />

mechanisms in which QSOX1 might play a key role, we suppressed<br />

QSOX1 protein expression using short hairpin (sh) RNA in ER+<br />

MCF7 and ER- BT549 breast cancer cell lines. Suppression of<br />

QSOX1 protein dramatically slowed cell proliferation but did not<br />

significantly effect apoptosis or cell cycle regulation. Inhibition of<br />

QSOX1 did however dramatically inhibit MCF7 and BT549 breast<br />

tumor cells from invading through Matrigel in a modified Boyden<br />

chamber assay. Inhibition of invasion could be rescued by the<br />

exogenous addition of recombinant QSOX1. Gelatin zymography<br />

indicated that QSOX1 plays an important role in activation of MMP-9<br />

a key mediator of breast cancer invasive behavior. Taken together, our<br />

results suggest that QSOX1 is a novel biomarker for risk of relapse<br />

and poor survival in Luminal B breast cancer, and has a pro-invasive<br />

role in malignant progression through post-translational activation<br />

of MMP-9.<br />

P1-05-05<br />

Podocalyxin is a key regulator of breast cancer progression and<br />

metastasis.<br />

Snyder KA, Hughes MR, Graves M, Roskelly C, McNagny KM.<br />

University of British Columbia, Vancouver, BC, Canada<br />

Podocalyxin (gene name Podxl) is a CD34-related sialomucin<br />

that has been shown to be important in regulating cell adhesion,<br />

migration, and polarity of hematopoietic progenitors and vascular<br />

endothelia. In breast cancer, we have shown that podocalyxin is<br />

overexpressed on a subset of node-negative tumors and its expression<br />

is strongly associated with poor overall survival. To determine<br />

whether podocalyxin directly regulates breast cancer cell behavior,<br />

we ectopically expressed podocalyxin in the human breast cancer cell<br />

line, MCF7, which expresses low levels of endogenous podocalyxin,<br />

is minimally invasive, and non-metastatic. Upon ectopic expression<br />

of podocalyxin, MCF7 “bulge apically”, produce apical and lateral<br />

microvilli, are less adhesive in vitro, and are delayed in targeting<br />

integrins to the basolateral surface. In contrast to MCF7, MDA.MB-<br />

231 is a basal-like breast cancer cell-line, which expresses high levels<br />

of endogenous podocalyxin, exhibits poorly polarized monolayer<br />

and mammosphere architecture in vitro and forms “metastatic” lung<br />

tumors in vivo. Using Podxl-targeted shRNA vectors we show that<br />

expression of podocalyxin is required for MDA.MB-231 cells to form<br />

distant metastases in a competitive in vivo assay utilizing humanized<br />

NOD/SCIDIL-2rγ-/- (NSG) mice. Previously, we observed that<br />

Podxl-deficient hematopoietic cells have impaired CXCR4-mediated<br />

chemotaxis to CXCL12, a known axis for tumor metastasis. Our<br />

current hypothesis is that podocalyxin regulates chemotaxis of tumorinitiating<br />

cells (TICs) to the CXCL12-rich tissues of the lungs, liver,<br />

and bone marrow. Up-regulation of podocalyxin on breast cancer<br />

cells may be a key event in tumor progression and likely enables<br />

these cells to sense chemokines and metastasize to peripheral sites.<br />

P1-05-06<br />

A novel mutation in the tyrosine kinase domain of ErbB2:<br />

molecular and proteomic investigation of its role in breast cancer<br />

invasion<br />

O’Hara J, Kast J. University of British Columbia, Vancouver, BC,<br />

Canada<br />

•Introduction<br />

A landmark genomic study revealed 17 somatic coding mutations<br />

in a lobular metastatic breast tumor, none of which were present<br />

in the patient’s primary tumor. Among these was a novel mutation<br />

in the tyrosine kinase domain of ErbB2, a gene often amplified in<br />

breast cancer. Other mutations in this domain have been shown<br />

to stimulate transformation, growth and invasion in breast cancer<br />

cells. We hypothesize that an accumulation of mutations in the<br />

primary tumor over time preceded the development of an invasive<br />

clone, which allowed it to metastasise. Therefore, we investigated<br />

the specific effects of ErbB2 on cellular signalling and invasion, in<br />

conjunction with proteomic analyses to show its effects on global<br />

protein expression.<br />

• Methods<br />

Wild-type and mutant ErbB2 cDNAs were cloned into V180 pLP<br />

3X FLAG-tagged expression vectors. T47D breast cancer cells were<br />

transfected with one of these constructs. Protein was detected by<br />

immunoblotting with an anti-ErbB2 antibody. Membranes were then<br />

immunoblotted with an anti-phospho-ERK1/2 antibody. Transfected<br />

cells were seeded onto Matrigel; cells that migrated through the matrix<br />

were stained and counted after 48h.<br />

To identify global proteomic changes caused by mutant ErbB2, T47D<br />

cells transfected with mutant/wild-type ErbB2 or vector control,<br />

were differentially labeled by growing them in medium containing<br />

various lysine/arginine isotopes (known as stable isotope labeling of<br />

amino acids in cell culture; SILAC). Protein lysates were mixed and<br />

subjected to LC-MS/MS mass spectrometry. Proteomic changes were<br />

analysed using MaxQuant and DAVID software.<br />

• Results<br />

Overexpression of the wild-type or mutant ErbB2 genes produced<br />

protein expression in T47D, a non-invasive breast cancer cell line,<br />

confirmed by western blotting. To measure signalling downstream<br />

of ErbB2, we measured levels of the MAP kinase phospho-ERK1/2.<br />

Increased phosphorylation of ERK1/2 was demonstrated in cells<br />

expressing mutated ErbB2, compared with cells transfected with<br />

wild-type. This indicates increased activation of mutant ErbB2,<br />

demonstrating distinct phenotypic effects between wild-type and<br />

mutant ErbB2 in breast cancer cells. To analyze whether increased<br />

activation was accompanied by enhanced invasive properties, invasion<br />

assays were performed using Matrigel. A greater number of cells (1.5-<br />

fold increase) expressing mutant ErbB2 migrated across this matrix<br />

compared with wild-type-expressing cells.<br />

Global proteomic analysis of T47D cells overexpressing wild-type<br />

or mutant ErbB2 yielded 1,357 total proteins, of which 543 could<br />

be quantified. Two subsets of proteins were identified, whose levels<br />

were either increased or decreased, in the mutant versus wild-typetransfected<br />

lysates (compared to control ratios). Proteins elevated<br />

in the mutant-expressing cells included candidates involved in cell<br />

migration, inhibition of apoptosis and promotion of cellular survival.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

168s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 1<br />

Proteins showing reduced expression were involved in pathways<br />

including adhesion and DNA damage response.<br />

• Conclusion<br />

This previously undescribed mutation in the tyrosine kinase domain<br />

of ErbB2 differentially effects downstream signalling pathways and<br />

may play a role in invasion of breast cancer cells.<br />

P1-05-07<br />

Prognostic relevance of Claudin-2 expression in metastatic breast<br />

cancer<br />

Hedenfalk I, Kimbung S, Kovacs A, Skoog L, Einbeigi Z, Walz T,<br />

Malmberg M, Loman N, Fernö M, Hatschek T, TEX Study Group.<br />

Lund University, Lund, Sweden; CREATE Health Strategic Center<br />

for Translational <strong>Cancer</strong> Research, Lund University, Lund, Sweden;<br />

Sahlgrenska University Hospital, Gothenburg, Sweden; Karolinska<br />

University Hospital, Solna, Sweden; Linköping University Hospital,<br />

Linköping, Sweden; Helsingborg General Hospital, Helsingborg,<br />

Sweden<br />

The site of relapse is known to be a strong prognostic factor of<br />

survival from metastatic breast cancer. The liver is the third most<br />

frequent site affected and patients with hepatic metastases have an<br />

inferior prognosis relative to patients with bone and lung metastases.<br />

Predicting the likelihood of liver metastasis after adjuvant therapy<br />

of primary breast cancer is challenging. The aim of this study was to<br />

identify and characterize genes associated with breast cancer liver<br />

metastases. We thus performed global transcriptional profiling of<br />

120 metastatic lesions from different anatomical sites from breast<br />

cancer patients enrolled in a randomized clinical trial 1 and found<br />

that while the expression of other matrix remodelling and tightjunction<br />

molecules was down-regulated in liver metastases, Claudin-2<br />

(CLDN2) was significantly up-regulated (P=0.001). Furthermore, high<br />

expression of CLDN2 was also observed amongst patients diagnosed<br />

with a liver metastasis, irrespective of the specific site of the metastasis<br />

that was profiled, compared to patients without hepatic metastases<br />

(P=0.001). We further investigated the prognostic significance of<br />

CLDN2 protein expression by immunohistochemical staining of<br />

primary tumours and matched lymph node metastases from a larger<br />

cohort of patients (n∼200) with metastatic breast cancer. We found<br />

CLDN2 to be significantly more frequently expressed in lymph<br />

node metastases compared to primary tumours (P=0.013). Also,<br />

consistent with mRNA expression levels, more frequent expression<br />

of CLDN2 protein was observed in the primary tumours of patients<br />

who eventually developed a liver metastasis (p=0.027). Interestingly,<br />

high expression of CLDN2 in the primary tumour was associated with<br />

a shorter time to recurrence (P=0.032). Specifically, high expression<br />

of CLDN2 in the primary tumour was associated with a shorter<br />

progression to liver metastasis (P=0.002). These data highlight a<br />

potential role of CLDN2 in the development and prognosis of breast<br />

cancer liver metastases.<br />

1. Hatschek, T., et al. Individually tailored treatment with epirubicin<br />

and paclitaxel with or without capecitabine as first-line chemotherapy<br />

in metastatic breast cancer: a randomized multicenter trial. <strong>Breast</strong><br />

<strong>Cancer</strong> Res Treat. 2012, 131(3):939-47.<br />

P1-05-08<br />

Targeting integrin signaling suppresses invasive recurrence in a<br />

three-dimensional model of radiation treated ductal carcinoma<br />

in situ<br />

Nam J-M, Ahmed KM, Costes S, Zhang H, Sabe H, Shirato H, Park<br />

CC. Hokkaido University Graduate School of Medicine, Sapporo,<br />

Hokkaido, Japan; Ernest Orlando Lawrence Berkeley National<br />

Laboratory, Berkeley, CA; UCSF, <strong>San</strong> Francisco, CA<br />

Background: DCIS (ductal carcinoma in situ) is comprised<br />

of cancerous cells that are contained within the milk duct and<br />

separated from the stroma by a basement membrane. Akt activation<br />

is implicated in breast cancer progression and also upregulated in<br />

∼30% of DCIS lesions. We have previously shown that β1-integrin<br />

inhibition enhanced the efficacy of ionizing radiation (IR) in invasive<br />

breast cancer, via an Akt-mediated effect. Radiation therapy (RT) is<br />

commonly used to treat DCIS, resulting in a decrease in recurrence<br />

risk by ∼50%. However, DCIS still recurs in a substantial number<br />

of patients, and as invasive disease half of the time. This study<br />

aimed to identify the effects of IR on Akt-driven DCIS, and possible<br />

mechanisms underlying invasive progression in surviving cells.<br />

Materials and Methods: To model DCIS, we took advantage of the<br />

ability of non-invasive mammary epithelial cells, MCF10A that form<br />

duct-like structures in 3-dimensional laminin-rich extracellular matrix<br />

(3D lrECM), and overexpressed an activated myristoylated form of the<br />

serine/threonine kinase, Akt. The 3D lrECM cultured MCF10A-Akt<br />

cells show filled lumen and retained basal polarity, characteristic of<br />

DCIS lesions in vivo. After DCIS structures formed in 3D lrECM,<br />

cultures were exposed to 8 Gy IR. Confocal microscopy analysis,<br />

western blot analysis, matrigel chemoinvasion assay and matix<br />

degradation assay were performed to characterize the IR effect on<br />

luminal or basal cells of the DCIS like structures.<br />

Results: MCF10A-Akt structures post-8 Gy IR shows a significant<br />

increase in apoptosis measured by cleaved caspase-3 (n=3, P <<br />

0.05). When we propagated cells post-IR in 3D lrECM, an invasive<br />

phenotype emerged in a sub-population of survivors (n=3, P <<br />

0.01). We also confirmed that inhibitory antibodies to β1-integrin<br />

suppressed the invasive phenotype that was induced by 8 Gy IR. In<br />

addition to this, invasion activity emerged by 8 Gy IR was inhibited<br />

by nuclear factor-kappaB (NF-κB) inhibitor JSH-23 (n=3, P < 0.01).<br />

Finally, inhibition of β1-integrins or NF-κB translocation completely<br />

suppresses the formation of invasive colonies.<br />

Discussion: In present study, we found that inhibition of β1-integrins<br />

abrogates the emergence of an invasive phenotype among surviving<br />

clones. Our current data suggest that regulation of β1-integrin<br />

signaling via NF-κB plays an important role in the emergence of<br />

invasive disease after radiation, and may be an important clinical<br />

target.<br />

P1-05-09<br />

FH535 inhibited migration and growth of breast cancer cells<br />

Dorchak JA, Iida J, Clancy R, Luo C, Chen Y, Hu H, Mural RJ, Shriver<br />

CD. Windber Research Institute, Windber, PA; Walter Reed National<br />

Military Medical Center, Bethesda, MD<br />

Background: Given the lack of effective targeted therapies for<br />

triple-negative (TN) breast cancer patients, a better understanding<br />

of the mechanisms of growth and invasion of these tumors will<br />

provide valuable insight into developing novel approaches to lower<br />

the mortality associated with TN breast cancer. There is substantial<br />

evidence indicating that the canonical Wnt signaling pathway is<br />

activated in TN breast cancer tissues. Previous studies report that<br />

FH535, a small molecule inhibitor of the Wnt signaling pathway,<br />

www.aacrjournals.org 169s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

decreases growth of cancer cells but not normal fibroblast cells,<br />

suggesting this pathway plays a role in tumor progression and<br />

metastasis. In this study, we test FH535 as a potential inhibitor of<br />

migration, invasion, and growth against malignant phenotypes of<br />

breast cancer.<br />

Methods and Results: We show that FH535 significantly inhibits<br />

growth, migration, and invasion of TN breast cancer cells (MDA-<br />

MB-231 and HCC38) using established protocols, implicating<br />

the Wnt signaling pathway plays a role in tumor progression and<br />

metastasis. FH535 is a potent growth inhibitor for TN breast cancer<br />

cells (MDA-MB-231 and HCC38), but not in other breast cancer cells<br />

(MCF-7, T-47D, or SK-BR-3) when cultured in three dimensional<br />

type I collagen gel. Additionally, western blotting analysis shows<br />

that treatment of MDA-MB-231 cells with FH535 markedly<br />

inhibits the expression of NEDD9, but not FAK, Src, or p130Cas,<br />

suggesting specificity of the Wnt signaling pathway in regulating gene<br />

expressions. Co-immunoprecipitation studies suggest that NEDD9 is<br />

specifically immunoprecipitated with CSPG4, but not with β1 integrin<br />

or CD44, implying CSPG4 and NEDD9 form a molecular complex<br />

for facilitating tumor migration, invasion, and growth. Statistical<br />

analysis (ANOVA) of The <strong>Cancer</strong> Genome Atlas (TCGA) project<br />

gene expression data (https://tcga-data.nci.nih.gov/) suggests that<br />

tumors from patients with basal-type breast cancer had significantly<br />

higher CSPG4 expression than tumors from patients with other<br />

subtypes, with all the p values


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 1<br />

Material and Methods:<br />

We enrolled 40 patients diagnosed with breast cancer from Ain<br />

Shams University Hospitals. Patients were sub-grouped into negative<br />

(zero) and positive (4) LN metastasis patients. All enrolled patients<br />

had not received neo-adjuvant chemotherapy. During modified<br />

radical mastectomy or conservative breast surgery, 15-20 ml of<br />

blood were collected from axillary vein tributaries for the isolation<br />

of tumor-associated leukocytes by Ficoll-Hypaque density gradient<br />

centrifugation. The immunophenotype of the isolated mononuclear<br />

cells was assessed using flow cytometry to identify the percentage of<br />

T lymphocytes (CD3+) and their subpopulations (CD4+ and CD8+),<br />

NK cells (CD56+) and B-lymphocytes (CD19+). Tumor-associated<br />

leukocytes isolated from positive and negative LN patients were<br />

seeded overnight in appropriate culture media at 37°C in a humidified<br />

CO2 incubator. Conditioned media containing secreted cytokines/<br />

chemokines were profiled using RayBio human cytokine antibody<br />

arrayIIIand then analyzed and quantified using ImageJ software. In<br />

addition, using immunoblottingwe assessed the level of expression<br />

of total and phospho NF-kβ in leukocyte lysates from positive and<br />

negative LN patients.<br />

Result:<br />

Flow cytometric analysis revealed a significant increase in the<br />

percentage of T lymphocytes and T helper(CD3+/CD4+) cells<br />

collected from axillary tributaries of positive LN patients as<br />

compared to negative LN patients. Leukocytesisolated from positive<br />

LN patients were characterized by more than five-foldincreases in<br />

IL-12,Thrombopoietin, IL-13, INF-ϒ, TARC, IGF-1, IL-3, TNF-α,<br />

IL-4, GCSF, IL-5 and IL-1α. Moreover, we detected an increase in<br />

cellular expression of total and phospho NF-kβ in leukocytes isolated<br />

from positive LN as compared to negative LN breast cancer patients.<br />

Conclusion: Our results demonstrate for the first time that tumorassociated<br />

leukocytes of positive LN patients possess different<br />

cytokine patterns than those ofnegative LN patients. Moreover, we<br />

identified major cytokines that may contribute to LN metastasis and<br />

could be therapeutically targeted.<br />

P1-05-12<br />

Metastatic xenograft models of human estrogen receptor negative<br />

breast cancer primary cultures are driven by the recruitment of<br />

myeloid-derived suppressor cells<br />

Drews-Elger K, Iorns E, Brinkman JA, Berry DL, Lippman ME,<br />

El-Ashry D. Sylvester Comprehensive <strong>Cancer</strong> Center, Braman<br />

Family <strong>Breast</strong> <strong>Cancer</strong> Institute, University of Miami, FL; Science<br />

Exchange, Palo Alto, CA; Georgetown University Medical Center,<br />

Washington, DC<br />

The study of early stage and metastatic breast cancer relies heavily<br />

on the use of established cell lines often derived from metastatic<br />

lesions rather than from primary tumors, limiting our understanding<br />

of primary tumor development. Study of the mechanisms governing<br />

the metastatic process in its entirety is not always feasible with<br />

these types of cellular and xenograft models, as often the primary<br />

tumor needs to be excised, or breast cancer cells are introduced into<br />

circulation of host mice. While this approach has allowed a better<br />

understanding of metastasis, it may unintentionally enhance the<br />

colonization efficiency and pass over important events occurring in<br />

the tumor microenvironment. In order to provide a cellular model that<br />

more closely recapitulates breast cancer development and progression,<br />

we generated breast cancer cellular cultures by dissociating specimens<br />

of human estrogen receptor-negative primary breast tumors and<br />

placing them in culture. Within our group of dissociated tumor (DT)<br />

cell cultures, we had shown that five cultures are formed by breast<br />

cancer cells and these were classified by PAM50 predictor analysis as<br />

belonging to the ER-/PR-/Her2+ and triple negative subtypes. DT cells<br />

are tumorigenic in NOD/Scid and NOD/Scid gamma (NSG) mice.<br />

Our first aim was to assess the metastatic potential of the DT cells in<br />

two mouse models. We show that 3 of the 5 cultures are metastatic to<br />

lymph nodes, liver and or lungs; common sites of metastasis in breast<br />

cancer patients. Spontaneous metastases were observed without the<br />

requirement of primary tumor excision. The metastatic frequency<br />

ranged from 25-90% in NOD/Scid mice, and from 80-100% in NSG<br />

mice. Moreover, expression of key proteins such as epidermal growth<br />

factor receptor (EGFR) detected in primary tumors was also found<br />

in the metastatic lesions. Because the DT cells have been isolated<br />

from primary breast tumors they represent a clinically relevant and<br />

valuable model to study breast cancer and metastasis.<br />

A critical factor influencing tumor progression and metastasis is the<br />

infiltration of immune cells including myeloid-derived suppressor<br />

cells (MDSCs), which have been shown to be increased in a tumordependent<br />

manner in xenograft models of breast cancer. Previous<br />

work from our lab has shown that stroma of tumor-bearing mice<br />

exhibited gene expression changes, including increased expression<br />

of calcium binding proteins S100A8 and S100A9, that are consistent<br />

with myeloid immune cell infiltration. Our second aim was thus to<br />

analyze the recruitment of S100A8+ MDSCs by xenografted DT<br />

tumors. We show that the recruitment of S100A8+ MDSCs is detected<br />

in DT tumors and sites of metastasis, and more importantly, their<br />

presence was significantly greater in the tumors that metastasized<br />

compared to those that did not. These data strongly suggest that the<br />

metastatic potential of breast cancer cells may be driven at least in<br />

part by the recruitment of S100A8+ MDSCs. Taken together, our<br />

observations and our closer-to-primary xenograft models may lead<br />

to a better understanding of metastatic disease, as well as to the<br />

development and pre-clinical screening of targeted breast cancer<br />

therapeutics, particularly in the metastatic setting.<br />

P1-05-13<br />

CLIC3 is associated with invasive behaviour and poorer prognosis<br />

in estrogen receptor-negative breast cancer.<br />

Macpherson IR, Dozynkiewicz M, Kalna G, Speirs C, Chaudhary<br />

S, Edwards J, Timpson P, Norman J. University of Glasgow, United<br />

Kingdom; Beatson Institute for <strong>Cancer</strong> Research, Glasgow, United<br />

Kingdom<br />

Background: We previously described a Chloride Intracellular<br />

Channel-3 (CLIC3)-dependent recycling pathway which trafficked<br />

active integrins from late endosomes to the cell surface and which was<br />

required for the migratory and invasive behaviour of A2780 ovarian<br />

cancer cells in vitro. We also demonstrated that elevated expression<br />

of CLIC3 was associated with poor prognosis in pancreatic cancer.<br />

We therefore set out to investigate the role of CLIC3 in breast cancer.<br />

Materials and Methods: CLIC3 expression was assessed by<br />

immunohistochemistry and the weighted histoscore method in a tissue<br />

microarray (TMA) consisting of triplicate cores from 141 patients<br />

diagnosed with invasive estrogen receptor (ER)-negative early<br />

breast carcinoma between 1995 and 1998. Full clinicopathological<br />

and follow-up data were available. Further data were obtained from<br />

publicly available gene expression datasets (Desmedt, GSE7390)<br />

and Oncomine TM . Knockdown of CLIC3 in the ER-ve MDA-MB231<br />

breast cancer cell line was achieved by nucleofection of 2 different<br />

siRNA sequences (Dharmacon). Inverse invasion assays measured<br />

invasion into a plug of matrigel supplemented with fibronectin over<br />

72 hours whilst organotypic invasion assays measured invasion into<br />

a fibroblast-containing collagen matrix over 6 days.<br />

www.aacrjournals.org 171s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Results: CLIC3 mRNA expression was significantly elevated in breast<br />

cancer in comparison to normal breast tissue in two independent data<br />

sets. High CLIC3 protein levels were associated with significantly<br />

shorter breast cancer specific survival (p=0.026) in a TMA of 141 ERve<br />

patients. This finding was corroborated by the observation that high<br />

CLIC3 mRNA levels were associated with shorter overall survival in<br />

198 patients in the Desmedt dataset (p=0.038). Transient silencing of<br />

CLIC3 expression using two independent siRNA sequences had no<br />

effect on proliferation of MDA-MB231 cells but significantly reduced<br />

their invasiveness by 46% and 93% respectively in an inverse invasion<br />

assay and by 36% and 42% respectively in an organotypic invasion<br />

assay. CLIC3 was found to co-localise with Rab7 and LAMP1 in late<br />

endosomes/lysosomes in MDA-MB231 cells.<br />

Conclusions: Our clinical and in vitro data indicate an important<br />

role for CLIC3 in the invasive and metastatic behaviour of some<br />

breast cancers. Further work to elucidate molecular mechanisms is<br />

underway and will be presented.<br />

P1-05-14<br />

Multiscale computational modeling of breast cancer invasion:<br />

Towards a predictive patient-based tool<br />

Trucu D, Thompson A, Chaplain MAJ. University of Dundee, United<br />

Kingdom; Ninewells Hospital, University of Dundee, United Kingdom<br />

Background<br />

Understanding the mechanisms of the growth and spread of cancer<br />

cells in the human breast and providing effective treatment continues<br />

to remain a significant challenge. <strong>Cancer</strong> cell invasion of breast<br />

tissue plays a pivotal role in the metastatic cascade through complex,<br />

coupled dynamics of the constituent processes occurring over a range<br />

of spatial and temporal scales, ranging from genetic events through<br />

intracellular signaling pathways through cell-matrix interactions to<br />

tissue scale changes.<br />

Method<br />

While most previous computational modeling has been carried out<br />

at the level of a single spatial or temporal scale, the work presented<br />

here has developed a novel multiscale modeling and computational<br />

simulation framework. Formulated in both two and three spatial<br />

dimensions, this approach is based on a new type of multiscale<br />

moving boundary computational model that explores the crucial role<br />

that matrix degrading enzymes, secreted by individual cancer cells<br />

(micro-scale) along the invasive edge of the tumour, play in the overall<br />

dynamics of the tissue-scale invasion (macro-scale).<br />

Results<br />

The computational simulation results we have obtained reveal a rich<br />

variety of cancer invasion patterns that emerge in the vicinity of a<br />

primary breast cancer. In the presence of heterogeneous tissue, our<br />

computational simulation results show well-defined “lobular” and<br />

“fingering” invasion patterns, consistent with the spread of a breast<br />

carcinoma, and which remain robust with respect to any changes made<br />

in the model parameters. These patterns are found to be compatible<br />

with respect to both the dynamic heterogeneity of cancer cell macroscale<br />

distributions and to the evolution of the heterogeneity of the<br />

surrounding breast tissue.<br />

Conclusions<br />

By exploring the dynamics of the molecular processes of matrix<br />

degrading enzymes and of tissue-scale spatio-temporal cancer tissue<br />

evolution, this novel multiscale computational modeling framework<br />

faithfully replicates the cancer invasion process. Development of this<br />

computational tool should allow pre-operative breast cancer patient<br />

data to be used to predict the extent of breast cancer invasion and<br />

hence the extent of surgical resection.<br />

P1-05-15<br />

Identification of a novel Hypoxia Inducible Factor-1 regulated<br />

gene involved in breast cancer growth and metastasis<br />

Peacock DL, Schwab LP, Seagroves TN. University of Tennessee<br />

Health Science Center, Memphis, TN<br />

Introduction: Hypoxia is a hallmark of most solid tumors. In response<br />

to hypoxic stress tumor cells adapt by regulating survival, metabolism<br />

and angiogenesis. The heterodimeric Hypoxia-Inducible Factor<br />

(HIF)-1 transcription factor is a master regulator of this response. HIF-<br />

1alpha protein is over-expressed in ∼30% of primary breast tumors<br />

and ∼70% of metastases, which independently correlates with poor<br />

prognosis and decreased survival in patients. Moreover, conditional<br />

deletion of Hif1a in the mammary epithelium of the MMTV-polyoma<br />

middle T (MMTV-PyMT) transgenic model of breast cancer<br />

significantly delays mammary tumor initiation and lung metastasis<br />

burden. Methods: Our lab has established primary mammary tumor<br />

epithelial cells (MTECs) from late stage carcinomas originating in<br />

PyMT+; Hif1α floxed mice (FVB/Nj strain, F11). These MTECs<br />

were exposed to either Adenovirus-eta-βgal or -Cre to create wildtype<br />

(WT) and knock-out (KO) cells, respectively. PyMT MTECs<br />

are transplantable into syngeneic FVB/Nj female recipients. Deletion<br />

of HIF-1 activity reduced primary tumor growth by ∼60% and the<br />

formation of lung macrometastases originating from mammary fat<br />

pad tumors by >90%. Microarray profiling was conducted to identify<br />

genes differentially expressed between WT and KO cells cultured<br />

at normoxia (21% O2) or hypoxia (0.5% O2) as well as between<br />

end-stage WT and KO tumors derived from the parental cell lines.<br />

Several genes were down-regulated in both gene sets in response to<br />

deletion of Hif1a. One mRNA of particular interest, creatine kinase<br />

brain isoform (Ckb) was down-regulated in KO cells >100 fold and >2<br />

fold in end-stage tumors. Pools of stable Ckb knockdown (KD) PyMT<br />

cells were generated via lentiviral transduction of two independent<br />

shRNA constructs (pLKO.1 shc59 or shc61, each producing >75%<br />

knockdown) to determine the effects of Ckb knockdown on cell<br />

proliferation, migration and metastasis. Results: No differences in<br />

cell proliferation were observed between WT and Ckb KD cells when<br />

cultured in standard culture conditions. In contrast, when cells were<br />

introduced to the mammary fat pad, initiation of palpable tumors<br />

was delayed by >30 days for each shRNA KD line, suggesting that<br />

CKB function may be modulated by the tumor microenvironment. A<br />

significant decrease in invasion through Matrigel was also observed<br />

for each shRNA Ckb cell line. Likewise, when cells were introduced<br />

into circulation via tail vein injection, 20% of mice injected with either<br />

shc59 or shc61 cells developed lung metastases whereas 100% of mice<br />

injected with WT cells developed metastases. Bioinformatic analyses<br />

indicate that multiple putative HIF-1 binding sites (HREs) are present<br />

in both the mouse and human CKB gene promoters; chromatin<br />

immunoprecipiation (ChIP) experiments are in progress to validate<br />

that CKB is a direct HIF-1 target gene. Conclusions: HIF-1alpha<br />

strongly promotes metastasis, the major cause of mortality in breast<br />

cancer patients. Further characterization of genes downstream of<br />

HIF-1alpha, such as CKB, that play a key role in driving invasion and<br />

tumor initiation may identify new pathways amenable to therapeutic<br />

intervention for patients with metastatic breast cancer.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

172s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 1<br />

P1-05-16<br />

Sushi Domain Containing 2 (SUSD2): a plasma membrane protein<br />

that increases immune evasion in breast tumorigenesis<br />

Watson AP, Davis EM, Egland KA. <strong>San</strong>ford Research/USD, Sioux<br />

Falls, SD<br />

Routinely used therapies are not adequate to treat the heterogeneity<br />

of breast cancer, and consequently, more therapeutic targets are<br />

desperately needed. To identify novel targets, we generated a breast<br />

cancer cDNA library enriched for breast cancer genes that encode<br />

membrane and secreted proteins. From this library we identified<br />

SUSD2 (Sushi Domain Containing 2), which encodes an 822 amino<br />

acid protein containing a transmembrane domain and functional<br />

domains inherent to adhesion molecules. Previous studies describe<br />

the mouse homolog, mSVS-1, but there are no studies on the human<br />

gene associated with breast cancer.<br />

Reverse-Transcriptase PCR analysis using total RNA from frozen<br />

breast cancer samples showed that SUSD2 is expressed in 19 of 24<br />

breast tumors. Immunohistochemistry (IHC) analysis was performed<br />

on human breast tissues using an anti-SUSD2 antibody. IHC showed<br />

weak or no expression of SUSD2 in normal epithelial cells, with the<br />

endothelial lining of vessels staining positive for SUSD2. However,<br />

staining was observed in pathological breast lesions and in lobular<br />

and ductal carcinomas.<br />

To explore the role of SUSD2 in breast tumorigenesis, we performed<br />

in vitro analyses of stable cell lines generated to over-express SUSD2.<br />

Our studies indicate that over-expression of SUSD2 increases the<br />

invasion of breast cancer cells through Matrigel and adhesion to<br />

fibronectin, an extracellular matrix protein. Additionally, contact<br />

between peripheral blood mononuclear cells (PBMCs) and breast<br />

cancer cells that express SUSD2 leads to diminished secretion<br />

of chemotactic cytokines and increased secretion of angiogenic<br />

cytokines compared to the empty-vector control. A similar co-culture<br />

experiment with Jurkat T cells showed increased induction of T cell<br />

apoptosis when cultured with cancer cells expressing SUSD2.<br />

Using an in vivo model with immunocompetent mice, we observed<br />

increased tumor growth and decreased survival in mice with<br />

mammary tumors expressing mSVS-1. Interestingly, increased tumor<br />

growth rate was not due to higher proliferative rates of the cells, as<br />

the Ki67 index was similar for all tumors. IHC staining with an anti-<br />

CD31 antibody revealed disordered and tortuous microvasculature<br />

in tumors expressing mSVS-1. We also characterized the population<br />

of lymphocytes in the blood, spleen and tumors of the mice as a<br />

measure of the immune response. While proportions of CD4 and<br />

CD8 cells were unchanged in the spleen and blood samples, we found<br />

significantly fewer CD4 tumor infiltrating lymphocytes in mice with<br />

tumors expressing mSVS-1.<br />

SUSD2 interacts with Galectin-1 (Gal-1), a 14-kDa secreted protein<br />

that is synthesized by carcinoma cells and promotes tumor immune<br />

evasion, angiogenesis and metastasis. Several previous studies suggest<br />

that cellular localization of Gal-1 is essential for its multiple roles in<br />

tumorigenesis. Interestingly, cell surface localization of Gal-1 was<br />

observed only in cell lines expressing SUSD2 but not the empty-vector<br />

control. Because the localization of Gal-1 on the surface of cells is<br />

dependent on the presence of SUSD2, using SUSD2 as a therapeutic<br />

target may disrupt this interaction and inhibit events required for<br />

tumorigenesis.<br />

P1-05-17<br />

S100A7/RAGE axis enhances breast cancer growth by activating<br />

Stat3 signaling<br />

Nasser MW, Wani NA, Qamri Z, Ahirwar D, Powell CA, Ganju RK.<br />

The Ohio State University, Columbus, OH<br />

S100A7 has been shown to be highly expressed in high grade ductal<br />

carcinoma in situ and invasive breast cancers. Its expression is also<br />

related to poor prognosis and associated with increased inflammatory<br />

infiltrates and various inflammatory disorders. However, the exact<br />

mechanism by which S100A7 enhances breast cancer growth and<br />

metastasis is not known. In the present study, we determined the<br />

molecular mechanisms by which S100A7 enhances growth and<br />

metastasis of breast cancer. Recently, we have shown that S100A7<br />

may mediate signaling by binding to receptor for advanced glycation<br />

end-products (RAGE). In this study, we first observed high expression<br />

of RAGE in ER-negative MDA-MB-453, MDA-MB-231 and highly<br />

metastatic SCP2 breast cancer cell lines by FACS analysis. We have<br />

also shown that S100A7 is secreted in the conditioned media of<br />

S100A7 overexpressing MDA-MB-231 cells. Recombinant soluble<br />

S100A7 was shown to induce cell migration and wound healing<br />

in MDA-MB-453, MDA-MB-231 and SCP2 cells. These effects<br />

were mediated through RAGE, as RAGE neutralizing antibody<br />

blocked S100A7-mediated functional effects. To further analyze<br />

the molecular mechanisms of S100A7-mediated signaling, we<br />

observed that recombinant S100A7 enhanced the activation of Stat3<br />

in MDA-MB-231 and SCP2 cell lines. Further analysis of molecular<br />

mechanisms revealed that S100A7 enhances ERK phosphorylation<br />

and Stat3 downstream target cyclinD1. These effects were attenuated<br />

by the treatment of soluble RAGE. We further analyzed the role of<br />

murine S100A7 (mS100a7a15) on Stat3 activation in in vivo inducible<br />

bi-transgenic MMTV-mS100a7a15 mice. These mice treated with<br />

Stat3 inhibitor showed reduced mS100a7a15-induced mammary<br />

hyperplasia and Stat3 activation in the mammary gland. We also<br />

observed that mS100a7a15 induction enhanced proliferation of<br />

mammary epithelial cells (MEC) isolated from bi-transgenic mice<br />

and this effect was attenuated by Stat3 inhibitor. Taken together,<br />

these studies reveal that S100A7 may enhance breast tumor growth<br />

and metastasis by binding to RAGE and activating Stat3 signaling.<br />

These studies may help in the development of S100A7/RAGE/Stat3<br />

as novel therapeutic targets for ER negative, especially triple-negative<br />

and basal-like cancers, which are highly aggressive and have currently<br />

been shown to enhance the Stat3 signaling pathway.<br />

P1-05-18<br />

Determining the molecular mechanism of the breast cancerinduced<br />

brain metastasis and a role of a novel pan-TGF-β<br />

inhibitor as a potential therapy for brain metastasis in a mouse<br />

xenograft model<br />

De Mukhopadhyay K, Elkahloun AG, Hinck AP, Yoon K, Cornell JE,<br />

Shu L, Yang J, Sun L. ; University of Texas Health Science Center,<br />

<strong>San</strong> <strong>Antonio</strong>, TX; National Human Genome Research Institute-NIH,<br />

Bethesda, MD<br />

<strong>Breast</strong> cancer (BCa) is the most common malignant disease in women<br />

in the U.S. Nearly 20% of patients with advanced BCa are eventually<br />

diagnosed with brain lesions, which is a devastating complication in<br />

patients with BCa over-expressing EGF receptor family members<br />

including Her2 positive and triple negative breast cancer. It is the<br />

most feared complication of BCa in part because are not capable of<br />

significantly treating the BCa-induced brain metastases due to the<br />

inability of the available treatment regimens to effectively penetrate<br />

the blood brain barrier (BBB) and also due to our limited knowledge<br />

www.aacrjournals.org 173s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

on cellular and molecular mechanisms that drive the homing to<br />

and growth in the brain of BCa cells. Therefore, there is a need of<br />

efficient model system that can significantly contribute towards<br />

our understanding of different factors from both host and tumor<br />

leading to brain metastasis. We have recently isolated a novel BCa<br />

cell line B6TC that was generated through fusion between human<br />

BCa, MDA-MB-231 and ZR-75-1 cells in mouse bone marrow.<br />

This B6TC cell line showed higher propensity to metastasize to<br />

brain than its parental cells when inoculated through intracardiac<br />

injection in female athymic nude mice. In order to generate a highly<br />

brain metastatic breast cancer model for mechanistic research, we<br />

subjected the B6TC cells through four rounds of selection for cells<br />

that were capable of trans-endothelial cell invasion to obtain cells<br />

that could invade through BBB. This in vitro selected cell line was<br />

further subjected through three rounds of in vivo selection for cells<br />

that were capable of metastasizing to the brain and the cells after third<br />

round selection was named N3LR, which has the highest potential<br />

to cause brain metastasis. In searching for genes and pathways that<br />

may contribute to the increased brain metastasis of N3-LR cell with<br />

microarray analysis, we found that the transforming growth factorbeta<br />

(TGFβ) signaling pathway is upregulated in N3-LR cell in<br />

comparison with B6TC cell, in addition to the EGF and prostaglandin<br />

signaling pathways that have been reported to be associated with brain<br />

metastatic breast cancer cells. Functional comparison also showed that<br />

N3-LR cell was more migratory than B6TC cell and more responsive<br />

to TGFβ-induced phosphorylation of Smad3 as well as migration,<br />

suggesting that TGFβ signaling may contribute to the increased<br />

brain metastatic potential. We next investigated whether metastatic<br />

tumor growth in the brain microenvironment can be inhibited by<br />

systemic administration of a potent pan-TGFβ inhibitor, BG E RIIa<br />

recombinant fusion protein containing the endoglin domain of<br />

betaglycan (BGE) and the extracellular domain of RII. The animals<br />

were inoculated intracardically with N3LR, the most potent subline<br />

of highly metastatic B6TC cells, and were then treated with vehicle<br />

or BG E RII systemically via i.p. injection right after the inoculation.<br />

After three weeks, the BG E RII treated group showed lower brain<br />

metastasis incidence and tumor burden as detected by whole mouse<br />

bioluminescence and GFP imaging. Further analyses to understand the<br />

underlying molecular and regulatory mechanism of brain metastasis<br />

and its intervention in our mouse model is underway for the discovery<br />

of novel molecularly targeted drugs to prevent and eradicate BCa<br />

metastasis initiation, progression and recurrence.<br />

P1-05-19<br />

Modeling <strong>Cancer</strong> Recurrence and the Therapeutic Effect of<br />

Adjuvant Systemic Therapy<br />

Ganesan S, Bhanot G, Boemo M, Lee JH. <strong>Cancer</strong> Institute of New<br />

Jersey- UMDNJ, New Brunswick, NJ; Rutgers University, Piscataway,<br />

NJ<br />

Despite complete surgical removal of early stage cancers, recurrence<br />

can occur at distant locations in time spans ranging from months to<br />

decades. This is due to the presence of disseminated cancer cells that<br />

can ultimately transition into active growth leading to recurrence.<br />

Systemic adjuvant therapy can decrease the incidence of distant<br />

relapse and lead to improved survival in some cancers. In this study,<br />

data on natural recurrence patterns of different breast cancer subtypes<br />

is used to build a probabilistic model of distant recurrence. The central<br />

hypothesis of this model is that after initial local therapy, a fraction of<br />

patients will have the presence of one or more disseminated dormant<br />

metastatic foci (DMF). The transition of DMF into actively growing<br />

metastatic foci (AMF) is described using a stochastic model. DMF<br />

of individual breast cancer subclasses each have a distinct probability<br />

per unit time of entering active growth. Fitting of this model to<br />

clinical trial data of early stage breast cancers treated with surgery<br />

alone demonstrates that DMF present in estrogen receptor negative<br />

(ER-) cancers and Luminal B cancer have greater probabilities<br />

of transition to active growth per unit time than Luminal A breast<br />

cancers. Clinical trial data were then used to determine the effect<br />

of adjuvant therapy on the dynamics of tumor recurrence. The data<br />

strongly support a model in which adjuvant chemotherapy works by<br />

eliminating only a fraction of DMF that have transitioned into AMF<br />

during the time of chemotherapy treatment. Thus short duration<br />

adjuvant chemotherapy would have greater efficacy in ER-, HER+ and<br />

Luminal B breast cancers, which have fast dynamics of recurrence,<br />

and little effect in Luminal A breast cancers with far slower dynamics<br />

of recurrence. Similarly, analysis of adjuvant hormonal therapy trial<br />

data, demonstrate that hormonal therapy also affects only DMF that<br />

transition into active growth during the period of hormonal therapy.<br />

This model can explain why extended duration therapy of up to 5<br />

years is more effective than shorter duration of adjuvant hormonal<br />

therapy. Interestingly some adjuvant treatments, such as trastuzumab<br />

for HER2+ breast cancers, affect recurrence rates in a way that<br />

suggest that these interventions directly kill DMF. This model of<br />

tumor recurrence has several clinical implications that could inform<br />

future clinical trial design.<br />

P1-05-20<br />

Fluorescent Hyaluronan Probes Distinguish Heterogeneous<br />

<strong>Breast</strong> <strong>Cancer</strong> Cell Subsets and Predict their Invasive Behavior<br />

Veiseh M, Kwon DH, Borowsky AD, Toelg C, Leong H, Lewis J, Turley<br />

EA, Bissell MJ. Lawrence Berkeley National Laboratories, Berkeley,<br />

CA; University of California Davis, Davis, CA; London Health<br />

Sciences Centre-London Regional <strong>Cancer</strong> Program; University of<br />

Western Ontario, London, ON, Canada<br />

Background: Tumor heterogeneity is a determining factor in diagnosis<br />

and therapy of breast cancer (Bca). We investigated Bca cellular<br />

heterogeneity by a functional fluorescent hyaluronan (HA) probe, and<br />

monitored its binding characteristics in relation to the degree of Bca<br />

aggression. HA is a polysaccharide ligand for CD44 and RHAMM<br />

receptors, which are linked to cell plasticity and predict poor clinical<br />

outcome in Bca. Methods: Fluorescent HA probes, multiplexed<br />

profiling, and cell sorting strategies were developed and used to detect/<br />

capture tumor cell subpopulations with differences in HA binding<br />

(HA -/low vs. HA high ) and HA receptors surface display. Whereas we<br />

concentrated on the most metastatic cell line, MDA-MB-231, we<br />

also monitored the presence of subpopulations in other Bca lines<br />

and showed heterogeneity in all. Results: The HA probe binding was<br />

highly heterogeneous, and the highest binding level was detected in<br />

highly invasive triple-negative basal subtypes such as MDA-MB-231.<br />

HA probes bound to the cell surface but also accumulated in cytoplasm<br />

and nucleus. Binding levels were dramatically reduced upon<br />

‘reversion’ of highly malignant cells to a non-malignant phenotype<br />

in three-dimensional cultures, suggesting that the level of cellular<br />

binding to fluorescent HA probe provides a measure of malignant<br />

behavior. Comparison between HA high and HA -/low subpopulations<br />

revealed surprising differences in morphology, proliferation and<br />

invasion in culture, which were retained in two in vivo models. HA high<br />

subpopulations exhibited higher levels of invasion but surprisingly<br />

lower levels of proliferation compared to either unsorted parental cells<br />

or the HA -/low subpopulation. Conclusions: Querying HA binding in<br />

Bca lines reveals an unexpected heterogeneity with respect to tumor<br />

phenotype in vivo, morphology in 3D, invasion and proliferation.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

174s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 1<br />

These results may aid in diagnosis and therapy of invasive Bca<br />

subpopulations and early sorting of cancer patients needing intensive<br />

chemotherapy.<br />

Keywords: Heterogeneity, 3D cultures, Hyaluronan, CD44,<br />

RHAMM/HMMR, Tumor phenotype, <strong>Breast</strong> cancer, Invasion.<br />

P1-05-21<br />

The role of epsin in promoting Epithelial-Mesenchymal Transition<br />

and metastasis by activating NF-kB signaling in breast cancer<br />

Cai X, Brophy ML, Hahn S, McManus J, Chang B, Pasula S, Chen<br />

H. Oklahoma Medical Research Foundation, Oklahoma City, OK;<br />

University of Oklahoma Health Science Center, Oklahoma City, OK<br />

Background: <strong>Breast</strong> <strong>Cancer</strong> progression and metastasis are multistep<br />

processes that involve local tumor growth and invasion followed<br />

by tumor dissemination to and re-establishment at distant sites.<br />

The ability of a tumor to metastasize is the major determinant of<br />

the mortality of cancer patients. Thus, elucidating the molecular<br />

pathways essential for tumor metastasis is of high priority in cancer<br />

biology. We have previously demonstrated that deficiency of epsins,<br />

a family of endocytic clathrin adaptor proteins, decreases tumor<br />

growth by enhancing VEGF signaling in vascular endothelial cells and<br />

subsequently promoting dysfunctional tumor angiogenesis. Research<br />

Objective: Our immediate aim in the current research is to determine<br />

the role of epsin in regulating breast cancer growth and metastasis<br />

by activating NF-κB signaling. Our ultimate goal is to seek better<br />

treatment for breast cancer patients. Rationale: Our preliminary data<br />

demonstrate that epsins are overexpressed in human cancers, including<br />

breast cancer. Knockdown of epsins in human breast cancer cell line<br />

MDA-MB-231 inhibits in vitro cell proliferation and migration.<br />

Xenograft or tail-vein injection of epsin-deficient breast cancer cells<br />

reveals marked reduction in in vivo tumorigenesis and lung metastasis.<br />

Moreover, E-cadherin is increased and vimentin decreased in epsindeficient<br />

MDA-MB-231 and in tumors derived from epsin-deficient<br />

MDA-MB-231 tumor models. Conversely, overexpression of epsin<br />

in breast epithelial cell line MCF10A and MDA-MB-231 results in<br />

decreased E-cadherin and increased vimentin expression. Hypothesis:<br />

We hypothesize that epsin regulates EMT through modulating NFκB<br />

signaling. Methods: we use combined techniques of western<br />

blot, confocal immunofluorescence and RT-PCR to examine NF-κB<br />

activation. Results: NF-κB phosphorylation, nuclear translocation<br />

and NF-κB target gene snail/slug expression is downregulated in<br />

epsin-deficient MDA-MB-231. Epsin overexpression in MDA-<br />

MB-231 increases NF-κB phosphorylation. Mechanistically, we show<br />

that epsin co-immunoprecipitates with components of TNF Receptor<br />

Signaling Complex (TNFR-SC) including TNFR1, TRADD, RIP,<br />

TRAF2 and NEMO in MDA-MB-231, which can be enhanced by<br />

TNFα stimulation. Conversely, recruitment of NEMO and TRAF2<br />

to TNFR1 is impaired in epsin-deficient MDA-MB-231 compared to<br />

control upon TNFα treatment. Given that ubiquitin-interacting motifs<br />

(UIM) of epsin is important for its interaction with ubiquitinated<br />

proteins, we show that wild type (WT) but not a UIM-deficient<br />

mutant epsin (epsin ∆UIM) coprecipitates with TNFR1 in 293T<br />

cells. Overexpression of epsin ∆UIM inhibits NF-κB activation in<br />

MDA-MB-231 and 293T cells compared to WT. In addition, UIM of<br />

epsin and polyubquitination of RIP are required for the interaction of<br />

epsin with RIP in 293T cells response to TNFα. Conclusion: epsin<br />

promotes breast cancer growth and metastasis by upregulating NFkB<br />

signaling and EMT. Epsin serves as scaffolding protein through<br />

UIM-polyubiquitin chain interaction to stabilize TNFR1 signaling<br />

complex (TNFR-SC) and subsequently enhance NF-kB signaling in<br />

breast cancer cells. Our study provides a basis for novel therapeutic<br />

targets for the development of anti-metastatic cancer treatments.<br />

P1-05-22<br />

<strong>Breast</strong> cancer cells that undergo an Epithelial-to-Mesenchymal<br />

transition co-opt LPP, a regulator of mesenchymal cell migration<br />

and invasion.<br />

Ngan E, Northey JJ, Ursini-Siegel J, Siegel PM. McGill University,<br />

Montreal, QC, Canada; Lady Davis Institute for Medical Research,<br />

Montreal, QC, Canada<br />

Transforming Growth Factor β (TGFβ) promotes breast cancer cell<br />

metastasis to multiple sites, including the bone and lungs. TGFβ is a<br />

strong inducer of Epithelial-to-Mesenchymal transitions (EMT) and<br />

breast cancers that exhibit features of an EMT acquire stem cell-like<br />

characteristics, are highly aggressive, are resistant to therapy and<br />

are refractory to tumor suppressive processes. While TGFβ itself is<br />

non-oncogenic, it is a potent modifier of the malignant phenotype in<br />

breast cancer and is capable of enhancing the migration, invasion and<br />

metastasis of ErbB2 expressing breast cancer cells.<br />

We have identified Lipoma Preferred Partner (LPP) as an<br />

indispensable regulator of TGFβ-induced migration and invasion of<br />

ErbB2 expressing breast cancer cells. LPP is ubiquitously expressed<br />

in smooth muscle cells where it mediates cell adhesion, migration<br />

and the formation of lamellipodial extensions. We hypothesize that<br />

breast cancer cells capable of undergoing an EMT can utilize novel<br />

mediators that are engaged in promoting the migration and invasion<br />

of mesenchymal cells. We propose that LPP is one such example,<br />

which promotes the migration and invasion of ErbB2 expressing<br />

breast cancer cells that have undergone a TGFβ-induced EMT.<br />

We demonstrate that ErbB2 expressing breast cancer cells display<br />

significant increases in cell migration and invasion upon TGFβ<br />

stimulation, and such responses are dependent on LPP expression.<br />

We show that LPP re-localizes to focal adhesion complexes following<br />

TGFβ-induced EMT and it is a critical determinant in focal adhesion<br />

turnover. Furthermore, we determined that LPP targeting to focal<br />

adhesions through its LIM1 domain requires the cooperation of<br />

ErbB2 and TGFβ signaling pathways. Finally, we demonstrate that<br />

LPP promotes TGFβ-induced migration and invasion of ErbB2<br />

expressing breast cancer cells through recruitment of α-Actinin, an<br />

actin cross-linking protein.<br />

Overall, we have identified LPP as a novel mediator that integrates<br />

TGFβ and ErbB2 signaling to promote the migration and invasion of<br />

breast cancer cells that undergo an EMT. Our data reveal that breast<br />

cancer cells, which can transition from an epithelial to mesenchymal<br />

phenotype, can engage a regulator of mesenchymal cell migration<br />

and invasion.<br />

P1-05-23<br />

Blockade of mTORC1 decreases CXCR4-mediated migration<br />

and metastasis<br />

Dillenburg Pilla P, Patel V, T. Dorsam R, Masedunskas A,<br />

Amornphimoltham P, Weigert R, A. Molinolo A, Gutkind JS. NIH,<br />

Bethesda, MD<br />

Tumor cells can co-opt the promigratory activity of chemokines and<br />

their cognate G protein–coupled receptors (GPCRs) to metastasize<br />

to regional lymph nodes or distant organs. Indeed, the migration<br />

toward SDF-1 (stromal cell–derived factor 1) of tumor cells bearing<br />

CXCR4 [chemokine (C-X-C motif) receptor 4] has been implicated<br />

in the lymphatic and organ-specific metastasis of various human<br />

malignancies, including breast cancer. The downstream mechanisms<br />

of CXCR4-mediated migration are not fully elucidated. The study<br />

of the precise signaling pathways involved in cell migration and<br />

metastasis in each tumor type may help identify novel therapeutic<br />

targets for improving cancer therapy. Here we show that blockade<br />

www.aacrjournals.org 175s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

of mTOR complex 1 (mTORC1) by Rapamycin or knocking down<br />

Raptor decreases CXCR4-Gαi mediated cell migration. Moreover,<br />

using a spontaneous lymph node metastasis model, we were able<br />

to show that pharmacological blockade of mTORC1 decreases<br />

lymph node metastasis in vivo. Taken together our data suggest that<br />

Rapamycin, a FDA-approved drug, could be beneficial to prevent<br />

tumor cells spreading from the primary tumor and metastasize.<br />

Molecular events underlying the effects of Rapamycin on CXCR4-<br />

induced migration will be presented.<br />

This research was supported [in part] by the Intramural Research<br />

Program of the NIH, NIDCR.<br />

P1-05-24<br />

Pharmacologic reversion of epigenetic silencing of the PRKD1<br />

promoter blocks breast tumor cell invasion and metastasis.<br />

Borges S, Doppler H, Andorfer CA, Perez EA, Sun Z, Anastasiadis<br />

PZ, Thompson AE, Geiger XJ, Storz P. Mayo Clinic, Jacksonville,<br />

FL; Mayo Clinic, Rochester, MN<br />

Epigenetic silencing of tumor suppressing genes by promoterspecific<br />

DNA methylation is common in many types of cancer.<br />

As an early event, this process has been well shown to promote<br />

tumor initiation and progression; however little is known how<br />

such epigenetic silencing can contribute to tumor metastasis. The<br />

PRKD1 gene encodes Protein Kinase D1 (PKD1), a serine/threonine<br />

kinase expressed in epithelial cells of the normal mammary gland<br />

that maintains the epithelial phenotype of normal breast cells and<br />

prevents epithelial-to-mesenchymal transition (EMT). PKD1 is<br />

also a critical suppressor of tumor cell invasion and is silenced in<br />

expression and activity during breast tumor progression. Here, we<br />

show that aberrant methylation of PRKD1 promoter region is not only<br />

correlated with the silencing of its expression but is also associated<br />

with invasiveness of breast cancer cell lines and with aggressiveness<br />

of breast tumors. Using the highly invasive MDA-MB-231 cells,<br />

we show that the inhibition of PRKD1 promoter methylation with<br />

the DNA methyltransferase inhibitor decitabine restores PKD1<br />

expression and significantly decreases their invasive abilities in<br />

vitro. More importantly, in a tumor xenograft model it dramatically<br />

blocks tumor spread and metastasis to the lung in a PKD1-dependent<br />

fashion. Our data suggest that the status of epigenetic regulation of the<br />

PRKD1 promoter can provide valid information on the invasiveness<br />

of breast tumors, and therefore could serve as an early diagnostic<br />

marker. Moreover, targeted upregulation of PKD1 expression may<br />

be used as a therapeutic approach to reverse the invasive phenotype<br />

of breast cancer cells.<br />

P1-06-01<br />

Upregulation of metabolism as a potential resistance mechanism<br />

to bevacizumab in primary breast cancer<br />

Mehta S, Hughes NP, Adams RF, Li SP, Han C, Kaur K, Taylor NJ,<br />

Padhani AR, Makris A, Buffa FM, Harris AL. Weatherall Institute of<br />

Molecular Medicine, Oxford, United Kingdom; Stanford University,<br />

Stanford, CA; Oxford University Hospitals NHS Trust, Oxford, United<br />

Kingdom; Mount Vernon <strong>Cancer</strong> Centre, Northwood, Middlesex,<br />

United Kingdom; <strong>Cancer</strong> and Haematology Centre, Churchill<br />

Hospital, Oxford, United Kingdom; Paul Strickland Scanner Centre,<br />

Mount Vernon Hospital, Northwood, Middlesex, United Kingdom<br />

Background: Recently the FDA has withdrawn the indication for<br />

bevacizumab in metastatic breast cancer after several clinical studies<br />

failed to demonstrate an overall survival benefit. These studies<br />

however did report an increase in response rates to chemotherapy<br />

and improvement in progression free survival, suggesting a pattern<br />

of response to the drug followed by the development of resistance.<br />

We have little knowledge of the molecular mechanisms driving the<br />

development of resistance to bevacizumab. To better understand these<br />

mechanisms, we have conducted a window of opportunity study<br />

using a single cycle of bevacizumab with detailed pharmacodynamic<br />

assessments using gene expression arrays and dynamic contrastenhanced<br />

magnetic resonance imaging (DCE-MRI).<br />

Methods: After ethical approval, 47 newly diagnosed locally<br />

advanced breast cancer patients were prospectively enrolled in this<br />

trial. Patients received single dose bevacizumab (15mg/ kg) 2 weeks<br />

prior to neoadjuvant chemotherapy and underwent core biopsies for<br />

gene expression and immunohistochemistry analysis and DCE-MRI<br />

scans before and 2 weeks after bevacizumab. 35 patients who had<br />

invasive ductal carcinoma together with good quality MRI scans and<br />

core biopsies before and after bevacizumab were included in this<br />

analysis. Pharmacokinetic (PK) modelling techniques were used to<br />

quantify PK parameters (K trans , k ep , v e ) from the DCE-MRI data. Gene<br />

expression profiling was performed using the Affymetrix Human<br />

Exon 1.0 ST arrays.<br />

Results: The majority of patients (28 / 35) showed a significant<br />

reduction in vessel permeability and blood flow of at least 30%<br />

following bevacizumab, with a mean decrease in the forward transfer<br />

constant (P < 0.0001) and the reverse rate constant k ep (P < 0.0001).<br />

From gene expression and immunohistochemistry analyses, we<br />

identified several key metabolism-related genes that are significantly<br />

up-regulated after bevacizumab treatment, including pyruvate<br />

dehydrogenase kinase isozyme 1 (PDK1) (fig.1) and carbonic<br />

anhydrase 9 (CA9). In addition, we found a number of interesting<br />

genes that are down-regulated after bevacizumab treatment, including<br />

sulfatase-1 (SULF1), and cyclin E1 (CCNE1).<br />

Discussion: This study highlights that the combination of DCE-MRI<br />

and gene expression arrays can lead to an improved understanding<br />

of the molecular mechanisms governing response and resistance to<br />

anti-angiogenic therapy. Heterogeneity of response to bevacizumab<br />

was demonstrated, with some tumours showing increases or no<br />

change in K trans and others marked reductions, which may be of value<br />

in early stratification for therapy maintenance. Furthermore, the gene<br />

expression analysis showed activation of pathways, which could<br />

contribute to the development of resistance. For example, we observed<br />

significant up regulation of genes involved in regulating the switch<br />

from mitochondrial metabolism to glycolysis, such as PDK1. This<br />

suggests that using bevacizumab with the other targeted agents such<br />

as Dichloroacetate, a PDK1 inhibitor might be helpful in overcoming<br />

the development of resistance and ultimately lead to improved patient<br />

survival. Our preclinical studies strongly support this possibility.<br />

P1-06-02<br />

A newly angiogenic biomarker Vasohibin-1 expression in ductal<br />

carcinoma in situ of the breast.<br />

Tamaki K, Tamaki N, Kamada Y, Uehara K, Miyashita M, Ishida T,<br />

Ohuchi N, Sasano H. Nahanishi Clinic Okinawa, Naha, Okinawa,<br />

Japan; Tohoku University Graduate School of Medicine, Sendai,<br />

Miyagi, Japan; Tohoku University Hospital, Sendai, Miyagi, Japan<br />

Vasohibin-1 is a recently identified negative feedback regulator of<br />

angiogenesis induced by VEGF-A and bFGF. Our previous study<br />

was the first study demonstrating the status of vasohibin-1 in human<br />

breast lesions, which indicates that vasohibin-1 is associated with<br />

neovascularization and may especially play important roles in the<br />

regulation of intratumoral angiogenesis in human breast cancer.In<br />

this study, we first evaluated mRNA expression of vasohibin-1 and<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

176s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 1<br />

CD31 in 39 Japanese emale breast carcinoma specimens including<br />

22 invasive ductal arcinoma (IDC) and 17 ductal carcinoma in situ<br />

(DCIS) using a real-time quantitative RT-PCR (QRT-PCR) with<br />

LightCycler system. Table shows the primer sequences used in realtime<br />

PCR in this study.<br />

Primer sequences used in real-time PCR in this study<br />

Gene (accession no.) Primer for PCR Size (bp)<br />

CD31 (NM000442) Forward: GATGTCAGAAACCATGCAA 199<br />

Reverse: AGCCTTCCGTTCTAGAGTATC<br />

Vasohibin-1 (KIAA1036) Foreward: AGATCCCCATACCGAGTGTG 167<br />

Reverse: GGGCCTCTTTGGTCATTTCC<br />

RPL13A (NM012523) Forward: CCTGGAGGAGAAGAGGAAAAG 125<br />

Reverse: TTGAGGACCTCTGTGGTATTT<br />

In addition, we also immunolocalized vasohibin-1 and CD31 and<br />

compared their immunoreactivity to nuclear grades and histological<br />

grades of 100 carcinoma cases (50 IDC and 50 DCIS). There were<br />

no statistically significant differences of CD31 mRNA expression<br />

and the number of CD31 positive vessels between DCIS and<br />

IDC (P = 0.250 and P = 0.191, respectively), whereas there was a<br />

statistically significant difference in vasohibin-1 mRNA expression<br />

and the number of vasohibin-1 positive vessels in DCIS and IDC (P<br />

= 0.022 and P


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

as 0, 1+, 2+ or 3+; however, scoring was tabulated in two ways. The<br />

first was an absolute HER2 score based solely on analysis of the<br />

carcinoma, and the second was a normalized HER2 score, obtained<br />

by subtracting the score of the non-neoplastic breast epithelium from<br />

that of the infiltrating cancer. FISH was performed using the Vysis<br />

PathVysion combined HER2 and CEP17 probe kit with morphometric<br />

analysis performed using a MetaSystems image analysis system that<br />

incorporated the Metafer software. A score of > 2.2 was required for<br />

classification as amplified, and 2.0), borderline (1.5 to 2.0)<br />

and negative. (2.0, 1.8 to 2.0 and


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 1<br />

P1-07-05<br />

Evaluation of tissue processing factors affecting HER2 IHC<br />

staining intensity in breast cancer cell lines<br />

Jensen K, Erickson J, Webster S, Pedersen HC. Dako Denmark,<br />

Glostrup, Denmark; Dako North America, Carpinteria, CA<br />

Background<br />

In breast cancer the HER2 status as established by IHC (e.g.<br />

HercepTest) determines eligibility for HER2 targeted therapy.<br />

Pre-analytical procedures including tissue processing create variation<br />

in HER2 intensity seen in breast cancer tissues. Therefore, there is a<br />

need for a greater understanding of the effects of the individual tissue<br />

processing steps on HER2 staining intensity.<br />

Control cell lines (CCL) are commonly used as staining run controls<br />

in immunohistochemistry (IHC) in both pharmacodiagnostic,<br />

routine and in clinical research. As a surrogate for cancer tissue,<br />

these immortalized cancer cells can serve as a model system for<br />

investigating the effect of process variables on staining intensity.<br />

This study was designed to investigate if variations in cell/tissue<br />

processing (fixation, embedding and sectioning) affect the IHC<br />

staining intensity in a HER2 formalin- fixed, paraffin-embedded<br />

(FFPE) control cell lines.<br />

To introduce a more objective scoring we have evaluated the<br />

application of digital image analysis combined with design of<br />

experiment (DOE) statistics.<br />

Methods<br />

The following tissue processing parameters were investigated:<br />

embedding medium (HistoGel or agarose), fixation time (18 or 48<br />

hours in neutral buffered formalin), density of cells (25% or 50%)<br />

and thickness of cut sections (2 µm, 4 µm or 6 µm). Factors were<br />

set up in a 2 k factorial experimental design; all slides were stained<br />

with Dako HercepTest. All slides were scanned, analyzed and<br />

compared on two different image analysis systems: ACIS®III (Dako)<br />

and Scanscope CS (Aperio).<br />

The DOE statistical analysis revealed the contribution of individual<br />

factors as well as any additive effect of a combination of factors.<br />

Analysis was based on the level of intensity as well as histoscores.<br />

Results<br />

In a 2 4 DOE experiment, several of the parameters described above<br />

were associated with decreased staining intensity: increased fixation<br />

time (p < 0.001), decreased section thickness (p < 0.001) and<br />

decreased cell density (p < 0.001). In a separate 2 3 experiment, it<br />

was shown that fixation media in combination with cell density was<br />

associated with changes in staining intensity (p values from 0.001 to<br />

0.022). Three different algorithms and two different slide scanners<br />

were tested and results were consistent regardless of the image<br />

analysis system or the algorithm used.<br />

Conclusions<br />

This study identified several tissue processing factors which impact<br />

IHC staining intensity in a HER2 expressing CCL. The discovery<br />

of these effects has advanced the understanding of processing<br />

methodology and the influence of pre-analytical factors on HER2<br />

IHC staining intensity.<br />

The application of a DOE setup with quantification by image<br />

analysis can be a useful tool for the systematic evaluation of several<br />

variables compared with the standard “one factor at a time” design.<br />

A multifactor effect between fixation media and cell density was<br />

detected which would not have been possible in a “one factor at a<br />

time” design. Implementing the knowledge achieved from this study<br />

provides a methodology for standardizing tissue processing and<br />

thereby improving HER2 testing in breast cancer.<br />

P1-07-06<br />

Effect of biospecimen variables on proteomic biomarker<br />

assessment in breast cancer<br />

Meric-Bernstam F, Akcakanat A, Chen H, Sahin A, Tarco E, Carkaci<br />

S, Adrada B, Singh G, Anh-Do K, Garces Z, Mittendorf EA, Babiera<br />

G, Wagner J, Bedrosian I, Krishnamurthy S, Symmans WF, Gonzalez-<br />

Angulo AM, Mills G. UT MD Anderson <strong>Cancer</strong> Center, Houston, TX;<br />

Ohio State University, Columbus, OH<br />

Background: PI3K/Akt/mTOR signaling is being actively pursued as<br />

a therapeutic target. We sought to determine how tumor heterogeneity<br />

and biospecimen variables affect assessment of PI3K/Akt/mTOR<br />

pathway activation.<br />

Methods: Intraoperative image-guided core-needle biopsies (CNB)<br />

of primary breast tumors were prospectively collected in 53 patients<br />

with invasive breast cancer. After surgery, specimens were collected<br />

from the center and periphery of the excised tumor. CNB, central<br />

and peripheral surgical specimens were assessed with reverse phase<br />

proteomic arrays (RPPA), H&E and immunohistochemistry (IHC).<br />

Results: The expression of standard of care markers ER, PR, and<br />

HER2 by RPPA correlated well between biospecimen types. Overall,<br />

there was a significant correlation between the expression of 132<br />

(86%) of 154 different markers in the center and periphery; the<br />

correlation was significantly higher for smaller tumors, and with<br />

shorter cold ischemia time. Expression of many investigational<br />

prognostic markers and druggable targets on CNB correlated with<br />

expression in the surgical specimen (average of center and periphery),<br />

while others, such as EGFR and c-MET, had a weak correlation. Of<br />

154 RPPA markers, 132 (86%) were not statistically different between<br />

the center and periphery, and 97 (67%) were not different between<br />

the CNB and the surgical specimen. On analysis of the PI3K/AKT/<br />

mTOR pathway, pAkt S473 and PTEN had a significant correlation<br />

between central and peripheral specimens, and between CNB and<br />

surgical specimens. However, pAkt S473, pS6 S235/236 and pS6<br />

240/244 levels were higher in CNB than the central specimens both<br />

by RPPA and by IHC. When patients were classified by RPPA PI3K<br />

pathway activation score, there was a moderate agreement between<br />

classification on the CNB and central specimens (Cohen’s Kappa<br />

0.539). However 9 of 20 tumors classified as having PI3K activation<br />

on CNB were classified as not having pathway activation on central<br />

specimens.<br />

Conclusions: There is remarkable homogeneity in expression<br />

of biomarkers within a tumor. However, proteomic markers are<br />

differentially expressed by biospecimen type and other preanalytic<br />

variables. PI3K pathway activation is greater in CNB compared to<br />

surgical samples.<br />

P1-07-07<br />

Inflammatory gene expression variations in the interval between<br />

core needle biopsy and excisional biopsy in early breast cancer<br />

Jeselsohn RM, Regan MM, Werner L, Fatima A, He HH, Brown M,<br />

Iglehart JD, Richardson AL, Come S. Beth Israel Deaconess Medical<br />

Center, Boston, MA; Dana Farber <strong>Cancer</strong> Institute, Boston, MA;<br />

Brigham and Women’s Hospital, Boston, MA<br />

Introduction: Advancements in molecular biology have unveiled<br />

multiple breast cancer promoting pathways and potential therapeutic<br />

targets. Large randomized clinical trials remain the ultimate means<br />

of validating therapeutic efficacy, but they require large cohorts of<br />

patients and are lengthy and costly. An alternative approach is to<br />

conduct a window of opportunity study in which patients are exposed<br />

to a drug pre-surgically during the interval between the core needle<br />

biopsy (CNB) and the definitive surgery (excisional biopsy (EB)).<br />

www.aacrjournals.org 179s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

These are non-therapeutic studies and the end point is not clinical or<br />

pathological response but rather evaluation of molecular changes in<br />

the tumor specimens that can predict response. However, since the end<br />

points of the non-therapeutic studies are biologic, it is critical to first<br />

define any biologic changes that occur in the absence of treatment. In<br />

this study, we compared the molecular profiles of breast cancer tumors<br />

at the time of the diagnostic biopsy versus the definitive surgery in<br />

the absence of any intervention.<br />

Methods: The study was conducted with DFCI/HCC IRB approval<br />

and patient consent. Post-menopausal women with a breast lesion<br />

suspected to be cancerous were eligible for this study. We obtained<br />

a tissue specimen at the time of a CNB and if determined to be<br />

consistent with invasive carcinoma a second specimen was obtained at<br />

the time of the EB. We used the Nanostring Ncounter system to study<br />

the expression level of 148 transcripts. Since we expected that most<br />

of the tumors will be hormone receptor positive (HR+), the library<br />

included; genes that have been shown to be prognostic in HR+ tumors<br />

(Oncotype DX®, PAM50), estrogen receptor (ER) modulators, ER<br />

responsive genes and inflammatory genes. The Wilcoxon’s signed<br />

rank test was used to evaluate for changes in gene expression levels<br />

between the paired samples.<br />

Results: 25 patients were enrolled in this study and paired tumor tissue<br />

samples were obtained from all patients. 21 of the paired samples<br />

were successfully analyzed by the nanostring system. 86% of the<br />

patients are HR+/Her2-.We found that the gene expression levels<br />

of 14 out of the 148 genes (9%) did change between the CNB and<br />

EB without any intervention (p


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 1<br />

Method. Patients with primary breast carcinoma treated at our center<br />

who had complete ER status from January 1990 to December 2011<br />

were included in this study. Patients were excluded if they presented<br />

with recurrent or metastatic disease. For statistical analyses, patients<br />

who underwent surgery for breast cancer were separated into three<br />

groups: ER-positive ≥10%, ER-positive 1-10% and ER negative.<br />

Analyses comparing various clinical and pathologic characteristics<br />

among patients with different ER status were performed. Survival<br />

rates were calculated by the Kaplan-Meier method.<br />

Result. Patients whose tumors were ER-positive 1-10% (2.7%)<br />

were younger (median age 53 Vs. 56 years, P


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

structures were analyzed at 100X. IHC was performed using Abs<br />

directed to the following PR phosphorylated sites (pAb) pSer 162,<br />

190, 294, 400 and 554. Six samples were selected for IHF using a<br />

secondary fluorescent Ab. RESULTS: All cases were PR [+]. At<br />

high magnification (100X), 2 PR nuclear distribution patterns were<br />

observed: a diffuse finely granular staining (D) or an aggregated<br />

pattern (A). This resulted in 3 tumor phenotypes (PR B Ab): A cells<br />

only in 2 cases, D cells only in 6, and a mixture of both A and D cells<br />

(AD) in 4. PR [-] malignant cells were present in various proportions.<br />

The IHF results were consistent with IHC studies. Only pAb to<br />

pSER190 was specific and sensitive, with a smaller number of [+]<br />

malignant cells relative to the standard PRB. CONCLUSION: We<br />

have developed an IHC method that utilizes formalin-fixed paraffinembedded<br />

tissue allowing the technique to be applied on a routine<br />

basis. Three classes of PR nuclear distribution have been defined: D,<br />

A and AD. D is consistent with the expression of PR that is weakly<br />

transcriptionally active or inactive and thus potentially predictive of<br />

poor patient response to antiPg. The A or AD pattern of PR nuclear<br />

distribution indicates active PR; patients with this pattern may exhibit<br />

a increased probability of benefiting from an antiPg. Preclinical<br />

studies have demonstrated that activated PR [+] cells can in turn<br />

stimulate PR [-] (stem or progenitor) mammary epithelial cells via<br />

secreted factors. In light of these findings, we are expanding the<br />

cohort to refine and confirm this biomarker technique. Additional<br />

preclinical work is ongoing to expand the biological rationale and to<br />

evaluate this technique alongside other technologies such as PR gene<br />

expression analysis. We anticipate that approximately 50% of PR+<br />

BC cases contain the A/AD phenotye and therefore may benefit from<br />

antiPg. The method of IHC described herein is a clinically applicable<br />

tool that may allow for selection of patients most likely to respond<br />

to antiPg treatment.<br />

and negation identification. The output from the NLP system was<br />

further processed in three steps: Identification of the HER2 concept,<br />

followed by extraction of the most likely HER2 value, and lastly, a<br />

decision module to select the most likely HER2 value if there were<br />

conflicting values.<br />

Results:<br />

Use the cancer registry data as the gold standard, for positive and<br />

negative HER2 values, the sensitivity and specificity of the NLP<br />

algorithm were 94.7% and 93.3%, respectively, including the cases<br />

where NLP did not select either positive or negative values. A manual<br />

chart review was performed on the discrepant cases. We found that the<br />

NLP were correct for many of these cases. For example, out of the 39<br />

NLP positive but registry negative cases, 4 were false positives and<br />

35 were true positives. Compared to the cancer registry data, NLP<br />

increased capture of positive and negative HER2 cases from 49% to<br />

73% of the cohort population.<br />

Discussion:<br />

NLP provides the opportunity to process clinical notes which are added<br />

to the EMR after the cancer registry has completed documenting the<br />

patients’ initial course of cancer treatment. On the other hand, NLP<br />

was not able to access some of the HER2 related clinical notes. For<br />

example, according to our chart review, some notes were stored as<br />

scanned images and not retrievable. In addition, clinical notes with<br />

incorrect filing dates so patients without any diagnosis date were not<br />

processed. We also identified key challenges in determining HER2<br />

results using NLP in this study. First, non-standard terminology for the<br />

HER2 receptor in the clinical notes hampered the NLP effectiveness.<br />

Second, clinicians described HER2 results in many different ways.<br />

NLP compared to the <strong>Cancer</strong> Registry<br />

HER2 Values # of Patients NLP Positive NLP Negative NLP neither Pos/Neg<br />

Registry Positive 266 252 9 5<br />

Registry Negative 1140 39 1064 30<br />

P1-07-12<br />

Using Natural Language Processing to Identify and Extract HER2<br />

Value from a large EMR system<br />

Zheng C, Avila C, Haque R. Kaiser Permanente Southern California,<br />

Pasadena, CA<br />

Background:<br />

HER2 status is important in breast cancer prognosis but has not been<br />

well collected in NCI-SEER affiliated cancer registries until recent<br />

years. For example, in a cohort of 2,846 women who were diagnosed<br />

with breast cancer from 1997 to 2011 in a large health plan, only<br />

49% of these patients had known HER2 status. However, many of<br />

these with missing HER2 status were documented in clinical notes.<br />

This study used Natural Language Processing (NLP) technology<br />

to identify and extract HER2 status. NLP results were validated by<br />

comparison with cancer registry data and any discrepancies were<br />

manually reviewed.<br />

Method:<br />

We assembled a cohort of 2,846 breast cancer patients from the<br />

membership of a health plan based in southern California, Kaiser<br />

Permanente. All free text notes including pathology reports, progress<br />

notes, and discharge summaries were extracted from our electronic<br />

medical record (EMR) system. A window of 9 months before and after<br />

the diagnosis date was applied to restrict the number of notes needed<br />

to be processed. Overall, 513,903 clinical notes were processed and<br />

indexed, which averages to 180 notes per patient. Separate ontologies<br />

were created for the HER2 terminology and HER2 status. HER2<br />

status values included positive, negative, borderline, test performed<br />

and test not performed. The NLP system employed additional<br />

components such as spelling correction, acronym recognition<br />

P1-07-13<br />

Prognostic relevance of statistically standardized estrogen<br />

receptor (ER), progesterone receptor (PR), and human epidermal<br />

growth factor receptor 2 (HER2) in tamoxifen(TAM)-treated<br />

NCIC CTG MA.14 patients<br />

Chapman J-AW, Sgroi D, Goss PE, Richardson E, Binns SN, Zhang Y,<br />

Schnabel CA, Erlander MG, Pritchard KI, Han L, Shepherd LE, Pollak<br />

MN. NCIC Clinical Trials Group, Queen’s University, Kingston, ON,<br />

Canada; Harvard University, Boston, MA; bioTheranostics, Inc.,<br />

<strong>San</strong> Diego, CA; Sunnybrook Odette <strong>Cancer</strong> Centre, University of<br />

Toronto, ON, Canada; Jewish General Hospital, McGill University,<br />

Montreal, QC, Canada<br />

Background: Poor inter-laboratory comparability of common<br />

clinically used breast cancer biomarkers led to a proposal of statistical<br />

standardization (SS) of laboratory results, similar to bone mineral<br />

density (BMD) z-scores. This analysis is the first utilization of SS in a<br />

trial where all women received TAM. Methods: MA.14 allocated 667<br />

postmenopausal women to TAM +/- Octreotide LAR (OCT) based on<br />

locally determined ER/PR, without HER2 status. At 9.8 yrs median<br />

follow-up, the secondary endpoint of relapse-free survival (RFS) had<br />

a non-significant hazard ratio (HR) for TAM-OCT to TAM of 0.87<br />

(95% CI 0.63-1.21; p=0.40). 299 patients who were representative of<br />

MA.14 patients by treatment and stratification factors (exact Fisher<br />

p-values=0.19-0.90) had their tumors centrally assessed for ER, PR,<br />

and HER2 by RT-PCR. Continuous values were used for SS of each<br />

biomarker. Univariate (uni) assessment used similar categorizations<br />

as those for BMD, assigning ER/PR/HER2 values by number of<br />

standard deviations (SD) about the mean (Group 1, z-score ≥1.0 SD<br />

below mean; Group 2, z-score


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 1<br />

≤1.0 SD above mean; Group 4, z-score >1.0 SD above mean). A logrank<br />

statistic was used to test for differences between SS biomarker<br />

groups with K-M plots for graphical description. Multivariate (multi)<br />

effects of SS biomarkers and baseline patient characteristics on RFS<br />

were examined with exploratory (un)stratified Cox step-wise forward<br />

regression, adding a factor if likelihood ratio criterion was p≤0.05.<br />

Sensitivity analyses used a prior external HER2+ cut-point of ≥1.32<br />

SD. Results: 292 patient samples passing internal analytical quality<br />

control were included in this analysis. Uni analyses indicated SS<br />

ER was not associated with RFS (p=0.31). SS PR had a significant<br />

uni effect on RFS [p=0.03; Group 4 compared to Group 1, HR of<br />

0.33 (95% CI 0.12-0.90); Group 3 compared to Group 1, HR of 0.42<br />

(95% CI 0.21-0.83); and Group 2 compared to Group 1 HR of 0.70<br />

(95%CI 0.36-1.37)]. SS HER2 also had a significant uni effect on<br />

RFS [p=0.004; Group 4 compared to Group 1, HR of 0.90 (95% CI<br />

0.37-2.16)]; Group 3 compared to Group 1, HR of 0.39 (95% CI<br />

0.18-0.84); and, Group 2 compared to Group 1, HR of 0.34 (95%<br />

CI 0.16-0.70)]. Multi stratified/unstratified Cox models indicated T1<br />

tumours (p=0.02/p=0.0002) and higher SS PR (p=0.02/0.01) were<br />

associated with significantly longer RFS; other unstratified results<br />

showed that N-ve patients had better RFS (p0.05). The HER2+ cut-point<br />

of ≥1.32 SD indicated directionally worse RFS (uni p-value=0.05;<br />

multi p-value=0.06). Discussion: In MA.14, all women received<br />

TAM. Local ER/PR status using categorical or semi-quantitative<br />

values did not impact RFS. A statistically standardized approach using<br />

continuous centralized ER, PR, HER2 by RT-PCR demonstrated that<br />

increasing PR values were associated with better RFS. Evaluation in<br />

other trials may provide support for this methodology.<br />

P1-07-14<br />

Enabling biomarker validation in breast cancer molecular<br />

subtypes: sensitivity and specificity of array-based subtype<br />

classification in 983 patients<br />

Györffy B, Lanczky A. Semmelweis University<br />

Background: The diverse breast cancer molecular subtypes have<br />

different clinicopathological characteristics and outcomes. Here,<br />

we assessed the correlation between microarray-defined and authorreported<br />

molecular subtypes using publicly available breast cancer<br />

datasets.<br />

Methods: GEO was searched for datasets where the authors<br />

determined molecular subtypes. The molecular subtype was recalculated<br />

for each patient using the StGallen guidelines by assessing<br />

the microarray-based expression of ER, HER2 and MKI67 (Basal:<br />

ER negative + HER2 negative, Luminal A: ER positive + HER2<br />

negative + low MKI67, HER2 enriched: HER2 positive + ER negative,<br />

Luminal B: ER positive + HER2 positive and ER positive + HER2<br />

negative + high MKI67). The thresholds used were 500 for ER (probe<br />

set 205225_at), 4800 for HER2 (216836_s_at) and 470 for MKI67<br />

(212021_s_at). Sensitivity and specificity were calculated for each<br />

subtype separately.<br />

Results: Molecular subtype was published in all together six datasets<br />

(GSE1456, GSE21653, GSE25066, GSE20711, GSE31519 and<br />

GSE17907) for 983 patients. In these, 380, 306, 169 and 128 were<br />

Basal, Luminal A, Luminal B and HER2 enriched, respectively. The<br />

microarray-based molecular subtype determination resulted in a<br />

sensitivity of 0.70, 0.55, 0.63 and 0.38 and a specificity of 0.96, 0.87,<br />

0.67 and 0.99 for Basal, Luminal A, Luminal B and HER2 enriched<br />

subtypes, respectively. Finally, the option to filter for molecular<br />

subtypes was implemented into our online biomarker validation<br />

platform at www.kmplot.com.<br />

Discussion. Microarray data provided highest sensitivity and<br />

specificity to independent subtype classification for Basal tumors,<br />

while the luminal B subtype displayed the highest discordance.<br />

Our registration-free online service enables the validation of gene<br />

expression based biomarkers in each subtype separately.<br />

P1-07-15<br />

Revisiting chromosome 17q copy number aberrations in early<br />

high-risk breast cancer<br />

Kotoula V, Bobos M, Eleftheraki AG, Timotheadou E, Razis E, Goussia<br />

A, Levva S, Kalogeras KT, Pectasides D, Fountzilas G. Hellenic<br />

Cooperative Oncology Group (HeCOG), Athens, Greece<br />

Background — Aim: HER2 and TOP2A gene status are of prognostic/<br />

predictive relevance and are broadly determined with fluorescent in<br />

situ hybridization (FISH). For this purpose, probes against (A)<br />

17p11.1-q11.1 (>5Mb, CENtromere), (B) 17q12 (600Kb, including<br />

HER2 among other genes), and (C) 17q21-22 (500Kb, similarly<br />

including TOP2A) are used, and gene pathology is determined by<br />

B/A and C/A signal ratios according to consensus cut-offs. However,<br />

chromosome 17 (chr17) and 17q pathology is complex and may be<br />

misinterpreted by this approach, especially in cases with low copy<br />

gains. Herein, we profiled A, B and C signals in 1027 adjuvantly<br />

treated, early breast cancer patients.<br />

Methods: Maximal A, B, C copy numbers (raw FISH data) were<br />

submitted to hierarchical clustering as continuous variables. Clusters<br />

were compared with clinicopathological parameters, conventional<br />

FISH ratios, and were evaluated for their prognostic significance<br />

(overall survival, OS) with respect to tumor Ki67 status (13% cut-off),<br />

TopoIIa protein (5% cut-off) and ER/PgR positivity (1% cut-off).<br />

Results: Clustered values resulted into 832 (81%) putatively chr17<br />

stable tumors (ABC low ) and 195 17q unstable tumors. Out of these,<br />

43 (4%) had possible chr17 gain (ABC high ), 30 (3%) HER2/TOP2A<br />

gain (BC high ), and 122 (12%) HER2 gain (B high ). Unstable 17q was<br />

more frequent in Ki67 high, ER/PgR negative, high-grade tumors, and<br />

were almost exclusively present in luminal-HER2 and HER2-enriched<br />

tumors (all Pearson’s p’s


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P1-07-16<br />

Liver derived epithelial cells as source of false positive circulating<br />

tumor cells in early breast cancer<br />

Habets L, Körber W, Frenken B, Danaei M, Kusche M, Peisker<br />

U, Kroll T, Pachmann K. Metares.e.V, Aachen, NRW, Germany;<br />

Brustzentrum Aachen Kreis Heinsberg, Aachen, NRW, Germany;<br />

Medizinische Universitätsklinik Jena, Thueringen, Germany<br />

The MAINTRAC technique as introduced by our coworkers from<br />

Jena (RBC lysis,fluorometric detection and analysis on Olympius<br />

ScanR) detects more circulating epithelial cells than techniques<br />

using enrichment. Also cells with a low EPCAM expression are<br />

detected and not only the typical cells with bright expression found<br />

after immunomagnetic enrichment. The relative cheapness and<br />

reproducibility allows frequent monitoring during and after therapy<br />

Using 3 colour detection (EPCAMfitc, DAPI, Vimentin PE) living and<br />

dead circulating epithelial cells in EMT, or cells in EMT with stemcell<br />

markers (EPCAMfitc, Vimentin-PE, CD44PacBlue) can be detected.<br />

In early breast cancer (n=135) cells can be found in 60% of patients<br />

and in 40% higher cell counts (>100 ml are detectable. A control<br />

population(n=100) showed low numbers in 98% (e.g (


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 1<br />

P1-07-18<br />

Association between Bone Turnover Markers in patients<br />

with breast cancer and bone metastases on treatment with<br />

bisphosphonates (ZOMAR study)<br />

Tusquets I, De la Piedra C, Manso L, Crespo C, Gómez P, Calvo L,<br />

Ruiz M, Martínez P, Perelló A, Antón A, Codes M, Margelí M, Murias<br />

A, Salvador J, Seguí MA, De Juán A, Gavilá J, Luque M, Pérez D,<br />

Zamora P, Arizcum A, Chacón JI, Heras L, Barnadas A. Hospital<br />

del Mar, Barcelona, Spain; Instituto de Investigación <strong>San</strong>itaria<br />

Fundación Jiménez Díaz, Madrid, Spain; Hospital 12 de Octubre,<br />

Madrid, Spain; Hospital Universitario Ramón y Cajal, Madrid, Spain;<br />

Hospital Universitario Vall d’Hebrón, Barcelona, Spain; Hospital<br />

Universitario A Coruña Juan Canalejo, A Coruña, Spain; Hospital<br />

Universitario Virgen del Rocío, Sevilla, Spain; Hospital de Basurto,<br />

Vizcaya, Spain; Hospital Son Dureta, Palma de Mallorca, Spain;<br />

Hospital Miguel Servet, Zaragoza, Spain; Hospital Virgen Macarena,<br />

Sevilla, Spain; H. Universitario Trias y Pujol, Barcelona, Spain;<br />

Hospital Universitario Insular de Gran Canaria, Gran Canaria,<br />

Spain; Hospital Nuestra Señora de Valme, Sevilla, Spain; Hospital<br />

Parc Taulí Sabadell, Barcelona, Spain; Hospital Marqués Valdecilla,<br />

<strong>San</strong>tander, Spain; Instituto Valenciano de Oncología, Valencia, Spain;<br />

Hospital General de Asturias, Oviedo, Spain; Hospital Costa del Sol,<br />

Málaga, Spain; Hospital La Paz, Madrid, Spain; Hospital Palencia<br />

Río Carrión, Palencia, Spain; Hospital Virgen de la Salud, Toledo,<br />

Spain; Hospital Cruz Roja Hospitalet del Llobregat, Barcelona,<br />

Spain; Hospital <strong>San</strong>ta Creu i <strong>San</strong>t Pau, Barcelona, Spain<br />

Background:<br />

The presence of bone metastases (BMe) alters the balance of bone<br />

remodeling and consequently, levels of bone turnover markers (BTM).<br />

Increased levels of these biomarkers are related to the risk of skeletalrelated<br />

events (SREs), disease progression and death. Treatment with<br />

bisphosphonates reduces the probability of SREs through osteoclastic<br />

activity inhibition. The aim of this study was to determine the relation<br />

between BTM, bone metastasis development and SREs, disease<br />

progression and death in patients with breast cancer (BC) and BMe.<br />

Patients and methods:<br />

Observational, prospective and multicenter study. Patients with BC<br />

and BMe; no previous bone treatment in the last 6 months prior to<br />

study entry. Urinary aminoterminal telopeptide of collagen I (NTX,<br />

Osteomark NTx Urine, Wampole Laboratories, USA); urinary<br />

alpha-alpha-isomer of carboxyterminal telopeptide of collagen I<br />

(αα-CTX, ALPHA Crosslaps EIA, ids, UK) and serum bone alkaline<br />

phosphatase (BALP, OSTASE BAP, ids, UK) were determined at<br />

baseline (V0) and every 3 mo along 18 months (V6). Patients were<br />

treated with zoledronic acid (ZA) at inclusion and every 3-4 weeks.<br />

Results:<br />

234 patients with BC and BMe were analyzed. BTM results were<br />

available for 219 patients at basal visit (V0) and every 3 mo of<br />

treatment along 18 months (V6). Population basal characteristics<br />

(234 patients): mean age: 59.8 years; ER+: 80.3%; PR+: 64.9%;<br />

HER 2+: 18.3%. Patients with pathologic baseline levels were: 49.8%<br />

NTX, 39.6% αα-CTX and 83.4% BALP. A significant decrease was<br />

observed in BTM at V2 vs V0 after 6 months: 13.7%, 8.4% and 58.4%<br />

presented pathologic values of NTX, αα-CTX and BALP respectively.<br />

Normalized levels remained steady throughout 18 mo follow-up,<br />

finding significant decrease for each BMT for each time point except<br />

at V6 for αα-CTX. Regarding association between BTM and SREs,<br />

progression and exitus, a significant association was observed between<br />

pathologic levels of BTM throughout follow-up: with SRE at V3,V4<br />

for NTX; with disease progression at V3,V4,V5,V6 for NTX, at V2<br />

for αα–CTX and at each follow up visit for BALP; and with death<br />

at V1,4,5 for NTX, at V5 for αα–CTX and at V1,2,3,4,5 for BALP.<br />

Conclusions:<br />

Addition of ZA to standard systemic therapy reduced BTM levels<br />

during the first 3 months of treatment and normalized levels remained<br />

steady throughout 18 months follow up except at Month 18 for αα-<br />

CTX. Pathological levels of BTM were significantly associated with<br />

SRE, disease progression and death.<br />

P1-07-19<br />

Mass Spectrometry Based Quantitative Analysis of the HER<br />

Family receptors in FFPE <strong>Breast</strong> <strong>Cancer</strong> Tissue<br />

Hembrough TA, Scaltriti M, Serra V, Jimenez J, Perez J, Liao<br />

W-L, Thyparambil S, Cortes J, Baselga J, Burrows J. OncoPlex<br />

Diagnostics, Inc., Rockville, MD; Vall d’Hebron Istitute of Oncology,<br />

Barcelona, Spain; Massachusetts General Hospital, Boston, MA<br />

The human EGF receptor family (HER’s) consists of two clinically<br />

validated drug targets (EGFR and HER2), a third (HER3) currently<br />

under investigation for its possible role in the acquisition of multidrug<br />

resistance and a fourth (HER4), the role of which is still matter of<br />

debate. Drugs inhibiting EGFR or HER2 show significant antitumor<br />

activity in the clinic, however, acquisition of resistance is a hallmark<br />

of these and most targeted therapies. In the case of EGFR and HER2,<br />

one of the emerging resistance mechanisms is the co-expression<br />

of HER3. Indeed, recent reports show that inhibition of the PI3K<br />

pathway leads to upregulation of HER3, and subsequent resistance.<br />

Clinical analysis of protein levels in formalin fixed paraffin embedded<br />

(FFPE) tissues is limited to immunohistochemistry (IHC), which<br />

is semi-quantitative and requires significant amounts of tissue.<br />

Moreover, the vast majority of research groups consider specific<br />

HER3 staining by IHC particularly challenging. However, accurate<br />

measurement of these targets is critical both for properly defining<br />

treatment groups and predicting patterns of resistance.<br />

In order to address these issues, we used trypsin digestion mapping<br />

and stable isotope labeled peptides to develop a panel of quantitative<br />

mass spectrometric (MS) assays to measure the levels of EGFR,<br />

HER2, HER3 and other clinically relevant targets in FFPE breast<br />

cancer tissue. These quantitative MS assays were then multiplexed<br />

to analyze 1µg of tumor protein.<br />

In this study, we multiplexed HER family analysis on 31 HER2<br />

positive breast cancers. Tumor tissue was microdissected from<br />

FFPE sections, and subjected to quantitative MS analysis of EGFR,<br />

HER2, HER3 as well as IGF1R and cMET. Quantitation of HER2<br />

showed a broad range of HER2 expression in these tissues. The<br />

highest expresser measured 26 fmol/ug tumor tissue, representing<br />

amplification and massive protein over expression. In contrast, five<br />

tissues showed low levels of HER2 expression, below 1 fmol/ug,<br />

similar to HER2 non-amplified cell lines. This suggests that MS<br />

quantitation can identify patients with low expression of HER2 who<br />

are unlikely to respond to trastuzumab therapy. As a matter of fact, 3<br />

of these 5 low expressing patients had outcome data and showed no<br />

response to trastuzumab treatment.<br />

In 28 of 31 patient tissue samples, HER3 showed low levels of<br />

expression (100-300 amol/ug tumor tissue) similar to HER3<br />

expression in cell lines, and comparable to low expressing EGFR<br />

and HER2 cell lines. The remaining 3 patients had no detectable<br />

HER3. This study demonstrates the feasibility of measuring HER3<br />

in multiplex in FFPE breast cancer tissue. Based on the low but<br />

widespread expression of HER3 in this cohort, it may be most useful<br />

to assess HER3 expression after initial treatment as a marker of<br />

potential resistance to targeted therapies.<br />

Taken together, these data demonstrate that a sensitive and quantitative<br />

assay to measure oncoproteins in FFPE clinical samples may help<br />

www.aacrjournals.org 185s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

stratify patients with variable expression of these targets. Our<br />

quantitation of oncogene expression from clinical samples uses a<br />

small amount of tissue, is clinically applicable and alleviates the<br />

problem of scoring either positive or negative for the expression of<br />

a given protein.<br />

P1-08-01<br />

Effects Of An Allosteric AKT Inhibitor (MK2206) Administered<br />

With Or Without The Aromatase Inhibitor Vorozole In An ER +<br />

Rat Mammary <strong>Cancer</strong> Model: Preventive And Therapeutic<br />

Effects<br />

Lubet RA, Ellis MJ, Grubbs CJ. National <strong>Cancer</strong> Institutes, Bethesda,<br />

MD; University of Washington at Saint Louis, MO; University of<br />

Alabama at Birmingham, AL<br />

The AKT inhibitors appear to hold some promise with regards to<br />

inhibition of ER + breast cancers which have relapsed. It has been<br />

hypothesized that the combination of an aromatase inhibitor plus an<br />

AKT inhibitor may be particularly effective. We, therefore, tested<br />

whether an allosteric AKT inhibitor (MK2206) alone or together<br />

with the aromatase inhibitor vorozole would be effective in a<br />

preventive and/or therapeutic setting. Female Sprague-Dawley rats<br />

were administered a single IV dose of methylnitrosourea (MNU)<br />

at 50 days of age. Beginning 5 days later, rats were administered<br />

the AKT inhibitor. In a preliminary study, two treatment regimens<br />

were employed: (1) dosing with inhibitor (100 mg/Kg BW/day, 7X/<br />

week) or (2) dosing with inhibitor (700 mg/Kg BW, 1X/week). These<br />

treatments minimally affected tumor multiplicity, but did significantly<br />

decrease tumor weights. We then examined the effects of combining<br />

the AKT inhibitor once weekly with a suboptimal dose of vorozole<br />

(0.12 mg/Kg BW/day) in a therapeutic setting. In a separate study,<br />

rats were treated with MNU and were allowed to develop their first<br />

palpable cancer. They were then treated with the combination of<br />

vorozole plus the AKT inhibitor or vehicle for 6 weeks. While tumors<br />

in vehicle treated rats grew an average of 600%, tumors treated with<br />

the inhibitor grew an average of only 95%; and, in fact, 5/12 showed<br />

a decrease in tumor size. Finally, we performed a similar combined<br />

study in a prevention setting. Dosing was: (a) AKT inhibitor (700 mg/<br />

Kg BW, 1X/week), (b) Vorozole (0.12 mg/Kg BW/day, 7X/week),<br />

(c) a plus b combined, and (d) vehicle. A, b, and c reduced tumor<br />

multiplicity by 26, 48 and 77%, respectively. Thus, combining an AKT<br />

inhibitor with an aromatase inhibitor showed synergy in prevention<br />

and treatment of ER + tumors.<br />

P1-08-02<br />

Gene expression changes in methylnitrosourea (MNU)-induced<br />

ER + mammary cancers following short-term treatment of rats<br />

with SERMs (Tamoxifen and Arzoxifene)<br />

Lubet R, Vedell P, Grubbs C, Bernard P, You M. National <strong>Cancer</strong><br />

Institute, Bethesda, MD; Medical School of Wisconsin, Milwaukee,<br />

WI; Hunstman <strong>Cancer</strong> Center, Salt Lake City, UT; University of<br />

Alabama at Birmingham, AL<br />

SERMs have proven to be highly effective in both therapy and<br />

prevention of ER + breast cancers. Tamoxifen has been the most<br />

commonly used SERM, but arzoxifene (another high- affinity<br />

competitive SERM agonist) showed strong activity in early clinical<br />

trials against ER + breast cancer; although further development was<br />

discontinued. Both SERMs have shown a dose dependent effect on<br />

prevention and therapy of rat mammary tumors in the ER + MNU<br />

model of cancer. Of interest, the doses required for prevention was<br />

significantly lower than the doses required for therapy. In the present<br />

study, rats bearing mammary cancers induced by MNU were treated<br />

with tamoxifen (0.66, 3.3, 20 and 100 ppm in diet) or arzoxifene (3.0<br />

ppm in diet) for 5 days. Global gene expression analysis showed<br />

that more than 100 genes were down-regulated and more than 100<br />

genes were up-regulated (p< 0.05 and fold change >1.5) in cancers<br />

treated with tamoxifen doses > 3 ppm; and that many of these gene<br />

changes were dose dependent. The genes modulated by tamoxifen<br />

and arzoxifene were enriched in the cell cycle pathway that were<br />

related to chromosome condensation in prometaphase [including<br />

Aurora-A, Aurora-B, Bub1B, non-SMC condensing I complex,<br />

subunit H (BRRN1), Condensin, CAP-G, CAP-G/G2, CAP-H/H2,<br />

CAP-D2/D3, CAP-E, TOP2, Cyclin A, Cyclin B, CDK1, Histone<br />

H1 and inter-centromere protein (INCENP]. Employing a different<br />

set of tamoxifen treated samples, we were able to confirm that many<br />

of the same genes were modulated employing a quantitative RT-<br />

PCR assay. Finally, we will compare certain of the gene changes<br />

obtained in the animal model with gene changes observed in human<br />

neoadjuvant trials.<br />

P1-09-01<br />

Long-term effect of tamoxifen use on the risk of contralateral<br />

breast cancer<br />

Mellemkjær L, Steding-Jessen M, Frederiksen K, Andersson M, Olsen<br />

JH. Danish <strong>Cancer</strong> Society Research Center, Copenhagen, Denmark;<br />

Copenhagen University Hospital, Rigshospitalet, Copenhagen,<br />

Denmark<br />

Background: Tamoxifen has been used widely in the treatment of<br />

breast cancer due to its beneficial effects on recurrences and mortality.<br />

Both randomized trials and observational studies give evidence that<br />

tamoxifen also reduces the incidence of contralateral breast cancer.<br />

The effect of tamoxifen seems to be present not only during treatment<br />

but also after termination of treatment, but the question is how long<br />

this effect lasts. Few studies have presented long-term follow-up<br />

data on risk of contralateral breast cancer after tamoxifen treatment.<br />

Material and Methods: In the Danish <strong>Breast</strong> <strong>Cancer</strong> Cooperative<br />

Group (DBCG) Database, we identified 50,323 women who had<br />

undergone surgery for invasive breast cancer and who had received<br />

treatment according to DBCG guidelines during 1978-2007. Among<br />

these women, 16,319 were users of tamoxifen while 34,004 were<br />

non-users. Contralateral breast cancer cases were identified in the<br />

Danish <strong>Cancer</strong> Registry and in the DBCG Database through 2009.<br />

The incidence of contralateral breast cancer among users of tamoxifen<br />

was compared to that of non-users by estimating incidence rate<br />

ratios (IRRs) in Cox regression analyses with adjustment for age at<br />

first breast cancer diagnosis, calendar period at first breast cancer<br />

diagnosis, histology of first breast cancer, radiotherapy, chemotherapy<br />

and other endocrine treatments besides tamoxifen.<br />

Results: During follow-up, 295 cases of contralateral breast cancer<br />

were observed among users of tamoxifen while 1,485 cases were<br />

observed among non-users resulting in an IRR of 0.82 (95% CI =<br />

0.71-0.94) after adjustments. Further analyses will include calculation<br />

of IRRs for contralateral breast cancer by time since breast cancer<br />

diagnosis, duration of tamoxifen and time since cessation of tamoxifen<br />

as well as calculation of IRRs after restriction to estrogen receptor<br />

positive breast cancer patients. Results of these analyses will be<br />

presented.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

186s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 1<br />

P1-09-02<br />

Pilot study of a 1-year intervention of high-dose vitamin D in<br />

women at high risk for breast cancer.<br />

Sivasubramanian PS, Hershman DL, Maurer M, Kalinsky K, Feldman<br />

S, Brafman L, Refice S, Kranwinkle G, Crew KD. Columbia University,<br />

New York, NY<br />

Background: Selective estrogen receptor modulators (SERMs)<br />

have been shown to decrease breast cancer incidence among highrisk<br />

women, but uptake for prevention has been poor. Observational<br />

studies have demonstrated that serum 25-hydroxyvitamin D (25-<br />

OHD) is inversely related to breast cancer risk, such that levels >40<br />

ng/ml are associated with about a 40% reduction in breast cancer risk<br />

compared to women who are vitamin D deficient (25-OHD 5 years) were assigned to a 1-year intervention of<br />

vitamin D3 20,000 or 30,000 IU weekly. Other eligibility criteria<br />

included baseline mammographic density (MD) ≥25%, serum 25-<br />

OHD ≤32 ng/ml, no current SERM use and no history of kidney<br />

stones. Women underwent a digital mammogram at baseline and<br />

12 months, and serial blood draws every 3 months. In addition,<br />

random core breast biopsies were conducted in premenopausal<br />

women, whereas postmenopausal women underwent a breast MRI<br />

at baseline and 12 months. Participants were monitored for toxicity,<br />

particularly hypercalcemia and hypercalciuria, every 3 months. The<br />

primary objective is to determine the safety and feasibility of highdose<br />

vitamin D in this study population. Secondary objectives are<br />

to determine changes in breast density and blood-based biomarkers<br />

(25-OHD, 1,25(OH)D, PTH, IGF-I, IGFBP-3). Serum 25-OHD was<br />

measured by Diasorin radioimmunoassay.<br />

Results: From November 2007 to January 2011, 292 women were<br />

screened and 142 were ineligible. Main reasons for ineligibility (%)<br />

included 25-OHD >32 ng/ml (27), opted for SERM (23), prior kidney<br />

stones (11), and MD 0.37) and 1 withdrew due to dyspepsia. Mean serum 25-OHD<br />

rose to 47 ng/ml at 3 months, 49.1 ng/ml at 6 months, and 53.7 ng/ml<br />

(range, 26-77) at 12 months. No significant hypercalcemia (serum Ca<br />

>10.5 mg/dl) occurred at either dose level. Imaging and biomarker<br />

analyses are ongoing.<br />

Discussion: We have demonstrated that a 1-year intervention of<br />

high-dose vitamin D3 is well tolerated and can increase serum 25-<br />

OHD above a target level of 40 ng/ml. This preliminary data has<br />

informed an ongoing phase IIb randomized placebo-controlled trial<br />

(SWOG 0812) of high-dose vitamin D in 200 high-risk premenopausal<br />

women and highlights the importance of early phase breast cancer<br />

chemoprevention trials with intermediate biomarker endpoints to<br />

test novel agents.<br />

P1-09-03<br />

Chemoprevention in patients with newly diagnosed breast cancers<br />

Refinetti AP, Chun J, Schnabel F, Guth A, Axelrod D. NYU Langone<br />

Medical Center, New York, NY<br />

Background<br />

Chemoprevention (including tamoxifen, raloxifene, and exemestane)<br />

is a strategy to reduce breast cancer incidence in high risk women.<br />

Studies have shown at least a 50% decrease in the incidence of breast<br />

cancer in users of these drugs. Despite this benefit, the majority<br />

of high risk, unaffected women who are offered chemoprevention<br />

decline the therapy. However, there is a growing population of women<br />

who have used these agents for primary prevention, and a larger<br />

population of survivors who have used these drugs as part of their<br />

systemic treatment. The purpose of this study was to identify a cohort<br />

of women with newly diagnosed breast cancers who had utilized<br />

chemoprevention and describe their patterns of disease.<br />

Methods<br />

The <strong>Breast</strong> <strong>Cancer</strong> Database of NYU Langone Medical Center was<br />

queried for patients who used chemopreventive drugs and developed<br />

breast cancer between 1/2010-1/2012. Patients were divided into<br />

primary and secondary chemoprevention groups (no previous history<br />

of breast cancer and previous history of breast cancer, respectively).<br />

Descriptive statistics were utilized.<br />

Results<br />

In the study period, 24 (2%) of 996 patients had used a chemopreventive<br />

agent. For 16 of the 24 (67%), the drug was part of systemic therapy<br />

for prior breast cancer, with a median of 12 years from the initial<br />

diagnosis to the diagnosis of a second breast cancer. The primary<br />

chemoprevention group included women with risk based on family<br />

history and atypical hyperplasias. The majority of patients were<br />

diagnosed with early stage disease (88% DCIS and stage I). This<br />

likely reflects their screening behaviors. In both groups, the majority<br />

of cancers were ductal in origin. Five of the 8 patients in the primary<br />

chemoprevention group were on treatment at the time of their cancer<br />

diagnosis, while 63% of patients in the secondary group were prior<br />

users. In the secondary group, the majority of cases were contralateral<br />

second primary breast cancers, with 31% ipsilateral recurrences.<br />

Interestingly, the majority of cancers in both groups were ER/PR<br />

positive.<br />

Clinical Characteristics<br />

VARIABLES TOTAL N=24 (%) PRIMARY N=8 (%)<br />

SECONDARY N=16<br />

(%)<br />

FHBC<br />

Positive 14 (58) 5 (63) 9 (56)<br />

Negative 10 (42) 3 (37) 7 (44)<br />

AH, LCIS<br />

AH 6 (25) 5 (63) 1 (6)<br />

LCIS 2 (8) 2 (25) 0 (0)<br />

HISTOLOGY<br />

DCIS 9 (38) 5 (63) 4 (25)<br />

IDC 13 (54) 2 (25) 11 (69)<br />

ILC 0 (0) 0 (0) 0 (0)<br />

Mixed 2 (8) 1 (12) 1 (6)<br />

ER<br />

Positive 19 (79) 7 (88) 12 (75)<br />

Negative 5 (21) 1 (12) 4 (25)<br />

PR<br />

Positive 15 (63) 6 (75) 9 (56)<br />

Negative 9 (37) 2 (25) 7 (44)<br />

CHEMOPREVENTION TYPE<br />

Tamoxifen 17 (71) 4 (50) 13 (81)<br />

Raloxifene 5 (21) 4 (50) 1 (6)<br />

AI 2 (8) 0 (0) 2 (13)<br />

CHEMOPREVENTION DURATION<br />


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

will need to be modified in light of prior hormonal treatment. Many<br />

of the patients in the secondary group were past users of prevention<br />

agents and further work is needed to clarify the duration of benefit<br />

of these drugs. In a similar vein, we look forward to research efforts<br />

to define the optimal age to initiate primary chemoprevention in<br />

high risk women.<br />

P1-09-04<br />

Down-regulation of trefoil protein 1(TFF1) in normal breast<br />

tissue of postmenopausal women at increased risk for breast<br />

cancer on exemestane<br />

Gatti-Mays M, Kallakury BVS, Makariou E, Venzon D, Permaul<br />

E, Isaacs C, Cohen P, Warren R, Gallagher A, Eng-Wong J.<br />

Georgetown University Hospital, Washington, DC; National <strong>Cancer</strong><br />

Institute, National Institutes of Health, Bethesda, MD; Lombardi<br />

Comprehensive <strong>Cancer</strong> Center, Georgetown University Medical<br />

Center, Washington, DC; Lombardi Comprehensive <strong>Cancer</strong> Center,<br />

Georgetown University Hospital, Washington, DC<br />

Background: Aromatase inhibitors (AI) are effective for breast cancer<br />

risk reduction in postmenopausal women. TFF1, also known as pS2,<br />

is an estrogen response gene present in normal mammary tissue with<br />

increased expression in estrogen receptor positive breast cancer.<br />

Previous studies have demonstrated down-regulation of TFF1 and Ki-<br />

67, a marker of proliferation, in postmenopausal women with locally<br />

advanced breast cancer who receive neoadjuvant AIs. TFF1 and<br />

proliferating cell nuclear antigen (PCNA) may serve as biomarkers<br />

of effect of AIs in women at increased risk for breast cancer.<br />

Methods: We conducted a single-arm phase II trial of exemestane<br />

in women at increased risk for breast cancer and examined the<br />

impact on TFF1 and PCNA. Postmenopausal women at increased<br />

risk for invasive breast cancer by clinical or histological criteria<br />

received 25mg of exemestane daily for 2 years. Subjects were<br />

required to have stopped any hormonal medication ≥ 3 months prior<br />

to enrollment. Image guided breast biopsies targeting dense breast<br />

tissue were performed at baseline and at 12 months. Core specimens<br />

were obtained under local anesthesia at each time point from the<br />

same area of the breast. One core biopsy sample was formalin-fixed,<br />

paraffin-embedded and examined for pathologic abnormalities, as<br />

well as TFF1 and PCNA. TFF1 was assessed by intensity of stain (0<br />

to 3+) and percent of cells with any staining; PCNA was assessed<br />

by percent of cells staining within the tissue section. The pathologist<br />

(B.K.) was blinded to the time of biopsy. Change in intensity and %<br />

positive cells were evaluated by paired t-test.<br />

Results: Thirty four subjects underwent both baseline & 12 month<br />

breast biopsies. Eight biopsies at baseline and 5 biopsies at 12 months<br />

did not contain any ductal or lobular tissue and were not analyzed.<br />

Twenty-two subjects had evaluable breast tissue at both time points<br />

for TFF1 analysis and 23 subjects for PCNA analysis. No high risk<br />

lesions or invasive cancers were identified. Of the baseline specimens,<br />

95.5% were positive for TFF1: 59.1% (13 of 22) were scored as<br />

3+(intense), 31.8% (7 of 22) were 2+(moderate) and 4.5% (1 of 22)<br />

were 1+(low). Percent of cells staining for TFF1 ranged from 0 to 20%<br />

(median=1%). After 1 year on exemestane TFF1 intensity decreased<br />

in 17 subjects (77.3%), 4 had no change and 1 increased. Mean TFF1<br />

change was -1.32 (95% CI -1.87 to -0.76; p < 0.001). The change in<br />

% positive cells for PCNA ranged from -15 to +30% (median = 0%).<br />

Discussion: Assessing tissue biomarkers with repeat core needle<br />

biopsies in a phase II prevention trial in high risk women is feasible.<br />

Since prevention agents are not universally protective, determining<br />

biomarkers of effect may allow tailored therapy. TFF1 is a biologically<br />

plausible biomarker of AI activity that was down-regulated in 77% of<br />

breast tissue following exemestane therapy. This is the first study to<br />

evaluate this tissue marker in the prevention setting. Further study is<br />

needed to correlate with other biomarkers of interest, e.g. change in<br />

mammographic density, serum hormone levels and clinical outcomes.<br />

P1-09-05<br />

The RAZOR trial: a phase II prevention trial of screening plus<br />

goserilin and raloxifene versus screening alone in pre-menopausal<br />

women at increased risk of breast cancer.<br />

Motion J, Ashcroft L, Dowsett M, Cuzick J, Hickman J, Evans G,<br />

Eccles D, Eeles R, Greenhalgh R, Affen J, Bundred S, Boggis C,<br />

Sergeant J, Fallowfield L, Adams J, Howell A. University Hospital<br />

South Manchester, Manchester, United Kingdom; The Christie NHS<br />

Foundation Trust, Manchester, United Kingdom; Royal Marsden<br />

Hospital, London, United Kingdom; Queen Mary, University of<br />

London, United Kingdom; Princess Anne Hospital, Southampton,<br />

United Kingdom; The Institute of <strong>Cancer</strong> Research, Royal Marsden<br />

Hospital, London, United Kingdom; University of Sussex -<br />

(SHORE-C), Brighton, United Kingdom; University of Manchester,<br />

United Kingdom; Manchester Royal Infirmary, Manchester, United<br />

Kingdom<br />

Background: Observational studies indicate that oophorectomy at<br />

about age 40 reduces breast cancer risk by approximately a half in<br />

high risk women. Widespread use of risk reducing oophorectomy is<br />

unlikely to be acceptable to these women. We explored the feasibility<br />

of giving goserelin to produce reversible ovarian suppression together<br />

with raloxifene to maintain bone mineral density (BMD). Objectives:<br />

The primary study objective was adherence to treatment. Secondary<br />

objectives were uptake of randomisation, side effects/quality of life<br />

and measures of effect on bone and in serum. Methods: Recruitment<br />

was from 3 UK Family History Clinics. Consenting women at ≥ 1 in 3<br />

lifetime risk of breast cancer were randomised to control or monthly<br />

subcutaneous goserelin 3.6 mg and raloxifene 60 mg/d orally for two<br />

years. Questionnaires (Endocrine Symptom Checklist, Trait & State<br />

Anxiety, Sexual Activity & <strong>Cancer</strong> Worry) measuring toxicity/quality<br />

of life were administered by nurses. Dual energy X-ray absorptiometry<br />

(DXA) BMD measurements were performed in the treatment arm<br />

annually. Lipids and collagen breakdown products were measured by<br />

standard methods. Results: 75 of 511 (14.7%) women approached<br />

agreed to randomisation (38 to treatment and 37 to control). The<br />

major reason for non-entry was fear of side effects (85%). Median<br />

age was 37 and 35 years, for the experimental (A) and control arm<br />

(B), respectively. Median follow up is 8.8 years. 20/38 in arm A and<br />

27/37 of controls completed the 24 m study. 18/38 women in arm A<br />

withdrew (13 [34%] because of side effects) and 10/37 in arm B for<br />

various reasons including the desire for risk reducing surgery (n=4).<br />

No significant differences were seen in the Endocrine Symptom Subscale,<br />

State or Trait anxiety or <strong>Cancer</strong> Worry. However, Hot flushes,<br />

night and cold sweats (together p


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 1<br />

significant symptoms and bone loss, thus methods to ameliorate these<br />

need to be developed if ovarian suppression is to play a role in breast<br />

cancer prevention.<br />

P1-09-06<br />

Biological Effects of Green Tea Capsule Supplementation in Presurgery<br />

<strong>Breast</strong> <strong>Cancer</strong> Patients<br />

Yu S, Spicer D, Hawes D, Wu A. University of Southern California,<br />

Los Angeles, CA<br />

Green tea is rich in antioxidant polyphenols consisting primarily<br />

of epicatechins, epigallocatechin, epicatechin gallate, and<br />

epigallocatechin gallate ( EGCG). There are suggestive evidence<br />

from observational epidemiologic studies that regular green tea<br />

consumers may have lower risk of breast cancer development and<br />

mortality. Inhibition of cell proliferation and angiogenesis and<br />

enhancement of apoptosis are some of the mechanisms that have been<br />

proposed to explain the anti-cancer actions of the bioactive extracts<br />

of green tea. We conducted a randomized pre-surgical pilot study<br />

of green tea capsules in women diagnosed with breast cancer at the<br />

Los Angeles County-University Southern California Medical Center.<br />

Women in both the green tea and control arms were non-green tea<br />

drinkers at baseline ( i.e. less than 1 cup of green tea per week at the<br />

time of breast cancer diagnosis). Women (n=17) who participated<br />

in the green tea arm took 3 green tea capsules daily (Pro Health<br />

Green Tea Mega EGCG, 3 capsules contained ∼ 870 mg EGCG;<br />

equivalent to ∼ 8 cups of green tea) between their breast cancer<br />

diagnosis until their scheduled surgery date. The average duration<br />

of green tea intake was 35 days. Women in the control arm did not<br />

drink green tea between diagnosis and surgery date. Participants in<br />

the green tea and control groups donated blood and urine specimens<br />

at enrollment and before surgery which were tested for urinary tea<br />

catechin levels as a measure of compliance. Baseline urinary tea<br />

catechin concentrations were low in both green tea and control arms.<br />

Tea catechin concentrations remained low for women in the control<br />

arm but increased significantly ( 2-10 folds for different catechins) for<br />

women in the green tea arm. The differences in pre-surgery urinary<br />

green tea levels were significantly different between these groups of<br />

participants We conducted standard immunohistochemical analysis<br />

of the formalin-fixed paraffin-embedded respective markers of cell<br />

proliferation (Ki67), apoptosis (BAX), and angiogenesis (CD34).<br />

Separate analysis was performed on benign and malignant breast<br />

cancer tissue. The changes in these three biomarkers pre- and posttreatment<br />

in the two groups will be presented.<br />

Supported by the WHH Foundation.<br />

P1-09-07<br />

Topical 4-OHT trial in women with DCIS of the breast: report of<br />

plasma and breast tissue concentration of tamoxifen metabolites<br />

Lee O, Chatterton RT, Muzzio M, Page K, Jovanovic B, Helenowski I,<br />

Dunn B, Heckman-Stoddard B, Foster K, Shklovskaya J, Skripkauskas<br />

S, Bergan R, Khan SA. Northwestern University, Chicago, IL; IIT<br />

Research Institute, Chicago, IL; National <strong>Cancer</strong> Institute, Bethesda,<br />

MD<br />

Background: Earlier studies have shown that 1-2mg of<br />

4-hydroxytamoxifen (4–OHT) gel applied to the breast skin reduced<br />

cell proliferation in estrogen receptor (ER) positive invasive cancers<br />

to a similar degree as oral tamoxifen (TAM), with significantly lower<br />

plasma levels. We now report results of a Phase IIB pre-surgical<br />

window trial of women with DCIS, designed to obtain pilot data in<br />

early lesions. Our ultimate goal is to develop transdermal 4-OHT as an<br />

alternative to oral TAM for women at high risk for breast cancer and<br />

those with DCIS. The study was closed early because the manufacturer<br />

discontinued the drug supply, but remains blinded until all biomarker<br />

analysis is complete. Here we report the plasma and breast adipose<br />

tissue concentration of TAM metabolites from the topical 4-OHT<br />

gel group (4 mg) in comparison with the oral TAM group (20mg).<br />

Methods: Women with DCIS were enrolled, and randomized to<br />

4-OHT gel (4mg/day, 2mg per breast, E: Z isomers =1:1,) or to oral<br />

(Z) TAM (20mg/day) for 4-10 weeks before surgery. Blood was<br />

collected on the day of surgery, and breast adipose tissue was collected<br />

at surgery. There were a total of 22 patients with matched blood and<br />

breast adipose tissue. The concentration of TAM metabolites in plasma<br />

and breast tissue was determined with liquid chromatography/tandem<br />

mass spectrometry. We assumed that the subjects with detectable<br />

N-desmethyl TAM (NDT) in plasma belong to the oral TAM group<br />

because NDT is not a product of 4-OHT metabolism. Under this<br />

assumption, 13 subjects were categorized into oral TAM group, and<br />

9 subjects into the topical 4-OHT group. Wilcoxon rank-sum test was<br />

used for statistical analysis.<br />

Results: The results are shown in the table. The concentration is<br />

presented as mean ± SD; the lowest quantitation limit (LQL) was 1<br />

ng/mL for plasma, and 3 ng/g for tissue. TAM and its metabolites<br />

were found in the plasma of the presumed oral TAM group, with high<br />

levels of TAM and NDT. In the presumed 4-OHT gel group, only (Z)<br />

4-OHT was found in the plasma although both (E) and (Z) forms were<br />

applied. The mean plasma level of 4-OHT in the gel group was 70%<br />

lower than the mean of 4-OHT in the oral TAM group (p=0.004).<br />

In breast tissue, similar amounts of (E) and (Z) forms of 4-OHT<br />

were found in the 4-OHT gel group, with the (Z) 4-OHT level being<br />

equivalent to that in the oral TAM group (p=0.48). Endoxifen was<br />

only found in the oral TAM group. We saw no evidence of further<br />

metabolic transformation of 4-OHT in the breast following topical<br />

administration.<br />

Compounds measured Plasma, ng/mL<br />

<strong>Breast</strong> tissue, ng/g<br />

Oral TAM(n=13) 4-OHT gel(n=9) Oral TAM(n=13) 4-OHT gel(n=9)<br />

(Z) TAM 92.8 ± 46.3


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

expression of MMP-9. MMP-9 mRNA levels were analyzed by realtime<br />

PCR. NF-κB and AP-1 DNA binding was analyzed by EMSA.<br />

Results: Our results showed that curcumin inhibits TPA-induced<br />

MMP-9 expression and cell invasion through suppressing NF-κB<br />

and AP-1 activation. Curcumin strongly repressed the TPA-induced<br />

phosphorylation of p38 and JNK and also inhibited TPA-induced<br />

translocation of PKCa from the cytosol to the membrane, but did not<br />

affect the translocation of PKCδ.<br />

Conclusion: It is concluded that curcumin inhibits the TPA-induced<br />

MMP-9 expression and cell invasion through the suppression of<br />

the PKCα/MAPK/NF-kB/AP-1 pathway in MCF-7 breast cancer<br />

cells. Accordingly, curcumin may have the therapeutic potential in<br />

restricting breast cancer metastasis.<br />

P1-10-02<br />

Comparative Preventive Efficacy of Select Chinese Herbs in<br />

<strong>Breast</strong> Carcinoma Derived Isogenic Cells with Modulated<br />

Estrogen Receptor Functions<br />

Telang NT, Li G, Katdare M, Sepkovic DW, Bradlow HL, Wong<br />

GYC. Palindrome Liaisons, Montvale, NJ; American Foundation<br />

for Chinese Medicine, New York, NY; Skin of Color Research<br />

Institute, Leroy T. Canoles Jr. <strong>Cancer</strong> Research Center, Hampton<br />

University, Eastern Virginia Medical School, Hampton, VA; David<br />

& Alice Jurist Institute for Research, Hackensack University Medical<br />

Center, Hackensack, NJ; David & Alice Jurist Institute for Research,<br />

Hackensack, NJ; American Foundation for Chinese Medicine, Beth<br />

Israel Medical Center, New York, NY<br />

Background: Human mammary carcinoma derived estrogen receptor<br />

positive (ER + ) MCF-7 cells depend on 17β-estradiol (E 2 ) for cellular<br />

proliferation in vitro and for tumor development in vivo. Traditionally,<br />

ER functions as a ligand activated nuclear transcription factor with E 2<br />

as its physiological ligand, and limited availability of E 2 modulates<br />

ER functions. We have recently demonstrated preventive efficacy of<br />

selected Chinese nutritional herbs in this MCF-7 cell culture model.<br />

The present study screened additional Chinese nutritional herbs for<br />

their preferential preventive efficacy on MCF-7 cells with modulated<br />

ER functions.<br />

Methods: MCF-7 cells were maintained in medium supplemented<br />

with either 0.7% serum,


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 1<br />

Conclusion:<br />

<strong>Breast</strong> cancer incidence is significantly reduced by both physical<br />

exercise and weight loss, especially in the postmenopausal population.<br />

Intensity, timing and nature of the exercise did not significantly alter<br />

the protective effect.<br />

P1-12-01<br />

Evaluation on efficacy and safety of capecitabine plus docetaxel<br />

versus docetaxel monotherapy in metastatic breast cancer patients<br />

pretreated with anthracycline: Results from a randomized phase<br />

III study (JO21095)<br />

Sato N, Yamamoto D, Rai Y, Iwase H, Saito M, Iwata H, Masuda<br />

N, Oura S, Watanabe J, Kuroi K. Niigata <strong>Cancer</strong> Center Hospital,<br />

Niigata, Japan; Kansai Medical University Hirakata Hospital,<br />

Hirakata, Osaka, Japan; Sagara Hospital, Kagoshima, Japan;<br />

Kumamoto University Hospital, Kumamoto, Japan; Juntendo<br />

University Hospital, Bunkyo, Tokyo, Japan; Aichi <strong>Cancer</strong> Center<br />

Hospital, Nagoya, Aichi, Japan; National Hospital Organization<br />

Osaka National Hospital, Osaka, Japan; Wakayama Medical<br />

University, Wakayama, Wakayama, Japan; Shizuoka <strong>Cancer</strong> Center,<br />

Nagaizumi-cho, Suntou-gun, Shizuoka, Japan; Tokyo Metropolitan<br />

<strong>Cancer</strong> and Infectious Diseases Center Komagome Hospital, Bunkyo,<br />

Tokyo, Japan<br />

Introduction: A previous large-scale phase III study demonstrated<br />

that, compared with docetaxel (T) alone, capecitabine (X) and T in<br />

combination (XT) offered significantly superior progression free<br />

survival (PFS) and overall survival (OS) in metastatic breast cancer<br />

(MBC). However, XT increased Grade 3/4 adverse events (AEs)<br />

which led to more frequent dose reductions than with T alone. Optimal<br />

dose of XT in Japanese was examined in a phase Ib study. Based<br />

on the background, we conducted a phase III randomized study in<br />

Japanese HER2 negative MBC patients pre-treated with anthracycline<br />

to compare efficacy and safety of XT therapy and T therapy.<br />

Methods: Eligible pts were HER2-negative MBC pts with<br />

anthracycline-pretreatment, a measurable tumor, and ECOG<br />

performance status of 0 or 1. Pts were randomly assigned to the XT<br />

group or the T→X group. The XT group received concurrent therapy<br />

of X (1650 mg/m 2 /day from day 1 to 14) and T (60 mg/m 2 ) in 3-week<br />

cycle. The T→X group received sequential therapy of T (70 mg/m 2 )<br />

in 3-week cycle followed at disease progression by X (2500 mg/m 2 /<br />

day from day 1 to 14 followed by 1-week rest). Primary endpoint<br />

was PFS. Secondary endpoints were OS, overall response rate (ORR),<br />

time to treatment failure (TTF), safety, and quality of life. The XT<br />

group and the T phase of the T→X group (T group) were compared<br />

in our evaluation.<br />

Results: Of 163 pts enrolled, 156 were eligible. Baseline<br />

characteristics of all pts in each group were well balanced. The<br />

median delivered dose was 79.0% and 95.1% of the planned dose<br />

respectively for X and T in the XT group, and it was 97.2% in the T<br />

group. Median PFS in the XT group was 10.5 months compared to<br />

9.8 months in the T group (hazard ratio [HR], 0.62; 95% confidence<br />

interval [CI], 0.40–0.97). The ORR was 70% and 61%; the median<br />

TTF was 9.6 months and 7.0 months in the XT group and the T<br />

group, respectively. Median OS has not been reached yet. Subgroup<br />

analysis showed PFS was longer in pts with liver metastasis (HR,<br />

0.39; 95% CI, 0.19–0.84) and in pts with lung metastasis (HR, 0.43;<br />

95% CI, 0.21–0.90) in the XT group. Incidence of treatment related<br />

AEs (TR-AEs) ≥Grade 3 was 74.4% (61 pts) in the XT group and<br />

76.3% (61 pts) in the T group. Frequently reported TR-AEs ≥Grade 3<br />

were; decrease in neutrophil count (XT, 57.3%; T, 60.0%), neutropenia<br />

(XT, 8.5%; T, 12.5%) and febrile neutropenia (XT, 6.1%; T, 10.0%).<br />

TR-AE ≥Grade 3 in the XT group with incidence at least 5% higher<br />

than the T group was hand-foot syndrome (XT, 7.3%; T, 0%). On the<br />

other hand, TR-AEs ≥Grade 3 in the T group with incidence at least<br />

5% higher than the XT group were fatigue (XT, 2.4%; T, 8.8%) and<br />

peripheral edema (XT, 1.2%; T, 6.3%).<br />

Conclusion: The concurrent therapy of XT demonstrated significant<br />

improvement of PFS compared with T alone. Superior efficacy of XT<br />

therapy was reported as same as the previously reported study on<br />

XT versus T although the dose was lower in our study. Considering<br />

the efficacy and tolerability, we consider concurrent Japanease dose<br />

XT therapy is a preferable treatment for MBC pts with liver or lung<br />

metastasis.<br />

P1-12-02<br />

Results of a phase 2, multicenter, single-arm study of eribulin<br />

mesylate as first-line therapy for locally recurrent or metastatic<br />

HER2-negative breast cancer<br />

Vahdat L, Schwartzberg L, Glück S, Rege J, Liao J, Cox D,<br />

O’Shaughnessy J. Weill Cornell Medical College, New York, NY; The<br />

West Clinic, Memphis, TN; Sylvester Comprehensive <strong>Cancer</strong> Center,<br />

Miami, FL; Eisai Inc, Woodcliff Lake, NJ; Texas Oncology-Baylor<br />

Charles A. Sammons <strong>Cancer</strong> Center, Dallas, TX<br />

Background: Eribulin mesylate is a novel nontaxane microtubule<br />

dynamics inhibitor that is approved in patients (pts) with metastatic<br />

breast cancer (MBC) who have previously received at least two<br />

chemotherapeutic regimens for MBC. We are reporting a preliminary<br />

planned analysis (first 6 cycles of treatment) of a phase 2 study that<br />

evaluates efficacy and safety of eribulin as first-line therapy for<br />

HER2-negative MBC.<br />

Methods: Pts with measureable HER2-negative locally recurrent or<br />

MBC with at least 12 months since prior neoadjuvant or adjuvant<br />

chemotherapy received eribulin mesylate at 1.4 mg/m 2 IV on days<br />

1 and 8 of each 3-week cycle. Endpoints include objective response<br />

rate (ORR) (primary), safety, progression free survival (PFS), time to<br />

response (TTR), and duration of response (DOR). Tumor assessments<br />

were evaluated every 6 weeks for the first 6 cycles and every 6-12<br />

weeks thereafter per RECIST 1.1.<br />

Results: 54 of 56 enrolled pts had at least 1 post-baseline assessment.<br />

The median number of cycles delivered was 7 (range 1,21). Pt<br />

characteristics: median age, 56 yrs (range 31-85); ECOG of 0, 32<br />

(57%); 29% had de novo stage IV; 38/56 (68%) had prior neo/adjuvant<br />

(45% had anthracycline and 45% taxane, the majority of which were<br />

given together). Seventy percent have visceral disease (45% liver,<br />

38% lung); 41 (73%) have estrogen receptor-positive (ER) disease<br />

and 12 (21%) have triple negative (TN) (ER-/PR-/HER2-) disease.<br />

Objective response data are found in Table 1.<br />

Table 1. Summary of Efficacy<br />

Efficacy<br />

ALL, n (%) (N=54) ER+, n (%) (N=40) ER-/PR-/HER2-, n (%) (N=11)<br />

ORR 17 (31) 13 (32) 4 (36)<br />

CR 0 0 0<br />

PR 17 (31) 13 (32) 4 (36)<br />

SD 26 (48) 21 (53) 4 (36)<br />

CBR (ORR+ ≥6<br />

mo SD)<br />

26 (48) 22 (55) 4 (36)<br />

Median months<br />

TTR 1.4 (1.2,2.7) 1.4 (1.3,2.7) 2.0 (1.1,5.6)<br />

DOR 5.8 (4.6,NE) 7.4 (3.3,NE) 5.8 (4.7,NE)<br />

PFS 6.1 (4.1,7.4) 6.8 (4.6,8.5) 3.5 (1.2,NE)<br />

3 pts were ER-/PR+ with no objective response (1 SD, 2 PD).<br />

www.aacrjournals.org 191s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

The most common treatment-related AEs are reported in Table 2.<br />

Table 2. Percentage of treatment-related adverse events<br />

Adverse Event (N=56) All events (%) Grade 3/4 (%)<br />

Leading to study drug discontinuation 9 9<br />

Leading to dose reduction 32 25<br />

Common AEs (≥25%)<br />

Alopecia 79 NA<br />

Neutropenia 66 48<br />

Fatigue 52 2<br />

Peripheral neuropathy 52 16<br />

Nausea 45 0<br />

Anemia 38 0<br />

Treatment-related serious AEs occurred in 5 (9%) pts, neutropenia<br />

(4%), and febrile neutropenia (5%). Five pts stopped study therapy<br />

due to adverse events. Fourteen pts remain on study treatment.<br />

Conclusions: The preliminary results of this first-line study suggest<br />

that eribulin has antitumor activity in ER+/HER2- and TN MBC with<br />

an acceptable safety profile. Further exploration of this treatment as<br />

part of neo/adjuvant therapy is warranted.<br />

P1-12-03<br />

Low-dose capecitabine monotherapy in HER-2 negative<br />

metastatic breast cancer: a retrospective study.<br />

Ambros T, Zeichner SB, Zaravinos J, Montero AJ, Ahn E, Mani A,<br />

Kronish L, Mahtani RL, Vogel CL. University of Miami, FL; Mount<br />

Sinai Medical Center, Miami Beach, FL; Memorial Hospital West,<br />

Pembroke Pines, FL; SUNY Downstate, Brooklyn, NY<br />

Background: Capecitabine (CAPE) has clinical activity in metastatic<br />

breast cancer (MBC) at an FDA approved dose of 1,250mg/m 2 twice<br />

daily for 14 days every 21 days (BID Q14). This schedule has been<br />

adopted by many United States oncologists, although the starting<br />

dose is often reduced. To determine if lower doses of CAPE have<br />

comparable clinical activity as the FDA approved dose, we performed<br />

a retrospective analysis. Methods: A retrospective review of records<br />

from a large breast cancer oncology practice and from Sylvester<br />

<strong>Cancer</strong> Center Deerfield Beach from 2000-2008 was performed after<br />

approval of the University of Miami IRB. Standard practice was<br />

CAPE low dose (CAPE-L) 1,000mg BID Q14. Primary outcome was<br />

clinical benefit rate (CBR) defined as complete response (CR), partial<br />

response (PR) or stable disease (SD) lasting for ≥ 6 months. Response<br />

rates (RR) in patients with measurable disease, progression free<br />

survival (PFS), overall survival (OS) defined as time period between<br />

beginning of CAPE and death, and adverse events (AE) defined<br />

according to the NCI CTCAE version 3.0 were secondary endpoints.<br />

A literature review was performed for comparison. Results: Data<br />

from 296 patients (pts) with HER-2 negative MBC were reviewed.<br />

Of those, 73 received CAPE-L at a starting dose between 303 mg/<br />

m 2 and 965 mg/m 2 (median 614 mg/m 2 ) BID Q14. Median number<br />

of prior lines of therapy was 1 (range 0 - 10); 34/73 (46.6%) of pts<br />

received CAPE-L as first line therapy. 23.3% of pts required dose<br />

reductions because of palmar-plantar erythrodysesthesia (PPE) (44%),<br />

diarrhea (17%), mucositis (11%) or other (28%). RR in 61 patients<br />

with measurable disease was 25%. PR occurred in 16/73 (22%),<br />

CR (0%), SD ≥ 6 months 21/73 (29%), and PD 31/73 (43%), with a<br />

CBR of 37/73 (51%). Median PFS and OS were 6.2 months (95% CI,<br />

4.4 to 8) and 21.4 months (95% CI, 14.4 to 28.6), respectively. AEs<br />

are reported in table 2. We recognized 12 trials that used the FDA<br />

approved dose in 1,949 patients. Bidimensionally measurable disease<br />

was present in 1,630 patients. CBR was reported as 62% in 1,006<br />

patients and RR as 24% in 398. Weighted averages of median PFS<br />

and OS were 5.1 months (95% CI, 4.5 to 5.7) and 12 months (95%<br />

CI, 9.6 to 14.4), respectively. Detailed toxicity data were available<br />

in 11 trials with 1,883 patients. A comparison of current series with<br />

literature review at standard dose follows (table 1). Conclusion:<br />

Compared with previously published data, CAPE-L appeared more<br />

tolerable with comparable clinical efficacy as package insert doses.<br />

Efficacy<br />

Current Series<br />

Literature<br />

CBR (%) 37/73 51 1006/1630 62<br />

RR (%) 15/61 25 398/1630 24<br />

Median prior line<br />

of Rx (range)<br />

1 (1 -10) 2 a (0 - 6)<br />

Median PFS (mo)<br />

6.2 (4.4 - 8)<br />

(95% CI)<br />

5.1 (4.5 - 5.7) b<br />

Median OS (mo)<br />

(95% CI)<br />

21.4 (14.4 - 28.6) 12 (9.6 - 14.4) c<br />

a<br />

n=759, 6 trials (n=1190) did not report individual data. b n=1949. c n=1411, 2 trials (n=538) did not<br />

reach 50% mortality.<br />

Adverse Events<br />

Current Series<br />

Literature (n=1883)<br />

n % n %<br />

PPE<br />

GI-II 20 27.4 709 37.7<br />

GIII-IV 4 5.5 215 11.4<br />

Diarrhea<br />

GI-II 12 16.4 509 27<br />

GIII-IV 4 5.5 193 10.2<br />

Mucositis<br />

GI-II 7 9.6 212 11.2<br />

GIII-IV 0 0 36 1.9<br />

Fatigue<br />

GI-II 12 16.4 343 18.2<br />

GIII-IV 0 0 57 3<br />

P1-12-04<br />

Carboplatin, nab-paclitaxel and bevacizumab as first-line<br />

treatment for metastatic breast cancer.<br />

Lo SS, Guo R, Czaplicki KL, Robinson PA, Gaynor E, Barhamand<br />

FB, Schulz WC, Kash JJ, Horvath LE, Bayer RA, Petrowsky C, De<br />

la Torre R, Park JH, Albain KS. Loyola University Medical Center,<br />

Maywood, IL; Hematology Oncology Consultants Ltd., Naperville,<br />

IL; Swedish Americal Regional <strong>Cancer</strong> Center, Rockford, IL; Edward<br />

<strong>Cancer</strong> Center, Naperville, IL; Central Dupage <strong>Cancer</strong> Center,<br />

Winfield, IL; CDPG Oncology at Delnor, Delnor, IL<br />

Background: Bevacizumab added to weekly paclitaxel resulted in<br />

improved progression free survival (PFS) and objective response rates<br />

(ORR) compared to weekly paclitaxel alone. Nab-paclitaxel and the<br />

platins are active in MBC. We conducted an efficacy and safety study<br />

of carboplatin, nab-paclitaxel and bevacizumab.<br />

Methods: A phase II open label prospective multi-site study enrolled<br />

patients (pts) who had measurable MBC according to RECIST 1.1<br />

criteria and no prior chemotherapy for advanced disease. The primary<br />

endpoint was PFS with secondary endpoints of overall survival<br />

(OS), ORR, and safety. Pts initially received carboplatin AUC 6 day<br />

1, 22,43, plus weekly nab-paclitaxel 100mg/m 2 and bevacizumab<br />

15mg/m 2 day 1,22,43 of a 56 day cycle. This was later changed to<br />

carboplatin AUC 6 day 1, nab-paclitaxel 100mg/m 2 day 1,8,15, and<br />

bevacizumab 10mg/m 2 day 1,15 of a 28 day cycle. Thirty-two pts<br />

were required to detect an increase in median PFS from 6.7 to 10.5<br />

mo with 80% power based on a one-sided p = 0.05. Kaplan–Meier<br />

analyses estimated PFS and OS. The log rank test was used for the<br />

comparison of survival curves between pts with triple negative MBC<br />

(TNBC) and pts with non-TNBC.<br />

Results: Thirty-two pts were enrolled between 2/2008 and 11/2011<br />

by 1 academic and 5 community oncology practices. Two pts were<br />

ineligible due to non-measurable disease and not included in the<br />

response analyses. The median age was 58 years (range 35-81), 22<br />

pts (69%) had an ECOG PS 0, 9 (28%) had a PS 1, 1 (3%) PS 2.<br />

Twenty-four (75%) pts had ER+ disease, 7 (22%) had TNBC, 1 (3%)<br />

had ER-HER2+ disease not eligible for trastuzumab-based therapy.<br />

Metastatic sites were bone (26%), liver (18%), loco-regional (16%),<br />

and lung (12%). One pt (3%) had bone and loco-regional disease only,<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

192s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 1<br />

19 (59%) had visceral dominant disease. The median number of weeks<br />

on treatment was 28.9 (range 5-131). The median PFS in all pts was<br />

13.6 months (mo) (95% CI 11.2-21.9), with a median OS of 26.8 mo<br />

(95% CI 13.3-41.2). The ORR (2 CR and 18 PR) was 66.7% (CI 47.2-<br />

82.7). There also were 6 (20%) unconfirmed PR and 3 (10%) stable<br />

disease, resulting in a clinical benefit rate of 96.7% (CI 82.78-99.92).<br />

There was no significant difference in PFS (median 13.6 vs 16.1mo,<br />

p=0.37) or OS (median 13.6 vs 26.8mo, p=0.32) in pts with TNBC<br />

versus non-TNBC disease. The most common toxicities of any grade<br />

(gr) include neutropenia and thrombocytopenia in 24 pts (75%) each,<br />

leukopenia and fatigue in 17 pts (53%) each, anemia in 15 (47%), and<br />

neuropathy in 10 (31%). Gr 4 neutropenia was seen in 7 pts (22%)<br />

without febrile neutropenia, and gr 4 thrombocytopenia occurred in<br />

6 (19%). There were no pts with gr 4 sensory neuropathy. All pts<br />

required chemotherapy dose delays, 15 (47%) had chemotherapy<br />

dose reductions.<br />

Conclusions: The carboplatin, nab-paclitaxel and bevacizumab<br />

combination is highly effective with good tolerance in first line<br />

MBC. As the role of anti-angiogenic therapy in first line metastatic<br />

breast cancer is being clarified, this would be an attractive regimen<br />

to test in the (neo)adjuvant setting and together with novel molecular<br />

targeted agents.<br />

P1-12-05<br />

First-line chemotherapy with pegylated liposomal doxorubicin<br />

versus capecitabine in elderly patients with metastatic breast<br />

cancer: results of the phase III OMEGA study of the Dutch <strong>Breast</strong><br />

<strong>Cancer</strong> Trialists’ Group (BOOG)<br />

Smorenburg CH, Seynaeve C, Wymenga MANM, Maartense E, de<br />

Graaf H, de Jongh FE, Braun HJ, Los M, Schrama JG, Portielje<br />

JEA, Hamaker M, van Tinteren H, de Groot SM, van Leeuwen-Stok<br />

EAE, Nortier HWR. Medical Center Alkmaar, Alkmaar, Netherlands;<br />

Erasmus Medical Center-Daniel den Hoed <strong>Cancer</strong> Center, Rotterdam,<br />

Netherlands; Medisch Spectrum Twente, Enschede, Netherlands;<br />

Reinier de Graaf Hospital, Delft, Netherlands; Medical Center<br />

Leeuwarden, Leeuwarden, Netherlands; Ikazia Hospital, Rotterdam,<br />

Netherlands; Vlietland Hospital, Schiedam, Netherlands; St.<br />

Antonius Hospital, Nieuwegein, Netherlands; Spaarne Hospital,<br />

Hoofddorp, Netherlands; Haga Hospital, The Hague, Netherlands;<br />

Diaconessehuis, Utrecht, Netherlands; Antoni van Leeuwenhoek<br />

Hospital/Netherlands <strong>Cancer</strong> Institute, Amsterdam, Netherlands;<br />

Comprehensive <strong>Cancer</strong> Centre the Netherlands, Amsterdam,<br />

Netherlands; Dutch <strong>Breast</strong> <strong>Cancer</strong> Trialists’ Group BOOG,<br />

Amsterdam, Netherlands; Leiden University Medical Center, Leiden,<br />

Netherlands<br />

Background The efficacy and feasibility of chemotherapy in elderly<br />

metastatic breast cancer (MBC) patients (pts) have been studied in<br />

various phase II studies. However, results of prospective randomized<br />

studies in elderly MBC pts are scarce.<br />

Methods In this phase III multicenter study, MBC pts ≥ 65 years<br />

eligible for first-line chemotherapy were randomized between<br />

pegylated liposomal doxorubicin (PEGdoxo) (45mg/m 2 , IV, q 4 wks)<br />

or capecitabine (Cape) (1000 mg/m 2 PO bid, days 1-14, q 3 wks).<br />

Other eligibility criteria were ECOG performance status (PS) ≤ 2 (3<br />

allowed if due to pain or pre existing comorbidity), adequate bone<br />

marrow and organ functions. Stratification factors were PS (0-1 vs<br />

2-3), HER2 status, visceral/non-visceral disease, adjuvant hormonal<br />

therapy (HTx), and HTx for MBC. Baseline geriatric assessment<br />

(GA) included functional status, instrumental activities of daily<br />

living, cognition, mood, comorbidity, polypharmacy and nutritional<br />

status. Chemotherapy was continued for 24 wks in the absence of<br />

progressive disease (PD) or unacceptable toxicity. Primary endpoint<br />

was progression-free survival (PFS), secondary endpoints were<br />

response rate, overall survival (OS), toxicity (CTC criteria) and<br />

compliance.<br />

Results Between April 2007 and August 2011, 78 pts were<br />

randomized to PEGdoxo (n=40) or Cape (n=38). The study was<br />

prematurely closed due to slow accrual and supply problems with<br />

PEGdoxo. Mean age was 74 years (range 65-86; 75+ 54%; 80+<br />

13%). Pt characteristics were balanced between the two arms: PS<br />

0-1 77%, ER+ 68%, HER2+ 5%, visceral/non-visceral disease<br />

76%/24%, adjuvant HTx 46%, HTx for MBC 56%, ≥ 3 metastatic<br />

sites 50%. Only 22 out of 75 pts with a baseline GA had no geriatric<br />

condition (29%), while 32 pts (43%) and 21 pts (28%) had one or<br />

≥ 2 geriatric conditions, respectively. Chemotherapy was given for<br />

6 months in 38%, with a mean dose intensity of 84% in both arms.<br />

Reasons for early treatment discontinuation were: PD (31%), toxicity<br />

(28%), pt withdrawal (3%). After a median follow up of 32 months,<br />

74 pts had PD and 56 pts had died. The median PFS was 5.7 and<br />

7.7 months with PEGdoxo and Cape (HR 0.68, 95% CI: 0.42-1.11,<br />

p=0.12) and the median OS was 13.8 and 16.8 months, respectively<br />

(HR 0.84, 95% CI: 0.49-1.42, p=0.51). Response was evaluable in<br />

64 pts, with a partial response (PR) in 7 (21%) and 6 pts (19%), and<br />

stable disease in 21 (64%) and 17 pts (55%) for PEGdoxo and Cape,<br />

respectively. Toxicity was acceptable, mainly being grade 1-2, with<br />

for PEGdoxo/Cape grade 1 alopecia in 14/4 pts (grade 2 in 1 PEGdoxo<br />

pt), grade 3 fatigue in 5/5 pts, grade 3 HFS in 4/6 pts and grade 3<br />

mucositis in 4/1 pts, respectively. Pts with ≥ 1 geriatric condition<br />

more frequently experienced grade 3-4 toxicity, after correcting for<br />

type of chemotherapy, age and PS (HR 2.24, 95% CI: 1.21-4.16). Pts<br />

aged 75+ had a twofold higher risk of dying, irrespective of treatment<br />

arm (HR 2.31, 95% CI: 1.31-4.07).<br />

Conclusions First-line chemotherapy with either PEGdoxo or Cape<br />

was feasible in elderly MBC pts, with adequate dose intensity and<br />

acceptable toxicity, even in non-fit pts or pts aged 75+. Baseline GA<br />

correlated with toxicity.<br />

P1-12-06<br />

N0937 (Alliance): Preliminary results of a phase II clinical trial<br />

of cisplatin and the novel agent brostallicin in patients with<br />

metastatic triple negative breast cancer (mTNBC)<br />

Moreno-Aspitia A, Rowland, Jr. KM, Allred JB, Liu H, Stella PJ, Gross<br />

HM, Soori GS, Karlin NJ, Perez EA. Mayo Clinic, Jacksonville, FL;<br />

Carle Foundation - Carle <strong>Cancer</strong> Center, Urbana, IL; Mayo Clinic,<br />

Rochester, MN; St. Joseph Mercy Health System, Ann Arbor, MI;<br />

Hematology & Oncology of Dayton, Inc., Dayton, OH; Missouri<br />

Valley <strong>Cancer</strong> Consortium CCOP, Omaha, NE; Mayo Clinic,<br />

Scottsdale, AZ<br />

Background: TNBC is characterized by unique molecular profiles,<br />

aggressive behavior, poor prognosis and lack of targeted therapies.<br />

Brostallicin is a novel synthetic compound from the class of DNA<br />

minor groove binding (MGB) anti-cancer agents, making it a logical<br />

agent to evaluate in the setting of TNBC. It retains activity in cancer<br />

cells resistant to alkylating agents, topoisomerase I inhibitors and<br />

is fully active against DNA-mismatch repair deficient tumor cells.<br />

Preclinical models using cell lines demonstrate that cells expressing<br />

relatively high glutathione/glutathione S-transferase (GSH/GST)<br />

levels are more susceptible to brostallicin’s antitumor efficacy.<br />

Cisplatin administration increases expression of GSH/GST in tumor<br />

cells, thus leading to an increased anti-tumor efficacy of brostallicin.<br />

Methods: Phase II cooperative group study in pts with mTNBC (³18<br />

years of age with measurable metastatic disease, ER/PR ≤1%; HER2<br />

www.aacrjournals.org 193s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

negative, who had received 0-4 prior chemotherapy regimens in the<br />

metastatic setting; with adequate hematologic, renal and hepatic<br />

functions; and no active CNS metastases; prior exposure to cisplatin<br />

allowed). Cisplatin on Day 1 followed by brostallicin on Day 2,<br />

repeated every 21 days. Aim: efficacy of brostallicin and proof of<br />

concept of its mechanism of action in mTNBC. Primary endpoint<br />

progression-free survival (PFS) at 3 months with 89% power (0.10<br />

significance level) to detect an absolute difference of 20% (35% vs<br />

55%), based on the median PFS of 60 days in pts with mTNBC from<br />

the N0234 trial of erlotinib and gemcitabine as 1 st /2 nd line. Secondary<br />

endpoints include ORR, duration of response (DOR), 6-month PFS,<br />

OS and AE profile. Tertiary endpoints include assessment of 1) GSH<br />

levels prior to the administration of cisplatin and of brostallicin;<br />

and 2) the prevalence of BCRA-1 mutation by IHC in primary or<br />

metastatic tumor.<br />

Results: Study closed on 3/28/12 and it accrued 48 pts (median<br />

f/u 2.3 mo; 0-15.3); 33 pts are off treatment and 15 pts remain on<br />

study; 38 pts evaluable for response, and 43 evaluable for AEs. 50%<br />

received therapy as 3rd to 5th line. Median number of cycles 2.5<br />

(off-treatment: 2; on-treatment: 3, range 0-15). There are currently 5<br />

confirmed responses (4 PR and 1 CR); DOR: 2.8-13.3 months. The<br />

6-mo PFS is currently 19.2% (95% CI: 8.9%, 41.3%); the median<br />

TTP is 3.0 months (95% CI: 1.7 months, 4.2 months). Current data<br />

are premature to determine the primary endpoint (3-mo PFS) but<br />

we expect to report such data by November 2012.Current toxicity<br />

data: 69.7% G3/4 heme toxicity. Non-heme toxicity G3 (30.2%) and<br />

G4 (9.3)% (febrile neutropenia 21%; fatigue G3 14%); and no G5<br />

non-heme AE.<br />

Conclusions: The current preliminary data of this trial show very<br />

encouraging activity of this regimen (brostallicin plus cisplatin) in<br />

mTNBC. Near 1/3 of pts are still currently receiving therapy, and we<br />

expect to provide primary and additional secondary endpoint data at<br />

SABCS 2012.<br />

P1-12-07<br />

A Retrospective Analysis of nab-Paclitaxel as First-Line Therapy<br />

for Metastatic <strong>Breast</strong> <strong>Cancer</strong> Patients With Poor Prognostic<br />

Factors<br />

O’Shaughnessy J, Gradishar W, Bhar P, Iglesias J. Baylor Sammons<br />

<strong>Cancer</strong> Center, Texas Oncology and US Oncology, Dallas, TX;<br />

Maggie Daley Center for Women’s <strong>Cancer</strong> Care, Robert H. Lurie<br />

Comprehensive <strong>Cancer</strong> Center, Northwestern University Feinberg<br />

School of Medicine, Northwestern Memorial Hospital, Chicago,<br />

IL; Celgene Corporation, Durham, NC; Celgene Corporation,<br />

Mississauga, ON, Canada<br />

Background: Solvent-based taxanes have demonstrated activity in<br />

metastatic breast cancer (MBC) patients with poor prognostic factors,<br />

an important subset of patients with aggressive disease who require<br />

special consideration in treatment optimization. nab-Paclitaxel has<br />

been compared with solvent-based (sb-) paclitaxel in a randomized<br />

phase III trial (CA012; N = 454) and docetaxel in a randomized<br />

phase II trial (CA024; N = 300) for the treatment of MBC, revealing<br />

a favorable clinical benefit for nab-paclitaxel in both intent-to-treat<br />

(ITT) populations. This retrospective analysis examines whether<br />

the safety and efficacy of nab-paclitaxel observed in these trials are<br />

maintained in patients with poor prognostic factors.<br />

Methods: This retrospective analysis evaluated safety and efficacy of<br />

the first-line treatment of patients with MBC with the poor prognostic<br />

factors of visceral dominant metastases (dominant metastatic lesion<br />

localized to visceral tissue [ie, not in bone or soft tissues]) and early<br />

disease recurrence (≤ 2 years after completing neoadjuvant or adjuvant<br />

treatment) in the CA012 and CA024 trials.<br />

Results: In CA012 (n = 186 first-line patients), nab-paclitaxel 260<br />

mg/m 2 every 3 weeks (q3w) demonstrated a higher overall response<br />

rate (ORR) vs sb-paclitaxel 175 mg/m 2 q3w in patients with visceral<br />

dominant metastases (42% vs 23%, P = .022) and in patients with<br />

early disease recurrence (43% vs 33%, P = not significant [NS]). In<br />

CA024 (n = 300 first-line patients), significantly higher ORRs for<br />

nab-paclitaxel at 100 mg/m 2 (63%, P = .002) and 150 mg/m 2 (76%,<br />

P < .001) the first 3 of 4 weeks (qw 3/4) vs docetaxel 100 mg/m 2 q3w<br />

(37%) were observed in patients with visceral dominant metastases.<br />

The difference in ORR in this subgroup between nab-paclitaxel 300<br />

mg/m 2 q3w (44%, P = NS) and docetaxel 100 mg/m 2 q3w was not<br />

statistically different. As in CA012, the differences in ORR among<br />

patients with early disease recurrence in CA024 were not statistically<br />

significant (35% to 64% for the nab-paclitaxel arms vs 21% for<br />

the docetaxel 100 mg/m 2 q3w arm, all P = NS). Progression-free<br />

survival (PFS) and overall survival (OS) showed trends similar to<br />

that of ORR in both trials. Statistical significance was only achieved<br />

for median PFS in CA024 among patients with visceral dominant<br />

metastases (13.1 months for nab-paclitaxel 150 mg/m 2 qw 3/4 vs<br />

7.5 months for nab-paclitaxel 100 mg/m 2 qw 3/4 [P = .010] and vs<br />

7.8 months for docetaxel 100 mg/m 2 q3w [P = .019]). Safety results<br />

were similar to those of previous reports for the ITT populations;<br />

in CA024, the lowest rates of neutropenia, sensory neuropathy, and<br />

fatigue were observed with nab-paclitaxel 100 mg/m 2 qw 3/4 in both<br />

patient subgroups.<br />

Conclusions: nab-Paclitaxel showed similar efficacy in patients with<br />

poor prognostic factors to that in the ITT populations of these 2 trials.<br />

In each trial, consistent trends in favor of nab-paclitaxel were observed<br />

for ORR, PFS, and OS vs a solvent-based taxane in the specified poor<br />

prognostic factor subgroups. No new safety signals were observed.<br />

P1-12-08<br />

Withdrawn by Author.<br />

P1-13-01<br />

A randomized trial of CEF versus dose dense EC followed by<br />

paclitaxel versus AC followed by paclitaxel in women with node<br />

positive or high risk node negative breast cancer, NCIC CTG<br />

MA.21: Results of the final relapse free survival analysis.<br />

Burnell MJ, Shepherd L, Gelmon K, Bramwell V, Walley B,<br />

Vandenberg E, Chalchal H, Pritchard K, Whelan T, Albain K, Perez<br />

E, Rugo H, O’Brien P, Chapman J, Levine M. NCIC Clinical Trials<br />

Group, Queen’s University, Kingston, ON, Canada; British Columbia<br />

<strong>Cancer</strong> Agency (BCCA), Vancouver, BC, Canada; University of<br />

Calgary, AB, Canada; Saint John Regional Hospital, Saint John, NB,<br />

Canada; London Regional <strong>Cancer</strong> Centre, London, ON, Canada;<br />

Sunnybrook <strong>Cancer</strong> Centre, Toronto, ON, Canada; Juravinski<br />

<strong>Cancer</strong> Centre, Hamilton Health Sciences Centre, Hamilton, ON,<br />

Canada; Loyola University Medical Center, Maywood, IL; Mayo<br />

Clinic Jacksonville, FL; University of California, <strong>San</strong> Francisco, CA;<br />

McMaster University, Hamilton, ON, Canada; Allan Blair <strong>Cancer</strong><br />

Centre, Regina, SK, Canada<br />

Background: In 2000, cyclophosphamide(C), epirubicin(E) and<br />

fluorouracil (F) (CEF) and doxorubicin (A) and C followed by<br />

paclitaxel (T) (AC/T) given every three weeks were commonly used<br />

regimens in Canada and the USA to treat women with node positive<br />

or high risk node negative operable breast cancer. In a previous trial in<br />

women with locally advanced breast cancer, 3 months of dose-dense<br />

EC was equivalent to six months of CEF. We hypothesized that the<br />

addition of 3 months of a taxane following dose-dense EC would<br />

be superior to CEF alone or AC/T. In a randomized trial in women<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

194s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 1<br />

with early breast cancer we compared CEF, dose dense EC/T and<br />

AC/T. An interim analysis (JCO, 2010) conducted when 50% of the<br />

events for the relapse-free-survival (RFS) analysis were observed,<br />

demonstrated that AC/T administered every 3 weeks was significantly<br />

inferior to CEF or EC/T. The results of the final RFS including CEF<br />

versus EC/T will be presented. Methods: Women, 60 years or younger,<br />

with operable axillary node positive or high risk node negative breast<br />

cancer were randomized to adjuvant CEF, EC/T or AC/T. CEF was<br />

given for 6 cycles and consisted of: C 75 mg/m2 orally on Days<br />

1-14, and E 60 mg/m2 and F 500 mg/m2, IV on Days 1 and 8. CEF<br />

patients received concurrent antibiotic prophylaxis. EC/T consisted<br />

of 6 cycles of EC every 14 days; E 120 mg/m2 and C 830 mg/m2,<br />

both IV on Day 1 with filgrastim 5 [ug]/kg/day subcutaneous from<br />

Days 2-13 and epoetin alpha 40,000 units subcutaneous once weekly.<br />

After completion of EC, 4 cycles of T 175 mg/m2 every 21 days with<br />

filgrastim and epoetin alpha were given. The AC/T regimen consisted<br />

of A 60 mg/m2 and C 600 mg/m2 IV every 21 days for 4 cycles.<br />

This was followed by T 175 mg/m2 every 21 days for 4 cycles. The<br />

final analysis for RFS was planned when 453 events were observed<br />

permitting the detection of a hazard ratio (HR) of 1.43 between CEF<br />

and AC/T and a HR of 1.43 between EC/T and the best of the other<br />

two arms. Quality of Life data using the EORTC C-30 and the <strong>Breast</strong><br />

<strong>Cancer</strong> Chemotherapy questionnaires were collected throughout the<br />

study. The results will be available at the time of the presentation.<br />

Results: A total of 2104 women were enrolled between December<br />

2000 and April 2005. The required 453 events for the RFS analysis<br />

were observed by the clinical cut-off date of March 30, 2012. At this<br />

point 369 deaths had been observed. The median follow-up is 87.5<br />

months. Outstanding data and queries are currently being collected<br />

and reviewed with an expected database lock and final analysis mid<br />

August, 2012. Febrile neutropenia remains higher on the CEF and<br />

EC/T arms while more neuropathy was seen in the taxane containing<br />

regimens. Conclusions: Although the two regimens considered a<br />

standard of care at the time the study was initiated are no longer widely<br />

used, the trial results will enhance our understanding of the magnitude<br />

of benefit of taxane following an anthracycline, the significance of<br />

cumulative anthracycline dose, and the role of dose density/ intensity<br />

as backbone of conventional chemo regimens.<br />

P1-13-02<br />

Withdrawn by Author<br />

P1-13-03<br />

Mature analysis of UK Taxotere as Adjuvant Chemotherapy<br />

(TACT) trial (CRUK 01/001); effects of treatment and<br />

characterisation of patterns of breast cancer relapse.<br />

Bliss JM, Ellis P, Kilburn L, Bartlett J, Bloomfield D, Cameron D,<br />

Canney P, Coleman RE, Dowsett M, Earl H, Verril M, Wardley A,<br />

Yarnold J, Ahern R, Atkins N, Fletcher M, McLinden M, Barrett-Lee P.<br />

Institute of <strong>Cancer</strong> Research, Sutton, Surrey, United Kingdom; Guy’s<br />

Hospital, Kings Health Partners AHSC, London, United Kingdom;<br />

Velindre NHS Trust <strong>Cancer</strong> Centre, Cardiff, United Kingdom;<br />

Edinburgh <strong>Cancer</strong> Research Centre, University of Edinburgh, United<br />

Kingdom; The Christie Hospital, Manchester, United Kingdom;<br />

Northern Centre for <strong>Cancer</strong> Care, Newcastle upon Tyne, United<br />

Kingdom; Brighton & Sussex University Hospitals, Brighton, United<br />

Kingdom; Ontario Institute for <strong>Cancer</strong> Research, Toronto, Canada;<br />

Beatson West of Scotland <strong>Cancer</strong> Centre, Glasgow, United Kingdom;<br />

Leeds Institute of Molecular Medicine, University of Leeds, Leeds,<br />

United Kingdom; ICR and Royal Marsden NHS Trust, London, United<br />

Kingdom; University of Cambridge, University of Cambridge and<br />

NIHR Cambridge Biomedical Research Centre, Cambridge, United<br />

Kingdom; Weston Park Hospital, Sheffield, United Kingdom; NHS<br />

National Services Scotland, Edinburgh, United Kingdom<br />

Introduction: TACT, an investigator-led study in 4162 women with<br />

node positive (N+ve) or high risk node negative (N-ve) early breast<br />

cancer (EBC), is the largest taxane trial unconfounded by treatment<br />

(trt) duration. At principal analysis, with 5 years follow-up (fup), no<br />

evidence of improved disease-free survival (DFS) was observed by<br />

switching to 4 cycles of docetaxel (D) after 4 cycles of FEC (Ellis,<br />

Lancet 2009). Results were provocative in suggesting differential<br />

effects according to ER & HER2 status. Longer fup provides<br />

opportunity to detect emergence of late trt effects overall & within<br />

phenotypic subgroups & explore patterns of recurrence, by tumor<br />

characteristics.<br />

Patients & methods: TACT recruited women with histologically<br />

confirmed completely resected invasive EBC from 104 centers (UK<br />

(103), Belgium (1)) between 02/2001 & 07/2003. Centers chose FEC<br />

(600/60/600 mg/m 2 q3wk x 8) or E-CMF (E 100mg/m 2 q3wk x 4 →<br />

CMF 100mg/m 2 PO d1-14 or 600mg/m 2 IV d1&8 /40/600 mg/m 2<br />

q4wk x 4) as their control, reflecting standard UK practice. Patients<br />

(pts) were randomized to FEC-D (FEC q3wk x 4 → D 100 mg/m 2<br />

q3wk x 4) or control. 2523 pts were from FEC centers (FEC=1265:<br />

FEC-D=1258) & 1639 from E-CMF centers (E-CMF=824;<br />

FEC-D=815). Endocrine therapy was given for 5 years. Few pts<br />

received HER2 directed therapy; 589 pts had unknown HER2 status.<br />

Median fup is now 97.5 months; this analysis updates DFS & overall<br />

survival in the ITT population. It also explores patterns of relapse<br />

by phenotypic & clinical characteristics. Analyses of trt effect are<br />

stratified by ER status due to issues of non-proportionality of hazard<br />

associated with length of fup.<br />

Results: DFS events have been reported for 1329 pts (FEC-D=640,<br />

Control=689) giving an unadjusted hazard ratio (HR) & 95%CI<br />

(stratified by control regimen & ER status) of 0.93 (0.83, 1.03) overall;<br />

p=0.16 in favor of FEC-D & for ER+ve/HER2-ve of 0.99 (0.84,<br />

1.17), for ER+ve/HER2+ve) 0.97 (0.73, 1.30), for ER-ve/HER2+ve<br />

0.74 (0.53, 1.03), & ER-ve/HER2-ve 0.93 (0.73, 1.17). 1017 patients<br />

have died (FEC-D=500, Control=517); unadjusted HR=0.98 (95%CI:<br />

0.86, 1.10); p=0.69 with intercurrent deaths (prior to distant relapse)<br />

reported for 80 pts (FEC-D=40, Control=40).<br />

Annual event rates show different pattern of disease relapse by<br />

phenotypic subgroup<br />

www.aacrjournals.org 195s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

DFS annual<br />

event rate N+ N-<br />

(years)<br />

2 5 8 2 5 8<br />

ER+ve &/<br />

or PgR+ve<br />

and HER2- 4.6 (3.7, 5.8) 4.5 (3.5,5.7) 5.1 (3.8,6.8) 1.6 (0.6,4.2) 2.2 (0.9,5.3) 1.3 (0.3,5.1)<br />

ve(N+=1745,<br />

N-=256)<br />

HER2+ve<br />

(N+=691, 15.0 (12.2, 18.5) 5.1 (3.4, 7.8) 2.8 (1.3,6.0) 5.3 (2.6,10.5) 1.5 (0.4,5.8) 0.0 (-,-)<br />

N-=158)<br />

ER-ve/<br />

HER2-ve<br />

(N+=397,<br />

N-=326)<br />

19.3 (15.1, 24.7) 4.3 (2.3,8.0) 3.5 (1.6,7.8) 4.9 (3.0,8.2) 4.1 (2.3,7.5) 2.0 (0.7,5.3)<br />

Graphical representation will further explore these patterns &<br />

associated sites of relapse.<br />

Discussion: With a median fup of >8 years no clear benefit has<br />

emerged for D over standard anthracyclines within the TACT pt<br />

group. Differential effects associated with different patterns of relapse<br />

remain of interest. TACT precedes use of antiHER2 therapy which<br />

is known to have impacted on early relapse risk in HER2+ve pts.<br />

The high relapse risk observed for pts with ER-ve/HER2-ve disease<br />

remains a current clinical challenge.<br />

P1-13-04<br />

Optimal duration of adjuvant chemotherapy for high risk node<br />

negative breast cancer patients: 6-year results of the prospective<br />

randomized phase III trial PACS 05.<br />

Kerbrat P, Coudert B, Asselain B, Levy C, Lortholary A, Marre A,<br />

Delva R, Rios M, Viens P, Brain E, Serin D, Edel M, Mauriac L,<br />

Campone M, Mouret-Reynier M-A, Bachelot T, Foucher-Goudier<br />

M-J, Roca L, Martin A-L, Roche H. Centre Eugene Marquis, Rennes,<br />

France; Centre Georges François Leclerc, Dijon, France; Institut<br />

Curie, Paris, France; Centre Francois Baclesse, Caen, France;<br />

Centre Catherine de Sienne, Nantes, France; Centre Hospitalier,<br />

Rodez, France; ICO Centre Paul Papin, Angers, France; Centre<br />

Alexis Vautrin, Vandoeuvre-les-Nancy, France; Institut Paoli<br />

Calmettes, Marseille, France; Institut Curie, Saint Cloud, France;<br />

Institut Sainte-Catherine, Avignon, France; Centre Hospitalier Emile<br />

Muller, Mulhouse, France; Institut Bergonié, Bordeaux, France;<br />

ICO Centre René Gauducheau, Saint Herblain, France; Centre<br />

Jean Perrin, Clermont-Ferrand, France; Centre Léon Berard, Lyon,<br />

France; Centre Hospitalier Bretagne-Sud, Lorient, France; Centre<br />

Val d’Aurelle, Montpellier, France; R&D Unicancer, Paris, France;<br />

Institut Claudius Regaud, Toulouse, France<br />

BACKGROUND In 2000, the NIH Consensus meeting concluded<br />

that 4 to 6 cycles of adjuvant chemotherapy appeared to provide an<br />

optimal benefit; So, we underwent a prospective randomized trial<br />

comparing 4 and 6 cycles of FEC 100 (JCO 2005; 23: 2686-2693)<br />

for high risk node negative breast cancer patients.<br />

METHODS This study enrolled 18-65 y women with operable breast<br />

cancer, without axillary lymph node involvement, or presence of<br />

isolated tumor cells, with size superior to 1 cm and another poor<br />

prognostic factor : T > 2 cm, HR –, SBR grade II or III, age < 35 y.<br />

After adequate breast surgery and axillary lymph node dissection or<br />

sentinel node technique, they were randomized between arm A, 6<br />

cycles of FEC 100, and arm B, 4 cycles, every three weeks. The local<br />

regional treatment was completed following usual recommendations.<br />

All HR+ patients received hormonal therapy for 5 years. After August<br />

2005, patients with HER2+ tumors were excluded from this study.<br />

The primary end point was PFS at 5 years. This study was powered<br />

to detect a 6% difference in favour of 6 cycles.<br />

Between August 2002 and September 2006, 1516 patients were<br />

randomized; 1515 are analysed in ITT. Three patients in the B group<br />

did not receive any chemotherapy. There is no significant difference<br />

between the two arms for tumor and patient characteristics.<br />

RESULTS At a median follow-up of 73 months we observed regarding<br />

PFS a low event rate, 197 for the entire population (13%) 91 in arm<br />

A median PFS, vs. 106 in arm B median PFS, without any difference<br />

between the two groups for DFS, DDFS, local relapse, overall<br />

survival. There was no unexpected toxicity. In the arm A we observed<br />

more grade III and IV neutropenia, without congestive heart failure.<br />

CONCLUSION At a follow-up of 73 months, we observed a low<br />

relapse rate, with no significant difference between the two arms.<br />

Duration of FEC100 does not induce different outcomes in this<br />

population. Question of length of adjuvant treatment is still open<br />

with and without taxanes.<br />

P1-13-05<br />

The association between timing in adjuvant chemotherapy<br />

administration and overall survival for women with breast cancer<br />

within the National Comprehensive <strong>Cancer</strong> Network (NCCN)<br />

Vandergrift JL, Breslin TM, Niland JC, Edge SB, Wolff AC, Marcom<br />

PK, Rugo HS, Moy B, Wilson JL, Ottesen RA, Weeks JC, Wong Y-N.<br />

National Comprehensive <strong>Cancer</strong> Network, Fort Washington, PA;<br />

University of Michigan Comprehensive <strong>Cancer</strong> Center, Ann Arbor,<br />

MI; City of Hope Comprehensive <strong>Cancer</strong> Center, Duarte, CA; Dana-<br />

Farber/Brigham Women’s <strong>Cancer</strong> Center, Boston, MA; Roswell Park<br />

<strong>Cancer</strong> Institute, Buffalo, NY; The Sidney Kimmel Comprehensive<br />

<strong>Cancer</strong> Center at Johns Hopkins, Baltimore, MD; Duke <strong>Cancer</strong><br />

Institute, Durham, NC; UCSF Helen Diller Family Comprehensive<br />

<strong>Cancer</strong> Center, <strong>San</strong> Francisco, CA; The Ohio State University<br />

Comprehensive <strong>Cancer</strong> Center and James <strong>Cancer</strong> Hospital and<br />

Solove Research Institute, Columbus, OH; Massachusetts General<br />

Hospital <strong>Cancer</strong> Center, Boston, MA; Fox Chase <strong>Cancer</strong> Center,<br />

Philadelphia, PA<br />

Introduction: Population based studies (eg, Hershman et al. BCRT<br />

2006 and Lohrisch et al. JCO 2006) showed poorer survival associated<br />

with long delays in adjuvant chemotherapy (CTX) initiation following<br />

definitive surgery (DS) for women with breast cancer (BC). Delays in<br />

CTX following diagnosis (DX) have not been evaluated. The ASCO/<br />

NCCN quality measures (QMs) recommend CTX 90-day (d) DS-to-<br />

CTX threshold, based on poor outcomes observed in prior studies,<br />

and a >120d DX-to-CTX threshold, based on the ASCO/NCCN QMs.<br />

Results: Median follow-up was 7.2 years and OS at 7 years was<br />

89%. Overall, 401 (8.7%) patients received CTX >120d after DX and<br />

113 (2.4%) patients received CTX >90d after DS. The DX-to-CTX<br />

interval was more strongly correlated with the DX-to-DS (r=0.74)<br />

interval than DS-to-CTX (r=0.54) interval. A >90d DS-to-CTX<br />

interval was significantly associated with poorer survival (HR: 1.65,<br />

95% CI 1.04-2.60, p=0.03) in adjusted analyses. Shorter DS-to-CTX<br />

thresholds of >60d (n=636, HR: 1.13, 95% CI: 0.89-1.43, p=0.319)<br />

or >75d (n=273, HR: 1.05, 95% CI 0.74-1.49, p=0.76) were not<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

196s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 1<br />

associated with OS. The association between a >120d DX-to-CTX<br />

interval and OS was not statistically significant (HR: 1.32, 95% CI<br />

0.99-1.76, p=0.06). Patients who received CTX >135d (n=231, HR<br />

1.25, 95% CI: 0.87-1.81, p=0.22) or >150d (n=128, HR 1.15, 95% CI:<br />

0.59-2.24, p=0.69) after DX did not display an increased risk of death.<br />

Excluding pathological staging factors from the model had no effect<br />

on the results. In subgroup analyses stratified by ER/PR, LVI, grade,<br />

T stage or N stage, a >120d delay in CTX did not display significant<br />

associations with OS. Among ER/PR negative patients, the association<br />

between a >120d delay and OS was borderline non-significant after<br />

adjusting the p-value for multiple hypothesis testing using the false<br />

discovery rate method (HR: 1.80, 95% CI: 1.16-2.79, p=0.09).<br />

Conclusion: Consistent with previous studies, CTX delays of >90<br />

days following surgery were associated with poorer survival. OS<br />

was not significantly compromised in patients with DX-to-CTX<br />

intervals >120 days although this analysis may have limited power<br />

to detect small effects. More variation in the DX-to-CTX interval<br />

was attributed to pre-surgery time which may explain the differences<br />

observed between the DX-to-CTX and DS-to-CTX intervals. Among<br />

patients with ER/PR negative disease, a non-significant association<br />

between OS and a >120 day DX-to-CTX interval was observed that<br />

warrants further examination.<br />

P1-13-06<br />

Withdrawn by Author<br />

P1-13-07<br />

Association Between Delayed Initiation of Adjuvant Chemotherapy<br />

and Survival in <strong>Breast</strong> <strong>Cancer</strong>: A Single-institution Study and A<br />

Systematic Review and Meta-analysis<br />

Yu K-D, Fan L, Yang C, Shao Z-M. <strong>Cancer</strong> Center and <strong>Cancer</strong><br />

Institute, Shanghai Medical College, Fudan University, Shanghai,<br />

China<br />

Objective<br />

Adjuvant chemotherapy (AC) improves survival among patients with<br />

operable breast cancer. However, the effect of delay in AC initiation on<br />

survival is unclear. We performed a single-institution data analysis and<br />

a systematic review and meta-analysis to determine the relationship<br />

between time to AC and survival outcomes.<br />

Methods<br />

PubMed, EMBASE, Cochrane Database of Systematic Reviews, and<br />

Web-of-Science databases (before March-20, 2012) were searched to<br />

identify relevant eligible studies. An additional retrospective analysis<br />

including 1,408 patients from the prospectively maintained database<br />

of Shanghai <strong>Cancer</strong> Center was performed and results were also<br />

included. Hazard ratios (HRs) for overall survival (OS) and diseasefree<br />

survival (DFS) from each study were converted to a regression<br />

coefficient (β) and its standard error corresponding to a continuous<br />

representation per 4-week delay of AC. Individual adjusted β were<br />

combined using a fixed-effects or random-effects model depending<br />

on heterogeneity.<br />

Results<br />

We included 8 eligible studies with 10 independent analytical groups<br />

involving 21,221 patients, 1 prospective observational study, 2<br />

secondary analyses in randomized trials (4 analytical groups), and<br />

5 hospital-/population-based retrospective studies. In our singleinstitution<br />

study, every 4-week delay in initiation of AC significantly<br />

decreased OS (HR=1.38; 95% confidence interval [CI], 1.03-1.87)<br />

and DFS (HR=1.35; 95% CI, 1.08-1.69) after adjustment for other<br />

prognostic variables. The overall meta-analysis demonstrated that<br />

a 4-week increase in time to AC was associated with a significant<br />

decrease in both OS (HR=1.20; 95% CI, 1.03-1.39; random-effects<br />

model) and DFS (HR=1.21; 95% CI, 1.08-1.36; fixed-effects model).<br />

One study caused a significant between-study heterogeneity for OS<br />

(P=0.001; I 2 =74.1%); after excluding that single study, there was no<br />

heterogeneity (P=0.345; I 2 =11.1%) and the HR was more significant<br />

(HR=1.24; 95% CI, 1.14-1.35; P


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

chemotherapy and those who had chemotherapy alone. There was also<br />

no statistically significant difference in B cell depletion or recovery<br />

between different age groups, initial tumour size or grade of tumour.<br />

Conclusions<br />

These results confirm that B cells are especially vulnerable to<br />

chemotherapy toxicity. Detailed phenotyping of B cells revealed<br />

that chemotherapy has a profound effect on B cell subpopulations.<br />

The significance of this on host immunity is under investigation. We<br />

have additionally identified two factors responsible for delayed B cell<br />

repopulation, which highlight the need for lymphocyte assessments<br />

to be performed during and after chemotherapy.<br />

P1-13-09<br />

A multicenter randomized study comparing the dose dense<br />

G-CSF-supported sequential administration of FEC followed by<br />

docetaxel versus paclitaxel as adjuvant chemotherapy in women<br />

with axillary lymph node positive breast cancer.<br />

Mavroudis D, Malamos N, Boukovinas I, Kakolyris S, Kourousis C,<br />

Athanasiadis A, Ziras N, Makrantonakis P, Polyzos A, Christophylakis<br />

C, Georgoulias V. Hellenic Oncology Research Group (HORG),<br />

Athens, Greece<br />

Purpose: To compare the efficacy of dose dense docetaxel versus<br />

paclitaxel following FEC as adjuvant chemotherapy in node positive<br />

early breast cancer.<br />

Patients and treatment: Women 18-75 years old with histologically<br />

confirmed HER2-negative invasive breast carcinoma surgically<br />

resected with at least one infiltrated axillary lymph node and<br />

absence of metastatic disease were randomized to receive 4 cycles of<br />

fluorouracil (700mg/m2), epirubicin (75mg/m2), cyclophosphamide<br />

(700mg/m2) followed by 4 cycles of either docetaxel (75mg/m2) or<br />

paclitaxel (175mg/m2). All chemotherapy cycles were administered<br />

every 14 days with G-CSF support. Stratification was based on<br />

menopausal status, number of involved nodes and hormone receptor<br />

expression. The primary endpoint of the study was to compare the<br />

disease-free survival (DFS) at 3 years and 239 patients were scheduled<br />

to enroll on each arm.<br />

Results: Between 2004-2007, 481 women were randomized and<br />

received FEC followed by docetaxel (arm A; n=240) or paclitaxel (arm<br />

B; n=241). The median age was 55 years in both arms, premenopausal<br />

status 31.3% versus 32.8%, more than 10 involved axillary nodes<br />

in 12.9% versus 12.4%, histological grade 3 tumor in 36.3% versus<br />

35.3% and hormone receptor negative disease in 14.6% versus 12%<br />

of patients in arms A and B, respectively. After a median follow up<br />

of 56.3 and 55.6 months (p=0.3) for arms A and B, respectively, there<br />

were 42 (17.5%) versus 47 (19.5%) disease relapses (p=0.5) and 20<br />

(8.3%) versus 22 (9.1%) disease-related deaths (p=0.7), respectively.<br />

The 3-year DFS rates were 88.1% versus 87.3% for arms A and B,<br />

respectively. Toxicity included grade 2-4 neutropenia in 31% versus<br />

21% (p=0.01), thrombocytopenia 3.5% versus 0.8% (p=0.06), febrile<br />

neutropenia 2.1% versus 1.2% (p=0.5), diarrhea 3.7% versus 2.5%<br />

(p=0.4), neurotoxicity 2.9% versus 4.6% (p=0.3) of patients in arms<br />

A and B, respectively. There were no toxic deaths.<br />

Conclusion: The dose dense administration with G-CSF support<br />

of FEC followed by either docetaxel or paclitaxel as adjuvant<br />

chemotherapy in women with node positive early breast cancer is<br />

well tolerated and results in a similar 3-year DFS rate.<br />

P1-13-10<br />

Efficacy, toxicity and quality of life in older patients with earlystage<br />

breast cancer treated with oral Tegafur-uracil or classical<br />

CMF (cyclophosphamide, methotrexate, and fluorouracil): an<br />

exploratory analysis of National Surgical Adjuvant Study for<br />

<strong>Breast</strong> <strong>Cancer</strong> (N-SAS BC) 01 Trial<br />

Hara F, Watanabe T, Shimozuma K, Ohashi Y. NHO Shikoku <strong>Cancer</strong><br />

Center, Matsuyama, Ehime, Japan; Hamamatsu Oncology Center,<br />

Hamamatsu, Shizuoka, Japan; College of Life Sciences, Ritsumeikan<br />

University, Kusatsu, Shiga, Japan; School of Public Health,<br />

University of Tokyo, Bunkyo-ku, Tokyo, Japan<br />

Background: It is critically important to consider the issue of<br />

treatment for older breast cancer patients in developed countries<br />

where aging has been rapidly advanced such as in Japan. According<br />

to Oxford Overview analysis of 15-year results, benefit of adjuvant<br />

chemotherapy in older than 70 years remains uncertain. Recently<br />

CALGB 49907 trial clearly showed standard chemotherapy of<br />

either cyclophosphamide, methotrexate, and fluorouracil (CMF) or<br />

doxorubicin plus cyclophosphamide (AC) is superior to capecitabine<br />

in patients with early-stage breast cancer who are 65 years or older.<br />

In contrast, we demonstrated equivalent efficacy between six cycles<br />

of classical CMF and 2 years of oral Tegafur-uracil (UFT) in phaseIII<br />

randomized controlled trial: N-SAS BC01 (Watanabe T et al, JCO<br />

2009). Of interest, UFT showed a trend toward better suppression<br />

of recurrence in patients over 50 years of age in this trial. In current<br />

exploratory analysis, we sought to examine whether UFT is not<br />

inferior to CMF in terms of efficacy, toxicity and quality of life (QOL)<br />

in older patients with early-stage breast cancer.<br />

Patients and Methods: N-SAS BC 01 trial was a randomly assigned<br />

trial comparing adjuvant oral UFT with classical CMF in patients with<br />

node negative, high risk breast cancer. In this exploratory analysis,<br />

patients of 65 years or older enrolled in N-SAS BC 01 trial were<br />

analyzed in terms of efficacy, toxicity and quality of life.<br />

Results: Of the 707 patients enrolled in N-SAS BC 01 trial, 97 patients<br />

(13.7%) were 65 years or older. Median age was 68 years (range, 65<br />

to 75 years). The 5-year relapse-free survival (RFS) rate was 92.5%<br />

in the CMF arm and 93.0% in the UFT arm. Overall survival (OS)<br />

rate at 5 years were 98.1% and 97.7%, respectively. The hazard ratios<br />

of the UFT arm relative to the CMF arm were 1.07 for RFS (95%<br />

CI, 0.31 to 3.55) and for OS (95% CI, 0.15 to 10.25). However 95<br />

% CIs were very wide due to the small sample size. Among patients<br />

who received CMF, frequency of grade 3/4 leukopenia (3.8%) and<br />

neutropenia (13.5%) were higher than 0% and 4.8% with UFT,<br />

respectively. Similarly, grade 3/4 increased liver enzyme and nausea/<br />

vomiting were more frequent with CMF than with UFT. In contrast,<br />

elevation of total bilirubin and diarrhea were more observed in UFT<br />

arm. Compared with patients received CMF, patients with UFT had<br />

better QOL scores assessed by EORTC QLQ-C30/BR23 and the<br />

FACT-B questionnaire. The rate of adherence was 79.2% (42/53) at 6<br />

months in the CMF arm and 74.4% (32/43) at 2 years in the UFT arm.<br />

Conclusion: The result of this study indicated that UFT might not<br />

be inferior to CMF in patient with early stage breast cancer who are<br />

65 years of age or older in terms of efficacy, toxicity and QOL. UFT<br />

would be a promising option for adjuvant chemotherapy in older<br />

women with node-negative, high-risk breast cancer. Further larger<br />

randomized clinical trial in this patient population would be warranted<br />

to validate these results.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

198s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 1<br />

P1-13-11<br />

Adjuvant treatment of early-stage breast cancer with eribulin<br />

mesylate following dose-dense doxorubicin and cyclophosphamide:<br />

preliminary results from a phase 2, single-arm feasibility study<br />

Traina TA, Hudis C, Fornier M, Lake D, Lehman R, Berkowitz AP,<br />

Rege J, Liao J, Cox D, Seidman AD. Memorial Sloan-Kettering<br />

<strong>Cancer</strong> Center, New York, NY; Eisai Inc, Woodcliff Lake, NJ<br />

Background: Despite recent improvements in breast cancer<br />

outcomes, patients (pts) with high-risk, early-stage breast cancer<br />

continue to experience recurrences and death due to disease.<br />

Chemotherapy regimens with the ability to extend survival remain an<br />

important drug development goal. Eribulin mesylate has demonstrated<br />

antitumor activity and improved overall survival (OS) in pts with<br />

heavily pretreated, locally recurrent or metastatic breast cancer when<br />

compared to treatment of physician’s choice. This study examines<br />

the feasibility of eribulin as adjuvant therapy following dose-dense<br />

(dd) doxorubicin (A) and cyclophosphamide (C) in patients with<br />

early-stage breast cancer.<br />

Methods: Eligible pts have histologically confirmed, HER2-normal,<br />

stage I-III invasive breast cancer and adequate bone marrow, liver, and<br />

renal function. Treatment consists of dd AC (A 60mg/m 2 IV; C 600mg/<br />

m 2 IV) on Day 1 of each 14-day cycle x4 cycles, followed by eribulin<br />

mesylate 1.4mg/m 2 IV over 2-5min on Days 1 and 8 every 21 days<br />

x4 cycles. Radiation/hormonal therapy were allowed per standard of<br />

care. The primary objective of feasibility is defined as the ability to<br />

complete 4 cycles of eribulin without a treatment-related dose delay<br />

(defined as >2 days) or reduction. Feasibility rates will be reported<br />

for pts with and without growth factor use. Secondary/exploratory<br />

endpoints include evaluation of the safety via NCI-CTCAEv4 of 4<br />

cycles of AC followed by 4 cycles of eribulin, and 3-year diseasefree<br />

survival and OS.<br />

Results: As of 5/22/12, 46 of 80 planned pts have been treated; 38<br />

pts have had ≥1 dose of eribulin and are evaluable for eribulin-related<br />

toxicity. Pt characteristics are as follows: median age 50 yrs (27-65<br />

yrs); 100% female; ECOG of 0=81.6%; breast cancer stage at study<br />

entry: stg 1: 7.5%; stg 2: 72.5%, stg 3: 20%. Select treatment-related<br />

AEs are reported as total (all cycles) and eribulin-related events; many<br />

AEs overlapped during treatment (Table).<br />

ALL CYCLES<br />

ERIBULIN CYCLES<br />

Adverse event (N=38) All events (%) Grade 3/4 (%) All events (%) Grade 3/4 (%)<br />

Peripheral neuropathy 50 3 42 3<br />

Fatigue 78 3 42 0<br />

Neutropenia 32 18 29 16<br />

Febrile neutropenia 8 8 3 3<br />

Leukopenia 16 13 18 10<br />

Alopecia 58 NA 21 NA<br />

Nausea 66 0 24 0<br />

Serious treatment-related AEs were reported in 2 pts, the most<br />

common (5.3%) being febrile neutropenia attributed to AC. Currently,<br />

13 pts have had eribulin dose modification or delays; 10 of the<br />

events were related to eribulin (7 reductions, 5 delays, 1 withdraw).<br />

Eribulin-related AEs associated with dose delay or reduction are:<br />

6 gr-3 neutropenia, 1 gr-3 febrile neutropenia, 1 gr-3 peripheral<br />

neuropathy, 1 gr-3 respiratory infection, 1 gr-3 fatigue. Six pts have<br />

discontinued (DC) treatment (2 AEs, 1 disease recurrence, 3 withdrew<br />

consent). Five of the 6 pts requiring eribulin delay/modification due<br />

to neutropenia were able to complete therapy with growth factor<br />

support. One pt DC eribulin therapy due to neuropathy.<br />

Conclusions: Preliminary results from this study suggest that adjuvant<br />

treatment with eribulin following dose-dense AC therapy has an<br />

acceptable safety profile. Accrual is ongoing and study completion<br />

is anticipated prior to SABCS 2012.<br />

P1-13-12<br />

Withdrawn by Author<br />

P1-13-13<br />

First planned efficacy analysis of the NNBC 3-Europe trial:<br />

Addition of docetaxel to anthracycline containing adjuvant<br />

chemotherapy in high risk node-negative breast cancer patients.<br />

Thomssen C, Kantelhardt EJ, Meisner C, Vetter M, Schmidt M, Martin<br />

P-M, Veyret C, Augustin D, Hanf V, Paepke D, Meinerz W, Hoffmann<br />

G, Wiest W, Sweep FCGJ, Schmitt M, Jaenicke F, von Minckwitz G,<br />

Harbeck N, On Behalf of the NNBC 3-Europe Study Group. Martin-<br />

Luther University Halle-Wittenberg, Halle an der Saale, Germany;<br />

Eberhard-Karls-University Tuebingen, Tuebingen, Germany;<br />

Johannes Gutenberg-Universität Mainz, Mainz, Germany; Medical<br />

Faculty Marseille, Marseille, France; Henri Becquerel Center, Rouen,<br />

France; Klinikum Deggendorf, Deggendorf, Germany; Klinikum<br />

Fürth, Fürth, Germany; Technical University Munich, München,<br />

Germany; St. Vincenz-Hospital, Paderborn, Germany; St. Josefs-<br />

Hospital, Wiesbaden, Germany; Katholisches Klinikum, Mainz,<br />

Germany; Radbourd University Nijmegen, Nijmegen, Netherlands;<br />

University Hamburg, Hamburg, Germany; German <strong>Breast</strong> Group,<br />

Neu-Isenburg, Germany; Ludwig-Maximilian-University, Munich,<br />

Germany<br />

Background: Risk assessment in node-negative (N0) breast<br />

cancer patients (pts) is routinely performed by established clinicopathological<br />

algorithms (CP) (Schmidt et al. Ann Oncol 2009). ASCO<br />

guidelines also recommend invasion markers uPA/PAI-1 (UP; Harris<br />

et al. JCO 2007). The prospective randomized multicenter NNBC<br />

3-Europe trial had two questions: 1) Does UP compared to the CP<br />

algorithm improve identification of low-risk node-negative breast<br />

cancer patients? 2) Does addition of taxanes to adjuvant chemotherapy<br />

improve DFS probability in high-risk node-negative breast cancer?<br />

Data are mature to report on the second question only.<br />

Methods: Between 2002 and 2009, 4,147 node-negative breast cancer<br />

pts were recruited. Risk assessment was performed by CP (43 %) or<br />

by UP (57 %). High-risk pts were randomized to receive FE 100 C*3-<br />

Doc*3 (FEC-D) versus FE 100 C*6 (FEC) as adjuvant chemotherapy.<br />

Primary endpoint was disease-free survival probability (DFS). It was<br />

planned to accrue 2,572 pts allowing detection of 4% reduction in<br />

recurrence probability at 5 years with a power of 80 % at a level of<br />

α=0.05 (LogRank) after a minimum observation time of 2.5 years<br />

for every recruited patient.<br />

Results: In the intent-to-treat population (ITT, n=2,541), 1,286 highrisk<br />

pts received FEC-D, and 1,255 received FEC. In July 2012, 90.8<br />

% of patients had reached the minimum observation time – median<br />

follow-up was 43.8, and 44.4 months. Main patients’ characteristics<br />

were distributed similarly between the two chemotherapy arms:<br />

median age was 53 yrs, 37.4 % of pts were premenopausal, median<br />

tumor size was 1.90 cm, grade 3 was seen in 53.8 %, hormone<br />

receptor status was negative in 30.1 % and HER2 was overexpressed<br />

in 20.0 %. Mastectomy was performed in 11 %. All planned courses<br />

of FEC-D were given in 84.4 %, of FEC in 91.4 % (p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P1-14-01<br />

Adding capecitabine and trastuzumab to neoadjuvant breast<br />

cancer chemotherapy - first survival analysis of the GBG/AGO<br />

intergroup-study GeparQuattro.<br />

von Minckwitz G, Rezai M, Loibl S, Fasching PA, Huober J, Tesch<br />

H, Bauerfeind I, Hilfrich J, Eidtmann H, Gerber B, Hanusch C,<br />

Blohmer J-U, Costa S-D, Jackisch C, Paepke S, Schneeweiss A,<br />

Kuemmel S, Denkert C, Mehta K, Untch M. German <strong>Breast</strong> Group,<br />

Neu-Isenburg; Louisenkrankenhaus Düsseldorf; University Erlangen;<br />

University Duesseldorf; Bethanien-Kankenhaus Frankfurt; Klinikum<br />

Landshut; Eilenriedeklinik Duesseldorf; University Kiel; University<br />

Rostock; Roteskreuzklinikum Muenchen; <strong>San</strong>kt Gertrauden Berlin;<br />

University Magdeburg; Klinikum Offenbach; Frauenklinik München;<br />

University Heidelberg; Kliniken Essen Mitte; Charite Berlin; Helios<br />

Kliniken Berlin<br />

Background: Previous results of the GeparQuattro study<br />

demonstrated that adding capecitabine either simultaneously or<br />

sequentially to EC-Docetaxel (D) neoadjuvant chemotherapy could<br />

not increase pathological complete response rates (pCR) (von<br />

Minckwitz G, JCO 2010). However, patients with HER2-positive<br />

disease treated simultaneously with trastuzumab showed a significant<br />

higher pCR rate than patients with HER2-negative disease treated with<br />

chemotherapy alone (Untch M, JCO 2010). We here report survival<br />

after a median follow up of 58 months including 279 relapses and<br />

191 deaths.<br />

Patients and methods: Patients with either large operable (cT3) and<br />

locally advanced (cT4) tumors, or hormone-receptor (HR)-negative<br />

receptor status, or HR-positive tumors but clinically node-positive<br />

disease were recruited to receive 4 cycles of EC (90mg/m 2 / 600mg/<br />

m 2 ) and randomized to either 4 cycles of D (100mg/m 2 ) or 4 cycles<br />

of DX (75mg/m 2 / 1800mg/m 2 ) or 4 cycles of D (75mg/m 2 ) followed<br />

by 4 cycles of X (1800mg/m 2 ) (D→X). Patients with HER-2 positive<br />

tumors received 1 year of trastuzumab, the first part concurrent to<br />

all chemotherapy cycles. All patients with HR+ tumors received<br />

endocrine therapy according to current standard. The intent-totreat<br />

survival analysis included 1421 patients for the chemotherapy<br />

question and 1495 patients for the trastuzumab question. Analyses<br />

were adjusted by age, stage, size, nodal status, histologic type, grade,<br />

hormone-receptor (HR) and HER2-status at baseline (if applicable).<br />

Results: No difference in DFS and OS was seen for patients receiving<br />

D, DX or D-X overall (hazard ratio 0.978, P=0.984 and hazard<br />

ratio 0.986, P=0.684, respectively) as well as by phenotype defined<br />

according to St. Gallen (all P>0.354).<br />

Patients with HER2-positive disease treated additionally with<br />

trastuzumab showed significantly better OS (P=0.015) compared<br />

to patients with HER2-negative disease treated with chemotherapy<br />

alone. DFS was significantly better for trastuzumab-treated patients<br />

with HR-negative tumors (P=0.046), but not with HR-positive<br />

tumors (P=0.790). OS after first relapse was significantly better in<br />

trastuzumab-re-treated patients with HER2-positive tumors (P=0.032)<br />

compared to relapsed patients with HER2-negative tumors.<br />

Patients with an early response after 4 cycles, with a clinical response<br />

at surgery and with a pCR showed a significantly better DFS and OS<br />

compared to patients without pCR (P=0.022, P


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 1<br />

breast cancer. Treatment discontinuation rate was significantly higher<br />

in TX than T group, however the pCR rate in patients in TX group<br />

who required treatment discontinuation or dose-reduction was similar<br />

to that in patients who completed as scheduled, which was different<br />

from T group. Determination of pre-/ post-treatment Ki67LI looks<br />

useful for predicting pCR and DFS.<br />

P1-14-03<br />

Overall survival results of a multicenter randomized phase II<br />

study in locally advanced breast cancer patients treated with<br />

or without neoadjuvant Trastuzumab for HER2 positive tumor<br />

(Remagus 02 trial)<br />

Giacchetti S, Pierga J-Y, Asselain B, Delaloge S, Brain E, Espié M,<br />

Mathieu M-C, Bertheau p, de Cremoux P, Tembo O, Marty M. Hôpital<br />

Saint Louis, Assitance Publique-Hôpitaux, Paris, France; Institut<br />

Curie, Paris, France; Institut Gustave Roussy, Villejuif, France;<br />

Institut Curie-Saint Cloud, Saint Cloud, France<br />

Background: Trastuzumab is indicated in neoadjuvant setting in<br />

locally advanced HER2 positive breast cancer patients (Gianni L.<br />

Lancet 2010). There is no data on the impact of the use of neoadjuvant<br />

Trastuzumab (T) compared to adjuvant T on survival.<br />

Patients and methods: From May 2004 to October 2007, 341<br />

stage II-III breast cancer patients were included in a phase II<br />

randomized trial and received 4 cycles (c) of epirubicin (75 mg/m2<br />

d1)–cyclophosphamide (750 mg/m2 d1) q 3 w followed by 4 (c) of<br />

docetaxel (100 mg/m2 d1) q 3 w. Pts with HER2+++ tumor (120<br />

pts) were randomized to receive or not neoadjuvant T combined<br />

with docetaxel. From September 2005, all pts with HER2+cancer<br />

received adjuvant T for a total of 18 c (106 pts). All pts with hormone<br />

receptors positive tumor received hormonal treatment according to<br />

menopausal status (Pierga et al BCRT 2010). We report here overall<br />

survival (OS) and disease free survival (DFS) data at 5 year and<br />

associated prognostic factors.<br />

Results: At a median follow up of 49 months, the median DFS was<br />

not reached for the whole population and was statistically superior<br />

for the HER2 positive cancer patients treated with chemotherapy plus<br />

neoadjuvant T compared to the other groups, p=0.018 . The median<br />

OS is not reached for the whole population and is statistically higher<br />

in HER2 positive tumor group compared to HER2 negative group<br />

(p=0.00077). For 106 HER2 positive breast cancer patients who had<br />

received one year of complete trastuzumab treatment, there was no<br />

significant difference in OS and DFS between pts who started T in<br />

neoadjuvant setting versus in adjuvant setting. DFS and OS were<br />

not significantly influenced by pathological Complete Response<br />

rate (pCR) (respectively, p = 0.22 and p = 0.56). At multivariate<br />

analysis including 6 factors (age, tumor size, clinical lymph node,<br />

ER, PgR), factors which influenced OS were tumor size (p =0.03)<br />

and ER expression (p = 0.06) and for DFS,clinical lymph node status<br />

(p=0.049) and PgR expression (p = 0.046).<br />

Conclusion: pCR is not a surrogate of survival in the HER2+subgroup.<br />

HER2 positive breast cancer pts receiving trastuzumab have a<br />

significant higher OS than those with HER2 negative tumors. OS<br />

and DFS do not seem to differ between the neoadjuvant T group and<br />

the T adjuvant group.<br />

P1-14-04<br />

Long- term outcome after neoadjuvant radiochemotherapy in<br />

locally advanced non-inflammatory breast cancer and predictive<br />

factors for a pathologic complete remission;<br />

results of a multi-variate analysis<br />

Matuschek C, Boelke E, Roth S, Bojar H, Audretsch W, Janni JW,<br />

Nestle-Kraemling C, Sauer R, Speer V, Budach W. Heinrich Heine<br />

University, Düsseldorf, Germany; European Institute for Molecular<br />

Oncology, Düsseldorf, Germany; Marien Hospital, Düsseldorf,<br />

Germany; Krankenhaus Gerresheim, Düsseldorf; University of<br />

Erlangen, Germany<br />

Background: An earlier published series of neoadjuvant radiochemotherapy<br />

(NRT-CHX) in locally advanced non-inflammatory<br />

breast cancer (LABC) has now been updated with a follow up of<br />

more than 15 years. Long- term outcome data and predictive factors<br />

for pathologic complete response (PCR) were analyzed.<br />

Patients and Methods: 315 LABC patients (cT1-cT4 /cN0-N1) were<br />

treated during 1991-1998 with NRT-CHX. Preoperative radiotherapy<br />

(RT) consisted of external beam radiation therapy (EBRT) of 50 Gy<br />

(5 × 2 Gy/week) to the breast and the supra-/infraclavicular lymph<br />

nodes combined with an electron boost in 214 cases afterwards or – in<br />

case of breast conservation – an 10-Gy interstitial boost with 192Ir<br />

afterloading before EBRT. Chemotherapy was administered prior to<br />

RT in 192 patients, and concomitantly in 113; 10 patients received no<br />

chemotherapy. The update of all follow up ended in November 2011.<br />

Age, tumor grade, nodal status, hormone receptor status, simultaneous<br />

vs. sequential CHX and the time interval between end of RT and<br />

surgery were examined in multivariate terms with as endpoint pCR<br />

and overall survival.<br />

Results: The total PCR rate after neoadjuvant RT-CHX reached 29.2<br />

% with LABC breast conservation becoming possible in 50.8%. In<br />

initially node-positive cases (cN+), a complete nodal response (pN0)<br />

after NRT-CHX was observed in 56% (89/159). The multivariate<br />

analysis revealed that a longer time interval to surgery increased<br />

the probability for a pCR (HR 1,17 [95% CI 1,05-1,31], p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

associated with a high incidence of cardiotoxicity. The risk of cardiac<br />

damage can be significantly reduced through liposomal encapsulation<br />

of anthracyclines. This phase I/II study was initiated to evaluate<br />

the combination of non-pegylated liposomal doxorubicin (NPLD),<br />

paclitaxel and lapatinib as primary treatment for patients with early<br />

stage, HER2-positive primary breast cancer.<br />

Patients and Methods: Patients with newly diagnosed HER2-positive<br />

(IHC 3+ or FISH+) early stage (T1c N1-2 or T2 N0-2) breast cancer<br />

were treated with NPLD (60mg/m 2 ; day 1), paclitaxel (175mg/m 2 , day<br />

1) and lapatinib (750-1500 mg orally daily) in 3-week intervals for up<br />

to 6 cycles. The primary endpoints were dose-limiting toxicities (DLT)<br />

and pathological complete response (pCR). Secondary endpoints<br />

include safety, incidence of cardiac events, and clinical response.<br />

Exploratory endpoints include molecular markers for sensitivity or<br />

resistance to chemotherapy and/or lapatinib evaluated.<br />

Results: A total of 84 patients have been included. No dose-limiting<br />

toxicity were observed and the maximum tolerated doses were NPLD<br />

60mg/m 2 , paclitaxel 175mg/m 2 and lapatinib 1500mg. Recommended<br />

phase 2 doses (P2D) were NPLD 60mg/m 2 , paclitaxel 175mg/m 2 and<br />

lapatinib 1250mg. The treatment was generally well tolerated and<br />

associated with toxicities that were consistent with the known sideeffects<br />

of the individual agents. No cardiac event has been observed<br />

to date. Preliminary efficacy data confirm a pCR breast rate of 41.7%<br />

and pCR rate in breast and lymph nodes of 37.5%, in 32 evaluable<br />

patients treated at ≥P2D.<br />

Conclusions: The combination of NPLD, paclitaxel and lapatinib is<br />

well tolerated and has high antitumor activity in patients with HER2-<br />

positive primary breast cancer. Updated results of all 84 patients will<br />

be presented.<br />

P1-14-06<br />

Significance of examining biomarkers of residual tumors after<br />

neoadjuvant chemotherapy using trastuzumab in combination<br />

with anthracycline and taxane in patients with primary HER2-<br />

positive breast cancer<br />

Kurozumi S, Takei H, Inoue K, Matsumoto H, Hayashi Y, Ninomiya<br />

J, Kubo K, Tsuboi M, Nagai S, Ookubo F, Oba H, Kurosumi M,<br />

Horiguchi J, Takeyoshi I. Saitama <strong>Cancer</strong> Center, Saitama, Japan;<br />

Gunma University Graduate School of Medicine, Gunma, Japan<br />

Background: Neoadjuvant chemotherapy (NAC) with taxane and<br />

FEC concurrently with trastuzumab is a potent regimen in women<br />

with HER2-positive breast cancer (BC), and several studies revealed<br />

high pCR rates in BC patients treated with this regimen. In the present<br />

study, we compared the status of biomarkers before and after NAC,<br />

and evaluated rates and patterns of discordant biomarker expression.<br />

We also evaluated differences of prognosis between patients<br />

with discordant biomarker expression and those with concordant<br />

expression.<br />

Patients and Methods: We investigated 118 Japanese women with<br />

invasive HER2 positive BC. Patients received 12 cycles of paclitaxel<br />

or 4 cycles of docetaxel followed by 4 cycles of FEC-75 with<br />

concomitant trastuzumab for 24 weeks and were followed for ≥1<br />

year after surgery. Of these, 27 patients with residual tumors 5 mm<br />

or larger were analyzed. HER2, ER, PgR, and Ki67 were examined<br />

in primary and residual tumors. Furthermore, recurrence-free survival<br />

(RFS) and overall survival (OS) were analyzed between patients<br />

classified based on these biomarkers.<br />

Results: Patients with pCR after NAC (75/118; 63.5%) had<br />

significantly better RFS than non-pCR patients (median follow-up:<br />

41 months). Residual tumors were obtained from 27 of 43 nonpCR<br />

patients and examined for immunohistochemical biomarker<br />

expression. In 14/27 non-pCR patients (51.9%), residual tumors were<br />

HER2 negative, despite being HER2 positive before NAC: HER2<br />

score changed from 3+ to 0 or 1+ in 8/18 patients (44.4%) and from<br />

2+ to 0 or 1+ in 6/9 (66.7%). ER expression changed in 2 patients (1<br />

positive to negative and 1 negative to positive). Patterns of biomarker<br />

expression in residual tumors were HER2 (+)/ER (–), 6 patients<br />

(22.2%); HER2 (+)/ER (+), 7 (25.9%); HER2 (–)/ER (+), 11 (40.7%);<br />

and triple negative (TN), 3 (11.1%). Recurrence was observed in 8/27<br />

(29.6%) non-pCR patients, and patterns of biomarker expression in<br />

residual tumors were HER2 (+)/ER (–), 3 patients; HER2 (+)/ER (+),<br />

2; and HER2 (–)/ER (+), 3. In addition, 1 patient with a HER2 (+)/<br />

ER (+) tumor and 1 patient with a HER2 (-)/ER (+) tumor died. RFS<br />

and OS were not statistically different between patients classified<br />

based on ER and Ki67 expressions. However, in the 18 non-pCR<br />

patients with primary tumor HER2 score of 3+ (overexpression of<br />

HER2 protein), the 10 with HER2-positive residual tumors showed<br />

significantly lower RFS than the 8 with HER2-negative (p


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 1<br />

Results In total, 201 patients (n=100 AC-T, n=101 TAC) were enrolled<br />

between February 2006 and April 2009. Baseline characteristics (AC-<br />

T/TAC) were well balanced. The clinical overall response rate (cCR<br />

and cPR) to 4 AC was comparable to that seen with 3 cycles TAC<br />

(48% versus 54%). Also, the clinical overall response rate to 8 AC-T<br />

was comparable to that seen with 6 TAC (81% versus 72%). In the<br />

AC-T arm, a third of the patients with cSD halfway reached a cCR<br />

or cPR after 8 cycles, whereas in the TAC arm only 18% of patients<br />

with cSD halfway reached a cCR or cPR after chemotherapy. In the<br />

sequential AC-T arm, 16% of the patients with cSD halfway reached<br />

a pCR after switching to docetaxel monotherapy. In contrast, in the<br />

TAC arm cSD halfway rarely resulted in pCR after 6 cycles. Patients<br />

with a cCR after chemotherapy had a pCR in 58% and 49% of cases,<br />

respectively. For patients with cPR these rates were 29% and 18%,<br />

respectively, and for patients with cSD 6% and 0%, respectively.<br />

Conclusion<br />

Clinical response rates are related to pCR rates. The meaning of<br />

cSD halfway should be differently interpreted for sequential versus<br />

concurrent chemotherapy. In concurrent chemotherapy schedules,<br />

a switch to a non-cross resistant drug may be worthwhile. Early<br />

clinical response may be used as a decision aid during neoadjuvant<br />

chemotherapy to recognize non-responders.<br />

Support: Unrestricted grants from sanofi-aventis NL BV and Amgen<br />

BV. Dutch breast cancer trialists’ group (BOOG).<br />

P1-14-08<br />

A prospective multicenter randomized phase II neo-adjuvant<br />

study of 5-fluorouracil, epirubicin and cyclophosphamide (FEC)<br />

followed by docetaxel, cyclophosphamide and trastuzumab (TCH)<br />

versus TCH followed by FEC versus TCH alone, in patients (pts)<br />

with operable HER2 positive breast cancer: JBCRG-10 study<br />

Masuda N, Sato N, Higaki K, Kashiwaba M, Matsunami N, Takano T,<br />

Yamamura J, Kaneko K, Takahashi M, Ohno S, Fujisawa T, Tsuyuki<br />

S, Miyoshi Y, Ohtani S, Yamamoto Y, Bando H, Onoda T, Kawabata<br />

H, Morita S, Ueno T, Toi M. NHO Osaka National Hospital, Osaka,<br />

Japan; Niigata <strong>Cancer</strong> Center Hospital, Niigata, Japan; Hiroshima<br />

City Hospital, Hiroshima, Japan; Iwate Medical University, Morioka,<br />

Japan; Osaka Rosai Hospital, Sakai, Japan; Toranomon Hospital,<br />

Tokyo, Japan; Hokkaido <strong>Cancer</strong> Center, Sapporo, Japan; National<br />

Kyushu <strong>Cancer</strong> Center, Fukuoka, Japan; Gunma Prefectural <strong>Cancer</strong><br />

Center, Ohta, Japan; Osaka Red Cross Hospital, Osaka, Japan;<br />

Hyogo College of Medicine, Nishinomiya, Japan; Kumamoto<br />

University Hospital, Kumamoto, Japan; University of Tsukuba,<br />

Faculty of Medicine, Tsukuba, Japan; Yokohama Asahi Central<br />

General Hospital, Yokohama, Japan; Yokohama City University<br />

Graduate School of Medicine and Medical Center, Yokohama, Japan;<br />

Kyoto University, Kyoto, Japan<br />

Background: The current standard treatment of primary systemic<br />

therapy (PST) in HER2 positive breast cancer is anthracyclines (A)<br />

and/or taxanes combined with trastuzumab (H) which demonstrates<br />

high pathological complete response (pCR). The pCR is considered<br />

as a predictive marker of prognosis although results are slightly<br />

different depending on the hormone receptor status. We conducted<br />

a randomized phase II study to examine sequence of treatments and<br />

necessity of A in the treatments using TCH to improve outcome and<br />

reduce cardiac toxicity in Japanese HER2 positive pts.<br />

Methods: Pts were treated with FEC (5FU 500 mg/m 2 , epirubicin 100<br />

mg/m 2 , cyclophosphamide 500 mg/m 2 ) and/or TCH (docetaxel 75 mg/<br />

m 2 , cyclophosphamide 600 mg/m 2 , H 6 mg/kg, loading by 8 mg) in<br />

3 groups: 4 cycles of FEC followed by 4 cycles of TCH (A-TCH); 4<br />

cycles of TCH followed by 4 cycles of FEC (TCH-A) or 6 cycles of<br />

TCH. An unplanned interim analysis was conducted due to one death<br />

by interstitial lung disease (ILD) in the A-TCH after completion of<br />

8 cycles. The pCR results suggested A containing regimens did not<br />

exceed benefit from the current standard regimen. The study was<br />

continued by limiting allocation only to the TCH group considering<br />

efficacy and safety. The primary endpoint was pCR and secondary<br />

endpoints were overall response rate (ORR) and safety.<br />

Results: A total of 103 pts were enrolled between Sep. 2009 and Sep.<br />

2011; 21 pts in the A-TCH, 22 pts in the TCH-A and 60 pts in the<br />

TCH including pts enrolled after termination of random allocation.<br />

Characteristics of the 103 pts were; median age of 54 (range, 33-70),<br />

median tumor size of 35 mm (range, 12-80), 42 pts with N(+) (40.8%)<br />

and 62 ER positive pts (60.2%). Characteristics of pts in the TCH<br />

were; median age of 54.5 (range, 33-67), median tumor size of 35.5<br />

mm (range, 12-80), 25 pts with N(+) (41.7%) and 34 ER positive pts<br />

(56.7%). No major difference was reported between groups treated<br />

with or without A. Per protocol population was 59 pts in the TCH and<br />

its pCR rate was 45.8% (95% CI, 32.2-59.3: ER negative, 61.5%; ER<br />

positive, 33.3%). ORR was 86.4% assessed by MRI or CT. Although<br />

it is an exploratory analysis, the pCR rate of A containing regimens<br />

was 39.0% (ER negative, 57.1%; ER positive, 29.6%). Adverse events<br />

≥grade 3 were reported in 50 pts (48.5%). Reported ILD was in 5 pts<br />

(A-TCH,1; TCH-A, 1; TCH, 3). The mean left ventricular ejection<br />

fraction (LVEF) decreased from 70.0% to 69.0% after treatment<br />

(A-TCH, 65.9%; TCH-A, 70.4%; TCH, 69.0%). Decrease of LVEF<br />

in the A-TCH was significant (p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

docetaxel (60 mg/m 2 on day 8 every 3 weeks) with capecitabine (1650<br />

mg/m 2 on days 1-14 every 3 weeks) followed by four cycles of FEC.<br />

Subtypes were classified into luminal A (lumA) (ER+ and/or PgR+,<br />

HER2-, Ki6714%), triple-positive (TP) (ER+ and/or PgR+, HER2+), HER2<br />

(ER-, PgR-, HER2+), and triple-negative (TN) (ER-, PgR-, HER2-)<br />

by using immunohistochemically stained specimen obtained by core<br />

needle biopsy. Absence of invasive tumor cells in the breast at the<br />

time of surgery was defined as pCR.<br />

Results<br />

We found 34 women in lumA, 54 in lumB, 15 in TP, 8 in HER2,<br />

33 in TN. The median age in each group was 53, 51, 50, 57 and<br />

53 respectively (p=.397). The mean tumor size was 3.2cm, 3.5cm,<br />

4.4cm, 3.9cm and 3.6cm respectively (p=.158). The clinical response<br />

rate measured by physical examination (calliper) was 80.6%, 93.9%,<br />

86.7%, 100% and 82.4% (p=.378). pCR rate was 5.9%, 14.8%.<br />

13.3%, 50%, 21.2% respectively (p=.031). The multivariate analysis<br />

showed ER negative, PgR negative and intrinsic subtype (TN) were<br />

the significant predictive factors of pCR (p=.027, p=.003, p=.004).<br />

Conclusion<br />

This study indicated that immunohistochemical classification of<br />

intrinsic subtype might be a useful predictive biomarker of pCR in<br />

breast cancer patients treated with preoperative chemotherapy.<br />

P1-14-10<br />

Final results of neoadjuvant trial of bevacizumab (B) and<br />

trastuzumab (T) in combination with weekly paclitaxel (P) as<br />

neoadjuvant treatment in HER2-positive breast cancer: A phase<br />

II trial (AVANTHER)<br />

Fernandez M, Calvo I, Martinez N, Herrero M, Quijano Y, Duran H,<br />

Garcia-Aranda M, Suarez A, Lopez-Rios F, Perez D, Perea S, Hidalgo<br />

M, Garcia-Estevez L. Centro Integral Oncológico Clara Campal,<br />

Madrid, Spain; Hospital Ramón y Cajal, Madrid, Spain; Hospital<br />

Costa del Sol, Marbella, Spain<br />

Background: B in combination with T has shown meaningful activity<br />

in patients (pts) with metastatic HER2-positive breast cancer.<br />

Pathological complete response (pCR) was defined as the absence<br />

of invasive disease at the time of histological study and there is a<br />

relationship between pCR and disease-free survival (DFS) and overall<br />

survival (OS). The complete pathological response rate is between 9<br />

to 34% of patients receiving primary treatment prior to surgery with<br />

chemotherapy and when the Herceptin was administrated, a pCR of<br />

39% was reached.<br />

AVANTHER is a Phase II trial of preoperative systemic therapy<br />

combining B with T and P in a weekly regimen in HER2 positive<br />

breast cancer to assess safety and efficacy of the combination.<br />

Methods: Pts with centrally-confirmed HER2- positive (IHC 3+<br />

or FISH positive) breast cancer (stage II or III including locally<br />

advanced) received neoadjuvant chemotherapy (NC) with weekly P<br />

(80mg/m2/week) for 12 weeks in combination with weekly T (4mg/<br />

kg loading dose and 2 mg/kg maintenance) and B (15mg/kg every<br />

3 weeks) for 4 cycles. After surgery all pts received T (1 year) and<br />

liposomal doxorubicin plus cyclophosphamide every 3 weeks (4<br />

cycles); primary endpoint was rate of pathological complete response<br />

(pCR) in breast and axilla. For all patients, a tissue sample at baseline<br />

as well as at surgery was collected for biomarker analyses.<br />

Results: A total of 44 pts have been enrolled. Median tumor size: 3.9<br />

cm. Nine (21%) pts had stage IIA; 19 (45%) stage IIB; 10 (24%) stage<br />

IIIA and 4 (10%) stage IIIB. Twenty-nine (69%) pts had estrogen<br />

positive receptors.<br />

Data from surgery of 42 patients from a total of 44 patients enrolled<br />

were presented in this abstract, but the final results will be present<br />

in the symposium. pCR was achieved in 18 (42.9%) pts. Safety and<br />

tolerability were good, with rare adverse events of grade 3 [1 (2.4%)<br />

episode of severe hypertension, 1 (2,4%) mucosal inflammation].<br />

We presented the results of relationship between magnetic resonance<br />

imaging (MRI) and pCR that is one of the secondary objectives of<br />

the study.<br />

100% of the patients without pathologic response had stable disease<br />

at resonance imaging. Of all patients who had pCR only 55.6% had<br />

complete radiological response.<br />

Conclusions: These data show that the combination of P with T and<br />

B without an anthracycline for 12 weeks is very effective as NC in<br />

HER2 positive breast cancer pts with a high rate of pCR and minimal<br />

side effects.<br />

And the MRI is useful for identifying the persistence of residual<br />

disease however it only predicts half of the complete pathological<br />

responses<br />

P1-14-11<br />

Assessing Prognosis and Therapy Response in Primary Systemic<br />

Therapy of <strong>Breast</strong> <strong>Cancer</strong> with Magnetic Resonance Spectroscopy<br />

Bolan PJ, Wey A, Eberly LE, Nelson MT, Haddad TC, Yee D, Garwood<br />

M. University of Minnesota, Minneapolis, MN; Masonic <strong>Cancer</strong><br />

Center, University of Minnesota, Minneapolis, MN<br />

PURPOSE: We have previously reported results of a pilot study<br />

in patients with locally advanced breast cancer receiving primary<br />

systemic chemotherapy (PST) showing that a decrease in tumor<br />

choline concentration ([tCho]) measured one day after starting<br />

treatment was associated with clinical response based on MRI. In<br />

this follow-up study with a larger cohort, we assessed whether early<br />

changes in [tCho] were associated with clinical response, based on<br />

both MRI and pathology, and survival.<br />

METHODS AND MATERIALS: Women with locally advanced<br />

breast cancer scheduled for PST were scanned using a high-field MRI/<br />

MRS protocol prior to treatment, day 1 after treatment, and at the end<br />

of the first course of chemotherapy. MRI/MRS was performed on a<br />

4 T Varian system with unilateral transmit/receive breast coils. MRI<br />

consisted of contrast-enhanced T1-weighted 3D gradient echo scans<br />

with one-minute temporal resolution. A single-voxel MR spectrum<br />

was acquired from the index lesion after MRI, and the concentration<br />

of total choline compounds ([tCho]) was quantified using water as<br />

an internal reference. Clinical response was assessed using RECIST<br />

criteria for measuring the longest diameter (LD) of the index lesion,<br />

and by pathologic complete response (pCR) at definitive surgery.<br />

Overall survival (OS) and invasive disease free survival (IDFS) were<br />

assessed by retrospective review of clinical records and the social<br />

security death index. The associations of change in [tCho] with pCR/<br />

LD response and with IDFS/OS were assessed using logistic/Cox<br />

regression, respectively, adjusted for node positivity and pCR status.<br />

RESULTS: Of the 74 women enrolled in the trial, 51 women (ages<br />

28-71, median 47 years) were scanned at all time points and included<br />

in the final analysis. Of these, 17 subjects had a partial or complete<br />

imaging RECIST response based on a ≥30% decrease of the LD of the<br />

index lesion. Pathologic complete response, defined as no detectable<br />

invasive disease in the breast, was observed in 13/51 overall subjects<br />

and in 6/35 hormone receptor positive (ER+ or PR+) subjects. Median<br />

follow-up time for survival analysis was 64 months (range 11-102<br />

months). Linear models showed no significant association between<br />

change in [tCho] and either pCR or RECIST response. An increase<br />

in [tCho] between the pretreatment scan and day 1 after treatment<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

204s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 1<br />

was found to be associated with reduced likelihood of both OS and<br />

IDFS. We found no association between pCR status and either OS or<br />

IDFS when considering all subjects and in the HR+ cohort (see Table<br />

1). No results could be reported for the HR- group (14 subjects) due<br />

to an insufficient number of events.<br />

Table 1 - Survival Hazard Ratios<br />

All<br />

HR+<br />

OS [tCho] ↑ 25% 1.94 (p=0.003) * 2.24 (p=0.020) *<br />

pCR 0.78 (p=0.746) 2.14 (p=0.334)<br />

IDFS [tCho] ↑ 25% 1.98 (p=0.001) * 2.02 (p=0.019) *<br />

pCR 1.15 (p=0.835) 1.87 (p=0.429)<br />

* statistically signficant at p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P1-14-14<br />

Neoadjuvant Sunitinib (S) Administered with Weekly Paclitaxel<br />

(P) /Carboplatin(C) in Patients (Pts) with Locally Advanced<br />

Triple-Negative <strong>Breast</strong> <strong>Cancer</strong> (TNBC): Preliminary Results<br />

from a Phase I/II Trial of the Sarah Cannon Research Institute.<br />

Yardley DA, Barton J, Hendricks C, Webb C, Priego V, Nimeh<br />

N, Gravenor D, Shastry M, Chirwa T, Burris HA. Sarah Cannon<br />

Research Institute, Nashville, TN; Tennessee Oncology, PLLC,<br />

Nashville, TN; National Capital Clinical Research Consortium,<br />

Bethesda, MD; Baptist Hospital East, Louisville, KY; Center for<br />

<strong>Cancer</strong> and Blood Disorders, Bethesda, MD; <strong>Cancer</strong> Center of<br />

Southwest Oklahoma Research, Lawton, OK; Family <strong>Cancer</strong> Center<br />

Foundation, Inc., Memphis, TN<br />

Background: S is an oral, small molecule, multi-targeted tyrosine<br />

kinase inhibitor of vascular endothelial growth factor receptor,<br />

platelet-derived growth factor receptor, stem cell factor receptor,<br />

and colony-stimulating factor-1 receptor with demonstrated single<br />

agent activity in multiple tumor types including breast cancer. S’s<br />

anti-proliferative and anti-angiogenic activity stimulated interest<br />

in combining it with a variety of chemotherapy regimens. In the<br />

previously reported Phase I portion of this trial, S 25mg PO daily was<br />

determined feasible in combination with weekly P and C, an active<br />

regimen for TNBC. We now report the feasibility and efficacy of the<br />

phase II evaluation of this combination as neoadjuvant therapy with<br />

pathologic complete response (pCR) as the primary endpoint.<br />

Methods: Eligibility criteria included: invasive adenocarcinoma,<br />

triple negative histology defined as ER/ PR negative ( 5% of pts.<br />

Conclusions: The addition of sunitinib to paclitaxel and carboplatin<br />

was accompanied by significant hematologic toxicity resulting<br />

in the inability to deliver the planned study therapy. Treatment<br />

discontinuations were frequent as was the need for dose adjustments<br />

and modifications. The sunitinib-paclitaxel/carboplatin combination<br />

evaluated in this study is not recommended for further use in locally<br />

advanced breast cancer.<br />

P1-14-15<br />

Recent experience of neoadjuvant chemotherapy according to<br />

breast cancer subtype: experience from a large United Kindgom<br />

teaching hospital<br />

Rattay T, Kaushik M, Ahmed S, Shokuhi S. University Hospitals of<br />

Leicester, United Kingdom<br />

INTRODUCTION:<br />

The most recent United Kingdom (NICE, 2009) guidelines<br />

recommend neoadjuvant chemotherapy as the most appropriate initial<br />

treatment for locally advanced breast cancer followed by mastectomy.<br />

Neoadjuvant chemotherapy should also be offered to patients with<br />

early breast cancer who are considering breast conserving surgery that<br />

is not advisable at presentation. However, the evidence base is still<br />

gathering. This study reviews the recent experience of neoadjuvant<br />

chemotherapy in a large United Kingdom breast unit according to<br />

breast cancer subtype.<br />

METHODS:<br />

Retrospective chart review of patients who received neoadjuvant<br />

chemotherapy for breast cancer at University Hospitals of Leicester<br />

between January 2008 and March 2011. Patients were identified<br />

from the unit database. <strong>Breast</strong> cancers were typed according to<br />

immunehistochemical marker profile: luminal A (ER or PR positive,<br />

HER2 negative), luminal B (ER or PR positive, HER2 positive), triple<br />

negative (ER and PR negative, HER2 negative), and HER2 positive<br />

(ER and PR negative, HER2 positive).<br />

RESULTS:<br />

Of 2,489 new patients with breast cancer seen during the study period,<br />

153 patients received neoadjuvant chemotherapy. 20 patients had<br />

lobular-type cancers, and the remaining 133 patients had ductal- or<br />

mixed-type histology. 101 patients had locally advanced disease<br />

and 52 patients presented as early breast cancer. 24 of all patients<br />

(15 %) achieved complete pathological response. 18 patients (12 %)<br />

never proceeded to surgery, eight of whom died during the study<br />

period. In total to date, 23 patients (15 %) have died including one<br />

death due to complications from chemotherapy. Of 120 patients<br />

initially scheduled for mastectomy, the tumor was downsized in 26<br />

patients (22 %) so that they were able to undergo breast conserving<br />

surgery (BCS), with margins involved in six patients. The outcome<br />

of neoadjuvant chemotherapy according to breast cancer subtype is<br />

presented (Tables 1 and 2).<br />

Table 2<br />

Subtype No of patients pCR No pathological response Deaths<br />

Luminal A 73 3 4 % 22 30 % 8 11 %<br />

Luminal B 28 7 25 % 4 14 % 1 4 %<br />

Triple negative 35 10 29 % 4 11 % 10 29 %<br />

HER2+ 17 4 24 % 2 12 % 4 24 %<br />

Table 2<br />

Subtype no surgery<br />

initially for<br />

converted to BCS<br />

mastectomy<br />

positive margins<br />

Luminal A 6 8 % 57 12 21 % 4 33 %<br />

Luminal B 2 7 % 24 5 21 % 1 20 %<br />

Triple<br />

negative<br />

5 14 % 27 6 22 % 1 17 %<br />

HER2+ 5 29 % 12 3 25 % 0 0 %<br />

CONCLUSIONS:<br />

In our experience, patients receive neoadjuvant chemotherapy for<br />

breast cancer for a variety of indications. Compared to our standard<br />

population of breast cancer patients, early mortality remains relatively<br />

high, particlularly in the hormone receptor negative subtypes. BCS<br />

conversion rates were similar across breast cancer subtypes, but BCS<br />

was less likely to be successful in the Luminal A group. Luminal<br />

A cancers were also significantly less likely to achieve pCR after<br />

neoadjuvant chemotherapy. <strong>Breast</strong> cancer subtype should be taken<br />

into account when scheduling patients for neoadjuvant chemotherapy.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

206s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 1<br />

P1-14-16<br />

Young age: predicts poor response rate after neoadjuvant<br />

chemotherapy in endocrine-responsive breast cancer<br />

Kim MK, Noh D-Y, Han W, Moon H-G, Ahn S-K, Kim JS, Kim T,<br />

Kim JY, Lee JW. Seoul National University Hospital, Seoul, Korea<br />

Background; <strong>Breast</strong> cancer rarely occurs in very young<br />

women(age0.05); and concentric shrinkage mode was<br />

observed only in 1 case (3.9%). More diffusive residual DCIS was<br />

observed in pts with diffusive microcalcifications on mammography<br />

before NAC than those without (76.5% vs. 7.7%, κ=0.670, p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Patients and methods/: From May 2004 to October 2007, 340<br />

stage II-III breast cancer patients were included in a phase II<br />

randomized trial and received 4 cycles (c) of epirubicin (75 mg/<br />

m2)–cyclophosphamide (750 mg/m2) q 3 w followed by 4 (c) of<br />

docetaxel (100 mg/m2) q 3 w. Pts with HER2 negative tumors (220<br />

pts) were randomized to receive or not neoadjuvant celecoxib (200<br />

mg bid) combined with docetaxel. All pts with hormone receptors<br />

positive tumor received hormonal treatment according to menopausal<br />

status (Pierga et al BCRT 2010). We report here overall survival<br />

(OS) and disease free survival (DFS) data and prognostic factors<br />

analyses at 5 years.<br />

Results/: At a median follow up of 49 months, the median DFS and<br />

OS are not reached for the whole population and none of them is<br />

significantly different between pts who received celecoxib or who did<br />

not (p = respectively 0.62 and 0.36). Celecoxib had no impact either<br />

on clinical and pathological complete response rate (pCR). DFS is<br />

significantly higher in patients who achieved pCR as compared to<br />

those who did not (p = 0.017; RR = 0.21 [0.051-0.88], whereas OS<br />

is borderline significant [p = 0.07; RR = 0.19 (0.026-1.4)]. Patients<br />

with triple negative (TN) tumors (78 pts) achieved worst DFS (p=<br />

0.02) and OS (p


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 1<br />

Conclusion :. This NCT provided high pCR, specially in the HRsubgroup<br />

with a very favourable toxicity profile and merits further<br />

exploration.<br />

P1-15-01<br />

Patterns of granulocyte colony stimulating factor (G-CSF) use in<br />

elderly breast cancer (BC) patients receiving myelosuppressive<br />

chemotherapy<br />

Blaes AH, Chia V, Solid C, Page J, Barron RL, Choi MR, Arneson TJ.<br />

University of Minnesota, Minneapolis, MN; Amgen, Inc., Thousand<br />

Oaks, CA; Minneapolis Medical Research Foundation, Minneapolis,<br />

MN<br />

Background: Febrile neutropenia (FN) is a common and potentially<br />

serious complication of myelosuppressive chemotherapy treatment<br />

in cancer patients. Oncology guidelines recommend primary G-CSF<br />

prophylaxis (PPG) in patients with a high risk of developing FN,<br />

which is risk >20% based on myelotoxicity of the regimen itself<br />

or from a combination of the therapy, older age, comorbidities and<br />

disease characteristics (Lyman <strong>Cancer</strong> 2011). Current patterns of<br />

G-CSF use and FN occurrence among elderly patients receiving<br />

myelosuppressive chemotherapy for BC have not been previously<br />

reported. To determine this, we performed a retrospective analysis<br />

using a subset of the Medicare 5% database.<br />

Methods: The Medicare 5% claims data set (includes a representative<br />

5% systematic sample of Medicare beneficiaries) was used to identify<br />

BC patients age 65+ initiating chemotherapy between 7/1/2003<br />

and 6/30/2009. Chemotherapy courses were identified for each<br />

patient; only the first course was used for this analysis. Using the<br />

National Comprehensive <strong>Cancer</strong> Network guidelines on Myeloid<br />

Growth Factors (NCCN V1.2012), chemotherapy course regimens<br />

were classified as high risk (HR) or intermediate risk (IR) for FN.<br />

Duration of first cycle was from date of first chemotherapy claim<br />

to the chemotherapy claim at day 21 or later, which defined the<br />

first day of the second cycle, etc., to a maximum of 9 cycles. First<br />

administration of G-CSF [filgrastim (NEUPOGEN®) or pegfilgrastim<br />

(Neulasta®)] was classified as either PPG (within first 5 days of<br />

first cycle), secondary prophylaxis (within first 5 days of second or<br />

subsequent cycles), or reactive (day 6 or later of first or subsequent<br />

cycles). FN assessed during the chemotherapy course was defined as<br />

hospitalization with a code for neutropenia in any position.<br />

Results: 885 courses with high FN risk and 1046 with IR FN risk were<br />

identified. The HR cohort was younger (71.4 vs 74.5 yrs) and had<br />

fewer comorbidities than the IR cohort. Selected aspects of G-CSF<br />

use patterns are summarized in the table. Among HR courses, 11.8%<br />

had ≥1 FN hospitalization and 2.1% had 2+; among IR courses 5.6%<br />

had ≥1 and 0.4% had 2+.<br />

Conclusion: NCCN recommends PPG be used with HR regimens and<br />

older age (notably >65 yr), an important risk factor for developing<br />

severe neutropenic complications. Despite this, PPG was used for<br />

elderly breast cancer patients in only 52% of chemotherapy courses<br />

with high risk of FN and in 10% of IR courses. More than 10%<br />

of patients with a HR regimen had an FN hospitalization. Careful<br />

attention to FN risk factors, including regimen and patient age, is<br />

needed when planning treatment strategy.<br />

Any G-CSF Use During Course<br />

Regimen’s FN Risk Any G-CSF Any filgrastim Any pegfilgrastim Both G-CSFs No G-CSF<br />

High (n=885) 73.8% 20.1% 64.0% 10.3% 26.2%<br />

IR (n=1046) 30.9% 13.8% 21.5% 4.4% 69.1%<br />

First G-CSF Use<br />

Regimen’s FN Risk Primary Prophylaxis Secondary Prophylaxis Reactive<br />

High (n=885) 70.6% 11.9% 17.5%<br />

IR (n=1046) 31.6% 30.0% 38.5%<br />

HR regimens: TAC (389); dose dense AC→T (345);<br />

docetaxel+trastuzumab (61); doxorubicin+docetaxel (50);<br />

doxorubicin+paciltaxel (21); docetaxel q14(19). IR regimens: CMF<br />

classic (481); paclitaxel q21 (337); docetaxel q21 (94); paclitaxel+<br />

trastuzumab (87); FEC (47).<br />

P1-15-02<br />

Febrile neutropenia (FN) risk assessment and granulocyte colonystimulating<br />

factor (G-CSF) guideline adherence in patients with<br />

breast cancer – results from a German prospective multicentre<br />

observational study (PROTECT)<br />

Steffens C-C, Eschenburg H, Kurbacher C, Goehler T, Schmidt M,<br />

Eustermann H, Schaffrik M, Otremba B. MVZ für Hämatologie/<br />

Onkologie, Klinik Dr. Hancken; Praxis für Hämatologie und<br />

internistische Onkologie, Güstrow; Praxis für Gynäkologie und<br />

gynäkologische Onkologie, Medizinisches Zentrum Bonn; Praxis für<br />

Hämatologie und internistische Onkologie, Dresden; Gynäkologische<br />

Onkologie, Universitätsfrauenklinik Mainz; WiSP Wissenschaftlicher<br />

Service Pharma GmbH, Langenfeld; Amgen GmbH, München;<br />

Onkologische Praxis Oldenburg/Delmenhorst<br />

Background: FN is a dose-limiting toxicity of chemotherapy which<br />

can affect patient (pt) outcomes. EORTC guidelines recommend<br />

G-CSF primary prophylaxis when the overall FN risk assessment<br />

is ≥20%. We evaluated risk assessment and guideline adherence<br />

(prophylaxis in high-risk pts) in clinical practice among breast cancer<br />

patients.<br />

Methods: Eligible pts were enrolled consecutively at centres selected<br />

based on experience and geographical spread. Key inclusion criteria<br />

were diagnosis of solid tumour or lymphoma, physician-assessed<br />

overall FN risk ≥10% (chemotherapy risk plus individual risk factors<br />

per EORTC guidelines), and planned primary (PPP) or secondary<br />

(PSP) prophylaxis with pegfilgrastim. Pegfilgrastim administered >3<br />

days after chemotherapy completion was classified as therapeutic use.<br />

The primary objective was to describe the proportion of pts with an<br />

investigator-assessed overall FN risk of >20% or 10-20% receiving<br />

PPP or PSP. Secondary objectives included evaluating FN incidence<br />

and chemotherapy dose reductions/delays. Data from breast cancer<br />

patients only are reported, and prophylaxis in high-risk patients was<br />

determined to evaluate guideline adherence.<br />

Results: 1003 pts were enrolled from Nov 2007 to Sept 2010. Mean<br />

(±SD) age was 54.4 (±11.2) years, almost all pts (99%) were female.<br />

Treatment intent was deemed curative by the investigator in 91% of<br />

pts. The investigator-assessed FN risk was >20% in 505 (50%) pts<br />

and 10-20% in 414 (41%) pts. Despite eligibility criteria, 84 (8%) pts<br />

were assessed by investigators as 20% (N=505)<br />

Planned prophylaxis PPP 470 (93%)<br />

PSP 35 (7%)<br />

Given prophylaxis PPP 404 (80%)<br />

PSP 80 (16%)<br />

Therapeutic use or use not according to label 21 (4%)<br />

www.aacrjournals.org 209s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Conclusion: Adherence to G-CSF guidelines, and reasons for<br />

differences between planned and administered prophylaxis warrants<br />

further investigation. Comparison of outcomes between prophylaxis<br />

groups should be interpreted with caution; however, FN incidence<br />

remained low with pegfilgrastim PP among breast cancer pts in<br />

German clinical practice.<br />

P1-15-03<br />

Comparison of efficacy of primary prophylaxis with pegfilgrastim,<br />

filgrastrim and a biosimilar filgrastim in TAC regimen (docetaxel,<br />

doxorubicin and cyclophosphamide)<br />

Brito M, Esteves S, Andre R, Isidoro M, Moreira A. Portuguese <strong>Cancer</strong><br />

Institute Francisco Gentil, Lisbon, Portugal<br />

Background<br />

Febrile neutropenia (FN) is a major toxicity of myelosupressive<br />

chemotherapy. Primary prophylactic use of granulocyte colony<br />

stimulating factors (G-CSF) is recommended in high risk FN<br />

regimens.<br />

The comparison of pegfilgrastim (Peg) and filgrastim (Fil) FN<br />

prophylactic effectiveness is still an issue of debate. Very recently<br />

Nivestim (Niv), a new biosimilar filgrastim, has also become<br />

commercially available.<br />

We aimed to compare the efficacy of the 3 mentioned types of G-CSF<br />

in the primary prophylaxis of FN.<br />

Methods:<br />

Single-center, retrospective study to evaluate the incidence of FN in<br />

women with breast cancer treated with adjuvant or neo-adjuvant TAC<br />

(FN risk ≥20%). Patients (Pt) were divided in 3 consecutive cohorts<br />

according to G-CSF primary prophylaxis (Fil, Peg and Niv) FN was<br />

defined as axillary temperature ≥38,3 °C and absolute neutrophil<br />

count < 500/ul.<br />

Results:<br />

We included a total of 421 women (median age 51 y, 25-76) with<br />

Stage II (56%) and Stage III (44%) breast cancer. Age and stage<br />

distribution were similar in the 3 cohorts.<br />

A single dose of Peg was administered in all 767 cycles (cy). The<br />

standard dose of Fil and Niv was 7 daily injections, only in in 13%<br />

Fil pt and 10% Niv pt < 7 administrations were done.<br />

The incidence of FN per patient and per cycle is presented in Table 1.<br />

In all cohorts, approximately half of NF episodes occurred in the 1st<br />

cycle (48% Fil, 59% Peg, 42% Niv).<br />

Table 1<br />

Fil Peg Niv P-value<br />

(147 pts; 840 cy) (140 pts; 767 cy) (134 pts; 710 cy) Fisher test<br />

% Pt FN 15,6% (23/147) 8,6% (12/140) 13,4% (18/134) Niv vs Peg<br />

IC95%:10,4-2,8% IC95%:4,5-14,5% IC95%:8,2-20,4% p= 0.25<br />

Niv vs Fil<br />

p = 0.62<br />

Fil vs Peg<br />

p = 0.07<br />

% Cy FN 3,2% (27/840) 2,2% (17/767) 3,7% (26/710) Niv vs Peg<br />

IC95%: 2,2-4,7% IC95%: 1,3-3,5% IC95%: 2,4-5,3% p = 0.12<br />

Niv vs Fil<br />

p = 0.68<br />

Fil vs Peg<br />

p = 0.28<br />

Conclusions:<br />

No differences in terms of efficacy existed between Biosimilar Niv<br />

and original biological reference Fil.<br />

Seven daily injections of Fil and Niv seem equivalent to single dose<br />

Peg.<br />

Besides efficacy, questions like cost-effectiveness and convenience<br />

of administration should be taken into account when approaching<br />

this topic.<br />

Our data showed a predominance of events in the 1st cycle (regardless<br />

of the type of G-CSF). This has been consistently described in the<br />

literature and may support the necessity to recommend other NF<br />

preventive measures in this cycle.<br />

P1-15-04<br />

The relationship of relative dose intensity and supportive care<br />

to febrile neutropenia rates in patients with early stage breast<br />

cancer receiving chemotherapy: a prospective cohort study of<br />

chemotherapy-associated toxicity<br />

Culakova E, Poniewierski MS, Wogu AF, Kuderer NM, Crawford J,<br />

Dale DC, Lyman GH. Duke University, Durham, NC; University of<br />

Washington, Seattle, WA<br />

Background: Febrile neutropenia (FN) represents a major doselimiting<br />

toxicity of cancer chemotherapy resulting in considerable<br />

morbidity, mortality, and cost. Patients have the highest risk of the<br />

initial neutropenic event in cycle 1 when most patients receive full<br />

dose chemotherapy. This study evaluates time course of neutropenic<br />

events in patients receiving chemotherapy for early-stage breast<br />

cancer (ESBC) and supportive care interventions that modify FN risk<br />

in ESBC patients treated in actual oncology practice.<br />

Methods: A prospective cohort study of adult cancer patients with<br />

solid tumors or lymphoma starting a chemotherapy regimen was<br />

conducted at 115 U.S. sites. Toxicities associated with chemotherapy<br />

were recorded in up to 4 cycles including severe neutropenia (SN),<br />

FN, and infection. Documented clinical interventions included<br />

reductions in chemotherapy relative dose intensity (RDI), the use of<br />

colony-stimulating factors (CSFs), and antibiotics.<br />

Results: A total of 1202 ESBC patients starting chemotherapy were<br />

analyzed, of which 1154, 1099, and 896 reached the midcycle of<br />

cycles 2, 3, and 4, respectively. While the majority of neutropenic<br />

and infection events occurred in cycle 1, decreasing rates of FN and<br />

infection in later cycles correlated with increasing reductions in dose<br />

intensity and increased use of CSFs and antibiotics.<br />

Table 1: Cycle specific and cumulative events (all patients)<br />

Cycle 1 Cycle 2 Cycle 3 Cycles 1-4<br />

% % % %<br />

FN 9.7 5.7 3.8 16.3<br />

SN or FN 32.1 22.7 18.5 44.0<br />

Infection 14.9 10.9 8.0 27.0<br />

RDI < 85% 13.4 19.4 19.8 32.5<br />

Cumulative CSFs 26.7 51.2 60.8 62.1<br />

Cumulative antibiotics 3.8 20.9 29.1 33.0<br />

The overall risk of FN in all patients combined was 16.3 %. It reached<br />

21.1% for patients who started with planned RDI≥85% and without<br />

primary CSF prophylaxis. There was no significant difference in FN<br />

rates by menopausal status or hormone receptors.<br />

Table 2: Patients with planned RDI≥85% and no primary CSFs prophylaxis<br />

Cumulative<br />

events<br />

Pre-menop Post-menop HR+ HR-<br />

% % % %<br />

FN 21.1 21.1 19.9 22.4<br />

SN or FN 56.8 58.1 55.1 62.5<br />

Infection 28.0 29.5 27.4 31.5<br />

RDI < 85% 16.6 31.4 25.9 24.6<br />

Cumulative CSFs 51.9 52.6 51.9 52.2<br />

Cumulative<br />

antibiotics<br />

34.3 36.2 33.6 40.5<br />

HR=hormone receptors<br />

Conclusions: While the risk of neutropenic complications is highest<br />

during the first cycle of chemotherapy, reductions in neutropenic<br />

events during subsequent cycles are associated with reduced<br />

chemotherapy dose intensity or increased use of supportive care<br />

measures. Nevertheless, the cumulative risk of neutropenic events<br />

remains high in ESBC patients receiving full dose chemotherapy<br />

without prophylactic measures overall and across menopausal and<br />

hormone receptor subgroups.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

210s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 1<br />

P1-15-05<br />

GSTP1 polymorphism is associated with chemotherapy induced<br />

neuropathy<br />

Miltenburg NC, Opdam M, Winter M, van Geer M, Oosterkamp HM,<br />

Boogerd W, Linn SC. The Netherlands <strong>Cancer</strong> Institute, Amsterdam,<br />

Netherlands; The Netherlands <strong>Cancer</strong> Institute<br />

Background:<br />

Peripheral neuropathy is a common and potentially debilitating<br />

side-effect of microtubule-stabilizing agents like docetaxel and is<br />

characterized by sensory symptoms like numbness, tingling and<br />

burning pain. Docetaxel induced neuropathy may occur in up to<br />

37% of patients and can be dose limiting, thus potentially reducing<br />

its chemotherapeutic efficacy. Doxorubicine has occasionally been<br />

described to be associated with neuropathy.<br />

With this background we investigated the role of various single<br />

nucleotide polymorphisms (SNPs) to predict chemotherapy-induced<br />

neuropathy. The exploration of SNPs and an association with<br />

incidence or severity of neuropathy is important, because it can<br />

result in identifying individuals being at higher risk of neurotoxicity.<br />

Methods:<br />

The MATADOR study is a prospective, non-blinded randomized<br />

phase II/III trial for patients with invasive breast cancer, comparing<br />

TAC (docetaxel 75 mg/m² as 1 hour i.v. infusion q 3 weeks, in<br />

combination with doxorubicin and cyclophosphamide for 6 cycles)<br />

with AC dose dense (doxorubicin and cyclophosphamide q 2 weeks)<br />

( ISRCTN61893718).<br />

In total 180 interesting SNPs were selected that are involved in<br />

metabolism, distribution and excretion of drugs and tested on DNA<br />

that was isolated from tumor or from tumor surrounding tissue. We<br />

selected 20 SNPs for further analyses because of reported associations<br />

with chemotherapy-induced neuropathy (see table).<br />

In the first analysis DNA of 45 patients was analyzed as a pilot,<br />

comparing DNA of 23 neuropathy patients with that of 22 controls.<br />

The primary hypothesis was to identify SNPs associated with NCI-<br />

CTC grade ≥2 neuropathy.<br />

Results:<br />

Interim toxicity data from the MATADOR study demonstrated that<br />

30 of 580 patients had grade ≥2 neuropathy. GSTP1 114Ala/114Val<br />

genotype was associated with grade ≥2 neuropathy. Twelve out of<br />

14 (86%) patients with this genotype had chemotherapy-induced<br />

neuropathy compared to 11 out of 31 (35%) without this genotype<br />

(chi-square test p=0.002).<br />

Conclusion:<br />

We found a correlation between GSTP1 114Ala/114Val genotype,<br />

involved in detoxification, and development of grade ≥2 neuropathy<br />

during treatment with chemotherapy. Updated results will be<br />

presented at the meeting.<br />

SNPs analyzed<br />

1. ABCB1 rs1045642<br />

2. ABCB1 rs1128503<br />

3. ABCB1 rs2032582<br />

4. ABCB1 rs3213619<br />

5. CYP2C8 rs1058930<br />

6. CYP2C8 rs10509681<br />

7. CYP2C8 rs11572080<br />

8. CYP2C8 rs11572103<br />

9. CYP2C8 rs72558195<br />

10. CYP2C8 rs72558196<br />

11. CYP3A4 rs2740574<br />

12. CYP3A5 rs776746<br />

13. CYP3A5 rs10264272<br />

14. CYP3A5 rs41303343<br />

15. CYP3A5 rs55965422<br />

16. GSTM1 rs1065411<br />

17. GSTP1 rs1695<br />

18. GSTP1 rs1138272<br />

19. RWDD3 rs2296308<br />

20. TECTA rs1829<br />

P1-15-06<br />

The impact of musculoskeletal toxicity on adherence to endocrine<br />

therapy in women with early stage breast cancer– observations<br />

in a non-trial setting<br />

Dent SF, Campbell MM, Crawley FL, Clemons MJ. The Ottawa<br />

Hospital Regional <strong>Cancer</strong> Center, Ottawa, ON, Canada<br />

Background: Aromatase inhibitor (AI) use is standard of care in the<br />

treatment of postmenopausal (PM) women with early stage breast<br />

cancer (EBC). Approximately 35% of patients (pts) discontinue their<br />

initially prescribed AI due to toxicity, the most commonly reported<br />

reason being musculoskeletal toxicity (MSKT). We report on the<br />

discontinuation rates of AI therapy based on MSKT in PM women<br />

with EBC treated at a tertiary care cancer centre.<br />

Methods: PM women with hormone receptor positive EBC treated<br />

with endocrine therapy (ET) that included an AI (upfront or after<br />

tamoxifen) at The Ottawa Hospital <strong>Cancer</strong> Center between 01/99<br />

and 02/06. Data included: demographics, type of ET, duration of<br />

treatment, incidence of patient-reported MSKT and treatment of<br />

MSKT. Comparisons between ETs were analyzed using Chi-square<br />

and Fischer’s t-tests.<br />

Results: A total of 626 pts, median 59 years (r: 30-92), median followup<br />

98 months, with stage: I (196 pts; 31%), II (341 pts; 54%) or III (89<br />

pts; 14%s) EBC. Treatment strategies included: AI(s) only (251 pts;<br />

40%); tamoxifen (TAM) followed by AI(s) (323 pts; 51.6%); AI(s)<br />

followed by TAM (16 pts; 2.6%); TAM-AI(s)-TAM (24 pts; 3.8%)<br />

and unknown (12 pts). Patient-reported MSKT was experienced by<br />

significantly more women treated with AIs than TAM (64% vs 36%,<br />

p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P1-15-07<br />

Ixabepilone-associated peripheral neuropathy in metastatic<br />

breast cancer patients and its effects on the ultrastructure of<br />

neurons<br />

Jain S, Carlson K, Chuang E, Cigler T, Moore A, Donovan D, Lam C,<br />

Cobham MV, Schneider S, Ramnarain A, Carey B, Ward M, Lane M,<br />

Strickland S, Vahdat L. Weill Cornell Medical College; Rockefeller<br />

University<br />

Background: Peripheral neuropathy is a dose-limiting toxicity of<br />

most microtubule-stabilizing chemotherapeutic agents. Ixabepilone,<br />

a semisynthetic analog of the natural epothilone B, has activity in<br />

a wide range of tumors including taxane-resistant disease. In this<br />

study, we sought to understand the effect of ixabepilone on the<br />

development of peripheral neuropathy both clinically and its effect<br />

at the ultrastructural level of the peripheral nerves and circulating<br />

factors over time. Parallel studies in animal models of neuropathy<br />

were performed at the same time (Proc AACR 2010 <strong>Abstract</strong> 4184).<br />

Methods: This open-label, non-randomized phase II study enrolled 14<br />

patients with metastatic breast cancer. Ixabepilone was administered<br />

by 2 schedules: the FDA approved dose of 40 mg/m² every 3 weeks<br />

(q3w) and 16 mg/m² on day 1, 8, and 15 of a 28-day cycle (weekly).<br />

Five controls, 2 with residual taxane-associated peripheral neuropathy<br />

and 3 with no prior chemotherapy or peripheral neuropathy, were<br />

also accrued. The primary objectives were to characterize the<br />

natural history of ixabepilone-associated peripheral neuropathy<br />

using the Total Neuropathy Score Clinical (TNSc) assessment tool<br />

prior to each cycle and to correlate changes in the ultrastructure<br />

of dermal myelinated nerve fibers via a 3 mm punch biopsy of an<br />

area 10 cm above the lateral malleolus every 2 cycles with electron<br />

microscopy (EM), as well as circulating factors (both inflammatory<br />

and neurotrophic) considered to be important in the pathogenesis of<br />

chemotherapy-induced peripheral neuropathy. Secondary objectives<br />

included progression-free survival (PFS) and non-neurologic toxicity.<br />

Results: 14 patients were enrolled and were equally divided<br />

between the 2 schedules of ixabepilone chemotherapy. There were<br />

no differences in baseline characteristics between the two groups.<br />

Mean age was 54 years (range 32-71). Mean number of previous<br />

chemotherapy regimens was 3.5 (range 0-8). 57% of patients had<br />

received a taxane in the adjuvant setting and 64% in the metastatic<br />

setting. The mean neuropathy score (TNSc) at baseline was 4.6<br />

(range 1-11). At a mean cumulative dose of 185 mg/m², the TNSc<br />

with ixabepilone q3w schedule was 3.7 points higher/worse (95%<br />

CI: 2.2-5.3, p=0.03) than the mean score observed in patients on the<br />

weekly schedule. The sensory component was most significantly<br />

affected, predominantly numbness. In 3 patients, the chemotherapy<br />

schedule was changed from every 3 weeks to weekly due to > grade<br />

2 toxicity at a mean cumulative dose of 107 mg/m², and TNSc<br />

decreased/improved by 2.7 points. PFS in patients on q3w ixabepilone<br />

was 133 days (range 28-280) and in patients on weekly ixabepilone<br />

was 179 days (range 66-336), nonsignificant. Evaluation of EM and<br />

circulating factors is ongoing.<br />

Conclusions: Weekly ixabepilone appears to have a more favorable<br />

neurotoxicity profile compared to the standard q3w schedule.<br />

Integration of the EM data and the circulating factor data are underway<br />

and will be presented. Ixabepilone-associated peripheral neuropathy<br />

may improve in patients switched to weekly ixabepilone without<br />

compromising efficacy.<br />

P1-15-08<br />

Higher toxicity of docetaxel for obese women with early breast<br />

cancer: lean body mass is a significant predictor of chemotherapy<br />

dose intensity reduction<br />

Gouerant S, Clatot F, Modzlewski R, Chaker M, Rigal O, Veyret<br />

C, Leheurteur M. Henri Becquerel Center, Rouen, France; Rouen<br />

University Hospital, Rouen, France<br />

Background: Prevalence of obesity has been increasing in the last<br />

years and obesity is known as a risk factor of post menopausal<br />

breast cancer. Despite the high number of obese women treated by<br />

docetaxel for breast cancer, few data are published about toxicity and<br />

its predictive factors in this population. A low lean body mass (LBM)<br />

was shown as a predictive factor of toxicity for other chemotherapy<br />

and can be measured by software on an abdominal computerized<br />

tomography (CT), we aimed to assess the predictive value of LBM<br />

in docetaxel toxicity.<br />

Methods: This is a monocentric retrospective analysis of patients<br />

treated for early breast cancer from January 2008 to December<br />

2010, who received a sequential chemotherapy with FEC 100 and<br />

docetaxel. All obese patients (BMI≥30 mg/m2) treated over these<br />

3 years were included (n=100), and the same number of non-obese<br />

patients were randomly selected among all those treated during this<br />

same period. We collected data from objective toxicity (hematologic<br />

and cardiologic toxicity, hospitalizations), and data related to dose<br />

intensity (dose diminution, delayed cure, modification drug, cessation<br />

of chemotherapy) to calculate the ratio [Total Received Dose (mg)/<br />

Total Planned Dose (mg)] (TRD/TPD ratio) for each drug. We<br />

measured the LBM for patients who had abdominal CT at diagnosis<br />

(n=89) to calculate the ratio [Planned Dose per cycle (mg) / LBM<br />

(kg)] (PD/LBM ratio).<br />

Results: Among 469 patients who received a sequential chemotherapy<br />

FEC 100/Docetaxel for an early breast cancer in these three years,<br />

100 were obese (21%). Obese and non-obese patients have similar<br />

clinical and histological characteristics except for T4d stage (10 in<br />

obese group and 1 in non-obese group, p=0,02).<br />

The TRD/TPD ratio of docetaxel is significantly lower in obese<br />

patients: 18 obese and 6 non-obese patients had a ratio less than<br />

1 (p=0,02), the main cause was cutaneous toxicities. On the other<br />

hand, there is no significant difference for the TRD/TPD ratio of<br />

epirubicine: 3 obese and 1 non-obese patients had a ratio less than 1<br />

(p=0,62). In univariate analysis, 4 factors are predictive of a TRD/<br />

TPD ratio of docetaxel less than 1: high age (p=0,01), high weight<br />

(p=0,02), high body surface area (p=0,05) and high BMI (p=0,006).<br />

In multivariate analysis, only BMI is predictive of a TRD/TPD ratio<br />

of docetaxel less than 1<br />

Patients with a TRD/TPD ratio of docetaxel less than 1 had a<br />

significantly higher PD/LBM compared to patients with a TRD/TPD<br />

ratio greater than 1, respectively 4,75 [3,02-6,04] and 3,75 [2,23-<br />

5,79] (p=0,01). In univariate analysis, the other predictive factors of<br />

TRD/TPD ratio of docetaxel less than 1 are high BMI (p=0,02), high<br />

weight (p=0,02) and high BSA (p=0,03). In multivariate analysis, only<br />

PD/LBM is predictive of a TRD/TPD ratio of docetaxel less than 1.<br />

Conclusion: Docetaxel is less well tolerated and the dose intensity<br />

received is lower for obese patients. The lean body mass measured<br />

by computed tomography is a new significative predictor of docetaxel<br />

dose intensity reduction in the early breast cancer.<br />

Key words: Early breast cancer-Chemotherapy-Docetaxel-<br />

Epirubicine-Body Mass Index-Lean Body Mass-Dose Intensity-<br />

Toxicity<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

212s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 1<br />

P1-15-09<br />

Multi-Institutional Evaluation of Bioimpedance Spectroscopy<br />

(BIS) in the Early Detection of <strong>Breast</strong> <strong>Cancer</strong> Related<br />

Lymphedema<br />

Vicini F, Arthur D, Shah C, Anglin BV, Curcio L, Laidley AL, Beitsch<br />

P, Whitworth P, Lyden M. Michigan Healthcare Professionals/21st<br />

Century Oncology; Virginia Commonwealth University; Beaumont<br />

Health System; The Medical Center of Plano; Advanced <strong>Breast</strong><br />

Care Specialists of Orange County; Texas <strong>Breast</strong> Specialists; Dallas<br />

Surgical Group; Nashville <strong>Breast</strong> Center; Biostat Inc.<br />

Background: Limited data is available examining the impact of early<br />

detection in the management of breast cancer related lymphedema<br />

(BCRL) with bioelectrical impedance spectroscopy (BIS) offering the<br />

potential for subclinical diagnosis of BCRL. The purpose of the study<br />

was to evaluate BIS’s ability to detect and monitor extracellular fluid<br />

accumulation of the upper limb in patients treated for breast cancer<br />

as it relates to the extent of loco-regional therapy.<br />

Methods: A total of 125 patients with breast cancer from 4 clinical<br />

practices were evaluated with BIS at baseline and following locoregional<br />

procedures. In order to assess the ability of BIS to detect<br />

sub-clinical changes by treatment modality, the change in L-Dex<br />

score from baseline to measurements taken within 180 days following<br />

surgery were calculated.<br />

Results: Mean age was 55 years with 68 patients (54.4%) undergoing<br />

sentinel lymph node (SLN) sampling while 57 (45.6%) underwent<br />

an axillary dissection (ALND). The mean number of nodes sampled<br />

was 7.9 (range:1.0-37.0) with 40.8%, 18.4%, 13.6%, and 27.2%<br />

of patients having 0-3, 4-6, 7-9, or greater than 10 lymph nodes<br />

sampled. Fifty-one patients (40.8%) underwent lumpectomy and 74<br />

(59.2%) mastectomy with 37 patients (29.6%) requiring more than<br />

one surgical procedure. Sixty-five patients (52%) underwent radiation<br />

therapy (RT). While controlling for baseline L-Dex score and the<br />

number of nodes removed, patients receiving RT had a significantly<br />

increased change (pre v. post radiation treatment) in mean L-Dex<br />

score (-2.5 v. 0.8, p=0.03) compared with those patients not receiving<br />

RT. When evaluating changes in mean L-Dex score from baseline<br />

BIS assessment to the first assessment within 180 days of surgery,<br />

no difference in the change in score was noted by surgical procedure<br />

(lumpectomy 1.8 v. mastectomy 2.2, p = 0.97); however, ALND was<br />

associated with a significantly increased change in mean L-Dex score<br />

(0.3 v. 5.0, p=0.003) compared with SLN biopsy. When stratifying<br />

by the number of nodes removed, a statistically significant increase<br />

in the change in mean L-Dex score was noted (0.4 v. 0.4 v. 4.3 v.<br />

6.4, p=0.04) for 0-3, 4-6, 7-10, and greater than 10 lymph nodes<br />

removed. Similar findings were noted when evaluating the impact<br />

of ALND (0.3 v. 4.0, p=0.0002) and nodes removed (0.5 v. 0.3 v. 2.9<br />

v. 4.9, p=0.006) when comparing the baseline BIS assessment to the<br />

first post-surgical assessment regardless of when the assessment was<br />

performed post-operatively.<br />

Conclusions: In this multi-institutional analysis, L-Dex scores<br />

generated from multi-frequency bioimpedance spectroscopy (BIS)<br />

mirrored the aggressiveness of loco-regional therapy with respect<br />

to nodal sampling and radiation therapy. These findings suggest<br />

that BIS can be used in the early detection of BCRL and should be<br />

implemented before and following loco-regional therapies. Further<br />

studies are needed to help validate the extent, degree, and chronologic<br />

time frame of these changes to help define recommendations for closer<br />

monitoring and possible early intervention and to compare them to<br />

concurrent clinical assessments.<br />

P1-15-10<br />

Chemotherapy-Induced Neutropenia in <strong>Breast</strong> <strong>Cancer</strong> Patients<br />

Receiving Sequential Anthracycline and Taxane Chemotherapy:<br />

An institutional experience<br />

Staudigl C, Seifert M, Tea M-K, Pfeiler G, Fink-Retter A, Fritzer<br />

N, Singer CF. Medical University of Vienna, Austria; Alps-Adria<br />

University Klagenfurt, Klagenfurt, Austria<br />

Background: Sequential regimen of anthracyclines and taxanes<br />

are commonly used in patients with early stage breast cancer (BC)<br />

and curative treatment intent. Due to the associated myelotoxicity,<br />

internationally accepted guidelines recommend granulocyte-colony<br />

stimulating factors (G-CSF) as primary or secondary prophylaxis,<br />

dependent on chemotherapy (CT) and patient-associated risk of febrile<br />

neutropenia. As part of a quality control initiative at our center, we<br />

have analyzed rates of grade 1 or 2 (1.0 x 109/L) and 3 or 4<br />

(


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P1-15-11<br />

The Kampo medicine Goshajinkigan prevents docetaxel-related<br />

peripheral neuropathy in breast cancer patients<br />

Abe H, Mori T, Kawai Y, Itoi N, Tomida K, Cho H, Kubota Y, Umeda<br />

T, Tani T. Shiga University of Medical Science Hospital, Otsu, Shiga,<br />

Japan; Shiga University of Medical Science, Otsu, Shiga, Japan<br />

Background: Although taxanes have become a key chemotherapeutic<br />

drug in breast cancer treatment. Taxanes inhibit the growth of cancer<br />

cells by disrupting the functioning of their microtubules; however,<br />

the microtubules of nerve cells are also affected by this process,<br />

which can cause neurological disorders. The Kampo medicine<br />

Goshajinkigan (GJG) is a traditional Japanese medicine that is used<br />

for the treatment of several neurological symptoms including pain<br />

and numbness, GJG is comprised of 10 herbs, each of which contains<br />

numerous active ingredients. Recently, GJG has been reported to<br />

prevent anticancer drug-induced peripheral neuropathy in colorectal<br />

cancer. We performed the present prospective randomized study to<br />

confirm the effects of GJG and mecobalamin (B12) against docetaxel<br />

(DOC)-associated peripheral neurotoxicity in breast cancer patients.<br />

Patients and method: Between May 2007 and April 2011, 60<br />

breast cancer patients were treated with DOC. Thirty-three patients<br />

(GJG group) received oral administration of 7.5 g/day GJG and 27<br />

patients (B12 group) received oral administration of 1500 µg/day<br />

B12. The patients were treated with TC (75mg/m 2 docetaxel and<br />

600 mg/m 2 cyclophosphamide) every 3 weeks for 4 cycles, docetaxel<br />

alone (100mg/m 2 ) every 3 weeks for 4 cycles, and XT (900mg/m 2<br />

capecitabine administered orally twice a day on days 1-14 plus 60 mg/<br />

m 2 docetaxel) every 3 weeks for 6 cycles. Peripheral neuropathy was<br />

evaluated during every course according to DEB-NTC (Neurotoxicity<br />

Criteria of Debiopharm), Common Terminology Criteria for Adverse<br />

Events (NCI-CTC) ver.3.0, and a visual analogue scale (VAS).<br />

Results: The median age of the GJG group was 58 years old (35 to<br />

70 years old), the B12 group was 55 years old (33 to 69 years old),<br />

and they were all females. For the regimens, in the GJG group, TC,<br />

DOC only, and XT were administered in 19 cases, 13 cases and 1<br />

case, respectively. In the B12 group, they were 15 cases, 11 cases and<br />

12 cases, respectively. The cumulative dose of DOC was 338.5 mg/<br />

m2 in the GJG group, and 340 mg/m2 in the B12 group. Peripheral<br />

neuropathy occurred significantly less frequently in the GJG group<br />

(39.3%) than the B12 group (88.9%) (p


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 2<br />

cells (CTCs) isolated by CellSearch® from peripheral blood of MBC<br />

patients was shown to be an additional strong independent prognostic<br />

factor for survival and progression of disease. The aim of this study<br />

was to combine CTC and other predictors in a nomogram that would<br />

allow clinicians to assess, on an individual basis, the prognosis of<br />

patients starting first-line chemotherapy for MBC.<br />

Patients and Methods: CTCs were counted before the start of treatment<br />

using the US Food and Drug Administration–cleared CellSearch®<br />

technology. We used a training set of 237 MBC patients starting<br />

a first line chemotherapy from the MD Anderson <strong>Cancer</strong> Center<br />

(MDACC, Houston, TX, USA), and nomograms were established<br />

to calculate the predicted probability of overall survivals (OS) at 1,<br />

2 and 3 years. These nomograms were externally validated using an<br />

independent data set of 260 patients starting a first line chemotherapy<br />

from the prospective Institut Curie 2006-04 study (Paris, France).<br />

Discriminatory ability of the models was assessed using Harrell’s<br />

c-statistic examining the model in the training sample (U.S. data) and<br />

the validation sample (French data). Accuracy of the nomograms was<br />

assessed both within the training sample and the validation sample<br />

via calibration plots at 1, 2 and 3 years. Covariates computed in the<br />

model were: age, subtype of disease, presence of visceral metastasis,<br />

bone metastasis, performance status, and CTCs.<br />

Results: Table1 describes patient characteristics. Median follow-up<br />

was of 24.5 and 29 months, respectively in the MDACC and Institut<br />

Curie. The C-statistics for OS was 0.7338 in the training set and<br />

0.7229 in the validation set. For the calibration plots, we averaged Cox<br />

predictions at two years within the quintiles of the ordered predictions.<br />

Within each quintile, we also estimated the unadjusted probability<br />

of death using Kaplan-Meier survival estimators. We then plotted<br />

unadjusted versus model average predictions at 1, 2 and 3 years. The<br />

nomograms discriminated OS prediction at 1, 2 and 3 years very well.<br />

Conclusions: Patients with first-line MBC present a wide range of<br />

survival duration, and our ability to assess prognosis and predict risks<br />

of progression and death on the basis of clinic-pathologic findings<br />

is limited. The validated nomograms represent an important tool for<br />

estimating prognosis, support treatment-decisions and clinical trial<br />

stratification in MBC.<br />

Patient characteristics<br />

MDACC cohort ICurie cohort<br />

Characteristics No. % No. % P value<br />

Total patients 236 100 210 100<br />

Age at diagnosis, years<br />

median 49.6 54.5 0.0004<br />

mean 50.3 58.4<br />

Receptor status<br />

ER+ or PR+, and HER2- 130 55.1 125 59.5 0.629<br />

HER2+ 43 18.2 36 17.1<br />

ER-, PR-, and HER2- 63 26.7 49 23.3<br />

Presence of visceral metastasis 144 61 169 80.5 40 35 14.8 28 13.3<br />

Missing data not included.<br />

P2-01-02<br />

Circulating Tumor Cells (CTC) may Express HER2/neu in<br />

Patients With Early HER2/neu Negative <strong>Breast</strong> <strong>Cancer</strong> – Results<br />

of the German SUCCESS C Trial<br />

Jaeger BAS, Rack BK, Andergassen U, Neugebauer JK, Melcher<br />

CA, Scholz C, Hagenbeck C, Schueller K, Lorenz R, Decker T,<br />

Heinrich G, Fehm T, Schneeweiss A, Lichtenegger W, Beckmann<br />

MW, Pantel K, Sommer HL, Friese K, Janni W. Klinikum der Ludwig-<br />

Maximilians-Universitaet, Munich, Germany; Heinrich Heine<br />

University, Duesseldorf, Germany; Stat-up Statistische Beratung<br />

und Dienstleistung, Munich, Germany; Gemeinschaftspraxis Dr.<br />

Lorenz/Hecker/Wesche, Braunschweig, Germany; Studienzentrum<br />

Onkologie Ravensburg, Ravensburg, Germany; Praxis Dr. Heinrich,<br />

Fuerstenwalde, Germany; Eberhard Karls Universitaet Tuebingen,<br />

Tuebingen, Germany; National Center for Tumor Disease and<br />

Department of Gynecology and Obstetrics, University Hospital<br />

Heidelberg, Germany; Charité Medical University, Berlin, Germany;<br />

Universitaet Erlangen, Germany; University Medical Center<br />

Hamburg-Eppendorf, Hamburg, Germany<br />

Background:<br />

There is growing evidence that the HER2/neu-status of distant<br />

metastases or minimal residual disease in blood and bone marrow<br />

may differ from the primary tumor in patients with breast cancer. The<br />

HER2/neu-status of CTCs was prospectively evaluated in patients<br />

with HER2/neu negative primary breast cancer randomized into the<br />

German multicenter SUCCESS C study.<br />

Methods:<br />

The SUCCESS C trial is a randomized Phase III study comparing<br />

FEC-Docetaxel (FEC-Doc) vs. Docetaxel-Cyclophosphamid (DC)<br />

as well as 2 years of a lifestyle-intervention in patients with early,<br />

HER2/neu negative, node positive or high-risk node negative primary<br />

breast cancer.<br />

As part of the translational research program, 23ml peripheral<br />

blood were drawn after adjuvant chemotherapy. In 505 samples, the<br />

prevalence of CTCs and their HER2/neu-status were assessed using<br />

the CellSearch System (Veridex, USA). After immunomagnetic<br />

enrichment with an anti-Epcam-antibody, cells were labelled with<br />

anti-CK8/18/19 and anti-CD45 antibodies. A fluorescein conjugate<br />

antibody with anti-CK-Fluorescein Isothiocyanate (FITC) was used<br />

for HER2/neu phenotyping. The cut-off for CTC-positivity was ≥ 1<br />

CTC and for HER2/neu ≥ 1 CTC with strong HER2/neu-staining<br />

(+++).<br />

Results:<br />

26,9% of pts (n=136) were positive for CTCs (mean 1.78; range 1-7;<br />

median = 1). The number of detected CTC was distributed as follows:<br />

1 CTC (n=76; 55.9%), 2 CTCs (n=35; 25.7%), 3 CTCs (n=13; 9.6%),<br />

4 CTCs (n=7; 5.2%) and ≥ 5 CTCs (n=5; 3.7%). HER2/neu staning<br />

of CTCs was not detectable or weak in 26.5% (n=36) and 4.4% (n=6)<br />

of CTC positive patients respectively and therefore categorized as<br />

HER2/neu negative. In 32.4% of the CTC-positive patients (n=44),<br />

we detected moderate and in 36.8% (n=50) strong HER2/neu-staining<br />

of ≥ 1 CTC per sample. No association was found between CTCs<br />

or the HER2/neu-status of CTCs with tumor size, histopathological<br />

grading, hormone receptor status or axillary lymph node involvement.<br />

Conclusions:<br />

The data of this trial confirm previous findings that patients with<br />

HER2/neu negative primary breast cancer can show HER2/neu<br />

positive minimal residual disease. These results underline the<br />

importance of frequent HER2/neu determination during follow up and<br />

disease progression. Survival data within the Success C trial will give<br />

further insight into the tumor biology of HER2/neu negative disease.<br />

www.aacrjournals.org 215s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P2-01-03<br />

Truncated HER2 receptor in circulating tumor cells (CTCs) of<br />

early and metastatic breast cancer patients<br />

Kallergi G, Nasias D, Papadaki M, Mavroudis D, Georgoulias V,<br />

Agelaki S. University of Crete, Heraklion, Crete, Greece; University<br />

General Hospital of Heraklion, Crete, Greece<br />

Introduction: Trastuzumab confers significant clinical benefit in<br />

patients with early and metastatic HER2-positive breast cancer.<br />

However, trastuzumab treated patients will eventually develop<br />

resistance to therapy. The presence of a truncated receptor with loss<br />

of the extracellular portion of HER2 (p95HER2) has been proposed<br />

as a potential mechanism of resistance. Preclinical and clinical<br />

data suggest that the expression of p95HER2 is associated with<br />

poor overall prognosis, resistance to trastuzumab and sensitivity to<br />

lapatinib, a dual EGFR and HER2 tyrosine kinase inhibitor. We have<br />

previously shown that approximately 50% of breast cancer patients<br />

express HER2 in their CTCs (Kallergi et al., 2007; Kallergi et al.,<br />

2008). The aim of the present study was to characterize, for the first<br />

time, the expression of p95HER2 in CTCs of breast cancer patients<br />

in comparison with the p95HER2 expression of the corresponding<br />

primary tumors. Methods: Two different cells lines (SKBR3 and<br />

SCOV3) were used as positive controls expressing the normal<br />

and truncated HER2 receptor, respectively. Western blot analysis<br />

revealed that SCOV3 was positive for p95 fragment of HER2<br />

receptor while SKBR3 had the full length protein. Consequently<br />

we developed a novel triple immunofluorescent (IF) assay for the<br />

simultaneous evaluation of the extracellular and intracellular domain<br />

of HER2 receptor using the corresponding anti-rabbit and anti-mouse<br />

antibodies respectively. Cells were also stained with pancytokeratin<br />

A45-B/B3 antibody (as a marker of CK-positive cells). Triple staining<br />

experiments were performed in peripheral blood mononuclear cells<br />

(PBMC) cytospins’ from patients with early (n=24) and metastatic<br />

(n=33) breast cancer. Slides were analysed with either confocal laser<br />

scanning microscopy or with the Ariol system. Results: Cytokeratin<br />

positive cells were identified in 17 (71%) out of 24 early and in<br />

20 (61%) out of 33 metastatic patients. HER2 positive CTCs were<br />

identified in 53% of early and 50% of metastatic, CTC-positive, breast<br />

cancer patients. HER2-positive CTCs lacking the extracellular domain<br />

of the receptor (p95HER positive CTCs) were identified in 7 out of<br />

20 (35%) of metastatic and 2 out of 17 (12%) of early, breast cancer<br />

patients. Exclusively p95HER2 positive CTCs were detected in 2 out<br />

of 20 (10%) patients with metastatic but not in patients with early<br />

disease. The corresponding primary tumors from 8 HER2-positive<br />

breast cancer patients were also triple stained. These experiments<br />

revealed that in two out of eight patients, p95HER2-positive cells<br />

were detected in a subgroup (5-10%) of cancer cells. Conclusions: Our<br />

results suggest that the truncated p95HER2 receptor is expressed in<br />

CTCs of both early and metastatic breast cancer patients. This finding<br />

has important therapeutic implications and may explain the observed<br />

increased efficacy of the trastuzumab plus lapatinib combination in<br />

HER2 positive breast cancer.<br />

P2-01-04<br />

Long term independent prognostic impact of circulating tumor<br />

cells detected before neoadjuvant chemotherapy in non-metastatic<br />

breast cancer: 70 months analysis of the REMAGUS02 study<br />

Bidard F-C, Delaloge S, Giacchetti S, Brain E, de Cremoux P, Vincent-<br />

Salomon A, Marty M, Pierga J-Y. Institut Curie, France; Institut<br />

Gustave Roussy, France; Hopital Saint Louis, France<br />

Background: Circulating tumor cells (CTCs) isolated by CellSearch ®<br />

from peripheral blood of metastatic breast patients have been shown<br />

as strong independent prognostic factor for progression-free and<br />

overall survival. With this technique, the REMAGUS02 prospective<br />

multicentric study was the first to report that CTC detection<br />

(≥1CTC/7.5ml) before and/or after neoadjuvant chemotherapy<br />

was independently associated with distant metastasis-free survival<br />

(DMFS, 18 months follow-up, Pierga CCR 2008) and with overall<br />

survival (OS, 36 months follow-up, Bidard Ann Oncol 2010).<br />

Patients and Methods: In 115 non-metastatic patients diagnosed with<br />

large operable or locally advanced breast cancer, we prospectively<br />

detected CTC using the CellSearch system before and after<br />

neoadjuvant chemotherapy in the REMAGUS02 trial (Pierga, BCRT<br />

2010). For this report, survival analyses were performed at a median<br />

follow-up of 70 months.<br />

Results: After a median follow-up of 70 months, 20 distant metastatic<br />

relapses and 14 deaths have been observed among the 115 patients<br />

included. CTC detection before chemotherapy (in 23% of patients) is<br />

an independent prognostic factor for both DMFS [P=0.03, relative risk<br />

(RR)=3.2] and OS [p=0.05, RR=3.7] in multivariate analyses, together<br />

with triple negative tumor status. At long-term, CTC detection after<br />

chemotherapy (17%) had no clear prognostic significance (p=0.15<br />

and 0.22, respectively). Complete pathological response as well as<br />

tumour size and all other variables tested did not appear as predictive<br />

in this model.<br />

Conclusions: Baseline CTC detection is a new independent<br />

prognostic factor in the neoadjuvant setting, and is, in this trial,<br />

superior to pathological tumor response to predict the survival of nonmetastatic<br />

breast cancer patients. We confirm therefore our previous<br />

results reported with shorter follow-up.<br />

Supported by PHRC AOM/2OO2/02117, Pfizer inc., Roche, sanofiaventis.ISRCTN10059974<br />

P2-01-05<br />

Parallel DNA and RNA profiling of EpCAM-positive cells in<br />

blood of metastatic breast cancer (MBC) patients confirm their<br />

malignant nature<br />

Magbanua MJM, Hauranieh L, Sosa EV, Pendyala P, Scott JH, Rugo<br />

HS, Park JW. University of California, <strong>San</strong> Francisco, CA<br />

Background: Molecular studies of EpCAM-positive cells found in<br />

the blood of cancer patients have been limited to immuno-enriched<br />

samples containing a high background of leukocytes. We developed<br />

a process to isolate rare EpCAM-positive cells and subjected them<br />

to DNA and RNA profiling.<br />

Methods: Blood was subjected to immunomagnetic enrichment<br />

using EpCAM beads followed by fluorescence activated cell sorting<br />

(IE/FACS) to isolate EpCAM-positive cells away from leukocytes<br />

(CD45+). Duplicate samples of 20 cells were isolated from the same<br />

enriched blood from 53 MBC patients and then subjected to DNA<br />

and RNA profiling in parallel. For DNA profiling, sorted cells were<br />

subjected to BAC array comparative genomic hybridization analysis<br />

following whole genome amplification. For RNA profiling, QPCR<br />

analysis was performed on sixty four (64) cancer-related genes using<br />

Taqman® low density arrays. For non-tumor controls, RNA profiling<br />

was performed on matched leukocytes (CD45+) isolated from the<br />

same enriched blood samples from 44 of the 53 patients.<br />

Results: Differential gene expression analysis between EpCAMpositive<br />

cells and matched leukocytes confirmed the up-regulation<br />

of EPCAM and other genes including MUC1 and KRT19 (adjusted p<br />


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 2<br />

hierarchical clustering analysis of RNA profiles showed that EpCAMpositive<br />

cells clustered away from the leukocytes. Genome-wide copy<br />

number analysis of EpCAM-expressing cells revealed gains (e.g. 1q<br />

and 8q), losses (e.g. 8p and 16q), and focal amplifications (e.g. on 8q<br />

and 11q including CCND1) frequently seen in primary breast cancers.<br />

Discussion: We demonstrate the feasibility of isolation and molecular<br />

analysis of highly pure EpCAM-positive cells in the blood. Our results<br />

strongly suggest that epithelial cells captured from blood of MBC<br />

patients are malignant in nature and are therefore circulating tumor<br />

cells. This approach can serve as a non-invasive source of highly pure<br />

metastatic tumor tissue for further molecular characterization. This<br />

study was supported by CALGB and BCRF.<br />

P2-01-06<br />

Association between Circulating Tumor Cells and Bone Turnover<br />

Markers in patients with breast cancer and bone metastases on<br />

treatment with bisphosphonates (ZOMAR study)<br />

Manso L, Barnadas A, Tusquets I, Crespo C, Gómez P, Calvo L, Ruiz<br />

M, Martínez P, Perelló A, Antón A, Codes M, Margelí M, Murias<br />

A, Salvador J, Seguí MA, De Juán A, Gavilá J, Luque M, Pérez D,<br />

Zamora P, Arizcum A, Chacón JI, Heras L, De la Piedra C. Hospital<br />

12 de Octubre, Madrid, Spain; Hospital <strong>San</strong>ta Creu i <strong>San</strong>t Pau,<br />

Barcelona, Spain; Hospital del Mar, Barcelona, Spain; Hospital<br />

Universitario Ramón y Cajal, Madrid, Spain; Hospital Universitario<br />

Vall d’Hebrón, Barcelona, Spain; Hospital Universitario A Coruña<br />

Juan Canalejo, A Coruña, Spain; Hospital Universitario Virgen del<br />

Rocío, Sevilla, Spain; Hospital Basurto, Vizcaya, Spain; Hospital<br />

Son Dureta, Palma de Mallorca, Spain; Hospital Miguel Servet,<br />

Zaragoza, Spain; Hospital Virgen Macarena, Sevilla, Spain; Hospital<br />

Universitario Trias y Pujol, Barcelona, Spain; Hospital Universitario<br />

Insular de Gran Canaria, Gran Canaria, Spain; Hospital Nuestra<br />

Señora de Valme, Sevilla, Spain; Hospital Parc Taulí Sabadell,<br />

Barcelona, Spain; Hospital Marqués Valdecilla, <strong>San</strong>tander, Spain;<br />

Instituto Valenciano de Oncología, Valencia, Spain; Hospital General<br />

de Asturias, Oviedo, Spain; Hospital Costa del Sol, Málaga, Spain;<br />

Hospital La Paz, Madrid, Spain; Hospital Palencia Río Carrión,<br />

Palencia, Spain; Hospital Virgen de la Salud, Toledo, Spain; Hospital<br />

Cruz Roja Hospitalet del Llobregat, Barcelona, Spain; Instituto de<br />

Investigación <strong>San</strong>itaria Fundación Jiménez Díaz, Madrid, Spain<br />

Background:<br />

Quantification of Circulating Tumor Cells (CTC) has demonstrated<br />

an important role in assessing disease progression and outcomes,<br />

and pathological CTC levels are an independent prognostic factor of<br />

disease progression. High CTC levels may be associated with bone<br />

turnover markers (BTM) levels and have also been associated with<br />

the risk of skeletal-related events (SREs), disease progression and<br />

death. The aim of this study was to determine the relation between<br />

CTC, BTM, and SREs, disease progression and death in patients in<br />

patients with breast cancer (BC) and bone metastasis (BMe) treated<br />

with zoledronic Acid.<br />

Patients and methods:<br />

Observational, prospective and multicenter study. Patients with<br />

BC and BMe; no previous bone treatment in the last 6 months<br />

prior to study entry. CTC (fluorescently labelled with nucleic acid<br />

dye 4,6-diamidino-2-phenylindole DAPI, monoclonal antibodies<br />

specific for leukocytes CD45-allophycocyanin and epithelial cells<br />

cytokeratin 8,18,19–phycoerythrin; Cell Search System Veridex);<br />

urinary aminoterminal telopeptide of collagen I (NTX, Osteomark<br />

NTx Urine, Wampole Laboratories, USA); urinary alpha-alpha-isomer<br />

of carboxyterminal telopeptide of collagen I (αα-CTX, ALPHA<br />

Crosslaps EIA, ids, UK) and serum bone alkaline phosphatase (BALP,<br />

OSTASE BAP, ids, UK) were determined at baseline (V0) and every<br />

3 mo along 18 months (V6). Patients were treated with zoledronic<br />

acid (ZA) at inclusion and every 3-4 weeks.<br />

Results:<br />

234 patients with BC and BMe were analyzed, being available CTC<br />

results for 114 patients and BTM results for 219 patients at basal visit<br />

(V0) and every 3 mo of treatment along 18 months (V6). Population<br />

basal characteristics (234 patients): mean age: 59.8 years; ER+:<br />

80.3%; PR+: 64.9%; HER 2+: 18.3%. 55.3% of patients (n=114)<br />

had detectable CTC (CTC≥1) at V0, and 32.5% of them presented<br />

pathological (CTC≥5) at V0. A significant decrease was observed at<br />

V1, with 39.8% of patients with detectable CTC levels and 14.0%<br />

of patients with pathologic CTC levels (p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

expressed ALDH1 both before and after chemotherapy, however<br />

high ALDH1 expression levels in CTCs were more frequently<br />

observed after treatment (91% vs 54%). The median percentage of<br />

ALDH1high expressing CTCs per patient was also increased after<br />

chemotherapy (86% vs 64%). TWIST expression was observed in<br />

the great majority of CTCs both before and after chemotherapy,<br />

however nuclear localization of TWIST in CTCs was more often<br />

encountered after treatment (92% vs 54%). The median percentage<br />

of TWISTnuclear expressing CTCs per patient also increased after<br />

chemotherapy (89% vs 48%). High ALDH1 expression in CTCs<br />

was positively correlated with nuclear localization of TWIST, both<br />

pre- and post-chemotherapy (p=0.0000001 and p=0.001, respectively,<br />

Spearman analysis). Concerning the co-expression of these molecules,<br />

the phenotype ALDH1high TWISTnuclear was the most abundant<br />

at both time points, however it was more frequently observed after<br />

treatment (90% vs 41% of CTCs). In addition, the median percentage<br />

of ALDH1high TWISTnuclear expressing CTCs per patient was also<br />

increased after chemotherapy (71% vs 0%).<br />

Conclusions: Stemness and EMT markers are co-expressed in CTCs<br />

from patients with metastatic breast cancer, both before and after the<br />

first-line chemotherapy. However chemotherapy seems to enrich these<br />

characteristics on CTCs, suggesting that CTCs possessing or acquiring<br />

these features are resistant to chemotherapy. Evaluation of the postchemotherapy<br />

samples is ongoing and final results will be presented.<br />

P2-01-08<br />

Different numbers and prognostic significance of circulating<br />

tumour cells in patients with metastatic breast cancer according<br />

to immunohistochemical subtypes.<br />

Peeters DJ, van Dam P-J, Wuyts H, Van den Eynden GG, Jeuris K,<br />

Prové A, Rutten A, Peeters M, Pauwels P, Van Laere SJ, Hauspy J,<br />

van Dam PA, Vermeulen PB, Dirix LY. GZA Hospitals Sint-Augustinus,<br />

Antwerp, Belgium; University of Antwerp, Belgium; Catholic<br />

University of Leuven, Leuven, Vlaams-Brabant, Belgium<br />

Introduction: The enumeration of circulating tumour cells (CTCs) with<br />

the EPCAM-based CellSearch system has prognostic significance in<br />

patients with metastatic breast cancer (MBC). However, breast<br />

cancer has been shown to be a molecularly heterogeneous disease.<br />

The aim of this study was to assess potential differences in the<br />

detection and prognostic significance of CTCs according to the<br />

immunohistochemically defined molecular subtypes of breast cancer.<br />

Methods: CellSearch CTC counts were obtained from 110 patients<br />

with MBC prior to first line systemic treatment, treated at GZA<br />

Hospitals Sint-Augustinus between november 2007 and december<br />

2011. Clinicopathological variables were prospectively entered in<br />

a database. Based on the St-Gallen surrogate definitions of intrinsic<br />

breast cancer subtypes (Goldhirsch et al. Ann Oncol 2011), patients<br />

were divided in 5 groups: luminal A (ER/PR+, HER2-, Bloom-<br />

Richardson histological grade I-II), luminal B – HER2 negative (ER/<br />

PR+, Her2-, grade III), luminal B – HER2 positive (ER/PR+, HER2+,<br />

any grade), HER2 positive – non luminal (ER/PR-, HER2+), and<br />

triple negative (TN) (ER/PR-, HER2-). Differences in progression<br />

free survival (PFS) and overall survival (OS) according to the FDA<br />

approved prognostic cut-off of ≥5 CTC/7.5 ml blood were estimated<br />

using Kaplan Meier and Cox proportional hazard statistics.<br />

Results: CTC were detected in 78 of 110 (71%) patients. Higher<br />

detection rates and numbers of CTC were observed in patients with<br />

luminal A and TN breast cancer as compared to patients with luminal<br />

B and HER2 positive disease. However, no differences in positivity<br />

rates were observed between molecular subtypes according to the<br />

5 CTC prognostic cut-off point (table 1). After a median FU time<br />

of 3.1 years, 39 patients had died. In the total study population, the<br />

presence of ≥5 CTC was an independent predictor of PFS and OS in<br />

multivariate analysis (PFS: HRCTC≥5=2.236 (1.366-3.658), p=0.001;<br />

OS: HRCTC≥5=3.180 (1.553-6.509), p=0.002). When analyzing<br />

subgroups separately, a lower prognostic power was observed in the<br />

HER2 positive and luminal B subgroups.<br />

N<br />

Median #CTC/<br />

7.5 ml (range)<br />

N (%) with<br />

≥1 CTC/<br />

7.5 ml<br />

N (%) with ≥5<br />

CTC/7.5 ml<br />

Log Rank P-value<br />

PFS (< vs ≥5<br />

CTC/7.5 ml)<br />

Log Rank<br />

P-value OS<br />

(< vs ≥5 CTC/<br />

7.5 ml)<br />

All 110 3.5 (0-2262) 78 (71%) 54 (49%) 0.003 0.0002<br />

LumA 37 10.0 (0-2262) 28 (76%) 21 (57%) 0.039 0.020<br />

LumB -<br />

HER2-<br />

22 0.5 (0-253) 11 (50%) 7 (32%) 0.362 0.098<br />

LumB -<br />

HER2+<br />

24 3.5 (0-115) 15 (63%) 12 (50%) 0.616 0.678<br />

HER2+ -<br />

12 5.0 (0-86)<br />

nonluminal<br />

10 (83%) 6 (50%) 0.197 0.157<br />

TN 15 7.0 (0-1101) 14 (93%) 8 (53%) 0.033 0.026<br />

Kruskal-Wallis: Chi square: Chi square:<br />

p=0.042 p=0.034 p=0.46<br />

Table 1<br />

Conclusion: Significant differences were observed in the detection<br />

and prognostic significance of EPCAM positive CTC according to the<br />

immunohistochemically defined breast cancer subtypes. Interestingly,<br />

CTC were detected more frequently in patients with luminal A and TN<br />

tumors. Furthermore, our data suggest a lower prognostic significance<br />

of CTC evaluation in HER2 positive patients with MBC. Our data<br />

independently confirm those reported by Giordano et al. (Ann Oncol<br />

2010) in a large clinically uniform population of patients with MBC<br />

before the start of first-line treatment.<br />

P2-01-09<br />

Tumor cell emboli in the lung and transcriptional profiles<br />

of circulating tumor cells derived from different vascular<br />

compartments in patients with metastatic breast cancer.<br />

Peeters DJ, Van den Eynden GG, Rutten A, Onstenk W, Sieuwerts<br />

AM, De Laere B, van Dam PA, Peeters M, Pauwels P, Van Laere SJ,<br />

Vermeulen PB, Dirix LY. GZA Hospitals Sint-Augustinus, Antwerp,<br />

Belgium; University of Antwerp, Belgium; Erasmus University<br />

Medical Center and <strong>Cancer</strong> Genomics Center, Rotterdam, South<br />

Holland, Netherlands; Catholic University of Leuven, Leuven,<br />

Vlaams-Brabant, Belgium<br />

Background: We have shown that in up to 50% of patients with<br />

metastatic breast cancer (MBC) significantly higher numbers of<br />

circulating tumor cells (CTCs) can be detected in central venous blood<br />

(CVB) as compared to peripheral venous blood (PVB), suggesting that<br />

the lungs might retain a substantial number of CTCs from the blood<br />

stream (Peeters et al. Br J <strong>Cancer</strong> 2011). The aim of this study was<br />

1) to investigate the relation between elevated numbers of CTCs and<br />

the presence of (intravascular) tumor cell emboli (TCE) in the lung<br />

in patients with advanced carcinomas, and 2) to investigate whether<br />

CTCs derived from CVB and PVB exhibit differential transcriptional<br />

characteristics.<br />

Methods: Seven patients with MBC and 1 patient with metastatic<br />

cervical carcinoma, all suffering from end-stage disease, were<br />

included in the first part of this study. CTCs were isolated and<br />

enumerated with the CellSearch system (Veridex, Raritan, NJ, USA)<br />

in 7.5 ml blood obtained from the central venous access catheter<br />

(CVB) and/or a peripheral vein (PVB). All blood samples were<br />

obtained within 5 days prior to death. The presence of TCE was<br />

studied in lung tissue samples obtained at autopsy. For the second<br />

study aim, paired CVB and PVB CTC samples were collected from<br />

an additional 10 MBC and 2 LABC patients. Transcriptional profiles<br />

were obtained for 91 breast cancer related genes as described by<br />

Sieuwerts et al. (Clin <strong>Cancer</strong> Res 2011).<br />

Results: Multiple TCE were observed in 4 out of 6 patients with highly<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

218s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 2<br />

elevated numbers of CTCs (>100 CTC/7.5 ml blood). These TCE were<br />

located exclusively intravascularly in 2 patients, while the other 2<br />

patients had a more diffuse infiltration pattern with perivascular and<br />

lymphovascular TCE. All 4 patients had a history of rapidly evolving<br />

respiratory distress in the last week of life although radiological<br />

examination of the lungs did not show significant interval changes.<br />

In another 2 MBC patients with >100 CTCs and 2 MBC patients<br />

with


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

significant correlations for the expressions of CK8 (p


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 2<br />

breast cancer cells within the bloodstream. Whereas only rare (0.1-<br />

10%) cells in the primary tumor expressed both mesenchymal and<br />

epithelial markers, such biphenotypic as well as purely mesenchymal<br />

cells were enriched among CTCs, across all histological subtypes of<br />

breast cancer. Analysis of the therapy response in 8 patients suggest<br />

an association of mesenchymal CTCs with disease progression. In<br />

an index patient followed longitudinally, fluctuation in epithelial and<br />

mesenchymal states was observed as a function of initial response<br />

and subsequent resistance to therapy. Mesenchymal markers were<br />

predominant in clusters of tumor cells, many of which had adherent<br />

platelets. Finally, RNA sequencing of mesenchymal CTC clusters<br />

identified TGF-B and other EMT-related signatures, which were<br />

absent from more epithelial CTCs. FOXC1, a known regulator of<br />

EMT, was abundantly expressed in mesenchymal CTCs and was<br />

detectable within localized regions of the primary breast tumor.<br />

Together, these data support a role for EMT in the blood-borne<br />

dissemination of breast cancer and point to the dynamic nature of<br />

this cell fate change.<br />

P2-02-01<br />

Accurate identification of metastatic breast cancer using<br />

methylated gene markers in circulating free DNA in peripheral<br />

blood<br />

Sukumar S, Fackler MJ, Lopez-Bujanda Z, Teo WW, Jeter S, Umbricht<br />

C, Visvanathan K, Wolff AC. Johns Hopkins University School of<br />

Medicine, Baltimore, MD<br />

Background: Preliminary studies from our lab have shown that<br />

a panel of methylation markers in tissue identifies 100% of tested<br />

breast cancer and 95% of tested DCIS, and has high accuracy in cells<br />

from ductal fluid and spontaneous nipple discharge 1,2 . Other groups<br />

have reported on the use of a single marker or a panel of markers to<br />

detect breast cancer in serum or plasma. Cell-free DNA studies in<br />

the peripheral blood of breast cancer patients with advanced disease<br />

or with early-stage disease after completion of local therapy support<br />

the hypothesis that methylated DNA markers in serum or plasma<br />

could be used to monitor response to therapy and for long-term<br />

surveillance. Validation studies to test these hypotheses have been<br />

hampered by assay methodological issues such as the very small<br />

amount of DNA shed in the serum by tumor compared to the total<br />

DNA shed by normal cells.<br />

Methods: To overcome this problem, we developed a modified<br />

quantitative methylation-specific PCR that directly measures the<br />

number of copies of methylated DNA markers in a small aliquot of<br />

serum (Serum-QM-MSP) and robustly detects less than 25 copies<br />

of DNA in 300 µL of serum. We then conducted a genome-wide<br />

methylome analysis to identify key markers that are preferentially<br />

methylated in serum from women with breast cancer and compared the<br />

profiles to those from women with no breast cancer. We then analyzed<br />

300 µL each of sera from 55 normal women (single time point) and<br />

43 women with metastatic breast cancer using this newly developed<br />

panel of markers and the Serum-QM-MSP assay. We also examined<br />

changes after therapy in a subset of patients with metastatic disease.<br />

Results: Methylation markers were quantitatively detected in sera<br />

of 39 out of 43 (91% sensitive) metastatic breast cancer patients<br />

with varying tumor burdens, and not in sera of any of 55 women<br />

(100% specific) for an AUC=0.95, using a laboratory threshold of<br />

7.2 cumulative methylation units. 28 of the 43 patients had sampling<br />

repeated 3-5 weeks after therapy started. Sera from patients whose<br />

tumors regressed and from those that had stable disease showed a<br />

quantitative reduction, while those with progressive disease showed<br />

an increase in methylation levels of several genes.<br />

Conclusion: Our results suggest that methylated DNA in serum<br />

accurately discriminates between blood samples from normal women<br />

and from metastatic breast cancer patients. Also, early changes after<br />

therapy initiation for metastatic disease may correlate with subsequent<br />

clinical outcome. Assay analytical validation studies are ongoing.<br />

Studies examining a potential role in surveillance in the adjuvant<br />

setting and therapeutic benefit in the metastatic setting are warranted.<br />

1. Fackler MJ et al. Genome-wide methylation analysis identifies<br />

genes specific to breast cancer hormone receptor status and risk<br />

of recurrence. <strong>Cancer</strong> Res. 2011 Oct 1;71(19):6195-207. PMCID:<br />

PMC3308629.<br />

2. Fackler MJ, et al. Hypermethylated genes as biomarkers of cancer<br />

in women with pathologic nipple discharge. Clin <strong>Cancer</strong> Res. 2009<br />

Jun 1;15(11):3802-11. PMID: 19470737<br />

P2-02-02<br />

Prognostic significance of the ratio of absolute neutrophil to<br />

lymphocyte counts for breast cancer patients in neoadjuvant<br />

setting<br />

Koh YW, Lee HJ, Ahn J-H, Lee JW, Gong G. University of Ulsan<br />

College of Medicine, Asan Medical Center, Seoul, Korea; Seoul<br />

National University Bundang Hospital, Seongnam, Bundang-Gu,<br />

Kyeonggi-Do, Korea<br />

Background: The baseline absolute neutrophil count/absolute<br />

lymphocyte count ratio (NLR) has potential prognostic importance in<br />

solid tumors, including breast cancer. However, NLR as a predictive<br />

factor of the pathologic complete response (pCR) or as a prognostic<br />

factor has not been fully studied in breast cancer patients who have<br />

received neoadjuvant chemotherapy.<br />

Materials and Methods: Using receiver operating characteristic curve<br />

analyses, we retrospectively examined the predictive and prognostic<br />

impact of the NLR at the diagnosis in 401 patients with primary breast<br />

cancer who were treated with neoadjuvant chemotherapy, followed by<br />

definitive surgical resection. Of the 401 included patients, 72 (18%)<br />

received anthracycline-based, 285 (71%) received anthracycline- and<br />

taxane-based, 37 (9.2%) received herceptin-based, and 7 (1.7%)<br />

received fluorouracil-based regimens. The significance of NLR<br />

was analyzed according to clinicopathologic variables and clinical<br />

outcomes.<br />

Results: Forty-three (10.7%) patients achieved pCR. On univariate<br />

analysis, high NLR (> 2.2) correlated with poor recurrence-free<br />

survival (RFS; P = 0.001) and overall survival (OS; P = 0.003).<br />

Subgroup analysis of the non-pCR group showed that NLR was<br />

significant for RFS and OS (P = 0.002 and P = 0.007, respectively).<br />

Multivariate analysis showed NLR to be an independent prognostic<br />

factor for RFS and OS (P = 0.002 and P = 0.007, respectively).<br />

Multivariate analyses of recurrence-free survival and overall survival according to clinicopathologic<br />

variables<br />

Multivariate<br />

Variables<br />

analysis<br />

Recurrencefree<br />

survival<br />

95%<br />

Harzard<br />

confidence<br />

ratio<br />

interval<br />

P<br />

Harzard<br />

ratio<br />

Overall<br />

survival<br />

95%<br />

confidence<br />

interval<br />

Lymphovascular<br />

(-) vs. (+) 1.48 0.912–2.402 0. 112 1.413 0.740–2.700 0. 295<br />

invasion<br />

ER and PR status (-) vs. (+) 0.32 0.198–0.517


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Conclusion: Our results suggest that the NLR is an independent<br />

prognostic factor for RFS and OS in breast cancer patients receiving<br />

neoadjuvant chemotherapy and may provide additional prognostic<br />

information for patients not achieving a pCR to determine who might<br />

profit from further therapy.<br />

P2-02-03<br />

The Collagen Receptor Endo180: A Metastatic Plasma Marker In<br />

<strong>Breast</strong> <strong>Cancer</strong> Modulated By Bisphosphonate Treatment<br />

Sturge J, Caley MP, Purshouse K, Fonseca A-V, Rodriguez-Teja M,<br />

Kogianni G, Waxman J, Palmieri C. Imperial College London, United<br />

Kingdom; The University of Hull, Hull, United Kingdom<br />

Purpose: To establish whether the collagen remodelling receptor,<br />

Endo180, should be given consideration as a useful plasma marker<br />

in metastatic breast cancer.<br />

Patients and Methods: Analysis of MCF-7 and MDA-MB-231 cell<br />

conditioned medium validated the anti-human (39.10) monoclonal<br />

antibody as suitable for the detection of soluble Endo180 in<br />

plasma. Eighty-seven breast cancer patients with early primary,<br />

locally advanced and metastatic disease were included in the study.<br />

Correlations between Endo180, CA 15-3 antigen (MUC1; mucin 1)<br />

and bisphosphonate treatment were investigated.<br />

Results: Endo180 was elevated in metastatic compared to early breast<br />

cancer (P


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 2<br />

disease-free survival (DFS) were calculated from the date of diagnosis<br />

and analyzed with Kaplan-Meier survival plots. Cox multivariate<br />

proportional hazards model was also performed.<br />

Results: Of the 103 evaluable patients, 45 patients received<br />

neoadjuvant chemotherapy, 35 adjuvant chemotherapy and 23 no<br />

chemotherapy. The median follow-up was 40 months (range 7-66<br />

months). Patients with CSCs in BM had a hazard ratio (HR) of<br />

8.8 for DFS (p = 0.002); patients with Aldefluor+ CSCs had a HR<br />

of 5.9 (p = 0.052) for DFS. Seven patients have expired and all of<br />

them had CSCs in BM. Moreover, we evaluated the presence of<br />

DTCs by FACS (CD326+CD45-) in 104 patients. Sixty-one (75%)<br />

of patients had positive BM samples for DTC (CD326+CD45- BM<br />

cells ≥ 0.53%). No association was shown between DTC in BM as<br />

determined by FACS and CTCs in PB. In a multivariate model, after<br />

adjusting for age (< 45 years old), clinical T stage, N stage, ER, PR,<br />

HER2 status, and nuclear grading, the presence of CSCs was found<br />

to be an independent predictor of DFS (HR = 15.8, P = 0.017) as<br />

was the presence of CTCs (HR = 13.9, P = 0.007). In this subset of<br />

patients, the presence of Aldefluor+ CSCs and DTCs was not found<br />

to be predictive of DFS in the multivariate model including the same<br />

factors as listed above.<br />

Conclusions: Our data indicate that the presence of BM metastasis<br />

is correlated to CSCs, and CSCs irrespective of ALDH activity are<br />

independent adverse prognostic factors in EBC patients. Moreover,<br />

the presence of CTCs was a strong independent predictor of DFS.<br />

P2-04-01<br />

Development of mouse breast cancer models based on induced<br />

cancer stem cells (iCSC).<br />

Takamoto Y, Onishi N, Kai K, Saya H. Institute for Advanced Medical<br />

Research (IAMR), School of Medicine, Keio university, Shinjyuku-ku,<br />

Tokyo, Japan; The University of Texas MD Anderson <strong>Cancer</strong> Center,<br />

Houston, TX<br />

<strong>Breast</strong> cancer is one of the leading causes of death in women<br />

worldwide. To develop novel therapeutic approaches for the<br />

refractory cases, the mouse models which recapitulate the tumor<br />

tissues biologically and pathologically similar to human breast<br />

cancer are required. Although xenograft models of established cell<br />

lines in immune-deficient mice are frequently used for preclinical<br />

experiments, such xenograft models are not sufficient because the<br />

heterogenous structure based on the microenvironment and the<br />

intrinsic characteristics of cancer cells is not correctly formed.<br />

In this study, we have established induced cancer stem cells (iCSC)<br />

from normal mouse mammary stem/progenitor cells through minimal<br />

required genetic manipulations and generated mouse breast cancer<br />

models by inoculating the iCSCs in the mammary fat pads. Initially,<br />

we established iCSC by introducing the H-RasV12 into Ink4a/Arfknockout<br />

mammary stem/progenitor cells and this iCSC formed tumor<br />

similar to human triple negative breast cancer in mouse. This finding<br />

suggested that two genetic events, an activation of oncogenic signal<br />

and a tumor suppressor inactivation, are required for generating the<br />

breast cancer iCSC.<br />

Anaplastic Lymphoma Kinase (ALK) gene, which encodes a receptor<br />

tyrosine kinase, was reported to be amplified and/or overexpressed<br />

up to 86% of patients in inflammatory breast cancer, and pleiotrophin<br />

(PTN), which is a physiological ligand for ALK, was also shown to be<br />

highly expressed in about 60% of human breast cancers. Therefore, we<br />

hypothesized that ALK pathway is involved in tumorigenesis of breast<br />

cancers and, then, attempted to generate iCSC by using ALK gene.<br />

Interestingly, we found that one of the naturally occurring mutations<br />

of ALK is sufficient for generating iCSC and tumor formation in<br />

vivo without any prior tumor suppressor inactivation. The ALKinduced<br />

iCSCs developed highly aggressive breast cancers in mice.<br />

Furthermore, the tumor formation was significantly suppressed when<br />

the ALK-induced iCSCs were generated by using mammary stem/<br />

progenitor cells derived from mouse deficient in CD44 which is a<br />

CSC marker. We have recently revealed a role of CD44, in particular<br />

that of a variant isoform (CD44v), in the protection of CSCs from<br />

high levels of oxidative stress derived from both tumor cells and their<br />

microenvironment (<strong>Cancer</strong> Cell 19: 387-400, 2011; <strong>Cancer</strong> Res 72:<br />

1438-1448, 2012; Nat Commun 3: 883, 2012). We will discuss the<br />

underlying mechanism of ALK-induced tumorigenesis and a role of<br />

CD44 in the CSC functions.<br />

P2-04-02<br />

Identification of genes associated with breast cancer<br />

micrometastatic disease in bone marrow using a human-inmouse<br />

xenograft system<br />

Aft R, Li S, Mudalagiriyappa C, Dasgupta N, Watson M, Fleming T,<br />

Ellis M, Pillai S. Washington University, St. Louis, MO<br />

Disseminated tumor cells (DTCs) found in the bone marrow (BM)<br />

of breast cancer patients portend a poor prognosis and are thought<br />

to be the intermediaries in the metastatic process. Study of these<br />

cells has been limited due to their scarcity. To develop a clinically<br />

relevant model to characterize hese cells, we have employed a human<br />

in mouse (HIM) xenograft model for propagating, isolating, and<br />

molecularly characterizing DTCs. Human breast adenocarcinomas<br />

were prospectively collected from 5 patients and implanted into<br />

humanized NOD/SCID mouse mammary fat pads. BM was collected<br />

from the long bones at varying passages of the tumors and analyzed<br />

for human-specific gene expression by qRT-PCR and gene expression<br />

microarray. Human-specific gene expression of SNAI1, GSC, FOXC2,<br />

KRT19, and STAM2, presumably originating from disseminated<br />

tumor cells, was detectable in the BM of all mice that had developed<br />

metastatic disease to other solid organs, but was not detectable<br />

in xenotransplanted mice that did not develop metastatic disease.<br />

Comparative gene expression microarray analysis of the HIM primary<br />

tumor, the corresponding BM from mice with metastatic disease,<br />

and BM from control mice identified additional patterns of gene<br />

expression enriched in BM-associated DTCs which included several<br />

genes associated with epithelial-mesenchymal transition, aggressive<br />

clinical phenotype, and metastatic disease development in primary<br />

human tumors. We have found that BM DTCs can be detected using<br />

the HIM xenograft model and have identified unique patterns of gene<br />

expression associated with BM DTCs, which may provide further<br />

insight into the biology and therapeutic vulnerability of metastatic<br />

tumor cell populations in breast cancer patients.<br />

P2-04-03<br />

A robust transgenic mouse model to study male breast cancer.<br />

Krause S, Lurvey HL, Tobin H, Ingber DE. Boston’s Children’s<br />

Hospital, Boston, MA; Wyss Institute of Biologically Inspired<br />

Engineering, Boston, MA<br />

Male breast cancer accounts for about 1% of all breast cancers and<br />

even though it is a rare disease, the incidence rate has increased<br />

steadily during the past 20 years. Study of the male form of the<br />

disease has been hampered by a lack of relevant animal models as<br />

it is commonly believed that male mammary glands do not contain<br />

ducts. Female FVB C3(1)-SV40Tag transgenic mice spontaneously<br />

develop hyperplastic mammary lesions that progress to invasive<br />

mammary tumors, whereas most of the transgenic males develop<br />

prostatic hyperplasia that progresses to adenocarcinoma. However,<br />

www.aacrjournals.org 223s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

upon closer examination, we discovered that approximately 45% of<br />

transgenic males develop mammary tumors by 40 weeks of age, often<br />

prior to developing prostate tumors. The course of male mammary<br />

development is similar to that observed in the females but with a<br />

later onset. Hyperplastic ducts form as early as 15 weeks of age<br />

and progress into DCIS-resembling tumors before developing into<br />

invasive tumors that invade locally into adipose tissue and muscle<br />

and occasionally form metastases to lung and other organs. We then<br />

examined male mammary development in the FVB background<br />

strain (wild type) and showed that all males develop mammary<br />

ducts that continue to grow and branch throughout adulthood. The<br />

morphology of these ducts was similar to that of females with a<br />

central lumen surrounded by concentric layers of luminal epithelium,<br />

myoepithelium, a basement membrane and surrounding stroma. A<br />

study of CD1 male mice confirmed that male mammary development<br />

was not specific to the FVB strain but rather, CD1 males showed<br />

similar mammary ductal development. Male mammary development<br />

in FVB wild type animals and tumor progression in the transgenic<br />

males was characterized in terms of cell proliferation, apoptosis and<br />

expression of various markers of differentiation including hormone<br />

receptor status. In summary, our results demonstrate that FVB C3(1)-<br />

SV40Tag transgenic males represent a highly robust and reproducible<br />

model for the study male breast cancer. Our long-term goal is to<br />

leverage the information we uncovered through this effort to design<br />

novel therapies for breast cancer treatment in men.<br />

P2-04-04<br />

Prolactin cooperates with loss of p53 to promote mammary<br />

tumorigenesis.<br />

O’Leary KA, Sullivan R, Rugowski DE, Schuler LA. University of<br />

Wisconsin, Madison, WI<br />

Prolactin (PRL) is critical for mammary development and lactation.<br />

Epidemiological studies also support a role for PRL in breast cancer.<br />

In order to study the contributions of PRL to tumor development and<br />

progression, we developed a transgenic mouse model that expresses<br />

PRL in the mammary epithelial cells (NRL-PRL), mimicking the<br />

mammary PRL production in women. Loss of function of the tumor<br />

suppressor p53 is a common occurrence in many human cancers<br />

including breast tumors. Interactions between PRL and p53 were<br />

examined by crossing the NRL-PRL mouse to p53 knockout mice<br />

which were made congenic on the FVB/N background. To circumvent<br />

the problem that p53 -/- mice are prone to multiple nonmammary<br />

tumors, mammary cells from 10-12 week old wild-type, NRL-<br />

PRL, p53 -/- , and NRL-PRL/ p53 -/- were transplanted to wild type<br />

mammary glands. Histological analysis of the donor glands showed<br />

that NRL-PRL glands displayed marked epithelial proliferation<br />

and focally dilated ducts, whereas p53 -/- glands exhibited irregular<br />

ductal epithelium with increased stromal density compared to<br />

wild type mice. In contrast, glands of donor PRL/ p53-/- females<br />

showed widespread epithelial hyperplasias and highly irregular<br />

ductal epithelium often surrounded by dense stroma. By one year of<br />

age, no tumors had developed from either wild-type or NRL-PRL<br />

transplanted epithelium. However, recipients of either p53 -/- or NRL-<br />

PRL/ p53 -/- mammary epithelium developed histologically similar<br />

anaplastic carcinomas. The presence of transgenic PRL decreased<br />

tumor latency (205 vs 244 days) and significantly increased tumor<br />

cell proliferation. In addition, these carcinomas appeared to be more<br />

aggressive: 5/16 (32%) of the NRL-PRL/ p53 -/- tumors invaded into<br />

the peritoneal cavity, compared to 0/10 p53 -/- tumors. Expression of<br />

matrix metalloproteinase 9 (MMP-9), but not MMP-2, was found to be<br />

significantly higher in the NRL-PRL/ p53 -/- , compared to p53 -/- tumors.<br />

MMP-9 is regulated by the transcription factor, AP-1, and PRL can<br />

signal through AP-1. NRL-PRL/ p53 -/- tumors displayed significantly<br />

increased expression of the AP-1 family members, c-Jun and FosL, but<br />

not JunD or c-Fos compared to tumors from p53 -/- mice. In addition,<br />

levels of c-Jun and FosL proteins increased in a similar pattern. In<br />

summary, interactions between PRL and loss of p53 promote breast<br />

cancer by increasing proliferation and invasiveness, potentially via<br />

AP-1 target genes. Supported by CDMRP BC053412.<br />

P2-04-05<br />

Prolonged targeted overexpression of Aurora-A in mammary<br />

epithelium promotes mammary adenocarcinoma with genomic<br />

instability<br />

Treekitkarnmongkol W, Katayama H, Sen S. University of Texas M.D.<br />

Anderson <strong>Cancer</strong> Center, Houston, TX<br />

Aurora kinase-A (referred to as Aurora-A) identified as a sereine/<br />

threonine kinase induces mitotic progression process including<br />

centrosome amplification, bipolar spindle formation and chromosome<br />

segregation through modulation of activity and localization of its<br />

interacting proteins during cell division. Beside its essential roles in<br />

mitosis, cancer profiling studies have revealed that Aurora-A function<br />

is implicated in tumor relevant signaling pathways and overexpressed<br />

in many human tumors. The findings showed highly frequent<br />

overexpression of Aurora-A in human breast ductal carcinoma in<br />

situ and primary invasive ductal breast carcinoma. Moreover, the<br />

Aurora-A overexpression correlates with poor prognosis in breast<br />

carcinoma, deregulated overexpression of Aurora-A associates with<br />

centrosome anomalies and chromosome instability in rodent models<br />

of mammary caricinogenesis.<br />

However, published studies in several transgenic mouse models<br />

attempting to address the role of Aurora-A in tumorigenesis and<br />

genomic instability have raised conflicting results. In this report we<br />

targeted expression of Aurora-A transgene in an inducible mouse<br />

model in which expression of Aurora-A was restricted to mammary<br />

epithelium during multiple pregnancy and lactation cycles under the<br />

activity of β-lactoglobulin promotor. The results demonstrate that<br />

overexpression of Aurora-A could induce tumorigenesis in mouse<br />

mammary epithelium. The tumor incidence was 70% of Aurora-A<br />

transgenic mice at 16 months of age. Of note, overexpression<br />

of Aurora-A led to genomic instability. Comparative genomic<br />

hybridization (CGH) array analyses revealed loss of Trim12a,<br />

Trim12c, Trim30b, Trim30d, Trim30e, Cdkn2d, Pten, Cep55 and<br />

gain of Adam6. Notably, Aurora-A overexpression caused nuclear<br />

accumulation of cyclin D1, activation of AKT, overexpression of<br />

TPX2 and PLK1and loss of ERα expression in tumors. Our findings<br />

indicated that prolonged Aurora-A expression in mammary epithelium<br />

leads to mammary adenocarcinomas with genomic instability.<br />

Aurora-A driven adenocarcinoma reveals deregulation of critical<br />

tumor relevant genetic pathways involving both oncogenes and tumor<br />

suppressor genes. These transgenic mouse model findings indicate that<br />

Aurora-A may be an important therapeutic target for mammary cancer.<br />

Keywords: Aurora-A, carcinogenesis.<br />

P2-04-06<br />

Transgenic expression of a breast-cancer specific GATA3 mutant<br />

leads to mammary hyperplasia<br />

Kenny PA, Chandiramani N. Albert Einstein College of Medicine,<br />

Bronx, NY<br />

GATA3 is expressed in normal luminal breast epithelial cells and is<br />

a key transcription factor in mammary gland development. In human<br />

breast tumors, high GATA3 protein levels are associated with estrogen<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

224s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 2<br />

receptor (ER) positivity and low grade. Recent published reports<br />

indicate that somatic mutations of GATA3 occur in up to 21% of ER+<br />

tumors. These authors suggest, but do not demonstrate, that these<br />

GATA3 mutations are loss-of-function null alleles and that GATA3<br />

is a haploinsufficient tumor suppressor gene.<br />

The published mutational data indicate a striking enrichment of<br />

GATA3 mutations in exons 5 and 6 (amino acids 309-443) which<br />

encode the second of two DNA binding zinc fingers. This suggests<br />

that mutant proteins lacking this zinc finger may confer a survival<br />

advantage on breast cancer cells. Given this apparently strong<br />

selective pressure for mutations in this hotspot, it seems unlikely that<br />

these mutations are loss-of-function alleles.<br />

We explored this directly by generating a MMTV-GATA3mutant<br />

mouse which specifically expresses a mutant GATA3 protein in<br />

the mammary gland. Non-parous pubertal transgenic mice exhibit<br />

a pronounced mammary hyperplasia and have increased ductal<br />

branching compared to wildtype littermates. These data confirm<br />

that the mutant GATA3 mutations commonly found in breast cancer<br />

are not null alleles, but that they instead have a gain-of-function<br />

phenotype. We are currently evaluating the effect of mutant GATA3<br />

on the kinetics of mammary tumorigenesis by crossing these mice<br />

to the MMTV-PyMT model.<br />

P2-04-07<br />

Modeling orthotopic and metastatic progression of mammary<br />

tumors to evaluate the efficacy of TGF-β inhibitors in a preclinical<br />

setting<br />

Biswas T, Yang J, Zhao L, Sun L. UTHSCSA, <strong>San</strong> <strong>Antonio</strong>, TX;<br />

UCSD, CA<br />

Preclinical models are elemental in understanding drug efficacy and<br />

safety in vivo. Many existing models of breast cancer metastasis use<br />

immune compromised animals and do not effectively characterize<br />

the differential stages of metastasis. Transforming growth factor-β<br />

(TGF-β) has been shown to be a major contributor in epithelialmesenchymal<br />

transition and enhances various steps during metastasis.<br />

The aim of this study is to develop mouse models of tumor metastasis<br />

using murine mammary cells in a syngeneic background. This will<br />

help illustrate the effect of TGF-β inhibitors on metastasis in an<br />

immune competent environment, and also allow determining the<br />

optimal time of drug administration. We demonstrate that TGF-β<br />

signaling has differential effects on a pair of murine mammary<br />

cell lines derived from PyMT tumors in C57Bl/6 mice. These cell<br />

lines represent luminal (Py 230) and basal (Py 8119) epithelial cell<br />

populations in the tumor. In vitro characterization revealed differences<br />

in viability, motility, and transcriptional and translational activation of<br />

Smad. Py 230 (clonal luminal) cells are growth inhibited and exhibit<br />

epithelial-mesenchymal transition on treatment with TGF-β, whereas<br />

Py 8119 (basal cells) are non-responsive to both effects. Furthermore,<br />

intra cardiac injections were performed using luciferase-GFP labeled<br />

PyMT cells in syngeneic background and animals were treated with<br />

either placebo or TβRI-KI (kinase inhibitor for TGF-β receptor I).<br />

Results from Py 8119 cell injections showed that TβRI-KI treatment<br />

enhanced bone and brain metastases. Further results will shed light on<br />

the effect of TβRI-KI during specific stages of metastasis progression.<br />

P2-04-08<br />

Targeted Expression of the Human Chaperone BAG3 to the<br />

Murine Mammary Gland Dysregulates Mammary Gland<br />

Development and Differentiation By Unrestricted Expansion of<br />

Luminal Cells<br />

Virador VM, Casagrange G, Raafat A, Callahan R, Kohn E. National<br />

<strong>Cancer</strong> Institute, Bethesda, MD<br />

BAG3 is a pro-survival pro-invasion chaperone. We hypothesized that<br />

human BAG3 overexpression in murine breast would contribute to<br />

mammary tumorigenesis. We generated transgenic mice ectopically<br />

expressing hBAG3 in the mammary gland under the control of the<br />

Mouse Mammary Tumor Viral promoter (MMTV-hBAG3). hBAG3<br />

was expressed in luminal cells throughout the ductal tree in multiple<br />

transgenic lines. Targeted expression of hBAG3 dysregulated<br />

mammary gland development by both increasing proliferation and<br />

differentiation. hBAG3 mammary glands had increased epithelial<br />

elongation at 5 weeks of age, premature glandular differentiation at 10<br />

weeks, and features of ductal hyperplasia at week 13 with increased<br />

Ki67 and p-Histone H3 in the luminal compartment. At week 13<br />

hBAG3 induced acinar differentiation. BAG3 pro-survival gain of<br />

function was recapitulated post-partum with delayed involution.<br />

Mammary glands of MMTV-hBAG3 had alveolar persistence<br />

three days after forced involution where non-transgenic controls<br />

demonstrated typical involution-driven apoptosis and remodeling.<br />

We crossed heterozygous MMTV-hBAG3 to WAP-Int3 mice with<br />

absent alveolar development in pregnancy. hBAG3 caused alveolar<br />

budding in the WAP-Int3 background. We hypothesized hBAG3 gain<br />

of function in the mammary gland would promote tumorigenesis.<br />

On aging, mammary glands of heterozygous MMTV-hBAG3<br />

displayed densely crowded acinar structures with atypia, consistent<br />

with hyperplastic alveolar nodule (HAN). We subjected MMTVhBAG3<br />

mice to either multiparity or chemical carcinogenesis. After<br />

two pregnancies, most MMTV-hBAG3 animals had hyperplastic<br />

mammary ductal lesions (MMTV-hBAG3 6/7, 86%; FVB/N 1/7,<br />

14%). Eighteen weeks post 7,12-dimethylbenz(a)antracene (DMBA)<br />

treatment, virgin females from five different transgenic lines had<br />

more hyperplastic lesions (MMTV-hBAG3 mean 6.3 lesions/gland,<br />

N=13, FVB/N mean 3.6 lesions/gland, N=9; p=0.04), some with<br />

atypical hyperplasia. Transgenic MMTV-hBAG3 females developed<br />

ER and PR (+) malignant tumors (5/19), 26% while non-transgenic<br />

controls had none (0/12). WAP-Int3 mice developed tumors as early<br />

as two pregnancies. Heterozygous WAP-Int3xMMTV-BAG3 had<br />

the same tumor frequency but preliminary data suggest increased<br />

aggressiveness and metastasis. Collectively, our results indicate that<br />

MMTV-hBAG3 promotes proliferation, dysregulates mammary gland<br />

development and promotes hyperplasia and tumorigenesis and may<br />

provide a novel mouse model to represent the human ER+ luminal<br />

breast cancer subtype.<br />

P2-05-01<br />

Gene expression changes associated with response to neoadjuvant<br />

chemotherapy are observed early in treatment: results from the<br />

I-SPY 1 TRIAL (CALGB 150007/150012; ACRIN 6657)<br />

Wolf DM, Yau C, Magbanua M, Boudreau A, Davis S, Haqq C, Park<br />

J, I-SPY TRIAL-1 Investigators, Esserman L, van’t Veer L. University<br />

of California, <strong>San</strong> Francisco, CA; I-SPY 1 TRIAL Institutions<br />

Background: Prior gene expression profiling studies have identified<br />

markers that are predictive of chemotherapy response using pretreatment<br />

biopsies. However, this approach does not account for<br />

chemotherapy-induced perturbation in signaling within tumors<br />

early in treatment that may predict response to therapy with<br />

www.aacrjournals.org 225s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

superior sensitivity to baseline signatures. We hypothesized that<br />

measuring early changes in gene expression induced by neoadjuvant<br />

chemotherapy might yield improved markers of treatment efficacy<br />

and patient outcome.<br />

Methods: Transcriptome data from 34K probe cDNA microarrays<br />

were assembled using serial biopsies obtained from 36 I-SPY<br />

1 TRIAL patients before treatment (T1), and 24-72 hours after<br />

beginning neoadjuvant anthracycline-based chemotherapy (T2).<br />

Outcome parameters included residual cancer burden (RCB) after<br />

therapy and recurrence free survival (RFS). Gene expression changes<br />

occurring between T1 and T2 (T2-T1) were compared between<br />

known responders (RCB 0/1) and non-responders (RCB 2/3) using<br />

a permutation test based on the t-statistic; Cox proportional hazards<br />

modeling was used to identify early response genes associated with<br />

RFS. Due to the small sample sizes, we adopted a relaxed significance<br />

threshold for outcome associations (p-value


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 2<br />

P2-05-04<br />

Differential potency of TORC1/C2 (INK/MLN-128) and<br />

pan-PI3K (GDC0941) inhibitors on breast cancer polysome<br />

composition and phosphoprotein response biomarkers<br />

Wilson-Edell KA, Yevtushenko M, Hanson I, Rogers AN, Scott GK,<br />

Benz CC. Buck Institute for Research on Aging, Novato, CA<br />

The PI3K (phosphoinositide-3 kinase)/AKT (protein kinase B)<br />

pathway is dysregulated in over 70% of breast cancers, including<br />

more than 40% of non-basal breast cancers that possess activating<br />

PIK3CA mutations and 15% of basal breast cancers with functional<br />

loss of PTEN (n=357 TCGA samples), suggesting that inhibitors of<br />

this signaling pathway may be clinically beneficial. Moreover, the<br />

recently reported BOLERO-2 trial (N Engl J Med, 2012) demonstrated<br />

the potential clinical benefit of using a TORC1 (target of rapamycin<br />

complex I) inhibitor, Everolimus, to treat patients with advanced<br />

non-basal, ER-positive breast cancers presumeably enriched with<br />

PIK3CA mutations. Among the new PI3K/AKT/TOR pathwaytargeted<br />

therapeutics now under clinical development are the dual<br />

TORC1/C2 inhibitor, INK/MLN-128, and the pan-PI3K inhibitor,<br />

GDC0941. Despite having comparable nanomolar target affinities,<br />

it is unclear whether similar subsets of breast cancers will respond<br />

similarly to these agents, or if targeting this PI3K/AKT/TOR pathway<br />

either upstream (e.g. by GDC0941) or downstream (e.g. by INK/<br />

MLN-128) will yield comparable antitumor effects. Many of the<br />

downstream cellular responses regulated by this pathway, including<br />

tumor cell survival, growth, metabolism, invasiveness and angiogenic<br />

potential, are mediated by phosphorylation of polysome effector<br />

proteins, including the known TORC1 regulated eukaryotic translation<br />

initiation factor 4 binding protein (4eBP1) and 40S ribosomal protein<br />

S6. Therefore, we compared the effects of INK/MLN-128 and<br />

GDC0941 on cell growth and polysome protein phosphorylation<br />

profiles using a model breast cancer cell line (SKBr3) with amplified<br />

upstream receptor (HER2) activation of the PI3K/AKT/TOR pathway.<br />

Continuous dosing of these two agents against SKBr3 cultures (0-625<br />

nM x5 days) revealed a 50-fold greater anti-proliferative efficacy for<br />

INK/MLN-128, with IC 50 = 9nM compared to the GDC0941 IC 50 =<br />

479nM. This difference in antiproliferative activity correlated with<br />

their 50-fold different doses required to inhibit polysome formation<br />

(relative to free 40S, 60S, and 80S ribosomes) and polysomal 40S<br />

Rack1 and 60S RPL24 content. Likewise, INK/MLN-128 more<br />

potently inhibited the phosphorylation of two polysome-associated<br />

proteins of 32 KD and 60 KD size, recognized by an antibody toward<br />

a motif phosphorylated by AKT and related kinases (RXXS*/T*); the<br />

32 KD substrate has been identified as S6. With equipotent dosing<br />

(0.1 mM INK/MLN-128 and 5 mM GDC0941), these two agents<br />

showed comparable inhibitory effects on polysome formation and<br />

protein phosphorylation. In summary, in a breast cancer model bearing<br />

upstream receptor activation of the PI3K/AKT/TOR pathway, distal<br />

pathway inhibition of TORC1/C2 by INK/MLN-128 appeared more<br />

potent and effective than more proximal inhibition of pan-PI3K by<br />

GDC0941. Furthermore, polysome inhibitory effects following INK/<br />

MLN-128 exposure were apparent within 8 h of treatment, and the<br />

most sensitive polysome response biomarker for this agent appeared<br />

to be phospho(T389)-S6 kinase, providing rationale for the use of<br />

this response biomarker in future clinical trials.<br />

P2-05-05<br />

Circulating HER2 extracellular domain (ECD) predicts a poor<br />

prognosis for metastatic breast cancer patients<br />

Wang X-j, Shao X-Y, Xu X-H, Chen Z-H, Wang S, Cai J-F, Huang J,<br />

Huang P, Lou C-J, Ling Z-Q, Han MX. Zhejiang <strong>Cancer</strong> Hospital,<br />

Hangzhou, Zhejiang, China; Hangzhou Polar Gene Company,<br />

Hangzhou, Zhejiang, China<br />

Background: Amplification or overexpression of HER2 has been<br />

shown to play an important role in approximately 30% of breast<br />

cancers and is strongly associated with increased recurrence and a<br />

worse prognosis. The HER2 extracellular domain (ECD) may be<br />

cleaved and shed from the surface of breast cancer cells and serum<br />

HER2 ECD levels can be detected by enzyme-linked immunosorbent<br />

assay (ELISA). In this study, we explored the relationship between<br />

circulating HER2 ECD and tissue HER2 status, then we also examined<br />

its predictive value in a cohort of metastatic breast cancer patients.<br />

Methods: Two hundred and seven metastatic breast cancer patients<br />

from March 2009 to July 2011 were involved in this prospective<br />

study at Zhejiang <strong>Cancer</strong> Hospital. The patients were identified<br />

histopathologically as having breast cancer by pathologists and<br />

staged mainly based on the pathology, the clinical manifestation, and<br />

the imaging findings of CT and MRI according to the classification<br />

system of the International Union Against <strong>Cancer</strong>. Serum HER2 ECD<br />

levels were measured by ELISA. Tissue HER2 was determined by<br />

IHC and FISH in tumor samples, respectively.<br />

Results: The level of serum HER2 ECD was at least more than 15 ng/<br />

ml in 31.4% (65/207) metastatic breast cancer patients, 39.1%(43/110)<br />

in HER2-positive patients and 23.4%(22/94) in HER2-negative cases,<br />

respectively, P=0.017. We also found that high serum HER2 ECD<br />

levels (≥15 ng/ml) were significantly associated with elevated serum<br />

CEA (52.1% v 21.5%, OR 3.978, 95%CI 2.138-7.401, P=0.000),<br />

CA125 (48.5% v 23.5%, OR 3.064, 95%CI 1.650-5.691, P=0.000),<br />

CA153 (53.2% v 17.1%, OR 5.48, 95%CI 2.897-10.366, P=0.000),<br />

LDH (53.3% v 23.1%, OR 3.798, 95%CI 2.011-7.173, P=0.000) and<br />

AKP (51.2% v 26.5%, OR 2.911, 95%CI 1.442-5.879, P=0.002),<br />

respectively. For the effect of HER2 ECD on the site of relapse, still<br />

there were statistically significant correlation between increased<br />

serum HER2 ECD levels and liver involvement (42.7% v 24.0%,<br />

OR 2.358, 95%CI 1.294-4.297, P=0.005), brain metastasis (50.0%<br />

v 26.7%, OR 2.744, 95%CI 1.397-5.391, P=0.003) and visceral<br />

involved (37.9% v 14.8%, OR 3.511, 95%CI 1.548-7.961, P=0.002),<br />

respectively. While serum HER2 ECD levels were not related to age,<br />

BMI, tumor size, lymph node involvement and hormone receptors<br />

status, respectively (P>0.05).<br />

Conclusions: Monitoring the circulating levels of the HER2 ECD in<br />

patients with metastatic breast cancer provides a real-time assessment<br />

of tumor burden and indicates poor prognosis, and may provide<br />

important information for reassessment of HER2 in HER2-negative<br />

metastatic breast cancer.<br />

www.aacrjournals.org 227s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P2-05-06<br />

Quantitative measurement of HER2 expression in breast cancers:<br />

comparison with “real world” HER2 testing in a multi-center<br />

Collaborative Biomarker Study (CBS) and correlation with<br />

clinicopathological features<br />

Yardley DA, Kaufman PA, Adams JW, Krekow L, Savin M, Lawler WE,<br />

Zrada S, Starr A, Einhorn H, Schwartzberg LS, Huang W, Weidler<br />

J, Lie Y, Paquet A, Haddad M, Anderson S, Brigino M, Bosserman<br />

L. Sarah Cannon Research Institute, Nashville, TN; Tennessee<br />

Oncology PLLC, Nashville, TN; Dartmouth Hitchcock Medical<br />

Center, Lebanon, NH; Arlington <strong>Cancer</strong> Center, Arlington, TX; Texas<br />

Oncology Bedford, Bedford, TX; Texas Oncology at Medical City<br />

Dallas 2, Dallas, TX; St. Jude Heritage Medical Group, Fullerton,<br />

CA; The Center for <strong>Cancer</strong> and Hematologic Disease, Cherry Hill,<br />

NJ; Monroe Medical Associates, Harvey, IL; Swedish American<br />

Regional <strong>Cancer</strong> Center, Rockford, IL; The West Clinic, Memphis,<br />

TN; Monogram Biosciences, Inc., So. <strong>San</strong> Francisco, CA; Center<br />

for Molecular Biology and Pathology, Laboratory Corporation<br />

of America, Inc., Research Triangle Park, NC; Wilshire Oncology<br />

Medical Group, Rancho Cucamonga, CA<br />

Background: Accurate determination of HER2 status is critical<br />

in determining appropriate therapy for breast cancer patients. The<br />

HERmark ® assay is a novel method to quantitatively measure HER2<br />

total protein expression (H2T) in breast cancer. In this study, we<br />

compared HERmark H2T with central laboratory HER2 retesting<br />

and local (site reported) HER2 testing of formalin-fixed, paraffinembedded<br />

(FFPE) breast cancer tissues. The quantitative total HER2<br />

measurements (H2T) by HERmark and results of local HER2 tests<br />

were correlated with tumor pathohistological characteristics and<br />

overall survival of breast cancer patients.<br />

Methods: 232 FFPE breast cancer tissues were provided by 11 CBS<br />

study sites for HER2 testing by the HERmark assay and central<br />

laboratory IHC re-testing performed in blinded fashion. Local HER2<br />

immunohistochemistry and/or fluorescence in situ hybridization<br />

(FISH) results and valid HERmark H2T and central HER2 IHC results<br />

were obtained in 192 cases for analysis.<br />

Results: H2T showed a significant correlation with central HER2 IHC<br />

staining intensity (P


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 2<br />

trastuzumab may convert HER2-positive (HER+) primary breast<br />

tumors to HER2 negative (HER-) after neoadjuvant chemotherapy.<br />

We compared the HER2 status in breast cancer patients before and<br />

after neoadjuvant treatment by using dual-color in situ hybridization<br />

(DISH).<br />

Methods<br />

We retrospectively identified 46 patients from the <strong>Breast</strong> <strong>Cancer</strong><br />

database at Tokai University Hospital, in whom HER2-positive<br />

primary breast cancer was diagnosed between 2000 and 2010, and<br />

who were treated with neoadjuvant chemotherapy or endocrine<br />

therapy with or without trastuzumab. Surgical specimens from<br />

patients achieving less than pCR were assessed to determine if<br />

there was enough residual tissue to evaluate the post-treatment<br />

HER2 status by DISH. HER2 status was defined as positive if DISH<br />

demonstrated a gene copy ratio of HER2:CEP17 >2.0. Paraffin<br />

tissue sections (4-µm thick) were mounted on glass slides (New<br />

Sliane III, Catalog No. 5126-25; MUTO PURE CHEMICALS, CO.<br />

LTD., Tokyo, Japan) and stained using the newly developed, fully<br />

automated HER2 Dual ISH assay on a BenchMark@ XT slide stainer<br />

according to the recommended procedure (Ventana Medical Systems<br />

Inc., Tucson, AZ). Thereafter, the stained slides were rinsed with<br />

tap water containing neutral detergent and then rinsed again with<br />

distilled water. The slides were dried at room temperature for at least<br />

60 min and cover-slipped with cover grass for SGC (MUTO PURE<br />

CHEMICALS). HER2 Dual ISH and H&E images were obtained<br />

using an Olympus BX51 microscope.<br />

Results<br />

A pCR was achieved in 9 of the 46 patients (19.6%). Specimens from<br />

core needle biopsy were not sufficient to assess the pretreatment<br />

HER2 status in 3 patients. In 9 patients, the post-treatment HER2<br />

status could not be assessed because residual tumors were DCIS only<br />

in 4 patients and there were less than 20 invasive cells in 5 patients.<br />

Residual tumor was sufficient to assess the post-treatment HER2<br />

status in 25 patients. In pretreatment specimens, of the 25 patients<br />

identified as HER2 + by immunohistochemical analysis, 3 patients<br />

were identified as HER2- by DISH. No post-treatment specimens<br />

were found to be HER2- by DISH among the tumors identified as<br />

HER2+ by DISH at pretreatment. Among the 3 pretreatment tumors<br />

identified as HER2- by DISH, 1 tumor was found to be HER2+ by<br />

DISH at post-treatment, and 2 showed a stable HER2 status.<br />

Conclusion<br />

DISH revealed a stable HER2 status in pretreatment breast tumors<br />

and in residual tumors. However, we found a discrepancy in the<br />

HER2 status between the immunohistochemical analysis and DISH.<br />

P2-05-09<br />

Identification and validation of a miRNA signature associated<br />

with breast cancer progression<br />

Waters PS, Dwyer RM, Kerin MJ. National University of Ireland,<br />

Galway, Ireland<br />

Introduction: MiRNAs are aberrantly expressed in both the<br />

circulation and tissue of patients with breast tumours, however little<br />

is currently known about the relationship between circulating and<br />

tissue miRNAs. The aim of this study was to quantify circulating<br />

and tumour tissue miRNA expression in a murine model of breast<br />

cancer and identify novel circulating markers of disease progression.<br />

Materials and Methods: Athymic nude mice (n=20) received<br />

either a mammary fat pad (n=8, MFP), or subcutaneous (n=7, SC)<br />

injection of MDA-MB-231 cells. Controls received no tumour cells<br />

(n=5). Tumour volume was monitored weekly and blood sampling<br />

performed at weeks 1, 3 and 6 following tumour induction (total<br />

n=60). Animals were sacrificed at week 6 and tumour tissue (n=15),<br />

lungs (n=20) and enlarged lymph nodes (n=3) harvested. RNA were<br />

extracted from all samples (n=98) and relative expression of miRNAs<br />

previously associated with tumour progression were quantified using<br />

RQ-RCR. A microRNA array was also preformed comparing animals<br />

with early (weeks 1, n=5) versus late (week 6, n=5) disease and results<br />

were analysed using RQ-PCR in all blood samples (n=60). Reverse<br />

transcription for the relevant targets and endogenous controls was<br />

carried out and expression was quantified using RQ-PCR.<br />

Results: MiR-10b expression was significantly higher in MFP<br />

compared to SC tumours (p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

lesions in bone, liver, lung and, to a lesser degree, lymph nodes. The<br />

50 mg dose resulted in approximately 75% decrease in liver uptake<br />

of 64 Cu and improved the visualization of hepatic metastasis. We have<br />

initiated the second phase of the study utilizing the pre-administration<br />

of 50 mg trastuzumab in women with HER2 1+ and 2 + disease. To<br />

date we have imaged one patient.<br />

Conclusion: 64 Cu-DOTA-trastuzumab PET imaging is feasible and<br />

image quality is improved with the pre-administration of 50 mg<br />

trastuzumab. Enrollment of women with IHC 1+ and 2+ disease<br />

continues. This work was supported by the Department of Defense<br />

grant # 1024511.<br />

P2-05-11<br />

Proteomic identification and in-silico verification of subtypespecific<br />

signatures in breast cancer.<br />

Pavlou MP, Dimitromanolakis A, Diamandis EP. University of<br />

Toronto, ON, Canada; Mount Sinai Hospital, Toronto, ON, Canada;<br />

University Health Network, Toronto, ON, Canada<br />

<strong>Breast</strong> cancer is a highly heterogeneous disease with different<br />

subtypes presenting distinct clinical characteristics. During the last<br />

decade has been shown that molecular classification of breast cancer<br />

holds the promise for the development of novel prognostic tools and<br />

the identification of novel treatment targets. However, proteins are the<br />

main mediators of biological processes and the molecular targets of<br />

the majority of drugs. Moreover, the proteome integrates the cellular<br />

genetic information and environmental influences. Hence coupling<br />

proteomic and transcriptomic studies may augment the development<br />

of targeted therapies and the identification of disease-relevant proteins<br />

networks.<br />

Here, we report an extensive mass spectrometry-based (MS)<br />

secretome analysis of eight breast cancer cell lines representing,<br />

the three main breast cancer subtypes (luminal, basal and HER2-<br />

neu amplified) in search of subtype-specific proteomic signatures.<br />

Following a bottom-up proteomic approach coupled to tandem mass<br />

spectrometry the conditioned media of 8 breast cancer cell lines were<br />

analyzed. More than 5,200 non-redundant proteins were identified in<br />

the conditioned media of the 8 cell lines with a false positive rate less<br />

than 1%. The mass spectrometry-based identification of 5 proteins<br />

was successfully verified by ELISA.<br />

For the generation of subtype-specific proteomic signatures, proteins<br />

common to all cell lines of the same subtype but not present in the<br />

other two subtypes were identified. Twenty-three, four and four<br />

proteins were found uniquely in basal, HER2-neu amplified and<br />

luminal breast cancer cells. Notably, ERBB2 was one of the proteins<br />

uniquely identified in the HER2-neu amplified subtype verifying the<br />

validity of our approach.<br />

Given that most of these proteins have no quantitative methods<br />

available at present, we opted to preliminarily examine the<br />

relationship of these candidates with breast cancer subtypes by using<br />

an in silico approach, based on transcriptomic data. We performed an<br />

in silico mRNA expression analysis using publicly available data from<br />

four independent experiments containing a total of 1039 patients with<br />

primary breast cancer. The expression profiles of 15 of 31 proteins<br />

investigated showed significant correlation with estrogen receptor<br />

expression and power to distinguish subtypes. The development<br />

of multiplex quantitative mass spectrometry-based assays for the<br />

quantification of the proteomic signatures in relevant biological<br />

specimens along with immunohistochemical studies for two of the<br />

31 candidates are currently ongoing.<br />

The significance of these subtype-specific proteins in breast cancer<br />

prognosis and disease progression warrants further investigation.<br />

P2-05-12<br />

Effects of de-escalated bisphosphonate therapy on bone turnover<br />

or metastasis markers and their correlation with risk of skeletal<br />

related events – A biomarker analysis in conjunction with the<br />

REFORM study.<br />

Addison CL, Zhao H, Mazzarello S, Mallick R, Amir E, Tannock<br />

I, Clemons M. Ottawa Hospital Research Institute, Ottawa, ON,<br />

Canada; Princess Margaret Hospital, Toronto, ON, Canada; The<br />

Ottawa Hospital <strong>Cancer</strong> Centre, Ottawa, ON, Canada<br />

Background: Despite variability in an individual’s risk of skeletal<br />

related events (SREs) from bone metastases (BM), all patients<br />

are treated using a similar dose and schedule (q3-4 wk) of IV<br />

bisphosphonate (BP). The REFORM trial (Amir et al., Am J Clin<br />

Oncol, in press) was a pilot randomised study evaluating the efficacy<br />

of de-escalated (q12 wk) versus standard (q3-4 wk) pamidronate<br />

in maintaining C-telopeptide (CTx) levels in the low risk range<br />

(


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 2<br />

P2-05-13<br />

Correlation of conventional versus experimental biomarkers of<br />

bone turnover and metastasis behaviour with skeletal related<br />

events – A biomarker analysis in conjunction with the TRIUMPH<br />

study.<br />

Addison CL, Kuchuk I, Bouganim N, Zhao H, Mazzarello S,<br />

Vandermeer L, Mallick R, Goss GD, Clemons M. Ottawa Hospital<br />

Research Institute, Ottawa, ON, Canada; The Ottawa Hospital<br />

<strong>Cancer</strong> Centre, Ottawa, ON, Canada; McGill University Health<br />

Centre, Montreal, QC, Canada<br />

BACKGROUND: Despite considerable variability in patient (pt) risk<br />

of skeletal related events (SREs) from bone metastases (BM), all pts<br />

are treated using a one size fits all approach, namely the same dose and<br />

dosing schedule (q3-4 wk) of IV bisphosphonate (BP). Identification<br />

of novel markers of individual SRE risk are thus required to better<br />

tailor treatment. TRIUMPH is an ongoing clinical trial evaluating<br />

q12 wk IV BP therapy for 1 year, following >3 months of standard<br />

q3-4 wk BP, in women with low risk bone metastases [defined by<br />

the bone resorption marker C-telopeptide (CTx) levels 600 ng/ml or SRE) or of SRE alone using<br />

logistic regression analysis.<br />

RESULTS: Although baseline CTx and NTx were elevated in pts who<br />

went on to develop SREs, this did not reach statistical significance.<br />

Baseline activinA trended towards total number of prior SREs<br />

(p=0.07). Baseline TGF-β correlated with duration of BM (p=0.004).<br />

Change in activinA (baseline to week 6) was the only biomarker that<br />

trended to predict coming off study early (p=0.043). Results of other<br />

baseline biomarkers and changes in biomarkers from baseline to wk<br />

12 will also be presented.<br />

CONCLUSIONS: This study further questions the role of CTx and<br />

NTx for driving treatment decisions around de-intensification of BP<br />

therapy (Coleman et al. J Clin Oncol 2012, suppl; abstr 511), and<br />

highlights the need for novel markers of SRE risk. Baseline levels of<br />

activinA was associated with the incidence of SREs in patients with<br />

BM and changes in levels from baseline to 6 weeks correlated with<br />

coming off study early. These findings warrant future studies in breast<br />

cancer pts assessing activinA as a predictor of SRE risk associated<br />

with breast cancer bone metastases.<br />

This study was supported by grants from the Ontario Institute for<br />

<strong>Cancer</strong> Research with funding from the Government of Ontario, and<br />

from the Ontario Chapter of the Canadian <strong>Breast</strong> <strong>Cancer</strong> Foundation.<br />

P2-05-14<br />

Clinical significance of Lysil oxidase-like 2 (LOXL 2) in breast<br />

cancer<br />

Ahn SG, Lee HM, Hwang SH, Lee SA, Jeong J, Lee H-E. Gangnam<br />

Severance Hospital, Yonsei University Medical College, Seoul,<br />

Republic of Korea<br />

Introduction<br />

Several preclinical studies provided relevant evidences that lysine<br />

oxidase-like 2 (LOXL2) is associated with invasiveness and<br />

metastasis in breast cancer. To investigate the clinical significance<br />

of LOXL2, we performed survival analysis with LOXL-2 in breast<br />

cancer patients.<br />

Method<br />

A cohort treated at Gangnam Severance hospital, Seoul, Korea<br />

between January 1996 and December 2004 was studied. The<br />

immunohistochemical analyses with LOXL-2 antibody were<br />

performed in the 309 patients. The patients was stratified into two<br />

groups based on the expression level of LOXL-2. Kaplan-Meier and<br />

Cox’s methods were used to define prognostic factors associated with<br />

breast cancer survival.<br />

Results<br />

LOXL-2 positive group was 50 patients (16.2%) and 259 patients<br />

(83.8%) are LOXL-2 negative group. The proportion of estrogen<br />

receptor-negative patients was significantly higher in LOXL-2<br />

positive group (54.0 vs. 37.0 %, P =0.040). The Kaplan-Meier overall<br />

survival estimate at 5 year for the breast cancer patients with negative<br />

LOXL-2 was higher than that for the patients with positive LOXL-2<br />

(88.0 vs. 71.8 %, P = 0.008). In the multivariate analysis for overall<br />

survival, lymph node metastasis (Hazard Ratio (HR) 0.32, 95%<br />

confidence interval (CI) 0.17-0.61) and positive LOXL-2 (HR 0.53,<br />

95% CI 0.29-0.97) were significantly associated. In the multivariate<br />

analysis for distant metastasis-free survival, age younger than 35 years<br />

(HR 0.52, 95% CI 0.30-0.91), lymph node metastasis (HR 0.29, 95%<br />

CI 0.16-0.54), high histologic grade (HR 0.54, 95% CI 0.32-0.89) and<br />

positive LOXL-2 (HR 0.55, 95% CI 0.31-0.97) were demonstrated<br />

as significant prognostic factors.<br />

Conclusion<br />

In breast cancer patients, overexpression of LOXL-2 is a poor<br />

prognostic factor for overall survival and distant metastasis-free<br />

survival.<br />

P2-05-15<br />

Assessment of Notch Signaling Pathway Components as<br />

Biomarkers for Triple Negative <strong>Breast</strong> <strong>Cancer</strong>: Comparison of<br />

Triple Negative <strong>Breast</strong> <strong>Cancer</strong> Cell Lines and Human <strong>Breast</strong><br />

<strong>Cancer</strong> Samples.<br />

Zlobin A, Olsauskas-Kuprys R, Hodge S, O’Toole M, Ersahin C,<br />

Osipo C. Loyola University Chicago, Cardinal Bernardin <strong>Cancer</strong><br />

Center, Maywood, IL<br />

Background: Treatment options for patients presenting with TNBC<br />

are limited, primarily because there is no approved targeted therapy<br />

available. Canonical breast cancer targets such as estrogen receptor<br />

and HER2/neu proteins are absent in TNBC. Furthermore, there is an<br />

urgent need to uncover biomarkers in this disease in light of its costly<br />

treatment as compared to non-TNBC treatment and, more importantly,<br />

its poor prognosis as evidenced by aggressive tumor metastasis and<br />

high patient mortality. While Notch-1 target gene expression has been<br />

reported to be linked to various types of malignancies, we propose that<br />

since Notch signaling pathway is involved in cancer cell proliferation<br />

and survival it could be a likely oncogenic driver and possible target<br />

in certain TNBC patients.<br />

www.aacrjournals.org 231s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Methods: Human TNBC cell lines BT-549, MDA-MB-231, and<br />

MDA-MB-468 were used to measure endogenous relative mRNA<br />

transcript levels of Notch genes and gene targets by means of real time<br />

PCR. In addition, the RNA from formalin-fixed, paraffin-embedded<br />

specimen from women-diagnosed with TNBC who had undergone<br />

breast surgery was also analyzed for comparable gene expression for<br />

similar targets following Laser capture micro-dissection.<br />

Results: The in vitro results show that mRNA transcripts of Notch-4,<br />

Deltex-1, Hes-1 and -Hes5, as well as upstream Notch regulator<br />

PEA3 targets IL-8 and MMP-9 were increased in MDA-MB-468<br />

and BT-549 cells as compared to MDA-MB-231 cells (n=3-5). In the<br />

clinical sample specimens, Notch-1, Jagged-1, and Hes-5 genes were<br />

up-regulated, while the Notch-4 gene was down-regulated in TNBC<br />

patients as compared to normal control specimens from reduction<br />

mammoplasty (n=4).<br />

Conclusions: Our data present the heterogeneity of TNBC disease.<br />

They also indicate that the Notch signaling pathway is up-regulated<br />

in certain TNBC cell lines and human TNBC breast cancer tissue.<br />

Lastly, these results suggest that some components of Notch signaling<br />

may be viable biomarkers to better predict Notch activity for future<br />

therapeutic interventions using Notch inhibitors.<br />

P2-05-16<br />

Expression of β-III tubulin, foxo 3 protein and deoxythymidine<br />

kinase in breast cancer patients receiving neoadjuvant<br />

chemotherapy<br />

Chow LWC, Loo WTY, Yip AYS, Ong EYY, Ng W-K. UNIMED Medical<br />

Institute, Hong Kong; Organisation for Oncology and Translational<br />

Research, Hong Kong; Central Hospital, Hong Kong<br />

Background:<br />

β-III tubulin is a major protein involved in cell division during<br />

metaphase by forming microtubules. Chemotherapeutic drugs target<br />

the dynamics of cytoskeleton, blocks mitosis and cell proliferation to<br />

induce apoptosis. FoxO1, FoxO3, FoxO4, and FoxO6 transcriptional<br />

factors suppress different types of human tumors. The transcriptional<br />

up-regulation of FoxO3a might be induced by chemo-drugs treatment<br />

and enhance apoptosis. Deoxythymidine kinase (DTYMK) is a<br />

key enzyme involved in DNA replication. Its expression level was<br />

identified as an independent prognostic marker and indicator of<br />

tumor burden. This study was to investigate the expression of β-III<br />

Tubulin, FOXO3 and DTYMK in breast cancer patients receiving<br />

neoadjuvant chemotherapy.<br />

Method:<br />

The expression of β-III tubulin, FOXO3 and DTYMK was evaluated<br />

by immunohistochemical assay (IHC) on 103 pre-treatment and 81<br />

post-treatment tumor samples from patients with locally advanced<br />

breast cancer. Samples were taken before and after 4 cycles of<br />

anthracycline-based neoadjuvant chemotherapy (5-fluorouracil,<br />

epirubicin, and cyclophosphamide). The expression of β-III tubulin,<br />

FOXO3 and DTYMK in tumor samples was measured in terms of<br />

percentage of cancer cells. T-test was applied to analyze the statistical<br />

significance of protein expression.<br />

Results:<br />

Median age of patients was 46 (range: 22-66) and the mean tumor<br />

size was 4.47±1.51 cm. Among 103 patients, 95 (92.2%), 5 (4.9%)<br />

and 2 (1.9%) patients had infiltrating ductal, lobular and mucinous<br />

carcinoma respectively, whereas 1 (1%) patient had unknown type<br />

of invasive tumor. At diagnosis, breast tumors were found to express<br />

β-III tubulin and DTYMK with a prevalence of 28.9% and 50%<br />

while the expression decreased to 11.1% and 25% after neoadjuvant<br />

treatment respectively. The expression of FOXO3 was 25% before<br />

treatment and increased to 75% post-treatment (p < 0.05). The change<br />

in expression before and after treatment was statistically significant.<br />

Conclusions:<br />

Comparison of the expression of β-III Tubulin, FOXO3 and DTYMK<br />

in breast cancer patients receiving neoadjuvant chemotherapy may<br />

serve as clinical assessing markers to predict the treatment outcome<br />

and prognosis.<br />

P2-05-17<br />

Correlation between cyclin D1, estrogen and progesterone<br />

receptors in invasive breast cancer after short-term treatment<br />

with tamoxifen or anastrozole<br />

Millen EC, Mattar A, Logullo AF, Nonogaki S, Soares FA, Gebrim<br />

LH. Federal University of Sao Paulo, Sao Paulo, SP, Brazil; Perola<br />

Byington Hospital, Sao Paulo, SP, Brazil; Federal University of Sao<br />

Paulo, Sao Paulo, SP; Arnaldo Vieira de Carvalho, Sao Paulo, SP,<br />

Brazil<br />

INTRODUCTION: Hormone therapy is associated with reduced<br />

breast cancer mortality. The use of biomarkers that are predictive of<br />

early cellular responses has been explored as a predictor of hormone<br />

resistance. Estrogen and progesterone receptor positivity and cyclin<br />

D1 have been associated with resistance to treatment with tamoxifen.<br />

OBJECTIVES: The aim of this study was to investigate the variations<br />

in the levels of cyclin D1 and the estrogen and progesterone receptors<br />

(ER and PR, respectively) in postmenopausal patients with ER- or<br />

PR-positive breast cancer after short-term treatment (26 days) with<br />

tamoxifen, anastrozole or a placebo.<br />

METHODS: This was a prospective, randomized double-blind study<br />

conducted in 71 patients with infiltrating ductal carcinoma (stages<br />

II and III) receiving care at either Pérola Byington Hospital or São<br />

Paulo Hospital (São Paulo, Brazil). The patients were divided into<br />

three groups based on their treatment during the preoperative period<br />

(26 days): P (placebo, N = 26), T (tamoxifen, 20 mg/day, N = 22)<br />

and A (anastrozole, 1 mg/day, N = 23). The biopsies were performed<br />

at diagnosis and after mastectomy (26th day), and the tumors were<br />

isolated by tissue microarray. Immunohistochemistry was performed<br />

using anti-cyclin D1 (Novocastra DCS-6), anti-ER (Dako-M7047)<br />

and anti-PR (Dako-M3569) antibodies. The semiquantitative analyses<br />

were performed using Allred criteria, and the statistical analyses were<br />

performed with the ANOVA parametric test (p ≤ 0.05).<br />

RESULTS: A reduction in the mean PR level from 4.22 (pre-treatment)<br />

to 1.94 (post-treatment) was observed only in patients treated with<br />

anastrozole (p = 0.01). A positive linear correlation between cyclin<br />

D1 and PR levels was observed in group A (p = 0.0001), whereas<br />

group T exhibited a negative correlation (p = 0.0001). No correlation<br />

was observed in group P (p = 0.35)*.<br />

CONCLUSION: PR and cyclin D1 are likely predictive of an early<br />

response to aromatase inhibitors in breast cancer.<br />

P2-05-18<br />

Withdrawn by Author<br />

P2-05-19<br />

Pathway-based analysis of breast cancer<br />

Song D, Cui M, Fan Z, Yang Y, Xue L, Zhang DY, Ye F. The First<br />

Hospital of Jilin University, Changchun, Jilin, China; The Mount<br />

Sinai Medical Center, New York, NY; Nanfang Hospital Southern<br />

Medical University, Guangzhou, China<br />

Objective: Although HER2 and ER pathways are predominant<br />

pathways altered in breast cancer, it is now well accepted that many<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

232s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 2<br />

other signaling pathways are also involved in the pathogenesis<br />

of breast cancer, especially in triple negative breast cancer. The<br />

understanding of these additional pathways may assist in identifying<br />

new therapeutic approaches for breast cancer.<br />

Methods: 13 invasive ductal carcinoma tissues and 5 benign breast<br />

tissues were analyzed for the mRNA expression level of 1243<br />

cancer pathway-related genes using SmartChip (WaferGen, CA),<br />

a real-time PCR-base method. In addition, the levels of 154 cancer<br />

pathway-related proteins and phosphoproteins were measured using<br />

our innovative Protein Pathway Array.<br />

Results: Out of 1,243 mRNAs, 68.7% were detected in breast cancer<br />

and 73 mRNAs were statistically significant between benign and<br />

cancer tissues. Of these mRNAs, 105 only expressed in breast cancer<br />

tissues and 33 mRNAs only expressed in normal breast tissues. Out<br />

of 154 proteins and phosphoproteins, 39% were detected in cancer<br />

tissues and 50 proteins were significantly differentiated between tumor<br />

and normal tissues. Interestingly, only 3 genes (CDK6, Vimentin and<br />

SLUG) showed decrease of both protein and mRNA. Six proteins<br />

(BCL6, CCNE1, PCNA, PDK1, SRC and XIAP) showed differentially<br />

expressed between tumor and normal tissues but no differences were<br />

observed at mRNA levels. Analyses of mRNA and protein data using<br />

Ingenuity Pathway Analysis showed more 15 pathways were altered<br />

in breast cancers and 6 of them shared between mRNAs and proteins,<br />

including p53, IL17, HGF, NGF, PTEN and PI3K/AKT.<br />

Conclusion: There is a broad dysregulation of various pathways in<br />

breast cancer both at protein levels and mRNA levels. It is important<br />

to note that mRNA expression does not correlate with protein level,<br />

suggesting that different regulation mechanisms between proteins<br />

and mRNAs.<br />

P2-05-20<br />

Validation and comparison of CS-IHC4 score with a nomogram<br />

based on Ki67 index, Adjuvant! Online, and St. Gallen risk<br />

stratification to predict recurrence in early Hormone Receptor<br />

(HR)-positive breast cancers<br />

Park YH, Im S-A, Cho EY, Ahn JH, Woo SY, Kim S, Keam B, Lee JE,<br />

Han W, Nam SJ, Park IA, Noh D-Y, Yang JH, Ahn JS, Im Y-H. Samsung<br />

Medical Center; Seoul National University College of Medicine<br />

Background: Recently, the information in the IHC score was reported<br />

to be similar to that in the 21-gene Genomic Health recurrence score<br />

(GHI-RS). The aim of this study is to develop a nomogram based<br />

on Ki67 index to predict recurrence and to validate the nomogram<br />

by comparison with CS-IHC4 as well as Adjuvant! Online and St.<br />

Galen risk stratification. In addition, we validated our nomogram<br />

with external cohort.<br />

Methods: We retrospectively analyzed the clinicopathologic<br />

characteristics and outcomes of 1,070 postoperative HR-positive<br />

breast cancer patients between 2004 and 2007 at the Samsung<br />

Medical Center to determine recurrence-free survival (RFS). We<br />

constructed nomogram using Cox proportional hazard model and<br />

validated externally in a cohort of 1,028 at Seoul National University<br />

Hospital. A prognostic model that used classical variables, Adjuvant!<br />

Online, St. Gallen risk stratification, and the four IHC markers (IHC4<br />

score) were created and assessed in our cohort by LR-χ 2 test using<br />

the bootstrapping method.<br />

Results: Nomogram showed an area under the receiver operating<br />

characteristic curve (AUC) of 0.70 (95% CI, 0.62-0.75) in the training<br />

set. The validation set showed a good discrimination with an AUC<br />

of 0.65 (95% CI, 0.58-0.72). In LR-χ 2 test, the nomogram score was<br />

found to be more informative than the IHC4 with CS (LR-χ 2 4.0539<br />

[df1], 95% CI; 0.1038-8.004 for CS-IHC4 + nomogram score vs.<br />

CS-IHC4).<br />

Average change in LR-χ² value and 95% Bootstrap CIs based on 1000 re-samplings for assessing<br />

the amount the amount of information added by the nomogram score or Adjuvant! Online or St.<br />

Galen risk stratification to the CS-IHC4 to a model containing the nomogram<br />

Model df LR-χ² 95% CI<br />

CS-IHC4 + nomogram score vs CS-IHC4 1 4.0539 0.1038 8.004<br />

CS-IHC4 + Adjuvant! Online vs CS-IHC4 1 0.2732 -4.1697 4.7162<br />

CS-IHC4 + St.Gallen vs Cs-IHC4 2 1.7558 -1.7859 5.2975<br />

RFS: Recurrence free survival, 95% CI: 95% confidence interval<br />

Prognostic significance was more prominent in N1 diseases than in the<br />

others (LR-χ 2 4.199, 95% CI; 1.496-6.902 for CS-IHC4 + nomogram<br />

score vs. CS-IHC4).<br />

Average change in LR-χ² value and 95% Bootstrap CIs based on 1000 re-samplings for assessing<br />

the amount the amount of information added by the nomogram score or Adjuvant! Online to the<br />

CS-IHC4 to a model containing the nomogram according to the nodal status (N0-3)<br />

CS-IHC4 + CS-IHC4 +<br />

Model<br />

Nomogram vs CS- Adjuvant! Online<br />

IHC4<br />

vs CS-IHC4<br />

CS-IHC4 + St.<br />

Gallen vs CS-IHC4<br />

N0 LR-χ² 0.442 0.272 0.566<br />

95% CI 0.109-0.776 -0.858-1.402 0.104-1.028<br />

N1 LR-χ² 4.199 1.212<br />

95% CI 1.496-6.902 -0.538-2.962<br />

N2 LR-χ² 0.659 1.573<br />

95% CI -0.131-1.448 1.435-1.710<br />

N3 LR-χ² 0.003 0.197<br />

95% CI -0.975-0.982 -0.439-0.832<br />

RFS: Recurrence free survival, 95% CI: 95% confidence interval<br />

However, Adjuvant! Online and St. Galen risk stratification did not<br />

show any definitive additional prognostic value.<br />

Conclusions: We developed and validated a nomogram based on Ki67<br />

index in external patients’ cohort. It was compared with CS-IHC4 in<br />

our patients’ cohort in early HR-positive breast cancers. This study<br />

implicates the amount of prognostic information contained in the<br />

nomogram is superior to that in the CS-IHC4 score.<br />

P2-05-21<br />

Molecular subtypes of invasive breast cancers show differential<br />

expression of the proliferation marker Aurora Kinase A (AURKA)<br />

Kovatich AJ, Luo C, Chen Y, Hooke JA, Kvecher L, Rui H, Shriver<br />

CD, Mural RJ, Hu H. Windber Research Institute, Windber, PA;<br />

Walter Reed National Military Medical Center, Bethesda, MD; MDR,<br />

Global Systems LLC, Windber, PA; Thomas Jefferson University,<br />

Philadelphia, PA<br />

Background: Invasive breast cancer (IBC) has been classified into<br />

four major subtypes based on gene expression profiling. The luminal<br />

A subtype (LA) has the best prognosis, when compared to luminal<br />

B (LB), HER2+, and basal-like (Basal). Ki67 by gene expression or<br />

immunohistochemistry (IHC) is commonly used as a proliferation<br />

index. The function of Ki67 in proliferation remains unknown.<br />

AURKA (STK15) is known to play an important role in mitosis, and<br />

is a component of the 21-gene recurrence score of the Oncotype Dx.<br />

With multiple platforms of molecular data available from hundreds of<br />

IBC tissues in The <strong>Cancer</strong> Genome Atlas project (TCGA), we sought<br />

to study the association of AURKA with different IBC subtypes and<br />

explore its use as a proliferation marker in IBCs.<br />

Methods: Gene expression (Agilent, log2 transformed), relative<br />

DNA copy number (CN, Affymetrix SNP 6.0), and exome sequence<br />

mutation (Illumina) data for 459 IBC cases were downloaded from the<br />

TCGA data portal. PAM50 classification results of all samples were<br />

obtained from the TCGA breast cancer AWG group and included 203<br />

LA, 113 LB, 51 HER2+, 84 Basal-like, and 8 Normal-like which were<br />

not used in this study due to the low numbers. Kruskal-Wallis tests<br />

were used to evaluate the differences among four subtypes on AURKA<br />

expression and CN, followed by Wilcoxon Mann-Whitney test with<br />

Bonferroni adjustment for pairwise analyses. Pearson’s Correlation<br />

Coefficient was used for correlation analyses. All statistical analyses<br />

were performed using SAS and R, and two-sided, p values


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

mRNA levels were significantly lower in LA (mean±SD, -2.61±0.63)<br />

compared to LB (-1.45±0.78), HER2+ (-1.38±0.61), and Basal<br />

(-1.26±0.62) subtypes (p values all < 0.0001). No significant<br />

difference was detected between other subtype pairs. In CN analysis,<br />

Basal (0.09±0.22) was lower than HER2+ (0.32±0.308, p


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 2<br />

To assess significance of the CLS, we found three primary/metastatic<br />

clonal pairs in the TCGA to serve as positive controls. To serve as<br />

negative controls, from 357 ER+ and 46 TN primary TCGA tumors,<br />

we formed a total of 16,422 independent ER+/TN pairs. For the 3<br />

clonal TCGA pairs, the CLS values were 39.3%, 58.5% and 60.0%.<br />

Most of the independent TCGA pairs had a CLS of zero (98.5%),<br />

with a maximum CLS of 2.8%. As the CLS for Case 1 lies above<br />

maximum observed CLS among 16,422 independent tumor pairs, we<br />

reject the hypothesis that this tumor pair is independent, at p80% of SNVs, consistent with clonal evolution of metastasis. The<br />

two tumors from Case 2 have few shared SNVs, consistent with<br />

independent origin. CNA results were consistent. This is the first<br />

clonality analysis reported from deep sequencing of phenotypically<br />

discordant synchronous bilateral breast cancers, and demonstrates that<br />

next-generation sequencing can distinguish clonal from independent<br />

tumor pairs with high confidence.<br />

Funding: The <strong>Breast</strong> <strong>Cancer</strong> Research Foundation<br />

P2-06-02<br />

Genomic evolution from primary breast cancers to distant<br />

metastases<br />

Hoefnagel LDC, Moelans CB, van der Groep P, van der Wall E, van<br />

Diest PJ. University Medical Center Utrecht, Netherlands<br />

Purpose: Genomic evolution is an accepted concept during cancer<br />

development and progression. However, little is known about the<br />

changes that occur during the metastatic process despite the fact<br />

that breast cancer metastases are the leading cause of death in breast<br />

cancer patients. The purpose of this study was therefore to investigate<br />

whether distant breast cancer metastases show progression in copy<br />

number changes of onco- and tumor suppressor genes compared to<br />

their primary tumor.<br />

Material and methods: Multiplex ligation-dependent probe<br />

amplification (MLPA) was used to compare copy number of 21<br />

established oncogenes and tumor suppressor genes between primary<br />

breast cancer samples and corresponding distant metastases in 55<br />

patients. Genes studied were ESR1, EGFR, FGFR1, ADAM 9, IKBKB,<br />

PRDM14, MTDH, MYC, CCND1, EMSY, CDH1, TRAF4, CPD,<br />

MED1, HER2, CDC6, TOP2A, MAPT, BIRC5, CCNE1 and AURKA.<br />

Results: Overall, there was no significant difference in mean copy<br />

number between primary tumors (1.26 +/- 0.6) and metastases (1.27<br />

+/- 0.64) (p=0.826), but on the individual gene level, mean copy<br />

number for PRDM14 (p=0,023), MED1 (p=0.001) and CCNE1<br />

(p=0.039) were significantly higher while TRAF4 (p=0.038) copy<br />

number was significantly lower in the metastases. Using dichotomized<br />

MLPA data, MTDH amplifications were significantly more frequent<br />

in the primary cancers compared to the metastases (27.3% vs.14.6%,<br />

p=0.039), while significantly more CDH1 losses were found in the<br />

metastases compared to the primary tumor (23.6% vs. 9.1%, p=0.039).<br />

Conclusion<br />

Distant breast cancer metastases show genomic evolution from<br />

the primary cancer as reflected by copy number changes in several<br />

established onco- and tumor suppressor genes.<br />

P2-06-03<br />

Specific Transcriptional Response of Four Blockers of Estrogen<br />

Receptors on Estrogen-Modulated Genes in ZR-75-1 <strong>Breast</strong><br />

<strong>Cancer</strong> Xenografts<br />

Calvo E, Luu-The V, Martel C, Labrie F. Laval University Hospital<br />

Research Center (CRCHUL), Quebec City, QC, Canada; Laval<br />

University, Quebec City, QC, Canada; EndoCeutics Inc., Quebec<br />

City, QC, Canada<br />

Introduction: Acolbifene (ACOL) is a novel compound completely<br />

free of estrogen-like activity in both the human and rat mammary<br />

gland and uterus. In previous studies, it was observed that ACOL<br />

could have a cytotoxic or tumoricidal rather only tumorostatic activity,<br />

with a lack of development of tumor resistance. The objective of the<br />

present study was to obtain information on the molecular basis of<br />

the tumoricidal properties of acolbifene, as well as on the identity<br />

of the genes potentially implicated in the resistance to tamoxifen<br />

(TAM). The specificity of action of ACOL was compared to the<br />

pure antiestrogen fulvestrant (FUL) as well as to tamoxifen (TAM)<br />

and raloxifene (RAL). Methods: The gene expression profile of the<br />

ZR-75-1 breast cancer xenografts was studied following treatment<br />

with ACOL, RAL, TAM and FUL. Ovariectomized female nude<br />

mice (10/group) bearing human ZR-75-1 breast cancer xenografts<br />

were supplemented with estrone (E 1 ) (subcutaneous silastic implants)<br />

and were injected daily with the vehicle alone or 50 µg of ACOL,<br />

RAL, TAM or FUL for 6 months. Mice were killed 24h after the last<br />

injection and ZR-75-1 tumors were collected and processed for RNA<br />

extraction and microarray analyses (Affymetrix GeneChip U133 Plus<br />

2.0). Results: Long-term exposure of ZR-75-1 xenografts to E 1 causes<br />

a massive modulation of E 1 -responsive genes. When the antiestrogens<br />

were administered simultaneously with E 1 , some compound-specific<br />

prevention of the effect of E 1 was observed. Globally, the efficacy of<br />

the compounds was ACOL>FUL or RAL>TAM. ACOL significantly<br />

prevented the effect of E 1 on 195 responsive genes. Among these,<br />

the most enriched gene ontological group in Molecular Function was<br />

Receptor Activity, including ER, insulin, retinoid and thrombospondin<br />

receptor activity. One of the transcriptional repressors of the<br />

estrogen signaling pathway, the NKX31, was down-regulated by<br />

E 1 and completely blocked by ACOL (fold-change -1.64 and 2.54,<br />

respectively). The most enriched biological processes were associated<br />

with mammary gland development, positive regulation of glucose<br />

metabolic processes and blood vessel morphogenesis. Between the<br />

genes implicated in cell death/apoptosis of breast cancer cells, the<br />

changes of expression of 57, 51, 48 and 31 genes were significantly<br />

neutralized by ACOL, RAL, FUL and TAM, respectively. Sixteen<br />

genes implicated in tumor resistance to TAM were significantly<br />

blocked by ACOL, including DEGS1, a gene implicated in the<br />

inhibition of EGF receptor biosynthesis. The other 15 genes in the<br />

group were up-regulated by E 1 , changes which were significantly<br />

blocked by ACOL. Conclusion: The present data offer a possible<br />

explanation for the potent tumoricidal action of acolbifene in<br />

human breast cancer xenografts, thus offering a new paradigm in the<br />

hormonal therapy of breast cancer.<br />

www.aacrjournals.org 235s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P2-06-04<br />

Impact of genomic testing on chemotherapy utilization<br />

Riley L, Nakijima H, Balinger K, Anderson D. St. Luke’s University<br />

Health Network, Bethlehem, PA<br />

Genomic testing for breast cancer has become a common practice. The<br />

two main tests at our institution are Oncotype DX ® and MammaPrint<br />

®<br />

. Each test has unique features and criteria. We sought to compare<br />

the practice pattern between these tests and compare them to patients<br />

who did not undergo any genomic testing.<br />

Methods: This is a retrospective review of all breast cancer patients<br />

between May 2008 and August 2011. Data was primarily thorough<br />

our tumor registry database. In some cases individual charts were<br />

also reviewed. Results: We analyzed 1093 breast cancers in 1064<br />

patients. The average age was 62. The stage at diagnosis was 19%<br />

DCIS, 41% stage I, 24% stage II, 11% stage III and 5% stage IV.<br />

Of the 1093 cancers, 70 (6%) had an Oncotype test performed, 180<br />

(16%) had a MammaPrint test done, 4 (0.4%) had both test, and 839<br />

(77%) had no genomic test performed. Patients with an Oncotype test<br />

were more likely to be stage I than those with a MammaPrint (66% vs<br />

48%). Additionally, patients who had a MammaPrint test more often<br />

received chemotherapy when compared to either the Oncotype test<br />

or no genomic testing.<br />

Percent of Patients that Received Chemotherapy<br />

Stage Oncotype Mammaprint Neither<br />

I 4% (2/46) 31% (27/86) 17% (51/306)<br />

II 30% (6/20) 66% (44/66) 55% (93/169)<br />

III 100% (4/4) 79% (15/19) 79% (78/99)<br />

Total 17% (12/70) 49% (89/180) 30% 248/839)<br />

Chemotherapy Utilization<br />

Low Intermediate High<br />

Oncotype 8% (3/38) 29% (6/21) 100% (2/2)<br />

Mammaprint 22% (18/82) NA 74% (64/86)<br />

With a mean follow-up of 1.5 years, there have been 28 recurrences (0<br />

in the Oncotype group, 5 in the MammaPrint group, and 23 in the no<br />

genomic testing group). All five of the MammaPrint recurrences were<br />

high risk and received adjuvant chemotherapy. Of the 23 recurrences<br />

in the no genomic testing group, eight patients were node negative<br />

and could have been considered appropriate for genomic testing, 6<br />

of these eight did not have adjuvant chemotherapy.<br />

Conclusions: There are distinct practice pattern differences between<br />

Oncotype DX ® and MammaPrint ® utilization. MammaPrint<br />

testing was ordered more often in higher staged patients and was<br />

associated with an increased use of chemotherapy. This may relate<br />

to MammaPrint’s previous requirement for fresh tissue that typically<br />

occurs before the final nodal status is known. This difference should<br />

diminish with the recent availability of MammaPrint on FFPE<br />

tissue. All patients who underwent genomic testing and developed a<br />

recurrence had been treated with adjuvant chemotherapy. In contrast,<br />

patients who did not have a genomic test and experienced a recurrence<br />

may have benefited from genomic testing.<br />

P2-06-05<br />

Single-cell RNA sequencing of paclitaxol-treated breast cancer<br />

cell lines to find individual cell response<br />

Vaske CJ, Lee W, Benz SC, <strong>San</strong>born JZ, Emerson BM, Pourmand N,<br />

Lopez Diaz F. Five3 Genomics, LLC, <strong>San</strong>ta Cruz, CA; University of<br />

California, <strong>San</strong>ta Cruz, CA; Salk Institute, La Jolla, CA<br />

<strong>Cancer</strong> treatments act on a population of cells, each of which<br />

may experience different individual responses to treatment. Such<br />

differential response will result in resistance to treatment even if a<br />

majority of cancerous cells are eliminated. To examine differential<br />

cell response, we simultaneously profiled the gene expression and<br />

mutation spectrum of individual cells from the MDAMB231 cell<br />

line using next generation sequencing of isolated RNA. A total of<br />

23 transcriptomes were characterized from paclitaxel-treated and<br />

paclitaxel-surviving cells. We found significant different changes in<br />

mutation rates between paclitaxel treated cells, with a dose-dependent<br />

increase in single nucleotide changes in RNA in paclitaxel-treated<br />

cells. Cells undergoing exposure to paclitaxel also showed higher<br />

pathway activity in SRC, as well as an integrin switch from ITGB1<br />

to ITGB3. In contrast, cells that survived a high dose of paclitaxel<br />

showed an insignificant number of single nucleotide changes,<br />

suggesting that these cells either evaded initial paclitaxel exposure or<br />

were better able to repair the effects of paclitaxel exposure. Despite the<br />

RNA sequence similarity between surviving and untreated cells, there<br />

were changes in gene expression and pathway activities including<br />

higher PI3K activity. Paclitaxel-surviving cells also showed activation<br />

of pathways associated with higher proliferation.<br />

P2-06-06<br />

Integrating multiple molecular profiles with pathways to learn<br />

sub-type specific interactions with PARADIGM<br />

Sedgewick AJ, Vaske CJ, Benz SC. Five3 Genomics, <strong>San</strong>ta Cruz, CA;<br />

University of Pittsburgh, PA<br />

High-dimensional omics profiling provides a detailed molecular view<br />

of individual breast cancers. We extended the Paradigm algorithm,<br />

a pathway analysis method for combining multiple omics data types<br />

with curated interactions on 7132 genes, to learn the strength and<br />

direction of curated interactions. Using only a single dataset, the<br />

568 samples from the TCGA breast cancer cohort, we found strong<br />

high-throughput support for the majority of curated interactions. Gene<br />

set enrichment found that targets of the strongest interactions were<br />

significantly enriched for apoptosis and cell morphogenesis, and that<br />

strong regulators were significantly associated with phosphorylation.<br />

Within the TCGA breast cancer cohort the interactions some of the<br />

strongest support centered around ER alpha, IL23, GREB1, AKT1,<br />

and p53. We also assessed different strength between breast cancer<br />

subtypes, and found interactions associated with the MYC pathway<br />

and the ER alpha network to be among the most differential between<br />

basal and luminal A subtypes. Finally, we extended the method to<br />

assess the contextual dependence of interactions and found significant<br />

correlation between FOXM1 and JUN in regulation of CDK1.<br />

P2-06-07<br />

Exome sequencing identifies somatic mutations in basal-like<br />

breast cancer before and after neoadjuvant chemotherapy<br />

Jiang Y-Z, Yu K-D, Shao Z-M. Shanghai <strong>Cancer</strong> Center, Shanghai,<br />

Shanghai, China<br />

Purpose<br />

Among the five breast cancer subtypes, basal-like breast cancers<br />

(BLBCs) have drawn much attention, because they are associated<br />

with poor clinicopathological features, lack of effective treatment<br />

targets, and have poor prognosis. We designed the experiment to test<br />

the hypothesis that significant mutational evolution of BLBCs might<br />

occur during chemotherapy.<br />

Methods<br />

We performed exome sequencing (100-fold coverage) to examine the<br />

exomes of 11 paired BLBC samples (pretreatment tumor biopsies and<br />

posttreatment tumors). The molecular subtype of BLBC was proven<br />

by gene-expression profile as well as by immunohistochemistry assay.<br />

Firstly, we sequenced the tumor genomic DNA obtained before and<br />

after neoadjuvant chemotherapy and their matched normal breast<br />

tissues. After analyzing and comparing potential somatic single<br />

nucleotide variations (SNVs) and insertions & deletions (indels),<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

236s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 2<br />

we focused on the functional mutations in certain genes or pathways.<br />

Furthermore, after validation of the mutations by <strong>San</strong>ger sequencing,<br />

we investigated their biological functions (e.g. drug-resistance, cell<br />

proliferation, etc.).<br />

Results<br />

Compared with pretreatment biopsies, analysis of whole-exome<br />

sequence data identified and validated TEKT4 (tektin 4) — a<br />

constitutive protein of microtubules in cilia, flagella, basal bodies<br />

and centrioles — as a significantly mutated gene in posttreatment<br />

tumors with a mutation frequency of approximately 17.6% in an<br />

independent extension cohort of 84 paired BLBC samples. TEKT4<br />

mutations are biologically relevant, as ectopic expression of mutant<br />

TEKT4 increased paclitaxel resistance of BLBC cell lines both in vitro<br />

and in vivo. Additionally, compared with the normal breast tissues, 16<br />

significantly mutated genes were identified and validated in the paired<br />

cancer samples, including TP53, ARID1A, BEND5, LCT, COL4A4,<br />

GAP43, COL6A6, LOC728369, STAP1, KCNK17, LZTS2, TST,<br />

IL9R, HDLBP, RASL11A and FAM22D. These genes have functions<br />

that could be broadly grouped into five biological classes: (i) MAPK<br />

signaling, (ii) PI3K/AKT signaling, (iii) cell cycle, (iv) metabolism<br />

and (v) loss of genome stability.<br />

Conclusion<br />

Our data reveal the novel concept that significant evolution might<br />

occur during chemotherapy and mutational heterogeneity could<br />

be a property of BLBCs. In addition, the identification of somatic<br />

mutations as biomarkers for pCR (pathologic complete remission)<br />

prediction and prognosis might help us optimize choice for sequential<br />

therapy and improve patients’ survival. Prospective clinical trials<br />

based on these findings might illustrate the drug-resistant mechanism<br />

and identify resistance pathways that can be exploited for therapeutic<br />

selection.<br />

P2-07-01<br />

BRCA1 regulates RHAMM function in breast cancer<br />

Fleisch MC, Yang Y, Mei Q, Sadat F, Brandi L, Iwaniuk KM,<br />

Honisch E, Maxwell CA, Niederacher D. Heinrich Heine University,<br />

Duesseldorf, Germany; University of British Columbia, Vancouver,<br />

BC, Canada<br />

Objectives: The receptor for hyaluronan-mediated motility (RHAMM,<br />

intracellular hyaluronan binding protein [IHABP], CD168), whose<br />

over-expression in tumors is associated with poor prognosis and<br />

early age at diagnosis, localizes to the centrosome, interacts with<br />

microtubules, functions in the maintenance of spindle pole stability<br />

and is required for centrosomal targeting. A previous study has shown<br />

that genetic variation at the HMMR locus (encoding for RHAMM)<br />

modifies breast cancer risk among BRCA1 mutation carriers and<br />

interplay between BRCA1, AURKA (aurora kinase A), RHAMM<br />

and phosphorylated RHAMM (pRHAMM) plays an important role<br />

in epithelial apicobasal polarization and breast carcinogenesis. Aim of<br />

this study was to investigate mechanisms by which BRCA1 regulates<br />

the function of RHAMM.<br />

Methods: Four pairs of shBRCA1 oligonucleotides containing target<br />

sequences against BRCA1 transcript were designed and synthesized.<br />

After annealing, these four double-strand shBRCA1 fragments were<br />

inserted into pLVTH vector and stably transfected into MCF-12A<br />

cells for knock-down of BRCA1 expression. The BRCA1 knockout<br />

breast cancer cell line HCC1937 with homozygous BRCA1<br />

c.5382insC mutation was transfected with the full length wt-BRCA1<br />

to restore BRCA1 function. HCC1937 cells were also transfected<br />

with the BRCA1-delExon11 isoform that lacks BRCA1 exon 11<br />

in its entirety. Western blotting analysis and immunohistochemical<br />

staining with anti-BRCA1, anti-RHAMM, anti-pRHAMM and anti-<br />

AURKA antibodies were performed. Real-time PCR was used to<br />

quantify RHAMM and AURKA expression in these different cell<br />

lines and transfectants .<br />

Results: Stable transfection of MCF-12A cells with the different<br />

shRNAs resulted in a significant decrease of BRCA1 transcript and<br />

protein levels. Real-time PCR showed increased RHAMM expression<br />

in MCF-12A cells transfected with shRNA BRCA1 knock-down<br />

constructs. Increased RHAMM gene expression was also determined<br />

in HCC1937 cells lacking BRCA1 function due to homozygous<br />

deleterious BRCA1 mutation c.5382insC. Restored wt-BRCA1<br />

expression in HCC1937 cells transfected with the full length BRCA1<br />

cDNA results in was significantly reduced RHAMM expression.<br />

Inhibition of RHAMM gene expression was even more pronounced<br />

in HCC1937 cells transfected with the BRCA1-delExon11variant<br />

lacking 1,143 amino acids of the BRCA1 protein. On the protein<br />

level loss of BRCA1 function affects nuclear localization of pT703-<br />

RHAMM the product of AURKA activity: pT703-RHAMM staining<br />

was revealed to be strong at the nuclear envelope in MCF12A BRCA1<br />

knock-down transfectants compared to the homogenous and less<br />

intense but clearly nuclear pT703-RHAMM staining in MCF12A<br />

wt-BRCA1 cells.<br />

Conclusions: BRCA1 negatively regulates RHAMM gene expression<br />

and might be involved in the nuclear localization of pRHAMM.<br />

Interestingly on the transcription level the BRCA1delexon11 isoform<br />

of BRCA1 displayed a stronger RHAMM repressor function than<br />

BRCA1 wild type. Taken together, our data provide insight into new<br />

functions of BRCA1 regulating RHAMM expression and mediating<br />

pRHAMM function.<br />

P2-08-01<br />

PIK3CA mutations associate with decreased Ki67 in early stage<br />

breast cancer (BC) and better outcomes in patients even among<br />

those with low Ki67 tumors.<br />

Datko FM, Patil S, Kalinsky K, Asher M, Wen YH, Hedvat C,<br />

Moynahan ME. Memorial Sloan-Kettering <strong>Cancer</strong> Center; Columbia<br />

University Medical Center<br />

Background: In studies of over 450 BC patients (pts) with > 10 years<br />

follow-up, pts with PIK3CA-mutated primary tumors had improved<br />

clinical outcome (Kalinsky et al 2009, Cizkova et al 2012). PIK3CA<br />

mutations associated with favorable clinicopathologic features: lower<br />

grade, smaller size, lymph node negativity (-), ER positivity (+),<br />

HER2-. In BC, several studies have suggested that PIK3CA mutations<br />

do not associate with PI3K/mTOR pathway activation (Loi et al 2010,<br />

Stemke-Hale et al 2008). To gain additional insight into the favorable<br />

biology imparted by a PIK3CA mutation in early stage BC and to<br />

identify predictive biomarkers, we performed immunohistochemistry<br />

(IHC) on tissue microarrays (TMA) constructed from 590 primary<br />

BCs.<br />

Methods: TMAs derived from FFPE tumors previously genotyped<br />

for PIK3CA mutations (rate of 32.5%, 192/590) were stained for ER,<br />

HER2 and AR (Kalinsky et al 2009). Here, we performed additional<br />

stains for MIB1 (Ki67 proliferation index, Ki67), p53 and markers<br />

of PI3K pathway activation (pS6, PTEN). Slides were scanned with<br />

Aperio ScanScope XT and segmented using TMALab (Aperio).<br />

Images were analyzed with the Aperio algorithm based on staining<br />

pattern (nuclear, cytoplasmic, membrane) and scored for % + and<br />

intensity. Manual review was performed for tumors with less than 3<br />

cores or discordant results. PTEN was scored manually: 0 (no tumor<br />

staining, PTEN loss), 1 (tumor < stroma), or 2 (tumor ≥ stroma). P<br />

www.aacrjournals.org 237s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

values were based on the log-rank test for comparison of Kaplan-<br />

Meier curves for disease free survival (DFS), Chi-square test for<br />

categorical variables and t-test for continuous variables.<br />

Results: On average, 66 cases (11%) were non-informative for each<br />

IHC stain, due to missing cores or insufficient tumor cells. In a binary<br />

analysis of Ki67 using a cutoff of ≥10% + cells, PIK3CA mutated<br />

tumors demonstrated significantly lower proliferation index with only<br />

9.4% of tumors (16/170) having high Ki67 as compared to 23.6% of<br />

wild-type PIK3CA tumors (82/347) (P=.001). Ki67 was also highly<br />

associated with PIK3CA genotype when analyzed with a cutoff of<br />

≥13.25% + cells or as a continuous variable. Importantly, DFS was<br />

significantly different when analyzed by PIK3CA genotype and<br />

Ki67 (P=.01). Among pts with low Ki67 tumors (n=419), PIK3CA<br />

mutation associated with longer DFS (10yr 79%, CI 69-85) as<br />

compared to wild-type PIK3CA (10yr 71%, CI 64-77). When PI3K<br />

pathway interactions were analyzed, PTEN loss associated with<br />

wild-type PIK3CA (P


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 2<br />

challenge because of ploidy changes induced by drugs. We made<br />

a HER2 genetic examination by Fluorescent in situ Hybridization<br />

(FISH) of invasive breast cancers before and after taxane treatment.<br />

After treatment we found supernumerary HER2 gene copies up to 15<br />

copies in the cases non-amplified at diagnosis.<br />

Material and methods: All 410 invasive breast carcinoma samples<br />

were collected in a single laboratory (Service de Pathologie, Centre<br />

Jean Perrin). Tumor samples were collected by biopsy at diagnosis or<br />

by surgical excision after taxanes chemotherapy. For all cases we had<br />

matched samples, one before and one after therapy. Tumor samples<br />

were formalin-fixed paraffin-embedded. IHC staining was done to<br />

detect the expression of HER2 protein. Only cases which were IHC<br />

2+ or IHC 1+ (with scattered HER2 heterogeneous staining) were<br />

checked by FISH.<br />

Results: After taxane-based neoadjuvant chemotherapy (4-8<br />

cycles) thirty patients (HER2 negative at Dg) out of 344 showed<br />

supernumerary HER2 gene copies detected in giant cells with large<br />

polyploid nuclei. The proportion of polyploid post-therapy cells<br />

among regular-size cancer cells (diploid) was from 15 to 30%. The<br />

FISH examination of polyploid cells revealed an increase of HER2<br />

copy number (and chromosome 17centromere) up to 15. The same<br />

copy number raise was observed with EGFR and ALK FISH probes.<br />

The ratio of all 30 cases with two distinct tumor cell populations<br />

(near diploid cells and giant cells) was up to 1.65 (not amplified).<br />

In one third of cases the average number of HER2 gene copies was<br />

higher than 6.<br />

Conclusions and recommendations: As a result of taxane<br />

administration, polyploid cells are formed and that most (if not<br />

all) chromosomes have been replicated simultaneously because<br />

of absent chromosomal segregation secondary to microtubule<br />

inhibition by taxanes. This HER2 copy increase is distinct from the<br />

mechanisms accounting for a more restricted gene amplification<br />

during carcinogenesis. The viability of giant polyploid cells is<br />

particularly low and most if not all of these cells succumb to mitotic<br />

catastrophe, an onco-suppressive mechanism causing cell cycle<br />

arrest and cell death. Irrespective of this consideration, it would be<br />

a critical error to classify cases as HER2 amplified solely only the<br />

bases of HER2 copy number >6. We recommend that the detection<br />

of more than 6 HER2 copies after taxane-based chemotherapy should<br />

be prompt a validation step involving the exclusion of polyploid cells<br />

from the analysis, as well as hybridization with a second and third<br />

probe specific for a chromosome other than 17. This validation step<br />

appears crucial to avoid unnecessary, costly treatments with HER2-<br />

targeted therapies after the failure of taxane-based chemotherapies.<br />

The clinico-pathological parameters of tumors with giant cells will<br />

be discussed.<br />

P2-09-01<br />

Reactivation of epigenetically silenced retinoic acid receptorbeta<br />

for therapy of breast cancer- from molecular mechanism to<br />

potential clinical applications<br />

Ordentlich P, Nguyen N, Jin K, Sadik H, Han L, Sukumar S. Syndax<br />

Pharmaceuticals; Sidney Kimmel <strong>Cancer</strong> Center, Johns Hopkins<br />

University School of Medicine<br />

Background: Triple negative breast cancer (TNBC) is a subgroup of<br />

breast cancer that is characterized by a lack of expression of targets<br />

for validated targeted therapies. Although responses to chemotherapy<br />

are observed, resistance develops rapidly. Genomic and molecular<br />

profiling of TNBC has identified multiple contributors to their<br />

uncontrolled growth including epigenetic dysregulation of genes<br />

important for normal cell growth and differentiation. Recent studies<br />

have shown that histone deacetylase (HDAC) inhibitors reverse the<br />

epigenetic profile of tumors, resulting in the re-expression of silenced<br />

genes encoding proteins such as ERα, EGFR, and RARβ. Entinostat is<br />

an oral, class 1 isoform selective HDACi recently shown in a phase 2<br />

study to be active in ER+ breast cancer. Clinical trials with entinostat<br />

in HER2+ and TNBC are also in progress. We hypothesized that<br />

combining epigenetic therapy using entinostat, with differentiation<br />

therapy using a retinoic acid receptor agonist- All Trans Retinoic<br />

Acid (ATRA) will provide an effective strategy for impeding the<br />

growth of TNBC and potentially sensitize tumors to commonly used<br />

chemotherapies (doxorubicin, carboplatin, paclitaxel).<br />

Methods: Combination studies of entinostat combined with ATRA<br />

with and without non-toxic doses (half clinical) of chemotherapy were<br />

carried out in vitro and in xenograft models of TNBC. Expression<br />

of RARb and downstream effectors were measured in cell lines and<br />

tumors from xenograft studies.<br />

Results: Nontoxic (half clinical) doses of doxorubicin when combined<br />

with entinostat and ATRA in vitro and in vivo exerted the best response<br />

when compared with combinations using paclitaxel and carboplatin.<br />

Re-expression of the silenced RARβ and downstream effectors was<br />

observed in the cell lines treated in vitro and in tumor xenografts<br />

of MDA-MB-231 and SUM159 cells in vivo. To gain insight into<br />

mechanism of action for the entinostat – doxorubicin combinations<br />

we examined the expression of enzyme targets of doxorubicin and<br />

determined that entinostat treatment reduced the expression of<br />

topoisomerase (Topo) II enzymes. Besides its conventional role in<br />

cell replication and division, Topo II has an important function in<br />

regulating transcription, especially in the nuclear receptor pathways<br />

such as ER and RAR. Our results demonstrated that Topo IIβ has a<br />

dual role in regulating the expression of RARβ. Inhibition of Topo IIβ<br />

in a time- and dose dependent manner up-regulated the expression of<br />

RARβ and its downstream genes. However, completely eliminating<br />

Topo IIβ decreased expression of RARβ.<br />

Conclusions: Our detailed study provides a new approach to treating<br />

TNBC using combinations of nontoxic doses of chemotherapeutic<br />

drugs with epigenetic therapy, and a deeper understanding of the<br />

molecular pathways involved in this response. These results suggest<br />

that combination of entinostat and a retinoic acid receptor agonist<br />

with low dose doxorubicin will provide an effective strategy for<br />

treatment of TNBC.<br />

P2-09-02<br />

Epigenetic Regulation of histone variants - a role in hormone<br />

therapy resistant breast cancer?<br />

Nayak SR, Oesterreich S, Pathiraja T. Magee Women’s Research<br />

Institute, University of Pittsburgh Medical Center, Pittsburgh, PA;<br />

Lester & Sue Smith <strong>Breast</strong> Center, Baylor College of Medicine,<br />

Houston, TX<br />

OBJECTIVE To utilize a genome wide approach to characterize<br />

epigenetic changes associated with resistance to estrogen deprivation,<br />

thus mimicking resistant to AIs.<br />

BACKGROUND Mechanisms of endocrine resistance include the<br />

activation of growth factor receptor pathways, overexpression of<br />

ER co-activators, and metabolic resistance due to polymorphisms<br />

in metabolizing enzymes. Recently, epigenetic mechanisms have<br />

been investigated to explain endocrine resistance. The majority of<br />

mechanisms for acquired resistance have been described following<br />

tamoxifen therapy, but the mechanisms for resistance to aromatase<br />

inhibitors (AI) remain less well known.<br />

HYPOTHESIS Estrogen deprivation results in altered methylation<br />

and altered expression of critical target genes in breast cancer cells,<br />

www.aacrjournals.org 239s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

which helps the cell to survive in the absence of estrogen, contributing<br />

to acquired AI resistance in breast cancer patients.<br />

MATERIALS AND METHODS We utilized derivatives of the<br />

ER_ MCF-7 cell line which were isolated after being cultured in<br />

estrogen-free media for 9 months (C4-12 cell line) and 18-24 months<br />

(LTED, Long term estrogen deprivation cell line). A genome-wide<br />

methylation screen was performed using Methyl CpG binding Domain<br />

(MBD) pull down assay followed by hybridization into Affymetrix<br />

Human Promoter 1.0R Array. Altered DNA methylation was validated<br />

by bisulfite genomic sequencing assays while gene expression of<br />

candidate genes was studied by qRT-PCR and Western blotting.<br />

RESULTS There were a significant amount of differentially<br />

methylated genes in C4-12 and LTED when compared to<br />

MCF7. Intriguingly, among the genes which were significantly<br />

hypomethylated in both endocrine resistant cell lines were a number<br />

of histone genes, with the homomorphic variant HIST1H2BE<br />

showing the strongest hypomethylation. Canonical histones have<br />

both homomorphic and heteromorphic variants, which differ in their<br />

timing of expression and similarity to the canonical histone protein’s<br />

amino acid sequence. Overexpression of various heteromorphic<br />

histone variants have been implicated in carcinogenesis, but to date,<br />

there have been no studies on homomorphic variants, and specifically<br />

H2B variants, in regards to their role in tumorigenesis. We show that<br />

hypomethylation of HIST1H2BE was associated with its upregulation<br />

in C4-12 and LTED cells. Furthermore, knockdown of of HIST2BE<br />

resulted in decreased growth in the endocrine resistant cells, but not<br />

in the parental MCF-7 cell lines.<br />

CONCLUSION & FUTURE DIRECTIONS The study is novel<br />

in two ways; first, this is the first study to show a potential role for<br />

histone variants in endocrine resistance. Second, this is one of very<br />

few examples where hypomethylation (instead of hypermethylation)<br />

results in altered expression of genes critical in tumorigenesis.<br />

Current studies include the detailed characterization of the phenotype<br />

following over-expression and knockdown of HIST1H2BE, the<br />

examination of the timing of expression of this variant in the S phase,<br />

and finally we are measuring methylation and expression of HIST2BE<br />

in a large collection of tumors from AI-treated breast cancer patients.<br />

P2-09-03<br />

Identifying a landscape of DNA methylation-driven genes in<br />

breast cancer using MethylMix<br />

Gevaert O, Plevritis S. Stanford University<br />

Background<br />

DNA methylation is being recognized as an important mechanism that<br />

contributes to oncogenesis. Hyper- and hypo-methylation of genes<br />

have been identified as critical events in multiple human cancers. A<br />

vast amount of data is now available that allows a comprehensive<br />

genome wide study of DNA methylation levels of cancer compared<br />

to normal tissue. Yet few statistical algorithms exist that exploit these<br />

vast datasets to quantify methylation levels and identify hypo- and<br />

hyper-methylated genes in cancer.<br />

Methods<br />

We developed a new computational method called MethylMix based<br />

on a Gaussian mixture modeling to identify DNA methylation states<br />

relative to normal tissue. We applied MethylMix on breast cancer<br />

data from The <strong>Cancer</strong> Genome Atlas (TCGA). Using paired gene<br />

expression data; we associate DNA methylation with gene expression<br />

data to identify functional DNA methylation events. Moreover, we<br />

searched for patient subgroups with similar methylation patterns and<br />

compare these with normal methylation data.<br />

Results<br />

MethylMix identified 344 and 172 significantly hyper- and<br />

hypo-methylated genes respectively. For example MTAP is<br />

hypermethylated in 30% of the samples and is known to be codeleted<br />

with the tumor suppressor gene CDKN2A. MTAP is also<br />

more frequently hypermethylated with increasing stage (Fisher<br />

exact test p-value 0.0007). Another interesting example is MUC1,<br />

hypermethylated in 17% of the samples but also significantly<br />

hypomethylated in the remaining 83% of the samples. MUC1’s<br />

methylation status differentiates between basal and luminal A tumors<br />

through hypermethylation in basal and hypomethylation in luminal A<br />

tumors (Fisher exact test p-value < 0.0001) consistent with MUC1’s<br />

role associated with ER positive breast cancer. Next, we observed<br />

hypomethylation of PTPN22 in the basal breast cancers (Fisher<br />

exact test, p-value < 0.0001) a tyrosine phosphatase involved in<br />

T cell receptor signaling. Similarly MMP7 is hypomethylated in<br />

basal breast cancers and hypermethylated in luminal (Fisher exact<br />

test, p-value < 0.0001). In contrast, a number of breast cancer genes<br />

previously reported to be methylated could not be confirmed due to<br />

various reasons. We found no functional or differential methylation<br />

for APC, CCND2, CDH1, CDH13, RARB, CDKN2A, HOXA5 and<br />

TWIST1 due to lack of correlation with gene expression or lack<br />

of significant difference with normal DNA methylation. We also<br />

confirmed a significant methylation pattern associated with basal<br />

breast cancer characterized by hypomethylation of genes related to the<br />

extracellular space. In addition, we observed a significant difference<br />

in the number of hypermethylated genes per molecular subtype. More<br />

specifically the luminal A and B subtypes together have significantly<br />

more hypermethylated genes compared to the basal subtype (164<br />

hypermethylated genes in luminal vs. 88 in basal, Wilcoxon rank<br />

sum test = -0.2, and ratio of<br />

tumor to normal mean beta >= 2.0).<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

240s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 2<br />

Estrogen receptor-negative BC is a more aggressive form than ER<br />

positive BC with approximately double the incidence in African<br />

Americans than in Caucasian Americans. The emerging differential<br />

methylation pattern within hormone receptor negative breast cancers<br />

would further help stratify them into distinct subgroups. Promotor<br />

methylation being potentially reversible, methylated genes may serve<br />

as future molecular targets for demethylating therapies.<br />

Support: Komen Foundation: KG110218<br />

P2-09-05<br />

Screening of significantly hypermethylated genes in breast cancer<br />

using MIRA-based microarray and identifying their expression<br />

levels<br />

Lian Z-q, Wang Q, Li W-p, Zhang A-q, Wu L. <strong>Breast</strong> Disease Center,<br />

Guangdong Women and Children Hospital of Guangzhou Medical<br />

College<br />

Background. Despite extensive studies in recent years, molecular<br />

biomarkers for early detection of breast cancer are urgently needed.<br />

This study aims to screen candidate methylation markers for early<br />

detection of breast cancer and explore the relationship between<br />

methylation and gene expression.<br />

Methods. We performed methylated-CpG island recovery<br />

assay(MIRA) combined with CpG island array on 61982 CpG sites<br />

across 4162 genes in 10 breast cancer tissues and 10 non-cancer<br />

breast tissues. Direct bisulfite sequencing and combined bisulfite<br />

restriction analysis (COBRA) were carried out in independent cancer<br />

and non-cancer samples. Gene expressions were analyzed by using<br />

microarray and validated by using RT-PCR.<br />

Results. We detected 70 significantly hypermethylated genes in breast<br />

cancer tissues, including many novel hypermethylated genes such<br />

as ITGA4, NFIX, OTX2 and FGF12. Direct bisulfite sequencing<br />

showed widespread methylations occurred in intragenic regions of<br />

WT1, PAX6 and ITGA4 genes and promoter region of OTX2 gene in<br />

breast cancer tissue. COBRA assay confirmed that WT1, OTX2 and<br />

PAX6 genes were hypermethylated in breast cancer tissues. Clustering<br />

analysis of gene expressions of 70 significantly hypermethylated<br />

genes revealed that most hypermethylated genes in breast cancer were<br />

not expressed in breast tissues. RT-PCR assay validated that WT1 and<br />

PITX2 were only weakly expressed in the breast cancer tissues and<br />

weren’t expressed in most non-cancer breast tissues. OTX2 and PAX6<br />

weren’t expressed in both breast cancer tissues and non-cancer tissues.<br />

Conclusions. Our results will expand our knowledge of<br />

hypermethylated genes and methylation sites for early detection<br />

of breast cancer and deepen our understanding of relationship<br />

between methylation and gene expression. MIRA approach can more<br />

effectively screen candidate methylated genes for further clinical<br />

validation than gene expression microarray-based strategy.<br />

P2-09-06<br />

The structure design and biological activities of inhibitory<br />

peptides, which block the interactions among polycomb repressive<br />

complex 2<br />

LI KK, Luo L, Kong X, Li L, Luo C. Rui Jin Hospital Affiliated with<br />

the Shanghai JiaoTong University School of Medicine, 197 Rui Jin<br />

Road II, Shanghai, China; Shanghai Institute of Materia Medica,<br />

Chinese Academy of Sciences, Shanghai, Shanghai, China<br />

Polycomb repressive complex (PRC2) contains several proteins,<br />

including embryonic ectoderm development (EED), suppressor<br />

of zeste 12 (SUZ12) and enhancer of zeste homolog 2 (EZH2).<br />

Excessive EZH2 concentrations have been reported as a marker of<br />

aggressive breast cancer and associated with invasion and cancer<br />

progression. EZH2 levels were elevated in patients with invasive<br />

breast carcinoma relative to normal or atypical hyperplasia. Except<br />

for the C-terminal SET domain, which functions as a histone-lysine<br />

N-methyltransferases, EZH2 has an N-terminal alpha helix region,<br />

which forms tight complex with EED protein. The in vivo enzymatic<br />

activity of EZH2 relies on and be tightly regulated by the interaction<br />

with EED. We designed series of artificial peptides trying to block<br />

the interaction between EZH2 and EED. By using label-free surface<br />

plasmon resonance (SPR) technology, the dynamic binding capacities<br />

of these peptides were tested. There were 3 leading structures standing<br />

out from the screening of more 80 peptides. After several round of<br />

co-crystal structure based optimization, the binding capacity of one<br />

inhibitor was reached to several nM levels, which indicated the<br />

availability for being a drug candidate. Finally, the biological activities<br />

of the inhibitory peptides were tested in several breast cancer cell<br />

lines. The PRC2 inhibitory peptides in this study are the first time<br />

reported PRC2 targeting epigenetic molecules with biological activity<br />

in breast cancer.<br />

P2-10-01<br />

PAM50 gene signature is prognostic for breast cancer patients<br />

treated with adjuvant anthracycline and taxane based<br />

chemotherapy<br />

Liu MC, Pitcher BN, Mardis ER, Davies SR, Snider JE, Vickery T,<br />

Reed JP, DeSchryver K, Singh B, Friedman PN, Gradishar WJ, Perez<br />

EA, Martino S, Citron ML, Norton L, Winer EP, Hudis CA, Perou CM,<br />

Ellis MJ, Barry WT. Georgetown University Lombardi Comprehensive<br />

<strong>Cancer</strong> Center, Washington, DC; Duke University Medical Center,<br />

Durham, NC; Washington University, St. Louis, MO; Washington<br />

University School of Medicine, St. Louis, MO; New York University<br />

Medical Center, New York, NY; University of Chicago, IL; Robert<br />

H. Lurie Comprehensive <strong>Cancer</strong> Center, Northwestern University<br />

Feinberg School of Medicine, Chicago, IL; Mayo Clinic, Jacksonville,<br />

FL; The Angeles Clinic and Research Institute, <strong>San</strong>ta Monica, CA;<br />

Hofstra North Shore-LIJ School of Medicine, ProHEALTH Care<br />

Associates, Lake Success, NY; Memorial Sloan-Kettering <strong>Cancer</strong><br />

Center, New York, NY; Dana Farber <strong>Cancer</strong> Institute, Harvard<br />

Medical School, Boston, MA; Lineberger Comprehensive <strong>Cancer</strong><br />

Center, University of North Carolina at Chapel Hill, NC; Siteman<br />

<strong>Cancer</strong> Center, Washington University School of Medicine, St. Louis,<br />

MO<br />

Background: Intrinsic breast cancer subtypes determined by the<br />

PAM50 assay are reported to be prognostic independent of standard<br />

clinicopathological variables. The CALGB (Alliance) 9741 adjuvant<br />

trial randomized treatment in a 2x2 factorial design to (i) 2-wk<br />

(dose dense; DD) vs 3-wk therapy and (ii) sequential vs concurrent<br />

doxorubicin, cyclophosphamide, paclitaxel (A-->T-->C vs AC -->T).<br />

DD therapy improved disease-free survival and overall survival (OS)<br />

(Citron et al. JCO 2003.). A significant interaction between intrinsic<br />

breast cancer subtypes and the use of DD therapy was hypothesized.<br />

Methods: C9741 was conducted in collaboration with ECOG,<br />

NCCTG, and SWOG. Biospecimens were collected from 1652 of<br />

2005 subjects. Multiplexed gene expression profiling (NanoString<br />

Technologies, Inc) generated PAM50 subtype calls (luminal A,<br />

luminal B, HER2-enriched, basal-like), risk of relapse (ROR) score,<br />

and proliferation score for 1321 cases (80%). Excluded samples were<br />

due to insufficient tumor (n=181) or RNA (n=150). The primary<br />

endpoint was relapse-free survival (RFS). The primary analysis<br />

to determine if the clinical benefit of DD therapy is dependent on<br />

intrinsic subtype was conducted as a test of interaction in a Cox<br />

proportional hazards model (3 degrees of freedom, df; alpha=0.05).<br />

www.aacrjournals.org 241s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Similar analyses were performed for ROR score and proliferation<br />

score as continuous measures of risk (1 df). PAM50 subtype, ROR<br />

score, and proliferation score were evaluated in separate multivariate<br />

models adjusting for number of positive nodes, menopausal status,<br />

dose density, and sequence of therapy.<br />

Results: Improved outcomes for DD therapy in the evaluable subset<br />

mirrored results from the complete dataset (HR=1.20; 95%CI 0.99-<br />

1.44) with a median follow-up of 12.3 yrs. Subtype analysis identified<br />

32% luminal A (n=417), 26% luminal B (n=341), 20% HER2-enriched<br />

(n=267), and 22% basal-like (n=296). Intrinsic subtypes were<br />

prognostic of RFS (p


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 2<br />

also observed with other breast signatures and RS, including an<br />

imputed 70-gene signature of MammaPrint (rho=0.59, p=3e-8) and<br />

the 50-gene PAM50 risk-of-relapse score (rho=0.54, p=8e-7), while<br />

poor correlation was seen for the GENIUS prognostic model (rho =<br />

0.06, p = 0.6). Discovery of gene-level associations to RS identified<br />

28 genes, including known cancer-associated genes such as BRCA2<br />

(FWER adj p=0.002), Cyclin E1 (FWER adj p=0.015), and CDCA5<br />

(FWER adj p=0.048). Pathway-level association in KEGG and GO<br />

Biologic Processes, identified 24 and 104 categories respectively,<br />

including Cell Cycle pathways KEGG:04110 (FDR adj p=0.002)<br />

and GO:0000085 (FDR adj p=0.0007). Among 22 in-vitro derived<br />

oncogenic pathway signatures, significant negative correlation is seen<br />

with p53 (rho=-0.56, adj p=4e-6) and positive correlation with betacatenin<br />

(rho=0.38, adj p=0.015). A multi-gene model of association<br />

to OncotypeDX RS classification is being developed using Prediction<br />

Analysis of Microarrays (PAM).<br />

Conclusions: Microarray-based prognostic breast cancer signatures<br />

are generally concordant. However, their application across platforms<br />

is currently sub-optimal. In the context of OncotypeDX RS, additional<br />

key cancer-associated genes and pathways are found to be associated<br />

with breast cancer prognosis, potentially providing insight into<br />

treatment opportunities for ER+ breast cancer. Validation efforts of<br />

these findings in an independent patient cohort are underway.<br />

Funding: NCI: RC2CA14041-01 and W81XWH-07-1-0394<br />

P2-10-04<br />

The Mammostrat Test is an Effective Tool to Stratify Patient<br />

Samples Previously Characterized as Intermediate by the<br />

Oncotype Dx Test<br />

Bloom KJ, Kyshtoobayeva A, Hilaire L, Gutekunst K. Clarient<br />

Diagnostic Services, Aliso Viejo, CA<br />

Mammostrat is a well validated, 5-antibody immunohistochemistry<br />

(IHC) based assay initially developed to assess the risk of recurrence<br />

in patients with early stage estrogen receptor (ER) positive breast<br />

cancer. Recent data from the TEAM trial demonstrated that<br />

Mammostrat predicted distant relapse free survival (DRFS) for both<br />

exemestane and tamoxifen-exemestane treated patients irrespective of<br />

nodal status and chemotherapy. OncoType Dx® is a recurrence assay<br />

frequently used by oncologists to aid in the selection of early stage<br />

breast cancer patients who may benefit from adjuvant chemotherapy.<br />

It is currently unclear whether patients whose tumors demonstrate an<br />

intermediate risk score should receive chemotherapy. We sought to<br />

determine if tumors with intermediate OncoType Dx® scores would<br />

be clearly classified as high or low risk with the Mammostrat assay.<br />

Formalin fixed paraffin embedded tissue blocks were retrieved from<br />

143 patients whose tumor were previously assessed as intermediate<br />

with the OncoType Dx® assay (Genomic Health, Redwood City,<br />

Ca). Six serial 5 micron sections were cut from the tissue block. One<br />

section was stained with H&E and the remaining five sections were<br />

stained with the Mammostrat antibodies, NDRG1 (AGI-3-3-12E<br />

clone, Clarient, Aliso Viejo, CA), TRMT2A (AGI-2-33-12E clone,<br />

Clarient, Aliso Viejo, Ca), P53 (D07 clone, Dako, Carpenteria, CA),<br />

CEACAM (S0235 08/28/06 clone, Clarient, Aliso Viejo, CA)and<br />

SLC7A5 (AGI-1-108-2E clone, Clarient, Aliso Viejo, CA). Slides<br />

were reviewed by a single pathologist (KJB) using previously<br />

published Mammostrat criteria and a risk score of low, moderate or<br />

high was generated.<br />

Of the 143 tumors, 62 (43%) were classified as low risk, 51 (36%)<br />

were classified as high risk and 30 (21%) were classified as moderate<br />

risk. Thus, 113 (79%) cases were defined as either low risk or high<br />

risk by the Mammostrat assay.<br />

The Mammostrat assay reclassified 79% of OncoType Dx®<br />

intermediate tumors into a clear high risk or low risk category. Based<br />

on data from the TEAM trial, node negative patients categorized<br />

as low risk by Mammostrat had a 5 year DRFS of 96%, while data<br />

from the NSABP-B20 trial showed that patients categorized as high<br />

risk by the Mammostrat assay derived a significant benefit from<br />

adjuvant chemotherapy. Mammostrat may be an effective tool to aid<br />

oncologists and patients in treatment decisions when Oncotype Dx®<br />

classifies a tumor as intermediate risk.<br />

P2-10-05<br />

Continuous association of a 200-gene prognostic risk score<br />

with probability of neoadjuvant chemotherapy response and<br />

translation from fresh frozen to FFPE tissue for clinical use.<br />

van Laar R, DiPaola J, Carbaugh H, Murphy T, DiRamio A, Flinchum<br />

R, Brown N, Albino T. Signal Genetics, New York, NY<br />

<strong>Breast</strong>PRS is a suite of complimentary gene signatures that resolves<br />

several of the significant limitations of current gene expression<br />

signatures in breast cancer. Notably, <strong>Breast</strong>PRS can accurately provide<br />

prognosis prediction, histological grade resolution and molecular<br />

subtyping of invasive breast cancer. The 200-gene prognostic<br />

component was previously validated in an independent retrospective<br />

series of 1,016 previously published fresh-frozen breast cancers and<br />

has now been applied to data from over 6,000 patients.<br />

In part one of this study, a meta-analysis of 890 previously published<br />

neoadjuvant treated breast cancer patients was performed. A<br />

highly significant association was observed between prognosis<br />

risk group (high vs low), continuous risk score (0-100), and a<br />

patient’s probability of achieving complete pathologic response with<br />

neoadjuvant chemotherapy. Data from both fine-needle-aspirate and<br />

biopsy tissue were included in the cohort. Notably, fewer than 10%<br />

of patients with the lowest (most favorable) pre-treatment <strong>Breast</strong>PRS<br />

prognosis scores achieve pCR. Probability of pCR was observed<br />

to increase steadily to over 40% for individuals with the highest<br />

prognosis scores.<br />

In part two of the analysis, 50 paired fresh-frozen and FFPE tumors<br />

were profiled on whole genome microarrays containing all three<br />

signatures, in a CLIA-certified high throughput genomics laboratory.<br />

Results will be presented that highlight the consistency of the<br />

prognosis, grade and subtype signatures in clinically relevant FFPE<br />

tissue.<br />

This suggests that <strong>Breast</strong>PRS, in addition to identifying 10-year<br />

recurrence risk, histologic grade and molecular subtype, may enable<br />

physicians to determine a patients benefit from receiving neoadjuvant<br />

chemotherapy with more granularity than current clinical or genomic<br />

methods.<br />

<strong>Breast</strong>PRS Prognostic Index vs Neoadjuvant Chemotherapy Response<br />

<strong>Breast</strong>PRS Prognostic Risk<br />

Score Percentile<br />

Percent of patients achieving pCR Percent of patients with RD<br />

0 to 10th 11% 89%<br />

10th to 20th 7% 93%<br />

20th to 30th 12% 88%<br />

30th to 40th 19% 81%<br />

40th to 50th 18% 82%<br />

50th to 60th 26% 74%<br />

60th to 70th 26% 74%<br />

70th to 80th 34% 66%<br />

80th to 90th 35% 65%<br />

90th to 100th 42% 58%<br />

www.aacrjournals.org 243s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P2-10-06<br />

Multicenter I-SPY 1 TRIAL (CALGB 150007/150012): <strong>Breast</strong><br />

cancer stem cells are associated with intrinsic chemoresistance<br />

and worse survival.<br />

Landis MD, Yau C, Neumeister V, Rimm DL, I-SPY 1 TRIAL<br />

Investigators, Esserman L, Chang JC. The Methodist Hospital,<br />

Houston, TX; University of California, <strong>San</strong> Francisco, CA; Yale<br />

University School of Medicine, New Haven, CT<br />

<strong>Breast</strong> cancer stem cells (CSCs) contribute to therapeutic resistance.<br />

I-SPY is a multi-center neoadjuvant trial for women with locally<br />

advanced breast cancers (3cm or greater) who received neoadjuvant<br />

doxorubicin and cyclophosphamide then paclitaxel. We hypothesized<br />

that CSC markers will be increased by neoadjuvant chemotherapy in<br />

patients who did not achieve a pathologic complete response, and that<br />

women with elevated CSC markers will have worse survival in this<br />

multicenter study. Methods: 215 women completed surgery, of those<br />

156 had residual tumor, of whom 56 had sufficient tumor sampled preand<br />

post-neoadjuvant chemotherapy to enable measurements of CSCs<br />

by automated quantitative analysis (AQUA) of immunofluourescent<br />

staining for aldehyde dehydrogenase (ALDH1), CD44, and ALDH1/<br />

CD44. A threshold of background staining was determined for each<br />

marker from a tissue microarray representing multiple cell lines<br />

and patient specimens and was used to define samples positive for<br />

CSCs. Effect of treatment on proportion of samples positive for CSCs<br />

was assessed using the McNemar test. Association between CSC<br />

markers and recurrence free survival (censored at 5 years) (RFS)<br />

were evaluated using Kaplan-Meier analysis of cohorts dichotomized<br />

at optimal marker thresholds as determined from ROC analysis for<br />

RFS prediction, as well as univariate Cox analysis of continuous<br />

CSC marker scores. Results: Of the 56 patients studied median<br />

tumor size was 6cm, 30% HR-neg, and 75% HER2 neg (by central<br />

and/or local IHC and/or FISH). In the HER2-neg subset, despite<br />

reducing tumor size in approximately 76% of cases, chemotherapy<br />

significantly increased the proportion of samples positive for CSCs:<br />

ALDH1 (38% to 74%), CD44 (67% to 88%), and ALDH1 in CD44<br />

compartment (43% to 76%) (McNemar test p = 0.0007, 0.03, and<br />

0.001, respectively). Within this HER2-neg subset, tumors with high<br />

pre-treatment CSC expression (i.e. CSC marker scores greater than<br />

optimal thresholds for RFS prediction – ALDH1 score ≥470.5, CD44<br />

score ≥8662, ALDH1 in CD44 compartment score ≥742.5) appeared<br />

to have worse RFS with a hazard ratio of 3.7, 2.8, and 2.1 respectively<br />

(log rank p=0.03, 0.07, 0.17). Interestingly, Cox regression analysis<br />

revealed that pretreatment ALDH1 score, as well as the ALDH1 in<br />

CD44 compartment score appeared significantly associated with<br />

RFS in the HR positive HER2 neg subset (n=33) (hazard ratio:<br />

2.2 (1.3 - 3.8), p=0. 003, and hazard ratio: 1.8 (1.1 – 2.8), p=0.01<br />

respectively). Surprisingly, post-treatment CSC markers expression<br />

did not show significant RFS associations within the HER2 neg<br />

or the HR positive HER2 neg subsets. Conclusion: These results<br />

from this multicenter study validate the hypothesis of treatmentresistant<br />

cancer stem cells, with an increase in CSC markers post<br />

chemotherapy, and suggest worse prognosis in women whose tumors<br />

have high expression of these stem cell markers, especially for HER2<br />

negative patients. Surprisingly, only pre- (and not post-) treatment<br />

CSC marker expression appears to associate with RFS. Studies to<br />

evaluate inhibitors of stem cell self-renewal and conventional therapy<br />

should be considered for HER2-neg breast cancers.<br />

P2-10-07<br />

Stem cell marker aldehyde dehydrogenase 1 in stromal cells<br />

strongly correlates with prognosis in breast cancer<br />

Sjöström M, Hartman L, Holmdahl J, Grabau D, Malmström P, Fernö<br />

M, Niméus-Malmström E. Lund University, Lund, Sweden<br />

Background: <strong>Breast</strong> cancer stem cells have the potential to provide<br />

prognostic information and be a therapeutic target. Aldehyde<br />

dehydrogenase 1 (ALDH1) is a widely used marker for cells with stem<br />

cell properties. The aim of this study was to evaluate the prognostic<br />

information of ALDH1, and its correlation with other clinical variables<br />

in early breast cancer.<br />

Material and methods: ALDH1 expression was investigated by<br />

immunohistochemistry in tissue microarrays of breast tumors from<br />

220 premenopausal node-negative breast cancer patients, mainly<br />

not subjected to any adjuvant systemic treatment. <strong>Cancer</strong> cells were<br />

evaluated for both staining intensity (negative, weak, moderate<br />

and strong) and percentage of stained cells. Stromal cells were<br />

evaluated for staining intensity. Spearman’s rank correlation was<br />

used to investigate correlation with continuous clinical variables, the<br />

Jonckheere-Terpstra Test for ordinal variables and Mann-Whitney for<br />

binary variables. For survival analysis with 5 years of follow-up the<br />

Kaplan-Meier method and Log-rank test for trend were used with<br />

distant disease-free survival (DDFS) as endpoint. Cox regression<br />

analysis was used to obtain hazard ratios, and for multivariate<br />

modeling.<br />

Results: ALDH1 staining intensity in stroma correlated positively<br />

with both ER and PR (p=0.004 and p=0.001). Stroma staining<br />

intensity was negatively correlated with Ki67 expression and high<br />

histological grade (p=0.002 and p=0.002). There was no significant<br />

correlation of stroma intensity with age, HER2 or tumor size. Stroma<br />

intensity showed a strong correlation with increased DDFS (Log-rank<br />

test for trend p=0.007).<br />

Both ALDH1 staining intensity in cancer cells as well as percentage<br />

of positively stained cancer cells correlated negatively with PR status<br />

(p=0.08 and p=0.03) while no correlation was found for ER (p=0.65<br />

and p=0.59). Staining intensity and percentage of positively stained<br />

cancer cells correlated positively with Ki67 expression (p=0.03<br />

and p=0.02). <strong>Cancer</strong> cell intensity showed a tendency of positive<br />

correlation with high histological grade (p=0.07). There was no<br />

correlation of percentage of positively stained cancer cells with<br />

histological grade (p=0.21), nor of staining intensity and percentage<br />

of positively stained cancer cells with HER2, tumor size, or age at<br />

diagnosis. <strong>Cancer</strong> cell intensity and percentage of positive cancer<br />

cells showed no correlation with DDFS (log rank test for trend<br />

p=0.79 and p=0.78).<br />

A multivariate model of ALDH1 in stroma including ER, HER2,<br />

Ki67, age (continuous), and tumor size showed that ALDH1 was an<br />

independent prognostic marker (p=0.036). Interestingly, ALDH1 in<br />

stroma (HR 0.58, mean per step in intensity) had stronger influence<br />

on the prognosis than ER status (HR 1.12), Ki67 (HR 1.62) and<br />

tumor size (HR 1.17).<br />

Conclusion: Strong ALDH1 staining intensity in stromal cells,<br />

in comparison with negative or weak staining, is a strong marker<br />

for increased DDFS in breast cancer and outperforms well-known<br />

prognostic markers such as ER, Ki67 and tumor size as an independent<br />

prognostic marker. The finding that stromal cell staining is correlated<br />

to DDFS may be an indication of the importance of the tumor<br />

microenvironment.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

244s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 2<br />

P2-10-08<br />

Prospective comparison of Recurrence Score and different<br />

definitions of luminal subtypes by central pathology assessment<br />

of single markers in early breast cancer: results from the phase<br />

III WSG-planB Trial<br />

Gluz O, Kreipe HH, Kates RE, Christgen M, Liedtke C, Shak S,<br />

Clemens M, Salem M, Liedtke B, Aktas B, Markmann S, Uleer C,<br />

Augustin D, Thomssen C, Nitz U, Harbeck N. West German Study<br />

Group, Moenchengladbach, NRW, Germany; Ev. Bethesda Hospital,<br />

Moenchengladbach, NRW, Germany; Medizinische Hochschule<br />

Hannover, Niedersachsen, Germany; University of Muenster,<br />

Muenster, Germany; Genomic Health, Inc., Germany; Clinics<br />

Mutterhaus, Trier, Germany; University of Cologne, Germany;<br />

Ev. Hospital, Bergisch Gladbach, Germany; University of Essen,<br />

Germany; Clinics Südstadt, Rostock, Germany; Gemeinschaftspraxis<br />

Hildesheim, Hildesheim, Germany; Clinics Deggendorf, Deggendorf,<br />

Germany; University of Halle, Germany; University of Munich,<br />

Munich, Bavaria, Germany<br />

Background: Routine use of multigene RT-PCR based assays as<br />

Recurrence Score (RS) vs. single markers (grade, uPA/PAI-1, HR,<br />

HER2, Ki-67) is controversially discussed in early BC. Several<br />

definitions of luminal A/B subtypes have been proposed by St. Gallen<br />

guidelines and individual researchers (grade 1/2 vs. 3, Ki-67 cut-offs<br />

14 or 20%). Recently, integration of PR>20% was proposed as an<br />

immunhistochemical luminal A subtype definition (Ki-67


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

adherence to internal protocols and mandatory participation in quality<br />

assurance programs. These results provide further confirmation that<br />

the vast majority of patients eligible for trastuzumab are not deprived<br />

from an effective treatment by using this algorithm.<br />

P2-10-10<br />

Clinical implications of molecular heterogeneity in highly<br />

proliferative, ER-positive, HER2-negative breast cancer<br />

Bianchini G, Pusztai L, Kelly CM, Iwamoto T, Callari M, Symmans<br />

WF, Gianni L. Ospedale <strong>San</strong> Raffaele, Milan, Italy; MD Anderson<br />

<strong>Cancer</strong> Center, Houston, TX; Mater Misericordiae University<br />

Hospital, Dublin, Ireland; Okayama University Hospital, Okayama,<br />

Japan; Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy<br />

Objectives: Different clinical behaviors are observed in tamoxifentreated<br />

and untreated ER-positive, HER2-negative highly-proliferative<br />

breast cancer (BC) that demonstrate either high (highERS) or low<br />

(lowERS) expression of estrogen-related genes (Bianchini SABCS<br />

2011). LowERS tumors are intrinsically endocrine resistant and<br />

at significant risk of relapse in the first 5 yrs after diagnosis. We<br />

studied lowERS and highERS BC in pts treated with neoadjuvant<br />

chemotherapy (NAC) and examined prognostic and predictive<br />

markers in the highest risk group of lowERS BC.<br />

Methods: We examined affymetrix gene expression data from<br />

193 ER+/HER2-, high proliferation BC from pts treated with<br />

taxane-anthracycline-based NAC followed by endrocrine therapy.<br />

Previously defined cut-offs for markers of proliferation (MKS),<br />

and estrogen-related genes were applied (Bianchini SABCS 2011).<br />

Within the lowERS group, we examined pts treated with no systemic<br />

adjuvant therapy (n=137; 50 events); adjuvant tamoxifen-only<br />

(n=141; 36 events); and NAC (n=127, 27 RCB0/I). We performed<br />

gene enrichment analysis for 2617 gene sets with known biological<br />

function (by 5000 random permutations). Primary endpoints were<br />

distant event free survival (DEFS) with follow-up censored at 5-yrs<br />

and pathological response (pathR) using the residual cancer burden<br />

(RCB) (Symmans JCO 2007).<br />

Results: The median follow-up of the NAC series was 3.1yrs. The<br />

DEFS at 4yrs was 0.94 [0.87-1.00] and 0.70 [0.60-0.81] in the high<br />

and low ERS groups, respectively (p=0.004) (despite the higher rate<br />

of pathR (RCB0/I) to NAC in the low ERS group (9.5% and 21.9%;<br />

p=0.04)). The pathR was prognostic in the lowERS group [HR 9.1<br />

(CI 1.23-67.4); p=0.009] but not in highERS (p=0.485). In contrast,<br />

a different outcome was observed in BC with RCBII-III, were the<br />

4-yrs DEFS was 0.93 [0.86-1.00] and 0.61 [0.49-0.76] in high and low<br />

ERS group, respectively (p=0.0007). In the lowERS group there was<br />

substantial overlap in biological functions associated with prognosis in<br />

both tamoxifen-treated and untreated pts. At a conservative threshold<br />

of p 1700) demonstrated a prognostic power of the EP score<br />

beyond what can be achieved by combining the commonly used<br />

clinicopathological parameters (Filipits M, 2011). The performance<br />

of the EP in chemotherapy-treated patients has not been evaluated<br />

yet. Here, we analyzed the EndoPredict score in node-positive ER+/<br />

HER2- BC patients from the GEICAM-9906 trial, treated with<br />

adjuvant chemotherapy followed by hormonal therapy.<br />

Methods: Patients included in this study participated in the<br />

GEICAM/9906 trial and were either treated with fluorouracil,<br />

epirubicin, and cyclophosphamide (FEC) or with FEC followed by<br />

weekly paclitaxel (FEC-P) (Martin M, 2008). ESR1 and ERBB2 gene<br />

expression were assessed by qRT-PCR in 800 formalin-fixed paraffin<br />

embedded (FFPE) tumor samples out of 1246 patients included in<br />

the GEICAM/9906 trial. The EndoPredict score (including eight<br />

prognostic genes) was successfully determined in 555 out of the<br />

566 ER+/HER2- patients. Patients were assigned into two categories<br />

(high/low), according to the predefined EP cut-off value (Filipits M,<br />

2011). The primary endpoint for the analysis was distant metastasis.<br />

Metastasis rates were estimated using the Kaplan–Meier method.<br />

Multivariate analysis was performed using Cox regression. Interaction<br />

between treatment effects and EP was tested as well.<br />

Results: Twenty-five percent of patients (n=141) were classified as<br />

EP-low-risk. Kaplan Meier analysis demonstrated that the metastasisfree<br />

survival (MFS) was 92% in the EP-low risk vs. 69% in the<br />

EP-high-risk group (absolute difference of 23%, HR 4.4 (2.3-8.4) p<br />

< 0.0001). Multivariate analysis showed that EP is an independent<br />

prognostic parameter after adjustment for age, grade, lymph node<br />

status and tumor size. EP was found to be prognostic in pre-<br />

(p=0.0002, HR = 5.5 (2.2-13.6)) and postmenopausal (p=0.0129, HR<br />

= 3.3 (1.3-8.2)) BC patients. There were not statistically significant<br />

differences in MFS between treatment arms (FEC vs. FEC-P) neither<br />

in the high nor in the low-risk groups. Interaction test between<br />

chemotherapy arm and EP score was not significant.<br />

Conclusions: The results of this study shows that the EndoPredict<br />

score is an independent prognostic parameter in node-positive, ER+/<br />

HER2- BC patients treated with adjuvant chemotherapy followed<br />

by hormonal therapy. EndoPredict was not found to be predictive of<br />

weekly paclitaxel efficacy. Novel predictive biomarkers are needed<br />

to identify the small subset of patients with ER+/HER2- tumors that<br />

actually benefit from weekly paclitaxel-containing regimens.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

246s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 2<br />

P2-10-12<br />

How well predict the 2011 St Gallen early breast cancer surrogate<br />

phenotypes metastatic survival?<br />

Vanderstichele A, Brouckaert O, Tuyls S, Amant F, Leunen K, Smeets<br />

A, Berteloot P, Van Limbergen E, Weltens C, Peeters S, Moerman P,<br />

Floris G, Paridaens R, Wildiers H, Vergote I, Christiaens M-R, Van<br />

Calster B, Neven P. University Hospitals, Leuven, Belgium<br />

BACKGROUND<br />

The five prognostic surrogate phenotypes, defined by the 2011 St<br />

Gallen consensus, predict metastatic relapse following early breast<br />

cancer treatment (Brouckaert et al Ann Oncol 2012). It is unknown to<br />

what extent these surrogate phenotypes defined on the primary tumor<br />

predict metastatic survival (MS) in a cohort of consecutive women<br />

with metachronous metastases.<br />

PATIENTS & METHODS<br />

All 4318 patients with primary operable breast cancer, diagnosed<br />

between 01-01-2000 and 31-12-2009 and treated in our center were<br />

prospectively entered in our institutional database. We included 345<br />

patients with a (distant) metastatic relapse during their follow-up.<br />

We observed metastatic survival as the time between the diagnosis<br />

of the relapse and death. Tumor subtype was defined according to<br />

the 2011 St Gallen recommendations in 5 groups: luminal A (ER+/<br />

HER2-, grade 1-2), luminal B1 (ER+/HER2-, grade 3), luminal B2 or<br />

luminal-HER2 (ER+/HER2+), HER2-like (ER-/HER2-) and triplenegative.<br />

We focused on the value of the primary tumor subtype as<br />

a major prognostic determinant, but also assessed age at diagnosis,<br />

tumor size, lymph node involvement, tumor subtype, PR-status,<br />

primary detection (screening vs. palpation), histology, adjuvant<br />

therapy, distant-metastasis free interval (DMFI), first-line metastatic<br />

hormonal therapy, recurrence sites (isolate or multiple) and metastasis<br />

detection (clinical vs. biochemical).<br />

RESULTS<br />

In our cohort, we recorded a median metastatic survival (MS) of<br />

22.3 months (95% CI: 19.7 – 25.6) and a 5-year survival probability<br />

of 15%. Significant differences in median MS were noted when<br />

considering the primary tumor subtype (cf. table).<br />

We observed an independent prognostic effect for multiple recurrence<br />

sites (HR = 1.78, 95% CI 1.37-2.32), first-line metastatic hormonal<br />

therapy (HR = 0.30, 95% CI 0.18 - 0.49), DMFI (HR = 0.92 CI 0.85-<br />

0.98) and, finally, primary tumor subtype.<br />

Independent prognostic factors in metastatic breast cancer survival<br />

VARIABLE N (%)<br />

DESCRIPTIVE<br />

UNIVARIATE<br />

MULTIVARIATE<br />

ANALYSIS<br />

ANALYSIS<br />

ANALYSIS<br />

Median MS P-value Hazard Ratio P-value Hazard Ratio P-value<br />

(95% CI) (Logrank) (95% CI) (Logrank) (95% CI) (Wald)<br />

Luminal A 100 (29.2) 2.61 (2.14 - 3.07)


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Results:<br />

34 675 non-TNBC patients achieved a DFS of 92,4 % at 2 years and<br />

81,2 % at 3 years whereas out of 3728 TNBC-patients only 79 %<br />

were disease-free at 2 years and 57,4 % at 3 years.<br />

With regards to overall survival 97,3 % of non-TNBC patients were<br />

alive at 2 years and 92,2 % at 3 years, whereas 89,3 % TNBC-patients<br />

were alive at 2 years and 82,9 % at 3 years.<br />

Conclusion:<br />

In this large registry of certified breast units from the West German<br />

<strong>Breast</strong> Center we could underline the significant differences of<br />

prognosis as to DFS and OS between the non-TNBC and TNBCsubgroup<br />

of patients, displaying survival rates with modern 3 rd<br />

generation chemotherapy, surgical treatment and radiotherapy in a<br />

controlled setting with prospectively collected data.<br />

To is, to our best knowledge, one of the largest data sets ever exploring<br />

the differences in prognosis between TNBC and non-TNBC-patients.<br />

P2-10-15<br />

Evaluation of Prognostic and Predictive Performance of <strong>Breast</strong><br />

<strong>Cancer</strong> Index and Its Components in Hormonal Receptor-Positive<br />

<strong>Breast</strong> <strong>Cancer</strong> Patients: A TransATAC Study<br />

Sgroi DC, Sestak I, Zhang Y, Erlander MG, Schnabel CA, Goss PE,<br />

Cuzick J, Dowsett M. Massachusetts General Hospital and Harvard<br />

Medical School, Boston, MA; Queen Mary University, London, United<br />

Kingdom; bioTheranostics Inc, <strong>San</strong> Diego, CA; Royal Marsden<br />

Hospital, London, United Kingdom<br />

Background: The Arimidex, Tamoxifen, Alone or in Combination<br />

(ATAC) trial compared the efficacy and safety of 5 years of<br />

anastrozole with tamoxifen as adjuvant treatment for postmenopausal<br />

women with localized HR+ breast cancer. At a median follow-up of<br />

10 years, a statistically significant improvement with anastrozole<br />

vs. tamoxifen for disease-free survival, time to recurrence and time<br />

to distant recurrence was observed. The HOXB13:IL17BR gene<br />

expression ratio (H/I) quantifies recurrence risk in ER positive (ER+)<br />

breast cancer patients and is predictive of benefit from endocrine<br />

therapy. Molecular Grade Index (MGI) is a five-gene index that<br />

provides quantitative and objective molecular assessment of tumor<br />

grade and proliferative status. <strong>Breast</strong> <strong>Cancer</strong> Index (BCI) combines<br />

H/I and MGI into a continuous risk model that provides a likelihood<br />

of distant recurrence in patients treated with endocrine therapy, and<br />

efficacy from neoadjuvant chemotherapy. In the current analysis,<br />

evaluation of the prognostic and predictive performance of BCI, H/I<br />

and MGI in the ATAC study cohort was conducted.<br />

Methods: Under the TransATAC protocol, formalin-fixed, paraffinembedded<br />

(FFPE) blocks of primary tumor were collected from HR+<br />

patients from each monotherapy arm. The current study examined<br />

samples collected from the United Kingdom, which constituted<br />

79% of the collection. RNA extracted from 1102 samples from the<br />

TransATAC study was amplified, converted to cDNA and subjected<br />

to RT-PCR with primers and probes to HOXB13, IL17BR, BUB1A,<br />

CENPA, NEK2, RACGAP1 and RRM2. H/I, MGI and BCI were<br />

calculated and risk groups were determined using pre-specified<br />

cutpoints.<br />

Results: Of 1102 tumor specimens assayed, 29 failed QC criteria,<br />

leaving 1073 samples for analysis. Detailed results on the prognostic<br />

and predictive performance of BCI, H/I and MGI will be presented.<br />

Data on whether BCI and its components provided independent<br />

prognostic information in the presence of classical variables, their<br />

prognostic value for risk of late recurrence, interaction by treatment<br />

arms, and comparative performance vs other models will also be<br />

discussed.<br />

Discussion: The ATAC trial has established the long-term efficacy and<br />

safety of anastrozole over tamoxifen as initial adjuvant treatment for<br />

post-menopausal early stage breast cancer patients. Continued efforts<br />

are needed to improve on quantification of residual risk in patients<br />

who were treated with endocrine therapy to guide decision-making<br />

in selecting additional adjuvant chemotherapy and/or administering<br />

extended endocrine treatment. This study will help to establish the<br />

strategy to more effectively select patients for adjuvant therapies.<br />

P2-10-16<br />

Quantitative HER3 protein expression and PIK3CA mutation<br />

status in matched samples from primary and metastatic breast<br />

cancer tissues and correlation with time to recurrence<br />

Sperinde J, Lara J, Michaelson R, Sun X, Conte P, Guarneri V,<br />

Barbieri E, Ali S, Leitzel K, Weidler J, Lie Y, Cook J, Haddad M,<br />

Paquet A, Winslow J, Howitt J, Hurley L, Eisenberg M, Petropoulos<br />

C, Huang W, Lipton A. Monogram Biosciences/Integrated Oncology/<br />

LabCorp, South <strong>San</strong> Francisco, CA; Saint Barnabas Medical Center,<br />

Livingston, NJ; University of Modena, Modena, Italy; Penn State/<br />

Hershey Medical Center, Hershey, PA; Lebanon VA Medical Center,<br />

Lebanon, PA; Laboratory Corporation of America, Research Triangle<br />

Park, NC<br />

Background: HER3 is thought to play a prominent role in resistance<br />

to HER2-directed breast cancer therapies. Recent data suggest that<br />

HER3 levels also influence HER2-normal breast tumor biology. HER3<br />

and PI3K signaling are linked in that in HER3 signaling activates<br />

PI3K and inhibition of PI3K activity can upregulate HER3 expression.<br />

Here, we measured quantitative HER3 protein expression levels and<br />

PIK3CA mutation status in matched tissues from the primary tumor<br />

and site of metastasis to assess correlations with time to recurrence.<br />

Methods: 44 pairs (8 HER2+ by HERmark®) of matched tissues from<br />

the primary tumor and the site of metastasis were evaluated for HER3<br />

protein expression using a sensitive, quantitative assay for HER3<br />

protein expression in FFPE tissue sections (VeraTag®). Matched<br />

samples were also evaluated for quantitative HER2 expression<br />

(HERmark) and for PIK3CA mutations at exon 9 (E542K and E545K)<br />

and exon 20 (H1047R).<br />

Results: HER3 protein expression at the metastatic site was largely<br />

independent of HER3 levels at the primary site (Spearman p=0.50) in<br />

contrast to HER2 expression (Spearman p=0.0004). HER3 expression<br />

in the primary tumor correlated with time to recurrence (TTR)<br />

(HR=2.0 per 2-fold increase in HER3; p


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 2<br />

tumor but not the metastatic tumor were associated with longer TTR.<br />

These observations suggest that HER3 protein expression may be an<br />

important prognostic factor for breast cancer progression.<br />

P2-10-17<br />

SET index predicts response to endocrine therapy rather than<br />

prognosis independently of other genomic signatures in a blinded<br />

validation study.<br />

Karn T, Hatzis C, Symmans F, Pusztai L, Ruckhäberle E, Schmidt M,<br />

Müller V, Hanker L, Heinrich T, Holtrich U, Kaufmann M, Rody A.<br />

Goethe-University; Nuvera Biosciences; The University of Texas MD<br />

Anderson <strong>Cancer</strong> Center; Gutenberg-University Mainz; University<br />

Hospital Hamburg-Eppendorf; Saarland-University, Homburg<br />

Background:<br />

A genomic index for sensitivity to endocrine therapy (SET) was<br />

previously derived from genes strongly positively and negatively<br />

coexpressed with estrogen receptor (Symmans et al. 2010, JCO<br />

28:4111). This SET index has been reported to predict survival benefit<br />

from adjuvant endocrine therapy independently of prognosis.<br />

Materials and Methods:<br />

Affymetrix gene expression profiling was performed on 307 ER<br />

positive primary breast cancers from a retrospective cohort treated<br />

solely with endocrine therapy. In a blinded study the SET index’ ability<br />

to predict survival was analyzed in this previously unpublished dataset.<br />

Affymetrix profiles (CEL files) were anonymized and provided to the<br />

developers of SET index (Nuvera Biosciences) without any clinical<br />

information. The 261 profiles that passed QC were categorized into<br />

SET classes based on the published prespecified cutoffs. Samples<br />

categorized as ER negative based on gene expression were excluded.<br />

Subsequently classifications were unblinded and clinical associations<br />

analyzed according to a predefined protocol. Exploratory analyses<br />

were performed on the relationship of SET index to other genomic<br />

signatures. We also assessed a potential pure prognostic value of<br />

SET index in an additional dataset of 164 node negative ER positive<br />

patients that did not receive any adjuvant treatment.<br />

Results:<br />

A lower SET index significantly correlated with higher grade<br />

(P=0.009) and negative PgR status (P=0.016). We detected no<br />

significant differences for age, tumor size, lymph node status, and<br />

HER2 status between patients with high, intermediate, and low SET<br />

index. In the lymph node negative (LNN) cohort (n=120) we observed<br />

a significant difference in DFS (5yr DFS 77.1±10.2% vs 94.4±2.2%;<br />

HR 4.20, 95% CI 1.72-10.2; P=0.002) and DMFS (HR 3.18, 95%<br />

CI 1.20-8.47; P=0.020) for patients with low SET index. In contrast,<br />

we found no prognostic value of SET index in lymph node positive<br />

patients (n=95). In multivariate analyses of LNN patients including<br />

SET, age, tumor size, histological grade, and PgR status, only SET<br />

was significantly associated with DFS (HR 3.37, 95% CI 1.25-9.01;<br />

P=0.016) and DMFS (HR 3.03, 95% CI 1.08-8.55; P=0.036). In<br />

exploratory analyses SET index was not correlated to other genomic<br />

signatures related to proliferation (as Recurrence Score, GGI, and<br />

NKI70) or immune response (e.g. 7IGS, SDPP). When we included<br />

all these genomic signatures as continous scores in a multivariate<br />

stepwise Cox regression model in the LNN endocrine treated cohort,<br />

only SET remained as significant (P=0.025) while Recurrence Score<br />

displayed a strong trend (P=0.054). We also verified that SET had<br />

no pure prognostic value in an additional dataset of 164 ER positive<br />

patients that did not receive any adjuvant treatment.<br />

Conclusions:<br />

In a blinded analysis the predictive ability of SET index was<br />

prospectively validated in an independent cohort of node-negative<br />

patients. Our exploratory analysis demonstrates that SET index is<br />

unrelated to other genomic signatures and delivers independent<br />

information on the response of patients to endocrine therapy rather<br />

than prognosis.<br />

P2-10-18<br />

Cell Cycle Profiling – risk score (C2P-RS) based on the specific<br />

activity of CDK1 and CDK2 predicts relapse in node-negative,<br />

hormone receptor-positive breast cancer treated with endocrine<br />

therapy<br />

Kim SJ, Masuda N, Tsukamoto F, Inaji H, Akiyama F, Sonoo H,<br />

Kurebayashi J, Yoshidome K, Tsujimoto M, Takei H, Masuda S,<br />

Nakamura S, Noguchi S. Graduate School of Medicine, Osaka<br />

University, Suita, Osaka, Japan; National Hospital Organization<br />

Osaka National Hospital, Osaka, Japan; Osaka Koseinenkin<br />

Hospital, Osaka, Japan; Osaka Medical Center for <strong>Cancer</strong> &<br />

Cardiovascular Diseases, Osaka, Japan; The <strong>Cancer</strong> Institute of<br />

Japan Foundation for <strong>Cancer</strong> Research, Tokyo, Japan; Kawasaki<br />

Medical School, Kurashiki, Okayama, Japan; Osaka Police Hospital,<br />

Osaka, Japan; Saitama <strong>Cancer</strong> Center, Kita-Adachi, Saitama, Japan;<br />

Nihon University School of Medicine, Tokyo, Japan; Showa University<br />

School of Medicine, Tokyo, Japan<br />

Background: We have previously reported on the Cell Cycle Profiling<br />

(C2P) assay for determining the specific activity (SA, activity/<br />

expression) of cyclin-dependent kinases (CDKs). The C2P-risk<br />

score (C2P-RS) based on CDK1 and CDK2 SAs was significantly<br />

associated with “tumor growth speed” in xenograft models and<br />

relapse in early breast cancer. This study was conducted to confirm<br />

the prognostic performance of C2P-RS classification in node-negative<br />

and hormone receptor (HR)-positive (ER- and/or PR-positive) breast<br />

cancers.<br />

Methods: This multicenter and blinded retrospective study included<br />

patients with node-negative, HR-positive tumors treated with<br />

endocrine therapy alone. Patients treated with adjuvant chemotherapy<br />

or trastuzumab or any kind of neoadjuvant treatments were excluded.<br />

C2P-RS was determined via the C2P assay (Sysmex Corporation,<br />

Kobe, Japan) using frozen samples obtained during surgery. C2P-RS<br />

classification based on C2P-RS and CDK1 SA classified patients into<br />

three groups (low, intermediate, and high C2P-RS groups) using the<br />

same cut-off values as previously reported. ER, PR, HER2, and Ki-<br />

67 expressions were centrally evaluated with immunohistochemistry.<br />

The primary endpoint was the 5y-relapse-free survival (RFS) rate.<br />

Results: Of 317 patients enrolled in this study, 266 (24-85 y) were<br />

eligible: menopausal status: pre- 44%, post- 56%; histologic grade:<br />

I 24%, II 58%, III16%; ER: positive 93%; PR: positive 82%; HER2:<br />

negative 96%. 22 (8.3%) patients relapsed within 5 y after surgery<br />

(median follow-up period, 73 mo). The distribution of each C2P-RS<br />

group was 71.8% in the low group, 12.0% in the intermediate group,<br />

and 16.2% in the high group. The Kaplan-Meier method showed that<br />

the 5y-RFS rate in the high C2P-RS group (74.4%) was significantly<br />

lower than that in the intermediate (84.3%) or the low C2P-RS<br />

groups (97.3%) (P


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

and Ki-67 expression (P=0.009) were significantly associated with<br />

relapse. However, multivariate Cox analysis showed that only C2P-<br />

RS classification was a significantly independent prognostic factor<br />

(the low C2P-RS group vs. the high C2P-RS group: P


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 2<br />

ER- disease in a large scale breast cancer cohort with long term follow<br />

up. The effect seems unrelated to systemic endocrine treatment or<br />

chemotherapy.<br />

P2-10-21<br />

Prognostic value of estrogen receptor status in women with<br />

synchronous or metachronous breast cancers<br />

Huo D. University of Chicago, IL<br />

Background: Estrogen receptor (ER) is an important prognostic<br />

and predictive factor for breast cancer (BC) patients. For women<br />

with two primary breast cancers, however, estrogen receptor status<br />

measured at the two tumors is not always the same, either due to<br />

tumor heterogeneity, or because of limited reproducibility of receptor<br />

assays. For women with metachronous breast cancers, it is unclear if<br />

estrogen receptor status of previous cancer has prognostic value. For<br />

women with synchronous breast cancers, it is important to know if<br />

inconsistent receptor status predicts worse clinical outcome.<br />

Methods: Data on ER status of two primary breast cancers were<br />

obtained from the Surveillance, Epidemiology, and End Results<br />

program 1990-2009. We used piecewise proportional hazard Cox<br />

models to examine the joint effect of ER statuses of two tumors on<br />

overall survival or disease-free survival. Hazard ratio (HR) and 95%<br />

confidence interval (CI) were calculated, after adjusting for age,<br />

stage, race/ethnicity, histology, grade, and treatments in Cox models.<br />

Synchronous and metachronous cases were analyzed separately.<br />

Results: There are 12,285 patients with synchronous breast cancers;<br />

81.2% had both tumors being ER positive, 10.2% had one tumor<br />

being ER positive but the other being ER negative, and 5.6% had<br />

both tumors being ER negative. As shown in the Table, patients with<br />

inconsistent ER status (heterogeneity) had worse short-term survival<br />

rate than patients with double ER positive tumors. Among the 14,470<br />

patients with metachronous breast cancers, 60.2% had two ER+<br />

tumors, 13.8% had two ER- tumors, 11.8% had ER status changing<br />

from negative to positive, and 14.1% had ER status changing from<br />

positive to negative. In addition to ER status of the index cancer, ER<br />

status of the first cancer has independent prognostic value in predicting<br />

both short-term and long-term outcome (p=0.0002).<br />

Table. Overall survival and estrogen receptors status in two primary breast cancers<br />

HR (95% CI), < 5 years HR (95% CI), >= 5 years<br />

ER in synchronous cases<br />

Both + 1.0 (ref.) 1.0 (ref.)<br />

One +, one - 1.72 (1.49-1.98) 0.99 (0.77-1.26)<br />

Both - 2.55 (2.22-2.93) 0.75 (0.57-0.99)<br />

ER in metachronous cases<br />

Both + 1.0 (ref.) 1.0 (ref.)<br />

- to + 0.96 (0.82-1.11) 0.75 (0.59-0.96)<br />

+ to - 1.46 (1.30-1.64) 0.67 (0.53-0.85)<br />

Both - 2.08 (1.85-2.35) 0.62 (0.47-0.82)<br />

Conclusions: Our study found that estrogen status of previous cancer<br />

has independent prognostic value for women with metachronous<br />

breast cancers. We also found that inconsistency in ER status between<br />

two tumors predicts worse survival for women with synchronous<br />

breast cancer. These findings suggest that heterogeneity or change<br />

in estrogen receptor expression in patients with two breast cancers<br />

should be considered during decision making on treatment or<br />

counseling on prognosis.<br />

P2-10-22<br />

Phosphorylation of Steroid Receptor Coactivator 3 (SRC3) at<br />

Ser543 is a novel independent prognostic marker in breast cancer<br />

Palmieri C, Gojis O, Rudraraju B, Abdel-Fatah TMA, Moore D,<br />

Shaw J, Green A, Ellis IO, Coombes RC, Ali S. Imperial College<br />

London, United Kingdom; Nottingham University City Hospital,<br />

Nottingham, United Kingdom; University of Leicester, United<br />

Kingdom; Nottingham University Hospitals, City Hospital Campus,<br />

Nottingham, United Kingdom<br />

Introduction:<br />

Steroid receptor coactivator 3 (SRC3) acts as a coactivator of nuclear<br />

receptors including estrogen receptor-alpha (ER). SRC3 has been<br />

implicated in the pathogenesis of breast cancer (BC) as well as in<br />

resistance to endocrine therapy. SRC3 is phosphorylated at a number<br />

of residues following the stimulation of growth factors or hormones.<br />

Tyr24 and Ser543 are both phosphorylated upon estrogen stimulation,<br />

while Tyr24 is modulated by JNK and Ser543 by both p38 and JNK.<br />

To date, the importance and potential role of phosphorylation at these<br />

residues has not been explored in BC. In this study we assessed the<br />

protein expression of SRC3, pTyr24 and pSer543 and association<br />

with clinico-pathological features and outcome in a well defined<br />

breast cancer series.<br />

Methods:<br />

The expressions of SRC3, pTyr24 and pSer543 were assessed in the<br />

Nottingham Tenovus Primary <strong>Breast</strong> <strong>Cancer</strong> Series which consists<br />

of 1650 cases of primary invasive. SRC3, pTyr24 and pSer543 were<br />

correlated with clinico-pathological data as well as outcome.<br />

Results:<br />

SRC3 expression was significantly associated with unfavourable<br />

clinicopathological features including ER -ve (p=0.02), PR –ve<br />

(p=.038), HER2 overexpression (p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Conclusion:<br />

Phosphorylation at Ser543 is associated with a luminal phenotype,<br />

positive prognostic factors and sensitivity to tamoxifen. Furthermore,<br />

it is an independent prognostic factor. pS543 is a novel prognostic<br />

marker in BC and warrants further investigation.<br />

P2-10-23<br />

Lymphovascular Invasion (LVI) and Overall Survival in Nodenegative<br />

and Node-positive <strong>Breast</strong> <strong>Cancer</strong> Patients: A Metaanalysis.<br />

Sahebjam S, Diaz-Padilla I, Ocana A, Seruga B, Amir E. Princess<br />

Margaret Hospital; Institute of Oncology Ljubljana<br />

Introduction: LVI is an important prognostic factor in patients with<br />

lymph node-negative invasive breast cancer. The prognostic value of<br />

LVI in lymph node-positive disease or in other subgroups remains<br />

unclear. Here we present a meta-analysis of studies assessing the<br />

impact of LVI on overall survival (OS) in different subgroups of<br />

breast cancer patients.<br />

Methods: A published data meta-analysis was conducted. Studies<br />

reporting hazard ratios (HR) for the association of LVI with OS in<br />

a multivariable model were included. Those not reporting HR or<br />

utilizing only univariable analyses were excluded. HR and 95%<br />

confidence intervals (CI) were extracted or computed and pooled<br />

using the DerSimonian and Laird random effects model. Subgroup<br />

analyses were conducted for the differential effect of lymph node<br />

involvement, estrogen receptor (ER) status, extent of LVI and decade<br />

in which the study was conducted (pre 1990, 1990-1999 and 2000<br />

onwards).<br />

Results: Twenty studies comprising 40,417 patients were included<br />

in the analysis. There was marked heterogeneity in the effect size<br />

between studies (Cochran Q p=5 U/ml had an 81% increased risk for<br />

recurrence (HR=1.810 [CI: 1.111-2.948]) and reduced overall<br />

survival (HR=1.020 [CI: 1.004 – 1.037]). In the multivariate analysis<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

252s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 2<br />

taking into account tumor size, nodal status, grading, age, hormonal<br />

and HER2/neu receptor status increasing CA27.29 levels were an<br />

independent prognostic marker.<br />

Conclusions: An increase of the tumor marker CA27.29 2 years after<br />

CHT compared to pre-chemotherapy baseline was associated with<br />

a worse prognosis. By using this approach, more patients at risk for<br />

recurrence were detected than with the standard threshold approach.<br />

Therefore, the use of relative change could help to identify more<br />

patients at risk for relapse who might benefit from an intensified<br />

follow up.<br />

P2-10-26<br />

Association between circulating tumor cells and molecular breast<br />

cancer subtypes<br />

Gang N, Haibo W, Funian L, Chen L, Xiaoyi L, Xingang W, Zhidong L.<br />

Affiliated Hospital of Medical College, Qingdao University, Qingdao,<br />

Shandong, China<br />

Background and Objective: Circulating tumor cells (CTCs) provide<br />

crucial information for the management of breast cancer patients.<br />

CTCs analysis can be used as a liquid biopsy approach for prognostic<br />

and predictive purposes in breast and other cancers. Several<br />

contradictory results presented about the breast cancer subtypes of<br />

CTCs recently. The aim of this study was therefore to analysis the<br />

association between the CTCs and molecular breast cancer subtypes.<br />

Methods: Blood samples were obtained from 143 breast cancer<br />

patients admitted to <strong>Breast</strong> Surgery Department, Affiliated Hospital of<br />

Medical College, Qingdao University. CTCs were detected by using<br />

immunomagnetic enrichment and a commercial kit, based on RT-PCR,<br />

was used to characterize CTCs. The expression of ER, PR, HER2<br />

was detected by immunohistochemistry. The molecular subtypes were<br />

defined as: luminal A (ER+ and/or PR+, HER2-), luminal B (ER+ and/<br />

or PR+, HER2+), TNBC (ER-, PR-, HER2-), and HER2+, (ER-,PR-,<br />

and HER2+). The relations between the positive rate of CTCs and<br />

clinicopathological characteristics of primary tumor and molecular<br />

subtypes of breast cancer patients were analyzed.<br />

Results: Overall positive rate of CTCs was 39.8% (57/143). The<br />

positive rate of CTCs in patients with Luminal A, Luminal B ,HER2+<br />

and TNBC subtype was 33.8% (26/77), 47.1% (8/17),50.0% (6/12)<br />

and 45.9%(17/37).There was significant difference in positive rate<br />

of CTCs by different molecular subtypes (P = 0.0415), especially<br />

at different AJCC stages (P = 0.004).The positive rate of CTCs in<br />

patients with Luminal A subtype was significantly lower than patients<br />

with the others breast cancer subtypes (P=0.0387).<br />

Conclusion: Our study suggests that presence of CTCs was more<br />

frequently found in patients with HER2+ and Luminal B subtypes.<br />

The presence of CTCs in patients with TNBC subtype was not as<br />

high as we wished. Theoretically, the positive rate of CTCs in TNBC<br />

subtype should be higher than that of others subtypes for being more<br />

aggressive histologically features and having increased potential to be<br />

invasive. One possible explanation for this might because of lacking<br />

epithelial markers expression and epithelial-mesenchyme transition<br />

(EMT) characteristics, CTCs of TNBC subtype are missed when using<br />

the standard EpCAM-based method. Shortage of patient’s number<br />

was another possible reason. Further research on the Association<br />

between CTCs and molecular breast cancer subtypes will contribute<br />

to a better understanding of the biology of metastatic development<br />

in cancer patients.<br />

P2-10-27<br />

No Discordant Receptor Status Results among 50 Paired <strong>Breast</strong><br />

and Axillary Metastasis Core Biopsies when Pre-analytic<br />

Variation is Controlled.<br />

Miller DV, Rosenthall RE, Avent JM, Carter CC, Hansen J, Hammond<br />

MEH. Intermountain Healthcare, Salt Lake City, UT<br />

Background: Discrepancy in Her2, estrogen receptor (ER), and<br />

progesterone receptor (PR) status among primary breast tumors and<br />

their axillary lymph node metastases has been reported in the range<br />

of 6-18%, leading to uncertainty as to the optimal sample for testing.<br />

It has been hypothesized this reflects heterogeneity within a given<br />

patient’s tumor, however the pre-analytic conditions (particularly<br />

time to fixation and fixation duration) for samples in these studies<br />

were either nonuniform for the sample pairs or else unspecified.<br />

Altered tissue antigenicity rather than true biologic alterations in<br />

receptor expression may account for some the discrepancy reported<br />

in the literature. This study of paired primary and metastatic tumors<br />

processed in a uniform fashion attempts to address this issue.<br />

Design: Core biopsy samples from 50 primary invasive ductal<br />

carcinomas and their corresponding ipsilateral axillary lymph node<br />

metastases obtained at the same procedure, using the same core biopsy<br />

sampling device, fixed under the same conditions, stained on the same<br />

staining runs, and analyzed using digital image analysis comprise the<br />

study cohort. Patient demographics and tumor characteristics (from<br />

subsequent excision specimens or clinical imaging) were obtained<br />

from medical records.<br />

Results: Complete agreement in Her2, ER, and PR scores was seen<br />

among all paired samples.The mean age of the 50 women was 51.72<br />

(27-86). Mean time to fixation was 2.55 minutes and the mean duration<br />

of fixation was 12.87 hours (6.4 -29.7). The Her2 scores were 0 = 2,<br />

1+ = 13, 2+ = 12, 3+ = 23. For 3+ cases, the mean tumor size was<br />

3.47 cm, 44% were ER negative and the mean tumor grade was 2.61<br />

(of 3). For 0/1+ cases, the mean tumor size was 3.86 cm, the mean<br />

tumor grade was 2.46 (38% were grade 3 and triple negative). The<br />

ER (Allred) scores were 0 = 17, 3 = 1, 4 = 1, 5 = 3, 6 = 9, 7 = 7, 8 =<br />

12. For ER negative tumors the mean tumor grade was 2.76. For ER<br />

positive tumors the mean tumor grade was 2.39 including all 3 grade<br />

1 tumors. Only minor discrepancies in Allred scores occurred between<br />

the paired samples; 5 with a difference of 1 (6-7, 7-8, 8-7, 7-6, 8-7);<br />

and one with a difference of 2 (8-6). The PR (Allred) scores were 0<br />

= 24, 2 = 3, 3 = 4, 4 = 8, 5 = 2, 6 = 6, 7 = 1. For PR negative tumors<br />

the mean tumor grade was 2.75. For PR positive tumors the mean<br />

tumor grade was 2.40, including all 3 grade 1 tumors. Only minor<br />

discrepancies in Allred scores occurred between the paired samples; 4<br />

with a difference of 1 (3-4, 4-5, 4-5, 6-7); and three with a difference<br />

of two (3-5, 4-6, 4-6). The mean age did not differ significantly by<br />

any receptor status.<br />

Conclusion: Under matched processing conditions, these 50 paired<br />

primary breast tumors and their corresponding lymph node metastases<br />

show congruent Her2 and ER staining results. This number of cases<br />

should have detected discrepancies in the range of 6-18%. Minor<br />

differences in staining intensity and %positive cells were noted, but<br />

none resulted in a positive vs negative mismatch. These data suggest<br />

the possibility that these minor differences, magnified through the<br />

influence of disparate fixation conditions, could account for the higher<br />

discrepancy rates reported in the literature.<br />

www.aacrjournals.org 253s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P2-10-28<br />

The Prognostic Index, KiGE, Combining Proliferation,<br />

Histological Grade and Estrogen Receptor Status Challenges<br />

Gene Profiling – A Study in 1,854 Chemo-Naïve Women with N0/<br />

N1 Primary <strong>Breast</strong> <strong>Cancer</strong>.<br />

Strand C, Bak M, Borgquist S, Chebil G, Falck A-K, Fjällskog M-L,<br />

Grabau D, Hedenfalk I, Jirström K, Klintman M, Malmtröm P, Olsson<br />

H, Rydén L, Stål O, Bendahl P-O, Fernö M. Lund University, Division<br />

of Oncology, Skåne University Hospital, Lund, Sweden; Odense<br />

University Hospital, Odense, Denmark; Mammography, Bergaliden,<br />

Helsingborg, Sweden; Helsingborg Hospital, Helsingborg, Sweden;<br />

Uppsala University Hospital, Uppsala, Sweden; Lund University,<br />

Skåne University Hospital, Lund, Sweden; Lund University, Division<br />

of Pathology, Lund, Sweden; Skåne University Hospital, Lund,<br />

Sweden; Linköping University, County Council of Östergötland,<br />

Linköping, Sweden; Lund University, Division of Surgery, Skåne<br />

University Hospital, Lund, Sweden<br />

Purpose<br />

The aim was to validate the prognostic value of a previously defined<br />

index (CAGE), combining proliferation (Cyclin A), histological<br />

grade, and estrogen receptor (ER) in different subsets of primary<br />

breast cancer patients. In the present study, Ki67 was included in the<br />

index (KiGE) instead of cyclin A, since it is the generally accepted<br />

proliferation marker in clinical routine.<br />

Patients and methods<br />

1,854 chemo-naive patients with primary breast cancer were included.<br />

The low KiGE group consisted of histological grade 1 cases and grade<br />

2 cases which were ER-positive and had low Ki67 expression. High<br />

KiGE consisted of all other cases.<br />

Results<br />

The KiGE index separated patients into groups with different<br />

prognosis. In multivariate analysis, KiGE was significantly associated<br />

with disease-free survival, when adjusted for age at diagnosis,<br />

tumor size and adjuvant endocrine treatment (hazard ratio: 3.5, 95%<br />

confidence interval: 2.6-4.7, P


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 2<br />

P2-10-30<br />

Expression of lipid metabolism genes in contralateral unaffected<br />

breast associated with estrogen receptor status of breast cancer<br />

Wang J, Scholtens D, Holko M, Ivancic D, Lee O, Hu H, Chatterton<br />

RT, Khan SA. Northwestern university, Chicago, IL<br />

Background: Risk biomarkers that are specific to ER subtypes of<br />

breast cancer would help developing distinct prevention strategies.<br />

The contralateral unaffected breast of women with unilateral breast<br />

is a good model for defining subtype specific risk as women with<br />

ER- primary tumor are at high risk for subsequent ER- tumor in<br />

contralateral breast.<br />

Methods: We performed random fine needle aspiration of unaffected<br />

breast of cases. RNA samples from 30 subjects (15 ER+ and 15 ER-)<br />

were used for Ilumina expression array analysis. Findings were<br />

validated by qRT-PCR using independent sample set consisted of<br />

36 subjects (12 ER+, 12 ER-, and 12 standard risk healthy controls).<br />

Results: Gene array studies identified 18 genes with significant<br />

higher expression in ER- case samples , 8 of which were associated<br />

with lipid metabolism. This pattern was confirmed by qRT-PCR<br />

in the same samples and in the 24 cases of validation set. When<br />

compared to the healthy controls, significant over-expression of 4<br />

genes (DHRS2, HMGCS2, HPGD, ACSL3) was observed in ERcases,<br />

with significant lower expression of UGT2B11 and APOD in<br />

ER+ cases, and decreased expression of UGT2B7 in both subtypes.<br />

Conclusion: Differential expression of lipid metabolism genes my<br />

be involved in the risk for subtypes of breast cancer, which might be<br />

potential biomarkers of ER specific breast cancer risk.<br />

P2-10-31<br />

Correlation of quantitative p95HER2 and total HER2 levels<br />

with clinical outcomes in a combined analysis of two cohorts of<br />

trastuzumab-treated metastatic breast cancer patients<br />

Duchnowska R, Sperinde J, Leitzel K, Szostakiewicz B, Paquet A, Ali<br />

SM, Jankowski T, Haddad M, Fuchs E-M, Arlukowicz-Czartoryska<br />

B, Winslow J, Singer C, Wysocki PJ, Lie Y, Horvat R, Foszczynska-<br />

Kloda M, Petropoulos C, Radecka B, Litwiniuk M, Debska S, Weidler<br />

J, Huang W, Biernat W, Köstler WJ, Jassem J, Lipton A. Military<br />

Institute of Medicine, Warsaw, Poland; Monogram Biosciences/<br />

Integrated Oncology/LabCorp, South <strong>San</strong> Francisco, CA; Penn State/<br />

Hershey Medical Center, Hershey, PA; Medical University of Gdansk,<br />

Poland; Lublin Oncology Center, Lublin, Poland; Medical University<br />

of Vienna, Austria; Bialystok Oncology Center, Bialystok, Poland;<br />

Greater Poland <strong>Cancer</strong> Center, Poznan, Poland; West Pomeranian<br />

Oncology Center, Szczecin, Poland; Opole Oncology Center, Opole,<br />

Poland; Poznan University of Medical Sciences, Poznan, Poland;<br />

Regional <strong>Cancer</strong> Center, Lódz, Poland<br />

Background: Expression of p95HER2 (p95), a truncated form of<br />

HER2 also known as p110 or M611-CTF, is a possible trastuzumab<br />

resistance mechanism and has been associated with poor prognosis in<br />

trastuzumab-treated HER2-positive metastatic breast cancer (MBC).<br />

Previously we reported on optimal clinical cutoffs for quantitative<br />

p95 (Clin <strong>Cancer</strong> Res, 16:4226, 2010) and quantitative HER2 protein<br />

expression (H2T) by HERmark® (<strong>Cancer</strong>, 116:5168, 2010) that<br />

defined patient subsets with different progression-free survival (PFS).<br />

These cutoffs were confirmed in an independent trastuzumab-treated<br />

MBC cohort (ASCO 2011, #586). Here, using individual patient data,<br />

we performed an analysis on the combined data set of 243 cases from<br />

the discovery and validation cohorts to derive optimal cutoffs for<br />

quantitative p95 and H2T.<br />

Methods: Both quantitative H2T (HERmark, Monogram Biosciences)<br />

and p95 assays employed the VeraTag® method to quantify protein<br />

expression in formalin-fixed, paraffin-embedded tumor samples from<br />

two cohorts of 101 and 142 cases of trastuzumab-treated MBC with<br />

7.4 and 9.2 months median PFS, respectively. All analyses were<br />

stratified by hormone receptor status, tumor grade (3 vs. 1+2) and<br />

cohort. H2T measurements were compared to pre-specified cutoffs for<br />

HERmark negative (H2T17.8 RF/mm 2 ), derived from<br />

the 95 th percentile of centrally determined HER2-negatives, respectively,<br />

within a reference database of 1,090 breast cancer patient samples.<br />

Results: Patients classified as HERmark-positive had longer PFS<br />

than those classified as HERmark-negative (HR=0.52; p=0.0006;<br />

medians 10.0 and 5.9 months). The previously determined optimal<br />

H2T cutoff of 13.8 RF/mm 2 in the center of the HERmark-equivocal<br />

zone, gave a similar result (HR=0.54; p=0.0005). This was close to<br />

the optimal cutoff of 12.75 RF/mm 2 (HR=0.48; p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Whitney tests. Spearman’s correlation determined the non-parametric<br />

correlation between the serum levels of sLeX and the inflammatory<br />

mediators. The receiver operating characteristic (ROC) curves and<br />

the corresponding area under the curve (AUC) analyses were used<br />

to determine the sensitivity and specificity of a given cut-off value<br />

for a particular serum marker.<br />

Results: The median sLeX level tended to increase with the stage of<br />

disease: MBC > EBC > DCIS albeit without significant differences<br />

among the disease stages. Among MBC patients, patients with sLeX<br />

below 1.75 U/mL had significantly improved overall survival (OS,<br />

mean survival 11.1 vs. 33.7 months, P = 0.002) and progression-free<br />

survival (PFS, mean survival 9.7 vs. 20.9 months, P= 0.042). The<br />

Hazard Ratio of high sLeX for OS was 5.5 (95% CI 1.6 to 18.9, p =<br />

0.007) and 2.3 for PFS (95% CI 1.0 to 5.2, P = 0.048). EBC and MBC<br />

patients have significantly higher serum levels of IL-1, IL-1RA, IL-<br />

6, IL-8, MCP-1, MCP-3, and MIP-1βthan those of HD. In addition,<br />

there were positive correlations between the serum levels of sLeX<br />

and cytokines IL-1β, IL-1RA, IL-2, IL-8, MIP-1β, and MCP-3. The<br />

AUC for sLeX was 0.598 (P = 0.016), and a cut-off of 3.13 pg/mL<br />

distinguished hormone receptor (HR)-positive from HR-negative<br />

patients (χ2 = 4.0, P = 0.045). Likewise, the AUC for TNF-α was<br />

0.620 (P = 0.003), and a cut-off 7.18 pg/mL distinguished HR-positive<br />

from HR-negative patients (χ2 =12.6, P < 0.001). Using a cut-off<br />

value established by ROC curves, few MBC patients (9 of 66, 13.6%)<br />

had a serum IL-2 level > 7.1 pg/mL compared to 57 of 185 (30.8%)<br />

non-MBC patients (χ2 =7.4, P = 0.007), suggesting that metastatic<br />

disease may be associated with immune suppression related to low<br />

serum IL-2. Conversely, 31of 66 (47%) MBC patients had a serum<br />

MCP-1 level > 750 pg/mL vs. 37 of 185 non-MBC patients (20%)<br />

(χ2 =23.8, P < 0.0001), suggesting that a high level of MCP-1 may<br />

play an important role in metastasis.<br />

Conclusion:<br />

Serum levels of sLeX were able to distinguish HR-positive from HRnegative<br />

patients and predict overall survival in metastatic patients.<br />

Serum sLeX and some inflammatory mediators tended to increase<br />

with the severity of disease, and together may facilitate local invasion<br />

of tumor cells. Furthermore, serum levels of MCP-1 and IL-2 may<br />

have prognostic value in breast cancer patients.<br />

P2-10-33<br />

Mitotic Component of Grade Can Distinguish <strong>Breast</strong> <strong>Cancer</strong><br />

Patients at Greatest Risk of Local Relapse<br />

Done SJ, Miller NA, Wei Shi W, Pintilie M, McCready DR, Liu F-F,<br />

Fyles A. University Health Network, Toronto, ON, Canada; Princess<br />

Margaret Hospital, University Health Network, Toronto, ON, Canada;<br />

University of Toronto, ON, Canada<br />

Background<br />

With the recent recognition of many different molecular subtypes of<br />

breast cancer a desire to more specifically categorize tumors to allow<br />

tailoring of treatment to individual patients has developed. This has<br />

largely involved the development of molecular tests rather than the<br />

re-examination of current pathologic criteria. We wanted to evaluate<br />

standard pathologic features to determine their ability to predict for<br />

local recurrence.<br />

Materials and Methods<br />

Slides were retrieved for review from 280 of 769 women who had<br />

participated in a trial of tamoxifen with or without breast irradiation<br />

between December 1992 and June 2000 and for whom outcome<br />

data up to 18 years was available. All women were 50 years of age<br />

or older at the time of enrollment and had T1 or T2 node negative<br />

breast cancer. The cases for which slides were obtained were<br />

representative of the whole group. The slides were reviewed by two<br />

breast pathologists (SJD and NAM). Several features were evaluated;<br />

modified Nottingham histologic grade and its components- degree of<br />

tubule formation, nuclear pleomorphism and mitotic count. Mitotic<br />

component of grade was calibrated to the microscopic field size used.<br />

The presence of lymphatic/vascular space invasion was also scored. A<br />

statistical analysis was performed to relate these pathologic features<br />

to local recurrence at up to 18 years.<br />

Results<br />

The strongest predictor of local recurrence was the mitotic component<br />

of the Nottingham histologic grade with 5.7% for mitotic score 1/3<br />

(n=200), 19.6% for mitotic score 2/3 (n=37) and 19.8% for mitotic<br />

score 3/3 (n=43)(Gray’s p-value = 0.0021). Overall grade was also<br />

able to predict for local recurrence with 2.6% for Grade 1 (n=49),<br />

10.6% for Grade 2 (n=162) and 17.9% for Grade 3 (n=71)(Gray’s<br />

p-value=0.026). However, neither architecture (0% vs. 9.5% vs.<br />

9.8%, Gray’s p-value=0.74) nor degree of nuclear pleomorphism<br />

(0% vs. 7.9% vs. 11.5%, Gray’s p-value=0.37), the other components<br />

of histologic grade, showed a statistically significant difference<br />

for recurrence. The presence or absence of endothelial lined space<br />

invasion was also found to be not statistically different (9.3% vs.<br />

13%, Gray’s p-value=0.55).<br />

Conclusion<br />

Within this cohort of tamoxifen treated T1 and T2 breast cancer<br />

patients 50 years of age or older, mitotic index could stratify women<br />

into groups with high and low risk of recurrence. If validated this may<br />

be a useful way of allocating patients to different treatment groups.<br />

Additional validation studies are planned on similar groups of patients.<br />

P2-10-34<br />

Development and validation of ClinicoMolecular Triad<br />

Classification (CMTC), a platform for breast cancer (BC)<br />

prognostic and predictive gene signature portfolios.<br />

Leong WL, Wang D-Y, Done S, McCready D. University Health<br />

Network, Toronto, ON, Canada<br />

Background: Numerous gene signatures have claimed prognostic<br />

significance in BCs. Each of these gene signatures was designed<br />

to answer a specific clinical or biological question, often by<br />

dichotomizing the targeted populations into a good and a bad risk<br />

group. None of these gene signatures on its own has sufficient degree<br />

of complexity to fully characterize this very heterogenous group of<br />

diseases, and hence lacks the flexibility to personalize treatments.<br />

To exploit the full potential of the genomic approach, we developed<br />

an 803-gene molecular classification, termed ClinicoMolecular<br />

Triad Classification (CMTC) that categorized BCs into 3 clinical<br />

treatment groups (triad) that can serve as a basic framework to<br />

guide management. CMTC also provide a detailed "portfolio" of 14<br />

other gene signatures and 19 oncogenic pathways to allow further<br />

customization of the treatments. The ability to get CMTC portfolio<br />

results at the time of initial diagnosis offers the unique advantage<br />

of early treatment planning, including the use of pre-operative<br />

chemotherapy to improve breast conservation in selected patients.<br />

This study aimed to validate the CMTC classification using an<br />

independent BC cohort.<br />

Study design/ results: RNA from fine needle aspirates were collected<br />

in a prospective BC cohort (n=340) between 2008 and 2010 at Princess<br />

Margaret Hospital and Mount Sinai Hospital, Toronto, we included<br />

all newly diagnosed BC patients going for surgery who consented<br />

to join the study. DNA microarray analyses were carried out using<br />

genome-wide Illumina Human Ref-8 version 3 Beadarrays, which<br />

contained >24K oligonucleotide probes. After excluding tumors with<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

256s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 2<br />

low RNA yield (n=8, success rate 97%), non-invasive cancers (n=27),<br />

insufficient follow-up data (n=21), CMTC divided the remaining<br />

284 BCs into 3 similar sized groups (triad). At a median followup<br />

of 32 months (range 6.3-52 months), the short-term recurrence<br />

was significantly worse (P=0.0048) in the poor prognostic groups.<br />

This result was similar to using an independent external validation<br />

cohort (n=2100) with long-term follow-up reported before, CMTC<br />

outperformed all other gene signatures in predicting prognosis and<br />

treatment response.<br />

Discussion/conclusion: This prospective validation cohort study<br />

demonstrated reproducibility of CMTC in classifying BCs into the<br />

three major treatment groups and its prognostic significance. CMTC<br />

can be used as a platform to personalize treatments: CMTC-1 BCs<br />

(ER+, low proliferation) in general can be treated with surgery and<br />

tamoxifen alone. CMTC-2 tumours (ER+, high proliferation) will<br />

require additional treatments, including chemotherapy, in addition to<br />

tamoxifen; other biologics can be prescribed based on the activities of<br />

additional oncogenic pathways. Neo-adjuvant chemotherapy should<br />

be considered for CMTC-3 tumours (triple negative and HER2+) with<br />

addition of trastuzumab in those that show activation of the HER2<br />

pathway. CMTC portfolio is being further developed into a genomic<br />

platform to guide personalized BC treatments..<br />

P2-10-35<br />

The grade of accompanying DCIS in IDC as important prognostic<br />

factor<br />

Kim JY, Moon H-G, Han WS, Noh DY. Seoul National University<br />

Hospital, <strong>Breast</strong> <strong>Cancer</strong> Center, Seoul, Korea<br />

Backgrounds: The issue of whether concomitant ductal carcinoma<br />

in situ (DCIS) affects the prognosis of with invasive breast cancer is<br />

important but studies of this have reported inconsistent results. So we<br />

aimed to assess the character and prognostic difference in infiltrating<br />

ductal carcinoma (IDC) according to accompanying DCIS status.<br />

Methods: We reviewed the medical records of the patients who<br />

underwent surgery for IDC. Male patients, patients who had received<br />

neoadjuvant chemotherapy, patients with synchronous bilateral breast<br />

cancer, follow-up periods


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Material and methods<br />

From an original cohort of 569 patients with primary breast cancer,<br />

tissue microarrays were constructed from archival tissue blocks of<br />

primary tumour (n=521), paired lymph node metastasis (n=147) and<br />

biopsies from distant metastasis (n=42). The samples were evaluated<br />

by two independent pathologists. The individual biomarker expression<br />

as well as subtype classification (Luminal A: ER+ and/or PR+, Ki67≤<br />

20% and HER2-. Luminal B: ER+ and/or PR+, Ki67> 20% and/or<br />

HER2+. HER2 type: ER/PR-, HER2+. Basal-like: ER/PR-, HER2-,<br />

CK5/6+ and/or EGFR+) were compared between primary tumour,<br />

lymph node metastasis and distant metastasis. Survival outcome<br />

were estimated using the Kaplan-Meier method and log rank test.<br />

The primary end-point was distant disease-free survival (DDFS).<br />

Results<br />

Primary tumour N (%) Lymph node N (%) Distant metastasis N(%)<br />

ER >10% 454/521 (87%) 117/147 (79%) 33/42 (79%)<br />

PR >10% 330/490 (67%) 85/146 (58%) 23/42 (55%)<br />

HER2+ (SISH) 103/510 (20%) 43/136 (32%) 18/39 (46%)<br />

Ki67 >20% 168/514 (33%) 66/144 (46%) 22/41 (54%)<br />

EGFR+ 85/517 (16%) 28/147 (19%) 9/40 (22%)<br />

CK5/6+ 127/517 (25%) 38/145 (26%) 8/42 (19%)<br />

Distribution of individual biomarker expression.<br />

Primary tumour N (%) Lymph node N (%)<br />

Distant metastasis<br />

N (%)<br />

luminal A 265 (55%) 53 (39%) 9 (23%)<br />

luminal B 160 (33%) 58 (42%) 22 (56%)<br />

HER2 type 18 (4%) 14 (10%) 4 (10%)<br />

Basal-like 33 (7%) 12 (9%) 4 (10%)<br />

Distribution according to subtype.<br />

The molecular subgroups in the primary tumours were associated<br />

with statistically significant prognostic information by log-rank<br />

test (p=0.005) validating luminal A as a subgroup with excellent<br />

prognosis. More detailed survival analysis comparing expression<br />

of biomarkers between primary tumour, lymph node metastasis and<br />

distant metastasis will be presented.<br />

Conclusion<br />

Prognostic information can be obtained by subtype classification using<br />

routine-based biomarker analysis in the primary tumour. Preliminary<br />

data suggest that the subtype distribution is associated with a worse<br />

prognostic profile in the lymph node- and distant metastasis. Detailed<br />

survival analysis will be presented at the meeting.<br />

P2-10-37<br />

Long-term prognosis of early breast cancer in a population-based<br />

cohort with a known BRCA1/2 mutation status.<br />

Nilsson MP, Werner Hartman L, Idvall I, Kristoffersson U, Olsson H,<br />

Johansson O, Borg Å, Loman N. Clinical Sciences, Lund University,<br />

Sweden; Lund University, Sweden; Landspitali University Hospital,<br />

Reykjavik, Iceland<br />

Background:<br />

The impact of germline BRCA1/2 mutations on breast cancer<br />

prognosis and treatment is currently not clear. We investigate how<br />

different factors, including BRCA1/2 mutation status, correlate with<br />

long-term outcome in early breast cancer in a population-based cohort.<br />

Methods:<br />

As previously reported, all women in the Southern Health Care Region<br />

in Sweden with breast cancer diagnosed before age 41 years between<br />

1990 and 1995 (n=262) were contacted in 1996 and offered mutation<br />

analysis of the BRCA1 and BRCA2 genes. Mutation analysis was<br />

performed on 234 of them, the others were excluded from further<br />

studies. 23 pathogenic mutations were found; 18 in BRCA1 and 5 in<br />

BRCA2. We will now present data on tumor and patient characteristics,<br />

treatment and long-term follow-up for the patients from this<br />

population-based cohort. Three patients have declined further study<br />

follow-up and were therefore excluded. Six patients had metastatic<br />

breast cancer at the time diagnosis (one of them a BRCA2 mutation<br />

carrier) and are also excluded, leaving 225 in the present analysis.<br />

Results:<br />

Among the 225 cases estrogen receptor (ER) status is known for<br />

191 (49% ER+, 51% ER-); histologic grade for 169 (24% grade I;<br />

33% grade II; 43% grade III); stage for 225 (36% stage I, 44% stage<br />

II, 20% stage III); type of surgery, adjuvant systemic treatment and<br />

postoperative radiotherapy for 225. In the cohort, 46% received<br />

adjuvant or neoadjuvant chemotherapy, whereas 15% were given<br />

adjuvant endocrine therapy. There is complete follow-up > 10 years<br />

for 205 and present vital status for 224.<br />

Of the 225 women, 128 (57%) have had a local, regional or distant<br />

recurrence of breast cancer (contralateral breast cancer not included)<br />

at the date of last follow-up. One hundred and six patients (47%)<br />

have died, 98 of whom from breast cancer and 2 from presumably<br />

radiation-induced malignancies caused by postoperative radiotherapy.<br />

Thirteen of the 22 BRCA1/2 mutation carriers (59 %) have died, all<br />

from breast cancer. Of the 9 mutation carriers that are alive today, 3<br />

have had an in-breast tumor recurrence and 3 have had a contralateral<br />

breast cancer; the 3 women that have had neither have all performed<br />

bilateral prophylactic mastectomy as well as prophylactic salpingoooforectomy.<br />

Data on how the long-term prognosis of early breast cancer is<br />

correlated to BRCA1/2 mutation status, adjuvant treatment, stage<br />

at diagnosis, histologic grade and ER status of the tumor will be<br />

presented.<br />

Conclusion:<br />

Women with early breast cancer in this cohort from the early 90ies<br />

had a poor long-term prognosis. The prognosis for women diagnosed<br />

with early breast cancer today is likely to be better, thanks to more<br />

modern chemotherapy regimens, endocrine and targeted treatments.<br />

The prognostic significance of BRCA1/2 mutation status will be<br />

further analyzed.<br />

P2-10-38<br />

Prognostic factors uPA/PAI-1: measurement in core needle<br />

biopsies<br />

Vetter M, Landstorfer B, Lantzsch T, Buchmann J, Große R, Ruschke<br />

K, Holzhausen H-J, Thomssen C, Kantelhardt E-J. Martin-Luther<br />

University Halle-Wittenberg, Halle/Saale, Germany; St. Elisabeth &<br />

St. Barbara Hospital, Halle/Saale, Germany; Martha-Maria Hospital,<br />

Halle/Saale, Germany<br />

Background<br />

The urokinase-type plasminogen activator (uPA) and its inhibitor PAI-<br />

1 are validated as prognostic factors at the highest level of evidence<br />

within the node-negative group of breast cancer patients (Harris L et<br />

al., JCO 2007; 25:5287).<br />

Usually, patients receive a core-needle biopsy before surgery. A prior<br />

feasibility study showed that the method of sampling (ex vivo from<br />

the surgical specimen using a small core needle or the larger excision<br />

specimen) doesn’t influence the uPA and PAI-1 results (Thomssen et<br />

al., J Natl <strong>Cancer</strong> 2009; 101:1028). With the current study, we assessed<br />

how well uPA and PAI-1 values obtained from the core needle biopsy<br />

(in vivo) predict the expression level of these prognostic factors in<br />

the later surgical specimen.<br />

Material and Methods<br />

Fresh frozen material of 2-3 core needle biopsies (min 10 mg each)<br />

and the corresponding tumor material (min 50 mg) removed by<br />

subsequent surgery were collected from 110 patients in two hospitals.<br />

Only areas of the surgical specimen were selected for testing that had<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

258s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 2<br />

a sufficient distance to the needle biopsy defect. The uPA and PAI-1<br />

concentrations were analysed using a commercially available ELISA<br />

test (FEMTELLE TM ; American Diagnostica, Inc., Stamford, CT).<br />

The predictive values (and sensitivity/specificity) of the core-needle<br />

biopsy data were calculated to forecast the uPA and PAI-1 values of<br />

the corresponding surgical specimen results.<br />

Results<br />

Sensitivity, specificity, the positive (ppv) and negative (npv) predictive<br />

values for uPA, PAI-1 and for the combination of uPA/PAI-1 are listed<br />

in table 1. Low concentrations of uPA in the core needle biopsies<br />

correlate with low concentrations in the surgical specimens across<br />

the entire study cohort as well as in the group with an intermediate<br />

(G2, N0) risk (npv=0.91).<br />

The clinical and pathological data of the cohort and the risk assessment<br />

will be presented.<br />

Table 1. Performance of uPA/PAI-1 testing in core needle biopsies with regard to forecast surgical<br />

specimen results.<br />

uPA sensitivity specificity ppv npv<br />

all, n=110 0.91 0.65 0.66 0.91<br />

95% C.I. 0.80 - 0.98 0.52 - 0.77 0.63 - 0.77 0.79 - 0.97<br />

intermediate group 0.98 0.63 0.57 0.91<br />

95% C.I. 0.52 - 1.00 0.35 - 0.85 0.29 - 0.99 0.59 - 1.00<br />

PAI-1<br />

all, n=110 0.81 0.60 0.73 0.70<br />

95% C.I. 0.71 - 0.91 0.44 - 0.74 0.61 - 0.83 0.55 - 0.85<br />

intermediate group 0.98 0.63 0.57 0.91<br />

95% C.I. 0.52 - 1.00 0.35 - 0.85 0.29 - 0.82 0.59 - 1.00<br />

uPA/PAI-1<br />

all, n=110 0.89 0.53 0.78 0.72<br />

95% C.I. 0.81 - 0.95 0.38 - 0.67 0.68 - 0.85 0.38 - 0.67<br />

intermediate group 0.92 0.67 0.75 0.89<br />

95% C.I. 0.64 - 1.00 0.35 - 0.90 0.48 - 0.93 0.52 - 1.00<br />

Conclusion<br />

Most important, low uPA/PAI-1 results in core-needle biopsies predict<br />

low results in the surgical specimen. In some cases, core-needle<br />

biopsies and surgical specimen show discordant data. These cases<br />

do not correlate with any clinical or pathological factors such as<br />

time between needling and surgery, grading, tumor size or hospital.<br />

We assume that these discordances are caused by pre-surgical<br />

manipulations, which change expression patterns in the remaining<br />

tissue as it is reported also from multi-gene tests.<br />

P2-10-39<br />

Does E-cadherin or N-cadherin or epithelial-mesenchymal<br />

transition have a probability of clinical implication of the<br />

prognostic marker in invasive ductal carcinoma?<br />

Lee J, Yang G, Paik SS, Chung MS. Hanyang University Medical<br />

Center, Seoul, Republic of Korea<br />

Purpose Epithelial to mesenchymal transition (EMT) is widely<br />

accepted hypothesis of progression and metastasis in carcinoma.<br />

EMT can be explained as a loss of epithelial features and gain<br />

of mesenchymal properties, and in the same manner, it can be<br />

characterized by down-regulation of epithelial cellular markers,<br />

such as E-cadherin (EC) and up-regulation of mesenchymal cellular<br />

markers, such as N-cadherin (NC). <strong>Breast</strong> cancer has a diverse clinical<br />

spectrum, treatments and prognosis according to clinicopathologic<br />

characteristics and immunohistochemical features. Identifying the<br />

presence of EMT also would be able to predict the prognosis and<br />

design better and customized therapy. Our objective was to evaluate<br />

expression of the EC/NC and EMT and correlation between EMT and<br />

clinicopathologic, immunohistochemical features and to investigate<br />

clinical implication of the EC/NC and EMT as prognostic marker<br />

in breast cancer. Methods Using the clinicopathologic data of 480<br />

patients who underwent operation for invasive ductal carcinoma<br />

(IDC) during February 2001 to December 2008. 282 patients with<br />

good quality of paraffin embedded tissue block were selected. The<br />

ER and PR immunohistochemistry was scored using the Allred Score.<br />

EC and NC expression were classified on the basis of intensity of<br />

immunohistochemistry staining. The gain of a mesenchymal marker<br />

or loss of an epithelial marker was interpreted as EMT-positive. In<br />

this study, breast cancer was classified into four types according to the<br />

immunohistochemical expression of ER, PR and HER2 expression<br />

on immunohistochemical analysis and/or with HER2 gene by FISH<br />

analysis: luminal A, luminal B, HER2-enriched, and triple negative<br />

type. Results Median follow-up period was 52.6 months (range 1-113)<br />

and median age was 49 years (range 27-79). Of total 282 cases, 152<br />

(53.9%) cases were classified as luminal A, 32 (11.3%) classified as<br />

luminal B, 43 (15.2%) cases were HER2-enriched and 55 (19.5%)<br />

cases were triple negative type. Expression of a mesenchymal marker,<br />

NC was observed in 22 (7.8%) patients and loss of an epithelial<br />

marker, EC was observed in 57 (20.2%) cases. A total of 75 (26.6%)<br />

were interpreted as EMT-positive. NC score was associated with<br />

HER2-enriched, and triple negative molecular subtype (P = 0.0234).<br />

NC status was associated with HER2 positive status (P =0.0456) and<br />

HER2-enriched, triple negative molecular subtype (P =0.0029). EMT<br />

correlated with ER negative status (P = 0.0199). T stage (P < 0.001),<br />

N stage (P < 0.001) and TNM stage (P < 0.001) and ER (P = 0.0403)<br />

and PR (P = 0.0041) status significantly influenced disease-free<br />

survival (DFS). Age (P = 0.0185) and T stage (P = 0.0017), N stage (P<br />

< 0.001) and ER (P = 0.0048), and PR status (P = 0.0072), molecular<br />

subtype (P = 0.0372), significantly influenced overall survival (OS).<br />

The status of the EC/NC and EMT did not significantly affect DFS<br />

or OS. Conclusion NC status was associated with HER2 positive<br />

status and HER2-enriched, triple negative molecular subtype. And<br />

EMT correlated with ER negative status. EC/NC and EMT status<br />

have relevance to a specific molecular subtype or a probability of<br />

clinical implication of the poor prognostic marker in breast cancer.<br />

P2-10-40<br />

Correlation between expression of the prognostic marker<br />

Progranulin (GP88) with Oncotype Dx Recurrence Score in<br />

estrogen receptor positive breast tumors<br />

Serrero G, Koka M, Goicochea L, Tkaczuk KR, Fernandez KL, Logan<br />

LS, Tuttle K, Yue B, Ioffe OB. A&G Pharmaceutical Inc, Columbia,<br />

MD; University of Maryland Greenebaum <strong>Cancer</strong> Center, Baltimore,<br />

MD; Franklin Square Hospital, Baltimore, MD; Mercy Hospital,<br />

Baltimore, MD<br />

Background:<br />

GP88 (Progranulin, acrogranin) is an 88kDa glycoprotein<br />

overexpressed in breast tumors and involved in their proliferation<br />

and survival. Biological studies have shown that GP88 in ER+ breast<br />

cancer cells was associated with estrogen independence and resistance<br />

to anti-estrogen therapies and aromatase inhibitors. In addition,<br />

GP88 stimulated migration, invasion and angiogenesis, hallmarks of<br />

metastasis. Pathological studies have demonstrated that GP88 was<br />

preferentially expressed in invasive ductal carcinoma while lobular<br />

carcinoma was mostly negative. No expression was found in normal<br />

mammary tissue. Two retrospective studies totaling 530 cases of<br />

ER+ formalin-fixed paraffin-embedded invasive ductal carcinoma<br />

(IDC) established that high GP88 expression was associated with a<br />

4-fold increase in risk of recurrence and 2.5-fold increase in death<br />

when compared to low GP88 scores. Multivariate analysis showed<br />

that GP88 remains a predictor of recurrence even when adjusted for<br />

prognostic factors such as tumor size, grade, stage, lymph node status<br />

and age. High level of GP88 was also found to be elevated in the serum<br />

of breast cancer patients when compared to healthy individuals. Since<br />

GP88 tumor tissue expression appears as a prognostic factor for ER+<br />

breast cancer, we proposed to investigate whether GP88 tumor tissue<br />

www.aacrjournals.org 259s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

expression would correlated to Oncotype Dx® Recurrence Score<br />

which is now for early stage ER+ node negative breast cancer patients.<br />

Methods:<br />

Eight five cases from women ages 37-77 with ER+ invasive<br />

mammary carcinoma from three different institutions and with<br />

an available Oncotype Dx recurrence score were selected with<br />

approval from each site’s IRB. GP88 expression was determined<br />

by immunohistochemistry (IHC) using the anti-human GP88 6B3<br />

monoclonal antibody (A&G) on a Ventana automated staining<br />

platform. For all cases examined, GP88 IHC scores (0, 1+, 2+, 3+)<br />

were compared to routine clinicopathologic factors (tumor size, grade,<br />

and stage), PR, HER2/neu and Ki67 expression (by image analysis)<br />

and to their Oncotype DX® recurrence score (Genomic Health).<br />

The associations of GP88 with the parameters described above were<br />

assessed by t test or ANOVA.<br />

Results:<br />

The GP88 tissue expression correlated with Oncotype Dx Recurrence<br />

score (p


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 2<br />

Results Through 10 years of follow-up, 80 locoregional recurrences<br />

occurred. Patients with a 70-gene signature high risk result (n = 492)<br />

had a locoregional recurrence risk of 8.8% (95%CI: 6.3-11.3) at 5<br />

years and 13.5 % (95%CI: 10.2-16.8) at 10 years, which was 2.6%<br />

(95%CI: 1.2-4.0) at 5 years and 5.7% (95%CI:3.5-7.9) at 10 years<br />

for patients with a low risk 70-gene signature (n = 561)(Logrank:<br />

p10%) were 12 cases (32.4%) which included only<br />

9 TP53 mutations. Five protein-truncating TP53 mutations were all<br />

negative staining of p53. Among BIRC5,BUB1 and PLK1 proteins,<br />

positive staining of PLK1 is the most correlated with TP53 mutations,<br />

sensitivity 0.78, specificity 0.84 and positive predictive value 0.78<br />

(p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Conclusion: These data support the hypothesis that the post<br />

chemotherapy ovary suffers both follicular and stromal dysfunction,<br />

as noted by lower Adione, which is specific to the ovarian stroma.<br />

Adione concentrations in the postmenopausal group women are<br />

similar to published reports of women post oophorectomy. This is the<br />

first longitudinal study we are aware of to evaluate ovarian stromal<br />

function in women undergoing chemotherapy. This total hormone<br />

depletion may be why women experiencing chemotherapy induced<br />

menopause report severe and distressing menopausal symptoms such<br />

as hot flashes. Estrogen is often implicated, but androgen deprivation<br />

in this population should be taken into consideration when planning<br />

interventions to improve health related quality of life.<br />

P2-11-02<br />

Perception and practice of reproductive specialists towards<br />

fertility preservation of young breast cancer patients<br />

Shimizu C, Kato T, Tamura N, Asada Y, Hiroko B, Mizota Y, Yamamoto<br />

S, Fujiwara Y. National <strong>Cancer</strong> Center Hospital, Tokyo, Japan; Asada<br />

Lady’s Clinic, Nagoya, Aichi, Japan; University of Tsukuba, Ibaragi,<br />

Japan; National <strong>Cancer</strong> Center, Tokyo, Japan<br />

Background: The potential for infertility caused by treatment is<br />

one of the important quality-of-life issues in young breast cancer<br />

(YBC) patients. Insufficient communication and partnership between<br />

oncologists and reproductive specialists has been identified as a<br />

major barrier to meeting their needs. However, the perception of<br />

reproductive specialists towards fertility preservation (FP) of YBC<br />

has not been evaluated.<br />

Objective: To investigate the perception and needs of reproductive<br />

specialists towards FP of YBC patients.<br />

Methods: A cross-sectional survey was developed and sent to 423<br />

certified reproductive specialists registered to the Japan Society for<br />

Reproductive Medicine to self-evaluate their perceptions and practices<br />

regarding FP in YBC patients.<br />

Results: Two hundred reproductive specialists (47%) responded to<br />

the survey.<br />

Demographic background of responding reproductive specialists (n=200)<br />

n (%)<br />

Age (year) mean 50 (range 35-71)<br />

Gender male 174 (87)<br />

female 42 (12)<br />

Spouse/ partner yes 190 (95)<br />

no 7 (4)<br />

Experience as a reproductive<br />

mean 25 (range 11-45)<br />

specialist (year)<br />

Experience of management of<br />

cancer patients<br />

yes 192 (96)<br />

no 4 (2)<br />

Affiliation academic hospital 75 (38)<br />

general hospital 42 (21)<br />

private clinic 77 (39)<br />

<strong>Breast</strong> Division in the same<br />

institution<br />

yes 104 (52)<br />

no 92 (46)<br />

99% responded that reproductive specialists should be engaged in<br />

FP of YBC patients. 46% responded that cancer treatment is more<br />

important than childbirth even if the patient was recurrence-free five<br />

years after primary treatment. 83% responded that they would like<br />

to treat YBC patients. Respondents affiliated with private clinics<br />

were more likely to accept both fertilized egg and unfertilized egg<br />

preservation than those affiliated with academic or general hospitals<br />

(p


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 2<br />

higher (worse) ACS and IOC health worry (p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P2-11-06<br />

Mortality among offspring of women diagnosed with breast<br />

cancer; a population based study<br />

Verkooijen HM, Ang X, Liu J, Czene K, Salim A, Hartman M.<br />

University Medical Center Utrecht, Netherlands; National University<br />

of Singapore, Singapore; Karolinska Institute, Stockholm, Sweden;<br />

National University Hospital, Singapore, Singapore<br />

Introduction: Survival after breast cancer has improved substantially<br />

over the past decades. As a result, an increasing number of breast<br />

cancer survivors is starting or extending their family post-diagnosis.<br />

We evaluated mortality risks of offspring from women with a history<br />

of breast cancer.<br />

Methods: From the Swedish Multi-Generation Register and the<br />

<strong>Cancer</strong> Register we identified all 63,205 children whose mother had<br />

ever been diagnosed with invasive breast cancer. For these children,<br />

we calculated relative mortality risks as compared to the background<br />

population (Standardized Mortality Ratios [SMRs]). We compared<br />

SMRs according to the mother’s parity status at diagnosis and timing<br />

of birth in relation to time of diagnosis, i.e. before (>1 year before),<br />

around (between 1 year before and 1 year after) and after (>1 year<br />

after) diagnosis.<br />

Results: Children born around their mother’s breast cancer diagnosis<br />

had an increased mortality risk as compared to the background<br />

population (SMR 2.70 95% CI [1.64-4.44]). This risk was particularly<br />

high for first-born children (SMR 11.07 (95% CI [2.09-27.13]), who<br />

had an absolute excess risk of 6.4% to die before the age of 5 years.<br />

Children born more than one year before or after their mother’s<br />

breast cancer diagnosis had similar mortality risks as the background<br />

population.<br />

Conclusion: Children born around their mother’s breast cancer<br />

diagnosis have significantly increased mortality risks. Offspring of<br />

nulliparous women have a particularly high relative mortality risk,<br />

suggesting that pregnant breast cancer patients may be keen to keep<br />

their first child in spite of potential treatment toxicity.<br />

P2-11-07<br />

Endothelial progenitor cells as novel markers of anthracycline<br />

induced cardiac injury<br />

Lustberg MB, Ruppert AS, Carothers S, Bingman A, McCarthy B,<br />

Raman S, Das M, Kanji S, Lu J, Das H, Cinar-Akakin H, Gurcan<br />

MN, Berger MJ, Wesolowski R, Olson EM, Ramaswamy B, Mrozek<br />

E, Layman RM, Binkley P, Shapiro CL. The OSU <strong>Breast</strong> Program at<br />

Stefanie Spielman Comprehensive <strong>Breast</strong> Center; OSU<br />

Background: Anthracyclines including doxorubicin (DOX) cause<br />

myocardial damage that manifests as either subclinical decrements<br />

of left ventricular ejection function (LVEF) or overt cardiomyopathy.<br />

LVEF changes and cardiac risk factors are insufficient predictors<br />

of future DOX cardiotoxicity. Bone marrow derived endothelial<br />

progenitor cells (EPCs) are mobilized and are homed to sites of<br />

myocardial injury to help with repair of damaged myocardium. We<br />

hypothesized that EPC levels would be indicative of early DOX<br />

cardiotoxicity. Hence, we prospectively collected serial blood samples<br />

to evaluate functional EPCs, Troponin I (Ti) and B-natriuretic peptide<br />

(BNP), in patients (pts) receiving DOX-based chemotherapy.<br />

Methods: Eligible pts were initiating adjuvant DOX for early stage<br />

breast cancer. Pts underwent cardiac magnetic resonance (CMR), Ti,<br />

BNP, and EPC at baseline, after 1 cycle of DOX, and after completion<br />

of DOX. CD133+ progenitor cells were isolated from the peripheral<br />

blood mononuclear cells (PBMC) using AutoMACS (automated<br />

magnetic cell sorting, Miltenyi Biotech). In vitro colony forming unit<br />

(CFU) assay was performed for isolated CD133+ progenitor cells<br />

on MethoCult (Stemcell Technology). After 8 days of culture, EPC<br />

colonies were counted using a two-step image analysis algorithm.<br />

Repeated measures analysis of variance modeled changes in cardiac<br />

markers over time. Logistic regression was used to correlate variables<br />

with abnormal Ti.<br />

Results: Forty two women were enrolled. The average age was 52<br />

years (range 33-68) and stage distribution was I (14%), II (58%) and<br />

III (28%). All but one patient received peg-fligrastim after DOX.<br />

Thirty six pts had EPC/cardiac biomarkers and twenty nine pts had<br />

CMRs at all three time points. LVEF decreased 1.6% following<br />

completion of DOX (95% CI: -3.8 to 0.6, p=0.16). There was a<br />

non-linear trend in EPCs over time (p=0.05), with an initial increase<br />

followed by a decrease, with average values of 59 (95% CI: 50-70),<br />

65 (95% CI: 55-75), and 50 (95% CI: 40-60), respectively, across the<br />

three time points. By the end of treatment, 54% (95% CI: 0.37-0.71)<br />

of women had abnormal troponins (median: 0.03, range: 0.02 to<br />

0.17). Variables associated with abnormal troponins included lower<br />

baseline EPCs (p=0.095), older age (p=0.075) and initial increase in<br />

BNP post cycle 1 (p


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 2<br />

Results: Accrual occurred during 42 clinic days between April 2011<br />

and February 2012. During that time, there were 129 postoperative<br />

appointments for patients with a diagnosis of breast cancer. 54 did not<br />

meet the eligibility requirements; 46 were DCIS, three had metastatic<br />

cancer, and five did not speak English. Of the patients who met the<br />

eligibility requirements 100% agreed to participate. Characteristics<br />

are shown in Table 1.<br />

Table 1 Enrollment Characteristics of Participants (N=73*)<br />

Age (years) 56.0 (12.7)<br />

Ethnicity (%)<br />

Non-Hispanic white 80.8<br />

African-American 4.1<br />

Hispanic 4.1<br />

Other 11.0<br />

Disease stage (%)<br />

Stage I 46.6<br />

Stage II 43.8<br />

Stage III 9.6<br />

Additional Treatment (%)<br />

None 15.9<br />

Radiation only 30.4<br />

Chemotherapy only 13.0<br />

Radiation and Chemotherapy 40.6<br />

Hormone Therapy (%)<br />

No 24.6<br />

Yes 75.4<br />

Treatment Location (%)<br />

Smilow <strong>Cancer</strong> Center 90.4<br />

Other 9.6<br />

*After further inquiry to participants were found to be pathologically DCIS and were therefore not<br />

included in analysis<br />

Only 39.4% of participants received both chemotherapy and radiation<br />

and 9.6% of our participants received their adjuvant therapy outside<br />

of Smilow <strong>Cancer</strong> Center.<br />

Conclusion: Women recently diagnosed with breast cancer are<br />

interested in receiving survivorship care plans after treatment, as<br />

demonstrated by 100% accrual rate of eligible patients approached<br />

in the postoperative visit. The postoperative visit in a surgical clinic<br />

may provide the starting point for tracking a patient through treatment.<br />

P2-11-09<br />

Weight Change and Risk of Incident Diabetes After <strong>Breast</strong> <strong>Cancer</strong><br />

Erickson KD, Patterson RE, Natarajan L, Lindsay SP, Heath D, Caan<br />

BJ. Moores UC <strong>San</strong> Diego <strong>Cancer</strong> Center, University of California,<br />

<strong>San</strong> Diego, La Jolla, CA; <strong>San</strong> Diego State University, <strong>San</strong> Diego, CA;<br />

Kaiser Permanente Northern California, Oakland, CA<br />

Weight gain after breast cancer treatment is common and may increase<br />

risk of diabetes. This analysis examined the association between<br />

weight change pattern and incident diabetes, adjusting for pre- and<br />

post-diagnosis BMI in breast cancer survivors (n=1,617) participating<br />

in the Women’s Healthy Eating & Living (WHEL) Study. Weight at 1<br />

year pre-diagnosis and ∼2 years post-diagnosis were used to calculate<br />

pre- to post-diagnosis body weight percentage change (categorized<br />

as: loss (>5%), stable (± 5%), moderate gain (>5% to


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

survivors, with a specific impact on PM. Women who complain of<br />

‘chemo fog’ should be evaluated carefully for PM deficits and their<br />

concerns should be acknowledged. An important finding from this<br />

study is the demonstration that fatigue is associated with memory<br />

deficits when memory as used in everyday life is evaluated. This<br />

finding which is suggestive of a common mechanism involved in<br />

fatigue symptoms and everyday memory disturbances raises the<br />

possibility that interventions targeted at improving fatigue may also<br />

improve memory functioning and quality of life in EBC survivors.<br />

Further longitudinal studies should be conducted to critically evaluate<br />

the role of chemotherapy by assessing PM before the initiation of<br />

chemotherapy and in EBC patients receiving only adjuvant hormonal<br />

therapy.<br />

P2-11-11<br />

Patient Reported Outcomes after <strong>Breast</strong> <strong>Cancer</strong> Surgery and<br />

Adjuvant Therapy from a German <strong>Breast</strong> <strong>Cancer</strong> Centre<br />

Feiten S, Dünnebacke J, Heymanns J, Köppler H, Thomalla J, van<br />

Roye C, Wey D, Weide R. Institute for Health Care Research in<br />

Oncology, Koblenz, Germany; Catholic Clinical Centre Koblenz-<br />

Montabaur, Koblenz, Germany; Oncology Group Practice Koblenz,<br />

Germany<br />

Objectives: Evaluation of the subjectively experienced physical,<br />

psychological, social and job-related consequences of breast cancer.<br />

Methods: Standardized paper pencil interview of patients with the<br />

initial diagnosis of breast cancer who had their primary surgery<br />

between 01/2006 and 12/2010 at an accredited breast cancer centre<br />

followed by systemic adjuvant treatment. The data collection was<br />

conducted with the help of a self-developed scanner-readable<br />

questionnaire which had been evaluated in a pretest.<br />

Results: 1260 patients were contacted, 871 completed questionnaires<br />

(return rate 72%) were analyzed. Median age of the patients (99.5%<br />

women) at the time of the interview was 65 years (30 - 91). 6%<br />

relapsed during the observation period. 91% were “satisfied” or<br />

“very satisfied” with the surgical result. 67% indicated a complete<br />

freedom of pain. 23% received lymphatic drainage at the time of the<br />

questioning (11/2011), 33% complained about limitations of arm<br />

and / or shoulder function. 76% received anti-hormonal therapy,<br />

13% stopped the anti-hormonal medication prematurely. Patients<br />

received a mean of 1.3 different anti-hormonal therapies, 54% took<br />

Tamoxifen. Psychological distress, cognitive limitations and physical<br />

consequences were rated on a scale from 1 – “not at all” to 4 – “very<br />

much”. The highest average values were found for the items sleep<br />

disturbances (2.3) and exhaustion (2.3), the lowest for depression (1.7)<br />

and word-finding difficulties (1.8). After therapy only 39% described<br />

a complete recovery of their physical capacity, 62% regained their<br />

previous mental capacity. 44% were in employment before their<br />

disease. 67% returned to their workplace but only 65% of them<br />

with their previous number of hours. 15% indicated disadvantages<br />

in their workplace due to the breast cancer disease. For 75% their<br />

partnership did not change, 12% experienced a deterioration, 13%<br />

an improvement. Before the illness 9% consulted a psychiatrist/<br />

psychotherapist, after the illness 18%. Before the diagnosis of breast<br />

cancer 13% received antipsychotic drugs, after the disease 25%.<br />

Conclusions: <strong>Breast</strong> cancer diagnosis and therapy leads to long<br />

lasting impairment of physical, psychological, social and job-related<br />

functioning in a significant number of patients.<br />

P2-11-12<br />

Proactive approach to risk reduction and early detection of breast<br />

cancer related lymphedema<br />

Fu MR, Guth A, Kleinman R, Cartwright F, Haber J, Axelrod D. New<br />

York University, New York, NY; NYU <strong>Cancer</strong> Center, New York, NY<br />

Advances in surgery and post-operative treatment continue to<br />

reduce the incidence of late effects of cancer treatment such as<br />

lymphedema. Yet, many breast cancer survivors still face considerable<br />

post-operative challenges as a result of developing lymphedema.<br />

The purpose of this study was to preliminarily evaluate a nursing<br />

educational and behavioral program “The Optimal YOU” to enhance<br />

lymphedema risk reduction, focusing on preventing inflammationinfection,<br />

promoting lymph drainage, and maintaining optimal Body<br />

Mass Index (BMI) as well as preoperative and subsequent follow-up<br />

assessment of limb volume changes and symptoms.<br />

A quasi-experimental design with repeated-measures was used.<br />

The Study outcomes included lymph volume changes by infrared<br />

perometer, lymphedema-related symptoms, and BMI by<br />

Bioimpedance at pre-operative baseline, post-operative, 6-month,<br />

and 12-month follow-up. A total of 147 patients were recruited and<br />

received the program, 141 patients completed the study (4% attrition<br />

rate).<br />

Majority (over 90%) of patients maintained preoperative limb volume<br />

and improved their symptom experience at 12-month follow-up.<br />

During the study period, only 7 patients (5%) had measurable<br />

lymphedema. At 12-month follow-up, among the 7 patients who<br />

developed measurable lymphedema, 2 patients’ limb volume<br />

returned to pre-operative level without compression therapy only<br />

by maintaining exercises of The Optimal YOU to promote daily<br />

lymph flow. About 80% of patients maintained or improved their<br />

preoperative BMI.<br />

Implementation of this nursing educational and behavioral<br />

intervention is effective to enhance lymphedema risk reduction and<br />

early detection. Findings of the study provided evidence to support<br />

the emerging policy change in lymphedema care, shifting treatmentfocus<br />

care to proactive risk reduction.<br />

P2-11-13<br />

The Effect of Positive Axillary Lymph Nodes on Symptoms,<br />

Physical Impairments, and Function<br />

Kesarwala AH, Pfalzer LA, O’Meara WP, Stout NL. National <strong>Cancer</strong><br />

Institute, Bethesda, MD; University of Michigan - Flint, MI; Lahey<br />

Clinic, Burlington, MA; Walter Reed National Military Medical<br />

Center, Bethesda, MD<br />

Purpose/Objective(s): The role of axillary lymph node (ALN)<br />

sampling in breast cancer (BC) treatment continues to evolve, and<br />

BC patients are recommended for post-operative regional nodal<br />

radiation therapy (RNRT) based on the number of positive ALN.<br />

RNRT is recommended for patients with 4 or more positive ALN,<br />

but it remains controversial in patients with 1-3 positive ALN and is<br />

rarely recommended for patients without positive ALN. Consideration<br />

of anticipated functional impairments often guides decision making.<br />

The purpose of this analysis is to investigate functional impairments<br />

in BC patients with varying numbers of positive ALN.<br />

Materials/Methods: 166 women were diagnosed with BC between<br />

2001-05 and enrolled and treated in a prospective surveillance<br />

physical therapy program. 110 had zero positive ALN, 37 had<br />

1-3 positive ALN, and 19 had 4 or more positive ALN on either<br />

sentinel LN biopsy or ALN dissection. Participants’ upper extremity<br />

(UE) range of motion, strength, and limb volume were assessed<br />

pre-operatively and at 1, 3, 6, 9, and 12+ months post-operatively<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

266s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 2<br />

by a physical therapist. Limb volume was assessed using infrared<br />

optoelectronic perometry. At 12+ months, overall health status, UE<br />

symptoms and function, and physical activity levels were reported<br />

using standardized questionnaires. Chi-square tests and one-way<br />

ANOVA analyses were used to determine significance between<br />

groups (p≤0.05).<br />

Results: Of these 166 patients, 94 received mastectomy and 72<br />

received lumpectomy, while 41 received RNRT and 58 received whole<br />

breast tangent RT. No significant differences were found between<br />

groups with regard to age or race. The number of dissected LN was not<br />

significantly different between those patients with 1-3 positive ALN<br />

and 4 or more positive ALN. Rates of lymphedema and seroma were<br />

not significantly different between those patients with zero positive<br />

ALN and 1-3 positive ALN, and rates of cording were not significantly<br />

different between any of the groups. Increased lymphedema (p=0.03)<br />

and seroma (p=0.005) were seen in those patients with 4 or more<br />

positive ALN compared to those patients with zero positive ALN,<br />

but this may also be related to a significantly greater number of<br />

dissected LN in the former group. By 12+ months post-operatively,<br />

there were no differences in shoulder abduction, shoulder flexion,<br />

internal rotation, or external rotation between groups. No differences<br />

were seen between groups in self-reported fatigue, UE swelling or<br />

weakness, arm stiffness, or ability to climb stairs.<br />

Conclusions: Functional impairments represent an important category<br />

of morbidity for BC survivors and should be considered in pretreatment<br />

decision making. The number of positive ALN may not<br />

correlate with increased impairment over the first year of treatment<br />

when a prospective surveillance physical therapy program is part of<br />

the plan of care. Additional research is needed to assess longer-term<br />

changes and the impact of axillary surgery and/or radiation in the<br />

context of aggregate effects of other BC treatment modalities.<br />

P2-11-14<br />

Symptoms, Physical Impairments, and Function in <strong>Breast</strong> <strong>Cancer</strong><br />

Patients with Negative Axillary Lymph Nodes<br />

Kesarwala AH, Pfalzer LA, O’Meara WP, Stout NL. National <strong>Cancer</strong><br />

Institute, Bethesda, MD; University of Michigan - Flint, MI; Lahey<br />

Clinic, Burlington, MA; Walter Reed National Military Medical<br />

Center, Bethesda, MD<br />

Purpose/Objective(s): <strong>Breast</strong> cancer (BC) patients are recommended<br />

for post-operative regional nodal radiation therapy (RNRT) based<br />

on the number of positive axillary lymph nodes (LN). While RNRT<br />

is recommended for patients with 4 or more positive LN, it remains<br />

controversial in patients with 1-3 positive LN. For these patients,<br />

consideration of anticipated functional impairments often guides<br />

decision making, but these considerations are confounded by the<br />

inseparable effects of disease in and treatment of the axilla. The<br />

purpose of this analysis is to investigate the effect of various therapies<br />

on functional impairments in BC patients without axillary disease.<br />

Materials/Methods: 166 women were diagnosed with BC between<br />

2001-05 and enrolled and treated in a prospective surveillance<br />

physical therapy program. 110 had zero positive axillary LN on<br />

either sentinel LN biopsy or axillary LN dissection and were analyzed<br />

for this report. Participants’ upper extremity (UE) range of motion,<br />

strength, and limb volume were assessed pre-operatively and at 1,<br />

3, 6, 9, and 12+ months post-operatively by a physical therapist.<br />

Limb volume was assessed using infrared optoelectronic perometry.<br />

At 12+ months, overall health status, UE symptoms and function,<br />

and physical activity levels were reported using standardized<br />

questionnaires. Chi-square tests and one-way ANOVA analyses were<br />

used to determine significance between groups (p≤0.05).<br />

Results: Of these 110 patients, 34 received mastectomy without RT, 21<br />

received mastectomy with RNRT, 10 received lumpectomy alone, and<br />

45 received lumpectomy with whole breast tangent RT. No significant<br />

differences were found between groups with regard to stage, ER/PR<br />

status, and number of dissected LN. Rates of lymphedema, cording,<br />

and seroma were not significantly different between groups. By<br />

12+ months post-operatively, there were no differences in shoulder<br />

abduction, shoulder flexion, internal rotation, or external rotation<br />

between groups. No differences were seen between groups in selfreported<br />

fatigue, UE swelling or weakness, arm stiffness, or ability<br />

to climb stairs.<br />

Conclusions: Functional impairments represent an important<br />

category of morbidity for BC survivors and should be considered in<br />

pre-treatment decision making. In patients without axillary disease,<br />

post-operative RNRT or whole breast tangent RT may not contribute<br />

significantly to impairment over the first year of treatment when a<br />

prospective surveillance physical therapy program is part of the plan<br />

of care. Additional research is needed to assess longer-term changes<br />

and the impact of radiation in the context of the aggregate effect of<br />

disease burden combined with other BC treatment modalities.<br />

P2-11-15<br />

Development of a web-based survey tool to assess change in<br />

breast cancer (BrCa) survivor knowledge after receipt of cancer<br />

treatment summary and survivorship care plan (SCP).<br />

Custer JL, Rocque GB, Wisinski KB, Jones NR, Donohue S, Koehn<br />

TM, Champeny TL, Terhaar AR, Chen KB, Peck KA, Tun MT,<br />

Wiegmann DA, Sesto ME, Tevaarwerk AJ. University of Wisconsin,<br />

Madison, WI<br />

Intro: The Institute of Medicine advocates survivorship care plans<br />

(SCPs) as tools to improve coordination of care by improving survivor<br />

knowledge of follow-up recommendations and future risks. No<br />

evidence exists to demonstrate that SCPs impact survivor knowledge<br />

of diagnosis, treatment, or future/chronic side effects. Furthermore,<br />

there is a lack of information on existing surveys and their ability to<br />

assess survivor knowledge regarding these issues, without change<br />

over time. The purpose of this research is to report on the development<br />

of a survey assessing knowledge of diagnosis, treatment, and side<br />

effects in BrCa survivors.<br />

Methods:Using existing literature, two oncologists created 24<br />

questions addressing knowledge of diagnosis, treatment, and<br />

side effects. Content experts including breast oncology providers<br />

(representing multiple subspecialties), Survey Research Shared<br />

Service (SRSS) and patient advocates reviewed and revised the<br />

questions. Next, potential questions were administered in a group<br />

setting to BrCa survivors to evaluate clarity of instructions and survey<br />

wording. The <strong>Breast</strong> <strong>Cancer</strong> Knowledge (BreaCK) survey was further<br />

revised based on survivor feedback.<br />

For pilot testing, BrCa survivors were recruited from clinic to test<br />

BreaCK survey content and clarity. Survey 1 was administered in<br />

clinic online. SRSS conducted verbal assessments regarding content<br />

after Survey 1. Four weeks later, survivors received Survey 2 via<br />

email and answered online. Correct answers were abstracted from<br />

the medical record.<br />

Results: Nine subjects completed both surveys. Qualitatively, little<br />

intra-subject variation was seen between surveys. Subjects did not feel<br />

that the survey was burdensome or intrusive. No subject was able to<br />

correctly answer all questions. Final survey adjustments were made<br />

based on subject feedback and common incorrect answers encountered<br />

when grading the surveys. Specifically, subjects had difficulty<br />

www.aacrjournals.org 267s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

understanding “endocrine or hormone therapy.” Furthermore, subjects<br />

reported guessing in response to some questions – additional answer<br />

categories were added, including “I don’t know.”<br />

Conclusion: Survivor knowledge did not change significantly between<br />

surveys. This suggests survivor knowledge was not impacted by<br />

the survey over the four-week interval. The revised BreaCK survey<br />

may be a useful tool for assessing survivor knowledge of diagnosis,<br />

treatment and side effects. A larger cohort of BrCa survivors is being<br />

recruited, starting Summer 2012, and will be evaluated using the<br />

survey.<br />

P2-11-16<br />

Cardiac Morbidity After Adjuvant Chemotherapy (CT) for Early<br />

<strong>Breast</strong> <strong>Cancer</strong> in the Community Setting.<br />

Patt D, Espirito J, Turnwald B, Denduluri N, Wang Y, Lina A,<br />

Hoverman R, Neubauer M, Bosserman L, Busby L, Brooks B,<br />

Cartwright T, Sitarik M, Schnadig I, Winter W, Garey J, Ginsburg-<br />

Arlen A, Bergstrom K, Beveridge R. Pathways Task Force, US<br />

Oncology Network, McKesson Specialty Health, Austin, TX; Pathways<br />

Task Force, US Oncology Network, McKesson Specialty Health,<br />

The Woodlands, TX; Pathways Task Force, US Oncology Network,<br />

McKesson Specialty Health, Arlington, VA; US Oncology Network,<br />

McKesson Specialty Health, The Woodlands, TX; Kansas City <strong>Cancer</strong><br />

Center, Pathways Task Force, US Oncology Network, McKesson<br />

Specialty Health, Overland Park, KS; Pathways Task Force, US<br />

Oncology Network, McKesson Specialty Health, Rancho Cucamonga,<br />

CA; Rocky Mountain <strong>Cancer</strong> Centers, Pathways Task Force, US<br />

Oncology Network, McKesson Specialty Health, Boulder, CO;<br />

Pathways Task Force, US Oncology Network, McKesson Specialty<br />

Health, Dallas, TX; Pathways Task Force, US Oncology Network,<br />

McKesson Specialty Health, Ocala, FL; Pathways Task Force, US<br />

Oncology Network, McKesson Specialty Health, Tualatin, OR;<br />

Pathways Task Force, US Oncology Network, McKesson Specialty<br />

Health, Portland, OR<br />

Cardiac morbidity after exposure to CT is a known and previously<br />

described risk. Anthracycline exposure can be complicated by acute<br />

or chronic cardiac toxicity. Trastuzumab exposure is largely associated<br />

with acute and reversible cardiac morbidity.<br />

Methods: We retrospectively queried the electronic health record<br />

(EHR) from our network of community oncology practices,<br />

iKnowMed, for patients (pts) diagnosed with Stage I-III breast cancer<br />

(BC) from 2007-2010 with at least 5 visits and follow-up (f/u) through<br />

2012. We stratified this group by CT utilization (yes/no), regimen type,<br />

age, and characterized the incidence of cardiac disease or initiation of<br />

cardiac medication through the f/u period to determine the association<br />

of cardiac disease or treatment with CT utilization. Cardiac diseases<br />

analyzed included congestive heart failure, valvular and ischemic<br />

heart disease, arrythmias, and hypertension. Cardiac medications<br />

included beta blockers, angiotensin-converting-enzyme inhibitors,<br />

angiotensin receptor II blockers, loop and thiazide diuretics. Hazard<br />

ratios by prespecified risk parameters were then analyzed by<br />

multivariate analysis for all pts who did not have cardiac disease<br />

preceding their diagnosis of BC.<br />

Results: We identified 20,900 pts with a median f/u of 3.2 yrs (1.4-<br />

5.4). 11,295 (54%) pts received adjuvant CT and 9,605 (46%) did<br />

not. Median age at diagnosis in the CT-treated arm and non CTtreated<br />

arm was 54 and 64 yrs, respectively (p < 0.0001). Among<br />

both the non-CT and CT-treated group, the baseline prevalence of<br />

cardiac disease was 14%. Among the CT-treated group, 3475 pts or<br />

31% (95% CI, 30 % - 32%) had or developed cardiac disease within<br />

the study period. In the non-CT group, 3790 pts or 39% (95% CI,<br />

38% - 40%) had or developed cardiac disease with the study period<br />

(p


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 2<br />

were noted for all other cardiorespiratory fitness testing outcomes.<br />

Significant improvements from baseline to 17 weeks were seen across<br />

both groups for all measures of body composition. Lean body mass<br />

did not significantly change from baseline to 17 weeks in both groups.<br />

There were no significant between group differences for change in<br />

body composition.<br />

Cardiorespiratory fitness at baseline and 17 weeks [mean (SD)]<br />

Variable Baseline 17 Weeks Change<br />

Withing Between group<br />

group p-value p-value<br />

Standard Exercise Group<br />

(n=9) VO2 Max (mL. 24.0 (7.1) 26.3 (6.8) 2.3 (2.9) 0.05<br />

kg-1.min-1)<br />

Structured Exercise<br />

Group (n=7)<br />

VO2max(mL.kg-1.<br />

24.6 (2.9) 29.8 (3.8) 5.3 (2.6) 0.002 0.05<br />

min-1)<br />

Standard Exercise<br />

Group (n=9) Treadmill 677.2 (319.6) 813.7 (271.2) 136.4 (101.9) 0.02<br />

Duration (seconds)<br />

Structured Exercise<br />

Group (n=7) Treatdmill 794.0 (131.3) 1051.4 (68.2) 257.4 (87.1) 0.02 0.02<br />

Duration (seconds)<br />

Standard Exercise<br />

Group (n=9) Maximum<br />

Achieved Heartrate<br />

167.4 (8.4) 166.5 (8.2) -1.0 (7.1) 0.71<br />

(bpm)<br />

Structured Exercise<br />

Group (n=7) Maximum<br />

Achieved Heartrate<br />

(bpm)<br />

171.3 (11.5) 175.4 (9.8) 4.1 (7.7) 0.21 0.21<br />

Conclusion: In this study, both standard exercise and structured<br />

exercise improved CR fitness measured by VO2max, but the structured<br />

exercise group experienced significantly greater improvements.<br />

P2-12-01<br />

Prospective Evaluation of Joint Symptoms in Postmenopausal<br />

Women Initiating Aromatase Inhibitors for Early Stage <strong>Breast</strong><br />

<strong>Cancer</strong><br />

Crew KD, Chehayeb Makarem D, Awad D, Kalinsky K, Maurer M,<br />

Kranwinkel G, Brafman L, Fuentes D, Hershman DL. Columbia<br />

University Medical Center, New York, NY<br />

Background: Aromatase inhibitors (AIs) are widely prescribed to<br />

postmenopausal women for the adjuvant treatment of hormonesensitive<br />

breast cancer (BC). However, musculoskeletal complaints<br />

can lead to nonadherence and early discontinuation. The aim of<br />

this study was to characterize the natural history of the AI-induced<br />

arthralgia syndrome and determine predictors of worsening symptoms.<br />

Methods: Postmenopausal women with stage I-III BC initiating<br />

adjuvant AI therapy were enrolled. All patients completed the<br />

following questionnaires at baseline and every 3 months for a year:<br />

Modified Brief Pain Inventory-Short Form (BPI-SF), Western Ontario<br />

and McMaster Universities Osteoarthritis (WOMAC) index and the<br />

Modified Assessment of Chronic Rheumatoid Affections of the Hands<br />

(M-SACRAH). Higher scores reflect worse symptoms. Quality of life<br />

was assessed using the Functional Assessment of <strong>Cancer</strong> Therapy-<br />

Endocrine Subscale (FACT-ES). Hand grip strength was measured at<br />

each visit with a Martin dynamometer. Paired t-tests were performed<br />

to compare follow-up evaluations to baseline. Linear Regression was<br />

performed to evaluate the association between baseline symptoms<br />

and change in symptoms.<br />

Results: A total of 169 patients have been consented, 3-month data<br />

is available on 102; 6-month data on 85. Median age: 63 (42-89);<br />

White/Black/Asian/Hispanic: 61.48/30.33/3.28/25.6; median BMI<br />

(kg/m2): 28 (12-50). Compared to baseline, there was a statistically<br />

significant increase in BPI pain severity and endocrine related<br />

symptoms on the FACT-ES at 3 and 6 months. Significant changes in<br />

the BPI pain interference, M-SACRAH pain and stiffness, WOMAC<br />

function, physical well-being on the FACT-ES, trial outcome index<br />

and pinch grip strength were seen at 3 months; however these changes<br />

did not remain significant at 6 months. Logistic regression models<br />

evaluating predictors of patient reported outcome measures and grip<br />

strength were performed. Baseline score was the strongest predictor<br />

of worsening symptoms (p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

but only the FACT-B social wellbeing was statistically significant<br />

at 3 months with a p=0.0164. Changes in other FACT-B and SF-36<br />

scores were not significantly different between exercise and usual care<br />

groups. There were significant improvements at 6 months in weight<br />

(p=0.0192), % body fat (p=0.0337), max strength (p=0.0029), and<br />

waist circumference (0.0359) and at 12 months in weight (p=0.0293),<br />

% body fat (p=0.0481), max strength (p=0.0097) and endurance<br />

(p=0.0037) in the exercise group compared to usual care.<br />

CONCLUSIONS: This randomized prospective study suggests<br />

benefit of exercise during chemotherapy. This benefit continued<br />

6 months beyond the completion of the exercise program with<br />

significant improvement in physical function, body composition,<br />

strength and endurance with no decline in quality of life. Regular<br />

moderate exercise may play an important role in improving function<br />

during adjuvant chemotherapy and should be further studied in a<br />

large randomized trial.<br />

P2-12-04<br />

Music Therapy Reduces Radiotherapy-Induced Fatigue in<br />

Patients with <strong>Breast</strong> or Gynecological <strong>Cancer</strong>: A Randomized<br />

Trial<br />

Freitas NMA, Silva TRMA, Freitas-Junior R, Paula Junior W,<br />

Silva DJ, Machado GDP, Ribeiro MKA, Carneiro JP. Araujo Jorge<br />

Hospital/ACCG, Goiania, Goias, Brazil; Federal University of<br />

Goias, Goiania, Goias, Brazil; Instituto Integrado de Neurociencias/<br />

IINEURO, Goiania, Goias, Brazil<br />

BACKGROUND: Fatigue is a frequent side effect of radiotherapy,<br />

and may interfere with self-esteem, social activities, and quality of<br />

life. The authors investigated the influence of music therapy (MT)<br />

on the reduction of fatigue in women with breast or gynecological<br />

malignant neoplasia during radiotherapy. METHODS: Randomized<br />

controlled trial (control group-CG and music therapy group-MTG)<br />

to assess fatigue, quality of life using the Functional Assessment of<br />

<strong>Cancer</strong> Therapy (FACT-F and FACT-G) version 4 and symptoms<br />

of depression using Beck Depression Inventory (BDI) in women<br />

undergoing radiotherapy in three distinct moments (during the first<br />

and the last week of radiotherapy, and on the week of the intermediary<br />

phase). Individual 30–40 minute sessions of music therapy were<br />

offered to participants of the MTG. RESULTS: In this study, 164<br />

women were randomized and 116 (63 CG and 53 MTG) were included<br />

in the analyses, with mean age of 52.9 years (CG) and 51.85 years<br />

(MTG). Participants in the MTG had an average of 10 MT sessions,<br />

during the study. FACT-F results were significant regarding Trial<br />

Outcome Index (TOI), synthesis of the Physical and functional wellbeing<br />

areas of FACT-F (p = 0.011), FACT-G (p = 0.005), FACT-F<br />

(p = 0.001) for MTG compared with CG. The depressive symptons<br />

were reduced to the minimum levels in the MTG (p = 0,005) with<br />

risk reduction of 74% (RR = 0,26; CI 0,10 - 0,70).<br />

Comparative analysis of categories (chi-square) during radiotherapy according to Beck Depression<br />

Inventory (BDI)<br />

Variable CG MTG P RR (CI 95%)<br />

N % N %<br />

BDI - Initial<br />

phase<br />

Minimum 36 65 34 64 0.88 1.00<br />

Mild to<br />

intense<br />

9 16 10 19<br />

BDI - Final<br />

phase<br />

Minimum 35 64 46 87 0.005 1.00<br />

Mild to<br />

intense<br />

11 20 5 9<br />

0.97 (0.65-<br />

1.44)<br />

0.26 (0.10-<br />

0.70)<br />

CG: Control Group; MTG: music therapy group; RR: relative risk; CI: confidence interval<br />

CONCLUSIONS: Individual MT sessions are effective to reduce<br />

fatigue related to cancer and symptoms of depression, as well as to<br />

improve quality of life for women with breast or gynecological cancer<br />

undergoing radiotherapy.<br />

P2-12-05<br />

Limited Absorption of Low Dose 10µg Intravaginal 17-β Estradiol<br />

(Vagifem®) in Postmenopausal Women with <strong>Breast</strong> <strong>Cancer</strong> on<br />

Aromatase Inhibitors<br />

Goldfarb SB, Dickler M, Dnistrian A, Patil S, Dunn L, Chang K,<br />

Berkowitz A, Tucker N, Carter J, Barakat R, Hudis C, Castiel M.<br />

Memorial Sloan-Kettering <strong>Cancer</strong> Center, New York, NY<br />

Background: Aromatase inhibitors (AIs) are used to treat<br />

postmenopausal women with hormone-receptor positive (HR+) breast<br />

cancer (BC). AIs block the peripheral conversion of androgens to<br />

estrogen (E), resulting in sub-physiologic levels of E that may lead<br />

to profound urogenital atrophy. Atrophic vaginitis in BC survivors<br />

is prevalent and its management is complex. An observational study<br />

(n=6) demonstrated elevated estradiol levels in 5/6 women on AIs after<br />

2 weeks (wks) of treatment with 25µg 17-β Estradiol (Vagifem ®),<br />

which raised questions regarding the safety of intravaginal estradiol<br />

in women with HR+ BC. However, the small sample size limited<br />

definitive conclusions, yet underscored the need for a clinical trial to<br />

evaluate concurrent use of AIs and intravaginal estradiol. In addition,<br />

a lower dose 17-β Estradiol (10µg) is now available and effectively<br />

treats healthy women with atrophic vaginitis. We hypothesized that<br />

the 10µg dose is effective and may have less systemic absorption<br />

than the 25µg dose. This is the first study to evaluate the 10µg dose<br />

in BC pts on AI therapy.<br />

Methods: A prospective longitudinal IRB-approved study was<br />

performed at MSKCC in postmenopausal women with stage I-III<br />

HR+ BC on adjuvant letrozole or anastrozole for at least 3 months<br />

and had urogenital atrophy. Patients on exemestane were not eligible<br />

due to cross-reactivity with the assay. All women were initiated on<br />

10µg intravaginal 17- β estradiol (Vagifem®). Serial estradiol/FSH<br />

levels were measured at baseline and wks 2, 7, 12, 18 & 24; we used<br />

a highly sensitive estradiol radioimmunoassay, ESTR-US-CT, from<br />

Cisbio US, Inc. Estradiol/FSH levels were checked approximately 12<br />

hrs after insertion, chosen to measure peak absorption. The primary<br />

endpoint was change in systemic estradiol level from baseline to<br />

wk 12. Patients also completed the Female Sexual Function Index<br />

(FSFI) and Menopausal Symptom Checklist (MSCL) at baseline<br />

and wks 12 & 24.<br />

Results: 26 pts have been treated and 18 are currently evaluable for<br />

the primary endpoint at wk 12. Wilcoxon signed rank test showed<br />

no statistically significant difference between baseline and wk 12<br />

estradiol levels (p= 0.49) or FSH levels (p= 0.28). The median change<br />

in estradiol from baseline to wk 12 was 0.3 with a range from -3 to<br />

14.6 (p=.49). Twelve wk results are anticipated for 6 additional pts on<br />

study; however 2 pts withdrew before wk 12. Based on the Wilcoxon<br />

signed rank test, estradiol levels were not elevated at wks 2 (n=17)<br />

or 7 (n=16) when compared to baseline. Graphical analysis showed<br />

a relationship with increasing estradiol coinciding with decreasing<br />

FSH, as physiologically expected. All patients reported being less<br />

bothered by menopausal symptoms on the MSCL from baseline to<br />

wk 12. Improvement in sexual function/FSFI scores was noted in all<br />

sexually active women.<br />

Conclusion: Treatment with intravaginal 10µg 17- β estradiol<br />

(Vagifem ®) did not elevate wks 2, 7 or 12 estradiol. It also provided<br />

relief of vaginal and menopausal symptoms and improvement in<br />

sexual function in postmenopausal women with HR+ BC on adjuvant<br />

AIs. This is consistent with findings in the general population.<br />

Updated data from this study will be presented for all patients treated<br />

on study.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

270s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 2<br />

P2-12-06<br />

Ultra-low dose vaginal estriol and Lactobacillus acidophilus<br />

(Gynoflor®) in early breast cancer survivors on aromatase<br />

inhibitors: Pharmacokinetic, efficacy and safety results from a<br />

phase I study.<br />

Neven P, Donders G, Mögele M, Lintermans A, Bellen G, Ortmann<br />

O, Buchholz S. University Hospital Gasthuisberg Leuven, Belgium;<br />

Femicare vzw, Clinical Research for Women, Tienen, Belgium;<br />

Universitätsklinikum Regensburg, Germany<br />

Introduction: Intra-vaginal estradiol (E2) is effective to treat<br />

symptomatic vaginal atrophy. However, in postmenopausal (PM)<br />

women on aromatase inhibitors (AI), this increases serum E2 levels.<br />

We here report pharmacokinetic (PK), efficacy and safety results of<br />

a phase I study using vaginal tablets with lyophilized lactobacilli and<br />

0.03 mg estriol (E3) (Gynoflor®) in PM breast cancer (BC) survivors<br />

on AI reporting atrophic vaginitis.<br />

Patients and Methods: In this open label phase I PK study, women<br />

being treated for ≥6 months with a non-steroidal AI and reporting<br />

non-hormone treatment-resistant vaginal atrophy were recruited. The<br />

primary objective was absorption and serum concentration of estrone<br />

(E1), E2, E3, FSH, LH and SHBG during initial therapy with one<br />

vaginal tablet of Gynoflor® daily for 28 days and during subsequent<br />

maintenance therapy of a daily tablet 3x per week for another 8 weeks.<br />

The secondary objective was to evaluate clinical symptoms, changes<br />

in vaginal pH, epithelium and microflora. Patients were assessed 6<br />

times for endpoints. Blood was taken pre-insertion (baseline samples)<br />

at study entry, week 2, 4, 8, 12. At entry and week 4, multiple PK<br />

blood samples were drawn 0.5 hours pre-insertion and 0.5, 1, 2, 3, 4,<br />

6, 8, 24 hours post-insertion. Serum samples were assayed by highly<br />

sensitive GC/MS.<br />

Results: The interim results of 8 of 16 enrolled patients who have<br />

completed the 12 week study period are presented. Basline estrogen<br />

concentrations of all women were below the levels of quantitation (E3:<br />

< 10 – 20 pg/ml; E2: < 1 pg/ml; E1: < 2 pg/ml). After first insertion,<br />

E3 serum levels transiently increased in 7 of 8 women; the single<br />

maximum concentration of all women was 132 pg/ml and highest<br />

levels (12-132 pg/ml) were observed usually 3 hours post-insertion.<br />

After 4 weeks of daily application, PK results showed an increase in<br />

E3 levels as compared to baseline in only 5 of 8 women. In addition,<br />

the single maximum concentration of all women was lower, 44 pg/ml,<br />

which was only reached after 8 hours. The baseline E3 concentrations<br />

did not change over the complete period of 12 weeks and were in the<br />

range of the limit of quantitation. The serum concentrations of E2 and<br />

E1 did not increase at any time point in any of the women. Clinical<br />

symptoms, vaginal pH and maturation index improved already after<br />

2 weeks of therapy. Global efficacy and tolerability were rated by the<br />

investigators in at least 75% of the cases as (very) good, and by at<br />

least 87.5 % of the women. No serious adverse events were observed.<br />

Conclusions: Gynoflor® vaginal tablet is an effective and safe option<br />

to treat vaginal atrophy in PM patients with early BC on AI. Our data<br />

showed only a transient increase in E3 serum concentration after first<br />

application, and this increase was lower after 4 weeks. Baseline E3<br />

concentrations did not increase demonstrating that no accumulation<br />

occurred. Serum E2 and E1 concentrations did not change and were<br />

always below limit of quantitation. The clinical significance of<br />

transient vaginal E3 absorption is unknown, however the absence of<br />

elevated E2 and E1 serum levels is compatible with the desired effect<br />

for BC patients taking an AI.<br />

*The first and second author have shared first authorship.<br />

P2-12-07<br />

A review of clinical endpoints and use of quality-of-life outcomes<br />

in phase III metastatic breast cancer clinical trials<br />

Tatla R, Landaverde D, Victor C, Miles D, Verma S. University of<br />

Toronto, ON, Canada; Sunnybrook Odette <strong>Cancer</strong> Center, Toronto,<br />

ON, Canada; Dalla Lana School of Public Health, University of<br />

Toronto, ON, Canada; Mount Vernon <strong>Cancer</strong> Centre, United Kingdom<br />

Background: The management of metastatic breast cancer (MBC)<br />

is often considered to be palliative, with most interventions intended<br />

to relieve disease symptoms, minimize treatment effects and prolong<br />

patient survival. The impact of disease and treatment on a patient’s<br />

functional abilities has led to the emphasis of incorporating quality<br />

of life (QoL) measures into clinical trials. The primary objective of<br />

this study is to evaluate phase III clinical trials in MBC, and assess<br />

the inclusion of QoL as an endpoint, in addition to conventional<br />

progression and survival endpoints.<br />

Methods: A structured PubMed search was conducted to identify<br />

phase III clinical trials published between Jan. 1990 and Aug.<br />

2011, which evaluated systemic treatment in MBC patients. Data<br />

pertaining to treatment regimens, study endpoints and clinical findings<br />

were collected, with a particular focus on progression-based (PB),<br />

overall survival (OS), and QoL endpoints. The instrument(s) used in<br />

evaluating QoL were also noted (when applicable).<br />

Results: Of 520 publications identified, 122 phase III MBC clinical<br />

trials met the inclusion criteria. Of these studies, 98.4% and 95.9%<br />

included PB and OS respectively, as clinical endpoints, while QoL was<br />

assessed in only 46 (37.7%) studies. 14 instruments were identified as<br />

QoL measurement tools among these studies, with EORTC QLQ-C30<br />

and FACT-B accounting for 54.7% of the instruments used. While<br />

the inclusion of QoL was not associated with the significance of PB<br />

results, there was an association between the inclusion of QoL and<br />

OS results, with 59% of significant OS studies and 32% of nonsignificant<br />

OS studies including QoL as a clinical endpoint (p=0.016).<br />

When stratified by treatment arm, it was found that studies favouring<br />

standard therapy were more likely to include QoL (75%, p=0.045),<br />

compared to those favouring the intervention (56%), and those without<br />

significant differences (32%).<br />

Conclusions: Although the importance of QoL is often emphasized<br />

in MBC management and treatment decisions, only one-third of<br />

identified phase III clinical trials included an assessment of QoL.<br />

About half of these trials showed no statistically significant differences<br />

in QoL endpoint; of note, instruments of varying validity were utilized.<br />

There needs to be greater emphasis on the evaluation of QoL, with<br />

the use of standard and validated QoL tools in MBC clinical trials,<br />

especially as we increasingly focus on progression-based endpoints.<br />

P2-12-08<br />

Sorafenib for treatment of breast-cancer related lymphedema<br />

Zambetti M, Guidetti A, Carlo-Stella C, De Benedictis E, Tessari A,<br />

Balzarini A, Caraceni A, Gianni L, Gianni AM. IRCCS Ospedale<br />

<strong>San</strong> Raffaele, Milan, Italy; Fondazione IRCCS Istituto Nazionale<br />

Tumori, Milan, Italy; Humanitas <strong>Cancer</strong> Center, IRCCS Istituto<br />

Clinico Humanitas, Rozzano, Italy<br />

BACKGROUND Lymphedema (LE) is a common complication<br />

of breast cancer (BC) treatments conditioning disability that affects<br />

quality of life. Decongestive therapy is the most popular treatment<br />

but it determines only a transient advantage, while pharmacologic<br />

therapy didn’t impact on LE. On the basis of clinical observations<br />

of LE regression in patients treated with sorafenib with antitumoral<br />

intent, we hypothesized that sorafenib could have an anti-LE activity<br />

through inhibition of vascular permeability by suppressing VEGFRs.<br />

www.aacrjournals.org 271s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

METHODS We conducted a single-arm, monoistitutional phase II<br />

study in BC patients with treatment-acquired LE of the arm. Major<br />

or uncontrolled cardiological disease, brain metastasis, history of<br />

thromboembolism were exclusion criteria. Concomitant chemo or<br />

hormonal therapy was allowed. Pts received sorafenib 200 mg daily<br />

for a maximum of 8 weeks. The primary end-point was to evaluate<br />

the efficacy of sorafenib as reduction of LE, defined by the percentage<br />

reduction (PR) of the difference between the total arm circumference<br />

(measured as the sum of the circumference at 12 points) of the affected<br />

and the controlateral arm (Starritt, Peterk JA <strong>Cancer</strong> 2001–10):<br />

[(Initial Difference – Final Difference)/Initial Difference] x 100.<br />

Secondary end-points were safety and duration of response (DOR).<br />

The study was designed to test the null hypothesis that the PR of<br />

edema observed with this therapy was at most 20% versus the<br />

alternative hypothesis that the PR obtained by this regimen was ≥40%.<br />

RESULTS From May 2009 to April 2011, 36 BC pts were enrolled.<br />

All pts underwent axillary dissection and 29 pts had received adjuvant<br />

radiotherapy, but none on the axilla. Median time from primary breast<br />

surgery and from occurrence of edema to study enrollement was<br />

65 and 49 months, respectively. All pts are evaluable for efficacy<br />

and toxicity. Most common toxicities included grade 1-2 gastralgia<br />

(17%), hypertension (17%) and rash (43%); one patient experienced<br />

grade 3 hand-foot syndrome. Twenty-five pts completed the planned<br />

8 weeks of therapy, 11 (31%) had early treatment discontinuation<br />

after 2 (n=6), 4 (n=4) and 6 (n=1) weeks of treatment due to recurrent<br />

grade 2 toxicity or to relapse of disease (n=1). The median PR of<br />

the difference between the two arms was 34% (range, 2-100), 14<br />

pts (39%) experienced a LE reduction ≥40%. Among 25 pts who<br />

completed therapy, 12 (48%) achieved a PR ≥40%. The median<br />

difference of total circumferences between the LE and controlateral<br />

arm was significantly reduced after treatment: 37 cm (range 8–88)<br />

vs 25 cm (range 1–62) (p = 0.006). Best response was achieved after<br />

a median of 5 weeks of therapy (range 1– 6) and the median DOR<br />

was 8 weeks (range 4-15). Reduction of LE was associated with<br />

improvement of related symptoms. After discontinuation of study<br />

drug 84% pts presented a progressive increase of total circumference<br />

of LE arm and returned to values similar to baseline after a median<br />

of 7 weeks (range 2–11).<br />

CONCLUSIONS: Low dose of sorafenib has a good toxicity profile<br />

and exerts a significant anti-LE activity in BC patients. The early but<br />

transient effect observed in this study suggests exploring different<br />

schedule of administration. Further studies are warranted in order to<br />

obtain a durable benefit in term of reduction of LE and quality of life.<br />

P2-12-09<br />

A randomized controlled trial of support group intervention<br />

after breast cancer treatment: Results on sick leave, health care<br />

utilization and health economy.<br />

Granstam Björneklett H, Rosenblad A, Lindemalm C, Ojutkangas<br />

M-L, Letocha H, Strang P, Bergkvist L. Centre for Clinical Research,<br />

Västerås, Västmanland, Sweden; <strong>Cancer</strong> Center Karolinska,<br />

Karolinska, Stockhoöm, Sweden; Karolinska Institutet, Stockholm,<br />

Sweden<br />

Background<br />

More than 50% of breast cancer patients are diagnosed before the<br />

age of 65. Returning to work after treatment is, therefore, of interest<br />

for both the individual and society. The aim was, therefore, to study<br />

the effect of support group intervention on sick leave, health care<br />

utilization and health economy.<br />

Material and Methods<br />

Of 382 patients with newly diagnosed breast cancer, 191 + 191<br />

patients were randomized to an intervention group or to a routine<br />

control group respectively.<br />

The intervention group received support intervention on a residential<br />

basis for one week, followed by four days of follow-up two months<br />

later. The support intervention included informative-educational<br />

sections, relaxation training, mental visualization and non-verbal<br />

communication. Patients answered a questionnaire at baseline, 2, 6<br />

and 12 months about sick leave and health care utilization.<br />

Result There was a trend towards longer sick leave and more health<br />

care utilization in the intervention group. The difference in total costs<br />

was statistically significantly higher in the intervention group after<br />

12 months (p= 0.0036)<br />

Conclusion There was no economic benefit from intervention in its<br />

present form. In fact we saw a tendency towards longer sick leave<br />

and more health care utilization and we could show that the total cost<br />

in the intervention group was actually higher and that this difference<br />

was statistically significant after twelve months.<br />

Key words: Support intervention, breast cancer, return to work, sick<br />

leave, health care utilization, health economy<br />

P2-12-10<br />

Psycho-spiritual therapy for improving the quality of life and<br />

spiritual well-being of women with breast cancer<br />

Loghmani A, Jafari N, Zamani A, Farajzadegan Z, Bahrami F, Emami<br />

H. Isfahan University of Medical Sciences, Isfahan, Islamic Republic<br />

of Iran; University of Isfahan, Islamic Republic of Iran<br />

Objective: Psychological distress and morbidity are common<br />

consequences of diagnosis and treatment of breast cancer and<br />

associated with poor quality of life (QOL). Among several<br />

approaches, spirituality has been shown to be significantly associated<br />

with improving the quality of life of these patients. The aim of this<br />

study was to assess the role of psycho-spiritual therapy intervention<br />

in improving the quality of life and spiritual well-being of patients<br />

with breast cancer.<br />

Methods: This was a randomized controlled trial study which was<br />

conducted in the <strong>Breast</strong> <strong>Cancer</strong> Research Center, St. S. Al-Shohada<br />

hospital, Isfahan, Iran. Sixty-eight patients with breast cancer were<br />

randomized to either psycho-spiritual therapy intervention group or<br />

control group who received routine management and educational<br />

programs. Before and after 6 weeks of psycho-spiritual therapy<br />

sessions, the quality of life was evaluated using <strong>Cancer</strong> quality-oflife<br />

questionnaire (QLQ-C30) and its supplementary breast cancer<br />

questionnaire (QLQ-BR23) and Spiritual well-being was measured<br />

using the Functional Assessment of Chronic Illness Therapy Spiritual<br />

Well-being scale (FACIT-Sp12). Multivariate, repeated-measures<br />

ANOVA, T-test and Paired T-test were used for analysis using<br />

Predictive Analytic Soft Ware (PASW, version 18) for windows.<br />

Results: In all sixty five patients actually completed the six-week<br />

intervention and were evaluated for the outcomes. The mean Global<br />

health status score/QOL reached from 44.37 (SD: 13.03) to 68.63<br />

(SD: 10.86), (p


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 2<br />

P2-12-11<br />

Use of the DigniCap System to Prevent Hair Loss in Women<br />

Receiving Chemotherapy (CTX) for Stage I <strong>Breast</strong> <strong>Cancer</strong> (BC)<br />

Rugo HS, Serrurier KM, Melisko M, Glencer A, Hwang J, D’Agostino,<br />

Jr. R, Hutchens S, Esserman LJ, Melin S. University of California<br />

<strong>San</strong> Francisco Helen Diller Comprehensive <strong>Cancer</strong> Center, <strong>San</strong><br />

Francisco, CA; Wake Forest Baptist Health Medical Center, Winston-<br />

Salem, NC<br />

Background: Alopecia is a non-life threatening complication of<br />

adjuvant CTX for early stage BC. CTX induced hair loss affects<br />

quality of life and potentially impacts decisions regarding the risks<br />

and benefits of treatment. Scalp cooling (SC), used internationally by<br />

thousands of patients (pts) including a growing number in the U.S., is<br />

thought to prevent hair loss through decreased follicular metabolic rate<br />

and vasoconstriction resulting in reduced delivery and cellular uptake<br />

of drugs. The DigniCap System is a self-contained SC system with<br />

circulating coolant. We sought to evaluate feasibility and efficacy of<br />

the DigniCap System in pts with stage I BC.<br />

Methods: We performed an initial prospective feasibility study before<br />

a planned pivotal trial. The primary endpoint was feasibility, defined<br />

as 75% HL). Pts assessed their own HL using the Dean’s<br />

scale, and pt-assessed quality of life, time to and quality of hair regrowth,<br />

and impact of HL on treatment decisions were also evaluated.<br />

Successful prevention of alopecia was defined as < grade 2 HL by the<br />

Dean’s scale. Eligibility included stage I disease and excluded use of<br />

anthracycline-taxane combination or sequential CTX.<br />

Results: 20 pts were enrolled. CTX regimens included: docetaxel and<br />

cyclophosphamide (TC) x 4-6 cycles (n=16), weekly paclitaxel and<br />

trastuzumab x 12 (n=2), and docetaxel, carboplatin, and trastuzumab<br />

(TCH) x 6 (n=2). 19 of 20 pts (95%) completed all CTX using the<br />

DigniCap System. By IP assessment, 15 pts (75%) had a maximum<br />

of ≤ grade 2 HL; 2 pts (10%) and 3 pts (15%) had a maximum grade<br />

3 or 4 HL respectively. By pt assessment, 11 pts (55%) reported<br />

≤ grade 2 HL. 68% and 32% of pts reported grade 1 or 2 toxicity,<br />

respectively, including head/scalp pain, feeling chilled, and rash. 85%<br />

of pts reported that SC made decisions about CTX easier. At a median<br />

follow-up of 15.4 months, no scalp metastases have been observed.<br />

Conclusions: Use of the DigniCap System is feasible, effective in<br />

preventing CTX-induced alopecia in the majority of users, and safe<br />

with short-term follow-up in pts with stage I BC. Additional studies<br />

should help to define potential causes for cap failure. This is the first<br />

prospective US trial to evaluate the effect of SC using an expert IP<br />

and blinded photographs to assess extent of alopecia. A larger trial is<br />

planned to confirm these results in pts with stage I/II disease.<br />

Support for this trial was provided by the Tauber Foundation (UCSF),<br />

the Madonia and Cooper Funds (WFBH), and Dignitana.<br />

P2-12-12<br />

Efficacy and safety of scalp cooling (SC) treatment for alopecia<br />

prevention in women receiving chemotherapy (CTX) for breast<br />

cancer (BC).<br />

Serrurier KM, Melisko ME, Glencer A, Esserman LJ, Rugo HS. UCSF<br />

Helen Diller Comprehensive <strong>Cancer</strong> Center<br />

Background: CTX induced hair loss (HL) is one of the most<br />

distressing side effects of BC treatment. Compared to women without<br />

CTX induced alopecia, those with alopecia have reported poorer body<br />

image, lower self-esteem and worse quality of life. In some cases,<br />

CTX induced HL may affect choice of therapy. Given the emotional<br />

impact of HL, the potential benefit of preventing CTX induced<br />

alopecia is great. We conducted a registry study tracking safety and<br />

efficacy in BC patients (pts) treated at our center who independently<br />

elected to use Penguin Cold Cap Therapy (PCCT).<br />

Methods: Pts who chose to use PCCT during CTX for early stage<br />

(ES) or advanced (M) disease signed informed consent to participate<br />

in the registry. HL was rated by practitioner assessment using the 5<br />

point Dean’s scale for HL: grade 0 (no HL), 1 (< 25% HL), 2 (25-50%<br />

HL), 3 (50-75% HL), and 4 (>75% HL), and by pt assessment using<br />

a visual analogue scale (VAS) from 0-100 where 0 = no HL and 100<br />

= 100% HL. Assessments were every 2-3 weeks at the start of each<br />

CTX cycle, and once in follow-up 1-6 months after completing CTX.<br />

Additional data collected included pt reported adverse events and pt<br />

satisfaction with the success, ease of use, and tolerability of PCCT.<br />

Results: 31 pts (29 with ES and 2 with M disease) have registered.<br />

25 have completed CTX and 20/25 are off study. 6 pts currently on<br />

CTX and 2 pts who stopped PCCT after cycle1 due to toxicity of<br />

CTX rather than issues with PCCT were excluded from the safety<br />

and efficacy analyses due to incomplete data. 2 additional pts were<br />

excluded from efficacy analysis due to beginning PCCT after the<br />

start of CTX, leaving 21 pts evaluable for efficacy and 23 pts for<br />

toxicity. CTX regimens included: docetaxel and cyclophosphamide<br />

(TC), docetaxel, carboplatin and trastuzumab (TCH), sequential<br />

doxorubicin and cyclophosphamide and paclitaxel, (AC/T), weekly<br />

nab-paclitaxel (nab). PCCT toxicities were all < grade 2 and included:<br />

15 pts (71%) with headaches and 5 (22%) with dandruff during PCCT.<br />

All pts experienced head pain, and 20 pts (95%) had scalp pain and<br />

felt chilled during the PCCT. Table 1 depicts clinician and patient<br />

reported efficacy.<br />

Table 1. Efficacy of PCCT by clinician and pt assessment<br />

Clinician reported HL by Dean’s scale<br />

CTX Regimen, N<br />

(N=pts)<br />

Pt reported HL by VAS (N=pts)<br />

Grade 0 or 1 Grade 2 Grade 3 or 4 VAS 0-25 VAS 26-50 VAS >50<br />

TC, 12 2 1 2 7 5 -<br />

TCH, 5 2 3 - 2 1 2<br />

AC/T, 2 1 - 1 - - 2<br />

Nab, 2 1 1 - - - 2<br />

Of the 20 pts assessed following completion of CTX and PCCT, 12<br />

(60%) “highly recommend” PCCT, and 8 (40%) “recommend to some<br />

degree”. No ESBC pts have worn a wig at any point.<br />

Conclusions: SC treatment (PCCT) is a viable but variably effective<br />

method of preventing HL for ESBC pts receiving CTX. The success of<br />

SC in hair preservation was dependent on the type and length of CTX<br />

regimen. Other patient factors impacting HL may exist. Short term<br />

toxicity is minimal. Given the emotional impact of CTX induced HL,<br />

ongoing study of PCCT and other SC devices, comparing different<br />

CTX regimens, is worthwhile. Ongoing studies at our center will<br />

include independently graded photographs to eliminate reporting<br />

bias by the clinician.<br />

Support for this trial was provided by the Tauber Foundation.<br />

P2-12-13<br />

Results of randomised controlled phase II study (KBCSG02 trial)<br />

of the efficiency of palonosetron, aprepitant, and dexamethasone<br />

for day1 with or without dexamethasone on days2 and 3.<br />

Kosaka Y, Sengoku N, Kikuchi M, Nishimiya H, Enomoto T, Kuranami<br />

M, Watanabe M. Kitasato University School of Medicine, Sagamihara,<br />

Japan<br />

Background: Emesis is one of the major non-hematologic toxicity<br />

caused by chemotherapy. The control of Chemotherapy Induced<br />

Nausea and Vomiting (CINV) is conducted to a life lengthening.<br />

www.aacrjournals.org 273s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Recently, CINV is controlled by the second generation 5-HT3 receptor<br />

blocker (Palonosetron ; PALO) and a NK-1 receptor antagonist<br />

(Aprepitant ; APR). It has been shown that dexamethasone (DEX)<br />

with 5-HT3 receptor blocker improves acute / delayed CINV.<br />

However, it has not been determined the medication schedule of<br />

DEX with PALO and APR. The purpose of this study is evaluated the<br />

efficiency of palonosetron, aprepitant and dexamethasone for day1<br />

with or without dexamethasone on days2 and 3.<br />

Methods: <strong>Breast</strong> cancer patients who administered anthracyclin<br />

drug regimen have been eligible from April, 2011 to June, 2012. The<br />

patients were randomised to group A (PALO/APR/DEX one day) and<br />

group B (PALO/APR/DEX three days). Eighty patients was estimate<br />

as study samples. The primary endpoint was a complete response<br />

(CR) rate of vomiting, the secondary endpoint was a complete control<br />

(CC) rate of vomiting.<br />

Trial design<br />

Aprepitant (125 mg) (80 mg) (80 mg)<br />

Palonosetron (0.75 mg)<br />

dexamethasone (9.9 mg) (6.6 mg or placebo) (6.6 mg or placebo)<br />

Chemotherapy do<br />

days 1 2 3 4 5<br />

Results: Forty patients were enrolled in this study, and patient<br />

recruitment is continued. CR rate was no significant difference in<br />

both groups (76.9% of group A, 73.3% of group B). CC rate of group<br />

A (61.5%) did not show inferiority compared with group B (40.0%).<br />

Conclusions: In this current study, we suggest that one day<br />

dexamethasone treatment could reduce enough the emesis of high<br />

emetogenic chemotherapeutic agents.<br />

P2-12-14<br />

Use of the MD Anderson Symptom Inventory to Screen for<br />

Depression in <strong>Breast</strong> <strong>Cancer</strong><br />

Kvale EA, Azuero CB, Azuero A, Fisch M, Ritchie C. Birmingham<br />

VA Medical Center, Birmingham, AL; University of Alabama at<br />

Birmingham, AL; University of Alabama, Tuscaloosa, AL; MD<br />

Anderson <strong>Cancer</strong> Center, Houston, TX; University of California,<br />

<strong>San</strong> Francisco, CA<br />

Background: The prevalence of depression among breast cancer<br />

patients is estimated to be twice that in the general population,<br />

is linked to diminished quality of life and impaired adherence to<br />

therapy. Depression is frequently underdiagnosed or misdiagnosed<br />

in this population. Efficient screening for depression is central to<br />

patient-centered care and enhanced clinical outcomes for breast<br />

cancer patients.<br />

Purpose: To evaluate the use of a commonly utilized symptom<br />

assessment instrument as a screen to enhance identification of breast<br />

cancer patients experiencing depression.<br />

Methods: Data from a longitudinal surveillance database in outpatient<br />

supportive and palliative care were utilized, 174 breast cancer<br />

patient contacts were evaluated. Patients completed both the MD<br />

Anderson Symptom Inventory (MDASI) and the 9-item Patient Health<br />

Questionnaire (PHQ-9) as components of a routinely collected patientreported<br />

outcomes battery. Performance of the MDASI (using the 1-10<br />

Depression question) in identifying cases of depression (defined as a<br />

score ≥ 15 on the PHQ-9) was determined using receiver operating<br />

characteristic (ROC) analysis.<br />

Results: Data were available on 174 patient contacts. When scored<br />

as a continuous measure, the MDASI performed well with an area<br />

under the ROC curve of 0.87 (95% confidence interval [CI], 0.81-<br />

0.94). An MDASI cutoff score of >= 6 provided a sensitivity of 73%<br />

(95% CI, 58%-88%), a specificity of 80% (95% CI, 74%-87%), a<br />

positive predictive value (PPV) of 46%, and a negative predictive<br />

value (NPV) of 93%.<br />

Conclusion: The “depression” component of the MDASI as a<br />

screening instrument for depression in breast cancer patients yields<br />

suboptimal sensitivity and specificity for use as a screening tool.<br />

Further efforts to evaluate subsequent iterations of the MDASI<br />

and combinations of elements in the MDASI that may enhance<br />

performance are indicated.<br />

P2-12-15<br />

Understanding the complex non face-to-face interventions<br />

delivered by the clinical nurse specialists in metastatic breast<br />

cancer.<br />

Warren M, Mackie D, Leary A. Royal Marsden NHS Foundation Trust,<br />

Sutton, Surrey, United Kingdom; Royal Marsden NHS Foundation<br />

Trust, London, United Kingdom; Independent Healthcare Consultant<br />

and Research Analyst<br />

Introduction: Despite improving survival rates in breast cancer,<br />

globally one third of patients diagnosed with primary breast cancer<br />

will go onto develop metastatic breast cancer. In recent years treatment<br />

for metastatic breast cancer in the United Kingdom has moved to the<br />

ambulatory setting using treatments such as, oral chemotherapies,<br />

oral biological and bisphosphonate therapies meaning telephone<br />

consultation and clinical management, pivotal to confluent care.<br />

Research Objectives: To determine the number and complexity of<br />

incoming telephone calls, the stage of the metastatic breast cancer<br />

disease pathway for example at diagnosis, treatment, follow-up, and<br />

palliative care, the nursing interventions, outcomes and the reason<br />

for contact from patients and carers.<br />

Rationale: Previous studies demonstrate much of the clinical work<br />

of the clinical nurse specialist is delivered via the telephone. The<br />

rationale of this study was to examine the complexity of non faceto-face<br />

patient interaction with the metastatic breast cancer clinical<br />

nurse specialist.<br />

Method: A prospective study conducted over a 1 – month period at a<br />

United Kingdom National Health Service Foundation Hospital Trust.<br />

Data was collected by two clinical nurse specialists on incoming calls<br />

using Excel and a bespoke interrelational structured query database.<br />

These data were then mined using standard data mining techniques<br />

and pattern recognition techniques.<br />

Results: The study collected 28 days of data. 229 patient and carer<br />

telephone contacts were recorded. Most calls were from patients<br />

(62.5%). Incoming calls resulted in the delivery of 1282 interventions,<br />

a mean of six interventions per call (range1-8) and clustered into<br />

four areas: meeting information needs (29%), symptom management<br />

(26%), psychological/social issues (33%) and other issues (12%).<br />

The incoming telephone work accounted for 63 h which represented<br />

30% of the total working time of the clinical nurse specialist. Calls<br />

primarily originated from patients who were in the follow-up phase<br />

(43% of calls), a group usually thought to prefer self management.<br />

P2-13-01<br />

Impact of Body Mass Index (BMI) on the efficacy of aromatase<br />

inhibitors to suppress estradiol serum levels in postmenopausal<br />

patients with early breast cancer: a prospective proof of principle<br />

Pfeiler G, Königsberg R, Hadji P, Fitzal F, Maroske M, Ban G,<br />

Zellinger J, Exner R, Seifert M, Singer C, Gnant M, Dubsky P. Medical<br />

University of Vienna, Austria; Applied <strong>Cancer</strong> Research – Institution<br />

for Translational Research Vienna (ACR-ITR VIEnna)/CEADDP,<br />

Austria; Universityhospital of Giessen and Marburg GmbH, Germany<br />

Background<br />

BMI impacts on the efficacy of aromatase inhibitors (AIs) regarding<br />

disease outcome in patients with early breast cancer. We hypothesized<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

274s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 2<br />

that this clinical impact of BMI is driven by the inability of AIs to<br />

suppress estradiol (E2) serum levels to the same low level in obese<br />

compared to non-obese patients.<br />

Methods<br />

68 postmenopausal ER positive patients with breast cancer, scheduled<br />

to receive anastrozole or letrozole as adjuvant endocrine treatment<br />

were prospectively included in this study. Serum was taken before<br />

(T1) and 3 months after start with aromatase inhibitors (T2). Serum<br />

levels of FSH, LH, E2, glucose and insulin were analyzed immediately<br />

in the clinical routine lab and in a dedicated central lab able to measure<br />

E2 in serum at low concentrations with high sensitivity.<br />

Results<br />

40 patients were normal - or overweight (non-obese; BMI 18.5-29.9<br />

kg/m2) and 28 patients were obese (BMI ≥ 30 kg/m2). A weak nonsignificant<br />

correlation between BMI and baseline E2 serum levels<br />

(r=0.2, p=0.2) translated into a strong negative, highly significant<br />

correlation between BMI and baseline FSH (r=-0.6, p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Methods: The Oncology Services Comprehensive Electronic Records<br />

(OSCER) database was used to identify women with BC (ICD-9<br />

174), ≥1 clinic visit, and confirmed AI (anastrozole, letrozole, or<br />

exemestane) in 2011. Women with metastases (ICD-9 196-198,<br />

stage IV, or M1 disease) were excluded. OSCER captures electronic<br />

medical record data on >500,000 cancer patients since 2004 from<br />

US outpatient community and hospital affiliated oncology practices.<br />

OSCER is projected nationally through methods of direct estimations<br />

utilizing claims data (Smith et al. submitted). To identify subgroups of<br />

women at increased risk of bone loss, estimates were stratified by age<br />

and number of AIs received. Bone loss was defined by diagnosis of<br />

osteoporosis (ICD-9 733.0), osteopenia (ICD-9 733.90), or receipt of<br />

bone tx either before initiation of first AI (proxy for pre-existing bone<br />

loss) or any time before or during 2011 (proxy for current bone loss).<br />

Results: When projected to the US population, it is estimated that<br />

489,664 (95% CI 467,642—511,686) women with non-metastatic<br />

BC were treated with an AI in 2011. Of these, 52% (253,672 [95%<br />

CI 239,229—268,115]) were 65 or older. The table summarizes<br />

results for the projected cohorts. These data indicate that 19% of<br />

women receiving AIs were in the pre-/peri-/early post-menopausal<br />

age group (55 or younger), and approximately 66% of women treated<br />

with AIs did not have evidence of pre-existing bone loss (i.e., either<br />

osteoporosis diagnosis or receipt of bone tx before AI).<br />

Total Age < 45 Age 45-55 Age 56-64 Age ≥ 65<br />

Total non-metastatic BC pts on AI 489,664 11,536 80,843 143,613 253,672<br />

Pts with >1 AI Rx 438,243 11,536 69,406 129,782 227,520<br />

No evidence of bone loss before<br />

initiation of first AI<br />

322,541 10,531 67,319 97,922 146,768<br />

Evidence of bone loss before or<br />

during AI tx<br />

199,683 3,573 22,907 59,881 113,322<br />

Conclusions: This is the first study to estimate prevalence of women<br />

with non-metastatic BC treated with AIs in the US. As a large<br />

proportion of women are diagnosed with ER+ve BC and eligible<br />

for AIs (Jemal et al. 2007), a growing number will face increased<br />

subsequent risk of fractures. These findings suggest that up to 500,000<br />

women annually are treated with AIs, and over 40% have evidence<br />

of bone loss before or during AI. These women will face increased<br />

risk of fractures, highlighting the need for prevention of fractures and<br />

intervention with targeted bone tx to increase bone mass and prevent<br />

or treat osteoporosis.<br />

P2-13-04<br />

Superior efficacy of anastrozole to tamoxifen as adjuvant therapy<br />

for postmenopausal patients with hormone-responsive breast<br />

cancer. Efficacy results of long-term follow-up data from N-SAS<br />

BC 03 trial<br />

Imoto S, Osumi S, Aogi K, Hozumi Y, Mukai H, Iwata H, Yokota I,<br />

Yamaguchi T, Ohashi Y, Watanabe T, Takatsuka Y, Aihara T. School<br />

of Medicine, Kyorin University, Tokyo, Japan; National Hospital<br />

Organization Shikoku <strong>Cancer</strong> Center, Ehime, Japan; Jichi Medical<br />

University, Tochigi, Japan; National <strong>Cancer</strong> Center Hospital East,<br />

Chiba, Japan; Aichi <strong>Cancer</strong> Center Hospital, Aichi, Japan; School of<br />

Public Health, University of Tokyo, Tokyo, Japan; Tohoku University<br />

School of Medicine, Miyagi, Japan; Hamamatsu Oncology Center,<br />

Shizuoka, Japan; Kansai Rosai Hospital, Hyogo, Japan; Aihara<br />

Hospital, Osaka, Japan<br />

Background: Aromatase inhibitors have been shown to be superior<br />

to tamoxifen as adjuvant therapy in postmenopausal patients with<br />

hormone-responsive breast cancer. Here we report the efficacy results<br />

of long-term follow-up data from N-SAS BC 03 trial (UMIN CTRID:<br />

C000000056), in which anastrozole was compared to tamoxifen in<br />

hormone-responsive postmenopausal early breast cancer patients<br />

who had taken tamoxifen for 1—4 years out of a total of 5 years of<br />

treatment as adjuvant therapy.<br />

Patients and methods: Out of a total of 706 recruited patients, 696<br />

patients (tamoxifen group, n=345; anastrozole group, n=351) were<br />

used as the full analysis set for the present study. The log-rank test<br />

was used to compare the two groups in terms of disease-free survival<br />

(DFS) and relapse-free survival (RFS), Kaplan-Meier estimates were<br />

calculated. The treatment effects were estimated by Cox’s proportional<br />

hazard model and were expressed as hazard ratios, with associated<br />

95% confidence intervals (CIs).<br />

Results: After a median follow-up of 76.1 months, (range: 1.3-110<br />

months), the unadjusted hazard ratio was 0.87 (95%Cl 0.60-1.26; logrank<br />

p=0.457) for DFS and 0.77 (95%Cl 0.49-1.22; log-rank p=0.266)<br />

for RFS, both in favor of anastrozole. The estimated hazard ratio<br />

(95%CIs) for DFS and for RFS until each time point (right censored<br />

data) was as follows: until 30 months: 0.54 (0.30-0.98) and 0.48 (0.23-<br />

0.98), until 42 months; 0.65 (0.40-1.06) and 0.53 (0.29-0.97), until 54<br />

months: 0.77 (0.50-1.19) and 0.63 (0.37-1.06), until 66 months: 0.82<br />

(0.55-1.24) and 0.72 (0.44-1.17), until 78 months: 0.83 (0.57-1.23)<br />

and 0.73 (0.46-1.17), respectively. The hazard ratios (95%CIs) for<br />

DFS and for RFS until 36 months were 0.72 (95%Cl 0.43-1.22) and<br />

0.58 (95%Cl 0.30-1.12), and those after 36 months were 1.06 (95%Cl<br />

0.62-1.81) and 0.98 (95%Cl 0.51-1.88), respectively.<br />

Conclusion: Superior efficacy of anastrozole to tamoxifen in the<br />

Japanese population was suggested over a long-term follow-up<br />

period. It was the greatest early after switching from tamoxifen and<br />

then decreased thereafter.<br />

P2-13-05<br />

A pilot prospective study of adherence to aromatase inhibitor<br />

adjuvant therapy in patients with stage 1-3 breast carcinoma.<br />

Heiss B, Thompson J, Nightingale G, Tait N, Kesmodel S, Bellavance<br />

E, Chumsri S, Bao T, Goloubeva O, Feigenberg S, Tkaczuk K. Marlene<br />

and Stewart Greenebaum <strong>Cancer</strong> Center, University of Maryland,<br />

Baltimore, MD; Thomas Jefferson University, Philadelphia, PA<br />

Introduction: The most practical approach to assessment of<br />

medication adherence in clinical practice is via patient self-reporting,<br />

which can be measured using the 8-item Morisky Medication<br />

Adherence Scale (MMAS). The 8-item MMAS was developed from<br />

a previously validated 4-item scale by supplementing additional items<br />

measuring specific medication-taking behaviors to better capture<br />

barriers surrounding adherence behavior and has been determined<br />

to have better reliability than 4-item scale. Validated medication<br />

adherence surveys such as an 8-item MMAS, are untested in oncology,<br />

but have been validated in HTN, HIV, DM and other disorders in the<br />

setting of multiple chronic medications. Thus this study will serve as<br />

a pilot study of the MMAS in the oncology population. Methods:<br />

This is a prospective study conducted at the University of Maryland<br />

Greenebaum <strong>Cancer</strong> Center (UMGCC). The primary objective was<br />

to identify the adherence level (high, medium, low) to adjuvant<br />

(AI) in early stage (1-3) breast cancer (BC) patients in order to<br />

identify predictors of adherence. Subjects were stratified according<br />

to duration of therapy with AI (≤ 2 yrs or >2 yrs) at the time of the<br />

survey. The secondary objective was to identify baseline predictors<br />

for adherence. Patients received a maximum 8 points for maximum<br />

adherence to medication treatment or high adherence, 6-7 points for<br />

medium adherence and < 6 points for low adherence. Eligibility:<br />

Adult females, postmenopausal, signed consent, hormone receptorpositive<br />

BC (ER and/or PR >=1%), pathologically confirmed Stage<br />

1-3 BC, completed surgery, adjuvant chemotherapy, and/or radiation<br />

therapy, and were recommended by their medical oncologist to take<br />

adjuvant AI for 5 years. Pts received a prescription for adjuvant AIs<br />

(anastrozole, letrozole or exemestane) after completion of adjuvant<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

276s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 2<br />

chemotherapy. Results: Patient characteristics: 73 BC pts were<br />

sequentially accrued so far at the UMGCC and filled out the selfreported<br />

questionnaire; Causcasian-62%, African American (AAF)-<br />

37%, Asian-1%, median age at diagnosis-54 (range33-84); stage<br />

1- 36%, stage 2- 47%, stage 3- 17%; mastectomy- 61%; lumpectomy-<br />

39%; local radiation therapy 62%; ER+ 95%; PR+ 78%, HER2+ 17<br />

%; tumor type ductal- 75%; lobular- 20%; mixed ductal/lobular- 5%;<br />

tumor grade-1- 46%, 2- 41%,3- 12%; adjuvant chemotherapy- 63%.<br />

Educational level- N/A- 3%, 2 years.<br />

Univariable regression models for continuous predictors and rxc<br />

contingency tables for categorical ones revealed that the adherence<br />

to AI therapy is not affected by age, marital status, education, prior<br />

cancer therapy, tumor stage, and concurrent medication. However,<br />

there was a trend for better adherence to AI therapy in Caucasian<br />

women vs. AAF (P=0.013). The study is ongoing.<br />

P2-13-06<br />

Effect of letrozole on bone and joints in collagen-induced arthritis<br />

in mice<br />

Lintermans A, Verhaeghe J, Van Bree R, Vanderschueren D,<br />

Vanderhaegen J, Braem K, Lories R, Neven P. University Hospitals<br />

Leuven, KU Leuven, Belgium; KU Leuven, Leuven, Belgium;<br />

University Hospitals Leuven, Belgium<br />

Background Aromatase inhibitors (AIs) are an established treatment<br />

modality for postmenopausal hormone-responsive breast cancer.<br />

Many of AI-treated patients experience AI-induced musculoskeletal<br />

syndrome (AIMSS), which may adversely affect quality of life and<br />

treatment adherence. The etiology underlying this syndrome remains<br />

unknown. Therefore, a well-established mouse model of human<br />

rheumatoid arthritis, collagen-induced arthritis (CIA), was used to<br />

evaluate how AIs affect bone and joints in inflammatory joint disease.<br />

Material and methods Thirty-two female DBA/1 mice were divided<br />

into 2 groups of 16 mice each; ovariectomy (OVX) or sham operation<br />

was performed at 7 weeks of age (=day 0). At day 14, arthritis was<br />

induced in all mice by injection of chicken sternal cartilage type<br />

II collagen emulsified with an equal volume of Freund’s complete<br />

adjuvant, followed by a booster injection at day 21. Afterwards,<br />

mice were treated with letrozole or vehicle during 30 days. Mice<br />

were sacrificed at day 54. Parameters analyzed included clinical<br />

CIA score of paws, dual-energy x-ray absorptiometry, microCT of<br />

femora, histopathological examination of ankles and paws and IGF-I<br />

levels. Analysis was performed with ANOVA, post-hoc tests, general<br />

linearized and mixed models.<br />

Results As expected, OVX led to an increased lean and total body<br />

weight relative to the sham group. Repeated measurements analysis<br />

with mixed models suggested interaction between weight, time and<br />

arthritis severity. Serum IGF-I concentrations were significantly<br />

higher in the OVX group compared with the sham group, but no<br />

differences were observed between letrozole treatment and controls.<br />

Collagen immunization resulted in progressive inflammation of the<br />

paws including the ankles that increased up to day 54. Over time,<br />

clinical scores for arthritis severity were highest in the OVX/vehicle<br />

group. Both letrozole groups had an intermediate course and the<br />

lowest scores were seen in the paws of the sham/vehicle group, as<br />

expected. Histopathological examination of hematoxylin and eosinstained<br />

sections were in line with these results.<br />

Subcapital BMD at the end of the arthritis experiments was<br />

significantly different between the four groups. Post-hoc test<br />

confirmed the negative effect of OVX versus sham procedure, in<br />

the presence or absence of letrozole treatment. MicroCT analysis<br />

showed that OVX induced a significant decrease in bone mineral<br />

density and content in both trabecular (volume and number) and<br />

cortical bone (volume, area, thickness and medullary area) compared<br />

to sham operation in vehicle-treated mice. In the letrozole group, only<br />

cortical bone (volume, area inside periost and medullary area) was<br />

significantly reduced following OVX. In addition, in the sham group,<br />

letrozole induced lower trabecular bone volume and less trabecular<br />

numbers compared to vehicle treatment. This was in contrast with<br />

the OVX groups; letrozole treatment resulted in significantly better<br />

trabecular bone parameters.<br />

Conclusion Not only bone loss but also severity of joint disease was<br />

increased by OVX. Remarkably, in contrast to what is observed in<br />

humans, letrozole treatment protected the bone and joints after OVX<br />

but had negative effects in its absence, thereby providing further<br />

evidence for a regulatory effect in joint and bone biology.<br />

P2-13-07<br />

Long-term follow-up data of the side effect profile of anastrozole<br />

compared with tamoxifen in Japanese women : findings from<br />

N-SAS BC03 trial.<br />

Iwata H, Ohsumi S, Aogi K, Hozumi Y, Imoto S, Mukai H, Yokota I,<br />

Yamaguchi T, Ohashi Y, Watanabe T, Takatsuka Y, Aihara T. Aichi<br />

<strong>Cancer</strong> Center Hospital, Nagoya, Aichi, Japan; NHO Shikoku<br />

<strong>Cancer</strong> Center, Matsuyama, Ehime, Japan; Jichi Medical University,<br />

Tochigi, Japan; School of Medicine, Kyorin University, Tokyo, Japan;<br />

National <strong>Cancer</strong> Center Hospital East, Chiba, Japan; School of<br />

Public Health, University of Tokyo, Tokyo, Japan; Tohoku University<br />

School of Medicine, Sendai, Japan; Hamamatsu Oncology Center,<br />

Hamamatsu, Japan; Kansai Rosai Hospital, Hyogo, Japan; Aihara<br />

Hospital, Osaka, Japan<br />

Background: Aromatase inhibitors (AIs) are standard treatment in<br />

the adjuvant setting such as initial, switch and extended treatment in<br />

postmenopausal hormone-sensitive breast cancer patients overseas.<br />

Non-steroidal AIs have usually been used as adjuvant treatment in<br />

Japanese practice. We already reported the efficacy and short term<br />

adverse events by switching therapy from tamoxifen (TAM) to<br />

anastrozole (ANA) in Japan (<strong>Breast</strong> <strong>Cancer</strong> Res Treat. 2010 ;121:379-<br />

87). However, the usage of adjuvant hormone therapy currently shows<br />

a trend to continue for a long time. Furthermore, long-term follow-up<br />

data is very important for the safe use of AIs. Off course, there are<br />

many data on safety profiles in Western countries. However, there are<br />

limited data in the Asian population. Methods: We investigated the<br />

planned analyses by using the data of a randomized adjuvant study<br />

of tamoxifen alone versus sequential tamoxifen and anastrozole in<br />

hormone-responsive postmenopausal breast cancer patients (N-SAS<br />

BC 03 trial. UMIN CTRID: C000000056). We examined the<br />

chronological changes from baseline to 36 months after randomization<br />

between the TAM arm (351 cases) and the ANA arm (345 cases)<br />

according to the case report forms (CRFs). We prospectively<br />

collected the data and compared data on hot flushes, nausea &<br />

vomiting, anorexia, fatigue, mood distress, headaches, arthralgia,<br />

leukocytopenia, liver transaminase changes, thromboembolism,<br />

vaginal bleeding, vaginal discharge, cardio-vascular events, second<br />

primary cancer and contra-lateral breast cancer between the two arms<br />

www.aacrjournals.org 277s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

every 3 months from baseline to 12 months, and every 6 months up<br />

to 36 months after randomization. The data were expressed as the<br />

difference in the incidence over the time course and were analyzed<br />

using chi-square tests and logistic regressions with the baseline<br />

value as a covariate via generalized estimating equation using robust<br />

standard errors. Results: The incidence of arthralgia are significantly<br />

better in the TAM arm than the ANA arm (P=0.00002). However,<br />

there are no differences concerning all events excluding arthralgia<br />

between the two arms. Ten and twelve cases of second primary<br />

cancer occurred in the TAM and ANA arms, respectively (P=0.6).<br />

Six and three cases of contralateral breast cancer occurred in the<br />

TAM and ANA arms, respectively (P=0.3). According to analyses of<br />

the chronological changes, significant changes were observed in hot<br />

flushes (P=0.001) and vaginal discharge (P.999)<br />

Conclusion: From our retrospective review, we did not observe<br />

any significant difference in the adherence to adjuvant anti-estrogen<br />

therapy in patients with EBC. The present process of discharging<br />

such patients to surgeons and family physicians for follow up does<br />

not seem to affect their compliance.<br />

P2-13-09<br />

Pharmacological impact of endoxifen in a laboratory simulation<br />

of tamoxifen therapy in postmenopausal breast cancer patients<br />

Maximov PY, McDaniel RE, Bhatta P, Brauch H, Jordan VC.<br />

Lombardi Comprehensive <strong>Cancer</strong> Center, Georgetown University<br />

Medical Center, Washington, DC; Dr. Margarete Fischer-Bosch-<br />

Institute of Clinical Pharmacology, Stuttgart, Germany<br />

Tamoxifen is a well-established adjuvant drug for the treatment and<br />

prevention of Estrogen Receptor (ER) positive breast cancer, effective<br />

in both pre- and postmenopausal women. However, the metabolic<br />

activation of tamoxifen to endoxifen by the CYP2D6 enzyme system<br />

still remains controversial to plan the treatment of patients with breast<br />

cancer. Since the discovery and description of the pharmacological<br />

properties of endoxifen, clinical trials were undertaken to determine<br />

the pharmacological relevance of endoxifen. The results of these trials,<br />

however, vary. In this study we demonstrate the pharmacological<br />

impact of endoxifen in a laboratory simulation of breast cancer<br />

therapy with tamoxifen in postmenopausal women in vitro in a<br />

panel of human breast cancer cell lines (MCF-7, T47D, ZR-75-1 and<br />

BT474). The concentrations of estrogens (E1/E2) used to simulate<br />

postmenopausal women treated with tamoxifen, were obtained<br />

from published studies, as well as the concentrations of tamoxifen<br />

and its metabolites (N-desmethyltamoxifen, 4-hydroxytamoxifen<br />

and endoxifen) based on the CYP2D6 genotype in postmenopausal<br />

breast cancer patients (Murdter TE et al, Clin Pharmacol Ther, 2011).<br />

Our results demonstrate that irrespective of CYP2D6 genotype,<br />

tamoxifen and its primary metabolites (N-desmethyltamoxifen and<br />

4-hydroxytamoxifen) are able to inhibit completely the estrogenstimulated<br />

growth of breast cancer cells in vitro. Additional endoxifen<br />

in any concentration corresponding to CYP2D6 genotype was not<br />

able to increase the antiestrogenic effect of tamoxifen and its primary<br />

metabolites. Moreover, we demonstrate that 4-hydroxytamoxifen<br />

is absolutely essentialforinhibition of estrogen action. Based on<br />

our results, we can conclude that endoxifen is pharmacologically<br />

supportive but not essential for any genotype of CYP2D6 in a<br />

postmenopausal setting. This study (PYM) was supported by Susan<br />

G. Komen for the Cure International Postdoctoral Fellowship under<br />

Award number SAC100009(VCJ), and the Department of Defense<br />

<strong>Breast</strong> Program under Award number W81XWH-06-1-0590 (VCJ).<br />

P2-13-10<br />

Prospective randomized and multicentric evaluation of cognition<br />

in menopausal breast cancer patients receiving adjuvant<br />

hormonotherapy: a phase III study (Preliminary results)<br />

Vanlemmens L, Delbeuck X, Servent V, Mailliez A, Vanlemmens L,<br />

Lefeuvre-Plesse C, Kerbrat P, Petit T, Fournier C, Vendel Y, Clisant S,<br />

Bonneterre J, Pasquier F, Le Rhun E. Centre Mémoire de Ressource<br />

et de Recherche - CHRU Lille, Lille, France; Centre Oscar Lambret,<br />

Université Lille Nord de France, Lille, France; Centre Eugène<br />

Marquis, Rennes, France; Centre Paul Strauss, Strasbourg, France;<br />

Centre Oscar Lambret, Lille, France; CHRU, Lille, France<br />

Background<br />

Cognitive impairment has been considered to be a possible adverse<br />

effect of aromatase inhibitors (AI). The aim of the study was to<br />

compare the impact of tamoxifen or AI on verbal memory (Rey<br />

auditory-verbal learning test) and other cognitive functions (memory,<br />

executive and attentional functions) after 6 months of treatment.<br />

Methods<br />

In this randomized, open-label phase III study, menopausal patients<br />

treated with adjuvant hormonotherapy for early breast cancer were<br />

enrolled at the end of the radiotherapy. Patients over 70 years, with<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

278s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 2<br />

a history of cognitive disorder or with prior chemotherapy were<br />

excluded. Detailed neuropsychological assessments and quality of<br />

life evaluations were performed before the 1st administration of<br />

hormonotherapy and then 6 months later. Considering the usual norms<br />

of the auditivo-verbal Rey test, an alpha risk of 5% and a 95% power,<br />

27 patients per arm had to be included. Statistical analyses included<br />

Chi2 test and Student tests when appropriate.<br />

Results<br />

62 consecutive evaluable patients were randomized in 2 arms between<br />

March 2009 and April 2011. Patients received tamoxifen in arm A<br />

(n=31) and AI in arm B (letrozole n=17; anastrozole n=12; exemestane<br />

n= 2). Median age at inclusion was 61.4 years. The median time<br />

since menopause was 10 years. Characteristics of the breast tumor<br />

and initial neuropsychological evaluations did not differ significantly<br />

between the 2 arms. After 6 months, we observed a significant decline<br />

of the performance at the episodic memory test (immediate recall of<br />

the Rey auditory verbal learning test) (p=0.0015) in arm A only and<br />

a significant improvement on executive measures (Trail Making Test<br />

and Stroop test) (respectively p=0.03/p=0.002) in arm B. Quality of<br />

life didn’t differ after 6 months of treatment.<br />

Conclusions<br />

These preliminary results do not support that AI have a worse<br />

adverse effect on cognitive functions than tamoxifen after 6 months<br />

of treatment. A confirmation is planned after one year of treatment.<br />

P2-14-01<br />

Fulvestrant vs exemestane for treatment of metastatic breast<br />

cancer in patients with acquired resistance to non-steroidal<br />

aromatase inhibitors – a meta-analysis of EFECT and SoFEA<br />

(CRUKE/03/021 & CRUK/09/007)<br />

Johnston SRD, Chia S, Kilburn LS, Gradishar WJ, Cameron D,<br />

Dodwell D, Ellis P, Howell A, Im Y-H, Coombes G, Piccart M, Dowsett<br />

M, Bliss J, On Behalf of the SoFEA and EFECT Investigators. The<br />

Royal Marsden Hospital NHS Foundation Trust & The Institute of<br />

<strong>Cancer</strong> Research, London, United Kingdom; British Columbia <strong>Cancer</strong><br />

Agency, University of British Columbia, Vancouver, BC, Canada; The<br />

Institute of <strong>Cancer</strong> Research, Sutton, Surrey, United Kingdom; Robert<br />

H. Lurie Comprehensive <strong>Cancer</strong> Center, Feinberg School of Medicine,<br />

Northwestern University, Chicago, IL; Christie Hospital NHS<br />

Trust, Manchester, United Kingdom; Edinburgh <strong>Cancer</strong> Research<br />

Centre, University of Edinburgh and NHS Lothian, Edinburgh,<br />

United Kingdom; Leeds Teaching Hospitals NHS Trust, St. James’s<br />

University Hospital, Leeds, United Kingdom; Guy’s and St Thomas’s<br />

NHS Foundation Trust, London, United Kingdom; Samsung Medical<br />

Center, Seoul, Korea; Jules Bordet Institute, Brussels, Belgium; The<br />

Royal Marsden NHS Foundation Trust, London, United Kingdom<br />

Background: Optimal endocrine treatment (trt) for post-menopausal<br />

women with ER+ advanced breast cancer (ABC) progressing on or<br />

following a non-steroidal (NS) aromatase inhibitor (AI) is unclear.<br />

The EFECT study showed no difference in efficacy between the<br />

steroidal antiestrogen fulvestrant (F) & steroidal AI exemestane (E)<br />

in this setting (HR=0.96, 95%CI: 0.82, 1.13; p=0.65). Pre-clinical<br />

data suggest F may be more effective in a low estrogen environment.<br />

SoFEA investigated F combined with anastrozole (F+A) in patients<br />

(pts) with acquired resistance to previous AI compared with F alone<br />

& F alone vs. E. The combination of F+A was no better than F<br />

(HR=1.00, 95%CI: 0.83, 1.21; p=0.98) nor F alone better than E<br />

(HR=0.95, 95%CI: 0.79, 1.14; p=0.56); the lack of added benefit for<br />

F+A is consistent with previous 1st-line studies that have assessed<br />

this combination versus A alone (FACT & SWOG-S0226).<br />

Methods: SoFEA is a multi-center partially blinded randomized phase<br />

III study postmenopausal women were allocated to F plus A (F+A<br />

n=243), F plus placebo (n=231) or E (n=249). Similarly, EFECT is a<br />

randomized, double-blind, placebo controlled, multi-center phase III<br />

trial of F (n=351) versus E (n=342) in postmenopausal women (see<br />

table). However, given the differences in prior endocrine therapy/<br />

responsiveness within SoFEA & EFECT populations, an individual<br />

pt meta-analysis combining data from SoFEA & EFECT will be<br />

conducted enabling exploration of putative effects within specific pt<br />

subgroups to establish evidence in support, or not, of a pt subgroup<br />

sensitive to F at the dose used in these trials. Subgroups to be analysed<br />

include receptor status, visceral involvement, AI sensitivity, age,<br />

NSAI setting & time on NSAI.<br />

Results: 723 pts (480 in F & E) were enrolled from 82 UK & 4 South<br />

Korean centers (03/2004-04/2010) in SoFEA. 693 pts were enrolled<br />

from 138 centers worldwide (08/2003-11/2005) in EFECT. Trt was<br />

well tolerated in both trials; serious adverse events were rare. The<br />

meta-analysis will be conducted in July 2012 & results presented.<br />

Conclusion: Combining individual pt data from SoFEA & EFECT<br />

via meta-analysis will provide definitive clinical information on pt’s<br />

response to F at the dose used in these studies, in particular whether<br />

certain pts with acquired resistance to NSAI do experience benefit<br />

of use of this antiestrogen as opposed to E.<br />

SoFEA & EFECT trial designs:<br />

SoFEA (EBCC 2012) EFECT (JCO 2008)<br />

Pts with HR+ ABC who have<br />

progressed or recurred after Pts with HR+ ABC progressing or<br />

Population<br />

NSAI, having demonstrated recurring after NSAI<br />

prior response<br />

Prior NSAI usage (adjuvant/<br />

19% / 81% 12% / 87%<br />

metastatic)<br />

Prior AI sensitivity 100% 63%<br />

Dose<br />

F=250mg monthly (IM) after<br />

500mg loading dose; E=25mg<br />

daily; A=1mg daily<br />

F=250mg monthly (IM) after<br />

500mg loading dose; E=25mg<br />

daily<br />

Median follow-up 38 months (all pts) 13 months (alive pts)<br />

P2-14-02<br />

Preclinical Evaluation of Enzalutamide in <strong>Breast</strong> <strong>Cancer</strong> Models<br />

Cochrane DR, Bernales S, Jacobsen BM, D’Amato NC, Guerrero J,<br />

Gómez F, Protter AA, Elias AD, Richer JK. University of Colorado<br />

Anschutz Medical Campus, Aurora, CO; Medivation Inc., <strong>San</strong><br />

Francisco, CA; Fundación Ciencia & Vida, <strong>San</strong>tiago, Chile<br />

Background: The vast majority of breast cancers express the<br />

androgen receptor (AR) with ∼80% of ER+ and 30% to50% of<br />

ER- tumors being positive for AR by immunohistochemistry (IHC).<br />

Approximately 30% of breast cancer patients with estrogen receptor<br />

positive (ER+) tumors do not respond to tamoxifen or aromatase<br />

inhibitors and AR signaling has been implicated as a possible<br />

mechanism of this insensitivity. AR overexpression has also been<br />

associated with resistance to either tamoxifen or aromatase inhibitors<br />

in cell line models.<br />

Hypothesis: ER+ breast cancers can switch from estrogen to<br />

androgen-dependent growth as they become resistant to traditional<br />

endocrine therapies and AR can serve as a therapeutic target in AR+<br />

breast cancers, irrespective of ER status.<br />

Methods: The proliferative effect of dihydrotestosterone (DHT) and<br />

estradiol (E2) in the presence enzalutamide (formerly MDV3100)<br />

was examined in vitro and in xenograft studies using models of both<br />

ER+ and ER- breast cancer that express AR. Furthermore, AR and<br />

ER were examined by IHC in clinical specimens from breast cancer<br />

patients treated with neoadjuvant exemestane and adjuvant tamoxifen.<br />

Tumor tissue was compared to adjacent normal breast epithelium.<br />

Results: In ER+ models, bicalutamide and enzalutamide both<br />

inhibited DHT-mediated proliferation, while enzalutamide<br />

uniquely inhibited E2-mediated proliferation. In vivo, enzalutamide<br />

www.aacrjournals.org 279s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

significantly reduced estrogen- and androgen-mediated growth of<br />

MCF7 (ER+/AR+) xenograft tumors. Remarkably, enzalutamide<br />

demonstrated an anti-proliferative effect comparable to tamoxifen.<br />

in vitro, enzalutamide inhibits E2-stimulated tumor cell proliferation<br />

and E2 bound ER activation of genes such as PR and SDF-1, despite<br />

no observed binding of enzalutamide to ER alpha or beta. In ER-/<br />

AR+ MDA-MB-453 xenografts, enzalutamide treatment resulted in<br />

decreased nuclear AR localization, increased apoptosis and tumor<br />

growth inhibition.<br />

Conclusions: Enzalutamide demonstrated significant anti-tumor<br />

activity in preclinical models of breast cancers that express AR,<br />

regardless of ER status. When enzalutamide opposes DHT, it<br />

functions by excluding AR from the nucleus, thereby reversing the<br />

anti-apoptotic effect of AR. In contrast, when opposing E2, it causes<br />

a decrease in tumor cell proliferation. Profiling of ER+ xenografts is<br />

underway to further elucidate the mechanism by which this occurs.<br />

P2-14-03<br />

“Ethinylestradiol” is beneficial for postmenopausal advanced<br />

breast cancer patients heavily pre-treated with endocrine agents<br />

Iwase H, Yamamoto Y, Murakami K-I, Yamamoto-Ibusuki M, Tomita<br />

S, Omoto Y. Kumamoto University, Kumamoto, Japan<br />

Purpose and Methods<br />

Estrogen deprivation therapy using aromatase inhibitors is a standard<br />

therapy in postmenopausal hormone-dependent breast cancer.<br />

Paradoxically, low-dose estradiol was reported to be beneficial for<br />

the postmenopausal patients who have been heavily pre-treated with<br />

long-term sequential anti-estrogen therapies. To determine efficacy<br />

and safety of ethinylestradiol (3mg /day oral), a phase 2 study has been<br />

performed for the postmenopausal patients with advanced or recurrent<br />

breast cancer who became resistant to sequential endocrine treatments.<br />

The primary endpoint was clinical benefit rate, secondary were safety,<br />

objective response rate and time to progression. The interim data will<br />

be reported because of the extremely beneficial results.<br />

Results<br />

Eighteen cases with ER-positive tumor which showed resistance to<br />

previous sequential-endocrine therapies including SERMs and/or<br />

aromatase inhibitors were registered from Oct 2010 to May 2012.<br />

Their mean age was 62 years-old and the mean observation time was<br />

9.2 months. Nine cases were evaluated as partial response, 1 case as<br />

long NC, 3 cases as stable disease, and another 2 cases as progressive<br />

disease. In three cases of all 18 cases, the estradiol administration<br />

was withdrawn within 1 week with their early endocrine-related<br />

symptoms, such as nausea, general fatigue and fever. Duration of<br />

effect in the case of the PR (including the 4 ongoing cases) was<br />

more than 24 weeks. All of 9 responders had high ER expression<br />

in the primary or metastatic tumor and had a history of long-term<br />

endocrine therapies in metastatic setting and had response to previous<br />

endocrine therapies. Serum estradiol levels elevated as 30-100pg/mL,<br />

and FSH was suppressed below premenopausal levels in 4 weeks<br />

later administration. Although the weight gain, irregular vaginal<br />

bleeding, or endometrial thickening was observed in patients treated<br />

with long-term treatment, there was no severe adverse event, such as<br />

deep venous thrombosis or other malignancies in this series.<br />

Discussion<br />

The mechanism of low-dose estrogen treatment can be considered<br />

the estrogen-induced apoptosis. The high ER expression and the<br />

response to previous endocrine therapies might be recognized as<br />

predictive factor of this treatment. This low-dose estrogen therapy<br />

may contribute to overturn the common sense of the endocrine therapy<br />

for breast cancer.<br />

P2-14-04<br />

A Phase Ib Dose Escalation Trial of RO4929097 (a γ-secretase<br />

inhibitor) in Combination with Exemestane in Patients with ER<br />

+ Metastatic <strong>Breast</strong> <strong>Cancer</strong>.<br />

Means-Powell JA, Minton SE, Mayer IA, Abramson VG, Ismail-<br />

Khan R, Arteaga CL, Ayers DA, <strong>San</strong>ders MS, Lush RM, Miele L.<br />

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

Center, Tampa, FL; University of Mississippi Health Care <strong>Cancer</strong><br />

Institute, Jackson, MS<br />

Background: RO4929097 is a potent and oral selective inhibitor of<br />

γ-secretase (GSI), producing inhibitory activity of Notch 1-4 signaling<br />

in tumor cells. Preclinical studies have demonstrated synergism<br />

of GSIs in combination with endocrine therapy, in ERα-positive<br />

xenografts. This suggests that optimal treatment of ER+ BC would<br />

benefit from an antiestrogen and GSI combination.<br />

Materials and Methods: To explore the safety, tolerability and<br />

preliminary antitumor activity of Exemestane (25 mg/d) with<br />

R04929097 combination, we conducted a Phase Ib trial in patients<br />

with ER+/ HER2- MBC, whose disease relapsed during treatment<br />

with or within 6 months of discontinuation of an adjuvant nonsteroidal<br />

aromatase inhibitor or Tamoxifen, or whose MBC progressed during<br />

treatment with 1 st or 2 nd line endocrine therapy. Goserelin (3.6 mg q28<br />

days Sub-Q) was given to all premenopausal patients. R04929097<br />

was administered orally daily for 3 consecutive days, followed by<br />

4 days off on a 21 day cycle. Doses of R04929097 were escalated<br />

on a standard 3+3 design over 5 doses (20, 30, 45, 90, and 140 mg).<br />

Patients were fully evaluated for dose limiting toxicity (DLT) for 3<br />

weeks. Patients were treated until disease progression or occurrence<br />

of unacceptable toxicity.<br />

Results: A total of 15 patients were enrolled. Median age was 56;<br />

average number of chemotherapies received in the adjuvant or<br />

metastatic setting was 3.3. Eleven patients had received adjuvant<br />

endocrine therapy. One patient received this as 1 st line endocrine<br />

therapy, eleven patients received this study as 2 nd line endocrine<br />

therapy; and three patients received this study as 3 rd line endocrine<br />

therapy in the metastatic setting. Of the 14 patients evaluable, 1<br />

had a partial response, 6 had stable disease, and 7 had progressive<br />

disease. All patients have discontinued the study at this time, 11 due<br />

to progression, 2 due to toxicity, and 1 withdrew after beginning<br />

protocol therapy. Twenty percent of patients had stable disease for six<br />

months or greater. Of the patients with stable disease for six months,<br />

one patient progressed at 9 months and 2 patients progressed at 10<br />

months. The most common toxicities were nausea (73.3%), anorexia<br />

(60%), hyperglycemia (53.3%), hypophosphatemia (46.7%), fatigue<br />

(66.7%), and cough (33.0%). Grade 3 toxicities were uncommon and<br />

included hypophosphatemia (13.0%) and rash (6.3%). There were no<br />

grade 4 toxicities. Rash was the only DLT and was seen at 140 mg.<br />

Conclusion: The combination of R04929097 and Exemestane was<br />

overall well tolerated, but MTD was not determined as access to<br />

drug ended. Further studies with GSIs in combination with endocrine<br />

therapy are warranted on the basis of a favorable safety profile and<br />

preliminary evidence of stable disease seen in patients with ER+ MBC<br />

refractory to previous endocrine therapies. PK, PD and biomarker<br />

analysis is ongoing.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

280s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 2<br />

P2-14-05<br />

A phase II study of combined fulvestrant and RAD001<br />

(everolimus) in metastatic estrogen receptor (ER) positive breast<br />

cancer after aromatase inhibitor (AI) failure.<br />

Croley JJ, Black EP, Romond E, Chambers M, Waynick S, Slone<br />

S, Waynick C, Stevens M, Weiss HL, Massarweh SA. University of<br />

Kentucky and Markey <strong>Cancer</strong> Center, Lexington, KY<br />

Background: Fulvestrant is used to treat women with metastatic<br />

ER-positive breast cancer after AI failure, but has a short duration<br />

of benefit. Pi3K/mTOR signaling has been implicated in preclinical<br />

models of fulvestrant resistance and recent trials suggest that<br />

everolimus, an oral inhibitor of mTOR, can overcome resistance to<br />

other forms of endocrine therapy. We hypothesized that everolimus<br />

may delay resistance to fulvestrant and prolong time to progression<br />

(TTP).<br />

Methods: We designed a phase II clinical trial of combined fulvestrant<br />

and everolimus in postmenopausal women with ER-positive breast<br />

cancer who relapsed or experienced metastatic disease progression<br />

within 6 months of AI use. Fulvestrant was given at 500 mg<br />

IM on day1, 250 mg d14, 250 mg d28, and monthly thereafter.<br />

Everolimus was given at 10 mg po daily. Patients were required to<br />

have measurable or evaluable disease with preserved performance<br />

status and adequate organ function. Primary endpoint is TTP, and<br />

secondary endpoints are safety, response rate, clinical benefit rate,<br />

and biomarker analysis. A sample size of 40 patients was calculated<br />

to meet a median TTP of 7.0 vs. 3.7 months for fulvestrant alone<br />

as reported in the EFECT trial. Patients were followed monthly for<br />

clinical and toxicity assessment and imaging was obtained every 2<br />

months. Tumor blocks were collected when available and biopsies<br />

were offered if disease was accessible.<br />

Results: To date, 30 patients enrolled on study with a median age of 56<br />

years (range 39-85). Most common metastatic disease sites were bone<br />

in 26 patients (87%), liver in 19 (63%), and lung in 16 (53%). Prior<br />

therapy included tamoxifen in 21 patients (70%), and chemotherapy<br />

in 20 (67%), of those 17 were in the adjuvant/neoadjuvant setting.<br />

6 patients (20%) received more than one AI. 2 patients were ruled<br />

ineligible immediately after enrollment and starting study treatment,<br />

one because of a creatinine level outside the reference range and<br />

one because of the need for palliative radiation. Of the remaining<br />

28 patients, 18 discontinued therapy because of disease progression,<br />

3 because of toxicity, 2 upon patient request, and 1 because of<br />

unrelated intercurrent illness, with 4 patients currently on therapy.<br />

Most common adverse events reported were mucositis in 13 patients<br />

(43%) and rash in 11 (36%). Most common laboratory abnormalities<br />

were elevated ALT/AST in 18 patients (60%), elevated cholesterol in<br />

13 (43%), and hypokalemia in 13 (43%). The majority of toxicities<br />

were grade I/II. Most common grade III toxicities, regardless of<br />

attribution, were infection requiring hospitalization in 3 patients<br />

(10%), hypokalemia in 3 (10%) and mucositis in 2 (7.4%). There was<br />

one grade 4 toxicity reported-hypokalemia. Overall, treatment was<br />

reasonably tolerated and toxicities manageable. Efficacy findings,<br />

including the primary endpoint of TTP, will be analyzed and presented<br />

at the meeting.<br />

Conclusions: Combined everolimus with fulvestrant is feasible and<br />

has manageable toxicities in this cohort of women with metastatic<br />

ER-positive breast cancer. Detailed efficacy analysis along with<br />

updated toxicity data will be presented at the meeting.<br />

P2-14-06<br />

A phase II trial of low dose estradiol in postmenopausal women<br />

with advanced breast cancer and acquired resistance to an<br />

aromatase inhibitor.<br />

Howell SJ, Seif MW, Armstrong AC, Cope J, Wilson G, Welch RS,<br />

Misra V, Ryder D, Blowers E, Palmieri C, Wardley AM. University of<br />

Manchester, United Kingdom; The Christie NHS Foundation Trust,<br />

Manchester, United Kingdom; Imperial College, London, United<br />

Kingdom<br />

Background High dose estrogen (HDE) is an effective but toxic<br />

treatment for postmenopausal women with advanced breast<br />

cancer (ABC). In vitro, prolonged periods of estrogen deprivation<br />

(ED) sensitises cancers to the inhibitory effects of estrogen. We<br />

hypothesised that the profound ED seen with third generation<br />

aromatase inhibitor (AI) would sensitise cancers to LDE which would<br />

be better tolerated.<br />

Methods Single arm phase II study in postmenopausal women<br />

with measurable ER+ ABC and demonstrated clinical benefit (CB)<br />

with a third generation AI. Treatment: estradiol valerate 2mg daily.<br />

Primary endpoint: CB rate (CBR) was correlated with baseline and<br />

dynamic LH/FSH levels and on-treatment estradiol levels. Secondary<br />

endpoints included TTF, PFS, toxicity and QoL. If LDE was effective,<br />

retreatment with the AI on which the cancer had progressed prior<br />

to study entry was offered on progression. If LDE was ineffective<br />

HDE was offered.<br />

Results 21/50 patients were recruited before early closure due to slow<br />

accrual. 19 were assessable for efficacy and toxicity (1 ineligible; 1<br />

no LDE). CBR was 5/19 (26%; 95%CI 9.1%,51.2%). Median TTF<br />

2.8months (range 0.5-29.8). CBR durations were 11.1, 16.8, 17.3*,<br />

19.8, 29.8 months (*censored). 4 stopped treatment early due to<br />

toxicity (G4 hypercalcaemia/ G2 vaginal bleeding (VB) plus G4<br />

hyponatraemia/ G2 mucositis/ G2 headaches) 1 with SD at 3 months<br />

and 3 before response assessment. Other common toxicities were all<br />

G1/2 and mainly limitted to nausea (8/19), breast pain (7/19) and<br />

tumour flare (7/19). VB occurred in 8/11 without prior hysterectomy.<br />

Baseline LH correlated with CBR by logistic regression (p=0.01). Of<br />

3 retreated with the same AI post LDE; 1 had PR, 1 SD ≥6months<br />

and 1 PD. One woman received HDE after failure of LDE and<br />

achieved a PR.<br />

Conclusion LDE is an effective and largely well tolerated treatment<br />

in women with acquired resistance to AI. VB is common without<br />

prior hysterectomy. Rechallenge with the same AI post LDE seems<br />

effective and may offer an extra line of endocrine therapy. This should<br />

be tested in future trials.<br />

P2-14-07<br />

Evaluation of Aromatase Inhibitor failure and total healthcare<br />

expenditures among post-menopausal women with metastatic<br />

ER+/HER2- breast cancer in the US<br />

Namjoshi M, Landsman-Blumberg P, Thomson E, Chu BC. Novartis<br />

Pharmaceuticals Corporation, East Hanover, NJ; Thomson Reuters,<br />

Washington, DC<br />

Aromatase Inhibitors (AIs) have replaced tamoxifen as first-line<br />

therapy for post-menopausal women with metastatic, ER+ breast<br />

cancer (BC). However, little information is available on the realworld<br />

use of AIs. This retrospective administrative claims study<br />

compared healthcare expenditures of post-menopausal women with<br />

metastatic ER+/HER2- BC treated with AIs and experiencing 0 or<br />

≥ 1 AI failures (AIF).<br />

Women ≥ 55 years of age newly diagnosed with stage IV BC (index)<br />

were identified in the 2006-2010 Thomson Reuters MarketScan<br />

www.aacrjournals.org 281s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

databases and followed until the earliest of chemotherapy,<br />

combination therapy, end of enrollment, or end of study (03/31/2011).<br />

ER+/HER2- disease was defined as any endocrine therapy (ET:<br />

tamoxifen, fulvestrant) or AI (anastrozole, letrozole, or exemestane)<br />

use and no trastuzumab, lapatinib or toremifene use anytime in the<br />

6-month pre-or variable length post-index periods. Those with postindex<br />

AI use except where use followed chemotherapy or combination<br />

therapy were retained for analysis. AIF post-index was defined as a<br />

switch to an alternative AI, ET, chemotherapy, or combination therapy.<br />

All-cause healthcare expenditures, defined as total provider payments<br />

(insurer + patient + COB) were examined by type of service and in<br />

total, and expressed as per patient per month (PPPM). A series of loggamma<br />

regressions were used to examine the relationship between<br />

AIF and PPPM expenditures as a form of cost ratios (CRs), adjusting<br />

for potential confounding factors such as patient demographic and<br />

clinical characteristics.<br />

Among 4,249 eligible patients, 36% had ≥1 AIF. At index, AIF<br />

patients were more likely to have liver (8% vs. 5%), lung (11% vs.<br />

8%), and bone metastases (60% vs. 49%), all p


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 2<br />

a secondary on-line screening before contact information is passed<br />

on to the researcher for the enrollment process.<br />

Results: Over 367,000 women have signed up, including survivors<br />

and women without a history of breast cancer, ranging from ages 18<br />

to 100, representing all 50 US states and 49 countries. To date, the<br />

AOW has recruited for 65 studies, both regional and national, that<br />

vary from biomarker and circadian rhythm research to psycho-social<br />

and quality of life studies. With over 73,000 AOW members having<br />

participated in the research process, this method of recruitment has<br />

been found to be effective and efficient. The diversity of the AOW<br />

members has proved beneficial for many studies, such as those<br />

needing to enroll racial/ethnic minorities, women of varying sexual<br />

orientations, or young survivors.<br />

A secondary goal of the AOW is to assist researchers new to research<br />

with human subjects. The AOW has successfully helped researchers<br />

cross the chasm, coaching them on what it takes to transition their<br />

research from animal models to human subjects. The AOW will<br />

guide researchers through IRB submission, patient recruitment, and<br />

sample collection.<br />

Conclusions: The AOW has proved to be a successful resource for<br />

scientists to accelerate accrual, expand the number and diversity of<br />

their subject population and to obtain exactly the type of specimens<br />

they need when they need it. This partnership between women and<br />

scientists has revolutionized research and accelerated efforts to<br />

eradicate breast cancer. The public is ready and willing to partner<br />

with the research community to find the answer to urgent clinical<br />

problems. The DSLRF will launch a new initiative this fall to harness<br />

the public’s desire to be active research participants: The Health of<br />

Women (HOW) Study, an online longitudinal cohort study that will<br />

reduce participant burden with multiple short modules at regular<br />

intervals. Preliminary data from HOW will be available later this year.<br />

P2-15-02<br />

The efficacy of recruitment and retention strategies for research<br />

subjects in an early phase investigator-initiated breast cancer<br />

trial.<br />

Reichow J, Higgins D, Parker S, Childs J, Disis ML, Salazar LG.<br />

University of Washington, Seattle, WA<br />

Background: One of the biggest challenges faced by investigators is<br />

the implementation of effective strategies to improve the recruitment<br />

and retention of research participants. This is especially true for<br />

investigator-initiated, federally funded (e.g. NIH and DOD), early<br />

phase clinical trials that involve the treatment of serious diseases such<br />

as metastatic breast cancer (MBC). These studies may face additional<br />

barriers to participation since patients have often already undergone<br />

maximal treatment and are usually not in a financial position that for<br />

allows the travel and lodging necessary to receive further investigative<br />

treatment. Moreover, efficacy and toxicology in early phase clinical<br />

trials are unknown. Thus, when study budget constraints do not<br />

allow monetary incentives to participation, it is difficult to provide<br />

motivation for patients to enroll and remain adherent to the protocol<br />

requirements. However, many MBC patients are motivated to join<br />

clinical trials for altruistic purposes alone, and evidence supports that<br />

the researcher-patient relationship may be the most important factor in<br />

clinical trial participation. Recognizing that many patients are willing<br />

to participate if provided the appropriate resources despite limited<br />

monetary incentives, we developed a system to improve patient<br />

recruitment and retention to our studies, which are primarily federally<br />

funded. We report here on the strategies developed and used by our<br />

group to recruit and retain patients in a federally-funded investigatorinitiated<br />

phase I/II vaccine study in MBC patients.<br />

Methods: This study was funded by the NIH/NCI and involved<br />

infusion of HER2 specific T cells in HER2+ MBC patients after<br />

completing in vivo priming with a HER2 vaccine. It required 11 visits<br />

to Seattle, Washington. Working with agencies that offer free services<br />

to patients enrolled in clinical trials, a list of available resources was<br />

compiled and a visit flowchart with specific information on travel and<br />

lodging resources (e,g, Angel Flights and ACS sponsorship), local<br />

transportation and entertainment was developed. During screening,<br />

patients were given the list of resources and trial information. An<br />

email system was used to quickly communicate and follow-up with<br />

patients. Eligible patients were given the visit flowchart to help with<br />

their planning of study visits. An enrollment packet was provided<br />

at the first visit with a calendar to keep track of the visit schedule.<br />

Coordination of care between the patient’s primary oncologist and<br />

the research staff was maintained throughout the study.<br />

Results: 17 of 19 patients enrolled were not from Washington State.<br />

Two out-of-state patients withdrew early from the trial for reasons<br />

unrelated to disease progression or toxicity; one subject completed 8<br />

visits and enrolled in another study and the other completed 2 visits<br />

and discontinued the trial to resume chemotherapy.<br />

Conclusion: We have developed a successful system to enroll and<br />

retain patients in a trial requiring multiple study visits. Development<br />

and implementation of site-specific standard procedures are critical to<br />

improve study participation and retention, especially when patients<br />

receive no financial benefit.<br />

P2-16-01<br />

Prospective breast cancer quality evaluation application<br />

developed for native Android tablet and web browser for<br />

improving mutltidisciplinary breast cancer care<br />

Grobmyer SR, Raum N, Sposato V. University of Florida, Gainesville,<br />

FL; University of Florida, Gaineville, FL<br />

Introduction: Easy to implement, prospective and user-friendly<br />

data collection tools are needed to insure optimal breast cancer<br />

care as defined by national quality measures. Prospective quality<br />

measurement can effectively allow rapid identification of gaps in<br />

care and identification for sources of programmatic improvement in<br />

multidisciplinary care.<br />

Methods: Our group has designed a novel prospective quality<br />

assessment tool system, which consists of 3 major integrated<br />

components: 1) a web application for administration and reporting<br />

of the data, 2) a native Android tablet application for real-time data<br />

collection of breast cancer quality parameters during tumor board, and<br />

3) a backend database where parameters are stored and reported on.<br />

Results: The web application is the primary interface for the breast<br />

cancer coordinator (BCC) to enter information for the applicable<br />

Tumorboards. Quality parameters for measurement as determined<br />

by the breast program leadership are entered into the system by the<br />

BCC on the web application. The BCC will also enter medical record<br />

numbers of patients to be presented at each tumor board in the web<br />

based system. In order to maximize work flow and reduce error, the<br />

system was designed to interface with the hospital electronic medical<br />

record for retrieval of patient demographic data based on medical<br />

record number entry.<br />

The Android tablet is the interface to the system during weekly<br />

breast cancer tumor board. The tablet application facilitates real time,<br />

prospective collection of: tumor board attendance, tumor staging,<br />

tumor type, and collection of center chosen and required breast cancer<br />

quality measures. Data collected on the android system is stored in<br />

real time a central and secure database.<br />

www.aacrjournals.org 283s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Real time quality data analysis and graphical display for user defined<br />

time periods is available on the web application and Android tablet.<br />

Required tumor hospital tumor registry data is also compiled in real<br />

time reports to faciliate communication with the hospital tumor<br />

registry.<br />

Conclusions: A novel real time Android based solution has been<br />

developed for real time breast cancer care quality and tumor registry<br />

information. Use of this technology greatly simplifies data collection<br />

required for participation national breast cancer accreditation<br />

organizations such as NAPBC as well as the American Joint<br />

Commission on <strong>Cancer</strong>. Its ease of implementation and use favor its<br />

widespread adoption. Efforts to quantify the impact of this unique<br />

tool on quality of performance in a multidisciplinary breast cancer<br />

program are underway.<br />

P2-16-02<br />

A web-based data management system for a multicenter<br />

international breast cancer oriented blood, tissue, and data<br />

biobank<br />

Margossian AL, Saadjian H, Mira A, Contreras A, Rimawi MF,<br />

Margossian ML, Scheurer M, Osborne K, Gutierrez C. <strong>Breast</strong> Center,<br />

Buenos Aires, Argentina; Dan Duncan <strong>Cancer</strong> Center/Smith <strong>Breast</strong><br />

Center, Baylor College of Medicine, Houston, TX<br />

The quality of biospecimens and associated data must be consistent<br />

and collected according to standardized methods in order to achieve<br />

international harmonization and coordination among biobanking<br />

networks.<br />

Sharing successful strategies between the Baylor College of Medicine<br />

(BCM) <strong>Cancer</strong> Center and <strong>Breast</strong> Center Buenos Aires (BCBA) is<br />

the driving force for the creation of an Argentinean <strong>Breast</strong> <strong>Cancer</strong><br />

oriented Biobank with a shared data management system.<br />

Objective: To create a bilingual web-based tool for biospecimen<br />

management, inventory, clinical and breast cancer data registration<br />

according to international standards of data security quality assurance<br />

(ISO 27001).<br />

Methods / System Characteristics: The system permits users to<br />

enter and retrieve data concerning the collection, storage, quality<br />

assurance and distribution of biospecimens, and provides clinical<br />

and other patient data for samples while maintaining strict patient<br />

confidentiality.<br />

It has a multi-tier architecture, developed using Service-Oriented<br />

Architecture (SOA) software design principles in .net 4.0, and uses a<br />

SQL server database backbone. The system allows multidimensional<br />

data analysis trough On Line Analytical Processes (OLAP) and<br />

operates with standard web clients, such as Internet Explorer.<br />

User Profiles/Access Levels:<br />

• Clinical staff (e.g., clinical research coordinator, phlebotomist,<br />

pathologist, PA, MD) conduct participant and consent registration<br />

and preregistration, epidemiological questionnaire data entry, barcode<br />

label generation, form printing, and clinical and pathological cancer<br />

data entry.<br />

• Biospecimen resource staff (e.g., lab technicians) track and store data<br />

about biospecimen collection, inventory, tracking and distribution,<br />

generate sample barcode labels, and maintain sample allocation and<br />

freezer maps for the system.<br />

• Scientists have the ability to search for biospecimens and associated<br />

data for their own translational research projects.<br />

<strong>Breast</strong> <strong>Cancer</strong> Database: Detailed clinical, pathological, treatment<br />

and follow-up breast cancer information are captured for each patient,<br />

including TNM, biopsy and definitive surgery of primary tumor and<br />

events, systemic and radiation treatment, recurrences and metastasis,<br />

follow-up. A summary screen provides a snapshot of these data to the<br />

viewer. Pathology reports and sample collection forms are attached<br />

in each corresponding biopsy or surgery screen for easy viewing and<br />

quality assurance.<br />

Conclusion: This Biobank Management System was designed by<br />

a multidisciplinary team to improve and streamline the workflow<br />

for each member of the biobank by: allowing preregistration<br />

capabilities before consenting individuals or collecting samples,<br />

automatic generation of clinical and sample ID labels, and real-time<br />

statistics with certified secure sensible information confidentiality.<br />

This system’s value is its wide range of uses, from the day-to-day<br />

management of a multicenter biobank to the storage of detailed<br />

clinical cancer-related data in a user-friendly and intuitive data entry<br />

environment.<br />

P2-16-03<br />

Creation of a “state-of-the-art” breast cancer data and biobank<br />

in Argentina<br />

Margossian AL, Gutierrez C, Saadjian H, May M, Ohanessian R,<br />

Bacigalupo S, Baravalle S, Margossian J, Osborne KC, Scheurer<br />

ME. <strong>Breast</strong> Center, Buenos Aires, Argentina; Dan Duncan <strong>Cancer</strong><br />

Center/Smith <strong>Breast</strong> Center, Baylor College of Medicine, Houston, TX<br />

Translational cancer research is highly dependent on having large<br />

series of cases with high-quality samples that were collected,<br />

processed, stored and annotated in a systematic fashion according<br />

to international standards, such as the ISBER Best Practices for<br />

Biorepositories. Sharing successful strategies between the Baylor<br />

College of Medicine (BCM) <strong>Cancer</strong> Center and <strong>Breast</strong> Center Buenos<br />

Aires (BCBA), we created this Argentinean biobank complete with<br />

breast cancer-oriented data, tumor and normal breast tissue and<br />

matched blood specimens according to best practices to facilitate<br />

international collaborative translational breast cancer research.<br />

Objective: Creation of a blood, breast tissue and tumor biobank in<br />

Argentina for research purposes with associated epidemiological,<br />

pathological, clinical, and follow-up data.<br />

Methods: Consented individuals from BCBA are classified by treating<br />

physician in four categories: 1) breast cancer; 2) benign breast disease<br />

by biopsy; 3) high-risk for breast cancer according to the Gail Model;<br />

and 4) healthy controls. Blood is collected at several time points<br />

during the breast cancer disease process: pre-surgical, pre-systemic<br />

treatment, and, if applicable, in the metastatic setting. Blood products<br />

are stored as whole blood, plasma, buffy coat, red blood cell pellet,<br />

serum and clot at -80°C or at room temperature in GenPlates. Fresh<br />

tissue and tumor sample is collected during surgical or core biopsy<br />

proceedings and stored at -80°C and paraffin embedded. Upon<br />

enrollment, participants complete an extensive epidemiological and<br />

risk factor questionnaire, which is supplemented by medical record<br />

abstraction for relevant pathological and clinical data. Participants<br />

are also re-contacted once a year for follow-up.<br />

The creation of this biobank in 2009 included the preparation and<br />

adaptation of the following processes from the Population Science<br />

Biorepository and Smith <strong>Breast</strong> Center Tumor Bank at BCM,<br />

including: 1) IRB-approved informed consent documents with specific<br />

language relevant to DNA-based research; 2) Epidemiological and<br />

risk factor questionnaires (10-page Core module and 6-page <strong>Breast</strong><br />

module); 3) Blood and tissue collection protocols, sample processing,<br />

sample bar coding and participant/sample registration system; and<br />

4) Web-based data management system, specifically designed for<br />

this biobank with encryption and data security (details presented<br />

in a separate abstract), including role-based access levels personal<br />

health information.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

284s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 3<br />

Conclusion: We started our endeavor in August 2008. The<br />

development of the consent forms, two questionnaires, and biobank<br />

database prototype took one year at BCM. The translation of consents<br />

and questionnaires into Argentinean Spanish took an additional 3<br />

months. Overall the Argentinean IRB approval process, training<br />

personnel, and equipment set-up lasted for 14 months. By April 2011,<br />

the first Argentinean participant was consented into the biobank. It<br />

took nearly 3 years from inception to realization for this biobank;<br />

however, the potential benefit to translation breast cancer research is<br />

large. The overall value of this biobank will depend on the number<br />

of individuals/samples accrued and the number of years of followup<br />

attained.<br />

P2-16-04<br />

Attitudes of metastatic breast cancer patients towards research<br />

biopsies<br />

Seah DS, Scott SM, Najita J, Openshaw T, Krag KJ, Frank E, Sohl<br />

J, Stadler ZK, Garrett M, Winer EP, Come S, Lin NU. Dana-Farber<br />

<strong>Cancer</strong> Institute, Boston, MA; Beth Isreal Deaconness Medical<br />

Center, Boston, MA; <strong>Cancer</strong> Care of Maine, Brewer, ME; Mass<br />

General North Shore <strong>Cancer</strong> Center, Danvers, MA; Memorial Sloan-<br />

Kettering <strong>Cancer</strong> Center, New York, NY<br />

Background:<br />

In the era of molecularly targeted therapy, developing an understanding<br />

of the molecular basis of cancer is a principal or secondary goal of<br />

many research studies. For this reason, studies collecting tissue for<br />

research purposes are increasingly common. Understanding patients’<br />

attitudes towards research biopsies may lead to improvement in<br />

accrual to research biopsy studies.<br />

Methods:<br />

Patients with metastatic breast cancer from two academic and two<br />

community hospitals completed a self-administered paper survey<br />

consisting of 29 questions in clinic to evaluate their willingness to<br />

consider providing additional biopsies (additional biopsy performed<br />

with a clinically indicated biopsy) and research purposes only biopsies<br />

(RPOB) (research biopsy performed as a stand alone procedure).<br />

Results:<br />

160 patients (n=80 academic, n=80 community) completed the survey,<br />

with a response rate of 98%. As expected, demographic variables<br />

differed between sites, with patients from academic sites likely to<br />

be younger (P=0.01), more educated (P=0.002), employed (P=0.01),<br />

have prior trial participation (P


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

1.88 (1.05-3.37) (adjusted for age, body mass index-BMI kg/m2)).<br />

Conclusion: Our study supports that insulin, IGF-1 and IGFBP-3<br />

independently, and in combination with cycling estrogen, predicts<br />

premenopausal mammographic density. These hormones may be<br />

important biomarkers in breast cancer control and clinical practice.<br />

P3-01-02<br />

Correlation of Mammographic breast density and tumor<br />

characteristics in Korean breast cancer patients<br />

Cho JY, Ahn SH, Lee JW, Yu JH, Koh BS, Kim HJ, Lee JW, Son BH,<br />

Gong G-y, Kim HH. College of Medicine, University of Ulsan, Asan<br />

Medical Center, Seoul, Republic of Korea<br />

Introduction: Western studies have demonstrated high breast density<br />

as a strong risk factor for breast cancer, it is poorly understood whether<br />

breast density affects the diverse phenotypes of breast cancer. We<br />

examined the association between various tumor characteristics and<br />

mammographic breast density in women with breast cancer. Methods:<br />

We conducted a cross-sectional analysis in 910 Korean women<br />

diagnosed with breast cancer to evaluate the associations between<br />

breast density and tumor size, lymph node status, lymphovascular<br />

invasion, histologic grade, estrogen receptor, progesterone receptor ,<br />

HER2. <strong>Breast</strong> density was classified as fatty (percent density less than<br />

50% by a computer-assisted thresholding program, named “Cumulus<br />

TM<br />

”; n = 470) or dense (percent density 50% or more; n = 440) for<br />

the cancer-free breast at the time of operation. Logistic regression<br />

was used to examine whether the relationships were modified by<br />

adjustment for body mass index, age at diagnosis, age at first birth,<br />

menopausal status, history of breast-feeding, and breast cancer<br />

staging. Results: Total 910 patients were involved, the mean age and<br />

median age at the operation was 48 years old (range 20-82), and the<br />

mean percent density was 48.09 (SD = 9.62 %: normally distributed,<br />

Kolmogorov-Smirnov test p=0.32). Crude analysis shows that tumor<br />

size over than 0.5cm were more likely to have dense breasts compared<br />

with women with a tumor size


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 3<br />

differences were observed in BC staging when we compared the A<br />

and B density with C and D (p=1.0). The same occurred with the<br />

genomic classification (p=0.48).<br />

Conclusion: Our screening population has a predominance of low<br />

mammogram densities, probably due to the high body mass index<br />

observed in previous studies of this cohort. With respect to cancer<br />

cases, the absence of statistical significance results suggests that<br />

longer follow up will be necessary for definitive conclusions.<br />

Density Stage 0/I % Stage II % Stage III % Total<br />

1 0 0 3 75 1 25 4<br />

2 5 83,3 1 16,7 0 0 6<br />

3 3 50 2 33,3 1 16,7 6<br />

4 1 100 0 0 0 0 1<br />

Table 1. <strong>Cancer</strong> staging versus breast density.<br />

Density Luminal A % Luminal B %<br />

Triple<br />

Negative<br />

% HER-2 % Total<br />

1 0 0 2 50 1 25 1 25 4<br />

2 4 66,7 2 33,3 0 0 0 0 6<br />

3 2 33,3 4 66,7 0 0 0 0 6<br />

4 1 100 0 0 0 0 0 0 1<br />

Table 2. Genomic classification versus breast density.<br />

P3-01-04<br />

Surgical management of mammographic microcalcification is<br />

improved by full field digital mammography (FFDM)<br />

Bundred SM, Zhou J, Hurley E, Wilson M, Morris J, Bundred NJ.<br />

University Hospital of South Manchester; University of Manchester<br />

Background: Preoperative diagnosis of impalpable breast lesions<br />

correlates closely with the number of surgical procedures required<br />

for treatment. Correct diagnosis of mammographic microcalcification<br />

as DCIS or invasive cancer is important because lesions upgraded to<br />

malignant diagnosis at surgery require repeat surgical procedures in<br />

44% of cases (BASO Audit 2011).<br />

Methods: To determine the impact of FFDM (FFDM only since April<br />

2010) on diagnostic accuracy, positive predictive value (PPV) and<br />

surgical management of MM. Screening and symptomatic women<br />

with mammographic microcalcification (n=1125) in one visit were<br />

reviewed. Demographic information; pre and post operative diagnosis<br />

and number of surgical procedures were recorded for pre FFDM (8/<br />

2007 to 3/2010: n=710) and post FFDM (4/2010 to 5/2011: n=413).<br />

Results: Overall 299 (106 invasive) and 144 (62 invasive) malignant<br />

lesions were diagnosed before and after FFDM introduction.<br />

Mammography SFM n=711 FFDM n = 413 p<br />

Positive Predictive value 42.6% 303 34.6% 143


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P3-01-06<br />

Ultrasound as single mode of imaging may miss significant<br />

pathology in symptomatic women aged 35-39<br />

Baker EL, Masudi T, Waterworth A. Calderdale and Huddersfield NHS<br />

Foundation Trust, Huddersfield, West Yorkshire, United Kingdom<br />

Recent guidelines in the UK stipulate that symptomatic women aged<br />

under 40 with clinically benign disease do not need mammography as<br />

a standard part of their assessment. 1 Previous practice at the Calderdale<br />

and Huddersfield NHS Trust has been to perform mammography on<br />

all women over 35 years old. This study was undertaken to determine<br />

if there would be an increase in false negative assessment under the<br />

new guidelines.<br />

All female patients 35-39 years old attending symptomatic breast<br />

clinic with benign clinical examination and undergoing both<br />

ultrasound and mammogram evaluation were eligible. Patients with<br />

previous breast cancer or a clinically suspicious lump mandating<br />

mammography were excluded. The information was gathered<br />

retrospectively using patient records. 96 women were included.<br />

Mammography and ultrasound results (including M and U valves)<br />

were compared for any discrepancy. If performed, biopsy results<br />

were also reviewed.<br />

In 70% of patients there was no discrepancy between mammographic<br />

and ultrasonic evaluation (M=U). In 25% U score was one higher than<br />

M score. On further analysis this group mainly comprised of those<br />

with fibroadenomas or fibrocystic breast changes seen on ultrasound<br />

(U2) with a normal mammogram (M1).<br />

In the remaining 5%, 3 patients had U score 2 greater than M score,<br />

1 had M>U by 1 and 1 had M>U by 2. On further analysis of these<br />

discrepancies most patients had benign disease. In two patients<br />

with U5 scores and lower M scores malignancy was confirmed on<br />

biopsy. In one patient ultrasound showed benign disease (U2) but<br />

mammogram demonstrated suspicious calcifications (M4) that were<br />

confirmed as high grade DCIS on stereo-core biopsy.<br />

In conclusion for the vast majority of patients in this group<br />

mammography does not add to the evaluation on presentation at<br />

symptomatic breast clinic. However this study identified a case (1<br />

patient in 96) where significant disease could have been missed had<br />

a mammogram not been undertaken. The acceptable level of such<br />

false negative assessments with ultrasound only imaging in women<br />

under 40 must be further evaluated.<br />

1<br />

Best practice diagnostic guidelines for patients presenting with<br />

breast symptoms Alexis M Willett, Michael J Michell, Martin J R<br />

Lee, November 2010<br />

Printed by Breakthrough <strong>Breast</strong> <strong>Cancer</strong>, www.dh.gov.uk/publications<br />

P3-01-07<br />

ESTIMATED RISK OF RADIATION-INDUCED BREAST<br />

CANCER FROM MAMMOGRAPHIC SCREENING<br />

Freitas-Junior R, Correa RS, Peixoto J-E, Ferreira RS, Tanaka<br />

RMN. Federal University of Goias, Goiania, Goias, Brazil;<br />

Comissão Nacional de Energia Nuclear, Goiania, Goias, Brazil;<br />

National <strong>Cancer</strong> Institute of Brazil, Rio de Janeiro, RJ, Brazil;<br />

Superintendência de Vigilância em Saúde do Estado de Goias,<br />

Goiania, Goias, Brazil<br />

Objective: Estimate the benefit–risk balance of mammography, as<br />

number of lives saved/lost in an opportunistic screening in the state<br />

of Goiás, Brazil, in 2010, according to type of technology available<br />

and age group indicated for screening. Methods: The number of lives<br />

saved was estimated considering the gross mortality rate for breast<br />

cancer, female population living in the state, estimated mortality rate<br />

reduction due to screening programs, and mammographic coverage in<br />

the state. For number of lives lost, the model adopted by the Biological<br />

Effects of Ionizing Radiation (BEIR VII) was used. Based on the mean<br />

glandular tissue dose (Dg), the following parameters were calculated:<br />

number of radiation-induced cancer cases with one exposure, with<br />

several exposures in screening programs and over the lifetime, number<br />

of deaths caused by radiation-induced cancer at a certain age and over<br />

the lifetime. Dg of each equipment was estimated according to the<br />

European protocol, using the incident air kerma (Ki) at the simulator<br />

entrance and the half-value layer (HVL) of the X-ray beam, and<br />

table values of coefficients to convert Ki into Dg. Ki and HVL were<br />

measured in 100 equipments in operation in the state. A standard breast<br />

simulator (5.3 cm thickness and 50% glandular tissue) was used. We<br />

considered a biannual mammographic screening with an exam routine<br />

composed of a cranio-caudal and a mediolateral oblique view and<br />

40–70 and 50–70-year age groups. Results: The mean Dg was 4.28<br />

(±1.06) and 6.61 (±3.67) for conventional and digital equipments,<br />

respectively (p


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 3<br />

Method: Conducted digitizer mammogram density assessment using<br />

quantitative rating system based on computer-assisted methods<br />

of measurement with cumulus program based on interactive<br />

thresholding. Polymorphism identification ESR1 XbaI and PvuII<br />

RFLP obtained through PCR process. The numbers of genotyped<br />

cases and controls for each marker were 50 cases and 58. Mean of<br />

density and frequencies of SNPs were compared between cases and<br />

controls to identify SNPs associated with cancer susceptibility. Using<br />

anova, independent t-test, cruskal wallis and mann whitney u test we<br />

calculated significant different between SNP genotype group.<br />

Results: The mean of mammographic density is higher in cases<br />

(52%) than in controls (0,41%) (p0,05). Women with one or two copies of the XbaI x allele had<br />

higher mean percentage density (AG/Xx and AA/xx, 49% and 47%,<br />

respectively) than those with the GG/XX genotype (32%,), AAVGG<br />

P 0.6, no distant metastasis observed at 5 years in the original 78<br />

patients with no adjuvant systemic treatment.) The aim of this study<br />

is to determine the proportion of biologically low and ultralow risk<br />

tumors among the screen-detected tumors. Second, to evaluate the<br />

impact of analog versus the more sensitive digital screening technique<br />

on the biology of tumors detected.<br />

Methods All Dutch breast cancer patients enrolled in the MINDACT<br />

trial (EORTC-10041) accrued 2007-2011, who were invited for<br />

the Dutch screening program (biennial, age 50-75), were included<br />

(n=1381). The proportions of 70-gene signature high, low and<br />

ultralow risk were calculated for patients with screen detected (n=<br />

694), interval (n=366), and symptomatic, non-screening (n=321)<br />

carcinomas, taking into account analog vs. digital technology. Covariants,<br />

such as age, tumor size, histological type, ER, progesterone<br />

receptor (PgR) and HER2/neu-oncoprotein (ERBB2) will be<br />

included in a multinominal logistic regression model to assess both<br />

confounding and effect modification by technology type; these data<br />

are pending and will be available during SABCS 2012.<br />

Results<br />

Proportion of 70-gene signature ultralow, low and high risk patients in all subgroups<br />

Ultralow risk Low risk High risk Total<br />

Chi Square<br />

Test<br />

Screen-detected carcinomas 240 (34,6%) 225 (32,4%) 229 (33%) 694 0,001<br />

Interval carcinomas 105 (28,7%) 99 (27%) 162 (44,3%) 366<br />

Non-screening related<br />

carcinomas<br />

108 (33,6%) 96 (29,9%) 117 (36,4%) 321<br />

Total 453 (32,8%) 420 (30,4%) 508 (36,8%) 1381<br />

Among the screen-detected tumors, 33% had a high risk 70-gene<br />

signature tumor and 67% had a low risk tumor (table 1). A significant<br />

difference was seen between the screen-detected carcinomas and<br />

interval carcinomas (p X 2 test = 0.001) in all 70-gene signature risk<br />

groups. Among the screen detected carcinomas, 41.5% was detected<br />

using analog mammography and 58.5% was detected using digital<br />

mammography. Analog mammography detected 26.4% high risk,<br />

31.9% low risk and 41.7% ultralow risk tumors. This is significantly<br />

different using digital mammography (p X 2 test = 0.001), which<br />

detected 37.7% high risk, 32.8% low risk and 29.6% ultralow risk<br />

tumors.<br />

Conclusion Screen detection was found to be associated with a higher<br />

likelihood of a biologically low risk tumor. Half of all screen-detected<br />

low risk tumors had an index score above 0.6, indicating an ultralow<br />

risk of distant metastasis. The transition from analog to digital<br />

mammography resulted in a decrease in detection of ultralow risk<br />

tumors and an increase in the detection of high risk tumors. Screendetected<br />

low risk biology tumors are frequent and comprehension<br />

may aid to minimize overtreatment.<br />

P3-02-02<br />

Use of contrast-enhanced computed tomography in clinical<br />

staging of asymptomatic breast cancer patients to detect<br />

asymptomatic distant metastases.<br />

Tanaka S, Sato N, Fujioka H, Takahashi Y, Kimura K, Iwamoto M,<br />

Uchiyama K. Osaka Medical College, Takatsuki City, Osaka, Japan<br />

Objective: The use of computed tomography (CT) with regards to the<br />

clinical staging of breast cancer (BC) patients has been on the increase<br />

in clinical practice. However, NCCN guidelines recommended the<br />

use of imaginng only in cases with locally advanced disease or signs<br />

of distant metastases (DM), and the benefits of routine CT have yet<br />

to be fully clarified. This study investigated the value of employing<br />

contrast-enhanced CT (CECT) to screen for DM in patients with<br />

asymptomatic BC.<br />

Methods: The clinical records of 483 patients with asymptomatic BC<br />

who underwent CECT, also in order to detect BC spread, between<br />

April 2006 and January 2011 were reviewed. The CECT results were<br />

classified into normal, true-positive (metastases) or false-positive<br />

findings.<br />

Results: Abnormal CECT findings, including true- and false-positive<br />

results, were detected in 65 patients (13.5%). Of these, 26 patients<br />

(5.4%) showed confirmed true metastatic disease, including 18 lung<br />

metastases, 11 liver metastases and 13 bone metastases. Upstaging to<br />

stage IV due to the results of the CECT was significantly associated<br />

with only larger tumos size (odds ratio, 33.4; 95% CI 12.1-92.5;<br />

P


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Clinical factors associated with upstaging to stage IV by CECT.<br />

n (%)<br />

Upstaged cases (true-positive) Others (normal/false-positive) P-value<br />

Age (years)<br />


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 3<br />

screen detected HER2 +ve cancers in this cohort are core grade I &<br />

II, node negative and ER +ve. Data will be presented on the systemic<br />

management and the outcome of these cases.<br />

P3-02-05<br />

Subtypes of screen-detected invasive breast cancer and<br />

symptomatic invasive breast cancer and their impact on survival<br />

Kobayashi N, Hikichi M, Miyajima S, Utsumi T. Fujita Health<br />

University, Toyoake, Aichi, Japan<br />

Background: <strong>Breast</strong> cancer is a worldwide problem which urgently<br />

requires solution, in many countries still being the most common<br />

cause of malignancy associated death. The screening program aims<br />

to detect early breast cancer to improve survival. We investigated<br />

the clinicopathrogical characteristic including intrinsic subtypes<br />

and outcome of patients with screen-detected invasive breast cancer<br />

(S-DIBC) compared with those with symptomatic invasive breast<br />

cancer (SIBC).<br />

Material and Methods: From January 2005 to December 2011 840<br />

patients with invasive breast cancer who underwent surgery at our<br />

hospital were included. There were 195 S-DIBCs and 645 SIBCs.<br />

We retrospectively reviewed the clinical and pathologic data. Ki-67<br />

LI was categorized as low ( or =15%). Tumors<br />

were classified into four groups based on the expression of ER,<br />

PgR, Ki-67 and HER2 as follows: luminal A (ER+ and/or PR+, and<br />

HER2- and Ki67 low), luminal B (ER+ and/or PR+, and HER2+ or<br />

Ki67 high), HER2 disease (ER-, PR-, HER2+), and triple negative<br />

(ER-, PR-, HER2-). Overall survival (OS) and relapse-free survival<br />

(RFS) curves were generated using the Kaplan- Meier method.<br />

Survival comparisons were made with the log-rank test, the level of<br />

significance was taken to be 0.05. SPSS 18.0 software package was<br />

used for statistical analysis.<br />

Results: S-DIBC was associated with smaller tumor size, less lymph<br />

node involvement, and earlier stage compared with SIBC (P < 0.001).<br />

Significantly more tumors were positive for hormone receptors in<br />

the S-DIBC group as compared to the SIBC group (ER+,83.5%<br />

vs.75.1%, P= 0.017; PgR+, 73.4% vs. 60.6%, P = 0.009), with a<br />

greater proportion of the luminal A subtype in the S-DIBC group<br />

(S-DIBC: Luminal A 59.0%, Luminal B 25.6%, HER2 disease 4.6%,<br />

triple negative 10.8%; SIBC group: Luminal A 46.5%, Luminal B<br />

30.0%, HER2 disease 6.6%, triple negative 16.8%). Patients with<br />

S-DIBC had better prognosis [5-year OS: 100% (S-DIBC) vs. 95.5%<br />

(SIBC), p=0.005, 5-year RFS: 98.5 vs. 92.1%, p=0.004]. Tumors<br />

detected with screening generally had more favorable outcomes than<br />

symptomatic cancers regardless of subtypes [5-year OS: (S-DIBC)<br />

Luminal A 100%, Luminal B 100%, HER2 disease 100%, triple<br />

negative 100%; (SIBC) Luminal A 95.9%, Luminal B 93.3%, HER2<br />

disease 95.5%, triple negative 84.1%)].<br />

Conclusions: Our results suggest that subtype distribution of S-DIBC<br />

differs from that of SIBC.<br />

P3-02-06<br />

Survival impact of early detection of recurrence after surgery in<br />

early breast cancer patients<br />

Hojo T, Tamura K, Masuda N, Inoue K, Kinoshita T, Fujisawa T,<br />

Hara F, Saji S, Asaga S, Anan K, Yamamoto N, Wada N, Takahashi<br />

M, Nakagami K, Kuroi K, Iwata H. National <strong>Cancer</strong> Center Hospital,<br />

Tokyo, Japan; Osaka National Hospital, Osaka, Japan; Saitama<br />

<strong>Cancer</strong> Center, Saitama, Japan; Gunma Prefectural <strong>Cancer</strong> Center,<br />

Gunma, Japan; Shikoku <strong>Cancer</strong> Cente, Shikoku, Japan; Kyoto<br />

University Graduate School of Medicine, Kyoto, Japan; Kitakyushu<br />

Municipal Medical Center, Kitakyushu, Japan; Chiba <strong>Cancer</strong> Center,<br />

Chiba, Japan; National <strong>Cancer</strong> Center Hospital East, Chiba, Japan;<br />

Hokkaido <strong>Cancer</strong> Center, Hokkaido, Japan; Shizuoka General<br />

Hospital, Shizuoka, Japan; Tokyo Metropolitan <strong>Cancer</strong> and Infectious<br />

diseases Center Komagome Hospital, Tokyo, Japan; Aichi <strong>Cancer</strong><br />

Center Hospital, Nagoya, Aichi, Japan<br />

Background and Object: Annual mammography and physical<br />

examination as the follow-up tests after surgery were recommended<br />

to early breast cancer patients based on the two randomized clinical<br />

trials (GIVIO and Rosselli Del Turco) which were reported in 1990s.<br />

Whereas, radiological imaging and blood test (serum tumor marker)<br />

for early detection of recurrence are not recommended due to the<br />

lack of evidence from clinical trial. However, the imaging techniques<br />

(helical CT, bone scan, PET/CT. MRI et al) to detect minute lesions and<br />

therapeutic options for metastatic breast cancer have been remarkably<br />

advanced since then. In fact, routine radiological examinations after<br />

surgery were performed in several Japanese hospitals for aiming early<br />

detection of recurrence as the clinical practice.<br />

We here evaluate the possible benefit of early detection of recurrence<br />

by radiological and laboratory examinations during post-operative<br />

follow-up period.<br />

Methods: Clinical information of breast cancer patients who were<br />

diagnosed as recurrence after surgery during 2005-2006 was collected<br />

from 30 hospitals in Japan. Clinical and pathological characteristics<br />

such as molecular subtype of breast cancer, survival time from<br />

initial therapy or 1 st recurrence, detection methods and symptomatic<br />

information when they diagnosed as metastasis were analyzed<br />

retrospectively.<br />

Results: As the routine examination of post-operative follow-up,<br />

serum tumor maker, chest x-ray/CT, abdominal US/CT and bone<br />

scan were done in 95%, 57%, 38%, 24% of 30 hospitals, respectively.<br />

Of the 698 patients individually evaluated in this analysis, 248 had<br />

loco-regional recurrences and 450 had distant metastases. The first<br />

distant metastatic site were 35% in bone, 30% in lung, 17% in liver<br />

and 11% in lymph node, respectively. All individual patients are<br />

divided into symptomatic (45.7%) or asymptomatic groups (54.3%) at<br />

the detection of metastases. Asymptomatic metastases were detected<br />

by serum tumor marker (26%), bone scan (18%), chest x-ray (17%),<br />

chest CT (17%), abdominal US (11%) and abdominal CT (5%),<br />

respectively. The median disease-free interval (DFI) was 3.0 years<br />

in both groups, but the median survival time after the diagnosis of<br />

recurrence to death were 3.7 years in asymptomatic patients and 3.0<br />

years in symptomatic patients, respectively. In addition, asymptomatic<br />

group had significantly superior overall survival (from primary<br />

surgery to death) than symptomatic group with oligo-metastases<br />

such as limited organ disease (P


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P3-02-07<br />

Rates of <strong>Cancer</strong> Detection and Abnormal Results among Clients<br />

Recruited for Mammography through Outreach and Education<br />

Supported by the Avon <strong>Breast</strong> Health Outreach Program<br />

Rose J, Opdyke KM, Gates-Ferris K, Hurlbert M. Cicatelli Associates<br />

Inc. (CAI), New York, NY; Avon Foundation for Women, New York, NY<br />

Background:<br />

The Avon <strong>Breast</strong> Health Outreach Program (BHOP) supports<br />

community-based organizations and safety-net providers in<br />

conducting outreach and education to recruit underserved, low-income<br />

and uninsured women for breast cancer screening mammography.<br />

Funded organizations report mammography outcomes as part of<br />

routine program monitoring. NCI <strong>Breast</strong> <strong>Cancer</strong> Consortium data for<br />

2009 reported a cancer detection rate of 3.92 per 1,000 for screening<br />

mammograms and 33.21 per 1,000 for diagnostic mammograms. In<br />

CY2009, CDC’s National <strong>Breast</strong> and Cervical <strong>Cancer</strong> Early Detection<br />

Program (NBCCEDP) reported 14 percent of mammograms had<br />

abnormal results requiring further investigation, with an overall cancer<br />

detection rate of 10.2 per 1,000 mammograms.<br />

Objective:<br />

To describe reported breast cancer detection rates and rates of<br />

abnormal mammography findings among Avon BHOP-funded<br />

programs.<br />

Methods:<br />

We reviewed routinely reported program outcomes data for 98<br />

organizations funded continuously through the Avon BHOP for a<br />

three-year period from 2009 to 2011. Data for 17,839 mammograms<br />

were available in aggregate by year by grantee agency including<br />

the number of mammograms reported, number with a preliminary<br />

abnormal finding, and number of confirmed cancer diagnoses.<br />

Results:<br />

Agencies reported an average of 981 mammogram outcomes per year<br />

over 3 years (range 108 to 5,946 by agency). The average agencyspecific<br />

rate of abnormal findings across 3 years was 13.2% (median<br />

11.2%; range 0.49% to 51.0%). The average agency-specific cancer<br />

detection rate was 8.1 per 1,000 (median 6.4 per 1,000; range zero to<br />

31.7 per 1,000). 28 organizations had 3-year average cancer detection<br />

rates in excess of 10 per 1,000, and 6 had rates exceeding 20 per<br />

1,000. Large fluctuations in the proportion of mammogram outcomes<br />

reported as abnormal by a given agency year-over-year were common.<br />

Conclusion:<br />

Avon BHOP agencies reported overall abnormal and cancer detection<br />

rates similar to those of the NBCCEDP, but higher than the general<br />

population. Fluctuations in year-over-year rates were common, and<br />

may indicate changes in the way program data were reported over<br />

time, changes in screening practices (e.g. new clinical providers or<br />

new equipment), and/or differences in breast cancer risk among clients<br />

recruited for screening over time. High rates of abnormal screening<br />

results could indicate delays in obtaining results from follow-up<br />

diagnostic testing, problems with the calibration of equipment, or<br />

other quality issues.<br />

Discussion:<br />

Grantees with unusually high or low rates of abnormal or cancer<br />

outcomes or significant year-over-year rate fluctuations would benefit<br />

from technical assistance to identify and explain the underlying<br />

causes of these trends. Ensuring that all clients recruited for breast<br />

cancer screening receive high quality mammography services –<br />

including accurate imaging and reliable interpretation by experienced<br />

radiologists – is critical to minimize client anxiety and to ensure that<br />

screening results in improved health outcomes.<br />

P3-02-08<br />

Is repetition of the contralateral mammogram of patients referred<br />

from breast cancer screening for unilateral findings necessary?<br />

Castro C, Schipper R-J, van Roozendaal L, van Goethem M, Lobbes<br />

M, Smidt M. Maastricht University Medical Center, Maastricht,<br />

Netherlands; Antwerp University Hospital, Antwerp, Belgium<br />

Introduction - The Netherlands started a nationwide breast cancer<br />

screening program in 1989, including women from the age of 50 until<br />

75. Screening mammograms are performed two yearly in a mobile<br />

unit and consist of a bilateral two-view mammogram of the breast<br />

(mediolateral oblique and craniocaudal views).<br />

If indicated, patients are referred to a breast clinic for diagnostic<br />

analysis. This work-up consist of among others repeating the bilateral<br />

two-view mammogram. Since the screening is digitalized, repeating<br />

of at least the mammogram of the non suspicious side might be<br />

unnecessary.<br />

The aim of this study is to determine the additional value of repeating<br />

the contralateral mammogram in patients referred for a suspicious<br />

unilateral lesion.<br />

Material and methods – 395 patients were referred from breast<br />

screening program to our institution for unilateral findings between<br />

October 2009 and August 2011. In all patients a bilateral mammogram<br />

was repeated and analyzed by an experienced breast radiologist. In<br />

the case of breast cancer a breast magnetic resonance imaging (MRI)<br />

was performed for preoperative staging. Anonymised data concerning<br />

the date of registration of the screening mammogram, the referred<br />

side (left/right or bilateral), age, screening’s BI-RADS classification,<br />

breast density, biopsy results and breast MRI results were collected.<br />

Results - Of the 395 patients referred for a suspicious unilateral<br />

finding, a malignancy on the referred side was observed in 144 patients<br />

(36.5%). In five patients bilateral breast cancer was detected. In one<br />

patient no malignancy was detected on the referred side, though on<br />

the contralateral side. Three of these six contralateral malignancies<br />

were directly mammographically detected. All six malignancies were<br />

detected with preoperative breast MRI.<br />

Conclusion - Repetition of the two-view mammogram of the<br />

contralateral side in patients referred with a unilateral suspicious<br />

finding seems unnecessary, since all contralateral malignancies<br />

were depicted on the preoperative staging breast MRI recommended<br />

according to the EUSOBI-guidelines. Omission of the contralateral<br />

mammogram could lead to reduction of associated health care costs,<br />

radiation exposure, and patient discomfort caused by the exam.<br />

P3-02-09<br />

Cost-effectiveness of screening with additional MRI for<br />

women with familial risk for breast cancer without a genetic<br />

predisposition<br />

Saadatmand S, Heijnsdijk EA, Rutgers EJ, Hoogerbrugge N,<br />

Oosterwijk JC, Tollenaar RA, Hooning M, Obdeijn I-M, de Koning<br />

HJ, Tilanus-Linthorst MM. Erasmus Medical Center, Rotterdam,<br />

Netherlands; The Netherlands <strong>Cancer</strong> Institute, Antoni van<br />

Leeuwenhoek Hospital, Amsterdam, Netherlands; Radboud University<br />

Medical Center, Nijmegen, Netherlands; University Medical Center<br />

Groningen, Netherlands; Leiden University Medical Center, Leiden,<br />

Netherlands; Erasmus Medical Center, Netherlands<br />

Background<br />

To reduce mortality risk, women with a family history of breast<br />

cancer are often screened with mammography before 50 years of<br />

age. Additional Magnetic Resonance Imaging (MRI) can improve<br />

sensitivity. MRI screening is cost-effective for BRCA1/2 mutation<br />

carriers. However, for women with a family history of breast cancer<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

292s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 3<br />

without a proven mutation cost-effectiveness is not clear. We<br />

evaluated the cost-effectiveness of additional MRI for women with<br />

a familial risk in the largest prospective MRI screening study: the<br />

Dutch MRI Screening Study (MRISC).<br />

Materials & Methods<br />

Between 1999 and 2007 a total of 1597 women (8370 women years at<br />

risk) between 25-70 years, with an estimated cumulative lifetime risk<br />

of 15-50% for breast cancer participated in the MRISC. Women were<br />

screened with clinical breast examination (CBE) every six months and<br />

annual mammography and MRI. We calculated the costs per detected<br />

breast cancer. In addition, MRISC data were incorporated into a<br />

micro simulation screening analysis model: MISCAN. This model<br />

simulates screening programs with different screening modalities and<br />

time intervals. Different screening schemes were evaluated and the<br />

cost per life-year gained (CLYG) estimated.<br />

Results<br />

Forty-seven breast cancers, including 9 Ductal Carcinoma in Situ,<br />

were detected. Screening with additional MRI leads to a cost per<br />

detected breast cancer treated of €101,962. In increasing age-cohorts<br />

the cost decreased, probably due to the higher breast cancer incidence.<br />

The cost per detected and treated breast cancer in age group 40-50<br />

years doubled in the age group >60 years. We will demonstrate these<br />

results more extensively.<br />

With MISCAN modeling we predicted that screening with this<br />

scheme from age 35 to 60 years reduces breast cancer mortality by<br />

30% at a CLYG of €119,945 (3.5% discounting), compared to 21%<br />

estimated mortality reduction at €45,707 CLYG with mammography<br />

and CBE alone.<br />

Conclusion<br />

Screening with MRI may improve survival for women with familial<br />

risk for breast cancer, but is expensive. However, it may be costeffective<br />

for a select group. We will discuss subgroups that may<br />

benefit from MRI screening and in which age category MRI was<br />

most effective in our study.<br />

P3-02-10<br />

Implementation and uptake of a provincial, population-based,<br />

organized breast screening program for high risk women in<br />

Ontario: The Ontario breast screening program (OBSP) high<br />

risk program.<br />

Eisen A, Carroll J, Chiarelli AM, Horgan M, Meschino W, Rabeneck<br />

L, Shumak R, Warner E. Sunnybrook Health Sciences Centre, Toronto,<br />

ON, Canada; University of Toronto, ON, Canada; Mount Sinai<br />

Hospital, Toronto, ON, Canada; <strong>Cancer</strong> Care Ontario, Toronto,<br />

ON, Canada; North York General Hospital, Toronto, ON, Canada<br />

Background: Genetic testing for mutations in BRCA1/2 has been<br />

clinically available in Ontario since 2000. Over 15000 individuals<br />

have been tested. Evidence from clinical trials has consistently shown<br />

that women at high risk of breast cancer (BrCa) benefit from BrCa<br />

screening that includes both magnetic resonance imaging (MRI) of<br />

the breast and mammography, yet access to MRI in Ontario was<br />

variable. In 2011, <strong>Cancer</strong> Care Ontario (CCO) established an expert<br />

panel to develop a protocol for expanding the OBSP, initially created<br />

for average risk women 50-74, to include MRI and mammography<br />

for eligible high risk women. Methods: The panel’s tasks included:<br />

1. determining high risk criteria 2. estimating the prevalence of<br />

high risk women 3. selecting a cancer risk model 4. developing a<br />

referral and assessment pathway for potentially eligible subjects 5.<br />

developing educational resources, training plan and communication<br />

strategy for relevant stakeholders 6. developing indicators for program<br />

evaluation, 7. providing guidance for post-implementation issues.<br />

Results: The program was initiated July, 2011 and now includes 28<br />

sites. Women aged 30-69 with and without a history of breast cancer<br />

are eligible if they 1. are BRCA1/2 mutation carriers or untested first<br />

degree relatives of carriers, 2. have a lifetime risk of breast cancer<br />

>=25% based on family history according to IBIS or BOADICEA<br />

risk models, or 3. received prior radiation therapy to the chest. It is<br />

estimated that 34000 high risk women in the target age group live<br />

in Ontario. Preliminary volume data for the first 9 mos are shown in<br />

Table 1. Of the 5037 women participating, 802 have been referred<br />

directly by their physician and 4,235 have been referred for genetic<br />

assessment. Of the 2,946 women who received genetic assessment,<br />

31% met the high risk criteria. To date, 729 high risk MRI scans have<br />

been performed. Conclusions: A population based organized screening<br />

program for high risk women that includes genetic risk assessment has<br />

been implemented in Ontario. Further evaluation of risk assessment<br />

and screen results are underway. To our knowledge, this is the first<br />

organized screening program for women at high risk of breast cancer.<br />

Table 1<br />

Category A* Category B ** Total<br />

# referrals to OBSP High Risk Program 802 4235 5037<br />

# women with family hx who received genetic<br />

assessment<br />

N/A N/A<br />

Counseling only 1769<br />

Counseling and BRCA1/2 testing 1177<br />

Total 2946<br />

Proportion of women with family hx determined<br />

N/A<br />

to be high risk by criteria<br />

31% N/A<br />

# women eligible for high risk screening 802 925 1727<br />

# women ineligible for high risk screening 0 2021 2021<br />

# high risk screens<br />

Mammo 649<br />

MRI 729<br />

Ultrasound (if MRI contraindicated) 6<br />

*category A: known BRCA1/2 carrier; 50% risk of being a carrier; prior XRT; known 25% lifetime<br />

breast cancer risk **category B: referred to OBSP program due to family hx without prior formal<br />

risk assessment<br />

P3-02-11<br />

Screening Magnetic Resonance Imaging (MRI) of the breast in<br />

women at increased lifetime risk for breast cancer: A retrospective<br />

single institution study.<br />

Ehsani S, Strigel R, Pettke E, Wilke L, Szalkucki L, Tevaarwerk<br />

AJ, Wisinski KB. University of Wisconsin Carbone <strong>Cancer</strong> Center,<br />

Madison, WI<br />

Background: Multiple factors are associated with an increased lifetime<br />

risk of breast cancer, including inheritance of an abnormal BRCA<br />

1/2 gene, history of lobular carcinoma in situ (LCIS) or atypical<br />

hyperplasia, family history of breast cancer or previous chest wall<br />

radiation. In 2007, the American <strong>Cancer</strong> Society released updated<br />

guidelines for breast cancer screening based on risk stratification.<br />

These guidelines added annual MRI screening to mammography<br />

for women with greater than or equal to a 20-25% lifetime risk.<br />

<strong>Breast</strong> MRI screening trials have consistently demonstrated a higher<br />

sensitivity of MRI for malignancy compared with mammography,<br />

with an additional cancer yield from MRI of approximately 3%.<br />

The purpose of this study was to evaluate MRI screening outcomes<br />

in women with an increased risk for breast cancer evaluated in<br />

an established breast subspecialty clinic within the University of<br />

Wisconsin (UW) Hospital and Clinics.<br />

Methods: Patients (Pts) were included if they were seen by a UW<br />

breast center nurse practitioner, medical or surgical oncologist<br />

between 1/1/2007 – 3/1/2011 with a diagnosis code of: family history<br />

of breast or ovarian cancer, genetic susceptibility to malignant<br />

neoplasm or genetic carrier, Hodgkin’s disease, LCIS, or atypical<br />

hyperplasia. Pts with a co-existing diagnosis of invasive breast cancer<br />

or ductal carcinoma in situ prior to initial encounter were excluded.<br />

Demographic information, breast cancer risk factors, estimated<br />

lifetime risk of breast cancer and screening recommendations<br />

www.aacrjournals.org 293s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

were abstracted from the medical record. Results of subsequent<br />

breast imaging examinations (including breast MRI, diagnostic and<br />

screening mammography, and image-guided biopsies) were analyzed<br />

with the use of the mammography information system (PenRad).<br />

Results: Of 276 women who met the inclusion criteria, 148 underwent<br />

at least 1 screening breast MRI. The majority of MRI screened pts<br />

were premenopausal (82%) and Caucasian (96.6%) with a mean age<br />

of 42.5 (range 20-68) at their initial encounter. Eighty five percent had<br />

a first degree relative with breast cancer and 72.3% of pts undergoing<br />

MRI screening had a documented lifetime risk of breast cancer of<br />

20% or greater using a validated model. Within this MRI-screened<br />

cohort, 18.2% had a known genetic predisposition to breast cancer.<br />

Over the time assessed, 307 MRIs were performed in the 148 pts.<br />

Biopsy was recommended and performed based on the results of<br />

the MRI in 31 of 307 exams (10%). Ten cancers were detected for<br />

a positive predictive value based on biopsy performed of 32% and<br />

an overall cancer yield of 3.3% (10 of 307 MRI exams). All cancers<br />

were stage 0 - II. All pts are currently with no evidence of disease.<br />

Conclusion: <strong>Breast</strong> MRI has a high positive predictive value and<br />

cancer yield with an acceptable biopsy rate in a diverse group of high<br />

risk women undergoing breast MRI at an academic center outside<br />

of a clinical trial.<br />

P3-02-12<br />

The Cost of Mammography Screening in the United States by<br />

Screening Policy<br />

Thorsen CM, Eklund M, Ozanne EM, Esserman LJ. University of<br />

California, <strong>San</strong> Francisco, CA; Karolinska Institute, Stockholm,<br />

Sweden<br />

Background:<br />

Multiple mammography screening strategies exist; however, the<br />

resources associated with screening strategies are not well known<br />

and are rarely discussed. Many organizations recommend annual<br />

mammography for women over the age of 40, while the United States<br />

Preventative Services Task Force (USPSTF) and most countries<br />

outside the US recommend biennial screening, focusing screening<br />

efforts on women over 50 years. Additionally, some suggest risk-based<br />

approaches to improve screening. This analysis seeks to estimate<br />

the aggregate cost of mammography screening in the US in 2010<br />

and to then further compare the costs of these proposed strategies,<br />

specifically comparing annual versus biennial screening.<br />

Methods:<br />

We simulated the annual cost of mammography in the US under the<br />

following screening strategies: 1) estimate of screening that occurred<br />

in 2010 under current policy, 2) annual screening of women older<br />

than 40 years, 3) biennial screening of women 50 to 75 years, and<br />

4) biennial screening of women 50 to 75 years and high risk women<br />

40-50 years following USPSTF guidelines. The simulated 2010<br />

screening policy was based on current screening rates ranging from<br />

51% of forty-year-old women to 66% of sixty-five-year-old women.<br />

Screening strategies 2-4 simulated screening 85% of women overall,<br />

and for the USPSTF policy 20% of women 40-50 years were estimated<br />

to be high risk. The simulation was carried out using R statistical<br />

software.<br />

Results:<br />

Aggregate costs of the four screening strategies were estimated. The<br />

total cost of mammography screening in the US in 2010 was estimated<br />

to be $7.95 billion. The projected cost under an annual screening<br />

policy beginning at 40 years was $10.71 billion. The projected cost<br />

of biennial screening from 50-75 years was $5.25 billion compared to<br />

$5.33 billion following the USPSTF guidelines. The biennial USPSTF<br />

policy cost $5.38 billion less than annual screening. The largest<br />

drivers of cost were the percent of women screened, the cost of a<br />

mammogram, and the percent of women recalled after mammography.<br />

Conclusions:<br />

The costs of mammography vary greatly by strategy and are<br />

relevant to screening policy. The basic biennial strategy was the<br />

least expensive; however for a slight increase in cost, the USPSTF<br />

2009 guideline strategy screened more age appropriate women for<br />

$2.62 billion less than current policy. For less cost, the modeled<br />

USPSTF guidelines screened 85% of women compared to current<br />

policies that screen less than 65% of women. Biennial screening<br />

provides screening to more women, lowers overall false-positive<br />

rates, and improves capacity for access to high-quality equipment<br />

and well-trained mammographers. As data to support evidenced<br />

based strategies increase, resources can be better allocated toward<br />

screening a broader, larger population of women that are most likely<br />

to benefit from screening. With no evidence for adverse outcomes,<br />

a biennial screening policy enables the population of US women to<br />

benefit from mammography for less cost.<br />

P3-02-13<br />

ANALYSIS OF INFRASTRUCTURE FOR MAMMOGRAPHY<br />

SCREENING IN THE STATE OF GOIAS, BRAZIL, 2010<br />

Freitas-Junior R, Correa RS, Peixoto J-E, Rodrigues DCN, Rahal<br />

RMS. Federal University of Goias, Goiania, Goias, Brazil; Comissão<br />

Nacional de Energia Nuclear, Goiania, Goias, Brazil; National<br />

<strong>Cancer</strong> Institute of Brazil, Rio de Janeiro, RJ, Brazil<br />

Objective: Assess the infrastructure for mammography screening<br />

in the state of Goiás, Brazil, taking into consideration the quality<br />

of the image generated by the equipment and the dose received by<br />

the patients, according to the type of technology (conventional and<br />

digital mammography). Methods: Cross-sectional study in which the<br />

observational units were the diagnostic imaging services that were<br />

in operation in 2010. In an in loco visit, we collected information on<br />

the equipment and performed tests to assess quality of the image,<br />

accuracy values for tube tension, quality of X-ray beam, and dose at<br />

the simulator entrance. The percentage of mammography equipments<br />

in conformity with the parameters evaluated was calculated, and the<br />

mean glandular tissue dose (Dg) was estimated based on the dose at<br />

the simulator entrance. To assess quality of the image and the radiation<br />

dose, we used a standard breast simulator (5.3 cm thickness and 50%<br />

glandular tissue). Results: In 2010, 102 services were in operation,<br />

using 106 equipments. Among these, 100 (94.3%) – 64 conventional<br />

and 36 digital mammography equipments – had their performance<br />

and dose assessed. The visualization of fibers (p = 0.044) and the<br />

dose at the simulator entrance (p = 0.042) were the parameters that<br />

presented differences between the types of technology.<br />

Frequency of mammography equipments in conformity with the parameters used to assess the<br />

quality of the image and the equipment performance according to the type of technology, in the<br />

state of Goiás, Brazil, in 2010.<br />

Variable Conventional Digital p<br />

(n = 64) (n = 36)<br />

Quality of the image 57 89.1 31 86.1 0.752<br />

Spatial resolution 63 98.4 32 88.9 0.055<br />

Microcalcifications 55 85.9 32 88.9 0.765<br />

Fibers 64 100 33 91.7 0.044<br />

Masses 64 100 35 97.2 0.36<br />

Low Contrast disks 46 71.9 29 80.6 0.471<br />

Dose at the simulator entrance 50 78.1 21 58.3 0.042<br />

Half-value layer (HVL) 62 96.9 36 100 0.535<br />

Tension precision 46 71.9 27 75 0.817<br />

The mean dose at the simulator entrance was 9.75 mGy (± 2.46)<br />

in conventional and 14.93 mGy (± 8.39) in digital equipments (p <<br />

0.001); 12% of the conventional and 50% of the digital equipments<br />

presented Dg above the limit established by the European guidelines.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

294s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 3<br />

Conclusion: The infrastructure for mammography screening in the<br />

state of Goias, Brazil, in 2010 presented acceptable quality standard<br />

according to international guidelines. However, the radiation doses<br />

were heterogeneous between the two types of technology, with a<br />

predominance of higher doses in digital equipments.<br />

P3-02-14<br />

INTEGRATED PROGRAM FOR BREAST CANCER<br />

CONTROL: PARTIAL PRESENTATION OF THE RESULTS<br />

OBTAINED INTHE FIRST 24MONTHS OF OPERATION<br />

Blanco EC, Borges MM, Pinotti M, Gennari MB, Ribeiro IM,<br />

Nascimento CCP, <strong>San</strong>tos LFG, Sahium RC. Oswaldo Cruz Alemão<br />

Hospital, São Paulo, Brazil<br />

Introduction <strong>Breast</strong> cancer is a public health problem in Brazil. This<br />

is due to the difficulty of primary prevention (eliminating risk factors<br />

or to diagnose and treat precursor lesions), with a consequent increase<br />

in the incidence and mortality from this cancer. The current challenge<br />

arises from the lack of access to a few specialized centers, which in<br />

turn ar e not always qualified for diagnosis and prompt treatment.<br />

Objective To develop a strategic plan aimed at two management<br />

priorities: resolubility diagnostic and fast treatment of women with<br />

clinical or secondary changes to mammographic screening.<br />

Methodology The Oswaldo Cruz Alemão Hospital (OCAH) in<br />

partnership with the Ministry of Health of Brazil has developed and<br />

deployed in the region of the Mooca - City of St. Paul the Integrated<br />

Program for <strong>Breast</strong> <strong>Cancer</strong> Control (IPBCC), which begins in the<br />

primary prevention (health education and removal of factors risk),<br />

continues with the mammographic detection and early diagnosis,<br />

modern instrumental in a Center for Diagnostic Detection and<br />

Tracking (CDDT), and culminates in the surgical and radiotherapy,<br />

ad juvant chemotherapy and hormone therapy or not, according to<br />

a standardized protocol. The care is coordinated in single query by<br />

mastologists trained in resolving service, with the participation of<br />

radiologists, oncologists, social workers, psychologists, nutritionists<br />

and health educators.<br />

Results From January 2010 to December 2011 were met 12266<br />

women, mean age 58 years (range, 22-89) of which 100 were<br />

diagnosed with breast cancer and the proportion of carcinomas was of<br />

8 per 1000. The detection rate mammography, ultrasound, and by both<br />

methods were: 0.30, 0.30 and 0.25, respectively. Forty-one percent of<br />

cancers corresponded to stages Tis N0M0 (3%) and T1N0M0 (38%)<br />

predominantly in women aged less than 50 years (38% of the sample).<br />

The average time between diagnosis and recorded start of surger y or<br />

chemotherapy was 42 days (range,19 - 68 ). As for quality of care,<br />

satisfaction was 99% (n = 12143).<br />

Conclusions The experience in resolving service provided a<br />

significant increase in the number of initial cases showing detection<br />

ratesper image consistent with the age distribution of population. It<br />

is expected that the tracking time and quality of service, allow the<br />

prevalent cancers cease to be diagnosed, privileging the incidents.<br />

The median time to diagnosis and early treatment of patients with<br />

cancer was significantly lower the average of 120 days recorded in<br />

some public reference services.<br />

P3-03-01<br />

3D mapping of total choline in human breast cancer using highspeed<br />

MR spectroscopic imaging at 3T: initial experience during<br />

neoadjuvant therapy<br />

Posse S, Zhang T, Royce M, Dayao Z, Lopez S, Sillerud L, Casey L,<br />

Eberhardt S, Lomo L, Rajput A, Russell J, Lee S-j, Bolan P. University<br />

of New Mexico School of Medicine and UNM <strong>Cancer</strong> Center,<br />

Albuquerque, NM; New Mexico <strong>Cancer</strong> Center, Albuquerque, NM;<br />

University of Minnesota, Minneapolis, MN<br />

OBJECTIVE: To assess the feasibility of quantitative high-speed MR<br />

spectroscopic imaging (MRSI) of total Choline (tCho) as an adjunct<br />

to dynamic-contrast enhanced MRI to improve lesion characterization<br />

and monitor treatment response in patients undergoing neoadjuvant<br />

chemotherapy (NAC).<br />

METHODS: Twelve patients with infiltrating ductal carcinoma<br />

(Table 1) were studied using a clinical 3T MR scanner (Siemens,<br />

Erlangen, Germany) equipped with 8- and 16-channel breast array<br />

(Hologic Inc., Bedford, MA). Four patients were studied before<br />

NAC. Four patients were studied once during NAC. Two patients<br />

were studied before and within 2-7 days of treatment initiation. One<br />

of these patients participated in an additional scan after 5 months of<br />

treatment. Two additional patients were studied at 3 time points during<br />

NAC. Measurements were performed using PRESS prelocalized<br />

3D Proton-Echo-Planar-Spectroscopic-Imaging (PEPSI) using TR/<br />

TE=2000ms/135ms, matrix size up to 32×16x8, voxel size=1cc, and<br />

total acquisition time of 10 minutes (including water reference scan).<br />

Additional data were collected at TE 60 ms to enhance sensitivity<br />

for detecting tCho and J-coupled resonances. TE-averaging (8 steps,<br />

DTE: 2.5 ms) was employed to minimize gradient sideband artifacts.<br />

Quantification of tCho in reference to tissue water was performed<br />

using spectral fitting and relaxation correction.<br />

RESULTS: Strongly elevated tCho with maximum concentration<br />

up to 5.3 mmol/kg was measured in 9 patients with enhancing<br />

lesions larger than 2 cc volume (Table 2). Decreases in tCho were<br />

measured in all four patients who were followed during neoadjuvant<br />

chemotherapy. Decreases in tCho were measurable during the first<br />

week of neoadjuvant treatment in responders, consistent with previous<br />

studies. Our preliminary data also indicate that the combination of<br />

concentration and spatial extent of detectable tCho may be the most<br />

sensitive marker of treatment response.<br />

Patient/tumor characteristics<br />

Patient<br />

Age<br />

Tumor<br />

grade<br />

Tumor stage<br />

ER & PR<br />

status<br />

HER2<br />

status<br />

Lesion size (cm)<br />

1 57 2 T4bN3cM0 ER- PR- POS 4.3x7.5x8.1 6 m<br />

2 50 2 T4dN3M0 ER+ PR+ POS 5 1 y<br />

3 55 3 T2N0M0 ER- PR- NEG 3.4 x 3.5<br />

4 45 1 T2N0M0 ER+ PR+ NEG 2.5 x 2<br />

5 28 3 T3N1M0 ER- PR+ POS 6.5 4 d<br />

6 54 3 T3N1M0 ER+ PR- NEG 7 X 5.9 21 d<br />

7 60 2 T1cN0M0 ER+PR + POS 1.2<br />

8 77 1 T4N2M0 ER+ PR+ POS 16 x 17 2 m<br />

9 50 3 T2N0M0 ER+ PR- POS 3.2 x 3.6 7 d<br />

10 60 3 T3N1M0 ER+ PR+ NEG 6 x 4.5 2 d<br />

11 52 3 T3N1M0 ER- PR- NEG 6.5 x 6.2 7 d<br />

12 35 3 T2N1M0 ER- PR- NEG 2.2<br />

Treatment<br />

duration at<br />

scan 1<br />

tChol by treatment duration<br />

Patient Before 1-7 days 8-21 days 3-12 weeks 3-6 months > 6 months<br />

1 0.7/0.7/1<br />

2 ND<br />

3 1.8/1.8/1<br />

4 0.5/0.5/1<br />

5 0.8/2.4/23 1.5/3.9/21 0.8/1.2/2<br />

6 1.0/2.4/7 0.9/1.6/4 ND<br />

7 ND (Clip)<br />

8 TF<br />

9 0.6/0.7/2 0.3/0.3/2 ND<br />

10 TF 0.9/1.6/15<br />

11 1.9/4.0/87 1.9/4.2/27<br />

12 1.7/5.3/47<br />

ND - Not detected; TF - technical failure<br />

www.aacrjournals.org 295s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

CONCLUSION: This study demonstrates feasibility of quantitatively<br />

mapping tCho in invasive breast carcinoma using high-speed MRSI.<br />

The long-term goals are to utilize high-speed MRSI as an early<br />

predictor of treatment failure in women undergoing neoadjuvant<br />

therapy (i.e. chemotherapy, endocrine therapy or biologic therapy)<br />

for breast cancer and to develop an improved screening protocol for<br />

high-risk patients.<br />

P3-03-02<br />

The metastatic rate of IMLN, when IMLN metastasis is suspected<br />

with PET CT.<br />

Lee J-H, Son G-T, Choi J-E, Kang S-H, Lee S-J. Yeungnam University<br />

College of Medicine, Daegu, Republic of Korea<br />

Background<br />

Metastatic status of internal mammary lymph nodes (IMLNs) has<br />

a clinical importance in assessing stage and prognosis of breast<br />

cancer. But, when metastasis of IMLN is suspected, the management<br />

is controversial. We reviewed 29 breast cancer patients who had<br />

IMLN dissection, retrospectively, and investigated the pathologic<br />

status of IMLNs.<br />

Methods<br />

From August 2005 to December 2011, at Yeungnam University<br />

Hospital, 43 patients underwent IMLN biopsy or dissection for<br />

suspected IMLN metastasis on lymphoscintigraphy (7 patients), breast<br />

ultrasound (1 patient) or PET CT (29 patients), when diagnosed with<br />

primary or recurred breast cancer. 6 patients who had stage IV at<br />

diagnosis or had too obscure data to identify exact location of IMLN,<br />

were excluded. Among them, we reviewed clinicopathologic features<br />

of IMLN detected on PET CT and metastatic status of IMLNs was<br />

investigated.<br />

Results<br />

Total 29 patients were included in this study. 19 patients and 10<br />

patients underwent IMLN dissection when diagnosed with primary<br />

or recurred breast cancer, respectively. Unlike conventional IMLN<br />

dissections, our IMLN biopsy or dissection was done during Radical<br />

mastectomy (in 2 pts.), modified radical mastectomy (in 14 pts.),<br />

using incision of breast conserving surgery (in 3 pts.) and separated<br />

incision ( in 10 pts.), with or without resection of ribs.<br />

The mean number of IMLNs was 2.76±2.15 and total metastatic rate<br />

of IMLN was 72.4% (21/29). The sensitivity, specificity, positive<br />

predictive value, and negative predictive value of PET CT was<br />

following: 91.3%, 42.8%, 72.4% and 27.6%. Mean standard uptake<br />

value (SUV) of metastatic and non-metastatic IMLN were 3.6±2.9<br />

and 3.9±2.6 and there was no statistical difference (p-value=0.821).<br />

In PET CT, non-metastatic vs metastatic IMLN<br />

Tumor location<br />

Tumor Size<br />

(cm)<br />

Number of<br />

metastatic<br />

ALN (n)<br />

Number of<br />

IMN (n)<br />

outer central/inner<br />

Non-metastatic IMLN<br />

4 (50.0%)<br />

(8 patients)<br />

4 (50.0%) 3.81±2.08 3.50±4.98 1.75±0.70 3.91±2.62<br />

Metastatic IMLN(21<br />

patients)<br />

10 (47.6%) 11 (52.4%) 3.54±2.01 8.28±7.74 3.14±2.39 3.68±2.90<br />

P-value 1.000 0.967 0.034 0.060 0.821<br />

During IMLN dissection, besides initial approach intercostals space<br />

(ICS), some metastatic IMLN was also found in upper or lower level<br />

ICS (42.9%, 6/14). Only IMLN metastasis without axillary nodes<br />

metastasis were found in 3 patients and the tumor location of these<br />

patients was all inner or central quadrant. Chest X-ray was done<br />

postoperatively as routine procedure, and there were no other specific<br />

complications such as pneumothorax or hemothorax.<br />

Conclusion<br />

IMLN dissection without radical mastectomy can be done safely<br />

without complications due to recent advance in diagnostic and surgical<br />

skills. If SUV on IMLN is shown on the PET-CT, IMLN dissection<br />

SUV<br />

is needed, regardless of SUV. If breast cancer is located at inner<br />

quadrant, more aggressive dissection of IMLN is needed. Further<br />

follow-up and studies are needed to assess locoregional recurrence and<br />

to compare improvement in overall survival and disease free survival.<br />

P3-03-03<br />

A tri-modality imaging assessment algorithm to evaluate<br />

neoadjuvant therapy response in patients with operable breast<br />

cancer<br />

Umphrey H, Bernreuter W, Bland K, Carpenter J, Falkson C, Forero<br />

A, Keene K, Krontiras H, Meredith R, Urist M, De Los <strong>San</strong>tos J.<br />

University of Alabama at Birmingham, AL<br />

Background: To determine the negative predictive value (NPV),<br />

positive predictive value (PPV), accuracy, sensitivity and specificity of<br />

a pre-surgical tri-modality imaging assessment algorithm to determine<br />

complete pathologic response (pCR) post neoadjuvant therapy in<br />

patients with operable breast cancer.<br />

Methods: A retrospective analysis was performed on data collected<br />

from patients receiving neoadjuvant therapy and pre-surgical<br />

breast magnetic resonance imaging (MRI), ultrasound (US) and<br />

mammography between 2004 and 2010 at our institution. Tri-modality<br />

imaging was reviewed by a single blinded breast radiologist and<br />

evaluated for predetermined modality specific parameters as defined<br />

in Table 1. The NPV, PPV, accuracy, sensitivity, and specificity were<br />

calculated on the basis of the final surgical pathology report with a<br />

complete pathologic response in the breast defined as no residual<br />

invasive disease or in situ disease.<br />

Results. Eighty-three tumors in 83 patients with a mean age of 50<br />

(range 27-70) were evaluated. Twenty-three patients had a pCR.<br />

The NPV, PPV, sensitivity, specificity, and accuracy of tri-modality<br />

imaging algorithm for pCR were 0.87, 0.95, 0.95, 0.87 and 0.93<br />

utilizing a cut-point of ≤ 5 for complete response by imaging. The<br />

mean score for patients with pCR was 4.61 (range 3-10) with 3 patients<br />

scoring above 5. The mean score for patients with residual disease<br />

was 7.73 (range 5-11).<br />

Conclusions: A tri-modality imaging scoring algorithm is predictive<br />

of complete pathologic response. This algorithm will be tested in a<br />

developing prospective trial that will also assess the additive value<br />

of tumor bed biopsy in patients who achieve a score of 5 or less.<br />

Table 1<br />

Imaging Modality<br />

Imaging Parameter Score<br />

0 1 2 3<br />

MR<br />

Early peak signal intensity ≤80 >80<br />

Internal enhancement pattern Homogeneous Heterogeneous<br />

Kinetic pattern Type I Type II Type III<br />

US<br />

Mass size Resolution of mass ≤20 mm >20 mm<br />

Mammography<br />

Residual mass Absent Present<br />

Malignant calcifications Absent Present<br />

P3-04-01<br />

Protein pathway activation mapping of the I-SPY 1 biopsy<br />

specimens identifies new network focused drug targets for patients<br />

with triple negative tumors<br />

Wulfkuhle JD, Wolf D, Gallagher RI, Yau C, Calvert V, Espina V,<br />

Illi J, Wu Q, Boe M, Yan Y, I-SPY TRIAL-1 Investigators, Liotta LA,<br />

van’t Veer L, Esserman L, Petricoin EF. George Mason University,<br />

Manassas, VA; University of California, <strong>San</strong> Francisco, CA;<br />

Genentech, South <strong>San</strong> Francisco, CA<br />

Background: Identification of new therapies and predictive<br />

biomarkers is needed for patients with triple negative (TN) breast<br />

cancer. Elucidating pathway activation in cancer is critical as these<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

296s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 3<br />

proteins represent targets for many of the new molecular therapeutics<br />

and companion diagnostic markers. The I-SPY TRIAL (CALGB<br />

150007/150012, ACRIN 6657) is a trial of neoadjuvant anthracyclineand<br />

taxane-based chemotherapy that provides longitudinal biopsy<br />

specimens and molecular and clinical/pathological characterization.<br />

Methods: Tumor epithelium was procured by Laser Capture<br />

Microdissection from 149 pretreatment frozen biopsy specimens<br />

(T1) and 102 frozen biopsy specimens collected 1-4 days after first<br />

chemotherapy treatment (T2). Reverse Phase Protein Microarray was<br />

used to measure the activation of 39 and 100 signaling proteins in<br />

the T1 and T2 biopsies, respectively, including drug targets for Phase<br />

I-III and FDA cleared therapeutics. Associations between pathway<br />

activation and response to chemotherapy (RCB 0/1 vs. 2/3) and<br />

relapse-free survival (RFS) were evaluated for the triple-negative<br />

(TN) patient population at each time point and for changes between<br />

T1 and T2. Significance thresholds for response and RFS were as<br />

follows: Wilcoxon rank sum test p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

positive lymph nodes). International recommendations for patients<br />

with 1-3 positive lymph nodes suggest that PMRT should be restricted<br />

to younger patients and patients with other poor prognostic features.<br />

However, large randomized trials, including the DBCG82 trials<br />

(Danish <strong>Breast</strong> <strong>Cancer</strong> Cooperative Group), have previously shown<br />

a substantial overall survival benefit after PMRT in patients with low<br />

risk of LR, and shown that the largest translation of LR reduction into<br />

breast cancer mortality reduction occurs within the most favorable<br />

prognosis group. Our hypothesis is that a more refined partitioning<br />

of patients likely to benefit from PMRT can be established through<br />

identification of genes whose transcription interacts with PMRT to<br />

modify the hazard of LR.<br />

Material and methods: The DBCG82bc cohort constitutes high risk<br />

patients (tumor size > 5 cm and/or positive nodes and/or invasion<br />

in skin or pectoral fascia) diagnosed between 1983-89, treated<br />

with mastectomy and partial axillary lymph nodes dissection and<br />

randomized to +/- PMRT. From 267 DBCG82bc patients, frozen<br />

tumor samples were available. Whole genome arrays (Applied<br />

Biosystem Human Genome Survey Microarray v2.0®, Applied<br />

Biosystem, Foster City, USA) were successful in 195 samples. Genes,<br />

whose expression levels interacted with PMRT on the association<br />

with LR, were identified through a two step Cox Proportional Hazard<br />

model with lasso penalty. 11 node negative patients were excluded<br />

from the subsequent analysis of 184 node positive patients (1-3 pos.<br />

nodes: 102 pts, ≥ 4 pos. nodes: 82 pts).<br />

Results: Seven genes were identified whose expression interact with<br />

the effect of PMRT, and a specialized index was generated based on<br />

the expression levels of these genes. Patients were ranked according<br />

to the size of the index, and divided into quartiles with 25% of the<br />

patients designated as having a “high index” and 75% a “low index”.<br />

Among patients not receiving PMRT, a low index was in both nodal<br />

groups (1-3 vs. ≥ 4 positive nodes) associated with a significantly<br />

higher risk of LR compared to patients with a high index. In both<br />

nodal groups, PMRT significantly reduced the risk of LR in patients<br />

with a low index; equalizing the risk to patients with a high index,<br />

who showed no additional LR reduction by PMRT. In the group of<br />

1-3 positive nodes, PMRT raised the local control rate (LCR) after<br />

10 years from 47.5 % to 91.8% in the low index group (p= 0.0001),<br />

whereas the change in the high index group was non-significant<br />

(92.3% vs. 100.0%). In the group of patients with ≥ 4 positive nodes,<br />

PMRT raised LCR from 16.6% to 80.0% in the low index group (p=<br />

0.0001), and no effect on LCR was seen in the high index group<br />

(83.3% vs. 87.5%, n.s.).<br />

Conclusion: A seven gene-profile attaining prognostic and predictive<br />

impact, irrespective of number of positive nodes, was identified. The<br />

profile allowed the identification of 25% of the patients not showing<br />

any additional benefit from PMRT in terms of LR. The gene-profile<br />

may provide a method to identify patients expected to benefit from<br />

PMRT.<br />

P3-04-04<br />

Identification of a “BRCAness” signature in triple negative breast<br />

cancer by Comparative Genomic Hybridization<br />

Toffoli S, Bar I, Abdulsater F, Delrée P, Hilbert P, Cavallin F, Moreau<br />

F, Clark J, Lacroix-Triki M, Campone M, Martin A-L, Roché H,<br />

Machiels J-P, Carrasco J, Canon J-L. Institute of Pathology and<br />

Genetics, Gosselies, Belgium; MDxHealth, Liège, Belgium; Institut<br />

Claudius Regaud, Toulouse, France; Institut de Cancérologie<br />

de l’Ouest-René Gauducheau, Saint-Herblain - Nantes, France;<br />

UNICANCER, Paris, France; Cliniques Universitaires Saint-Luc,<br />

Brussels, Belgium; Grand Hôpital de Charleroi (GHdC), Charleroi,<br />

Belgium<br />

Background:<br />

Triple Negative <strong>Breast</strong> <strong>Cancer</strong>s (TNBC) represent 12% to 20% of total<br />

<strong>Breast</strong> <strong>Cancer</strong>s (BC) and have a worse outcome compared with other<br />

breast cancer subtypes. TNBC often show a deficiency in DNA double<br />

strand break repair mechanisms. This deficiency is generally related<br />

to inactivation of a repair enzymatic complex involving BRCA1<br />

caused either by genetic mutations, epigenetic modifications, or by<br />

post-transcriptional regulations. The identification of BC presenting<br />

a BRCA1 deficiency could be useful to select patients that could<br />

benefit from PARP inhibitors, alkylant agents or platinum-based<br />

chemotherapy.<br />

In this study, we have identified by Comparative Genomic<br />

Hybridization array (CGH-array) a recurrent gain in 17q25.3<br />

characteristic of BRCA1 mutated or methylated TNBC.<br />

Methods:<br />

130 formalin-fixed paraffin embedded (FFPE) tumours including<br />

TNBC (unknown-BRCA1-status, BRCA1 mutated and nonmutated),<br />

Luminal A, Luminal B, Her2-neu amplified (Her2+) BC<br />

and BRCA1-mutated non-TNBC (mutated-Luminal A, Luminal B,<br />

Her2+) were obtained from our local tumour collection. DNA was<br />

extracted and genomic copy-number alterations were analysed by<br />

CGH-array (Agilent, SuperPrint G3 Human CGH 8x60K Oligo<br />

Microarrays). FISH analyses were performed on section from FFPE<br />

samples to validate some chromosomal aberrations belonging to the<br />

17q25.3 amplified region evidenced by CGH-array. The study of<br />

BRCA1 promoter methylation status in all tumours was carried out<br />

by MDxHealth (Liege,Belgium).<br />

Results:<br />

In this study, we have identified by CGH-array a genomic region<br />

(17q25.3) amplified in 90% of the BRCA1 mutated tumours (29/32).<br />

This chromosomal gain was studied in other subtypes of BC by<br />

CGH-array and it was only evidenced in 30% (6/20) of BRCA1<br />

non-mutated TNBC, 26.67% (4/15) of unknown-BRCA1-status<br />

TNBC, 13.64% (3/22) of Luminal B, 19.05% (4/21) of Her2+ and<br />

0% (0/20) of Luminal A breast cancers. FISH assays confirmed these<br />

chromosomal amplifications and evidenced like CGH array analyses<br />

a significant difference between BRCA1 mutated and non-mutated<br />

BC for the 17q25.3 gain.<br />

BRCA1 methylation was found only in TNBC (11/58) and was<br />

not found in the BRCA1-mutated BC cohort, nor in the Luninal<br />

A, Luminal B and Her2+ samples. In BRCA1 non-mutated TNBC,<br />

the methylation was found in 8 cases including 4 with 17q25.3<br />

amplification. In the unknown-BRCA1-status TNBC, 3 methylated<br />

samples were found with 1 case with co-amplification of the 17q25.3<br />

region.<br />

Recurrence of 17q25.3 amplification in BRCA1 mutated tumours as<br />

well as its detection in BC having a methylation in BRCA1 promoter<br />

suggests that the 17q25.3 gain could be a marker of the BRCA1<br />

deficiency. Identification of relevant genes whose expression is upregulated<br />

within the recurrently gained region is underway.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

298s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 3<br />

Conclusions:<br />

The CGH signature observed in 17q25.3 chromosomal region and<br />

FISH assay developed in this study could allow the identification of<br />

“BRCAness” breast tumours, improving the diagnostic performance<br />

and orienting the selection of the appropriate therapy. The up-regulated<br />

genes themselves might also represent potential therapeutic targets.<br />

Acknowledgements:<br />

This work was financed through the “Plan National <strong>Cancer</strong>-Action<br />

29” (Belgium) and supported by the UNICANCER-PACS08 trial<br />

(France).<br />

P3-04-05<br />

Kinomic and phospho-proteomic analysis of breast cancer stemlike<br />

cells<br />

Leth-Larsen R, Christensen AG, Ehmsen S, Moeller M, Palmisano<br />

G, Larsen MR, Ditzel HJ. University of Southern Denmark, Odense,<br />

Denmark; Odense University Hospital, Odense, Denmark<br />

<strong>Cancer</strong> stem cells are thought to be responsible for tumorigenic<br />

potential and to possess resistance mechanisms against chemotherapyand<br />

radiation-induced cancer cell death, while the bulk of a tumor<br />

lacks these capacities. The resistance mechanisms may cause these<br />

cells to survive and become the source of later tumor recurrence,<br />

highlighting the need for therapeutic strategies that specifically target<br />

pathways central to these cancer stem cells. The CD44 hi /CD24 -/low<br />

compartment of human breast cancer is enriched in tumor-initiating<br />

cells; however the functional heterogeneity within this subpopulation<br />

remains poorly defined.<br />

From a triple-negative breast cancer cell line we isolated and<br />

cloned CD44 hi single-cells that exhibited functional heterogeneity<br />

according to cancer stem cell features, such as tumor formation<br />

in immunodeficient mice, mammosphere formation, invasion and<br />

migration abilities and sensitivity to chemotherapeutics.<br />

Quantitative proteomic analysis of these clones revealed several<br />

proteins, including kinases, exhibiting altered expression levels,<br />

while other proteins exhibited altered phosphorylation levels. The<br />

quantitative proteomic approach was based on post digestion dimethyl<br />

labeling followed by TiO 2 enrichment of phosphopeptides. Moreover,<br />

analysis of the flowthrough fraction revealed quantitative changes<br />

in the total proteome. Transcriptomic analysis using Affymetrix<br />

Human Gene 1.0 ST Arrays revealed similar alterations at the gene<br />

expression level.<br />

We used these single cell clones to further investigate the molecular<br />

mechanisms underlying the association between selected proteins,<br />

some of them phosphorylated, and cancer stem cell features.<br />

Furthermore, phosphosite-expression was investigated with<br />

NetworKIN that predicts in vivo kinase-substrate relationships.<br />

Our studies reveal pathways that are regulated by protein<br />

phosphorylation and include relevant kinase targets for drug<br />

discovery. Our results support functional heterogeneity in the breast<br />

cancer cell compartment and hold promise for further refinements of<br />

prognostic marker profiling of cells with cancer stem cell-like features.<br />

P3-04-06<br />

Higher gene expression of CSPG4 in the basal-like subtype of<br />

invasive breast cancer and its negative association with lymph<br />

node metastasis<br />

Luo C, Iida J, Chen Y, Dorchak J, Kovatich AJ, Mural RJ, Hu H,<br />

Shriver CD. Windber Research Institute, Windber, PA; Walter Reed<br />

National Military Medical Center, Bethesda, MD; MDR, Global<br />

Systems LLC, Windber, PA<br />

Background: Chondroitin sulfate proteoglycan 4 (CSPG4) was<br />

first identified as a melanoma specific antigen and demonstrated to<br />

stabilize cell-substratum interactions during early events of tumor cell<br />

spreading on endothelial cells and matrix proteins. In invasive breast<br />

cancers (IBCs), CSPG4 has been reported to be highly expressed in<br />

the triple-negative (TN) subtype. Western blotting analyses suggest<br />

that CSPG4 is expressed on TN IBC lines. Here we further explored<br />

the specific gene expression (GE) profiles associated with CSPG4<br />

using the available IBC cases from The <strong>Cancer</strong> Genome Atlas<br />

(TCGA) project towards identification of potential drug targets for<br />

patients with TN IBC.<br />

Methods: The GE data (Agilent, log2 transformed) of 459 IBC tumors<br />

were downloaded from the TCGA data portal. PAM50 classification<br />

results of all samples were obtained from the TCGA breast cancer<br />

AWG group, including 203 Luminal A (LA), 113 Luminal B (LB),<br />

51 HER2+, 84 basal-like (Basal), and 8 Normal-like (not used).<br />

Clinicopathologic data (age, race, T-, N-, and M-stages) were also<br />

obtained from the AWG group. Non-parametric Kruskal-Wallis<br />

test was used for cross-subtype analysis of CSPG4 expression,<br />

followed by Wilcoxon-Mann-Whitney test for pairwise comparison<br />

with Bonferroni adjustment. Chi-square tests were performed for<br />

categorical variation analyses. Differentially Expressed Gene (DEG)<br />

analysis was performed using the Bioconductor “samr” package for<br />

two class unpaired comparison, with p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P3-04-07<br />

Comparison of breast tumors with HER2 amplification and<br />

polysomy 17<br />

Field LA, Deyarmin B, Ellsworth RE, Shriver CD. Windber<br />

Research Institute, Windber, PA; Henry M. Jackson Foundation for<br />

the Advancement of Military Medicine, Windber, PA; Walter Reed<br />

National Military Medical Center, Bethesda, MD<br />

Background: Approximately 20% of invasive breast cancers<br />

overexpress the human epidermal growth factor receptor, Her2, and<br />

are associated with poor prognosis including decreased relapse-free<br />

and overall survival. Overexpression of the Her2 protein is primarily<br />

due to amplification of the HER2 oncogene on chromosome 17,<br />

and these patients are typically treated with targeted therapies such<br />

as trastuzumab or lapatinib. However, increased HER2 copies may<br />

also result from chromosome 17 polysomy in which the entire<br />

chromosome has been duplicated one or more times. The clinical<br />

benefit of treating patients with polysomy 17 in the absence of HER2<br />

amplification with targeted therapy remains unclear. Herein, we<br />

compared the clinical characteristics as well as the gene expression<br />

profiles of breast tumors with increased HER2 copy numbers due to<br />

HER2 amplification or chromosome 17 polysomy.<br />

Methods: Patients for this study were enrolled in the Clinical <strong>Breast</strong><br />

Care Project. HER2 and CEP17 copy numbers were determined<br />

using the PathVysion HER-2 DNA Probe Kit (Abbott Laboratories).<br />

Only those patients with at least 4 HER2 copies per cell were<br />

included in the analysis; those with a HER2/CEP17 ratio 2.2 (N=44)<br />

were considered amplified. Microarray data were generated on HG<br />

U133A 2.0 arrays (Affymetrix) for those samples with tissue available<br />

(14 polysomy and 18 amplified cases) and analyzed using Partek<br />

Genomics Suite.<br />

Results: Tumor grade and size did not differ between polysomic and<br />

amplified samples. Although amplified cases tended to be younger<br />

at diagnosis and less likely to be diagnosed with Stage I tumors, this<br />

difference did not reach statistical significance. HER2 amplified<br />

tumors were more likely to be ER negative (P=0.027) and have lymph<br />

node metastases (P=0.028) than polysomic tumors. Comparison of<br />

polysomic and amplified cases by expression microarray identified<br />

67 genes that were differentially expressed (P2) between the two groups. Thirty-nine genes were more highly<br />

expressed and 28 genes more lowly expressed in tumors with HER2<br />

gene amplification when compared to tumors from patients with<br />

chromosome 17 polysomy.<br />

Discussion: ER and lymph node status of breast tumors from women<br />

with increased HER2 copy number differed depending on whether<br />

the increased copy number was due to HER2 gene amplification or<br />

conversely, polysomy of chromosome 17. Likewise, gene expression<br />

profiles also differed depending on whether increased HER2 was<br />

due to gene amplification or chromosomal gain. Those genes with<br />

significant levels of differential expression between these two groups<br />

are involved in cell cycle control, cell adhesion and migration, cell<br />

differentiation, cytoskeleton organization, signal transduction, protein<br />

synthesis and degradation, and ion transport. Interestingly, the top<br />

4 most significant genes with higher expression in the polysomic<br />

samples, including TUBG1, CPD, BLMH, and JUP, are all located<br />

on 17q. Currently, it is not clear whether treatment with targeted<br />

Her2 therapy is beneficial in patients with chromosome 17 polysomy.<br />

However, the genes identified here may represent novel targets for<br />

developing new treatments that are effective in these women and<br />

require further study.<br />

P3-04-08<br />

Expression of hormone-responsive genes in benign breast tissue<br />

varies with menstrual cycle phase and menopausal status<br />

Hu H, Wang J, Lee O, Shidfar A, Iyer S, Ivancic D, Chatterton RT,<br />

Stearns V, Sukumar S, Khan SA. Northwestern University, Chicago,<br />

IL; Johns Hopkins University School of Medicine<br />

Background: The expression of many genes is known to be regulated<br />

by ambient hormone levels. In a preliminary study of random fineneedle<br />

aspirate (rFNA) samples from benign breast tissue, we found<br />

several genes that were highly correlated with the serum levels of<br />

progesterone (P4) and estradiol (E2). We now present data to further<br />

validate these genes as markers of menstrual cycle phase (MCP) and<br />

menopausal status (MPS) in benign breast tissue which may allow<br />

retrospective classification of archived breast samples with respect<br />

to MCP and MPS at the time of sampling.<br />

Methods: 240 rFNA samples from healthy women with recorded<br />

hormonal data at the time of sampling were analyzed. We divided<br />

these subjects by menstrual cycle phase (MCP) and menopausal<br />

status (MPS): 41 early follicular: (low circulating E2 and P4); 48<br />

mid-cycle (high E2 and low P4); 31 luteal (moderate E2 and high<br />

P4). 120 post-menopausal (low E2 and low P4). 100 ng of RNA from<br />

rFNA samples of the breast was reverse transcribed. Amplicons of<br />

interest were linearly amplified to 14 cycles for 35 genes related<br />

to hormone responsiveness. qPCR reactions were carried out<br />

using the TaqMan OpenArray (Applied Biosystems). For each<br />

gene of interest, expression levels were normalized to the average<br />

expression of GAPDH. Gene expression difference between groups<br />

were conducted using the Mann-Whitney Test. P-values from gene<br />

expression difference were adjusted via the Benjamini-Hochberg<br />

(1995) approach.<br />

Results: The mean value of TNFSF11 expression level was 13.19<br />

fold higher in luteal phase subjects than in post-menopausal subjects<br />

(P=0.0003) where there was also the biggest difference of serum<br />

P4 level between groups. The expression of DIO2 and MYBPC1<br />

was also significantly higher in luteal phase group than in the postmenopausal<br />

group (P=0.005 , P= 0.02, respectively). These 3 genes<br />

also demonstrated a higher expression pattern in luteal phase than<br />

mid-cycle and follicular phase but analysis is still ongoing. All<br />

comparisons between these groups will be presented at the meeting.<br />

Conclusion: The expression levels of TNFSF11, DIO2 and MYBPC1<br />

vary with MCP and MPS. These hormone-responsive genes are<br />

candidate MCP classifiers which could be applied to archived breast<br />

samples to assess whether biomarkers of breast cancer risk are stable<br />

across the menstrual cycle, since MCP and MPS variation is likely<br />

an important source of biological noise in studies of archived breast<br />

biopsy material.<br />

P3-04-09<br />

Withdrawn by Author<br />

P3-04-10<br />

Comparison between RNA-Seq and Affymetrix gene expression<br />

data<br />

Fumagalli D, Haibe-Kains B, Michiels S, Brown DN, Gacquer D,<br />

Majjaj S, Salgado R, Larsimont D, Detour V, Piccart M, Sotiriou<br />

C, Desmedt C. Institut Jules Bordet, Brussels, Belgium; Institut<br />

de Recherches Cliniques de Montréal, Montréal, QC, Canada;<br />

Université Libre de Bruxelles, Campus Erasme, Brussels, Belgium<br />

Background: Microarrays have revolutionized breast cancer research<br />

by enabling gene expression comparisons on a transcriptome-wide<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

300s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 3<br />

scale. They have provided clinically useful information such as the<br />

development of the intrinsic classification as well as several gene<br />

prognostic signatures. Recently, with the advent of next generation<br />

sequencing technology, RNA-Seq has emerged as a promising<br />

technology for digital read-out of the transcriptome. Here, we will<br />

present a comparison of microarray and RNA-Seq data on a series<br />

of 58 breast cancer samples.<br />

Methods: 58 tumors representing the different molecular subtypes [17<br />

triple negative (ER-, PgR-, HER2-); 14 HER2 positive (HER2+); 17<br />

luminal A (ER+, HER2-,histological grade 1), 10 luminal B (ER+,<br />

HER2-,histological grade 3)] were obtained from patients recruited<br />

between 2007 and 2011 at Jules Bordet Institute. Their RNA was<br />

profiled using the Affymetrix HG-U133 Plus 2.0 chips and sequenced<br />

with the ILLUMINA platform, producing ∼30 million 2*50 bp<br />

paired-end reads per sample. The reads alignment and expression<br />

quantification were performed using Tophat 2 and Cufflinks 2,<br />

respectively, on the basis of the Ensembl genome annotations<br />

hg19. Affymetrix microarray data were normalized using fRMA<br />

and probesets were selected using JetSet. A selection of 7 clinically<br />

relevant gene signatures was evaluated.<br />

Results: About 16000 genes were retained for analysis. Comparison<br />

of Affymetrix and RNA-Seq data revealed that 50%, 25% and 5%<br />

of these genes had a Spearman correlation superior to 0.7, 0.82 and<br />

0.92, respectively. The genes with the highest correlation coefficients<br />

between Affymetrix and RNA-Seq tended to have medium/high<br />

expression levels (median Affymetrix log2 expression of 7.5)<br />

consistent with the fact that microarrays have a limited dynamic<br />

range compared to RNA-Seq. We observed an excellent correlation<br />

between Affymetrix and RNA-Seq for the clinically relevant genes<br />

ER (0.97), PgR (0.95) and HER2 (0.90) respectively. We observed,<br />

however, a few discordances between ER and PgR quantification<br />

by immunohistochemistry and RNA-Seq/Affymetrix. We next<br />

measured the consistency of two molecular classifiers, SCMGENE<br />

(Haibe-Kains JNCI2012) and PAM50 (Parker JCO2009), between<br />

Affymetrix arrays and RNA-seq. A kappa coefficient of 0.93 (CI 0.85-<br />

1) and 0.79 (CI 0.66-0.91) was observed for SCMGENE and PAM50,<br />

respectively, with the majority of the discrepancies for PAM50<br />

concerning the HER2 subtype. The first generation proliferationbased<br />

gene expression signatures showed an excellent correlation<br />

between Affymetrix and RNA-Seq: Genomic Grade Index (0.98),<br />

Mammaprint (0.98) and Oncotype (0.97). The immune and stroma<br />

gene signatures (Desmedt et al. 2008) showed a correlation of 0.97<br />

and 0.79 respectively. The lower correlation for the stroma signature<br />

could partly be explained by the fact that RNA-Seq and Affymetrix<br />

are measuring different splice variants.<br />

Conclusions: This is to our knowledge the first study to report a<br />

systematic comparison between RNA-Seq and Affymetrix HG-<br />

U133 Plus2.0 data in breast cancer for individual genes and several<br />

prognostic gene signatures. According to these results, RNA-Seq can<br />

reliably be used for the quantification of breast cancer transcriptome.<br />

P3-04-11<br />

Systemic treatment decision making for patients with stage I and<br />

II, hormone receptor positive, her2/neu negative breast cancer<br />

Zhu X, Graham N, Paquet L, Dent S, Song X. University of Ottawa,<br />

ON, Canada; The Ottawa Hospital <strong>Cancer</strong> Centre, Ottawa, ON,<br />

Canada; Carleton University, Ottawa, ON, Canada<br />

Background: Oncotype DX is a clinically validated risk stratification<br />

tool that can predict the risk of recurrence and the benefit of adjuvant<br />

chemotherapy in women with hormone receptor positive (HR+),<br />

HER2/neu negative early stage breast cancer (EBC). This tool has<br />

been available to oncologists in Ontario since April 2010 at significant<br />

cost, yet no guidelines exist regarding their use. This retrospective<br />

chart review examined the factors that were associated with use of<br />

Oncotype DX at a tertiary care cancer centre.<br />

Materials and methods: One hundred patients (pts) diagnosed with<br />

HR+, HER2/neu negative EBC (stage I-II), who underwent Oncotype<br />

DX testing between April 1, 2010, and June 30, 2011 were included<br />

in the study. A second control group of 100 patients with similar<br />

disease characteristics but who did not receive Oncotype DX testing<br />

were randomly selected. Data collection included demographics,<br />

tumor grade and stage, and Adjuvant! Online recurrence risk scores.<br />

The distribution of patients in each category was compared using the<br />

chi-square test to detect statistically significant differences between<br />

distributions.<br />

Results: Median age in the Oncotype DX group was 58 years (r: 26-<br />

77) and 63 years (r: 30-81) in the control group. 20 patients in the<br />

Oncotype DX group were aged 35-49, 57 patients were aged 50-64,<br />

and 23 patients were aged 65 or older, while the control group had 16,<br />

43, and 41 patients, respectively (p=0.02). The Oncotype DX group<br />

had 72 pre- and perimenopausal pts and 28 postmenopausal patients,<br />

while the control group had 81 and 19 patients, respectively (p=0.13).<br />

20, 56, and 24 pts in the Oncotype DX group had grade 1, 2, and 3<br />

histology, respectively, vs. 44, 44, and 12, respectively in the control<br />

group (p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Methods: We have developed a user-friendly web-based system to<br />

allow the identification and evaluation of genes that are significantly<br />

associated with disease progression and survival for breast cancer in<br />

general and also with respect to molecular subtype. The underlying<br />

algorithm combines gene expression data from multiple DNA<br />

microarray experiments and detailed clinical data to correlate<br />

outcome with gene expression levels. This algorithm integrates<br />

gene expression and survival data from 21 datasets on 10 different<br />

microarray platforms corresponding to 20,017 gene sequences across<br />

3,519 samples.<br />

Results: We demonstrate the robustness of our approach in comparison<br />

to two commercially available prognostic tests in breast cancer.<br />

Our algorithm complements these prognostic tests and is consistent<br />

with their findings. In addition, <strong>Breast</strong>Mark can act as a powerful<br />

reductionist approach to these more complex gene signatures,<br />

eliminating superfluous genes, potentially reducing the complexity<br />

and cost of these multi-index assays. We also applied the algorithm to<br />

examine expression of 58 receptor tyrosine kinases in the basal-like<br />

subtype of breast cancer, identifying seven receptor tyrosine kinases<br />

associated with poor disease-free survival and/or overall survival in<br />

this subtype (EPHA5, FGFR1, FGFR3, VEGFR1, PDGFRβ, ROS,<br />

TIE1). A web application for using this algorithm is currently available<br />

at http://glados.ucd.ie/<strong>Breast</strong>Mark/index.html.<br />

Conclusions: <strong>Breast</strong>Mark is a useful tool for examining putative<br />

prognostic markers at the RNA level in breast cancer. The value of<br />

this tool will be in the preliminary assessment of putative biomarkers<br />

in breast cancer as a whole and within its molecular subtypes. It will<br />

be of particular use to clinical and academic research groups with<br />

limited bioinformatics facilities.<br />

P3-05-01<br />

Molecular subtyping improves stratification of patients into<br />

diagnostically more meaningful risk groups<br />

Cristofanilli M, Turk M, Kaul K, Weaver J, Wesseling J, Stork-Sloots<br />

L, de Snoo F, Yao K. Fox Chase <strong>Cancer</strong> Center; NorthShore University<br />

HealthSystem; Netherlands <strong>Cancer</strong> Institute; Agendia NV<br />

Background: Microarray-based gene expression profiling<br />

demonstrated that breast cancer is a heterogeneous group of different<br />

diseases characterized by distinct molecular aberrations, rather than<br />

one disease. Improved understanding of the molecular phenotypes<br />

of the disease has already shown prognostic and predictive value<br />

and, when prospectively applied, could have dramatic implications<br />

in establishing a more personalized approach to the management<br />

of early-stage breast cancer. Combined use of MammaPrint and a<br />

molecular subtyping profile (BluePrint) identifies disease subgroups<br />

with marked differences in long-term outcome and response to neoadjuvant<br />

therapy [Glück et al. ASCO 2012]. The aim of this study<br />

was to evaluate the implication of accurate molecular subtyping using<br />

MammaPrint and BluePrint in women with early-stage breast cancer<br />

treated at US Institutions following National Comprehensive <strong>Cancer</strong><br />

Network (NCCN) standard guidelines.<br />

Methods: 208 frozen tumor samples from consecutive BC patients<br />

(TI-III, N0-Ib) were obtained from 2 US centers. Median age at<br />

diagnosis was 56 years (range 28–89 years). Between 1992 and<br />

2010 patients were treated either with breast-conserving therapy or<br />

mastectomy with axillary lymph node dissection followed by systemic<br />

adjuvant therapy when indicated. Sixty-three percent of patients<br />

received adjuvant endocrine therapy (ET), 58% received adjuvant<br />

chemotherapy (CT) and 32% received both. Hormone Receptor (HR)<br />

and HER2 status were assessed by immunohistochemistry (IHC)<br />

and fluorescent in-situ hybridization (FISH), following standard<br />

guidelines. Median follow-up was 11.3 years. Survival was assessed<br />

for patient groups according to local pathological assessment and<br />

compared with molecular classification of patients (centrally assessed<br />

full genome expression at Agendia laboratory).<br />

Results: Standard HR and HER2 status assessment revealed that<br />

57% of all tumors examined were luminal-like (ER/PR positive,<br />

HER2 negative), 20% HER2 positive and 24% triple negative.<br />

Molecular classification demonstrated discordance in the following<br />

clinical groups: 16 out of 41 patients previously identified as HER2<br />

positive were reclassified as Luminal-type, with 97% 5-year distant<br />

metastases-free survival (DMFS) for Luminal A (MammaPrint Low<br />

Risk/Luminal-type) and 98% for Luminal B (MammaPrint High<br />

Risk/Luminal-type). Ten patients identified with clinical triplenegative<br />

tumors were reclassified with molecular subtyping as HER2<br />

positive (n=6) and Luminal-type (n=4). Of the patients classified<br />

with BluePrint Basal-type tumors, 58 (28%) had a 5-year DMFS of<br />

82% (81% received adjuvant CT). Of those with HER2-type tumors,<br />

25 (12%) had a 5-year DMFS of 76% (88% received adjuvant CT<br />

without trastuzumab). Discordant cases are being centrally re-assessed<br />

for ER, PR and HER2.<br />

Conclusions: This retrospective study showed that Molecular<br />

Subtyping with BluePrint and MammaPrint leads to clinically<br />

significant prognostic and molecular stratification. The use of<br />

MammaPrint and BluePrint in the management of patients with<br />

primary breast cancer should be considered for a more accurate<br />

selection of adjuvant therapies in this era of personalized medicine.<br />

P3-05-02<br />

Pathological assessment of discordant cases for molecular<br />

(BluePrint and MammaPrint) versus clinical subtypes for breast<br />

cancer among 621 patients from the EORTC 10041/BIG 3-04<br />

(MINDACT) trial<br />

Viale G, Slaets L, de Snoo F, van’t Veer L, Rutgers E, Piccart M,<br />

Bogaerts J, van den Akker J, Stork-Sloots L, Engelen K, Russo L,<br />

Dell’Orto P, Cardoso F. European Institute of Oncology; European<br />

Organisation for Research and Treatment of <strong>Cancer</strong>; Agendia NV;<br />

Netherlands <strong>Cancer</strong> Institute; Jules Bordet Institute; Champalimaud<br />

<strong>Cancer</strong> Center<br />

Background: Biology has become the main driver of breast cancer<br />

therapy. Intrinsic biological subtypes by gene expression profiling<br />

have been identified. Pathology can be used to define surrogates<br />

of these subtypes but these are not always concordant, which may<br />

lead to different treatment plans. We investigated the concordance<br />

between BluePrint and MammaPrint (microarray-based) breast<br />

cancer subtypes and pathological surrogates (based on ER, PR,<br />

HER2 & Ki67).<br />

Methods: Using available data (centrally assessed pathology and<br />

genomics) from the MINDACT pilot phase [Rutgers et al. EJC 2011]<br />

621 tumors were analyzed. Patients were classified according to<br />

4-category based pathology (ER, PR, HER2 and Ki67); additionally,<br />

classification was performed adhering to the recent St. Gallen<br />

recommendations [Goldhirsch et al. 2011], which recognizes an<br />

additional category (Luminal B HER2+). Based on BluePrint and<br />

MammaPrint 4 subtypes are formed: Luminal A (Luminal-type/<br />

MammaPrint Low Risk); Luminal B (Luminal-type/MammaPrint<br />

High Risk); HER2-type; and Basal-type. This study is an analysis of<br />

discordant patient groups (i.e. clinical HER2+/BluePrint Luminaltype;<br />

clinical Hormone Receptor (HR)-positive/BluePrint Basal-type)<br />

providing comparison of centrally assessed tumor heterogeneity as<br />

well as comparison of quantified ER, PR and HER2 results.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

302s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 3<br />

Results: Ki67 is often used as biomarker to distinguish Luminal A<br />

from Luminal B subgroups. The concordance between MammaPrint<br />

and centrally assessed Ki67 in Luminal-type patients is 71%, with<br />

a κ score of 0.35 (95% CI 0.26–0.45) indicating that Ki67 and<br />

MammaPrint cannot reliably substitute for each other. There is a<br />

relatively large group of clinical HER2+ cases that are BluePrint<br />

Luminal-type (29 out of 76; 38%) indicating that tumor expression<br />

of the Luminal profile is dominant compared with expression of the<br />

HER2 profile. These patients have high IHC ER results and fall into<br />

the group that St Gallen separately defines as Luminal B HER2-<br />

type. These patients may have lower response to trastuzumab [von<br />

Minckwitz et al. JCO 2012]. 12 out of 76 BluePrint Basal-type patients<br />

are clinical HR+. These patients have low centrally assessed IHC ER<br />

and PR expression (≥1% and


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Conclusion: The genomic variability prior to therapy was higher in<br />

the partial-responders vs. the complete responders, and the remaining<br />

tumor was even more heterogeneous after treatment than prior to<br />

treatment. Double immunoFISH is a valuable tool for visualization<br />

of both phenotypic and genomic alterations in the same cell in FFPE<br />

sections. The cohort will be expanded to explore the diversity further,<br />

and the results will be presented.<br />

P3-05-05<br />

HER2 Expression and Gene copy analysis by Immunofluorescence<br />

and Fluorescence in situ Hybridization, on a Single formalin-fixed<br />

paraffin-embedded tissue section<br />

Ha T, Seppo A, Ginty F, Kenny K, Henderson D, Kyshtoobayeva A,<br />

Gerdes M, Larriera A, Liu X, Corwin A, Zingelewicz S, Lazare M,<br />

Jun N, Kyshtoobayeva A, Chow C, Al-Kofahi Y, Hollman D, Bloom K.<br />

GE Global Research, Niskayuna, NY; Clarient Diagnostics Services,<br />

Aliso Viejo, CA<br />

Introduction<br />

<strong>Breast</strong> cancer is the most common cancer for women worldwide.<br />

HER2 expression and gene copy number are important when<br />

determining eligibility for adjuvant therapy and/or chemotherapy<br />

medications. One challenging issue for breast cancer testing is<br />

intratumoral heterogeneity of HER2 gene amplification. Intratumoral<br />

heterogeneity can make it difficult to localize target cells of interest.<br />

Serial tissue sections used for independent H&E, IHC and FISH stains<br />

also increase the difficulty to localize targets due to cellular truncation.<br />

We have developed a system to assess both HER2 expression and<br />

gene copy number on the same cell.<br />

Method<br />

Immunofluorescence (IF) and Fluorescence in situ Hybridization<br />

(FISH) were performed on tissue sections from 19 patients with<br />

invasive ductal breast carcinoma. Cases were selected based on prior<br />

HER2 FISH results (HER2:Chromosome 17 = ratio) representing<br />

unamplified (


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 3<br />

were case dependent. For instance, the main cause of over-counting<br />

errors was image noise and artifacts. On the other hand, the main<br />

cause of under-counting was low image contrast, especially in highly<br />

amplified cases. These results are an early indication of the promise<br />

of automatic dot counting applied to breast cancer slides multiplexed<br />

for Her2 IF and FISH.<br />

P3-05-07<br />

Poor prognosis early breast cancer: pathological characteristics<br />

of the Unicancer-PACS08 trial including patients treated with<br />

docetaxel or ixabepilone in adjuvant setting<br />

Lacroix-Triki M, Delrée P, Filleron T, Penault-Llorca F, Bor C, Mery<br />

E, Maisongrosse V, Génin P, Jacquemier J, Reyre J, Caveriviere P,<br />

Quintyn-Ranty M-L, Escourrou G, Mesleard C, Lemonnier J, Martin<br />

A-L, Campone M. Institut Claudius Regaud, Toulouse, France;<br />

Institut de Pathologie et de Génétique, Gosselies, Belgium; Centre<br />

Jean Perrin, Clermont-Ferrand, France; Centre Francois Baclesse,<br />

Caen, France; Centre Alexis Vautrin, Vandoeuvre les Nancy, France;<br />

Institut Paoli Calmettes, Marseille, France; Laboratoire Anatomie<br />

et de Cytologie des Feuillants, Toulouse, France; CHU Rangueil,<br />

Toulouse, France; R&D Unicancer, Paris, France; ICO Centre René<br />

Gauducheau, Saint Herblain, France<br />

Background: The PACS08 trial aimed to compare adjuvant FEC100-<br />

Docetaxel regimen to FEC100-Ixabepilone (Ixa) in poor prognosis<br />

early breast cancer (BC). The study population included BC<br />

patients presenting with triple-negative (TN) [i.e. estrogen receptor<br />

(ER)-/progesterone receptor (PR)-/HER2-] or ER+/PR-/HER2-<br />

tumor, which are subgroups significantly associated with worse<br />

prognosis. Central review was performed and detailed pathological<br />

characteristics of the cohort are reported herein.<br />

Patients and method: Between 2007 and 2010, 762 patients with<br />

unilateral TNBC (n=592, 78%) or ER+/PR-/HER2- BC (n=170, 22%)<br />

were enrolled. Recruitment was interrupted due to BMS decision to<br />

stop Ixa development in adjuvant setting. As defined by inclusion<br />

criteria, TNBC were either node+ or node-, and ER+/PR-/HER2-<br />

BC only node+. Following the validation of ER, PR and HER2<br />

status on whole sections prior to inclusion, paraffin blocks (n=754)<br />

were sent for central pathology review, tissue microarray (TMA)<br />

construction and constitution of the trial collection for translational<br />

research studies. Review of the cases (n=754) was performed by a<br />

board of expert breast pathologists on a one-week working session<br />

with discussion of the difficult cases under a multihead microscope.<br />

Tumor characteristics were assessed on whole tissue sections.<br />

Immunohistochemical detection of Ki67, EGFR, cytokeratins<br />

(CK)5/6 and 14, was performed on TMAs.<br />

Results: TNBC were significantly associated with younger age at<br />

diagnosis (median age 51yr vs 57.5yr in the ER+/PR- subgroup,<br />

p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

“all-high” expression group /n=70/; 5) CLDN3-low (CLDN4, -7<br />

and ECDH-high) group /n=68/ and 6) “all-low” group /n=15/. The<br />

latter was characterised by the CLDN4-low feature as compared to<br />

the other groups.<br />

Conclusion: Expression of JPs can have prognostic relevance in breast<br />

carcinomas. The groups with low expression of ECDH have good<br />

prognosis, while gradually increasing JP expression outlines poor<br />

prognostic tumours. Strikingly the drop in all JPs expression - most<br />

likely resembling the claudin-low subtype - specifies the subgroup<br />

with the poorest outcome, which still remains an infrequent disease<br />

entity.<br />

Grant support: TÁMOP-4.2.2/B-10/1-2010-0013.<br />

P3-05-09<br />

The role of TGF-beta receptor type 3 in breast cancer progression<br />

Jovanovic B, Ashby WJ, Zijlstra A, Pietenpol JA, Moses HL.<br />

Vanderbilt University, Nashville, TN<br />

<strong>Breast</strong> cancer is a diverse group of diseases. Gene expression profiling<br />

has identified luminal A, luminal B, Her2-like, Normal breast-like<br />

and Basal-like as major classifications of breast cancer diseases.<br />

These correlate to the overall patient survival with the basal-like<br />

subtype, clinically termed as triple-negative breast cancers (TNBC),<br />

demonstrating the poorest overall outcome. The poor patient outcome<br />

is primarily due to absence of hormonal receptor targets ER, PR<br />

and HER2 which are the major drug-targets for currently available<br />

therapies. Considering that TNBC lacks well-defined molecular<br />

targets, our group pursued extensive genomic molecular and<br />

biological analysis of over 500 tumor tissues from TNBC patients.<br />

Ultimately, this led to the discovery of six, TNBC subtypes; among<br />

which are Mesenchymal-Stem Like (MSL) and Mesechymal (M).<br />

These two subtypes share a unique biological driver, the TGF-beta<br />

pathway.<br />

To gain insight into the role of the TGF-β pathway signaling in<br />

TNBC we have performed gene expression analysis on all six<br />

TNBC subtypes. A major finding was that among all TGF-β<br />

pathway-associated genes, TGF-beta receptor type 3 (TBR3) is most<br />

differentially expressed in MSL and M tumor subtypes of TNBC.<br />

Based on these findings, we are hypothesizing that TBR3 is required<br />

for maintenance of tumorigenicity in MSL and M subtypes of the<br />

triple-negative breast cancer.<br />

In order to test our hypothesis we have screened 15 TNBC cell lines<br />

for both RNA and protein levels of TBR3 and validated that TBR3<br />

is differentially expressed in M and MSL subtypes. Furthermore,<br />

we have manipulated TBR3 expression in M and MSL TNBC cell<br />

lines and tested their ‘biologies’ using both an in vitro and in vivo<br />

model systems. Preliminary results indicate that upon silencing TBR3<br />

expression, cell motility as well as ability to cluster together in 3D<br />

culture system is significantly changed. In addition, our xenograft<br />

mouse model also demonstrated a significant change in tumor onset<br />

and growth.<br />

We are interested in validating the prognostic and functional<br />

contribution of TBR3 to the breast cancer progression. The results<br />

of this study will provide insight into the potential use of the TGF-β<br />

signaling axis for prognostic or therapeutic utility in breast cancer<br />

patients.<br />

P3-06-01<br />

Hotspot mutations in PIK3CA are predictive for treatment<br />

outcome on aromatase inhibitors but not for tamoxifen<br />

Ramirez Ardila DE, Helmijr JC, Lurkin I, Look M, Ruigrok-Ritstier<br />

K, Simon I, Van Laere S, Sweep F, Span P, Linn S, Foekens J, Sleijfer<br />

S, Berns EMJJ, Jansen MPHM. Erasmus MC - Daniel den Hoed,<br />

Rotterdam, Zuid Holland, Netherlands; Agendia BV, Amsterdam,<br />

Netherlands; Antwerp University/Oncology Centre, GZA Hospitals<br />

St-Augustinus, Antwerp, Belgium; Radboud University Nijmegen<br />

Medical Center, Nijmegen, Netherlands; Netherlands <strong>Cancer</strong><br />

Institute, Amsterdam, Netherlands<br />

Introduction: PIK3CA is the most frequent (30%) mutated oncogene<br />

in breast cancer and may lead to an activation of the PI3K/AKT/<br />

mTOR-pathway. Cell lines resistant to tamoxifen have an activated<br />

PI3K-pathway. Phase III clinical trials show substantial benefit when<br />

mTOR-inhibitors are added to aromatase inhibitor treatment. On<br />

the other hand, patients with PIK3CA exon 20 mutation expression<br />

signature show better treatment outcome after adjuvant tamoxifen<br />

therapy. To address this controversy, we evaluated the PIK3CA<br />

mutation status in 1423 primary breast cancer specimens for its<br />

relationship with prognosis and treatment outcome after first-line<br />

endocrine treatment.<br />

Methods: Hotspot mutations in exon 9 and 20 of PIK3CA were<br />

detected by multiplex snapshot analyses. Mutation status in ERpositive<br />

tumors was related to metastasis free survival (MFS) in<br />

292 untreated lymph node negative (LNN) patients and time to<br />

progression (TTP) in patients with metastatic disease treated with<br />

first-line tamoxifen (N=482) or aromatase inhibitors (AIs; N=103).<br />

Whole genome mRNA and miRs expression profiling was performed<br />

in the latter patient subset to develop a PIK3CA gene signature in<br />

64 specimens. This was validated in 28 independent ER-positive<br />

specimens.<br />

Results: We could evaluate 1371 specimens and detected 437 hotspot<br />

mutations for PIK3CA (32%). Mutations in exon 20 were detected in<br />

256 patients (59%), of which 40 cases with a H1047L (16%) and 216<br />

with a H1047R (84%) mutation. Mutations in PIK3CA exon 9 were<br />

discovered in 174 patients (41%), with E542K and E545K mutations<br />

in 59 (34%) and 105 (60%) cases, respectively, as the most prevalent<br />

ones. Finally, 6 patients had PIK3CA double mutations for both exon<br />

9 and 20 and one patient had a E542K and E545K mutation.<br />

Evaluation of the untreated LNN patients for prognosis showed no<br />

relationship between MFS and PIK3CA mutations (HR= 1.07 [95%<br />

CI: 0.73-1.56]; P=0.73), neither for exon 9 nor exon 20 compared to<br />

wild-type. In the multicenter cohort of 482 patients with advanced<br />

disease treated with first-line tamoxifen no link with treatment<br />

outcome and PIK3CA mutation status was observed (HR= 1.13 [95%<br />

CI: 0.92-1.38]; P=0.24). However, patients with advanced disease<br />

treated with first-line AIs (N=64) showed a significant longer TTP<br />

for patients with a PIK3CA mutation compared to wild-types (HR=<br />

0.46 [95% CI: 0.25-0.87]; P=0.017).<br />

Expression profiles of mRNAs and miRs were integrated and<br />

resulted in signatures for PIK3CA status discovered by pathway (17<br />

genes) and expression analyses (10 genes and 9 miRs). Validation<br />

and comparison with published signatures in the 28 independent<br />

tumors showed that our 10-genes signature had the highest PIK3CA<br />

prediction accuracy (75%), with 60% sensitivity and 78% specificity.<br />

Moreover, this signature better associates with TTP (HR= 0.38 [95%<br />

CI: 0.20-0.72]; P = 0.003).<br />

Conclusion: Mutations in PIK3CA are not prognostic value in<br />

untreated ER-positive LNN patients, not predictive in ER-positive<br />

patients with advanced disease treated with first-line tamoxifen<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

306s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 3<br />

therapy, however, are predictive for favourable outcome after first-line<br />

AIs. Moreover, we propose an expression signature of 10 genes as<br />

a putative biomarker to predict the PI3K status and response to AIs.<br />

P3-06-02<br />

Genetic polymorphisms from genome-wide association study<br />

associated with the metabolic and cell proliferation pathways<br />

affect the time to distant metastases of hormone receptor-positive<br />

and Her2-negative early breast cancer<br />

Huang C-S, Kuo S-H, Yang S-Y, Lien H-C, Lin C-H, Lu Y-S, Cheng<br />

A-L, Chang K-J. National Taiwan University Hospital and National<br />

Taiwan University College of Medicine, Taipei, Taiwan; College of<br />

Public Health, National Taiwan University, Taipei, Taiwan<br />

Background:<br />

Single nucleotide polymorphisms (SNPs) identified from genomewide<br />

association study (GWAS) have been found to be associated<br />

with breast cancer risk. We hypothesized that candidate genes and<br />

genes derived from GWAS involving in the Estrone/Estradiol (E2)/<br />

Tamoxifen biosynthesis may influence the adjuvant hormonal therapy<br />

effect and the survival. In this study, we sought to investigate whether<br />

these SNPs are associated with prognosis of hormone receptor (HR)-<br />

positive breast cancer patients, especially in HER2-negative patients.<br />

Patients and methods:<br />

We selected breast cancer susceptibility SNPs identified by GWAS,<br />

SNPs in tamoxifen metabolizing related genes, and SNPs in estrogen<br />

receptor genes and estrogen metabolism genes, and genotyped for<br />

variations of above genes, including ALDH3A1, CYP2C19, COMT,<br />

CYP19, MAP3K1, FGFR2, TNRC9, HCN1, ERCC4, CYP3A5,<br />

UGT1A1, ER, ABCG2, CYP2B6, CYP2D6, 5p12 in 171 hormone<br />

receptor-positive, and Her2-negative early breast cancer patients<br />

(127 with negative lymph node [LN], and 44 with 1-3 positive LN).<br />

All patients received adjuvant hormonal therapy. The associations<br />

were examined between SNPs and distance disease-free survival<br />

(DDFS), and overall survival (OS) by using the log-rank test and<br />

Cox’s proportional hazard model. Furthermore, we combined<br />

clinicopathologic features and SNPs into the risk score analysis to<br />

further validate above identified genetic markers.<br />

Results:<br />

We found that SNPs of CYP2B6 (rs3211371), FGFR2 (rs2981582),<br />

and MAP3K1 (rs889312) were significantly associated with DDFS<br />

and OS. Furthermore, in lymph node-negative patients, CYP2B6<br />

(rs3211371), FGFR2 (rs2981582), MAP3K1 (rs889312) and 5p12<br />

(rs10941679 and rs4415084) were significantly associated with DDFS<br />

and OS, while CYP3A5 (rs776746) was significantly associated with<br />

OS but not DDFS. We further assessed the associations of disease<br />

prognosis with the number of high-risk genotypes in CYP3A5,<br />

FGFR2, and MAP3K1, and showed significant dose-response<br />

relationships between the number of high-risk alleles at these 3 loci<br />

and DDFS (P = 0.005 for trend) and OS (P = 0.0008 for trend). When<br />

combining the clinicopathologic features and SNPs into the risk<br />

score analysis, patients were divided into 3 subgroups (subgroup 1,<br />

risk score1.708, n=43 [LN-positive, n=11]). We found that around 20 %<br />

of subgroup 3 patients had early development of distant metastases<br />

(DM) in the upfront 3 years (the trend of DM appeared to persist at<br />

least 10 years), and subgroup 2 patients had higher risk of DM after<br />

5 years, whereas subgroup 1 patients had no development of DM<br />

even after 12 years.<br />

Conclusion:<br />

Our results indicate that in addition to drug metabolic genes, genes<br />

related to cell proliferation, anti-apoptosis, and signaling transduction,<br />

for example, CYP3A5, CYP2B6, FGFR2, and MAP3K1 genes were<br />

associated with DDFS and OS in HR-positive/Her2-negative breast<br />

cancer patients. These findings provide additional insight that the<br />

genetic variants, or host factors, may affect the prognosis of breast<br />

cancer.<br />

P3-06-03<br />

Association between PAM50 breast cancer intrinsic subtypes and<br />

effect of gemcitabine in advanced breast cancer patients<br />

Jørgensen CLT, Nielsen TO, Bjerre KD, Liu S, Wallden B, Balslev<br />

E, Nielsen DL, Ejlertsen B. Herlev University Hospital, Herlev,<br />

Denmark; Danish <strong>Breast</strong> <strong>Cancer</strong> Cooperative Group, Copenhagen,<br />

Denmark; University of British Columbia, Vancouver, BC, Canada;<br />

NanoString Technologies, Seattle, WA<br />

Background<br />

<strong>Breast</strong> cancer can be subclassified into luminal A and B, basal-like<br />

and HER2-enriched subtypes, each with distinct biology, prognosis<br />

and response to therapy. Pre-clinical studies have suggested that a<br />

basal-like subtype is associated with responsiveness to gemcitabine.<br />

Additional data suggest that patients with highly proliferating tumors<br />

(non-luminal A) might be particularly responsive to gemcitabine.<br />

We examined these hypotheses on SBG0102, a randomized trial<br />

comparing gemcitabine plus docetaxel (GD) to single agent docetaxel<br />

(D) in patients with advanced breast cancer. Secondarily, we analyzed<br />

prognosis by PAM50 subtype, which has not previously been<br />

addressed in a metastatic setting.<br />

Patients and Methods<br />

RNA was isolated from archival formalin-fixed, paraffin-embedded<br />

primary breast tumor tissue from patients randomly assigned to<br />

GD or D, and analyzed with NanoString Technologies’ nCounter<br />

system and PAM50 intrinsic subtyping algorithm. Using time to<br />

progression (TTP) as primary endpoint, overall survival (OS) and<br />

response rate (RR) as secondary endpoints, relationships between<br />

subtypes and outcome after chemotherapy were analyzed by methods<br />

prespecified in writing as a formal prospective-retrospective clinical<br />

trial correlative study. Data analysis was performed independently<br />

by the DBCG statistical core.<br />

Results<br />

RNA from 270 patients was evaluable. 84 patients (31%) were<br />

classified as luminal A, 97 (36%) luminal B, 43 (16%) basal-like,<br />

and 46 (17%) as HER2-enriched. PAM50 intrinsic subtype was<br />

significantly associated with TTP (P=.0006) and OS (P=.0083) by<br />

Kaplan-Meier analysis. In the adjusted Cox model PAM50 remained<br />

independently prognostic. RR was not significantly different by<br />

subtype, and PAM50 was not a significant predictor of TTP by<br />

treatment arm. PAM50 was however a highly significant predictor of<br />

OS following GD compared to D (P interaction =.0008). The 43 patients<br />

with a basal-like subtype had a significant reduction in OS events<br />

(hazard ratio (HR)=0.27; 95% CI 0.14-0.52; P interaction =.0004), although<br />

TTP events were not significant by interaction test (HR=0.37; 95%<br />

CI 0.18-0.76; P interaction =.19). Kaplan-Meier estimates revealed a<br />

gain in median OS of 10 months in the doublet arm compared to<br />

the monotherapy arm (18.7 (95% CI, 12.4-23.0) v 8.5 (95% CI,<br />

4.2-11.8) months).<br />

Conclusion<br />

A significantly improved and clinical important prolongation of<br />

survival was seen from the addition of gemcitabine to docetaxel in<br />

advanced basal-like breast cancer patients. However, we could not<br />

www.aacrjournals.org 307s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

show a similar significant reduction in TTP events. These results need<br />

validation in a second similar trial or a prospective study stratified<br />

for basal-like breast cancer to obtain convincing evidence that<br />

identification of a basal-like profile may be used to direct the use of<br />

gemcitabine in combination with docetaxel.<br />

P3-06-04<br />

Role of pMAPkinase, pAKT, p27 & IGF-IR as predictive markers<br />

of response to trastuzumab in patients with HER2-positive<br />

invasive breast cancer treated with neoadjuvant chemotherapy<br />

+ trastuzumab in the REMAGUS02 trial<br />

Mathieu MC, Goubar A, Sigal B, Bertheau P, Guinebretière JM, André<br />

F, Pierga JY, Delaloge S, Giacchetti S, Brain E, Marty M. Institut<br />

Gustave Roussy - INSERM U981, Villejuif, France; Institut Curie,<br />

Paris, France; Hopital Saint-Louis, Paris, France<br />

Background: Predicting a benefit from trastuzumab in patients with<br />

HER2+ breast cancer remains an important goal. Possible mechanisms<br />

of resistance include altered receptor antibody interaction, Akt and<br />

MAPK pathways, and loss of p27. The objective of this study was to<br />

determine the correlation between pMAPkinase (pMAPK), pAKT,<br />

p27, IGF-IR protein expression and the benefit of trastuzumab for<br />

patients randomized to chemotherapy (CT) alone and CT with<br />

trastuzumab.<br />

Patients and methods: From May 2004 to October 2007, 120<br />

patients with stage II and III HER2+ breast carcinomas were<br />

enrolled in a phase II trial of neoadjuvant chemotherapy (CT) with<br />

epirubicin-cyclophosphamide (4 courses) followed by docetaxel<br />

± trastuzumab (T) (4 courses). A complete pathological response<br />

(pCR) was defined by the absence of residual invasive carcinoma<br />

in the breast and axillary lymph nodes. A tissue microarray was<br />

constructed from paraffin-embedded tumor samples collected prior to<br />

neoadjuvant chemotherapy. Patients’ tumours were scored HER2 3+<br />

immunohistochemically (IHC) or 2+ IHC with HER2 amplification<br />

by FISH. Immunohistochemical analysis of pMAPK, pAKT, p27 and<br />

IGF-IR was performed on tumor tissue microarrays before CT. The<br />

H-score (intensity x %) was evaluated. Specimens were classified<br />

as exhibiting high or low expression based on a median value as the<br />

cut-off point for each marker. A logistic regression model, including<br />

the marker and its interaction with treatment, was used to analyse the<br />

markers predictive of a treatment effect on the pCR. The independent<br />

predictive value was analysed in a multivariate logistic regression<br />

adjusting on the lymph node and ER status.<br />

Results: 117/120 (97.5%) patients had sufficient tumor for the<br />

analysis. The pCR rate was 19% in the CT arm and 25% in the CT+T<br />

arm. The median H-score was : pMAPK = 28, pAKT= 25, p27= 50 and<br />

IGF-IR = 15. No significant difference was observed in the pCR rate<br />

between the two arms according to pAKT, p27, IGF-IR expression.<br />

The pCR rate was higher in CT+T compared to CT alone in patients<br />

with high pMAPK expression (OR= 4.7 (0.9- 24.2); interaction p =<br />

0.03). No difference was observed in the pCR rate in patients with<br />

low pMAPK expression (OR= 0.5 (0.1- 1.8).<br />

Conclusions: In HER2-positive breast cancers, pMAPK expression<br />

evaluated by IHC was significantly associated with a pathological<br />

response in the arm with neoadjuvant trastuzumab. High pMAPK<br />

expression could be a predictive marker of response to trastuzumab<br />

in a CT +T regimen.<br />

P3-06-05<br />

Expression of SPARC in human breast cancer and its predictive<br />

value in the GeparTrio neoadjuvant trial.<br />

Untch M, Prinzler J, Fasching P, Müller BM, Gade S, Meinhold-<br />

Heerlein I, Huober J, Karn T, Liedtke C, Loibl S, Müller V, Rack<br />

B, Schem C, Darb-Esfahani S, von Minckwitz G, Denkert C. Helios<br />

Klinikum Berlin-Buch, Germany; Charité Universitätsmedizin<br />

Berlin, Germany; Universitätsklinikum Erlangen, Germany; German<br />

<strong>Breast</strong> Group, Germany; Universitätsklinikum Aachen, Germany;<br />

Universitätsklinikum Düsseldorf, Germany; Universitätsklinikum<br />

Frankfurt am Main, Germany; Universitätsklinikum Münster,<br />

Germany; Universitätsklinikum Hamburg-Eppendorf, Germany;<br />

Universitätsklinikum München; Universitätsklinikum Schleswig-<br />

Holstein, Germany<br />

Background: Secreted protein acidic and rich in cysteine (SPARC)<br />

is an albumin-binding protein and associated with poor prognosis in<br />

multiple cancers.<br />

The aim of this analysis was to determine the frequency of SPARC<br />

expression among different molecular breast cancer subtypes and to<br />

evaluate its predictive value for therapy response after neoadjuvant<br />

anthracycline/taxane based chemotherapy (CTX) in participants of<br />

the GeparTrio trial.<br />

Methods: We evaluated tumoral SPARC expression by<br />

immunohistochemistry on tissue microarrays (TMAs) constructed<br />

from formalin-fixed paraffin-embedded (FFPE) pre-treatment core<br />

biopsies from 667 patients (pts) of the GeparTrio trial. The details of<br />

the GeparTrio study design are described elsewhere (von Minckwitz,<br />

JNCI 2008). Cutoffs for SPARC expression were determined using the<br />

web-based software Cutoff Finder (http://molpath.charite.de/cutoff/).<br />

Results: SPARC protein expression was measurable by IHC on<br />

the TMAs and 176 (26.4 %) of 667 tumors were SPARC positive.<br />

SPARC expression was increased in pts with triple-negative breast<br />

cancer (TNBC) compared to hormone receptor or HER2 positive<br />

subtypes (p = 0.039).<br />

SPARC positivity was associated with an increased pCR rate in the<br />

overall population (p < 0.001). SPARC negative tumors had a pCR<br />

rate of 15%, which increased to 27% in SPARC positive tumors.<br />

Similarly, in TNBC the pCR rate increased from 26% in SPARC<br />

negative tumors to 47% in SPARC positive tumors (p = 0.032).<br />

In multivariate logistic regression analysis adjusted for standard<br />

clinicopathological factors, SPARC was independently predictive<br />

in the overall population (p = 0.010) as well as the subgroups of pts<br />

with TNBC (p = 0.036).<br />

Conclusions: SPARC is expressed in all biological breast cancer<br />

subtypes with TNBC revealing the highest expression rate. Our data<br />

suggest that SPARC expression may provide predictive information<br />

for response to neoadjuvant CTX.<br />

As SPARC is an albumin-binding protein and might mediate<br />

intratumoral accumulation of nab-Paclitaxel, prospective analysis of<br />

SPARC expression is planned in the GeparSepto trial.<br />

P3-06-06<br />

Lactate dehydrogenase B in breast cancer contributes to glycolytic<br />

phenotype and predicts response to neoadjuvant chemotherapy<br />

Dennison JB, Molina JR, Mitra S, Gonzalez-Angulo AM, Brown RE,<br />

Mills GB. MD Anderson <strong>Cancer</strong> Center, Houston, TX; University of<br />

Texas Health Science Center, Houston, TX<br />

Purpose: Although breast cancers are known to be molecularly<br />

heterogeneous, their metabolic heterogeneity is less well understood.<br />

This study aimed to identify and evaluate metabolic biomarkers in<br />

breast cancers and determine their ability to predict outcomes.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

308s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 3<br />

Methods: mRNA microarray data from breast cancer cell lines were<br />

used to identify bimodal genes, those with the highest potential for<br />

robust high/low classification in a clinical setting. Using a panel of<br />

breast cancer cell lines, expression and activity of the highest scoring<br />

bimodal metabolism gene, lactate dehydrogenase B (LDHB), was<br />

quantified and associated with glycolytic phenotype. The contribution<br />

of LDHB to glycolysis was evaluated using MDA-MB-231 and<br />

HCC1937 cell lines with stable lentiviral knockdown of LDHB.<br />

mRNA expression of LDHB was evaluated for association with<br />

neoadjuvant chemotherapy response within clinical and PAM50-<br />

derived subtypes.<br />

Results: LDHB was highly expressed in cell lines with glycolytic,<br />

basal-like phenotypes. Knockdown of LDHB in cell lines reduced<br />

glycolytic dependence, linking LDHB expression directly to<br />

metabolic function. Using four independent patient datasets, LDHB<br />

mRNA expression was positively associated with basal subtype and<br />

negatively associated with luminal and HER2 subtypes. Furthermore,<br />

LDHB predicted basal phenotype independently of hormone-receptor<br />

(HR) clinical status (OR = 21.6 for HR-positive/HER2-negative<br />

and OR = 18.2 for triple-negative). While LDHB expression could<br />

predict basal phenotype, high LDHB expression identified aggressive<br />

breast cancer tumors that were primarily but not exclusively basal.<br />

Importantly, high LDHB expression predicted pathological complete<br />

response to neoadjuvant chemotherapy for both hormone receptor<br />

(HR) positive/HER2-negative (OR = 4.0, P = .0002) and triplenegative<br />

(OR = 3.0, P = .003) cancers. Consistent with increased<br />

response to chemotherapy, LDHB expression in basal cancers within<br />

the triple-negative group was associated with the proliferative marker<br />

CCNB1 (P < .0001).<br />

Conclusion: mRNA expression of LDHB as a single marker predicted<br />

glycolytic phenotype in cell lines and response to neoadjuvant<br />

chemotherapy in breast cancers independently of HR status. These<br />

observations support prospective clinical evaluation of LDHB as a<br />

predictive marker of response for breast cancer patients treated with<br />

neoadjuvant chemotherapy.<br />

P3-06-07<br />

Ki67 as a Predictive Marker of Response to Neoadjuvant<br />

Chemotherapy in Patients with Early-Stage <strong>Breast</strong> <strong>Cancer</strong><br />

(ESBC): A Systematic Review and Evidence Summary<br />

Lyman GH, Culakova E, Poniewierski MS, Wogu AF, Barry W,<br />

Ginsburg GS, Marcom PK, Ready N, Abernethy A, Geradts J, Hwang<br />

S, Kuderer NM. Duke University<br />

Background: Immunohistochemical (IHC) assessment of the<br />

proportion of cells staining for the KI67 nuclear antigen is being<br />

increasing utilized in the management of patients with early-stage<br />

breast cancer (ESBC). A comprehensive systematic review and<br />

evidence synthesis of biomarkers potentially predictive of response to<br />

systemic therapy was initiated as a part of an NCI-funded comparative<br />

effectiveness research program.<br />

Methods: Studies of chemotherapy response prediction based on<br />

baseline IHC assessment of Ki67 in patients with ESBC receiving<br />

neoadjuvant systemic therapy were identified. Response was specified<br />

as pathologic complete response (pCR) or clinical response (ClinR).<br />

Assay predictive performance for response was assessed on the basis<br />

of sensitivity, specificity, predictive value and predictive odds ratio<br />

(POR±95%CLs) utilizing mixed effects models. Study results were<br />

fitted in an ROC analysis based on the method of DerSimonian and<br />

Laird. Publication bias was evaluated on the basis of funnel plot<br />

asymmetry assessed by Egger’s regression intercept and Begg and<br />

Mazumdar’s rank correlation.<br />

Results: Of 469 potentially eligible studies, dual blind full text review<br />

identified 42 eligible studies reporting 44 independent cohorts with<br />

6,716 patients (21–979). While Ki67 cutpoints varied considerably,<br />

they were most commonly between 10%-30% (median 20%, range<br />

1-50%). The analysis prsented here is limited to the 30 studies of<br />

ESBC patients (N=3,343) receiving neoadjuvant therapy of which<br />

14 reported fewer than 100 patients. The proportion of patients with<br />

elevated Ki67 across studies ranged from 0.20-0.92 (median=0.54).<br />

Sensitivity and specificity for treatment response in patients with high<br />

vs. low baseline Ki67 was 0.65 [0.61, 0.68] and 0.52 [0.50, 0.54],<br />

respectively. Estimated response rates across studies in patients with<br />

high vs. low Ki67 were 31% [29%, 34%] and 19% [17%, 21%],<br />

respectively. The estimated POR for response across studies was<br />

2.82 [2.14, 3.72; P


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

bead-based extraction method (STRATIFYER XTRAKT kits). Ki-67,<br />

TOP2A and RACGAP1 as well as CALM2 as a house keeping gene<br />

were measured via a multiplex quantitative RT-PCR (qPCR). Kaplan-<br />

Meier survival estimates, patitioning test and correlation analyses<br />

were performed using the SAS JMP® 9.0.0 software.<br />

Results<br />

There was only a moderate correlation between Ki-67 mRNA or<br />

Ki-67 measured by immunohistochemistry (IHC) and histologic<br />

grade (Spearman r=0.52 p


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 3<br />

associated with achievement of pCR in univariable analysis (OR<br />

1.01, 95% CI 0.96, 1.05) and after adjustment for hormone receptor<br />

status (HRS; OR 1.02, 95%CI 0.97, 1.07), tumor grade (OR 1.01,<br />

95%CI 0.96, 1.06), Ki67 (OR 1.02, 95%CI 0.97, 1.07) or BMI (OR<br />

1.00, 95%CI 0.96, 1.05). Vitamin D was independently associated<br />

with Ki67 (OR 0.95, 95%CI 0.91, 0.99). Vitamin D level was also<br />

not associated with 3 year RFS in univariable analysis (HR 0.98, 95%<br />

CI 0.95, 1.02), and after adjustment for HRS (HR 0.99, 95%CI 0.95,<br />

1.03), tumor grade (HR 0.99, 95%CI 0.95, 1.02), Ki-67 (HR 0.99,<br />

95%CI 0.95, 1.03), BMI (HR 0.96, 95% CI 0.92, 1.01) or pCR (HR<br />

0.99, 0.95, 1.02); no interaction was seen when stratifying on HRS<br />

(p-interaction= 0.71).<br />

Conclusions: The majority of patients in this study had insufficient<br />

vitamin D levels (2, and deletion was defined as FISH ratio


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

1. Multivariate analyses of age, nodal status, ER status, and gene<br />

expression shown below in Table 2 indicate that neither cMYC nor<br />

TOP2A status had an effect on outcome. Only nodal and ER status<br />

affected outcome.<br />

Table 1<br />

Parameter n 3-Yr DFS (%) 3-Yr OS (%)<br />

TOP2A Amplified 190 97.1 98.5<br />

Normal 130 97.6 98.5<br />

Deletion 118 94.8 97.9<br />

cMYC Amplified 99 96.8 99.00<br />

Normal 246 97.8 99.6<br />

Deletion 91 94.2 96.0<br />

ER Status Positive 320 98.3 99.4<br />

Negative 173 93.7 97.0<br />

Positive Node Yes 102 92.9 97.1<br />

No 391 97.8 98.9<br />

Table 2. Summary of Multivariate Analysis<br />

Parameter Hazard Ratio p-value<br />

Age Every Year 1.03 0.26<br />

TOP2A Amp. vs. Not-Amp 0.96 0.95<br />

cMYC Amp. vs.Not-Amp 1.01 0.99<br />

Nodes Positive vs. Negative 2.87 0.06<br />

ER Positive vs. Negative 0.33 0.05<br />

T status T2/T3 vs. T1 2.06 0.20<br />

Conclusion:<br />

Outcome (DFS + OS) in a phase II study of a nonanthracycline<br />

regimen (TC) coupled with H was unaffected by cMYC or TOP2A<br />

gene copy number status. Only ER and nodal status showed an effect<br />

in a multivariate analysis.<br />

P3-06-13<br />

Expression of Phosphorylated Activating Transcription factor<br />

2 (ATF2) is associated with sensitivity to endocrine therapy in<br />

breast cancer<br />

Palmieri C, Rudraraju B, Abdel-Fatah TMA, Moore DA, Shaw J,<br />

Green A, Ellis IO, Coombes RC, Simak A. Imperial College London,<br />

United Kingdom; Nottingham University City Hospital, Nottingham,<br />

United Kingdom; University of Leicester, United Kingdom<br />

Background<br />

The Activating Transcription Factor 2 (ATF2) gene is a member of<br />

the ATF and CREB group of bZIP transcription factors. It is involved<br />

in in transcriptional control, chromatin remodelling and the DNA<br />

damage response. Two threonine residues (Thr69 and Thr71) within<br />

the activation domain (AD) are important for its transcriptional<br />

activation and high pThr69/pThr71-ATF2 has been correlated with<br />

a well-differentiated phenotype in breast cancer (BC). In this study<br />

the protein expression of ATF2 & pThr71-ATF2 (pATF2) and its<br />

association with clinico-pathological features and outcome in a well<br />

defined BC series were studied.<br />

Methods<br />

The expression of ATF2 and pATF2 was assessed in the Nottingham<br />

Tenovus Primary <strong>Breast</strong> <strong>Cancer</strong> Series which consists of 1650 cases<br />

of primary invasive BC. ATF2 and pATF2 were correlated with<br />

clinico-pathological data as well as outcome in those high risk cases<br />

(NPI>3.4) based on exposure to tamoxifen.<br />

Results<br />

A total of 1351 tumours were suitable for analysis of both ATF2<br />

and pATF2; 436/1351 (32%) of BC co-expressed ATF2 and pATF2<br />

while 414/1351(31%) were negative for both ATF2 and pThr71-<br />

ATF2. Co-expression ATF2 and pATF2 was highly significantly<br />

associated luminal breast cancer phenotype, well differentiation,<br />

low proliferation (low mitotic index, low Ki67 and low SPAG5;<br />

ps


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 3<br />

discovery cohort, the signature showed positive predictive value<br />

(PPV; i.e., cumulative relapse rate of patients predicted to relapse<br />

in 3 years) of 95.4% and negative predictive values (NPV; i.e.,<br />

relapse-free survival of patients predicted not to relapse in 3 years)<br />

of 100%. In the validation cohort, the classifier predicted correctly<br />

with PPV of 75.0% and NPV of 76.9% at 3 years. Compared with<br />

patients predicted not to relapse, those predicted to relapse had a<br />

hazard ratio of 3.37 (95% CI, 1.15-9.85) for disease recurrence or<br />

death in 3 years. In an extended validation cohort of 269 patients, our<br />

signature discriminated chemoresistant TNBC in overall cohort (PPV,<br />

52.4%; NPV, 77.7%; log rank P


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

was significantly associated with higher pCR rate(19.5 vs 73.2%;<br />

p=0.026; in patients with p53- and p53+ breast cancer). In a<br />

multivariate regression analysis, after adjusting for tumor and node<br />

status, treatment administered, p53 status maintained an independent<br />

predictive role for pCR.[relative risk(RR)=6.247, 95% confidence<br />

interval (CI)=0.9-39.1] In our homogeneous patient population, pCR<br />

was not associated with initial clinical stage, nodal status as well<br />

as other clinicopathologic factors. The status of the p53, which is<br />

mutated in a high percentage of breast tumors, is a predictive factor<br />

for cytotoxic drug activity. By contrast, bcl-2 failed to be related to<br />

chemotherapy activity.<br />

P3-06-17<br />

Withdrawn by Author<br />

P3-06-18<br />

Increase of serum androgen and its metabolites in postmenopausal<br />

primary breast cancer patients with disease progression during<br />

neo-adjuvant exemestane treatment; JFMC 34-0601 TR<br />

Takada M, Saji S, Honma N, Masuda N, Yamamoto Y, Kuroi K,<br />

Yamashita H, Ohno S, Aogi K, Ueno T, Toi M. Graduate School<br />

of Medicine, Kyoto University, Kyoto, Japan; Tokyo Metropolitan<br />

Institute of Gerontology, Tokyo, Japan; Osaka National Hospital,<br />

Osaka, Japan; Kumamoto University Hospital, Kumamoto, Japan;<br />

Tokyo Metropolitan Canwithdrcer and Infectious Diseases Center,<br />

Komagome Hospital, Tokyo, Japan; Hokkaido University, Sapporo,<br />

Japan; National Hospital Organization Kyushu <strong>Cancer</strong> Center,<br />

Fukuoka, Japan; National Hospital Organization Shikoku <strong>Cancer</strong><br />

Center, Ehime, Japan<br />

BACKGROUND: We reported a positive correlation between body<br />

mass index (BMI) and clinical response to neo-adjuvant hormonal<br />

therapy (NAH) with steroidal aromatase inhibitor, exemestane, in<br />

post-menopausal breast cancer patients (Takada M et al. <strong>Breast</strong> 2012).<br />

Here, we examined the relationship between serum concentrations<br />

of sex steroids and BMI, and explored their predictive value for<br />

clinical response.<br />

PATIENTS AND METHODS: Among the 116 post-menopausal<br />

patients enrolled in the JFMC 34-0601 clinical trial of 24 weeks (wks)<br />

of NAH with exemestane, serums from 60 patients at pre-treatment,<br />

at 4wks and end of the treatment (24wks) were subjected to this<br />

study. Estradiol (E2), estrone (E1), dehydroepiandrosterone (DHEA),<br />

androstenedione (A-dione), 5-androstene-3β, 17β-diol (Aenediol),<br />

5α-androstane-3β, 17β-diol (Aanediol) were measured using LC-MS/<br />

MS analysis and E1- sulfate (E1S) was by RIA.<br />

RESULTS: There were no significant correlations between pretreatment<br />

concentrations of sex steroids and BMI, except for moderate<br />

correlation of E2 and BMI (p=0.004). In multivariate analysis, E1 was<br />

an independent predictive factor for objective response [odds 6.0, 95%<br />

confidence interval (CI) 1.5 — 34.6, p=0.011], as well as BMI. All of<br />

the estrogens decreased to under-detection levels (0.5 for E1 and E2,<br />

5 pg/assay for E1S) at 4 wks of treatment, and maintained through to<br />

the end of treatment in almost all patients independently of clinical<br />

response. The geometric mean percentage changes in androgens after<br />

NAH were: DHEA -0.2% (95%CI -15.3% — +17.6%) for patients<br />

without progressive disease (non-PD) and +44.8% (95%CI +22.1%<br />

— +71.8%) for patients with progressive disease (PD); A-dione<br />

-2.3% (95%CI -17.3% — +15.4%) for non-PD and +45.6% (95%CI<br />

+28.3% — +65.3%) for PD; Aenediol -11.5% (95%CI -20.6% —<br />

-1.4%) for non-PD and +24.9% (95%CI +1.9% — +53.0%) for PD;<br />

Aanediol +21.3% (95%CI -5.5% — +55.8%) for non-PD and +56.3%<br />

(95%CI +5.3% — +132.0%) for PD, respectively. The changes in the<br />

concentrations of both DHEA and A-dione in patients with PD were<br />

statistically significant (p=0.008 and p=0.002, respectively). In all<br />

of the PD patients, the serum concentrations of DHEA and A-dione<br />

were increased after NAH.<br />

CONCLUSION: Pretreatment serum concentration of E1 was an<br />

independent predictive factor for clinical response to NAH with<br />

exemestane. Measurement of dynamics of the serum androgen<br />

concentrations might be helpful for monitoring treatment response,<br />

and mechanism of increase of androgens has a value for further<br />

investigation. Our results should be validated using a larger dataset.<br />

(UMIN ID; C000000345)<br />

P3-06-19<br />

Ki-67 mRNA as a predictor for response to neoadjuvant<br />

chemotherapy in primary breast cancer<br />

Aigner J, Schneeweiss A, Marme F, Eidt S, Altevogt P, Sinn P, Wirtz R.<br />

National Center for Tumor Diseases, University-Hospital Heidelberg,<br />

Germany; Institut of Pathology at the St.-Elisabeth-Hospital,<br />

Cologne, Germany; German <strong>Cancer</strong> Research Center, Heidelberg,<br />

Germany; University of Heidelberg, Germany; STRATIFYER<br />

MolecularPathology GmbH, Cologne, Germany<br />

Background<br />

Stratification of molecular subtypes is of outmost importance for<br />

clinical decision making according to the 12th St Gallen Consensus<br />

conference (1). The molecular subtypes were originally identified<br />

by genome-wide mRNA analysis of fresh tissue specimen (2).<br />

Conventional immunehistochemistry assessment using ER, PR ,<br />

HER2 and Ki67 approximate these subtypes with ∼ 80% concordance<br />

(3). Here we have tested a predefined Single-Sample-Predictor for<br />

molecular subtyping based on mRNA assessment of ESR1, PGR,<br />

HER2, Ki67 and RACGAP1 in a prospective-retrospective study<br />

design.<br />

Materials and Methods<br />

Pretreatment core cut biopsies from n=100 patients with PBC treated<br />

within a randomized phase II trial (1) of anthracylin/taxane based<br />

NAC were examined. Immunohistochemistry was performed for the<br />

Ki67 antigen on an automated IHC platform (Dako Techmate 500).<br />

Ki-67 were assessed either by visual scoring (vIHC) or by quantitative<br />

image analysis (qIHC). RNA from formalin-fixed, paraffin embedded<br />

(FFPE) routine biopsies was extracted using a bead-based extraction<br />

method (STRATIFYER XTRAKT kits). Fresh tissue samokes were<br />

prepared by using Qiagen kits. ESR1, PGR, HER2, Ki-67 and<br />

RACGAP1 as well as CALM2 as a house keeping gene were measured<br />

via a multiplex quantitative RT-PCR (qPCR). Stratification into<br />

molecular subtypes was done by stepwise analysis of singular genes<br />

using predefined cut-off values. Kaplan-Meier survival estimates,<br />

patitioning test and correlation analyses were performed using the<br />

SAS JMP® 9.0.0 software.<br />

Results<br />

Concordance between RNA based and IHC-based subtyping into<br />

Luminal A, Luminal B, HER2 positive and Triple Negative breast<br />

cancer by using ESR1, PGR, HER2, Ki-67 and RACGAP1 was at<br />

85%. mRNA based analysis of fixed tumor specimen performed<br />

equally well as fresh tissue analysis. The rate of pathological complete<br />

Remission for the predefined molecular subtypes has been for Luminal<br />

A 0%, Luminal B 19%, HER2 positive 21% (without Trastuzumab)<br />

und Triple Negative Tumors 22%. qPCR from fresh tissue specimen<br />

resulted in almost identical classifications. The 5-year overall survival<br />

rate was significantly different between subtypes as expected: Luminal<br />

A 100%, Luminal B 85%, HER2 positiv 80% und triple negativ 60%.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

314s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 3<br />

Luminal tumors predominantly metastasized into the bones. <strong>Cancer</strong><br />

related deaths within the first three years were restricted tot he Triple<br />

negative Tumortype.<br />

Conclusion<br />

The Single-Sample-Predictor based on assessment of only five<br />

mRNA Markers (ESR1, PGR, HER2, Ki-67 and RACGAP1) reliably<br />

stratified into molecular subtypes by using either fixed or fresh<br />

pretreatment core needle biopsies. The mRNA based assessment<br />

enables objective and highly reproducible subtyping for clinical<br />

routine diagnostics.<br />

1) Goldhirsch et al., Annals of Oncology 2011<br />

2) Perou et al., Nature 2000<br />

3) Sotiriou&Pusztai, NEJM 2009<br />

4) Schneeweiss et al, Ann Oncol 2011<br />

P3-06-20<br />

Is it possible to predict the efficacy of a combination of<br />

Panitumumab plus FEC 100 followed by docetaxel (T) for patients<br />

with triple negative breast cancer (TNBC)? Final biomarker<br />

results from a phase II neoadjuvant trial.<br />

Nabholtz J-M, Dauplat M-M, Abrial C, Weber B, Mouret-Reynier<br />

M-A, Gligorov J, Tredan O, Vanlemmens L, Petit T, Guiu S, Jouannaud<br />

C, Tubiana-Mathieu N, Kwiatkowski F, Cayre A, Uhrhammer N,<br />

Privat M, Desrichard A, Chollet P, Chalabi N, Penault-Llorca F.<br />

Centre Jean Perrin, Clermont-Ferrand, France; Centre Alexis<br />

Vautrin, Vandoeuvre les Nancy, France; Hôpital Tenon, Paris, France;<br />

Centre Leon Berard, Lyon, France; Centre Oscar Lambret, Lille,<br />

France; Centre Paul Strauss, Strasbourg, France; Centre Georges<br />

François Leclerc, Dijon, France; Institut Jean Godinot, Reims,<br />

France; CHU Dupuytren, Limoges, France<br />

Background : TNBC is an heterogeneous group of tumors for some<br />

of which the Epithelial Growth Factor Receptor pathway (EGFR)<br />

may play an important role. We evaluated the efficacy and toxicity<br />

of an anti-EGFR antibody (panitumumab) combined with a standard<br />

neoadjuvant chemotherapy in order to identify predictive biomarkers<br />

of efficacy and target biologically defined subpopulations for potential<br />

further development.<br />

Methods : Sixty patients with stage II-IIIA disease were prospectively<br />

included in this multicentre neoadjuvant study. Systemic therapy (ST)<br />

consisted of panitumumab (9 mg/kg q.3 weeks x8) combined with<br />

FEC 100 (500/100/500 mg/m 2 q.3 weeks x4) followed by 4 cycles<br />

of T (100 mg/m 2 q.3weeks x4). All patients underwent surgery at<br />

completion of ST.<br />

Paraffin-embedded and frozen samples were systematically collected<br />

before and after ST for biologic studies.<br />

Patients characteristics are as follows : mean age 50 [27-72] ; median<br />

tumor size : 40 mm [20-120] ; invasive ductal carcinoma : 96.7% ;<br />

Scarff-Bloom-Richardson Grade III : 71.7%, grade II : 28.3%.<br />

Complete pathological response (pCR) rate was 52.3% [95% IC :<br />

37.3-67.5] (Sataloff’s classification) and 46.7% [95% IC : 31.6-61.4]<br />

(Chevallier’s classification). Conservative surgery was performed in<br />

88% of cases.<br />

Skin toxicity was the main side-effect : Cutaneous toxicity grade IV<br />

: 5%, grade III : 30%, grade II : 20%. Neutropenia grade IV : 27% ;<br />

febrile neutropenia : 5%. Infection : 0%. Hand-foot syndrome grade<br />

III : 3.3%. Ungueal toxicity grade III : 1.6%, grade II : 20%.<br />

Results :<br />

We performed a ROC curve to identify the best cut-off value for<br />

KI-67, EGFR, cytokeratin 5-6 and p53 in order to predict a pCR.<br />

Tumors with more than 40% of positive cells for KI-67 and tumors<br />

with a score for EGFR greater than 70 tend to be associated with<br />

pCR according to Chevallier’s classification (p=0.06). No predictive<br />

value was identified for Cytokeratin 5-6 and p53 (p=0.61 and p=0.27,<br />

respectively).<br />

Immunohistochemistry results show that two thirds of tumors have<br />

more than 40% of positive cells for KI-67 and that two thirds of<br />

tumors present a score for EGFR greater than 70.<br />

About half of the tumors express cytokeratin 5-6 and p53 (cut off: 1%).<br />

Chi-squared tests were performed to assess relations between<br />

cutaneous toxicities and pCR.<br />

The cutaneous toxicities were not predictive of pCR (p=0.94) and no<br />

correlations were found with KI-67, EGFR, Cytokeratin 5-6 and p53.<br />

In terms of BRCA1 and BRCA2 status, 35 tumors were analysed so<br />

far: BRCA1: 6 mutations (17%); BRCA2 (30 patients): 1 mutation<br />

(3.3%).<br />

Conclusions : These results suggest the possibility to identify a<br />

subpopulation with high probability of pCR (KI-67 > 40%, EGFR<br />

score > 70).<br />

Further biological studies are ongoing and will be presented at<br />

the meeting, including EGFR polymorphisms, C-met, ALDH1,<br />

pCadherine and PTEN.<br />

This will help us further define subpopulations of TNBC patients,<br />

potential targets for antiEGFR development.<br />

P3-06-21<br />

Unique Molecular Subtypes of Triple Negative <strong>Breast</strong> Carcinomas<br />

by Routine IHC: Implications for Treatment and Prognosis<br />

Ashfaq R, Wright B, Russell K, Voss A. Caris Life Sciences, Phoenix,<br />

AZ<br />

Background: Triple negative breast cancer (TNBC) is defined by<br />

lack of expression for Estrogen Receptor, Progesterone Receptor<br />

and Her2. Recent gene expression profiling has shown that triple<br />

negative tumors are a heterogeneous group of tumors with variable<br />

prognosis. The objective of this study is to identify unique subsets<br />

of triple negative lesions based on routine IHC.<br />

Method: Previous breast cancer cases received for molecular testing<br />

by Caris Target Now (Caris Life Sciences, Phoenix, AZ) undergo<br />

standard testing for ER, PR, Her2 by IHC and Her2 FISH based<br />

on which molecular subtype is assigned (ER/PR+, Her2+ Triple<br />

Negative). Triple Negative breast carcinomas were further assessed<br />

by IHC for additional markers including CK 5/6, CK7, CK14, P53,<br />

AR, PTEN, Ki-67 and Cav-1 to help identify unique molecular<br />

subsets of TNBC.<br />

Results: Based on IHC profiles for 215 TNBC, 42% can be<br />

categorized as Basal subtype based on positivity for CK5/6.CK14<br />

and CK17, 40% are Androgen Receptor positive, 55% have loss of<br />

Caveolin , 7% of TNBC have P53 alterations. Additionally 46% of<br />

TNBC have loss of PTEN. The highest loss of PTEN occurs in the<br />

Stromal subtype (77%). PTEN loss is rarely seen in p53 positive cases.<br />

The large majority of TNBC are proliferative (90%) as assessed by<br />

Ki-67 index.<br />

Conclusions: Routine IHC markers define at least four unique<br />

subgroups of TNBC; Basal subtype, P53- Positive subtype, AR<br />

Positive subtype , Stromal subtype (Cav-1-negative), Proliferative<br />

Subtype. This study shows that with routine IHC it is possible to<br />

identify TNBC with more favorable prognosis such as AR-Positive<br />

subtype and Low proliferation TNBC versus those TNBC with poor<br />

prognosis, such as P53 and Stromal subtypes. In addition to their<br />

unique prognostic profiles, these subtypes may require different<br />

therapeutic interventions.<br />

www.aacrjournals.org 315s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P3-06-22<br />

Mechanisms behind trastuzumab resistance as neoadjuvant<br />

therapy in HER2-positive operable breast cancer<br />

Jinno H, Sato T, Hayashida T, Takahashi M, Hirose S, Kitagawa Y.<br />

Keio University School of Medicine, Shinjuku, Tokyo, Japan<br />

Background<br />

The development of trastuzumab has had a major impact in treatment<br />

of breast cancer patients with HER2 overexpression. Despite<br />

clinical usefulness, intrinsic or acquired resistance to trastuzumab<br />

is a common clinical phenomenon. However, the mechanisms of<br />

resistance to trastuzumab have not been fully elucidated. Potential<br />

mechanisms include aberrant downstream signaling caused by loss of<br />

PTEN and/or PIK3CA mutation, alternative signaling from IGF-1R<br />

and masking with MUC4. The objective of this study was to determine<br />

the possible mechanisms of resistance to trastuzumab as neoadjuvant<br />

therapy in women with HER2-overexpressing operable breast cancer.<br />

Patients & methods<br />

Patients with operable breast cancer received 12 cycles of weekly<br />

paclitaxel (80 mg/m2 IV) plus weekly trastuzumab (4mg/kg loading<br />

dose followed by 2 mg/kg IV) for 12 weeks before surgery. All<br />

tumors were HER2-positive by immunohistochemistry (IHC)<br />

or fluorescence in situ hybridization. Expressions of ER, PgR,<br />

Ki67, PTEN, phosphorylated IGF-1R (pIGF-1R) and MUC4 were<br />

performed by IHC in core needle biopsy samples at baseline. ER<br />

and PgR status was assessed using Allred score. For Ki67 labeling<br />

index, a total of 400 cells were counted from three consecutive highpower<br />

magnifications. PTEN, pIGF-1R and MUC4 expression level<br />

was scored semiquantitatively based on staining intensity. PIK3CA<br />

mutation status was evaluated by sequencing of PIK3CA exons 9 and<br />

20 using PCR amplification and direct sequencing. pCR was defined<br />

as no residual invasive carcinoma in the breast.<br />

Results<br />

Thirty-seven patients were enrolled and assessable for clinical and<br />

pathologic responses. ER and PgR were positive in 18 (48.6%) and 16<br />

(43.2%) patients, respectively. The overall response rate was 86.5%,<br />

including a complete response in 21 patients and a partial response in<br />

11 patients. The pCR rate was 48.6% (18/37). Negative, moderate and<br />

strong membranous expression of MUC4 was observed in 3 (8.1%),<br />

18 (48.6%) and 12 (32.4%) patients, respectively. Membranous<br />

staining for pIGF1-R was negative in 24 (64.9%) patients. PTEN<br />

loss was observed in 33.3% (8/24) of the tumor examined. PIK3CA<br />

sequence analysis of the 13 tumors identified 2 mutations in exon<br />

20 and 2 mutations in exon 9, corresponding to a PIK3CA mutation<br />

frequency of 30.8%. MUC4 and pIGF1-R status did not affect the<br />

pCR rate. PTEN loss and/or PIK3CA mutation were not significantly<br />

associated with pCR rate. pCR rate was significantly correlated with<br />

PgR negativity (p=0.004) and higher Ki67 (p=0.01).<br />

Conclusions<br />

These data indicate that aberrant downstream signaling caused by<br />

loss of PTEN and/or PIK3CA mutation, alternative signaling from<br />

IGF-1R and masking with MUC4 were not important mechanisms<br />

of resistance to trastuzumab.<br />

P3-06-23<br />

Predicting response to neoadjuvant letrozole<br />

Larionov A, Turnbull A, Sims A, Renshaw L, Kay C, Harrison D, Dixon<br />

JM. University of Edinburgh, Scotland, United Kingdom<br />

Background<br />

Approximately 75% of older postmenopausal patients with large<br />

operable or locally advanced oestrogen receptor positive breast<br />

cancer respond to treatment with neoadjuvant letrozole. Prediction of<br />

response is required as approximately 25% fail to respond. The aim<br />

of this study was to design a response predicting classifier with two<br />

data sets, one published previously [1] and secondly a new dataset.<br />

Methods<br />

Patients were treated with Letrozole (Femara, 2.5 mg daily) and<br />

tissues were collected before treatment, 2 or 3 weeks on treatment,<br />

and between 3 and 5 months on treatment. Response was based on<br />

a 3D ultrasound, with response being classified as a 50% reduction<br />

in tumour volume at 3 months. To ensure biological relevance of<br />

the response groups we also separated patients into Quick Stable<br />

Responders (QSR) and Long-Term Non-responders (LNR), as<br />

summarised in Table 1.<br />

Datasets summary<br />

Dataset publication<br />

Complete triplets<br />

(Total patients)<br />

Assessable clinical responses Extreme response groups<br />

(Resp vs Non-resp)* (QSR vs LNR)*<br />

JCO-2009 [1] 57 (62) 52 (37 vs 15) 23 (16 vs 7)<br />

SABCS-2012 51 (76) 41 (31 vs 10) 24 (15 vs 9)<br />

*Responses numbers are shown for triplets only<br />

Illumina HT-12 BeadArrays were used for gene expression profiling,<br />

allowing platform-independent cross-validation with our previous<br />

study using Affymetrix HGU-133 chips [1]. The probe IDs were reannotated<br />

to Ensemble gene IDs and XPN batch correction has been<br />

applied to make the datasets directly comparable. A combination of<br />

Rank-Product, SAM and Random Forrest analyses have been used<br />

to design several alternative signatures separating extreme response<br />

groups, the patients who responded rapidly vs those who did not<br />

respond for a long time. One of the best performing signatures<br />

included 48 informative features: 24 at baseline, 12 at 2-weeks and<br />

12 changes between 0 and 2 weeks. This signature was validated on<br />

the independent previously published test set by classifying extreme<br />

responses (quick stable responders vs long term non responders)<br />

using Support vector machines (SVM) with different kernel functions.<br />

Results<br />

Are summarised in Table 2.<br />

Response prediction<br />

SVM kernel function<br />

Baseline genes 2-week genes Changed genes<br />

All features combined (48)<br />

alone (24) alone (12) alone (12)<br />

Radial 18/23 (78%)* 18/23 (78%) 19/23 (83%) 21/23 (91%)<br />

Linear 16/23 (70%) 13/25 (57%) 20/23 (87%) 21/23 (91%)<br />

*Number of correct classifications/ Total number of cases (% of correct classifications)<br />

Conclusions<br />

- A new sample series has been collected to study neo-adjuvant<br />

endocrine response. It can be used for independent validation of<br />

previous results [1] and for design of new platform-independent<br />

response predictors.<br />

- Multi-gene predictors based upon change in gene expression<br />

outperform those from baseline or 14 day measurements alone. The<br />

best classifiers include all 3 time points.<br />

- A classifier has been derived with a 91% accuracy rate of predicting<br />

extreme response group in 21 of 23 patients.<br />

References<br />

[1] Miller 2009 J Clin Oncol 27:1382.<br />

P3-06-24<br />

Early activation of IFN/STAT signaling in tumor cells of patientderived<br />

triple negative breast cancer xenografts predicts tumor<br />

sensitivity to chemotherapy<br />

Legrier M-E, Yvonnet V, Beurdeley A, Stephant G, Le Ven E, Banis<br />

S, Lassalle M, Deas O, Cairo S, Judde J-G. Xentech, Evry, Ile de<br />

France, France<br />

Triple negative breast cancer (TNBC) is a tumor subtype characterized<br />

by the absence of overexpressed estrogen receptor-alpha (ER),<br />

progesterone receptor (PR), and HER2 receptor, encoded by<br />

ERBB2, a known proto-oncogene. This type of tumors account for<br />

approximately 15-25% of breast cancers at diagnosis, and is one of<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

316s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 3<br />

the most aggressive subtypes, with 77% of patients that live free of<br />

disease 5 years post-diagnosis. One of the most reliable predictive<br />

markers of patient outcome is the pathological complete response<br />

(pCR), which indicates that the surgical specimen removed after<br />

neoadjuvant chemotherapy contains no viable tumor cells detectable<br />

at histopathological level. For patients with pCR, the probability of<br />

surviving the disease is very high, however, pCR is observed only<br />

in about 20-30% of TNBC. On the other hand, for patients with no<br />

pCR the probablity of developing recurrent disease at 5 years is 50%.<br />

As pCR is strongly correlated with treatment efficacy, it is mandatory<br />

to develop methods that allow to tell as quick as possible if the<br />

treatment chosen for a given patient is efficiently working or if it<br />

should be abandoned in favor of an alternative strategy that could<br />

prove more efficacious. XenTech collection of breast cancer patientderived<br />

xenografts (PDXs) includes 25 models of TNBC that display<br />

heterogeneous response to different chemotherapy agents. We used<br />

our models to investigate if transcriptional changes could be detected<br />

in PDXs that responded well to genotoxic agents. To do this we<br />

analyzed the gene expression profile of laser-microdissected residual<br />

tumor nodules interspersed in the murine stroma upon very efficient<br />

response to Adriamycin/Cyclophosphamide (AC). When doing so,<br />

we identified several genes of the IFN/STAT1 pathway that were<br />

over-expressed when compared to untreated tumors. This activation<br />

seems to be a transient event, as it was lost in tumors relapsing after<br />

the residual tumor nodule stage.<br />

The finding that residual cells from tumors strongly responding to<br />

AC treatment over-expressed IFN/STAT1 pathway-related genes<br />

prompted us to investigate whether this effect could be detected as<br />

an early event upon tumor exposure to chemotherapy. All TNBC<br />

models tested that were good responders to AC treatment displayed<br />

over-expression of IFN/STAT1 pathway-related genes as early as 3<br />

days post-treatment, most of them reaching a plateau of intensity<br />

at day 7 post-treatment. By contrast, TNBC insensitive or low<br />

responders to AC treatment failed to show over-expression of IFN/<br />

STAT1 pathway-related genes.<br />

To verify if the selective over-expression of these genes in<br />

TNBC models sensitive to AC was independent of the treatment<br />

administered, Irinotecan and Capecitabine were used to treat TNBC<br />

models with heterogeneous response to these drugs. Again, we<br />

found that overexpression of IFN/STAT1 pathway-related genes<br />

was specifically identified at early stages only in TNBC models that<br />

responded well to these drugs.<br />

These results suggest that genes of the IFN/STAT1 pathway could be<br />

early predictors of tumor response in patients receiving neoadjuvant<br />

chemotherapy. Prospective clinical validation studies are warranted<br />

to confirm the findings from this preclinical study.<br />

P3-06-25<br />

PTEN mRNA positivity using in situ measurements is associated<br />

with better outcome in Tamoxifen treated breast cancer patients.<br />

Schalper KA, Li K, Rimm DL. Yale School of Medicine, New Haven,<br />

CT<br />

Background: Aberrant phosphatidylinositol 3-kinase (PI3K) pathway<br />

signaling occurs in a high proportion of human breast cancers and<br />

is involved in carcinogenesis, tumor progression and resistance to<br />

targeted therapies. Reduced levels of the tumor suppressor PTEN<br />

has been associated with PI3K pathway activation and is observed<br />

in ∼40% of breast carcinomas. PTEN levels have traditionally been<br />

determined in breast tumors by qualitative immunohistochemistry<br />

(IHC) and its prognostic and/or predictive value has not been<br />

clearly established. We used a novel method of quantitative in situ<br />

measurements of PTEN mRNA and protein to determine their possible<br />

prognostic and predictive value in breast cancer.<br />

Materials and methods: PTEN mRNA and protein were measured<br />

on formalin fixed paraffin embedded (FFPE) cell lines and tumor<br />

samples on tissue microarrays (TMAs) representing a retrospective<br />

cohort of 404 breast cancer cases collected from the Yale Pathology<br />

archives between 1975 and 2005. The RNAscope paired-primer<br />

method was used for PTEN mRNA in situ hybridization (ISH), with<br />

Ubiquitin C and DapB ISH probes used as positive and negative<br />

controls for each run. PTEN protein measurements were performed<br />

with monoclonal antibody 138G6 (Cell Signaling Technology).<br />

Both mRNA and protein were measured using the AQUA method of<br />

quantitative immunofluorescence.<br />

Results: Both PTEN mRNA ISH signal and protein were highly<br />

reproducible in FFPE tissues and cell lines. PTEN mRNA levels<br />

were lower in cell lines with known PTEN downregulation. In breast<br />

carcinoma samples, PTEN mRNA scores showed a positive non-linear<br />

association with protein levels. Neither PTEN mRNA nor protein<br />

showed prognostic value. However, high PTEN mRNA expression<br />

using a visually defined cutoff was associated with better outcome<br />

in a subset of Tamoxifen treated breast cancer patients.<br />

Conclusion: Initial data suggests that PTEN mRNA expression may<br />

be predictive of favorable response to endocrine therapy in breast<br />

carcinomas. However, validation in larger independent cohorts is<br />

required.<br />

P3-06-26<br />

Serum anti-p53 antibody titers predict pathological response to<br />

preoperative chemotherapy in women with HER2 positive or<br />

triple negative breast cancer.<br />

Yoshiyama T, Nakamura Y, Igawa A, Saruwatari A, Shigechi T, Ueda<br />

N, Kuba S, Ishida M, Nishimura S, Nishiyama K, Ohno S. National<br />

Kyushu <strong>Cancer</strong> Center, Fukuoka, Japan<br />

Background: In SABCS 2009, we reported that the high level<br />

of serum anti-p53 antibody titers was associated with response to<br />

preoperative chemotherapy in cases with primary breast cancer<br />

(SABCS 2009). Prognostic meaning of pathological complete<br />

response (pCR) after preoperative chemotherapy differs among<br />

various subtype, and there is a close relationship between pCR and<br />

prognosis in cases with HER2 positive or triple negative disease. The<br />

aim of this study is to evaluate the association between the high level<br />

of serum anti-p53 antibody titers and pCR in patients with HER2-<br />

positive or triple negative breast cancer.<br />

Patients and Methods: In this study, we analyzed 196 women<br />

with operable early stage breast cancer (T1-T3, N0-1) treated with<br />

preoperative chemotherapy and definitive curative surgery since 2002<br />

to 2011. All of the patients received four cycles of FEC (fluorouracil<br />

500 mg/m2, epirubicin 100 mg/m2, cyclophosphamide 500 mg/<br />

m2 q3w) followed by four cycles of docetaxel (75 mg/m2 q3w).<br />

The serum anti-p53 antibody titers were assessed by ELISA with<br />

the anti-p53 EIA Kit II (MESACUP anti-53 Test: MBL) in the pretreatment<br />

serum samples. The test’s cut-off value was determined<br />

to be 1.3 IU/ml based on reference distribution in healthy control<br />

individuals. The association between serum anti-p53 antibody titers<br />

and pCR was analyzed.<br />

Results: The subtype of tumors of 196 patients were classified to<br />

86 of ER+/HER2-, 24 of ER+/HER2+, 29 of ER-/HER2+ and 57 of<br />

ER-/HER2-. The pCR was achieved 49 of 196 patients (25%). The<br />

pCR rate were 7%(6/86), 33%(8/24), 48%(14/29) and 37%(21/57)<br />

in the subtype of ER+/HER2-, ER+/HER2+, ER-/HER2+ and ER-/<br />

www.aacrjournals.org 317s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

HER2- respectively. The range of serum anti-p53 antibody titers in<br />

the serum samples of 196 patients was between 0.40 and 5610 ( the<br />

median was 0.40 and the average was 85.2). Excluding 86 cases with<br />

ER+/HER2-, relationship between serum anti-p53 antibody titers<br />

and pCR was evaluated. According to serum anti-p53 antibody titers<br />

with the patients of pCR, cut-off value of serum anti-p53 antibody<br />

titers was set to be 6 IU/ml. The pCR was observed in nine of 12<br />

cases (75%) with high serum anti-p53 antibody titers, which was<br />

significantly higher than those with low titers (35%: 34/98 ; p=0.01).<br />

Multivariate analysis showed that the only high anti-p53 antibody titer<br />

was an independent predictive factor for pCR due to preoperative<br />

chemotherapy (p=0.01).<br />

Conclusion: p53 mutation analysis using serum anti-p53 antibody<br />

titers might be a useful predictive test for response to preoperative<br />

chemotherapy in patients especially with HER2 positive or triple<br />

negative breast cancer.<br />

P3-06-27<br />

Dynamic tomographic optical breast imaging (TOBI) to monitor<br />

response to neoadjuvant therapy in breast cancer<br />

Carp SA, Wanyo CM, Specht M, Schapira L, Moy B, Finkelstein DM,<br />

Boas DA, Isakoff SJ. Massachusetts General Hospital, Charlestown,<br />

MA; Massachusetts General Hospital, Boston, MA<br />

Background: Near-infrared optical measurements have been<br />

recently shown to offer a promising non-invasive way for monitoring<br />

breast neoadjuvant chemotherapy (NAC) and predicting outcome.<br />

In particular, snapshots of tissue oxy and deoxy-hemoglobin<br />

concentration as well as water and lipid content have been<br />

demonstrated to be sensitive to therapy-induced changes. In this study,<br />

we extend optical measurements to capture additional hemodynamic<br />

and metabolic biomarkers revealed by dynamically imaging breast<br />

tissue during fractional mammographic compression. Using our<br />

dynamic tomographic optical breast imaging (TOBI) system we<br />

evaluate the early prediction performance of this advanced technology.<br />

Methods: We are conducting a pilot feasibility study in female<br />

patients with unilateral locally advanced breast cancer undergoing<br />

standard-of-care NAC. Pre-treatment and day 7 post-treatment TOBI<br />

scans are obtained, with additional (optional) scans on day 1 of each<br />

subsequent chemotherapy cycle. Both breasts are compressed in<br />

turn to 4-8 lbs of force, and optical images are acquired once every<br />

2 seconds over two minutes. Time-resolved oxy-(HbO), deoxy-<br />

(HbR), and total-(HbT) hemoglobin concentration and hemoglobin<br />

oxygen saturation (SO 2 ) are calculated. The compression-induced<br />

rate of change of HbT correlates with changes in tissue blood volume<br />

indicative of biomechanical properties. The evolution of tissue SO 2<br />

is modeled to obtain an index of the ratio of oxygen metabolism to<br />

blood flow. Therapy induced changes are quantified, and comparisons<br />

between changes in responders vs. non-responders are performed<br />

(response is defined here as >50% reduction in the largest tumor<br />

diameter).<br />

Results: We have enrolled 20 patients so far, of which 90%<br />

(N=18) completed both the day 0 and day 7 scans. 17 patients have<br />

undergone surgery at this point. We focused our initial analysis on<br />

5 HER2+ patients, of which two were non-responders, and three<br />

were responders according to our criteria. Four patients received<br />

taxol+herceptin+lapatinib, while the other received taxol+lapatinib<br />

only. In this small subgroup, the non-responders had an average<br />

increase of 1% in total hemoglobin concentration (HbT) from day<br />

0 to day 7, while the responders had an average 12% decrease in<br />

HbT, respectively. We also noted different trends in the evolution of<br />

the tissue oxygen consumption to blood flow ratio, which increased<br />

32% in non-responders from day 0 to day 7, while decreasing 11%<br />

in responders.<br />

Conclusions: The large percentage of enrolled patients that completed<br />

both initial scans demonstrates the feasibility of using dynamic optical<br />

breast tomography for breast neoadjuvant chemotherapy monitoring.<br />

Results in a small cohort of 5 HER2+ patients suggested a decreasing<br />

trend in HbT for responders as observed by previous studies. We<br />

also report for the first time an increase in the metabolic ratio of<br />

oxygen consumption to blood flow in non-responders vs. a decrease<br />

in responders. These initial results of our on-going study suggest that<br />

dynamic TOBI can detect changes due to treatment and may have<br />

predictive value for the treatment outcome and supports further studies<br />

of this non-invasive and portable tool for chemotherapy monitoring.<br />

P3-06-28<br />

Use of the MiCK drug-induced apoptosis assay improves clinical<br />

outcomes in recurrent breast cancer (BRCA)<br />

Bosserman L, Rogers K, Davidson D, Whitworth P, Karimi M,<br />

Upadhyaya G, Rutledge J, Hallquist A, Perree M, Presant C.<br />

Wilshire Oncology Medical Group-US Oncology; Nashville Oncology<br />

Associates; Tennessee Plateau Oncology; Nashville <strong>Breast</strong> Center;<br />

Data Vision; DiaTech Life Sciences<br />

Background: The microculture kinetic (MiCK) assay (Correct<br />

Chemo) correlates with outcomes in acute myelocytic leukemia<br />

and ovarian cancer (<strong>Cancer</strong> Research, in press). A prior trial suggested<br />

that its use in breast cancer could improve clinical outcomes (<strong>Cancer</strong>,<br />

in press). This study was designed to correlate MiCK assay results<br />

with clinical outcomes in recurrent or metastatic BRCA.<br />

Methods: 30 patients with recurrent or metastatic breast cancer in 4<br />

different institutions were evaluated. Each patient (pt) had a BRCA<br />

biopsy sent to a central laboratory, tumor cells were purified to over<br />

90% homogeneity, and were then cultured with individual drugs or<br />

drug combinations. The induction of apoptosis was measured every<br />

five minutes continuously for 48 hours. The amount of apoptosis<br />

was expressed in kinetic units (KU), and results were sent to the<br />

attending oncologist within 72 hours of submission. Physicians were<br />

free to choose any treatment plan for the pts, and were free to add<br />

hormonal therapy or biotherapy. Clinical results were evaluated by<br />

oncologists using clinical criteria and the results of the MiCK assay<br />

were correlated with outcomes of complete (CR) or partial (PR)<br />

response, time-to-relapse (TTR), and overall survival (OS).<br />

Results: Median age was 57 years, and number of lines of prior therapy<br />

was a median of 2 (range 1-8). Median ECOG performance status was<br />

1. The total number of drugs tested in the assay was a median of 12<br />

(range 3-31). The MiCK assay was used to help select therapy in 22<br />

pts (73%). There was change between drugs originally planned before<br />

MiCK assay and drugs used after MiCK in 15 pts (50%). The best<br />

therapy from the MiCK assay was used for treatment in 16 pts (53%).<br />

In five pts (17%), a single drug was used in place of a combination.<br />

Generic drugs were used in place of proprietary drugs in nine pts<br />

(30%). Hormonal therapy was added to drugs selected based on the<br />

MiCK assay in seven pts (23%), and bio-therapy drugs were added<br />

to chemotherapy drugs in eight pts (27%). If the MiCK results were<br />

used to help select therapy, eight pts had a CR or PR (27%), compared<br />

to 0 pts with CR or PR if MiCK was not used (p=0.04). If the MiCK<br />

assay was used to determine therapy, 17 pts (59%) had a CR, PR or<br />

stable disease compared to only 2 pts (6.9%) in whom the MiCK<br />

assay was not used (p< 0.01). The TTR was significantly longer if the<br />

MiCK assay was used to select chemotherapy, 7.4 months, compared<br />

to only 2.2 months if the MiCK assay was not used (p< 0.01). There<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

318s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 3<br />

was a trend toward longer survival if the MiCK assay was used, 16.8<br />

months, compared to 13.1 months if the MiCK assay was not used,<br />

but the difference was not statistically significant (p=0.3). If the best<br />

chemotherapy from the MiCK assay was used, there were trends for<br />

increased TTR (7.3 vs 3.9 mo if best not used p=0.13) and increased<br />

rate of CR or PR or stable (54% vs 17% p=0.11).<br />

Conclusions: Use of the MiCK assay to determine chemotherapy was<br />

associated with a higher response rate and a longer time to relapse<br />

in pts with recurrent or metastatic BRCA. It is possible that OS is<br />

also improved, but longer follow up is needed. There was a trend for<br />

improved outcomes if the best chemotherapy based on the MiCK<br />

assay was used.<br />

P3-06-29<br />

Change of circulating tumor cells before and after neoadjuvant<br />

chemotherapy in patients with primary breast cancer<br />

Horiguchi J, Takata D, Rokutanda N, Nagaoka R, Tokiniwa H, Tozuka<br />

K, Sato A, Kikuchi M, Oyama T, Takeyoshi I. Gunam University<br />

Hospital, Marbashi, Gunma, Japan; Gunma University Hospital,<br />

Maebashi, Gunma, Japan<br />

Background: Circulating tumor cells (CTCs) in peripheral blood<br />

may represent the possible presence of early tumor dissemination.<br />

However, relatively few studies were designed to investigate the<br />

change of CTC status by adjuvant chemotherapy in operable breast<br />

cancer patients. Patients and methods: Peripheral blood (7.5ml)<br />

was collected from 95 patients with stage II/III breast cancer before<br />

neoadjuvant chemotherapy (NAC). NAC consisted of anthracycline<br />

and paclitaxel chemotherapy and additional trastuzumab treatment<br />

for patients with HER2-positive tumors. Results: The average age<br />

of patients was 52.6 year old (median 52.0). One or more CTCs<br />

were detected in 18 (18.9%) of 95 patients. CTCs were detected<br />

in 6 (12.0%) of 50 patients with clinical stage II disease and 12<br />

(26.7%) of 45 patients with clinical stage III disease. According to<br />

tumor subtypes, CTCs were detected in 5 (17.9%) of 28 patients<br />

with hormone receptor (HR)-positive and HER2-negative tumors<br />

(L subtype), 3 (12.5%) of 24 patients with HR-positive and HER2-<br />

positive tumors (L-H subtype), 4 (22.2%) of 18 patients with HRnegative<br />

and HER2-positive tumors (H subtype), and 6 (24.0%) of 25<br />

patients with HR-negative and HER2-negative tumors (TN subtype).<br />

After NAC, 17 (94.4%) of 18 patients who were CTC-positive before<br />

chemotherapy changed into negative status. 30 (31.6%) of 95 patients<br />

had a pathologic complete response (pCR) after NAC. There was<br />

no correlation between CTC status before NAC and pathological<br />

response. At the median follow up of 27 months, distant metastasis<br />

was observed in 9 patients (9.5%). Patients with clinical stage III, TN<br />

subtype, or non-pCR had a significantly worse disease-free survival<br />

(DFS). However, CTC status before NAC was not a significant<br />

prognostic factor. Conclusion: NAC has a significant impact on CTC<br />

status irrespective of tumor subtypes. CTC status before NAC was<br />

not a significant prognostic factor in this study. The reason of which<br />

may be that most of patients showing positive for CTCs before NAC<br />

have changed into negative after NAC.<br />

P3-06-30<br />

Predictors of Pathologic Complete Response to Chemotherapy<br />

and Antiangiotherapy in <strong>Breast</strong> <strong>Cancer</strong>.<br />

Makhoul I, Griffin RJ, Dhakal I, Raj V, Hennings L, Kadlubar SA.<br />

University of Arkansas for Medical Sciences, Little Rock, AR<br />

Vascular endothelial growth factor (VEGF) is a central mediator<br />

of angiogenesis in breast cancer. We have conducted a clinical trial<br />

using bevacizumab (B) in breast cancer in the neoadjuvant setting<br />

and showed improved pathologic complete response (pCR) when B<br />

was added to chemotherapy (CT). 41% of patients (Pts) who received<br />

B+CT achieved pCR. Blood and tumor samples were collected at<br />

baseline and serially on treatment from 39 patients who participated<br />

in the study. The correlative study was approved by the IRB and<br />

consent forms were obtained at the time of enrollment. Since patients<br />

do not equally benefit from B, it has become necessary to define the<br />

profile of patients who will benefit and those who are more likely<br />

to have major side effects from the drug. This study was conducted<br />

to evaluate for the presence of specific SNPs and CNV in the germ<br />

line and a serum protein profile that might be predictive of pCR in<br />

the neoadjuvant setting in breast cancer patients receiving B and CT.<br />

Methods: To test for the presence of functional germ line SNPs and<br />

CNVs that may influence pCR, DNA was extracted from buffy coats<br />

of study participants and SNPs/CNVs were determined using Illumina<br />

technology. To test the hypothesis that individuals who achieved<br />

pCR were those with differential expression of selected proteins<br />

in their blood, custom multiplex was performed for the following<br />

serum proteins: ANG-1, ANG-2, bFGF, IL-1a, MMP-9, PDGF-BB,<br />

PECAM-1, Tie-2, VEGF, and VEGFR2.<br />

Statistical test: The association between genotype and CNV of each<br />

SNP vs. pCR status were analyzed using Cochran-Armitage Trend<br />

Test. Differences in serum protein levels between pCR and non-pCR<br />

groups were examined by Wilcoxon rank sum test. Significance<br />

of association was evaluated in two settings of α (i) α=0.05 (ii)<br />

α=0.00001.<br />

Results: Genotype and CNV vs. pCR (n=38). In genotype vs. pCR<br />

status by SNPs tests, 131618 (∼13%) SNPs with allele frequency<br />


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

q21d demonstrated a 29% overall response rate (J Clin Oncol 29:<br />

2011 (suppl; abstr 1034)). Here we present the relationship between<br />

the dynamics of serum tumor marker CA27.29, etirinotecan pegol<br />

pharmacokinetics, and tumor response.<br />

Methods: Data from 45 pts with at least two CA27.29 measurements<br />

(pre- & post-first dose), were fitted with a PK/PD model developed<br />

to correlate CA27.29 dynamics with SN38 exposure predicted from<br />

individual pt dosing history: d[CA27.29]/dt = Kin*exp(beta*t)*(1-<br />

[SN38]/(IC50+[SN38]))-Kout*[CA27.29]. Correlation of CA27.29<br />

and RECIST response was investigated; % change of CA27.29 from<br />

baseline was also correlated with progression-free survival (PFS).<br />

Results: CA27.29 vs time profiles were well described by the model,<br />

with a population mean plasma SN38 IC50 of 1.6 ng/mL. Typical<br />

minimum/maximum SN38 concentrations during Tx were 1.5/ 3<br />

ng/mL for q14d and 0.9/ 2.4 ng/mL for q21d, indicating that both<br />

schedules resulted in SN38 exposure near the IC50. The half-life of<br />

CA27.29 decline was 15 days. 41 pts had pre- and post-Tx tumor<br />

measurements for correlation of CA27.29 response with tumor size.<br />

Of the 15 pts with RECIST CR or PR, 14 (93%) exhibited at least<br />

10% declines in CA27.29 during Tx; all 5 pts with RECIST SD ≥ 6<br />

months (mths) also manifested ≥10% declines, whereas only 45%<br />

(5/11) of pts with SD < 6 mths showed ≥10% decline in CA27.29.<br />

Among the 10 pts with RECIST progressive disease (PD), 80%<br />

showed ≥10% CA27.29 elevations during Tx. The median maximum<br />

% reduction in observed CA27.29 from baseline during the course of<br />

Tx ranged from -55% to +30% and correlated with responder status<br />

as tabulated below.<br />

Median maximum % reduction of CA27.29 from baseline by RECIST response<br />

CR (N=2) PR (N=13) SD ≥ 6 mths (N=5) SD < 6 mths (N=11) PD (N=10)<br />

-55 -55 -31 -4 30<br />

To investigate whether response in CA27.29 can serve as early<br />

indicator of Tx outcome, we assessed the impact of a reduction of<br />

≥25% after 6 weeks (wks) of Tx (2 or 3 Tx cycles for q21d and q14d<br />

schedules, respectively) on PFS. Among the 26 pts who had not<br />

had tumor progression or discontinued Tx by wk 6, median PFS for<br />

pts with ≥ 25% reduction in CA27.29 (n=9) was 12 mths. Pts with<br />


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 3<br />

P3-06-33<br />

Effect of trastuzumab-based therapy on serum activin A levels<br />

in metastatic breast cancer.<br />

Zubritsky LM, Ali SM, Leitzel K, Koestler W, Fuchs E-M, Costa L,<br />

Knight R, Laadem A, Sherman ML, Lipton A. Penn State Hershey<br />

Medical Center, Hershey, PA; Lebanon VA Medical Center, Lebanon,<br />

PA; Medical University of Vienna, Austria; <strong>San</strong>ta Maria Hospital,<br />

Lisbon, Portugal; Celgene Corp., Summit, NJ; Acceleron Pharma,<br />

Cambridge, MA<br />

Background: Only half of HER2-positive metastatic breast cancer<br />

patients will respond to first-line trastuzumab-containing therapy, but<br />

of these, most will progress within a year. Trastuzumab resistance<br />

remains a continuing clinical problem, and better biomarkers and<br />

therapies are needed. Activin A is a TGF-beta superfamily member<br />

that regulates cell proliferation, apoptosis, differentiation, and immune<br />

response. We have previously reported that higher pretreatment serum<br />

activin A level predicted reduced progression-free survival (PFS) and<br />

overall survival (OS) to first-line trastuzumab, independent of age, line<br />

of therapy, CA 15-3, and hormone receptor status (ASCO Ab ID 607,<br />

2012). Methods: Serum activin A was measured using ELISA (R&D<br />

Systems, Minneapolis, MN) in 60 metastatic breast cancer patients<br />

before and 1 month after starting first-line trastuzumab-containing<br />

therapy. PFS and OS were analyzed using the Kaplan-Meier method<br />

and Cox modeling with both continuous and dichotomous (median)<br />

serum activin A analyses. Results: Pretreatment serum activin A levels<br />

had a median of 629 pg/mL and an inter-quartile range of 406 to 1791<br />

pg/mL. There was no significant change in activin A levels between<br />

pretreatment and one month after starting trastuzumab therapy.<br />

Median activin A level at one month was 655 pg/mL, with an interquartile<br />

range of 405 to 1517 pg/mL. 83% of patients who had low<br />

pretreatment activin A levels (below median) had low activin levels<br />

at one month, and 83% who had high activin levels (above median)<br />

at baseline had high activin A levels at one month. Patients who had<br />

high activin A levels at baseline and one month had the worst outcome<br />

for both PFS (HR 3.9; median 134 days vs.776 days; p Q3 25/139 22/150 0.68<br />

VEGFR-2 10.2 ng/mL ≤ median 42/291 53/296 1.27 0.0237<br />

> median 45/278 34/309 0.61<br />

Cosmetic results one year after surgery<br />

Conclusions: Unlike trials in metastatic BC (AVADO, AVEREL), in<br />

the adjuvant setting, baseline pVEGF-A concentration did not show<br />

predictive value for BEV efficacy with a median cut-off. However,<br />

analyses using the Q3 cut-off suggest a trend toward predictive<br />

value. High baseline pVEGFR-2 was associated with greater BEV<br />

treatment effect, consistent with previous results in AVADO and<br />

AVEREL. The impact of differing biology in the adjuvant setting,<br />

lower median VEGF-A concentration than in the metastatic setting<br />

(77.0 vs 127.0-129.1 pg/mL), and the possible influence of surgery<br />

immediately before treatment require further investigation. Additional<br />

exploratory analyses are ongoing to provide better understanding of<br />

the BEATRICE dataset and the complex biology of angiogenesis,<br />

including additional markers, changes over time, and combination<br />

signatures.<br />

P3-06-35<br />

Topoisomerase 1 gene copy aberrations in 52 human breast cancer<br />

cell lines and association to gene expression<br />

Stenvang J, Smid M, Nielsen S, Timmermans M, Rømer M, Nielsen<br />

D, Foekens J, Brünner N, Martens J. University of Copenhagen,<br />

Denmark; Erasmus University Medical Center/Daniel den Hoed<br />

<strong>Cancer</strong> Center, Rotterdam, Netherlands; Herlev University Hospital,<br />

Copenhagen, Denmark<br />

Background: Topoisomerase 1 (TOP1) targeting drugs, e.g.<br />

irinotecan, are included in treatment regimens for different cancer<br />

www.aacrjournals.org 321s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

types. Two clinical phase II studies 1,2 suggested a role for irinotecan<br />

in the treatment of metastatic breast cancer patients being refractory<br />

to anthracyclines and taxanes. Access to predictive biomarker<br />

for irinotecan would increase response rates and secure that only<br />

patients with a high likelihood of obtaining benefit will receive the<br />

drug. However, no predictive biomarkers for TOP1 targeting drugs<br />

are available.<br />

Experimental design: A TOP1/CEN-20 fluorescence in situ<br />

hybridization (FISH) probe mix 3 (Dako A/S, Denmark) was applied to<br />

analyze formalin fixed paraffin embedded (FFPE) tissue sections from<br />

52 human breast cancer cell lines 4 . The FISH data were correlated<br />

to the gene expression levels (Affymetrix u133a GeneChip) of ER,<br />

HER2, TOP2A and molecular subtypes. Furthermore, gene expression<br />

data from 8 TOP1 high copy and 8 TOP1 low copy cell lines were<br />

applied for Kegg pathway analysis.<br />

Results: To categorize the cell lines, we initially analyzed the TOP1<br />

copy number and CEN-20 counts in 100 normal breast epithelium<br />

sections and defined the normal diploid range as the mean plus/minus<br />

0.5 times the haploid gene copy number. Thereby the normal diploid<br />

range for TOP1 was defined as 1.7 (range 1.45-1.90) and for CEN-<br />

20 it was 1.6 (range 1.38-1.82). By applying these cut-off values we<br />

found that among the 52 breast cancer cell lines 61.5% had TOP1<br />

gene gains and 59.6% had increased CEN-20 copy numbers. However,<br />

the Pearson correlation between TOP1 and CEN-20 counts was<br />

0.54 resulting in only 11.5% of the cell lines having TOP1/CEN-20<br />

ratios > 1.5. A significant association exist between TOP1 gene copy<br />

number (FISH) and TOP1 copy numbers obtained from SNP array<br />

analysis (p


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 3<br />

Patients and Methods:<br />

Using the population-based Swedish Multi-Generation and <strong>Cancer</strong><br />

Registers we selected 8,111 women diagnosed with in situ breast<br />

cancer between 1980 and 2004. We estimated the relative risks<br />

of subsequent ipsi- and contralateral invasive breast cancer or a<br />

contalateral in situ expressed as standardized incidence ratios (SIRs),<br />

in relation to age, year, time since diagnosis and family history (1 st<br />

degree relative) for breast cancer. The relative risk of death was<br />

expressed as standardized mortality ratio, (SMR).<br />

Results:<br />

Of 8,111 women identified with first in situ, 859 had a family history<br />

for breast cancer. The overall risk of a subsequent invasive breast<br />

cancer is over four fold and the risk for contralateral in situ breasts<br />

cancer was almost seven fold, compared with the risk in healthy<br />

women.<br />

Table 1. SIR of second breast event after diagnosis of first carcinoma in situ.<br />

All No family history Family history<br />

No.<br />

of SIR (95% CI)<br />

cases<br />

No.<br />

of SIR (95% CI)<br />

cases<br />

No.<br />

of SIR (95 % CI)<br />

cases<br />

Incidence<br />

Rate<br />

Ratio*<br />

(95% CI)<br />

2nd breast<br />

1.18 (0.96,<br />

886 4.34 (4.06, 4.63) 781 4.22 (3.93, 4.53) 105 5.44 (4.45, 6.59)<br />

event<br />

1.46)<br />

2nd in situ<br />

1.08 (0.61,<br />

118 6.70 (5.55, 8.02) 104 6.62 (5.41, 8.02) 14 7.41 (4.05, 12.44)<br />

contralateral 1.91)<br />

2nd invasive<br />

(ipsi- and<br />

contra<br />

latera+<br />

missing<br />

side)<br />

2nd<br />

ipsilateral<br />

invasive<br />

768 4.03 (3.75, 4.33) 677 3.92 (3.63, 4.22) 91 5.13 (4.13, 6.30)<br />

376 1.90 (1.72, 2.10) 334 1.86 (1.6, 2.08) 42 2.27 (1.63, 3.06)<br />

2nd<br />

contralateral 303 1.53 (1.36, 1.71) 262 1.46 (1.29, 1.65) 41 2.20 (1.58, 2.98)<br />

invasive<br />

1.21 (0.97,<br />

1.51)<br />

1.01 (0.73,<br />

1.40)<br />

1.49 (1.06,<br />

2.09)<br />

∗Reference group is No family history. Incidence rate ratio has been adjusted for attend age, calendar<br />

period and year of first diagnosis of carcinoma in situ and time since first diagnosis.<br />

Women with a family history had almost a 50% increased risk for<br />

a contralateral invasive breast cancer, IRR 1.49 (95% CI 1.06-<br />

2.09) compared to women without, but had no increased risk for a<br />

contralateral in situ cancer or ipsilateral invasive breast cancer.<br />

The risk for subsequent breast cancer decreased over time after<br />

diagnosis, but still 15 years after first in situ diagnosis the risk was<br />

over three times higher compared to the general population. Women<br />

below 40 years at diagnosis had the highest risk for a subsequent<br />

breast event, SIR 8.54 (95% CI 6.07-11.67).<br />

The overall mortality for women with no second invasive event was<br />

the same as for women in the general population, SIR 1.01 (95% CI<br />

0.95-1.08). Women below 50 years at first in situ diagnosis, with a<br />

second invasive cancer, had a higher mortality compare to women<br />

above 50 years, SIR 8.3 (95% CI 5.38-11.54) and SIR 1.70 (95% CI<br />

1.39-2.06) respectively.<br />

Conclusion:<br />

The risk for a subsequent invasive breast cancer, as well as mortality<br />

was substantially higher in younger women, which should be taken<br />

into account when planning their treatment and follow-up. Family<br />

history did not increase the risk for a subsequent ipsilateral invasive<br />

cancer.<br />

P3-07-04<br />

Cigarette smoking and postmenopausal breast cancer risk: results<br />

from the NIH-AARP Diet and Health Study<br />

Nyante SJ, Gierach GL, Dallal CM, Park Y, Hollenbeck AR, Brinton<br />

LA. National <strong>Cancer</strong> Institute, Rockville, MD; AARP, Washington, DC<br />

Epidemiologic evidence regarding the relationship between smoking<br />

and breast cancer risk is inconsistent. Some studies suggest that<br />

the relationship depends on interaction with other factors, such as<br />

alcohol use, body mass index (BMI), and menopausal hormone<br />

therapy (MHT). We investigated the relationship between smoking<br />

and breast cancer risk and interactions with breast cancer risk factors<br />

in the NIH-AARP Diet and Health Study, a large prospective cohort.<br />

Postmenopausal women ages 50-71 years (N=192,076) in six US<br />

states and two metropolitan areas were followed from 1995-1996<br />

through 2006. Risk factor information was self-reported at baseline.<br />

Smoking status was based on whether participants smoked ≥ 100<br />

cigarettes in their lifetime and whether they currently smoked (current<br />

- 15%, former - 40%, never - 45%). Alcohol use was estimated from<br />

a dietary questionnaire, and categorized based on drinking ≤ 5 or ><br />

5 g/day. BMI was calculated from reported height and weight and<br />

categorized as ≥ 30 or < 30 kg/m2. MHT use was categorized as<br />

current, former, or never use of any estrogen or progestin preparation.<br />

<strong>Cancer</strong> diagnosis, estrogen receptor (ER), and progesterone receptor<br />

(PR) data were reported by state registries. After a mean 9.6 years<br />

of follow-up, 7,698 women were diagnosed with primary invasive<br />

breast cancer. Multivariable-adjusted hazard ratios (HRs) and 95%<br />

confidence intervals (CIs) were calculated using Cox proportional<br />

hazards regression. Multiplicative interactions between smoking<br />

and covariates were evaluated using the likelihood ratio test (LRT).<br />

Overall, smokers were at an increased risk of breast cancer compared<br />

to women who never smoked (current HR 1.19, 95% CI 1.10, 1.28;<br />

former HR 1.08, 95% CI 1.02, 1.13). Excess risk diminished as time<br />

since quitting increased and was close to null for women who quit<br />

smoking ≥ 10 years prior to study enrollment compared to never<br />

smokers (HR 1.04, 95% CI 0.98, 1.11). Relative risks differed<br />

significantly based on alcohol use (P-LRT < 0.01), but not BMI or<br />

MHT use (P-LRT > 0.05). The HR associated with current smoking<br />

was 1.15 (95% CI 1.05, 1.25) among women who drank ≤ 5 g/day,<br />

but was higher among women who drank > 5 g/day (HR 1.41, 95% CI<br />

1.22, 1.61). The relationship for those who drank > 5 g/day persisted<br />

after adjustment for the amount of alcohol (5-10, 10-20, 20-35, >35<br />

g/day) consumed (HR 1.36, 95% CI 1.18, 1.56). Among women who<br />

drank > 5 g/day, current smoking was associated with increased risks<br />

of hormone receptor-positive tumors (ER+/PR+ HR 1.29, 95% CI<br />

1.01, 1.64; ER+/PR- HR 2.11, 95% CI 1.27, 3.50), but not ER-/PRtumors<br />

(HR 1.07, 95% CI 0.64, 1.79). In summary, we found that<br />

smoking was associated with elevated breast cancer risk which was<br />

strongest among women who drank > 5 g of alcohol per day. Among<br />

these women, smoking-associated increases in breast cancer risk were<br />

limited to hormone receptor-positive tumors, consistent with the<br />

known relationship between alcohol use and ER+ breast cancer risk.<br />

Findings were similar after additional adjustment for the amount of<br />

alcohol consumed, suggesting that the increased risks were not due to<br />

residual confounding by alcohol dose, although further analyses are<br />

needed to fully understand the interaction between these two factors.<br />

P3-07-05<br />

Bisphosphonate use after primary breast cancer and risk of<br />

contralateral breast cancer using pharmacy data<br />

Kwan M, Habel L, Song J, Weltzien E, Chung J, Sun Y, Fletcher<br />

S, Haque R. Division of Research, Kaiser Permanente Northern<br />

California; Kaiser Permanente Southern California; Harvard<br />

Medical School<br />

Background: It is not clear if bisphosphonates (BP) are associated<br />

with improved breast cancer prognosis in women with early breast<br />

cancer. Clinical trials have reported mixed results, yet BPs may be<br />

most beneficial in patients with low estrogen levels. However, BPs<br />

and risk of second (contralateral) breast cancer has been minimally<br />

studied. We examined the association of oral BP use (ever/never<br />

www.aacrjournals.org 323s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

and duration) on risk of contralateral breast cancer (CBC) in 17,224<br />

women with early stage breast cancer treated with tamoxifen.<br />

Materials and Methods: A cohort was assembled of women<br />

diagnosed with their first primary breast cancer (Stage 0, I, II) from<br />

1996 to 2007 on tamoxifen and followed through 31 December<br />

2009 at Kaiser Permanente Northern and Southern California.<br />

Demographic, tumor, pharmacy, and cancer treatment information<br />

was extracted from electronic medical records and SEER-affiliated<br />

cancer registries at each site. Second (contralateral) tumors were<br />

identified from the cancer registries. Detailed information on oral<br />

BP use (before and after initial breast cancer diagnosis) was obtained<br />

from pharmacy databases. A record of >90 days supply was considered<br />

the minimum exposure. Initiation and duration of post-diagnosis use<br />

was categorized as 1) non-use (≤90 days supply) and use (>90 days<br />

supply) and 2) non-use (≤90 days supply), 91 days–90<br />

days supply) was associated with a modestly lower CBC risk<br />

(HR=0.80; 95% CI: 0.61, 1.16). However, stratified analyses by age at<br />

breast cancer diagnosis (


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 3<br />

patients during 2000-2009, by cancer stage (localized, regional,<br />

distant and unknown/unstaged) and race (White, Black, Other<br />

[American Indians/Alaska Natives or Asians/Pacific Islanders]).<br />

RESULTS: A total of 543,539 cases of female BC were reported to<br />

SEER during 2000-2009, with 452,047 cases among Whites (W),<br />

54,310 cases among Blacks (B), and 37,182 among other races (O).<br />

The overall incidence was highest for Whites (130.2 per 100,000<br />

population), followed by Blacks (120.1) and other races (59.6-93.2).<br />

Similarly, incidence of localized BC was highest among Whites<br />

(81.1), followed by Blacks (63.0) and other races (35.4-58.7). Blacks<br />

had the highest incidence of regional (43.6 vs. 18.8-40.1), distant (9.7<br />

vs. 3.7-6.0), and unknown stage of BC (3.8 vs. 1.6-3.0) compared<br />

to Whites and other races. Incidence of localized BC decreased<br />

slightly for Whites (average annual change -0.4%) during 2000-2009;<br />

however it increased for all other groups (1.4-1.8%). Incidence of<br />

distant BC increased for all races (W: 1.0%, B: 2.3%, O: 3.1-3.8%).<br />

Blacks had the lowest one-year and five-year survival for localized,<br />

regional and distant BC.<br />

Table 1. Survival for female breast cancer patients, by stage and race<br />

Localized Regional Distant Unknown<br />

1-year survival<br />

Whites 100% 98% 66% 79%<br />

Blacks 99% 96% 59% 80%<br />

Other 100% 99% 70% 88%<br />

5-year survival<br />

Whites 98% 84% 24% 56%<br />

Blacks 92% 70% 15% 46%<br />

Other 96% 84% 24% 59%<br />

One-year and five-year survival rates were relatively stable during<br />

2000-2009 for regional and localized BC (annual average change:<br />

-0.6% to 0.7%), for all races. For distant BC, Blacks had least<br />

improvement during 2000-2009 in one-year survival (annual average<br />

change 0.03%, vs. 0.6% for Whites and 3.6% for other races), and<br />

decreasing five-year survival (annual average change -5.4% vs. 2.2%<br />

for Whites and 1.7% for other races).<br />

CONCLUSIONS: Based on US population-based registry data,<br />

Blacks had higher incidence of more advanced breast cancer and<br />

lower long-term survival for all stages of BC. There is considerable<br />

room to improve survival rates for patients with regional and distant<br />

BC, especially among Blacks. Further studies evaluating the survival<br />

experience of later-stage BC patients based on other key factors,<br />

such as age, socioeconomic status, treatment patterns, and tumor<br />

characteristics (e.g. ER/HER2 status) are warranted.<br />

P3-07-08<br />

Recent trends in the characteristics of patients with distant stage<br />

breast cancer in the United States Surveillance, Epidemiology<br />

and End Stage Disease (SEER) program<br />

Li J, Dalvi T, Pawaskar M, Tsai K, Caspard H, Fryzek J. Medimmune,<br />

Gaithersburg, MD; Epistat Institute, Gaithersburg, MD<br />

Objectives. Even though advances in screening and contemporary<br />

diagnostic techniques have led to earlier detection, a proportion<br />

of breast cancer patients are still diagnosed with distant stage<br />

disease. The objective of this research was to describe the changing<br />

demographic and clinical characteristics of patients with distant stage<br />

breast cancer.<br />

Methods. We identified women aged 20 years or older who were<br />

diagnosed with distant stage primary breast cancer between 1995 and<br />

2009 with non-missing hormone receptor (HR) status in the SEER 18<br />

Registry (1973-2009) database. As staging systems changed across<br />

the years of the study, we opted to use the SEER historic stage in<br />

our analyses. Trends in demographic and clinical characteristics were<br />

evaluated by time period of diagnosis (1995-1999, 2000-2004, 2005-<br />

2009) and HR status (HR+ includes ER+/PR+, ER+/PR- and ER-/<br />

PR+; HR- includes ER-/PR- only). Statistically significant changes<br />

were examined using the Cochran Armitage test.<br />

Results. A total of 30,161 women with distant stage breast cancer<br />

were identified. Over the 15 years, the incidence rate (IR) of distant<br />

stage disease was stable (10.6 to 10.9 per 100,000 pys), as well as<br />

the proportion of HR+ cases (68%).<br />

Age did not vary significantly over time; 55% of the HR+ cases<br />

and 45% of the HR- cases were 60 years or older. The proportion of<br />

Black and Hispanic patients increased significantly over time (18 to<br />

24% for HR+; 24 to 33% for HR-) -as did the proportion of patients<br />

diagnosed with grades I and II cancers (44 to 54% for HR+; and 16<br />

to 20% for HR-). The proportion of ductal histologic diagnoses was<br />

stable over time, at 77% among HR+ cases and 93% among HR- cases<br />

in 2005-2009. <strong>Cancer</strong>-directed surgeries (from 63 to 47% for HR+;<br />

72 to 56% for HR-) and radiation therapies (43 to 37% for HR+; 42<br />

to 38% for HR-) became less common over the time. Information<br />

on chemotherapy/systemic therapies is unavailable in this dataset.<br />

Conclusions. Although, incidence rate of distant stage breast<br />

cancer was stable over a time, there was a higher proportion of<br />

diagnosis of grade I/II stage cancer cases potentially due to earlier<br />

cancer detections by screening programs. The growing proportion<br />

of minorities suggested that screening programs be adjusted to the<br />

changing demographics in the United States.<br />

P3-07-09<br />

Prevalence and effects of off-label use of chemotherapeutic agents<br />

in elderly breast cancer patients: estimates from Surveillance,<br />

Epidemiology and End Results-Medicare data<br />

Eaton AA, Sima CS, Panageas KS. Memorial Sloan-Kettering <strong>Cancer</strong><br />

Center, New York, NY<br />

Purpose: To gain FDA approval, drug companies submit evidence<br />

from clinical trials demonstrating that a drug is safe and efficacious<br />

in a certain setting. Current regulations allow oncologists to prescribe<br />

approved drugs for uses other than those for which the drug was<br />

approved (off-label use). While there seems to be a consensus that<br />

off-label use is common in oncology, there is a lack of published data<br />

on recent trends and on utilization in different populations.<br />

The strength of evidence demonstrating safety and efficacy varies<br />

for drugs used in the off-label setting, and patients may be at risk for<br />

unacceptable toxicity. The risk of toxicity may be higher among older<br />

patients, whose ability to tolerate chemotherapy is compromised by<br />

age-related organ decline and comorbidities. Our objective was to<br />

describe the prevalence of off-label use of chemotherapeutic agents<br />

in elderly breast cancer patients with Medicare coverage and to<br />

compare toxicity between patients using off-label drugs and patients<br />

using only approved drugs.<br />

Materials and Methods: We identified women 65 and older with a<br />

first primary breast cancer diagnosed in 2000-2007 from Surveillance,<br />

Epidemiology and End Results (SEER) cancer registry data linked<br />

with Medicare claims. Specific chemotherapeutic agents used<br />

following diagnosis were identified in Medicare claims and classified<br />

as on- or off-label use based on whether the drug was approved<br />

for breast cancer as of May 2012. Thirty-day toxicity (ER visit,<br />

hospitalization or death) was compared between patients whose first<br />

line regimen contained only approved drugs and patients whose first<br />

line regimen contained at least one off-label drug using the Cochran-<br />

Mantel-Haenszel test.<br />

www.aacrjournals.org 325s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Results:<br />

Use of chemotherapy in breast cancer patients, by stage at diagnosis<br />

Stage 0/1 Stage 2 Stage 3 Stage 4 Overall<br />

N 59997 29170 7420 5237 101824<br />

Chemotherapy use<br />

Any chemotherapy 6% 36% 52% 38% 20%<br />

Any approved chemotherapy 6% 36% 52% 37% 20%<br />

Any off-label chemotherapy 0.6% 4% 8% 12% 3%<br />

30-day toxicity<br />

Approved drugs only 5% 7% 10% 15% 6%<br />

At least one off-label drug 6% 8% 16% 17% 8%<br />

The most commonly-used approved drugs were cyclophosphamide<br />

(used in 14% of patients), doxorubicin (10%), docetaxel (5%),<br />

fluorouracil (5%), and paclitaxel (5%).<br />

In total, forty-five off-label agents were used. Vinorelbine and<br />

carboplatin were each used in 1% of patients. No other off-label drug<br />

was used in more than 109 (0.11%) of patients.<br />

Toxicity was significantly higher for patients treated with off-label<br />

agents compared to patients treated with only approved drugs, after<br />

adjusting for stage at diagnosis (p=0.01).<br />

Conclusions: In this population-based cohort, rates of chemotherapy<br />

use were generally low, and rates of off-label use were even lower,<br />

with only 3% of patients treated off-label. Off-label use became more<br />

common as stage at diagnosis increased, with use increasing from 1%<br />

in stage 0/1 patients to 12% in stage 4 patients. The higher toxicity<br />

rates observed in patients treated with off-label drugs may indicate<br />

that these drugs are less safe than approved agents. This result will<br />

be confirmed in a matched analysis accounting for age, stage and<br />

comorbidities.<br />

P3-07-10<br />

Effect of lifestyle and single nucleotide polymorphisms on breast<br />

cancer risk: A case-control study in Japanese women<br />

Mizoo T, Taira N, Nishiyama K, Nogami T, Iwamoto T, Motoki T,<br />

Shien T, Matuoka J, Doihara H, Ishihara S, Kawai N, Kawasaki K,<br />

Ishibe Y, Ogasawara Y. Okayama Medical University, Okayama,<br />

Japan; Okayama Saiseikai General Hospital, Okayama, Japan;<br />

Okayama Rousai Hospital, Okayama, Japan; Kagawa Prefectural<br />

<strong>Cancer</strong> Detection Center, Takamatsu, Kagawa, Japan; Mizushima<br />

Kyodo Hospital, Kurashiki, Okayama, Japan; Kagawa Prefectural<br />

Central Hospital, Takamatsu, Kagawa, Jordan<br />

[Background] Lifestyle, including diet and physical activity, and birth/<br />

breastfeeding history are known to affect the risk for breast cancer.<br />

A correlation between single nucleotide polymorphisms (SNPs) and<br />

breast cancer risk has also been suggested in some reports, but the<br />

gene-environment interaction with breast cancer risk has not been<br />

examined widely.<br />

[Methods] The subjects were 476 breast cancer patients and 528<br />

controls who attended a health check and had no history of breast<br />

cancer. Lifestyle was examined using a questionnaire with 48<br />

questions about diet, physical activity, smoking habits, alcohol<br />

intake, and birth/breastfeeding history, etc. Based on past reports,<br />

we analyzed 6 SNPs (TNRC9-rs3803662, LSP1/11q-rs3817198,<br />

MAP3K1-rs889312, 8q24-rs13281615, 5p12-rs981782, and TGFβ1-<br />

rs1800470) using blood samples, and calculated age-adjusted odds<br />

ratios in a multiple logistic regression analysis.<br />

[Results] The study was performed from December 2010 to<br />

November 2011. Lifestyle factors (age-adjusted odds ratio, [95%<br />

confident interval]) found to have a significant correlation with<br />

development of breast cancer included BMI (1.041, [1.00 - 1.08]),<br />

smoking history (2.28, [1.45 - 3.65]), small number of births (1.18,<br />

[1.05 - 1.34]), no exercise during leisure time (1.36, [1.06 - 1.77]),<br />

low intake of green and yellow vegetables (1.69, [1.01 - 2.85]), and<br />

low intake of mushrooms (1.58, [1.05 - 2.39]). Stratified analysis<br />

based on menopause status showed that smoking history (1.85,<br />

[1.00 - 3.49]) and low intake of green and yellow vegetables (2.5,<br />

[1.03 - 5.88]) were significant risk factors before menopause, and<br />

that smoking history (2.28, [1.45 - 3.65]), low intake of green and<br />

yellow vegetables (2.24, [1.09 - 4.68]), no exercise during leisure<br />

time (1.67, [1.19 - 2.36]), small number of births (1.49, [1.25 - 1.78]),<br />

and no breastfeeding history (1.03, [1.01 - 1.06]) were significant risk<br />

factors after menopause. The results of SNP analysis suggested that<br />

TNRC9-rs3803662 was a significant risk factor in women before<br />

menopause (2.29, [1.25 - 4.25]). However, in multivariate analysis<br />

including lifestyle factors and SNPs, only smoking history emerged<br />

as a significant risk factor in women before menopause.<br />

[Conclusion] A correlation between lifestyle and breast cancer risk<br />

was found in this study, consistent with previous findings. Lifestyle<br />

and environmental factors such as births and breastfeeding may be<br />

more important than SNPs as risk factors for breast cancer.<br />

P3-07-11<br />

Retrospective database review of outcomes in invasive lobular<br />

carcinoma and invasive ductal carcinoma of the breast<br />

Shih Y-C, Dillon P. University of Virginia, Charlottesville, VA<br />

Background. Invasive lobular carcinoma (ILC) and invasive ductal<br />

carcinoma (IDC) are histologically distinct breast cancer subtypes.<br />

While ILC and IDC have similar disease-free survival (DFS) and<br />

overall survival (OS), the patterns of metastatic recurrence are<br />

suspected to differ due to differing adhesion markers.<br />

Methods. Retrospective review of a cohort of breast cancer patients<br />

treated at a tertiary academic medical center from May 1999 to April<br />

2012 was performed. Demographic, pathologic, treatment and followup<br />

data were collected from a cancer database and the institutional<br />

medical records.<br />

Results. There were 179 ILC and 358 IDC (1:2 stage-matched)<br />

patients in the study period. Mean age (59 vs. 58 years), gender,<br />

race (88% vs. 80% Caucasian), menopause state (71% vs. 66%<br />

post-menopausal), and family history (48% vs. 42% positive for<br />

breast cancer) were similar in the two groups. Median follow-up<br />

was 4.7 years. ILC patients had more negative mammograms upon<br />

diagnosis (6.8% vs. 1.4%, p=0.001), and their breast cancers were<br />

more likely to be hormone-receptor-positive/HER2-negative (90%<br />

vs. 69%, p


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 3<br />

P3-07-12<br />

Risk Factors for <strong>Breast</strong> <strong>Cancer</strong> in Women Served in Odete<br />

Valadares Maternity, Belo Horizonte-MG<br />

Sediyama CMNO, Peluzio MCG, Abranches MV. Universidade<br />

Federal de Viçosa, Viçosa, MG, Brazil<br />

With changes in lifestyle of the population and changes in women<br />

reproductive patterns in the last century, the profile of mortality<br />

among women has changed, today the prevailing non-infectious<br />

chronic diseases, like cancer, including breast cancer. This paper is<br />

a case-control population-based, with the objective of evaluating the<br />

risk factors and their association with breast cancer in a population of<br />

women seen in Odete Valadares Maternity, in the Hospital Foundation<br />

of the State of Minas Gerais, in Belo Horizonte, between January and<br />

July 2006. Inclusion criteria were age over 18 years and referral for<br />

mammography service. Women with abnormal mammography and<br />

confirmation of malignancy of the breast were invited to participate<br />

in the study, and those with normal mammography were invited<br />

to compose the control group. We evaluated 710 women and 456<br />

selected to participate in the study, 261 with breast cancer and 195<br />

healthy controls.<br />

Table1: Characterization of the demographic, anthropometric and reproductive patient cases and<br />

controls<br />

Variables Control Cases p<br />

Mean(SD) Median Mean(SD) Median<br />

65.55(39.80-<br />

Weight (kg) 67.12(±14.82)<br />

66.74(±14.13) 64.30(39-116) 0.27*<br />

113.68)<br />

Age (years) 47.82(±9.62) 47(16-73) 54.8(±11.93) 53(30-92) 0.8 1.191 0.715 - 1.984<br />

Alcoholism 0.5483 0.351 - 0.856#<br />

Smoking 1.084 0.694 - 1.693<br />

* A positive association. # Protective Association.<br />

This study found a positive association between physical inactivity,<br />

menopause, nulliparity, lack of breastfeeding and family history of<br />

breast cancer and the diagnosis of breast cancer, and alcohol conferred<br />

a protective effect in this population.<br />

P3-07-13<br />

Importance of socioeconomic status in relation to breast cancer<br />

risk and prognostic factors in Argentina.<br />

Croce MV, Demichelis SO, Cermignani L, Zwenger A, Segal-Eiras<br />

A, Giacomi N. Faculty of Medical Sciences, UNLP, La Plata, Buenos<br />

Aires, Argentina<br />

In Argentina, there are no studies evaluating neither breast cancer<br />

screening nor risk and prognostic factors in relation to socioeconomic<br />

status among women in metropolitan areas. Taking into account<br />

that Argentina presents social and economical disparities and that<br />

there is a mixture of features of both developed and developing<br />

societies, it is interesting to compare prognostic and risk factors<br />

in disadvantaged and advantaged women as it would clarify the<br />

influence of socioeconomic factors in breast cancer biology. The<br />

purpose of this study was to compare risk and prognostic factors of<br />

invasive breast cancer in two different Argentine populations. Study<br />

participants and data collection. A total of 625 women who had a<br />

histologically confirmed diagnosis of invasive primary breast cancer<br />

were included; 270 patients belonged to a private clinic of the city<br />

of La Plata (province of Buenos Aires) belonging to an Advantaged<br />

Population (AP) and 355 patients belonged to a public hospital of<br />

the city of Neuquén (province of Neuquén, Patagonia) belonging<br />

to a Disadvantaged Population (DP). Women of these geographical<br />

regions and first diagnosed with invasive primary breast carcinoma<br />

from 2002 until 2007 were eligible as cases. There were no racial or<br />

ethnic differences between the two groups of women; all of them were<br />

born in Argentina. Risk factors included age at diagnosis, menarche<br />

and menopause status, breastfeeding and parity, while prognostic<br />

factors were: disease stage, number of metastatic lymph nodes, tumor<br />

size, histological and nuclear grade, vascular invasion, ER, PR, and<br />

Her2neu statuses. Methods: Statistical analysis included frequency<br />

analysis and ANOVA (p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P3-07-14<br />

Initial treatment and survival among elderly breast cancer<br />

patients in the United States by estrogen receptor status and<br />

cancer stage at diagnosis: an analysis of national registry data<br />

2000-2009<br />

Lang K, Huang H, Namjoshi M, Federico V, Menzin J. Boston Health<br />

Economics, Waltham, MA; Novartis Pharmaceuticals Corporation,<br />

East Hanover, NJ<br />

BACKGROUND: There are few recent studies of initial treatment<br />

and survival among elderly, newly diagnosed breast cancer (BC)<br />

patients by estrogen receptor (ER) status and cancer stage at diagnosis.<br />

METHODS: The linked Surveillance and Epidemiology End Results-<br />

Medicare (SEER-Medicare) database was used for this analysis. SEER<br />

is an epidemiologic surveillance system consisting of populationbased<br />

tumor registries designed to track cancer incidence and survival<br />

in geographically defined areas that represent approximately 25% of<br />

the US population. The SEER registry file (available through 2007)<br />

is linked to Medicare claims and enrollment data available through<br />

2009. We identified female patients newly diagnosed with Stage I, II,<br />

III, or IV breast cancer in a SEER registry between January 2002 and<br />

December 2007. Study patients were required to be aged 66+ years<br />

with no prior history of any other (non-breast) cancer. Patients were<br />

followed from the date of breast cancer diagnosis through death or<br />

December 31, 2009. Patients were identified as ER+ (ER-) if the ER<br />

assay was reported to SEER as positive/elevated (negative/normal).<br />

Demographics, initial treatment (surgery or radiation within 4 months<br />

of diagnosis), and survival (proportion of patients who died during<br />

the study period, survival at 96 months and median survival) were<br />

evaluated by ER status and cancer stage. Kaplan-Meier (KM) survival<br />

curves were estimated by ER status and stage.<br />

RESULTS: 70,845 female BC patients (59,243 ER+, and 11,602<br />

ER-) were identified. Age at diagnosis was similar for ER+ and ERpatients<br />

(mean [SD]: 75-77 [7] years; median: 74-76 years) across all<br />

stages. More ER+ patients were White (86% vs. 80% for ER-), and<br />

diagnosed at Stage I (56% vs. 39%). Fewer ER+ than ER- patients<br />

were diagnosed at Stage II (32% vs. 39%), Stage III (8% vs. 15%)<br />

and Stage IV (4% vs. 7%). Most patients had surgery in the first 4<br />

months following diagnosis (96% ER+; 94% ER-), with Stage IV<br />

patients least likely (41% ER+ vs. 47% ER-). ER+ patients were more<br />

likely to receive initial radiation than ER- patients (48% vs. 42%),<br />

with a similar trend by stage (Stage I: 54% vs. 50%; Stage II: 40%<br />

vs. 35%; Stage III: 46% vs. 44%; Stage IV: 30% vs. 29%). Fewer<br />

ER+ patients died during follow-up (23% vs. 36% of ER- patients).<br />

KM survival analyses suggest that survival rates were higher for ER+<br />

than ER- patients for the entire study period, overall (survival at 96<br />

months: 64% vs. 52%), and by stage (Stage I: 73% vs. 70%; Stage<br />

II: 60% vs. 50%; Stage III: 38% [median survival 72 months] vs.<br />

27% [median survival 41 months]; Stage IV: 12% [median survival<br />

23 months] vs. 8% [median survival 9 months]).<br />

CONCLUSIONS: Among elderly patients diagnosed with BC,<br />

those with ER+ status were diagnosed at earlier stages and had better<br />

survival outcomes. Further study of treatment patterns and outcomes<br />

by ER status is warranted.<br />

P3-08-01<br />

The prospective risk of ovarian cancer in 1433 women in a breast<br />

cancer family history clinic: no increased risk in families testing<br />

negative for BRCA1 and BRCA2.<br />

Evans DGR, Ingham SL, Sahin S, Buchan I, Warwick J, O’Hara C,<br />

Moran A, Howell A. St Mary’s Hospital, Manchester, United Kingdom;<br />

Wythenshawe Hospital, Manchester, United Kingdom; The University<br />

of Manchester, Manchester, United Kingdom; Imperial College<br />

London, United Kingdom; North West <strong>Cancer</strong> Intelligence Service,<br />

NHS Foundation Trust, Manchester, United Kingdom; Christie<br />

Hospital, Manchester, United Kingdom<br />

Since the identification of the BRCA1 and BRCA2 genes speculation<br />

has continued regarding the breast and ovarian cancer risk associated<br />

with mutations in these genes. Also, as to whether mutations in these<br />

genes account for the majority of the association between breast and<br />

ovarian cancer. Identification of RAD51C and RAD51D mutations<br />

in breast/ovarian cancer families has reawakened the debate as to<br />

whether it is safe to reassure women from BRCA negative families<br />

that they are not at increased risk of ovarian cancer. 1433 women<br />

from breast cancer families that had tested negative for BRCA1/2<br />

were followed for a total of between 0.04 and 25 years and checked<br />

against a cancer registry for ovarian cancer incidence. Data was<br />

censored at ovarian cancer diagnosis, oophorectomy or death. During<br />

16358 women years of follow up no invasive epithelial ovarian cancer<br />

occurred, although 2 borderline tumors were diagnosed. Expected<br />

rates for this cohort from cancer registry data were 2.75 epithelial<br />

ovarian tumors with 2.52 for invasive cancer and 0.23 for borderline<br />

tumors. The upper confidence limit for invasive RR was 1.3 and for<br />

borderline tumors 0.97-31. In conclusion this the largest prospective<br />

follow up of a BRCA negative cohort has demonstrated that there is<br />

no increase in risk of invasive ovarian cancer in families that have<br />

tested negative for BRCA1/2.<br />

P3-08-02<br />

Common variants at 10p14 and 1p11.2 display heterogeneity in<br />

breast cancer associations by E-cadherin tumor tissue expression<br />

in two independent datasets<br />

Horne HN, Sherman ME, Garcia-Closas M, Pharoah PD, Blows<br />

FM, Yang XR, Lissowska J, Brinton LA, Chanock SJ, Figueroa JD.<br />

National <strong>Cancer</strong> Institute, Rockville, MD; The Institute of <strong>Cancer</strong><br />

Research, Sutton, Surrey, United Kingdom; University of Cambridge,<br />

United Kingdom; M. Sklodowska-Curie Memorial <strong>Cancer</strong> Center and<br />

Institute of Oncology, Warsaw, Poland<br />

Background: E-cadherin is a tumor suppressor gene involved in<br />

cell-cell adhesion, epithelial-to-mesenchymal transitions (EMT) and<br />

invasion. Loss of E-cadherin expression is strongly associated with<br />

lobular breast cancers, which exhibit single cell patterns of infiltration<br />

and are often estrogen receptor positive. We sought to determine if<br />

relative risk estimates for 19 established breast cancer susceptibility<br />

loci were modified by E-cadherin breast tumor tissue expression.<br />

Methods: Case-control analyses included up to 1885 invasive breast<br />

cancer cases and 2366 age and site matched controls aged 20-74 years<br />

from the Polish <strong>Breast</strong> <strong>Cancer</strong> Study (PBCS), a population based casecontrol<br />

study conducted in Poland from 2000-2003. Genotyping of<br />

the 19 single nucleotide polymorphisms (SNPs) was performed using<br />

TaqMan® assays. Tissue expression of E-cadherin was assessed using<br />

immunohistochemical (IHC) staining of tissue microarrays and IHC<br />

results were scored as the product of percent positive tumor cells x<br />

intensity. Tumors having a score of


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 3<br />

to estimate odds ratios (OR) and 95% confidence intervals (95% CI)<br />

for each breast cancer subtype, defined by E-cadherin expression<br />

levels, compared to controls. Case-only data from the PBCS (N = 797)<br />

and the Study of Epidemiology and Risk Factors in <strong>Cancer</strong> Heredity,<br />

SEARCH (N = 2155) was used in logistic regression models to test<br />

for heterogeneity of SNPs by E-cadherin expression.<br />

Results: Three SNPs suggested significant heterogeneity by<br />

E-cadherin expression in the PBCS: rs2046210 at 6q25.1(ESR1)<br />

[per-allele ORs (95% CI); 1.53 (1.19-1.98) for E-cadherin low tumors<br />

and 0.99 (0.86-1.14) for E-cadherin high tumors, P-heterogeneity<br />

= 0.002]; rs1045485 at 10p14 (CASP8) [per-allele ORs (95% CI);<br />

0.62 (0.41-0.93) for E-cadherin low tumors and 0.98 (0.81-1.18) for<br />

E-cadherin high tumors, P-heterogeneity = 0.04]; and rs11249433<br />

at 1p11.2 (NOTCH2/FCGR1B) [per-allele ORs (95% CI); 1.29<br />

(1.02-1.64) for E-cadherin low tumors and 1.01 (0.88-1.15) for<br />

E-cadherin high tumors, P-heterogeneity = 0.06]. Combined caseonly<br />

analysis of PBCS and SEARCH for these three SNPs showed<br />

significant heterogeneity by E-cadherin expression for rs11249433<br />

[Interaction OR (95% CI); 1.19 (1.05-1.36), P-heterogeneity =<br />

0.007] and rs1045485 [Interaction OR (95% CI); 0.69 (0.53-0.90),<br />

P-heterogeneity = 0.007]. The association with rs2046210 [Interaction<br />

OR (95% CI); 1.12 (0.61-2.03), P-heterogeneity = 0.73] did not<br />

remain significant in combined analyses.<br />

Conclusion: Our findings provide evidence that associations for<br />

breast cancer susceptibility loci vary by E-cadherin tumor tissue<br />

expression, which has not been described previously. Specifically, our<br />

results suggest that the genetic markers rs11249433 and rs1045485<br />

may preferentially modify risk for tumors with low or absent<br />

E-cadherin expression in two independent data sets.<br />

P3-08-03<br />

Urinary levels of prostaglandin E 2 metabolite and postmenopausal<br />

breast cancer<br />

Kim S, Taylor JA, <strong>San</strong>dler DP. Georgia Health Sciences University,<br />

Augusta, GA; National Institute of Environmental Health Sciences,<br />

Research Triangle Park, NC<br />

Estrogens are considered a key player in the mammary carcinogenesis.<br />

In postmenopausal women, peripheral aromatization of androgens<br />

is the predominant source of estrogens. Given that prostaglandin<br />

E 2 (PGE 2 ) is a potent activator of aromatase expression, we<br />

hypothesized that endogenous level of PGE 2 would influence estrogen<br />

bioavailability and subsequent breast cancer risk in postmenopausal<br />

women. In addition, because PGE 2 synthesis is suppressed by<br />

treatment of non-steroidal anti-inflammatory drugs (NSAIDs)<br />

via inhibition of cyclooxygenase enzyme activity, we further<br />

hypothesized that regular use of NSAIDs may modify the association<br />

between PGE 2 and postmenopausal breast cancer risk. We carried out<br />

a case-cohort analysis within the NIEHS Sister Study, a prospective<br />

cohort study of 50,884 women. Postmenopausal women who were<br />

aged 50 or older and did not report current use of hormones were<br />

eligible for the present analysis (N=11,338). Urinary levels of a major<br />

PGE 2 metabolite (PGE-M), which is generally accepted as the most<br />

accurate index of endogenous PGE 2 formation, were quantified using<br />

liquid chromatography/tandem mass spectrometry in 302 incident<br />

breast cancer cases and 305 subcohort members who were randomly<br />

selected by frequency matching for age among cases. Hazard ratios<br />

(HR) and 95% confidence intervals (95% CI) were estimated using<br />

Prentice’s pseudo-likelihood approach with Barlow’s weighting<br />

scheme. Several factors were associated with higher levels of urinary<br />

PGE-M in the multivariable analysis, including current smoking,<br />

obesity, high caloric intake from saturated fat and surgical menopause.<br />

No associations were observed for dietary intake of omega 3 fatty<br />

acids, use of vitamin supplements and physical activity. Overall, there<br />

was no association between urinary PGE-M and postmenopausal<br />

breast cancer risk. After including the interaction terms between<br />

NSAID use and urinary levels of PGE-M in the model, however, high<br />

levels of urinary PGE-M were associated with an increased risk of<br />

breast cancer in postmenopausal women who did not regularly use<br />

NSAIDs (likelihood ratio test p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

of endogenous and exogenous hormones and hormonal agents such<br />

as tamoxifen, used in the treatment of breast cancer this association<br />

may have wider implications.<br />

P3-08-05<br />

Title: Copy number variation and risk of chemotherapy-related<br />

infection.<br />

Abraham JE, Rueda OM, Chin S-F, Guo Q, Harrington P, Earl<br />

HM, Pharoah PD, Caldas C. Strangeway’s Research Laboratory,<br />

University of Cambridge, Cambridgeshire, United Kingdom;<br />

University of Cambridge NHS Foundation Hospitals, Cambridge,<br />

Cambridgeshire, United Kingdom; <strong>Cancer</strong> Research UK Cambridge<br />

Research Institute, Li Ka Shing Centre, Cambridge, Cambridgeshire,<br />

United Kingdom<br />

Background<br />

Copy-number variations (CNVs) are DNA changes that result in<br />

regions of the genome being either duplicated (copy number gains)<br />

or deleted (copy number losses). CNVs have been identified in 12%<br />

of the human genome. Although CNVs do not appear to have a major<br />

role in disease susceptibility, it is known that they are involved in drug<br />

metabolism, toxicity and response. Infection is a common dose and/or<br />

treatment limiting chemotherapy-related toxicity. Infections can be a<br />

major cause of morbidity and mortality. Although a limited number<br />

of studies have investigated the association of single nucleotide<br />

polymorphisms (SNPs) with severity of infection, no studies so<br />

far have identified CNV associations with chemotherapy-related<br />

infection. In a non-oncological setting, CNVs have been noted to be<br />

associated with altered risk of certain types of infections and severe<br />

sepsis. This study aims to evaluate whether CNV are associated<br />

with the risk of chemotherapy-related infection in a genome wide<br />

association study (GWAS) of breast cancer patients recruited to<br />

clinical trials and treated with chemotherapy regimens including<br />

paclitaxel, epirubicin, cyclophosphamide, gemcitabine, 5fluorouracil<br />

and methotrexate.<br />

Method<br />

DNA from blood and saliva samples were collected as part of the<br />

pharmacogenetics GWAS, PGSNPS. Chemotherapy-related infection<br />

was assessed using the NCI Common Toxicity Criteria (CTC).<br />

Patients (n=1921 samples) were classified as cases (NCI CTC grades<br />

2-4) or as controls (NCI CTC grades 0-1). The Affymetrix SNP6.0<br />

array, which contains more than 946,000 probes for the detection of<br />

CNVs and more than 906,600 SNPs, was used as the copy number<br />

estimation platform. DNA was segmented using DNAcopy (circular<br />

binary segmentation). CNVs were called based on their median and<br />

standard deviation. Log-ratios larger than the median plus 2 times<br />

the standard deviation were considered gains and log-ratios with<br />

values smaller than 2.5 times the standard deviation were considered<br />

as losses. Only regions with a copy number alteration present in<br />

at least a 10% of the samples were considered for the analysis.<br />

Fisher’s exact test was performed on each of these regions and the<br />

variable ‘infection’. 2-side p-values were obtained and adjusted using<br />

Benjamin-Hochberg correction.<br />

Results<br />

One CNV region (frequency of gain 0.16) showed an association<br />

with increased risk of infection (Odds Ratio=1.79, 95% Confidence<br />

Interval [1.34-2.40], P=6.37x10 -5 ). Five other CNV regions<br />

demonstrated copy number gains and three CNV regions showed<br />

copy number losses that were associated with an increased risk of<br />

infection (P=10 -4 ). All regions had a frequency of gain or loss > 0.10<br />

and were therefore not rare variants.<br />

Conclusion<br />

These data suggest that CNVs may play a role in increased risk<br />

of chemotherapy-related toxicity. The most significant CNVs<br />

associated with infection are currently undergoing validation using<br />

an independent cohort of adjuvant breast cancer patients with<br />

similar exposure to chemotherapy and comparable documentation<br />

of chemotherapy-related infection.<br />

P3-08-06<br />

Triple Negative <strong>Breast</strong> <strong>Cancer</strong> and BRCA Status: Implications<br />

for Genetic Counseling<br />

Haque R, Alvarado M, Ahmed SA, Shi JM, Chung JWL, Avila CC,<br />

Zheng CX, Tiller GE. Kaiser Permanente Southern California,<br />

Pasadena, CA<br />

BACKGROUND<br />

Controversy exists whether women newly diagnosed with triple<br />

negative breast cancer (TNBC) should be referred to genetic<br />

counseling as they may be more likely to be BRCA carriers. However,<br />

prior studies included small numbers of carriers and their results have<br />

had limited influence on practice guidelines. The objective of this<br />

study was to determine the association of breast cancer molecular<br />

subtype and BRCA status in a large group of medically insured<br />

women.<br />

METHODS<br />

We examined a cohort of 2,105 women with breast cancer history<br />

tested for BRCA mutations in a large California health plan from<br />

1997-2011. BRCA test results were recorded in the health plan’s<br />

clinical genetics registry. Of the 2,105 breast cancer patients, 249<br />

were BRCA mutation carriers (143 BRCA1 carriers, and 106 BRCA2<br />

carriers). We conducted data linkages of all patients with the health<br />

plan’s NCI-SEER affiliated tumor registry and identified ER, PR,<br />

and HER2. HER2 status was also captured from pathology reports<br />

using natural language processing. ER, PR, and HER2 status were<br />

assessed by immunohistochemical or FISH techniques. Patients were<br />

classified into four main biologic subtypes: triple negative (ER-/PR-/<br />

HER2-); luminal A (ER+ and/or PR+/HER2-); luminal B (ER+ and/or<br />

PR+/HER2+); and HER2-enriched (HER2+/ER-). We examined the<br />

association between molecular subtypes (collapsed into TNBC vs. non<br />

TNBC categories) and BRCA1/2 mutation status using contingency<br />

table analyses. P-values (two-sided) were estimated using chi-square<br />

analysis. Multivariable logistic regression was used to estimated<br />

adjusted odds ratios (OR) and 95% confidence intervals.<br />

RESULTS<br />

TNBC subtype was strongly associated with BRCA status (P


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 3<br />

CONCLUSION<br />

TNBC was strongly associated with BRCA1 status, but not with<br />

BRCA2 status. Statistically significant numbers of patients with<br />

BRCA mutations have a TNBC profile. These patients should<br />

therefore be referred to clinical genetics for further evaluation and<br />

possible testing.<br />

P3-08-07<br />

HER2 positive breast carcinomas: trend in evolution between<br />

1998 and 2008 and relationship with clinico-pathological<br />

characteristics in a population based study.<br />

Beltjens F, Bertaut A, Pigeonnat S, Pouget N, Guiu S, Poillot M-L,<br />

Charon-Barra C, Coudert B, Dabakuyo S, Fumoleau P, Arveux P,<br />

Arnould L. Centre GF Leclerc, Dijon, France<br />

Background:<br />

Determination of HER2 status in invasive breast carcinomas is now<br />

mandatory for the optimal treatment of the patient. A vast majority<br />

of initial published studies described positivity in about 20 to 25<br />

% of breast carcinomas, but the proportion of HER2 positive gene<br />

expression in women with breast cancer varies in the literature<br />

from 9 to 74%. The population of the tumor analyzed (for example<br />

histologic types, stage, age), the type of the assays used and the<br />

lack of uniformity or quality controls in detection assays may partly<br />

explain these differences. Our study describes the evolution of HER2<br />

positive invasive breast cancer proportions during a period of 10<br />

years and identifies clinical and pathological characteristics of these<br />

positive tumors.<br />

Methods:<br />

Based on the regional, population-based breast cancer registry, we<br />

identify 2396 patients diagnosed with invasive breast cancer in the<br />

Côte-d’Or department and treated in a single institution between 1998<br />

and 2008. The HER2 status was determined by immunohistochemistry<br />

and by fluorescence in situ hybridization by a single team that<br />

performed regular internal quality controls and participated to<br />

multiannual external international quality control programs.<br />

Results:<br />

During the 1998-2008 period, 12.8% of invasive breast cancers<br />

were identified as HER2 positive, with no trend to annual variations<br />

(p=0.27). During the same period, despite an increase of 45% of<br />

the annual diagnosed cancers in the institution, the clinical and<br />

pathological characteristics such as age, tumor size, grade, hormonal<br />

receptors expression remain stable. We only noticed a slight but<br />

significant increase of lobular carcinomas (p=0.036). In multivariate<br />

analysis, HER2 positivity was as expected associated with : hormonal<br />

receptor negative status (OR=1.7 [IC95% : 1.2-2.4], p=0.0035),<br />

metastatic presentation at diagnosis (OR=2 [IC95% : 1.2-3.4],<br />

p=0.0092), ductal subtype (OR=1.9 [IC95% : 1.1-3.1], p=0.0178)<br />

and high SRB grade (OR=2.8 [IC95% : 1.7-4.5] for SBR II, OR=5.4<br />

[IC95% : 3.2-9.1] for SBR III, p=0.0035). Conversely, patients older<br />

than 75 years presented mostly HER2 negative tumors (OR=0.5<br />

[IC95% : 0.3-0.8], p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P3-08-09<br />

Clinical and Pathological Characteristics of <strong>Breast</strong> <strong>Cancer</strong> in<br />

Women with Neurofibromatosis Type 1<br />

Yap Y-S, Wong J, Chan M, Yong W-S, Ong K-W, Lo S-K, Ngeow J,<br />

Tan B, Madhukumar P, Tan M-H, Ang P, Teh B-T, Tan P-H, Lee AS-<br />

G. National <strong>Cancer</strong> Centre Singapore; Singapore General Hospital<br />

Background:<br />

Neurofibromatosis type 1(NF1) is an autosomal dominant genetic<br />

disorder affecting 1 in 3,000 individuals worldwide. Risk of cancer,<br />

including breast cancer, is increased, as NF1 is a tumour suppressor<br />

gene. There is limited data on the characteristics of breast cancer in<br />

individuals with NF1.<br />

Objectives:<br />

To study the clinical and pathological characteristics of breast cancer<br />

in women with NF1.<br />

Methodology:<br />

Patients with both NF1 and breast cancer were identified retrospectively<br />

from hospital records as well as prospectively when seen at National<br />

<strong>Cancer</strong> Centre Singapore (NCCS) and Singapore General Hospital<br />

(SGH). All women had histologically proven breast cancer and<br />

fulfilled at least 2 of the 7 criteria developed by the NIH Consensus<br />

Conference for clinical diagnosis of NF1. Germline NF1 mutation<br />

sequencing is being performed on patients with blood specimens<br />

available. Details on patient demographics, tumour grade, stage,<br />

receptor status and clinical outcome were evaluated. The pathological<br />

characteristics of NF1-associated breast cancer were then compared<br />

to sporadic breast cancers in our 2001-2004 cohort, with a focus on<br />

grade and HER2 positivity rate.<br />

Results:<br />

We identified 13 women with NF1 and breast cancer seen at our<br />

institutions from 2001 to present date. Average age of breast cancer<br />

diagnosis for women testing positive was 48 years (range 30-64).<br />

All 13 patients had invasive ductal carcinoma; of which 5 were<br />

stage 3 or 4 at diagnosis. To date, 4 out of 12 patients with stage<br />

1-3 breast cancer have relapsed. Grade, estrogen, progesterone and<br />

HER2 receptor status were fully available for 12 patients. Ten out<br />

of 12 tumours (83%) were grade 3, compared to 45% (705/1578)<br />

among our sporadic breast cancers (p=0.007). Eight out of 12(67%)<br />

NF1-associated breast cancers were HER2 positive (IHC 3+ or FISH<br />

positive), compared to 27% (367/1367) in sporadic cancers (p=0.002).<br />

Two other NF1-associated tumours were triple negative, and two<br />

were hormone receptor positive and HER2 negative. Six out of 12<br />

(50%) NF1-associated cancers were estrogen receptor (ER) positive,<br />

compared to 67% (1124/1689) in sporadic cancers (p=0.2).<br />

Conclusion:<br />

Our findings suggest that NF1-associated breast cancer tends to<br />

be more aggressive, with a higher frequency of grade 3 and HER2<br />

positive tumours compared to sporadic breast cancer. Further genomic<br />

profiling of these tumours (in progress) may elucidate the role of<br />

NF1 in breast cancer. This may also have implications for sporadic<br />

tumours with somatic NF1 mutations in individuals without NF1.<br />

P3-09-01<br />

Odds of the triple negative subtype and survival of stages 1-3<br />

breast cancer: Variation by race/ethnicity<br />

Parise C, Caggiano V. Sutter Institute for Medical Research,<br />

Sacramento, CA<br />

Background<br />

Triple negative (TN) breast cancer is known to be more common in<br />

black and Hispanic women and associated with poor survival. This<br />

study assesses the differences in the odds and survival of the TN<br />

versus the ER+/PR+/HER2- (the most common subtype) by race/<br />

ethnicity separately for AJCC stages 1, 2, and 3.<br />

Methods<br />

Using the California <strong>Cancer</strong> Registry for years 2000-2010, we<br />

examined 91,393 cases of stages 1-3 TN and ER+/PR+/HER2- first<br />

primary female invasive breast cancer. Race/ethnicity was defined<br />

as white, black, Hispanic, and Asian/Pacific Islander (API). Logistic<br />

regression was used to assess the association of race with odds of<br />

the TN subtype. Kaplan-Meier was used to compute 6-year survival.<br />

Cox proportional hazards was used to assess the risk of mortality of<br />

the TN subtype by race adjusting for age, grade, year of diagnosis,<br />

and socioeconomic status. All analyses were run separately for stages<br />

1, 2, and 3.<br />

Results<br />

The table shows the distribution of the TN and ER+/PR+/HER2-<br />

subtypes by stage and race/ethnicity.<br />

The distribution of the TN and ER+/PR+/HER2-subtypes by stage and race/ethnicity<br />

Stage 1 Stage 2 Stage 3 Total (%)<br />

Race/ ER+/PR+/<br />

ER+/PR+/<br />

ER+/PR+/<br />

TN<br />

TN<br />

TN<br />

ethnicity HER2-<br />

HER2-<br />

HER2-<br />

White 29,647 3,815 18,797 4,677 4,353 1,432 62,721(68.6)<br />

Black 1,438 565 1,392 987 410 377 5,169(5.7)<br />

Hispanic 4,834 977 4,283 1,771 1,409 623 13,897(15.2)<br />

API 4,086 568 3,146 834 747 225 9,606(10.5)<br />

Total (%) 40,005(43.8) 5,925(6.5) 27,618(30.2) 8,269(9.0) 6,919(7.6) 2,657(2.9) 91,393<br />

Blacks had increased odds of the TN compared with whites for stages<br />

1 (OR=2.24; 95%CI=1.96-2.56); 2 (OR=2.01; 95%CI=1.81-2.25) and<br />

3 (OR=2.00; 95%CI=1.66-2.40).<br />

Hispanics had increased odds for the TN in stages 1 (OR=1.23;<br />

95%CI=1.11-1.35) and 2(OR=1.20; 95%CI=1.11-1.31). APIs had<br />

decreased odds of the TN in stages 2 (OR=0.85; 95%CI=0.77-0.93)<br />

and 3 (OR=0.77; 95%CI=0.64-0.92).<br />

KM survival showed that APIs had the best 72 month survival for<br />

the TN in stages 1 (86%), 2 (78%), and 3(59%).<br />

Hazard ratios (HRs) indicated that all races with TN had the same risk<br />

of mortality in stage 1. Only in stage 2 did blacks have an increased<br />

risk of mortality (HR=1.17; 95%CI=1.02-1.36) while APIs had a<br />

decreased risk (HR=0.78; 95%CI=0.65-0.94). In stage 3, Hispanics<br />

(HR=0.85; 95%CI=0.72-0.99) and APIs (HR=0.70; 95%CI=0.55-<br />

0.89) had decreased risk of mortality compared with whites.<br />

Conclusions<br />

Race/ethnicity is an important factor for survival of the TN subtype:<br />

1. Although blacks and Hispanics are more likely to have the TN<br />

subtype, blacks have an increased risk of mortality compared with<br />

whites only in stage 2.<br />

2. For stage 1, all races have the same risk of mortality.<br />

3. Hispanics have decreased risk of mortality in stage 3.<br />

4. APIs have decreased risk of mortality in both stages 2 and 3.<br />

P3-09-02<br />

The HER2 –positive subtypes by stage and race/ethnicity<br />

Caggiano V, Parise C. Sutter Institute for Medical Research,<br />

Sacramento, CA<br />

HER2-positivity is often associated with poor survival. The purpose<br />

of this study is to determine if there are differences in mortality among<br />

the HER-positive subtypes when stratified by stage and race/ethnicity.<br />

Methods<br />

Using the California <strong>Cancer</strong> Registry for years 2000-2010, we<br />

examined 99,897 cases of stages 1-3 ER+/PR+/HER2- and all<br />

HER2-positive first primary female invasive breast cancers. Race/<br />

ethnicity was defined as white, black, Hispanic, and Asian/Pacific<br />

Islander (API). Kaplan-Meier (KM) and the Log-Rank test were used<br />

to compute 6-year survival. Cox proportional hazards was used to<br />

assess the risk of mortality of each of the HER2-positive subtypes<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

332s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 3<br />

when compared with the ER+/PR+/HER2- subtype (the most common<br />

subtype). All analyses were adjusted for age, grade, year of diagnosis<br />

(


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

luminal B, TN, and Her2-type). Demographics (age at diagnosis,<br />

menopausal status, and BMI) were collected from medical records.<br />

Case-only odds ratios (ORs) and 95% confidence intervals (CIs) were<br />

estimated using logistic regression, adjusting for age at diagnosis.<br />

Results In all cases, the patients were Japanese. Of the 531 cases<br />

with IHC marker data, 333 (62.7%) were classified as luminal A,<br />

85 (16.0%) were luminal B, 43 (8.1%) were Her2-type, and the<br />

remaining 70 cases (13.2%) were TN. The distribution of patient<br />

demographics (age at diagnosis, menopausal status, and BMI) did<br />

not differ significantly by breast cancer tumor subtype. Case-only<br />

ORs comparing each subtype to luminal A were caluculated. Of<br />

the TN cases, postmenopausal TN cases were more likely to be<br />

underweight (OR = 3.14, 95% CI = 1.19 to 8.01). Although some<br />

epidemiological studies have reported that higher BMI was associated<br />

with premenopausal TN cases compared with luminal A cases, this<br />

association was not found among premenopausal TN cases analyzed<br />

using BMI 18.5 to 24.9 kg/m 2 as the reference in this analysis.<br />

However, there were no underweight cases (BMI < 18.5 kg/m 2 )<br />

among the premenopausal TN cases in the present study. Therefore,<br />

the association between BMI and the TN subtype was also analyzed<br />

using BMI < 25 kg/m 2 as the reference. Compared to luminal A cases,<br />

premenopausal TN cases were more likely to be obese (OR = 4.11,<br />

95% CI = 1.10 to 14.40), similar to reports from Western countries.<br />

Compared to luminal A cases, premenopausal luminal B cases were<br />

likely to be underweight (OR = 3.27, 95% CI = 0.88 to 11.39) or obese<br />

(≥ 25 kg/m 2 ) (OR = 3.32, 95% CI = 0.98 to 10.81), yet this association<br />

was of borderline significance. Compared to luminal A cases, luminal<br />

B and Her2-type cases were likely to be underweight (BMI < 18.5<br />

kg/m 2 ), yet this association was of borderline significance (luminal<br />

B: OR = 2.12, 95% CI = 0.97 to 4.46; Her2-type: OR = 2.53, 95%<br />

CI = 0.92 to 6.36).<br />

Conclusions In the present study, significant heterogeneity of<br />

associations between BMI and tumor subtypes was observed. <strong>Breast</strong><br />

cancer subtypes may have different etiologies associated with each<br />

subtype.<br />

P3-09-05<br />

The role of breast size in detection of breast cancer in asian women<br />

Lee S, <strong>San</strong>dhu H, Zheng Y. Holy Name Medical Center<br />

Introduction. Asian women typically have smaller breast sizes than<br />

those of Caucasian women and a recent survey revealed a common<br />

belief in the Korean community that smaller breast means lesser risk<br />

of developing breast cancer. However, since breast density plays<br />

a strong role in breast cancer risk in women with smaller breasts,<br />

Korean women who follow the trend of higher breast density may<br />

have increased risk of breast cancer, with smaller breast size.<br />

Materials and Methods. Mammograms from 2,050 Korean American<br />

women were obtained from Holy Name Medical Center during the<br />

years 2008 to 2011. The breast density was analyzed and compared to<br />

that reported for Caucasian women. The role of smaller breast size was<br />

further evaluated by randomly selecting 85 patients whose breast sizes<br />

were obtained by using image analysis of the mammograms using<br />

ImageJ, an image processing software developed by the National<br />

Institute of Health.<br />

Results. The data from 2050 Korean American women revealed<br />

that Korean American women have denser breast tissue than those<br />

reported for Caucasian women. In addition, 61% of these women<br />

who developed breast cancer were in high (BI-RADS 3 or 4) breast<br />

density groups. The high breast density was associated with the<br />

smallest average breast size and 50% of breast cancer patients fell<br />

into the smallest breast size category.<br />

Conclusion. This study shows that Korean women with smaller breast<br />

size follow the trend of higher breast density, and therefore increased<br />

risk of breast cancer. The association of the risk of breast cancer with<br />

smaller breast size and higher breast density needs to be addressed<br />

when counseling Asian women in the prevention of breast cancer.<br />

P3-10-01<br />

Epidemiological risk factors and normal breast tissue markers<br />

in inflammatory breast cancer<br />

Atkinson RL, Sexton KR, Ueno NT, El-Zein R, Brewster AM,<br />

Krishnamurthy SA, Woodward WA. University of Texas MD Anderson<br />

<strong>Cancer</strong> Center, Houston, TX; Dan L Duncan <strong>Cancer</strong> Center, Baylor<br />

College of Medicine, Houston, TX<br />

Background: Inflammatory breast cancer (IBC) is a rare form of<br />

aggressive breast cancer with no existing identifiers for screening or<br />

prevention strategies. Women with triple-negative (TNBC, ER–,PR–<br />

,Her2–) non-inflammatory breast cancer are less likely to breastfeed,<br />

and we have shown in adjacent normal breast tissue that this tissue<br />

has more foci of stem cells compared to non-TNBC cancers. A<br />

disproportionately higher percentage of women with IBC have TNBC<br />

relative to women with non-IBC. We hypothesized that adjacent<br />

normal tissue in TNBC IBC vs. TN non-IBC may also display unique<br />

biological features based on epidemiologic characteristics. Methods:<br />

We examined epidemiologic factors by breast cancer receptor subtype<br />

in 144 patients diagnosed with IBC in 1991–2011 at MD Anderson<br />

<strong>Cancer</strong> Center. <strong>Breast</strong> cancer risk factors including parity and<br />

breastfeeding were compared between patients with TN and non-TN<br />

IBC with chi-square or Wilcoxon rank sum tests. Normal adjacent<br />

tissues were stained for stem cell markers CD44+CD49f+CD133/2+<br />

and macrophage marker CD68. Results: The mean age at diagnosis<br />

was 52.3 years (range=23-80) and 83% of patients were non-Hispanic<br />

white, 80% were overweight or obese (BMI >25), and 36% were<br />

TN IBC. Patients with TN IBC had significantly lower frequency of<br />

breastfeeding compared with non-TN IBC, 28% vs. 55%, (p=0.01).<br />

No differences were found in the frequency of other breast cancer risk<br />

factors. All 8 IBC adjacent tissue samples showed a distinct spatial<br />

distribution of stem cell staining, not limited to the triple negative<br />

patients. Compared with 0/60 non-IBC cases, 0/8 triple negative<br />

non-IBC, (p=0.001 Pearson chi-square). Given the high BMI among<br />

IBC patients, we further examined normal tissues for the presence<br />

of CD68+ cells distributed individually or as clusters exhibiting a<br />

“crown-like” pattern (multiple CD 68+ macrophages found around<br />

dead adipocytes), and found that 7 of the 8 IBC adjacent tissues<br />

were CD68+. Benign biopsies collected from 2 patients at 10 years<br />

before diagnosis displayed similar staining, including both stem cell<br />

and CD68 staining. Compared with 12/60 non-IBC adjacent tissues<br />

were positive for CD68, with 1/8 TN non-IBC, (p=0.001 Pearson<br />

chi-square). Conclusion: We describe for the first time a stem-cell<br />

staining pattern unique to IBC present in all IBC tissues examined,<br />

including pre-cancer biopsies. Tissue samples from additional patients<br />

will be examined to further explore the relationship between stem<br />

cells and CD68 positivity with IBC subtypes.<br />

P3-10-02<br />

Modulation of mRNA and miRNAs by the novel histone<br />

deacetylase inhibitor, CG-1521 disrupts cytokinesis in SUM149PT<br />

inflammatory breast cancer cells<br />

Chatterjee N, Tenniswood MPR. University at Albany, Rensselaer, NY<br />

Inflammatory breast cancer (IBC) comprises only 2.0% of all breast<br />

cancer cases; however it accounts for 8.0% of breast cancer-specific<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

334s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 3<br />

deaths. IBC is highly aggressive, appears at a younger age, and is<br />

more difficult to treat than the common forms of breast cancer. By<br />

the time the disease is diagnosed, IBC is usually stage 3 and has<br />

already metastasized to neighboring lymphatics. Though many tumors<br />

are p53 wild type and/or Her2 positive, the median overall survival<br />

among women with IBC is less than 4 years even with aggressive<br />

multimodal therapy. To develop IBC-specific therapies, we have<br />

compared the effects of two hydroxamic acid histone deacetylase<br />

(HDAC) inhibitors, Trichostatin A (TSA) and CG-1521 on the biology<br />

of the triple negative SUM149PT IBC cell line. In these cells, CG-<br />

1521 and TSA induce dose (0-10 µM) and time dependent (0-96 h)<br />

increases in the proportion of cells undergoing cell cycle arrest and<br />

cell death, in the presence or absence of 17β-estradiol. To identify<br />

the genomic targets of CG-1521, we have performed concurrent<br />

microarray analyses of mRNA and miRNA expression followed<br />

by qPCR validation. In SUM149PT cells, CG-1521 modulates the<br />

expression of approximately 935 transcripts: 341 of these transcripts<br />

are up-regulated and 594 are down-regulated after 48h of treatment.<br />

Gene ontology analysis demonstrates that many of the down-regulated<br />

mRNA transcripts encode proteins that are associated with the spindle<br />

assembly checkpoint, chromosome segregation and microtubule based<br />

processes including KIF4, PRC1, AURKB and KIF23 (MKLP-1).<br />

Strikingly, many genes in these ontologies are potentially targeted<br />

by a small number of miRNAs (miR-22, miR-1207-5p & miR-494)<br />

which are significantly up-regulated by CG-1521, suggesting that the<br />

ability of CG-1521 to down-regulate the transcripts associated with<br />

spindle formation and cytokinesis may be significantly modulated by<br />

miRNA expression. Immunofluorescence microscopy demonstrates<br />

that treatment of SUM149PT cells with CG-1521 results in the<br />

appearance of elongated midzone structures visualized by phalloidin<br />

staining, similar to the phenotype seen by others after siRNAmediated<br />

knockdown of KIF4 in HeLa cells. These data suggest<br />

that CG-1521 affects the central mitotic spindle formation, disrupts<br />

furrow formation and abscission, arresting the cells in cytokinesis<br />

ultimately leading to cell death. Whether these effects are solely<br />

mediated through transcriptional mechanisms or whether CG-1521<br />

also directly affects the activity of these proteins through acetylation<br />

remains to be determined. Nevertheless, the data show that histone<br />

deacetylase inhibitors induce cell death in IBC cells, and suggest that<br />

these small molecules may be useful addition to the armamentarium<br />

as adjuvants for the treatment of IBC.<br />

P3-10-03<br />

Bartonella henselae Infection Detected in Patients with<br />

Inflammatory <strong>Breast</strong> <strong>Cancer</strong>.<br />

Fernandez SV, Aburto L, Maggi R, Breitschwerdt EB, Cristofanilli M.<br />

Fox Chase <strong>Cancer</strong> Center, Philadelphia, PA; North Caroline State<br />

University, Raleigh, NC<br />

Inflammatory breast cancer (IBC) is a very aggressive type of<br />

advanced breast cancer with a poor prognosis. Clinical symptoms<br />

involve a rapid onset of changes in the skin overlying the breast,<br />

including edema, redness and swelling including a wrinkled and<br />

orange peel appearance in the skin. This particular presentation is<br />

due to the invasion of the skin dermal lymphatics by breast cancer<br />

cells that obstructed the lymph channels producing the characteristic<br />

skin changes that mimic an inflammatory process. Mouse Mammary<br />

Tumor-associated Virus (MMTV) and other infectious agents have<br />

been consider as possible etiological agents of IBC particularly<br />

related to the initial description of higher incidence in women living<br />

in rural areas in North Africa. Although, the etiopathological role<br />

of bacteria in this disease has never been explored in spite of the<br />

evidence that chronic infections with certain bacteria can facilitate<br />

tumors development.<br />

Several bacterial infections promote cell proliferation and could<br />

increase the rate of cell transformation. Bartonella spp. is an emerging<br />

pathogen that can cause conditions which are characterized by<br />

the development of proliferative lesions. Bartonella might cause<br />

vasculoproliferative disorders by triggering the proliferation of<br />

endothelial cells and inducing the secretion of proliferative cytokines<br />

from infected host cells. It cause persistent infection of erythrocytes<br />

and endothelial cells in their mammalian hosts and their transmission<br />

occur mainly by blood-sucking arthropods. Bartonella are Gramnegative<br />

bacteria usually associated with cat-scratch disease, urban<br />

trench fever, bacillary angiomatosis-peliosis and endocarditis. Some<br />

reports have showed some similarities between cat scratch disease and<br />

inflammatory breast cancer (Povoski et al., <strong>Breast</strong> J 9: 497-500, 2003).<br />

Methods and Results: In the present work, we report the identification<br />

of Bartonella henselae in the pleural effusions of two patients with<br />

IBC. These patients had both estrogen-receptor negative (ER-) and<br />

progesterone-receptor negative (PR-) disease and have failed a<br />

number of previous chemotherapy regimens. Furthermore, they had<br />

metastatic disease to the pleura, skin, lymph nodes and lung. Cells<br />

from the pleural fluids of these patients stained positive for Bartonella<br />

sp. using the Warthin-Starry staining kit. Moreover, we used PCR and<br />

sequencing of the 16S-23S intergenic spacer (ITS) region to identify<br />

the etiological agent as Bartonella henselae. By immunofluorescence<br />

using an antibody that specifically recognized Bartonella henselae,<br />

the bacteria were found in the nucleus of IBC tumor cells confirming<br />

the infection and their intracellular localization.<br />

Conclusions: These results suggested that bacteria infections such<br />

as the one produced by Bartonella henselae might contribute to the<br />

clinical and pathological characteristics of IBC, associated with rapid<br />

spread of the breast tumor cells through the lymphatic system. The<br />

infection and activation of endothelial lymphatics by Bartonella might<br />

contribute to the highly metastatic process observed in IBC patients.<br />

An acute inflammatory reaction triggered by the Bartonella infected<br />

endothelium may be crucial for initiating the chronic inflammation<br />

in IBC patients and the rapid spread of tumor cells.<br />

P3-10-04<br />

Regulation of inflammatory breast cancer cell invasion through<br />

Akt1/PKBα phosphorylation of RhoC GTPase<br />

Lehman HL, Van Laere SJ, van Golen CM, Vermeulen PB, Dirix<br />

LY, van Golen KL. The University of Delaware, Newark, DE; Sint<br />

Augustine Hospital, Antwerp, Belgium; Catholic University, Leuven,<br />

Belgium; Delaware State University, Dover, DE<br />

With a 42% and 18% 5- and 10-year respective disease-free survival<br />

rate, inflammatory breast cancer (IBC) is arguably the deadliest<br />

form of breast cancer. IBC invades the dermal lymphatic vessels<br />

of the skin overlying the breast and as a consequence nearly all<br />

women have lymph node involvement and ∼1/3 have gross distant<br />

metastases at the time of diagnosis. One year after diagnosis ∼90%<br />

of patients have detectable metastases, making IBC a paradigm for<br />

lymphovascular invasion. Understanding the underlying mechanisms<br />

of the IBC metastatic phenotype is essential for new therapies. Work<br />

from our laboratory and others show distinct molecular differences<br />

between IBC and non-inflammatory breast cancers. Previously we<br />

demonstrated that RhoC GTPase is a metastatic switch responsible<br />

for the invasive phenotype of IBC. In the current study we integrate<br />

observations made in IBC patients with in vitro analysis. We<br />

demonstrate that the PI3K/Akt signaling pathway is crucial in IBC<br />

www.aacrjournals.org 335s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

invasion. Key molecules involved in cytoskeletal control and cell<br />

motility are specifically upregulated in IBC patients compared with<br />

stage and cell-type-of-origin matched non-inflammatory breast cancer<br />

patients. Distinctively, RhoC GTPase is a substrate for Akt1 and its<br />

phosphorylation is absolutely essential for IBC cell invasion. Further<br />

our data show that Akt3, not Akt1 has a role in IBC cell survival.<br />

Together our data demonstrate a unique and targetable pathway for<br />

IBC invasion and survival.<br />

P3-10-05<br />

Response to neoadjuvant systemic therapy (NST) in inflammatory<br />

breast cancer (IBC) according to estrogen receptor (ER) and<br />

HER2 expression.<br />

Masuda H, Iwamoto T, Brewer T, Hsu L, Kai K, Woodward WA,<br />

Reuben JM, Valero V, Alvarez RH, Willey J, Hortobagyi GN, Ueno<br />

NT. Morgan Welch Inflammatory <strong>Breast</strong> <strong>Cancer</strong> Research Program<br />

and Clinic, The University of Texas MD Anderson <strong>Cancer</strong> Center,<br />

Houston, TX; The University of Texas MD Anderson <strong>Cancer</strong> Center,<br />

Houston, TX; Okayama University Hospital, Okayama, Japan<br />

Background: Inflammatory breast cancer (IBC) is the most aggressive<br />

form breast cancer. NST, followed by local therapy (surgery and<br />

radiation therapy), is considered the current standard therapy for IBC.<br />

Among noninflammatory breast cancers, sensitivity to NST differs<br />

based on ER and HER2 status. However, whether the sensitivity<br />

to NST also differs in primary IBC based on ER status or other<br />

prognostic factors has not been studied in a large cohort.<br />

Methods: We retrospectively reviewed 1078 patients (pts) newly<br />

diagnosed with IBC from April 1989 to January 2011. Of these, 838<br />

pts met our inclusion criterion of stage III disease at diagnosis, and 713<br />

of these pts had received NST and surgery. Among this population,<br />

545 pts had information available on both ER and HER2 status. We<br />

compared pathological complete response (pCR) rates (defined as<br />

no evidence of invasive disease in the breast and ipsilateral axillary<br />

limph nodes) and clinical characteristics between ER and HER2-status<br />

subgroups and analyzed their clinical outcome. We used the Kaplan-<br />

Meier method to estimate the median recurrence-free survival (RFS)<br />

after surgery and overall survival (OS), and the Cox proportional<br />

hazards regression model to test the statistical significance of potential<br />

prognostic factors in each group.<br />

Results: Overall 177 pts had ER+HER2- tumors; 75, ER+HER2+; 134,<br />

ER-HER2+; and 159, ER-HER2-. NST consisted of anthracyclinebased<br />

[A] alone, a taxane [T] alone or with A+T; HER2 targeting<br />

therapies (H) were administered to 117 patients with HER2-positive<br />

breast cancer after 1998. Overall pCR rate was 14.7%. pCR rates<br />

are shown by marker subtype and NST received in the table below.<br />

pCR rate, nuclear grade, vascular invasion, clinical response to NST,<br />

adjuvant treatment, radiation therapy, and adjuvant hormonal therapy<br />

differed significantly among subgroups.<br />

The median RFS and OS for all patients was 19.2 and 33.2 months,<br />

respectively. In multivariate analysis, BMI, ER status, lymphatic<br />

invasion, radiation therapy, and pCR rate were associated with<br />

RFS, and ER status, vascular invasion, radiation therapy, and pCR<br />

rate were associated with OS. Except in the ER+HER2- group,<br />

pCR was associated with better prognosis compared to non-pCR.<br />

Adjuvant hormonal therapy improved RFS both in ER+HER2+ and<br />

ER+HER2- groups, but did not improve OS in the ER+HER2+ group.<br />

Among 209 patients with HER2+ IBC, 134 received HER2 targeting<br />

therapies in neoadjuvant or adjuvant chemotherapy, and had a trend<br />

to improvement in RFS compared to chemotherapy alone (P=0.082).<br />

The ER-HER2- group showed poorest outcome compared to other<br />

subgroups (P


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 3<br />

P3-10-07<br />

Lymph node status and survival in inflammatory breast cancer<br />

Sieffert MR, Pedersen RC, Tereffe W, Cui H, Woods RR, Viscusi<br />

RK, LeBeau-Grasso L, Lang JE. University of Arizona College of<br />

Medicine, Tucson, AZ; University of Texas MD Anderson <strong>Cancer</strong><br />

Center, Houston, TX; University of Arizona <strong>Cancer</strong> Center, Tucson,<br />

AZ; University of Arizona Health Sciences Center, Tucson, AZ;<br />

University of Southern California Norris Comprehensive <strong>Cancer</strong><br />

Center, Los Angeles, CA<br />

Objectives: Positive lymph node status in breast cancer is known<br />

to be associated with poor outcomes when compared with nodenegative<br />

disease, but the effect of lymph node status on outcomes in<br />

inflammatory breast cancer (IBC) has not been evaluated. This study<br />

was designed to investigate the association between lymph-node status<br />

and overall survival (OS) in individuals with inflammatory breast<br />

cancer using prospective data from the Surveillance, Epidemiology,<br />

and End Results (SEER) database.<br />

Methods: We identified 750 patients in the April 2012 edition of the<br />

SEER 17 registry who had non-metastatic IBC diagnosed from 1973-<br />

2008. Patients were included only if they met a stringent definition<br />

of IBC (ICD-O-2 morphology code 8503) and their pathologic nodal<br />

status was known. Patients who did not receive mastectomy as part<br />

of their local therapy were excluded, to minimize the likelihood of<br />

inadvertently including patients with metastatic disease at or shortly<br />

after diagnosis. A total of 711 patients were deemed evaluable for<br />

analysis (145 node-negative, 566 node-positive). Survival analysis<br />

was performed using the Kaplan–Meier method. Cox proportional<br />

hazard regression was performed to evaluate univariate and<br />

multivariate associations between treatment and OS. Information<br />

regarding receipt of systemic therapy and human epidermal growth<br />

factor receptor 2 (HER2) status was not available in this version of<br />

the SEER database.<br />

Results: Positive lymph node status was associated with a significant<br />

decrease in OS (p=0.01) when compared with node negative status.<br />

In lymph node-positive patients, ER or PR positivity was associated<br />

with better OS than ER or PR negativity (adjusted HR 0.56 (p=0.005)<br />

for ER+ vs. ER-, and adjusted HR 0.55 (p=0.009) for PR+vs. PR-).<br />

In patients with positive lymph nodes, the combination of surgery<br />

and radiation therapy improved overall survival when compared with<br />

surgery alone (adjusted HR 0.56, p=0.003). In node-negative patients,<br />

the combination of surgery and radiation therapy was not clearly<br />

superior to surgery alone, possibly due to the rarity of node-negative<br />

IBC (adjusted HR 0.53, 95% CI 0.23-1.19, p=0.13).<br />

Conclusions: Our findings provide a better understanding of the<br />

characteristics of inflammatory breast cancer, and creates the<br />

opportunity for future studies to evaluate the prognostic significance<br />

and treatment implications of lymph node status in inflammatory<br />

breast cancer. Our study is limited by lack of knowledge of use of<br />

systemic therapy, the HER2 status for each patient, and information<br />

regarding locoregional recurrence. However, institutions participating<br />

in the SEER registry are likely to follow guidelines set forth by the<br />

National Comprehensive <strong>Cancer</strong> Network, recommending induction<br />

chemotherapy followed by surgery followed by radiation for IBC.<br />

Nearly 80% of the IBC patients included in this study had nodal<br />

metastasis, reflecting the inherently aggressive biology of this disease.<br />

Further studies are required to characterize the biology of IBC and<br />

guide the optimal treatment of this disease.<br />

P3-10-08<br />

A new in vitro method of growing and studying inflammatory<br />

breast cancer emboli<br />

Lehman HL, Daasner EJ, Vermeulen PB, Dirix LY, Van Laere S, van<br />

Golen KL. The University of Delaware, Newark, DE; Sint Augustine<br />

Hospital, Antwerp, Belgium; Catholic University, Leuven, Belgium<br />

Inflammatory breast cancer (IBC) is the deadliest form of breast<br />

cancer, presenting as intralymphatic emboli. Emboli within the dermal<br />

lymphatic vessels are thought to contribute to rapid metastasis. The<br />

lack of appropriate in vitro models has made it difficult to accurately<br />

study how IBC emboli metastasize. To date, attempts at creating IBC<br />

tumor emboli in vitro have used 3-dimensional culture on a solid layer<br />

of MatrigelTM, which does not resemble the physical properties of<br />

the lymphatic system. Dermal lymphatic fluid produces oscillatory<br />

fluid shear forces and is 1.5-1.7-fold more viscous than water with<br />

a pH range of 7.5-7.7. We have established a method for forming<br />

tumor emboli by culturing the IBC cell lines in suspension with<br />

either polyethylene glycol- or hyaluronic acid-containing medium<br />

and oscillatory fluid shear forces. Non-IBC cells do not form emboli<br />

under identical conditions. In vitro IBC emboli were analyzed for<br />

expression of markers associated with patient emboli and their ability<br />

to undergo amoeboid movement. In a direct comparison, the in vitro<br />

IBC emboli closely resemble IBC patient emboli with respect to<br />

size, composition and E-cadherin expression. Further, cells from the<br />

emboli are able to invade in clusters via RhoC GTPase-dependent<br />

amoeboid movement. Invasion by clusters of IBC cells is disrupted<br />

by exposure to TGFβ. This study provides a biologically relevant in<br />

vitro model to accurately grow and study inflammatory breast cancer<br />

biology and metastasis.<br />

P3-10-09<br />

Peptide-based molecular targeting of inflammatory breast cancer.<br />

Eckhardt BL, Miao RY, Cao Y, Driessen WH, Krishnamurthy S, Arap<br />

W, Ueno N, Anderson RL, Pasqualini R. The University of Texas at<br />

MD Anderson <strong>Cancer</strong> Center, Houston, TX; Stanford University<br />

School of Medicine; Trescowthick Research Laboratories, Peter<br />

MacCallum <strong>Cancer</strong> Center; The University of Texas at MD Anderson<br />

<strong>Cancer</strong> Center<br />

Inflammatory breast cancer (IBC) is a subtype of breast cancer<br />

that has a frequent association with metastatic disease and a poorer<br />

prognosis than comparative non-inflammatory breast cancers. While<br />

IBC is now considered a distinct subclass of breast cancer, the lack<br />

of molecular characterization, both at the genomic and proteomic<br />

levels, has hampered the development of rationalized and targeted<br />

therapies. Inherent receptor/ligand interactions that can occur on<br />

the surface of tumor cells can act as a dynamic molecular address<br />

that can enable targeted delivery of drugs and imaging agents to<br />

tumors. We hypothesize that such molecular addresses within IBC<br />

can be exploited for ligand-based imaging and early detection of<br />

disease sites. To this end, it is our goal to generate targeted imaging<br />

and therapeutic agents by combining ligand-directed targeting with<br />

efficient transduction of IBC cells by hybrid gene delivery vectors.<br />

Our strategy utilizes a hybrid vector with genomic elements from<br />

adeno-associated virus (AAV) and an M13-derived phage. Ligandtargeted,<br />

AAV/phage (AAVP) chimeras can display tumor-homing<br />

peptides that mediate selective internalization of viral particles<br />

through specific ligand-receptor interactions in vitro and in vivo. Such<br />

targeted vehicles are suited for the delivery of different reporter genes<br />

that can be used for imaging, diagnosis and therapy of breast cancer<br />

As a part of our ongoing studies we have identified, characterized<br />

and evaluated a peptide (WIFPWIQL, amino acid sequence) that<br />

www.aacrjournals.org 337s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

can target GRP78, a stress-response protein that is expressed in<br />

IBC tumors and elevated during metastatic progression. Indeed, we<br />

found that this GRP78-targeting peptide can bind to, and internalize<br />

within IBC cells. As a result, we sought to characterize the ability of<br />

this peptide to mediate the delivery of fluorescent-based compounds<br />

and toxic moieties in preclinical models of IBC and breast cancer<br />

metastasis. Using amine-based chemical coupling, we conjugated<br />

near-infrared dyes on both WIFPWIQL-phage and on a WIFPWIQLpeptide<br />

engrafted antibody. When these fluorescent construct<br />

were administered into mice bearing IBC or IBC-like tumors, we<br />

could visualize tumor-specific targeting of the vectors in vivo. To<br />

demonstrate efficacy of GRP78-targeted therapeutics, we conjugated<br />

the tumor-homing, GRP78 ligand to a cell-death inducing domain<br />

(creating a compound called BMTP-78, bone metastasis targeting<br />

peptide-78), which can selectively kill cells upon internalization. We<br />

show here that BMTP78 therapy in mice with established GRP78-<br />

positive tumors, but not matched GRP78-negative tumors, could<br />

effectively reduce tumor growth and metastatic burden. Finally, we<br />

demonstrate a WIFPWIQL-AAVP construct that expresses a suicide<br />

gene (HSVtk) under the control of either CMV or GRP78 promoter,<br />

could sensitize IBC tumor xenografts to the pro-drug ganciclovir.<br />

Collectively, our results demonstrate an in vivo receptor/ligand system<br />

that has the potential for imaging and therapeutic targeting of IBC<br />

and aggressive breast tumors.<br />

P3-10-10<br />

STATUS OF ANAPLASTIC LYMPHOMA KINASE (ALK)<br />

GENE IN INFLAMMATORY BREAST CARCINOMA<br />

Krishnamurthy S, Woodward W, Reuben JM, Tepperberg J, Ogura<br />

D, Niwa S, Ueno NT. MD Anderson <strong>Cancer</strong> Center, Houston, TX;<br />

LabCorp, Durham, NC; Link Genomics, Toyoka, Japan<br />

The ALK gene located on chromosome 2p23 belongs to the insulin<br />

receptor superfamily and encodes a receptor tyrosine kinase that<br />

is normally expressed in brain, testis and small intestine. There is<br />

limited data regarding ALK gene rearrangements in breast cancers. We<br />

performed a comprehensive investigation of ALK in IBC using array<br />

comparative genomic hybridization (CGH), transcriptional profiling,<br />

fluorescence in situ hybridization (FISH) and immunohistochemistry<br />

(IHC) for ALK protein expression.<br />

Materials and Methods: We used formalin fixed and paraffin<br />

embedded core biopsies of IBC for IHC and FISH and microdissected<br />

frozen tissues for array CGH and mRNA analysis. ALK protein<br />

was evaluated by IHC using primary antibody against ALK<br />

(DAKO cytomation). The Vysis ALK Break Apart FISH probe kit<br />

(Abbott Laboratories) was used to detect chromosome 2p23 gene<br />

rearrangements. Genomic DNA was applied to whole genome DNA<br />

microarray (Link Genomics) which covered the entire human genome<br />

using 12310 individually amplified bacterial artificial chromosome<br />

clones (BAC) clones. Thresholds of gains and losses were set at 1.2<br />

and 0.8 respectively. Analysis of mRNA expression was performed by<br />

labeling cRNA from the specimens to an oligo-nucleotide microarray<br />

(whole genome 4x44K Agilent technologies), scanned with an Agilent<br />

microarray scanner and analyzed with features extraction software<br />

(Agilent Technologies).<br />

Results: We performed IHC and FISH for ALK in 29 IBC patients<br />

(ER/PR +, HER2- in 12, HER2+ in 13, triple negative in 4). Array<br />

CGH and mRNA analysis was conducted in 20 of these 29 cases. All<br />

the 29 cases were negative for ALK protein expression by IHC. FISH<br />

analysis revealed intact bi-color flanking signals in all the tumor cell<br />

nuclei from the 29 specimens indicating absence of EML4-ALK gene<br />

rearrangements in any of the cases. We found mild gain of intact ALK<br />

signals in a small percentage of interphase cells in 90% of the cases.<br />

FISH for CEP2 performed in 8 specimens showed normal CEP2<br />

levels in 6/8 indicating mild amplification of the ALK gene in these<br />

cases. A total of 20 DNAs analyzed by array CGH focusing on the 2<br />

BACs that covered the ALK gene region showed no gain in 17 cases<br />

indicating no change in copy number of ALK, gain in 1 BAC in 2<br />

cases and gain of 2 BACS in 1 case. Transcriptional profiling revealed<br />

that mRNA expression of ALK was not significantly higher than the<br />

5 normal breast controls (P >0.05, t test) including the 3 patients with<br />

possible mild increase in copy numbers of ALK gene by array CGH.<br />

Conclusion: Gene rearrangements involving the ALK gene were not<br />

noted in any of the IBC patients. Mild gain of intact ALK signals<br />

as evidenced by FISH suggests low level amplification in majority<br />

of IBC patients. The lack of gain in copy numbers of ALK gene in<br />

85% of the IBC patients together with the absence of significant<br />

elevation of mRNA levels and absence of ALK protein expression<br />

by IHC suggests absence of transcription and translation of the mild<br />

amplification of the ALK gene in patients with IBC.<br />

P3-11-01<br />

Matched-pair analysis of patients with male and female breast<br />

cancer<br />

Kim M, Lee SK, Choi M-Y, Kim S, Kim J, Jung SP, Bae SY, Kil WH,<br />

Lee JE, Nam SJ. Samsung Medical Center, Sungkyunkwan University<br />

School of Medicine, Seoul, Republic of Korea<br />

Purpose: Male breast cancer (MBC) is extremely rare, accounting for<br />

less than 1% of all malignancies in men and only 1% of all breast<br />

carcinomas. The treatment and surveillance guidelines on male breast<br />

cancers are less recognized. The aim of this study is to evaluate our<br />

single institution’s experience with MBC over the past 15 years and<br />

to contrast differences between female and MBC.<br />

Methods: MBC diagnosed from 1994 to 2010 at the Department of<br />

Surgery, Samsung Medical Center (Seoul, Korea) was retrospectively<br />

analyzed. Clinical data and tumor characteristics were examined. Each<br />

MBC was matched with female counterparts by 1:N varied matching<br />

ratio that showed accordance in seven variables (year of diagnosis,<br />

age, tumor stage, nodal stage, tumor grade, estrogen receptor(ER),<br />

progesterone receptor(PR)).<br />

Results: 39 male/184 female matched-pairs were available for<br />

analysis. The median duration of follow-up was 3.8 years. The median<br />

age of MBC patients was 50 years and the median size of tumor was<br />

2.0cm. The proportion of positivity of ER and PR status was 97.4%<br />

and 84.6%, respectively. Despite of higher positive rate of hormone<br />

receptor, the rate of hormone therapy in MBC patients was significant<br />

lower than female conterpart (p=0.002). Men and women with breast<br />

cancer had similar disease-free survival (DFS) and disease-specific<br />

overall survival (DSS). Five MBC patients had a recurrence during<br />

follow up period and 4 of them were expired. The 10-years DFS was<br />

73.1% in men and 80.5% in women (p=0.348). The 10-years DSS<br />

was 74.1% in men and 87.1% in women, respectively (p=0.207).<br />

Conclusion: This study showed no disease free and overall survival<br />

differences between male and female breast cancer patients and<br />

revealed that gender is no predictor for survival in breast cancer. Male<br />

patients receive obviously less adjuvant treatment compared their<br />

female matched patients. It would be better to do more aggressive<br />

treatment in MBC to improve the survival outcome.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

338s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 3<br />

Table1. Matching criteria of male and female breast cancer patients<br />

Male (n=39) Female (n=184) p<br />

Median age (range) 50 (33-74) 50 (33-75) 0.936<br />

Tumor stage pTis 4 10.3% 13 7.1%<br />

pT1 23 59.0% 111 60.3%<br />

pT2 12 30.8% 60 32.6% 0.789<br />

Nodal stage pN0 21 53.8% 99 53.8%<br />

pN1 8 20.5% 40 21.7%<br />

pN2 7 17.9% 31 16.8%<br />

pN3 2 5.1% 10 5.4%<br />

pNx 1 2.6% 4 2.2% 1.000<br />

Tumor grade G1 11 28.2% 46 25.0%<br />

G2 23 59.0% 113 61.4%<br />

G3 5 12.8% 25 13.6% 0.916<br />

ER-expression ER- 1 2.6% 1 0.5%<br />

ER+ 38 97.4% 183 99.5% 0.320<br />

PR-expression PR- 6 15.4% 26 14.1%<br />

PR+ 33 84.6% 158 85.9% 0.839<br />

Table 2. Not-matched tumor characteristics and treatment features of<br />

Male (n=39)<br />

Female<br />

(n=184)<br />

p<br />

Histology DCIS 4 10.3% 13 7.1%<br />

Invasive ductal carcinoma 28 71.8% 154 83.7%<br />

Invasive lobular carcinoma 0 0% 3 1.6%<br />

Mucinous carcinoma 6 15.4% 6 3.3%<br />

Others* 1 2.6% 8 4.3% 0.109<br />

HER2 negative 34 87.2% 165 89.7%<br />

positive 3 7.7% 18 9.8%<br />

unknown 2 5.1% 1 0.5% 0.119<br />

Operation Mastectomy 36 92.3% 68 37.0%<br />

<strong>Breast</strong> conserving surgery 3 7.7% 116 63.0% < 0.001<br />

Adjuvant<br />

radiotherapy<br />

12 30.8% 127 69.0% < 0.001<br />

Adjuvant<br />

systemic Chemotherapy 24 61.5% 125 67.9% 0.550<br />

therapy<br />

Hormone therapy 32 82.1% 177 96.2% 0.002<br />

* apocrine carcinoma, tubular carcinoma, micropapillary carcinoma, mixed<br />

P3-11-02<br />

Male breast cancer: A comparison between BRCA mutation<br />

carriers and non-carriers in Hong Kong, Southern China<br />

Kwong A, Chau WW, Wong CHN, Law FBF, Ng EKO, Suen DTK,<br />

Kurian AW, West DW, Ford JM, Ma ESK. University of Hong Kong,<br />

Pokfulam, Hong Kong; Hong Kong Hereditary <strong>Breast</strong> <strong>Cancer</strong> Family<br />

Registry, Happy Valley, Hong Kong; Stanford University School of<br />

Medicine, Stanford, CA; Hong Kong <strong>San</strong>itorium & Hospital, Happy<br />

Valley, Hong Kong<br />

Background: Male breast cancer is suggested to be biologically<br />

different from female breast cancer. The differences in clinicopathology<br />

between male and female breast cancer raise the issues of establishing<br />

specific strategies and treatment regime for male breast cancer<br />

patients. The single most significant risk factor for male breast<br />

cancer is a mutation in the BRCA2 gene. The lack of information<br />

on hereditary breast cancer in male, particularly in Asians, leaves<br />

great but forgiven research area on epidemiological studies for this<br />

group of patients.<br />

Methods: All male breast cancer patients and their family members,<br />

from a Hong Kong Hereditary and High Risk <strong>Breast</strong> <strong>Cancer</strong> Program<br />

since year 2007, were recruited in this study. All received genetic<br />

counseling and BRCA mutation testing using DNA extracted from<br />

blood samples. A questionnaire was administered at their first visit<br />

which included questions on their demographics and socioeconomic<br />

status. Other information including family history of breast cancer<br />

or other kinds of cancer, method of diagnosis, surgical strategies,<br />

pathological results, treatment regime, relapse, metastasis, and<br />

outcomes were obtained from their medical records. Descriptive<br />

analysis was performed describing the background characteristics.<br />

Chi-square test and Student’s t-test were applied to calculate the<br />

associations between BRCA mutation and risk factors. Survival<br />

analysis was performed to look for their survival patterns.<br />

Results: Thirty-six male breast cancer patients were recruited between<br />

year 2007 and 2012, while 21 were diagnosed before year 2007 (range:<br />

1996 to 2012). Mean, standard deviation, and median follow-up time<br />

were 5.75±4.31 and 5.25 years. Seven were found to carry the BRCA<br />

mutation. All were BRCA2 mutation and the mutation rate was 19.4%<br />

(N = 7). Family history of cancer was found in 52.8% (N = 19). Male<br />

BRCA mutation carriers were found to have higher risk of secondary<br />

cancer, and their first and second degree family members had higher<br />

risk of either breast cancer or other kinds of cancers. T stage in BRCA<br />

patients was significantly higher than non-BRCA patients (p = 0.028).<br />

All BRCA mutation carriers had ER positive cancers compared with<br />

96.2% who were non-carriers. Half of the male BRCA patients were<br />

PR positive compared with higher percentage in non-BRCA patients<br />

(50% vs. 80.8%, p = 0.117). Both groups had similar overall (p =<br />

0.962) and disease-free survivals (p = 0.919). The means and standard<br />

deviations of 5-year overall survival between BRCA and non-BRCA<br />

patients were 2.08±0.25 and 4.24±0.12 years respectively, and<br />

2.08±3.03 and 4.41±1.46 years for disease-free survival.<br />

Conclusions: The prevalence of male breast cancer patients with<br />

BRCA2 mutation in Hong Kong is comparable with other similar<br />

studies. Male breast cancer patients with BRCA2 mutation are<br />

suspected to have higher chance of secondary cancer and familial<br />

cancer. Although percentage of ER positive cancers are similar to the<br />

two groups, BRCA2 mutation carriers tend to have less PR positive<br />

cancers which may suggest a poorer prognosis although due to a small<br />

sample size this cannot be shown in this cohort. Further collaborative<br />

studies to better understand male breast cancer patients carrying the<br />

BRCA mutation is warranted.<br />

P3-11-03<br />

Treatment outcomes for early stage male breast cancer: a single<br />

centre retrospective case-control study<br />

Kwong A, Visram H, Graham N, Balchin K, Petrcich W, Dent S. The<br />

Ottawa Hospital, Ottawa, ON, Canada; Ottawa Hospital Research<br />

Institute, Ottawa, ON, Canada<br />

Background:<br />

Male breast cancer (BC) is a rare disease with a reported incidence of<br />

1%, and treatment strategies are therefore driven by studies conducted<br />

in the female BC population. There is also limited information<br />

with regards to treatment outcomes for men with early stage BC,<br />

particularly when compared with their female counterparts. The<br />

objective of this study is to compare the disease free survival (DFS)<br />

and overall survival (OS) of males with early stage BC compared to<br />

age and stage matched females in a single institution.<br />

Methods:<br />

A retrospective matched case-control study was conducted to compare<br />

male and female pts treated for stage 0 to IIIB BC, at the Ottawa<br />

Hospital <strong>Cancer</strong> Centre, between 1981 and 2009. Matching of male<br />

and female pts was done based on age at diagnosis (+/- 2 years),<br />

year of diagnosis (+/- 1 year), and disease stage. Data regarding<br />

surgery, adjuvant radiation, chemotherapy, and endocrine therapy was<br />

collected. Overall survival and disease-free survival were calculated<br />

using Kaplan-Meir analysis.<br />

Results:<br />

A total of 144 pts (72 female; 72 male) were eligible for this study;<br />

median age at diagnosis for both groups was 74 years (r: 30-85).<br />

Median follow-up was 54 months (r: 2-204) for males and 54<br />

months (r: 4-241) for females. All 72 men underwent mastectomy<br />

compared to 38 females (53%). Forty-four (61%) females received<br />

adjuvant radiation therapy compared to 29 (40%) males. Twenty<br />

(28%) females received adjuvant chemotherapy, compared to 23<br />

(31%) males. All female pts had estrogen receptor positive (ER+)<br />

disease, while 59 males (13 unknown) had ER+ disease. An equal<br />

number of males and females (79%) received endocrine treatment.<br />

The mean DFS for females was 127 months (25th percentile 33<br />

www.aacrjournals.org 339s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

months; median 104 months; 75th percentile 174 months), compared<br />

to 93 months (25th percentile 46 months; median 88 months; 75th<br />

percentile 112 months) for males (p=0.62). Mean OS for females was<br />

117 months (25th percentile 40 months; median 107 months; 75th<br />

percentile 136 months), and 124 months (25th percentile 48 months;<br />

median 90 months; 75th percentile 128 months) for males (p=0.35).<br />

In multivariate analysis, the only parameter that affected both DFS<br />

and OS was stage at diagnosis (hazard ratio 0.42, 95% CI 0.18-1.00;<br />

hazard ratio 0.25, 96% CI 0.09-0.68).<br />

Conclusions:<br />

This is one of the largest case-cohort studies to report on treatment<br />

outcomes of early stage male BC pts treated in a non-trial setting. Male<br />

pts received comparable systemic therapy (chemotherapy/endocrine)<br />

as their female counterparts and DFS and OS were similar. These<br />

results add to current evidence from population studies that male sex<br />

is not a poor prognostic factor for treatment outcomes in early stage<br />

BC. The development of a national male breast cancer registry would<br />

facilitate the evaluation of modern treatment strategies and patient<br />

outcomes in this population.<br />

P3-11-04<br />

Male breast cancer: Overall survival in a single institution.<br />

Bello MA, Bergmann A, Costa CRA, Pinto RR, Millen EC, Thuler<br />

LCS, Bender PFM. Brazilian National <strong>Cancer</strong> Institute, Rio de<br />

Janeiro, Brazil<br />

Background: Male breast cancer is an uncommon disease and the<br />

therapy is mainly based on what is know from female breast cancer.<br />

Objective: To investigate the clinicopathologic characteristics of male<br />

breast cancer and the overall survival in a single institution.<br />

Methods: The clinical data and survival status of 75 male breast<br />

cancer treated in a Brazilian public cancer hospital from 2000 to<br />

2009 were collected. The association with clinicopathological<br />

characteristics and overall survival was analyzed using Kaplan-Meier<br />

curves and the Cox proportional hazards regression (enter method)<br />

was used to assess survival differences after adjusting for confounders.<br />

The study was approved by National <strong>Cancer</strong> Institute Research and<br />

Ethics Committee (number 128/11).<br />

Results: The median patient age was 64 years (range 33-86). Estrogen<br />

receptor (ER) was positive in 58 (77.3%) patients, while progesterone<br />

receptor (PR) were positive in 47 (62.7%). Histology type was<br />

ductal infiltrant carcinoma for 57 (76.0%) and 51 (68.0%) patients<br />

underwent surgery. The median follow-up period was 43,1 months<br />

(range 2.7-147.8). The median survival from the diagnosis of breast<br />

cancer was 97.0 months (95%CI 53.6 -140.4) with a 61.7% 5-year<br />

overall survival (OS). In the final Cox regression model, independent<br />

factors associated with increased risk of death were metastasis at<br />

diagnosis (HR = 18.1; 95%CI: 5.9-55.2), ≥ 65 years old (HR = 4.3;<br />

95%CI: 1.7-10.5); tumor stages ≥ IIb (HR = 3.5; 95%CI: 1.3-9.7)<br />

and smoking (HR = 1.6; 95%CI: 1.04-2.6).<br />

Conclusions: Invasive ductal carcinoma is the main pathologic type.<br />

The median survival from the diagnosis of breast cancer was 97.0<br />

months and metastasis at diagnosis, patient age, tumor stage and<br />

smoking are independent prognostic factors.<br />

P3-12-01<br />

Serum biomarkers identification using quantitative proteomics<br />

in patients (pts) with untreated brain metastases from HER2-<br />

positive breast cancer receiving capecitabine (C) and lapatinib<br />

(L) (UNICANCER LANDsCAPE trial)<br />

Gonçalves A, Camoin L, Ben Younès I, Romieu G, Campone M, Diéras<br />

V, Cropet C, Mahier Aït-Oukhatar C, Dalenc F, Le Rhun E, Labbe-<br />

Devilliers C, Borg J-P, Bachelot T. Institut Paoli Calmettes, Marseille,<br />

France; Université de la Méditerranée, Marseille, France; Centre Val<br />

d’Aurelle, Montpellier, France; Institut de Cancérologie de l’Ouest,<br />

Nantes, France; Institut Curie, Paris, France; Centre Léon Bérard,<br />

Lyon, France; Unicancer, Paris, France; Institut Claudius Regaud,<br />

Toulouse, France; Centre Oscar Lambret, Lille, France<br />

Background<br />

The LANDsCAPE phase II study showed that C+L had a significant<br />

antitumor activity in previously untreated brain metastases (BM)<br />

from HER2-positive breast cancer (BC), with a central nervous<br />

system-objective response rate (CNS-ORR) of 67% and a median<br />

time to whole-brain radiotherapy (WBRT) of 8.3 months (43 analysed<br />

pts). Thus initial C+L combination might be a viable alternative to<br />

immediate WBRT in this setting. In this study, serum samples were<br />

collected before and during treatment for proteomic-based approaches<br />

to identify predictive biomarkers of treatment response.<br />

Methods<br />

Baseline (BL) and day 21 (D21) serum samples from highly<br />

responsive (R, CNS lesions -volumetric response ≥ 75%, n=6) and<br />

non-responsive (NR, CNS-stable or progressive disease, n=6) pts were<br />

subjected to isobaric Tag for Relative and Absolute Quantification<br />

(iTRAQ)-based proteomics. Samples from each condition were<br />

pooled to constitute 4 independent mixes ((BL-R, D21-R, BL-NR<br />

and D21-NR). Each of them underwent immuno-depletion of highly<br />

abundant proteins, concentration, reduction, alkylation and tryptic<br />

digestion. Then, each mix was fractionated and subjected to iTRAQ<br />

identification and quantitation using nano-liquid chromatography<br />

(LC) and electrospray ionisation (ESI)-orbitrap tandem mass<br />

spectrometry (MS/MS) (LTQ-orbitrap, Thermofisher). Differentially<br />

expressed proteins were analysed using IPA (Ingenuity® Systems)<br />

to highlight biological functions and signalling pathways that were<br />

most significantly enriched.<br />

Results<br />

iTRAQ-based measurements identified serum proteins with significant<br />

(fold-change > 1.5 and p-value < 0.05) differential expression between<br />

BL and D21, in R- (n= 38 proteins) and NR- pts (n= 18 proteins). At<br />

baseline, 30 proteins were differentially expressed between R-and<br />

NR-pts. Among the most differentially expressed proteins, some had<br />

been previously described as involved in cancer metastases (between<br />

BL and D21 : tenascin C, neuropilin-1, Serpin A1, Cathepsin S,<br />

prostaglandin D2 synthase, melanoma cell adhesion molecules,<br />

procollagen C-endopeptidase enhancer; between R and NR: carnosine<br />

dipeptidase 1, endothelial protein C receptor, matrix metallopeptidase<br />

9, cystatin C, lumican, plexin B2, Insulin-like growth factor binding<br />

protein acid labile subunit, pro-platelet basic protein, cadherin 5,<br />

Protein S). Proteins with differential expression during treatment<br />

were involved in various biological processes, including lipid<br />

metabolism, small molecule biochemistry, molecular transport, gene<br />

expression, cellular growth and proliferation, cellular movement and<br />

cancer, as well as various canonical pathways such as acute phase<br />

response signalling, LXR/RXR activation and complement system.<br />

Interestingly, and as opposed to NR-pts, R-pts specifically showed an<br />

overall increase in proteins of acute phase response, with a significant<br />

decrease in C-reactive protein (CRP).<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

340s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 3<br />

Conclusion<br />

iTRAQ-based quantitative proteomics identify serum proteins<br />

that could predict the therapeutic response to C+L combination in<br />

untreated BM from HER2-positive BC. If validated, such biomarkers<br />

may help to select the best therapeutic strategy in this setting.<br />

P3-12-02<br />

The impact of estrogen receptor status on treatment outcomes<br />

following gamma knife radiosurgery for brain metastases of<br />

primary breast cancer<br />

Cupelo EC, Aridgides PD, Bogart JA, Hahn SS, Shapiro AJ. SUNY<br />

Upstate Medical University, Syracuse, NY<br />

INTRODUCTION:<br />

Estrogen receptor (ER) status is a critical component of determining<br />

initial therapy of newly diagnosed breast cancer. Although 70% of<br />

primary breast malignancies are ER+, there remains limited data on<br />

whether ER status influences treatment outcomes for patients with<br />

brain metastases. Gamma Knife Radiosurgery (GKS) is increasingly<br />

being used in the management of brain metastases. The goal of this<br />

study is to determine the impact of ER status in patients receiving<br />

GKS for breast cancer metastases to brain.<br />

METHODS:<br />

A database of consecutive breast cancer patients receiving GKS at<br />

our institution from January 2000 to March 2012 was reviewed for<br />

hormone receptor status, dates of initial brain metastasis and GKS,<br />

presence of neurologic symptoms, and recurrence of GKS-treated<br />

lesions. The main cohort was subdivided by receptor status at initial<br />

tissue diagnosis. Progression or loss of local control of the treated<br />

lesions was defined as radiographic evidence of growth (greater than<br />

20% increase in the sum of the longest diameters) or treated lesions<br />

requiring additional treatment until death or last follow-up. Analysis<br />

for neurological symptom improvement was performed for patients<br />

with available follow-up information, with at least 1 month follow–up.<br />

Comparisons were made between subgroups using Kaplan-Meier<br />

method, Chi-square analysis and univariate comparisons with logrank<br />

test. Significance was set at p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

metastases, including its combination with trastuzumab in patients<br />

with HER2-positive disease. This trial may provide clinical evidence<br />

for the approach presented here.<br />

P3-12-04<br />

A phase 2, multi-center, open label study evaluating the efficacy<br />

and safety of GRN1005 alone or in combination with trastuzumab<br />

in patients with brain metastases from breast cancer<br />

Lin NU, Schwartzberg LS, Kesari S, Yardley DA, Verma S, Anders<br />

CK, Shih T, Shen Y, Miller K. UC <strong>San</strong> Diego, La Jolla, CA; Indiana<br />

University Melvin and Bren Simon <strong>Cancer</strong> Center, Indianapolis, IN;<br />

Sunnybrook Odette <strong>Cancer</strong> Centre, Toronto, ON, Canada; Dana-<br />

Farber <strong>Cancer</strong> Institute, Boston, MA; The West Clinic, Memphis,<br />

TN; Geron, Menlo Park, CA; University of North Carolina at Chapel<br />

Hill, NC; Sarah Cannon Research Institute, Nashville, TN; Tennessee<br />

Oncology, PLLC., Nashville, TN<br />

Rationale: Treatment of brain metastasis (BM) remains an unmet<br />

medical need due to the inability of most anti-cancer agents to<br />

effectively cross the blood-brain barrier (BBB). GRN1005 is a<br />

peptide-drug conjugate (PDC) consisting of 3 molecules of paclitaxel<br />

conjugated to a 19-amino acid peptide, Angiopep-2. GRN1005<br />

effectively penetrates the BBB by targeting the low-density<br />

lipoprotein receptor-related protein 1 (LRP1), which is expressed<br />

on the surface of the BBB and on certain neoplastic cells. In a phase<br />

1 study, GRN1005 demonstrated encouraging anti-tumor activity in<br />

breast cancer (BC) patients with BM.<br />

Methods: Up to 157 patients (pts) with measurable BM from BC,<br />

with or without extra-cranial disease, are currently being enrolled<br />

in an ongoing open label study. Prior cranial irradiation is allowed<br />

but lesions previously treated with stereotactic radiosurgery (SRS)<br />

are excluded from the efficacy analysis. All currently enrolled pts<br />

received GRN1005 at a starting dose of either 650 or 550 mg/m 2<br />

IV every three weeks, until intra-cranial disease progression (iPD),<br />

unacceptable toxicity, or initiation of non-protocol systemic therapy.<br />

HER2 positive pts received concomitant trastuzumab. The primary<br />

endpoint is intra-cranial objective response rate (iORR), as assessed<br />

by independent review using CNS RECIST 1.1. Additional endpoints<br />

include safety, duration of intra-cranial response, 3-month intracranial<br />

progression free survival (iPFS3), extra-cranial ORR, and<br />

neurocognitive function.<br />

Results: As of 4 June 2012, 24 pts (13 at 650 mg/m 2 and 11 at 550<br />

mg/m 2 ) have been treated with a median of 2 cycles of GRN1005<br />

(range 1-5). Median age was 51.5 years (34-68) and 14 (58%) pts<br />

have HER2-positive disease. Pts received a median of 6 (3-14) prior<br />

systemic regimens; 19 had received prior whole brain radiation<br />

therapy (WBRT) ± SRS and 2 had received prior SRS only (data<br />

pending on 3 pts). Six (25%) pts have discontinued treatment with<br />

GRN1005, including 2 due to iPD and 2 due to adverse events<br />

(AE). One additional pt had clinical progression and discontinued<br />

GRN1005.<br />

The AEs observed at 650 mg/m 2 are qualitatively consistent with<br />

conventional paclitaxel but are more frequent and more severe than<br />

observed with q3 week paclitaxel which led to a protocol amendment<br />

and dose reduction to 550 mg/m 2 for all subsequent pts. Among all<br />

treated pts, the most frequent AEs were neutropenia (75%), fatigue<br />

(50%), mucositis (33%), neuropathy (29%), and diarrhea (25%).<br />

Notable Grade 3-4 AEs were neutropenia in 12 pts (including 1 case<br />

of febrile), and arthralgia, fatigue, and rash in 3 pts each. Preliminary<br />

anti-tumor activity has been observed in both intra-cranial and extracranial<br />

lesions.<br />

Conclusions: GRN1005 is a promising treatment for BM associated<br />

with breast cancer and demonstrates a safety profile qualitatively<br />

similar to paclitaxel dosed every 3 weeks. Due to tolerability concerns<br />

at the 650 mg/m 2 dose, the 550 mg/m 2 dose is currently being<br />

evaluated in these heavily pretreated BC pts. Updated efficacy and<br />

safety data will be presented at the meeting.<br />

P3-12-05<br />

Clinical features and outcomes of leptomeningeal metastasis in<br />

patients with breast cancer: a single center experience<br />

Jo J-C, Kang MJ, Ahn J-H, Jung KH, Kim JE, Gong G, Kim HH,<br />

Ahn SD, Kim SS, Son BH, Ahn SH, Kim S-B. Asan Medical Center,<br />

University of Ulsan College of Medicine, Seoul, Korea<br />

Background: Leptomeningeal metastasis (LM) is one of the major<br />

problems in managing metastatic breast cancer because of LM<br />

typically carries a devastating prognosis and often represents a<br />

terminal event. We analyzed the clinical features and outcomes of<br />

LM in patients with breast cancer.<br />

Methods: We retrospectively reviewed the medical records of patients<br />

who were diagnosed with LM from breast cancer between 2002 and<br />

2012 at Asan Medical Center.<br />

Results: Of the 95 LM patients by cytologically proven (n=81)<br />

or radiologically diagnosed (n=14), 57 (60%) had an ECOG<br />

performance status (PS) ≥ 3, and the median age was 47 years<br />

(range, 26-72 years). The patients were diagnosed with LM after<br />

a median of 10.3 months (95% CI, 5.5-15.0 months) from the<br />

time of diagnosis of metastatic breast cancer. LM was present in<br />

2 patients at the time of initial diagnosis. Twenty-three patients<br />

(24.2%) had isolated CNS metastasis, and 6 (6.3%) had only LM<br />

without any detectable metastasis sites. At the time of diagnosis<br />

of LM, 46 patients (48.4%) presented with coincidental failure of<br />

systemic disease control. Seventy-eight patients (82.1%) underwent<br />

intrathecal chemotherapy (methotrexate; n=78, thiotepa; n=11),<br />

resulting in one-third of cytologic negative conversion (n=26), and<br />

41 (43.2%) received systemic chemotherapy. The overall median<br />

survival time was 3.3 months (95% CI, 2.5-4.2 months) and 7.8% of<br />

the patients survived for more than 1 year. Overall survival tended<br />

to be better in patients who achieved cytologic negative conversion<br />

to intrathecal chemotherapy than those did not (median 4.5 months<br />

versus 3.2 months, P=0.241). Overall survival was not different<br />

according to subtypes; hormone receptor (+), HER2 (+), and triple<br />

negative (median 3.6 months, 3.3 months, and 3.2 months, P=0.937).<br />

Multivariate analysis demonstrated that ECOG PS ≥ 3 (HR=2.09,<br />

95% CI 1.21-3.58, P=0.007), coincidental failure of systemic disease<br />

control at LM (HR=3.01, 95% CI 1.76-5.15, P


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 3<br />

In addition, improvement in our understanding of breast cancer<br />

biology changed the treatment strategy, possibly leading to improved<br />

outcomes in patients with breast cancer and brain metastases.<br />

Object:<br />

The aim of this study was to investigate the correlation between<br />

biology of primary tumors and prognosis of patients with breast<br />

cancer metastasis to the brain.<br />

Patients and method:<br />

Among 3,171 patients with primary breast cancer undergoing<br />

surgery at out hospital from 2003 to 2010, 35 patients (1.1%) who<br />

developed brain metastases, excluding stage IV at the initial diagnosis.<br />

We reviewed 35 patients for a correlation between survival and<br />

clinicopathological variables, including stage, hormonal sensitivity,<br />

HER2 expression status, the number of brain metastases, and<br />

treatment for brain metastases.<br />

Results:<br />

The mean age of 35 patients was 52.1 years (range,22-68) and the<br />

median follow-up period was 41.0 months. The subtypes of primary<br />

tumors included triple-negative (TN) in 14 patients (40%), luminal<br />

in 11 patients (31.4%), luminal HER2-positive (luminal HER2) in<br />

2 patients (5.7%), and HER2-positive (HER2) in 2 patients (5.7%).<br />

The duration from surgery to recurrence (disease-free survival, DFS,<br />

months) was 14.7 in TN, 26.5 in luminal, 29.8 in luminal HER2, and<br />

31.0 in HER2. The duration from the first recurrence to the detection<br />

of brain metastases (months) was 7.2 in TN, 14.4 in luminal, 15.8 in<br />

luminal HER2, and 4.0 in HER2. The mean survival time after the<br />

diagnosis of brain metastases was 8.6 months (range, 0-40) with a<br />

survival time of ³ 1 year in 8/35 patients (23%) and that of < 1 year<br />

in 21/35 patients (60%). Both DFS and the duration from the first<br />

recurrence to the diagnosis of brain metastases were shorter in TN<br />

compared with other subtypes. In HER2 subtype, DFS was long but,<br />

once recurrence was detected, the duration to the development of brain<br />

metastases was short. In multivariate analysis of clinicopathological<br />

variables (age, DFS, ER, HG, HER2, the number of brain metastases<br />

(solitary versus multiple), the site of initial metastasis (brain versus<br />

other organs), and the presence or absence of chemotherapy after<br />

brain metastases, revealed solitary brain metastasis (P=0.002) and<br />

the initial metastatic site of brain (P=0.003) to be independent factors<br />

predicting survival.<br />

Table 1. Clinicopathological variables and survival in patients with breast cancer metastasis to the<br />

brain (multivariate analysis)<br />

odd ratio 95%CI p<br />

age 0.534 0.2532-1.1274 0.100<br />

DFS 1.34 0.4842-3.7451 0.568<br />

HG 0.61 0.1864-0.2354 0.430<br />

ER 0.578 0.1869-1.7897 0.342<br />

HER2 0.551 0.1878-1.1640 0.287<br />

The site of initial metastasis(Brain versus other<br />

0.164<br />

organs)<br />

1.9856-2.4003 0.003<br />

The number of brain metastases(solitary versus<br />

6.904<br />

multiple)<br />

0.0498-0.5489 0.002<br />

The presence or absence of chemotherapy after<br />

0.389<br />

brain metastases<br />

0.1192-0.2715 0.118<br />

Conclusion:<br />

The clinical outcomes of brain metastases were different among<br />

tumor subtypes. Factors predicting survival in patients with breast<br />

cancer and brain metastases may be the site of initial metastasis, the<br />

number of brain metastasis, and local treatment for brain metastases.<br />

P3-12-07<br />

Voyagers and their aids: The role of interactions between tumor<br />

and endothelial cells in brain metastasis<br />

Shah KN, Faridi JS. University of the Pacific, Stockton, CA<br />

Clinically, brain metastases are most commonly observed in breast,<br />

lung, and melanoma cancers. When a tumor cell leaves its site of<br />

origin, it enters a nearby blood vessel on a voyage to find a new tissue<br />

to invade. These circulating tumor cells will act as voyagers and they<br />

interact with a endothelial cell. For example, in the case of brain<br />

metastasis, tumor cells need to adhere to brain endothelial cells before<br />

they can invade into the surrounding brain tissue. Thus, investigating<br />

the mechanisms of tumor and endothelial cell interactions can serve<br />

as a very powerful tool to inhibit the voyage of these tumor cells<br />

and prevent the formation of metastases. In this study, we performed<br />

trans-endothelial migration assay using HBMVEC cells to study<br />

tumor-endothelial interactions. In general, HBMVEC cells were<br />

seeded to BD-Falcon cell culture inserts at a density of 2 × 105 cells<br />

and allowed to grow for 24 hours to form a confluent monolayer and<br />

GFP expressing tumor cells (MCF-7, Akt3 overexpressing MCF7,<br />

MDA-MB-231 and MDA-MB-231-BR) were incubated at suitable<br />

interval at 37°C. At the end of incubation period, cells were fixed and<br />

number of tumor cells that will penetrate the monolayer of HBMEC<br />

were counted using Phase-contrast microscope in 12 different visual<br />

fields. Transmigrating tumor cells are thought to be able to overcome<br />

the endothelial barrier by inducing changes within endothelial cells.<br />

We identified the molecular changes responsible for such adhesion<br />

and invasion by cell sorting for gene expression analysis. After coculturing<br />

the HBMEC (after transfecting with Mcherry protein) with<br />

GFP expressing tumor cells and Mcherry labeled HBMEC, we used<br />

cell sorting to separate tumor and HBMEC cells. After sorting the<br />

tumor cells and endothelial cells from co-culture, (MCF-7 monolayer<br />

vs MCF-7 co-cultured, MDA-MB-231 monolayer vs MDA-MB-231<br />

co-cultured, HBMEC monolayer vs HBMEC co-cultured) gene<br />

expression analysis was performed using SABiosciences cell adhesion<br />

assay kit to accurately capture the genetic changes that occured in the<br />

tumor and endothelial cells upon co-culture.<br />

P3-12-08<br />

Incidence, predictive factors and outcome of brain metastases<br />

(BM) in a single institution cohort of breast cancer patients.<br />

Perelló A, Alarcon J, Garcias C, Duran J, Colom B, Clemente G,<br />

Avella A, Canet R, Torrecabota J, Rifa J. Hospital Universitari Son<br />

Espases, Palma, Balearic Islands, Spain<br />

Background<br />

The aim of the study was to determine the cumulative incidence of<br />

BM, predictive factors of BM, and survival from diagnosis of BM in<br />

a cohort of breast cancer patients in a single institution.<br />

Patients and Methods<br />

We collected data from a cohort of 793 breast cancer patients between<br />

January 2000 and December 2005 in our institution. Variables<br />

recorded include age at diagnosis, hormonal receptor status (HRS),<br />

human epidermal growth factor receptor 2 status (HER2), histological<br />

grade, T stage and N stage. We analyzed the 5 and 10-year cumulative<br />

incidence of BM. Time to detection of BM and survival from BM<br />

were estimated using Kaplan-Meier method. Cox regression model<br />

was used to analyze the variables associated with time to BM<br />

Results<br />

With a median follow-up of 100 months, 49 patients went on to<br />

develop BM. The overall 5 and 10 year cumulative incidence of BM<br />

were 4.9% and 8.2% respectively. The table shows the 5 and 10 year<br />

cumulative incidence and hazard ratio (HR) of the predictive factors<br />

for the development of BM in the univariable analysis.<br />

www.aacrjournals.org 343s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

5 and 10 year cumulative incidence and hazard ratio (HR) of the predictive factors for the<br />

development of BM in the univariable analysis<br />

Variables % 5-year incidence % 10-year incidence HR (95%CI) P value<br />

HER 2 + 15.4 24.1 3.22 (1.80-5.79)


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 3<br />

35 years-old (HR 2.09; 95% CI: 1.15-3.81, p=0.016) were strongly<br />

associated with higher incidence of brain metastasis. Table 1 showed<br />

the important characteritics according to different age group.<br />

Characteristics of patients according to diagnosis age<br />

Characteristics Total, N=2248 Age ≤35, N=104 Age 36-59, N=1503 Age ≥60, N=641<br />

<strong>Breast</strong> cancer subtype, N (%)<br />

Luminal A 1104 (49.1) 47 (45.2) 744 (49.5) 313 (48.8)<br />

Luminal B 448 (19.9) 24 (23.1) 299 (19.9) 125 (19.5)<br />

HER2-enriched 301 (13.4) 10 (9.6) 202 (13.4) 89 (13.9)<br />

Triple negative<br />

phenotype<br />

395 (17.6) 23 (22.1) 258 (17.2) 114 (17.8)<br />

Brain metastasis<br />

164 (7.3) 19 (18.3) 109 (7.3) 36 (5.6)<br />

event, N (%)<br />

Cumulative incidence rate of brain metastasis event, % (95% CI)<br />

At 1 year 1.2 (1.0- 1.4) 3.0 (1.3- 4.7) 1.0 (0.7- 1.3) 1.2 (0.8- 1.6)<br />

At 3 year 4.9 (4.4- 5.4) 10.4 (7.1- 13.7) 4.7 (4.1- 5.3) 4.5 (3.6- 5.4)<br />

At 5 year 8.2 (7.5- 8.9) 20.5 (15.3- 25.7) 7.8 (6.9- 8.7) 7.5 (6.1- 8.9)<br />

Luminal A= ER+ and/or PR+ and grade 1-2; Luminal B= ER+ and/or PR+ and HER2+ or grade 3;<br />

HER2- enriched= HER2+ by IHC or FISH and ER/PR -, Triple negative= ER-. PR- and HER2-<br />

Furthermore the influence of each biological subtype amoung different<br />

age group were investigated. For patient younger than 35 years-old,<br />

risk of the developement of brain metastasis was independent to<br />

biological subtype (p=0.507); while the risk of brain metastasis was<br />

influenced by biological subtype in older age groups. For patients<br />

older than 35, patients with triple negative and HER2 enriched disease<br />

had significantly increased risk for brain metastasis compared to<br />

luminal A; but for patients with luminal B disease, such incremental<br />

risk for the development brain metastasis was only observed in<br />

patients older than 60 years-old.<br />

Conclusion<br />

Our study found that each biological subtypes of breast cancer had<br />

distinct impact on the development of brain metastasis among different<br />

age group. Further studies were needed to disclose the biology<br />

underneath. Also the findings should be taken into consideration when<br />

developing strategy for brain metastasis prevention.<br />

P3-12-11<br />

Clinical outcome in patients with surgically resected brain<br />

metastases from breast cancer: prognostic considerations<br />

regarding molecular status and established prognostic<br />

classification systems.<br />

Tabouret E, Metellus P, Tallet A, Figarella-Branger D, Charaffe-<br />

Jauffret E, Viens P, Gonçalves A. Institut Paoli Calmettes, Marseille,<br />

France; APHM, Marseille, France; Insitut Paoli-Calmettes,<br />

Marseille, France<br />

Introduction: Incidence of brain metastases (BM) from breast cancer<br />

(BC) is increasing, paralleling increasing life expectancy of patients.<br />

However, biology, prognostic evaluation and treatment strategy are<br />

still a matter of debate. We analysed here a series of patients with<br />

resected brain metastases from BC to assess the actual prognostic<br />

factors in this specific population and the relevancy of widely accepted<br />

BM prognostic classification systems.<br />

Methods: we reviewed retrospectively all patients (n=49) referred to<br />

our institution between December 2001 and July 2011 with available<br />

tumor tissue from resected breast cancer brain metastases. All patients<br />

underwent initial breast tumor surgery and surgical resection of the<br />

BM. Patients, tumor and treatment characteristics were recorded.<br />

Prognostic value of ER, PR, HER2 status of BM, age, Karnofsky<br />

performance status, Mini Mental Score (MMS), clinical features,<br />

number and topography of BM, systemic disease characteristics<br />

(response, metastatic sites), delay between primary tumor and BM<br />

diagnoses and treatment after neurosurgery were analyzed using<br />

uni- and multi-variate analyses.<br />

Results: Median age was 54 years (28-75). Median KPS was 80 (50-<br />

90). MMS was abnormal for 7 pts (14.3%). At BM diagnosis, 12 pts<br />

(24.5%) did not present extracranial disease and 15 (20.6%) had only<br />

one systemic metastatic site. After surgery, 83.7% of pts benefited<br />

from whole-brain radiotherapy and 79.6% from systemic therapy.<br />

Median follow-up was 25.6 months. Median cerebral progression-free<br />

survival (C-PFS) was 11.3 months (IC95:6.0-16.6) and median overall<br />

survival (OS) was 19.4 months (IC95:16.1-22.7). By univariate<br />

analysis, neither hormonal or HER2 BM status impacted survival.<br />

Similarly, molecular subtyping by IHC approximation (“basal”,<br />

ER- and PR-/HER2-;”luminal A”, ER and/or PR+/HER2-; “luminal<br />

B”, ER and/or PR+/HER2+; “HER2”, ER and PR-/HER2+) was not<br />

significantly associated with survival(p=0.946). Abnormal MMS and<br />

multiple systemic metastases were associated with poor C-PFS (p=<br />

0.009 for both) and poor OS (p=0.006 and p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

was 34 and 8 m respectively (p=0.001). Endocrine therapy did not<br />

improve survival in ER and/or PR positive pts, (median 14 vs 20 m,<br />

p=0.437). In the entire group, median survival in RPA classes I, II<br />

and III was 24, 18 and 3 m, respectively (p


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 3<br />

P3-13-03<br />

Molecular factors associated with bone metastases in breast<br />

cancer patients<br />

Winczura P, Sosinska-Mielcarek K, Duchnowska R, Badzio A, Lakomy<br />

J, Majewska H, Peksa R, Pieczynska B, Radecka B, Debska S, Zok J,<br />

Rogowski W, Strzelecka M, Kulma-Kreft M, Blaszczyk P, Litwiniuk<br />

M, Jesien-Lewandowicz E, Rutkowski T, Jaworska-Jankowska M,<br />

Adamowicz K, Foszczynska-Kloda M, Biernat W, Jassem J. Medical<br />

University of Gdansk, Poland; Regional Oncology Center, Gdansk,<br />

Poland; Regional Oncology Center, Opole, Poland; Military Institute<br />

of Medicine, Warsaw, Poland; Medical University of Lódz, Poland;<br />

Warmia and Masuria Oncology Center, Olsztyn, Poland; Maritime<br />

Hospital, Gdynia, Poland; Oncology Center, Bydgoszcz, Poland;<br />

Maria Sklodowska-Curie Memorial <strong>Cancer</strong> Center and Institute of<br />

Oncology, Gliwice, Poland; Regional Hospital in Wroclaw, Poland;<br />

West Pomeranian Oncology Center, Szczecin, Poland<br />

Background. Bones constitute the most frequent localization of<br />

distant failure in breast cancer patients. Treatment of bone metastases<br />

is virtually palliative, but in a proportion of patients can effectively<br />

prolong survival and improve quality of life. Currently, there are<br />

no effective strategies to prevent bone dissemination with systemic<br />

adjuvant therapies. Thus, better molecular selection and identification<br />

of patients who have particularly high risk of skeletal metastases may<br />

facilitate designing future clinical trials. In this study we analyzed<br />

expression of selected tumor proteins potentially associated with<br />

skeletal metastases in breast cancer patients.<br />

Patients and methods. The study group included 184 metastatic<br />

breast cancer patients; 113 with clinically diagnosed bone metastases<br />

and 71 with exclusively other sites of metastases, respectively. In<br />

all patients, using tissue microarrays technology, IHC expression of<br />

the following proteins was evaluated in the primary tumor: estrogen<br />

receptor (ER), progesterone receptor (PR), human epidermal<br />

growth factor receptor 2 (HER2), Ki67, cyclooxygenase 2 (COX2),<br />

chemokine receptor CXCR4, osteopontin (OPN), calcium sensing<br />

receptor (CaSR), cytokeratins 5/6 (CK5/6) and parathyroid hormonerelated<br />

protein receptor 1 (PTHrPR1). Additionally based on ER/<br />

PR, HER2 and Ki67 expression, following molecular breast cancer<br />

subtypes were selected: luminal A, luminal B HER2-negative, luminal<br />

B HER2-positive, nonluminal HER2-positive and triple negative.<br />

Results. Median survival in patients with bone vs. other site of<br />

metastases was 56 vs. 37 months respectively (p=0.0098). ER<br />

expression was more common in patients who did, compared<br />

with those who did not develop bone metastases (74.% vs. 45%<br />

respectively; p=0.0001), whereas cytoplasmic overexpression of<br />

OPN (1.9% vs. 14% respectively; p=0.002) and PTHrPR1 (16%<br />

vs. 34% respectively; p=0.007) was more common in patients with<br />

other sites of metastases. The impact of ER and cytoplasmic OPN<br />

expression on the risk of bone dissemination was confirmed in the<br />

multivariate analysis (Table 1). Luminal A breast cancer subtype<br />

(43% vs. 23% respectively; p=0.009) and luminal B HER2-positive<br />

(16% vs. 4.9%, respectively; p=0.032) were more common among<br />

patients with bone dissemination, whereas triple negative tumors<br />

prevailed in patients with other sites of metastases (16% vs. 38%;<br />

p=0.002). Median survival of patients with luminal A subtype was<br />

significantly longer than that with remaining subtypes (67 vs. 38<br />

months respectively; p=0.0004).<br />

Conclusions. These results suggest that ER expression and lack of<br />

OPN expression are independent factors predicting increased risk of<br />

bone dissemination in breast cancer patients. Bone metastases are<br />

specifically associated with luminal A and luminal B HER2-positive<br />

subtypes.<br />

Table 1. Multivariate analysis<br />

Marker Regression coefficient Standard error P 95% CI<br />

ER 0.412 0.135 0.002 0.147 - 0.677<br />

OPN cytoplasmic -0.988 0.416 0.018 -1.804 - -0.171<br />

P3-13-04<br />

Skeletal related Events and Bone Metastasis Patients with <strong>Breast</strong><br />

<strong>Cancer</strong> in Japan. A Retrospective Study<br />

Yamashiro H, Takada M, Imai S, Yamauch A, Tsuyuki S, Inamoto T,<br />

Matsutani Y, Sakata S, Wada Y, Okamura R, Harada T, Tanaka F,<br />

Moriguchi Y, Kato H, Higashide S, Kan N, Yoshibayashi H, Suwa<br />

H, Okino T, Nakayama I, Ichinose Y, Yamagami K, Hashimoto T,<br />

Nakatani E, Nagata Y, Kudo Y, Toi M. National Hospital Organization<br />

KURE Medical Center, Kure, Hiroshima, Japan; Graduate School<br />

of Medicine, Kyoto University, Kyoto, Japan; Kurashiki Central<br />

Hospital, Kurashiki, Okayama, Japan; Kitano Hospital (The Tazuke<br />

Kofukai Medical Research Institute), Osaka, Japan; Osaka Red<br />

Cross Hospital, Osaka, Japan; Tenri Hospital, Tenri, Nara, Japan;<br />

National Hospital Organization Kyoto Medical Center, Kyoto, Japan;<br />

National Hospital Organization Himeji Medical Center, Himeji,<br />

Hyogo, Japan; Yamatotakada Municipal Hospital, Yamatotakada,<br />

Nara, Japan; Osaka Saiseikai Noe Hospital, Osaka, Japan; Fukui<br />

Red Cross Hospital, Fukui, Japan; Kyoto City Hospital, Kyoto, Japan;<br />

Nagahama City Hospital, Nagahama, Shiga, Japan; Kan Norimichi<br />

Clinic, Kyoto, Japan; Japanese Red Cross Society Wakayama Medical<br />

Center, Wakayama, Wakayama, Japan; Hyogo Prefectural Tsukaguchi<br />

Hospital, Amagasaki, Hyogo, Japan; Kouga Public Hospital, Kouga,<br />

Shiga, Japan; Kyoto Min-iren Chuo Hospital, Kyoto, Japan; Takatsuki<br />

Red Cross Hospital, Takatsuki, Osaka, Japan; Shinko Hospital, Kobe,<br />

Hyogo, Japan; Hashimoto Clinic, Kobe, Hyogo, Japan; Translational<br />

Research Informatics Center, Foundation for Biomedical Research<br />

and Innovation, Kobe, Hyogo, Japan; Kobe City Medical Center<br />

General Hospital, Kobe, Hyogo, Japan<br />

Objective: Since skeletal related events (SRE) worsen QOL, the<br />

information relating to the SRE is important in the patient’s care. We<br />

have examined the incidences of bone metastasis (BM) and SRE in<br />

Japan and explored the prognostic factors which may affect the BM<br />

or SRE free period or survival.<br />

Patients and Methods: A retrospective, multicenter (21 institutes)<br />

study was performed. Patients with node positive or node negative<br />

with moderate to severe recurrence risk primary breast cancer<br />

were included. The primary endpoint was SRE-free period, and the<br />

secondary endpoints were BM-free period and the over-all survival<br />

period. By using a proportional hazard model (Cox regression),<br />

factors which affect these endpoints were examined in the population<br />

excluding stage IV patients.<br />

Results: 1,779 patients were registered and 1,708 patients’ data were<br />

used for analysis. The median follow-up duration was 5.71 years.<br />

SRE occurred in 133 (68.9%) of 193 patients who developed BM.<br />

The median SRE-free period from the initial treatment was 1,006<br />

days. The SRE-free rate and SRE-free survival rate at 5-years were<br />

92.6% and 85.4%, respectively. The median period from the initial<br />

SRE to the second one was 112 days, and the second to the third one<br />

was 147 days. The median BM-free period was 767 days. The BMfree<br />

and BM-free survival rates at 5-years were 89.2% and 83.7%,<br />

respectively. The over-all survival rate at 5-years was 89.4%. The<br />

SRE-free period was significantly affected by the number of lymph<br />

node metastasis (LN mets) (Hazard Ratio; HR [95%CI]; 1.09 [1.042-<br />

1.143], p=0.0002) and clinical stage (when stage I was assumed<br />

as reference, HR in stage II; 4.54 [1.296-15.933], stage III; 7.36<br />

[1.171-31.539], p=0.0262). SRE-free survival period was affected by<br />

the number of LN mets (HR; 1.10 [1.066-1.131], p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

(stage I as reference, HR in stage II: 2.93 [1.349-6.349], and stage<br />

III: 4.28 [1.694-10.815], p=0.008), and tumor phenotype (Luminal<br />

[L] A as reference, HR in LB; 1.89 [0.868-4.117], L-HER2: 2.81<br />

[1.334-5.923], HER2: 2.68 [1.266-5.659], Triple negative (TN);<br />

5.38 [3.031-9.537], p


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 3<br />

on cancer cell migration were not observed when using osteoblasts.<br />

Together, these results suggest the functional importance of osteocytic<br />

Cx43 hemichannels in attenuating cancer cell migration and growth.<br />

P3-14-01<br />

Molecular definition of the transition of ductal carcinoma in situ<br />

(DCIS) to invasive ductal carcinoma (IDC).<br />

Colavito S, Stepansky A, Madan A, Harris LN, Hicks J, Bossuyt V,<br />

Rimm D, Lannin D, Stern DF. Yale University, New Haven, CT; Cold<br />

Spring Harbor Laboratory, Cold Spring Harbor, NY; Case Western<br />

Reserve University, Cleveland, OH<br />

Background: Ductal carcinoma in situ (DCIS) is thought to be a<br />

precursor lesion to invasive ductal carcinoma and sometimes occurs<br />

in combination with invasive disease. However, the majority of DCIS<br />

lesions do not progress to invasive disease. To date no molecular<br />

markers have been identified that associate with the potential for<br />

development to invasive disease. Our work has sought to identify<br />

molecular markers that can be used to determine the likelihood of<br />

DCIS being associated with invasive disease. Identification of such<br />

markers could be of direct therapeutic benefit, since patients with a<br />

high likelihood of associated or subsequent invasive disease could<br />

be managed more aggressively.<br />

Methods: We have compiled matched pairs, consisting of patients<br />

that have recurred with DCIS, compared to those who have DCIS plus<br />

invasive disease. Laser-capture microdissection was used to isolate in<br />

situ and invasive components of the latter, and in situ components of<br />

the former. We analyzed these samples by parallel cDNA-mediated<br />

Annealing, Selection, Extension, and Ligation (DASL) analysis of<br />

transcription, and DNA copy number analysis by resequencing. In<br />

addition, exome capture deep re-sequencing has been conducted on<br />

multiple cored in situ versus invasive components of DCIS from the<br />

same tumor sample.<br />

Results: Gene expression analysis revealed candidate genes that<br />

were specific to DCIS and others that were expressed only in the<br />

adjacent invasive component of the tissue. These genes are currently<br />

being evaluated to identify interesting candidates. Additionally,<br />

we characterized the gene expression signatures from tumors that<br />

recurred as DCIS only, to the DCIS component of those tumors that<br />

recurred with adjacent invasive disease. We identified genes that were<br />

differentially expressed between these data sets.<br />

The DNA copy number analysis of laser-captured samples indicated<br />

a single dominant clone in each DCIS. In contrast to previously<br />

reported observations, the profiles of invasive versus non-invasive<br />

lesions differ significantly from one another. Observations of noninvasive<br />

versus invasive components of adjacent disease in two of<br />

the pairs indicate that the profile of the invasive lesions differ from<br />

that of the non-invasive component, however the profiles share many<br />

individual features.<br />

Discussion: Our data identify differences between in situ and<br />

neighboring invasive tumor that may mark features associated with<br />

progression. Integration of the copy number and transcription profiling<br />

datasets will reveal the extent to which genomic alterations drive<br />

changes in gene expression. Identification of markers that distinguish<br />

indolent and aggressive subsets of DCIS that can be used to predict<br />

an association with invasive disease has the potential for near term<br />

clinical utility and may identify therapeutic targets for aggressive<br />

DCIS.<br />

P3-14-02<br />

Withdrawn by Author<br />

P3-14-03<br />

Differences in pathological and biological factors between DCIS,<br />

DCIS with microinvasion (DCIS-MI) and DCIS with concomitant<br />

invasive ductal carcinoma (DCIS-IDC).<br />

MacGrogan G, Baranzelli MC, Picquenot JM, Penault-Llorca F,<br />

Mathieu MC, Tas P, Fermeaux V, Mery E, Sagan C, Blanc-Fournier<br />

C, Brabencova E, Arnould L, Jacquemier J, Ettore F, Velasco<br />

V, Gonzalves B, Brouste V, Tunon de Lara C. Institut Bergonié,<br />

Bordeaux, France; Centre Oscar Lambret, Lille, France; Centre<br />

Henri Becquerel, Rouen, France; Centre Jean Perrin, Clermont-<br />

Ferrand, France; Institut Gustave Roussy, Villejuif, France; Centre<br />

Eugène Marquis, Rennes, France; CHU de Limoges, Limoges,<br />

France; Institut Claudius Regaud, Toulouse, France; Centre René<br />

Gauducheau, Saint Herblain, France; Centre François Baclesse,<br />

Caen, France; Institut Jean Godinot, Reims, France; Centre Georges<br />

François Leclerc, Dijon, France; Institut Paoli Calmettes, Marseille,<br />

France; Centre Antoigne Lacassagne, Nice, France<br />

Background<br />

Specific pathological and biological factors differentiating pure DCIS,<br />

DCIS with microinvasion (DCIS-MI) and DCIS with concomitant<br />

invasive ductal carcinoma (DCIS-IDC) are currently unknown.<br />

Identification of such factors would enable us to better understand<br />

the transition from an in situ to an invasive carcinoma and improve<br />

patient management for patients diagnosed with extensive DCIS.<br />

Methods<br />

We previously performed a prospective surgical study demonstrating<br />

the benefit of performing upfront sentinel lymph node (SLN) biopsy<br />

in patients with extensive DCIS or DCIS-MI undergoing mastectomy.<br />

In that study, 196 DCIS and 31 DCIS-MI diagnosed on vacuumassisted<br />

macrobiopsies underwent mastectomy and SLN biopsy. Final<br />

pathological status after mastectomy examination revealed 117 DCIS,<br />

38 DCIS-MI, 69 DCIS-IDC, and in 3 cases no residual disease was<br />

found. The mastectomy specimens of 216 cases (111 DCIS, 37 DCIS-<br />

MI, 68 DCIS-IDC) were available for central pathological review and<br />

a TMA containing DCIS lesions of 214 cases (110 DCIS, 36 DCIS-<br />

MI, 68 DCIS-IDC) was performed. DCIS morphological features<br />

(histological size, nuclear grade, comedo necrosis, inflammation)<br />

and immunohistochemical factors (ER, PR, Ki-67 and Her2) were<br />

assessed in each of the three groups and compared using Chi2 or<br />

Wilcoxon tests.<br />

Results<br />

Median histological DCIS sizes did not significantly differ<br />

between DCIS and DCIS-MI groups (70mm[4-160] and 60mm[4-<br />

180], respectively, p=0.33), nor between DCIS and DCIS-<br />

IDC(70mm[4-160] and 65mm[10-140], respectively, p=0.98). DCIS<br />

nuclear grade was significantly higher in DCIS-MI compared to DCIS<br />

(p=0.01), but there was no difference in nuclear grade between DCIS<br />

and DCIS-IDC (p=0.22). Comedo necrosis was more often present in<br />

DCIS-MI compared to DCIS (p=0.04), and no such difference was<br />

found between DCIS and DCIS-IDC (p=0.81). Stromal inflammation<br />

surrounding DCIS was more often found in DCIS-MI and DCIS-IDC<br />

compared to DCIS (p=0.03 and p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Conclusions<br />

In patients with extensive DCIS there are few morphological or<br />

current immunohistochemical factors differentiating in situ cases<br />

from those that have associated microinvasion or invasive ductal<br />

carcinoma. DCIS-MI cases have more aggressive histological features<br />

than DCIS. Stromal inflammation surrounding DCIS was the only<br />

feature distinguishing DCIS from DCIS-IDC.<br />

P3-14-04<br />

Symptomatic DCIS is a risk factor for invasion and should be<br />

managed accordingly<br />

Dimopoulos N, Williams K, Kirwan CC, Johnson R, Howe M, Bundred<br />

NJ. University Hospital of South Manchester<br />

Background: The National ABS Guidelines indicate axillary surgery<br />

is unwarranted in DCIS but should be offered to patients undergoing<br />

mastectomy. Up to 30% of pre-operatively diagnosed DCIS lesions<br />

harbour occult invasive cancers at mastectomy.<br />

Methods: To determine if DCIS mode of presentation, size (or other<br />

clinical factors such as Grade or HER2 status) predict invasion or<br />

nodal involvement, 380 patients undergoing surgery with a DCIS preoperative<br />

(B5) diagnosis were studied. In total 162 patients underwent<br />

mastectomy +/- axillary staging for DCIS and 216 conservation<br />

surgery between 2008-2011 in one unit.<br />

HER2, oestrogen receptor (ER) and progesterone receptor<br />

(PR) expression within primary DCIS were established using<br />

immunohistochemistry. HER2 was scored 0 (absent) to 3 (maximum).<br />

Scores ≥2 were taken as positive if amplified on FISH testing. ER<br />

and PR scored positive if ≥5% of cells stained.<br />

Results: In total 216 had <strong>Breast</strong> Conservation and 162 Mastectomy<br />

with 127 undergoing sentinel lymph node biopsy (111) or subsequent<br />

axillary surgery (16).<br />

Of 162 patients undergoing mastectomy for DCIS, 121 (75%)<br />

underwent axillary staging, with 9 (7%) having involved nodes.<br />

Screening mammography detected 117 (72%) but 45 (28%) presented<br />

symptomatically with a breast mass. Median age at diagnosis was<br />

57.8 years (range 29-94). Women less than 35 years of age had<br />

tumours sized greater than 4 cm in 72.3% compared to 59% in older<br />

women (p


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 4<br />

P3-14-06<br />

The Utility of Margin Index to Predict Residual DCIS Following<br />

<strong>Breast</strong> Conserving Surgery<br />

Aneja S, Lannin DR, Killelea B, Horowitz NR, Chagpar AB. Yale<br />

University School of Medicine, New Haven, CT<br />

Background: Although breast conserving surgery (BCS) is<br />

commonly performed for ductal carcinoma in situ (DCIS), patients<br />

may require re-excision for close or positive margins to rule out<br />

residual disease. The concept of margin index (defined as margin<br />

distance/tumor size) has been found to predict residual disease in<br />

patients with invasive breast cancer, but this concept has yet to be<br />

evaluated for patients with DCIS. We sought to determine if DCIS<br />

margin index (defined as the closest margin of DCIS/extent of DCIS)<br />

was predictive of residual disease.<br />

Methods: We performed a chart review of all in situ breast cancers<br />

treated with BCS at an academic breast center in 2009. DCIS margin<br />

index was calculated as closest margin distance of DCIS (mm) / extent<br />

of DCIS (mm). Non-parametric statistical analyses were performed<br />

to evaluate the relationship between margin index and residual DCIS.<br />

Results: 177 patients with DCIS were treated with BCS in 2009.<br />

Of these, 87 (49.1%) underwent resection of additional tissue for<br />

close margins or as routine practice, and had recorded quantitative<br />

margin distances and tumor sizes such that margin index could be<br />

calculated. Of these, 18 (20.7%) had residual DCIS in the additional<br />

tissue excised. Median patient age was 60.4 years (range; 31.9-91.4).<br />

Of the 50 patients with available immunohistochemistry analysis, 42<br />

(84%) were ER-positive and 36 (72%) were PR-positive respectively.<br />

12 (13.8%) had low grade DCIS, 53 (60.9%) intermediate grade<br />

DCIS, and 22 (25.3%) high grade DCIS. Median tumor size was<br />

16mm (range; 1-80 mm); and median margin distance was 2 mm<br />

(range; 0-18 mm). Median margin index was 0.125 (range; 0 to 18).<br />

Factors correlating with residual DCIS on univariate analysis were<br />

high tumor grade (33.3% vs. 23.2%, p=0.044), PR-negativity (66.7%<br />

vs. 22.7%, p=0.044), increasing tumor size (median 27.5 mm vs.<br />

15.0 mm, p=0.003), decreasing margin distance (median 1.0 mm vs.<br />

2.1 mm, p=0.004), and lower margin index (median 0.032 vs. 0.200,<br />

p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P4-01-02<br />

Association of DCE-MRI texture features with molecular<br />

phenotypes and neoadjuvant therapy response in breast cancer<br />

Banerjee N, Maity S, Varadan V, Janevski A, Kamalakaran S, Sikov<br />

W, Abu-Khalaf M, Bossuyt V, Lannin D, Harris L, Cornfeld D,<br />

Dimitrova N. Philips Research North America; Yale Comprehensive<br />

<strong>Cancer</strong> Center; Warren Alpert Medical School of Brown University;<br />

Yale-New Haven Hospital; Yale <strong>Breast</strong> <strong>Cancer</strong> Program; Seidman<br />

<strong>Cancer</strong> Center<br />

Purpose: MRI imaging phenotype features such as volume and<br />

morphology are used to characterize tumor heterogeneity and tumor<br />

response. Texture-based imaging features are important in lesion<br />

characterization but their relationship to molecular phenotypes and<br />

response is unclear. Molecular stratification of breast cancer into<br />

luminal, basal, ERBB2, and normal-like can be made based on<br />

gene expression profiles. We investigate how texture-based imaging<br />

features relate to tumor biology, genetic subtypes and neoadjuvant<br />

therapy response using MRI, histopathological and RNA-sequencing<br />

data.<br />

Materials and Methods: Data from 74 Stage IIA to IIIB breast<br />

cancer patients enrolled in neo-adjuvant clinical trials NCT00617942<br />

and NCT00723125 were retrospectively reviewed. We evaluated 37<br />

gray-level co-occurrence matrix features (GLCM) on post-contrast T1<br />

fat-suppressed images of 38 HER2- tumors and 35 HER2+ tumors.<br />

The texture features included angular second moment, contrast,<br />

correlation, first diagonal moment, entropy, regularity, roughness,<br />

line likeness and other statistical summaries. We also performed<br />

RNA-sequencing on 23 tumors and compared RNAseq-based PAM50<br />

clustering with texture-based clustering. Patients with pCR and RCB<br />

class=I were determined to be responders and the rest were labeled<br />

non-responders. Wilcoxon signed rank test was used to compare<br />

luminal vs. basal, ER+ vs. ER- and PR+ vs. PR- tumors and determine<br />

the discriminative power of the texture features. We then performed<br />

hierarchical clustering on our patient data set based on the significant<br />

texture features and evaluated their association with subtypes,<br />

hormone receptor status and response. Statistical significance of<br />

clusters was determined by hypergeometric test.<br />

Results: We found five MRI texture features to be significantly<br />

associated with tumor subtypes: first diagonal moment, contrast range,<br />

correlation range and entropy range (p


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 4<br />

[1] Arasu et al. Acad Radiol 2011;18:716-721.<br />

[2] Yankeelov & Gore. CMIR 2009;3:91-107.<br />

[3] Hylton et al. Radiology. 2012;263:663-672.<br />

P4-01-04<br />

MRI enhancement in stromal tissue surrounding breast tumors:<br />

Association with RFS following neoadjuvant chemotherapy<br />

Jones EF, Sinha SP, Newitt D, Klifa C, Kornak J, Park CC, Hylton<br />

NM. University of California, <strong>San</strong> Francisco, CA<br />

Objective: Stromal tissue surrounding breast tumors can harbor<br />

abnormalities that predict increased risk of recurrence. The purpose<br />

of this study was to evaluate low-level contrast enhancement patterns<br />

in normal appearing breast stroma surrounding tumors using dynamic<br />

contrast-enhanced magnetic resonance imaging (DCE-MRI).<br />

A secondary objective was to investigate whether enhancement<br />

patterns varied with distance from tumor and whether proximitydependent<br />

enhancement was associated with recurrence-free survival<br />

following neoadjuvant chemotherapy (NACT). Materials and<br />

Methods: Sixty-three patients with locally-advanced breast cancer<br />

were imaged with DCE-MRI before (V1) and after one cycle (V2)<br />

of adriamycin-cytoxan (AC) therapy. Normal-appearing stromal<br />

tissue on MR images was defined as fibroglandular tissue outside<br />

of tumor regions. Fibroglandular tissue was segmented from precontrast<br />

T1-weighted images using a fuzzy C-means clustering<br />

method 1 . Tumor regions were identified using a percent enhancement<br />

threshold of 70% in the first post-contrast image (PE1). Distance<br />

from tumor (proximity) was defined for each stromal voxel as the<br />

minimum three-dimensional distance from any tumor voxel. Stromal<br />

enhancement was characterized by calculating mean PE and mean<br />

signal enhancement ratio (SER=PE1/PE2) in distance shells of 5<br />

mm, from 0 to 40 mm outside of tumor tissue. Global PE and SER<br />

were calculated by averaging all stromal voxels from 5 to 40 mm<br />

from tumor. Proximity-dependent PE and SER were analyzed using<br />

a linear mixed effects model and Cox proportional hazards model for<br />

recurrence-free survival. Results: The mixed effects model displayed<br />

a decreasing radial trend in PE at both V1(estimated mean = -0.39<br />

per mm, 95% CI (-0.50, -0.29), p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

from prior studies (7 lesions). Six lesions were malignant (all IDC)<br />

and 10 were benign (5 fibroadenomas, 4 cysts, 1 papilloma). Scan<br />

time (5 minutes) and in-plane resolution were equivalent between<br />

the CUBE and FSE images while slice thickness in CUBE (2 mm)<br />

was half that of FSE (4 mm). Lesion-to-fibroglandular tissue signal<br />

ratios (S L /S F ) were calculated for both FSE and CUBE; S L is lesion<br />

signal and S F is fibroglandular tissue signal. The signal ratios were<br />

assessed with ordinary least squares linear regression. Along with<br />

lesion signal intensity with respect to surrounding tissue, depiction<br />

of lesion morphology on T2-weighted images can also contribute<br />

to differential diagnosis in breast MRI. A radiologist with breast<br />

MRI expertise evaluated the resolution difference between the two<br />

sequences based on the depiction of lesion morphology and the<br />

alignment of lesions between the T2-weighted and DCE images<br />

Results: S L /S F showed a correlation coefficient between the two<br />

methods of 0.93. Mean values of S L /S F for different methods and<br />

lesions were, FSE malignant: 1.01 ± 0.21, FSE benign: 2.05 ± 0.65,<br />

CUBE malignant 0.99 ± 0.25, CUBE benign 2.16 ± 0.62. Depiction<br />

of lesion morphology and signal intensity in small lesions improved<br />

in the CUBE images due to the reduced partial volume effect in the<br />

slice direction. Alignment of structures between T2-weighted and<br />

DCE images was facilitated with CUBE due to the higher throughplane<br />

resolution and the ability to reformat the images in orientations<br />

other than the acquisition plane.<br />

Conclusion: CUBE provides equivalent contrast and improved<br />

resolution in the breast in comparison to FSE. While T2-weighted<br />

images will continue to be an adjunct to DCE images, 3D T2-weighted<br />

sequences like CUBE have the potential to expand the contribution of<br />

T2-weighted images to the characterization of benign and malignant<br />

lesions in the breast.<br />

P4-01-07<br />

The Relationship of <strong>Breast</strong> Density in Mammography and<br />

Magnetic Resonance (MR) Imaging in a High Risk Population<br />

Chun J, Refinetti AP, Schnabel F, Leite AP, Price A, Billig J, Schwartz<br />

S, Moy L. NYU Langone Medical Center, New York, NY<br />

Background<br />

Mammographic breast density (BD) is associated with a 4 to 6-fold<br />

increased risk for developing breast cancer. Increased breast density<br />

on mammography is also known to decrease the diagnostic sensitivity<br />

of the exam, which is of great concern to women at increased risk for<br />

breast cancer. There is well-established medical literature describing<br />

the benefit of MRI in screening high risk women. A previous study has<br />

shown that background parenchymal enhancement (BPE) as measured<br />

on MRI can be correlated with breast cancer risk. The purpose of this<br />

study was to evaluate the relationship between BD, BPE, and FGT<br />

(assessment of the amount of fibroglandular tissue with contiguous<br />

MR images through both breasts) in pre and post-menopausal women<br />

who are at high risk for developing breast cancer.<br />

Methods<br />

The High Risk <strong>Breast</strong> <strong>Cancer</strong> Consortium (HRBCC) database at NYU<br />

Langone Medical Center was queried for our study population. A total<br />

of 56 women had both screening mammograms and MRIs completed<br />

at our center. Variables of interest included BD, BPE, FGT, BMI, Gail<br />

scores, and menopausal status. BD was defined by ACR classifications<br />

1-4. FGT was assessed on a similar scale 1-4. BPE was categorized<br />

as minimal, mild, moderate, or marked. BMI (kg/m2) was grouped<br />

into 4 categories: underweight (≤18), normal (19-24), overweight<br />

(25-29), and obese (≥30). Statistical analyses were performed using<br />

Spearman Correlation Coefficients.<br />

Results<br />

The median age in our cohort was 48 years (range 24-70 years). The<br />

majority of women were Caucasian (74%) with a median 5-year<br />

Gail score of 3.35. There was no correlation between BD and BPE<br />

(r=0.08) and BPE and FGT (r=0.18). However, there was a moderate<br />

positive correlation between BD and FGT (r=0.65). When we stratified<br />

by menopausal status, the correlation for BD and FGT was stronger<br />

for post-menopausal women (r=0.73) versus premenopausal women<br />

(r=0.57). When we stratified by BMI, we found a stronger positive<br />

association for BD and FGT among women who were overweight<br />

and obese (r=0.62).<br />

Clinical Characteristics<br />

VARIABLES TOTAL N=56 (%)<br />

FHBC<br />

Positive 35 (63)<br />

Negative 21 (37)<br />

BRCA 1,2<br />

Positive 17 (30)<br />

Negative 39 (70)<br />

AH, LCIS<br />

AH 18 (32)<br />

LCIS 12 (21)<br />

BD<br />

fatty 3 (5)<br />

scattered 11(20)<br />

heterogeneously dense 20 (36)<br />

dense 22 (39)<br />

BPE<br />

minimal 19 (34)<br />

mild 13 (23)<br />

moderate 14 (25)<br />

marked 10 (18)<br />

FGT<br />

fatty 10 (18)<br />

scattered 8 (14)<br />

heterogeneously dense 21 (38)<br />

dense 17 (30)<br />

BMI<br />

underweight ≤18 0 (0)<br />

normal 19-24 43 (77)<br />

overweight 25-29 9 (16)<br />

obese ≥30 4(7)<br />

Conclusions<br />

In our study cohort of patients at high risk for developing breast<br />

cancer, BD and BPE were not correlated, even after adjusting for<br />

menopausal status. This implies that BD and BPE may represent<br />

different characteristics of breast tissue and may have different<br />

implications. We found a strong correlation between BD and FGT.<br />

This association was strongest in women who were overweight and<br />

obese. FGT is a more objective and quantitative measurement of<br />

breast density and may be more useful in quantitative breast cancer<br />

risk assessment. Further studies are necessary to determine if BPE<br />

and FGT are independent risk factors for breast cancer.<br />

P4-01-08<br />

Effect of bilateral salpingo-oophrectomy on breast MRI<br />

fibroglandular volume and background parenchymal<br />

enhancement for BRCA 1/2 mutation carriers.<br />

DeLeo, III MJ, Domchek S, Kontos D, Conant E, Weinstein S. Hospital<br />

of the University of Pennsylvania, Philadelphia, PA<br />

PURPOSE: Bilateral salpingo-oophrectomy (BSO) reduces breast<br />

cancer risk in women with BRCA1/2 mutations by approximately<br />

50%. Annual screening for these high-risk women includes breast<br />

MRI and mammography. It is unknown whether BSO affects<br />

fibroglandular volume (FG) and background enhancement (BG) on<br />

breast MRI in this population. The purpose of this study is to assess<br />

the difference in FG and BG in BRCA1/2 mutation carriers before<br />

and after BSO on breast MRI.<br />

METHOD AND MATERIALS: We compared FG and BG on<br />

contrast-enhanced breast MRI before and after BSO. Two readers<br />

blinded to both clinical and imaging history scored FG according<br />

to the BI-RADS criteria from 1-4: fatty, scattered, heterogeneously<br />

dense, or extremely dense. BG was graded based on the MRI BI-<br />

RADS scale from 1-4: minimal, mild, moderate, and marked. Average<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

354s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 4<br />

BIRADS scores of each reader were used to calculate mean FG and<br />

BG (±SD) before and after BSO, and compared using a paired t-test<br />

for significance.<br />

RESULTS: We examined 60 women with BRCA1/2 mutations who<br />

underwent breast MRI before and after BSO from 2001-2011 at our<br />

institution. Five patients were excluded from the final analysis as they<br />

underwent mastectomy or bilateral radiation therapy before post-BSO<br />

MRI. Mean time to post-BSO MRI was 8.3 months ± 7 months.<br />

Mean pre- and post-operative FG were 2.64±0.78 and 2.58±0.75,<br />

respectively (p=0.622). Mean pre- and post-operative BG were<br />

2.46±0.93 and 1.87±0.81, respectively (p=0.0001). <strong>Breast</strong> cancer was<br />

detected in 8 women at a median time of 3.4 years following BSO.<br />

In these 8 women, mean pre- and post-operative FG were 3.10±0.57<br />

and 2.55±0.69, respectively. Mean pre- and post-operative BG were<br />

3.20±0.67 and 2.35±0.71, respectively.<br />

CONCLUSION: In this population of 55 BRCA1/2 mutation carriers<br />

who underwent breast MRI before and after BSO, there was a<br />

significant reduction in BG following BSO. There was no difference<br />

in FG on MRI following BSO. For the 8 patients in this cohort who<br />

were diagnosed with breast cancer after BSO, there was a trend toward<br />

higher FG and BG both before and after BSO, in comparison to the<br />

patients who were not diagnosed with breast cancer, suggesting that<br />

rates of higher FG and BG may be associated with increased risk for<br />

developing breast cancer.<br />

P4-01-09<br />

Screening breast magnetic resonance imaging (MRI) in earlystage<br />

breast cancer survivors<br />

Dowton AA, Meyer ME, Ruddy KJ, Yeh ED, Partridge AH. Dana-<br />

Farber <strong>Cancer</strong> Institute, Boston, MA; Brigham & Women’s Hospital,<br />

Boston, MA<br />

Background:<br />

Limited evidence is available regarding the value of screening breast<br />

MRI in female early stage breast cancer survivors. We sought to<br />

evaluate outcomes of index screening breast MRI in breast cancer<br />

survivors in the modern era of breast cancer treatment and imaging.<br />

Methods:<br />

Using a large prospective institutional database, we identified all<br />

women who underwent breast MRI between 7/2006 and 6/2008, were<br />

≥ 1 year and ≤ 3 years after diagnosis of stage I-IIIa breast cancer, had<br />

remaining breast tissue after treatment, and had consented to a record<br />

review protocol at our center (95% uptake rate). Sociodemographic,<br />

disease, staging and treatment information (see table 1), indications<br />

and outcomes for breast MRI, as well as disease status at 2 years after<br />

the index MRI were ascertained.<br />

Results:<br />

643 patients were identified as having undergone at least 1 MRI of the<br />

breast(s); 248 (38.5%) were diagnostic MRIs (had identified clinical<br />

indication or imaging abnormality); 12 (2%) were missing clinical<br />

data; 383 (59.5%) were screening MRIs. 338/383 (88%) patients who<br />

had screening MRI had stage I-IIIa invasive breast cancer. 220/338<br />

(65%) MRIs performed were index MRIs (index MRI defined as<br />

first MRI >1 year post diagnosis). For this preliminary analysis, we<br />

include only the 155 patients (70%) who were within 1-3 years of<br />

diagnosis at the time of index MRI (see table 1 for patient and disease<br />

characteristics).<br />

24/155 (15.5%) screening MRIs were considered not normal. Five<br />

women (21%) underwent biopsy based on abnormal MRI finding; 1<br />

resulted in the detection of a local recurrence (4.2%, 95% CI=0.1%-<br />

21.1%). 3/131 patients who had a normal index MRI were found<br />

to have local/regional recurrence or contralateral breast cancer<br />

diagnosed within 2 years of the MRI (2.3%, 95% CI=0.5%-6.6%). A<br />

statistically significant difference in the proportions was not observed<br />

based on Fisher’s test (p=0.50). 6/155 patients (3.9 %) had a systemic<br />

recurrence in the 2 years following index MRI.<br />

Table 1. Patient and Disease Characteristics<br />

Characteristic (N=155) n (%)<br />

Age at diagnosis (yrs)<br />

Median (Min-Max) 48 (28-77)<br />

0 - =70 2 (1%)<br />

Race<br />

Caucasian Non-Hispanic 144 (93%)<br />

African-American Non-Hispanic 1 (1%)<br />

Other 10 (6%)<br />

Genetic Mutation Status<br />

BRCA 1 Positive 0 (0%)<br />

BRCA 2 Positive 1 (0.6%)<br />

Indeterminate mutation of unknown significance 6 (3.9%)<br />

Negative 35 (22.6%)<br />

Not Tested 113 (72.9%)<br />

Family History of <strong>Breast</strong> <strong>Cancer</strong><br />

Yes 77 (49.7%)<br />

No 78 (50.3%)<br />

Tumor Stage<br />

I 83 (54%)<br />

IIA 34 (22%)<br />

IIB 26 (17%)<br />

IIIA 12 (8%)<br />

Type of definitive surgery<br />

BCS 92 (59%)<br />

Mastectomy 63 (41%)<br />

Hormone receptor status<br />

HR+ 125 (81%)<br />

HR- 30 (19%)<br />

HER2 status<br />

High pos 27 (17%)<br />

Negative 123 (79%)<br />

Low pos 4 (3%)<br />

Unknown 1 (1%)<br />

Conclusion:<br />

The vast majority of first screening breast MRIs obtained 1-3 years<br />

following treatment in early stage breast cancer survivors are normal,<br />

and very few detect cancer. Planned further analysis in this dataset<br />

will evaluate frequency of cancer detection in longer-term survivors.<br />

Additional research is necessary to determine the risks and benefits of<br />

screening breast MRIs being performed in this population.<br />

P4-01-10<br />

High-resolution diffusion weighted imaging for the separation<br />

of benign from malignant BI-RADS 4/5 lesions found on breast<br />

MRI at 3 Tesla<br />

Wisner DJ, Rogers N, Deshpande VS, Newitt DN, Laub GA, Porter<br />

DA, Joe BN, Hylton NM. University of California, <strong>San</strong> Francisco, CA;<br />

Siemens Medical Solutions, USA, Inc, <strong>San</strong> Francisco, CA; Siemens<br />

Medical Solutions, Erlangen, Germany<br />

Diffusion weighted imaging (DWI) has shown potential for separating<br />

malignant from benign lesions on breast MRI, theoretically improving<br />

the specificity of the breast MRI exam. However, standard DWI<br />

is hampered by multiple factors in the breast which limit utility<br />

in clinical practice. A readout segmented diffusion technique<br />

(RESOLVE)(1) permits the use of extremely short echo spacing and<br />

resamples uncorrectable data using a real-time navigator, reducing<br />

image distortion and potentially providing more accurate apparent<br />

diffusion coefficient (ADC) within lesions. In order to study the<br />

potentially utility of this method against standard diffusion, we<br />

compared ADC values from retrospectively-identified BI-RADS 4/5<br />

(suspicious) lesions with both standard single shot-spin echo EPI (ss-<br />

EPI) and RESOLVE diffusion at 3 Tesla over a 10-month period. All<br />

patients subsequently went to image-directed biopsy.<br />

The imaging parameters were as follows: TR/TE=7500-10000/60 ms<br />

(ss-EPI) and 8000-12000/64 ms (RESOLVE), averages=8 (ss-EPI)<br />

and 1 (RESOLVE), readout segmentation factor 5 with echo spacing<br />

of 0.3 ms (RESOLVE), slices = 47-50, b-values 0, 800, resolution<br />

= 1.8x1.8 x2.4 mm3, imaging times of approximately 5 minutes for<br />

both techniques. Freehand ROI’s were drawn on each suspicious<br />

lesion based on b=800 maps in close reference to the post-contrast<br />

T1 series by a board-certified radiologist who was blinded to final<br />

pathology and sequence. Similar ROI’s were drawn in normal tissue<br />

www.aacrjournals.org 355s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

as a control. For each lesion, mean ADC values and signal intensities<br />

at b=800 were recorded. ADC values were averaged by technique<br />

and pathologic outcome (benign or malignant). Signal intensity was<br />

normalized by dividing mean signal intensity of the lesion ROI by<br />

that of the control. Significance was determined using Wilcoxon<br />

rank-sum test for ADC, and paired t-test for signal intensity measures.<br />

Of the final cohort of 38 lesions in 31 patients, 9 were malignant<br />

and the remainder were benign. Two lesions were no longer present<br />

at breast MRI biopsy, and hence deemed benign. The lesion-tobackground<br />

signal intensity on diffusion was higher (p=0.05) on<br />

RESOLVE (1.9±0.1) compared to standard diffusion (1.7±0.1).<br />

Statistically significant differences between benign and malignant<br />

lesion were observed for mean ADC obtained by both methods<br />

(Table 1; p


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 4<br />

a focus or foci. Referring physicians provided information regarding<br />

the follow-up status of 88/154 (57%) lesions in noncompliant patients,<br />

of which 44/88 (50%) were followed by mammogram instead of MRI.<br />

Among compliant patients, 7/178 (3.9%) lesions seen on follow-up<br />

MRI were found to be high risk or malignant.<br />

Conclusion<br />

Compliance with follow-up MRI recommendation after core needle<br />

biopsy is low. Three characteristics were found to be associated with<br />

noncompliance with follow-up imaging: referral from non-affiliated<br />

physician, screening MRI, and a focus lesion. Moreover, patients who<br />

do not comply with recommended MRI follow-up are more likely to<br />

have follow-up mammography. Follow-up imaging among compliant<br />

patients found high-risk lesions and malignancy. Facilities performing<br />

MRI-guided core biopsies should therefore be aware of a high risk<br />

of noncompliance with follow-up.<br />

P4-01-13<br />

Practice patterns of MRI utilization for breast cancer treatment<br />

within the University of California system as part of the Athena<br />

initiative<br />

Tokin CA, Ojeda H, Mayadev JS, Hylton NM, Fowble BL, Rugo HS,<br />

Hwang S, Hurvitz S, Wells C, Blair SL. University of California, <strong>San</strong><br />

Diego; University of California, Davis; University of California, <strong>San</strong><br />

Francisco; Duke University; University of California, Los Angeles<br />

Background: The appropriate utilization of <strong>Breast</strong> MRI in breast<br />

cancer care remains controversial. As part of a quality improvement<br />

initiative for breast cancer screening and treatment, we sent out a<br />

survey to physicians who treat breast cancer patients. All respondents<br />

are participants in the ATHENA initiative, a program which unites<br />

physicians, researchers, and patients at the five University of<br />

California medical centers.<br />

Objective: To use the ATHENA infrastructure to perform a qualitative<br />

analysis of variations in breast cancer care.<br />

Methods: Surveys were sent to 50 physicians in the ATHENA network<br />

whose practices are focused on breast cancer. Respondents were<br />

presented with clinical scenarios, and asked whether they would<br />

recommend MRI always/usually or sometimes/never. Differences<br />

were compared by Chi square.<br />

Results: 39 physicians completed the survey (78% response rate).<br />

Of these physicians 29% were surgeons, 26% radiation oncologists<br />

and 45% medical oncologists. Athena physicians were more likely<br />

to order MRI for high risk screening of mutation carriers than not<br />

(85% yes vs. 15% no, p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P4-01-15<br />

Impact of Preoperative MRI on the Surgical Treatment of <strong>Breast</strong><br />

<strong>Cancer</strong>: A SEER-Medicare Analysis<br />

Thorsen CM, Weiss JE, Kerlikowske K, Ozanne EM, Buist DS,<br />

Hubbard RA, Tosteson AN, Henderson LM, Virnig BA, Goodrich ME,<br />

Onega TL. University of California, <strong>San</strong> Francisco, CA; Dartmouth<br />

Medical School, Lebanon, NH; University of Washington, Seattle,<br />

WA; University of North Carolina, Chapel Hill, NC; University of<br />

Minnesota, Minneapolis, MN<br />

Background:<br />

Preoperative magnetic resonance imaging (MRI) use has increased<br />

for women with invasive breast cancer and ductal carcinoma in situ<br />

(DCIS). Prior studies have disputed whether preoperative MRI is<br />

associated with increased rates of mastectomy. We evaluated the<br />

rates of mastectomy versus breast conserving surgery (BCS) and their<br />

association with preoperative MRI in older women.<br />

Methods:<br />

We identified women in SEER-Medicare 66 years or older diagnosed<br />

with DCIS or invasive breast cancer between 2002 and 2007 treated<br />

within 6 months of diagnosis with mastectomy or breast conserving<br />

surgery (BCS) with or without radiotherapy (RT). Preoperative<br />

MRI was defined as MRI occurring before a woman’s first surgery<br />

after her initial diagnosis. We looked for surgical treatment (BCS or<br />

mastectomy) and radiotherapy, age at diagnosis, year of diagnosis and<br />

cancer type, overall and separately by receipt of MRI. We examined<br />

the association of surgical treatment with MRI using multivariable<br />

logistic regression adjusting for age at diagnosis, year of diagnosis,<br />

cancer type and radiotherapy.<br />

Results:<br />

Among the 70,758 women identified, 5,126 (7.2%) had a preoperative<br />

MRI. The overall use of MRI increased from 1.2% in 2002 to 18.0%<br />

in 2007 (p


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 4<br />

P4-02-01<br />

18F-FDG PET/CT for the assessment of locoregional lymph node<br />

involvement and radiotherapy indication in stage II-III breast<br />

cancer treated with neoadjuvant chemotherapy<br />

Koolen BB, Valdés Olmos RA, Vogel WV, Vrancken Peeters M-JTFD,<br />

Rodenhuis S, Rutgers EJT, Elkhuizen PHM. Netherlands <strong>Cancer</strong><br />

Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, Netherlands<br />

Background: Risk for locoregional recurrence (LRR) in breast cancer<br />

depends on tumor size and on number and location of tumor-positive<br />

locoregional nodes. Postoperative irradiation of chest wall and/or<br />

locoregional nodes is advised in patients at high risk for LRR, but<br />

remains controversial in patients at intermediate risk. Because the<br />

number of tumor-positive axillary nodes in patients treated with<br />

neoadjuvant chemotherapy (NAC) can no longer be determined from<br />

the axillary lymph node dissection (ALND) and conventional staging<br />

of N3-disease with ultrasound is inaccurate, planning of radiotherapy<br />

in these patients is hampered. The aim of the present study was to<br />

assess the accuracy of 18F-FDG PET/CT for locoregional staging in<br />

stage II-III breast cancer patients scheduled for NAC. Second, we<br />

wished to assess the value of pre-chemotherapy PET/CT in estimating<br />

the risk for LRR and determining chest wall and/or locoregional<br />

radiotherapy indication, based on the detection of ≥4 FDG-avid<br />

axillary nodes or occult N3-disease.<br />

Methods: 278 patients underwent PET/CT of the thorax in prone<br />

position. Presence and number of FDG-avid nodes were evaluated,<br />

blinded for other diagnostic procedures.<br />

First, PET/CT findings were compared with histopathology before<br />

NAC (ultrasound with fine needle aspiration and/or sentinel lymph<br />

node biopsy (SLNB)) to determine the diagnostic accuracy for<br />

detection of axillary metastases and newly discovered N3-disease.<br />

Second, risk estimation and radiotherapy planning were evaluated<br />

with and without PET/CT. Conventional locoregional staging<br />

consisted of ultrasound with fine needle aspiration before, SLNB<br />

before, and ALND after NAC. Patients were classified as low-risk<br />

(cT2N0), intermediate-risk (cT0N1, cT1N1, cT2N1, and cT3N0), or<br />

high-risk (cT3N1, cT4, cN2-3, and (y)pN2-3) for LRR. Emphasis was<br />

on the number of patients upstaged to the high-risk group by PET/<br />

CT, requiring postoperative locoregional irradiation.<br />

Results: Sensitivity, specificity, positive predictive value, and negative<br />

predictive value in the detection of axillary metastases were 80%,<br />

92%, 98%, and 53%, respectively. Occult lymph node metastases in<br />

the internal mammary chain and periclavicular area were detected in<br />

17 (6%) and 25 (9%) patients, respectively.<br />

PET/CT detected occult N3-disease in 5 (11%) of 47 low-risk patients.<br />

In 116 intermediate-risk patients, PET/CT detected ≥4 FDG-avid<br />

nodes in 14 (12%) patients and occult N3-disease in 13 (11%) patients,<br />

upstaging 25 (22%) of intermediate-risk patients. In total, 30 (18%) of<br />

278 low- and intermediate-risk patients were upstaged to the high-risk<br />

group, requiring chest wall and/or locoregional irradiation.<br />

Conclusion: In breast cancer patients scheduled for NAC, PET/CT<br />

renders pre-chemotherapy SLNB unnecessary in case of an FDGavid<br />

axillary node and detects occult N3-disease in 15% of patients.<br />

Pre-chemotherapy PET/CT is a valuable tool for selection of highrisk<br />

patients who will benefit from locoregional radiotherapy. In<br />

our population, 18% of patients had an indication for locoregional<br />

irradiation based on PET/CT information, which was missed by<br />

conventional staging.<br />

P4-02-02<br />

Comparison of 18F-FDG PET-CT and 11C-MET PET-CT for<br />

assessment of response to neoadjuvant chemotherapy in locally<br />

advanced breast carcinoma.<br />

Dinesh A, Ramteke VK, Chander J, Tripathi M, Mahajan S. Maulana<br />

Azad Medical College, New Delhi, Delhi, India; Institute of Nuclear<br />

Medicine & Allied Sciences, New Delhi, Delhi, India<br />

INTRODUCTION<br />

Neo-adjuvant chemotherapy plays an important role in treatment of<br />

breast cancer by decreasing the tumor load & it offers an opportunity<br />

to evaluate response of primary tumor to chemotherapy. This makes<br />

response monitoring crucial in the management of breast carcinoma.<br />

Standard anatomical imaging modalities are unable to accurately<br />

reflect the response to chemotherapy until several cycles of drug<br />

treatment have been completed. Metabolic imaging using tracers like<br />

18F-fluorodeoxyglucose (FDG) as a marker of glucose metabolism<br />

or amino acid tracers like L-methyl-11C methionine (MET) have<br />

potential role for the measurement of treatment response in breast<br />

cancer.<br />

AIMS AND OBJECTIVES<br />

To compare FDG PET-CT with MET PET-CT for assessment of<br />

response to neoadjuvant chemotherapy, in locally advanced breast<br />

carcinoma and to correlate it with clinico-pathological response.<br />

METHODS<br />

In our prospective study, 20 patients with histology proven locally<br />

advanced breast carcinoma were enrolled. All patients underwent<br />

clinical assessment and PET-CT imaging using FDG and MET before<br />

and after 3 cycles of neoadjuvant chemotherapy (CAF regimen).<br />

Thereafter, all patients were taken for modified radical mastectomy<br />

and the resected breast with dissected axillary nodes was sent for<br />

histo-pathological analysis. Tumor response to the neoadjuvant<br />

chemotherapy was evaluated clinically using WHO criteria and by<br />

PET-CT imaging using PERCIST criteria. Responses calculated were<br />

compared for statistical significance using paired t- test.<br />

RESULTS<br />

Mean SUV in FDG-PET for primary tumor (p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

for axillary metastasis by FDG-PET and MET-PET shows higher<br />

accuracy than clinical examination. Thus, making PET-CT a valuable<br />

monitoring tool for guiding the choice of surgical management of<br />

axilla.<br />

P4-02-03<br />

FDG PET/CT for early monitoring of response to neoadjuvant<br />

chemotherapy in breast cancer patients.<br />

Andrade W, Soares F, Lima E, Maciel MdS, Toledo C, Iyeyasu H, Cruz<br />

M, Fanelli M. A. C. Camargo <strong>Cancer</strong> Hospital, São Paulo, SP, Brazil<br />

Introduction: Neoadjuvant chemotherapy (NAC), initially used only<br />

for locally advanced breast cancer, is now commonly used in patients<br />

with operable but large breast cancer or unfavorable tumor/breast size<br />

index because increases the chances of performing breast conservative<br />

surgery (BCS) instead of mastectomy in this group of patients.<br />

Patients and Methods<br />

This is a prospective unicenter trial. FDG PET/CT were performed in<br />

40 patients at baseline and after the second cycle of NAC. The level<br />

and relative changes in standardized uptake value (SUV) of FDG<br />

uptake were assessed regarding their ability to predict histopathologic<br />

response.<br />

Pathologic complete response (pCR) was defined as no malignant<br />

cells (no invasive ductal carcinoma and no ductal carcinoma in situ)<br />

identifiable in sections from the site of the tumor in the breast and in<br />

the axillary lymph node.<br />

Results<br />

This prospectively study analyzed forty patients undergoing NAC,<br />

all female, age ranged 27-64 years (mean 41.0 years and median 38<br />

years), all tumors were invasive ductal carcinoma, histological tumor<br />

grade 1, 2 and 3 were present respectively at 5%, 38.5% and 100%<br />

of the sample and nuclear grade 2 and 3 were present respectively at<br />

12.5% and 87.5%. Estrogen receptor was positive in 60% of patients<br />

and the progesterone receptor was positive in 47.5% of patients. Her<br />

2 was overexpressed in 12 patients (30%). Phenotype in this sample<br />

had the following distribution: 12.5% luminal A (5 patients), 50%<br />

Luminal B (20 patients: 14 patients with Ki-67> 14% and 6 cases<br />

with HER 2 overexpression), 15 % HER 2 (6 patients) and cases triple<br />

negative 9 (22.5%). size of the primary tumor ranged from 4.10 cm to<br />

12.0 cm (mean 7.10 cm). The size of the primary tumor ranged from<br />

had a mean of 6.7 cm and a median of 6.0 cm. This group showed<br />

great uniformity in relation to primary chemotherapy. NAC with<br />

cyclophosphamide and adriamycin were administered to 38 patients.<br />

In this study, pCR was obtained in 12 patients (30%). Baseline FDG<br />

SUV referring to pCR group was 11.26 and 7.26 in non-pCR group (p<br />

= 0.04). FDG SUV after second cycle was 2.73 in pCR group and 4.64<br />

in non-pCR group (p = 0.048). When analyzing ∆SUV (difference<br />

between baseline SUV and SUV after second cycle), pCR group<br />

had a mean reduction of 81.58% and non-pCR group had a mean<br />

reduction of 81.58% (p = 0.001). Receiver operating curve analyses<br />

were performed to deter mine optimal differentiation cut-off values<br />

of ∆SUV for differentiation of pCR and non-pCR. After two courses<br />

of NAC the optimal cutoff value to early differentiation between pCR<br />

from non-pCR were 59,1% in decrease of SUV. The sensitivity and<br />

specificity were 64,3% and 100,0%, respectively.<br />

Conclusion<br />

The optimum role of FDG PET in predicting the response of breast<br />

cancers to neoadjuvant chemotherapy is still not clearly defined and<br />

the SUV cut-off needs to be validated. FDG-PET allows for prediction<br />

of treatment response by the level of FDG uptake in terms of SUV at<br />

baseline and after two cycles of chemotherapy and FDG-PET may be<br />

helpful for individual treatment stratification in breast cancer patients.<br />

P4-02-04<br />

Tissue oxyhemoglobin dynamics measured with functional optical<br />

imaging immediately after starting chemotherapy correlates<br />

with markers of cellular proliferation and inflammation in a rat<br />

breast tumor model.<br />

Ueda S, Roblyer D, Cerussi A, Saeki T, Tromberg B. Beckman Laser<br />

Institute, University of California, Irvine, Irvine, CA; Saitama Medical<br />

University, Hidaka, Saitama, Japan<br />

We have reported that a transient increase of oxyhemoglobin level<br />

measured 24 hours after the start of neoadjuvant chemotherapy using<br />

noninvasive Diffuse Optical Spectroscopic Imaging (DOSI) may be<br />

useful in discriminating nonresponding from reponding breast cancer<br />

patients (PNAS. 2011 Aug 30;108(35):14626-31). To elucidate the<br />

underlying biological mechanism, we compared optical measured<br />

hemoglobin levels to tissue biomarkers in a rat tumor model.<br />

Method: An orthotopic mammary tumor model using Fischer 344<br />

rats and MAT3B cells was used for this experiment. 16 mg of<br />

cyclophosphamide was administered (i.p.) in 15 rats after tumors<br />

had grown to a significant size. A wide-field functional imaging<br />

device that utilizes structured near-infrared light was used to measure<br />

superficial absolute concentrations of oxyhemoglobin (ctO2Hb) and<br />

deoxyhemoglobin (ctHHb) of the tumors. Rats were imaged and<br />

sacrificed (two per timepoint) at each of the following timepoints:<br />

baseline, 2, 6, 12, 24, 48, 96 hours, and day 8 following the injection.<br />

Resected tumors were sectioned after paraffin-embedding and<br />

representative slides were used for immunohistochemistry. Cellular<br />

markers of proliferation (measured by Ki67), apoptosis (Caspase3),<br />

NK cells (ANK61), and macrophages (MAC387) were counted in<br />

each slide using Image J software.<br />

Result: Average tumors baseline values of ctO2Hb and ctHHb were<br />

81.4 µM (±9.2 SD) and 22.4 µM (±4.1 SD) respectively. In most<br />

rats, both levels gradually increased from baseline to 24 hours after<br />

the injection and reached a maximum at 48 hours and then declined<br />

until Day 8. At 24 hours, ctO2Hb was significantly higher than<br />

baseline [96.8 µM (±8.8 SD) vs. 81.4µM (±9.2 SD), p = 0.003 using<br />

Student’s t-test]. This trend was mimicked in the tissue markers of<br />

cellular proliferation and apoptosis, which peaked at 24 through 48<br />

hours and then dropped. The average NK cell count was also found<br />

to rise consistently through the course of monitoring and reached the<br />

highest at day 8, while macrophage counts reached a peak at 48 hours<br />

and then declined. The differences between baseline and 24 hours for<br />

these were statistically significant: Ki67 [294 (±107 SD) vs. 115 (±68<br />

SD), p = 0.0002], Caspase3 [2916 (±1383 SD) vs. 136 (±150 SD), p<br />

< 0.0001], NK cells [635 (±329 SD) vs. 159 (±110 SD), p = 0.0001],<br />

and macrophage markers [281 (±260 SD) vs. 39 (±31 SD), p = 0.01].<br />

Average values of hemoglobin and biological markers at baseline and after injection in 15 rats.<br />

Unit Baseline 12hrs 24hrs 48hrs 96hrs Day8<br />

ctHbO2 uM 81.4 86.6 96.8* 113.6* 71.5 57.8*<br />

ctHHb uM 22.4 22.8 25.6 30.8* 19.8 25<br />

Size mm 5.7 6.4 7.4* 8.5* 4.8 5.7<br />

Ki67 counts/HPF 115 62 294* 225* 22*<br />

Caspase3 counts/HPF 136 445* 2916* 3307* 140<br />

NK counts/HPF 159 541* 635* 587* 1380*<br />

Mac counts/HPF 39 125* 281* 1143* 171*<br />

* Significant difference between baseline and measurement time, p ≤ 0.01<br />

Conclusion: We observed an increase in oxyhemoglobin early after<br />

chemotherapy, closely linked to changes in cellular proliferation,<br />

apoptosis, NK cells and macrophage accumulation. Early optical<br />

changes may be linked to chemotherapy-induced tumor metabolic<br />

and acute inflammatory responses.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

360s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 4<br />

P4-02-05<br />

A novel 64Cu-liposomal PET agent (MM-DX-929) predicts<br />

response to liposomal chemotherapeutics in preclinical breast<br />

cancer models<br />

Lee H, Zheng J, Gaddy D, Kirpotin D, Dunne M, Drummond D, Allen<br />

C, Jaffray D, Hendriks B, Wickham T. Merrimack Pharmaceuticals,<br />

Boston, MA; Princess Margaret Hospital, University Health Network,<br />

Toronto, ON, Canada; Leslie Dan Faculty of Pharmacy, University<br />

of Toronto, ON, Canada<br />

Background: Liposomal anthracyclines (such as pegylated liposomal<br />

doxorubicin (PLD) or HER2-targeted liposomal doxorubicin (MM-<br />

302)) are being used and/or evaluated for the clinical management<br />

of breast cancer, but responses vary from patient to patient. It<br />

is hypothesized that variability in the deposition of liposomal<br />

therapeutics within tumors leads to differences in drug exposure,<br />

thereby directly influencing tumor response. We have developed<br />

MM-DX-929, a novel 64Cu-liposomal PET imaging agent, as a<br />

clinically-implementable tool to investigate whether image-based<br />

quantification of liposome deposition in tumors can predict treatment<br />

response to liposomal chemotherapeutics, including PLD, MM-302<br />

and/or liposomal irinotecan (MM-398).<br />

Objectives: Our primary objective is to demonstrate that the extent<br />

of tumor uptake of MM-DX-929 is predictive of tumor response to<br />

MM-302 in preclinical breast cancer xenograft models. A secondary<br />

objective is to enable clinical translation of MM-DX-929 to enable<br />

incorporation into existing therapeutic trials.<br />

Methods: Mice bearing BT474-M3 mammary and subcutaneous<br />

tumors were injected intravenously with MM-DX-929 prior to dosing<br />

with MM-302. PET/CT imaging was performed at 16h post MM-<br />

DX-929 injection, and tumor uptake was determined. Response to<br />

treatment was quantified as tumor volume changes measured over a<br />

2-month period by MRI.<br />

Results: Tumor deposition of MM-DX-929 administration correlated<br />

well with treatment response to MM-302 (Spearman correlation<br />

coefficient of -0.891 and a p-value of 0.0004). MM-DX-929<br />

accumulation in tumors prior to the start of the MM-302 treatment<br />

successfully predicted improved tumor growth inhibition following<br />

MM-302 treatment.<br />

Conclusion: These findings support trials of MM-DX-929 as a<br />

predictive imaging agent to select patients who are most likely to<br />

respond to liposomal therapies. The clinical development of MM-<br />

DX-929 for identification of breast cancer patients likely to respond<br />

to liposomal therapeutics is currently being pursued.<br />

P4-02-06<br />

Molecular imaging with trastuzumab and pertuzumab of HER2-<br />

positive breast cancer in mice: a step towards personalized<br />

medicine<br />

Collin B, Oudot A, Vrigneaud J-M, Moreau M, Raguin O, Duchamp<br />

O, Tizon X, Denat F, Varoqueaux N, Brunotte F, Fumoleau P.<br />

Centre Georges François Leclerc, Dijon, France; Institut de<br />

Chimie Moléculaire de l’université de Bourgogne, Dijon, France;<br />

Oncodesign, Dijon, France; Roche France, Boulogne Billancourt,<br />

France<br />

Introduction<br />

The accurate and reliable assessment of HER2 status is an essential<br />

step in the diagnostic workup and selection of targeted treatment<br />

strategy in breast cancer patients. However, current ex vivo methods<br />

do not evaluate HER2 status possible discordance between primary<br />

tumor and distant metastases, nor immediate response to therapeutic<br />

intervention. Non-invasive imaging of HER2 expression could<br />

be a relevant alternative with additional benefits such as a better<br />

selection of patients for HER2-targeted therapies and the possible<br />

optimization of treatment strategies. Recent clinical trials suggest<br />

a better efficacy of trastuzumab / pertuzumab doublet but a lower<br />

activity of pertuzumab monotherapy over trastuzumab alone.<br />

Aim<br />

The aim of our study was therefore to evaluate preclinically the<br />

use of radiolabeled DOTAGA (2,2’,2’’-(10-(2,6-dioxotetrahydro-<br />

2H-pyran-3-yl)-1,4,7,10-tetraazacyclododecane-1,4,7-triyl)triacetic<br />

acid) conjugated trastuzumab and pertuzumab for SPECT/CT (Single<br />

Photon Emission Computed Tomography/Computed Tomography)<br />

HER2 imaging purposes.<br />

Methods<br />

Trastuzumab and pertuzumab were conjugated to a new bifunctional<br />

chelating agent: DOTAGA anhydride (DOTA analog) and<br />

subsequently radiolabeled with the gamma-emitter Indium 111. The<br />

functionality of [111In]-DOTAGA-antibodies analogs was evaluated<br />

by in vitro saturation assays using HER2-overexpressing human breast<br />

cancer cell line (HCC1954). In vivo biodistribution was studied by<br />

SPECT/CT imaging at 24h, 48h and 72h after intravenous injection<br />

of [111In]-DOTAGA-antibodies in subcutaneous BT474 tumorbearing<br />

rodents.<br />

Results<br />

[111In]-DOTAGA-trastuzumab and [111In]-DOTAGA-pertuzumab<br />

showed respectively 2.6 and 2.7 DOTAGA /antibody. Both [111In]-<br />

DOTAGA-trastuzumab and [111In]-DOTAGA-pertuzumab achieved<br />

a radiochemical purity > 95%. Biological activity of [111In]-<br />

DOTAGA-trastuzumab and [111In]-DOTAGA-pertuzumab was<br />

maintained: the affinity determined on HER2 expressing HCC1954<br />

cell lines, was 5.5 and 5.6 nM. SPECT/CT imaging experiments in<br />

tumor-bearing rodents and gamma-counting of organs both showed<br />

that at 72h post-injection, more than 60 % of the activity was localized<br />

in the tumor (66.9±0.9 %ID/g for [111In]-DOTAGA-trastuzumab<br />

and 63.8±6.3 %ID/g for [111In]-DOTAGA-pertuzumab) and that<br />

an excess of non-radiolabeled corresponding antibody significantly<br />

shifted down tumor-targeting (to 17.6±2.4 %ID/g for [111In]-<br />

DOTAGA-trastuzumab and 8.7±0.8 %ID/g for [111In]-DOTAGApertuzumab.<br />

Conclusions<br />

Our results demonstrate a similar HER2 binding of trastuzumab<br />

and pertuzumab. Both radiolabeled [111In]-DOTAGA-trastuzumab<br />

and [111In]-DOTAGA-pertuzumab should be considered and<br />

might be promising tools for molecular imaging diagnosis of HER2<br />

overexpression in breast cancer.<br />

P4-02-07<br />

Early Optical Tomography Changes Predict <strong>Breast</strong> <strong>Cancer</strong><br />

Response to Neoadjuvant Chemotherapy<br />

Lim EA, Gunther JE, Flexman M, Kim HK, Hibshoosh H, Kalinsky<br />

K, Crew K, Maurer M, Taback B, Feldman S, Brown M, Refice S,<br />

Alvarez-Cid M, Hielscher A, Hershman DL. Columbia University<br />

Medical Center, New York, NY; Columbia University, New York,<br />

NY; Herbert Irving Comprehensive <strong>Cancer</strong> Center, New York, NY;<br />

Mailman School of Public Health, New York, NY<br />

Background: Pathologic complete response (pCR) or a low Residual<br />

<strong>Cancer</strong> Burden (RCB) score following neoadjuvant chemotherapy<br />

(NACT) predicts a superior survival in breast cancer (BC) patients.<br />

An early predictive marker of tumor response during NACT would<br />

provide a way to optimize treatment for non-responders; however,<br />

no ideal technology currently exists. Diffuse optical tomography<br />

www.aacrjournals.org 361s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

(DOT) is a novel, fast, safe, and low-cost technique that uses near<br />

infrared light to provide 3D data on tissue vascularity without the use<br />

of radiation, making it a promising technology for assessing early<br />

tumor response to NACT. We hypothesized that a 2-week change in<br />

DOT parameters would predict response to NACT as measured by<br />

the RCB score.<br />

Methods: Women with stage II-IIIc invasive BC scheduled to<br />

undergo NACT with 12 cycles of a weekly taxane followed by 4<br />

cycles of doxorubicin with cyclophosphamide (AC) were enrolled.<br />

Treatment with additional biologic therapies was allowed. DOT<br />

measurements were made before starting NACT, 2 weeks into<br />

treatment, and before surgery. Concentrations of oxyhemoglobin<br />

[HbO2], deoxyhemoglobin [Hb], total hemoglobin [HbT], and tissue<br />

scattering (SC) were measured by DOT. Final pathology specimens<br />

were scored for the RCB index (continuous measure), RCB class (0,<br />

1, 2, 3), and a dichotomized RCB score (RCB class 0 or 1: responders<br />

to NACT; RCB class 2 or 3: non-responders). Correlation analysis,<br />

ANOVA testing, and two sample t-tests were used to evaluate the<br />

relationship between the two-week changes in DOT parameters and<br />

the RCB score.<br />

Results: Since July 2011, we have recruited 11 pts, of whom 7 have<br />

undergone surgery. Complete data is available for 6 pts. Two of 7<br />

pts had a pCR (RCB 0), 1 had RCB 1, 3 had RCB 2, and 1 had RCB<br />

3. The Pearson correlation between the 2-week change in [Hb] and<br />

the continuous RCB index was 0.94 (p=0.0047), and that between<br />

the 2-week change in SC and the RCB index was 0.93 (p=0.0073).<br />

At 2 weeks, the [Hb] decreased by 6.7% for pts whose pathology<br />

demonstrated an RCB 0 (pCR), 1.8% for RCB 1, 0.6% for RCB 2, and<br />

increased 0.7% for RCB 3. ANOVA and Tukey testing demonstrated a<br />

significant difference in the [Hb] change for pts with RCB 0 compared<br />

to pts with RCB 1, 2, or 3 (p


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 4<br />

Sensitivity for the detection of breast malignancy<br />

Sensitivity<br />

Mammography 72%<br />

Ultrasound 63%<br />

BSGI 82%<br />

Mammography + Ultrasound 90%<br />

Mammography + Ultrasound + BSGI 98%<br />

A subsequent breast MRI detected one of the lesions missed by the<br />

three modalities while two lesions were found by pathology alone.<br />

Conclusions<br />

Of the three imaging modalities, BSGI provided the highest<br />

independent sensitivity and when added to the diagnostic workup,<br />

BSGI detected an additional 14 malignancies, increasing the<br />

sensitivity from 90% to 98%. Although it is beyond the scope of this<br />

work, it is interesting to note that the cost of the BSGI procedure<br />

is relatively low, about $320, and that in this population the BSGI<br />

specificity was 74%. In summary, when added to the diagnostic work<br />

up of patients in the community breast cancer, BSGI can improve<br />

the detection of breast malignancy when compared to mammography<br />

and ultrasound alone.<br />

P4-02-10<br />

Molecular <strong>Breast</strong> Imaging: A multicenter clinical registry to<br />

compare breast-specific gamma imaging (BSGI) and breast MRI<br />

in the detection of breast carcinoma<br />

Weigert JM, Kieper DA, Stern LH, Böhm-Vélez M. Hampton<br />

University, Hampton, VA; Thomas Jefferson University, Methodist<br />

Division, Philadelphia, PA; Mandell and Blau MDs PC, New Britain,<br />

CT; Weinstein Imaging Associates, Pittsburgh, PA<br />

Purpose<br />

<strong>Breast</strong>-Specific Gamma Imaging (BSGI), also known as molecular<br />

breast imaging (MBI), is a diagnostic breast imaging procedure<br />

using that is becoming more common in clinical practice. In the<br />

BSGI procedure, a patient receives an intravenous injection of<br />

Tc99m-Sestamibi (MIBI) and imaging is conducted with a breastoptimized<br />

gamma camera. According to the clinical literature, the<br />

largest advantage of BSGI when compared to mammography is that<br />

the sensitivity of the examination is not influenced by breast tissue<br />

density. The goal of this work is to quantify the clinical sensitivity<br />

of BSGI against that of MRI, another diagnostic imaging modality<br />

capable of detecting mammographically occult malignancies.<br />

Methods<br />

A multi-center patient registry was maintained for all patients<br />

routinely sent to BSGI and MRI as part of their diagnostic work<br />

up. All patients who had a malignant finding on subsequent needle<br />

biopsy or surgery within 12 months post imaging were included in<br />

this analysis. The BIRADS rating schematic was used for MRI and<br />

a similar, 6-category system was used for the BSGI images. For each<br />

modality, the reports were classified as Negative: category 1 – 3 and<br />

positive: category 4 - 6.<br />

Results<br />

There were 60 patients with 63 malignant breast lesions. MRI was<br />

negative in 6 lesions (2 DCIS, 3 IDC and 1 ILC) while BSGI was<br />

negative in 6 lesions (5 IDC and one mixed IDC/DCIS). Both BSGI<br />

and MRI detected 56 of the 63 malignancies providing a sensitivity<br />

of 89% for both modalities. The combination of BSGI and MRI<br />

detected 63 of the 63 malignancies for a sensitivity of 100%.[table 1]<br />

Conclusions<br />

According to the results of this patient registry, BSGI provides equal<br />

sensitivity to that of MRI however each modality was false-negative<br />

in different patients. As a result, while each modality provided a<br />

sensitivity of 89%, the combination of BSGI and MRI provided a<br />

sensitivity of 100%.<br />

Sensitivity for BSGI/MBI, breast MRI and the combination of both modalities<br />

Sensitivity<br />

BSGI/MBI 89%<br />

MRI 89%<br />

Combined BSGI + MRI 100%<br />

The MBI/BSGI procedure does involve a relatively small dose of<br />

radiation, but this dose is less than the dose patients receive from<br />

other common diagnostic examinations such as a chest CT scan.<br />

According to RadiologyInfo.org, a website co-developed by the<br />

American College of Radiology and Radiological Society of North<br />

America, “…there are no known long-term adverse effects from such<br />

low-dose exposure.” In comparison, MRI does not involve exposure<br />

to radiation but utilizes a contrast agent that has been reported to cause<br />

adverse reactions in a very small number of patients, especially those<br />

with insufficient kidney or liver function. In addition, MRI cannot<br />

be performed in some types of patients such as those with metal<br />

implants, or those with claustrophobia. In our experience, BSGI is<br />

a low-cost imaging procedure (the procedure costing approximately<br />

1/3 that of MRI) that is well tolerated by patients and can be used in<br />

patients who for whom MRI is contraindicated or very challenging.<br />

P4-03-01<br />

Distance of breast cancer from the skin influence axillary nodal<br />

metastasis<br />

Kim EJ, Chae BJ, Song BJ, Kwak HY, Chang EY, Kim SH, Jung SS.<br />

Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul,<br />

Republic of Korea<br />

Background<br />

In breast cancer, axillary lymph node status is one of the most<br />

important prognostic variables and a crucial component to the staging<br />

system. Moreover, cancers underlying skin are willing to access to<br />

the lymphatics and cause lymphatic dissemination. Therefore, there<br />

seems an association between proximity of breast cancer to the skin<br />

and the incidence of axillary node positivity or local recurrence.<br />

The aim of this study was to determine whether distance of breast<br />

cancers from the skin affects the the clinical and pathologic features<br />

including the likelihood of axillary nodal metastases, ipsilateral breast<br />

cancer recurrence, and recurrence free survival.<br />

Methods<br />

Between January 2003 and December 2009, data were collected<br />

prospectively regarding 1005 consecutive breast cancer patients<br />

who underwent surgical treatment. The exclusion criteria were noninvasive<br />

carcinoma (e.g. Ductal Carcinoma In Situ), prior breast<br />

surgery, previous radiotherapy or neo-adjuvant chemotherapy and<br />

previous endocrine therapy. The distance of the tumor from the skin<br />

surface was measured from the skin to the most anterior hypoechoic<br />

leading edge of the lesion.<br />

Results<br />

A total of 891 patients were included in the statistical analysis,<br />

603(68%) had no axillary nodal metastasis, 288(32%) had axillary<br />

nodal metastasis. Distance of breast cancer from the skin less than<br />

3mm induced more axillary nodal metastasis (P=0.039). However,<br />

there was no statistical significant correlation between distance of<br />

breast cancer from the skin less than 3mm and ipsilateral breast<br />

tumor recurrence (P=0.788) or recurrence-free survival (P=0.353).<br />

Conclusion<br />

<strong>Breast</strong> cancers located closer to the skin have a higher incidence of<br />

axillary nodal metastasis. When evaluating a breast cancer patient<br />

and considering the risk of nodal metastasis the distance of the tumor<br />

from the skin should be considered.<br />

www.aacrjournals.org 363s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P4-03-02<br />

Automatic BI-RADS Diagnosis of <strong>Breast</strong> Lesions by<br />

CAD(computer-aid diagnosis)<br />

Kuo W-H, Chuang S-C, Yang S-H, Chen C-N, Chen A, Chang K-J.<br />

National Taiwan University Hospital and National Taiwan University<br />

College of Medicine, Taipei, Taiwan; Graduate Institute of Industrial<br />

Engineering, National Taiwan University, Taipei, Taiwan<br />

Objective<br />

Ultrasound is one of the most effective non-invasive tool for breast<br />

tumor detection and diagnosis. However, acquisition and observation<br />

of ultrasound images are mostly subjective and highly dependent on<br />

personal experience and judgment. The inter-observer variation often<br />

results in significantly different decisions. Objective quantification<br />

of sonographic features has become a pressing issue facing the<br />

medical staff.<br />

This research is to develop a robust, well-performing CAD system<br />

which can provide objective suggestion for BI-RADS categorization.<br />

Materials & Methods<br />

Sonographic tumor features of 264 tissue-proved BI-RADS 3<br />

to 5 breast lesions (65 cancers, 199 benign cases)were collected<br />

consecutively by well-experienced single investigator using Siemens<br />

S2000 in National Taiwan University Hospital. There are initially<br />

30 attributes extracted from only one representative view of each<br />

tumor. These attributes based on the lexicon descriptions of BI-RADS<br />

includes characters such as shape, margin irregularity, heterogeneity,<br />

boundary, acoustic shadow, flexibility. After feature selection, 9<br />

attributes, including 6 B-mode attributes and 3 elastography-related<br />

attributes are taken into practical model building. All 9 attributes<br />

used in this research are quantitative features and are computed by<br />

computer. The only necessarily manual effort is to outline the tumor<br />

lesion on the ultrasound image by investigator.<br />

We randomly take 175 cases(42 cancers, 133 benign cases), about<br />

two-third of the sample, for model training and leave 89 cases<br />

(23 cancers, 66 benign cases)as the independent test data. In both<br />

training data and independent data, the ratio between benignancy<br />

and malignancy is kept even.<br />

Using “Parameterized Three-Phased Ensemble Model”, we transform<br />

the original binary predicting result into probabilistic form so that<br />

the model can be applied to BI-RADS category which is based on<br />

malignancy probability. In each ensemble model, every component<br />

classifier (Multi-Layer FLD Classification Tree) will be assigned<br />

a weight according to its performance by AdaBoost algorithm.<br />

The classifier gives back a real value as its predicting result. The<br />

categorizing probability threshold we adopt in this research follows<br />

standard BI-RADS system: BI-RADS 3


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 4<br />

represents a significant and unmet need for improved margin<br />

assessment. High reoperation rates present both increased treatment<br />

risk to patients and increased burden on healthcare systems. In the<br />

USA alone, over 150,000 lumpectomies are performed per year at<br />

an average cost between $11,000 and $19,000 USD per procedure.<br />

Assuming a relatively modest average repeat operation rate of 25%,<br />

potentially preventable repeat surgeries represent an approximate cost<br />

to the US healthcare system of $500M (USD) annually.<br />

Reducing the prevalence of repeat surgeries may be accomplished by<br />

providing faster and more accurate intraoperative tools for assessing<br />

margin widths during the time of the first surgery. One such potential<br />

technique involves the use of Optical Coherence Tomography (OCT)<br />

imaging, which uses light to produce images in much the same way<br />

that ultrasound produces images with sound. Compared to ultrasound,<br />

OCT provides decreased depth of penetration, but increased resolution<br />

capabilities. The increased resolution that OCT provides allows for the<br />

visualization of the internal cellular structure within a tissue sample<br />

and therefore, provides the potential ability to differentiate cancerous<br />

from normal or benign cells. We propose the use of an intraoperative<br />

OCT imaging system to provide near real-time imaging information<br />

about the internal structure of tissue samples excised during BCS<br />

procedures. Our hypothesis is that the overall rate of repeat operations<br />

can be reduced by providing a tool to assist surgeons with the task of<br />

margin width estimation during the time of surgery.<br />

We have developed an early stage prototype OCT imaging system<br />

that has completed laboratory phantom and preclinical studies. This<br />

paper will present the capabilities of an OCT imaging system to<br />

provide margin assessment information in biological breast tissue<br />

mimicking phantoms. The phantoms were designed to encompass<br />

imaging characteristics across a wide range of human breast densities.<br />

The paper will go on to describe preclinical imaging that was done in<br />

tumor specimens excised from human breast cancer rat models. The<br />

results obtained in the phantom and preclinical studies suggest the<br />

potential for OCT as a near-real time, intraoperative imaging tool to<br />

aid surgeons with breast lumpectomy margin width estimation. To<br />

help realize this potential, further research is required in to the use<br />

of OCT during BCS.<br />

P4-03-05<br />

The Clinical Trial of New Optical Mammography<br />

Ogura H, Yoshimoto K, Nasu H, Hosokawa Y, Matsunuma R, Ide<br />

Y, Yamaki E, Yamashita D, Suzuki T, Oda M, Ueda Y, Yamashita Y,<br />

Sakahara H. Hamamatsu University School of Medicine, Hamamatsu,<br />

Shizuoka, Japan; Hamamatsu Photonics K.K., Hamamatsu, Shizuoka,<br />

Japan<br />

Objectives: Optical mammography (O-MMG), which utilizes nearinfrared<br />

light for the detection of abnormal breast tissue, can be a<br />

potentially useful and non-invasive tool for the screening of breast<br />

cancer patients. Here we report on the preliminary results of our<br />

clinical trial using a time-resolved O-MMG system developed by<br />

Hamamatsu Photonics (Hamamatsu, Japan). In our past examination,<br />

negative results included cases with mismatch for breast cup because<br />

of small breast, lesion outside of cup because of big breast, and bad<br />

positioning. So, size and shape of a gantry was improved (3 cup sizes;<br />

S, M and L cup). We reported the initial experience of the improved<br />

O-MMG systems.<br />

Materials and Methods: In principle, the technique involves sequential<br />

irradiation of the breast tissue with a pulsed laser with a wavelength<br />

of 760, 800 and, 830nm, which is then detected by the variable<br />

capacity gantry at multiple sites after passing through the breast<br />

tissue. Measurement time has improved to about 7 minutes (a third<br />

of the conventional measurement time). The data are analyzed and<br />

tomographic images of the absorption coefficient of the breast are then<br />

reconstructed based on our tomographic reconstruction algorithm.<br />

Between October 2011 and May 2012, fifteen patients with breast<br />

cancer and one patient with fibroadenoma participated in the trial.<br />

Results: In patients with breast cancer, S cup was used for one patient,<br />

M cup for 12 patients and L cup for two patients. Five patients after<br />

neo-adjuvant chemotherapy were excluded from the analysis. The<br />

lesions were depicted as an area of high hemoglobin concentration<br />

in 7 of 10 patients with cancer. One lesion of fibroadenoma was also<br />

identified as an area of high hemoglobin concentration.<br />

Conclusion: Further examination with our new O-MMG systems<br />

will be of interest.<br />

P4-03-06<br />

Non-invasive classification of microcalcifications by the use of<br />

X-ray phase contrast mammography as a novel tool in breast<br />

diagnostics<br />

Hauser N, Wang Z, Kubik-Huch RA, Singer G, Trippel M, Roessl<br />

E, Hohl MK, Stampanoni M. Interdisciplinary <strong>Breast</strong> Center<br />

Kantonsspital Baden, Baden, Switzerland; Paul Scherrer Institut,<br />

Villigen, Switzerland; Kantonsspital Baden, Baden, Switzerland;<br />

Philips Research Laboratories, Hamburg, Germany<br />

Purpose: Phase-contrast and scattering-based x-ray imaging are<br />

known to provide additional and complementary information to<br />

conventional, absorption-based methods. The goal of this study is to<br />

evaluate this method to distinguish between benign and malignant<br />

microcalcifications with the benefit to increase accuracy of early<br />

breast cancer diagnosis. Phase-contrast mammography has been<br />

shown to increase image quality of native breast samples when<br />

compared to conventional mammography.<br />

Two major types of microcalcifications are found within breast<br />

tissue. Type I consist of calcium oxalate dehydrate and type II<br />

microcalcifications are composed of calcium phosphates. Type I is<br />

seen most frequently in benign lesions whereas type II is indicative<br />

for proliferative lesions, including carcinomas. Phase contrast<br />

mammography is shown here to distinguish between the two types of<br />

microcalcifications and therefore indicates a step forward to improve<br />

early breast cancer diagnosis.<br />

Material and Methods: Freshly dissected breast specimens (n=50)<br />

were imaged using a Talbot-Lau interferometer equipped with a<br />

conventional x-ray tube; the interferometer was operated at the fifth<br />

Talbot distance, tube voltage of 40 kVp with mean energy of 28 keV,<br />

and current of 25 mA. The device simultaneously recorded absorption,<br />

differential phase and small-angle scattering signals. These quantities<br />

were combined into novel, high-frequency-enhanced radiographic<br />

images. Histopathological analysis was performed and regions of<br />

interests correlated with the findings of phase contrast mammography.<br />

Results: Our novel imaging approach yields complementary and<br />

otherwise inaccessible information on electron density distribution<br />

and small-angle scattering power of the sample at microscopic scale.<br />

Recently we generated the world’s first phase contrast mammograms<br />

of native, not-fixed human whole breast samples. Our results<br />

indicate the superiority of the new technique with respect to image<br />

quality and lesion conspicuity. A clinical reader study is currently<br />

carried out. Exploiting the multiple, complementary information<br />

obtained by grating-based interferometry, we are able to classify<br />

microcalcifications by a non-invasive technique within the clinical<br />

environment.<br />

By considering the small-angle scattering signal as a complement<br />

to the absorption signal, our method can analyze the differences in<br />

www.aacrjournals.org 365s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

the attenuation coefficient as well as in the crystal structure of the<br />

microcalcifications. Further, the scattering signal is used to decouple<br />

the thickness parameter. We demonstrate that type I and type II<br />

microcalcifications give opposite absorption and scattering signals<br />

and in addition the small-angle scattering signal helps to determine<br />

the type of microcalcification.<br />

Conclusions: The potential clinical significance of phase-contrast<br />

enhanced mammography has been evaluated by our team. This<br />

technique yields improved diagnostic capabilities when compared<br />

with conventional mammography, can provide superior contrast,<br />

inaccessible and complementary information, and potentially<br />

also reduce dose deposition. The non-invasive classification of<br />

microcalcifications is an important step toward early diagnosis and<br />

differentiation of breast lesions.<br />

P4-03-07<br />

Angiogenic effect of bevacizumab and paclitaxel in metastatic<br />

breast cancer: evaluation by contrast-enhanced ultrasonography<br />

using Sonazoid®<br />

Ito T, Mizuno H, Iiboshi Y, Yamamura N, Fujii H, Hitora T, Fujii R,<br />

Ohashi T, Nakagawa T, Izukura M. Rinku General Medical Center,<br />

Izumisano, Osaka, Japan<br />

Agents targeting vascular endothelial growth factor (VEGF) have<br />

been validated as breast cancer therapeutics, yet efficacy can differ<br />

between tumor types and individual patients. Rapid noninvasive<br />

determination of response could provide significant benefits. We<br />

tested if response to the VEGF antibody bevacizumab and chemo<br />

drug paclitaxel could be detected using contrast-enhanced ultrasound<br />

imaging (CEUS).<br />

The objective of this study was to evaluate contrast-enhanced<br />

ultrasound (CEUS) with contrast agent Sonazoid® injection as a<br />

predictor of tumor response.<br />

Patients and methods: Ten patients with advanced and metastatic breast<br />

carcinomas treated were evaluated. Examinations were performed at<br />

baseline, after 2 and 4 weeks and every one month on antiangiogenic<br />

drug bevacizumab and chemo drug paclitaxel in tumor targets that<br />

were accessible to CEUS. Histological findings were obtained in all<br />

cases by ultrasound-guided 14 gauge core needle biopsy or 10 gauge<br />

vacuum-assisted biopsy. Each examination included a bolus injection<br />

of Sonazoid® and 40-60 seconds of raw linear data with an Aplio<br />

(Toshiba), Logiq (GE) and Ascendus (Hitachi-ALOKA) with a 8-12<br />

MHz linear transducer. A low reduced mechanical index (MI) of less<br />

than 0.24 was chosen to avoid early gas bubble destruction.<br />

The ethic committee of our institution approved this study. An<br />

informed consent was signed by each patient for the ultrasound<br />

examination and before administering for the contrast medium.<br />

Results: A total of 60 examinations were performed. Various<br />

intratumoral perfusion parameters changed over time, and the signal<br />

intensity was significantly impaired in the treated tumors before the<br />

treatment efficacy on tumor size could be observed.<br />

Conclusion: CEUS has advantages for real-time visualization and is a<br />

new noninvasive imaging technique which might be an effective tool<br />

for evaluating antiangiogenic drugs and chemo drugs in breast cancer.<br />

The use of a noninvasive ultrasound approach may allow for earlier<br />

and more effective determination of efficacy of antiangiogenic<br />

therapy.<br />

P4-03-08<br />

A new real-time image fusion technique, a coordinated sonography<br />

and MRI using magnetic position tracking system, improves the<br />

sonographic identification of enhancing lesions in breast MRI<br />

Nakano S, Fujii K, Yoshida M, Kousaka J, Mouri Y, Fukutomi T,<br />

Ishiguchi T. Aichi Medical University, Nakakute, Aichi, Japan<br />

<strong>Breast</strong> magnetic resonance imaging (MRI) is a method that can<br />

detect clinically and mammographically occult cancer foci. For<br />

lesions suspected to be malignant on the basis of breast MRI,<br />

targeted sonography can help determine whether the lesion is<br />

amenable to tissue biopsy using sonographic guidance. However,<br />

the reproducibility of sonography as well as interexaminer reliability<br />

in targeted sonography is still a clinical issue. Recently, we have<br />

developed real-time virtual sonography (RVS) that can coordinate a<br />

sonographic image and a MRI multiplanar reconstruction (MRI-MPR)<br />

of the same section in real time. Although RVS has been reported to<br />

be useful for breast cancers, only a few studies have investigated the<br />

size and positioning error between a real-time sonographic image<br />

and a MRI-MPR image. The purpose of this study was to evaluate<br />

the accuracy of RVS to sonographically identify enhancing lesions<br />

by breast MRI. Between December 2008 and May 2009, RVS was<br />

performed in 51 consecutive patients with 63 enhancing lesions.<br />

MRI was performed with the patients in the supine position using a<br />

1.5-T imager with a body surface coil to achieve the same position<br />

as with sonography. To assess the accuracy of the RVS, the following<br />

three issues were analyzed: (i) The sonographic detection rate of<br />

enhancing lesions, (ii) the comparison of the tumor size measured by<br />

sonography and the MRI-MPR and (iii) the positioning errors as the<br />

distance from the actual sonographic position to the expected MRI<br />

position in 3-D. Among the 63 enhancing lesions, 42 (67%) lesions<br />

were identified by conventional B-mode, whereas the remaining 21<br />

(33%) initial conventional B-mode occult lesions were identified<br />

by RVS alone. The sonographic size of the lesions detected by<br />

RVS alone was significantly smaller than that of lesions detected<br />

by conventional B-mode (p


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 4<br />

value (PPV), and negative predictive value (NPV) of each imaging<br />

modality were calculated using pathologic complete response (pCR)<br />

as the outcome of interest.<br />

RESULTS:<br />

Both MRI and ultrasonography displayed higher sensitivity (77%<br />

and 79%, respectively) than specificity (38% and 25%) in predicting<br />

primary tumor response, while PET-CT was associated with higher<br />

specificity (100%) than sensitivity (68%). All three imaging<br />

modalities exhibited higher PPVs (84%, 88%, and 100%) than NPVs<br />

(31%, 14%, 40%) in relation to primary tumor response. Both MRI<br />

and PET-CT were less sensitive for predicting the response of nodal<br />

disease than of primary tumor (p < 0.001).<br />

CONCLUSIONS:<br />

Out of the three radiologic tools, examined in the present study, no<br />

single test was clearly superior in evaluating the tumor response of<br />

either the primary tumor or nodal metastases. Future investigations<br />

are warranted to identify the tumor and treatment factors which may<br />

influence the specificity, sensitivity and predictive values of each test.<br />

P4-03-10<br />

Sonographic-pathological correlation in contrast-enhanced<br />

ultrasonography in the diagnosis of breast cancers.<br />

Kato K, Miyamoto Y, Kamio M, Nogi H, Imawari Y, Mimoto R, Toriumi<br />

Y, Nakata N, Takeyama H, Uchida K. The Jikei University School of<br />

Medicine, Tokyo, Japan<br />

Background: As contrast-enhanced ultrasonography (CEUS) in<br />

the diagnosis of breast masses has been performed less frequently<br />

than other imaging modalities, it’s efficacy evaluation has not been<br />

established. CEUS with Sonazoid, which is microbubble-based<br />

contrast agent, allows visualization of the mass bloodstream, therefore<br />

the classification of enhancement patterns in CEUS could increase<br />

diagnostic yield for breast masses. To achieve this, a clear standard<br />

for the CEUS evaluation is needed. The purpose of this study was to<br />

identify CEUS findings for malignancies and clarify sonographicpathological<br />

correlation of breast cancers.<br />

Material and Method: Present study included 20 patients with<br />

invasive ductal cancer who underwent operation for primary therapy<br />

without any chemotherapy or hormonal therapy. We contrasted CEUS<br />

features with Sonazoid with surgical specimens retrospectively. As<br />

malignant findings, the presence of focal perfusion defects inside<br />

masses and enhancement of outside masses were chosen.<br />

Result: All of the 20 cases displayed focal perfusion defects. 9<br />

cases of those could be successfully assessed in the same sections<br />

histopathologically and ultrasonographically. Histopathological<br />

findings showed those defects to be fibrotic foci inside masses in<br />

7cases (77.7%). While, 6 cases of all (30%) showed stains outside<br />

tumor, and only one case was found out to be fibrotic growth with<br />

cancer cells.<br />

Conclusion: focal perfusion defects of breast masses in CEUS<br />

were considered to be specific for malignancy, and they seemed to<br />

be fibrotic foci histopathologically. Desmoplastic reaction has been<br />

observed in epithelial solid tumor biology, and if those defects reflect<br />

the desmoplasia defects could be the proof of malignancy.<br />

P4-03-11<br />

SUVmax of FDG-PET/CT is Associated with Chemotherapy<br />

Response Assay Test Results and Prognostic Factors in <strong>Breast</strong><br />

<strong>Cancer</strong> Patients<br />

Lee A, Chang J, Bang B, Lee J, Han D, Min S, Yun C, Kim BS, Lim<br />

W, Paik N, Moon B-I. Ewha Womans University Hospital, Seoul,<br />

Republic of Korea<br />

Backgrounds: 18 F-fluorodeoxyglucose positron emission tomography<br />

with computed tomography ( 18 F-FDG PET/CT) is widely used for<br />

cancer evaluation and there are several studies which suggested<br />

maximum standard uptake value (SUVmax) may reflect the cancer<br />

patients’ prognosis. Adenosine triphosphate-based chemotherapy<br />

response assay (ATP-CRA) is commonly used as a chemosensitivity<br />

assay modality for the treatment of various cancers in clinical settings<br />

and there have been several studies on its usefulness in breast cancer.<br />

We previously reported chemosensitivity (ATP-CRA) results might<br />

reflect patients’ prognosis. So we supposed that SUVmax could<br />

reflect chemosenitivity (ATP-CRA) results of patients, so performed<br />

this study to analyze the relationship between PET/CT SUVmax and<br />

chemosenstivity (ATP-CRA) results of these drugs. Furthermore, we<br />

evaluated prognostic factors for breast cancer according to SUVmax.<br />

Materials and Methods: 102 breast cancer patients, who underwent<br />

PET/CT and chemosensitive (ATP-CRA) test between December<br />

2010 and April 2012, were enrolled in this study. We analyzed,<br />

retrospectively, the correlation between SUVmax of PET/CT and<br />

chemosenitivity (ATP-CRA) results of doxorubicin/paclitaxel.<br />

SUVmax according to prognostic factors were also assessed.<br />

Results: Chemosensitivity (ATP-CRA) results of doxorubicin and<br />

paclitaxel have significant positive correlation with SUVmax. Their<br />

correlation coefficient were 0.236 (p=0.020) and 0.216 (p=0.030),<br />

respectively. Patients with larger tumor size, higher histologic and<br />

nuclear grade, estrogen receptor negative, progesterone receptor<br />

negative, HER2 positive, and Ki67 positive (≥14%) have higher<br />

mean SUVmax values (p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

make pathological examination with tumor tissues removed by needle<br />

biopsy. Then we make a retrospective analysis and evaluation of<br />

patients scanned completely by qualitative and semi-quantitative<br />

methods. All patients have signed the informed consent form before<br />

the test, together with the recognition of the Ethics Committee.<br />

Results: From December 2011 to May 2012, a total of 245 patients<br />

were scanned and operated with clear paraffin pathological diagnosis<br />

by intravenous injection of 99 mTc -3PRGD 2 . Among them, a total of<br />

106 patients (167 developing lesions) were positive with radionuclide<br />

imaging results, 139 patients were negative. Diseases of 106 patients<br />

are divided into four series: breast cancer, inflammation mastitis,<br />

phyllodes tumor and partly with fibroadenoma. They have in common<br />

that there is a clear flow signal in and around lesions by color Doppler<br />

ultrasound.<br />

Discussion: After study on 245 patients, we created a molecular<br />

imaging tool to show expression of integration αvβ3 in order to<br />

assess tumor angiogenesis.Integrin αvβ3 plays an important role in<br />

physiological and pathological processes of malignant tumor, such<br />

as inflammation, tumor angiogenesis, invasion and metastasis etc.<br />

Therefore, its high expression is easy to find in cancer, mastitis, most<br />

of the phyllodes tumor and part of fiber adenoma with peripheral<br />

lesions. For patients with clear imaging, we make comprehensive<br />

analysis based on their ultrasound, mammography, MRI or CT results,<br />

which can generally contributes to the early diagnosis of disease in<br />

clinical. The detection of sentinel lymph node should be avoided<br />

for the patients with axillary lymph node imaging, who are clearly<br />

diagnosed as breast carcinoma with axillary lymph node metastases.<br />

These peptides are not only tumor receptor-targeted imaging agents<br />

with potential clinical value, but also laid a solid foundation for<br />

further development of receptor-targeted radionuclide therapy to solid<br />

tumors. The method marked with high expression of integrin αvβ3 in<br />

the tumor vascular endothelial cells by 99 mTc-3PRGD 2 and SPECT/<br />

CT imaging mode has a broad prospect in the early diagnosis of<br />

tumor, lesion detection and efficacy evaluation. In the future, we may<br />

also find the new imaging agent with more accuracy and specificity.<br />

We believe that there will be more and more breast cancer patients<br />

benefit from the technology development of ECT/CT examination.<br />

P4-04-01<br />

Combination of intratumoral CpG with systemic anti-OX40 and<br />

anti-CTLA4 mAbs eradicates established triple negative breast<br />

tumors in mice<br />

Li J, Tandon V, Levy R, Esserman L, Campbell M. University of<br />

California, <strong>San</strong> Francisco, CA; Stanford University, Stanford, CA<br />

Background: Evidence is mounting across diverse datasets and<br />

scientific techniques implicating immune dysregulation in poor<br />

outcomes of high grade and/or triple negative breast cancers.<br />

A multimodal strategy to stimulate the immune system could<br />

be an effective therapeutic approach for breast cancer. CpG<br />

oligodeoxynucleotides are single-stranded DNA that stimulate<br />

dendritic cells and B cells by binding to Toll-like receptor 9 (TLR9).<br />

OX40 (CD134) is predominantly expressed on activated T cells and<br />

the binding of OX40 with an agonistic antibody (anti-OX40) leads<br />

to the proliferation and survival of T cells. Agonistic anti-OX40<br />

antibodies can also reverse the suppressive effects of Treg cells. A<br />

third avenue is to target Cytotoxic T lymphocyte associated antigen<br />

(CTLA4) , which plays a negative regulatory role in T cell activation<br />

and is also constitutively expressed on Treg cells. Blocking CTLA4<br />

with an antagonistic antibody (anti-CTLA4) has been shown to<br />

enhance anti-tumor responses. In this study, we tested the combination<br />

of intratumoral (IT) CpG with systemic anti-OX40 plus anti-CTLA4<br />

in a mouse model of triple negative breast cancer.<br />

Methods: 4T1, an aggressive, triple-negative mammary carcinoma,<br />

was implanted orthotopically into syngeneic BALB/C mice.<br />

Treatment was initiated when tumors were ∼0.5 cm in the largest<br />

dimension. Mice were divided into 8 groups. Various combinations<br />

of PBS control, CpG, taxol, and anti-OX40+anti-CTLA4 mAbs<br />

were tested (Tables 1 and 2). CpG, 100 µg, was injected IT for 5<br />

consecutive days (days 1-5); Taxol, 16 mg/kg, was administered IP on<br />

day 1; anti-OX40 (400 µg) and anti-CTLA4 (100 µg) were given IP<br />

on days 1 and 5. Mice were sacrificed once tumors became ulcerated<br />

or reached a diameter >2 cm.<br />

Results: As shown in Table 1, CpG alone, taxol alone, or the anti-<br />

OX40 + anti-CTLA4 combination each delayed the growth of<br />

established 4T1 tumors when compared to the untreated control group.<br />

However, the majority of the mice (except one in CpG treated group)<br />

died from tumor progression. The combination of CpG + anti-OX40<br />

+ anti-CTLA4 had the best anti-tumor effects, with 4 out of 5 mice<br />

having demonstrated complete regression of their tumors (Table 2).<br />

The addition of taxol to the combination of CpG + anti-OX40 + anti-<br />

CTLA4 did not improve the anti-tumor effect, and while there was<br />

a substantial delay in growth, only one animal achieved a complete<br />

regression.<br />

Table 1: Single Agent<br />

Groups Treatments<br />

Average tumor size at the time of<br />

# of tumor free mice<br />

completion(mm3)<br />

Group 1 Control 1388 0/5<br />

Group 2 Taxol 1112 0/5<br />

Group 3 CpG 305 1/5<br />

Group 4 anti-OX40+anti-CTLA4 739 0/5<br />

Table 2: Combined Treatment<br />

Groups Treatments<br />

Average tumor size at the time of # of tumor<br />

completion(mm3)<br />

free mice<br />

Group 5 Taxol+CpG 450 0/5<br />

Group 6 Taxol+ anti-OX40+antiCTLA4 341 2/5<br />

Group 7 CpG+ anti-OX40+antiCTLA4 119 4/5<br />

Group 8 Taxol+CpG+ anti-OX40+antiCTLA4 327 1/5<br />

Conclusion: Our study demonstrated that the combination of IT CpG<br />

with systemic anti-OX40 + anti-CTLA4 mAbs can cure mice with<br />

established triple negative breast tumors, supporting the possibilities<br />

of applying immune modulation approach in human triple negative<br />

tumors. Studies are on-going to further dissect the immunological<br />

mechanisms involved.<br />

P4-04-02<br />

Tumor-initiated peripheral myeloid cell expansion is reversed<br />

by radiation therapy<br />

Gough MJ, Savage T, Bahjat KS, Redmond W, Bambina S, Kasiewicz<br />

M, Cottam B, Newell P, Crittenden MR. Earle A. Chiles Research<br />

Institute, Providence <strong>Cancer</strong> Center, Portland, OR; Providence<br />

<strong>Cancer</strong> Center, Portland, OR; The Oregon Clinic, Portland, OR<br />

Myeloid lineage cells, particularly monocytes and macrophages,<br />

have an important role in the development and progression of cancer.<br />

Within established tumors, macrophages are critical for angiogenesis,<br />

invasion, metastasis, immunosuppression and response to therapy.<br />

In addition, both transplantable and transgenic mouse models of<br />

mammary cancer are associated with myeloid expansions detectible<br />

in the tumor, spleen and peripheral blood, and these myeloid cells are<br />

able to suppress T cell activation in vitro. Reducing the tumor burden<br />

through chemotherapy and resection has been shown to control the<br />

myeloid expansion. However, chemotherapies that reduce tumor<br />

burden also have direct effects on myeloid cells, and surgical trauma<br />

has been shown to mobilize myeloid cells. Radiation therapy is the<br />

third major option to treat cancer, but the consequence of radiation<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

368s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 4<br />

therapy to systemic myeloid populations has not been described. We<br />

applied tumor-specific radiation therapy to test the hypothesis that the<br />

frequency of myeloid lineage cells correlates with the primary tumor<br />

burden. We demonstrate in the MMTV-PyMT model of mammary<br />

cancer that myeloid expansion in the blood closely correlates with<br />

myeloid involvement in the tumor and with tumor progression. We<br />

demonstrate that radiation therapy of 4T1 mammary tumors leads<br />

to a decline in myeloid cell numbers in the blood and a decrease in<br />

spleen size. The frequency of myeloid cells does not decline to the<br />

level seen in tumor-free mice: we demonstrate that metastatic disease<br />

can prevent myeloid cell numbers from returning to baseline, and that<br />

tumor recurrence from residual disease correlates with re-expansion<br />

of myeloid lineage cells. Despite these changes in the frequency<br />

of myeloid lineage cells, T cell numbers in the blood and spleen<br />

remains constant. However, radiation therapy improves the myeloid<br />

cell:CD8+ T cell ratio and results in increased proliferation of T cells<br />

in the spleen. Finally, we demonstrate that T cell responses to foreign<br />

antigens are not altered by tumor burden or myeloid cell expansion,<br />

while T cell responses to tumor-associated antigens are increased<br />

after radiation therapy of the tumor. These data demonstrate that<br />

myeloid cell numbers are directly linked to primary tumor burden,<br />

that this population contracts following radiation therapy, and that<br />

radiation therapy may open a therapeutic window for immunotherapy<br />

of residual disease.<br />

P4-04-03<br />

Monocytic immature myeloid cells permit tumor immune evasion<br />

during postpartum involution<br />

Schedin P, Martinson H, Callihan E, Jindal S, Borges V. University<br />

of Colorado, Aurora, CO<br />

Young women with breast cancer diagnosed in the postpartum period,<br />

defined as up to 5 years from last child delivery, have a poorer<br />

prognosis compared to women who have never been pregnant, and<br />

represent ∼30% of all young women’s breast cancer cases. Our lab<br />

has proposed that the poor prognosis associated with postpartum<br />

breast cancer is due to gland remodeling following pregnancy,<br />

known as postpartum involution. Using rodent models and human<br />

breast tissue, it has been demonstrated recently that postpartum<br />

gland involution utilizes wound healing programs to remodel the<br />

gland to a non-secretory state. For example, macrophages with tumor<br />

promotional characteristics have been identified as essential mediators<br />

of epithelial cell death during postpartum involution. We hypothesize<br />

that the immune cells within the normal involuting gland set up an<br />

immunosuppressive environment to protect the host from negative<br />

sequel resulting from potential self-antigen exposure during epithelial<br />

cell death. A consequence would be the establishment of an immune<br />

microenvironment that favors the growth and metastasis of breast<br />

cancer cells. To characterize the role of immune cells during normal<br />

postpartum involution as well as in the promotion of mammary cancer,<br />

we developed an immunocompetent mouse model of postpartum<br />

breast cancer using the Balb/c mouse.<br />

By flow cytometry, we found that dendritic cells, GR1+ monocytic<br />

immature myeloid cells (IMCs), and T cells increase exponentially<br />

in the mammary gland during involution. Functional assays using<br />

myeloid cells from the mammary gland indicate the IMCs isolated<br />

from involuting glands are capable of suppressing T cell activation<br />

ex vivo, consistent with immune suppression in vivo. To begin to<br />

investigate whether IMCs are able to promote tumor progression in<br />

the postpartum breast cancer setting, D2A1 mammary carcinoma<br />

were injected into mammary fat pads of Balb/c mice. D2A1 cells<br />

injected into hosts with actively involuting mammary glands had a<br />

significant growth advantage over tumor cells injected into nulliparous<br />

host glands, demonstrating that mammary gland involution promotes<br />

the growth of mammary carcinoma cells in this immune competent<br />

model. T cells identified by flow cytometry are significantly lower<br />

in number systemically and within the tumors of the involutiongroup<br />

compared to the nulliparous-group, indicating systemic<br />

immunosuppression specific to the involution-group. To demonstrate<br />

immunosuppressive function, IMCs were isolated from tumors and<br />

incubated with T cells ex vivo. IMCs from involution tumors were<br />

significantly more suppressive then IMCs from nulliparous mice.<br />

These data indicate IMCs play an immunomodulatory role in the<br />

mammary gland during involution, which allows tumor cells to<br />

undergo immune evasion in the postpartum period. We are currently<br />

investigating how GR1+ IMCs suppress T cell activation and hope<br />

to determine if reprograming these cells could prevent the promotion<br />

of breast cancer during involution. (DOD-BC10904 and BC100910)<br />

P4-04-04<br />

Immunohistochemical analysis of <strong>Cancer</strong> Testis antigens and<br />

Topoisomerase 2-alpha expression in triple negative breast<br />

carcinomas: a retrospective study<br />

Juretic A, Mrklic I, Spagnoli GC, Pogorelic Z, Tomic S. Zagreb<br />

University Hospital Centre, Zagreb, Croatia; Split University Hospital<br />

Centre, Split, Croatia; University of Basel, Switzerland<br />

Aim. The aim of this study is to assess the expression of cancer testis<br />

(C/T) antigens in the TNBC group, expression of topoisomerase<br />

2-alpha (TOPO2A), basal-like (BL) immunophenotype defined by<br />

basal markers (CK5/6, CK14, and EGFR), BL morphology and<br />

conventional clinicopathologic factors as well as to demonstrate their<br />

prognostic relevance with respect to this group of tumours.<br />

Methods. The study includes 83 patients who underwent surgery<br />

between January 2003 and December 2009. Slides were stained<br />

immunohistochemically with CK5/6, CK14, EGFR, Ki-67, TOPO2A,<br />

MAGE-A1, MAGE-A10, NY-ESO and multi-MAGE. Survival-time<br />

and multivariate survival analyses were performed for the purpose<br />

of identifying prognostic markers for tumours with more aggressive<br />

behaviour.<br />

Results. Of the 83 triple negative tumours, 55 (66.3%) have BL<br />

immunophenotype and 40 (48.2%) BL morphology. MAGE-A1<br />

show the most frequent expression (69.0%), followed by multi-<br />

MAGE (58.0%), NY-ESO (27.1%) and MAGE-A10 (13, 2%).<br />

MAGE-A10 expression is significantly correlated with tumour size<br />

(p=0.026). The expressions of MAGE-A1, MAGE-A10 and multi-<br />

MAGE are significantly correlated with clinical stage (p=0.024,<br />

p=0.041, p=0.031, respectively). A significant correlation has been<br />

found between the expressions of MAGE-A10 and NY-ESO and the<br />

expression of TOPO2A (p=0.005, p=0.013). There is no significant<br />

correlation between the expression of C/T antigens and disease-free<br />

survival (DFS) or overall survival (OS).<br />

Conclusions. High expression levels of MAGE and NY-ESO<br />

observed in TNBC are of potential clinical relevance, especially in<br />

the adjuvant setting of treatment, and may be of therapeutic value<br />

as a vaccine-based treatment in the TN group of breast tumours for<br />

which therapeutic options are limited.<br />

Key words: triple negative breast cancer, basal-like breast cancer,<br />

cancer-testis antigen, topoisomerase 2-alpha, prognosis, survival<br />

analysis.<br />

www.aacrjournals.org 369s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P4-04-05<br />

Listeria monocytogenes-based bivalent Lm-LLO immunotherapy<br />

for the treatment of HER2/neu positive and triple negative breast<br />

cancer and its impact on immunosuppression<br />

Wallecha A, Ramos K, Malinina I, Singh R. Advaxis Inc, Princeton, NJ<br />

Overexpression of tumor associated antigens (TAA) such as HER2/<br />

neu and high molecular weight melanoma associated antigen<br />

(HMW-MAA) are associated with aggressive high-grade tumors<br />

leading to disease progression and reduced survival. HMW-MAA has<br />

been reported as a TAA in triple negative breast tumors and is also<br />

expressed at high levels by activated pericytes and pericytes in tumor<br />

angiogenic vasculature that are associated with neovascularization<br />

in vivo. Previously published reports have shown that bioengineered<br />

Listeria monocytogenes (Lm) secreting either a chimeric HER2/neu<br />

antigen (cHER2), or HMW-MAA fragment, fused to a truncated<br />

highly immunogenic fragment of the adjuvant protein listeriolysin<br />

O (LLO) were found to impact the growth of both transplantable and<br />

transgenic mouse breast tumor models. Therefore, we hypothesized<br />

that an Lm-LLO immunotherapy capable of delivering two different<br />

antigens would likely have a synergistic effect of decreasing tumor<br />

growth by targeting two independent mechanisms that support tumor<br />

growth; 1) tumor angiogenesis, and 2) tumor cell surface marker, thus<br />

improving the therapeutic efficacy of the agent. Here we report the<br />

construction of a bivalent recombinant Lm-LLO immunotherapy that<br />

expresses and secretes both the cHER2 and HMW-MAA antigens<br />

concomitantly as fusion proteins. Initial characterization of the<br />

bivalent immunotherapy indicates that both the antigens cHER2<br />

and HMW-MAA are stably expressed and secreted after two in<br />

vivo mouse passages. Preliminary studies support the hypothesis<br />

that bivalent immunotherapy causes efficient reduction of tumor<br />

growth in both HER2 expressing and triple negative breast tumors.<br />

Further, mechanistic studies demonstrate that treatment with Lm-LLO<br />

immunotherapy resulted in a decreased proportion of regulatory T<br />

cells (Tregs) as well as Myeloid Derived Suppressor Cells (MDSC)<br />

particularly in the tumor microenvironment and not in spleen, tumor<br />

draining lymph nodes, or peripheral blood. Interestingly, both residual<br />

Tregs and MDSCs isolated from the tumor microenvironment were<br />

found to have a decreased ability to suppress the division of T cells<br />

after Lm-LLO immunotherapy, a finding which warrants further<br />

investigation. We will present data on the efficacy of bivalent Lm-LLO<br />

immunotherapy and the mechanisms that are likely responsible for<br />

the observed tumor regression. Currently Lm-LLO immunotherapies<br />

are being evaluated in Phase 2 clinical trials for HPV-associated<br />

dysplasia and malignancies such as CIN2/3, cervix cancer and head<br />

& neck cancer.<br />

P4-04-06<br />

Young women’s breast cancer is characterized by increased<br />

immune suppression through circulating myeloid derived<br />

supressor cells<br />

Borges VF, Ramirez O, Borakove M, Manthey E, Diamond JR, Elias<br />

AD, Finlayson C, Kounalakis N, Jordan K. University of Colorado<br />

Denver, Aurora, CO; University of Colorado <strong>Cancer</strong> Center, Aurora,<br />

CO<br />

Background: Women age 20-40 have a higher ten-year risk for<br />

developing breast cancer than the five other leading cancers in this<br />

gender and age group combined. Women currently in their 30s have<br />

a 1/203 chance of developing breast cancer over the next ten years.<br />

Moreover, cancers diagnosed in younger women have a signficantly<br />

higher risk of death for reasons that are not fully identified. Myeloid<br />

derived suppressor cells (MDSC) have a role in suppressing antitumor<br />

immunity through Jak/Stat pathway activation by generation<br />

of reactive oxygen species (ROS) and arginase-1, and correlate<br />

preclinically with cancer progression and in human canceres with<br />

poorer prognositic features and outcomes. Our Young Women’s<br />

<strong>Breast</strong> <strong>Cancer</strong> Translational Program seeks to identify immunologic<br />

differences potentially contributing to the poorer prognosis and<br />

potential targets for development of immunomodulatory treatment.<br />

Hypothesis: We hypothesized that newly diagnosed, treatmentnaive<br />

young breast cancer cases would have higher percentage of<br />

circulating MDSC with T cell suppressive function than similar<br />

age women without breast cancer. Methods: We conducted IRB<br />

approved, prospective translational studies of women age 40 and<br />

under both affected and unaffected by breast cancer. Exclusion criteria<br />

included pregnancy, known autoimmunity or immunosuppressive<br />

medications, or other cancers. MDSC were isolated from peripheral<br />

blood and phenotyped through standard protocols. T cell suppressive<br />

abilites were tested in coculture assays for expression of activation<br />

markers CD25 and CD69, and production of gamma-interferon.<br />

Identification of mechanism of suppression was assayed through<br />

secretion of arginase-1and generation of ROS. Results: No difference<br />

in percentage of MDSC was identified between young women with<br />

breast cancer (n=61) and unaffected subjects (n=18). However, a<br />

statistically significant increase in the MDSC ability to suppress<br />

T cell activation was identified in the breast cancer cohort with<br />

diminished CD25 and CD69 expression and diminished production<br />

of gamma-interferon. MDSC from young breast cancer subjects<br />

secreted significantly more arginase-1 from MDSC. No significant<br />

difference in generation of ROS was found between the two cohorts.<br />

Conclusions: The presence of equal rather than higher numbers of<br />

circulating MDSCs between the young breast cancer versus unaffected<br />

subjects was unexpected in comparison to prior publications on<br />

MDSC in cancer. Our results may differ due to our larger sample<br />

size or the alignment by gender and age the cohorts being more<br />

representative. Also, these are the first data on MDSC in a young<br />

female population. Despite equal numbers of MDSCs, the functional<br />

analyses demonstrate MDSC isolated from breast cancer have<br />

enhanced T cell suppression capabilities compared with normal<br />

controls. These data provacatively suggest a functional difference<br />

inherent to MDSC in the young breast cancer-bearing host. Moreover,<br />

they indicate that normal young women have realtively high levels<br />

of MDSC that are available for cancer to ursurp and induce immune<br />

suppression. Identification of secretion of arginine-1 as a potential<br />

mechanism of immune supporession in young women’s breast cancer<br />

warrants further investigation.<br />

P4-04-07<br />

Tartrate-resistant Acid Phosphatase (TRAcP)-Expressed Tumor-<br />

Associated Macrophages Promote <strong>Breast</strong> <strong>Cancer</strong> Progression<br />

Dai M-S, Wu C-C, Chang P-Y, Ho C-L, Hsieh Y-F, Lo K-Y, Kao W-Y,<br />

Chao T-Y, Yu J-C. Tri-Service General Hospital, Taipei, Taiwan;<br />

Taipei Medical University, Taipei, Taiwan<br />

Purpose<br />

Tumor-associated macrophages (TAMs) can promote or dampen<br />

breast cancer progression, depending on phenotypic changes within<br />

the cancer microenvironment. Two contrasting activation states<br />

were described in the literature: the M1 anti-tumoral and M2 protumoral<br />

subtype of TAMs. In clinical observation, we analyzed the<br />

TRAcP expression in breast cancer TAM and correlated to patient’s<br />

chemotherapy responses and survival. In animal models, we aim<br />

to investigate the tumor-promoting or inhibiting properties among<br />

tartrate-resistant acid phosphatase (TRAcP) expressed TAMs.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

370s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 4<br />

Methods<br />

TRAcP expression, T lymphocytes, and other macrophage relevant<br />

markers were determined by immunohistochemical staining in the<br />

human breast cancer tissue from the patients before neoadjuvant<br />

chemotherapy. The expression of TRAcP within the TAMs were<br />

calculated and correlated to the patient’s chemotherapy responsiveness<br />

and outcome. The polarized M1/M2 macrophages and breast cancer<br />

hematogenous metastasis were created in the animal model. Activated<br />

macrophages from TRAcP+/+ or TRAcP-/- mice were injected with<br />

JC (Balb/c breast cancer) cells in the metastatic model.<br />

Results<br />

The presence of Foxp3+ regulatory T cell (Treg) correlates to<br />

the tumor proliferation (Ki-67 and Her2 expression) and higher<br />

Foxp3+ Treg prevalence and tumor Ki-67 expression inferred<br />

poorer chemotherapy response. TRAcP staining within the CD68+<br />

macrophages were observed with better chemotherapy response.<br />

In animal model, M2 polarized macrophages have higher TRAcP<br />

expression, promote breast cancer metastasis, and attract Treg<br />

infiltrating in tumor, consistent with the clinical observation.<br />

Conclusion<br />

TRAcP-expressing TAMs, representing M2-polarized macrophage,<br />

demonstrat local tumor-promoting and immune-suppressive effects,<br />

and chemotherapy resistance. TRAcP expression within TAM can be<br />

a potential prognostic marker for breast cancer.<br />

P4-04-08<br />

A Potential non-viral vector to transfect dendritic cell and thereby<br />

MHC-Class I antigen presentation might be a potential use in<br />

carcinoma<br />

Shaheen S, Akita H, Souichirou I, Miura N, Harashima H. Hokkaido<br />

University, Sapporo, Hokkaido, Japan<br />

To date almost no remarkable potential non-viral vector was<br />

developed to transfect promisingly primary dendritic cell like Bone<br />

Marrow Derived Dendritic Cell (BMDC) so that it might generate<br />

antigen presentation while epitopic-plasmid was delivered. Here we<br />

have introduced a potential non-viral vector named Stearyl-KALA<br />

MEND (100-130 nm, zeta potential 35-40 mV), which expressed<br />

to date the highest transgene expression while firefly luciferase was<br />

introduced (sub-cloned), and thereby promising antigen presentation<br />

in vitro while OVA plasmid containing MHC Class-I restricted<br />

SIINFEKL epitope was introduced and co-cultured with B3Z T-cell<br />

hybridoma. On the basis of these results, we used this vector to<br />

transfect BMDC and harvested the DC to chase the corresponding<br />

EG7-OVA induced tumor ex vivo. Later we further immunized mice<br />

directly with STR-KALA MEND containing OVA plasmid and<br />

challenged against the tumor (EG7-OVA induced). In this in vivo<br />

study we found also a significant antitumor activity. To evaluate<br />

the promptness of our vector we further sub-cloned Mart-1 gene, in<br />

our sub-cloned plasmid vector and immunized the mice as before.<br />

Thereafter inserting B16F10 melanoma cells to the immunized<br />

mice we found also a significant antitumor activity after 24 days of<br />

inoculation. Thus the vector, STR-KALA MEND might be a future<br />

potential use as DNA vaccine in anti-tumor methodology.<br />

P4-05-01<br />

Oncolytic Herpes Simplex Virus Vector G47∆ Effectively Targets<br />

<strong>Breast</strong> <strong>Cancer</strong> Stem Cells<br />

Liu R, Zeng W, Hu P, Wu J, Li J, Wang J, Lei L. The Third Affiliated<br />

Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China;<br />

The Sichuan Province <strong>Cancer</strong> Hospital, Chengdu, Sichuan, China<br />

<strong>Breast</strong> tumors can be classified according to their gene-expression<br />

profiles into different molecular subtypes with distinct biological<br />

and clinical features. Moreover, different subtypes show distinct<br />

responses to different therapeutic regimens, thereby resulting in<br />

markedly different outcomes. Recently, accumulating evidence has<br />

suggested that breast cancer originates from cancer stem cells (CSCs),<br />

which comprise a small percentage of the overall tumor but are highly<br />

tumorigenic and pluripotent with unlimited proliferation potential.<br />

Furthermore, cancer stem cells are highly resistant to conventional<br />

treatment, which potentially explains certain difficulties in treating<br />

cancer with current therapy options. The use of oncolytic viruses is<br />

a relatively new strategy in cancer therapy, oncolytic herpes simplex<br />

virus (oHSV) has been shown to be a safe and effective therapeutic<br />

approach for a variety of different cancers.<br />

In this study, different types of breast cancer cell lines were cultured<br />

in anchorage-independent conditions, the cells typically form<br />

mammospheres within 10 days of anchorage-independent culture,<br />

which showed properties of CSCs. The mammospheres are capable of<br />

being passaged indefinitely and single dissociated mammosphere cells<br />

were able to regenerate spheres. Moreover, compared to the parental<br />

cell lines, mammoshpere cells were enriched for CD44 + CD24 - cells<br />

and highly expression of the alternative breast cancer stem cell<br />

marker ALDH-1.<br />

The third generation oHSV vector G47∆ effectively killed different<br />

subtypes of breast cancer cells in vitro, with more than 98% of the<br />

tumor cells killed by day 5, even at multiplicities of infection(MOI)<br />

0.01. Moreover, G47∆ equally targeted non-cancer stem cells<br />

(NCSCs) and CSCs which showed resistance to paclitaxel. We also<br />

demonstrated that G47∆ effectively replicated and spread among<br />

CSCs. Moreover, G47∆ impaired the self-renewal ability of CSCs,<br />

as the viable cells unable to form secondary tumor spheres. We<br />

also showed that G47∆ was able to induce the regression of tumor<br />

xenografts in BALB/c nude mice and demonstrated the ability of<br />

G47∆ to synergize with paclitaxel for killing both NCSCs and CSCs.<br />

This is the first report that oHSV effectively targets different breast<br />

cancer NCSCs and CSCs, thereby suggesting that oHSV may be an<br />

effective treatment modality for patients with breast cancer.<br />

P4-06-01<br />

JAK2/STAT3 activity in inflammatory breast cancer supports<br />

the investigation of JAK2 therapeutic targeting<br />

Overmoyer BA, Almendro V, Shu S, Peluffo G, Park SY, Nakhlis F,<br />

Bellon JR, Yeh ED, Jacene HA, Hirshfield-Bartek J, Polyak K. Dana<br />

Farber <strong>Cancer</strong> Institute, Harvard Medical School, Boston, MA; Seoul<br />

National University, Seoul, Korea<br />

Background: Inflammatory breast cancer (IBC) is a virulent subtype<br />

of locally advanced breast cancer that accounts for 1-5% of all breast<br />

cancers diagnosed in the United States. Conventional therapy for<br />

IBC (preoperative chemotherapy, mastectomy, radiation therapy)<br />

results in a median 5 year overall survival of 50%, which supports<br />

the need for investigation into the molecular distinctiveness of<br />

IBC, with the ultimate goal of developing effective therapeutic<br />

agents that target these molecular changes. Some of the distinctive<br />

molecular characteristics of IBC include the presence of a substantial<br />

proportion of CD44+/CD24- breast cancer cells that express stem<br />

www.aacrjournals.org 371s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

cell-like characteristics, over-expression of E-cadherin, and extensive<br />

formation of tumor emboli. Examination of breast tumors and preclinical<br />

data suggests that the JAK2/STAT3 pathway is active in<br />

CD44+/CD24- breast cancer cells, and is stimulated by various<br />

mechanisms associated with IBC, such as high IL-6 levels. We<br />

investigated the role of JAK2/STAT3 activation in IBC as preclinical<br />

evidence to pursue JAK2 targeted therapy for IBC in a clinical setting.<br />

Methods: Using de-identified breast tissue specimens from both<br />

untreated and treated IBC patients obtained on a protocol approved by<br />

our institutional review board; we performed immunohistochemical<br />

(IHC) and multicolor immunofluorescence analysis to detect the<br />

presence of CD44+/CD24- cells and the fraction of activated<br />

phospho-STAT3 (pSTAT3). The number of cells positive for pSTAT3<br />

was assessed pre and post-treatment. We also analyzed the status of<br />

CD44+/CD24- cells and pSTAT3 in SUM149 and SUM190 IBC cell<br />

lines, and an IBC xenograft model (IDC31) derived from primary<br />

tumor obtained from an IBC patient. We tested the efficacy of multiple<br />

different JAK2 inhibitors to inhibit the growth and viability of cell<br />

cultures and xenografts.<br />

Results: All IBCs were highly enriched in CD44+/CD24- cells, with<br />

approximately 80% of all cancer cells within tumors displaying this<br />

phenotype. About 85% of IBC cases show activation of pSTAT3, and<br />

a significant fraction (40%) of CD44+/CD24- cells were pSTAT3<br />

positive. Interestingly, the fraction of pSTAT3+ cells was lower in the<br />

CD44+/CD24- cells in post treatment samples from triple negative<br />

but not from luminal tumors. JAK2 inhibition significantly decreased<br />

the proliferation of pSTAT3+ IBC cells lines in vitro and the growth<br />

of pSTAT3+ xenografts including the IBC model ICD31.<br />

Discussion: Specific molecular characteristics of IBC result in a<br />

unique and virulent disease course. Based upon a predominance<br />

of CD44+/CD24- breast cancer stem cells present in IBC, we<br />

investigated the JAK2/STAT3 pathway in IBC tissue specimens, cell<br />

lines and an IBC xenograft model. Our preclinical data demonstrates<br />

a highly active JAK2/STAT3 pathway in IBC which lends itself to<br />

exploration of the efficacy of a JAK2 inhibitor in the treatment of<br />

this disease. A phase I/II study is underway to evaluate combination<br />

ruxolitinib and chemotherapy for the primary treatment of triple<br />

negative IBC.<br />

P4-06-02<br />

Identification of novel G-protein coupled receptor targets in<br />

HER2-positive breast cancer.<br />

Yadav P, Bhat RR, Chayanam S, Christiny PI, Nanda S, Hu H,<br />

Creighton C, Osborne CK, Schiff R, Trivedi MV. University of<br />

Houston, TX; Lester & Sue Smith <strong>Breast</strong> Center, Dan Duncan <strong>Cancer</strong><br />

Center, Houston, TX; Baylor College of Medicine, Houston, TX<br />

Background: HER2 is a member of human epidermal growth factor<br />

receptor (HER) superfamily and HER2-overexpressing (HER2+)<br />

breast cancer (BC) is an aggressive tumor. Despite the clinical success<br />

of anti-HER2 therapies, de novo and acquired drug resistance occur<br />

in many patients. Identification of novel drug targets to overcome<br />

anti-HER2 therapy resistance is an unmet need. Since G-protein<br />

coupled receptors (GPCRs) are known to cross-talk with the HER<br />

superfamily, it is possible that some GPCRs may signal to modulate<br />

the HER2 pathway. GPCRs are considered excellent drug targets<br />

due to their plasma membrane localization, unique ligand-binding<br />

pocket, and availability of high throughput assays for drug screening.<br />

The expression and function of the majority of GPCRs are largely<br />

unknown in HER2+ BC. The goal of this study was to identify novel<br />

GPCR targets in HER2+ BC, in the context of anti-HER2 therapy<br />

resistance.<br />

Methods: We examined the differential GPCRs expression in BC stem<br />

cells, suggested to be involved in resistance, as well as in anti-HER2<br />

treatment-resistant BT474 cell line model of HER2+ BC. BC stem<br />

cells were identified as aldehyde dehydrogenase-positive (ALDH+)<br />

cells using the Aldefluor assay. Brightly fluorescent ALDH+ cells<br />

were separated from ALDH- cells using FACS Aria II cell sorter.<br />

Anti-HER2 resistant derivatives of BT474 cells were established by<br />

long-term exposure to increasing drug concentration of trastuzumab<br />

(T), lapatinib (L), or their combination (T+L). RNA was isolated and<br />

subjected to profiling using TaqMan real time RT-PCR GPCR 384-<br />

well microarray to quantify the expression of mRNA encoding 343<br />

GPCRs from 50 different subfamilies. Only overexpressed GPCRs<br />

were considered of interest as drug targets, and the overexpression of<br />

GPCRs was verified by RT-PCR and western blotting for the selected<br />

targets. Publically available TCGA dataset for mRNA expression<br />

was also interrogated to determine differential expression of selected<br />

GPCRs in HER2+ vs. other subtypes of BC.<br />

Results: Nine GPCRs [BAI3, EDNRA, GPR110, GPR116, GPR124,<br />

MTNR1A, EDG2, EMR2, GCGR] were upregulated in ALDH+<br />

compared to ALDH- BT474 cells. In addition, 11 GPCRs [CCBP2,<br />

CCR9, F2RL1, GALR2, GPR1, GPR24, GPR87, GPR110, GPR183,<br />

LGR4, OXER1] were over-expressed in the resistant derivatives (T,<br />

L, and T+L) compared to the parental BT474 cells. Out of these, 13<br />

belong to Class A and 6 to Class B, designated by The International<br />

Union of Basic and Clinical Pharmacology (IUPHAR). GPR110<br />

was the only GPCR common to BC stem cells as well as resistant<br />

derivatives of BT474 cells. In TCGA dataset, GPR110 expression was<br />

significantly higher in HER2+ and basal subtypes of BC compared to<br />

ER+ luminal A and B subtypes. GPR110 as well as other differentially<br />

expressed GPCRs are currently being investigated as potential targets<br />

by determining the effects of their downregulation or exogenous<br />

over-expression on the growth and drug-sensitivity of these BC cells.<br />

Conclusions: We report for the first time the differential expression<br />

of a large panel of GPCRs in the BC stem cell population and in anti-<br />

HER2 resistant derivatives of the BT474 cell line model of HER2+<br />

BC. Results of the functional studies will guide novel strategies to<br />

improve the treatment for HER2+ BC.<br />

P4-06-03<br />

Zinc Finger Nuclease Genome Engineering Reveal Multiple<br />

Functions of Secretory Leukocyte Peptidase Inhibitor in<br />

Regulating Pleuripotency of <strong>Cancer</strong> Stem Cells in Inflammatory<br />

<strong>Breast</strong> <strong>Cancer</strong><br />

Robertson FM, Hibbs S, Boley KM, Chu K, Ye Z, Wright MC, Liu<br />

H, Luo AZ, Cristofanilli M, Wemhoff G. The University of Texas MD<br />

Anderson <strong>Cancer</strong> Center, Houston, TX; Sigma-Aldrich, St. Louis,<br />

MO; Fox Chase <strong>Cancer</strong> Center, Philadelphia, PA<br />

Background: Inflammatory breast cancer (IBC) is the most aggressive<br />

and lethal variant of this disease and is known to be enriched for<br />

cells with a cancer stem cell phenotype. IBC is characterized by the<br />

presence of cell aggregates, defined as tumor emboli, that metastasize<br />

into skin and chest wall. The only documented biomarker of tumor<br />

emboli is the surface glycoprotein, E-cadherin. We previously<br />

demonstrated that IBC tumor emboli express the alarm anti-protease,<br />

secretory leukocyte peptidase inhibitor (SLPI), a metastasis related<br />

gene highly expressed in IBC patient tumors. Since the function<br />

of SLPI in IBC is unknown, the present studies used zinc finger<br />

technology to knockout (KO) copies of SLPI in the SUM149 IBC<br />

cell line to define the role of SLPI in IBC.<br />

Materials and Methods: Using CompoZr zinc finger nuclease (ZFN)<br />

technology (Sigma-Aldrich), SLPI KO cell lines were generated by<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

372s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 4<br />

disrupting all alleles (3) in exon 1 of SUM149 cells derived from<br />

an IBC primary tumor with a high CD44+cell population. The<br />

target-specific ZFNs bound DNA at a sequence-specific location<br />

and created double strand breaks repaired by non-homologous end<br />

joining, resulting in deletions at the SLPI locus. Single cell SLPI KO<br />

clones were isolated and serially passaged to establish stable cell<br />

lines. Functional assays were used to assess proliferation, invasion,<br />

tube formation and clonogenicity. Global transcriptional profiling<br />

was performed to identify genes and signaling pathways directly<br />

regulated by SLPI.<br />

Results: SUM149 SLPI KO clones did not produce SLPI protein<br />

and had a significantly slower turn-over time of 75 hrs compared<br />

with 24 hrs in SUM149 wild type clones. Loss of SLPI blocked<br />

invasion by 50%, and completely inhibited formation of tube-like<br />

structures, an activity defined as vasculogenic mimicry, characteristic<br />

of IBC. The loss of only 1 SLPI allele resulted in the inability of<br />

SUM149 cells to grow as anchorage independent clones in soft agar,<br />

commonly used as a predictor of in vivo tumorigenicity. SLPI directly<br />

regulated expression of multiple genes within the embryonic stem cell<br />

pleuripotency canonical pathway, including WNT, Frizzled, PDK-1,<br />

platelet derived growth factor receptor and sphingosine-1-phosphate<br />

receptor. Studies are underway to determine the specific role of SLPI<br />

in IBC tumor growth and metastasis.<br />

Conclusions: Our previous studies demonstrated that SLPI is<br />

expressed by IBC tumor emboli and can be used as a biomarker of<br />

tumor emboli in IBC core and skin punch biopsies. SLPI was found to<br />

not only regulate critical functional activities of IBC tumor cells but<br />

also to directly regulate genes within pathways critically important<br />

to maintenance of pleuripotency of tumors with a cancer stem cell<br />

phenotype. Collectively, these studies demonstrate the power and<br />

utility of the zinc finger technology, which enables the interrogation<br />

of tumor cells to discern the direct function and role of specific genes<br />

of interest.<br />

P4-06-04<br />

Gamma-secretase inhibitors suppress the activation of NFκB<br />

and the expression of TNFα, IL-6 and IL-8 in triple negative<br />

breast cancer cells<br />

Gu J-W, King J, Makey KL, Chinchar E, Gibson J, Miele L. University<br />

of Mississippi Medical Center, Jackson, MS<br />

Background: Inflammation mediators, such as NF-kappa B (NFκB)<br />

and tumor necrosis factor alpha (TNFα), play major role in breast<br />

cancer pathogenesis, progression, and relapse. Triple negative breast<br />

cancer (TNBC) is a heterogeneous group of aggressive breast cancers<br />

and often has a poor outcome, in which Notch pathways are highly<br />

activated. However, role of crosstalk between Notch and inflammation<br />

mediators in TNBC progression and recurrence is poorly understood.<br />

The present study determines: 1) whether NFκB is highly activated<br />

in TNBC cells such as MDA-MB-231 (claudin-low-like) and MDA-<br />

MB-468 (basal-like), compared to ER-positive cells (MCF-7); 2)<br />

whether TNFα, IL-6 and IL-8 are highly expressed in TNBC cells,<br />

compared to MCF-7 cells; 3) whether Notch inhibition by gammasecretase<br />

inhibitors (GSIs) significantly decrease NFκB activation and<br />

the expression of TNFα, IL-6 and IL-8 in TNBC cells; 4) whether<br />

GSI inhibits TNBC cell migration.<br />

Material and Methods: MDA-MB-231, MDA-MB-468, MCF-7<br />

cells were cultured using RPMI 1640 media with 10% FBS. The cells<br />

were exposed to GSIs such as DAPT and RO4929097 for 18 hours.<br />

Nuclear NFκB activation was determined by using the TransAM<br />

NFκB p65 kit (Active Motif). The expressions of TNFα, IL-6, IL-<br />

8, and IFNγ were determined by the ELISA kits (R&D Systems).<br />

Migration was determined using BD BioCoat Matrigel Invasion<br />

Chamber (BD Bioscience Discovery Labware, Sedford, MA).<br />

Results: Nuclear NFκB p65 activations (A450) were 0.292±0.015,<br />

0.222±0.005, and 0.132±0.004 in cultured MDA-MB-231, MDA-<br />

MB-468, and MCF-7 cells, respectively, in which the negative<br />

control was 0.125±0.008 and the difference between the groups<br />

were significant (P


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

S1 cells, suggesting that IR induces the formation of α5β1-integrin<br />

complexes leading to increased survival post-IR. Together these<br />

results provide the first evidence that NF-κB-induced β1-integrin<br />

transactivation is responsible for increased survival post IR in breast<br />

cancer. Since β1-integrin is shown to activate NF-κB, our data further<br />

suggest a loop-like β1-integrin–NF-κB–β1-integrin pathway in tumor<br />

radioresistance. This novel pathway may serve as an efficient drug<br />

target to re-sensitize radioresistant tumor cells.<br />

P4-06-06<br />

RNAi screen of the breast cancer genome identifies KIF14 and<br />

TLN1 as genes that modulate chemosensitivity in breast cancer<br />

Singel SM, Cornelius C, Batten K, Fasciani G, Wright WE, Lum L,<br />

Shay JW. University of Texas Southwestern, Dallas, TX<br />

We conducted a RNA-interference screen for genes whose loss-offunction<br />

enhanced doxorubicin and docetaxel chemosensitivity in a<br />

“triple-negative,” estrogen receptor negative, progesterone receptor<br />

negative, and Her2 negative (ER- PR- Her2-) breast cancer cell line,<br />

MDA-MB-231. From ranking chemosensitivity of 334 short-hairpin<br />

RNA (shRNA) MDA-MB-231 cell lines (targeting 134 genes with<br />

known somatic mutations in breast cancer), we focused on two genes,<br />

kinesin family member 14 (KIF14) and talin (TLN1) that not only<br />

enhanced chemosensitivity but also have oncogenic annotations.<br />

KIF14 has robust expression in breast cancer cells compared to<br />

normal mammary cells. TLN1 expression is important for migration<br />

of breast cancer cells. In TLN1-deficient cells, CK19 and ZO-1 are<br />

upregulated while CK14 and snail are downregulated, suggesting that<br />

TLN1 is important for the maintenance of epithelial-mesenchymal<br />

transition in MDA-MB-231 cells. KIF14 and TLN1 loss-of-function<br />

also enhanced chemosensitivity in 3 other triple negative breast cancer<br />

cell lines (HCC1937, HCC38, and Hs578T) but not in normal human<br />

mammary epithelial cells (HMECs). Examining protein-proteininteraction<br />

networks, we identified RIP2 as a target whose inhibition<br />

via SB203580 or PP2 can further enhance chemosensitization in<br />

KIF14-deficient cells. Knock-down of a number of other known<br />

protein-protein interaction partners of KIF14 and TLN1, including<br />

FAK, CIT, ARRB2, PSTPiP1, PRC1, SVIL, ITGA2B, ITGB3,<br />

VCL and PXN, do not significantly alter doxorubicin or docetaxel<br />

chemosensitivity in MDA-MB-231 cells. Mammary fat pad xenografts<br />

of KIF14- and TLN1- deficient MDA-MB-231 cells into NOD/SCID<br />

mice demonstrated significantly less tumor mass compared to control<br />

MDA-MB-231 cells after chemotherapy. Expressions of KIF14 and<br />

TLN1 from breast cancer expression arrays improve prognostic<br />

predictions compared to clinicopathological features alone. In<br />

summary, screenings for therapeutic targets using chemotherapy and<br />

genes with known somatic mutations in breast cancer not only provide<br />

a rational targeted screen, but also present possible up-front novel<br />

treatment combinations for patients with triple negative breast cancer.<br />

P4-06-07<br />

Isoform specific antibodies against the fibroblast growth factor<br />

receptor 2 have predictive and therapeutic implications in the<br />

treatment of human breast cancers<br />

Ettyreddy AR, Somarelli J, Bitting R, Armstrong A, Garcia Blanco<br />

MA. Duke University, Durham, NC<br />

Background: Aberrant induction of the epithelial to mesenchymal<br />

transition cascade (EMT) has a defining role in shaping cancer<br />

malignancy. Despite the complexity of this process, the alternative<br />

splicing of the fibroblast growth factor receptor 2 (FGFR2), between<br />

the epithelial-specific FGFR2-IIIb and mesenchymal-specific FGFR2-<br />

IIIc isoforms, has been shown to play a pivotal role in the EMT<br />

cascade. Aberrant splicing of FGFR2 in carcinomas has been shown<br />

to mark invasive cancerous cells that have undergone an EMT event<br />

and have gained the ability to thrive in diverse microenvironments.<br />

In addition to improper splicing patterns, overexpression of either<br />

isoform is sufficient to induce tumor formation by constitutively<br />

activating common oncogenic pathways. For example, a recent<br />

microarray analysis identified amplification of the FGFR2-IIIb locus<br />

as a common oncogenic signature in a significant number of treatmentresistant<br />

triple negative breast cancers (TNBC). Furthermore,<br />

inhibition of FGFR2 signaling in candidate TNBCs was sufficient to<br />

induce tumor regression. Due to the intimate link that exists between<br />

FGFR2 and breast cancer (BC), isoform-specific antibodies against<br />

the receptor may be able to identify tumors that possess deregulated<br />

FGFR2 signaling and subsequently inhibit their growth by blocking<br />

FGF ligand binding.<br />

Methods: To develop FGFR2-IIIb and —IIIc specific antibodies,<br />

we initially cloned and expressed the alternatively spliced region of<br />

each isoform and provided sufficient amount to ThermoScientific<br />

for rabbit immunizations. After the 72-day protocol was completed,<br />

the raw anti-IIIb/IIIc serum was passed through a column containing<br />

immobilized GST-IIIc/IIIb, respectively, to remove cross-reactive<br />

antibodies. Antibody specificity was assessed through western blotting<br />

and immunofluorescence (IF). Therapeutic validity of serum was<br />

determined by its ability to inhibit the growth of cancers in-vitro.<br />

Results: Through western blotting, we were able to demonstrate that<br />

the anti-IIIb serum was able to specifically recognize the FGFR2-<br />

IIIb isoform. However, the anti-IIIc serum exhibited substantial<br />

cross-reactivity to FGFR2-IIIb and was therefore not included in<br />

downstream studies. By IF, the anti-IIIb serum specifically recognized<br />

FGFR2-IIIb expressing cells while exhibiting background levels of<br />

reactivity to those that expressed FGFR2-IIIc, FGFR1, or FGFR4.<br />

To test the therapeutic validity of the anti-IIIb serum, we conducted<br />

cell inhibition assays using MCF-7 (FGFR2-IIIb specific), BT-549<br />

(FGFR2-IIIc specific), and PC3 (FGFR2 negative) cancer lines to<br />

test whether the growth inhibitory effects of the antibody could<br />

specifically target FGFR2-IIIb expressing cancer cell lines. After<br />

5 days of treatment with anti-IIIb or pre-immune rabbit serum, we<br />

observed potent growth inhibition of the MCF-7 BC cells (p=.02) in<br />

anti-IIIb treated wells and minimal effects on BT-549 and PC3 cells.<br />

Conclusion: Based upon the observed data, the anti-IIIb serum<br />

that we developed not only identified FGFR2-IIIb expressing cells<br />

through IF, but also inhibited their growth in vitro. Taken together,<br />

our data suggest that FGFR2-IIIb may be a promising therapeutic<br />

and predictive marker.<br />

P4-06-08<br />

Clinical utility of functional analysis of FGFR kinase family for<br />

selecting patients who may benefit from FGFR inhibitors<br />

Hoe N, Zhou J, Kuy C, Jin K, Srikrishnan R, Soundararajan A, Ma<br />

Y, Liu X, Singh S. Prometheus Labs, <strong>San</strong> Diego, CA<br />

Background:<br />

Fibroblast growth factor receptor (FGFR) family of receptor tyrosine<br />

kinases (RTKs) are currently under investigation as therapeutic<br />

targets for the treatment of breast cancer. Members of this kinase<br />

family have been associated with breast cancer progression and<br />

resistance to current therapeutics. FGFR1 amplifications range from<br />

7.5-17% in breast cancers and are associated with a shorter overall<br />

survival. FGFR1 amplified cell lines drive activation of AKT, ERK,<br />

and RSK and are highly sensitive to FGFR1 inhibition, suggesting<br />

an oncogenic addiction to FGFR1. FGFR2 is activated in lapatinibresistant,<br />

HER2-positive cells. Likewise, FGFR3 expression levels<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

374s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 4<br />

are elevated in tamoxifen-resistant, ER positive patients. FGFR 4<br />

overexpression correlated with poor response to chemotherapy due<br />

to activation of MAPK and increase in B-cell lymphoma extra large<br />

(BCL-XL) levels. Using an immuno-microarray system, this study<br />

seeks to determine FGFR signal transduction pathway modulation<br />

post FGFR inhibitor treatment in breast cancer cell lines expressing<br />

varying levels of FGFR1, 2, 3 or 4.<br />

Method:<br />

FGFR 1, 2, 3, and 4 amplified cell lines (MDA-MB-134VI (KRAS),<br />

SNU16, RT112, and MDA-MB-453) were treated with varying<br />

dosages (1, 10, 100, and 1000nM) of several FGFR inhibitors<br />

(AZD4547 (pan FGFR), PD173074 (FGFR1,3) and Ponatinib-<br />

AP24534 (FGFR1)) either in presence or absence of corresponding<br />

FGF (FGF1, FGF7, FGF9, and FGF19) stimulation. Expression<br />

and activation of FGFR 1, 2, 3, and 4 and downstream signaling<br />

proteins FRS2, AKT, ERK, MEK, and RSK were measured utilizing<br />

a proximity based immuno-microarray.<br />

Results:<br />

FGFR1, 2, 3, 4 protein expression was detected in each of the<br />

corresponding four FGFR1, 2, 3, 4 amplified cell lines. Total protein<br />

levels remained unchanged with or without FGFs stimulation. Levels<br />

of FGFR1 phosphorylation increased 5 folds in MDA-MB-134VI<br />

stimulated with FGF1. FGFR2 and FGFR3 activation increased by<br />

2-3 folds in SNU16 and RT112 stimulated with FGF7, and FGF9<br />

respectively. FGFR4 activation levels in MDA-MB-453 remained<br />

unchanged due to Y367C mutation. ERK was activated in both FGF1<br />

stimulated and unstimulated group in MDA-MB134 due to the KRAS<br />

mutation. RSK activation increased in both SNU16 and RT112 when<br />

stimulated with FGF7, and FGF9 respectively. FGFR inhibitors<br />

treated data analysis is currently in progress and will be presented.<br />

Conclusion:<br />

We have demonstrated utility of specific FGFR analysis using well<br />

defined model systems. FGFR- profiling should be considered during<br />

the clinical work-up in order to select patients who may benefit from<br />

regimen containing specific FGFR inhibitors.<br />

P4-06-09<br />

Cell surface receptor CDCP1 as a potential marker of triple<br />

negative breast cancers progression<br />

Campiglio M, Sasso M, Bianchi F, De Cecco L, Turdo F, Triulzi<br />

T, Orlandi R, Morelli D, Aiello P, Ghirelli C, Agresti R, Tagliabue<br />

E. Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy;<br />

Fondazione IRCCS Istituto Nazionale dei Tumori<br />

The risk of recurrence and death in triple negative breast cancer<br />

(TNBC) peaks between the first and the third year after surgery and<br />

to date, there is still an urgent need to define specific markers for<br />

therapeutic targeting. Surgical wound healing process was shown<br />

to contribute to tumor recurrence since molecules release during<br />

wounding, which can contribute to create a suitable environment<br />

for tumor progression, are released into the blood and may stimulate<br />

disseminated metastatic cells. Therefore, wound-healing fluids (WHF)<br />

from breast cancer patients has been used as a tool representing the<br />

repertoire of growth factors and cytokines released after surgery<br />

able to stimulate TNBC progression. By treating TNBC cell lines<br />

with WHFs collected 24 hr post-surgery of breast cancer patients,<br />

we identified CDCP1, a cell surface receptors with CUB domain<br />

significantly up modulated by WHFs in 4 out of 5 TNBC cell lines.<br />

CDCP1, reported to be a regulator of tumor metastasization and<br />

a resistant factor to anoikis, was found highly expressed in some<br />

epithelial tumors, i.e. colon and lung and associated to a poor<br />

prognosis. By in silico analysis of gene expression public datasets of<br />

breast cancer specimens, we found a significant correlation between<br />

CDCP1 expression level and risk of relapse. Additionally, in a<br />

dataset of breast cancer cell lines we found an increased expression<br />

of CDCP1 in basal respect to luminal cancer cells. To move forward<br />

with the investigation of the CDCP1 role in the TNBC subtype, we<br />

analyzed CDCP1 protein in 12 TNBC and 8 luminal breast cancers<br />

and found a strong membrane staining in all the TNBCs, while in<br />

only 2 luminal cases faint membrane reactivity was observed. TNBC<br />

cells CDCP1-silenced by specific siRNA were not affected in their<br />

in vitro growth, while they were strongly and significantly impaired<br />

in their capability to migrate and invade; CDCP1-shRNA of TNBC<br />

cells was performed to investigate CDCP1 impact on growth and<br />

metastatization in SCID mice. In conclusion, our data suggest that<br />

CDCP1 is a new potential receptor promoting TNBC progression.<br />

Supported by AIRC (Associazione Italiana Ricerca sul Cancro).<br />

P4-06-10<br />

Epigenetic silencing of glutamine synthetase (Glul) defines<br />

glutamine depletion therapy<br />

Cavicchioli F, Shia A, O’Leary K, Haley V, Crook TR, Thompson<br />

AM, Lackner M, Lo Nigro C, Schmid P. Brighton and Sussex Medical<br />

School, University of Sussex, Brighton, United Kingdom; Ninewells<br />

Hospital, University of Dundee, United Kingdom; Genentech, Inc.,<br />

<strong>San</strong> Francisco; S. Croce General Hospital, Cuneo, Italy<br />

Background: Methylation-dependent transcriptional silencing of<br />

genes involved in amino acid synthesis can provide potential targets<br />

for novel synthetic lethality strategies. Glutamine synthetase (Glul)<br />

is the key enzyme in the biosynthesis of glutamine. We identified<br />

Glul as a novel gene subject to methylation-dependent transcriptional<br />

silencing in breast cancer cell lines using a combined functional screen<br />

with methylation reversal assays and methylation arrays.<br />

Methods: Methylation reversal assays were performed using<br />

5-aza-2-deoxycytidine and/or trichostatin treatment coupled with<br />

whole genome mRNA microarrays (Illumina HT-12 v4 Expression<br />

BeadChip Kit). Expression of Glul with and without pharmacological<br />

methylation reversal with azacytidine and/or trichostatin was<br />

validated using qRT-PCR and Western Blot. Methylation of Glul was<br />

analysed using methylation microarrays (Illumina 450K Methylation<br />

BeadChip), bisulphite sequencing and pyrosequencing. Sensitivity<br />

to glutamine deprivation was assessed using an MTT assay after<br />

culturing cells in media with various glutamine concentrations or in<br />

complete absence of glutamine. We used a panel of 55 breast cancer<br />

cell lines and formalin-fixed paraffin-embedded tissue from a series<br />

of 116 stage I-III primary breast cancers with linked mature clinical<br />

outcome data that were randomly selected from the Cuneo Tissue<br />

Bank. Tissue samples were subject to histopathological review to<br />

ensure adequate representation of cancer cells.<br />

Results: Dense methylation of the CpG-island of Glul was detected<br />

in 45% of cell lines across all subtypes. Methylation of the CpG<br />

island was linked with absent or down-regulated expression of Glul<br />

in some but not all cell lines, and Glul expression could be reactivated<br />

by azacytidine and trichostatin in these cell lines. Methylation of<br />

shore areas was detected in several cell lines but was not associated<br />

with transcriptional silencing. Cells with methylation-dependent<br />

low or absent Glul expression were highly sensitive to glutamine<br />

deprivation, whereas cell lines without Glul methylation were rescued<br />

by compensatory up-regulation of Glul. Using pyrosequencing, dense<br />

methylation of the CpG island of Glul was found in 32.8% of patients,<br />

with an additional 17.2% of patients showing partial methylation.<br />

No significant association with a specific breast cancer subtype or<br />

outcome was found.<br />

www.aacrjournals.org 375s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Conclusions: This is the first report of methylation-dependent<br />

transcriptional silencing of Glul expression in cancer. Our data<br />

demonstrate that a significant proportion of primary breast cancers<br />

show methylation of Glul and suggest that glutamine deprivation<br />

could be a novel synthetic lethality strategy for these cancers.<br />

P4-06-11<br />

Expression of genes spanning a breast cancer susceptibility locus<br />

on 6q25.1 is modulated by epigenetic modification<br />

Dunbier AK, White J, Van Huffel S. University of Otago, Dunedin,<br />

Otago, New Zealand<br />

Background:<br />

Genome wide association studies have identified single nucleotide<br />

polymorphisms around C6ORF97, an open reading frame (ORF)<br />

immediately upstream of the gene encoding ERα (ESR1) on 6q25.1,<br />

to be associated with increased risk of breast cancer 1,2 . C6ORF97<br />

and two neighbouring genes, RMND1 and C6ORF211, are also<br />

co-expressed with ESR1 in oestrogen receptor positive (ER+ve)<br />

breast cancers and C6ORF97 expression is inversely associated<br />

with proliferation and recurrence free survival in a tamoxifen-treated<br />

patient cohort 3 . Epigenetic modification represents one potential<br />

mechanism through which expression of co-localised genes could<br />

be modulated.<br />

Aim:<br />

To investigate the role of epigenetic modification in the co-expression<br />

of ESR1, RMND1, C6ORF211 and C6ORF97.<br />

Methods:<br />

Methylation status and expression of genes in the ESR1/C6ORFs locus<br />

was assessed in postmenopausal breast cancers and ER+ve cell lines.<br />

Two ER+ve cell lines, MCF7 and T47D, were cultured in oestrogendepleted<br />

media and samples were taken fortnightly for 6 months.<br />

DNA and RNA from ER+ve tumours was also analysed in addition to<br />

control ER-ve cell lines. Methylation analysis of CpG Islands within<br />

the ESR1/C6ORFs locus was carried out using methylation sensitive<br />

sequencing analysis. Expression levels of ESR1, RMND1, C6ORF211<br />

and C6ORF97 were determined using quantitative real-time PCR.<br />

Results:<br />

Variation in methylation patterns was observed at a number of key<br />

CpG islands in the ESR1/C6ORFs region. ER+ve cell lines and<br />

highly ER+ve tumours were hypomethylated. Hypomethylation was<br />

associated with increased expression of ESR1, RMND1, C6ORF211<br />

and C6ORF97. Temporal changes in methylation and expression<br />

of these genes were also observed over the period of oestrogen<br />

deprivation in both MCF7 and T47D.<br />

Conclusions:<br />

Methylation at the ESR1/C6ORFs locus is associated with expression<br />

levels of ESR1, RMND1, C6ORF211 and C6ORF97. This suggests<br />

that methylation is involved in modulating the expression of these<br />

four genes and that it could partially explain the previously observed<br />

co-expression of the genes. Our finding that hypomethylation in this<br />

region occurs in tumours with high ERα expression suggests that it<br />

could be a mechanism through which tumours increase ERα levels<br />

to maximise growth stimulation from oestradiol. Our observation<br />

that RMND1, C6ORF211 and C6ORF97 also increase in expression<br />

suggests that the proteins encoded by these genes may contribute to<br />

the phenotype exhibited by ER+ve breast tumours.<br />

References:<br />

1. Turnbull C et al. Nature Genetics 2010;42:504-7.<br />

2. Zheng W et al. Nature Genetics 2009;41:324-8.<br />

3. Dunbier A et al. PLoS Genetics 2011;7:e1001382.<br />

P4-06-12<br />

Monoclonal antibodies against nicastrin for the treatment of<br />

breast canmcer: in vitro and in vivo characterisation and function<br />

Filipovic A, Lombardo Y, Deonarain M, Giamas G, Cordingley H,<br />

Tralau-Stewart C, Coombes RC. Imperial College London, United<br />

Kingdom<br />

Nicastrin (NCT) is a crucial structural and functional constituent of the<br />

gamma-secretase (GS) enzyme which activates a range of substrates<br />

(Notch1-4, CD-44, Her-4, E-cadherin, EpCAM etc.), with a causative<br />

role in development and progression of malignant disease. Nicastrin<br />

is the only GS component with a single-pass transmembrane- and a<br />

large extracellular domain (ECD). This makes NCT a potential target<br />

for monoclonal antibody (McAb) therapy.<br />

Using genetic immunization with the NCT-ECD cDNA., we raised<br />

and cloned rat hybridomas expressing anti-NCT monoclonal Abs<br />

(McAbs) (Genovac). Four clones (NMA1-4) were taken to complete<br />

McAb production and purification, based on their ability to recognise<br />

cell surface NCT on breast cancer cells and potency to inhibit breast<br />

cancer cell invasion. We have established the binding affinities of<br />

the McAbs using non cell based and cell based assays, as well as<br />

performed competition binding experiments using surface plasmon<br />

resonance, which have revealed that the two McAbs bind distinct<br />

epitopes on NCT ECD. Although nicastrin is a heavily glycosylated<br />

protein, dyglycosylation experiments using EndoH and PNGase have<br />

revealed that the McAbs1-4 most likely bind peptide sequences of<br />

nicastrin ECD. We will also present further data from the PEPScreen<br />

experiment used to further delineate the binding epitopes of the antinicastrin<br />

McAbs. In terms of mechanism of action, we have shown<br />

that McAb2 affects the Akt pathway, while the McAb4 acts on the<br />

Cdc42-RhoGTPases signalling axis.<br />

Importantly, we have performed an in vivo mammary fat pad xenograft<br />

mouse model of triple negative breast cancer (MDA-MB-231), where<br />

the i.v. injected McAb2 inhibited tumour growth by 50% over 25<br />

days (p = 0.007). We will present the data from genome microarray<br />

analyses used to assess tumour tissues (control arm and McAb2 arm)<br />

from the mouse xenograft model.<br />

In patient samples we have previously shown (Filipovic et al, <strong>Breast</strong><br />

<strong>Cancer</strong> Research and Treatment, 2011) that nicastrin is overexpressed<br />

in breast cancer tissue compared to normal breast, (n= 1050) where it<br />

confers worse overall survival in estrogen receptor negative patients.<br />

We have now established that nicastrin can be detected in the serum<br />

of breast cancer patients by ELISA, where as a circulaing marker, its’<br />

levels are increased in mataststic patients compared to the primary<br />

BC patients.<br />

We conclude that anti-nicastrin McAb therapy may be an efficient<br />

strategy to target nicastrin expressing breast cancer.<br />

P4-06-13<br />

Effects of Statin on triple-negative breast cancer (TNBC) with<br />

Ets-1 overexpression<br />

Lee S, Jung HH, Park YH, Ahn JS, Im Y-H. Samsung Medical Center<br />

Background: Triple negative breast cancer (TNBC) is diagnosed<br />

accounting for approximately 15-20% of all breast cancer diagnoses<br />

and an aggressive clinical phenotype characterized by lack of<br />

expression of estrogen receptor (ER) and progesterone receptor (PR)<br />

as well as the absence of human epidermal growth factor receptor-2<br />

(HER-2) overexpression. Because of its expression profile, TNBC is<br />

not amenable to treatment with hormone therapy or the anti-HER2<br />

monoclonal antibody trastuzumab, and systemic treatment options<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

376s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 4<br />

are limited to cytotoxic chemotherapy. At present, there is not a clear,<br />

proven effective single agent that targets a defining vulnerability in<br />

TNBC.<br />

The proto-oncogene Ets-1 is a member of the Ets family of<br />

transcription factors which share a unique DNA binding domain, the<br />

Ets domain. Ets proteins are targets for phosphorylation in response<br />

to stimulation by a variety of different growth modulators, including<br />

intracellular calcium, activators of protein kinase C pathways,<br />

growth factors and cytokines. The importance Ets genes in human<br />

carcinogenesis is supported by the observations that Ets genes<br />

have altered expression patterns, are chromosomally amplified or<br />

deleted, or are located at translocation breakpoints in leukemia and<br />

solid tumors. In model systems, increased expression of Ets-1 was<br />

found to be associated with enhanced angiogenesis and the invasive<br />

phenotype. Studies in breast cancer cell lines have implicated Ets-1<br />

in the progression of breast cancer.<br />

The present study was conducted to better understand the molecular<br />

mechanisms underlying statin-induced suppression associated with<br />

transcription factor Ets-1 overexpressed TNBC.<br />

Methods: We evaluated the anti-tumor effects of simvastatin on<br />

TNBC cells using a MTT assay, invasion assay, siRNA transfection,<br />

western blotting and xenograft study which were used to address<br />

the role of Ets-1 activity and the Erk/Akt pathway on the effect of<br />

simvastatin.<br />

Results: We demonstrated that the expression of Ets-1 was increased<br />

particular in TNBC cells among various breast cancer cell lines<br />

and the simvastatin statistically significantly enhanced antitumor<br />

activity in Ets-1 overexpressed TNBC cells. In a mouse model, the<br />

growth of Ets-1 expressed TNBC xenograft tumors was statistically<br />

significantly inhibited when simvastatin was treated. Furthermore,<br />

our data demonstrated for the first time that simvastatin inhibited the<br />

growth of TNBC cells by inhibiting Ets-1 activity via Erk and Akt<br />

pathway in a dose-dependent manner.<br />

Conclusion: Our results suggest that the inhibition of Ets-1 acitivity<br />

via Erk and Akt pathway may be a novel mechanism by which<br />

simvastatin suppresses the growth of TNBC cells. The ability of<br />

simvastatin to induce cell death via Ets-1, as well as its ability<br />

to downregulate signaling through Ras/Raf/MEK/Erk and PI3K/<br />

Akt pathway, suggested translational value. Exploitation of this<br />

activity might include a combination of Ras/Raf/MEK/Erk or PI3K/<br />

Akt/mTOR inhibitors and simvastatin to induce cell death or the<br />

combination of simvastatin in these signaling pathways. Further<br />

preclinical and clinical studies are warranted to further investigate<br />

the application of simvastatin for the treatment of TNBC.<br />

P4-06-14<br />

CD146-suppresses breast tumor invasion via a novel transcription<br />

target TIMPv<br />

Shanmuganathan S, AbdElmageed Z, Fernando A, Gaur R, Ramkumar<br />

A, Bhat S, Gupta I, Muzumdar S, Hakkim L, Ouhtit A. Sultan Qaboos<br />

University, Al-Khod, Seeb, Oman<br />

The function of the cell adhesion receptor CD146, a recently<br />

discovered marker of endothelial cells and a tumor promoter of<br />

melanoma and other cancers, is controversial in breast cancer (BC).<br />

However several lines of evidence supports its role as a tumor<br />

suppressor in BC. Further, the molecular mechanisms underpinning<br />

this suppression are unknown, neither has the ligand for CD146 been<br />

identified. Using a novel validated Enhanced Green Fluorescent<br />

Protein (EGFP)-inducible systems of CD146 expression both in vitro<br />

and in vivo, we provide here molecular and functional evidence of<br />

CD146 and its novel transcriptional target TIMPv (a variant of tissue<br />

inhibitor of metallo-proteinases) in underpinning the suppression of<br />

BC invasion.<br />

Tetracycline (tet-on) CD146 system was developed in both MCF-7<br />

and MDA-231 BC founder cell lines, and validated using time course<br />

RT-PCR and western blot analyses, and fluorescent microscopy.<br />

In functional experiments, induction of CD146 inhibited BC cell<br />

migration and invasion. TIMPv, the only endogenous protein<br />

inhibitor known for metallocarboxypeptidases, was identified by<br />

expression profiling as a novel transcriptional target of CD146-<br />

signaling, an association validated by quantitative PCR and<br />

immunoblotting experiments in a range of breast and melanoma<br />

cancer cells. However, siRNA inhibition of CD146 in the SKMel-28<br />

melanoma cell line increased TIMPv expression, suggesting that<br />

while TIMPv is a positive transcriptional target of CD146 in BC<br />

cells, it is negatively regulated in melanoma cells. Furthermore,<br />

using invasion assay, the functional relevance of TIMPv to CD146-<br />

suppressed metastasis was demonstrated by selective suppression<br />

of TIMPv in CD146-expressing BC inducible cells using RNAi.<br />

More interestingly, induction of CD146 expression in vivo, using<br />

the tet-on CD146 expression system in BC Xenograft model resulted<br />

in suppression of breast tumor growth. Further, Clinical analysis of<br />

breast tissue samples by Immunohistochemistry showed that TIMPv<br />

expression patterns paralleled those of CD44s during breast tumor<br />

progression. Pharmacological and molecular approaches revealed<br />

that the activation of NFκB via Akt pathway couples CD146 to the<br />

transcription of TIMPv in BC cells.<br />

Our study is the first report to provide a functional molecular link<br />

of a novel transcriptional target of CD146, TIMPv, to cancer via a<br />

unique axis that underpin CD146-suppressed BC progression; TIMPv<br />

is a potential target for guiding the development of novel therapeutic<br />

strategies for BC.<br />

P4-06-15<br />

Mifepristone modifies the tumor microenvironment increasing<br />

the therapeutic efficiency of low doses of Doxorubicin liposomes<br />

or paclitaxel-albumin nanoparticles in a murine model of breast<br />

cancer.<br />

Sequeira GR, Vanzulli SI, Lamb CA, Rojas PA, Lanari C. Instituto<br />

de Biología y Medicina Experimental, Ciudad Autónoma de Buenos<br />

Aires, Argentina; Academia Nacional de Medicina, Ciudad Autónoma<br />

de Buenos Aires, Argentina<br />

Chemotherapy is the standard treatment for metastatic breast cancer<br />

today. To overcome undesired side effects and improve drug efficacy in<br />

tumor cells, different nanoparticle formulations have been developed.<br />

Paclitaxel has been bound with human albumin (Nab-paclitaxel), and<br />

pegylated liposomes have been developed for doxorubicin delivery<br />

(PEG-LD). The aim of our study was to evaluate the effect of these<br />

therapeutic agents in experimental mammary carcinomas expressing<br />

different ratios of progesterone receptor (PR) isoforms and to evaluate<br />

the effect of combined treatments of chemotherapeutic agents with<br />

the antiprogestin mifepristone (MFP). Nab-paclitaxel (Abraxane,<br />

Abraxis), at a dosage of 30 and 60 mg/kg body weight administered<br />

in three doses every four days, completely inhibited the growth of the<br />

antiprogestin-resistant C4-2-HI mouse mammary carcinoma, which<br />

is associated with high levels of PR isoform B (PR-B) expression.<br />

C4-HI, an antiprogestin-responsive mouse tumor that shows high<br />

levels of PR isoform A (PR-A) expression, was not inhibited by<br />

the low Nab-paclitaxel dose. However, the combination of MFP<br />

(6 mg silastic pellet) with Nab-paclitaxel improved the efficacy of<br />

both single treatments. Similar experiments were conducted using<br />

www.aacrjournals.org 377s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

PEG-LD (Doxopeg; Laboratorios Raffo). In this case, all tumors<br />

tested (C4-HI, CC4-3-HI, 59-2-HI and C4-2-HI) were all highly<br />

responsive to high (18 mg/kg/week) or low (9 mg/kg/week) PEG-LD<br />

doses. When PEG-LD concentrations were lowered to 4.5 mg/kg/<br />

week and they were combined with MFP, an improved response was<br />

observed with combined treatments only in tumors with high PR-A<br />

levels (p


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 4<br />

June 2008]. After assessing how common was CRG disregulation in<br />

different epithelial cancers, via comparative genomic analysis of CRG<br />

expression patterns in tumor vs. normal tissue, we found that 34 CRGs<br />

(∼35%) were significantly disregulated in breast cancer. Because of<br />

the need for novel intervention targets in basal-like breast cancer, we<br />

focused our functional analysis there. We have tested a subset of CRGs<br />

disregulated in breast tumors for their contribution to malignancy in<br />

basal-like breast cancer cells, re-setting CRG levels toward normal<br />

cell levels by stable cDNA expression or shRNA-mediated knockdown<br />

and testing tumor formation capacity of perturbed cells. Our<br />

studies reveal that CRGs play an essential role in controlling tumor<br />

formation of basal-like breast cancer cells, with significant reductions<br />

in tumor volume, and in some cases reduced tumor incidence, upon<br />

CRG modulation. Disruption of cell behavior appears to be limited to<br />

cancer cells, as non-transformed mammary cells harboring analogous<br />

perturbations retain capacity to form acinar structures in 3D culture.<br />

Several FDA approved pharmacological agents, identified to reverse<br />

the CRG expression pattern, display inhibitory effects on basal-like<br />

breast cancer cell lines, with minimal effects on non-transformed<br />

cells. CRGs thus represent cancer cell-specific vulnerabilities in basallike<br />

breast cancer, providing novel and unexpected opportunities for<br />

intervention against this difficult-to-target disease.<br />

P4-06-19<br />

Astemizole and calcitriol: A novel targeted therapeutic strategy<br />

for breast cancer<br />

García J, García R, Villanueva O, <strong>San</strong>tos N, Barrera D, Avila E,<br />

Ordaz D, Halhali A, Camacho J, Larrea F, Díaz L. Instituto Nacional<br />

de Ciencias Médicas y Nutrición Salvador Zubirán, México, DF,<br />

Mexico; Centro de Investigación y de Estudios Avanzados del I.P.N.,<br />

México, DF, Mexico<br />

Calcitriol antiproliferative effects include inhibition of the oncogenic<br />

ether-à-go-go-1 potassium channel (Eag1), which expression and<br />

activity are necessary for cell cycle progression and tumorigenesis.<br />

Astemizole, a new promising antineoplastic drug, targets Eag1 by<br />

blocking ion currents. In vitro, we showed that astemizole synergized<br />

calcitriol antiproliferative effects by downregulating CYP24A1, the<br />

enzyme that degrades calcitriol, and upregulating the vitamin D<br />

receptor (VDR), which mediates calcitriol actions. In this study we<br />

investigated the antineoplastic effect of astemizole and calcitriol in a<br />

murine model xenografted with T-47D and invasive ductal carcinomaderived<br />

breast cancer cells (IDC). Tumor-bearing nude mice were<br />

treated with oral astemizole (50 mg/kg/day) and/or i.p. calcitriol<br />

(30 µg/kg/twice a week) during 3 weeks. Astemizole and calcitriol<br />

significantly inhibited, while the coadministration of both drugs<br />

further reduced, tumor growth compared to untreated controls. These<br />

results were supported by immunohistochemistry studies of Ki-67, a<br />

proliferation marker, which revealed less staining in tumors from mice<br />

with the co-treatment compared to controls and calcitriol or astemizole<br />

alone. In a similar manner as reported in vitro, we observed that the<br />

concomitant administration of astemizole and calcitriol upregulated<br />

VDR tumor expression (P < 0.05). Conclusion: Coadministration of<br />

calcitriol and astemizole increased the antineoplastic effects of both<br />

drugs. Our results indicate that astemizole may improve calcitriol<br />

bioactivity by upregulating tumoral VDR. In addition, the findings<br />

in this study suggest that VDR-negative tumors could be sensitized<br />

to calcitriol antineoplastic effects by concomitant administration with<br />

astemizole, which would also allow for less calcitriol dosing without<br />

sacrificing overall therapeutic efficacy. Herein we suggest a novel<br />

combined adjuvant therapy for the management of breast cancer<br />

tumors. These results provide scientific bases for further studies<br />

designed to test both compounds simultaneously in patients bearing<br />

solid or metastatic tumors.<br />

P4-07-01<br />

The Class I Selective PI3K Inhibitor GDC-0941 Enhances<br />

the Efficacy of Docetaxel in Human <strong>Breast</strong> <strong>Cancer</strong> Models by<br />

Increasing the Rate of Apoptosis<br />

Wallin JJ, Guan J, Prior WW, Lee LB, Ross L, Belmont LD, Koeppen<br />

H, Belvin M, Sampath D, Friedman LS. Genentech, Inc., South <strong>San</strong><br />

Francisco, CA<br />

Docetaxel (DTX) is commonly used as a front-line treatment option<br />

for breast cancer but many patients ultimately relapse and succumb to<br />

disease progression. Phosphatidylinositol 3-kinases (PI3K) are lipid<br />

kinases that regulate breast tumor cell growth, migration and survival.<br />

Given that phosphatidylinositol 3-kinase (PI3K) is frequently<br />

activated in breast cancer and induces chemo-resistance, it is an<br />

attractive target for combination therapy with standard of care drugs<br />

such as DTX. The current study was intended to determine if GDC-<br />

0941, an orally bioavailable class I selective PI3K inhibitor, enhances<br />

the anti-tumor activity of DTX in human breast cancer models in vitro<br />

and in vivo. A panel of 25 breast tumor cell lines representing luminal,<br />

HER2+ and basal sub-types were treated with GDC-0941, DTX or<br />

the combination of both drugs and assayed for cellular viability,<br />

modulation of PI3K pathway markers and apoptosis induction. The<br />

combination of GDC-0941 and DTX decreased the cellular viability<br />

of breast tumor cell lines in vitro but with variable drug synergy.<br />

The addition of both drugs resulted in stronger synergistic effects in<br />

a sub-set of tumor cell lines that was not predicted by breast cancer<br />

sub-type or mutations within the PI3K pathway. Human xenografts<br />

of breast cancer cell lines and patient-derived tumors were utilized<br />

to assess efficacy of GDC-0941 and DTX in vivo. In these models,<br />

the best combination efficacy was detected when the two drugs were<br />

dosed within 1 hour of each other. We also observed that GDC-0941<br />

increased the rate of apoptosis in cells arrested in mitosis upon cotreatment<br />

with DTX. Our data provides a preclinical rationale for<br />

evaluating GDC-0941 in combination with DTX for breast cancer<br />

treatment.<br />

P4-07-02<br />

Withdrawn by Author<br />

P4-07-03<br />

Rapamycin and Dalotuzumab in combination inhibit parental<br />

and endocrine resistant breast cancer cells.<br />

Beckwith H, Fettig-Anderson L, Yueh N, Douglas Y. University of<br />

Minnesota, Minneapolis, MN<br />

The PI3K/Akt/mTOR pathway is involved in breast cancer resistance<br />

to endocrine therapy. Inhibitors of mTOR have been shown to have<br />

benefit for patients with ER positive tumors that have developed<br />

endocrine therapy resistance. Inhibition of mTOR triggers a negative<br />

feedback loop resulting in enhanced phosphorylation of Akt and<br />

subsequent breast cancer cell proliferation via the IGF signaling<br />

pathway. Therefore, blockade of both IGF-1R and mTOR may be<br />

necessary to completely suppress this pathway. Our laboratory studied<br />

the effect of dual inhibition of mTOR and IGF-1R with rapamycin<br />

and dalotuzumab respectively on parental and endocrine resistant<br />

MCF-7 breast cancer cell tumorigenesis.<br />

Immunoblotting demonstrates parental MCF-7L cells treated with<br />

rapamycin alone demonstrate increased phosphorylation of Akt.<br />

www.aacrjournals.org 379s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Inhibition of phosphorylation of Akt can be achieved with combined<br />

treatment with rapamycin and dalotuzumab at concentrations of 5 nM<br />

and 2 µg/mL respectively. In addition to inhibition of pAkt, combined<br />

treatment inhibits phosphorylation of S6K1 and the translational<br />

repressor protein, 4eBP1. Furthermore, MTT proliferation assay and<br />

soft agar assay demonstrate inhibition of parental cell proliferation and<br />

colonization respectively with combined rapamycin and dalotuzumab<br />

treatment. In order to examine the effect of combination therapy in<br />

endocrine resistant cell lines, we established tamoxifen resistant<br />

(TamR) and long term estrogen deprived (LTED) cell lines. TamR<br />

cells were generated by culturing MCF-7L cells in the presence<br />

of tamoxifen for more than 1 year. LTED cells were generated by<br />

culturing MCF-7L cells in the absence of estrogen for more than 6<br />

months. Proliferation of TamR and LTED cells treated with rapamycin<br />

at a concentration of 5nM and dalotuzumab at a concentration of 2µg/<br />

mL in combination was significantly inhibited. We conclude that<br />

combination of IGF1R and mTOR inhibition might be necessary to<br />

inhibit endocrine resistant breast cancers. Ongoing clinical trials will<br />

test this combination of drugs.<br />

P4-07-04<br />

Methioninase cell-cycle synchronization potentiates chemotherapy<br />

for breast cancer<br />

Yano S, Li S, Han Q, Tan Y, Fujiwara T, Hoffman RM. Anti<strong>Cancer</strong><br />

Inc., <strong>San</strong> Diego, CA; Okayama University Graduate School of<br />

Medicine and Dentistry, Okayama, Japan; University of California,<br />

<strong>San</strong> Diego, CA<br />

Deprivation of methionine selectively arrests cancer cells during late<br />

S-phase (Proc. Natl. Acad. Sci. USA 77, 7306-7310, 1980; Biochim.<br />

Biophys. Acta, Reviews on <strong>Cancer</strong> 738, 49-87, 1984), where they<br />

are highly sensitive to chemotherapy drugs which damage DNA (J.<br />

Natl. <strong>Cancer</strong> Inst. 76, 629-639, 1986). <strong>Cancer</strong> cells, transformed to<br />

express different color fluorescent reporters during specific phases<br />

of the cell cycle (Cell 132, 487-498, 2008), were used to monitor the<br />

onset of the S/G 2 -phase block due to methionine deprivation effected<br />

by recombinant methioninase (rMETase). The S/G 2 -phase blocked<br />

cancer cells fluoresced yellow or green in contrast to cancer cells in<br />

G 1 which fluoresced red. <strong>Cancer</strong> cells, including MCF-7 breast cancer,<br />

synchronously blocked in S/G 2 -phase by rMETase, were identified<br />

by their yellow-green fluorescence and allowed to accumulate to the<br />

maximum extent. At the point of maximum yellow/green cells in<br />

the culture, the cells were administered chemotherapy drugs which<br />

interact with DNA or block DNA synthesis such as doxorubicin. We<br />

termed this procedure color-coded chemotherapy (CCC). CCC was<br />

highly effective against the cancer cells (90% cell kill). In contrast,<br />

treatment of cancer cells with drugs only, and without rMETaseeffected<br />

S/G 2 -phase synchrony, led to the majority of the cancer cell<br />

population being blocked in G 1 phase (red fluorescent) where they<br />

were resistant to the drugs (40% cell kill). CCC, which identifies,<br />

by fluorescent color, when cancer cells are blocked in S/G 2 -phase<br />

by a unique cell-cycle-blocking agent, rMETase, demonstrates<br />

the potential of cell-synchronization-based chemotherapy for<br />

breast cancer.<br />

P4-07-05<br />

MEDI3379, an antibody against HER3, is active in HER2-driven<br />

human breast tumor models<br />

Xiao Z, Rothstein R, Carrasco R, Wetzel L, Kinneer K, Chen H, Tice<br />

D, Hollingsworth R, Steiner P. MedImmune, LLC, Gaithersburg, MD<br />

HER3 (ERBB3) is a member of the EGFR/HER family of receptor<br />

tyrosine kinases (RTK), consisting of EGFR, HER2, HER3 and<br />

HER4. Unlike the other HER family members, HER3 lacks intrinsic<br />

tyrosine kinase activity and therefore needs to form heterodimers<br />

with EGFR, HER2 or other kinase-proficient RTKs to be functionally<br />

active. Dimerization is induced by overexpression of EGFR or HER2<br />

in a ligand-independent (LI) manner. In this process HER3 acts as<br />

a driver in divergent cancer types including HER2-positive breast<br />

cancer (BC) via induction of the PI3K-AKT pathway. Alternatively,<br />

heregulin (NRG1/HRG), the major HER3 ligand, induces a<br />

conformational shift in HER3 which leads to dimer formation with<br />

a partner RTK in a ligand-dependent (LD) manner.<br />

We have developed an antagonistic human monoclonal antibody<br />

against HER3, termed MEDI3379, and tested it in multiple breast<br />

cancer cell lines. We observed effective suppression of constitutive<br />

pHER3 and pAKT with MEDI3379, leading to anti-proliferation<br />

effects in cell culture models. Preclinical evaluation of MEDI3379<br />

demonstrated antitumor activity in several orthotopic BC xenografts in<br />

nude mice which did not express HRG. For example, the BC xenograft<br />

model BT474 with amplified HER2 responded to MEDI3379 (65%<br />

dTGI). In conclusion, our findings with targeting of HER3 in mouse<br />

models support continued development of MEDI3379 for cancer.<br />

P4-08-01<br />

PI3K/mTOR inhibition overcomes in vitro and in vivo trastuzumab<br />

resistance independent of feedback activation of pAKT<br />

O’Brien NA, McDonald K, Tong L, Von Euw E, Conklin D, Kalous O,<br />

Di Tomaso E, Schnell C, Linnartz R, Hurvitz SA, Finn RS, Hirawat<br />

S, Slamon DJ. UCLA, Los Angeles, CA; Novartis Pharmaceuticals,<br />

Cambridge, MA; Novartis Pharmaceuticals, Basel, Switzerland<br />

Background: Aberrant activity of the PI3K/mTOR pathway has been<br />

implicated in resistance to trastuzumab and anti-hormonal therapy for<br />

HER2-amplified and ER-positive breast cancer, respectively. Previous<br />

studies in our laboratory and others have shown that increased PI3K/<br />

mTOR signaling, either through PTEN loss or activating PIK3CA<br />

mutations, can confer resistance to trastuzumab therapy. In this study,<br />

we assessed the potential of targeting the PI3K/mTOR pathway in<br />

overcoming both de novo and acquired trastuzumab resistance.<br />

Materials and Methods: The in vitro activity of the pan-PI3K<br />

inhibitor BKM120, the mTORC1 inhibitor RAD001 and the dual<br />

PI3K/mTORC1/2 inhibitor BEZ235 were evaluated in a panel of<br />

49 human breast cancer and immortalized cell lines. The in vivo<br />

activity of these molecules was assessed in six cell line xenografts<br />

models representing, ER+/HER2- (KPL-1, ZR75-1), ER+/HER2+<br />

(UACC812, MDA361), ER-/HER2+ (SUM190), PIK3CA mutant<br />

(SUM190, MDA361), PTEN-null (ZR75-1) and trastuzumab resistant<br />

(BT-TR) breast cancer. Finally, the effect of PI3K/mTOR inhibition<br />

on feedback activation of PI3K signaling and compensatory pathways<br />

was measured by Western blot, immunohistochemistry (IHC) and<br />

reverse phase protein analysis (RPPA) of control and treated cell<br />

lysates/tumors.<br />

Results: Using a sensitivity cut-off of an IC 50 of < 1 µmol/L, 16 of<br />

the 18 HER2-amplified breast cancer cell lines and 8 of 10 cell lines<br />

with activating mutations in PIK3CA were sensitive to the pan-<br />

PI3K inhibitor BKM120. BEZ235 showed the most potent efficacy<br />

across the panel with IC 50 s < 100 nmol/L for each of the 49 cell<br />

lines tested. The HER2-amplified/PIK3CA mutant cell lines were<br />

also unexpectedly sensitive to the mTORC1 inhibitor RAD001, this<br />

was despite the silencing of mTORC1 signaling being followed by a<br />

feedback increase in phospho-AKT signaling. Furthermore, each of<br />

these molecules showed remarkable in vivo activity across the panel<br />

of xenografts models. BKM120, RAD001 and BEZ235 induced both<br />

tumor stabilization and regression independent of PTEN, PI3K, ER<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

380s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 4<br />

and HER2 status. Pharmacodynamic analysis of tumor tissue revealed<br />

that BEZ235 and RAD001 both inhibited mTORC1 signaling as<br />

indicated by a reduction in the levels of phosphorylated ribosomal<br />

protein S6 (pS6). However, in contrast to BKM120 and BEZ235,<br />

RAD001 did not induce a reduction in the levels of pAKT (S473<br />

or TH308) yet showed comparable in vivo efficacy to each of these<br />

molecules. Finally, combined targeting of HER2 and PI3K/mTOR in<br />

vitro increased the anti-proliferative activity of the molecules and led<br />

to an increased induction of apoptosis. The efficacy of these molecules<br />

(alone or in combination with trastuzumab) was assessed in a model of<br />

in vivo trastuzumab resistance generated through long-term treatment<br />

of the trastuzumab sensitive BT474 cells (BT-TR). All PI3Ki induced<br />

complete inhibition of tumor proliferation in monotherapy, while the<br />

combination of trastuzumab and each of these molecules induced<br />

tumor regression in the trastuzumab resistant tumors.<br />

Discussion: These pre-clinical data indicate that targeting the PI3K/<br />

mTOR pathway either alone or in combination with trastuzumab is<br />

effective strategy for overcoming trastuzumab resistance.<br />

P4-08-02<br />

Reactivation of oncogenic signaling through mTOR inhibitorsinduced<br />

feedback adaptations.<br />

Rodrik-Outmezguine V, Chandarlapaty S, Poulikakos P, Scaltriti M,<br />

Baselga J, Rosen N. MSKCC, New York, NY; MGH, Charlestown, MA<br />

mTORC1 inhibitors (or rapalogs) relieve feedback inhibition of<br />

receptor tyrosine kinases (RTKs) and cause mTORC2-dependent<br />

phosphorylation of AKT S473 and activation of AKT kinase and<br />

signaling. The relief of feedback inhibition of mTORC2/AKT<br />

signaling has been shown to abolish the antitumor activity of rapalogs<br />

and has often been proposed as a potential mechanism of resistance<br />

to these compounds.<br />

mTOR kinase inhibitors that inhibit both mTORC1 and mTORC2<br />

complexes have now been developed to circumvent these problems.<br />

mTOR kinase inhibitors block mTORC1 and mTORC2 and thus, do<br />

not cause the mTORC2 activation of AKT observed with rapamycin.<br />

We now show, however, that these novel classes of drugs have a<br />

biphasic effect on AKT. Inhibition of mTORC2 leads to AKT S473<br />

dephosphorylation and a rapid but transient inhibition of AKT T308<br />

phosphorylation and AKT signaling. However, inhibition of mTOR<br />

kinase, as observed with rapalogs, also relieves feedback inhibition of<br />

RTKs leading to subsequent PI3K activation and rephosphorylation of<br />

AKT but only at T308, which is sufficient to reactivate AKT activity<br />

and signaling. Thus, catalytic inhibition of mTOR kinase leads to a<br />

new steady state characterized by profound inhibition of mTORC1 and<br />

accumulation of activated AKT phosphorylated on T308 but not S473.<br />

We now confirm that the reactivation of AKT signaling is a mechanism<br />

of acquired resistance to rapalogs. We observe an induction of<br />

phospho-RTKs in rapamycin-resistant cells leading to both AKT<br />

and ERK signaling pathway activation enhancing cell survival.<br />

Combined inhibition of mTOR and the induced RTKs can prevent<br />

AKT reactivation and fully abolishes AKT signaling. These results<br />

reveal the adaptive capabilities of oncogenic signaling networks,<br />

highlighting the possible need for combinatorial approaches to block<br />

feedback-regulated pathways.<br />

P4-08-03<br />

The impact of the heregulin-HER receptor signaling axis on<br />

response to HER tyrosine kinase inhibitors.<br />

Gwin WR, Liu L, Zhao S, Xia W, Spector NL. Duke University,<br />

Durham, NC<br />

Background: HER receptor tyrosine kinases (EGFR/HER1; HER2;<br />

HER3; HER4) and their soluble ligands represent a robust system<br />

involved in the regulation of a diverse array of cellular processes.<br />

Deregulation of HER receptors, notably HER2, has been linked<br />

to the initiation and progression of breast cancer and other solid<br />

tumors. Relatively less is known about the role of HER receptor<br />

soluble ligands in tumorigenesis and responsiveness to HER targeted<br />

therapies. Here we will discuss the impact of the HER3 ligand<br />

heregulin (HRG) on the sensitivity of breast cancers to HER tyrosine<br />

kinase inhibitors (TKIs), and identify TKI strategies to treat HRGdriven<br />

breast cancers. Methods: The effects of exogenous HRG<br />

(50ng/ml) on the antitumor activity of a panel of TKIs with different<br />

enzymatic properties e.g. a reversible, selective inhibitor (lapatinib)<br />

and irreversible, pan-HER inhibitors (neratinib; CI-1033) were<br />

assessed in HER2+ breast cancer (BC) cell lines. The concentration<br />

of TKIs used was in the 1-2.5 uM range, and cells were treated over<br />

a 72 hr course. Vehicle treatment alone served as controls. The impact<br />

of the above mentioned treatments on total HER receptor and specific<br />

EGFR, HER2, and HER3 phosphotyrosine sites, in addition to the<br />

phosphorylation state of components of downstream MAPK and PI3K<br />

signaling pathways was analyzed through western blot. In addition<br />

to studying the effects of exogenous HRG, the impact of TKIs on a<br />

model of triple negative BC (TNBC) known to produce HRG in an<br />

autocrine manner was also evaluated. QRT-PCR was used to assess<br />

the effects of TKIs on HER receptor mRNA levels. Results: Lapatinib<br />

inhibited proliferation and phosphorylation of EGFR, HER2, HER3,<br />

and downstream MAPK and PI3K signaling pathways in HER2+<br />

SKBR3 and BT474 cell lines. Pre-treatment with HRG abrogated the<br />

antitumor effects of a therapeutic concentration of lapatinib (1 uM),<br />

and reversed the inhibitory effects of lapatinib on the phosphorylation<br />

of HER receptors and components of their downstream signaling<br />

pathways e.g. Erk1/2 and Akt. Neratinib also blocked proliferation<br />

and phosphorylation in HER2+ BC cells. In contrast to lapatinib,<br />

the antitumor effects of neratinib were not reversed by exogenous<br />

HRG. Interestingly, treatment with neratinib and similar pan-HER<br />

irreversible TKIs, but not reversible TKIs, resulted in loss of HER2<br />

and EGFR protein expression. QRT-PCR was used to evaluate if<br />

this effect was at the level of transcription. Furthermore, neratinib<br />

inhibited cell proliferation and blocked EGFR signaling in a TNBC<br />

model driven by autocrine produced HRG. Conclusions: Our findings<br />

suggest that HRG is a mediator of therapeutic resistance to lapatinib in<br />

HER2+ and TNBC. Neratinib demonstrates profound effects on HER<br />

signaling by markedly reducing HER2 and EGFR protein expression<br />

and blocking the pro-tumorigenic effects of autocrine or paracrine<br />

expression of HRG. In contrast to HRG, we previously showed that<br />

EGF, an EGFR specific ligand did not reverse the antitumor effects of<br />

lapatinib in breast cancer cells. Thus, the selection of HER targeted<br />

therapies in a given tumor should take into account not only the<br />

HER receptor expression profile, but also the presence of autocrine<br />

or paracrine derived ligands activating HER receptors.<br />

www.aacrjournals.org 381s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P4-08-04<br />

Abcc10 status affects proliferation, metastases and tumor<br />

sensitivity<br />

Domanitskaya N, Paulose C, Jacobs J, Foster K, Hopper-Borge E.<br />

Fox Chase <strong>Cancer</strong> Center, Philadelphia, PA<br />

Background: It has been proposed that therapy-induced overexpression<br />

of ATP-binding cassette (ABC) drug efflux pumps promotes resistance<br />

in many drug-treated cancers. While early studies focused on<br />

overexpression and activity of the P-glycoprotein (Pgp) pump,<br />

a structurally distinct group of ABC efflux pumps, known as the<br />

Multidrug Resistance Proteins (MRPs;ABCCs), have been gaining<br />

increasing attention as alternative sources of resistance. There are 9<br />

ABCC family members (ABCC1-6, 10-12), and we have previously<br />

shown that ABCC10 is unique because it confers resistance to a wide<br />

variety of anticancer agents including taxanes, vinca alkaloids and<br />

nucleoside-based analogs (Hopper-Borge et al, 2004, Hopper-Borge<br />

et al, 2009). Importantly we have shown that Abcc10 null mice are<br />

sensitized to taxanes (Hopper-Borge et al, <strong>Cancer</strong> Research, 2011).<br />

This is the first transporter whose loss results in in vivo tissue<br />

sensitivity to taxanes.<br />

Methods & Results: We recently bred a PyVmT mammary tumor<br />

model to our Abcc10 gene disrupted mice to investigate Abcc10’s<br />

in vivo activities regarding breast cancer resistance mechanisms.<br />

We have observed that Abcc10+/+ (WT) status limits the rate of<br />

tumorigenesis (p=0.020) and that WT tumors are more metastatic<br />

compared to Abcc10-/- (KO), (p=0.0489). To better understand the<br />

potential resistance mechanisms in this model we determined the<br />

expression levels of relevant transporters with qRT-PCR and found no<br />

significant differences between WT and KO. The PI3K-AKT-mTOR,<br />

a major signaling pathway involved in breast cancer progression has<br />

been connected to ABCCs (Tazzari, et al, 2007), and we observed<br />

upregulation of AKT in KO versus WT mammary tumor cell lines via<br />

western analyses. Therefore, we decided to ask if Abcc10 status affects<br />

proliferation, attachment, and/or migration (CyQuant, Invitrogen and<br />

xCELLigence, Roche). We showed that WT cell lines grow 3 times<br />

slower compared to KO. For migration kinetic we observed that KO<br />

cell lines were more active compared to WT.<br />

To determine the impact of Abcc10 loss in vivo we injected SCID<br />

mice with 3 WT or KO mammary tumor derived cell lines and<br />

treated tumors with docetaxel or vehicle. Tumors derived from WT<br />

and KO mice were both responsive to docetaxel (WT, p=0.003 and<br />

KO, p=0.001). However, over 21 days, the WT tumors were reduced<br />

by 50% compared to KO reduction of 86%. Kaplan-Meier survival<br />

analysis of treated/untreated SCID mice reveals that Abcc10 status is<br />

related to a poorer prognosis (KO p


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 4<br />

P4-08-06<br />

Notch-dependent Regulation of Novel Genes Associated with<br />

Trastuzumab Resistance<br />

Osipo C, Baumgartner A, Zlobin A, O’Toole M. Loyola University<br />

Chicago, Maywood, IL<br />

Background: We have shown previously that Notch-1 is critical for the<br />

development and maintenance of trasuzumab or lapatinib resistance<br />

in HER2 positive breast cancer cell lines. Here we sought to identify<br />

the critical genes that are regulated by Notch to promote anti-HER2<br />

targeted resistance.<br />

Methods: We measured expression of 84 known breast cancerassociated<br />

transcripts using a real-time PCR array in both trastuzumab<br />

sensitive and resistant BT474 cell lines. A 4 fold increase or decrease<br />

in transcripts levels were measured to be statisitically significant.<br />

To confirm the change in levels, we designed primers to each<br />

gene of interest and repeated the PCR at least three independent<br />

times. Furthermore, we specifically asked whether Notch-1 or<br />

it’s transcriptional mediator, CBF-1, directly regulated any of the<br />

identified genes in HER2 positive breast cancer cells using a genetic<br />

knocked down approach. An ANOVA for multiple comparisons was<br />

used to compute statistical significance.<br />

Results: We identified more than 4 fold increase in APC, ABCG2,<br />

ABCB1, Gata-3, Bcl-2, Id2, and HRG-2 transcripts in trastuzumab<br />

resistant versus sensitive cells. Conversley, Gli1 was downregulated<br />

in resistant versus sensitive cells. Interestingly, siRNA directed against<br />

Notch-1 or CBF-1 decreased ABCG2 and Gata-3, but conversely<br />

increased ABCB1 and HRG-2 in resistant cells.<br />

Conclusions: Our findings demonstrate APC, ABCG2, ABCB1,<br />

Gata-3, Bcl-2, Id2, and HRG-2 transcripts are signficantly increased<br />

in trastuzumab resistant breast cancer cells compared to sensitive<br />

cells. More importantly, Notch-1 or it’s direct transcriptional activator<br />

CBF-1 may contribute to traztuzumab resistance by directly regulating<br />

critical genes that contribute to multidrug resistance: ABCB1 and<br />

ABCG2 and/or genes associated with stem cell survival: Gata-3<br />

and HRG-2.<br />

P4-08-07<br />

Novel insight into the tumor “flare” phenomenon and lapatinib<br />

resistance<br />

Piede JA, Zhao S, Liu L, Lyerly HK, Osada T, Wang T, Xia W, Spector<br />

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

BACKGROUND: Resistance to lapatinib generally develops in<br />

approximately half of patients within one year of initiating treatment.<br />

When lapatinib is withdrawn, there is often a rapid progression of<br />

disease which is associated with high mortality rates. This “flare”<br />

phenomenon has also been observed upon discontinuation of other<br />

tyrosine kinase inhibitors (TKIs) in lung cancer, renal cell carcinoma,<br />

and gastrointestinal stromal tumors.<br />

METHODS: Using the HER2+ cell lines SKBR3 and BT474, we<br />

developed both in vitro and in vivo models demonstrating lapatinib<br />

resistance and the tumor “flare” phenomenon. Parental HER2+ cell<br />

lines were gradually exposed to increasing doses of lapatinib (100nM<br />

to 5uM) to develop a lapatinib-resistant form which was ultimately<br />

maintained in 1uM of lapatinib. Lapatinib-”released” cell lines were<br />

developed by removing lapatinib exposure from resistant cell lines<br />

and allowing the cells to grow for at least two weeks. Cell lines were<br />

grown in vitro using traditional monolayer cell culturing techniques<br />

and mammosphere technology. A comprehensive analysis of parental,<br />

lapatinib-resistant, and lapatinib-released cell lines was performed<br />

using microscopy, protein/phosphoprotein analysis, cell cycle, cancer<br />

stem cell markers by FACS, and invasion assays. Parental cells served<br />

as the control in all experiments. In vivo models were performed by<br />

injecting 10,000 cells of parental, resistant, and released cell lines in<br />

the mammary fat pads of SCID mice. Tumors were surgically resected<br />

after approximately sixty days and volume recorded. All experiments<br />

were repeated three times and calculated for statistical significance.<br />

RESULTS: Cell cycle analysis and proliferation assays demonstrate<br />

that lapatinib-resistant and released cell lines continue to proliferate<br />

despite the addition of 1uM lapatinib. Released cells also demonstrate<br />

less of a response to retreatment with lapatinib. Tumor volume of<br />

lapatinib-released cell lines were significantly larger than parental<br />

and resistant counterparts [parental: 39.9mm 3 , SD 9.48; resistant:<br />

71.8mm 3 , SD 62.33; released: 943.4mm 3 , SD 100.1] and this<br />

difference was statistically significant (p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

their proliferation and arrested them at the G1 phase, by inactivating<br />

the RB/E2F pathway, decreasing the number of new origins and<br />

eventually reducing the polyploidy population.<br />

Cell survival assays after different chemotherapeutic drug treatment<br />

showed that overexpression of the exogenous HOXC10 in MCF10A<br />

led to less susceptibility to most drugs. This was partially due to a<br />

protection from apoptosis by upregulating and activating the antiapoptotic<br />

machinery such as the NF-kb pathway. Further investigation<br />

revealed the involvement of HOXC10 in DNA repair (and not initial<br />

response), especially after DNA crosslink damage. Interestingly, the<br />

binding of HOXC10 to CDK7 in a region outside its homeodomain<br />

activates CDK7 activity towards RNA polymerase II mainly in<br />

response to DNA damage. Since HOX genes are difficult to target<br />

therapeutically, one potential approach to overcome chemoresistance<br />

in HOXC10 overexpressing cells is by including CDK7 inhibitors<br />

(already in clinical trials).<br />

All these results were confirmed in the SUM159 model which stably<br />

expresses a HOXC10-shRNA.<br />

Finally, HOXC10 was found to be significantly overexpressed in<br />

MCF7 isogenic cell lines gradually selected to be resistant to some<br />

chemotherapeutic drugs. By knocking down HOXC10 in these<br />

sublines, resistance to the drug was reduced. Further, SUM159 and<br />

MDAMD231 xenografts that were treated with chemotherapy over<br />

weeks and that show partial to no response tend to have a higher<br />

expression of HOXC10.<br />

Conclusion: This study shows that HOXC10, a homeobox protein<br />

previously shown to be regulated during the cell cycle and to have a<br />

positive effect on proliferation, is overexpressed in the majority of<br />

breast cancers. This upregulation may have clinical implications since<br />

cells with higher expression of HOXC10 tend to have more genomic<br />

instability and activation of anti-apoptotic and DNA repair pathways,<br />

which eventually modulate the response to some chemotherapy drugs.<br />

P4-08-09<br />

Targeting Thyroid Receptor b in Estrogen Receptor Negative<br />

<strong>Breast</strong> <strong>Cancer</strong><br />

Gu G, Covington K, Rechoum Y, O’Malley B, Mangelsdorf D, Minna<br />

J, Webb P, Fuqua S. Baylor College of Medicine, Houston, TX; UT<br />

Southwestern Medical Center; The Methodist Hospital Research<br />

Institute<br />

Background:<br />

The treatment of estrogen receptor (ER)-negative breast cancer (BC)<br />

is a major clinical problem due to the lack of useful therapeutic targets.<br />

Nuclear receptors (NRs) are potential targets in these patients because<br />

they regulate global transcriptional events and many already have<br />

agonists/antagonists available.<br />

Material and Methods:<br />

We used microarray analysis of 227 ER-negative tumors to identify<br />

NR targets, and performed hierarchical clustering using 41 NRs.<br />

Expressed receptors were scored using prediction analysis of<br />

microarrays (PAM) across clustered groups. Cell lines were matched<br />

to subtypes using previously described data (Neve et al. 2006).<br />

Candidate gene expression levels were confirmed by qRT-PCR<br />

using TaqMan probes. pGIPZ lentiviral vectors encoding shRNA<br />

were used to knockdownselected candidates. MTT and soft agar<br />

assays were used to measure chemosensitivity and growth following<br />

treatment with Docetaxel (Doc), Doxorubicin (Dox), or Cisplatin<br />

(Cis). Statistical analysis was performed using Red-R.<br />

Results:<br />

The 41 NRs clustered tumors into 5 groups. For each group we<br />

selected genes representing the highest ranked discriminators, and<br />

examined their effects in cell lines matching each groups’ gene<br />

signature. Thyroid hormone receptor b (THRβ) was selected from<br />

group V. The expression levels of this receptor were confirmed by<br />

qRT-PCR and Western blot analysis.<br />

Knockdown of THRβ in ER-negative HCC2185 cells rendered cells<br />

more resistant to all chemotherapeutics by using MTT assay. Similar<br />

results were confirmed in ER-negative MDA-MB-453 and HCC202<br />

cells. Knockdown of THRβ enhanced colony forming potential<br />

in anchorage-independent soft agar assays in MDA-MB-453 and<br />

HCC202 cells. Statistical analysis using clinical data from Sabatier et<br />

al. (BCRT 2011) showed that patients with low THRb have a worse<br />

clinical outcome. In order to translate these findings into the clinic,<br />

we treated cells with a specific THRβ agonists, GC-1 and KB-141.<br />

GC-1 inhibited cell growth in growth assays, and synergistic effects<br />

were observed when cells were treated with GC-1 and Docetaxel in<br />

combination. Re-expression of ERα protein was observed in ERnagative<br />

cells lines after treatment with GC-1 and KB141, suggesting<br />

that modulation of THRβ may also extend hormonal therapy to this<br />

hormonally insensitive group of patients.<br />

Conclusion:<br />

Clinical targeting of NRs in ER-negative BCs is a novel strategy since<br />

receptors can be specifically targeted with ligands. Our data suggest<br />

that chemotherapy response in ER-negative patients overexpressing<br />

THRβ could be enhanced with a THRβ agonist. Similarly, functional<br />

re-activation of ERα by activating THRβ might extend hormonal<br />

therapies to these patients as well.<br />

P4-08-10<br />

Systematic expression analysis of the genes related to drugresistance<br />

in isogenic docetaxel- and adriamycin-resistance breast<br />

cancer cell lines.<br />

Tang J, Li W, Zhong S, Xu J, Zhao J. Jiangsu <strong>Cancer</strong> Hospital,<br />

Nanjing, Jiangsu, China; Jiangsu <strong>Cancer</strong> Hospital, Nnajing, Jiangsu,<br />

China<br />

Background: Docetaxel (Doc) and Adriamycin (Adr) are two of the<br />

most effective chemotherapeutic agents in the treatment of breast<br />

cancer. However, their efficacy is often limited by the emergence<br />

of multidrug resistance (MDR). The purpose of this study was to<br />

investigate MDR mechanisms through analyzing systematically the<br />

expression changes of genes related to MDR in the induction process<br />

of isogenic drug resistant MCF-7 cell lines.<br />

Material and Methods: Isogenic resistant sublines selected at 100<br />

and 200nM docetaxel (MCF-7/100nM Doc and MCF-7/200nM<br />

Doc) or at 500 and 1500 nM adriamycin (MCF-7/500nM Adr and<br />

MCF-7/1500nM) were developed from human breast cancer parental<br />

cell line MCF-7/S, by exposing MCF-7/S to gradually increasing<br />

concentrations of docetaxel or adriamycin in vitro. Flow cytometry,<br />

MTT cytotoxicity assay and cell growth curves were preformed to<br />

demonstrate the MDR characteristics developed in sublines. Some<br />

key genes on the pathways related to drug resistance (including<br />

drug-transporters: MDR1, MRP1 and BCRP; drug metabolizingenzymes:<br />

CYP3A4 and GST pi; target gene: Topo IIα and β-tubulin<br />

III; apoptosis genes: Bcl-2 and Bax) were analyzed at RNA and protein<br />

expression levels by real time RT-qPCR and western blot, respectively.<br />

Results: MCF-7/100nM Doc and MCF-7/200nM Doc was 22- and<br />

37-fold resistant to docetaxel, and 18- and 32-fold to adriamycin,<br />

respectively. MCF-7/500nM Adr and MCF-7/1500nM Adr was 61-<br />

and 274-fold resistant to adriamycin, and 3- and 12-fold to docetaxel,<br />

respectively. Meantime, they also showed cross-resistance to the other<br />

anticancer drugs in different degree. Compared to sensitive MCF-<br />

7/S, RT-qPCR and Western blot results revealed that the expression<br />

of MDR1, MRP1, BCRP, Bcl-2 were elevated in both MCF-7/<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

384s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 4<br />

Doc and MCF-7/Adr, and Topo IIα, Bax were down-regulated in<br />

both the sublines, while CYP3A4 and β-tubulin III were increased<br />

only in MCF-7/Doc and GST pi was increased only in MCF-7/<br />

Adr. Furthermore, these changes above were dose-dependent, i.e.<br />

acquirement of MDR is dependent on docetaxel or adriamycin<br />

concentrations in breast carcer model resistant MCF-7 cells.<br />

Conclusions: The established MCF-7/Doc or MCF-7/Adr has the<br />

typical MDR characteristics, which can be used as the models<br />

for resistance mechanism study. The acquired process of MCF-<br />

7/S resistance to docetaxel or adriamycin is gradual, and is also<br />

complicated with the various pathways involved in. There are some<br />

common mechanisms as well as drug-special mechanisms between<br />

MCF-7/Doc and MCF-7/Adr.<br />

P4-09-01<br />

Retrospective evaluation of precision of gene-expression-based<br />

signatures of prognosis and tumor biology in replicate surgical<br />

biospecimens from patients with breast cancer<br />

Barry WT, Marcom PK, Geradts J, Datto MB. Duke University<br />

Medical Center, Durham, NC<br />

Background: Numerous gene-expression signatures have been<br />

developed for breast cancer. However, assessments of their validity<br />

continue to largely ignore the impact of intratumor heterogeneity and<br />

technical variation on clinical utility. Here, the collection of replicate<br />

specimen in Barry et al (2010) is used to evaluate a broad collection<br />

of gene-expression signatures of prognosis and tumor biology.<br />

Methods: Eighteen patients with multiple frozen cores (one patient<br />

with quadruplicate, twelve with triplicate, and five with doublet<br />

samples) were previously identified in the Duke <strong>Breast</strong> SPORE<br />

tissue repository. Cores were assessed for percent invasive cancer<br />

cellularity, and tumor size, grade, and ER/PR status. RNA was<br />

extracted and hybridized to AffymetrixH133Plus2.0 microarrays.<br />

Expression signatures of prognosis and tumor biology were developed<br />

or translated to the Affymetrix platform using routines by Prat et al.<br />

(2012) and Haibe-Keins et al. (2012). Association between signatures,<br />

and precision among replicates are evaluated using Pearson and<br />

intraclass correlation. Coefficients are reported with 95% confidence<br />

intervals.<br />

Results: Among prognostic signatures, an imputed 70-gene signature<br />

of MammaPrint had the highest level of precision (ICC=0.96, 0.91-<br />

0.98); strong concordance is seen with Affymetrix-based PAM50<br />

risk-of-relapse (ICC=0.82, 0.65-0.92); 16 of 18 patients had constant<br />

subtype calls. Substantially lower concordance was seen in the<br />

GENIUS prognostic model (ICC=0.62, 0.35-0.82). In ER+ patients<br />

(n=13), two algorithms to impute the 21-gene model of OncotypeDX<br />

recurrence score were concordant (r =0.98, ICC= 0.90 and 0.83) and<br />

distinct from the GENIUS ER+ score (average r=0.74, ICC=0.78).<br />

Imputed models for the Rotterdam 76-gene model (+ER status),<br />

GGI (+grade), and ROR-T (+tumor size) showed lower levels of<br />

correlation (0.47


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

develop prognostic signatures for other cancer types, and are using<br />

similar multiplexed algorithms to develop gene signatures able to<br />

predict response to neoadjuvant therapy.<br />

P4-09-03<br />

Withdrawn by Author<br />

P4-09-04<br />

Gene expression profile that predict outcome of Tamoxifentreated<br />

estrogen receptor-positive, high-risk, primary breast<br />

cancer patients: a DBCG study.<br />

Lyng MB, Lænkholm A-V, Tan Q, Vach W, Gravgaard KH, Knoop<br />

A, Ditzel HJ. University of Southern Denmark, Odense, Denmark;<br />

Slagelse Hospital, Slagelse, Denmark; Odense University Hospital,<br />

Odense, Denmark; University Medical Center Freiburg, Germany<br />

Background<br />

Tamoxifen significantly improves outcome for estrogen receptorpositive<br />

(ER+) breast cancer, but the 15-year recurrence rate remains<br />

30%. The aim of this study was to identify gene profiles that accurately<br />

predicted the outcome of ER+ breast cancer patients who received<br />

adjuvant Tamoxifen mono-therapy.<br />

Methods<br />

Post-menopausal breast cancer patients diagnosed no later than 2002,<br />

being ER+ as defined by >1% IHC staining and having a frozen tumor<br />

sample with >50% tumor content were included. Tumor samples<br />

from 108 patients treated with adjuvant Tamoxifen were analyzed for<br />

the expression of 59 genes using quantitative-PCR. End-point was<br />

clinically verified recurrence to distant organs or ipsilateral breast.<br />

Gene profiles were identified using a model building procedure based<br />

on conditional logistic regression and leave-one-out cross-validation,<br />

followed by a non-parametric boostrap (1000x re-sampling). The<br />

optimal profiles were further examined in 5 previously-reported<br />

datasets containing similar patient populations that were either treated<br />

with Tamoxifen or left untreated (N=623).<br />

Results<br />

Three gene profiles were identified, the strongest being a 2-gene<br />

combination of BCL2-CDKN1A exhibiting an accuracy of 75% for<br />

prediction of outcome. Independent examination using 4 previouslyreported<br />

microarray datasets of Tamoxifen-treated patient samples<br />

(N=503) confirmed the potential of BCL2-CDKN1A (average<br />

accuracy: 68%, range: 53%-85%). The predictive value was<br />

further determined by comparing the ability of the genes to predict<br />

recurrence in an additional, previously-published, cohort consisting<br />

of Tamoxifen-treated (n=58, p=0.015) and untreated patients (n=62,<br />

p=0.25).<br />

Conclusion<br />

A novel gene expression profile predictive of outcome of Tamoxifentreated<br />

patients was identified. The validation suggests that BCL2-<br />

CDKN1A exhibit strong predictive potential.<br />

P4-09-05<br />

Microarray anlyses of breast cancers identify CH25H, a<br />

cholesterol gene, as a potential marker and target for late<br />

metastatic reccurences.<br />

Saghatchian M, Mittempergher L, Michiels S, Wolf D, Canisius<br />

SV, Dessen P, Delaloge S, Lazar V, Benz SC, Roepman P, Glas AM,<br />

Tursz T, Bernards R, van’t Veer LJ. The Netherlands <strong>Cancer</strong> Institute,<br />

Amsterdam, Netherlands; Institut Jules Bordet, Brussels, Belgium;<br />

UCSF Helen Diller Family Comprehensive <strong>Cancer</strong> Center, <strong>San</strong><br />

Francisco; Institut Gustave Roussy, Villejuif, France; University of<br />

California, <strong>San</strong>ta Cruz; Agendia, Amsterdam, Netherlands<br />

Background<br />

However hormone receptor–positive, early-stage breast cancer is<br />

a disease with a long natural history and improved survival with<br />

5-year adjuvant endocrine treatments. Yet late recurrence remains<br />

an important issue in adjuvant therapy. Predictors of late recurrence<br />

are not yet well characterized.<br />

Method<br />

A total of 252 breast primary tumors were selected at the Netherlands<br />

<strong>Cancer</strong> Institute from retrospective series of ER+, HER2- breast<br />

cancer patients with a follow-up of at least 10 years. Gene expression<br />

analysis was performed using Agilent 4x44K microarrays. In order<br />

to identify genes associated to late survival differences, we used the<br />

survdiff function implemented in the R package survival and we set<br />

the parameter rho to -1 to give weight to the later part of the survival<br />

curve. The survdiff function was applied to each probe individually<br />

for DMFS time considering the probe as a covariate dichotomized<br />

into 2 groups (above and below the median expression across all<br />

samples). The parameter “strata” was used to stratify the 140 patients<br />

based on additional clinico-pathological parameters (Grade, Diameter,<br />

Lymph node status and MammaPrint), in order to find genes that add<br />

prognostic value to those parameters already known.This approach<br />

uses the distant-metastasis free survival (DMFS) time as a continuous<br />

variable.<br />

Results<br />

After univariate analysis, MammaPrint, diameter, lymph node status<br />

and grade were significantly associated to late DMFS differences<br />

(Chi-square test p-values equal to 0.016, 0.004,


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 4<br />

gene a potential target for late metastasis control in breast cancer.<br />

These results warrant further prospective investigation and functional<br />

characterization of CH25H in this setting.<br />

P4-09-06<br />

miR-187 is an independent prognostic factor in lymph nodepositive<br />

breast cancer patients.<br />

O’Connor DP, Mulrane L, Brennan DJ, Madden S, Gremel G, McGee<br />

SF, McNally S, Martin FM, Crown JP, Jirstrom K, Higgins DG,<br />

Gallagher W. UCD Conway Institute, Dublin, Ireland; Molecular<br />

Therapeutics for <strong>Cancer</strong> Ireland, Dublin City University, Dublin,<br />

Ireland; St Vincent’s University Hospital, Dublin, Ireland; Lund<br />

University, Lund, Sweden<br />

Purpose<br />

MicroRNAs (miRNAs) involved in cancer progression have now<br />

become the focus of much attention as they represent a new class of<br />

biomarkers and potential drug targets. Here, we describe an integrated<br />

bioinformatics, functional analysis and translational pathology<br />

approach for the identification of novel miRNAs involved in breast<br />

cancer progression.<br />

Experimental Design<br />

Differential gene expression can, in part, be attributed to the activity<br />

of specific miRNAs. Given a database of miRNA binding site<br />

motifs and gene expression levels determined by transcriptomic<br />

profiling, correspondence analysis, between group analysis and<br />

co-inertia analysis can be combined to produce a ranked list of<br />

miRNAs associated with a specific gene signature and phenotype.<br />

Here, using two independent breast cancer cohorts, this approach<br />

was employed to produce a ranked list of miRNAs associated with<br />

disease progression. Functional studies were subsequently carried out<br />

in MCF7 cells assessing for alterations in growth, tumorigenicity and<br />

agressiveness and miRNA expression was evaluated in two cohorts<br />

of breast cancer patients by locked nucleic acid in situ hybridisation<br />

on tissue microarrays.<br />

Results<br />

CIA identified miR-187 as a key miRNA associated with poor<br />

outcome in breast cancer.<br />

Ectopic expression of miR-187 in MCF7 cells resulted in a more<br />

aggressive phenotype (evidenced by increased anchorage-independent<br />

growth, migratory and invasive potential).<br />

In a test cohort (n=117) breast cancer patients, high expression of<br />

miR-187 was associated with a trend towards reduced breast cancerspecific<br />

survival (BCSS) (p=0.058), and a significant association<br />

with reduced BCSS in lymph node-positive patients (p=0.036). In a<br />

validation cohort (n=470), high miR-187 was significantly associated<br />

with reduced BCSS in the entire cohort (p=0.021) and, again, in lymph<br />

node-positive patients (p=0.012).<br />

Multivariate cox regression analysis revealed that miR-187 is an<br />

independent prognostic factor in both TMA cohorts (Cohort 1 HR-<br />

7.369 (95% CI 2.048-26.509, p=0.002); Cohort 2 HR-2.798 (95%<br />

CI 1.518-5.157, p=0.001).<br />

Conclusions<br />

miR-187 expression in breast cancer leads to the formation of a more<br />

aggressive, invasive phenotype and acts as an independent predictor<br />

of outcome.<br />

P4-09-07<br />

ING1 Expression Measured by AQUA can be an Independent<br />

Prognostic Marker in <strong>Breast</strong> <strong>Cancer</strong><br />

Thakur S, Klimowicz A, Pohorelic B, Dean M, Konno M, Bose P,<br />

Magliocco A, Riabowol K. University of Calgary, AB, Canada; Tom<br />

Baker <strong>Cancer</strong> Center, Calgary, AB, Canada<br />

Background:<br />

<strong>Breast</strong> cancer accounts for around 45,000 cancer related deaths in<br />

North America per year. The high incidence of breast cancer observed<br />

in this region is most likely due to the availability of various screening<br />

programs used to detect breast cancer, which otherwise may never get<br />

diagnosed. There is an inverse relation between the cost of treatment<br />

and patient survival as the breast cancer progresses to higher grades.<br />

Therefore, screening and diagnosing breast cancers at earlier stages<br />

is needed which can improve patient survival.<br />

The INhibitor of Growth (ING) family of tumour suppressor proteins<br />

are implicated in diverse cellular processes including chromatin<br />

remodelling and apoptosis. ING proteins are stoichiometric members<br />

of histone acetyl transferase (HAT) and histone deacetylase (HDAC)<br />

complexes. Their expression is downregulated in several cancers,<br />

including breast carcinoma. Alternatively, ING function may be<br />

compromised by mislocalization to the cytoplasm. However, the<br />

association of expression of ING proteins and cancer specific survival<br />

has not been studied yet. In this study, we aimed to evaluate the<br />

prognostic significance of ING1 in breast cancer.<br />

Patients and Methods:<br />

This study included 443 <strong>Breast</strong> <strong>Cancer</strong> patients diagnosed in Calgary,<br />

Canada from 1985-2000. Clinical data were obtained from the Alberta<br />

<strong>Cancer</strong> Registry and chart review. Tissue microarrays (TMAs)<br />

were assembled from triplicate cores of formalin-fixed paraffinembedded<br />

tumour tissue. ING1 protein expression was quantified<br />

using fluorescent immunohistochemistry and automated quantitative<br />

analysis (AQUA) in normal breast epithelial cells and breast cancer<br />

samples. 5 year progression free survival (PFS) was analyzed using<br />

Kaplan-Meier plots and Cox proportional hazard modelling.<br />

Results:<br />

In our study, we found that ING1 expression is significantly<br />

downregulated in the breast cancer patient samples relative to<br />

normal breast epithelia. High or low ING1 cytoplasmic/nuclear<br />

(C/N) ratio results in poor 5 year PFS. Also, the low tumor/stromal<br />

(t/s) ratio of ING1 results in poor prognosis. In univariate analysis,<br />

t/s ING1 expression showed a negative correlation with tumor stage<br />

[p=0.008]; tumor grade [p=0.000]; nodal status [p=0.000] and 5 year<br />

PFS [p=0.004]. However, no correlation was observed with ER, PR<br />

or HER2 status. In multivariate analysis, t/s ING1 proved to be an<br />

independent and better prognostic marker [HR 0.582, p=0.037] than<br />

HER2 [HR 2.230, p=0.057]; Age at diagnosis [HR 2.010, p=0.205]<br />

and tumor size [HR 1.471, p=0.189].<br />

Discussion and Conclusions:<br />

This is the first study to evaluate ING1 expression in breast cancer<br />

using the AQUA technique. ING1 expression high or low in the<br />

C/N ratio might disrupt the stoichiometry of chromatin-modifying<br />

complexes and hamper ING1s extra nuclear functions, leading to poor<br />

survival. The significantly improved survival observed in patients<br />

with high t/s ING1 expression further strengthens the role of ING1<br />

as a tumor suppressor.<br />

www.aacrjournals.org 387s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P4-09-08<br />

HOXB9, a gene promoting tumor angiogenesis and proliferation,<br />

is significantly associated with poor clincal outcomes in ERpositive<br />

breast cancer patients<br />

Seki H, Hayashida T, Jinno H, Takahashi M, Suzuki K, Kaneda M,<br />

Hara H, Osaku M, Asanuma F, Yamada Y, Mukai M, Kitagawa Y.<br />

Kitasato University Kitasato Institute Hospital, Tokyo, Japan; Keio<br />

University School of Medicine, Tokyo, Japan; Keio University School<br />

of Medicine, Japan<br />

Background: Studies have suggested that HOXB9 expression<br />

in breast cancer cells promotes cellular invasiveness, metastatic<br />

ability, and tumor neovascularization in the surrounding tissue<br />

in in vitro and in vivo assays. These findings imply that HOXB9<br />

overexpression may alter tumor-specific cell fates and the tumor<br />

stromal microenvironment, contributing to breast cancer progression<br />

(Hayashida et al., PNAS 2010). We previously reported that clinical<br />

outcomes were significantly decreased in HOXB9-positive patients<br />

(Seki et al., Ann Surg Oncol. 2012). In this study, it was demonstrated<br />

that HER2 type and basal-like breast cancers were more associated<br />

with HOXB9 positivity than the luminal type. Therefore, it is required<br />

to identify more detail clinicopathological features most likely to<br />

be associated with HOXB9 overexpression. We investigated the<br />

correlation between HOXB9 overexpression and clinicopathological<br />

variables, and clinical outcomes in estrogen receptor (ER)-positive<br />

breast cancer patients.<br />

Patients and methods: A consecutive series of 118 ER-positive breast<br />

cancer patients who underwent surgical treatment were examined.<br />

HOXB9 expression was analyzed immunohistochemically using the<br />

anti-human HOXB9 polyclonal antibody. Immunostaining of Ki-67<br />

and CD31 were also performed to evaluate tumor proliferation and<br />

angiogenesis.<br />

Results: The median age was 59 years and median observation<br />

period was 62 months. Of 118 tumor specimens immunostained for<br />

HOXB9, 49 specimens (41.5%) were positive staining. Univariate<br />

logistic regression revealed high nuclear grade (p


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 4<br />

P4-09-10<br />

Prospective Analysis of Fatty Acid Synthase (FASN) in <strong>Breast</strong><br />

<strong>Cancer</strong> Tissue of Early-Stage <strong>Breast</strong> <strong>Cancer</strong> Patients<br />

Puig T, Blancafort A, Casoliva G, Oliveras G, Casas M, Buxo M,<br />

Saiz E, Viñas G, Dorca J, Porta R. University of Girona and Girona<br />

Biomedical Research Institute (IDIBGi), Girona, Spain; Assistència<br />

<strong>San</strong>itària Institute, Girona, Spain; Catalan Institute of Oncology,<br />

Girona, Spain; Dr. Josep Trueta Hospital, Girona, Spain; Dr. Josep<br />

Trueta Hospital and Girona Biomedical Research Institute (IDIBGi),<br />

Girona, Spain; Health Inequalities Research Group - Employment<br />

Conditions Knowledge Network (GREDS-EMCONET), Universitat<br />

Pompeu Fabra, Barcelona, Spain; Dr. Josep Trueta Hospitaland<br />

Girona Biomedical Research Institute (IDIBGi), Girona, Spain<br />

Background: <strong>Cancer</strong> cells require nutrients to survive in the<br />

unfavorable microenvironment of primary solid tumors or metastases<br />

before angiogenesis development. Fatty acid synthase (FASN) is a<br />

multi-enzyme protein that catalyzes fatty acid synthesis. Expression<br />

levels of FASN are low or undetectable in normal human tissues<br />

except for the liver and the adipose tissue. In contrast, high levels<br />

of FASN expression have been detected in breast cancer tumors<br />

and other human carcinomas. Several reports highlight that FASN<br />

overexpression in tumor samples correlates with progression,<br />

aggressiveness and metastatic potential of the disease. In adition,<br />

some studies have suggested the same correlation with serum levels of<br />

FASN. Our aim was to analyze the association between the expression<br />

of tumor and serum levels of FASN with clinical and pathological<br />

prognostic factors in early-stage breast cancer patients.<br />

Methods: Fifty-five patients with early-stage breast cancer treated<br />

with surgery and post-operative chemotherapy were included in the<br />

study. We prospectively measured the levels of FASN in tumor and<br />

serum samples. Clinical data included demographic characteristics,<br />

menarche, pregnancy, breast feeding, menopausal status and body<br />

mass index (BMI). Pathological and molecular data included:<br />

pathological state, histological grade, estrogen and progesterone<br />

receptors, HER2 status, p53 mutation and Ki 67 levels. FASN tissue<br />

expression levels were determined by IHC and circulating FASN<br />

levels were determined by ELISA. FASN expression was graded from<br />

0 to 3+, meaning 0-1+ normal amounts of FASN protein compared<br />

to non-tumor breast tissue, 2+ moderate amounts and 3+ the highest<br />

levels of FASN expression. Baseline characteristics were summarized<br />

descriptively. Categorical variables were compared by c 2 or Fisher’s<br />

exact. For continuous variables, if the data are approximately normal,<br />

the two groups were compared using ANOVA. If the normality<br />

assumption is not warranted, then the Kruskall-Wallis test has been<br />

used.<br />

Results: Median age was 49 (rage 33-77). 51% of the patients were<br />

menopausal and median BMI was 24,75. Thirty-four percent of the<br />

patients had stage I, 51% stage II and 15% stage III. We observed a<br />

statistically significant association between FASN over expression<br />

and the lack of progesterone receptors (p=0.027) in tumor samples. In<br />

contrast, we found no relation between FASN and estrogen receptor<br />

nor between FASN and HER2 tumor expression in this setting.<br />

Menopause and age were strongly related to higher levels of FASN<br />

tumor expression (p< 0.001). Patients with higher BMI had higher<br />

levels of FASN in tumor tissue although this association was not<br />

statistically significant (p= 0.07). Finally, we observed a positive<br />

relation between breast cancer stage and the levels of FASN tumor<br />

(p=0.05). In contrast, circulating FASN levels were not associated<br />

with any pathological or clinical prognostic factor.<br />

Conclusions: Our study suggests that FASN overexpression is<br />

significantly related to age, menopausal status, more advanced stages<br />

and lack of progesterone receptor expression in early-stage breast<br />

cancer patients. However, no relation between serum levels of FASN<br />

and the clinical or molecular prognostic factors have been observed.<br />

P4-09-11<br />

Fatty Acid Synthase (FASN) expression in Triple-Negative <strong>Breast</strong><br />

<strong>Cancer</strong><br />

Viñas G, Oliveras G, Perez-Bueno F, Giro A, Blancafort A, Puig-Vives<br />

M, Marcos-Gragera R, Dorca J, Brunet J, Puig T. Catalan Institute<br />

of Oncology and Girona Biomedical Research Institute (IDIBGi),<br />

Girona, Spain; University of Girona and Biomedical Research<br />

Institute (IDIBGi), Girona, Spain; Dr. Josep Trueta Hospital and<br />

Catalan Institute of Health (ICS), Girona, Spain; Catalan Institute<br />

of Oncology, Girona, Spain; University of Girona, Spain<br />

Background:<br />

In approximately 15-20% of patients with breast cancer, the tumors<br />

do not express estrogen receptor (ER), progesterone receptor (PR)<br />

and do not have amplification of HER2. These tumors are called<br />

triple-negative breast cancer (TNBC) and patients with these tumors<br />

have a poor prognosis. There is no clinically validated, molecularly<br />

targeted therapy for TNBC patients and they can be treated only with<br />

chemotherapy. Thus, the identification of a novel targeted therapies for<br />

TNBC patients would be of great benefit. Fatty acid synthase (FASN)<br />

is a multi-enzyme protein that catalyzes fatty acid synthesis. Its main<br />

reaction is to synthesize palmitate from acetyl-CoA and malonyl-<br />

CoA, in the presence of NADPH, into long-chain saturated fatty<br />

acids. Expression levels of FASN are low or undetectable in normal<br />

human tissues except for the liver and the adipose tissue. In contrast,<br />

high levels of FASN expression have been detected in several human<br />

carcinomas. Several reports highlight that FASN overexpression<br />

in tumor samples correlates with progression, aggressiveness and<br />

metastatic potential of the disease. Previously, our group has shown<br />

that FASN activity inhibition induces apoptosis in several human<br />

cancer cells. The aim of our study was to evaluate the expression<br />

of tumor levels of FASN in triple-negative breast cancer patients.<br />

Methods:<br />

FASN tumor expression was retrospectively evaluated in 30<br />

paraffin-embedded core-biopsies of 30 patients with TNBC using<br />

the Fatty Acid Synthase polyclonal antibody (Assay design,<br />

Enzo Life Sciences, Exeter United Kingdom) and the detection<br />

kit EnVision (DAKO, Glostrup, Denmark). FASN expression<br />

levels were determined by immunohistochemistry (IHC) using the<br />

AutostainerPlus Link (DAKO). FASN expression was graded from 0<br />

to 3+, meaning 0-1+ normal amounts of FASN protein compared to<br />

non-tumor breast tissue, 2+ moderate amounts and 3+ highest levels<br />

of FASN expression. In vitro, we have determined the effect of the<br />

FASN-inhibitors, EGCG and G28UCM, on cell viability (measured by<br />

the MTT assay) and apoptosis [as assessed by cleavage of poly(ADPribose)<br />

polymerase (PARP)] of two TNBC cell lines, MDA-MB-231<br />

and MDA-MB-468.<br />

Results:<br />

From 30 patients with TNBC, 66.6% (20/30) of the patients had<br />

a high expression of FASN (FASN 3+) and 33.3% (10/30) had a<br />

moderate expression of FASN (FAS 2+). None of the tumors had lack<br />

of FASN expression analysed by immunohistochemistry. In vitro, the<br />

FASN-inhibitor G28UCM induced apoptosis and showed low IC 50<br />

values of citotoxicity in both, MDA-MB-231 and MDA-MB-468<br />

TNBC cell lines.<br />

www.aacrjournals.org 389s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Conclusions:<br />

FASN is overexpressed in triple-negative breast cancer tumors. The<br />

absence of target therapies for this breast cancer subtype and its poor<br />

prognosis lead to the exploration of FASN as therapeutic target for<br />

TNBC patients.<br />

P4-09-12<br />

TIMP-4 – Prognostic Marker and Treatment Target for Triple-<br />

Negative <strong>Breast</strong> <strong>Cancer</strong>s.<br />

Wallon UM, Sabol JL, Gilman PB, Zemba-Palko V, DuHadaway<br />

JB, Ciocca RM, Ali ZA, Carp NZ, Choy NS, Wojciechowski BS,<br />

Prendergast GC. Lankenau Medical Center, Wynnewood, PA<br />

BACKGROUND — Tissue inhibitor of metalloproteinase-4 (TIMP-<br />

4) is a secreted multi-functional protein associated with poor survival<br />

prognosis among early-stage triple-negative breast cancers (TNBC)1.<br />

Triple-negative breast cancers (TNBC) represent a highly aggressive<br />

form of this disease with few treatment options available. Current<br />

standard chemotherapy (CTX) includes taxane or adriamycin based<br />

regiments.<br />

Extracellular TIMP-4 binds to the membrane bound tetraspanin CD63<br />

and induces the activation of PI3K/AKT/mTOR survival pathway.<br />

Here we report that TIMP-4 induced aggressive tumor growth and<br />

metastasis can be adverted by targeting TIMP-4 either directly by<br />

sequestering extracellular pools of TIMP-4 or indirectly by blocking<br />

the activation of downstream survival pathways.<br />

METHODS — Prospectively collected patient samples, in accordance<br />

with the IRB approved protocol, were tested for circulating levels of<br />

TIMP-4 using a commercially available ELISA assay in samples from<br />

time of surgery and at each treatment cycle. The medical oncology<br />

staff recommended therapy without knowledge of TIMP-4 status.<br />

The role of elevated TIMP-4 in TNBC cell behavior was tested in cell<br />

culture and animal experiments using the human breast cancer line<br />

MDA-MB-468. Cells with or without TIMP-4 added to the medium<br />

were used to determine the effects on growth, clonogenic survival<br />

and response to chemotherapeutic agents such as adriamycin, Taxol,<br />

the new anti-TIMP-4 antibody and the PI3K/AKT/mTOR inhibitor<br />

GDC-0941. The same cell-line was used to induce tumor growth in<br />

nude mice with or without TIMP-4 containing slow-release pellets<br />

implanted into the mammary fatpad (mfp). Tumor growth and<br />

response to therapy was followed over a six-week period.<br />

RESULTS — Results from patient samples demonstrated that<br />

circulating TIMP-4 levels in breast cancer patients remain unaffected<br />

after surgical removal of the primary tumor. Adriamycin containing<br />

regiments was the only CTX to suppress the TIMP-4 levels<br />

independent of primary tumor size and nodal status.<br />

Adding TIMP-4 to cell culture medium or the mfp of mice resulted<br />

in an 1.5-fold increased tumor growth rate. Elevated TIMP-4 in mice<br />

also resulted in liver metastasis in 25% of animals (N=8).<br />

In cell cultures, the TIMP-4 induced effects were completely adverted<br />

by addition of GDC-0941. Adding the TIMP-4 antibody, to cell culture<br />

medium or i.p. injections to mice, resulted in a decelerated growth rate.<br />

CONCLUSIONS — On the basis of these clinical and experimental<br />

data we suggest that TIMP-4 may represent a simple prognostic and<br />

predictive marker for TNBC patients at highest risk. The presence of<br />

TIMP-4 identifies a patient population likely to recur quickly based on<br />

the continuous activation of the PI3K/AKT/mTOR pathway. Though<br />

adriamycin therapy can reduce the TIMP-4 levels, the toxicity of this<br />

agent suggests that targeted therapy of the PI3K/AKT pathway and/or<br />

a biological therapeutic approach directed against TIMP-4 may be of<br />

benefit in this subset of pts and should be further explored.<br />

a Liss, M et.al. Am. J. Pathol. 2009<br />

b Donover, P et.al. J. Cell. Biochem. 2010<br />

P4-10-01<br />

Edgetic perturbation of BRCT-mediated interactions caused by<br />

the BRCA1 H1686Q sequence variant<br />

De Nicolo A, Pathania S, Parisini E, Joukov V. Dana-Farber <strong>Cancer</strong><br />

Institute, Boston, MA; Harvard Medical School, Boston, MA; Italian<br />

Institute of Technology, Milan, Italy<br />

Background: The ever-increasing number of variants of uncertain<br />

significance (VUS) that result from BRCA1/BRCA2 genetic testing<br />

poses a challenge to healthcare providers, thus impacting clinical<br />

decision-making. Though a quantitative posterior probability model,<br />

mostly reliant on genetic data, has been developed to estimate the<br />

clinical relevance of individual VUS, indirect evidence derived from<br />

functional tests continues to be a sought-after adjunct to achieve<br />

VUS categorization. Using a multimodal approach, we previously<br />

demonstrated the deleterious effect on protein localization and<br />

function of the Italian founder sequence alteration BRCA1 V1688del,<br />

and the neutral effect of a neighboring variant, BRCA1 V1687I,<br />

identified in a patient with early onset triple negative breast cancer.<br />

Disease-causality has been suggested for the adjacent BRCA1<br />

H1686Q variant that occurs in the evolutionarily conserved THV<br />

motif of the first BRCT repeat and was described in a breast/ovarian<br />

cancer family (Giannini et al., 2008). We aimed at substantiating<br />

the predicted pathogenicity of BRCA1 H1686Q by assessing its<br />

biological significance.<br />

Methods: To investigate the structural and functional consequences<br />

of the BRCA1 H1686Q sequence alteration, we used a strategy that<br />

combined homology modeling with analysis of protein interactions<br />

(by immunoprecipitation and GST pull down techniques) and function<br />

(by a homologous recombination assay).<br />

Results: The three-dimensional model of the BRCA1 H1686Q<br />

BRCT domain suggested that the site and nature of the amino<br />

acid change could alter the interaction and relative orientation of<br />

secondary structure elements in the core region of the protein, thus<br />

possibly affecting the overall stability of the protein and triggering a<br />

long-range allosteric effect with repercussions on residues involved<br />

in phosphopeptide binding. Indeed, in transfected 293T cells, the<br />

recombinant BRCA1 H1686Q protein was less abundant than its<br />

wild-type counterpart. Moreover, while efficiently heterodimerizing,<br />

via its N-terminus, with the binding partner BARD1, the BRCA1<br />

H1686Q protein displayed a peculiar behavior with regards to<br />

C-terminal, BRCT-mediated interactions as it bound BRIP1/FANCJ<br />

and not CtIP. These results, repeatedly observed in different cell lines,<br />

were confirmed by reciprocal immunoprecipitations as well as by<br />

GST pull down experiments using a recombinant BRCA1 H1686Q<br />

BRCT domain. Notably, the BRCA1 H1686Q protein displayed a<br />

homologous recombination defect similar to that observed for the<br />

clinically ascertained BRCA1 M1775R mutant protein.<br />

Conclusions: Our data suggest that the BRCA1 H1686Q variant,<br />

by affecting the BRCT ligand recognition ability, causes an edgetic<br />

perturbation (i.e. selective disruption/retention) of BRCT-mediated<br />

interactions, which is yet sufficient to impair the homologous<br />

recombination function of the encoded protein. These findings provide<br />

valuable structure/function insights that could help elucidate the link<br />

between BRCA1 DNA repair and tumor suppression functions and aid<br />

development of functional tests for VUS screening and classification.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

390s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 4<br />

P4-10-02<br />

Targeted resequencing of BRCA1 and BRCA2 in inherited breast<br />

cancer<br />

Wong-Brown MW, Li S, Wilkins M, Avery-Kiejda KA, Bowden NA,<br />

Scott RJ. University of Newcastle, NSW, Australia; University of New<br />

South Wales, Sydney, NSW, Australia; Hunter Area Pathology Service<br />

(HAPS), Newcastle, NSW, Australia<br />

<strong>Breast</strong> cancer is the leading cause of cancer-related deaths in women<br />

worldwide, affecting about 13,000 women every year in Australia.<br />

Inherited loss-of-function mutations in BRCA1 and BRCA2<br />

predispose to high risk of breast and/or ovarian cancer. Since the<br />

discovery of breast cancer susceptibility genes BRCA1 and BRCA2<br />

two decades ago, there have not been any other genes identified that<br />

play a significant role in predisposition to inherited breast cancer.<br />

A large proportion of individuals with inherited breast cancer are<br />

negative for BRCA mutations and despite numerous research efforts,<br />

further breast cancer susceptibility genes still remain elusive.<br />

We hypothesize that genetic anomalies are present in the BRCA1 and<br />

BRCA2 genes in a subset of individuals with inherited breast cancer<br />

where no genetic anomalies where identified using traditional <strong>San</strong>ger<br />

sequencing. This study aims were to identify genetic anomalies in<br />

BRCA1 and BRCA2 by completely re-sequencing 200 kilobases<br />

surrounding BRCA1 and BRCA2 using targeted massively parallel<br />

sequencing, or next-generation sequencing.<br />

For this study, DNA was used from 10 individuals referred for genetic<br />

testing after meeting the criteria for inherited breast cancer, and had<br />

been screened for BRCA1 and BRCA2 mutations by the Hunter<br />

Area Pathology Service (Newcastle, NSW, Australia). All individuals<br />

used for this study did not harbour causative genetic changes in the<br />

coding regions of BRCA1 or BRCA2. Targeted next-generation<br />

paired-end sequencing of regions containing BRCA1 and BRCA2<br />

was performed using Agilent SureSelect and an Illumina GAIIx<br />

(The Ramaciotti Centre for Gene Function Analysis). An average of<br />

50x coverage was achieved across the targeted genomic region for<br />

all samples. The sequence data was aligned to the Human Reference<br />

Sequence 37.2. Single nucleotide polymorphisms present in dbSNP<br />

or the 1000 genomes project were removed from further analyses.<br />

Genetic differences in the form of single nucleotide variants (SNVs)<br />

and insertions/deletions (indels) were identified in most individuals<br />

tested in regions that had previously remained unexplored, such as the<br />

non-coding regions of BRCA1 and BRCA2, the 5’UTR and promoter<br />

sites. Structural variations including large deletions, duplications,<br />

insertions, inversions and rearrangements were also investigated, but<br />

require further analysis and validation for confirmation.<br />

This study has comprehensively investigated BRCA1 and BRCA2 and<br />

surrounding genomic regions in a mutation negative inherited breast<br />

cancer population. The issue of accuracy of mutation detection by<br />

traditional methods, such as <strong>San</strong>ger sequencing alone, has also been<br />

addressed by this study. The outcome of this study is the increase<br />

in current knowledge of the genetic variations that results in the<br />

development and/or progression of inherited breast cancer, aid in the<br />

management of individuals with breast cancers by providing a more<br />

specific diagnosis of disease risk and provide information required<br />

for the development of personalized treatment.<br />

P4-11-01<br />

Rapid genetic counseling and testing in newly diagnosed breast<br />

cancer patients, findings from an RCT<br />

Wevers MR, Ausems MG, Bleiker EM, Rutgers EJ, Witkamp AJ, Hahn<br />

DE, Brouwer T, Kuenen MA, van der <strong>San</strong>den-Melis J, van der Luijt RB,<br />

Hogervorst FB, van Dalen T, Theunissen EB, van Ooijen B, de Roos<br />

MA, Borgstein PJ, Vrouenraets BC, Huisman JJ, Bouma WH, Rijna<br />

H, Vente JP, Valdimarsdottir H, Verhoef S, Aaronson NK. Netherlands<br />

<strong>Cancer</strong> Institute - Antoni van Leeuwenhoek Hospital, Amsterdam,<br />

Netherlands; University Medical Center, Utrecht, Netherlands;<br />

Diakonessen Hospital, Utrecht, Netherlands; St. Antonius Hospital,<br />

Nieuwegein, Netherlands; Meander Medical Center, Amersfoort,<br />

Netherlands; Rivierenland Hospital, Tiel, Netherlands; Onze Lieve<br />

Vrouwe Gasthuis, Amsterdam, Netherlands; St. Lucas Andreas<br />

Hospital, Amsterdam, Netherlands; Tergooi Hospitals, Blaricum,<br />

Netherlands; Gelre Hospitals, Apeldoorn, Netherlands; Kennemer<br />

Gasthuis, Haarlem, Netherlands; Zuwe Hofpoort Hospital, Woerden,<br />

Netherlands; Mount Sinai School of Medicine, New York<br />

Introduction<br />

Female breast cancer patients carrying a BRCA1/2 mutation have<br />

an increased risk of second primary breast and ovarian tumors.<br />

Rapid genetic counseling and testing (RGCT) may aid in making<br />

informed decisions about therapeutic and preventive surgery and<br />

adjuvant treatment. Little is known about the effects of RGCT on<br />

treatment decisions and psychosocial well-being. We have performed<br />

a randomized controlled trial to investigate these issues.<br />

Methods<br />

Newly diagnosed breast cancer patients from 12 Dutch hospitals with<br />

at least a 10% risk of carrying a BRCA1/2 mutation were randomized<br />

to an intervention group (RGCT) or a usual care control group (ratio<br />

2:1). Study outcomes included uptake of RGCT, choice of type of<br />

surgery, cancer risk perception, cancer-specific distress, quality of life<br />

and decisional satisfaction. Assessments took place at study entry,<br />

and at 6 and 12 months follow-up.<br />

Results<br />

Between 2008 and 2010, 271 patients were recruited, of whom 3<br />

subsequently withdrew. The remaining 268 patients were randomized<br />

to the intervention (n=181) or control (n=87) group. Complete<br />

questionnaire data were available for 250 (93%) and 243 (91%)<br />

patients at 6 and 12 months follow-up, respectively. Of the 181<br />

women in the intervention group, 180 (98%) underwent genetic<br />

counseling after a median of 4 days. One-hundred thirteen (63%)<br />

of them opted for accelerated DNA test procedures, of whom 72<br />

underwent rapid testing (results available in


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

control group, respectively). Data on additional surgery performed<br />

during follow-up and psychosocial outcomes will be available at the<br />

time of the conference.<br />

Conclusion<br />

The uptake of rapid genetic counseling among high-risk breast<br />

cancer patients was high, and the majority of patients underwent<br />

accelerated DNA-testing procedures. However, RGCT did not have<br />

a significant effect on choice of type of primary surgery. In part, this<br />

may be explained by the fact that surgeons and patients often did<br />

not wait for DNA test results before primary surgery. Conclusions<br />

regarding the psychosocial impact of RGCT will be presented at the<br />

time of the conference.<br />

P4-11-02<br />

Novel BRCA1 and BRCA2 genomic rearrangements in Southern<br />

Chinese breast/ovarian cancer patients<br />

Kwong A, Ng EKO, Law FBF, Wa A, Wong CLP, Wong CHN,<br />

Kurian AW, West DW, Ford JM, Ma ESK. University of Hong Kong,<br />

Pokfulam, Hong Kong; Hong Kong <strong>San</strong>itorium & Hospital, Happy<br />

Valley, Hong Kong; Hong Kong Hereditary <strong>Breast</strong> <strong>Cancer</strong> Family<br />

Registry, Happy Valley, Hong Kong; Stanford University School of<br />

Medicine, Stanford, CA<br />

Background and Aims: Germline mutations in the two breast<br />

cancer susceptibility genes, BRCA1 and BRCA2 account for a<br />

significant portion of hereditary breast/ovarian cancer. Most of the<br />

BRCA mutations reported in Southern Chinese patients were point<br />

mutations, small deletions, and insertions. The spectrum of large<br />

genomic rearrangement (LGR) is largely unknown. Here we perform<br />

the first study on the LGR of BRCA genes in a Hong Kong Chinese<br />

population. We aimed to determine the spectrum of BRCA LGRs in<br />

Southern Chinese patients with breast cancer.<br />

Methods: A total of 555 clinically high-risk breast and/or ovarian<br />

cancer patients were recruited from the Hong Kong Hereditary <strong>Breast</strong><br />

<strong>Cancer</strong> Family Registry, diagnosed from March 2007 to November<br />

2011. Multiplex ligation-dependent probe amplification (MLPA)<br />

for detecting BRCA LGRs together with comprehensive BRCA1 and<br />

BRCA2 gene sequencing of all coding exons were performed. cDNA<br />

sequencing of the LGRs was performed to locate the breakpoint of<br />

the deletions.<br />

Results: Overall BRCA1/2 mutation prevalence among this cohort<br />

was 12.4% (69/555). Among the 69 mutations identified, 4 novel<br />

LGRs (2 in BRCA1 and 2 in BRCA2) were detected only by MLPA<br />

but not full gene sequencing. Overall the LGR genes accounted for<br />

5.8% (4/69) of all BRCA mutations in our cohort, 6.9% (2/29) of all<br />

BRCA1 mutations and 5% (2/40) of all BRCA2 mutations.<br />

Conclusions: These findings highlight the LGR spectrum of BRCA1<br />

and BRCA2 genes in Southern Chinese breast cancer patients. LGR<br />

testing together with BRCA1/2 full gene sequencing is superior<br />

to other methods for comprehensive BRCA1/2 analysis in clinical<br />

settings.<br />

P4-11-03<br />

Single nucleotide polymorphism testing for breast cancer risk<br />

assessment: patient trust and willingness to pay<br />

Howe R, Omer Z, Hanoch Y, Miron-Shatz T, Thorsen C, Ozanne EM.<br />

Universiy of California <strong>San</strong> Francisco, CA; Massachusetts General<br />

Hospital, Boston, MA; Plymouth University, United Kingdom; Ono<br />

Academic College, Israel<br />

Background<br />

The field of breast cancer risk assessment is advancing rapidly<br />

with recent discoveries about risk conferring single nucleotide<br />

polymorphisms (SNPs). While these discoveries can promote<br />

personalized medicine, they are often brought to the market with<br />

direct to consumer (DTC) testing before data can support their<br />

widespread use and before reliable options for dealing with testing<br />

outcomes can be offered. In this context, and knowing that patients<br />

often misunderstand risk information, it is unclear how patients will<br />

respond to these options.<br />

Methods<br />

We surveyed high risk women’s interest in SNP testing. Participants<br />

were recruited from the <strong>Cancer</strong> Genetics Network (CGN), a national<br />

network of cancer centers that maintains a database of individuals<br />

with a family history of cancer. Participants were asked to answer<br />

questions regarding their interest in SNP testing including: whether<br />

they trust it, how much they would be willing to pay for testing, how<br />

they prefer to be tested, and how they would proceed with information<br />

identifying them as below or above average risk.<br />

Results<br />

189 women without a history of breast cancer or SNP testing<br />

completed the questionnaire. The average age of the participants was<br />

49, ranging from 30 to 65. All participants had at least one relative<br />

with breast or ovarian cancer. 13% had previously tested positive for<br />

a BRCA mutation, and 33% had received BRCA testing. Most women<br />

(90%) did not know what SNP testing was prior to the survey. Once<br />

SNP testing was described, 68% of women were interested in DTC<br />

SNP testing; at the same time, only 38% of the participants reported<br />

that they trusted DTC SNP testing.<br />

Survey Question<br />

Participant Responses<br />

Know what SNP testing is? 18 (10%)<br />

Interested in DTC SNP testing? 128 (68%)<br />

Willingness to pay for SNP testing? $125, range: $0-1000<br />

Trust DTC SNP testing? 70 (38%)<br />

Above average<br />

risk results<br />

Below average<br />

risk results<br />

How they would proceed with results<br />

Discuss with doctor 146 (77%) 112 (59%)<br />

Discuss with genetic counselor 118 (62%) 67 (35%)<br />

Research on own 74 (39%) 56 (30%)<br />

Discuss with testing company 28 (15%) 16 (8%)<br />

Skeptical of results 21 (11%) 35 (19%)<br />

No further action 2 (1%) 43 (23%)<br />

None of the above 2 (1%) 0 (0%)<br />

Intervention choices<br />

Lifestyle changes after contacting doctor 139 (74%) 90 (48%)<br />

Regular mammograms 93 (49%) 108 (57%)<br />

Surgery 69 (37%) 41 (22%)<br />

Alternative medicine 60 (32%) 33 (17%)<br />

Lifestyle changes without contacting doctor 42 (22%) 40 (21%)<br />

None 15 (8%) 70 (38%)<br />

Conclusion<br />

While our results show that women are interested in DTC SNP<br />

testing, their willingness to pay is lower than the DTC cost (∼$300).<br />

Involvement of genetic counselors and providers in SNP testing<br />

discussions may be needed to overcome the current lack of trust<br />

of DTC testing among patients. Many women showed interest in<br />

lifestyle interventions, suggesting that these interventions should be<br />

incorporated as part of standard follow-up recommendations. When<br />

identified by SNP testing as “below average” risk, women do not<br />

seem to trust the results enough to forego regular mammograms. As<br />

DTC testing becomes more common, and as more SNP tests become<br />

available, it will be necessary for the medical community to address<br />

patients’ interest in these tests and to assist in interpreting results.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

392s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 4<br />

P4-11-04<br />

A Structured Genetic Risk Evaluation and Testing Program in<br />

the Community Oncology Practice Increases Identification of<br />

Individuals at Risk for BRCA Mutations<br />

Langer L, Clark L, Gress J, Patt D, Denduluri N, Wang Y, Andersen<br />

J, Solti M, Wheeler A, Delamelena T, Smith, II JW, <strong>San</strong>dbach J.<br />

Compass Oncology, Portland, OR; Texas Oncology, Austin, TX;<br />

Virginia <strong>Cancer</strong> Specialists, Arlington, VA; McKesson Specialty<br />

Health/The US Oncology Network, The Woodlands, TX<br />

Background: Genetic risk assessment is an important component of<br />

the care of the community oncology breast cancer patient. However,<br />

identification of at-risk patients is largely an ad-hoc process and<br />

practices lack a systematic approach to genetic risk evaluation. The<br />

US Oncology Network Genetic Risk Evaluation and Testing (USON<br />

GREAT) Program provides a structured approach to implementation<br />

of genetic risk evaluation, testing, and triage for appropriate<br />

intervention.<br />

Methods: In 2009, our multi-disciplinary community oncology<br />

practice implemented the USON GREAT Program. The practice’s<br />

program has a single dedicated nurse practitioner and physician lead,<br />

trained in part through a core educational curriculum and utilizing<br />

US Oncology Network-wide genetics resources (web-based MD,<br />

midlevel, and genetic counselor conferencing; discussion Portal;<br />

published guidelines and office procedures). NCCN guidelines were<br />

used to guide testing recommendations. Sequential risk evaluations<br />

were documented prospectively. We retrospectively analyzed how<br />

evaluation patterns changed over a 4 year time period. We also sought<br />

to capture descriptive characteristics of the evaluated population.<br />

Results: Overall, between 2008 and 2011, our practice evaluated<br />

1018 patients at potential risk for a BRCA mutation (mut), based on<br />

personal history of breast cancer under age 50; ovarian, fallopian or<br />

peritoneal cancer; known family history of malignancy; or known<br />

BRCA mutation in the family.<br />

Total Risk Evaluations by Year<br />

Year New Patients* Total Evaluated (% of new pt)<br />

2008 1116 71 (6%)<br />

2009 1168 270 (23%)<br />

2010 1095 353 (32%)<br />

2011 924 324 (35%)<br />

* Patients with DCIS, invasive breast cancer, ovarian/fallopian/peritoneal cancers<br />

In 2008, 6% of potential at-risk individuals were identified vs 35%<br />

in 2011. NCCN guideline exclusions for BRCA testing in invasive<br />

breast cancer were 8% in 2008 and 3% in 2010.<br />

Test Results by Diagnosis<br />

Diagnosis Number Evaluated BRCA1 BRCA2 VUS Negative<br />

Invasive <strong>Breast</strong> <strong>Cancer</strong> 655 24 32 20 477<br />

DCIS 55 2 0 1 48<br />

Ovarian/Fallopian/Peritoneal 143 16 9 5 96<br />

Unaffected 165 12 23 6 96<br />

Totals 1018 54 64 32 717<br />

150 deleterious mut and variants of uncertain significance (VUS)<br />

were identified. There was an 14.7% overall identification rate for<br />

BRCA1/2 (B1, B2) mut and VUS. Among mut and VUS identified<br />

by cancer type, B1 mut was more commonly identified in patients<br />

with a gynecologic malignancy (53% B1 vs 30% B2, 17% VUS); mut<br />

in invasive breast cancer were more likely to be in B2 (42% B2 vs<br />

32% B1, 26% VUS). 7% of all tests for individuals with malignancy<br />

were declined or cancelled due to insurance or finances, vs 37% for<br />

unaffecteds, despite their high risk of mutation carrier status.<br />

Conclusions: We report a single practice’s four-year experience with<br />

implementation of the USON GREAT Program. The results from this<br />

experience demonstrate that the USON GREAT Program results in<br />

higher rates of identification of at-risk individuals, and promotes<br />

more appropriate guidelines-based testing in the community oncology<br />

setting. The relative frequency of BRCA2 vs BRCA1 in invasive<br />

breast cancer is of unclear significance at this time and warrants further<br />

analysis. Cost of testing remains a barrier to appropriate utilization.<br />

P4-11-05<br />

Optimal age to start preventive measures in women with<br />

BRCA1/2 mutations or high familial breast cancer risk<br />

Tilanus-Linthorst MMA, Lingsma H, Evans GD, Kaas R, Manders<br />

P, Hooning MJ, van Asperen CJ, Thompson D, Eeles R, Oosterwijk<br />

JC, Leach MO, Steyerberg EJ. Erasmus University MC, Rotterdam,<br />

Netherlands; Erasmus University MC, Netherlands; University of<br />

Manchester, United Kingdom; NKI/AVL, Amsterdam, Netherlands;<br />

UMC St Radboud, Netherlands; Leiden University Medical Centre,<br />

Netherlands; Centre for <strong>Cancer</strong> Genetic Epidemiology, Cambridge,<br />

United Kingdom; Royal Marsden NHS Foundation, United Kingdom;<br />

University Medical Centre Groningen, Netherlands; Institute of<br />

<strong>Cancer</strong> Research, Royal Marsden, Sutton, United Kingdom<br />

Background<br />

Women from high risk breast cancer families consider preventive<br />

measures including screening. Guidelines on screening differ<br />

considerably regarding starting age. We investigated whether age<br />

at diagnosis in affected family members is predictive for age at<br />

diagnosis of a relative.<br />

Methods<br />

We analyzed the age of breast cancer detection of 1304 first and<br />

second degree relatives of 314 BRCA1, 164 BRCA2 and 244 highrisk<br />

participants of the Dutch MRI-SCreening study. The within<br />

and between family variance in age at diagnosis was analyzed with<br />

a random effect linear regression model. We compared the starting<br />

age of screening based on risk-group (25 years for BRCA1, 30 years<br />

for BRCA2 and 35 years for familial risk), on family history, and on<br />

the model, which combines both. The findings were validated in 63<br />

families from the UK- MARIBS study.<br />

Results<br />

Mean age at diagnosis in the family varied between families; 95%<br />

range of mean family ages was 35-55 in BRCA1 -, 41-57 in BRCA2-<br />

and 44-60 in high-risk families.14% of the variance in age at diagnosis<br />

was explained by family history, 7% by risk group.<br />

Approaches to determining start of screening based on the model<br />

and on risk-group were similar in terms of cancers missed and years<br />

of screening. The approach based on familial history only, missed<br />

more cancers and required more screening years in both the Dutch<br />

and UK datasets.<br />

Conclusion<br />

Age at breast cancer diagnosis is in part dependent on family history<br />

which may assist planning starting age for preventive measures.<br />

Keywords: BRCA1, BRCA2 , breast cancer, prevention, age at onset<br />

P4-11-06<br />

Automatic referral to genetic counseling for identification of<br />

BRCA1/2 mutations: a pilot program at Norton <strong>Cancer</strong> Institute,<br />

Louisville, KY.<br />

Lewis AL, Alabek ML, Dreher C, Goldberg JM, Brooks SE. Norton<br />

Healthcare, Louisville, KY<br />

BACKGROUND: Identifying a hereditary cancer syndrome has the<br />

potential to significantly impact a patient’s treatment and long-term<br />

management, as well as prevent future malignancies among relatives.<br />

Therefore, referral of patients appropriate for genetic counseling<br />

and testing is critical. Providers traditionally initiate patient referral;<br />

however, this can lead to inconsistent referrals and results in failure<br />

to refer more than 50% of appropriate individuals, according to recent<br />

www.aacrjournals.org 393s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

studies. In an attempt to address these quality issues, we developed<br />

an automatic referral program using the National Comprehensive<br />

<strong>Cancer</strong> Network guidelines, which was approved through our cancer<br />

committee and implemented 10/1/10. This pilot was conducted within<br />

a genetic counseling program staffed by board certified genetic<br />

counselors and associated with a private, multi-disciplinary oncology<br />

practice that is part of an American College of Surgeons accredited<br />

Network <strong>Cancer</strong> Center. Our <strong>Cancer</strong> Center is part of a five-hospital<br />

integrated healthcare system and a part of the National <strong>Cancer</strong> Institute<br />

Community <strong>Cancer</strong> Centers Program network.<br />

METHODS: We conducted weekly reviews of oncology patients<br />

scheduled for upcoming appointments to identify individuals<br />

diagnosed with breast cancer before age 50 or with ovarian, fallopian,<br />

or primary peritoneal cancer at any age. Patients previously referred<br />

to our genetic counseling program were excluded. Providers were<br />

notified weekly of their patients identified via this program, with the<br />

option to decline referral. We undertook a retrospective review of<br />

the outcomes of individuals identified from 10/1/10 through 10/1/11,<br />

with follow up as of 6/1/12. IRB approval was obtained through the<br />

University of Louisville.<br />

RESULTS: We identified 521 patients for referral, 24 (4.6%) of<br />

whom were declined by a provider, resulting in 497 referrals. Three<br />

hundred forty one (69%) referrals had breast cancer and 156 (31%)<br />

had other malignancies. Of referrals, 139 (28%) have been seen,<br />

223 (45%) have declined, and 135 (27%) are in process. Testing was<br />

pursued by 108 (78%) of referrals seen, all of whom had BRCA1/2<br />

testing and 5 (4.6%) of whom had additional testing. We identified<br />

11 (10%) individuals with a BRCA1/2 mutation. An additional 17<br />

(16%) individuals who completed testing were counseled to consider<br />

enhanced surveillance in the absence of a confirmed hereditary cancer<br />

syndrome. The total number of first degree relatives with potential to<br />

benefit from this program is 62 among individuals with a BRCA1/2<br />

mutation and 86 among individuals without a confirmed hereditary<br />

cancer syndrome.<br />

CONCLUSIONS: Initiation of an automatic genetic counseling<br />

referral program in the setting of a large oncology practice is feasible<br />

and has the potential to identify individuals with a BRCA1/2 mutation<br />

who might otherwise go undetected. Few providers object to referral,<br />

and the majority of patients are receptive to referral. The impact of this<br />

program extends beyond those identified with a BRCA1/2 mutation.<br />

This project was funded in part with federal funds: NCI, NIH Contract<br />

No. HHSN261200800001E.<br />

P4-12-01<br />

A nomogram based on clinical, imaging and histological data to<br />

predict the risk of upgrades to malignancy at surgery in biopsydiagnosed<br />

premalignant lesions of the breast<br />

Uzan C, Mazouni C, Balleyguier C, Mathieu M-C, Ferchiou M,<br />

Delaloge S. Institut Gustave Roussy, Villejuif, France<br />

Background: Population-based mammography screening has resulted<br />

in the increased detection of suspicious, non-palpable lesions that<br />

require histopathological assessment. When a “pre-malignant”<br />

lesion, such as lobular carcinoma in situ (LCIS), atypical ductal or<br />

lobular hyperplasia (ADH and ALH) or flat epithelial atypia (FEA), is<br />

discovered on the primary biopsy, a surgical excision is recommended<br />

due to the risk of underestimation of the lesion. DCIS or invasive<br />

carcinoma is finally retrieved in 10-30% of the patients, meaning a<br />

large part receive unnecessary surgery. The aim of this study was to<br />

define a nomogram integrating clinical, imaging and histological data<br />

to predict for the risk of underestimation of the lesion. Intentionally,<br />

we decided to include all the premalignant lesions which implied<br />

the same management, whereas most of the previous models of the<br />

literature are focused in only one or another type of atypia.<br />

Patients and Methods: We collected complete clinical, radiological<br />

and double-reading histological data on all patients with a diagnosis<br />

of pure atypical lesion (CLIS, ADH, ALH, FEA) on image-guided<br />

biopsy performed at the One-Stop Unjt of our breast care center<br />

from 2004 to 2011. Univariate and multivariate logistic regression<br />

analyses were used to develop a model predicting for the presence of<br />

DCIS or invasive carcinoma on the final surgical sample, and build<br />

a nomogram. This nomogram was evaluated on a training set of 205<br />

patients treated at IGR.<br />

Results: 205 patients were eligible for the study. Of these 205 patients,<br />

50 cancers (24.4%) were diagnosed at definitive surgery (21 DCIS,<br />

20 ductal and 9 lobular invasive carcinoma). Univariate analysis<br />

retrieved age (p=0.03), number of biopsy cores (p=0.02) and type<br />

of radiological anomalies (p=0.02) as factors associated with cancer<br />

at surgery. The presence of microcalcifications on biopsies of limit<br />

significance was included in the multivariate analysis (p = 0.09). The<br />

final most informative nomogram included information on patient<br />

age (p = 0.03), type of radiological anomalies (p= 0.02) and number<br />

of biopsy cores (p=0.04). The predictive accuracy of the nomogram<br />

was 0.71 and 0.68 in the training set before and after bootstrapping,<br />

respectively. The calibration of the nomogram was good. The<br />

sensitivity, specificity, positive predictive value, and negative<br />

predictive values were 68%, 73%, 45%, and 88%, respectively.<br />

Conclusion: This nomogram could help identify a subset of patients<br />

with premalignant disease for whom surgery could be spared and who<br />

would be eligible for exclusive clinical follow-up. A validation study<br />

is currently undergoing in an external dataset.<br />

P4-12-02<br />

Serum 25-hydroxyvitamin D3 is associated with decreased risk<br />

of postmenopausal breast cancer in whites: the Multiethnic<br />

Cohort Study<br />

Kim Y, Franke AA, Shvetsov YB, Wilkens LR, Lurie G, Cooney RV,<br />

Maskarinec G, Hernandez BY, Le Marchand L, Henderson B, Kolonel<br />

LN, Goodman MT. University of Hawai’i <strong>Cancer</strong> Center; University of<br />

Hawaii; Keck School of Medicine, University of Southern California<br />

Ecological studies support a positive relation of sunlight exposure to<br />

a reduced incidence of breast cancer, but findings from prospective<br />

studies of the association of circulating levels of 25-hydroxyvitamin<br />

D (25(OH)D), the established marker of vitamin D status, with the<br />

risk of breast cancer have been null. We conducted a nested casecontrol<br />

study within the Multiethnic Cohort of prediagnostic serum<br />

levels of 25-hydroxyvitamin D2 and 25-hydroxyvitamin D3 among<br />

707 postmenopausal breast cancer cases and matched controls from<br />

Hawaii and Los Angeles. A 5 ng/mL increase in serum levels of<br />

25-hydroxyvitamin D3, the major component of 25(OH)D, was<br />

associated with a significant 28% decreased risk of breast cancer<br />

among white women, but not among women from other ethnic groups<br />

(pheterogeneity = 0.01). Among whites, women considered deficient<br />

in vitamin D (i.e., 25(OH)D


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 4<br />

P4-12-03<br />

Towards a risk prediction model for breast cancer that utilizes<br />

breast tissue risk features<br />

Pankratz VS, Degnim AC, Visscher DW, Frank RD, Vierkant RA,<br />

Ghosh K, Aziza N, Vachon CM, Frost M, Radisky DC, Hartmann LC.<br />

Mayo Clinic, Rochester, MN; Mayo Clinic, Jacksonville, FL<br />

Background: Optimal early detection and prevention strategies for<br />

breast cancer are predicated on our ability to accurately identify<br />

women at increased risk for disease. Current breast cancer risk<br />

prediction models rely on clinical and epidemiologic features. As<br />

demonstrated with other cancers, such as those of the cervix and<br />

colon, cancer risk assessment is enhanced when the tissue at risk<br />

is examined for evidence of premalignant abnormalities. Here, we<br />

present a multivariate model for individualized breast cancer risk<br />

prediction that incorporates tissue features identified from biopsies<br />

from women with benign breast disease (BBD).<br />

Methods: We used our cohort of more than 9000 women diagnosed<br />

with BBD at the Mayo Clinic from 1967-1991, and identified a nested<br />

case:control series of 377 women who developed cancer (cases)<br />

matched with 734 women who did not (controls). We identified a set<br />

of variables that either alone or in two-way interactions contributed to<br />

a multivariable risk prediction model. We combined this relative risk<br />

model with the baseline incidence of breast cancer and the baseline<br />

hazard of death from our full BBD cohort to develop a risk prediction<br />

tool that computes absolute risk estimates for breast cancer. We<br />

calculated 5-year risk predictions for these cases and controls using<br />

our model and using the Gail model and compared c-statistics from<br />

these risk predictions. We validated our model using an independent<br />

set of 378 breast cancer cases and 728 matched controls drawn from<br />

our BBD cohort.<br />

Results: Cases and controls in the model development data set differed<br />

significantly on a number of features when examined individually.<br />

Those included in a multivariable relative risk model for breast cancer<br />

among women with BBD are shown below.<br />

Factors in risk model<br />

Variable<br />

Levels<br />

Histologic Impression<br />

Proliferative vs. not<br />

Involuted Lobules 1-74% vs.


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P4-12-05<br />

Enrichment of aldosterone synthase (CYP11B2) gene C-allele<br />

carriers in women at high risk in the OncoVue® polyfactorial<br />

risk model<br />

Jupe ER, Pugh TW, Knowlton NS, DeFreese DC. InterGenetics<br />

Incorporated, Oklahoma City, OK; NSK Statistical Solutions,<br />

Choctaw, OK<br />

Background: Models for accurately assessing breast cancer risk<br />

are utilized for guiding clinical decisions regarding early detection<br />

and prevention. We developed a polyfactorial risk model (PFRM;<br />

OncoVue) from analysis of a large case-control database including<br />

both functional gene polymorphism and clinical factor data. A<br />

multivariate logistic regression based statistical protocol was used<br />

to derive a single risk model consisting of 22 single nucleotide<br />

polymorphisms (SNPs) in 19 genes and 5 traditional risk factors.<br />

Several studies have shown that the PFRM is significantly improved<br />

in individualized risk estimation compared to the <strong>Breast</strong> <strong>Cancer</strong> Risk<br />

Assessment Tool (BCRAT) or Gail model. Here we have examined<br />

the genotype frequencies of women placed in the highest decile of risk<br />

by the PFRM compared to both the overall control population and to<br />

women in the control population placed at the lowest decile of risk.<br />

Materials and Methods: Analyses were performed on 1671 breast<br />

cancer cases and 3351 controls that enrolled in a case-control study<br />

conducted in six regions of the United States. DNAs were genotyped<br />

for the 22 genetic polymorphisms and combined with clinical risk<br />

factor information to calculate PFRM risk estimates for the individual<br />

participants. The PFRM lifetime scores were used to stratify the<br />

upper and lower deciles of risk. The genotype frequencies for these<br />

stratified risk groups were determined and compared to the overall<br />

control population and to each other (upper vs. lower decile). Odds<br />

Ratios (OR) were determined for the CYP11B2 genotypes because<br />

they exhibited the largest differences in frequency among the groups.<br />

Results: For CYP11B2 comparison of the genotype frequencies in<br />

the highest decile risk group to the overall genotype frequencies<br />

of controls identified significant enrichment of the C-allele carriers<br />

in the highest risk group. The OR for C-allele carriers (C/C+C/T)<br />

compared to the most common T/T genotype was 3.0 (1.9, 4.8; 95%<br />

C.I.) with a p-value


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 4<br />

odds ratio was consistent across both case control studies (OR 1.25,<br />

95% CI 1.06-1.46) and cohort studies (OR 1.26, 95% CI 1.09-1.45).<br />

Heterogeneity was significant (I² = 73.40, p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

mammogram history), and Correlates of <strong>Breast</strong> Health (e.g., smoking<br />

behavior, alcohol consumption), we found that clients were between<br />

3.2 and 7.2 times more likely to complete the sections via ACASI<br />

as opposed to SAPI.<br />

Rates of CIF completion by interview mode<br />

Number of items ACASI, % complete FFI, % complete SAPI, % complete<br />

in section (n=101)<br />

(n=100) (n=100)<br />

Overall CIF 34 73.3 64.0 37.0<br />

Demographics 12 82.2 85.0 59.0<br />

<strong>Breast</strong> Health<br />

Information<br />

11 94.1 93.0 80.0<br />

Correlates of <strong>Breast</strong><br />

Health<br />

11 93.1 78.0 65.0<br />

Discussion:<br />

Overall, we found that use of ACASI resulted in significantly higher<br />

rates of overall form completion and lower rates of missing data than<br />

use of SAPI or FFI, with the greatest identified disparity in form<br />

completion between ACASI and SAPI. This study has important<br />

implications for breast health specialists or any health practitioners<br />

who regularly rely on self-administered questionnaires and/or<br />

face-to-face interviews to collect important health information. We<br />

recommend that where feasible, ACASI be utilized as an effective<br />

means of collecting high quality client-level data.<br />

P4-13-03<br />

Hormone replacement therapy, is there an increased risk of in<br />

situ breast cancer ? data from a french cohort<br />

Coussy F, Giacchetti S, Hamy A-S, Porcher R, Cuvier C, Lalloum<br />

M, de Roquancourt A, Albiter M, Espié M. Hopital St Louis, Paris,<br />

France; Hôpital St Louis, Paris, France; Assistance Publique-<br />

Hôpitaux de Paris, Hôpital St Louis, Paris, France<br />

Background: Use of hormone replacement therapy (HRT) has been<br />

associated with an increased risk of breast cancer. Few studies focus<br />

on correlation between HRT and in situ breast cancer A large cohort<br />

study, the Million Women Study (Reeves GK et al. Lancet Oncol.<br />

2006) have shown that the use of HRT has been associated with<br />

an increased incidence of in situ breast cancer (RR= 1, 55 , IC=1,<br />

4-1,72, p=0,03).<br />

Purpose: We investigated the association between HRT use and<br />

duration and in situ breast cancer incidence from a data base of<br />

infraclinic breast lesions.<br />

Materials & Methods: From 2007 to 2011, 2708 patients (pts) with a<br />

non palpable breast lesion were referred to our breast disease unit for<br />

exploration (Saint Louis Hospital, Paris). Radiological abnormalities<br />

were screened by mammography and/or breast ultrasound, and/or<br />

MRI. Out of 2708 pts, 1668 had a biopsy. All biopsies were seen by<br />

an anatomopathologist dedicated to breast. Here, we focus on the<br />

1017 postmenopausal women (61%).<br />

Results: Biopsies revaled invasive breast cancers in 222 pts (22%),<br />

in situ breast cancers in 164 pts (16%) [10 lobular in situ carcinoma<br />

and 154 ductal in situ carcinoma], high risk breast lesions in 103 pts<br />

(10%) and benign breast lesions in 528 patients (52%). Characteristics<br />

of the 164 patients with in situ breast cancer: median age 62 [IQR:<br />

57 to 68], at least one pregnancy (mean number of pregnancies per<br />

woman, 1.6) 75,6%, family history of breast cancer:30,2%. Among<br />

these 164 patients, 42.6 % had used an oral contraceptive and 53%<br />

of them a HRT, with a mean duration of use of 7 years.<br />

The HRT use was not significantly associated with in situ breast<br />

cancer (p=0.66) as well as the duration of HRT.<br />

Benin In situ breast cancer p<br />

Nb of patients 528 164<br />

Median age (IQR), ys 59 (55-66) 62 (57-68) 0.008<br />

Mean nb of pregnancy (SD) 1.8 (1.4) 1.6 (1.3) 0.24<br />

Median duration of oral contraception (IQR), ys 0 (0-7) 0 (0-8) 0.97<br />

Median duration of HRT (IQS) , ys 0 (0-6) 1 (0-6) 0.66<br />

Duration of HRT, nb (%) 0.89<br />

never use 260 (49.6) 77 (47)<br />


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 4<br />

TABLE 1 - Number of avoided events due to Estrogen, over 10 year follow up<br />

Estrogen- Placebo<br />

GROUP - Ages 50-59 Annualized -Annualized RR [95% C.I.]<br />

(%) (%)<br />

CHD: 0.18 0.31 0.59 [0.38-0.90] - 1,300<br />

iBrCA 0.24 0.30 0.80 [0.53-1.19] - 600<br />

All Deaths 0.35 0.48 0.73 [0.53-1.00] - 1,300<br />

GROUP - ALL AGES<br />

iBrCa 0.27 0.35 0.77 [0.62-0.95] - 800<br />

NAE / 100,000 over<br />

10 years<br />

iBrCa: Women without<br />

0.23 0.38 0.61 [0.47-0.79] - 1,500<br />

PHBBD (80% of all)<br />

CONCLUSION.<br />

1. Estrogen therapy could reduce thousands of BrCa, CHD and AC-<br />

Mortality events annually, just in North America. These gains are in<br />

addition to the established quality of life improvement for millions<br />

of women due to [E].<br />

2. Of particular importance is the [E] effect on reduction of iBrCa<br />

rates, particularly significant for women without PHBBD, confirming<br />

the new paradigm of Dual E effect for human BrCa [Ref 3].<br />

3. These substantial Public Health gains associated with [E] may<br />

justify changing policy to incorporate [E] into HRT guidelines for<br />

appropriately selected women.<br />

4. Accelerated research to optimize [E] formulations, and identifying<br />

subsets that benefit most, is urgently required for optimum HRT use<br />

in Prevention of iBrCa, CHD, and reducing AC- mortality.<br />

REFERENCES<br />

1. LaCroix AZ, et.al. Health outcomes after stopping conjugated equine<br />

estrogens among postmenopausal women with prior hysterectomy: a<br />

randomized controlled trial. JAMA 2011;305:1305-14.<br />

2. Anderson GL, et.al. Conjugated equine oestrogen and breast<br />

cancer incidence and mortality in postmenopausal women with<br />

hysterectomy: extended follow-up of the Women’s Health Initiative<br />

randomised placebo-controlled trial Lancet Oncol 2012;13:476-86.<br />

3. Ragaz J, et.al. Dual estrogen effects on breast cancer: endogenous<br />

estrogen stimulates, exogenous estrogen protects. Further investigation<br />

of estrogen chemoprevention is warranted. <strong>Cancer</strong> Res 2010;70.<br />

P4-13-05<br />

The gap between perceptions of risk and actual risk for breast<br />

cancer.<br />

Kaplan CP, Lopez M, Tice J, Pasick R, Kerlikowske K, Chen A,<br />

Karliner L. University of California, <strong>San</strong> Francisco, CA<br />

Background: Estimating a woman’s individual risk for breast cancer<br />

is essential for tailoring appropriate screening and risk reduction<br />

strategies. Prior research suggests that women often misperceive<br />

their risk as either higher or lower than it actually is. As a result,<br />

average risk women who perceive their risk to be high may experience<br />

unnecessary anxiety. In contrast, high risk women who perceive their<br />

risk to be average/low may not utilize needed screening or available<br />

risk reduction options. The overall aim of this study is to compare<br />

perception of breast cancer risk to actual risk among a sample of<br />

multi-ethnic women with a range of breast cancer risk.<br />

Methods: As part of BREASTCARE, a randomized controlled<br />

trial designed to evaluate a PC-tablet based intervention providing<br />

multi-ethnic women and their primary care physicians with<br />

tailored information about breast cancer risk, we collected baseline<br />

information regarding perception of risk. Women visiting general<br />

internal medicine primary care clinics at two sites (one academic<br />

practice and one safety net practice) during the study period, between<br />

the ages of 40 and 74, who spoke English, Spanish or Chinese, and<br />

had no personal history of breast cancer were eligible to participate.<br />

Participants were asked how concerned they were about getting breast<br />

cancer: very concerned, somewhat concerned, a little concerned, or<br />

not at all concerned. They were also asked what they believed their<br />

chances of getting breast cancer were compared to other women their<br />

age: higher, the same, or lower. We classified women as high risk if<br />

they had a positive family history based on the Referral Screening<br />

Tool (Bellcross, et al. 2009), or were in the top 5% estimated 5-year<br />

risk for their age group based on either the Gail Model (Gail et al.<br />

1999) or the <strong>Breast</strong> <strong>Cancer</strong> Surveillance Consortium Model when<br />

breast density data was available (Tice et al. 2008). We then compared<br />

women’s perception of breast cancer risk with their actual risk.<br />

Results: To date, data has been obtained for 913 participants. One<br />

third of the women were white, 23% Black, 23% Latina and 17%<br />

Asian. Twenty percent (N=182) of the women were classified as<br />

high risk. Among average risk women (N=727) 48% were very or<br />

somewhat concerned about getting breast cancer. In contrast, 35%<br />

of high risk women were a little or not at all concerned. When asked<br />

how their chances of getting breast cancer compared to other women<br />

their age, 20% of average risk women thought their risk was higher<br />

than other women their age, while 71% of those at high risk indicated<br />

that they had the same or lower risk than other women their age.<br />

Conclusions: Our results suggest that both average and high risk<br />

women have an incorrect perception of their personal breast cancer<br />

risk compared to their actual risk. Interventions that address this<br />

misunderstanding can help those at high risk to initiate appropriate risk<br />

reduction practices and those at average risk to reduce unnecessary<br />

anxiety.<br />

P4-13-06<br />

Association of Age, Obesity and Incident <strong>Breast</strong> <strong>Cancer</strong><br />

Phenotypes<br />

Scheri R, Power S, Marks J, Seewaldt V, Marcom K, Hwang S. Duke<br />

University Medical Center, Durham, NC<br />

Background: Obesity has been shown to be associated with increased<br />

risk of breast cancer in postmenopausal women. However, it is unclear<br />

whether this risk is isolated to a specific phenotype. We hypothesized<br />

that the increased levels of insulin, IGF-1 and estrogens associated<br />

with obesity preferentially drive an increased proportion of the<br />

luminal A phenotype of breast cancer.<br />

Methods: Between 2008 and 2011, 1001 women were diagnosed<br />

with invasive breast cancer at Duke University Medical Center and<br />

had weight and height recorded at the time of diagnosis. Tumor<br />

phenotypes were based on hormone receptor (HR: ER- and/or PRpositive)<br />

and Her2 testing with subtypes defined as follows: Luminal<br />

A: HR(+), Her2(-); Luminal B: HR(+), Her2(-); Her2: HR(-), Her2(+);<br />

triple negative: HR(-), Her2(-).<br />

Results: Median age of the cohort was 55.7 years; median BMI was<br />

27.7. As expected, increasing BMI was associated with older age, with<br />

BMI almost evenly distributed between three groups: ≤ 25, 25-30, and<br />

>30. In the overall group, proportion of luminal B, Her2, and triple<br />

negative subtypes did not differ by BMI; however the proportion of<br />

patients with luminal A cancer increased from 65% for the lowest<br />

BMI group to 70% for the highest BMI group. Analysis stratified by<br />

age ≤ 40, 40 to 59, and ≥60 years showed a lower proportion of the<br />

triple-negative (19% vs. 6% vs. 4%, p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P4-13-07<br />

Meta-analysis of epidemiological studies of Insulin Glargine and<br />

<strong>Breast</strong> <strong>Cancer</strong> Risk<br />

Boyle P, Koechlin A, Boniol M, Bota M, Robertson C, Rosenstock J,<br />

Bolli GB. International Prevention Research Institute, Lyon, France;<br />

University of Strathclyde, Glasgow, United Kingdom; Dallas Diabetes<br />

and Endocrine Center, Dallas; University of Perugia, Italy<br />

After having lain dormant for a while, the association between<br />

diabetes, its risk factors and treatments, and cancer risk and death<br />

is now high on the clinical and research agenda. The microscope<br />

is currently focused on the relationships between Pioglitazone and<br />

Bladder <strong>Cancer</strong>, Exenatide and Pancreas <strong>Cancer</strong>, Liraglutide and<br />

pancreas cancer and insulin use and lung cancer. The potential<br />

association between use of insulin glargine and breast cancer risk<br />

has been the subject of recent major studies.<br />

All data regarding cancer risk and use of insulin glargine has been<br />

assembled and meta-analyses performed using state-of-the-art<br />

statistical methodology. A random effects model was employed with<br />

tests for heterogeneity (I 2 ) and publication bias. These meta-analyses<br />

are based on reports from 21 epidemiological studies involving over<br />

one million patients and 3 million person-years of observation.<br />

Based on independent estimates from these studies, the Summary<br />

Relative Risk (SRR) for all forms of cancer was (SRR=0.91, 95% CI<br />

(0.84, 0.99)), and for breast cancer SRR=1.11 (95% CI (1.00, 1.48)).<br />

For new users of glargine, the SRR for breast cancer was SRR=1.22<br />

(95% CI (1.00, 1.48)). For colorectal cancer the SRR=0.83 (95% CI<br />

(0.74, 0.94)) and for prostate cancer SRR=1.14 (95% CI (0.93, 1.39)).<br />

Overall, the risk of developing cancer among users of insulin glargine<br />

is reduced compared to the risk of users of other insulins. Similarly,<br />

the risk of colorectal cancer is reduced among users of glargine.<br />

While the lower bound of the 95% confidence interval is 1.00, the<br />

risk of breast cancer does not increase with increasing duration of<br />

use of glargine. In some studies the trend in risk with increasing<br />

duration of use goes in opposite directions. The development of a<br />

detectable breast cancer from the initial carcinogenic event depends<br />

on the tumour doubling time. The time for a de novo breast cancer to<br />

become detectable ranges from 12.3 years for a doubling time of 150<br />

days; 16.4 years for a doubling time of 200 days; and 20.5 years for a<br />

doubling time of 250 days. Most published studies have a maximum<br />

of 3-4 years of glargine use.<br />

The databases employed in these analyses were not designed for<br />

such epidemiological investigation. A major limitation is the absence<br />

of knowledge as to why a potential treatment was prescribed for<br />

an individual and why a change in therapy was indicated. Further<br />

potential limitations to this meta-analysis include that the comparison<br />

group was not the same in all studies but this could also be seen as<br />

a strength. The meta-analysis of the randomized trials had several<br />

insulin comparators and the retinopathy study had NPH as the<br />

comparator. This is not likely to invalidate the findings of this<br />

analysis nor would the fact that different adjustments were made in<br />

the individual studies.<br />

The current evidence gives no support to the hypothesis that insulin<br />

glargine is associated with an increased risk of cancer as compared<br />

to other insulins and should give reassurance to physicians and their<br />

patients. In respect to breast cancer, there is no indication of a causal<br />

association between use of insulin glargine and increased risk of<br />

breast cancer.<br />

P4-13-08<br />

Diabetes, Related Factors and <strong>Breast</strong> <strong>Cancer</strong> Risk<br />

Boyle P, Boniol M, Koechlin A, Bota M, Robertson C, Leroith D,<br />

Rosenstock J, Bolli GB, Autier P. International Prevention Research<br />

Institute, Lyon, France; University of Strathclyde, Glasgow, United<br />

Kingdom; Mt. Sinai, New York; Dallas Diabetes and Endocrine<br />

Center, Dallas; University of Perugia, Italy<br />

Diabetes and breast cancer are both extremely common conditions in<br />

women and may share common risk factors. It is natural to investigate<br />

any potential common risk factors and to seek biological clarification<br />

and improve prospects for prevention.<br />

Therefore, in order to help clarify the potential association between<br />

diabetes, related factors and breast cancer risk, a comprehensive<br />

literature review and formal meta-analysis was carried out, planned,<br />

conducted and reported following PRISMA guidelines regarding<br />

meta-analysis of observational studies. Variables studies in relation<br />

to breast cancer risk were adiposity, physical activity, glycaemic load,<br />

glycaemic index, diabetes, IGF-1, fasting glucose, fasting insulin<br />

and C-peptide, adiponectin and metformin and glargine use among<br />

patients with diabetes. For all variables except diabetes and breast<br />

cancer, only prospective studies were included in meta-analyses.<br />

Summary Relative Risks (SRR) and corresponding 95% Confidence<br />

Intervals (CI) were calculated from random effect models.<br />

For breast cancer at all ages, the calculated risks were as follows:<br />

diabetes (SRR=1.27 95% CI (1.16, 1.39); physical activity<br />

(SRR=0.88, 95% CI (0.85, 0.92)); glycaemic load (SRR=1.06,<br />

95% CI (1.00, 1.12)); glycaemic index (SRR=1.04, 95% CI (0.99,<br />

1.10)); fasting glucose (SRR=1.12, 95% CI (1.01, 1.24)); serum<br />

insulin (SRR=1.18, 95% CI (0.75, 1.85)); c-peptide (SRR=1.29,<br />

95% CI (0.91, 1.82)); adiponectin (SRR=1.16, 95% CI (0.93,<br />

1.46)); metformin (SRR=1.00, 95% CI (0.69, 1.46)); and glargine<br />

(SRR=1.11, 95% CI (1.00, 1.24)). An increase of 5 units in Body<br />

Mass Index (a weight increase if 14.5 kg in a person 1.70 metres tall)<br />

was associated in post-menopausal breast cancer (SRR=1.12, 95%<br />

CI (1.08, 1.16)) but not at pre-menopausal ages (SRR=0.83, 95%<br />

CI (0.72, 0.95)). Serum insulin was associated with breast cancer at<br />

post-menopausal ages but not at pre-menopausal ages whereas with<br />

c-peptide there was a significant association at pre-menopausal ages<br />

but not post-menopausal. For IGF-1, Hodge’s Standardised Mean<br />

Difference (HSMD) was calculated in cohort studies and there was no<br />

significant association with breast cancer at all ages (HSMD=0.003,<br />

95% CI (-0.059, 0.065)), at post-menopausal ages (HSMD=-0.014,<br />

95% CI (-0.106, 0.077)) or at pre-menopausal ages (HSMD=0.039,<br />

95% CI (-0.038, 0.117)).<br />

The risk of breast cancer is increased among post-menopausal women<br />

who have diabetes. Among those factors related to diabetes, key risk<br />

factors for breast cancer appear to be adiposity and lack of physical<br />

activity which are both related to the risk of developing diabetes.<br />

Action on these lifestyle factors should form the basis of a common<br />

prevention strategy. There is a need to re-evaluate potential biological<br />

mechanisms to explain the increased risk in post-menopausal women<br />

with diabetes.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

400s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 4<br />

P4-13-09<br />

The effect of weight change on breast adipose and dense tissue<br />

Graffy HJ, Harvie MN, Warren RM, Boggis CR, Astley SM, Evans<br />

GD, Adams JE, Howell A. University Hospital of South Manchester,<br />

Manchester, United Kingdom; Addenbrooke’s Hospital, Cambridge,<br />

United Kingdom; University of Manchester, United Kingdom; Central<br />

Manchester University Hospitals NHS Foundation Trust, Manchester,<br />

United Kingdom<br />

Background Observational studies indicate that weight gain or loss<br />

alters breast cancer risk. Hormonal prevention with tamoxifen or<br />

ovarian suppression reduces mammographic breast density, but has<br />

no impact on breast adipose tissue. 1 The aim of this study was to<br />

assess whether change in weight is associated with change in breast<br />

density, breast fat or both. Methods 74 premenopausal women aged<br />

between 35 and 45 years, with a family history of breast cancer,<br />

were recruited from our Family History Clinic. 36 were allocated to<br />

a 25% calorie-restricted Mediterranean diet and exercise intervention;<br />

a further 38 women were separately recruited into a control group<br />

and given written diet and exercise advice only. At baseline and 12<br />

months breast dense and non-dense (largely adipose) components of<br />

the breast were measured: area using Cumulus 2 and volume using the<br />

Manchester ‘stepwedge’ method. 3 Visual assessment of percentage<br />

density was reported to the nearest 5%. 4 We used correlations to<br />

relate percentage change in absolute dense and non-dense (area and<br />

volume) to percentage change in: weight, total body fat (DXA),<br />

intra-abdominal and subcutaneous abdominal fat (MRI), as well<br />

as serum total IGF-I. Results At 12 months, 40 (54%) women<br />

lost >1.5% body weight, 14 (19%) gained >1.5% and 21 (28%)<br />

remained weight stable,


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

comparison of the main prognostic factors was performed to<br />

investigate differences in younger women with primary BC in contrast<br />

to older women over time.<br />

Patients and Methods: In this retrospective analysis a consecutive<br />

pts cohort of 4010 pts was analyzed. Pts were documented and treated<br />

for primary invasive breast cancer between 1963 and 2003 at two<br />

University Hospitals in Germany. To be eligible, pts were required to<br />

have identified tumor characteristics, including TNM-status. Pts with<br />

carcinoma in situ or distant metastases were excluded. The cohort<br />

was divided in two age groups, ≤40y and >40y. Furthermore to reveal<br />

trends and changes over the duration of 41 years the period of analysis<br />

was split into 3 time frames: 1963-1976, 1977-1989 and 1990-2003.<br />

We analyzed the main prognostic factors for BC including tumor<br />

size, grading, nodal status and HR-status in longitudinal comparison<br />

regarding the three time frames, respectively. During 1963-77 HRstatus<br />

was determined in just 12.6% of pts. Thus, this time frame was<br />

excluded in the analysis of HR-status.<br />

Results: In 41 yrs, 747 (18.6%) pts were treated between 1963-76,<br />

1722 pts (42.9%) in 1977-89 and 1541 pts (38.4%) in 1990-2003.<br />

Overall 358 pts were ≤40y and 3652 pts were over the age of 40.<br />

A significant reduction of tumor size (metric assessment) at primary<br />

diagnosis was observed for both age groups (pts≤40y: p=0.012;<br />

pts>40y: p40<br />

y (p=0.001) whereas no difference could be seen in pts aged ≤40 y<br />

(p=0.991).<br />

In both age groups the number of G2/3 tumors increased over the<br />

yrs (pts≤40y: p=0.001; pts>40y: p40y 38.4% and 21.7% were HR-negative,<br />

respectively (p


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 4<br />

also collected and used to identify women at potentially increased<br />

risk. Upon enrollment, women are given the opportunity to provide<br />

a blood or saliva sample for research purposes.<br />

Women who meet Athena-defined criteria that identify them to be<br />

at increased risk receive a referral to a <strong>Breast</strong> Health Specialist<br />

(BHS). The BHS identifies individual patient needs for prevention<br />

and screening services, including genetic counseling and testing,<br />

provides referrals to a High Risk <strong>Breast</strong> Clinic or nurse practitioner,<br />

and conducts lifestyle modification counseling. BHS have special<br />

training in breast cancer risk assessment, and some are licensed<br />

genetic counselors. Primary care and/or referring providers are<br />

directly informed of risk assessment results through mailings or the<br />

electronic medical record.<br />

Results<br />

The recruitment goal enrollment for Athena is 150,000 and to date<br />

more than 17,000 women have been enrolled across the five centers.<br />

Of those enrolled, 32% indicated that they have a family history of<br />

cancer. 56% of the cohort consented to participate in research, and<br />

40% provided a biospecimen for research purposes. Across the five<br />

centers, 32 educational outreach sessions about Athena were held,<br />

reaching approximately 375 providers.<br />

Site UCLA UCI UCD UCSF UCSD Total<br />

Enrollment Period<br />

12/16/2010- 3/28/2011- 11/9/2011- 12/2/2011- 12/19/2011-<br />

5/21/2012 5/21/2012 5/21/2012 5/21/2012 5/21/2012<br />

Completed questionnaire 13,600 1,564 965 319 621 17,069<br />

Indicated a family history* 4,500 255 415 71 273 5,514 (32%)<br />

Consented to participate in<br />

7,150<br />

research<br />

1,090 841 230 315 9,626 (56%)<br />

Consented to provide<br />

biospecimen<br />

4,826 868 563 196 305 6,758 (40%)<br />

Outreach sessions 9 12 0 5 6 32<br />

Providers contacted in<br />

outreach*<br />

250 39 0 38 48 375<br />

* Includes estimates<br />

Conclusion<br />

Successful implementation of the Athena risk assessment and decision<br />

support process will enable the identification of high risk women who<br />

are most likely to benefit from tailored screening or risk reducing<br />

interventions and who otherwise may not have been referred for risk<br />

reducing measures. By identifying women at the highest risk and<br />

connecting them to screening and prevention resources, the Athena<br />

<strong>Breast</strong> Health Network aims to ultimately reduce the incidence of<br />

breast cancer in its participant cohort.<br />

P4-13-14<br />

Lung cancer after treatment of breast cancer:retrospective study<br />

from Curie Institut<br />

Sebbagh S, Cosquer M, Kirova YM, Livartowski A. Institut Curie,<br />

Paris, France; Institut Curie<br />

Background: Few studies have evaluated the effects of adjuvant<br />

radiotherapy (RT)of breast cancer(BC).The relation between the risk<br />

of lung carcinoma and radiotherapy have been controversial.<br />

Methods and materials: We retrospectively studied 127 patients<br />

treated at the Institut Curie with non metastatic breast cancer and<br />

lung carcinoma between 2000 and 2011(2/3 of BC apperead befor<br />

lung).Confirmation Diagnosis bronchial cancer obtained by: biopsy:<br />

histological data:architecture, IHC (HR, HER 2, TTF1), EGFR,<br />

Kras statut clinical and radiological Correlation. Comparison with<br />

breast tumor<br />

Results: BC: median age at diagnosis 54 years, predominatly invasive<br />

ductual carcinoma(IDC), lumpectomy 78%, mastectomy 21%. Lung<br />

cancer: median age at diagnosis: 63 years, 67 smokers. histology: 52<br />

% Adenocarcinoma, 18.1 % scamous cell carcinoma , 18.1% large<br />

cell carcinoma,13.4 % small cell carcinoma.EGFR mutation in 4.3%.<br />

109 patients underwent RT (3 cases of lung cancer befor BC):Region:<br />

internal mammary chain: 46, supracalvicular: 42 , axillary: 21.<br />

Technique: lateral decubitus position: 44 , dorsal decubitus position:<br />

57. Interval between breast and lung cancer: 0-3 years: 24.4%, 3-5<br />

years: 15%, 6-10years: 16.5%, 11-20 years: 28%, >20 years: peak of<br />

incidence of lung cancer in the 3 years of diagnosis of breast cancer:<br />

24.4 %. There was no apparent relation between treatment of BC<br />

and relative risk of developing lung carcinoma. 2nd peak between<br />

11-20 years: 32 % patients, suggest that RT may increase risk of lung<br />

carcinoma (latency period for radiation induced second malignancy).<br />

Conclusion: This study suggest that adjuvant RT is associated with<br />

a real but small risk of developing lung carcinoma.<br />

P4-14-01<br />

Incidence rate of nipple areolar complex ischemia after nipple<br />

sparing mastectomy. Analysis of the American Society of <strong>Breast</strong><br />

Surgeons Nipple Sparing Mastectomy Registry.<br />

Mitchell SD, Willey SC, Feldman SM, Beitsch PD, Unzeitig GW,<br />

Manasseh DME, Neumayer LA, Laidley AL, Grutman SB, Habal N,<br />

Busch-Devereaux E, Dupont EL, Ashikari AY. White Plains Hospital<br />

Medical Center, White Plains, NY; Georgetown, Washington, DC;<br />

Columbia University College of Physicians & Surgeons, New York,<br />

NY; Dallas <strong>Breast</strong> Center, Dallas, TX; Laredo <strong>Breast</strong> Care, Laredo,<br />

TX; Maimonides Medical Center, Brooklyn, NY; University of<br />

Utah, Salt Lake City, UT; Texas <strong>Breast</strong> Specialists, Texas Oncology,<br />

Dallas, TX; American Society of <strong>Breast</strong> Surgeons, Columbia, MD;<br />

Carolina <strong>Breast</strong> & Oncologic Surgery, Greenville, NC; <strong>Breast</strong> Surgery<br />

Associates, Greenlawn, NY; Watson Clinic, Lakeland, FL; Ashikari<br />

<strong>Breast</strong> Center, Dobbs Ferry, NY<br />

Objectives:<br />

The American Society of <strong>Breast</strong> Surgeons (ASBS) Nipple Sparing<br />

Mastectomy Registry (NSMR) is a prospective, non- randomized,<br />

IRB approved multi-center registry. The ASBS NSMR has been<br />

designed to facilitate compilation of information on metrics utilized,<br />

techniques utilized, aesthetic outcomes, as well as oncologic outcomes<br />

for the nipple sparing mastectomy. The NSMR will be ongoing for 10<br />

years with an initial N of 1000. Patient selection entails all patients<br />

undergoing a NSM at the participating institutions who have agreed<br />

to participate. We evaluated the incidence rate of nipple or nipple<br />

areolar complex ischemia after a nipple sparing mastectomy.<br />

Method:<br />

The first 173 patients entered into the ASBS NSMR (2011-2012)<br />

were analyzed for incidence rate of ischemia of the nipple or nipple<br />

areolar complex (NAC). Ischemia is defined as epidermolysis (partial<br />

thickness necrosis) or full thickness necrosis. Metrics analyzed<br />

included: postoperative NAC status, indication for surgery, patient<br />

characteristics, incision utilized, sub areolar and flap dissection<br />

technique, as well as cosmetic outcomes.<br />

Results:<br />

(Thirty-five) surgeons entered 265 NSMs on 173 patients into the<br />

ASBS NSM Registry. Indications included invasive carcinoma, DCIS,<br />

and prophylaxis. Incisions utilized included radial +/- peri -areolar<br />

extension, infra mammary fold, peri- areolar ellipse/ hemi batwing,<br />

previous lumpectomy site, and previous mastopexy incisions. Sub<br />

areolar and flap dissection technique included sharp +/- tumescent<br />

injection, electrocautery, or plasma blade. Reconstruction type<br />

included tissue expander, immediate implant, or autologous flap.<br />

Median follow-up was 5 months (range 1-16 months).<br />

The nipple or NAC in 33 (12%) of 265 mastectomies experienced<br />

some degree of ischemia. Out of a total of 265 mastectomies 3 (1%)<br />

required surgical debridement, 1 (0.3%) required surgical excision,<br />

and 29 (11%) experienced epidermolysis with full recovery (17<br />

topical treatment and 12 no treatment). No correlation in incision<br />

www.aacrjournals.org 403s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

type, method of dissection, utilization of separate axillary incision for<br />

SLNBx/AXLND, size or location of tumor, type of reconstruction,<br />

initial fill volume if tissue expanders utilized, previous breast surgery,<br />

previous radiation therapy, previous chemotherapy, smoking history,<br />

cup size, degree of ptosis, or indication for surgery was found in NACs<br />

undergoing ischemia versus those that did not. Cosmetic outcome<br />

assessed by the surgeon as well as patient satisfaction did not vary<br />

from the population that did not experience nipple/NAC ischemia.<br />

Cosmetic results were rated consistently as good or excellent.<br />

Conclusion:<br />

We report a 12% incidence rate of nipple or nipple areolar complex<br />

ischemia ranging from epidermolysis to full thickness necrosis: 3<br />

(1%) requiring surgical debridement, 1 (0.3%) requiring excision,<br />

and 29 (11%) exhibiting epidermolysis with full recovery. Surgical<br />

debridement or excision was rarely necessary. Epidermolysis was<br />

found to resolve with no bearing on perceived cosmetic outcome.<br />

P4-14-02<br />

National Trends and Indications for Nipple-Sparing Mastectomy:<br />

An Analysis Using the Surveillance, Epidemiology, and End<br />

Results (SEER) Database<br />

Agarwal S, Agarwal S, Agarwal J. Wayne State University, Detroit,<br />

MI; University of Michigan, Ann Arbor, MI; University of Utah, Salt<br />

Lake City, UT<br />

Introduction: Nipple-sparing mastectomy (NSM) is an evolving<br />

procedure occasionally used for breast cancer treatment. However,<br />

indications for NSM are institution-specific and lack consensus<br />

in the literature. This study uses the Surveillance, Epidemiology,<br />

and End Results (SEER) database to investigate the oncologic and<br />

demographic characteristics of breast cancer patients treated with<br />

NSM in the United States from 2005-2009.<br />

Methods: Female breast cancer patients treated from 2005-2009 with<br />

nipple-sparing/subcutaneous mastectomy (SEER code 30), defined as<br />

removal of breast tissue with preservation of nipple-areolar complex<br />

and overlying skin, were included. Variables analyzed included<br />

patient age, race, tumor stage, tumor size, lymph node status, and<br />

radiotherapy delivery.<br />

Results: A total of 449 female breast cancer patients who underwent<br />

NSM were isolated from the SEER database. The number of patients<br />

in the SEER database who underwent NSM increased steadily each<br />

year, from 66 in 2005 to 133 in 2009. Mean age was 52 years (s.d.<br />

12 years). Breakdown by race showed that 369 (82%) patients were<br />

White women, 42 (9%) were Black women, 36 (8%) were of Asian<br />

descent. A majority of patients (402) were documented to have ductal<br />

or lobular carcinoma. Documented tumor diameter was less than 3<br />

cm in 63% (284) of patients. Invasive cancer was documented in 283<br />

(63%) patients; in situ cancer was documented in 166 (37%) patients.<br />

Lymph node involvement was negative in 374 (83%) patients.<br />

Radiation therapy was delivered to 91 (20%) patients; however, 64<br />

patients receiving radiation therapy were node-negative. Radiation<br />

was delivered to 6% (10) of patients with in situ cancer and 27% (77)<br />

of patients with invasive cancer (p < 0.0001).<br />

Conclusions: The number of NSM procedures reported in SEER for<br />

breast cancer has increased from 2005-2009. However, there is a lack<br />

of consensus in the literature as to which patients may be candidates<br />

for NSM. By utilizing a national database, we are able to aggregate<br />

and report on the national experience with therapeutic NSM, to<br />

date. Patients undergoing NSM for breast cancer were characterized<br />

predominantly by tumors less than 3 cm in diameter and with negative<br />

lymph node involvement. Patients with both invasive and in situ<br />

tumors were considered suitable candidates for NSM. A majority of<br />

patients had either ductal or lobular carcinomas. A minority of patients<br />

received radiation therapy, although there was a tendency towards<br />

providing patients with invasive tumors with radiation therapy. This<br />

national data should help guide patient selection for NSM, although<br />

future studies are needed to report on the long-term oncologic safety<br />

of this procedure.<br />

Number of Reported NSM Procedures by Year<br />

Year<br />

# of NSM Procedures<br />

2005 66<br />

2006 76<br />

2007 82<br />

2008 92<br />

2009 133<br />

P4-14-03<br />

Nipple-sparing mastectomy and intra-operative nipple biopsy:<br />

To freeze or not to freeze?<br />

Guth AA, Blechman K, Samra F, Shapiro R, Axelrod D, Choi M, Karp<br />

N, Alperovich M. NYU School of Medicine, New York, NY<br />

Background: Advances in breast cancer screening and treatment have<br />

fostered the use of surgical procedures that increasingly optimize<br />

cosmetic outcome, while ensuring oncologic safety remains the<br />

primary concern of the oncologic surgeon. The role of nipple-sparing<br />

mastectomy (NSM) for risk-reducing surgery and breast cancer is<br />

evolving. It can be difficult to demonstrate involvement of the nippleareolar<br />

complex (NAC) preoperatively, and and in this report we<br />

examine the utility of intraoperative subareolar frozen section (FS).<br />

Methods: Records of patients undergoing NSM at the NYU Langone<br />

Medical Center from 2006-2011 were reviewed retrospectively. Use<br />

of subareolar FS was at surgeon’s discretion.<br />

Results: 237 NSM were performed (146 prophylacytic, 91 theraputic).<br />

FC was not utilized in 58 mastectomies (28 prophylactic), with 2 (+)<br />

on paraffin. Among the remaining 180 mastectomies, 11 biopsies<br />

were (+)(7.2%); 5 subareolar biopsies were (+) on FS and paraffin<br />

histologic slides (PS)(2.8%); 6 were negative on FS and (+) on PS.<br />

Among the 3 prophylactic NSM with (+) subareolar biopsies there<br />

was 1 (+) FS, 1 (-) FS, and 1 with no FS performed. There were no<br />

false (+) FS. Four of the 5 patients with (+)FS underwent simultaneous<br />

excision of the NAC. The 5th patient had atypia on FS and DCIS<br />

on PS, and returned to the OR during the same hospitalization for<br />

removal of NAC. The remaining patients underwent subsequent<br />

excision of the NAC either during planned 2nd stage reconstruction<br />

or following completion of chemotherapy. 8 NAC were free of disease<br />

and 5 were positive for in situ or invasive disease. There has been no<br />

local recurrence in these patients to date.<br />

Conclusions: The rate of NAC involvement is low, 5.5% in this series,<br />

and FS, while utilized in the majority of these cases, detected only<br />

46% of subareolar disease. While FS can direct intraoperative decision<br />

making, the predictive power is low, and these considerations must be<br />

addressed in preoperative patient discussions. Furthermore, among<br />

those patients with (+) subareolar biopsies, only 39% had residual<br />

disease on NAC excision. Thus, optimal oncologic management of<br />

the NAC must still be considered an unresolved issue.<br />

P4-14-04<br />

Total skin-sparing mastectomy in BRCA mutation carriers<br />

Warren Peled A, Hwang ES, Ewing CA, Alvarado M, Esserman LJ.<br />

University of California, <strong>San</strong> Francisco; Duke University Medical<br />

Center<br />

BACKGROUND: Total skin-sparing mastectomy (TSSM) with<br />

preservation of the nipple-areolar complex skin has become<br />

increasingly accepted as an oncologically safe procedure for both<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

404s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 4<br />

prophylactic and therapeutic indications. The goal of this study was<br />

to evaluate the oncologic outcomes after TSSM in BRCA mutation<br />

carriers.<br />

METHODS: We identified 53 BRCA-positive patients who underwent<br />

bilateral TSSM for prophylactic (27 patients) or therapeutic (26<br />

patients) indications from 2001 to 2011. Cases were age-matched<br />

(for prophylactic cases) or age- and stage-matched (for therapeutic<br />

cases) with non-BRCA-positive patients who underwent bilateral<br />

TSSM during this time period. Outcomes included tumor involvement<br />

of the resected nipple tissue, the development of new breast cancers<br />

in patients who underwent bilateral risk-reducing TSSM, and the<br />

development of any local-regional recurrence in patients who<br />

underwent therapeutic TSSM.<br />

RESULTS: Outcomes from 212 TSSM procedures in 53 cases<br />

and 53 controls were analyzed. In patients undergoing TSSM for<br />

prophylactic indications, in situ cancer was found in 1 (1.9%) of the<br />

nipple specimens in the BRCA-positive patients vs. 2 specimens<br />

(3.7%) in the non-BRCA-positive cohort (p = 1). At a mean followup<br />

of 56 months, no new cancers developed in the BRCA-positive<br />

or the non-BRCA-positive cohorts. In patients undergoing TSSM for<br />

therapeutic indications, in situ or invasive cancer was found in 0 of<br />

the nipple specimens in the BRCA-positive patients vs. 2 specimens<br />

(3.9%) in the non-BRCA-positive cohort (p = 0.49). At a mean<br />

follow-up of 33 months, there were no local-regional recurrences in<br />

the BRCA-positive cohort.<br />

CONCLUSIONS: TSSM is an oncologically safe procedure in<br />

BRCA-positive patients, as is evidenced by the low rates of tumor<br />

involvement of the nipple tissue and local-regional recurrence after<br />

therapeutic mastectomy. In BRCA-positive patients undergoing<br />

TSSM as a risk-reducing strategy, five-year follow-up demonstrates<br />

no increased risk for the development of new breast cancers; longerterm<br />

follow-up is anticipated to further confirm its safety.<br />

P4-14-05<br />

Skin sparing mastectomy – thorough breast tissue removal leads<br />

to a low local recurrence rate of breast cancer<br />

Paily AJ, Drabble EH. Derriford Hopsital, Plymouth, Devon, United<br />

Kingdom<br />

Introduction<br />

Skin sparing mastectomy (SSM) has the dual advantage of being<br />

an acceptable treatment for breast cancer as well as preserving a<br />

skin envelope for better aesthesis and cosmesis. However, SSM and<br />

immediate breast reconstruction for early breast cancer is not based<br />

on level-1 evidence. Even though oncological recurrence is equivalent<br />

to that with skin removing mastectomies, most studies have only<br />

short term follow-ups. Local Recurrence (LR) or the threat of it is<br />

becoming an increasing issue for us as 1 in 4 will succumb from local<br />

recurrence. We present local recurrence (LR) and systemic recurrence<br />

(SR) rates and the aesthetic results following SSM with or without<br />

immediate reconstruction.<br />

Methods and materials<br />

Over a period of 10 years, 249 patients undergoing SSM with or<br />

without immediate reconstruction were identified from a prospectively<br />

recorded database and complete details of all their treatments and<br />

outcomes were assessed. The reconstructive methods employed<br />

included Transverse Rectus Abdominus Myocutaneous (TRAM)<br />

flap, Latissimus Dorsi (LD) flap in isolation or along with an implant<br />

and subpectoral implant insertion. LR and SR rates were determined.<br />

Pathological examination of biopsies from the skin flaps after SSM,<br />

were also analysed.<br />

Results<br />

Of the 249 patients who underwent SSM, only 34 did not have<br />

immediate breast reconstruction. The overall LR and SR rates<br />

were 0.4% and 4.4% respectively. 76 patients underwent SSM and<br />

TRAM flap reconstruction (1 LR and 5 SR). 33 patients had LD flap<br />

reconstruction (no LR and 1 SR). 61 patients were given LD flap<br />

reconstruction with implants (no LR and 4 SR). SSM and insertion<br />

of sub-pectoral implants was performed on 45 patients (no LR and no<br />

SR). 34 patients underwent SSM without immediate reconstruction<br />

(no LR, 1 SR). All 249 patients had no evidence of residual breast<br />

tissue in subcutaneous flap biopsies. Of the 215 patients who<br />

underwent immediate reconstruction, 140 had a good aesthetic result<br />

(65%). Individually, TRAM flap and LD flap with implant insertion<br />

each produced the best aesthetic result (74%). While reconstructions<br />

with LD flaps alone did result in a good aesthetic result in 70% of<br />

patients, it also had the poorest aesthetic result (12%) compared to<br />

the other three groups.<br />

Conclusion<br />

SSM with or without immediate breast reconstruction performed in<br />

our unit has shown a very low rate of LR compared to that in literature.<br />

In addition, flap biopsies have shown no residual breast tissue in our<br />

patients highlighting that thorough removal of breast tissue at skin<br />

sparing mastectomy leads to low LR rates. Our systemic recurrence<br />

rates are much higher than local recurrence rates, but are still as<br />

expected or slightly lower than expected. Our approach therefore<br />

achieves thorough removal of breast tissue and we believe that this<br />

leads to a low local recurrence rate, but with good aesthetic results.<br />

P4-14-06<br />

Withdrawn by Author<br />

P4-14-07<br />

Impact of preservation of the intercostobrachial nerve during<br />

axillary dissection on sensory change and health-related quality<br />

of life two years after breast cancer surgery<br />

Taira N, Shimozuma K, Ohsumi S, Kuroi K, Shiroiwa T, Watanabe<br />

T, Saito M. Okayama University Hospital, Okayama, Japan;<br />

Ritsumeikan University, Japan; National Shikoku <strong>Cancer</strong> Center,<br />

Japan; Tokyo Metropolitan <strong>Cancer</strong> and Infectious Diseases Center<br />

Komagome Hospital, Japan; Teikyo University, Japan; Sendai<br />

Medical Center, Japan; Juntendo University, Japan<br />

Background: Sensory loss or paresthesia due to division of the<br />

intercostobrachial nerve (ICBN) is a complication of axillary lymph<br />

node dissection (ALND). Preservation of the ICBN may be of value,<br />

but few prospective studies have shown an impact of preservation<br />

on sensory changes or health related quality of life (HRQOL) after<br />

breast cancer surgery.<br />

Methods: Prospective study was performed to evaluate the association<br />

of ICBN preservation with sensory change and HRQOL at 1<br />

(baseline), 6, 12 and 24 months after breast cancer surgery in 140<br />

patients. The sensory examination included dysesthesia, paresthesia,<br />

abnormal touch and pain sensation in the upper arm.<br />

Results: Division of the ICBN did not influence the frequency or<br />

severity of subjective dysesthesia and paresthesia. There was no<br />

marked difference in touch or pain sensation at baseline between<br />

patients with a preserved (group P) and divided (group D) ICBN. In<br />

group P, the percentage of patients aware of a sensory deficit or loss<br />

decreased with time, and that of patients aware of a hypersensitive<br />

sensation increased. These changes did not occur in group D, leading<br />

to a significant difference between the groups at 24 months. The main<br />

difference between the groups was the area with reduced touch or<br />

www.aacrjournals.org 405s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

pain sensation. This area decreased with time in group P, but not in<br />

group D. ICBN preservation or division did not influence HRQOL.<br />

Conclusion: ICBN preservation in ALND has a benefit of a reduced<br />

area with long-term axillary hypoesthesia, but has no influence on<br />

improvement of pain and HRQOL.<br />

P4-14-08<br />

Intraoperative ultrasound guidance for excision of non-palpable<br />

breast cancer<br />

Barentsz MW, van Dalen T, Gobardhan PD, Bongers V, Perre CI,<br />

Pijnappel RM, van den Bosch MA, Verkooijen HM. University Medical<br />

Center Utrecht, Netherlands; Diakonessenhuis, Utrecht, Netherlands;<br />

Amphia Hospital, Breda, Netherlands<br />

Background<br />

The effectiveness of intraoperative ultrasound (IOUS) for<br />

preoperative localization of non-palpable breast cancers within the<br />

operation theatre has not been studied extensively. In this prospective<br />

cohort study, we compared margin status, re-excision rate and excised<br />

volume of IOUS to guidewire localization (GWL).<br />

Methods<br />

A total of 258 consecutive patients with non-palpable invasive breast<br />

cancer underwent breast conserving surgery between 1999-2010.<br />

GWL was performed in 138 (54%) and IOUS in 120 (46%) patients.<br />

Tumour dimensions, resection volume, margin status and re-excision<br />

rates were compared by means of multivariate regression analysis.<br />

Calculated resection ratios, i.e. indicating the amount of excess tissue<br />

resection, were calculated by dividing the total resection volume<br />

by the optimal resection volume (the tumor diameter plus a 1.0 cm<br />

margin) and compared between the groups.<br />

Results<br />

The groups were similar in terms of age, histological subtype and<br />

presence of DCIS. Lesions in the IOUS group were larger (1.24 vs.<br />

0.98 cm), while lesions in the GWL group consisted more often of<br />

microcalcifications only (19% vs. 3%). Tumour free resection margins<br />

were obtained in more than 93% of patients (93.5% with GWL vs.<br />

93.3% with IOUS, P = .958) and re-excision was performed in 11.0%<br />

of patients undergoing GWL and 12.5% of patients undergoing IOUS<br />

(P = .684). In both groups, resection volumes were similar, but IOUS<br />

led to more optimal tissue resection (calculated resection ratio 4.33<br />

vs 3.30, P = .018). After adjustment for tumor diameter, radiological<br />

findings and presence of DCIS, the difference in calculated resection<br />

volumes was no longer significant.<br />

Conclusion<br />

For localization of non-palpable breast cancer, IOUS is a reliable<br />

alternative to GWL, as it achieves similar results in terms of complete<br />

tumour removal, re-excision rate and excised volume.<br />

P4-14-09<br />

Feasibility of liposuction for treatment of arm lymphedema from<br />

breast cancer.<br />

Doren EL, Smith PD, Sun W, Lacevic M, Fulp W, Reid R, Laronga<br />

C. University of South Florida, Tampa, FL; Moffitt <strong>Cancer</strong> Center,<br />

Tampa, FL<br />

Background: Lymphedema is a dreaded complication of breast cancer<br />

treatment affecting 20% of women having axillary node dissection.<br />

Liposuction minimizes unwanted fat in targeted areas. Our objective<br />

was to explore the feasibility of liposuction to reduce fat volume and<br />

thus arm lymphedema.<br />

Methods: An IRB-approved prospective trial was conducted of<br />

women having unilateral arm lymphedema resulting from breast<br />

cancer treatment. At enrollment there was no evidence of cancer<br />

recurrence or arm cellulitis. Arm measurements (circumferential),<br />

volumes (water displacement and geometric calculation), and muscle<br />

strength differences between the affected and unaffected arms and<br />

quality of life/functionality were measured pre-operatively and<br />

post-operatively at 6 weeks, 6 months and one year(s). Descriptive<br />

statistical analysis was performed.<br />

Results: Six breast cancer survivors underwent the liposuction<br />

procedure from 12/2008- 4/2011. Median age was 54 yrs (range: 43-<br />

60) and median volume of fat aspirated was 700mls (range: 350-700).<br />

Average volume difference between the affected and unaffected arms<br />

at baseline was 522.5 mls (176-867) (geometric) and 589.2mls (280-<br />

770) (water displacement). No immediate complications; 1 cellulitis<br />

at 4 months post-operative. Average percent volume reductions for<br />

5 of the 6 women at 6 weeks, 6 months and 1 year were 70%, 47%,<br />

71% mls geometrically and 63%, 18%, 54% by water displacement<br />

respectively. Quality of life and functionality improved in all patients.<br />

Muscle strength remained unchanged. Pain lessened. Average followup<br />

is 15.49 months (range: 1.8-24.84 months).<br />

Conclusion: Liposuction can safely reduce volume of arm<br />

lymphedema and improve functionality/quality of life. Larger studies<br />

(longer follow-up) are required to validate the durability of these<br />

early results.<br />

P4-14-10<br />

Atypical Ductal Hyperplasia diagnosed on directional vacuumassisted<br />

biopsy: is surgical excision mandatory?<br />

Chauvet M-P, Rivaux G, Farre I, Houpeau J-L, Giard S, Ceugnart<br />

L. Centre Oscar Lambret, Lille, France<br />

Background: The incidence of atypical ductal hyperplasia (ADH) is<br />

increasing due to mass screening. Because of the underestimation risk<br />

of malignancy, the management remains controversial. Our goal was<br />

to analyze clinicopathologic features of patients with ADH diagnosed<br />

on directional vacuum-assisted biopsy (DVAB). The objectives of this<br />

continuous retrospective study were to evaluate the underestimation<br />

rate of malignancy and to identify predictors of upgrade to carcinoma.<br />

Methods: Between 2003 and 2010, 3159 patients underwent<br />

stereotactic DVAB in our institute. We retrospectively evaluate<br />

clinical, mammographic and pathological features of 298 cases of<br />

ADH who underwent surgical excision in our center (93.1%). Patients<br />

with concurrent history of breast cancer, intraductal carcinoma (DCIS)<br />

associated, or with no follow-up or surgical excision on place were<br />

excluded. Histological scar of macrobiopsy was systematically<br />

searched in surgical specimens. A pathologic upgrade was defined by<br />

presence of invasive cancer or DCIS on surgical specimen. Statistical<br />

tests used were the chi-square or Fisher’s exact test.<br />

Results: Among the 298 studied DVAB, 224 ADH were isolated<br />

(75.2%), 46 associated to flat epithelial atypia (15.4%) and 28 to<br />

lobular neoplasia (9.4%). 98.4% patients presented microcalcifications<br />

at diagnosis. In 52 cases, lesions were upgraded to DCIS (n=38) or<br />

invasive cancer (n=14). The underestimated rate was 17.5%. In 67.3%<br />

cases, upgrade lesions were low or intermediate grade DCIS. Only the<br />

history of contralateral breast cancer was significantly correlated with<br />

the underestimated rate (p=0.04). There was no statistical difference<br />

between these 52 cases and the 246 others for: family history, size<br />

of calcification, sampling number, histological lesion, and quality of<br />

calcifications removal.<br />

Conclusions: In this study, DVAB may not be considered a therapeutic<br />

procedure in case of ADH, even in the case of complete removal of<br />

microcalcifications. It is still challenging to identify a subgroup of<br />

ADH cases with a low upgrade rate.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

406s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 4<br />

P4-14-11<br />

Risk reducing mastectomy for women with high personal breast<br />

cancer risk<br />

Onyekwelu O, Shetty G, Baildam A. Nightingale & Genesis <strong>Breast</strong><br />

<strong>Cancer</strong> Prevention Centre, University Hospital of South Manchester,<br />

Wythenshawe, Manchester, United Kingdom; St. Bartholomew’s<br />

Hospital (Bart’s), West Smithfield, London, United Kingdom<br />

Introduction<br />

For high-risk familial history of breast cancer, risk reducing<br />

mastectomy (RRM) is now regarded as one of the management<br />

options. The aim of this study is to analyse the outcome of women<br />

undergoing RRM from 1994 to 2010 during which surgical techniques<br />

evolved and were refined.<br />

Methods<br />

A cohort of women undergoing RRM was followed from this single<br />

centre. Surgical and further details about demographics, comorbidities,<br />

and follow-up were retrieved form medical records. Analysis was<br />

carried out using the IBM®-SPSS®-Statistics-19system.<br />

Results<br />

Of 154 women 139 had bilateral RRM and 15 had unilateral surgery<br />

after earlier contralateral breast cancer. Median age at operation was<br />

39.4 years (22-63years). 54 (45.5%) women were BRCA1/2 gene<br />

mutation carriers. Most women had nipple sacrificing (86) skinsparing<br />

mastectomy with immediate reconstruction. 132 (85.7%) had<br />

submuscular expander/implant reconstruction, whilst 9 had either LD<br />

flap reconstruction or TRAM flap reconstruction.<br />

31 (20.1%) women had some complication including haematoma<br />

requiring evacuation, minor skin/nipple necrosis, but implant loss<br />

was rare (2). 52 (33.8%) women had revisional surgeries in 5 years<br />

post RRM of whom 29 had one unanticipated secondary operation.<br />

Complications were more common in the early years of the cohort.<br />

On histology, 3 women were identified with lobular carcinoma in-situ<br />

and one was associated with 1mm of microinvasion. 4 had ductal<br />

carcinoma in-situ and 2 of them were BRCA carriers. Two (1.2%)<br />

primary breast cancers were subsequently identified during a mean<br />

follow-up period of 9.82 years (2-18 years) but this was substantially<br />

less than prediction modelling. Both were BRCA gene mutation<br />

carriers - one had nipple sparing RRM and the other did not.<br />

Conclusions<br />

In asymptomatic high-risk women, RRM is safe and efficacious in<br />

reducing medium term future breast cancer incidence. Surgery carries<br />

the risk of complications and cosmetic revisions can be anticipated.<br />

P4-14-12<br />

Evaluation of the effect of pasireotide LAR administration in<br />

the lymphocele prevention after mastectomy with axillary lymph<br />

node dissection for breast cancer: results of a phase 2 randomized<br />

study.<br />

Chereau E, Uzan C, Zohar S, Bezu C, Mazouni C, Ballester M, Gouy<br />

S, Rimareix F, Garbay J-R, Darai E, Uzan S, Rouzier R. Tenon,<br />

APHP, UPMC - Paris 6, Paris, France; Institut Gustave Roussy,<br />

Villejuif, France; Hopital Saint-Louis, APHP, U444-INSERM, Paris<br />

7, Paris, France<br />

Background: Lymphocele is the principal post-operative morbidity<br />

following axillary node dissection. According to the literature, the<br />

incidence can vary from 4% to 89%. Encouraging results in terms<br />

of reducing postoperative lymphoceles as well as the volume and<br />

duration of drainage using octreotide LAR has been recently reported.<br />

Pasireotide LAR, a long acting drug designed to target multiple<br />

somatostatin receptors, was evaluated in this trial.<br />

Trial design: A phase II, two centers, randomized, double-blind,<br />

non-comparative pilot study was carried out in order to evaluate<br />

efficacy and safety of a single injection of pasireotide LAR 60 mg<br />

administered 7-10 days before scheduled mastectomy with axillary<br />

dissection surgery. This study included a parallel placebo arm to<br />

assess the natural course of the disease.<br />

Eligibility criteria: Adult female breast cancer patients planned to<br />

undergo a mastectomy (without reconstruction at the same time) and<br />

axillary node dissection .<br />

Specific aims: To assess the efficacy and safety of a single injection<br />

of pasireotide LAR 60 mg or placebo prior to mastectomy with<br />

axillary lymph node dissection surgery in reducing symptomatic<br />

lymphocele development. Symptomatic lymphocele was evaluated<br />

and was defined as: 1. total lymphocele drainage/aspiration volume<br />

(unique or iterative) >60 cc inclusive within the 28 days after surgery<br />

(excluding post-surgery drain) or; 2. a systematic aspiration volume<br />

at day 28 > 120 cc.<br />

Statistical methods: The statistical analysis was carried out<br />

sequentially after observing the absence of symptomatic lymphocele<br />

for each patient. It involves estimating the probability of a response in<br />

each group using a Bayesian design based on a beta-binomial model.<br />

The probability of response was considered random and its prior<br />

distribution was centered on 80% in the pasireotide group and 60%<br />

in the placebo group according to the investigators initial guesses.<br />

The distribution of the probability of response was updated after the<br />

observation of the patients included in the trial.<br />

Results: A total of 90 patients were included over 18 months: 42 in<br />

the treatment group and 48 in the placebo group. In the treatment<br />

group, the posterior mean estimation of the response rate (i.e. patients<br />

who did not experience a symptomatic lymphocele) was 62.4%<br />

(95% CI: 48.6%-75.3%) and 50.2% in the placebo group (95% CI:<br />

37.6%-62.8%%). In the treatment group, one serious adverse event<br />

occurred in a patient with known insulin dependent diabetes requiring<br />

hospitalization for hyperglycaemia.<br />

Conclusion: A one time injection of pasireotide LAR to prevent<br />

symptomatic lymphocele development in women undergoing<br />

mastectomy with axillary dissection is promising. Further clinical<br />

studies are warranted.<br />

P4-14-13<br />

Therapeutic mammaplasty does not cause a delay in the delivery<br />

of chemotherapy in high risk breast cancer patients<br />

Khan J, Barrett S, Stallard S, Forte C, Weiler-Mithoff E, Reid I,<br />

Winter A, Doughty J, Romics L. Victoria Infirmary, Glasgow, United<br />

Kingdom; Beatson West of Scotland <strong>Cancer</strong> Centre, Glasgow, United<br />

Kingdom; Western Infirmary, Glasgow, United Kingdom; Royal<br />

Infirmary, Glasgow, United Kingdom<br />

Introduction: oncosurgical safety of therapeutic mammaplasty (TM)<br />

is widely investigated. The interval between surgery and delivery<br />

of adjuvant chemotherapy is an integral part of overall oncological<br />

safety. Therefore, we examined the time between TM and AC, and<br />

compared it to wide local excision (WLE) and mastectomy (Mx)<br />

with or without immediate breast reconstruction (IBR), respectively.<br />

Methods: data of 174 patients who underwent TM, WLE and Mx±IBR<br />

was analyzed retrospectively. All patients were operated within three<br />

breast units of Glasgow during a period of 48 months. Time between<br />

decision to offer adjuvant chemotherpay and delivery of the first<br />

cycle of chemotherapy was analyzed. Significance was calculated<br />

with Mann-Whitney and Kruskal-Wallis tests (two and four groups<br />

compared, respectively).<br />

www.aacrjournals.org 407s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Results: median time to adjuvant chemotherapy after TM (n=36)<br />

was 29 [16-58] days, WLE (n=66) was 29.5 [15-105], Mx only<br />

(n=56) was 29 [15-57], and Mx and IBR (n=16) was 31 [15-58]<br />

days. No significant difference was found in terms of time to adjuvant<br />

chemotherpay in patients treated with TM compared to WLE<br />

(p=0.384), Mx only (p=0.828) or Mx and IBR (p=0.366). Further,<br />

there was no significant difference when a cumulative comparison<br />

of the four groups was carried out (p=0.507).<br />

Conclusions: our data indicate that oncosurgical safety of TM in terms<br />

of time to chemotherapy is similar to other high risk breast cancer<br />

patients treated WLE and Mx with or without IBR. This also suggests<br />

that there is no significant difference in postoperative complication<br />

rates after these four ways of surgical treatment of breast cancer,<br />

which would possibly be the primary cause for a delay in delivering<br />

adjuvant chemotherapy.<br />

P4-14-14<br />

Phyllodes tumour of the breast: A retrospective analysis of 87<br />

cases<br />

Walter HS, Esmail F, Krupa J, Ahmed SI. Leicester Royal Infirmary,<br />

Leicester, United Kingdom; Glenfield Hospital, Leicester, United<br />

Kingdom<br />

Background: Phyllodes tumour is a rare fibroepithelial tumour of the<br />

breast that accounts for only 0.5% of all breast neoplasms.<br />

Methods: We retrospectively reviewed the case notes of all patients<br />

with phyllodes tumour treated between 1987-2010. Clinical data was<br />

analysed with respect to pathological and treatment outcome data.<br />

Results: Between 1987-2010, 87 patients were treated for tumours,<br />

histologically classified as benign (60), borderline (10) and malignant<br />

(17). Median age at presentation was 55 years for patients with<br />

malignant phyllodes, 45 years for benign phyllodes and 48 years for<br />

patients with borderline tumours. Primary surgery for patients with<br />

benign or borderline phyllodes in 68 cases was breast conserving with<br />

2 patients undergoing mastectomy. Of those with malignant phyllodes,<br />

primary surgery for 6 patients was mastectomy and 11 patients had<br />

breast conserving surgery. 2 patients with malignant phyllodes<br />

received adjuvant radiotherapy, to the chest wall for recurrence<br />

following mastectomy in one patient and following local excision<br />

in the other patient. 2 patients with malignant phyllodes proceeded<br />

to axillary surgery, with no involvement of lymph nodes. 6 patients<br />

(8.6 %) with benign disease and 4 (23.5 %) with malignant phyllodes<br />

had a local recurrence. Of those with malignant phyllodes who had a<br />

recurrence, 2 had systemic metastases and all had had a local excision<br />

at presentation. Median time to development of metastatic disease<br />

from initial presentation was 2.5 years. 1 patient received 1 cycle of<br />

palliative doxorubicin but progressed through treatment. Relapse free<br />

survival to date for patients with benign, borderline and malignant<br />

phyllodes was 91.7 %, 90 % and 70.6 % respectively.<br />

Conclusion: Phyllodes tumours form a heterogenous group with<br />

a spectrum of behaviour in terms of risk of local recurrence and<br />

metastases. From this series, malignant phyllodes have a higher<br />

recurrence rate than benign and borderline tumours. Local excision in<br />

this group was associated with a 36.4 % local failure rate and 18.2 %<br />

risk of systemic metastasis. Treatment for malignant phyllodes should<br />

be individually based, but the greater risk of relapse both locally and<br />

systemically, should be taken into consideration and the options of<br />

mastectomy or oncoplastic techniques considered.<br />

P4-15-01<br />

Second conservative treatment for ipsilateral breast tumor<br />

recurrence: GEC-ESTRO <strong>Breast</strong> WG study<br />

Hannoun-Levi J-M, Resch A, Gal J, Niehoff P, Loessl K, Kovács G,<br />

Van Limbergen E, Polgar C. Antoine Lacassagne <strong>Cancer</strong> Center,<br />

Univerity of Nice-Sophia, Nice, France; Medical University, General<br />

Hospital of Vienna, Austria; Antoine Lacassagne <strong>Cancer</strong> Center,<br />

Nice, France; University Erlangen-Nuremberg, Erlangen, Germany;<br />

University Hospital Bernes, Switzerland; University Hospital<br />

Schleswig-Holstein Campus Lübeck, Germany; University Hospital<br />

Gasthuisberg, Leuven, Belgium; National Institute of Oncology,<br />

Budapest, Hungary<br />

For ipsilateral breast cancer tumor (IBTR), radical mastectomy<br />

represents the treatment option frequently proposed to the patient.<br />

A second conservative treatment (SCT) has been proposed using<br />

either lumpectomy alone or associated with a second irradiation of<br />

the tumor bed. However, in both clinical situations, the proof level<br />

of such therapeutic approaches remains low, based on cased-series<br />

or retrospective studies.<br />

To analyze the clinical outcome of a SCT using lumpectomy and<br />

multicatheter interstitial brachytherapy (MIB) for IBTR, the GEC-<br />

ESTRO <strong>Breast</strong> Working Group retrospectively analyzed the results of<br />

217 patients (pts) with an IBTR treated between 09/00 and 09/10 in<br />

8 European institutions by lumpectomy and MIB (low - LDR, pulse<br />

- PDR, or high-dose rate - HDR). Survival rates without 2nd local<br />

(2nd LR) and metastatic recurrence, disease free survival (DFS) and<br />

specific and overall survivals were analyzed as well as late effects<br />

and cosmetic results. Dosimetric data were reported according to the<br />

dose rate used. Univariate and multivariate analysis were performed<br />

to find local, metastatic and/or DFS progression prognostic factors.<br />

With a median follow-up of 14.5 years [3.5-38.2] and 3.9 years [1.1-<br />

10.3] from primary tumor and IBTR respectively and a median time<br />

interval of 9.4 years [1.1-35.4] between primary and IBTR, 20.7%<br />

of the local recurrence were observed at distance from the primary<br />

tumor. Median tumor sizes were 15 mm [1-60] and 11 mm [1-40]<br />

for the primary and IBTR respectively. Thirty-nine percent of the<br />

patient underwent an axillary lymph node dissection at the time of<br />

IBTR. Median radiotherapy dose for the primary was 56 Gy [30-<br />

69.6]. Positive hormonal receptor status for IBCR was 72.8% while<br />

65% and 19.8% received hormonal and chemotherapy respectively<br />

as adjuvant therapy for the IBTR. Five and 10-year actuarial 2nd LR<br />

rates were 5.6% [1.5-9.5] and 7.2% [2.1-12.1] respectively. Five and<br />

10-year actuarial metastatic recurrence rates were 9.6% [5.7-15.2]<br />

and 19.1% [7.8-28.3] respectively. Five and 10-year actuarial DFS<br />

rates were 84.6% [78.9-90.6] and 77.2% [67.5-88.3] respectively. Five<br />

and 10-year actuarial overall/specific survival (OS) rates were 88.7%<br />

[83.1-94.8] and 76.4% [66.9-87.3] respectively. 141 pts developed<br />

193 complications. Fibrosis was the most frequent complication with<br />

11% of G3-4 complications. Cosmetic result was jugged as excellent/<br />

good in 85%. Focusing on multivariate analysis, prognostic factors<br />

for 2nd LR, metastatic recurrence and DFS are reported.<br />

MVA results<br />

IBTR 2nd LR Distant mets DFS<br />

Age 0.09 - -<br />

Grade 0.02 - -<br />

pT - 0.01 0.001<br />

HR - - 0.003<br />

pT: tumor size; HR: hormone status<br />

The results of this study suggest that in case of IBTR, a SCT<br />

combining lumpectomy plus MIB is feasible with an overall survival<br />

rate at least equivalent to those obtain after salvage mastectomy. The<br />

rate of complication remains acceptable with encouraging cosmetic<br />

results.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

408s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 4<br />

The literature analysis suggests that the rate of 2nd LR is 10% [3-<br />

32], 25% [7-36] and 10% [2-26], after salvage mastectomy, salvage<br />

lumpectomy alone or combined with a second irradiation respectively.<br />

However, the 5-y OS rates after salvage mastectomy and SCT seem<br />

to be equivalent (75%) mainly influenced by distant metastatic<br />

progression.<br />

P4-15-02<br />

Timing of infectious complications following breast conserving<br />

therapy with catheter-based accelerated partial breast irradiation<br />

Haynes AB, Bloom ES, Bedrosian I, Kuerer HM, Hwang RF, Caudle<br />

AS, Hunt KK, Graviss L, Chemaly RF, Tereffe W, Shaitelman SF,<br />

Babiera GV. University of Texas MD Anderson <strong>Cancer</strong> Center,<br />

Houston, TX<br />

Background:<br />

Accelerated partial breast irradiation (APBI) has been introduced<br />

as an alternative to whole breast irradiation as part of breast<br />

conserving therapy for selected patients. The long-term outcomes<br />

remain under investigation. Previous publications have emphasized<br />

early postoperative infections with APBI with less focus on delayed<br />

infection. In the current study, we evaluated patients enrolled on a<br />

prospective registry trial for infectious complications after treatment<br />

with catheter-based APBI.<br />

Methods:<br />

Patients who underwent single-entry catheter-based APBI were<br />

identified from a single-institution prospective registry from 2009 to<br />

2011. Data regarding treatment, patient comorbidities, complications,<br />

and outcomes were obtained from registry and retrospective chart<br />

review. Infectious complications were assessed from the date of APBI<br />

catheter insertion and were classified as early (≤30 days) or delayed<br />

(>30 days). All patients were maintained on oral antibiotics while<br />

the catheter was in place.<br />

Results:<br />

A total of 91 patients with 92 cases of primary breast cancer were<br />

enrolled on a prospective registry at a comprehensive cancer center<br />

between 2009 and 2011 and treated with single-entry catheter-based<br />

APBI. The median follow-up time was 76.2 weeks. A temporary<br />

catheter was placed at the time of initial operation in 40 cases (43.5%)<br />

and left in place a median of 6 days prior to definitive catheter<br />

insertion. There were 20 patients (21.7%) who required re-excision.<br />

Overall, breast infection occurred in 13 (14.1%) patients. Three (3.3%)<br />

patients had infection within 30 days of catheter placement and 10<br />

(10.9%) occurred more than 30 days after catheter insertion (median<br />

112.5 days, interquartile range 51-154). Eight patients were managed<br />

with oral antibiotics alone on an outpatient basis. The remainder<br />

required a combination of admission, intravenous antibiotics, and<br />

aspiration of abscess. One patient underwent operative drainage.<br />

Conclusion:<br />

We found an overall infection rate of 14.1% in patients treated with<br />

catheter-based APBI. This is consistent with other reports; however,<br />

we found that the majority of infections occurred more than 30<br />

days after definitive catheter placement. Vigilance for infectious<br />

complications must continue beyond the immediate treatment<br />

period in patients undergoing catheter-based APBI. Most infections<br />

following APBI can be managed on an outpatient basis without<br />

operative intervention.<br />

P4-15-03<br />

Patterns of relapse following re-irradiation of the breast using<br />

partial breast brachytherapy (PBB)<br />

Chadha M, Boachie-Adjei K, Boolbol SK, Kirstein L, Osborne MP,<br />

Tarter P, Harrison LB. Beth Israel Medical Center, New York, NY<br />

Purpose: This study is undertaken to evaluate the patterns of relapse<br />

among patients with a prior history of irradiation where the RT field<br />

included the breast, and who received re-irradiation using PBB for<br />

a localized second cancer event in the breast using partial breast.<br />

Materials and Methods: Twenty-seven patients were enrolled in an<br />

IRB approved study. Nineteen patients had prior history of breast<br />

cancer treated with lumpectomy and external beam therapy (ERT),<br />

and 8 patients had prior history of mantle RT. All patients underwent<br />

lumpectomy with negative margins and received a median dose of<br />

45Gy PBB. The median time interval between the primary breast<br />

cancer diagnosis and the second cancer event in the ipsilateral breast<br />

is 94-months (range 28-months to 211-months). The median time<br />

between mantle RT and the breast cancer is 245 months.<br />

Results: At a median follow up of 73 months following lumpectomy<br />

and PBB 14.8 % (4/27) patients developed a local recurrence and were<br />

salvaged by mastectomy. The 5-year Kaplan Meier local disease-free<br />

survival and mastectomy-free survival is 100% and 81%, respectively.<br />

Remarkably, one third of patients with prior history of mantle RT<br />

developed third primary cancers (pancreas, lung and renal). All<br />

patients with the third primary cancers died free of any relapse from<br />

the breast cancer. None of the patients with prior history of breast<br />

cancer developed a second malignancy, and 3/21 developed distant<br />

metastases.<br />

Conclusions: In appropriately selected patients with prior history<br />

of lumpectomy and ERT for breast cancer we observed a high rate<br />

of local control and freedom from mastectomy. Patterns of relapse<br />

among patients with prior mantle RT suggest that breast conservation<br />

and PBB was appropriate for treating early stage disease. <strong>Breast</strong><br />

conservation therapy did not have a detrimental effect on breast cancer<br />

specific outcome and eventual patient survival. Patients motivated<br />

for breast conservation may have an alternative to mastectomy at<br />

initial diagnosis.<br />

P4-15-04<br />

Impact of awake brest cancer surgery on postoperative<br />

lymphocyte responses<br />

Vanni G, De Felice V, Buonomo C, Esser A, Petrella G, Buonomo<br />

OC. Tor Vergata University, Rome, Italy<br />

Background. Surgical stress and general anesthesia can have a<br />

detrimental effects on postoperative immune system. We sought<br />

to evaluate comparatively postoperative lymphocytes response in<br />

patients undergoing quadrantectomy and lymph node sentinel biopsy<br />

(LNSB) under local or general anesthesia.<br />

Methods. Between December 2011 and May 2012, 50 patients<br />

with breast cancer were randomized by computer to undergo<br />

quadrantectomy and LNSB under local anesthesia (awake group,<br />

n=25) or general anesthesia (control group, n=25). In both groups,<br />

assessment of total lymphocytes count and changes in proportion of<br />

specific lymphocyte-subsets including CD19+, CD3+, CD4+/CD8+<br />

ratio, CD4+, CD8+ and CD16+CD56+ (Natural-killer) were evaluated<br />

at baseline and on postoperative day 1, 2 and 3.<br />

Results. Study groups were well matched in baseline and surgical<br />

data. On postoperative day 1, a significantly higher amount of<br />

Natural-Killer proportion was found in the awake group versus the<br />

control group (median: 12 Vs 4, P=0.002); this difference was still<br />

www.aacrjournals.org 409s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

significant on postoperative day 2 (median: 11 Vs 7, P=0.02). Yet, in<br />

awake group there was a higher CD4+/CD8+ ratio on postoperative<br />

day 1 (median: 2.6 Vs 1.6, P=0.05). No difference was found among<br />

groups in the remaining lymphocyte subsets. On postoperative day<br />

3, total lymphocytes count was significantly decreased from baseline<br />

value in the control group only (median-∆: 460/mm3, P=0.01) while<br />

it remained unchanged in the awake group.<br />

Conclusions. In this study, quadrantectomy and LNSB under local<br />

anesthesia resulted in a lesser impact on postoperative lymphocyte<br />

responses when compared to the equivalent procedure performed<br />

under general anesthesia.<br />

P4-15-05<br />

Long-term outcome of breast cancer patients treated with<br />

radiofrequency ablation<br />

Earashi M, Noguchi M, Motoyoshi A, Komiya H, Fujii H. Yatsuo<br />

General Hospital, Toyama, Japan; Kanazawa Medical University<br />

Hospital, Uchinada-Daigaku, Ishikawa, Japan<br />

Background: Radiofrequency ablation (RFA) is considered to be the<br />

most promising non-surgical ablation technique for the treatment<br />

of small breast cancer. In feasibility studies, the ablated tumor was<br />

surgically removed after RFA, and the tumor cell viability were<br />

assessed by histological and/or immunohistological examinations.<br />

However, these assessments of tumor viability do not take the place<br />

of long-term follow-up in patients treated with RFA alone, because<br />

they do not allow for them to be followed up for recurrence after<br />

RFA, or to determine if there were any undesirable complications of<br />

this procedure. At present, few data are available regarding long-term<br />

follow-up of patients treated with this modality.<br />

Methods: Since 2005, we have performed RFA and sentinel lymph<br />

node (SLN) biopsy in 19 cases with invasive breast cancer less than<br />

2.0 cm in greatest diameter. After SLN biopsy, a primary electrode<br />

(seven-array model 70 Starburst needle electrode; RITA Medical<br />

Systems, Mountain View, CA) was inserted into the tumor under<br />

real-time US guidance. Then, the prongs of the needle electrode<br />

were deployed over a distance of 3 cm in all cases, and the RF<br />

generator (RITA model 1500) was activated and set to automatic,<br />

with power at 20 W, temperature of 95°C, and an ablation time of<br />

15 min. Axillary lymph node dissection (ALND) was performed in<br />

patients with positive SLNs. Several months after RFA therapy, the<br />

ablated tumor tissue was excised by multiple mammotome biopsy<br />

and examined histologically or immunohistochemically with H&E<br />

staining, nicotinamide adenine dinucleotide (NADH)-diaphorase<br />

staining, and single-stranded (ss) DNA staining. All cases were<br />

followed-up after breast radiation and systemic therapies.<br />

Results: The mean tumor size based on the ultrasonographic maximum<br />

dimension was 1.3 cm (range: 0.5 – 2.0 cm). Although complete<br />

response was histologically confirmed in only 8 cases, NADHdiaphorase<br />

and ssDNA staining did not demonstrate any viable tumor<br />

cells in any of the ablated lesions. At a mean follow-up of 60 months<br />

(follow-up range, 37 – 82 months), there were no cases of in-breast<br />

recurrence, although one patient died due to hepatic metastases.<br />

Cosmesis of the conserved breast was excellent or good in all of the<br />

cases, but a hard lump was persistent after RFA in half of the cases.<br />

Conclusions: The long-term outcome of patients treated with RFA<br />

is encouraging with regard to cosmesis and local control. However,<br />

a persisted lump can cause patient discomfort, anxiety and fear.<br />

Therefore, further studies are needed to establish the optimal<br />

technique. Moreover, a prospective study will be required to determine<br />

the equivalency in local recurrence rates between the RFA therapy<br />

and conventional breast-conserving treatment.<br />

P4-16-01<br />

Accelerated hypofractionated whole breast radiotherapy for<br />

localized breast cancer: the effect of a boost on patient reported<br />

long-term cosmetic outcome<br />

Chan EK, Tabarsi N, Tyldesley S, Khan M, Woods R, Speers C, Weir<br />

L. British Columbia <strong>Cancer</strong> Agency, Vancouver Centre, Vancouver,<br />

BC, Canada; University of British Columbia, Vancouver, BC, Canada<br />

PURPOSE<br />

Equivalent long-term local control and cosmetic outcomes between<br />

conventional and accelerated, hypofractionated whole breast<br />

radiotherapy (AWBRT) for early-stage breast cancer have been<br />

demonstrated. However, there is uncertainty about the long-term<br />

cosmetic outcome of a boost to the tumor bed following AWBRT<br />

(AWBRT+B). The primary outcome of this study was to evaluate<br />

the cosmetic effect of a boost using a patient reported questionnaire.<br />

The cosmetic subscale in the questionnaire was used to compare the<br />

appearance of the treated versus non treated breast between the boost<br />

and non-boost groups.<br />

MATERIALS AND METHODS<br />

Between 2000 and 2005, 4392 women 75 years and under with<br />

unilateral early-stage breast cancer received AWBRT alone or<br />

AWBRT+B. Random samples of 800 women treated with AWBRT<br />

alone and 800 women treated with AWBRT+B were identified from<br />

the 3960 women still alive at least 5 years after treatment without<br />

contralateral disease. The women were contacted by mail to complete<br />

a questionnaire based on the <strong>Breast</strong> <strong>Cancer</strong> Treatment Outcomes Scale<br />

(22 questions regarding cosmetic, pain and functional outcomes).<br />

Cochrane-Armitage (CA) trend test and Wilcoxon Rank-sum (WR)<br />

were used to compare baseline patient and treatment variables to<br />

long-term cosmetic outcomes between the two treatment groups.<br />

RESULTS<br />

312 women (154 received AWBRT alone and 158 received<br />

AWBRT+B) completed the questionnaire. The median (range) age<br />

of respondents was 57 (40-75) years in the AWBRT alone group and<br />

52 (32-75) years in the AWBRT+B group (p


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 4<br />

CONCLUSION<br />

Similar to conventionally fractionated WBRT, patients who receive<br />

a boost after AWBRT self-report long-term slightly worse cosmetic<br />

and pain outcomes compared AWBRT alone.<br />

P4-16-02<br />

A survival benefit from locoregional radiotherapy for nodepositive<br />

and CMF treated breast cancer is most significant in<br />

Luminal A tumors<br />

Voduc D, Cheang MCU, Tyldesley S, Chia S, Gelmon K, Speers C,<br />

Nielsen TO. BC <strong>Cancer</strong> Agency, Vancouver, BC, Canada; University<br />

of British Columbia, Vancouver, BC, Canada<br />

Background: Between 1978-1986, 318 premenopausal women<br />

treated with mastectomy for lymph node positive breast cancer, were<br />

randomized to CMF chemotherapy alone vs. CMF chemotherapy<br />

and adjuvant radiotherapy (RT) to the chest wall and regional<br />

lymph nodes. After 15 years of follow-up, post-mastectomy RT was<br />

associated with a statistically significant 29% relative risk reduction in<br />

mortality. Recent evidence suggests that Luminal A tumors, identified<br />

using hormone receptors and Ki67, have a particularly favorable<br />

prognosis. We retrospectively identified the Luminal A tumors from<br />

this clinical trial cohort to determine if the response to postmastectomy<br />

RT differed among Luminal A and non-Luminal A tumors.<br />

Methods: 203 archival breast tumor samples from this study were used<br />

to construct a tissue microarray. Luminal A tumors were identified<br />

using an immunopanel consisting of: estrogen receptor, progestorone<br />

receptor, Her2, and Ki67. Luminal A tumors were defined as either<br />

ER or PR positive, Her2 negative, and Ki67 < 14%. Kaplan-Meier<br />

estimates and the log-rank test were used to test the differences in<br />

locoregional relapse free survival (LRFS) and breast cancer specific<br />

survival (BCSS). Interaction between treatment and Luminal A/Nonluminal<br />

A were tested using Cox regression analysis.<br />

Results: The intrinsic subtype was successfully determined in 144<br />

breast tumors, and 49 were classified as Luminal A (34%). Survival<br />

outcomes at 10 years are summarized in Table 1:<br />

Table 1: Survival outcomes at 10 years<br />

<strong>Breast</strong> <strong>Cancer</strong> Specific Survival Luminal A Non-Luminal A<br />

RT 82% 54%<br />

No RT 36% 49%<br />

Log-rank p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P4-16-04<br />

Outcome of stage II/III breast cancer treated with neoadjuvant<br />

versus adjuvant radiotherapy in British Columbia.<br />

Lohmann AE, Voduc D, Speers C, Chia S. British Columbia <strong>Cancer</strong><br />

Agency, Vancouver, BC, Canada<br />

Backgroud: Neoadjuvant radiotherapy (NRT) is generally<br />

recommended after systemic therapy for inoperable stage III breast<br />

cancer. At the BCCA, neoadjuvant radiation is also frequently offered<br />

for patients with operable node positive disease after neoadjuvant<br />

chemotherapy. There is a lack of randomised trial data comparing<br />

outcomes in stage II/III breast cancer when radiation therapy is<br />

delivered neoadjuvantly versus adjuvantly.<br />

Aim: The primary objective of this study is to assess the clinical<br />

outcomes, as measured by relapse-free survival (RFS), overall<br />

survival (OS), and breast cancer-specific survival (BCSS), of women<br />

with stage II/III breast cancer treated with neoadjuvant chemotherapy<br />

and either neoadjuvant or adjuvant radiotherapy.<br />

Methods: Patients were identified by linking the <strong>Breast</strong> <strong>Cancer</strong><br />

Outcomes Unit (BCOU) with the British Columbia <strong>Cancer</strong> Agency<br />

Pharmacy data repository. Inclusion criteria included: Female,<br />

referred to BCCA with newly diagnosed disease, clinical stage II or<br />

III breast cancer, neoadjuvant chemotherapy, breast surgery performed<br />

as part of the initial treatment plan, RT (given adjuvantly or neoadjuvantly<br />

to the breast/chest wall +/- regional nodes). Patients were<br />

excluded if they had a previous or synchronous in situ or invasive<br />

breast cancer. Demographic data, treatment characteristics, ER, PR<br />

and HER-2 status (when available) were extracted. Data was analyzed<br />

using descriptive statistics and Kaplan Méier curves survival analyses<br />

were produced using SPSS, V. 14.<br />

Results: Between Jan 1, 1995 and Dec 31, 2008, 687 patients with<br />

stage II/III disease were identified. 394 patients received neo-adjuvant<br />

and 293 patients received adjuvant radiation. Patients treated with<br />

neoadjuvant vs. adjuvant RT differed in age (median: 51yrs vs.<br />

49yrs, p


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 4<br />

P4-16-06<br />

Radiotherapy to the Primary Tumor Is Associated with Improved<br />

Survival in Stage IV <strong>Breast</strong> <strong>Cancer</strong><br />

Morgan SC, Caudrelier J-M, Clemons MJ. University of Ottawa, ON,<br />

Canada; The Ottawa Hospital <strong>Cancer</strong> Centre, Ottawa, ON, Canada<br />

Background: In patients found to have metastatic disease at the time<br />

of breast cancer diagnosis, the role of local therapy is undefined.<br />

Numerous retrospective analyses have suggested that surgery and/<br />

or external beam radiotherapy (EBRT) directed at the primary tumor<br />

may improve overall survival (OS). All these analyses, however, are<br />

subject to significant selection bias. The current retrospective analysis<br />

of a large registry dataset attempts to limit the effect of this bias.<br />

Methods: The study population consisted of women in the<br />

Surveillance, Epidemiology, and End Results (SEER) program<br />

database diagnosed with stage IV breast cancer between 1988 and<br />

2009. Only those patients for whom surgery to the primary tumor<br />

was recommended but was not undertaken (due to patient refusal<br />

or other uncategorized reasons) were included. In this population<br />

of patients deemed candidates for surgery, the association between<br />

receipt of primary tumor-directed EBRT and overall survival was<br />

studied. Descriptive statistics were used to characterize the study<br />

population. OS was estimated using the Kaplan-Meier (KM) method.<br />

Univariate and multivariate Cox regression were used to identify<br />

factors associated with OS.<br />

Results: A total of 3,529 cases were analyzed. EBRT was received in<br />

768 cases. Median age at diagnosis was 68 years (IQR, 56-79 years).<br />

Median follow-up by reverse KM estimate was 98 months (range,<br />

0-252 months). On univariate analysis, EBRT was associated with<br />

improved OS (hazard ratio 0.80, 95% CI 0.74-0.87, p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

With a mean follow-up of 49 months (range 31-66 months), only one<br />

case of local recurrence has been reported. The observed toxicity was<br />

considered acceptable.<br />

As to cosmetic results, at 6 months after the end of IORT, 71/226<br />

patients (31.4.%) had a score of 2 for symmetry and contour<br />

(asymmetry exhibited by 1/3 or less of volume breast), while 19/226<br />

(8.4%) had a score 3 (asymmetry greater than 1/3 of breast volume).<br />

These findings remained unchanged at the following examinations. No<br />

breast oedema, discoloration at site or scar prominence were observed.<br />

Discussion<br />

IOERT has significant advantages compared to other post-operative<br />

APBI approaches. The surgical re-approximation of the tumor bed,<br />

combined with the high quality of electron beam radiation, generates<br />

substantially more uniform dose distributions.<br />

IOERT offers the advantage of an excellent delineation of the tumor<br />

bed under visual control and high sparing of normal tissue, including<br />

the skin. IOERT delivers a very high biologically dose at the time of<br />

the surgery, when residual tumor cells are more rapidly proliferating.<br />

IOERT is insensitive to chemotherapy sequencing since all of the<br />

radiation is given during the surgery. IOERT offers low-risk women<br />

the possibility of a one-day procedure to treat their cancer and<br />

preserve their breast.<br />

The results of our study are very encouraging compared with other<br />

series with IOERT or APBI. The absolute recurrence rate of 0.4%<br />

and the recurrence rate per year of 0.2% of the present study are<br />

lower compared to the ones from the largest series with IOERT ever<br />

published (3.6% and 1.2%, respectively) (1).<br />

Conclusion<br />

APBI using a single dose of IOERT can be delivered safely in women<br />

with early, low risk breast cancer. A longer follow-up is needed to<br />

ascertain its efficacy compared to that of the current standard treatment<br />

of whole breast irradiation.<br />

References<br />

1. Veronesi U, Orecchia R, Luini A, et al. Intraoperative radiotherapy<br />

during breast conserving surgery: a study on 1,822 cases treated with<br />

electrons. <strong>Breast</strong> <strong>Cancer</strong> Res Treat 2010; 124:141-151.<br />

P4-16-09<br />

Dosimetric feasibility and acute toxicity in a prospective trial of<br />

ultra-short course accelerated partial breast irradiation (APBI)<br />

using a multi-lumen balloon brachytherapy device<br />

Khan AJ, Vicini FA, Brown S, Haffty BG, Kearney T, Dale R, Arthur<br />

DW. <strong>Cancer</strong> Institute of New Jersey, New Brunswick, NJ; Michigan<br />

Healthcare Professionals, Farmington Hills, MI; Wellstar Kennestone<br />

Hospital, Marietta, GA; Imperial College London, United Kingdom;<br />

Virginia Commonwealth University, Massey <strong>Cancer</strong> Center,<br />

Richmond, VA<br />

Background:<br />

Shorter courses of APBI with novel fractionation schedules are being<br />

investigated; a large randomized trial from Europe has recently shown<br />

the safety and efficacy of a single-fraction adjuvant approach (with<br />

limited follow-up). We designed a prospective trial to identify and<br />

address the potential radiobiological and logistical limitations of<br />

single-fraction, intraoperative APBI.<br />

Methods:<br />

We designed a single-arm, multi-institutional, prospective phase<br />

II trial that sequentially treats three cohorts of women (each n=30)<br />

with three progressively hypofractionated schedules. Eligible women<br />

were age ≥ 50 years with unifocal invasive or in situ tumors ≤ 3.0<br />

cm, excised with negative margins, and with negative lymph nodes<br />

and positive hormone-receptors. Using a reference schedule of 60 Gy<br />

delivered in 2 Gy fractions, and assuming tumor parameters: a/b = 4<br />

Gy; a = 0.27 Gy-1, and repopulation parameters of: Teff = 26 days;<br />

delay time = 0 days, the reference tumor BED is ∼ 86 Gy4. We began<br />

with a schedule of 4 fractions of 7 Gy delivered twice-daily using<br />

a Contura MLB multi-lumen device. We defined very conservative<br />

dosimetric criteria for acceptability: maximum skin and rib dose to not<br />

exceed 100% of prescription dose, and V150 and V200 to not exceed<br />

40 cc and 10 cc, respectively. Subsequent schedules are 3 fractions<br />

of 8.25 Gy and 2 fractions of 10.25 Gy, both delivered over 2 days.<br />

The primary endpoint is to exclude a local failure rate exceeding 10%<br />

with the upper limit of a 95% confidence interval.<br />

Results:<br />

A total of 30 patients have been enrolled at the 7 Gy x 4 fractions<br />

dose-level and followed for a minimum of 6 months. The median<br />

skin dose as a percent of prescription dose (PD) was 84% (40-100)<br />

and the median rib dose was 71% (16-119). 96% of the PTV_eval<br />

received a median of 95% of PD (range 85-103). The V150 (range<br />

14-48cc) and V200 (range 0-29cc) criteria were met in all cases. One<br />

breast infection occurred and was treated; 2 cases of symptomatic<br />

fat necrosis and 2 cases of symptomatic seromas occurred. No acute<br />

toxicities greater than CTCAE grade 2 have been observed.<br />

Conclusion:<br />

Short-course APBI is dosimetrically feasible using the Contura MLB<br />

and appears to be tolerable in terms of acute toxicities. Our approach<br />

is based on well-defined radiobiological parameters and allows for<br />

an abbreviated course of treatment that is guided by full pathological<br />

review and the ability to objectively achieve and validate acceptable<br />

dosimetric criteria in each case. We have opened enrollment to the<br />

next schedule of 8.25 Gy for three fractions.<br />

P4-16-10<br />

Positive sentinel lymph nodes (SLNs) without axillary dissection<br />

in breast cancer: the tangent fields of irradiation may not allow<br />

optimal coverage and dose distribution in level I and II of the<br />

axilla<br />

Qiong P, Khodari W, Bigorie V, Bosc R, Dao TH, Totobenazara J-L,<br />

Assaf E, Caillet P, Diana C, Lagrange J-L, Calitchi E, Belkacemi<br />

Y. APHP GH Henri Mondor et Université de Paris-Est, Créteil,<br />

France; APHP GH Henri Mondor, Créteil, France; Association of<br />

Radiotherapy and Oncology of the Mediterranean Area, France;<br />

French <strong>Breast</strong> Intergroup, France<br />

Purpose<br />

The recent Z0011 trial demonstrated equivalent survival in patients<br />

(pts) with breast cancer (BC) and 1 to 2 positive SLNs who were<br />

randomly assigned to SLN biopsy (SLNB) alone or followed by<br />

axillary lymph node dissection (ALND). More importantly, regional<br />

recurrence with SLNB alone was less than 1%, despite the fact that an<br />

estimated 27% of patients had additional metastases in the undissected<br />

axillary nodes. Currently, many teams do not recommend anymore<br />

ALND when the pts receive systemic therapy and whole breast<br />

irradiation (WBI). However, the optimal design of radiation fields for<br />

patients with positive SLNs who do not undergo ALND is uncertain.<br />

This prospective study was designed to define the dose distribution<br />

and coverage of level I and II of the axilla using only tangent fields<br />

after conservative surgery (CS) or total mastectomy (TM).<br />

Methods and Materials<br />

Thirty four pts were included in this analysis. 5 (14.7%) had TM and<br />

29 (85.3%) had CS. Median age was 54 (30-90) years. All pts had<br />

3D conformal RT. The level I/II/III axillary volumes were contoured<br />

using the RTOG contouring atlas. The total dose delivered was 50Gy<br />

in 25 fractions (n=11) or the same dose (D) followed by a boost of<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

414s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 4<br />

10 to 16Gy (n=23). Pts were analyzed according to the total dose<br />

delivered in these 2 groups: group 1 (50Gy; n=11), group 2 (60 to<br />

66Gy; n=23). A subgroup analysis was also performed in the group<br />

2 regarding the initial tumor site and delivered dose: 66Gy to the<br />

upper quadrant (n=14 group 2a) vs the lower quadrant (n=4; group<br />

2b) vs 60Gy to the upper quadrant (n=5, group 2c). All dose-volumehistograms<br />

were analyzed. Mean, median values derived from level I<br />

and level II volumes were compared using t-test. Level of significance<br />

was set at p-value=0.05.<br />

Results<br />

In the whole population, the median D delivered to level I and II<br />

was 16.2 and 1.7 Gy, respectively. The mean D at these levels were<br />

21.2 Gy (rang: 1.0 – 48.1) and 5.0 Gy (0.1 – 40.9), respectively. The<br />

volume of 95% and 50%-isodose coverage were 0 cm 3 and 17.2 cm 3<br />

for level I and 0 cm 3 for level II. The mean values for level I and II<br />

volumes were 1.4 cm 3 and 21.9 cm 3 , respectively.<br />

According to the total D delivered (with or without boost), no<br />

difference was found between group 1 and group 2 in terms mean,<br />

median, and minimal D delivered to axilla levels (p = 0.5 and 0.9).<br />

Conversely, the difference was statistically significant for maximal D<br />

(p = 0.01). Furthermore, there was no difference according to tumor<br />

site and total dose between group 2a and 2b and group 2c (p = 0.07<br />

and 0.7). In addition, no difference was observed between irradiation<br />

of whole breast or chest wall at 50Gy (p = 0.3 and 0.7).<br />

Conclusion<br />

In patients undergoing conservative surgery or post-mastectomy<br />

radiotherapy, tangent fields provide a limited coverage of level I and<br />

II of the axilla as defined by the RTOG contouring atlas. In addition,<br />

these regions were mainly underdosed with au total dose < 30Gy even<br />

for pts who received 66Gy at the upper quadrant. These data should be<br />

considered when only tangent fields are planed to target the axilla in<br />

patients with metastases in SLNB positive without axillary dissection.<br />

P4-16-11<br />

Impact of receptor status on prognosis among breast cancer<br />

patients with brain metastases treated with Cyberknife<br />

radiosurgery<br />

Fasola CE, Gibbs IC, Soltys SG, Horst KC. Stanford <strong>Cancer</strong> Center,<br />

Stanford, CA<br />

Purpose: Stereotactic radiosurgery (SRS) is increasingly used in the<br />

management of brain metastases. Receptor status is an important<br />

prognostic factor among women with breast cancer. The aim of this<br />

study was to evaluate the effect of receptor status on clinical outcomes<br />

in breast cancer patients with brain metastases treated with SRS.<br />

Materials and Methods: We performed a retrospective review of<br />

102 primary breast cancer patients with brain metastases treated<br />

with Cyberknife radiosurgery at Stanford University Medical Center<br />

between 2004 and 2011. The clinical characteristics, pathologic<br />

features, treatment details and clinical outcomes were reviewed.<br />

Median follow-up time was 18 months from the date of treatment<br />

with Cyberknife radiosurgery. All analyses were performed using<br />

SAS software, version 9.3 (SAS Institute, Cary, NC).<br />

Results: Patients with Her-2/neu positive receptor status treated with<br />

trastuzumab had a significantly improved median overall survival after<br />

treatment with stereotactic radiosurgery compared to patients with<br />

Her-2/neu negative receptor status (27 months vs 7 months, p=0.002)<br />

or patients with Her-2/neu positive receptor status who did not<br />

receive trastuzumab (27 months vs 12 months, p=0.04). In contrast,<br />

patients with estrogen receptor, progesterone receptor and Her-2/<br />

neu negative receptor status had significantly worse median overall<br />

survival after treatment with stereotactic radiosurgery compared to<br />

patients with at least one positive receptor (8 months vs 17 months,<br />

p=0.02). On multivariable analysis, Her-2/neu status emerged as the<br />

only significant predictor of survival (p=0.01).<br />

Conclusions: Her-2/neu receptor status was found to be a significant<br />

prognostic factor influencing survival among breast cancer patients<br />

with brain metastases treated with SRS. Importantly, the addition<br />

of treatment with trastuzumab among Her-2 neu positive patients<br />

significantly improved clinical outcomes.<br />

P4-16-12<br />

Outcomes of low- risk Ductal Carcinoma in situ in South East<br />

Asian women treated with breast conservation surgery.<br />

Wong FY, Wang FQ, Chen JJ, Tan CH, Tan PH. National <strong>Cancer</strong><br />

Centre Singapore; Singapore General Hospital, Singapore, Singapore<br />

Background<br />

Singapore is the first South East Asian country to establish a<br />

nationwide breast cancer screening programme in 2002. Since then,<br />

the incidence of breast cancer and pre-invasive disease, in particular,<br />

has increased tremendously. Ductal carcinoma in situ (DCIS)<br />

constitutes over a quarter of all screen-detected cancers.<br />

In Singapore, most patients with DCIS are managed with breast<br />

conserving surgery (BCS). In almost all cases, this is followed by<br />

adjuvant radiotherapy (RT) to the whole breast. However, the optimum<br />

management of low-risk DCIS remains contentious. Non-randomized<br />

data in Caucasian women suggests benefits in the reduction of local<br />

recurrence despite already low recurrence rates. Similar information<br />

in Asian women is lacking. We examined the outcomes of South East<br />

Asian women with low-risk DCIS treated with BCS.<br />

Methods<br />

Retrospective chart reviews of patients treated with BCS for DCIS<br />

from 1995 to 2011 was performed. Patients meeting the pathological<br />

criteria from ECOG 5194 were included: Low-Intermediate grade<br />

(LG) DCIS ≥0.3cm but ≤2.5cm excised with final margins ≥3mm<br />

and High grade (HG) DCIS ≥3mm but ≤1.0cm.<br />

Most patients received adjuvant RT consisting of whole breast RT to<br />

50Gy followed by a 10Gy boost to the tumour bed. Patients deemed<br />

to have especially low recurrence risks did not receive RT and some<br />

patient declined adjuvant RT. Patients who are oestrogen receptor<br />

positive were offered 5 years of tamoxifen.<br />

Results<br />

A total of 744 patients with pathological diagnosis of pure DCIS were<br />

identified, of which 273 meets the selection criteria: LG (N=219);<br />

HG (N=54). Median follow-up for these patients is 60 months.<br />

Median age at diagnosis is 50 years old. About 70% of patients were<br />

screen detected; only 21 of which (7.7%) were diagnosed before the<br />

introduction of breast screening.<br />

There were 8 ipsilateral breast tumour recurrences (IBTR) in total;<br />

7 of which were DCIS. The estimated actuarial IBTR rates at 5- and<br />

10-year for the entire cohort are 1.8% and 4.3% respectively. In<br />

comparison, 10 contralateral new breast primaries were diagnosed in<br />

this cohort, giving 5- and 10-year contralateral relapse rates of 1.9%<br />

and 7.2% respectively.<br />

All patients with LG DCIS received RT except for 9. There were<br />

7 recurrences in this group, two among the 9 patients who did not<br />

receive RT. The estimated IBTR rates at 5- and 10-year are 2.3% and<br />

4.2% respectively. All patients with HG DCIS received RT. There<br />

was only 1 IBTR occurring beyond 5 years giving an estimated IBTR<br />

rate of 4.5% at 10-year. These results compare favourably with the<br />

www.aacrjournals.org 415s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

5-year IBTR rates of 6.1% and 15.3% in the LG and HG patients<br />

respectively in the ECOG 5194 study where patients were managed<br />

with BCS alone.<br />

One patient died of metastatic breast cancer from the contralateral<br />

breast giving a 10-year overall survival of 98.5%.<br />

Conclusions<br />

South East Asian women with screen detected DCIS have exceedingly<br />

low rates of IBTR after breast conservation therapy. The addition<br />

of adjuvant RT approximately halved the IBTR rates in our study<br />

patients. The high proportion of recurrence in the small group of<br />

patients who did not receive RT warns of the potential hazards of<br />

managing even these low-risk patients without radiation.<br />

P4-16-13<br />

Relationship between coverage of axillary lymph nodes, with<br />

tangential breast irradiation, and body mass index<br />

Inokuchi M, Furukawa H, Fujimura T, Ohta T, Ohashi S, Takanaka<br />

T. Kanazawa University Hospital, Kanazawa, Ishikawa, Japan<br />

Background:<br />

The ACOSOG Z0011 trial confirmed axillary clearance is not<br />

necessary in select, clinically lymph node-negative women with<br />

positive sentinel lymph node biopsies (SLNBs) who undergo breastconserving<br />

surgery(BCS) or receive whole-breast radiotherapy(RT)<br />

and systemic therapy. The results suggested that the receipt of adjuvant<br />

breast RT and systemic therapy is adequate for loco-regional control.<br />

But it was unknown whether the low regional recurrence rates in<br />

these patients were caused by the treatment of a portion of the axilla<br />

with the tangent fields. In any case, axillary lymph node dose with<br />

tangential breast irradiation differs for each patient. It could depend<br />

on the proportions or body mass index(BMI) of each patient. If the<br />

difference of BMI impacts axillary coverage in breast RT, it could<br />

also influence loco-regional control. The purpose of this study was<br />

the dosimetric analyses of the axillary coverage with standard breast<br />

tangential irradiation, by BMI categories.<br />

Methods and materials:<br />

Between April 2007 and September 2011, 256 breast cancer patients<br />

were treated with breast tangential irradiation after BCS. They were<br />

classified using BMI categories as follows: underweight (


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 4<br />

were deceased or developed distant metastasis, 5 patients were alive<br />

with no evidence of disease, 3 patients had partial tumour response<br />

with surgery pending.<br />

Conclusions: Patients with locally advanced, triple-negative breast<br />

cancer who progressed during or shortly after NAC had a high clinical<br />

response rate when treated with salvage XRT+ CDDP. In patients<br />

who underwent surgery after salvage treatment, 50% achieved pCR.<br />

Salvage treatments appeared to be relatively well tolerated and safe.<br />

This subset of patients is at very high risk for tumour recurrence.<br />

Prospective, multi-centre studies of concurrent cisplatin and<br />

radiotherapy in the setting of locally advanced, triple negative breast<br />

cancer are needed to further assess efficacy and toxicity.<br />

P4-16-15<br />

Withdrawn by Author<br />

P4-17-01<br />

The effect of body mass index on breast reconstruction outcomes.<br />

Lopez JJ, Laronga C, Doren EL, Sun W, Fulp WJ, Smith PD.<br />

University of South Florida, Tampa, FL; H. Lee Moffitt <strong>Cancer</strong><br />

Center, Tampa, FL<br />

Introduction<br />

With increasing numbers of women choosing mastectomy for breast<br />

cancer treatment, breast reconstruction is consequentially on the rise.<br />

Obesity, a known predictor for wound healing complications, is also<br />

on the rise. Our objective is to review our institutional experience<br />

with the association between Body Mass Index (BMI) and breast<br />

reconstruction complications.<br />

Methods<br />

An IRB approved retrospective review of prospectively gathered<br />

patients having mastectomy with reconstruction was conducted.<br />

Data including patient demographics, stage at diagnosis, adjuvant<br />

treatment, type of mastectomy, type of reconstruction, and<br />

complications were collected. Patients were stratified by BMI into<br />

two categories: Normal weight (BMI 18.5 – 24.9), and overweight/<br />

obese (BMI 25 or greater). The statistical analysis was preformed<br />

using Wilcoxon Rank-Sum Test and Chi Squared Test, both using<br />

exact method with Monte Carlo estimation.<br />

Results<br />

From 06/1996 to 08/2011, 443 patients were identified having<br />

mastectomy and reconstruction. Of these, 218 patients had a normal<br />

weight at the time of mastectomy; 225 patients were overweight/<br />

obese. The overall median age was 49 years (range: 18 - 82). 780<br />

mastectomies with reconstruction (106 unilateral, 337 bilateral) were<br />

performed. The most common reconstruction types included 477<br />

tissue expander with implant reconstructions (62.8% of breasts), 106<br />

latissimus flap with prosthesis (14.0%), and 103 pedicled TRAM flaps<br />

(13.6%). 245 patients (55.3%) experienced at least one complication;<br />

the most common complications were fat necrosis (80 patients, 18.1%<br />

of patients), infection (57, 12.9%), epidermolysis (41, 9.3%), and<br />

skin necrosis (39, 8.8%).<br />

The overweight/obese group had a significantly higher prevalence<br />

of diabetes (6.3% vs. 0.9%, p = 0.0036) and hypertension (26.7%<br />

vs. 11.1%, p < 0.0001) and was more likely to receive neoadjuvant<br />

chemotherapy (16.2% vs. 4.8% p = 0.0005), possibly due to<br />

presentation at a later stage, but no significant differences were<br />

identified. Other comorbid conditions, including smoking, history of<br />

breast cancer, adjuvant chemotherapy, and history of or postsurgical<br />

radiation, were similar between the two groups. One significant<br />

difference was that the overweight/obese group was significantly<br />

older than the normal weight group (p = 0.0005).<br />

There were no significant differences identified in the incidence of any<br />

individual complication between the two BMI groups. Additionally,<br />

the incidence of a patient having any complication was similar<br />

between the two groups (128 overweight/obese patients for 56.9%<br />

and 117 normal weight patients for 53.7%, p = 0.4918). When using<br />

breasts instead of patients as the unit of measure, when an overweight/<br />

obese patient had a complication they were significantly more likely<br />

to require an unanticipated return to the OR (93/160 breasts with a<br />

complication for 58.1% vs. 67/154 for 43.5%, p = 0.0124).<br />

Conclusion<br />

Obesity does not increase the risk of breast reconstruction<br />

complications, but increases the severity of complications if one<br />

should arise. This supports the continued use of breast reconstruction<br />

in patients regardless of their weight, and emphasizes that when<br />

the correct procedure is selected for an overweight/obese patient,<br />

outcomes can be similar to patients that are of a normal weight.<br />

P4-17-02<br />

Radiation therapy and expander-implant breast reconstruction:<br />

an analysis of timing and comparison of complications<br />

Lentz RB, Higgins SA, Matthew MK, Kwei SL. Yale University School<br />

of Medicine, New Haven, CT<br />

Background: The optimal timing of expander/implant exchange in<br />

the setting of post mastectomy radiation (PMRT) remains unclear.<br />

Studies investigating the sequencing of reconstruction and PMRT<br />

are inconclusive, with some reporting exchange prior to PMRT<br />

results in fewer complications, while others report no difference.<br />

When the exchange follows PMRT, most reconstructive surgeons<br />

wait between 3-6 months, however, little is known about the optimal<br />

time for exchange. The primary aim of the study was to characterize<br />

complications associated with sequencing expander/implant breast<br />

reconstruction before or after PMRT. The secondary aim was to<br />

compare the outcomes between early (4<br />

months) expander/implant exchange in the subset of patients who<br />

received PMRT prior to exchange.<br />

Materials and Methods: The medical records of all patients<br />

undergoing PMRT in the setting of expander/implant breast<br />

reconstruction between June 2004-June 2011 at our institution were<br />

reviewed retrospectively. For aim 1, patients were classified as having<br />

undergone exchange prior to the initiation of PMRT (group I) or after<br />

completion of PMRT (group II). For aim 2, patients who underwent<br />

exchange after PMRT were then classified as having undergone<br />

exchange less than 4 months after PMRT (group III) or more than 4<br />

months after PMRT (group IV). Complications requiring additional<br />

surgery or hospitalization were recorded. Statistical analysis was<br />

performed using students t test and Fischer exact test.<br />

Results: Fifty-five eligible patients were identified having undergone<br />

56 two-stage tissue expander/implant breast reconstructions by 6<br />

different plastic surgeons, 22 in group I and 34 in group II. There<br />

was no significant difference in overall complication (54.55% vs<br />

47.06%, p = 0.584) or reconstruction failure rates (13.64% vs 20.59%,<br />

p = 0.724). The most commonly encountered complications were<br />

infection, wound dehiscence and capsular contracture. There was<br />

no significant difference in rate of infection (18.18% vs 23.53%, p =<br />

0.746) or wound dehiscence (4.55% vs 14.71%, p = 0.386). Group<br />

I did experience a higher rate of capsular contracture (40.91% vs<br />

11.76%, p = 0.021). From the 34 breast reconstructions that underwent<br />

expander/implant exchange following PMRT, 20 were in group III<br />

and 14 were in group IV. There was no significant difference in<br />

overall complication (40% vs 57.14%, p = 0.487) or reconstruction<br />

failure rate (25% vs 14.29%, p = 0.672). Trends suggest a higher rate<br />

www.aacrjournals.org 417s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

of infection in group III (30% vs 14.29%, p = 0.422) and a higher<br />

rate of capsular contracture in group IV (5% vs 21.43%, p = 0.283),<br />

however statistical significance was not reached.<br />

Conclusions: Our findings suggest that neither the sequencing nor<br />

timing of expander/implant exchange in the setting of PMRT has<br />

an impact on overall complication or reconstruction failure rate.<br />

Patients who underwent exchange prior to PMRT, thus radiating<br />

their permanent implants, experienced a higher incidence of capsular<br />

contracture. With respect to optimal timing following PMRT, our<br />

results suggest that implant exchange can occur earlier without<br />

resulting in an increase in complications, however there may be an<br />

association between timing and type of complication.<br />

P4-17-03<br />

Towards the standardisation of outcome reporting in<br />

reconstructive breast surgery: Initial results of the BRAVO<br />

(<strong>Breast</strong> Reconstruction and Valid Outcome) Study–A multicentre<br />

consensus process to develop a core outcome set for reconstructive<br />

breast surgery<br />

Potter S, Ward J, Cawthorn S, Holcombe C, Warr R, Wilson S, Tillett<br />

R, Weiler-Mithoff E, Winters Z, Barker J, Oates C, Harcourt D,<br />

Brookes S, Blazeby J. University of Bristol, United Kingdom; North<br />

Bristol NHS Trust, Bristol, United Kingdom; Linda McCartney <strong>Breast</strong><br />

Centre, Royal Liverpool and Broadgreen University Hospitals NHS<br />

Trust, Liverpool, United Kingdom; NHS Greater Glasgow and Clyde,<br />

Glasgow, United Kingdom; University of the West of England, Bristol,<br />

United Kingdom<br />

Introduction: Appropriate outcome selection is essential if research<br />

is to guide decision-making for patients, professionals and policy<br />

makers. Systematic reviews evaluating the clinical, cosmetic and<br />

patient-reported outcomes of breast reconstruction, however, have<br />

demonstrated marked heterogeneity of outcome reporting such that<br />

results from individual studies cannot be compared or combined.<br />

Standardising end-points by developing and using core outcome<br />

sets - an agreed minimum set of outcomes that should be measured<br />

and reported in all research and audit studies – is one way by which<br />

outcome reporting may be improved. We therefore report the initial<br />

results of the BRAVO (<strong>Breast</strong> Reconstruction and Valid Outcomes)<br />

Study which aims to use a scientifically rigorous Delphi consensus<br />

process to develop a core outcome set for reconstructive breast<br />

surgery.<br />

Methods: The Delphi process involves the sequential completion of<br />

questionnaires to allow stakeholder opinions to be synthesised using<br />

item responses to prioritise outcome domains.<br />

The questionnaire was developed from a long list of 148 outcomes<br />

generated from literature reviews and qualitative work with<br />

stakeholders. The outcomes were categorised into 34 domains in six<br />

categories (short-term complications; late complications; symptoms;<br />

psychosocial issues; practical issues and cosmesis) and each domain<br />

operationalised.<br />

Key stakeholders were identified as patients, surgeons, specialist<br />

nurses and psychologists and participants were sampled purposively<br />

to ensure a breadth of perspectives. Each participant was sent a<br />

questionnaire and asked to prioritise the outcomes on a nine-point<br />

likert scale from 1(not important) to 9(extremely important).<br />

The number of respondents in each group rating each outcome as not<br />

important(scores 1-3); equivocal(scores 4-6) or very important(score<br />

7-9) were calculated for each item and compared between groups. The<br />

proportions of respondents rating each item as very important(score<br />

7-9) was used to rank the items.<br />

Results: 213 of the 430 questionnaires were returned(126/274 patients<br />

and 87/156 professionals) giving a response rate of 49.5%.<br />

Patient participants had a median age of 53.4 years(range 34-76)<br />

and had undergone a full range of reconstructive procedures. The<br />

professional group included 39 breast surgeons, 20 plastic surgeons<br />

and 18 clinical nurse specialists.<br />

There was agreement between 7 of the 10 outcomes that each group<br />

rated most highly. Items with consensus included patient-reported<br />

cosmesis,cosmetic satisfaction and early complications. Patients, but<br />

not professionals, considered generic complications such as bleeding<br />

to be important while professionals valued psychosocial issues such<br />

as self-esteem more highly than patients.<br />

Conclusions: Patients and professionals prioritise similar<br />

outcomes, but areas of discrepancy with regard to complications<br />

and psychosocial outcomes remain. A further Delphi round asking<br />

participants to re-prioritise outcomes and a consensus meeting to<br />

ratify the final decisions will be necessary to determine a final core<br />

outcome set for reconstructive breast surgery.<br />

P4-17-04<br />

BRECONDA: Development and acceptability of an interactive<br />

decisional support tool for women considering breast<br />

reconstruction<br />

Sherman KA, Harcourt D, Lam T, Boyages J. Macquarie University,<br />

Sydney, NSW, Australia; Westmead Hospital, University of Sydney,<br />

Westmead, NSW, Australia; University of the West of England, Bristol,<br />

United Kingdom<br />

Introduction<br />

Women needing a mastectomy for breast cancer, or cancer<br />

prophylaxis, are faced with the difficult decision regarding whether,<br />

and how, to restore breast shape after surgery. To a large extent<br />

this decision is based on personal preferences and values. In view<br />

of limited support resources available in this context, we have<br />

developed an online interactive decision aid, BRECONDA, to assist<br />

with decision-making. BRECONDA uses a multi-media platform to<br />

provide up-to-date information about surgical choices, interactive<br />

decision sheets encouraging women to weigh-up perceived benefits<br />

and risks and identify personal values and preferences, and video<br />

recorded patient stories. Since psychological stress can hamper<br />

decision-making, BRECONDA also demonstrates videoed stress<br />

management relaxation techniques. The aim of this study was to<br />

assess the user acceptability of this intervention.<br />

Methods<br />

Following diagnosis, and prior to surgery, 54 women with breast<br />

cancer who were eligible for breast reconstruction following<br />

mastectomy were randomly assigned into one of two conditions: 1)<br />

Intervention group which received access to the BRECONDA program<br />

as well as a standard information booklet about breast surgery given<br />

to all such patients; and, 2) Control/Usual care group which received<br />

the standard information booklet alone. User ratings of satisfaction<br />

and reactions to BRECONDA were documented at 6-week followup<br />

for the Intervention group through quantitative measures and<br />

telephone interviews. Additionally, perceived decisional conflict,<br />

distress (intrusive and avoidant thoughts), knowledge and satisfaction<br />

with information at the 6-week assessment were documented for<br />

all participants. ANCOVAs were used to identify between group<br />

differences on these key variables at follow-up.<br />

Results<br />

Intervention participants’ ratings of BRECONDA demonstrated<br />

high user acceptability, with high scores on perceived usefulness,<br />

ease of use and provision of sufficient information. Interview data<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

418s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 4<br />

indicated that Intervention participants perceived BRECONDA to be<br />

well-balanced, informative, and beneficial to the decision making<br />

process and that it helped them feel more secure in their decision<br />

and to prepare questions for their surgeon. Interactive decision<br />

sheets, patient testimonials and photo galleries were highly valued<br />

by all interviewees. At follow-up, 40% of participants had undergone<br />

immediate reconstruction, with fewer Intervention participants<br />

electing this surgery. Furthermore, Intervention participants reported<br />

lower decisional conflict compared with Usual Care participants at<br />

follow-up (p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Results: 237 patients underwent implant/expander reconstruction.<br />

Median time from implant placement until last follow-up was 173<br />

days. 21.5% developed major complications (needing operative time<br />

or infection needing intravenous treatment). Diabetes was the most<br />

consistent factor associated with major complications (46.7 vs 20.1%,<br />

p = 0.02 and p = 0.009 in multivariable analysis). Radiation was linked<br />

to capsular contractures (18.6 vs 10.1%, p = 0.02). Chemotherapy<br />

(25.0 vs 19.0%, p = 0.26) or radiation (26.3 vs 19.1%, p = 0.21) did<br />

not predict major complications. Among patients receiving PMRT<br />

(80 patients), 26.2% had major complications, 34 had immediate<br />

PMRT on the expander and 44 had PMRT to the chest wall, followed<br />

by delayed reconstruction with expander/implant placement. In<br />

these, delayed reconstruction increased dehiscence (0 vs 18.2%, p =<br />

0.009) compared to immediate reconstruction, with a trend for higher<br />

incidence of major complications in the delayed reconstruction group<br />

(14.7 vs 34.1%, p = 0.05). 40 Gy/16 versus 50-50.4 Gy/25-28 (28 vs<br />

24%, p = 0.78) was not associated with major complications. Diabetes<br />

and smoking were associated with several complications.<br />

Conclusions: Diabetes is associated with a higher rate of major<br />

complications after expander/implant reconstruction while radiation<br />

increases capsular contractures. If PMRT is indicated, putting in<br />

an expander before radiation results in less morbidity than delayed<br />

reconstruction. Also, 40 Gy/16 versus 50-50.4 Gy/25-28 do not differ<br />

much in terms of complication rates.<br />

P4-17-09<br />

Efficacy and safety of lipomodelling and adipose tissue derived<br />

degenerative stem cells(ADRC) for breast reconstruction –<br />

Medium term follow up<br />

Noor L, Bhaskar P, Hennessy C. University Hospital of North Tees,<br />

Cleveland, United Kingdom<br />

Background: Past and current literature support Lipomodelling as<br />

a tool for breast reconstruction. Lipomodelling with Adipose tissue<br />

Derived Regenerative Stem Cells dramatically improves the graft<br />

survival through both adipogenesis and angiogenesis. Literature<br />

regarding safety of this procedure for breast reconstruction is limited.<br />

Our aim was to assess the safety and efficacy of ADRC-enriched fat<br />

grafts for breast reconstruction.<br />

Material & Methods: A prospective study from September 2008<br />

– April 2012 at a District General Hospital UK including patients<br />

operated by two breast surgeons. The indications and the fat grafting<br />

procedures used are analyzed (Cytori Celution 800/CRS or Cytori<br />

pure graft system). Patients with benign indications were followed<br />

for 1 year and those with cancer were followed for 5 years according<br />

to regional guide lines with breast imaging.<br />

Results: Out of total 50 patients, 10 had benign diagnosis while 40<br />

had breast cancer treatment. 30 patients had correction after breast<br />

conservation, 17 had revision of reconstruction (8 TRAM, 4 LD<br />

and 5 implant based reconstruction) while 3 had other procedures.<br />

19/40 had history of breast irradiation as adjuvant treatment after<br />

surgery for breast cancer. Median age was 52 years with range 18-<br />

72 years. ADRC enriched fat grafting was done in 40 cases and pure<br />

graft in10 patients. Graft volume injected was 80-420 mls (average<br />

247 mls). 90% patients are satisfied with outcome of surgery with<br />

good to excellent cosmetic outcome. Only 4 patients had noticeable<br />

fat resorption, 3 underwent redo grafting. At average follow up of<br />

21 months (1-48 months) no significant complications were seen,<br />

imaging showed fat necrosis in 3 and oil cyst in 1 patient though no<br />

one required biopsy. No patient developed local recurrence, although<br />

4 subsequently found to have metastatic disease.<br />

Conclusion: We found it a useful, safe and effective technique for<br />

breast reconstruction with high patient satisfaction. We recommend<br />

a central database of all patients with this procedure to substantiate<br />

findings of this study and as recommended by Association of <strong>Breast</strong><br />

Surgery (ABS) & British Association of Plastic Reconstructive &<br />

Aesthetic Surgeons (BAPRAS).<br />

P5-01-01<br />

Predicting OncoDX Recurrence Scores with Immunohistochemical<br />

Markers<br />

Bradshaw SH, Gravel DH, Song X, Marginean EC, Robertson SJ.<br />

The Ottawa Hospital, Ottawa, ON, Canada<br />

Background: Standard immunohistochemistry (IHC) performed for<br />

invasive breast carcinoma includes ER, PR and Her2/Neu status, and<br />

these markers are used in conjunction with other patient and tumour<br />

factors to determine prognosis and guide treatment. Many, but not<br />

all, low stage, lymph node (LN) negative, ER positive patients have<br />

a good prognosis without chemotherapy. Thus a demand exists for<br />

predictive tools to stratify patient risk within this subgroup. It has<br />

been reported that, for a subset of ER positive Her2/Neu negative<br />

patients, the 21 gene OncotypeDX recurrence score (Onco-RS) adds<br />

independent prognostic information to that obtained from these<br />

standard IHC markers (1). As several genes analyzed for the Onco-RS<br />

relate to ER, PR, HER2/neu and proliferative status, it is reasonable<br />

to try to incorporate clinical-pathological variables and these IHC<br />

scores into a predictive model. Indeed, recent studies suggest that most<br />

of the additional information provided by the OncoDX-RS may be<br />

obtained more cost effectively using the Ki-67 IHC based proliferation<br />

percentage combined with a semi-quantitative assessment of standard<br />

IHC markers including ER and PR and Her2/neu (2). The aim of this<br />

study is to assess the ability of a simple combined IHC recurrence<br />

score (IHC-RS) to predict Onco-RS. The IHC-RS was derived from<br />

a simple semi-quantitative assessment of ER and PR combined with<br />

Ki-67 proliferation percentage.<br />

Design: A cohort of 159 women aged 27-78 with ER positive, HER2/<br />

neu negative breast cancer completed Oncodx testing between March<br />

2010 and May 2012. This sample reflects the population selected at<br />

our institution for Oncotype testing. The variables investigated for<br />

inclusion in a model to predict RS score included tumor grade, stage,<br />

patient age, Allred ER & PR and Ki-67 percentage.<br />

Results and Discussion: A predictive model was developed to<br />

generate a recurrence score (IHC-RS) using stepwise multiple<br />

regression incorporating Allred ER score, Allred PR score and Ki-<br />

67 percentage. The best subset model (Schwartz BIC) accounted<br />

for 60.7% of the Onco-RS variability (adjusted R 2 =60.7, p = 0.05).<br />

In addition, analysis of individual cases where the IHC-RS was not<br />

in agreement with the Onco-RS reveals that the Onco-RS, although<br />

technically highly reproducible, may suffer from sampling error.<br />

The IHC-RS is more robust with respect to sampling error, owing to<br />

the retention of tumor architecture inherent in IHC. IHC, however,<br />

can lack the technical reproducibility and transportability inherent<br />

in the Onco-RS methodology. There are clearly advantages to an<br />

ICH derived multi-score such as IHC-4 (3) or the simpler IHC-RS<br />

proposed in this study. Full utility of any IHC-based recurrence score<br />

will require standardization of testing and scoring both within and<br />

across different testing laboratories. In addition, full utility of any IHC<br />

based model will require direct correlation to patient outcome, rather<br />

than to a surrogate marker such as the Onco-RS used in this study.<br />

1 - M Dowsett et al. J Clin Oncol. 2010 Apr 10; 1829-1834<br />

2 – Cuzick J et al. J Clin Oncol. 2011 Nov 10;4273-8.<br />

3 –S Barton et al. British Journal of <strong>Cancer</strong> (2012) 106, 1760–1765.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

420s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 5<br />

P5-01-02<br />

Inter-observer concordance of Ki-67 labeling index in breast<br />

cancer: Japan <strong>Breast</strong> <strong>Cancer</strong> Research Group (JBCRG) Ki-67<br />

Ring Study<br />

Ueno T, Mikami Y, Yoshimura K, Tsuda H, Kurosumi M, Masuda S,<br />

Horii R, Toi M, Sasano H. Kyoto University Hospital, Kyoto, Japan;<br />

National <strong>Cancer</strong> Center Hospital, Tokyo, Japan; Saitama <strong>Cancer</strong><br />

Center, Saitama, Japan; Nihon University School of Medicine, Tokyo,<br />

Japan; The <strong>Cancer</strong> Institute Hospital of the Japanese Foundation<br />

for <strong>Cancer</strong> Research, Tokyo, Japan; Tohoku University School of<br />

Medicine, Sendai, Japan<br />

Background: The standardized assessment of Ki-67 labeling index<br />

(LI) plays pivotal roles in identifying the patients (pts) with primary<br />

breast cancer who could benefit from systemic chemotherapy, in<br />

particular among pts with estrogen receptor(ER)-positive cancers.<br />

Therefore, in this study, we evaluated the inter-observer concordance<br />

of the assessment of Ki-67 LI in archival materials.<br />

Methods: Six surgical pathologists specializing in breast pathology<br />

from different Japanese institutions participated in this study. All<br />

slides were prepared from archival tissues of breast cancer fixed<br />

in 10% buffered formalin for 24 hours in a single institution (KU).<br />

Three independent studies were conducted. Study 1) Six consecutive<br />

slides were prepared from 5 cases. A slide from each case was stained<br />

with MIB-1 (DAKO, Denmark) in each institution according to their<br />

routine methods. Total of 30 stained slides were assessed for Ki-67<br />

LI by each pathologist using two different modes of assessment. One<br />

is the scoring system in which the rate of positive cells was scored<br />

from 1 (0 – 9 %) to 10 (90 – 100%) without counting the cell number.<br />

The second one is the counting system in which approximately 1000<br />

cells in total were counted in the hot spots and the positive rate was<br />

calculated. Study 2) Twenty tumors with Ki-67 LI ranging from 5 to<br />

25 (15 ± 10) %, stained in a single institution (KU) were assessed by<br />

each pathologist by the counting system. Study 3) In order to avoid<br />

variations by assessment in varied microscopic fields and to further<br />

evaluate the variation of threshold of immunointensity interpreted<br />

as positive by different pathologists, fifteen printed photographs of<br />

Ki-67-stained slides were sent and assessed for Ki-67 LI by each<br />

participating breast pathologist.<br />

Results:<br />

Study 1) The counting system demonstrated a better correlation<br />

of Ki-67 LI among six pathologists than the scoring system {the<br />

intraclass correlation coefficient (ICC), 0.66 (95% confidence interval<br />

0.52-0.78) for the counting system, 0.57 (0.42-0.72) for the scoring<br />

system}. The two assessment systems showed a moderate correlation<br />

{ICC, 0.68 (0.60-0.75)}. Study 2) The assessment of Ki-67 LI in 20<br />

slides with Ki-67 LI of 5 to 25 % demonstrated a correlation similar<br />

to that in the specimens with an unrestricted range of Ki-67 LI in<br />

the study 1 {ICC, 0.68 (0.50-0.81) for the study 2, 0.66 (0.52-0.78)<br />

for the study 1}. Study 3) The assessment of Ki-67 LI in the same<br />

photographs yielded a considerably significant concordance among<br />

six pathologists {ICC, 0.94 (0.88-0.97)}.<br />

Conclusion:<br />

The counting system turned out better than the scoring system in<br />

terms of the inter-observer agreement of the Ki-67 LI assessment.<br />

The degree of concordance was by no means influenced by the range<br />

of Ki-67 LI. The concordance of the Ki-67 LI assessment among six<br />

participating pathologists was significantly high when the assessed<br />

field was fixed using the same photographs for evaluation, suggesting<br />

that the selection of the fields for evaluation is critical. These results<br />

suggest that identification of hot spots for evaluation is pivotal for<br />

obtaining the accurate Ki-67 LI of breast cancer and still images of<br />

these hot spots could provide reproducible results.<br />

P5-01-03<br />

Discordant Estrogen and Progesterone Receptor Status in <strong>Breast</strong><br />

<strong>Cancer</strong><br />

Hefti MM, Hu R, Knoblauch N, Collins L, Tamimi RM, Beck AH.<br />

Beth Israel Deaconess Medical Center, Boston, MA; Brigham and<br />

Women’s Hospital, Boston, MA<br />

Introduction: Hormone receptor status is used to guide clinical<br />

therapy in breast cancer. Estrogen receptor (ER) assays from<br />

pathology reports have been shown to be in agreement with ER<br />

measured by a centralized laboratory on tissue microarray 87% of<br />

the time (Kappa = 0.64); however, little is known of reproducibility<br />

within subsets of patients defined by ER/PR status (1).<br />

Methods: We computed percent agreement and Kappa statistics for<br />

ER/PR status as reported in pathology reports from Nurse’s Health<br />

Study (NHS) participants with breast cancer from 1976 to 2000, and<br />

as scored by immunohistochemistry (IHC) on tissue microarrays at a<br />

central laboratory (n=2011). Statistics were computed separately for 4<br />

subsets of patients stratified by ER/PR status as defined by pathology<br />

reports (ER+/PR+, ER+/PR-, ER-/PR+, ER-/PR-). We also used a<br />

curated compilation of publicly available gene expression data sets<br />

providing both IHC and microarray data on ER and PR expression<br />

(n=1236).<br />

Results: We observed significant heterogeneity in Kappa values<br />

across the four groups of patients, with ER+/PR+ and ER-/PRshowing<br />

the strongest agreement, ER+/PR- showing fair agreement,<br />

and ER-/PR+ showing no significant agreement: 89.1% of ER+/PR+<br />

cases from medical record were ER+/PR+ by TMA (κ=.60), 69.4% of<br />

ER-/PR- cases from medical record were ER-/PR- by TMA (κ=.63),<br />

42% of ER+/PR- cases from medical record were ER+/PR- by TMA<br />

(κ=.37), and only 6% of cases of ER-/PR+ cases from medical record<br />

were ER-/PR+ by TMA (κ=.06). Using publicly available microarray<br />

data, we observed similar results comparing IHC to gene expression<br />

data in the same samples. Expression values were scaled across<br />

patients, and a patient was classified as positive for the marker by<br />

microarray if the scaled expression value was greater than zero. The<br />

agreement between IHC data and gene expression-based classification<br />

was 50.0% (k=.50), 80.8% (κ=.54), 42.0% (κ=.15), and 7.2% (κ=-<br />

.006) for ER+/PR+, ER-/PR-, ER+/PR-, and ER-/PR+, respectively.<br />

Conclusion: ER-/PR+ is by far the most rare and least reproducible<br />

subset of breast cancer cases. Given the lack of reproducibility of<br />

the ER-/PR+ group and the minimal reported benefit from hormonal<br />

therapy in these patients (2), these data question the clinical utility<br />

of assessment of PR, particularly in ER- breast cancer.<br />

1. Collins LC, Marotti JD, Baer HJ, Tamimi RM. Comparison of<br />

estrogen receptor results from pathology reports with results from<br />

central laboratory testing. Journal of the National <strong>Cancer</strong> Institute.<br />

2008;100(3):218-21.<br />

2. Anderson H, Hills M, Zabaglo L, A’Hern R, Leary AF, Haynes BP,<br />

et al. Relationship between estrogen receptor, progesterone receptor,<br />

HER-2 and Ki67 expression and efficacy of aromatase inhibitors in<br />

advanced breast cancer. Annals of oncology: official journal of the<br />

European Society for Medical Oncology/ESMO. 2011;22(8):1770-6.<br />

P5-01-04<br />

Clinico-pathological features of low-grade triple negative early<br />

breast cancers.<br />

Nourieh M, Furhmann L, Feron J-G, Caly M, Diéras V, Sastre-Garau<br />

X, Chavrier P, Vincent-Salomon A. Institut Curie, Paris, France<br />

Aims: To characterize clinico-pathological features and clinical<br />

outcome of low grade triple negative early breast carcinomas.<br />

www.aacrjournals.org 421s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Material and methods: Between January 2005 and December 2006,<br />

300 tumours were classified as triple negative (ER-ve, PR-ve, HER2<br />

0/1+) out of 3000 patients treated for a breast cancer at the Institut<br />

curie. Patients with a low-grade (grade 1 and 2 according to Ellis<br />

and Elston) T1 and small T2 (< 3cm) breast carcinoma treated with<br />

surgery first were considered in our study. Immunohistochemistry<br />

was performed with ER, PR, Androgen Receptor (AR), HER2,<br />

Ki67, EGFR, CK5/6, CK14 and CK8/18 antibodies for all tumours;<br />

GCDFP15 for invasive lobular carcinomas (ILC) and invasive ductal<br />

carcinoma (IDC) with apocrine features, and CA15-3 to assess the<br />

inverted polarity if needed.<br />

Results: We identified 36 low-grade carcinomas out of 186 triple<br />

negative breast carcinomas (19%). Thirty-two tumours were grade<br />

2 and four tumours grade 1 and associated with a low or a moderate<br />

mitotic score. Ki67 proliferation index was high (≥20%) in 22 cases<br />

(61%) (median: 22%-mean: 29%). Thirty-one (86%) tumours showed<br />

a basal- like phenotype (EGFR+ or CK5/6+ or CK14 +). Interestingly,<br />

the five “nul” (non basal-like) triple negative tumours demonstrated<br />

a low proliferation index (Ki67 ≤20%). All but one tumors expressed<br />

CK8/18. The tumour’s size varied between 3 and 37 mm (mean =<br />

17.2). Lympho- vascular invasion was present in 10 cases (27%).<br />

High and intermediate grade ductal carcinoma in situ component was<br />

associated in 28 cases (77%). Stroma was abundant and associated<br />

with a lymphocytic infiltrate in all cases. Various histological types<br />

were observed: 16 IDC (44%), 7 ILC (19%), 5 IDC with apocrine<br />

differentiation, 2 micropapillary, 1 papillary, 1 mucinous, 3 adenoid<br />

cystic and one low grade adeno-squamous carcinomas. Three out of<br />

16 IDC showed a week nuclear positivity with AR. Five and six out<br />

of the seven ILC were positive for AR and GCDFP15 respectively.<br />

All carcinomas with apocrine differentiation were AR and CGDFP15<br />

positive. Conversely, all micropapillary, papillary and mucinous<br />

carcinomas were AR negative. The mean age of patients was 61.6<br />

years. Distant metastases occurred in five patients. Thirty-three (91%)<br />

women were alive and 29 (80%) of them without evidence of cancer<br />

at 6 years of follow-up.<br />

Conclusion: Low grade invasive carcinomas represent 19% of the<br />

cases in our series of 186 triple negative early breast carcinomas and<br />

were diagnosed in patients older than 60 years. These cases were<br />

of various histological types, were all associated with a marked<br />

lymphocytic infiltrate and in the majority of the cases with high grade<br />

ductal carcinomas in situ. Despite a low mitotic activity, proliferation<br />

index was generally higher than 20% except for “nul” non-basal<br />

cases. However, patient’s overall survival seemed to be better than<br />

that reported for high grade triple negative breast cancer. Histological<br />

types, proliferation and age should be taken into account for treatment<br />

decision in triple negative early breast carcinoma patients.<br />

P5-01-05<br />

Could c-myc amplification replace Cahan’s criterias to<br />

discriminate secondary from primary angiosarcoma of the<br />

breast?<br />

Laé M, Hamel F, Hadj-Hamou S, Malfoy B, Kirova YM. Institut<br />

Curie, Paris, France<br />

Background: Angiosarcomas (AS) are a relatively rare histological<br />

subtype of sarcomas. Despite their rarity, AS display remarkable<br />

clinical heterogeneity. These tumors can occur in any location in the<br />

body, but the breast AS are important part of these tumours. Cahan et<br />

al. defined the criteria for the diagnosis of radiation induced sarcomas<br />

(RIS), used since 1948: i) prior history of RT; ii) asymptomatic<br />

latent period of several years; iii) the occurrence of a sarcoma<br />

within a previously irradiated field; iiii) histological confirmation of<br />

sarcomatous nature of the post-irradiation lesion.<br />

Purpose: The aim of this study is to show a new biological data which<br />

can help us to recognize these rare tumors.<br />

Material and Methods: Forty-one breast AS were studied<br />

(pathological review, clinical and follow-up information obtained),<br />

as well as the transcriptome signatures of the radiation tumorigenesis.<br />

For all RIS, the patients’ radiotherapy plans and hospital records were<br />

reviewed and their radiation related nature was confirmed if they<br />

respected the classic Cahan’s criteria. Interphase FISH was performed<br />

by hybridization of probes covering C-MYC (chromosome 8q24.21)<br />

and CEP8 on tissue sections from each of the forty-one tumors.<br />

Results: Among forty-one breast AS, thirty one were secondary<br />

tumors and ten primary tumors. Amplification (5 to 20 fold) of the<br />

MYC oncogene was found in all breast secondary AS (31 cases) but<br />

in none of the 10 primary AS except one. The relationships between<br />

clinical, pathological and biological features are studied. These data<br />

point the pathways preferentially involved in the pathogenesis of<br />

secondary AS of the breast and may provide the basis for an additional<br />

targeted therapy.<br />

Conclusion: The presence of c-myc amplification on biopsy<br />

or surgical specimen represents an important additional tool to<br />

discriminate secondary from primary angiosarcoma of the breast<br />

when radiotherapy data are not available.<br />

P5-01-06<br />

Differences in FGF1 and FGFR2 expression in BRCA1-associated,<br />

BRCA2-associated, and sporadic breast carcinomas<br />

Vos S, van der Wall E, van Diest PJ, van der Groep P. University<br />

Medical Center Utrecht, Netherlands<br />

Introduction. In contrast to BRCA1-associated breast cancer, a<br />

distinctive phenotype for BRCA2-associated carcinomas has not<br />

been identified yet as there is no clear distinction between BRCA2-<br />

and non-BRCA mutation related or sporadic carcinomas. Recently,<br />

studies suggest overexpression of fibroblast growth factor 1 (FGF1)<br />

and fibroblast growth factor receptor 2 (FGFR2) in BRCA2 related<br />

cancers. The aim of the study was therefore to investigate whether<br />

there is differential expression of FGF1 and/or FGFR2 between<br />

BRCA2 related and sporadic and BRCA1 related cancers. This would<br />

reveal the usefulness of FGF1 and FGFR2 immunohistochemistry in<br />

daily pathology practise.<br />

Method. Invasive breast carcinomas of 33 BRCA1 and 22 BRCA2<br />

germline mutation carriers and a tissue microarray containing 104<br />

sporadic invasive breast carcinomas were immunohistochemically<br />

stained for FGF1, FGFR2, estrogen receptor (ER), progesterone<br />

receptor (PR), HER2, epidermal growth factor receptor 1 (EGFR),<br />

cytokeratin (CK) 5/6 and CK14.<br />

Results. FGFR2 expression was seen in 68.2% and 79.0% of BRCA2-<br />

associated and sporadic carcinomas respectively, in contrast to 22.6%<br />

of BRCA1-associated tumors (p = 0.000). FGF1 expression was seen<br />

in 72.7% of BRCA2-associated carcinomas and in 45.2% and 41.8% in<br />

BRCA1-associated and sporadic carcinomas, respectively (p = 0.032).<br />

Conclusion. FGFR2 expression differs significantly between BRCA1-<br />

and BRCA2 associated breast carcinomas but not between BRCA2<br />

and sporadic cancers. FGF1 expression differs significantly between<br />

BRCA2-associated and sporadic carcinomas and could be used as a<br />

BRCA2-specific biomarker.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

422s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 5<br />

P5-01-07<br />

Fibroadenomatoid changes are more prevalent in middle-aged<br />

women and have a positive association with invasive breast cancer<br />

Chen Y, Bekhash A, Kovatich AJ, Hooke JA, Kvecher L, Mitchell EP,<br />

Rui H, Mural RJ, Shriver CD, Hu H. Windber Research Institute,<br />

Windber, PA; Walter Reed National Military Medical Center,<br />

Bethesda, MD; MDR, Global Systems LLC, Windber, PA; Thomas<br />

Jefferson University, Philadelphia, PA<br />

Background: The role of benign breast diseases (BBDs) in the<br />

development of invasive breast cancers (IBCs) has been studied for<br />

many years. Some BBDs have been studied comprehensively (e.g.,<br />

fibrocystic changes (FCC)) while less is known about other BBDs<br />

(e.g., fiboadenomatoid changes (FAC)). FAC has been considered by<br />

some researchers as a precursor of fibroadenoma (FA). Conclusions<br />

from different studies vary, partially due to different interpretation<br />

methods and diagnostic criteria when multiple hospitals and<br />

pathologists were involved. In this study, we used subjects in the<br />

Clinical <strong>Breast</strong> Care Project (CBCP) from a military medical center<br />

where pathology slides were reviewed by a single breast pathologist<br />

to study FAC, FA, and FCC in comparison to the published literature.<br />

Methods: Subjects were enrolled in the study following IRBapproved,<br />

HIPAA-compliant protocols. All the clinicopathologic<br />

data are available from the CBCP data warehouse (DW4TR). In the<br />

CBCP, FCC is composed of 4 components: stromal fibrosis, cysts,<br />

apocrine metaplasia, and sclerosing adenosis. Two modeling studies<br />

were performed. i) For the BBDs and IBC association study, two<br />

groups of subjects were identified: 1136 subjects diagnosed with<br />

“Benign” or “Atypical” diseases, and 619 cases diagnosed with<br />

IBCs. A logistic regression model was developed for the prediction<br />

of IBCs by the 3 BBDs and 2 well-established risk factors (RF): age<br />

(younger, 60) and race (Caucasian,<br />

African American, Asian, and other). ii) For the RF association study<br />

with the BBDs, 6 additional RFs reported to be associated with<br />

these BBDs were identified from the literature: current use of oral<br />

contraceptives, number of live births, education, body mass index,<br />

hormonal replacement therapy, and IBC family history. These 8 RFs<br />

were used to develop a logistic regression model for each of the BBDs.<br />

The analyses were performed in SAS.<br />

Results: In the first study, age and race were confirmed as RFs<br />

for IBCs. FAC was positively associated with IBC (OR=3.04,<br />

95%CI=2.06 to 4.50). FA was negatively associated with IBC,<br />

and the level of the association was stronger in women without<br />

FCC (OR=0.15, 95%CI=0.08 to 0.28), compared to women with<br />

FCC (OR=0.40, 95%CI=0.24 to 0.65). FCC was not significantly<br />

associated with IBC. Results from the second study indicated that,<br />

age was significantly associated with FAC (p=0.015), specifically<br />

the middle-aged women were more likely to have FAC compared<br />

to younger women (OR=2.03, 95%CI=1.23 to 3.34), while the<br />

older women were at a non-significantly increased risk. Trends of<br />

association with FAC were also noted for the number of live birth<br />

(p=0.095), ethnicity (p=0.096), and current oral contraceptive pill use<br />

(p=0.077). The FCC model results were in general consistent with the<br />

literature, and we also confirmed that age was negatively associated<br />

with the diagnosis of FA.<br />

Discussion: Our study was consistent with FCC findings in the<br />

literature. We observed that FAC was positively associated with IBC,<br />

whereas FA was negatively associated. Also, FAC occurred more<br />

often in middle-aged women while FAs occurrence was higher in<br />

younger women. Our results suggest that FAC and FA may be two<br />

different diseases.<br />

P5-01-08<br />

Complex fibroadenoma is not an independent risk marker for<br />

breast cancer<br />

Nassar A, Visscher DW, Degnim AC, Frank RD, Vierkant RA,<br />

Hartmann LC, Frost MH, Ghosh K. Mayo Clinic, Rochester, MN<br />

Background: Fibroadenoma (FA) is a relatively common benign<br />

breast tumor that can occur in women of any age, with a peak<br />

incidence during the second and third decades. The only previous<br />

study of this lesion reported that FAs are associated with a 2.2 times<br />

increased risk of developing invasive breast cancer (BC) compared<br />

to matched controls [1]. This relative risk may increase to 3.1 among<br />

patients with complex FA, and remains elevated for decades after<br />

diagnosis. However, this study did not thoroughly account for other<br />

forms of concomitant risk factors. Our investigative team sought to<br />

examine breast cancer risk among women with non-complex and<br />

complex FA, overall and stratified by other BC risk factors.<br />

Materials and Methods: The study cohort included women between<br />

ages 18 to 85 in the Mayo Benign <strong>Breast</strong> Disease (BBD) Cohort who<br />

underwent excisional breast biopsy between 1967and1991 and were<br />

found to have a FA. FA was defined histologically as a combination<br />

of epithelial and stromal proliferation. Complex FA was defined as<br />

FA associated with any of the following features: sclerosing adenosis,<br />

epithelial calcifications, papillary apocrine change, and microcysts<br />

greater than 3.0 mm. The primary endpoint was a diagnosis of<br />

BC, determined using the Mayo medical record and questionnaire<br />

information from study participants. A single breast pathologist,<br />

blinded to the initial diagnosis and clinical outcome, performed<br />

pathology review. Observed vs. expected BC risk across levels of<br />

FA was assessed via standardized incidence ratios (SIRs), using agestratified<br />

incidence rates from the Iowa Surveillance, Epidemiology,<br />

and End Results (SEER) registry. Analyses were carried out overall<br />

and within subgroups of involution status (none, partial, complete)<br />

and overall histology (non-proliferative disease [NP], proliferative<br />

disease without atypia [PDWA] or atypical hyperplasia [AH]).<br />

Results: Of 9097 women in the Mayo BBD Cohort, FA were identified<br />

in 2139- non-complex in 1903 (20.9%) and complex in 236 (2.6%).<br />

The greatest proportion of FA occurred in the 40-69 age range. The<br />

mean ages for women with non-complex FA and complex FA were<br />

45.7 and 50.2 years respectively. The SIR of breast cancer in the<br />

overall BBD cohort was 1.5 (95% CI [1.4-1.6]). The SIR among<br />

women with non-complex FA was 1.5 (95% CI [1.3-1.8]), and for<br />

complex FA increased to 2.41 (95% CI [1.7-3.4]). However, women<br />

with complex FA were more likely to have other concomitant highrisk<br />

histologic features such as PDWA and incomplete involution.<br />

In stratified analyses accounting for involution status and PDWA,<br />

complex FA did not demonstrate an independent increase in BC risk.<br />

Conclusion: Complex FA does not confer an increased risk for BC<br />

beyond other established histologic features. Therefore, women with<br />

complex FA should be managed based upon a risk level consistent<br />

with the major histologic category of PDWA and/or AH.<br />

P5-01-09<br />

Identification of Molecular Apocrine Triple Negative <strong>Breast</strong><br />

<strong>Cancer</strong> Using a Novel 2-Gene Assay and Comparison with<br />

Androgen Receptor Protein Expression and Gene Expression<br />

Profiling by DASL<br />

Alvarez J, Schaffer M, Karkera J, Martinez G, Gaffney D, Bell<br />

K, Sharp M, Wong J, Hertzog B, Ricci D, Platero S. Janssen<br />

Pharmaceuticals<br />

Background: The molecular apocrine (MA) subtype of breast cancer<br />

is identified by gene expression profiling. MA tumors are estrogen<br />

www.aacrjournals.org 423s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

receptor (ER) negative and progesterone receptor (PR) negative, but<br />

still express estrogen responsive genes. The androgen receptor (AR)<br />

pathway may be driving growth in these tumors because androgen<br />

responsive genes are expressed in tumors with the MA gene signature.<br />

The MA gene signature is identified in approximately 10% of triple<br />

negative breast cancer (TNBC) and may predict patients with<br />

tumors responsive to agents that inhibit the AR pathway. AR protein<br />

expression, measured by immunohistochemistry (IHC), may be a<br />

surrogate for the MA gene signature, but to date, a careful comparison<br />

of gene expression profiles and AR protein expression has not been<br />

conducted. In this study, cohorts of TNBCs were assessed for the<br />

MA gene signature and these results were compared with AR IHC<br />

expression and with a novel gene expression assay that may predict<br />

tumors with the MA gene signature.<br />

Methods: Formalin fixed, paraffin-embedded (FFPE) TNBC samples<br />

were commercially obtained. ER, PR and HER2 status of these<br />

samples was confirmed by IHC. AR expression was detected by<br />

IHC using two different antibody clones. Both staining intensity<br />

and percent positive cells were recorded for each sample. Gene<br />

expression data was collected from a cohort of TNBC FFPE samples<br />

using cDNA-mediated Annealing, Selection, extension, and Ligation<br />

(DASL) technology. A 2-gene classifier of the MA gene expression<br />

signature was derived by interrogating publically available gene<br />

expression data from ER-negative breast cancers. A reversetranscriptase<br />

polymerase chain reaction (RT-PCR) assay to detect<br />

the 2-gene classifier was developed. Cell lines predicted to have the<br />

MA gene signature by the 2-gene assay were tested for sensitivity<br />

to R-1881 in vitro.<br />

Results: Using computational approaches and publically available<br />

datasets, we confirmed the validity of the MA gene signature and<br />

estimated the prevalence to be between 12% and 37% in ER-negative<br />

breast tumors. The 2-gene classifier was 100% specific in determining<br />

MA tumors in a training set using gene expression data as a standard.<br />

In a validation set, the 2-gene assay was 66% correlative with AR<br />

IHC positivity when the IHC cut-off was set at 10% positive tumor<br />

cells. Cell lines predicted to express the MA gene signature by the<br />

2-gene classifier proliferated in response to androgen. This effect was<br />

blocked by Flutamide.<br />

Conclusions: These results indicate that AR IHC using a 10% cut-off<br />

may not completely correlate with the MA gene signature. Further<br />

refinement of AR IHC scoring criteria may produce greater specificity.<br />

Cell proliferation data suggests the 2-gene assay can predict tumors<br />

that will proliferate in response to androgen. Work is ongoing to<br />

determine the correlation between the 2-gene assay results, AR IHC<br />

and DASL gene expression data to fully understand the predictability<br />

of this assay. Understanding this correlation may allow use of simple<br />

clinical assays to accurately select patients responsive to agents that<br />

block AR signaling.<br />

P5-01-10<br />

Clinical-pathological features and outcomes of Invasive lobular<br />

(ILC) vs Invasive ductal (IDC) breast cancer (BC): a monoinstitutional<br />

series.<br />

Ferro A, Eccher C, Triolo R, Caldara A, Di Pasquale MC, Russo LM,<br />

De Carli NL, Cuorvo LV, Barbareschi M, Gasperetti F, Berlanda G,<br />

Pellegrini M, Moroso S, Galligioni E. S Chiara Hospital, Trento,<br />

Italy; FBK, Trento, Italy<br />

Purpose:<br />

The aim of our study was to investigate different clinico-biological<br />

behavior associated to ILC compared to IDC evaluating implications<br />

on survival outcomes<br />

Methods: We analyzed data from 3749 cases of IBC treated from<br />

1995 to 2008 and categorized as having ILC, IDC and mixed/other.<br />

Associations between clinical-pathological variables and the ILC/<br />

IDC histotype were assessed using a χ 2 test. Log-rank test and Cox<br />

regression model were performed to evaluate the impact of hystologic<br />

types on overall survival (OS), Event Free Survival (EFS) and Post-<br />

Progression Survival (PPS).<br />

Results: We identified 445 (12%) ILC, 3021 (80.5%) IDC, 149<br />

mixed (ductal-lobular) BC (3.9%) and 134 other hystotypes (3.6%).<br />

Median age was 61 (25-97) years. Compared with IDC, ILC occurred<br />

significantly (p3 ILC pts, in whom a trend of worse prognosis (55.6<br />

vs 60.2%) was observed.<br />

Conclusions: Despite the quite favorable biological pattern, ILC did<br />

not show a better outcomes than IDC.<br />

P5-01-11<br />

‘Same-day diagnosis’ of women suspected of breast cancer:<br />

success rate and impact on diagnostic quality and patients’<br />

anxiety levels<br />

Barentsz MW, Wessels H, van Diest PJ, Pijnappel RM, van der Pol<br />

CC, Witkamp AJ, van den Bosch MA, Verkooijen HM. University<br />

Medical Center Utrecht<br />

Introduction:<br />

Uncertainty about a breast cancer diagnosis is a stressful event. As<br />

such, many initiatives have been undertaken to reduce the time of<br />

uncertainty by providing ‘same-day diagnosis’ for patients suspected<br />

of breast cancer. However, only few studies have evaluated the<br />

feasibility of ‘same-day diagnosis’ diagnosis, while the impact of<br />

‘same-day diagnosis’ on diagnostic accuracy and patient anxiety has<br />

hardly been studied at all. This study aims to evaluates feasibility,<br />

diagnostic accuracy and impact on patients’ anxiety of ‘same-day<br />

diagnosis’ of patients suspected of breast cancer.<br />

Materials and Methods:<br />

In November 2011, the ‘same-day diagnosis’ system was implemented<br />

in our hospital. This system consists of completing the following<br />

items on the day of the first visit:<br />

1. Clinical examination, imaging (mammography, ultrasound), and<br />

(if necessary) FNA or core needle biopsy<br />

2. High speed processing of biopsy specimens using Leica Peloris<br />

tissue processor<br />

3. Multidisciplinary evaluation of clinical, imaging and histological<br />

findings<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

424s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 5<br />

4. Communication of the final diagnosis and treatment plan to the<br />

patient<br />

All patients presenting for ‘same-day diagnosis’ are entered<br />

prospectively in a database. Feasibility of ‘same-day diagnosis’ is<br />

evaluated, and reasons for failure are scrutinized. Diagnostic quality<br />

of ‘same-day diagnosis’ is evaluated by linking all patients to PALGA<br />

(Dutch National Pathology Database) six months following the day<br />

of diagnosis, in order to identify false negative results (i.e. missed<br />

cancers) and false positive results (i.e. ‘cancers’ turned out to be<br />

benign during further work-up). Impact of ‘same-day diagnosis’<br />

on patient anxiety is measured by the 4-item State Trait Anxiety<br />

Inventory, which is filled out by patients on several occasions on<br />

the day of diagnosis as well as during the first seven days following<br />

diagnosis.<br />

Results<br />

Between November 2011 and May 2012, 266 women were eligible<br />

for ‘same-day diagnosis’. Fifty-eight patients (22%) were diagnosed<br />

with breast cancer. Overall, 80% of the patients received a final<br />

diagnosis within one day. For patients with cancer, this proportion<br />

was somewhat lower, i.e. 71%. Accuracy of ‘same-day diagnosis’ and<br />

impact on patients’ anxiety are currently under study.<br />

Conclusion<br />

’Same-day diagnosis’ is feasible for the far majority of patients<br />

suspected of breast cancer. Evaluation of implementation of ‘sameday<br />

diagnosis’ on diagnostic quality and impact on patient anxiety is<br />

crucial. Updated results will be presented in December 2012.<br />

P5-01-12<br />

Over- and Undergrading of <strong>Breast</strong> <strong>Cancer</strong> on Core Biopsies in<br />

Comparison to Surgical Specimens<br />

Decker T, Focke CM, Gläser D, Finsterbusch K. Bonhoeffer Medical<br />

Center, Neubrandenburg, Germany<br />

Background<br />

In numerous studies histological grade (HG) remained an independent<br />

prognostic factor for ER positive tumours even after the inclusion<br />

of gene signatures in the multivariate models. Moreover, grade 3 is<br />

judged as clear argument in favour of chemotherapy. Therefore, there<br />

are many reasons to provide patients and doctors with HG as important<br />

prognostic and predictive information already preoperatively. To<br />

evaluate the relevance of histological grading (HG) based on core<br />

biopsies (CB) for clinical decision making in breast cancer (BC) we<br />

evaluated the concordance with HG from surgical specimen (SSP)<br />

and the reasons for under- or overgrading.<br />

Methods<br />

CB and related SSP of 398 BCs were prospectively graded according<br />

to the Nottingham grading system (SSP: 65 G1, 162 G2 and 171<br />

G3). CB/SSP agreement rates and positive predictive values (PPV)<br />

of CB based HG were calculated. Rates of over- and underestimation<br />

of glandular differentiation (GD), nuclear pleomorphism (NP) and<br />

mitotic activity (MA) were calculated.<br />

Results<br />

CB/SSP agreement rates came out with 95% for G1, 73% for G2, and<br />

56 % for G3. The PPVs of CB based HG were 56% for G1, 61% for<br />

G2 and 100% for G3. The overgrading and undergrading rates on CB<br />

were 1% and 37%, respectively. Main causes for undergrading in CB<br />

were underestimated MA (40%) and a combination of underestimated<br />

NP and MA (32%).<br />

Conclusions<br />

Whereas overgrading on CB is an exception, undergrading derives<br />

largely from underestimation of proliferation. G3 assessed on<br />

CB came out with the highest PPV, providing a reliable base for<br />

preoperative decisions i.e. about primary chemotherapy. Reversely,<br />

because of the relative low PPV of G1 on CB one has to be aware of<br />

potential upgrading on surgical specimen later on.<br />

P5-01-13<br />

Flat Epithelial Atypia: Management and outcome in three Dutch<br />

teaching hospitals<br />

Ghuijs PM, de Vries B, Strobbe LJA, van Deurzen CHM, Heuts EM,<br />

Keymeulen KBMI, Lobbes MBI, Wauters CAP, Van de Vijver KKBT,<br />

Smidt ML. Maastricht University Medical Centre, Maastricht,<br />

Netherlands; Canisius-Wilhelmina Hospital, Nijmegen, Netherlands;<br />

Erasmus Medical Centre, Rotterdam, Netherlands<br />

Background: Flat Epithelial Atypia (FEA) is a presumably<br />

neoplastic alteration of terminal duct-lobular units, characterized<br />

by the replacement of native luminal epithelium by ductal cells<br />

demonstrating low-grade cytologic atypia. The architecture shows<br />

stratification of epithelial cells. FEA is often accompanied by<br />

microcalcifications and therefore discovered in biopsies following<br />

screening mammography. FEA is frequently seen in association with<br />

ADH (atypical ductal hyperplasia), DCIS (ductal carcinoma in situ),<br />

lobular neoplasia and invasive tubular carcinomas. There is emerging<br />

evidence suggesting FEA may represent a precursor to DCIS. The<br />

risk of subsequent breast carcinoma remains to be defined. The aim of<br />

this study is therefore to inventorise the management and outcome of<br />

solitary FEA in histological biopsies in three Dutch teaching hospitals.<br />

Materials and Methods: Data of this retrospective multicentre<br />

study were collected in a database. Local pathology databases were<br />

screened with the terms: ‘FEA’, ‘Flat Epithelial Atypia’, ‘columnar<br />

atypia’ and Dutch equivalents. Results were manually screened, only<br />

including solitary FEA.<br />

Patient files were viewed for information on presentation,<br />

mammography, ultrasound and management: surgery vs follow-up.<br />

In case of excision, definitive pathology was added.<br />

Results: We included 103 patients showing only solitary FEA in the<br />

primary biopsy. Management of these patients consisted of followup<br />

for 60 patients (58,3%) and surgery for 43 patients (41,7%, 49<br />

excisions): lumpectomy (42) or mastectomy (7). Reason for choosing<br />

mastectomy was preventive in case of contralateral breast cancer or<br />

increased familial or genetic risk.<br />

Definitive pathology of lumpectomy or mastectomy showed no<br />

abnormalities or solitary FEA in 31 patients; other findings were<br />

ADH in 7, LCIS in 4 and DCIS in 7 patients. Some patients showed<br />

more than one finding. Invasive breast cancer (IBC) was detected in<br />

3 patients. Only one mastectomy showed invasive disease, located<br />

in a different lobe, however.<br />

No incidents occurred in the follow-up group.<br />

Conclusions: No consistent management exists concerning solitary<br />

FEA in these three hospitals. Also, one hospital used the diagnosis<br />

of FEA inconsistently and interchangingly with other terms. Lack of<br />

this study is the retrospective gathering of data, making it difficult<br />

to detect the reasons for the chosen management. DCIS or IBC was<br />

discovered in 20,4% of all surgical specimens. It was concluded that<br />

FEA should be seen as a red flag, indicating the possible presence of<br />

a more malignant lesion. Additional research is warranted concerning<br />

long term follow-up for this patient group.<br />

www.aacrjournals.org 425s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P5-01-14<br />

Discrepancy between routine and expert pathologists in<br />

histological assessment of early stage non palpable breast cancer<br />

and its impact on loco regional and systemic treatment<br />

Postma EL, Verkooijen HM, van Diest PJ, Willems SM, van den Bosch<br />

MA, van Hillegersberg R. UMC Utrecht<br />

Background<br />

Histopathological parameters are essential for deciding on further<br />

adjuvant treatment in patients with breast cancer after surgery.<br />

Here, we assessed the impact of inter observer variability on further<br />

treatment strategy in patients with clinically node negative, non<br />

palpable breast carcinomas.<br />

Methods<br />

Clinical and histopathological data of 314 patients with clinically<br />

node negative non palpable invasive breast cancer were analysed.<br />

Histological assessment of the primary tumour and sentinel nodes was<br />

first performed in a routine setting, subsequently central review took<br />

place. In cases of discordance between local en central assessment,<br />

we determined the impact on locoregional and systemic treatment<br />

strategy.<br />

Results<br />

Discordance between local and central review was seen in 13%<br />

of the patients (kappa 0.60 95% CI 0.50-0.71) for type, in 12%<br />

(k=0.796 95% CI 0.73-0.86) for grade, in 1% for ER status (k=0.898<br />

95% CI 0.80-1.0), in 2% for PR status(0.940 95% CI 0.89-0.99).<br />

Discrepancy in the assessment of the sentinel node(s) was seen in<br />

2% of the patients. (k=0.954 95% CI 0.92-0.98). Practising current<br />

Dutch Guidelines, loco regional treatment would be affected in 2%<br />

(7/310), systemic treatment in 5% (16/310) and both in 1% (2/310)<br />

of the patients.<br />

For patients in whom central review would have led to additional<br />

systemic treatment (3%), the 10 years mortality and recurrence rate<br />

would decrease with a median of 4.6% and 15% respectively.<br />

Conclusion<br />

Inter observer variation in the histopathological assessment of nonpalpable<br />

breast carcinoma specimens and sentinel nodes results in<br />

a different loco regional or systemic treatment advice in 8% of the<br />

patients.<br />

P5-01-15<br />

Anti-ER monoclonal antibody SP1 seems more sensitive in the<br />

identification of ER alpha positive breast cancer cases than anti-<br />

ER monoclonal antibody 1D5<br />

Madeira KP, Daltoé RD, Sirtoli GM, Rezende LCD, Carvalho AA,<br />

Silva IV, Rangel LBA. Universidade Federal do Espírito <strong>San</strong>to,<br />

Vitória, ES, Brazil<br />

Background:<br />

Accurate pathological analysis of biopsies is crucial for conclusive<br />

and precise disease diagnosis, prognosis and therapeutic guidelines.<br />

Evaluation of estrogen receptor (ER) expression in breast cancer<br />

(BC) is imperative in determining the disease prognosis, and patients’<br />

eligibility for hormonotherapy. Herein, ER alpha status was evaluated<br />

in 61 BC cases using two different anti-ER alpha monoclonal<br />

antibodies, SP1 (rabbit) and 1D5 (mouse).<br />

Methods:<br />

Primary BC cases registered in two reference hospitals, with<br />

correspondent written informed consent, were used to generate in<br />

house tissue microarray platforms. ER alpha expression was accessed<br />

by immunohistochemistry using the monoclonal antibodies anti-ER<br />

alpha, SP1 and 1D5. Staining scoring followed the Allred method (PS,<br />

proportion score or percentage of stained cells; IS, intensity score).<br />

Concordance and correlation parameters were calculated using Kappa<br />

factor; Pearson, Spearman’s, or intraclass correlation coefficient (CF).<br />

Staining patterns were compared by paired T-Test and Wilcoxon test.<br />

Findings:<br />

SP1 and 1D5 provided equivalent results (Concordance rate,<br />

96.7%; Kappa factor, 0.921), whereas SP1 was more prone for<br />

positive results than 1D5 (2 samples diverged). Total concordance<br />

of PS was obtained (Pearson and intraclass CF, 0.7351 and 0.6193,<br />

respectively); however, concordance between the antibodies seems<br />

more accurate in higher PS values. Excellent SI correlation between<br />

antibodies was observed throughout the population (Spearman’s<br />

CF, ρ=0.9150). Following the Allred score, 17 out of 42 positive<br />

BC samples diverged; always pointing 1D5 to weaker staining than<br />

SP1. When calculating Spearman’s CF of Total Score (TS) within<br />

the population, excellent correlation between both the antibodies<br />

(ρ=0.9238) was noted; nonetheless, results were less concordant<br />

result among the BC positive cases (ρ=0.7743). Indeed, 20 samples<br />

were differentially classified using the antibodies (only 3 had higher<br />

TS with 1D5). Considering the mean TS of all samples, or of invasive<br />

ductal carcinoma, SP1 provided higher scores than 1D5 (p


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 5<br />

microscope show a regular shape. In fibroadenoma, phyllodes tumor<br />

and mastitis team, they shows unobviously sonographic features and<br />

the cell morphology contrast with breast cancer are quite different.<br />

Conclusions :Ultrasonography appearance of breast masses is related<br />

to pathologic characteristics and can be a predictor of histopathologic<br />

pattern preoperatively.<br />

P5-02-01<br />

Discrepancy between CT and FDG-PET/CT in the staging of<br />

patients with inflammatory breast cancer: Implications for<br />

radiation therapy treatment planning<br />

Jacene H, DiPiro P, Bellon J, Nakhlis F, Hirshfield-Bartek J, Yeh<br />

E, Overmoyer B. Dana-Farber <strong>Cancer</strong> Institute, Harvard Medical<br />

School, Boston, MA<br />

Background: To compare suspected areas of cancer involvement based<br />

on the findings of CT and FDG-PET/CT in patients with inflammatory<br />

breast cancer (IBC) and to determine if discrepancies would modify<br />

radiation therapy treatment fields.<br />

Methods: This is a retrospective study of 29 consecutive female<br />

patients with IBC (median age 49 years, range 28-78) who had both<br />

CT and PET/CT scans prior to the initiation of systemic therapy.<br />

CT and PET/CT scans were obtained within a median of 3 days<br />

(range 0-64). CT and PET/CT scans were independently reviewed<br />

by a board-certified radiologist (PD) and a board-certified nuclear<br />

medicine physician (HJ), respectively. Findings were recorded by<br />

anatomic site and graded as negative, equivocal or positive for<br />

malignancy. Radiation fields were then determined by a breast<br />

radiation oncologist (JB) after separately reviewing the CT and PET/<br />

CT images. Discrepancies between the two modalities were recorded.<br />

The study was approved by the hospital institutional review board.<br />

Results: Seven of 29 patients (24%) had discrepant findings that<br />

likely would have resulted in a modification of radiation treatment<br />

fields and/or radiation dose. In four patients, internal mammary<br />

nodal involvement was suspected on PET/CT but not on CT. In<br />

two of these four patients, PET/CT also detected additional disease<br />

(supraclavicular in one patient and chest wall in the other) that<br />

likely would have required a change in radiation field and/or dose.<br />

Another patient had subpectoral adenopathy on PET/CT that was not<br />

considered abnormal on CT. One patient had equivocal findings in<br />

the infraclavicular region on CT which was negative on PET/CT. In<br />

one patient, body habitus limited the CT evaluation; more extensive<br />

infiltrative soft tissue and nodal disease was seen on PET/CT.<br />

In four patients, there were discrepant findings for distant disease<br />

that likely would have resulted in a change in management or<br />

additional imaging studies. In two of these cases, PET/CT showed a<br />

clearly positive finding that was negative or equivocal on CT. In an<br />

additional two cases, equivocal PET/CT findings would have resulted<br />

in additional testing that would not have been recommended by CT<br />

alone. In another 4 cases, metastatic disease was suspected based<br />

on CT, but additional sites (all in bone and not seen on CT) were<br />

suspected on PET/CT.<br />

Conclusions: Inflammatory breast cancer patients have a high<br />

probability of presenting with extensive local/regional disease and<br />

distant metastases. In our IBC population, PET/CT imaging would<br />

have likely resulted in a change of radiation field or dose in 7 of 29<br />

(24%) patients. Additionally, in four instances PET/CT suspected<br />

additional metastatic disease (14%). These discrepancies most<br />

commonly involved inclusion of an internal mammary nodal radiation<br />

field and identification of distant metastases. Pathologic verification<br />

of abnormal findings is necessary to verify these results. PET/CT<br />

imaging should be considered a standard component of radiographic<br />

staging for patients with IBC.<br />

P5-02-02<br />

Factors influencing time to seeking medical advice and start of<br />

treatment in breast cancer (BC) patients – an International survey<br />

Jassem J, Ozmen V, Bacanu F, Drobniene M, Eglitis J, Kahan Z,<br />

Lakshmaiah K, Mardiak J, Pienkowski T, Semiglazova T, Stamatovic<br />

L, Timcheva C, Vasovics S, Vrbanec D, Zaborek P. Medical University<br />

of Gdansk, Poland; University of Istanbul, Turkey; Sf Maria Hospital,<br />

Bucharest, Romania; Institute of Oncology, Vilnius University, Vilnus,<br />

Lithuania; Riga East University Hospital, Riga, Latvia; University of<br />

Szeged, Hungary; Kidwai Memorial Institute of Oncology, Bangaluru,<br />

India; National <strong>Cancer</strong> Institute and Medical School of Comenius<br />

University, Bratislava, Slovakia (Slovak Republic); Medical Center<br />

of Postgraduate Education, ECZ, Otwock, Poland; Petrov Research<br />

Institute of Oncology, St. Petersburg, Russian Federation; Institute of<br />

Oncology and Radiology, Belgrade, Serbia; Chemiotherapy Clinic,<br />

Sofia, Bulgaria; Zagreb University Hospital Center, Zagreb, Croatia;<br />

Warsaw School of Economics, Warsaw, Poland<br />

Background. We earlier reported patient-related factors influencing<br />

the delay in first medical advice for signs of BC in selected countries (J<br />

Clin Oncol 2012;30[15S]:578s). The present analysis, which includes<br />

additional countries, addresses the factors influencing time from first<br />

symptoms of BC to initiation of treatment (Total Delay Time; TDT).<br />

Methods. A total of 6,588 female BC patients from 11 countries<br />

were surveyed using a uniform questionnaire translated into local<br />

languages. TDT was determined using 8 individual scales, including<br />

one pertaining to patient delay and 7 related to subsequent steps in<br />

a typical diagnostic process. Regression models were constructed<br />

using 18 variables concerning diverse contextual and personal patient<br />

characteristics. Due to expected differences in the relevant sets of<br />

predictors, time between first symptoms and first medical visit (Patient<br />

Delay Time; PDT) and time between first medical visit and start of<br />

therapy (System Delay Time; SDT) were modeled separately, with<br />

multilevel regression.<br />

Results. Mean TDT varied in individual countries from 11.5 to 29.4<br />

weeks (grand mean of 14.3 weeks; see table), with 43% of cases<br />

with a delay of >12 weeks. Multilevel regression equation indicated<br />

that factors significantly correlated with longer PDT were distrust in<br />

the medical system and ignoring disease. Patients with fear of the<br />

disease, stronger self-examination habits, at least secondary education,<br />

being employed, reporting more support from friends and family, and<br />

living in cities of >100,000 inhabitants had shorter PDT. Predictors<br />

of shorter SDT included being diagnosed by an oncologist vs. other<br />

physician, having at least secondary education, being older than 60<br />

years, having a history of cancer in female relatives and having breast<br />

lump vs. other BC symptoms. Indicators of longer SDT were PDT >4<br />

weeks, more than one BC symptom detected by patient, distrust in<br />

the medical system, disregard of BC signs, less support from friends<br />

and family, and having first medical examination in a public vs.<br />

private medical center. Individual countries differed significantly with<br />

regard to intercept of the multilevel models and slopes of regression<br />

coefficients for selected psychological and behavioral attributes.<br />

Conclusions. An extensive set of variables potentially impacting<br />

delay times, mainly related to psychological and behavioral patient<br />

attributes, was examined. Several of them strongly determined<br />

both PDT and SDT, but their strength differed between individual<br />

countries. Both models (for PDT and SDT), although statistically<br />

significant, accounted for approximately 20% of variance in time;<br />

www.aacrjournals.org 427s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

therefore other variables, e.g. related to the differences in national<br />

healthcare systems (not addressed in this study) might have a stronger<br />

impact on delays in the initiation of BC therapy and warrant further<br />

analyses.<br />

Country Number of patients Mean PDT (weeks) Mean SDT (weeks) Mean TDT (weeks)<br />

Poland 1000 3.6 9.5 11.5<br />

Lithuania 458 4.9 8.3 12.2<br />

Serbia 800 4.5 9.3 12.9<br />

Slovakia 253 4.0 10.7 13.2<br />

Croatia 250 4.9 10.2 13.4<br />

Turkey 1031 4.8 10.5 13.8<br />

Russia 1059 4.8 12.4 15.7<br />

Bulgaria 644 4.8 12.5 15.9<br />

Latvia 156 6.2 13.1 17.5<br />

Romania 319 6.0 20.4 25.5<br />

India 268 6.1 24.7 29.4<br />

P5-02-03<br />

Large-scale genomic instability consistently identifies BRCA1/2<br />

inactivation in breast cancers<br />

Stern M-H, Popova T, Manié E, Dubois T, Sigal-Zafrani B, Bollet M,<br />

Sastre-Garau X, Vincent-Salomon A, Houdayer C, Stoppa-Lyonnet<br />

D. Institut Curie, Paris, France<br />

The goal of this study was to identify genomic markers predicting<br />

actual BRCA1/2 inactivation in breast cancers. By comparing<br />

genomic profiles of BRCA1/2 mutated and wildtype basal-like<br />

carcinomas (BLC) we developed a signature of BRCAness. The<br />

signature accounts for the level of the large scale genomic instability<br />

and was demonstrated to predict BRCA1/2 impairment with 100%<br />

sensitivity and 90% specificity in the large series of BLCs. We<br />

introduced a bioinformatics procedure evaluating the number of large<br />

scale rearrangements (denoted as Large Scale Transition or LST)<br />

in the genome and showed that the number of LSTs discriminate<br />

homologous recombination deficient and proficient tumors (Popova<br />

et al, <strong>Cancer</strong> Research 2012 Aug).<br />

The efficiency of the signature of BRCAness represented by the<br />

number of LSTs was then evaluated for the luminal breast tumors and<br />

ovarian carcinomas. Luminal breast tumors and ovarian carcinomas<br />

could be stratified into two groups according to the number of LSTs.<br />

The performance of the LST signature in all types of breast cancers<br />

will be presented.<br />

Measurement of LST is a highly reliable, simple and cost effective<br />

method to identify patients who should benefit of a BRCA1/2 testing,<br />

and to select tumors which may respond to treatment targeting DNA<br />

repair deficiencies (platinium salts, PARP inhibitors).<br />

P5-03-01<br />

<strong>Cancer</strong> stem cells predict engraftment and poor prognosis of<br />

primary breast tumors<br />

Charaffe-Jauffret E, Ginestier C, Bertucci F, Cabaud O, Wicinski<br />

J, Finetti p, Josselin E, Adelaide J, Nguyen T-T, Monville F,<br />

Jacquemier J, Thomassin-Piana J, Pinna G, Jalaguier A, Lambaudie<br />

E, Houvenaeghel G, Xerri l, Harel-bellan A, Chaffanet M, Viens P,<br />

Birnbaum D. CRCM-IPC; CEA<br />

<strong>Breast</strong> cancer is a major health problem and heterogeneity of the<br />

disease has been considered as a strong limitation to find the best<br />

therapies to cure cancer, overcome recurrences and metastases. The<br />

establishment of models that reflect tumor biology and metastatic<br />

progression is critical to develop successful new therapeutic strategies.<br />

In the breast, orthotopic xenografts currently appear as the best models<br />

to study tumor growth, metastasis and develop tools for prognosis<br />

prediction. Furthermore, mouse transplant assays have been used to<br />

assess cancer stem cell (CSC) activity and demonstrate that leukemia<br />

and many solid tumors are organized along a hierarchical model.<br />

Despite the promise of the CSC model sustained by mouse<br />

transplantation assays, the clinical relevance of xenografts studies to<br />

identify determinants of stemness able to influence clinical outcome<br />

remains challenging. In breast cancer, transcriptional programs from<br />

functionally validated CSC populations remain to be deciphered.<br />

Here, we report the establishment of a bank of primary breast tumorderived<br />

xenografts (xenobank). We showed that the xenografts retain<br />

the main features of primary tumors, that engraftment is correlated<br />

with the presence of CSC in tumors, and that engraftment in the<br />

mouse is able to predict prognosis in patients. This suggests that<br />

CSCs may govern breast cancer prognosis. We established the gene<br />

expression profiles of functionally validated ALDEFLUOR-positive<br />

CSC populations (breast CSC-GES) and demonstrated their clinical<br />

relevance. Among 2609 patients with breast cancers, we validated<br />

that he expression of the breast CSC-GES is correlated with poor<br />

outcome and metastasis in uni-and multivariate analysis (5-year<br />

MFS was 70% CI95 [67-74] in the breast CSC-positive class and<br />

80% (CI95 [77-83]) in the breast CSC-negative class (p=5.5E-04<br />

with log-rank test). Furthermore, we identified a core of 19 genes<br />

commonly expressed in breast CSC, murine embryonic, neural and<br />

hematopoietic stem cells programs and demonstrated for each gene<br />

its ability to modulate breast CSC population, being implicated in<br />

self-renewing or differentiation programs. We found that the core of<br />

genes in common between four stem cell gene expression studies<br />

(CE-BCSC-3SC) displayed an adverse prognostic impact for patients<br />

with breast cancer. The core contained genes implicated in oxidative<br />

phosphorylation, detoxification, lipid metabolism, and genomic<br />

stability, and these shared determinants of stemness influenced<br />

clinical outcome.<br />

Thus, we show ed that CSCs from orthotopically engrafted primary<br />

breast tumors have clinical and biological relevance. This functionally<br />

validated CSC population is highly correlated with survival and<br />

express genes governing main stem cell functions, substantiating a<br />

major prediction of the CSC model and opening further promises for<br />

new CSC therapies using valid preclinical models.<br />

P5-03-02<br />

Targeting mRNA Translation to Enhance the Radiosensitivity of<br />

Inflammatory <strong>Breast</strong> <strong>Cancer</strong> Stem Cells<br />

Silvera D, Connolly EP, Volta V, Arju R, Venuto T, Schneider RJ.<br />

NYU School of Medicine, New York, NY; NYU <strong>Cancer</strong> Institute, NYU<br />

School of Medicine, New York, NY; Columbia University College of<br />

Physicians and Surgeons, New York, NY<br />

Purpose/Objective(s): Inflammatory breast cancer (IBC) is a highly<br />

aggressive and radiation resistant malignancy with a dismal prognosis<br />

despite multimodality therapy, including ionizing radiation. We have<br />

previously shown that the unique pathogenic properties of IBC result<br />

in part from over-expression of translation initiation factor eIF4G1,<br />

which is part of the eIF4F translation initiation complex, along with<br />

eIF4E and eIF4A. eIF4F is regulated by mTOR, providing a promising<br />

target for anti-cancer therapeutics. We demonstrated that protein<br />

synthesis is highly regulated during IR by the DNA-damage response<br />

(DDR) pathway through mTOR signaling. Many key proteins required<br />

for the DDR pathway are encoded by mRNAs that require high levels<br />

of the eIF4F complex and mTOR activity for their efficient translation.<br />

We hypothesized that upregulation of eIF4F in IBC plays a crucial<br />

role in the radio-resistance of disease.<br />

Materials/Methods: Experiments were conducted in IBC SUM149<br />

cells. eIF4G1, eIF4E and eIF4A were silenced through the generation<br />

of stable cell lines that express tetracycline-inducible shRNAs. eIF4A<br />

was also inhibited using the pharmacologic investigational inhibitor<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

428s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 5<br />

DAMD-PatA. Radiation sensitivity in vitro was determined by cell<br />

survival assay. Tumor xenografts were generated by the injection<br />

of stable shRNA inducible cell lines into nude mice. IBC SUM149<br />

cancer stem cells (CSC) from both in vitro and in vivo experiments<br />

were analyzed by a combination of cell surface marker analysis,<br />

mammosphere formation and Aldefluor assays.<br />

Results: We show that moderate inhibition by silencing of individual<br />

components (or by pharmacologic inhibition of eIF4A) of the<br />

eIF4F complex prevents IBC xenograft tumor growth and strongly<br />

enhances radiosensitivity. In contrast to results obtained for nontransformed<br />

breast epithelial cells, reducing the high levels of eIF4G1<br />

in epithelial IBC cells in 2D cultures provides no enhancement in<br />

radiation sensitivity. Rather, SUM149 IBC cells harbor a substantial<br />

population of CSCs, which are the cells that are strongly dependent<br />

on high levels of eIF4G1, and which are selectively radio-sensitized<br />

as a result of eIF4G1-silencing. CSCs also require eIF4E and eIF4A<br />

activity in order to survive radiation treatment. We also demonstrate<br />

that silencing of eIF4G1 radio-sensitizes the stem cell population<br />

within IBC tumor xenografts. Radio-resistance of IBC cells is likely<br />

mediated by differential responses to the DDR in the stem-cell<br />

populations and by selective mRNA translation of proteins involved<br />

in the DDR pathway.<br />

Conclusions: Our results demonstrate that regulation of mRNA<br />

translation plays an important role in conferring radio-resistance to<br />

advanced breast cancers, particularly by allowing the survival of the<br />

CSC compartment. While inhibition of eIF4F enhances radiation<br />

sensitivity in non-transformed cells, this process is abrogated in<br />

IBC due to enrichment of a radiation resistant CSC population,<br />

demonstrating translational control of the breast cancer stem cell<br />

population, and providing a novel understanding of the role of the<br />

regulation of mRNA translation in radiation resistance of breast<br />

cancer.<br />

P5-03-03<br />

Antitumor Activity and <strong>Cancer</strong> Stem Cells Effect of Cetuximab<br />

in Combination with Ixabepilone in Triple Negative <strong>Breast</strong><br />

<strong>Cancer</strong>s (TNBC)<br />

Tanei T, Rodriguez AA, Dobrolecki L, Choi DS, Landis M, Chang JC.<br />

The Methodist Hospital Research Institute and Weill Cornell Medical<br />

School, Houston, TX<br />

Purpose: The ErbB family, including EGFR, has been demonstrated to<br />

play key roles in metastasis, tumorigenesis, cell proliferation, and drug<br />

resistance. Recently, these characteristics have been linked to a small<br />

subpopulation of cells classified as cancer stem cells (CSCs) which<br />

are believed to be responsible for tumor initiation and maintenance.<br />

Ixabepilone is the microtubule-stabilizing agent has been expected<br />

to be more sensitive than the conventional taxanes. The aim of this<br />

study was to investigate whether the EGFR monoclonocal antibody<br />

cetuximab, in combination with ixabepilone is a more effective<br />

treatment, and kill cancer stem cells more effectively as compared<br />

to chemotherapy alone in TNBC.<br />

Experimental Design and Results: <strong>Breast</strong> CSC populations were<br />

evaluated with FACS analysis (CD44+ and CD24-/low, or Aldefluor+)<br />

and mammosphere formation efficiency (MSFE). In vitro, we<br />

demonstrated that in triple negative cell lines (MDA-MB-231 and<br />

SUM159), cancer stem cell populations were decreased after treatment<br />

of cetuximab, or cetuximab plus ixabepilone. In vivo, cetuximab in<br />

combination with ixabepilone treatment caused significant tumor<br />

regression (cetuximab vs. cetuximab+ ixabepilone; tumor volume<br />

fold change P


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P5-03-05<br />

Histone deacetylase (HDAC)-inhibitor mediated reprogramming<br />

drives cancer cells to the pentose phosphate metabolic pathway<br />

Debeb BG, Larson RA, Lacerda L, Xu W, Smith DL, Ueno NT, Reuben<br />

JM, Gilcrease M, Krishnamurthy S, Buchholz TA, Woodward WA. MD<br />

Anderson <strong>Cancer</strong> Center, Houston, TX<br />

Recent studies have shown that energy metabolism in human<br />

pluripotent cells contrasts sharply with energy metabolism in<br />

differentiated cell types. Specifically, it has been shown that nuclear<br />

reprogramming from somatic cells to induced pluripotent stem cells<br />

is associated with a switch from oxidative to glycolytic metabolism.<br />

Whether a metabolic switch also occurs in reprogrammed/<br />

dedifferentiated breast cancer cells is unknown. Moreover, the<br />

function of the metabolic state in stemness is poorly understood and<br />

no data are available on whether breast cancer stem cells (CSCs)<br />

are metabolically different from committed cancer cells. Herein we<br />

demonstrated that HDAC inhibitors reprogram committed single<br />

aldefluor negative breast cancer cells into aldefluor positive cells<br />

(10.3 ± 2.8 vs 21.3 ±3.7% untreated vs treated P


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 5<br />

paclitaxel (ip on days 1, 4 and 8) or both drugs when tumors reached<br />

0.5 cm 2 . Statistical analysis was performed using one-way ANOVA<br />

with the Bonferroni multiple comparison test. RESULTS: Activity of<br />

the HH measured by the expression of Gli1 was increased in BCSC<br />

cell lines as compared to E cell line (p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

HIF-1α DN construct. Moreover, SUM-149 cells expressing the HIF-<br />

1α DN construct exhibited delayed tumor growth in vivo (p = 0.05).<br />

Standard clonogenic assays demonstrated that PyMT HIF-1α KO<br />

cells and SUM-149 cells expressing the HIF-1α DN construct were<br />

more sensitive to radiation therapy (SF6 of PyMT, HIF-1α:Control,<br />

0.016:0.087), but that the PyMT KO and SUM-149 mammospheres<br />

that persisted after radiation were completely radioresistant (SF6<br />

of PyMT, HIF-1 α: Control, 0.48:0.44). In contrast, MCF-7 cells<br />

were not radiosensitized in either standard or mammosphere assays.<br />

Interestingly, in HIF-1α DN MCF-7 cells, molecular features of<br />

IBC were observed, such as the increased expression of E-Cadherin<br />

and loss of Wisp3. But, Notch1 protein expression was unchanged<br />

between HIF-1α DN MCF7 or SUM149 cells. Moreover, concurrent<br />

radiation in the presence of a gamma secretase inhibitor or with<br />

a p53-MDM2 inhibitor nutlin failed to radiosensitize HIF KO<br />

mammosphere clonogens. We conclude that downregulation of HIF-1<br />

activity selectively radiosensitizes IBC clonogenic cells but fails to<br />

radiosensitize the residual mammospheres. These data suggest that<br />

the known HIF-1α mediated mechanisms that favor radiosensitivity,<br />

such as the promotion of glycolysis and proliferation under stress,<br />

may predominate in mammospheres, which ultimately leads to<br />

radioresistance in residual mammospheres after HIF inhibition.<br />

P5-03-11<br />

Possible role for cancer stem cells: results from a pilot neoadjuvant<br />

trial of HER-2 positive breast cancer patients treated with a<br />

combination of (Nab)-paclitaxel and lapatinib.<br />

Siziopikou KP, Gradishar WJ, Kaklamani VG. Northwestern<br />

University Feinberg School of Medicine, Chicago, IL<br />

Background: Lapatinib, a dual kinase inhibitor against both<br />

epidermal growth factor (EGFR) and human epidermal factor 2<br />

(HER-2) and nanoparticle albumin bound (nab) paclitaxel was<br />

recently used by our group in a pilot study of early stage HER-2<br />

positive breast cancer. We reported that the combination was well<br />

tolerated and showed good efficiency in this patient group. <strong>Cancer</strong><br />

stem cells were recently identified in solid tumors including breast<br />

carcinomas. These tumor initiating stem cells are reported to be<br />

increased in HER-2 positive breast cancers. Lapatininb is postulated<br />

to reduce the percentage of breast cancer stem cells following HER-<br />

2 inhibition. This study aimed to investigate such an effect in this<br />

patient population of early stage HER-2 positive breast cancer cases<br />

uniquely treated by a combination of nab-paclitaxel and lapatininb.<br />

Design: 30 patients with stage I-III HER-2 positive breast cancer<br />

were treated in a neoadjuvant setting with lapatinib 1,000mg/day and<br />

nab-paclitaxel 260 mg/m2 every 2 weeks for 4 cycles. The expression<br />

of the stem cell markers aldehyde dehydroganase (ALDH1) (BD),<br />

CD24 and CD44 (both <strong>San</strong>ta Cruz Biotechnology) was assessed<br />

immunohistochemically in breast cancer specimens prior to treatment<br />

and at the time of definitive surgery. Staining was considered negative<br />

if < or = 1%, and positive if >1%.<br />

Results: Of the 30 patients, 28 underwent surgery and were evaluated<br />

for pathologic response. Complete pathologic response (pCR) was<br />

observed in 5/28 (17.9%) of the patients. Of the 17 patients for whom<br />

pre-treatment material was available 13 (76.5%) were positive for<br />

ALDH1 expression and 4 (23.5%) were negative. Of the 22 patients<br />

with material available for testing at the time of surgery, 5 showed<br />

pCR, 3 were negative and 14 (63.6%) were positive for ALDH-A1<br />

expression. The CD44+/CD24- phenotype was variable and showed<br />

no difference between groups.<br />

Conclusions: 1. Combination of lapatinib and nab-paclitaxel resulted<br />

in a complete pathologic response in almost 1/5 of the HER-2 positive<br />

early stage breast cancer patients. 2. Overall, there is a reduction in<br />

the number cancer stem cells following neoadjuvant treatment with<br />

lapatinib and nab-paclitaxel. Our results support the hypothesis that<br />

lapatinib may have an effect in manipulating the numbers of cancer<br />

stem cells in patients with HER-2 positive breast cancer. Additional<br />

studies are currently under way to further characterize these findings.<br />

P5-03-12<br />

Targeting breast cancer stem cells using the autopahgy inhibitor<br />

N-Acetyl cysteine<br />

Dave B, Granados S, Mitra S, Chang JC. Methodist Hostpital<br />

Research Institute, Houston, TX<br />

Introduction: One in eight women is diagnosed with breast cancer<br />

in the United States. The most aggressive form of breast cancer is<br />

the “triple negative breast cancer” (TNBCs), where there is a lack<br />

of expression of all three receptors, namely ER, PR and HER2 and<br />

a lack of targeted therapies lead to the highest relapse rate in breast<br />

cancer. This makes it imperative to identify and target the mechanism<br />

of relapse of this cancer. We have recently identified autophagy<br />

as a mechanism of tumor resistant cell survival in breast cancer.<br />

We have demonstrated an increase in autophagy in chemotherapy<br />

treated patients. Further, we have shown that addition of a stem<br />

cell inhibitor against NOTCH reduces autophagy and stem cells<br />

population. In order to eliminate these tumor resistant cells from<br />

surviving chemotherapy, we plan to target the cell survival pathway of<br />

autophagy using N-acetyl csyteine as a novel inhibitor. Materials and<br />

Methods: We treated three triple negative cell lines (SUM159, BT549<br />

and MDA-MB231) with varying concentrations of N-acetyl cysteine<br />

and determined its impact on tumor initiating cells via mammosphere<br />

formation and FACS sorting of CD44hi/CD24low cells. N-acetyl<br />

cysteine affects mitochondrial metabolism so we tested its impact<br />

on mitochondrial DNA mass. Results: N-acetyl cysteine significantly<br />

decreased TIC population as evidenced by the remarkable reduction<br />

in mammosphere formation efficiency and levels of CD44hi/24low<br />

cells at 1 and 10uM in all three cell lines. In two cells lines we have<br />

demonstrated that there is a significant increase in mitochondrial mass<br />

upon treatment of NAC. Conclusion: We have currently determined<br />

that N-acetyl cysteine works via autophagy and eliminates tumor<br />

initiating cell population. We have also demonstrated that this<br />

involves changing the mitochondrial mass and overall changes in the<br />

metabolism of these cells. This novel interlink between mitochondrial<br />

metabolism and autophagy provided a new insight into the role of<br />

tumor initiating cells in breast cancer and possible new approaches<br />

to treat therapy resistance in triple negative breast cancer.<br />

P5-03-13<br />

Detection of tumor initiating cells (TIC) among the peripherally<br />

circulating epithelial tumor cells from patients with breast cancer<br />

Pachmann K, Zimon D, Pizon M, Carl S, Rabenstein C, Camara<br />

O, Pachmann U. University Hospital Jena, Germany; Transfusion<br />

Center, Bayreuth, Germany<br />

Background: The detection of tumor cells circulating in the peripheral<br />

blood of patients with breast cancer is a sign that cells have been able<br />

to leave the primary tumor and survive in the circulation. However,<br />

in order to form metastases they require additional properties such<br />

as the ability to adhere, self renew and grow. Here we present data<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

432s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 5<br />

that a variable fraction among the circulating tumor cells detected by<br />

the maintrac® approach express mRNA of the stem cell gene nanog<br />

and of the adhesion molecule vimentin and are capable of forming<br />

tumor spheres a property ascribed to TIC.<br />

Patients and methods: Between 10 to 50 circulating epithelial antigen<br />

positive cells detected by the maintrac® approach were selected<br />

randomly from each of 20 patients with breast cancer before and<br />

after surgery and isolated using automated capillary aspiration and<br />

deposited individually onto slides for expression profiling. In addition,<br />

the circulating tumor cells were cultured without isolation among the<br />

white blood cells from 5 patients with breast cancer in different stages<br />

of disease using culture methods favoring growth of epithelial cells.<br />

Results: Although no EpCAM mRNA positive cells expressing stem<br />

cell genes or the adhesion molecule vimentin were detected before<br />

surgery, 10 to 20% of the cells were found to be positive for mRNA<br />

of these genes after surgery. Most surprisingly, it was possible to grow<br />

tumor spheres (Fig 1) from these circulating cells without previous<br />

isolation in all patients in a fraction comprising between 1% of the<br />

epithelial antigen positive cells in patients with newly diagnosed<br />

tumor up to 10% in a patient with advanced breast cancer.<br />

Conclusion: We here show, that among the peripherally circulating<br />

tumor cells a variable fraction is able to express stem cell and adhesion<br />

properties and can be grown into tumor spheres, a property ascribed<br />

to cells capable of initiating tumors and metastases.<br />

P5-03-14<br />

Expression of ALDH1 in metastasizing axillary lymphnodes in<br />

breast cancer<br />

Shi A, Dong Y, Bi L, Xu N, Fan Z, Li S, Yang H, Li Y. First Hospital of<br />

Bethune Medical College, Jilin University, Changchun, Jilin, China;<br />

Lester & Sue Smith <strong>Breast</strong> Center, Baylor College of Medicine,<br />

Houston, TX<br />

Background: There is increasing evidences that a wide variety of<br />

malignancies, including breast cancer, may be driven by a small subset<br />

of ‘tumor-initiating cells’ or ‘cancer stem cells’ (CSC) which are able<br />

to form tumors in immunocompromised mice as well as to generate<br />

the phenotypic heterogeneity of the initial tumor. Enzyme aldehyde<br />

dehydrogenase (ALDH1) has been reported as a possible marker<br />

for mammary CSC. These cells are a source of tumor recurrence<br />

and metastasis, and are resistant to chemotherapy, radiotherapy and<br />

hormone therapy.<br />

Objective: Assuming that the detection of CSC in axillary lymph<br />

nodes is more effective to predicting cancer outcome than the widely<br />

used detection of cancer cells in axillary lymph nodes, we measure<br />

ALDH1 levels to predict their presence into axillary lymph nodes on<br />

development of cancer and anticipate outcomes.<br />

Methods: ALDH1 protein was detected by an immunohistochemical<br />

technique in 229 cases of breast cancer diagnosed from 2002 to 2011<br />

Follow-up ranged from 11.5 months to 96.9 months, with a mean of<br />

73.9 months. A survival assay was used to determine the relationship<br />

between distant metastatic rate and survival rate.<br />

Results: ALDHl expression was detected in 79cases and the Positive<br />

rate in metastatic axillary lymph nodes was 34.5%. Negative ER,<br />

PR status were related to the ALDH1 positive cases(P= 0.012). See<br />

Table 1. Mortality rate between ALDH1 positive cases (50.8%) and<br />

negative cases (28.8%) were significantly different (P= 0.001). See<br />

Table 2. Further, survival analysis of recurrence-free survivals (RFS)<br />

and survival rate decreased significantly between ALDHl positive and<br />

negative cases (P =0.001) (see table 2) and COX analysis shows that<br />

ALDH1 expression is an independent predictor of poor outcome in<br />

breast cancer(P =0.011).<br />

Discussion<br />

What cancer stem cells migrate to the axillary nodes have more<br />

important prediction than that the matastesis of normal cancar cells<br />

in axillary node. It might be a role resulting in dying in breast cancer.<br />

table1 Relationship between ALDH1 expression and clinicopathological parameters.<br />

clinical parameters ALDH+ ALDH- P VALUE<br />

All cases 79 150<br />

Age(years) 0.719<br />

≤50 42 76<br />

>50 37 74<br />

Menopause status 0.622<br />

pre 43 77<br />

post 34 70<br />

tumor size(cm) 0.384<br />

≤2 16 27<br />

2-5 50 106<br />

>5 13 16<br />

The number of 0.843<br />

≤3 36 73<br />

4-9 31 53<br />

>9 12 24<br />

STAGE(II, III, IV) 0.137<br />

II(Iia,Iib) 29 73<br />

III (IIIa, IIIb, IIIc) 47 75<br />

IV 3 2<br />

ER 0.012<br />

positive 46 113<br />

negative 31 36<br />

PR 0.139<br />

positive 49 109<br />

negative 28 40<br />

Her-2 0.192<br />

positive 22 31<br />

negative 55 118<br />

table 2 Relationship between ALDH1 expression and status.<br />

Status ALDH1+ ALDH1- P-VALUE<br />

Alive(group1) 48 121 0.001<br />

Death(group2) 31 29<br />

P5-04-01<br />

Expression of epithelial-mesenchymal transition (EMT)-related<br />

markers in primary tumors and matched lymph node metastases<br />

in breast cancer patients<br />

Zaczek AJ, Ahrends T, Markiewicz A, Seroczynska B, Szade J,<br />

Welnicka-Jaskiewicz M, Jassem J. Intercollegiate Faculty of<br />

Biotechnology, University of Gdansk and Medical University of<br />

Gdansk, Gdansk, Poland; Medical University of Gdansk, Pomorskie,<br />

Poland<br />

Background. Malignant tumors may include multiple cancer cell<br />

populations with various metastatic potential. More aggressive<br />

subpopulations might easier be captured in lymph nodes metastases<br />

(LNM) than in primary tumors (PT). We evaluated mRNA and protein<br />

levels of master EMT regulators: TWIST1, SNAIL and SLUG,<br />

protein levels of EMT-related markers: E-cadherin, vimentin, and<br />

expression of classical breast cancer receptors: HER2, ER and PR<br />

in PT and corresponding LNM. The results were correlated with<br />

clinicopathological data and patient outcomes.<br />

Methods. Formalin-fixed paraffin-embedded (FFPE) samples from<br />

PT and matched LNM from 42 stage II-III breast cancer patients<br />

were examined. Expression of TWIST1, SNAIL and SLUG was<br />

measured by RT-qPCR, with median as cut off for positive result.<br />

Protein expression was examined by immunohistochemistry on tissue<br />

microarrays. For TWIST1, SNAIL, SLUG, E-cadherin and vimentin<br />

>10% of positively stained cells defined positive result. ER and PR<br />

were scored according to Allred system, and HER2 - according to<br />

HercepTest criteria. Results were considered concordant if PT and<br />

LNM were both positive or both negative. Concordance was measured<br />

by estimating Cohen’s kappa coefficient (κ), with κ value equal 1<br />

indicating perfect agreement. Disease-free survival (DFS) and overall<br />

survival (OS) were compared using F-Cox test. Hazard ratios (HRs)<br />

with 95% confidence intervals (95% CI) were computed using Cox<br />

regression analysis.<br />

www.aacrjournals.org 433s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Results. On average, expression of TWIST1, SNAIL and SLUG<br />

was significantly higher in LNM compared to PT (P


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 5<br />

cancers with adjacent DCIS, expression of mesenchymal markers<br />

was increased in invasive component compared to DCIS component.<br />

CONCLUSIONS: Our study confirmed that EMT is an intrinsic<br />

characteristic of basal-like subtype and is associated with breast<br />

cancer with stem cell phenotype. However, increased expression of<br />

EMT-related markers in invasive carcinoma compared to pure DCIS,<br />

especially in basal-like subtype, and in the invasive component of<br />

basal-like invasive carcinoma with DCIS component also suggests<br />

a role of EMT in the transition of in situ- to invasive carcinoma in<br />

basal-like breast cancer.<br />

P5-04-04<br />

Significance of PELP1/HDAC2/microRNA-200 regulatory<br />

network in EMT and metastasis of breast cancer<br />

Roy SS, Gonugunta VK, Bandyopadhyay AM, Rao M, Goodall G, Sun<br />

L, Tekmal RR, Vadlamudi RK. UTHSCSA, <strong>San</strong> <strong>Antonio</strong>, TX; Centre<br />

for <strong>Cancer</strong> Biology, Adelaide, Australia<br />

Tumor metastasis remains a significant clinical problem and is the<br />

leading cause of death among breast cancer patients. Estrogen receptor<br />

(ER)-coregulators play an essential role in cancer progression and<br />

metastatic tumors express increased levels of coregulators. Proline<br />

glutamic acid rich protein (PELP1) is an ER coregulator, its expression<br />

is upregulated during breast cancer progression to metastasis and is an<br />

independent prognostic predictor of shorter survival of breast cancer<br />

patients. MicroRNA (miR) mediated regulation of tumorigenesis<br />

is emerging as a new paradigm in cancer biology and widespread<br />

misexpression of miRs has been reported in breast cancer. The<br />

objective of this study is to examine the mechanism and therapeutic<br />

significance of PELP1 regulation of miRs leading to breast cancer<br />

metastasis. We have used both ER+ve (ZR75, MCF7) and ER-ve<br />

(MDAMB231, MDAMB468) models that either stably overexpress<br />

PELP1 or PELP1shRNA. Boyden chamber, and invasion assays<br />

demonstrated that PELP1 down regulation significantly affect<br />

migration of both ER+ve and ER-ve cells. Epithelial to Mesenchymal<br />

Transition (EMT) real time qPCR Array studies identified PELP1<br />

modulate expression of EMT genes Snail, Twist, ZEB1, ZEB2,<br />

Vimentin and MMPs. Importantly, whole genome microRNA array<br />

analysis using PELP1 model cells revealed that miR200a and miR141<br />

were significantly upregulated in cells expressing PELP1-shRNA<br />

compared to control cells. Accordingly, over expression of PELP1<br />

in low metastatic model cells decreased expression of miR200a and<br />

miR141. PELP1 regulation of miRs was further confirmed by ZEB1<br />

and ZEB2 3’UTR luciferase reporter assays. ChIP analysis revealed<br />

recruitment of PELP1 to the proximal promoter region of miR-200a<br />

and miR141 and promoter reporter assays further confirmed PELP1<br />

regulation of miRs. Interestingly, PELP1 down regulated expression<br />

of miR200a and miR141 by promoting repressive chromatin<br />

modifications via HDAC2. Supporting this, HDAC inhibitors reversed<br />

PELP1 driven repressive effects. Further, ectopic expression of<br />

miR200a and miR141 mimetic decreased PELP1 mediated invasion/<br />

metastatic functions. Prognostic significance of PELP1-miRNA<br />

axis was determined using Tissue micro-array (TMA) and in situ<br />

hybridization (ISH assays) of Locked Nucleic Acid (LNA)-based<br />

microarray approach in 102 human breast tumors. To test therapeutic<br />

potential in vivo, we have generated ZR-PELP1- and MCF7-PELP1-<br />

shMIMIC of miR200a and miR141 stable cells. In vitro gene<br />

expression and Boyden chamber assays using these model cells<br />

revealed that shMIMIC of miR200a and miR141 reversed PELP1<br />

mediated alterations in gene expression and reduced PELP1 driven<br />

migration/invasion. Proof of principle studies using IVIS imaging<br />

of nude mice based assays of GFP-Luc labeled cells demonstrated<br />

therapeutic efficacy of miRIDIAN shMIMIC of miR200a and<br />

miR141 on PELP1 driven in vivo metastasis. Collectively, these<br />

novel findings demonstrate for the first time a previously unknown<br />

role for PELP1 in epigenetically controlling the functions of tumor<br />

metastasis suppressor miR-200a and miR141. These results suggest<br />

that PELP1-miR axis may be crucial stimulus for promoting EMT and<br />

breast cancer metastasis. This study is funded by NIH T32CA148724<br />

Postdoc Fellowship Grant.<br />

P5-04-05<br />

E-Cadherin is Required for In Vivo Growth of Inflammatory<br />

<strong>Breast</strong> <strong>Cancer</strong>: Importance of Mesenchymal-Epithelial Transition<br />

(MET) and Role of HIF1α<br />

Chu K, Boley KM, Cristofanilli M, Robertson FM. The University of<br />

Texas MD Anderson <strong>Cancer</strong> Center, Houston, TX; Fox Chase <strong>Cancer</strong><br />

Center, Philadelphia, PA<br />

Background: Inflammatory breast cancer (IBC) is the most aggressive<br />

and deadly form of breast cancer and is phenotypically distinct from<br />

other forms of locally advanced breast cancer. The 5-year survival for<br />

IBC patients (40%) has not improved even with the implementation<br />

of multi-disciplinary care and targeted therapies (e.g. anti-HER2 or<br />

anti-estrogen therapies). Lymph node involvement is common at first<br />

diagnosis and the accelerated metastasis program that characterizes<br />

IBC account for a disproportionate number of the 40,000 women<br />

who will die from breast cancer. The most common molecular<br />

marker for IBC is the cell adhesion surface glycoprotein, E-Cadherin.<br />

Classically, E-Cadherin is thought to be a tumor suppressor and its<br />

loss is associated with the epithelial-mesenchymal transition (EMT)<br />

and acquisition of an invasive phenotype. The importance of EMT in<br />

metastasis is well accepted in numerous tumor models although the<br />

presence of EMT in patient samples is rarely demonstrated. Given<br />

the propensity of IBC to metastasize, one would expect a strong EMT<br />

signature in IBC cell lines, however the persistent robust expression<br />

of E-Cadherin in IBC patient tumors and in IBC cell lines, with a<br />

lack of expression of ZEB1, a transcriptional repressor of E-cadherin<br />

and diminished expression of genes within the transforming growth<br />

factor beta signaling pathway, suggests otherwise.<br />

Methods: To further define the role of E-Cadherin in IBC, we used<br />

the SUM149 IBC cell line to manipulate E-Cadherin via shRNA<br />

knockdown and by overexpression of ZEB1 a known transcriptional<br />

repressor of E-cadherin.<br />

Results: While the lack of E-Cadherin or ZEB1 overexpression in<br />

SUM149 cells led to an EMT phenotype in vitro, E-Cadherin was<br />

required for in vivo growth of SUM149 tumor cells. Gene profiling<br />

identified novel E-Cadherin signaling pathways including an hypoxia<br />

gene signature through HIF1a as well as cytokine responsive<br />

pathways. The requirement of HIF-1a for in vivo growth of SUM149<br />

cells was confirmed by shRNA knockdown of HIF-1a.<br />

Conclusions: The absolute requirement for E-Cadherin and<br />

maintenance of the mesenchymal-epithelial transition in IBC<br />

tumor growth is demonstrated. Furthermore, a novel functional link<br />

between E-Cadherin and HIF-1a and in their critical role in survival<br />

of IBC tumors is identified for the first time and warrants further<br />

investigation.<br />

Supported in part by The Susan G Komen Organization Promise<br />

Grant KG081287 (FMR and MC)<br />

www.aacrjournals.org 435s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P5-04-06<br />

Soluble factors from activated immune cells induce epithelial<br />

mesenchymal transition in inflammatory breast cancer cells<br />

Cohen EN, Gao H, Anfossi S, Giordano A, Tin S, Wu Q, Lee B-N,<br />

Luthra R, Krishnamurthy S, Hortobagyi GN, Ueno NT, Woodward WA,<br />

Reuben JM. The University of Texas MD Anderson <strong>Cancer</strong> Center,<br />

Houston, TX; The University of Texas at Houston Health Science<br />

Center, Houston, TX; The Morgan Welch Inflammatory <strong>Breast</strong> <strong>Cancer</strong><br />

Research Program and Clinic, The University of Texas MD Anderson<br />

<strong>Cancer</strong> Center, Houston, TX<br />

Rationale:<br />

Inflammatory breast cancer (IBC) is the most insidious form of<br />

locally advanced disease. Emerging evidence suggests that host<br />

factors in the microenviromement may interact with underlying IBC<br />

genetics to promote the aggressive nature of the tumor. An integral<br />

part of the metastatic process involves epithelial to mesenchymal<br />

transition (EMT) where primary breast cancer cells gain motility and<br />

stem cell features that allow distant seeding. Interestingly, the IBC<br />

consortium microarray data found no clear evidence for EMT in IBC<br />

tumor tissues. However, it is unknown if soluble factors secreted by<br />

activated immune cells mediate EMT in the IBC microenvironment<br />

that may account for the absence of EMT in studies of the tumor cells<br />

themselves. Therefore, we tested whether the conditioned media of<br />

activated immune cells were capable of inducing EMT in IBC cells.<br />

Methods:<br />

Conditioned media (CM) were generated using healthy donor<br />

peripheral blood mononuclear cells that were activated with anti-CD3<br />

antibody immobilized to plastic and soluble anti-CD28 antibody to<br />

activate T cells through the T-cell receptor (TCR) or left unstimulated<br />

for 48 hours. Thereafter, CM from each of the cultures was harvested<br />

and filtered. Next, 48-hour pre-seeded SUM149 IBC cells were grown<br />

in culture medium consisting of 25% CM and 75% IBC culture<br />

medium for an additional 2 days. Unconditioned media and TGF-β<br />

were used as negative and positive controls, respectively for EMT.<br />

Following treatment with CM, RNA was extracted from the target<br />

cells and analyzed for the presence of EMT-related transcription<br />

factors (EMT-TF) and markers of epithelial and mesenchymal states<br />

by TaqMan® qRT-PCR. Subsequently, a panel of 24 genes was tested<br />

on 4 IBC cell lines (SUM149, SUM190, KPL4 and IBC-3) and 5<br />

non-IBC cell lines (MCF-10a, MCF-7, MDA-231, and MDA-453)<br />

treated with immune-activated CM using the Fluidigm® Dynamic<br />

Array integrated fluidic circuit (“chip”) gene expression platform<br />

which allows for the simultaneous quantification of 2,304 data points<br />

using TaqMan® assays. Formalin-fixed, paraffin embedded blocks<br />

were prepared from trypsinized cells for immunohistochemical (IHC)<br />

staining to detect E-cadherin and vimentin expression.<br />

Results:<br />

SUM149 cells cultured in the presence of TCR-activated CM for two<br />

days showed upregulation in EMT-TFs (SNAIL1, ZEB1, and TG2),<br />

vimentin and fibronectin by qRT-PCR. IHC staining showed increases<br />

in both vimentin and E-cadherin expression after 48-hour exposure to<br />

anti-TCR CM. Fluidigm® gene expression analysis of multiple cell<br />

lines exposed to anti-TCR CM showed that E-cadherin expression<br />

was unchanged or slightly decreased in non-IBC cell lines, whereas<br />

3 of 4 IBC cell lines showed an increase in E-cadherin.<br />

Discussion:<br />

These data suggest that soluble factors secreted by activated immune<br />

cells are capable of inducing EMT in IBC cells and may mediate the<br />

persistent E-cadherin expression observed in IBC. Such processes<br />

may contribute to the highly aggressive nature of the disease; however,<br />

an immune competent in vivo model is warranted to fully understand<br />

the implications of these findings.<br />

P5-04-07<br />

Epithelial-mesenchymal transition phenotype in breast cancers<br />

is associated with clinicopathologic factors indicating aggressive<br />

biologic behavior and poor clinical outcome<br />

Bae YK, Kim A, Choi JE, Kang SH, Lee SJ. Yeungnam University<br />

College of Medicine, Daegu, Korea<br />

Epithelial-mesenchymal transition (EMT) is defined by the loss<br />

of epithelial characteristics and the acquisition of a mesenchymal<br />

phenotype. EMT in epithelial tumors is a reversible process and<br />

expected to be involved in invasion, metastasis and chemoresistance<br />

of cancer cells. EMT-inducing factors down-regulate E-cadherin and<br />

up-regulate extracellular matrix proteins such as fibronectin. <strong>Cancer</strong><br />

tissue may display a wide spectrum of expression phenotypes of<br />

EMT-related proteins. However, little is known about the clinical<br />

significance of the different EMT phenotypes in breast cancers. We<br />

investigated expression pattern of EMT-related proteins, E-cadherin<br />

and fibronectin in 1,498 invasive breast carcinomas by using<br />

immunohistochemistry on tissue microarrays. EMT phenotypes were<br />

divided into complete type (E-cadherin-negative and fibronectinpositive);<br />

incomplete type, including hybrid (E-cadherin-positive and<br />

fibronectin-positive) and null (E-cadherin-negative and fibronectinnegative)<br />

types; and a wild type (E-cadherin-positive and fibronectinnegative).<br />

We correlated EMT phenotype with clinicopathologic<br />

characteristics and patients survival. Loss of E-cadherin was observed<br />

in 140 (9.3%) cases and fibronectin was expressed in cancer cells of<br />

320 (21.4%) cases. Twenty three (1.5%) cases were categorized as<br />

complete type, 414 (27.6%) as incomplete type (hybrid type, 297;<br />

null type, 117), and 1,061 (70.8%) as wild type. Complete EMT<br />

phenotype was significantly associated with advanced pT stage,<br />

lymph node metastasis, high histological grade, and triple negativity<br />

(p


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 5<br />

increased expression of some EMT transcriptional regulators, such<br />

as ZEB1, Snail and Slug, which suggested our selected kinases may<br />

regulate EMT through these transcription factors. Interestingly,<br />

FACS analysis also showed a substantial increase in CD44 hi CD24 -<br />

population in these kinase-transducted HMLER cells. The increase<br />

of CD44 hi CD24 - mesenchymal phenotype could be attributed to<br />

increase of standard form CD44 (CD44s) and decrease of the variant<br />

form CD44 (CD44v8-10). And this alternation was associated with<br />

down-regulation of ESRP1 and ESRP2, two splicing factors that<br />

regulate CD44 splicing and EMT. Therefore, these selected kinases<br />

may be considered as novel regulators of EMT, and may aid the<br />

development of new prognostic markers or drug targets for breast<br />

cancer progression.<br />

P5-04-09<br />

Squamous Cell Carcinoma Antigen 1 (SCCA1) Upregulation<br />

Causes Epithelial to Mesenchymal Transition (EMT) and<br />

Promotes Mammary Tumorigenesis<br />

Sheshadri N, Ullman E, Catanzaro J, Chen E, Zong W-X. Stony Brook<br />

University, Stony Brook, NY<br />

SCCA1, a member of the family of serine protease inhibitors, was first<br />

identified as a serological marker for cervical cancer but has since been<br />

shown to be upregulated in several other cancers including lung, head<br />

and neck, liver, skin, and breast. SCCA1 is believed to protect cells<br />

from unscheduled cathepsin release and activation upon lysosomal<br />

injury. In order to understand the molecular role of SCCA in cancer,<br />

we ectopically expressed it in the immortalized yet non-tumorigenic<br />

mammary epithelial cell line MCF10A. Expression of SCCA1 impairs<br />

the efficiency of lysosomal as well as proteasomal protein degradation<br />

pathways. Transcriptional profiling of SCCA expressing cells by<br />

microarray demonstrates an altered gene expression pattern that<br />

resembles EMT. Phenotypically, SCCA1 expression leads to spindle<br />

shaped cell morphology, increased migratory potential, resistance<br />

to anoikis, and the ability to form colonies in soft agar. SCCA1-<br />

expressing MCF10A cells can form tumors when injected into the<br />

mammary fat pad of immune-compromised mice. Correlative to the<br />

transforming activity, SCCA1 expressing cells have enhanced Erk and<br />

Akt signaling pathways, and have bypassed the requirement of growth<br />

factors and hormones that are essential for proliferation of MCF10A<br />

cells. In a transgenic mouse model, where SCCA1 is conditionally<br />

expressed in mammary epithelial cells under the control of MMTV<br />

promoter, increased mammary gland ductal branching and hyperplasia<br />

are observed. These findings indicate a previously unknown role of<br />

SCCA1 in promoting EMT and tumorigenesis, possibly by inducing<br />

chronic misfolded protein stress that contributes to enhance tumorpromoting<br />

signaling.<br />

P5-05-01<br />

Catalogued phospho-sensing peptides identifying active,<br />

oncogenic kinase signatures<br />

Mori M, Chen Z, Boudreau A, van’t Veer L, Coppé J-P. University<br />

of California, <strong>San</strong> Francisco, CA; Kinogea Inc., Shanghai, China;<br />

Lawrence Berkeley National Laboratory, Berkeley, CA<br />

Background: Phospho-signaling networks represent one of the most<br />

clinically and therapeutically important frontiers of biology and<br />

medicine. Yet, measuring the phospho-catalytic activity of kinases,<br />

and monitoring the functionality of the entire kinome network at once<br />

remains largely unexplored. In particular, a highly convenient tool<br />

would be the advent of a microarray-like chip to comprehensively map<br />

phosphorylation networks to identify kinase signatures underlying<br />

cancer types and guide therapeutic interventions. In order to create<br />

such a kinase-sensing platform, we developed a library of peptide<br />

probes capable of reporting on the catalytic activity of kinases. The<br />

well-studied EGFR and SRC kinase network measurements serve<br />

as proof of concept.<br />

Methods: We developed a human kinase protein and peptide<br />

repertoire from thousands of publications and >30 public databases<br />

computationally curated, filtered and merged, suitable to act as peptide<br />

beacons to evaluate enzymes’ activity. Our database catalogues 916<br />

kinase enzymes, 1913 substrate proteins, 6173 distinctive kinasesubstrate<br />

active nodes, and compiles 2702 unique biological kinase<br />

peptide targets that all represent biologically relevant, latent nanosensors<br />

usable to comprehensively track kinase signaling networks.<br />

Here, a representative set of 33 out of 417 peptides predicted to be<br />

specifically targeted by EGFR and SRC tyrosine kinase families<br />

and 17 control peptides, is tested for their potential kinase-activity<br />

reporting ability using a common luminescence ATP-consumption<br />

assay. We further validated the peptide screen in biological extracts<br />

using an isogenic culture model of basal like breast cancer (HMT-<br />

3522 S1 and T4-2).<br />

Results: The differential phosphorylation activity of recombinant,<br />

active kinase enzymes (EGFR, ERBB2, SRC, HCK, FYN, FRK, LYN,<br />

LCK) was successfully detected by ATP-consumption measurement<br />

using the 50 peptides in time course experiments (0.25-4 hours) with<br />

evidence of selectively. Subsets of these data were confirmed by SRC<br />

kinase inhibitor treatments and dilution assays (0.1-5ng/ul kinase<br />

enzymes). Increased presence of activated SRC kinase in biological<br />

extracts from T4-2 breast cancer cells compared to nonmalignant<br />

S1 cells was shown by significantly elevated phosphorylation of 10<br />

tested peptides (maximum elevation was shown with a SRC -specific<br />

reporting peptide and increased from 4 to 73% ATP consumption in<br />

S1 versus T4-2 cells, respectively).<br />

Conclusion: These data demonstrate that the tested peptides can<br />

monitor kinases’ activity. The peptide database can serve as a resource<br />

to provide sensitive and specific phospho-sensing probes, used to<br />

directly and specifically evaluate the activity of kinases, and can<br />

potentially support the identification of the entire, active, oncogenic<br />

kinome. Using this resource, we were able to identify peptides with<br />

evidence of specificity in in vitro and ex culture assays. Our efforts<br />

now concentrate on expanding this approach into a functional<br />

kinomic-screening platform in multiplex array format. Ultimately, we<br />

hope to translate such device into the clinic, and map phosphorylation<br />

signatures that cause breast malignancies and to guide tailor-made<br />

therapies.<br />

P5-05-02<br />

The role of cellular heterogeneity on the therapeutic response<br />

of breast cancer: Clinical insights from a hybrid multiscale<br />

computational model<br />

Powathil GG, Thompson A, Chaplain MAJ. University of Dundee,<br />

Scotland, United Kingdom; Dundee <strong>Cancer</strong> Centre, University of<br />

Dundee, Scotland, United Kingdom<br />

Background:<br />

The therapeutic control of cancer progression critically depends on<br />

the responses of the individual cells that constitute the entire tumor<br />

mass. A key reason for poor therapeutic responses is the emergence<br />

of drug resistance due to intracellular and extracellular cellular<br />

heterogeneity that seriously impair the efficacy of chemotherapy<br />

and radiation therapy.<br />

Method:<br />

We have developed a hybrid multiscale computational model of breast<br />

cancer growth and treatment that incorporates individual cell-cycle<br />

www.aacrjournals.org 437s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

dynamics and the effects of a changing microenvironment through<br />

the oxygen dynamics to consider optimum treatment protocols<br />

for breast cancer. In this model, the internal cell-cycle dynamics<br />

determine the growth strategy of the cancer cells, making the model<br />

more biologically relevant. This also helps classify the cancer cells<br />

according to their cell-cycle state and to analyse the effect of various<br />

cell-cycle-dependent chemotherapeutic drugs. The incorporation of<br />

oxygen dynamics within this hybrid model allows study of the effects<br />

of the microenvironment in cell-cycle regulation and drug resistance<br />

due to a hypoxia-induced quiescence (G0/G1 arrest) of the cancer<br />

cells. Furthermore, the effects of radiation therapy are included in<br />

the model using a modified linear quadratic model for the radiation<br />

damage, incorporating the effects of hypoxia and cell-cycle in<br />

determining the cell based radiosensitivity. The model is then used to<br />

study various combinations of multiple doses of cell-cycle-dependent<br />

chemotherapies and radiation therapy given that radiation may work<br />

better by the partial synchronisation of cells in a radiosensitive phase<br />

of the cell-cycle.<br />

Results:<br />

Using this computational model, we have shown that the cytotoxic<br />

effect of combination therapy is very much dependent on the timing of<br />

drug delivery, the time delay between the doses of chemotherapeutic<br />

drug and cell-cycle heterogeneity. The effectiveness of the drugs is<br />

also dependent on the spatial distribution of the tumor cell mass as<br />

this greatly influences the surrounding tumor microenvironment and<br />

drug distribution. After each dose of the chemotherapeutic drug, the<br />

changing spatial dynamics of the tumor cells redistributes the tissue<br />

oxygen and subsequently delivered drug doses within the tumor.<br />

Moreover, the cell-cycle phase-based chemotherapy also helps in<br />

favor of radiation therapy through the partial synchronisation of cells<br />

in the most radiosensitive phase of the cell-cycle.<br />

Conclusions:<br />

The incorporation of the spatial distribution of the breast cancer cells<br />

along with the internal and external cellular heterogeneity is critical<br />

to optimising scheduling strategies for chemotherapy and combined<br />

chemotherapy and radiation therapy. Overall, the computational<br />

simulation results highlight the potential usefulness of the model in<br />

developing patient-specific optimal treatment strategies.<br />

P5-05-03<br />

The 5p12 breast cancer susceptibility locus is associated with<br />

MRPS30 expression in estrogen receptor - positive tumors<br />

Quigley DA, Van Loo P, Alnæs GG, Tost J, Zelenika D, Balmain<br />

A, Børresen-Dale A-L, Kristensen VN. Institute for <strong>Cancer</strong><br />

Research, Oslo University Hospital, Oslo, Norway; Wellcome Trust<br />

<strong>San</strong>ger Institute, Hinxton, United Kingdom; Helen Diller Family<br />

Comprehensive <strong>Cancer</strong> Center, University of California, <strong>San</strong><br />

Francisco, CA; CEA - Institut de Génomique, Evry, France<br />

Genome-wide association studies (GWAS) have identified numerous<br />

loci linked to breast cancer susceptibility, but few of these loci have<br />

been convincingly linked to causal variants. We identified genes<br />

whose mRNA expression is linked to germline variation in normal<br />

mammary tissue and breast tumors by performing a genome-wide<br />

expression Quantitative Trait Locus (eQTL) analysis in 97 samples<br />

of normal mammary tissue and 286 breast adenocarcinomas. In the<br />

tumors we found 164 cis-acting loci, which affect expression of<br />

nearby genes. Twenty nine of these loci are previously unreported<br />

in other eQTL studies, including a cis-acting locus at 5p12 affecting<br />

expression of the mitochondrial ribosomal 28S subunit protein<br />

MRPS30 in estrogen receptor-positive but not estrogen receptornegative<br />

tumors. This locus has been associated with susceptibility<br />

to estrogen receptor-positive breast cancer in several GWAS, with<br />

MRPS30 being the suggested candidate gene. One of these studies<br />

found that the rs771660 variant was most significantly associated<br />

with breast cancer in the 5p12 region, and we found that rs771660<br />

was most strongly associated with MRPS30 gene expression. We<br />

provide the first evidence that this breast cancer susceptibility locus<br />

affects MRPS30 expression specifically in estrogen receptor-positive<br />

tumors and suggest a mechanism for estrogen activation of MRPS30<br />

via proteins in the AP-1 complex.<br />

P5-05-04<br />

Intraductal delivery of RNAi-based therapeutics to gene targets<br />

identified through computational systems modeling<br />

Brock A, Krause S, Li H, Kowalski M, Collins J, Ingber D. Wyss<br />

Institute, Harvard University, Boston, MA; Boston Children’s<br />

Hospital, Boston, MA<br />

We utilize a computational systems biology approach to identify<br />

gene targets with the potential to serve as targets for therapeutic<br />

RNAi. Inferred gene regulatory network models were constructed<br />

using whole transcriptome expression profiling for progressive<br />

stages of mammary tumorigenesis in FVB C3(1)-SV40Tag mice.<br />

These computational models predicted key mediator transcription<br />

factors that are critical for hyperplasia and ductal filling in the early<br />

stages of disease progression. Putative mediator transcription factors<br />

were targeted by RNAi first in a mammary spheroid assay. Based<br />

on their ability to restore lumen formation and normal apical-basal<br />

polarization to mammary tumor cells, two transcription factors, Hoxa1<br />

and Hoxb13, were selected for validation in vivo. siRNA targeting<br />

each of these transcription factors was formulated with lipidoid<br />

nanoparticles and delivered to the mammary gland via intraductal<br />

injection. Silencing Hoxa1 resulted in a significant reduction in tumor<br />

incidence in FVB C3(1)-SV40Tag. This represents a novel strategy<br />

to achieve localized silencing of a target gene in the mammary<br />

epithelium.<br />

P5-05-05<br />

Computational modeling of breast cancer growth and spread:<br />

the role of matrix degrading enzymes<br />

Deakin NE, Thompson AM, Chaplain MAJ. University of Dundee,<br />

United Kingdom; Dundee <strong>Cancer</strong> Centre, University of Dundee,<br />

United Kingdom<br />

Background<br />

A “hallmark” of cancer growth and metastatic spread is the process of<br />

local invasion of the surrounding tissue. <strong>Cancer</strong> cells achieve this by<br />

the secretion of enzymes involved in proteolysis (tissue degradation)<br />

including matrix metalloproteinases (MMPs). These over-expressed<br />

proteolytic enzymes then proceed to degrade the host tissue allowing<br />

the cancer cells to disseminate throughout the microenvironment by<br />

active migration and interaction with components of the extracellular<br />

matrix such as collagen. The implications of MMP-2 activation by<br />

MMP-14 and the tissue inhibitor of metalloproteinases-2 (TIMP2) can<br />

be considered alongside the suitability of the matrix to be invaded.<br />

The suitability of the matrix incorporates pore size, since in some<br />

highly dense collagen structures such as breast tissue, the cancer<br />

cells are unable to physically fit through a porous region and rely on<br />

membrane-bound MMPs to forge a path through which degradation<br />

by other MMPs and movement of cancer cells can occur.<br />

Method<br />

We have developed a multi-scale mathematical model of breast<br />

cancer cell invasion of host tissue by combining both micro- (cell)<br />

and macro- (tissue) scale dynamics. The model considers cancer cells<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

438s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 5<br />

and two matrix-degrading enzymes from the MMP family of differing<br />

mechanistic actions, namely MMP-2 and the membrane bound MMP-<br />

14 (also called MT1-MMP), and their interaction with, and effect on,<br />

the extracellular matrix (ECM) in a 2-D spatial domain.<br />

For the macro-scale we use a system of reaction-diffusion-taxis<br />

partial differential equations to capture the qualitative dynamics of<br />

the migratory response of the cancer cells. The micro-scale operates<br />

over a smaller spatial and temporal scale, and as such allows us<br />

to explicitly examine the stochastic role which is inherent within<br />

the region of enzyme operation. Computational simulations of the<br />

equations predict the spatio-temporal evolution of the cancer cell<br />

density, the concentration levels of the various enzymes and the<br />

density of the extracellular matrix.<br />

Results<br />

The model exhibits a rich range of dynamic and heterogeneous<br />

spatio-temporal solutions, which qualitatively match experimental<br />

and clinically observed results for aggressive breast carcinoma<br />

invasion and can be used to determine the speed of invasion through<br />

a variety of structured mediums such as the highly dense collagen<br />

constitution of some peritumoral stroma. Using the “worst case”<br />

scenario, an estimate for the maximum region of invasion over time<br />

from the initial data can be obtained suggesting the likely extent of<br />

breast cancer cell invasion into the surrounding tissue.<br />

Conclusion<br />

This modeling is most relevant to and informs the biological processes<br />

occurring in early stage breast cancer, in which local invasion is a<br />

crucial aspect of tumor behavior.<br />

P5-05-06<br />

Spatio-temporal computational modeling of the p53-Mdm2<br />

pathway<br />

Sturrock M, Thompson AM, Chaplain MAJ. University of Dundee,<br />

United Kingdom; Dundee <strong>Cancer</strong> Centre, University of Dundee,<br />

United Kingdom<br />

Background<br />

The p53 transcription factor is a well-established regulator of the cell<br />

cycle and key to preventing cancer. In response to a variety of cellular<br />

stresses (e.g., DNA damage or oncogene activation) p53 is activated<br />

and translocates to the nucleus where it activates transcription of target<br />

genes which can induce cell cycle arrest, senescence or apoptosis. One<br />

of the target genes for p53 is the gene for Mdm2, a negative regulator<br />

of p53 function in cells. Mdm2 represses p53 function through two<br />

main mechanisms: by promoting p53 ubiquitination and proteasomal<br />

degradation and, through direct inhibition of p53 transcriptional<br />

activity. Experimental data have revealed that in response to gamma<br />

irradiation, p53 and Mdm2 concentrations exhibit spatial and temporal<br />

oscillatory dynamics in individual MCF7 breast cancer cells. The<br />

precise function of these oscillations is still under investigation.<br />

Method<br />

Using in vitro laboratory data, we have developed a computational<br />

model to study the spatio-temporal evolution of the p53-Mdm2<br />

pathway. The model comprises a system of coupled nonlinear<br />

partial differential equations, including transport terms and reaction<br />

kinetics. The transport is assumed to include both random (diffusion)<br />

and active mechanisms with proteins convected in the cytoplasm<br />

toward the nucleus, modeling transport along microtubules. Internal<br />

structures such as ribosomes and the nuclear membrane are also<br />

explicitly modeled.<br />

Results<br />

Using computational simulations we have found ranges of values<br />

for the model parameters such that sustained oscillatory dynamics<br />

occur, consistent with available experimental measurements. To<br />

bridge the gap between in vitro and in silico experiments realistic<br />

cell geometries using imported images of cells have informed our<br />

computational domain. The effects of microtubule-disrupting drugs,<br />

proteasome inhibitor drugs, and mutations in the DNA binding site<br />

of p53, have been modeled yielding data in agreement with published<br />

experimental laboratory studies.<br />

Conclusions<br />

This mathematical computational model enables us to compare the<br />

effects of clinically relevant therapeutic approaches at the single cell<br />

level. In particular, the precise temporal and spatial distributions of<br />

p53 can be determined and the cellular consequences modeled.<br />

P5-06-01<br />

Active pak6 induces aneuploidy in breast cancer cells.<br />

Paul BA, Lu ML. University of Miami Miller School of Medicine,<br />

Miami, FL; Florida Atlantic University, Boca Raton, FL<br />

The natural history of breast cancer remains largely undefined. More<br />

than 50% of clinical specimens exhibit aneuploidy. Among these<br />

aneuploid cancers, nearly half have chromosome copy numbers<br />

approaching tetraploid. The high incidence of aneuploidy in advanced<br />

breast cancer is thought to be a byproduct of chromosome missegregation<br />

resulting from mutations in tumor suppressor genes<br />

regulating cell cycle checkpoints and corresponding to disease<br />

progression. However, recent large-scale sequencing projects have<br />

revealed that most tumors contain approximately 14-20 different<br />

mutant genes, suggesting heterogeneity exists throughout the genomes<br />

of cancer cells. This notion revives the concept that aneuploidy<br />

resulting from chromosome mis-segregation or other similar<br />

mechanisms may play an active role leading to transformation and<br />

disease progression.<br />

A novel p21-activated kinase 6 (PAK6) was recently identified to be<br />

an androgen receptor (AR) and estrogen receptor (ER) interacting<br />

protein. An estrogen-stimulated PAK6 activation was observed in<br />

BT474 breast cancer cells. This activation is mediated by estrogenstimulated<br />

activation of PKA. At the cellular level, active PAK6<br />

promotes transformation to metastatic phenotypes in MCF7 breast<br />

cancer cells as evidenced by increases in cell motility, matrigel<br />

invasion, and soft agar growth. Further characterization of MCF7<br />

cells expressing a constitutively active PAK6 mutant revealed an<br />

accumulation of cells with higher ploidy as compared to that of<br />

parental cells. Using a proteomic approach to determine the PAK6<br />

downstream target, we identified that Cdt1, a DNA pre-replicative<br />

complex subunit, is overexpressed in response to PAK6 activation.<br />

In eukaryotes, the expression and stability of Cdt1 are strictly<br />

regulated to ensure only one round of DNA replication in each cell<br />

cycle. Deregulated overexpression of Cdt1 has previously been<br />

demonstrated to induce aneuploidy and malignant transformation<br />

due to re-initiation of DNA replication within the same S-phase. The<br />

identification of Cdt1 overexpression in response to PAK6 activation<br />

provides a plausible mechanism for the observed accumulation of<br />

higher ploidy cells in MCF7 cells expressing active PAK6. Taken<br />

together these findings suggest a direct link between hormonal signals,<br />

DNA replication, and maintenance of genomic integrity.<br />

P5-06-02<br />

The Role of <strong>Breast</strong> Tumor Kinase (Brk) in Cell Cycle Control<br />

Nimnual AS, Chan E. Stony Brook University, Stony Brook, NY<br />

Brk promotes cell proliferation and has been associated with multiple<br />

mitogenic pathways. However, the role of Brk in the cell cycle is still<br />

elusive. Here, we present a mechanism of Brk-induced cell cycle<br />

www.aacrjournals.org 439s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

progression. Brk downregulates the cell cycle inhibitor p27 by altering<br />

its transcription factor FoxO3a subcellular localization. p27 is a key<br />

component of the G1/S cell cycle checkpoint and downregulation of<br />

p27 promotes S phase entry. The progression of the cell from G1 to<br />

S phase, induced by Brk, is independent of growth factors suggesting<br />

a role for Brk in malignant transformation. FoxO3a appears to be a<br />

major component in p27 down-regulation by Brk as the effect of Brk<br />

on p27 expression is significantly abrogated in cells transfected with<br />

siRNA targeting FoxO3a. The induction of FoxO3a nuclear exclusion<br />

by Brk is inhibited by LY294002 indicating that the mechanism is<br />

mediated by the PI3-kinase/Akt pathway. The mechanism underlying<br />

PI3-kinase mediated Brk function is still under investigation with<br />

evidence suggesting the involvement of the translocation of epidermal<br />

growth factor receptor. These data suggest that perturbation of p27<br />

expression induced by Brk causes S phase entrance. Deregulation of<br />

the cell cycle is a key event in neoplasia, and thus, the mechanism<br />

presented here likely contributes to breast cancer development.<br />

P5-07-01<br />

Defining the mechanism of breast cancer growth by chemokine<br />

receptor CXCR7 and Epidermal Growth Factor Receptor<br />

coupling interaction.<br />

Salazar N, Muñoz D, Singh RK, Lokeshwar BL. University of Miami<br />

Miller School of Medicine, Miami, FL; Miami VA Medical Center,<br />

Miami, FL; Dow Corning Inc., Midland, MI<br />

Introduction: <strong>Breast</strong> cancer (BrCa) ranks second in both incidence<br />

and cancer deaths for women in the USA. Recent advances have<br />

revealed a significant contribution of chemokines and their receptors<br />

in tumor growth, survival after chemotherapy, and organ-specific<br />

metastasis. CXCR7 is the latest CXC-chemokine receptor implicated<br />

in BrCa growth. It is endogenously over expressed in BrCa, but<br />

its mechanism of tumor growth enhancement is still not clearly<br />

defined. CXCR7 heterodimerizes with CXCR4 and binds to SDF-1<br />

(CXCL12) and CXCL11. No direct ligand mediated physiological<br />

action has been implicated for CXCR7 in BrCa other than facilitating<br />

CXCR4-mediated cell motility, however, studies in other cancers have<br />

implicated CXCR7 in cell proliferation, anti-apoptotic activity and<br />

cell-cell adhesion. The expression pattern of CXCR7 in BrCa tissues<br />

and its potential contribution in BrCa cell proliferation and survival<br />

were investigated in representative BrCa cell lines using knock down<br />

and knock-in experiments.<br />

Results: CXCR7 was only expressed in ER-α positive BrCa cells<br />

(e.g., MCF-7) and was not found in BrCa cells with known metastatic<br />

potential (MDA-MB231 and SKBr3). Depletion of CXCR7 in MCF-7<br />

BrCa cells by both siRNA and shRNA decreased cell proliferation<br />

and caused cell cycle arrest. Since CXCR7 is an atypical G-protein<br />

coupled receptor where ligand activation does not elicit conventional<br />

intracellular signaling, we hypothesized that CXCR7 regulates<br />

BrCa proliferation through interaction with EGFR or activating<br />

other potential targets of MAP-kinase activation. CXCR7 depletion<br />

reduced site-specific phosphorylation of EGFR at Tyrosine 1110 after<br />

EGF-stimulation. CXCR7 depletion also reduced phosphorylation of<br />

ERK-1/2, indicating a potentially direct interaction with mitogenic<br />

signaling in MCF-7 cells. We tested for direct interaction of CXCR7<br />

with EGFR by co-immunoprecipitation and immunofluorescence<br />

studies. We found EGFR and CXCR7 co-immunoprecipitate and<br />

co-localize as demonstrated by confocal microscopy. Interestingly,<br />

our results also demonstrate CXCR7 and EGFR are co-localized<br />

before EGF stimulation. Further corroboration of CXCR7-EGFR<br />

interaction was established using the proximity ligation assay (PLA)<br />

technique that allowed us to visualize CXCR7-EGFR co-localization<br />

both in cell culture and in human normal and breast cancer tissue.<br />

Conclusion: This data demonstrates an important role for CXCR7 in<br />

BrCa growth and explores the mechanism of action of direct coupling<br />

of EGFR with CXCR7.<br />

P5-07-02<br />

Analysis of a novel synergistic relationship between the FK506<br />

binding protein FKBP52 and beta catenin in androgen receptor<br />

signaling pathways<br />

Storer CL, Olivares K, Fletterick RJ, Webb P, Cox MB. The University<br />

of Texas at El Paso, El Paso, TX; The University of California, <strong>San</strong><br />

Francisco, CA; The Methodist Hospital Research Institute, Houston,<br />

TX<br />

Hormone-dependent cancers depend largely on folding and regulation<br />

by a chaperone-steroid receptor complex. The final stage of the<br />

receptor-chaperone complex consists of a receptor, a heat shock<br />

protein 90 (Hsp90) dimer, p23, and an immunophilin. Hormonedependent<br />

prostate cancer progresses due to key interactions between<br />

the androgen receptor complex and the ligand dihydrotestosterone.<br />

While current treatments block androgen receptor-ligand interactions,<br />

these therapies are no longer effective in advanced stage, hormone<br />

independent prostate cancer (HRPC), when the receptor is activated<br />

even in miniscule levels of hormone. Therefore, we are interested<br />

in targeting other members of the androgen receptor complex and<br />

signaling cascade, namely the immunophilin FK506 binding protein<br />

(FKBP52) and the Wnt cell signaling protein β-catenin. FKBP52 has<br />

the ability to potentiate activity of the androgen, progesterone, and<br />

glucocorticoid receptors and is a prostate cancer biomarker. β-catenin<br />

is a member of the cellular adhesion complex with E-cadherin in<br />

healthy cells, but in aberrant situations, β-catenin actively promotes<br />

progression and invasion in a number of cancers. Our data from<br />

FKBP52 knockout mouse embryonic fibroblast cell lines suggests<br />

that FKBP52 and β-catenin work in tandem to synergize androgen<br />

receptor (AR) transcriptional activity with the probasin promoter.<br />

When AR, FKBP52, and β-catenin are co-transfected, a synergistic<br />

up-regulation of AR signaling is observed, as assessed by luciferase<br />

reporter assays. According to pull down assays, FKBP52 and<br />

β-catenin can interact in the absence of other proteins. Luciferase<br />

assays of mutant FKBP52 proteins suggest that FKBP52’s binding<br />

to β-catenin involves the FKBP52 proline-rich loop and may act<br />

independently of Hsp90 binding. Interestingly, it appears that this<br />

synergistic effect is promoter-specific, suggesting that regulation by<br />

these proteins occurs at the transcriptional level. We have successfully<br />

abrogated the synergism of these proteins using the FKBP52-specific<br />

inhibitor MJC13, a small molecule developed for use in our laboratory.<br />

According to surface plasmon resonance studies, this molecule binds<br />

the androgen receptor at the BF-3 regulatory surface, the interaction<br />

surface for FKBP52 regulation of receptor. According to recent<br />

findings, AR, Wnt, and Human Epidermal Growth Factor Receptor<br />

2 signaling pathways are linked in molecular apocrine breast cancer<br />

progression. We therefore hypothesize that our findings could be<br />

relevant to breast cancer as well as prostate cancer.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

440s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 5<br />

P5-07-03<br />

Gallotannins and Gallic acid From Mango Fruit (Mangifera<br />

Indica L) Suppress <strong>Breast</strong> <strong>Cancer</strong> Tumor Growth by Targeting<br />

Phosphatidylinositol3-kinase (PI3K)-Akt-NF-kB Pathway and<br />

Associated microRNAs<br />

Banerjee N, Kim H, Krenek K, Talcott S, Talcott SM. Texas A&M<br />

University, College Station, TX<br />

The cytotoxic and anti-inflammatory properties of polyphenolics have<br />

been demonstrated in numerous types of cancer and the objective<br />

of this research was to understand the post-transcriptional targets<br />

involved in anti-inflammatory and pro-apoptotic pathways in the<br />

breast cancer cell line BT474, treated with a polyphenolic extract<br />

(2.5-40µg/ml) rich in gallotannins and gallic acid from mango fruit<br />

in vitro and in athymic nude mice bearing BT474 cells as xenografts.<br />

Results show that cell proliferation as well as tumor size and weight<br />

in vivo was decreased by polyphenolics 60% and 70% in vitro and<br />

vivo, respectively. NF-kB protein and mRNA expression as well as<br />

activation were decreased in a dose-dependent manner (0.6 fold).<br />

The extract also inhibited PI3K-dependent phosphorylation of AKT<br />

and expression and this was accompanied by down-regulation of<br />

miRNA 126 of BT474 (2.25 fold) and in vivo. NFκB-dependent<br />

genes involved in apoptosis-inhibition, metastasis and angiogenesis,<br />

such as survivin, VCAM-1, and VEGF were also decreased, in vitro<br />

and in vivo.<br />

A microRNA-array was also used to investigate whether microRNAs<br />

with target regions within affected genes were affected by the<br />

polyphenolic extract and results show that in addition to miR-126,<br />

miR-let-7a and miR-let-7b among others were significantly increased<br />

by the extract.<br />

In summary the anti-carcinogenic and anti-inflammatory activity of<br />

mango polyphenolics in breast cancer cells were at least in part due<br />

to targeting miR-126 -Phosphatidylinositol 3-kinase (PI3K)-Akt-NFkB<br />

which plays an important role in the proliferative/ inflammatory<br />

phenotype exhibited in this breast cancer cell line.<br />

P5-07-04<br />

Aurora kinase regulates PKC-mediated MMP-9 expression and<br />

invasion in MCF-7 breast cancer cells<br />

Kim JS, Noh EM, Lee YR, Hwang B-M, Jung SH, Youn HJ, Lee SJ.<br />

Institute for Medical Sciences Chonbuk National University Medical<br />

School, Jeonju, Republic of Korea; Chonbuk National University<br />

Medical School, Jeonju, Republic of Korea; School of Dentistry,<br />

Wonkwang University, Iksan, Republic of Korea; College of Phamacy,<br />

Ewha Womans University, Seoul, Republic of Korea<br />

Aurora kinase is a novel family of serine/threonine kinases. Elevated<br />

expression of aurora kinase A and B is observed in many tumor cells,<br />

and dysregulation of aurora kinase has been linked to tumorigenesis.<br />

Therefore, a number of studies focused in their oncogene activities<br />

as anti-tumor targets. Matrix metalloproteinase-9 (MMP-9), which<br />

degrades the extracellular matrix (ECM) and it is important process for<br />

breast cancer cell invasion. MMP-9 can be stimulated by activation of<br />

various cellular signaling pathways including protein kinase C (PKC),<br />

and mitogen-activated protein kinase (MAPKs), activator protein-1<br />

(AP-1) and nuclear factor-kappaB (NF-κB) signaling pathways.<br />

Here, we show that 12-O-tetradecanoylphorbol-13-acetate (TPA), a<br />

directly PKC activator stimulation resulted in an up-regulation and<br />

phosphorlyation of aurora kinases in MCF-7 breast cancer cells.<br />

Also, Results showed that inhibition of the aurora kinases suppressed<br />

TPA-induced MMP-9 secretion/expression and cell invasion through<br />

suppression of NF-κB, AP-1, and MAPKs in MCF-7 breast cancer<br />

cell. In conclusion, this study provides new insight into the novel role<br />

of aurora kinase for expression of MMP-9 by TPA and regulation of<br />

aurora kinase by TPA through MAPKs signaling pathway.<br />

P5-07-05<br />

Insulin-like growth factor binding protein-3 is a key component<br />

of the breast cancer cell response to DNA-damaging therapy<br />

Baxter RC, Lin MZ, Marzec KA, Martin JL. University of Sydney,<br />

Sydney, NSW, Australia<br />

Introduction. <strong>Breast</strong> cancer cells frequently develop resistance to<br />

DNA-damaging therapies, such as radiation and the cytotoxic drugs<br />

doxorubicin and etoposide, and activation of the epidermal growth<br />

factor receptor (EGFR) may play an important role in this process.<br />

However many estrogen receptor (ER)-negative breast tumors<br />

respond poorly to DNA-damaging drugs even when combined with<br />

an EGFR inhibitor. EGFR can modulate the repair of DNA double<br />

strand breaks (DSB) by non-homologous end-joining (NHEJ), by<br />

forming protein complexes that include the catalytic subunit of<br />

DNA-dependent protein kinase (DNA-PKcs). In previous studies we<br />

have shown that the growth-regulatory protein, insulin-like growth<br />

factor binding protein-3 (IGFBP-3), can translocate to the nucleus,<br />

is a substrate for DNA-PKcs, and can transactivate EGFR. The aim<br />

of this study was to delineate the role of IGFBP-3 in the response of<br />

breast cancer cells to DSB-inducing chemotherapeutic agents.<br />

Results. In the triple-negative breast cancer cell lines MDA-MB-468<br />

and Hs578T, which express IGFBP-3 highly, nuclear localization<br />

of EGFR and IGFBP-3, and of their complex as measured by<br />

coimmunoprecipitation (coIP), was enhanced by exposure to<br />

etoposide or doxorubicin, peaking 2 h after etoposide treatment.<br />

This effect was blocked by the EGFR kinase inhibitor gefitinib.<br />

Concomitantly, the co-location of EGFR-IGFBP-3 complexes with<br />

lipid rafts, visualized by proximity ligation assay (PLA) and confocal<br />

microscopy, decreased in response to drug treatment, consistent<br />

with the loss of this complex from the plasma membrane. Similar<br />

to the nuclear DNA-PKcs-EGFR complex, the nuclear DNA-PKcs-<br />

IGFBP-3 complex (seen by both coIP and PLA) was greatest 4 h<br />

after treatment. DNA-PKcs activation at serine-2056, required for<br />

the repair of DNA DSB and stimulated by etoposide treatment, was<br />

decreased by IGFBP-3 downregulation using two separate siRNAs,<br />

and this treatment also attenuated the enhanced DNA-PKcs-EGFR<br />

complexes seen in response to etoposide. Collectively these data<br />

suggest that IGFBP-3 has an obligatory role in the DNA repair<br />

response to DNA-damaging therapy through its interactions with<br />

EGFR and DNA-PKcs.<br />

Conclusion. IGFBP-3 co-translocation to the nucleus of breast<br />

cancer cells and its complex formation with DNA-PKcs and EGFR<br />

in response to DNA damage demonstrate its previously unrecognized<br />

involvement in the regulation of DNA DSB repair by NHEJ. These<br />

novel findings suggests the possibility of a therapeutic approach for<br />

sensitizing ER-negative breast cancers to chemo- or radiotherapy<br />

by targeting the DNA repair function of IGFBP-3. Supported by the<br />

Australian Research Council.<br />

www.aacrjournals.org 441s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P5-07-06<br />

Triple negative receptor status is associated with low DNA repair<br />

capacity in women with breast cancer<br />

Matta J, Ortiz C, Vargas W, Echenique M, <strong>San</strong>chez F, Ramirez<br />

E, Torres A, Ortiz J, Bolaños G, Gonzalez J, Laboy J, Barnes R,<br />

<strong>San</strong>tiago S, Romero A, Martinez R, Alvarez-Garriga C, Bayona M.<br />

Ponce School of Medicne and Health Sciences, Ponce, Puerto Rico;<br />

Auxilio Mutuo Hospital, <strong>San</strong> Juan, Puerto Rico; Ponce School of<br />

Medicine and Health Sciences and Damas Hospital, Ponce, Puerto<br />

Rico; Private Office, Ponce, Puerto Rico; Ponce School of Medicine<br />

and Health Sciences and Private Practice, Ponce, Puerto Rico; Ponce<br />

School of Medicine and Health Sciences and <strong>San</strong> Lucas Hospital,<br />

Ponce, Puerto Rico; Ponce School of Medicine and Health Sciences,<br />

Ponce, Puerto Rico; Dr. Pila Hospital, Ponce, Puerto Rico; Food and<br />

Drug Administration, Silver Spring, MD<br />

BACKGROUND: <strong>Breast</strong> cancer (BC) is a heterogeneous disease that<br />

manifests different etiologies based on its hormone-receptor status,<br />

which is used to determine BC risk, prognosis, and therapy. Although<br />

many studies have shown important associations between hormone<br />

receptor status and BC, it is unknown how or whether DNA repair<br />

capacity (DRC) correlates with, or is influenced by hormone receptor<br />

status Even though diminished DNA repair capacity is recognized as<br />

a risk factor for BC, no studies to date have evaluated the relationship<br />

of DRC and hormone receptor status. OBJECTIVE: The objective<br />

of this study was to evaluate the association between estrogen,<br />

progesterone, and HER2/neu receptors and DRC measured in<br />

lymphocytes of breast cancer patients. METHODS: Study participants<br />

were pre and post-menopausal women histopathologically confirmed<br />

BC (n =271) were recruited over the last 6 years, primarily through<br />

clinicians from the cities of Ponce, <strong>San</strong> Juan, and Yauco, and other<br />

selected cities throughout Puerto Rico. We recruited only patients<br />

who (1) were recently diagnosed histopathologically with primary<br />

breast cancer and (2) were treatment-naïve (i.e., had not received<br />

chemotherapy, blood transfusions, or radiotherapy). All participants<br />

signed informed consent forms for interviewing, drawing blood<br />

samples, and obtaining tumor material and pathology reports. DRC<br />

was measured in purified lymphocytes with a host cell reactivation<br />

assay and a luciferase reporter gene. Hormone receptor status for ER,<br />

PR and Her2neu, were determined through immunohistochemistry.<br />

DRC levels were dichotomized in ?high? and ?low? using the median<br />

as a cutoff point (2.0% DRC level). <strong>Breast</strong> cancer cases with high<br />

and low DRC were compared to receptor status and other selected<br />

variables such as age, BMI, multivitamin use, that were adjusted for<br />

potential confounding. Crude and multiple logistic regression adjusted<br />

odds ratios were used to assess the associations between low DRC<br />

and each of the receptors and selected variables after adjusting for<br />

age, overweight and obesity, age at menopause, multivitamin use.<br />

RESULTS: Pre-menopausal- Triple negative BC patients were more<br />

likely to have a low DRC (p


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 5<br />

P5-09-01<br />

Evaluation of BCL2 sequence variant in Iranian women patients<br />

with <strong>Breast</strong> cancer<br />

Motahari B, Ghaffarpour M, Javadi GH, Houshmand M. Science and<br />

Research branch, Islamic Azad University, Tehran, Islamic Republic<br />

of Iran; National Institute of Genetic Engineering and Biotechnology,<br />

Tehran, Islamic Republic of Iran; Iranian Research Organization for<br />

Science and Technology, Tehran, Islamic Republic of Iran; Special<br />

Medical Center, Tehran, Islamic Republic of Iran<br />

Introduction:<br />

Despite recent advancements in the treatment and management of<br />

cancers, <strong>Breast</strong> cancer still remains the most common malignancy and<br />

second leading cause of cancer-related deaths among women in Iran.<br />

Apoptosis and cellular proliferation play role an important role during<br />

normal mammary development and carcinogenesis of the mammary<br />

gland. In normal tissues, the homeostasis between apoptosis and cell<br />

proliferation is regulated by Bcl-2 family proteins. Tumor cells tend to<br />

interfere with the mechanism of Apoptosis by activating anti apoptotic<br />

genes such as bcl-2. BCL-2 gene, located at 18q21.3 and consists of<br />

three exons and two promoters (P1 and P2), which have different<br />

functions. The second promoter, P2, is located 1,400 bp upstream of<br />

the translation initiation site and functions as a negative regulatory<br />

element to the P1 promoter. Bcl-2 is the one of the most important<br />

anti apoptotic genes.It has been shown that the -938AA genotype s<br />

associated with higher Bcl-2 expression levels in B-CLL and poor<br />

prognosis. Bcl2 gene has been also demonstrated with breast cancer<br />

development and a single nucleotide polymorphism (SNP-938C >A)<br />

has been identified recently and can provide a susceptible factor for<br />

causing breast cancer.<br />

The main objective of this study was to evaluation of the frequency of<br />

specific genetic alterations in the BCL-2 gene (-938,-758 and 21) and<br />

the risk of breast cancer in Iranian women patients with <strong>Breast</strong> cancer.<br />

Materials and Methods:<br />

Patients; 27 paired Fresh tumor and adjacent normal samples were<br />

obtained from consecutive patients with BC who underwent surgery<br />

for mama mastroctomy from IRANIAN National Tumor Bank,<br />

National <strong>Cancer</strong> Institute, Imam Khomeini Hospital Complexes,<br />

Medical Tehran University, Tehran, Iran. Histopathological<br />

examinations were performed, and all tumors were confirmed as<br />

adenocarcinoma.<br />

Mutational analysis: Fresh tumors and their adjacent were extracted<br />

for genomic DNA using the QIA amp Mini Kit according to the<br />

manufacturer’s instructions. We searched DNA samples of Iranian<br />

patients for identification of SNP; -938C>A, - 758T insertion in the<br />

inhibitory P2 promoter and 21A/G in exon 2 of the BCL-2 gene by<br />

means of PCR sequencing.<br />

Results and conclusions:<br />

In All cases, the frequency of genetic alterations in the BCL-2<br />

gene (-938,-758 and 21) was 16 of27 (59.29%) heterozygous in-<br />

938(C, A) and 5 of 27(18.51%) SNP-938 C >A ,0 of 27(0%)and 8<br />

of 23(34.78%)heterozygous in +21(A,G)and 2 of 23(8.69%)SNP<br />

21A>G respectively. According to our findings, point mutation in<br />

-938C and 21A alleles displayed a positive relation with increasing<br />

tumorgenesis in breast cancer. Our result indicated that the regulatory<br />

BCL2 promoter polymorphism (-938C, A) and coding BCl2 gene<br />

(21A, G) may be useful as a tumor marker for increasing tumorgenesis<br />

in Iranian women breast cancer.<br />

P5-10-01<br />

MicroRNAs -181 and -135a modulate tumour infiltrating immune<br />

cell programs in distinct molecular breast cancer subtypes<br />

Paladini L, Truglia M, Arcangeli A, De Mattos Aruda L, Bianchini<br />

G, Iwamoto T, Bottai G, Pusztai L, Calin GA, Di Leo A, <strong>San</strong>tarpia<br />

L. Istituto Toscano Tumori, Prato, Italy; Istituto Toscano Tumori -<br />

Hospital of Prato, Prato, Italy; University of Florence, Italy; Vall<br />

d’Hebron Institute of Oncology, Vall d’Hebron University Hospital,<br />

Barcelona, Spain; Fondazione Centro <strong>San</strong> Raffaele del Monte Tabor,<br />

Milan, Italy; Okayama University Hospital, Okayama, Japan; The<br />

University of Texas M.D. Anderson <strong>Cancer</strong> Center, Houston<br />

Background and Aims: MicroRNAs (miRNAs) are short noncoding<br />

RNA whose expression levels have diagnostic and prognostic<br />

implications in cancer. However, the relevance of aberrantly expressed<br />

miRNAs to breast tumour-immune response has not been established.<br />

We aimed to assess the association between deregulated miRNAs<br />

and tumour immune response in breast molecular cancer subtypes.<br />

Methods: One hundred and seventy ductal infiltrating breast<br />

carcinomas (BC) (ER+/HER2- n=85, HER2+ n=40, ER-/HER2-<br />

n=45) were evaluated for the presence of cellular immune components<br />

by IHC assay. CD3, CD20 were used as markers for T lymphocytes<br />

infiltration (TIL), and B lymphocytes infiltration (BIL), respectively,<br />

whereas CD68 for the presence of tumour-associated macrophage<br />

(TAM). Total RNA (including miRNA) was extracted from all<br />

tumours and 32 highly specific miRNAs and 276 immune genes were<br />

evaluated by array and qRT-PCR assays. We derived an integrated<br />

immune miRNA-regulatory gene profile to tease out relationships<br />

of these genes as potential targets of specific miRNAs. In vitro<br />

experiments on BC cell lines were performed to confirm potential<br />

links between deregulated miRNAs and immune genes.<br />

Results: Lymphocytes infiltration (LI) was most common in HER2+<br />

and ER-/HER2- BC samples (P < .004 and < .001) although B<br />

and T cells components were differently distributed in these 2 BC<br />

groups. TAM was a frequent event in ER-/HER2- BC subtype (P<br />

< .005). By gene and integration analyses we identified important<br />

miRNA immune-modulated target genes associated with prognostic<br />

factors and with molecular BC subtypes. We focused our studies<br />

on miR-181 and miR-135a that were significantly associated with<br />

ER-/HER2- (P < .001) and HER2+ BC (P < .002), respectively.<br />

BCs with abnormal expression of miR-181 and miR-135a showed<br />

an important association with distinct TIL and TAM components<br />

and specific immune genes. Importantly, preclinical data on BC cell<br />

lines demonstrated that miR-181 and miR-135a were able to regulate<br />

important cell immune-modulators such as SEMA3B and STAT6<br />

signalling pathways, respectively. These 2 cellular pathways in turn<br />

were capable to regulate specific cascades of immune gene networks.<br />

Conclusion: We identified specific miRNAs associated with immune<br />

cellular components and immune-related tumour response in the 3<br />

main molecular BC subtypes (ER+/HER2-, HER2+ and ER-/HER2).<br />

Aberrant expression of miR-181 and miR-135a can contribute to BC<br />

tumour immune response programs by regulating important gene<br />

targets such as SEMA3B and STAT6 signalling pathways in ER-/<br />

HER2- and HER2+ tumours, respectively. Modulation of miRNAs<br />

activating tumour immune response programs may have important<br />

implications for novel therapeutic strategies in BC.<br />

www.aacrjournals.org 443s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P5-10-02<br />

High serum levels of miR-19a are associated with poor outcome<br />

in metastatic inflammatory breast cancer<br />

Anfossi S, Giordano A, Cohen EN, Gao H, Woodward W, Ueno NT,<br />

Valero V, Alvarez RH, Hortobagyi GN, Lee B-N, Cristofanilli M,<br />

Reuben JM. The University of Texas MD Anderson <strong>Cancer</strong> Center;<br />

Morgan Welch Inflammatory <strong>Breast</strong> <strong>Cancer</strong> Research Program and<br />

Clinic, The University of Texas MD Anderson <strong>Cancer</strong> Center; Fox<br />

Chase <strong>Cancer</strong> Center<br />

Background: Increased expression of miR-19a plays an important<br />

role in tumor progression by regulating angiogenesis and apoptosis.<br />

As miRs can be a valuable diagnostic and prognostic serum biomarker,<br />

we assessed if high serum levels of miR-19a were associated with the<br />

shorter overall survival (OS) of patients with metastatic inflammatory<br />

breast cancer (MIBC) and metastatic non-inflammatory breast cancer<br />

(MNIBC).<br />

Methods: We analyzed 27 MNIBC (10 HER2 normal [HER2 - ] and<br />

17 HER2 amplified [HER2 + ]), 37 MIBC (18 HER2 - and 19 HER2 + )<br />

patients, and 30 healthy donors (HDs). Patients’ sera were collected<br />

before starting a new line of treatment. MiR-19a levels were measured<br />

in patients’ sera and cell lines (MCF-7, SKBR3, MA-231, KPL-4,<br />

SUM-149) by qRT-PCR to assess differences in expression levels<br />

between IBC and non-IBC. MiR-192 and U6 snRNA were used<br />

to normalize miR-19a expression levels in serum and cell lines,<br />

respectively. Fold-changes in expression of miR-19a were calculated<br />

using the 2 -DCt method. Mann-Whitney U test, ROC curve analysis,<br />

Kaplan-Meier, and Student’s t-test were used for statistical analysis.<br />

Results: Median levels of miR-19a were higher in sera of<br />

both MNIBC and MIBC patients than in HDs (p=.001, p1.62) had a shorter<br />

median OS than MNIBC patients with HER2 - tumors (n=9) and low<br />

serum level of miR-19a ( 1.62) and HER2 + MNIBC patients with low<br />

miR-19a (


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 5<br />

P5-10-04<br />

Metformin mediated upregulation of microRNA-193 triggers<br />

apoptosis by decreasing fatty acid synthase<br />

Cochrane DR, Wahdan-Alaswad RS, Edgerton SM, Terrell KL,<br />

Spoelstra NS, Thor AD, Anderson SM, Richer JK. University of<br />

Colorado Denver School of Medicine, Aurora, CO<br />

Background: We have previously shown that the anti-diabetic drug<br />

metformin kills triple negative (TN) breast cancer cells lines via<br />

apoptosis, at lower IC50 than luminal and HER2 subtypes (presented<br />

at ENDO, manuscript in preparation). Metformin reportedly shows<br />

greater efficacy against stem cells than differentiated cells, reducing<br />

mammosphere formation in in non-adherent culture. Fatty acid<br />

synthase (FASN) is an enzyme critical for de novo fatty acid synthesis<br />

that is overexpressed in breast cancer.<br />

Hypothesis: We hypothesized that miRNAs might be involved in<br />

the ability of metformin to preferentially kill TN cell lines at a lower<br />

IC50 than luminal A lines.<br />

Methods: TN and luminal A (LA) breast cancer cell lines (MDA-<br />

468, HCC70 and MCF7, T47D, respectively) were used to study<br />

the effects of 10mM metformin action in vitro, at physiological and<br />

hyperglycemic culture conditions. Affymetrix chips Human Gene<br />

1.1 and the miRNA 2.0 were utilized to profile gene and miRNA<br />

expression at 6 and 24 hrs. Mimics and lentiviral expression vectors<br />

were utilized to manipulate miRNA expression and a luciferase<br />

reporter was used to confirm miR-193 direct targeting of the FASN<br />

3’UTR.<br />

Results: miR-193 is significantly higher in untreated LA as compared<br />

to TN cells. MiR-193 increases 2-4 fold within 6 hrs of metformin<br />

treatment in both TN, but not LA cell lines. A predicted target of miR-<br />

193, fatty acid synthase (FASN), is decreased by 8 fold following 24<br />

hrs 10mM metformin treatment of the TN breast cancer cells. miR-193<br />

directly targets FASN via a binding site in the 3’UTR, downregulating<br />

gene expression. Restoration of miR-193 to TN lines, causes a<br />

dramatic decrease in FASN protein in a dose dependent fashion.<br />

Conclusions and future directions: Metformin stimulates an increase<br />

in mature miR-193, which mediates the dramatic downregulation of<br />

FASN. This occurs coincident with apoptotic cell death. It remains to<br />

be determined if FASN is the only relevant direct target of miR-193<br />

in TN cell lines. The addition of exogenous non-targetable FASN,<br />

lacking its 3’UTR, will demonstrate whether this pathway is a primary<br />

mechanism of metformin action in TN cells. Future studies will<br />

investigate the ability of metformin to inhibit FASN in non-adherent<br />

mammosphere (stem cell-like) subpopulations, the role of fatty acid<br />

metabolism in mammosphere maintenance and anoikis resistance.<br />

Supported by Komen <strong>Breast</strong> <strong>Cancer</strong> Foundation Grants KG100575<br />

(ADT, JKR, SME, NSS) and KG090415 (JKR, DRC)<br />

P5-10-05<br />

Novel Mechanisms of Metformin Action in TN <strong>Breast</strong> <strong>Cancer</strong>:<br />

Upregulation of miRNA 141 and 192, are Associated with a<br />

Decrease in Targets GRB2 and MSN Involved in Signaling and<br />

Motility Respectively<br />

Edgerton SM, Richer JK, Fan Z, Spoelstra NS, Wahdan-Alsawad RS,<br />

Arnadottir SS, Thor AD. University of Colorado Denver School of<br />

Medicine, Aurora, CO; Aarhus University, Aarhus, Denmark<br />

BACKGROUND: We have previously shown that the anti-diabetic<br />

agent, metformin, inhibits cellular proliferation, colony formation and<br />

cell signaling, with an induction of S-phase arrest and apoptosis in<br />

triple negative (TN) breast cancer cell lines in vitro, and cell outgrowth<br />

and proliferation in vivo. These changes are associated with profound<br />

shifts in phosphorylated cell signaling intermediates, including EGFR,<br />

IGF-1R, Akt, MAPK and others (Liu, et al Cell Cycle 2009). We<br />

have also shown that Stat3 activity (via phosphorylation at serine and<br />

tyrosine sites) is critical to the effects of metformin against TN cells<br />

(Deng, et al Cell Cycle 2012). In unpublished studies, we have noted<br />

that metformin significantly inhibits cellular motility in vitro. In this<br />

study we focus on mechanisms underlying the effects of metformin<br />

in TN cancer cell signaling and motility.<br />

METHODS: TN breast cancer cell lines MDA-MB-468 and MDA-<br />

MB-231were used to study mechanisms of metformin action in vitro.<br />

Invasion chamber and motility assays were used to quantitate the<br />

effects of metformin as compared to untreated controls, with and<br />

without a variety of inhibitors at normal or supraphysiological glucose<br />

concentrations. Affymetrix chips Human Gene 1.1 and miRNA 2.0<br />

were used to identify genes and miRNA up or down regulated by<br />

metformin treatment at 6 and 24 hr at glucose concentrations of 5, 10<br />

or 17 mM. Appropriate primers and antibodies were used to validate<br />

these data at the transcript and protein levels respectively by qRT-<br />

PCR and western blot analyses. Lentiviral expression vectors and<br />

mimics were used to manipulate miRNA expression levels. Direct<br />

targeting of the GRB2 and MSN 3’UTR’s were performed using<br />

luciferase reporters.<br />

RESULTS: TargetScan and Diana miR-Path analysis of miRNA’s<br />

up-regulated at 6 hr following metformin treatmetns identified genes<br />

that were down regulated with metformin treatment such as GRB2<br />

(a predicted target of miR-141) and MSN (a predicted target of miR-<br />

192), as possible mechanisms of drug action. GRB2 regulated the<br />

phosphorylation of numerous receptor tyrosine kinase (RTK) gene<br />

pathway intermediates, including those downstream of EGFR, IGF-<br />

1R and MAPK. Knock-down of miR-141 by an antagomir showed<br />

that miR-141 regulated GRB2. Invasion chamber and motility assays<br />

confirm that the metformin associated reduction in TN cell motility<br />

and invasion capacity involved miR-193 and MSN.<br />

CONCLUSIONS: Metformin acts by diverse molecular mechanisms;<br />

some of these may be cell type specific. In TN breast cancer cells,<br />

metformin causes a rapid increase in miR-141, which targets GRB2, a<br />

gene that encodes a critical regulator upstream of many growth factors.<br />

Metformin treatment also upregulates miR-192, which represses MSN<br />

to reduce motility and invasion.<br />

Supported by Komen <strong>Breast</strong> <strong>Cancer</strong> Foundation Grant KG100575<br />

(SME, JKR, ZF, NSS, ADT)<br />

P5-10-06<br />

A functional role for miR-150 in breast cancer<br />

D’Amato NC, Gu H, Lee M, Heinz R, Spoelstra NS, Jean A, Cochrane<br />

DR, Richer JK. University of Colorado Anschutz Medical Campus,<br />

Aurora, CO<br />

Background: During mammary gland development, massive<br />

coordinated changes in protein expression govern the progression<br />

through pregnancy to lactation and involution. These dramatic<br />

changes are likely regulated in part at the translational level by<br />

changes in microRNAs (miRNAs). We profiled miRNA expression<br />

in mammary epithelial cells (MECs) isolated from mice at pregnancy<br />

day 14 (P14) or lactation day 2 (L2) and found that miR-150 is the<br />

most significantly downregulated miRNA between pregnancy and<br />

lactation. Interestingly, miR-150 was recently discovered to be<br />

decreased in mouse mammary tumors compared to normal mammary<br />

tissue in numerous transgenic models. However, a causal role for<br />

miR-150 has yet to be studied in human breast cancer and little is<br />

known about its functional role and relevant targets in the normal<br />

breast or breast cancer.<br />

www.aacrjournals.org 445s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Hypothesis: We hypothesized that miR-150 may be a tumor<br />

suppressor whose loss in breast cancer cells is an important event that<br />

allows for expression of multiple pro-tumorigenic genes.<br />

Methods: miR-150 levels in human breast cancers were evaluated in<br />

specimens of both ER+ and triple-negative breast cancer (TNBC) as<br />

compared to adjacent, non-involved normal breast epithelium by in<br />

situ hybridization. miR-150 levels were also measured by qRT-PCR<br />

in cell lines representative of multiple breast cancer subtypes. We<br />

stably restored miR-150 in TNBC cell lines by lentiviral infection<br />

and evaluated its effects on clonogenicity, growth in 3D culture,<br />

migration/invasion, and tumorigenicity. Expression of predicted<br />

miR-150 targets was assessed by immunoblot. Crossing of miR-<br />

150 fl/fl mice with BLG-Cre or MMTV/NIC transgenic mice will be<br />

utilized to determine the effects of MEC-specific loss of this miRNA<br />

on mammary tumorigenesis.<br />

Results: In clinical samples, in situ hybridization reveals that<br />

miR-150 levels are lower in both ER+ and TNBC tumor specimens<br />

compared to adjacent normal epithelium, with triple-negative tumors<br />

having the lowest expression. All breast cancer cell lines tested also<br />

have low miR-150 expression as compared to normal mammary<br />

epithelial tissue, with TNBC cell lines expressing the lowest levels.<br />

Exogenous expression of miR-150 in breast cancer cell lines caused<br />

a dramatic decrease in migration and invasion in vitro, and we are<br />

testing predicted miR-150 targets for their role in this phenotype.<br />

Experiments with transgenic models will determine if loss of miR-150<br />

in mammary epithelial cells in the MMTV/NIC mouse model results<br />

in decreased latency or increased tumor formation and metastasis, or<br />

if miR-150 loss during pregnancy results in alterations in lactation,<br />

hyperplasia, or tumor formation.<br />

Conclusions: TNBC specimens and cell lines have the lowest<br />

expression of miR-150, though decreased miR-150 expression<br />

compared to normal mammary epithelium is a common feature of<br />

breast cancers regardless of subtype. miR-150 expression dramatically<br />

inhibits breast cancer cell migration and invasion in vitro, and ongoing<br />

experiments will determine the relevant target genes.<br />

Supported by Susan G. Komen Grant KG090415 and NIH NICHD<br />

P01 PAR-10-245 (Project 3) to JKR<br />

P5-10-07<br />

Luminal A breast cancer: identification of novel circulating<br />

miRNA biomarkers<br />

McDermott AM, Miller N, Ball G, Sweeney KJ, Kerin MJ. National<br />

University of Ireland, Galway, Galway, Ireland; Nottingham Trent<br />

University, Nottingham, United Kingdom<br />

Introduction<br />

<strong>Breast</strong> cancer is a prevalent disease in need of a reliable circulating<br />

biomarker with a role in combination with mammography to facilitate<br />

early diagnosis. Mi(cro)RNAs are a group of small molecules with<br />

ideal biomarker characteristics. The knowledge that miRNAs are<br />

aberrantly expressed in cancer, and can be detected in the circulation<br />

supports their potential role in minimally invasive cancer diagnostics.<br />

Intrinsic breast cancer subtypes provide well characterized phenotypes<br />

to study the biomarker potential of miRNAs. The aim of this study<br />

was to identify systemic miRNAs differentially expressed in Luminal<br />

A (ER+PR+HER2/neu-) breast cancer, and to study their utility as<br />

oncologic biomarkers in the clinical setting.<br />

Material and Method<br />

Whole blood samples were collected prospectively from women<br />

diagnosed with Luminal A breast cancer (n=57) and healthy controls<br />

(n=57). Luminal A phenotype was confirmed by immunohistochemistry<br />

and fluorescence in situ hybridization (FISH). RNA was extracted,<br />

reverse transcribed and a Taqman Low-Density Array (TLDA,<br />

microarray) conducted on a test cohort (10 Luminal A; 10 Control).<br />

Differentially expressed miRNAs were identified using Artificial<br />

Neural Networks (ANN). Expression of specific miRNAs were<br />

validated using RQ-PCR on an independent whole blood cohort (n=47<br />

Luminal A; n=47 Control) and tissue (n=30). Results were analyzed<br />

using Q-Base and Minitab V16.0.<br />

Results<br />

The microarray performed on the test cohort identified 77 differentially<br />

expressed miRNAs. Artificial Neural Networking highlighted seven<br />

miRNAs for further analysis (Table 1).<br />

Table 1: Top 7 Ranked miRNAs<br />

Rank-Order* miRNA Median-Train-Performance** p-value***<br />

1 miR-181a 0.75 0.005<br />

2 miR-301a 0.833333 0.116<br />

3 miR-182 0.75 0.551<br />

4 miR-423-5p 0.75 0.08<br />

5 miR-93 0.708333 0.924<br />

6 miR-652 0.75 0.001<br />

*Ranked according to ANN ** Based on Test Cohort *** Based on Validation Cohort, obtained<br />

using 2-sample t-test<br />

RQ-PCR quantification of expression of these seven candidate<br />

miRNAs in the validation group confirmed the biomarker potential<br />

of two miRNAs. MiR-181a and miR-652 were significantly<br />

under-expressed in the cancer group compared to the control<br />

group (p=0.005 and p=0.001 respectively). Circulating miR-181a<br />

expression correlated with invasive tumour size (r=-0.592, p=0.008).<br />

Furthermore, a combination of these two miRNAs provided a<br />

sensitivity and specificity for detection of Luminal A breast cancer<br />

of 70% and 65%, respectively (Area Under the Curve, AUC=0.77).<br />

MiR-181a and miR-652 were also under-expressed in Luminal A<br />

tumor tissue compared to Tumor Associated Normal (TAN, p=0.019<br />

and p=0.001, respectively).<br />

Conclusion<br />

This study provides insight into the underlying molecular portrait<br />

of the Luminal A subtype by identifying 77 miRNAs with altered<br />

systemic expression levels. Two novel miRNA oncologic biomarkers<br />

are identified, which are altered in both the tumor and circulation.<br />

These miRNAs may have a role in combination with mammography<br />

to facilitate accurate subtype-specific breast tumor detection.<br />

P5-10-08<br />

Hyperactive MAPK signaling alters expression of miR-221/222<br />

and miR-30 families, which impacts multiple pathways and<br />

contributes to breast cancer aggressiveness and progression<br />

Miller P, El-Ashry D. University of Miami, FL<br />

We have previously identified a patient-derived microRNA signature<br />

indicative of hyperactive MAPK (hMAPK) signaling in breast<br />

cancer. This signature associates with reduced estrogen receptor<br />

(ER) expression, and altered protein expression of a multitude of<br />

genes associated with breast cancer aggressiveness and progression.<br />

Additionally, this signature predicts poor clinical outcome with<br />

reduced disease free survival and overall survival in all comers, and<br />

especially among patients with ER+ tumors. Many of the proteins<br />

whose expression is altered in tumors with this hMAPK-miRNA<br />

signature are predicted or validated targets of miRNAs within<br />

the signature. We hypothesize that alterations in expression of<br />

hMAPK-miRNAs directly affect post-transcriptional regulation of<br />

a subset of these proteins. Using a hMAPK cell line model of breast<br />

cancer generated from an MCF-7 background, we investigated the<br />

involvement of hMAPK-miRNAs in post-transcriptional regulation<br />

and resulting dynamics of expression of several proteins identified<br />

as being differentially expressed between hMAPK-tumors and nothMAPK-tumors.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

446s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 5<br />

Here we have investigated the hMAPK regulation of one<br />

miRNA family that is upregulated and one miRNA family that is<br />

downregulated in the hMAPK signature. Each of these families has<br />

key validated targets important in breast cancer, and many other<br />

putative gene targets. The miR-221/222 family is upregulated in the<br />

hMAPK-miRNA signature, and is a negative regulator of ER and the<br />

cell cycle regulator p27. In our breast cancer cell line model, we have<br />

previously demonstrated that miR-221/222 are upregulated and ER<br />

and p27 are downregulated in hMAPK cells vs control transfected<br />

MCF-7 cells. We now show that inhibiting MAPK signaling with<br />

pharmacological inhibitors reduces the expression of miR-221/222,<br />

and results in enhanced expression of ER and p27; we confirm that<br />

this regulation occurs at the post-transcriptional level using a dualluciferase<br />

reporter construct containing the ER 3’UTR. Additionally,<br />

stimulation of MAPK signaling dynamically increases the expression<br />

of miR-221/222 and decreases ER and p27 expression. These results<br />

indicate that hMAPK signaling directly alters expression of ER and<br />

p27 by altering miRNA expression. Members of the miR-30 family are<br />

downregulated in the hMAPK-miRNA signature, and several proteins<br />

that upregulated in hMAPK-miRNA tumors vs not-hMAPK-miRNA<br />

tumors, including SNAIL, NOTCH1, and FOXO3, are all targets<br />

of the miR-30 family. Using our cell line model, we have shown<br />

that members of the miR-30 family are downregulated by hMAPK<br />

signaling, that SNAIL is upregulated, and that abrogation of hMAPK<br />

signaling reverses these changes.<br />

It is known that SNAIL and ER exhibit inverse expression in breast<br />

cancer cells, and these data suggest that this is due to hMAPK<br />

mediated miRNA activity. Thus, hMAPK signaling may contribute<br />

to increased EMT, ER-negativity, and poor clinical outcome observed<br />

in breast tumors with this hMAPK-miRNA signature via the direct<br />

action of these hMAPK-regulated miRNAs. These results suggest<br />

novel mechanisms by which hMAPK signaling may contribute to<br />

breast cancer disease aggressiveness and progression.<br />

P5-10-09<br />

Prediction of Trastuzumab treatment response for HER2-positive<br />

breast cancer by microRNA profiling<br />

Sato F, Wang Z, Ueno T, Myomoto A, Takizawa S, Masuda N, Mikami<br />

Y, Shimizu K, Tsujimoto G, Toi M. Graduate School of Medicine, Kyoto<br />

University, Kyoto, Japan; Toray Industry, Kamakura, Kanagawa,<br />

Japan; Kyoto University Hospital, Kyoto, Japan; Graduate School<br />

of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan<br />

[Background and Aim] Although Trastuzumab has been used for<br />

HER2(+) breast cancer, the treatment response of Trastuzumab<br />

therapy depends on unknown mechanisms among individual cases.<br />

In order to avoid unnecessary adverse events and to lighten financial<br />

burden for patients, pre-treatment prediction of trastuzumab treatment<br />

response would be beneficial for patients. Thus, in this study, we<br />

develop a prediction algorithm using microRNA expression profile<br />

of breast cancer tissues. [Materials and Methods] Eighty-three breast<br />

cancer patients who underwent trastuzumab-chemo combined therapy<br />

before operations were enrolled with written informed consent.<br />

FFPE specimens of pre-treatment core needle biopsy samples were<br />

collected, and regions containing cancer and adjacent stromal cells<br />

were laser-microdissected. Total RNA samples extracted from the<br />

microdissected specimens were subjected for microRNA microarray<br />

(3D-Gene®, Toray, Japan) analysis. Among these 83 patients, 39 cases<br />

had pCR (complete response in IDC regions regardless presense of<br />

DCIS without lymphnode metastasis), and the other 44 cases did not.<br />

According to the pCR/non-pCR information, we develop a prediction<br />

model using 35 signature microRNAs by a SVM technique. Prediction<br />

accuracy assessed by Leave-one-out validation was AUROC=0.889.<br />

The 35 signature microRNAs for trastuzumab treatment response<br />

included 7 out of 8 let-7 family members and miR-125a-5p/b-5p.<br />

[Conclusion] microRNA profile could predict treatment response of<br />

trastuzumab-chemo combined therapy for HER2(+) breast cancer,<br />

and the developed prediction algorithm might be a useful tool for<br />

clinical decision making.<br />

P5-10-10<br />

The miR-34a is down-regulated in breast cancer and breast stem<br />

cells and a potential to eradicating breast cancer via a systemic<br />

delivery of a VISA –miR-34a nanoparticle system<br />

Xie X, Li L, Xie X, Wei W, Kong Y, Wu M, Yang L, Gao J, Xiao X, Tang<br />

J, Xie Z, Wang X, Liu P, Li X, Guo J. Sun Yat-Sen University <strong>Cancer</strong><br />

Center, Guangzhou, China<br />

<strong>Breast</strong> cancer is the most common disease in women around the world<br />

and the current treatment strategies are not potent enough for the<br />

patients, especially those who are the triple-negative type of molecular<br />

classifications. Therefore, novel and more effective treatments are<br />

pressingly needed. Of the current methods, target therapy, which<br />

not only retains cancer-specific expression but also limits toxicity,<br />

is a new strategy for treatment of cancers. In this study, it was to<br />

investigate miR-34a expression in breast caner specimen and its<br />

relationship with patient’s clinical status, and develop targeted miR-<br />

34a delivery system as a potential method for breast cancer therapy.<br />

miR-34a expression was investigated by qRT-PCR and related to<br />

clinicopathologic significance and found to be down-regulated in<br />

breast cancer as compared with normal adjacent tissues and involved<br />

in breast cancer stem cell through down-regulation of CD44, ZEB1,<br />

and Bmi1. We designed targeted miR-34a expression using T-VISA<br />

system (hTERT promoter driven VP16-Gal4-WPRE Integrated<br />

Systemic Amplifier) liposomed-based nanoparticles and evaluated<br />

the antitumor effect in breast cancer cells in vitro and in orthotopic<br />

animal model as well as its systemic toxicity. The T-VISA-miR-34a<br />

system robust targeted to breast cancer cells and breast cancer stem<br />

cells and prolonged duration expression of miR-34a. Furthermore, a<br />

systemic delivery of a VISA –miR-34a nanoparticle system targeted<br />

efficiently expression of miR-34a to tumors and could significantly<br />

inhibit tumor growth and prolong mouse survival in multiple living<br />

imaging xenograft and syngeneic models of orthotopic breast cancer<br />

and without toxicity in intact mice. Our study demonstrated that<br />

miR-34a expression was down-regulated in breast cancer and breast<br />

cancer stem cells through down-regulation of CD44, ZEB1, and Bmi1.<br />

The T-VISA-miR-34a nanoparticle system showed robust antitumor<br />

effects in breast cancer and could be a potential therapeutic approach<br />

to eradicating breast cancer.<br />

P5-10-11<br />

Clinical significance of microRNA expression as a prognostic<br />

factor in early N+ breast cancer (BC)<br />

Ciruelos EM, de Velasco GA, Castañeda C, Rodriguez-Peralto JL,<br />

Gamez A, Sepúlveda JM, Cortés-Funes H, Castellano DE, Fresno<br />

JA. Hospital Universitario 12 de Octubre, Madrid, Spain; Instituto<br />

de Enfermedades Neoplásicas, Lima, Peru; Hospital Universitario<br />

La Paz, Madrid, Spain<br />

Background: microRNAs (miRNAs) are small noncoding RNA<br />

molecules that post-transcriptionally regulate gene expression. In the<br />

present study, we describe miRNA expression in early node-positive<br />

BC and identify a 8-miRNA score that could contribute to prognosis<br />

assessment.<br />

www.aacrjournals.org 447s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Methods: First, microarray analysis was performed using RNA<br />

samples extracted from primary breast cancer. The expression of<br />

miRNAs was analyzed by RT-qPCR using the TaqMan Arrays<br />

from Applied Biosystems. After a suitable normalization of the 677<br />

miRNAs analyzed, 123 presented a good correlation between their<br />

levels of expression in frozen tissue and paraffin tissue. A supervised<br />

analysis was done in order to identify MIR that could predict relapse.<br />

Results: 172 patients with node-positive chemotherapy-treated early<br />

BC were included in the present study. The median age at diagnosis<br />

was (53.7) years, 128 cases (74.4%) had positive estrogen receptor,<br />

and 163 patients ( 94,7%) received adjuvant chemotherapy. After a<br />

median follow up of 8,1 years, 71p (41.2%) relapsed. Supervised<br />

analyses identified 8 microRNAs (has miR-30e, miR-30a, miR-21,<br />

miR-210, miR-93, miR-150, miR-99b, miR-572) associated with<br />

higher risk of relapse (5 year PFS 50% vs 90%, HR 3.75, 95% CI<br />

2.27 – 6.19) and with estimated overall survival (median not reached,<br />

HR 4.51, 95% CI 2.36 – 8.61). Interestingly, miRNAs defined 2<br />

prognostic groups (high and low risk) regardless of type of adjuvant<br />

chemotherapy (with or without anthracyclines) and histological<br />

subtype (luminal A, B or triple negative).<br />

DFS and OS in the global population clasiffied as 8miRNA-low and high risk<br />

8miRNA<br />

DFS OS<br />

risk (n)<br />

median (years) HR (95% CI) p median (years) HR (95% CI) p<br />

Low (103) not reached 3.7 (2.27 - 6.19)


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 5<br />

comparisons between means and ANOVA followed by post-hoc test<br />

to compare the mean response between subject factors of interest. All<br />

tests were 2-tailed; statistical significance was set at p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

that are differentially expressed between chemo-resistant and sensitive<br />

breast cancer cells. In particular, through high-throughput miRNA<br />

inhibitor library screens, we have identified miRNA inhibitors that<br />

sensitize resistant TNBC cells to paclitaxel, a drug commonly used to<br />

treat triple negative breast cancers. Interestingly, our studies revealed<br />

that specific miRNAs including miR-185 (patent pending) that<br />

sensitize resistant TNBCs to paclitaxel are expressed at significantly<br />

lower levels in relapsed metastatic TNBC patient sera compared to<br />

sera from their healthy siblings. Furthermore, using liposome- or<br />

biocompatible PLGA nanoparticle-based approaches, we show that<br />

systemic delivery of one of the sensitizer miRNAs suppresses breast<br />

cancer lung metastasis without any hepatotoxicity in preclinical mouse<br />

tumor models. Importantly, we show that one of the mechanisms<br />

by which sensitizer miRNAs regulate paclitaxel sensitivity is by<br />

targeting Stathmin, a microtubule destabilizer protein that can bind<br />

to tubulin dimers and stimulate microtubule destabilization. These<br />

findings suggest that tumor-specific miRNAs render selective cell<br />

cytotoxicity in a drug-specific manner, that these miRNAs may serve<br />

as detection markers for identifying patients who might benefit most<br />

from specific drug treatment, and that these miRNAs may represent<br />

novel therapeutic tools for the treatment of the TNBCs. Since<br />

miRNAs are endogenously expressed and can be easily manipulated<br />

using synthetic oligoribonucleotides, we believe that they represent<br />

more attractive targets than the single gene or gene product targeted<br />

by conventional cancer treatments that are typically prone to drug<br />

resistance. Since paclitaxel is used for the treatment of many cancers,<br />

the long-term implications of this study are likely not limited to breast<br />

cancer alone but may apply to other tumor types.<br />

P5-10-17<br />

Radiotherapy-induced miR expression influences the formation<br />

of local recurrence in breast cancer<br />

Fabris L, Berton S, Massarut S, D’Andrea S, Roncadin M, Perin T,<br />

Calin G, Belletti B, Baldassarre G. CRO, National <strong>Cancer</strong> Institute;<br />

MD Anderson <strong>Cancer</strong> Center<br />

In early breast cancer (EBC) patients, local disease relapse occurs<br />

at the site of the original primary tumor in 90% of cases and the<br />

addition of external beam radiotherapy (EBRT) after surgery is<br />

necessary to reduce recurrence rate. This observation suggested that<br />

surgery itself could represent a perturbing factor, possibly by altering<br />

the local microenvironment by inducing a wound healing response.<br />

Recent evidences indicate that, in selected EBC patients, 5 weeks<br />

of EBRT can be successfully replaced by a single dose of Targeted<br />

Intraoperative Radiotherapy (TARGIT). Interestingly, TARGIT acts<br />

not only affecting tumor cell survival itself, but is also able to alter<br />

the tumor microenvironment, by modifying cytokines secretion and<br />

activation of several intracellular pathways in breast cancer cells.<br />

In order to understand the significance of these modifications in<br />

breast tumor microenvironment after exposure to TARGIT we set<br />

up a mouse model of TARGIT treatment. Our results highlighted<br />

that the act of surgery alone strongly stimulates breast cancer cell<br />

growth and the addition of local irradiation completely reverses the<br />

surgery-induced breast cancer cell growth. Moreover, we evaluated<br />

the impact of TARGIT by analyzing microRNA (miR) expression<br />

both in our mouse model and in human specimens. To this aim, we<br />

collected paired specimens of peri-tumoral mammary tissues from 30<br />

early breast cancer (EBC) patients, before and after treatment with<br />

TARGIT. Microarray approach followed by qRT-PCR validation<br />

revealed the presence of specific TARGIT-regulated miRs. Among<br />

the others, miR-223 was the most highly and significantly modified.<br />

In silico analyses indicated that epidermal growth factor (EGF) is a<br />

potential candidate of mR-223 regulation. In vitro experiments, using<br />

both normal and tumor derived cell lines validated the bioinformatic<br />

prediction, demonstrating that the 3’UTR of EGF is a target of miR-<br />

223 and this binding results in the control of EGF production by<br />

the cells. Importantly, miR-223 overexpression was able to impair,<br />

at least in part, the wound-induced growth of normal and tumor<br />

derived breast-cancer cells, eventually influencing breast cancer cell<br />

proliferation and survival.<br />

Our results suggest that the effects of radiotherapy in the wounded<br />

tissue go far beyond the mere killing of residual tumor cells and is able<br />

to balance the negative effects of the wound healing process, both in<br />

human and mouse breast tissue. TARGIT-induced microenvironment<br />

modifications, in particular miR-223 up-regulation, significantly<br />

impair breast cancer response to surgery-induced inflammation<br />

eventually resulting in recurrence control. Our experimental findings<br />

strongly support the notion that the timely application of radiotherapy<br />

to the tumor bed, immediately after tumor removal, is critical to<br />

counteract the effects of surgery and wound healing process on<br />

residual breast cancer cells and normal peri-tumoral breast tissue.<br />

P5-11-01<br />

Extending breast conservation choices for women with breast<br />

cancer<br />

Egbeare DM, Chan HY. Cheltenham General Hospital, Cheltenham,<br />

Gloucestershire, United Kingdom<br />

Introduction<br />

Long term outcomes for women treated with breast conserving surgery<br />

are similar to women having mastectomy. The adoption of modern<br />

oncoplastic techniques (e.g. volume displacement) has increased the<br />

proportion of women having breast conserving surgery compared<br />

with mastectomy. The widespread use of therapeutic reduction<br />

mammoplasty (TRM) can further reduce the need for mastectomy<br />

in a modern oncoplastic breast unit. We analyse if the introduction of<br />

TRM has led to an increase in breast conservation and more choice<br />

for women treated at our unit.<br />

Methods<br />

Operative logbooks were reviewed. The two-year period April 2007<br />

- April 2009 (before widespread adoption of TRM) was compared to<br />

April 2010 - April 2012. Length of stay, re-admission rates, additional<br />

operative procedures and cancer recurrences were analysed.<br />

Results<br />

During the 2007-2009 time period, a single surgeon undertook<br />

209 primary operations for breast cancer. 64 women underwent<br />

mastectomy (30.6%). Of these, 26 (40.6%)had an immediate<br />

reconstruction with latissimus dorsi flap (+/- an implant). 4 women<br />

in this period had a TRM as their primary surgery for breast cancer<br />

(1.9%).<br />

During the 2010-2012 period, the same surgeon undertook 201 primary<br />

operations for breast cancer. 55 women underwent mastectomy<br />

(27.4%): 27 had immediate latissimus dorsi reconstruction; 13 had<br />

implant based reconstruction (overall reconstruction rate 72%). 32<br />

patients underwent TRM as their primary breast cancer operation<br />

(15.9%), all with simultaneous contralateral breast reduction.<br />

Of the immediate reconstruction group in 2007-09, 11 women (42%)<br />

have undergone another operative procedure – nipple reconstruction,<br />

contralateral breast operations, capsulectomy or “tidying-up”<br />

procedures. One patient has had a recurrence of her breast cancer and<br />

one patient has died of breast cancer in the 5 year follow-up period.<br />

One patient has developed contralateral breast cancer.<br />

In the 2010-12 TRM group, 6 patients (18.8%) underwent a second<br />

operation – 3 re-excisions of positive margins and 3 axillary<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

450s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 5<br />

clearances (positive sentinel lymph node biopsy at first operation).<br />

The median length of stay for women treated with TRM was 1 day<br />

compared to a median stay of 6 days for patients undergoing latissimus<br />

dorsi reconstruction.<br />

Conclusions<br />

Our unit already had high breast conserving surgery rates and a<br />

high reconstruction:mastectomy ratio. Introduction of TRM became<br />

widespread approximately 2 years ago. Using a TRM technique 25-<br />

50% of the breast volume can be removed. This enables some patients<br />

who would have been advised mastectomy to be offered a choice<br />

of breast conserving surgery; hence the further modest reduction in<br />

mastectomy rate seen.<br />

TRM enables breast (and nipple) conservation and a quicker recovery<br />

period. TRM may appeal to women thinking about breast reduction<br />

before a breast cancer diagnosis. It is useful after neoadjuvant<br />

treatment, for multifocal tumours and in women where a high tumour<br />

volume:breast volume ratio would make good cosmetic results<br />

difficult to achieve with traditional breast conservation techniques.<br />

A further advantage of TRM is the simultaneous contralateral breast<br />

reduction; thus reducing re-operation rates.<br />

In conclusion, most importantly, TRM extends breast conserving<br />

surgery choices for women with breast cancer.<br />

P5-11-02<br />

<strong>Breast</strong> <strong>Cancer</strong> Radiation Therapy and the Risk of Developing<br />

Bronchiolitis Obliterans Organizing Pneumonia (BOOP):<br />

Communication of BOOP Risk by <strong>Breast</strong> <strong>Cancer</strong> Information<br />

Websites and General Medical Information Websites<br />

Kelly EM. Kelly Consulting, Smithtown, NY<br />

Background: BOOP is an inflammatory pulmonary disorder<br />

consisting of organized polypoid granulation tissue in the distal<br />

airways extending into the alveolar ducts and alveoli. Clinical<br />

manifestations of BOOP include fever, cough, dyspnea on exertion,<br />

and fatigue. Patients with severe BOOP require hospitalization. The<br />

mainstay of BOOP treatment is high dose corticosteroids. Many<br />

patients relapse and require long term corticosteroids, often in<br />

combination with another immunosuppressant. Significant morbidity<br />

and disability may be associated with both the diagnosis of BOOP<br />

and the toxicities of treatment. The mortality rate of BOOP has been<br />

estimated to be about 5%.<br />

Radiation Therapy (RT) is an important component in the treatment<br />

of breast cancer (<strong>Breast</strong> Ca). Beginning in 1995, reports of BOOP<br />

occurring in <strong>Breast</strong> Ca RT patients began to appear in the medical<br />

literature. From 1995 to the present, 43 case study reports described<br />

121 <strong>Breast</strong> Ca RT patients who developed BOOP within one year of<br />

their RT. From 1999 to 2011, seven epidemiological studies were<br />

published that suggested that the incidence of BOOP in <strong>Breast</strong> Ca<br />

RT patients was in the range of 2% to 3%.<br />

Objectives: The primary objective of this study was to determine<br />

if internet sources of <strong>Breast</strong> Ca information targeted to <strong>Breast</strong> Ca<br />

patients include BOOP information in their description of RT risks.<br />

The secondary objective was to determine if general medical websites<br />

conveyed BOOP/<strong>Breast</strong> Ca RT risk information.<br />

Methods: Eight websites specifically targeted to <strong>Breast</strong> Ca patients<br />

were reviewed. Sponsors of these websites included the US<br />

government, <strong>Breast</strong> Ca advocacy groups, and medical organizations.<br />

Seven general medical websites that contained BOOP/<strong>Breast</strong> Ca RT<br />

information were also reviewed.<br />

Results: There was no mention of BOOP in any of the websites<br />

targeted to <strong>Breast</strong> Ca patients. The websites included those from<br />

the National <strong>Cancer</strong> Institute, American <strong>Cancer</strong> Society, American<br />

Society of Therapeutic Radiology and Oncology, Susan G. Komen,<br />

and <strong>Breast</strong><strong>Cancer</strong>.Org. The internet search identified seven general<br />

medical websites that did include information about the risk of BOOP<br />

from <strong>Breast</strong> Ca RT.<br />

Discussion: It is perplexing that none of the eight website sources<br />

for <strong>Breast</strong> Ca patients included any mention of the risk of BOOP<br />

associated with <strong>Breast</strong> Ca RT, whereas seven general medical internet<br />

sources did disclose this information. This disparity raises important<br />

issues and questions. The majority of BOOP/<strong>Breast</strong> Ca RT reports<br />

were published in pulmonology journals. Is this information being<br />

disseminated to providers of <strong>Breast</strong> Ca treatment? Many of the <strong>Breast</strong><br />

Ca information websites stated that their information was up to date<br />

and was reviewed by leading <strong>Breast</strong> Ca physicians, so it remains an<br />

enigma that the BOOP/RT association was not disclosed. The lack of<br />

any information about BOOP and <strong>Breast</strong> Ca RT may indicate <strong>Breast</strong><br />

Ca physician unawareness. An alternative explanation for this finding<br />

may be that these physicans are aware of the BOOP/RT association,<br />

but they believe that the rarity of BOOP diagnoses justifies omitting<br />

this information in internet-based information sources.<br />

P5-12-01<br />

ARE WEB-BASED RESOURCES THE BREAST? : AN<br />

EVALUATION OF THE QUALITY OF ONLINE RESOURCES<br />

FOR BREAST CANCER PATIENTS<br />

Nguyen S, Regehr G, Brar B, Lin J, Ingledew P. British Columbia<br />

<strong>Cancer</strong> Centre, Fraser Valley <strong>Cancer</strong> Centre, Surrey, BC, Canada;<br />

Centre for Health Education Scholarship, Vancouver, BC, Canada;<br />

UBC, Vancouver, BC, Canada<br />

Background: <strong>Cancer</strong> patients are increasingly using the internet to<br />

seek disease specific information. Our prior studies indicate that a<br />

majority of breast cancer patients use the internet to aid decision<br />

making and to inform themselves about their disease. Although<br />

there is a substantial amount of information available on the internet<br />

regarding breast cancer, there are no comprehensive studies evaluating<br />

the quality of this information and whether it is adequate to support<br />

patients’ decision making. The purpose of this study was to apply a<br />

validated evaluation tool to evaluate the quality of online breast cancer<br />

patient information. Methods: Using the principles of design-based<br />

research, an evaluation tool was developed and validated for the<br />

purposes of evaluation of web-based patient information. To review<br />

the quality of breast cancer information, a list of the top 100 breast<br />

cancer websites was systematically compiled using the meta-search<br />

engines Yippy and Dogpile, and the search engine Google. The<br />

websites were assessed for administration, accountability, authorship,<br />

organization, readability, and content. Inter-rater reliability was<br />

evaluated. Results were analyzed using descriptive statistics. Results:<br />

Over 2000 hits were initially obtained using the search term “breast<br />

cancer” (the most common search term according to our previous<br />

studies) in the three search engines. After applying the pre-specified<br />

inclusion and exclusion criteria the “top 100” breast cancer sites were<br />

evaluated. The majority of breast cancer websites were administered<br />

by commercial businesses (47%) and non-profit organizations<br />

(33%). 86% of the websites disclosed ownership, sponsorship, and/<br />

or advertising. Only 34% of websites identified the author and 39%<br />

cited sources. The average readability of websites was a grade 9 level.<br />

The majority of the information was out of date and only 27% of<br />

websites had updated their content within the last two years. While<br />

88% of the websites sufficiently covered breast cancer, only 18%<br />

addressed prognosis. Conclusions: While a majority of breast cancer<br />

patients use the internet to obtain information on their diagnosis and<br />

treatment options, this study demonstrates that web-based information<br />

www.aacrjournals.org 451s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

is variable and there are distressing gaps in the information available.<br />

Although the assessed websites were mostly accurate, there were<br />

significant deficits in authorship, attribution, and currency. Of<br />

concern, our research has previously demonstrated that patients use<br />

authorship and attribution to determine the reliability of web-based<br />

information. Additionally, very few websites provided information<br />

regarding prognosis, an area research has identified as an important<br />

topic sought by breast cancer patients. The results of this study can<br />

be used to counsel patients on the strengths and weaknesses of webbased<br />

breast cancer information and to empower patients to choose<br />

sites likely to enhance their personal knowledge.<br />

P5-12-02<br />

Tangled in the <strong>Breast</strong> <strong>Cancer</strong> Web: An Evaluation of the Usage<br />

of Web-based Information Resources by <strong>Breast</strong> <strong>Cancer</strong> Patients<br />

Nguyen SKA, Ingledew P. British Columbia <strong>Cancer</strong> Agency, FVCC,<br />

Surrey, BC, Canada<br />

Background: Increasingly cancer patients are researching web-based<br />

information to inform their clinical encounter with physicians. The use<br />

of the internet by breast cancer patients is incompletely characterized<br />

in the literature. The purpose of this study was to characterize the<br />

use of the internet by breast cancer patients describing the way they<br />

search for, and the impact of, web-based information on the clinical<br />

encounter.<br />

Methods: From September 2011 to January 2012, breast cancer<br />

patients presenting at a tertiary cancer centre waiting to see their<br />

oncologist completed a written 23-question survey. Answers were<br />

closed and open-ended. Mixed-methods were used to interpret the<br />

quantitative and qualitative data.<br />

Results: 81 patients were approached and 56 completed the survey.<br />

Forty-five (80%) respondents used the internet and of those, 32 (71%)<br />

respondents searched for breast cancer information. Patients who<br />

searched for internet breast cancer information (users) were younger<br />

than those who did not use the internet (p = 0.01). All respondents<br />

used Google as their principal search engine and “breast cancer” was<br />

the most commonly used search term. Twenty (62%) users accessed<br />

sites from a reputable source and 19 (59%) users viewed top search<br />

engine hits. To evaluate quality, 15 (47%) users referred to website<br />

author credentials and 13 (41%) users examined references. All<br />

users sought information with respect to treatment and three-quarters<br />

sought information on prognosis. The internet influenced treatment<br />

decision making for 30 (66%) users. Twenty-six (80%) users felt<br />

the information increased their knowledge of breast cancer and it<br />

influenced treatment decision making for 16 (53%) patients.<br />

Conclusions: This study highlights search patterns and factors<br />

used by patients in seeking web-based breast cancer information.<br />

Physicians must appreciate that breast cancer patients use the internet<br />

for information and address discrepancies between the information<br />

sought and that which is available.<br />

P5-12-03<br />

Developing the ePromotora: Increasing Promotora’s Access to<br />

<strong>Breast</strong> <strong>Cancer</strong> Electronic Resources<br />

Lopez AM, Ryan J, Valencia A, El-Khayat Y, Nunez A. University of<br />

Arizona, Tucson, AZ<br />

Background: Disparities in breast cancer screening and survival have<br />

been attributed to multiple factors including limited access to breast<br />

health education. Innovative information technologies facilitate health<br />

education; however, access to information technology is not uniform.<br />

The resultant systematic gap in health knowledge has led to a new<br />

health disparity where higher socioeconomic individuals acquire<br />

health information electronically more easily than the disadvantaged.<br />

Promotoras or Community Health Workers (CHWs) facilitate access<br />

to culturally appropriate cancer health information, screening and care.<br />

Hypothesis: An electronic asynchronous breast cancer educational<br />

intervention developed by the Arizona Telemedicine Program and<br />

the Arizona Health Sciences Library will improve CHW computer<br />

literacy and will enhance health education communication. Specific<br />

Aims: to develop, implement and assess a 6-session e-promotora<br />

training curriculum with a focus on breast health.<br />

Methods: The first step in developing a training curriculum was a<br />

needs assessment and a computer literacy assessment. This assessment<br />

served as the foundation for the computer literacy curriculum which<br />

focused on breast health content. The educational intervention was<br />

delivered as a combination of on-line, webinar, and face-to-face<br />

seminars. To meet the needs of the participants all sessions were<br />

delivered bilingually, Spanish and English. Program evaluation<br />

included pre and post knowledge assessments, session assignments,<br />

program satisfaction and 6-month follow up focus groups or structured<br />

interviews to evaluate the utility, implementation and dissemination<br />

of knowledge gained.<br />

Results: 26 CHWs have completed the training. All participants<br />

identified as Latinas, average age--51 years of age and educational<br />

attainment--58% (15/26) reported high school or less. Pre and post<br />

assessment data reveal high program satisfaction scores, increased<br />

participant knowledge and confidence in knowledge, consistent and<br />

ongoing use of information learned and computer skills gained and<br />

effective dissemination of health information gained.<br />

Conclusions: The e-promotora training curriculum effectively<br />

improves CHW computer literacy, facilitates dissemination of<br />

electronic health information and helps address the disparities in<br />

electronic health information.<br />

P5-13-01<br />

Does empowering patients improve accrual to breast cancer<br />

trials?<br />

Arnaout A, Kuchuk I, Bouganim N, Verma S, Clemons M. Ottawa<br />

Hospital, Ottawa, ON, Canada; Mcgill University and Segal <strong>Cancer</strong><br />

Centre, Jewish General Hospital, Montreal, QC, Canada<br />

BACKGROUND Many international oncology professional and<br />

patient advocacy groups recommend that a minimum of 5% of eligible<br />

new patients should be entered into a clinical trial. Unfortunately,<br />

physician and patient related barriers translates to a much lower<br />

accrual rate in reality. We performed a prospective single arm pilot<br />

study evaluating the efficacy of implementing various physician and<br />

patient related strategies in enhancing clinical trial accrual.<br />

METHODS All patients with newly diagnosed breast cancer seen<br />

at the breast surgery clinic were eligible for the study. Patients were<br />

offered information packages by their surgeons on non-surgical<br />

clinical trials that they might be eligible for, prior to their initial<br />

oncologist visit. Oncologists were informed of the information given<br />

to the prospective patient consultation via email and chart flagging.<br />

Patient were then given a questionnaire assessing the feedback on this<br />

method of introducing clinical trials. The primary outcome was the<br />

number of patients consenting to clinical trials. Secondary outcomes<br />

included the number of patients actually enrolling in clinical trials,<br />

screen failure rate, and overall patient satisfaction with this method<br />

of potentially enhancing accrual to clinical trials.<br />

RESULTS 36 patients consented to this pilot study. 51% of<br />

oncologists mentioned the clinical trials to the patients. For those<br />

patients with which clinical trials were discussed, 72% went on to<br />

consent for a trial of which 31% were ultimately found to be ineligible<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

452s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 5<br />

(screen failure rate). The overall 14% clinical trial enrolment was<br />

significantly higher than our historical average of 7%. 100% of the<br />

patients found that the information package very helpful and was<br />

noted to reduce anxiety (39%) and empower (31%) the patients. 19%<br />

of the patients felt that this information should have been offered by<br />

the oncologist during the initial consultation as opposed to the surgeon<br />

prior to the oncology visit.<br />

CONCLUSIONS The findings of this study could have a major<br />

impact on the way that cancer centres across the world approach<br />

patients for clinical trial options. Physicians remain an important<br />

barrier to trial accrual. The results of this study demonstrate that<br />

combined patient and physician centered approach to clinical trial<br />

enrolment may be the most effective.<br />

P5-13-02<br />

Decision making from multidisciplinary team meetings to<br />

bedside: factors predicting for physicians’ and breast cancer<br />

patients’ acceptance of clinical trials proposed by MTMs<br />

Deneuve J, Mazouni C, Arnedos M, Prenois F, Saghatchian M, André<br />

F, Bourrgier C, Delaloge S. Institut Gustave Roussy, Villejuif, France<br />

Background: In University hospitals or Comprehensive <strong>Cancer</strong><br />

Centers (CCC), breast cancer (BC) patients have a theoretically large<br />

access to clinical trials as part of the management and treatment of<br />

their localized or advanced disease. Though, most patients are not<br />

proposed inclusion into suitable trials or studies or do not accept<br />

them. We aimed at determining the factors that influence physicians<br />

and patient’s choice to respectively propose or enter clinical trials for<br />

which the patients were considered eligible by MTM.<br />

Methods: Patients who were considered eligible for a clinical trial<br />

by a BC-specific multidisciplinary team meeting (MTM) assessing<br />

their cases were prospectively registered during a 6 months period.<br />

The complete files of these patients were prospectively registered<br />

following initial written proposal by the MTM. The 27 clinical trials<br />

proposed were classified in 5 categories: cognitive studies, imaging,<br />

radiation therapy, diagnostic/prognostic biology, or interventional<br />

therapeutic studies. Detailed analysis of factors predicting physician’s<br />

proposal and patient’s final inclusion was conducted. Candidate<br />

factors were age, socio-demographic characteristics, PS, type of<br />

clinician, time intervals, stage of disease, type of disease, type of<br />

study, single vs multiple study proposal.<br />

Results: MTM proposed 547 inclusions into clinical trials for<br />

397 patients between March 3rd and Sept 21 st , 2011. 267 pts were<br />

considered eligible for 1 trial, 100 for 2, 25 for 3, 4 for 4 and 1 for<br />

5 trials; 211 for cognitive studies, 13 for imaging, 71 for radiation<br />

therapy, 136 for diagnostic/prognostic biology, 116 for interventional<br />

therapeutic studies. Upon referral physician’s visit, the trial was<br />

only proposed in 39.3% and pts were finally included in 29.1% of<br />

the cases. Reasons for non inclusion were patients’ refusal in only<br />

5-11%, but absence of proposal by physician in 45-81%, according to<br />

the types of studies. The only factor predicting both proposal by the<br />

referral physician’s and final inclusion into trials was type of study<br />

(both p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Results: We are planning to enroll 480 participants for the quantitative<br />

survey portion and 48 participants for in-depth qualitative data<br />

collection, including audio-recorded patient-physician interactions<br />

and in-depth patient interviews, along with 8 in-depth provider<br />

interviews. Data collection is ongoing and results will be presented.<br />

Acknowledgement: This research was supported by a grant from the<br />

National <strong>Cancer</strong> Institute (2P50CA106743)<br />

P5-14-01<br />

Withdrawn by Author<br />

P5-14-02<br />

Women’s responses to changes in US Preventive Services Task<br />

Force mammography screening guidelines: results from focus<br />

groups among ethnically diverse women<br />

Bluethmann SM, Allen JD, Hernandez C, Opdyke KM, Gates-Ferris K,<br />

Hurlbert M, Harden E. University of Texas School of Public Health;<br />

Dana Farber <strong>Cancer</strong> Institute; Avon Foundation for <strong>Breast</strong> <strong>Cancer</strong><br />

Crusade; Cicatelli and Associates<br />

Introduction: In 2009, the US Preventive Services Task Force<br />

(USPSTF) changed its recommendations on the age and frequency for<br />

routine mammography. To this point, responses to this change among<br />

ethnically diverse women have been not been well examined. The<br />

objective of this qualitative study is to describe women’s awareness<br />

of the change in mammography screening guidelines by the U.S.<br />

Preventive Services Task Force and to describe their attitudes toward<br />

this change.<br />

Methods: White, Black and Hispanic women ages 40-49 years<br />

were recruited from a variety of community settings in the Greater<br />

Boston, MA area to participate in focus groups (k=10; N=73).<br />

Groups were segmented by race/ethnicity (Black=29%; White=29%;<br />

Hispanic=15%). Women were asked if they were aware of the change<br />

in USPSTF guidelines, and if so, their understanding about reasons<br />

for this change and intention to comply. Focus groups were audiotaped<br />

and transcribed verbatim. Thematic content analysis was used<br />

to cull recurring discussion themes.<br />

Results: Most women in this study were not aware of changes in the<br />

USPSTF mammography screening guidelines. Those who were aware<br />

of the guideline change were highly suspicious that it was motivated<br />

by a desire for cost savings on the part of insurance companies and/or<br />

providers. Concerns regarding the accuracy of mammography, pain<br />

associated with screening, and fear of receiving positive test results<br />

were prevalent. Nevertheless, most said that they did not intend<br />

to comply with changes in guidelines; many believed that regular<br />

(yearly) mammography should start at a young age (40 or before)<br />

and continue indefinitely.<br />

Conclusion: Most women in this sample were unaware about changes<br />

in mammography screening guidelines and lacked understanding<br />

regarding underlying reasons for the change. Communication about<br />

the rationale for changes in mammography screening guidelines has<br />

left many women unconvinced about the potential downsides of<br />

screening and has generated a high degree of mistrust of insurance<br />

companies and medical providers.<br />

P5-14-03<br />

<strong>Breast</strong> cancer screening and follow-up of abnormal mammogram<br />

results: A population-based study comparing results from an<br />

urban university cancer center to a national database.<br />

Mitchell EP, Avery TP, Jaslow RC, Berger AC, Lee SY, Vaughan-Briggs<br />

C, Bonat J. Thomas Jefferson University, Philadelphia, PA<br />

Background: To improve access to screening, the National <strong>Breast</strong><br />

and Cervical <strong>Cancer</strong> Early Detection Program (NBCCEDP) was<br />

developed through the Centers for Disease Control and Prevention<br />

(CDC) to provide low-income, and uninsured underserved women<br />

access to timely breast and cervical cancer screening and diagnostic<br />

services. This population-based study investigates the demographics<br />

and diagnostic outcomes of women who underwent breast cancer<br />

screening through this program established at an urban university<br />

cancer center compared to data obtained from the national program<br />

database.<br />

Methods: The Kimmel <strong>Cancer</strong> Center at Jefferson (KCC) launched<br />

its screening program with resources from the CDC, the State of<br />

Pennsylvania, and the Susan G. Komen Foundation in 2008. The<br />

core of the program is staff lead by a Social Worker/Navigator who<br />

connects patients to education and screening services, institutional<br />

information and guidance, and follow-up to minimize barriers<br />

to access, lessen dropout, and ensure follow-through for timely<br />

diagnosis and treatment. Working with our Community-Based partner<br />

organizations, uninsured and underinsured women in Philadelphia<br />

are able to seamlessly travel from education and screening through<br />

to treatment and support. All KCC patients evaluated through this<br />

program from 2008-2011 were included and records of the NBCCDP<br />

database 2006-2010 for this study and analyses.<br />

Results: KCC has a substantially larger African American (54.44%<br />

vs. 13.8%), smaller Hispanic (8.59% vs. 27.6%), larger percentage<br />

of abnormal mammograms (25.96% vs. 14%), higher breast cancer<br />

diagnosed per mammogram (2.13 vs 1.0) and a much younger<br />

population than the national cohort. Fewer than 1% of KCC patients<br />

have been lost to follow-up.<br />

Conclusions: The KCC <strong>Breast</strong> and Cervical Screening and Treatment<br />

Program Services reaches a highly vulnerable and at-risk population,<br />

has a higher abnormal mammogram and breast cancer detection rate,<br />

and a higher continued participation rate than the national cohort. The<br />

Social/Worker/Navigator has a distinct role in providing follow-up<br />

for abnormal findings to minimize no-shows by providing creative<br />

problem-solving, support, counseling, finding resources to minimize<br />

barriers, and contributes significantly to the ease of operation and the<br />

continued participation of patients in the program.<br />

P5-14-04<br />

Alleviate the pain in the system: Using process improvement and<br />

systems design tools to strive for a high quality breast healthcare<br />

system in the District of Columbia metro area.<br />

Brousseau MK, Graham L, Kaye P, Nolan T, Moraras N. Primary<br />

Care Coalition of Montgomery County, Maryland, Silver Spring,<br />

MD; Regional Primary Care Coalition, Washington, DC; Associates<br />

in Process Improvement, Silver Spring, MD<br />

Background: Throughout the District of Columbia Metro Area,<br />

breast cancer mortality rates are higher than the national average due<br />

in part to limited service coordination and lack of access to breast<br />

health services along the continuum. The Primary Care Coalition of<br />

Montgomery County (PCC) and the Regional Primary Care Coalition<br />

(RPCC) are collaborating to improve the efficiency and effectiveness<br />

of breast cancer screening, referral, and follow-up so that jurisdictions<br />

and clinics in the DC metro area are better positioned to provide<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

454s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 5<br />

100% low-income women residents with access to breast healthcare.<br />

The Regional Initiative builds upon the successes of the PCC <strong>Breast</strong><br />

Healthcare Process Improvement Project.<br />

Objectives: 1) Adopt the Primary Care-Based Model in three clinics<br />

in three other jurisdictions and implement process improvement<br />

approaches to show improvements in breast cancer screening,<br />

referral, and follow-up; and 2) Develop a cross-jurisdiction learning<br />

community to enhance spread throughout the Region.<br />

Methods: The Regional Initiative employs a multi-tiered learning<br />

community framework to share data, address systems level<br />

barriers, and design new systems for the provision of high-quality<br />

care regardless of insurance status. Partners use the Model for<br />

Improvement to improve processes at the micro-system level and<br />

system design theory drives larger systems changes.<br />

Results:<br />

Objective 1: The three adaptation sites represent Prince Georges<br />

County, Maryland, Washington, DC, and Fairfax Virginia. After a<br />

year of focused process improvement, partnership development, and<br />

data collection, initial analysis demonstrates:<br />

- All three clinics show increased screening rates approaching 65%,<br />

representing the 90th percentile performance in the Healthcare<br />

Effectiveness Data and Information Set (HEDIS) for Medicaid breast<br />

cancer screening.<br />

- At Greater Baden Medical Services in Prince Georges County,<br />

Maryland, cycle time between referral and mammography screening<br />

decreased from 48 to 27 days.<br />

- The Community Health Care Network, Fairfax County, Virginia,<br />

worked closely with one mammography provider to decrease the<br />

no-show rate for mammography appointments from 26% to 6%.<br />

- The Project Team developed a change package to share<br />

recommended interventions and successful strategies that have been<br />

tested and documented, and can be used by clinics certified as primary<br />

care medical homes or in the process of adopting those standards.<br />

Objective 2: The Cross-Jurisdiction Learning Collaborative brings<br />

representatives from each jurisdiction together to share breast health<br />

measures, success stories, and plan/discuss process improvement<br />

activities from a regional perspective. Participants include over 50<br />

breast health providers from around the region:<br />

- 17 safety-net clinics<br />

- 11 hospital and/or mammography providers<br />

- All County and/or State National <strong>Breast</strong> and Cervical <strong>Cancer</strong> Early<br />

Detection Programs in the Region<br />

Next Steps: Spread learnings regionally and nationally to foster<br />

collaboration, replicate the PCC Primary Care-Based Model using<br />

the change package, and further improve breast health outcomes for<br />

the safety-net population.<br />

P5-14-05<br />

Compliance with radiation therapy in breast cancer patients in<br />

Southeastern Kentucky<br />

Elsoueidi R, Adane E, Dignan M. Appalachian Regional Healthcare,<br />

Hazard, KY; University of Kentucky, Lexington, KY<br />

Background: Nearly a quarter of Americans live in rural areas,<br />

which consistently report higher cancer mortality rates than urban<br />

and suburban areas. The worse outcome for cancer patients in<br />

these areas has been attributed in part to compliance with treatment<br />

recommendations. Southeastern Kentucky is a rural area with a low<br />

income population and barriers such as distance and lack of public<br />

transportation to access health care and compliance with treatment<br />

is a major concern.<br />

Methods: Data on patients with breast cancer treated with radiation<br />

therapy at Appalachian Regional Healthcare cancer center which<br />

serves the area of Southeastern Kentucky were collected. Age,<br />

economic status (measured by availability of medical insurance),<br />

distance of patient’s residence from the hospital, were evaluated as<br />

potential predictors of compliance with recommendations to obtain<br />

radiation therapy. Data were analyzed with Fisher’s exact test.<br />

Results: A total of 80 patients with breast cancer received adjuvant<br />

radiation treatment from 2008 to 2011. Of these 80 patients, 24 (30%)<br />

had Medicaid, 31 (39%) had Medicare, and 25 (31%) had commercial<br />

health insurance. Only two (2.5%) patients did not complete treatment<br />

due to transportation. An additional 10 patients (13%) had treatment<br />

interruption due to weather, transportation or other financial reasons.<br />

In patients who completed treatment, a total of 2640 days of treatment<br />

were provided. A total of 55 (2.1%) days of treatment were missed.<br />

Of the 10 patients who had interruption, 5 of them had Medicaid and<br />

the number of missed days by the Medicaid patients was (37/55),<br />

which is 67% of the total days missed. No statistically significant<br />

difference was detected in the number of patients missing treatment<br />

between Medicaid and other insurance types (p=0.16). None of the<br />

factors evaluated predicted patient compliance, although a trend<br />

toward lower compliance was noted in patients who had Medicaid.<br />

Conclusion: Despite being a rural area with multiple barriers to access<br />

healthcare, the rate of full compliance (defined as completion of the<br />

entire course of radiation therapy) with adjuvant radiation therapy in<br />

breast cancer patients in Southeastern Kentucky was 97.5%, which is<br />

similar to the rates in clinical trials that generally exceed 90%. A trend<br />

toward lower compliance was noted in patients who had Medicaid<br />

although it was not statistically significant.<br />

P5-14-06<br />

Analysis of test-therapy concordance for biomarkers uPA and<br />

PAI-1 in primary breast cancer in clinical hospital routine:<br />

Results of a prospective multi-center study at Certified <strong>Breast</strong><br />

<strong>Cancer</strong>s in Germany<br />

Jacobs VR, Augustin D, Wischnik A, Kiechle M, Hoess C, Steinkohl O,<br />

Rack B, Kapitza T, Krase P. Paracelsus Medical University, Salzburg,<br />

Austria; Klinikum Deggendorf, Deggendorf; Klinikum Augsburg,<br />

Augsburg, Germany; Technical University Munich (TUM), Germany;<br />

Cooperative <strong>Breast</strong> Center Ebersberg-Rosenheim. Kreisklinikum<br />

Ebersberg, Ebersberg, Germany; Klinikum Dritter Orden, Munich,<br />

Germany; Ludwig-Maximilian-University (LMU), Munich, Germany;<br />

Top-Expertise, Germering/Munich, Germany; AOK Bayern, Munich<br />

Objective: Biomarkers uPA and PAI-1 are guideline recommended<br />

by ASCO, USA, and AGO, Germany, to be used in primary breast<br />

cancer to avoid unnecessary CTX in medium risk recurrence patients<br />

[G2, N-, HR+, Her2neu-, >35 years]. For quality assurance of uPA/<br />

PAI-1 testing an analysis of test-therapy concordance was performed.<br />

Methods: Prospective, non-interventional, multi-center study<br />

over two years among six Certified <strong>Breast</strong> Centers in Germany to<br />

analyze application of uPA/PAI-1 and use of the result in consecutive<br />

decision making in Tumor Board decision and actual therapy in the<br />

clinical setting for AOK Bayern-insured patients. Concordance and<br />

discordance rates of uPA/PAI-1 testing in daily clinical setting were<br />

calculated and individual reasons for decision making analyzed.<br />

Results: Among n=93 uPA/PAI-1 tests evaluated n=42/93 (45.2%)<br />

were uPA+PAI-1 negative and n=51/93 (54.8%) uPA and/or PAI-1<br />

positive. In the group of uPA+PAI-1 negative test result in n=35/42<br />

(83.3%) CTX was avoided and and in n=7/42 (16.7%) CTX<br />

performed. In the group of uPA and/or PAI-1 positive test result<br />

in n=26/51 (51.0%) CTX was performed and in n=25/51 (49.0%)<br />

www.aacrjournals.org 455s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

not. Concordance of uPA/PAI-1 test result vs. consecutive therapy<br />

was (35+26)/93=65.6% and discordance of uPA/PAI-1 test-therapy<br />

(7+25)/93 = 34.4%. However, discordance of uPA/PAI-1 test result vs.<br />

Tumor Board recommendation was only n=21/93 (22.6%). As reasons<br />

for discordance a variety of influencing and/or interference factors<br />

were indentified, affecting use of test and test results or even changing<br />

therapy decision, e.g. individual therapy decision by physician,<br />

parallel use of competitive biomarkers, study participation, missing<br />

CTX therapy option in advance (like advanced age), wrong target<br />

group, timing of uPA/PAI-1 test too early, test result in contradiction<br />

of physicians’ expectations (expected negative result) and rejection of<br />

CTX recommended by patients despite uPA/PAI-1 test-proven benefit.<br />

Conclusions: Individual process optimization of uPA/PAI-1 test<br />

indication, performance and result assessment could improve quality<br />

of care, reduce costs and lead to an improved application of uPA/PAI-1<br />

test results as well as more stringency and consistency of test-therapy<br />

decisions in daily clinical setting.<br />

P5-15-01<br />

Cost-effectiveness of gene expression profiling for ductal<br />

carcinoma in-situ (Oncotype DCIS Score)<br />

Alvarado MD, Harrison BL, Solin LJ, Ozanne EM. University of<br />

California, <strong>San</strong> Francisco, CA; Albert Einstein Medical Center,<br />

Philadelphia, PA<br />

BACKGROUND: Ductal carcinoma in-situ (DCIS) affects nearly<br />

50,000 women each year. More than 75% of women who receive<br />

lumpectomy (BCS) for DCIS undergo whole breast radiation (XRT),<br />

which is known to prevent local recurrences. A molecular assay for<br />

DCIS has recently been validated, that identifies low-risk biology<br />

and may give insight into the need for adjuvant XRT. We sought to<br />

determine the cost-effectiveness of the Oncotype DCIS Score for<br />

risk stratification in newly diagnosed DCIS.<br />

METHODS: We conducted a cost-effectiveness analysis of using<br />

this molecular assay (validated in the E5194 study) compared to<br />

standard clinical assessment to determine treatment recommendation<br />

for XRT. A Markov model was developed in TreeAge Pro 2011 and<br />

simulated relevant outcomes over a lifetime horizon for 55-year-old<br />

women. For those in the intervention arm who are stratified by the<br />

assay, it was assumed that 75% of women (low DCIS score) did not<br />

receive XRT and had local risk of recurrence (LRR) of 12%; 25%<br />

were intermediate or high risk by the DCIS score (LRR of 26%) and<br />

received 6 weeks of XRT with an assumed LRR of 13%. For those<br />

in the standard care arm who are stratified by clinical assessment,<br />

an assumed 25% did not receive XRT and had LRR of 15.4%; 75%<br />

received 6 weeks of XRT with an estimated LRR of 7.7%. Recurrence<br />

rates were based on ECOG 5194 and assumed 50% reduction with<br />

XRT. Utilities were derived from literature. Direct medical costs<br />

were obtained from Medicare fee schedule; indirect costs (time and<br />

transportation for XRT) were ascertained.<br />

RESULTS: On average, the intervention (assay) strategy was less<br />

costly than the clinical assessment strategy by approximately $1000/<br />

patient, with similar life expectancies (17.15 vs 17.11, respectively)<br />

and quality-adjusted life-years (QALYs) (16.777 vs 16.789). The<br />

incremental cost-effectiveness ratio (ICER) for changing strategy<br />

from the assay to clinical assessment was approximately $95,000/<br />

QALY, at the upper limit of the societal accepted willingness-to-pay<br />

threshold.<br />

CONCLUSIONS: Based on the conservative assumptions that the<br />

benefit of XRT is independent of biology and that at least 75% of<br />

patients in E5194 would typically be offered XRT, the assay strategy<br />

is more cost-effective than standard clinical assessment. Additional<br />

research is needed to better understand health state utilities associated<br />

with less treatment as it pertains to risk of recurrence. Further<br />

validation studies of the assay are needed to accurately assess radiation<br />

benefit across all risk groups.<br />

P5-15-02<br />

The benefit of targeted therapeutics in medical oncology since<br />

the development of trastuzumab<br />

Conter HJ, Conter D, Wolff RA, Valero V, Zwelling L. University of<br />

Texas M.D. Anderson <strong>Cancer</strong> Center, Houston, TX; Huron College,<br />

London, ON, Canada<br />

Background:<br />

Trastuzumab has achieved widespread approval and funding across<br />

much of the developed world for metastatic and, later, adjuvant<br />

HER2/neu positive breast cancer. This success may be attributed<br />

to a favorable therapeutic index and incremental cost-effectiveness<br />

ratio (ICER). Has the success of trastuzumab been replicated by<br />

newer therapeutics?<br />

Methods:<br />

The societal marginal benefit gained from a new drug can be estimated<br />

by calculating the difference between the incremental quality-adjusted<br />

life-years gained (QALY) and the maximum willingness-to-pay<br />

(WTP) for the increase in health by society. A systematic review was<br />

employed to amass all English-language cost-effectiveness analyses<br />

(CEA) on small-molecule inhibitors and monoclonal antibodies<br />

approved for the treatment of solid malignancies. Searches of<br />

PubMed and EMBASE provided citations published between 1995<br />

and February 5, 2012. Two reviewers each independently assessed<br />

the eligibility of all abstracts and subsequently abstracted data<br />

from published abstracts and manuscripts. CEAs comparing two<br />

experimental treatments to each other were excluded. Incremental<br />

costs and ICERs were converted from their native currency to U.S.<br />

dollars according to their average exchange-rates since publication.<br />

Results:<br />

Of the 1,576 citations identified, 60 were included in the final<br />

analysis. Tumor-types studied included breast, colorectal, gastric,<br />

gastrointestinal stromal, head and neck, non-small cell lung, ovarian,<br />

hepatocellular, and renal cancers, and pancreatic neuro-endocrine<br />

tumor. Studies originated from USA (14%), continental Europe<br />

(37%), England (12%), Canada (12%), Asia (11%), and Latin America<br />

(12%). Median WTP was $65,000 (range $30,000-$297,000). 92%<br />

of all CEAs included considered to be cost-effective by the CEA’s<br />

authors. Trastuzumab was studied for breast cancer treatment by 13<br />

CEAs in the adjuvant setting and by 3 CEAs in the metastatic setting.<br />

Trastuzumab is significantly more cost-effective than other targeted<br />

treatments (mean ICER $32,000 vs. $108,000, p=0.001). 84% of<br />

trastuzumab studies found its ICER


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 5<br />

can be made or future therapies produce greater incremental clinical<br />

benefits for the same cost, it seems unlikely that the societal success<br />

of trastuzumab will be replicated. However, this analysis assumes<br />

that maximizing societal health, not profit, is of primary concern.<br />

P5-15-03<br />

Cost-effectiveness of ‘radioguided occult lesion localization’<br />

(ROLL) versus ‘wire-guided localization’ (WGL) in breast<br />

conserving surgery for non-palpable breast cancer: results from<br />

a randomized controlled multicentre trial<br />

Postma EL, Koffijberg E, Verkooijen HM, Witkamp AJ, van den Bosch<br />

MA, van Hillegersberg R. UMC Utrecht; Julius Center Utrecht<br />

Background<br />

Accurate pre-operative localization of non-palpable breast cancer is<br />

essential to achieve complete resection. Radio-guided occult lesion<br />

localization (ROLL) has been introduced as an alternative for wireguided<br />

localization (WGL). Although efficacy of ROLL has been<br />

established in a RCT, cost-effectiveness of ROLL compared with<br />

WGL is not yet known.<br />

Objective. To determine whether ROLL has acceptable costeffectiveness<br />

compared with WGL.<br />

Methods. An economic evaluation was performed alongside<br />

a randomized controlled trial (ClinicalTrials.gov, number<br />

NCT00539474). Women (>18 years) with histologically proven nonpalpable<br />

breast cancer and eligible for breast conserving treatment<br />

with sentinel node procedure were randomized to ROLL(n=162) or<br />

WGL (n=152). Empirical data on direct medical costs was collected,<br />

and changes in quality of life were measured over a 6 months period.<br />

Bootstrapping was used to assess uncertainty in cost-effectiveness<br />

estimates, and sensitivity of the results to the missing data approach<br />

was investigated.<br />

Results. In total, 314 patients with 316 invasive breast cancers were<br />

enrolled. On average ROLL required the same time as WGL for<br />

the initial procedure (119 vs. 118 min.), resulted in a 7% higher reinterventions<br />

risk (27% vs. 20%, p=0.163), a 13% higher complication<br />

risk (30% vs. 17%, p=0.006) but showed similar health effects<br />

(difference 0.00 QALYs 95%CI (-0.04 to 0.05). Total costs were also<br />

similar for ROLL and WGL (+ €26 per patient 95% CI -250 to 311).<br />

Conclusion. ROLL is comparable to WGL with respect to both costs<br />

and quality of life, and will therefore not lead to more cost-effective<br />

medical care.<br />

P5-15-04<br />

CTX and CTX-related direct medication costs saved by testing<br />

biomarkers uPA and PAI-1 in primary breast cancer: Results<br />

of a prospective multi-center study at Certified <strong>Breast</strong> Centers<br />

in Germany<br />

Jacobs VR, Augustin D, Wischnik A, Kiechle M, Hoess C, Steinkohl O,<br />

Rack B, Kapitza T, Krase P. Paracelsus Medical University, Salzburg,<br />

Austria; Klinikum Deggendorf, Deggendorf, Germany; Klinikum<br />

Augsburg, Augsburg, Germany; Technical University Munich<br />

(TUM), Germany; Cooperative <strong>Breast</strong> Center Ebersberg-Rosenheim.<br />

Kreisklinikum Ebersberg, Ebersberg, Germany; Klinikum Dritter<br />

Orden, Munich, Germany; Ludwig-Maximilian-University (LMU),<br />

Munich, Germany; Top Expertise, Germering/Munich, Germany;<br />

AOK Bayern, Munich, Germany<br />

Background: Biomarkers uPA and PAI-1 are guideline recommended<br />

by ASCO, USA, and AGO, Germany, to be used in primary breast<br />

cancer to avoid unnecessary CTX in medium risk recurrence patients<br />

[G2, N-, HR+, HER2neu-, >35 years]. This study was performed to<br />

verify in normal clinical settings how many CTX cycles and how<br />

much CTX-related direct medication costs can be avoided by uPA/<br />

PAI-1 testing.<br />

Methods: Prospective, non-interventional, multi-center study over<br />

two years among six Certified <strong>Breast</strong> Centers in Germany to analyze<br />

application of uPA/PAI-1 and consecutive decision making in the<br />

clinical setting for AOK Bayern-insured patients. CTX regimen<br />

and cycles avoided were identified for each case and direct costs for<br />

CTX and CTX-related medication costs for concomitant medication<br />

calculated for each patient individually according to body weight<br />

and body surface as well as potential FN prophylaxis according<br />

to physicians’ decision. All medication costs were taken from the<br />

pharmaceutical price list for Germany Rote Liste of 2012. EURO [€]<br />

to US Dollar conversion rate as of June 12 2012: € 1.00 = US$ 1.25.<br />

Results: In n=93 breast cancers n=35 CTX (37.6%; FEC n=25,<br />

FEC+DOC n=8 and TC n=2) were avoided according to uPA/PAI-<br />

1 test result. Consecutively 210 CTX cycles or 12.1 years of CTX<br />

application were saved improving patients’ quality of life. uPA/<br />

PAI-1 testing saved direct medication costs for avoided CTX of US$<br />

221.816, CTX-related concomitant medication of US$ 34.353 and<br />

G-CSF prophylaxis of US$ 25.749, overall US$ 281.918. At process<br />

costs for a single uPA/PAI-1 test calculated at US$ 359, uPA/PAI-<br />

1 testing resulted in additional costs of US$ 33.387 for all breast<br />

cancer cases. Overall, testing of uPA/PAI-1 has been proven to be<br />

cost-effective regarding direct medication costs alone at a returnon-investment<br />

ratio of 8.4:1. Indirect cost savings further increase<br />

this ROI.<br />

Conclusions: Innovative and individual cancer diagnostics like<br />

biomarkers uPA/PAI-1 can decrease need of CTX and increase<br />

patients’ quality of life and thereby reduce costs for health care<br />

systems. Since application of this test is inadequate at present time<br />

measures are suggested to fully implement such cost-effective<br />

diagnostics.<br />

P5-15-05<br />

Budget impact analysis of everolimus for estrogen receptor<br />

positive, human epidermal growth factor receptor-2 negative<br />

metastatic breast cancer patients in the United States<br />

Xie J, Diener M, De G, Yang H, Wu EQ, Namjoshi M. Analysis Group,<br />

Inc., New York, NY; Analysis Group, Inc., Boston, MA; Novartis<br />

Pharmaceuticals Corporation, East Hanover, NJ<br />

BACKGROUND: A phase III randomized clinical trial demonstrated<br />

significantly longer progression-free survival of everolimus and<br />

exemestane combination therapy compared to exemestane alone in<br />

postmenopausal women with estrogen receptor positive (ER+), human<br />

epidermal growth factor receptor-2 negative (HER2-) metastatic<br />

breast cancer (MBC) who failed letrozole or anastrozole. This study<br />

estimates the budget impact of adding everolimus as the first or second<br />

treatment after failure of letrozole or anastrozole for post-menopausal,<br />

ER+ HER2- MBC patients from a third-party payer perspective in<br />

the United States.<br />

METHODS: Pharmacy and medical budget impacts were estimated<br />

over the first year of everolimus use in this indication. Published<br />

epidemiology data were used to estimate target population size. Market<br />

share was assumed to be divided between exemestane, fulvestrant<br />

and tamoxifen before everolimus entry based on market research<br />

data on file. Market share of combination therapy of everolimus and<br />

exemestane was assumed to increase linearly during the one-year<br />

period and replace 10% of overall market share by the end of the year,<br />

proportional to the pre-entry market share distribution of the other<br />

three treatments. Pharmacy budget inputs (wholesale acquisition cost<br />

www.aacrjournals.org 457s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

of the therapies, daily dose, treatment duration, dispensing fee and<br />

copayment) were obtained from published and unpublished sources.<br />

Patients were assumed to be on treatment until progression or death.<br />

Medical costs were estimated as the average costs before and after<br />

progression, weighted by time in each state and adjusted for survival.<br />

Pre- and post-progression costs, time to progression and survival rates<br />

were derived from published literature. Compliance was assumed to<br />

be 100% until progression and adverse events were not considered.<br />

All costs were reported in 2011 US dollars.<br />

RESULTS: In a hypothetical health plan with 1 million members,<br />

there would be 72 patients expected to use one of the study drugs<br />

(exemestane, tamoxifen, fulvestrant or everolimus+exemestane) as<br />

the first treatment option immediately after letrozole or anastrozole<br />

failure, and 159 expected to use one of the study drugs as the second<br />

treatment option after letrozole or anastrozole failure. For the first<br />

treatment option after letrozole or anastrozole failure, total budget<br />

impact was $0.013 per member per month (PMPM), including a<br />

$0.017 PMPM increase to the pharmacy budget and $0.004 PMPM<br />

decrease to the medical budget one year after everolimus entry. For<br />

the second treatment option after letrozole or anastrozole failure, total<br />

budget impact was $0.029 PMPM, including a $0.037 PMPM increase<br />

to the pharmacy budget and $0.009 PMPM decrease to the medical<br />

budget one year after everolimus entry. Total budget impact was<br />

$0.042 PMPM, including a $0.055 PMPM increase to the pharmacy<br />

budget and $0.013 PMPM decrease to the medical budget one year<br />

after everolimus entry.<br />

CONCLUSION: Use of everolimus for ER+, HER2- MBC is<br />

expected to reduce the medical budget due to improved efficacy<br />

but increase the pharmacy budget due to higher drug cost. The total<br />

budget impact is relatively small.<br />

P5-15-06<br />

Societal economics of the 21-gene Recurrence Score® in estrogenreceptor-positive<br />

early-stage breast cancer in Japan<br />

Yamauchi H, Nakagawa C, Yamashige S, Takei H, Yagata H, Yoshida<br />

A, Hayashi N, Hornberger J, Yu T, Chien R, Chao C, Yoshizawa C,<br />

Nakamura S. St. Luke’s International Hospital, Chu-o-ku, Tokyo,<br />

Japan; Hitotsubashi University, Tokyo, Japan; Saitama <strong>Cancer</strong><br />

Center, Saitama, Japan; Cedar Associates LLC, Menlo Park, CA;<br />

Stanford University School of Medicine, Stanford, CA; Mailman<br />

School of Public Health, Columbia University, New York, NY;<br />

Genomic Health, Inc., Redwood City, CA; Showa University, Tokyo,<br />

Japan<br />

Background: <strong>Breast</strong> cancer incidence and breast-cancer mortality<br />

have been increasing in Japan. In estrogen-receptor-positive breast<br />

cancer, adding chemotherapy as adjuvant treatment is not definitive<br />

benefit for all women. Primary data on the clinical validity and<br />

decision impact of the 21-gene Recurrence Score for guiding the<br />

use of adjuvant chemotherapy has been studied among a Japanese<br />

population.<br />

Objective: The cost-effectiveness of the clinically validated 21-<br />

gene Recurrence Score for estrogen-receptor-positive, lymph-nodenegative,<br />

early-stage breast cancer (ESBC) was assessed from a<br />

Japanese societal perspective.<br />

Method: The proportion of patients with low, intermediate, and high<br />

risk of recurrence by the 21-gene Recurrence Score and the influence<br />

of the Recurrence Score on use of adjuvant chemotherapy were<br />

obtained in a study of 104 patients. A Markov model was used to<br />

track time until distant recurrence and time from distant recurrence to<br />

death. Effect of adjuvant chemotherapy based on 21-gene Recurrence<br />

Score was based on published clinical validation studies. Direct and<br />

indirect medical costs were obtained from the referral center. Utilities<br />

associated with progression and adverse events associated with<br />

chemotherapy were extracted from the literature. Sensitivity analyses<br />

were performed to assess the key drivers of cost-effectiveness and<br />

the robustness of the findings to variations in the input estimates.<br />

Results: Forty-eight percent of patients were identified by the<br />

21-gene Recurrence Score as low-risk, 36% as intermediate-risk,<br />

and 16% as high-risk. In treatment decision by using the 21-gene<br />

Recurrence Score, chemotherapy use overall decreased by 19%,<br />

resulting in an average increase of 0.235 QALYs. Testing with the<br />

21-gene Recurrence Score cost ¥350,000 per patient with ¥153,490<br />

lower acute costs because fewer women were treated with adjuvant<br />

chemotherapy. Eighteen percent more women who were identified as<br />

high risk were recommended adjuvant chemotherapy, which results<br />

in longer progression-free survival. On average across all risk groups,<br />

cost of monitoring until recurrence per patient increased by ¥3,572<br />

and costs associated with recurrence declined by ¥43,687. Use of the<br />

21-gene Recurrence Score resulted in a net cost of ¥665,455 ($8,386)<br />

per QALY gained.<br />

Conclusions: The change in decisions resulting from use of the 21-<br />

gene Recurrence Score in women with estrogen-receptor-positive,<br />

lymph-node-negative, ESBC is projected to be societally costeffective<br />

in Japan.<br />

P5-15-07<br />

Time savings with trastuzumab subcutaneous (SC) injection<br />

vs. trastuzumab intravenous (IV) infusion: First results from a<br />

Time-and-Motion study (T&M)<br />

de Cock E, Tao S, Alexa U, Pivot X, Knoop A. United Biosource<br />

Corporation, Barcelona, Spain; United Biosource Corporation,<br />

Montreal, Canada; F Hoffmann-La Roche Ltd, Basel, Switzerland;<br />

CHU Jean Minjoz, Besancon, France; Odense University Hospital,<br />

Odense, Denmark<br />

Objective:<br />

To quantify resource utilization in terms of “active” health care<br />

professional (HCP) time and consumables, as well as associated costs,<br />

related to trastuzumab (TRA) SC injection and TRA IV infusion in<br />

the treatment of patients with HER2-positive EBC within the PrefHer<br />

trial setting. Using time and resource use inputs, we will estimate<br />

average time and cost for SC and IV processes on a per patient and<br />

per site basis, as well as estimate potential time and cost savings with<br />

a switch from IV to SC administration route per site and per country.<br />

Methods:<br />

This is a multi-country (6 countries), multi-centre (17 centres),<br />

prospective, T&M study, run as a sub-study to the PrefHer trial<br />

(ClinicalTrials.gov identifier NCT01401166). Using a structured<br />

questionnaire, we conducted on-site interviews with a nurse and<br />

pharmacy member in each centre to elicit practice pattern flow and<br />

to obtain time estimates for trastuzumab-related tasks for both IV and<br />

SC processes. The information served to tailor generic Case Report<br />

Forms (CRFs) to reflect centre-specific practices. For each task we<br />

defined adequate “start” and “stop” descriptions for pre-specified<br />

tasks to ensure accurate and unbiased collection of T&M data. Data<br />

collection is performed by trained observers who measure time<br />

using a stop watch and record onto paper CRFs. This is a descriptive<br />

non-comparative study with convenience-based sample sizes<br />

ranging between 10-30 observations per process per site. While data<br />

collection is currently ongoing (final data expected during Q4 2012<br />

and are planned to be reported at the meeting), first results presented<br />

in this abstract are based on interview data and for SC process on<br />

preliminary data from the T&M study (in the absence of staff-elicited<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

458s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 5<br />

time estimates for SC process due to lack of experience). Estimated<br />

total time is calculated as the sum of individual task times and results<br />

are pooled across all sites.<br />

Results:<br />

Patient hospital arrival/registration, blood sampling, and physician<br />

visit account for a median 39 minutes (min) and are expected to be<br />

similar for both processes (i.e. “non-observed” time; not included<br />

in T&M data collection). Median total HCP time for tasks included<br />

in the T&M study (i.e. “observed” time) is estimated at 59 and 20<br />

min, respectively (estimated 66% reduction with SC). For IV, the<br />

majority of “observed” task time is for account of TRA pharmacy<br />

reconstitution (25%) and infusion initiation (17%), with the remaining<br />

58% distributed across other care unit tasks. For SC, injection<br />

(35%) and TRA pharmacy dispensing (30%) constitute the bulk of<br />

“observed” task time, with 35% for other care unit tasks. Time savings<br />

are expected because of avoiding time related mainly to infusion line<br />

(dis)connection, and IV pharmacy reconstitution, tasks which are only<br />

partially being replaced by SC injection and discarding.<br />

Conclusion: A switch from IV to SC TRA may result in important<br />

care unit and pharmacy time savings to be reinvested in improving<br />

overall patient care. Patients could potentially be moved out of<br />

the chemotherapy care unit to receive SC administration in other<br />

settings and free up valuable chair time, thereby increasing the unit’s<br />

throughput and overall efficiency.<br />

P5-16-01<br />

Survival advantage in patients with metastatic breast cancer<br />

receiving endocrine therapy plus Sialyl Tn-KLH vaccine: post<br />

hoc analysis of a large randomized trial<br />

Ibrahim NK, Murray JL, Zhou D, Mittendorf EA, Sample D, Tautchin<br />

M, Miles D. University of Texas MD Anderson <strong>Cancer</strong> Center,<br />

Houston, TX; Biomira, Inc., AB, Canada; Mount Vernon <strong>Cancer</strong><br />

Center, Northwood, Middlesex, United Kingdom<br />

Background<br />

A multicenter, double blinded, randomized phase III trial of<br />

the therapeutic cancer vaccine STn-KLH was completed in an<br />

international cohort of 1,028 women with metastatic breast cancer who<br />

had either no evidence of disease or nonprogressive disease following<br />

first-line chemotherapy. This trial is registered with ClinicalTrials.<br />

gov (No. NCT00003638). The outcomes showed that STn-KLH<br />

was safe and relatively well tolerated but had neither a positive nor<br />

negative effect on time to progression (TTP) or overall survival (OS)<br />

duration in the intent-to-treat population when compared with KLH<br />

control alone. The purpose of this post hoc analysis is to explore the<br />

potential benefit of combining an antiestrogen with MUC1 vaccines<br />

in metastatic breast cancer patients.<br />

Methods<br />

The data were further explored to determine if a retrospective,<br />

reassigned endocrine subset patient stratification produces subgroups<br />

that may have experienced benefit in TTP or survival compared with<br />

the phase III trial ITT analysis.<br />

Results<br />

Women treated with concomitant endocrine therapy, a pre stratified<br />

subset comprising approximately one third of the original study<br />

population, achieved a clinical benefit both in terms of TTP and<br />

survival compared with women who did not receive endocrine<br />

therapy. Moreover, women in the endocrine-treatment subset who<br />

mounted a median or greater antibody response (titer >1:320 toward<br />

bovine submaxillary mucin) to the STn-KLH vaccine experienced<br />

significantly longer median survival than their trial counterparts who<br />

mounted a below-median antibody response.<br />

Conclusion<br />

Unlike maintenance chemotherapy, with its associated cumulative<br />

toxicity, the combination of endocrine and STn-KLH therapy may<br />

offer clinical benefit with few adverse effects for women with<br />

metastatic breast cancer.<br />

P5-16-02<br />

Final Results of the Phase I/II Trials of the E75 Adjuvant <strong>Breast</strong><br />

<strong>Cancer</strong> Vaccine<br />

Vreeland TJ, Clifton GT, Hale DF, Sears AK, Patil R, Holmes JP,<br />

Ponniah S, Mittendorf EA, Peoples GE. <strong>San</strong> <strong>Antonio</strong> Military Medical<br />

Center, <strong>San</strong> <strong>Antonio</strong>, TX; Joyce Murtha <strong>Breast</strong> Care Center, Windber,<br />

PA; Redwood Regional Medical Group, <strong>San</strong>ta Rosa Memorial<br />

Hospital, <strong>San</strong>ta Rosa, CA; Uniform Services University of Health<br />

Sciences, Bethesda, MD; MD Anderson <strong>Cancer</strong> Center, Houston, TX<br />

Background: We have completed phase I/II clinical trials vaccinating<br />

breast cancer patients (pts) with E75, a HLA-A2/A3-restricted HER2/<br />

neu (HER2) peptide vaccine. The vaccine was administered in the<br />

adjuvant setting to prevent recurrences in high risk patients rendered<br />

disease-free with standard of care therapy. We have previously<br />

reported preliminary results indicating that the vaccine (including<br />

booster inoculations) is safe and effective in stimulating an antitumor<br />

immune response. Here, we report the final 5 year results<br />

from these trials.<br />

Methods: The phase I/II trials were performed as dose-escalation/<br />

schedule-optimization trials enrolling node positive and high-risk,<br />

node negative breast cancer patients with tumors expressing any level<br />

of HER2. HLA-A2/A3+ pts were enrolled into the vaccine group<br />

(VG) while HLA-A2/A3- pts were followed prospectively as the<br />

untreated control group (CG). The VG pts were given 4-6 monthly<br />

intradermal inoculations of E75 with GM-CSF during the primary<br />

vaccine series (PVS). In addition, a voluntary booster program was<br />

initiated during the trial, with booster inoculations being offered every<br />

6 months after completion of the PVS. Patients were monitored for<br />

local and systemic toxicity (graded by NCI Common Terminology<br />

Criteria for Adverse Events). In vivo immune response was assessed<br />

in the VG by delayed type hypersensitivity (DTH) reactions to both<br />

E75 and saline, pre- and post-PVS. VG and CG pts were followed<br />

for 60 months (mo) and recurrences were documented. Demographic<br />

differences were compared with the Fisher’s exact test and diseasefree<br />

survival was determined using the Kaplan-Meier method and<br />

compared by log-rank test.<br />

Results: 195 pts were enrolled, 6 withdrew (2 from VG, 4 from CG),<br />

1 was lost to follow-up prior to vaccination, and 1 was found to be<br />

ineligible, leaving 187 evaluable pts; 108 in the VG and 79 in the<br />

CG. 53 pts volunteered for the booster program and received at least<br />

one booster inoculation. The VG and CG were well-matched with the<br />

only statistically significant difference being ER-/PR- status (31.1%<br />

in VG vs 17.7% in CG, p=0.04). Vaccination was well tolerated<br />

(maximum local toxicity: 73.1% Grade 1, 26.9% Grade 2, 0% Grade<br />

3; maximum systemic toxicity: 72.2% Grade 1, 15.7% Grade 2, and<br />

2.8% Grade 3). In the VG, pre- to post-PVS E75 DTH significantly<br />

increased (mean 3.8 ±1.0 vs 14.8±1.4, p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

At the end of the 5 year follow-up period, the E75 vaccine shows a<br />

strong trend toward preventing breast cancer recurrence in vaccinated<br />

patients. To investigate this vaccine (now known as NeuVax) further,<br />

the PRESENT trial, a prospective, randomized, double-blind, placebocontrolled,<br />

multi-center phase III registration trial has been initiated<br />

and is actively enrolling.<br />

P5-16-03<br />

Phase II study of autologous dendritic cell vaccination in patients<br />

with HER2 negative breast cancer combined with neoadjuvant<br />

chemotherapy<br />

Castillo A, Salgado E, Inoges S, Arevalo E, Castañon E, López Díaz<br />

de Cerio A, Sola JJ, Pina L, Nuñez J, Rodriguez-Spiteri N, Espinós<br />

J, Aramendia JM, Fernandez Hidalgo OA, <strong>San</strong>tisteban M. Clínica<br />

Universidad de Navarra, Pamplona, Navarra, Spain; Complejo<br />

Hospitalario de Navarra, Pamplona, Navarra, Spain; Universidad de<br />

Navarra, Pamplona, Navarra, Spain; Clínica Universidad de Navarra<br />

Background The limited efficacy of neoadjuvant schedules to obtain<br />

pCR in breast cancer together with a prompt relapse of triple negative<br />

breast cancers has opened up the development of new strategies<br />

involving the immune system, which is essential in cancer control.<br />

We have elaborated an autologous vaccine with dendritic cells loaded<br />

with patients’ own tumor antigens. Based on the synergistic effect<br />

between immunotherapy and chemotherapy our hypothesis is that the<br />

combination of both strategies could improve pCR with an excellent<br />

tolerance and could increase DFS and OS. Methods Two centers are<br />

enrolling patients from February 2011. Twenty-one patients with<br />

stage II-III HER2 negative breast cancer have started on sequential<br />

neoadjuvant chemotherapy based on dose dense antracyclines (E<br />

100mg/m 2 and C 600 mgr/m 2 ) x4 cycles plus GM-CSF followed by<br />

taxanes (DOC 75-100 mgr/m 2 ) x4 cycles plus vaccination. Vaccine<br />

calendar was started after the 4 th EC and alternated with taxane<br />

schedule and after chemotherapy up to a maximum of a 2 year-period.<br />

SNB biopsy was performed before neoadjuvant chemotherapy.<br />

pCR in the breast and the axilla is the main endpoint, measured by<br />

Miller&Paine classification. Data were analyzed using SPSS and<br />

alpha was set at 0.05 (two-tailed). We have compared these results in<br />

the vaccinated cohort (V) with our historic cohort (C) of 19 patients<br />

treated in the same way but without the vaccine. Results Both cohorts<br />

(V and C) were well balanced for age, histologyc type, differentiation<br />

grade, Ki67>15%, tumor size ≤ 5 cm, stage and expression of hormone<br />

receptors. All patients except one in the vaccine cohort achieved 8<br />

cycles of chemotherapy. According to grade ¾ toxicities we did not<br />

find significant differences except for hand-foot syndrome in the V<br />

cohort (19% vs 0%; p=0.04) and vomiting in the C cohort (0% vs<br />

21%; p=0.02). Lymphopenia was the most common adverse event<br />

(81% in V vs 89.5% in C; p= 0.45). Other toxicities were anemia (0%<br />

vs 5.3%; p=0.28), thrombopenia (0% vs 10% p=0.12), leukopenia<br />

(9.5% vs 15.8% p=0.55) and neutropenia (14% vs 21%, p=0.57).<br />

Gastrointestinal toxicity was similar in both groups: mucositis (0%<br />

vs 5.3%, p=0.28); gastritis (5% in both cohorts), nausea (19% vs<br />

16%, p= 0.78), diarrhea (5% in both cohorts), constipation (0% vs<br />

15% in C, p=0.06). Myalgias were seen in 19% in V vs 11% in C<br />

(p=0.45). We did not see any local grade ¾ toxicity related to the<br />

intradermal vaccines. Regarding efficacy data, clinical complete<br />

response by MRI was shown in 38% in the V as compared to 16% in<br />

the C (p=0.06). <strong>Breast</strong>-conserving surgery was more common in the<br />

V cohort (62% vs 53%, p=0.55). Total pCR (T plus N) was superior<br />

with the addition of the vaccine (24% vs 5% p=0.014). Moreover<br />

in three more patients in the V group we find isolated tumor cells in<br />

the breast (T itc) . Conclusions The addition of autologous dendritic<br />

cell vaccines to neoadjuvant chemotherapy significantly increased<br />

the rate of pathological complete responses among patients with<br />

HER2-negative breast cancer. Vaccination does not seem to add any<br />

toxicity to the schedule.<br />

P5-16-04<br />

A phase I study of a DNA plasmid based vaccine encoding the<br />

HER-2/neu intracellular domain in subjects with HER2+ breast<br />

cancer.<br />

Salazar LG, Slota M, Higgens D, Coveler A, Dang Y, Childs J, Bates<br />

N, Guthrie K, Waisman J, Disis ML. University of Washington, Seattle,<br />

WA; BREASTLINK, Hawthorne, CA<br />

HER2+ breast cancer (BC) is associated with early disease relapse,<br />

usually to distant sites. This would suggest relapse is due to residual<br />

microscopic disease. Generation of vaccine-induced HER2-specific<br />

CD4+ T helper immunity (Th1) may result in immunologic<br />

eradication of residual HER2+ tumor cells and subsequent<br />

development of immunologic memory and epitope spreading (ES),<br />

which has been associated with a survival benefit in vaccinated BC<br />

patients. We have shown HER2 peptide-based vaccines can generate<br />

immunity in BC however, more recently we developed a plasmid<br />

DNA based vaccine (pNGVL3-hICD) which may have additional<br />

advantages over synthetic peptides. DNA vaccines offer a strategy<br />

to immunize against multiple tumor antigens and are able to elicit<br />

both CTL and Th1 immunity. Plasmid DNA can also remain at the<br />

vaccine site, providing a constant source of antigen. Intradermal<br />

(i.d.) delivery of DNA vaccines with GM-CSF as adjuvant may<br />

enhance immunogenicity due to local influx of dermal Langerhans<br />

cells. We have recently completed a phase I trial utilizing pNGVL3-<br />

hICD in optimally treated stage III and IV HER2+ BC patients and<br />

have defined vaccine safety profile, optimal dose and schedule; and<br />

demonstrated vaccine biologic activity.<br />

Methods: A total of 66 subjects with stage III and IV HER2+ BC in<br />

complete remission were enrolled sequentially into 1 of 3 pNGVL3-<br />

hICD dose arms (22 subjects/arm): Arm 1=10µg, Arm 2=100 µg, and<br />

Arm 3 =500µg. All vaccines were admixed with 100µg GM-CSF and<br />

given i.d. monthly for a total of 3 vaccines. Toxicity was assessed at<br />

baseline, during vaccination and at follow-up. Immune responses to<br />

HER ICD and ECD were assessed with IFN-γ ELISPOT at baseline<br />

and serially through week 60 post-vaccination. Linear regression<br />

analysis was used to compare differences in immune responses from<br />

baseline over the whole study period between dose arms. Vaccine site<br />

skin biopsies and peripheral lymphocytes were serially analyzed for<br />

plasmid persistence via RT-PCR.<br />

Results: 64 subjects (20 in Arm 1; 22 in Arm 2; 22 in Arm 3)<br />

completed 3 vaccines. Age, stage/status, number of previous<br />

chemotherapy regimens, and use of bisphosphonate and trastuzumab<br />

therapies was similar across dose arms. Vaccine-related toxicity was<br />

primarily Grade 1/2 injection site reactions, myalgias, arthralgias<br />

and not significantly different between arms; no cardiac or grade<br />

IV toxicity was observed. Immune responses to HER2 ICD were<br />

significantly better in Arms 2 and 3 vs Arm 1 (p=0.001 and 0.002,<br />

respectively) but not statistically different between Arms 2 and 3. 38<br />

patients had DNA plasmid persistence at the vaccination site with no<br />

difference between arms. There has been no detection of DNA plasmid<br />

in lymphocytes from patients in all arms. Analyses of survival and ES<br />

(HER ECD immune responses) are on-going and will be presented.<br />

Conclusions: pNGVL3-hICD was safe and effectively induced<br />

persistent HER2 ICD specific Th1 immunity without increased cardiac<br />

toxicity. Moreover, immunity was present more than 1 year after end<br />

of vaccination, indicative of vaccine-induced immunologic memory.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

460s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 5<br />

P5-16-05<br />

The combination of trastuzumab and HER2-directed peptide<br />

vaccines is safe in HER2-expressing breast cancer patients<br />

Hale DF, Vreeland TJ, Perez SA, Berry JS, Ardavanis A, Trappey<br />

AF, Tzonis P, Sears AK, Clifton GT, Shumway NM, Papamichail<br />

M, Ponniah S, Peoples GE, Mittendorf EA. Brooke Army Medical<br />

Center, <strong>San</strong> <strong>Antonio</strong>, TX; <strong>Cancer</strong> Immunology and Immunotherapy<br />

Center, Athens, Greece; MD Anderson <strong>Cancer</strong> Center, Houston, TX;<br />

Uniformed Services University of the Health Sciences, Bethesda, MD<br />

Background: Cardiotoxicity is the most concerning toxicity associated<br />

with the commonly used HER2-directed immunotherapy, trastuzumab<br />

(Tz). In general, a significant decline of left ventricular ejection<br />

fraction (EF) in asymptomatic patients is accepted as a decrease of<br />

at least 10% or an absolute value of below 50%. We are currently<br />

conducting multiple trials of HER2-directed peptide vaccines, often<br />

given either concurrently or in close temporal proximity to Tz. This<br />

has raised the issue that combining therapies could increase the risk<br />

of cardio-toxicity. Here, we present safety data from multiple trials<br />

in which the combination of these HER2-directed therapies was<br />

administered.<br />

Methods: Phase I and II trials were conducted in disease-free breast<br />

cancer patients after completion of chemotherapy when indicated.<br />

Patients (pts) who were determined by treating oncologists to qualify<br />

for Tz received this therapy per standard-of-care. These pts were<br />

enrolled onto HER2-directed peptide vaccine trials per each trial’s<br />

inclusion criteria, with vaccinated (VG) pts receiving peptide + GM-<br />

CSF and control (CG) pts receiving GM-CSF alone. All patients were<br />

monitored for local and systemic toxicity to peptide inoculations<br />

(graded by the NCI’s Common Terminology Criteria for Adverse<br />

Events). In addition, patients who received Tz had EF tracked<br />

through either echocardiogram or MUGA according to local standard<br />

of practice. Our database was queried for patients who received Tz<br />

and peptide, and had documented measures of EF pre-vaccine (Pre),<br />

during vaccine (D) and post-vaccine (Post). These pts were then<br />

placed in two groups based on the timing of Tz and vaccine therapy:<br />

concurrent(C) group and sequential(S) group. Mean EF at each time<br />

point was compared using a t-test.<br />

Results: Overall, the peptide vaccines were well tolerated (max local<br />

tox: 1% Grade 0, 65% Gr 1, 33% Gr 2, 1% Gr 3; max systemic tox:<br />

19% Gr 0, 63% Gr 1, 18% Gr 2, 0% Gr 3). These toxicities are likely<br />

secondary to the GM-CSF immunoadjuvant as control pts receiving<br />

GM-CSF alone have similar toxicity profiles (max local tox: 0%<br />

Gr 0, 76% Gr 1, 23% Gr 2, 1% Gr 3; max systemic tox: 20% Gr 0,<br />

65% Gr 1, 15% Gr 2, 0% Gr 3). There have been no serious or nonserious<br />

cardiac-related adverse events in our trials. In total, 71 pts<br />

treated with Tz and enrolled in a vaccine trial had EF measurements<br />

available for analysis; 54 in the S group (35 VG, 19 CG) and 17 in<br />

the C group (10 VG, 7 CG). Overall, neither VG nor CG pts had<br />

significant changes in EF (VG Pre: 65±0.8%, D: 63±0.9%, Post:<br />

64±0.3%; CG Pre: 63±1.2%, D: 64±1.8%, Post: 63±1.1%). Separating<br />

VG pts into C and S pts, there were again no significant changes in<br />

EF, (C Pre: 65±1.0%, D: 63±1.0%, Post: 63±1.4%; S Pre: 65±1.7%,<br />

D: 61±1.3%, Post: 65±1.8%).<br />

Conclusions: HER2-directed peptide vaccines are safe and well<br />

tolerated. Initial data indicate that the combination of Tz and HER2-<br />

directed peptide vaccines, whether concurrent or sequential, does not<br />

cause significant cardiac toxicities as measured by changes in the EF<br />

during and after therapy. We will continue to track this safety data<br />

to confirm early findings as we pursue additional combination trials.<br />

P5-16-06<br />

A phase 2 randomized trial of docetaxel (DOC) alone or in<br />

combination with therapeutic cancer vaccine, CEA-, MUC-1-<br />

TRICOM (PANVAC)<br />

Heery CR, Ibrahim NK, Mohebtash M, Madan RA, Arlen PM, Bilusic<br />

M, Kim JW, Singh NK, Hodge S, McMahon S, Steinberg SM, Hodge<br />

JW, Schlom J, Gulley JL.<br />

Background<br />

A previous phase 1/2 trial of PANVAC, a poxviral based cancer<br />

vaccine, suggested clinical efficacy in some patients (pts) with breast<br />

and ovarian cancer and evidence of immunologic activity. Preclinical<br />

data showed DOC can modify tumor phenotype, making tumor cells<br />

more amenable to T-cell mediated killing. The goal was to determine<br />

if DOC and PANVAC could synergize and improve clinical outcomes<br />

compared with DOC alone.<br />

Methods<br />

This is an open-label randomized phase 2 multi-center trial designed<br />

to enroll 48 pts with metastatic breast cancer to receive DOC in<br />

combination with PANVAC (A) or alone (B). Cross-over was<br />

allowed so that pts randomized to B could receive the vaccine upon<br />

progression. Eligibility included ECOG performance status


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P5-17-01<br />

Bevacizumab (B) in the adjuvant treatment of breast cancer -<br />

first toxicity results from Eastern Cooperative Oncology Group<br />

trial E5103.<br />

Miller KD, O’Neill A, Dang C, Northfelt D, Gradishar W, Sledge GW.<br />

Indiana University Melvin and Bren Simon <strong>Cancer</strong> Center; Dana<br />

Farber <strong>Cancer</strong> Institute; Memorial Sloan Kettering <strong>Cancer</strong> Center;<br />

Mayo Clinic; Northwestern University<br />

Background: A previous feasibility trial (E2104 – Ann Oncol<br />

23(2):331-7,2012) suggested incorporation of B into anthracyclinecontaining<br />

adjuvant therapy was feasible but ongoing cardiac<br />

monitoring was required to define the true impact of B on cardiac<br />

function. Methods: Patients (pts) were assigned 1:2:2 to one of three<br />

treatment arms. In addition to doxorubicin and cyclophosphamide<br />

followed by weekly paclitaxel, patients received either placebo (Arm<br />

A – AC>T) or B during chemotherapy (Arm B - BAC>BT), or B<br />

during chemotherapy followed by B monotherapy (15 mg/kg q3wk)<br />

for an additional 10 cycles (Arm C – BAC>BT>B). Randomization<br />

was stratified and B dose adjusted for choice of AC schedule (classical<br />

q3wk - 15 mg/kg; dose dense(dd) q2 wk - 10 mg/kg). When indicated,<br />

radiation and hormonal therapy were administered concurrently<br />

with B (for Arm C pts). The primary cardiac endpoint was the<br />

incidence of clinically apparent cardiac dysfunction (CHF)defined<br />

as symptomatic decline in left ventricular ejection fraction (LVEF)<br />

to below the lower limit of normal (LLN) or symptomatic diastolic<br />

dysfunction as assessed by independent review. Cumulative toxicity<br />

data as of Jan 23, 2012 are presented. Results: From 11.07 to 2.11,<br />

4994 pts were enrolled. Median age was 52; 80% received ddAC.<br />

Chemotherapy associated toxicities including myelosuppression<br />

(Grade 4 neutropenia 16/20/19%) and neuropathy (Grade ≥ 3<br />

8/8/8%) were similar across all arms. Grade ≥ 3 hypertension/<br />

thrombosis/proteinuria/hemorrhage was reported by 7/3/10% & LVEF < LLN 1% 2% 2%<br />

Post C8 LVEF Arm A Arm B Arm C<br />

Baseline and post Cycle 8 LVEF available 847 1685 1674<br />

Median post Cycle 8 LVEF 61 60 60<br />

% with post Cycle 8 LVEF decline > 10% 12% 18% 18%<br />

% with post Cycle 8 drop >10% & LVEF < LLN 2% 6% 6%<br />

Conclusion: Incorporation of B into anthracycline and taxane<br />

containing adjuvant therapy results in a significant but small increase<br />

in clinical CHF. The rate of clinical CHF is similar to that predicted<br />

by E2104 (2.5-2.9%) and reported In the FDA label for anthracycline<br />

pre-treated pts(3.8%). No unexpected toxicities were encountered.<br />

P5-17-02<br />

Withdrawn by Author<br />

P5-17-03<br />

Quality of life (QoL) results from the TURANDOT trial<br />

comparing two bevacizumab (BEV)-containing regimens as firstline<br />

treatment for HER2-negative metastatic breast cancer (mBC)<br />

Láng I, Inbar MJ, Kahan Z, Greil R, Beslija S, Stemmer SM,<br />

Kaufman B, Ahlers S, Brodowicz T, Zielinski C. National Institute of<br />

Oncology, Budapest, Hungary; Tel Aviv Sourasky Medical Centre,<br />

Tel Aviv, Israel; University of Szeged, Hungary; Paracelsus Medical<br />

University, Salzburg, Austria; Institute of Oncology, Sarajevo, Bosnia<br />

and Herzegowina; Rabin Medical Center, Petah-Tikva, Israel;<br />

Sheba Medical Center, Ramat Gan, Israel; IST GmbH Mannheim,<br />

Mannheim, Germany; Medical University of Vienna and Central<br />

European Cooperative Oncology Group (CECOG), Vienna, Austria<br />

Background: BEV has been shown to significantly improve the<br />

efficacy of both paclitaxel (PAC) and capecitabine (CAP) as firstline<br />

therapy for HER2-negative mBC. TURANDOT, a randomized<br />

phase III trial comparing BEV-PAC vs BEV-CAP, includes extensive<br />

QoL evaluation.<br />

Methods: Patients (pts) with HER2-negative mBC who had received<br />

no prior chemotherapy for mBC were randomized to receive either<br />

BEV-PAC (BEV 10 mg/kg d1 & 15 + PAC 90 mg/m 2 d1, 8, & 15<br />

q4w) or BEV-CAP (BEV 15 mg/kg d1 + CAP 1000 mg/m 2 bid d1-14<br />

q3w) until disease progression, unacceptable toxicity, or withdrawal of<br />

consent. The primary endpoint is overall survival (OS); QoL, assessed<br />

using the EORTC QLQ-C30, is a secondary endpoint. Pts were asked<br />

to complete QLQ-C30 at baseline, q12w during study therapy, at the<br />

end of treatment, and 28 days after discontinuing from the study.<br />

Results: On-treatment QoL questionnaires were available from all 561<br />

of the treated pts at baseline, 394 at week 12, 266 at week 24, 186 at<br />

week 36, 129 at week 48, and 50 pts ranged from +1.5 to +2.6. In the BEV-CAP arm, mean<br />

baseline score was 56.0; the mean score remained above the baseline<br />

mean score until week 96. Mean change from baseline ranged from<br />

-3.9 at week 84 to +4.9 at week 96. Mean scores for most individual<br />

symptom scales were low (typically


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 5<br />

P5-17-04<br />

Management of Antiangiogenics’ Renovascular Safety in breast<br />

cancer. Subgroup and intermediate results of the MARS Study.<br />

Launay-Vacher V, Janus N, Daniel C, Rey J-B, Ray-Coquard I,<br />

Gligorov J, Spano J-P, Thery J-C, Jouannaud C, Goldwasser F, Mir<br />

O, Morere J-F, Oudard S, Azizi M, Dorent R, Deray G, Beuzeboc P.<br />

Institut Curie, Paris, France; Hôpital de la Pitié-Salpêtrière, Paris,<br />

France; Institut Jean Godinot, Reims, France; Centre Léon Bérard,<br />

Lyon, France; Hôpital Tenon, Paris, France; Hôpital Cochin, Paris,<br />

France; Hôpital Avicenne, Bobigny, France; Hôpital Européen<br />

Georges Pompidou, Paris, France<br />

Background:<br />

Hypertension (HTN) and proteinuria (Pu) are class-side-effects<br />

of anti-VEGF drugs (AVD), related to the inhibition of the VEGF<br />

pathway. The MARS study has been conducted to assess the<br />

renovascular tolerance of these drugs in the clinical setting.<br />

Methods:<br />

The MARS study is a multicentric prospective observational study of<br />

the renovascular safety of AVD in AVD-naive patients. in 7 centres<br />

from 2009 to 2011. There was no intervention on the choice of the<br />

AVD. Data collected included: gender, age, serum creatinine (SCr),<br />

diabetes, HTN, hematuria (Hu) and dipstick Pu. This sub-group<br />

analysis presents the intermediate results for the first 184 patients<br />

with breast cancer (BC) receiving bevacizumab who completed the<br />

1-year follow-up (f/u) (out of 337 BC patients in total).<br />

Results:<br />

All these 184 patients received bevacizumab (mean and median<br />

durations of treatment: 7.8 and 8 months, respectively). Median age<br />

at inclusion was 60 years. Bone, visceral and cerebral metastasis<br />

frequencies were 75.0, 52.2 and 7.1%, respectively. Diabetes and<br />

HTN prevalences were 4.3% and 10.3%, respectively. Baseline<br />

renal assessment retrieved: Pu 13.0%, Hu 8.2%, mean aMDRD<br />

98.8 ml/min/1.73m 2 and 1.1% had aMDRD


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P5-17-06<br />

The deficient eNOS c.894G>T genotype is not associated with<br />

increased severity of hypertension and proteinuria in breast<br />

cancer patients receiving bevacizumab<br />

Del Re M, Ferrarini I, Fontana A, <strong>San</strong>toro M, Bona E, Del Re I, Stasi<br />

I, Bertolini I, Laurà F, Landucci E, Salvadori B, Falcone A, Danesi<br />

R. University Hospital, Pisa, Italy<br />

Background. Tumor angiogenesis is a complex process involving a<br />

wide array of effector molecules and stromal cells. In tumor tissue,<br />

vasculature is structurally and functionally abnormal, causing<br />

elevated interstitial pressure and irregular perfusion. The expression<br />

of vascular endothelial growth factor (VEGF), the most important<br />

angiogenic factor, is enhanced in many tumors. VEGF may induce<br />

nitric oxide (NO) production via up-regulation of the endothelial NO<br />

synthase (eNOS, NOS3) and the resultant overproduction of NO is<br />

associated with vasodilation and edema within tumors (Goel S et al.<br />

Physiol Rev 2011;91:1071). eNOS plays an important physiologic<br />

role in maintaining blood pressure homeostasis and vascular<br />

integrity by providing constitutive release of NO in endothelial<br />

cells. Functional variants of the eNOS gene, including the singlenucleotide<br />

polymorphism rs1799983 (c.894G>T, p.Asp298Glu) at<br />

codon 298, have been associated with reduced function of eNOS<br />

and higher incidence of hypertension (HT) (Niu W, Qi Y. PLoS One<br />

2011;6:e24266).<br />

Purpose. Since suppression of VEGF-eNOS axis by anti-angiogenic<br />

therapies is considered a causative factor of HT in patients, the<br />

purpose of this study was to examine whether the major eNOS nonsynonymous<br />

variant c.894G>T may be associated with increased<br />

risk of developing hypertension (HT) and proteinuria (PU) in breast<br />

cancer patients treated with bevacizumab.<br />

Patients and methods. Forty-one metastatic breast cancer patients<br />

given bevacizumab as per standard of care were enrolled. Main<br />

characteristics were: median age 49.5 years (range 29-73) at first<br />

diagnosis, 53 years (range 34-74) at metastatic disease; PS 0-1 in all<br />

patients; 4 subjects with hypertension and 1 patient with compensated<br />

cardiovascular disease at diagnosis. Twenty-six subjects had received<br />

neoadjuvant or adjuvant chemotherapy based on anthracycline and<br />

taxane; first-line chemotherapy for metastatic disease was paclitaxel<br />

plus bevacizumab for all patients; 14 subjects received hormonetherapy<br />

for metastatic disease (range 1-5 lines). Germline DNA was<br />

extracted from peripheral blood and used to screen patients for eNOS<br />

c.894G>T variant by automatic sequencing. The study was approved<br />

by the local Ethics Committee.<br />

Results. Three patients (7.3%) were homozygous variant c.894TT,<br />

12 (29.3%) homozygous wild-type c.894GG and the remaining 26<br />

(63.4%) were heterozygous c.894GT. The c.894TT patients had no HT<br />

or PU at baseline and developed grade (G) 1, 2, 2 HT, respectively,<br />

and in one case PU during treatment. G1, 2 and 3 HT developed in 4, 5<br />

and 2 c.894GG subjects, respectively, while PU was observed in 7/12<br />

(58%) patients. The full range of HT grades and PU were observed<br />

in heterozygous subjects. Thirty-seven patients achieved one of the<br />

following: partial remission, minimal response or stable disease upon<br />

treatment with bevacizumab in combination with chemotherapy; 3<br />

subjects had progressive disease and 1 was not evaluable.<br />

Conclusions. The presence of the mutant T allele of c.894G>T is<br />

not associated with increased severity of HT and PU; therefore,<br />

bevacizumab can be administered at no increased risk in TT patients<br />

with respect to the wild-type GG population.<br />

P5-17-07<br />

Influence of bevacizumab on local-regional recurrence in triple<br />

negative breast cancer<br />

Prendergast BM, De Los <strong>San</strong>tos JF, Barrett OC, Forero A, Falkson<br />

CI, Urist MM, Krontiras H, Bland KI, Meredith RF, Keene KS, You<br />

Z, Carpenter JT. University of Alabama Birmingham, AL<br />

Background:<br />

Triple negative breast cancer (TNBC) comprises 15-20% of newly<br />

diagnosed invasive breast cancers and has been associated with an<br />

elevated risk of both local-regional recurrence (LRR) as well as distant<br />

failure. Bevacizumab (BEV) has been shown to improve pathologic<br />

complete response rates in the setting of neoadjuvant chemotherapy<br />

(NCT), but its effect on LRR remains undefined. This study reports<br />

the impact of adding BEV to standard breast cancer therapy in TNBC<br />

patients treated at a single institution.<br />

Methods:<br />

One hundred and eighty-five consecutive TNBC patients treated at the<br />

University of Alabama Birmingham from May 2005 to August 2010<br />

were reviewed. Thirty-one TNBC patients treated with chemotherapy<br />

(CT) and BEV on prospective studies were matched (1:2) with 62<br />

TNBC patients treated with CT alone, controlling for age, stage, and<br />

use of radiation (RT). The Kaplan-Meier method was used to estimate<br />

LRR, distant metastasis-free survival (DMFS), and overall survival<br />

(OS), and cohorts were compared using Cox proportional hazards<br />

models and the 2-sided log-rank test.<br />

Results:<br />

Mean age was 47 and 46 years in the cohorts with and without BEV,<br />

respectively. Stage in each cohort was as follows: 32% stage I, 61%<br />

stage II, and 7% stage III. Use of adjuvant RT was 81% in each cohort<br />

(25/31 BEV, 50/62 no BEV). BEV was delivered with NCT in 17<br />

patients (55%) and with adjuvant CT in the remaining 14 patients<br />

(45%). Eleven patients (35%) completed an additional 1 year of<br />

maintenance therapy with BEV. LRR occurred in 2 (6%) patients<br />

treated with BEV vs. 16 (26%) treated with CT alone (HR=0.25,<br />

95% CI 0.06-1.07, p=0.04). Distant recurrence occurred in 3 (10%)<br />

patients treated with BEV vs. 11 (18%) patients in the CT group<br />

HR=0.6, 95% CI 0.2-1.8, p=0.48). There were 2 (6%) deaths in the<br />

BEV group vs. 12 (19%) in the CT alone group (HR=0.55, 95% CI<br />

0.13-2.3, p=0.19).<br />

Conclusions:<br />

In a matched-pair analysis of TNBC patients, the addition of BEV<br />

to conventional breast cancer management was associated with a<br />

reduction in LRR. Further prospective study is necessary to examine<br />

the impact of BEV on local-regional control in TNBC.<br />

P5-18-01<br />

Pertuzumab (P) in combination with trastuzumab (T) and<br />

docetaxel (D) in elderly patients with HER2-positive metastatic<br />

breast cancer in the CLEOPATRA study<br />

Miles D, Baselga J, Amadori D, Sunpaweravong P, Semiglazov V,<br />

Knott A, Clark E, Ross G, Swain SM. Mount Vernon <strong>Cancer</strong> Centre,<br />

Middlesex, United Kingdom; Massachusetts General Hospital <strong>Cancer</strong><br />

Center and Harvard Medical School, Boston, MA; Istituto Scientifico<br />

Romagnolo per lo Studio e la Cura dei Tumori (IRST), Meldola, Italy;<br />

Songklanagarind Hospital, Prince of Songkla University, Hat Yai,<br />

Thailand; NN Petrov Research Institute of Oncology, St Petersburg,<br />

Russian Federation; Roche Products Limited, Welwyn, United<br />

Kingdom; MedStar Washington Hospital Center, Washington, DC<br />

Background: The incidence of cancer increases with age as does<br />

the risk of treatment-related adverse events (AEs) due to underlying<br />

comorbidities. A better understanding of cancer therapy-related AEs<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

464s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 5<br />

in elderly pts may help identify the optimal therapy by balancing<br />

treatment benefit and risk. CLEOPATRA, a double-blind Phase III<br />

trial, compared placebo (Pla)+T+D with P+T+D in pts with HER2-<br />

positive 1L MBC (Baselga 2012). Here we report safety and efficacy<br />

by age group.<br />

Methods: P/Pla: 840 mg initial dose, 420 mg q3w iv; T: 8 mg/kg<br />

initial dose, 6 mg/kg q3w iv; D: 75 mg/m 2 q3w iv, escalating to 100<br />

mg/m 2 if tolerated. P/Pla+T were given until progressive disease (PD)<br />

or unacceptable toxicity. At least 6 cycles of D were recommended; 6 cycles were<br />

allowed at investigators’ discretion. At baseline, pts were required<br />

to have ECOG PS of 0 or 1, LVEF ≥50% and no decline to


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

The issues related to the ITT and dependent censored analyses will be<br />

reviewed and discussed. Alternative analytic approaches designed to<br />

estimate the treatment effect that would have been observed had there<br />

been no selective crossover will be presented. The methods include<br />

the inverse probability of censoring weighted (IPCW) approach,<br />

and randomization-based estimators under the accelerated failure<br />

time model.<br />

HERA data to about 8y MFU (available fall 2012) will be used to<br />

illustrate approaches.<br />

CONCLUSION: Alternative methods addressing selective crossover<br />

are required to estimate the trastuzumab effect for updated analyses<br />

of DFS and OS for HERA, and for any other large randomized trial<br />

with positive interim results.<br />

P5-18-03<br />

Clinicopathological features among patients with HER2-positive<br />

breast cancer with prolonged response to trastuzumab based<br />

therapy<br />

Vaz-Luis I, Seah D, Olson E, Metzger O, Wagle N, Sohl J, Litsas G,<br />

Burstein H, Krop I, Winer E, Lin NU. Dana Farber <strong>Cancer</strong> Institute,<br />

Boston, MA; Ohio State University<br />

Background: HER2-positivity is a predictor of benefit from<br />

trastuzumab (TRZ), but fails to depict the observed interpatient<br />

variability in terms of treatment (tx) duration. In this study we<br />

described the relationship between clinicopathological features and<br />

TRZ tx duration.<br />

Methods: A retrospective consecutive series of 343 HER2+ breast<br />

cancer (BC) patients (pts) treated with TRZ at the Dana-Farber <strong>Cancer</strong><br />

Institute from 1999-2008 was identified. 139 pts treated with 1st line<br />

TRZ-based tx were selected for analysis. Pts who received any non-<br />

TRZ prior tx for metastatic disease were excluded. TRZ tx duration<br />

was defined as time from start of 1st line therapy to the 1st day of 2nd<br />

line therapy or death. Central nervous system (CNS) progression with<br />

TRZ maintenance was not considered change of tx. Pts were divided<br />

equally into 3 groups based on the duration of 1st line tx distribution.<br />

Short-term responders (STR) were on the 1st line tx for 15m. An additional group of extremely LTR (ELTR) was defined<br />

as being in the 90th percentile of tx duration (>37m). Descriptive<br />

analysis was performed; fisher exact test, Kruskal-Wallis and logistic<br />

regression methods were used to compare groups.<br />

Results: Median follow-up time since metastatic diagnosis was 4 years<br />

(y) (range 0-11). Median age at diagnosis was 47y (22-83), 25% of<br />

stage I-III pts at diagnosis received adjuvant/neoadjuvant TRZ. The<br />

median disease free interval (DFI) was 20m (0-172), median number<br />

of metastatic sites was 2(1-5), 68% of pts had visceral disease. Median<br />

duration of 1st line tx was 10m (2-105). TRZ was given with CT<br />

in 86%, hormone tx in 6% and as monotherapy in 9%. 25% of pts<br />

developed CNS progression and continued tx. There were only small<br />

absolute differences for clinicopathological characteristics among<br />

STR, IR and LTR.<br />

(N) STR(46) IR (46) LTR (47) p<br />

Duration of 1st line tx(median; range) 3; 2-7 10; 7-14 25; 15-105<br />

HR(n;%) 0.10<br />

Positive 18; 39 28; 61 26; 55<br />

Negative 28; 61 18; 39 21; 45<br />

Stage I-III(n;%) 37; 80 36; 78 33; 70 0.54<br />

Adjuv/neoadjuv TRZ(n;%) 12; 32 9; 25 6; 18 0.40<br />

DFI(median; range) 21; 0-172 17; 0-153 23; 0-161 0.88<br />

Number of sites of 1st<br />

2.5; 1-5 2; 1-4 2; 1-5 0.94<br />

recurrence(median; range)<br />

Sites of 1st recurrence: Visceral(n;%) 32; 70 26; 57 36; 77 0.11<br />

Type of 1st line tx(n;%) 0.10<br />

TRZ monotherapy 7; 15 1; 2 4; 9<br />

TRZ-hormone tx 1; 2 5; 11 2; 4<br />

TRZ-chemotherapy 38; 83 40; 87 41; 87<br />

Complete response(n;%) 0; 0 3; 7 2; 4 0.19<br />

CNS progression during 1st line tx;<br />

non-CNS disease controlled(n;%)<br />

3; 7 6; 13 25; 53


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 5<br />

Results: Eight patients enrolled in the phase I trial and received a<br />

median of 5 (range 1-24) cycles of therapy. All patients had received<br />

trastuzumab and a median of 5 (range 2-12) prior lines of therapy.<br />

Grade 3 diarrhea was the dose-limiting toxicity. The MTD was<br />

determined to be T at 8 mg IV weekly with neratinib at 240 mg<br />

daily. As of June 1, 2012, nineteen patients (target accrual thirty-four<br />

patients) have enrolled in the phase II trial and received a median<br />

of 2 (range 1-8) cycles of therapy. Among the 25 patients treated at<br />

the MTD (phase I n=6; phase II n=19), the most frequent treatmentrelated<br />

grade 2/3 events were diarrhea (gr 2 44%/gr 3 24%), mucositis<br />

(20%/12%), hyperglycemia (24%8%), and rash (20%/0%). Twenty<br />

patients treated at the MTD are evaluable for response; 11 patients<br />

had PR (8 confirmed PRs) and 1 had SD≥6 months for a RR of<br />

40%. Molecular analyses revealed a set of patients with more than<br />

1 mutation in the HER2-PI3K-AKT-mTOR pathway had prolonged<br />

responses to T-N therapy.<br />

Conclusions: Temsirolimus and neratinib is a tolerable regimen that<br />

has clinical activity in trastuzumab-refractory HER2+ MBC patients.<br />

The phase II study is ongoing and additional efficacy, safety, and<br />

biomarker data will be presented.<br />

P5-18-05<br />

Interim Results from a Phase 1b/2a Study of Trastuzumab<br />

Emtansine and Docetaxel, With and Without Pertuzumab, in<br />

Patients With HER2-Positive Locally Advanced or Metastatic<br />

<strong>Breast</strong> <strong>Cancer</strong><br />

Martin M, García-Sáenz JÁ, Dewar JA, Albanell J, Limentani<br />

SA, Strasak A, Patre M, Branle F, Fumoleau P. Hospital General<br />

Universitario Gregorio Marañón; Hospital <strong>San</strong> Carlos; Ninewells<br />

Hospital; Hospital del Mar; Levine <strong>Cancer</strong> Institute; F. Hoffmann-La<br />

Roche Limited; Centre Georges François Leclerc<br />

Introduction<br />

Trastuzumab emtansine (T-DM1) is an antibody-drug conjugate<br />

comprising trastuzumab, a stable linker, and DM1 (a microtubule<br />

inhibitor). In 2 randomized trials in patients (pts) with HER2-positive<br />

metastatic breast cancer (MBC), T-DM1 significantly prolonged<br />

progression-free survival (PFS) compared with standard therapy<br />

(Hurvitz, ESMO 2011; Blackwell, ASCO 2012). The combination<br />

of T-DM1 and docetaxel (D) or pertuzumab (P) has shown enhanced<br />

antitumor activity in preclinical models of HER2-positive BC (Lewis<br />

Phillips, AACR 2008; Fields, AACR 2010).<br />

Methods<br />

This phase 1b/2a dose-escalation study examines the safety,<br />

tolerability, efficacy, and pharmacokinetics (PK) of T-DM1 (starting<br />

dose 2.4 mg/kg every 3 weeks [q3w]) combined with D (starting<br />

dose 75 mg/m 2 q3w) in pts with MBC, and T-DM1 with D±P (840<br />

mg loading dose, then 420 mg q3w) in pts with newly diagnosed<br />

locally advanced BC (LABC; clinical stage IIa-IIIc). The maximum<br />

tolerated dose (MTD) for T-DM1+D is established in pts with MBC<br />

and used as the starting dose for feasibility of T-DM1+D±P (up to 6<br />

cycles) in pts with LABC. Eligibility criteria include HER2-positive<br />

MBC (locally assessed) or LABC (centrally confirmed). This interim<br />

report includes data from pts who completed at least 1 treatment<br />

cycle in phase 1b.<br />

Results<br />

During MBC feasibility, 21 pts were enrolled; median age was 47.0<br />

years (yr; range, 33–70); median number of prior lines of MBC<br />

therapy was 2 (range, 0–10). Four pts experienced 6 DLTs (Table 1).<br />

The MTD was T-DM1 3.6 mg/kg q3w + D 60 mg/m 2 q3w. Common<br />

grade (G) 3/4 adverse events (AEs): neutropenia (81%), leukopenia<br />

(52%), and thrombocytopenia (29%). ORR was 76.2% (16/21);<br />

median PFS was 11.8 months (range, 1.6–26.9). In LABC feasibility,<br />

18 pts were enrolled; median age was 49.5 yr (range, 34–74). Four<br />

pts experienced 4 DLTs (Table 2). The MTD for T-DM1+D was 3.6<br />

mg/kg q3w + 75 mg/m 2 q3w; MTD for T-DM1+D+P was 3.6 mg/kg<br />

q3w + 75 mg/m 2 q3w + 840 mg (with G-CSF primary prophylaxis).<br />

Common G3/4 AEs: T-DM1+D, neutropenia (2/9; 22%), increased<br />

alanine aminotransferase (ALT; 2/9; 22%), thrombocytopenia (TCP;<br />

1/9; 11%); T-DM1+D+P, neutropenia (5/9; 56%), TCP (2/9; 22%),<br />

increased ALT (2/9; 22%), and cytolytic hepatitis (1/9; 11%). With<br />

9 pts enrolled in the T-DM1+D LABC cohorts and 8 pts currently<br />

evaluable, pCR rate (ypT0N0) is 87.5%.<br />

Table 1. MBC Feasibility<br />

Cohort 1 (n=6) Cohort 2 (n=6) Cohort 3 (n=3) Cohort 4 (n=6)<br />

T-DM1 dose, mg/kg 2.4 (day 2) 2.4 (day 2) 2.4 (day 1) 3.6 (day 1)<br />

Docetaxel dose, mg/m 2 75 (day 1) 60 (day 1) 60 (day 1) 60 (day 1)<br />

DLTs<br />

G3 hepatitis (n=2); G3<br />

febrile neutropenia; G3 hepatitis<br />

G4 TCP<br />

None<br />

G3 increased<br />

aspartate<br />

aminotransferase<br />

Table 2. LABC Feasibility<br />

Cohort 1<br />

(n=3)<br />

Cohort 2<br />

(n=3)<br />

Cohort 3 (n=3) Cohort 4 (n=3) Cohort 5 (n=6)<br />

T-DM1 dose, mg/kg 3.6 3.6 3.6 3.6 3.6<br />

Docetaxel dose, mg/m 2 60 75 100 60 75<br />

Pertuzumab dose, mg — — — 840 840<br />

DLTs None None<br />

G4 TCP; G3<br />

G4 TCP; G4<br />

mucosal None<br />

neutropenia<br />

inflammation<br />

Conclusions<br />

Full-dose T-DM1 can be safely combined with D in pts with MBC,<br />

and with D±P in pts with LABC. G-CSF is needed with higher doses<br />

of D. Pathological assessment for all LABC patients and PK analyses<br />

will be available for the full presentation.<br />

P5-18-06<br />

Trastuzumab emtansine in HER2-positive metastatic breast<br />

cancer: pooled safety analysis from seven studies<br />

Diéras V, Harbeck N, Budd GT, Greenson JK, Guardino E, Samant<br />

M, Chernyukhin N, Smitt M, Krop IE. Institut Curie; <strong>Breast</strong> Center,<br />

University of Munich; Cleveland Clinic, Lerner College of Medicine;<br />

University of Michigan; Genentech, Inc.; Dana-Farber <strong>Cancer</strong><br />

Institute<br />

Background<br />

The antibody-drug conjugate (ADC) trastuzumab emtansine (T-DM1)<br />

combines the antitumor activities of trastuzumab with intracellular<br />

delivery of the cytotoxic agent DM1 and represents a new approach<br />

to therapy for HER2-positive MBC. Recently, the first phase 3 data<br />

have been reported with significant improvements in PFS (9.6 vs<br />

6.4 months; HR=0.650; P


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Results<br />

Among all pts (N=882), median age was 53 years (range 25–85 years);<br />

52.7% had ≥3 metastatic sites; 81.7% had received prior treatment<br />

for MBC; and the median number of prior non-endocrine agents for<br />

MBC was 3.0 (range 0–19). All-grade AEs occurring in ≥25% of<br />

patients were fatigue (45.4%), nausea (42.3%), headache (28.7%),<br />

thrombocytopenia (28.7%), and constipation (25.5%). Grade ≥3 AEs<br />

occurring in ≥2% were thrombocytopenia, increased AST, increased<br />

ALT, fatigue, hypokalemia, and anemia. Table 1 describes the grade<br />

≥3 incidence of these AEs, as well as selected AEs with known<br />

association to T-DM1 or potential risk based on the components of<br />

T-DM1 or on preclinical data.<br />

Table 1: Incidence of grade ≥3 adverse events by NCI CTCAE, v 3.0 in T-DM1-treated patients<br />

EMILIA (prior<br />

treatment with<br />

taxane,<br />

All T-DM1<br />

exposed patients<br />

(N=882)<br />

Single-arm trials<br />

(prior MBC<br />

treatment)<br />

(n=288)<br />

TDM4450g<br />

(no prior MBC<br />

treatment) (n=69)<br />

trastuzumab)<br />

(n=490)<br />

Adverse event Grade ≥3, % Grade ≥3, % Grade ≥3, % Grade ≥3, %<br />

Thrombocytopenia 10.2 7.6 7.2 12.9<br />

Increased AST 4.1 3.1 8.7 4.3<br />

Fatigue 3.2 3.8 4.3 2.4<br />

Hypokalemia 2.9 4.2 2.9 2.2<br />

Increased ALT 2.8 1.4 10.1 2.9<br />

Anemia 2.5 2.4 2.9 2.7<br />

Peripheral neuropathy 1.2 1.0 0 1.6<br />

Nausea 1.0 1.0 2.9 0.8<br />

Constipation 0.6 0.3 1.4 0.4<br />

Headache 0.6 0.3 0 0.8<br />

Epistaxis 0.5 1.0 0 0.2<br />

Cardiac dysfunction a 0.2 0.3 1.4 0<br />

Eye disorders a 0.1 0.3 0 0<br />

Nodular regenerative<br />

hyperplasia of the liver<br />

0.2 0.3 0 0.2<br />

Pneumonitis a 0.2 0.3 1.4 0<br />

Infusion-related reactions 0.1 0.3 0 0<br />

a<br />

AE basket defined based on MedDRA SMQ.<br />

Conclusions<br />

The safety profile of T-DM1 3.6 mg/kg q3w was consistent across<br />

the studies and the different patient populations with HER2-positive<br />

MBC. Additional analyses on hepatic transaminase increases and<br />

those exploring potential associations between thrombocytopenia<br />

and hemorrhage will be presented.<br />

P5-18-07<br />

Completed SN33 Trial: 60 month follow-up of Early Stage Node<br />

Positive HER2 Negative patients with NeuVax (E75) and<br />

sargramostim.<br />

Mazanet R, Schwartz MW, Ramadan D, Lavin PT. Galena Biopharma,<br />

Lake Oswego, OR; Aptiv Solutions, Southborough, MA<br />

Background: Despite recent advances in adjuvant treatment, the<br />

prognosis for HER2 negative patients has not improved. Unlike<br />

HER2 positive patients, the benefit of Herceptin has not been shown.<br />

Here we present the final 60-month results of a phase 1/I2 clinical<br />

trial vaccinating early stage node positive breast cancer patients with<br />

NeuVax (E75) and sargramostim following adjuvant chemotherapy<br />

and radiation. Previously, data reported from this trial was combined<br />

with a node negative trial that used the same treatment schema. Those<br />

reports showed the vaccine was safe, and effective at stimulating<br />

HER2-specific immunity. We report safety and efficacy (Disease Free<br />

Survival, DFS) data through 60 months. Methods: Eligible patients<br />

had excised early stage node-positive breast cancer with any level of<br />

HER2 expression. After completion of standard of care chemotherapy<br />

and radiation, patients were enrolled but not randomized: patients<br />

were assigned to the vaccine group (VG) only if they were HLA<br />

A2/A3+. The non-treated patients were followed as a control group<br />

(CG). Hormone receptor positive patients received appropriate<br />

adjuvant therapy concomitant with vaccine. The phase I/II trial was<br />

performed with phase 1 as a dose escalation/schedule optimization<br />

where VG was given 3-6 monthly inoculations of NeuVax (E75) with<br />

the immunoadjuvant, sargramostim. The phase 2 portion utilized<br />

the optimal dose and schedule carried forward. Herceptin became<br />

commercially available during the conduct of the trial, and HER2<br />

positive patients were offered this treatment but remained on trial.<br />

DFS was summarized by Kaplan-Meier lifetables and analyzed<br />

by the log-rank test. Due to waning immunity after the scheduled<br />

monthly vaccinations, a voluntary booster program was initiated to<br />

be integrated into the trial process. Results: 97 patients were enrolled;<br />

53 in the VG and 44 in the CG. The VG and CG were well-matched<br />

with the only difference being trastuzumab therapy (18.5% in VG<br />

vs. 4.8% in CG, p=0.03). There were no HER2 0 patients in the VG.<br />

Vaccination was well tolerated with primarily grade 1 and grade 2<br />

toxicity. No VG patients relapsed during the vaccination period. ITT<br />

patients in this phase 1/2 trial favored VG in DFS at 24 months (VG<br />

90.6%, CG 79.5%, p=0.1194); at 60 months, the VG continues to have<br />

fewer recurrences than the CG (VG 84.5% vs. 77.1%, p=0.3046).<br />

In the patients who were treated at the optimum (Phase 2) dose, the<br />

difference in the DFS at 24 months observed was (VG 96.2%, CG<br />

79.5%, p=0.0597); at 60 months, the VG continues to have fewer<br />

recurrences than the CG (92.3% vs. 77.1%). When the HER2 positive<br />

patients were removed from the population, the Phase 2 HER2<br />

Negative VG had a significantly improved DFS at 24 months (100%<br />

vs. 80.6%, p=0.045); at 60 months, VG retains a 17% difference (VG<br />

94.7% vs. CG 77.4%). Conclusions: NeuVax (E75) and sargramostim<br />

vaccine is safe and well–tolerated in early stage breast cancer patients.<br />

With follow-up at 60 months, the vaccine used as adjuvant therapy<br />

continues to reduce relapses in node positive HER2 negative patients<br />

treated at an optimal dose. A phase 3 multicenter, multinational<br />

double-blind trial is underway.<br />

P5-18-08<br />

Identification of ErbB2 function in the heart: implication for<br />

anti-ErbB2 therapy in breast cancer<br />

Perry M-C, Eichner LJ, Dufour CR, Muller WJ, Giguère V. Rosalind<br />

and Morris Goodman <strong>Cancer</strong> Research Centre, McGill University,<br />

Montréal, QC, Canada; McGill University, Montréal, QC, Canada<br />

The ErbB2 receptor tyrosine kinase is essential for cardiac<br />

development during embryogenesis. The importance of its signaling in<br />

the adult heart was revealed by an unexpected cardiotoxic side effect<br />

of trastuzumab, a monoclonal antibody against ErbB2 used in the<br />

treatment of breast cancer. Considering that trastuzumab-associated<br />

cardiotoxicity is usually largely reversible, we hypothesized that<br />

ErbB2 signaling in the heart is important to maintain cardiac<br />

homeostasis.<br />

As genetic experiments showed that ErbB2-deficient embryos die at<br />

mid-gestation, whereas conditional inactivation of ErbB2 in the heart<br />

leads to early and severe cardiac dysfunction, the role of ErbB2 in<br />

the mature heart can not be analyzed in those animal models. Thus,<br />

we chose an ErbB2 hypomorph (HP) mouse model in which ErbB2<br />

is expressed at only 10% of its endogenous level to study the role<br />

of ErbB2 in the adult heart. Histopathological analyses of the heart<br />

in this model revealed that at birth, the ErbB2 HP mice have similar<br />

heart mass and cardiomyocite size as the control animals. However,<br />

we found that the rapid growth of the heart during early postnatal<br />

development is impaired in ErbB2 HP mice, indicating that the heart<br />

is unable to increase cell size in order to adapt to the pressure overload<br />

that mice face following birth. This incapacity of the ErbB2 HP heart<br />

to maintain homeostasis eventually leads to the development of<br />

cardiac dysfunction in this model, as characterized by a decrease of<br />

the left ventricular function. Microarray and ChIP-seq analyses were<br />

applied to identify the ErbB2-responsive pathways which may explain<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

468s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 5<br />

the phenotype observed. Taken together, our results demonstrate that<br />

ErbB2 signaling is required for the physiological adaptive response<br />

of the mature heart to pressure burden. The results generated by this<br />

study reveal the biological function of Erbb2 in the adult heart, but<br />

also provide important information for devising strategies to prevent<br />

and mitigate the cardiotoxic effects of Trastuzumab treatment,<br />

allowing for the potential use of this drug to treat all cancer patients<br />

overexpressing ErbB2.<br />

P5-18-09<br />

A Phase I Study of MM-302, a HER2-targeted Liposomal<br />

Doxorubicin, in Patients with Advanced, HER@- Positive <strong>Breast</strong><br />

<strong>Cancer</strong><br />

Wickham T, Futch K. Merrimack Pharmmaceuticals; Merrimack<br />

Pharmaceuticals<br />

Background: Anthracyclines have been an effective backbone of<br />

breast cancer therapies for decades. However, cardiotoxicity issues<br />

associated with free anthracyclines have limited their effective<br />

use. While liposomal doxorubicin formulations have succeeded in<br />

reducing cardiotoxicity, they have failed to demonstrate clear-cut<br />

efficacy advantages. To address these safety and efficacy limitations,<br />

we have designed a pegylated liposomal doxorubicin formulation,<br />

MM-302, with anti-HER2 antibody fragments coupled to its surface.<br />

MM-302 specifically targets tumor cells overexpressing HER2 with<br />

minimal uptake into normal cells such as cardiomyocytes which<br />

express low levels of HER2. This first-in-human Phase 1 study<br />

evaluates the safety of MM-302 in patients with HER2+ advanced<br />

breast cancer (ABC). Methods: Patients aged > 18 years with<br />

histologically confirmed HER2+ advanced breast cancer that have<br />

progressed or recurred on standard therapy or for which no standard<br />

therapy exists who have adequate performance status, bone marrow<br />

reserve, and organ function, were eligible for the study. A standard 3 +<br />

3 dose escalation design is used to determine the maximum tolerated<br />

dose (MTD) followed by expansion arms to evaluate activity. The<br />

primary endpoint is determination of the MTD. Secondary endpoints<br />

included determination of dose-limiting toxicity, adverse event(s),<br />

and pharmacokinetic and immunogenicity profiles of MM-302, as<br />

well as overall response and clinical benefit rates of MM-302. MM-<br />

302 is administered intravenously weekly in 4-week cycles. Results:<br />

We report current findings containing data from four cohorts (8, 16,<br />

30 and 40 mg/m2 Q4W). 14 patients have been enrolled in these<br />

cohorts; median age 55 (range 34-65), median cycles 2 (range 1-6).<br />

The most frequent AEs were constipation (53.8%), Fatigue (69.2%)<br />

and decreased appetite (46.2%). Five patients have achieved SD and<br />

one patient has achieved a PR. A pharmacokinetic analysis indicates<br />

a half-life of approximately 50 hours. Conclusions: To date, results<br />

suggest that single agent MM-302 administered to patients with ABC<br />

is tolerable, without cardiotoxicity, in patients up to 40 mg/m2 Q4W.<br />

Updated data on additional dosing cohorts and expansion arms will<br />

be presented.<br />

P5-18-10<br />

Chemotherapy can enhance trastuzumab-mediated ADCC<br />

Tagliabue E, Sfondrini L, Regondi V, Casalini P, Pupa SM, Sommariva<br />

M, Balsari A, Triulzi T. Fondazione IRCCS Istituto Nazionale dei<br />

Tumori; Università degli Studi di Milano<br />

Trastuzumab, a recombinant humanized monoclonal antibody<br />

directed to the HER2 protein, has shown survival benefits in<br />

women with HER2-positive breast cancer, and treatment is now<br />

FDA-approved in combination with chemotherapy. However, some<br />

patients do not respond clinically to trastuzumab, pointing to the need<br />

for further definition of trastuzumab killing activity on tumor cells<br />

to optimize this therapy. Recent clinical data indicate a synergistic<br />

therapeutic effect between trastuzumab and taxanes in HER2-positive<br />

breast cancer patients, but the mechanism(s) underlying this synergy<br />

remains unclear. Because drug-stressed cells can dynamically<br />

regulate factors that favor activity of immune effector cells, and<br />

because trastuzumab-mediated ADCC is crucially dependent on<br />

NK cell activity, we hypothesize that this synergy reflects enhanced<br />

trastuzumab-mediated ADCC on tumor cells due to improved rather<br />

than impaired participation of immune effectors resulting from druginduced<br />

stress. To test this hypothesis, we investigated the effect of<br />

chemotherapy in HER2-positive breast carcinoma models on NK<br />

receptor ligands and correlated the changes in these molecules with<br />

the ability of trastuzumab to mediate ADCC.<br />

Flow cytometry revealed a 4-fold upmodulated surface expression of<br />

NKG2D (MICA, MICB, ULBP1, ULBP2) and DNAM-1 (CD112 and<br />

CD155) ligands in HER2-positive breast carcinoma cell lines BT474<br />

and MDA-MB-361 treated for 6 hr with taxotere at a 72-hr IC 50 dose,<br />

consistent with results of Western blot analysis using soluble lysates<br />

obtained from chemotherapy-treated HER2-positive breast carcinoma<br />

xenografts. The enhanced expression of NKG2D and DNAM-1<br />

ligands in breast carcinoma cells was accompanied by about a 50%<br />

increase in trastuzumab-mediated in vitro ADCC in chemotherapytreated<br />

versus untreated cells. Antibodies blocking NKG2D and<br />

DNAM-1, but not control monoclonal antibody, abolished the<br />

chemotherapy-induced increase of in vitro trastuzumab-dependent<br />

ADCC. Increased expression of NK ligands in taxotere-treated BT474<br />

cells was associated with cytoskeletal damage assessed by confocal<br />

microscopy; at 72 hr after chemotherapy, NK ligands returned to<br />

basal levels, pointing to the importance of duration of NK ligand<br />

enhancement for the chemotherapy-induced trastuzumab-mediated<br />

ADCC.<br />

Together, our results indicate that chemotherapy can modulate<br />

expression levels of NK-activating ligands on human breast carcinoma<br />

cells in correlation with trastuzumab-mediated ADCC, raising the<br />

possibility of identifying optimal chemotherapy administration<br />

schedules to maximize activity of trastuzumab-mediated ADCC<br />

effectors. (Partially supported by AIRC)<br />

P5-18-11<br />

Pharmacokinetics and exposure-efficacy relationship of<br />

trastuzumab emtansine in EMILIA, a phase 3 study of<br />

trastuzumab emtansine vs capecitabine and lapatinib in HER2-<br />

positive locally advanced or metastatic breast cancer<br />

Wang B, Jin J, Wada R, Fang L, Saad O, Olsen S, Althaus B, Swain<br />

S, Untch M, Girish S. Genentech, Inc.; Quantitative Solutions;<br />

Washington Hospital Center; HELIOS Hospital Berlin-Buch<br />

Background<br />

Trastuzumab emtansine (T-DM1) is an antibody-drug conjugate<br />

composed of trastuzumab (T), the cytotoxic agent DM1, and a<br />

stable thioether linker; it is being evaluated for the treatment (tx) of<br />

HER2-positive cancer. EMILIA is a randomized, multicenter, openlabel<br />

phase 3 study assessing the efficacy and safety of T-DM1 vs<br />

capecitabine (X) and lapatinib (L) in pts with HER2-positive locally<br />

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

T and a taxane. We report the pharmacokinetics (PK) of T-DM1 in<br />

EMILIA and compare these data with historical data of single-agent<br />

T-DM1. The effects of T-DM1 exposure on efficacy outcomes in<br />

EMILIA are also reported.<br />

www.aacrjournals.org 469s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Methods<br />

In EMILIA, pts were randomized 1:1 to receive T-DM1 3.6 mg/kg<br />

q3w (n=495) or X + L (n=496) in 21-day cycles. At least 1 PK sample<br />

was collected from 350 pts in the T-DM1 arm; 307 of these had<br />

adequate PK profiles for the estimation of ≥1 PK parameter in cycle<br />

1. PK parameters were estimated by noncompartmental analysis for<br />

serum T-DM1, serum total T (conjugated and unconjugated T), and<br />

plasma DM1. A logistic regression model was used to evaluate the<br />

relationship between T-DM1 exposure (ie, T-DM1 AUC and DM1<br />

C max ) and objective response rates (ORR). Analyses of progressionfree<br />

survival (PFS) and overall survival (OS), stratified by T-DM1<br />

exposure, were conducted. Cox regression was used to evaluate the<br />

effect of T-DM1 exposure and other potential covariates on PFS<br />

and OS.<br />

Results<br />

The PK parameters for T-DM1, total T, and DM1 in EMILIA and<br />

other studies of single-agent T-DM1 are summarized in Table 1.<br />

The PK parameters appear similar regardless of prior tx for MBC.<br />

No significant differences in mean serum T-DM1 AUC (P=0.091)<br />

or plasma DM1 C max (P=0.812) were observed between responders<br />

and non-responders following T-DM1 tx. Furthermore, variability in<br />

T-DM1 AUC and DM1 C max does not appear to affect the probability<br />

of response to therapy (P=0.23 for T-DM1 AUC and P=0.74 for DM1<br />

C max ). Based on Cox proportional hazards analysis, no significant<br />

exposure-efficacy relationship was observed between T-DM1 AUC<br />

(P=0.23 for PFS and P=0.53 for OS) or DM1 C max (P=0.96 for PFS<br />

and P=0.34 for OS) and PFS or OS, respectively.<br />

Table 1. PK parameters of single-agent T-DM1<br />

T-DM1 T-DM1 T-DM1 T-DM1 T-DM1 Total T DM1<br />

Mean (SD)<br />

AUC ,<br />

Study<br />

inf<br />

CL, mL/<br />

C max , µg/mL day •µg/ t ½ , day V ss , mL/kg<br />

day/kg<br />

mL<br />

C max , µg/mL C max , ng/mL<br />

EMILIA (n=307) 83.4 (16.5) 489 (122) 3.68 (0.886) 29.5 (14.6) 7.81 (2.18) 86.3 (20.1) 4.61 (1.61)<br />

TDM4450g (no prior<br />

MBC tx) (n=64)<br />

84.9 (30.6) 476 (119) 3.58 (0.666) 30.0 (8.21) 8.09 (2.50) 81.5 (20.6) 5.08 (2.41)<br />

TDM4258g (prior T for<br />

MBC) (n=101)<br />

80.9 (20.7) 457 (129) 3.53 (0.714) 28.4 (12.9) 8.51 (2.69) 88.0 (30.2) 5.35 (2.03)<br />

TDM4374g (prior T and<br />

L for MBC) (n=105)<br />

79.5 (21.1) 486 (141) 3.96 (0.964) 31.2 (10.9) 8.04 (2.97) 89.9 (31.3) 5.36 (2.56)<br />

Conclusions<br />

T-DM1 PK data in EMILIA pts were similar to those observed in<br />

previous phase 2 studies, which include pts who have and have not<br />

received prior therapy for MBC. Variation in T-DM1 exposure among<br />

pts who received T-DM1 3.6 mg/kg q3w had no significant impact on<br />

ORR, PFS, or OS. Detailed analyses will be presented.<br />

P5-18-12<br />

Comparison of treatment patterns and outcomes in metastatic<br />

breast cancer patients initiated on trastuzumab vs. lapatinib; a<br />

retrospective analysis<br />

Gauthier G, Guérin A, Styles AL, Wu EQ, Masaquel A, Brammer<br />

MG, Lalla D. Analysis Group Inc., Boston, MA; Genentech, Inc.,<br />

South <strong>San</strong> Francisco, CA<br />

Background: Trastuzumab-or lapatinib-based therapies are<br />

recommended by the National Comprehensive <strong>Cancer</strong> Network<br />

(NCCN) guidelines as preferred agents for metastatic HER2-positive<br />

breast cancer. Few studies have compared treatment patterns,<br />

healthcare resource utilization (HRU), and costs in metastatic breast<br />

cancer (MBC) patients receiving trastuzumab vs. lapatinib. The<br />

objective of this study was to compare discontinuation rates, HRU,<br />

and costs in MBC patients (pts) initiated on trastuzumab vs. lapatinib.<br />

Methods: MBC adult women initiated on trastuzumab or lapatinib on/<br />

after 03/13/2007 (lapatinib FDA-approval date) were selected from the<br />

US-based PharMetrics® Integrated Database (2000-2011). Selected<br />

pts were required to be continuously eligible in their healthcare plan<br />

≥6 months prior to and ≥30 days following trastuzumab or lapatinib<br />

initiation date. Pts were observed over the period spanning from<br />

trastuzumab or lapatinib initiation date (without restriction on line<br />

of therapy) up to the end of continuous health plan enrollment or<br />

data availability, whichever occurred first. Trastuzumab or lapatinib<br />

discontinuation rates (gap ≥45 consecutive days) were compared<br />

using multivariate Cox proportional-hazards models and reported<br />

as hazard ratios (HRs). Incremental HRU was estimated using<br />

negative binomial regression models and reported as incidence rate<br />

ratios (IRRs). Monthly cost difference (2010 USD; measured from<br />

a payer perspective) were estimated using multivariate generalized<br />

linear or two-part models. Since a large proportion of lapatinib users<br />

were previously treated with trastuzumab, a sensitivity analysis was<br />

conducted to control for the impact of prior trastuzumab use.<br />

Results: Among the 643 pts selected, 381 and 262 pts were initiated<br />

on trastuzumab and lapatinib, respectively. Of the 262 pts initiated<br />

on lapatinib, 171 (65%) were previously treated with trastuzumab.<br />

After adjustment for confounders (demographics, index year, prior<br />

MBC therapies, comorbidities, baseline HRU, and MBC duration),<br />

compared to trastuzumab users, lapatinib users had a higher rate of<br />

treatment discontinuation (HR=1.57; p


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 5<br />

(n=10), capecitabine (n=8), T-DM1 (n=6), and pertuzumab (n=1).<br />

Therapy was well tolerated; no treatment related grade 4 toxicities<br />

were observed. Myelosuppression was limited to one pt with Grade<br />

2 anemia and one each with Grade 2/3 thrombocytopenia. MTD was<br />

not reached. There were no objective responses; 2 pts. in cohort 3<br />

had SD. Serial tumor biopsies and bone marrow aspirates have been<br />

analyzed for 7 pts (cohort 1: 001-004; cohort 2: 005-007; cohort 3:<br />

analysis ongoing). Tumor hTERT (p=ns) and phosphorylated HER2<br />

(p=0.03) decreased after treatment in nearly all pts. Bone marrow<br />

S-phase did not change consistently with treatment.<br />

Relative pHER2<br />

Relative hTERT<br />

Patient Baseline Pre-cycle 2 Baseline Pre-cycle 2<br />

001 0.109 0.089 0.380 0.867<br />

002* 0 0 0.924 0<br />

003 0.302 0.131 0.574 0.366<br />

004 0.290 0,158 0.224 0.014<br />

005 1.709 0.418 0.806 0.583<br />

006 0.890 0.244 1.017 0.314<br />

007 0.945 0.388 0.253 0.191<br />

*Her2+ at diagnosis but baseline sample at study entry HER2-.<br />

Conclusion: The combination of trastuzumab + imetelstat is well<br />

tolerated and results in decreases in HER2 phosphorylation similar<br />

to that seen in preclinical models. Additional biologic correlates and<br />

PK analyses are ongoing. Further study of this combination in less<br />

heavily pre-treated patients is planned.<br />

P5-18-14<br />

Cardiac monitoring during adjuvant trastuzumab therapy for<br />

breast cancer<br />

Ng D, Ferrusi I, Khong H, Earle C, Trudeau M, Marshall D, Leighl N.<br />

University of Toronto, ON, Canada; McMaster University, Hamilton,<br />

ON, Canada; University of Calgary, AB, Canada; Ontario Institute<br />

for <strong>Cancer</strong> Research, Toronto, ON, Canada<br />

Background: Adjuvant trastuzumab improves survival in HER2+<br />

breast cancer. Cardiac toxicity is an important potential side effect.<br />

We report real world practice patterns in cardiac monitoring and<br />

outcomes during adjuvant trastuzumab therapy in Ontario. Methods:<br />

A cohort of female patients diagnosed with early breast cancer from<br />

Jan 1, 2006 to Dec 31, 2007 and who received trastuzumab was<br />

identified retrospectively through linkage of provincial administrative<br />

and registry databases. Demographic, pathology, treatment, hospital<br />

admissions, claims for cardiac tests (MUGA or echo), and outcomes<br />

were extracted for individuals. Pre-existing cardiac disease (CHF,<br />

MI, angina, valve disorder, arrhythmia, cardiomyopathy) and risk<br />

factors (diabetes, lipid disorder, hypertension) were classified using<br />

ICD-10 codes. Appropriate cardiac monitoring definitions were<br />

based on published trials and expert opinion. Symptomatic cardiac<br />

toxicity was defined as a physician claim or hospital admission with<br />

a cardiac diagnosis occurring within 2 years of the first trastuzumab<br />

dose. Asymptomatic cardiac toxicity was defined as temporary or<br />

permanent cessation of trastuzumab with additional cardiac tests.<br />

Patients were categorized into treatment groups: G1 received at<br />

least 17 doses (standard 3-weekly administration for 51 weeks)<br />

with no complications; G2 stopped early for non-cardiac reasons<br />

(no additional cardiac tests); G3 had symptomatic or asymptomatic<br />

cardiac toxicity. Analyses of patient, treatment and system factors<br />

possibly affecting cardiac monitoring and toxicity were performed.<br />

Results: 1,357 patients diagnosed with early breast cancer between<br />

2006-7 received trastuzumab (median=18 doses). 77% received<br />

anthracyclines; 4.1% had at least 1 cardiac risk factor. The majority<br />

(91%) had a baseline cardiac test, including 96% of those with<br />

cardiac risks. Geographic region was associated with baseline testing,<br />

but multivariable analysis of other factors including urbanicity,<br />

left lateral radiation, age, income, and anthracycline use, did not<br />

explain the variation in baseline testing patterns. The majority,<br />

81%, had ≥3 cardiac tests, 62.2% had ≥4. Cardiac monitoring was<br />

deemed appropriate in 80.7% of patients without cardiac events, and<br />

73.4% in those with symptomatic or asymptomatic cardiac events.<br />

Multivariable analysis revealed duration of trastuzumab to be the<br />

most significant factor associated with appropriateness of cardiac<br />

monitoring (G1 - OR 0.62, p=0.018, 95% CI 0.45-0.92; G3 - OR<br />

4.56, p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P5-18-16<br />

A Multicenter Phase 2 Study (JO22997) Evaluating the Efficacy<br />

and Safety of Trastuzumab Emtansine in Japanese Patients With<br />

Heavily Pretreated HER2-Positive Metastatic <strong>Breast</strong> <strong>Cancer</strong><br />

Masuda N, Ito Y, Takao S, Doihara H, Rai Y, Horiguchi J, Kohno<br />

N, Fujiwara Y, Tokuda Y, Watanabe J, Iwata H, Ishiguro H, Miyoshi<br />

Y, Matsubara M, Kashiwaba M. NHO Osaka National Hospital,<br />

Osaka, Japan; The <strong>Cancer</strong> Institute Hospital of JFCR, Tokyo, Japan;<br />

Hyogo <strong>Cancer</strong> Center, Akashi, Japan; Okayama University Hospital,<br />

Okayama, Japan; Sagara Hospital, Kagoshima, Japan; Gunma<br />

University, Maebashi, Japan; Tokyo Medical University, Tokyo,<br />

Japan; National <strong>Cancer</strong> Center Central Hospital, Tokyo, Japan;<br />

Tokai University, Isehara, Japan; Shizuoka <strong>Cancer</strong> Center, Sunto-gun,<br />

Japan; Aichi <strong>Cancer</strong> Center, Nagoya, Japan; Kyoto University, Kyoto,<br />

Japan; Hyogo College of Medicine, Nishinomiya, Japan; Chugai<br />

Pharmaceutical Co.,Ltd., Tokyo, Japan; Iwate Medical University,<br />

Morioka, Japan<br />

Background: Trastuzumab emtansine (T-DM1) is an anti-human<br />

epidermal growth factor receptor 2 (HER2) antibody-drug conjugate<br />

in development for the treatment of HER2-positive recurrent, locally<br />

advanced or metastatic breast cancer (MBC). T-DM1 comprises<br />

trastuzumab, DM1—a microtubule inhibitory cytotoxic agent derived<br />

from maytansine—and the stable linker ([N-maleimidomethyl]<br />

cyclohexane-1-carboxylate) that conjugates DM1 and trastuzumab. A<br />

phase 1 study in Japanese patients determined a maximum tolerated<br />

dose for T-DM1 of 3.6 mg/kg every 3 weeks (q3w), which was<br />

identical to that reported in Western populations (TDM3569g). Singleagent<br />

T-DM1 has demonstrated robust clinical efficacy in Western<br />

phase 2 and phase 3 clinical studies (TDM4258g, TDM4374g and<br />

EMILIA). Since the efficacy of T-DM1 in Japanese patients has not<br />

previously been investigated, this study examined the efficacy and<br />

safety of T-DM1 in Japanese patients with pretreated HER2-positive<br />

MBC.<br />

Methods: JO22997 is a phase 2, multicenter, single arm clinical<br />

study assessing the efficacy and safety of single-agent T-DM1<br />

given at 3.6 mg/kg q3w to patients with HER2-positive MBC. Key<br />

eligibility criteria were prior treatment with trastuzumab and at least<br />

1 chemotherapy, ECOG performance status (PS) of ≤2, and adequate<br />

organ function. Patients were required to have target lesions according<br />

to the guidelines of the Response Evaluation Criteria In Solid Tumors,<br />

v1.1. The primary objective of the study was to assess the objective<br />

response rate (ORR) by independent review committee (IRC);<br />

secondary objectives were progression-free survival (PFS), safety,<br />

and to obtain pharmacokinetic data in Japanese patients.<br />

Results: 73 patients received T-DM1. Median age was 58 years<br />

(range, 36–82 years); 61 (83.6%), 10 (13.7%), and 2 (2.7%) patients<br />

had ECOG PS of 0, 1, and 2, respectively; 39 (53.4%) patients had<br />

tumors that were estrogen receptor– and/or progesterone receptor–<br />

positive. The median number of prior chemotherapy regimens for<br />

MBC was 3 (range, 1–8) including lapatinib in 43 (58.9%) patients.<br />

Median duration of treatment with T-DM1 was 23.1 weeks (range,<br />

0.1–63.3 weeks). The ORR by IRC was 38.4% (90% confidence<br />

interval [CI], 28.8%–48.6%; partial response only), and clinical<br />

benefit rate (partial response + stable disease ≥24 weeks) was 45.2%<br />

(95% CI, 33.5%–57.3%). Median PFS by IRC was 5.6 months<br />

(95% CI, 4.6–8.2 months). The most frequently observed grade ≥3<br />

adverse events were thrombocytopenia (21.9%), increased aspartate<br />

aminotransferase (13.7%), increased alanine aminotransferase (8.2%)<br />

and vomiting (5.5%). One patient (1.4%) discontinued treatment due<br />

to thrombocytopenia. No patient received platelet transfusion. Grade<br />

3/4 hemorrhage was observed in one patient (1.4%). PK parameters<br />

for T-DM1 and its metabolites were consistent with previous phase<br />

I results.<br />

Conclusion: Single-agent T-DM1 has promising activity in Japanese<br />

patients with previously treated HER2-positive MBC and was well<br />

tolerated. Although the incidence of grade 3/4 thrombocytopenia<br />

tends to be higher than in Western studies, this was not associated<br />

with clinically important manifestations. (JapicCTI-101277)<br />

P5-18-17<br />

Impact of compliance with National Comprehensive <strong>Cancer</strong><br />

Network guidelines on adjuvant trastuzumab administration for<br />

patients with HER2-positive breast cancer.<br />

Mullins DN, Maly J, Abdel-Rasoul M, Shapiro CL, Olson EM.<br />

Comprehensive <strong>Cancer</strong> Center, The Ohio State University Wexner<br />

Medical Center, Arthur G. James <strong>Cancer</strong> Hospital and Richard J.<br />

Solove Research Institute, Columbus, OH; The Ohio State University<br />

Wexner Medical Center, Columbus, OH; The Ohio State University,<br />

Columbus, OH<br />

Background:<br />

The National Comprehensive <strong>Cancer</strong> Network (NCCN) breast cancer<br />

guidelines recommend trastuzumab as a component of adjuvant<br />

therapy for patients with stage I-III HER2-positive breast cancer.<br />

Reasons for noncompliance with adjuvant trastuzumab therapy and<br />

the impact of noncompliance with national guidelines are unknown.<br />

Methods:<br />

We retrospectively identified 331 patients with stage I-III HER2<br />

-positive breast cancer treated at the Ohio State University James<br />

<strong>Cancer</strong> Hospital 2005-2011 who were eligible for adjuvant<br />

trastuzumab. Medical records were reviewed to obtain age at<br />

diagnosis, race, co-morbidity score, stage, hormone positivity, type<br />

of therapy administered, date of disease recurrence, vital status and<br />

date of last follow up. Available clinician-documented reasons for not<br />

administering adjuvant trastuzumab were also recorded. Multivariate<br />

logistic regression modeling was used to examine the effect of these<br />

variables on completion of 1 year of adjuvant trastuzumab. Cox<br />

regression modeling was used to estimate the effect of completing 1<br />

year of trastuzumab on disease-free and overall survival, respectively.<br />

Results:<br />

Median follow up was 39.9 months (range 12.0-85.1). Of the 331<br />

patients, the majority of (289; 87%) received at least 1 dose of<br />

trastuzumab while 251 (76%) patients completed the recommended 1<br />

year of trastuzumab therapy. In multivariate modeling, age ≥ 70 years<br />

(Odds Ratio 0.18, 95% CI 0.07 to 0.47; p


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 5<br />

Conclusion:<br />

Age ≥ 70 years and stage I disease are predictors of noncompliance<br />

with NCCN guidelines recommending administration of adjuvant<br />

trastuzumab therapy for one year. Failure to complete one year<br />

of adjuvant trastuzumab is significantly associated with disease<br />

recurrence and worse overall survival in patients with stage I-III<br />

HER2-amplified breast cancer.<br />

P5-18-18<br />

Lapatinib versus trastuzumab, or both, added to preoperative<br />

chemotherapy for breast cancer: A meta-analysis of randomized<br />

evidence<br />

Valachis A, Nearchou A, Lind P. University of Uppsala, Sweden;<br />

Mälarsjukhuset, Eskilstuna, Sweden; Karolinska Institute, Stockholm,<br />

Sweden<br />

Purpose: We compared the efficacy and safety of the addition of<br />

lapatinib versus trastuzumab, or the combination, to neoadjuvant<br />

chemotherapy in HER2-positive breast cancer.<br />

Methods: Potentially eligible trials were identified through PubMed<br />

and Cochrane Library searches and abstracts of major international<br />

conferences. The endpoints that we assessed were pathologic<br />

complete response (pCR) rate and toxicity. Pooled relative risks<br />

(RR) were estimated for each endpoint with fixed or random effects<br />

models, depending on between-studies heterogeneity.<br />

Results: Six trials were identified with 1494 eligible patients. The<br />

probability to achieve pCR was higher for the trastuzumab plus<br />

chemotherapy arm than lapatinib plus chemotherapy (RR 1.25, 95%<br />

Confidence Interval (CI): 1.08-1.43; p-value = 0.003) (6 trials; 1494<br />

patients). Furthermore, the probability for pCR was significantly<br />

higher in the group given both lapatinib and trastuzumab than in<br />

the group given trastuzumab alone (RR 1.39, 95% CI: 1.20-1.63;<br />

p-value < 0.001) (4 trials; 779 patients). Diarrhea and dermatologic<br />

toxicities were statistically more frequent in patients receiving<br />

lapatinib than trastuzumab. The combination was associated with a<br />

higher rate of diarrhea than trastuzumab alone. No differences were<br />

observed regarding cardiac adverse events among patients receiving<br />

trastuzumab, lapatinib, or the combination.<br />

Conclusion: These data support the superiority of a dual-HER2<br />

inhibition for the treatment of HER2-positive breast cancer in the<br />

neoadjuvant setting. The direct comparison of trastuzumab and<br />

lapatinib demonstrated that lapatinib is inferior in terms of pCR and<br />

toxicity.<br />

P5-18-19<br />

The 2006 Adjuvant Trastuzumab Convention in Belgium: 5<br />

years later<br />

Vanderhaegen J, Paridaens R, Piccart M, Lalami Y, Machiels J-P,<br />

Louis E, Borms M, Mebis J, Dirix L, Lintermans A, Brouckaert O,<br />

Neven P. University Hospital Leuven, Belgium; University Hospital<br />

Leuven/Catholic University Leuven, Belgium; Institut Jules Bordet,<br />

Brussels, Belgium; Cliniques Universitaires St-Luc, Brussels,<br />

Belgium; CHU de Liège, Liege, Belgium; AZ Groeninge, Kortrijk,<br />

Belgium; Limburgs Oncologisch Centrum, Limburg, Belgium; St<br />

Augustinus Hospital, Wilrijk, Belgium<br />

Background: Outside clinical trials, long term follow-up data from<br />

large series of women treated with adjuvant trastuzumab are lacking.<br />

We here report such data from a Belgian cohort for premature<br />

trastuzumab discontinuation and 5 year outcome.<br />

Patients and Methods: A year prior to its reimbursement (June 1 st<br />

2007), the Belgian health authorities offered adjuvant trastuzumab<br />

to women with a HER-2 amplified breast cancer as given in HERA:<br />

following completion of loco-regional therapy, at least 4 cycles<br />

of chemotherapy (CT) (neo-adjuvant or adjuvant) or radiotherapy<br />

whichever was last. Demographics, histopathologic features, baseline<br />

(≥55%) and follow-up left ventricular ejection fraction (LVEF)<br />

together with 5 yr breast cancer related events (BCRE) were centrally<br />

recorded. Outcome was reported by grade (< or > grade 3), lymph<br />

node (pN) status (neg/pos), hormone receptor (HR) status (neg/pos)<br />

and timing of CT (adjuvant/neo-adjuvant).<br />

Results: 917 pts were included. Files on histopathology, demographics,<br />

LVEF and therapy are available for 887 (97%) pts, outcome for<br />

738 (80%). Median age at diagnosis was 53 years (25-81); 56%<br />

of tumors were >pT1, 57% pN+, 72% grade 3, 59% HR-positive<br />

for estrogen receptor (ER) and/or progesterone receptor (PR). CT<br />

was neo-adjuvant in 20% and adjuvant in 80%. Radiotherapy and<br />

endocrine therapy were given respectively in 86% and 50%. Median<br />

follow-up was 63 (range 8-90) months. Premature (


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P5-18-20<br />

Phase II study of pertuzumab, trastuzumab, and weekly paclitaxel<br />

in patients with metastatic HER2-overexpressing metastatic<br />

breast cancer.<br />

Datko F, D’Andrea G, Dickler M, Theodoulou M, Goldfarb S, Lake<br />

D, Fornier M, Modi S, Sklarin N, Comen E, Fasano J, Gajria D,<br />

Drullinsky P, Gilewski T, Murphy C, Syldor A, Lau A, Hamilton N,<br />

Patil S, Liu J, Chandarlapaty S, Hudis C, Dang C. Memorial Sloan-<br />

Kettering <strong>Cancer</strong> Center, New York, NY; Bon Secours Hospital,<br />

Cork, Ireland<br />

Background: Pertuzumab (P) is a monoclonal antibody which binds<br />

to extracellular domain II of HER2 distally from trastuzumab (H),<br />

disrupting HER2 dimerization and signaling. The CLEOPATRA<br />

phase III trial showed that HP + docetaxel in HER2+ metastatic breast<br />

cancer (MBC) prolonged progression-free survival (PFS) compared<br />

to placebo + H + docetaxel. We report preliminary results of a phase<br />

II study to evaluate the safety and efficacy of weekly paclitaxel with<br />

HP (THP).<br />

Methods: Patients (pts) with HER2+ MBC with 0-1 prior treatment<br />

(Rx) are eligible. Pts receive weekly (w) paclitaxel (80mg/m 2 ), q3w<br />

trastuzumab (loading dose 8mg/kg → 6mg/kg), and q3w pertuzumab<br />

(flat loading dose 840mg → flat dose 420mg). The primary endpoint is<br />

PFS at 6 months (mo). Secondary endpoints include response, safety<br />

(including cardiac events), and tolerability. Evaluable pts are those<br />

who have started study Rx and are assessed at 6 mo for PFS. Left<br />

ventricular ejection fraction (LVEF) is monitored by echocardiogram<br />

every 3 mo. Cardiac events are defined as symptomatic LV systolic<br />

dysfunction (LVSD), non-LVSD cardiac death, or probable cardiac<br />

death.<br />

Results: As of 6-1-12, 38 of the planned 69 pts were enrolled and<br />

20 pts were evaluable at 6 mo. Median age is 52 years (range 32 to<br />

72). A total of 11 pts (55%) previously received trastuzumab in the<br />

adjuvant or metastatic setting, and 8 pts (40%) were being treated<br />

in the second-line metastatic setting. Of the 20 evaluable pts, G 3/4<br />

toxicities were sepsis (1 pt, 5%), cholecystitis (1 pt, 5%), fatigue (1<br />

pt, 5%), skin ulceration (1 pt, 5%) and cystic macular degeneration<br />

(1 pt with prior prolonged Rx with paclitaxel, 5%). Common G 1/2<br />

toxicities included alopecia (20 pts, 100%), peripheral neuropathy<br />

(20 pts, 100%), fatigue (18 pts, 90%), ALT/AST elevation (17 pts,<br />

85%), diarrhea (15 pts, 75%), rash (13 pts, 65%), nail changes (10<br />

pts, 50%), mucositis (9 pts, 45%), dry skin (8 pts, 40%), and nausea<br />

(8 pts, 40%). Median LVEF was 64% at baseline (range 50% to 69%),<br />

60% at 3mo (range 50% to 73%), and 60% at 6mo (range 49% to<br />

67%). There were no cardiac events. At 6 mo, 15/20 pts (75%) were<br />

progression-free (2 CR, 8 PR and 5 SD); 5 pts had progressed. The 6<br />

mo PFS results for all 38 enrolled patients will be updated.<br />

Conclusions: Our single-center phase II study continues to accrue,<br />

with no clinically significant diarrhea or signal of increased cardiac<br />

toxicity to date. Pertuzumab was recently FDA-approved in<br />

combination with trastuzumab and docetaxel. If the estimate of safety<br />

and activity is similar to results with docetaxel in CLEOPATRA, this<br />

study will provide support for weekly paclitaxel as an alternative<br />

option in combination with trastuzumab and pertuzumab in this<br />

setting.<br />

P5-18-21<br />

A Phase II randomized trial of lapatinib with either vinorelbine<br />

or capecitabine as first- and second-line therapy for ErbB2-<br />

overexpressing metastatic breast cancer (MBC)<br />

Janni W, Sarosiek T, Pikiel J, Karaszewska B, Staroslawska E,<br />

Salat C, Caglevic C, Potemski P, Brain E, Briggs K, de Silvio<br />

M, Sapunar F, Papadimitriou C. Klinikum der Heinrich-Heine-<br />

Universität Düsseldorf, Düsseldorf, Germany; Centrum Medyczne<br />

Ostrobramska, NZOZ Magodent, Warsaw, Poland; Wojewodzkie<br />

Centrum Onkologii, Centrum Badan Klinicznych, Gdansk, Poland;<br />

Przychodnia Lekarska NZOZ “KOMED”, Konin, Poland; Centrum<br />

Onkologii Ziemii Lubelskiej, Lublin, Poland; Hämato-Onkologische<br />

Gemeinschaftspraxis, München, Germany; Mariano <strong>San</strong>chez<br />

Fontecilla, Las Condes, Chile; Wojewodzki Szpital Specjalistyczny,<br />

Kopernika, Poland; Institut Curie - Hôpital René Huguenin, Saint-<br />

Cloud, France; GlaxoSmithKline Oncology, Uxbridge, Middlesex,<br />

United Kingdom; GlaxoSmithKline Oncology, Collegeville, PA;<br />

Therapeutic Clinic, General Hospital of Athens, Greece<br />

Background: Lapatinib (L), a dual kinase inhibitor of epidermal<br />

growth factor receptor and ErbB2, is effective in the treatment of<br />

ErbB2+ MBC in combination with capecitabine (C) following<br />

progression after trastuzumab, anthracyclines, and taxanes.<br />

Vinorelbine (V) is an important chemotherapy option in MBC, and<br />

multiple Phase II trials have been conducted in combination with<br />

trastuzumab. This randomized, open-label, multicenter, Phase II<br />

study (LAP112620, VITAL) evaluated the efficacy and safety of L<br />

with either V or C in women with ErbB2+ MBC.<br />

Methods: Patients with MBC who had received ≤1 chemotherapy<br />

regimen in the metastatic setting were randomized 2:1 to either L<br />

1250 mg orally once daily (QD) continuously plus V 20 mg/m2<br />

intravenously on Days 1 and 8, every third week, or L 1250 mg<br />

orally QD continuously plus C 2000 mg/m2/day orally in 2 doses 12<br />

hours apart on Days 1-14 every third week. Patients were stratified<br />

by prior receipt of therapy for MBC (Y/N) and site of metastatic<br />

disease (visceral/soft tissue or bone-only). The primary endpoint<br />

of progression-free survival (PFS) was assessed once all subjects<br />

had been followed for a minimum of 6 months or had otherwise<br />

progressed, died or withdrawn, if sooner. The primary focus was to<br />

evaluate PFS in the L plus V arm with a descriptive intent only. Other<br />

endpoints included overall response rate, overall survival, and safety.<br />

Patients progressing on one treatment arm were given the option of<br />

crossing over to the other arm.<br />

Results: 112 patients were randomized. The results and conclusions<br />

sections will be updated once the primary analysis has been completed<br />

in September 2012.<br />

clinicaltrials.gov - NCT01013740<br />

P5-18-22<br />

Long-term survival of patients with HER2 metastatic breast<br />

cancer treated by targeted therapies.<br />

Fiteni F, Villanueva C, Bazan F, Chaigneau L, Cals L, Dobi E,<br />

Montcuquet P, Nerich V, Limat S, Pivot X. Besançon University<br />

Hospital, Besançon, Doubs, France<br />

Purpose<br />

HER2 targeted therapies have substantially improved outcomes of<br />

patients with HER2 positive metastatic breast cancer (MBC). Medical<br />

records of patients with HER2 positive MBC achieving long survival<br />

were reviewed to describe their clinical characteristics and to assess<br />

whether curability is possible.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

474s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 5<br />

Patients and methods<br />

One thousand and seven hundred patients with MBC were treated<br />

between 01/01/2003 and 31/05/2012 in the 5 hospitals from the<br />

Franche-Comte region. All 217 patients with HER2 positive MBC<br />

region were retrospectively studied.<br />

Results<br />

Among this population of HER2 positive MBC, 56 patients (26%)<br />

survived longer than 5 years. The characteristics of these long<br />

survivors were the following: median age at diagnosis was 55 years<br />

old (range, 33-87); 20 patients (35,7%) were metastatic at presentation;<br />

20 patients (35,7%) had received adjuvant chemotherapy; the<br />

median disease-free interval was 17 months; brain, liver, lung, bone<br />

metastases were observed in 4 (7%), 19 (34%), 13 (23%), 23 (41%)<br />

in cases at occurrence of metastatic disease, respectively; tumours<br />

were hormonal receptors positive in 39 cases (70%) of these patients.<br />

A total of 16 (29%), 7 (12,5%), 7 (12,5%) and 26 (46%) cases had<br />

received 1, 2, 3 or more lines of chemotherapy at metastatic setting.<br />

The proportion of HER2 targeted treatment was 79%, 61%, 50%<br />

in first, second and third line, respectively. All patients received<br />

trastuzumab or lapatinib for their disease and 18 patients (32%) were<br />

given an anthracycline containing regimen.<br />

There were 7 complete response (CR), there were all observed after<br />

one line of chemotherapy based on trastuzumab combined with a<br />

taxane in all cases. Among these 7 patients, all continued trastuzumab<br />

after CR and all RH+ patients received hormonotherapy after CR.<br />

Among these 56 patients, 35 (62,5%) were still alive at the date<br />

cutoff in 31/05/2012.<br />

Conclusion<br />

A significant proportion of patients with HER2 positive MBC were<br />

long survivors. Prospective clinical trials in this subset of patients<br />

should take into account this high number of patients with favorable<br />

outcome to achieve conclusion in term of survival. Can one consider<br />

the curabiblity of some MBC patients with HER2 overexpression?<br />

P5-18-23<br />

The prognosis of T1a, T1b N0 M0, HER2+ patients in Korea.<br />

Lee JW, Moon H-G, Han W, Noh D-Y. Seoul National University<br />

Hospital, Seoul, Korea<br />

Backgrounds: Increased expression of HER-2 or amplification of<br />

the HER-2/neu gene has been associated with a worse prognosis<br />

than with other breast cancer phenotypes. Especially in cases of size<br />

of 0.5∼1cm and node-negative status, adjuvant chemotherapy with<br />

or without Trastuzumab treatment is suggested by recent National<br />

Comprehensive <strong>Cancer</strong> Network(NCCN) guidelines. In Korea, the<br />

breast cancer patient pool is more younger than western data, and<br />

there could be some ethnic differences.<br />

Methods: The Korean <strong>Breast</strong> <strong>Cancer</strong> Society (KBCS) has been<br />

collaborating with the Korean Central <strong>Cancer</strong> Registry (KCCR) to<br />

improve the completeness and validity of the breast cancer registry<br />

and the mortality data. Data of T1a,bN0M0 breast cancer patients<br />

from 1999 to 2006 from KBCS on-line registry was obtained, and a<br />

total of 2,962 patients were included for survival analysis.<br />

Results: 711 patients (24.0%) was HER-2 positive. Hormone receptor<br />

and HER2 status was used to classify 4 molecular subtypes; Hormone<br />

receptor(HR)+/HER2-, HR+/HER2+, HR-/HER2+, HR-/HER2-. The<br />

median follow-up was for 55.82 month (±24.196). <strong>Breast</strong> cancer<br />

specific survival rate was lowest at HR-/HER2+ subtype(97.9%).<br />

It’s 1.0∼1.8% lower than other subtypes. Between all T1N0M0 cases,<br />

especially T1b HR-/HER2+ cases showed lowest survival of 96.4%.<br />

Conclusion: This study calculated the nationwide survival rates of<br />

Korean breast cancer patients with T1a,bN0M0 disease for the first<br />

time. HER2 gene positivity was associated with worse prognosis<br />

and it’s concordant with western studies. But prognosis of T1N0M0<br />

lesion is much better than that of other nations. So, the usage of<br />

chemotherapy and Trastuzumab treatment should be considered more<br />

carefully in Korea. Further analysis of the long-term survival and a<br />

nationwide collaborative study should be performed to estimate the<br />

survival trend of Korean breast cancer patients.<br />

P5-18-24<br />

Population pharmacokinetics of trastuzumab emtansine, a HER2-<br />

targeted antibody-drug conjugate, in patients with HER2-positive<br />

metastatic breast cancer: clinical implications of the effect of<br />

various covariates<br />

Lu D, Girish S, Gao Y, Wang B, Yi J-H, Guardino E, Samant M,<br />

Cobleigh M, Rimawi M, Conte P, Jin J. Genentech, Inc.; Quantitative<br />

Solutions; Rush University Medical Center; Baylor College of<br />

Medicine; University of Modena and Reggio Emilia<br />

Background<br />

Trastuzumab emtansine (T-DM1) is a HER2-targeted antibodydrug<br />

conjugate composed of the humanized monoclonal antibody<br />

trastuzumab, the potent cytotoxic agent DM1 (a microtubule inhibitor),<br />

and a stable thioether linker. To estimate typical pharmacokinetic<br />

(PK) parameter values and interpatient variability, a population PK<br />

model for T-DM1 was previously developed from 1 phase 1 (0.3 to<br />

4.8 mg/kg in qw or q3w regimens) and 2 phase 2 (3.6 mg/kg q3w)<br />

trials (Gupta, J Clin Pharmacol 2012). The model reported here has<br />

been updated with additional data from 2 randomized trials (phase 2<br />

TDM4450g and phase 3 EMILIA, 3.6 mg/kg q3w). Another phase 2<br />

trial (TDM4688g) was used for external validation of the model. The<br />

effect of demographic and pathophysiological covariates on the PK<br />

of T-DM1 was explored to better understand the clinical factors that<br />

might affect exposure and clinical outcome for individual patients.<br />

Methods<br />

For the current analysis, 9934 T-DM1 serum concentration-time<br />

data points from 671 patients were simultaneously fitted using<br />

NONMEM® software. T-DM1 concentration-time data to date are<br />

best described using a 2-compartment linear model. All relevant<br />

and plausible covariates likely to have an effect on T-DM1 systemic<br />

exposure, or likely to have clinical relevance, were explored for<br />

possible correlation with the key T-DM1 PK parameters of clearance<br />

(CL) and central volume of distribution (V c ). These covariates include<br />

those related to demographics, renal and hepatic function, disease<br />

status, and treatment history.<br />

Results<br />

The estimated CL for T-DM1 is 0.68 L/day, V c is 3.13 L, and the<br />

terminal half-life is 3.94 days. Interindividual variability (IIV) of<br />

the base model is 25.6% and 17.5% for CL and V c , respectively.<br />

Patients with greater body weight, sum of longest dimension of target<br />

lesions, serum concentration of shed HER2 extracellular domain, and<br />

aspartate aminotransferase concentrations, as well as patients with<br />

lower serum albumin and baseline trastuzumab concentrations, have<br />

statistically faster CL. Patients with greater body weight also have<br />

statistically larger V c . Incorporation of these covariates (P


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Conclusions<br />

A relatively small IIV for the estimated T-DM1 PK parameters of CL<br />

and V c was observed. None of the evaluated covariates had a clinically<br />

meaningful magnitude of effect on T-DM1 exposure (


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 5<br />

between arms. Causes of death remained unchanged from the first<br />

interim OS analysis, with the most common cause being progressive<br />

disease. Adverse events leading to death were rare and balanced<br />

between arms.<br />

Conclusions: Treatment of pts with HER2-positive 1L MBC with<br />

P+T+D compared with Pla+T+D was associated with an improvement<br />

in OS, which was both statistically significant and clinically<br />

meaningful. These results show that combined HER2 blockade and<br />

chemotherapy using the P+T+D regimen can be considered a standard<br />

of care for pts with HER2-positive MBC in the 1L setting.<br />

P5-19-01<br />

Targeting the HER3-phosphatidyl inositol-3 kinase pathway in<br />

breast cancers<br />

Cook RS, Morrison MM, Arteaga CL, Perou CM. Vanderbilt<br />

University, Nashville, TN; University of North Carolina<br />

Background: Using a mouse model of HER3 ablation in the mammary<br />

epithelium, we previously showed that HER3 is required for the<br />

genesis of transgenic HER2-overexpressing breast cancers. However,<br />

increasing evidence suggests that transgenic models of HER2<br />

overexpression result in luminal B-like mammary tumors, which<br />

resemble human breast tumors that express estrogen receptor (ER)<br />

but are often not responsive to endocrine therapy. Furthermore, we<br />

found that HER3 is required in the untransformed mouse mammary<br />

epithelium for differentiation and survival of luminal cells, but not<br />

basal cells. Herein we report that HER3 drives growth and survival of<br />

luminal breast cancers in a manner that parallels its role in maintaining<br />

the untransformed luminal epithelium in the breast.<br />

Results: Expression profiling of a large human breast cancer dataset<br />

demonstrated that HER3 expression was highest in Luminal breast<br />

cancers as compared to different molecular subtypes of breast cancers,<br />

including HER2-enriched, claudin-low, and basal-like breast cancers.<br />

In luminal breast cancer cell lines MCF7, ZR75-1, and MDA-MB-361,<br />

HER3 targeting using siRNA or a monoclonal antibody against HER3<br />

decreased Akt, S6, and Erk1/2 phosphorylation, cell growth and cell<br />

survival under conditions of estrogen deprivation. Luminal breast<br />

tumor xenografts treated with U3-1287, a humanized monoclonal<br />

anti-ErbB3 antibody, or EZN-3920, an anti-sense oligonucleotide<br />

against ErbB3, decreased tumor growth and phosphatidyl inositol-3<br />

kinase (PI3K)/mTOR signaling in vivo. Interestingly, luminal breast<br />

tumor xenografts responded to the ER inhibitor fulvestrant with<br />

increased HER3 expression and phosphorylation, and with increased<br />

PI3K/mTOR signaling as shown by enhanced S6 phosphorylation.<br />

Fulvestrant-mediated upregulation of HER3 was also seen in matched<br />

clinical breast tumor specimens harvested before treatment and after<br />

21 days of fulvestrant treatment.AMG-888 blocked fulvestrantinduced<br />

HER3 and phospho-S6 upregulation in vivo, resulting in<br />

regression of luminal breast tumor xenografts.<br />

Conclusions: These data suggest that HER3 targeting may improve the<br />

outcome of luminal breast cancers treated with endocrine therapy, and<br />

are consistent with our findings that a HER3-dependent expression<br />

signature positively correlates with relapse in tamoxifen-treated<br />

patients. Taken together, these results define a novel role for HER3<br />

in luminal breast cancer cells, and support future investigations into<br />

therapeutic strategies to target HER3 in luminal breast cancers.<br />

P5-19-02<br />

Selective PI3K and dual PI3K/mTOR inhibitors enhance the<br />

efficacy of endocrine therapies in breast cancer models<br />

Friedman L, Ross LB, Wallin J, Guan J, Prior WW, Wu E, Nannini<br />

M, Sampath D. Genentech, Inc., South <strong>San</strong> Francisco, CA<br />

Purpose: Phosphoinositide 3-kinases (PI3K) are lipid kinases that can<br />

regulate breast tumor cell growth, migration and survival. Standard<br />

of care drugs such as estrogen receptor (ER) antagonists including<br />

fulvestrant and tamoxifen, and aromatase inhibitors such as letrozole<br />

are indicated for the treatment of hormone receptor positive breast<br />

cancer. The current study is focused on investigating preclinical<br />

activity in breast cancer models, for GDC-0941, a class I PI3K<br />

inhibitor, GDC-0032, a PI3K inhibitor, and GDC-0980, a dual mTOR<br />

kinase and class I PI3K inhibitor. Investigation into PI3K inhibitor<br />

efficacy in combination with endocrine therapies is also explored.<br />

Experimental Design: A panel of ER+ breast cancer cell lines were<br />

treated with GDC-0941, GDC-0032 and GDC-0980 either as single<br />

agents or in combination with fulvestrant or tamoxifen and assayed for<br />

cellular effects. MCF-7 cells ectopically expressing aromatase were<br />

utilized to test the efficacy of aromatase inhibitors in combination<br />

with PI3K inhibitors in vitro. In addition, human xenografts of breast<br />

cancer cell lines were employed to assess combination efficacy of<br />

PI3K inhibitors with fulvestrant and tamoxifen in vivo.<br />

Results: Combination of GDC-0941, GDC-0032 or GDC-0980 with<br />

endocrine therapies resulted in a decrease in cellular viability and<br />

an increase in cell death. Synergy of PI3K inhibitor combinations<br />

with fulvestrant or tamoxifen was assessed using Combination<br />

Index (C.I.), and C.I. values as low as 0.1 indicated strong synergy<br />

in some contexts. Combination activity of PI3K inhibitors and<br />

letrozole was also observed in MCF7 cells expressing aromatase.<br />

In MCF-7 xenografts, the combination of GDC-0980, GDC-0032<br />

and GDC-0941 enhanced activity of fulvestrant resulting in tumor<br />

regressions and tumor growth delay (116% tumor growth inhibition<br />

(TGI) for GDC-0980 and 91% TGI for GDC-0941 and GDC-0032). In<br />

addition, the combination of GDC-0941 or GDC-0032 with tamoxifen<br />

enhanced the efficacy of tamoxifen in vivo (83%TGI for GDC-0941<br />

and 102%TGI for GDC-0032). Mechanism of action and biomarker<br />

studies are underway.<br />

Conclusion: Collectively, the non-clinical efficacy data provide a<br />

strong rationale to evaluate the combination of PI3K inhibitors with<br />

anti-estrogen therapy in hormone receptor positive breast cancer.<br />

P5-19-03<br />

Olaparib plus carboplatin in combination with vandetanib<br />

inhibited the growth of BRCA-wt triple negative breast tumors<br />

in mice: Outside BRCA-box<br />

Dey N, Sun Y, De P, Leyland-Jones B. <strong>San</strong>ford Research/USD, Sioux<br />

Falls, SD<br />

Introduction: PARP inhibition has emerged as one of the most<br />

exciting and promising ‘targeted’ therapeutic strategies in the<br />

treatment of advanced triple negative breast cancer (TNBC) – the<br />

intended ‘target,’ being DNA repair (Anders CK, 2010). Although<br />

olaparib is known to have antitumor activity in BRCA-related TNBC<br />

cells, a limited number of preclinical and clinical studies have shown<br />

antitumor efficacy of olaparib in non-BRCA-related BC (Shimo<br />

T, 2012). Understanding the biology of TNBC cells has identified<br />

molecular targets including RTK(s), such as EGFR.<br />

Purpose: Here we tested the combination of a PARP inhibitor,<br />

olaparib (O) (AstraZeneca) plus carboplatin (C) with the EGFR/<br />

VEGFR inhibitor, vandetanib (V) (AstraZeneca) in a BRCA-wt<br />

TNBC model.<br />

www.aacrjournals.org 477s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Methods: Athymic mice bearing TNBC (BRCA-wt VEGFR<br />

expressing MDA-MB231& MDA-MB468 [EGFR amplified/<br />

overexpressed]) xenograft tumors (200 mm 3 ) were treated with O<br />

plus C in combination with V (Arms: vehicle control 1, vehicle<br />

control 2, C+O, V, C+O+V). In vitro effects of V in combination<br />

with O plus C (or temozolomide) on clonogenic growth (3D on-TOP<br />

assay), proliferation (MTT and CelltiterGLO), apoptosis (Apoptosis<br />

Array), cell signaling marker(s) (Western Blot), and tumor cell<br />

phenotypes (fibronectin-directed migration, matrigel-invasion, and<br />

vascular mimicry) were investigated in a panel of five BRCA-wt and<br />

BRCA-mutated TNBC cell lines. The effects of V were tested on (a)<br />

cell signaling marker(s), (b) angiogenesis marker (HIF-1alpha), and<br />

(c) angiogenesis related phenotypes (vitronectin-directed migration,<br />

and cord formation) in HUVEC.<br />

Results: (1) O plus C in combination with V caused a regression of<br />

the in vivo growth of established tumors by 50% which was evident<br />

in both the BRCA-wt TNBC models tested. Interestingly, a marked<br />

suppression of the progression of tumor-growth was observed in the<br />

O plus C arm. (2) In vitro, V alone (10 µM) inhibited baseline as<br />

well as EGF-induced phosphorylation of AKT (S473 / T308), S6RP,<br />

4EBP1 and ERK. (3) TNBC cells exhibited higher sensitivity to V in<br />

clonogenic assays when combined with a 10 µM fixed dose of O and<br />

C / temozolomide. (4) A combination of V with O plus C increased<br />

cleaved caspase-3, PARP cleavage, and pro-apoptotic signals, while<br />

inhibiting vascular mimicry, migration, and invasion in MDA-MB231,<br />

MDA-MB468, and SUM149 cells. (5) Treatment with V blocked<br />

cord formation, migration, EGF-induced HIF-1α accumulation, and<br />

phosphorylation of AKT, 4EBP1, and ERK in HUVEC.<br />

Conclusion: A profound anti-tumor efficacy of O plus C in<br />

combination with V in BRCA-wt TNBC model can be explained by<br />

a significant anti-proliferative/pro-apoptotic and anti-migratory/antiinvasive<br />

actions of the drugs (alone or in combination), as observed<br />

both in tumor cells as well as in endothelial compartments. The<br />

combination of O plus C plus V merits further investigation in TNBC.<br />

P5-20-01<br />

A randomized double-blind phase II study of the combination<br />

of oral WX-671 plus capecitabine vs. capecitabine monotherapy<br />

in first-line HER2- negative metastatic breast cancer (MBC).<br />

Goldstein LJ, Oliveria CT, Heinrich B, Stemmer SM, Mala C, Selder<br />

S, Bevan P, Harbeck N. Fox Chase <strong>Cancer</strong> Center, Philadelphia,<br />

PA; Instituto Brasilerio Controle <strong>Cancer</strong>, Sao Paulo, Brazil;<br />

Hamatologisch-Onkologische-Praxis Augsburg, Augsburg, Germany;<br />

Rabin Medical Center, Petah Tikva, Israel; Wilex, Munich, Germany;<br />

Univeristy of Munich, Munich, Germany<br />

Background: uPA and its inhibitor PAI-1 play a key role in tumor<br />

invasion, metastasis and tumor growth. High levels of uPA and PAI-<br />

1 in breast tumors are statistically significant prognostic factors of<br />

disease-free (DFS) and overall survival (OS), which were validated<br />

at the highest level of evidence, as well as predictors for benefit<br />

of adjuvant chemotherapy. WX-UK1 is an active site competitive<br />

inhibitor of uPA with an inhibition constant in the submicromolar<br />

range. WX-671 (upamostat) is an oral prodrug of WX-UK1. In<br />

preclinical animal tumor models, both WX-UK1 and WX-671 have<br />

been shown to reduce the growth rate of implanted tumors, to inhibit<br />

invasion, and reduce metastases. This current proof of concept study<br />

is designed to substantiate the anti-metastatic properties of upamostat<br />

for patients appropriate for first line therapy for MBC.<br />

Methods: Female patients aged >18, with HER2 negative MBC<br />

appropriate for first line monotherapy with capecitabine, with<br />

adequate performance status, organ function, bone marrow reserve<br />

without brain metastases were eligible. Patients were randomized in a<br />

double-blind fashion to receive upamostat (200mg orally daily for 21<br />

days) plus capecitabine (1000 mg/m2 orally twice daily for 14 days)<br />

vs. capecitabine (1000 mg/m2 orally twice daily for 14 days) in 3 week<br />

treatment cycles until progressive disease or unacceptable toxicity.<br />

The primary endpoint is to evaluate the efficacy of the combination<br />

of upamostat plus capecitabine compared to monotherapy as assessed<br />

by comparison of progression free survival. The secondary objectives<br />

are OS, objective response rates, safety and tolerability, and to assess<br />

the pharmacokinetics (PK) of upamostat and capecitabine when<br />

combined.<br />

Results: Between August 2008 and April 2011,132 patients were<br />

enrolled. 17 patients are still receiving treatment. 26% of the patients<br />

are characterized as triple negative, 13% as only Estrogen Receptor<br />

(ER) positive and 4% as only Progesteron Receptor (PR) positive.<br />

57 % of the patients are ER and PR positive.<br />

Conclusions: Progression free survival, response rates and safety<br />

will be reported. This abstract is being submitted as a placeholder. A<br />

completed abstract will be submitted when the analyses are completed.<br />

P5-20-02<br />

Predictors of long-term survival in a large cohort of patients<br />

with HER2-positive (HER2+) metastatic breast cancer (MBC).<br />

Chavez Mac Gregor M, Lei X, Giordano SH, Valero V, Esteva F,<br />

Mittendorf EA, Gonzalez-Angulo AM, Hortobagyi GN. University<br />

of Texas MD Anderson <strong>Cancer</strong> Center, Houston, TX<br />

Background: Numerous studies have examined prognostic factors for<br />

survival in breast cancer patients (pts), but few have focused on longterm<br />

survival among pts with HER2+ MBC receiving HER2-targeted<br />

therapy. In this study we sought to evaluate the clinical characteristics<br />

and predictive factors of long-term survival in this group of pts.<br />

Methods: All pts with HER2+ MBC treated with HER2-directed<br />

therapy at MD Anderson <strong>Cancer</strong> Center were identified by<br />

retrospective review of the Institutional <strong>Breast</strong> Medical Oncology<br />

database. HER2 status was determined by IHC or FISH. Patient<br />

characteristics were analyzed using descriptive statistics. Overall<br />

survival (OS) was measured from the date of diagnosis of the first<br />

distant metastasis to the date of death or last contact. Pts were grouped<br />

according to OS and categorized as long term-survivors (LTS, OS<br />

≥5 years), or non-long term survivors (non-LTS, OS 50, p=0.005);<br />

stage at diagnosis (25% for stage I-III vs. 36% for stage IV de novo,<br />

p


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 5<br />

breast surgery among pts with stage IV de novo (OR=2.88; 95% CI<br />

1.47-5.66) were significantly associated with long-term survival.<br />

Conclusions: Some patients with HER2+positive MBC have an<br />

excellent outcome, particularly younger pts with HR-positive<br />

tumors, low burden of disease and non-visceral metastases in which<br />

multidisciplinary treatment is favored. In the era of HER2-targeted<br />

therapies, 14.5% of the patients are long-term survivors. To our<br />

knowledge this is the largest series identifying the characteristics of<br />

this group of patients.<br />

P5-20-03<br />

A phase 2, double-blind, randomized, placebo-controlled, dosefinding<br />

study of sotatercept for the treatment of patients with<br />

chemotherapy-induced anemia and metastatic breast cancer.<br />

Auerbach M, Osborne CRC, Klesczewski K, Laadem A, Sherman<br />

ML, Bianca R. Auerbach Hematology/Oncology, Baltimore, MD;<br />

Texas Oncology, P.A., Sammons <strong>Cancer</strong> Center, Dallas, TX; Celgene<br />

Corporation, Summit, NJ; Acceleron Pharma, Cambridge, MA<br />

Background: Sotatercept is a recombinant activin receptor IIA<br />

(ActRIIA) ligand trap and is comprised of the extracellular domain<br />

of ActRIIA linked to the Fc domain of human IgG1 (ActRIIA-IgG1).<br />

Results of preclinical and early clinical studies provide evidence that<br />

sotatercept increases the concentration of hemoglobin (Hb) in blood<br />

and may constitute a novel treatment for chemotherapy-induced<br />

anemia (CIA), a condition that results in significant morbidity.<br />

Methods: This study evaluated the safety of, and hematopoietic<br />

response to, sotatercept in patients with CIA and metastatic breast<br />

cancer (MBC). Response was defined as increase in Hb ≥1 g/dL for<br />

≥28 consecutive days during treatment or ≤2 months following the last<br />

dose, in the absence of red blood cell (RBC) transfusion or treatment<br />

with an erythropoiesis stimulating agent (ESA). If RBC transfusion<br />

or treatment with an ESA was required, no Hb measurements within<br />

28 days of the RBC transfusion or administration of ESA were<br />

used to determine the Hb response. Subjects were randomized to<br />

subcutaneous injection of sotatercept (0.1, 0.3, or 0.5 mg/kg) or<br />

placebo and were treated on day 1 of each of four 28-day cycles.<br />

Results: Thirty (30) subjects were enrolled and treated (5 placebo,<br />

25 sotatercept). Increases in mean Hb in the 0.3 and 0.5 mg/kg<br />

groups were greater than in 0.1 mg/kg and placebo groups. Increases<br />

peaked approximately 15 days following treatment and declined<br />

over the remaining interval between treatments. Among 23 subjects<br />

with confirmed CIA and administered ≥1 dose and followed for ≥57<br />

days (per-protocol analysis), 5/18 (28%) administered sotatercept<br />

responded (0/5 [0%] at 0.1 mg/kg; 3/9 [33%] at 0.3 mg/kg; 2/4<br />

[50%] at 0.5 mg/kg) vs 1/5 [20%] administered placebo. Among 13<br />

nonresponders administered sotatercept, 5 (39%) experienced ≥1<br />

dose interruption/reduction, as per protocol, due to elevated Hb. The<br />

incidence of adverse events (AEs) was consistent with that reported<br />

in patients with MBC. No dose-limiting or dose-related toxicity was<br />

observed.<br />

Conclusions: Sotatercept demonstrated dose-dependent hematopoietic<br />

activity in subjects with CIA and MBC. The safety findings were<br />

generally consistent with the known safety profile of sotatercept.<br />

These data suggest that a greater dose of sotatercept or a dose<br />

interval


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P5-20-05<br />

A Phase 2 trial of RAD 001 and Carboplatin in patients with triple<br />

negative metastatic breast cancer<br />

Singh JC, Volm M, Novik Y, Speyer J, Adams S, Omene CO, Meyers<br />

M, Smith JA, Schneider R, Formenti S, Goldberg JD, Li X, Davis S,<br />

Beardslee B, Tiersten A. New York University, New York, NY<br />

BACKGROUND: RAD001 is an oral mTOR inhibitor that has<br />

exhibited activity in breast cancer. Triple negative breast cancer<br />

cells are unable to repair double stranded DNA breaks and hence<br />

have sensitivity to platinum agents that cause interstrand crosslinks.<br />

Rapamycin acts synergistically with platinum agents to induce<br />

apoptosis and inhibit proliferation in at least two different breast<br />

cancer cell lines (including ER/PR negative cell lines). We propose<br />

that combination RAD001 and carboplatin may have activity in<br />

triple-negative breast cancer.<br />

MATERIALS AND METHODS: The primary objective of the study<br />

is to determine clinical benefit (complete remission (CR) + partial<br />

remission (PR) + stable disease (SD more than 6 months)) and the<br />

toxicity of this combination in triple negative metastatic breast cancer<br />

who have had 0-3 prior chemotherapy regimens for metastatic disease.<br />

Twenty-five subjects were to be entered in this Phase II study. This<br />

design has greater than 80% power to test the null hypothesis that the<br />

clinical benefit rate is less than or equal to 10% versus the alternative<br />

hypothesis that clinical benefit rate is greater than or equal to 30%.<br />

Prior carboplatin is allowed. Women with treated brain metastasis<br />

are eligible. Secondary objectives are to determine progression free<br />

survival and relationship between pretreatment sensitivity (biopsy<br />

at baseline) and clinical response (biopsy post 2 cycles) using IHC<br />

staining for abundance of key proteins in the Akt-mTOR pathway and<br />

their activity using surrogate phosphorylation site-specific antibodies.<br />

According to the original study plan, carboplatin AUC 6, was to be<br />

given intravenously every three weeks. Five mg of RAD001 was<br />

to be given daily with a 3 patient run-in and then 10 mg daily if<br />

there were no dose-limiting toxicities. Due to a surprising amount<br />

of thrombocytopenia with this combination the dose of carboplatin<br />

was first amended to AUC 5 and most recently to AUC 4 with 5 mg<br />

of RAD001 (and no plan to escalate to 10 mg).<br />

RESULTS: 23 patients of a planned 25 have been recruited thus far.<br />

Median age is 59. Of the 20 patients assessable for response at this<br />

time, there have been 1 CR, 5 PRs, 8 SDs and 6 PDs. One SD was<br />

achieved in a patient progressing on single agent Carboplatin at study<br />

entry. Median duration of CR+ SD +PR thus far is 13 weeks (range:<br />

6-74 weeks). 5 of 22 patients assessable for toxicity had grade 3/4<br />

thrombocytopenia and 4 patients had grade 3 neutropenia (no febrile<br />

neutropenia). 13 out of eighteen patients have had treatment held and/<br />

or dose reductions secondary to hematological toxicity, however, since<br />

amendment for starting dose of Carboplatin to AUC 4 the regimen<br />

has been very well tolerated with only 1 out of eleven patients with<br />

grade 3 neutropenia and grade 3 thrombocytopenia. 1 patient suffered<br />

from grade 3 dehydration. The estimated clinical benefit rate is 45%<br />

(95% confidence interval: 23%, 67%). Median time to progression<br />

or death is 85 days from start of treatment.<br />

CONCLUSIONS: Our study has met the primary end point of<br />

demonstrating clinical benefit in triple negative metastatic breast<br />

cancer. Dose limiting thrombocytopenia was an unexpected side<br />

effect requiring protocol amendment. We continue to accrue study<br />

subjects at the amended dosing.<br />

P5-20-06<br />

RAD001 (Everolimus) in combination with Letrozole in the<br />

treatment of postmenopausal women with estrogen receptor<br />

positive Metastatic <strong>Breast</strong> cancer after failure of hormonal<br />

therapy – a phase II study<br />

Safra T, Kaufman B, Ben Baruch N, Kadouri-Sonenfeld L, Nisenbaum<br />

B, Greenberg J, Ryvo L, Yerushalmi R, Evron E. Tel Aviv Sourasky<br />

MC, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;<br />

Sheba Medical Center, Ramat Gan, Israel; Kaplan Medical Center,<br />

Rehovot, Israel; Hadassah Ein Karem Medical Center, Jerusalem,<br />

Israel; Meir Kfar Saba Medical Center, Kfar Saba, Israel; Rabin<br />

Medical Center Belinson Campus, Petach Tikva, Israel; Assaf<br />

Harofeh Medical Center, Assaf Harofeh, Israel<br />

Background and Objectives: Approximately 70% of postmenopausal<br />

women (PMW) present with hormone receptor positive Metastatic<br />

<strong>Breast</strong> cancer (MBC) and there is a need for new treatment modalities<br />

after failure of prior endocrine therapy. Activation of the mTOR<br />

pathway is a key adaptive change driving endocrine resistance. Preclinical<br />

and early clinical data suggest synergistic effect between<br />

RAD001 (Everolimus, a Novartis mTOR inhibitor) and Letrozole<br />

results in more profound effects on proliferation arrest and induction<br />

of tumor cell kill.<br />

Our objectives are to assess overall response rate (ORR) as well as<br />

progression free survival (PFS), overall survival (OS) and safety<br />

profile with treatment of Letrozole and RAD001 in PMW with MBC<br />

after failure of endocrine therapies.<br />

Methods and materials: A multi-center, Israeli phase II open label<br />

study evaluating treatment of RAD001 (10 mg daily) combined with<br />

Letrozole (2.5 mg daily) in PMW with MBC after recurrence or<br />

progression on Tamoxifen and/or Anastrozole and/or Letrozole and/<br />

or Fulvestarnt and/or Exemestane.<br />

Sixty five patients were enrolled in 7 institutes. This abstract presents<br />

data and preliminary results of 24 patients in 2 of the study centers.<br />

Median age was 65.5 (54-80) years. Hormonal status were: 50% of<br />

the patients had ER+/PR+, 40% ER+/PR- and 10% ER-/PR+. All<br />

had MBC with 1 to 3 metastatic sites i.e. bones (90%), liver (30%),<br />

lung (12%) and skin (12%).<br />

Patients were previously exposed to a median of 1 (1-3) therapy line<br />

for MBC, all previously treated with hormonal therapy for MBC<br />

and the majority were also exposed to adjuvant hormonal therapy.<br />

Results: With a median follow up of 4 months (range 3-12), partial<br />

response was observed in 22%, stable disease in 28% and progressive<br />

disease in 50% of patients. Median time to progression (TTP) was 4<br />

months (3-11) with 65% still on treatment. Median OS is too early<br />

to evaluate (all but 2 are still alive).<br />

Grade III bone marrow toxicity was observed in about 5%. Nonhematological<br />

toxicities were: grade I fatigue in about 90%, rash and<br />

irritation(grade I-II) in about 70%, glucose level, liver function tests,<br />

blood cholesterol and triglycerides increased and/or renal function<br />

alterations (grade I-III) in about 80%, all the above disappeared after<br />

treatment delays or dose reduction, 5% developed non-inflammatory<br />

pneumonitis.<br />

The 2 most common toxicities were mucositis; 30% grade I and 50%<br />

grade II and significant weight loss (5-10% weight reduction) in 90%.<br />

Conclusions: Our study shows significant activity with the<br />

combination of RAD001 and Letrozole in MBC previously treated<br />

several lines of hormonal treatment with an acceptable toxicity.<br />

Combining RAD001 with Letrozolein the treatment of MBC,<br />

potentially inhibit tumor cell growth\proliferation and act as anti<br />

angiogenesis while at the same time potentially preventing the<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

480s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 5<br />

development of Letrozole resistance. Longer follow-up and further<br />

evaluation is warranted, additional data will be collected and provided<br />

towards the 2012 SABCS.<br />

P5-20-07<br />

Phase II Trial of Dasatinib in Combination With Weekly<br />

Paclitaxel for Patients with Metastatic <strong>Breast</strong> Carcinoma<br />

Morris PG, Lake D, McArthur HL, Gilewski T, Dang C, Chaim J,<br />

Patl S, Lim K, Norton L, Hudis CA, Fornier MN. Memorial Sloan-<br />

Kettering <strong>Cancer</strong> Center, New York<br />

Background: Src kinase plays an important role in proliferation,<br />

survival, angiogenesis and metastasis in several malignancies<br />

including breast cancer. Therefore, inhibition of Src and other<br />

tyrosine kinases (TKs) represents a novel therapeutic approach.<br />

Dasatinib is a potent inhibitor of 5 oncogenic TKs, inhibits VEGFstimulated<br />

proliferation, has potent bone anti-resorptive activity and<br />

selectively inhibits basal-type breast cancer growth. Preclinically,<br />

the combination of dasatinib and paclitaxel had superior antitumor<br />

activity to either agent alone. In a previous phase I study, we<br />

determined that, in combination with weekly paclitaxel, the optimum<br />

dose of dasatinib was 120mg. Of note, 4/9 (44%) patients treated at<br />

or above this dasatinib dose level had objective tumor response. We<br />

now present results from the phase II trial of this combination.<br />

Methods: Patients with MBC, ECOG PS 0-1, normal hepatic, renal,<br />

marrow function were eligible. Patients had measurable, HER2-<br />

negative metastatic breast cancer (MBC), ≤2 prior therapies for MBC.<br />

Treatment consisted of weekly paclitaxel 80 mg/m 2 IV 3/4 weeks +<br />

Dasatinib 120mg orally daily. Response was assessed by RECIST<br />

after every 8 weeks of therapy. Simon’s two-stage optimal design<br />

was used to test the null hypothesis of a 15% response rate (RR)<br />

against the alternative of a 30% RR. In stage I, planned enrollment<br />

was 23 patients based on Type I and Type II errors of 10%. If 4 or<br />

more responses are observed, enrollment will be extended to 55<br />

patients. Exploratory correlative biomarkers of clinical benefit include<br />

Src phosphorylation (p-Src) in peripheral blood mononuclear cells,<br />

plasma levels of VEGFR2 and collagen Type IV, circulating tumor<br />

cells (CTCs) and tumor gene expression profiling.<br />

Results: 21 patients (19 females, 2 male) have enrolled; median<br />

age 48 (range 30-79). Patients received a median of 1 prior therapy<br />

for MBC (range 0-2). 6 patients are not assessable for response: 1<br />

has received 15% of pts included anemia<br />

(45%), neutropenia (32%), nausea (51%), vomiting (32%) diarrhea<br />

(51%), fatigue (53%), anorexia (25%) dysgeusia (32%), headache<br />

(28%), peripheral neuropathy (34%) cough (28%) alopecia (21%)<br />

and hypokalemia (19%). Grade 3/4 adverse events occurred in 30<br />

patients, with only four experiencing grade 4 AEs, three neutropenia<br />

and one chest pain. No grade 3/4 febrile neutropenia occurred. Grade<br />

3 AEs of interest included two pts with peripheral neuropathy and 1<br />

with pleural effusion.<br />

Conclusions: The combination of Ixabepilone weekly + Dasatinib<br />

orally daily is an active and well tolerated regimen in pretreated MBC<br />

with manageable toxicity. Updated results will be reported.<br />

www.aacrjournals.org 481s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P5-20-09<br />

Tumor mutational analysis and therapy outcomes for patients<br />

(pts) with metastatic/unresectable locally advanced myoepithelial/<br />

metaplastic breast cancer treated with PI3K targeted therapy<br />

Moulder S, Wheler J, Albarracin C, Gilcrease M, Falchook G, Naing<br />

A, Hong D, Fu S, Piha-Paul S, Tsimberidou A, Janku F, Kurzrock R.<br />

University of Texas, MD Anderson <strong>Cancer</strong> Center<br />

Background: Metaplastic breast cancers are considered a<br />

chemorefractory subset of triple negative breast cancers. Molecular<br />

profiling has demonstrated that metaplastic tumors are enriched for<br />

epithelial-to-mesenchymal transition (EMT), frequently express<br />

myoepithelial differentiation, make up a component of the ‘claudinlow’<br />

subtype, and harbor relatively high rates of mutations/activation<br />

of the PI3kinase pathway (Hennessy, <strong>Cancer</strong> Research, 2009; Prat,<br />

<strong>Breast</strong> <strong>Cancer</strong> Res, 2010).<br />

Methods: Data from pts with myoepithelial/metaplastic breast cancer<br />

treated within the Center for Targeted Therapy using regimens with<br />

known inhibitors of the PI3K pathway were evaluated to determine<br />

response to therapy. Mutational analyses were performed in archived<br />

tumor samples when available.<br />

Results: 23 pts have been treated using 6 different therapy regimens<br />

and one pt was treated on two separate clinical trials for a total<br />

of 24 analyzable outcomes. Patients were treated with liposomal<br />

doxorubicin, bevacizumab and the mTOR inhibitor, temsirolimus<br />

(DAT, n=17); liposomal doxorubicin and temsirolimus (DT, n=1);<br />

paclitaxel, bevacizumab and temsirolimus (TAT, n=3); paclitaxel<br />

and temsirolimus (TT, n=1), paclitaxel in combination with an<br />

experimental PI3K inhibitor (TEx, n=1), or temsirolimus alone<br />

(tem, n=1). Response was measured every two cycles using RECIST<br />

criteria. Most pts had received prior chemotherapy, median of 2<br />

prior regimens (range 0-7). Three patients were not evaluated for<br />

response, one who died of pneumonia during cycle 2 (DAT) and<br />

two who have not yet completed 2 cycles of therapy (DAT, TEx).<br />

Response rate (CR+PR) was 35% (CR=2, PR=4, SD≥6 months=2,<br />

SD


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 5<br />

P5-20-11<br />

Bevacizumab in metastatic breast cancer: a retrospective<br />

matched-pair analysis.<br />

Gampenrieder SP, Romeder F, Muß C, Pircher M, Ressler S,<br />

Rinnerthaler G, Bartsch R, Sattlberger C, Mlineritsch B, Greil R.<br />

Paracelsus Medical University, Salzburg, Austria; Comprehensive<br />

<strong>Cancer</strong> Center Vienna, Medical University of Vienna, Austria;<br />

Hospital of Vöcklabruck, Vöcklabruck, Austria<br />

Background: In November 2011 the FDA withdrew accelerated<br />

approval of the breast cancer indication for bevacizumab, because “the<br />

drug has not been shown to be safe and effective for this use” (FDA<br />

Commissioner M. A. Hamburg, MD). The objective of this analysis<br />

was to evaluate efficacy and safety of bevacizumab in non-selected<br />

patients with stage IV breast cancer.<br />

Patients and methods: All patients with metastatic breast cancer<br />

treated with bevacizumab in combination with chemotherapy in our<br />

institution between 2005 and 2011 were retrospectively analysed.<br />

At least 1 cycle of bevacizumab was required for safety analysis, 3<br />

applications for efficacy evaluation. A control group was matched<br />

according to the following criteria: receptor status, line of treatment,<br />

chemotherapy backbone, visceral or non-visceral disease and age.<br />

Results: A total of 213 patients fulfilled the inclusion criteria. All<br />

of them were evaluable for toxicity, 199 for response. 503 potential<br />

controls allowed a complete matching for 103 patients. Fifty-eight<br />

percent of patients received first-line, 15% second-line and 27%<br />

of patients beyond second-line treatment. Visceral metastases<br />

were present in 69% of patients in both groups. The most common<br />

chemotherapy backbones were paclitaxel, capecitabine and docetaxel<br />

with 44%, 32% and 19%, respectively.<br />

Control group (n = 103) Bevacizumab group (n = 103) p-value<br />

PFS [mo] 6.0 (95 % CI 4.0-7.9) 8.9 (95 % CI 7.5-10.4) .009<br />

OS [mo] 24.4 (95 % CI 21.1-27.8) 25.4 (95 % CI 15.9-34.8) .498<br />

ORR (CR+PR) 37 (36%) 47 (46%), RR 1.26 (95% CI .88-1.80) .170<br />

CBR (ORR+SD) 68 (67%) 87 (85%), RR 1.27 (95% CI 1.07-1.47) .003<br />

Most common grade 3/4 toxicities related to bevacizumab were<br />

hypertension (27%), proteinuria (1%), thromboembolism (9%) and<br />

bleeding events (1%). Interestingly, patients developing hypertension<br />

during bevacizumab treatment had a more favourable outcome (PSF<br />

11.7 vs. 5.7 months, p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Methods: This is a single center, open-label study to evaluate the<br />

safety and tolerability of every other week entinostat in combination<br />

with a 28-day cycle of Lapatinib. Patients with metastatic breast<br />

cancer in whom trastuzumab has failed were included. The phase<br />

I portion of the study is a conventional 3+3 dose-escalation design.<br />

Dose levels include 0 (starting dose) Entinostat 10 mg orally every<br />

other week, I Entinostat 12 mg, and II Entinostat 15 mg. Lapatinib<br />

1,250 mg orally is given every day without dose escalation. Toxicities<br />

are evaluated at the end of each cycle.<br />

Results: Here we report the phase I portion of the study. To date, 9<br />

patients were enrolled, 3 were in level 0, and 6 were in level I. In Level<br />

0, 2 patients were taken off study due to disease progression (PD) at<br />

the end of cycle one and 1 patient was taken off study due to PD at<br />

the end of cycle two. In Level I, 1 patient was taken off study due to<br />

PD at the end of cycle one and 2 patients were taken off study due<br />

to PD at the end of cycle 2. 1 patient had stable disease. The median<br />

age is 41 (range, 26 – 69). Seven of the nine patients are evaluable for<br />

toxicity. Most common toxicities reported by the patients are nausea<br />

grade 3 (1), fatigue grade 3 (1), muscle aches/pain grade 2 (3), skin<br />

rash grade 3 (1), paresthesias grade 2 (2), heartburn grade 1 (4), and<br />

diarrhea Grade 2 (1). Lapatinib dose was reduced in 2 patients. The<br />

most common hematological toxicities were neutropenia grade 1 (3),<br />

anemia grade 2 (1), and thrombocytopenia grade 4 (1).<br />

Conclusions: Overall, patients have tolerated the combination<br />

regimen relatively well. We have not reached the maximum tolerated<br />

dose, so patient enrollment will continue until the phase I portion of<br />

the study is complete, most likely in July 2012. We plan to proceed<br />

with phase II portion in two parallel cohorts (HER2+ inflammatory<br />

and non-inflammatory metastatic breast cancer).<br />

P5-21-01<br />

pT1a,bpN0M0 breast cancer: clinicopathological characteristics<br />

and their impact on treatment decision. Central review of the<br />

prospective ODISSEE cohort<br />

Lacroix-Triki M, Radosevic-Robin N, Louis B, Roche-Comet I,<br />

Soubeyrand M-S, Bourgeois H, Chauvet M-P, Fourrier-Réglat A,<br />

Gligorov J, Peyrat J-P, Dalenc F, Belkacemi Y, Penault-Llorca F.<br />

Institut Claudius Regaud, Toulouse, France; Centre Jean Perrin,<br />

Clermont-Ferrand, France; Laboratoire d’Anatomie et Cytologie<br />

Pathologiques, Strasbourg, France; Institut Histo-Cyto-Pathologie,<br />

Le Bouscat, France; Cabinet CY-PATH, Villeurbanne, France;<br />

Clinique Victor Hugo, Le Mans, France; Centre Oscar Lambret,<br />

Lyon, France; Université Victor Segalen, Bordeaux, France; Hôpital<br />

Tenon, Paris, France; CHU Henri Mondor-UPEC, Créteil, France<br />

Background: The incidence of infra-centimetric breast cancer (BC)<br />

is increasing due to mass screening. The management remains<br />

controversial opposing locoregional treatment only based on low<br />

stage and potentially aggressive tumor biology (especially for triple<br />

negative and HER2 positive tumors). The objectives of the prospective<br />

multicenter ODISSEE study were i) to describe daily practice<br />

management of pT1ab BC, ii) to identify potential new biomarkers<br />

and iii) to describe long term outcome (10-year follow up).<br />

Methods: 616 patients with unifocal pT1a,b pN0M0 BC were included<br />

after surgery from May 2009 to March 2010 in 116 centers. Paraffin<br />

blocks were available for central pathology review in 350 patients.<br />

Due to the small tumor size, tissue microarray (TMA) construction<br />

was finally achieved for 287 cases. Collection of additional tumor<br />

blocks is ongoing. Review of the cases was performed independently<br />

by two pathologists and discordant cases were reviewed under a<br />

multihead microscope. Histological characteristics (histological<br />

type, tumor size and grade, presence of in situ component, presence<br />

of lymphovascular invasion) were assessed on whole tissue sections.<br />

Immunohistochemical detection of ER, PR, HER2, Ki67, EGFR,<br />

cytokeratin 5/6, Bcl-2 was performed on TMAs.<br />

Results: Clinicopathological characteristics of the 287 centrally<br />

reviewed cases were similar to those of the 616 patients included in<br />

the cohort. Median age: 60.1 years (range [31–89] years). Median<br />

tumor size: 7.8 mm (range [1–10] mm) with 11% pT1a and 89%<br />

pT1b. Histological types: 72% ductal, 11% lobular and 17% other<br />

types (among which 53% of tubular). Minor in situ component was<br />

found in 36% of the cases, and invasive carcinoma with an extensive<br />

in situ component in 1.9%. Most of tumors were histological grade I<br />

and II (52% and 40% respectively), 8% were grade III. Proliferation<br />

was low as assessed by mitotic count (low 82%, intermediate 13%<br />

and high 5%) or by Ki67 index (


December 4-8, 2012 <strong>Abstract</strong>s: Poster Session 5<br />

(63%), these HER2+ pts do not differ from the general BC population.<br />

Both hormone receptor types are positive in 47%, while ER+ only or<br />

PR+ only was detected in 13% and 3%, respectively. Adjuvant ET<br />

of any type, starting before or during T treatment, was given in 87%<br />

of HR+ and in 3% of HR- pts. 6% of HR+ pts received T and ET<br />

without concomitant CT. Details on the adjuvant treatment options<br />

are provided in the table:<br />

Concomitant adjuvant<br />

treatment<br />

HER2+/HR- HER2+/HR+ Total<br />

CT and ET 3% 81% 52%<br />

ET only 0% (4 pts) 6% 4%<br />

CT only 92% 12% 41%<br />

Trastuzumab single drug therapy 5% 1% 3%<br />

Anthracycline 87%* 88%* 88%*<br />

Taxane 66%* 63%* 64%*<br />

CT simultaneous to trastuzumab 16%* 16%* 16%*<br />

* of pts receiving CT<br />

After a median follow-up of 26.4 months and the observation of a<br />

total of 296 relapse events (9%; HR-: 12%; HR+: 7%) relapse-free<br />

survival (RFS) was significantly shorter in the HR- compared to the<br />

HR+ group (at 3 years: 86 vs 92%, hazard ratio = 0.56 , p < 0.0001).<br />

Conclusion: The vast majority of HER2+ BC pts receives concomitant<br />

endocrine therapy in case of a positive HR status. Regardless of the<br />

regulatory status in Germany, a small proportion of patients with<br />

co-positive early BC received T and ET without concomitant CT.<br />

No differences between receptor subgroups could be detected with<br />

respect to type of CT and its onset relative to T. HR status remains<br />

a paramount prognostic factor in this HER2+ sub-population of BC<br />

pts, with risk of early relapse (after a median follow-up of 2 years)<br />

almost twice as high in HR- tumors.<br />

P5-21-03<br />

Concurrent loco-regional radiotherapy and trastuzumab in<br />

early-stage breast cancer: Long term results of prospective<br />

single-institution study<br />

Jacob J, Belin L, Pierga J-Y, Vincent-Salomon A, Dendale R, Beuzeboc<br />

P, Cottu P-H, Campana F, Fourquet A, Kirova YM. Institut Curie,<br />

Paris, France<br />

Purpose: To evaluate the early and late heart and skin toxicities, as<br />

well as the efficacy of concurrent adjuvant trastuzumab-radiotherapy<br />

(RT) for breast cancer (BC), with group with internal mammary<br />

chain (IMC) RT.<br />

Material and methods: Prospective study of 308 patients (pts)<br />

treated between 2000 and 2009 by concurrent trastuzumab with<br />

normofractionated RT for non-metastatic BC. Left ventricular ejection<br />

fraction (LVEF) was assessed by echocardiography or myocardial<br />

scintigraphy at baseline, before and after RT, every three months<br />

for one year, then annually. A LVEF strictly lower than 55%, was<br />

considered as altered. Trastuzumab was delivered every three weeks<br />

(8 mg/kg in the first infusion then 6 mg/kg) during a median time of<br />

12 months (3-62). All toxicities were evaluated using the Common<br />

Terminology Criteria for Adverse Effects version 3.0. Survival data<br />

were defined as the time from the histological diagnosis of BC until<br />

occurrence of the event. Loco-regional recurrence free- and alive pts<br />

were censored at the date of their last known contact. Survival and<br />

interval rates as well as their confidence interval (CI) were calculated<br />

using the Kaplan–Meier method.<br />

Results: Median age was 52 years (25-83). Median follow-up was<br />

50 months (13-126). The clinical tumour staging was: T0 or T1 in<br />

131 pts (43.4%), T2 in 122 pts (40.4%), T3 or T4 in 49 pts (16.2%).<br />

The regional lymph nodes were histologically involved in 132 pts<br />

(43.0%). The left breast was treated in 155 pts (50.3%). The IMC<br />

was irradiated in 227 pts (73.7%), among them 114 (37.0%) in the<br />

left side. Before trastuzumab, anthracycline-based regimens were<br />

administered in 280 pts (90.9%). The median dose of trastuzumab<br />

was 6145 mg (1845-29180). Acute skin toxicity was acceptable with<br />

226 (74.1%) grade 1, 67 (22.0%) grade 2 and 12 (3.9%) grade 3 skin<br />

reactions. Esophagitis was observed in 31 pts (10.0%): 26 (8.4%)<br />

grade 1; 4 (1.3%) grade 2, and 1 (0.3%) grade 3. Out of 286 pts with<br />

assessments at the median time of 23 months, late telangiectasia grade<br />

1 occurred in 14 pts (4.9%) and grade 2 in 10 pts (3.5%), local pain<br />

grade 1 in 39 pts (13.7%) and grade 2 in 8 pts (2.8%), fibrosis grade<br />

1 in 53 pts (18.6%) and grade 2 in 20 pts (7.0%). At the completion<br />

of RT, 11 pts (3.6%), whose cardiologic assessment at baseline was<br />

normal, presented a clinically silent LVEF alteration. During followup,<br />

44 patients experienced cardio-vascular morbidity revealed by:<br />

asymptomatic LVEF alteration (50.0%), thrombo-embolic event<br />

(18.2%), tachycardia (15.9%), ischemic cardiomyopathy (6.8%),<br />

pericarditis (4.5%), hypertrophic cardiomyopathy (2.3%) or arterial<br />

hypertension (2.3%). The cumulative incidence of these events at 48<br />

months was 13.3% CI95% [9.4; 17.2]. A symptomatic congestive<br />

heart failure was reported for 3 pts (1.0%). No death of cardiac origin<br />

was observed. At 48 months, loco-regional control was 95% CI95%<br />

[92; 98] and overall survival was 98% CI95% [96; 100].<br />

Conclusions: In this prospective study of BC pts treated with<br />

trastuzumab and RT, both the rates of skin and esophageal toxicities<br />

were acceptable with excellent local control. Further follow-up is<br />

warranted to assess late cardiac toxicity.<br />

P5-21-04<br />

Real-world use and effectiveness of adjuvant trastuzumab in 2665<br />

consecutive breast cancer patients<br />

Tjan-Heijnen VCG, Seferina SC, Lobbezoo DJA, Voogd AC,<br />

Dercksen MW, van den Berkmortel F, van Kampen RJW, van de<br />

Wouw AJ, Joore MA, Borm GF. Maastricht University Medical<br />

Centre, Maastricht, Limburg, Netherlands; GROW-School for<br />

Oncology and Developmental Biology, Maastricht University<br />

Medical Centre, Maastricht, Limburg, Netherlands; Máxima Medical<br />

Centre, Veldhoven, Brabant, Netherlands; Atrium Medical Centre<br />

Parkstad, Heerlen, Limburg, Netherlands; Orbis Medical Center,<br />

Sittard-Geleen, Limburg, Netherlands; VieCuri Medical Centre,<br />

Venlo, Limburg, Netherlands; Radboud University Medical Center,<br />

Nijmegen, Gelderland, Netherlands<br />

Background The impact of drug prescriptions in the real world as<br />

opposed to strict trial prescription is only rarely assessed even though<br />

it is well recognized that incorrect use may harm a patient and may<br />

have a huge impact on health care resources. We aimed to assess<br />

how and to whom trastuzumab was given since its introduction in<br />

September 2005 as adjuvant treatment, and to assess its efficacy in<br />

daily practice.<br />

Patient and methods We set up a registry of all patients diagnosed<br />

with stage I-III invasive breast cancer in 5 Dutch hospitals during the<br />

years 2005-2007. Patients were included irrespective of HER2 status<br />

and of treatment received. We present the main patient and tumor<br />

characteristics, treatment delivered and 5-year treatment outcomes.<br />

Results Of 2665 patients included, n=479 (18%) had a HER2 positive<br />

tumor. As of September 2005, adjuvant trastuzumab was given to<br />

96% of patients with a HER2 positive tumor receiving adjuvant<br />

chemotherapy. The median age of these patients was 51 (27 - 72)<br />

years. Their median tumor size was 23 (5 - 65) mm, 55% had nodepositive,<br />

57% ER-positive and 47% PR-positive disease. In 83% of<br />

patients, trastuzumab was delivered in sequence after (taxane-based)<br />

chemotherapy, mainly in the first years after introduction, and in 91%<br />

in a 3-weekly regimen. Trastuzumab was delivered for a median of<br />

49 (IQR 45 - 52) weeks. For patients with a HER2 positive tumor<br />

www.aacrjournals.org 485s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

treated with adjuvant trastuzumab (n=232), the five-year disease-free<br />

survival was 81%. For patients with a HER2 positive tumor who<br />

were not treated with trastuzumab (n=247) the five-year disease-free<br />

survival was 75% (p=0.13). Corresponding 5-year overall survival<br />

rates were 91% and 78%, respectively (p=0.0002).<br />

Conclusion In the Netherlands, trastuzumab was rapidly implemented<br />

since its introduction in September 2005. Characteristics of patients<br />

treated with trastuzumab resembled those included in the HERA study.<br />

Similarly, outcomes were in agreement with reported phase III studies.<br />

More detailed and multivariate analyses will be shown at the meeting.<br />

Funding Netherlands Organization for Health Research and<br />

Development (ZonMw: 80-82500-98-9056) and Roche Netherlands.<br />

P6-01-01<br />

Metastatic and non-metastatic isogenic breast cancer cells lines<br />

show different metabolic signatures in response to intermittent<br />

hypoxia and transient glutamine/glucose deprivation<br />

Simoes RV, Ackerstaff E, Serganova I, Kruchevsky N, Sukenick G,<br />

Blasberg R, Koutcher JA. Memorial Sloan-Kettering <strong>Cancer</strong> Center,<br />

New York, NY; Cornell University, New York, NY<br />

INTRODUCTION. <strong>Cancer</strong> cells quickly adapt to their<br />

microenvironment by reprogramming their metabolism. Specific<br />

metabolic responses to defined stress conditions, mimicking tumor<br />

microenvironmental changes, may therefore unveil signatures of<br />

cancer phenotype, not detected in standard tissue culture.<br />

PURPOSE. To identify distinct metabolic signatures associated with<br />

metastatic ability, we investigated in the two isogenic breast cancer<br />

cell lines, 4T1 (highly metastatic) and 67NR (non-metastatic), how<br />

glutamine supply/deprivation affects cellular glucose metabolism,<br />

and vice-versa, in aerobic or hypoxic conditions, and the effects on<br />

cell growth.<br />

METHODS. 4T1 and 67NR cells were cultured in medium A, i.e.<br />

DME containing 1% P/S, 10% FBS, 25 mM glucose (Glc), and 6 mM<br />

glutamine (Gln). Cell growth was determined in tissue culture from<br />

cells cultured for 48 h in various media A-D: A, all nutrients available;<br />

B: A but no Gln; C: A but 2 mM Gln; D: A but no Glc. Cellular<br />

mitochondrial function was also assessed in both 4T1 and 67NR cells<br />

using an XF96 Analyzer (Sea Horse Bioscience, Billerica, MA) and<br />

the specific inhibitors: oligomycin, FCCP, antimycin and rotenone.<br />

Metabolism was studied in cells grown on microcarriers in the various<br />

media A-D under aerobic (∼21% O2) and hypoxic conditions (∼1%<br />

O2), using our magnetic resonance (MR)-compatible cell perfusion<br />

system on a 500 MHz spectrometer. For the cell perfusion studies,<br />

Glc or Gln were exchanged for 1-13C-Glc or 3-13C-Gln respectively.<br />

The metabolic fate of 13C-labeled nutrients was followed under the<br />

various environmental conditions by 13C MR spectroscopy (MRS),<br />

while energy metabolism was observed by 31P MRS.<br />

RESULTS. Deprivation of either Glc or Gln for 48 h reduced<br />

significantly the cell growth of 4T1 and 67NR cells, resulting in a<br />

similar growth rate for the nutrient-deprived 4T1 and 67NR cells.<br />

As determined from the MR experiments, each cell line adopted<br />

different metabolic strategies to cope when exposed to specific<br />

metabolite stress conditions, including on the level of oxygen<br />

availability. Unlike in 67NR cells, glycolysis is glutamine-dependent<br />

in the more aggressive 4T1 cell line (∼2-fold higher glucose-derived<br />

lactate synthesis rate was observed in the presence of glutamine),<br />

which decreases when oxygen becomes available to fuel TCA cycle<br />

activity, as monitored by glucose-derived glutamate synthesis. The<br />

studies further indicate an impaired TCA cycle activity in 67NR<br />

cells, reflected in a high accumulation rate of glucose- or glutaminederived<br />

succinate (significantly higher compared to 4T1 cells). This is<br />

consistent with the mitochondrial functional analysis, showing higher<br />

mitochondrial TCA activity in 4T1 cells than in 67NR, most likely<br />

due to impairment of succinate dehydrogenase (SDH, respiratory<br />

Complex II) in the latter.<br />

CONCLUSIONS. Our results support the association between<br />

increased mitochondrial metabolism and metastatic potential,<br />

observed recently. We are investigating the SDH status in these cells<br />

lines and in additional metastatic and non-metastatic breast cancer<br />

cell lines, to establish whether SDH, and glutamine metabolism, could<br />

be potential targets for therapy.<br />

P6-01-02<br />

Adipose tissue from breast cancer patients with the metabolic<br />

syndrome promotes proliferation and invasion of tumor cells<br />

and influences expression of genes involved in carcinogenesis.<br />

McGarrigle SA, Carroll PA, Healy LA, Boyle T, Pidgeon GP, Kennedy<br />

MJ, Connolly EM. Trinity College Dublin and St. James’s Hospital,<br />

Dublin, Ireland<br />

Background<br />

Obesity is a public health issue of global proportions and is recognised<br />

as a risk factor for post-menopausal breast cancer. Similarly, the<br />

metabolic syndrome (MetS) is recognised as a high risk state for<br />

cancer in general. Previously we have shown that the MetS is common<br />

in postmenopausal breast cancer patients and is associated with a more<br />

aggressive tumor biology. However, the molecular mechanisms by<br />

which obesity and/or the MetS promote breast cancer remain unclear.<br />

Adipose tissue, including mammary fat, is a functionally active<br />

endocrine organ. The aims of this study were to determine whether<br />

factors secreted by mammary adipose tissue could affect tumor cell<br />

biology and to assess the effect of the MetS on this adipose depot<br />

and its subsequent effect on tumor cells.<br />

Methods<br />

Adipose tissue from fresh mastectomy specimens was cultured in<br />

serum free media for 72 h to produce adipose conditioned media<br />

(ACM). MCF-7 and MDA-MB 231 cell lines were treated with ACM<br />

for 24-48 h. Tumor cell function was then assessed by measuring cell<br />

proliferation (BrDU assay) and cell invasion. In addition, expression<br />

of 84 genes implicated in pathways involved in carcinogenesis<br />

was examined in these cell lines following ACM treatment, using<br />

quantitative PCR arrays.<br />

Results<br />

In the estrogen receptor (ER) positive MCF-7 cell line, ACM<br />

from MetS breast cancer patients promoted significantly greater<br />

proliferation compared to ACM from normal weight patients (203.6<br />

± 34.23 vs 136.8 ± 11.58%, p =0.022). Similarly, ACM from MetS<br />

patients significantly increased invasion of MCF-7 cells compared to<br />

ACM from normal weight patients (153.4 ± 6.027 vs 126.3 ± 6.03%<br />

RFU, p =0.006). No differences in cell proliferation or invasion<br />

between cells treated with ACM from MetS patients compared to<br />

ACM from normal weight patients were found in the ER negative<br />

MDA-MB-231 cell line. Treatment of MCF-7 cells with ACM from<br />

MetS patients resulted in significant alterations (>2 fold up/down<br />

regulation) in expression of 11 genes involved in carcinogenesis.<br />

Primarily, genes implicated in invasion/metastasis and adhesion<br />

were differentially expressed between ACM-treated cells and cells<br />

treated with control media. On the other hand, when MDA-MB-231<br />

cells were treated with ACM from the same patients only one gene,<br />

SERPIN B5 was significantly up-regulated > 2 fold.<br />

Conclusions<br />

These data demonstrate that factors secreted from mammary adipose<br />

tissue from metabolically unhealthy patients promote proliferation<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

486s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: General Session 6<br />

and invasion of ER positive tumor cells and influence expression of<br />

genes involved in carcinogenesis in these cells. These effects were<br />

not observed in ER negative tumor cells suggesting that they may<br />

be mediated, at least in part by the estrogen receptor. These results<br />

have provided insight into how mammary adipose tissue may act via<br />

a paracrine mechanism to influence aspects of carcinogenesis and into<br />

how the metabolic syndrome may modulate this.<br />

P6-01-03<br />

Decreasing the metastatic potential in Triple Negative <strong>Breast</strong><br />

<strong>Cancer</strong> through the miR-17 cluster<br />

Jin L, Simone B, <strong>San</strong>o Y, Lim M, Zhao S, Savage JE, Baserga<br />

R, Camphausen K, Simone NL. Thomas Jefferson University,<br />

Philadelphia, PA; National <strong>Cancer</strong> Institute, Bethesda, MD<br />

Background: In animal models of breast cancer, caloric restriction<br />

(CR), or a 25-30% reduction of calories, has decreased tumor initiation<br />

and progression. We sought to determine if CR could be used as a<br />

novel therapeutic intervention to enhance cytotoxic therapies such as<br />

radiation (RT) and alter the molecular profile of triple negative breast<br />

cancer (TNBC), which displays a poor prognosis.<br />

Methods/Results: In two murine models of TNBC (4T1 which is<br />

highly metastatic and 67NR which is locally aggressive), mice with<br />

palpable tumors were treated with radiation, caloric restriction (30%<br />

intake reduction) or both. 3 times per week measurements revealed<br />

significant tumor regression with RT or diet modification, and an even<br />

greater regression is observed combining diet modification with RT.<br />

Concurrently, many reports have shown that TNBC is most distinct<br />

from other breast tumor subtypes due to up-regulation of the miR-<br />

17-92 cluster which we have shown by RT-PCR is downregulated<br />

with both CR and radiation (RT) and even further decreased with<br />

the combination. cDNA arrays done on all conditions revealed that<br />

the top 5 statistically significant GO terms with high fold changes<br />

were all associated with extracellular matrix and metastases. In silico<br />

analysis demonstrated 5 ECM related genes that were targets of the<br />

miR-17 cluster: collagen 4 alpha 1 (COL4A1), collagen 4 alpha 3<br />

(COL4A3), laminin alpha 3 (LAMA3), metallopeptidase inhibitor<br />

2 and 3 (TIMP2 and TIMP3) and these were inversely correlated<br />

with the miRs.<br />

Conclusions: CR, or altering the metabolism of breast cancer, can<br />

be combined with RT to decrease progression of TNBC tumors and<br />

that the miR-17-92 cluster is also decreased with these conditions.<br />

cDNA arrays suggest that extracellular matrix (ECM) proteins play<br />

a large role in the mechanism of action of CR and RT combined.<br />

Combination therapy with CR and RT can decrease the metastatic<br />

potential in TNBC and in part does this through the miR-17-92 cluster<br />

by way of controlling ECM.<br />

P6-01-04<br />

DDB2 a new regulator of metabolism and cell death in human<br />

breast tumor cells<br />

Klotz R, Besancenot V, Brunner E, Becuwe P, Grandemange S.<br />

Université de Lorraine, Vandoeuvre les Nancy, France<br />

The protein Damaged DNA Binding-2 (DDB2) is well known for<br />

its role in DNA repair by nucleotide excision repair. Interestingly,<br />

DDB2 is differentially expressed in breast cancer expressing the<br />

estrogen receptor alpha or not. Recent works performed in our<br />

laboratory showed a new role of DDB2 in the control of proliferation<br />

and invasive abilities in different breast tumor cells through its<br />

involvement in the transcriptional regulation of target genes. Two<br />

genes involved in tumorigenic processes, MnSOD (manganese<br />

superoxide dismutase) and IκBα (inhibitor alpha of Nuclear Factorkappa<br />

B), have been found to be regulated by DDB2. In addition,<br />

transcriptomic analyses on cells that differentially express DDB2<br />

showed that several genes involved in the regulation of cellular<br />

metabolism are modulated. Our aim is now to focus on the effects of<br />

DDB2 expression on cellular metabolism and glycolysis. The results<br />

indicated that the overexpression of DDB2 leads to a respiratory chain<br />

dysfunction and an increase of the glycolytic pathway. Moreover,<br />

we observed an increased production of reactive oxygen species in<br />

these cells, compared to parental cells. As mitochondria are involved<br />

in cell death, we performed different experiments to evaluate the<br />

impact of DDB2 in the response to anticancer agents (Doxorubicin,<br />

and 5-fluorouracile (5-FU)) commonly used in the treatment of breast<br />

cancer. Interestingly, the cells exhibit a greater sensitivity to anticancer<br />

drugs when DDB2 is overexpressed. As these two agents are related<br />

to DNA damaged, we have also used other molecules, the apoptotic<br />

inducer, TNFα (Tumor Necrosis Factor alpha), and the Paclitaxel<br />

(an antimicrotubule agent) to precise the role of DDB2 on cell death.<br />

Similar results were obtained, thus demonstrating the influence of<br />

DDB2 overexpression in the response to cell death. The identification<br />

of molecular mechanisms responsible for these cellular modifications<br />

could place DDB2 and these target genes as predictive markers of<br />

sensitivity to anticancer drugs in breast cancer.<br />

P6-01-05<br />

Enhancement of 18 F-FDG uptake and glycolysis by epidermal<br />

growth factor via PI3K activation in T47D breast cancer cells<br />

Lee EJ, Park JW, Cung Q, Jung K-H, Lee JH, Paik J-Y, Lee K-H.<br />

Samsung Medical Center, Sungkyunkwan University School of<br />

Medicine, Seoul, Korea<br />

It is well known that the binding of epidermal growth factor (EGF)<br />

to EGF receptors (EGFR) stimulates proliferation of breast cancer<br />

cells via activation of mitogen-activated protein kinase (MAPK) and<br />

phosphatidylinositol 3-kinase (PI3K)-protein kinase B (Akt) signaling<br />

pathways. There was a report that glucose consumption and lactate<br />

production were increased in MDA-MB-468 breast cancer cells<br />

following EGF exposure. However, the effect of EGF in enhancing<br />

glucose uptake and signaling pathways that are involved have not been<br />

clearly revealed. We thus investigated the role of EGF in stimulation<br />

of 18 F-FDG uptake in T47D breast cancer cells, and further elucidated<br />

the molecular mechanisms that are involved.<br />

Exposure of T47D cells to 100 ng/mL EGF for 24 h caused a<br />

substantial augmentation of 18 F-FDG uptake to even greater extents<br />

to 310.8% ± 30.1% of control cells (P < 0.001), and this effect<br />

of EGF showed dose- and time-dependency. Increased glucose<br />

uptake by EGF occurred through enhanced membrane GLUT-1<br />

expression as well as hexokinase activity. Inhibition experiments with<br />

cycloheximide showed that the metabolic effect by EGF required<br />

new protein biosynthesis. Stimulation of glucose uptake by EGF<br />

was dependent on sufficient EGFR expression because 18 F-FDG<br />

uptake in MCF-7 cells weakly expressing EGFR was not affected<br />

by EGF treatment. Although EGF was also a stimulator of T47D<br />

cell proliferation (137.3 ± 7.3% of basal levels at 24 h, P < 0.001),<br />

the metabolic effect occurred in a manner that clearly preceded and<br />

surpassed the proliferative effect. EGF-stimulated proliferation was<br />

significantly inhibited by the EGFR inhibitor BIBX1382, the PI3K<br />

inhibitor LY294002 and the specific MAPK inhibitor PD98059.<br />

Unlike proliferation, the ability of EGF to augment 18 F-FDG uptake<br />

was found to be dependent on PI3K but not on MAPK activity.<br />

These findings yield the insight into our understanding of the role of<br />

EGF and the molecular basis that are involved in glucose metabolism<br />

of breast cancer cells.<br />

www.aacrjournals.org 487s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P6-02-01<br />

Apoptotic cell clearance lies at the interface of post-lactational<br />

involution and breast cancer<br />

Cook RS, Stanford JC, Earp S. Vanderbilt University; University of<br />

North Carolina<br />

Background. Cytokines that promote wound healing and immune<br />

tolerance are profoundly upregulated during post-lactational<br />

involution of the breast and promote tumor growth and malignancy.<br />

This observation is supported by increasing evidence that postlactational<br />

involution produces a highly malignant microenvironment<br />

contributing to poor outcome of pregnancy associated breast cancers<br />

(PABCs) which arise within 5 years of a full-term pregnancy. We<br />

have discovered that one mechanism driving cytokine modulation<br />

during post-lactational involution is efferocytosis, or the phagocytic<br />

clearance of apoptotic cells (ACs). Cell death is a prominent feature<br />

of the post-lactational breast, when up to 90% of the entire epithelial<br />

content dies and must be removed, making this an ideal model to study<br />

both apoptosis and AC clearance. Without efferocytosis, residual ACs<br />

undergo secondary necrosis, which stimulates acute inflammation<br />

and expression of Th1 cytokines. Thus, efferocytosis prevents<br />

secondary necrosis of dying cells, enhances immune tolerance and<br />

wound healing through production of Th2 cytokines. In healthy postlactational<br />

breast tissue this is an optimal physiological response that<br />

promotes breast remodeling. However, in the context of breast cancer,<br />

efferocytosis may be pro-tumorigenic.<br />

Results. We have shown that post-lactational efferocytosis is<br />

necessary to re-establish breast tissue homeostasis after lactation, and<br />

for re-initiating successful lactation after future pregnancies. We have<br />

also demonstrated that during the earliest phases of post-lactational<br />

involution, the breast epithelium is the primary phagocytic resource<br />

in the breast, although it is clear that macrophages engulf dying cells<br />

during later stages of involution. We have identified the receptor<br />

tyrosine kinase MerTK as a necessary mediator of efferocytosis and<br />

subsequent cytokine modulation during post-lactational involution.<br />

Further, MerTK-mediated cytokine changes in the post-lactational<br />

mammary microenvironment promotes malignant progression of<br />

spontaneously forming PABC in a transgenic mouse model, as<br />

genetic targeting of MerTK decreased tumor growth and metastasis<br />

without altering tumor latency. Post-lactational tumors expressed<br />

abundant Th2 and wound healing cytokines as compared to tumors<br />

in nulliparous mice. However, loss of MerTK in post-lactational<br />

mammary tumors impaired the expression of Th2 and wound healing<br />

cytokines, while inducing the expression of Th1 cytokines.<br />

Conclusion. These results suggest that efferocytosis-mediated<br />

cytokine regulation is a key driver of malignant severity in pregnancy<br />

associated mammary tumors. These data are consistent with our<br />

previous demonstrations that MerTK in the mammary tumor<br />

microenvironment increases tumor malignancy. Targeted inhibition<br />

of MerTK may decrease efferocytosis-mediated cytokine modulation<br />

in breast tumors to limit pro-tumorigenic cytokine production<br />

and stimulate production of cytokines associated with anti-tumor<br />

immunity.<br />

P6-02-02<br />

Altered matrix homeostasis regulates estrogen biosynthesis in<br />

adipose tissue<br />

Ghosh S, Ashcraft K, Li R. UT Health Science Center at <strong>San</strong> <strong>Antonio</strong>,<br />

TX<br />

Several estrogen-dependent pathological conditions including breast<br />

cancer, uterine fibroids, and endometriosis are associated with local<br />

overexpression of aromatase, a key enzyme in estrogen biosynthesis<br />

and important therapeutic target for postmenopausal ER+ breast<br />

cancer. In addition, excessive aromatase expression in adipose tissue<br />

at least partially accounts for obesity-associated breast cancer risk<br />

among postmenopausal women. While altered matrix homeostasis<br />

is associated with these aromatase-overexpressing tissues, little is<br />

known as to whether it can directly impact local steroidogenic gene<br />

expression and estrogen biosynthesis. A paucity of knowledge in<br />

this area is partly due to the lack of proper model systems that can<br />

recapitulate the mechanical properties of a cell’s microenvironment.<br />

Our data indicate that matrix alone can significantly stimulate<br />

aromatase transcription in breast stromal cells (BSCs) via distinct<br />

signaling pathways. With the help of a repertoire of molecular and<br />

biophysical tools, such as micropost, micropattern and polyacrylamide<br />

gel and 3-dimensional matrix culture, we determined that matrix<br />

rigidity and cellular shape play important role in the regulation of<br />

aromatase transcription in the BSCs. We further discovered that<br />

cytoskeleton dynamics can significantly alter the ECM-stimulated<br />

aromatase transcription in BSCs. Following careful and elaborate<br />

studies with the help of siRNA-mediated knockdown and overexpression<br />

of the individual factors, we found that a non-canonical<br />

collagen receptor member of discoidin domain receptor (DDR) family,<br />

DDR1, transduce the external signal inside the cells to facilitate<br />

the aromatase transcription. Further studies with siRNA-mediated<br />

knockdown and specific inhibitors revealed that DDR1 activates a<br />

signaling cascade, which promotes activation of the cJun kinase (JNK-<br />

1) by phosphorylation and subsequently the activation of Activation<br />

Protein-1 family member JunB. Activated JunB physically binds at<br />

the cancer-specific promoters of aromatase and induce transcription<br />

in stromal cells. To determine the consequence of the altered matrix<br />

generated aromatase on the growth of estrogen receptor (ER)-<br />

positive breast cancer cell lines we set up in vitro co-culture system.<br />

Proliferation of ZR75-1 and its ability to produce estrogen-stimulated<br />

genes were enhanced in multiple fold, when the cells were co-cultured<br />

physically with BSCs inside collagen or fed with BSC conditioned<br />

media in the presence of testosterone.<br />

The link between matrix homeostasis and hormone metabolism is<br />

a vastly under-explored topic. Results from our work has bridged a<br />

major gap of knowledge in this field. Findings from the study also<br />

identified new factors that are key for the tissue-specific activation<br />

of estrogen biosynthesis and provide novel prognostic tools and<br />

markers, as well as new therapeutic targets for reducing local estrogen<br />

production, thus overcoming the side effects often associated with<br />

systemic inhibition of aromatase. The proposed work promises to<br />

establish a novel paradigm for regulation of steroidogenic gene<br />

expression and estrogen production and may have a far-reaching<br />

impact on the etiology and treatment of endocrine diseases that are<br />

associated with altered matrix homeostasis.<br />

P6-02-03<br />

Mouse Models Of <strong>Breast</strong> <strong>Cancer</strong> Identify Oncogene-Specific<br />

Stroma Associated With Human <strong>Breast</strong> <strong>Cancer</strong> Molecular<br />

Subtypes<br />

Saleh SM, Laferriere J, Cory S, Souleimanova M, Zacksenhaus E,<br />

Muller W, Hallett M, Park M. McGill University, Montreal, QC,<br />

Canada; Toronto General Hospital, Toronto, ON, Canada<br />

<strong>Breast</strong> <strong>Cancer</strong> is a highly heterogeneous disease that involves a<br />

complex cross talk between the tumor cells and their surrounding<br />

microenvironment. Previous work in our lab has utilized laser capture<br />

microdissection (LCM) to isolate breast tumor epithelium as well as<br />

tumor associated stroma cells. These isolated cell populations were<br />

subjected to gene expression analysis, which separated the stroma<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

488s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: General Session 6<br />

samples into distinct subtypes. Pairing these stroma subtypes with<br />

genomic subtypes resulted in pairs having different outcomes hinting<br />

at the importance of these interactions (Finak et al, In preparation).<br />

Although this and other data now support a role for stroma in tumor<br />

progression, it is still unclear how these distinct differences in the<br />

stroma are induced.<br />

We hypothesized that specific changes within stroma are related to<br />

the oncogenic event occurring in the tumor epithelium. The use of<br />

human data for analysis generates correlative conclusions due to<br />

compounding factors such as genetic variance and living conditions.<br />

In an attempt to get more causative results, we used transgenic mouse<br />

models of breast cancer within the same strain (FVB) and where<br />

the gene is driven by the same promoter (MMTV), changing only<br />

the oncogene: Wnt1, NeuNDL2-5, PyVMT and activated Met. This<br />

allowed us to link differences observed in the stroma back to the<br />

epithelial oncogenic change.<br />

Through the use of LCM, linear amplification of the RNA and<br />

hybridization to Agilent microarrays, we generated a combined dataset<br />

of 33 samples including adjacent tumor epithelial, tumor stroma,<br />

in addition to pooled whole tumour samples and a pooled normal<br />

mammary fat pad sample, as control tissue. Unsupervised clustering<br />

of the most variable genes confirmed that the LCM isolated epithelial<br />

and stroma compartments specifically.<br />

Hierarchal clustering of the most variable genes was also used to<br />

determine the number of stroma groups. This approach revealed that<br />

there were 4 oncogene associated stroma groups, confirming that each<br />

of the oncogenes generated distinct stroma subtypes.<br />

Using the Rank Product package to compare each stroma group<br />

against the others identified over 1000 differentially expressed<br />

genes defining the stroma subtypes. Pathway analysis of these genes<br />

identified differences in both cell recruitment and pathway activation.<br />

To understand if these stroma classes were relevant to the human<br />

disease, the mouse stromal genes were used to order a large human<br />

whole tumour dataset using the BreSAT ordering (Lesurf et al, In<br />

preparation). This showed that the mouse models shared similarities<br />

with human samples from different molecular subtypes.<br />

P6-02-04<br />

TMEM (Tumor MicroEnvironment of Metastasis) in human<br />

breast cancer is a blood vessel associated intravasation<br />

microenvironment unrelated to lymphatics<br />

Ginter PS, Robinson BD, D’Alfonso TM, Oktay MH, Gertler FB,<br />

Rohan TE, Condeelis JS, Jones JG. Weill Cornell Medical College,<br />

New York, NY; Albert Einstein College of Medicine, Bronx, NY;<br />

Massachusetts Institute of Technology, Cambridge, MA<br />

In breast cancer, both lymph node and distant metastasis represent<br />

dissemination of tumor cells from a primary site, but the mechanism<br />

of spread and the subsequent risk of mortality may not be the<br />

same. Historically, lymphatic spread has been documented both<br />

descriptively, as presence or absence of lymphovascular invasion<br />

(LVI), and as a formal part of TNM staging. Until recently, however,<br />

there has been no way to directly assess the risk of hematogenous<br />

dissemination by the primary tumor.<br />

Observations from multiphoton-based intravital imaging of rodent<br />

models of breast cancer and the analysis of Mena function in tumor<br />

cells in vivo have characterized an intravasation microenvironment<br />

(ME) involved in the systemic dissemination of tumor cells from<br />

primary breast tumors. We have identified the corresponding structure<br />

in FFPE tissue and called it TMEM (Tumor MicroEnvironment of<br />

Metastasis). This microanatomic landmark is defined as the direct<br />

apposition of a Mena-overexpressing intravasation competent<br />

carcinoma cell, a perivascular macrophage, and an endothelial cell.<br />

In a case control study of 30 case-control pairs, where each matched<br />

pair differed only in their metastatic status – non-metastatic vs.<br />

metastatic – we found that the density of TMEM was significantly<br />

associated with development of systemic metastasis (p = 0.00006).<br />

The relationship of hematogenous- and lymphatic-mediated tumor<br />

cell spread is not understood. Using the previously described cohort<br />

in which we showed that TMEM was associated with metastasis,<br />

the purpose of this study was to 1) assess intratumoral lymphatic<br />

density, 2) determine if TMEM- lymphatic structures associated with<br />

lymphatics exist, and 3) determine if TMEM- lymphatic structures<br />

correlate with systemic metastatic risk. Cases were stained with a<br />

triple immunostain identical to that used in our earlier study except<br />

that D2-40 (a lymphatic marker) was used, rather than CD31 (a blood<br />

vessel marker). The marker for macrophages (CD68) and invasive<br />

tumor cells (Mena) remained the same. Two pathologists, blinded to<br />

outcome, evaluated the presence or absence of intratumoral lymphatics<br />

and quantitated the number of TMEM-lymphatic structures per 10<br />

high power (400x) fields in areas of highest intratumoral lymphatic<br />

density. A TMEM-lymphatic structure was defined as the direct<br />

apposition of a lymphatic (D2-40) endothelial cell with a macrophage<br />

and invasive tumor cell.<br />

Intratumoral lymphatics were absent in a majority of tumors in each<br />

of the 2 groups (18 of 30 non-metastatic, 16 of 30 metastatic; p=0.6).<br />

TMEM-lymphatic structures were rare and were equally present in<br />

the 2 groups (3 metastatic and 3 non-metastatic cases). Using the<br />

Wilcoxon (paired) signed-rank test, we found no significant difference<br />

in the density of these structures between the two groups (p = 0.4).<br />

Furthermore, TMEM-lymphatic structures did not correlate with<br />

the presence of lymph node metastases (p=0.8). We conclude that<br />

lymphatic vessels do not participate in the TMEM assembly that<br />

has been associated with hematogenous metastasis. TMEM density<br />

assessment reflects a hematogenous intravasation ME and offers a<br />

novel approach to the assessment of metastatic risk.<br />

P6-02-05<br />

A novel culturing 3-D model to evaluate the role of tumor<br />

microenvironment in IBC.<br />

Dong X, Franco-Barraza J, Mu Z, Alpaugh RK, Cristofanilli M,<br />

Cukierman E. Fox Chase <strong>Cancer</strong> Center, Philadelphia, PA<br />

Introduction: Inflammatory breast cancer (IBC) is a highly aggressive<br />

form of breast cancer associated with extremely poor outcomes.<br />

The clinical and pathological characteristics of the disease are the<br />

peculiar invasion of the dermal lymphatics as tumor emboli and the<br />

development of early recurrences. We aimed to establish a 3D model<br />

to evaluate the role of tumor microenvironment.<br />

Methods: We used human tumor-associated fibroblasts (or fibroblasts<br />

derived from metastatic skin) from IBC patients to build a multilayer<br />

extracellular matrix structure which effectively mimics aspects of<br />

the mesenchymal microenvironment of IBCs. Using this in vivo-like<br />

microenvironment we proceeded to test both matrix effects upon<br />

IBC’s phenotypes and IBC modifications upon the cell-derived 3D<br />

matrices.<br />

We seeded the IBC cells into the matrix and cultured for 3 days,<br />

then tested the characterization markers cancer cells and ECM<br />

e.g.Phalloidin, E-cadherin, Ki67, α5β1 integrin and fibronectin by<br />

immunofluorescence and the expression of E-cadherin and vimentin as<br />

marker of epithelial-mesenchymal transition (EMT) by western blot.<br />

Results: We divided six IBC cell lines into 2 groups depending<br />

on the phenotypes acquired when cultured in the IBC fibroblastderived<br />

ECM. SUM149 (EGF receptor positive and aggressive<br />

www.aacrjournals.org 489s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

phenotype), BR016 and LG018 (harvested from patient’s pleural<br />

effusions) presented a single cell organization with a spindle-like or<br />

mesenchymal type (as opposed to cluster) morphology. In comparison,<br />

SUM190 (HER2 positive and non aggressive tumorigenesis),<br />

MDA-IBC-3 and FC-IBC-02 (abstracted from patient’s pleural<br />

effusion) presented a phenotype resembling mammospheres or in<br />

vivo emboli. Moreover, this last group of cells showed a peculiar<br />

capability for ECM modifications which greatly differed from the<br />

ECM modifications that were apparent following 3 day culturing of<br />

the above mentioned group represented by SUM149. In addition,<br />

proliferation measurements by Ki67 expression demonstrated a<br />

significant increased in 3D culture for SUM149, BR016 and LG018<br />

compared with that in 2D culture, while no differences in proliferation<br />

were observed in the other three cell lines. Moreover, the expression<br />

of E-cadherin known to be upregulated in IBC tumors was increased<br />

in all cancer cells when seeded into the human fibroblast-derived<br />

3D matrix indicating a potential role of the microenvironment in<br />

promoting proliferation, growth and invasion.<br />

Conclusion: The present study demonstrated the establishment of<br />

a novel IBC stromal 3D model using extracellular matrix produced<br />

from human fibroblasts of patients with advanced IBC. We showed a<br />

dynamic interaction between cancer cells and the microenvironment<br />

and potential sorting of IBC cells into two discrete groups which also<br />

correlate with their aggressive behaviors in vivo. We believe that these<br />

system may serve to predict levels of IBC tumorigenesis. We will<br />

proceed to further study the two identified responsive phenotypes with<br />

the goal of uncovering mechanisms of IBC tumor-stromal interactions<br />

and better understand ECM influences upon IBC development and<br />

progression. The ultimate goal will be to use the system to study IBC<br />

biology and better design drugs that will specifically affect the newly<br />

identified phenotypes.<br />

P6-02-06<br />

A 3D tri-culture model of normal mammary gland. A tool for<br />

breast cancer initiation studies<br />

Nash CE, Holliday DL, Mavria G, Tomlinson DC, Hanby AM, Speirs<br />

V. Leeds Institute of Molecular Medicine, The University of Leeds,<br />

Leeds, West Yorkshire, United Kingdom<br />

The mechanisms involved in breast cancer initiation are not well<br />

understood. This may in part be due to a lack of an in vitro model<br />

that faithfully recapitulates the morphology, phenotype and in vivo<br />

architecture of the human mammary gland. Most in vitro models<br />

of normal breast have relied on the use of reconstituted basement<br />

membrane gels to induce luminal epithelial cell polarity and have<br />

neglected the role of myoepithelial cells and fibroblasts in this process.<br />

We aimed to develop a 3D in vitro culture system of normal breast<br />

which includes three of the major functional cell types embedded<br />

in a more physiologically relevant collagen 1 matrix. We used this<br />

system to investigate the mechanisms behind breast cancer initiation<br />

via genetic manipulation of some well known oncogenes and tumour<br />

suppressors involved in breast cancer progression.<br />

Myoepithelial cells (Myo1089) and fibroblasts (generated in house)<br />

were isolated and immortalised from breast reduction mammoplasty<br />

samples collected with ethical approval. Following characterisation,<br />

these were virally transfected with Enhanced Green Fluorescent<br />

Protein (EGFP) and dsRed protein respectively to enable tracking.<br />

3D cultures were established in collagen 1 and included the nontumorigenic<br />

luminal epithelial cell line HB2, EGFP Myo1089 cells<br />

and dsRed fibroblasts. Cells were cultured for 3 weeks in Transwell<br />

cell culture inserts. Following fixation these were analysed by H&E,<br />

confocal microscopy and immunohistochemistry. Morphology and<br />

immunostaining profiles were compared to sections of normal breast<br />

tissue.<br />

Immunohistochemical characterisation using the following antibodies:<br />

E-cadherin, epithelial membrane antigen, vimentin, laminin 5,<br />

collagen 4 plus luminal and basal cytokeratins, demonstrated polarised<br />

epithelial structures with lumen formation and basement membrane<br />

production with a similar immunostaining profile to normal breast<br />

tissue. The importance of including myoepithelial cells and fibroblasts<br />

in maintaining these structures was demonstrated. We established this<br />

model is amenable to genetic manipulation by overexpressing Her2 in<br />

HB2 cells, and knocking out ERβ1 in EGFP Myo1089 cells and Rac1<br />

and Dock4 in dsRed fibroblast cell lines using shRNA techniques.<br />

These were included in separate models with morphological and<br />

phenotypic effects determined by confocal microscopy.<br />

In summary we have developed an in vitro model of normal breast<br />

tissue that includes three of the major functional breast cell types<br />

cultured in a physiologically relevant 3D matrix. We have validated<br />

the morphology and protein expression profile against human breast<br />

tissue specimens and confirm that this is a suitable model of normal<br />

breast. The model is suitable for experimental manipulation and<br />

cell behaviour can be easily visualised using standard laboratory<br />

techniques. We conclude this is a robust in vitro model of normal<br />

breast tissue offering an alternative cost-effective method of studying<br />

genes involved in breast cancer initiation.<br />

P6-02-07<br />

In vitro 3D Model of <strong>Breast</strong> Tumor Stroma<br />

Jaganathan H, Mitra S, Dave B, Godin B. The Methodist Hospital<br />

Research Institute, Houston, TX<br />

One in eight women is diagnosed with breast cancer in the United<br />

States every year. Despite advancements for early diagnosis and<br />

initial treatment of breast cancer, the complexity and multiplicity of<br />

biological factors and barriers in tumor microenvironment prevent<br />

potent active agents from reaching their targets in cancer cells.<br />

There has been a growing body of evidence about the importance of<br />

three-dimensional (3D) culture systems in discovery and modeling<br />

of biological processes with in vivo relevance. A few examples that<br />

have been reported in 3D monocultures include different growth<br />

patterns of normal and malignant breast cancer cells in 3D cultures of<br />

laminin-rich gels and effect on cell polarity pathways. These effects<br />

are not observed in conventional two-dimensional (2D) cultures,<br />

showing that cells organized in a 3D matrix can shed light on novel<br />

mechanisms in breast cancer biology. Moreover, neither 2D nor<br />

3D monoculture models available today mimic the complexity of<br />

clinically observed tumor microenvironment (tumor stroma), which<br />

also contains fibroblasts, macrophages and endothelial cells and<br />

represents an important physiological barrier for efficient delivery of<br />

therapeutics to breast tumors. In this work we propose and investigate<br />

an in vitro system mimicking the tumor microenvironment in which<br />

relevant cell populations in tumor microenvironment are co-cultured<br />

with breast cancer cells*. The system uses the Nano3D® platform<br />

based on magnetic levitation. 3D tumor models for breast cancer were<br />

designed and optimized by co-culturing SUM159 and MDA-MB-231,<br />

triple negative breast cancer cells, with 293T fibroblasts. The cells<br />

were co-cultured at different ratios and for a various time periods.<br />

Further, to assess the similarity of the in vitro system to the in vivo<br />

disease, we tested an an orthotopic model of breast cancer in SCID<br />

beige mice. Histological analysis of frozen tissues from in vitro and<br />

in vivo studies confirmed that the in vitro 3D system based on the<br />

mixture of fibroblasts and cancer cells has structural similarities to<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

490s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: General Session 6<br />

the in vivo observed tumor stroma. Engineering an in vitro 3D tumor<br />

model that accurately represents the in vivo tumor will further benefit<br />

research efforts in breast cancer diagnosis and therapy.<br />

*Patent pending – inventors: B. Godin, G. Souza, and B. Dave<br />

Acknowledgments: The authors acknowledge a financial support<br />

from the Susan G. Komen Postdoctoral Fellowship #<strong>PDF</strong>12229449.<br />

P6-02-08<br />

Molecular drivers of adipogenotoxicosis in breast tumorassociated<br />

adipose<br />

Ellsworth RE, Field LA, van Laar R, Deyarmin B, Hooke JA, Shriver<br />

CD. Windber Research Institute, Windber, PA; Signal Genetics, New<br />

York, NY; Walter Reed National Military Medical Center, Bethesda,<br />

MD; Henry M. Jackson Foundation, Windber, PA<br />

Background: Having long been thought to function only as an inert<br />

energy storage depot, the role of adipose tissue in tumorigenesis has<br />

been largely ignored; however, adipose is an active endocrine organ<br />

that can directly influence tumor growth. Improved understanding of<br />

the role of adipose in tumorigenesis is crucial given the association<br />

between obesity and breast cancer risk in post-menopausal women,<br />

increasing rates of obesity and use of autologous fat transfer in breast<br />

reconstruction.<br />

Methods: Adipose, adjacent to and distant from (>3 cm from<br />

the closest tumor margin) invasive breast tumors, was laser<br />

microdissected from 20 post-menopausal women, and from 22<br />

post-menopausal women with non-malignant breast disease. Gene<br />

expression data were generated using U133A 2.0 microarrays. Data<br />

were analyzed to identify significant patterns of differential expression<br />

between adipose classes at the individual gene and molecular pathway<br />

level. Gene expression differences were validated using qRT-PCR in<br />

an additional set of 29 specimens.<br />

Results: SPP1, RRM2, MMP9 and PLA2G7 were expressed at >3-<br />

fold (P3-fold higher expression<br />

of EGFL6 and ITGB2 and >3-fold lower levels of PIP, which are<br />

involved in growth, proliferation, and cellular adhesion in adjacent<br />

compared to non-malignant adipose. Pathway analysis revealed that<br />

immune response differs between non-malignant, distant and tumoradjacent<br />

adipose with an enhanced B- and T-cell response detected in<br />

adjacent compared to distant or non-malignant adipose. Inflammatory<br />

response as well as DNA transcription and replication pathways were<br />

differentially expressed in distant compared to non-malignant adipose.<br />

Conclusions: Gene expression levels differ in breast adipose<br />

depending on presence of and proximity to tumor cells. Adipose<br />

adjacent to the tumor demonstrated the largest immune response,<br />

supporting the idea of adipogenotoxicosis, which through proinflammatory<br />

and genotoxic responses, promotes tumor development.<br />

In addition, genes involved in cellular proliferation, degradation of the<br />

extracellular matrix and angiogenesis are differentially expressed in<br />

adjacent compared to distant or non-malignant adipose, thus tumoradjacent<br />

adipose may be contributing to the growth and invasion of<br />

the primary tumor. These data thus suggest that adipose is not an<br />

inert component of the breast microenvironment but plays an active<br />

role in tumorigenesis.<br />

P6-02-09<br />

Role of HGF in obesity-associated tumorigenesis: C3(1)-Tag mice<br />

as a model for human basal-like breast cancer<br />

Sundaram S, Freemerman AJ, Hamilton J, McNaughton K, Galanko<br />

JA, Darr DB, Perou CM, Troester MA, Makowski L. University of<br />

North Carolina at Chapel Hill, Chapel Hill, NC<br />

Obesity is associated with basal-like breast cancer (BBC), an<br />

aggressive breast cancer subtype. The objective of this study was to<br />

determine whether high fat diet-induced obesity promotes BBC onset<br />

in adulthood and to evaluate the role of stromal-epithelial interactions<br />

in obesity-associated tumorigenesis. Specifically, we hypothesized<br />

that hepatocyte growth factor (HGF) plays a promoting role in BBC,<br />

which tends to express the HGF receptor, c-Met. In C3(1)-Tag mice,<br />

a murine model of BBC, we demonstrate that obesity leads to a<br />

significant increase in HGF secretion and an associated decrease in<br />

tumor latency. By immunohistochemical analysis, normal mammary<br />

tissue exhibit obesity-induced hepatocyte growth factor (HGF) in<br />

stroma and corresponding epithelial expression of c-Met. HGF<br />

secretion was also increased in primary mammary fibroblasts isolated<br />

from normal mammary and tumors of obese mice compared to lean.<br />

These results show that diet-induced elevation of HGF expression is a<br />

stable phenotype, maintained after several passages and after removal<br />

of dietary stimulation. In cocultures, neutralization of HGF blunted<br />

tumor cell migration; further linking diet-mediated HGF-dependent<br />

effects in tumor aggressiveness. In sum, these results suggest that<br />

HGF plays an important role in obesity-associated carcinogenesis and<br />

raise the hypothesis that diet may affect lasting changes in stroma,<br />

potentially through epigenetic means. Understanding the effects of<br />

obesity on risk and progression is important given epidemiologic<br />

studies that imply half of BBC could be eliminated by reducing<br />

obesity. Our findings suggest the possibility that reducing obesity may<br />

be useful in preventing obesity-associated breast cancer mediated by<br />

the HGF/c-Met pathway.<br />

P6-03-01<br />

Molecular Dissection of <strong>Breast</strong> Luminal Cell Transcription<br />

Factor Networks<br />

Bargiacchi FG, Earp HS, Perou CM. University of North Carolina<br />

Chapel Hill, NC<br />

During mammary development, cellular differentiation and lineage<br />

commitment are driven by distinct growth cycles under the control<br />

of local epithelial and mesenchymal paracrine signaling mechanisms.<br />

Within the mammary gland are multipotent stem cells, which give rise<br />

to luminal and myoepithelial lineages. The mechanisms that govern<br />

differentiation into these distinct cell populations are not completely<br />

understood, yet critically important for a complete understanding of<br />

normal development and breast tumorigenesis. Recent studies have<br />

shown that retroviral transduction of mouse and human fibroblasts<br />

with four transcription factors can initiate the conversion of a somatic<br />

cell into an embryonic stem cell-like state, with capabilities of<br />

differentiating into cell types of all three germ layers. The generation<br />

of induced pluripotent stem cells (iPS) illustrates that transformation<br />

of a mature cell into a more immature state can erase most epigenetic<br />

programming critical for the differentiation of that cell. Our goal is<br />

to determine whether mammary specific transcription factors can<br />

directly transdifferentiate fibroblasts or human mammary epithelial<br />

cells (HMEC) into functionally differentiated ER+/luminal cells.<br />

To address this goal of creating ER+/luminal cells via an iPS<br />

type approach, we have developed a model in which an hTERT –<br />

immortalized human mammary epithelial cell line are transduced<br />

utilizing a multifunctional lentiviral expression vector system,<br />

www.aacrjournals.org 491s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

which allows the simultaneous expression and/or silencing of<br />

multiple candidate genes. We are investigating the abilities of various<br />

combinations of transcription factors (TFs) to convert somatic<br />

cells to fully functional differentiated mammary epithelial cells.<br />

Nine candidate TFs (including ER , GATA3, FOXA1, XBP1 and<br />

CDKN2C) were chosen based upon their differential expression in<br />

genomic transcriptional data set of primary human breast tumors,<br />

and in fluorescence-activated cell sorting (FACS) of normal breast<br />

cell populations. These TFs are highly expressed in ER+/luminal<br />

cells, and some are known to be important lineage-specific factors.<br />

Ectopic overexpression of these cDNAs in HMECs indicated<br />

that these transcription factors have both unique and overlapping<br />

contributions towards inducing the expression of genes responsible<br />

for luminal differentiation. Examining the expression patterns of<br />

individually transduced cells using a classifier of differentiation<br />

status (Differentiation Score, Prat et al. 2010) identified several genes,<br />

including ESR1 and GATA3, as causing a transition towards the<br />

luminal subtype. These individual gene data will guide the selection<br />

of combinations, since we anticipate that, as was seen for iPS cells,<br />

multiple genes will be required for transdifferentiation into the luminal<br />

cell lineage. Since there are presently no cell lines or mouse models<br />

of Luminal A/ER+ breast cancers, the artificial creation of such of a<br />

cell line would be of great value.<br />

P6-04-01<br />

Global analysis of breast cancer metastasis suggests cellular<br />

reprogramming is central to the endocrine resistant phenotype.<br />

Bolger JC, McCartan D, Walsh CA, Hao Y, Hughes E, Byrne C, Hill<br />

ADK, O’Gaora P, Young LS. Royal College of Surgeons in Ireland,<br />

Dublin 2, Ireland; University College Dublin, Ireland<br />

The development of breast cancer resistance to endocrine therapy<br />

results from altered cellular plasticity leading to the emergence of<br />

a hormone independent tumour. We have previously shown that the<br />

endocrine resistant phenotype is poorly differentiated and has greater<br />

metastatic potential when compared with an endocrine sensitive<br />

phenotype. The underlying mechanism of how an ER positive,<br />

endocrine sensitive tumour can turn on these adaptive mechanisms<br />

remains unresolved. We hypothesise that cellular reprogramming<br />

can occur as a result of long-term exposure to endocrine treatment<br />

which manifests clinically as tumour recurrence. Our aim was to<br />

identify the mechanism of breast cancer cellular reprogramming in<br />

a clinical context.<br />

In this study we adopted a global approach to define transcriptional<br />

networks significantly elevated in the breast cancer metastatic<br />

setting. Using RNAseq we determined the gene expression profile<br />

of three ER positive patients who developed tumour recurrence on<br />

tamoxifen treatment. RNAseq was performed on primary tumours,<br />

axillary node metastases and subsequent distant metastases. Following<br />

bioinformatic analysis, we defined the genes that were significantly<br />

elevated in the metastatic tumours. In the metastases, genes clustered<br />

away from those in the primary and node, with 209 genes uniquely<br />

elevated in the metastatic context, suggesting altered gene expression<br />

in response to endocrine therapy. Pathway analysis of genes<br />

significantly elevated in the metastatic setting included developmental<br />

pathways (WNT signalling and TGFbeta), growth factor signalling<br />

pathways (MAPkinase), cell adhesion molecules, junction adherens<br />

and pathways in cancer. Further analysis provided a list of clinically<br />

relevant transcriptional networks which may facilitate tumour cell<br />

de-differentiation. Transcription factors including Myc, SMAD2,<br />

ESR, TCF3, CUX1 and CLOCK were identified which potentially<br />

drive reprogramming in response to endocrine treatment.<br />

We interrogated this data to identify downstream targets of this ‘dedifferentiation’<br />

network for new predictive markers of response to<br />

endocrine treatment. Bioinformatic analysis identified PAWR, an<br />

inhibitor of progenitor cell expansion as a potential SMAD2 target.<br />

In a xenograft model of endocrine resistance, PAWR expression was<br />

found to be reduced in metastatic tissue from lung, liver and bone<br />

compared with the primary tumour. In human breast cancer patients<br />

PAWR significantly associated with a good response to endocrine<br />

treatment and luminal A status (p=0.0008), establishing PAWR as a<br />

predictor of good response to endocrine therapies.<br />

We have identified a transcriptional network which may drive a<br />

de-differentiated phenotype following endocrine treatment. Data<br />

presented here proposes a mechanism by which apparently less<br />

aggressive Luminal A type tumours may fail endocrine treatment and<br />

develop subsequent recurrence. This represents a fundamental shift in<br />

how we approach the development of resistance to endocrine therapy,<br />

establishing a number of biomarkers which will allow tracking of<br />

response to endocrine therapy or allow accurate early prediction of<br />

those who will respond to treatment.<br />

P6-04-02<br />

Final progression-free survival analysis of BOLERO-2: a phase<br />

III trial of everolimus for postmenopausal women with advanced<br />

breast cancer<br />

Piccart M, Baselga J, Noguchi S, Burris H, Gnant M, Hortobagyi G,<br />

Mukhopadhyay P, Taran T, Sahmoud T, Rugo H. Institut Jules Bordet,<br />

Université Libre de Bruxelles, Brussels, Belgium; Massachusetts<br />

General Hospital <strong>Cancer</strong> Center and Harvard Medical School,<br />

Boston, MA; Osaka University, Osaka, Japan; Sarah Cannon<br />

Research Institute, Nashville, TN; Comprehensive <strong>Cancer</strong> Center,<br />

Medical University of Vienna, Vienna, Austria; The University<br />

of Texas MD Anderson <strong>Cancer</strong> Center, Houston, TX; Novartis<br />

Pharmaceuticals Corporation, East Hanover, NJ; University of<br />

California, <strong>San</strong> Francisco Helen Diller Family Comprehensive<br />

<strong>Cancer</strong> Center, UCSF, <strong>San</strong> Francisco, CA<br />

Introduction: Treatment options for postmenopausal women with<br />

hormone receptor-positive (HR + ) breast cancer (BC) who relapse or<br />

progress on a nonsteroidal aromatase inhibitor (NSAI) are limited.<br />

Interim analyses of the BOLERO-2 trial demonstrated that combining<br />

the oral mammalian target of rapamycin (mTOR) inhibitor, everolimus<br />

(EVE), with the steroidal aromatase inhibitor, exemestane (EXE),<br />

significantly prolonged progression-free survival (PFS) in this patient<br />

population. Protocol-specified final PFS analyses are presented.<br />

Methods: The phase III, double-blind, BOLERO-2 trial randomized<br />

postmenopausal women with HR + BC progressing or recurring after<br />

NSAIs to EVE (10 mg once daily; n = 485) plus EXE (25 mg once<br />

daily) or placebo (PBO; n = 239) plus EXE. The primary endpoint<br />

was PFS. Secondary endpoints included overall survival, safety, bone<br />

turnover, and overall response rate.<br />

Results: At a median follow-up of 18 months, 510 PFS events were<br />

reported. The addition of EVE to EXE significantly prolonged median<br />

PFS versus EXE monotherapy as per local assessment (7.8 vs 3.2 mo,<br />

respectively; HR = 0.45 [95% CI, 0.38-0.54]; log-rank P < .0001).<br />

Furthermore, these data were consistent with results based on central<br />

assessment (11.0 mo for EVE + EXE vs 4.1 mo for EXE alone;<br />

HR = 0.38 [95% CI, 0.31-0.48]; log-rank P < .0001). Fewer deaths<br />

were reported with EVE + EXE (25.4%) vs PBO + EXE (32.2%).<br />

In addition to significantly improving PFS and delaying disease<br />

progression in bone, PFS benefits with EVE + EXE versus PBO +<br />

EXE were consistent in all prospectively defined subgroups, including<br />

subsets of patients with visceral metastases (n = 406), 3 or more sites<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

492s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: General Session 6<br />

of metastasis (n = 271), and Eastern Cooperative Oncology Group<br />

(ECOG) performance status 1 or 2 (n = 274). The safety profile of<br />

EVE + EXE was consistent with that reported at the interim analysis<br />

and with data for EVE from other oncology settings. Updated overall<br />

survival data will be presented.<br />

Conclusions: Adding EVE to EXE markedly prolonged PFS in the<br />

overall population and in all patient subsets, validating the rationale of<br />

targeting the mTOR pathway to improve clinical outcome in patients<br />

with HR + advanced BC progressing during or after NSAI therapy.<br />

P6-04-03<br />

Changes in breast tumor metabolism and estradiol binding<br />

as measured by FES PET in patients treated with the histone<br />

deacetylace inhibitor vorinostat and aromatase inhibitor therapy<br />

Linden HM, Kurland BF, Specht JM, Vijayakrishn GK, Gralow JR,<br />

Peterson LM, Schubert EK, Link JM, David MA, Eary JF, Krohn<br />

KA. University of Washington, Seattle, WA; Fred Hutchinson<br />

<strong>Cancer</strong> Research Center, Seattle, WA; University of Pennsylvania,<br />

Philadelphia, PA<br />

Background: Some estrogen receptor-positive (ER+) metastatic breast<br />

cancers are bone and soft tissue dominant, indolent, and controlled<br />

by endocrine therapy. However, these tumors eventually become<br />

refractory to endocrine therapy and need a mechanism to reset the<br />

“estrogen-dependence” to allow continued benefit upon progression.<br />

Histone deacetylase inhibitors (HDACi) act as modulators of gene<br />

expression that are promising therapeutic agents for this group of<br />

tumors (Huang 2000, Sabnis 2011). Preclinical and clinical data<br />

demonstrate in ER-poor tumors and cell lines ER up-regulation and<br />

consequently enhanced lethality to endocrine agents. The optimal dose<br />

and schedule are not known, but two promising phase II studies show<br />

benefit in a continuous schedule (Yardley 2011, Munster 2011). FES<br />

PET is a promising imaging agent used as a biomarker to determine<br />

which patients will benefit from endocrine therapy, and to monitor<br />

estradiol binding during therapy (Mortimer 2001, Linden 2011).<br />

Methods: Patients with ER+ HER2- metastatic breast cancer with<br />

prior aromatase inhibitor (AI) exposure and clinical benefit of<br />

endocrine therapy were eligible for a phase II study of HDACi therapy<br />

to restore sensitivity to AI therapy. Following baseline FDG PET,<br />

FES PET and standard imaging (CT, MRI, ultrasound and/or bone<br />

scan as indicated by tumor location), patients received 2 weeks of<br />

vorinostat therapy (400 mg po daily). FES PET was performed at 2<br />

weeks while on HDACi therapy. Patients then received 6 weeks of<br />

AI monotherapy. FDG PET, FES PET and response assessment were<br />

performed at 8 weeks. Patients with clinical benefit (stable disease or<br />

response) continued on the regimen, 2 weeks of vorinostat followed<br />

by 6 weeks of AI.<br />

Results: To date, 8 patients have been enrolled of whom 6 have<br />

completed the first 8 weeks of treatment and all correlative imaging<br />

studies. FES biomarker imaging results are mixed, with some patients<br />

showing an increase in tumor estradiol concentrating ability by FES<br />

PET on HDACi therapy, and decline in metabolic activity by FDG.<br />

Two patients continue on treatment with clinical benefit. Results will<br />

be updated as accrual continues.<br />

Conclusions: Changes in estradiol binding are measured by serial<br />

FES PET in patients on HDACi therapy support preclinical concept<br />

of HDACi modulation of ER expression in metastatic breast cancer.<br />

Molecular imaging is a promising tool to monitor Estradiol binding<br />

pharmacodynamics, and Vorinostat HDACi therapy is a promising<br />

novel approach to allow patients to avoid toxicities of traditional<br />

chemotherapy once their tumor has progressed on endocrine therapy.<br />

Funding: P01, MKA, Merck<br />

P6-04-04<br />

Upregulation of the androgen agonist prosaposin in aromatase<br />

inhibitor resistant breast cancer; mediation by the developmental<br />

protein HOXC11<br />

McIlroy M, Hao Y, Bane FT, Young L, O’Gaora P. Royal College<br />

of Surgeons in Ireland, Dublin, Ireland; University College Dublin,<br />

Ireland<br />

Aromatase inhibitors work by abrogating the activity of the enzyme<br />

cyp 19 (Aromatase) which converts adrenal androgens into Estrogen<br />

within mammary adipose tissue. It is the synthesized steroid<br />

estrone that plays a pivotal role in promoting breast cancer growth<br />

in postmenopausal women. The development of resistance to AI<br />

therapy is still being elucidated, however studies have shown that<br />

tumour adaptability is of vital importance in the cancer cells ability<br />

to evade treatment. This may occur via the switch from estrogen<br />

dependent to estrogen-independent growth which manifests as the loss<br />

of positive estrogen receptor and progesterone receptor status. The<br />

levels of androgens generated by the adrenal glands of women decline<br />

gradually with age but are not dramatically impacted by menopause.<br />

As breast cancer cells are treated AI drugs to block the conversion of<br />

androstendione to estrogen these androgenic steroids will continue to<br />

circulate. Elucidation of the adaptive changes in the cancer cells in<br />

response to the altered steroid environment is therefore of interest.<br />

Research from this lab has identified the homeobox protein HOXC11<br />

as an interacting partner of the steroid receptor coactivator SRC1 in<br />

endocrine resistance. HOXC11 target genes are undefined and so<br />

motif-mapping and RNA-seq experiments were undertaken to help<br />

elucidate the role of HOXC11 in the development of resistance and<br />

metastatic spread. From these studies we have identified Prosaposin<br />

(PSAP) as a putative HOXC11 regulated gene. PSAP is a known<br />

androgen agonist associated with metastatic potential in prostate<br />

cancer and endocrine resistance in breast cancer. Studies performed<br />

confirmed the ability of HOXC11 to regulate PSAP in AI resistant<br />

breast cancer. At a translational level PSAP is an extremely attractive<br />

target for clinical investigation due to its secretory nature which<br />

facilitates quantification from blood serum. Studies exploring the<br />

levels of secreted PSAP in resistant cell line models were undertaken<br />

and only the AI (letrozole) resistant cells had detectable levels of<br />

PSAP. Furthermore when we looked at levels of PSAP in breast<br />

cancer patients 18% had detectable amounts of the protein in their<br />

serum and the sample with the most elevated level was discovered<br />

to be from a patient who had suffered disease recurrence whilst<br />

undergoing AI therapy. High levels of serum PSAP also associated<br />

with strongly positive staining for androgen receptor in matched<br />

patient tissue. Survival analysis on a cohort of breast cancer patients<br />

(n=680) suggests that in AI treated patients there is a trend indicting<br />

that positivity for androgen receptor may result in reduced diseasefree<br />

survival. As a secreted protein PSAP will be readily detectable<br />

in breast cancer patients’ serum from a simple blood sample and<br />

may indicate that the tumour cells are adapting to an androgen rich<br />

environment promoting disease progression.<br />

www.aacrjournals.org 493s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P6-04-05<br />

Tamoxifen dose escalation based on endoxifen level: a prospective<br />

trial with genotyping, phenotyping and pharmacokinetics over<br />

4 months.<br />

Zaman K, Dahmane E, Perey L, Bodmer A, Anchisi S, Wolfer A,<br />

Galmiche M, Stravodimou A, Buclin T, Eap C, Decosterd L, Csajka<br />

C, Leyvraz S. University Hospital CHUV, Lausanne, Switzerland;<br />

Ensemble Hospitalier de la Côte, Morges, Switzerland; University<br />

Hospital CHUV, University of Geneva, Lausanne, Switzerland;<br />

University Hospital, Geneva, Switzerland; Hôpital Cantonal, Sion,<br />

Switzerland<br />

Background<br />

Retrospective studies assessing the impact of tamoxifen (Tam)<br />

metabolism and its active metabolite, endoxifen, on the efficacy of<br />

the treatment produced conflicting results. The prospective CYPTAM-<br />

BRUT 2 trial is ongoing 1 . In the present study we assessed if the level<br />

of Tam metabolites could be improved by doubling tamoxifen dose in<br />

breast cancer patients (pts) with any CYP2D6 genotype, poor (PM),<br />

intermediate (IM) and also extensive metabolizer (EM).<br />

Patients and methods:<br />

This multicenter, prospective, open-label trial included pts treated<br />

with Tam for ≥ 4 months. CYP2D6 activity was determined centrally<br />

by genotyping and phenotyping (dextromethorphan test). Liquid<br />

chromatography-tandem-mass spectrometry was used to measure<br />

Tam, N-desmethyltamoxifen (N-DMT), 4-hydroxytamoxifen (4-HT)<br />

and endoxifen twice at baseline (Tam 20 mg qd), then at days 30,<br />

90 and 120 after having increased the dose to 20 mg bid. Endoxifen<br />

increase and the differences between genotype/phenotype subgroups<br />

were analyzed by ANOVA.<br />

Results:<br />

76 pts were analyzed. Steady-state concentrations for Tam and its<br />

metabolites were reached in 30 days after doubling the dose. A<br />

range of 1.6 to 1.8 fold increase was observed. Geometric mean<br />

plasma concentrations in ng/ml (CV%) were: at baseline and day 30<br />

respectively 134 (48) and 246 (46) for tamoxifen (p < 0.0001); 246<br />

(53) and 413 (48) for N-DMT (p < 0.0001); 2.3 (44) and 3.7 (51) for<br />

4HT (p < 0.0001); 18.7 (89) and 31.1 (92) for endoxifen (p = 0.005).<br />

The level of endoxifen increased 1.4 to 1.7 folds in all genotype<br />

subgroups with geometric mean plasma concentrations in ng/ml<br />

(CV%): 6.9 (36) to 9.7 (24) in PMs (p = 0.7); 14.2 (69) to 20.7 (76)<br />

in IMs (p < 0.0001); and 22.6 (76) to 38.7 (85) in EMs (p < 0.0001).<br />

Similar results were obtained while considering phenotype subgroups.<br />

Genotypes and phenotypes explained less than 30% of the variability<br />

in endoxifen levels.<br />

The occurrence of hot flashes and night sweating were followed<br />

prospectively. Endoxifen levels did not predict an increase in HF/<br />

NS events’ overall occurrence (OR = 1.01, CI 95% 0.78 – 1.31 for HF<br />

and 1.01, CI 95% 0.79 – 1.29 for NS). Twelve pts received CYP2D6<br />

inhibitors. Nine pts did not complete the planned 4 months with<br />

tamoxifen 20 mg bid. The main reasons were mood disorders, hot<br />

flashes, headache and nausea. Self-reported treatment compliance<br />

assessed by monthly anonymous questionnaire was ≥ 95%, except<br />

80-95% in 4 pts.<br />

Conclusions:<br />

This is the first trial reporting the impact of the increase of tamoxifen<br />

dose in all CYP2D6 genotypes, including EMs. Dose escalation of<br />

tamoxifen increased significantly the plasma level of endoxifen by<br />

similar ratio in all genotype subgroups.<br />

Because of a huge inter-individual variability genotyping and<br />

phenotyping are not adequate surrogate markers of endoxifen level.<br />

Very low endoxifen levels are observed even in pts classified as EM.<br />

Future trials aiming to improve the plasma level of endoxifen<br />

should consider direct measurement of the metabolite in plasma and<br />

adjust tamoxifen dose according to the initial level of the metabolite<br />

independently of the genotype.<br />

Reference 1: A. Dieudonné, Journal of Clinical Oncology, 2011 ; vol<br />

29, No 15, suppl (May 20, 2011) : TPS 140<br />

P6-04-06<br />

Inverse regulation of Neuregulin1 and HER-3 during treatment<br />

with aromatase inhibitors of estrogen receptor-positive breast<br />

cancer<br />

Flågeng MH, Larionov A, Geisler J, Dixon JM, Lønning PE, Mellgren<br />

G. Haukeland University Hospital, Bergen, Norway; University<br />

of Edinburgh, United Kingdom; Akershus University Hospital,<br />

Lørenskog, Norway; University of Bergen, Norway<br />

Background: Resistance to endocrine therapy may be promoted<br />

by deregulation of growth factor signaling pathways in addition to<br />

its intimate bidirectional crosstalk with the estrogen receptor (ER)<br />

signaling pathway. We have previously shown HER-2/neu mRNA<br />

upregulation in HER-2/neu non-amplified tumors responding<br />

to therapy with aromatase inhibitors (AIs) [1]. Targeting HERdownstream<br />

signaling pathways may represent a strategy to overcome<br />

acquired endocrine resistance. Here, we aim at exploring the effect<br />

of treatment with AIs on the HER-receptor family members and<br />

the HER-3/-4 ligand neuregulin1 (NRG1) in ER+ breast cancers.<br />

Methods: Matched tumor biopsies were collected from 85 ER+<br />

breast cancers before and after three months of neo-adjuvant treatment<br />

with the AIs letrozole or anastrozole. For 64 of the patients, tumor<br />

biopsies were also available at two weeks of treatment. The patients<br />

were classified as either responders or non-responders depending<br />

on more or less than 50% reduction in tumor size, respectively.<br />

RNA was extracted from tumors and mRNA expression analyses<br />

of HER-1-4 and NRG1 were performed by real-time PCR using<br />

gene-specific primers. Changes in mRNA levels during treatment<br />

were estimated using the non-parametric Wilcoxon sign-rank test,<br />

while the correlations between mRNA levels were analyzed using<br />

2-tailed Spearman rank test. Results: Among all the ER+ HER-2/<br />

neu non-amplified tumors we observed a significant increase in<br />

mRNA expression of EGFR/HER-1 (51/64 tumors, p


December 4-8, 2012 <strong>Abstract</strong>s: General Session 6<br />

1. Flageng, M.H., L.L. Moi, J.M. Dixon, J. Geisler, E.A. Lien, W.R.<br />

Miller, P.E. Lonning, and G. Mellgren, Br J <strong>Cancer</strong>, 2009.101:<br />

1253-60.<br />

2. Makhija, S., L.C. Amler, D. Glenn, F.R. Ueland, M.A. Gold, D.S.<br />

Dizon, V. Paton, C.Y. Lin, T. Januario, K. Ng, A. Strauss, S. Kelsey,<br />

M.X. Sliwkowski, and U. Matulonis, J Clin Oncol, 2010. 28:1215-23.<br />

P6-04-07<br />

Significance and therapeutic potential of PELP1-mTOR axis in<br />

breast cancer progression and therapy resistance<br />

Gonugunta VK, Cortez V, Sareddy GR, Roy SS, Zhang H, Tekmal<br />

RR, Vadlamudi RK. UTHSCSA, <strong>San</strong> <strong>Antonio</strong>, TX; Shantou University<br />

Medical College, Shantou, China<br />

Proline, Glutamic-acid and Leucine-rich Protein 1 (PELP1) is a<br />

proto-oncogene that modulates ER signaling by functioning as an<br />

ER-coregulator. Emerging studies demonstrated that in a subset of<br />

breast tumors, PELP1 is predominantly localized in the cytoplasm<br />

and that PELP1 participates in extranuclear signaling by facilitating<br />

ER interactions with Src, PI3K, and AKT. PELP1 expression is<br />

upregulated in breast cancer, its deregulation contributes to therapy<br />

resistance, and PELP1 is a prognostic marker of poor survival.<br />

However, the mechanism by which PELP1 extranuclear actions<br />

contributes to cancer progression and therapy resistance remains<br />

unknown. We have recently discovered that PELP1 has the potential<br />

to interact with mammalian target of rapamycin (mTOR), a serine/<br />

threonine kinase that forms two distinct complexes called mTORC1<br />

(containing Raptor and PRAS40) and mTORC2 (containing<br />

Rictor and Protor). The objective of this application is to test<br />

whether crosstalk occurs between mTOR and PELP1 signaling<br />

axis and to test whether mTOR targeting drugs can be used to<br />

target PELP1 oncogenic functions. We have used breast cancer<br />

cells with PELP1 overexpression (MCF7-PELP1, ZR75-PELP1,<br />

T47D-PELP1) or PELP1 down regulation (MCF7-PELP1shRNA,<br />

ZR75-PELP1shRNA) along with controls to study the role of PELP1<br />

in the regulation of mTOR axis. PELP1 knockdown significantly<br />

reduced downstream mTOR signaling components as analyzed by<br />

Western analysis using phospho-S6K, -4EBP1, -mTOR and -Akt,<br />

antibodies. Overexpression of PELP1 activated mTOR signaling<br />

components. Using immunoprecipitation, we have demonstrated<br />

that PELP1 interacts with mTOR. Further immunopreciptation<br />

analysis using Rictor and Raptor specific antibodies revealed that<br />

PELP1 associates with both TORC1 and TORC2 complexes. Using<br />

PELP1WT and PELP1cyto (that predominantly localizes in the<br />

cytoplasm), we have demonstrated the differential activation of mTOR<br />

signaling components: PELP1WT activated both TORC1 and TORC2<br />

pathways, while PELP1cyto uniquely activated TORC2. mTOR<br />

targeting drugs (Rapamycin or AZD8055) showed a significant effect<br />

on the in vitro proliferation of PELP1 model cells. AZD8055 is more<br />

potent in reducing PELP1 driven tumor growth in vivo compared<br />

to rapamycin. Immunohistochemical studies on xenografts derived<br />

from MCF7, MCF7-PELP1WT and MCF7-PELP1cyto models<br />

demonstrated that PELP1 signaling modulates mTOR signaling<br />

in vivo and inhibition of mTOR signaling rendered PELP1 driven<br />

tumors to be highly sensitive to therapeutic inhibition. Further,<br />

mTOR inhibitors sensitized tamoxifen therapy resistant PELP1cyto<br />

model cells to hormonal therapy. IHC analysis of mammary glands<br />

and mammary tumors from PELP1Tg mice revealed deregulation<br />

of mTOR signaling components with excessive activation of S6K<br />

and 4EBP1. Using breast tumor tissue arrays (n=100), we found<br />

significant correlation of PELP1 cytosolic localization with mTOR<br />

signaling. Collectively, the experimental results from these studies<br />

identified PELP1-mTOR axis as a novel component of PELP1<br />

oncogenic functions and suggests, mTOR inhibitor(s) will be effective<br />

chemotherapeutic agents for down regulating PELP1 oncogenic<br />

functions and for blocking PELP1-mediated therapy resistance.<br />

P6-04-08<br />

FOXA1 expression: regulated by EZH2 and associated with<br />

favorable outcome to tamoxifen in advanced breast cancer<br />

Reijm EA, Helmijr JCA, Soler CMJ, Beekman R, Gerritse FL, Pragervan<br />

der Smissen WJC, Ruigrok-Ritstier K, van IJcken WFJ, Stevens<br />

MG, Smid M, Look MP, Meijer ME, Sieuwerts AM, Sleijfer S, Foekens<br />

JA, Berns EMJJ, Jansen MPHM. Erasmus MC - Daniel den Hoed,<br />

Rotterdam, Netherlands; Erasmus MC, Rotterdam, Netherlands<br />

Background: Enhancer of Zeste Homolog 2 (EZH2) is a member of<br />

the polycomb complex 2 and serves as a histone methyltransferase.<br />

Through trimethylation of histone 3 of lysine 27, EZH2 regulates<br />

‘genome wide’ gene transcription silencing.<br />

Downregulation of EZH2 is associated with increased expression<br />

of the estrogen receptor (ER) and favorable outcome to tamoxifen<br />

in metastatic breast cancer. The binding of ER to its target genes is<br />

enhanced by Forkhead Box A1 (FOXA1), a pioneer factor that binds<br />

to and opens chromatized DNA. The aim of this study is to investigate<br />

the potential association between EZH2 and FOXA1 and their relation<br />

with treatment outcome in patients with advanced breast cancer.<br />

Experimental design: EZH2 and FOXA1 mRNA levels were<br />

analysed using available gene expression profile-data of 344 primary<br />

breast cancer specimens of lymph-node-negative (LNN) patients and<br />

in 109 ER-positive (ER+) tumors of patients with metastatic disease<br />

treated with first-line tamoxifen. To functionally analyze EZH2, cell<br />

lines were generated with different knockdown-constructs for EZH2,<br />

followed by evaluation of mRNA and protein expression levels and<br />

chromatin immunoprecipitation (ChIP) experiments combined with<br />

qPCR and/or next generation sequencing (NGS; ChIPseq).<br />

Results: In the 344 LNN breast tumors, EZH2 was highly expressed<br />

in breast tumors of the basal subtype. In contrast, FOXA1 was hardly<br />

expressed in this subtype, but highly in the luminal A and B subtypes.<br />

FOXA1 was related to a prolonged time to progression (TTP) after<br />

tamoxifen (HR=0.51 [0.35-0.74], P


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P6-04-09<br />

Lack of response to aromatase inhibitors involves distinct<br />

mechanisms<br />

Turnbull AK, Larionov AA, Renshaw L, Kay C, Sims AH, Dixon JM.<br />

University of Edinburgh, United Kingdom<br />

Background: Estrogen is a key hormone involved in cancer cell<br />

development. It forms a complex with estrogen receptor alpha (ER)<br />

which binds to estrogen response elements (EREs) in the promoter<br />

regions of genes under its transcriptional control. Letrozole inhibits<br />

local production of estrogen. Despite the prevalent use of such drugs<br />

little is known about their effect at molecular and transcriptomic level.<br />

A better understanding of the molecular mechanisms underlying why<br />

approximately 75% of ER rich cancers respond might help to elucidate<br />

the mechanisms characterising a lack of response in some patients.<br />

Methods: A new gene expression dataset was generated from<br />

sequential core biopsy/theatre samples (before treatment, 10-14 days<br />

on treatment and at surgery) taken from ER+ patients undergoing<br />

neo-adjuvant letrozole treatment. Clinical response to treatment<br />

was assessed by changes in tumour volume based on 3D ultrasound<br />

(performed by a single operator).<br />

Results: On letrozole the majority of ER+ tumours respond with<br />

dramatic early transcriptomic changes. A significant reduction in<br />

proliferation is seen through down-regulation of key cell cycle control<br />

genes such as CyclinD1, CyclinA, CyclinB1, CyclinB2 and CDK2,<br />

and genes involved with the initiation phase of DNA replication<br />

such as the MCM complex. Several of these (including CyclinD1,<br />

CyclinB2 and MCM2) have been shown to contain candidate ERE<br />

sequences in their promoter region and therefore down-regulation<br />

of these would be concordant with deprivation of estrogen. There<br />

is also a significant up-regulation of genes including collagens,<br />

lamanins, integrins and others involved with intra-cellular adhesion<br />

and extra-cellular matrix (ECM) remodelling. Preliminary analysis<br />

suggests the involvement of a local immune response as a potential<br />

mechanism for tumour cell death, as genes involved with antigen<br />

presentation, leukocyte trans-endothelial mediation, natural killer cell<br />

mediated cytotoxicity and T-cell mediated cell death are significantly<br />

up-regulated within the clinically responding group.<br />

Two distinct subsets were seen in non-responders. The first subgroup<br />

shows a ‘classical non-response profile’ with very little change in<br />

expression of genes involved with cell cycle, ECM remodelling,<br />

cellular adhesion and antigen presentation between baseline and<br />

14-days of treatment. The second subgroup have a gene expression<br />

profile similar to the responding group with a down-regulation of cell<br />

cycle and up-regulation of genes involved with ECM remodelling,<br />

cellular adhesion and antigen presentation. Whether these are true<br />

non-responders is not clear.<br />

Conclusion: Approximately half of clinically non-responding patients<br />

have molecular profiles similar to responding patients whilst the<br />

other half have a distinct pattern of expression with significantly less<br />

change in genes associated with cell cycle control, ECM remodelling,<br />

intra-cellular adhesion and antigen presentation. On-going work will<br />

elucidate long term outcomes in these two groups.<br />

P6-04-10<br />

Comprehensive gene and protein assessment of the role of Her2 in<br />

the response to neoadjuvant Letrozole suggests patients without<br />

amplification may also benefit from anti-Her2 treatment<br />

Webber V, Turnbull AK, Larionov AA, Sims AH, Harrison D, Renshaw<br />

L, Dixon JM. Melville Trust for Care and Cure of <strong>Cancer</strong>, Edinburgh;<br />

Western General Hospital, Edinburgh<br />

Background:<br />

Older postmenopausal patients with large operable or locally<br />

advanced oestrogen receptor positive, invasive breast cancers are<br />

candidates for treatment with neoadjuvant letrozole. HER2 positivity<br />

is a potential marker for early endocrine therapy resistance. In this<br />

study we have evaluated the effects of HER2 amplification, gene<br />

and protein expression at diagnosis and following 14 and 90 days of<br />

treatment with neoadjuvant Letrozole.<br />

Methods:<br />

70 postmenopausal women with large operable and locally advanced<br />

oestrogen receptor (ER) positive invasive breast cancers were treated<br />

with neoadjuvant letrozole. Response was assessed by 3D ultrasound.<br />

Sequential core biopsies were taken at 0, 14 days and 3 months of<br />

treatment. 12 patients were HER2 positive (either 3+ or 2+ FISH<br />

positive), 20 were HER2 2+ FISH negative and 38 were HER2<br />

negative (0 or +).<br />

Results:<br />

43 patients were responders to letrozole and 17 were non-responders.<br />

7 of the 17 non-responders were HER2 positive. Analysing response<br />

in two groups (HER2 2+ and 3+ together versus 0 or +) there was<br />

a greater difference in response rate between these two groups than<br />

when splitting into conventional HER2 positive and negative groups<br />

(P


December 4-8, 2012 <strong>Abstract</strong>s: General Session 6<br />

not all patients with ER+ BC respond to hormonal therapy (e.g.<br />

tamoxifen and aromatase-inhibitor) but a large number will eventually<br />

develop resistance to long term therapy.<br />

Purpose: Development of resistance to endocrine therapy is a<br />

clinical issue in ER+ BC. Studies with human cell lines have shown<br />

that upregulation of PI3K-AKT-mTOR and MEK-ERK pathways<br />

contributes to resistance to endocrine therapy (Miller TW et al JCO<br />

2011; <strong>San</strong>chez CG et al <strong>Breast</strong> <strong>Cancer</strong> Res 2011). Using leterozoleresistant<br />

ER+ BC model, we investigated the effects of PI3K-AKTmTOR<br />

pathway inhibitor alone and in combination with MEK1/2<br />

inhibitor.<br />

Methodology: The anti-proliferative, anti-migratory, and intracellular<br />

signaling (pAKT, pS6RP, p4EBP1 and pERK) effects of GDC-0941<br />

(Class I PI3K inhibitor) or GDC-0980 (dual PI3K/mTOR inhibitor)<br />

alone and in combination with GDC-0973 (MEK1/2 inhibitor) were<br />

evaluated in aromatase overexpressed (MCF-7aro), letrozole-resistant<br />

(MCF-7aro/LetR) and long-term-estrogen-deprived (LTEDaro) BC<br />

cell lines by MTT assays, integrin-mediated migration assay and<br />

Western blots.<br />

Results: 1) Both GDC-0980 and GDC-0941 exhibited excellent in<br />

vitro cell killing activity in MTT assay in all cell lines with IC50’s<br />

10-30 nM & 0.2-1.5 µM respectively. PI3K pathway inhibitors<br />

were more potent when combined with GDC-0973; 2) Integrinmediated<br />

migration was significantly higher in both MCF-7aro/LetR<br />

andLTEDaro cells than MCF-7 and MCF-7aro cells; 3) Both MCF-<br />

7aro/LetR and LTEDaro cells movement were significantly abrogated<br />

following the treatment of GDC-0980, and the combination of GDC-<br />

0980 plus GDC-0973 more effectively blocked integrin-mediated<br />

migration; 4) Inhibition of phosphorylation of AKT (S473/T308),<br />

S6RP and 4EBP1 (T37/46) was observed following PI3K pathway<br />

inhibitors; 5) pERK was upregulated following the treatment of<br />

GDC-0941 (PI3K inhibitor); 6) Basal level of pERK was significantly<br />

high in MCF7/LetR and MCF7/LTED cells, and GDC-0980 failed to<br />

change the level of pERK in those cell lines; and 7) Combination of<br />

PI3K pathway inhibitor plus MEK1/2 inhibitor significantly blocked<br />

activation of both pathways.<br />

Conclusion: Our in vitro data suggest that therapeutic targeting of<br />

the PI3K-AKT-mTOR and the MEK-ERK signaling pathways should<br />

be effective in abrogating aromatase-inhibitor-resistance in ER+ BC.<br />

P6-04-12<br />

Novel selective estrogen receptors degraders regress tumors in<br />

pre-clinical models of endocrine-resistant breast cancer<br />

Hager JH, Darimont B, Joseph J, Govek S, Grillot K, Aparicio A,<br />

Bischoff E, Kahraman M, Kaufman J, Lai A, Lee K-J, Lu N, Nagasawa<br />

J, Prudente R, Qian J, Sensintaffar J, Shao G, Heyman R, Rix P, Smith<br />

ND. Aragon Pharmaceuticals, <strong>San</strong> Diego, CA<br />

80% of all breast cancers express the estrogen receptor alpha (ERα)<br />

and thus are treated with anti-hormonal therapies that directly block<br />

ER function (e.g.Tamoxifen) or hormone synthesis (Aromatase<br />

Inhibitors). While these therapies are initially effective, acquired<br />

resistance invariably emerges. Importantly, the majority of these<br />

tumors continue to depend on ERα for growth and survival. The<br />

emerging evidence that ERα can signal in both a ligand-dependent<br />

and ligand-independent manner supports the development of agents<br />

that are not only competitive ERα antagonists but also reduce steady<br />

state levels of the receptor and thus limit both modes of signaling.<br />

We have identified novel, non-steroidal ERα antagonists that<br />

induce degradation of ERα in breast cancer cell lines at picomolar<br />

concentrations resulting in significant reduction in steady state ERα<br />

protein levels. Using peptide-based conformational profiling, we show<br />

that these Selective Estrogen Receptor Degraders (SERDs) induce<br />

estrogen receptor conformations that are distinct from both fulvestrant<br />

and tamoxifen indicating a unique mechanism of action. This unique<br />

biological profile coupled with good oral pharmacokinetics produces<br />

tumor regressions in both tamoxifen-sensitive and -resistant models<br />

of breast cancer in vivo. Recent pre-clinical and clinical data indicate<br />

that PI3K pathway signaling can contribute to the endocrine resistant<br />

state. In a preclinical model of tamoxifen resistant breast cancer in<br />

which SERD monotherapy only produces tumor growth inhibition,<br />

SERD therapy in combination with torc1/2 inhibition results in frank<br />

tumor regressions. These orally bioavailable SERDs hold promise as<br />

a next generation therapy for the treatment of ER+ breast cancer as<br />

monotherapy, as well as in combination with agents that target other<br />

pathways involved in both intrinsic and acquired endocrine resistance.<br />

P6-04-13<br />

HOXB7 functions as a co-activator of estrogen receptor in the<br />

development of tamoxifen resistance<br />

Jin K, Teo WW, Yoshida T, Park S, Sukumar S. Johns Hopkins<br />

University School of Medicine, Baltimore, MD<br />

Background: Multiple factors including long term treatment with<br />

tamoxifen are involved in the development of selective estrogen<br />

receptor modulator (SERM)-resistance in ERα positive breast cancer.<br />

Increased expression of HER family members can also directly alter<br />

cellular responses to tamoxifen, but the mechanism underlying the<br />

increased expression of the HERs is not clear. In this report we show<br />

that HOXB7 is an upstream regulator of HER2 and ER expression.<br />

Results: Overexpression of HOXB7 results in upregulation of HER2<br />

and ERα target genes, while knockdown of HOXB7 with siRNA in<br />

tamoxifen-resistant cell lines causes decrease of HER2 and ERα<br />

target genes expression and loss of tamoxifen resistance. To mediate<br />

upregulation of HER2 and ER-target genes occurring in tamoxifen<br />

resistance, the HOXB7-ERα complex recruits ERα coactivators and<br />

promotes HER2- and ER-target genes transcriptional activity through<br />

direct binding to the HER2 enhancer and regulatory regions of ER<br />

target genes. However, depletion of HOXB7 negatively affects the<br />

binding affinity of its cofactors to the HER2 enhancer element and<br />

ER-binding sites. Also, it is likely that miR196a controls HOXB7<br />

expression; reducing miR196a levels in tamoxifen-resistant cells<br />

causes an increase of HOXB7 expression. Overexpression of miR196a<br />

sensitizes MCF-7 tamoxifen resistant cells to tamoxifen through<br />

downregulation of HER2 and ER-target genes while inhibition of<br />

miR196a by anti-miR196a inhibitors increases tamoxifen resistance<br />

through the upregulation of HER and ER target genes in MCF-7<br />

cells. Further, miRNA 196a is regulated by c-MYC which undergoes<br />

stabilization through the EGFR-HER2 signaling pathway. Depletion<br />

of c-MYC by MYC shRNA enhances tamoxifen sensitivity by<br />

increasing the level of miR196a transcripts. Also, by c-MYC ChIP<br />

assay, we found that c-MYC inhibits miR196a expression through<br />

direct binding to its promoter region.<br />

Conclusions: Overexpression of HOXB7 is a key event in the<br />

initiation and maintenance of tamoxifen resistance. These studies<br />

suggest that HOXB7 acts as a key regulator orchestrating two<br />

major groups of target molecules, ERα and HER2, in the oncogene<br />

hierarchy. Blocking MYC expression or reexpression of miR196a<br />

in TAM-R cells may provide novel strategies for reversing SERMresistance.<br />

Therefore, we have provided additional evidence that<br />

supports the thesis that functional antagonism of HOXB7 has the<br />

potential to circumvent tamoxifen resistance.<br />

www.aacrjournals.org 497s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Reference: Jin K, Kong X, Shah T, Penet MF, Wildes F, Sgroi DC,<br />

Ma XJ, Huang Y, Kallioniemi A, Landberg G, Bieche I, Wu X, Lobie<br />

PE, Davidson NE, Bhujwalla ZM, Zhu T, Sukumar S. The HOXB7<br />

protein renders breast cancer cells resistant to tamoxifen through<br />

activation of the EGFR pathway. Proc Natl Acad Sci U S A. 2012<br />

Feb 21;109(8):2736-41.<br />

P6-04-14<br />

Targeting the PI3K/mTOR pathway in patient-derived xenograft<br />

models of endocrine resistant luminal breast cancer<br />

Cottu PH, Bagarre T, Assayag F, Bièche I, Chateau-Joubert S,<br />

Fontaine J-J, Decaudin D, Slimane K, Vincent-Salomon A, Marangoni<br />

E. Institut Curie, Paris, France; Institut Curie, Saint-Cloud, France;<br />

Ecole Veterinaire d’Alfort, Maisons-Alfort, France; Novartis Pharma,<br />

Rueil Malmaison, France<br />

Background. Most ER+ tumors will eventually escape endocrine<br />

treatments. PI3K/mTOR pathway targeting has improved clinical<br />

results in endocrine treatments resistant (ETR) patients, however with<br />

unsustained responses. Our laboratory has developed several models<br />

of ETR luminal breast cancer (LBC) patient derived xenografts (PDX)<br />

suitable for biological studies (Cottu et al, AACR 2012 & IMPAKT<br />

2012). The aims of the present study were 1) to analyze the biological<br />

factors associated with acquired ETR in relevant models of LBC;<br />

2) to determine the therapeutic interest of combining everolimus<br />

with different ET models resistant to different endocrine treatments<br />

(ET). Methods. Implementation of LBC models has been reported<br />

(Cottu et al, BCRT 2012). ETR models were derived by selecting<br />

tumors growing under continuous ET, subsequently re-engrafted<br />

and rechallenged with endocrine therapies (tamoxifen, letrozole,<br />

fulvestrant), and with everolimus alone and combined with ETs. When<br />

possible, initial sensitive tumors were simultaneously treated with the<br />

same protocols. At progression, or after at least 120 days of continuous<br />

therapies, tumor tissue was retrieved. Tissue microarrays (TMA)<br />

were performed to analyze the biological modifications associated<br />

to ET and mTOR targeting. Triplicate immunohistochemistry of<br />

P-AKT/AKT, P-mTOR/mTOR and P-S6 were performed in addition<br />

to standard markers. Transcriptomic and RTPCR analyses are also<br />

being conducted. Results. Three models resistant to tamoxifen and 2<br />

models resistant to surgical ovariectomy used a surrogate to estrogen<br />

deprivation, were developed from 3 initial PDX, and were maintained<br />

as independent hormone-resistant variants. All these models retained<br />

a luminal phenotype, based on IHC and transcriptomic analyses<br />

and had a wt PI3KCA. ETR models had an expression profile very<br />

similar to the initial PDX, suggesting that minor changes may be<br />

associated with the acquisition of an ETR phenotype. Three of these<br />

models have been tested. One tamoxifen resistant model confirmed<br />

a resistance specific to tamoxifen only and a high level of sensitivity<br />

to a fulvestrant everolimus combination. A second PDX yielded 2<br />

other models resistant to tamoxifen and ovariectomy. These 2 models<br />

developed a resistance to all ET modalities. Therapeutic testing<br />

with combinations of everolimus and ETs were highly efficient, but<br />

everolimus alone was as efficient in these 2 models with complete and<br />

durable responses. Activation of the PI3K pathway was demonstrated<br />

in all cases by IHC analyses (expression of pAKT and pS6), and<br />

complementary ongoing RTPCR analyses. Everolimus based effects<br />

were associated with tumor fibrosis, a strong inhibition of Ki67 and<br />

pS6, but not always pAKT. The expression of ER was not modified<br />

by treatments, except in the fulvestrant-treated mice where it was<br />

strongly inhibited. Conclusion. We have developed and validated a<br />

platform of ETR models derived from our LBC original PDX. These<br />

models retained a luminal phenotype although highly resistant to ET.<br />

Activation of the PI3K pathway has been confirmed in ETR models,<br />

and everolimus alone, or combined with ET confirms high and durable<br />

efficacy. Updated mechanistic analyses performed at different time<br />

points will be presented at the meeting.<br />

P6-04-15<br />

The involvement of LMTK3 in endocrine resistance is mediated<br />

via multiple signaling pathways<br />

Giamas G, Xu Y, Filipovic A, Grothey A, Coombes CR, Stebbing J.<br />

Imperial College, London, United Kingdom<br />

We have previously identified new kinases that are able to<br />

phosphorylate and/or modulate ERα activity via a high-throughput<br />

RNA interference screening. Amongst the most potent novel<br />

regulators was LMTK3, a previously uncharacterized molecule. We<br />

found that LMTK3 confers its anti-tumor effects predominantly via<br />

ERα-regulated signaling. Evolutionary analyses revealed LMTK3<br />

was the only kinase that has adaptively evolved and subjected to<br />

Darwinian positive selection, an intriguing and suggestive result given<br />

the unique susceptibility of humans to ERα + breast cancer compared<br />

to the great apes (especially for chimpanzees [Pan troglodytes], our<br />

closest living relatives). Furthermore, in a large cohort of human<br />

breast cancer patients (n=613), LMTK3 protein levels and intronic<br />

polymorphisms were significantly associated with disease-free and<br />

overall survival and predicted response to endocrine therapy. Finally,<br />

using an established orthotopic mouse model we demonstrated<br />

that inhibition of LMTK3 significantly reduces tumor volume and<br />

bioluminescence.<br />

Now, by performing: i) mass spectrometry (MS), ii) SILAC-based<br />

quantitative proteomics and iii) genome microarray experiments of<br />

ERα + breast cancer cell lines (Tam-sensitive and Tam-resistant) in<br />

the presence or absence of various treatments (Tam, E2, ICI), we<br />

reveal certain genes, including well-defined oncogenes and tumorsuppressors<br />

that are statistically up- or down-regulated.<br />

Interestingly, amongst the newly identified LMTK3-regulated genes/<br />

proteins were targets belonging to already established signalling<br />

pathways implicated in cell survival, cytoskeletal rearrangement,<br />

transformation, and autophagy.<br />

In addition, inhibition of LMTK3 re-sensitised a Tam treatment breast<br />

cancer mouse model, to Tam treatment, leading to decreased tumour<br />

growth. Moreover, in patients’ plasma samples, acquired LMTK3 gene<br />

amplification (copy number variation) was associated with relapse<br />

whilst receiving tamoxifen. In aggregate, these data support a role for<br />

LMTK3 in both innate (intrinsic) and acquired (adaptive) endocrine<br />

resistance in breast cancer.<br />

Further results and mechanistic data concerning LMTK3 actions will<br />

be presented, placing for the first time this new kinase in the maps<br />

of signal transduction.<br />

P6-04-16<br />

The role of RIP140 and FOXA1 in breast cancer endocrine<br />

sensitivity and resistance<br />

Harada-Shoji N, Coombes RC, Lam EW-F. Imperial College London,<br />

United Kingdom<br />

Background<br />

The estrogen receptor-α (ERα) is implicated in the initiation and<br />

progression of breast cancer. Endocrine therapeutics , including<br />

tamoxifen (OHT) and fulvestrant (ICI182780; ICI) improve diseasefree<br />

survival. Despite positive effects, de novo or acquired resistance<br />

to endocrine therapies frequently occurs. It has recently been<br />

published that nuclear receptor–interacting protein 1, NRIP1, also<br />

called RIP140, encodes an ER corepressor RIP140 and has a role in the<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

498s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: General Session 6<br />

regulation of breast cancer cell growth in response to anti-estrogens.<br />

FOXA1 has previously been shown to function as a ‘pioneer factor’<br />

and dictate ER dependent gene expression in both normal and<br />

malignant breast epithelial cells. In this study we investigated the<br />

expression of ERα cofactors RIP140 and FOXA1 and their roles in<br />

hormonal therapy and endocrine resistance.<br />

Methods<br />

Tamoxifen sensitive or resistant breast cancer cell lines were analysed<br />

for protein and mRNA expression by western blotting and quantitative<br />

real-time PCR with or without the inhibition of ERα with tamoxifen or<br />

ICI. To determine the functional role of RIP140 and FOXA1 as well<br />

as their interaction in breast cancer, tamoxifen sensitive breast cancer<br />

cells (eg. MCF-7 and ZR-75-1) were transfected with either RIP140<br />

or FOXA1 expression vector and examined for the expression of ERtarget<br />

genes by Western blot and RT-qPCR analysis. The consequence<br />

of depletion of RIP140 and FOXA1 by RNA interference (RNAi) was<br />

also studied in these breast cancer cells. The expression of RIP140<br />

and FOXA1 and potential target genes were also validated in breast<br />

cancer patient samples by Immunohistochemistry.<br />

Results<br />

We first examined the basal expression levels of RIP140 and FOXA1<br />

in tamoxifen sensitive as well as resistant breast cancer cell lines. The<br />

expression levels of RIP140 were low in the resistance cells compared<br />

with their parental sensitive counterparts, while that of FOXA1 had<br />

no significant difference between the cell lines. The degradation of<br />

ERα by ICI treatment resulted in the down-regulation of RIP140 and<br />

FOXA1 expression in the endocrine sensitive. Our experiments also<br />

demonstrated the functional interaction between RIP140 and FOXA1<br />

and showed that depletion of RIP140 by RNA interference (RNAi) in<br />

sensitive cells resulted in reduction of FOXA1 expression.<br />

We also indentified from this work, a number of ER-regulated genes<br />

which are activated by FOXA1 and repressed by RIP140 expression.<br />

The regulation of these target genes by ERα, FOXA1 and RIP140<br />

were also confirmed in the breast cancer patient samples.<br />

Conclusion<br />

The results from this study demonstrated that RIP140 and FOXA1<br />

play a pivotal role in dictating hormonal sensitivity and resistance<br />

in breast cancer. Moreover, this work also identified a number of<br />

ERα-regulated genes which are downstream targets of FOXA1 and<br />

RIP140, and as such, they may be reliable prognostic markers for<br />

endocrine sensitivity in breast cancer.<br />

P6-04-17<br />

The androgen metabolite-dependent growth in hormone receptor<br />

positive breast cancer as a novel aromatase inhibitor-resistance<br />

mechanism<br />

Hanamura T, Niwa T, Nishikawa S, Konno H, Ghono T, Kobayashi Y,<br />

Kurosumi M, Takei H, Yamaguchi Y, Ito K-I, Hayashi S-I. Graduate<br />

School of Medicine, Tohoku University, Sendai, Japan; Shinshu<br />

University School of Medicine, Matsumoto, Japan; Saitama <strong>Cancer</strong><br />

Center, Saitama, Japan<br />

Introduction: Aromatase inhibitors (AIs) have been reported<br />

to exert their anti-proliferative effects not only by reducing the<br />

production of estrogens but also by unmasking the inhibitory effect<br />

of androgens such as testosterone (TS) or dihydrotestosterone (DHT)<br />

in postmenopausal women with hormone receptor-positive breast<br />

cancer. The behavior of androgens in AI-resistance mechanisms is<br />

not sufficiently understood. 5α-androstane-3β,17β-diol (3β-diol)<br />

generated from DHT by 3β-hydroxysteroid dehydrogenase type 1<br />

(3β-HSD type 1: HSD3B1) has androgenic activity and substantial<br />

estrogenic activity, representing a potential mechanism of AI<br />

resistance.<br />

Methods: To investigate these issues, ERE-GFP-transfected MCF-<br />

7-E10 cells were cultured for 3 months under steroid-depleted, TSsupplemented<br />

conditions which is the similar as the AI treatment.<br />

Among the surviving cells, two stable variants that show ER activity<br />

depending on androgen metabolites were selected as AD-EDR<br />

(androgen metabolite-dependent and estrogen depletion-resistant) by<br />

monitoring GFP expression. Using these cell lines, we investigated<br />

the process of adaptation to androgen-abundant conditions and the<br />

role of androgens in AI-resistance mechanisms.<br />

Results: AD-EDR cell lines showed increased growth and induction<br />

of estrogen-responsive genes rather than androgen-responsive<br />

genes by androgens or 3β-diol. Further analysis revealed increased<br />

expressions of HSD3B1 and reduced expression of androgen receptor<br />

(AR) in these cell lines. In parental MCF-7-E10 cells, ectopic<br />

expression of HSD3B1 or inhibition of AR resulted in adaptation to<br />

estrogen-deprived and androgen-abundant conditions. In coculture<br />

with stromal cells replicating the local estrogen production from<br />

androgen, AD-EDR cell lines showed AI resistance compared with<br />

parental MCF-7-E10 cells. Immunohistochemistry and real-time PCR<br />

analyses on 9 pairs of primary and recurrent tissue samples from AIresistant<br />

breast cancer revealed the decrease of AR protein expression<br />

in all cases and increase of HSD3B1 mRNA expression in 5.<br />

Conclusion: In the present study, we successfully cloned two stable<br />

variants that show ER activity depending on androgen metabolites.<br />

Investigation of these cell lines suggested that the increased function<br />

of 3β-HSD type 1 and reduced function of AR contribute to AI<br />

resistance by enhancing the androgen metabolite-dependent growth<br />

and reducing the inhibitory effect of androgens. Our data of clinical<br />

samples suggest that this mechanism also acts as an AI-resistance in<br />

clinical breast cancer in some cases.<br />

P6-04-18<br />

Evaluation of the molecular mechanisms behind fulvestrant<br />

resistant breast cancer<br />

Kirkegaard T, Hansen SK, Reiter BE, Sorensen BS, Lykkesfeldt AE.<br />

Danish <strong>Cancer</strong> Society Research Center, Copenhagen, Denmark;<br />

Aarhus University Hospital, Aarhus, Denmark<br />

To study the mechanisms behind treatment resistance, we have<br />

previously established a large panel of antiestrogen resistant breast<br />

cancer cell lines based on the estrogen receptor (ER)-positive breast<br />

cancer cell line, MCF-7. It seems to be a general phenomenon that<br />

MCF-7 cells switch from ER driven to human epidermal growth<br />

factor receptor (HER) driven cell growth upon development of<br />

fulvestrant resistance. To obtain a broader knowledge of the resistance<br />

mechanisms, we have established four fulvestrant resistant subclones<br />

from the ER-positive breast cancer cell line, T47D.<br />

The parental T47D cells were confirmed to be estrogen responsive<br />

and short-term treatment of the parental T47D cells with fulvestrant<br />

severely reduced cell growth as well as protein expression of ER<br />

and the estrogen regulated proteins, insulin-like growth factor 1<br />

receptor alpha (IGF-1Rα) and progesterone receptor (PR). All<br />

four fulvestrant resistant cell lines had reduced mRNA and protein<br />

expression of HER1 and HER4 compared to T47D, while HER2 was<br />

highly up regulated in the fulvestrant resistant cell lines and ER was<br />

lost. No difference in response to the HER2 inhibitor trastuzumab<br />

or the specific HER2 kinase inhibitor AG825 was observed between<br />

the fulvestrant resistant cell lines and T47D, indicating that HER2<br />

is not the main driver of fulvestrant resistance in T47D cells. We<br />

found no indication of involvement of the two classical signaling<br />

pathways downstream of the HER receptors (Ras/Raf/Erk and PI3K/<br />

Akt). However, the JNK inhibitor SP600125 preferentially inhibited<br />

www.aacrjournals.org 499s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

growth of two tested fulvestrant resistant cell lines. Withdrawal of<br />

fulvestrant for several weeks did not result in regain of ER expression,<br />

or in change of HER receptor expression, supporting a stable resistant<br />

phenotype.<br />

In conclusion, loss of ER expression explains the lack of effect of<br />

fulvestrant in the T47D fulvestrant resistant cell lines. Noteworthy,<br />

the observed over expression of HER2 in the fulvestrant resistant<br />

T47D cells does not seem to be the main driver of cell growth, as<br />

HER2 targeted therapies like trastuzumab and HER2 kinase inhibitor<br />

did not exert a preferential growth inhibition of resistant cells. These<br />

breast cancer cells with high endogenous HER2 expression will be<br />

used as a model mimicking HER2 positive breast cancer which does<br />

not respond to standard HER2 targeted therapies.<br />

P6-04-19<br />

Neutralizing antibody to human GP88 (progranulin) restores<br />

sensitivity to tamoxifen and inhibits breast tumor growth in<br />

mouse xenografts<br />

Serrero G, Dong J, Hayashi J. A&G Pharmaceutical Inc, Columbia,<br />

MD<br />

The 88 kDa glycoprotein GP88 (Progranulin, PCDGF, acrogranin)<br />

is the largest member of the granulin/epithelin family of growth<br />

modulators characterized by 7.5 cysteine-rich granulin/epithelin<br />

repeats. Our laboratory has demonstrated that GP88 represents<br />

an ideal therapeutic and diagnostic target in breast cancer. Our<br />

published studies have demonstrated that: 1) GP88 expression<br />

increases with tumorigenesis; 2) inhibition of GP88 expression<br />

by antisense transfection in MDA-MB-468 cells inhibited by 90%<br />

tumor formation in nude mice; 3) in ER+ breast cancer cells, GP88<br />

stimulates proliferation and its overexpression confers estrogen<br />

independence and resistance to several anti-estrogens and aromatase<br />

inhibitor; 4) In Her-2 overexpressing breast tumors, GP88 stimulated<br />

Her-2 phosphorylation and conferred trastuzumab resistance; 5) GP88<br />

is expressed in 80% invasive ductal carcinoma and 60% of ductal<br />

carcinoma whereas it is negative in lobular carcinoma, benign lesions<br />

and normal mammary epithelial cells; 6) pathological studies with<br />

530 cases of ER+ invasive ductal carcinoma showed that GP88 tumor<br />

expression measured by immunohistochemistry (IHC) is a prognostic<br />

indicator of recurrence in early stage breast cancer patients; High<br />

GP88 tissue expression was associated with a 4-fold increase in risk<br />

of recurrence at 5 years; 7) GP88 is secreted and can be detected<br />

in the serum of breast cancer patients at an increased level when<br />

compared to healthy subjects. Based on these results, we developed<br />

a neutralizing anti-GP88 antibody AG1. AG1 was expressed in a high<br />

yield CHO cell line and formulated. AG1 inhibits GP88 biological<br />

effect (proliferation and migration) in a dose-dependent fashion in<br />

vitro. Here we will report the results of studies investigating the effect<br />

of AG1 on the tumor development of tamoxifen sensitive (MCF-7)<br />

and resistant (TamR) estrogen receptor positive breast cancer cells<br />

lines injected in nude mice. We show that AG1 alone (5mg/kg)<br />

inhibited tumor growth of MCF-7 cells injected into nude mice in<br />

a similar efficacy as tamoxifen. Interestingly, in tamoxifen resistant<br />

cells derived from MCF-7, the anti-GP88 antibody AG1 inhibited by<br />

more than 50% tumor formation of TamR cells in combination with<br />

tamoxifen, whereas tamoxifen only had no effect.<br />

These data suggest that inhibiting GP88 provides a novel and<br />

alternative therapeutic pathway for the restoration of tamoxifen<br />

sensitivity in ER+ breast cancer.<br />

This works is supported by grant 2R44CA124179 from the National<br />

<strong>Cancer</strong> Institute and 02-2010-010 from the Avon Foundation for<br />

Women.<br />

P6-04-20<br />

Endocrine resistance in invasive lobular carcinoma cells parallels<br />

unique estrogen-mediated gene expression<br />

Sikora MJ, Luthra S, Chandran UR, Dabbs DJ, Welm AL, Oesterreich<br />

S. University of Pittsburgh <strong>Cancer</strong> Institute, Pittsburgh, PA;<br />

University of Pittsburgh, PA; Magee-Womens Hospital, Pittsburgh,<br />

PA; University of Utah Huntsman <strong>Cancer</strong> Institute, Salt Lake City, UT<br />

Invasive lobular carcinoma (ILC) represents ∼10% of newly<br />

diagnosed breast tumors, accounting for ∼30,000 cases annually in the<br />

US. However, ILC has been understudied as a breast cancer subtype.<br />

ILC-specific signaling pathways and responses to endocrine therapies<br />

are not well characterized. Recent retrospective analyses of luminaltype<br />

breast cancers suggest that ILC patients treated with endocrine<br />

therapy have poorer disease-free survival and overall survival than<br />

invasive ductal carcinoma (IDC) patients with similar biomarkers. We<br />

hypothesize that unique transcriptional control of estrogen receptoralpha<br />

(ERα) by estrogens and anti-estrogens in ILC cells drive a<br />

differential response of ILC tumors to endocrine therapies.<br />

The ILC cell lines MDA MB 134VI and SUM44PE were used as in<br />

vitro models of ILC tumors to examine responses to estradiol (E2)<br />

and the anti-estrogens tamoxifen (Tam), 4-hydroxytamoxifen (4OHT),<br />

endoxifen (Bx), and fulvestrant (ICI). We examined cell growth and<br />

expression of canonical ERα-regulated genes in response to E2. Cells<br />

were also treated with anti-estrogens +/- E2 to examine their ability<br />

to inhibit E2-induced growth. To establish a genome-wide profile of<br />

ERα-mediated gene regulation in ILC cells, ILC cells were subjected<br />

to gene expression microarray analysis following 0-24hrs treatment<br />

with 1nM E2. In parallel to these in vitro studies, in vivo models of ILC<br />

are being assessed for endocrine responsiveness, including MDA MB<br />

134VI xenografts and the primary human tumor xenograft HCI-013.<br />

We observed that both MDA MB 134VI and SUM44PE are growthinduced<br />

by E2 treatment. However, both cell lines also present de novo<br />

resistance to Tam, 4OHT, and Bx. While ICI treatment completely<br />

blocked E2-induced growth in MDA MB 134VI, Tam, 4OHT, and<br />

Bx acted as partial agonists. Treatment with these compounds alone<br />

induced ∼25% growth relative to E2, and E2-induced growth could<br />

only be inhibited ∼75%. Similarly, SUM44PE cells are strongly<br />

growth-inhibited by ICI treatment, whereas growth is not reduced<br />

by Tam, 4OHT, or Bx treatment. Consistent with these observations,<br />

novel patterns of gene expression are induced by E2 treatment in ILC<br />

cells. E2-treatment induced canonical targets (e.g. GREB1, IGFBP4)<br />

in both ILC cell lines. However, in MDA MB 134VI cells only ∼35%<br />

of genes regulated by E2 at 24hrs overlap with those regulated in the<br />

IDC cell line MCF-7. Further, a subset of the overlapping genes is<br />

differentially regulated between cell types, including ESR1 (1.25-<br />

fold and 0.67-fold versus control in MDA MB 134VI and MCF-7,<br />

respectively), HOXC6, PMP22, and L1CAM. Examination of these<br />

observations in in vivo models is currently ongoing.<br />

These data are consistent with the hypothesis that E2 and antiestrogens<br />

differentially regulate ERα-mediated gene expression<br />

in ILC cells versus IDC cells. The de novo resistance to tamoxifen<br />

observed in ILC cells may correlate with the worse outcomes in<br />

ILC patients observed in retrospective analyses. We hypothesize<br />

that elucidation of the mechanisms responsible for differential ERα<br />

regulation may reveal novel therapeutic targets for treating ILC<br />

patients and overcoming endocrine resistance.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

500s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: General Session 6<br />

P6-04-21<br />

AIB1 expression specifically predicts breast cancer patient<br />

response to aromatase inhibitor therapy<br />

Vareslija D, O’Hara J, Tibbitts P, McBryan J, Hao Y, Hill A, Young<br />

L. Royal College of Surgeons Ireland, Dublin, Ireland<br />

Aromatase inhibitors (AI) have evolved over the last decade into an<br />

effective therapeutic regime for postmenopausal women with primary<br />

or advanced breast cancer. Despite their remarkable success in the<br />

clinic, intrinsic resistance to therapy occurs in a proportion of patients,<br />

while other patients who respond initially to treatment will relapse<br />

with recurrent disease. Previous studies suggest that this may be due,<br />

at least in part to estrogen receptor (ER) hypersensitivity.<br />

We undertook ER ChIPseq analysis on AI resistant cell model and<br />

data for this suggests that ER transcriptional regulation alone may<br />

not be responsible for the development of the resistant phenotype.<br />

We examined the role of the established ER coactivator protein<br />

AIB1. AIB1 has previously been associated with initiation of breast<br />

cancer and resistance to endocrine therapy. In tamoxifen treated<br />

patients, expression of AIB1 in conjunction with an activated HER2<br />

cascade has been associated with treatment resistance and early<br />

disease recurrence. By contrast, we have observed that AIB1 alone<br />

can predict response to AIs. In our TMA the expression of AIB1<br />

associated with disease recurrence (p=0.025) and reduced disease<br />

free survival time (p=0.0471) in patients treated with AIs as firstline<br />

therapy. Reflecting increased growth factor activity reported in<br />

AI resistance, AIB1 expression associated with the growth factor<br />

second messenger signaling proteins, p-Src and pERK1/2, but not the<br />

receptor HER2. These results suggest that AIB1 may utilize additional<br />

transcription factors other than ER to drive endocrine resistance.<br />

Additionally, we show that AIB1 is highly expressed in AI resistant<br />

metastases; therefore, monitoring AIB1 expression may be useful<br />

to screen for disease progression and detect disease advancement<br />

before metastases appear.<br />

Our studies of cell line models of AI resistance suggest that AIB1<br />

may play a functional role in aggressive, migratory, phenotype of<br />

AI resistance. We have generated cell line models of resistance to<br />

letrozole (LetR) and anastrozole (AnaR). Our resistance models<br />

have higher levels of AIB1 and have increased migratory capacity.<br />

Interestingly, knockdown of AIB1 reduces the migratory capacity of<br />

the resistant cells.<br />

Furthermore, we have observed that AIB1 regulation of ER target<br />

genes is selectively enhanced in AI resistant cells in a promoter<br />

specific context. AIB1 recruitment to ER target genes such as pS2 and<br />

Myc becomes insensitive to letrozole. By contrast, AIB1 recruitment<br />

to cyclinD1 retained letrozole sensitivity. Our evidence suggests that<br />

steroidal regulation of transcription factors such as Jun and Fos may<br />

contribute to this promoter-specific regulation of ER target genes.<br />

We establish a role for AIB1 in AI-resistant breast cancer and describe<br />

a new mechanism of ERalpha/AIB1 gene regulation which could<br />

contribute to the development of an aggressive tumour phenotype.<br />

We provide evidence of a central role for AIB1 in regulating selective<br />

ER transcriptional activity and driving tumour recurrence in AI<br />

treated patients. Tackling the emerging problem of AI resistance in a<br />

timely fashion will enable us to tailor existing therapies and improve<br />

outcome in specific patient groups before disease recurrence becomes<br />

a clinical issue.<br />

P6-04-22<br />

Regulation of Notch localization by endocrine therapy in Estrogen<br />

Receptor positive breast cancer cells: Clinical implications for<br />

endocrine resistance<br />

Espinoza I, Caskey M, Baker RC, Miele L. University of Mississippi,<br />

Jackson, MS<br />

Background: Estrogen receptors occur in about two-thirds of breast<br />

tumors and endocrine therapy is probably the most important systemic<br />

therapy for hormone receptor positive breast cancer. However,<br />

after an initial response to hormonal therapy, most tumors develop<br />

resistance leading to disease progression. Central mechanisms<br />

thought to be involved in this process include: 1) alterations in tumor<br />

cell physiology causing insensitivity to drug-induced apoptosis or<br />

induction of drug-detoxifying mechanisms; 2) expression of energydependent<br />

transporters that eject anti-cancer drugs from cells and<br />

3) the emergence of cancer stem-like cell clones that are estrogenindependent<br />

and/or antiestrogen resistant. We and others have shown<br />

that Notch signaling mediates survival signals in ER+ breast cancer<br />

cells; induces expression of drug transporter ABCG2 (BCRP),<br />

activates ERα in the absence of estrogen and mediates survival of<br />

breast cancer stem-like cells. Previously, we demonstrated a crosstalk<br />

between ERα and Notch signaling whereby estrogen inhibits Notch<br />

activation and estrogen withdrawal or tamoxifen re-activate Notch,<br />

a possible mechanism of resistance. The mechanism of these effects<br />

has remained elusive. Methods: we used sub-cellular fractionation and<br />

immunofluorescence staining to investigate the total expression and<br />

cellular distribution of Notch signaling components in MCF-7 cells<br />

treated with 17-β-estradiol, or estrogen-free medium. Results: Our<br />

data shows that in cells treated with estrogen-free medium, Notch1<br />

is located in the Lipid Rafts (LR) in the plasma membrane, along<br />

with Notch signaling components such as presenilin 1 (the catalytic<br />

subunit of γ-secretase), ADAM10 and Notch negative regulator<br />

NUMB. There is published evidence that γ-secretase is most active<br />

in lipid rafts.17-β-Estradiol modifies the membrane lipid composition<br />

and change the localization of Notch and Notch regulators (such as<br />

ADAM10, presenilin-1 and NUMB) at the plasma membrane, thereby<br />

altering the activation of Notch. These observations are consistent<br />

with a model in which the lipid composition of the plasma membrane<br />

is a critical factor in regulating the rate of Notch activation. FDAapproved<br />

cholesterol lowering drugs affect Notch signaling in ways<br />

consistent with this model. Conclusions: Our findings indicate that<br />

endocrine therapy affects Notch signaling at least in part through<br />

alterations in membrane composition and lipid raft localization of<br />

Notch signaling components.<br />

P6-04-23<br />

Targeting of endocrine therapy resistance through combined<br />

mTOR and histone deacetylase inhibition<br />

Ordentlich P, Flechsig S, Hoffmann J. Syndax Pharmaceuticals,<br />

Waltham, MA; EPO-GmbH Berlin, Berlin, Germany<br />

Background: Primary and acquired resistance to endocrine therapy<br />

in advanced breast cancer presents a significant barrier to maximizing<br />

the clinical effectiveness of these agents. Preclinical data has<br />

demonstrated that up-regulation and/or constitutive activation of<br />

estrogen independent growth signaling pathways drive resistance<br />

to aromatase inhibitors and that targeting of these pathways is an<br />

effective strategy to overcoming the resistance. We recently reported<br />

in a randomized, placebo controlled phase 2 study that entinostat<br />

(ENT), an oral class 1 selective HDAC inhibitor, combined with<br />

exemestane extended PFS and OS in postmenopausal women with<br />

ER+ positive breast cancer that had progressed on a prior nonsteroidal<br />

www.aacrjournals.org 501s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

aromatase inhibitor. Likewise, everolimus (EVE), a mTORi,<br />

combined with exemestane or tamoxifen (TAM) also significantly<br />

extended PFS in a similar patient population. In order to determine the<br />

potential benefit of combining ENT with EVE plus hormone therapy<br />

we have piloted a series of xenograft studies in primary human tumor<br />

models of TAM sensitivity and resistance.<br />

Methods: Athymic nude mice bearing xenografts of tissue derived<br />

directly from patient tumors (MaCa4049 – TAM sensitive; MaCa3366/<br />

TAM – partially resistant to TAM) were treated with A) vehicle B)<br />

2 mg/kg EVE C) 2 mg/kg EVE + 10 mg/kg TAM D) 15 mg/kg ENT<br />

E) 2mg/kg EVE + 10mg/kg TAM + 15 mg/kg ENT. Tumor samples<br />

taken at the end of the study were cryoconserved for analysis of gene<br />

and protein expression and protein phosphorylation.<br />

Results: Growth of MaCa4049 tumors was inhibited in all treatment<br />

groups relative to vehicle in two independent studies conducted<br />

in this tumor model. Groups B, C and D demonstrated a similar<br />

level of activity while maximal inhibition was observed in Group<br />

E (combination of EVE + ENT + TAM). All treatments were<br />

generally well tolerated with weight loss similar in groups C and E.<br />

Treatment of MaCa3366/TAM tumors resulted in similar levels of<br />

tumor growth inhibition in groups B, C and E which were all highly<br />

effective in suppressing tumor growth in this model. As EVE alone<br />

was highly effective as a single agent in the MaCa3366/TAM model<br />

the contribution of ENT will be assessed through molecular analysis<br />

of tumors upon study completion. Efficacy results and analysis of<br />

tumor samples from completed studies will be presented to provide<br />

insight into mechanism of action for the triple combination.<br />

Conclusions: Preclinical data demonstrating enhanced activity of<br />

entinostat combined with everolimus and hormone therapy provides<br />

the rationale for clinical investigation of this novel therapeutic<br />

approach to targeting hormone therapy resistance.<br />

P6-04-24<br />

Overexpression of protein kinase C alpha differentially activates<br />

transcription factors in T47D breast cancer cells in the presence<br />

of 17β-estradiol both in the 2D and 3D environments.<br />

Pham TND, Asztalos S, Weiss MS, Shea LD, Tonetti DA. University<br />

of Illinois at Chicago, IL; Northwestern University, Evanston, IL<br />

Background: Our lab has previously shown that overexpression of<br />

protein kinase C alpha (PKCα) results in a hormone independent,<br />

tamoxifen resistant phenotype in the T47D:A18 breast cancer cell<br />

line. Moreover, 17β-estradiol (E2) inhibits colony formation of<br />

T47D/PKCα in 3D Matrigel and tumor growth in vivo but not in the<br />

2D environment (Zhang et al, Mol. <strong>Cancer</strong> Res, 2009). Differential<br />

transcriptional activation may account for the phenotypic changes<br />

observed in T47D/PKCα cells. In this study, we sought to identify<br />

transcription factors (TF) differentially activated in T47D:A18/neo<br />

versus T47D:A18/PKCα grown in 3D Matrigel in the presence and<br />

absence of E2, and compare these TFs with those activated in the 2D<br />

environment using a high-throughput screening (HTS) technology.<br />

Methods: A system for rapid, non-invasive, large scale, dynamic<br />

quantification of TF activity was developed (Weiss et al, PLoS ONE,<br />

2010). Reporter constructs were created containing a TF binding site<br />

that preceeds a basal promoter to produce firefly luciferase (Fluc)<br />

using a lentiviral backbone. The control construct contained only<br />

the basal promoter and was used for normalization. T47D/PKCα and<br />

T47D/neo cells were infected with lentiviral reporter constructs. Cells<br />

were seeded either on top of Matrigel or without Matrigel in 384 well<br />

plates in E2-free media. After 3 days, cells were treated with either<br />

E2 (10-9M) or vehicle (0.1% ethanol). Fluc activity was assessed by<br />

bioluminescence imaging at 24, 48 and 72 hours following treatment<br />

using Xenogen IVIS Spectrum imaging system.<br />

Results: Initial findings showed differential transcriptional activities<br />

between the parental T47D/neo and T47D/PKCα cell lines at the basal<br />

level (without E2 treatment) in the 3D environment. Specifically,<br />

TFs involved in tumorigenesis (such as ETS1, STAT5) or the EMT<br />

process (such as SNAI1) were more highly activated in T47D/PKCα<br />

cells. Furthermore, E2 has a modulating effect on the activities of<br />

TFs in the two cell lines over time; observed both in the 2D and 3D<br />

environments. In particular, some TFs, such as Oct4, ETS1, and<br />

Stat5; were modulated (either induced or suppressed) by E2 only in<br />

T47D/PKCα cells.<br />

Conclusion: These findings suggest that overexpression of PKCα<br />

alters TF activity in T47D breast cancer cells both basally and in<br />

response to E2. This HTS technology allowed us to reveal the dynamic<br />

regulation of TF activation in PKCα overexpressing T47D breast<br />

cancer cells. We are now beginning to understand the mechanism<br />

whereby PKCα may mediate differential response to E2 dependent<br />

on the microenvironment. This HTS approach is likely to lead to the<br />

identification of new therapeutic targets.<br />

P6-04-25<br />

Genomic Deregulation and Therapeutic Role of Nemo-like Kinase<br />

in Luminal B <strong>Breast</strong> <strong>Cancer</strong><br />

Cao X, Qin L, Kim J-A, Tan Y, Wang X, Schiff R. Lester & Sue Smith<br />

<strong>Breast</strong> Center, Baylor College of Medicine, Houston, TX<br />

Luminal B breast tumors are notoriously known as high grade<br />

breast cancer prone to Tamoxifen resistance and early recurrence.<br />

Chromosomal abnormalities are a crucial class of drug targets<br />

in cancer which can be classified into numerical and structural<br />

rearrangements. Gene amplification is the result of numerical<br />

rearrangement that generates multiple copies of an otherwise normal<br />

human gene, such as Her2 amplification observed in breast cancer.<br />

Gene fusion is the result of structural rearrangement that fuses<br />

together pieces of two different genes, such as BCR-ABL gene fusion<br />

identified in leukemia.<br />

Due to their critical role in causing human cancers and their presence<br />

exclusively in the tumor tissues, gene amplifications and fusions<br />

provide highly specific drug targets with only minimal side effects.<br />

In order to identify these chromosome aberrations that can serve<br />

as new therapeutic targets in breast cancer, we developed a novel<br />

bioinformatics analysis called “copy number signature analysis”.<br />

This is based on the observation that driving gene fusions and their<br />

original (wild-type) genes are often amplified in fusion-positive<br />

tumors, or tumors without the fusions respectively. This suggests that<br />

gene fusions and amplifications may synergize to activate important<br />

oncogenes; if directly druggable, these aberrations will provide<br />

substantial opportunities for new therapies.<br />

Applying this analysis to the newly emerged genomic data from the<br />

<strong>Cancer</strong> Genome Atlas (TCGA) project nominated Nemo-like kinase<br />

(NLK) as a lead therapeutic target for invasive breast cancer. NLK<br />

encodes a serine-threonine mitogen activated protein kinase that<br />

plays a pivotal role in transforming growth factor beta pathway and<br />

embryo development. Genomic amplifications and rearrangements<br />

at NLK locus happen in 7%-9% of breast tumors, which may lead<br />

to deregulated serine-threonine kinase activity. Of note, these<br />

aberrations preferentially present in luminal B or Her2 positive<br />

breast tumors, suggesting their association with these high grade<br />

tumors. Most interestingly, transient inhibition of NLK through<br />

small RNA interference in multiple breast cancer cell lines harboring<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

502s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: General Session 6<br />

NLK deregulations, we observed potent inhibition of cell growth<br />

and capability of forming tumor-like colonies, as well as increased<br />

sensitivity to Tamoxifen treatment.<br />

These data suggest that NLK might be deregulated by genetic events<br />

which may promote growth-factor signaling and Tamoxifen resistant<br />

phenotype. Molecular assays detecting NLK chromosomal aberrations<br />

may serve as a predictive biomarker for endocrine resistance in breast<br />

cancer as well as selection of patients for appropriate treatment.<br />

P6-04-26<br />

Tamoxifen may block estrogen induced secretion of certain<br />

cytokines to interrupt tumor associated macrophage infiltration<br />

in breast cancer<br />

Ding J, Chen CM, Jin W, Shao Z, Wu J. <strong>Breast</strong> <strong>Cancer</strong> Institute, Fudan<br />

University Shanghai <strong>Cancer</strong> Center, Shanghai, China; Shanghai<br />

Medical College, Fudan University, Shanghai, China<br />

The role of tumor microenvironment during the initiation and<br />

progression of breast cancer is now realized to be of critical<br />

importance, both for understanding of the fundamental cancer<br />

biology and exploiting relatively new mechanism for breast cancer<br />

metastasis. Macrophage is a one of the most common components<br />

in the breast cancer microenvironment. Normally, the macrophage in<br />

tumor microenvironment is referred to tumor associated macrophage<br />

(TAM), which shares some attributes of alternatively activated<br />

macrophage. Many pre-clinical and clinical studies demonstrate an<br />

inverse correlation between TAM infiltration and patients’ prognosis<br />

indicating a macrophage supporting role for tumor progression. It is<br />

well documented that selective patients with breast cancer can benefit<br />

from anti-estrogen therapy. However, the mechanisms involved are<br />

still not fully elucidated. Our previous study indicates that highly<br />

tumorigenic breast cancer cell line MDA-MB-231 can educate<br />

monocyte differentiation into alternatively activated macrophage,<br />

while weakly tumorigenic cell line MCF-7 (without estrogen<br />

supplementation) can not. In the present study, we explored the effects<br />

of estrogen and tamoxifen on the secreting of certain cytokines, which<br />

are required for monocyte chemotaxis and differentiation. <strong>Breast</strong><br />

cancer cell lines with different estrogen receptor status were applied<br />

to estimate the level of cytokines (IL4, IL6, M-CSF, GM-CSF, and<br />

MCP-1) before and after cultured with 17β-estradiol, tamoxifen or<br />

both. We found that weakly invasive breast cancer cell lines (MCF-<br />

7, T47D, MDA-MB-468, and SKBR3) generally expressed lower<br />

levels of such cytokines compared with highly invasive breast cancer<br />

cell lines (MDA-MB-231, MDA-MB-231-BO, MDA-MB-231<br />

HM, Bcap 37, BT549, and Hs578T) at baseline. However, the<br />

discrepancies could be compensated partially by exposure to 10nM<br />

17β-estradiol in estrogen receptor (ER) positive cell lines (MCF-7<br />

and T47D). Moreover, the compensation was substantially blocked<br />

by 2µM tamoxifen. On the contrary, tamoxifen, alone, didn’t affect<br />

the secretion of such cytokines mentioned above in vitro. Based on<br />

these findings, we tentatively concluded that some patients with<br />

ER positive breast cancers benefiting from anti-estrogen therapy<br />

may partially attributes to blocking monocyte differentiation into<br />

TAM, which destroyed the tumor microenvironment. These findings<br />

suggest the future possibility of using TAM as a novel therapeutic<br />

target in patients with anti-estrogen resistance and primary triplenegative<br />

breast cancers (TNBC) with no effectively therapeutic<br />

measures nowadays, which are defined by lack of estrogen receptor,<br />

progesterone receptor and ERBB2 gene amplification, representing<br />

approximately 16% of all breast cancers.<br />

P6-04-27<br />

EBAG9 immunoreactivity is a potential prognostic factor for<br />

poor outcome of breast cancer patients with adjuvant tamoxifen<br />

therapy<br />

Shigekawa T, Ijichi N, Ikeda K, Miyazaki T, Horie-Inoue K, Shimizu C,<br />

Saji S, Aogi K, Tsuda H, Osaki A, Saeki T, Inoue S. Research Center<br />

for Genomic Medicine, Saitama Medical University; International<br />

Medical Center, Saitama Medical University; Tokyo Metropolitan<br />

<strong>Cancer</strong> and Infectious Disease Center, Komagome Hospital; National<br />

<strong>Cancer</strong> Center Hospital; Graduate School of Medicine, Kyoto<br />

University; National Shikoku <strong>Cancer</strong> Center; Graduate School of<br />

Medicine, The University of Tokyo<br />

Estrogen receptor-binding fragment associated gene 9 (EBAG9) is<br />

a primary estrogen responsive gene originally cloned from MCF-7<br />

human breast carcinoma cells. Upregulation of EBAG9 expression<br />

has been observed in several malignant tumors such as advanced<br />

breast cancers, indicating that EBAG9 may contribute to tumor<br />

progression. In the present study, the immunohistochemical analyses<br />

of EBAG9 were performed using the specimens from breast cancer<br />

patients treated with tamoxifen as adjuvant therapy. To investigate the<br />

association between EBAG9 expression and the therapeutic effect of<br />

tamoxifen in breast cancer, we used adjuvant tamoxifen-treated breast<br />

cancer patient tissues from 3 institutions. The study was approved<br />

by the institutional review board. Patients with distant metastases<br />

within 5 years after surgery followed by tamoxifen treatment were<br />

defined as relapse cases, whereas those without distant metastases<br />

were defined as relapse-free cases. The immunoreactivity of EBAG9<br />

was predominantly detected in the cytoplasm of breast cancer cells.<br />

Notably, EBAG9 immunoreactivity level was substantially elevated<br />

in the breast cancer samples from the relapse patients, and the positive<br />

immunoreactivity of EBAG9 was correlated with poor prognosis<br />

of the patients with adjuvant therapy. These results suggest that<br />

EBAG9 expression in tumor regions is associated with unfavorable<br />

prognosis of patients with adjuvant tamoxifen therapy and EBAG9<br />

immunoreactivity can serve as a biomarker for adjuvant tamoxifen<br />

efficacy.<br />

P6-04-28<br />

Changes in BAG-1 expression level affect the Tamoxifen-induced<br />

apoptosis and Gegitinb-induced apoptosis in breast cancer cells<br />

Lu S, Zhuang X, Liu H. Tianjin Medical University <strong>Cancer</strong> Institute<br />

and Hospital, Tianjin, China; Tianjin Medical University, Minstry of<br />

Education, Tianjin, China<br />

BAG-1 (bcl-2-associated athanogene-1) is a multifunctional<br />

protein which can interact with a number of molecules to regulate<br />

diverse biological processes including apoptosis, proliferation,<br />

signal transduction and transcription. BAG-1 protects cells from<br />

a wide range of apoptotic stimuli including radiation, hypoxia and<br />

chemotherapeutic agents and growth factor withdrawal. Therefore,<br />

BAG-1 may contribute to reduced cell death in cancer development,<br />

as well as to resistance to important therapeutic agents. Our study<br />

showed that the TAM sensitive cell line MCF-7 expressed lower<br />

levels of BAG-1 than the TAM resistant cell line MCF-7/TAMR,<br />

which was induced from MCF-7 cell line by culturing in medium<br />

containing 4-OH TAM (1×10- 7 M) for 6 months. Our study also<br />

found that down-regulation of BAG-1 significantly enhanced the<br />

sensitivity of the TAMR/MCF-7 cells to TAM treatment. Additionally,<br />

we used vectors carrying the individual BAG-1 isoforms to transfect<br />

the MCF-7 cells, and found that BAG-1 p50 was the only isoform<br />

that inhibited the TAM-induced apoptosis in the MCF-7 cells, while<br />

the other isoforms had little effect. Besides, we detected strong<br />

www.aacrjournals.org 503s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

expression of BAG-1 in several breast cancer cell lines including<br />

T47D, MCF-7, MDA-MB231 and SK-BR-3. We even observed a<br />

1.56 fold increase of EGFR in T47D cells transfected with BAG-1<br />

siRNA when compared with the negative control. To further explore<br />

the association between BAG-1 expression and Gefitinb response in<br />

breast cancer cell lines, we used Gefitinb of different concentrations<br />

to treat cells for 24 hours, 48 hours and 72 hours respectively. The<br />

results of cell viability assay indicated that the down-regulation of<br />

BAG-1 in breast cancer cell line T47D significantly enhanced the<br />

T47D cells’ sensitivity to Gefitinb-induced apoptosis.<br />

In summary, our results indicated that BAG-1 may have influence in<br />

the development of TAM resistance and Gefitinb resistance in breast<br />

cancer. However, our study was conducted in breast cancer cells in<br />

vitro; further studies on the relationship between BAG-1 expression<br />

and response to TAM and Gefitinb in a large cohort of breast cancer<br />

patients may be needed to confirm our findings.<br />

P6-04-29<br />

Vitamin D induces expression of estrogen receptor and restores<br />

endocrine therapy response in estrogen receptor-negative breast<br />

cancer<br />

<strong>San</strong>tos N, Diaz L, Ordaz D, Garcia J, Barrera D, Avila E, Halhali<br />

A, Medina H, Camacho J, Larrea F, Garcia R. Instituto Nacional<br />

de Ciencias Médicas y Nutrición Salvador Zubirán, México, DF,<br />

Mexico; Centro de Investigación y de Estudios Avanzados del I.P.N.,<br />

México, DF, Mexico<br />

Background: Approximately 30% of all breast tumors do not express<br />

estrogen receptor (ER) and patients with these tumors present poor<br />

prognosis and respond poorly to hormone therapy. Calcitriol through<br />

its vitamin D receptor (VDR) exerts antiproliferative, apoptotic and<br />

pro-differentiating effects in cancer. Calcitriol´s effects upon ER<br />

expression in breast cancer cells is controversial. Therefore, in order<br />

to clarify this issue, the aim of the present study was to determine if<br />

calcitriol induces ERα expression in ERα-negative breast cancer cells<br />

and could restore antiestrogen responses. The evaluation of calcitriol<br />

effects was performed in terms of proliferation and regulation of the<br />

following genes: Cyclin D1, involved in cell cycle, and Ether-à-go-go<br />

1 (Eag1), related to cell proliferation and tumor progression.<br />

Methods: Cultured cells derived from ERα-negative breast tumors<br />

and an established ERα-negative breast cancer cell line (SUM 229)<br />

were used in this study. These cells were treated with calcitriol<br />

and reverse transcription-PCR or western blotting analyses were<br />

performed to assess ERα expression. Growth assays with XTT were<br />

used to evaluate the antiproliferative response to the antiestrogens<br />

fulvestran and tamoxifen. Gene expression analysis for Cyclin D1 and<br />

Eag1 was evaluated by real time PCR in cells treated simultaneously<br />

with calcitriol plus estradiol or fulvestran.<br />

Results: The treatment with calcitriol in ER-negative breast cancer<br />

cells resulted in the induction of ERα. This effect was specifically<br />

mediated through the vitamin D3 receptor (VDR), since the VDR<br />

antagonist TEI-9647 effectively inhibited the ability of calcitriol to<br />

stimulate ERα gene expression. Consequently, the induction of ERα<br />

by calcitriol restores the response to antiestrogens in breast cancer<br />

cells by inhibiting cell proliferation. Co-treatment of calcitriol and<br />

antiestrogens down-regulated Cyclin D1 and Eag1 gene expression.<br />

Conclusion: Calcitriol induced the expression of ERα and restored<br />

antiestrogenic responses in ERα-negative breast cancer cells.<br />

Moreover, fulvestran down regulated mRNA expression of Cyclin D1<br />

and Eag1 when ERα-negative cells were pre-treated with calcitriol.<br />

These results suggest that the combined treatment with calcitriol and<br />

antiestrogens could be a new therapeutic strategy for ERα-negative<br />

breast cancer patients.<br />

P6-04-30<br />

COUP-TFII suppresses NFκB activation in endocrine-resistant<br />

breast cancer cells<br />

Litchfield LM, Klinge CM. University of Louisville School of<br />

Medicine, Louisville, KY<br />

COUP-TFII is an orphan nuclear receptor that functions as either a<br />

transcriptional activator or repressor. We previously reported that<br />

COUP-TFII expression is reduced in endocrine-resistant LCC9<br />

breast cancer cells and that overexpression of COUP-TFII restored<br />

the ability of the antiestrogen tamoxifen to inhibit cell proliferation.<br />

An increase in NFκB activity and p65/RELA expression has been<br />

reported in endocrine-resistant breast cancer cells. Because NFκB<br />

activation results in increased expression of anti-apoptotic and proproliferative<br />

genes, increased NFκB signaling promotes the survival<br />

of cancer cells. The goal of the current study was to further elucidate<br />

the mechanism by which COUP-TFII enables cells to maintain<br />

endocrine sensitivity and to determine if COUP-TFII can modulate<br />

NFκB activity in tamoxifen-resistant breast cancer cells. To examine<br />

whether COUP-TFII can suppress NFκB activity, MCF-7 (tamoxifensensitive,<br />

TAM-S) and LCC9 (tamoxifen-resistant, TAM-R) breast<br />

cancer cells were transfected with a NFκB luciferase reporter and<br />

treated with TNFα to activate NFκB. NFκB activity was elevated in<br />

LCC9 TAM-R cells and co-transfection with COUP-TFII reduced<br />

NFκB activity. COUP-TFII overexpression led to a statistically<br />

significant reduction in the expression of NFκB subunits RELA,<br />

RELB, NFKB1, and REL in LCC9 cells. Significantly reduced<br />

expression of NFκB target genes IL6, ICAM1, TNFAIP3, and CCL2<br />

was also evident upon COUP-TFII overexpression, reflecting the<br />

suppression of the NFκB pathway by COUP-TFII. Inhibition of NFκB<br />

activation by COUP-TFII also led to a reduction in cell viability in<br />

TAM-R cells in response to tamoxifen treatment. Together these<br />

data indicate a novel role for COUP-TFII in suppression of NFκB<br />

activity which may explain the ability of COUP-TFII to promote<br />

tamoxifen-sensitivity. COUP-TFII may be both a useful biomarker to<br />

predict tamoxifen-sensitivity as well as a target to restore endocrine<br />

sensitivity to resistant cells.<br />

Supported by NIEHS T32 ES011564 predoctoral fellowship to L.M.L.<br />

and Susan G. Komen for the Cure grant KG080365 to C.M.K.<br />

P6-05-01<br />

Evaluation of the prognostic significance of androgen receptor<br />

(AR) expression in relation to ER expression in breast cancer (BC)<br />

Gucalp A, Datko FM, Patil S, Hedvat CV, Kalinsky K, Hudis CA,<br />

Traina TA, Moynahan ME. Memorial Sloan-Kettering <strong>Cancer</strong> Center;<br />

Columbia University Medical Center<br />

Background: AR is expressed in 60-80% of invasive BC. Emerging<br />

data suggest that AR expression may have a prognostic role in BC<br />

irrespective of ER and that the androgen-signaling pathway may<br />

be involved in BC pathogenesis. We previously identified an ARdependent<br />

subset of ER/PR(-) BC and reported clinical benefit<br />

from AR-inhibition (Gucalp, ASCO 2012). We now describe the<br />

prevalence, clinicopathologic characteristics, and survival outcomes<br />

of AR(+) BC in relation to expression of ER in a single-institution<br />

retrospective cohort.<br />

Methods: We identified 590 patients (pts) with resectable BC (tumor<br />

>1cm) who had surgery at MSKCC from 1992-1996. IRB approval<br />

was obtained. Tissue microarray (TMA) construction/scoring has been<br />

described (Kalinsky CCR 2009). TMAs were stained, scanned and<br />

digitally scored (Aperio ScanScope XT) for nuclear steroid receptors<br />

(ER, AR), HER2, p53, and MIB1 (Ki67). Using digital quantification<br />

of tumor cell nuclear staining, pts were categorized into the following<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

504s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: General Session 6<br />

7 groups: ER dominant (ERD, both ER and AR >10% with ER>AR<br />

by >5%), ER only (ER>10%, AR 10%, 10%, AR>ER by >5%), AR<br />

only (AR>10%, ER


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

NVP-BEZ235. Interestingly, we observed that all AR-positive TNBC<br />

cell lines examined contain PIK3CA mutations (H1047R) suggesting<br />

selection for deregulation of the PI3K pathways during evolution of<br />

AR expressing breast cancers. Further investigation of a panel of<br />

AR-positive TNBC tumors showed that PIK3CA kinase mutations<br />

are a frequent event in this TNBC subtype (69.2% vs. 26.9%) and<br />

not just an artifact of the LAR cell line models. We show that PI3K<br />

pathway activation plays a role in resistance to AR antagonists,<br />

as there are elevated levels of activated AKT in residual tumors<br />

after bicalutamide treatment of xenograft tumors. Furthermore, we<br />

demonstrate that genetic/pharmacological targeting of AR synergizes<br />

with PI3K/mTOR inhibition in both two- and three-dimensional cell<br />

culture models. Moreover, the combination of bicalutamide with<br />

PI3K/mTOR inhibitors GDC0980 or NVP-BEZ235 decreased tumor<br />

volume in mouse xenograft studies . Our preclinical data provide<br />

the rationale for pre-selecting AR-positive TNBC patients for future<br />

clinical trials evaluating the combination of AR antagonists with<br />

PI3K/mTOR inhibitors.<br />

P6-05-04<br />

Expression of the Androgen Receptor in triple negative tumors<br />

and its modulation by receptor tyrosine kinases and downstream<br />

pathways<br />

Cuenca D, Montero JC, Morales JC, Prat A, Pandiella A, Ocana<br />

A. Albacete University Hospital, Albacete, Spain; <strong>Cancer</strong> Research<br />

Center, CIC-CSIC, Salamanca, Spain; Lineberger <strong>Cancer</strong> Center,<br />

University of North Carolina<br />

Background: Androgen receptor (AR) plays a central role in some<br />

tumor types like prostate cancer. Its expression and function in breast<br />

cancer is relatively unknown. Studies have linked tyrosine kinase (TK)<br />

receptors and their intracellular signaling pathways with AR status. In<br />

the present work we study the presence of AR in breast cancer human<br />

samples and how TKs can control its expression.<br />

Methods: Tumor samples from patients were used to assess the<br />

expression of AR by western-blot. Membrane and intracellular<br />

kinases were evaluated using phosphoprotein arrays. Cell lines<br />

representative of all breast cancer subtypes were used to assess the<br />

expression of the AR and other signaling proteins using western-blot<br />

and phosphoprotein arrays. Proliferation studies with several kinase<br />

inhibitors were measured by MTT assays. We also interrogated<br />

publicly available gene expression microarray data sets with annotated<br />

clinical-pathological data.<br />

Results: Across all the intrinsic subtypes of breast cancer, the<br />

expression of AR gene was found highly expressed in HER2-enriched<br />

and Luminal tumors and lowly expressed in Basal-like and/or triplenegative<br />

(TN) tumors. In TN tumors, PDGFRβ and EGFR were<br />

found highly activated, together with AKT and Erk1/2. A positive<br />

correlation was observed between activation of EGFR and PDGFRβ<br />

and the expression of AR. In vitro, AR protein expression varied<br />

across cell lines and was found more expressed in MCF7 (Luminal)<br />

and HS578T (TN), BT549 (TN), HCC70 (TN) and no expressed<br />

in HBL100 (TN) and MDA-MB231 (TN). Administration of the<br />

antiandrogen bicalutamide produced an anti-proliferative effect that<br />

was increased by association of an Erk1/2 inhibitor. Inhibition of the<br />

PI3K-mTOR pathway in BT549 and HS578T reduced the expression<br />

of AR but did not augment the action of bicalutamide. In TN tumors,<br />

AR gene and protein was found more expressed in older compared<br />

to younger patients.<br />

Conclusions: In breast cancer, AR is expressed and is controlled by<br />

receptor TKs through the PI3K-mTOR pathway. An increased antiproliferative<br />

effect is observed with the combination of bicalutamide<br />

and Erk1/2 inhibitors. AR expression correlates with age at diagnosis.<br />

P6-05-05<br />

Triple receptor comparison between primary breast cancer and<br />

metachronous or synchronous liver metastasis<br />

Tuyls S, Brouckaert O, Vanderstichele A, Vanderhaegen J, Amant<br />

F, Leunen K, Smeets A, Berteloot P, Van Limbergen E, Weltens C,<br />

Peeters S, Vanbeckevoort D, Floris G, Moerman P, Paridaens R,<br />

Wildiers H, Vergote I, Christiaens M-R, Neven P. University Hospitals<br />

Leuven, Belgium<br />

Background: Decisions about systemic treatment in women with<br />

metastatic breast cancer are currently based on the presence of<br />

estrogen receptors (ER), progesterone receptors (PR) or Human<br />

Epidermal growth factor Receptor 2 (HER2 receptors) in the primary<br />

tumor. Recently, several studies have reported significant discordances<br />

in ER, PR and HER2 status between the primary tumor and metastatic<br />

lesions and this may vary by metastatic site. Prognostic implications<br />

remain unclear although alterations in ER, PR and/or HER2 can<br />

influence metastatic management. This study represents one of the<br />

largest studies evaluating how frequent receptor discordances occur<br />

in liver metastasis, whether this alters therapeutic options and impacts<br />

prognosis.<br />

Patients and methods: 246 breast cancer patients with histological<br />

confirmed liver metastasis were analyzed in this retrospective study.<br />

Immunohistochemistry (IHC) and/or FISH were used to determine<br />

ER, PR and HER2 receptor status. We excluded patients when<br />

comparison between receptors of primary tumor and metastasis was<br />

impossible due to missing data (n=85), when liver metastasis did not<br />

originate from breast cancer (n=36) and when pathology was obtained<br />

from autopsy specimens (n=38).<br />

Results: 87 patients had matched tissue samples of primary tumor<br />

and liver metastasis with possible comparison of at least one of the<br />

receptors. Table 1 summarizes changes in ER, PR, HER2 between<br />

primary and metastatic lesion. Discordance in receptor status was<br />

associated with shorter time to death (63.3 months) compared to the<br />

concordant group (75.5 months).<br />

table1<br />

- to + + to - total discordance<br />

ER 2/12 (16.7%) 16/75 (21.3%) 18/87 (20.7% with 95% CI 12.8-30.7)<br />

PR 6/23 (26.1%) 32/63 (50.8%) 38/86 (44.2% with 95% CI 33.5-55.3)<br />

HER2 1/50 (2.0%) 0/13 (0%) 1/63 (1.6% with 95% CI 0.04-8.53)<br />

Conclusions: A significant proportion of ER and PR show discordance<br />

between primary tumor and liver metastasis. However we could not<br />

establish the same level of discordance for the HER2 receptors as<br />

in other studies. In general, only about 1 in 5 patients gained new<br />

(endocrine or targeted) therapeutic options. Tissue confirmation<br />

remains important to evaluate whether metastatic disease has become<br />

endocrine insensitive (in approximately 1 in 5 patients), avoiding<br />

unnecessary delay in chemotherapy. Discordance in receptors may<br />

be associated with inferior prognosis.<br />

P6-05-06<br />

Serum estradiol levels in postmenopausal ER+/PR- breast cancer<br />

are lower than those in postmenopausal ER+/PR+<br />

Yamamoto Y, Ibusuki M, Goto H, Murakami K, Iwase H. Kumamoto<br />

University Hospital, Kumamoto, Japan<br />

(Background) Activation of growth factor receptor (GFR) signaling<br />

is related with ER+/PR- breast cancer (BC). Proportion of ER+/PRphenotype<br />

in ER+ BC in postmenopausal patients is higher than that<br />

in premenopausal. However, this phenomenon cannot be explained<br />

by only GFR signaling activation. We hypothesized that ER+/PRphenotype<br />

in postmenopausal patients has at least two subtypes, one<br />

is related with activation of GFR signaling and the other is related<br />

with suppression of PR expression due to low estrogenic stimulation.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

506s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: General Session 6<br />

(Purpose) The aim of this study is to clarify out hypothesis.<br />

(Patients and methods) We measured serum E2 levels with<br />

radioimmuno assay (range of E2 levels : 1.3 to 50 pg/ml) in 154<br />

postmenopausal ER+ BC patients. Cut-off levels of ER and PR<br />

were used 10% or more expression in tumor cells. We analyzed<br />

the correlation between serum levels of E2 and clinicopathological<br />

factors including PR expression. We also examined the relationship<br />

between HER2 expression levels and ki67 labeling index in ER+/<br />

PR- BC. In addition, we analysed the difference in ki67 LI between<br />

in premenopausal patients (n=65) and in postmenopausal patients.<br />

(Results) Median serum levels of E2 in ER+/PR+ phenotype (6.7<br />

pg/ml) were significantly higher than in ER+/PR- phenotype (4.5<br />

pg/ml)(p=0.036). Serum levels of E2 was positively associated<br />

with body mass index (p=0.01). However, there was no significant<br />

difference between serum E2 levels and other clinicopathological<br />

factors including tumor size, nodal status, nuclear grade, levels of<br />

ER expression and ki67 LI. In ER+/PR- phenotype, ki67 LI were<br />

varied (Median: 7.5%, range: 0.5 to 85%). Ki67 LI in Postmenopausal<br />

patients with ER+/PR- phenotype was significantly lower than that<br />

in premenopausal patients with ER+/PR- phenotype (p=0.0022).<br />

Expression levels of HER2 was positively associated with ki67<br />

LI(p=0.002).<br />

(Conclusions) ER+/PR- cancer in postmenopausal patients may<br />

have two types of phenotype. One is highly proliferative phenotype<br />

that is stimulated with GFR signaling. The other is low proliferative<br />

one that PR expression is suppressed by without sufficient estrogen<br />

supply. Serum levels of E2 might affect development of ER+ BC in<br />

postmenopausal women, especially in case of no activation of GFR<br />

signaling.<br />

P6-05-07<br />

Amplified in breast cancer 1 and ankyrin repeat containing<br />

cofactor 1 mediate estrogen induced repression of the ErbB2<br />

oncogene<br />

Garee JP, Riegel AT. Georgetown University, Washington, DC<br />

The oncogene amplified in breast cancer 1 (AIB1/SRC3) is a nuclear<br />

receptor coactivator and plays an important role in the regulation<br />

of gene transcription. Additionally, AIB1 is important in malignant<br />

progression as AIB1 has been shown to be a gene amplified in breast<br />

cancer and is also over-expressed in a variety of cancer types. Previous<br />

work from our laboratory has identified a splice variant of AIB1<br />

termed AIB1-∆4. This splice variant arises from the splicing of exon 4<br />

from the mature mRNA. This leads to the formation of an N terminally<br />

truncated form of the AIB1 protein, which is a more potent coactivator<br />

of steroid dependent transcription compared to the full length AIB1.<br />

This suggests that the N-terminal fragment of AIB1, which is not<br />

present in AIB1-∆4, may act to dampen AIB1 mediated transcriptional<br />

activation. The presence of this N terminal region suggests this region<br />

may be a site of interaction with a repressive protein, which could<br />

interact with AIB1 and not AIB1-∆4. We have identified ANCO1 as<br />

an N terminal AIB1 binding protein that acts to repress AIB1 mediated<br />

transcriptional activity. Co-immunoprecipitation experiments have<br />

indicated interaction between AIB1 and ANCO1 but not between<br />

AIB1-∆4 and ANCO1 indicating ANCO1 binds in the N terminal<br />

region of AIB1. AIB1 has a clinical implications and association<br />

with the oncogene ErbB2, which is an estrogen repressed gene with<br />

hormone effect acting through an estrogen response element (ERE)<br />

in intron 1 of the ErbB2 gene. We wanted to determine if the AIB1/<br />

ANCO1 complex played a role in hormone regulation of ErbB2.<br />

Silencing of AIB1 and ANCO1 by shRNA led to a reversal of<br />

estrogen-induced repression of ErbB2 indicating this protein complex<br />

may play a role in mediating this repression. Examining binding of<br />

these proteins to the intronic ERE of ErbB2 we determined that AIB1<br />

and ANCO1 are recruited in the presence of estrogen. Additionally,<br />

repressive proteins such as HDAC3, HDAC4 and N-CoR are recruited<br />

along with the AIB1/ANCO1 complex. While AIB1-∆4 can still be<br />

recruited to the intronic ERE no additional repressive proteins are<br />

recruited. This suggests a mechanism by which the AIB1/ANCO1<br />

complex can recruit repressive proteins to repress ErbB2 gene<br />

transcription, while AIB1-∆4 can escape this repressive mechanism.<br />

This may potentially have interesting clinical implications, as high<br />

levels of AIB1-∆4 may be responsible for increasing the expression<br />

levels of the ErbB2 oncogene.<br />

P6-05-08<br />

Significant heterogeneity in ERalpha and PR receptor status in<br />

different distant breast cancer metastases of the same patient<br />

Hoefnagel LDC, van der Groep P, Broers JE, van der Wall E, van<br />

Diest PJ. UMC Utrecht, Utrecht, Netherlands<br />

Purpose: Receptor conversion for estrogen receptor alpha (ERα),<br />

progesterone receptor (PR) and human epidermal growth factor<br />

receptor 2 (HER2) in single distant breast cancer metastases has<br />

been described before. However, most patients develop multiple<br />

metastases. Heterogeneity in receptor status between different<br />

metastases of the same patient has not been studied, probably since<br />

such material is very rare. The consequence of such heterogeneity<br />

would mean that as many metastases as possible need to be biopsied<br />

to re-assess receptor status.<br />

We therefore aimed to study heterogeneity of receptor status between<br />

different distant metastases of the same patient in a relatively large<br />

group by re-staining all primary tumors and metastases with current<br />

optimal immunohistochemical methods on full sections.<br />

Material and methods: Formalin fixed paraffin embedded tissue of<br />

primary breast carcinomas and corresponding multiple (≥2) distant<br />

metastases from 38 female patients were collected from several<br />

pathology departments in The Netherlands and stained for ERα, PR<br />

and HER2 by IHC. We evaluated the heterogeneity of receptor status<br />

between the metastases and the correlation with the primary tumor.<br />

Results: In the 4 cases that showed ER conversion, 2 (50%) showed<br />

heterogeneity in receptor expression between different metastases<br />

of the same patient. Of the 17 cases that showed PR conversion, 12<br />

cases (71%) showed heterogeneity in expression between metastases.<br />

There were no changes in the receptor profile of primary breast cancer<br />

to their metastases for HER2 in this group.<br />

Conclusion: In a significant number of metastatic breast cancer<br />

patients, there is heterogeneity in ER and PR receptor status between<br />

different breast cancer metastases of the same patient. This implies<br />

that as many metastases as possible need to be biopsied to re-assess<br />

receptor status and set the optimal indication for hormonal therapy in<br />

these patients. Alternatively, non-invasive assessment of the receptor<br />

status by molecular imaging may be an easier future way to assess<br />

receptor status of all metastatic lesions.<br />

www.aacrjournals.org 507s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P6-05-09<br />

Unravelling the global effect of estrogen on breast cancer cell<br />

proteome using quantitative proteomics.<br />

Pavlou MP, Drabovich AP, Dimitromanolakis A, Diamandis EP.<br />

University of Toronto, ON, Canada; Mount Sinai Hospital, Toronto,<br />

ON, Canada; University Health Network, Toronto, ON, Canada;<br />

Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto,<br />

ON, Canada<br />

Estrogens exert their function through genomic and non-genomic<br />

pathways mediated mainly by estrogen receptors. Estrogen signalling<br />

is highly complex and once activated initiates cancer cell proliferation<br />

and survival, playing a pivotal role in breast cancer development and<br />

progression. The identification of estrogen regulated genes is the<br />

first step towards elucidating the mechanisms of estrogen function.<br />

Although numerous studies have investigated the effect of estradiol<br />

at the mRNA level, assessment of global changes at the protein level<br />

has been limited mainly due to technological limitations.<br />

Here, we present the most extensive proteomic study in the quest of<br />

proteins whose expression is regulated or associated with estradiol<br />

action. To facilitate accurate quantification of proteins, we first<br />

developed and optimized a proteomic protocol for cell lysis and<br />

sample preparation, based on a mass spectrometry-compatible<br />

detergent. Following our protocol coupled to stable isotope labelling<br />

with amino acids in cell culture (SILAC) of MCF-7 breast cancer<br />

cells, we quantified approximately 4,000 proteins and identified 153<br />

proteins differentially expressed upon estradiol stimulation. Notably,<br />

known estrogen regulated proteins such as trefoil 1(TFF1) and<br />

progesterone receptor (PGR) showed a four- and three-fold increase<br />

respectively, at 48 hours after estradiol stimulation. Forty eight of<br />

153 differentially expressed proteins have been found to contain<br />

estrogen receptor elements (EREs) indicating direct regulation by<br />

estradiol. Protein-protein network analysis (STRING) revealed an<br />

extensive protein network related to cell proliferation. Differential<br />

expression of proteins was verified in three estrogen receptor positive<br />

breast cancer cell lines (MCF-7, HCC-1428, BT-483) using a targeted<br />

mass spectrometry-based quantitative approach; selected reaction<br />

monitoring (SRM). A multiplex SRM assay was developed for the<br />

simultaneous quantification of 56 proteins. The same assay was<br />

utilized to study the kinetics of these proteins in time and in presence<br />

of inhibitors of estrogen receptor facilitating the distinction between<br />

direct and indirect protein targets.<br />

To our knowledge, such an extensive network of estrogenregulated<br />

genes has never been previously studied at the protein<br />

level. Interestingly, numerous differentially expressed proteins<br />

identified in the present study have not been connected to estrogen<br />

signalling or breast cancer before. The role of these proteins in breast<br />

carcinogenesis and their potential as therapeutic targets warrants<br />

further investigation.<br />

P6-05-10<br />

Progestins exert divergent growth effects and regulate tumorunique<br />

gene cohorts in patient derived breast cancer xenografts<br />

Sartorius CA, Finlay-Schultz J, Li C, Rosen RB, Hendricks P, Wisell<br />

J, Finlayson C, Elias A, Kabos P. University of Colorado Denver<br />

Anschutz Medical Campus, Aurora, CO<br />

Background: Progesterone is an important hormone for development<br />

and normal function of the breast; however, its role in established<br />

breast cancers is less clear. Progestins have been implicated in<br />

regulating tumor cell growth, signaling, differentiation state, and<br />

stem/progenitor properties in breast cancer cells. High dose progestin<br />

treatment can be an effective therapy for some advanced breast tumors,<br />

through an unknown mechanism. Progesterone receptors (PRs) are<br />

considered positive prognostic indicators, although their levels vary<br />

considerably among luminal subtype breast tumors. Progestin/PR<br />

regulated genes in tumor cells have only been determined in cultured<br />

breast cancer cells and mouse mammary tumor models. Here we<br />

define tumor unique progestin-dependent growth effects and gene<br />

regulation profiles in patient-derived luminal breast tumor xenografts.<br />

Methods: For these studies, three luminal estrogen receptor<br />

(ER)+PR+ transplantable xenografts were used that were derived<br />

from one pleural effusion and two previously untreated primary<br />

tumors. All three tumors were estrogen dependent to various degrees<br />

and contained variable levels of ER (5-90%) and PR (5-90%).<br />

Tumors were grown in vivo under continuous placebo, estrogen,<br />

or estrogen plus progestin conditions for 8-10 weeks and growth<br />

parameters monitored. Gene expression profiles were determined<br />

from dissected tumors using Affymetrix® GeneChip human gene 1.1<br />

ST microarrays and results analyzed using Partek Genomics Suite<br />

software. Specific gene regulation was confirmed by q-RT-PCR and<br />

immunohistochemistry.<br />

Results: Progestins had an inhibitory, neutral, and stimulatory effect<br />

on estrogen dependent growth in each of the three tumor lines.<br />

Accordingly, there was relatively little overlap in PR regulated genes<br />

between the three tumors. In the tumor in which progestin treatment<br />

had a potent anti-tumor effect, progestins were strong repressors of<br />

estrogen/ER-regulated genes. Gene expression patterns between<br />

the natural hormone progesterone and the synthetic drug MPA were<br />

compared in one tumor line and found to be mostly similar. The<br />

percent of PR+ tumor cells may have influenced the potency of gene<br />

regulation. Unique progestin regulated genes were discovered in this<br />

human tumor model system that were involved in immune response,<br />

cytokine signaling, and stem/progenitor cell features.<br />

Conclusions: Women with breast cancer are exposed to progestins<br />

naturally or through hormonal therapies and these may have a<br />

profound effect on tumor biology. In some tumors, progestins are<br />

potently anti-proliferative, while in others they elicit the opposite<br />

effect and potentiate estrogen induced tumor growth. The diversity of<br />

progestin-regulated genes in each tumor underscores the hormone’s<br />

context dependent effects. Determining progestin dependent gene<br />

signatures in patient tumors may pinpoint appropriate candidates for<br />

progestin therapy in advanced tumors and/or provide a prognostic<br />

tool for predicting tumor progression.<br />

Funded by: NIH R01 CA140985 (C.A.S.), the Grohne Fund (P.K.),<br />

and the University of Colorado <strong>Cancer</strong> Center (C.A.S., P.K.)<br />

P6-05-11<br />

Leptin and Leptin receptor expression in human breast cancer<br />

Wazir U, Al Sarakbi W, Jiang WG, Mokbel K. The London <strong>Breast</strong><br />

Institute, The Princess Grace Hospital, London, United Kingdom;<br />

Cardiff University-Peking University Oncology Joint Institute,<br />

Cardiff, United Kingdom<br />

INTRODUCTION: Leptin is a protein that plays a key role in<br />

regulating energy intake and energy expenditure, including appetite<br />

and metabolism. It is one of the most important adipose derived<br />

hormones and has been suggested to act as a growth factor in<br />

neoplasms. Obesity has been linked to increased risk of breast<br />

cancer in postmenopausal women. Increased peripheral production<br />

of oestrogen has been regarded as the main cause for this association,<br />

but other factors affecting fat metabolism may be implicated. There<br />

is recent evidence that Leptin stimulates cancer cell proliferation in<br />

MCF-7 cancer cells. Both Cox2 and hTERT have been proposed as<br />

mediators of Leptin’s carcinogenic role.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

508s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: General Session 6<br />

Our objective was to determine, using quantitative PCR, whether<br />

the mRNA expression levels of Leptin and Leptin receptor were<br />

consistent with a tumour proliferative function and whether levels<br />

from these genes were positively correlated with clinical outcome<br />

in breast cancer.<br />

METHODS: A total of 153 samples were analysed. The levels<br />

of transcription of Leptin and its receptor were determined using<br />

quantitative PCR and normalised against (CK19). Transcript levels<br />

within breast cancer specimens were compared to normal background<br />

tissues and analysed against conventional pathological parameters<br />

and clinical outcome over a 10 year follow-up period.<br />

RESULTS: The levels of Leptin and Leptin receptor mRNA were<br />

significantly higher in malignant samples (p = 0.0011 and 0.0014<br />

respectively). This difference remained statistically significant<br />

when comparing levels between normal samples and all malignant<br />

subgroups according to their NPI, grade, stage, local recurrence, and<br />

distant metastasis in both ER receptor positive and negative samples.<br />

It was also present across all histological types for both Leptin and<br />

its receptor.<br />

There was no significant change in mRNA transcription levels<br />

correlating with tumour grade, stage, disease free survival, or<br />

development of loco regional recurrence.<br />

There was also no statistically significant difference in levels of<br />

expression of both Leptin and its receptor when comparing ER<br />

positive to negative patients (p= 0.74 and 0.27 respectively).<br />

Leptin levels showed a significantly positive correlation with Leptin<br />

receptors (r=0.504, 0.0000000222). We have observed no significant<br />

correlations between Leptin and hTERT or Cox2.<br />

CONCLUSION: This study demonstrates compelling evidence that<br />

both Leptin and Leptin receptor mRNA transcription levels are more<br />

readily expressed in cancerous tissues providing further evidence<br />

that it plays a significant role in the process of breast carcinogenesis.<br />

However, Leptin levels did nor correlate with tumor’s stage, clinical<br />

outcome, hTERT or Cox-2 levels.<br />

P6-05-12<br />

Lack of Frequent Estrogen Receptor Mutation in Primary <strong>Breast</strong><br />

Tumors<br />

Oesterreich S, Kitchens C, Gavin P, Wu C-C, Riehle K, Coarfa C,<br />

Edwards D, Schiff R, Milosavljevic A, Lee A. University of Pittsburgh<br />

<strong>Cancer</strong> Institure, Pittsburgh, PA; Baylor College of Medicine,<br />

Houston, TX; NSABP, Pittsburgh, PA<br />

The estrogen receptor alpha (ESR1) is a critical driver of breast<br />

tumorigenesis, and as such has been a target for therapy for many<br />

years. Early reports using <strong>San</strong>ger sequencing and conformational<br />

assays reported that there were little if any mutations in ER-positive<br />

tumors, although a few somatic mutations have recently been described<br />

in tumors and cell lines. We set out to validate the authenticity of<br />

these reported somatic mutations by performing analysis of ER<br />

DNA sequence variants (DSVs) in 66 ER+ breast tumors, and 39<br />

breast cancer cell lines. We utilized a combined approach of target<br />

capture sequencing of ESR1, and subsequent testing for novel and<br />

previously identified DSVs using an orthogonal mass spectrometry<br />

based sequencing approach. A DNA capture approach was designed<br />

to capture all exons, flanking splice sites, and the 3’UTR of ESR1.<br />

DNA was captured and sequenced using SOLiD.<br />

Using stringency with a cut-off with at least 2 variant reads, variants<br />

in at least 20% of the reads, and requiring evidence for the variants in<br />

both strands, we identified 4 previously reported SNPs (rs2077647;<br />

rs46432; rs1801132; rs2228480), and two previously reported somatic<br />

mutations H6Y and N532Y. We developed mass-spectrometry assays,<br />

including controls, for these DSVs, and in addition, we included 10<br />

DSVs which had previously been reported as somatic mutations in<br />

breast (and other) cancer. Applying mass-spec based analysis to breast<br />

tumors and cell lines, we were able to confirm previously reported<br />

SNPs, and one previously reported mutations (H6Y). This mutation<br />

was found in one breast tumor and one cell line. None of the other<br />

reported somatic mutations could be confirmed in tumors or cell<br />

lines. Functional assays on the ESR1H6Y DSV failed to identify<br />

differences to wildtype receptor. The lack of a phenotype, and the<br />

infrequent occurrence of this DSV do not support a major driver role<br />

for ESR1H6Y.<br />

This analysis suggests that ESR1 mutations are a rare event in ER+<br />

primary breast cancer.<br />

P6-05-13<br />

ESR1 gene amplification in breast cancer: influence of RNAse<br />

treatment on FISH results<br />

Moelans CB, Holst F, Hellwinkel O, Simon R, van Diest PJ. University<br />

Medical Center Utrecht, Netherlands; University Medical Center<br />

Hamburg Eppendorf, Hamburg, Germany<br />

Introduction The frequency of estrogen receptor 1 (ESR1)<br />

amplification in breast cancer has long been controversial. High<br />

frequencies of amplification were more likely to be reported when<br />

measured by FISH compared to other methods such as qPCR, CGH<br />

and multiplex-ligation dependent probe amplification (MLPA). It was<br />

recently reported that HSR-like FISH clusters were RNAse sensitive<br />

which could lead to overestimation of ESR1 amplification frequency<br />

by FISH. We therefore compared copy numbers by FISH without<br />

and with RNAse treatment with MLPA in 53 tumor samples from 24<br />

tumors with ESR1 copy number increase by FISH.<br />

Methods Seventy five tumour areas (for each tumor with increased<br />

ESR1 copy number at least one area with the highest amplified copy<br />

number and cell density by FISH (Zytovysion)) from 38 breast cancers<br />

were laser microdissected and subjected to MLPA (kit P078-B1).<br />

Twenty four of these tumors (15 amplified and 9 gained; 53 areas)<br />

were also treated with RNAse A to compare copy numbers without<br />

and with RNAse treatment (blinded).<br />

Results RNAse treatment removed cloudy clusters but did not<br />

significantly change ESR1 gene copy numbers (p=0.47; average copy<br />

number was 4.60 pre- and 4.44 post-RNAse). However, CEP6 copy<br />

numbers were slightly higher after RNAse treatment (p=0.09; average<br />

copy number 2.17 pre- and 2.23 post-RNAse) leading to a significant<br />

decrease in ESR1/CEP6 ratios (p=0.006; average ratio 2.16 pre- and<br />

1.98 post-RNAse). 12/15 FISH-amplified cases were still amplified<br />

after RNAse treatment, whereas three cases were re-classified as<br />

gains (cut-off ratio 1.3). 7/9 FISH-gained cases remained gained<br />

after RNAse treatment, one case was reclassified as amplified and<br />

the other case as non-amplified. Spearman rho correlation between<br />

MLPA and FISH prior and after RNAse treatment was 0.57 and 0.56,<br />

respectively (both p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Also, mosaic heterogeneity (cell-to-cell variation) was frequently<br />

observed by FISH (present in 60% of the samples in this study).<br />

Conclusion Cloudy clusters by FISH are RNAse sensitive and could<br />

lead to overestimation of copy numbers. However, after RNAse<br />

treatment, ESR1 copy numbers remain increased. These copy number<br />

increases could at least in some cases be confirmed by MLPA,<br />

suggesting that they are DNA-based. ESR1 copy number increases<br />

seem to occur quite frequently in breast cancer, but the levels are in<br />

most cases not high enough to designate the sample as “amplified”<br />

by all techniques. PCR-based methods could be influenced by<br />

heterogeneity or contamination with normal cells and reported results<br />

depend on the cut-offs used and on the definition of amplification.<br />

P6-05-14<br />

Estrogen-induced genes in ductal carcinoma in situ(DCIS): their<br />

comparison with invasive ductal carcinoma.<br />

Ebata A, Suzuki T, Takagi K, Miki Y, Onodera Y, Nakamura Y,<br />

Fujishima F, Ishida K, Watanabe M, Tamaki K, Ishida T, Ohuchi N,<br />

Sasano H. Tohoku University Graduate School of Medicine, Sendai,<br />

Miyagi, Japan; Tohoku University Hospital, Sendai, Miyagi, Japan<br />

It is well known that estrogens play important roles in both the<br />

pathogenesis and development of invasive ductal carcinoma (IDC) of<br />

human breast. However, molecular features of estrogen actions have<br />

remained largely unclear in pure ductal carcinoma in situ (pDCIS),<br />

regarded as a precursor lesion of many IDCs. This is partly due to<br />

the fact that gene expression profiles of estrogen-responsive genes<br />

have not been examined in pDCIS. Therefore, we first examined the<br />

profiles of estrogen-induced genes in estrogen receptor (ER)-positive<br />

pDCIS and DCIS (DCIS-c) and IDC (IDC-c) components of IDC<br />

cases (n = 4, respectively) by microarray analysis. Estrogen-induced<br />

genes identified in this study were tentatively classified into three<br />

different groups in the hierarchical clustering analysis, and 33%<br />

of the genes were predominantly expressed in pDCIS rather than<br />

DCIS-c or IDC-c cases. Among these genes, the status of MYB<br />

(c-MYB), RBBP7 (RbAp46) and BIRC5 (survivin) expression in<br />

carcinoma cells was significantly higher in ER-positive pDCIS(n =<br />

53) than that in ER-positive DCIS-c (n = 27) or IDC-c (n = 27) by<br />

subsequent immunohistochemical analysis of the corresponding genes<br />

(P < 0.0001, P = 0.03 and P = 0.0003, respectively). In particular,<br />

the status of c-MYB immunoreactivity was inversely (P = 0.006)<br />

correlated with Ki-67 in the pDCIS cases. These results suggest<br />

that expression profiles of estrogen-induced genes in pDCIS may be<br />

different from those in IDC, and c-MYB, RbAp46 and survivin may<br />

play particularly important roles among estrogen induced genes in<br />

ER-positive pDCIS.<br />

P6-05-15<br />

Glucocorticoids has diverse effect on the tumorigenicity in<br />

estrogen receptor positive and estrogen recptor negative cancer<br />

cells<br />

Gao M, Yeh L-C, Chang C-P, Cheng A-L, Lu Y-S. National Taiwan<br />

University Hospital, Taipei, Taiwan<br />

Background: Glucocorticoids (GCs) exerts its biologic effects via<br />

GC receptor (GR), which has been linked to signal transduction<br />

pathways inducing apoptosis in many hematological malignancies.<br />

Generally, GCs are considered not affecting the growth of gross<br />

tumor of most non-hematological solid tumor. However, a recent<br />

study using meta-analysis of primary breast tumor gene expression<br />

from 1378 early breast cancer patients with long-term follow-up,<br />

showing that high levels of GR expression significantly correlated<br />

with shorter relapse-free survival in estrogen receptor (ER) negative<br />

patients. On the contrary, in ER+ breast cancer patients, high levels<br />

of GR expression in tumors were significantly associated with better<br />

outcome relative to low levels of GR expression. (<strong>Cancer</strong> Res. 2011,<br />

15;71: 6360-70). We hypothesized that GC has differential effect on<br />

the tumorigenicity in ER+ and ER- breast cancer cells.<br />

Methods: Twelve ER- breast cancer cell lines and 5 ER+ breast<br />

cancer cell lines were tested. MTT assay, clonogenic assay and soft<br />

agar assay was performed with cancer cells treated with or without<br />

dexamethasone (DEX). Flow cytometry were applied to detect the<br />

effect of DEX on the expression of cancer stem cell markers after<br />

treatment with DEX. Nude mice tail vein injection assay of cancer<br />

metastasis was performed to evaluate the in vivo tumorigenicity.<br />

Transfection with ER expression vector to MDA-MB231, a triple<br />

negative cancer cell, by lipofectamine was carried to determine the<br />

role of ER in this scenario.<br />

Results. DEX significantly enhanced colony forming ability and/or<br />

tumorigenicity in 12 ER- breast cancer cell lines. However, DEX<br />

did not affect the proliferation rate of these cancer cells according<br />

to MTT assay. We further explored whether DEX could affect the<br />

population of cancer stem-like cell, and thereby resulted in increase<br />

of tumorigenicity. By flow cytometry study, we found treatment of<br />

DEX significantly increased the number stem cell marker + (for<br />

examples, ALDH+) cells in ER- cancer cell lines, but not in ER+<br />

cell lines. Sorting and collection of these stem cell marker + cells<br />

demonstrated that they had much higher colony forming efficiency<br />

and tumorigenicity. Further, in nude mice model by tail vein<br />

injection with MDA-MB-231-Lu2 (derived from lung metastasis of<br />

MDA-MB-231 cells after two in vivo passages in nude mice using<br />

intracardiac injection), treatment of DEX significantly increased the<br />

lung metastatic foci number and size. In contrast, DEX significantly<br />

decreased the colony forming ability and tumorigenicity in 5 ER+<br />

cancer cell lines. Short term exposure (72hr) of DEX resulted in<br />

increased p21 expression and cell cycle G1 arrest in MCF7, an ER+<br />

cancer cell. Prolong incubation of MCF7 cells with DEX (more than<br />

10 days) induced cell apoptosis. When MDA-MB231 cells were<br />

stably transfected with ER, MDA-MB231/ER cells showed similar<br />

phenotype as those of ER+ cancer cell lines, i.e., DEX decreased<br />

the clonogenic and tumorigenicity ability. Studies on underlying<br />

mechanisms are ongoing, and preliminary result showed that there<br />

is cross talk between ER and GR.<br />

Conclusions: GC increased tumorigenicity in ER- breast cancer cells,<br />

but decrease tumorigenicity in ER+ cancer cells.<br />

P6-06-01<br />

Lobular involution reduces breast cancer risk through<br />

downregulation of invasive and proliferative cellular processes<br />

Radisky DC, Visscher DW, Stallings-Mann ML, Frost MH, Allers TM,<br />

Degnim AC, Hartmann LC. Mayo Clinic, Jacksonville, FL; Mayo<br />

Clinic, Rochester, MN<br />

Background: Age-related lobular involution (LI) is a physiological<br />

process, usually accelerated with menopause, in which the epithelial<br />

tissue of the breast gradually regresses. Our previous analysis of a<br />

cohort of more than 14,000 women who had a benign breast biopsy<br />

at the Mayo Clinic revealed that women who had undergone LI were<br />

at significantly reduced risk of subsequent breast cancer development.<br />

In our studies of postmenopausal women specifically, we found<br />

that more than 40% had not completed the process of LI, and that<br />

the increased breast cancer incidence in older women was strongly<br />

associated with incomplete LI. We sought to determine the potential<br />

protective mechanistic effects of lobular involution.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

510s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: General Session 6<br />

Methods: We generated primary human mammary epithelial cell lines<br />

(HMECs) from breast biopsies of women who showed no LI (N=6),<br />

partial LI (N=7), or complete LI (N=6). These cell lines were analyzed<br />

for proliferation in culture and profiled for genomic signatures using<br />

Affymetrix gene arrays. We also analyzed RNA isolated from a<br />

cohort of archived formalin-fixed, paraffin-embedded (FFPE) benign<br />

breast biopsy samples from postmenopausal women who had either<br />

completed the process of LI (N=4) or who had not initiated LI (N=8)<br />

for genomic signatures using Whole-Genome DASL Assays.<br />

Results: We found that LI was associated with decreased cellular<br />

proliferation and inhibition of processes associated with tumor<br />

development and progression. Specifically, we found that HMECs<br />

derived from benign breast biopsies in which LI was not evident<br />

showed increased proliferation and elevated expression of Twist<br />

(P=0.002), a transcription factor associated with epithelialmesenchymal<br />

transition (EMT) and breast cancer development. We<br />

are assessing differentially expressed genes for association with breast<br />

cancer risk using a nested case:control group of benign breast biopsies<br />

from women in our cohort who subsequently developed breast cancer<br />

(N=85) vs those who did not (N=142) and for whom LI status at the<br />

time of initial biopsy was known.<br />

Conclusions: LI may decrease cancer risk through inhibition of key<br />

tumorigenic processes. Identification of processes associated with<br />

the physiologic risk reduction seen with LI may provide insight into<br />

novel avenues for risk reduction.<br />

P6-06-02<br />

Characterization of an exosome-associated apoptosis-inducing<br />

activity produced by triple negative breast cancer cells.<br />

Georgoulia NE, Iliopoulos D, Mitchison TJ. Harvard Medical School,<br />

Boston, MA; Dana Farber <strong>Cancer</strong> Institute, Boston, MA<br />

Purpose: We sought to investigate whether breast cancer cells can<br />

suppress the proliferation of other breast cancer cells in order to<br />

understand the biological significance of cell heterogeneity in breast<br />

cancer.<br />

Materials and Methods: 20 breast cancer cell lines, corresponding<br />

to all molecular profiles of breast cancer, were cultured in a 20x20<br />

conditioned medium protocol. Cell proliferation and apoptosis were<br />

evaluated by EdU (5-ethynyl-2’-deoxyuridine)<br />

and PARP (Poly ADP ribose) cleaved labeling respectively, and<br />

documented by fluorescence microscopy. Serum free conditioned<br />

media (CM) were prepared from all the cell lines, 24hrs of incubation,<br />

and screened for their anti-proliferative effect against the different<br />

breast cancer tumor cells. Active media were ultra-centrifuged and<br />

both the supernatants as well as the pellets were assessed for the<br />

anti-proliferative activity.<br />

Results: Using this 20Χ20 matrix, CM obtained from two out of<br />

six triple negative breast cancer cells displayed anti-proliferative<br />

activity against the majority of cell lines, irrespectively of their<br />

molecular phenotype including triple negative cells. No significant<br />

anti-proliferative activity was observed in CMs from luminal or<br />

HER2(+) cell lines. However the two triple negative CMs induced a<br />

significant decrease in the cell lines’ proliferation index, which was<br />

associated with a concomitant increase in cells’ apoptotic index.<br />

Ultracentrifugation revealed that the apoptosis-inducing activity was<br />

retained in the pellet but not the supernatant of the CM. Negative<br />

staining electron microscopy (EM) revealed the presence of exosomes<br />

in the CM’s ultra-centrifuged pellet, which stained positive for cd63<br />

surface protein marker using an immunogold assay.<br />

Conclusions: These data strongly suggest that some triple negative<br />

breast cancer cell lines posses exosome-mediated apoptosis-inducing<br />

activity. This activity may have biological and clinical relevance<br />

since it could be a factor participating in the selection of tumor<br />

cell subpopulations of different degrees of aggressiveness. Further<br />

proteomic and microRNA profiling of the exosomes is ongoing in<br />

order to characterize this exosome-mediated apoptosis-inducing<br />

activity.<br />

P6-06-03<br />

Caveolin-1: A potential mediator of RhoC GTPase driven<br />

Inflammatory breast cancer cell invasion.<br />

Joglekar M, van Golen K. University of Delaware, Newark, Delaware<br />

The proposed study focuses on Inflammatory <strong>Breast</strong> <strong>Cancer</strong> (IBC),<br />

which is one of the most aggressive forms of locally advanced<br />

breast cancer. IBC is an understudied disease in terms of identifying<br />

various molecular entities that could potentially be responsible for<br />

the aggressiveness of this disease. Recent advancements reveal that<br />

IBC has a unique molecular profile when compared with non-IBC.<br />

Caveolin-1 is found to have opposite expression pattern in IBC<br />

versus non-IBC. Our lab has shown that Caveolin-1 is found to be<br />

over-expressed in IBC whereas in non-IBC its expression is much<br />

reduced compared to normal mammary epithelial cells.<br />

Caveolin-1 is known to be associated with specialized lipid rafts<br />

in the plasma membrane called ‘caveolae’. ‘Caveolin scaffolding<br />

domain (CSD)’ from amino acids 82-101 concentrates variety of<br />

signaling molecules such as growth factor receptors, protein kinases,<br />

heterotrimeric G proteins and Rho GTPases. Thus it is known as a<br />

potential regulator of many signaling pathways. Rho GTPases are<br />

members of the Ras superfamily of small GTP binding proteins<br />

involved in cytoskeletal rearrangements during cell motility and<br />

invasion. Our lab has previously shown that RhoC GTPase is over<br />

expressed in IBC and that exogenous expression of RhoC GTPase<br />

produces motile and invasive phenotype in human mammary<br />

epithelial cells (HMECs) similar to SUM149 IBC cells. Thus the<br />

overall objective of this study is to determine if caveolin-1 over<br />

expression mediates the RhoC GTPase driven invasive phenotype<br />

of IBC via its scaffolding action.<br />

The first objective of this study is to elucidate the effect of caveolin-1<br />

over expression on SUM149 IBC cell invasion. This was done by<br />

down regulating caveolin-1 to a level that is comparable to human<br />

mammary epithelial cells (HMECs) by using si caveolin-1. With<br />

the same si caveolin-1 or exogenous CSD synthetic peptide, we<br />

performed Matrigel invasion assays followed by cell viability and<br />

proliferation assay. Si caveolin-1 did not produce significant changes<br />

in cell viability over the period of four days as well as in Ki-67<br />

staining which was used as a proliferation marker. Caveolin-1 down<br />

regulation reduced cell invasion by 90% compared to untreated<br />

cells. Similarly, exogenous CSD synthetic peptide treatment reduced<br />

SUM149 cell invasion significantly. In order to understand caveolin-1<br />

localization in SUM149 cells, we performed immuno gold labeling<br />

of caveolin-1 by transmission electron microscopy as well as sucrose<br />

gradient centrifugation. Both these methods showed distinct pool of<br />

cytoplasmic caveolin-1 in addition to plasma membrane associated<br />

caveolin-1. Co-localization of caveolin-1 and RhoC GTPase was<br />

studied by co-transfecting cells with GFP-caveolin-1 and RFP-RhoC<br />

GTPase.<br />

Thus, this data suggests potential role of caveolin-1 over expression<br />

in regulating IBC cell invasion. Due to its ability to concentrate<br />

variety of signaling molecules, caveolin-1 could act as a principle<br />

regulator of several downstream events that make cells motile and<br />

www.aacrjournals.org 511s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

invasive through RhoC GTPase activation. Thus studying caveolin-1<br />

localization and effect of its regulation on the signaling pathways<br />

could help understand the important molecular mechanisms in IBC.<br />

P6-06-04<br />

Withdrawn by Author<br />

P6-06-05<br />

Down-regulation of the circadian factor Period 2 by the oncogenic<br />

E3 ligase Mdm2: Relevance of circadian components for cell<br />

cycle progression<br />

Liu J, Gotoh T, Vila-Caballer M, <strong>San</strong>tos CS, Yang J, Finkielstein CV.<br />

Virginia Tech, Blacksburg, VA<br />

Circadian rhythms are mechanisms that measure time on a scale<br />

of about 24 h and that adjusts our body to external environmental<br />

signals. Core circadian clock genes are defined as genes whose protein<br />

products are necessary components for the generation and regulation<br />

of circadian rhythms. Circadian proteins also regulate genes involved<br />

in either cell division or death; and a perturbation of the balance<br />

among these processes leads to cancer development and progression.<br />

A key aspect of cancer research is identifying new regulatory<br />

pathways involved in proliferation and differentiation of cell.<br />

Disruption of circadian rhythm has recently emerged as a new<br />

potential risk factor in the development of cancer, pointing to the<br />

core gene period 2 (per2) as a tumor suppressor. However, it remains<br />

unclear how the circadian network regulates tumor suppression, nor<br />

which, if any, of its components is either the ultimate effector that<br />

influences the fate of the cell.<br />

Initial experiments were devoted to identifying new interacting<br />

partners for Per2 using a two-hybrid system. Interestingly, among the<br />

positive clones analyzed was the oncogenic protein Mdm2. This result<br />

was validated by immunoprecipitation of recombinant and endogenous<br />

Per2/Mdm2 complexes from unstressed cells. Pull-down assays using<br />

tagged-expressed proteins fragments and labeled proteins were later<br />

used to map the interacting regions between Per2 and Mdm2. Our<br />

results show Mdm2 binds to the central flexible region of Per2 known<br />

to interact with various protein partners. Thus, we hypothesized that<br />

binding of Mdm2 to Per2 might act by mediating its ubiquitination<br />

and therefore altering Per2 stability. We next examined the formation<br />

of the Mdm2/p53/Per2 complex by immunoprecipitation. Our data<br />

show anti-p53 antibody is able to co-immunoprecipitate Per2 and<br />

Mdm2. Moreover, in vitro and in vivo ubiquitination assays show<br />

that binding of Per2 to p53 prevented ubiquitination of p53 by Mdm2<br />

without altering their binding. Immunofluorescence studies using<br />

H1299 cells (p53-) confirmed Per2 role in p53 stabilization and for<br />

localization. Overall our results suggest that Mdm2 modulates the<br />

stability of Per2 and p53 in unstressed cells, and might be responsible<br />

for the oscillatory levels of these proteins observed in a 24 h cycle.<br />

P6-06-06<br />

The therapeutic potential and hazard of exposure to Cerbera<br />

odollam leaf extracts<br />

Chung F, Chan K, Lim YY, Li B. Monash University Sunway Campus,<br />

Subang Jaya, Selangor, Malaysia; Taylor’s University, Subang Jaya,<br />

Selangor, Malaysia<br />

Background: Despite its significance in leading to the reduction of<br />

breast cancer mortality rates, therapeutic regiments for breast cancer<br />

still face two major problems: (1) the heterogeneity of the tumours and<br />

(2) the prevalence of drug resistance towards anti-estrogen therapy.<br />

The leaf extracts from Cerbera odollam were screened for active<br />

compounds with the potential to overcome these problems.<br />

Methods: Hormone responsive (MCF7) and hormone-independent<br />

cell lines (MDA-MB-231) were treated with the fractionated<br />

methanolic extracts of C. odollam followed by analyses on cell<br />

cycle perturbations and gene expression changes resulting from the<br />

treatment. The stated analyses were performed by flow cytometry,<br />

semi-quantitative RT-PCR, western blotting and immunofluorescence.<br />

The active fraction was identified, upon which HPLC separation was<br />

performed to identify the putative active compound.<br />

Results: Initial experiments revealed that a fractionated extract<br />

contains a potent cytostatic agent which is active towards hormoneresponsive<br />

but not hormone-independent breast cell lines [the<br />

Estrogen Receptor-alpha (ERα) positive MCF7 cells remained<br />

static in growth for 10 days]. Comparison of the effects of the active<br />

and inactive fractions on MCF7 cells revealed key differences in<br />

cell growth and expression of ERα and its downstream regulated<br />

genes. The active compound was identified to be neriifolin (a cardiac<br />

glycoside) which also exhibited prolonged cytostatic effects (for 10<br />

days upon treatment of 18.7 nM) similar to the active fraction. Gene<br />

expression analyses suggested that the ERα/Estrogen (E2) regulatory<br />

pathway and p53-dependent p21 WAF1 transactivaton could be the<br />

cellular targets.<br />

Conclusion: Although neriifolin could be a potential drug for breast<br />

cancer treatment, its severity in down regulation of the ERα/E2<br />

regulatory pathway which is critical during female development<br />

indicates the potential health hazard from our exposure to C. odollam.<br />

We are now focusing on establishing assay to evaluate whether<br />

neriifolin could cause drug resistance (similar to tamoxifen) by<br />

silencing ERα expression through hypermethylation of the ERα<br />

promoter.<br />

P6-07-01<br />

Withdrawn by Author<br />

P6-07-02<br />

Prediction of oncotype DX ® recurrence score using pathology<br />

generated equations<br />

Bhargava R, Klein ME, Shuai Y, Brufsky AM, Puhalla SL, Jankowitz<br />

R, Dabbs DJ. Magee-Womens Hospital of UPMC, Pittsburgh, PA;<br />

University of Wisconsin-Madison School of Medicine and Public<br />

Health, Madison, WI; University of Pittsburgh <strong>Cancer</strong> Institute,<br />

Pittsburgh, PA<br />

BACKGROUND: Oncotype DX ® is a quantitative reverse<br />

transcription polymerase chain reaction based assay that has been<br />

shown to have prognostic and predictive value in estrogen receptor<br />

(ER) positive breast cancers. The result is reported as a recurrence<br />

score (RS) ranging from 0-100, divided into low risk (


December 4-8, 2012 <strong>Abstract</strong>s: General Session 6<br />

New Magee Equation 1 (nME1): RS = 15.31385 + Nottingham<br />

score*1.4055+ ER H-score*(-0.01924) + PR H-score*(-0.02925) +<br />

(0 for HER2 negative, 0.77681 for equivocal, 11.58134 for HER2<br />

positive) + Tumor size*0.78677 + KI67 index*0.13269.<br />

New Magee Equation 2 (nME2): RS = 18.8042+ Nottingham<br />

score*2.34123 + ER H-score*(-0.03749) + PR H-score*(-0.03065)<br />

+ (0 for HER2 negative, 1.82921 for equivocal, 11.51378 for HER2<br />

positive) + Tumor size*0.04267.<br />

New Magee Equation 3 (nME3): RS = 24.30812+ ER<br />

H-score*(-0.02177) + PR H-score*(-0.02884) + (0 for HER2<br />

negative, 1.46495 for equivocal, 12.75525 for HER2 positive) +<br />

KI-67*0.18649.<br />

RESULTS: The concordance between the risk category of oncotype<br />

DX ® and our equations was 55.8%, 59.4%, and 54.4% for nME1,<br />

nME2, and nME3 respectively. With exclusion of the intermediate risk<br />

categories for both the actual RS and estimated RS, the concordance<br />

for each equation increased to more than 95%, reflecting the very<br />

low two step discordance (100% {76/76}, 98.6% {75/76}, and<br />

98.7% {79/80} for nME1, nME2, and nME3 respectively). Even<br />

when the estimated RS fell in the intermediate category with any of<br />

the equations, the actual RS was either intermediate or low in more<br />

than 85% of the cases. The Pearson correlation coefficient between<br />

estimated and actual RS was similar for each of the equations<br />

(0.61661, 0.60386 and 0.59407 for nME1, nME2 and nME3,<br />

respectively).<br />

CONCLUSIONS: Any of the 3 equations can be used to estimate the<br />

RS depending on available data. If the estimated RS is clearly high or<br />

low, the oncologists should not expect a dramatically different result<br />

from oncotype DX®, and the oncotype DX® test may not be needed.<br />

Conversely, an oncotype DX® result that is dramatically different<br />

from what is expected based on standard morpho-immunohistologic<br />

variables should be thoroughly investigated.<br />

P6-07-03<br />

Risk classification of Early Stage <strong>Breast</strong> <strong>Cancer</strong> as Assessed by<br />

MammaPrint and Oncotype DX Genomic Assays<br />

Clough KB, Poulet B, Jamshidian F, Butler S, Svedman C, Levy E.<br />

L’Institut du Sein-Paris <strong>Breast</strong> Centre, Paris, Ile de France, France;<br />

Genomic Health, Inc., Redwood City, CA<br />

Background: The 21-gene Oncotype DX® Recurrence Score® assay<br />

has been validated for prediction of 10-year risk of distant recurrence<br />

and likelihood of benefit from chemotherapy (CT) in patients (Pts)<br />

with estrogen receptor-positive (ER+) early stage breast cancer<br />

(ESBC). MammaPrint ® is a multi-gene assay that has been validated<br />

as a prognosticator in a heterogenous Pt population that includes ER+,<br />

ER-, triple negative and HER2+ Pts. While the development and<br />

validation cohorts for these genomic assays are significantly different,<br />

the tests are frequently believed to provide equivalent information.<br />

The aim of the present study was to assess how these tests classify<br />

patients when compared side by side in the same Pt specimen.<br />

Materials and methods: Pts with ER+, HER2- , ESBC in which the<br />

MammaPrint assay had been sent were identified at the Institut du<br />

Sein, Paris, France. Clinical and pathological characteristics and<br />

the MammaPrint results (failure, low or high risk) were collected.<br />

Oncotype DX quantitative RT-PCR analysis was performed at<br />

Genomic Health and was blinded to the clinical and MammaPrint<br />

data. Descriptive statistics were calculated for failure rates, cross<br />

classification, and tumor characteristics.<br />

Results: Informed consent and sufficient tumor material for analysis<br />

was available from 67 Pts who were predominantly low and<br />

intermediate risk by clinicopathologic features: Tumor grade- 24<br />

low, 36 intermediate and 7 high; Histology- 49 ductal, 14 lobular<br />

and 4 other. MammaPrint analysis had failed to deliver result for 10<br />

of the 67 Pts. Recurrence Score results were generated in all 67 Pts.<br />

The risk classification of the MammaPrint and Oncotype DX tests in<br />

the 57 patients where both results were available is presented in the<br />

Table below. Of note, there were only 2 high Recurrence Score results<br />

compared with 22 high risk MammaPrint results. Furthermore, 45%<br />

of the Pts with high risk MammaPrint result had a low Recurrence<br />

Score result; they also had high quantitative ER expression of more<br />

than 9.5 expression units by RT-PCR, which is associated with likely<br />

hormonal therapy benefit. There was no clear association between<br />

tumor characteristics and MammaPrint failure or differences in risk<br />

classification.Conclusions: This direct comparison demonstrates that<br />

the MammaPrint and Oncotype DX tests classify a large proportion<br />

of Pts differently. Of note, nearly half of the Pts with high risk<br />

MammaPrint result had a low Recurrence Score indicating minimal,<br />

if any, benefit from chemotherapy.<br />

Table 1. Mammaprint and Recurrence Score classification<br />

Mammaprint<br />

Low Risk High Risk Total<br />

Low Recurrence Score 23 (65%) 10 (45%) 33 (58%)<br />

Intermediate Recurrence Score 11 (31%) 11 (50%) 22 (39%)<br />

High Recurrence Score 1 (3%) 1 (5%) 2 (4%)<br />

Total 35 (100%) 22 (100%) 57 (100%)<br />

P6-07-04<br />

Evaluation of Adjuvant! Online for primary operable grade 2<br />

breast cancers with 10-year follow-up<br />

Van Calster B, Brouckaert O, Wildiers H, Christiaens M-R,<br />

Weltens C, Paridaens R, Van Limbergen E, Neven P. On behalf of<br />

Multidisciplinary <strong>Breast</strong> Centre, UZ Leuven<br />

Background: In primary operable breast cancers, Adjuvant!Online<br />

(AOL) uses demographics and clinic-pathological markers to<br />

predict the 10-year breast cancer specific survival (BCSS) with and<br />

without adjuvant systemic therapy. Grade 2 BC is a difficult group<br />

for adjuvant treatment decisions. We compared the observed with<br />

the AOL-estimated BCSS rate in our centre in consecutive women<br />

primary operated for a grade 2 BC.<br />

Patients and Methods: All patients with grade 2 primary operable<br />

breast cancer from UZL diagnosed between 1/1/2000 and 31/3/2002.<br />

After 10 years of follow-up, patients were evaluated for BCSS<br />

and dead from other cause. For each patient, the adjuvant! Online<br />

algorithm (version 8) was applied to predict BCSS taking the patients’<br />

adjuvant therapy into account. Adjuvant! Online was validated by<br />

assessing the calibration and discrimination. Discrimination was<br />

assessed by the area under the ROC curve. The expected BCSS rate<br />

was obtained by averaging the predicted probabilities of BCSS. Next,<br />

logistic calibration was performed by predicting observed BCSS<br />

within 10 years of follow-up with the odds of the predicted probability<br />

of BCSS (odds BCSS). In this analysis, the intercept should be 0 and<br />

the coefficient for odds BCSS 1 for perfect calibration of the predicted<br />

probabilities. A likelihood ratio test is used to test the null hypothesis<br />

of perfect calibration.<br />

Outcome after 10 years follow-up N (%)<br />

Alive 358 (80.5%)<br />

Death due to BC 29 (6.5%)<br />

Death due to other cause 58 (13.0%)<br />

Lost to follow-up 6<br />

Percentages exclude the cases that were lost to follow-up.<br />

Results: Six patients were lost to follow-up. The median age at<br />

diagnosis of the remaining 445 patients was 56 years (IQR 48 to 67<br />

years, range 26 to 89 years). In 433/445 cases (97%), the prediction<br />

from AOL was available. The median AOL predicted probability for<br />

BCSS was 89.1% (IQR 83.2 to 94.6%, range 20.3 to 98.9%). The<br />

observed BCSS rate was 93.3% whereas the AOL-expected rate was<br />

www.aacrjournals.org 513s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

86.7%. The likelihood ratio test for perfect calibration was statistically<br />

significant (p


December 4-8, 2012 <strong>Abstract</strong>s: General Session 6<br />

Methods: 162 pts were enrolled in IBCSG 27-02, BIG 1-02, NSABP<br />

B-37. RT was recommended but not required if a completion/<br />

salvage mastectomy was performed after an ipsilateral breast tumor<br />

recurrence. Hormonal therapy was required for ER+ and/or PR+<br />

tumors and anti-HER2 therapy was given at investigator discretion.<br />

Stratification criteria were hormone receptor status of recurrence, site<br />

of recurrence (breast, mastectomy scar/chest, regional lymph nodes),<br />

and use of prior C.<br />

Results: Primary surgery was breast conserving: 98 (60%) and<br />

mastectomy: 64 (40%). Nodal surgery for primary cancer was sentinel<br />

node resection: 19 (12%), axillary node (AN) sampling: 37 (23%), AN<br />

dissection: 88 (56%), more extensive nodal surgery: 6 (4%), and no<br />

nodal surgery: 8 (5%). The sites of first ILRR were breast 87 (54%),<br />

mastectomy scar/chest wall 55 (34%), and regional lymph nodes 20<br />

(12%), and were similarly distributed by treatment arms. Of the 98<br />

pts who originally had a lumpectomy, 92 (94%) received RT. Among<br />

these 92, ILRR were in-breast: 82 (89%), chest wall: 3 (3%), and<br />

regional node: 7 (8%). Salvage mastectomy was performed in 76/98<br />

(78%) with prior lumpectomy. Of the other 22 pts, 16 had repeat<br />

lumpectomies, 5 had AN resection, and 1 had chest wall resection. Of<br />

the 64 who originally had mastectomy, 52 (81%) recurred in the scar/<br />

chest wall and 12 (19%) in the regional nodes. 15 pts (23%) received<br />

post-mastectomy RT, 4 of whom had regional nodal recurrences. Of<br />

the 139 pts who had ER status reported for the primary tumor, 18<br />

(13%) had a different ER status reported for the ILRR (table). PR<br />

expression was discordant in 29 (25%).<br />

Estrogen Receptor Status<br />

ILLR<br />

Primary Negative Positive Total<br />

Negative 42(89%) 5(11%) 47<br />

Positive 13(14%) 79(86%) 92<br />

Tot. known 55 84 139<br />

Missing 3 20 23<br />

Average time from surgery for ILRR to surgery for second LRR was<br />

1.6 yrs (range: 0.08 -5.8). The average pt age for second LRR was<br />

60.4 yrs versus 54.5 yrs for those who developed distant failures. At<br />

median follow up of 4.5 yrs, 15 pts developed a second LRR (9 local,<br />

6 regional), 7 (47%) of whom have died. Of the 32 (20%) pts who<br />

developed a distant failure, 17 (53%) have died. C following ILRR<br />

appears to reduce second LRR (5 v 10).<br />

Conclusions Similar to the effect observed in distant failure rates,<br />

C reduced the rate of second LRR by half. A second LRR following<br />

multimodality therapy for an ILRR is a strong prognostic indicator of<br />

subsequent death in this population, comparable to pts experiencing<br />

distant metastasis.<br />

Funding: NCI PHS grants U10-CA-37377, -69974, -12027, -69651,<br />

and CA-75362.<br />

P6-07-07<br />

Withdrawn by Author<br />

P6-07-08<br />

Associations between lifestyle parameters and prognostic factors<br />

used for stratifying for adjuvant treatment in breast cancer<br />

Guldberg TL, Christensen S, Ravnsbaek A, Zachariae B, Jensen AB.<br />

Aarhus University Hospital, Aarhus, Denmark<br />

Background: At present, tumour related factors and age at time of<br />

diagnosis are used to determine type of adjuvant treatment for women<br />

diagnosed with early stage breast cancer (ESBC).<br />

Aim: To investigate associations between lifestyle parameters and<br />

prognostic factors used for stratifying for adjuvant treatment, by<br />

correlating known clinical prognostic factors to comorbidity, body<br />

mass index (BMI), and physical activity.<br />

Method: 4917 women who had been treated for ESBC in Denmark in<br />

2001-2004 were identified. Disease-and treatment data were obtained<br />

from The Danish <strong>Breast</strong> <strong>Cancer</strong> Cooperative Group. Data concerning<br />

comorbidity was collected at the surgical departments. Health related<br />

behaviours were assessed by questionnaires three months after<br />

surgery, using the Physical Activity Scale for the Elderly to assess<br />

level of physical activity.<br />

Results: All lifestyle parameters were significantly associated with<br />

one or more tumour related prognostic factors at time of surgery:<br />

Increasing BMI was associated with unfavourable nodal status (OR:<br />

1.026; 95%CI:1.009-1.043), with not having grade I malignancy<br />

(OR:1.028; 95%CI:1.008-1.048) and with tumour size >20mm<br />

(OR:1.083; 95%CI: 1.065-1.102) but not with oestrogen receptor (ER)<br />

status (OR:1.002; 95%CI:0.981-1.023). Similarly, decreasing levels of<br />

physical activity was associated with nodal status (OR: 1.001; 95%CI:<br />

1.001-1.003), with tumour size (OR: 1.002; 95%CI: 1.001-1.003) and<br />

with higher grade of malignancy (OR: 1.001; 95%CI: 1.000-1.003;<br />

p=0.014), but not with ER status (OR: 1.000; 95%CI:0.999-1.002).<br />

Comorbidity was significantly associated with tumour size>20mm<br />

(OR: 1.137; 95%CI: 1.002-1.275), but not with nodal status (OR:<br />

0.989; 95%CI: 0.878-1.115), higher grade of malignancy (OR: 0.918;<br />

95%CI: 0.803-1.050) or ER status (OR: 1.176; 95%CI: 0.992-1.393).<br />

Some of the significant associations between lifestyle parameters and<br />

tumour related prognostic factors only pertained to premenopausal<br />

women. This pattern was observed in the associations between<br />

BMI and nodal status (Premenopausal: χ2: 14.55; p=0.002.<br />

Postmenopausal: χ2:2.39; p=0.495), and physical activity and tumour<br />

size (Premenopausal: χ2:13.72; p=0.008. Postmenopausal: χ2: 7.10;<br />

p=0.130). No such differences were found for comorbidity.<br />

Conclusion: Higher levels of BMI and comorbidity were found to<br />

be associated with poorer prognostic factors at time of surgery in<br />

women suffering from ESBC. Furthermore, inactivity and higher<br />

BMI was associated with significantly poorer prognostic factors in<br />

the premenopausal population than it was in the postmenopausal<br />

population. Physically inactive women had poorer prognostic factors<br />

at time of surgery; however, caution is needed due to possible<br />

confounding by treatment parameters. Future data on the relapsepattern<br />

in this cohort will shed further light on whether specific<br />

lifestyle patterns can predict disease outcome and the potential<br />

relevance of stratifying women suffering from ESBC to relevant<br />

adjuvant treatment according to lifestyle parameters at time of<br />

diagnosis, especially in the younger premenopausal population.<br />

P6-07-09<br />

Identification of Trophinin associated protein (TROAP) as a<br />

novel biological marker in breast cancer (BC): Co-expression of<br />

TROAP and TOPO2A predicts response of anthracycline based<br />

chemotherapy (ATC-CT)<br />

Abdel-Fatah TMA, Balls G, Miles AK, Moseley P, Green A, Rees<br />

R, Ellis IO, Chan SYT. Nottinigah City Hospital NHS Trust; The<br />

Van Geest <strong>Cancer</strong> Research Center, Nottingham Trent University;<br />

University of Nottingham<br />

Introduction<br />

Recently, TOPO2A alteration was found to be a predictor for ATC-CT<br />

and by using neural network and pathways analysis of gene expression<br />

array data , TROAP gene was revealed as a major hub in TOPO2A<br />

pathway and strongly related to genes that are involved in mitotic<br />

cell cycle regulation. In addition, we found that TROAP gene was<br />

among top 10 ranked genes out of 48,000 of transcripts, that accurately<br />

predicted worse clinical outcome, and differentiated between low and<br />

high grade based on a 10-fold external cross-validation analysis with<br />

www.aacrjournals.org 515s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

an average classification accuracy of >99.999%. TROAP protein is<br />

essential for centrosome integrity and proper bipolar organisation<br />

of spindle assembly during mitosis and plays essential role in cell<br />

proliferation.<br />

In the current study the molecular and clinicopathological functions<br />

of TROAP expression and its effect on management of breast cancer<br />

have been investigated.<br />

Methods<br />

The co-expression of TROAP and TOPO2A protein was evaluated by<br />

using dual immunoflurescent in BC cell lines. In addition both TROAP<br />

and TOP2A protein expressions were immunohistochemically (IHC)<br />

assessed in 40 normal breast tissues and a well characterised series<br />

of 1650 primary BC and were correlated to clinicopathological and<br />

other biomarkers. IHC staining was performed using Anti-TROAP<br />

rabbit polyclonal (HPA044102; Sigma).<br />

The association between TROAP and response to chemotherapy was<br />

investigated in 350 ER negative BC treated with adjuvant ATC-CT<br />

and 260 locally advanced BC treated with neoadjuvant ATC-CT. In<br />

addition the clinical outcome of TROAP expression was evaluated in a<br />

series of 180 ER- high risk BC patients who did not received any CT.<br />

Results<br />

No expression of TROAP protein was observed in normal breast tissue<br />

while, 25% of BC showed TROAP protein overexpression. By using<br />

dual immunoflurescent in BC cell lines, The MCF7 cells showed<br />

strong cytoplasmic TROAP staining with no TOPO2A expression,<br />

while The T47D cells did not express TROAP but expressed<br />

TOPO2A. SKBr3, MDA468 and MDA231 cell lines showed coexpression<br />

of TROAP and TOPO2A. TROAP overexpression was<br />

significantly associated with aggressive clinico-pathological features<br />

including; high grade, high mitotic rate, absence of hormonal<br />

receptors, overexpression of HER2, TOP2A and EGFR (p


December 4-8, 2012 <strong>Abstract</strong>s: General Session 6<br />

invasive ductal carcinoma (IDC). The prognostic factors for IMPC<br />

are not well characterized due to the relative scarcity of cases reported<br />

in the literature.<br />

Methods: We analyzed the National <strong>Cancer</strong> Institute’s Surveillance,<br />

Epidemiology, and End Results (SEER) database to evaluate<br />

prognostic factors of a population of 645 breast IMPC patients<br />

and 300,060 breast IDC patients reported between 2001 and 2008.<br />

Using univariate and multivariate analyses, hazard ratios (HR) were<br />

calculated for disease-specific (DSS) and overall survival (OS) for<br />

these patients using parameters such as patient age at diagnosis,<br />

histological grade, ER status, PR status, tumor size, and degree of<br />

lymph node positivity. Subset analysis of high grade, lymph nodepositive<br />

patients was performed to compare DSS and OS between<br />

IMPC and IDC.<br />

Results: The 5-year DSS and OS for IMPC patients were 92.1% and<br />

84.6% compared to 5-year DSS and OS of 88.5% and 80.2% for IDC<br />

patients. At presentation, TNM staging of IMPC cases was similar<br />

to IDC except for a higher percentage of LN metastases (52.4% in<br />

IMPC vs. 34.7% in IDC). Of those with known estrogen receptor<br />

(ER) status, 84.2% of IMPC cases were ER-positive, which was<br />

associated with better DSS (Hazard Ratio (HR) 0.36, p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

series with local recurrence, Grade 3 , >3 nodes involved, tumour<br />

diameter >40mm, coded dichotomously, and age coded as above as<br />

independent variables.<br />

Results<br />

The results of the analyses are given in the table below.<br />

Regression analyses (case matched subset and whole series)<br />

Variable Hazard ratio 95% CI P value<br />

Matched cases (n = 340)<br />

Local recurrence 1.63 1.17 - 2.28 0.0041<br />

Age deviation 1.03 per year 1.01 - 1.05 0.015<br />

Whole series (n = 2945)<br />

Local recurrence 1.33 1.01 - 1.75 0.0410<br />

Age deviation 1.05 per year 1.04 - 1.06 40mm 1.66 1.31 - 2.13 3 +ve nodes 2.99 2.46 - 3.66


December 4-8, 2012 <strong>Abstract</strong>s: General Session 6<br />

by analysing the differences in the published taxane effect in the<br />

EBCTCG Chemotherapy overview (4) according to tumour grade.<br />

Methods : Annual event rate ratios and 95% CIs were calculated<br />

from data on over 20,000 early breast cancer patients treated with<br />

chemotherapy (4). P-values for trend in taxane effect were calculated<br />

across 3 tumour grade categories or for heterogeneity between 2<br />

categories.<br />

Results : The table shows annual event rate ratios (CIs) for grade<br />

categories, for taxane-plus-anthracycline-based regimen versus<br />

the same, or more (< doubled or ∼doubled) non-taxane cytotoxic<br />

chemotherapy. Significantly different taxane effects were observed<br />

across categories of tumour grade. For both endpoints taxane benefit<br />

increased with lower tumour grade, there was a smaller effect in high<br />

grade tumours and no evidence the treatment benefit decreased with<br />

follow up time.<br />

Taxane Effect by Differentiation status<br />

Event rate ratio<br />

Grade<br />

(95%CI)<br />

N Grade 3 Grade 2 Grade 1/2 Grade 1 P-value<br />

BRCa Mortality 20,770 1.08(.94-1.25) 0.77(.65-0.90) 0.68(.47-0.98)


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

predictive biomarkers to detect these non-responsive breast cancers at<br />

time of diagnosis may allow for improved clinical outcome for these<br />

patients. FKBP4 (FKBP52) is a co-chaperone protein that has been<br />

shown to regulate progesterone but not estrogen receptor activity, and<br />

was recently found to be highly expressed in early stage breast cancer<br />

compared to benign breast tissue.This evidence suggests that FKBP4<br />

may play an important role in endocrine-responsive breast cancers.<br />

Methods: Global proteomic screening for biomarkers of aggressive<br />

breast cancer was performed on tissue samples from 24 patients with<br />

lymph node negative (LN-) disease and 24 patients with lymph node<br />

positive (LN+) disease randomly selected from the Calgary Tamoxifen<br />

Cohort, a retrospective cohort of breast cancer patients treated with<br />

adjuvant tamoxifen [n=511] from 1985-2000 at the Tom Baker <strong>Cancer</strong><br />

Centre (Calgary, Canada). Liquid tissue lysates from FFPE tissue were<br />

analyzed by mass spectrometry and differentially expressed proteins<br />

were identified by the semi-quantitative spectral count method.<br />

FKBP4 was identified as an upregulated target in LN+ patients. The<br />

mRNA expression database of Kao et al. (BMC <strong>Cancer</strong>, 2011) was<br />

used to assess the prognostic potential of targets identified in the<br />

proteomic screen. FKBP4 protein expression was evaluated in tissue<br />

microarrays built from FFPE samples from the Calgary Tamoxifen<br />

Cohort using quantitative fluorescence immunohistochemistry and<br />

HistoRx AQUA analysis. Ten-year overall survival (OS) or five-year<br />

disease free survival (DFS) were the primary outcomes. Continuous<br />

variable FKBP4 data was dichotomized at the top quartile for both<br />

mRNA and protein expression analysis.<br />

Results: Univariate analysis demonstrated a significant association<br />

between high levels of FKBP4 mRNA and worse OS in ER+Her2-<br />

patients [n=182, HR=2.118 (1.070-4.192), p=0.031] within the Kao<br />

et al database. Similarly, univariate analysis demonstrated that high<br />

levels of FKBP4 protein expression was associated with significantly<br />

worse DFS in ER+ Her2- patients [n=358, HR=1.632 (1.001-2.659),<br />

p=0.049] in the Calgary Tamoxifen Cohort. FKBP4 was found to be an<br />

independent prognostic factor in both mRNA and protein expression<br />

cohorts using multivariate analysis adjusted for age, T stage, and<br />

lymph node status [OS: HR=2.786 (1.394-5.570), p=0.004], or age,<br />

tumor size, tumor grade, and lymph node status [DFS: HR=1.875<br />

(1.021-3.442), p=0.043], respectively.<br />

Conclusions: Proteomic screening from FFPE tissue can identify new<br />

breast cancer biomarker candidates. Using this technique we have<br />

identified FKBP4 as an independent prognostic factor in ER+Her2-<br />

breast cancers, measured either by mRNA expression analysis or by<br />

quantitative protein expression analysis. Further studies are required<br />

to determine if FKBP4 may also be a predictive biomarker for<br />

tamoxifen response in ER+HER2- patients.<br />

P6-07-18<br />

Identification of Sperm Associated Antigen 5 (SPAG5) as a novel<br />

biological and predictive biomarker in <strong>Breast</strong> cancer<br />

Abdel-Fatah TMA, Ball G, Miles AK, Moseley P, Green A, Rees B,<br />

Ellis IO, Chan SYT. Nottingham City Hospital NHS Trust, Nottingham;<br />

The John van Geest <strong>Cancer</strong> Research Centre, Nottingham Trent<br />

University; University of Nottingham<br />

Introduction<br />

SPAG5 has been found to be involved in the functional and dynamic<br />

regulation of mitotic spindles, and to be essential for chromosome<br />

segregation fidelity. Recently we found by using neural network<br />

and pathways analysis of a gene expression array data that SPAG5<br />

was among top 10 ranked genes out of 48,000 of transcripts, that<br />

accurately predicted worse clinical outcome based on a 10-fold<br />

external cross-validation analysis with an average classification<br />

accuracy of >99.999%. Moreover we found that 5% of BC showed<br />

amplification of SPAG5 locus at chromosome 17q11.2 and SPAG5<br />

mRNA expression levels displayed a statistically significant<br />

correlation with its copy number.<br />

Methods<br />

In the current study the molecular and clinicopathological features<br />

of SPAG5 expression and its effect on management of BC have been<br />

investigated in 2800 BC patients with primary operable invasive BCs<br />

constituted four cohorts:<br />

1) A series of 1650 BC patients received adjuvant endocrine and/or<br />

CMF chemotherapy according to NPI.<br />

2) A series of 256 BC received adjuvant anthracycline-based<br />

chemotherapy (ATC-CT)<br />

3) A series of 140 primary BC HER2+ patients treated with ATC-CT+<br />

Herceptin 4) To validate SPAG5 as a predictor factor for ATC-CT,<br />

260 patients with locally advanced primary breast cancer treated with<br />

neoadjuvant ATC-CT were included and the pathological complete<br />

response (pCR) was used to evaluate the response to chemotherapy.<br />

Immunohistochemical staining was performed using Anti--SPAG5<br />

rabbit polyclonal (HPA022479; Sigma).<br />

Results: i) By using dual immunoflurescent in BC cell lines, coexpression<br />

of SPAG5 and proliferating cell nuclear antigen (PCNA)<br />

was detected in 4 out of 5 of the breast cancer cell lines screened<br />

(MCF7, T47D, MDA468 and MDA231) providing evidence for the<br />

importance of SPAG5 in cell proliferation. ii) 20% of breast cancer<br />

showed SPAG5 protein overexpression. SPAG5 overexpression<br />

showed a statistically significant association with ER-, PR-, triple<br />

negative phenotype, high grade tumour, high ki67, basal like<br />

phenotype and epithelial mesenchymal transition phenotype, p53<br />

mutation and absence of DNA repair genes (BRCA1, ATM and<br />

XRCC1); p values


December 4-8, 2012 <strong>Abstract</strong>s: General Session 6<br />

Patients and methods<br />

Primary operable breast cancer patients diagnosed between 1/1/2000<br />

and 31/12/2009 treated in UZ Leuven were retrieved from the<br />

prospectively managed database (n=4318). Her-2 status was missing<br />

in 93 and ER/PR in 2 patients. Of the remaining patients, 3330 were<br />

luminal (ER and/or PR positive) HER2 negative (full cohort details:<br />

Brouckaert O., et al. Ann Oncol. 2012 Apr 6). Missing values were<br />

observed in 137 patients (4.1%), and these were imputed using<br />

‘multivariate imputation by chained equations’. A competing risks<br />

proportional hazards model was used for multivariable analysis<br />

(includes age, size, grade, screening, palpability, nodal status,<br />

histology, type of surgery, radiotherapy, endocrine and chemotherapy)<br />

of distant metastasis free interval (DMFI) and breast cancer specific<br />

survival (BCSS). We used a predefined set of predictors, investigated<br />

non-linearity of the predictor effects, checked the proportional hazards<br />

assumption, and tested for interactions with grade.<br />

Results<br />

Median follow-up was 76 months and 5-year survival probability<br />

93%. PR is an independent prognostic variable but this is grade<br />

dependent for DMFI, but not for BCSS (interaction p=0.15), although<br />

PR positivity was mainly protective for grade 3 breast cancers again<br />

(table 1). Palpability is a strong prognostic variable for DMFI and<br />

BCSS and was strongly correlated with screen detection (yes/no) and<br />

here, we found no evidence of grade dependency.<br />

Table 1: descriptive statistics and multivariable adjusted hazard ratios (within a competing risk<br />

framework) for distant metastatic free interval (DMFI) and breast cancer specific survival (BCSS)<br />

N (%) DMFI BCSS<br />

Age (+10y) 58 (median) 0.87 (0.77-0.98) 0.85 (0.73-0.99)<br />

Tumor size (+1cm) 20mm (median) 1.06 (1.01-1.12) 1.01 (0.94-1.09)<br />

Palpable 2603 (78.2%) 2.07 (1.13-3.79) 2.69 (1.16-6.22)<br />

pN0 2041 (61.3%) Reference Reference<br />

pN1 944 (28.3%) 1.59 (1.11-2.27) 1.96 (1.25-3.06)<br />

pN2 228 (6.8%) 3.16 (1.97-5.05) 4.12 (2.26-7.50)<br />

pN3 117 (3.5%) 8.12 (4.94-13.4) 10.79 (5.88-19.8)<br />

Grade (+1 unit) - 5.78 (2.74-12.2) 3.22 (2.39-4.36)<br />

PR (in grade 1) - 4.03 (1.01-16.12)* -<br />

PR (in grade 2) - 1.49 (0.76-2.91) -<br />

PR (in grade 3) - 0.55 (0.35-0.85) -<br />

PR (overall) - - 0.57 (0.38-0.87)<br />

Chemotherapy 985 (29.6%) 0.60 (0.41-0.89) 0.48 (0.29-0.80)<br />

* Although marginally significant, this result is clinically unreliable due to low numbers (10<br />

metastatic events in 568 PR+ grade 1 breast cancers, but 0 events in only 56 PR- grade 1 breast<br />

cancers.<br />

Conclusion<br />

PR status and palpability may further refine prognosis of patients with<br />

luminal HER2 negative breast cancers. For PR, prognostic value is<br />

grade dependent, but for palpability, this is grade independent.<br />

P6-07-20<br />

Variables measured at Central Nervous System (CNS) relapse, but<br />

not Immunophenotype, identify groups of breast cancer patients<br />

with shorter post CNS-relapse survival<br />

Gómez HL, Pinto JA, Schwarz JL, Vigil CE, Vallejos CS. Instituto<br />

Nacional de Enfermedades Neoplásicas, Lima, Peru; Oncosalud,<br />

Lima, Peru<br />

Objective: We evaluated breast cancer immunophenotype and<br />

variables measured at CNS-relapse as prognostic factors in a cohort<br />

of Hispanic patients.<br />

Methods: We reviewed data from 2602 breast cancer women in<br />

stages I-III diagnosed between 2000 and 2005 at the Instituto<br />

Nacional de Enfermedades Neoplásicas, Lima, Perú. According to<br />

immunophenotype, tumors were categorized in luminal A (RE+ and/or<br />

RP+, HER2-), luminal B (RE+ and/or RP+, HER2+), HER2 (RE-, RP-,<br />

HER2+) and triple negative (RE-, RP-, HER2-). Clinicopathological<br />

data at diagnosis and at CNS relapse were evaluated and used to<br />

calculate prognostic scores according to Recursive Partitioning<br />

Analysis (RPA) score, Graded Prognostic Assessment (GPA) Score,<br />

and Basic Score for Brain Metastases. Endpoints were time to CNS<br />

metastasis and Post CNS-relapse survival.<br />

Results: With a median follow-up of 7.5 years, 818 (31.4%) patients<br />

had locoregional or distant recurrence. In total, 159 (6.1%) patients<br />

developed CNS metastases, of whom 92 (3.5%) as the first site of<br />

recurrence and 22 meningeal metastases were detected. In triple<br />

negative, 51 (32.1%) developed SNC metastases, 46 (28.9%) in<br />

luminal A, 37 (23.3%) in HER2 and 25 (15.7%) in luminal B patients.<br />

Median time since breast cancer diagnosis to SNC metastases was<br />

28.1 months (mo) and the variables influencing it were Clinical T<br />

(P


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Results: A total of 157 cases had CNS relapse from which 124 had<br />

only brain metastases, 19 only leptomeningeal carcinomatosis (LMC),<br />

and 14 both. Forty three cases were [HR+, HER2-] (5 LMC), 68<br />

were TN (13 LMC) and 51 were [HR+/-, HER2 +] (15 LMC). 152<br />

cases were in clinical stages I-III at diagnosis (96.8% of all SNC<br />

relapses). There was no association between IHC phenotypes and age<br />

at CNS relapse, ECOG Performance Status (P=0.837), extracraneal<br />

metastases (P=0.304), control of primary tumor (P=0.164), number<br />

or volume of brain metastases (P=0.706). Significant association was<br />

found between phenotype and time from diagnosis to development of<br />

CNS metastases (≤8 months: 16.3% in TN, 7.3% in Her-2 and 0% for<br />

[HR+, HER2-]; P=0.017 ) and histological grade (grade III: 74.4% in<br />

TN, 55.% in Her-2 and 30% for [HR+, HER2-]; P=0.006). Symptoms/<br />

signs commonest in LMC patients were cephalea (7 cases, 72%),<br />

meningism (6, 24%), nausea (6, 24%), vomiting (5, 20%), ataxia (5,<br />

20%), facial palsy (4, 16%), somnolence (4, 16%), paraparesia (3,<br />

12%), hiporeflexia (3, 12%), apraxia (3, 12%), arreflexia (3, 12%),<br />

poor sphincter control (3, 12%), hemiparesia (2, 8%), seizures (2,<br />

8%), bradypsichia (2, 8%), neuropathic pain (1, 4%). Meningism<br />

was most frequent in TN cases (50%). Post recurrence survival was<br />

shorter in LMC TN patients (3.61mo vs 4.89mo to [HR+, HER2-]<br />

vs 5.95mo to [HR+/-, HER2 +; P=0.044] and in LMC patients with<br />

meningism (1.38mo vs 5.26mo; P=0.02). Multivariate analysis<br />

identify meningism as the worse risk factor for survival in patients<br />

relapsed with LMC (HR: 4.33 compared with patients without LMC;<br />

P=0.066). Conclusions: In LMC patients, meningism was identified<br />

as a sign related to shorter survival after the SNC relapse.<br />

P6-07-22<br />

Association between SPARC mRNA expression, prognosis and<br />

response to neoadjuvant chemotherapy in early breast cancer<br />

(BC): a pooled in-silico analysis<br />

Azim Jr HA, Singhal S, Michiels S, Ignatiadis M, Djazouli K,<br />

Fumagalli D, Desmedt C, Piccart M, Sotiriou C. Universite Libre de<br />

Bruxelles, Brussels, Belgium; Institut Jules Bordet, Brussels, Belgium;<br />

Celgene International<br />

Background: SPARC (Secreted Protein Acidic and Rich in Cysteine)<br />

is known to regulate cell growth and to inhibit cell-cycle progression.<br />

It has high affinity in binding albumin and hence has been suggested<br />

to predict benefit of albumin-bound cytotoxic agents. We conducted<br />

a pooled analysis to elucidate SPARC expression according to BC<br />

molecular subtypes and its association with clinical outcome in<br />

early BC.<br />

Methods: We used publically available datasets and normalized<br />

microarray data as published by the original studies. Eligible patients<br />

were those who received no adjuvant systemic therapy (untreated<br />

series), were treated with tamoxifen (tam-treated series) or were<br />

treated with neoadjuvant anthracyclines ± paclitaxel or docetaxel<br />

(neoadjuvant series). We computed 2 SPARC modules, SPARC7 and<br />

SPARC8 that were composed of genes with an absolute correlation<br />

above 0.7 and 0.8 with SPARC, respectively. In the untreated series,<br />

we examined the expression of SPARC according to BC subtype<br />

defined by PAM50. We investigated the correlation with other gene<br />

modules representing diverse biological processes; proliferation<br />

(AURKA, GGI), immune (STAT1, IRM), and stroma (DCN, PLAU).<br />

We investigated the association with relapse-free survival (RFS) in<br />

univariate and multivariate models, both in the untreated and tamtreated<br />

series. This was performed in all patients and according to BC<br />

subtype. In the neoadjuvant series, we investigated the association<br />

with pathological complete response (pCR) in all patients and<br />

according to BC subtype. All multivariate models were adjusted for<br />

tumor size, nodal status, age and histological grade.<br />

Results: 1008, 393 and 996 patients were included in the untreated,<br />

tam-treated and neoadjuvant series, respectively. SPARC expression<br />

was highest in luminal-A, small (< 2 cm) and low histological grade<br />

tumors (all p


December 4-8, 2012 <strong>Abstract</strong>s: General Session 6<br />

For all cut off points, there was no significant difference in survival<br />

rate and recurrence rate. We observed high-rates of lymphatic invasion<br />

and lymph node metastasis even in patients with a low proportion<br />

of IMPC lesion.<br />

Conclusion<br />

Our results provide no evidence to support a relationship between<br />

proportion of IMPC lesion and breast cancer prognosis. However,<br />

lymphatic invasion and lymph node metastasis was a high frequency<br />

with low proportion of the IMPC.<br />

The number of cases, lymphatic invasion, LN positive, recurrence rate and overall survival<br />

# of cases


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Conclusion<br />

In this large dataset with long follow up our results indicate that<br />

high annexin A1 expression among women with breast cancer was<br />

associated with favorable patient and tumor characteristics and an<br />

improved RFS. Annexin A1 should be explored further as a potential<br />

prognostic biomarker.<br />

P6-07-26<br />

Prognostic significance of the maximal value of the baseline<br />

standardized uptake value on fluorine 18 fluorodeoxyglucose<br />

positron emission tomography /computed tomography for<br />

predicting pathologic malignancy of operable breast cancer with<br />

neoadjuvant chemotherapy<br />

Kadoya T, Akimoto E, Emi A, Shigematsu H, Masumoto N, Okada<br />

M. Hiroshima University, Hiroshima, Japan<br />

PURPOSE: [18F]-fluorodeoxyglucose (FDG) positron emission<br />

tomography (PET)/computed tomography (CT) is potentially useful<br />

in predicting pathological complete response (pCR), disease free<br />

survival (DFS) and overall survival (OS) of breast cancer patients<br />

with neoadjuvant chemotherapy.<br />

MATERIALS AND METHODS: 77 breast cancer patients (mean<br />

age ± SD: 52.6 ± 11.2 years) with clinical Stage I∼III between<br />

January 2006 and December 2011, were prospectively evaluated<br />

(median follow up period:26.5 months). Neoadjuvant chemotherapy<br />

of an anthracycline-based regimen and taxane was performed, and<br />

patients underwent a whole-body FDG PET/CT before and after<br />

chemotherapy. The maximal value of the baseline standardized<br />

uptake values (SUVmax) were assessed for predicting pCR, DFS and<br />

OS. For the evaluation of relationship between SUVmax values and<br />

prognosticators such as hormone receptors, human epidermal growth<br />

factor receptor 2 (HER2), nuclear grade, lymph node metastasis and<br />

tumor size, statistical analyses were performed using Student t test<br />

and log-rank test, and p values of less than 0.05 were considered to<br />

indicate statistically significant differences.<br />

RESULTS: Clinical Stage included were I (n = 2, 2.6%), II (n=62,<br />

80.5%) and III (n=12, 15.6%). Tumors with estrogen receptor positive<br />

were 52 (67.5%) and negative were 24 (31.2%). Therapeutic response<br />

by neoadjuvant chemotherapy was obtained in 15 patients (19.5%)<br />

with pCR and 60 (77.9%) without pCR. Patients were divided into<br />

two groups according to cut-off SUVmax established on the basis<br />

of receiver operating characteristic (ROC) analysis (6.0 Odds ratio(95%Cl) P<br />

(n=46) (n=31) (Student t test)<br />

ER negative 9 15 3.75 0.02<br />

ER positive 36 16 (1.36-10.35)<br />

PgR negative 13 15 2.31 0.083<br />

PgR positive 32 16 (0.89-6.00)<br />

HER2 negative 31 18 1.51 0.70<br />

HER2 positive 8 7 (0.47-4.85)<br />

There was a significant difference in OS between two groups (p=0.05),<br />

but, pCR (OR:1.07, 95%Cl:0.34-3.40, P=0.86) and DFS (P=0.07) did<br />

not show strong relationship with SUVmax values.<br />

Comparison of prognosis<br />

SUV max≤6.0 SUV max>6.0 Odds ratio P<br />

(n=46) (n=31) (95%Cl)<br />

pCR 9 6 1.07 0.86<br />

Non pCR 35 25 (0.34-3.40) (Student t test)<br />

Disease free 44 27 0.07<br />

Recurrence 1 4 (log-rank test)<br />

Alive 45 28 0.02<br />

Dead 0 3 (log-rank test)<br />

CONCLUSION: SUVmax on FDG PET/CT before neoadjuvant<br />

chemotherapy have a predictive value for high-grade malignancy<br />

and prognosis in clinical Stage I∼III breast cancer.<br />

P6-07-27<br />

Body mass index and survival after breast cancer diagnosis in<br />

Japanese women<br />

Kawai M, Minami Y, Nishino Y, Ohuchi N, Kakugawa Y. Tohoku<br />

University Graduate School of Medicine, Sendai, Miyagi, Japan;<br />

Miyagi <strong>Cancer</strong> Center Research Institute, Natori, Miyagi, Japan;<br />

Miyagi <strong>Cancer</strong> Center Hospital, Natori, Miyagi, Japan<br />

Background: Body mass index (BMI) may be an important factor<br />

affecting breast cancer outcome. Studies conducted mainly in Western<br />

countries have reported a relationship between higher BMI and a<br />

higher risk of all-cause death or breast cancer-specific death among<br />

women with breast cancer, but only a few studies have been reported<br />

in Japan so far. In the present prospective study, we investigated<br />

the associations between BMI and the risk of all-cause and breast<br />

cancer-specific death among breast cancer patients overall and by<br />

menopausal status and hormone receptor status. Methods: The study<br />

included 653 breast cancer patients admitted to a single hospital<br />

in Japan, between 1997 and 2005. BMI was assessed using a selfadministered<br />

questionnaire. The patients were completely followed<br />

up until December, 2008. Hazard ratios (HRs) and 95% confidence<br />

intervals (CIs) were estimated according to quartile points of BMI<br />

categories, respectively:


December 4-8, 2012 <strong>Abstract</strong>s: General Session 6<br />

P6-07-28<br />

Assessment of T stage in the multiple breast carcinomas<br />

Gong G, Jo J-H, Lee HJ, Kang J. University of Ulsan College of<br />

Medicine, Asan Medical Center, Republic of Korea; University of<br />

Ulsan College of Medicine, Gangneung Asan Hospital, Republic of<br />

Korea; Seoul National University Bundang Hospital, Republic of<br />

Korea; Haeundae Paik Hospital, Inje University College of Medicine,<br />

Republic of Korea<br />

Background: Tumor size is an important predictor of nodal metastasis,<br />

recurrence, distant metastasis in invasive breast carcinomas. T stage<br />

assignment of the multiple simultaneous ipsilateral breast carcinomas<br />

is based on the largest tumor with the suffix “(m)” to indicate<br />

multiplicity in the AJCC staging, and sum of the sizes should not<br />

be used. The aim of this study is to compare current AJCC staging<br />

and two test models using the aggregate of all tumor foci and the<br />

aggregate of all tumor volume in the single and multiple breast<br />

carcinomas. Methods: We reviewed original pathologic reports of<br />

1145 consecutive patients with invasive ductal carcinoma, 2003-2004.<br />

131 (11.4%) patients had multiple masses. Three factors were used to<br />

compare T stages; (1) the greatest dimension of the largest invasive<br />

component (T), (2) the aggregate of all tumor foci (TA), and (3) the<br />

aggregate of all tumor volume (TV). T stage follows AJCC (7th ed,<br />

2010) system. TA and TV were categorized as followings: TA1 < 2cm,<br />

2cm ≤ TA2 < 5cm and TA3 ≥ 5cm; TV1 < 8cm³ (average diameter <<br />

2cm), 8 cm³ ≤ TV2 < 125cm³ and TV3 ≥ 125cm³ (average diameter<br />

≥ 5cm). Clinicopathologic parameters were analyzed according to T<br />

stage. Results: Axillary lymph node metastasis was more common<br />

in the cases with multiple masses (64/131, 48.9%) than single mass<br />

(397/1014, 39.2%) and the number of the metastatic lymph node was<br />

also higher in multiple cases. Multiple breast carcinomas tended to<br />

show frequent recurrence, and disease specific death compared with<br />

the single cases in the same stage. (p < 0.05) Among 606 cases of<br />

T1, 34 cases were reclassified to TA2 and in 487 cases of the T2, 23<br />

cases were reclassified to TA3. TV also showed reclassification of the<br />

cases. Multiplicity, T stage, TA stage, and TV stage were significant<br />

factors for nodal positivity in the univariate analysis (p < 0.05). Nodal<br />

positivity was remained in the multivariate analysis for recurrence and<br />

disease specific death, but the other factors were not significant for<br />

nodal involvement, recurrence and disease specific death. Conclusion:<br />

Multiple breast carcinomas has adverse patients’ outcome than single<br />

carcinoma with same T stage of AJCC system and a better staging<br />

method is required to cover multiple carcinomas.<br />

P6-07-29<br />

Independent prognostic value of age depends on breast cancer<br />

subtype<br />

Brouckaert O, Salihi R, Laenen A, Vanderhaegen J, Amant F, Leunen<br />

K, Smeets A, Berteloot P, Van Limbergen E, Weltens C, Moerman P,<br />

Peeters S, Paridaens R, Floris G, Wildiers H, Vergote I, Christiaens<br />

M-R, Neven P. UZ Leuven; Interuniversity Centre for Biostatistics<br />

and Statistical Bioinformatics<br />

Background: Whether age is an independent prognostic factor in<br />

early breast cancer remains controversial. Different cut-off values<br />

have been suggested in different studies and few studies assessed its<br />

value in triple negative breast cancer and other phenotypes.<br />

Methods: We included all primary operable breast cancer patients<br />

from our prospectively managed database in UZ Leuven, Belgium.<br />

We excluded male patients, patients treated elsewhere first, patients<br />

treated with primary systemic treatment first. We assessed the effect<br />

of age as a continuous and categorical variable for distant metastasis<br />

free interval (DMFI), locoregional free interval (LRRFI) and breast<br />

cancer specific survival (BCSS) in multivariable analysis (correcting<br />

for phenotype, tumor size, nodal status, chemo-, endocrine-,<br />

radiotherapy, type of breast and axillary surgery). We assessed the<br />

effect of age for DMFI, LRRFI and BCSS in basal-like breast cancer<br />

and other phenotypes.<br />

Results: We included 4318 patients with a mean/median age of 58/ 57<br />

years (1170 patients were younger than 50 years) and with a median<br />

follow-up of 6,0 years. Multivariate analysis confirmed age as an<br />

independent prognostic variable for LLRFI (HR1.91; 95% CL:1.33-<br />

2.74; P=0.0005), DMFI (HR:1.77; 95% CL:1.33-2.36; P


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

In logistic regression analysis, significantly fewer HER2+HR+ pts<br />

achieved pCR compared to HER2+HR- pts regardless of the pCR<br />

definitions.<br />

Logistic regression analysis of pCR rate using diffrent definitions according to HR status<br />

HER2+HR- HER2+HR+ Odds ratio P<br />

N=74 % N=89 %<br />

ypT0 ypN0 20 27 12 14 0.42 0.033<br />

ypT0/is ypN0 27 36 17 19 0.37 0.005<br />

ypT0/is ypN0/+ 31 42 19 21 0.38 0.005<br />

(Conclusions) HER2+HR+ breast cancer pts had lower achievement<br />

rate of pCR, which was less prognostic compared to that in<br />

HER2+HR- pts regardless of pCR definitions. pCR is a suitable<br />

surrogate end point for HER2+HR- pts but not for HER2+HR+ pts<br />

in clinical trial settings.<br />

P6-07-31<br />

Assessing germline Homologous Recombination pathway<br />

deficiency in BRCA1 mutation carriers using Single Cell Network<br />

Profiling.<br />

Rosen DB, Leung LY, Louie B, Evensen E, Fields SZ, Cesano A,<br />

Shapira I, Hawtin RE. Nodality Inc, South <strong>San</strong> Francisco, CA;<br />

Monter <strong>Cancer</strong> Center, North Shore Long Island Jewish Medical<br />

School, Lake Success, NY<br />

Background: Inherited alterations in BRCA1/2 genes increase<br />

genomic instability and cancer susceptibility. DNA sequencing detects<br />

BRCA1/2 mutations, but has the following limitations; 1) mutations<br />

may have unknown functional significance, 2) epigenetic alterations<br />

and mutations in other Homologous Recombination (HR) pathway<br />

components are not detected, and 3) the combined effects of pathway<br />

mutations are not understood. Thus a functional assessment of HR<br />

competence at the single cell level remains an unmet need as BRCA1/2<br />

sequencing does not holistically inform on functionality of the HR<br />

pathway. Single Cell Network Profiling (SCNP) is a multiparametric<br />

flow cytometry-based assay that simultaneously measures, at the<br />

single cell level, extracellular surface markers and functional changes<br />

in intracellular signaling in response to extracellular modulators<br />

(Kornblau et al. Clin <strong>Cancer</strong> Res 2010). In this study, we tested the<br />

ability of SCNP to detect and quantify functional changes in HR<br />

signaling using peripheral blood mononuclear cell (PBMC) samples<br />

from BRCA1 mutation carriers (MUT) and wild type (WT) subjects.<br />

Methods: HR pathway activity was examined in PBMCs from<br />

BRCA1 MUT (n=21) or WT (n=20) subjects. Cell lines carrying<br />

BRCA1 MUTor WT genes were used as controls. PBMCs were<br />

stimulated with anti-CD3 and anti-CD28 for 24 hours to induce T<br />

cell proliferation then treated with PARP inhibitor (PARPi) AZD2281<br />

+/- Temozolomide (TMZ) for 48h or 72h to induce DNA damage.<br />

DNA damage response (DDR) readouts were measured in both<br />

CyclinA2- and CyclinA2+ T cell subsets. Measurements included<br />

induced levels of p21, p53 and phosphorylation (p-) of p-H2AX,<br />

p-DNA-PKcs, p-RPA2/32, and p-BRCA1.<br />

Results: As expected based on the mechanism of action of PARPi,<br />

higher levels of induced p-H2AX and p53 were observed in<br />

CyclinA2+ cells of BRCA1 MUT versus WT cell line controls. In<br />

PBMCs, T cell proliferation (%CyclinA2+) was positively associated<br />

with PARPi induced DDR readouts. After controlling for proliferation,<br />

statistically significant differences in PARPi induced DDR signaling<br />

were observed between BRCA1 MUT and WT samples in many<br />

simultaneously assessed readouts including p-H2AX, p53 and p21<br />

(increased in MUT), particularly in CyclinA2+ cells. Additionally,<br />

BRCA1 MUT samples displayed lower basal p-BRCA1 but higher<br />

induced p-BRCA1 levels compared to BRCA1 WT samples.<br />

Conclusions: SCNP was able to detect and quantify functional<br />

differences between PBMC samples from BRCA1 MUT<br />

(haploinsufficient) and WT donors by quantitatively assessing DDR<br />

signaling in CyclinA2+ T cells. Once verified on a larger data set, the<br />

assay could form the basis for the development of screening tests to<br />

identify subjects at higher risk of developing cancer or stratification<br />

tests to inform on cancer patient selection for treatment with PARP<br />

inhibitors.<br />

P6-07-32<br />

Prognosis of metastatic breast cancer subtypes: the hormone<br />

receptor/HER2 positive subtype is associated with the most<br />

favorable outcome<br />

Tjan-Heijnen VCG, Lobbezoo DJA, van Kampen RJW, Voogd AC,<br />

Dercksen MW, van den Berkmortel F, Smilde TJ, van de Wouw AJ,<br />

Peters FPJ, van Riel JMGH, Peters NAJB, Borm GF. Maastricht<br />

University Medical Center, Maastricht, Netherlands; Maxima<br />

Medical Center, Eindhoven, Netherlands; Atrium Medical Center<br />

Parkstad, Heerlen, Netherlands; Jeroen Bosch Hospital, Den<br />

Bosch, Netherlands; Viecuri Medical Center, Venlo, Netherlands;<br />

Orbis Medical Center, Sittard, Netherlands; St Elisabeth Hospital,<br />

Tilburg, Netherlands; St Jans Hospital, Weert, Netherlands; Radboud<br />

University Medical Center, Nijmegen, Netherlands<br />

Background<br />

In early breast cancer the different subtypes and their prognostic<br />

relevance are well known, contrary to the knowledge on prognostic<br />

relevance of subtypes in metastatic breast cancer. This study<br />

presents survival estimates after diagnosis of distant metastases of<br />

breast cancer subtypes in a recent cohort in which the treatment is<br />

representative of current clinical practice.<br />

Patients and methods<br />

All patients diagnosed with metastatic breast cancer in one of eight<br />

participating hospitals in the South-East part of the Netherlands<br />

between 2007-2009 were included and all medical charts were<br />

reviewed. Patients were categorized in 4 subtypes based on the<br />

hormone receptor (HR) and human epidermal growth factor receptor<br />

2 (HER2) status of the primary tumor; HR positive/HER2 negative,<br />

HR positive/HER2 positive, HR negative/HER2 positive and triple<br />

negative (TN). Metastatic survival was estimated using the Kaplan-<br />

Meier product-limit method. Cox proportional hazards model was<br />

used to determine the prognostic impact of breast cancer subtype,<br />

adjusted for possible confounders.<br />

Results<br />

A total of 815 patients were included; the HR+/HER2- subtype<br />

comprising 66%, the HR-/HER2+ subtype 8%, the TN subtype 15%<br />

and the HR+/HER2+ subtype 11% of patients. The four subtypes were<br />

associated with different metastatic survival times; the HR+/HER2+<br />

subtype was associated with the best metastatic survival (median,<br />

32.9 months), compared to 22.1 months for the HR+/HER2- subtype,<br />

19.5 months for the HR-/HER2+ subtype and 7.7 months for the TN<br />

subtype. Aside from subtype, other prognostic factors of metastatic<br />

survival were site of metastases and metastatic-free interval.<br />

Conclusion<br />

Of the four subtypes, the HR+/HER2+ subtype was associated with<br />

the most favorable outcome, in terms of metastatic survival.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

526s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: General Session 6<br />

P6-07-33<br />

A clinicopathologic analysis of 45 patients with metaplastic<br />

breast cancer<br />

Verma S, Cimino-Mathews A, Figueroa Magalhaes MC, Zhang Z,<br />

Stearns V, Connolly RM. Sidney Kimmel Comprehensive <strong>Cancer</strong><br />

Center at Johns Hopkins, Baltimore, MD; St. Agnes Hospital,<br />

Baltimore, MD; Johns Hopkins Hospital, Baltimore, MD<br />

Background:<br />

Metaplastic breast carcinoma (MBC) is a rare tumor, accounting for<br />

< 1% of all breast cancers. The 5-year survival rate is approximately<br />

65% vs. 89% for conventional invasive ductal cancer (IDC), therefore<br />

improved treatment options are required. Evidence as to how to<br />

manage patients with MBC is derived from a number of single<br />

institution case series, and usually follows the treatment paradigm for<br />

conventional IDC. We aimed to review the clinicopathologic features<br />

of women with MBC treated at our institution to add to the current<br />

literature, and to identify potential prognostic biomarkers of outcome.<br />

Patients and Methods: The study was approved by the Institutional<br />

Review Board of John Hopkins University. We retrospectively<br />

identified patients > 18 yrs of age with histologic confirmed diagnosis<br />

of MBC through the Johns Hopkins Pathology archives from March<br />

1999-February 2012. We collected and reviewed data relating to<br />

clinicopathological features, local and systemic treatments, and<br />

patient outcomes. Survival probabilities at specific time points were<br />

estimated using the Kaplan-Meier method and confidence intervals<br />

(CI) obtained using the Greenwood formula.<br />

Results: Of 45 cases identified, median age was 59 (range 38-93),<br />

62% of patients were white and 36% were black. Histologic subtypes<br />

identified were chondroid (24%), spindle (20%), sarcomatoid<br />

(16%), squamous (11%) and mixed (29%). Median tumor size<br />

3cm (range 1-14.8cm); lymph node status was positive (24.4%) /<br />

negative (51.2%) /unknown (24.4%); 60% had grade 3 tumors;<br />

69% were estrogen receptor (ER)/progesterone receptor (PR)/<br />

HER2-negative (triple-negative), 20% ER/PR-positive, with 4<br />

HER2 positive tumors and 3 HER2-unknown. Two patients had<br />

metastatic disease at diagnosis. Neoadjuvant therapy (anthracyclinetaxane<br />

based in 83%) was administered in 6 patients, with one<br />

patient achieving complete pathological response to dose dense<br />

adriamycin and cyclophosphamide followed by weekly paclitaxel.<br />

All patients underwent surgery (82% negative margins) and 60%<br />

received adjuvant radiation. Adjuvant chemotherapy, predominantly<br />

anthracycline +/- taxane-based was administered in 58% cases<br />

(excluding neoadjuvant). 5 year recurrence-free survival (n=43)<br />

was 63.3% (95% CI, 0.382-0.884) with median follow up of 26.2<br />

months (range 1.7-137.2 months). 5 year overall survival (n=45)<br />

was 68.7% (95% CI, 0.448-0.926) at median follow up 28 months<br />

(range 2.4-137.9 months).<br />

Conclusion: We report one of the largest single institution case series<br />

of clinicopathologic features associated with MBC in the literature.<br />

MBC is associated with a worse prognosis than conventional IDC,<br />

despite a relatively low rate of positive nodal involvement. The<br />

majority of patients in this series had high grade, triple-negative<br />

tumors and were treated with optimal local and systemic therapy. We<br />

will report results of subgroup analyses at the time of presentation. We<br />

also aim in the future to analyze available archival tumors to identify<br />

potential prognostic and predictive biomarkers, and anticipate that<br />

these results will provide the rationale for clinical trials with novel<br />

investigational agents in this area of unmet need.<br />

P6-07-34<br />

Disease-related outcomes with adjuvant chemotherapy in HER2<br />

positive or triple negative T1a/bN0 breast cancers<br />

Shao T, Olszewski AJ, Boolbol SK, Migdady Y, Boachie-Adjei K, Sakr<br />

BJ, Klein P, Sikov W. Beth Israel Medical Center, Continuum <strong>Cancer</strong><br />

Centers of New York, New York, NY; The Warren Alpert Medical<br />

School of Brown University, Providence, RI<br />

Background: Previous studies reported higher relapse rates in<br />

T1a/bN0 breast cancers characterized by high-risk biology (HER2<br />

positive or triple negative). The benefits of adjuvant chemotherapy<br />

in this group have not been evaluated. The purpose of our study<br />

was to determine potential impact of chemotherapy on incidence of<br />

relapses and to define the population appropriate for treatment based<br />

on observational data.<br />

Methods: We pooled data from two multi-institutional databases<br />

spanning the period of 1996-2008 (Beth Israel Medical Center) and<br />

2000-2010 (Brown University). We fitted a propensity score model<br />

to adjust for unbalanced confounders between the groups treated or<br />

untreated with adjuvant chemotherapy and, in case of HER2+ disease,<br />

with trastuzumab. Competing risk analysis was employed to study the<br />

effect of chemotherapy on cancer relapses in the matched population.<br />

Results: The study included 321 patients (160 from Beth Israel and<br />

161 from Brown) with a median age of 57 years (range 28-88). 111<br />

(35%) cases were triple negative (TN) and 210 (65%) were HER2+ (of<br />

which 64% were ER+). 41% patients received adjuvant chemotherapy<br />

and 22% of HER2+ cases received trastuzumab. The treated<br />

group differed from untreated with regards to distribution of age,<br />

menopausal status, year of diagnosis, histological subtype and grade,<br />

tumor size, rates of mastectomy, and adjuvant endocrine therapy. All<br />

confounders were successfully balanced with the propensity score<br />

analysis. With a median follow-up of 56 months, 20 relapses (12<br />

locoregional and 8 distant) and 10 unrelated deaths occurred. The<br />

cumulative incidence of relapse at 5 years was 7.3% (95% CI, 4.3-<br />

11.4) and relapse-free survival was 90.2% (95% CI, 85.2-93.6). Age<br />

less then 35 years (46.4% vs. 6.1%, p=0.0004) and TN status (12.9%<br />

for TN versus 4.8% for HER2+, p=0.03) were associated with higher<br />

risk of relapse. No significant differences with regards to tumor size<br />

(T1a or T1b), surgery type and other variables were observed. There<br />

was no significant effect of chemotherapy on incidence of relapse<br />

(Hazard ratio 1.2, 95% CI 0.34-4.23, p=0.78), relapse-free survival<br />

or distant recurrence-free interval after propensity score adjustment.<br />

Additionally in the HER2+ patients, no benefit of trastuzumab was<br />

detected (hazard ratio 0.78, 95% CI 0.06-9.85, p=0.85). Adjuvant<br />

endocrine therapy was associated with lower risk of relapse in the<br />

ER+ subgroup (p=0.04).<br />

Conclusion: Survival outcomes in very early stage breast cancer<br />

are excellent with current therapy even in biologically aggressive<br />

subtypes. Triple negative status and very young age correlate with<br />

higher incidence of relapse. While adjuvant endocrine therapy may be<br />

effective in ER positive subset, the risk/benefit ratio of chemotherapy<br />

with or without trastuzumab remains uncertain.<br />

P6-07-35<br />

Dragonfly Effect or Ironic Paradox: Prognostic Implication of<br />

Postsurgical Drain in <strong>Breast</strong> <strong>Cancer</strong> Patients<br />

Yin W, Lin Y, Shen Z, Shao Z-M, Lu J. Fudan University Shanghai<br />

<strong>Cancer</strong> Center, Shanghai, China<br />

Background: Surgical removal of primary breast cancer may result<br />

in subsequent alterations of host milieu. Massive clinical data have<br />

indicated that surgical extirpation favorably modifies the natural<br />

history for some patients, but it may also precipitate the metastatic<br />

www.aacrjournals.org 527s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

outgrowth for others. Drainage fluid, harvested from a post-surgical<br />

wound, appears to be a physiological response to surgical trauma.<br />

However, few clinicians have recognized that the volume of wound<br />

drain might mirror the postoperative recurrence kinetics.<br />

Methods: A total of 1439 patients were selected retrospectively<br />

from a large database of patients who underwent surgery between<br />

Jan 1, 2000 and Dec 31, 2002 in Fudan University Shanghai <strong>Cancer</strong><br />

Center, Shanghai, China. The Spearman rank correlation analysis<br />

was conducted to evaluate the association between drainage volume<br />

of postoperative day 1 (POD1) and clinicopathological parameters,<br />

which included age at diagnosis, body weight at diagnosis, total<br />

number of excised axillary lymph nodes (ALNs), time to drainage<br />

volume less than 30ml (TTV30), total protein and albumin<br />

concentrations in serum. Survival curves were performed with<br />

Kaplan–Meier method and annual recurrence hazard was estimated<br />

by hazard function.<br />

Results: Drainage volume of POD1 was positively correlated with age<br />

at diagnosis (Spearman’s P < 0.001), weight at diagnosis (P


December 4-8, 2012 <strong>Abstract</strong>s: General Session 6<br />

P6-07-38<br />

The Value of Progesterone Receptor in Predicting the Recurrence<br />

Score for Hormone-Receptor–Positive Invasive <strong>Breast</strong> <strong>Cancer</strong><br />

Patients<br />

Onoda T, Yamauchi H, Yagata H, Hayashi N, Yoshida A, Nakamura<br />

S. St Luke’s International Hospital, Tokyo, Japan; Showa University,<br />

Tokyo, Japan; Yokohama Asahi Central General Hospital, Kanagawa,<br />

Japan<br />

Background: Molecular genomic profiling currently plays a<br />

major role in treatment decisions for breast cancer patients. The<br />

21-gene signature (Oncotype DX ® [ODX]) is one of the most wellvalidated<br />

assays. Herein we evaluated the associations between high<br />

Recurrence Score (RS) measured by the ODX and pathological<br />

factors. Methods: From October 2007 to October 2010, the ODX<br />

assay was performed for 139 hormone-receptor–positive invasive<br />

breast cancer patients at single institution. Correlations between the<br />

RS and the following pathological factors were analyzed: tumor size<br />

(T), lymph node metastasis (N), nuclear atypia (NA), mitotic counts<br />

(MC), nuclear grade (NG), lymphatic and vascular invasion (LI and<br />

VI), estrogen receptor (ER), progesterone receptor (PgR), HER2, and<br />

Ki-67. Expression of ER, PgR, HER2, and Ki-67 was examined by<br />

immunohistochemistry. The Ki-67 labeling index was automatically<br />

counted with an Ariol-SL50 instrument (APPLIED IMAGING). The<br />

other pathological factors were estimated under the supervision of<br />

one experienced pathologist. Spearman rank correlation coefficients<br />

were calculated for the analyses. When the r was >0.4 or


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

and PgR-negative status might indicate poor prognosis. However,<br />

this study was a retrospective analysis of a limited, heterogeneous<br />

patient group. A large-scale cohort study in the Japanese population<br />

is, therefore, recommended.<br />

P6-07-40<br />

Initial Neutrophil-to-Lymphocyte ratio in primary breast cancer<br />

patients: a simple and useful biomarker as prognostic factor<br />

Han A, Noh H, Lee J. Yonsei Univeristy Wonju Medical School, Wonju,<br />

Gangwon, Republic of Korea<br />

Background: Increasing neutrophil/lymphocyte ratios (NLR) on<br />

preoperative blood tests have been associated with worse survival<br />

of patients with colorectal, gastric, hepatocellular, pancreatic and<br />

lung cancer. However the utility of NLR to predict mortality in<br />

breast cancer patients has not been studied. The aim of our study<br />

was to determine whether the NLR is predictive of disease specific<br />

mortality in breast cancer and subtype-adjusted prognostic effect in<br />

breast cancer patients.<br />

Methods: We retrospectively identified all the patients with primary<br />

breast cancer diagnosed at Yonsei University Wonju Medical School<br />

between January 2000 and June 2011 who underwent primary<br />

surgery and completed all phase of their treatment. <strong>Breast</strong> cancer<br />

with infiltrating ductal histology was included and divided into 4<br />

subtypes: Luminal A, Luminal B, Her2-enriched, and basal type. Each<br />

immunohistochemical characteristics are as followings: Luminal A:<br />

ER and/or PgR positive and negative Her2, Luminal B: ER and/or<br />

PgR positive with positive HER2, Her2-enriched: negative ER and<br />

PR with positive Her2, and triple negative as basal.<br />

Results: Patients with NLR≥2.5 showed significantly lower 5-year,<br />

10-year breast cancer specific survival than patients with NLR


December 4-8, 2012 <strong>Abstract</strong>s: General Session 6<br />

Patients diagnosed as invasive breast cancer without Paget’s disease<br />

were recruited in control group for clinical and pathologic features<br />

comparison.<br />

Since tumors in PD group expressed more high risk factors (see<br />

results), a matched study was done to investigate whether the poor<br />

survival of PD group was by nature or due to the overexpression of<br />

high risk factors. The matched group was derived from all the patients<br />

who received mastectomy for invasive breast cancer (with no Paget’s<br />

disease) during 2001 to 2005 in FUSCC. The match was conducted<br />

according to four variables: dimension of tumor, lymph node status,<br />

hormone receptor status and Her2 status. After matching, patients<br />

were randomly selected in a 3:1 ratio to patients in PD group.<br />

[Results]<br />

52 patients were recruited in PD group. 22 (42.3%) patients’ primary<br />

symptom were nipple change, 25 (48.1%) patients’ primary symptom<br />

were mass in breast. Physical examination found no nipple-areola<br />

change in 24 patients at diagnosis, their Paget’s disease were found<br />

unexpectedly by routine nipple-areola section examination after<br />

mastectomy.<br />

More than 8000 invasive breast cancers were diagnosed during 2001<br />

to 2005. 700 patients (140 consecutive patients per year, average<br />

monthly patient amount) were recruited in control group. Compared<br />

with control group, tumors in PD group had larger size, more axillary<br />

lymph node involvement, lower hormone receptor expression, higher<br />

HER2 expression and worse survival.<br />

Comparison of Pathologic Features (Control Group) and Survival (Matched Group) with PD Group<br />

PD group (n=52) Control Group (n=700) Matched Group (n=156)<br />

Dimension ≤2cm 24 (46.2%) 423 (60.4%)∗ 72 (46.2%)<br />

>2cm 28 (53.8%) 277 (39.6%) 84 (53.8%)<br />

Lymph Node + 28 (53.8%) 250 (35.7%)∗ 84 (53.8%)<br />

- 24 (46.2%) 450 (64.3%) 72 (46.2%)<br />

Hormone receptor + 18 (34.6%) 488 (69.7%)∗ 54 (34.6%)<br />

- 34 (65.4%) 212 (30.3%) 102 (65.4%)<br />

Her2 + 40 (76.9%) 149 (21.3%)∗ 120 (76.9%)<br />

- 12 (23.1%) 551 (78.7%) 36 (23.1%)<br />

5y Relapse-Free Survival 52.2% 86.7%∗ 81.4%∗<br />

5y Overall Survival 62.1% 91.8%∗ 85.9%∗<br />

∗compared with PD group, P


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

of the breast, diagnosed between 2000 to 2009 that had undergone<br />

treatment in the National <strong>Cancer</strong> Institute of Brazil.<br />

OBJECTIVES<br />

Analyze overall survival and the main prognostic factors among<br />

women with infiltrating lobular carcinoma of the breast, diagnosed<br />

from 2000 to 2009 that had undergone treatment in the National<br />

<strong>Cancer</strong> Institute of Brazil.<br />

MATERIALS AND METHODS<br />

The survival curves were obtained in a hospital cohort of 843 patients<br />

with infiltrating lobular carcinoma of the breast diagnosed and treated<br />

between 2000 to 2009. The study variables were: age, race, tobacco,<br />

alcohol consumption , tumor-related variables, and treatment-related<br />

variables. Survival functions were calculated by the Kaplan-Meier<br />

method.<br />

RESULTS<br />

The mean follow up time was 64,27 months (sd 33,76), with 30,2% of<br />

deaths occurred in the period where the mean patients age was 58,58<br />

years (sd 13,22). The overall survival (OS) was 105,61 months (95%<br />

CI, 101,68 to 109,54). The OS for patients with 4 or more positives<br />

axillary lymph nodes was 91,66 months (95% CI, 83,19 to 100,13;<br />

p


December 4-8, 2012 <strong>Abstract</strong>s: General Session 6<br />

RESULTS: 47 PR and 89 HR girls have completed surveys. Age did<br />

not differ between groups (Mage=15.6; SD=2.4). 30% of HR girls<br />

have a mother with BC. 67% of HR girls vs. 30% of PR girls reported<br />

self-perceived risk for adult BC to be “higher than other girls my<br />

age,” (p=


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P6-08-03<br />

Informational needs and psychosocial assessment of patients in<br />

their first year after metastatic breast cancer diagnosis<br />

Seah DS, Lin NU, Curley C, Winer E, Partridge A. Dana-Farber<br />

<strong>Cancer</strong> Institution, Boston, MA<br />

Background:<br />

Psychosocial distress is common after a diagnosis of breast cancer.<br />

Little is known about the informational needs and the psychosocial<br />

adjustment of patients diagnosed with metastatic breast cancer (MBC)<br />

within the first year of their diagnosis.<br />

Methods:<br />

Patients with MBC from a single academic institution completed a<br />

cross-sectional self-administered paper survey. The survey included<br />

demographics, the Medical Outcomes Study Short Form-36 (SF-36),<br />

the Hospital Anxiety and Depression Scale (HADS), and Toronto<br />

Informational Needs Questionnaire-<strong>Breast</strong> <strong>Cancer</strong> (TINQ). Medical<br />

history was obtained by chart review. The Spearman correlation<br />

coefficient assessed the relationship between TINQ and the following:<br />

age at MBC diagnosis, disease free interval (DFI), time between<br />

survey completion and MBC diagnosis, number of lines of therapy,<br />

and HADS.<br />

Results:<br />

Fifty-two (90%, 50F 2M) patients completed the survey. Median<br />

age at MBC diagnosis was 52 yrs (range 22-81). Thirty-nine (75%)<br />

patients had completed college, 92% were Caucasian. Median time<br />

between MBC diagnosis and survey completion was 6 months (range<br />

1-12). Sixteen (31%) patients had de novo stage 4 disease. At time of<br />

survey completion, 36 (69%) patients were on 1st line therapy with<br />

some patients were receiving their 4th line of therapy. SF-36 scores<br />

were lower in all 8 subscales compared to the general population.<br />

In particular, role limitations due to physical health (Norm-based<br />

transformation mean score 39.3, SD=12.1), social functioning (Mean<br />

41.8, SD=12.7), role limitations due to emotional problems (Mean<br />

43.3, SD=13.3), vitality (Mean 44.1, SD=10.8) and general health<br />

(Mean 44.3, SD=12.1) were diminished. The Physical and Mental<br />

Component Summary norm-based transformation scores were 43.2<br />

(SD=11.7) and 45.4 (SD=11.3) respectively.9/48 (19%) patients<br />

met criteria for anxiety, and 4/48 (8%) patients met criteria for<br />

depression by HADS criteria (scores > 11). TINQ scores range from<br />

51 to 255, with 35/52 (69%) having a total score > 200, suggesting<br />

high informational need. Of the 5 subscales, treatment information<br />

was most important, followed by information about disease, physical<br />

care, psychosocial needs and investigative tests. The most important<br />

informational issues for patients were: if there was cancer anywhere<br />

else in their body (Mean score 4.78), how to deal with side effects<br />

(Score 4.78), and if there were ways to prevent treatment side effects<br />

(Score 4.77), with a score of 5=extremely important, and 1= not<br />

important.<br />

Only DFI correlated with TINQ (Spearman coefficient -0.413,<br />

P=0.011), with patients who had a shorter DFI having greater<br />

informational needs. Age at MBC diagnosis, time of completion of<br />

survey, number of lines and HADS were not significant.<br />

Conclusion:<br />

Based on this study, patients with recently diagnosed MBC have<br />

high informational needs and poor psychosocial adjustment. The<br />

overall quality of life appears to be worse in this population of<br />

patients compared to the general population. There is also a subset<br />

of patients who are dealing with significant anxiety and depression.<br />

Additional research, education, and supportive care services aimed at<br />

meeting the informational and psychosocial needs of women living<br />

with MBC are warranted.<br />

P6-08-04<br />

The Impact of a <strong>Breast</strong> <strong>Cancer</strong> Diagnosis in Young Women on<br />

their Relationship with their Mothers<br />

Ali A, Fergus K, Wright F, Pritchard K, Kiss A, Warner E. Sunnybrook<br />

Health Sciences Centre, Odette <strong>Cancer</strong> Centre, University of Toronto,<br />

Toronto<br />

Background: <strong>Breast</strong> cancer in younger women (≤ 40) is associated<br />

with greater physical and psychological morbidity than in older<br />

women. No study to our knowledge has examined the effect of a<br />

breast cancer diagnosis in young women on their relationship with<br />

their mothers, or the support needs of these mothers. Methods: We<br />

developed and pre-tested a self-administered questionnaire on 10<br />

survivors of breast cancer diagnosed ≤ age 40 for clarity, content<br />

and sensitivity (<strong>San</strong> <strong>Antonio</strong>, 2011). Then, consecutive recurrencefree<br />

women age ≤ 40 at diagnosis, within 4 years of a breast cancer<br />

diagnosis, whose mothers were alive at diagnosis, were asked to<br />

complete the modified questionnaire in a medical oncology follow-up<br />

clinic. Results: Of 110 eligible daughters approached from 07/2011<br />

– 05/2012, 90 with a mean age 36 (range 21-43) participated in the<br />

study. Stage at diagnosis: 1- 23 (26%), 2- 46 (51%) 3- 21 (23%).<br />

Ethnicity: Asian: 29 (32%), Caucasian 43 (48%), Black 2 (2%), Mix/<br />

Other 16 (18%). At diagnosis, 15 were living with their mothers, 44<br />

were not living in the same city including 23 who were in different<br />

countries, 7 of whom were not informed of the diagnosis as the<br />

daughters did not want to worry them. Mean age of the 88 mothers<br />

alive at time of study was 64 (range 48-84) and 16 had previously<br />

had breast cancer. Illness attributions: Eight blamed their mothers<br />

for their developing breast cancer, and 22 believed their mothers<br />

felt responsible to some extent (overlap in 4). Supports: Of the 43<br />

daughters who had turned to their mothers for emotional support<br />

over the year prior to diagnosis, all but one did so after diagnosis,<br />

as did 20% of those who had not turned to mothers over the year<br />

prior to diagnosis. In 11 cases, the daughters turned to their mothers<br />

before approaching anyone else for support. Of the 83 daughters<br />

who informed mothers of their diagnosis, 76 (92%) reported their<br />

mothers were emotionally and/or practically supportive. The 4 most<br />

difficult issues faced by daughters were fatigue, anxiety, breast loss<br />

and menopausal symptoms, with 70 daughters discussing at least<br />

some issues with their mothers. Of the 35 working mothers, 27 took<br />

time off to support their daughters. Nineteen mothers slept over or<br />

moved in with the daughters during their treatment, 8 of whom had<br />

been living in a different country. Forty-four (44/83=53%) daughters<br />

reported that the breast cancer diagnosis had a favorable impact on<br />

their relationship with their mothers. Formal supports for mothers:<br />

Thirty-two (32/83=39%) reported their mothers did not have adequate<br />

psychosocial support, and 59 (59/90=66%) indicated health care<br />

professionals could help mothers by providing brochures on caring<br />

for a daughter with breast cancer, having professionally led education<br />

sessions, as well as support groups. Conclusion: Mothers are an<br />

important source of support for young breast cancer daughters, and<br />

most daughters perceived that the diagnosis resulted in the motherdaughter<br />

relationship becoming closer. However, the physical and<br />

emotional toll on mothers appears to be high. Future studies should<br />

address the effects of a breast cancer diagnosis in a young daughter<br />

from the mothers’ perspective, and the benefit of formal, culturally<br />

sensitive supports for these mothers.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

534s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: General Session 6<br />

P6-08-05<br />

Changes in Cognitive Function in Older Women With <strong>Breast</strong><br />

<strong>Cancer</strong> Treated with Standard Chemotherapy and Capecitabine<br />

within CALGB 49907<br />

Freedman RA, Pitcher B, Keating NL, Barry WT, Ballman KV,<br />

Kornblith A, Mandelblatt J, Kimmick GG, Hurria A, Winer EP, Hudis<br />

CA, Cohen HJ, Muss HB. Dana-Farber <strong>Cancer</strong> Institute, Boston, MA;<br />

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

School, Boston, MA; Mayo Clinic, Rochester, MN; Georgetown<br />

University, Washington, DC; City of Hope, Duarte, CA; Memorial<br />

Sloan-Kettering, New York, NY; University of North Carolina, Chapel<br />

Hill, NC<br />

Background<br />

Cognitive changes for older women who receive chemotherapy are<br />

poorly understood.<br />

Methods<br />

<strong>Cancer</strong> and Leukemia Group B (CALGB) 49907 enrolled 633 women<br />

≥65 years with early stage breast cancer and randomized them to<br />

standard adjuvant chemotherapy (AC or CMF) or capecitabine (N<br />

Eng J Med 360:2055, 2009). Patients enrolled on the Quality of Life<br />

companion study (N = 350) included women who had cognitive<br />

and psychological functioning sufficient for providing informed<br />

consent and participation in the trial (J Clin Oncol 29:1022, 2011).<br />

Participants completed an 18 item questionnaire focusing on their<br />

subjective report of cognitive function (attention, problem-solving,<br />

speed, new learning, and remote memory) at 6 time points (baseline,<br />

mid-treatment, within 1 month post treatment, and at 12, 18, and 24<br />

months post treatment). Possible scores for each item ranged from 1-7<br />

(‘above average’ - ‘severe problem’). Cognitive function score (CFS)<br />

was defined as the mean score of items 1-18. Cognitive impairment<br />

was defined as CFS 1.5 standard deviations above the overall average<br />

baseline. Univariate associations were tested between baseline CFS<br />

and patient characteristics (treatment, race, education, number of<br />

positive nodes, hormone receptor status, HER2 status, performance<br />

status, number of comorbidities, fatigue and depression) using<br />

Kruskal-Wallis testing. Frequency distributions were used to describe<br />

cognitive impairment. Multivariate analyses were not completed<br />

because few women reported subjective cognitive impairment during<br />

the study period.<br />

Results<br />

297 assessments were received at baseline, 280 at both mid-treatment<br />

and within 1 month post-treatment, 259 at 12 months, 245 at 18<br />

months and 240 at 24 months post-treatment. The median age was 72<br />

(65-85); 32% received AC, 21% CMF, and 47% capecitabine. Most<br />

women were white (87%) and had a performance status of 0 (73%).<br />

Approximately half of women had a high school degree or less and<br />

the majority had stage I-II (78%), hormone-receptor positive (68%)<br />

cancers and most did not experience depression or fatigue at baseline.<br />

At baseline, the overall mean CFS was 2.1 (‘normal ability’) and 19<br />

women (6%) had mild to severe impairment. Baseline scores did not<br />

differ by treatment (p=0.35). Worse cognitive function at baseline<br />

was associated with less education (p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P6-08-07<br />

Quality of life of women with breast cancer: A Middle East<br />

perspective<br />

Jassim GA, Whitford DL. Royal College of Surgeons in Ireland-<br />

Medical University of Bahrain, Busaiteen, Bahrain<br />

Background: <strong>Breast</strong> cancer is the most common cancer among<br />

women worldwide. Scarce information is currently available on the<br />

quality of life (QoL) of breast cancer survivors in the Middle East<br />

region. The objective of this study is to describe QoL of Bahraini<br />

women with breast cancer and investigate the association between<br />

their QoL and their sociodemographic and clinical data.<br />

Methods: This is a cross sectional study in which the Arabic version<br />

of the European Organization for Research and Treatment of <strong>Cancer</strong><br />

general quality of life questionnaire (QLQ-C30) and the disease<br />

specific questionnaire (QLQ-BR23) were administered to a random<br />

sample of 337 Bahraini women with breast cancer.<br />

Statistical analysis: The collected data were coded, entered and<br />

analyzed using the statistical package SPSS. Relevant descriptive<br />

statistics were computed for all items. A higher score represents a<br />

“better” level of functioning, or a “worse” level of symptoms. The<br />

“Score” served as the dependent (Outcome) variable in the study<br />

for the purpose of data analysis. Sociodemographics and clinical<br />

information represented the independent variables. The equality of<br />

means across categories of each categorical independent variable<br />

was tested using ANOVA and independent t-test. Non-parametric<br />

tests (Kruskal Wallis and Mann Whitney tests) were used instead if<br />

the statistical assumptions of using the parametric tests were violated.<br />

Results: Of the total sample only 239 consented to participation. The<br />

mean age of participants was 54.4 (SD±10.7). Participants had a mean<br />

score for global health of 63.9 (SD±21.3). Among functional scales,<br />

social functioning scored the highest mean (77.5) whereas emotional<br />

functioning scored the lowest (63.4). The most distressing symptom<br />

on the symptom scales of QLQ-C30 was fatigability (Mean 35.2).<br />

Using the disease specific tool (QLQ-BR23) it was found that sexual<br />

functioning scored the lowest mean (25.9) whereas “upset due to hair<br />

loss” symptom scored the highest mean (46.3). Significant mean<br />

differences for various functional and symptom scales were observed<br />

among categories of age, marital status, type of surgery, stage of<br />

disease, monthly income, educational status and time since diagnosis.<br />

Discussion: Comparable global QoL mean was reported by<br />

participants in this study despite the limited structured psychological<br />

support available for breast cancer Bahraini women compared to<br />

advanced services in the western community. Women showed poor<br />

performance on sexual functioning and enjoyment. One of the reasons<br />

might be that single women were under less pressure to worry about<br />

their partner’s opinion because traditionally and religiously the local<br />

society is restricting dating and premarital sex. On the other hand,<br />

married women were constantly intimidated by the second wife’s<br />

option.<br />

Conclusion: Bahraini women showed good functioning on most<br />

functional and symptom scales. Poorest functioning was reported for<br />

emotional and sexual domains. On the symptom scales, fatigue and<br />

arm morbidities were the most bothersome symptoms. Differences<br />

in several QoL domains are discussed in view of sociocultural and<br />

religious aspects. Health professionals need to develop a culturally<br />

sensitive clinical approach to enhance QoL of women with breast<br />

cancer in the Middle East region.<br />

P6-08-08<br />

Three months of adjuvant hormone therapy does not increase<br />

fatigue or cognitive failure: results of a prospective early stage<br />

breast cancer trial.<br />

Kennedy D, Lower EE. Oncology Hematology Consultants,<br />

Cincinnati, OH<br />

INTRODUCTION: The diagnosis and treatment of breast cancer<br />

may alter a patient’s mental and physical quality of life. Fatigue<br />

and impaired cognition have been reported after chemotherapy and<br />

hormone therapy. The purpose of this study was to prospectively<br />

determine the incidence of fatigue and cognitive changes in early<br />

stage breast cancer patients during adjuvant hormone therapy and<br />

to evaluate the relationship of dysfunction with other risk factors.<br />

METHODS: Fifty-four consecutive patients of a single medical<br />

oncologist with newly diagnosed hormone dependent breast cancer<br />

prescribed adjuvant hormone therapy enrolled in this prospective<br />

study. Data collected included age, race, menopausal status, breast<br />

cancer stage, body mass index, Functional Assessment of Chronic<br />

Illness Therapy-Fatigue (FACIT)-F, Fatigue Assessment Scale (FAS),<br />

short form 36 (SF-36), cognitive failure questionnaire (CFQ), Beck<br />

depression inventory (BDI-2) , Epworth sleepiness scale, and facial<br />

recognition (FR) and sheep dash time (SDT) to assess executive<br />

functioning. All tests were administered by one nurse prior to hormone<br />

initiation (0 mo) and after 3 months (3 mo) of therapy. Statistical<br />

analysis was performed using Student t test for paired data with a p<br />

value of


December 4-8, 2012 <strong>Abstract</strong>s: General Session 6<br />

P6-08-09<br />

Overcoming <strong>Breast</strong> <strong>Cancer</strong>: The Importance of Connecting with<br />

Fellow Survivors<br />

Geoghegan C, Whitman B. Y-ME National <strong>Breast</strong> <strong>Cancer</strong><br />

Organization, Chicago, IL; Whitman Insight Strategies, New York, NY<br />

Y-ME National <strong>Breast</strong> <strong>Cancer</strong>’s Organization, founded in 1978,<br />

is not-for-profit organization with a mission to “To assure through<br />

information, empowerment, and peer support, that no one faces breast<br />

cancer alone.” Each year, Y-ME’s 24x7x365 <strong>Breast</strong> <strong>Cancer</strong> Hotline<br />

receives approximately 40,000 contacts. Callers to Y-ME’s toll-free<br />

Hotline are referred by clinicians, community groups and global<br />

cancer organizations that recognize the importance of providing<br />

emotional relief to patients and their loved ones. Trained and certified<br />

peer counselors, all breast cancer survivors, staff the hotline. Callers<br />

can speak to a Spanish or English speaking peer counselor or have an<br />

interpreter in any of over 150 languages added to the line.<br />

An online survey conducted by Whitman Insight Strategies on behalf<br />

of the Y-ME National <strong>Breast</strong> <strong>Cancer</strong> Organization was conducted to<br />

better understand the psychosocial needs of breast cancer patients and<br />

survivors. Surveys were conducted on April 3-5, 2012, among 400<br />

breast cancer patients and survivors 18 years of age and older across<br />

the United States. The margin of error for total sample is ±4.9% at<br />

the 95% confidence interval. Respondents received an email invite<br />

to participate in a short online survey; their emails were selected at<br />

random among online market research panelists who had previously<br />

indicated that they have or had breast cancer.<br />

The results indicate that talking to a breast cancer survivor can be<br />

instrumental in overcoming breast cancer: 84% of breast cancer<br />

patients and survivors say this is one of the most important ways<br />

of coping with breast cancer and 68% wish that they could have<br />

been connected to other survivors. Access to a 24x7x365 hotline,<br />

where breast cancer survivors provide emotional support and<br />

practical advice, is considered important by virtually all women<br />

surveyed (95%)--73% think this is “extremely” or “very” important.<br />

Unfortunately, although Y-ME provides such a hotline; however,<br />

awareness of it is extremely low (14%). We encourage all healthcare<br />

providers to encourage patients to reach our to Y-ME by calling 800-<br />

221-2141 and time of the day or night.<br />

Those who had another survivor to talk were asked to select among<br />

a number of relevant responses to the question: What did talking<br />

to another breast cancer patient/survivor provide you with? The<br />

following table provides the proportion that selected each response.<br />

Proportion Selecting Each Response<br />

Response<br />

% of Responders<br />

A feeling that I was not alone 70%<br />

A better idea of what to expect 61%<br />

Emotional support 60%<br />

Hope 53%<br />

Optimism 44%<br />

Better understanding of what diagnosis means 41%<br />

Peace of mind 38%<br />

Questions to ask doctor/healthcare provider 37%<br />

Someone I could count on 34%<br />

Needed information about how to cope 32%<br />

Help for my family 17%<br />

P6-08-10<br />

<strong>Cancer</strong> as self: a novel assessment of patient identity as it relates<br />

to a cancer diagnosis<br />

Horst KC, Fero KE, Haimovitz K, Dweck CS. Stanford University,<br />

Stanford, CA<br />

Introduction:<br />

Previous studies have demonstrated that an individual’s attitude<br />

towards self may impact how one faces challenges. Those who view<br />

themselves as malleable (growth mindset) approach challenges and<br />

problems as a learning opportunity and an opportunity to improve<br />

and change. In contrast, those who are more fixed in their approach<br />

(fixed mindset) have difficulty with challenges or setbacks. In the<br />

present study, we applied this conceptual framework to a cohort of<br />

breast cancer patients to evaluate their attitudes towards the cancer<br />

diagnosis. The goal of this study is to test the internal validity of<br />

this questionnaire and to correlate its findings with patient-reported<br />

measures of depression, anxiety and insomnia.<br />

Methods:<br />

Twenty consecutive breast cancer patients undergoing radiation<br />

therapy completed an electronic survey consisting of 4 scales: the<br />

Hospital Anxiety and Depression Scale (HADS; 14 items), the<br />

Insomnia Severity Index (ISI; 7 items), a validated psychological<br />

questionnaire assessing fixed versus growth mindset (8 items), and<br />

an experimental questionnaire evaluating whether the patient views<br />

their disease as self-defining. Demographic and clinical variables<br />

were also collected. A principal components factor (PCF) analysis<br />

was used to examine the internal structure of the new questionnaire.<br />

Hierarchical linear regression analyses were performed to assess its<br />

relationship to other variables.<br />

Results:<br />

Twenty patients completed the survey during the first week of<br />

radiotherapy. Median age was 57 (range, 31-81). Five patients (25%)<br />

had Stage 0 disease, 9 (45%) Stage I, 0 (0%) Stage II, 5 (25%) Stage<br />

III, and 1 (5%) Stage IV. Seven patients (35%) received chemotherapy<br />

while 13 (65%) did not. Eleven patients (55%) had significant others<br />

while undergoing treatment and 9 (45%) were single. The PCF<br />

analysis confirmed that the items of the experimental questionnaire<br />

were internally consistent (α = .88), such that all were retained in<br />

the final scale for subsequent analyses. The extent to which patients<br />

saw cancer as self defining was statistically significantly associated<br />

with their reported levels of depression (r = .60, p < .005) and<br />

anxiety (r = .60, p < .005). There was a similar trend with reported<br />

levels of insomnia which was not statistically significant (p=0.2).<br />

Of the demographic and clinical variables, only stage of cancer was<br />

associated with depression and anxiety.<br />

Conclusions:<br />

Patients undergoing treatment for breast cancer who view the cancer<br />

diagnosis as self-defining are more likely to self-report depression,<br />

anxiety, and insomnia. In this cohort, the experimental questionnaire<br />

demonstrated strong internal validity. An intervention that alters<br />

patients’ perception of identity as it relates to the cancer diagnosis<br />

could be a promising compliment to the standard treatment of<br />

depression, anxiety, and insomnia.<br />

P6-09-01<br />

Investigating the effectiveness of a psycho-educational behavioral<br />

intervention for cancer-related cognitive dysfunction in women<br />

with breast and gynecological cancer: Knowledge, self-efficacy,<br />

and behavioral change.<br />

Bernstein LJ, Dissanayake D, Tirona KM, Nyhof-Young JM, Catton<br />

PA. Princess Margaret Hospital, Toronto, ON, Canada; University<br />

of Toronto, ON, Canada<br />

Background: Approximately one third of individuals receiving<br />

chemotherapy for breast cancer experience cancer-related cognitive<br />

dysfunction (CRCD). CRCD significantly impacts quality of life,<br />

employment, and social relationships, and can hinder ability to<br />

return to pre-cancer activities. The cause is likely multi-factorial,<br />

and currently no medical treatment exists. The <strong>Cancer</strong> Survivorship<br />

Program at Princess Margaret Hospital provides a behavioralintervention<br />

for patients with CRCD. The single, one-on-one, hour<br />

www.aacrjournals.org 537s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

long session provides information and compensatory strategies to<br />

improve knowledge, self-management skills, and decrease the impact<br />

of cognitive dysfunction on daily life.<br />

Methods: A mixed-method study assessed patient perceptions.<br />

Eligible women had been or were currently being treated for a<br />

diagnosis of breast or gynecological cancer and were English<br />

speakers. Likert-scale based questionnaires were administered<br />

immediately before, immediately after, and 6 weeks post-intervention.<br />

Six parameters were evaluated quantitatively: i) program delivery, ii)<br />

perception of knowledge, iii) distress, iv) self-efficacy, v) behavior<br />

change, and vi) behavioral change effectiveness. Ten individuals also<br />

participated in qualitative, semi-structured audio-recorded interviews<br />

1-6 weeks post questionnaire completion. Responses were analyzed<br />

using the constant comparative method.<br />

Results: 72 female cancer survivors (61 breast; 11 gynecological;<br />

mean age 51.3) completed questionnaires. Immediately postintervention,<br />

patients reported significant increases in perception of<br />

knowledge, self-efficacy, and decreases in CRCD-related distress, and<br />

these improvements were maintained at 6 week follow up (p < .01 in<br />

all cases). Participants reported numerous benefits associated with<br />

increased knowledge, including a sense of validation and reassurance,<br />

and increased confidence and success in managing their symptoms.<br />

No statistically significant differences in intervention effectiveness<br />

were observed as a function of patient age (older or younger than<br />

50), education (greater or less than grade 12), or employment status<br />

(skilled worker or higher vs lower). However, individuals of low<br />

employment status were less satisfied with the volume of information<br />

in the intervention. Interviews indicated their preference for an<br />

additional session to reinforce the techniques discussed. Thematic<br />

analysis also indicated that adoption of new compensatory strategies<br />

was associated with a sense of control, confidence and pride for all<br />

patients; although barriers were identified for some that prevented<br />

long-term use (e.g. techniques take time and effort). Employment<br />

distress was mainly tied to difficulties with work tasks.<br />

Conclusions: A one-hour, psycho-educational behavioral intervention<br />

can be effective for breast and gynecological cancer patients suffering<br />

from CRCD. Benefits can last at least 6 weeks. Results suggest a need<br />

for better intervention integration with oncology appointments, a<br />

follow-up session to help individuals assimilate CRCD information,<br />

and increased session personalization to address individual difficulties<br />

and job concerns.<br />

P6-09-02<br />

Pre-treatment cognitive function (CF) in women with locally<br />

advanced breast cancer (LABC) and in healthy controls.<br />

Bernstein LJ, Seruga B, Pond G, Tirona KM, Dodd A, Tannock<br />

IF. Princess Margaret Hospital, Toronto, ON, Canada; University<br />

of Toronto; Institute of Oncology Ljubljana, Slovenia; McMaster<br />

University, Hamilton, ON, Canada<br />

Background: Several studies have reported that women with breast<br />

cancer may have cognitive dysfunction prior to chemotherapy<br />

(ChT), and that cognitive dysfunction may be related to the cancer<br />

itself or to the psychological impact of a cancer diagnosis. Previous<br />

studies assessing CF in breast cancer patients pre-ChT have done<br />

so after surgery. Women with LABC receive ChT prior to surgery,<br />

and therefore provide a unique opportunity to assess CF without the<br />

confounding factor of surgery. This study reports CF in women with<br />

newly diagnosed LABC compared to healthy controls.<br />

Methods: Baseline information including participant demographics,<br />

smoking/alcohol history, co-morbidities, and medications were<br />

collected. Women with LABC underwent a 2-hour battery of CF tests<br />

prior to neoadjuvant ChT. Six cognitive domains using standardized,<br />

validated, and reliable neuropsychological tests were assessed: verbal,<br />

visual-spatial, memory, attention, processing speed, and executive<br />

function. Performance on each test was transformed to z-scores<br />

using normative data (average range z-score = -1 to 1). Participants<br />

also completed self-report questionnaires for CF (FACT-COG3<br />

and PAOFI), fatigue (FACIT-F subscale) and affect (HADS). Data<br />

obtained were compared to those for age-matched healthy women<br />

who were administered the same battery of tests and measures.<br />

Results: Forty-seven women with LABC and 22 controls were<br />

assessed. All women completed the CF tests and questionnaires<br />

within 2 hours. There were no significant differences between patients<br />

and controls in age (pts median age=49; controls=48), education<br />

(median years in school=16 for both), smoking or alcohol history,<br />

co-morbidities, mood disorders (anxiety, depression), previous history<br />

of concussion, or current medication for mood or sleep problems.<br />

Objective CF testing demonstrated no significant difference in the<br />

mean performance of women with LABC vs that of controls in any<br />

objective domain, nor were there statistically significant differences<br />

in the frequency of deficits (e.g., 96% of pts had < 2 deficits; 86%<br />

of controls had < 2 deficits). Women with LABC did not report<br />

worse CF, fatigue, or depressive symptoms compared to controls.<br />

Anxiety was the only self-reported measure where patients had more<br />

symptoms (mean HADS 7.6 vs 5.4, p < .036). Consistent with the<br />

literature, only weak to no association was observed between any<br />

of the objective measures of CF and the self-reported measures.<br />

Multivariate regression analyses found for both patients and controls,<br />

low education level (≤12 years), smoking history (≥10 pack year),<br />

alcohol intake (≥10 drinks/week), and currently taking medication<br />

for anxiety, depression, or sleep to be associated with poorer CF as<br />

measured on at least one objective domain.<br />

Conclusion: When assessed prior to ChT and surgery, no significant<br />

difference in objectively assessed or self-reported CF was observed<br />

between women with LABC and healthy controls. The effects of<br />

cancer treatment will be evaluated as part of a longitudinal study.<br />

P6-09-03<br />

Fatigue after breast cancer may be related to conditions other<br />

than the cancer. The impact of comorbidity is essential.<br />

Reidunsdatter RJ, Hjermstad M, Oldervoll L, Lundgren S. HiST/<br />

NTNU, Trondheim, Norway; HIST, Trondheim, Norway; NTNU,<br />

Trondheim, Norway; Oslo University Hospital, Oslo, Norway;<br />

Røros Rehabilitation, Røros, Norway; St. Olav University Hospital,<br />

Trondheim, Norway<br />

Purpose: Fatigue after treatment for breast cancer is common, but is<br />

also prevalent in people with chronic diseases such as heart failure,<br />

diabetes, and depression etc. The primary objective was to compare<br />

the level of fatigue between BC patients one year after end of<br />

treatment with data from a representative survey of the Norwegian<br />

population (GenPop). Secondary aim was to explore the association<br />

between chronic conditions and fatigue and in both samples.<br />

Design and Method: 245 patients treated with chemo –and/or<br />

radiotherapy after surgery, were assessed one year after treatment.<br />

Comorbidity was recorded by clinical examinations in patients and<br />

by self-report in the GenPop (N=652). Fatigue was measured by the<br />

3-item subscale of the EORTC QLQ-C30, with higher scores on the<br />

0-100 scale implying more fatigue. Analysis of covariance was applied<br />

to compare age-adjusted mean scores between groups.<br />

Results: Mean age was 58 (9) years in patients and 52 (14) years<br />

in GenPop. 23% of patients and 32% of GenPop had one or more<br />

of the following conditions; cardiovascular- or pulmonary disease,<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

538s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: General Session 6<br />

diabetes or depression. No significant differences were found in<br />

fatigue between BC patients and GenPop (mean score 26.7 vs.<br />

29.7). Comorbidity was the greatest determinant of increased<br />

fatigue, regardless of BC treatment. Patients with comorbidity were<br />

significantly more fatigued than those without comorbidity (mean<br />

difference 10.4), as found in the GenPop (mean difference 13.3).<br />

Conclusion: Similar fatigue levels in BC patients and GenPop one<br />

year after treatment is promising, but longer follow-up is needed.<br />

Comorbidity conditions should always be assessed when evaluating<br />

fatigue and other patient related outcomes. Prevention or treatment<br />

of common chronic conditions might be considered in rehabilitation<br />

programs to reduce fatigue in cancer survivors.<br />

P6-09-04<br />

The Association of Low Level Arm Volume Increases with<br />

Lymphedema Symptoms Following Treatment for <strong>Breast</strong> <strong>Cancer</strong><br />

Skolny MN, Miller CL, Shenouda M, Jammallo LS, O’Toole J,<br />

Niemierko A, Taghian AG. Massachusetts General Hospital, Boston,<br />

MA<br />

Purpose/Objective: The symptoms associated with breast cancerrelated<br />

lymphedema are well-documented, and include sensations<br />

of heaviness, swelling, and tightness in the upper extremity and<br />

trunk. However, the clinical significance of low-level arm volume<br />

changes frequently experienced by breast cancer patients is not well<br />

understood. We sought to determine the association of low level arm<br />

volume changes with patient-reported lymphedema symptoms in<br />

women treated for breast cancer.<br />

Methods: 267 patients who underwent surgical treatment for breast<br />

cancer from 2010- 2012 were identified from a cohort of patients<br />

prospectively screened for lymphedema at our institution. Patients<br />

were assessed with perometer arm volume measurements and a<br />

survey of lymphedema symptoms pre and post operatively, and at<br />

3-7 month intervals thereafter. Inclusion in this analysis was limited<br />

to unilaterally affected women with ≥ 3 assessments and ≥ 6 months<br />

of post-surgical follow-up. Arm volume changes were quantified as<br />

Relative Volume Change (RVC): RVC= (A2*U1)/ (U2*A1) - 1, where<br />

A1 is pre-operative arm volume and A2 is post-operative arm volume<br />

on the affected side, and U1 and U2 are arm volumes on the unaffected<br />

side at these time points. Low level arm volume change was defined as<br />

a measurement with RVC ≥ 5% -


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P6-09-06<br />

Family Members’ Burden in Patients with Metastatic and Early<br />

Stage <strong>Breast</strong> <strong>Cancer</strong><br />

Wan Y, Gao X, Mehta S, Zhang Z, Faria C, Schwartzberg L. Pharmerit<br />

North America; Eisai, Inc.; The West Clinic<br />

BACKROUND: No previous studies have assessed the indirect<br />

costs attributable to family members of patients with metastatic<br />

breast cancer (MBC). This retrospective database study compared<br />

productivity loss and cost associated with family members of patients<br />

with MBC vs. early-stage breast cancer (EBC).<br />

METHODS: The Thomson Reuters MarketScan Health and<br />

Productivity Management (HPM) data (2005-2009) were used. MBC<br />

and EBC patients with no other tumors and 12-month continuous<br />

enrollment in medical insurance after the index diagnosis date were<br />

identified. Adult working family members of these MBC/EBC patients<br />

were identified based on their enrollee ID numbers since the enrollees<br />

from the same family were assigned the same ID except for the last<br />

two digits. The patients’ working family members who were eligible<br />

for absenteeism and did not have any cancer diagnosis were included<br />

and used as a proxy for “caregivers”. The reasons for absenteeism<br />

included sickness, personal leave, disability, recreation, Family and<br />

Medical Leave Act (FMLA) and others. The productivity loss was<br />

measured as the leave days taken under FMLA or personal leave by<br />

the family members during a 12-month follow-up period. Associated<br />

indirect costs were estimated by multiplying leave days with daily<br />

wages obtained from the 2011 Bureau of Labor Statistics. Descriptive<br />

analyses and a generalized linear model with log link and gamma<br />

distribution were conducted.<br />

RESULTS: A total of 209 MBC/1,166 EBC patients’ family members<br />

were eligible for absenteeism, with a mean age of 50/51 years. 1,373<br />

out of the 1,375 family members were male. Majority of the family<br />

members (52%) worked in manufacturing industry, followed by<br />

transportation/communication/utilities (28%) and oil gas extraction/<br />

mining industry (15%). 52% of the family members had PPO health<br />

insurance. Family members of MBC and EBC patients had similar<br />

clinical and demographic characteristics. Similar proportions of<br />

MBC and EBC patients’ family members had overall and individual<br />

type of absenteeism. However, MBC patients’ family members had<br />

more FMLA/personal leave days and higher costs vs. EBC patients<br />

(overall mean: 2.8±6.1 vs. 2.1±4.8; $2,366±5,095 vs. $1,741±3,995,<br />

both p=.047). Among those who had non-zero FMLA/personal<br />

leave days (MBC vs. BC: N=92/44% vs. 502/43%), mean FMLA/<br />

personal leave days and costs were 6.4±7.8 vs. 4.8±6.3; $5,375±6,556<br />

vs. $4,043±5,272 respectively, both p=.033. After adjusting for<br />

covariates, MBC patients’ family members incurred 40% (p=.06)<br />

more absenteeism-related costs due to FMLA/personal leave vs.<br />

EBC patients’ family members. Additionally, occupation in oil/gas<br />

extraction/mining (vs. manufacturing non-durable goods industry,<br />

p


December 4-8, 2012 <strong>Abstract</strong>s: General Session 6<br />

P6-09-08<br />

COMPliance and Arthralgia in Clinical Therapy: The COMPACT<br />

trial, assessing the incidence of arthralgia, therapy costs and<br />

compliance within the first year of adjuvant anastrozole therapy<br />

Harbeck N, Blettner M, Bolten WW, Hindenburg H-J, Jackisch<br />

C, Klein P, König K, Kreienberg R, Rief W, Wallwiener D, Zaun<br />

S, Hadji P. University of Munich, Germany; University of Mainz,<br />

Germany; Klaus-Miehlke-Hospital for Rheumatology, Wiesbaden,<br />

Germany; Head of BNGO e.V., Germany; Hospital for Gynecology<br />

and Obstetrics, Offenbach, Germany; d.s.h. Statistical Services,<br />

Rohrbach, Germany; Gynecological Society Germany e.V., Germany;<br />

University Women’s Hospital, Ulm, Germany; Outpatient Clinic for<br />

Psychotherapy, Philipps-University, Marburg, Germany; University<br />

Women’s Hospital, Tübingen, Germany; AstraZeneca GmbH, Wedel,<br />

Germany; Women’s Hospital Phillips-University, Marburg, Germany<br />

Background: Aromatase Inhibitors (AI) are well established as<br />

adjuvant treatment for postmenopausal (PMP) women with hormone<br />

receptor positive (HR+) early breast cancer (EBC). However, phase<br />

III clinical trials have reported higher rates of arthralgia associated<br />

with AI than tamoxifen. This study aims to collect real world data on<br />

the effects of AI-associated arthralgia on patient compliance, patient<br />

outcomes and on treatment of arthralgia.<br />

Methods: COMPACT is an open, prospective, non-interventional,<br />

non-randomized study (NCT00857012) run in Germany. PMP women<br />

with HR+ EBC who had been on adjuvant anastrozole (ANA) for<br />

3-6 months were enrolled and stratified by initial adjuvant ANA or<br />

switch from tamoxifen. All patients received regular standardized<br />

information about breast cancer from baseline to week 20 to support<br />

treatment compliance. Data on demographics, arthralgia, related<br />

therapies, other side effects and QoL were collected at baseline,<br />

3, 6 and 9 months. Primary endpoints are scaled data on arthralgia<br />

and compliance within the first year of ANA therapy. Secondary<br />

endpoints include incidence of arthralgia, therapy costs, reasons for<br />

non-compliance, and influence of arthralgia on clinical outcome.<br />

Results: Between Apr 2009 and Mar 2011, 2313 patients were<br />

enrolled, 2007 on upfront ANA and 306 on switch from tamoxifen.<br />

The mean age at baseline was 64.5 years, mean BMI 27.7. Only<br />

17.0% of patients had received HRT prior to their EBC. At baseline,<br />

41.9% reported symptoms relating to skeleton or musculature. 12.0%<br />

reported arthralgia existing prior to ANA treatment and 13.2% stated<br />

a worsening of pre-existing arthralgia or new arthralgia after starting<br />

ANA. Predictors for non-adherence to AI therapy were former nonadherence,<br />

general symptom load on the side effect scale GASE,<br />

and low benefit expectation at treatment start. Risk of arthralgia was<br />

related to BMI (lowest for patients with BMI ≤24.1 kg/m², highest<br />

with BMI >30.5 kg/m² at all time points; OR>1) and upfront therapy<br />

(switch patients had a reduced risk of 68% at 6 and 61% at 9 months<br />

compared to patients with ANA upfront, p=0.002). Patients with<br />

prior chemotherapy had lower rates of arthralgia before start of ANA<br />

(10.4% vs 13.3%, p=0.036) but higher rates after the start of ANA and<br />

before study start (27.0% vs 22.5%, p=0.013). Patients with arthralgia<br />

showed higher compliance rates at all time points (p


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P6-09-10<br />

Informational needs among women considering breast<br />

reconstruction post-mastectomy.<br />

Ahmed I, Harvey A, Amsellem M. <strong>Cancer</strong> Support Community,<br />

Washington, DC<br />

For many women, the complexity of processing and learning about<br />

their breast cancer diagnosis is further complicated by decisions to<br />

be made about breast reconstruction post-mastectomy. For women<br />

considering reconstruction post-mastectomy, obtaining information<br />

about reconstruction procedures and outcomes is a process, and<br />

multiple factors may influence the decision-making process. Little is<br />

known about the extent to which patient’s knowledge and expectations<br />

related to breast reconstruction procedures weigh on their decisionmaking<br />

process.<br />

To better understand and meet the needs of women facing<br />

reconstruction, the <strong>Cancer</strong> Support Community surveyed 1,185<br />

women diagnosed with breast cancer. Women answered questions<br />

either: online (n=840) in August, 2010; or via paper and pencil (n=<br />

345) after attending a breast reconstruction workshop held at multiple<br />

sites nationwide in 2011. In addition to demographics, and diagnostic<br />

and treatment history, women reported their decision regarding<br />

reconstruction, experience searching for and receiving information,<br />

and expectations about reconstruction.<br />

Respondents were primarily Caucasian (81.9%), and the mean age at<br />

diagnosis was 48.9. 21.2% of respondents were first diagnosed with<br />

breast cancer in the past year, and 30.0% were diagnosed at least five<br />

years ago. Online respondents were more likely to be Caucasian, less<br />

likely to have been recently (less than one year) diagnosed with breast<br />

cancer, and more likely to have already made a decision regarding<br />

reconstruction, as compared to workshop attendees, but were similar<br />

in all other respects for these analyses.<br />

Most respondents (57.3%) had either undergone or had decided to<br />

undergo breast reconstruction. 18.4% of respondents reported they<br />

decided not to undergo reconstruction, and 8.9% reported they were<br />

not eligible. Additionally, 15.5% of respondents were currently<br />

considering or planning to consider their options for reconstruction<br />

at a later time.<br />

Of the women who were currently considering or planning to consider<br />

breast reconstruction, information-seeking patterns and informational<br />

needs were reported. Aside from their health care team, most women<br />

sought additional information about reconstruction from other women<br />

with breast cancer (40.9%), medical literature (46.0%), patient support<br />

groups (35.6%), friends and family (41.3%), and the Internet (57.2%).<br />

Over one-third of respondents considering reconstruction (34.5%)<br />

would have liked to have had more information about the risks and<br />

benefits of reconstruction at the time of mastectomy versus at a later<br />

time. These women also reported a number of factors influencing their<br />

current point in the decision-making process, including: concerns<br />

about implant safety (83.3%); concerns about additional treatment<br />

and recovery (85.5%); concerns about failed procedures (91.5%); and<br />

confusion about the reconstruction decision-making process (72.2%).<br />

Survey responses suggest women considering breast reconstruction<br />

have a variety of informational needs, some of which are not being<br />

met. Results suggest there is still work to be done with regard to<br />

establishing realistic expectations about the procedures and outcomes,<br />

providing comprehensive information at various stages throughout<br />

the process, and across the various choices.<br />

P6-09-11<br />

Examining patient treatment choices involving efficacy, toxicity,<br />

and cost tradeoffs in the metastatic breast cancer setting<br />

White CB, Smith ML, Abidoye O, Lalla D. Carol B. White &<br />

Associates; Research Advocacy Network; Genentech, Inc.<br />

Background: Most patients with metastatic breast cancer are treated<br />

with chemotherapies and/or targeted therapies. These therapies have<br />

toxicity profiles that vary with agent(s) used. Patient attitudes towards<br />

different adverse events (AE’s) may vary and factor into treatment<br />

decisions. Different patients may have specific feelings about tolerable<br />

and unacceptable AE’s, especially when balanced against possible<br />

treatment benefit. As more agents/combinations become available, it<br />

becomes increasingly important to understand which adverse events<br />

impact treatment decisions. Previous research has shown that conjoint<br />

analysis (CA) is a valid methodology that allows patients to express<br />

preferences and is particularly useful when designed based on specific<br />

treatment profiles (Smith, ASCO 2011; Smith, ASCO 2012).<br />

Methods: The objective of this study was to assess patient<br />

preferences using CA based on profiles of two MBC regimens<br />

(trastuzumab+docetaxel and T-DM1). Patients were presented pairs<br />

of hypothetical treatments (describing benefit, AE’s, and cost) and<br />

asked what their preferred alternative was; a follow-up question asked<br />

if they would take the treatment if it were the only option available.<br />

Five AE’s (alopecia, peripheral neuropathy, diarrhea, fatigue, and<br />

neutropenia) with differing likelihood, severity, and/or duration were<br />

included. There were 3 stages in preparing the CA survey: the first<br />

comprised two online focus groups conducted with patients with<br />

metastatic disease. Stage 2 included the development of the CA<br />

survey using patient language to describe the AE’s and their impact,<br />

as well as images to represent likelihoods, progression-free survival<br />

(PFS), and costs. Stage 3 is initiating and will recruit patients with<br />

the assistance of several breast cancer organizations (target n= 600).<br />

Analysis of response patterns allows study of the influence of each<br />

variable and provides a basis for prediction of treatment choice for<br />

any combination of benefit, AE’s, and cost. Final analysis will be<br />

complete in September 2012.<br />

Results: Findings from the focus groups facilitated an understanding<br />

of PFS, of experience with and impact of the AE’s on decisionmaking,<br />

and of attitudes. In Stage 2, the survey was pretested with<br />

seven patients and took approximately 20 minutes to complete.<br />

Feedback suggested the questions were relevant and realistic.<br />

Suggestions allowed for improvement of the CA explanatory<br />

material, as well as refinement of a few answer choices to questions<br />

outside the CA section of the survey. Final study results will present<br />

the proportion of patients who are predicted to prefer each of two<br />

treatment profiles, the impact of each attribute level on treatment<br />

preference and differing preferences seen in patient subgroups. This<br />

information will provide valuable insight into patient preferences and<br />

inform future development of new therapies. In addition, these results<br />

may generate discussion and consideration of patient preferences in<br />

conversations about patient care and treatment selection.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

542s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: General Session 6<br />

P6-10-01<br />

A randomized phase 2 study of the antibody-drug conjugate<br />

CDX-011 in advanced GPNMB-overexpressing breast cancer:<br />

The EMERGE study<br />

Yardley DA, Weaver R, Melisko ME, Saleh MN, Arena FP, Forero<br />

A, Cigler T, Stopeck A, Citron D, Oliff I, Bechhold R, Loutfi R,<br />

Garcia A, Crowley E, Green J, Yellin MJ, Davis TA, Vahdat LT.<br />

Florida <strong>Cancer</strong> Specialists, Tampa, FL; University of California,<br />

<strong>San</strong> Francisco Helen Diller Family Comprehensive <strong>Cancer</strong> Center,<br />

<strong>San</strong> Francisco, CA; Sarah Cannon Research Institute/Tennessee<br />

Oncology, PLLC, Nashville, TN; Georgia <strong>Cancer</strong> Specialists PC,<br />

<strong>San</strong>dy Springs, GA; Arena Onc Assoc PC, Lake Success, NY; Bium<br />

Inc, Birmingham, AL; Weill Cornell Medical College, New York, NY;<br />

Celldex Therapeutics, Inc., Needham, MA; Arizona <strong>Cancer</strong> Center<br />

at the University of Arizona, Tucson, AZ; <strong>Cancer</strong> Treatment Centers<br />

of America/Midwestern Regional Medical Center, Zion, IL; Orchard<br />

Healthcare Research Inc., Skokie, IL; Oncology Hematology Care,<br />

Cincinnati, OH; Henry Ford Health System, Detroit, MI; USC/Norris<br />

Comprehensive <strong>Cancer</strong> Center, Los Angeles, CA<br />

Background: GPNMB is an internalizable transmembrane<br />

glycoprotein overexpressed in multiple tumor types, including breast<br />

cancer (BC). GPNMB promotes BC metastases in a murine model<br />

and is a poor prognostic marker in patients (pts) (Rose 2010). CDX-<br />

011 (glembatumumab vedotin) is a fully human GPNMB-specific<br />

monoclonal antibody conjugated to the potent cellular toxin MMAE<br />

via a protease-sensitive peptide linker, designed to cleave upon<br />

cellular internalization. In Phase I/II studies of CDX-011 in BC<br />

(n=42), an objective response rate (ORR) (including confirmed and<br />

unconfirmed response) of 12% overall and 20% for “triple-negative”<br />

(ER/PR/HER2-; TN) pts was observed (Saleh, ASCO 2010).<br />

Methods: The Phase II EMERGE study examined the efficacy of<br />

CDX-011 by level of GPNMB expression. Refractory BC (2-7 prior<br />

cytotoxic regimens in metastatic setting), GPNMB expression in ≥ 5%<br />

of tumor or stroma cells (as centrally determined on archival tumor),<br />

and no persistent treatment-related toxicity (including neuropathy) of<br />

≥ Grade 2 severity were required for enrollment. Pts were stratified<br />

by GPNMB expression pattern (any tumor, low stromal or high<br />

stromal) and randomized 2:1 to CDX-011 (1.88 mg/kg IV q 21 days)<br />

or “investigator’s choice” (IC) single-agent chemotherapy, and treated<br />

until progression (PD) or intolerance with IC pt crossover permitted to<br />

CDX-011 at PD. Endpoints: ORR (primary), progression-free survival<br />

(PFS), safety, pharmacokinetics, pharmacodynamics.<br />

Results: 99% of screened tumor samples expressed GPNMB. 122<br />

treated pts (81 CDX-011 vs. 41 IC) had median age = 56 vs. 56 years;<br />

median # prior anticancer regimens = 6 vs. 5; visceral disease (liver/<br />

lung) = 83% vs. 80%; TN = 33% vs 27%, respectively. To date, 15<br />

IC pts have crossed over to CDX-011, and 14 pts (8 CDX-011, 6<br />

IC) continue on treatment. CDX-011 was well tolerated with less<br />

hematologic toxicity than IC; CDX-011-related toxicity included<br />

rash (overall = 38%; Grade 1 = 20%; Grade 2 = 15%; Grade 3 =<br />

3%) and neuropathy (overall = 18%; Grade 1 = 8%; Grade 2 = 7%;<br />

Grade 3 = 2%).<br />

ORR<br />

Median PFS (months)<br />

CDX-011 IC CDX-011 IC p-value<br />

All pts 19% (15/81) 14% (5/36) 2.1 1.7 0.6763<br />

TN 21% (5/24) 0% (0/9) 2.3 1.6 0.4071<br />

hGPNMB 32% (8/25) 13% (1/8) 2.5 1.4 0.0996<br />

TN & hGPNMB 36% (4/11) 0% (0/3) 3.0 1.3 0.0032<br />

hGPNMB = high tumor GPNMB expression (in ≥25% of tumor cells). CDX-011 includes 15 pts<br />

who crossed over after PD on IC. ORR analysis includes confirmed and unconfirmed response and<br />

excludes pts who discontinued study without evaluable post-baseline imaging (n=15 for CDX-011;<br />

n=5 for IC).<br />

Conclusions: CDX-011 was a well-tolerated and active agent in this<br />

heavily pre-treated BC pt population. Preliminary analysis of the<br />

EMERGE study demonstrates enhanced activity of CDX-011 in TN<br />

as well as high GPNMB expressing BC tumors, confirming that CDX-<br />

011 is a targeted agent. This noted promising activity in advanced TN<br />

and hGPNMB enriched pt populations warrants further evaluation.<br />

P6-10-02<br />

MLN8237 (alisertib), an investigational Aurora A Kinase<br />

inhibitor, in patients with breast cancer: Emerging phase 2 results<br />

Alvarez RH, DeMichele A, Mailliez A, Benaim E, Fingert H,<br />

Schusterbauer C, Zhang B, Melichar B. The University of Texas<br />

MD Anderson <strong>Cancer</strong> Center, Houston, TX; Abramson <strong>Cancer</strong><br />

Center, Philadelphia, PA; Centre Oscar Lambret, Lille, Cedex 59,<br />

France; Millennium Pharmaceuticals, Inc., Cambridge, MA; Fakultní<br />

nemocnice Olomouc - Onkologická klinika, Olomouc, Czech Republic<br />

Background: Aurora A Kinase (AAK) is a key mitotic regulator<br />

amplified or overexpressed in a variety of tumor types including preinvasive<br />

and invasive breast cancer. In mouse mammary epithelium,<br />

AAK overexpression induces genetic instability and tumor formation.<br />

In breast cancer patients (pts), AAK overexpression is often associated<br />

with poor prognosis. Therefore, AAK is an attractive target in cancer<br />

treatment. MLN8237 is an oral, selective AAK inhibitor under<br />

evaluation in pts with different advanced tumors. An ongoing multiindication<br />

phase 2 (NCT01045421) trial evaluates MLN8237 in pts<br />

with solid tumors; here we present preliminary phase 2 data on the<br />

breast cancer cohort.<br />

Methods: Eligible pts were female ≥18 years of age with stage IV<br />

breast cancer, ECOG PS 0–1, measurable disease by RECIST, and<br />

≤4 prior cytotoxic chemotherapy regimens. Stratification on receptor<br />

subtype was performed to ensure 8–10 pts with triple negative disease<br />

and 8–10 patients with HER2 positive disease. Oral MLN8237 50 mg<br />

was administered twice-daily for 7 days, followed by 14 days’ rest<br />

in 21-day cycles. The primary objective was overall response rate;<br />

response was determined by RECIST v1.1. Secondary objectives<br />

included assessment of safety. A Simon’s optimal 2-stage design was<br />

used; 20 pts were initially enrolled, and this cohort was expanded if<br />

≥2 partial responses (PR) were observed in response-evaluable pts.<br />

Results: In the first 20 pts, 2 had PR and the cohort was expanded<br />

to a total of 53 (data cut-off: March 29, 2012). Median age was 60<br />

years (range 33–81). Pts received a median of 3 cycles (range 1-14)<br />

and 20 pts (38%) received ≥6 treatment cycles. Of 45 responseevaluable<br />

pts, 6 pts (13%) had a best response of PR and 26 (58%)<br />

had stable disease (SD). Median duration of SD was 68 days, of which<br />

2 pts (4%) had SD for ≥6 months (clinical benefit rate [PR+SD ≥6<br />

months] =18%). One HER2+ patient with 2 prior lines of therapy<br />

(carboplatin/docetaxel/trastuzumab + trastuzumab maintenance;<br />

gemcitabine/lapatinib/ trastuzumab) had PR 6 months. Fifty-three pts<br />

were evaluable for toxicity. Drug-related adverse events (AEs) were<br />

reported in 49 pts (92%) and were commonly grade 1–2 in severity.<br />

The most frequent drug-related AEs included alopecia (n=26, 49%),<br />

neutropenia (n=25, 47%), diarrhea (n=23, 43%), fatigue (n=21, 40%),<br />

and stomatitis (n=19, 36%). A total of 34 pts (64%) reported grade ≥3<br />

drug-related AEs, including neutropenia (n=23, 43%) and leukopenia<br />

(n=8, 15%). Two pts (4%) discontinued due to AEs; there were no<br />

on-study deaths. Treatment is ongoing in 21 pts.<br />

Conclusions: Emerging phase 2 data reported here provide<br />

preliminary evidence of antitumor activity of oral MLN8237 in<br />

pts with relapsed/refractory breast cancer. AEs were frequent but<br />

generally reversible and manageable by dose reduction and supportive<br />

care.<br />

www.aacrjournals.org 543s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

P6-10-03<br />

The PKC inhibitor PKC412 antagonizes breast cancer cell growth<br />

and enhances tamoxifen sensitivity<br />

Shou J, Chew SA, Mitsiades N, Kumar V, Fu X, Chamness G, Osborne<br />

K, Schiff R. Lester & Sue Smith <strong>Breast</strong> Center, Baylor College of<br />

Medicine, Houston, TX; Baylor College of Medicine, Houston,<br />

TX; Dan L Duncan <strong>Cancer</strong> Center, Baylor College of Medicine,<br />

Houston, TX<br />

Background: AIB1 (SRC-3, NCoA3), a member of the p160/steroid<br />

receptor coactivators family, plays a critical role in cell growth and<br />

proliferation. In estrogen receptor-alpha positive (ER+) breast cancer<br />

(BC) cells, it coactivates estrogen- and additional transcription<br />

factors-dependent gene transcription, reducing the antagonistic<br />

activity of tamoxifen and resulting in tamoxifen resistance (TR). We<br />

have previously shown that BC patients whose tumors expressed high<br />

levels of both AIB1 and HER-2 had worse outcomes with tamoxifen<br />

therapy, suggesting that AIB1 may be an important diagnostic and<br />

therapeutic target. Our findings that knocking down AIB1 attenuates<br />

ER signaling and inhibits breast cancer cell growth further indicate<br />

that the manipulation of AIB1 level could be an approach to treating<br />

BC and overcoming TR. Recently, it has been shown that protein<br />

kinase C (PKC) isoforms phosphorylate AIB1 and prevent its<br />

proteasome-mediated degradation. The present study was carried<br />

out to test if the multi-targeted kinase and PKC inhibitor PKC412<br />

(midostaurin) is capable of promoting degradation of AIB1, inhibiting<br />

BC cell growth, and promoting tamoxifen antagonistic activity.<br />

Methods: The ER+ MCF7, T47D, and ZR75-B BC cells and their<br />

tamoxifen-resistant derivatives (TR) were used. The Methylene<br />

Blue assay was employed to measure cell viability of BC cell lines<br />

after treatment with PKC412 or the combination of PKC412 with<br />

tamoxifen. To determine the impact of PKC412 on AIB1 and ER<br />

proteins and mRNA levels, we used immunoblotting and RT-qPCR,<br />

respectively.<br />

Results: PKC412 successfully inhibited the growth of MCF7,<br />

T47D, and ZR75-B cells, and their tamoxifen-resistant derivatives.<br />

Treatment with PKC412 depleted AIB1 protein and reduced the<br />

level of ER protein without significant alteration in AIB1 mRNA<br />

level. Consequently, ER signaling was disrupted, as reflected by<br />

decreased expression of ER target genes such as GREB1. PKC412<br />

also enhanced tamoxifen’s antagonistic activity in the parental cell<br />

lines and sensitized tamoxifen-resistant MCF7 and ZR75-B cells to<br />

tamoxifen.<br />

Conclusions: The results of this study suggest that the multi-targeted<br />

kinase and PKC inhibitor PKC412 can post-translationally destabilize<br />

AIB1 protein and ER, inhibiting BC cell growth and viability.<br />

PKC412 enhances tamoxifen’s antagonistic activity on BC growth;<br />

furthermore, it sensitizes tamoxifen-resistant cells to tamoxifen. Thus,<br />

PKC412 is a promising agent to treat BC and, in combination with<br />

tamoxifen, to delay or overcome TR.<br />

P6-10-04<br />

The Presence of Anaplastic Lymphoma Kinase Recapitulates<br />

Formation of <strong>Breast</strong> Tumor Emboli with Encircling<br />

Lymphovasculogenesis<br />

Liu H, Chu K, Ochoa AE, Ye Z, Zhang X, Jin J, Wright MC, Barsky<br />

SH, Cristofanilli M, Robertson FM. The University of Texas MD<br />

Anderson <strong>Cancer</strong> Center, Houston, TX; University of Nevada School<br />

of Medicine, Reno, NV; Fox Chase <strong>Cancer</strong> Center, Philadelphia, PA<br />

Background: Genetic abnormalities in the anaplastic lymphoma<br />

kinase (ALK) gene result in activation of signaling pathways<br />

including Akt, mTor, and JAK/Stat3. ALK has been shown to be a<br />

primary oncogenic driver in a variety of human tumors, including both<br />

hematologic malignancies such as anaplastic large cell lymphoma,<br />

as well as solid tumors including neuroblastoma, non-small cell lung<br />

cancer, myofibroblastic tumors and most recently, high grade serous<br />

ovarian carcinoma. While only ∼3% of all breast cancers have been<br />

reported to have ALK genetic abnormalities, our studies revealed that<br />

inflammatory breast cancer (IBC), the most lethal variant of breast<br />

cancer, is characterized by prevalent ALK gene amplification with<br />

activated ALK signaling. The present studies investigated the role<br />

of ALK in breast cancer by expressing full-length wild type ALK<br />

in MCF-7 cells.<br />

Materials and Methods: Clones of MCF-7 breast cancer cells<br />

expressing wild type ALK or non-target vector were produced by<br />

lentivirus infection and selection of single cell clones. MCF-7 ALK<br />

clones were evaluated using live cell and phase contrast imaging,<br />

immunofluorescent staining with confocal imaging, gene profiling,<br />

phospho-protein array analysis, western blot and ELISA validation.<br />

In vivo studies were performed by injection of MCF-7 ALK clones<br />

into using NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ mice using IACUC<br />

approved animal protocols.<br />

Results: When cultured on plastic substrates, MCF-7 ALK clones<br />

formed tumor cell aggregates instead of monolayer cultures, and when<br />

cultured as tumor spheroids under non-adherent 3D conditions had<br />

a distinct cellular phenotype with significantly greater clonogenicity<br />

than either non-target vector MCF-7 clones or the parental cell line.<br />

Whole transcriptome analysis, with validation using protein arrays,<br />

western blots and ELISA analysis revealed that the presence of<br />

ALK up-regulated phospho-src. In vivo studies revealed that ALK<br />

expressing MCF-7 clones formed tumor emboli that were enwrapped<br />

by dermal lymphatic vessels, essentially recapitulating the phenotype<br />

of IBC tumor emboli that exhibit encircling lymphovasculogenesis.<br />

Enforced expression of wild type ALK in another breast cancer cell<br />

line resulted in similar formation of tumor emboli.<br />

Discussion: These studies provide first time evidence for the<br />

association between full length ALK and formation of highly invasive<br />

tumor emboli enwrapped by lymphatic vessels, which is a primary<br />

characteristic of IBC. These studies, taken together with discovery of<br />

the prevalence of ALK gene amplification in IBC patients, indicate<br />

that ALK represents an important therapeutic target for IBC, with<br />

the availability of new ALK targeted therapies to evaluate as single<br />

agents and in combinations with other agents that may effectively<br />

target IBC tumor emboli that we have now linked to ALK and which<br />

represent the metastatic lesion of this lethal variant of breast cancer.<br />

Funding by Susan G. Komen Organization Promise Grant KG081287<br />

(FMR and MC).<br />

P6-10-05<br />

SU2C Phase 1b Trial of Dual PI3K/ mTOR Inhibitor BEZ235<br />

with Letrozole in ER+/HER2- Metastatic <strong>Breast</strong> <strong>Cancer</strong> (MBC)<br />

Mayer IA, Abramson VG, Balko JM, Isakoff SJ, Forero A, Kuba MG,<br />

<strong>San</strong>ders ME, Li Y, Winer E, Arteaga CL. Vanderbilt-Ingram <strong>Cancer</strong><br />

Center, Nashville, TN; Massachussets General Hospital, Boston, MD;<br />

University of Alabama at Birmingham, AL; MD Anderson <strong>Cancer</strong><br />

Center, Houston, TX; Dana-Farber <strong>Cancer</strong> Institute, Boston, MD<br />

Background: Mutations in PIK3CA, the gene encoding the p110alpha<br />

subunit of PI3K, have been associated with antiestrogen resistance<br />

in ER+ BC. In general, antiestrogen-resistant cancers retain ER and<br />

responsiveness to estradiol, suggesting that treatment of ER+/PI3K<br />

mutant BC should include PI3K inhibitors plus endocrine therapy.<br />

Methods: We are conducting a phase Ib open-label trial of letrozole<br />

(2.5 mg/d) with the dual PI3K/ mTOR inhibitor BEZ235 (400 mg<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

544s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: General Session 6<br />

BID, sachet formulation) in post-menopausal patients with ER+/<br />

HER2 negative MBC. Upon toxicity, BEZ235 was reduced to 400 mg<br />

in AM/ 200 mg in PM (dose level -1), and 200 mg BID (dose level<br />

-2), in a standard 3+3 de-escalation design. Treatment continues until<br />

unacceptable toxicity or disease progression. Tumor assessments are<br />

performed every 2 months. All tumor biopsies are being analyzed for<br />

PI3K pathway alterations.<br />

Results: To date, 9 patients have been accrued; 6 on dose level 1 (400<br />

mg BID) and 3 on dose level -1 (400 mg in AM/ 200 mg in PM).<br />

Of note, the latter 3 patients have been on treatment for less than 2<br />

weeks at the time of abstract submission. Median age was 55 years<br />

(range 40 - 65); all patients had disease progression on previous<br />

endocrine therapy, and 100% had bone metastases and 30% had<br />

visceral metastases. Preliminary toxicities for dose level 1 cohort (400<br />

mg BID) are summarized in the table below. The DLT was grade 3<br />

mucositis, which happened less than 3 weeks of study initiation, in<br />

a total of 3 out of 6 patients. All grade 3 mucositis cases improved<br />

after BEZ235 interruption for 7 days and subsequent dose reduction.<br />

These 3 events triggered activation of the dose level -1 cohort (400<br />

mg AM/ 200 mg PM). The first 3 patients on dose level 1 cohort (2<br />

of which have PIK3CA mutated tumors) had their tumor assessment<br />

scans at 8 weeks post treatment initiation; all patients were found to<br />

have stable disease.<br />

Toxicity: Percentage (%) of Patients by Grade<br />

Toxicity Grade 1 (%) Grade 2 (%) Grade 3 (%) Total<br />

Mucositis 0 22 22* 44<br />

Nausea 44 0 0 44<br />

Diarrhea 44 11 0 55<br />

Dyspepsia 0 11 0 11<br />

Fatigue 11 11 0 22<br />

* DLT<br />

Discussion: The combination of letrozole/BEZ235 is still being<br />

evaluated in patients with ER+/HER2MBC. Safety, tolerability and<br />

preliminary antitumor activity data will be updated at the meeting.<br />

P6-10-06<br />

Rational combination therapy against triple-negative breast<br />

cancer<br />

Al-Ejeh F, Miranda M, Simpson PT, Chenevix-Trench G, Lakhani SR,<br />

Khanna KK. Queensland Institute of Medical Research, Brisbane,<br />

QLD, Australia; The University of Queensland, Brisbane, QLD,<br />

Australia<br />

Background: Triple negative breast cancer (TNBC) is an aggressive<br />

subtype of breast cancer with higher incidence of recurrence,<br />

more distant metastasis, and poorer survival. This subtype is also<br />

characterized by complex genomes where little of their genomes<br />

remain at normal copy number but without high, focal copy number<br />

amplifications. At the transciptome level, the majority of TNBC<br />

(∼75%) are classified as basal-like breast cancer (BLBC) according<br />

to the five intrinsic subtypes. Despite considerable genomic and<br />

gene expression characterization of TNBC, proteomic and phosphoproteomic<br />

investigations of this disease are limited with no available<br />

targeted therapies in clinical use.<br />

Methods & Results: We used the Kinex TM antibody array (http://<br />

www.kinexus.ca/) to interrogate protein/phosphoproteins levels in<br />

43 primary breast cancer biopsies (16 TNBC, 16 ER/PR positive<br />

and 11 HER2-positive) and 16 breast cancer cell lines. Unsupervised<br />

hierarchical clustering of protein/phosphoprotein levels revealed<br />

two subgroups of TNBC in comparison to other subtypes. Western<br />

blotting and Proteome Profiler TM Arrays (R&D Systems) were used<br />

to validate deregulated proteins/phosphoproteins in TNBC. Pathway<br />

analysis revealed that one subgroup of TNBC exploits overlapping<br />

and cross-talking networks for survival. These signaling networks<br />

are downstream from elevated activation of EGFR, integrins and<br />

Insulin-like growth factor 1 receptor (IGF1R).<br />

Targeted molecular inhibitors of activated kinases in these pathways<br />

showed specificity against basal-like/TNBC cell lines compared to<br />

other subtypes in vitro. These activated kinases/networks represent<br />

druggable targets for the treatment of TNBC but may be limited by<br />

compensatory effect of the complex cross-talking signaling networks.<br />

To overcome compensatory downstream signaling that would limit<br />

the inhibition of a given pathway; we developed EGFR-targeted<br />

radioimmunotherapy (RIT) strategy to systemically deliver cytotoxic<br />

loads of beta particles ( 177 Lu) that would kill targeted cells and<br />

surrounding cells by crossfire effect. The combination of EGFRdirected<br />

RIT with chemotherapy and PARP inhibition successfully<br />

treated orthotopic and metastatic TNBC models established from<br />

cell lines and patient-derived xenografts. The superior efficacy of<br />

this triple-agent combination therapy is explained by enhanced DNA<br />

damage and reduced DNA repair response, higher apoptotic cell death<br />

and the elimination of putative breast cancer stem cells.<br />

Conclusion: Proteomic analysis of TNBC provides a powerful tool<br />

to elucidate druggable signaling networks with therapeutic potential.<br />

TNBC utilizes complex interacting signaling networks and rational<br />

combination therapies are required for effective therapy.<br />

P6-10-07<br />

Phase I study of BYL719, an alpha-specific PI3K inhibitor, in<br />

patients with PIK3CA mutant advanced solid tumors: preliminary<br />

efficacy and safety in patients with PIK3CA mutant ER-positive<br />

(ER+) metastatic breast cancer (MBC)<br />

Juric D, Argiles G, Burris HA, Gonzalez-Angulo AM, Saura C, Quadt<br />

C, Douglas M, Demanse D, De Buck S, Baselga J. Massachussetts<br />

General Hospital <strong>Cancer</strong> Center, Boston, MA; Vall d’Hebron<br />

University Hospital, Barcelona, Spain; Sarah Cannon Research<br />

Institute, Nashville, TN; M.D. Anderson <strong>Cancer</strong> Center, Houston, TX;<br />

Novartis Pharma Corporation, East Hanover, NJ; Novartis Pharma<br />

AG, Basel, Switzerland<br />

Background:<br />

Deregulation of the PI3K pathway occurs in >50% of breast cancers.<br />

Mutations in PIK3CA, the gene encoding the p110alpha subunit of<br />

PI3K contributes to resistance to endocrine and anti-HER2 therapies.<br />

BYL719 is a potent, specific oral inhibitor of the alpha subunit of<br />

PI3K, which inhibits wild-type p110alpha and its most common<br />

somatic mutations (IC 50 =5 nM) much more potently than other PI3K<br />

isoforms.<br />

Methodology:<br />

BYL719 has been investigated in a phase I study in patients with<br />

PIK3CA mutant advanced solid tumors. BYL719 has a favorable<br />

safety and PK profile; the MTD was established at 400 mg once<br />

daily (Juric et al., AACR 2012). As of 5 September 2012, 79 patients<br />

have been enrolled in this phase I study, among them 20 patients<br />

with PIK3CA mutant ER+ MBC. 5 of these patients had ER+/<br />

HER2 positive (HER2+) MBC. Presence of PIK3CA mutations was<br />

determined by either local or central assessment.<br />

Results:<br />

20 patients with ER+ PIK3CA mutant MBC were treated once daily<br />

with BYL719 at doses of 30 mg (n=1), 180 mg (n=1), 270 mg (n=1),<br />

400 mg (n=14) and 450 mg (n=3). Median age was 55 years (range<br />

40-78). 15/20 patients with ER+ PIK3CA mutant MBC had HER2<br />

negative and 5/20 had HER2+ MBC. Patients had received a median<br />

of 5 previous treatment regimens (range 1-11). Most patients had<br />

received multiple prior lines of endocrine therapy including tamoxifen<br />

www.aacrjournals.org 545s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

and aromatase inhibitors; 9 patients also received fulvestrant and 16<br />

patients received prior chemotherapy. HER2+ patients have been<br />

previously treated with trastuzumab and/or lapatinib.<br />

Based on a recent analysis of 19 patients (data cut-off 5 July 2012), the<br />

safety of BYL719 in patients with ER+ MBC was comparable to the<br />

overall study population. Most frequent adverse events (AEs; ≥10%)<br />

suspected to be related to BYL719 were hyperglycemia, diarrhea,<br />

nausea, fatigue, decreased appetite, vomiting, rash and erythematous<br />

rash, dysgeusia, dry mouth, and stomatitis. The majority of AEs were<br />

CTCAE grade 1/2 and were clinically well manageable. Most frequent<br />

CTCAE grade 3/4 events were hyperglycemia and erythematous rash.<br />

18/20 PIK3CA mutant ER+ MBC patients were treated at potentially<br />

effective doses of ≥270 mg/day. 6/18 patients (33%) achieved tumor<br />

shrinkage > 20%, among them 2 patients achieved partial responses<br />

(PR) by RECIST. Median progression free survival (PFS) analyzed<br />

in 17 patients treated at ≥270 mg/day in the ER+ BC patients was<br />

7.3 months (CI [4.6,9.5]), compared to 3.7 months (CI [1.8,5.5]) in<br />

38 patients with other PIK3CA mutant solid tumors (data cut-off 5<br />

July 2012)<br />

Conclusion: BYL719 has a favorable safety profile with manageable<br />

side effects, which were mainly related to its mechanism of action.<br />

BYL719 shows promising preliminary clinical activity as single agent<br />

in patients with PIK3CA mutant ER+ MBC, warranting further clinical<br />

development. The investigation of BYL719 continues in patients with<br />

ER+ MBC as single agent, and in combination with endocrine therapy.<br />

P6-11-01<br />

Intermittent High Dose Proton Pump Inhibitor Improves<br />

Progression Free Survival as Compared to Standard<br />

Chemotherapy in the First Line Treatment of Patients with<br />

Metastatic <strong>Breast</strong> <strong>Cancer</strong><br />

Hu X, Wang B, Sun S, Chiesi A, Wang J, Zhang J, Fais S. Fudan<br />

University Shanghai <strong>Cancer</strong> Center, Shanghai, China; National<br />

Institute of Health, Roma, Italy<br />

Purpose<br />

High dose proton pump inhibitor (PPI) has proved to be potentially<br />

effective when combined with chemotherapy in preclinical data.<br />

This study (NCT01069081) was designed to investigate whether the<br />

efficacy of chemotherapy could be improved with the addition of<br />

proton pump inhibitor (PPI) in patients with metastatic breast cancer.<br />

Patients and Methods<br />

Females elder than 18 years old with histologically confirmed<br />

metastatic breast cancer were eligible for participation. Patients<br />

enrolled were randomly assigned to three arms: Arm A, docetaxol 75<br />

mg/m 2 on d4, followed by cisplatin 75 mg/m 2 on d4, repeated every<br />

21 days until maxinum 6 cycles or presence of disease progression<br />

or intolerable toxicity; Arm B, the same chemotherapy plus<br />

esomeprazole 80 mg p.o. bid three days on and the subsequent four<br />

days off, beginning on d1 repeated weekly up to disease progression,<br />

intolerable toxicity, patient’s withdrawal, or a maximum of 66<br />

weeks; Arm C, the same as Arm B with the only difference being<br />

dose of esomeprazole at 100 mg p.o. bid. The primary endpoint was<br />

progression free survival (PFS), and the secondary endpoints were<br />

time to progression (TTP), objective response rate (ORR), safety<br />

profile, and overall survival (OS).<br />

Results<br />

From Aug. 2009 to Dec. 2011, 100 women signed informed consent<br />

form (ICF) and 94 patients have undergone at least one injection of<br />

chemotherapy. Three patients had severe hypersensitivity reactions,<br />

two occurring after the first injection of docetaxol and one in the<br />

second cycle. After a median follow up of 17 months, 68 (72.3%)<br />

patients got disease progression and 23 (24.5%) patients died. Median<br />

PFS for the whole group (n=94), arm A (n=33), arm B (n=30), arm C<br />

(n=31) were 8.9, 7.5, 10.9, and 9.5 months, respectively (p=0.082).<br />

A significant difference was observed between patients who had<br />

taken PPI and who not with median PFS of 9.5 and 7.5 months,<br />

respectively (p=0.030). Among 17 patients with triple negative<br />

breast cancer, this difference was bigger with median PFS of 9.5 and<br />

3.3 months, respectively (p=0.014). The overall response rates for<br />

the whole group, arm A, arm B, arm C were 58.5%, 51.5%, 63.3%,<br />

61.3%, respectively.<br />

Conclusion<br />

This trial is the first randomized study to demonstrate antitumor effects<br />

of intermittent high dose PPI in patients with metastatic breast cancer.<br />

Docetaxel and cisplatin doublet is comparable to the other first-line<br />

chemotherapeutical regimens and the addition of proton pump<br />

inhibitor to the doublet improves efficacy with no adding toxicity,<br />

especially in patients with triple negative breast cancer.<br />

Key Words Proton Pump Inhibitor (PPI), Triple Negative <strong>Breast</strong><br />

<strong>Cancer</strong> (TNBC), Phase II Study, Metastatic <strong>Breast</strong> <strong>Cancer</strong>,<br />

Chemotherapy<br />

P6-11-02<br />

Inhibition of mTOR and Fatty Acid Synthase (FASN) overcome<br />

acquired resistance to Trastuzumab, Lapatinib and both in<br />

HER2+ breast cancer<br />

Puig T, Blancafort A, Giro A, Viqueira A, Viñas G, Massaguer A,<br />

Carrion-Salip D, Bolos MV, Urruticoechea A, Oliveras G. University<br />

of Girona and Girona Biomedical Research Institute (IDIBGi),<br />

Girona, Spain; Catalan Institute of Oncology (ICO) and Girona<br />

Biomedical Research Institute (IDIBGi), Girona, Spain; Pfizer,<br />

Madrid, Spain; University of Girona, Spain; Catalan Institute of<br />

Oncology (ICO), Hospitalet de Llobregat, Barcelona, Spain<br />

Introduction:<br />

Resistance to dual biological blockade of HER2 positive breast cancer<br />

arises as a novel therapeutic challenge with high clinical relevance.<br />

We have shown that inhibition of fatty acid synthase (FASN) has<br />

anticancer activity and enhances the effect of chemotherapy and<br />

anti-HER2 drugs in pre-clinical breast cancer models. Furthermore,<br />

our group has recently observed that inhibition of FASN can reverse<br />

resistance to anti-HER2 therapies.<br />

The development of FASN inhibitors has consequently appeared as a<br />

novel anti-target modality for treating cancer. However, the clinical<br />

use of FASN inhibitors, such as Cerulenin, C75 and Epigallocatechin<br />

3-gallate (EGCG) is limited by anorexia and induced body weight<br />

loss or by its low in vivo potency and stability. We synthesized novel<br />

EGCG-related inhibitors to improve their use as anti-tumor agents.<br />

Within these, G28UCM was selected for its inhibitory effect of FASN<br />

activity and selective cytotoxicity in tumor cells.<br />

Recently, it has been reported that mTOR blockade acts synergistically<br />

with HER2 inhibition to induce cell death and tumor regression in<br />

resistant breast cancer models.<br />

Materials and Methods:<br />

We have developed long term HER2+/ FASN+ breast cancer cell lines<br />

(SKBr3) resistant to the HER2-monoclonal antibody Trastuzumab<br />

(SKTR), the EGFR/HER2-tyrosine kinase inhibitor Lapatinib<br />

(SKLR) or both (SKLTR). Once established, we have characterized<br />

these cells by studying a panel of EGF receptors signaling proteins<br />

with western blot analysis, changes in adherence to extracellular<br />

matrix proteins and invasion capacity with colorimetric assays. Using<br />

MTT assay, we have assessed the effect of the mTOR-inhibitor,<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

546s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: General Session 6<br />

Temsirolimus, and G28UCM on viability of parental and resistant<br />

cells. The isobologram method has been used to estimate the possible<br />

synergistic effect between both treatments.<br />

Results:<br />

Resistant cells maintained downstream HER2 pathway activation<br />

by stimulating the expression/activation of alternative EGF family<br />

receptors and/or those specific ligands. Moreover, these cells increased<br />

adherence to extracellular matrix proteins and invasion capacity.<br />

Different combination regiment of Temsirolimus with G28UCM<br />

displayed a strong synergistic effect in inducing cell death of both,<br />

parental and resistant HER2+ breast cancer cells.<br />

SKBr3 SKTR SKLR SKLTR<br />

Trastuzumab 30,5 +/- 0,7 n.s. n.s. n.s.<br />

Lapatinib 1,3 +/- 1,1 9,5 +/- 2,1 n.s. n.s.<br />

Temsirolimus 4,8 +/- 0,9 6,0 +/- 0,8 5,3 +/- 0,7 5,7 +/- 1,2<br />

G28UCM 11,6 +/- 1,9 10,3 +/- 1,5 11,2 +/- 1,4 5,5 +/- 1,1<br />

Temsirolimus + Ix=0,02 +/- 0,02** Ix=0,71 +/- 0,02** Ix=0,03 +/- 0,09** Ix=0,55 +/- 0,11**<br />

G28UCM (synergism) (synergism) (synergism) (synergism)<br />

Table 1. IC30 value (µM). ns, no significant cell proliferation inhibition. Interaction index (Ix): Ix ><br />

1 is antagonism, Ix = 1 is additivism and Ix < 1 is sinergism. Data are means ± SE. * (p < 0.05) and<br />

** (p < 0.01) indicate the level of statistical significance of the Ix compared with an Ix of 1.<br />

Conclusions:<br />

The inhibition of mTOR and FASN is a potential novel therapeutic<br />

strategy in dual resistant HER2 positive breast cancer.<br />

P6-11-03<br />

Site-Specific, Concomitant Delivery of Rapamycin and Paclitaxel<br />

in <strong>Breast</strong> <strong>Cancer</strong>: Consequent Synergistic Efficacy Enhancement<br />

Blanco E, <strong>San</strong>gai T, Hsiao A, Ruiz-Esparza GU, Ferrari M, Meric-<br />

Bernstam F. The Methodist Hospital Research Institute, Houston, TX;<br />

The University of Texas MD Anderson <strong>Cancer</strong> Center, Houston, TX<br />

Background: The phosphatidylinositol 3-kinase (PI3K)/Akt/<br />

mammalian target of rapamycin (mTOR) (PI3k/Akt/mTOR) pathway<br />

is dysregulated in certain breast cancers. Ongoing clinical trials aim<br />

to therapeutically exploit this pathway through administration of<br />

rapamycin (RAP), an mTOR inhibitor, in combination with paclitaxel<br />

(PTX). However, actual drug synergy in clinical settings may not<br />

be fully realized due to disparate pharmacokinetic parameters of<br />

individual drug formulations, wherein drugs or their effects may never<br />

be present in the tumor at the same time. Our objective was to generate<br />

a nanoparticle platform capable of site-specifically delivering precise<br />

amounts of rapamycin and paclitaxel to breast tumors with hopes<br />

of increasing synergistic targeting of the PI3k/Akt/mTOR pathway.<br />

Materials and Methods: Drug-containing nanoparticles composed<br />

of amphiphilic block copolymers of pegylated poly(ε-caprolactone)<br />

(PEG-PCL, MW = 5k-5k) were fabricated and nanoparticle size and<br />

drug loading efficiency was determined. In vitro growth inhibition<br />

of nanoparticle formulations of varying ratios was evaluated in<br />

MCF-7 and MDA-MB-468 breast cancer cells via sulforhodamine<br />

B assays, after which median-effect plot analyses and combination<br />

index calculations were conducted. Antitumor efficacy studies were<br />

performed in female nude mice bearing MDA-MB-468 tumors, in<br />

which nanoparticles were administered intravenously twice a week<br />

for the duration of three weeks. Biodistribution of drug-containing<br />

nanoparticles in extracted tumors were examined, as well as reverse<br />

phase protein array (RPPA) analysis to gain insights into site-specific<br />

synergy.<br />

Results: Nanoparticles were spherical, with an average diameter of<br />

9 nm. Both rapamycin and paclitaxel loaded favorably, allowing for<br />

customization of different ratios within nanoparticles. Combination<br />

indices demonstrated that a 3:1 ratio of RAP:PTX had the most<br />

synergy in MDA-MB-468 breast cancer cells in vitro, a synergy<br />

found to be preserved in vivo. Significant tumor regression (> 1.5<br />

fold reduction from initial tumor volume) was observed in vivo upon<br />

administration of 3:1 RAP:PTX (15:5 mg/kg) nanoparticles. The<br />

precise ratio of rapamycin and paclitaxel (3:1) was found maintained<br />

in tumors 24 h after administration, an effect not achievable with free<br />

drug formulations. RPPA analysis demonstrated effective blocking<br />

of mTOR and Akt 24 h after administration of nanoparticles, key<br />

events in drug synergy.<br />

Discussion: Site-specific delivery of synergistic agents in preciselycontrolled<br />

drug ratios, possible through their incorporation into<br />

nanoparticles, was shown to be highly efficacious against breast<br />

tumors. Findings demonstrate the ability to deliver specific drug ratios<br />

to tumors, potentially precluding the need to administer maximal<br />

doses of both agents in order to achieve synergy, lessening patient<br />

side-effects. This study demonstrates the potential for prediction<br />

of in vivo therapeutic outcomes from in vitro synergistic findings.<br />

Nanoparticle delivery of drugs may also yield enhanced understanding<br />

of mechanisms of synergy between molecular-targeted drugs and<br />

traditional chemotherapeutics in vivo, resulting in novel and more<br />

efficacious treatment regimens.<br />

P6-11-04<br />

Targeting the tumor microenvironment: tetrathiomolybdate<br />

decreases circulating endothelial progenitor cells in women with<br />

breast cancer at high risk of relapse.<br />

Jain S, Kornhauser N, Lam C, Ward MM, Chuang E, Cigler T, Moore<br />

A, Donovan D, Cobham MV, Schneider S, Hurtado Rua SM, Lane<br />

ME, Mittal V, Vahdat LT. Weill Cornell Medical College<br />

Background: Bone marrow-derived endothelial progenitor cells<br />

(EPCs) constitute an important part of the tumor microenvironment<br />

and are critical for metastatic progression in preclinical models and<br />

breast cancer patients (Jain et al, <strong>Breast</strong> <strong>Cancer</strong> Res Treat, 2012).<br />

Tetrathiomolybdate (TM), a copper-depleting compound inhibits<br />

angiogenesis, tumor growth, and metastasis. This study explores<br />

the effect of TM on EPCs in patients at high risk for breast cancer<br />

recurrence.<br />

Methods: This phase II study enrolled stage 3, 4 without evidence of<br />

disease (NED), and any node-positive triple negative breast cancer<br />

patient. Only concomitant hormone therapy was allowed. Patients<br />

received induction TM 180 mg daily at baseline followed by an<br />

equal or lower daily dose (median 100 mg, range 0-140) to maintain<br />

ceruloplasmin (Cp) level < 17 mg/dl (target for copper depletion).<br />

We monitored EPCs (CD45dim/CD133+/VEGFR2+), Cp, CEA, and<br />

CA15-3 at baseline and monthly. Wilcoxon signed-rank was used to<br />

compare Cp and EPC levels between baseline and subsequent time<br />

points. All p-values were two-sided with statistical significance<br />

evaluated at the 0.05 alpha level.<br />

Results: 50 patients (33 adjuvant, 17 Stage 4 NED, and 22 triple<br />

negative) were enrolled. In the first 40 patients enrolled who had<br />

received at least 24 months of TM, EPC and Cp data were available<br />

for analysis. Of these 40 patients, 1 patient did not take TM due to<br />

patient preference, and 736 cycles of TM (average 18.9 per patient)<br />

were administered. Median age was 50 years (range 29-66). Median<br />

number of tumor size and positive lymph nodes among adjuvant<br />

patients were 3.5 cm (range 1.2-7) and 9 (range 0-42), respectively. Of<br />

the patients receiving hormone therapy, 11 patients were on tamoxifen<br />

and 16 patients were on an aromatase inhibitor. Median baseline Cp<br />

level was 30 mg/dL (range 20-47). 71% patients adequately copper<br />

depleted at month 1 to a mean Cp of 14.8 mg/dL. A larger proportion of<br />

triple negative patients copper depleted (82%) compared to hormone<br />

receptor positive subtypes (47%) and HER2/neu positive subtypes<br />

(67%). Median EPCs/ml decreased from baseline to last dose by<br />

16 in patients that achieved the copper depletion target, p=0.014.<br />

www.aacrjournals.org 547s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Conversely, in patients that did not copper deplete, median EPCs/ml<br />

increased by 136, p=0.005. Of the 50 patients on study, 7 patients<br />

relapsed in which a significant increase in EPCs preceded an objective<br />

clinical relapse and a tumor marker rise by a median of 1 month. Only<br />

grade 3/4 toxicity was hematologic, occurred in 49 cycles (6.7%),<br />

and resolved in 5-13 days with TM held and resumed at a lower dose.<br />

Conclusions: TM is a well-tolerated oral copper chelator that may<br />

contribute to maintaining EPCs below baseline in copper-depleted<br />

patients. Molecular subtype may impact on the ability to copper<br />

deplete. EPCs may have potential as a surrogate marker for early<br />

relapse and as a therapeutic target for interrupting the metastatic<br />

progression.<br />

P6-11-05<br />

Novel sorafenib-derivatives induce apoptosis in breast cancer<br />

cells through STAT3 inhibition<br />

Liu C-Y, Tseng L-M, Chang K-C, Chu P-Y, Su J-C, Shiau C-W, Chen<br />

K-F. Taipei Veterans General Hospital, Taipei, Taiwan; National<br />

Yang-Ming University, Taipei, Taiwan; St. Martin De Porres Hospital,<br />

Chia-Yi, Taiwan; National Taiwan University Hospital, Taipei, Taiwan<br />

Background: STAT3 has emerged as a novel potential anti-cancer<br />

target and its signaling is constitutively activated in various cancers<br />

including breast cancer. Our previous study has shown that STAT3 is<br />

a major kinase-independent target of sorafenib in HCC. (J Hepatol.<br />

2011). Moreover, recent evidence has shown that p-STAT3 can be<br />

down-regulated by phosphatases, such as SHP-1 tyrosine phosphatase.<br />

We have designed and synthesized a series of sorafenib analogues<br />

devoid of sorafenib’s kinase inhibition activity, some of which showed<br />

stronger p-STAT3 inhibition and apoptosis-inducing effects than<br />

sorafenib in HCC cells (Eur J Med Chem. 2011). Here, we tested<br />

the efficacy of novel sorafenib derivatives, SC-1 and SC-43, in breast<br />

cancer cells and examined the drug mechanism.<br />

Methods: Six <strong>Breast</strong> cancer cell lines were used for in vitro studies.<br />

Cell viability was examined by MTT-assay. Apoptosis was examined<br />

by both flow cytometry and Western blot. Signal transduction<br />

pathways in cells were assessed by Western Blot. SHP-1 activity<br />

was measured by a phosphatase activity assay kit. In vivo efficacy<br />

of Sorafenib, and sorafenib derivatives were tested in xenografted<br />

nude mice.<br />

Results: Sorafenib, SC-1 and SC-43 induced apoptosis in association<br />

with down-regulation of P-STAT3 and its downstream proteins Cyclin<br />

D1 and survivin in a dose-dependent manner in breast cancer cell<br />

lines (HCC-1937, MDA MB-468, MDA MB-231, MDA MB-453,<br />

SKBR-3, MCF-7) and the apoptotic effects induced by SC-1 and<br />

SC-43 were more potent than Sorafenib. Over-expression of STAT3<br />

in MDA MB-468 cells protected cells from apoptosis induced by<br />

sorafenib, SC-1 and SC-43. Moreover, SC-1 and SC-43 up-regulated<br />

higher SHP-1 activity than sorafenib by in vitro phosphatase assays.<br />

Knockdown of SHP-1 by siRNA reduced apoptosis induced by SC-1<br />

and SC-43. Importantly, SC-1 and SC-43 showed in vivo efficacy in<br />

MDA-468 xenografted tumor.<br />

Conclusions: Our data indicated that inhibiton of p-STAT3 by upregulating<br />

SHP-1 activity mediated apoptotic effects of sorafenib,<br />

SC-1 and SC-43 in breast cancer cells.<br />

P6-11-06<br />

A phase Ib study of LCL161, an oral inhibitor of apoptosis (IAP)<br />

antagonist, in combination with weekly paclitaxel in patients with<br />

advanced solid tumors<br />

Dienstmann R, Vidal L, Dees EC, Chia S, Mayer EL, Porter D,<br />

Baney T, Dhuria S, Sen SK, Firestone B, Papoutsakis D, Cameron<br />

S, Infante JR. Vall d’Hebron University Hospital, Barcelona, Spain;<br />

Hospital Clinic i Provincial, Barcelona, Spain; University of North<br />

Carolina Lineberger Comprehensive <strong>Cancer</strong> Center, Chapel Hill, NC;<br />

British Columbia <strong>Cancer</strong> Agency, University of British Columbia,<br />

Vancouver, BC, Canada; Dana-Farber <strong>Cancer</strong> Institute, Boston, MA;<br />

Novartis Pharmaceuticals Corporation, Cambridge, MA; Novartis<br />

Pharmaceuticals Corporation, Florham Park, NJ; Sarah Canon<br />

Research Institute, Nashville, TN<br />

Background: Impaired apoptosis is a common feature of cancer<br />

cells and may contribute to chemoresistance. LCL161 is an oral<br />

small molecule antagonist of Inhibitor of Apoptosis Proteins<br />

(IAPs) that sensitizes a subset of tumors from diverse lineages to<br />

treatment with cytotoxic therapies, including paclitaxel. Multiple<br />

breast cancer models are sensitive to LCL161 as a single agent and<br />

LCL161 acts synergistically with paclitaxel in these models. A phase<br />

I study established an LCL161 dose of 1800 mg once weekly as<br />

well tolerated, with strong evidence of pharmacodynamic activity<br />

at doses ≥320 mg. This ongoing phase Ib study defines the dose<br />

limiting toxicities (DLTs), maximum tolerated dose (MTD), safety,<br />

and pharmacokinetics (PK) of LCL161 in combination with weekly<br />

paclitaxel.<br />

Methods: Patients with advanced/metastatic solid tumors were treated<br />

with paclitaxel 80 mg/m 2 each week followed by escalating doses of<br />

LCL161 administered once weekly immediately following paclitaxel.<br />

PK and biomarker sampling was performed.<br />

Results: Thirty-two patients have received LCL161 doses of 600 mg<br />

(n=3), 1200 mg (n=5), 1500 mg (n=4), and 1800 mg (n=20). The most<br />

frequent adverse events considered LCL161-related included diarrhea<br />

(n=11; 1 Grade 3), nausea (n=8), fatigue (n=7; 2 Grade 3), peripheral<br />

neuropathy (n=6; 1 Grade 3), vomiting (n=6), decreased appetite<br />

(n=5), alopecia (n=4), and anemia (n=4). The principal DLTs were<br />

neutropenia, fatigue, and neuropathy. Significant cytokine release<br />

syndrome, the DLT of single-agent LCL161, has not been observed<br />

likely due to the use of dexamethasone as a premedication. No PK<br />

interaction between LCL161 and paclitaxel was observed. RECIST<br />

partial responses have been observed in 4 patients with diverse tumor<br />

types, including breast cancer. Preliminary antitumor activity in the<br />

expansion cohort with breast cancer patients will be presented.<br />

Discussion: LCL161 and paclitaxel combination therapy is well<br />

tolerated, with manageable toxicities and no evidence of a PK<br />

interaction that might interfere with the activity of either agent.<br />

Enrollment of additional patients with breast and ovarian cancer into<br />

an expansion cohort is ongoing, utilizing an approach to identify those<br />

more likely to respond to treatment with IAP antagonists.<br />

P6-11-07<br />

The <strong>Cancer</strong> Stem Cell-Targeting Wnt Inhibitor VS-507 Reduces<br />

<strong>Breast</strong> <strong>Cancer</strong> Growth and Metastasis<br />

Ring JE, Kolev VN, Padval MV, Keegan M, Vidal CM, Neill AA,<br />

Shapiro IM, Pachter JA, Xu Q. Verastem, Inc., Cambridge, MA<br />

<strong>Cancer</strong> stem cells (CSCs) support tumor viability and growth through<br />

their ability to self-renew and differentiate into heterogeneous tumor<br />

tissue. CSCs are typically resistant to standard cytotoxic agents,<br />

leading to their enrichment and the consequent regrowth of refractory<br />

tumors. While this population has been challenging to directly target<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

548s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: General Session 6<br />

therapeutically, we are actively developing novel agents, such as VS-<br />

507, that selectively target the cancer stem cell subpopulation in vivo.<br />

Previously, we and others have shown that VS-507, a cancer stem-cell<br />

specific agent, inhibits Wnt signaling with corresponding reduction<br />

of the LRP6 protein, a Frizzled co-receptor upregulated in breast<br />

cancer cell lines. In the current study, we have continued to examine<br />

the effect of VS-507 on the Wnt/Beta-catenin signaling pathway to<br />

further elucidate its mechanism of action. We determined that VS-<br />

507 also decreases expression of the second Frizzled co-receptor<br />

LRP5 in MDA-MB-231 breast cancer cells stimulated by Wnt3A.<br />

The combined inhibition of the co-receptors LRP5 and LRP6 may<br />

contribute to the observed inhibition of Beta-catenin-mediated<br />

transcription by VS-507 in a TOP-Flash assay. Accordingly, VS-507<br />

reduced expression of Axin2, a transcriptional target of Beta-catenin.<br />

Because human breast cancer cell lines represent a mixed population<br />

of CSC and non-CSC cells, we evaluated VS-507 across a panel of cell<br />

lines by monitoring changes in viability and in the percentage of cancer<br />

stem cells. In SUM159 triple negative breast cancer cells, VS-507<br />

had micromolar potency against the bulk population with preferential<br />

nanomolar potency against the ALDEFLUOR+ CSC population with<br />

similar effects observed in the Hoechst side population (SP) CSC<br />

assay. In contrast, cytotoxic anticancer drugs such as paclitaxel and<br />

cisplatin increased the percentage of ALDEFLUOR+ cancer stem<br />

cells under similar conditions. In vivo, oral administration of VS-507<br />

(15 mg/kg, QD daily) as a single agent partially inhibited MDA-<br />

MB-231 tumor growth, consistent with its observed inhibition of the<br />

cancer stem cell subpopulation. Furthermore, VS-507 inhibited lung<br />

metastasis in a 4T1.2 murine breast cancer model. In summary, these<br />

results further elucidate the mechanism by which VS-507 inhibits<br />

Wnt/Beta-catenin signaling. VS-507 reduces tumor cell proliferation<br />

in vitro with preferential effects on cancer stem cells which translate<br />

to inhibition of tumor growth, enhancement of efficacy of cytotoxic<br />

agents such as docetaxel, and inhibition of metastasis in mouse<br />

models. These data provide additional support for the development<br />

of VS-507 as a novel anti-cancer stem cell agent.<br />

P6-11-08<br />

A multicenter, open-label Ph IB/II study of BEZ235, an oral dual<br />

PI3K/mTOR inhibitor, in combination with paclitaxel in patients<br />

with HER2-negative, locally advanced or metastatic breast cancer<br />

Campone M, Fumoleau P, Gil-Martin M, Isambert N, Beck JT,<br />

Becerra C, Shtivelband M, Duval V, di Tomaso E, Roussou P, Urban<br />

P, Urruticoechea A. Centre René Gauducheau, Nantes, France;<br />

Centre Georges François Leclerc, Dijon, France; Institut Català<br />

d’Oncologia, Barcelona, Spain; Highlands Oncology Group,<br />

Fayetteville, AR; Baylor University Medical Center, Dallas, TX;<br />

Ironwood <strong>Cancer</strong> and Research Centers, Chandler, AZ; Novartis<br />

Pharma AG, Basel, Switzerland; Novartis Institutes for BioMedical<br />

Research, Inc., Cambridge, MA<br />

Background: The PI3K/AKT/mTOR pathway is frequently altered<br />

in breast cancer (BC). Alterations in PIK3CA or inactivation of<br />

PTEN are observed in 10–40% and in up to 50% of breast tumors,<br />

respectively. The PI3K/AKT/mTOR pathway can be further activated<br />

through various receptor classes or cross-talk with other pathways,<br />

making it a rational target for therapeutic intervention in BC. BEZ235<br />

is an oral dual inhibitor of mTOR and PI3K. It has demonstrated antiproliferative<br />

activity, substantial growth inhibition, and induction of<br />

apoptosis in preclinical studies. In a Ph I study, single-agent BEZ235<br />

(600 mg BID) was shown to have less toxicity than the equivalent<br />

once-daily dosing, and have preliminary evidence of activity in<br />

advanced solid tumors (Arkenau, et al. ASCO 2012:#3097).<br />

Methods: This is a multicenter, open-label Ph IB/II study of<br />

continuous, oral BEZ235 twice daily (BID) in combination with<br />

paclitaxel (80 mg/m 2 ; IV weekly [QW]) in women with HER2-<br />

negative (HER2–), metastatic or inoperable locally advanced BC<br />

(NCT01495247).<br />

For the Ph IB part, women with HER2–, metastatic or inoperable<br />

locally advanced BC, who are suitable for treatment with paclitaxel,<br />

are eligible for enrollment. The primary objective is to determine the<br />

maximum tolerated dose (MTD)/recommended Ph II dose (RP2D)<br />

of BEZ235 in combination with paclitaxel based on dose-limiting<br />

toxicities (DLTs) using an adaptive 5-parameter Bayesian logistic<br />

regression model with overdose control. The MTD is defined as the<br />

highest drug dosage not causing medically unacceptable DLTs in<br />

more than 35% of the treated patients during Cycle 1 (1 cycle = 28<br />

days). Secondary objectives include safety (CTCAE), preliminary<br />

activity (RECIST), and pharmacokinetics (PK). Estimated enrollment<br />

is 15–30 patients into the Ph IB part.<br />

Results: As of June 2012, 13 pts have been enrolled into the Ph IB<br />

part of the trial. The first cohort (n=7 pts) received BEZ235 200 mg<br />

BID + 80 mg/m 2 QW paclitaxel. Of these 7 pts, 3 are ongoing, with 2<br />

pts having received treatment for more than 12 weeks so far, and 4 pts<br />

have discontinued (2 due to an adverse event [AE]; 1 due to an AE/<br />

pt’s decision; and 1 due to disease progression). Of the 6 evaluable<br />

pts in the first cohort, 2 experienced DLTs: Grade 3 stomatitis (1<br />

pt) and Grade 2/3 neutropenia (1 pt). Most common AEs included<br />

stomatitis and GI toxicity (e.g. diarrhea, nausea/vomiting). To date,<br />

reported Grade 3 AEs related to study drug were stomatitis (2 pts),<br />

neutropenia (1 pt), and skin rash (1 pt). Among the 3 pts with at least<br />

one tumor evaluation, 1 pt with a triple-negative metastatic BC, who<br />

had previously been treated with paclitaxel, experienced a RECIST<br />

partial response which was confirmed on second tumor evaluation.<br />

PK analysis is ongoing.<br />

Conclusions: Additional patients have been enrolled at BEZ235 200<br />

mg BID/paclitaxel 80 mg/m 2 QW to provide further information on<br />

the safety and activity profile of this combination. Updated safety<br />

and efficacy results will be presented. Upon determination of the<br />

MTD/RP2D, the randomized Ph II part will begin to compare<br />

weekly paclitaxel given with or without BEZ235 BID as the first-line<br />

treatment of HER2– metastatic BC.<br />

P6-11-09<br />

FAK Inhibitor VS-4718 Attenuates <strong>Breast</strong> <strong>Cancer</strong> Stem Cell<br />

Function In Vitro and In Vivo<br />

Kolev VN, Vidal CM, Shapiro IM, Pavdal M, Keegan M, Xu Q, Pachter<br />

JA. Verastem, Cambridge, MA<br />

Focal adhesion kinase (FAK) is a cytoplasmic tyrosine kinase that<br />

mediates signal transduction by integrins as well as growth factor<br />

receptors. FAK has been implicated in multiple steps of carcinogenesis<br />

including tumor initiation, growth and metastasis. Amplification and<br />

overexpression of FAK have been observed in aggressive human<br />

cancers including breast cancer. We have now observed that VS-4718,<br />

a selective FAK kinase inhibitor, exhibits preferential inhibitory<br />

effects on breast cancer stem cells.<br />

VS-4718 is a potent and selective FAK kinase inhibitor that blocks<br />

fibronectin-stimulated FAK autophosphorylation at Tyr397 at low<br />

nanomolar concentrations. To determine if FAK plays a role in the<br />

biology of breast cancer stem cells, VS-4718 was evaluated in a<br />

multitude of cancer stem cell assays both in vitro and in vivo. In<br />

parallel, treatment of SUM159 triple negative breast cancer cells<br />

in vitro with FAK shRNA inhibits tumorsphere formation. These<br />

data taken together indicate a role of FAK in breast cancer stem cell<br />

www.aacrjournals.org 549s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

renewal. Similarly, pre-treatment of SUM159 cells with VS-4718 in<br />

matrigel attenuated secondary tumorsphere formation. Furthermore,<br />

VS-4718 reduced the side population (SP) and the percentage of<br />

ALDEFLUOR+ cancer stem cells in SUM159 breast cancer cells in<br />

vitro. In direct contrast, standard-of-care agent paclitaxel increased<br />

the percentage of ALDEFLUOR+ cancer stem cells in all three assays.<br />

The in vivo effect of VS-4718 on cancer stem cells was evaluated<br />

in SUM159 and MDA-MB-231 human triple negative breast cancer<br />

xenograft models. Following systemic administration, VS-4718<br />

induced significant reduction of cancer stem cells in tumors as<br />

assessed by a decrease in ALDEFLUOR+ cells and tumorsphereforming<br />

efficiency relative to vehicle-treated tumors.<br />

In summary, our results indicate the importance of FAK in the selfrenewal<br />

of breast cancer stem cells in vitro and in vivo, and support the<br />

clinical development of the selective FAK inhibitor VS-4718 to target<br />

cancer stem cells for the treatment of triple negative breast cancer.<br />

P6-11-10<br />

IBL2001: Phase I/II study of a novel dose-dense schedule of oral<br />

indibulin for the treatment of metastastic breast cancer.<br />

Traina TA, Hudis C, Seidman A, Gajria D, Gonzalez J, Anthony SP,<br />

Smith DA, Chandler JC, Jac J, Youssoufian H, Korth CC, Barrett<br />

JA, Sun L, Norton L. Memorial Sloan-Kettering <strong>Cancer</strong> Center,<br />

New York, NY; Evergreen Hematology and Oncology, Spokane, WA;<br />

Compass Oncology, Vancouver, WA; The West Clinic, Memphis, TN;<br />

US Oncology Research, Woodlands, TX; ZIOPHARM Oncology, Inc.,<br />

Boston, MA; Harmon Hill Consulting, New York, NY<br />

Background: Indibulin (ZI0-301) is a novel, oral, synthetic small<br />

molecule microtubule inhibitor which binds tubulin at a different<br />

site than taxanes and vinca alkaloids. Preclinical data demonstrate<br />

indibulin does not interact with acetylated (neuronal) tubulins and in<br />

clinical studies has not exhibited the neurotoxicity associated with<br />

other tubulin binders. Indibulin has potent antitumor activity in human<br />

cancer cell lines, including multidrug-, taxane-, and vinblastineresistant.<br />

Norton-Simon modeling based on cell line data suggested<br />

that dose dense (dd) administration could optimize efficacy while<br />

limiting toxicity.<br />

Methods: Eligible patients (pts) have metastatic or unresectable<br />

locally advanced breast cancer, ECOG performance status ≤ 2,<br />

adequate organ function, measurable or nonmeasurable disease<br />

and any number of prior therapies. Uncontrolled gastrointestinal<br />

malabsorption syndrome and grade 2 or higher peripheral neuropathy<br />

are the principal exclusions. Adverse events (AEs) are graded by<br />

CTCAE v. 4.0. Objective disease status is evaluated according to<br />

RECIST 1.1. The primary objective of the phase (Ph) I portion of the<br />

study is to determine the maximum tolerated dose (MTD) of indibulin<br />

when given in dd fashion 5 days treatment, 9 days rest using standard<br />

3+3 dose escalation schema.<br />

Phase I Dose Cohorts<br />

Cohort Dose (mg) Q1D x 5<br />

1 25<br />

2 50<br />

3 100<br />

4 150<br />

5 200<br />

6 275<br />

7 350<br />

8 450<br />

The secondary objectives are to evaluate safety profile at various<br />

dosing levels, pharmacokinetics (PK) and preliminary activity of<br />

indibulin. Once the MTD is defined, a food effect cross- over group<br />

(N=12) will be enrolled. Two groups of 6 pts each will be treated<br />

in either the fed or fasted state during the first cycle. A subgroup of<br />

13 pts consisting of 12 pts from the food effect group plus the last<br />

pt from the MTD cohort will be evaluated for PFS at 4 months and<br />

will serve as the population for the first stage of a Simon two-stage<br />

design. If 4 or more out of 13 pts do not progress at 4 months, the Ph<br />

II portion of the study will be opened.<br />

Results: Twenty one pts (20 F, 1 M) have been enrolled to cohorts<br />

1 through 6 and the dose escalation is ongoing. Preliminary safety<br />

and efficacy data have been analyzed for 18 pts treated in cohorts 1<br />

through 5 and are presented henceforth. No DLT has been observed<br />

and no MTD has been reached. Median age 58 years (32-81). PS<br />

0=4, 1=12, 2=2. Median number of prior therapies 5 (1-12). Most<br />

frequent treatment-emergent AEs were: anorexia, constipation, cough,<br />

nausea (each in 39% pts); dyspnea (33%); fatigue, vomiting (each<br />

28%). There were no related grade 3-4 AEs. PK analysis revealed<br />

that indibulin plasma exposures increased approximately dose<br />

proportionally from 25 to 200 mg with C max of 165 ± 89 ng/mL and<br />

AUC 0-24 of 1411 ± 111 ng•h/mL at 200 mg. There were no objective<br />

responses. Stable disease was seen in 1 pt in the 150 mg cohort.<br />

Longest duration on-study was 4 months.<br />

Conclusions: Oral indibulin was well tolerated in the doses up to 200<br />

mg and the dose-proportional PK with lack of DLTs allows for further<br />

dose-escalation. Stable disease observed at sub-MTD dose may be a<br />

sign of activity in this heavily pre-treated population.<br />

P6-11-11<br />

Multistage Delivery of Paclitaxel: Increased Drug Stability and<br />

Sustained Release Results in Enhanced Efficacy in <strong>Breast</strong> <strong>Cancer</strong><br />

Blanco E, <strong>San</strong>gai T, Hsiao A, Ferrati S, Bai L, Liu X, Meric-Bernstam<br />

F, Ferrari M. The Methodist Hospital Research Institute, Houston, TX;<br />

The University of Texas MD Anderson <strong>Cancer</strong> Center, Houston, TX<br />

Background: A significant challenge for effectively treating cancer<br />

is overcoming biological barriers that reduce circulation times<br />

and increase degradation of possible treatments. We established<br />

an innovative approach to address this issue by embedding drugcontaining<br />

nanoparticles within the pores of a larger mesoporous<br />

silicon particle (MSP) in order to optimize site-specific localization<br />

and release of therapeutic agents. The objective is to develop<br />

a nanotherapeutic-based multistage platform for breast cancer<br />

treatment, wherein paclitaxel, a mitotic inhibitor used in the treatment<br />

of breast tumors, will be loaded into polymeric micelles, which in<br />

turn will be loaded within MSPs. We hypothesize that this nested<br />

incorporation of drugs within MSPs, combined with enhanced tumor<br />

transport, will result in a more pronounced and sustained antitumor<br />

effect.<br />

Materials and Methods: Micelles were assembled from amphiphilic<br />

block copolymers consisting of poly(ethylene glycol)-poly(εcaprolactone)<br />

(PEG-PCL, MW = 5k-5k). Nanoparticle size, zeta<br />

potential, and morphology was determined, and PTX loading and<br />

release kinetics from micelles analyzed. Drug-containing micelles<br />

were incorporated into MSPs by a previously established dry-loading<br />

method, wherein nanoparticles were incorporated into pores via<br />

capillary action. Loading of fluorescent micelles was used to verify<br />

loading within MSPs via fluorescence microscopy and flow cytometry<br />

analysis. Sulforhodamine B assays were used to evaluate the in vitro<br />

antitumor efficacy of the platform in MCF-7 and MDA-MB-468<br />

breast cancer cells. In vivo efficacy was evaluated in MDA-MB-468<br />

breast tumors in female nu/nu mice.<br />

Results: Resulting micelles had an average size of 20 nm, as confirmed<br />

through TEM, with paclitaxel loaded into micelles very effectively.<br />

Release kinetics showed that 50% of the drug was released within<br />

4 hours and 80% released within 24 hours. Loading of micelles into<br />

MSPs depended largely on electrostatic interactions, with micelles<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

550s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: General Session 6<br />

loading better within pores of MSPs displaying increased positive<br />

charge. Micelle loading into MSPs was successful as demonstrated<br />

by flow cytometry, and release was significantly retarded (< 30% of<br />

drug released over 4 d). Incubation of micelle-containing MSPs with<br />

breast cancer cells in vitro showed that MSPs could be internalized<br />

by cells, after which a sustained and delayed release of the payload<br />

was observed in cells. <strong>Breast</strong> tumors treated with MSPs demonstrated<br />

sustained tumor suppression (169 mm 3 compared to initial starting<br />

volume of 200 mm 3 ) at day 35 following a single injection. It is<br />

important to note that sustained tumor efficacy was achieved with<br />

nanoparticle and free drug formulations, however, with the caveat of<br />

repeated administrations.<br />

Discussion: A novel multistage approach to chemotherapy effectively<br />

allows a secondary payload to be loaded and preserved within the<br />

MSPs until reaching the tumor site. This prevents premature release<br />

of the drug and allows for a sustained release which may potentially<br />

result in fewer patient side effects. Future studies will involve loading<br />

of multiple nanoparticle types into MSPs and addition of targeting<br />

and diagnostic components.<br />

P6-11-12<br />

Nanoparticle-enhanced chemotherapeutics delivery in drugresistant<br />

triple-negative breast cancer<br />

van de Ven AL, Landis MD, Paskett LA, Meyn A, Frieboes HB, Chang<br />

JC, Ferrari M. The Methodist Hospital Research Institute, Houston,<br />

TX; University of Louisville, KY<br />

Chemotherapeutics delivery is generally poor in tumors characterized<br />

by rapid perfusion and low blood volume fraction. Systemically<br />

administered nanoparticles can be engineered to overcome abnormal<br />

flow conditions to act as intravascular drug depots for the localized<br />

delivery of high concentrations of chemotherapeutics. The feasibility<br />

of this approach was first demonstrated using melanoma, and is<br />

now being further investigated using well-characterized human<br />

triple-negative breast cancer biopsies implanted into mice. Intravital<br />

microscopy studies of human cancer-in-mice selected for differing<br />

vascular morphologies have yielded intriguing preliminary data<br />

regarding the role of tumor vascularity in drug and particle delivery.<br />

The first-pass perfusion of a 40kDa dextran tracer revealed that<br />

BCM-2665 tumors are perfused ∼6x more rapidly than BCM-4195<br />

tumors (11.3 ± 2.3s vs. 67.6 ± 11.0) and contain ∼30% lower volume<br />

fraction of blood (0.046 ± 0.011 vs. 0.062 ± 0.015). Interestingly, flow<br />

parameters that adversely impact drug accumulation appear to favor<br />

plateloid particle accumulation. BCM-2665 xenografts receiving<br />

an i.v. injection of 1000x400nm particles show ∼10x more particle<br />

accumulation than BCM-4195 tumors (24.8 ± 3.2 x103/mm3 vs. 3.5<br />

± 0.4 x103/mm3). The ability of these therapeutic particles to reach<br />

tumors appears to be primarily driven by flow-related parameters,<br />

which we characterize using a combination of intravital microscopy,<br />

computed tomography, and mathematical modeling. The total number<br />

of particles accumulated within a given tumor appears to be largely<br />

driven by the number of particles entering the tumor, since ∼65-70% of<br />

entering plateloid particles are retained by the tumor vasculature. This<br />

suggests that cytotoxic intravascular drug depots may be a promising<br />

strategy for increasing the efficiency of chemotherapeutics delivery<br />

to drug-resistant tumors and is the premise of ongoing therapeutic<br />

response studies. Clearly if more drugs can be delivered to the tumors,<br />

better outcomes can be expected for the patients.<br />

P6-11-13<br />

In vitro antineoplastic evaluation of rationally designed<br />

naphtoquinone-derived drugs in triple-negative breast cancer<br />

cell line<br />

Madeira KP, Daltoé RD, Herlinger AL, Guimarães IS, Allochio Filho<br />

JF, Teixeira SF, Valadão IC, Greco S, Rangel LBA. Federal University<br />

of Espirito <strong>San</strong>to, Brazil<br />

Background: <strong>Breast</strong> cancer (BC) comprises multiple diseases<br />

harboring different genetic alterations, which subtypes respond<br />

differently to treatment, and are associated to distinct clinical<br />

outcomes. Likewise, triple negative breast cancer (TNBCs) are a<br />

heterogeneous subgroup of BC, immunophenotypically negative<br />

for estrogen and progesterone receptor, and HER2, that account for<br />

10-15% of all invasive BC, affecting mostly African-American and<br />

Hispanic pre-menopausal women. Considering that TNBC have poor<br />

prognosis, and cannot be effectively treated with current targeted<br />

therapies, the discovery of new treatment options is imperative.<br />

Methods: Novel naphtoquinone-derived drugs were rationally<br />

designed to act through multiple pathways aiming the avoidance<br />

of drug-resistant phenotype acquisition by tumor cells, and were<br />

synthesized following high efficiency and low cost method. Drugs<br />

antineoplastic efficacy (AE) was accessed in claudin-low TNBC cell<br />

line, MDA-MB-231, through the evaluation of cellular metabolic<br />

viability (CMV) (MTT method). Drugs structures are protected by<br />

patent. Cells were cultured in RPMI media supplemented with 10%<br />

(v/v) FBS and antibiotics until subconfluence; then, 1.5x10 5 cells/<br />

well were subcultured for 72h prior to treatment with drugs (10 -4 , 10 -5 ,<br />

10 -6 , 10 -7 , and 10 -8 M). After 24h, CMV was assessed. Experiments<br />

in which the lineage was treated with cisplatin, doxorubicin or<br />

paclitaxel were run in parallel. The mean and standard-deviation<br />

of the absorbancies were used to calculate CMV and drugs IC 50<br />

(PrismaGraphPad version 5.1).<br />

Findings: We screened the AE of 43 novel naftoquinones-derived<br />

drugs in MDA-MB-231 lineage; seven have decreased its CMV by, at<br />

least, 50%, as: PIC1 (IC 50 1.15x10 -4 M; CMV decrease of 50%); PIC6<br />

(IC 50 4.24x10 -5 M; CMV decrease of 70%); PIC10 (IC 50 5.07x10 -5 M;<br />

CMV decrease of 70%); PIC 20 (IC 50 1.38x10 -5 M; CMV decrease<br />

of 90%); PIC21 (IC 50 5.00x10 -5 M; CMV decrease of 70%); S5 (IC 50<br />

7.26x10 -5 M; CMV decrease of 60%); M20 (IC 50 7.94x10 -5 M; CMV<br />

decrease of 60%). Their AE was significantly higher than that of<br />

cisplatin (IC 50 1.56x10 -4 M; CMV decrease < 10%), doxorubicin (IC 50<br />

1.76x10 -4 M; CMV decrease of 38%), and paclitaxel (IC 50 5.05x10 -7 M;<br />

CMV decrease of 20%).<br />

Interpretation: Altogether, our results present potential novel<br />

antineoplastic drugs to treat TNBC from a panel of in house<br />

rationally designed naftoquinones-derived compounds, developing<br />

an innovative and economically viable project.<br />

P6-11-14<br />

Post-hoc safety and tolerability assessment in patients receiving<br />

palliative radiation during treatment with eribulin mesylate for<br />

metastatic breast cancer<br />

Yardley DA, Vahdat L, Rege J, Cortés J, Wanders J, Twelves C. Sarah<br />

Cannon Research Institute, Nashville, TN; Weill Cornell Medical<br />

College, New York, NY; Eisai Inc, Woodcliff Lake, NJ; Vall d’Hebron<br />

University Hospital, Barcelona, Spain; European Knowledge Center,<br />

Eisai Ltd., Hatfield, Hertfordshire, United Kingdom; St James<br />

University Hospital, Leeds, United Kingdom<br />

Background: Management of advanced breast cancer frequently<br />

includes palliative radiation therapy (RT) and many chemotherapy<br />

agents are radiation-sensitizers. We assessed whether the safety profile<br />

www.aacrjournals.org 551s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

of eribulin differed between metastatic breast cancer (MBC) patients<br />

receiving eribulin alone and those who also received palliative RT.<br />

Methods: Two previous phase 2 and 3 eribulin trials (Studies 211<br />

and 305, respectively) enrolled a total of 794 patients with locally<br />

recurrent or MBC who had received 2-5 prior chemotherapy<br />

regimens. In both trials eribulin mesylate (1.4 mg/m 2 ) was given on<br />

Days 1 and 8 of a 21-day cycle and intercurrent palliative RT was<br />

permitted. In this post-hoc analysis, patient-level data were pooled<br />

for a descriptive comparison of adverse events (AEs) between the<br />

patients who received RT during their study treatment and those<br />

who did not. RT delivered within 3 weeks of initiation of eribulin or<br />

RT encompassing >30% of marrow was excluded. If palliative RT<br />

was utilized on study, the protocol requirements were as follows:<br />

indications included bone pain, bronchial obstruction, ulcerating skin<br />

lesions; the total field for palliative RT was not to involve >10% of<br />

total bone marrow; and the irradiated lesion was not to be used for<br />

tumor response assessment. During palliative RT, eribulin treatment<br />

was to be delayed and then resumed when the patient had recovered<br />

from any RT-associated toxicities.<br />

Results: Of the 794 patients (291 patients in Study 211 and 503 in<br />

Study 305) who received eribulin, 44 (5.5%) received palliative RT.<br />

Site of radiation therapy<br />

Frequency*<br />

Arm/shoulder 3<br />

Back/spine 19<br />

Brain (whole) 7<br />

Chest 3<br />

Head/neck 2<br />

Leg/hip 17<br />

Undisclosed 9<br />

7 patients received RT at multiple sites.<br />

The majority (26/44, 60%) received ≥10 cGys. Baseline demographics<br />

were similar between the RT (n=44) and no RT (n=750) subgroups,<br />

as was the use of concomitant medications. Six of 44 patients (14%)<br />

continued eribulin treatment during the palliative RT, contrary to<br />

protocol recommendation. The AE profiles over the course of the<br />

study were similar between subgroups. The most common events<br />

reported in both subgroups (RT vs no RT) were neutropenia (50% vs<br />

55%), alopecia (48% vs 51%), nausea (43% vs 40%), fatigue (39%<br />

vs 32%), and asthenia (27% vs 32%). There were no significant<br />

differences in Grade 3/4 treatment-emergent AEs, or other AEs (total<br />

AEs: 56.8% vs 58.9%). Potential localized events that were more<br />

frequent in the RT subgroup were bone pain (27% vs 15%), back<br />

pain (27% vs 10%), and musculoskeletal pain (14% vs 8%); many<br />

represent potential indications for RT. Half (22/44) of patients started<br />

RT 30 days after the start of eribulin treatment. Palliative RT was<br />

short in duration; 33 (75%) patients received ≤7 days, and 11 (25%)<br />

patients received 8-20 days. Six of the 44 irradiated patients (14%)<br />

had a skin-related toxicity, but most were alopecia (7/8 events) and<br />

all were associated with eribulin.<br />

Conclusions: This post-hoc subgroup analysis suggests palliative RT<br />

while receiving eribulin treatment does not appear to have an effect<br />

on the AE profile of eribulin. Further studies to define the optimal<br />

use of radiotherapy with eribulin are warranted.<br />

P6-12-01<br />

Adjuvant treatment in breast cancer patients aged 70 years or<br />

older during three years. A systematic review of patients charts.<br />

Nätterdal T, Klint L, Holmberg E, Gunnarsdottír KA, Linderholm BK.<br />

Institution of Medical Sciences, Sahlgrenska Univeristy Hospital,<br />

Gothenburg, Sweden; Sahlgrenska University Hospital, Gothenburg,<br />

Sweden; Karolinska Institute Science Park, Stockholm, Sweden<br />

Background and aims: In general, previous studies on adjuvant<br />

chemotherapy have not included the elderly breast cancer (BC)<br />

population. Our aim was to investigate the actual recommended<br />

adjuvant therapy for patients’ age ≥70 years in clinical routine. We<br />

also determined the proportion of patients that terminated therapy<br />

in correlation to documented BC relapses. Material and Methods:<br />

Charts from consecutive patients diagnosed with a primary stage I-III<br />

BC from 1 st January 2005 through 31 st December 2007, age ≥70 years<br />

were studied (n=358). Data regarding standard BC parameters at<br />

diagnosis, adherence to recommended adjuvant therapy, reasons for<br />

suspension of treatment and outcome were documented. Comparisons<br />

were performed with patients divided into three age groups (70-74;<br />

75-79; and ≥80 years) and between different treatment modalities<br />

(i.e. endocrine therapy; chemotherapy; trastuzumab). Results: Higher<br />

age was statistically significantly correlated with axillary metastases<br />

(70-74y n=30, [30%]; 75-79 n=41, [49%]; and ≥80y n=120 [42%],<br />

(p=0.011), while tumor size were equal (p=0.103). No difference was<br />

found regarding routine BC parameters as histological type (p=0.636)<br />

or grade (p=0.662); vascular invasion (p=0.681); ER (p=0.892);<br />

PgR (p=0.718) or HER2 status (p=0.542). A total of 286 patients<br />

were advised adjuvant systemic therapy; only two patients received<br />

trastuzumab. A higher proportion of the oldest patients ≥80 (n=32<br />

[22%]) were left without any treatment compared with 75-79y (n=17<br />

[17%] and 70-74y (n=13 [13%], (p=0.0002), this was explained by<br />

differences regarding chemotherapy. Data on compliance was lacking<br />

for five patients, however there was a trend not reaching statistically<br />

significance revealing a correlation between higher age and a more<br />

frequent treatment termination (p=0.061). Treatment suspension was<br />

for chemotherapy (70-74y n=21, [52%]; 75-79 n=17, [71%]; and<br />

≥80y n=3 [100%] and for ET (70-74y n=69, [25%]; 75-79 n=66,<br />

[32%]; and ≥80y n=104 [46%]. There was a statistically significant<br />

correlation between higher age and more breast cancer relapses<br />

(BCR) (p=0.015) and RFS (p=0.038). We found a higher proportion<br />

of BCR among patients terminating planned treatment (n=112; 27%<br />

BCR) compared with patients adherent to therapy (n=168; 8% BCR)<br />

(p=0.015). Patients not recommended any treatment (n=62) had an<br />

intermediate outcome with 16% BCR. This observation was found<br />

in all age groups; patients terminating therapy; 70-74y 21% BCR,<br />

75-79y 42% BCR and ≥80y 20% BCR respectively compared with<br />

patients’ adherent to therapy 70-74y 6% BCR, 75-79y 10% BCR and<br />

≥80y 7% respectively. Conclusions: Elderly patients are diagnosed<br />

with BC at a later stage and adjuvant chemotherapy, as well as,<br />

endocrine therapy prematurely terminated. Insufficient treatment<br />

was significantly associated with increased risk of relapse. Less toxic<br />

regimens should be carefully investigated.<br />

P6-12-02<br />

Use of cytochrome P450 interacting medications in the setting of<br />

adjuvant therapy for breast cancer.<br />

Njiaju UO, Kolesar JM, Johnston SA, Eickhoff JC, Osterby KR,<br />

Poggi LE, Tevaarwerk AJ, Millholland RJ, Oliver KA, Heideman<br />

JL, Wisinski KB. University of Wisconsin-Madison, WI; University of<br />

Wisconsin-Madison; University of Wisconsin Hospitals and Clinics<br />

Background: In the current era of personalized medicine, oral<br />

targeted therapies are increasingly used in cancer treatment. In<br />

breast cancer, oral anti-estrogen agents have historically been part<br />

of standard treatment for hormone receptor positive disease. More<br />

recently, other targeted agents have been introduced in the metastatic<br />

setting, and are being evaluated as adjuvant therapies. Many oral<br />

medications, including anticancer therapies, are metabolized by<br />

cytochrome P450 (CYP450) enzymes raising possibility of drug-drug<br />

interactions that may affect toxicities or breast cancer outcomes.<br />

We sought to evaluate concomitant CYP450 medication use among<br />

women seeing a medical oncologist to discuss adjuvant systemic<br />

therapy for breast cancer.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

552s<br />

<strong>Cancer</strong> Research


December 4-8, 2012 <strong>Abstract</strong>s: General Session 6<br />

Methods: We performed an electronic medical record database<br />

extraction. Adult women diagnosed with breast cancer from 1/2008-<br />

7/2011 were identified from the University of Wisconsin Hospital and<br />

Clinics <strong>Cancer</strong> Registry. Medication lists were extracted from the first<br />

encounter with a medical oncologist after the initial breast cancer<br />

diagnosis. Non-systemic medications were excluded. Cytochrome<br />

P450 (CYP450) enzyme-interacting medications were categorized as<br />

inhibitors, inducers, and/or substrates of specific enzymes including<br />

CYP1A2, CYP2A6, CYP2B6, CYP2C19, CYP2C8, CYP2C9,<br />

CYP2D6, CYP2E1, and CYP3A4. CYP450 inhibitors and inducers<br />

were further characterized as strong, moderate or weak acting.<br />

Results: A total of 455 women with non-metastatic breast cancer<br />

were identified. Mean age was 56.6 years (range of 23-90) and<br />

413 (91%) were Caucasian. Polypharmacy, defined as use of 3-5<br />

medications, was seen in 123 (27.0%) women. A total of 236 (51.9%)<br />

women were on 0-4; 109 (24.0%) on 5-10; and 13 (2.9%) on > 10<br />

medications at the time of first encounter with a medical oncologist<br />

after a breast cancer diagnosis. 23 (5.05%) women were on strong<br />

CYP450 enzyme inhibitors while 72 (15.8%) were on strong inducers.<br />

CYP450 enzymes most commonly affected were CYP3A4, CYP2C9,<br />

and CYP2D6. Among medications taken on a fixed schedule,<br />

levothyroxine and simvastatin were the most commonly used, while<br />

simvastatin and ranitidine were the most common CYP450 interacting<br />

medications. Further classification of potential CYP450 interactions<br />

is ongoing.<br />

Conclusions: A significant proportion of patients were on one or more<br />

CYP450 interacting medications in the setting of adjuvant therapy for<br />

breast cancer. Given the number of new oral cancer agents that are<br />

also CYP450 interacting, the potential for drug interactions should<br />

be recognized and appropriate management strategies implemented.<br />

P6-12-03<br />

Effects of high dose of bisphosphonate therapy on bone<br />

microarchitecture of the peripheral skeleton in women with early<br />

stage breast cancer<br />

Shao T, Shane ES, McMahon D, Crew KD, Kalinsky K, Maurer M,<br />

Brown M, Gralow JR, Hershman DL. Beth Israel Medical Center,<br />

Continuum <strong>Cancer</strong> Centers of New York, New York, NY; Columbia<br />

University Medical Center, New York, NY; University of Washington/<br />

Seattle <strong>Cancer</strong> Care Alliance, Seattle, WA<br />

Background: Randomized studies investigating adjuvant<br />

bisphosphonates in women with breast cancer are ongoing. While<br />

bisphosphonates would be expected to prevent the deterioration of<br />

bone microarchitecture that accompanies hormone or chemotherapy,<br />

complete suppression of osteoclast activity for prolonged periods<br />

of time can decrease repair of micro-cracks, and possibly lead to<br />

decreased bone strength. While bone strength is governed by the<br />

amount of bone present, trabecular and cortical components of bone<br />

microarchitecture also contribute independently to bone strength.<br />

We aimed to characterize the effects of long-term bisphosphonates<br />

on bone microarchitecture in women with breast cancer using highresolution<br />

peripheral quantitative computed tomography (HR-pQCT)<br />

of the distal radius and tibia.<br />

Methods: We conducted a cross-sectional study involving early stage<br />

breast cancer patients treated with bisphosphonates on the S0307<br />

clinical trial. Women were randomized to receive zoledronic acid,<br />

oral clodronate or oral ibandronate in doses far higher than those<br />

used in osteoporosis treatment as per protocol. After 18-36 months of<br />

bisphosphonate therapy, participates underwent a one-time evaluation<br />

of areal bone mineral density (aBMD) of the 1/3 radius, lumbar spine,<br />

and hip by dual energy x-ray absorptiometry (DXA), and cortical and<br />

trabecular volumetric BMD (vBMD) and trabecular microarchitecture<br />

of the radius and tibia by HR-pQCT. HR-pQCT measurements were<br />

compared to healthy young premenopausal women and age-matched<br />

Caucasian women.<br />

Results: Baseline characteristics of the 12 enrolled patients: median<br />

age of 53 (range 40-67); white/Hispanics 7/5; pre/postmenopausal<br />

4/8; mean body mass index 28.7 kg/m2 (20.9-34.8); average time<br />

on bisphosphonates 20 months (18-30); zoledronic acid/clodronate/<br />

ibandronate 5/6/1. The median aBMD DXA T-score of the 1/3<br />

radius, lumbar spine and total hip were normal at +0.3, +0.1, and<br />

+0.2, respectively. Mean total, cortical, and trabecular vBMD of the<br />

radius as measured by HR-pQCT were 330±71, 905±55, and 146±35<br />

mg hydroxyapatite/cm3, respectively. Mean cortical thickness was<br />

0.803±0.170 mm, and mean trabecular number was 1.9±0.2. Mean<br />

total, cortical, and trabecular vBMD of the tibia were 285±54, 880±54,<br />

and 150±38 mg hydroxyapatite/cm3, respectively. Mean cortical<br />

thickness was 1.135±0.264 mm, and mean trabecular number was<br />

1.7±0.3. There were no statistically significant differences between<br />

study group and each control. However, results were more similar to<br />

healthy young premenopausal control than the age-matched control.<br />

Conclusion: Women on long-term bisphosphonate therapy for breast<br />

cancer had normal aBMD by DXA and normal cortical and trabecular<br />

vBMD, cortical thickness and trabecular number at the peripheral<br />

skeleton compared to healthy young women and age-matched women.<br />

This preliminary data is reassuring for cancer survivors if benefits<br />

from this therapy are established in the adjuvant setting.<br />

Radius Study Group Mean (SD) Premenopausal Control Mean (SD)<br />

vBMD (mg HA/cm3)<br />

Total 330 (71) 330 (57)<br />

Cortical 905 (55) 904 (44)<br />

Trabecular 146 (35) 160 (33)<br />

Cortical thickness (mm) 0.803 (0.170) 0.804 (0.149)<br />

Trabecular number 1.9 (0.2) 1.7 (0.2)<br />

P6-13-01<br />

Lyso-thermosensitive liposomal doxorubicin + local hyperthermia<br />

for radiation-pretreated chest wall recurrence<br />

Formenti S, Rugo H, Myerson R, Diederich C, Straube W, O’Conner<br />

B, Matzkowitz AJ, Goodman RL, Muggia F. New York University<br />

<strong>Cancer</strong> Institute, New York, NY; UC <strong>San</strong> Francisco, <strong>San</strong> Francisco,<br />

CA; Siteman <strong>Cancer</strong> Center, Saint Louis, MO; Rhode Island Hospital,<br />

Providence, RI; New Hope <strong>Cancer</strong> Center, Hudson, FL; Saint<br />

Barnabas Medical Center, Livingston, NJ<br />

Background: Chest wall recurrence (CWR) causes much morbidity<br />

when not controlled by local measures. Lyso-thermosensitive<br />

liposomal doxorubicin (LTLD) provides accelerated release of<br />

doxorubicin (Dox) at ≥ 39° C, and is being studied for CWR based<br />

on the pioneering thermochemotherapy work of Dewhirst and<br />

colleagues at Duke. We now report on its tolerance, pharmacology,<br />

and preliminary antitumor effects in a dose-escalation study<br />

combined with local hyperthermia (LH) for radiation-refractory<br />

CWR. LH selectively increases liposomal permeability in tumor<br />

microvasculature, and promotes release of Dox from LTLD, and Dox<br />

tumor uptake, in addition to LH anti-tumor effects.<br />

Methods: This phase I study entered patients (pts) with CWRs <<br />

3 cm deep failing all standard Tx including surgery, radiation, and<br />

chemotherapy: pts received up to 6 LTLD/LH treatments every 21<br />

days at a starting dose of 40 mg/m 2 (cohort 1) and escalated to 50 mg/<br />

m 2 (cohort 2). LTLD was infused IV over 30 minutes (min); followed<br />

within 30 min by microwave or ultrasound LH. The thermal dose<br />

goal was 40°C-42°C for 60 min. Pharmacokinetic samples for total<br />

www.aacrjournals.org 553s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

plasma Dox and doxorubicinol (Doxol) were taken at 0.5, 5, 10 and<br />

24 hours after starting infusion. Left ventricular ejection fraction was<br />

monitored every other cycle.<br />

Results: Eleven pts with a median of 4 prior chemotherapy regimens<br />

(range 2 — 12) were enrolled; all but one had prior anthracycline<br />

(AC). All pts received ≥ 2 cycles. The within subject variability in<br />

Dox and Doxol exposure was small with mean Cycle 2 vs Cycle 1<br />

ratios ranging from 0.99 to 1.06.<br />

Cmax/dose (ng/ml)/(mg/m 2 ) Cycle 1 Cycle 2<br />

Dox 499.82 512.00<br />

Doxol 0.46 0.45<br />

AUClast/dose ((ng*hr/ml)/(mg/m 2 )<br />

Dox 1338.12 1381.82<br />

Doxol 7.96 8.04<br />

Grade 3 and 4 toxicities included reversible neutropenia in 17 (40.5%)<br />

and one case (each) of mucositis (grade 1), chest wall thermal burn,<br />

and chest wall cellulitis (both grade 4); these occurred in only ≥ 5%<br />

of 42 cycles given. No cardiomyopathy or hand-foot toxicity occurred.<br />

The rate of clinically-significant (≥ 6 point) QoL improvement on<br />

the FACT-B after 2 cycles was 54.5% (95% CI: 25.1% - 83.9%),<br />

including 1 lasting > 3 months. The local objective response rate was<br />

encouraging: 45.5% (95% CI: 16.1% - 74.9%), with 1 complete and<br />

4 partial local responses.<br />

Conclusion: LTLD + LH is safe in CWR after prior radiation<br />

and doxorubicin. A phase II study is planned for pharmacologic<br />

evaluations and to add pts with less or no anthracycline treatment.<br />

P6-13-02<br />

Overcoming therapy resistance of metastatic breast cancer by<br />

enhanced tumor delivery of polymeric doxorubicin<br />

Shen H, Xu R, Mai J, Huang Y, Ferrari M. The Methodist Hospital<br />

Research Institute, Houston, TX<br />

Metastatic breast cancers are treated with chemotherapy drugs in<br />

clinic. However, patients usually develop therapy resistance quickly<br />

due to intrinsic and acquired mechanisms. Poor drug delivery to<br />

tumor cells may also contribute to therapy resistance. To address<br />

both biological and mass transport barriers to effective therapy,<br />

we developed a new doxorubicin-based formulation, polymeric<br />

doxorubicin (pDox), to be delivered using a porous silicon drug<br />

carrier, the multistage vector (MSV). We show the novel, rationally<br />

designed MSV/pDox drug enriches in tumor tissues and has<br />

considerable therapeutic efficacy in two animal models of triple<br />

negative breast cancer lung metastasis, conferring a significant<br />

survival advantage to animals treated with MSV/pDox over all other<br />

control groups (80% survival for the MSV/pDox group at 24 weeks<br />

post treatment vs. 0% survival for all others, log-rank test, p


December 4-8, 2012 <strong>Abstract</strong>s: General Session 6<br />

P6-14-01<br />

Estrogen/progestogen use after <strong>Breast</strong> <strong>Cancer</strong> – a long-term<br />

follow-up of the Stockholm randomized trial.<br />

Fahlén MC, Fornander T, Johansson H, Rutqvist L-E, Wilking N,<br />

von Schoultz E. Capio St Görans Hospital, Stockholm, Sweden;<br />

Karolinska Institutet, Stockholm, Sweden<br />

Background. The management of hormonal deficiency symptoms in<br />

breast cancer survivors is still an unsolved problem. While several<br />

studies have shown that menopausal hormone therapy (MHT) may<br />

increase the risk of breast cancer in healthy women, its effects on<br />

recurrence for women already affected by breast cancer is unclear.<br />

Observational studies have even suggested that there may be a<br />

decreased recurrence rate for women using MHT. The few clinical<br />

trials in this field have all been closed preterm.<br />

Methods. The Stockholm trial was started in 1997 as a randomized<br />

open study with two parallel groups. Postmenopausal women who had<br />

undergone primary surgery for breast cancer were asked to participate.<br />

After inclusion they were randomized to either treatment with MHT<br />

(n=188) or not (n=190). The study was designed to minimize the<br />

dose of progestogen in the MHT arm. The trial was stopped in 2003<br />

when another Swedish study (HABITS), with a similar arrangement,<br />

reported increased recurrence. However the Stockholm material<br />

showed no excess risk after 4 years of follow-up. A long-term followup<br />

has now been performed.<br />

The aim. We wanted to determine if it was possible to treat the<br />

menopausal symptoms experienced by women with breast cancer<br />

and on breast cancer medication, without compromising their safety.<br />

Statistical method. The median follow-up time was calculated<br />

using the reversed Kaplan-Meyer technique. Proportional hazards<br />

regression was used to model event specific hazards. Results from the<br />

regression models were presented as hazard ratios together with 95%<br />

confidence intervals. Differences in time to specific event distributions<br />

were tested using the log-rank test. All analyses were based on the<br />

intention-to-treat principle.<br />

Findings. The mean duration of hormone use in the MHT group was<br />

2.6±1.2 years. After a median of 10.8 years of follow-up, there was no<br />

difference in new breast cancer events: 60 in the MHT group versus 48<br />

among controls (HR=1.3;95%CI=0.9-1.9). Among women on MHT,<br />

11 had local recurrence and 12 distant metastases versus 15 and 12 for<br />

the controls. There were 14 contra-lateral breast cancers in the MHT<br />

group and 4 in the control group (HR=3.6;95%CI=1.2-10.9;p=0.013).<br />

There was no difference in overall mortality between the groups. The<br />

total number of deaths in the menopausal hormone therapy group<br />

was 19 (10 from breast cancer) and in the control group 18 (11 from<br />

breast cancer). Also there was no difference in the occurrence of new<br />

primary malignancies other than contra-lateral breast cancer – 21 in<br />

the MHT group and 16 in the non MHT group.<br />

Interpretation. The number of new events did not differ between the<br />

groups, in contrast to previous reports. The increased recurrence<br />

in HABITS has been attributed to this trials higher progestogen<br />

exposure. As both trials were prematurely closed, data do not allow<br />

firm conclusions. In neither of the studies there were any increased<br />

mortality from breast cancer or other causes in the MHT-group.<br />

Current guidelines typically consider MHT contraindicated in breast<br />

cancer survivors. Our findings suggest that, in some women symptom<br />

relief may outweigh the potential risks of MHT.<br />

P6-14-02<br />

Withdrawn by Author<br />

P6-14-03<br />

Statin and aspirin use is not associated with a reduced risk of<br />

VTE’s in breast cancer patients<br />

Shai A, Rennert HS, Ballan Haj M, Lavie O, Steiner M, Rennert<br />

G. Lin Medical Center, Haifa, Israel; Carmel Lady Davis Medical<br />

Center, Haifa, Israel<br />

Introduction: Statins and aspirin have been shown to reduce the risk<br />

of venous thromboembolic events (VTE’s) in the general population<br />

in randomized trials.<br />

Aim: To assess whether statins and aspirin reduce the incidence of<br />

deep vein thrombosis and pulmonary embolism in patients diagnosed<br />

with breast cancer.<br />

Methods: The <strong>Breast</strong> <strong>Cancer</strong> in Northern Israel Study (BCINIS) is an<br />

on-going population-based case-control study of consecutive breast<br />

cancer cases and matched controls diagnosed in the northern part of<br />

Israel since 2000.<br />

Only cases insured by the Clalit Health Services (64%) were included<br />

in this analysis. Data on medication use and VTE were extracted<br />

from the computerized database. Patients taking Warfarin or LMWH<br />

were excluded.<br />

Statistical analysis was performed using SPSS (v 18). Use of<br />

medications was analyzed as a time dependent covariate in a Cox<br />

Regression model.<br />

Results: Of 3552 patients 261 (7.3%) had a VTE during a median<br />

follow up of 4.7 years.<br />

In a multivariate analysis age (HR 1.03, 95% CI 1.02-1.04), BMI (HR<br />

2.12, 95% CI 1.43-3.14 for BMI >30), chemotherapy (HR 3.87, 95%<br />

CI 2.50-5.98) and metastatic disease (HR 2.27, 95% CI 1.10-6.53)<br />

were associated with an increased risk for VTE’s.<br />

Statins, mainly simvastatin and pravastatin, were used by 55.7%<br />

of the patients, and 45.7% used aspirin. Neither statins nor aspirin<br />

were associated with a significantly reduced risk for a VTE. After<br />

controlling for age, BMI, stage, chemotherapy and tamoxifen use the<br />

HR for statins was 0.80, CI 0.56-1.13, p=0.2, and the HR for aspirin<br />

was 1.004, CI 0.74-1.76, p=0.98.<br />

Conclusion: In contrast to the findings in the general population, statin<br />

and aspirin use were not associated with a reduced risk for VTE’s in<br />

our cohort of breast cancer patients. Our results might be explained<br />

by an alternate mechanism of VTE formation in breast cancer and the<br />

use of low potency statins (simvastatin and pravastatin) in this cohort.<br />

P6-14-04<br />

Rosehip extracts prevent triple negative breast cancer cell<br />

proliferation by regulation the phosphorylation of p70S6 Kinase.<br />

Coburn TL, Cagle PD, Shofoluwe AI, Martin PM. North Carolina<br />

Agricultural and Technical State University, Greensboro, NC<br />

Triple Negative <strong>Breast</strong> <strong>Cancer</strong> (TNBC) is an aggressive form of<br />

breast cancer, characterized by its lack of the human epidermal<br />

growth factor receptor-2 (HER-2), the estrogen receptor (ER), and<br />

the progesterone receptor (PR). Due to the lack of these hormone<br />

receptors, TNBC has abnormal therapeutic results; leading to the<br />

need for unique treatment methods. TNBC has a partial response<br />

to chemotherapy, which is associated with the lack of clinically<br />

established targeted therapies. Plant extracts have shown to be useful<br />

as chemopreventive agents due to their disruption of pro-survival<br />

and proliferative mechanisms on cancer cells. Rosehip extracts have<br />

shown potential anti-proliferative activity against cancer cells in vitro.<br />

www.aacrjournals.org 555s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

This study investigated the effect of rosehip extracts in TNBC cell<br />

lines (HCC1806 and MB157) and basal-like breast cancer cell lines<br />

(HCC70 and HCC1500). To determine the inhibitory role of rosehip<br />

extracts in cell proliferation, MTT assays were conducted, following<br />

treatment with varying concentrations of rosehip extracts (1mg/ml,<br />

250ug/ml, 25ug/ml, and 25ng/ml) for 48 hours. The data suggests<br />

that rosehip extracts, can significantly decrease cell proliferation in<br />

all the breast cancer cell lines assayed. Preliminary studies suggest<br />

that this is accomplished without toxic effects. Western blot analysis<br />

was used to investigate which signaling pathway rosehip extracts<br />

were affecting. Western Blot analysis suggests that rosehip extracts<br />

are inhibiting p70 S6 kinase phosphorylation and its ability to act as<br />

a kinase for S6 ribosomal protein. Based on these findings, rosehip<br />

extracts prevent cell proliferation by interfering with the p70 S6<br />

kinase signaling.<br />

P6-14-05<br />

Impact of breast cancer CME: Physician practice pattern,<br />

knowledge, and competence assessments<br />

Haas M, Heintz A, Stacy T. Educational Concepts Group, LLC,<br />

Atlanta, GA<br />

Background<br />

Several scientific advances for women with breast cancer have taken<br />

place in the last decade. The National <strong>Cancer</strong> Institute reports that<br />

approximately 2.5 million women with a history of breast cancer are<br />

alive today due to advances in screening, detection, treatment, and<br />

supportive care.<br />

Due to the volume and pace of scientific advances within the<br />

specialty of oncology, the timely dissemination of clinical trial<br />

results and discussions regarding the appropriate incorporation of<br />

advances into therapeutic strategy is necessary to provide the highest<br />

quality patient care. Furthermore, increasing awareness in the breast<br />

cancer community on similarities and differences amongst practice<br />

management strategies helps to identify individual practice patterns<br />

and benchmark individual knowledge and competence against one’s<br />

professional peers.<br />

Methods<br />

Between November 2011 and November 2012, educational outcomes<br />

assessments were gathered during 95 independent continuing medical<br />

education (CME) activities held within community and academic<br />

institutions and practices across the United States. The activities<br />

educated oncology healthcare providers on recent updates to the<br />

development and integration of new therapies in the management of<br />

ER+, HER2+ and/or HER2- breast cancer.<br />

Participants were asked a series of case-based questions via an<br />

audience response system to assess baseline knowledge, competence,<br />

and identify practice patterns. Assessments were repeated post a<br />

1-hour independent CME certified activity, with an additional 6-week<br />

electronic follow-up. Barriers to implementing the information<br />

learned were captured at 6-week follow-up. Educational gaps were<br />

identified through documentation and comparison of current best<br />

practices versus actual participant responses.<br />

Results<br />

To date, a total of 568 healthcare providers have participated in the<br />

initiative of which 326 (57%) were physicians. Number of years in<br />

practice included < 10 years (22%), 10-20 years (50%), 21-30 years<br />

(13%), > 30 years (15%). The number of breast cancer patients seen<br />

per month included ≤ 10 (45%), 11-20 (17%), 21-30 (15%), 31-40<br />

(8%), and > 40 (15%). This represents a potential impact of 3,706<br />

patient lives.<br />

Provider competency in applying recent literature to the patient care<br />

setting was both assessed and measured. Results of patient therapy<br />

selection in the first-line and relapsed MBC setting will be reported.<br />

Knowledge and awareness regarding recent clinical trials as well as<br />

self-reported competence in discussing the mechanism of action,<br />

efficacy, and safety data from recent clinical trials will be presented.<br />

Participants reported a number of barriers to applying data learned<br />

at the activities into clinical practice. Responses included lack of<br />

reimbursement (24%), treatment side effects (24%), newness of<br />

treatment data (40%), perceived efficacy (8%) and treatment not<br />

commercially available (4%).<br />

Conclusion<br />

The results suggest the education of providers on the most currently<br />

available clinical trial results and peer discussion of how to optimize<br />

translation of this information into patient care increases breast<br />

cancer provider knowledge and competence. <strong>Breast</strong> cancer CME<br />

plays an important role in improving the care of patients and practice<br />

of medicine.<br />

OT1-1-01<br />

A phase II study of neoadjuvant epirubicin/cyclophosphamide<br />

(EC) followed by weekly nanoparticle albumin-bound paclitaxel<br />

with or without trastuzumab for node-positive breast cancer<br />

Hirano A, Hattori A, Kamimura M, Ogura K, Kim N, Setoguchi<br />

Y, Okubo F, Inoue H, Jibiki N, Miyamoto R, Kinoshita J, Kimura<br />

K, Fujibayashi M, Shimizu T. Tokyo Women’s Medical University,<br />

Medical Center East, Tokyo, Japan; Tokyo Women’s Medical<br />

University, Yachiyo Medical Center, Yachiyo, Japan<br />

Background<br />

Paclitaxel is considered standard treatment for metastatic breast cancer<br />

(MBC) and is often used as adjuvant and neoadjuvant chemotherapy<br />

for patients with early-stage disease. Conventional paclitaxel requires<br />

solvents such as polyoxyethylated castor oil; however, such solvents<br />

are associated with toxicity including peripheral neuropathy and<br />

hypersensitivity reaction. Moreover, the use of the drug requires<br />

special tubing and in-line filters. Therefore, nanoparticle albuminbound<br />

paclitaxel (nab-PTX) requiring no solvent has been developed.<br />

Nab-PTX was effective in patients with MBC and as a neoadjuvant<br />

therapy. A comparison between weekly and triweekly nab-PTX<br />

suggested that weekly nab-PTX was superior in progression-free<br />

survival.<br />

Trial design<br />

This is a phase II trial to evaluate the efficacy and toxicity of<br />

neoadjuvant epirubicin/cyclophosphamide (EC) (epirubicin/<br />

cyclophosphamide) followed by weekly nab-PTX with or without<br />

trastuzumab for node-positive breast cancer. Patients receive four<br />

cycles of epirubicin (90 mg/m 2 ) and cyclophosphamide (600 mg/<br />

m 2 ) every 3 weeks, followed by four cycles of nab-PTX (125 mg/m 2 )<br />

on days 1, 8 and 15 in a 28-day cycle. Fifteen cycles of trastuzumab<br />

(2 mg/kg, loading 4 mg/kg) are added to the nab-PTX regimen in<br />

HER2-positive patients every week.<br />

Eligibility criteria<br />

Patients with histologically diagnosed invasive breast cancer based<br />

on a core needle biopsy of the T1-4 N1-3 without previous operation<br />

or chemotherapy are included in this trial. Eligible patients are aged<br />

between 20 years and 70 years with a performance status of 0 to 2<br />

and adequate organ functions.<br />

Specific aims<br />

The primary endpoint is the pathologic complete response (pCR) rate<br />

in the breast and axilla, and the secondary endpoints are the breast<br />

conserving rate, toxicities, feasibility and overall survival.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

556s<br />

<strong>Cancer</strong> Research


December 4-8, 2012<br />

<strong>Abstract</strong>s: Poster Session OT1<br />

Statistical methods<br />

The sample size was calculated using the Simon method, with a type<br />

I error of 10% and a study power of 80%.<br />

1. HER2-negative patients<br />

The expected rate of pCR was 25% and the required number of<br />

patients was estimated to be 33.<br />

2. HER2-positive patients<br />

The expected rate of pCR was 50%, and the required number of<br />

patients was estimated to be 21.<br />

Present and target accrual<br />

Patient accrual within two medical centers started in April 2011<br />

with 20 patients being on study to date (2012, June 12). A total of<br />

56 patients (22 are HER2-positive and 34 are HER2-negative) are<br />

planned to be enrolled in the trial.<br />

OT1-1-02<br />

A single-arm phase IIIb study of pertuzumab and trastuzumab<br />

with a taxane as first-line therapy for patients with HER2-positive<br />

advanced breast cancer (PERUSE)<br />

Bachelot T, Ciruelos E, Peretz-Yablonski T, Schneeweiss A, Puglisi<br />

F, Mitchell L, Dünne A, Miles D. Centre Leon Bérard, Lyon, France;<br />

Hospital Universitario 12 Octubre, Madrid, Spain; Hadassah-<br />

Hebrew University Medical Center, Jerusalem, Israel; National<br />

Center for Tumor Diseases, University-Hospital Heidelberg,<br />

Germany; University Hospital of Udine, Italy; F. Hoffmann-La Roche,<br />

Basel, Switzerland; Mount Vernon <strong>Cancer</strong> Centre, Mount Vernon<br />

Hospital, Middlesex, United Kingdom<br />

Background: Pertuzumab (P), a humanized monoclonal antibody,<br />

inhibits signaling downstream of HER2 by binding to the dimerization<br />

domain of the receptor and preventing heterodimerization with<br />

other HER family members. The epitope recognized by P is distinct<br />

from that bound by trastuzumab (H) and so their complementary<br />

mechanisms of action result in a more comprehensive HER2 blockade.<br />

Data from the phase III trial CLEOPATRA showed significantly<br />

improved PFS in patients (pts) receiving P + H + docetaxel compared<br />

with H + docetaxel + placebo as first-line treatment for HER2-positive<br />

metastatic breast cancer (BC).<br />

Trial design: PERUSE is a phase IIIb, multicenter, open-label,<br />

single-arm study in pts with HER2-positive metastatic or locally<br />

recurrent BC who have not been treated with systemic nonhormonal<br />

anticancer therapy for metastatic cancer. Pts will receive, P: 840 mg<br />

initial dose, 420 mg q3w IV; H: 8 mg/kg initial dose, 6 mg/kg q3w<br />

IV; taxane: docetaxel, paclitaxel, or nab-paclitaxel according to local<br />

guidelines. Treatment will be administered until disease progression<br />

or unacceptable toxicity. A planned protocol amendment will allow<br />

hormone receptor-positive pts to receive endocrine therapy alongside<br />

P+H after completion of taxane therapy, in line with clinical practice.<br />

Eligibility criteria: At baseline, pts must have an LVEF of ≥50%,<br />

an ECOG PS of 0, 1, or 2, a disease-free interval of ≥6 months, and<br />

must not have received prior anti-HER2 agents for the treatment of<br />

metastatic BC. Prior H and/or lapatinib in the (neo)adjuvant setting<br />

is permitted, providing there was no disease progression during<br />

treatment. Pts must not have experienced other malignancies within<br />

the last 5 yrs other than carcinoma in situ of the cervix or basal cell<br />

carcinoma. There must be no clinical or radiographic evidence of CNS<br />

metastases or clinically significant cardiovascular disease.<br />

Specific aims: As H was not widely available in the (neo)adjuvant<br />

setting prior to CLEOPATRA recruitment, a relatively low proportion<br />

of pts in CLEOPATRA had previously received H. PERUSE will<br />

assess the safety and tolerability of P+H + choice of taxane as first-line<br />

therapy for pts with HER2-positive metastatic or locally advanced<br />

BC in a pt population likely to have experienced wider exposure to<br />

prior H therapy.<br />

Statistical methods: The primary endpoints of the PERUSE study<br />

are safety and tolerability. Secondary endpoints include PFS, OS,<br />

ORR, CBR, duration of response, time to response and QoL. The final<br />

analysis will be performed when 1500 pts have been followed up for<br />

at least 12 months after the last pt receives last study treatment unless<br />

they have been lost to follow-up, withdrawn consent, or died, or if the<br />

study is prematurely terminated by the sponsor. Safety analyses are<br />

planned after enrollment of ∼350, 700, and 1000 pts. Additionally,<br />

a data and safety monitoring board will review safety data after ∼50<br />

pts have been enrolled and then every 6 months.<br />

Current and target accrual: Enrollment of the first pt is expected in<br />

June 2012 with a total of 1500 pts planned to be recruited.<br />

OT1-1-03<br />

PERSEPHONE: Duration of Trastuzumab with Chemotherapy<br />

in women with HER2 positive early breast cancer.<br />

Earl HM, Cameron DA, Miles D, Wardley AM, Ogburn ERM, Vallier<br />

A-L, Loi S, Higgins HB, Hiller L, Dunn JA. University of Cambridge,<br />

Cambridge, United Kingdom; NIHR Cambridge Biomedical Research<br />

Centre, Cambridge, United Kingdom; Edinburgh University,<br />

Edinburgh, United Kingdom; Mount Vernon <strong>Cancer</strong> Centre,<br />

Middlesex, United Kingdom; The Christie Hospital, Manchester,<br />

United Kingdom; Warwick Clinical Trials Unit, University of<br />

Warwick, Coventry, United Kingdom; Cambridge <strong>Cancer</strong> Trials<br />

Centre, Cambridge<br />

Background: Persephone is a phase III randomised controlled trial<br />

comparing six months of trastuzumab to the standard 12 month<br />

duration in patients with HER2 positive early breast cancer in respect<br />

of disease free survival, safety and cost-effectiveness. A Persephone<br />

sister study, the PHARE trial run by the National Institute for <strong>Cancer</strong>,<br />

successfully closed to recruitment in 2010. A prospective metaanalysis<br />

is planned once each trial has reported individually.<br />

Methods: A total of 4000 patients will be randomised into each of the<br />

two treatment groups. Eligible participants must be Her2 positive with<br />

a histological diagnosis of invasive breast cancer and no evidence<br />

of metastatic disease. Patients will receive neo-adjuvant or adjuvant<br />

chemotherapy and have no previous diagnosis of malignancy unless<br />

managed by surgical treatment only and disease-free for 10 years.<br />

Patients can be randomised at any time prior to receiving their 10th<br />

cycle of trastuzumab.<br />

The power calculations assume that the disease-free survival (DFS)<br />

of the standard treatment of 12 months trastuzumab will be 80% at<br />

4 years. On this basis, with 5% 1-sided significance and 85% power,<br />

a trial randomising 2000 in each arm will have the ability to prove<br />

non-inferiority of the experimental arm defining non-inferiority as<br />

‘no worse than 3%’ below the control arm 4 year DFS. Primary<br />

outcome is disease-free survival non-inferiority (equivalence) of 6<br />

months trastuzumab compared with 12 months in women with early<br />

breast cancer. Secondary outcomes are overall survival non-inferiority<br />

(equivalence); expected incremental cost effectiveness; cardiology<br />

function and analysis of predictive factors for development of cardiac<br />

damage. Two mandatory sub-studies are: Tumour block collection to<br />

discover molecular predictors of survival with respect to duration of<br />

trastuzumab treatment and blood sample collection, used to discover<br />

single nucleotide polymorphisms (SNPs) as genetic/pharmaco-genetic<br />

determinants of prognosis, toxicity and treatment outcome. A third<br />

optional sub-study is the quality of life questionnaires.<br />

www.aacrjournals.org 557s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Results: Persephone opened to recruitment in October 2007. To date,<br />

2152 patients (54%) of its total have been randomised from 147 UK<br />

sites. Recruitment is due to complete by December 2013 and the first<br />

planned interim analysis of the primary outcome will be mid-2016.<br />

Conclusion: The IDSMC last reviewed the trial in December 2011 and<br />

congratulated the Trial Management Group on the conduct of the trial<br />

and the quality of the data. No safety concerns were identified, and<br />

the IDSMC proposed that the trial continue to planned recruitment.<br />

OT1-1-04<br />

ALTERNATIVE: safety and efficacy of lapatinib (L), trastuzumab<br />

(T), or both in combination with an aromatase inhibitor (AI)<br />

for the treatment of hormone receptor-positive (HR+), human<br />

epidermal growth factor receptor 2 positive (HER2+) metastatic<br />

breast cancer<br />

Johnston S, Wroblewski S, Huang Y, Harvey C, Nagi F, Franklin N,<br />

Gradishar W. Royal Marsden NHS Foundation Trust and Institute<br />

of <strong>Cancer</strong> Research, London, United Kingdom; GlaxoSmithKline,<br />

Collegeville, PA; GlaxoSmithKline, Stockley Park, United Kingdom;<br />

Northwestern University, Chicago, IL<br />

Background: Overexpression of the human epidermal growth<br />

factor receptor 2 (HER2) gene in breast cancer is associated with an<br />

aggressive phenotype, poor prognosis, and resistance to endocrine<br />

therapies. Of HER2+ patients, ∼50% are also hormone receptorpositive<br />

(HR+). For patients who are both HER2+ and HR+,<br />

combining the AI letrozole with the dual tyrosine kinase inhibitor L<br />

has been shown to improve outcomes compared with letrozole alone.<br />

The combination of L and T, a humanized monoclonal antibodytargeting<br />

HER2, has been shown to improve outcomes compared<br />

with L alone.<br />

Trial design: The ALTERNATIVE study is a Phase III, randomized,<br />

open-label, multicenter trial, which will examine the efficacy of L/T/<br />

AI in combination versus T/AI alone. Patients will be randomized<br />

to 1 of 3 treatment arms: L 1000 mg po QD plus T (loading dose of<br />

8 mg/kg followed by maintenance with 6 mg/kg IV q3w plus an AI<br />

po QD); T plus an AI; or L 1500 mg po QD plus an AI. Choices of<br />

AI include letrozole, anastrozole, or exemestane.<br />

Eligibility criteria: Postmenopausal female patients with HER2+/<br />

HR+ metastatic breast cancer (MBC) who have received neo/adjuvant<br />

T and endocrine therapy, are treatment naïve for MBC, and are not<br />

candidates for chemotherapy.<br />

Specific aims: The primary efficacy endpoint is overall survival<br />

(OS), defined as the time from randomization until death due to any<br />

cause, for L/T/AI compared with T/AI alone. Secondary efficacy<br />

objectives include comparisons of OS between T/AI and L/AI as<br />

well as between T/L/AI and L/AI in addition to comparisons of<br />

progression-free survival, overall response rate, time to response, and<br />

duration of response. The safety objective is to evaluate the safety<br />

and tolerability for all 3 treatment groups.<br />

A 4-year recruitment is anticipated. More than 200 centers across<br />

37 countries are planned; approximately 110 centers are currently<br />

open for enrollment.<br />

Statistical methods: The study is powered to detect a 42% reduction<br />

in risk of death (hazard ratio=0.70) in patients who receive L/T/AI<br />

(median 28.5 months) versus T/AI (median 20 months) using a 1-sided<br />

test for superiority with α=0.025. The required number of total events<br />

to achieve a power of 80% is 249. Secondary comparisons are not<br />

powered and will be based on the intent-to-treat population.<br />

Present and target accrual: Twenty-six (26) of 525 patients have<br />

been randomized. Patients who have participated in previous neo-/<br />

adjuvant trials including a T regimen are eligible, provided they meet<br />

all other inclusion criteria.<br />

The study is currently recruiting patients, with an anticipated target<br />

accrual of 525 patients by March 2016.<br />

Clinical trial registry number: NCT01160211<br />

OT1-1-05<br />

A Phase I pharmacokinetics trial comparing PF-05280014 and<br />

trastuzumab in healthy volunteers (REFLECTIONS B327-01)<br />

Ricart AD, Zacharchuk C, Reich SD, Meng X, Barker KB, Taylor CT,<br />

Hansson AG. Pfizer Inc., <strong>San</strong> Diego, CA; Pfizer Inc., Cambridge, MA;<br />

Pfizer Inc., New Haven, CT<br />

Background: Trastuzumab is a humanized recombinant monoclonal<br />

antibody that selectively binds the extracellular domain of human<br />

epidermal growth factor receptor 2 (HER2) and is approved for<br />

treatment of breast and gastric cancers. PF05280014 is being<br />

developed as a potential biosimilar to trastuzumab. In nonclinical<br />

evaluations, PF-05280014 has an identical amino acid sequence to<br />

trastuzumab and similar physicochemical and in vitro functional<br />

properties. The goal of this phase I trial is to demonstrate the<br />

pharmacokinetic similarity of PF-05280014 to trastuzumab sourced<br />

from both the United States (trastuzumab-US) and European Union<br />

(trastuzumab-EU).<br />

Trial design: In this double-blind, parallel group, single dose<br />

trial, subjects will be randomized 1:1:1 into 3 arms: PF-05280014;<br />

trastuzumab-US, and trastuzumab-EU (NCT01603264).<br />

Eligibility: Healthy male volunteers, 18-55 years of age with normal<br />

left ventricular ejection fraction are eligible. Multiple exclusion<br />

criteria common to Phase 1 trials are in effect. All subjects must<br />

provide informed consent.<br />

Aims: The primary objectives are to demonstrate the pharmacokinetic<br />

similarity of PF-05280014 to trastuzumab-US and trastuzumab-EU.<br />

Secondary objectives include evaluating the safety, tolerability, and<br />

immunogenicity of PF-05280014 compared with US-licensed and<br />

EU-approved trastuzumab products.<br />

Statistical methods: Pharmacokinetic similarity will be demonstrated<br />

if the 90% confidence interval of the ratio of the area under the<br />

concentration-versus-time curve from time 0 to the last time point<br />

with quantifiable concentration (AUC T ) and maximum concentration<br />

(C max ) of PF-05280014 to both trastuzumab-US and trastuzumab-EU<br />

are within 80% – 125%. At least 93 subjects, 31/arm, will be needed<br />

to provide >81% power to demonstrate pharmacokinetic similarity<br />

for all comparisons. The planned enrolment is 105 subjects to account<br />

for subjects who may not complete the full follow-up period. The<br />

intent-to-treat (ITT) population is defined as all subjects who are<br />

randomized to receive treatment. The modified ITT population is<br />

defined as all subjects who are randomized and receive at least one<br />

dose of treatment and will be used to assess safety, tolerability, and<br />

immunogenicity. The per-protocol population is defined as all subjects<br />

who are randomized to and receive treatment and do not have any<br />

major protocol violations and will be used for the primary evaluation<br />

of pharmacokinetic parameters.<br />

Accrual: The target accrual is 105 subjects; present accrual is<br />

17subjects.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

558s<br />

<strong>Cancer</strong> Research


December 4-8, 2012<br />

<strong>Abstract</strong>s: Poster Session OT1<br />

OT1-1-06<br />

A phase III randomized study of Paclitaxel and Trastuzumab<br />

versus Paclitaxel, Trastuzumab and Lapatinib in first line<br />

treatment of HER2 positive metastatic breast cancer.<br />

Crown JP, Moulton B, O’Donovan N. St Vincent’s University<br />

Hospital, Elm Park, Dublin, Ireland; ICORG (All Ireland Cooperative<br />

Oncology Research Group), Dublin 4, Ireland; National Institute for<br />

Cellular Biotechnology, Dublin City University, Dublin 9, Ireland<br />

Background: Preclinical studies have shown that dual targeting<br />

of the extracellular domain and the kinase domain of HER2 using<br />

trastuzumab (H) and lapatinib (L) produces greater growth inhibition<br />

than single agent treatment. The combination of trastuzumab<br />

and lapatinib has shown improved clinical outcome compared to<br />

lapatinib alone in the pre-treated metastatic setting (EGF104900),<br />

and compared to trastuzumab in the neo-adjuvant setting (Neo-Altto).<br />

This international phase III randomised trial will compare the efficacy<br />

of trastuzumab and paclitaxel (T) with trastuzumab, paclitaxel and<br />

lapatinib in first line treatment of HER2 positive metastatic breast<br />

cancer, and will examine potential predictive biomarkers of response<br />

to trastuzumab and/or lapatinib.<br />

Study Design and Eligibility: Six hundred patients with invasive<br />

HER2 positive (3+ or FISH positive) metastatic breast cancer<br />

(measurable disease per RECIST 1.1), who have not received prior<br />

systemic therapy for metastatic disease, will be randomised to receive<br />

(A) weekly paclitaxel (80 mg/m², for 3 weeks of a 4 week cycle) plus<br />

trastuzumab (8 mg/kg loading dose day 1 and 4mg/kg every 2 weeks)<br />

or (B) weekly paclitaxel (80 mg/m², for 3 weeks of a 4 week cycle)<br />

plus trastuzumab (8 mg/kg loading dose day 1 and 4 mg/kg every<br />

2 weeks) plus lapatinib (1,000 mg daily), until disease progression,<br />

unacceptable toxicity or consent withdrawal.<br />

Objectives: The primary objective of the study is to compare the<br />

efficacy of THL versus TH in first line treatment of metastatic HER2<br />

positive breast cancer. Secondary objectives include: (i) examining<br />

the objective tumour response rate and overall survival; (ii) assessing<br />

the safety and tolerability of lapatinib when administrated with both<br />

paclitaxel and trastuzumab; (iii) examining the effects of the TH<br />

regimen versus the THL regimen on health-related quality of life<br />

(FACT-B); (iv) examining potential biomarkers in tumour tissue<br />

and serum samples; (v) determining if prophylactic loperamide<br />

significantly reduces the number of diarrhoea-related adverse events.<br />

Statistical Methods: The expected median progression free survival<br />

time for the control arm is 6.9 months based on the trastuzumab plus<br />

paclitaxel arm of the phase III trastuzumab plus chemotherapy trial.<br />

A total of 600 evaluable patients (and 485 observed events) would be<br />

sufficient to detect an increase to 8.9 months in median PFS time for<br />

the THL combination, with 80% power and a two sided significance<br />

level of 0.05. Progression-free survival will be analysed at nine<br />

months after enrolment ends and overall survival will be analysed<br />

at 30 months after the end of enrolment. Objective response will be<br />

defined as the proportion of patients who receive a complete or partial<br />

response as defined by RECIST 1.1.<br />

Accrual: This study will accrue six hundred patients across forty<br />

International centres. Countries signed up to participate include<br />

Finland, France, Germany, Greece, Iceland, Ireland, Israel, Italy,<br />

Netherlands, Norway, Poland, Portugal, Switzerland and Spain. The<br />

study opened to accrual in Ireland Feb 12 and six patients have been<br />

accrued to date.<br />

Funding: Trial supported by GlaxoSmithKline. Lapatinib kindly<br />

supplied by GlaxoSmithKline.<br />

OT1-1-07<br />

Human epidermal growth factor receptor 2 (HER2) suppression<br />

with the addition of lapatinib to trastuzumab in HER2-positive<br />

metastatic breast cancer (LTP112515)<br />

Lin N, Danso MA, David AK, Muscato J, Rayson D, Houck, III WA,<br />

Ellis C, DeSilvio M, Garofalo A, Nagarwala Y, Winer E. Dana-<br />

Farber <strong>Cancer</strong> Institute, Boston, MA; Virginia Oncology Associates,<br />

Norfolk, VA; Augusta Oncology Associates, Augusta, GA; Missouri<br />

<strong>Cancer</strong> Associates, Columbia, MO; QEII Health Sciences Centre,<br />

Halifax, NS, Canada; Virginia <strong>Cancer</strong> Specialists, PC, Winchester,<br />

VA; GlaxoSmithKline Oncology, Collegeville, PA<br />

Background: The combination of lapatinib (L) with trastuzumab (T)<br />

results in enhanced antitumor activity in preclinical models of HER2-<br />

positive breast cancer (BC), due to the complementary mechanisms<br />

of action of the two agents. In patients with prior T-treated HER2-<br />

positive metastatic BC (MBC), treatment with T plus L was<br />

associated with longer progression-free survival (PFS) and overall<br />

survival (OS) compared with L alone. The combination also had an<br />

acceptable safety and tolerability profile. In patients with stage II/III<br />

BC, preoperative treatment with the combination plus paclitaxel (P)<br />

resulted in significantly higher pathologic complete response rates<br />

compared with P combined with either agent alone. This evidence<br />

supports the concept of dual HER2 blockade as a treatment strategy<br />

for HER2-positive MBC. Current common clinical practice for most<br />

patients experiencing clinical benefit from first-line treatment with<br />

T plus chemotherapy is to continue T as maintenance therapy after<br />

completion of the recommended number of chemotherapy cycles.<br />

The present study is designed to evaluate whether the addition of L<br />

improves PFS among women with HER2-positive MBC receiving<br />

T as maintenance therapy.<br />

Methods: In this open-label, Phase III study, 280 patients will be<br />

stratified by line of treatment (first/second) and hormone receptor<br />

status (positive/negative), then randomized 1:1 to receive maintenance<br />

treatment with either L (1000 mg qd, continuously) in combination<br />

with T (6 mg/kg once every 3 weeks [q3w]), or T (6 mg/kg q3w)<br />

alone. Patients will receive study treatment until disease progression,<br />

death, discontinuation due to adverse events, or patient withdrawal.<br />

The primary endpoint is PFS; secondary endpoints are; OS, clinical<br />

benefit rate, and safety.<br />

Key eligibility criteria include females, age ≥18 years with HER2-<br />

positive MBC who have completed 12 to 24 weeks of first- or<br />

second-line treatment with T plus chemotherapy with an objective<br />

response or stable disease at time of chemotherapy discontinuation.<br />

Patients with stable brain metastases are eligible if entering the study<br />

on second-line treatment.<br />

Efficacy endpoints will be analyzed in the intent-to-treat population. A<br />

total of 193 PFS events is required to detect a 50% increase in median<br />

PFS (hazard ratio=0.667) from 18 weeks (T alone) to 27 weeks ( L<br />

plus T). The hypothesis will be tested using a 1-sided test with 80%<br />

power and a type I error of 0.025. One interim analysis is planned<br />

for futility when approximately 97 PFS events (50% of the required<br />

events) have been observed. Safety endpoints will be analyzed in<br />

all randomized patients who receive ≥1 dose of study medication.<br />

The trial is currently open for accrual in the United States and Canada.<br />

clinicaltrials.gov - NCT00968968<br />

www.aacrjournals.org 559s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

OT1-1-08<br />

Clinical outcomes among ErbB2+ MBC patients treated with<br />

lapatinib-capecitabine after trastuzumab progression: Role of<br />

early switch to lapatinib (TYCO study).<br />

Abulkhair O, Uslu R, Sezgin C, Büyükberber S, Darwish T, Isikdogan<br />

A, Gumus M, Dane F, Sevinc A, Halawani H, Uncu D, Marrero N,<br />

Tobler J, Soares C, Landis S, Moraes E, Gidekel R, <strong>San</strong>tillana S,<br />

Nunez P, Cagnolati S, Rodriguez JG. Ege University Medical Faculty,<br />

Izmir, Turkey; Gazi University Medical Faculty, Ankara, Turkey;<br />

King Abdulaziz Medical City National Guard Health Affairs, Riyadh,<br />

Saudi Arabia; King Abdullah Medical City - Oncology Centre,<br />

Jeddah, Saudi Arabia; Dicle University Medical Faculty, Diyarbakir,<br />

Turkey; Kartal Training and Research Hospital, Istanbul, Turkey;<br />

Marmara University Training and Research Hospital, Istanbul,<br />

Turkey; Gaziantep University Medical Faculty, Gaziantep, Turkey;<br />

King Fahad Specialist Hospital, Dammam, Saudi Arabia; Numune<br />

Training and Research Hosptial, Ankara, Turkey; Instituto Docente<br />

de Urología, Valencia, Venezuela; GlaxoSmithKline, Rio de Janeiro,<br />

Brazil; GlaxoSmithKline, Buenos Aires, Argentina; GlaxoSmithKline,<br />

Stockley Park, United Kingdom; Hospital Oncológico Padre<br />

Machado, Caracas, Venezuela; Centro Oncologico-FIDES-La Plata,<br />

Buenos Aires, Argentina<br />

Background: Lapatinib in combination with capecitabine is a standard<br />

of care treatment for ErbB2+ metastatic breast cancer (MBC) patients<br />

who have progressed after anthracyclines, taxanes and trastuzumab<br />

treatment. Results from the lapatinib pivotal trial showed that the<br />

addition of lapatinib to capecitabine increased median time to<br />

progression (TTP) even among heavily pre-treated patients (median of<br />

4 prior lines of therapy). A post-hoc exploratory sub-group analysis of<br />

this trial suggested that earlier administration of lapatinib-capecitabine<br />

in MBC patients who progress after trastuzumab may produce better<br />

clinical outcomes. The TYCO study was designed to evaluate if early<br />

initiation of lapatinib-capecitabine in patients with ErbB2+ MBC who<br />

have progressed on trastuzumab-containing regimen improves TTP<br />

in comparison with a delayed start of the therapy.<br />

Trial design: TYCO is an international, multicenter, prospective,<br />

observational study in 269 ErbB2+ MBC patients whose disease has<br />

progressed after treatment with trastuzumab in the metastatic setting.<br />

Two cohorts will be compared; Group 1: patients receiving lapatinibcapecitabine<br />

just after the first trastuzumab progression, and Group<br />

2: patients receiving lapatinib-capecitabine after two or more lines<br />

of treatment after first trastuzumab progression. The study duration<br />

is of 12 months with data collection at baseline and approximately<br />

every 3 months thereafter.<br />

Major Eligibility Criteria:<br />

1. Females ≥18y with confirmed ErbB2+ MBC who have progressed<br />

after a previous trastuzumab-containing regimen,<br />

2. Pts eligible for standard therapy with lapatinib-capecitabine at<br />

approved conventional doses, as per local approved label.<br />

3. Pts eligible to start standard treatment with Lapatinib-capecitabine<br />

at conventional doses, or receiving standard treatment with Lapatinibcapecitabine<br />

at conventional doses, for no longer than 10 weeks from<br />

the start of the treatment to the date of inclusion in the study;<br />

Aims: Primary objective of this study is to determine if early switch<br />

to lapatinib-capecitabine in patients with ErbB2+ metastatic breast<br />

cancer who have progressed on trastuzumab containing regimen<br />

improves time to disease progression as determined by treating<br />

physician either clinically or radiologically. Secondary objectives<br />

include overall response rate and overall survival.<br />

Statistical Methods: Kaplan-Meier plots will be used to describe the<br />

median TTP after start of lapatinib-capecetabine. Cox proportional<br />

hazard model will be developed to estimate the adjusted hazard ratio<br />

(and 95% confidence intervals) comparing TTP for the two treatment<br />

group using propensity score methods (trimmed sample, adjustment<br />

for the continuous propensity score measure, and doubly robust<br />

adjustment) to adjust for potential confounding by indication that<br />

may arise due to the non-randomised design.<br />

Present and Target Accrual: Enrollment began in February 2010,<br />

and as per May 2012, 266 patients have been included from Turkey,<br />

Venezuela, Argentina, Saudi Arabia and Colombia.<br />

Country<br />

Enrollment<br />

Turkey 134<br />

Venezuela 44<br />

Argentina 29<br />

Saudi Arabia 38<br />

Colombia 21<br />

Total 266<br />

OT1-1-09<br />

Opti-HER HEART: A prospective, multicenter, single-arm,<br />

phase II study to evaluate the safety of neoadjuvant liposomal<br />

doxorubicin plus paclitaxel, trastuzumab, and pertuzumab in<br />

patients with operable HER2-positive breast cancer.<br />

Gavilá J, Llombart A, Guerrero A, Ruiz A, Guillem V. Fundación<br />

Instituto Valenciano de Oncología, Valencia, Spain; Hospital Arnau<br />

de Vilanova, Valencia, Spain; SOLTI <strong>Breast</strong> <strong>Cancer</strong> Research Group,<br />

Barcelona, Spain<br />

Background:<br />

In recent years, the concomitant use of two anti-HER2 therapies<br />

has been generating great expectations. Of note, despite the high<br />

activity reported by several studies, none has ever included a<br />

chemotherapeutic regimen with anthracyclines. The therapeutic value<br />

of anthracyclines in HER2-positive breast cancer (BC) has been<br />

well established. However, the feasibility of their combination with<br />

trastuzumab is still controversial. Liposomal anthracyclines, on the<br />

other hand, may offer a safer alternative. Of note, in our institutional<br />

experience using a regimen of liposomal doxorubicin plus weekly<br />

paclitaxel and trastuzumab as standard neoadjuvant therapy for stage<br />

II-III breast cancer (BC) patients reach a pathological complete<br />

response (pCR) rate of 65%, without any grade 3/4 cardiac toxicity.<br />

Trial Design: This study seeks to optimize the treatment of patients<br />

with operable HER2-positive BC, while minimizing cardiac risk, by<br />

combining dual anti-HER2 blockade plus a taxane and a liposomal<br />

anthracycline. In this single-arm, phase II clinical trial, patients will<br />

receive neoadjuvant therapy for six 21-day cycles. Regimen consists<br />

of: trastuzumab 4 mg/kg loading dose on Day 1 of Cycle 1, then 2<br />

mg/kg on Days 8 and 15 and on Days 1, 8, and 15 of subsequent<br />

cycles; pertuzumab 840 mg loading dose on Day 1 of Cycle 1, then<br />

420 mg on Day 1 of the following cycles; liposome-encapsulated<br />

doxorubicin 50 mg/m2 on Day 1 of each cycle; and, paclitaxel 80<br />

mg/m2 on Days 1, 8, and 15 of each cycle. ECG will be performed<br />

at baseline and every planned visit. Left ventricular ejection fraction<br />

(LVEF) decline will be assessed at baseline, weeks 6 and 12, before<br />

surgery, and every 3 months thereafter during the adjuvant period,<br />

for a total of 12 months. In order to assess biomarkers of cardiac<br />

injury, blood samples will be collected at the same time points. An<br />

initial safety run-in phase is included, in which the first ten enrolled<br />

patients undergo an intensified safety monitoring for cardiac and<br />

hematological adverse events.<br />

Eligibility: Female patients 18-74 years old with primary HER2-<br />

positive invasive breast cancer eligible for definitive surgery, with<br />

adequate cardiac function.<br />

Specific aims: The primary objective is to assess cardiac safety,<br />

measured by the incidence of symptomatic cardiac events and by<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

560s<br />

<strong>Cancer</strong> Research


December 4-8, 2012<br />

<strong>Abstract</strong>s: Poster Session OT1<br />

asymptomatic LVEF decline. Secondary objectives include efficacy,<br />

breast conservation rate, additional safety and tolerability, and<br />

predictive biomarkers of cardiotoxicity.<br />

Statistical methods: Assuming that the incidences of cardiac events<br />

with regimens containing anti-HER2 agents are 3% for symptomatic<br />

and 15% for asymptomatic, 83 patients will be required to reject the<br />

null hypothesis with 80% confidence. Additionally, efficacy will be<br />

evaluated by pCR rates.<br />

Target accrual: Recruitment started in September 2012 and will<br />

include 83 patients across 20 sites in Spain. Trastuzumab and<br />

pertuzumab are kindly provided by Roche. The trial is supported by<br />

a grant from TEVA.<br />

OT1-1-10<br />

DETECT III - A multicenter, randomized, phase III study to<br />

compare standard therapy alone versus standard therapy plus<br />

lapatinib in patients with initially HER2-negative metastatic<br />

breast cancer but with HER2-positive circulating tumorcells<br />

Melcher CA, Janni JW, Schneeweiss A, Fasching PA, Hagenbeck CD,<br />

Aktas B, Pantel K, Solomayer EF, Ortmann U, Jaeger BAS, Mueller<br />

V, Rack BK, Fehm TN. University Hospital Duesseldorf, Germany;<br />

National Center for Tumor Diseases, Heidelberg, Germany;<br />

University Hospital Erlangen; University Hospital Essen, Germany;<br />

University Medical Center Hamburg-Eppendorf, Hamburg, Germany;<br />

University Hospital Homburg, Germany; Ludwig-Maximilians-<br />

University Munich, Munich, Germany; University Hospital Hamburg-<br />

Eppendorf, Germany; University Hospital Tuebingen, Germany<br />

Background: Despite a HER2-negative primary tumor approximately<br />

20-30% of patients develop HER2-positive metastases (Zidan et al.<br />

2005; Tewes et al. 2009). As previously described in the DETECT I<br />

trial (Fehm et al. 2010) determination of HER2 status on circulating<br />

tumor cells (CTCs) is one option for re-evaluating HER2-status<br />

in the metastatic setting. Currently it is unclear if HER2-targeted<br />

therapy based on the assessment of HER2-status of CTCs reveals a<br />

clinical benefit.<br />

Trial design: DETECT III is a randomized, open-label, two arm phase<br />

III study comparing standard treatment alone vs. standard treatment<br />

plus HER2-targeted therapy with lapatinib in HER2-negative<br />

metastatic breast cancer patients with HER2-positive CTCs. Choices<br />

of chemotherapy and endocrine therapy include: docetaxel, paclitaxel,<br />

capecitabine, vinorelbine, non pegylated liposomal doxorubicin,<br />

letrozole, exemestane and anastrozole.<br />

Main eligibility criteria:<br />

1. metastatic breast cancer with HER2-negative primary tumor tissue<br />

and/or biopsies from metastatic sites or locoregional recurrences<br />

2. evidence of ≥ 1 HER2-positive CTC<br />

3. ≥ 1 evaluable metastatic lesion according to RECIST<br />

4. Tumor evaluation within 6 weeks before randomization<br />

Specific aims:<br />

Objective<br />

The objective of the trial is to prove the clinical efficacy of lapatinib in<br />

patients with metastasizing breast cancer who exhibit HER2-positive<br />

circulating tumor cells (CTC) although the primary tumor tissue and/<br />

or biopsies from metastatic sites were investigated for HER2 status<br />

and showed HER2-negativity.<br />

Primary endpoint:<br />

· Progression free survival<br />

Secondary endpoints:<br />

· Overall response rate<br />

· Clinical benefit rate<br />

· Overall survival<br />

· Dynamic of CTC<br />

· Quality of life<br />

· Safety and tolerability of lapatinib<br />

Statistical methods: The primary endpoint will be analyzed by<br />

Kaplan-Meier method using the logrank test in order to compare<br />

the PFS distributions of the two arms. Efficacy, toxicity and other<br />

event rates are calculated, providing confidence intervals. In case<br />

of comparison between patient groups, these rates will be analyzed<br />

by Fisher’s exact test or test. The Kaplan Meier analysis for all<br />

event related data will be carried out overall for the whole patient<br />

population. Furthermore a Cox regression analysis will be done using<br />

the following covariates<br />

· Hormone receptor status (positive/negative)<br />

· Number of prior chemotherapy lines for metastatic disease<br />

· Prior endocrine therapy for metastatic disease<br />

· Endocrine treatment vs. cytotoxic treatment<br />

· One metastatic site vs. multiple metastatic sites<br />

· Bone metastases vs. no bone involvement<br />

· Performance status ECOG Score (0 / > 0)<br />

Present accrual and target accrual: As only half of the patients<br />

with HER2-negative metastatic breast cancer show CTC-positivity<br />

and of those approximately 32% will exhibit HER2-positive CTC<br />

(Fehm et al. 2010), screening of about 1420 patients is required to<br />

enroll 228 patients. First patient was screened in February 2012. As<br />

of July 27 th 2012 117 patients were screened and 21 were found to<br />

have HER2-positive CTCs<br />

OT1-1-11<br />

TBCRC 022: Phase II Trial of Neratinib for Patients with Human<br />

Epidermal Growth Factor Receptor 2 (HER2)-Positive <strong>Breast</strong><br />

cancer and Brain Metastases<br />

Freedman RA, Gelman RS, Wefel JS, Krop IE, Melisko ME, Ly A, Agar<br />

NYR, Connolly RM, Blackwell KL, Nabell LM, Ingle JN, Van Poznak<br />

CH, Puhalla SL, Niravath PA, Ryabin N, Wolff AC, Winer EP, Lin N.<br />

Dana-Farber <strong>Cancer</strong> Institute, Boston, MA; The University of Texas<br />

MD Anderson <strong>Cancer</strong> Center, Houston, TX; University of California,<br />

<strong>San</strong> Francisco, CA; Brigham and Women’s Hospital, Boston, MA;<br />

Johns Hopkins University, Baltimore, MD; Duke University, Durham,<br />

NC; University of Alabama, Birmingham, AL; Mayo Clinic, Rochester,<br />

MN; University of Michigan, Ann Arbor, MI; Univerity of Pittsburgh,<br />

Pittsburgh, PA; Baylor, Houston, TX<br />

Background: 1/3 of women with metastatic HER2+ breast cancer<br />

will develop central nervous system (CNS) metastases yet evidencebased<br />

treatments for women with progressive CNS disease are limited.<br />

Neratinib is an irreversible inhibitor of erbB1, HER2, and erbB4<br />

which has promising activity in HER2+ breast cancer. Preclinical<br />

evidence suggests it may cross the blood brain barrier.<br />

Trial Design: This is a multicenter, phase II, open-label study of<br />

neratinib for patients with HER2+ breast cancer and brain metastases.<br />

Neratinib is administered at 240 mg orally daily during a 28 day<br />

cycle. Two cohorts will be enrolled: Cohort 1 will enroll 40 patients<br />

with progressive CNS disease; cohort 2 will enroll ≤5 patients who<br />

are candidates for surgical excision of intracranial disease. Surgical<br />

candidates receive neratinib 7-21 days preoperatively and resume<br />

postoperatively. All patients are re-staged every 2 cycles. Those who<br />

develop non-CNS progression have an option to extend therapy with<br />

trastuzumab+neratinib. Circulating tumor cells (CTC) are collected at<br />

baseline and progression; neurocognitive testing, HADS and EORTC<br />

QLQ30/BN20 measures are administered at baseline, cycle 2, cycle<br />

3, and progression (cohort 1). Intracranial tumor, cerebrospinal fluid<br />

(CSF), and plasma are collected at surgery (cohort 2).<br />

www.aacrjournals.org 561s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Specific Aims: The primary endpoint is CNS objective response<br />

rate (ORR) by composite criteria. Additional endpoints include:<br />

non-CNS ORR, progression-free survival, overall survival (OS),<br />

site of 1st progression, and toxicity. Correlative and exploratory<br />

endpoints include association of CTC count and OS and longitudinal<br />

neurocognitive function and quality of life. In an exploratory<br />

analysis (cohort 2), we will quantify neratinib concentrations in<br />

CSF, intracranial tissue, and plasma and examine associations with<br />

response.<br />

Eligibility: Patients must have confirmed HER2+ metastatic disease<br />

with ≥1 parenchymal brain lesion measuring ≥10 mm that is new<br />

or progressed after completing ≥1 line of standard CNS-directed<br />

treatment (cohort 1) or CNS disease that is amenable for surgery,<br />

including those without prior CNS treatments (cohort 2). Additional<br />

eligibility criteria (cohorts 1,2) include: adequate performance status<br />

and end organ/marrow function, and ejection fraction ≥50%. Any<br />

number of prior lines of therapy is allowed, including prior lapatinib.<br />

Statistical Methods: Cohort 1 has a 2-stage design with up to 40<br />

patients. CNS ORR is defined as ≥50% reduction in sum volume of<br />

CNS target lesions, without evidence of new lesions, progression of<br />

non-target CNS lesions, non-CNS disease progression, worsening<br />

neurological symptoms, or increase in corticosteroids. CNS lesion<br />

measurements are performed centrally by the Harvard Tumor Imaging<br />

Metrics Core. If 1/18 patients have a CNS response in the 1st stage,<br />

another 22 patients will enroll. With this design, if ≥5 of 40 patients<br />

achieve a CNS response, the drug will be deemed worthy of future<br />

study. This 2-stage design has 92% power to distinguish between<br />

a true CNS ORR of 20% and a null of 6% (one-sided type I error<br />

rate=9%).<br />

Accrual: Accrual has begun. Target=45 (cohort 1=40, cohort 2=5)<br />

OT1-1-12<br />

NSABP FB-7 Trial: A Phase II Randomized Clinical Trial<br />

Evaluating Neoadjuvant Therapy Regimens with Weekly<br />

Paclitaxel and Neratinib or Trastuzumab or Neratinib and<br />

Trastuzumab Followed by Doxorubicin and Cyclophosphamide<br />

with Postoperative Trastuzumab in Women with Locally<br />

Advanced HER2-Positive <strong>Breast</strong> <strong>Cancer</strong><br />

Lu J, Jacobs SA, Buyse ME, Paik S, Wolmark N. State University<br />

of New York at Stony Brook, Stony Brook, NY; National Surgical<br />

Adjuvant <strong>Breast</strong> and Bowel Project, Pittsburgh, PA<br />

Neratinib is an oral, small molecule which acts as an irreversible<br />

inhibitor of the pan ErbB receptor tyrosine kinase. Dual ErbB<br />

blockade combined with chemotherapy improves efficacy in Her-<br />

2 positive breast cancer. Both NeoALTTO (Lancet 2012) and<br />

Neosphere (Lancet Oncol 2012) trials demonstrated higher pCR for<br />

Her-2 positive breast cancer receiving dual anti-Her-2 neoadjuvant<br />

blockade therapy. The purpose of this trial is to determine the activity<br />

and safety profile of Neratinib as mono-blockade or in combination<br />

with Trastuzumab as dual blockade in neoadjuvant therapy of locally<br />

advanced breast cancer (stage IIB, III A, B and C).<br />

Methods: This NSABP Foundation Research Program study (FB-<br />

7) is designed as a Phase II, multi-center, three arm clinical trial for<br />

patients with Her-2 positive locally advanced breast cancer. 126<br />

patients will be enrolled. The initial two- arm study of Neratinib<br />

or Trastuzumab in combination with Paclitaxel began in December<br />

2010. 30 patients were enrolled by December of 2011 at which time<br />

the study was placed on hold awaiting the recommended phase II<br />

dose of the three drug combination, Neratinib, Trastuzamab and<br />

Paclitaxel ( NSABP FB-8 trial), which is a Phase I dose-escalation<br />

study in women with metastatic Her-2 positive breast cancer. The<br />

amended FB -7 trial is now a three arm trial of weekly Paclitaxel<br />

and Neratinib or Trastuzumab or Neratinib and Trastuzumab for 4<br />

cycles followed by Doxorubicin and Cyclophosphamide for 4 cycles<br />

prior to surgical resection. Patients will then receive postoperative<br />

Trastuzumb to complete one total year of Her-2 blockade therapy. The<br />

primary goal of this study is to determine the pathologic complete<br />

response in breast and axillary lymph nodes following completion<br />

of neoadjuvant therapy.<br />

OT1-1-13<br />

Dual blockade with Afatinib and Trastuzumab as neooadjuvant<br />

treatment for patients with locally advanced or operable breast<br />

cancer receiving taxane-anthracycline containing chemotherapy<br />

(DAFNE)-GBG70<br />

Hanusch C, Schneeweiss A, Untch M, Paepke S, Kuemmel S,<br />

Jackisch C, Huober J, Hilfrich J, Gerber B, Eidtmann H, Denkert C,<br />

Costa S-D, Blohmer J-U, Loibl S, Nekljudova V, von Minckwitz G.<br />

Rotkreuzklinikum Muenchen; University Heidelberg; Helios Klinik<br />

Berlin; Frauenklinik Muenchen; Kliniken Essen Mitte; Klinikum<br />

offenbach; University Duesseldorf; Eilenriedeklinik Düsseldorf;<br />

University Rostock; University Kiel; Charite Berlin; University<br />

Magdeburg; <strong>San</strong>kt Gertrauden Berlin; German <strong>Breast</strong> Group, Neu-<br />

Isenburg<br />

Background: Anthracycline/taxane based combination chemotherapy<br />

of at least 18 weeks represents the standard of care in the neoadjuvant<br />

setting. In HER2 positive disease trastuzumab is given concurrently<br />

to chemotherapy and achieves a pCR rate (no invasive residuals in<br />

breast and nodes) of approx. 40% which can be increased by double<br />

anti HER2 blockade by approximately 20%. There are no data on the<br />

combination of afatinib (BIBW 2992), an irreversible HER family<br />

inhibitor with trastuzumab.<br />

Methods: This is a multi-centre, prospective, open-label phase II<br />

study evaluating the efficacy and safety of afatinib in combination<br />

with weekly paclitaxel + trastuzumab followed by epirubicin/<br />

cyclophosphamide/trastuzumab as neoadjuvant therapy in<br />

patients with untreated HER2-positive early breast cancer. Pts<br />

with histologically confirmed, centrally reviewed HER2 positive,<br />

unilateral, primary operable or locally advanced breast cancer can be<br />

included. Tumor size has to be at least 2cm by sonography.<br />

All patients will be treated for a total duration of 30 weeks (6 weeks<br />

with afatinib (20mg) and trastuzumab (8/6mg/kg) alone, 12 weeks<br />

with weekly paclitaxel (80mg/m²), afatinib and trastuzumab and 12<br />

weeks with epirubicin/cyclophosphamide/trastuzumab according to<br />

standard). During the first 2 weeks afatinib 20 mg will be given only<br />

every other day to reduce the risk of diarrhea and skin toxicities.<br />

Primary prophylaxis with loperamide 2x 2 mg daily is obligatory<br />

during the first 4 weeks of afatinib/trastuzumab and the first 2 weeks<br />

of afatinib/trastuzumab/paclitaxel. Thereafter prophylaxis with<br />

loperamide can be stopped if no diarrhea grade > 1 occurred.<br />

Primary objective is pathological complete response (pCR = ypT0/<br />

is ypN0). Secondary objectives are pCR by other definitions, clinical<br />

response rates, rate and type of surgery, toxicity and compliance, pCR<br />

related to skin toxicity and diarrhoea and pre-specified molecular<br />

markers. An extensive biomaterial collection is integreated, including<br />

obligatory biomaterial (e.g. skin biopsies) collection at baseline, prior<br />

to start of paclitaxel at the end of paclitaxel and prior to surgery.<br />

Neoadjuvant anthracycline-taxane-based chemotherapy given<br />

simultaneously with trastuzumab after central HER 2-testing results in<br />

a pCR rate of approx. 50%. The addition of a dual anti HER2 blockade<br />

to chemotherapy increased the pCR by absolute 20%. A pCR rate of<br />

70% is expectedand and a pCR rate of 55% or lower excluded; with<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

562s<br />

<strong>Cancer</strong> Research


December 4-8, 2012<br />

<strong>Abstract</strong>s: Poster Session OT1<br />

α=0.1 and 1-ß=80%, this requires 65 evaluable patients for two-sided<br />

one group χ2-test. An integrated safety phase is planned for the first<br />

15 patients entering the study.<br />

The trial is registered under NCT015591477. It is financially<br />

supported by Boehringer Ingelheim.<br />

Results: Centres have been initiated after approval by ethics<br />

committee and authorities. Three patients have been recruited. It is<br />

planned to recruit 12 months in 15 sites in Germany.<br />

OT1-1-14<br />

Open-label, Phase II trial of afatinib, with or without vinorelbine,<br />

for the treatment of HER2-overexpressing inflammatory breast<br />

cancer (IBC)*<br />

Swanton C, Cromer J, On Behalf of the 1200.89 Trial Group. CR-UK<br />

London Research Institute, London, United Kingdom; Boehringer<br />

Ingelheim, North Ryde, Australia<br />

Background: IBC is a rare but aggressive form of BC accounting<br />

for 1–5% of all BC. Despite recent treatment advances, prognosis<br />

remains poor, with 3-year survival rate of 40% vs. 85% in non-IBC.<br />

IBC commonly lack ER/PR and more frequently harbor HER2 gene<br />

amplification (∼40%), and EGFR overexpression (∼30%) that are<br />

associated with poor prognosis. Recent data reported clinical efficacy<br />

of lapatinib, a reversible EGFR/HER2 inhibitor, in trastuzumab<br />

naïve/resistant HER2-positive IBC. Afatinib is an oral irreversible<br />

ErbB Family Blocker that blocks signaling through activated EGFR<br />

(ErbB1), HER2 (ErbB2), ErbB4 receptors, and transphosphorylation<br />

of ErbB3. Afatinib demonstrated preclinical activity in trastuzumabresistant<br />

cell lines, HER2-positive tumor xenografts and HER2-<br />

negative SUM149 xenograft model derived from an IBC cell line.<br />

Its clinical efficacy was shown in heavily pretreated, HER2-positive<br />

metastatic BC patients (pts) who progressed after trastuzumab, with<br />

a partial response in 10% and a clinical benefit in 46% of pts. The<br />

combination of afatinib and vinorelbine has been shown to be more<br />

effective than either compound alone in reducing tumor volume in<br />

the SUM190 xenograft model. The purpose of this biomarker-driven<br />

trial is to investigate the efficacy and safety of afatinib alone and<br />

in combination with vinorelbine following progression on afatinib<br />

monotherapy and the genomic changes that occur during treatment in<br />

pts with trastuzumab pretreated or naïve HER2-positive IBC.<br />

Methods: Pts are recruited in two parallel cohorts of 20 pts<br />

each – trastuzumab-naïve and trastuzumab failure cohorts. Key<br />

eligibility criteria: Histologically-confirmed HER2-positive BC;<br />

investigator-confirmed IBC characterized by diffuse erythema and<br />

edema (peau d’orange) – dermal lymphatic emboli or palpable mass<br />

are not necessary for diagnosis; no prior anti-HER2 therapy except<br />

trastuzumab in the trastuzumab failure cohort; no prior vinorelbine;<br />

ECOG performance status 0–2; disease must be biopsiable. All pts<br />

start afatinib monotherapy (40 mg/day oral) in 4-week cycles (Part<br />

A). Following disease progression, pts may receive afatinib (40 mg/<br />

day) + vinorelbine (i.v. 25 mg/m 2 /week) (Part B). Primary endpoint:<br />

Clinical benefit defined as stable disease (for ≥6 months), partial<br />

response or complete response (RECIST 1.1). Secondary endpoints:<br />

Objective response, duration of objective response and progressionfree<br />

survival. Other endpoints include overall survival and safety.<br />

Endpoints are assessed separately for Part A and Part B. In Part A<br />

(afatinib monotherapy) fresh tumor biopsies are taken pretreatment<br />

and following progressive disease, and blood samples are taken<br />

pretreatment to explore predictive markers of response/resistance to<br />

afatinib, to describe the IBC population that may benefit most and<br />

for tumor DNA next-generation sequencing and proteomic analysis.<br />

This is one of the first prospective clinical trials to integrate deep<br />

exome sequencing into trial design to assess proof of principle of a<br />

personalized cancer medicine approach to improve individualized<br />

pt treatment. The trial is planned to accrue 40 pts worldwide and<br />

recruitment is ongoing since August 2011.<br />

*Updated abstract from ASCO 2012.<br />

OT1-1-15<br />

LUX-<strong>Breast</strong> 3: Randomized Phase II trial of afatinib (BIBW<br />

2992) alone or with vinorelbine versus investigator’s choice of<br />

treatment in patients (pts) with HER2-positive breast cancer<br />

(BC) with progressive brain metastases after trastuzumab and/<br />

or lapatinib-based therapy*<br />

Joensuu H, Ould-Kaci M, On Behalf of the 1200.67 Trial Group.<br />

Helsinki University Central Hospital, Helsinki, Finland; Boehringer<br />

Ingelheim, Paris, France<br />

Background: BC is one of the most common causes of central<br />

nervous system (CNS) metastases and approximately a third of pts<br />

with HER2-positive, advanced BC will develop brain metastases<br />

with a poor prognosis. Efficacy of systemic therapies is limited<br />

since few drugs can readily cross the blood-brain barrier. BC brain<br />

metastases represent a high unmet medical need for which novel<br />

therapies are needed. Afatinib is an oral irreversible ErbB Family<br />

Blocker that blocks signaling through activated EGFR (ErbB1), HER2<br />

(ErbB2) and ErbB4 receptors and transphosphorlyation of ErbB3.<br />

Afatinib has shown preclinical activity in trastuzumab-resistant cell<br />

lines and HER2-positive tumor xenografts. Its clinical efficacy has<br />

been demonstrated in pts with heavily pretreated, HER2-positive<br />

metastatic BC (MBC), who progressed after trastuzumab therapy,<br />

with partial responses (PRs) in 10% and clinical benefit in 46%<br />

of pts. In a Phase I afatinib trial, a sustained PR was observed in a<br />

non-small cell lung cancer (NSCLC) pt with brain metastasis and<br />

in a Phase II trial in NSCLC, pts with or without brain metastases<br />

achieved comparable objective response on afatinib. The purpose of<br />

this trial (NCT01441596) is to investigate whether afatinib alone or<br />

in combination with vinorelbine has any effect on HER2-positive BC<br />

with brain metastases after prior trastuzumab and/or lapatinib therapy.<br />

Methods: Key eligibility criteria: Histologically confirmed HER2-<br />

positive BC; recurrent/progressive brain metastases during/after prior<br />

trastuzumab and/or lapatinib therapy; at least one measurable and<br />

progressive lesion in the CNS (≥10 mm on T1-weighted, gadoliniumenhanced<br />

MRI) after prior systemic and/or radiation therapy; extra-<br />

CNS lesions allowed; prior surgery, whole brain radiotherapy or<br />

stereotactic radiosurgery allowed; ECOG performance status (PS)<br />

0–2; no prior treatment with HER2-tyrosine kinase inhibitor other<br />

than lapatinib. Eligible pts are stratified by: ECOG PS score 0–1<br />

vs. 2; number of brain metastases ≤3 vs. >3; prior lapatinib therapy.<br />

Pts are randomized to (n=40/arm): Arm A: Afatinib (40 mg/day<br />

oral); Arm B: Afatinib (40 mg/day oral) + vinorelbine (25 mg/m 2 /<br />

week); Arm C: Investigator’s choice of medical treatment approved<br />

for MBC. All treatments are administered in a 3-weekly course.<br />

Primary endpoint: Pt benefit at 12 weeks (i.e. absence of CNS/extra<br />

CNS disease progression as per RECIST 1.1 and no tumor-related<br />

worsening of neurological signs/symptoms or increase in steroid<br />

dosage). Secondary and other endpoints: Progression-free survival,<br />

overall survival, safety and afatinib concentration in plasma and<br />

cerebrospinal fluid in ≥12 pts who consent to participate in the PK<br />

study. The trial is planned to accrue 120 pts worldwide. When 20<br />

pts in each treatment arm have been treated for at least 12 weeks<br />

(or progressed before Week 12), a Data Monitoring Committee will<br />

www.aacrjournals.org 563s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

evaluate the benefit–risk ratio to decide whether to proceed with the<br />

trial or stop one of the treatment arms for futility. The study began<br />

in October 2011 and enrollment is ongoing.<br />

*Updated abstract from ASCO 2012.<br />

OT1-1-16<br />

LUX-<strong>Breast</strong> 1: Randomized, Phase III trial of afatinib (BIBW<br />

2992) and vinorelbine vs. trastuzumab and vinorelbine in patients<br />

with HER2-overexpressing metastatic breast cancer (MBC)<br />

failing one prior trastuzumab treatment*<br />

Xu B, Im S-A, Huang C-S, Im Y-H, Ro J, Zhang Q, Arora R, Mehta<br />

A, Jung K, Yeh D-C, Lee S, Jassem J, Wojtukiewicz M, Chen S-C,<br />

Lahogue A, Uttenreuther-Fischer M, Hurvitz S-A, Harbeck N,<br />

Piccart-Gebhart M, On Behalf of the LUX-<strong>Breast</strong> 1 Study Group.<br />

Chinese Academy of Medical Sciences, Beijing, China; Seoul National<br />

University Hospital, Seoul, Korea; National Taiwan University<br />

Hospital, Taipei, Taiwan; Samsung Medical Center, Seoul, Korea;<br />

National <strong>Cancer</strong> Center, Goyang, Korea; Third Affiliated Hospital<br />

of Harbin Medical University, Heilongjiang Province, China; Sujan<br />

Surgical <strong>Cancer</strong> Hospital & Amravati <strong>Cancer</strong> Foundation, Amravati,<br />

India; Central India <strong>Cancer</strong> Research Institute, Nagpur, India; Asan<br />

Medical Center, Seoul, Korea; Taichung Veterans General Hospital,<br />

Taichung, Taiwan; Severance Hospital, Seoul, Korea; Medical<br />

University, Gdansk, Poland; Medical University, Bialystok, Poland;<br />

Chang Gung Memorial Hospital - Linkou Branch, Taoyuan County,<br />

Taiwan; SCS Boehringer-Ingelheim Comm.V, Brussels, Belgium;<br />

Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany;<br />

UCLA, Los Angeles, CA; University of Munich, Munich, Germany;<br />

Institut Jules Bordet, Brussels, Belgium<br />

Background: Afatinib is a ErbB Family Blocker that irreversibly<br />

blocks signaling from all relevant ErbB Family dimers. Afatinib<br />

is being developed in EGFR (ErbB1)-driven (non-small cell lung<br />

cancer [NSCLC]/head and neck squamous cell carcinoma) and<br />

HER2 (ErbB2)-driven (breast) malignancies, and has shown clinical<br />

efficacy in NSCLC patients with EGFR activating mutations. 1,2<br />

In trastuzumab-resistant, HER2-positive (SUM190) xenografts,<br />

afatinib showed antitumor activity which was superior to lapatinib.<br />

Afatinib monotherapy activity in this SUM190 model was increased<br />

by the addition of intravenous (i.v.) vinorelbine. Clinically, afatinib<br />

monotherapy demonstrated activity in an open-label, single-arm,<br />

Phase II trial in patients with HER2-positive MBC after progression<br />

on trastuzumab, with a progression-free survival (PFS) of 15.1<br />

weeks, an overall survival (OS) of 61.0 weeks and 10% objective<br />

response (OR). 3 HER-reprogramming appears to play an important<br />

role in resistance 4 and recent clinical trial results have indicated that<br />

targeting more than one ErbB Family member increases efficacy. 5<br />

Thus afatinib, as an ErbB Family Blocker, should add to the portfolio<br />

of drugs available to treat trastuzumab resistance in HER2-positive<br />

BC patients. This is currently being tested in the LUX-<strong>Breast</strong> 1 trial.<br />

Methods: LUX-<strong>Breast</strong> 1 (NCT01125566) is a Phase III, openlabel,<br />

multicenter trial evaluating the efficacy and safety of afatinib<br />

+ vinorelbine vs. trastuzumab + vinorelbine in patients with<br />

HER2-overexpressing MBC who progressed on, or after one prior<br />

trastuzumab-based treatment regimen. Patients are randomized 2:1<br />

to afatinib (40 mg/day oral) + vinorelbine (i.v. 25 mg/m 2 /week)<br />

or trastuzumab (i.v. 2 mg/kg/week after 4 mg/kg loading dose)<br />

+ vinorelbine (i.v. 25 mg/m 2 /week). Patients receive continuous<br />

treatment in the absence of disease progression or unacceptable<br />

toxicity. Key eligibility criteria include histologically-confirmed<br />

HER2-positive MBC; no prior treatment with vinorelbine or HER2-<br />

targeted treatment other than trastuzumab; progression on one prior<br />

trastuzumab based regimen in either the adjuvant (or


December 4-8, 2012<br />

<strong>Abstract</strong>s: Poster Session OT1<br />

retreatment with paclitaxel (i.e. should not have been pretreated with<br />

paclitaxel within the past 12 months), or are eligible for treatment with<br />

vinorelbine (i.e. should not have been pretreated with vinorelbine).<br />

Exclusion criteria include inadequate cardiac, renal, hepatic and<br />

hematological function, pre-existing gastrointestinal dysfunction,<br />

rapidly progressing visceral MBC, interstitial lung disease, and active<br />

brain metastases. The primary endpoint is objective response (OR)<br />

and secondary endpoints include best overall response, duration of<br />

OR, progression-free survival (PFS) and safety. PFS and safety will<br />

be assessed separately for afatinib mono- and combination therapy.<br />

An early stopping rule was deployed to minimize the number of<br />

pts treated should afatinib be ineffective; once 20 evaluable pts<br />

(according to RECIST 1.1) completed at least two courses of afatinib<br />

(or progressed during the first course), a meeting was held to evaluate<br />

the objective tumor response rate and to decide whether to proceed<br />

with the trial or stop due to futility. If at least one unconfirmed OR<br />

had been witnessed from all available information at the time, then<br />

the trial was to continue to full accrual. This early stopping rule for<br />

futility has been passed and the trial will continue to full accrual. Pt<br />

enrollment began in May 2011 in ∼40 sites and five countries.<br />

1. Lin NU, et al. <strong>Breast</strong> <strong>Cancer</strong> Res Treat 2012. DOI: 10.1007/<br />

s10549-012-2003-y.<br />

*Updated abstract from ASCO 2012.<br />

OT1-2-01<br />

NSABP B-43: A phase III clinical trial to compare trastuzumab<br />

(T) given concurrently with radiation therapy (RT) to RT alone<br />

for women with HER2+ DCIS resected by lumpectomy (Lx)<br />

Cobleigh MA, Anderson SJ, Julian TB, Siziopikou KP, Arthur DW,<br />

Rabinovitch R, Zheng P, Mamounas EP, Wolmark N. National<br />

Surgical Adjuvant <strong>Breast</strong> and Bowel Project, Pittsburgh, PA; Rush<br />

University Medical Center, Chicago, IL; University of Pittsburgh<br />

Graduate School of Public Health, Pittsburgh, PA; Allegheny General<br />

Hospital, Pittsburgh, PA; Northwestern University Feinberg School<br />

of Medicine, Chicago, IL; Virginia Commonwealth University,<br />

Richmond, VA; University of Colorado, Aurora, CO; Aultman<br />

Hospital, Canton, OH<br />

Background: A significant amount of DCIS is ER negative and/or<br />

overexpresses HER2. This provides an opportunity to test molecular<br />

therapy in DCIS.<br />

In xenograft models and cell lines, T boosts RT effectiveness. In<br />

T-treated HER2+ patients, apoptosis occurs within 1 wk of single<br />

agent T use, with T found in ductal aspirates. Ample safety evidence<br />

for T exists. T given during whole breast irradiation (WBI) may<br />

improve results for lumpectomy (Lx) resected HER2+ DCIS. A trial<br />

to examine this question will enhance the understanding of breast<br />

tumor biology and the prevention of such tumors and could possibly<br />

extend breast-conserving surgery benefits for women with DCIS.<br />

Method: After Lx for pure DCIS, each patient’s DCIS lesion is<br />

centrally tested for HER2 by IHC analysis. HER2 2+ tumors undergo<br />

FISH analysis. HER2 3+ or FISH+ patients can be randomly assigned<br />

to 2 doses of T, 3 weeks apart during WBI or to WBI alone.<br />

Women ≥18 yrs. with a margin-clear Lx for pure DCIS, with ECOG<br />

status 0/1 who are clinically or pathologically node negative are<br />

eligible. Centrally tested DCIS must be HER2 +. ER and/or PR status<br />

must be known before randomization.<br />

Primary aims are to determine if T decreases ipsilateral breast cancer<br />

recurrence, ipsilateral skin cancer recurrence, or ipsilateral DCIS.<br />

Secondary aims are to determine the benefit of T in preventing<br />

regional or distant recurrence and contralateral invasive breast cancer<br />

or DCIS. B-43 will determine if DFS, recurrence-free interval, and<br />

OS can be improved with the use of T. 2000 patients will be accrued<br />

over 7.9 yrs, with a definitive analysis of primary endpoints performed<br />

at163 ipsilateral breast cancer events (7.5 - 8 yrs. after protocol<br />

initiation) with an 80% power to detect a hazard reduction of 36%,<br />

from 1.73 ipsilateral breast cancer events per 100 pt-yrs to 1.11 events<br />

per 100 pt-yrs. The 36% observed reduction in the hazard of IIBCR-<br />

SCR-DCIS on the T arm is based on a projection of 40% hazard<br />

reduction if the compliance were perfect, with a 10% noncompliance<br />

rate. As of 5-31-12, 939 patients have been randomized.<br />

NCT00769379Grant support: PHS NCI-U10-CA-69651, -12027, and<br />

NCI P30-CA-14599 from the US NCI and Genentech, Inc.<br />

OT1-2-02<br />

SHARE. A French multiceter phase III trial comparing<br />

accelerated partial irradiation (APBI) versus standard or<br />

hypofractionated whole breast irradiation in low risk of local<br />

recurrence breast cancer<br />

Belkacemi Y, Bourgier C, Kramar A, Lemonier J, Auzac G, Dumas I,<br />

Lacornerie T, Mijonnet S, Mege J-P, Lartigau E. APHP; GH Henry<br />

Mondor and UPEC, Paris, Créteil, France; Institut Gustave Roussy,<br />

Villejuif, France; Oscar Lambret Center, Lille, France; UNICANCER,<br />

Paris, France<br />

Introuction<br />

The six worldwilde ongoing APBI phase III trials are characterized<br />

by a significant heterogeneity regarding inclusion criteria and<br />

stratifications. One non-inferiority trial (SHARE) is currently ongoing<br />

in France.<br />

SHARE Trial design<br />

SHARE will randomize 2800 patients in 3 arms: standard (Arm A;<br />

6.5 weeks) versus hypofractionated (Arm B; 3 weeks) versus APBI<br />

(Arm C; 1 week) using only 3D conformal radiotherapy (3D CRT). In<br />

this trial high quality and homogeneous criteria in surgery, pathology<br />

and RT for the 3 arms is planed.<br />

Primary objective<br />

To estimate and compare the rates of local recurrences between the<br />

experimental and control arms.<br />

Inclusion criteria<br />

Post menopausal women aged ≥ 50y (stratification: < 70 yrs vs ≥<br />

70 yrs)<br />

Menopausal status confirmed since ≥ 12 months (Clinically and/or<br />

biologically)<br />

No previous ipsilateral breast and/or mediastinal irradiation<br />

Pathologic confirmation of invasive carcinoma (all types of invasive<br />

carcinomas)<br />

Unifocal tumor confirmed on the pathologic specimen<br />

Pathologic tumor size of the carcinoma ≤ 2cm (including the in situ<br />

component)<br />

All pathologic grades (stratification: HER2/triple negative vs others)<br />

Clear lateral margins confirmed on the final pathology report. The<br />

minimal size from the invasive and in situ disease should be 2 mm<br />

(≥ 2mm)<br />

pN0 (i+/-) (stratification: pN0 vs pN(i+))<br />

Chemotherapy and trastuzumab are not allowed - RT should be started<br />

≥ 4weeks and ≤ 12 weeks after surgery (including the date of second<br />

excision for close or involved margins)<br />

Clips in the tumor bed placed during surgery (4 to 5 clips)<br />

www.aacrjournals.org 565s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Treatment<br />

Arm A<br />

3D CRT should be started within 12 weeks after the last surgery. A<br />

total of 50 Gy is delivered in 25 fractions, one fraction of 2 Gy per<br />

day, 5 days a week. In this arm, the boost of 10 to 16 Gy is delivered<br />

in 5 to 8 fractions.<br />

Dose prescription (arm B)<br />

The main point is that in both schedules the total dose is delivered<br />

in 3 weeks:<br />

- The Canadian schedule described by Whelan et al. delivers 42.5 Gy<br />

in 16 fractions (2.65Gy/fraction, 5 fractions / week).<br />

- The UK schedule, reported in START B trial delivers 40 Gy in 15<br />

fractions (2.66 Gy/ fraction, 5 fractions / week). Nodal and boost<br />

RT is not allowed.<br />

Arm C<br />

3D CRT should be started within 12 weeks after the last surgery.<br />

Intensity modulated RT (IMRT) and brachytherpayare not allowed.<br />

A total dose of 40 Gy in 10 fractions over 1 week (4Gy per fraction<br />

twice a day with minimal delay of 6h).<br />

Conclusion<br />

In summary, the French trial will allow:<br />

- Selected patients according to age and low risk of local recurrence<br />

parameters.<br />

- To evaluate in a subgroup of patients (4 to 5 centers), the impact of<br />

magnetic resonance imaging (MRI) for patient selection and the rate<br />

of occult disease (multifocality) not detected by standard imaging<br />

evaluation which is considered as an exclusion criteria.<br />

- To determine APBI parameters adapted to the surgical procedure in<br />

the tumor bed remodeling setting.<br />

- To determine homogeneous criteria for the surgical procedure,<br />

minimal requirements and optimal criteria that have to be mentioned<br />

in the final pathology report in all 3 arms of the study.<br />

- To test hypofractionation using 3 weeks-schedules as compared<br />

to APBI<br />

OT2-1-01<br />

Feasibility of sentinel node detection after neoadjuvant<br />

chemotherapy for patient with proved axillary lymph node<br />

involvement: the French prospective multiinstitutional GANEA<br />

2 ongoing trial<br />

Classe J-M, Bordes V, Gimbergues P, Tunon de Lara C, Faure C,<br />

Belichard C, Houpeau J-L, Raro P, Dupré P-F, Houvenaeghel G,<br />

Barranger E, Marchal F, Deblay P, Rouanet P, Lefebvre C, Bourcier<br />

C, Alran S. Institut de <strong>Cancer</strong>ologie de l’Ouest, Nantes Saint Herblain,<br />

France; Centre Jean Perrin, Clermont-Ferrand, France; Institut<br />

Bergonié, Bordeaux, France; Centre Leon Berard, Lyon, France;<br />

Centre Huguenin, Saint Cloud, France; Centre Oscar Lambret, Lille,<br />

France; Centre Hospitalier Universitaire Morvan, Brest, France;<br />

Paoli Calmettes, Marseille, France; CHU Lariboisière, Paris,<br />

France; Centre Alexis Vautrin, Nancy, France; Centre


December 4-8, 2012<br />

<strong>Abstract</strong>s: Poster Session OT2<br />

OT2-1-02<br />

Clinical node negative breast cancer patients undergoing breast<br />

conserving therapy: follow-up versus sentinel lymph node biopsy<br />

van Roozendaal LM, Smidt ML, de Wilt HHW, van Dalen T, Strobbe<br />

LJA, van der Hage J, Tjan-Heijnen VCG, Linn SC, Serroyen JL.<br />

Maastricht University Medical Center, Maastricht, Netherlands;<br />

Radboud University Nijmegen Medical Center, Nijmegen,<br />

Netherlands; Diakonessen Hospital Utrecht, Utrecht, Netherlands;<br />

Canisius-Wilhelmina Hospital, Nijmegen, Netherlands; Netherlands<br />

<strong>Cancer</strong> Institute - Antoni van Leeuwenhoek Hospital, Amsterdam,<br />

Netherlands; Maastricht University, Maastricht, Netherlands<br />

Background<br />

The ACOSOG-Z0011 study demonstrated no additional value of<br />

complementary axillary lymph node dissection in case of limited<br />

axillary sentinel lymph node (SLN) metastases in breast cancer<br />

patients undergoing breast conserving therapy 1 . Since axillary<br />

ultrasound can exclude advanced axillary metastatic disease preoperative<br />

2 , there is ample opportunity to evaluate whether SLN<br />

biopsies are still indicated in a selected group of breast cancer patients.<br />

Trial design<br />

A prospective non-inferiority randomized multicenter trial was<br />

designed.<br />

<strong>Breast</strong> cancer patients with cT1-2N0 disease, with no evidence for<br />

axillary metastases on ultrasound, treated with breast conserving<br />

therapy (lumpectomy + whole breast radiotherapy), will be<br />

randomized between follow-up and SLN biopsy. If adjuvant systemic<br />

therapy is not indicated by tumor or patient characteristics, gene<br />

expression profiling may be used for evaluation. To assess Quality<br />

of Life, 3 validated questionnaires will be used: QLQ-C30, QLQ-BR<br />

23 and Lymph-ICF 3-5 .<br />

Eligibility criteria:<br />

- Women with histological confirmed cT1-2 invasive unilateral breast<br />

carcinoma<br />

- Clinical node negative: no palpable nodes at physical examination<br />

and an axillary ultrasound without signs of lymph node metastases<br />

(cyto-/histology if indicated)<br />

- Neoadjuvant systemic therapy is allowed.<br />

Specific aims<br />

Primary endpoint is the axillary recurrence rate. The number of<br />

delayed axillary dissections will be registered. Secondary endpoints<br />

are distant-disease free survival, overall survival, local recurrence,<br />

morbidity and Quality of Life.<br />

Statistical methods<br />

Based on a 5-year axillary recurrence free survival rate: failure rate<br />

of 0.99 for controls and a true failure rate of 0.96 (as considered<br />

acceptable) for study subjects. Overall, 2086 patients will be<br />

included to be able to reject the null hypothesis that the failure rate<br />

for experimental and control subjects is inferior by at least 5% (D =<br />

-5%) with probability of 0.8 and alpha of 5%.<br />

Present accrual and target accrual<br />

This study is expected to start in late 2012 after approval by the<br />

Ethical Medical Committee.<br />

References<br />

1. Giuliano, A.E., et al., Axillary dissection vs no axillary dissection<br />

in women with invasive breast cancer and sentinel node metastasis:<br />

a randomized clinical trial. JAMA, 2011. 305(6): p. 569-75.<br />

2. Neal, C.H., et al., Can preoperative axillary US help exclude N2<br />

and N3 metastatic breast cancer? Radiology, 2010. 257(2): p. 335-41.<br />

3. Aaronson, N.K., et al., The European Organization for Research<br />

and Treatment of <strong>Cancer</strong> QLQ-C30: a quality-of-life instrument for<br />

use in international clinical trials in oncology. J Natl <strong>Cancer</strong> Inst,<br />

1993. 85(5): p. 365-76.<br />

4. Sprangers, M.A., et al., The European Organization for Research<br />

and Treatment of <strong>Cancer</strong> breast cancer-specific quality-of-life<br />

questionnaire module: first results from a three-country field study.<br />

J Clin Oncol, 1996. 14(10): p. 2756-68.<br />

5. Devoogdt, N., et al., Lymphoedema Functioning, Disability and<br />

Health questionnaire (Lymph-ICF): reliability and validity. Phys<br />

Ther, 2011. 91(6): p. 944-57.<br />

OT2-1-03<br />

The Z11 design for breast cancer patients undergoing a<br />

mastectomy<br />

van Roozendaal LM, Smidt ML, de Wilt HHW, van Dalen T, Strobbe<br />

LJA, van der Hage J, Tjan-Heijnen VCG, Linn SC, Serroyen JL.<br />

Maastricht University Medical Center, Maastricht, Netherlands;<br />

Radboud University Nijmegen Medical Center, Nijmegen,<br />

Netherlands; Diakonessen Hospital Utrecht, Utrecht, Netherlands;<br />

Canisius-Wilhelmina Hospital, Nijmegen, Netherlands; Netherlands<br />

<strong>Cancer</strong> Institute - Antoni van Leeuwenhoek Hospital, Amsterdam,<br />

Netherlands; Maastricht University, Maastricht, Netherlands<br />

Background<br />

The diagnostic work-up and treatment of axillary lymph nodes in breast<br />

cancer patients is an ongoing topic of research. The ACOSOG-Z0011<br />

study demonstrated no additional value of complementary axillary<br />

lymph node dissection (cALND) in case of limited axillary sentinel<br />

lymph node (SLN) metastases in breast cancer patients undergoing<br />

breast conserving therapy 1 . It is questionable whether these results<br />

can be applied to patients undergoing a mastectomy 2 .<br />

Trial design<br />

A prospective non-inferiority randomized multicenter trial was<br />

designed.<br />

<strong>Breast</strong> cancer patients with cT1-2N0 disease treated with mastectomy<br />

and limited axillary SLN metastases will be randomized for followup<br />

versus complementary axillary treatment. To assess the Quality<br />

of Life and morbidity benefits of this experimental treatment, 3<br />

validated questionnaires will be used: QLQ-C30, QLQ-BR 23 and<br />

Lymph-ICF 3-5 .<br />

Eligibility criteria<br />

- Women with histological confirmed cT1-2 invasive unilateral breast<br />

carcinoma<br />

- Clinical node negative: no palpable nodes in physical examination<br />

and the axillary ultrasound without signs of lymph node metastases<br />

(cyto-/histology if indicated)<br />

- Sentinel lymph node biopsy must contain at least one and a<br />

maximum of 3 (micro)metastases<br />

- Neoadjuvant systemic therapy is allowed<br />

Specific aims<br />

Primary endpoint is the axillary recurrence rate. The number of<br />

delayed axillary dissections will be registered. Secondary endpoints<br />

are distant-disease free survival, overall survival, local recurrence,<br />

morbidity and Quality of Life.<br />

www.aacrjournals.org 567s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Statistical methods<br />

Based on 5-year axillary recurrence free survival rate, a failure<br />

rate of 0.98 among controls and a true failure rate of 0.96 for study<br />

subjects are considered acceptable. Overall, 1114 patients will be<br />

included to be able to reject the null hypothesis that the failure rate<br />

for experimental and control subjects is inferior by at least 5% (D =<br />

-5%) with probability of 0.8 and alpha of 5%.<br />

Present accrual and target accrual<br />

This study is expected to start in late 2012 after approval by the<br />

Ethical Medical Committee.<br />

References<br />

1. Giuliano, A.E., et al., Axillary dissection vs no axillary dissection<br />

in women with invasive breast cancer and sentinel node metastasis:<br />

a randomized clinical trial. JAMA, 2011. 305(6): p. 569-75.<br />

2. Morrow, M. and A.E. Giuliano, To cut is to cure: can we really<br />

apply Z11 in practice? Ann Surg Oncol, 2011. 18(9): p. 2413-5.<br />

3. Aaronson, N.K., et al., The European Organization for Research<br />

and Treatment of <strong>Cancer</strong> QLQ-C30: a quality-of-life instrument for<br />

use in international clinical trials in oncology. J Natl <strong>Cancer</strong> Inst,<br />

1993. 85(5): p. 365-76.<br />

4. Sprangers, M.A., et al., The European Organization for Research<br />

and Treatment of <strong>Cancer</strong> breast cancer-specific quality-of-life<br />

questionnaire module: first results from a three-country field study.<br />

J Clin Oncol, 1996. 14(10): p. 2756-68.<br />

5. Devoogdt, N., et al., Lymphoedema Functioning, Disability and<br />

Health questionnaire (Lymph-ICF): reliability and validity. Phys<br />

Ther, 2011. 91(6): p. 944-57.<br />

OT2-1-04<br />

Intraoperative assessment of tumor margins with a new optical<br />

imaging technology: A multi-center, randomized, blinded clinical<br />

trial<br />

Jacobs LK, Carney PS, Cittadine AJ, McCormick DT, Somera<br />

AL, Darga DA, Putney JL, Adie SG, Ray P, Cradock KA, Tafra L,<br />

Gabrielson EW, Boppart SA. The Johns Hopkins University School<br />

of Medicine, Baltimore, MD; University of Illinois, Urbana, IL;<br />

Diagnostic Photonics, Inc, Chicago, IL; Carle Foundation Hospital,<br />

Urbana, IL; AdvancedMEMS, <strong>San</strong> Francisco, CA; Anne Arundel<br />

Medical Center, Annapolis, MD<br />

Background<br />

Partial mastectomy is the most commonly performed procedure<br />

for invasive breast cancer and is associated with a reexcision rate<br />

commonly ranging from 20% to 40% in the literature. This high rate<br />

of reexcision is associated with significant additional cost (estimated<br />

over $4,000 per reexcision) and lower quality outcomes.<br />

Optical coherence tomography (OCT) is a high-resolution imaging<br />

technology that images tissue structure with micron-scale resolution<br />

– on the same scale as histopathology. It is similar to ultrasound<br />

except it uses near infra-red light waves instead of sound waves to<br />

create detailed images several millimeters deep into tissue. Although<br />

widely used in ophthalmology with growing use in cardiovascular<br />

imaging, high-resolution OCT imaging has a narrow depth of focus<br />

and requires instrumentation that is not well suited for intraoperative<br />

use. Drawing from OCT technology, interferometric synthetic aperture<br />

microscopy (ISAM) is a computational imaging technique that creates<br />

high-resolution, always in-focus images in software with basic optical<br />

instrumentation. A high-resolution ISAM probe and imaging system<br />

has been developed for intraoperative imaging of tissue structure and<br />

has the potential to broadly impact intraoperative assessment of tumor<br />

margins. Intraoperative ISAM imaging of the excised breast cancer<br />

specimen margins and in vivo imaging within the surgical cavity may<br />

reduce the high rate of reexcision associated with partial mastectomy.<br />

Trial Design<br />

The trial design is a prospective, multi-center, randomized, double<br />

arm study comparing the reexcision rate of standard of care partial<br />

mastectomy versus the reexcision rate of standard of care partial<br />

mastectomy plus intraoperative ISAM imaging.<br />

Inclusion Criteria<br />

• Women histologically diagnosed with invasive carcinoma of the<br />

breast (invasive ductal or lobular)<br />

• Undergoing partial mastectomy (lumpectomy) procedure<br />

• Age 18 years or more<br />

Exclusion Criteria<br />

• Multicentric disease<br />

• Bilateral disease<br />

• Neoadjuvant systemic therapy<br />

• All T4 tumors<br />

• Previous radiation in the operated breast<br />

• Prior surgical procedure in the same quadrant<br />

• Implants in the operated breast<br />

• Pregnancy<br />

• Lactation<br />

• Participating in any other investigational study which can influence<br />

collection of valid data<br />

Primary Endpoints<br />

• Measure of surgical reexcision rate<br />

• Rate of tumor at final surgical margins<br />

Secondary Endpoints<br />

• Volume of tissue excised<br />

• Clinical and economic measures of addressing asymmetry<br />

Statistical Methods<br />

The trial is designed to show superiority of the ISAM imaging arm<br />

to the standard of care. Statistical design is two group, continuity<br />

corrected chi-squared test of equal proportions with 90% power and<br />

alpha=0.05. The trial design assumes a baseline reoperation rate in<br />

the standard of care arm of 24% with at least a 50% reduction in the<br />

ISAM imaging arm.<br />

Present Accrual and Target Accrual<br />

Not yet recruiting. Target accrual is 230 patients in the partial<br />

mastectomy + imaging arm and 230 patients in the standard of care<br />

partial mastectomy arm.<br />

OT2-2-01<br />

SOFT and TEXT: Trials of tamoxifen and exemestane with and<br />

without ovarian function suppression for premenopausal women<br />

with hormone receptor-positive early breast cancer<br />

Zickl L, Francis P, Fleming G, Pagani O, Walley B, Price KN, Gelber<br />

RD, Regan MM. International <strong>Breast</strong> <strong>Cancer</strong> Study Group, North<br />

American <strong>Breast</strong> <strong>Cancer</strong> Group<br />

Background<br />

The SOFT and TEXT randomized phase 3 trials address two primary<br />

questions for endocrine treatment of premenopausal women with<br />

hormone receptor-positive breast cancer. 1) In combination with<br />

ovarian function suppression (OFS), does an aromatase inhibitor<br />

(exemestane, E) improve outcome compared with tamoxifen (T)? 2)<br />

Does addition of OFS to T improve outcome compared with T alone?<br />

Trial Designs<br />

SOFT compares 5y of T to OFS+T or OFS+E. OFS can be GnRH<br />

analog (triptorelin) x 5y, oophorectomy or ovarian irradiation. Median<br />

age was 43y (11%


December 4-8, 2012<br />

<strong>Abstract</strong>s: Poster Session OT2<br />

TEXT compares 5y of OFS+T to OFS+E. Patients were enrolled<br />

prior to CT (if planned). Median age was 43y (9% 5y. The estimated power to<br />

detect a 20%, 25%, or 30% reduction in the hazard with OFS+E vs<br />

OFS+T is 63%, 84%, and 95%, respectively.<br />

The comparison of OFS+T to T alone is planned at 5y MFU (SOFT,<br />

n=2045). The estimated power to detect a 20%, 25%, 30%, or 33.5%<br />

reduction in the hazard is 34%, 52%, 69%, and 80%, respectively.<br />

Accruals<br />

SOFT Target: 3000; Final: 3066<br />

TEXT Target: 2639; Final: 2672<br />

Enrollment 2003-2011; primary analyses expected late 2013/early<br />

2014.<br />

Related Research<br />

Quality of Life (QL) component evaluates QL, menopausal symptoms<br />

and sexual impairment.<br />

TEXT Translational Research investigates patient and tumor features<br />

that may contribute to treatment effectiveness and side effects.<br />

TEXT-Bone investigates changes in bone mineral density and the<br />

role of serial serum markers of bone remodeling as predictors of<br />

bone side effects.<br />

Co-SOFT evaluates changes in cognitive function during the first year.<br />

SOFT-EST evaluates estrogen levels during the first 4y of GnRH<br />

analogue and whether there is a suboptimally estrogen-suppressed<br />

subgroup.<br />

North American Pharmacogenetics study investigates whether genetic<br />

variations that affect T and E metabolism influence efficacy.<br />

OT2-2-02<br />

Prospective multicentre study evaluating the effect of impaired<br />

tamoxifen metabolization on efficacy in breast cancer patients<br />

receiving tamoxifen in the neo-adjuvant or metastatic setting -<br />

The CYPTAM-BRUT 2 trial.<br />

Lintermans A, Dieudonné A-S, Blomme C, Lambrechts D, Wildiers H,<br />

Christiaens M-R, Timmerman D, Van Calster B, Decloedt J, Berteloot<br />

P, Joerger M, Zaman K, Dezentjé V, Neven P. University Hospitals<br />

Leuven, Belgium; Vesalius Research Center and VIB, Catholic<br />

University Leuven, Leuven, Belgium; AZ Sint-Blasius, Dendermonde,<br />

Belgium; AZ Sint-Maarten, Duffel, Belgium; Cantonal Hospital,<br />

Sint-Gallen, Switzerland; University Hospital CHUV, Lausanne,<br />

Switzerland; Leiden University Medical Center, Leiden, Netherlands<br />

Background<br />

Tamoxifen is often used for the prevention and treatment of hormone<br />

receptor positive breast cancer. It is metabolized in the liver through<br />

cytochrome P450 (CYP) enzymes to more active metabolites, of<br />

which endoxifen is considered the most important. It is known<br />

that certain genetic variants in CYP enzymes result in lower serum<br />

endoxifen levels, but their effect on tamoxifen efficacy is not yet clear<br />

as results remain contradictory.<br />

Trial design<br />

CYPTAM-BRUT 2 is a prospective multicentre open label single<br />

arm non randomized observational trial approved by the UZ Leuven<br />

Ethics Committee. Eligibility criteria are postmenopausal women with<br />

estrogen receptor positive invasive breast cancer receiving tamoxifen<br />

as first line therapy in the metastatic or neo-adjuvant setting. Prior<br />

adjuvant endocrine therapy is allowed if there is more than 12 months<br />

after completion of the adjuvant therapy.<br />

Primary endpoint is the relation between endoxifen levels and<br />

objective response rate. Secondary endpoints are the relation between<br />

endoxifen levels and the proportion of patients who are failure-free at<br />

6 months, clinical benefit, tolerability of tamoxifen and the predictive<br />

value of the tamoxifen activity score, based on the presence of genetic<br />

variants and CYP inhibiting drugs. Patients with bone-only lesions<br />

will only be included in the analysis of the secondary endpoints.<br />

The trial is designed to show an improvement of the objective tumor<br />

response rate from 5% in patients with an unfavorable endoxifen<br />

profile (steady-state concentrations < 90nM) to 20% in patients with<br />

a favorable endoxifen profile (steady-state concentrations ≥ 90nM).<br />

With a type I error of 5% and a power of 90%, 231 patients have<br />

to be included into the study. The main secondary study endpoint is<br />

progression-free survival (PFS) at 6 months. With a total number<br />

260 patients eligible for PFS, the study has a power of 80% to detect<br />

a 20% improvement of PFS at 6 months (i.e. from 50% to 70%) in<br />

patients with a favorable endoxifen as compared to patients with an<br />

unfavorable endoxifen profile, and a type-I error of 5%.<br />

Patient accrual<br />

Total sample size is 260 and currently 202 patients from 22<br />

participating centres in Belgium and Switzerland are included (May<br />

2012).<br />

www.aacrjournals.org 569s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

OT2-2-03<br />

Dovitinib (TKI258) or placebo in combination with fulvestrant in<br />

postmenopausal, endocrine-resistant HER2–/HR+ breast cancer:<br />

a phase II study<br />

Andre F, Greil R, Denduluri N, Barrios C, Campone M, Cortes<br />

J, Neven P, Reddick C, Squires M, Zhang Y, Yovine A, Blackwell<br />

K. Institut Gustave Roussy, Villejuif, France; Medizinische<br />

Universitätsklinik Salzburg mit Hämatologie, Salzburg, Austria;<br />

Virginia <strong>Cancer</strong> Specialists, US Oncology, Arlington, VA; Pontifícia<br />

Universidade Católica do Rio Grande do Sul School of Medicine,<br />

Porto Alegre, Brazil; Institut de Cancérologie de l’Ouest-René<br />

Gauducheau, Saint-Herblain, France; Vall d’Hebron Institute<br />

of Oncology, Barcelona, Spain; Hospital Gasthuisberg, Leuven,<br />

Belgium; Novartis Pharmaceuticals Corporation, East Hanover, NJ;<br />

Novartis Pharma AG, Basel, Switzerland; Duke University Medical<br />

Center, Durham, NC<br />

Background: Overcoming endocrine resistance is a critical goal<br />

in the treatment of hormone receptor-positive (HR+) breast cancer.<br />

Molecular mechanisms associated with endocrine resistance include<br />

adaptive cross-talk between the estrogen receptor and the fibroblast<br />

growth factor receptor (FGFR). Up to 8% of HR+/ human epidermal<br />

growth factor receptor 2 negative (HER2–) breast cancer patients<br />

have amplification of the FGFR1 gene, which is associated with<br />

resistance to endocrine therapy. In preclinical models, resistance to<br />

endocrine therapy can be overcome via FGFR1 inhibition. Dovitinib<br />

is a potent oral inhibitor of receptor tyrosine kinases, including FGFR,<br />

vascular endothelial growth factor receptor (VEGFR), and platelet<br />

derived growth factor receptor (PDGFR), that demonstrated antitumor<br />

activity in heavily pretreated breast cancer patients with FGF-pathway<br />

amplification (FGFR1, FGFR2, or ligand FGF3; Andre et al, ASCO<br />

2011). Dovitinib may reverse resistance to endocrine therapy related<br />

to FGF-pathway amplification and may also inhibit angiogenesis,<br />

which plays an essential role in breast cancer development. Dovitinib<br />

is studied here in combination with fulvestrant to determine if it<br />

can improve outcomes in postmenopausal patients with endocrine<br />

resistant HER2-/HR+ breast cancer.<br />

Methods: This is a multicenter, randomized, double-blind, placebocontrolled,<br />

phase II trial that will enroll postmenopausal HER2–/<br />

HR+ locally advanced or metastatic breast cancer patients (N ≈ 150)<br />

progressing within 12 months of completion of adjuvant endocrine<br />

therapy or after ≤ 1 prior endocrine therapy in the advanced setting.<br />

Patients prospectively undergo molecular screening to enrich for<br />

FGF amplification (FGFR1, FGFR2, or FGF3 amplification by<br />

qualitative polymerase chain reaction (qPCR); 45 amplified and 30<br />

nonamplified patients per arm). Patients are randomized 1:1 (stratified<br />

by FGF-amplification and presence of visceral disease) to receive<br />

fulvestrant intramuscularly (500 mg q4w [with an additional dose<br />

2 weeks after the initial dose]) in combination with oral dovitinib<br />

(500 mg, 5 days on/2 days off) or placebo until disease progression,<br />

unacceptable toxicity, death or discontinuation due to any reason<br />

(eg, withdrawal). Crossover is not permitted. The primary endpoint<br />

is progression-free survival, with tumor assessments performed<br />

q8w. Secondary endpoints include overall response rate per RECIST<br />

v1.1, duration of response, overall survival, Eastern Cooperative<br />

Oncology Group performance status and patient-reported outcome<br />

scores over time, and safety. Additionally, the pharmacodynamic<br />

effect of dovitinib on FGFR-associated angiogenic pathways in<br />

tumor specimens and potential predictive biomarkers of response to<br />

dovitinib will be explored.<br />

OT2-2-04<br />

A phase III randomized, placebo-controlled clinical trial<br />

evaluating the use of adjuvant endocrine therapy +/- one year of<br />

everolimus in patients with high-risk, hormone receptor- (HR)<br />

positive and HER2-negative breast cancer: SWOG/NSABP S1207.<br />

Chavez-Mac Gregor M, Barlow WE, Gonzalez-Angulo AM, Rastogi<br />

P, Mamounas EP, Ganz PA, Schott AF, Paik S, Lew DL, Bandos H,<br />

Hortobagyi GN. The University of Texas MD Anderson <strong>Cancer</strong> Center,<br />

Houston, TX; SWOG Statistical Center, Seattle, WA; NSABP/Four<br />

Allegheny Center, Pittsburgh, PA; Aultman <strong>Cancer</strong> Center, Canton,<br />

OH; UCLA Jonsson Comprehensive <strong>Cancer</strong> Center, Los Angeles,<br />

CA; University of Michigan, Ann Arbor, MI; NSABP Division of<br />

Pathology, Pittsburgh, PA; NSABP Biostatistical Center, Graduate<br />

School of Public Health, University of Pittsburgh, PA<br />

BACKGROUND: Abnormalities of the PI3kinase/AKT/mTOR<br />

signaling network are some of the most common molecular anomalies<br />

in breast cancer. This pathway has been associated with resistance to<br />

endocrine therapies among HR-positive breast tumors. Everolimus,<br />

an mTOR-inhibitor, has been shown to increase the biological activity<br />

of aromatase inhibitors. In the metastatic setting, everolimus in<br />

combination with tamoxifen or exemestane increased the progressionfree<br />

survival in patients previously treated with endocrine therapy.<br />

S1207 proposes to evaluate the role of everolimus used in combination<br />

with endocrine therapy in the adjuvant setting.<br />

SPECIFIC AIMS/TRIAL DESIGN: S1207 is a SWOG/NSABP<br />

randomized phase III double-blind, placebo-controlled clinical trial.<br />

The primary objective of the study is to assess whether the addition<br />

of one year of everolimus to standard adjuvant endocrine therapy<br />

improves invasive disease-free survival (DFS) among patients with<br />

high risk HR-positive breast cancer. Secondary objectives include<br />

overall survival, distant recurrence-free survival, safety, adherence<br />

and quality of life. Submission of tissue specimens/blood samples<br />

is required for translational studies. Patients will be randomized to<br />

receive standard adjuvant endocrine therapy (selected by treating<br />

physician) in combination with one year of everolimus (10 mg PO<br />

daily) or standard adjuvant endocrine therapy in combination with<br />

one year of matched placebo.<br />

ELIGIBILITY CRITERIA: Patients with histologically confirmed<br />

invasive breast cancer HER2-negative and HR-positive with high risk<br />

features including: 1) node-negative disease with tumors ≥2cm and<br />

a recurrence score (RS) >25; 2) 1-3 positive nodes and RS >25; 3)<br />

patients with ≥4 positive lymph nodes regardless of RS. Patients with<br />

≥4 positive lymph nodes after completing neoadjuvant chemotherapy<br />

are also eligible. All patients must have completed surgery, adjuvant/<br />

neoadjuvant chemotherapy, and radiation therapy (if indicated) before<br />

registration. Prior exposure to mTOR inhibitors is not allowed.<br />

STATISTICAL METHODS/TARGET ACCRUAL: This is a<br />

parallel randomization design with equal allocation to the two<br />

treatment groups (everolimus and placebo). Randomization is<br />

stratified by 4 risk groups. All analyses are intent-to-treat by<br />

randomized assignment of eligible patients. The study plans to<br />

randomize 3,500 patients over a 3.5-year accrual period with the<br />

primary analysis conducted 3 years after the last patient is randomized.<br />

The study has 90% power (with 2-sided α=0.05) to detect an effective<br />

hazard ratio of 0.75 for everolimus versus placebo, corresponding<br />

to a gain in DFS of approximately 4.3% at 5 years. All patients will<br />

be followed for 10 years to assess overall survival and late adverse<br />

events. The expected trial duration from activation to reporting of<br />

DFS is about 7 years.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

570s<br />

<strong>Cancer</strong> Research


December 4-8, 2012<br />

<strong>Abstract</strong>s: Poster Session OT2<br />

OT2-2-05<br />

A prospective, randomised multi-centre phase II study evaluating<br />

the adjuvant, neoadjuvant or palliative treatment with tamoxifen<br />

+/- GnRH analogue versus aromatase inhibitor + GnRH analogue<br />

in male breast cancer patients (GBG-54 MALE)<br />

Linder M, von Minckwitz G, Kamischke A, Rudlowski C, Eggemann<br />

H, Nekljudova V, Loibl S. German <strong>Breast</strong> Group, Neu-Isenburg;<br />

Kinderwunschzentrum Münster, Germany; Universitätsfrauenklinik<br />

Bonn; Universitätsfrauenklinik Magdeburg<br />

Background<br />

<strong>Breast</strong> cancer (BC) in men is a rare disease accounting for 0.5-1%<br />

of all BC. The only available information on endocrine therapies<br />

derives from few retrospective case series and studies with small<br />

numbers of patients (pts). Treatment strategies are not based on<br />

data from prospective, randomised clinical studies and optimal<br />

therapy is unknown. Current clinical management is extrapolated<br />

from principles established for female BC. As 90% of male pts have<br />

hormone receptor positive BC, they receive tamoxifen 20mg as<br />

standard adjuvant therapy.<br />

Although women benefit from treatment with aromatase inhibitors<br />

(AI), only case reports exist of men treated with AI. Data from other<br />

entities show, that AI only suppresses oestradiol of 40-50% with<br />

an increase of testosterone. Among men on AIs, the hypothalamicpituitary<br />

feedback loop results in an increase substrate for<br />

aromatisation. By adding a gonadotropin-releasing hormone analogue<br />

(GnRH), the feedback loop would be interrupted and complete<br />

oestrogen suppression may be achieved.<br />

Patients and Methods<br />

The MALE study is a prospective, randomized, phase II study in<br />

which 48 male pts will be included in the adjuvant, neoadjuvant<br />

and metastatic setting. In a 3-arm design, endocrine therapies are<br />

compared consisting of tamoxifen 20mg daily versus tamoxifen<br />

daily+GnRH on day 1 and after 3 months versus exemestane<br />

25mg daily+GnRH on day 1 and after 3 months. The treatment<br />

duration is 6 months. The primary aim is to determine the oestradiol<br />

suppression after 3 months. Secondary endpoints are to compare the<br />

oestradiol suppression after 6 months and to assess the compliance<br />

(with standardized questionnaires as Aging Male Symptom Score,<br />

International Index of Erectile function and International Prostate<br />

Symptom Score), safety and toxicity profile. The determination of<br />

the predefined hormones at baseline and after 3 and 6 months will be<br />

measured centrally. After the antihormonal treatment, all pts should<br />

be treated according to local recommendations. Included can be men<br />

with primary or metastatic endocrine sensitive BC with an indication<br />

of an endocrine therapy who did not receive prior antihormonal<br />

therapy; prior chemotherapy is allowed.<br />

16 evaluable pts per group are needed for the Kruskal-Wallis-Test to<br />

have 80% power to detect at the 5% significance level a difference.<br />

The translational research programme includes the determination of<br />

cytochrome P450 polymorphisms, the hormone receptor activity and<br />

aromatase expression of the tumour tissue.<br />

Results<br />

The time of recruitment should not exceed 2 years with 36 recruiting<br />

sites in Germany. As no study specific treatment or investigation is<br />

planned after the end of treatment, surgery and follow up are not<br />

part of this study, however information on the health status of the<br />

pts will be collected. The trial has been opened in May 2012 and is<br />

currently recruiting.<br />

Conclusion<br />

This is the first study randomizing men with BC to investigate the<br />

ability to suppress estradiol of different endocrine regimen.<br />

OT2-3-01<br />

Phase Ib pilot study to evaluate reparixin in combination with<br />

chemotherapy with weekly paclitaxel in patients with HER-2<br />

negative metastatic breast cancer (MBC)<br />

Schott AF, Wicha M, Cristofanilli M, Ruffini P, McCanna S, Reuben<br />

JM, Goldstein LJ. University of Michigan, Ann Arbor, MI; Fox Chase<br />

<strong>Cancer</strong> Center, Philadelphia, PA; Dompé s.p.a., Milano, Italy; MD<br />

Anderson <strong>Cancer</strong> Center, Houston, TX<br />

Background: <strong>Breast</strong> cancer stem cells (BCSCs) have the ability<br />

to self renew and generate the full range of cells that make up a<br />

bulk tumor. Chemokine receptor 1 (CXCR1) is almost exclusively<br />

expressed in the aldehyde dehydrogenase positive (ALDH+) BCSC<br />

population compared with its expression in bulk tumor cells. CXCR1<br />

is a receptor for CXCL8 (previously IL8), a proinflammatory<br />

chemokine implicated in the metastasis and progression of multiple<br />

malignancies, including breast cancers. CXCL8 has also been shown<br />

to stimulate self-renewal of BCSCs in vitro. Tissue damage induced<br />

by chemotherapeutic agents may induce CXCL8 as part of the injury<br />

response. This suggests that strategies aimed at interfering with the<br />

CXCL8/CXCR1 axis, activated by conventional chemotherapy (CT),<br />

may be able to target BCSCs and increase the efficacy of current<br />

therapies. Reparixin, a low molecular weight blocker of CXCL8<br />

biological activity, reduced ALDH-1+ cells in a human breast cancer<br />

xenograft when administered alone or in combination with taxane<br />

chemotherapy, and reduced metastases. Based on these findings, a<br />

Phase Ib study to determine the safety of paclitaxel plus reparixin<br />

therapy, and to explore the effects of reparixin on BCSCs and the<br />

tumor microenvironment, was initiated. Design and Treatment:<br />

Patients will receive a three-day run-in with reparixin oral tablets<br />

3 times/day (tid) followed by paclitaxel 80 mg/m2/week (Days 1,<br />

8, and 15 for 28-day cycle) + reparixin oral tablets tid.for 21 days.<br />

Three dose levels of 3-6 subjects will be explored in total: Dose level<br />

1 = 400 mg oral reparixin tid; Dose level 2 = 800 mg reparixin tid;<br />

and Dose level 3 = 1200 mg reparixin tid. An additional 6 subjects<br />

will be enrolled at the highest tolerated dose. Safety will be assessed<br />

following one cycle. Treatment continues as long as clinical benefit<br />

is observed. Main Eligibility Criteria: Patients must be female aged<br />

> 18 years with HER-2 negative MBC, eligible for treatment with<br />

paclitaxel (not taxane-refractory), have had up to 3 prior CT lines for<br />

advanced breast cancer (not including neo/adjuvant chemotherapy),<br />

must have measurable disease according to RECIST criteria version<br />

1.1, have ECOG PS of 0-1, have adequate organ function, and have<br />

no brain metastases. Objectives: Primary: To evaluate the safety<br />

and pharmacokinetic (PK) profile of the combination treatment.<br />

Secondary: 1) To evaluate tumor biopsies for the effects of reparixin<br />

on BCSC markers and tumoral microenvironment; 2) To evaluate<br />

blood samples for a) enumeration and molecular characterization<br />

of circulating tumor cells (CTCs), biomarker profiling for BCSC<br />

and epithelial-mesenchymal transition (EMT), and b) inflammatory<br />

cytokines; 3) To assess disease response for indication of efficacy.<br />

Statistical Methods: All tumor data collected will be reviewed and<br />

listed. No formal statistical analysis of this data is planned. Safety<br />

and tolerability analysis will be applied on the safety population with<br />

descriptive statistics for the safety variables.Summary statistics will<br />

be performed for each PK parameter and correlative study parameter.<br />

Accrual status: To date, four subjects have been enrolled at the first<br />

dose level. Target accrual is 15-24 subjects.<br />

www.aacrjournals.org 571s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

OT2-3-02<br />

Phase Ib/II study of an oral PI3K/mTOR inhibitor plus letrozole<br />

compared with letrozole (L) in pre-operative setting in patients<br />

with Estrogen Receptor-positive, HER2-negative early breast<br />

cancer (BC): Phase Ib preliminary data.<br />

Canon JL, Bergh J, Saura C, Oliveira M, Houk B, Millham R, Barton<br />

J, Dowsett M, Giorgetti C. Grand Hopital de Charleroi, Charleroi,<br />

Belgium; Karolinska Institutet and University Hospital, Stockholm,<br />

Sweden; Vall d’Hebron University Hospital, Vall d’Hebron Institute<br />

of Oncology (VHIO), Barcelona, Spain; Pfizer Oncology, La Jolla;<br />

Pfizer Oncology, Groton; Pfizer Biotechnology Unit & Oncology<br />

Clinical Research, <strong>San</strong> Diego; Royal Marsden Hospital, London,<br />

United Kingdom; Pfizer Oncology, Milan, Italy<br />

Background: Acquired hormone independent BC cell growth is<br />

associated with upregulation of the PI3K/mTOR pathway in vitro<br />

and in neo-adjuvant studies of aromatase inhibition (Ghazoui AACR<br />

<strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong>, 2011). Preclinical and clinical studies<br />

suggest that the combination of hormone therapy and a PI3K/mTOR<br />

inhibitor can restore sensitivity to hormone resistant tumours. Luminal<br />

B BCs are more frequently associated with endocrine resistance,<br />

hyperactivation of the PI3K signaling pathway, and increased<br />

expression of the Ki-67 proliferation marker than Luminal A BCs.<br />

This particular population may be a suitable target for PI3K directed<br />

therapy. A reduction in Ki-67 in the neo-adjuvant setting by endocrine<br />

therapy has been shown to correlate with efficacy of that same<br />

therapeutic in the adjuvant setting. Thus, a pre-operative study in high<br />

Ki-67-expressing BCs with short term reduction in Ki-67 as a primary<br />

endpoint could be used to support a study in the adjuvant setting with<br />

a conventional disease free endpoint. Study Design: The study is<br />

conducted in 2 phases, a Phase Ib followed by a randomized Phase<br />

II. The Phase Ib is designed to assess the tolerability of PF-04691502,<br />

an oral PI3K/mTOR antagonist, combined with L in postmenopausal<br />

women with ER positive, HER-2 negative advanced BC. A total of 10<br />

patients will be enrolled in this portion of the study. PF-04691502+L<br />

will be administered until progression of disease. After up to 2 months<br />

of treatment, the safety profile of PF-04691502+L will be evaluated<br />

prior to the initiation of the Phase II portion in a pre-operative setting.<br />

A total of 204 postmenopausal women with ER positive, HER-2<br />

negative early BC selected based on a high level of Ki-67 (>10%)<br />

will be enrolled in the Phase II portion. Patients will be randomized<br />

to receive PF-04691502+L or L alone for 6 weeks. The Phase II is<br />

designed to test the hypothesis that the addition of PF-04691502 to L<br />

can reduce Ki-67 levels after 6 weeks of administration to a greater<br />

degree than L alone, thus supporting the concept that the compound<br />

might mitigate the intrinsic or acquired resistance to hormonal<br />

therapy in a high risk patient population in the adjuvant setting.<br />

Patients will undergo 3 sequential core-biopsies (baseline, weeks 2<br />

and 6) to compare the changes in Ki-67 value from baseline to the<br />

end of the treatments and to test pharmacodynamic effects associated<br />

with PI3K/mTOR pathway modulation. Results: As of May 2012,<br />

8 postmenopausal women with advanced BC were enrolled in the<br />

phase Ib. All patients received PF-04691502 8 mg/day and L 2.5<br />

mg/day. Treatment is ongoing in all patients. The combination has a<br />

manageable safety profile although the median time of observation<br />

is short (47 days). A full set of safety data of patients enrolled in the<br />

Phase Ib will be presented. Also pharmacokinetic profile of L alone<br />

and in combination with PF-04691502 will be shown.<br />

OT2-3-03<br />

Denosumab versus placebo as adjuvant treatment for women<br />

with early-stage breast cancer at high risk of disease recurrence<br />

(D-CARE): An international, randomized, double-blind, placebocontrolled<br />

phase 3 clinical trial<br />

Goss PE, Barrios CH, Chan A, Finkelstein DM, Iwata H, Martin<br />

M, Braun A, Ke C, Maniar T, Coleman RE. Massachusetts General<br />

Hospital, Boston, MA; PUCRS School of Medicine, Porto Alegre,<br />

Brazil; <strong>Breast</strong> <strong>Cancer</strong> Research Centre, Perth, WA, Australia; Aichi<br />

<strong>Cancer</strong> Center, Nagoya, Chikusa-ku, Japan; Hospital Gregorio<br />

Maranon, Madrid, Spain; Amgen Inc., Thousand Oaks, CA; CR-UK/<br />

YCR Sheffield <strong>Cancer</strong> Research Centre, Sheffield, United Kingdom<br />

Bone represents approximately 40% of all first recurrences in women<br />

with early-stage breast cancer and is a common site of distant<br />

recurrence. In preclinical studies, RANKL inhibition significantly<br />

delays skeletal tumor formation, reduces skeletal tumor burden, and<br />

prolongs survival of tumor-bearing mice. Denosumab is approved<br />

for the prevention of skeletal-related events (SREs) in patients with<br />

established bone metastases from solid tumors.<br />

Purpose: The D-CARE trial is designed to assess if denosumab<br />

treatment prolongs bone metastasis-free survival (BMFS) and diseasefree<br />

survival (DFS) in the adjuvant breast cancer setting. The primary<br />

endpoint of this event-driven trial is BMFS. Secondary endpoints<br />

include DFS and overall survival. Additional endpoints are safety,<br />

breast density, incidence of SREs (following the development of bone<br />

metastasis) patient reported outcomes, and biomarkers.<br />

Methods: In this international, randomized, double-blind, and placebo<br />

controlled phase 3 trial, approximately 4,500 women with stage II or<br />

III breast cancer, at high risk for recurrence and with known hormone<br />

and HER-2 receptor status, are eligible. High risk is defined as biopsy<br />

evidence of breast cancer in regional lymph nodes, tumor size > 5<br />

cm, (T3), or locally advanced disease (T4). Standard-of-care adjuvant<br />

or neoadjuvant chemo-, endocrine, or HER-2 targeted therapy, alone<br />

or in combination, must be planned. Patients with a prior history of<br />

breast cancer (except ductal carcinoma in situ or lobular carcinoma<br />

in situ) or distant metastasis, oral bisphosphonate (BP) use within 1<br />

year of randomization, or any intravenous BP use, are not eligible.<br />

Patients are randomized 1:1 to receive denosumab 120 mg or placebo<br />

subcutaneously monthly for 6 months, then every 3 months for a<br />

total of 5 years of treatment. Supplemental vitamin D (≥ 400 IU)<br />

and calcium (≥ 500 mg) will be administered. The trial, sponsored<br />

by Amgen Inc. and registered with the ClinicalTrials.gov identifier<br />

NCT01077154, began enrolling patients in June 2010 and is expected<br />

to complete enrollment in late 2012.<br />

OT2-3-04<br />

A PILOT PHASE II STUDY TO EVALUATE THE IMPACT OF<br />

DENOSUMAB ON DISSEMINATED TUMOR CELLS (DTC) IN<br />

PATIENTS WITH EARLY STAGE BREAST CANCER (ESBC)<br />

Li J, Rugo HS. University of California, <strong>San</strong> Francisco, CA<br />

Background: Bone is the most common site of metastases in breast<br />

cancer (BC). The detection of keratin + cells (DTC) in bone marrow<br />

(BM) has been shown to correlate with an increased risk of disease<br />

recurrence. Receptor activator of nuclear factor kappa b (RANK)<br />

is critical to bone turnover. Pre-clinical studies have shown that<br />

the ligand to RANK (RANKL) can induce metastases of RANKexpressed<br />

tumor cells, and that treatment with a RANKL inhibitor can<br />

reduce BC metastases. Our previous study suggests that higher RANK<br />

expression in tumor at the time of diagnosis correlates with worse<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

572s<br />

<strong>Cancer</strong> Research


December 4-8, 2012<br />

<strong>Abstract</strong>s: Poster Session OT2<br />

recurrent free survival (RFS). Denosumab, a humanized RANKL<br />

inhibitor, has been approved in patients (pts) with osteoporosis and<br />

metastatic BC to bone.<br />

Trial design: We designed a nonrandomized, open label phase II<br />

pilot study evaluating the impact of denosumab on DTCs in pts with<br />

ESBC and DTCs following (neo) adjuvant chemotherapy. A total<br />

of 45 pts will be enrolled; treatment (Rx) consists of denosumab by<br />

subcutaneous injection monthly for 6 months, then every 12 weeks for<br />

6 months for a total of one year. BMA will be repeated at 6 and 12 mo;<br />

samples will be evaluated for DTC and for specific gene expression.<br />

Eligibility criteria: Eligible pts have completed adjuvant<br />

chemotherapy and at least 3 months (mo) of hormonal Rx as<br />

indicated, and have >10 DTC on screening bone marrow aspiration<br />

by immunomagnetic enrichment + flow cytometry (IE/FC).<br />

Specific aims: The primary efficacy endpoint is the reduction of<br />

DTC/ml from >10DTC/ml to ≤ 10DTC/ml at 6 mo and one year,<br />

and the feasibility of denosumab use in this setting. Secondary and<br />

exploratory endpoints are to evaluate the correlation of DTC at each<br />

time point with BC recurrence, and to analyze gene expression of<br />

markers related to bone destruction and inflammation in sorted DTC.<br />

Statistical methods: A total of 45 pts will be enrolled. Based on<br />

results from a previous trial testing zoledronic acid in a group of pts<br />

with similar eligibility, we expect to have evaluable BMA on 37 (82%)<br />

at 1 year. With standard Rx we expect 5% or fewer will have counts<br />

≤10 DTC/ml at 1 year. Using STATA routine sample size (SAMPSI:<br />

sample size and power for means and proportions), as long as 20% of<br />

pts in treated group become DTC-negative, we will have 89% power<br />

to detect this degree of efficacy. These calculations are based on an<br />

“exact” test of the proportion of successes; those with reductions to<br />

≤10DTC/ml in sample sizes of 37 based on one-sided testing at a 5%<br />

level of significance. We will compare the DTC/ml at 6 and 12 mo<br />

to baseline to evaluate reduction in counts.<br />

Present accrual and target accrual: trial will be open in July, 2012<br />

with target accrual of 45 pts.<br />

OT2-3-05<br />

AVASTEM: a phase II randomized trial evaluating anti-cancer<br />

stem cell activity of pre-operative bevacizumab and chemotherapy<br />

in breast cancer<br />

Gonçalves A, Pierga J-Y, Brain E, Tarpin C, Cure H, Esterni B,<br />

Boyer-Chammard A, Bertucci F, Jalaguier A, Houvenaeghel G, Viens<br />

P, Charaffe-Jauffret E, Extra J-M. Institut Paoli Calmettes, Marseille,<br />

France; Institut Curie-Centre René Huguenin, Paris, France; Institut<br />

Jean Godinot, Reims, France<br />

Background:<br />

Bevacizumab (BEV) in combination with chemotherapy (CT)<br />

improved response rate and progression-free survival without<br />

detectable impact on overall survival (OS) in metastatic breast cancer<br />

(BC). BC contains population of cells that display stem-cell properties.<br />

These cancer stem cells (CSC) are known to be relatively insensitive<br />

to cytotoxic chemotherapies and might play a major role in treatment<br />

resistance and relapse. Complex interactions exist between CSC and<br />

angiogenesis: whereas CSC may secrete VEGF and pro-angiogenic<br />

factors and may stay in VEGF-dependant vascular niche, recent<br />

preclinical data indicate that antiangiogenics may increase breast CSC<br />

population via hypoxia. CSC proportion may be monitored in human<br />

BC using IHC staining of ALDH1. AVASTEM trial was designed to<br />

evaluate whether the addition of BEV to conventional CT alters the<br />

proportion of CSC in BC patients receiving neo-adjuvant (NA) CT.<br />

Trial design:<br />

In this open-label phase II randomized study, patients are stratified<br />

by HER2 status and randomized 1:2 to receive NACT:<br />

- arm A (experimental): FEC100 (5FU 500 mg/m², Epirubicin 100<br />

mg/m², Cyclophosphamide 500 mg/m²) + BEV (15 mg/kg, 3-weekly)<br />

x 4 followed by Docetaxel (100 mg/m²) + BEV +/- trastuzumab (T, 8<br />

mg/kg and then 6 mg/kg, 3-weekly) if HER2-positive, x 4<br />

- arm B (control): FEC100 x 4 followed by Docetaxel 100 +/- T if<br />

HER2-positive, x 4.<br />

Tumor biopsies are required at baseline and after the first 4 cycles<br />

of FEC. After NACT, appropriate surgery and radiation therapy are<br />

performed, followed by adjuvant T and hormonal therapy, if indicated.<br />

Eligibility criteria:<br />

- Primary HER2-positive or -negative BC candidate to NACT<br />

(inflammatory breast cancer and synchronous metastatic disease<br />

are eligible)<br />

- Primary BC accessible to initial biopsy<br />

- Left ventricular ejection fraction ≥ 55%<br />

- Appropriate haematological, liver, renal and coagulation functions<br />

Specific aims:<br />

-Primary endpoint: to measure the variations of CSC proportion<br />

(by measuring the amount of IHC ALDH1-positive cells) in tumor<br />

biopsies obtained before C1 and after C4 in patients treated with<br />

BEV-based NACT or with NACT alone.<br />

- Secondary endpoints: tolerance, pCR, disease-free survival,<br />

recurrence-free survival and OS; ancillary studies (CSC<br />

characterization by CD44/CD24 IHC staining and evaluation of<br />

tumor-initiating properties of fresh tumor cells; CTC counts; contrastbased<br />

breast ultra-sonography; transcriptomics and proteomics)<br />

Statistical design:<br />

The difference between percentage of ALDH1-positive cells after 4<br />

cycles of BEV-based NACT and before treatment will be calculated.<br />

In the experimental arm, an equivalence test will be used to validate<br />

the hypothesis that the cell percentage does not increase more than<br />

5% during treatment (under the following hypothesis: no difference<br />

of expression, difference variability σ = 13, significance level of 0.05,<br />

power of 0.8). With the assumption of approximately 20% of missing<br />

data, 50 patients will be included in the tested arm. The control arm<br />

(n=25) is needed to confirm CSC increase during conventional CT.<br />

Present and target accrual:<br />

By May June 2012, 69 patients have been included. A total of 75<br />

patients are to be enrolled. Funded by INCA-PHRC 2009.<br />

OT2-3-06<br />

A phase II, non-randomized, multicenter, exploratory trial of<br />

single agent BKM120 in patients with triple-negative metastatic<br />

breast cancer.<br />

Saura C, Lin N, Ciruelos E, Maurer M, Lluch A, Gavilá J, Winer E,<br />

Baselga J, Rodón J. Vall d’Hebron University Hospital, Barcelona,<br />

Spain; Dana-Farber <strong>Cancer</strong> Institute, Boston, MA; Hospital 12<br />

de Octubre, Madrid, Spain; Columbia University Medical Center,<br />

New York; Hospital Clínico de Valencia, Valencia, Spain; Instituto<br />

Valenciano de Oncología, Valencia, Spain; Massachusetts General<br />

Hospital, Boston; SOLTI <strong>Breast</strong> <strong>Cancer</strong> Research Group, Barcelona,<br />

Spain<br />

Despite best available therapy, triple-negative breast cancer (TNBC)<br />

continues to be associated with poorer outcomes when compared to<br />

other breast cancer subtypes and poses a serious therapeutic challenge.<br />

Therefore, novel and more efficacious therapies are in great need. It<br />

is widely accepted nowadays, that TNBC is not homogeneous, but<br />

comprised of a number of subpopulations very different in genetic<br />

www.aacrjournals.org 573s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

make-up, oncogene dependence, and response to treatment. Among<br />

these, multiple core phosphatidylinositol-3-kinase (PI3K) pathway<br />

components are known to be altered in TNBC. The relevance of<br />

activating mutations in the PI3KCA gene or PTEN/INPP4B loss of<br />

protein expression on the virulence of breast cancer and response to<br />

PI3K inhibitors is yet to be elucidated. PI3K hyperactivity due to<br />

these alterations suggests that inhibitors of the PI3K pathway may<br />

be used to reverse resistance.<br />

BKM120 is a potent and highly specific oral pan-class I PI3K inhibitor<br />

currently in clinical development. This non-randomized, two-stage,<br />

open-label, single-arm exploratory trial is designed to elucidate the<br />

preliminary therapeutic value of BKM120 in patients with metastatic<br />

TNBC and to identify subpopulations that may mostly benefit.<br />

Women 18 years and above with confirmed mTNBC who have<br />

received at least two prior chemotherapy regimens in the adjuvant or<br />

metastatic setting are eligible to participate. Availability of a tumor<br />

block from the primary tumor or accessible metastasis for biopsy is<br />

mandatory. The primary objective is to determine the clinical activity<br />

of BKM120 defined as the clinical benefit rate (CR, PR or SD for<br />

more than 4 months per RECIST 1.1). Secondary objectives are to<br />

determine progression-free survival, overall survival, safety profile,<br />

pharmacodynamic effects, and to identify biomarkers predictive of<br />

response. BKM120 will be administered orally at a 100-mg dose, once<br />

daily, in a continuous schedule. Treatment will continue until disease<br />

progression, unacceptable toxicity, or decision of the physician or the<br />

patient to terminate participation.<br />

Stage 1 will include up to 50 patients with an intermediate analysis<br />

after the enrollment of the first 29 subjects. If no sufficient activity<br />

is observed, the clinical trial will be terminated. If drug activity is<br />

observed, enrollment will continue up Stage 1 completion. After<br />

50 patients are enrolled, a comprehensive analysis of molecular<br />

markers using technology and knowledge gathered in the Stand Up<br />

to <strong>Cancer</strong> Dream Team will take place. This analysis will be focused<br />

in identifying molecular markers predictive of response that would<br />

guide patient selection for Stage 2. Two clone investigator-initiated<br />

protocols (one for the US, one for Spain) are enrolling in parallel. A<br />

total of 110 patients will take part in this study in four sites in Spain<br />

and in 2 in the US: 50 patients in Stage 1 and 60 in Stage 2, exploring<br />

a maximum of 3 subgroups of 20 patients each. The trial is funded by<br />

a Stand Up to <strong>Cancer</strong> Dream Team Translational Research Grant, a<br />

Program of the Entertainment Industry Foundation (SU2C-AACR-<br />

DT0209) and supported by Novartis. Patient enrollment started in<br />

June 2012.<br />

OT2-3-07<br />

A randomized, phase 2 study of the poly (ADP-ribose) polymerase<br />

(PARP) inhibitor veliparib (ABT-888) in combination with<br />

temozolomide (TMZ) or in combination with carboplatin (C) and<br />

paclitaxel (P) versus placebo plus C/P in subjects with BRCA1 or<br />

BRACA2 mutation and metastatic breast cancer<br />

Isakoff SJ, Pulhalla S, Shepherd SP, Falotico N, Kaufman B,<br />

Friedlander M, Robson M, Domchek S, Garber J, McKeegan E, Chyla<br />

B, Qian J, Giranda VL. Massachusetts General Hospital, Boston, MA;<br />

University of Pittsburgh, PA; Abbott, Abbott Park, IL, Virgin Islands,<br />

British; Sheba Medical Center, Israel; Prince of Wales Hospital,<br />

Sydney, Australia; Memorial Sloan-Kettering <strong>Cancer</strong> Center, New<br />

York, NY; University of Pennsylvania, Philadelphia, PA; Dana Farber<br />

<strong>Cancer</strong> Institute, Boston, MA<br />

Background: Veliparib is a potent, oral PARP inhibitor that inhibits<br />

DNA repair, primarily through single strand break repair. BRCA1/2<br />

mutated tumors are defective in homologous recombination, leading<br />

to more error prone mechanisms of DNA repair and increased<br />

sensitivity to PARP inhibition. Veliparib and TMZ recently<br />

demonstrated promising activity in BRCA1/2 mutation carriers, and<br />

veliparib and C/P is being evaluated in a CTEP Phase 1 study. This<br />

Phase 2 multinational study will evaluate veliparib in combination<br />

with TMZ and in combination with C/P compared to an active,<br />

placebo-controlled arm of C/P in subjects with BRCA1 or BRCA2<br />

mutation and metastatic breast cancer.<br />

Methods: Approximately 240 subjects will be randomized in a 1:1:1<br />

ratio to: 1) Veliparib 40 mg BID D1-7 + TMZ (150 - 200 mg/m 2 QD<br />

D1-5) of every 28 day cycle; or 2) Veliparib 120 mg BID D1-7 +<br />

C (AUC 6, D3) and P (175 mg/m 2 , D3) of every 21 day cycle or 3)<br />

placebo BID D1-7 + C/P. Key eligibility includes known deleterious<br />

BRCA1/2 mutation, ≤1 prior chemotherapy for metastatic disease, and<br />

no prior platinum agent. Randomization will be stratified by hormone<br />

receptor positive versus negative, prior vs. no prior cytotoxic therapy,<br />

and ECOG 0-1 vs 2.<br />

The primary objective is progression-free survival (PFS) using<br />

RECIST 1.1. Secondary objectives include overall survival, objective<br />

response rate, duration of response and safety/tolerability. Optional<br />

paired tissue biopsies will be obtained at baseline and at progression<br />

to evaluate markers of response and resistance. Additional correlatives<br />

for response include assessment of peripheral blood, CTCs, and<br />

archived tissue for markers of disease status, gene methylation and<br />

mutational status, and tumor-specific alteration of cellular proteins/<br />

peptides and/or nucleic acids. Using the log-rank test for PFS, with<br />

150 PFS events the study will have ≥ 80% power at 2-sided α level<br />

of 0.05 to detect a statistically significant treatment effect assuming<br />

a true hazard ratio of 0.58 favoring the 2 treatment groups containing<br />

veliparib. As of June 11, 2012, 5 subjects have been enrolled.<br />

OT2-3-08<br />

Phase III randomized study of the oral pan-PI3K inhibitor<br />

BKM120 with fulvestrant in postmenopausal women with HR+/<br />

HER2– locally advanced or metastatic breast cancer, treated with<br />

aromatase inhibitor, and progressed on or after mTOR inhibitorbased<br />

treatment – BELLE-3<br />

Di Leo A, Germa C, Weber D, Di Tomaso E, Dharan B, Massacesi<br />

C, Hirawat S. <strong>San</strong>dro Pitigliani Hospital of Prato, Prato, Italy;<br />

Novartis Oncology, Paris, France; Novartis Institutes for BioMedical<br />

Research, Inc., Cambridge, MA; Novartis Pharmaceuticals<br />

Corporation, East Hanover, NJ<br />

Background: The PI3K/AKT/mTOR pathway is frequently activated<br />

in breast cancer (BC), and has been implicated in resistance to<br />

endocrine therapies and mTOR inhibitors (mTORi). BKM120 is<br />

an oral inhibitor of all 4 isoforms of class I PI3K (α, β, γ, δ). In<br />

combination with fulvestrant, BKM120 has demonstrated proapoptotic<br />

effects in vitro and anti-tumor activity in vivo. Preliminary<br />

clinical activity was observed with BKM120 in pts with BC, both as<br />

a single agent and in combination with letrozole (Mayer, et al. ASCO<br />

2012:#510), or trastuzumab (Saura, et al. SABCS 2011:#PD09-03).<br />

Use of the pan-PI3K inhibitor BKM120 may overcome resistance to<br />

mTORi in BC by targeting the PI3K pathway upstream; this will be<br />

explored in the current study. Stratification of pts according to PI3K<br />

activation status will definitively conclude whether this biomarker<br />

is predictive of response.<br />

Study design: This is a Phase III randomized, double-blind, placebocontrolled<br />

trial of BKM120 or placebo in combination with fulvestrant<br />

(BELLE-3; CBKM120F2303). Postmenopausal women with HR+/<br />

HER2– locally advanced or metastatic BC whose disease has been<br />

treated with aromatase inhibitor (AI), and is refractory to endocrine<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

574s<br />

<strong>Cancer</strong> Research


December 4-8, 2012<br />

<strong>Abstract</strong>s: Poster Session OT2<br />

and mTORi combination therapy (progression while on mTORi and<br />

endocrine therapy or within 30 days of end of treatment) are eligible<br />

for enrollment. Other eligibility criteria include: ≤1 previous line of<br />

chemotherapy for advanced disease; and available archival tumor<br />

tissue for analysis of PI3K-related biomarkers. Pts are randomized<br />

(2:1) to receive continuous BKM120 (100 mg) or placebo daily, and<br />

fulvestrant (500 mg). Randomization is stratified according to PI3K<br />

pathway activation (PIK3CA mut and/or PTEN mut and/or loss of<br />

PTEN protein expression by immunohistochemistry), and visceral<br />

disease status. Study treatment is given until disease progression or<br />

until study discontinuation for any reason.<br />

Co-primary endpoints are progression-free survival (PFS) in both full<br />

and PI3K pathway-activated populations based on local investigator<br />

assessment (RECIST 1.1). Key secondary endpoints are overall<br />

survival (OS) in both full and PI3K pathway-activated populations.<br />

Other secondary endpoints are PFS and OS (PI3K non-activated/<br />

unknown population), overall response rate and clinical benefit rate,<br />

safety (CTCAE 4.03), pharmacokinetics of BKM120, and patientreported<br />

quality of life (EORTC QLQ-C30 and QLQ-BR23).<br />

Statistical methods: PFS and OS will be estimated using the Kaplan–<br />

Meier method for full and PI3K pathway-activated populations. Both<br />

primary and key secondary endpoints will be tested using a stratified<br />

log-rank test at levels α governed by a graphical gate-keeping<br />

procedure. A stratified Cox regression will be used to estimate the<br />

hazard ratio, along with two-sided 95% CI.<br />

Target accrual: Estimated enrollment for BELLE-3 is 615 pts.<br />

Recruitment in this Global study is ongoing with planned enrollment<br />

of patients in North America, South America, Europe and Asia.<br />

OT2-3-09<br />

Phase III randomized study of the oral pan-PI3K inhibitor<br />

BKM120 with fulvestrant in postmenopausal women with HR+/<br />

HER2– locally advanced or metastatic breast cancer resistant to<br />

aromatase inhibitor – BELLE-2<br />

Baselga J, Campone M, Cortes J, Iwata H, de Laurentiis M, Jonat W,<br />

Di Tomaso E, Hachemi S, Goteti S, Germa C, Massacesi C, Arteaga<br />

C. Massachusetts General Hospital, Boston, MA; Centre René<br />

Gauducheau, Nantes, France; Vall d’Hebron Institute Oncology,<br />

Barcelona, Spain; Aichi <strong>Cancer</strong> Center Hospital, Nagoya, Aichi,<br />

Japan; Università degli Studi di Napoli Federico II, Naples, Italy;<br />

Christian-Albrechts-Universität zu Kiel, Kiel, Germany; Novartis<br />

Institutes for BioMedical Research, Inc., Cambridge, MA; Novartis<br />

Oncology, Paris, France; Novartis Pharmaceuticals Corporation,<br />

East Hanover, NJ; Vanderbilt-Ingram <strong>Cancer</strong> Center, Nashville, TN<br />

Background: The PI3K/AKT/mTOR pathway has been reported<br />

altered in up to 40% of hormone receptor-positive (HR+) breast<br />

cancers (BC). Furthermore, pathway activation has been implicated in<br />

resistance to endocrine therapy, including aromatase inhibitors (AI).<br />

BKM120 is an oral inhibitor of all 4 isoforms of class I PI3K (α, β,<br />

γ, δ). In combination with fulvestrant, BKM120 has demonstrated<br />

pro-apoptotic effects in vitro, and achieved near-complete tumor<br />

regression in HR+ PIK3CA-mutated estrogen-independent xenografts.<br />

Preliminary clinical activity was observed with BKM120 in patients<br />

with BC, both as a single agent and in combination with letrozole<br />

(Mayer, et al. ASCO 2012:#510) or trastuzumab (Saura, et al. SABCS<br />

2011:#PD09-03).<br />

Study design: This is a Phase III randomized, double-blind, placebocontrolled<br />

trial of BKM120 or placebo in combination with fulvestrant<br />

in postmenopausal women with HR+/HER2– locally advanced or<br />

metastatic BC that progressed on or after AI treatment (BELLE-2;<br />

CBKM120F2302; NCT01610284). After a 14-day fulvestrant runin<br />

phase to allow determination of tumor PI3K pathway activation<br />

status, patients are randomized (1:1) to receive continuous BKM120<br />

(100 mg/d) or placebo, and fulvestrant 500 mg at C1D15, and D1<br />

of all cycles thereafter; 1 cycle=28 days. Randomization is stratified<br />

according to PI3K pathway activation (PIK3CA mut and/or PTEN<br />

mut and/or loss of PTEN protein expression by IHC), and visceral<br />

disease status. Study treatment is given until disease progression or<br />

until study discontinuation for any reason.<br />

Eligibility criteria include: postmenopausal women with HR+/HER2–<br />

locally advanced or metastatic BC refractory to AI (progression while<br />

on AI, or within 4 wks [metastatic] or 12 mos [adjuvant] of last AI<br />

dose); no more than 1 previous line of chemotherapy for advanced<br />

disease; and available archival tumor tissue for analysis of PI3Krelated<br />

biomarkers.<br />

Co-primary endpoints: PFS in the full and PI3K pathway-activated<br />

populations based on local investigator assessment (RECIST 1.1).<br />

Key secondary endpoints: overall survival (OS) in both full and<br />

PI3K pathway-activated populations. Other secondary endpoints:<br />

PFS and OS (PI3K non-activated/unknown population), overall<br />

response rate and clinical benefit rate (both in full, PI3K-activated,<br />

PI3K non-activated/unknown populations), safety (CTCAE 4.03),<br />

pharmacokinetics of BKM120 and fulvestrant, and patient-reported<br />

quality of life (EORTC QLQ-C30 and QLQ-BR23).<br />

Statistical methods: PFS and OS will be estimated using the Kaplan–<br />

Meier method for both full and PI3K pathway activated populations.<br />

Both primary and key secondary endpoints will be tested using a<br />

stratified log-rank test at levels α governed by a graphical gatekeeping<br />

procedure. A stratified Cox regression will be used to estimate<br />

the hazard ratio, along with two-sided 95% confidence interval.<br />

Target accrual: Estimated enrollment for BELLE-2 is 842 patients.<br />

Recruitment in this Global study is ongoing with planned enrollment<br />

of patients in North America, South America, Europe, Asia and Africa.<br />

OT2-3-10<br />

Phase II study of panitumumab, nab-paclitaxel, and carboplatin<br />

for patients with primary inflammatory breast cancer (IBC)<br />

without HER2 overexpression<br />

Willey JS, Alvarez RH, Valero V, Lara JM, Parker CA, Hortobagyi<br />

GN, Ueno NT. Morgan Welch Inflammatory <strong>Breast</strong> <strong>Cancer</strong> Research<br />

Program and Clinic, The University of Texas MD Anderson <strong>Cancer</strong><br />

Center, Houston, TX; University of TX, MD Anderson <strong>Cancer</strong> Center,<br />

Houston, TX<br />

Background: Inflammatory breast cancer (IBC) is the most<br />

aggressive form of primary breast cancer. The outcome for patients<br />

with IBC is bleak despite multimodality treatment approaches.<br />

10-year disease-free survival rates after combined anthracycline<br />

and taxane-containing chemotherapy, surgery, and radiation are<br />

only 20%-25%. Our recent study found EGFR overexpression, a<br />

predictive factor of poor outcome, in 12 of 40 (30%) patients with<br />

IBC. Panitumumab has shown activity against EGFR overexpressing<br />

breast cancer xenograft model.<br />

Trial design: This is a single center, open-label, phase II study to<br />

evaluate the safety and efficacy of panitumumab in combination<br />

with preoperative chemotherapy. The treatment regimen consists<br />

panitumumab 2.5 mg/Kg given intravenously alone for the first week,<br />

followed by weekly panitumumab, nab-paclitaxel (100mg/m 2 ) and<br />

carboplatin (2 AUC) (PNC) for 12 weeks. Patients then will receive<br />

5-FU, epirubicin, and cyclophosphamide (FEC) every 3 weeks for 4<br />

cycles prior to surgery.<br />

Eligibility criteria: 1) Histological confirmation of breast carcinoma<br />

with pathologic evidence of dermal lymphatic invasion and clinical<br />

www.aacrjournals.org 575s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

diagnosis of IBC, including diffuse erythema, heat, ridging, and peau<br />

d’orange; 2) Normal HER2 expression; 3) No prior therapies for<br />

IBC; 4) Adequate hematologic, cardiac, renal and hepatic functions.<br />

Specific aims: 1) Primary objective is to determine the pathologic<br />

complete response (pCR) rate in patients with primary IBC without<br />

HER2 overexpression; 2) Secondary objectives are to determine the<br />

disease-free survival (DFS), overall survival (OS), the safety and<br />

tolerability of PNC regimens and the correlates of pathologic response<br />

rate and EGFR expression level.<br />

Statistical methods: 1) Previous studies have shown that this IBC<br />

patient population achieved a 13% pCR rate on the standard of care.<br />

We assume a beta (0.26, 1.74) prior distribution for the pCR rate.<br />

This prior distribution has a mean of 13% and a standard deviation<br />

of 19%. 2) We will stop the trial early if P (pCR rate >/= 13%) is<br />

< 0.01. If we determine that there is less than a 1% chance that the<br />

pCR rate is 13% or more we will consider stopping the trial. 3) Once<br />

we have completed the study we will estimate the pCR rate with a<br />

90% credible interval. If we have pCR in 4 of the 40 patients (10%),<br />

then our 90% credible interval for the pCR rate will be 4.0-19.6%.<br />

If we have pCR in 8 of the 40 patients (20%), then our 90% credible<br />

interval for the pCR rate will be 10.6-30.4%. We will also report the<br />

posterior probability that the pCR rate is 13% or more. For example,<br />

if we have pCR in 8 of the 40 patients (20%), then the probability<br />

that the pCR rate is 13% or more is 0.869.<br />

Present accrual and target accrual: To date, 13 patients have been<br />

enrolled. Target accrual is 40 patients.<br />

OT2-3-11<br />

Tivozanib in combination with paclitaxel vs placebo with<br />

paclitaxel in patients with locally advanced or metastatic triplenegative<br />

breast cancer<br />

Mayer EL, Miller K, O’Shaughnessy J, Dickler M, Vogel C,<br />

Leyland-Jones B, Steelman L, Robinson M, Kuriyama N, Agarwal<br />

S. <strong>Breast</strong> Oncology Center, Dana-Farber <strong>Cancer</strong> Institute, Boston,<br />

MA; Indiana University Melvin and Bren Simon <strong>Cancer</strong> Center,<br />

Indianapolis, IN; Baylor-Charles A. Sammons <strong>Cancer</strong> Center, Texas<br />

Oncology and US Oncology, Dallas, TX; Memorial Sloan-Kettering<br />

<strong>Cancer</strong> Center, New York, NY; Sylvester Comprehensive <strong>Cancer</strong><br />

Center, Miami, FL; <strong>San</strong>ford Research/USD, Sioux Falls, SD; AVEO<br />

Oncology, Cambridge, MA<br />

Background: Triple-negative breast cancer (TNBC) is an aggressive<br />

cancer with inferior survival outcomes. Although weekly paclitaxel<br />

(WP) is effective in the treatment (tx) of metastatic breast cancer<br />

(MBC), optimization of therapies for patients (pts) with TNBC is<br />

essential. Angiogenesis is a hallmark of advanced cancer, with subset<br />

analyses suggesting activity of angiogenesis inhibitors in TNBC.<br />

Tivozanib (TIVO) is a potent and selective inhibitor of vascular<br />

endothelial growth factor receptors (VEGFR) 1, 2, and 3 with a<br />

promising role in metastatic renal cell carcinoma, and established<br />

safety in Phase I combination with WP in MBC.<br />

Purpose: This Phase II trial will assess the efficacy and safety of<br />

TIVO + WP in the first-line setting for pts with advanced or metastatic<br />

TNBC and evaluate the performance of candidate angiogenesis<br />

biomarkers.<br />

Objectives: The primary objective of this study is to compare<br />

progression-free survival (PFS) of pts treated with TIVO + WP vs<br />

pts treated with placebo (PB) + WP. Secondary objectives include<br />

objective response rate (ORR), overall survival (OS), safety and<br />

tolerability, quality of life, and correlative candidate biomarker<br />

endpoints. The pharmacokinetics of TIVO + WP also will be<br />

characterized.<br />

Study Design and Methods: This multicenter, randomized,<br />

PB-controlled, two-arm study will enroll pts with metastatic or<br />

unresectable TNBC (evaluable per RECIST) and no prior systemic<br />

therapy. Pts must have confirmed available archival tumor tissue.<br />

Pts will be stratified by ECOG performance score and number of<br />

metastatic sites, then randomized to receive either oral TIVO 1.5 mg<br />

once daily for 3 weeks (wks) on/1 wk off and intravenous WP 90<br />

mg/m 2 for 3 wks on/1 wk off, or PB + WP. One cycle will be 4 wks;<br />

tx will continue until disease progression or unacceptable toxicity.<br />

Archival tumor tissue and blood samples will be evaluated for<br />

response biomarkers, including a hypoxia sensitivity gene signature,<br />

a myeloid resistance gene signature, and angiogenic ligands. All pts<br />

will be followed for survival until death. Adverse events will be<br />

monitored throughout the study. Pharmacokinetic samples will be<br />

collected during cycles 1 and 2. PAM-50–defined intrinsic molecular<br />

subtype populations also will be evaluated retrospectively.<br />

Recruitment of 130 patients is planned, with an interim analysis after<br />

80 pts to measure ORR (130 pts with a total of 82 investigator-assessed<br />

PFS events provides 80% power to detect statistically significant<br />

PFS differences between tx arms). Endpoint analyses will use the<br />

intent-to-treat population. The primary efficacy analysis will use<br />

investigator assessments of response and a two-sided 95% confidence<br />

interval for the hazard ratio produced using Cox proportional hazards<br />

regression models. OS will be compared using the log-rank test.<br />

Analyses of candidate biomarkers and determination of an optimal<br />

predictive cutoff for response also are planned. Trial enrollment will<br />

commence in fall 2012.<br />

Conclusion: This study will determine whether TIVO, a selective<br />

and potent VEGFR inhibitor, combined with WP improves clinical<br />

outcomes in pts with TNBC, and whether clinical activity is associated<br />

with candidate angiogenesis biomarkers.<br />

OT3-1-01<br />

A pilot study of single-dose ipilimumab and/or cryoablation in<br />

women with early-stage breast cancer scheduled for mastectomy<br />

Diab A, McArthur HL, Solomon S, Comstock C, Maybody M, Sacchini<br />

V, Durack J, Gucalp A, Yuan J, Patil S, Thorne A, Sung J, Kotin A,<br />

Morris E, Brogi E, Morrow M, Wolchok J, Allison J, Hudis C, Norton<br />

L. Memorial Sloan-Kettering <strong>Cancer</strong> Center, New York, NY<br />

Background: With cryoablation, probes are inserted into tumors to<br />

induce crystallization, osmotic changes, vascular stasis, and ultimately<br />

tumor necrosis. The resultant release of tumor antigens could activate<br />

a tumor-specific immune response through antigen presentation by<br />

dendritic cells (DC) to T-cells. Cryoablation is an effective treatment<br />

modality in many cancers (Habash 2007), and feasibility studies have<br />

shown that thermal ablation can be performed safely and effectively<br />

in small breast cancers (Noguchi 2007).<br />

To amplify immune response, we use ipilimumab, a fully human<br />

monoclonal antibody that blocks cytotoxic T lymphocyte antigen-4<br />

(CTLA4), a co-inhibitory molecule present on the surface of<br />

activated T cells. Durable and clinically significant responses to<br />

ipilimumab have been reported in prostate, bladder, and renal cell<br />

cancers. Ipilimumab has been most extensively studied in malignant<br />

melanoma, with 10-20% response rates and significant survival<br />

benefits reported (Wolchok 2010, Hodi 2010).<br />

Cryoablation-mediated tumor destruction exposes DCs to sufficient<br />

quantities of tumor antigens and inflammatory cytokines to induce<br />

DC maturation and activation. Furthermore, in preclinical murine<br />

models, the combination of cryoablation with CTLA4 blockade<br />

successfully mediates rejection of metastatic prostate cancer lesions<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

576s<br />

<strong>Cancer</strong> Research


December 4-8, 2012<br />

<strong>Abstract</strong>s: Poster Session OT3<br />

and prevents growth of secondary tumors (Waitz 2012). Combined<br />

ablative therapy with CTLA4 blockade is appealing due to the relative<br />

safety of percutaneous ablation in early stage breast cancer and the<br />

efficacy and tolerability of ipilimumab administration in numerous<br />

settings. We therefore hypothesize that this strategy could confer<br />

long-term immunity, and ultimately cure, for women with early<br />

stage breast cancer.<br />

In this pilot study, the safety and tolerability of pre-operative<br />

cryoablation alone, single dose ipilimumab (10mg/kg) alone, or the<br />

combination is being evaluated in women with early stage breast<br />

cancer.<br />

Design: Three sequential test groups of 6 patients are being enrolled<br />

(total N=18). The study groups are: pre-operative cryoablation alone<br />

(A), pre-operative single dose ipilimumab alone (B), and pre-operative<br />

single-dose ipilimumab followed by cryoablation (C). Groups A and<br />

B must meet the safety primary endpoint before enrollment to study<br />

arm C is begun. The timing of all study interventions is defined by<br />

the pre-determined, non-study surgery date.<br />

Primary aim: To determine the safety and tolerability of preoperative<br />

ipilimumab and/or cryoablation in patients with resectable<br />

breast cancer.<br />

Secondary aims: To explore and characterize pre- and postintervention<br />

radiographic and immunological (peripheral blood and<br />

tumor tissue) correlates.<br />

Statistics: If at least 5 of the 6 patients in each group proceed with<br />

surgery without delay, the regimen will be considered safe/tolerable<br />

for further study. Secondary aims are exploratory.<br />

Eligibility: Eligible women are age ≥18y with early-stage, operable,<br />

≥1.5 cm invasive breast cancer, no history of autoimmune disease<br />

and planning mastectomy.<br />

Study Status: As of June 12, 2012, Study Arm A is filled: 4 women<br />

have undergone cryoablation and mastectomy and 2 have undergone<br />

cryoablation and are awaiting surgery.<br />

OT3-1-02<br />

Phase II randomized study of combination immunotherapy with<br />

or without Polysaccharide Krestin (PSK®) concurrently with a<br />

HER2 ICD peptide-based vaccine and trastuzumab in patients<br />

with stage IV breast cancer<br />

Childs JS, Higgins DM, Parker S, Reichow J, Lu H, Standish L,<br />

Disis ML, Salazar LG. University of Washington, Seattle, WA; Bastyr<br />

University, Kenmore, WA<br />

Background: Endogenous immunity in patients with HER2+<br />

metastatic breast cancer (MBC) is likely dampened by an immunesuppressive<br />

tumor microenvironment and not sufficient to control<br />

tumor growth. Thus, most patients have disease relapse after achieving<br />

complete remission with standard therapies. Immunomodulation<br />

directed at enhanced stimulation of tumor specific immunity could<br />

result in immunologic eradication of residual HER2+ tumor cells and<br />

prevent BC relapse. We have shown PSK to be a potent TLR-2 agonist<br />

that stimulates both innate and adaptive immunity in a BC mouse<br />

model. Additionally, we have shown combination immunotherapy<br />

with HER2 peptide vaccines and trastuzumab (TRAZ) to be safe and<br />

able to elicit HER2 specific Th1 immunity and epitope spreading (ES)<br />

which has been associated with survival in vaccinated patients. Lastly,<br />

decreased serum TGF-β elicited by HER2 vaccination correlates<br />

with Th1 ES and may serve as a biomarker to predict cancer vaccine<br />

efficacy. We hypothesize that PSK, when given with TRAZ can<br />

augment vaccine induced HER2 specific TH1 immunity and prevent<br />

disease relapse in patients with optimally treated HER2+ MBC.<br />

Trial design: Phase II randomized two-arm clinical trial. Patients will<br />

be enrolled and randomly assigned in equal numbers to 1 of 2 arms<br />

(15 patients/arm) as follows: Arm 1:HER2 ICD vaccine, TRAZ and<br />

placebo or Arm 2:HER2 ICD vaccine, TRAZ and PSK. All patients<br />

will receive 3 monthly HER2 ICD vaccines plus TRAZ and 4 months<br />

of concomitant PSK or placebo. Serial blood draws for immunologic<br />

monitoring will be done.<br />

Eligibility criteria: Patients with Stage IV HER2+ BC who have been<br />

treated with definitive therapy and are: (1) without evidence of disease<br />

or have stable-bone only disease, (2) receiving TRAZ monotherapy,<br />

and (3) without clinically significant autoimmune disease. Patients<br />

must have normal LVEF per MUGA scan or echocardiogram.<br />

Aims: (1) Evaluate safety of PSK when given with a HER2 vaccine<br />

and TRAZ (2) Evaluate the effect of PSK on serum TGF-β levels when<br />

given with a HER2 vaccine and TRAZ and (3) Evaluate the effect<br />

of PSK on intermolecular ES when given with a HER2 vaccine and<br />

TRAZ. A secondary objective is to evaluate progression free survival<br />

(PFS) and overall survival (OS).<br />

Statistical methods: (1) Toxicity will be determined by clinical and<br />

chemical parameters and grading will be done per CTEP CTCAE<br />

4.0.; (2) Evaluation of TGF-β levels, pre and post-PSK treatment will<br />

be assessed with linear regression models; and analysis of multiple<br />

post-baseline measurements will be performed using generalized<br />

estimating equations; (3) A positive antigen-specific immune response<br />

will be defined as a precursor frequency >1:20,000 antigen-specific<br />

peripheral blood mononuclear cells. Differences in the levels of HER2<br />

immunity will be evaluated between arms using a two-tailed T test.<br />

The degree of ES in each arm will be evaluated with generalized linear<br />

modeling; (4) Large differences in PFS and OS observed between<br />

groups will be noted and described.<br />

Target accrual: 30 patients.<br />

OT3-2-01<br />

Influence of strength and endurance training on selected physical<br />

and psychological parameters and on immune system, metabolism<br />

and circulating tumor cells during adjuvant chemotherapy<br />

Mundhenke C, Weisser B, Keller L, <strong>San</strong>ders L, Summa B, Duerkop<br />

J, Schmidt T, Jonat W. University of Kiel, SH, Germany; Institute<br />

of Sport Science, University of Kiel, SH, Germany; Comprehensive<br />

<strong>Cancer</strong> Center North, Kiel, SH, Germany<br />

Background: Reduced quality of life, limited performance and<br />

fatigue are commonly observed side effects of breast cancer systemic<br />

treatment. In previous studies sport therapy could improve these<br />

symptoms. Sport has also been reported to positively impact the<br />

clinical course of the disease. So far it has remained unclear if these<br />

influences on clinical outcome are caused by alterations in the immune<br />

system or in tumor biology or in glucose or fat metabolism.<br />

The aim of this study is to analyse the implications of individual<br />

interventions in exercise therapy on endurance, muscle strength,<br />

fatigue and quality of life, alterations in the immune system or in<br />

tumor biology or in glucose or fat metabolism for breast cancer<br />

patients during chemo therapy.<br />

Method: The trial is prospectively randomised and controlled and will<br />

involve 60 breast cancer patients receiving adjuvant chemotherapy.<br />

20 patients each are randomized into a strength training group, an<br />

endurance training group or a control group (with standard care). At<br />

the beginning and after 12 weeks the following tests are undertaken:<br />

isometric maximum strength test, a PWC 150 test to determine<br />

the endurance of patients. The standardised questionnaire EORTC<br />

QLQ C30 including module BR23 to measure quality of life and the<br />

questionnaire MFI-20 to determine symptoms of fatigue are filled<br />

www.aacrjournals.org 577s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

in. Questionnair D2 measures ability to concentrate. Immunologic<br />

parameters as CD4, CD8 and T-REG-cells (FACS-Analysis), the<br />

number of the circulating tumour cells (measured by density gradient<br />

centrifugation) and also the concentration of glucose, insulin, leptin<br />

and adiponectin are being measured at study entry and after 12 weeks.<br />

Momentarily patients are being screened, recruited and randomised<br />

into the trial. The final evaluation of the results is planned II/ 2013.<br />

Discussion: The purpose of this study is to show if additional value<br />

can be added by individualized strength- or endurance training.<br />

Stabilisation or improvement of maximum strength and endurance<br />

during chemotherapy, as well as increase of quality of life and<br />

reduction of symptoms of fatigue would be a major improvement.<br />

Knowledge about effects of physical activity on circulating tumour<br />

cells as well as on immunologic alterations and on glucose and<br />

fat metabolism under chemotherapy could lead to a more specific<br />

counseling of patients and an individualized training program during<br />

chemotherapy.<br />

OT3-2-02<br />

The PREDICT Study ( Prospective, Randomized Early Detection<br />

and Intervention after <strong>Breast</strong> <strong>Cancer</strong>-Treatment, for women at<br />

risk of lymphedema)<br />

Taghian AG, Skolny MN, O’Toole J, Miller CL, Jammallo LS, Specht<br />

MC. Massachusetts General Hospital, Boston, MA<br />

Introduction: It is well-documented that lymphedema is one of the<br />

most feared long-term side effects of breast cancer (BC) treatment.<br />

However, to date, a standardized approach for the quantification and<br />

treatment of breast cancer-related lymphedema (BCRL) has yet to<br />

be established.<br />

Aims: We propose a screening and intervention trial that will assess<br />

the efficacy of early detection and intervention for BCRL. Intervention<br />

comprises the use of compression garments for mild lymphedema<br />

and compression garments +/- nighttime bandaging for moderate<br />

lymphedema. Other factors to be evaluated include: symptom clusters,<br />

treatment adherence, fear avoidance behavior, quality of life (QOL),<br />

upper extremity function, and risk factors for BCRL.<br />

Eligibility Criteria: Women 18 years + with a confirmed BC diagnosis,<br />

no history of BC, no known metastatic or locally advanced disease,<br />

no history of primary lymphedema, sentinel lymph node biopsy or<br />

axillary lymph node dissection as part of definitive breast surgery.<br />

Study Design: A two-stage study which includes a Screening and<br />

an Intervention trial. The screening arm will evaluate arm volume<br />

change during and after BC treatment with target accrual of 8000.<br />

Patients will undergo measurements via perometry and complete<br />

the MGH Lymphedema Evaluation Following Treatment for<br />

<strong>Breast</strong> <strong>Cancer</strong> (LEFT-BC) Survey at each screening appointment<br />

to evaluate changes in functionality, upper extremity utilization<br />

(fear associated avoidance), and QOL. Screening visits will occur<br />

pre- and post - operatively, at the conclusion of chemotherapy and<br />

radiation therapy and every 3-7 months thereafter. Patients will<br />

become eligible for enrollment into the intervention trial if, during<br />

the course of screening, they develop a relative arm volume change<br />

(RVC) of ≥ 5% which persists at a verification measurement within<br />

4-8 weeks. Eligible subjects are enrolled into one of two groups based<br />

on verification RVC: Group I – Mild Lymphedema (5-10% RVC) or<br />

Group II – Moderate Lymphedema (11-20% RVC). Subjects are then<br />

randomized within each group. Group I subjects are randomized to<br />

one of two arms: I-A – Observation, I-B – Compression, and Group<br />

II subjects are randomized to one of two arms: II-A – Compression,<br />

II-B – Compression + Night Compression Bandaging. Target accrual<br />

for the intervention trial is 336 subjects (Group I: 208, Group II: 128).<br />

Clinical Relevance: The results of this study will yield level I evidence<br />

on the effectiveness of early detection and intervention for BCRL.<br />

Findings may shape clinical practice in diagnosis and treatment, as<br />

well as provide insight regarding the risk factors, symptoms, upper<br />

extremity function, and quality of life (QOL) associated with BCRL.<br />

*Funding by award #s R01CA139118 &3P5OCA089393, AGT<br />

OT3-3-01<br />

Eniluracil + 5-fluorouracil + leucovorin (EFL) vs. capecitabine<br />

phase 2 trial for metastatic breast cancer<br />

Rivera E, Chang JC, Semiglazov V, Gorbunova V, Manikhas A,<br />

Krasnozhon D, Kirby G, Spector T. Banner MD Anderson <strong>Cancer</strong><br />

Center, Gilbert, AZ; The Methodist Hospital Research Institute,<br />

Houston, TX; Road Clinical Hospital of the Russian Railways, St.<br />

Petersburg, Russian Federation; Russian Oncological Research<br />

Center n.s.Blokhin RAMS, Moscow, Russian Federation; City Clinical<br />

Oncology Center, St. Petersburg, Russian Federation; Institution<br />

Leningrad Regional Oncology Center, Leningrad Region, Russian<br />

Federation; Adherex Technologies, Inc., Research Triangle Park, NC<br />

Based on a modified dosing protocol designed to optimize efficacy,<br />

an open-label EFL vs. capecitabine (4:3 randomization) Phase 2<br />

trial for metastatic breast cancer is in progress. Eniluracil inactivates<br />

dihydropyrimidine dehydrogenase, thereby preventing the formation<br />

of α-fluoro-β-alanine, and conferring 100% oral bioavailability<br />

and a 5 hr half-life on 5-fluorouracil (5-FU). Study drugs are<br />

administered orally for 1 st - or 2 nd -line treatment for metastatic disease<br />

in patients previously treated with an anthracycline and a taxane.<br />

Arm 1: eniluracil (40 mg) taken 11-16 hr before 5-FU (30 mg/m 2 );<br />

leucovorin (30 mg) taken with 5-FU and the next day. The regimen<br />

is administered once/week for 3 weeks/4 weeks. Arm 2: capecitabine<br />

(1000 mg/m 2 ) taken bid for 14 days/21days. Arm 2 patients with<br />

disease progression could crossover to take EFL in Arm X. Two sites<br />

in the USA and 19 in Russia are enrolling. Currently, 115 patients<br />

(21% are 1 st -line, 70% had previous 5-FU treatment) are enrolled<br />

and 83 have had tumor assessments. EFL was well tolerated with no<br />

unexpected toxicities. As of May 2012, there were 11, 7, & 1 partial<br />

responses in Arms 1, 2, & X, respectively. The primary endpoint,<br />

progression-free survival, will be determined approximately 7.5<br />

months after the trial is enrolled with 140 evaluable patients.<br />

OT3-3-02<br />

ADAPT - Adjuvant Dynamic marker-Adjusted Personalized<br />

Therapy trial optimizing risk assessment and therapy response<br />

prediction in early breast cancer<br />

Gluz O, Hofmann D, Kates RE, Harbeck N, Nitz U. West German<br />

Study Group, Moenchengladbach, NRW, Germany; Ev. Bethesda<br />

Hospital, Moenchengladbach, NRW, Germany; University Clinic<br />

Großhadern, Munich, Bavaria, Germany<br />

Background: Appropriate indication for chemotherapy (CTx) in<br />

patients with hormone receptor (HR) positive breast cancer (BC),<br />

prediction of CTx response, and addition of novel targeted therapies<br />

are the most important questions in adjuvant therapy of early BC. Few<br />

well evaluated prognostic tests identify patients (pts) with a low-risk<br />

profile resulting in a low enough CTx benefit to justify omission of<br />

CTx. Several clinical trials with results still pending have already<br />

addressed this question such as TAILORx, MINDACT, NNBC-3,<br />

WSG-planB.<br />

However, proliferation seems to be a key part of all prognostic<br />

genomic signatures. Moreover, dynamic changes of proliferation<br />

(as a result of endocrine, cytotoxic and targeted therapy) during<br />

therapy have been shown to be most important for outcome prediction<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

578s<br />

<strong>Cancer</strong> Research


December 4-8, 2012<br />

<strong>Abstract</strong>s: Poster Session OT3<br />

in patients with pathological complete response to neoadjuvant<br />

chemotherapy in distinct BC subtypes (luminal B, TNBC, HER2+).<br />

Trial design: ADAPT is one of the first new generation adjuvant<br />

trials dealing with individualization of adjuvant decision-making<br />

in early BC. The WSG-ADAPT trial combines static assessment of<br />

prognosis of patients by a genomic signature (Recurrence Score in<br />

HR+ disease) and conventional prognostic markers (nodal status)<br />

with dynamic measurement of proliferation/apoptosis changes during<br />

a short course of preoperative therapy. ADAPT will establish early<br />

predictive molecular surrogate markers for outcome by assessing<br />

response to a short 3-week induction treatment, using a baseline<br />

diagnostic core biopsy and a second biopsy after induction treatment.<br />

ADAPT consists of an umbrella trial and four different sub-protocols<br />

(HR+/HER2-, HR+/HER2+, HR-/HER2+ HR-/HER2-) and is set up<br />

as a prospective, multi-center, controlled, non-blinded, randomized<br />

phase II/III trial.<br />

Eligibility criteria: Pre-/postmenopausal women with histologically<br />

confirmed unilateral primary invasive early BC. Women identified<br />

to require chemo- or targeted (anti-HER2) therapy must have<br />

adequate laboratory values and organ function. Pts must have no<br />

contraindications for the planned treatment.<br />

Specific aims: Primary endpoints will include evaluation of the<br />

dynamic test for outcome prediction and prospective comparison<br />

of 5yr EFS in responders (intermediate risk (RS 12-25) and good<br />

response to short-term endocrine therapy in HR+/HER2- as well as<br />

pts with pCR in HER2+/triple negative (TN) disease) compared to<br />

low risk HR+/HER2- (RS≤11, N0-1) pts (control group). Secondary<br />

endpoints will include overall survival.<br />

Statistical methods: Assumption across sub-protocols is that adjuvant<br />

CTx can be spared in HR+/HER2- BC or pCR be achieved in<br />

HER2+/TN in expected 1120 (HR+/HER2-) or 170 (HER2+/TN) pts<br />

respectively within the main phase. Their outcome will be compared<br />

to the control group (expected n=640 HR+/HER2- pts: low risk (by<br />

RS) and thus no CTx). Assuming 94% 5yr survival in control group,<br />

one-sided test of non-inferiority at 95% CI will have 80% power for<br />

a survival non-inferiority margin corresponding to 3.2% (i.e. 90.8%<br />

survival).<br />

Present and target accrual: By June 2012, 11 of 12 initial sites have<br />

been initiated, and 7 pts recruited. Target accrual for run-in phase is<br />

400 pts (4000 for run-in and main phase).<br />

OT3-3-03<br />

Withdrawn by Author<br />

alopecia more than 24 months after their last chemotherapy infusion.<br />

Optimal information of patients about alopecia and persisting alopecia<br />

appears to be mandatory before treatment : 47% of patients undergo<br />

a psychological shock during hair loss. Morevover, BCIRG 001<br />

study (TAC versus FAC) led to the conclusion that docetaxel 75 mg/<br />

msq is responsible for persisting alopecia for 3% of patients. So, by<br />

extrapolating, in France, each year, docetaxel could induce a persisting<br />

alopecia in 300 patients. To confirm this figure, “ERALOP” study is<br />

in progress to evaluate precisely the incidence of persisting alopecia<br />

after docetaxel in early breast cancer patients and then theses data will<br />

be communicated to ANSM. It must be noticed that 8% of patients<br />

refused to be treated because of the risk of persistant alopecia (Trueb<br />

et al, 2010). That’s why it is a worldwide public health problem, with<br />

personal and societal implications.<br />

As alopecia is universal after FEC100 regimen, the cooling scalp has<br />

been supposed to be very effective during docetaxel 1 hour infusion<br />

and to allow a better hair regrowth but with a high risk in term of<br />

tolerance. Since 2012, a prospective clinical trial “ALOPREV” has<br />

begun in Western France to evaluate tolerance of cooling scalp,<br />

after 3-4 courses of FEC100 regimen and during 3-4 courses of<br />

docetaxel 100 mg/msq, for patients treated for early breast cancer.<br />

So, ALOPREV is designed as a safety trial. First, cooling scalp<br />

major toxicities (headache, neck pain, discomfort associated with<br />

cold, sinusitis) are evaluated during each docetaxel course by the<br />

patient. Then, auto-questionnary about hair loss and regrowth, nails<br />

disorders and others toxicities due to docetaxel and questionnary about<br />

quality of life (EORTC QLQ-C30 and BR-23 scales) is completed<br />

by the patients before, during and after treatment. Patients will<br />

be followed-up 12 months and final toxicities will be reported by<br />

physicians. According to our daily practice, it will demonstrate that<br />

60% of patients could tolerate two consecutive cooling scalps during<br />

docetaxel infusion, and with statistical considerations (alpha-beta<br />

risks and 10% of ineligibility). So 160 patients must be included<br />

during inclusion period (18 months). 10 public or private care centers<br />

have been opened (45 investigators). This trial implicates particularly<br />

nurses teams and all patients will sign an informed consent before<br />

FEC100 courses. ALOPREV started two months ago and 25 patients<br />

are already enrolled.<br />

In conclusion, cooling scalp is known to be safe, manageable and<br />

partially effective before hair loss. ALOPREV will evaluate safety<br />

and efficacy to avoid persisting alopecia after hair loss.<br />

OT3-3-04<br />

ALOPREV : first cooling scalp trial for prevention of persisting<br />

alopecia after docetaxel for early breast cancer patients.<br />

Bourgeois H, Soulié P, Lucas B, Mercier Blas A, Zannetti A, Delecroix<br />

V, L’haridon T, Blot E, Delaloge S, Grudé F. Clinique Victor Hugo,<br />

Le Mans, France; Observatoire dédié au <strong>Cancer</strong> Bretagne Pays de<br />

Loire, Angers, France; Institut de Cancérologie de l’Ouest, Angers<br />

Nantes, France; CHRU Morvan, Brest, France; CHP, Saint Grégoire,<br />

France; CHD, Cholet, France; Pôle Hospitalier Mutualiste, Saint<br />

Nazaire, France; CHD, La Roche sur Yon, France; Centre Hospitalier<br />

Bretagne Atlantique, Vannes, France; Clinique Océane, Centre Saint<br />

Yves, Vannes, France; Institut Gustave Roussy, Villejuif, France<br />

During SABCS 2009, Western France Observatory of <strong>Cancer</strong><br />

has presented “ALOPERS” results from more than one hundred<br />

patients with persisting alopecia or suboptimal hair regrowth after<br />

chemotherapy for early breast cancer. Docetaxel 75-100 mg/msq<br />

was concerned for majority of patients. 43% of patients experienced<br />

www.aacrjournals.org 579s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

OT3-3-05<br />

Neurotoxicity characterization phase II randomized study of nabpaclitaxel<br />

versus conventional paclitaxel as first-line therapy of<br />

metastatic HER2-negative breast cancer. An ONSOCUR Study<br />

Group.<br />

Ciruelos E, Bueno C, Cantos B, Carrión R, Echarri M, Enrech S,<br />

García Saenz JA, Guerra JA, Lara MA, Martínez N, Rodríguez-<br />

Antona C, Domínguez-González C, <strong>San</strong>z JL, Baquero J, Cortés-Funes<br />

H, Sepúlveda JM. Hospital 12 de Octubre, Madrid, Spain; Hospital<br />

Universitario Ramón y Cajal, Madrid, Spain; Hospital Universitario<br />

del Sureste, Arganda del Rey (Madrid), Spain; Hospital Universitario<br />

Clínico <strong>San</strong> Carlos, Madrid, Spain; Hospital Universitario Severo<br />

Ochoa, Leganés (Madrid), Spain; Hospital Universitario Puerta<br />

de Hierro Majadahonda, Majadahonda (Madrid), Spain; Hospital<br />

Universitario Infanta Cristina, Parla (Madrid), Spain; Hospital<br />

Universitario Infanta Leonor, Madrid, Spain; Hospital Universitario<br />

de Getafe, Getafe (Madrid), Spain; Hospital Universitario de<br />

Fuenlabrada, Fuenlabrada (Madrid), Spain; APICES, Madrid,<br />

Spain; Celgene, Madrid, Spain; Centro Nacional de Investigaciones<br />

Oncologicas, Madrid, Spain<br />

Background: Nab-paclitaxel (Nab-P) is a nanoparticle albumin-bound<br />

form of paclitaxel that is thought to exploit natural albumin pathways<br />

to enhance the selective uptake and accumulation of paclitaxel at<br />

the site of the tumour, thus reducing its diffusion to normal tissues.<br />

Nab-P has been approved for the treatment of metastatic breast cancer<br />

patients who have failed first-line treatment for metastatic disease and<br />

for whom standard, anthracycline-containing therapy is not indicated.<br />

Toxicity profile of Nab-P is well characterized with significantly less<br />

haematological toxicities compared with conventional paclitaxel<br />

(P). Nab-P derived grade III neuropathy is short-lasting and more<br />

reversible than conventional P-derived neuropathy, probably due to<br />

absence of Cremophor solvent, or due to P itself. However there is<br />

still a lack of clinical and physiological characterisation of Nab-P<br />

induced neuropathy. The Current used tools for early detection<br />

and continuous evaluation of neurotoxicity are not optimal. Most<br />

used toxicity scales are limited, as they do not provide a detailed<br />

information of the severity of the neuropathy, its impact on quality of<br />

life, or physiopathology mechanisms. In addition, an inter-individual<br />

variability exists in terms of neurotoxicity predisposition when<br />

taxanes are used; it seems to be related to polymorphic differences in<br />

genes implicated in transport and metabolism of these drugs.<br />

Specific aims: Primary objective is to characterize neurotoxicity<br />

according to Total Neuropathy Score (TNS) and electromyographic<br />

changes. Secondary endpoints are determine the rate of neuropathy<br />

chemo-induced, the predictive value of some genetic variants (SNPs)<br />

for the development of neuropathy, clinical activity, toxicity profile<br />

and safety of treatments and quality of life (EORTC QLQ-C30 and<br />

CIPN20).<br />

Trial design: Phase II open-label randomised clinical trial with four<br />

parallel arms: a) conventional paclitaxel 80 mg/m 2 on days 1, 8 and<br />

15; b) Nab-P 100 mg/m 2 on days 1, 8 and 15; c) Nab-P 150 mg/m 2<br />

on days 1, 8 and 15; d) Nab-P 150 mg/m 2 on days 1 and 15. Study<br />

treatments will be administered in a 28-day cycle until progression<br />

disease, intolerable toxicity or investigator’s decision.<br />

Eligibility criteria: Women >18 years, with cytological or<br />

histological confirmation of HER2-negative breast cancer, metastatic<br />

disease, not prior chemotherapy for advanced disease, no prior grade<br />

> 1 neuropathy from any cause, measurable or evaluable disease,<br />

ECOG >2 and adequate bone marrow, renal and hepatic functions.<br />

No Relevant comorbidities.<br />

Statistical methods: This is an exploratory safety study to better<br />

characterize neurotoxicity in patients treated with Nab-P compared<br />

toP. Therefore no formal sample size or power calculations will be<br />

performed. We expect to recruit 60 patients, 15 in each treatment arm,<br />

in 18 months. Recruitment will start on October 2012.<br />

OT3-3-06<br />

NeoEribulin: A Phase II, non-randomized, open-label, single-arm,<br />

multicenter, exploratory pharmacogenomic study of single agent<br />

eribulin as neoadjuvant treatment for operable Stage I-II HER2<br />

non-overexpressing breast cancer.<br />

Prat A, Llombart A, de la Peña L, Di Cosimo S, Ortega V, Rubio I,<br />

Muñoz E, Harbeck N, Cortés J. Vall d’Hebron University Hospital,<br />

Barcelona, Spain; Hospital Arnau de Vilanova, Valencia, Spain;<br />

SOLTI <strong>Breast</strong> <strong>Cancer</strong> Research Group, Barcelona, Spain; Istituto<br />

Nazionale dei Tumori, Milan, Italy; Brustzentrum am Klinikum der<br />

Universität München, Munich, Germany<br />

Background: The neoadjuvant setting provides a unique opportunity<br />

to identify predictive biomarkers for novel therapeutic molecules.<br />

Eribulin, a novel anti-microtubule agent, has recently been approved<br />

for the treatment of metastatic breast cancer and is currently under<br />

investigation in earlier stages of the disease.<br />

Trial design: This is a Phase II, non-randomized, open-label, singlearm<br />

exploratory pharmacogenomic study of eribulin as monotherapy<br />

that seeks to identify predictive biomarkers of response to this agent.<br />

Eribulin will be administered at 1.23 mg/m 2 (equivalent to 1.4 mg/m 2<br />

of eribulin mesylate) intravenously on Days 1 and 8 of every 21-day<br />

cycle, for 4 cycles. <strong>Breast</strong> surgery will be carried out 3-5 weeks after<br />

completion of study treatment.<br />

Eligibility criteria: Female adults with operable primary Stage I-II<br />

HER2 non-overexpressing breast cancer.<br />

Specific aims: The primary objective is to evaluate the correlation<br />

between pre-treatment mRNA expression from breast tumors and<br />

pathological complete response in the breast (pCR B ) at the time of<br />

surgery. Secondary objectives aim to assess the rate of pCR B and pCR<br />

in the breast and axilla, the clinical and radiological overall response<br />

rates, and the biological activity of the treatment; also, to estimate<br />

the sensitivity and specificity of gene expression to predict clinical<br />

response, to determine the correlation of pCR B with intrinsic breast<br />

cancer subtypes, and to evaluate the safety and tolerability of the<br />

regimen. Additional molecular characterizations by gene expression<br />

and exome or genome sequencing will be performed in order to further<br />

identify biomarkers of response and/or safety.<br />

Statistical methods: This is an exploratory study and sample size is<br />

not based on statistical power. Assuming a pCR in the breast of 15%,<br />

a sample size of 200 patients is estimated to provide a 90% probability<br />

of detecting a gene signature whose expression is associated with a<br />

two-fold increase in odds of achieving a pCR in the breast, assuming<br />

5% of patients would be lost to follow-up and 5% would have<br />

insufficient quality or quantity of mRNA. The rate of pCR in breast<br />

is calculated assuming a pCR in luminal A tumors of 0-2%, in luminal<br />

B tumors of 6-10% and in triple-negative of approximately 20-30%.<br />

Biomarker analyses: Baseline, 21-day treated, and post-treatment<br />

(surgical) primary breast tumor tissue samples will be obtained from<br />

each patient. The expression of 542 genes involved in breast cancer<br />

will be explored with the NanoString technology using formalin–<br />

fixed, paraffin–embedded tissue. The expression of single genes and<br />

the score of each signature in the pre-treatment and 21-day treated<br />

samples will be correlated with the clinical/pathologic outcome using<br />

a variety of statistical approaches.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

580s<br />

<strong>Cancer</strong> Research


December 4-8, 2012<br />

<strong>Abstract</strong>s: Poster Session OT3<br />

Target accrual: 200 patients, including a minimum of 100 subjects<br />

with triple-negative breast cancer, in 32 sites across Spain, Portugal,<br />

France, and Germany. The trial is funded by Eisai and registered with<br />

ClinicalTrials.gov. Patient enrollment began in July 2012.<br />

OT3-3-07<br />

Neoadjuvant bevacizumab with weekly nanoparticle albumin<br />

bound (nab)-paclitaxel plus carboplatin followed by doxorubicin<br />

plus cyclophosphamide (AC) for triple negative breast cancer.<br />

Snider JN, Allen JW, Young R, Schwartzberg L, Javed Y, Jahanzeb<br />

M, Sachdev JC. University of Tennessee, Memphis, TN; The West<br />

Clinic, Memphis, TN; The Center for <strong>Cancer</strong> and Blood Disorders,<br />

Fort Worth, TX<br />

Background:<br />

Triple negative breast cancer (TNBC) predominantly clusters with<br />

“basal like” subtype on genomic profiling. Over-expression of<br />

Secreted Protein Acidic and Rich in Cysteine (SPARC) has been<br />

observed in basal like breast cancer (Charaffe-Jaufrett et al. Oncogene<br />

2006; 2273-84). Endothelial transcytosis of nab- paclitaxel occurs<br />

via albumin- gp60 receptor-caveolin 1 interaction. SPARC entraps<br />

the albumin resulting in higher intratumoral accumulation, which<br />

may explain the increased efficacy of nab-paclitaxel (Desai et al.<br />

Translational Oncology 2009; 59-63). Exploiting this mechanism &<br />

the dysfunctional BRCA mediated DNA repair in basal tumors, we<br />

hypothesize that nab-paclitaxel + the DNA damaging drug carboplatin<br />

would produce high response rates in TNBC. Adding bevacizumab<br />

may enhance efficacy by blocking angiogenesis. In TNBC, pathologic<br />

complete remission (pCR) to neo-adjuvant therapy correlates with<br />

better disease-free survival (DFS). We hypothesize that high pCR<br />

rates can be achieved for patients with TNBC with this combination<br />

translating to an improved DFS than seen historically.<br />

Methods: Patients with palpable & operable TNBC ≥ 2 cm are eligible<br />

for this single stage phase II trial. pCR (defined as the absence of<br />

invasive tumor cells) in the breast is the primary end point, while<br />

pCR in the breast + axillary nodes & pCR + near pCR (residual<br />

tumor < 5mm) in the breast are secondary end points. 57 evaluable<br />

patients are needed, assuming a pCR rate of 25% vs. 40% for the null<br />

& alternate hypotheses respectively. 35 patients have been accrued<br />

to date. Patients receive carboplatin AUC 6 day 1 & nab-paclitaxel<br />

100mg/m 2 days 1, 8 & 15 of a 28 day cycle for 4 cycles and then dose<br />

dense AC for 4 cycles. Bevacizumab is given at 10mg/kg Q 2 weeks<br />

with chemotherapy for the first 6 cycles. Surgery & radiation are per<br />

institutional standards. Bevacizumab is continued postoperatively to<br />

complete 1 year of treatment. A core biopsy for collection of fresh<br />

tumor tissue is required prior to the start of study treatment. Blood is<br />

collected at baseline, and after 4 & 8 cycles for biomarker analysis.<br />

Genomic and expression profiling will be undertaken for correlation<br />

with response.<br />

This study was approved and funded by the National Comprehensive<br />

<strong>Cancer</strong> Network (NCCN) from general research support from Celgene<br />

Corporation.<br />

OT3-3-08<br />

Eribulin/Cyclophosphamide versus Docetaxel/Cyclophosphamide<br />

as Neoadjuvant Therapy in Locally Advanced HER2-Negative<br />

<strong>Breast</strong> <strong>Cancer</strong>: A Randomized Phase II Trial of the Sarah Cannon<br />

Research Institute<br />

Yardley DA, Hainsworth JD, Shastry M, Finney L, Burris HA.<br />

Tennessee Oncology, PLLC, Nashville, TN; Sarah Cannon Research<br />

Institute, Nashville, TN<br />

Background: Neoadjuvant chemotherapy for locally advanced breast<br />

cancer improves survival and rates of breast-conserving surgery.<br />

Pathologic complete response (pCR) after neoadjuvant therapy<br />

strongly correlates with improved disease free survival (DFS) and<br />

provides an early indicator of treatment efficacy. The expected pCR<br />

rate with a standard taxane-containing combination is approximately<br />

18%. Eribulin is a non-taxane synthetic analogue of halichondrin B<br />

that inhibits microtubule dynamics by a novel mechanism of action<br />

distinct from other tubulin-targeting agents. Eribulin is active against<br />

taxane and anthracycline pretreated metastatic breast cancer (MBC)<br />

and is well tolerated with a predictable toxicity profile. Substitution<br />

of eribulin for docetaxel in a neoadjuvant combination regimen<br />

may therefore improve efficacy. This study will evaluate the nonanthracycline<br />

eribulin/cyclophosphamide (ErC) and docetaxel/<br />

cyclophosphamide (TC) combinations as neoadjuvant therapy. The<br />

first ten patients (pts) will be evaluated for tolerability and feasibility<br />

of standard prescribed eribulin monotherapy dosing in combination<br />

with standard dose cyclophosphamide.<br />

Study Objectives: This randomized phase II trial is designed to<br />

determine the pCR rate of locally advanced, HER2-negative breast<br />

cancer treated with 6 cycles of ErC or TC. The secondary objectives<br />

are to evaluate the clinical response rate of ErC as neoadjuvant therapy<br />

and to determine the 2 year DFS of pts treated with ErC and TC.<br />

Eligibility: Females ≥ 18 years with untreated, locally advanced,<br />

HER2-negative breast cancer appropriate for neoadjuvant<br />

chemotherapy are eligible. Eligibility criteria include: adenocarcinoma<br />

histology; clinical T1-3, N0-2, M0 breast tumors; ECOG PS 0- 2;<br />

known hormone receptor status at study entry; adequate bone marrow<br />

and organ function; willingness to provide archived biopsy specimen<br />

for correlative testing. Clinical N3, T1N0M0, and T4 tumors are<br />

excluded. Upfront axillary lymph node sampling and/or definitive<br />

nodal surgery is permitted, and demonstrated pN3a disease is allowed.<br />

Trial Design: A lead-in phase of the trial will enroll 10 pts to be treated<br />

with ErC to determine safety and feasibility of the combination. If<br />

the safety is confirmed, subsequent pts will be stratified by hormone<br />

receptor status (positive vs. triple- negative) and will be randomized<br />

in a 2:1 ratio to Arm 1: ErC or Arm 2: TC. Pts on Arm 1 will receive<br />

eribulin 1.4 mg/m 2 IV (Days 1 & 8) and cyclophosphamide 600<br />

mg/m 2 IV (Day 1). Pts on Arm 2 will receive docetaxel 75 mg/m 2<br />

IV (Day 1) and cyclophosphamide 600 mg/m 2 IV (Day 1). Both<br />

regimens are repeated every 21 days for a total of 6 cycles. After<br />

completion of neoadjuvant chemotherapy, pts will undergo definitive<br />

local surgery, as determined by the treating surgeon. Archival tumor<br />

samples and residual tumor tissue at surgery will be collected for<br />

biomarker evaluations. A total of 66 pts (Arm 1: 44; Arm 2: 22) will<br />

be enrolled to this study. A pCR rate ≥ 18% in patients treated with<br />

ErC is considered a study result that merits further evaluation. This<br />

trial is pending activation and has a total accrual goal of 76 pts.<br />

www.aacrjournals.org 581s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

OT3-3-09<br />

ARTemis trial - randomised trial with neo-adjuvant chemotherapy<br />

for patients with early breast cancer<br />

Earl HM, Blenkinsop C, Grybowicz L, Vallier A-L, Cameron DA,<br />

Bartlett JMS, Murray N, Caldas C, Thomas J, Dunn JA, Higgins HB,<br />

Hiller L, Hayward L. University of Cambridge, United Kingdom;<br />

NIHR Cambridge Biomedical Research Centre, Cambridge, United<br />

Kingdom; University of Warwick, Coventry, United Kingdom;<br />

Cambridge University Hospitals NHS Foundation Trust, Cambridge,<br />

United Kingdom; Edinburgh University, Edinburgh, United Kingdom;<br />

University of Edinburgh, United Kingdom; Royal Adelaide Hospital,<br />

Adelaide, SA, Australia; <strong>Cancer</strong> Research UK Cambridge Research<br />

Institute, Cambridge, United Kingdom; Western General Hospital,<br />

Edinburgh, United Kingdom<br />

Background: Bevacizumab, a humanised monoclonal antibody<br />

which targets vascular endothelial growth factor (VEGF), has<br />

shown promising anti-tumour effect when given concurrently<br />

with taxane-based chemotherapy in breast cancer. However two<br />

neoadjuvant breast cancer trials have produced conflicting results. The<br />

GEPARQUINTO trial reported significant benefit only in the triple<br />

negative patients (pCR 27.8% vs 36.4% p=0.021). The NSABP-B40<br />

study showed significant benefit for bevacizumab only in the ER+ve<br />

patients (pathological complete response (pCR) in ER+ve population<br />

15.2% vs 23.3%, p=0.008).<br />

Trial design: ARTemis is a phase III randomised trial to determine<br />

whether the addition to neo-adjuvant chemotherapy of bevacizumab<br />

is more effective than standard chemotherapy alone in patients with<br />

HER2-negative early breast cancer. A total of 400 patients will be<br />

randomised into each of the two treatment arms which will allow an<br />

absolute difference in the pCR rates in excess of 10% to be detected<br />

at the 5% (2-sided) level of significance with an 85% power. Primary<br />

outcome is pathological complete response rates after neo-adjuvant<br />

chemotherapy, i.e. no residual invasive carcinoma in the breast, and<br />

no evidence of metastatic disease within the lymph nodes. Secondary<br />

outcomes are disease free survival, overall survival, complete<br />

pathological response rates in breast alone, radiological response<br />

after 3 and 6 cycles of chemotherapy and rate of breast conservation<br />

and toxicities. Translational work (ARTemis-Science): collection of<br />

blood samples, and paraffin-embedded formalin-fixed and fresh tissue<br />

samples to investigate whether markers for chemotherapy response/<br />

resistance, and pharmacogenetic and pharmacogenomic markers can<br />

be correlated with outcomes (pathCR and RFS) in patients randomised<br />

to receive bevacizumab versus those that do not.<br />

Eligibility criteria: Histologically confirmed HER2-negative<br />

invasive breast cancer; T2 to T4 tumour and/or large/fixed axillary<br />

nodes (>20mm / clinical N2 or N3), or inflammatory disease;<br />

suitable for, and fit to receive neo-adjuvant chemotherapy and an<br />

anti-angiogenic.<br />

Results: Recruitment into ARTemis began in April 2009 and as of<br />

June 2012 had recruited 600 (75%) patients and 65 sites are open for<br />

recruitment. ARTemis is due to complete recruitment by first quarter<br />

of 2013 and the first planned interim analysis of the primary outcome<br />

will be December 2013.<br />

Conclusion: The IDSMC reviewed the trial in June 2012 and<br />

recommended continuation of recruitment to this important trial given<br />

there were no safety concerns.<br />

OT3-3-10<br />

ASTER 70s: Benefit of adjuvant chemotherapy for oestrogen<br />

receptor-positive HER2-negative breast carcinoma in women<br />

over 70 according to Genomic Grade. A French UNICANCER<br />

Geriatric Oncology Group (GERICO) and <strong>Breast</strong> Group (UCBG)<br />

multicentre phase III trial.<br />

Brain E, Baffert S, Bonnefoi HR, Cottu P, Girre V, Lacroix-Triki<br />

M, Latouche A, Leger-Falandry C, Peyro Saint Paul HP, Rollot F,<br />

Romieux G, Servent V, Orsini C, Bonnetain F. Institut Curie, Saint-<br />

Cloud, France; Institut Curie, Paris, France; Institut Bergonié,<br />

Bordeaux, France; Centre Hospitalier La Roche sur Yon, La Roche<br />

sur Yon, France; Institut Claudius Regaud, Toulouse, France; CNAM,<br />

Paris, France; Centre Hospitalier Lyon Sud, Lyon, France; Ipsogen<br />

SA, Marseille, France; Centre Val d’Aurelle, Montpellier, France;<br />

Centre Oscar Lambret, Lille, France; R&D Unicancer, Paris, France;<br />

Centre Georges François Leclerc, Dijon, France<br />

Background<br />

Despite a high incidence of oestrogen receptor-positive (ER+)<br />

HER2-negative (HER2-) breast carcinoma (BC) in elderly women,<br />

rationale and modalities of adjuvant chemotherapy (CT) vary greatly<br />

and remain heterogeneous after 70 years, precluding evaluation<br />

of its benefit and impact on functional status and outcome. As for<br />

trials investigating innovative treatments, elderly patients are often<br />

excluded from programs evaluating new modern prognosis classifiers.<br />

Trial design<br />

ASTER 70s is a prospective multicentre trial that investigates the<br />

impact on overall survival (OS) of adjuvant CT in elderly ER+<br />

HER2- BC patients selected with a modern prognosis classifier while<br />

taking into account competing risks for mortality (EudraCT 2011-<br />

004744-22). After surgery, 2,000 patients over 70 years will have<br />

genomic grade (GG) assessed centrally by qRT-PCR on formalin-fixed<br />

paraffin-embedded samples (Ipsogen). Only those with a high GG<br />

or Equivocal (estimation∼700, group I) will be randomized between<br />

endocrine treatment alone and CT followed by endocrine treatment.<br />

CT regimen is left to the choice of investigators amongst 3 regimen<br />

of same duration [4 q3w cycles, docetaxel+cyclophosphamide<br />

(TC), doxorubicin or non pegylated liposomal doxorubicin<br />

(Myocet®)+cyclophosphamide (AC or MC), all with G-CSF],<br />

as well as endocrine treatment (aromatase inhibitor±tamoxifen).<br />

Radiotherapy will follow standard guidelines. Patients with low GG<br />

or not randomized for any reason (estimation∼1,300, group II) will be<br />

followed up as an observational cohort, with endocrine treatment only.<br />

Eligibility criteria<br />

Any ER+ HER2- BC after complete surgery, M0, any pT or pN.<br />

Normal organ functions. No specific BC treatment before surgery. No<br />

exclusion for underlying medical condition unless potential contraindication<br />

to study drugs.<br />

Specific aims<br />

OS is the primary endpoint. Secondary endpoints address competing<br />

risks for mortality, include cost-effectiveness and QTWiST analysis,<br />

geriatric questions, acceptability and quality of life (QLQ C30 and<br />

specific elderly scale ELD15). The Lee’s 4-year mortality score<br />

is calculated. The G8 screening tool is also assessed and used<br />

for stratification. Translational research will focus on ageing and<br />

prognostic biomarkers, and pharmacogenetic.<br />

Statistical methods<br />

Sample size based on 4-year OS (87.5 vs 80%), bilateral test, α=0.05,<br />

β=0.20 and HR= 0.60.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

582s<br />

<strong>Cancer</strong> Research


December 4-8, 2012<br />

<strong>Abstract</strong>s: Poster Session OT3<br />

Present accrual and target accrual<br />

The trial has been opened to inclusion in April 2012. 14 centres have<br />

been activated with 14 patients included: 7 randomized in group I,<br />

2 included in the cohort (group II) and 4 with GG determination<br />

ongoing.<br />

OT3-3-11<br />

A RANDOMIZED PHASE III TRIAL COMPARING<br />

NANOPARTICLE-BASED PACLITAXEL WITH SOLVENT-<br />

BASED PACLITAXEL AS PART OF NEOADJUVANT<br />

CHEMOTHERAPY FOR PATIENTS WITH EARLY BREAST<br />

CANCER (GeparSepto) GBG 69<br />

Untch M, Jackisch C, Blohmer J-U, Costa S-D, Denkert C,<br />

Eidtmann H, Gerber B, Hanusch C, Hilfrich J, Huober J, Kuemmel<br />

S, Schneeweiss A, Paepke S, Loibl S, Nekljudova V, von Minckwitz<br />

G. Helios Kliniken Berlin; Klinikum Offenbach; <strong>San</strong>kt-Gertrauden<br />

Berlin; University Magdeburg; Charite Berlin; University Kiel;<br />

University Rostock; Rotkreuzklinikum Muenchen; Eilenriedeklinik<br />

Dueseldorf; University Duesseldorf; Kliniken Essen Mitte;<br />

Frauenklinik Muenchen; German <strong>Breast</strong> Group, Neu-Isenburg<br />

Background: Anthracycline/taxane based regimen are standard of<br />

care for neoadjuvant therapy in breast cancer. Recent data from the<br />

neo-Tango study suggest that a reverse sequence of taxane followed<br />

by the anthracycline can achieve higher pCR rates. Solvent-based<br />

taxanes (paclitaxel, docetaxel) cause severe toxicities most likely<br />

by the solvents such as cremophor. Nab-paclitaxel is a solvent-free<br />

formulation of paclitaxel encapsulated in albumin. It is believed that<br />

nab-Paclitaxel compared to solvent based paclitaxel followed by<br />

conventional dosed EC might further improve the pCR rate in breast<br />

cancer patients receiving neoadjuvant treatment. Previous studies<br />

have shown that dual anti-HER blockade is superior to trastuzumab<br />

alone resulting in an increase of pCR rate by 20%.<br />

Patients and Methods: The GeparSepto trial, a neoadjuvant,<br />

randomized phase III study, planned to include 1200 pts, randomized<br />

to nab-paclitaxel versus conventional, solvent based paclitaxel given<br />

weekly for 12 weeks followed in both arms by 4 cylces conventionally<br />

dosed EC. The primary objective is to compare the rate of pCR (ypT0<br />

+ ypN0). Further objectives are to compare the pCR rate in predefined<br />

subgroups, pCR by other definition, the clinical response rate and<br />

the rate of breast conserving surgery after chemotherapy in the two<br />

different treatment arms.<br />

Women with untreated, histologically confirmed uni- or bilateral, cT2-<br />

cT4d carcinoma, and no clinically relevant cardiovascular and other<br />

co-morbidities are randomized to receive either paclitaxel (80mg/<br />

m²) or nab-paclitaxel (150 mg/ m²) day 1, 8, 15, q d 22 for 4 cycles<br />

followed by conventional EC (E (90mg/m²)+C (600 mg/m²)) on day 1<br />

q day 22 for 4 cycles. HER2 positive pts receive trastuzumab (loading<br />

dose 8mg/kg followed by 6 mg/kg) and pertuzumab (loading dose 840<br />

mg followed by 420 mg) q3w concomitantly to the chemotherapy.<br />

Biomaterial including FFPE form core biopsy, serum, plasma, full<br />

blood is collected before randomization, after the 12 cycles for (nab-)<br />

paclitaxel therapy and after the 4 cycles of EC before surgery. The<br />

HER2,estrogen receptor, progesterone receptor, Ki67 and SPARC<br />

status will be centrally tested by immunohistochemistry prior to<br />

randomization for stratification. A broad translational program is<br />

planned. It has been assumed that solvent based taxane will achieve an<br />

overall pCR rate of 33% to be increased using nab-paclitaxel to 41%,<br />

corresponding to an odds ratio of 1.41. If 596 patients are enrolled into<br />

each arm, a χ2-test will have an 80% power with a 2-sided significance<br />

level α=0.05 to show the superiority of nab-paclitaxel. Closed test<br />

procedure will be used to test for non-inferiority of nab-paclitaxel first.<br />

The trial is registered under NCT01583426. It is financially supported<br />

by Roche and Celgene.<br />

Results: The centres have been initiated after approval by ethics<br />

committee and authorities.<br />

First patient in will be this months. It is planned to recruit 18 months<br />

in 100 sites in Germany.<br />

Conclusion: Geparsepto will investigate the efficacy of neoadjuvant<br />

nab-paclitaxel compared to solvent based paclitaxel given weekly<br />

and the dual blockade with Trastuzumab and Pertuzumab in HER<br />

2 positive BC.<br />

OT3-4-01<br />

PROMIS: Prospective Registry Of MammaPrint in breast cancer<br />

patients with an Intermediate recurrence Score<br />

Soliman H, Untch S, Stork-Sloots L. Moffitt <strong>Cancer</strong> Center; Agendia<br />

Inc; Agendia NV<br />

Background: Gene expression profiling in breast cancer offers the<br />

potential to improve prognostic accuracy, treatment choice, and<br />

health outcomes in women diagnosed with early-stage breast cancer.<br />

Numerous gene-profiling assays are now available, which can be<br />

applied to a single tumor specimen to provide physicians with a more<br />

complete basis for treatment decisions.<br />

• MammaPrint is a DNA microarray-based in vitro diagnostic that<br />

measures the activity of 70 genes to estimate whether patients are<br />

at high risk or low risk of developing metastases within the first 10<br />

years after curative surgery.<br />

• BluePrint is an 80-gene molecular subtyping profile that<br />

discriminates between three breast cancer subtypes: Luminal, HER2,<br />

and Basal.<br />

• TargetPrint provides a quantitative measurement of estrogen receptor<br />

(ER), progesterone receptor (PR), and HER2, and can serve as a<br />

reliable second pathology assessment for locally-assessed parameters.<br />

• Oncotype DX measures expression of five reference genes and 16<br />

cancer-related genes, quantifying the risk of distant recurrence in<br />

patients with ER+ early breast cancer who are treated with adjuvant<br />

hormonal therapy, and predicting clinical benefit with additional<br />

adjuvant chemotherapy.<br />

Trial design: PROMIS is a prospective, observational, case-only<br />

study that will investigate the additional value of MammaPrint,<br />

BluePrint and TargetPrint in women with an intermediate Oncotype<br />

DX score. An initial CRF – capturing baseline patient characteristics,<br />

pathology information, Oncotype DX score and the recommended<br />

treatment plan – will be completed before receiving the MammaPrint<br />

result. A second CRF – capturing the recommended treatment – will<br />

be completed within 4 weeks after receiving the MammaPrint result.<br />

Eligibility: The study will include women aged ≥18 years with<br />

histologically proven invasive stage I-II, node-negative, hormone<br />

receptor-positive, HER2-negative breast cancer and Oncotype DX<br />

score of 18–30 who signed informed consent.<br />

Objectives: The objectives of the study are to:<br />

1. Describe the frequency of chemotherapy + endocrine versus<br />

endocrine-alone decisions in Oncotype DX intermediate score<br />

patients.<br />

2. Assess the impact of MammaPrint on chemotherapy + endocrine<br />

versus endocrine-alone treatment decisions.<br />

3. Assess the distribution of MammaPrint Low and High Risk in<br />

patients with an intermediate recurrence score.<br />

4. Assess concordance of TargetPrint ER, PR and HER2 results with<br />

Oncotype DX ER, PR and HER2 and with locally assessed IHC/<br />

FISH ER, PR and HER2.<br />

www.aacrjournals.org 583s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

5. Compare clinical subtype based on IHC/FISH ER, PR, HER2 and<br />

Ki-67 (if available) with BluePrint molecular subtype.<br />

Statistical methods: As the project is exploratory, sample size<br />

calculations do not apply as only descriptive statistics will be used.<br />

The frequency of chemotherapy + endocrine versus endocrinealone<br />

decisions in Oncotype DX intermediate score patients will<br />

be calculated before and after receiving the MammaPrint result.<br />

A Chi-square test will be performed for the comparison of the two<br />

proportions.<br />

Accrual: A total of ∼300 eligible patients will be enrolled from<br />

multiple institutions.<br />

Clinical trial registry number: NCT01617954<br />

OT3-4-02<br />

MINT I: Multi-Institutional Neo-adjuvant Therapy, MammaPrint<br />

Project I<br />

Cox C, Blumencranz P, Reintgen D, Saez R, Howard N, Gibson J,<br />

Stork-Sloots L, Glück S. University of South Florida; Morton Plant<br />

Hospital; Florida Hospital North Pinellas; Plano <strong>Cancer</strong> Institute;<br />

Agendia Inc; Agendia NV; Miller School of Medicine, University<br />

of Miami<br />

Background: Women with locally advanced breast cancer (LABC)<br />

are often treated with neo-adjuvant chemotherapy to reduce the size<br />

of the tumor prior to surgery, to enable breast conserving surgery<br />

and to observe the clinical effect of therapy in real time. Studies<br />

have shown that the 25–27% of individuals who have a pathologic<br />

complete response (pCR) to neoadjuvant therapy have a survival<br />

advantage of 80% in 5 years, which is double the expected survival<br />

of the remaining patients without pCR. If patients who are likely<br />

to show a pCR could be identified prior to initiation of therapy, it<br />

would enable more informed treatment decisions – patients likely to<br />

respond would be served well by current neoadjuvant chemotherapy<br />

protocols, while those unlikely to respond may be better suited to<br />

innovative new strategies for drug discovery [von Minckwitz et al.<br />

JCO 2006]. Genomics assays, which are widely used to provide<br />

prognostic and predictive information in early breast cancer, have the<br />

potential to provide information on the likelihood of a patient with<br />

LABC responding to neo-adjuvant therapy [Glück et al. ASCO 2012].<br />

Trial design: MINT I is a prospective study designed to test the<br />

ability of molecular profiling, as well as traditional pathologic and<br />

clinical prognostic factors, to predict responsiveness to neo-adjuvant<br />

chemotherapy in patients with LABC. MammaPrint risk profile,<br />

BluePrint molecular subtyping profile, TargetPrint estrogen receptor<br />

(ER), progesterone receptor (PR) and HER2 single gene readout, and<br />

the 56-gene TheraPrint Research Gene Panel will be analyzed on a<br />

fresh tumor specimen using the whole genome array. Patients will<br />

receive neo-adjuvant chemotherapy pre-specified in the protocol.<br />

Response will be measured centrally. pCR is defined as the absence<br />

of invasive carcinoma in both the breast and axilla at microscopic<br />

examination of the resection specimen, regardless of the presence of<br />

carcinoma in situ.<br />

Eligibility: The study will include women ≥18 years with<br />

histologically-proven invasive breast cancer T2 (≥3.5cm)-T4, N0M0<br />

or T2-T4N1M0, adequate bone marrow reserves and normal renal<br />

and hepatic function who signed an IRB approved informed consent.<br />

Objectives: The objectives of the study are to:<br />

1. Determine the predictive power of MammaPrint and BluePrint<br />

for sensitivity to neo-adjuvant chemotherapy as measured by pCR.<br />

2. Compare TargetPrint ER, PR and HER2 with local and centralized<br />

IHC and/or CISH/FISH assessment.<br />

3. Identify correlations between TheraPrint and response to neoadjuvant<br />

chemotherapy.<br />

4. Identify and/or validate predictive gene expression profiles of<br />

clinical response or resistance to neo-adjuvant chemotherapy.<br />

5. Compare BluePrint with IHC-based subtype classification.<br />

Statistical methods: Standard statistical tests such as the Pearson Chisquare<br />

test will be used to characterize and evaluate the relationship<br />

between chemoresponsiveness and gene expression patterns.<br />

Accrual: A total of 226 eligible patients will be enrolled from multiple<br />

institutions. To date (June 06, 2012), 31 patients have been enrolled.<br />

Clinical trial registry number: NCT01501487.<br />

OT3-4-03<br />

Prospective neo-adjuvant registry trial linking MammaPrint,<br />

subtyping and treatment response: Neo-adjuvant <strong>Breast</strong> Registry<br />

– Symphony Trial (NBRST)<br />

Whitworth P, Gittleman M, Akbari S, Nguyen B, Baron P, Rotkiss M,<br />

Beatty J, Gibson J, Stork-Sloots L, de Snoo F, Beitsch P. Nashville<br />

<strong>Breast</strong> Center; <strong>Breast</strong> Care Specialists; Virginia Hospital Center;<br />

Todd <strong>Cancer</strong> Institute, Long Beach Memorial Medical Center;<br />

<strong>Cancer</strong> Specialists of Charleston; Northern Indiana <strong>Cancer</strong> Research<br />

Consortium; The <strong>Breast</strong> Place Charleston; Agendia Inc; Agendia NV;<br />

Dallas Surgical Group<br />

Background: The FDA-approved MammaPrint 70 gene profile is<br />

performed on a diagnostic multi-gene array. This platform enables<br />

additional gene expression profiles to be analyzed simultaneously on<br />

one tumor specimen. One such gene expression profile is BluePrint,<br />

an 80 gene molecular subtyping profile that discriminates between<br />

three distinctive subtypes; Basal-type, Luminal-type, and HER2-type.<br />

By combining MammaPrint and BluePrint, patients can be stratified<br />

into the following subgroups: Luminal-type/MammaPrint Low<br />

Risk; Luminal-type/MammaPrint High Risk; HER2-type and Basaltype.<br />

Marked differences are observed in response to neo-adjuvant<br />

treatment in groups stratified by MammaPrint and BluePrint [Glück<br />

et al. ASCO 2012].<br />

Trial design: NBRST is a prospective observational, case-only<br />

study linking MammaPrint and BluePrint to neo-adjuvant treatment<br />

response and Distant Metastases-Free Survival (DMFS) and Relapse-<br />

Free Survival (RFS). MammaPrint and BluePrint are assessed on fresh<br />

or formalin-fixed, paraffin-embedded core needle biopsy samples.<br />

Treatment is at the discretion of the physician, adhering to NCCN<br />

approved regimens or a recognized alternative.<br />

Eligibility: The study will include women aged 18–90 with<br />

histologically proven breast cancer, who have started or are scheduled<br />

to start neo-adjuvant chemotherapy or neo-adjuvant hormonal therapy,<br />

after successful MammaPrint and BluePrint assessment, and who<br />

provide written informed consent. Additional inclusion criteria are<br />

that women must have had no excisional biopsy or axillary dissection,<br />

no confirmed distant metastatic disease, and no prior therapy for the<br />

treatment of breast cancer.<br />

Objectives: The main objectives of this registry study are to:<br />

1. Measure chemosensitivity (as defined by pathological complete<br />

response [pCR]) or endocrine sensitivity (as defined by decrease<br />

in longest tumor diameter or RCB1) in the molecular subgroups as<br />

determined by combining MammaPrint and BluePrint results.<br />

2. Compare local IHC and FISH results with TargetPrint results and<br />

BluePrint results.<br />

3. Document impact of MammaPrint, TargetPrint and BluePrint<br />

results on treatment decisions.<br />

4. Assess the 2–3 and 5-year DMFS and RFS for the different<br />

molecular subgroups.<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

584s<br />

<strong>Cancer</strong> Research


December 4-8, 2012<br />

<strong>Abstract</strong>s: Poster Session OT3<br />

Statistical methods: Comparison of response rates between different<br />

molecular subgroups will be conducted using the Pearson Chi-square<br />

test.<br />

Accrual: The trial started in August 2011 and there are currently<br />

36 participating sites in the US. To date (June 06, 2012), 158 of<br />

the planned 400 patients have been enrolled.Clinical trial registry<br />

number: NCT01479101.<br />

OT3-4-04<br />

InVite: an observational pilot study evaluating the feasibility of<br />

genome-wide association studies using self-reported data from<br />

patients with metastatic breast cancer treated with bevacizumab<br />

Leyland-Jones B, Faoro L, Barnholt K, Kiefer A, Yager S, Yi J, Turner<br />

B, Keane A, Wang L, Eriksson N, Milián ML, O’Neill V. Genentech,<br />

Inc.; 23andMe; <strong>San</strong>ford Research/USD<br />

Background: Personalized healthcare tailors treatments to patients<br />

and their disease characteristics through the use of genetics and other<br />

biomarkers. Genetic differences among individuals may explain<br />

variations in drug treatment response, including side effects. With such<br />

information physicians could make more informed decisions about<br />

drugs and dosing for a given individual, thereby improving patient<br />

care. Although there has been some success, to a large extent genetic<br />

variation related to drug response remains unexplained.<br />

Bevacizumab, a humanized monoclonal antibody against the<br />

angiogenic factor VEGF, has demonstrated activity in patients with<br />

metastatic breast cancer (MBC). The InVite study will evaluate the<br />

feasibility of performing genomewide association studies using selfreported<br />

information collected via an online platform from patients<br />

with MBC who have been treated with bevacizumab. Using novel<br />

methodology in a convenient, user-friendly, and scientifically rigorous<br />

format, InVite ultimately aims to identify potential pharmacogenetic<br />

associations in this patient population.<br />

Trial design: InVite is a pilot, non-interventional, observational, webbased,<br />

prospective cohort study designed to collect patient-reported<br />

safety, efficacy, and genetic data from patients with MBC treated with<br />

bevacizumab. Data on demographics, breast cancer disease status,<br />

cancer treatment history, bevacizumab-related outcomes, and certain<br />

safety events will be collected directly from patients entirely via<br />

online surveys. Patients will be asked to complete surveys at the time<br />

of enrollment and then every 3 months for 1 year after enrollment. A<br />

saliva sample for DNA collection will be gathered using an at-home<br />

kit. Evaluations of data quality and collection feasibility will be<br />

conducted intermittently. There will be an optional substudy to allow<br />

for blood sample collection for DNA analysis.<br />

Eligibility criteria: ≥18 years of age, residing in the US, locally<br />

recurrent breast cancer or MBC, currently being or having been<br />

treated with bevacizumab starting on or before Dec 31, 2011, fluent<br />

in English, and access to a computer with an internet connection.<br />

Specific aims: The primary objective is to assess the feasibility of<br />

recruiting subjects and collecting biospecimens and self-reported<br />

data online. The secondary objective is to characterize the patient<br />

population. Exploratory objectives include analyzing potential<br />

associations between genetic polymorphisms and 1) bevacizumabinduced<br />

hypertension, the most common bevacizumab-related adverse<br />

event, and 2) patient-reported time-to-progression.<br />

Statistical methods: Baseline demographics, clinical and treatment<br />

characteristics of enrolled patients will be summarized. Each<br />

polymorphism genotyped will be tested for association with the<br />

defined endpoint using appropriate statistical modeling.<br />

Present and target accrual: Accrual as of May 23, 2012 is 82 patients.<br />

Target accrual is 1000 patients.<br />

OT3-4-05<br />

Use of early CIRculating tumor CElls count changes to guide<br />

the use of chemotherapy in advanced metastatic breast cancer<br />

patients: the CirCe01 randomized trial<br />

Bidard F-C, Asselain B, Baffert S, Brain E, Campone M, Delaloge<br />

S, Bachelot T, Tubiana-Mathieu N, Pelissier S, Pierga J-Y. Institut<br />

Curie; Institut de Cancérologie de l’Ouest; Institut Gustave Roussy,<br />

Villejuif; Centre Léon Bérard, Lyon; CHU Limoges<br />

Background<br />

After the SWOG0500 trial, the CirCe01 trial (NCT01349842) is the<br />

second randomized attempt to demonstrate that patients who received<br />

a first cycle of chemotherapy and whose CTC count did not drop<br />

should be switched off this chemotherapy.<br />

The clinical setting used here is a population with high chemoresistance<br />

prevalence, i.e. patients starting a third line chemotherapy; the CTC<br />

test will be also repeated at the beginning of every new chemotherapy<br />

line in patients randomized in the CTC arm (i.e. to evaluate the 3 rd ,<br />

4 th , 5 th … lines).<br />

In the CTC arm, it has been hypothesized that many patients with<br />

chemoresistant tumor will experience repeated early chemotherapy<br />

changes (e.g. 3 chemotherapy regimens tested in 9 weeks), giving<br />

support to the discontinuation of inefficient, toxic and costly<br />

chemotherapies and the start of palliative care. It is believed, for this<br />

subgroup of patients, that such de-escalating management will benefit<br />

both patients and healthcare systems. For some other patients, it is<br />

also expected that they will benefit from the early chemotherapy turnaround<br />

and find earlier an efficient therapy among all those tested.<br />

Methods<br />

The main inclusion criteria are metastatic breast cancer patients<br />

starting a third line of chemotherapy and an elevated CTC count at<br />

baseline.<br />

304 metastatic breast cancer patients will be randomized between the<br />

standard arm, in which the treatment management is made following<br />

the current standard of care, and the CTC-driven arm, in which the<br />

“response” after every first cycle of any new chemotherapy line is<br />

assessed by CTC count made before the second cycle. Chemotherapies<br />

which did not lead to a significant reduction of CTC count are<br />

discontinued, and the patient might be offered another chemotherapy<br />

regimen (which will be also evaluated by CTC).<br />

The medical primary endpoint of the trial is the overall survival,<br />

whereas a medico-economic study as a co-primary endpoint. Several<br />

secondary endpoints are pre-planned, including progression-free<br />

survival, quality of life, anxiety/depression and comparison with<br />

serum markers.<br />

OT3-4-06<br />

Circulating tumor cells to guide the choice between chemotherapy<br />

and hormone therapy as first line treatment for hormone<br />

receptors positive metastatic breast cancer patients: the STIC<br />

CTC METABREAST trial<br />

Bidard F-C, Baffert S, Hajage D, Brain E, Armanet S, Simondi C,<br />

Mignot L, Asselain B, Tresca P, Pierga J-Y. Institut Curie, France<br />

Background<br />

Baseline CTC count already demonstrated its accuracy as an<br />

independent prognostic marker in metastatic breast cancer (MBC),<br />

before the start of any treatment (Cristofanilli, NEJM 2004; Pierga<br />

Ann Oncol 2011) for progression free survival and overall survival.<br />

Multivariate analyses performed in both a pooled analysis (Liu, ASCO<br />

2011) and in the IC 2006-04 study showed that the other independent<br />

prognostic factors were the performance status and hormone-receptor<br />

(HR) status. Oppositely, the other criteria that are frequently used to<br />

www.aacrjournals.org 585s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

choose between hormone therapy and chemotherapy for the treatment<br />

of first line hormone receptor-positive MBC patients (e.g. number of<br />

metastatic sites, visceral disease, metastasis-free interval…) were not<br />

independent prognostic factors. As CTC count is highly correlated to<br />

PFS, we designed a large pivotal phase III trial to evaluate the use of<br />

CTC to determine the disease aggressiveness and the choice of first<br />

line treatment in potentially hormonosensitive MBC.<br />

Trial design<br />

First line metastatic breast cancer patients will be randomized between<br />

the clinician choice and CTC count-driven choice. In the CTC arm,<br />

patients with ≥5 CTC/7.5ml will receive chemotherapy whereas<br />

patients with


December 4-8, 2012<br />

Author Index<br />

1200.67 Trial Group OT1‐1‐15<br />

1200.89 Trial Group OT1‐1‐14<br />

A. Molinolo, A P1‐05‐23<br />

Aaronson, NK<br />

P4‐11‐01<br />

Abadie‐Lacourtoisie, S P1‐14‐21<br />

Abbruzzese, BC<br />

P1‐01‐04<br />

ABCSG<br />

P2‐10‐02<br />

Abdel‐Fatah, TMA P2‐10‐22, P3‐06‐13,<br />

P6‐07‐09, P6‐07‐18<br />

AbdElmageed, Z P4‐06‐14, P4‐06‐16<br />

Abdel‐Rasoul, M<br />

P5‐18‐17<br />

Abd‐El‐Tawab, R<br />

P1‐05‐11<br />

Abdraboh, M<br />

P4‐06‐16<br />

Abdul‐Hussein, M<br />

PD03‐09<br />

Abdulsater, F<br />

P3‐04‐04<br />

Abe, H<br />

PD06‐02, P1‐15‐11<br />

Abe, K<br />

P1‐01‐27<br />

Abernethy, A<br />

P3‐06‐07<br />

Abidoye, O<br />

P6‐09‐11<br />

Abkevich, V<br />

PD09‐04<br />

Abraham, J PD07‐09, PD09‐06<br />

Abraham, JE<br />

P3‐08‐05<br />

Abramson, RG<br />

P4‐01‐03<br />

Abramson, VG P2‐14‐04, P4‐01‐03,<br />

P6‐10‐05<br />

Abranches, MV<br />

P3‐07‐12<br />

Abrial, C<br />

P3‐06‐20<br />

Abu‐Khalaf, M PD05‐05, P4‐01‐02<br />

Abulkhair, O<br />

OT1‐1‐08<br />

Aburto, L<br />

P3‐10‐03<br />

Ackerstaff, E<br />

P6‐01‐01<br />

Adamowicz, K P3‐12‐09, P3‐13‐03<br />

Adams, J<br />

P1‐09‐05<br />

Adams, JE<br />

P4‐13‐09<br />

Adams, JW<br />

P2‐05‐06<br />

Adams, RF P1‐06‐01, P4‐01‐01<br />

Adams, S<br />

P5‐20‐05<br />

Adane, E<br />

P5‐14‐05<br />

Addison, C<br />

P3‐13‐05<br />

Addison, CL P2‐05‐12, P2‐05‐13<br />

Adelaide, J<br />

P5‐03‐01<br />

Ademar, BJ<br />

P3‐01‐03<br />

Ademuyiwa, FO<br />

P3‐07‐02<br />

Adie, SG<br />

OT2‐1‐04<br />

Adnet, P‐Y<br />

S6‐2, P3‐05‐03<br />

Adrada, B<br />

P1‐07‐06<br />

Aebi, S<br />

S3‐2, P6‐07‐06<br />

Affan, AM<br />

P4‐17‐06<br />

Affen, J<br />

P1‐09‐05<br />

Aft, R<br />

P2‐04‐02<br />

Agabiti, E P1‐01‐10, P1‐01‐11<br />

Agalliu, I<br />

PD03‐09<br />

Agar, NYR<br />

OT1‐1‐11<br />

Agarwal, J<br />

P4‐14‐02<br />

Agarwal, P<br />

P2‐10‐03<br />

Agarwal, S<br />

P4‐14‐02,<br />

P4‐14‐02, OT2‐3‐11<br />

Agelaki, S P2‐01‐03, P2‐01‐07<br />

Ager, E<br />

P3‐12‐03<br />

Agrawal, R S4‐1<br />

Agresti, R<br />

P4‐06‐09<br />

A’Hern, R<br />

PD07‐07,<br />

P1‐13‐03, P3‐06‐09<br />

A’Hern, RP<br />

P6‐07‐15<br />

Aherne, ST<br />

P3‐04‐12<br />

Ahirwar, D<br />

P1‐05‐17<br />

Ahlers, S<br />

P5‐17‐03<br />

Ahmed, I<br />

P6‐09‐09, P6‐09‐10<br />

Ahmed, KM P1‐05‐08, P4‐06‐05<br />

Ahmed, S<br />

P1‐14‐15<br />

Ahmed, SA<br />

P3‐08‐06<br />

Ahmed, SI<br />

P4‐14‐14<br />

Ahn, E<br />

P1‐12‐03<br />

Ahn, JH<br />

P2‐05‐20<br />

Ahn, J‐H PD09‐05, P2‐02‐02, P3‐12‐05<br />

Ahn, JS PD09‐05, P2‐05‐20, P4‐06‐13<br />

Ahn, SD<br />

P3‐12‐05<br />

Ahn, SG<br />

P2‐05‐14<br />

Ahn, SH<br />

P3‐01‐02, P3‐12‐05<br />

Ahn, SK<br />

PD05‐07, P1‐01‐08<br />

Ahn, S‐K<br />

P1‐14‐16<br />

Ahrends, T<br />

P5‐04‐01<br />

Ahrendt, GM S2‐1<br />

Aiello, P<br />

P4‐06‐09<br />

Aigner, J<br />

P3‐06‐08, P3‐06‐19<br />

Aihara, T<br />

P2‐13‐04, P2‐13‐07<br />

Aird, E S4‐1<br />

Aivazyan, L<br />

PD03‐04<br />

Akazawa, K<br />

PD07‐05<br />

Akbari, S<br />

OT3‐4‐03<br />

Akcakanat, A<br />

P1‐07‐06<br />

Akens, MK<br />

P4‐03‐04<br />

Akhtar, A<br />

P6‐05‐02<br />

Akimoto, E<br />

P6‐07‐26<br />

Akita, H<br />

P4‐04‐08<br />

Akiyama, F P1‐01‐12, P2‐10‐18<br />

Aktas, B<br />

S1‐7, P1‐14‐05,<br />

P2‐10‐08, OT1‐1‐10<br />

Al Riyami, H<br />

P4‐06‐16<br />

Al Sarakbi, W<br />

P6‐05‐11<br />

Alabek, ML<br />

P4‐11‐06<br />

Alarcon, J<br />

P3‐12‐08<br />

Alazawi, D<br />

P1‐01‐15<br />

Alba, E<br />

P2‐10‐11<br />

Albain, K<br />

PD10‐02, P1‐13‐01<br />

Albain, KS<br />

P1‐12‐04<br />

Albanell, J<br />

P5‐18‐05<br />

Albarracin, C<br />

P5‐20‐09<br />

Albarracin, CT<br />

P6‐07‐25<br />

Albino, T<br />

P2‐10‐05<br />

Albiter, M<br />

P4‐13‐03<br />

Al‐Ejeh, F<br />

P6‐10‐06<br />

Alexa, U<br />

P5‐15‐07<br />

Ali, A<br />

P6‐08‐04<br />

Ali, RA<br />

P4‐17‐06<br />

Ali, S<br />

P2‐10‐16, P2‐10‐22<br />

Ali, SM<br />

P2‐10‐31, P3‐06‐33<br />

Ali, ZA<br />

P4‐09‐12<br />

Al‐Janabi, S<br />

PD02‐03<br />

Alkhayyat, SS<br />

P2‐13‐08<br />

Al‐Kofahi, Y P3‐05‐05, P3‐05‐06<br />

Allen, C<br />

P4‐02‐05<br />

Allen, JD<br />

P5‐14‐02<br />

Allen, JW<br />

OT3‐3‐07<br />

Allers, TM<br />

P6‐06‐01<br />

Allevi, G<br />

PD07‐03<br />

Allison, J<br />

OT3‐1‐01<br />

Allison, MAK<br />

P3‐06‐12<br />

Allochio Filho, JF<br />

P6‐11‐13<br />

Allred, C<br />

PD07‐01<br />

Allred, JB<br />

P1‐12‐06<br />

Allum, W P1‐01‐10, P1‐01‐11<br />

Almendro, V P3‐05‐04, P4‐06‐01<br />

Alnæs, GG<br />

P5‐05‐03<br />

Alpaugh, KR<br />

PD05‐01<br />

Alpaugh, RK PD05‐03, P6‐02‐05<br />

Alperovich, M P4‐14‐03, P4‐17‐07<br />

Alran, S<br />

PD07‐04, P1‐01‐07,<br />

P1‐01‐19, OT2‐1‐01<br />

Alsner, J<br />

P3‐04‐03<br />

Alterovitz, G<br />

P1‐15‐12<br />

Altevogt, P P3‐06‐08, P3‐06‐19<br />

Althaus, B<br />

P5‐18‐11<br />

Alvarado, M<br />

S4‐2, P3‐08‐06,<br />

P4‐14‐04, P4‐16‐07<br />

Alvarado, MD<br />

P5‐15‐01<br />

Alvarez, J<br />

P5‐01‐09<br />

Alvarez, RH P2‐10‐32, P3‐10‐05,<br />

P5‐10‐02, P5‐20‐13,<br />

P6‐10‐02, OT2‐3‐10<br />

Alvarez, RMa<br />

PD08‐06<br />

Alvarez de Lacerda, LC P5‐03‐10<br />

Alvarez‐Cid, M<br />

P4‐02‐07<br />

Alvarez‐Garriga, C<br />

P5‐07‐06<br />

Amadori, D<br />

P5‐18‐01<br />

Amant, F P2‐10‐12, P6‐05‐05,<br />

P6‐07‐05, P6‐07‐29<br />

Ambros, T<br />

P1‐12‐03<br />

Ambrosone, CB<br />

P3‐07‐02<br />

Ameer‐Beg, S<br />

P2‐10‐29<br />

Ames, M<br />

PD10‐08<br />

Amir, E P2‐05‐12, P2‐10‐23, P3‐13‐05<br />

Amornphimoltham, P P1‐05‐23<br />

Amsellem, M P6‐09‐09, P6‐09‐10<br />

Anan, K<br />

P3‐02‐06<br />

Anastario, M<br />

P4‐13‐02<br />

Anastasiadis, PZ<br />

P1‐05‐24<br />

Anchisi, S<br />

P6‐04‐05<br />

Andergassen, U<br />

P2‐01‐02,<br />

P2‐01‐11, P4‐13‐11<br />

Anders, CK<br />

P3‐12‐04<br />

Andersen, J<br />

P4‐11‐04<br />

Anderson, D<br />

P2‐06‐04<br />

Anderson, EE<br />

P5‐13‐03<br />

Anderson, L<br />

P2‐08‐02<br />

Anderson, N<br />

PD03‐04<br />

Anderson, RL<br />

P3‐10‐09<br />

Anderson, S<br />

P2‐05‐06<br />

Anderson, SJ S3‐2, P6‐07‐06, OT1‐2‐01<br />

Anderson, SK<br />

PD10‐04<br />

Anderson, SM<br />

P5‐10‐04<br />

Andersson, M<br />

P1‐09‐01<br />

Andorfer, CA<br />

P1‐05‐24<br />

Andrade, VP PD04‐05, PD05‐02<br />

Andrade, W<br />

P4‐02‐03<br />

André, F<br />

P3‐06‐04,<br />

P5‐13‐02, OT2‐2‐03<br />

Andre, R<br />

P1-15-03<br />

Andreis, D<br />

PD07‐03<br />

Andrieux, N<br />

P1‐01‐21<br />

Aneja, S<br />

P3‐14‐06<br />

Anfossi, S<br />

P2‐03‐01,<br />

P5‐04‐06, P5‐10‐02<br />

Ang, D<br />

P2‐08‐03<br />

Ang, P<br />

P3‐08‐09<br />

Ang, X<br />

P2‐11‐06<br />

Anglin, BV<br />

P1‐15‐09<br />

Angulo‐Gonzalez, AM P5‐20‐13<br />

Anh‐Do, K<br />

P1‐07‐06<br />

Anthony, SP<br />

P6‐11‐10<br />

Antón, A S1‐7 , P1‐07‐18, P2‐01‐06<br />

Anton‐Culver, H<br />

P4‐13‐13<br />

Antúnez, JR<br />

P1‐01‐29<br />

Aogi, K<br />

P1‐14‐02, P2‐13‐04,<br />

P2‐13‐07, P3‐06‐18, P6‐04‐27<br />

Aparicio, A<br />

P6‐04‐12<br />

Aparicio, S<br />

P2‐10‐20<br />

Apple, SK<br />

P4‐08‐05<br />

Appleman, LJ<br />

PD09‐06<br />

Aramendia, JM<br />

P5‐16‐03<br />

Arap, W<br />

P3‐10‐09<br />

Araujo Neto, JT<br />

P1‐01‐28<br />

Arcangeli, A<br />

P5‐10‐01<br />

Arce Salinas, C<br />

P4‐16‐14<br />

Ardavanis, A<br />

P5‐16‐05<br />

Areaga, CL S3‐6<br />

Arena, FP<br />

P6‐10‐01<br />

Arevalo, E<br />

P5‐16‐03<br />

Argiles, G<br />

P6‐10‐07<br />

Aridgides, PD<br />

P3‐12‐02<br />

Arizcum, A P1‐07‐18, P2‐01‐06<br />

Arju, R<br />

P5‐03‐02<br />

Arlen, PM<br />

P5‐16‐06<br />

Arlinghaus, LR<br />

P4‐01‐03<br />

Arlukowicz‐Czartoryska, B P2‐10‐31<br />

Armanet, S<br />

OT3‐4‐06<br />

Armstrong, A<br />

P4‐06‐07<br />

Armstrong, AC<br />

P2‐14‐06<br />

Arnadottir, SS<br />

P5‐10‐05<br />

Arnaout, A PD09‐01, P5‐13‐01<br />

Arnedos, M<br />

P5‐13‐02<br />

Arneson, TJ<br />

P1‐15‐01<br />

Arnoud, L<br />

PD05‐08<br />

Arnould, L P3‐08‐07, P3‐14‐03<br />

Arora, R<br />

OT1‐1‐16<br />

Arteaga, C<br />

OT2‐3‐09<br />

Arteaga, CL PD01‐04, P2‐14‐04,<br />

P5‐19‐01, P6‐10‐05<br />

Arthur, D<br />

P1‐15‐09, P4‐16‐03<br />

Arthur, DW P4‐16‐09, OT1‐2‐01<br />

Arun, BK<br />

P2‐10‐32<br />

Arveux, P<br />

P3‐08‐07<br />

Aryandono, T<br />

P3‐01‐08<br />

Asada, Y<br />

P2‐11‐02<br />

Asaga, S<br />

P3‐02‐06<br />

Asano, T<br />

P4‐12‐06<br />

Asanuma, F<br />

P4‐09‐08<br />

Ashby, WJ<br />

P3‐05‐09<br />

Ashcraft, K<br />

P6‐02‐02<br />

Ashcroft, L<br />

P1‐09‐05<br />

Asher, M<br />

P2‐08‐01<br />

Ashfaq, R<br />

P3‐06‐21<br />

Ashikari, AY<br />

P4‐14‐01<br />

Ashworth, A PD05‐08, P3‐08‐04<br />

Askren, MK S6‐3<br />

Asmann, YW<br />

PD10‐04<br />

Assaf, E<br />

P4‐16‐10<br />

Assayag, F<br />

P6‐04‐14<br />

Asselain, B P1‐13‐04, P1‐14‐03,<br />

P1‐14‐18, P2‐01‐13,<br />

OT3‐4‐05, OT3‐4‐06<br />

Astley, SM<br />

P4‐13‐09<br />

Asztalos, S<br />

P6‐04‐24<br />

Atale, R<br />

PD05‐06, P1‐03‐02<br />

Athanasiadis, A<br />

P1‐13‐09<br />

Atkins, N<br />

P1‐13‐03<br />

Atkinson, RL<br />

P3‐10‐01<br />

ATLAS Collaborators<br />

Worldwide S1‐2<br />

Audretsch, W<br />

P1‐14‐04<br />

Auerbach, M<br />

P5‐20‐03<br />

Augereau, P<br />

P1‐14‐21<br />

Augustin, D P1‐13‐13, P2‐10‐08,<br />

P5‐14‐06, P5‐15‐04<br />

Auman, JT S6‐1<br />

Auricchio, N S5‐7<br />

Ausems, MG<br />

P4‐11‐01<br />

Autier, P<br />

P4‐13‐08<br />

Auzac, G<br />

OT1‐2‐02<br />

Avella, A<br />

P3‐12‐08<br />

Avent, JM<br />

P2‐10‐27<br />

Avery, TP<br />

P5‐14‐03<br />

Avery‐Kiejda, KA<br />

P4‐10‐02<br />

Avila, C<br />

P1‐07‐12<br />

Avila, CC<br />

P3‐08‐06<br />

Avila, E<br />

P4‐06‐19, P6‐04‐29<br />

Awad, D<br />

P2‐11‐03, P2‐12‐01<br />

Awada, A S6‐6<br />

Awolala, Y<br />

P5‐13‐03<br />

Axelrod, D P1‐09‐03, P2‐11‐12,<br />

P4‐14‐03, P4‐17‐07<br />

Ayala de la Peña, F<br />

P3‐06‐15<br />

Ayers, DA<br />

P2‐14‐04<br />

Ayers, GD<br />

P4‐01‐03<br />

Azim Jr, HA S4‐4, P6‐07‐14, P6‐07‐22<br />

Aziza, N<br />

P4‐12‐03<br />

Azizi, M<br />

P5‐17‐04<br />

Azizian, N<br />

P5‐04‐08<br />

Azuero, A<br />

P2‐12‐14<br />

Azuero, CB<br />

P2‐12‐14<br />

Baak, J<br />

P2‐10‐19<br />

Babiera, G PD07‐01, P1‐07‐06<br />

Babiera, GV<br />

P4‐15‐02<br />

Bacanu, F<br />

P5‐02‐02<br />

Bachelot, T P1‐13‐04, P2‐01‐13,<br />

P3‐12‐01, OT1‐1‐02, OT3‐4‐05<br />

Bacigalupo, S<br />

P2‐16‐03<br />

Bading, JR<br />

P2‐05‐10<br />

Badovinac Crnjevic, T P2‐13‐02<br />

Badve, S<br />

P1‐03‐02<br />

Badzio, A P3‐12‐09, P3‐13‐03<br />

Bae, SY<br />

P3‐11‐01<br />

Bae, YK<br />

P5‐04‐07<br />

Bae, Y‐K<br />

P1‐01‐05<br />

Baehner, FL S1‐10<br />

Baffert, S<br />

OT3‐3‐10,<br />

OT3‐4‐05, OT3‐4‐06<br />

Bagarre, T<br />

P6‐04‐14<br />

Bahjat, KS<br />

P4‐04‐02<br />

Bahrami, F<br />

P2‐12‐10<br />

Bai, L<br />

P6‐11‐11<br />

Baier, M<br />

PD07‐02<br />

Baildam, A<br />

P4‐14‐11<br />

Bak, M<br />

P2‐10‐28<br />

Baker, EL<br />

P3‐01‐06<br />

Baker, K<br />

P1‐04‐06<br />

Baker, RC<br />

P6‐04‐22<br />

Balchin, K<br />

P3‐11‐03<br />

Baldassarre, G<br />

P5‐10‐17<br />

Balinger, K<br />

P2‐06‐04<br />

Balko, JM S3‐6, PD01‐04, P6‐10‐05<br />

Ball, G<br />

P5‐10‐07, P6‐07‐18<br />

Ballan Haj, M<br />

P6‐14‐03<br />

Ballester, M<br />

P4‐14‐12<br />

Balleyguier, C<br />

P4‐12‐01<br />

www.aacrjournals.org 587s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Ballman, KV<br />

S5‐4, PD10‐04,<br />

P1‐01‐06, P6‐08‐05<br />

Balls, G<br />

P6‐07‐09<br />

Balmain, A<br />

P5‐05‐03<br />

Balmaña, J<br />

P6‐13‐03<br />

Balsari, A<br />

P5‐18‐10<br />

Balslev, E P3‐06‐03, P3‐06‐32<br />

Balzarini, A<br />

P2‐12‐08<br />

Bambina, S<br />

P4‐04‐02<br />

Ban, G<br />

P2‐13‐01<br />

Bando, H<br />

P1‐14‐02, P1‐14‐08<br />

Bandos, H<br />

OT2‐2‐04<br />

Bandyopadhyay, A P5‐03‐04, P5‐03‐06<br />

Bandyopadhyay, AM P5‐04‐04<br />

Bane, FT<br />

P6‐04‐04<br />

Banerjee, N<br />

PD05‐05,<br />

P4‐01‐02, P5‐07‐03<br />

Banerji, J S3‐3<br />

Baney, T<br />

P6‐11‐06<br />

Bang, B<br />

P4‐03‐11<br />

Banis, S<br />

P3‐06‐24<br />

Banzi, M<br />

PD05‐03<br />

Bao, L<br />

P2‐06‐01<br />

Bao, T<br />

P2‐13‐05<br />

Baquero, J<br />

OT3‐3‐05<br />

Bar, I<br />

P3‐04‐04<br />

Barakat, N<br />

PD02‐01<br />

Barakat, R<br />

P2‐12‐05<br />

Baranyai, Z<br />

P3‐05‐08<br />

Baranzelli, MC PD05‐08, P3‐14‐03<br />

Baravalle, S<br />

P2‐16‐03<br />

Barbareschi, M<br />

P5‐01‐10<br />

Barbeau, E<br />

PD08‐03<br />

Barber, P<br />

P2‐10‐29<br />

Barbieri, E<br />

P2‐10‐16<br />

Barbosa, EM<br />

P1‐01‐28<br />

Bardia, A<br />

P2‐01‐14<br />

Barentsz, MW P4‐14‐08, P5‐01‐11<br />

Bargiacchi, FG<br />

P6‐03‐01<br />

Barginear, M<br />

P5‐10‐13<br />

Barhamand, FB<br />

P1‐12‐04<br />

Barinoff, J<br />

S4‐5, PD06‐01<br />

Barker, J<br />

P4‐17‐03<br />

Barker, KB<br />

OT1‐1‐05<br />

Barlow, WE<br />

OT2‐2‐04<br />

Barnadas, A P1‐07‐18, P2‐01‐06<br />

Barnes, NLP<br />

PD04‐06<br />

Barnes, PJ<br />

P2‐10‐09<br />

Barnes, R<br />

P5‐07‐06<br />

Barnett, CM<br />

PD03‐08<br />

Barnholt, K<br />

OT3‐4‐04<br />

Baron, P<br />

OT3‐4‐03<br />

Barranger, E<br />

OT2‐1‐01<br />

Barrera, D P4‐06‐19, P6‐04‐29<br />

Barrett, J<br />

S4‐1, P2‐10‐29<br />

Barrett, JA<br />

P6‐11‐10<br />

Barrett, OC<br />

P5‐17‐07<br />

Barrett, S<br />

P4‐14‐13<br />

Barrett‐Lee, P S3‐3, S4‐1, P1‐13‐03<br />

Barrett‐Lee, PJ<br />

PD07‐09<br />

Barrios, C<br />

OT2‐2‐03<br />

Barrios, CH<br />

OT2‐3‐03<br />

Barron, RL<br />

P1‐15‐01<br />

Barry, P<br />

P1‐01‐10, P1‐01‐11<br />

Barry, W<br />

P3‐06‐07<br />

Barry, WT P2‐10‐01, P2‐10‐03,<br />

P4‐09‐01, P6‐08‐05<br />

Barsky, SH<br />

P6‐10‐04<br />

Bartlett, J<br />

S3‐3, P1‐13‐03<br />

Bartlett, JMS S1‐5, S4‐6, P1‐07‐17,<br />

OT3‐3‐09<br />

Barton, DL<br />

P2‐11‐01<br />

Barton, J<br />

P1‐14‐14, OT2‐3‐02<br />

Barton, Jr., J<br />

P5‐17‐05<br />

Bartsch, R<br />

P5‐20‐11<br />

Baselga, J S1‐11, S5‐1, S5‐2, S5‐7,<br />

P1‐07‐19, P2‐01‐14, P4‐08‐02,<br />

P5‐18‐01, P5‐18‐02, P5‐18‐26,<br />

P6‐04‐02, P6‐10‐07, OT2‐3‐06,<br />

OT2‐3‐09<br />

Baserga, R<br />

P6‐01‐03<br />

Bass, BL<br />

P6‐07‐11<br />

Basso, R<br />

P1‐01‐28<br />

Bastin, J<br />

P6‐09‐07<br />

Bates, N<br />

P5‐16‐04<br />

Batten, K<br />

P4‐06‐06<br />

Battista, M<br />

P2‐10‐13<br />

Batzoglou, S<br />

PD05‐09<br />

Bauer, JA<br />

P6‐05‐03<br />

Bauerfeind, I<br />

P1‐14‐01<br />

Bauerfeind, IGP S2‐2<br />

Baum, M S4‐2<br />

Baumgartner, A<br />

P4‐08‐06<br />

Bautista‐Pina, V<br />

P5‐10‐12<br />

Baxter, RC<br />

P5‐07‐05<br />

Bayer, RA<br />

P1‐12‐04<br />

Bayona, M<br />

P5‐07‐06<br />

Bazan, F<br />

P5‐18‐22<br />

Bazzola, L<br />

PD07‐03<br />

Beadling, C<br />

P2‐08‐03<br />

Bealin, L<br />

P6‐08‐01<br />

Beardslee, B<br />

P5‐20‐05<br />

Beatty, J<br />

OT3‐4‐03<br />

Becerra, C<br />

P6‐11‐08<br />

Becette, V<br />

PD07‐04<br />

Bechhold, R<br />

P6‐10‐01<br />

Beck, AH<br />

P5‐01‐03<br />

Beck, JT<br />

P6‐11‐08<br />

Beck, T<br />

P2‐10‐25<br />

Beckmann, MW<br />

PD07‐02,<br />

P2‐01‐02, P2‐10‐25<br />

Beckwith, H<br />

P4‐07‐03<br />

Becuwe, P<br />

P6‐01‐04<br />

Bedard, M<br />

P2‐11‐10<br />

Bedrosian, I<br />

P1‐07‐06,<br />

P1‐07‐09, P4‐15‐02<br />

Beekman, R<br />

P6‐04‐08<br />

Beelen, KJ PD01‐02, PD01‐03<br />

Beitsch, P P1‐15‐09, OT3‐4‐03<br />

Beitsch, PD<br />

P2‐10‐42,<br />

P4‐14‐01, P4‐16‐03<br />

Bekhash, A<br />

P5‐01‐07<br />

Belani, CP<br />

PD09‐06<br />

Belau, AK<br />

P1‐14‐05<br />

Belichard, C<br />

P1‐01‐07,<br />

P1‐01‐19, OT2‐1‐01<br />

Belin, L<br />

P5‐21‐03<br />

Belkacemi, Y<br />

P4‐16‐10,<br />

P5‐21‐01, OT1‐2‐02<br />

Bell, K<br />

P5‐01‐09<br />

Bell, R<br />

S5‐2, S6‐4,<br />

S6‐5, P3‐06‐34, P5‐18‐02<br />

Bellardini, P<br />

P1‐01‐25<br />

Bellavance, E<br />

P2‐13‐05<br />

Bellen, G<br />

P2‐12‐06<br />

Bellet, M<br />

P6‐13‐03<br />

Belletti, B<br />

P5‐10‐17<br />

Bellin, LS<br />

P1‐01‐17<br />

Bello, MA P3‐11‐04, P6‐07‐44<br />

Bellon, J<br />

P5‐02‐01<br />

Bellon, JR P3‐10‐06, P4‐06‐01<br />

Belmont, LD<br />

P4‐07‐01<br />

Belousov, A S6‐7<br />

Beltjens, F<br />

P3‐08‐07<br />

Belvin, M<br />

P4‐07‐01<br />

Ben Baruch, N<br />

P5‐20‐06<br />

Ben Younès, I<br />

P3‐12‐01<br />

Benaim, E<br />

P6‐10‐02<br />

Benca, J<br />

P2‐01‐12<br />

Bendahl, P‐O<br />

PD03‐07,<br />

P2‐10‐28, P2‐10‐36<br />

Bender, PFM<br />

P3‐11‐04<br />

Bendifallah, S<br />

P1‐01‐14<br />

Bengier, A<br />

P4‐13‐12<br />

Benke, Z<br />

P2‐10‐24<br />

Benko, L<br />

P1‐07‐17<br />

Benson, JR<br />

P1‐01‐01<br />

Benyunes, MC<br />

P5‐18‐26<br />

Benz, CC<br />

P2‐05‐04<br />

Benz, SC<br />

P2‐06‐05,<br />

P2‐06‐06, P4‐09‐05<br />

Berg, PE<br />

P2‐05‐22<br />

Bergan, R<br />

P1‐09‐07<br />

Berger, AC<br />

P5‐14‐03<br />

Berger, MF<br />

PD05‐02<br />

Berger, MJ<br />

P2‐11‐07<br />

Berger, N<br />

P3‐08‐08<br />

Bergh, J<br />

S1‐11,<br />

PD06‐08, OT2‐3‐02<br />

Bergkvist, L<br />

P2‐12‐09<br />

Bergmann, A P3‐11‐04, P6‐07‐44<br />

Bergstrom, K<br />

P2‐11‐16<br />

Berkowitz, A<br />

P2‐12‐05<br />

Berkowitz, AP<br />

P1‐13‐11<br />

Berlanda, G<br />

P5‐01‐10<br />

Berman, MG S6‐3<br />

Bernales, S<br />

P2‐14‐02<br />

Bernard, P P1‐08‐02, P6‐07‐10<br />

Bernards, R<br />

P4‐09‐05<br />

Bernreuter, W<br />

P3‐03‐03<br />

Berns, EMJJ<br />

PD01‐03,<br />

P3‐06‐01, P6‐04‐08<br />

Bernstein, L<br />

PD08‐02<br />

Bernstein, LJ P6‐09‐01, P6‐09‐02<br />

Berruti, A<br />

PD07‐03<br />

Berry, DL<br />

P1‐05‐12<br />

Berry, JS<br />

P5‐16‐05<br />

Bertaut, A<br />

P3‐08‐07<br />

Berteloot, P P2‐10‐12, P6‐05‐05,<br />

P6‐07‐29, OT2‐2‐02<br />

Bertheau, P<br />

P1‐14‐03<br />

Bertheau, P<br />

P3‐06‐04<br />

Bertolini, I<br />

P5‐17‐06<br />

Berton, S<br />

P5‐10‐17<br />

Bertschinger, J<br />

P5‐18‐25<br />

Bertucci, F<br />

P3‐05‐03,<br />

P5‐03‐01, OT2‐3‐05<br />

Beruti, S<br />

PD02‐01, PD06‐03<br />

Besancenot, V<br />

P6‐01‐04<br />

Beslija, S<br />

P5‐17‐03<br />

Beumer, JH<br />

PD09‐06<br />

Beurdeley, A<br />

P3‐06‐24<br />

Beutler, M<br />

P2‐10‐29<br />

Beuzeboc, P P5‐17‐04, P5‐21‐03<br />

Bevan, P<br />

P5‐20‐01<br />

Beveridge, R<br />

P2‐11‐16<br />

Bezu, C<br />

P1‐01‐14, P4‐14‐12<br />

Bhalla, KN S3‐7<br />

Bhanot, G<br />

P1‐05‐19<br />

Bhar, P<br />

P1‐12‐07<br />

Bhargava, R<br />

P6‐07‐02<br />

Bhaskar, P<br />

P4‐17‐09<br />

Bhat, RR<br />

P4‐06‐02<br />

Bhat, S<br />

P4‐06‐14, P4‐06‐16<br />

Bhatta, P<br />

P2‐13‐09<br />

Bi, L<br />

P5‐03‐14<br />

Bianca, R<br />

P5‐20‐03<br />

Bianchi, F<br />

P4‐06‐09<br />

Bianchini, G S6‐7, P2‐10‐10, P5‐10‐01<br />

Bickell, N<br />

PD08‐01<br />

Bidard, F‐C P2‐01‐01, P2‐01‐04,<br />

P2‐01‐13, OT3‐4‐05, OT3‐4‐06<br />

Bièche, I<br />

P6‐04‐14<br />

Biernat, W<br />

P2‐10‐31,<br />

P3‐12‐09, P3‐13‐03<br />

Bigorie, V<br />

P4‐16‐10<br />

Billig, J<br />

P4‐01‐07<br />

Bilusic, M<br />

P5‐16‐06<br />

Bingham, C<br />

PD05‐03<br />

Binghman, C<br />

PD05‐01<br />

Bingman, A<br />

P2‐11‐07<br />

Binkley, P<br />

P2‐11‐07<br />

Binns, SN<br />

P1‐07‐13<br />

Birch, KE<br />

P4‐13‐10<br />

Birnbaum, D<br />

P5‐03‐01<br />

Bischoff, E<br />

P6‐04‐12<br />

Bischoff, FZ<br />

P2‐01‐10<br />

Bischoff, J<br />

P1‐14‐05<br />

Bishop, JM<br />

P1‐04‐01<br />

Bissell, MJ<br />

P1‐05‐20<br />

Biswal, NC<br />

PD01‐01<br />

Biswas, T<br />

P2‐04‐07<br />

Bitting, R<br />

P4‐06‐07<br />

Bjarnadottir, O<br />

PD03‐07<br />

Bjarnason, G<br />

P2‐12‐03<br />

Bjerre, KD<br />

P3‐06‐03<br />

Black, DM S2‐3<br />

Black, EP<br />

P2‐14‐05<br />

Blackford, A S3‐5<br />

Blackwell, K<br />

OT2‐2‐03<br />

Blackwell, KL P6‐05‐02, OT1‐1‐11<br />

Blaes, AH<br />

Blair, SL<br />

Blancafort, A<br />

Blanc‐Fournier, C<br />

Blanco, E<br />

Blanco, EC<br />

Bland, K<br />

Bland, KI<br />

Blasberg, R<br />

Blaszczyk, P<br />

Blazeby, J<br />

Blazer, KR<br />

Blechman, K<br />

Bleicher, RJ<br />

Bleiker, EM<br />

Blenkinsop, C<br />

Blettner, M<br />

Blinder, VS<br />

Bliss, J<br />

Bliss, JM<br />

P1‐15‐01<br />

P4‐01‐13<br />

P4‐09‐10,<br />

P4‐09‐11, P6‐11‐02<br />

P3‐14‐03<br />

P6‐11‐03, P6‐11‐11<br />

P3‐02‐14<br />

P3‐03‐03<br />

P5‐17‐07<br />

P6‐01‐01<br />

P3‐12‐09, P3‐13‐03<br />

P4‐17‐03<br />

PD08‐06<br />

P4‐14‐03, P4‐17‐07<br />

P1‐01‐16<br />

P4‐11‐01<br />

OT3‐3‐09<br />

P6‐09‐08<br />

P2‐11‐05<br />

PD07‐07, P2‐14‐01<br />

S3‐3, S4‐1,<br />

P1‐13‐03, P6‐07‐15<br />

Blohmer, J S1‐11<br />

Blohmer, JU S4‐5<br />

Blohmer, J‐U S3‐1, PD06‐01, P1‐14‐01,<br />

OT1‐1‐13, OT3‐3‐11<br />

Blomme, C<br />

OT2‐2‐02<br />

Bloom, ES<br />

P4‐15‐02<br />

Bloom, K<br />

P3‐05‐05<br />

Bloom, KJ P2‐10‐04, P3‐05‐06<br />

Bloomfield, D<br />

S3‐3, P1‐13‐03<br />

Blosser, RJ<br />

P1‐03‐02<br />

Blot, E<br />

OT3‐3‐04<br />

Blowers, E<br />

P2‐14‐06<br />

Blows, FM<br />

P3‐08‐02<br />

Blue, MS<br />

P1‐01‐04<br />

Bluethmann, SM<br />

P5‐14‐02<br />

Blumencranz, P<br />

OT3‐4‐02<br />

Blumenthal, G S1‐11<br />

Boachie‐Adjei, K<br />

P3‐08‐08,<br />

P4‐15‐03, P6‐07‐34<br />

Boas, DA<br />

P3‐06‐27<br />

Bobos, M<br />

P1‐07‐15<br />

Bock, C<br />

PD03‐09<br />

Bode, H<br />

P6‐09‐07<br />

Bodmer, A<br />

P6‐04‐05<br />

Boe, M<br />

P3‐04‐01, P3‐04‐02<br />

Boehm, D<br />

P2‐10‐13<br />

Boelke, E<br />

P1‐14‐04<br />

Boemo, M<br />

P1‐05‐19<br />

Boer, K S1‐6<br />

Bogaerts, J<br />

S1‐11,<br />

P3‐02‐01, P3‐05‐02<br />

Bogart, JA<br />

P3‐12‐02<br />

Boggis, C<br />

P1‐09‐05<br />

Boggis, CR<br />

P4‐13‐09<br />

Böhme, M<br />

P1‐14‐05<br />

Böhm‐Vélez, M P4‐02‐09, P4‐02‐10<br />

Bohnsteen, B<br />

P5‐21‐02<br />

Bojar, H<br />

P1‐14‐04<br />

Bolan, P<br />

P3‐03‐01<br />

Bolan, PJ<br />

P1‐14‐11<br />

Bolaños, G<br />

P5‐07‐06<br />

Boley, KM P4‐06‐03, P5‐04‐05<br />

Bolger, JC<br />

P6‐04‐01<br />

Bollet, M<br />

P5‐02‐03<br />

Bolli, GB<br />

P4‐13‐07, P4‐13‐08<br />

Bolos, MV<br />

P6‐11‐02<br />

Bolten, WW<br />

P6‐09‐08<br />

Bona, E<br />

P5‐17‐06<br />

Bonanni, B<br />

PD03‐01,<br />

PD04‐07, PD06‐06<br />

Bonardi, S<br />

PD07‐03<br />

Bonat, J<br />

P5‐14‐03<br />

Bondarenko, IM S1‐6<br />

Bondarenko, IN S1‐4<br />

Bongers, V<br />

P4‐14‐08<br />

Boniol, M P4‐13‐07, P4‐13‐08<br />

Bonnefoi, H S1‐1, S1‐11, S5‐3<br />

Bonnefoi, HR<br />

OT3‐3‐10<br />

Bonnet, F<br />

PD05‐04<br />

Bonnetain, F<br />

OT3‐3‐10<br />

Bonneterre, J P1‐07‐11, P2‐13‐10<br />

Bono, P<br />

PD10‐06<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

588s<br />

<strong>Cancer</strong> Research


December 4-8, 2012<br />

Author Index<br />

Bontenbal, M<br />

P6‐07‐05<br />

Boogerd, W<br />

P1‐15‐05<br />

Boolbol, SK P3‐08‐08, P4‐15‐03,<br />

P6‐07‐34<br />

Booser, DJ<br />

P5‐20‐13<br />

Boothe, DL<br />

P4‐03‐09<br />

Boppart, SA<br />

OT2‐1‐04<br />

Bor, C<br />

P3‐05‐07<br />

Boraas, M<br />

P1‐01‐16<br />

Borakove, M<br />

P4‐04‐06<br />

Bordes, V<br />

OT2‐1‐01<br />

Borg, Å<br />

P2‐10‐37<br />

Borg, J‐P<br />

P3‐12‐01<br />

Borges, MM<br />

P3‐02‐14<br />

Borges, S<br />

P1‐05‐24<br />

Borges, V<br />

P4‐04‐03<br />

Borges, VF<br />

P4‐04‐06<br />

Borgquist, S PD03‐07, P2‐10‐28<br />

Borgstein, PJ<br />

P4‐11‐01<br />

Borm, GF<br />

P1‐14‐07,<br />

P5‐21‐04, P6‐07‐32<br />

Borms, M<br />

P5‐18‐19<br />

Borowsky, AD<br />

P1‐05‐20<br />

Børresen‐Dale, A‐L P2‐10‐20, P3‐04‐03,<br />

P3‐05‐04, P5‐05‐03, P5‐10‐03<br />

Bosc, R<br />

P4‐16‐10<br />

Bose, P<br />

P4‐09‐07<br />

Bose, R S5‐6<br />

Bosq, J<br />

P1‐07‐11<br />

Bosserman, L<br />

P2‐05‐06,<br />

P2‐11‐16, P3‐06‐28<br />

Bossuyt, V<br />

PD05‐05,<br />

P3‐14‐01, P4‐01‐02<br />

Bota, M<br />

P4‐13‐07, P4‐13‐08<br />

Bottai, G<br />

P5‐10‐01<br />

Botteri, E<br />

PD04‐07<br />

Bottini, A<br />

PD07‐03<br />

Boudreau, A<br />

P2‐05‐01,<br />

P4‐09‐02, P5‐05‐01<br />

Bouganim, N<br />

P2‐05‐13,<br />

P3‐13‐05, P5‐13‐01<br />

Boughey, JC S2‐1<br />

Boukovinas, I<br />

P1‐13‐09<br />

Bouma, WH<br />

P4‐11‐01<br />

Bourcier, C<br />

OT2‐1‐01<br />

Bourgeois, H P5‐21‐01, OT3‐3‐04<br />

Bourgier, C PD07‐04, OT1‐2‐02<br />

Bourrgier, C<br />

P5‐13‐02<br />

Bouton, M<br />

PD08‐04<br />

Bowden, NA<br />

P4‐10‐02<br />

Bowden, S S3‐3<br />

Boyages, J<br />

P4‐17‐04<br />

Boyer‐Chammard, A OT2‐3‐05<br />

Boyle, P<br />

P4‐13‐07, P4‐13‐08<br />

Boyle, T<br />

P6‐01‐02<br />

Brabencova, E<br />

P3‐14‐03<br />

Brachtel, E<br />

P2‐01‐14<br />

Brack, S<br />

P5‐18‐25<br />

Brackstone, M P1‐14‐13, P4‐16‐14<br />

Bradbury, AR<br />

P6‐08‐01<br />

Bradley, T<br />

P5‐10‐13<br />

Bradlow, HL<br />

P1‐10‐02<br />

Bradshaw, SH<br />

P5‐01‐01<br />

Braem, K<br />

P2‐13‐06<br />

Brafman, L P1‐09‐02, P2‐12‐01<br />

Brain, E P1‐13‐04, P1‐14‐03, P1‐14‐18,<br />

P2‐01‐04, P2‐01‐13, P3‐06‐04,<br />

P5‐18‐21, OT2‐3‐05, OT3‐3‐10,<br />

OT3‐4‐05, OT3‐4‐06<br />

Brammer, MG<br />

P5‐18‐12<br />

Bramwell, V<br />

P1‐13‐01<br />

Brandi, L<br />

P2‐07‐01<br />

Branle, F<br />

P5‐18‐05<br />

Brar, B<br />

P5‐12‐01<br />

Brase, JC<br />

S4‐3, P2‐10‐11<br />

Brauch, H<br />

P2‐13‐09<br />

Braun, A<br />

OT2‐3‐03<br />

Braun, HJ<br />

P1‐12‐05<br />

Breazna, A S1‐6<br />

Breitschwerdt, EB<br />

P3‐10‐03<br />

Brennan, DJ<br />

P4‐09‐06<br />

Brennan, K<br />

PD02‐07<br />

Breslin, T<br />

P1‐01‐13<br />

Breslin, TM<br />

P1‐13‐05<br />

Brewer, T<br />

PD03‐06, P3‐10‐05<br />

Brewer, TM<br />

PD03‐08<br />

Brew‐Graves, C S4‐2<br />

Brewster, AM<br />

P3‐10‐01<br />

Briggs, K<br />

P5‐18‐21<br />

Brigino, M<br />

P2‐05‐06<br />

Briley, LP<br />

PD10‐05<br />

Brindley, JH<br />

P3‐02‐04<br />

Brink, A<br />

PD07‐01<br />

Brinkman, JA<br />

P1‐05‐12<br />

Brinton, LA P3‐07‐04, P3‐08‐02<br />

Brito, M<br />

P1-15-03<br />

Brock, A<br />

P5‐05‐04<br />

Broderick, J<br />

P6‐08‐06<br />

Broderick, P<br />

P3‐08‐04<br />

Brodowicz, T<br />

P5‐17‐03<br />

Broers, JE<br />

P6‐05‐08<br />

Brogi, E<br />

OT3‐1‐01<br />

Bronson, RT<br />

P4‐08‐05<br />

Brookes, S<br />

P4‐17‐03<br />

Brooks, B<br />

P2‐11‐16<br />

Brooks, SE<br />

P4‐11‐06<br />

Brooks‐Brafman, L<br />

P2‐11‐03<br />

Brophy, ML<br />

P1‐05‐21<br />

Brouckaert, O P2‐10‐12, P5‐18‐19,<br />

P6‐05‐05, P6‐07‐04,<br />

P6‐07‐19, P6‐07‐29<br />

Brousseau, MK<br />

P5‐14‐04<br />

Brouste, V<br />

P3‐14‐03<br />

Brouwer, T<br />

P4‐11‐01<br />

Brown, D S4‐2, S6‐2, P3‐05‐03<br />

Brown, DN<br />

P3‐04‐10<br />

Brown, J<br />

S4‐1, S6‐5,<br />

PD01‐07<br />

Brown, JE S6‐4<br />

Brown, M PD01‐08, P1‐07‐07,<br />

P4‐02‐07, P6‐12‐03<br />

Brown, N<br />

P2‐10‐05<br />

Brown, RE<br />

P3‐06‐06<br />

Brown, S<br />

P4‐16‐09<br />

Brucker, C<br />

P5‐21‐02<br />

Brufsky, AM PD09‐06, P6‐07‐02<br />

Brugge, JS<br />

P4‐08‐05<br />

Brunet, J<br />

P4‐09‐11<br />

Brunner, A<br />

PD05‐09<br />

Brunner, E<br />

P6‐01‐04<br />

Brünner, N P3‐06‐32, P3‐06‐35<br />

Brunotte, F<br />

P4‐02‐06<br />

Brunt, M S3‐3<br />

Bubuteishvili‐Pacaud, L S6‐5, P3‐06‐34<br />

Buchan, I<br />

P3‐08‐01<br />

Buchholz, S<br />

P2‐12‐06<br />

Buchholz, TA<br />

S2‐1, S2‐3,<br />

PD03‐06, P5‐03‐05<br />

Buchmann, J<br />

P2‐10‐38<br />

Buclin, T<br />

P6‐04‐05<br />

Budach, W<br />

P1‐14‐04<br />

Budczies, J S4‐5<br />

Budd, GT<br />

PD02‐04,<br />

P2‐13‐02, P5‐18‐06<br />

Budd, T<br />

P6‐07‐45<br />

Budlovsky, J<br />

P4‐13‐04<br />

Budman, D<br />

P5‐10‐13<br />

Budrys, N<br />

PD03‐09<br />

Bueno, C<br />

OT3‐3‐05<br />

Buffa, FM P1‐06‐01, P4‐01‐01<br />

Bui, S<br />

PD02‐04<br />

Buist, DS<br />

P4‐01‐15<br />

Bulloch, KJ<br />

P2‐11‐08<br />

Bulsara, M S4‐2<br />

Bundred, N<br />

PD07‐07<br />

Bundred, NJ PD04‐06, P1‐01‐26,<br />

P3‐01‐04, P3‐14‐04<br />

Bundred, S<br />

P1‐09‐05<br />

Bundred, SM<br />

P3‐01‐04<br />

Buonomo, C<br />

P4‐15‐04<br />

Buonomo, OC<br />

P4‐15‐04<br />

Burgin, C<br />

PD04‐04<br />

Burnell, MJ<br />

P1‐13‐01<br />

Burnett, D<br />

P2‐11‐17<br />

Burris, H<br />

P6‐04‐02<br />

Burris, HA P1‐14‐14, P5‐17‐05,<br />

P6‐10‐07, OT3‐3‐08<br />

Burrows, J<br />

P1‐07‐19<br />

Burstein, H<br />

P5‐18‐03<br />

Busby, L<br />

P2‐11‐16<br />

Busch‐Devereaux, E P4‐14‐01<br />

Butler, S<br />

P6‐07‐03<br />

Butler, SM S1‐10<br />

Buxo, M<br />

P4‐09‐10<br />

Buyse, ME<br />

OT1‐1‐12<br />

Büyükberber, S<br />

OT1‐1‐08<br />

Byrd, DR S2‐1<br />

Byrne, C<br />

P6‐04‐01<br />

Caan, BJ<br />

P2‐11‐09<br />

Caanen, BA<br />

P6‐08‐02<br />

Caballero, R<br />

P2‐10‐11<br />

Cabaud, O<br />

P5‐03‐01<br />

Caggiano, V P3‐09‐01, P3‐09‐02<br />

Cagle, PD<br />

P6‐14‐04<br />

Caglevic, C<br />

P5‐18‐21<br />

Cagnolati, S<br />

OT1‐1‐08<br />

Cai, G<br />

P3‐12‐12<br />

Cai, J‐F<br />

P2‐05‐05<br />

Cai, X<br />

P1‐05‐21<br />

Caillet, P<br />

P4‐16‐10<br />

Cairo, S<br />

P3‐06‐24<br />

Caldara, A<br />

P5‐01‐10<br />

Caldarella, P<br />

PD06‐06<br />

Caldas, C<br />

P2‐10‐20,<br />

P3‐08‐05, OT3‐3‐09<br />

Caleffi, M<br />

P3‐01‐03<br />

Caley, MP<br />

P2‐02‐03<br />

Calhoun, E<br />

P5‐13‐03<br />

Calin, G<br />

P5‐10‐17<br />

Calin, GA<br />

P5‐10‐01<br />

Calitchi, E<br />

P4‐16‐10<br />

Callahan, R<br />

P2‐04‐08<br />

Callari, M<br />

P2‐10‐10<br />

Callihan, E<br />

P4‐04‐03<br />

Cals, L<br />

P5‐18‐22<br />

Calvert, V P3‐04‐01, P3‐04‐02<br />

Calvo, E<br />

P2‐06‐03<br />

Calvo, I<br />

P1‐14‐10, P5‐18‐15<br />

Calvo, L P1‐07‐18, P2‐01‐06, P2‐10‐11<br />

Caly, M<br />

P5‐01‐04<br />

Camacho, J P4‐06‐19, P6‐04‐29<br />

Camara, O<br />

P5‐03‐13<br />

Cameron, D S1‐11, S3‐3, S5‐2, S6‐4,<br />

S6‐5, P1‐13‐03,<br />

P2‐14‐01, P3‐06‐34, P5‐18‐02<br />

Cameron, DA OT1‐1‐03, OT3‐3‐09<br />

Cameron, S<br />

P6‐11‐06<br />

Camoin, L<br />

P3‐12‐01<br />

Campana, F<br />

P5‐21‐03<br />

Campbell, M<br />

P4‐04‐01<br />

Campbell, MM<br />

P1‐15‐06<br />

Campbell, P S6‐2<br />

Campbell, R<br />

P5‐13‐03<br />

Camphausen, K<br />

P6‐01‐03<br />

Campiglio, M<br />

P4‐06‐09<br />

Campone, M P1‐13‐04, P2‐01‐13,<br />

P3‐04‐04, P3‐05‐07, P3‐12‐01,<br />

P5‐18‐26, P6‐11‐08, OT2‐2‐03,<br />

OT2‐3‐09, OT3‐4‐05<br />

Candelaria, PV<br />

P4‐06‐18<br />

Canet, R<br />

P3‐12‐08<br />

Canisius, SV<br />

P4‐09‐05<br />

Cannas, P<br />

P1‐01‐25<br />

Canney, P<br />

S3‐3, P1‐13‐03<br />

Canon, JL<br />

OT2‐3‐02<br />

Canon, J‐L<br />

P3‐04‐04<br />

Cantos, B<br />

OT3‐3‐05<br />

Cao, X<br />

PD09‐08, P6‐04‐25<br />

Cao, Y<br />

P3‐10‐09<br />

Cappelletti, MR<br />

PD07‐03<br />

Capuano, LG<br />

P1‐01‐25<br />

Caraceni, A<br />

P2‐12‐08<br />

Carbaugh, H<br />

P2‐10‐05<br />

Cardoso, F P3‐02‐01, P3‐05‐02<br />

Carey, B<br />

P1‐15‐07<br />

Carey, L<br />

P6‐05‐02<br />

Carey, LA PD09‐03, PD10‐07<br />

Carillo, A<br />

P5‐13‐03<br />

Carkaci, S<br />

P1‐07‐06<br />

Carl, S<br />

P5‐03‐13<br />

Carlo‐Stella, C<br />

P2‐12‐08<br />

Carlson, JW<br />

PD06‐08<br />

Carlson, K<br />

P1‐15‐07<br />

Carmeliet, P<br />

P3‐06‐34<br />

Carmo, PAO<br />

P6‐07‐44<br />

Carneiro, JP<br />

P2‐12‐04<br />

Carney, PS<br />

OT2‐1‐04<br />

Carothers, S<br />

P2‐11‐07<br />

Carp, NZ<br />

P4‐09‐12<br />

Carp, SA<br />

P3‐06‐27<br />

Carpenter, J<br />

P3‐03‐03<br />

Carpenter, JT<br />

P5‐17‐07<br />

Carrasco, E<br />

S1‐7, P2‐10‐11<br />

Carrasco, J<br />

P3‐04‐04<br />

Carrasco, R<br />

P4‐07‐05<br />

Carrillo, JE<br />

PD08‐05<br />

Carrión, R<br />

OT3‐3‐05<br />

Carrion‐Salip, D<br />

P6‐11‐02<br />

Carroll, J<br />

P3‐02‐10<br />

Carroll, M<br />

P2‐05‐10<br />

Carroll, PA<br />

P6‐01‐02<br />

Carstensen, J<br />

P6‐07‐12<br />

Carter, CC<br />

P2‐10‐27<br />

Carter, CRD<br />

P1‐13‐08<br />

Carter, J<br />

P2‐12‐05<br />

Cartun, RW<br />

P1‐07‐10<br />

Cartwright, F<br />

P2‐11‐12<br />

Cartwright, T<br />

P2‐11‐16<br />

Carvalho, AA<br />

P5‐01‐15<br />

Carvalho, RM<br />

P6‐07‐44<br />

Casagrange, G<br />

P2‐04‐08<br />

Casalini, P<br />

P5‐18‐10<br />

Casas, M<br />

P2‐10‐11, P4‐09‐10<br />

Casbard, A<br />

PD07‐09<br />

Caseras, M<br />

P5‐13‐03<br />

Casey, L<br />

P3‐03‐01<br />

Caskey, M<br />

P6‐04‐22<br />

Casoliva, G<br />

P4‐09‐10<br />

Caspard, H<br />

P3‐07‐08<br />

Castañeda, C<br />

P5‐10‐11<br />

Castañon, E<br />

P5‐16‐03<br />

Castellano, DE<br />

P5‐10‐11<br />

Castiel, M<br />

P2‐12‐05<br />

Castillo, A<br />

P5‐16‐03<br />

Castro, C<br />

P3‐02‐08<br />

Catanzaro, J<br />

P5‐04‐09<br />

Catton, PA<br />

P6‐09‐01<br />

Caudle, AS<br />

P4‐15‐02<br />

Caudrelier, J‐M<br />

P4‐16‐06<br />

Cauley, JA<br />

PD03‐09<br />

Cavallin, F<br />

P3‐04‐04<br />

Cavedon, C<br />

P4‐16‐08<br />

Caveriviere, P<br />

P3‐05‐07<br />

Cavicchioli, F<br />

P4‐06‐10<br />

Cawthorn, S<br />

P4‐17‐03<br />

Cayre, A<br />

P2‐08‐04, P3‐06‐20<br />

Cazzaniga, M PD03‐01, PD06‐06<br />

Ceban, T<br />

P1‐14‐21<br />

Cermignani, L<br />

P3‐07‐13<br />

Cerussi, A<br />

P4‐02‐04<br />

Cesano, A<br />

P6‐07‐31<br />

Ceugnart, L<br />

P4‐14‐10<br />

Chacón, JI P1‐07‐18, P2‐01‐06<br />

Chadha, M P3‐08‐08, P4‐15‐03<br />

Chae, BJ<br />

P2‐05‐02, P4‐03‐01<br />

Chaffanet, M<br />

P5‐03‐01<br />

Chagpar, A<br />

P2‐11‐08<br />

Chagpar, AB<br />

PD04‐04,<br />

P3‐14‐05, P3‐14‐06<br />

Chaigneau, L<br />

P5‐18‐22<br />

Chaim, J<br />

P5‐20‐07<br />

Chaker, M<br />

P1‐15‐08<br />

Chakravarthy, AB<br />

P4‐01‐03<br />

Chakravarty, D<br />

P1‐05‐10<br />

Chalabi, N<br />

P3‐06‐20<br />

Chalchal, H<br />

P1‐13‐01<br />

Chaluvally‐Raghavan, P P5‐10‐03<br />

Chambers, A<br />

P1‐14‐13<br />

Chambers, M<br />

P2‐14‐05<br />

Chamness, G<br />

P6‐10‐03<br />

Chamness, GC S5‐8<br />

Champeny, TL<br />

P2‐11‐15<br />

Chan, A<br />

OT2‐3‐03<br />

Chan, E<br />

P5‐06‐02<br />

Chan, EK<br />

PD04‐02, P4‐16‐01<br />

Chan, HY<br />

P5‐11‐01<br />

www.aacrjournals.org 589s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Chan, K<br />

P6‐06‐06<br />

Chan, M<br />

P3‐08‐09<br />

Chan, SYT P6‐07‐09, P6‐07‐18<br />

Chandarlapaty, S PD04‐05, P4‐08‐02,<br />

P5‐18‐04, P5‐18‐20<br />

Chander, J<br />

P4‐02‐02<br />

Chandiramani, N<br />

P2‐04‐06<br />

Chandler, JC<br />

P6‐11‐10<br />

Chandran, UR<br />

P6‐04‐20<br />

Chang, B<br />

P1‐05‐21<br />

Chang, C‐P<br />

P6‐05‐15<br />

Chang, EY<br />

P4‐03‐01<br />

Chang, J<br />

P4‐03‐11<br />

Chang, JC<br />

S5‐8, P2‐10‐06,<br />

P3‐06‐14, P5‐03‐03, P5‐03‐12,<br />

P6‐07‐11, P6‐11‐12, OT3‐3‐01<br />

Chang, K<br />

P2‐12‐05<br />

Chang, K‐C<br />

P6‐11‐05<br />

Chang, K‐J P3‐06‐02, P4‐03‐02<br />

Chang, M<br />

PD10‐03<br />

Chang, MC<br />

PD03‐02,<br />

PD03‐05, P2‐10‐09<br />

Chang, P‐Y<br />

P4‐04‐07<br />

Chang, Y‐F P5‐10‐14, P5‐10‐16<br />

Chanock, SJ<br />

P3‐08‐02<br />

Chao, C<br />

P5‐15‐06<br />

Chao, J<br />

P2‐14‐08<br />

Chao, KC<br />

PD09‐09<br />

Chao, T‐C<br />

P3‐12‐10<br />

Chao, T‐Y<br />

P4‐04‐07<br />

Chaplain, MAJ P1‐05‐14, P5‐05‐02,<br />

P5‐05‐05, P5‐05‐06<br />

Chapman, J<br />

P1‐13‐01<br />

Chapman, J‐AW P1‐07‐13, P2‐13‐02<br />

Charaffe‐Jauffret, E P3‐12‐11,<br />

P5‐03‐01, OT2‐3‐05<br />

Charitansky, H<br />

P1‐01‐21<br />

Charon‐Barra, C<br />

P3‐08‐07<br />

Chateau‐Joubert, S<br />

P6‐04‐14<br />

Chatterjee, N<br />

P3‐10‐02<br />

Chatterton, RT P1‐09‐07, P2‐10‐30,<br />

P3‐04‐08, P4‐12‐04<br />

Chau, WW<br />

P3‐11‐02<br />

Chaudhary, S<br />

P1‐05‐13<br />

Chauhan, L S3‐7<br />

Chauvet, M‐P P4‐14‐10, P5‐21‐01<br />

Chaves Benito, A<br />

P3‐06‐15<br />

Chavez Mac Gregor, M P5‐20‐02<br />

Chavez‐Mac Gregor, M OT2‐2‐04<br />

Chavrier, P<br />

P5‐01‐04<br />

Chayanam, S PD01‐01, P4‐06‐02<br />

Cheang, MCU P4‐16‐02, P6‐07‐10<br />

Chebil, G<br />

P2‐10‐28, P2‐10‐36<br />

Cheema, K<br />

PD04‐06<br />

Chehayeb Makarem, D P2‐12‐01<br />

Che‐Lehman, J<br />

P1‐14‐18<br />

Chemaly, RF<br />

P4‐15‐02<br />

Chen, A<br />

P4‐03‐02, P4‐13‐05<br />

Chen, AC<br />

P6‐07‐11<br />

Chen, AP<br />

PD09‐06<br />

Chen, B S5‐4<br />

Chen, CM P3‐01‐05, P6‐04‐26<br />

Chen, C‐M<br />

P4‐16‐05<br />

Chen, C‐N<br />

P4‐03‐02<br />

Chen, E<br />

P5‐04‐09<br />

Chen, H<br />

P1‐05‐21, P1‐07‐06,<br />

P4‐07‐05, P6‐07‐25<br />

Chen, J P3‐06‐10, P3‐12‐12, P3‐12‐12<br />

Chen, JJ<br />

P4‐16‐12<br />

Chen, K<br />

P1‐01‐09<br />

Chen, KB<br />

P2‐11‐15<br />

Chen, K‐F<br />

P6‐11‐05<br />

Chen, KM<br />

P2‐09‐04<br />

Chen, L<br />

P2‐10‐26<br />

Chen, PY<br />

P4‐16‐03<br />

Chen, S P2‐14‐08, P6‐04‐11, P6‐07‐36<br />

Chen, S‐C<br />

OT1‐1‐16<br />

Chen, Y<br />

P1‐05‐09, P2‐05‐21,<br />

P3‐04‐06, P3‐09‐03, P5‐01‐07<br />

Chen, Z<br />

P5‐05‐01<br />

Chen, Z‐H P2‐05‐05, P4‐09‐09<br />

Chenevix‐Trench, G P6‐10‐06<br />

Cheng, A‐L P3‐06‐02, P6‐05‐15<br />

Cheng, H S5‐4<br />

Cheng, H‐CS<br />

P4‐16‐05<br />

Cherbavaz, DB S1‐10<br />

Chereau, E P1‐01‐14, P4‐14‐12<br />

Chernyukhin, N<br />

P5‐18‐06<br />

Cheung, KJ<br />

P1‐05‐02<br />

Chew, SA<br />

P6‐10‐03<br />

Chi, D<br />

PD01‐08<br />

Chia, S<br />

P2‐14‐01, P4‐16‐02,<br />

P4‐16‐04, P6‐11‐06<br />

Chia, V<br />

P1‐15‐01<br />

Chia, YL<br />

P3‐06‐31<br />

Chiarelli, AM<br />

P3‐02‐10<br />

Chiba, A<br />

P1‐01‐20<br />

Chien, R<br />

P5‐15‐06<br />

Chiesi, A<br />

P6‐11‐01<br />

Chilcott‐Burns, S<br />

P3‐08‐04<br />

Childs, J<br />

P2‐15‐02, P5‐16‐04<br />

Childs, JS<br />

OT3‐1‐02<br />

Chin, S‐F<br />

P3‐08‐05<br />

Chinchar, E<br />

P4‐06‐04<br />

Chirwa, T P1‐14‐14, P5‐20‐10<br />

Chitale, D<br />

P2‐09‐04<br />

Chiu, J‐H<br />

P3‐12‐10<br />

Chlebowski, R<br />

PD03‐09<br />

Cho, EY<br />

P2‐05‐20<br />

Cho, H<br />

P1‐15‐11<br />

Cho, JH<br />

P1‐01‐23, P2‐05‐03<br />

Cho, JY<br />

P3‐01‐02<br />

Cho, S‐H<br />

PD02‐06<br />

Choi, DS<br />

P5‐03‐03<br />

Choi, HH<br />

P1‐01‐24<br />

Choi, JE<br />

P5‐04‐07<br />

Choi, J‐E<br />

P1‐01‐05, P3‐03‐02<br />

Choi, M P1‐03‐02, P4‐14‐03, P4‐17‐07<br />

Choi, MR<br />

P1‐15‐01<br />

Choi, M‐Y<br />

P3‐11‐01<br />

Choi, Y<br />

P5‐04‐03<br />

Choi, Y‐L<br />

P2‐05‐02<br />

Chollet, P<br />

P3‐06‐20<br />

Choo, E<br />

PD04‐03<br />

Choridah, L<br />

P3‐01‐08<br />

Chow, C<br />

P3‐05‐05<br />

Chow, LWC P1‐14‐02, P2‐05‐16<br />

Chow, W<br />

P2‐14‐08<br />

Choy, NS<br />

P4‐09‐12<br />

Christaki, G P1‐01‐10, P1‐01‐11<br />

Christensen, AG<br />

P3‐04‐05<br />

Christensen, I<br />

P3‐06‐32<br />

Christensen, S<br />

P6‐07‐08<br />

Christgen, M<br />

P2‐10‐08<br />

Christiaens, M‐R P2‐10‐12, P6‐05‐05,<br />

P6‐07‐04, P6‐07‐19,<br />

P6‐07‐29, OT2‐2‐02<br />

Christiansen, J PD02‐01, PD06‐03<br />

Christianson, D<br />

P5‐20‐08<br />

Christie, A<br />

P1‐07‐04<br />

Christiny, PI<br />

P4‐06‐02<br />

Christman, LA<br />

P1‐01‐04<br />

Christopherson, C<br />

PD10‐03<br />

Christophylakis, C<br />

P1‐13‐09<br />

Chu, BC<br />

P2‐14‐07, P5‐20‐12<br />

Chu, E<br />

PD09‐06<br />

Chu, K P4‐06‐03, P5‐04‐05, P6‐10‐04<br />

Chu, N‐M<br />

P4‐16‐05<br />

Chu, P‐Y<br />

P6‐11‐05<br />

Chuang, E P1‐15‐07, P6‐11‐04<br />

Chuang, S‐C<br />

P4‐03‐02<br />

Chumsri, S<br />

P2‐13‐05<br />

Chun, J<br />

P1‐09‐03, P4‐01‐07<br />

Chung, C<br />

P2‐14‐08<br />

Chung, E<br />

P3‐12‐03<br />

Chung, F<br />

P6‐06‐06<br />

Chung, J<br />

P3‐07‐05<br />

Chung, JWL<br />

P3‐08‐06<br />

Chung, MS<br />

P2‐10‐39<br />

Church, TD<br />

P4‐01‐11<br />

Chyla, B<br />

OT2‐3‐07<br />

Cianni, R<br />

P1‐01‐25<br />

Ciciliano, JC<br />

P2‐01‐14<br />

Cigler, T<br />

P1‐15‐07,<br />

P6‐10‐01, P6‐11‐04<br />

Cimino‐Mathews, A P6‐07‐33<br />

Cimprich, B S6‐3<br />

Cinar‐Akakin, H<br />

P2‐11‐07<br />

Ciocca, RM<br />

P4‐09‐12<br />

Ciruelos, E P5‐18‐26, OT1‐1‐02,<br />

OT2‐3‐06, OT3‐3‐05<br />

Ciruelos, EM<br />

P5‐10‐11<br />

Citron, D<br />

P6‐10‐01<br />

Citron, ML<br />

P2‐10‐01<br />

Cittadine, AJ<br />

OT2‐1‐04<br />

Clague, J<br />

PD08‐06<br />

Clancy, R<br />

P1‐05‐09<br />

Clare, SE<br />

PD05‐06, P1‐03‐02<br />

Clarissa, A<br />

P3‐01‐03<br />

Clark, AS<br />

P3‐06‐10<br />

Clark, E S5‐1, P5‐18‐01, P5‐18‐26<br />

Clark, J<br />

P3‐04‐04<br />

Clark, L<br />

P4‐11‐04<br />

Clark, LS<br />

PD10‐07<br />

Clarke, C<br />

P3‐04‐12<br />

Classe, J‐M P1‐01‐21, OT2‐1‐01<br />

Clatot, F<br />

P1‐15‐08<br />

Cleator, S<br />

P3‐02‐04<br />

Clemens, M<br />

P2‐10‐08<br />

Clemente, G<br />

P3‐12‐08<br />

Clemons, M P2‐05‐12, P2‐05‐13,<br />

P2‐13‐02, P5‐13‐01<br />

Clemons, MJ<br />

P1‐15‐06,<br />

P3‐13‐05, P4‐16‐06<br />

Clifton, GT P5‐16‐02, P5‐16‐05<br />

Clisant, S<br />

P2‐13‐10<br />

Clough, KB<br />

P6‐07‐03<br />

Clynes, M<br />

P3‐04‐12<br />

Coarfa, C<br />

P6‐05‐12<br />

Coates, AS S1‐1<br />

Cobham, MV P1‐15‐07, P6‐11‐04<br />

Cobleigh, M<br />

P5‐18‐24<br />

Cobleigh, MA<br />

OT1‐2‐01<br />

Coburn, TL<br />

P6‐14‐04<br />

Cochrane, DR<br />

P2‐14‐02,<br />

P5‐10‐04, P5‐10‐06<br />

Codes, M<br />

P1‐07‐18, P2‐01‐06<br />

Cohen, EN P2‐03‐01, P2‐10‐32,<br />

P5‐04‐06, P5‐10‐02<br />

Cohen, HJ<br />

P6‐08‐05<br />

Cohen, J<br />

P6‐08‐06<br />

Cohen, P<br />

P1‐09‐04<br />

Colavito, S<br />

P3‐14‐01<br />

Colcher, D<br />

P2‐05‐10<br />

Coleman, R<br />

S3‐3, PD07‐09<br />

Coleman, RE<br />

S6‐4, P1‐13‐03,<br />

P2‐02‐04, OT2‐3‐03<br />

Collea, R<br />

P3‐06‐12<br />

Colleoni, M S1‐1<br />

Collin, B<br />

P4‐02‐06<br />

Collins, B<br />

P2‐11‐10<br />

Collins, IM<br />

P4‐13‐10<br />

Collins, J<br />

P5‐05‐04<br />

Collins, L<br />

P5‐01‐03<br />

Collins, R<br />

P1‐07‐04<br />

Colom, B<br />

P3‐12‐08<br />

Colon‐Otero, G S5‐5<br />

Come, S<br />

PD09‐03,<br />

P1‐07‐07, P2‐16‐04<br />

Comen, E<br />

P5‐18‐20<br />

Comstock, C<br />

OT3‐1‐01<br />

Conant, E<br />

P4‐01‐08<br />

Concannon, KF<br />

P2‐01‐14<br />

Condeelis, J<br />

P2‐10‐29<br />

Condeelis, JS<br />

P6‐02‐04<br />

Conklin, D<br />

P4‐08‐01<br />

Connolly, EM<br />

P6‐01‐02<br />

Connolly, EP PD09‐09, P5‐03‐02<br />

Connolly, J<br />

PD04‐04<br />

Connolly, RM P6‐07‐33, OT1‐1‐11<br />

Connor, CS<br />

PD09‐02<br />

Conte, P<br />

P2‐10‐16, P5‐18‐24<br />

Conter, D<br />

P5‐15‐02<br />

Conter, HJ<br />

P5‐15‐02<br />

Conti, P<br />

P2‐05‐10<br />

Contreras, A<br />

P2‐16‐02<br />

Cook, J<br />

P2‐10‐16<br />

Cook, RS<br />

P5‐19‐01, P6‐02‐01<br />

Coolen, A<br />

P2‐10‐29<br />

Coombes, CR<br />

P6‐04‐15<br />

Coombes, G PD07‐07, P2‐14‐01<br />

Coombes, RC P1‐04‐04, P2‐10‐22,<br />

P3‐06‐13, P4‐06‐12, P6‐04‐16<br />

Cooney, RV<br />

P4‐12‐02<br />

Cope, FO<br />

P1‐01‐04<br />

Cope, J<br />

P2‐14‐06<br />

Coppé, J‐P<br />

P5‐05‐01<br />

Cordingley, H<br />

P4‐06‐12<br />

Corica, T S4‐2<br />

Corless, CL<br />

P2‐08‐03<br />

Cornelius, C<br />

P4‐06‐06<br />

Cornell, JE<br />

P1‐05‐18<br />

Cornfeld, D<br />

P4‐01‐02<br />

Correa, RS P3‐01‐07, P3‐02‐13<br />

Cortazar, P S1‐11<br />

Cortes S6‐6<br />

Cortes, J S5‐1, P1‐07‐19, P5‐18‐26,<br />

P6‐11‐14, P6‐13‐03,<br />

OT2‐2‐03, OT2‐3‐09, OT3‐3‐06<br />

Cortés‐Funes, H P5‐10‐11, OT3‐3‐05<br />

Cortez, V<br />

P6‐04‐07<br />

Corwin, A P3‐05‐05, P3‐05‐06<br />

Cory, S<br />

P6‐02‐03<br />

Cosquer, M<br />

P4‐13‐14<br />

Costa, CRA<br />

P3‐11‐04<br />

Costa, L<br />

P3‐06‐33<br />

Costa, S‐D<br />

S3‐1, P1‐14‐01,<br />

OT1‐1‐13, OT3‐3‐11<br />

Costantino, J S1‐11<br />

Costantino, JP S1‐10<br />

Costes, S<br />

P1‐05‐08<br />

Cote, M<br />

PD03‐09<br />

Cottam, B<br />

P4‐04‐02<br />

Cottu, P<br />

PD05‐08, OT3‐3‐10<br />

Cottu, PH P2‐01‐13, P6‐04‐14<br />

Cottu, P‐H<br />

P5‐21‐03<br />

Coudert, B P1‐13‐04, P3‐08‐07<br />

Coupe, V<br />

PD04‐01<br />

Coupland, B<br />

P3‐08‐04<br />

Coussy, F<br />

P4‐13‐03<br />

Coutant, C P1‐01‐07, P1‐01‐14<br />

Couturaud, B<br />

P1‐01‐19<br />

Coveler, A<br />

P5‐16‐04<br />

Covington, K<br />

P4‐08‐09<br />

Cowens, JW<br />

P2‐10‐02<br />

Cox, C<br />

OT3‐4‐02<br />

Cox, D P1‐12‐02, P1‐13‐11, P5‐20‐04<br />

Cox, MB<br />

P5‐07‐02<br />

Cradock, KA<br />

OT2‐1‐04<br />

Crain, B<br />

P2‐06‐01<br />

Crawford, B<br />

P4‐13‐13<br />

Crawford, J<br />

P1‐15‐04<br />

Crawley, FL<br />

P1‐15‐06<br />

Creighton, C<br />

P4‐06‐02<br />

Creighton, CJ<br />

PD01‐01<br />

Crespo, C S1‐7, P1‐07‐18, P2‐01‐06,<br />

P2‐10‐11<br />

Crew, K<br />

P4‐02‐07<br />

Crew, KD PD03‐03, P1‐09‐02,<br />

P2‐11‐03, P2‐12‐01, P6‐12‐03<br />

Cristofanilli, M PD05‐01, PD05‐03,<br />

P2‐01‐01, P2‐03‐01, P2‐10‐32,<br />

P3‐05‐01, P3‐10‐03, P4‐06‐03,<br />

P5‐04‐05, P5‐10‐02, P6‐02‐05,<br />

P6‐10‐04, OT2‐3‐01<br />

Crittenden, MR<br />

P4‐04‐02<br />

Crivello, ML<br />

P1‐01‐16<br />

Croce, MV<br />

P3‐07‐13<br />

Crockett, M<br />

P3‐06‐12<br />

Croley, JJ<br />

P2‐14‐05<br />

Cromer, J<br />

OT1‐1‐14<br />

Cronin, M<br />

PD02‐07<br />

Cronin, MT S3‐6<br />

Crook, TR<br />

P4‐06‐10<br />

Cropet, C<br />

P3‐12‐01<br />

Croucher, PI<br />

PD07‐08<br />

Crow, J<br />

P1‐01‐06<br />

Crow, JR<br />

P1‐07‐09<br />

Crowley, E<br />

P6‐10‐01<br />

Crown, J<br />

P3‐04‐12<br />

Crown, JP S1‐6, P4‐09‐06, OT1‐1‐06<br />

Crozier, C S1‐5<br />

Cruz, M<br />

P4‐02‐03<br />

Csajka, C<br />

P6‐04‐05<br />

Cuenca, D P4‐06‐17, P6‐05‐04<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

590s<br />

<strong>Cancer</strong> Research


December 4-8, 2012<br />

Author Index<br />

Cui, H<br />

P3‐10‐07<br />

Cui, M<br />

P2‐05‐19<br />

Cukierman, E<br />

P6‐02‐05<br />

Culakova, E P1‐15‐04, P3‐06‐07<br />

Cung, Q<br />

P6‐01‐05<br />

Cunliffe, HE<br />

P1‐05‐04<br />

Cuorvo, LV<br />

P5‐01‐10<br />

Cupelo, EC<br />

P3‐12‐02<br />

Curcio, L<br />

P1‐15‐09<br />

Cure, H<br />

OT2‐3‐05<br />

Curley, C<br />

P6‐08‐03<br />

Custer, JL<br />

P2‐11‐15<br />

Cuvier, C<br />

P4‐13‐03<br />

Cuzick, J<br />

S1‐9, PD04‐08,<br />

P1‐09‐05, P2‐10‐15<br />

Czaplicki, KL<br />

P1‐12‐04<br />

Czene, K<br />

P2‐11‐06, P3‐07‐03<br />

Daasner, EJ<br />

P3‐10‐08<br />

Dabakuyo, S<br />

P3‐08‐07<br />

Dabbas, B PD02‐01, PD06‐03<br />

Dabbs, DJ P6‐04‐20, P6‐07‐02<br />

Dafni, U<br />

P5‐18‐02<br />

D’Agostino, Jr., R<br />

P2‐12‐11<br />

Dahl, M‐L<br />

PD10‐09<br />

Dahmane, E<br />

P6‐04‐05<br />

Dai, M‐S<br />

P4‐04‐07<br />

Dai, Y<br />

P2‐06‐01<br />

Dakir, DF<br />

P3‐01‐03<br />

Dale, DC<br />

P1‐15‐04<br />

Dale, R<br />

P4‐16‐09<br />

Dalenc, F<br />

P3‐12‐01, P5‐21‐01<br />

Daley, F<br />

P3‐06‐09<br />

D’Alfonso, TM<br />

P6‐02‐04<br />

Dall, B<br />

P4‐01‐14<br />

Dall, P<br />

P5‐21‐02<br />

Dallal, CM<br />

P3‐07‐04<br />

Dall’Oglio, S<br />

P4‐16‐08<br />

Daltoé, RD P5‐01‐15, P6‐11‐13<br />

Dalvi, T<br />

P3‐07‐08<br />

Daly, M<br />

P6‐08‐01<br />

D’Amato, NC P2‐14‐02, P5‐10‐06<br />

Danaei, M<br />

P1‐07‐16<br />

D’Andrea, G<br />

P5‐18‐20<br />

D’Andrea, S<br />

P5‐10‐17<br />

Dane, F<br />

OT1‐1‐08<br />

Danesi, R<br />

P5‐17‐06<br />

Dang, C P5‐17‐01, P5‐18‐20, P5‐20‐07<br />

Dang, Y<br />

P5‐16‐04<br />

Daniel, B<br />

P5‐20‐10<br />

Daniel, BL P4‐01‐06, P4‐01‐12<br />

Daniel, C<br />

P5‐17‐04<br />

Danso, MA<br />

OT1‐1‐07<br />

Dao, TH<br />

P4‐16‐10<br />

Darai, E<br />

P4‐14‐12<br />

Darb‐Esfahani, S<br />

P3‐06‐05<br />

Darer, J<br />

P4‐13‐12<br />

Darga, DA<br />

OT2‐1‐04<br />

Darimont, B<br />

P6‐04‐12<br />

Darnay, BG<br />

P5‐03‐09<br />

Darr, DB<br />

P6‐02‐09<br />

Darwish, T<br />

OT1‐1‐08<br />

Das, H<br />

P2‐11‐07<br />

Das, M<br />

P2‐11‐07<br />

Das Gupta, S S3‐7<br />

Dasgupta, N<br />

P2‐04‐02<br />

Datko, F<br />

P5‐18‐20<br />

Datko, FM P2‐08‐01, P6‐05‐01<br />

Datto, M<br />

P2‐10‐03<br />

Datto, MB<br />

P4‐09‐01<br />

Dauplat, M‐M<br />

P3‐06‐20<br />

Dave, B<br />

P5‐03‐12, P6‐02‐07<br />

David, AK<br />

OT1‐1‐07<br />

David, MA<br />

P6‐04‐03<br />

Davidson, C<br />

P6‐07‐24<br />

Davidson, D<br />

P3‐06‐28<br />

Davidson, N S5‐5<br />

Davidson, NE S5‐4<br />

Davies, C S1‐2<br />

Davies, S<br />

P3‐06‐09, P6‐07‐10<br />

Davies, SR<br />

P2‐10‐01<br />

Davis, EM<br />

P1‐05‐16<br />

Davis, S<br />

P2‐05‐01, P5‐20‐05<br />

Davis, TA<br />

P6‐10‐01<br />

Dawood, S<br />

P6‐07‐25<br />

Dayao, Z<br />

PD07‐01, P3‐03‐01<br />

De, G<br />

P5‐15‐05<br />

De, P<br />

P5‐19‐03, P6‐04‐11<br />

de Azambuja, E S5‐2, P5‐18‐02<br />

De Benedictis, E<br />

P2‐12‐08<br />

De Blaye, P<br />

P1‐01‐21<br />

De Brot, M<br />

PD04‐05,<br />

PD05‐02, P1‐06‐03<br />

De Buck, S<br />

P6‐10‐07<br />

De Carli, NL<br />

P5‐01‐10<br />

De Cecco, L<br />

P4‐06‐09<br />

de Cock, E<br />

P5‐15‐07<br />

de Cremoux, P P1‐14‐03, P2‐01‐04<br />

De Felice, V<br />

P4‐15‐04<br />

de Graaf, H<br />

P1‐12‐05<br />

de Grève, J<br />

P6‐09‐07<br />

de Groot, SM<br />

P1‐12‐05<br />

de Jongh, FE<br />

P1‐12‐05<br />

De Juán, A P1‐07‐18, P2‐01‐06<br />

de Koning, HJ<br />

P3‐02‐09<br />

De la Haba, JR S1‐7<br />

de la Peña, L<br />

OT3‐3‐06<br />

De la Piedra, C P1‐07‐18, P2‐01‐06<br />

De la Torre, R<br />

P1‐12‐04<br />

De Laere, B<br />

P2‐01‐09<br />

de Laurentiis, M<br />

OT2‐3‐09<br />

De Los <strong>San</strong>tos, J<br />

P3‐03‐03<br />

De Los <strong>San</strong>tos, JF P5‐17‐07, P6‐07‐37<br />

De Masi, C<br />

P1‐01‐25<br />

De Mattos, L<br />

P6‐13‐03<br />

De Mattos Aruda, L<br />

P5‐10‐01<br />

De Mukhopadhyay, K P1‐05‐18<br />

De Nicolo, A<br />

P4‐10‐01<br />

de Roos, MA<br />

P4‐11‐01<br />

de Roquancourt, A<br />

P4‐13‐03<br />

De Rycke, Y<br />

P1‐01‐07<br />

de Silvio, M<br />

P5‐18‐21<br />

de Snoo, F P2‐10‐42, P3‐05‐01,<br />

P3‐05‐02, P3‐06‐11, OT3‐4‐03<br />

de Velasco, GA<br />

P5‐10‐11<br />

de Vries, B P1‐14‐07, P5‐01‐13<br />

de Wilt, HHW OT2‐1‐02, OT2‐1‐03<br />

Deakin, NE<br />

P5‐05‐05<br />

Dean, M<br />

P4‐09‐07<br />

Deas, O<br />

P3‐06‐24<br />

Debeb, B<br />

PD03‐06<br />

Debeb, BG P5‐03‐05, P5‐03‐10<br />

Deblay, P<br />

OT2‐1‐01<br />

Debled, M<br />

PD05‐04<br />

Debska, S<br />

P2‐10‐31,<br />

P3‐12‐09, P3‐13‐03<br />

Decaudin, D<br />

P6‐04‐14<br />

DeCensi, A PD03‐01, PD06‐06<br />

Decker, T PD06‐04, PD06‐05,<br />

P2‐01‐02, P5‐01‐12<br />

Decloedt, J<br />

OT2‐2‐02<br />

Decoster, L<br />

P6‐09‐07<br />

Decosterd, L<br />

P6‐04‐05<br />

Dedeurwaerder, S S6‐2<br />

Dees, EC<br />

PD10‐07, P6‐11‐06<br />

Defrance, M S6‐2<br />

DeFreese, DC<br />

P4‐12‐05<br />

Degnim, AC P4‐12‐03, P5‐01‐08,<br />

P6‐06‐01<br />

Del Castillo, A<br />

PD04‐07<br />

Del Re, I<br />

P5‐17‐06<br />

Del Re, M<br />

P5‐17‐06<br />

Delabaye, I<br />

P3‐13‐01<br />

Delaloge, S P1‐14‐03, P1‐14‐18,<br />

P2‐01‐04, P2‐01‐13, P3‐06‐04,<br />

P4‐09‐05, P4‐12‐01, P5‐13‐02,<br />

OT3‐3‐04, OT3‐4‐05<br />

Delamelena, T<br />

P4‐11‐04<br />

Delattre, O<br />

PD05‐08<br />

Delbeuck, X<br />

P2‐13‐10<br />

Delecroix, V<br />

OT3‐3‐04<br />

DeLeo, III, MJ<br />

P4‐01‐08<br />

Delforge, M<br />

P3‐13‐01<br />

Delgado Sánchez, JJ P1‐01‐29<br />

Dell’Orto, P<br />

PD03‐01,<br />

P3‐05‐02, P6‐07‐14<br />

Delrée, P<br />

P3‐04‐04, P3‐05‐07<br />

Delva, R<br />

P1‐13‐04, P1‐14‐21<br />

Demanse, D<br />

P6‐10‐07<br />

DeMichele, A<br />

P3‐06‐10,<br />

P4‐13‐01, P6‐10‐02<br />

Demichelis, SO<br />

P3‐07‐13<br />

Denat, F<br />

P4‐02‐06<br />

Dendale, R<br />

P5‐21‐03<br />

Denduluri, N PD10‐08, P2‐11‐16,<br />

P4‐11‐04, OT2‐2‐03<br />

Deneuve, J<br />

P5‐13‐02<br />

Deng, G<br />

PD03‐04<br />

Denkert, C S3‐1, S4‐5, PD06‐01,<br />

P1‐14‐01, P3‐06‐05,<br />

OT1‐1‐13, OT3‐3‐11<br />

Dennison, JB<br />

P3‐06‐06<br />

Dent, R S6‐5, P2‐12‐03, P3‐06‐34<br />

Dent, S P3‐04‐11, P3‐11‐03, P3‐13‐05<br />

Dent, SF<br />

P1‐15‐06<br />

Deonarain, M<br />

P4‐06‐12<br />

Deray, G<br />

P5‐17‐04<br />

Dercksen, MW P5‐21‐04, P6‐07‐32<br />

Dercksen, W<br />

PD07‐06<br />

Desai, P<br />

PD03‐09<br />

DeSchryver, K P2‐10‐01, P6‐07‐10<br />

Deshpande, VS<br />

P4‐01‐10<br />

DeSilvio, M<br />

OT1‐1‐07<br />

Desmedt, C S4‐4, S6‐2, P3‐04‐10,<br />

P3‐05‐03, P6‐07‐14, P6‐07‐22<br />

Desrichard, A<br />

P3‐06‐20<br />

Dessen, P<br />

P4‐09‐05<br />

Detour, V<br />

P3‐04‐10<br />

Detre, S<br />

PD07‐07<br />

Deurloo, RJ<br />

P3‐06‐34<br />

DeVries, C<br />

P2‐11‐04<br />

Dewar, J<br />

S4‐1, PD03‐02<br />

Dewar, JA<br />

S4‐2, P5‐18‐05<br />

Dey, N<br />

P5‐19‐03, P6‐04‐11<br />

Deyarmin, B P3‐04‐07, P6‐02‐08<br />

Dezentjé, V<br />

OT2‐2‐02<br />

Dhakal, I<br />

P3‐06‐30<br />

Dharan, B<br />

OT2‐3‐08<br />

Dhuria, S<br />

P6‐11‐06<br />

Di, GH<br />

P3‐01‐05<br />

Di Cosimo, S<br />

OT3‐3‐06<br />

Di Legge, P<br />

P1‐01‐25<br />

Di Leo, A S1‐4, P5‐10‐01, OT2‐3‐08<br />

Di Pasquale, MC<br />

P5‐01‐10<br />

Di Tomaso, E<br />

P4‐08‐01<br />

di Tomaso, E<br />

P6‐11‐08<br />

Di Tomaso, E OT2‐3‐08, OT2‐3‐09<br />

Diab, A<br />

OT3‐1‐01<br />

Diamandis, EP P2‐05‐11, P6‐05‐09<br />

Diamond, JR<br />

P4‐04‐06<br />

Diana, C<br />

P4‐16‐10<br />

Diaz, L<br />

P4‐06‐19, P6‐04‐29<br />

Diaz‐Padilla, I<br />

P2‐10‐23<br />

Dickinson, M<br />

P4‐02‐08<br />

Dickler, M<br />

P2‐12‐05,<br />

P5‐18‐20, OT2‐3‐11<br />

Dickson, N<br />

P5‐17‐05<br />

Diederich, C<br />

P6‐13‐01<br />

Diener, M<br />

P5‐15‐05<br />

Dienstmann, R<br />

P6‐11‐06<br />

Diéras, V P2‐01‐13, P3‐12‐01,<br />

P5‐01‐04, P5‐18‐06<br />

Dietze, O S4‐3<br />

Dieudonné, A‐S<br />

OT2‐2‐02<br />

Dignan, M<br />

P5‐14‐05<br />

Dillenburg Pilla, P<br />

P1‐05‐23<br />

Dillon, P<br />

P3‐07‐11<br />

Dimitromanolakis, A P2‐05‐11,<br />

P6‐05‐09<br />

Dimitrova, N PD05‐05, P4‐01‐02<br />

Dimopoulos, N PD04‐06, P3‐14‐04<br />

Dinesh, A<br />

P4‐02‐02<br />

Ding, J<br />

P6‐04‐26<br />

Ding, L S5‐6<br />

Dinh, P<br />

P1‐07‐08<br />

Dinniwell, R<br />

P4‐16‐14<br />

DiPaola, J<br />

P2‐10‐05<br />

DiPiro, P<br />

P5‐02‐01<br />

DiRamio, A<br />

P2‐10‐05<br />

Dirix, L<br />

S1‐5, P5‐18‐19<br />

Dirix, LY P2‐01‐08, P2‐01‐09,<br />

P3‐10‐04, P3‐10‐08<br />

Disalvatore, L<br />

PD04‐07<br />

Disis, ML<br />

P2‐15‐02,<br />

P5‐16‐04, OT3‐1‐02<br />

Dissanayake, D<br />

P6‐09‐01<br />

Ditsch, N S1‐11<br />

Ditzel, H S4‐4<br />

Ditzel, HJ P3‐04‐05, P4‐09‐04<br />

Diver, J<br />

PD06‐03<br />

Divine, G<br />

P2‐09‐04<br />

Dixon, JM P3‐06‐23, P6‐04‐06,<br />

P6‐04‐09, P6‐04‐10<br />

Djazouli, K<br />

P6‐07‐22<br />

Dnistrian, A<br />

P2‐12‐05<br />

Dobbs, J S4‐1<br />

Dobi, E<br />

P5‐18‐22<br />

Dobrolecki, L<br />

P5‐03‐03<br />

Dodd, A<br />

P6‐09‐02<br />

Dodwell, D<br />

P2‐14‐01<br />

Dogan, A<br />

PD02‐05<br />

Doihara, H P3‐07‐10, P5‐18‐16<br />

Doimi, F S3‐6<br />

Domanitskaya, N<br />

P4‐08‐04<br />

Domchek, S P4‐01‐08, OT2‐3‐07<br />

Domínguez‐González, C OT3‐3‐05<br />

Donders, G<br />

P2‐12‐06<br />

Done, S<br />

P2‐10‐34<br />

Done, SJ PD03‐02, PD03‐05, P2‐10‐33<br />

Dong, J<br />

P6‐04‐19<br />

Dong, Q<br />

P5‐03‐04, P5‐03‐06<br />

Dong, X<br />

P6‐02‐05<br />

Dong, Y<br />

P5‐03‐14<br />

Donohue, S<br />

P2‐11‐15<br />

Donovan, D P1‐15‐07, P6‐11‐04<br />

Doppler, H<br />

P1‐05‐24<br />

Dorca, J<br />

P4‐09‐10, P4‐09‐11<br />

Dorchak, J<br />

P3‐04‐06<br />

Dorchak, JA<br />

P1‐05‐09<br />

Doren, EL P4‐14‐09, P4‐17‐01<br />

Dorent, R<br />

P5‐17‐04<br />

Doridot, V<br />

P1‐01‐07<br />

dos <strong>San</strong>tos, I<br />

P3‐08‐04<br />

Doughty, J<br />

P4‐14‐13<br />

Douglas, M<br />

P6‐10‐07<br />

Douglas, Y<br />

P4‐07‐03<br />

Dowling, RJO PD03‐02, PD03‐05<br />

Dowsett, M<br />

S1‐9, S4‐6, S5‐2,<br />

PD01‐05, PD07‐07, P1‐09‐05,<br />

P1‐13‐03, P2‐10‐15, P2‐14‐01,<br />

P3‐08‐04, P5‐18‐02, OT2‐3‐02<br />

Dowton, AA<br />

P4‐01‐09<br />

Doxey, DK<br />

P1‐03‐02<br />

Doyen, C<br />

P3‐13‐01<br />

Dozynkiewicz, M<br />

P1‐05‐13<br />

Drabble, EH<br />

P4‐14‐05<br />

Drabovich, AP<br />

P6‐05‐09<br />

Drake, C S1‐5<br />

Dranitsaris, G<br />

P3‐13‐05<br />

Dreher, C<br />

P4‐11‐06<br />

Drews‐Elger, K<br />

P1‐05‐12<br />

Driessen, WH<br />

P3‐10‐09<br />

Drobish, A<br />

PD10‐07<br />

Drobniene, M<br />

P5‐02‐02<br />

Drosick, DR<br />

P5‐17‐05<br />

Drukker, CA P2‐10‐42, P3‐02‐01<br />

Drullinsky, P P5‐18‐04, P5‐18‐20<br />

Drummond, D<br />

P4‐02‐05<br />

D’Souza, R<br />

P4‐13‐02<br />

Du, Y<br />

P5‐10‐15<br />

du Bois, A S3‐4<br />

Duan, X<br />

PD10‐02<br />

Dubois, T<br />

P5‐02‐03<br />

Dubsky, P S4‐3, P2‐10‐02, P2‐13‐01<br />

Duchamp, O<br />

P4‐02‐06<br />

Duchnowska, R P2‐10‐31, P3‐12‐09,<br />

P3‐13‐03<br />

Duck, L<br />

P3‐13‐01<br />

Dueck, AC<br />

PD10‐04<br />

Duerkop, J<br />

OT3‐2‐01<br />

Dufour, CR<br />

P5‐18‐08<br />

DuHadaway, JB<br />

P4‐09‐12<br />

Dumas, I<br />

OT1‐2‐02<br />

Dunbier, AK<br />

P4‐06‐11<br />

Dunn, B<br />

P1‐09‐07<br />

Dunn, JA OT1‐1‐03, OT3‐3‐09<br />

Dunn, L<br />

P2‐12‐05<br />

www.aacrjournals.org 591s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Dünne, A<br />

OT1‐1‐02<br />

Dunne, M<br />

P4‐02‐05<br />

Dünnebacke, J<br />

P2‐11‐11<br />

Dupiot‐Chiron, J<br />

PD05‐04<br />

Dupont, EL<br />

P4‐14‐01<br />

Dupré, P‐F P1‐01‐21, OT2‐1‐01<br />

Durack, J<br />

OT3‐1‐01<br />

Duran, H<br />

P1‐14‐10, P5‐18‐15<br />

Duran, J<br />

P3‐12‐08<br />

Dutcus, CE S6‐6<br />

Duval, V<br />

P6‐11‐08<br />

Duvall‐Noelle, NL<br />

P1‐05‐01<br />

Dweck, CS<br />

P6‐08‐10<br />

Dwyer, RM<br />

P2‐05‐09<br />

Eap, C<br />

P6‐04‐05<br />

Earashi, M<br />

P4‐15‐05<br />

Earl, H<br />

S3‐3, P1‐13‐03<br />

Earl, HM<br />

P3‐08‐05,<br />

OT1‐1‐03, OT3‐3‐09<br />

Earle, C<br />

P5‐18‐14<br />

Earp, HS<br />

P6‐03‐01<br />

Earp, S<br />

P6‐02‐01<br />

Eary, JF<br />

P6‐04‐03<br />

Eaton, AA<br />

P3‐07‐09<br />

Eaton, CL<br />

PD07‐08<br />

Ebata, A<br />

P6‐05‐14<br />

Eberhardt, S<br />

P3‐03‐01<br />

Eberle, CE<br />

P2‐11‐05<br />

Eberly, LE<br />

P1‐14‐11<br />

Eccher, C<br />

P5‐01‐10<br />

Eccles, D<br />

P1‐09‐05<br />

Echarri, M<br />

OT3‐3‐05<br />

Echenique, M<br />

P5‐07‐06<br />

Eckel‐Passow, JE<br />

PD10‐04<br />

Eckhardt, BL<br />

P3‐10‐09<br />

Edel, M<br />

P1‐13‐04<br />

Edge, S<br />

P1‐01‐13<br />

Edge, SB<br />

P1‐13‐05<br />

Edgerton, SM P5‐10‐04, P5‐10‐05<br />

Edgren, G<br />

P3‐07‐03<br />

Edirimanne, S P1‐11‐01, P4‐12‐07<br />

Edwards, D<br />

P6‐05‐12<br />

Edwards, J<br />

P1‐05‐13<br />

Eeles, R<br />

P1‐09‐05, P4‐11‐05<br />

Egbeare, DM<br />

P5‐11‐01<br />

Eggemann, H<br />

OT2‐2‐05<br />

Egland, KA<br />

P1‐05‐16<br />

Egleston, B P1‐05‐03, P6‐08‐01<br />

Egleston, BL P1‐01‐16, P2‐01‐01<br />

Eglitis, J<br />

P5‐02‐02<br />

Ehmsen, S<br />

P3‐04‐05<br />

Ehsani, S<br />

P3‐02‐11<br />

Eichner, LJ<br />

P5‐18‐08<br />

Eickhoff, JC<br />

P6‐12‐02<br />

Eidt, S PD10‐06, P3‐06‐08, P3‐06‐19<br />

Eidtmann, H S1‐11, S3‐1, S4‐5,<br />

P1‐14‐01, OT1‐1‐13, OT3‐3‐11<br />

Eiermann, W<br />

S1‐11, S4‐2,<br />

S4‐5, PD06‐01,<br />

Einbeigi, Z<br />

P1‐05‐07<br />

Einhorn, H<br />

P2‐05‐06<br />

Eisen, A<br />

P2‐12‐03, P3‐02‐10<br />

Eisenberg, M<br />

P2‐10‐16<br />

Ejlertsen, B<br />

S1‐1, P3‐06‐03<br />

Eklund, M<br />

P3‐02‐12<br />

El Mamlouk, T<br />

P1‐05‐11<br />

El Sharouni, MA<br />

P4‐01‐17<br />

El‐Ashry, D P1‐05‐12, P5‐10‐08<br />

Eldad, Z<br />

P1‐04‐05<br />

Eldon, MA<br />

P3‐06‐31<br />

Eleftheraki, AG<br />

P1‐07‐15<br />

ElGhonaimy, EA<br />

P1‐05‐11<br />

Elias, A<br />

P6‐05‐10<br />

Elias, AD<br />

P2‐14‐02, P4‐04‐06<br />

Elias, SG<br />

P2‐10‐42, P4‐09‐02<br />

Eliasson, E<br />

PD10‐09<br />

Elkahloun, AG<br />

P1‐05‐18<br />

El‐Khayat, Y<br />

P5‐12‐03<br />

Elkhuizen, PHM<br />

P4‐02‐01<br />

Elling, D<br />

P1‐14‐05<br />

Elliott, C<br />

P2‐13‐02<br />

Ellis, C<br />

OT1‐1‐07<br />

Ellis, IO<br />

P2‐10‐22, P3‐06‐13,<br />

P6‐07‐09, P6‐07‐18<br />

Ellis, M<br />

P2‐04‐02, P6‐07‐10<br />

Ellis, MJ S5‐6, PD07‐01, P1‐08‐01,<br />

P2‐10‐01, P2‐13‐02<br />

Ellis, P<br />

S3‐3, P1‐13‐03,<br />

P2‐10‐29, P2‐14‐01<br />

Ellisen, LW<br />

PD09‐03<br />

Ellison, PT<br />

P3‐01‐01<br />

Ellsworth, RE PD02‐02, P3‐04‐07,<br />

P6‐02‐08<br />

El‐Shinawi, M<br />

P1‐05‐11<br />

Elsoueidi, R<br />

P5‐14‐05<br />

El‐Zein, R<br />

P3‐10‐01<br />

Emami, H<br />

P2‐12‐10<br />

Emerson, BM<br />

P2‐06‐05<br />

Emi, A<br />

P6‐07‐26<br />

Endo, Y<br />

P4‐12‐06<br />

Engelen, K<br />

P3‐05‐02<br />

Engelman, JA<br />

P3‐12‐03<br />

Eng‐Wong, J<br />

P1‐09‐04<br />

Ennis, M<br />

PD03‐05<br />

Enomoto, T<br />

P2‐12‐13<br />

Enrech, S<br />

OT3‐3‐05<br />

Epperson, A<br />

P5‐20‐08<br />

Erbstoesser, E<br />

S4‐5, PD06‐01<br />

Erickson, J<br />

P1‐07‐05<br />

Erickson, KD<br />

P2‐11‐09<br />

Eriksson, N<br />

OT3‐4‐04<br />

Erlander, MG S1‐9, P1‐07‐13, P2‐10‐15<br />

Ersahin, C PD10‐02, P2‐05‐15<br />

Escallon, JM<br />

PD03‐05<br />

Eschenburg, H<br />

P1‐15‐02<br />

Escourrou, G<br />

P3‐05‐07<br />

Eslick, GD P1‐11‐01, P4‐12‐07<br />

Esmail, F<br />

P4‐14‐14<br />

Espié, M S5‐3, P1‐14‐03, P4‐13‐03<br />

Espina, V<br />

P3‐04‐01, P3‐04‐02<br />

Espinós, J P5‐03‐07, P5‐16‐03<br />

Espinosa‐Bravo, M<br />

P1‐01‐29<br />

Espinoza, I<br />

P6‐04‐22<br />

Espirito, J<br />

P2‐11‐16<br />

Esser, A<br />

P4‐15‐04<br />

Esserman, L<br />

S4‐2, P2‐05‐01,<br />

P2‐10‐06, P3‐04‐01,<br />

P3‐04‐02, P4‐04‐01, P4‐13‐13<br />

Esserman, LJ P2‐12‐11, P2‐12‐12,<br />

P3‐02‐01, P3‐02‐12, P3‐06‐10,<br />

P4‐14‐04, P4‐16‐07<br />

Estabrook, A<br />

P3‐08‐08<br />

Esterni, B<br />

OT2‐3‐05<br />

Esteva, F<br />

P5‐20‐02<br />

Esteves, S<br />

P1-15-03<br />

Estevez, LG<br />

P5‐18‐15<br />

Ettl, J S1‐6<br />

Ettore, F<br />

P3‐14‐03<br />

Ettyreddy, AR<br />

P4‐06‐07<br />

Euhus, D<br />

P1‐07‐04<br />

Eustermann, H S3‐4, P1‐15‐02<br />

Evans, A<br />

PD07‐07<br />

Evans, AC<br />

P2‐02‐04<br />

Evans, DGR<br />

P3‐08‐01<br />

Evans, G<br />

P1‐09‐05<br />

Evans, GD P4‐11‐05, P4‐13‐09<br />

Evensen, E<br />

P6‐07‐31<br />

Evron, E<br />

P5‐20‐06<br />

Ewald, AJ<br />

P1‐05‐02<br />

Ewing, CA P4‐14‐04, P4‐16‐07<br />

Exner, R<br />

P2‐13‐01<br />

Extra, J‐M<br />

OT2‐3‐05<br />

Fabian, CJ PD09‐02, P2‐11‐17<br />

Fabris, L<br />

P5‐10‐17<br />

Fackler, MJ P2‐02‐01, P4‐12‐04<br />

Fagerland, M<br />

P3‐01‐01<br />

Fahlén, MC<br />

P6‐14‐01<br />

Fais, S<br />

P6‐11‐01<br />

Faisal, A<br />

P3‐01‐08<br />

Fakhrejahani, E<br />

P1‐14‐02<br />

Falchook, G<br />

P5‐20‐09<br />

Falck, A‐K P2‐10‐28, P2‐10‐36<br />

Falcone, A<br />

P5‐17‐06<br />

Falkson, C<br />

P3‐03‐03<br />

Falkson, CI<br />

P5‐17‐07<br />

Fallowfield, L<br />

P1‐09‐05<br />

Falotico, N<br />

OT2‐3‐07<br />

Falzon, M S4‐2<br />

Fan, L<br />

P1‐13‐07<br />

Fan, Z P2‐05‐19, P5‐03‐14, P5‐10‐05<br />

Fanelli, G<br />

P1‐01‐25<br />

Fanelli, M<br />

P4‐02‐03<br />

Fang, L<br />

P5‐18‐11<br />

Faoro, L<br />

OT3‐4‐04<br />

Farajzadegan, Z<br />

P2‐12‐10<br />

Faria, C<br />

P6‐09‐06<br />

Faridi, JS<br />

P3‐12‐07<br />

Farley, J<br />

P4‐01‐03<br />

Farrar, C<br />

P3‐12‐03<br />

Farrar, JT<br />

P4‐13‐01<br />

Farre, I<br />

P4‐14‐10<br />

Fasano, J<br />

P5‐18‐20<br />

Fasching, P<br />

S1‐11, P3‐06‐05<br />

Fasching, PA PD07‐02, P1‐14‐01,<br />

OT1‐1‐10<br />

Fasciani, G<br />

P4‐06‐06<br />

Fasola, CE<br />

P4‐16‐11<br />

Fatayer, H<br />

P4‐01‐14<br />

Fatima, A<br />

P1‐07‐07<br />

Faure, C<br />

P1‐01‐21, OT2‐1‐01<br />

Favret, A<br />

P3‐06‐12<br />

Fazzio, RT<br />

P1‐01‐22<br />

Federico, V<br />

P3‐07‐06,<br />

P3‐07‐07, P3‐07‐14<br />

Fehm, T S2‐2, S4‐5, PD06‐01,<br />

P2‐01‐02, P6‐07‐05<br />

Fehm, TN<br />

OT1‐1‐10<br />

Fei, K<br />

PD08‐01<br />

Feigenberg, S<br />

P2‐13‐05<br />

Feigin, KN<br />

P6‐05‐02<br />

Feiten, S<br />

P2‐11‐11<br />

Feldman, S P1‐09‐02, P4‐02‐07<br />

Feldman, SM PD03‐03, P4‐14‐01<br />

Felzener, MM<br />

P1‐01‐28<br />

Feng, J‐g<br />

P4‐09‐09<br />

Fennessy, PJ<br />

P1‐01‐15<br />

Ferchiou, M<br />

P4‐12‐01<br />

Fergus, K<br />

P6‐08‐04<br />

Fermeaux, V<br />

P3‐14‐03<br />

Fernandes, L<br />

P2‐10‐29<br />

Fernandez, KL<br />

P2‐10‐40<br />

Fernandez, M<br />

P1‐14‐10<br />

Fernandez, ME<br />

PD08‐05<br />

Fernandez, SV<br />

P3‐10‐03<br />

Fernandez Hidalgo, OA P5‐03‐07,<br />

P5‐16‐03<br />

Fernández‐Zapico, M P5‐03‐07<br />

Fernando, A<br />

P4‐06‐14<br />

Fernö, M P1‐05‐07, P2‐10‐07,<br />

P2‐10‐19, P2‐10‐28, P2‐10‐36<br />

Fero, KE<br />

P6‐08‐10<br />

Feron, J‐G<br />

P1‐01‐07,<br />

P1‐01‐19, P5‐01‐04<br />

Ferrari, M P6‐11‐03, P6‐11‐11,<br />

P6‐11‐12, P6‐13‐02<br />

Ferrarini, I<br />

P5‐17‐06<br />

Ferrati, S<br />

P6‐11‐11<br />

Ferree, S<br />

P2‐10‐02<br />

Ferreira, RS<br />

P3‐01‐07<br />

Ferrero, J‐M<br />

P5‐18‐26<br />

Ferreyros, G<br />

P6‐07‐21<br />

Ferro, A<br />

P5‐01‐10<br />

Ferrusi, I<br />

P5‐18‐14<br />

Fersis, N<br />

P1‐14‐05<br />

Festa, A<br />

PD08‐03<br />

Festy, F<br />

P2‐10‐29<br />

Fettig‐Anderson, L<br />

P4‐07‐03<br />

Field, LA<br />

P3‐04‐07, P6‐02‐08<br />

Fields, SZ<br />

P6‐07‐31<br />

Figarella‐Branger, D P3‐12‐11<br />

Figueroa, JD<br />

P3‐08‐02<br />

Figueroa Magalhaes, MC P6‐07‐33<br />

Filho, EC<br />

P1‐01‐28<br />

Filipits, M<br />

S4‐3, P2‐10‐02<br />

Filipovic, A<br />

P1‐04‐04,<br />

P4‐06‐12, P6‐04‐15<br />

Filleron, T<br />

P3‐05‐07<br />

Finas, D<br />

P2‐10‐25<br />

Fineberg, S<br />

PD08‐01<br />

Finetti, p<br />

P5‐03‐01<br />

Fingert, H<br />

P6‐10‐02<br />

Finkelstein, D<br />

PD09‐03<br />

Finkelstein, DM P3‐06‐27, OT2‐3‐03<br />

Finkielstein, CV<br />

P6‐06‐05<br />

Fink‐Retter, A<br />

P1‐15‐10<br />

Finlay‐Schultz, J<br />

P6‐05‐10<br />

Finlayson, C P4‐04‐06, P6‐05‐10<br />

Finn, RS<br />

S1‐6, P4‐08‐01<br />

Finney, L P5‐17‐05, OT3‐3‐08<br />

Finstad, SE<br />

P3‐01‐01<br />

Finsterbusch, K PD06‐04, P5‐01‐12<br />

Firestone, B<br />

P6‐11‐06<br />

Fisch, K<br />

S4‐3, P2‐10‐11<br />

Fisch, M<br />

P2‐12‐14<br />

Fiskus, W S3‐7<br />

Fiteni, F<br />

P5‐18‐22<br />

Fitoussi, A<br />

P1‐01‐19<br />

Fittipaldi, P<br />

PD05‐03<br />

Fitzal, F<br />

P2‐10‐02, P2‐13‐01<br />

Fjällskog, M‐L<br />

P2‐10‐28<br />

Flågeng, MH<br />

P6‐04‐06<br />

Flamaing, J<br />

P6‐09‐07<br />

Flechsig, S<br />

P6‐04‐23<br />

Fleige, B S2‐2<br />

Fleisch, MC<br />

P2‐07‐01<br />

Fleming, G<br />

OT2‐2‐01<br />

Fleming, T<br />

P2‐04‐02<br />

Fletcher, M<br />

S3‐3, P1‐13‐03<br />

Fletcher, O<br />

P3‐08‐04<br />

Fletcher, S<br />

P3‐07‐05<br />

Fletterick, RJ<br />

P5‐07‐02<br />

Flexman, M<br />

P4‐02‐07<br />

Flinchum, R<br />

P2‐10‐05<br />

Flippo‐Morton, TS S2‐1<br />

Flockhart, D<br />

PD10‐08<br />

Flores, ER<br />

P5‐10‐03<br />

Floris, G P2‐10‐12, P6‐05‐05, P6‐07‐29<br />

Flory, K<br />

PD01‐07<br />

Flote, VG<br />

P3‐01‐01<br />

Flyger, H S4‐2<br />

Focke, CM<br />

PD06‐04,<br />

PD06‐05, P5‐01‐12<br />

Foekens, J<br />

P3‐06‐01,<br />

P3‐06‐32, P3‐06‐35<br />

Foekens, JA<br />

P6‐04‐08<br />

Fohlin, H<br />

P6‐07‐12<br />

Folkerd, E<br />

P3‐08‐04<br />

Fonseca, A‐V<br />

P2‐02‐03<br />

Fontaine, C<br />

P6‐09‐07<br />

Fontaine, J‐J<br />

P6‐04‐14<br />

Fontana, A<br />

P5‐17‐06<br />

Ford, JM PD09‐04, P3‐11‐02, P4‐11‐02<br />

Forero, A P3‐03‐03, P5‐17‐07,<br />

P6‐05‐02, P6‐10‐01, P6‐10‐05<br />

Formenti, S P5‐20‐05, P6‐13‐01<br />

Fornander, T P6‐07‐12, P6‐14‐01<br />

Fornier, M P1‐13‐11, P5‐18‐20<br />

Fornier, MN<br />

P5‐20‐07<br />

Foroni, C<br />

PD07‐03<br />

Forstbauer, H<br />

P2‐10‐25<br />

Forte, C<br />

P4‐14‐13<br />

Foster, K<br />

P1‐09‐07, P4‐08‐04<br />

Foszczynska‐Kloda, M P2‐10‐31,<br />

P3‐12‐09, P3‐13‐03<br />

Foucher‐Goudier, M‐J P1‐13‐04<br />

Fountzilas, G<br />

S5‐4, P1‐07‐15<br />

Fourchotte, V P1‐01‐07, P1‐01‐19<br />

Fournier, C<br />

P2‐13‐10<br />

Fourquet, A<br />

P5‐21‐03<br />

Fourrier‐Réglat, A<br />

P5‐21‐01<br />

Fowble, B<br />

P4‐16‐07<br />

Fowble, BL<br />

P4‐01‐13<br />

Fowles, AM<br />

PD07‐08<br />

Fox, EM<br />

PD01‐04<br />

Franchini, Z<br />

P4‐16‐08<br />

Francis, P<br />

OT2‐2‐01<br />

Franco, R<br />

PD08‐01<br />

Franco‐Barraza, J<br />

P6‐02‐05<br />

Frank, E<br />

P2‐16‐04<br />

Frank, RD P4‐12‐03, P5‐01‐08<br />

Franke, AA<br />

P4‐12‐02<br />

Frankel, PH<br />

P2‐14‐08<br />

Franklin, N<br />

OT1‐1‐04<br />

Fraternali, F<br />

P2‐10‐29<br />

Frazer, KA<br />

P2‐06‐01<br />

Frederiksen, K<br />

P1‐09‐01<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

592s<br />

<strong>Cancer</strong> Research


December 4-8, 2012<br />

Author Index<br />

Freedman, RA P6‐08‐05, OT1‐1‐11<br />

Freeman, JV<br />

P2‐02‐04<br />

Freemerman, AJ<br />

P6‐02‐09<br />

Freitas, NMA<br />

P2‐12‐04<br />

Freitas‐Junior, R<br />

P2‐12‐04,<br />

P3‐01‐07, P3‐02‐13<br />

Frenken, B<br />

P1‐07‐16<br />

Fresno, JA<br />

P5‐10‐11<br />

Fresquet, V<br />

P5‐03‐07<br />

Frieboes, HB<br />

P6‐11‐12<br />

Friedlander, M P4‐13‐10, OT2‐3‐07<br />

Friedman, L<br />

P5‐19‐02<br />

Friedman, LS P4‐07‐01, P6‐04‐11<br />

Friedman, PN<br />

P2‐10‐01<br />

Friedrichs, K<br />

P5‐21‐02<br />

Friese, K<br />

P2‐01‐02, P2‐01‐11<br />

Frigessi, A<br />

P3‐04‐03<br />

Frisell, J<br />

P3‐07‐03<br />

Fritel, X<br />

P1‐01‐07<br />

Fritzer, N<br />

P1‐15‐10<br />

Frost, M<br />

P2‐11‐01, P4‐12‐03<br />

Frost, MH P5‐01‐08, P6‐06‐01<br />

Fruhwirth, G<br />

P2‐10‐29<br />

Frydenberg, H<br />

P3‐01‐01<br />

Fryzek, J<br />

P3‐07‐08<br />

Fu, MR<br />

P2‐11‐12<br />

Fu, S<br />

P5‐20‐09<br />

Fu, X<br />

PD01‐01, P6‐10‐03<br />

Fuchs, E‐M P2‐10‐31, P3‐06‐33<br />

Fuentes, D P2‐11‐03, P2‐12‐01<br />

Fujibayashi, M<br />

OT1‐1‐01<br />

Fujii, H<br />

P4‐03‐07, P4‐15‐05<br />

Fujii, K<br />

P4‐03‐08<br />

Fujii, R<br />

P4‐03‐07<br />

Fujii, Y<br />

P4‐12‐06<br />

Fujimura, T<br />

P4‐16‐13<br />

Fujioka, H<br />

P3‐02‐02<br />

Fujisawa, T P1‐14‐08, P3‐02‐06<br />

Fujishima, F<br />

P6‐05‐14<br />

Fujita, T<br />

P1‐02‐02, P3‐12‐06<br />

Fujiwara, T<br />

P4‐07‐04<br />

Fujiwara, Y P2‐11‐02, P5‐18‐16<br />

Fuks, F S6‐2<br />

Fukumura, D<br />

P3‐12‐03<br />

Fukutomi, T<br />

P4‐03‐08<br />

Fulp, W<br />

P4‐14‐09<br />

Fulp, WJ<br />

P4‐17‐01<br />

Fumagalli, D S6‐2, P1‐07‐08, P3‐04‐10,<br />

P3‐05‐03, P6‐07‐22<br />

Fumoleau, P<br />

S5‐3, P3‐08‐07,<br />

P4‐02‐06, P5‐18‐05, P6‐11‐08<br />

Funian, L<br />

P2‐10‐26<br />

Fuqua, S<br />

P4‐08‐09<br />

Furberg, A‐S<br />

P3‐01‐01<br />

Furhmann, L<br />

P5‐01‐04<br />

Furukawa, H<br />

P4‐16‐13<br />

Futch, K<br />

P5‐18‐09<br />

Fyles, A<br />

P2‐10‐33<br />

Gabbani, M<br />

P4‐16‐08<br />

Gabram, S<br />

P1‐01‐06<br />

Gabrielson, EW<br />

OT2‐1‐04<br />

Gacquer, D<br />

P3‐04‐10<br />

Gaddy, D<br />

P4‐02‐05<br />

Gade, S<br />

S3‐1, P3‐06‐05<br />

Gaffney, D<br />

P5‐01‐09<br />

Gajria, D P5‐18‐04, P5‐18‐20, P6‐11‐10<br />

Gal, J<br />

P4‐15‐01<br />

Galanko, JA<br />

P6‐02‐09<br />

Galant, C<br />

P3‐05‐03<br />

Gallagher, A<br />

P1‐09‐04<br />

Gallagher, RI P3‐04‐01, P3‐04‐02<br />

Gallagher, W<br />

P4‐09‐06<br />

Gallagher, WM<br />

P3‐04‐12<br />

Galligioni, E<br />

P5‐01‐10<br />

Gallion, K<br />

PD08‐05<br />

Galmiche, M<br />

P6‐04‐05<br />

Gamez, A<br />

P5‐10‐11<br />

Gampenrieder, SP<br />

P5‐20‐11<br />

Gandhi, S<br />

P4‐16‐14<br />

Gandini, S<br />

PD06‐06<br />

Ganesan, S<br />

P1‐05‐19<br />

Gang, N<br />

P2‐10‐26<br />

Ganju, RK<br />

P1‐05‐17<br />

Gany, F<br />

P2‐11‐05<br />

Ganz, PA<br />

OT2‐2‐04<br />

Gao, H P2‐03‐01, P5‐03‐04, P5‐03‐06,<br />

P5‐04‐06, P5‐10‐02<br />

Gao, J<br />

P5‐10‐10<br />

Gao, M<br />

P6‐05‐15<br />

Gao, S<br />

P4‐03‐12<br />

Gao, X<br />

P6‐09‐06<br />

Gao, Y<br />

P5‐18‐24<br />

Garbay, J‐R<br />

P4‐14‐12<br />

Garber, J<br />

PD01‐08, OT2‐3‐07<br />

Garber, JE<br />

PD09‐03<br />

García García, T<br />

P3‐06‐15<br />

García Garre, E<br />

P3‐06‐15<br />

García, J<br />

P4‐06‐19<br />

García, R<br />

P4‐06‐19<br />

García Saenz, JA<br />

OT3‐3‐05<br />

García‐Martínez, E<br />

P3‐06‐15<br />

García‐Sáenz, JÁ<br />

P5‐18‐05<br />

Garces, Z<br />

P1‐07‐06<br />

Garcia, A<br />

P6‐10‐01<br />

Garcia, E<br />

P5‐18‐15<br />

Garcia, J<br />

P6‐04‐29<br />

Garcia, R<br />

P6‐04‐29<br />

Garcia Blanco, MA<br />

P4‐06‐07<br />

Garcia‐Aranda, M<br />

P1‐14‐10<br />

Garcia‐Closas, M<br />

P3‐08‐02<br />

Garcia‐Estevez, L<br />

P1‐14‐10<br />

Garcias, C<br />

P3‐12‐08<br />

Garcia‐Saenz, JA S1‐7<br />

Garee, JP<br />

P6‐05‐07<br />

Garey, J<br />

P2‐11‐16<br />

Gargi, P<br />

P2‐10‐29<br />

Garnett, S S1‐4<br />

Garofalo, A<br />

OT1‐1‐07<br />

Garrett, M<br />

P2‐16‐04<br />

Garwood, M<br />

P1‐14‐11<br />

Gasperetti, F<br />

P5‐01‐10<br />

Gates‐Ferris, K P3‐02‐07, P4‐13‐02,<br />

P5‐14‐02<br />

Gattelli, A<br />

PD01‐06<br />

Gatti‐Mays, M<br />

P1‐09‐04<br />

Gaule, P<br />

P3‐04‐12<br />

Gaur, R<br />

P4‐06‐14<br />

Gauthier, A<br />

PD05‐08<br />

Gauthier, G<br />

P5‐18‐12<br />

Gavilá, J P1‐07‐18, P2‐01‐06, OT1‐1‐09,<br />

OT2‐3‐06<br />

Gavin, P<br />

P6‐05‐12<br />

Gaynor, E<br />

P1‐12‐04<br />

Gaynor, ER<br />

PD10‐02<br />

GBG (German <strong>Breast</strong> Group) S1‐7<br />

Gebrim, LH<br />

P2‐05‐17<br />

Gehrmann, M<br />

P2‐10‐13<br />

GEICAM (Spanish <strong>Breast</strong> <strong>Cancer</strong><br />

Research Group) S1‐7<br />

Geiger, XJ<br />

P1‐05‐24<br />

Geisler, J<br />

P6‐04‐06<br />

Gelber, RD S1‐1, S3‐2, S5‐2, P5‐18‐02,<br />

P6‐07‐06, OT2‐2‐01<br />

Gelber, S<br />

S3‐2, P6‐07‐06<br />

Gelman, RS<br />

OT1‐1‐11<br />

Gelmon, K<br />

P1‐13‐01,<br />

P2‐13‐02, P4‐16‐02<br />

Generali, D<br />

PD07‐03<br />

Génin, P<br />

P3‐05‐07<br />

Gennari, MB<br />

P3‐02‐14<br />

Gentilini, OD<br />

PD03‐01<br />

Geoghegan, C<br />

P6‐08‐09<br />

Georgoulia, NE<br />

P6‐06‐02<br />

Georgoulias, V<br />

P1‐13‐09,<br />

P2‐01‐03, P2‐01‐07<br />

Geradts, J<br />

P2‐10‐03,<br />

P3‐06‐07, P4‐09‐01<br />

Gerber, B<br />

S1‐11, S3‐1, S4‐5,<br />

PD06‐01, P1‐14‐01,<br />

OT1‐1‐13, OT3‐3‐11<br />

Gerdes, M<br />

P3‐05‐05<br />

Geretti, E<br />

P1‐07‐03<br />

Germa, C OT2‐3‐08, OT2‐3‐09<br />

Gerritse, FL<br />

P6‐04‐08<br />

Gertler, FB<br />

P6‐02‐04<br />

Gevaert, O<br />

P2‐09‐03<br />

Geyer, C S1‐11<br />

Geyer, CE<br />

P6‐07‐06<br />

Geyer, Jr., CE S3‐2, S5‐5<br />

Ghaffarpour, M<br />

P5‐09‐01<br />

Ghazoui, Z<br />

PD01‐05<br />

Ghimbovschi, S<br />

P2‐05‐22<br />

Ghirelli, C<br />

P4‐06‐09<br />

Ghono, T<br />

P6‐04‐17<br />

Ghosh, K P1‐01‐22, P4‐12‐03, P5‐01‐08<br />

Ghosh, S<br />

P6‐02‐02<br />

Ghuijs, PM<br />

P5‐01‐13<br />

Giacchetti, S P1‐14‐03, P1‐14‐18,<br />

P2‐01‐04, P3‐06‐04, P4‐13‐03<br />

Giacomi, N<br />

P3‐07‐13<br />

Giamas, G<br />

P1‐04‐04,<br />

P4‐06‐12, P6‐04‐15<br />

Gianni, AM<br />

P2‐12‐08<br />

Gianni, L<br />

S1‐11, S5‐2, S6‐7,<br />

P2‐10‐10, P2‐12‐08, P5‐18‐02<br />

Giard, S<br />

P4‐14‐10<br />

Gibbs, IC<br />

P4‐16‐11<br />

Gibson, J<br />

P4‐06‐04,<br />

OT3‐4‐02, OT3‐4‐03<br />

Gibson, L<br />

P3‐08‐04<br />

Gidekel, R<br />

OT1‐1‐08<br />

Gielen, M<br />

P6‐08‐02<br />

Gierach, GL<br />

P3‐07‐04<br />

Giermek, J<br />

P6‐07‐05<br />

Giguère, V<br />

P5‐18‐08<br />

Gil, M S1‐7<br />

Gilcrease, M P5‐03‐05, P5‐20‐09<br />

Gilewski, T P5‐18‐20, P5‐20‐07<br />

Gilks, B<br />

P2‐10‐09<br />

Gillego, A<br />

P3‐08‐08<br />

Gilles, E<br />

P1‐07‐11<br />

Gillett, C<br />

P2‐10‐29<br />

Gilman, PB<br />

P4‐09‐12<br />

Gil‐Martin, M<br />

P6‐11‐08<br />

Gilmer, TM<br />

P4‐08‐05<br />

Gimbergues, P<br />

OT2‐1‐01<br />

Ginestier, C<br />

P5‐03‐01<br />

Ginsburg, GS P2‐10‐03, P3‐06‐07<br />

Ginsburg‐Arlen, A<br />

P2‐11‐16<br />

Ginter, PS<br />

P6‐02‐04<br />

Ginty, F<br />

P3‐05‐05<br />

Giobbie‐Hurder, A S1‐1, S4‐4<br />

Giordano, A P2‐01‐01, P2‐03‐01,<br />

P5‐04‐06, P5‐10‐02<br />

Giordano, SH P5‐20‐02, P5‐20‐13<br />

Giorgetti, C<br />

OT2‐3‐02<br />

Giovanni, C<br />

PD04‐05<br />

Giranda, V<br />

PD09‐06<br />

Giranda, VL<br />

OT2‐3‐07<br />

Giri, D<br />

PD04‐05<br />

Giri, DD<br />

PD05‐02, P6‐05‐02<br />

Girish, S<br />

P5‐18‐11, P5‐18‐24<br />

Giro, A<br />

P4‐09‐11, P6‐11‐02<br />

Girre, V<br />

OT3‐3‐10<br />

Gittleman, M<br />

OT3‐4‐03<br />

Giuliano, AE<br />

P1‐01‐06<br />

Giuliano, M S5‐8<br />

Glas, AM<br />

P4‐09‐05<br />

Gläser, D<br />

PD06‐04, P5‐01‐12<br />

Glassey, E<br />

P1‐01‐26<br />

Glazebrook, KN<br />

P1‐01‐22<br />

Glencer, A P2‐12‐11, P2‐12‐12<br />

Gligorov, J<br />

P3‐06‐20,<br />

P5‐17‐04, P5‐21‐01<br />

Glück, S P1‐12‐02, P3‐06‐11, OT3‐4‐02<br />

Gluz, O<br />

P2‐10‐08, OT3‐3‐02<br />

Gnant, M<br />

S4‐3, P2‐10‐02,<br />

P2‐13‐01, P6‐04‐02<br />

Gobardhan, PD<br />

P4‐14‐08<br />

Gobbi, H<br />

P1‐06‐03<br />

Godellas, C PD10‐02, P1‐02‐01<br />

Godin, B<br />

P6‐02‐07<br />

Godwin, J S1‐2<br />

Goehler, T<br />

P1‐15‐02<br />

Goel, S<br />

P3‐12‐03<br />

Goes, JCS<br />

P1‐01‐28<br />

Goetz, MP<br />

PD10‐08<br />

Goga, A S3‐8<br />

Goicochea, L<br />

P2‐10‐40<br />

Gojis, O<br />

P2‐10‐22<br />

Goka, E<br />

P1‐04‐06<br />

Goldberg, JD<br />

P5‐20‐05<br />

Goldberg, JM<br />

P4‐11‐06<br />

Goldfarb, S<br />

P5‐18‐20<br />

Goldfarb, SB<br />

P2‐12‐05<br />

Goldhirsch, A<br />

S1‐1, S5‐2,<br />

P5‐18‐02, P6‐07‐14<br />

Goldstein, LC<br />

P1‐07‐01<br />

Goldstein, LJ S3‐5, P5‐20‐01, OT2‐3‐01<br />

Goldwasser, F<br />

P5‐17‐04<br />

Golemis, EA<br />

P1‐05‐03<br />

Goloubeva, O<br />

P2‐13‐05<br />

Gomes, DO<br />

P6‐07‐44<br />

Gómez, F<br />

P2‐14‐02<br />

Gomez, H S3‐6<br />

Gómez, HL P6‐07‐20, P6‐07‐21<br />

Gómez, P<br />

P1‐07‐18,<br />

P2‐01‐06, P6‐13‐03<br />

GomezGarcia, EB<br />

P6‐08‐02<br />

Gonçalves, A<br />

P3‐12‐01,<br />

P3‐12‐11, OT2‐3‐05<br />

Gondo, N<br />

P1‐02‐02<br />

Gondou, N<br />

P3‐12‐06<br />

Gong, G<br />

P1‐03‐03, P2‐02‐02,<br />

P3‐12‐05, P6‐07‐28<br />

Gong, G‐y<br />

P3‐01‐02<br />

Gonugunta, VK P5‐04‐04, P6‐04‐07<br />

Gonzalez, J<br />

P5‐07‐06,<br />

P6‐05‐02, P6‐11‐10<br />

Gonzalez, ME<br />

P1‐04‐11<br />

Gonzalez‐Angulo, AM P1‐07‐06,<br />

P3‐06‐06, P5‐20‐02,<br />

P6‐07‐25, P6‐10‐07, OT2‐2‐04<br />

Gonzalves, B<br />

P3‐14‐03<br />

Goodall, G<br />

P5‐04‐04<br />

Goodman, D<br />

P4‐13‐13<br />

Goodman, MT<br />

P4‐12‐02<br />

Goodman, RL<br />

P6‐13‐01<br />

Goodrich, ME<br />

P4‐01‐15<br />

Goodwin, PJ PD03‐02, PD03‐05<br />

Gorbunova, V<br />

OT3‐3‐01<br />

Gore, I<br />

P3‐06‐12<br />

Goss, GD<br />

P2‐05‐13<br />

Goss, PE<br />

S1‐9, PD09‐03,<br />

PD10‐05, P1‐07‐13, P2‐10‐15,<br />

P2‐13‐02, OT2‐3‐03<br />

Gossiel, F S6‐4<br />

Goteti, S<br />

OT2‐3‐09<br />

Goto, H<br />

P6‐05‐06<br />

Gotoh, T<br />

P6‐06‐05<br />

Goubar, A<br />

P3‐06‐04<br />

Gouerant, S<br />

P1‐15‐08<br />

Gough, MJ<br />

P4‐04‐02<br />

Goussia, A<br />

P1‐07‐15<br />

Gouy, S<br />

P4‐14‐12<br />

Govek, S<br />

P6‐04‐12<br />

Gown, AM<br />

P1‐07‐01<br />

Gozalbo, F<br />

P1‐01‐29<br />

Grabau, D<br />

PD03‐07,<br />

P2‐10‐07, P2‐10‐28<br />

Grabulovski, D<br />

P5‐18‐25<br />

Gradishar, W<br />

P1‐12‐07,<br />

P5‐17‐01, OT1‐1‐04<br />

Gradishar, WJ<br />

P2‐10‐01,<br />

P2‐14‐01, P5‐03‐11<br />

Graffy, HJ<br />

P4‐13‐09<br />

Graham, L<br />

P5‐14‐04<br />

Graham, N P3‐04‐11, P3‐11‐03<br />

Gralow, J S5‐4, S5‐5, PD10‐04<br />

Gralow, JR P6‐04‐03, P6‐12‐03<br />

Granados, S<br />

P5‐03‐12<br />

Grandemange, S<br />

P6‐01‐04<br />

Granstam Björneklett, H P2‐12‐09<br />

Granville, T<br />

PD04‐04<br />

Grau, AM<br />

P4‐01‐03<br />

Gravel, DH<br />

P5‐01‐01<br />

Gravenor, D<br />

P1‐14‐14<br />

Graves, M<br />

P1‐05‐05<br />

Gravgaard, KH<br />

P4‐09‐04<br />

Graviss, L<br />

P4‐15‐02<br />

Gray, K<br />

PD10‐03<br />

Gray, R S1‐2<br />

Greco, S<br />

P6‐11‐13<br />

Green, A<br />

P2‐10‐22, P3‐06‐13,<br />

P6‐07‐09, P6‐07‐18<br />

Green, J<br />

P6‐10‐01<br />

www.aacrjournals.org 593s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Greenberg, J<br />

P5‐20‐06<br />

Greenhalgh, R<br />

P1‐09‐05<br />

Greenlee, H<br />

P2‐11‐03<br />

Greenson, JK<br />

P5‐18‐06<br />

Gregory, WM S6‐4<br />

Greil, R S4‐3, P2‐10‐02, P5‐17‐03,<br />

P5‐20‐11, OT2‐2‐03<br />

Gremel, G<br />

P4‐09‐06<br />

Gress, J<br />

P4‐11‐04<br />

Grieve, R<br />

PD07‐09<br />

Griffin, RJ<br />

P3‐06‐30<br />

Griffiths, G<br />

PD07‐09<br />

Grillot, K<br />

P6‐04‐12<br />

Grizzle, WE<br />

P2‐05‐22<br />

Grobmyer, SR<br />

P2‐16‐01<br />

Groman, A<br />

P3‐07‐02<br />

Gronesova, P<br />

P2‐01‐12<br />

Grose, VA<br />

P4‐06‐18<br />

Gross, HM<br />

P1‐12‐06<br />

Grosse, R<br />

P2‐10‐38<br />

Grothey, A<br />

P6‐04‐15<br />

Grubbs, C<br />

P1‐08‐02<br />

Grubbs, CJ<br />

P1‐08‐01<br />

Grudé, F<br />

OT3‐3‐04<br />

Grutman, SB<br />

P4‐14‐01<br />

Gruver, AM<br />

PD02‐04<br />

Gryaznov, SM<br />

P5‐08‐01<br />

Grybowicz, L<br />

OT3‐3‐09<br />

Gu, G<br />

P4‐08‐09<br />

Gu, H<br />

P5‐10‐06<br />

Gu, J‐W<br />

P4‐06‐04<br />

Gu, Y<br />

P1‐03‐03<br />

Guan, J<br />

P4‐07‐01, P5‐19‐02<br />

Guardino, E P5‐18‐06, P5‐18‐24<br />

Guariglia, S<br />

P4‐16‐08<br />

Guarneri, V<br />

P2‐10‐16<br />

Guastalla, J‐P S5‐3<br />

Gucalp, A<br />

P6‐05‐01,<br />

P6‐05‐02, OT3‐1‐01<br />

Gudlaugsson, E<br />

P2‐10‐19<br />

Guenther, M<br />

PD07‐01<br />

Guérin, A<br />

P5‐18‐12<br />

Guerra, JA<br />

OT3‐3‐05<br />

Guerrero, A<br />

S1‐7, OT1‐1‐09<br />

Guerrero, J<br />

P2‐14‐02<br />

Guerrieri‐Gonzaga, A PD03‐01,<br />

PD04‐07, PD06‐06<br />

Guggisberg, N<br />

P1‐07‐10<br />

Gui, G<br />

P1‐01‐10, P1‐01‐11<br />

Guidetti, A<br />

P2‐12‐08<br />

Guilhen, N<br />

P1‐01‐07<br />

Guillem, V<br />

OT1‐1‐09<br />

Guimarães, IS<br />

P6‐11‐13<br />

Guinebretière, JM P1‐14‐18, P3‐06‐04<br />

Guiu, S<br />

P3‐06‐20, P3‐08‐07<br />

Guldberg, TL<br />

P6‐07‐08<br />

Gulley, JL<br />

P5‐16‐06<br />

Gumus, M<br />

OT1‐1‐08<br />

Gunnarsdottír, KA<br />

P6‐12‐01<br />

Gunther, JE<br />

P4‐02‐07<br />

Guo, B<br />

P1‐14‐13<br />

Guo, J<br />

P5‐10‐10<br />

Guo, Q<br />

P3‐08‐05<br />

Guo, R<br />

PD10‐02, P1‐12‐04<br />

Guo, X<br />

PD05‐09<br />

Gupta, I<br />

P4‐06‐14, P4‐06‐16<br />

Gurcan, MN<br />

P2‐11‐07<br />

Gusterson, B S1‐1<br />

Gutekunst, K<br />

P2‐10‐04<br />

Guth, A<br />

P1‐09‐03, P2‐11‐12<br />

Guth, AA PD08‐01, P4‐14‐03, P4‐17‐07<br />

Guthrie, K<br />

P5‐16‐04<br />

Gutierrez, C S5‐8, P2‐16‐02, P2‐16‐03<br />

Gutierrez‐Barrera, AM P2‐10‐32<br />

Gutin, A<br />

PD09‐04<br />

Gutkind, JS<br />

P1‐05‐23<br />

Gwin, WR<br />

P4‐08‐03<br />

Györffy, B P1‐07‐14, P2‐10‐24<br />

Ha, T<br />

P3‐05‐05, P3‐05‐06<br />

Haas, M<br />

P6‐14‐05<br />

Habal, N<br />

P4‐14‐01<br />

Habel, L<br />

P3‐07‐05<br />

Haber, D S5‐7<br />

Haber, DA<br />

P2‐01‐14<br />

Haber, J<br />

P2‐11‐12<br />

Habets, L<br />

P1‐07‐16<br />

Hachemi, S<br />

OT2‐3‐09<br />

Hadad, SM<br />

PD03‐02<br />

Haddad, M<br />

P2‐05‐06,<br />

P2‐10‐16, P2‐10‐31<br />

Haddad, TC<br />

P1‐14‐11<br />

Hadj‐Hamou, S<br />

P5‐01‐05<br />

Hadji, P<br />

P2‐13‐01, P6‐09‐08<br />

Hadjiminas, D<br />

P3‐02‐04<br />

Haffty, BG S2‐1, P4‐16‐03, P4‐16‐09<br />

Hagenbeck, C P2‐01‐02, P4‐13‐11<br />

Hagenbeck, CD<br />

OT1‐1‐10<br />

Hager, JH<br />

P6‐04‐12<br />

Hahn, DE<br />

P4‐11‐01<br />

Hahn, S<br />

P1‐05‐21<br />

Hahn, SS<br />

P3‐12‐02<br />

Haibe‐Kains, B<br />

P3‐04‐10<br />

Haibo, W<br />

P2‐10‐26<br />

Haimovitz, K<br />

P6‐08‐10<br />

Hainsworth, JD<br />

OT3‐3‐08<br />

Hajage, D<br />

P2‐01‐01,<br />

P2‐01‐13, OT3‐4‐06<br />

Hakkim, L P4‐06‐14, P4‐06‐16<br />

Halawani, H<br />

OT1‐1‐08<br />

Hale, DF<br />

P5‐16‐02, P5‐16‐05<br />

Haley, B<br />

P1‐07‐04<br />

Haley, V<br />

P4‐06‐10<br />

Halhali, A P4‐06‐19, P6‐04‐29<br />

Hall, J<br />

PD02‐01<br />

Hallett, M<br />

P6‐02‐03<br />

Hallquist, A<br />

P3‐06‐28<br />

Hallum‐Montes, R<br />

P4‐13‐02<br />

Haloua, M<br />

PD04‐01<br />

Hamaker, M<br />

P1‐12‐05<br />

Hamel, F<br />

P5‐01‐05<br />

Hamilton, J<br />

P6‐02‐09<br />

Hamilton, N PD03‐04, P5‐18‐20<br />

Hammond, MEH<br />

P2‐10‐27<br />

Hamy, A‐S<br />

P4‐13‐03<br />

Han, A<br />

P6‐07‐40<br />

Han, C<br />

P1‐06‐01, P4‐01‐01<br />

Han, D<br />

P4‐03‐11<br />

Han, L P1‐07‐13, P2‐09‐01, P2‐13‐02<br />

Han, MX<br />

P2‐05‐05, P4‐09‐09<br />

Han, Q<br />

P4‐07‐04<br />

Han, SN<br />

P6‐07‐05<br />

Han, SW<br />

PD09‐05<br />

Han, W PD05‐07, P1‐01‐08, P1‐14‐16,<br />

P1‐14‐19, P2‐05‐20, P3‐06‐16,<br />

P5‐18‐23<br />

Han, WS<br />

P2‐10‐35<br />

Hanamura, T<br />

P6‐04‐17<br />

Hanby, A<br />

P2‐10‐29<br />

Hanby, AM<br />

P6‐02‐06<br />

Handricks, C<br />

P5‐20‐10<br />

Hanf, V<br />

PD07‐02, P1‐13‐13<br />

Hanker, L<br />

P2‐10‐17<br />

Hanna, GG<br />

P6‐07‐24<br />

Hanna, W P1‐07‐17, P2‐10‐09<br />

Hannah, A<br />

P3‐06‐31<br />

Hannoun‐Levi, J‐M<br />

P4‐15‐01<br />

Hanoch, Y<br />

P4‐11‐03<br />

Hansen, J<br />

P2‐10‐27<br />

Hansen, N<br />

P1‐01‐06<br />

Hansen, SK<br />

P6‐04‐18<br />

Hanson, I<br />

P2‐05‐04<br />

Hansson, AG<br />

OT1‐1‐05<br />

Hanusch, C<br />

S3‐1, P1‐14‐01,<br />

OT1‐1‐13, OT3‐3‐11<br />

Hao, Y P6‐04‐01, P6‐04‐04, P6‐04‐21<br />

Haqq, C<br />

P2‐05‐01<br />

Haque, R P1‐07‐12, P3‐07‐05, P3‐08‐06<br />

Hara, F<br />

P1‐13‐10, P3‐02‐06<br />

Hara, H<br />

P4‐09‐08<br />

Harada, T<br />

P3‐13‐04<br />

Harada‐Shoji, N<br />

P6‐04‐16<br />

Harashima, H<br />

P4‐04‐08<br />

Harbeck, N P1‐13‐13, P2‐10‐08,<br />

P5‐18‐06, P5‐20‐01, P6‐07‐05,<br />

P6‐09‐08, OT1‐1‐16, OT3‐3‐02,<br />

OT3‐3‐06<br />

Harcourt, D P4‐17‐03, P4‐17‐04<br />

Hardefeldt, PJ P1‐11‐01, P4‐12‐07<br />

Harden, E<br />

P5‐14‐02<br />

Harel‐Bellan, A<br />

P5‐03‐01<br />

Hargreaves, BA<br />

P4‐01‐06<br />

Harismendy, O<br />

P2‐06‐01<br />

Harland, R<br />

P6‐07‐13<br />

Harper‐Wynne, C<br />

PD07‐07<br />

Harrigal, CL<br />

P4‐01‐12<br />

Harrington, P<br />

P3‐08‐05<br />

Harris, AL P1‐06‐01, P4‐01‐01<br />

Harris, E S4‐2<br />

Harris, L<br />

P4‐01‐02<br />

Harris, LN PD05‐05, P3‐14‐01<br />

Harrison, BL<br />

P5‐15‐01<br />

Harrison, D P3‐06‐23, P6‐04‐10<br />

Harrison, LB<br />

P4‐15‐03<br />

Harry, A<br />

P4‐17‐06<br />

Hartman, A‐R PD09‐03, PD09‐04<br />

Hartman, L<br />

P2‐10‐07<br />

Hartman, M P2‐11‐06, P3‐07‐03<br />

Hartman, RD<br />

P1‐01‐04<br />

Hartmann, LC<br />

P4‐12‐03,<br />

P5‐01‐08, P6‐06‐01<br />

Harvey, A P6‐09‐09, P6‐09‐10<br />

Harvey, C<br />

OT1‐1‐04<br />

Harvie, MN<br />

P4‐13‐09<br />

Harwin, W<br />

P5‐20‐10<br />

Hasegawa, Y<br />

PD07‐05<br />

Hasenburg, A S1‐5<br />

Hashimoto, T<br />

P3‐13‐04<br />

Hasteh, F<br />

P2‐06‐01<br />

Hatachi, T<br />

P1‐01‐27<br />

Hatschek, T<br />

P1‐05‐07<br />

Hattori, A<br />

OT1‐1‐01<br />

Hattori, M P1‐02‐02, P3‐12‐06<br />

Hatzis, C<br />

P2‐10‐17<br />

Hauranieh, L<br />

P2‐01‐05<br />

Hauschild, M<br />

PD07‐02<br />

Hauser, N<br />

P4‐03‐06<br />

Hauspy, J<br />

P2‐01‐08<br />

Haviland, J<br />

P3‐06‐09<br />

Haviland, JS S4‐1<br />

Hawes, D<br />

P1‐09‐06<br />

Hawtin, RE<br />

P6‐07‐31<br />

Hayashi, J<br />

P6‐04‐19<br />

Hayashi, M<br />

PD07‐05<br />

Hayashi, N<br />

P5‐15‐06,<br />

P6‐07‐23, P6‐07‐38<br />

Hayashi, S‐I<br />

P6‐04‐17<br />

Hayashi, T<br />

P1‐01‐27<br />

Hayashi, Y<br />

P1‐14‐06<br />

Hayashida, T P1‐14‐09, P3‐06‐22,<br />

P4‐03‐03, P4‐09‐08<br />

Hayes, DF S4‐6, S6‐3<br />

Hayes, DN S6‐1<br />

Hayes, MK<br />

P4‐03‐09<br />

Haynes, AB<br />

P4‐15‐02<br />

Haynes, B PD07‐07, P3‐08‐04<br />

Hayward, L<br />

OT3‐3‐09<br />

Hazra, S S5‐7<br />

He, HH<br />

PD01‐08, P1‐07‐07<br />

He, J<br />

PD02‐07<br />

He, Y S6‐6<br />

Healy, LA<br />

P6‐01‐02<br />

Heath, D<br />

P2‐11‐09<br />

Heckman‐Stoddard, B P1‐09‐07<br />

Hedenfalk, I P1‐05‐07, P2‐10‐28<br />

Hedvat, C<br />

P2‐08‐01<br />

Hedvat, CV<br />

P6‐05‐01<br />

Hee‐Dong, H<br />

P5‐10‐03<br />

Heery, CR<br />

P5‐16‐06<br />

Hefti, MM<br />

P5‐01‐03<br />

Heguy, A<br />

PD04‐05<br />

Hei, TK<br />

PD09‐09<br />

Heideman, JL<br />

P6‐12‐02<br />

Heijnsdijk, EA<br />

P3‐02‐09<br />

Heinrich, B<br />

P5‐20‐01<br />

Heinrich, G<br />

P2‐01‐02<br />

Heinrich, T<br />

P2‐10‐17<br />

Heintz, A<br />

P6‐14‐05<br />

Heinz, R<br />

P5‐10‐06<br />

Heinz, RE<br />

P4‐12‐04<br />

Heinzmann, D<br />

P5‐18‐02<br />

Heiss, B<br />

P2‐13‐05<br />

Helderman‐van den Enden, A P6‐08‐02<br />

Helenowski, I<br />

P1‐09‐07<br />

Helgadottir, H<br />

PD09‐10<br />

Helland, Å<br />

P3‐05‐04<br />

Hellwinkel, O<br />

P6‐05‐13<br />

Helmijr, JC<br />

P3‐06‐01<br />

Helmijr, JCA<br />

P6‐04‐08<br />

Helms, G S2‐2<br />

Hembrough, TA<br />

P1‐07‐19<br />

Henderson, B<br />

P4‐12‐02<br />

Henderson, D P3‐05‐05, P3‐05‐06<br />

Henderson, K<br />

PD08‐02<br />

Henderson, LM<br />

P4‐01‐15<br />

Hendricks, C P1‐14‐14, P5‐17‐05<br />

Hendricks, P<br />

P6‐05‐10<br />

Hendriks, B P1‐07‐03, P4‐02‐05<br />

Hengstler, JG<br />

P2‐10‐13<br />

Hennessy, C<br />

P4‐17‐09<br />

Hennessy, S<br />

P4‐13‐01<br />

Hennings, L<br />

P3‐06‐30<br />

Henry, NL<br />

PD10‐08<br />

Henschel, V<br />

S6‐5, P3‐06‐34<br />

Hepp, P<br />

P2‐10‐25, P4‐13‐11<br />

HERA Study Team S5‐2, P5‐18‐02<br />

Heras, L<br />

P1‐07‐18, P2‐01‐06<br />

Herbert, B‐S P5‐08‐01, P5‐18‐13<br />

Hergenroeder, G<br />

PD01‐01<br />

Herlinger, AL<br />

P6‐11‐13<br />

Hermann, R<br />

P5‐20‐08<br />

Hernandez, BY<br />

P4‐12‐02<br />

Hernandez, C<br />

P5‐14‐02<br />

Hernandez, RK<br />

P2‐13‐03<br />

Herrera, LA<br />

PD08‐06<br />

Herrero, M P1‐14‐10, P5‐18‐15<br />

Hershman, DL PD03‐03, P1‐09‐02,<br />

P2‐11‐03, P2‐12‐01,<br />

P4‐02‐07, P6‐12‐03<br />

Hertz, DL<br />

PD10‐07<br />

Hertzog, B<br />

P5‐01‐09<br />

Herzog, J<br />

PD08‐06<br />

Hess, KR<br />

P2‐03‐01<br />

Hesselberth, J<br />

PD01‐07<br />

Heuts, EM<br />

P5‐01‐13<br />

Heyer, DM<br />

P4‐06‐18<br />

Heyman, R<br />

P6‐04‐12<br />

Heymanns, J<br />

P2‐11‐11<br />

Hibbs, S<br />

P4‐06‐03<br />

Hibshoosh, H PD03‐03, P4‐02‐07<br />

Hickish, T<br />

OT1‐1‐17<br />

Hickman, J<br />

P1‐09‐05<br />

Hicks, J<br />

P3‐14‐01<br />

Hidalgo, M P1‐14‐10, P5‐18‐15<br />

Hidalgo‐Miranda, A<br />

P5‐10‐12<br />

Hiddingh, L<br />

P3‐12‐03<br />

Hieken, TJ<br />

P1‐01‐22<br />

Hielscher, A<br />

P4‐02‐07<br />

Higaki, K P1‐14‐08, P1‐14‐12, P4‐01‐05<br />

Higashide, S<br />

P3‐13‐04<br />

Higgens, D<br />

P5‐16‐04<br />

Higgins, D<br />

P2‐15‐02<br />

Higgins, DG<br />

P4‐09‐06<br />

Higgins, DM<br />

OT3‐1‐02<br />

Higgins, HB OT1‐1‐03, OT3‐3‐09<br />

Higgins, MJ<br />

P2‐13‐02<br />

Higgins, SA<br />

P4‐17‐02<br />

Hikichi, M<br />

P3‐02‐05<br />

Hilaire, L<br />

P2‐10‐04<br />

Hilbert, P<br />

P3‐04‐04<br />

Hilfrich, J S3‐1, S4‐5, PD06‐01,<br />

P1‐14‐01, OT1‐1‐13, OT3‐3‐11<br />

Hill, A<br />

P6‐04‐21<br />

Hill, ADK<br />

P6‐04‐01<br />

Hiller, L OT1‐1‐03, OT3‐3‐09<br />

Hiller, Ral<br />

P2‐01‐11<br />

Hillier, S<br />

P3‐08‐04<br />

Hills, M<br />

PD07‐07<br />

Hilsenbeck, SG S5‐8, PD01‐01<br />

Hilton, JF<br />

P3‐13‐05<br />

Hinck, AP<br />

P1‐05‐18<br />

Hindenburg, H‐J<br />

P6‐09‐08<br />

Hinke, A<br />

P5‐21‐02<br />

Hinrichs, JW<br />

PD02‐03<br />

Hinzey, A<br />

P2‐11‐04<br />

Hirakawa, H<br />

P1‐01‐27<br />

Hirano, A<br />

OT1‐1‐01<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

594s<br />

<strong>Cancer</strong> Research


December 4-8, 2012<br />

Author Index<br />

Hirawat, S P4‐08‐01, OT2‐3‐08<br />

Hiroji, I<br />

P1‐02‐02<br />

Hirokaga, K P6‐07‐30, P6‐07‐39<br />

Hiroko, B<br />

P2‐11‐02<br />

Hirose, S<br />

P1‐14‐09, P3‐06‐22<br />

Hirshfield‐Bartek, J<br />

P3‐10‐06,<br />

P4‐06‐01, P5‐02‐01<br />

Hisamatsu, K<br />

P1‐14‐02<br />

Hitora, T<br />

P4‐03‐07<br />

Hjermstad, M<br />

P6‐09‐03<br />

Ho, C‐L<br />

P4‐04‐07<br />

Hoadley, KA S6‐1<br />

Hoch, U<br />

P3‐06‐31<br />

Hodge, JW<br />

P5‐16‐06<br />

Hodge, S<br />

P2‐05‐15, P5‐16‐06<br />

Hoe, N<br />

P4‐06‐08<br />

Hoefnagel, LDC<br />

PD01‐02,<br />

P2‐06‐02, P6‐05‐08<br />

Hoess, C<br />

P5‐14‐06, P5‐15‐04<br />

Hoffman, A<br />

P1‐03‐01<br />

Hoffman, RM<br />

P4‐07‐04<br />

Hoffmann, G<br />

P1‐13‐13<br />

Hoffmann, J<br />

P6‐04‐23<br />

Hofmann, D<br />

OT3‐3‐02<br />

Hogarth, M<br />

P4‐13‐13<br />

Hogben, K<br />

P3‐02‐04<br />

Hogervorst, FB<br />

P4‐11‐01<br />

Hohl, MK<br />

P4‐03‐06<br />

Hojo, T<br />

P3‐02‐06<br />

Holcombe, C<br />

P4‐17‐03<br />

Holden, AE<br />

PD08‐05<br />

Holen, I<br />

PD07‐08, P2‐02‐04<br />

Holko, M<br />

P2‐10‐30<br />

Hollenbeck, AR<br />

P3‐07‐04<br />

Holliday, DL<br />

P6‐02‐06<br />

Hollingsworth, R<br />

P4‐07‐05<br />

Hollman, D P3‐05‐05, P3‐05‐06<br />

Holmberg, E<br />

P6‐12‐01<br />

Holmberg, L<br />

P2‐10‐29<br />

Holmdahl, J<br />

P2‐10‐07<br />

Holmes, FA<br />

P3‐06‐12<br />

Holmes, JP<br />

P5‐16‐02<br />

Holst, F<br />

P6‐05‐13<br />

Holt, S<br />

P1‐01‐18<br />

Holtrich, U<br />

P2‐10‐17<br />

Holtveg, H S4‐2<br />

Holz, MK<br />

P1‐04‐02<br />

Holzhausen, H‐J<br />

P2‐10‐38<br />

Hong, C‐C<br />

P3‐07‐02<br />

Hong, D<br />

P5‐20‐09<br />

Honisch, E<br />

P2‐07‐01<br />

Honma, N<br />

P3‐06‐18<br />

Hood, K<br />

PD07‐09<br />

Hood, N<br />

PD03‐05<br />

Hoog, J<br />

PD07‐01<br />

Hoogerbrugge, N<br />

P3‐02‐09<br />

Hooke, JA<br />

P2‐05‐21,<br />

P5‐01‐07, P6‐02‐08<br />

Hooning, M<br />

P3‐02‐09<br />

Hooning, MJ<br />

P4‐11‐05<br />

Hopkins, S<br />

P3‐13‐05<br />

Hopper, JL<br />

P4‐13‐10<br />

Hopper‐Borge, E<br />

P4‐08‐04<br />

Hopwood, P S4‐1<br />

Horgan, K P1‐13‐08, P4‐01‐14<br />

Horgan, M<br />

P3‐02‐10<br />

Horie‐Inoue, K<br />

P6‐04‐27<br />

Horiguchi, J PD07‐05, P1‐14‐06,<br />

P2‐05‐07, P3‐06‐29, P5‐18‐16<br />

Horii, R<br />

P1‐01‐12, P5‐01‐02<br />

Horio, A<br />

P1‐02‐02, P3‐12‐06<br />

Horiuchi, D S3‐8<br />

Hornberger, J<br />

P5‐15‐06<br />

Horne, HN<br />

P3‐08‐02<br />

Horng, C‐F<br />

P4‐16‐05<br />

Horowitz, NR P3‐14‐05, P3‐14‐06<br />

Horst, KC P4‐16‐11, P6‐08‐10<br />

Horstman, A<br />

PD02‐03<br />

Hortobagyi, G<br />

P6‐04‐02<br />

Hortobagyi, GN PD03‐06, PD03‐08,<br />

P1‐07‐09, P2‐01‐01, P2‐03‐01,<br />

P3‐10‐05, P5‐03‐09, P5‐04‐06,<br />

P5‐10‐02, P5‐20‐02, P5‐20‐13,<br />

OT2‐2‐04, OT2‐3‐10<br />

Horvat, R<br />

P2‐10‐31<br />

Horvath, LE<br />

P1‐12‐04<br />

Hoskin, P S4‐1<br />

Hoskins, KF<br />

P5‐13‐03<br />

Hosokawa, Y<br />

P4‐03‐05<br />

Hostetter, G<br />

P1‐05‐04<br />

Houck, III, WA<br />

OT1‐1‐07<br />

Houdayer, C<br />

P5‐02‐03<br />

Houk, B<br />

OT2‐3‐02<br />

Houlston, R<br />

P3‐08‐04<br />

Houpeau, J‐L<br />

P1‐01‐21,<br />

P4‐14‐10, OT2‐1‐01<br />

Houshmand, M<br />

P5‐09‐01<br />

Houvenaeghel, G P1‐01‐21, P5‐03‐01,<br />

OT2‐1‐01, OT2‐3‐05<br />

Hoverman, R<br />

P2‐11‐16<br />

Howard, N<br />

OT3‐4‐02<br />

Howe, M<br />

P3‐14‐04<br />

Howe, R<br />

P4‐11‐03<br />

Howell, A P1‐09‐05, P2‐14‐01,<br />

P3‐08‐01, P4‐13‐09<br />

Howell, L<br />

P4‐13‐13<br />

Howell, SJ<br />

P2‐14‐06<br />

Howitt, J<br />

P2‐10‐16<br />

Hozumi, Y P2‐13‐04, P2‐13‐07<br />

Hsiao, A<br />

P6‐11‐03, P6‐11‐11<br />

Hsieh, Y‐F<br />

P4‐04‐07<br />

Hsu, C‐H<br />

PD08‐04<br />

Hsu, L<br />

P3‐10‐05<br />

Hu, H P1‐05‐09, P2‐05‐21, P2‐10‐30,<br />

P3‐04‐06, P3‐04‐08, P3‐09‐03,<br />

P4‐06‐02, P5‐01‐07<br />

Hu, J<br />

P6‐07‐17<br />

Hu, P<br />

P4‐05‐01<br />

Hu, R<br />

P5‐01‐03<br />

Hu, X<br />

P6‐11‐01<br />

Hu, X‐C<br />

P6‐07‐43<br />

Huang, AT<br />

P4‐16‐05<br />

Huang, C<br />

P5‐03‐06<br />

Huang, C‐S<br />

P3‐06‐02,<br />

OT1‐1‐16, OT1‐1‐17<br />

Huang, H<br />

P3‐07‐06,<br />

P3‐07‐07, P3‐07‐14<br />

Huang, J<br />

P2‐05‐05<br />

Huang, L<br />

P2‐10‐29, P6‐07‐36<br />

Huang, P<br />

P2‐05‐05<br />

Huang, W<br />

P2‐05‐06,<br />

P2‐10‐16, P2‐10‐31<br />

Huang, Y<br />

P3‐12‐03,<br />

P6‐13‐02, OT1‐1‐04<br />

Hubbard, RA<br />

P4‐01‐15<br />

Hubert, C<br />

PD05‐04<br />

Hucke, J<br />

P2‐10‐25<br />

Hudis, C S5‐4, S5‐5, P1‐13‐11,<br />

P2‐12‐05, P5‐18‐04, P5‐18‐20,<br />

P6‐11‐10, OT3‐1‐01<br />

Hudis, CA P2‐10‐01, P5‐20‐07,<br />

P6‐05‐01, P6‐05‐02, P6‐08‐05<br />

Hudry, D<br />

P1‐01‐07<br />

Hughes, E<br />

P6‐04‐01<br />

Hughes, M<br />

P1‐01‐13<br />

Hughes, ME S3‐5<br />

Hughes, MR<br />

P1‐05‐05<br />

Hughes, NP P1‐06‐01, P4‐01‐01<br />

Hughes, TA<br />

P1‐13‐08<br />

Hui, M<br />

P3‐07‐03<br />

Huisman, JJ<br />

P4‐11‐01<br />

Hung, M‐H<br />

P3‐12‐10<br />

Hunt, K<br />

PD07‐01<br />

Hunt, KK<br />

S2‐1, P1‐01‐06,<br />

P1‐07‐09, P4‐15‐02<br />

Huo, D<br />

P2‐10‐21<br />

Huo, L<br />

P1‐07‐09<br />

Huober, J<br />

S3‐1, S3‐4, S4‐5,<br />

PD06‐01, P1‐14‐01, P3‐06‐05,<br />

OT1‐1‐13, OT3‐3‐11<br />

Hurd, T<br />

P1‐01‐06<br />

Hurlbert, M<br />

P3‐02‐07,<br />

P4‐13‐02, P5‐14‐02<br />

Hurley, E<br />

P3‐01‐04<br />

Hurley, L<br />

P2‐10‐16<br />

Hurria, A<br />

P2‐14‐08, P6‐08‐05<br />

Hurtado Rua, SM<br />

P6‐11‐04<br />

Hurvitz, S<br />

P4‐01‐13<br />

Hurvitz, SA P4‐08‐01, P4‐08‐05<br />

Hurvitz, S‐A<br />

OT1‐1‐16<br />

Hutchens, S<br />

P2‐12‐11<br />

Huws, A<br />

P1‐01‐18<br />

Hwang, B‐M<br />

P5‐07‐04<br />

Hwang, ES P4‐14‐04, P4‐16‐07<br />

Hwang, H<br />

P1‐01‐23<br />

Hwang, HC<br />

P1‐07‐01<br />

Hwang, J<br />

P2‐12‐11<br />

Hwang, RF<br />

P4‐15‐02<br />

Hwang, S P1‐01‐13, P2‐10‐03,<br />

P3‐06‐07, P4‐01‐13, P4‐13‐06<br />

Hwang, SH<br />

P2‐05‐14<br />

Hwang, SO<br />

P1‐01‐24<br />

Hwangbo, SM<br />

P1‐01‐24<br />

Hylton, NM<br />

P4‐01‐04,<br />

P4‐01‐10, P4‐01‐13<br />

Hynes, NE<br />

PD01‐06<br />

Ibarra‐Drendall, C<br />

P1‐03‐01<br />

Ibrahim, NK P5‐16‐01, P5‐16‐06<br />

Ibusuki, M<br />

P6‐05‐06<br />

Ichinose, Y<br />

P3‐13‐04<br />

Ide, Y<br />

P4‐03‐05<br />

Idvall, I<br />

P2‐10‐37<br />

Igawa, A<br />

P3‐06‐26<br />

Iglehart, JD<br />

P1‐07‐07<br />

Iglesias, J<br />

P1‐12‐07<br />

Ignatiadis, M S4‐4, P6‐07‐14, P6‐07‐22<br />

Iiboshi, Y<br />

P4‐03‐07<br />

Iida, J<br />

P1‐05‐09, P3‐04‐06<br />

Ijichi, N<br />

P6‐04‐27<br />

Ikeda, DM<br />

P4‐01‐12<br />

Ikeda, K<br />

P6‐04‐27<br />

Iliopoulos, D<br />

P6‐06‐02<br />

Illi, J<br />

P3‐04‐01, P3‐04‐02<br />

Im, S‐A S5‐1, PD09‐05, P1‐14‐19,<br />

P2‐05‐20, OT1‐1‐16<br />

Im, Y‐H PD09‐05, P2‐05‐20, P2‐14‐01,<br />

P4‐06‐13, OT1‐1‐16<br />

Imai, S<br />

P3‐13‐04<br />

Imawari, Y P4‐03‐10, P5‐04‐02<br />

Imoto, S<br />

P2‐13‐04, P2‐13‐07<br />

In, Y‐H<br />

PD05‐07<br />

Inaba, M<br />

P1‐01‐20<br />

Inaji, H<br />

P2‐10‐18<br />

Inamoto, T<br />

P3‐13‐04<br />

Inbar, MJ<br />

P5‐17‐03<br />

Incio, J<br />

P3‐12‐03<br />

Infante, JR<br />

P6‐11‐06<br />

Infante, KP<br />

P1‐01‐28<br />

Ingber, D<br />

P5‐05‐04<br />

Ingber, DE<br />

P2‐04‐03<br />

Ingham, SL<br />

P3‐08‐01<br />

Ingle, J<br />

P6‐05‐02<br />

Ingle, JN<br />

P2‐13‐02, OT1‐1‐11<br />

Ingledew, P P5‐12‐01, P5‐12‐02<br />

Inhorn, RC<br />

P5‐17‐05<br />

Inoges, S<br />

P5‐16‐03<br />

Inokuchi, M<br />

P4‐16‐13<br />

Inoue, H<br />

OT1‐1‐01<br />

Inoue, K<br />

P1‐14‐06, P3‐02‐06<br />

Inoue, S<br />

P6‐04‐27<br />

International Ki67 in <strong>Breast</strong><br />

<strong>Cancer</strong> Working Group of the<br />

BIG‐NABCG Collaboration S4‐6<br />

Ioffe, OB<br />

P2‐10‐40<br />

Iorns, E<br />

P1‐05‐12<br />

Ip, P S5‐7<br />

Irvin, WJ<br />

PD10‐08<br />

Irwin, M<br />

P2‐11‐08<br />

Isaacs, C<br />

P1‐09‐04<br />

Isakoff, SJ PD09‐03, P2‐01‐14,<br />

P3‐06‐27, P6‐05‐02,<br />

P6‐10‐05, OT2‐3‐07<br />

Isambert, N<br />

P6‐11‐08<br />

Ishibe, Y<br />

P3‐07‐10<br />

Ishida, K<br />

P6‐05‐14<br />

Ishida, M<br />

P3‐06‐26<br />

Ishida, T P1‐06‐02, P2‐10‐43, P6‐05‐14<br />

Isidoro, M<br />

P1-15-03<br />

Ishiguchi, T<br />

P4‐03‐08<br />

Ishiguro, H<br />

P5‐18‐16<br />

Ishihara, S<br />

P3‐07‐10<br />

Ishikawa, T<br />

PD07‐05<br />

Ishioka, C<br />

P2‐10‐43<br />

Isikdogan, A<br />

OT1‐1‐08<br />

Ismail‐Khan, R<br />

P2‐14‐04<br />

Isner, P<br />

P2‐10‐03<br />

Isola, J<br />

PD10‐06<br />

I‐SPY 1 TRIAL Investigators P2‐10‐06<br />

I‐SPY TRIAL‐1 Investigators P2‐05‐01,<br />

P3‐04‐01,<br />

P3‐04‐02, P3‐06‐10<br />

Ito, K‐I<br />

P6‐04‐17<br />

Ito, M<br />

P1‐14‐12, P4‐01‐05<br />

Ito, T<br />

P4‐03‐07<br />

Ito, Y<br />

P1‐14‐02, P5‐18‐16<br />

Itoi, N<br />

P1‐15‐11<br />

Itoyanagi, N<br />

P1‐01‐27<br />

Ivancic, D P2‐10‐30, P3‐04‐08<br />

Iversen, A<br />

P3‐01‐01<br />

Iwamoto, M P3‐02‐02, P3‐09‐04<br />

Iwamoto, T PD03‐08, P2‐05‐08,<br />

P2‐10‐10, P3‐07‐10,<br />

P3‐10‐05, P5‐10‐01<br />

Iwaniuk, KM<br />

P2‐07‐01<br />

Iwasa, M<br />

P4‐12‐06<br />

Iwase, H P1‐12‐01, P2‐14‐03, P6‐05‐06<br />

Iwase, T<br />

P1‐01‐12<br />

Iwata, H P1‐12‐01, P1‐14‐02, P2‐13‐04,<br />

P2‐13‐07, P3‐02‐06, P3‐12‐06,<br />

P5‐18‐16, P6‐07‐16,<br />

OT2‐3‐03, OT2‐3‐09<br />

Iyer, S<br />

P3‐04‐08<br />

Iyeyasu, H<br />

P4‐02‐03<br />

Izukura, M<br />

P4‐03‐07<br />

Izumchenko, E<br />

P1‐05‐03<br />

Jac, J<br />

P6‐11‐10<br />

Jacene, H<br />

P5‐02‐01<br />

Jacene, HA P3‐10‐06, P4‐06‐01<br />

Jackisch, C S3‐1, S3‐1, S3‐4, S4‐5,<br />

S5‐2, S6‐5, PD06‐01,<br />

P1‐14‐01, P5‐18‐02, P6‐09‐08,<br />

OT1‐1‐13, OT3‐3‐11<br />

Jackson, SA<br />

P5‐20‐13<br />

Jacob, J<br />

P5‐21‐03<br />

Jacobs, J<br />

P4‐08‐04<br />

Jacobs, LK<br />

OT2‐1‐04<br />

Jacobs, SA<br />

OT1‐1‐12<br />

Jacobs, VR P5‐14‐06, P5‐15‐04<br />

Jacobsen, BM<br />

P2‐14‐02<br />

Jacquemier, J<br />

P3‐05‐07,<br />

P3‐14‐03, P5‐03‐01<br />

Jaeger, BAS<br />

P2‐01‐02,<br />

P4‐13‐11, OT1‐1‐10<br />

Jaenicke, F<br />

P1‐13‐13<br />

Jafari, N<br />

P2‐12‐10<br />

Jaffray, D<br />

P4‐02‐05<br />

Jaganathan, H<br />

P6‐02‐07<br />

Jahanzeb, M<br />

OT3‐3‐07<br />

Jain, M<br />

OT1‐1‐17<br />

Jain, RK<br />

P3‐12‐03<br />

Jain, S<br />

P1‐15‐07, P6‐11‐04<br />

Jakesz, R<br />

S4‐3, P2‐10‐02<br />

Jalaguier, A P5‐03‐01, OT2‐3‐05<br />

James, CR<br />

P6‐07‐24<br />

Jammallo, LS P6‐09‐04, OT3‐2‐02<br />

Jamshidian, F S1‐10, P6‐07‐03<br />

Janevski, A PD05‐05, P4‐01‐02<br />

Jankowitz, R<br />

P6‐07‐02<br />

Jankowski, T<br />

P2‐10‐31<br />

Janku, F<br />

P5‐20‐09<br />

Janni, JW<br />

P1‐14‐04,<br />

P4‐13‐11, OT1‐1‐10<br />

Janni, W<br />

P2‐01‐02,<br />

P2‐10‐25, P5‐18‐21<br />

Jansen, MPHM P3‐06‐01, P6‐04‐08<br />

Janssen, E<br />

P2‐10‐19<br />

Janus, N<br />

P5‐17‐04<br />

Jarosz, M<br />

PD02‐07<br />

Jarrett, BL<br />

P2‐11‐04<br />

Jasin, M<br />

PD09‐10<br />

Jaslow, RC<br />

P5‐14‐03<br />

Jassem, J P2‐10‐31, P3‐12‐09, P3‐13‐03,<br />

P5‐02‐02, P5‐04‐01, OT1‐1‐16<br />

Jassim, GA<br />

P6‐08‐07<br />

Javadi, GH<br />

P5‐09‐01<br />

Javed, Y<br />

OT3‐3‐07<br />

www.aacrjournals.org 595s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Jaworska‐Jankowska, M P3‐12‐09,<br />

P3‐13‐03<br />

Jay, A<br />

PD03‐09<br />

Jean, A<br />

P5‐10‐06<br />

Jefferys, SR S6‐1<br />

Jen, J<br />

PD10‐04<br />

Jenkins, RB PD02‐05, PD10‐04<br />

Jensen, AB<br />

P6‐07‐08<br />

Jensen, K<br />

P1‐07‐05<br />

Jensen, KC<br />

PD09‐04<br />

Jensen, RA<br />

PD09‐02<br />

Jeong, J<br />

P2‐05‐14<br />

Jeong, J‐H S1‐10, S5‐5<br />

Jerusalem, G S1‐4<br />

Jeschke, U<br />

P2‐01‐11<br />

Jeselsohn, RM<br />

P1‐07‐07<br />

Jesien‐Lewandowicz, E P3‐12‐09,<br />

P3‐13‐03<br />

Jeter, S<br />

P2‐02‐01<br />

Jeuris, K<br />

P2‐01‐08<br />

Ji, L<br />

P4‐01‐11<br />

Jia, Z<br />

P6‐07‐43<br />

Jian, J‐MJ<br />

P4‐16‐05<br />

Jiang, J S2‐3<br />

Jiang, JX<br />

P3‐13‐06<br />

Jiang, W<br />

P1‐04‐08<br />

Jiang, WG P1‐04‐03, P1‐04‐07,<br />

P1‐04‐09, P6‐05‐11<br />

Jiang, Y‐Z<br />

P2‐06‐07<br />

Jibiki, N<br />

OT1‐1‐01<br />

Jimenez, J<br />

P1‐07‐19<br />

Jin, J P5‐18‐11, P5‐18‐24, P6‐10‐04<br />

Jin, K P2‐09‐01, P4‐06‐08, P6‐04‐13<br />

Jin, L<br />

P1‐01‐09, P6‐01‐03<br />

Jin, W<br />

P6‐04‐26<br />

Jindal, S<br />

P4‐04‐03<br />

Jinno, H<br />

P1‐14‐09, P3‐06‐22,<br />

P4‐03‐03, P4‐09‐08<br />

Jirstrom, K<br />

PD03‐07,<br />

P2‐10‐28, P4‐09‐06<br />

Jiwa, M<br />

PD02‐03<br />

Jo, J‐C<br />

P3‐12‐05<br />

Jo, J‐H<br />

P6‐07‐28<br />

Joe, BN<br />

P4‐01‐10<br />

Joensuu, H PD10‐06, OT1‐1‐15<br />

Joerger, M<br />

OT2‐2‐02<br />

Joffres, M<br />

P4‐17‐08<br />

Joglekar, M P1‐04‐10, P6‐06‐03<br />

Johansson, H PD06‐06, P6‐14‐01<br />

Johansson, O<br />

P2‐10‐37<br />

Johnson, N<br />

P3‐08‐04<br />

Johnson, R<br />

P3‐14‐04<br />

Johnston, S<br />

PD07‐07,<br />

P6‐07‐15, OT1‐1‐04<br />

Johnston, SA<br />

P6‐12‐02<br />

Johnston, SRD<br />

P2‐14‐01<br />

Jonat, W OT2‐3‐09, OT3‐2‐01<br />

Jones, EF<br />

P4‐01‐04<br />

Jones, JG<br />

P6‐02‐04<br />

Jones, KN<br />

P1‐01‐22<br />

Jones, LA<br />

P1‐01‐01<br />

Jones, M<br />

P3‐08‐04<br />

Jones, N<br />

PD05‐04<br />

Jones, NR<br />

P2‐11‐15<br />

Jones, RA<br />

P1‐04‐05<br />

Jones, S<br />

P3‐06‐12<br />

Joore, MA<br />

P5‐21‐04<br />

Jordan, K<br />

P4‐04‐06<br />

Jordan, LB<br />

PD03‐02<br />

Jordan, VC<br />

P2‐13‐09<br />

Jørgensen, CLT<br />

P3‐06‐03<br />

Jose, V<br />

P1‐07‐08<br />

Joseph, D S4‐2<br />

Joseph, J<br />

P6‐04‐12<br />

Joshi, A<br />

PD01‐01<br />

Josselin, E<br />

P5‐03‐01<br />

Jouannaud, C P3‐06‐20, P5‐17‐04<br />

Joukov, V<br />

P4‐10‐01<br />

Jovanovic, B P1‐09‐07, P3‐05‐09<br />

Joy, MT<br />

P1‐01‐04<br />

Jud, SM<br />

PD07‐02<br />

Judde, J‐G<br />

P3‐06‐24<br />

Jueckstock, J<br />

P4‐13‐11<br />

Juhasz, E<br />

P3‐05‐08<br />

Julian, TB S1‐10, S2‐1, OT1‐2‐01<br />

Jun, N<br />

P3‐05‐05, P3‐05‐06<br />

Jung, HH<br />

P4‐06‐13<br />

Jung, JH<br />

P1‐01‐24<br />

Jung, K<br />

OT1‐1‐16<br />

Jung, KH<br />

P3‐12‐05<br />

Jung, K‐H<br />

P6‐01‐05<br />

Jung, ME<br />

PD03‐04<br />

Jung, MS S6‐3<br />

Jung, S<br />

P2‐11‐05<br />

Jung, SH<br />

P1‐10‐01, P5‐07‐04<br />

Jung, SP<br />

P3‐11‐01<br />

Jung, SS<br />

P2‐05‐02, P4‐03‐01<br />

Jung, S‐Y<br />

P1‐01‐03<br />

Jungberg, P<br />

P5‐21‐02<br />

Junghaenel, DU<br />

P6‐08‐06<br />

Jupe, ER<br />

P4‐12‐05<br />

Juretic, A<br />

P4‐04‐04<br />

Juric, D<br />

S5‐7, P6‐10‐07<br />

Justice, R S1‐11<br />

Jyrkkiö, S<br />

PD10‐06<br />

Kaanumalle, L<br />

P3‐05‐06<br />

Kaas, R<br />

P4‐11‐05<br />

Kabos, P<br />

PD01‐07, P6‐05‐10<br />

Kadlubar, SA<br />

P3‐06‐30<br />

Kadouri‐Sonenfeld, L P5‐20‐06<br />

Kadoya, T<br />

P6‐07‐26<br />

Kahan, Z<br />

P5‐02‐02, P5‐17‐03<br />

Kahraman, M<br />

P6‐04‐12<br />

Kai, K PD03‐08, P2‐04‐01, P3‐10‐05<br />

Kaklamani, VG<br />

P5‐03‐11<br />

Kakolyris, S<br />

P1‐13‐09<br />

Kakugawa, Y<br />

P6‐07‐27<br />

Kalinsky, K PD03‐03, P1‐09‐02,<br />

P2‐01‐10, P2‐08‐01,<br />

P2‐11‐03, P2‐12‐01,<br />

P4‐02‐07, P6‐05‐01, P6‐12‐03<br />

Kallakury, BVS<br />

P1‐09‐04<br />

Kallergi, G P2‐01‐03, P2‐01‐07<br />

Kalna, G<br />

P1‐05‐13<br />

Kalogeras, KT<br />

P1‐07‐15<br />

Kalous, O<br />

P4‐08‐01<br />

Kamada, Y<br />

P1‐06‐02<br />

Kamalakaran, S PD05‐05, P4‐01‐02<br />

Kamath, V<br />

P3‐05‐06<br />

Kamigaki, S<br />

P1‐14‐02<br />

Kamimura, M<br />

OT1‐1‐01<br />

Kamio, M P4‐03‐10, P5‐04‐02<br />

Kamischke, A<br />

OT2‐2‐05<br />

Kammler, R<br />

PD10‐03<br />

Kamoun, W<br />

P3‐12‐03<br />

Kan, N<br />

P3‐13‐04<br />

Kandalaft, PL<br />

P1‐07‐01<br />

Kaneda, M<br />

P4‐09‐08<br />

Kaneko, K<br />

P1‐14‐08<br />

Kang, E<br />

P5‐04‐03<br />

Kang, J<br />

P6‐07‐28<br />

Kang, MJ<br />

P3‐12‐05<br />

Kang, SH<br />

P5‐04‐07<br />

Kang, S‐H P1‐01‐05, P3‐03‐02<br />

Kanji, S<br />

P2‐11‐07<br />

Kannarkat, G<br />

P5‐20‐10<br />

Kantelhardt, EJ<br />

P1‐13‐13<br />

Kantelhardt, E‐J<br />

P2‐10‐38<br />

Kao, W‐Y<br />

P4‐04‐07<br />

Kapitza, T P5‐14‐06, P5‐15‐04<br />

Kaplan, C<br />

P4‐13‐13<br />

Kaplan, CP<br />

P4‐13‐05<br />

Kapoor, S<br />

P3‐06‐10<br />

Karaba, M<br />

P2‐01‐12<br />

Karaszewska, B<br />

P5‐18‐21<br />

Karimi, M<br />

P3‐06‐28<br />

Karkera, J<br />

P5‐01‐09<br />

Karlin, NJ<br />

P1‐12‐06<br />

Karliner, L<br />

P4‐13‐05<br />

Karlis, D<br />

P5‐18‐02<br />

Karn, T<br />

P2‐10‐17, P3‐06‐05<br />

Karp, JE S3‐5<br />

Karp, N<br />

P4‐14‐03, P4‐17‐07<br />

Kasem, A P1‐04‐08, P1‐04‐09<br />

Kash, JJ<br />

P1‐12‐04<br />

Kashef‐Haghighi, D<br />

PD05‐09<br />

Kashiwaba, M<br />

P1‐14‐02,<br />

P1‐14‐08, P5‐18‐16<br />

Kasiewicz, M<br />

P4‐04‐02<br />

Kass, EM<br />

PD09‐10<br />

Kast, J<br />

P1‐05‐06<br />

Kataja, V<br />

PD10‐06<br />

Katayama, H<br />

P2‐04‐05<br />

Katchman, BA<br />

P1‐05‐04<br />

Katdare, M<br />

P1‐10‐02<br />

Kates, RE P2‐10‐08, OT3‐3‐02<br />

Kato, H<br />

P3‐13‐04<br />

Kato, K<br />

P4‐03‐10, P5‐04‐02<br />

Kato, T<br />

P2‐11‐02<br />

Katzorke, N<br />

P4‐13‐11<br />

Kaufman, B<br />

P5‐17‐03,<br />

P5‐20‐06, OT2‐3‐07<br />

Kaufman, CS<br />

PD04‐04<br />

Kaufman, J<br />

P6‐04‐12<br />

Kaufman, PA<br />

S6‐6, P2‐05‐06<br />

Kaufmann, M P2‐10‐17, P6‐07‐05<br />

Kaul, K<br />

P3‐05‐01<br />

Kaur, K<br />

P1‐06‐01<br />

Kaushik, M<br />

P1‐14‐15<br />

Kavuri, SM S5‐6<br />

Kawabata, H<br />

P1‐14‐08<br />

Kawai, M<br />

P6‐07‐27<br />

Kawai, N<br />

P3‐07‐10<br />

Kawai, Y<br />

P1‐15‐11<br />

Kawasaki, K<br />

P3‐07‐10<br />

Kay, C PD10‐02, P3‐06‐23, P6‐04‐09<br />

Kaye, P<br />

P5‐14‐04<br />

Kazmi, N<br />

PD03‐04<br />

Ke, C<br />

OT2‐3‐03<br />

Keam, B<br />

PD09‐05, P2‐05‐20<br />

Keane, A<br />

OT3‐4‐04<br />

Kearney, T<br />

P4‐16‐09<br />

Keating, NL<br />

P6‐08‐05<br />

Keegan, M P6‐11‐07, P6‐11‐09<br />

Keene, K<br />

P3‐03‐03<br />

Keene, KS P5‐17‐07, P6‐07‐37<br />

Keisch, M<br />

P4‐16‐03<br />

Kelleher, M<br />

P2‐10‐29<br />

Keller, L<br />

OT3‐2‐01<br />

Kelley, MC<br />

P4‐01‐03<br />

Kellokumpu‐Lehtinen, P‐L PD10‐06<br />

Kelly, CM<br />

P2‐10‐10<br />

Kelly, EM<br />

P5‐11‐02<br />

Kenis, C<br />

P6‐09‐07<br />

Kennedy, D<br />

P6‐08‐08<br />

Kennedy, MJ<br />

P6‐01‐02<br />

Kenney, K<br />

P4‐12‐04<br />

Kenny, K<br />

P3‐05‐05<br />

Kenny, PA<br />

P2‐04‐06<br />

Keogh, M<br />

P5‐10‐13<br />

Kerbrat, P<br />

S5‐3, PD07‐04,<br />

P1‐13‐04, P2‐13‐10<br />

Kere, D<br />

P1‐01‐21<br />

Kerin, M<br />

P3‐08‐04<br />

Kerin, MJ<br />

P1‐01‐15,<br />

P2‐05‐09, P5‐10‐07<br />

Kerlikowske, K<br />

P3‐02‐01,<br />

P4‐01‐15, P4‐13‐05<br />

Kern, P<br />

P2‐10‐14<br />

Kesari, S<br />

P3‐12‐04<br />

Kesarwala, AH P2‐11‐13, P2‐11‐14<br />

Keshtgar, M S4‐2<br />

Kesmodel, S<br />

P2‐13‐05<br />

Kessels, LW<br />

PD07‐06<br />

Keymeulen, KB<br />

P6‐08‐02<br />

Keymeulen, KBMI<br />

P5‐01‐13<br />

Khan, AJ<br />

P4‐16‐09<br />

Khan, J<br />

P4‐14‐13<br />

Khan, M<br />

P4‐16‐01<br />

Khan, QJ<br />

PD09‐02<br />

Khan, SA P1‐09‐07, P2‐10‐30,<br />

P3‐04‐08, P4‐12‐04<br />

Khanna, KK<br />

P6‐10‐06<br />

Khasanov, R S1‐4<br />

Khawaja, S<br />

P1‐01‐18<br />

Khayat, D S5‐3<br />

Khodari, W<br />

P4‐16‐10<br />

Khong, H<br />

P5‐18‐14<br />

Kieback, DG S1‐5<br />

Kiechle, M P5‐14‐06, P5‐15‐04<br />

Kiefer, A<br />

OT3‐4‐04<br />

Kieper, DA P4‐02‐09, P4‐02‐10<br />

Kiermaier, A S5‐1<br />

Kikuchi, M P2‐12‐13, P3‐06‐29<br />

Kil, WH<br />

P3‐11‐01<br />

Kilburn, L<br />

P1‐13‐03<br />

Kilburn, LS<br />

P2‐14‐01<br />

Killelea, B P3‐14‐05, P3‐14‐06<br />

Kim, A<br />

P5‐04‐07<br />

Kim, B<br />

P4‐01‐14<br />

Kim, BS<br />

P4‐03‐11<br />

Kim, CA<br />

P1‐05‐10<br />

Kim, D‐C<br />

PD02‐06<br />

Kim, EJ<br />

P4‐03‐01, P5‐04‐03<br />

Kim, E‐K<br />

P1‐01‐23<br />

Kim, H<br />

P5‐07‐03<br />

Kim, HH<br />

P3‐01‐02, P3‐12‐05<br />

Kim, HJ<br />

PD09‐05, P3‐01‐02<br />

Kim, HK<br />

P4‐02‐07<br />

Kim, J<br />

PD05‐07, P3‐11‐01<br />

Kim, J‐A<br />

PD09‐08, P6‐04‐25<br />

Kim, JE<br />

P3‐12‐05<br />

Kim, JH<br />

PD09‐05<br />

Kim, JS P1‐01‐08, P1‐14‐16, P5‐07‐04<br />

Kim, JW<br />

P5‐16‐06<br />

Kim, JY<br />

PD05‐07, P1‐01‐08,<br />

P1‐14‐16, P2‐10‐35<br />

Kim, M<br />

P2‐10‐41, P3‐11‐01<br />

Kim, MJ<br />

P1‐01‐23<br />

Kim, MK PD05‐07, P1‐01‐08, P1‐14‐16<br />

Kim, N<br />

OT1‐1‐01<br />

Kim, P S5‐7<br />

Kim, S<br />

PD05‐07, P2‐05‐20,<br />

P3‐08‐03, P3‐11‐01<br />

Kim, SB<br />

PD09‐05<br />

Kim, S‐B<br />

P3‐12‐05, P5‐18‐26<br />

Kim, SH<br />

P4‐03‐01<br />

Kim, SI<br />

P1‐01‐23<br />

Kim, S‐I<br />

P2‐05‐03<br />

Kim, SJ<br />

PD07‐05, P2‐10‐18<br />

Kim, SK<br />

P1‐10‐01<br />

Kim, S‐K<br />

P1‐01‐03<br />

Kim, S‐R S1‐10<br />

Kim, SS<br />

P3‐12‐05<br />

Kim, ST S1‐6<br />

Kim, SW<br />

P1‐01‐03<br />

Kim, S‐W<br />

P5‐04‐03<br />

Kim, S‐Y<br />

PD09‐05<br />

Kim, T<br />

PD05‐07, P1‐01‐08,<br />

P1‐14‐16, P3‐06‐16<br />

Kim, WW<br />

P1‐01‐24<br />

Kim, Y<br />

P4‐12‐02<br />

Kim, YW<br />

P1‐10‐01<br />

Kimbung, S<br />

P1‐05‐07<br />

Kimler, BF<br />

PD09‐02<br />

Kimmick, GG<br />

P6‐08‐05<br />

Kimura, K<br />

P3‐02‐02,<br />

P3‐09‐04, OT1‐1‐01<br />

King, DW<br />

P1‐01‐04<br />

King, J<br />

P4‐06‐04<br />

King, K‐L<br />

P3‐12‐10<br />

King, KS<br />

PD10‐05<br />

King, T<br />

P5‐18‐04<br />

King, TA<br />

PD04‐05, PD05‐02<br />

Kinneer, K<br />

P4‐07‐05<br />

Kinoshita, J<br />

OT1‐1‐01<br />

Kinoshita, N<br />

P1‐01‐27<br />

Kinoshita, T P1‐01‐12, P3‐02‐06<br />

Kirby, G<br />

OT3‐3‐01<br />

Kirkegaard, T<br />

P6‐04‐18<br />

Kirova, YM P1‐01‐19, P4‐13‐14,<br />

P5‐01‐05, P5‐21‐03<br />

Kirpotin, D<br />

P4‐02‐05<br />

Kirstein, L<br />

P4‐15‐03<br />

Kirwan, CC P1‐01‐26, P3‐14‐04<br />

Kishimoto, M<br />

P6‐07‐39<br />

Kiss, A<br />

P6‐08‐04<br />

Kitagawa, Y P1‐14‐09, P3‐06‐22,<br />

P4‐03‐03, P4‐09‐08<br />

Kitao, A<br />

P6‐07‐30<br />

Kitchens, C<br />

P6‐05‐12<br />

Kiyokawa, I<br />

P2‐10‐32<br />

Klauschen, F<br />

PD06‐01<br />

Kleer, CG<br />

P1‐04‐11<br />

Klein, ME<br />

P6‐07‐02<br />

Klein, P P3‐08‐08, P6‐07‐34, P6‐09‐08<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

596s<br />

<strong>Cancer</strong> Research


December 4-8, 2012<br />

Author Index<br />

Kleine‐Tebbe, A<br />

P2‐10‐25<br />

Kleinman, R<br />

P2‐11‐12<br />

Klein‐Szanto, AA<br />

P1‐05‐03<br />

Klemens, AE<br />

PD08‐04<br />

Klemp, JR PD09‐02, P2‐11‐17<br />

Klepczyk, LC<br />

P6‐07‐37<br />

Klesczewski, K<br />

P5‐20‐03<br />

Klifa, C<br />

P4‐01‐04<br />

Klimowicz, A<br />

P4‐09‐07<br />

Klimowicz, AC P1‐07‐10, P6‐07‐17<br />

Kline, E<br />

PD01‐07<br />

Klinge, CM<br />

P6‐04‐30<br />

Klint, L<br />

P6‐12‐01<br />

Klintman, M P2‐10‐19, P2‐10‐28<br />

Klotz, R<br />

P6‐01‐04<br />

Kluding, P<br />

P2‐11‐17<br />

Knauer, M<br />

S4‐3, P2‐10‐02<br />

Knight, R<br />

P3‐06‐33<br />

Knoblauch, N<br />

P5‐01‐03<br />

Knoop, A<br />

P4‐09‐04, P5‐15‐07<br />

Knott, A<br />

P5‐18‐01, P5‐18‐26<br />

Knowlton, NS<br />

P4‐12‐05<br />

Knutson, KL<br />

P5‐03‐07<br />

Kobayashi, N<br />

P3‐02‐05<br />

Kobayashi, Y<br />

P6‐04‐17<br />

Koboldt, DC S5‐6<br />

Kochi, M<br />

P1‐14‐12, P4‐01‐05<br />

Koczywas, M<br />

P2‐14‐08<br />

Kodack, DP<br />

P3‐12‐03<br />

Koechlin, A P4‐13‐07, P4‐13‐08<br />

Koehn, TM<br />

P2‐11‐15<br />

Koelbl, H<br />

P2‐10‐13<br />

Koenig, KB<br />

P1‐07‐09<br />

Koeppen, H<br />

P4‐07‐01<br />

Koestler, W<br />

P3‐06‐33<br />

Koffijberg, E<br />

P5‐15‐03<br />

Kofi, P<br />

P2‐05‐10<br />

Kogianni, G<br />

P2‐02‐03<br />

Koh, BS<br />

P3‐01‐02<br />

Koh, YW<br />

P2‐02‐02<br />

Kohn, E<br />

P2‐04‐08<br />

Kohno, N<br />

PD07‐05, P5‐18‐16<br />

Kohno, S<br />

P6‐07‐39<br />

Kojima, I<br />

P1‐01‐20<br />

Koka, M<br />

P2‐10‐40<br />

Kokufu, I<br />

P6‐07‐39<br />

Kölbl, AC<br />

P2‐01‐11<br />

Kolesar, JM<br />

P6‐12‐02<br />

Kolev, VN P6‐11‐07, P6‐11‐09<br />

Kolonel, LN<br />

P4‐12‐02<br />

Komenaka, IK<br />

PD08‐04<br />

Komiya, H<br />

P4‐15‐05<br />

Kondo, N P1‐02‐02, P3‐12‐06<br />

Kong, X<br />

P2‐09‐06<br />

Kong, Y<br />

P5‐10‐10<br />

König, K<br />

P6‐09‐08<br />

Königsberg, R<br />

P2‐13‐01<br />

Konishi, M<br />

PD07‐05<br />

Konno, H<br />

P6‐04‐17<br />

Konno, M<br />

P1‐07‐10,<br />

P4‐09‐07, P6‐07‐17<br />

Kontos, D<br />

P4‐01‐08<br />

Koo, JS<br />

P1‐01‐23, P2‐05‐03<br />

Koolen, BB<br />

P4‐02‐01<br />

Koornstra, RHT<br />

PD01‐03<br />

Köppler, H<br />

P2‐11‐11<br />

Korah, V<br />

P5‐13‐03<br />

Körber, W<br />

P1‐07‐16<br />

Kornaga, EN<br />

P1‐07‐10<br />

Kornak, J<br />

P4‐01‐04<br />

Kornblith, A<br />

P6‐08‐05<br />

Kornhauser, N<br />

P6‐11‐04<br />

Korth, CC<br />

P6‐11‐10<br />

Kosaka, Y<br />

P2‐12‐13<br />

Kosel, M<br />

PD10‐08<br />

Koslow, S<br />

PD04‐05<br />

Koslow Mautner, S<br />

PD05‐02<br />

Köstler, WJ<br />

P2‐10‐31<br />

Kotin, A<br />

OT3‐1‐01<br />

Kotoula, V<br />

P1‐07‐15<br />

Koukel, L<br />

PD10‐09<br />

Kounalakis, N<br />

P4‐04‐06<br />

Kourousis, C<br />

P1‐13‐09<br />

Kousaka, J<br />

P4‐03‐08<br />

Koutcher, JA<br />

P6‐01‐01<br />

Kovács, AK<br />

P3‐05‐08<br />

Kovács, G<br />

P4‐15‐01<br />

Kovacs, A<br />

P1‐05‐07<br />

Kovalenko, N<br />

OT1‐1‐17<br />

Kovatich, AJ<br />

P2‐05‐21,<br />

P3‐04‐06, P5‐01‐07<br />

Kowalski, M<br />

P5‐05‐04<br />

Krag, KJ<br />

PD09‐03, P2‐16‐04<br />

Kramar, A<br />

S5‐3, OT1‐2‐02<br />

Kranwinkel, G P2‐11‐03, P2‐12‐01<br />

Kranwinkle, G<br />

P1‐09‐02<br />

Krappmann, K<br />

P2‐10‐11<br />

Krase, P<br />

P5‐14‐06, P5‐15‐04<br />

Krasnozhon, D<br />

OT3‐3‐01<br />

Krause, S<br />

P2‐04‐03, P5‐05‐04<br />

Kreienberg, R<br />

S3‐4, P6‐09‐08<br />

Kreipe, HH<br />

P2‐10‐08<br />

Krekel, N<br />

PD04‐01<br />

Krekow, L<br />

P2‐05‐06<br />

Krenek, K<br />

P5‐07‐03<br />

Krishnamurthy, S P1‐07‐06, P2‐03‐01,<br />

P3‐10‐09, P3‐10‐10,<br />

P5‐03‐05, P5‐04‐06<br />

Krishnamurthy, SA<br />

P3‐10‐01<br />

Krishnan, SR<br />

PD09‐07<br />

Kristensen, VN<br />

P5‐05‐03<br />

Kristoffersson, U<br />

P2‐10‐37<br />

Krizman, D<br />

P6‐07‐17<br />

Krocker, J<br />

P1‐14‐05<br />

Kroemer, G<br />

P2‐08‐04<br />

Kroep, JR<br />

PD07‐06<br />

Krohn, KA<br />

P6‐04‐03<br />

Kroll, T<br />

P1‐07‐16<br />

Kronenwett, R S4‐3, P2‐10‐11<br />

Kronish, L<br />

P1‐12‐03<br />

Krontiras, H P3‐03‐03, P5‐17‐07<br />

Krop, I<br />

P3‐05‐03, P5‐18‐03<br />

Krop, IE<br />

P5‐18‐06, OT1‐1‐11<br />

Kruchevsky, N<br />

P6‐01‐01<br />

Kruitwagen, RF<br />

P6‐08‐02<br />

Krupa, J<br />

P4‐14‐14<br />

Kruper, L<br />

PD08‐02<br />

Kuba, MG<br />

S3‐6, P6‐10‐05<br />

Kuba, S<br />

P3‐06‐26<br />

Kubik‐Huch, RA<br />

P4‐03‐06<br />

Kubo, K<br />

P1‐14‐06<br />

Kubota, Y<br />

P1‐15‐11<br />

Kuchiki, M<br />

P3‐02‐03<br />

Kuchuk, I<br />

P2‐05‐13,<br />

P3‐13‐05, P5‐13‐01<br />

Kuderer, NM<br />

P1‐15‐04,<br />

P2‐10‐03, P3‐06‐07<br />

Kudla, A<br />

P1‐07‐03<br />

Kudo, Y<br />

P3‐13‐04<br />

Kuehn, T S2‐2<br />

Kuemmel, S<br />

S3‐1, P1‐14‐01,<br />

OT1‐1‐13, OT3‐3‐11<br />

Kuenen, MA<br />

P4‐11‐01<br />

Kuerer, HM S2‐3, P1‐07‐09, P4‐15‐02<br />

Kuffel, M<br />

PD10‐08<br />

Kuhn, W S3‐4<br />

Kuijpers, CC<br />

PD02‐03<br />

Kulka, J<br />

P3‐05‐08<br />

Kulma‐Kreft, M P3‐12‐09, P3‐13‐03<br />

Kulyk, SO S1‐6<br />

Kumaki, N<br />

P2‐05‐08<br />

Kumar, R<br />

P4‐08‐08<br />

Kumar, S<br />

P3‐07‐02<br />

Kumar, V<br />

PD01‐01, P6‐10‐03<br />

Kümmel, S PD07‐02, P1‐14‐05<br />

Kuo, S‐H<br />

P3‐06‐02<br />

Kuo, W‐H<br />

P4‐03‐02<br />

Kuranami, M<br />

P2‐12‐13<br />

Kurbacher, C<br />

S3‐4, P1‐15‐02<br />

Kurebayashi, J<br />

P2‐10‐18<br />

Kurian, AW P3‐11‐02, P4‐11‐02<br />

Kuriyama, N<br />

OT2‐3‐11<br />

Kurland, BF<br />

P6‐04‐03<br />

Kuroi, K P1‐12‐01, P1‐14‐02, P3‐02‐06,<br />

P3‐06‐18, P4‐14‐07, P6‐07‐16<br />

Kurokawa, M<br />

P1‐05‐03<br />

Kurosumi, M<br />

P1‐14‐06,<br />

P5‐01‐02, P6‐04‐17<br />

Kurozumi, S<br />

P1‐14‐06<br />

Kurzrock, R<br />

P5‐20‐09<br />

Kusche, M<br />

P1‐07‐16<br />

Kuy, C<br />

P4‐06‐08<br />

Kvale, EA<br />

P2‐12‐14<br />

Kvecher, L P2‐05‐21, P5‐01‐07<br />

Kwak, HY<br />

P4‐03‐01<br />

Kwan, M<br />

P3‐07‐05<br />

Kwan, W<br />

P4‐17‐08<br />

Kwasny, W<br />

P2‐10‐02<br />

Kwei, SL<br />

P4‐17‐02<br />

Kwiatkowski, F<br />

P3‐06‐20<br />

Kwok, S<br />

PD10‐03<br />

Kwon, DH<br />

P1‐05‐20<br />

Kwon, H‐C<br />

PD02‐06<br />

Kwon, Y<br />

P1‐01‐03<br />

Kwong, A<br />

P3‐11‐02,<br />

P3‐11‐03, P4‐11‐02<br />

Kyshtoobayeva, A P2‐10‐04, P3‐05‐05,<br />

P3‐05‐05, P3‐05‐06<br />

Laadem, A P3‐06‐33, P5‐20‐03<br />

Labbe‐Devilliers, C<br />

P3‐12‐01<br />

Laboy, J<br />

P5‐07‐06<br />

Labrie, F<br />

P2‐06‐03<br />

Lacerda, L PD03‐06, P5‐03‐05<br />

Lacevic, M<br />

P4‐14‐09<br />

Lackner, M<br />

P4‐06‐10<br />

Lacornerie, T<br />

OT1‐2‐02<br />

Lacroix‐Triki, M PD05‐08, P3‐04‐04,<br />

P3‐05‐07, P5‐21‐01,<br />

OT3‐3‐10<br />

Laé, M<br />

P5‐01‐05<br />

Laenen, A<br />

P6‐07‐29<br />

Lænkholm, A‐V<br />

P4‐09‐04<br />

Laeufle, R S6‐5<br />

Laferriere, J<br />

P6‐02‐03<br />

Lafon, D<br />

PD05‐04<br />

Lagier, R<br />

PD10‐03<br />

Lagrange, J‐L<br />

P4‐16‐10<br />

Lahogue, A<br />

OT1‐1‐16<br />

Lai, A<br />

P6‐04‐12<br />

Laidley, AL P1‐15‐09, P4‐14‐01<br />

Lake, D<br />

P1‐13‐11,<br />

P5‐18‐20, P5‐20‐07<br />

Lake, DF<br />

P1‐05‐04<br />

Lakhani, A<br />

PD10‐02<br />

Lakhani, SR<br />

P6‐10‐06<br />

Laki, F<br />

P1‐01‐19<br />

Lakomy, J P3‐12‐09, P3‐13‐03<br />

Lakshmaiah, K<br />

P5‐02‐02<br />

Lalami, Y<br />

P5‐18‐19<br />

Lalla, D<br />

P5‐18‐12, P6‐09‐11<br />

Lalloum, M<br />

P4‐13‐03<br />

Lam, C<br />

P1‐15‐07, P6‐11‐04<br />

Lam, EW‐F<br />

P6‐04‐16<br />

Lam, T<br />

P4‐17‐04<br />

Lamb, CA<br />

P4‐06‐15<br />

Lambaudie, E<br />

P5‐03‐01<br />

Lambrechts, D<br />

P3‐05‐03,<br />

P6‐07‐14, OT2‐2‐02<br />

Lambros, MBK<br />

PD05‐08<br />

Lamoureux, J<br />

PD06‐03<br />

Lampe, D<br />

P1‐14‐05<br />

Lanari, C<br />

P4‐06‐15<br />

Lanchbury, J<br />

PD09‐04<br />

Lanczky, A<br />

P1‐07‐14<br />

Landaverde, D<br />

P2‐12‐07<br />

Landberg, G<br />

PD04‐06<br />

Landis, M<br />

P5‐03‐03<br />

Landis, MD P2‐10‐06, P6‐11‐12<br />

Landis, S<br />

OT1‐1‐08<br />

Landsman‐Blumberg, P P2‐14‐07,<br />

P5‐20‐12<br />

Landstorfer, B<br />

P2‐10‐38<br />

Landua, J<br />

P4‐02‐08<br />

Landucci, E<br />

P5‐17‐06<br />

Lane, D<br />

PD03‐09<br />

Lane, J<br />

P6‐07‐41<br />

Lane, M<br />

P1‐15‐07<br />

Lane, ME<br />

P6‐11‐04<br />

Lang, I S1‐6, S3‐2, P5‐17‐03, P6‐07‐06<br />

Lang, JE<br />

P3‐10‐07<br />

Lang, K P3‐07‐06, P3‐07‐07, P3‐07‐14<br />

Langbaum, TS S3‐5<br />

Lange, C<br />

P1‐07‐11<br />

Langer, L<br />

P4‐11‐04<br />

Lanigan, C<br />

P6‐07‐45<br />

Lannin, D P3‐14‐01, P4‐01‐02<br />

Lannin, DR P3‐14‐05, P3‐14‐06<br />

Lansdown, M<br />

P4‐01‐14<br />

Lantzsch, T P1‐14‐05, P2‐10‐38<br />

Lapetino, S<br />

P1‐02‐01<br />

Lara, J<br />

P2‐10‐16<br />

Lara, JM<br />

OT2‐3‐10<br />

Lara, MA<br />

OT3‐3‐05<br />

Laredo, S<br />

P2‐12‐03<br />

Larionov, A P3‐06‐23, P6‐04‐06<br />

Larionov, AA P6‐04‐09, P6‐04‐10<br />

Laronga, C<br />

P1‐01‐13,<br />

P4‐14‐09, P4‐17‐01<br />

Larrea, F<br />

P4‐06‐19, P6‐04‐29<br />

Larriera, A<br />

P3‐05‐05<br />

Larsen, L<br />

P4‐01‐11<br />

Larsen, MR<br />

P3‐04‐05<br />

Larsimont, D S4‐4, S6‐2, P1‐07‐08,<br />

P3‐04‐10, P3‐05‐03<br />

Larson, R<br />

PD03‐06<br />

Larson, RA P5‐03‐05, P5‐03‐10<br />

Lartigau, E<br />

OT1‐2‐02<br />

Lasry, S<br />

P1‐01‐19<br />

Lassalle, M<br />

P3‐06‐24<br />

Latouche, A<br />

OT3‐3‐10<br />

Lau, A<br />

P5‐18‐20<br />

Laub, GA<br />

P4‐01‐10<br />

Launay‐Vacher, V<br />

P5‐17‐04<br />

Laurà, F<br />

P5‐17‐06<br />

Lavie, O<br />

P6‐14‐03<br />

Lavin, PT<br />

P5‐18‐07<br />

Law, FBF<br />

P3‐11‐02, P4‐11‐02<br />

Lawler, K<br />

P2‐10‐29<br />

Lawler, WE<br />

P2‐05‐06<br />

Lawton, P S4‐1<br />

Layman, RM<br />

P2‐11‐07<br />

Lazar, V<br />

P4‐09‐05<br />

Lazare, M<br />

P3‐05‐05<br />

Lazzeroni, M PD04‐07, PD06‐06<br />

Le Marchand, L<br />

P4‐12‐02<br />

Le Rhun, E P2‐13‐10, P3‐12‐01<br />

Le Ven, E<br />

P3‐06‐24<br />

Leach, MO<br />

P4‐11‐05<br />

Leader, JB<br />

P4‐13‐12<br />

Leary, A<br />

PD07‐07, P2‐12‐15<br />

Lebas, N<br />

PD07‐04<br />

Lebeau, A S2‐2<br />

LeBeau‐Grasso, L<br />

P3‐10‐07<br />

Lebrecht, A<br />

P2‐10‐13<br />

Lecuru, F<br />

P1‐01‐21<br />

Lee, A P4‐03‐11, P5‐10‐13, P6‐05‐12<br />

Lee, AS‐G<br />

P3‐08‐09<br />

Lee, B‐N P2‐10‐32, P5‐04‐06, P5‐10‐02<br />

Lee, C<br />

PD04‐03<br />

Lee, E<br />

P1‐01‐03<br />

Lee, EJ<br />

P6‐01‐05<br />

Lee, H<br />

P4‐02‐05<br />

Lee, HE<br />

P5‐04‐03<br />

Lee, H‐E<br />

P2‐05‐14<br />

Lee, HJ P2‐02‐02, P5‐04‐03, P6‐07‐28<br />

Lee, HM<br />

P2‐05‐14<br />

Lee, H‐S<br />

PD05‐07<br />

Lee, H‐W<br />

PD02‐06<br />

Lee, J<br />

P1‐01‐03, P2‐10‐39,<br />

P4‐03‐11, P6‐07‐40<br />

Lee, JE P2‐05‐20, P2‐10‐41, P3‐11‐01<br />

Lee, JH<br />

P1‐05‐19, P6‐01‐05<br />

Lee, J‐H PD02‐06, P1‐01‐05, P3‐03‐02<br />

Lee, JJ<br />

P1‐01‐24<br />

Lee, JW PD05‐07, P1‐01‐08, P1‐14‐16,<br />

P2‐02‐02, P3‐01‐02, P3‐01‐02,<br />

P4‐16‐14, P5‐18‐23<br />

Lee, K‐C<br />

PD02‐06<br />

Lee, KE<br />

PD09‐05<br />

Lee, K‐H<br />

PD09‐05, P6‐01‐05<br />

Lee, K‐J<br />

P6‐04‐12<br />

Lee, KS<br />

PD09‐05<br />

Lee, LB<br />

P4‐07‐01<br />

Lee, M<br />

P5‐10‐06<br />

Lee, MH<br />

PD09‐05<br />

Lee, M‐R<br />

PD02‐06<br />

www.aacrjournals.org 597s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Lee, O<br />

P1‐09‐07, P2‐10‐30,<br />

P3‐04‐08, P4‐12‐04<br />

Lee, S<br />

P1‐01‐03, P3‐09‐05,<br />

P4‐06‐13, OT1‐1‐16<br />

Lee, SA<br />

P2‐05‐14<br />

Lee, S‐E<br />

PD02‐06<br />

Lee, SJ<br />

P5‐04‐07, P5‐07‐04<br />

Lee, S‐J P1‐01‐05, P3‐03‐01, P3‐03‐02<br />

Lee, SK<br />

P2‐10‐41, P3‐11‐01<br />

Lee, SY<br />

P5‐14‐03<br />

Lee, W<br />

P2‐06‐05<br />

Lee, YH<br />

P1‐01‐24<br />

Lee, YR<br />

P5‐07‐04<br />

Lefebvre, C P1‐01‐21, OT2‐1‐01<br />

Lefeuvre‐Plesse, C<br />

P2‐13‐10<br />

Leger‐Falandry, C<br />

OT3‐3‐10<br />

Legrier, M‐E<br />

P3‐06‐24<br />

Leheurteur, M<br />

P1‐15‐08<br />

Lehman, HL P3‐10‐04, P3‐10‐08<br />

Lehman, R<br />

P1‐13‐11<br />

Lehmann, BD<br />

P6‐05‐03<br />

Lei, L<br />

P4‐05‐01, P4‐09‐09<br />

Lei, X<br />

P5‐20‐02<br />

Leighl, N<br />

P5‐18‐14<br />

Leinonen, M<br />

PD10‐06<br />

Leitch, AM S2‐1<br />

Leitch, M<br />

PD07‐01, P1‐07‐04<br />

Leite, AP<br />

P4‐01‐07<br />

Leitzel, K<br />

P2‐10‐16,<br />

P2‐10‐31, P3‐06‐33<br />

Lembersky, BC S1‐10<br />

Lemonier, J<br />

OT1‐2‐02<br />

Lemonnier, J PD07‐04, P3‐05‐07<br />

Lenhard, M<br />

P6‐07‐05<br />

Lentz, RB<br />

P4‐17‐02<br />

Leonardi, MC<br />

PD04‐07<br />

Leong, H<br />

P1‐05‐20<br />

Leong, L<br />

P2‐14‐08<br />

Leong, W<br />

PD03‐05<br />

Leong, WL<br />

P2‐10‐34<br />

Le‐Petross, HT S2‐1<br />

Lerebours, F<br />

PD07‐04<br />

Leroith, D<br />

P4‐13‐08<br />

Leth‐Larsen, R<br />

P3‐04‐05<br />

Letocha, H<br />

P2‐12‐09<br />

Leunen, K<br />

P2‐10‐12,<br />

P6‐05‐05, P6‐07‐29<br />

Leung, LY<br />

P6‐07‐31<br />

Leung, S<br />

P6‐07‐10<br />

Leung, SCY S4‐6<br />

Levine, EG<br />

P3‐07‐02<br />

Levine, M<br />

P1‐13‐01<br />

Levva, S<br />

P1‐07‐15<br />

Levy, C<br />

P1‐13‐04<br />

Levy, E<br />

P6‐07‐03<br />

Levy, R<br />

P4‐04‐01<br />

Lew, DL<br />

OT2‐2‐04<br />

Lewis, AL<br />

P4‐11‐06<br />

Lewis, CR<br />

PD04‐03<br />

Lewis, J<br />

P1‐05‐20<br />

Lewis, M<br />

P4‐02‐08, P5‐03‐08<br />

Leyland‐Jones, B<br />

S4‐4, S5‐2,<br />

PD10‐03, P5‐18‐02, P5‐19‐03,<br />

P6‐04‐11, OT2‐3‐11, OT3‐4‐04<br />

Leyvraz, S<br />

P6‐04‐05<br />

Lezon‐Geyda, K<br />

PD05‐05<br />

L’haridon, T<br />

OT3‐3‐04<br />

Li, B<br />

P6‐06‐06<br />

Li, C<br />

P6‐05‐10<br />

Li, F<br />

P4‐02‐08<br />

Li, G<br />

P1‐10‐02<br />

Li, H<br />

P5‐05‐04<br />

Li, J<br />

P3‐07‐08, P4‐04‐01,<br />

P4‐05‐01, OT2‐3‐04<br />

Li, JJ<br />

P3‐01‐05<br />

Li, JL<br />

P5‐03‐10<br />

Li, K<br />

P3‐06‐25<br />

LI, KK<br />

P2‐09‐06<br />

Li, L P2‐09‐06, P5‐04‐08, P5‐10‐10<br />

Li, R<br />

P6‐02‐02<br />

Li, S<br />

S5‐6, P2‐04‐02,<br />

P4‐07‐04, P4‐10‐02, P5‐03‐14<br />

Li, SP<br />

P1‐06‐01<br />

Li, W<br />

P4‐08‐10, P5‐04‐08<br />

Li, W‐P<br />

P2‐09‐05<br />

Li, X<br />

P1‐04‐11, P4‐01‐03,<br />

P5‐10‐10, P5‐20‐05<br />

Li, Y<br />

P5‐03‐14, P6‐07‐25,<br />

P6‐07‐37, P6‐10‐05<br />

Lian, Z‐Q<br />

P2‐09‐05<br />

Liao, J P1‐12‐02, P1‐13‐11, P5‐20‐04<br />

Liao, W‐L<br />

P1‐07‐19<br />

Lichinitser, MR S1‐4<br />

Lichtenegger, W<br />

P2‐01‐02<br />

Lie, Y P2‐05‐06, P2‐10‐16, P2‐10‐31<br />

Liede, A<br />

P2‐13‐03<br />

Liedtke, B<br />

P2‐10‐08<br />

Liedtke, C S1‐7, S2‐2, P2‐10‐08,<br />

P3‐06‐05<br />

Liefers, G‐J<br />

PD07‐06<br />

Lien, H‐C<br />

P3‐06‐02<br />

Liesche, F<br />

P2‐01‐11<br />

Lillemoe, HA<br />

P1‐03‐02<br />

Lim, E<br />

PD01‐08<br />

Lim, EA<br />

P4‐02‐07<br />

Lim, K<br />

P5‐20‐07<br />

Lim, M<br />

P6‐01‐03<br />

Lim, W<br />

P4‐03‐11<br />

Lim, YY<br />

P6‐06‐06<br />

Lima, DT<br />

P6‐07‐44<br />

Lima, E<br />

P4‐02‐03<br />

Limat, S<br />

P5‐18‐22<br />

Limentani, SA<br />

P5‐18‐05<br />

Lin, C‐H<br />

P3‐06‐02<br />

Lin, J<br />

P5‐12‐01<br />

Lin, MZ<br />

P5‐07‐05<br />

Lin, N OT1‐1‐07, OT1‐1‐11, OT2‐3‐06<br />

Lin, NU<br />

P2‐16‐04, P3‐12‐04,<br />

P5‐18‐03, P6‐08‐03<br />

Lin, Y<br />

P6‐07‐35<br />

Lina, A<br />

P2‐11‐16, P3‐06‐12<br />

Lind, P<br />

P5‐18‐18<br />

Lindemalm, C<br />

P2‐12‐09<br />

Linden, HM<br />

P6‐04‐03<br />

Linder, M<br />

OT2‐2‐05<br />

Linderholm, BK<br />

P6‐12‐01<br />

Lindh, J<br />

PD10‐09<br />

Lindquist, DL<br />

P3‐06‐12<br />

Lindsay, SP<br />

P2‐11‐09<br />

Lindström, LS<br />

PD06‐08<br />

Ling, H<br />

P6‐07‐42<br />

Ling, Z‐Q P2‐05‐05, P4‐09‐09<br />

Lingle, WL<br />

PD10‐04<br />

Lingsma, H<br />

P4‐11‐05<br />

Link, JM<br />

P6‐04‐03<br />

Linn, S<br />

P3‐06‐01<br />

Linn, SC PD01‐02, PD01‐03, P1‐15‐05,<br />

P6‐07‐05, OT2‐1‐02, OT2‐1‐03<br />

Linnartz, R<br />

P4‐08‐01<br />

Lintermans, A P2‐12‐06, P2‐13‐06,<br />

P5‐18‐19, OT2‐2‐02<br />

Lionaki, A<br />

P6‐07‐13<br />

Liotcheva, V<br />

P2‐10‐03<br />

Liotta, LA P3‐04‐01, P3‐04‐02<br />

Lippman, ME P1‐04‐06, P1‐05‐12<br />

Lipson, D<br />

PD02‐07, PD05‐01<br />

Lipson, JA<br />

P4‐01‐12<br />

Lipton, A<br />

P2‐10‐16,<br />

P2‐10‐31, P3‐06‐33<br />

Lissowska, J<br />

P3‐08‐02<br />

Litchfield, LM<br />

P6‐04‐30<br />

Litsas, G<br />

P5‐18‐03<br />

Little, JL<br />

P1‐05‐03<br />

Litwiniuk, M<br />

P2‐10‐31,<br />

P3‐12‐09, P3‐13‐03<br />

Liu, C S1‐8<br />

Liu, C‐Y<br />

P3‐12‐10, P6‐11‐05<br />

Liu, D<br />

PD03‐06<br />

Liu, F‐F<br />

P2‐10‐33<br />

Liu, G<br />

P1‐14‐17<br />

Liu, H<br />

P1‐12‐06, P4‐06‐03,<br />

P6‐04‐28, P6‐10‐04<br />

Liu, J P2‐11‐06, P5‐18‐20, P6‐06‐05<br />

Liu, JC<br />

P1‐04‐05<br />

Liu, L<br />

P4‐08‐03, P4‐08‐07<br />

Liu, MC PD09‐03, P2‐10‐01, P6‐05‐02<br />

Liu, M‐C<br />

P4‐16‐05<br />

Liu, P<br />

PD03‐08, P5‐10‐10<br />

Liu, Q S1‐10<br />

Liu, R<br />

P4‐05‐01<br />

Liu, S P2‐10‐02, P3‐06‐03, P6‐07‐25<br />

Liu, SX<br />

PD01‐08<br />

Liu, X P3‐05‐05, P4‐06‐08, P6‐11‐11<br />

Liu, Y<br />

P6‐07‐36<br />

Liu, Yb<br />

P6‐07‐36<br />

Liu, Z‐B<br />

P6‐07‐42<br />

Livartowski, A<br />

P4‐13‐14<br />

Llombart, A OT1‐1‐09, OT3‐3‐06<br />

Llop, JR<br />

P4‐06‐18<br />

Lluch, A<br />

OT2‐3‐06<br />

Lo, K‐Y<br />

P4‐04‐07<br />

Lo, S‐K<br />

P3‐08‐09<br />

Lo, SS<br />

PD10‐02, P1‐12‐04<br />

Lo Nigro, C<br />

P4‐06‐10<br />

Lobbes, M<br />

P3‐02‐08<br />

Lobbes, MBI<br />

P5‐01‐13<br />

Lobbezoo, DJA P5‐21‐04, P6‐07‐32<br />

Lobelle, J‐P<br />

P6‐09‐07<br />

Loddo, M<br />

PD04‐08<br />

Loessl, K<br />

P4‐15‐01<br />

Logan, LS<br />

P2‐10‐40<br />

Loghmani, A<br />

P2‐12‐10<br />

Logullo, AF<br />

P2‐05‐17<br />

Loh, S‐W<br />

P1‐01‐01<br />

Lohmann, AE<br />

P4‐16‐04<br />

Loi, S P1‐07‐08, P6‐07‐14, OT1‐1‐03<br />

Loibl, S<br />

S1‐7, S1‐11, S3‐1,<br />

S4‐5, PD06‐01,<br />

P1‐14‐01, P3‐06‐05, P6‐07‐05,<br />

OT1‐1‐13, OT2‐2‐05, OT3‐3‐11<br />

Lokeshwar, BL<br />

P5‐07‐01<br />

Loman, N<br />

PD03‐07,<br />

P1‐05‐07, P2‐10‐37<br />

Lombardo, Y P1‐04‐04, P4‐06‐12<br />

Lomo, L<br />

P3‐03‐01<br />

Longy, M<br />

PD05‐04<br />

Lønning, PE<br />

P6‐04‐06<br />

Loo, WTY<br />

P2‐05‐16<br />

Look, M<br />

P3‐06‐01<br />

Look, MP<br />

P6‐04‐08<br />

Lopez, AM<br />

P5‐12‐03<br />

Lopez, JJ<br />

P4‐17‐01<br />

Lopez, M<br />

P4‐13‐05<br />

Lopez, S<br />

P3‐03‐01<br />

Lopez Diaz, F<br />

P2‐06‐05<br />

López Díaz de Cerio, A P5‐16‐03<br />

Lopez‐Bujanda, Z<br />

P2‐02‐01<br />

Lopez‐Rios, F P1‐14‐10, P5‐18‐15<br />

Lord, CJ<br />

PD05‐08<br />

Lorenz, R<br />

P2‐01‐02<br />

Lories, R<br />

P2‐13‐06<br />

Lortholary, A<br />

S5‐3, P1‐13‐04<br />

Los, M<br />

P1‐12‐05<br />

Lou, C‐J<br />

P2‐05‐05<br />

Louie, B<br />

P6‐07‐31<br />

Louis, B<br />

P5‐21‐01<br />

Louis, E<br />

P5‐18‐19<br />

Loutfi, R<br />

P6‐10‐01<br />

Love, SM<br />

P2‐15‐01<br />

Lower, EE<br />

P6‐08‐08<br />

Lu, D<br />

P5‐18‐24<br />

Lu, H<br />

OT3‐1‐02<br />

Lu, J<br />

P2‐11‐07, P5‐10‐15,<br />

P6‐07‐35, OT1‐1‐12<br />

Lu, JS<br />

P3‐01‐05<br />

Lu, ML<br />

P5‐06‐01<br />

Lu, N<br />

P6‐04‐12<br />

Lu, S<br />

P6‐04‐28<br />

Lu, Y<br />

P4‐08‐05, P5‐10‐03<br />

Lu, Y‐S<br />

P3‐06‐02, P6‐05‐15<br />

Lubet, R<br />

P1‐08‐02<br />

Lubet, RA<br />

P1‐08‐01<br />

Lucas, B<br />

OT3‐3‐04<br />

Lucci, A<br />

P1‐01‐06, P2‐03‐01<br />

Lueck, H‐J S3‐4<br />

Luengo Gil, G<br />

P3‐06‐15<br />

Lum, L<br />

P4‐06‐06<br />

Lundgren, S<br />

P6‐09‐03<br />

Luo, AZ<br />

P4‐06‐03<br />

Luo, C<br />

P1‐05‐09, P2‐05‐21,<br />

P2‐09‐06, P3‐04‐06, P3‐09‐03<br />

Luo, J<br />

PD03‐09<br />

Luo, L<br />

P2‐09‐06<br />

Luo, R<br />

PD07‐01<br />

Luo, Y<br />

PD02‐04<br />

Luque, M P1‐07‐18, P2‐01‐06<br />

Lurie, G<br />

P4‐12‐02<br />

Lurkin, I<br />

P3‐06‐01<br />

Lurvey, HL<br />

P2‐04‐03<br />

Lush, RM<br />

P2‐14‐04<br />

Lussiez, A<br />

P3‐12‐03<br />

Lustberg, MB P2‐11‐04, P2‐11‐07<br />

Luthra, R<br />

P5‐04‐06<br />

Luthra, S<br />

P6‐04‐20<br />

Luu, T<br />

P2‐14‐08<br />

Luus, L<br />

P1‐07‐03<br />

Luu‐The, V<br />

P2‐06‐03<br />

Lux, MP<br />

PD07‐02<br />

LUX‐<strong>Breast</strong> 1 Study Group OT1‐1‐16<br />

LUX‐<strong>Breast</strong> 2 Study Group OT1‐1‐17<br />

Ly, A<br />

OT1‐1‐11<br />

Lyden, M<br />

P1‐15‐09, P4‐16‐03<br />

Lyerly, HK<br />

P4‐08‐07<br />

Lykkesfeldt, AE<br />

P6‐04‐18<br />

Lyman, GH P1‐15‐04, P2‐10‐03,<br />

P3‐06‐07<br />

Lyng, M S4‐4<br />

Lyng, MB<br />

P4‐09‐04<br />

Ma, C<br />

PD07‐01<br />

Ma, ESK<br />

P3‐11‐02, P4‐11‐02<br />

Ma, J<br />

P4‐02‐08<br />

Ma, Q<br />

P4‐03‐12<br />

Ma, X‐J<br />

PD02‐04<br />

Ma, Y<br />

P4‐06‐08<br />

Maartense, E PD07‐06, P1‐12‐05<br />

MacBeath, G<br />

P1‐07‐03<br />

MacGrogan, G<br />

P3‐14‐03<br />

Machado, GDP<br />

P2‐12‐04<br />

Machiels, J‐P P3‐04‐04, P5‐18‐19<br />

Maciel, MdS<br />

P4‐02‐03<br />

Mackay, A<br />

PD05‐08<br />

Mackey, J<br />

S6‐5, P3‐06‐34<br />

Mackie, D<br />

P2‐12‐15<br />

MacNeill, F P1‐01‐10, P1‐01‐11<br />

Macpherson, IR<br />

P1‐05‐13<br />

Madan, A<br />

P3‐14‐01<br />

Madan, RA<br />

P5‐16‐06<br />

Madani, S<br />

P4‐06‐16<br />

Madaras, L<br />

P3‐05‐08<br />

Madden, S P3‐04‐12, P4‐09‐06<br />

Madeira, KP P5‐01‐15, P6‐11‐13<br />

Madhukumar, P<br />

P3‐08‐09<br />

Madlensky, L<br />

P4‐13‐13<br />

Maekawa, Y<br />

P6‐07‐30<br />

Maetens, M<br />

S6‐2, P1‐07‐08<br />

Maffuz‐Aziz, A<br />

P5‐10‐12<br />

Magbanua, M<br />

P2‐05‐01<br />

Magbanua, MJM<br />

P2‐01‐05<br />

Magee, B S4‐1<br />

Maggi, R<br />

P3‐10‐03<br />

Magliocco, A P2‐10‐09, P4‐09‐07<br />

Magliocco, AM P1‐07‐10, P6‐07‐17<br />

Mahajan, S<br />

P4‐02‐02<br />

Mahesh, S<br />

P6‐07‐41<br />

Maheswaran, S<br />

P2‐01‐14<br />

Mahier Aït‐Oukhatar, C P3‐12‐01<br />

Mahler, L<br />

P6‐08‐06<br />

Mahmood, N<br />

P6‐07‐13<br />

Mahtani, RL<br />

P1‐12‐03<br />

Mai, H<br />

PD10‐02<br />

Mai, J<br />

P6‐13‐02<br />

Maibach, R<br />

S3‐2, P6‐07‐06<br />

Maillart, P<br />

P1‐14‐21<br />

Mailliez, A P2‐13‐10, P6‐10‐02<br />

Maisongrosse, V<br />

P3‐05‐07<br />

Maity, S<br />

P4‐01‐02<br />

Majewska, H P3‐12‐09, P3‐13‐03<br />

Majjaj, S<br />

S6‐2, P1‐07‐08,<br />

P3‐04‐10, P6‐07‐14<br />

Makariou, E<br />

P1‐09‐04<br />

Makey, KL<br />

P4‐06‐04<br />

Makhoul, I<br />

P3‐06‐30<br />

Makowski, L<br />

P6‐02‐09<br />

Makrantonakis, P<br />

P1‐13‐09<br />

Makris, A P1‐06‐01, P3‐06‐14<br />

Mala, C<br />

P5‐20‐01<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

598s<br />

<strong>Cancer</strong> Research


December 4-8, 2012<br />

Author Index<br />

Malamos, N<br />

P1‐13‐09<br />

Malamud, S<br />

P3‐08‐08<br />

Malfoy, B<br />

P5‐01‐05<br />

Malhotra, S<br />

P6‐08‐01<br />

Malinina, I<br />

P4‐04‐05<br />

Mallick, R<br />

P2‐05‐12,<br />

P2‐05‐13, P3‐13‐05<br />

Mallon, E S1‐1<br />

Mallon, P<br />

P1‐01‐19<br />

Malmberg, M<br />

P1‐05‐07<br />

Malmström, P P2‐10‐07, P2‐10‐19<br />

Malmtröm, P<br />

P2‐10‐28<br />

Maluta, S<br />

P4‐16‐08<br />

Maly, J<br />

P5‐18‐17<br />

Mamet, R<br />

P1‐01‐13<br />

Mammen, JMW<br />

PD09‐02<br />

Mamounas, E S1‐11<br />

Mamounas, EP S1‐10, S3‐2, S5‐5,<br />

P6‐07‐06, OT1‐2‐01, OT2‐2‐04<br />

Manasova, D<br />

P2‐01‐12<br />

Manasseh, DME<br />

P4‐14‐01<br />

Mandelblatt, J<br />

P6‐08‐05<br />

Manders, P<br />

P4‐11‐05<br />

Mangelsdorf, D<br />

P4‐08‐09<br />

Mani, A<br />

P1‐12‐03<br />

Maniar, T<br />

OT2‐3‐03<br />

Manié, E<br />

P5‐02‐03<br />

Manikhas, A<br />

OT3‐3‐01<br />

Mann, M<br />

PD09‐07, P1‐05‐10<br />

Mansfield, AS<br />

PD02‐05<br />

Manso, L<br />

P1‐07‐18, P2‐01‐06<br />

Manson, J<br />

PD03‐09<br />

Manthey, E<br />

P4‐04‐06<br />

Mañu, A<br />

P5‐03‐07<br />

Manuel, D<br />

P4‐01‐14<br />

Mao, JJ<br />

P4‐13‐01<br />

Marangoni, E<br />

P6‐04‐14<br />

Marchal, F P1‐01‐21, OT2‐1‐01<br />

Marciai, N<br />

P4‐16‐08<br />

Marcom, K<br />

P4‐13‐06<br />

Marcom, P<br />

P1‐01‐13<br />

Marcom, PK P1‐13‐05, P2‐10‐03,<br />

P3‐06‐07, P4‐09‐01<br />

Marcos‐Gragera, R<br />

P4‐09‐11<br />

Mardiak, J P2‐01‐12, P5‐02‐02<br />

Mardis, E<br />

P6‐07‐10<br />

Mardis, ER<br />

S5‐6, P2‐10‐01<br />

Margelí, M P1‐07‐18, P2‐01‐06<br />

Marginean, EC<br />

P5‐01‐01<br />

Margolin, S<br />

PD10‐09<br />

Margossian, AL P2‐16‐02, P2‐16‐03<br />

Margossian, J<br />

P2‐16‐03<br />

Margossian, ML<br />

P2‐16‐02<br />

Markiewicz, A<br />

P5‐04‐01<br />

Markmann, S<br />

P2‐10‐08<br />

Markopoulos, C S1‐5<br />

Marks, J<br />

P4‐13‐06<br />

Marme, F P3‐06‐08, P3‐06‐19<br />

Maroske, M<br />

P2‐13‐01<br />

Márquez‐Garbán, DC PD03‐04<br />

Marre, A<br />

P1‐13‐04<br />

Marrero, N<br />

OT1‐1‐08<br />

Marshall, D<br />

P5‐18‐14<br />

Marshall, HC S6‐4<br />

Martel, C<br />

P2‐06‐03<br />

Martens, J P3‐06‐32, P3‐06‐35<br />

Martin, A‐L PD07‐04, P1‐13‐04,<br />

P3‐04‐04, P3‐05‐07<br />

Martin, FM<br />

P4‐09‐06<br />

Martin, JL<br />

P5‐07‐05<br />

Martin, L<br />

PD03‐09<br />

Martin, L‐A<br />

PD01‐05<br />

Martin, M S1‐4, S1‐7, S3‐2, P2‐10‐11,<br />

P5‐18‐05, P6‐07‐06, OT2‐3‐03<br />

Martin, PM<br />

P6‐14‐04<br />

Martin, P‐M<br />

P1‐13‐13<br />

Martinez, G<br />

P5‐01‐09<br />

Martinez, ME<br />

PD08‐04<br />

Martínez, N P1‐14‐10,OT3‐3‐05<br />

Martínez, P P1‐07‐18, P2‐01‐06<br />

Martínez‐Climent, JA P5‐03‐07<br />

Martinez, R<br />

P5‐07‐06<br />

Martino, S<br />

S5‐5, P2‐10‐01<br />

Martinson, H<br />

P4‐04‐03<br />

Marty, M P1‐14‐03, P1‐14‐18,<br />

P2‐01‐04, P3‐06‐04<br />

Marzec, KA<br />

P5‐07‐05<br />

Marzolini, S<br />

P2‐12‐03<br />

Masaquel, A<br />

P5‐18‐12<br />

Masedunskas, A<br />

P1‐05‐23<br />

Maskarinec, G<br />

P4‐12‐02<br />

Mason, C<br />

P5‐10‐13<br />

Massacesi, C OT2‐3‐08, OT2‐3‐09<br />

Massaguer, A<br />

P6‐11‐02<br />

Massarut, S<br />

S4‐2, P5‐10‐17<br />

Massarweh, SA<br />

P2‐14‐05<br />

Mastropasqua, MG S4‐6<br />

Masuda, H PD03‐06, PD03‐08,<br />

P3‐10‐05, P5‐03‐09<br />

Masuda, M<br />

P1‐01‐20<br />

Masuda, N P1‐12‐01, P1‐14‐02,<br />

P1‐14‐08, P2‐10‐18,<br />

P3‐02‐06, P3‐06‐18,<br />

P5‐10‐09, P5‐18‐16, P6‐07‐16<br />

Masuda, S P2‐10‐18, P5‐01‐02<br />

Masudi, T<br />

P3‐01‐06<br />

Masumoto, N<br />

P6‐07‐26<br />

Mathieson, T<br />

P1‐03‐02<br />

Mathieu, MC P3‐06‐04, P3‐14‐03<br />

Mathieu, M‐C<br />

P1‐14‐03,<br />

P1‐14‐18, P4‐12‐01<br />

Matielle, CR<br />

P1‐01‐28<br />

Matsubara, M<br />

P5‐18‐16<br />

Matsui, H<br />

P2‐06‐01<br />

Matsumoto, H<br />

P1‐14‐06<br />

Matsumoto, K<br />

P6‐07‐30<br />

Matsumoto, M<br />

P1‐01‐27<br />

Matsunami, N<br />

P1‐14‐08<br />

Matsunuma, R<br />

P4‐03‐05<br />

Matsutani, Y<br />

P3‐13‐04<br />

Matsuura, H<br />

P1‐01‐20<br />

Matsuzu, K<br />

P1‐01‐20<br />

Matta, J<br />

P5‐07‐06<br />

Mattar, A<br />

P2‐05‐17<br />

Matthew, MK<br />

P4‐17‐02<br />

Matthews, A<br />

S4‐2, PD04‐03<br />

Matuoka, J<br />

P3‐07‐10<br />

Matuschek, C<br />

P1‐14‐04<br />

Matzkowitz, AJ<br />

P6‐13‐01<br />

Maurer, M PD03‐03, P1‐09‐02,<br />

P2‐11‐03, P2‐12‐01,<br />

P4‐02‐07, P6‐12‐03, OT2‐3‐06<br />

Mauriac, L<br />

P1‐13‐04<br />

Mavria, G<br />

P6‐02‐06<br />

Mavroudis, D P1‐13‐09, P2‐01‐03,<br />

P2‐01‐07<br />

Maximov, PY<br />

P2‐13‐09<br />

Maxwell, CA<br />

P2‐07‐01<br />

May, M<br />

P2‐16‐03<br />

Mayadev, JS<br />

P4‐01‐13<br />

Maybody, M<br />

OT3‐1‐01<br />

Mayer, EL<br />

PD09‐03,<br />

P6‐11‐06, OT2‐3‐11<br />

Mayer, IA<br />

P2‐14‐04,<br />

P4‐01‐03, P6‐10‐05<br />

Mayer, JA<br />

P2‐01‐10<br />

Mazanet, R<br />

P5‐18‐07<br />

Mazouni, C<br />

P4‐12‐01,<br />

P4‐14‐12, P5‐13‐02<br />

Mazzarello, S<br />

P2‐05‐12,<br />

P2‐05‐13, P3‐13‐05<br />

McArthur, HL P5‐20‐07, OT3‐1‐01<br />

McBride, ML<br />

PD04‐02<br />

McBryan, J<br />

P6‐04‐21<br />

McCall, L<br />

S2‐1, PD07‐01<br />

McCall, LM<br />

P1‐01‐06<br />

McCanna, S<br />

OT2‐3‐01<br />

McCartan, D<br />

P6‐04‐01<br />

McCarthy, B<br />

P2‐11‐07<br />

McCormick, DT<br />

OT2‐1‐04<br />

McCready, D<br />

P2‐10‐34<br />

McCready, DR PD03‐05, P2‐10‐33<br />

McCulloch, C<br />

P3‐05‐06<br />

McCurtin, RE<br />

P1‐13‐08<br />

McDaniel, RE<br />

P2‐13‐09<br />

McDermott, AM<br />

P5‐10‐07<br />

McDonagh, C<br />

P1‐07‐03<br />

McDonald, K<br />

P4‐08‐01<br />

McDonnell, SK<br />

PD10‐05<br />

McGarrigle, SA<br />

P6‐01‐02<br />

McGee, SF<br />

P4‐09‐06<br />

McGinness, MK<br />

PD09‐02<br />

McIlroy, M<br />

P6‐04‐04<br />

McKeegan, E<br />

OT2‐3‐07<br />

McLachlan, SA<br />

P4‐13‐10<br />

McLaughlin, SA S2‐1<br />

McLeod, HL<br />

PD10‐07<br />

McLinden, M<br />

P1‐13‐03<br />

McMahon, D<br />

P6‐12‐03<br />

McMahon, S<br />

P5‐16‐06<br />

McManus, J<br />

P1‐05‐21<br />

McMurray, HR<br />

P4‐06‐18<br />

McNagny, KM<br />

P1‐05‐05<br />

McNally, S<br />

P4‐09‐06<br />

McNaughton, K<br />

P6‐02‐09<br />

McShane, LM S4‐6<br />

McTiernan, A<br />

P3‐01‐01<br />

Means‐Powell, J<br />

P4‐01‐03<br />

Means‐Powell, JA<br />

P2‐14‐04<br />

Mebis, J<br />

P5‐18‐19<br />

Medina, H<br />

P6‐04‐29<br />

Meerbrey, KL<br />

PD01‐01<br />

Meershoek‐Klein<br />

Kranenbarg, EM<br />

PD07‐06<br />

Mege, J‐P<br />

OT1‐2‐02<br />

Mego, M<br />

P2‐01‐12<br />

Mehta, A OT1‐1‐16, OT1‐1‐17<br />

Mehta, K<br />

S1‐7, S1‐11,<br />

S3‐1, P1‐14‐01<br />

Mehta, S P1‐06‐01, P4‐01‐01, P6‐09‐06<br />

Mei, Q<br />

P2‐07‐01<br />

Meijer, ME<br />

P6‐04‐08<br />

Meijer, S<br />

PD04‐01<br />

Meinerz, W<br />

P1‐13‐13<br />

Meinhold‐Heerlein, I P3‐06‐05<br />

Meisner, C<br />

P1‐13‐13<br />

Melcher, CA<br />

P2‐01‐02,<br />

P4‐13‐11, OT1‐1‐10<br />

Meliadò, G<br />

P4‐16‐08<br />

Melichar, B<br />

P6‐10‐02<br />

Melin, S<br />

P2‐12‐11<br />

Melisko, M<br />

P2‐12‐11<br />

Melisko, ME P2‐12‐12, P4‐16‐07,<br />

P6‐10‐01, OT1‐1‐11<br />

Mellemkjær, L<br />

P1‐09‐01<br />

Mellgren, G<br />

P6‐04‐06<br />

Mena, M<br />

P5‐03‐07<br />

Meng, X<br />

OT1‐1‐05<br />

Menzin, J<br />

P3‐07‐06,<br />

P3‐07‐07, P3‐07‐14<br />

Mercier Blas, A<br />

OT3‐3‐04<br />

Meredith, R<br />

P3‐03‐03<br />

Meredith, RF P5‐17‐07, P6‐07‐37<br />

Meric‐Bernstam, F P1‐07‐06, P1‐07‐09,<br />

P6‐11‐03, P6‐11‐11<br />

Mery, E<br />

P3‐05‐07, P3‐14‐03<br />

Meschino, W<br />

P3‐02‐10<br />

Mesleard, C<br />

P3‐05‐07<br />

Messer, K<br />

P2‐06‐01<br />

Meszoely, IM<br />

P4‐01‐03<br />

Metaxas, M S4‐2<br />

Metellus, P<br />

P3‐12‐11<br />

Metz, WL<br />

P1‐01‐04<br />

Metzger, O S1‐1, P3‐05‐03, P5‐18‐03<br />

Meyer, ME<br />

P4‐01‐09<br />

Meyers, M<br />

P5‐20‐05<br />

Meyn, A<br />

P6‐11‐12<br />

Miao, RY<br />

P3‐10‐09<br />

Michaelson, R<br />

P2‐10‐16<br />

Michiels, S<br />

S4‐4, P1‐07‐08,<br />

P3‐04‐10, P3‐05‐03,<br />

P4‐09‐05, P6‐07‐14, P6‐07‐22<br />

Miele, L P2‐14‐04, P4‐06‐04, P6‐04‐22<br />

Migdady, Y<br />

P6‐07‐34<br />

Mignot, L<br />

OT3‐4‐06<br />

Mijonnet, S PD07‐04, OT1‐2‐02<br />

Mikami, Y P5‐01‐02, P5‐10‐09<br />

Miki, M<br />

P6‐07‐30<br />

Miki, Y<br />

P6‐05‐14<br />

Milani, M<br />

PD07‐03<br />

Milbourn, DE<br />

P1‐01‐17<br />

Miles, AK P6‐07‐09, P6‐07‐18<br />

Miles, D<br />

P2‐12‐07, P5‐16‐01,<br />

P5‐18‐01, OT1‐1‐02, OT1‐1‐03<br />

Miles, J<br />

P2‐05‐10<br />

Milián, ML<br />

OT3‐4‐04<br />

Milisen, K<br />

P6‐09‐07<br />

Millen, EC P2‐05‐17, P3‐11‐04<br />

Miller, A<br />

P3‐07‐02<br />

Miller, CL P6‐09‐04, OT3‐2‐02<br />

Miller, DV<br />

P2‐10‐27<br />

Miller, K<br />

P3‐12‐04, OT2‐3‐11<br />

Miller, KD P5‐17‐01, P5‐18‐13<br />

Miller, N<br />

P5‐10‐07<br />

Miller, NA<br />

P2‐10‐33<br />

Miller, P<br />

P1‐04‐06, P5‐10‐08<br />

Miller, SM<br />

P6‐09‐05<br />

Miller, V<br />

PD02‐07<br />

Miller, VA S3‐6<br />

Millham, R<br />

OT2‐3‐02<br />

Millholland, RJ<br />

P6‐12‐02<br />

Mills, G<br />

P1‐07‐06<br />

Mills, GB PD01‐01, P3‐06‐06,<br />

P4‐08‐05, P5‐10‐03<br />

Mills, J S4‐1<br />

Milne, RL<br />

P4‐13‐10<br />

Milosavljevic, A<br />

P6‐05‐12<br />

Miltenburg, NC<br />

P1‐15‐05<br />

Mimoto, R P4‐03‐10, P5‐04‐02<br />

Min, S<br />

P4‐03‐11<br />

Min, S‐H<br />

P2‐11‐05<br />

Minami, Y<br />

P6‐07‐27<br />

Minarik, G<br />

P2‐01‐12<br />

Minca, E<br />

P6‐07‐45<br />

Minna, J<br />

P4‐08‐09<br />

Minton, SE<br />

P2‐14‐04<br />

Mir, O<br />

P5‐17‐04<br />

Mira, A<br />

P2‐16‐02<br />

Miranda, M<br />

P6‐10‐06<br />

Miro, C<br />

P5‐18‐15<br />

Miron‐Shatz, T<br />

P4‐11‐03<br />

Mirza, FN<br />

P2‐13‐08<br />

Miskimen, K<br />

PD05‐05<br />

Misra, V<br />

P2‐14‐06<br />

Mitchell, EP P5‐01‐07, P5‐14‐03<br />

Mitchell, L<br />

OT1‐1‐02<br />

Mitchell, SD<br />

P4‐14‐01<br />

Mitchell, T<br />

PD01‐01<br />

Mitchison, TJ<br />

P6‐06‐02<br />

Mitra, S P3‐06‐06, P5‐03‐12, P6‐02‐07<br />

Mitsiades, N<br />

P6‐10‐03<br />

Mitsopoulos, C<br />

PD05‐08<br />

Mittal, V<br />

P6‐11‐04<br />

Mittempergher, L<br />

P4‐09‐05<br />

Mittendorf, EA S2‐1, P1‐01‐06,<br />

P1‐07‐06, P1‐07‐09, P5‐16‐01,<br />

P5‐16‐02, P5‐16‐05, P5‐20‐02<br />

Miura, D<br />

PD07‐05<br />

Miura, N<br />

P4‐04‐08<br />

Miura, T<br />

P2‐10‐32<br />

Miyajima, S<br />

P3‐02‐05<br />

Miyamoto, R<br />

OT1‐1‐01<br />

Miyamoto, Y<br />

P4‐03‐10<br />

Miyashita, M PD07‐05, P1‐06‐02,<br />

P6‐07‐39<br />

Miyazaki, T<br />

P6‐04‐27<br />

Miyoshi, Y<br />

P1‐14‐08,<br />

P5‐18‐16, P6‐07‐39<br />

Mizoo, T<br />

P3‐07‐10<br />

Mizota, Y<br />

P2‐11‐02<br />

Mizuno, H<br />

P4‐03‐07<br />

Mizuno, Y<br />

PD06‐02<br />

Mlineritsch, B P2‐10‐02, P5‐20‐11<br />

Modesto, MG<br />

P1‐01‐28<br />

Modi, S<br />

P5‐18‐04, P5‐18‐20<br />

Modzlewski, R<br />

P1‐15‐08<br />

Moebus, V S3‐4<br />

Moelans, CB<br />

PD02‐03,<br />

P2‐06‐02, P6‐05‐13<br />

Moeller, M<br />

P3‐04‐05<br />

Moerman, P<br />

P2‐10‐12,<br />

P6‐05‐05, P6‐07‐29<br />

Mögele, M<br />

P2‐12‐06<br />

Mohamed, MM<br />

P1‐05‐11<br />

Mohammed, H<br />

P3‐04‐03<br />

Mohar, A<br />

PD08‐06<br />

www.aacrjournals.org 599s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Mohebtash, M<br />

P5‐16‐06<br />

Mokbel, K P1‐04‐03, P1‐04‐07,<br />

P1‐04‐08, P1‐04‐09, P6‐05‐11<br />

Molina, JR<br />

P3‐06‐06<br />

Momen, L<br />

P6‐05‐02<br />

Monsey, J S5‐6<br />

Montcuquet, P<br />

P5‐18‐22<br />

Monteiro, SO<br />

P6‐07‐44<br />

Montero, AJ<br />

P1‐12‐03<br />

Montero, JC P4‐06‐17, P6‐05‐04<br />

Monville, F<br />

P5‐03‐01<br />

Moon, B‐I<br />

P4‐03‐11<br />

Moon, HG<br />

P1‐01‐08<br />

Moon, H‐G PD05‐07, P1‐14‐16,<br />

P1‐14‐19, P2‐10‐35,<br />

P3‐06‐16, P5‐18‐23<br />

Moore, A<br />

P1‐15‐07, P6‐11‐04<br />

Moore, C<br />

P6‐08‐01<br />

Moore, D<br />

P2‐10‐22<br />

Moore, DA<br />

P3‐06‐13<br />

Moore, HM<br />

P1‐04‐11<br />

Moraes, E<br />

OT1‐1‐08<br />

Morales, JC<br />

P6‐05‐04<br />

Morales, S S1‐7<br />

Moran, A<br />

P3‐08‐01<br />

Moran, CJ<br />

P4‐01‐06<br />

Moraras, N<br />

P5‐14‐04<br />

Morden, JP S3‐3<br />

Moreau, F<br />

P3‐04‐04<br />

Moreau, M<br />

P4‐02‐06<br />

Moreira, A<br />

P1-15-03<br />

Morelli, D<br />

P4‐06‐09<br />

Moreno, L<br />

P5‐13‐03<br />

Moreno‐Aspitia, A<br />

P1‐12‐06<br />

Morere, J‐F PD08‐03, P5‐17‐04<br />

Morgan, D S4‐1<br />

Morgan, SC<br />

P4‐16‐06<br />

Mori, M<br />

P5‐05‐01<br />

Mori, T<br />

P1‐15‐11<br />

Moriguchi, Y<br />

P3‐13‐04<br />

Morimoto, T<br />

P1‐14‐02<br />

Morioka, T<br />

P3‐13‐02<br />

Moris, F<br />

P4‐06‐17<br />

Morita, S<br />

P1‐14‐08<br />

Moroso, S<br />

P5‐01‐10<br />

Morris, D<br />

P6‐07‐17<br />

Morris, DG<br />

P1‐07‐10<br />

Morris, E<br />

OT3‐1‐01<br />

Morris, J<br />

P3‐01‐04<br />

Morris, PG<br />

P5‐20‐07<br />

Morrison, L<br />

P6‐07‐45<br />

Morrison, MM<br />

P5‐19‐01<br />

Morrogh, M<br />

PD05‐02<br />

Morrow, M PD04‐05, OT3‐1‐01<br />

Mortimer, JE P2‐05‐10, P2‐14‐08<br />

Morton‐Gittens, JA<br />

P4‐17‐06<br />

Mose, LE S6‐1<br />

Moseley, P P6‐07‐09, P6‐07‐18<br />

Moses, HL<br />

P3‐05‐09<br />

Motahari, B<br />

P5‐09‐01<br />

Motion, J<br />

P1‐09‐05<br />

Motoki, T<br />

P3‐07‐10<br />

Motoyoshi, A<br />

P4‐15‐05<br />

Motsinger‐Reif, AA<br />

PD10‐07<br />

Moulder, S<br />

P5‐20‐09<br />

Moulis, S<br />

P1‐07‐03<br />

Moulton, B<br />

OT1‐1‐06<br />

Mouret‐Fourme, E<br />

PD07‐04<br />

Mouret‐Reynier, M‐A PD07‐04,<br />

P1‐13‐04, P3‐06‐20<br />

Mouri, Y<br />

P4‐03‐08<br />

Mourlane, F<br />

P5‐18‐25<br />

Moy, B<br />

S3‐5, P1‐01‐13,<br />

P1‐13‐05, P3‐06‐27<br />

Moy, L<br />

P4‐01‐07<br />

Moynahan, ME<br />

PD09‐10,<br />

P2‐08‐01, P6‐05‐01<br />

Mrklic, I<br />

P4‐04‐04<br />

Mrozek, E<br />

P2‐11‐07<br />

Mu, D‐B<br />

P1‐14‐17<br />

Mu, Z<br />

P6‐02‐05<br />

Mudalagiriyappa, C<br />

P2‐04‐02<br />

Mueller, BM<br />

PD06‐01<br />

Mueller, V<br />

OT1‐1‐10<br />

Muggia, F<br />

P6‐13‐01<br />

Muhsen, S<br />

PD04‐05<br />

Mukai, H<br />

P2‐13‐04, P2‐13‐07<br />

Mukai, M<br />

P4‐09‐08<br />

Mukaibashi, T<br />

P1‐01‐20<br />

Mukhopadhyay, P<br />

P6‐04‐02<br />

Mullarkey, PJ<br />

P4‐01‐12<br />

Müller, BM<br />

S4‐5, P3‐06‐05<br />

Müller, V<br />

P2‐10‐17,<br />

P3‐06‐05, P6‐07‐05<br />

Muller, W<br />

P6‐02‐03<br />

Muller, WJ<br />

P5‐18‐08<br />

Mullins, DN<br />

P5‐18‐17<br />

Mulrane, L<br />

P4‐09‐06<br />

Munarriz, B<br />

P2‐10‐11<br />

Mundhenke, C<br />

OT3‐2‐01<br />

Muñoz, D<br />

P5‐07‐01<br />

Muñoz, E PD08‐05, OT3‐3‐06<br />

Muñoz, M S1‐7<br />

Muñoz‐Cousuelo, E<br />

P6‐13‐03<br />

Murakami, K<br />

P6‐05‐06<br />

Murakami, K‐I<br />

P2‐14‐03<br />

Murakami, S<br />

P1‐14‐02<br />

Mural, RJ P1‐05‐09, P2‐05‐21,<br />

P3‐04‐06, P3‐09‐03, P5‐01‐07<br />

Murias, A P1‐07‐18, P2‐01‐06<br />

Muris, JJ<br />

PD01‐03<br />

Murphy, C<br />

P5‐18‐20<br />

Murphy, D<br />

PD02‐01<br />

Murphy, DA<br />

PD06‐03<br />

Murphy, T<br />

P2‐10‐05<br />

Murray, JL<br />

P5‐16‐01<br />

Murray, N PD07‐09, OT3‐3‐09<br />

Muscato, J<br />

OT1‐1‐07<br />

Muschler, B<br />

P4‐13‐11<br />

Muss, C<br />

P5‐20‐11<br />

Muss, HB<br />

P6‐08‐05<br />

Muth, M<br />

PD07‐02<br />

Muzumdar, S P4‐06‐14, P4‐06‐16<br />

Muzzio, M<br />

P1‐09‐07<br />

Myerson, R<br />

P6‐13‐01<br />

Myhre, S<br />

P3‐04‐03<br />

Myomoto, A<br />

P5‐10‐09<br />

Nabell, L<br />

P6‐05‐02<br />

Nabell, LM<br />

OT1‐1‐11<br />

Nabholtz, J‐M<br />

P3‐06‐20<br />

Nadi, K<br />

P1‐01‐18<br />

Naeim, A<br />

P4‐13‐13<br />

Nagai, S<br />

P1‐14‐06<br />

Nagaoka, R P2‐05‐07, P3‐06‐29<br />

Nagar, H<br />

P4‐03‐09<br />

Nagarwala, Y<br />

OT1‐1‐07<br />

Nagasawa, J<br />

P6‐04‐12<br />

Nagata, Y<br />

P3‐13‐04<br />

Nagayama, A<br />

P1‐14‐09<br />

Nagayasu, T<br />

P1‐01‐27<br />

Nagi, F<br />

OT1‐1‐04<br />

Naing, A<br />

P5‐20‐09<br />

Nair, BC<br />

PD09‐07<br />

Najita, J<br />

P2‐16‐04<br />

Nakagami, K<br />

P3‐02‐06<br />

Nakagawa, C<br />

P5‐15‐06<br />

Nakagawa, T<br />

P4‐03‐07<br />

Nakamura, S P1‐01‐12, P1‐14‐02,<br />

P2‐10‐18, P5‐15‐06,<br />

P6‐07‐23, P6‐07‐38<br />

Nakamura, Y P3‐06‐26, P6‐05‐14<br />

Nakano, S<br />

P4‐03‐08<br />

Nakao, K<br />

P1‐01‐27<br />

Nakashima, M<br />

P1‐01‐27<br />

Nakata, N<br />

P4‐03‐10<br />

Nakatani, E<br />

P3‐13‐04<br />

Nakayama, H<br />

P1‐01‐20<br />

Nakayama, I<br />

P3‐13‐04<br />

Nakayama, K<br />

P6‐07‐23<br />

Nakayama, T<br />

P1‐14‐02<br />

Nakhlis, F<br />

P3‐10‐06,<br />

P4‐06‐01, P5‐02‐01<br />

Nakijima, H<br />

P2‐06‐04<br />

Nalvarte, I<br />

PD01‐06<br />

Nam, J‐M<br />

P1‐05‐08<br />

Nam, SJ P2‐05‐20, P2‐10‐41, P3‐11‐01<br />

Namjoshi, M P2‐14‐07, P3‐07‐06,<br />

P3‐07‐07, P3‐07‐14,<br />

P5‐15‐05, P5‐20‐12<br />

Nanda, S<br />

PD01‐01, P4‐06‐02<br />

Nannini, M<br />

P5‐19‐02<br />

Nascimento, CCP<br />

P3‐02‐14<br />

Nash, CE<br />

P6‐02‐06<br />

Nasias, D<br />

P2‐01‐03<br />

Nassar, A<br />

P5‐01‐08<br />

Nasser, MW<br />

P1‐05‐17<br />

Nasu, H<br />

P4‐03‐05<br />

Natarajan, L<br />

P2‐11‐09<br />

Natori, T<br />

PD06‐02<br />

Natrajan, R<br />

PD05‐08<br />

Nätterdal, T<br />

P6‐12‐01<br />

Naume, B<br />

P3‐05‐04<br />

Nayak, SR<br />

P2‐09‐02<br />

Nazareth, LV<br />

P2‐06‐01<br />

Nearchou, A<br />

P5‐18‐18<br />

Neciosup, SP<br />

P6‐07‐21<br />

Negoro, S<br />

P6‐07‐30<br />

Neill, AA<br />

P6‐11‐07<br />

Nekljudova, V S2‐2, S3‐1, P6‐07‐05,<br />

OT1‐1‐13, OT2‐2‐05, OT3‐3‐11<br />

Nelson, MT<br />

P1‐14‐11<br />

Nemeth, Z<br />

P3‐05‐08<br />

Nemoto, M<br />

P4‐03‐03<br />

Nerenberg, M<br />

PD02‐01<br />

Nerich, V<br />

P5‐18‐22<br />

Nerurkar, A P1‐01‐10, P1‐01‐11<br />

Nestle‐Kraemling, C P1‐14‐04<br />

Neubauer, M<br />

P2‐11‐16<br />

Neugebauer, J<br />

P2‐01‐11<br />

Neugebauer, JK<br />

P2‐01‐02<br />

Neumayer, L S3‐5<br />

Neumayer, LA<br />

P4‐14‐01<br />

Neumeister, V<br />

P2‐10‐06<br />

Neven, P P2‐10‐12, P2‐12‐06, P2‐13‐06,<br />

P5‐18‐19, P6‐05‐05, P6‐07‐04,<br />

P6‐07‐05, P6‐07‐19, P6‐07‐29,<br />

P6‐09‐07, OT2‐2‐02, OT2‐2‐03<br />

Newburger, D<br />

PD05‐09<br />

Newell, P<br />

P4‐04‐02<br />

Newitt, D<br />

P4‐01‐04<br />

Newitt, DN<br />

P4‐01‐10<br />

Ng, CKY<br />

PD05‐08<br />

Ng, D<br />

P5‐18‐14<br />

Ng, EKO<br />

P3‐11‐02, P4‐11‐02<br />

Ng, T<br />

P2‐10‐29<br />

Ng, W‐K<br />

P2‐05‐16<br />

Ngan, E<br />

P1‐05‐22<br />

Ngeow, J<br />

P3‐08‐09<br />

Ngo, C<br />

P1‐01‐07, P1‐01‐19<br />

Nguyen, B<br />

OT3‐4‐03<br />

Nguyen, N P2‐09‐01, P4‐08‐08<br />

Nguyen, S<br />

P5‐12‐01<br />

Nguyen, SKA P4‐17‐08, P5‐12‐02<br />

Nguyen, TH<br />

P2‐05‐22<br />

Nguyen, T‐T<br />

P5‐03‐01<br />

Nichol, A<br />

PD04‐02<br />

Niederacher, D<br />

P2‐07‐01<br />

Niehoff, P<br />

P4‐15‐01<br />

Nielsen, D P3‐06‐32, P3‐06‐35<br />

Nielsen, DL<br />

P3‐06‐03<br />

Nielsen, S P3‐06‐32, P3‐06‐35<br />

Nielsen, T P2‐10‐02, P6‐07‐10<br />

Nielsen, TO S4‐6, P3‐06‐03, P4‐16‐02<br />

Niemierko, A<br />

P6‐09‐04<br />

Nightingale, G<br />

P2‐13‐05<br />

Niikura, N P2‐05‐08, P3‐13‐02<br />

Nijenhuis, MV<br />

P2‐10‐42<br />

Nik‐Zainal, S S6‐2<br />

Niland, JC<br />

P1‐13‐05<br />

Nilsson, MP<br />

P2‐10‐37<br />

Nimeh, N<br />

P1‐14‐14<br />

Niméus‐Malmström, E P2‐10‐07<br />

Nimnual, AS<br />

P5‐06‐02<br />

Ninomiya, J<br />

P1‐14‐06<br />

Niraula, S<br />

PD03‐05<br />

Niravath, PA<br />

OT1‐1‐11<br />

Nisenbaum, B<br />

P5‐20‐06<br />

Nishikawa, S<br />

P6‐04‐17<br />

Nishimiya, H<br />

P2‐12‐13<br />

Nishimur, R<br />

PD06‐07<br />

Nishimura, S<br />

P3‐06‐26<br />

Nishino, Y<br />

P6‐07‐27<br />

Nishiyama, K P3‐06‐26, P3‐07‐10<br />

Nishiyama, S<br />

P1‐01‐20<br />

Nishiyama, T<br />

P4‐12‐06<br />

Nitz, U S3‐4, P2‐10‐08, OT3‐3‐02<br />

Niu, CJ<br />

P4‐03‐04<br />

Niwa, S<br />

P3‐10‐10<br />

Niwa, T<br />

P6‐04‐17<br />

Njiaju, UO<br />

P6‐12‐02<br />

NNBC 3‐Europe Study Group P1‐13‐13<br />

Nodora, J<br />

PD08‐04<br />

Nofech‐Mozes, S P1‐07‐17, P2‐10‐09<br />

Nogami, T<br />

P3‐07‐10<br />

Nogi, H<br />

P4‐03‐10, P5‐04‐02<br />

Noguchi, M<br />

P4‐15‐05<br />

Noguchi, S P2‐10‐18, P6‐04‐02<br />

Noh, DY<br />

P2‐10‐35<br />

Noh, D‐Y PD05‐07, P1‐01‐08,<br />

P1‐14‐16, P1‐14‐19,<br />

P2‐05‐20, P3‐06‐16, P5‐18‐23<br />

Noh, EM<br />

P5‐07‐04<br />

Noh, H<br />

P6‐07‐40<br />

Nolan, T<br />

P5‐14‐04<br />

Noll, DC S6‐3<br />

Nonogaki, S<br />

P2‐05‐17<br />

Noor, L<br />

P4‐17‐09<br />

Nordenskjöld, B<br />

P6‐07‐12<br />

Norman, J<br />

P1‐05‐13<br />

Northey, JJ<br />

P1‐05‐22<br />

Northfelt, D<br />

P5‐17‐01<br />

Nortier, HWR<br />

P1‐12‐05<br />

Nortier, JWR S3‐2, PD07‐06, P6‐07‐06<br />

Norton, L P2‐10‐01, P5‐18‐04,<br />

P5‐20‐07, P6‐11‐10, OT3‐1‐01<br />

Nourieh, M<br />

P5‐01‐04<br />

Novik, Y<br />

P5‐20‐05<br />

Nunez, A<br />

P5‐12‐03<br />

Nuñez, J<br />

P5‐16‐03<br />

Nuñez, LE<br />

P4‐06‐17<br />

Nunez, P<br />

OT1‐1‐08<br />

Nyante, SJ<br />

P3‐07‐04<br />

Nygaard, S<br />

P3‐06‐32<br />

Nyhof‐Young, JM<br />

P6‐09‐01<br />

Oates, C<br />

P4‐17‐03<br />

Oba, H<br />

P1‐14‐06<br />

Obdeijn, I‐M<br />

P3‐02‐09<br />

O’Brien, NA<br />

P4‐08‐01<br />

O’Brien, P<br />

P1‐13‐01<br />

Ocal, T<br />

P1‐05‐04<br />

Ocana, A P2‐10‐23, P4‐06‐17, P6‐05‐04<br />

O’Cathail, S<br />

P3‐02‐04<br />

Ochoa, AE<br />

P6‐10‐04<br />

O’Conner, B<br />

P6‐13‐01<br />

O’Connor, DP<br />

P4‐09‐06<br />

Oda, M<br />

P4‐03‐05<br />

O’Donoghue, JM<br />

P4‐17‐06<br />

O’Donovan, N P3‐04‐12, OT1‐1‐06<br />

Oesterreich, S<br />

P2‐09‐02,<br />

P6‐04‐20, P6‐05‐12<br />

Ofo, E<br />

P2‐10‐29<br />

O’Gaora, P P6‐04‐01, P6‐04‐04<br />

Ogasawara, Y<br />

P3‐07‐10<br />

Ogburn, ERM<br />

OT1‐1‐03<br />

Ogilvie, T<br />

P1‐07‐10<br />

Ogiya, R<br />

P3‐13‐02<br />

Ogura, D<br />

P3‐10‐10<br />

Ogura, H<br />

P4‐03‐05<br />

Ogura, K<br />

OT1‐1‐01<br />

Oh, D‐Y<br />

PD09‐05<br />

Oh, P<br />

P2‐12‐03<br />

Ohanessian, R<br />

P2‐16‐03<br />

O’Hara, C<br />

P3‐08‐01<br />

O’Hara, J<br />

P1‐05‐06, P6‐04‐21<br />

Ohashi, S<br />

P4‐16‐13<br />

Ohashi, T<br />

P4‐03‐07<br />

Ohashi, Y P1‐13‐10, P2‐13‐04, P2‐13‐07<br />

Ohde, S<br />

P6‐07‐23<br />

Ohno, S<br />

P1‐14‐02, P1‐14‐08,<br />

P3‐06‐18, P3‐06‐26<br />

Ohsumi, S P2‐13‐07, P4‐14‐07<br />

Ohta, T<br />

P4‐16‐13<br />

Ohtani, S<br />

P1‐14‐08, P1‐14‐12<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

600s<br />

<strong>Cancer</strong> Research


December 4-8, 2012<br />

Author Index<br />

Ohuchi, N P1‐06‐02, P2‐10‐43,<br />

P6‐05‐14, P6‐07‐27<br />

Oikawa, M<br />

P1‐01‐27<br />

Ojeda, H<br />

P4‐01‐13, P4‐13‐13<br />

Ojutkangas, M‐L<br />

P2‐12‐09<br />

Okada, M<br />

P6‐07‐26<br />

Okamura, R<br />

P3‐13‐04<br />

Okamura, T P2‐05‐08, P3‐13‐02<br />

Okino, T<br />

P3‐13‐04<br />

Oktay, MH<br />

P6‐02‐04<br />

Okubo, F<br />

OT1‐1‐01<br />

Okuno, T<br />

P6‐07‐39<br />

Oldervoll, L<br />

P6‐09‐03<br />

O’Leary, K<br />

P4‐06‐10<br />

O’Leary, KA<br />

P2‐04‐04<br />

Oliff, I<br />

P6‐10‐01<br />

Olivares, K<br />

P5‐07‐02<br />

Oliveira, M P6‐13‐03, OT2‐3‐02<br />

Oliver, KA<br />

P6‐12‐02<br />

Oliveras, G<br />

P4‐09‐10,<br />

P4‐09‐11, P6‐11‐02<br />

Oliveria, CT<br />

P5‐20‐01<br />

Olivo, MS S6‐6<br />

Ollila, DW S2‐1<br />

Olsauskas‐Kuprys, R P2‐05‐15<br />

Olsen, JH<br />

P1‐09‐01<br />

Olsen, L<br />

PD08‐04<br />

Olsen, S<br />

P5‐18‐11<br />

Olson, E<br />

P5‐18‐03<br />

Olson, EM P2‐11‐07, P5‐18‐17<br />

Olson, J<br />

PD07‐01<br />

Olsson, H<br />

P2‐10‐28,<br />

P2‐10‐36, P2‐10‐37<br />

Olszewski, AJ<br />

P6‐07‐34<br />

Olvera, N<br />

PD04‐05, PD05‐02<br />

Omagari, T<br />

P1‐01‐27<br />

O’Malley, B<br />

P4‐08‐09<br />

O’Meara, WP P2‐11‐13, P2‐11‐14<br />

Omene, CO<br />

P5‐20‐05<br />

Omer, Z<br />

P4‐11‐03<br />

Omoto, Y<br />

P2‐14‐03<br />

Onega, TL<br />

P4‐01‐15<br />

O’Neill, A<br />

P5‐17‐01<br />

O’Neill, V<br />

OT3‐4‐04<br />

Ong, EYY<br />

P2‐05‐16<br />

Ong, K‐W<br />

P3‐08‐09<br />

Onishi, N<br />

P2‐04‐01<br />

Onoda, T<br />

P1‐14‐08, P6‐07‐38<br />

Onodera, Y<br />

P6‐05‐14<br />

Onstenk, W<br />

P2‐01‐09<br />

Onsum, M<br />

P1‐07‐03<br />

Onyekwelu, O<br />

P4‐14‐11<br />

Ookubo, F<br />

P1‐14‐06<br />

Oosterkamp, HM<br />

P1‐15‐05<br />

Oosterwijk, JC P3‐02‐09, P4‐11‐05<br />

Opdam, M<br />

PD01‐02,<br />

PD01‐03, P1‐15‐05<br />

Opdyke, KM P3‐02‐07, P5‐14‐02<br />

Openshaw, T<br />

P2‐16‐04<br />

Ordaz, D<br />

P4‐06‐19, P6‐04‐29<br />

Ordentlich, P P2‐09‐01, P6‐04‐23<br />

Orlandi, R<br />

P4‐06‐09<br />

Orr, N<br />

P3‐08‐04<br />

Orsini, C<br />

OT3‐3‐10<br />

Ortega, V P6‐13‐03, OT3‐3‐06<br />

Ortiz, C<br />

P5‐07‐06<br />

Ortiz, J<br />

P5‐07‐06<br />

Ortmann, O<br />

P2‐12‐06<br />

Ortmann, U<br />

OT1‐1‐10<br />

Osada, T<br />

P4‐08‐07<br />

Osaki, A<br />

P6‐04‐27<br />

Osako, T<br />

P1‐01‐12<br />

Osaku, M<br />

P4‐09‐08<br />

Osborne, CK S5‐8, PD01‐01, P4‐06‐02<br />

Osborne, CRC<br />

P5‐20‐03<br />

Osborne, K P2‐16‐02, P6‐10‐03<br />

Osborne, KC<br />

P2‐16‐03<br />

Osborne, MP<br />

P4‐15‐03<br />

O’Shaughnessy, J P1‐12‐02, P1‐12‐07,<br />

P3‐06‐12, P5‐20‐04, OT2‐3‐11<br />

Oshitanai, R<br />

P3‐13‐02<br />

Osin, P<br />

P1‐01‐10, P1‐01‐11<br />

Osipo, C<br />

P2‐05‐15, P4‐08‐06<br />

Ossher, L S6‐3<br />

Osterby, KR<br />

P6‐12‐02<br />

Osumi, S<br />

P2‐13‐04<br />

Oswald, M<br />

P5‐10‐13<br />

Ota, D<br />

PD07‐01<br />

Otani, S<br />

P4‐01‐05<br />

O’Toole, J P6‐09‐04, OT3‐2‐02<br />

O’Toole, M P2‐05‐15, P4‐08‐06<br />

Otremba, B<br />

P1‐15‐02<br />

Otsubo, R<br />

P1‐01‐27<br />

Ottesen, RA<br />

P1‐13‐05<br />

Otteson, R<br />

P1‐01‐13<br />

Ottewell, PD<br />

PD07‐08<br />

Oudard, S<br />

P5‐17‐04<br />

Oudot, A<br />

P4‐02‐06<br />

Ouhtit, A<br />

P4‐06‐14, P4‐06‐16<br />

Ould‐Kaci, M<br />

OT1‐1‐15<br />

Oura, S<br />

P1‐12‐01, P6‐07‐16<br />

Overgaard, J<br />

P3‐04‐03<br />

Overmoyer, B<br />

P5‐02‐01<br />

Overmoyer, BA P3‐10‐06, P4‐06‐01<br />

Owen, R<br />

S4‐1, P3‐06‐09<br />

Owonikoko, TK<br />

PD09‐06<br />

Oxley, P<br />

P4‐17‐08<br />

Oyama, T<br />

P3‐06‐29<br />

Ozanne, EM P3‐02‐12, P4‐01‐15,<br />

P4‐11‐03, P4‐13‐13, P5‐15‐01<br />

Ozmen, V<br />

P5‐02‐02<br />

Pacchiarotti, A<br />

P1‐01‐25<br />

Pachmann, K P1‐07‐16, P5‐03‐13<br />

Pachmann, U<br />

P5‐03‐13<br />

Pachter, JA P6‐11‐07, P6‐11‐09<br />

Pacia, E<br />

PD06‐03<br />

Padfield, D<br />

P3‐05‐06<br />

Padhani, AR<br />

P1‐06‐01<br />

Padval, MV<br />

P6‐11‐07<br />

Paepke, D<br />

P1‐13‐13<br />

Paepke, S<br />

S3‐1, P1‐14‐01,<br />

OT1‐1‐13, OT3‐3‐11<br />

Pagani, G<br />

PD03‐01<br />

Pagani, O<br />

OT2‐2‐01<br />

Page, J<br />

P1‐15‐01<br />

Page, K<br />

P1‐09‐07<br />

Paik, J‐Y<br />

P6‐01‐05<br />

Paik, N<br />

P4‐03‐11<br />

Paik, S<br />

S1‐10, S1‐11, S5‐5,<br />

OT1‐1‐12, OT2‐2‐04<br />

Paik, SS<br />

P2‐10‐39<br />

Paillocher, N<br />

P1‐14‐21<br />

Paily, AJ<br />

P4‐14‐05<br />

Paladini, L<br />

P5‐10‐01<br />

Pallaud, C<br />

P3‐06‐34<br />

Palles, C<br />

P3‐08‐04<br />

Palmer, G S3‐6, PD05‐01, PD05‐03<br />

Palmieri, C P2‐02‐03, P2‐10‐22,<br />

P2‐14‐06, P3‐02‐04, P3‐06‐13<br />

Palmisano, G<br />

P3‐04‐05<br />

Pan, H S1‐2<br />

Panageas, KS<br />

P3‐07‐09<br />

Panday, H<br />

P4‐08‐08<br />

Pandiella, A P4‐06‐17, P6‐05‐04<br />

Pandita, T<br />

P4‐08‐08<br />

Pankratz, VS<br />

P4‐12‐03<br />

Panneerdoss, S<br />

P5‐10‐14<br />

Pannu, H<br />

P5‐18‐04<br />

Pantel, K P2‐01‐02, OT1‐1‐10<br />

Papadaki, M<br />

P2‐01‐03<br />

Papadaki, MA<br />

P2‐01‐07<br />

Papadimitriou, C<br />

P5‐18‐21<br />

Papamichail, M<br />

P5‐16‐05<br />

Papoutsakis, D<br />

P6‐11‐06<br />

Paquet, A<br />

P2‐05‐06,<br />

P2‐10‐16, P2‐10‐31<br />

Paquet, L<br />

P2‐11‐10, P3‐04‐11<br />

Paragas, V<br />

P1‐07‐03<br />

Pardo, I<br />

P1‐03‐02<br />

Parham, LR<br />

PD10‐05<br />

Paridaens, R P2‐10‐12, P5‐18‐19,<br />

P6‐05‐05, P6‐07‐04, P6‐07‐19,<br />

P6‐07‐29, P6‐09‐07<br />

Parise, C<br />

P3‐09‐01, P3‐09‐02<br />

Parisini, E<br />

P4‐10‐01<br />

Parissenti, A<br />

P1‐14‐13<br />

Park, B‐W P1‐01‐23, P2‐05‐03<br />

Park, CC P1‐05‐08, P4‐01‐04, P4‐06‐05<br />

Park, H<br />

PD03‐09, P4‐13‐13<br />

Park, HS<br />

P1‐01‐23, P2‐05‐03<br />

Park, H‐S<br />

PD09‐05<br />

Park, HY<br />

P1‐01‐24<br />

Park, IA<br />

P2‐05‐20<br />

Park, J<br />

P2‐05‐01<br />

Park, JH<br />

P1‐12‐04<br />

Park, JM<br />

P1‐01‐23, P2‐05‐03<br />

Park, JW P2‐01‐05, P4‐16‐07, P6‐01‐05<br />

Park, M<br />

P6‐02‐03<br />

Park, S<br />

P1‐01‐23, P2‐05‐02,<br />

P2‐05‐03, P6‐04‐13<br />

Park, S‐J<br />

P1‐14‐19<br />

Park, SY<br />

P4‐06‐01, P5‐04‐03<br />

Park, Y<br />

P3‐07‐04<br />

Park, YH<br />

P2‐05‐20, P4‐06‐13<br />

Parker, BA P2‐06‐01, P4‐13‐13<br />

Parker, CA<br />

OT2‐3‐10<br />

Parker, J<br />

P6‐07‐10<br />

Parker, JS S6‐1<br />

Parker, S<br />

P2‐15‐02, OT3‐1‐02<br />

Parkes, EE<br />

P6‐07‐24<br />

Parmar, M S6‐5<br />

Partridge, A<br />

P6‐08‐03<br />

Partridge, AH<br />

P4‐01‐09<br />

Pasick, R<br />

P4‐13‐05<br />

Paskett, LA<br />

P6‐11‐12<br />

Pasqualini, R<br />

P3‐10‐09<br />

Pasquier, F<br />

P2‐13‐10<br />

Pasula, S<br />

P1‐05‐21<br />

Patel, R S1‐6<br />

Patel, V<br />

P1‐05‐23<br />

Pathania, S<br />

P4‐10‐01<br />

Pathiraja, T<br />

P2‐09‐02<br />

Patil, R<br />

P5‐16‐02<br />

Patil, S P2‐08‐01, P2‐11‐05, P2‐12‐05,<br />

P5‐18‐04, P5‐18‐20, P6‐05‐01,<br />

P6‐05‐02, OT3‐1‐01<br />

Patl, S<br />

P5‐20‐07<br />

Patre, M<br />

P5‐18‐05<br />

Patrick‐Miller, L<br />

P6‐08‐01<br />

Patt, D<br />

P2‐11‐16, P4‐11‐04<br />

Patterson, RE<br />

P2‐11‐09<br />

Paul, BA<br />

P5‐06‐01<br />

Paul, D<br />

P3‐06‐12<br />

Paula Junior, W<br />

P2‐12‐04<br />

Pauletti, G<br />

P2‐08‐02<br />

Paulino, AC<br />

P6‐07‐11<br />

Paulose, C<br />

P4‐08‐04<br />

Pauporte, I S5‐3<br />

Pauwels, P P2‐01‐08, P2‐01‐09<br />

Pavdal, M<br />

P6‐11‐09<br />

Pavlou, MP P2‐05‐11, P6‐05‐09<br />

Pawaskar, M<br />

P3‐07‐08<br />

Pazdur, R S1‐11<br />

Peacock, DL<br />

P1‐05‐15<br />

Peacock, N<br />

P5‐20‐10<br />

Peccatori, FA<br />

P6‐07‐14<br />

Pechan, J<br />

P2‐01‐12<br />

Peck, AR S1‐8<br />

Peck, KA<br />

P2‐11‐15<br />

Pectasides, D<br />

P1‐07‐15<br />

Pedersen, HC<br />

P1‐07‐05<br />

Pedersen, RC<br />

P3‐10‐07<br />

Pedrini, JL S1‐4<br />

Peeters, DJ P2‐01‐08, P2‐01‐09<br />

Peeters, J<br />

P3‐06‐11<br />

Peeters, M P2‐01‐08, P2‐01‐09<br />

Peeters, S<br />

P2‐10‐12,<br />

P6‐05‐05, P6‐07‐29<br />

Peg, V<br />

P1‐01‐29<br />

Peisker, U<br />

P1‐07‐16<br />

Peixoto, J‐E P3‐01‐07, P3‐02‐13<br />

Peksa, R<br />

P3‐12‐09, P3‐13‐03<br />

Pelissier, S<br />

OT3‐4‐05<br />

Pellegrini, M<br />

P5‐01‐10<br />

Peltier, S S6‐3<br />

Peluffo, G<br />

P4‐06‐01<br />

Peluzio, MCG<br />

P3‐07‐12<br />

Pemberton, K<br />

OT1‐1‐17<br />

Penault‐Llorca, F PD05‐08, P2‐08‐04,<br />

P3‐05‐07, P3‐06‐20,<br />

P3‐14‐03, P5‐21‐01<br />

Pendergrass, K S1‐4<br />

Pendleton, CS<br />

P6‐05‐03<br />

Pendyala, P<br />

P2‐01‐05<br />

Penedo, FJ<br />

PD08‐05<br />

Peoples, GE P5‐16‐02, P5‐16‐05<br />

Pepels, MJ<br />

PD07‐06<br />

Perea, F<br />

P1‐14‐13<br />

Perea, S<br />

P1‐14‐10, P5‐18‐15<br />

Perelló, A<br />

P1‐07‐18,<br />

P2‐01‐06, P3‐12‐08<br />

Peretz‐Yablonski, T OT1‐1‐02<br />

Perey, L<br />

P6‐04‐05<br />

Perez, C<br />

PD10‐02, P1‐02‐01<br />

Pérez, D P1‐07‐18, P1‐14‐10, P2‐01‐06<br />

Perez, E<br />

P1‐13‐01<br />

Perez, EA S5‐4, S5‐5, S6‐6, PD10‐04,<br />

P1‐05‐24, P1‐12‐06, P2‐10‐01<br />

Perez, J<br />

P1‐07‐19<br />

Perez, SA<br />

P5‐16‐05<br />

Perez‐Bueno, F<br />

P4‐09‐11<br />

Perez‐Garcia, J<br />

P6‐13‐03<br />

Perez‐Plasencia, C<br />

PD08‐06<br />

Perez‐Stable, EJ<br />

PD08‐05<br />

Pérez‐Tenorio, G<br />

P6‐07‐12<br />

Perin, T<br />

P5‐10‐17<br />

Periyasamy, M<br />

P1‐04‐04<br />

Permaul, E<br />

P1‐09‐04<br />

Perou, C<br />

S1‐11, P6‐07‐10<br />

Perou, CM S6‐1, P2‐10‐01, P5‐19‐01,<br />

P6‐02‐09, P6‐03‐01<br />

Perre, CI<br />

P4‐14‐08<br />

Perree, M<br />

P3‐06‐28<br />

Perren, T<br />

P4‐01‐14<br />

Perry, A<br />

P4‐17‐06<br />

Perry, M‐C<br />

P5‐18‐08<br />

Pertsemlidis, A<br />

P5‐10‐14<br />

Peters, A<br />

P3‐12‐03<br />

Peters, FPJ<br />

P6‐07‐32<br />

Peters, NAJB<br />

P6‐07‐32<br />

Peters, U<br />

PD03‐09<br />

Petersen, V<br />

P5‐21‐02<br />

Peterson, LM<br />

P6‐04‐03<br />

Petit, A<br />

P1‐01‐29<br />

Petit, T<br />

P2‐13‐10, P3‐06‐20<br />

Peto, J<br />

P3‐08‐04<br />

Peto, R S1‐2<br />

Petrcich, W<br />

P3‐11‐03<br />

Petrella, G<br />

P4‐15‐04<br />

Petrella, TM<br />

P2‐12‐03<br />

Petricoin, EF P3‐04‐01, P3‐04‐02<br />

Petropoulos, C P2‐10‐16, P2‐10‐31<br />

Petrowsky, C<br />

P1‐12‐04<br />

Petrucelli, N<br />

PD03‐09<br />

Petruse, A<br />

P4‐13‐13<br />

Petruzelka, L S1‐4<br />

Petry, C<br />

S4‐3, P2‐10‐11<br />

Pettke, E<br />

P3‐02‐11<br />

Peyrat, J‐P<br />

P5‐21‐01<br />

Peyro Saint Paul, HP OT3‐3‐10<br />

Pfalzer, LA P2‐11‐13, P2‐11‐14<br />

Pfeiler, G<br />

P1‐15‐10, P2‐13‐01<br />

Pham, T<br />

P2‐01‐10<br />

Pham, TND<br />

P6‐04‐24<br />

Pharoah, PD P3‐08‐02, P3‐08‐05<br />

Phillips, K‐A<br />

P4‐13‐10<br />

Phipps, EA<br />

P5‐08‐01<br />

Piccart, M S1‐11, S4‐4, S6‐2, P1‐07‐08,<br />

P2‐14‐01, P3‐04‐10, P3‐05‐02,<br />

P3‐05‐03, P5‐18‐19, P6‐04‐02,<br />

P6‐07‐14, P6‐07‐22<br />

Piccart‐Gebhart, M<br />

OT1‐1‐16<br />

Piccart‐Gebhart, MJ S5‐2, P5‐18‐02<br />

Picquenot, JM<br />

P3‐14‐03<br />

Pidgeon, GP<br />

P6‐01‐02<br />

Pieczynska, B P3‐12‐09, P3‐13‐03<br />

Piede, JA<br />

P4‐08‐07<br />

Pienkowski, T<br />

P5‐02‐02<br />

Pierga, JY<br />

P3‐06‐04<br />

Pierga, J‐Y<br />

S5‐3, P1‐01‐07,<br />

P1‐01‐19, P1‐14‐03, P1‐14‐18,<br />

P2‐01‐01, P2‐01‐04, P2‐01‐13,<br />

P5‐21‐03, OT2‐3‐05,<br />

OT3‐4‐05, OT3‐4‐06<br />

Pietenpol, JA P3‐05‐09, P6‐05‐03<br />

Pietrarota, P<br />

P4‐16‐08<br />

www.aacrjournals.org 601s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Pietras, RJ<br />

PD03‐04<br />

Pigeonnat, S<br />

P3‐08‐07<br />

Pigorsch, S S4‐2<br />

Piha‐Paul, S<br />

P5‐20‐09<br />

Pijnappel, RM P4‐14‐08, P5‐01‐11<br />

Pikiel, J<br />

P5‐18‐21<br />

Pillai, MM<br />

PD01‐07<br />

Pillai, S<br />

P2‐04‐02<br />

Pina, L<br />

P5‐16‐03<br />

Pinder, S<br />

P2‐10‐29<br />

Pinna, G<br />

P5‐03‐01<br />

Pinotti, M<br />

P3‐02‐14<br />

Pinter, T S1‐6<br />

Pintilie, M<br />

P2‐10‐33<br />

Pinto, JA S3‐6, P6‐07‐20, P6‐07‐21<br />

Pinto, RR<br />

P3‐11‐04, P6‐07‐44<br />

Pinto, W<br />

P1‐01‐29<br />

Piper, T S1‐5<br />

Pircher, M<br />

P5‐20‐11<br />

Pirolli, M<br />

P2‐13‐03<br />

Pitcher, B<br />

P6‐08‐05<br />

Pitcher, BN<br />

P2‐10‐01<br />

Pivot, X<br />

S5‐3, S6‐5,<br />

P5‐15‐07, P5‐18‐22<br />

Pizon, M<br />

P5‐03‐13<br />

Platero, S<br />

P5‐01‐09<br />

Plevritis, S<br />

P2‐09‐03<br />

Plichta, JK<br />

P1‐02‐01<br />

Pluard, T<br />

PD07‐01<br />

Plyler, JR<br />

P5‐10‐16<br />

Poggi, LE<br />

P6‐12‐02<br />

Pogorelic, Z<br />

P4‐04‐04<br />

Pohorelic, B P4‐09‐07, P6‐07‐17<br />

Poillot, M‐L<br />

P3‐08‐07<br />

Polgar, C<br />

P4‐15‐01<br />

Polistena, A<br />

P1‐01‐25<br />

Pollak, MN<br />

P1‐07‐13<br />

Polley, M‐YC S4‐6<br />

Polyak, K P3‐05‐04, P4‐06‐01<br />

Polyzos, A<br />

P1‐13‐09<br />

Pond, G<br />

P6‐09‐02<br />

Poniewierski, MS P1‐15‐04, P3‐06‐07<br />

Ponniah, S P5‐16‐02, P5‐16‐05<br />

Popova, T<br />

P5‐02‐03<br />

Porcher, R<br />

P4‐13‐03<br />

Port, D<br />

PD08‐06<br />

Porta, R<br />

P4‐09‐10<br />

Porter, C<br />

P2‐11‐17<br />

Porter, D<br />

P6‐11‐06<br />

Porter, DA<br />

P4‐01‐10<br />

Portielje, JEA<br />

P1‐12‐05<br />

Portier, BP PD02‐04, P6‐07‐45<br />

Portillo, RM<br />

P3‐06‐12<br />

Posse, S<br />

P3‐03‐01<br />

Postma, EL<br />

P4‐01‐17,<br />

P5‐01‐14, P5‐15‐03<br />

Potemski, P<br />

P5‐18‐21<br />

Potter, BM<br />

P1‐01‐04<br />

Potter, S<br />

P4‐17‐03<br />

Potvin, K<br />

P1‐14‐13<br />

Potyka, I S4‐2<br />

Pouget, N<br />

P3‐08‐07<br />

Poulet, B<br />

P6‐07‐03<br />

Poulikakos, P<br />

P4‐08‐02<br />

Pourmand, N<br />

P2‐06‐05<br />

Powathil, GG<br />

P5‐05‐02<br />

Powell, CA<br />

P1‐05‐17<br />

Power, S<br />

P4‐13‐06<br />

Pradeep, S<br />

P5‐10‐03<br />

Prager‐van der Smissen, WJC P6‐04‐08<br />

Prasad, N<br />

P5‐18‐13<br />

Prat, A P6‐05‐04, P6‐07‐10, OT3‐3‐06<br />

Prathap, P<br />

P6‐07‐13<br />

Pratt, C<br />

PD09‐01<br />

Prendergast, BM<br />

P5‐17‐07<br />

Prendergast, GC<br />

P4‐09‐12<br />

Prenois, F<br />

P5‐13‐02<br />

Presant, C<br />

P3‐06‐28<br />

Press, M<br />

PD02‐01<br />

Press, MF<br />

P4‐08‐05<br />

Price, A<br />

P4‐01‐07<br />

Price, JT<br />

P6‐07‐17<br />

Price, KN<br />

OT2‐2‐01<br />

Priego, V<br />

P1‐14‐14<br />

Prinzler, J<br />

S4‐5, P3‐06‐05<br />

Prior, WW P4‐07‐01, P5‐19‐02<br />

Pritchard, K<br />

P1‐13‐01,<br />

P2‐12‐03, P6‐08‐04<br />

Pritchard, KI P1‐07‐13, P2‐13‐02<br />

Privat, M<br />

P3‐06‐20<br />

Procter, M S5‐2<br />

Proctor, I<br />

PD04‐08<br />

Protter, AA<br />

P2‐14‐02<br />

Prové, A<br />

P2‐01‐08<br />

Prowell, T S1‐11<br />

Prudente, R<br />

P6‐04‐12<br />

Pruneri, G PD03‐01, PD04‐07,<br />

PD06‐06, P3‐05‐03<br />

Psyrri, A S5‐4<br />

Pu, M<br />

P2‐06‐01<br />

Puccio, A<br />

PD03‐01<br />

Pugh, TW<br />

P4‐12‐05<br />

Puglisi, F<br />

OT1‐1‐02<br />

Puhalla, SL<br />

PD09‐06,<br />

P6‐07‐02, OT1‐1‐11<br />

Puig, T P4‐09‐10, P4‐09‐11, P6‐11‐02<br />

Puig‐Vives, M<br />

P4‐09‐11<br />

Pulhalla, S<br />

OT2‐3‐07<br />

Puntoni, M PD03‐01, PD06‐06<br />

Pupa, SM<br />

P5‐18‐10<br />

Purdie, CA<br />

PD03‐02<br />

Purnomosari, D<br />

P3‐01‐08<br />

Purshouse, K<br />

P2‐02‐03<br />

Purushotham, A<br />

P2‐10‐29<br />

Pusztai, L<br />

S6‐7, P2‐10‐10,<br />

P2‐10‐17, P5‐10‐01<br />

Putney, JL<br />

OT2‐1‐04<br />

Putter, H<br />

PD07‐06<br />

Qamri, Z<br />

P1‐05‐17<br />

Qian, J<br />

P6‐04‐12, OT2‐3‐07<br />

Qin, J<br />

P4‐09‐09<br />

Qin, L<br />

P6‐04‐25<br />

Qin, L‐X<br />

PD05‐02<br />

Qiong, P<br />

P4‐16‐10<br />

Quach, D<br />

P2‐13‐03<br />

Quadt, C<br />

P6‐10‐07<br />

Quigley, DA<br />

P5‐05‐03<br />

Quigley, J<br />

P2‐13‐03<br />

Quijano, Y P1‐14‐10, P5‐18‐15<br />

Quinaux, E<br />

PD02‐01<br />

Quintanar‐Jurado, V P5‐10‐12<br />

Quintayo, MA<br />

P1‐07‐17<br />

Quintyn‐Ranty, M‐L P3‐05‐07<br />

Raafat, A<br />

P2‐04‐08<br />

Rabaglio, M<br />

P2‐13‐02<br />

Rabeneck, L<br />

P3‐02‐10<br />

Rabenstein, C<br />

P5‐03‐13<br />

Rabinovitch, R<br />

OT1‐2‐01<br />

Rack, B<br />

P2‐10‐25, P3‐06‐05,<br />

P5‐14‐06, P5‐15‐04, P6‐07‐05<br />

Rack, BJ<br />

P2‐01‐11<br />

Rack, BK<br />

P2‐01‐02,<br />

P4‐13‐11, OT1‐1‐10<br />

Radecka, B<br />

P2‐10‐31,<br />

P3‐12‐09, P3‐13‐03<br />

Radisky, DC P4‐12‐03, P6‐06‐01<br />

Radosevic‐Robin, N P5‐21‐01<br />

Radovich, M PD05‐06, P1‐03‐02<br />

Ragaz, J<br />

P4‐13‐04, P6‐07‐43<br />

Raghavendra, A<br />

P4‐01‐11<br />

Raguin, O<br />

P4‐02‐06<br />

Rahal, RMS<br />

P3‐02‐13<br />

Rahnenführer, J<br />

P2‐10‐13<br />

Rai, Y<br />

P1‐12‐01, P5‐18‐16<br />

Raj, V<br />

P3‐06‐30<br />

Rajan, P<br />

P1‐02‐01<br />

Rajput, A<br />

P3‐03‐01<br />

Rakha, E<br />

P3‐06‐09<br />

Rakovitch, E<br />

P1‐07‐17<br />

Ramadan, D<br />

P5‐18‐07<br />

Ramalingam, SS<br />

PD09‐06<br />

Raman, D<br />

P1‐05‐01<br />

Raman, S<br />

P2‐11‐07<br />

Ramaswamy, B<br />

P2‐11‐07<br />

Ramaswamy, S<br />

P2‐01‐14<br />

Ramirez, AG<br />

PD08‐05<br />

Ramirez, E<br />

P5‐07‐06<br />

Ramirez, J<br />

P2‐11‐05<br />

Ramirez, O<br />

P4‐04‐06<br />

Ramirez Ardila, DE<br />

P3‐06‐01<br />

Ramkumar, A P4‐06‐14, P4‐06‐16<br />

Ramnarain, A<br />

P1‐15‐07<br />

Ramos, K<br />

P4‐04‐05<br />

Ramos, M S1‐7<br />

Rampaul, R<br />

P4‐17‐06<br />

Ramteke, VK<br />

P4‐02‐02<br />

Randolph, S S1‐6<br />

Rangel, LBA P5‐01‐15, P6‐11‐13<br />

Rao, M P5‐04‐04, P5‐10‐14, P5‐10‐16<br />

Rao, R<br />

S3‐7, P1‐07‐04<br />

Rappold, E<br />

PD10‐05<br />

Raro, P P1‐01‐21, P1‐14‐21, OT2‐1‐01<br />

Rastegar, R<br />

P4‐17‐08<br />

Rastogi, P S1‐11, S5‐5, OT2‐2‐04<br />

Rathbone, EJ S6‐4<br />

Rattay, T<br />

P1‐14‐15<br />

Raubitschek, AA<br />

P2‐05‐10<br />

Rauch, P<br />

P6‐08‐01<br />

Raum, N<br />

P2‐16‐01<br />

Ravichandran, P<br />

P6‐07‐41<br />

Ravnsbaek, A<br />

P6‐07‐08<br />

Ray, P<br />

OT2‐1‐04<br />

Ray‐Coquard, I<br />

P5‐17‐04<br />

Rayson, D<br />

OT1‐1‐07<br />

Razis, E<br />

P1‐07‐15<br />

Rea, D S1‐5<br />

Ready, N<br />

P2‐10‐03, P3‐06‐07<br />

Rebollar‐Vega, RG<br />

P5‐10‐12<br />

Rechoum, Y<br />

P4‐08‐09<br />

Reddick, C<br />

OT2‐2‐03<br />

Reddy, J<br />

PD03‐06<br />

Redmond, W<br />

P4‐04‐02<br />

Reed, J<br />

P6‐07‐10<br />

Reed, JP<br />

P2‐10‐01<br />

Reedijk, M<br />

PD03‐05<br />

Rees, B<br />

P6‐07‐18<br />

Rees, H<br />

P2‐10‐09<br />

Rees, R<br />

P6‐07‐09<br />

Reeves, KJ<br />

PD07‐08<br />

Refice, S PD03‐03, P1‐09‐02, P4‐02‐07<br />

Refinetti, AP P1‐09‐03, P4‐01‐07<br />

Regan, M<br />

S4‐4, PD10‐03<br />

Regan, MM P1‐07‐07, P3‐10‐06,<br />

P5‐18‐02, OT2‐2‐01<br />

Rege, J<br />

P1‐12‐02, P1‐13‐11,<br />

P5‐20‐04, P6‐11‐14<br />

Regehr, G<br />

P5‐12‐01<br />

Regondi, V<br />

P5‐18‐10<br />

Reich, SD<br />

OT1‐1‐05<br />

Reichow, J P2‐15‐02, OT3‐1‐02<br />

Reid, I<br />

P4‐14‐13<br />

Reid, J<br />

PD10‐08<br />

Reid, R<br />

P4‐14‐09<br />

Reidunsdatter, RJ<br />

P6‐09‐03<br />

Reijm, EA<br />

P6‐04‐08<br />

Reinholz, MM<br />

PD10‐04<br />

Reintgen, D<br />

OT3‐4‐02<br />

Reis‐Filho, J<br />

P3‐06‐09<br />

Reis‐Filho, JS<br />

PD05‐08<br />

Reiter, BE<br />

P6‐04‐18<br />

Re‐Louhau, F<br />

P4‐06‐17<br />

Renne, G<br />

P6‐07‐14<br />

Rennert, G<br />

P6‐14‐03<br />

Rennert, HS<br />

P6‐14‐03<br />

Renshaw, L<br />

P3‐06‐23,<br />

P6‐04‐09, P6‐04‐10<br />

Resch, A<br />

P4‐15‐01<br />

Ressler, S<br />

P5‐20‐11<br />

Reuben, JM PD03‐08, P2‐01‐01,<br />

P2‐01‐12, P2‐03‐01, P2‐10‐32,<br />

P3‐10‐05, P3‐10‐10, P5‐03‐05,<br />

P5‐03‐09, P5‐04‐06, P5‐10‐02,<br />

OT2‐3‐01<br />

Reuter‐Lorenz, PA S6‐3<br />

Rey, J‐B<br />

P5‐17‐04<br />

Reyal, F<br />

P1‐01‐19<br />

Reyes, ME<br />

P5‐03‐09<br />

Reynolds, C<br />

P1‐01‐22<br />

Reyre, J<br />

P3‐05‐07<br />

Rezai, M<br />

P1‐14‐01,<br />

P2‐10‐14, P2‐10‐25<br />

Rezende, LCD<br />

P5‐01‐15<br />

Riabowol, K<br />

P4‐09‐07<br />

Ribas, R<br />

PD01‐05<br />

Ribeiro, IM<br />

P3‐02‐14<br />

Ribeiro, MKA<br />

P2‐12‐04<br />

Ribeiro, R<br />

P3‐01‐03<br />

Ricart, AD<br />

OT1‐1‐05<br />

Ricci, D<br />

P5‐01‐09<br />

Ricci, F<br />

P1‐01‐25<br />

Richardson, A<br />

PD01‐08<br />

Richardson, AL<br />

P1‐07‐07<br />

Richardson, E<br />

P1‐07‐13<br />

Richer, JK P2‐14‐02, P5‐10‐04,<br />

P5‐10‐05, P5‐10‐06<br />

Richmond, A<br />

P1‐05‐01<br />

Ricker, C<br />

P4‐01‐11<br />

Rief, W<br />

P6‐09‐08<br />

Riegel, AT<br />

P6‐05‐07<br />

Riehle, K<br />

P6‐05‐12<br />

Rifa, J<br />

P3‐12‐08<br />

Rigal, O<br />

P1‐15‐08<br />

Rijna, H<br />

P4‐11‐01<br />

Riley, L<br />

P2‐06‐04<br />

Rimareix, F<br />

P4‐14‐12<br />

Rimawi, M<br />

P5‐18‐24<br />

Rimawi, MF<br />

S5‐8, P2‐16‐02<br />

Rimm, D<br />

S5‐4, P3‐14‐01<br />

Rimm, DL<br />

PD02‐01,<br />

P2‐10‐06, P3‐06‐25<br />

Ring, A<br />

P6‐07‐05<br />

Ring, JE<br />

P6‐11‐07<br />

Rinnerthaler, G<br />

P5‐20‐11<br />

Rino, Y<br />

P1‐01‐20<br />

Rios, M<br />

P1‐13‐04<br />

Ritchie, C<br />

P2‐12‐14<br />

Rivain, A‐L<br />

P1‐01‐19<br />

Rivaux, G<br />

P4‐14‐10<br />

Rivera, E<br />

OT3‐3‐01<br />

Rix, P<br />

P6‐04‐12<br />

Ro, J PD09‐05, P5‐18‐26, OT1‐1‐16<br />

Roarty, K<br />

P5‐03‐08<br />

Robbins, D<br />

P3‐13‐05<br />

Robert, NJ<br />

P3‐06‐12<br />

Robertson, C P4‐13‐07, P4‐13‐08<br />

Robertson, FM<br />

P4‐06‐03,<br />

P5‐04‐05, P6‐10‐04<br />

Robertson, S<br />

P2‐10‐09<br />

Robertson, SJ<br />

P5‐01‐01<br />

Robidoux, A<br />

S3‐2, P6‐07‐06<br />

Robinson, A S3‐3<br />

Robinson, BD<br />

P6‐02‐04<br />

Robinson, M<br />

OT2‐3‐11<br />

Robinson, P<br />

PD10‐02<br />

Robinson, PA<br />

P1‐12‐04<br />

Roblyer, D<br />

P4‐02‐04<br />

Robson, M<br />

OT2‐3‐07<br />

Roca, L<br />

P1‐13‐04<br />

Rocha, RM<br />

P1‐06‐03<br />

Roche, H<br />

P1‐13‐04, P3‐04‐04<br />

Roche, N<br />

P1‐01‐10, P1‐01‐11<br />

Roche‐Comet, I<br />

P5‐21‐01<br />

Rocque, GB<br />

P2‐11‐15<br />

Rodenhuis, S<br />

P4‐02‐01<br />

Rodier, J‐F<br />

P1‐01‐21<br />

Rodón, J<br />

OT2‐3‐06<br />

Rodrigues, DCN<br />

P3‐02‐13<br />

Rodrigues, DN PD05‐08, P3‐06‐09<br />

Rodriguez, AA<br />

P3‐06‐14,<br />

P5‐03‐03, P6‐07‐11<br />

Rodriguez, CA<br />

P2‐10‐11<br />

Rodriguez, EM<br />

P1‐04‐01<br />

Rodriguez, JG<br />

OT1‐1‐08<br />

Rodríguez‐Antona, C OT3‐3‐05<br />

Rodriguez‐Cuevas, S P5‐10‐12<br />

Rodriguez‐Lescure, A P2‐10‐11<br />

Rodriguez‐Peralto, JL P5‐10‐11<br />

Rodriguez‐Spiteri, N P5‐16‐03<br />

Rodriguez‐Teja, M<br />

P2‐02‐03<br />

Rodrik‐Outmezguine, V P4‐08‐02<br />

Rody, A<br />

P2‐10‐17<br />

Roepman, P<br />

P4‐09‐05<br />

Roessl, E<br />

P4‐03‐06<br />

Rogers, AN<br />

P2‐05‐04<br />

Rogers, K<br />

P3‐06‐28<br />

Rogers, N<br />

P4‐01‐10<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

602s<br />

<strong>Cancer</strong> Research


December 4-8, 2012<br />

Author Index<br />

Rogowski, W P3‐12‐09, P3‐13‐03<br />

Rohan, TE<br />

P6‐02‐04<br />

Rojas, LA<br />

P5‐18‐13<br />

Rojas, M<br />

P5‐13‐03<br />

Rojas, PA<br />

P4‐06‐15<br />

Rokutanda, N P2‐05‐07, P3‐06‐29<br />

Rollot, F<br />

OT3‐3‐10<br />

Roloff, T<br />

PD01‐06<br />

Romeder, F<br />

P5‐20‐11<br />

Rømer, M P3‐06‐32, P3‐06‐35<br />

Romero, A<br />

P5‐07‐06<br />

Romero, Q<br />

PD03‐07<br />

Romero‐Cordoba, SL P5‐10‐12<br />

Romics, L<br />

P4‐14‐13<br />

Romieu, G<br />

S5‐3, P3‐12‐01<br />

Romieux, G<br />

OT3‐3‐10<br />

Romond, E<br />

S5‐5, P2‐14‐05<br />

Roncadin, M<br />

S4‐2, P5‐10‐17<br />

Rong, H‐M<br />

P2‐08‐02<br />

Rose, C<br />

PD03‐07<br />

Rose, J<br />

P3‐02‐07<br />

Rosen, DB<br />

P6‐07‐31<br />

Rosen, J<br />

P4‐02‐08, P5‐03‐08<br />

Rosen, N PD04‐05, P4‐08‐02, P5‐18‐04<br />

Rosen, RB<br />

P6‐05‐10<br />

Rosenberg, AL S1‐8<br />

Rosenberg, CA<br />

PD03‐09<br />

Rosenblad, A<br />

P2‐12‐09<br />

Rosenstock, J P4‐13‐07, P4‐13‐08<br />

Rosenthall, RE<br />

P2‐10‐27<br />

Roskelly, C<br />

P1‐05‐05<br />

Ross, G<br />

S5‐1, S6‐7,<br />

P5‐18‐01, P5‐18‐26<br />

Ross, GM<br />

P3‐08‐04<br />

Ross, J<br />

PD02‐07, PD05‐01<br />

Ross, L<br />

P4‐07‐01<br />

Ross, LB<br />

P5‐19‐02<br />

Roth, S<br />

P1‐14‐04<br />

Rothé, F S6‐2<br />

Rothstein, R<br />

P4‐07‐05<br />

Rotkiss, M<br />

OT3‐4‐03<br />

Rotmensz, N PD04‐07, P6‐07‐14<br />

Rouanet, P<br />

OT2‐1‐01<br />

Roussi, P<br />

P6‐09‐05<br />

Roussou, P<br />

P6‐11‐08<br />

Rouzier, R P1‐01‐07, P1‐01‐14,<br />

P1‐01‐19, P4‐14‐12<br />

Rowan, B<br />

P6‐08‐01<br />

Rowland, C<br />

PD10‐03<br />

Rowland, Jr., KM<br />

P1‐12‐06<br />

Roy, S<br />

PD10‐07<br />

Roy, SS PD09‐07, P5‐04‐04, P6‐04‐07<br />

Royce, M<br />

P3‐03‐01<br />

Roylance, R<br />

P6‐07‐15<br />

Rubin, P<br />

P5‐20‐08<br />

Rubio, I<br />

P1‐01‐29, OT3‐3‐06<br />

Ruckhäberle, E<br />

P2‐10‐17<br />

Rudas, M<br />

S4‐3, P2‐10‐02<br />

Ruddy, KJ<br />

P4‐01‐09<br />

Rüdiger, T S4‐5<br />

Rudlowski, C<br />

OT2‐2‐05<br />

Rudraraju, B P2‐10‐22, P3‐06‐13<br />

Rueda, OM P2‐10‐20, P3‐08‐05<br />

Ruffini, P<br />

OT2‐3‐01<br />

Rugo, H PD09‐03, P1‐01‐13, P1‐13‐01,<br />

P6‐04‐02, P6‐05‐02, P6‐13‐01<br />

Rugo, HS S3‐5, P1‐13‐05, P2‐01‐05,<br />

P2‐12‐11, P2‐12‐12, P4‐01‐13,<br />

P4‐16‐07, OT2‐3‐04<br />

Rugowski, DE<br />

P2‐04‐04<br />

Rui, H S1‐8, P2‐05‐21, P5‐01‐07<br />

Ruigrok‐Ritstier, K P3‐06‐01, P6‐04‐08<br />

Ruiz, A<br />

P2‐10‐11, OT1‐1‐09<br />

Ruiz, M<br />

P1‐07‐18, P2‐01‐06<br />

Ruiz‐Borrego, M<br />

P2‐10‐11<br />

Ruiz‐Esparza, GU<br />

P6‐11‐03<br />

Rukazenkov, Y S1‐4<br />

Rumas, N<br />

P1‐02‐01<br />

Ruppert, AS<br />

P2‐11‐07<br />

Rusby, J<br />

P1‐01‐10<br />

Rusby, JE<br />

P1‐01‐11<br />

Ruschke, K<br />

P2‐10‐38<br />

Russell, J<br />

P3‐03‐01<br />

Russell, K<br />

P3‐06‐21<br />

Russell, NS<br />

P2‐10‐42<br />

Russnes, HG<br />

P3‐05‐04<br />

Russo, L<br />

P3‐05‐02<br />

Russo, LM<br />

P5‐01‐10<br />

Rutgers, E<br />

P3‐05‐02<br />

Rutgers, EJ P3‐02‐09, P4‐11‐01<br />

Rutgers, EJT<br />

P2‐10‐42,<br />

P3‐02‐01, P4‐02‐01<br />

Ruth, K<br />

P1‐01‐16<br />

Rutkowski, T P3‐12‐09, P3‐13‐03<br />

Rutledge, J<br />

P3‐06‐28<br />

Rutqvist, L‐E<br />

P6‐14‐01<br />

Rutten, A<br />

P2‐01‐08, P2‐01‐09<br />

Ryabin, N<br />

OT1‐1‐11<br />

Ryan, J<br />

P5‐12‐03<br />

Ryan, PD<br />

PD09‐03<br />

Rydén, L PD03‐07, P2‐10‐28, P2‐10‐36<br />

Ryder, D<br />

P2‐14‐06<br />

Rye, IH<br />

P3‐05‐04<br />

Ryvo, L<br />

P5‐20‐06<br />

Saad, O<br />

P5‐18‐11<br />

Saadatmand, S<br />

P3‐02‐09<br />

Saadjian, H P2‐16‐02, P2‐16‐03<br />

Sabe, H<br />

P1‐05‐08<br />

Sabine, VS S1‐5<br />

Sabol, JL<br />

P4‐09‐12<br />

Sacchini, V<br />

OT3‐1‐01<br />

Sachdev, JC<br />

OT3‐3‐07<br />

Sackey, H<br />

P3‐07‐03<br />

Sadat, F<br />

P2‐07‐01<br />

Sadewa, AH<br />

P3‐01‐08<br />

Sadik, H<br />

P2‐09‐01, P4‐08‐08<br />

Saeki, T<br />

P4‐02‐04, P6‐04‐27<br />

Sætersdal, A<br />

P3‐05‐04<br />

Saez, R<br />

P5‐20‐10, OT3‐4‐02<br />

Safgren, S<br />

PD10‐08<br />

Safra, T<br />

P5‐20‐06<br />

Sagan, C<br />

P3‐14‐03<br />

Sagara, Y<br />

P6‐07‐16<br />

Saghatchian, M P4‐09‐05, P5‐13‐02<br />

Sahadevan, M<br />

P4‐12‐04<br />

Sahebjam, S<br />

P2‐10‐23<br />

Sahin, A<br />

P1‐07‐06<br />

Sahin, S<br />

P3‐08‐01<br />

Sahium, RC<br />

P3‐02‐14<br />

Sahmoud, T<br />

P6‐04‐02<br />

Sahoo, R<br />

PD07‐07<br />

Sai‐Girdhar, P<br />

P1‐01‐18<br />

Saini, K<br />

P3‐05‐03<br />

Sainsbury, R<br />

PD04‐08<br />

Saito, M P1‐12‐01, P4‐14‐07, P6‐07‐16<br />

Saito, Y<br />

P3‐13‐02<br />

Saiz, E<br />

P4‐09‐10<br />

Saji, S P3‐02‐06, P3‐06‐18, P6‐04‐27<br />

Sakahara, H<br />

P4‐03‐05<br />

Sakai, Y<br />

PD02‐05<br />

Sakata, S<br />

P3‐13‐04<br />

Sakr, BJ<br />

P6‐07‐34<br />

Sakr, R<br />

P5‐18‐04<br />

Sakr, RA<br />

PD04‐05, PD05‐02<br />

Salari, R<br />

PD05‐09<br />

Salat, C<br />

P5‐18‐21<br />

Salazar, LG<br />

P2‐15‐02,<br />

P5‐16‐04, OT3‐1‐02<br />

Salazar, N<br />

P5‐07‐01<br />

Saleh, MN<br />

P6‐10‐01<br />

Saleh, SM<br />

P6‐02‐03<br />

Salem, M<br />

P2‐10‐08<br />

Salgado, E<br />

P5‐16‐03<br />

Salgado, R<br />

S6‐2, P1‐07‐08,<br />

P3‐04‐10, P3‐05‐03<br />

Salihi, R<br />

P6‐07‐29<br />

Salim, A<br />

P2‐11‐06<br />

Salmon, R<br />

PD07‐04<br />

Salomon, A<br />

P1‐01‐07<br />

Salvador, J P1‐07‐18, P2‐01‐06<br />

Salvadori, B<br />

P5‐17‐06<br />

Samant, M P5‐18‐06, P5‐18‐24<br />

Sammel, MD<br />

P4‐13‐01<br />

Sampath, D P4‐07‐01, P5‐19‐02<br />

Sample, D<br />

P5‐16‐01<br />

Samra, F<br />

P4‐14‐03, P4‐17‐07<br />

Samson, S S3‐8<br />

<strong>San</strong>born, JZ<br />

P2‐06‐05<br />

<strong>San</strong>chez, F<br />

P5‐07‐06<br />

Sánchez, S<br />

P5‐03‐07<br />

<strong>San</strong>chez‐Olle, G<br />

P6‐13‐03<br />

<strong>San</strong>cho de Salas, M<br />

P1‐01‐29<br />

<strong>San</strong>dbach, J<br />

P4‐11‐04<br />

<strong>San</strong>ders, L<br />

OT3‐2‐01<br />

<strong>San</strong>ders, ME<br />

S3‐6, P4‐01‐03,<br />

P6‐05‐03, P6‐10‐05<br />

<strong>San</strong>ders, MS<br />

P2‐14‐04<br />

<strong>San</strong>dhu, H<br />

P3‐09‐05<br />

<strong>San</strong>dler, DP<br />

P3‐08‐03<br />

<strong>San</strong>ds, C<br />

P6‐08‐01<br />

<strong>San</strong>ft, T<br />

P2‐11‐08<br />

<strong>San</strong>gai, T<br />

P6‐11‐03, P6‐11‐11<br />

<strong>San</strong>Miguel, SA<br />

PD08‐05<br />

<strong>San</strong>o, Y<br />

P6‐01‐03<br />

<strong>San</strong>sano, I<br />

P1‐01‐29<br />

<strong>San</strong>tarpia, L<br />

P5‐10‐01<br />

<strong>San</strong>tiago, S<br />

P5‐07‐06<br />

<strong>San</strong>tibanez, M<br />

PD08‐06<br />

<strong>San</strong>tillana, S<br />

OT1‐1‐08<br />

<strong>San</strong>tisteban, M P5‐03‐07, P5‐16‐03<br />

<strong>San</strong>toro, M<br />

P5‐17‐06<br />

<strong>San</strong>tos, CS<br />

P6‐06‐05<br />

<strong>San</strong>tos, LFG<br />

P3‐02‐14<br />

<strong>San</strong>tos, N<br />

P4‐06‐19, P6‐04‐29<br />

<strong>San</strong>z, JL<br />

OT3‐3‐05<br />

Sapunar, F<br />

P5‐18‐21<br />

Saralli, E<br />

P1‐01‐25<br />

Saranathan, M<br />

P4‐01‐06<br />

Sareddy, GR PD09‐07, P6‐04‐07<br />

Sarode, V<br />

P1‐07‐04<br />

Sarosiek, T<br />

P5‐18‐21<br />

Sartorius, C<br />

PD01‐07<br />

Sartorius, CA<br />

P6‐05‐10<br />

Saruwatari, A<br />

P3‐06‐26<br />

Sasano, H P1‐06‐02, P1‐14‐02,<br />

P5‐01‐02, P6‐05‐14<br />

Sasi, W<br />

P1‐04‐03<br />

Sasso, M<br />

P4‐06‐09<br />

Sastre‐Garau, X PD05‐08, P1‐01‐19,<br />

P5‐01‐04, P5‐02‐03<br />

Satele, DV<br />

P2‐11‐01<br />

Sato, A<br />

P2‐05‐07, P3‐06‐29<br />

Sato, F<br />

P5‐10‐09<br />

Sato, K<br />

PD06‐02<br />

Sato, N<br />

P1‐12‐01, P1‐14‐02,<br />

P1‐14‐08, P3‐02‐02, P6‐07‐16<br />

Sato, T<br />

P3‐06‐22<br />

Sattlberger, C<br />

P5‐20‐11<br />

Sau, A<br />

PD09‐01<br />

Sauder, CAM<br />

P1‐03‐02<br />

Sauer, R<br />

P1‐14‐04<br />

Saunders, C S4‐2<br />

Saura, C<br />

P6‐10‐07, P6‐13‐03,<br />

OT2‐3‐02, OT2‐3‐06<br />

Savage, JE<br />

P6‐01‐03<br />

Savage, T<br />

P4‐04‐02<br />

Savin, M<br />

P2‐05‐06<br />

Sawaki, M P1‐02‐02, P3‐12‐06<br />

Sawyer, E<br />

P3‐08‐04<br />

Saya, H<br />

P2‐04‐01<br />

Scala, T<br />

P1‐01‐25<br />

Scaltriti, M S5‐7, P1‐07‐19, P4‐08‐02<br />

Schaefer, R<br />

P2‐10‐24<br />

Schafer, JM<br />

P6‐05‐03<br />

Schaffer, M<br />

P5‐01‐09<br />

Schaffrik, M<br />

P1‐15‐02<br />

Schaid, DJ<br />

PD10‐05<br />

Schalper, KA<br />

P3‐06‐25<br />

Schaper, C<br />

P2‐10‐02<br />

Schapira, L<br />

P3‐06‐27<br />

Schedin, P<br />

P4‐04‐03<br />

Schem, C<br />

P3‐06‐05<br />

Schenk, K<br />

P6‐08‐02<br />

Scherer, SJ<br />

P3‐06‐34<br />

Scheri, R<br />

P4‐13‐06<br />

Scheurer, M<br />

P2‐16‐02<br />

Scheurer, ME<br />

P2‐16‐03<br />

Schiff, R<br />

S5‐8, PD01‐01,<br />

PD09‐08, P4‐06‐02, P6‐04‐25,<br />

P6‐05‐12, P6‐10‐03<br />

Schimmer, AA<br />

P1‐04‐05<br />

Schindlbeck, C<br />

P2‐01‐11<br />

Schipper, R‐J<br />

P3‐02‐08<br />

Schlehe, B<br />

P6‐07‐05<br />

Schlichting, E<br />

P3‐01‐01<br />

Schlom, J<br />

P5‐16‐06<br />

Schmid, P P1‐14‐05, P4‐06‐10<br />

Schmidheiser, H<br />

P6‐08‐01<br />

Schmidt, M S1‐6, PD10‐06, P1‐13‐13,<br />

P1‐15‐02, P2‐10‐13, P2‐10‐17<br />

Schmidt, MK<br />

P3‐02‐01<br />

Schmidt, P<br />

P5‐21‐02<br />

Schmidt, T<br />

OT3‐2‐01<br />

Schmitt, M<br />

P1‐13‐13<br />

Schmitz, KJ<br />

P2‐11‐17<br />

Schnabel, CA S1‐9, P1‐07‐13, P2‐10‐15<br />

Schnabel, F P1‐09‐03, P4‐01‐07<br />

Schnadig, I<br />

P2‐11‐16<br />

Schneeweiss, A S3‐1, S3‐4, P1‐14‐01,<br />

P2‐01‐02, P3‐06‐08, P3‐06‐19,<br />

P5‐18‐26, OT1‐1‐02, OT1‐1‐10,<br />

OT1‐1‐13, OT3‐3‐11<br />

Schneider, BP<br />

PD05‐06<br />

Schneider, R<br />

P5‐20‐05<br />

Schneider, RJ<br />

P5‐03‐02<br />

Schneider, S P1‐15‐07, P6‐08‐06,<br />

P6‐11‐04<br />

Schnell, C<br />

P4‐08‐01<br />

Schnell, FM<br />

P5‐20‐08<br />

Schnitt, S<br />

PD01‐08<br />

Schoenegg, W S1‐7<br />

Scholtens, D<br />

P2‐10‐30<br />

Scholz, C<br />

P2‐01‐02<br />

Schott, AF OT2‐2‐04, OT2‐3‐01<br />

Schrader, I<br />

P2‐10‐25<br />

Schrama, JG<br />

P1‐12‐05<br />

Schrenk, P S2‐2<br />

Schubert, EK<br />

P6‐04‐03<br />

Schueller, K<br />

P2‐01‐02<br />

Schuler, LA<br />

P2‐04‐04<br />

Schulz, WC<br />

P1‐12‐04<br />

Schulz‐Wendtland, R PD07‐02<br />

Schusterbauer, C<br />

P6‐10‐02<br />

Schütte, M<br />

PD07‐02<br />

Schwab, LP<br />

P1‐05‐15<br />

Schwab, RB<br />

P2‐06‐01<br />

Schwartz, L<br />

P6‐08‐01<br />

Schwartz, MR<br />

P6‐07‐11<br />

Schwartz, MW<br />

P5‐18‐07<br />

Schwartz, S<br />

P4‐01‐07<br />

Schwartzberg, L P1‐12‐02, P5‐20‐04,<br />

P6‐09‐06, OT3‐3‐07<br />

Schwartzberg, LS P1‐15‐12, P2‐05‐06,<br />

P3‐12‐04, P5‐20‐08<br />

Schwarz, JL<br />

P6‐07‐20<br />

Schwarz, LJ<br />

P6‐07‐21<br />

Schwba, LP<br />

P5‐03‐10<br />

Scott, GK<br />

P2‐05‐04<br />

Scott, JH<br />

P2‐01‐05<br />

Scott, RJ<br />

P4‐10‐02<br />

Scott, SM<br />

P2‐16‐04<br />

Seagroves, TN P1‐05‐15, P5‐03‐10<br />

Seah, D<br />

P5‐18‐03<br />

Seah, DS<br />

P2‐16‐04, P6‐08‐03<br />

Searleman, AC S5‐6<br />

Sears, AK P5‐16‐02, P5‐16‐05<br />

Sebbagh, S<br />

P4‐13‐14<br />

Sedgewick, AJ<br />

P2‐06‐06<br />

Sediyama, CMNO<br />

P3‐07‐12<br />

Sedlacek, S<br />

P3‐06‐12<br />

Sedlackova, T<br />

P2‐01‐12<br />

Sedletcaia, A<br />

P1‐04‐02<br />

Seewaldt, V P1‐03‐01, P4‐13‐06<br />

Seferina, SC<br />

P5‐21‐04<br />

Segal‐Eiras, A<br />

P3‐07‐13<br />

Seguí, MA P1‐07‐18, P2‐01‐06<br />

Seidman, A P5‐18‐04, P6‐11‐10<br />

Seidman, AD<br />

P1‐13‐11<br />

Seif, MW<br />

P2‐14‐06<br />

Seifert, M P1‐15‐10, P2‐13‐01<br />

Seki, H<br />

P4‐09‐08<br />

Selder, S<br />

P5‐20‐01<br />

Selfors, LM<br />

P4‐08‐05<br />

Semiglazov, V S1‐11, P5‐18‐01,<br />

P5‐18‐26, OT3‐3‐01<br />

Semiglazova, T<br />

P5‐02‐02<br />

www.aacrjournals.org 603s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Sen, S<br />

P2‐04‐05<br />

Sen, SK<br />

P6‐11‐06<br />

Sener, S<br />

P4‐01‐11<br />

Sengoku, N<br />

P2‐12‐13<br />

SenGupta, SK<br />

P2‐10‐09<br />

Senn‐Reeves, JN<br />

P2‐11‐01<br />

Sensintaffar, J<br />

P6‐04‐12<br />

Senter, L<br />

P4‐13‐02<br />

Seo, S<br />

P1‐05‐03<br />

Sepkovic, DW<br />

P1‐10‐02<br />

Seppo, A<br />

P3‐05‐05, P3‐05‐06<br />

Sepúlveda, JM P5‐10‐11, OT3‐3‐05<br />

Sequeira, GR<br />

P4‐06‐15<br />

Sequist, LV<br />

P2‐01‐14<br />

Serganova, I<br />

P6‐01‐01<br />

Sergeant, J<br />

P1‐09‐05<br />

Sergenson, N S3‐3<br />

Serin, D<br />

P1‐13‐04<br />

Seroczynska, B<br />

P5‐04‐01<br />

Serra, V<br />

P1‐07‐19<br />

Serrano, D PD04‐07, PD06‐06<br />

Serrero, G P2‐10‐40, P6‐04‐19<br />

Serroyen, JL OT2‐1‐02, OT2‐1‐03<br />

Serrurier, KM P2‐12‐11, P2‐12‐12<br />

Seruga, B P2‐10‐23, P6‐09‐02<br />

Servent, V P2‐13‐10, OT3‐3‐10<br />

Serzhanova, VA<br />

P1‐05‐03<br />

Sestak, I<br />

S1‐9, P2‐10‐15<br />

Sesto, ME<br />

P2‐11‐15<br />

Setoguchi, Y<br />

OT1‐1‐01<br />

Sevenet, N<br />

PD05‐04<br />

Severson, TM<br />

PD01‐03<br />

Sevinc, A<br />

OT1‐1‐08<br />

Sexton, KR<br />

P3‐10‐01<br />

Seynaeve, C<br />

S1‐5, P1‐12‐05<br />

Sezgin, C<br />

OT1‐1‐08<br />

Sfondrini, L<br />

P5‐18‐10<br />

Sgroi, D<br />

P1‐07‐13, P2‐01‐14<br />

Sgroi, DC<br />

S1‐9, P2‐10‐15<br />

Shabaan, A<br />

P4‐01‐14<br />

Shah, AM<br />

P2‐01‐14<br />

Shah, C<br />

P1‐15‐09, P4‐16‐03<br />

Shah, KN<br />

P3‐12‐07<br />

Shah, RR<br />

P1‐07‐09<br />

Shaheen, S<br />

P4‐04‐08<br />

Shai, A<br />

P6‐14‐03<br />

Shaitelman, SF P4‐15‐02, P4‐16‐03<br />

Shak, S<br />

S1‐10, P2‐10‐08<br />

Shaker, H<br />

P1‐01‐26<br />

Shakeraneh, S<br />

P4‐13‐04<br />

Shamill, E<br />

P2‐10‐29<br />

Shan, Q<br />

P1‐01‐09<br />

Shan, T<br />

P2‐05‐10<br />

Shane, ES<br />

P6‐12‐03<br />

Shanmuganathan, S P4‐06‐14, P4‐06‐16<br />

Shao, G<br />

P6‐04‐12<br />

Shao, T P3‐08‐08, P6‐07‐34, P6‐12‐03<br />

Shao, X‐Y P2‐05‐05, P4‐09‐09<br />

Shao, Z<br />

P6‐04‐26<br />

Shao, Z‐M P1‐13‐07, P2‐06‐07,<br />

P3‐01‐05, P3‐06‐14,<br />

P6‐07‐35, P6‐07‐36, P6‐07‐42<br />

Shapira, I<br />

P5‐10‐13, P6‐07‐31<br />

Shapiro, AJ<br />

P3‐12‐02<br />

Shapiro, CL<br />

P2‐11‐04,<br />

P2‐11‐07, P5‐18‐17<br />

Shapiro, IM P6‐11‐07, P6‐11‐09<br />

Shapiro, R P4‐14‐03, P4‐17‐07<br />

Sharaiha, Y<br />

P1‐01‐18<br />

Sharma, A<br />

P1‐04‐03<br />

Sharma, AK P1‐04‐07, P1‐04‐08,<br />

P1‐04‐09<br />

Sharma, N<br />

P4‐01‐14<br />

Sharma, P<br />

S3‐7, PD09‐02<br />

Sharp, M<br />

P5‐01‐09<br />

Shastry, M P1‐14‐14, P5‐17‐05,<br />

P5‐20‐10, OT3‐3‐08<br />

Shaw, J<br />

P2‐10‐22, P3‐06‐13<br />

Shay, JW<br />

P4‐06‐06<br />

Shea, LD<br />

P6‐04‐24<br />

Shea, M<br />

PD01‐01<br />

Sheeba, I<br />

P2‐10‐29<br />

Sheehan, C<br />

PD02‐07<br />

Shen, D S5‐6<br />

Shen, H<br />

P6‐13‐02<br />

Shen, W S5‐6<br />

Shen, Y<br />

P3‐12‐04<br />

Shen, Z<br />

P6‐07‐35<br />

Shenouda, M<br />

P6‐09‐04<br />

Shepherd, L<br />

P1‐13‐01<br />

Shepherd, LE P1‐07‐13, P2‐13‐02<br />

Shepherd, SP PD09‐06, OT2‐3‐07<br />

Sherman, KA P4‐17‐04, P6‐09‐05<br />

Sherman, ME<br />

P3‐08‐02<br />

Sherman, ML P3‐06‐33, P5‐20‐03<br />

Sheshadri, N<br />

P5‐04‐09<br />

Sheth, P<br />

P4‐01‐11<br />

Shetty, G<br />

P4‐14‐11<br />

Shi, A<br />

P5‐03‐14<br />

Shi, JM<br />

P3‐08‐06<br />

Shia, A<br />

P4‐06‐10<br />

Shiau, C‐W<br />

P6‐11‐05<br />

Shiau, C‐Y<br />

P3‐12‐10<br />

Shibata, K<br />

P1‐01‐27<br />

Shidfar, A<br />

P3‐04‐08<br />

Shien, T<br />

P3‐07‐10<br />

Shigechi, T<br />

P3‐06‐26<br />

Shigekawa, T<br />

P6‐04‐27<br />

Shigematsu, H<br />

P6‐07‐26<br />

Shigeoka, Y PD07‐05, P6‐07‐39<br />

Shih, T<br />

P3‐12‐04<br />

Shih, Y‐C<br />

P3‐07‐11<br />

Shimizu, C P2‐11‐02, P6‐04‐27<br />

Shimizu, K<br />

P5‐10‐09<br />

Shimizu, S<br />

P1‐01‐20<br />

Shimizu, T<br />

OT1‐1‐01<br />

Shimozuma, K P1‐13‐10, P4‐14‐07<br />

Shin, H‐C<br />

P1‐14‐19<br />

Shin, I‐s<br />

P1‐01‐03<br />

Shin, YK<br />

P2‐05‐02<br />

Shipley, D<br />

P5‐17‐05<br />

Shiraki, N<br />

P4‐12‐06<br />

Shirato, H<br />

P1‐05‐08<br />

Shiroiwa, T<br />

P4‐14‐07<br />

Shklovskaya, J<br />

P1‐09‐07<br />

Shoemaker, M<br />

P3‐08‐04<br />

Shofoluwe, AI<br />

P6‐14‐04<br />

Shokuhi, S<br />

P1‐14‐15<br />

Shorter, R<br />

P6‐08‐01<br />

Shou, J<br />

P6‐10‐03<br />

Shparyk, Y S1‐6<br />

Shriver, CD PD02‐02, P1‐05‐09,<br />

P2‐05‐21, P3‐04‐06, P3‐04‐07,<br />

P3‐09‐03, P5‐01‐07, P6‐02‐08<br />

Shtivelband, M<br />

P6‐11‐08<br />

Shu, L<br />

P1‐05‐18<br />

Shu, S<br />

P4‐06‐01<br />

Shuai, Y<br />

P6‐07‐02<br />

Shumak, R<br />

P3‐02‐10<br />

Shumway, NM<br />

P5‐16‐05<br />

Shuping, JL<br />

P1‐01‐04<br />

Shvetsov, YB<br />

P4‐12‐02<br />

Shyamala, M S5‐7<br />

Shyr, Y‐M<br />

P3‐12‐10<br />

Sicilia, P<br />

P6‐08‐01<br />

Sicking, I<br />

P2‐10‐13<br />

Sideras, K<br />

PD10‐08<br />

Sidow, A<br />

PD05‐09<br />

Sieberova, G<br />

P2‐01‐12<br />

Sieffert, MR<br />

P3‐10‐07<br />

Siegel, PM<br />

P1‐05‐22<br />

Sieuwerts, AM P2‐01‐09, P6‐04‐08<br />

Sigal, B<br />

P1‐14‐18, P3‐06‐04<br />

Sigal‐Zafrani, B P1‐01‐19, P5‐02‐03<br />

Sigurdson, ER<br />

P1‐01‐16<br />

Sikora, MJ<br />

P6‐04‐20<br />

Sikov, W PD05‐05, P4‐01‐02, P6‐07‐34<br />

Sillerud, L<br />

P3‐03‐01<br />

Silva, DJ<br />

P2‐12‐04<br />

Silva, IV<br />

P5‐01‐15<br />

Silva, TRMA<br />

P2‐12‐04<br />

Silver, S<br />

P1‐01‐13<br />

Silvera, D<br />

P5‐03‐02<br />

Sima, CS<br />

P3‐07‐09<br />

Simak, A<br />

P3‐06‐13<br />

Simmons, P<br />

PD07‐09<br />

Simmons, S S4‐1<br />

Simoes, RV<br />

P6‐01‐01<br />

Simon, I<br />

P3‐06‐01<br />

Simon, MS<br />

PD03‐09<br />

Simon, R<br />

P6‐05‐13<br />

Simondi, C<br />

OT3‐4‐06<br />

Simone, B<br />

P6‐01‐03<br />

Simone, NL<br />

P6‐01‐03<br />

Simpson, G<br />

P3‐08‐04<br />

Simpson, PT<br />

P6‐10‐06<br />

Sims, A<br />

P3‐06‐23<br />

Sims, AH<br />

P6‐04‐09, P6‐04‐10<br />

Sing, AP S1‐10<br />

Singel, SM<br />

P4‐06‐06<br />

Singer, C<br />

P2‐10‐02,<br />

P2‐10‐31, P2‐13‐01<br />

Singer, CF<br />

S4‐3, P1‐15‐10<br />

Singer, G<br />

P4‐03‐06<br />

Singh, B<br />

P2‐10‐01<br />

Singh, G<br />

P1‐07‐06<br />

Singh, JC<br />

P5‐20‐05<br />

Singh, NK<br />

P5‐16‐06<br />

Singh, R<br />

P4‐04‐05<br />

Singh, RK<br />

P5‐07‐01<br />

Singh, S<br />

S5‐7, P4‐06‐08<br />

Singhal, S<br />

S6‐2, P1‐07‐08,<br />

P3‐05‐03, P6‐07‐22<br />

Sinha, SP<br />

P4‐01‐04<br />

Sinn, P<br />

P3‐06‐08, P3‐06‐19<br />

Sirtoli, GM<br />

P5‐01‐15<br />

Sitarik, M<br />

P2‐11‐16<br />

Sivaraman, I<br />

P4‐12‐04<br />

Sivasubramanian, PS P1‐09‐02<br />

Siziopikou, KP P5‐03‐11, OT1‐2‐01<br />

Sjöström, M<br />

P2‐10‐07<br />

Skaland, I<br />

P2‐10‐19<br />

Sklarin, N<br />

P5‐18‐20<br />

Skolny, MN P6‐09‐04, OT3‐2‐02<br />

Skoog, L<br />

P1‐05‐07, P6‐07‐12<br />

Skripkauskas, S<br />

P1‐09‐07<br />

Slaets, L S1‐11, P3‐02‐01, P3‐05‐02<br />

Slamon, D<br />

PD02‐01<br />

Slamon, DJ<br />

S1‐6, P2‐08‐02,<br />

P4‐08‐01, P4‐08‐05<br />

Sledge, G<br />

P2‐13‐02<br />

Sledge, GW<br />

PD05‐06,<br />

PD10‐04, P5‐17‐01<br />

Sledge, Jr., GW S5‐5<br />

Sleijfer, S P3‐06‐01, P6‐04‐08<br />

Slimane, K<br />

P6‐04‐14<br />

Sloane, BF<br />

P1‐05‐11<br />

Slone, S<br />

P2‐14‐05<br />

Slota, M<br />

P5‐16‐04<br />

Smalle, N<br />

P1‐13‐08<br />

Smas, ME<br />

P2‐01‐14<br />

Smeets, A<br />

P2‐10‐12,<br />

P6‐05‐05, P6‐07‐29<br />

Smeets, D<br />

P3‐05‐03<br />

Smid, M P3‐06‐32, P3‐06‐35, P6‐04‐08<br />

Smidt, M<br />

P3‐02‐08<br />

Smidt, ML<br />

P5‐01‐13,<br />

OT2‐1‐02, OT2‐1‐03<br />

Smilde, TJ P1‐14‐07, P6‐07‐32<br />

Smirnova, I S1‐4<br />

Smit, VTHBM<br />

PD07‐06<br />

Smith, BD S2‐3<br />

Smith, DA<br />

P6‐11‐10<br />

Smith, DL P5‐03‐05, P5‐03‐10<br />

Smith, I<br />

S5‐2, P5‐18‐02<br />

Smith, IE<br />

PD07‐07<br />

Smith, JA<br />

P5‐20‐05<br />

Smith, ML<br />

P6‐09‐11<br />

Smith, ND<br />

P6‐04‐12<br />

Smith, PD P4‐14‐09, P4‐17‐01<br />

Smith, R<br />

PD04‐03<br />

Smith, II, JW<br />

P4‐11‐04<br />

Smitt, M<br />

P5‐18‐06<br />

Smorenburg, CH<br />

P1‐12‐05<br />

Snider, J<br />

P6‐07‐10<br />

Snider, JE<br />

P2‐10‐01<br />

Snider, JN<br />

OT3‐3‐07<br />

Sninsky, J<br />

PD10‐03<br />

Snuderl, M<br />

P3‐12‐03<br />

Snyder, KA<br />

P1‐05‐05<br />

Soares, C<br />

OT1‐1‐08<br />

Soares, F<br />

P4‐02‐03<br />

Soares, FA P1‐06‐03, P2‐05‐17<br />

Soeling, U<br />

P2‐10‐25<br />

SoFEA and EFECT<br />

Investigators<br />

P2‐14‐01<br />

Sohl, J<br />

P2‐16‐04, P5‐18‐03<br />

Sohn, J<br />

PD09‐05<br />

Sola, JJ<br />

P5‐16‐03<br />

Solbach, C<br />

P2‐10‐13<br />

Soler, CMJ<br />

P6‐04‐08<br />

Solid, C<br />

P1‐15‐01<br />

Soliman, H<br />

OT3‐4‐01<br />

Solin, LJ<br />

P5‐15‐01<br />

Solomayer, EF<br />

OT1‐1‐10<br />

Solomon, S<br />

OT3‐1‐01<br />

Soloway, MG S6‐1<br />

Solti, M<br />

P4‐11‐04<br />

Soltys, SG<br />

P4‐16‐11<br />

Somaiah, N<br />

P3‐06‐09<br />

Somarelli, J<br />

P4‐06‐07<br />

Somera, AL<br />

OT2‐1‐04<br />

Somlo, G<br />

P2‐14‐08, P3‐06‐11<br />

Sommariva, M<br />

P5‐18‐10<br />

Sommer, HL<br />

P2‐01‐02<br />

Son, BH<br />

P3‐01‐02, P3‐12‐05<br />

Son, G‐T<br />

P1‐01‐05, P3‐03‐02<br />

Song, BJ<br />

P2‐05‐02, P4‐03‐01<br />

Song, D<br />

P2‐05‐19<br />

Song, J<br />

P3‐07‐05<br />

Song, X<br />

P2‐11‐10, P3‐04‐11,<br />

P3‐13‐05, P5‐01‐01<br />

Song, Y P3‐12‐03, P4‐03‐12, P5‐01‐16<br />

Sonis, ST<br />

P1‐15‐12<br />

Sonoo, H<br />

P2‐10‐18<br />

Sood, AK<br />

P5‐10‐03<br />

Soori, GS<br />

P1‐12‐06<br />

Sorensen, BS<br />

P6‐04‐18<br />

Sørlie, T<br />

P3‐04‐03<br />

Sosa, EV<br />

P2‐01‐05<br />

Sosinska‐Mielcarek, K P3‐12‐09,<br />

P3‐13‐03<br />

Sotiriou, C S4‐4, S6‐2, P1‐07‐08,<br />

P3‐04‐10, P3‐05‐03,<br />

P6‐07‐14, P6‐07‐22<br />

Soubeyrand, M‐S<br />

P5‐21‐01<br />

Souichirou, I<br />

P4‐04‐08<br />

Souleimanova, M<br />

P6‐02‐03<br />

Soulié, P<br />

P1‐14‐21, OT3‐3‐04<br />

Soundararajan, A<br />

P4‐06‐08<br />

Spagnoli, GC<br />

P4‐04‐04<br />

Span, P<br />

P3‐06‐01<br />

Spano, J‐P<br />

P5‐17‐04<br />

Specht, JM<br />

P6‐04‐03<br />

Specht, M<br />

P3‐06‐27<br />

Specht, MC<br />

OT3‐2‐02<br />

Speck, RM<br />

P4‐13‐01<br />

Spector, N<br />

P4‐08‐07<br />

Spector, NL<br />

P4‐08‐03<br />

Spector, T<br />

OT3‐3‐01<br />

Speer, V<br />

P1‐14‐04<br />

Speers, C PD04‐02, P4‐16‐01,<br />

P4‐16‐02, P4‐16‐04<br />

Speirs, C<br />

P1‐05‐13<br />

Speirs, V<br />

P6‐02‐06<br />

Spencer, P<br />

P3‐13‐05<br />

Sperinde, J P2‐10‐16, P2‐10‐31<br />

Sperk, E S4‐2<br />

Speyer, J<br />

P5‐20‐05<br />

Spicer, D<br />

P1‐09‐06<br />

Spoelstra, NS<br />

P5‐10‐04,<br />

P5‐10‐05, P5‐10‐06<br />

Sposato, V<br />

P2‐16‐01<br />

Sposto, R<br />

P4‐01‐11<br />

Spraggs, CF<br />

PD10‐05<br />

Spyratos, F<br />

PD07‐04<br />

Squires, M<br />

OT2‐2‐03<br />

Sridhara, R S1‐11<br />

Srikrishnan, R<br />

P4‐06‐08<br />

Sroussi, J<br />

P1‐01‐19<br />

Stacy, T<br />

P6‐14‐05<br />

Stadler, ZK<br />

P2‐16‐04<br />

Staebler, A S2‐2<br />

Stål, O<br />

P2‐10‐28, P6‐07‐12<br />

Stallard, S<br />

P4‐14‐13<br />

Stallings‐Mann, ML P6‐06‐01<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

604s<br />

<strong>Cancer</strong> Research


December 4-8, 2012<br />

Author Index<br />

Stamatovic, L<br />

P5‐02‐02<br />

Stambolic, V PD03‐02, PD03‐05<br />

Stampanoni, M<br />

P4‐03‐06<br />

Standish, L<br />

OT3‐1‐02<br />

Stanford, JC<br />

P6‐02‐01<br />

Starczynski, J<br />

P1‐07‐17<br />

Stark, A<br />

P4‐13‐12<br />

Stark, G<br />

P1‐04‐06<br />

Staroslawska, E<br />

P5‐18‐21<br />

Starr, A<br />

P2‐05‐06<br />

START Trialists S4‐1<br />

Stasi, I<br />

P5‐17‐06<br />

Staudigl, C<br />

P1‐15‐10<br />

Stearns, V PD09‐03, PD10‐08,<br />

P3‐04‐08, P4‐12‐04,<br />

P6‐05‐02, P6‐07‐33<br />

Stebbing, J P1‐04‐04, P6‐04‐15<br />

Stecklein, S<br />

S3‐7, PD09‐02<br />

Steding, CE<br />

P5‐18‐13<br />

Steding‐Jessen, M<br />

P1‐09‐01<br />

Steelman, L<br />

OT2‐3‐11<br />

Steffens, C‐C<br />

P1‐15‐02<br />

Steger, G S6‐5<br />

Stein, R S3‐3<br />

Steinberg, MS<br />

P3‐06‐12<br />

Steinberg, SM<br />

P5‐16‐06<br />

Steiner, M<br />

P6‐14‐03<br />

Steiner, P<br />

P4‐07‐05<br />

Steinkohl, O P5‐14‐06, P5‐15‐04<br />

Stella, PJ<br />

P1‐12‐06<br />

Stemmer, SM P5‐17‐03, P5‐20‐01<br />

Stenvang, J P3‐06‐32, P3‐06‐35<br />

Stepansky, A<br />

P3‐14‐01<br />

Stephant, G<br />

P3‐06‐24<br />

Stephens, P PD02‐07, PD05‐01<br />

Stephens, PJ S3‐6<br />

Stern, DF<br />

P3‐14‐01<br />

Stern, LH P4‐02‐09, P4‐02‐10<br />

Stern, M‐H<br />

P5‐02‐03<br />

Stessin, A<br />

P4‐03‐09<br />

Stevens, M<br />

P2‐14‐05<br />

Stevens, MG<br />

P6‐04‐08<br />

Steyerberg, EJ<br />

P4‐11‐05<br />

Stineman, MG<br />

P4‐13‐01<br />

Stitt, L<br />

P2‐13‐08<br />

Stockerl‐Goldstein, K S3‐5<br />

Stoeber, K<br />

PD04‐08<br />

Stokoe, C<br />

P3‐06‐12<br />

Stoller, RG<br />

PD09‐06<br />

Stolley, M<br />

P5‐13‐03<br />

Stone, A<br />

P6‐08‐06<br />

Stopeck, A<br />

P6‐10‐01<br />

Stoppa‐Lyonnet, D<br />

P5‐02‐03<br />

Storer, CL<br />

P5‐07‐02<br />

Storhoff, J<br />

P2‐10‐02<br />

Stork‐Sloots, L P3‐05‐01, P3‐05‐02,<br />

P3‐06‐11, OT3‐4‐01,<br />

OT3‐4‐02, OT3‐4‐03<br />

Storniolo, AMV<br />

P1‐03‐02<br />

Storz, P<br />

P1‐05‐24<br />

Stott, SL<br />

P2‐01‐14<br />

Stout, NL<br />

P2‐11‐13, P2‐11‐14<br />

Stover, AC<br />

P4‐16‐07<br />

Strand, C<br />

P2‐10‐19, P2‐10‐28<br />

Strang, P<br />

P2‐12‐09<br />

Strasak, A<br />

P5‐18‐05<br />

Straube, W<br />

P6‐13‐01<br />

Stravodimou, A<br />

P6‐04‐05<br />

Strickland, S<br />

P1‐15‐07<br />

Strigel, R<br />

P3‐02‐11<br />

Strina, C<br />

PD07‐03<br />

Strobbe, LJA P5‐01‐13, OT2‐1‐02,<br />

OT2‐1‐03<br />

Strzelecka, M P3‐12‐09, P3‐13‐03<br />

Stuller, KA<br />

P2‐11‐04<br />

Sturge, J<br />

P2‐02‐03<br />

Sturrock, M<br />

P5‐05‐06<br />

Styles, AL<br />

P5‐18‐12<br />

Su, F<br />

P1‐01‐09<br />

Su, J‐C<br />

P6‐11‐05<br />

Su, N<br />

PD02‐04<br />

Suarez, A<br />

P1‐14‐10, P5‐18‐15<br />

Subbarayalu, P<br />

P5‐10‐16<br />

Sue, GR<br />

P3‐14‐05<br />

Suen, DTK<br />

P3‐11‐02<br />

Suganuma, N<br />

P1‐01‐20<br />

Sugiura, H<br />

P4‐12‐06<br />

Sukenick, G<br />

P6‐01‐01<br />

Sukov, WR<br />

PD02‐05<br />

Sukumar, S P2‐02‐01, P2‐09‐01,<br />

P3‐04‐08, P4‐08‐08, P4‐12‐04,<br />

P6‐04‐13<br />

Sullivan, R<br />

P2‐04‐04<br />

Suman, V<br />

PD07‐01, PD10‐08<br />

Suman, VJ S2‐1, S5‐5<br />

Summa, B<br />

OT3‐2‐01<br />

Sun, G<br />

P4‐03‐12, P5‐01‐16<br />

Sun, L P1‐05‐18, P2‐04‐07, P4‐03‐12,<br />

P5‐01‐16, P5‐03‐04, P5‐03‐06,<br />

P5‐04‐04, P6‐11‐10<br />

Sun, S<br />

P6‐11‐01<br />

Sun, W<br />

P4‐14‐09, P4‐17‐01<br />

Sun, X<br />

P2‐10‐16<br />

Sun, X‐Y<br />

P1‐14‐17<br />

Sun, Y PD09‐09, P3‐07‐05, P5‐19‐03,<br />

P6‐04‐11<br />

Sun, Z<br />

P1‐05‐24<br />

Sundaram, S<br />

P6‐02‐09<br />

Sung, J<br />

OT3‐1‐01<br />

Sunpaweravong, P<br />

P5‐18‐01<br />

Suter, T S6‐5<br />

Sutterlin, M S4‐2<br />

Suwa, H<br />

P3‐13‐04<br />

Suzuki, K P4‐09‐08, P6‐07‐23<br />

Suzuki, T<br />

P4‐03‐05, P6‐05‐14<br />

Suzuki, Y P2‐05‐08, P3‐13‐02<br />

Svedman, C<br />

P6‐07‐03<br />

Swain, S<br />

P5‐18‐11<br />

Swain, SM<br />

S1‐11, S5‐1, S5‐5,<br />

P5‐18‐01, P5‐18‐26<br />

Swanton, C P6‐07‐15, OT1‐1‐14<br />

Sweeney, KJ P1‐01‐15, P5‐10‐07<br />

Sweep, F<br />

P3‐06‐01<br />

Sweep, FCGJ<br />

P1‐13‐13<br />

Swerdlow, A<br />

P3‐08‐04<br />

Sydenham, M<br />

S4‐1, P3‐06‐09<br />

Syldor, A<br />

P5‐18‐04, P5‐18‐20<br />

Symmans, F P1‐07‐08, P2‐10‐17<br />

Symmans, WF S2‐1, P1‐07‐06,<br />

P1‐07‐09, P2‐10‐10, P3‐06‐14<br />

Szade, J<br />

P5‐04‐01<br />

Szalkucki, L<br />

P3‐02‐11<br />

Szallasi, Z<br />

P6‐07‐15<br />

Szasz, AM<br />

P3‐05‐08<br />

Szostakiewicz, B<br />

P2‐10‐31<br />

Sztupinszki, Z<br />

P2‐10‐24<br />

T. Dorsam, R P1‐05‐23<br />

Taback, B S2‐1, PD03‐03, P4‐02‐07<br />

Tabarsi, N<br />

P4‐16‐01<br />

Tabouret, E<br />

P3‐12‐11<br />

Tafra, L<br />

OT2‐1‐04<br />

Taghian, AG P6‐09‐04, OT3‐2‐02<br />

Tagliabue, E P4‐06‐09, P5‐18‐10<br />

Taioli, E<br />

P5‐10‐13<br />

Taira, N<br />

P3‐07‐10, P4‐14‐07<br />

Tait, N<br />

P2‐13‐05<br />

Takada, M P3‐06‐18, P3‐13‐04<br />

Takagi, K<br />

P6‐05‐14<br />

Takahashi, F<br />

P1‐14‐02<br />

Takahashi, M P1‐14‐08, P1‐14‐09,<br />

P3‐02‐06, P3‐06‐22,<br />

P4‐03‐03, P4‐09‐08, P6‐07‐39<br />

Takahashi, S<br />

P4‐12‐06<br />

Takahashi, Y<br />

P3‐02‐02<br />

Takahasi, S<br />

P2‐10‐43<br />

Takamoto, Y<br />

P2‐04‐01<br />

Takanaka, T<br />

P4‐16‐13<br />

Takano, T<br />

P1‐14‐08<br />

Takao, S<br />

PD07‐05, P5‐18‐16,<br />

P6‐07‐30, P6‐07‐39<br />

Takata, D<br />

P2‐05‐07, P3‐06‐29<br />

Takatsuka, Y P2‐13‐04, P2‐13‐07<br />

Takei, H<br />

P1‐14‐06, P2‐10‐18,<br />

P5‐15‐06, P6‐04‐17<br />

Takei, J<br />

P6‐07‐23<br />

Takeyama, H P4‐03‐10, P5‐04‐02<br />

Takeyoshi, I P1‐14‐06, P2‐05‐07,<br />

P3‐06‐29<br />

Takizawa, S<br />

P5‐10‐09<br />

Talavera, GG<br />

PD08‐05<br />

Talcott, S<br />

P5‐07‐03<br />

Talcott, SM<br />

P5‐07‐03<br />

Tallet, A<br />

P3‐12‐11<br />

Tamaki, K P1‐06‐02, P6‐05‐14<br />

Tamaki, N<br />

P1‐06‐02<br />

Tamimi, RM<br />

P5‐01‐03<br />

Tamura, K<br />

P3‐02‐06<br />

Tamura, N<br />

P2‐11‐02<br />

Tan, B<br />

P3‐08‐09<br />

Tan, CH<br />

P4‐16‐12<br />

Tan, M‐H<br />

P3‐08‐09<br />

Tan, PH<br />

P4‐16‐12<br />

Tan, P‐H<br />

P3‐08‐09<br />

Tan, Q<br />

P4‐09‐04<br />

Tan, Y PD09‐08, P4‐07‐04, P6‐04‐25<br />

Tanaka, F<br />

P3‐13‐04<br />

Tanaka, RMN<br />

P3‐01‐07<br />

Tanaka, S<br />

P3‐02‐02, P3‐09‐04<br />

Tandon, V<br />

P4‐04‐01<br />

Tanei, T<br />

P5‐03‐03<br />

Tang, G S1‐10<br />

Tang, J<br />

P4‐08‐10, P5‐10‐10<br />

Tang, L<br />

P6‐05‐03<br />

Tang, S<br />

S1‐11, P4‐01‐11<br />

Tani, T<br />

P1‐15‐11<br />

Taniguchi, H<br />

P1‐01‐27<br />

Tanimoto, A<br />

P4‐03‐03<br />

Tanino, H<br />

PD07‐05<br />

Tanioka, M<br />

P6‐07‐30<br />

Tannock, I<br />

P2‐05‐12<br />

Tannock, IF<br />

P6‐09‐02<br />

Tannous, BA<br />

P3‐12‐03<br />

Tao, J S5‐7<br />

Tao, S<br />

P5‐15‐07<br />

Taran, T<br />

P6‐04‐02<br />

Tarco, E<br />

P1‐07‐06<br />

Tarpin, C<br />

OT2‐3‐05<br />

Tarter, P<br />

P4‐15‐03<br />

Tas, P<br />

P3‐14‐03<br />

Tashima, R<br />

PD06‐07<br />

Tatla, R<br />

P2‐12‐07<br />

Tauer, KW<br />

P5‐20‐08<br />

Tautchin, M<br />

P5‐16‐01<br />

Tawbi, H<br />

PD09‐06<br />

Tawfik, OW<br />

PD09‐02<br />

Taylor, CT<br />

OT1‐1‐05<br />

Taylor, JA<br />

P3‐08‐03<br />

Taylor, NJ<br />

P1‐06‐01<br />

Tea, M‐K<br />

P1‐15‐10<br />

Teh, BS<br />

P6‐07‐11<br />

Teh, B‐T<br />

P3‐08‐09<br />

Teixeira, SF<br />

P6‐11‐13<br />

Tejedo, S<br />

P5‐13‐03<br />

Tekmal, RR P5‐04‐04, P6‐04‐07<br />

Telang, NT<br />

P1‐10‐02<br />

Telli, ML<br />

PD09‐04<br />

Tembo, O<br />

P1‐14‐03<br />

Tenniswood, MPR<br />

P3‐10‐02<br />

Teo, WW<br />

P2‐02‐01, P6‐04‐13<br />

Tepperberg, J<br />

P3‐10‐10<br />

Ter Haar‐van Eck, SA P6‐08‐02<br />

Terada, M<br />

P3‐13‐02<br />

Terao, M<br />

P3‐13‐02<br />

Tereffe, W P3‐10‐07, P4‐15‐02<br />

Terhaar, AR<br />

P2‐11‐15<br />

Terrell, KL<br />

P5‐10‐04<br />

Tesch, H<br />

S4‐5, PD06‐01,<br />

P1‐14‐01, P2‐10‐25<br />

Tessari, A<br />

P2‐12‐08<br />

Tevaarwerk, AJ<br />

P2‐11‐15,<br />

P3‐02‐11, P6‐12‐02<br />

TEX Study Group<br />

P1‐05‐07<br />

Thakur, S<br />

P4‐09‐07<br />

Theodoropoulos, PA P2‐01‐07<br />

Theodoulou, M<br />

P5‐18‐20<br />

Therrien, B S6‐3<br />

Thery, J‐C<br />

P5‐17‐04<br />

Theunissen, EB<br />

P4‐11‐01<br />

Thomalla, J<br />

P2‐11‐11<br />

Thomas, F<br />

PD03‐09<br />

Thomas, J<br />

OT3‐3‐09<br />

Thomas, M S6‐7<br />

Thomassin‐Piana, J<br />

P5‐03‐01<br />

Thompson, A S4‐2, P1‐05‐14, P5‐05‐02<br />

Thompson, AE<br />

P1‐05‐24<br />

Thompson, AM PD03‐02, P4‐06‐10,<br />

P5‐05‐05, P5‐05‐06<br />

Thompson, D<br />

P4‐11‐05<br />

Thompson, EA<br />

PD10‐04<br />

Thompson, J<br />

P2‐13‐05<br />

Thompson, MO<br />

P4‐01‐12<br />

Thompson, PA<br />

PD08‐04<br />

Thompson, SL<br />

P2‐11‐01<br />

Thomson, E P2‐14‐07, P5‐20‐12<br />

Thomssen, C<br />

S3‐4, P1‐13‐13,<br />

P2‐10‐08, P2‐10‐38<br />

Thor, AD<br />

P5‐10‐04, P5‐10‐05<br />

Thorat, M<br />

PD04‐08<br />

Thorén, L<br />

PD10‐09<br />

Thorne, A<br />

OT3‐1‐01<br />

Thorsen, C<br />

P4‐11‐03<br />

Thorsen, CM P3‐02‐12, P4‐01‐15<br />

Thuler, LCS P3‐11‐04, P6‐07‐44<br />

Thummala, AR S1‐6<br />

Thune, I<br />

P3‐01‐01<br />

Thürlimann, B S1‐1<br />

Thyparambil, S<br />

P1‐07‐19<br />

Tibbitts, P<br />

P6‐04‐21<br />

Tice, D<br />

P4‐07‐05<br />

Tice, J<br />

P4‐13‐05<br />

Tiersten, A<br />

P5‐20‐05<br />

Tilanus‐Linthorst, MM P3‐02‐09<br />

Tilanus‐Linthorst, MMA P4‐11‐05<br />

Tiller, GE<br />

P3‐08‐06<br />

Tillett, R<br />

P4‐17‐03<br />

Timar, J<br />

P2‐10‐24<br />

Timcheva, C<br />

P5‐02‐02<br />

Timmerman, D<br />

OT2‐2‐02<br />

Timmermans, M P3‐06‐32, P3‐06‐35<br />

Timms, K<br />

PD09‐04<br />

Timotheadou, E<br />

P1‐07‐15<br />

Timpson, P<br />

P1‐05‐13<br />

Tin, S P2‐03‐01, P2‐10‐32, P5‐04‐06<br />

Ting, DT<br />

P2‐01‐14<br />

Tirona, KM P6‐09‐01, P6‐09‐02<br />

Tizon, X<br />

P4‐02‐06<br />

Tjan‐Heijnen, VCG P1‐14‐07,<br />

P5‐21‐04, P6‐07‐32,<br />

OT2‐1‐02, OT2‐1‐03<br />

Tkaczuk, K<br />

P2‐13‐05<br />

Tkaczuk, KR<br />

P2‐10‐40<br />

Tobias, JS S4‐2<br />

Tobin, H<br />

P2‐04‐03<br />

Tobin, NP<br />

PD06‐08<br />

Tobler, J<br />

OT1‐1‐08<br />

Toelg, C<br />

P1‐05‐20<br />

Toesca, A<br />

PD06‐06<br />

Toffoli, S<br />

P3‐04‐04<br />

Toi, M<br />

S6‐5, P1‐14‐02,<br />

P1‐14‐08, P3‐06‐18, P3‐13‐04,<br />

P5‐01‐02, P5‐10‐09<br />

Tokes, A‐M<br />

P3‐05‐08<br />

Tokin, CA<br />

P4‐01‐13<br />

Tokiniwa, H P2‐05‐07, P3‐06‐29<br />

Tokuda, Y<br />

P2‐05‐08,<br />

P3‐13‐02, P5‐18‐16<br />

Tolaney, S<br />

P6‐05‐02<br />

Toledo, C<br />

P4‐02‐03<br />

Tollenaar, RA<br />

P3‐02‐09<br />

Toller, I<br />

P5‐18‐25<br />

Tomczyk, K<br />

P3‐08‐04<br />

Tomic, S<br />

P4‐04‐04<br />

Tomida, K<br />

P1‐15‐11<br />

Tomita, S<br />

P2‐14‐03<br />

Tomlinson, DC<br />

P6‐02‐06<br />

Tomlinson, I<br />

P3‐08‐04<br />

Toner, M<br />

P2‐01‐14<br />

Tonetti, DA<br />

P6‐04‐24<br />

Tong, L<br />

P4‐08‐01<br />

Toriumi, Y P4‐03‐10, P5‐04‐02<br />

Torrecabota, J<br />

P3‐12‐08<br />

Torrejon‐Castro, D<br />

P6‐13‐03<br />

Torres, A<br />

P5‐07‐06<br />

Torres, R S1‐4<br />

Tost, J<br />

P5‐05‐03<br />

Tosteson, AN<br />

P4‐01‐15<br />

www.aacrjournals.org 605s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Totobenazara, J‐L<br />

P4‐16‐10<br />

Toy, KA<br />

P1‐04‐11<br />

Toyama, T<br />

P4‐12‐06<br />

Tozuka, K P2‐05‐07, P3‐06‐29<br />

Traina, T<br />

PD09‐03<br />

Traina, TA P1‐13‐11, P6‐05‐01,<br />

P6‐05‐02, P6‐11‐10<br />

Tralau‐Stewart, C<br />

P4‐06‐12<br />

Tramm, T<br />

P3‐04‐03<br />

Trappey, AF<br />

P5‐16‐05<br />

Trassard, M<br />

PD07‐04<br />

Tredan, O<br />

P3‐06‐20<br />

Treekitkarnmongkol, W P2‐04‐05<br />

Tresca, P<br />

OT3‐4‐06<br />

Triolo, R<br />

P5‐01‐10<br />

Tripathi, M<br />

P4‐02‐02<br />

Tripathy, D<br />

P4‐01‐11<br />

Trippel, M<br />

P4‐03‐06<br />

Triulzi, T<br />

P4‐06‐09, P5‐18‐10<br />

Trivedi, MV<br />

S5‐8, P4‐06‐02<br />

Troester, MA<br />

P6‐02‐09<br />

Tromberg, B<br />

P4‐02‐04<br />

Troxell, M<br />

PD05‐09<br />

Troxell, ML<br />

P2‐08‐03<br />

Trucu, D<br />

P1‐05‐14<br />

Trudeau, M P2‐12‐03, P5‐18‐14<br />

Truglia, M<br />

P5‐10‐01<br />

Tsai, K<br />

P3‐07‐08<br />

Tsai, W‐Y<br />

P2‐11‐03<br />

Tsai, Y‐CS<br />

P4‐16‐05<br />

Tsai, Y‐F<br />

P3‐12‐10<br />

Tse, CH<br />

P1‐07‐01<br />

Tseng, L‐M<br />

P3‐12‐10,<br />

P6‐11‐05, OT1‐1‐17<br />

Tsimberidou, A<br />

P5‐20‐09<br />

Tsou, M‐H<br />

P4‐16‐05<br />

Tsuboi, M<br />

P1‐14‐06<br />

Tsuda, B<br />

P2‐05‐08, P3‐13‐02<br />

Tsuda, H<br />

P1‐01‐12,<br />

P5‐01‐02, P6‐04‐27<br />

Tsugawa, K<br />

P1‐01‐12<br />

Tsujimoto, G<br />

P5‐10‐09<br />

Tsujimoto, M<br />

P2‐10‐18<br />

Tsukamoto, F<br />

P2‐10‐18<br />

Tsuyuki, S P1‐14‐08, P3‐13‐04<br />

Tubbs, RR PD02‐04, P6‐07‐45<br />

Tubiana‐Mathieu, N P3‐06‐20,<br />

OT3‐4‐05<br />

Tuchman, L<br />

P6‐08‐01<br />

Tucker, N<br />

P2‐12‐05<br />

Tullis, I<br />

P2‐10‐29<br />

Tun, MT<br />

P2‐11‐15<br />

Tunon de Lara, C PD05‐04, P1‐01‐21,<br />

P3‐14‐03, OT2‐1‐01<br />

Turdo, F<br />

P4‐06‐09<br />

Turk, M<br />

P3‐05‐01<br />

Turley, EA<br />

P1‐05‐20<br />

Turman, YJ S6‐1<br />

Turnbull, A<br />

P3‐06‐23<br />

Turnbull, AK P6‐04‐09, P6‐04‐10<br />

Turner, B<br />

OT3‐4‐04<br />

Turnwald, B<br />

P2‐11‐16<br />

Turpeenniemi‐Hujanen, T PD10‐06<br />

Tursz, T<br />

P4‐09‐05<br />

Tusquets, I P1‐07‐18, P2‐01‐06<br />

Tuttle, K<br />

P2‐10‐40<br />

Tuyls, S<br />

P2‐10‐12, P6‐05‐05<br />

Tweardy, D<br />

P5‐03‐08<br />

Twelves, C<br />

S6‐6, P6‐11‐14<br />

Tyldesley, S<br />

PD04‐02,<br />

P4‐16‐01, P4‐16‐02<br />

Tzeng, C‐H<br />

P3‐12‐10<br />

Tzonis, P<br />

P5‐16‐05<br />

Uchida, K P4‐03‐10, P5‐04‐02<br />

Uchida, S<br />

P2‐05‐07<br />

Uchiyama, K P3‐02‐02, P3‐09‐04<br />

Udovitsa, D<br />

OT1‐1‐17<br />

Ueda, N<br />

P3‐06‐26<br />

Ueda, S<br />

P4‐02‐04<br />

Ueda, Y<br />

P4‐03‐05<br />

Uehara, K<br />

P1‐06‐02<br />

Ueno, N<br />

P3‐10‐09<br />

Ueno, NT PD03‐06, PD03‐08, P2‐03‐01,<br />

P2‐10‐32, P3‐10‐01, P3‐10‐05,<br />

P3‐10‐10, P5‐03‐05, P5‐03‐09,<br />

P5‐03‐10, P5‐04‐06, P5‐10‐02,<br />

P5‐20‐13, OT2‐3‐10<br />

Ueno, T P1‐14‐02, P1‐14‐08, P3‐06‐18,<br />

P5‐01‐02, P5‐10‐09<br />

Uhlén, M<br />

PD03‐07<br />

Uhrhammer, N<br />

P3‐06‐20<br />

Uleer, C<br />

P2‐10‐08<br />

Ullman, E<br />

P5‐04‐09<br />

Umbricht, C<br />

P2‐02‐01<br />

Umeda, T<br />

P1‐15‐11<br />

Umphrey, H<br />

P3‐03‐03<br />

Uncu, D<br />

OT1‐1‐08<br />

Unger, HA<br />

P1‐04‐02<br />

Untch, M S1‐11, S2‐2, S3‐1, S3‐4,<br />

S5‐2, P1‐14‐01, P3‐06‐05, P5‐18‐02,<br />

P5‐18‐11, OT1‐1‐13, OT3‐3‐11<br />

Untch, S<br />

OT3‐4‐01<br />

Unzeitig, G<br />

PD07‐01<br />

Unzeitig, GW<br />

P4‐14‐01<br />

Upadhyaya, G<br />

P3‐06‐28<br />

Urban, P<br />

P6‐11‐08<br />

Urist, M<br />

P1‐01‐06, P3‐03‐03<br />

Urist, MM<br />

P5‐17‐07<br />

Urruticoechea, A P6‐11‐02, P6‐11‐08<br />

Ursin, G<br />

P3‐01‐01<br />

Ursini‐Siegel, J<br />

P1‐05‐22<br />

Ushio, A<br />

P1‐02‐02, P3‐12‐06<br />

Uslu, R<br />

OT1‐1‐08<br />

Utama, FE S1‐8<br />

Utsumi, T<br />

P3‐02‐05<br />

Uttenreuther‐Fischer, M OT1‐1‐16,<br />

OT1‐1‐17<br />

Uzan, C<br />

P4‐12‐01, P4‐14‐12<br />

Uzan, S<br />

P4‐14‐12<br />

Vach, W<br />

P4‐09‐04<br />

Vachon, CM<br />

P4‐12‐03<br />

Vadakkan, T<br />

P4‐02‐08<br />

Vadlamudi, RK PD09‐07, P1‐05‐10,<br />

P5‐04‐04, P6‐04‐07<br />

Vahdat, L P1‐12‐02, P1‐15‐07,<br />

P5‐20‐04, P6‐11‐14<br />

Vahdat, LT P6‐10‐01, P6‐11‐04<br />

Vaidya, JS S4‐2<br />

Valachis, A<br />

P5‐18‐18<br />

Valadão, IC<br />

P6‐11‐13<br />

Valagussa, P S1‐11, S6‐7<br />

Valdés Olmos, RA<br />

P4‐02‐01<br />

Valdimarsdottir, H<br />

P4‐11‐01<br />

Valencia, A<br />

P5‐12‐03<br />

Valent, A<br />

P2‐08‐04<br />

Valente, AL<br />

PD02‐02<br />

Valero, V P2‐10‐32, P3‐10‐05, P5‐10‐02,<br />

P5‐15‐02, P5‐20‐02, P5‐20‐13,<br />

OT2‐3‐10<br />

Vallejos, CS P6‐07‐20, P6‐07‐21<br />

Vallier, A‐L OT1‐1‐03, OT3‐3‐09<br />

Valo, I<br />

P1‐14‐21<br />

van Asperen, CJ<br />

P4‐11‐05<br />

Van Bree, R<br />

P2‐13‐06<br />

Van Calster, B P2‐10‐12, P6‐07‐04,<br />

P6‐07‐05, P6‐07‐19, OT2‐2‐02<br />

Van Calsteren, K<br />

P6‐07‐05<br />

van Dalen, T P4‐11‐01, P4‐14‐08,<br />

OT2‐1‐02, OT2‐1‐03<br />

van Dam, PA P2‐01‐08, P2‐01‐09<br />

van Dam, P‐J<br />

P2‐01‐08<br />

van de <strong>San</strong>dt, L<br />

P2‐10‐13<br />

van de Velde, CJH S1‐5, PD07‐06<br />

van de Ven, AL<br />

P6‐11‐12<br />

van de Ven, S<br />

PD07‐06<br />

Van de Vijver, KKBT P5‐01‐13<br />

van de Wouw, AJ P5‐21‐04, P6‐07‐32<br />

van Decker, S<br />

P6‐08‐01<br />

van den Akker, J<br />

P3‐05‐02<br />

van den Berkmortel, F P5‐21‐04,<br />

P6‐07‐32<br />

van den Bosch, MA P4‐01‐17,<br />

P4‐14‐08, P5‐01‐11,<br />

P5‐01‐14, P5‐15‐03<br />

Van den Eynden, GG P2‐01‐08,<br />

P2‐01‐09<br />

van den Tol, P<br />

PD04‐01<br />

Van den Wyngaert, T P3‐13‐01<br />

van der Groep, P P2‐06‐02, P5‐01‐06,<br />

P6‐05‐08<br />

van der Hage, J OT2‐1‐02, OT2‐1‐03<br />

van der Luijt, RB<br />

P4‐11‐01<br />

van der Pol, CC<br />

P5‐01‐11<br />

van der <strong>San</strong>den‐Melis, J P4‐11‐01<br />

van der Wall, E P2‐06‐02, P5‐01‐06,<br />

P6‐05‐08<br />

van Deurzen, CHM<br />

P5‐01‐13<br />

van Diest, PJ PD02‐03, P2‐06‐02,<br />

P4‐01‐17, P5‐01‐06, P5‐01‐11,<br />

P5‐01‐14, P6‐05‐08, P6‐05‐13<br />

van Gastel, SM<br />

P1‐14‐07<br />

van Geer, M<br />

P1‐15‐05<br />

van Goethem, M<br />

P3‐02‐08<br />

van Golen, CM<br />

P3‐10‐04<br />

van Golen, K P1‐04‐10, P6‐06‐03<br />

van Golen, KL P3‐10‐04, P3‐10‐08<br />

van Hillegersberg, R P4‐01‐17,<br />

P5‐01‐14, P5‐15‐03<br />

Van Huffel, S<br />

P4‐06‐11<br />

van IJcken, WFJ<br />

P6‐04‐08<br />

van Kampen, RJW P5‐21‐04, P6‐07‐32<br />

van Laar, R P2‐10‐05, P6‐02‐08<br />

van Laarhoven, HW<br />

P1‐14‐07<br />

van Laarhoven, HWM PD07‐06<br />

Van Laere, S P3‐06‐01, P3‐10‐08<br />

Van Laere, SJ P2‐01‐08, P2‐01‐09,<br />

P3‐10‐04<br />

van Leeuwen‐Stok, E PD07‐06<br />

van Leeuwen‐Stok, EAE P1‐12‐05<br />

Van Limbergen, E P2‐10‐12, P4‐15‐01,<br />

P6‐05‐05, P6‐07‐04,<br />

P6‐07‐19, P6‐07‐29<br />

Van Loo, P<br />

P5‐05‐03<br />

van Ooijen, B<br />

P4‐11‐01<br />

Van Poznak, CH<br />

OT1‐1‐11<br />

van Puyvelde, K<br />

P6‐09‐07<br />

van Riel, JMGH<br />

P6‐07‐32<br />

van Roozendaal, L<br />

P3‐02‐08<br />

van Roozendaal, LM OT2‐1‐02,<br />

OT2‐1‐03<br />

van Roye, C<br />

P2‐11‐11<br />

van Slooten, H‐J<br />

PD02‐03<br />

van Spronsen, DJ<br />

P1‐14‐07<br />

van Tinteren, H<br />

P1‐12‐05<br />

van Warmerdam, LJ PD07‐06, P1‐14‐07<br />

Van Zee, KJ<br />

PD04‐05<br />

van’t Veer, L<br />

P2‐05‐01,<br />

P3‐04‐01, P3‐04‐02, P3‐05‐02,<br />

P3‐06‐11, P4‐13‐13, P5‐05‐01<br />

van’t Veer, LJ P2‐10‐42,P3‐02‐01,<br />

P4‐09‐02, P4‐09‐05<br />

Vanbeckevoort, D<br />

P6‐05‐05<br />

Vandenberg, E<br />

P1‐13‐01<br />

Vandenberg, T<br />

P1‐14‐13<br />

Vandergrift, JL<br />

P1‐13‐05<br />

Vanderhaegen, J P2‐13‐06, P5‐18‐19,<br />

P6‐05‐05, P6‐07‐29<br />

Vandermeer, L P2‐05‐13, P3‐13‐05<br />

Vanderschueren, D<br />

P2‐13‐06<br />

Vanderstichele, A P2‐10‐12, P6‐05‐05<br />

VandeWydeven, K<br />

P5‐13‐03<br />

vanDiest, PJ PD01‐02, PD01‐03<br />

Vanlemmens, L<br />

P2‐13‐10,<br />

P2‐13‐10, P3‐06‐20<br />

Vanni, G<br />

P4‐15‐04<br />

Vanzulli, SI<br />

P4‐06‐15<br />

Varadan, V PD05‐05, P4‐01‐02<br />

Vareslija, D<br />

P6‐04‐21<br />

Vargas, W<br />

P5‐07‐06<br />

Varma, S<br />

PD05‐09<br />

Varoqueaux, N<br />

P4‐02‐06<br />

Varricchio, C<br />

PD04‐07<br />

Vaske, CJ P2‐06‐05, P2‐06‐06<br />

Vasovics, S<br />

P5‐02‐02<br />

Vásquez, J<br />

P6‐07‐21<br />

Vassilakopoulou, M S5‐4<br />

Vaughan‐Briggs, C<br />

P5‐14‐03<br />

Vaysse, C<br />

P1‐01‐19<br />

Vaz‐Luis, I<br />

P5‐18‐03<br />

Vedell, P<br />

P1‐08‐02<br />

Veiseh, M<br />

P1‐05‐20<br />

Vejborg, I<br />

P3‐02‐03<br />

Velasco, V<br />

P3‐14‐03<br />

Veldman, T<br />

P2‐10‐03<br />

Velikova, G<br />

S3‐3, P4‐01‐14<br />

Venables, K S4‐1<br />

Venat Bouvet, L<br />

PD07‐04<br />

Vendel, Y<br />

P2‐13‐10<br />

Vente, JP<br />

P4‐11‐01<br />

Venuto, T<br />

P5‐03‐02<br />

Venzon, D<br />

P1‐09‐04<br />

Vergote, I<br />

P2‐10‐12,<br />

P6‐05‐05, P6‐07‐29<br />

Verhaeghe, J<br />

P2‐13‐06<br />

Verhoef, S<br />

P4‐11‐01<br />

Verhoeven, D S1‐4<br />

Verkooijen, HM P2‐11‐06, P4‐01‐17,<br />

P4‐14‐08, P5‐01‐11,<br />

P5‐01‐14, P5‐15‐03<br />

Verma, R<br />

P1‐13‐08<br />

Verma, S P2‐11‐10, P2‐12‐03, P2‐12‐07,<br />

P3‐12‐04, P5‐13‐01, P6‐07‐33<br />

Vermeulen, PB P2‐01‐08, P2‐01‐09,<br />

P3‐10‐04, P3‐10‐08<br />

Vermorken, JB<br />

PD01‐03<br />

Verriele, V<br />

P1‐14‐21<br />

Verril, M<br />

P1‐13‐03<br />

Verrill, M S3‐3<br />

Vetter, M<br />

P1‐13‐13, P2‐10‐38<br />

Veyret, C<br />

P1‐13‐13, P1‐15‐08<br />

Veys, I<br />

S4‐4, P3‐05‐03<br />

Viñas, G P4‐09‐10, P4‐09‐11, P6‐11‐02<br />

Viale, G<br />

S1‐1, S4‐4, S4‐6,<br />

PD04‐07, PD10‐03, P3‐05‐02,<br />

P3‐05‐03, P6‐07‐14<br />

Vicente Conesa, AM P3‐06‐15<br />

Vicente García, V<br />

P3‐06‐15<br />

Vicini, F<br />

P1‐15‐09<br />

Vicini, FA P4‐16‐03, P4‐16‐09<br />

Vickery, T P2‐10‐01, P6‐07‐10<br />

Victor, C<br />

P2‐12‐07<br />

Vidal, CM P6‐11‐07, P6‐11‐09<br />

Vidal, L<br />

P6‐11‐06<br />

Vidal, M<br />

P6‐13‐03<br />

Vidaurre, T<br />

P6‐07‐21<br />

Vieites, B<br />

P1‐01‐29<br />

Viens, P<br />

P1‐13‐04, P3‐12‐11,<br />

P5‐03‐01, OT2‐3‐05<br />

Vierkant, RA P4‐12‐03, P5‐01‐08<br />

Vigil, CE<br />

P6‐07‐20<br />

Vijayakrishn, GK<br />

P6‐04‐03<br />

Vila‐Caballer, M<br />

P6‐06‐05<br />

Vilardell, F<br />

P1‐01‐29<br />

Villanueva, C<br />

P5‐18‐22<br />

Villanueva, O<br />

P4‐06‐19<br />

Villarin, E<br />

P2‐01‐10<br />

Villarreal‐Garza, C<br />

PD08‐06<br />

Vinayak, S<br />

PD09‐04<br />

Vincent, A PD01‐02, PD01‐03<br />

Vincent, D S6‐2, P1‐07‐08, P3‐05‐03<br />

Vincent‐Salomon, A PD05‐08,<br />

P1‐01‐19, P2‐01‐04, P5‐01‐04,<br />

P5‐02‐03, P5‐21‐03, P6‐04‐14<br />

Vingiani, A<br />

PD03‐01<br />

Violante, A<br />

P1‐01‐25<br />

Viqueira, A<br />

P6‐11‐02<br />

Virador, VM<br />

P2‐04‐08<br />

Virani, S<br />

PD04‐02<br />

Virnig, BA<br />

P4‐01‐15<br />

Viscusi, RK<br />

P3‐10‐07<br />

Visram, H<br />

P3‐11‐03<br />

Visscher, DW<br />

P4‐12‐03,<br />

P5‐01‐08, P6‐06‐01<br />

Visvanathan, K S3‐5, P2‐02‐01<br />

Vo, H‐T<br />

PD02‐04<br />

Voduc, D P4‐16‐02, P4‐16‐04, P6‐07‐10<br />

Vogel, C<br />

OT2‐3‐11<br />

Vogel, CL<br />

P1‐12‐03<br />

Vogel, VG<br />

P4‐13‐12<br />

Vogel, WV<br />

P4‐02‐01<br />

Vojnovic, B<br />

P2‐10‐29<br />

Vollan, HKM<br />

P2‐10‐20<br />

Volm, M<br />

P5‐20‐05<br />

Volta, V<br />

P5‐03‐02<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

606s<br />

<strong>Cancer</strong> Research


December 4-8, 2012<br />

Author Index<br />

von der Assen, A<br />

P5‐21‐02<br />

von Euler‐Chelpin, M P3‐02‐03<br />

Von Euw, E<br />

P4‐08‐01<br />

von Minckwitz, G S1‐7, S2‐2, S3‐1,<br />

S4‐5, PD06‐01, P1‐13‐13, P1‐14‐01,<br />

P3‐06‐05, P6‐07‐05, OT1‐1‐13,<br />

OT2‐2‐05, OT3‐3‐11<br />

von Schoultz, E<br />

P6‐14‐01<br />

vonMincwitz, G S1‐11<br />

Voogd, AC P5‐21‐04, P6‐07‐32<br />

Vos, S<br />

P5‐01‐06<br />

Voss, A<br />

P3‐06‐21<br />

Voytko, NL S1‐6<br />

Vrancken Peeters, M‐JTFD P4‐02‐01<br />

Vrbanec, D<br />

P5‐02‐02<br />

Vreeland, TJ P5‐16‐02, P5‐16‐05<br />

Vriens, B<br />

PD07‐06<br />

Vriens, BE<br />

P1‐14‐07<br />

Vrigneaud, J‐M<br />

P4‐02‐06<br />

Vrouenraets, BC<br />

P4‐11‐01<br />

Wa, A<br />

P4‐11‐02<br />

Wachsmann, G S1‐7<br />

Wactawski‐Wende, J PD03‐09<br />

Wada, N<br />

P3‐02‐06<br />

Wada, R<br />

P5‐18‐11<br />

Wada, Y<br />

P3‐13‐04<br />

Wagle, N<br />

P5‐18‐03<br />

Wagner, J<br />

P1‐07‐06<br />

Wagner, K‐U S1‐8<br />

Wahdan‐Alaswad, RS P5‐10‐04<br />

Wahdan‐Alsawad, RS P5‐10‐05<br />

Waheed, S P1‐01‐10, P1‐01‐11<br />

Wai, ES<br />

PD04‐02<br />

Waisman, J<br />

P5‐16‐04<br />

Wakita, K<br />

P6‐07‐39<br />

Walker, K<br />

P3‐08‐04<br />

Walker, M<br />

P5‐20‐08<br />

Walker, MS<br />

P1‐15‐12<br />

Wallace, AM<br />

P2‐06‐01<br />

Wallden, B<br />

P3‐06‐03<br />

Wallecha, A<br />

P4‐04‐05<br />

Walley, B P1‐13‐01, OT2‐2‐01<br />

Wallin, J<br />

P5‐19‐02<br />

Wallin, JJ<br />

P4‐07‐01<br />

Wallon, UM<br />

P4‐09‐12<br />

Wallwiener, D<br />

P6‐09‐08<br />

Wals, J<br />

P1‐14‐07<br />

Walsh, CA<br />

P6‐04‐01<br />

Walsh, FJ<br />

PD02‐05<br />

Walsh, T<br />

P6‐07‐10<br />

Walter, HS<br />

P4‐14‐14<br />

Walters, EA<br />

P4‐06‐18<br />

Walz, T<br />

P1‐05‐07<br />

Wan, Y<br />

P6‐09‐06<br />

Wanders, J<br />

S6‐6, P6‐11‐14<br />

Wang, A<br />

PD03‐03, PD10‐03<br />

Wang, B P5‐18‐11, P5‐18‐24, P6‐11‐01<br />

Wang, D<br />

P5‐03‐04, P5‐03‐06<br />

Wang, D‐Y<br />

P2‐10‐34<br />

Wang, F<br />

P4‐16‐07<br />

Wang, FQ<br />

P4‐16‐12<br />

Wang, H<br />

PD02‐04<br />

Wang, J<br />

P2‐10‐30, P3‐04‐08,<br />

P4‐05‐01, P6‐11‐01<br />

Wang, K<br />

S3‐6, P4‐03‐12<br />

Wang, L<br />

OT3‐4‐04<br />

Wang, N<br />

PD07‐08<br />

Wang, Q<br />

P2‐09‐05<br />

Wang, S<br />

P2‐05‐05, P4‐06‐18<br />

Wang, T<br />

S5‐8, P4‐08‐07<br />

Wang, X PD09‐08, P5‐10‐10, P6‐04‐25<br />

Wang, X‐j P2‐05‐05, P4‐09‐09<br />

Wang, Y P2‐11‐16, P3‐06‐12, P4‐11‐04<br />

Wang, YC S5‐8<br />

Wang, Y‐L<br />

P3‐12‐10<br />

Wang, Y‐S<br />

P1‐14‐17<br />

Wang, Z<br />

PD02‐04, P4‐03‐06,<br />

P5‐10‐09, P6‐07‐45<br />

Wani, NA<br />

P1‐05‐17<br />

Wantanabe, A<br />

P1‐05‐04<br />

Wanyo, CM<br />

P3‐06‐27<br />

Wapnir, I S3‐2<br />

Wapnir, IL<br />

P6‐07‐06<br />

Ward, J<br />

P4‐17‐03<br />

Ward, M<br />

P1‐15‐07<br />

Ward, MM<br />

P6‐11‐04<br />

Ward, P<br />

P5‐20‐10<br />

Wardley, A<br />

S3‐3, P1‐13‐03<br />

Wardley, AM P2‐14‐06, OT1‐1‐03<br />

Warm, M<br />

PD07‐02<br />

Warnecke, R<br />

P5‐13‐03<br />

Warner, E<br />

P2‐12‐03,<br />

P3‐02‐10, P6‐08‐04<br />

Warr, R<br />

P4‐17‐03<br />

Warren, LE<br />

P3‐10‐06<br />

Warren, M<br />

P2‐12‐15<br />

Warren, R<br />

P1‐09‐04<br />

Warren, RM<br />

P4‐13‐09<br />

Warren Peled, A P4‐14‐04, P4‐16‐07<br />

Warrick, A<br />

P2‐08‐03<br />

Warwick, J<br />

P3‐08‐01<br />

Wasser, MNJM<br />

PD07‐06<br />

Watanabe, G<br />

P2‐10‐43<br />

Watanabe, J<br />

P1‐12‐01,<br />

P5‐18‐16, P6‐07‐16<br />

Watanabe, K<br />

P6‐07‐39<br />

Watanabe, M<br />

P2‐10‐43,<br />

P2‐12‐13, P6‐05‐14<br />

Watanabe, T P1‐13‐10, P2‐13‐04,<br />

P2‐13‐07, P4‐14‐07<br />

Waters, PS P1‐01‐15, P2‐05‐09<br />

Waterworth, A<br />

P3‐01‐06<br />

Watson, AP<br />

P1‐05‐16<br />

Watson, M PD07‐01, P2‐04‐02<br />

Wauters, CAP<br />

P5‐01‐13<br />

Waxman, J<br />

P2‐02‐03<br />

Waynick, C<br />

P2‐14‐05<br />

Waynick, S<br />

P2‐14‐05<br />

Wazer, D<br />

P4‐16‐03<br />

Wazir, U<br />

P1‐04‐07, P1‐04‐08,<br />

P1‐04‐09, P6‐05‐11<br />

Weaver, J<br />

P3‐05‐01<br />

Weaver, R<br />

P6‐10‐01<br />

Webb, C<br />

P1‐14‐14<br />

Webb, P<br />

P4‐08‐09, P5‐07‐02<br />

Webber, V<br />

P6‐04‐10<br />

Weber, B<br />

P3‐06‐20<br />

Weber, D<br />

OT2‐3‐08<br />

Weber, HA S5‐2<br />

Weber, JJ<br />

P1‐01‐17<br />

Weber, KE<br />

S4‐3, P2‐10‐11<br />

Webster, MA<br />

P1‐07‐10<br />

Webster, S<br />

P1‐07‐05<br />

Weeks, J<br />

P1‐01‐13<br />

Weeks, JC<br />

S3‐5, P1‐13‐05<br />

Wefel, JS<br />

OT1‐1‐11<br />

Wei, W P4‐02‐08, P5‐03‐08, P5‐10‐10<br />

Wei Shi, W<br />

P2‐10‐33<br />

Weide, R<br />

P2‐11‐11<br />

Weideman, PC<br />

P4‐13‐10<br />

Weidler, J<br />

P2‐05‐06,<br />

P2‐10‐16, P2‐10‐31<br />

Weidner, SM<br />

P1‐15‐12<br />

Weigelt, B<br />

PD05‐08<br />

Weigert, JM P4‐02‐09, P4‐02‐10<br />

Weigert, R<br />

P1‐05‐23<br />

Weiler‐Mithoff, E P4‐14‐13, P4‐17‐03<br />

Weinstein, P<br />

P5‐20‐08<br />

Weinstein, S<br />

P4‐01‐08<br />

Weir, L<br />

P4‐16‐01<br />

Weiss, E<br />

P2‐10‐25<br />

Weiss, HL<br />

P2‐14‐05<br />

Weiss, JE<br />

P4‐01‐15<br />

Weiss, L<br />

P4‐13‐13<br />

Weiss, MS<br />

P6‐04‐24<br />

Weisser, B<br />

OT3‐2‐01<br />

Weitsman, G<br />

P2‐10‐29<br />

Weitzel, JN<br />

PD08‐06<br />

Welch, RS<br />

P2‐14‐06<br />

Wells, C<br />

P4‐01‐13<br />

Wells, MN<br />

P2‐01‐14<br />

Welm, AL<br />

P6‐04‐20<br />

Welnicka‐Jaskiewicz, M P5‐04‐01<br />

Weltens, C P2‐10‐12, P6‐05‐05,<br />

P6‐07‐04, P6‐07‐19, P6‐07‐29<br />

Weltz, B<br />

P2‐10‐24<br />

Weltzien, E<br />

P3‐07‐05<br />

Wemhoff, G<br />

P4‐06‐03<br />

Wen, YH<br />

P2‐08‐01<br />

Weng, Z<br />

PD05‐09<br />

Wenger, N<br />

P4‐13‐13<br />

Wennmalm, K<br />

PD06‐08<br />

Wenz, F S4‐2<br />

Werner, L<br />

P1‐07‐07<br />

Werner Hartman, L<br />

P2‐10‐37<br />

Wesolowski, R<br />

P2‐11‐07<br />

Wesseling, J<br />

PD01‐03,<br />

P2‐10‐42, P3‐05‐01<br />

Wessels, H<br />

P5‐01‐11<br />

West, DW P3‐11‐02, P4‐11‐02<br />

West, RB<br />

PD05‐09<br />

Westbrook, TF<br />

PD01‐01<br />

Westenend, PJ<br />

P6‐07‐05<br />

Wetzel, L<br />

P4‐07‐05<br />

Wevers, MR<br />

P4‐11‐01<br />

Wey, A<br />

P1‐14‐11<br />

Wey, D<br />

P2‐11‐11<br />

Wheatley, D<br />

PD07‐09<br />

Wheeler, A<br />

P4‐11‐04<br />

Whelan, T<br />

P1‐13‐01<br />

Wheler, J<br />

P5‐20‐09<br />

White, A<br />

P3‐02‐04<br />

White, CB<br />

P6‐09‐11<br />

White, J<br />

P4‐06‐11<br />

Whitford, DL<br />

P6‐08‐07<br />

Whitman, B<br />

P6‐08‐09<br />

Whitworth, P PD07‐01, P1‐15‐09,<br />

P3‐06‐28, OT3‐4‐03<br />

Wicha, M<br />

OT2‐3‐01<br />

Wicinski, J<br />

P5‐03‐01<br />

Wickerham, DL S1‐10<br />

Wickerham, L S1‐11<br />

Wickham, T<br />

P1‐07‐03,<br />

P4‐02‐05, P5‐18‐09<br />

Wiegmann, DA<br />

P2‐11‐15<br />

Wienert, S<br />

PD06‐01<br />

Wiest, W<br />

P1‐13‐13<br />

Wiktor, AE<br />

PD02‐05<br />

Wildiers, H P2‐10‐12, P3‐13‐01,<br />

P6‐05‐05, P6‐07‐04,<br />

P6‐07‐05, P6‐07‐19, P6‐07‐29,<br />

P6‐09‐07, OT2‐2‐02<br />

Wilke, L<br />

P3‐02‐11<br />

Wilke, LG S2‐1<br />

Wilkens, LR<br />

P4‐12‐02<br />

Wilkerson, MD S6‐1<br />

Wilking, N<br />

P6‐14‐01<br />

Wilkins, M<br />

P4‐10‐02<br />

Wilkinson, JB<br />

P4‐16‐03<br />

Wilks, S<br />

P5‐20‐04<br />

Willems, SM<br />

P5‐01‐14<br />

Willey, J<br />

P3‐10‐05<br />

Willey, JS P5‐20‐13, OT2‐3‐10<br />

Willey, SC<br />

P4‐14‐01<br />

Williams, G<br />

PD04‐08<br />

Williams, K<br />

P3‐14‐04<br />

Williams, KE<br />

PD04‐06<br />

Williams, NR S4‐2<br />

Willianson, J<br />

P3‐08‐04<br />

Wilson, BC<br />

P4‐03‐04<br />

Wilson, C<br />

S6‐4, P2‐02‐04<br />

Wilson, G<br />

P2‐14‐06<br />

Wilson, J<br />

P1‐01‐13<br />

Wilson, JL<br />

P1‐13‐05<br />

Wilson, K<br />

P4‐13‐04<br />

Wilson, M<br />

P3‐01‐04<br />

Wilson, S<br />

P4‐17‐03<br />

Wilson‐Edell, KA<br />

P2‐05‐04<br />

Winchester, D<br />

PD04‐04<br />

Winchester, L<br />

P4‐01‐01<br />

Winczura, P P3‐12‐09, P3‐13‐03<br />

Winer, E S1‐1, S5‐5, P3‐05‐03,<br />

P5‐18‐03, P6‐08‐03, P6‐10‐05,<br />

OT1‐1‐07, OT2‐3‐06<br />

Winer, EP PD09‐03, PD10‐04,<br />

P2‐10‐01, P2‐16‐04,<br />

P6‐08‐05, OT1‐1‐11<br />

Winslow, J P2‐10‐16, P2‐10‐31<br />

Winter, A<br />

P4‐14‐13<br />

Winter, M<br />

P1‐15‐05<br />

Winter, MC<br />

P2‐02‐04<br />

Winter, W<br />

P2‐11‐16<br />

Winters, Z<br />

P4‐17‐03<br />

Wirtz, R<br />

P3‐06‐19<br />

Wirtz, RM PD10‐06, P3‐06‐08<br />

Wischnik, A P5‐14‐06, P5‐15‐04<br />

Wisell, J<br />

P6‐05‐10<br />

Wishart, GC<br />

P1‐01‐01<br />

Wisinski, KB<br />

P2‐11‐15,<br />

P3‐02‐11, P6‐12‐02<br />

Wisner, DJ<br />

P4‐01‐10<br />

Wist, EA<br />

P3‐01‐01<br />

Witkamp, AJ P4‐01‐17, P4‐11‐01,<br />

P5‐01‐11, P5‐15‐03<br />

Witteveen, PO<br />

P6‐07‐05<br />

Wittner, BS<br />

P2‐01‐14<br />

Wogu, AF P1‐15‐04, P3‐06‐07<br />

Wojciechowski, BS<br />

P4‐09‐12<br />

Wojtukiewicz, M<br />

OT1‐1‐16<br />

Wolchok, J<br />

OT3‐1‐01<br />

Wolf, D P3‐04‐01, P3‐04‐02, P4‐09‐05<br />

Wolf, DM P2‐05‐01, P4‐09‐02<br />

Wolfer, A<br />

P6‐04‐05<br />

Wolff, AC S3‐5, P1‐13‐05, P2‐02‐01,<br />

OT1‐1‐11<br />

Wolff, RA<br />

P5‐15‐02<br />

Wolmark, N S1‐10, S1‐11, S3‐2, S5‐5,<br />

P6‐07‐06, OT1‐1‐12, OT1‐2‐01<br />

Won, HH<br />

PD05‐02<br />

Wong, C<br />

P2‐14‐08<br />

Wong, CHN P3‐11‐02, P4‐11‐02<br />

Wong, CLP<br />

P4‐11‐02<br />

Wong, FY<br />

P4‐16‐12<br />

Wong, GYC<br />

P1‐10‐02<br />

Wong, H<br />

P4‐13‐04<br />

Wong, J P1‐01‐17, P3‐08‐09, P5‐01‐09<br />

Wong, KL<br />

P2‐01‐10<br />

Wong, S<br />

P4‐02‐08<br />

Wong, ST<br />

P3‐06‐14<br />

Wong, Y‐N P1‐01‐13, P1‐13‐05<br />

Wong-Brown, MW<br />

P4‐10‐02<br />

Woo, SY<br />

P2‐05‐20<br />

Woodman, N<br />

P2‐10‐29<br />

Woods, R<br />

PD04‐02, P4‐16‐01<br />

Woods, RR<br />

P3‐10‐07<br />

Woodward, W<br />

P3‐10‐10,<br />

P5‐03‐09, P5‐10‐02<br />

Woodward, WA PD03‐06, PD03‐08,<br />

P2‐03‐01, P3‐10‐01, P3‐10‐05,<br />

P5‐03‐05, P5‐03‐10, P5‐04‐06<br />

Worsham, MJ<br />

P2‐09‐04<br />

Wouters, C<br />

P3‐13‐01<br />

Wouters, K<br />

P3‐13‐01<br />

Wright, B<br />

P3‐06‐21<br />

Wright, F<br />

P6‐08‐04<br />

Wright, MC P4‐06‐03, P6‐10‐04<br />

Wright, WE<br />

P4‐06‐06<br />

Wroblewski, S<br />

OT1‐1‐04<br />

Wu, A P1‐09‐06, P5‐03‐04, P5‐03‐06<br />

Wu, C<br />

PD03‐09<br />

Wu, C‐C<br />

P4‐04‐07, P6‐05‐12<br />

Wu, E<br />

P5‐19‐02<br />

Wu, EQ<br />

P5‐15‐05, P5‐18‐12<br />

Wu, H<br />

PD05‐01<br />

Wu, J P3‐01‐05, P4‐05‐01, P6‐04‐26<br />

Wu, L<br />

P2‐09‐05<br />

Wu, M<br />

P5‐10‐10<br />

Wu, Q P3‐04‐01, P3‐04‐02, P5‐04‐06<br />

Wulfkuhle, JD P3‐04‐01, P3‐04‐02<br />

Wuyts, H<br />

P2‐01‐08<br />

Wymenga, MANM<br />

P1‐12‐05<br />

Wysocki, PJ<br />

P2‐10‐31<br />

Xerri, l<br />

P5‐03‐01<br />

Xia, G S1‐8<br />

Xia, W<br />

P4‐08‐03, P4‐08‐07<br />

Xiang, D<br />

P1‐07‐04<br />

Xiao, X<br />

P5‐10‐10<br />

Xiao, Z<br />

P4‐07‐05<br />

Xiaoyi, L<br />

P2‐10‐26<br />

Xie, H<br />

PD10‐09<br />

Xie, J<br />

P5‐15‐05<br />

Xie, X<br />

P5‐10‐10, P5‐10‐10<br />

Xie, Z<br />

P5‐10‐10<br />

Xingang, W<br />

P2‐10‐26<br />

Xu, B<br />

OT1‐1‐16<br />

Xu, J<br />

P4‐08‐10<br />

www.aacrjournals.org 607s<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012


CTRC-AACR <strong>San</strong> <strong>Antonio</strong> <strong>Breast</strong> <strong>Cancer</strong> <strong>Symposium</strong><br />

Xu, N<br />

P5‐03‐14<br />

Xu, Q<br />

P6‐11‐07, P6‐11‐09<br />

Xu, R<br />

P6‐13‐02<br />

Xu, W PD03‐06, P5‐03‐05, P5‐03‐10<br />

Xu, X<br />

P2‐01‐10<br />

Xu, X‐H<br />

P2‐05‐05<br />

Xu, X‐L<br />

P6‐07‐42<br />

Xu, XX<br />

PD08‐02<br />

Xu, Y<br />

P6‐04‐15<br />

Xu, Z P1‐03‐03, P4‐03‐12, P5‐01‐16<br />

Xue, L<br />

P2‐05‐19<br />

Xynos, Y<br />

P3‐02‐04<br />

Yadav, P<br />

P4‐06‐02<br />

Yagata, H P1‐01‐12, P5‐15‐06,<br />

P6‐07‐23, P6‐07‐38<br />

Yager, S<br />

OT3‐4‐04<br />

Yamada, J<br />

PD06‐02<br />

Yamada, Y<br />

P4‐09‐08<br />

Yamagami, K PD07‐05, P3‐13‐04<br />

Yamaguchi, T P2‐13‐04, P2‐13‐07<br />

Yamaguchi, Y<br />

P6‐04‐17<br />

Yamaki, E<br />

P4‐03‐05<br />

Yamamoto, D P1‐12‐01, P6‐07‐16<br />

Yamamoto, N<br />

P3‐02‐06<br />

Yamamoto, S<br />

P2‐11‐02<br />

Yamamoto, Y P1‐14‐02, P1‐14‐08,<br />

P2‐14‐03, P3‐06‐18,<br />

P6‐05‐06, P6‐07‐16<br />

Yamamoto‐Ibusuki, M P2‐14‐03<br />

Yamamura, J<br />

P1‐14‐08<br />

Yamamura, N<br />

P4‐03‐07<br />

Yamanaka, A<br />

P1‐01‐20<br />

Yamanaka, T<br />

P1‐01‐20<br />

Yamashige, S<br />

P5‐15‐06<br />

Yamashiro, H<br />

P3‐13‐04<br />

Yamashita, D<br />

P4‐03‐05<br />

Yamashita, H P1‐14‐02, P3‐06‐18,<br />

P3‐12‐03, P4‐12‐06<br />

Yamashita, Y<br />

P4‐03‐05<br />

Yamauch, A<br />

P3‐13‐04<br />

Yamauchi, H P1‐01‐12, P5‐15‐06,<br />

P6‐07‐23, P6‐07‐38<br />

Yan, FJ<br />

P1‐07‐17<br />

Yan, Y<br />

P3‐04‐01, P3‐04‐02<br />

Yanac, AF S1‐8<br />

Yang, C<br />

P1‐13‐07<br />

Yang, G<br />

P2‐10‐39<br />

Yang, H P5‐01‐16, P5‐03‐14, P5‐15‐05<br />

Yang, J P1‐05‐18, P2‐04‐07, P6‐06‐05<br />

Yang, JH<br />

P2‐05‐20<br />

Yang, J‐H<br />

P2‐10‐41<br />

Yang, L<br />

P5‐10‐10<br />

Yang, N S1‐8<br />

Yang, P<br />

PD03‐08<br />

Yang, S‐H<br />

P4‐03‐02<br />

Yang, S‐Y<br />

P3‐06‐02<br />

Yang, W<br />

P3‐06‐14<br />

Yang, XR<br />

P3‐08‐02<br />

Yang, Y<br />

P2‐05‐19, P2‐07‐01<br />

Yang, Z<br />

P3‐12‐12<br />

Yankeelov, TE<br />

P4‐01‐03<br />

Yano, H<br />

P1‐01‐27<br />

Yano, S<br />

P4‐07‐04<br />

Yao, K<br />

P3‐05‐01<br />

Yap, Y‐S<br />

P3‐08‐09<br />

Yardley, DA P1‐14‐14, P2‐05‐06,<br />

P3‐12‐04, P5‐17‐05, P5‐20‐10,<br />

P6‐10‐01, P6‐11‐14, OT3‐3‐08<br />

Yarnold, J<br />

P1‐13‐03<br />

Yarnold, JR<br />

S4‐1, P3‐06‐09<br />

Yasmeen, S<br />

P4‐13‐13<br />

Yau, C P2‐05‐01, P2‐10‐06, P3‐04‐01,<br />

P3‐04‐02, P4‐09‐02<br />

Ye, F<br />

P2‐05‐19<br />

Ye, L<br />

P1‐04‐03<br />

Ye, W‐w<br />

P4‐09‐09<br />

Ye, Z<br />

P4‐06‐03, P6‐10‐04<br />

Yee, D<br />

P1‐14‐11<br />

Yeh, D‐C<br />

OT1‐1‐16<br />

Yeh, E<br />

P5‐02‐01<br />

Yeh, ED P3‐10‐06, P4‐01‐09, P4‐06‐01<br />

Yeh, I‐T<br />

P5‐03‐04, P5‐03‐06<br />

Yeh, L‐C<br />

P6‐05‐15<br />

Yelensky, R<br />

S3‐6, PD02‐07<br />

Yelle, L S6‐6<br />

Yellin, MJ<br />

P6‐10‐01<br />

Yerushalmi, R<br />

P5‐20‐06<br />

Yeung, TY<br />

PD01‐08<br />

Yevtushenko, M<br />

P2‐05‐04<br />

Yi, J<br />

OT3‐4‐04<br />

Yi, J‐H<br />

P5‐18‐24<br />

Yi, M<br />

P1‐07‐09<br />

Yin, W<br />

P6‐07‐35<br />

Yip, AYS<br />

P2‐05‐16<br />

Yokota, I<br />

P2‐13‐04, P2‐13‐07<br />

Yong, W‐S<br />

P3‐08‐09<br />

Yoon, K<br />

P1‐05‐18<br />

Yoshibayashi, H<br />

P3‐13‐04<br />

Yoshida, A P1‐01‐20, P5‐15‐06,<br />

P6‐07‐23, P6‐07‐38<br />

Yoshida, K<br />

P5‐04‐02<br />

Yoshida, M<br />

P4‐03‐08<br />

Yoshida, N<br />

P1‐14‐02<br />

Yoshida, S<br />

P6‐07‐30<br />

Yoshida, T<br />

P6‐04‐13<br />

Yoshidome, K<br />

P2‐10‐18<br />

Yoshimoto, K<br />

P4‐03‐05<br />

Yoshimoto, N<br />

P4‐12‐06<br />

Yoshimura, K<br />

P5‐01‐02<br />

Yoshiyama, T<br />

P3‐06‐26<br />

Yoshizawa, C<br />

P5‐15‐06<br />

You, M<br />

P1‐08‐02<br />

You, Z<br />

P5‐17‐07<br />

Youn, HJ<br />

P1‐10‐01, P5‐07‐04<br />

Young, L<br />

P6‐04‐04, P6‐04‐21<br />

Young, LS<br />

P6‐04‐01<br />

Young, R<br />

OT3‐3‐07<br />

Younus, J<br />

P2‐13‐08<br />

Youssoufian, H<br />

P6‐11‐10<br />

Yovine, A<br />

OT2‐2‐03<br />

Yu, B‐L<br />

P4‐16‐05<br />

Yu, J<br />

P2‐08‐02<br />

Yu, J‐C<br />

P4‐04‐07<br />

Yu, JH<br />

P3‐01‐02<br />

Yu, K‐D P1‐13‐07, P2‐06‐07, P3‐06‐14<br />

Yu, M<br />

S5‐7, P2‐01‐14<br />

Yu, S<br />

P1‐09‐06<br />

Yu, T<br />

P5‐15‐06<br />

Yu, X‐y<br />

P4‐09‐09<br />

Yuan, J<br />

OT3‐1‐01<br />

Yue, B<br />

P2‐10‐40<br />

Yueh, N<br />

P4‐07‐03<br />

Yuki, S<br />

P2‐05‐08<br />

Yun, C<br />

P4‐03‐11<br />

Yvonnet, V<br />

P3‐06‐24<br />

Zabaglo, LA S4‐6<br />

Zaborek, P<br />

P5‐02‐02<br />

Zacharchuk, C<br />

OT1‐1‐05<br />

Zachariae, B<br />

P6‐07‐08<br />

Zacksenhaus, E<br />

P6‐02‐03<br />

Zaczek, AJ<br />

P5‐04‐01<br />

Zafra Poves, M<br />

P3‐06‐15<br />

Zahm, D‐M<br />

P2‐10‐25<br />

Zaman, K P6‐04‐05, OT2‐2‐02<br />

Zamani, A<br />

P2‐12‐10<br />

Zambetti, M<br />

P2‐12‐08<br />

Zamora, E<br />

P6‐13‐03<br />

Zamora, P P1‐07‐18, P2‐01‐06<br />

Zannetti, A<br />

OT3‐3‐04<br />

Zanoni, V<br />

PD07‐03<br />

Zaravinos, J<br />

P1‐12‐03<br />

Zaun, S<br />

P6‐09‐08<br />

Zebisch, M<br />

P2‐01‐11<br />

Zehnbauer, B<br />

P6‐07‐10<br />

Zeichner, SB<br />

P1‐12‐03<br />

Zelek, LH<br />

PD08‐03<br />

Zelenika, D<br />

P5‐05‐03<br />

Zellinger, J<br />

P2‐13‐01<br />

Zemba‐Palko, V<br />

P4‐09‐12<br />

Zeng, W<br />

P4‐05‐01<br />

Zhan, M<br />

P3‐06‐14, P4‐02‐08<br />

Zhang, A‐Q<br />

P2‐09‐05<br />

Zhang, B<br />

P6‐10‐02<br />

Zhang, D<br />

P5‐03‐09<br />

Zhang, DY<br />

P2‐05‐19<br />

Zhang, F<br />

P5‐10‐03<br />

Zhang, H P1‐04‐04, P1‐05‐08,<br />

P4‐06‐05, P6‐04‐07<br />

Zhang, J<br />

P6‐07‐43, P6‐11‐01<br />

Zhang, L<br />

S1‐11, P2‐12‐03<br />

Zhang, M S6‐3, P4‐02‐08, P5‐03‐08<br />

Zhang, Q P3‐12‐12, OT1‐1‐16<br />

Zhang, T<br />

P3‐03‐01<br />

Zhang, X<br />

P6‐10‐04<br />

Zhang, Y S1‐9, P1‐03‐03, P1‐07‐13,<br />

P2‐10‐15, P4‐06‐18, OT2‐2‐03<br />

Zhang, Y‐J<br />

P6‐07‐43<br />

Zhang, Y‐M<br />

P4‐09‐09<br />

Zhang, Y‐P<br />

P6‐07‐43<br />

Zhang, Z<br />

P6‐07‐33, P6‐09‐06<br />

Zhang, Z‐P<br />

P1‐14‐17<br />

Zhao, H<br />

P2‐05‐12, P2‐05‐13<br />

Zhao, J<br />

P4‐08‐10<br />

Zhao, L<br />

P2‐04‐07<br />

Zhao, S P4‐08‐03, P4‐08‐07, P6‐01‐03<br />

Zhe, J<br />

P1‐04‐05<br />

Zheng, C<br />

P1‐07‐12<br />

Zheng, CX<br />

P3‐08‐06<br />

Zheng, J<br />

P4‐02‐05<br />

Zheng, P<br />

OT1‐2‐01<br />

Zheng, Y<br />

P3‐09‐05<br />

Zheng, Y‐b<br />

P4‐09‐09<br />

Zhidong, L<br />

P2‐10‐26<br />

Zhong, S<br />

P4‐08‐10<br />

Zhou, D<br />

P5‐16‐01<br />

Zhou, J<br />

P3‐01‐04, P4‐06‐08<br />

Zhou, JZ<br />

P3‐13‐06<br />

Zhou, M<br />

P6‐07‐43<br />

Zhou, Rj<br />

P6‐07‐36<br />

Zhu, L<br />

P1‐01‐09<br />

Zhu, R<br />

P3‐06‐14<br />

Zhu, S<br />

PD05‐09<br />

Zhu, X<br />

P3‐04‐11<br />

Zhu, Y<br />

P4‐02‐08<br />

Zhuang, X<br />

P6‐04‐28<br />

Zickl, L<br />

OT2‐2‐01<br />

Zielinski, C<br />

P5‐17‐03<br />

Zignani, J<br />

P3‐01‐03<br />

Zijlstra, A<br />

P3‐05‐09<br />

Zimon, D<br />

P5‐03‐13<br />

Zingelewicz, S<br />

P3‐05‐05<br />

Ziras, N<br />

P1‐13‐09<br />

Zlobin, A<br />

P2‐05‐15, P4‐08‐06<br />

Zoeller, JJ<br />

P4‐08‐05<br />

Zoghi, B<br />

P5‐10‐14, P5‐10‐16<br />

Zohar, S<br />

P4‐14‐12<br />

Zok, J<br />

P3‐12‐09, P3‐13‐03<br />

Zong, W‐X<br />

P5‐04‐09<br />

Zrada, S<br />

P2‐05‐06<br />

Zubritsky, LM<br />

P3‐06‐33<br />

Zujewski, JA S1‐11, S5‐5<br />

Zvelebil, M PD05‐08, P3‐08‐04<br />

Zwelling, L<br />

P5‐15‐02<br />

Zwenger, A<br />

P3‐07‐13<br />

<strong>Cancer</strong> Res; 72(24 Suppl.) December 15, 2012<br />

608s<br />

<strong>Cancer</strong> Research

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!