23.03.2016 Views

Best of AACR Journals

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

THE BEST OF<br />

THE <strong>AACR</strong> JOURNALS<br />

Learn more at<br />

<strong>AACR</strong>journals.org


Personalis®<br />

ACE Services for Cancer Research & Clinical Trials<br />

ACE Extended Cancer Panel for DNA and RNA<br />

The Most Complete and Accurate Tumor Pr<strong>of</strong>iling<br />

Designed for both cancer research and clinical trials<br />

The Accuracy and Content Enhanced (ACE) Extended Cancer Panel has been designed<br />

to support both cancer research and clinical trials. It provides more coverage <strong>of</strong> gene<br />

pathways and functions known to be involved in cancer biology than any other panel<br />

commercially available. Up until now discovery researchers have had to make a choice<br />

between the breadth <strong>of</strong> sequencing used to find novel variants in cancer-related genes,<br />

and the depth <strong>of</strong> sequencing needed to identify variants present at low allelic fraction.<br />

The Personalis ACE Extended Cancer Panel is the first service to combine the breadth<br />

<strong>of</strong> coverage for novel variant discovery across key cancer pathway genes, with the<br />

sequencing depth needed to detect variants present at low allelic fraction.<br />

Comprehensive coverage <strong>of</strong> key cancer pathway genes<br />

>500x mean coverage over the extended footprint (DNA)<br />

enhanced<br />

coverage <strong>of</strong><br />

over 1,300+<br />

cancer genes<br />

and<br />

over 200+<br />

miRNA genes<br />

Sensitivity for both known and novel fusion events (RNA)<br />

The most advanced and complete informatics analysis<br />

www.personalis.com/cancer<br />

info@personalis.com<br />

+1 855 436-6634 | +1 650 752-1300<br />

© 2015 Personalis, Inc. All rights reserved. Personalis ® , ACE Exome ® , ACE Clinical Exome, and ACE Genome ® are<br />

registered trademarks or trademarks <strong>of</strong> Personalis, Inc., (“Personalis”) in the United States and/or other countries.


The <strong>Best</strong> <strong>of</strong> the <strong>AACR</strong> <strong>Journals</strong><br />

The <strong>AACR</strong> is proud to present “The <strong>Best</strong> <strong>of</strong> the <strong>AACR</strong> <strong>Journals</strong>,” a collection <strong>of</strong> the most-cited articles published in 2013 across the<br />

<strong>AACR</strong> journal portfolio. We congratulate the authors <strong>of</strong> these outstanding articles for their tireless efforts and contributions to the<br />

cancer research field.<br />

Each journal’s section in the collection includes the most-cited review article and the four most-cited research articles based on the<br />

total number <strong>of</strong> citations from date <strong>of</strong> publication through January 2015.<br />

Table <strong>of</strong> Contents<br />

Cancer Discovery<br />

The Basic Principles <strong>of</strong> Chimeric Antigen Receptor Design<br />

Michel Sadelain, Renier Brentjens, and Isabelle Rivière .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .1<br />

Activating HER2 Mutations in HER2 Gene Amplification Negative Breast Cancer<br />

Ron Bose, Shyam M. Kavuri, Adam C. Searleman, Wei Shen, Dong Shen, Daniel C. Koboldt, John Monsey, Nicholas Goel,<br />

Adam B. Aronson, Shunqiang Li, Cynthia X. Ma, Li Ding, Elaine R. Mardis, and Matthew J. Ellis .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .2<br />

First-in-Humans Trial <strong>of</strong> an RNA Interference Therapeutic Targeting VEGF<br />

and KSP in Cancer Patients with Liver Involvement<br />

Josep Tabernero, Ge<strong>of</strong>frey I. Shapiro, Patricia M. LoRusso, Andres Cervantes, Gary K. Schwartz, Glen J. Weiss, Luis Paz-Ares,<br />

Daniel C. Cho, Jeffrey R. Infante, Maria Alsina, Mrinal M. Gounder, Rick Falzone, Jamie Harrop, Amy C. Seila White,<br />

Iva Toudjarska, David Bumcrot, Rachel E. Meyers, Gregory Hinkle, Nenad Svrzikapa, Renta M. Hutabarat, Valerie A. Clausen,<br />

Jeffrey Cehelsky, Saraswathy V. Nochur, Christina Gamba-Vitalo, Akshay K. Vaishnaw, Dinah W.Y. Sah, Jared A. Gollob,<br />

and Howard A. Burris III .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .2<br />

Amplification <strong>of</strong> the MET Receptor Drives Resistance to Anti-EGFR Therapies in Colorectal Cancer<br />

Alberto Bardelli, Simona Corso, Andrea Bertotti, Sebastijan Hobor, Emanuele Valtorta, Giulia Siravegna,<br />

Andrea Sartore-Bianchi, Elisa Scala, Andrea Cassingena, Davide Zecchin, Maria Apicella, Giorgia Migliardi,<br />

Francesco Galimi, Calogero Lauricella, Carlo Zanon, Timothy Perera, Silvio Veronese, Giorgio Corti,<br />

Alessio Amatu, Marcello Gambacorta, Luis A. Diaz Jr, Mark Sausen, Victor E. Velculescu, Paolo Comoglio,<br />

Livio Trusolino, Federica Di Nicolantonio, Silvia Giordano, and Salvatore Siena .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .3<br />

Identification <strong>of</strong> Targetable FGFR Gene Fusions in Diverse Cancers<br />

Yi-Mi Wu, Fengyun Su, Shanker Kalyana-Sundaram, Nickolay Khazanov, Bushra Ateeq, Xuhong Cao, Robert J. Lonigro, Pankaj Vats,<br />

Rui Wang, Su-Fang Lin, Ann-Joy Cheng, Lakshmi P. Kunju, Javed Siddiqui, Scott A. Tomlins, Peter Wyngaard, Seth Sadis,<br />

Sameek Roychowdhury, Maha H. Hussain, Felix Y. Feng, Mark M. Zalupski, Moshe Talpaz, Kenneth J. Pienta, Daniel R. Rhodes,<br />

Dan R. Robinson, and Arul M. Chinnaiyan .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .3<br />

Cancer Epidemiology, Biomarkers & Prevention<br />

Oral Contraceptive Use and Risk <strong>of</strong> Breast, Cervical, Colorectal, and Endometrial Cancers: A Systematic Review<br />

Jennifer M. Gierisch, Remy R. Coeytaux, Rachel Peragallo Urrutia, Laura J. Havrilesky, Patricia G. Moorman,<br />

William J. Lowery, Michaela Dinan, Amanda J. McBroom, Vic Hasselblad, Gillian D. Sanders, and Evan R. Myers . .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .4<br />

Cancer Survivors in the United States: Prevalence across the Survivorship<br />

Trajectory and Implications for Care<br />

Janet S. de Moor, Angela B. Mariotto, Carla Parry, Catherine M. Alfano, Lynne Padgett, Erin E. Kent, Laura Forsythe,<br />

Steve Scoppa, Mark Hachey, and Julia H. Rowland .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .5<br />

Vegetarian Diets and the Incidence <strong>of</strong> Cancer in a Low-risk Population<br />

Yessenia Tantamango-Bartley, Karen Jaceldo-Siegl, Jing Fan, and Gary Fraser . .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .5<br />

Genetic Susceptibility Loci for Subtypes <strong>of</strong> Breast Cancer in an African American Population<br />

Julie R. Palmer, Edward A. Ruiz-Narvaez, Charles N. Rotimi, L. Adrienne Cupples, Yvette C. Cozier,<br />

Lucile L. Adams-Campbell, and Lynn Rosenberg .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .6<br />

Drinking Water Arsenic in Northern Chile: High Cancer Risks 40 Years after Exposure Cessation<br />

Craig M. Steinmaus, Catterina Ferreccio, Johanna Acevedo Romo, Yan Yuan, Sandra Cortes, Guillermo Marshall,<br />

Lee E. Moore, John R. Balmes, Jane Liaw, Todd Golden, and Allan H. Smith .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .6<br />

Cancer Immunology Research<br />

Getting Personal with Neoantigen-Based Therapeutic Cancer Vaccines<br />

Nir Hacohen, Edward F. Fritsch, Todd A. Carter, Eric S. Lander, and Catherine J. Wu .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .7<br />

Anti-CTLA-4 Antibodies <strong>of</strong> IgG2a Isotype Enhance Antitumor Activity through Reduction<br />

<strong>of</strong> Intratumoral Regulatory T Cells<br />

Mark J. Selby, John J. Engelhardt, Michael Quigley, Karla A. Henning, Timothy Chen, Mohan Srinivasan, and Alan J. Korman .. .. .. .. .. .. .. .. .. .. .8<br />

The <strong>Best</strong> <strong>of</strong> the <strong>AACR</strong> <strong>Journals</strong><br />

i


Table <strong>of</strong> Contents<br />

An Abscopal Response to Radiation and Ipilimumab in a Patient with Metastatic Non–Small Cell Lung Cancer<br />

Encouse B. Golden, Sandra Demaria, Peter B. Schiff, Abraham Chachoua, and Silvia C. Formenti .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .8<br />

T Cells Expressing Chimeric Antigen Receptors Can Cause Anaphylaxis in Humans<br />

Marcela V. Maus, Andrew R. Haas, Gregory L. Beatty, Steven M. Albelda, Bruce L. Levine,<br />

Xiaojun Liu, Yangbing Zhao, Michael Kalos, and Carl H. June . .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .9<br />

PD-L1 Expression in the Merkel Cell Carcinoma Microenvironment: Association with<br />

Inflammation, Merkel Cell Polyomavirus, and Overall Survival<br />

Evan J. Lipson, Jeremy G. Vincent, Myriam Loyo, Luciane T. Kagohara, Brandon S. Luber, Hao Wang, Haiying Xu,<br />

Suresh K. Nayar, Timothy S. Wang, David Sidransky, Robert A. Anders, Suzanne L. Topalian, and Janis M. Taube .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .9<br />

Cancer Prevention Research<br />

New Perspectives <strong>of</strong> Curcumin in Cancer Prevention<br />

Wungki Park, A.R.M. Ruhul Amin, Zhuo Georgia Chen, and Dong M. Shin .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. ..10<br />

Durable Antibody Responses Following One Dose <strong>of</strong> the Bivalent Human Papillomavirus<br />

L1 Virus-Like Particle Vaccine in the Costa Rica Vaccine Trial<br />

Mahboobeh Safaeian, Carolina Porras, Yuanji Pan, Aimee Kreimer, John T. Schiller, Paula Gonzalez, Douglas R. Lowy,<br />

Sholom Wacholder, Mark Schiffman, Ana C. Rodriguez, Rolando Herrero, Troy Kemp, Gloriana Shelton, Wim Quint,<br />

Leen-Jan van Doorn, Allan Hildesheim, and Ligia A. Pinto, for the CVT Group .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. ..11<br />

MicroRNA Expression Signatures during Malignant Progression from Barrett’s Esophagus<br />

to Esophageal Adenocarcinoma<br />

Xifeng Wu, Jaffer A. Ajani, Jian Gu, David W. Chang, Weiqi Tan, Michelle A.T. Hildebrandt, Maosheng Huang,<br />

Kenneth K. Wang, and Ernest Hawk .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. ..11<br />

MUC1 Vaccine for Individuals with Advanced Adenoma <strong>of</strong> the Colon: A Cancer<br />

Immunoprevention Feasibility Study<br />

Takashi Kimura, John R. McKolanis, Lynda A. Dzubinski, Kazi Islam, Douglas M. Potter, Andres M. Salazar,<br />

Robert E. Schoen, and Olivera J. Finn . .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. ..12<br />

Increased Levels <strong>of</strong> Urinary PGE-M, a Biomarker <strong>of</strong> Inflammation, Occur in Association<br />

with Obesity, Aging, and Lung Metastases in Patients with Breast Cancer<br />

Patrick G. Morris, Xi Kathy Zhou, Ginger L. Milne, Daniel Goldstein, Laura C. Hawks, Chau T. Dang, Shanu Modi,<br />

Monica N. Fornier, Clifford A. Hudis, and Andrew J. Dannenberg .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. ..12<br />

Cancer Research<br />

Genome-Wide Epigenetic Regulation <strong>of</strong> miRNAs in Cancer<br />

Constance Baer, Rainer Claus, and Christoph Plass .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. ..13<br />

The Noncoding RNA MALAT1 Is a Critical Regulator <strong>of</strong> the Metastasis Phenotype <strong>of</strong> Lung Cancer Cells<br />

Tony Gutschner, Monika Hämmerle, Moritz Eißmann, Jeff Hsu, Youngsoo Kim, Gene Hung, Alexey Revenko,<br />

Gayatri Arun, Marion Stentrup, Matthias Groß, Martin Zörnig, A. Robert MacLeod, David L. Spector, and Sven Diederichs .. .. .. .. .. .. .. .. .. .. ..14<br />

Androgen Receptor Splice Variants Mediate Enzalutamide Resistance<br />

in Castration-Resistant Prostate Cancer Cell Lines<br />

Yingming Li, Siu Chiu Chan, Lucas J. Brand, Tae Hyun Hwang, Kevin A.T. Silverstein, and Scott M. Dehm .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. ..14<br />

Complex Tumor Genomes Inferred from Single Circulating Tumor Cells by<br />

Array-CGH and Next-Generation Sequencing<br />

Ellen Heitzer, Martina Auer, Christin Gasch, Martin Pichler, Peter Ulz, Eva Maria H<strong>of</strong>fmann, Sigurd Lax, Julie Waldispuehl-Geigl,<br />

Oliver Mauermann, Carolin Lackner, Gerald Höfler, Florian Eisner, Heinz Sill, Hellmut Samonigg, Klaus Pantel, Sabine Riethdorf,<br />

Thomas Bauernh<strong>of</strong>er, Jochen B. Geigl, and Michael R. Speicher .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. ..15<br />

Targeting Tumor-Infiltrating Macrophages Decreases Tumor-Initiating Cells, Relieves<br />

Immunosuppression, and Improves Chemotherapeutic Responses<br />

Jonathan B. Mitchem, Donal J. Brennan, Brett L. Knolh<strong>of</strong>f, Brian A. Belt, Yu Zhu, Dominic E. Sanford, Larisa Belaygorod,<br />

Danielle Carpenter, Lynne Collins, David Piwnica-Worms, Stephen Hewitt, Girish Mallya Udupi, William M. Gallagher,<br />

Craig Wegner, Brian L. West, Andrea Wang-Gillam, Peter Goedegebuure, David C. Linehan, and David G. DeNardo .. .. .. .. .. .. .. .. .. .. .. .. .. .. ..15<br />

Clinical Cancer Research<br />

The Definition <strong>of</strong> Primary and Secondary Glioblastoma<br />

Hiroko Ohgaki and Paul Kleihues .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. ..16<br />

BRAF Inhibition Is Associated with Enhanced Melanoma Antigen Expression and a More Favorable Tumor<br />

Microenvironment in Patients with Metastatic Melanoma<br />

Dennie T. Frederick, Adriano Piris, Alexandria P. Cogdill, Zachary A. Cooper, Cecilia Lezcano, Cristina R. Ferrone, Devarati Mitra,<br />

Andrea Boni, Lindsay P. Newton, Chengwen Liu, Weiyi Peng, Ryan J. Sullivan, Donald P. Lawrence, F. Stephen Hodi, Willem W. Overwijk,<br />

Gregory Lizée, George F. Murphy, Patrick Hwu, Keith T. Flaherty, David E. Fisher, and Jennifer A. Wargo .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. ..17<br />

ii<br />

aacrjournals.org


Table <strong>of</strong> Contents<br />

Analysis <strong>of</strong> Tumor Specimens at the Time <strong>of</strong> Acquired Resistance to EGFR-TKI Therapy<br />

in 155 Patients with EGFR-Mutant Lung Cancers<br />

Helena A. Yu, Maria E. Arcila, Natasha Rekhtman, Camelia S. Sima, Maureen F. Zakowski, William Pao,<br />

Mark G. Kris, Vincent A. Miller, Marc Ladanyi, and Gregory J. Riely .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. ..17<br />

Durable Cancer Regression Off-Treatment and Effective Reinduction Therapy with an Anti-PD-1 Antibody<br />

Evan J. Lipson, William H. Sharfman, Charles G. Drake, Ira Wollner, Janis M. Taube, Robert A. Anders, Haiying Xu,<br />

Sheng Yao, Alice Pons, Lieping Chen, Drew M. Pardoll, Julie R. Brahmer, and Suzanne L. Topalian .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. ..18<br />

An Epithelial–Mesenchymal Transition Gene Signature Predicts Resistance to EGFR and PI3K<br />

Inhibitors and Identifies Axl as a Therapeutic Target for Overcoming EGFR Inhibitor Resistance<br />

Lauren Averett Byers, Lixia Diao, Jing Wang, Pierre Saintigny, Luc Girard, Michael Peyton, Li Shen, Youhong Fan, Uma Giri, Praveen<br />

K. Tumula, Monique B. Nilsson, Jayanthi Gudikote, Hai Tran, Robert J.G. Cardnell, David J. Bearss, Steven L. Warner, Jason M. Foulks,<br />

Steven B. Kanner, Varsha Gandhi, Nancy Krett, Steven T. Rosen, Edward S. Kim, Roy S. Herbst, George R. Blumenschein, J. Jack Lee, Scott<br />

M. Lippman, K. Kian Ang, Gordon B. Mills, Waun K. Hong, John N. Weinstein, Ignacio I. Wistuba, Kevin R. Coombes, John D. Minna,<br />

and John V. Heymach .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. ..18<br />

Molecular Cancer Research<br />

Radiation Survivors: Understanding and Exploiting the Phenotype following Fractionated Radiation Therapy<br />

Adeola Y. Makinde, Molykutty John-Aryankalayil, Sanjeewani T. Palayoor, David Cerna, and C. Norman Coleman .. .. .. .. .. .. .. .. .. .. .. .. .. .. ..19<br />

A Modified HSP70 Inhibitor Shows Broad Activity as an Anticancer Agent<br />

Gregor M. Balaburski, Julia I.-Ju Leu, Neil Beeharry, Seth Hayik, Mark D. Andrake, Gao Zhang, Meenhard Herlyn,<br />

Jessie Villanueva, Roland L. Dunbrack, Tim Yen, Donna L. George, and Maureen E. Murphy .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. ..20<br />

The Heterochronic microRNA let-7 Inhibits Cell Motility by Regulating the Genes in the<br />

Actin Cytoskeleton Pathway in Breast Cancer<br />

Xiaowen Hu, Jinyi Guo, Lan Zheng, Chunsheng Li, Tim M. Zheng, Janos L. Tanyi, Shun Liang, Chiara Benedetto,<br />

Marco Mitidieri, Dionyssios Katsaros, Xia Zhao, Youcheng Zhang, Qihong Huang, and Lin Zhang .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. ..20<br />

Procollagen Lysyl Hydroxylase 2 Is Essential for Hypoxia-Induced Breast Cancer Metastasis<br />

Daniele M. Gilkes, Saumendra Bajpai, Carmen C. Wong, Pallavi Chaturvedi, Maimon E. Hubbi, Denis Wirtz,<br />

and Gregg L. Semenza .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. ..21<br />

Activation <strong>of</strong> the FGF2-FGFR1 Autocrine Pathway: A Novel Mechanism <strong>of</strong> Acquired Resistance to Gefitinib in NSCLC<br />

Hideki Terai, Kenzo Soejima, Hiroyuki Yasuda, Sohei Nakayama, Junko Hamamoto, Daisuke Arai, Kota Ishioka,<br />

Keiko Ohgino, Shinnosuke Ikemura, Takashi Sato, Satoshi Yoda, Ryosuke Satomi, Katsuhiko Naoki, and Tomoko Betsuyaku . .. .. .. .. .. .. .. .. .. ..21<br />

Molecular Cancer Therapeutics<br />

FOX(M1) News—It Is Cancer<br />

Marianna Halasi and Andrei L. Gartel . .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. ..22<br />

Genome and Transcriptome Sequencing in Prospective Metastatic Triple-Negative<br />

Breast Cancer Uncovers Therapeutic Vulnerabilities<br />

David W. Craig, Joyce A. O’Shaughnessy, Jeffrey A. Kiefer, Jessica Aldrich, Shripad Sinari, Tracy M. Moses,<br />

Shukmei Wong, Jennifer Dinh, Alexis Christ<strong>of</strong>orides, Joanne L. Blum, Cristi L. Aitelli, Cynthia R. Osborne,<br />

Tyler Izatt, Ahmet Kurdoglu, Angela Baker, Julie Koeman, Catalin Barbacioru, Onur Sakarya,<br />

Francisco M. De La Vega, Asim Siddiqui, Linh Hoang, Paul R. Billings, Bodour Salhia, Anthony W. Tolcher,<br />

Jeffrey M. Trent, Spyro Mousses, Daniel Von H<strong>of</strong>f, and John D. Carpten .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. ..23<br />

EGFR Exon 20 Insertion Mutations in Lung Adenocarcinomas: Prevalence,<br />

Molecular Heterogeneity, and Clinicopathologic Characteristics<br />

Maria E. Arcila, Khedoudja Nafa, Jamie E. Chaft, Natasha Rekhtman, Christopher Lau,<br />

Boris A. Reva, Maureen F. Zakowski, Mark G. Kris, and Marc Ladanyi . .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. ..23<br />

NF-kB2/p52 Induces Resistance to Enzalutamide in Prostate Cancer:<br />

Role <strong>of</strong> Androgen Receptor and Its Variants<br />

Nagalakshmi Nadiminty, Ramakumar Tummala, Chengfei Liu, Joy Yang, Wei Lou,<br />

Christopher P. Evans, and Allen C. Gao .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. ..24<br />

Inhibition <strong>of</strong> Mutant GNAQ Signaling in Uveal Melanoma Induces<br />

AMPK-Dependent Autophagic Cell Death<br />

Grazia Ambrosini, Elgilda Musi, Alan L. Ho, Elisa de Stanchina, and Gary K. Schwartz .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. .. ..24<br />

The <strong>Best</strong> <strong>of</strong> the <strong>AACR</strong> <strong>Journals</strong><br />

iii


JOIN THE <strong>AACR</strong> NETWORK<br />

CONTENT • CONVENIENCE • CONNECTIONS<br />

OUR SOCIAL NETWORK<br />

Facebook.com/<strong>AACR</strong><br />

#<strong>AACR</strong><br />

LinkedIn.com<br />

YouTube.com/<strong>AACR</strong>News<br />

http://blog.<strong>AACR</strong>.org<br />

DON<br />

DONATE<br />

OUR <strong>AACR</strong> NETWORK<br />

<strong>AACR</strong>.org<br />

<strong>AACR</strong>journals.org<br />

CancerCareers.org<br />

CancerProgressReport.org<br />

CancerTodayMag.org<br />

CONNECT WITH US<br />

Follow <strong>AACR</strong> on social media, receive notification <strong>of</strong> significant findings published in<br />

<strong>AACR</strong> journals, and converse with other attendees during our conferences.


Cancer<br />

Cancer Discovery Discovery<br />

Cancer Discovery<br />

CANCER<br />

DISCOVERY<br />

FEBRUARY<br />

www.aacrjournals.org<br />

Lewis C. Cantley, PhD<br />

Editor-in-Chief<br />

José Baselga, MD, PhD<br />

Editor-in-Chief<br />

Scope<br />

Cancer Discovery publishes high-impact, peerreviewed<br />

articles describing major advances in<br />

research and clinical trials. As the premier cancer<br />

information resource, the Journal presents<br />

Review Articles, Perspectives and Commentaries,<br />

News stories, and Research Watch summaries<br />

<strong>of</strong> important journal articles to its readers to<br />

keep them informed about the latest findings<br />

in the field. Topics span the spectrum <strong>of</strong> cancer<br />

research and medicine from the laboratory to the<br />

clinic and epidemiologic studies.<br />

Review Article<br />

The Basic Principles <strong>of</strong> Chimeric Antigen Receptor Design<br />

Michel Sadelain, Renier Brentjens, and Isabelle Rivière<br />

Abstract<br />

Chimeric antigen receptors (CAR) are recombinant receptors<br />

that provide both antigen-binding and T-cell–activating<br />

functions. A multitude <strong>of</strong> CARs has been reported over the past<br />

decade, targeting an array <strong>of</strong> cell surface tumor antigens. Their<br />

biologic functions have dramatically changed following the<br />

introduction <strong>of</strong> tripartite receptors comprising a costimulatory<br />

domain, termed second-generation CARs. These have recently<br />

shown clinical benefit in patients treated with CD19-targeted<br />

autologous T cells. CARs may be combined with costimulatory<br />

ligands, chimeric costimulatory receptors, or cytokines to<br />

further enhance T-cell potency, specificity, and safety. CARs<br />

represent a new class <strong>of</strong> drugs with exciting potential for cancer<br />

immunotherapy.<br />

Michel Sadelain<br />

Significance: CARs are a new class <strong>of</strong> drugs with great potential for<br />

cancer immunotherapy. Upon their expression in T lymphocytes,<br />

CARs direct potent, targeted immune responses that have recently<br />

shown encouraging clinical outcomes in a subset <strong>of</strong> patients with<br />

B-cell malignancies. This review focuses on the design <strong>of</strong> CARs,<br />

including the requirements for optimal antigen recognition and<br />

different modalities to provide costimulatory support to targeted T<br />

cells, which include the use <strong>of</strong> second- and third-generation CARs,<br />

costimulatory ligands, chimeric costimulatory receptors, and<br />

cytokines. Cancer Discov; 3(4); 388–98. ©2013 <strong>AACR</strong>.<br />

Cancer Discov April 2013 3:388–98; Published OnlineFirst April 2,<br />

2013; doi:10.1158/2159-8290.CD-12-0548<br />

The <strong>Best</strong> <strong>of</strong> the <strong>AACR</strong> <strong>Journals</strong><br />

1


Cancer Discovery<br />

Research Article<br />

Activating HER2 Mutations in HER2 Gene<br />

Amplification Negative Breast Cancer<br />

Ron Bose, Shyam M. Kavuri, Adam C. Searleman, Wei Shen, Dong Shen, Daniel C. Koboldt,<br />

John Monsey, Nicholas Goel, Adam B. Aronson, Shunqiang Li, Cynthia X. Ma, Li Ding,<br />

Elaine R. Mardis, and Matthew J. Ellis<br />

Ron Bose<br />

Matthew J. Ellis<br />

Abstract<br />

Data from 8 breast cancer genome-sequencing projects identified<br />

25 patients with HER2 somatic mutations in cancers lacking<br />

HER2 gene amplification. To determine the phenotype <strong>of</strong> these<br />

mutations, we functionally characterized 13 HER2 mutations<br />

using in vitro kinase assays, protein structure analysis, cell<br />

culture, and xenograft experiments. Seven <strong>of</strong> these mutations<br />

are activating mutations, including G309A, D769H, D769Y,<br />

V777L, P780ins, V842I, and R896C. HER2 in-frame deletion<br />

755–759, which is homologous to EGF receptor (EGFR) exon<br />

19 in-frame deletions, had a neomorphic phenotype with<br />

increased phosphorylation <strong>of</strong> EGFR or HER3. L755S produced<br />

lapatinib resistance, but was not an activating mutation in our<br />

experimental systems. All <strong>of</strong> these mutations were sensitive to<br />

the irreversible kinase inhibitor, neratinib. These findings show<br />

that HER2 somatic mutation is an alternative mechanism to<br />

activate HER2 in breast cancer and they validate HER2 somatic<br />

mutations as drug targets for breast cancer treatment.<br />

Significance: We show that the majority <strong>of</strong> HER2 somatic<br />

mutations in breast cancer patients are activating mutations<br />

that likely drive tumorigenesis. Several patients had mutations<br />

that are resistant to the reversible HER2 inhibitor lapatinib, but<br />

are sensitive to the irreversible HER2 inhibitor, neratinib. Our<br />

results suggest that patients with HER2 mutation–positive breast<br />

cancers could benefit from existing HER2-targeted drugs. Cancer<br />

Discov; 3(2); 224–37. ©2013 <strong>AACR</strong>.<br />

Cancer Discov February 2013 3:224–37; Published OnlineFirst<br />

December 7, 2012; doi:10.1158/2159-8290.CD-12-0349<br />

Research Article<br />

First-in-Humans Trial <strong>of</strong> an RNA Interference Therapeutic Targeting<br />

VEGF and KSP in Cancer Patients with Liver Involvement<br />

Josep Tabernero, Ge<strong>of</strong>frey I. Shapiro, Patricia M. LoRusso, Andres Cervantes, Gary K. Schwartz, Glen J. Weiss, Luis Paz-Ares,<br />

Daniel C. Cho, Jeffrey R. Infante, Maria Alsina, Mrinal M. Gounder, Rick Falzone, Jamie Harrop, Amy C. Seila White,<br />

Iva Toudjarska, David Bumcrot, Rachel E. Meyers, Gregory Hinkle, Nenad Svrzikapa, Renta M. Hutabarat, Valerie A. Clausen,<br />

Jeffrey Cehelsky, Saraswathy V. Nochur, Christina Gamba-Vitalo, Akshay K. Vaishnaw, Dinah W.Y. Sah, Jared A. Gollob,<br />

and Howard A. Burris III<br />

Josep Tabernero<br />

Abstract<br />

RNA interference (RNAi) is a potent and specific mechanism for<br />

regulating gene expression. Harnessing RNAi to silence genes<br />

involved in disease holds promise for the development <strong>of</strong> a new<br />

class <strong>of</strong> therapeutics. Delivery is key to realizing the potential<br />

<strong>of</strong> RNAi, and lipid nanoparticles (LNP) have proved effective<br />

in delivery <strong>of</strong> siRNAs to the liver and to tumors in animals. To<br />

examine the activity and safety <strong>of</strong> LNP-formulated siRNAs in<br />

humans, we initiated a trial <strong>of</strong> ALN-VSP, an LNP formulation<br />

<strong>of</strong> siRNAs targeting VEGF and kinesin spindle protein (KSP),<br />

in patients with cancer. Here, we show detection <strong>of</strong> drug in<br />

tumor biopsies, siRNA-mediated mRNA cleavage in the liver,<br />

pharmacodynamics suggestive <strong>of</strong> target downregulation, and<br />

antitumor activity, including complete regression <strong>of</strong> liver<br />

metastases in endometrial cancer. In addition, we show that<br />

biweekly intravenous administration <strong>of</strong> ALN-VSP was safe<br />

and well tolerated. These data provide pro<strong>of</strong>-<strong>of</strong>-concept for<br />

RNAi therapeutics in humans and form the basis for further<br />

development in cancer.<br />

Significance: The findings in this report show safety,<br />

pharmacokinetics, RNAi mechanism <strong>of</strong> action, and clinical<br />

activity with a novel first-in-class LNP-formulated RNAi<br />

therapeutic in patients with cancer. The ability to harness RNAi to<br />

facilitate specific multitargeting, as well as increase the number<br />

<strong>of</strong> druggable targets, has important implications for future drug<br />

development in oncology. Cancer Discov; 3(4); 406–17. ©2012 <strong>AACR</strong>.<br />

Cancer Discov April 2013 3:406–17; Published OnlineFirst January<br />

28, 2013; doi:10.1158/2159-8290.CD-12-0429<br />

2<br />

aacrjournals.org


Cancer Discovery<br />

Research Article<br />

Amplification <strong>of</strong> the MET Receptor Drives Resistance to<br />

Anti-EGFR Therapies in Colorectal Cancer<br />

Alberto Bardelli, Simona Corso, Andrea Bertotti, Sebastijan Hobor, Emanuele Valtorta, Giulia Siravegna,<br />

Andrea Sartore-Bianchi, Elisa Scala, Andrea Cassingena, Davide Zecchin, Maria Apicella, Giorgia Migliardi,<br />

Francesco Galimi, Calogero Lauricella, Carlo Zanon, Timothy Perera, Silvio Veronese, Giorgio Corti,<br />

Alessio Amatu, Marcello Gambacorta, Luis A. Diaz Jr, Mark Sausen, Victor E. Velculescu, Paolo Comoglio,<br />

Livio Trusolino, Federica Di Nicolantonio, Silvia Giordano, and Salvatore Siena<br />

Alberto Bardelli<br />

Silvia Giordano<br />

Abstract<br />

EGF receptor (EGFR)-targeted monoclonal antibodies are<br />

effective in a subset <strong>of</strong> metastatic colorectal cancers. Inevitably,<br />

all patients develop resistance, which occurs through emergence<br />

<strong>of</strong> KRAS mutations in approximately 50% <strong>of</strong> the cases. We show<br />

that amplification <strong>of</strong> the MET proto-oncogene is associated<br />

with acquired resistance in tumors that do not develop KRAS<br />

mutations during anti-EGFR therapy. Amplification <strong>of</strong> the MET<br />

locus was present in circulating tumor DNA before relapse was<br />

clinically evident. Functional studies show that MET activation<br />

confers resistance to anti-EGFR therapy both in vitro and in<br />

vivo. Notably, in patient-derived colorectal cancer xenografts,<br />

MET amplification correlated with resistance to EGFR blockade,<br />

which could be overcome by MET kinase inhibitors. These results<br />

highlight the role <strong>of</strong> MET in mediating primary and secondary<br />

resistance to anti-EGFR therapies in colorectal cancer and<br />

encourage the use <strong>of</strong> MET inhibitors in patients displaying<br />

resistance as a result <strong>of</strong> MET amplification.<br />

Significance: Amplification <strong>of</strong> the MET proto-oncogene is<br />

responsible for de novo and acquired resistance to anti-EGFR<br />

therapy in a subset <strong>of</strong> colorectal cancers. As multiple anti-<br />

MET therapeutic strategies are available, these findings <strong>of</strong>fer<br />

immediate novel opportunities to design clinical studies. Cancer<br />

Discov; 3(6); 658–73. ©2013 <strong>AACR</strong>.<br />

Cancer Discov June 2013 3:658–73; Published OnlineFirst June 6,<br />

2013; doi:10.1158/2159-8290.CD-12-0558<br />

Research Brief<br />

Identification <strong>of</strong> Targetable FGFR Gene Fusions in<br />

Diverse Cancers<br />

Yi-Mi Wu, Fengyun Su, Shanker Kalyana-Sundaram, Nickolay Khazanov, Bushra Ateeq, Xuhong Cao,<br />

Robert J. Lonigro, Pankaj Vats, Rui Wang, Su-Fang Lin, Ann-Joy Cheng, Lakshmi P. Kunju, Javed Siddiqui,<br />

Scott A. Tomlins, Peter Wyngaard, Seth Sadis, Sameek Roychowdhury, Maha H. Hussain, Felix Y. Feng,<br />

Mark M. Zalupski, Moshe Talpaz, Kenneth J. Pienta, Daniel R. Rhodes, Dan R. Robinson, and<br />

Arul M. Chinnaiyan<br />

Dan R. Robinson<br />

Arul M. Chinnaiyan<br />

Abstract<br />

Through a prospective clinical sequencing program for<br />

advanced cancers, four index cases were identified which<br />

harbor gene rearrangements <strong>of</strong> FGFR2, including patients with<br />

cholangiocarcinoma, breast cancer, and prostate cancer. After<br />

extending our assessment <strong>of</strong> FGFR rearrangements across<br />

multiple tumor cohorts, we identified additional FGFR fusions<br />

with intact kinase domains in lung squamous cell cancer, bladder<br />

cancer, thyroid cancer, oral cancer, glioblastoma, and head and<br />

neck squamous cell cancer. All FGFR fusion partners tested<br />

exhibit oligomerization capability, suggesting a shared mode<br />

<strong>of</strong> kinase activation. Overexpression <strong>of</strong> FGFR fusion proteins<br />

induced cell proliferation. Two bladder cancer cell lines that<br />

harbor FGFR3 fusion proteins exhibited enhanced susceptibility<br />

to pharmacologic inhibition in vitro and in vivo. Because <strong>of</strong> the<br />

combinatorial possibilities <strong>of</strong> FGFR family fusion to a variety<br />

<strong>of</strong> oligomerization partners, clinical sequencing efforts, which<br />

incorporate transcriptome analysis for gene fusions, are poised<br />

to identify rare, targetable FGFR fusions across diverse cancer<br />

types.<br />

Significance: High-throughput sequencing technologies facilitate<br />

defining the mutational landscape <strong>of</strong> human cancers, which will<br />

lead to more precise treatment <strong>of</strong> patients with cancer. Here,<br />

through integrative sequencing efforts, we identified a variety <strong>of</strong><br />

FGFR gene fusions in a spectrum <strong>of</strong> human cancers. FGFR fusions<br />

are active kinases. Cells harboring FGFR fusions showed enhanced<br />

sensitivity to the FGFR inhibitors PD173074 and pazopanib,<br />

suggesting that patients with cancer with FGFR fusions may<br />

benefit from targeted FGFR kinase inhibition. Cancer Discov; 3(6);<br />

636–47. ©2013 <strong>AACR</strong>.<br />

Cancer Discov June 2013 3:636–47; Published OnlineFirst April 4,<br />

2013; doi:10.1158/2159-8290.CD-13-0050<br />

The <strong>Best</strong> <strong>of</strong> the <strong>AACR</strong> <strong>Journals</strong> 3


In collaboration with the<br />

<strong>AACR</strong> Molecular<br />

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

Preventive Oncology<br />

Cancer<br />

Epidemiology,<br />

Biomarkers &<br />

Prevention<br />

Cancer Epidemiology,<br />

Biomarkers & Prevention<br />

Cancer<br />

Epidemiology,<br />

Biomarkers<br />

& Prevention<br />

April 2013<br />

Volume 22 • Number 4<br />

Pages 479–744<br />

Translating Science to Populations<br />

www.aacrjournals.org<br />

SPECIAL CONTENT<br />

IN THIS ISSUE!<br />

Epidemiology Group<br />

and the<br />

Timothy R. Rebbeck, PhD<br />

Editor-in-Chief<br />

Scope<br />

Cancer Epidemiology, Biomarkers & Prevention publishes original peerreviewed,<br />

population-based research on cancer etiology, prevention,<br />

surveillance, and survivorship. The following topics are <strong>of</strong> special interest:<br />

descriptive, analytical, and molecular epidemiology; biomarkers including<br />

assay development, validation, and application; chemoprevention and<br />

other types <strong>of</strong> prevention research in the context <strong>of</strong> descriptive and<br />

observational studies; the role <strong>of</strong> behavioral factors in cancer etiology<br />

and prevention; survivorship studies; risk factors; and the science <strong>of</strong> cancer<br />

health disparities. Besides welcoming manuscripts that address individual<br />

subjects in any <strong>of</strong> the relevant disciplines, CEBP editors encourage the<br />

submission <strong>of</strong> manuscripts with a transdisciplinary approach.<br />

Review Article<br />

Oral Contraceptive Use and Risk <strong>of</strong> Breast, Cervical,<br />

Colorectal, and Endometrial Cancers: A Systematic Review<br />

Jennifer M. Gierisch, Remy R. Coeytaux, Rachel Peragallo Urrutia, Laura J. Havrilesky, Patricia G. Moorman,<br />

William J. Lowery, Michaela Dinan, Amanda J. McBroom, Vic Hasselblad, Gillian D. Sanders, and Evan R. Myers<br />

Jennifer M. Gierisch<br />

Abstract<br />

Oral contraceptives may influence the risk <strong>of</strong> certain cancers.<br />

As part <strong>of</strong> the AHRQ Evidence Report, Oral Contraceptive Use<br />

for the Primary Prevention <strong>of</strong> Ovarian Cancer, we conducted<br />

a systematic review to estimate associations between oral<br />

contraceptive use and breast, cervical, colorectal, and<br />

endometrial cancer incidence. We searched PubMed, Embase,<br />

and Cochrane Database <strong>of</strong> Systematic Reviews. Study inclusion<br />

criteria were women taking oral contraceptives for contraception<br />

or ovarian cancer prevention; includes comparison group with<br />

no oral contraceptive use; study reports quantitative associations<br />

between oral contraceptive exposure and relevant cancers;<br />

controlled study or pooled patient-level meta-analyses; sample<br />

size for nonrandomized studies ≥100; peer-reviewed, Englishlanguage;<br />

published from January 1, 2000 forward. Randomeffects<br />

meta-analyses were conducted by estimating pooled ORs<br />

with 95% confidence intervals (CIs). We included 44 breast, 12<br />

cervical, 11 colorectal, and 9 endometrial cancers studies. Breast<br />

cancer incidence was slightly but significantly increased in users<br />

(OR, 1.08; CI, 1.00–1.17); results show a higher risk associated<br />

with more recent use <strong>of</strong> oral contraceptives. Risk <strong>of</strong> cervical<br />

cancer was increased with duration <strong>of</strong> oral contraceptive use<br />

in women with human papillomavirus infection; heterogeneity<br />

prevented meta-analysis. Colorectal cancer (OR, 0.86; CI, 0.79–<br />

0.95) and endometrial cancer incidences (OR, 0.57; CI, 0.43–0.77)<br />

were significantly reduced by oral contraceptive use. Compared<br />

with never use, ever use <strong>of</strong> oral contraceptives is significantly<br />

associated with decreases in colorectal and endometrial cancers<br />

and increases in breast cancers. Although elevated breast cancer<br />

risk was small, relatively high incidence <strong>of</strong> breast cancers means<br />

that oral contraceptives may contribute to a substantial number<br />

<strong>of</strong> cases. Cancer Epidemiol Biomarkers Prev; 22(11); 1931–43.<br />

©2013 <strong>AACR</strong>.<br />

Cancer Epidemiol Biomarkers Prev November 2013 22:1931–43;<br />

Published OnlineFirst September 6, 2013; doi:10.1158/1055-9965.<br />

EPI-13-0298<br />

4<br />

aacrjournals.org


Cancer Epidemiology, Biomarkers & Prevention<br />

Research Article<br />

Cancer Survivors in the United States: Prevalence across the<br />

Survivorship Trajectory and Implications for Care<br />

Janet S. de Moor, Angela B. Mariotto, Carla Parry, Catherine M. Alfano, Lynne Padgett, Erin E. Kent, Laura Forsythe,<br />

Steve Scoppa, Mark Hachey, and Julia H. Rowland<br />

Janet S. de Moor<br />

Abstract<br />

Background: Cancer survivors represent a growing population,<br />

heterogeneous in their need for medical care, psychosocial<br />

support, and practical assistance. To inform survivorship<br />

research and practice, this manuscript will describe the prevalent<br />

population <strong>of</strong> cancer survivors in terms <strong>of</strong> overall numbers and<br />

prevalence by cancer site and time since diagnosis.<br />

Methods: Incidence and survival data from 1975–2007 were<br />

obtained from the Surveillance, Epidemiology, and End Results<br />

Program and population projections from the United States<br />

Census Bureau. Cancer prevalence for 2012 and beyond was<br />

estimated using the Prevalence Incidence Approach Model,<br />

assuming constant future incidence and survival trends but<br />

dynamic projections <strong>of</strong> the U.S. population.<br />

Results: As <strong>of</strong> January 1, 2012, approximately 13.7 million<br />

cancer survivors were living in the United States with prevalence<br />

projected to approach 18 million by 2022. Sixty-four percent <strong>of</strong><br />

this population have survived 5 years or more; 40% have survived<br />

10 years or more; and 15% have survived 20 years or more after<br />

diagnosis. Over the next decade, the number <strong>of</strong> people who have<br />

lived 5 years or more after their cancer diagnosis is projected to<br />

increase approximately 37% to 11.9 million.<br />

Conclusions: A coordinated agenda for research and practice<br />

is needed to address cancer survivors’ long-term medical,<br />

psychosocial, and practical needs across the survivorship<br />

trajectory.<br />

Impact: Prevalence estimates for cancer survivors across the<br />

survivorship trajectory will inform the national research agenda<br />

as well as future projections about the health service needs <strong>of</strong><br />

this population. Cancer Epidemiol Biomarkers Prev; 22(4); 561–70.<br />

©2013 <strong>AACR</strong>.<br />

Cancer Epidemiol Biomarkers Prev April 2013 22:561–70;<br />

Published OnlineFirst March 27, 2013; doi:10.1158/1055-9965.<br />

EPI-12-1356<br />

Research Article<br />

Vegetarian Diets and the Incidence <strong>of</strong> Cancer in a<br />

Low-risk Population<br />

Yessenia Tantamango-Bartley, Karen Jaceldo-Siegl, Jing Fan, and Gary Fraser<br />

Abstract<br />

Background: Cancer is the second leading cause <strong>of</strong> death in<br />

the United States. Dietary factors account for at least 30% <strong>of</strong><br />

all cancers in Western countries. As people do not consume<br />

individual foods but rather combinations <strong>of</strong> them, the assessment<br />

<strong>of</strong> dietary patterns may <strong>of</strong>fer valuable information when<br />

determining associations between diet and cancer risk.<br />

Methods: We examined the association between dietary patterns<br />

(non-vegetarians, lacto, pesco, vegan, and semi-vegetarian) and the<br />

overall cancer incidence among 69,120 participants <strong>of</strong> the Adventist<br />

Health Study-2. Cancer cases were identified by matching to cancer<br />

registries. Cox proportional hazard regression analysis was conducted<br />

to estimate hazard ratios, with “attained age” as the time variable.<br />

Results: A total <strong>of</strong> 2,939 incident cancer cases were identified. The<br />

multivariate HR <strong>of</strong> overall cancer risk among vegetarians compared<br />

with non-vegetarians was statistically significant [HR, 0.92; 95%<br />

confidence interval (CI), 0.85–0.99] for both genders combined.<br />

Also, a statistically significant association was found between<br />

vegetarian diet and cancers <strong>of</strong> the gastrointestinal tract (HR, 0.76;<br />

Yessenia Tantamango-<br />

Bartley<br />

95% CI, 0.63–0.90). When analyzing the association <strong>of</strong> specific<br />

vegetarian dietary patterns, vegan diets showed statistically<br />

significant protection for overall cancer incidence (HR, 0.84; 95%<br />

CI, 0.72–0.99) in both genders combined and for female-specific<br />

cancers (HR, 0.66; 95% CI, 0.47–0.92). Lacto-ovo-vegetarians<br />

appeared to be associated with decreased risk <strong>of</strong> cancers <strong>of</strong> the<br />

gastrointestinal system (HR, 0.75; 95% CI, 0.60–0.92).<br />

Conclusion: Vegetarian diets seem to confer protection against<br />

cancer.<br />

Impact: Vegan diet seems to confer lower risk for overall and<br />

female-specific cancer than other dietary patterns. The lactoovo-vegetarian<br />

diets seem to confer protection from cancers<br />

<strong>of</strong> the gastrointestinal tract. Cancer Epidemiol Biomarkers Prev;<br />

22(2); 286–94. ©2012 <strong>AACR</strong>.<br />

Cancer Epidemiol Biomarkers Prev February 2013 22:286–94;<br />

Published OnlineFirst November 20, 2012; doi:10.1158/1055-9965.<br />

EPI-12-1060<br />

The <strong>Best</strong> <strong>of</strong> the <strong>AACR</strong> <strong>Journals</strong> 5


Cancer Epidemiology, Biomarkers & Prevention<br />

Research Article<br />

Genetic Susceptibility Loci for Subtypes <strong>of</strong> Breast Cancer in<br />

an African American Population<br />

Julie R. Palmer, Edward A. Ruiz-Narvaez, Charles N. Rotimi, L. Adrienne Cupples, Yvette C. Cozier,<br />

Lucile L. Adams-Campbell, and Lynn Rosenberg<br />

Julie R. Palmer<br />

Abstract<br />

Background: Most genome-wide association studies (GWAS)<br />

have been carried out in European ancestry populations; no<br />

risk variants for breast cancer have been identified solely from<br />

African ancestry GWAS data. Few GWAS hits have replicated in<br />

African ancestry populations.<br />

Methods: In a nested case–control study <strong>of</strong> breast cancer in the<br />

Black Women’s Health Study (1,199 cases/1,948 controls), we<br />

evaluated index single-nucleotide polymorphisms (SNP) in 21 loci<br />

from GWAS <strong>of</strong> European or Asian ancestry populations, overall,<br />

in subtypes defined by estrogen receptor (ER) and progesterone<br />

receptor (PR) status (ER+/PR+, n = 336; ER−/PR−, n = 229), and<br />

in triple-negative breast cancer (TNBC, N = 81). To evaluate the<br />

contribution <strong>of</strong> genetic factors to population differences in breast<br />

cancer subtype, we also examined global percent African ancestry.<br />

Results: Index SNPs in five loci were replicated, including three<br />

associated with ER−/PR− breast cancer (TERT rs10069690 in<br />

5p15.33, rs704010 in 10q22.3, and rs8170 in 19p13.11): per allele<br />

ORs were 1.29 [95% confidence interval (CI) 1.04–1.59], P = 0.02,<br />

1.52 (95% CI 1.12–2.08), P = 0.01, and 1.30 (95% CI 1.01–1.68),<br />

P = 0.04, respectively. Stronger associations were observed for<br />

TNBC. Furthermore, cases in the highest quintile <strong>of</strong> percent<br />

African ancestry were three times more likely to have TNBC than<br />

ER+/PR+ cancer.<br />

Conclusions: These findings provide the first confirmation <strong>of</strong><br />

the TNBC SNP rs8170 in an African ancestry population, and<br />

independent confirmation <strong>of</strong> the TERT ER− SNP. Furthermore,<br />

the risk <strong>of</strong> developing ER− breast cancer, particularly TNBC,<br />

increased with increasing proportion <strong>of</strong> global African ancestry.<br />

Impact: The findings illustrate the importance <strong>of</strong> genetic factors<br />

in the disproportionately high occurrence <strong>of</strong> TNBC in African<br />

American women. Cancer Epidemiol Biomarkers Prev; 22(1); 127–<br />

34. ©2012 <strong>AACR</strong>.<br />

Cancer Epidemiol Biomarkers Prev January 2013 22:127–34;<br />

Published OnlineFirst November 7, 2012; doi:10.1158/1055-9965.<br />

EPI-12-0769<br />

Research Article<br />

Drinking Water Arsenic in Northern Chile: High Cancer<br />

Risks 40 Years after Exposure Cessation<br />

Craig M. Steinmaus, Catterina Ferreccio, Johanna Acevedo Romo, Yan Yuan, Sandra Cortes, Guillermo Marshall,<br />

Lee E. Moore, John R. Balmes, Jane Liaw, Todd Golden, and Allan H. Smith<br />

Craig M. Steinmaus<br />

Abstract<br />

Background: Millions <strong>of</strong> people worldwide are exposed to<br />

arsenic-contaminated water. In the largest city in northern Chile<br />

(Ant<strong>of</strong>agasta), more than 250,000 people were exposed to high<br />

arsenic drinking water concentrations from 1958 until 1970 when a<br />

water treatment plant was installed. Because <strong>of</strong> its unique geology,<br />

limited water sources, and good historical records, lifetime exposure<br />

and long-term latency patterns can be assessed in this area with<br />

better accuracy than in other arsenic-exposed areas worldwide.<br />

Methods: We conducted a population-based case–control study<br />

in northern Chile from October 2007 to December 2010 involving<br />

232 bladder and 306 lung cancer cases and 640 age- and gendermatched<br />

controls, with detailed information on past exposure<br />

and potential confounders, including smoking and occupation.<br />

Results: Bladder cancer ORs for quartiles <strong>of</strong> average arsenic<br />

concentrations in water before 1971 (335 μg/L) were 1.00, 1.36 [95% confidence interval (CI),<br />

0.78–2.37], 3.87 (2.25–6.64), and 6.50 (3.69–11.43), respectively.<br />

Corresponding lung cancer ORs were 1.00, 1.27 (0.81–1.98), 2.00<br />

(1.24–3.24), and 4.32 (2.60–7.17). Bladder and lung cancer ORs in<br />

those highly exposed in Ant<strong>of</strong>agasta during 1958 to 1970 but not<br />

thereafter were 6.88 (3.84–12.32) and 4.35 (2.57–7.36), respectively.<br />

Conclusions: The lung and bladder cancer risks that we found<br />

up to 40 years after high exposures have ended are very high.<br />

Impact: Our findings suggest that prevention, treatment, and<br />

other mortality reduction efforts in arsenic-exposed countries<br />

will be needed for decades after exposure cessation. Cancer<br />

Epidemiol Biomarkers Prev; 22(4); 623–30. ©2013 <strong>AACR</strong>.<br />

Cancer Epidemiol Biomarkers Prev April 2013 22:623–30; Published<br />

OnlineFirst January 25, 2013; doi:10.1158/1055-9965.EPI-12-1190<br />

6<br />

aacrjournals.org


In collaboration with the<br />

Cancer<br />

Immunology<br />

Research<br />

Cancer Immunology Research<br />

Cancer<br />

Immunology<br />

Research<br />

July 2013 • Volume 1 • Number 1 • Pages 1–76<br />

Illuminating the Interplay <strong>of</strong><br />

Cancer and the Immune System<br />

www.aacrjournals.org<br />

Cancer Research Institute<br />

Glenn Dran<strong>of</strong>f, MD<br />

Editor-in-Chief<br />

Scope<br />

Cancer Immunology Research publishes outstanding original articles<br />

reporting major advances in cancer immunology that span the discipline<br />

from basic investigations in host-tumor interactions to developmental<br />

therapeutics in model systems, early translational studies in patients,<br />

and late-stage clinical trials. The journal disseminates knowledge <strong>of</strong><br />

immunology to the cancer research community, catalyzing crossdisciplinary<br />

work that yields a deeper understanding <strong>of</strong> the host-tumor<br />

relationship, more potent cancer treatments, and improved clinical<br />

outcomes.<br />

Review Article<br />

Getting Personal with Neoantigen-Based<br />

Therapeutic Cancer Vaccines<br />

Nir Hacohen, Edward F. Fritsch, Todd A. Carter, Eric S. Lander, and Catherine J. Wu<br />

Abstract<br />

Despite years <strong>of</strong> preclinical efforts and hundreds <strong>of</strong> clinical<br />

studies, therapeutic cancer vaccines with the routine ability<br />

to limit or eliminate tumor growth in humans have been<br />

elusive. With advances in genome sequencing, it is now<br />

possible to identify a new class <strong>of</strong> tumor-specific antigens<br />

derived from mutated proteins that are present only in the<br />

tumor. These “neoantigens” should provide highly specific<br />

targets for antitumor immunity. Although many challenges<br />

Catherine J. Wu<br />

remain in producing and testing neoantigen-based vaccines<br />

customized for each patient, a neoantigen vaccine <strong>of</strong>fers a<br />

promising new approach to induce highly focused antitumor<br />

T cells aimed at eradicating cancer cells. Cancer Immunol Res;<br />

1(1); 11–15. 2013 <strong>AACR</strong>.<br />

Cancer Immunol Res July 2013; 1:11–15; Published OnlineFirst April<br />

7, 2013; doi: 10.1158/2326-6066.CIR-13-0022<br />

The <strong>Best</strong> <strong>of</strong> the <strong>AACR</strong> <strong>Journals</strong> 7


Cancer Immunology Research<br />

Research Article<br />

Anti-CTLA-4 Antibodies <strong>of</strong> IgG2a Isotype Enhance Antitumor<br />

Activity through Reduction <strong>of</strong> Intratumoral Regulatory T Cells<br />

Mark J. Selby, John J. Engelhardt, Michael Quigley, Karla A. Henning, Timothy Chen, Mohan Srinivasan, and<br />

Alan J. Korman<br />

Alan J. Korman<br />

Abstract<br />

Antitumor activity <strong>of</strong> CTLA-4 antibody blockade is thought<br />

to be mediated by interfering with the negative regulation <strong>of</strong><br />

T-effector cell (Teff) function resulting from CTLA-4 engagement<br />

by B7-ligands. In addition, a role for CTLA-4 on regulatory T<br />

cells (Treg), wherein CTLA-4 loss or inhibition results in reduced<br />

Treg function, may also contribute to antitumor responses<br />

by anti-CTLA-4 treatment. We have examined the role <strong>of</strong> the<br />

immunoglobulin constant region on the antitumor activity <strong>of</strong><br />

anti-CTLA-4 to analyze in greater detail the mechanism <strong>of</strong> action<br />

<strong>of</strong> anti-CTLA-4 antibodies. Anti-CTLA-4 antibody containing<br />

the murine immunoglobulin G (IgG)2a constant region exhibits<br />

enhanced antitumor activity in subcutaneous established MC38<br />

and CT26 colon adenocarcinoma tumor models compared<br />

with anti-CTLA-4 containing the IgG2b constant region.<br />

Interestingly, anti-CTLA-4 antibodies containing mouse IgG1<br />

or a mutated mouse IgG1-D265A, which eliminates binding to<br />

all Fcγ receptors (FcγR), do not show antitumor activity in these<br />

models. Assessment <strong>of</strong> Teff and Treg populations at the tumor<br />

and in the periphery showed that anti-CTLA-4-IgG2a mediated<br />

a rapid and dramatic reduction <strong>of</strong> Tregs at the tumor site,<br />

whereas treatment with each <strong>of</strong> the isotypes expanded Tregs in<br />

the periphery. Expansion <strong>of</strong> CD8 + Teffs is observed with both the<br />

IgG2a and IgG2b anti-CTLA-4 isotypes, resulting in a superior<br />

Teff to Treg ratio for the IgG2a isotype. These data suggest that<br />

anti-CTLA-4 promotes antitumor activity by a selective reduction<br />

<strong>of</strong> intratumoral Tregs along with concomitant activation <strong>of</strong> Teffs.<br />

Cancer Immunol Res; 1(1); 32–42. ©2013 <strong>AACR</strong>.<br />

Cancer Immunol Res July 2013; 1:32–42; Published OnlineFirst April<br />

7, 2013; doi: 10.1158/2326-6066.CIR-13-0013<br />

Research Article<br />

An Abscopal Response to Radiation and Ipilimumab in a<br />

Patient with Metastatic Non–Small Cell Lung Cancer<br />

Encouse B. Golden, Sandra Demaria, Peter B. Schiff, Abraham Chachoua, and Silvia C. Formenti<br />

Abstract<br />

A posteriori evidence suggests that radiotherapy to a targeted<br />

tumor can elicit an immune-mediated abscopal (ab-scopus,<br />

away from the target) effect in nontargeted tumors, when<br />

combined with an anti-CTL antigen-4 (CTLA-4) monoclonal<br />

antibody. Concurrent radiotherapy and CTLA-4 blockade<br />

induced immune-mediated abscopal effects in poorly<br />

immunogenic preclinical tumor models and patients with<br />

metastatic melanoma. However, no such reports exist for<br />

patients with metastatic lung adenocarcinoma. We report the<br />

first abscopal response in a treatment-refractory lung cancer<br />

Silvia C. Formenti<br />

patient treated with radiotherapy and ipilimumab (a human<br />

anti-CTLA-4 monoclonal antibody). A posttreatment increase in<br />

tumor-infiltrating cytotoxic lymphocytes, tumor regression, and<br />

normalization <strong>of</strong> tumor markers was observed. One year after<br />

treatment with concurrent radiotherapy and ipilimumab, the<br />

patient is without evidence <strong>of</strong> disease. Cancer Immunol Res; 1(6);<br />

365–72. ©2013 <strong>AACR</strong>.<br />

Cancer Immunol Res December 2013; 1:365–72; Published<br />

OnlineFirst October 9, 2013; doi: 10.1158/2326-6066.CIR-13-0115<br />

8<br />

aacrjournals.org


Cancer Immunology Research<br />

Research Article<br />

T Cells Expressing Chimeric Antigen Receptors Can Cause<br />

Anaphylaxis in Humans<br />

Marcela V. Maus, Andrew R. Haas, Gregory L. Beatty, Steven M. Albelda, Bruce L. Levine, Xiaojun Liu, Yangbing Zhao,<br />

Michael Kalos, and Carl H. June<br />

Marcela V. Maus<br />

Abstract<br />

T cells can be redirected to overcome tolerance to cancer by<br />

engineering with integrating vectors to express a chimeric antigen<br />

receptor (CAR). In preclinical models, we have previously shown<br />

that transfection <strong>of</strong> T cells with mRNA coding for a CAR is an<br />

alternative strategy that has antitumor efficacy and the potential<br />

to evaluate the on-target <strong>of</strong>f-tumor toxicity <strong>of</strong> new CAR targets<br />

safely due to transient mRNA CAR expression. Here, we report<br />

the safety observed in four patients treated with autologous T<br />

cells that had been electroporated with mRNA coding for a CAR<br />

derived from a murine antibody to human mesothelin. Because<br />

<strong>of</strong> the transient nature <strong>of</strong> CAR expression on the T cells, subjects<br />

in the clinical study were given repeated infusions <strong>of</strong> the CAR-T<br />

cells to assess their safety. One subject developed anaphylaxis and<br />

cardiac arrest within minutes <strong>of</strong> completing the third infusion.<br />

Although human anti-mouse immunoglobulin (Ig)G antibodies<br />

have been known to develop with CAR-transduced T cells, they<br />

have been thought to have no adverse clinical consequences. This<br />

is the first description <strong>of</strong> clinical anaphylaxis resulting from CARmodified<br />

T cells, most likely through IgE antibodies specific to the<br />

CAR. These results indicate that the potential immunogenicity <strong>of</strong><br />

CARs derived from murine antibodies may be a safety issue for<br />

mRNA CARs, especially when administered using an intermittent<br />

dosing schedule. Cancer Immunol Res; 1(1); 26–31. ©2013 <strong>AACR</strong>.<br />

Cancer Immunol Res July 2013; 1:26–31; Published OnlineFirst April<br />

7, 2013; doi: 10.1158/2326-6066.CIR-13-0006<br />

Research Article<br />

PD-L1 Expression in the Merkel Cell Carcinoma<br />

Microenvironment: Association with Inflammation,<br />

Merkel Cell Polyomavirus, and Overall Survival<br />

Evan J. Lipson, Jeremy G. Vincent, Myriam Loyo, Luciane T. Kagohara, Brandon S. Luber, Hao Wang,<br />

Haiying Xu, Suresh K. Nayar, Timothy S. Wang, David Sidransky, Robert A. Anders, Suzanne L. Topalian,<br />

and Janis M. Taube<br />

Suzanne L. Topalian<br />

Janis M. Taube<br />

Abstract<br />

Merkel cell carcinoma (MCC) is a lethal, virus-associated cancer<br />

that lacks effective therapies for advanced disease. Agents<br />

blocking the PD-1/PD-L1 pathway have shown objective, durable<br />

tumor regressions in patients with advanced solid malignancies<br />

and efficacy has been linked to PD-L1 expression in the tumor<br />

microenvironment. To investigate whether MCC might be a<br />

target for PD-1/PD-L1 blockade, we examined MCC PD-L1<br />

expression, its association with tumor-infiltrating lymphocytes<br />

(TIL), Merkel cell polyomavirus (MCPyV), and overall survival.<br />

Sixty-seven MCC specimens from 49 patients were assessed with<br />

immunohistochemistry for PD-L1 expression by tumor cells and<br />

TILs, and immune infiltrates were characterized phenotypically.<br />

Tumor cell and TIL PD-L1 expression were observed in 49%<br />

and 55% <strong>of</strong> patients, respectively. In specimens with PD-L1(+)<br />

tumor cells, 97% (28/29) showed a geographic association with<br />

immune infiltrates. Among specimens with moderate-severe TIL<br />

intensities, 100% (29/29) showed PD-L1 expression by tumor cells.<br />

Significant associations were also observed between the presence<br />

<strong>of</strong> MCPyV DNA, a brisk inflammatory response, and tumor cell<br />

PD-L1 expression: MCPyV(-) tumor cells were uniformly PD-<br />

L1(-). Taken together, these findings suggest that a local tumorspecific<br />

and potentially MCPyV-specific immune response<br />

drives tumor PD-L1 expression, similar to previous observations<br />

in melanoma and head and neck squamous cell carcinomas.<br />

In multivariate analyses, PD-L1(-) MCCs were independently<br />

associated with worse overall survival [HR 3.12; 95% confidence<br />

interval, 1.28–7.61; P = 0.012]. These findings suggest that an<br />

endogenous immune response promotes PD-L1 expression in the<br />

MCC microenvironment when MCPyV is present, and provide<br />

a rationale for investigating therapies blocking PD-1/PD-L1 for<br />

patients with MCC. Cancer Immunol Res; 1(1); 54–63. ©2013 <strong>AACR</strong>.<br />

Cancer Immunol Res July 2013; 1:54–63; Published OnlineFirst May<br />

21, 2013; doi: 10.1158/2326-6066.CIR-13-0034<br />

The <strong>Best</strong> <strong>of</strong> the <strong>AACR</strong> <strong>Journals</strong> 9


Cancer<br />

Prevention<br />

Research<br />

Cancer Prevention Research<br />

The Forefront <strong>of</strong> Prevention Science<br />

www.aacrjournals.org<br />

Cancer<br />

Prevention<br />

Research<br />

November 2013 • Volume 6 • Number 11 • Pages 1151–1250<br />

Scott M. Lippman, MD<br />

Editor-in-Chief<br />

Scope<br />

Cancer Prevention Research is devoted exclusively to cancer prevention. The<br />

journal publishes important original studies, reviews, and commentaries<br />

within the major topic areas <strong>of</strong> biology <strong>of</strong> premalignancy, risk factors<br />

and risk assessment, early detection research, immunoprevention and<br />

chemopreventive and other interventions, including the basic science<br />

behind them. The journal includes preclinical, clinical and translational<br />

research, with special attention given to molecular discoveries and<br />

an emphasis on building a translational bridge between the basic and<br />

clinical sciences.<br />

Review Article<br />

New Perspectives <strong>of</strong> Curcumin in Cancer Prevention<br />

Wungki Park, A.R.M. Ruhul Amin, Zhuo Georgia Chen, and Dong M. Shin<br />

Dong M. Shin<br />

Abstract<br />

Numerous natural compounds have been extensively investigated<br />

for their potential for cancer prevention over the decades.<br />

Curcumin, from Curcuma longa, is a highly promising<br />

natural compound that can be potentially used for chemoprevention<br />

<strong>of</strong> multiple cancers. Curcumin modulates multiple<br />

molecular pathways involved in the lengthy carcinogenesis<br />

process to exert its chemopreventive effects through several<br />

mechanisms: promoting apoptosis, inhibiting survival<br />

signals, scavenging reactive oxidative species (ROS), and reducing<br />

the inflammatory cancer microenvironment. Curcumin<br />

fulfills the characteristics for an ideal chemopreventive<br />

agent with its low toxicity, affordability, and easy accessibility.<br />

Nonetheless, the clinical application <strong>of</strong> curcumin is currently<br />

compromised by its poor bioavailability. Here, we review the<br />

potential <strong>of</strong> curcumin in cancer prevention, its molecular targets,<br />

and mechanisms <strong>of</strong> action. Finally, we suggest specific<br />

recommendations to improve its efficacy and bioavailability<br />

for clinical applications. Cancer Prev Res; 6(5); 387–400. ©2013<br />

<strong>AACR</strong>.<br />

Cancer Prev Res May 2013 6:387–400; Published OnlineFirst March<br />

6, 2013; doi:10.1158/1940-6207.CAPR-12-0410<br />

10<br />

aacrjournals.org


Cancer Prevention Research<br />

Research Article<br />

Durable Antibody Responses Following One Dose <strong>of</strong> the Bivalent<br />

Human Papillomavirus L1 Virus-Like Particle Vaccine in the Costa<br />

Rica Vaccine Trial<br />

Mahboobeh Safaeian, Carolina Porras, Yuanji Pan, Aimee Kreimer, John T. Schiller, Paula Gonzalez, Douglas R. Lowy,<br />

Sholom Wacholder, Mark Schiffman, Ana C. Rodriguez, Rolando Herrero, Troy Kemp, Gloriana Shelton, Wim Quint,<br />

Leen-Jan van Doorn, Allan Hildesheim, and Ligia A. Pinto, for the CVT Group<br />

Mahboobeh Safaeian<br />

Abstract<br />

The Costa Rica HPV16/18 Vaccine Trial (CVT) showed that fouryear<br />

vaccine efficacy against 12-month HPV16/18 persistent<br />

infection was similarly high among women who received one,<br />

two, or the recommended three doses <strong>of</strong> the bivalent HPV16/18 L1<br />

virus-like particle (VLP) vaccine. Live-attenuated viral vaccines,<br />

but not simple-subunit vaccines, usually induce durable lifelong<br />

antibody responses after a single dose. It is unclear whether<br />

noninfectious VLP vaccines behave more like live-virus or simplesubunit<br />

vaccines in this regard. To explore the likelihood that<br />

efficacy will persist longer term, we investigated the magnitude<br />

and durability <strong>of</strong> antibodies to this vaccine by measuring HPV16-<br />

and HPV18-specific antibodies by VLP-ELISA using serum from<br />

enrollment, vaccination, and annual visits through four years in<br />

four vaccinated groups; one-dose (n = 78), two-doses separated<br />

by one month (n = 140), two doses separated by six months<br />

(n = 52), and three scheduled doses (n = 120, randomly selected).<br />

We also tested enrollment sera from n = 113 HPV16- or HPV18<br />

L1-seropositive women prevaccination, presumably from natural<br />

infection. At four years, 100% <strong>of</strong> women in all groups remained<br />

HPV16/18 seropositive; both HPV16/18 geometric mean titers<br />

(GMT) among the extended two-dose group were non-inferior<br />

to the three-dose group, and ELISA titers were highly correlated<br />

with neutralization titers in all groups. Compared with the natural<br />

infection group, HPV16/18 GMTs were, respectively, at least 24<br />

and 14 times higher among the two-dose and 9 and 5 times higher<br />

among one-dose vaccinees. Antibody levels following one-dose<br />

remained stable from month 6 through month 48. Results raise the<br />

possibility that even a single dose <strong>of</strong> HPV VLPs will induce longterm<br />

protection. Cancer Prev Res; 6(11); 1242–50. ©2013 <strong>AACR</strong>.<br />

Cancer Prev Res November 2013 6:1242–1250; doi:10.1158/1940–6207.<br />

CAPR-13-0203<br />

Research Article<br />

MicroRNA Expression Signatures during Malignant Progression<br />

from Barrett’s Esophagus to Esophageal Adenocarcinoma<br />

Xifeng Wu, Jaffer A. Ajani, Jian Gu, David W. Chang, Weiqi Tan, Michelle A.T. Hildebrandt, Maosheng Huang,<br />

Kenneth K. Wang, and Ernest Hawk<br />

Xifeng Wu<br />

Abstract<br />

Barrett’s esophagus is the precursor lesion <strong>of</strong> esophageal<br />

adenocarcinoma, whose progression follows sequential stages. However,<br />

the low progression rate and the inadequacy and subjective<br />

interpretation <strong>of</strong> histologic grading in predicting Barrett’s esophagus<br />

progression call for more objective biomarkers that can improve risk<br />

prediction. We conducted a genome-wide pr<strong>of</strong>iling <strong>of</strong> 754 human<br />

microRNAs (miRNA) in 35 normal epithelium, 34 Barrett’s esophagus,<br />

and 36 esophageal adenocarcinoma tissues using TaqMan real-time<br />

PCR-based pr<strong>of</strong>iling. Unsupervised hierarchical clustering using 294<br />

modestly to highly expressed miRNAs showed clear clustering <strong>of</strong> two<br />

groups: normal epithelium versus Barrett’s esophagus/esophageal<br />

adenocarcinoma tissues. Moreover, there was an excellent clustering<br />

<strong>of</strong> Barrett’s metaplasia (without dysplasia) tissues from normal<br />

epithelium tissues. However, Barrett’s esophagus tissues <strong>of</strong> different<br />

stages and esophageal adenocarcinoma tissues were interspersed.<br />

There were differentially expressed miRNAs at different stages. The<br />

majority <strong>of</strong> miRNA aberrations involved upregulation <strong>of</strong> expression<br />

in Barrett’s esophagus and esophageal adenocarcinoma tissues,<br />

with the most dramatic alterations occurring at the Barrett’s<br />

metaplasia stage. Known oncomiRs, such as miR-21, miR-25, and<br />

miR-223, and tumor suppressor miRNAs, including miR-205, miR-<br />

203, let-7c, and miR-133a, showed progressively altered expression<br />

from Barrett’s esophagus to esophageal adenocarcinoma. We also<br />

identified a number <strong>of</strong> novel miRNAs that showed progressively<br />

altered expression, including miR-301b, miR-618, and miR-23b. The<br />

significant miRNA alterations that were exclusive to esophageal<br />

adenocarcinoma but not Barrett’s esophagus included miR-375<br />

downregulation and upregulation <strong>of</strong> five members <strong>of</strong> the miR-<br />

17-92 and its homologue clusters, which may become promising<br />

biomarkers for esophageal adenocarcinoma development. Cancer<br />

Prev Res; 6(3); 196–205. ©2013 <strong>AACR</strong>.<br />

Cancer Prev Res March 2013 6:196–205; doi:10.1158/1940-6207.<br />

CAPR-12-0276<br />

The <strong>Best</strong> <strong>of</strong> the <strong>AACR</strong> <strong>Journals</strong> 11


Cancer Prevention Research<br />

Research Article<br />

MUC1 Vaccine for Individuals with Advanced Adenoma <strong>of</strong><br />

the Colon: A Cancer Immunoprevention Feasibility Study<br />

Takashi Kimura, John R. McKolanis, Lynda A. Dzubinski, Kazi Islam, Douglas M. Potter, Andres M. Salazar,<br />

Robert E. Schoen, and Olivera J. Finn<br />

Robert E. Schoen<br />

Olivera J. Finn<br />

Abstract<br />

Cancer vaccines based on human tumor-associated antigens<br />

(TAA) have been tested in patients with advanced or recurrent<br />

cancer, in combination with or following standard therapy. Their<br />

immunogenicity and therapeutic efficacy has been difficult to<br />

properly evaluate in that setting characterized by multiple highly<br />

suppressive effects <strong>of</strong> the tumor and the standard therapy on the<br />

patient’s immune system. In animal models <strong>of</strong> human cancer,<br />

vaccines administered in the prophylactic setting are most<br />

immunogenic and effectively prevent cancer development and<br />

progression. We report results <strong>of</strong> a clinical study that show that<br />

in patients without cancer but with a history <strong>of</strong> premalignant<br />

lesions (advanced colonic adenomas, precursors to colon cancer),<br />

a vaccine based on the TAA MUC1 was highly immunogenic in<br />

17 <strong>of</strong> 39 (43.6%) <strong>of</strong> vaccinated individuals, eliciting high levels <strong>of</strong><br />

anti-MUC1 immunoglobulin G (IgG) and long-lasting immune<br />

memory. Lack <strong>of</strong> response in 22 <strong>of</strong> 39 individuals was correlated<br />

with high levels <strong>of</strong> circulating myeloid-derived suppressor<br />

cells (MDSC) prevaccination. Vaccine-elicited MUC1-specific<br />

immune response and immune memory were not associated<br />

with significant toxicity. Our study shows that vaccines based on<br />

human TAAs are immunogenic and safe and capable <strong>of</strong> eliciting<br />

long-term memory that is important for cancer prevention. We<br />

also show that in the premalignant setting, immunosuppressive<br />

environment (e.g., high levels <strong>of</strong> MDSC) might already exist in<br />

some individuals, suggesting an even earlier premalignant stage or<br />

preselection <strong>of</strong> nonimmunosuppressed patients for prophylactic<br />

vaccination. Cancer Prev Res; 6(1); 18–26. ©2012 <strong>AACR</strong>.<br />

Cancer Prev Res January 2013 6:18–26; Published OnlineFirst<br />

December 17, 2012; doi:10.1158/1940-6207.CAPR-12-0275<br />

Research Article<br />

Increased Levels <strong>of</strong> Urinary PGE-M, a Biomarker <strong>of</strong><br />

Inflammation, Occur in Association with Obesity, Aging,<br />

and Lung Metastases in Patients with Breast Cancer<br />

Patrick G. Morris, Xi Kathy Zhou, Ginger L. Milne, Daniel Goldstein, Laura C. Hawks, Chau T. Dang,<br />

Shanu Modi, Monica N. Fornier, Clifford A. Hudis, and Andrew J. Dannenberg<br />

Patrick G. Morris<br />

Andrew J. Dannenberg<br />

Abstract<br />

Elevated levels <strong>of</strong> COX-derived prostaglandin E 2 (PGE 2 ) occur<br />

in inflamed tissues. To evaluate the potential links between<br />

inflammation and breast cancer, levels <strong>of</strong> urinary prostaglandin E<br />

metabolite (PGE-M), a stable end metabolite <strong>of</strong> PGE 2 , were quantified.<br />

We enrolled 400 patients with breast cancer: controls with early breast<br />

cancer (n = 200), lung metastases (n = 100), and metastases to other<br />

sites (n = 100). Patients completed a questionnaire, provided urine,<br />

and had measurements <strong>of</strong> height and weight. Urinary PGE-M was<br />

quantified by mass spectrometry. Ever smokers with lung metastasis<br />

who had not been exposed to nonsteroidal anti-inflammatory drugs<br />

(NSAIDs) had the highest PGE-M levels. PGE-M levels were increased<br />

in association with elevated body mass index (BMI; P < 0.001), aging<br />

(P < 0.001), pack-year smoking history (P = 0.02), lung metastases<br />

(P = 0.02), and recent cytotoxic chemotherapy (P = 0.03). Conversely,<br />

use <strong>of</strong> NSAIDs, prototypic inhibitors <strong>of</strong> COX activity, was associated<br />

with reduced PGE-M levels (P < 0.001). On the basis <strong>of</strong> the current<br />

findings, PGE-M is likely to be a useful biomarker for the selection <strong>of</strong><br />

high-risk subgroups to determine the use <strong>of</strong> interventions that aim to<br />

reduce inflammation and possibly the development and progression<br />

<strong>of</strong> breast cancer, especially in overweight and obese women. Cancer<br />

Prev Res; 6(5); 428–36. ©2013 <strong>AACR</strong>.<br />

Cancer Prev Res May 2013 6:428–436; Published OnlineFirst March<br />

26, 2013; doi:10.1158/1940-6207.CAPR-12-0431<br />

12<br />

aacrjournals.org


Cancer<br />

Research<br />

Cancer Research<br />

George C. C. Prendergast, PhD<br />

Editor-in-Chief<br />

Scope<br />

Cancer Research is the most frequently cited cancer journal in the<br />

world. The journal publishes original studies, reviews, and opinion<br />

pieces <strong>of</strong>fering significance and broad impact to a diverse audience<br />

spanning basic, preclinical, clinical, prevention, and epidemiologic<br />

research. Cancer Research seeks manuscripts that <strong>of</strong>fer pathobiological<br />

and translational impact to inform the personal, clinical, and societal<br />

problems posed by cancer. The main scope <strong>of</strong> the journal is captured<br />

in its primary subsections, which focus on molecular and cellular<br />

pathobiology, tumor and stem cell biology, therapeutics and targets,<br />

microenvironment and immunology, prevention and epidemiology, and<br />

integrated systems and technology.<br />

Review Article<br />

Genome-Wide Epigenetic Regulation <strong>of</strong> miRNAs in Cancer<br />

Constance Baer, Rainer Claus, and Christoph Plass<br />

Abstract<br />

Aberrant microRNA (miRNA) expression contributes to tumorigenesis<br />

and cancer progression. Although the number <strong>of</strong> reported<br />

deregulated miRNAs in various cancer types is growing fast, the<br />

underlying mechanisms <strong>of</strong> aberrant miRNA regulation are still<br />

poorly studied. Epigenetic alterations including aberrant DNA<br />

methylation deregulate miRNA expression, which was first shown by<br />

reexpression <strong>of</strong> miRNAs upon pharmacologic DNA demethylation.<br />

However, studying the influence <strong>of</strong> DNA methylation on miRNA<br />

transcription on a genome-wide level was hampered by poor miRNA<br />

promoter annotation. Putative miRNA promoters were identified<br />

on a genome-wide level by using common promoter surrogate<br />

markers (e.g., histone modifications) and were later validated as<br />

such in different tumor entities. Integrating promoter datasets and<br />

Christoph Plass<br />

global DNA methylation analysis revealed an extensive influence<br />

<strong>of</strong> DNA hyper- as well as hypomethylation on miRNA regulation.<br />

In this review, we summarize the current knowledge <strong>of</strong> the field<br />

and discuss recent efforts to map miRNA promoter sequences<br />

and to determine the contribution <strong>of</strong> epigenetic mechanisms<br />

to the regulation <strong>of</strong> miRNA expression. We discuss examples <strong>of</strong><br />

tumor suppressive and oncogenic miRNAs such as the miR-34 and<br />

miR-21 family, respectively, which highlight the complexity and<br />

consequences <strong>of</strong> epigenetic miRNA deregulation. Cancer Res; 73(2);<br />

473–7. ©2012 <strong>AACR</strong>.<br />

Cancer Res January 15, 2013 73:473–7; Published OnlineFirst<br />

January 10, 2013; doi: 10.1158/0008-5472.CAN-12-3731<br />

The <strong>Best</strong> <strong>of</strong> the <strong>AACR</strong> <strong>Journals</strong> 13


Cancer Research<br />

Research Article<br />

The Noncoding RNA MALAT1 Is a Critical Regulator <strong>of</strong> the<br />

Metastasis Phenotype <strong>of</strong> Lung Cancer Cells<br />

Tony Gutschner, Monika Hämmerle, Moritz Eißmann, Jeff Hsu, Youngsoo Kim, Gene Hung, Alexey Revenko,<br />

Gayatri Arun, Marion Stentrup, Matthias Groß, Martin Zörnig, A. Robert MacLeod, David L. Spector,<br />

and Sven Diederichs<br />

Sven Diederichs<br />

Abstract<br />

The long noncoding RNA MALAT1 (metastasis-associated<br />

lung adenocarcinoma transcript 1), also known as MALAT-1<br />

or NEAT2 (nuclear-enriched abundant transcript 2), is a highly<br />

conserved nuclear noncoding RNA (ncRNA) and a predictive<br />

marker for metastasis development in lung cancer. To uncover<br />

its functional importance, we developed a MALAT1 knockout<br />

model in human lung tumor cells by genomically integrating<br />

RNA destabilizing elements using zinc finger nucleases. The<br />

achieved 1,000-fold MALAT1 silencing provides a unique loss<strong>of</strong>-function<br />

model. Proposed mechanisms <strong>of</strong> action include<br />

regulation <strong>of</strong> splicing or gene expression. In lung cancer,<br />

MALAT1 does not alter alternative splicing but actively regulates<br />

gene expression including a set <strong>of</strong> metastasis-associated genes.<br />

Consequently, MALAT1-deficient cells are impaired in migration<br />

and form fewer tumor nodules in a mouse xenograft. Antisense<br />

oligonucleotides (ASO) blocking MALAT1 prevent metastasis<br />

formation after tumor implantation. Thus, targeting MALAT1<br />

with ASOs provides a potential therapeutic approach to<br />

prevent lung cancer metastasis with this ncRNA serving as both<br />

predictive marker and therapeutic target. Finally, regulating<br />

gene expression, but not alternative splicing, is the critical<br />

function <strong>of</strong> MALAT1 in lung cancer metastasis. In summary, 10<br />

years after the discovery <strong>of</strong> the lncRNA MALAT1 as a biomarker<br />

for lung cancer metastasis, our loss-<strong>of</strong>-function model unravels<br />

the active function <strong>of</strong> MALAT1 as a regulator <strong>of</strong> gene expression<br />

governing hallmarks <strong>of</strong> lung cancer metastasis. Cancer Res;<br />

73(3); 1180–9. ©2012 <strong>AACR</strong>.<br />

Cancer Res February 1, 2013 73:1180–9; Published OnlineFirst<br />

December 14, 2012; doi: 10.1158/0008-5472.CAN-12-2850<br />

Research Article<br />

Androgen Receptor Splice Variants Mediate Enzalutamide<br />

Resistance in Castration-Resistant Prostate Cancer Cell Lines<br />

Yingming Li, Siu Chiu Chan, Lucas J. Brand, Tae Hyun Hwang, Kevin A.T. Silverstein, and Scott M. Dehm<br />

Scott M. Dehm<br />

Abstract<br />

Persistent androgen receptor (AR) transcriptional activity underlies<br />

resistance to AR-targeted therapy and progression to lethal<br />

castration-resistant prostate cancer (CRPC). Recent success in<br />

retargeting persistent AR activity with next generation androgen/<br />

AR axis inhibitors such as enzalutamide (MDV3100) has validated<br />

AR as a master regulator during all stages <strong>of</strong> disease progression.<br />

However, resistance to next generation AR inhibitors limits<br />

therapeutic efficacy for many patients. One emerging mechanism <strong>of</strong><br />

CRPC progression is AR gene rearrangement, promoting synthesis<br />

<strong>of</strong> constitutively active truncated AR splice variants (AR-V) that<br />

lack the AR ligand-binding domain. In this study, we show that cells<br />

with AR gene rearrangements expressing both full-length and AR-<br />

Vs are androgen independent and enzalutamide resistant. However,<br />

selective knock-down <strong>of</strong> AR-V expression inhibited androgenindependent<br />

growth and restored responsiveness to androgens<br />

and antiandrogens. In heterogeneous cell populations, AR gene<br />

rearrangements marked individual AR-V–dependent cells that<br />

were resistant to enzalutamide. Gene expression pr<strong>of</strong>iling following<br />

knock-down <strong>of</strong> full-length AR or AR-Vs showed that AR-Vs drive<br />

resistance to AR-targeted therapy by functioning as constitutive<br />

and independent effectors <strong>of</strong> the androgen/AR transcriptional<br />

program. Further, mitotic genes deemed previously to be unique<br />

AR-V targets were found to be biphasic targets associated with<br />

a proliferative level <strong>of</strong> signaling output from either AR-Vs or<br />

androgen-stimulated AR. Overall, these studies highlight AR-Vs<br />

as key mediators <strong>of</strong> persistent AR signaling and resistance to the<br />

current arsenal <strong>of</strong> conventional and next generation AR-directed<br />

therapies, advancing the concept <strong>of</strong> AR-Vs as therapeutic targets in<br />

advanced disease. Cancer Res; 73(2); 483–9. ©2012 <strong>AACR</strong>.<br />

Cancer Res January 15, 2013 73:483–9; Published OnlineFirst<br />

November 1, 2012; doi: 10.1158/0008-5472.CAN-12-3630<br />

14<br />

aacrjournals.org


Cancer Research<br />

Research Article<br />

Complex Tumor Genomes Inferred from Single<br />

Circulating Tumor Cells by Array-CGH and<br />

Next-Generation Sequencing<br />

Jochen B. Geigl<br />

Ellen Heitzer, Martina Auer, Christin Gasch, Martin Pichler, Peter Ulz, Eva Maria H<strong>of</strong>fmann, Sigurd Lax,<br />

Julie Waldispuehl-Geigl, Oliver Mauermann, Carolin Lackner, Gerald Höfler, Florian Eisner, Heinz Sill,<br />

Hellmut Samonigg, Klaus Pantel, Sabine Riethdorf, Thomas Bauernh<strong>of</strong>er, Jochen B. Geigl, and Michael R. Speicher<br />

Michael R. Speicher<br />

Abstract<br />

Circulating tumor cells (CTC) released into blood from primary<br />

cancers and metastases reflect the current status <strong>of</strong> tumor<br />

genotypes, which are prone to changes. Here, we conducted<br />

the first comprehensive genomic pr<strong>of</strong>iling <strong>of</strong> CTCs using array–<br />

comparative genomic hybridization (CGH) and next-generation<br />

sequencing. We used the U.S. Food and Drug Administration–<br />

cleared CellSearch system, which detected CTCs in 21 <strong>of</strong> 37<br />

patients (range, 1–202/7.5 mL sample) with stage IV colorectal<br />

carcinoma. In total, we were able to isolate 37 intact CTCs from<br />

six patients and identified in those multiple colorectal cancer–<br />

associated copy number changes, many <strong>of</strong> which were also<br />

present in the respective primary tumor. We then used massive<br />

parallel sequencing <strong>of</strong> a panel <strong>of</strong> 68 colorectal cancer–associated<br />

genes to compare the mutation spectrum in the primary tumors,<br />

metastases, and the corresponding CTCs from two <strong>of</strong> these<br />

patients. Mutations in known driver genes [e.g., adenomatous<br />

polyposis coli (APC), KRAS, or PIK3CA] found in the primary<br />

tumor and metastasis were also detected in corresponding<br />

CTCs. However, we also observed mutations exclusively in<br />

CTCs. To address whether these mutations were derived from<br />

a small subclone in the primary tumor or represented new<br />

variants <strong>of</strong> metastatic cells, we conducted additional deep<br />

sequencing <strong>of</strong> the primary tumor and metastasis and applied<br />

a customized statistical algorithm for analysis. We found that<br />

most mutations initially found only in CTCs were also present<br />

at subclonal level in the primary tumors and metastases from<br />

the same patient. This study paves the way to use CTCs as a<br />

liquid biopsy in patients with cancer, providing more effective<br />

options to monitor tumor genomes that are prone to change<br />

during progression, treatment, and relapse. Cancer Res; 73(10);<br />

2965–75. ©2013 <strong>AACR</strong>.<br />

Cancer Res May 15, 2013 73:2965–75; Published OnlineFirst March<br />

7, 2013; doi: 10.1158/0008-5472.CAN-12-4140<br />

Research Article<br />

Targeting Tumor-Infiltrating Macrophages Decreases<br />

Tumor-Initiating Cells, Relieves Immunosuppression,<br />

and Improves Chemotherapeutic Responses<br />

Jonathan B. Mitchem, Donal J. Brennan, Brett L. Knolh<strong>of</strong>f, Brian A. Belt, Yu Zhu, Dominic E. Sanford, Larisa Belaygorod,<br />

Danielle Carpenter, Lynne Collins, David Piwnica-Worms, Stephen Hewitt, Girish Mallya Udupi, William M. Gallagher,<br />

Craig Wegner, Brian L. West, Andrea Wang-Gillam, Peter Goedegebuure, David C. Linehan, and David G. DeNardo<br />

David G. DeNardo<br />

Abstract<br />

Tumor-infiltrating immune cells can promote chemoresistance<br />

and metastatic spread in aggressive tumors. Consequently, the<br />

type and quality <strong>of</strong> immune responses present in the neoplastic<br />

stroma are highly predictive <strong>of</strong> patient outcome in several cancer<br />

types. In addition to host immune responses, intrinsic tumor cell<br />

activities that mimic stem cell properties have been linked to<br />

chemoresistance, metastatic dissemination, and the induction <strong>of</strong><br />

immune suppression. Cancer stem cells are far from a static cell<br />

population; rather, their presence seems to be controlled by highly<br />

dynamic processes that are dependent on cues from the tumor<br />

stroma. However, the impact immune responses have on tumor<br />

stem cell differentiation or expansion is not well understood. In<br />

this study, we show that targeting tumor-infiltrating macrophages<br />

(TAM) and inflammatory monocytes by inhibiting either the<br />

myeloid cell receptors colony-stimulating factor-1 receptor (CSF1R)<br />

or chemokine (C–C motif) receptor 2 (CCR2) decreases the number<br />

<strong>of</strong> tumor-initiating cells (TIC) in pancreatic tumors. Targeting CCR2<br />

or CSF1R improves chemotherapeutic efficacy, inhibits metastasis,<br />

and increases antitumor T-cell responses. Tumor-educated<br />

macrophages also directly enhanced the tumor-initiating capacity<br />

<strong>of</strong> pancreatic tumor cells by activating the transcription factor<br />

STAT3, thereby facilitating macrophage-mediated suppression <strong>of</strong><br />

CD8 + T lymphocytes. Together, our findings show how targeting<br />

TAMs can effectively overcome therapeutic resistance mediated by<br />

TICs. Cancer Res; 73(3); 1128–41. ©2012 <strong>AACR</strong>.<br />

Cancer Res February 1, 2013 73:1128–41; Published OnlineFirst<br />

December 5, 2012; doi: 10.1158/0008-5472.CAN-12-2731<br />

The <strong>Best</strong> <strong>of</strong> the <strong>AACR</strong> <strong>Journals</strong> 15


Clinical<br />

Cancer<br />

Research<br />

Clinical Cancer Research<br />

Kenneth C. Anderson, MD<br />

Editor-in-Chief<br />

Scope<br />

Clinical Cancer Research publishes innovative clinical and translational<br />

cancer research studies that bridge the laboratory and the clinic.<br />

The journal is especially interested in clinical trials evaluating new<br />

treatments, accompanied by research on pharmacology, and molecular<br />

alterations or biomarkers that predict response or resistance to<br />

treatment. The journal also prioritizes laboratory and animal studies<br />

<strong>of</strong> new drugs and molecule-targeted agents with the potential to lead<br />

to clinical trials, and studies <strong>of</strong> targetable mechanisms <strong>of</strong> oncogenesis,<br />

progression <strong>of</strong> the malignant phenotype, and metastatic disease.<br />

Review Article<br />

The Definition <strong>of</strong> Primary and Secondary Glioblastoma<br />

Hiroko Ohgaki and Paul Kleihues<br />

Abstract<br />

Glioblastoma is the most frequent and malignant brain tumor.<br />

The vast majority <strong>of</strong> glioblastomas (~90%) develop rapidly<br />

de novo in elderly patients, without clinical or histologic<br />

evidence <strong>of</strong> a less malignant precursor lesion (primary<br />

glioblastomas). Secondary glioblastomas progress from lowgrade<br />

diffuse astrocytoma or anaplastic astrocytoma. They<br />

manifest in younger patients, have a lesser degree <strong>of</strong> necrosis,<br />

are preferentially located in the frontal lobe, and carry a<br />

significantly better prognosis. Histologically, primary and<br />

secondary glioblastomas are largely indistinguishable, but they<br />

differ in their genetic and epigenetic pr<strong>of</strong>iles. Decisive genetic<br />

signposts <strong>of</strong> secondary glioblastoma are IDH1 mutations, which<br />

are absent in primary glioblastomas and which are associated<br />

with a hypermethylation phenotype. IDH1 mutations are the<br />

earliest detectable genetic alteration in precursor low-grade<br />

Hiroko Ohgaki<br />

diffuse astrocytomas and in oligodendrogliomas, indicating that<br />

these tumors are derived from neural precursor cells that differ<br />

from those <strong>of</strong> primary glioblastomas. In this review, we summarize<br />

epidemiologic, clinical, histopathologic, genetic, and expression<br />

features <strong>of</strong> primary and secondary glioblastomas and the biologic<br />

consequences <strong>of</strong> IDH1 mutations. We conclude that this genetic<br />

alteration is a definitive diagnostic molecular marker <strong>of</strong> secondary<br />

glioblastomas and more reliable and objective than clinical criteria.<br />

Despite a similar histologic appearance, primary and secondary<br />

glioblastomas are distinct tumor entities that originate from<br />

different precursor cells and may require different therapeutic<br />

approaches. Clin Cancer Res; 19(4); 764–72. ©2012 <strong>AACR</strong>.<br />

Clin Cancer Res February 15, 2013 19:4 764–72; Published<br />

OnlineFirst December 3, 2012; doi:10.1158/1078-0432.CCR-12-3002<br />

16<br />

aacrjournals.org


Clinical Cancer Research<br />

Research Article<br />

BRAF Inhibition Is Associated with Enhanced Melanoma Antigen<br />

Expression and a More Favorable Tumor Microenvironment in<br />

Patients with Metastatic Melanoma<br />

Jennifer A. Wargo<br />

Dennie T. Frederick, Adriano Piris, Alexandria P. Cogdill, Zachary A. Cooper, Cecilia Lezcano, Cristina R. Ferrone,<br />

Devarati Mitra, Andrea Boni, Lindsay P. Newton, Chengwen Liu, Weiyi Peng, Ryan J. Sullivan, Donald P. Lawrence,<br />

F. Stephen Hodi, Willem W. Overwijk, Gregory Lizée, George F. Murphy, Patrick Hwu, Keith T. Flaherty, David E. Fisher, and Jennifer A. Wargo<br />

Abstract<br />

Purpose: To evaluate the effects <strong>of</strong> BRAF inhibition on the tumor<br />

microenvironment in patients with metastatic melanoma.<br />

Experimental Design: Thirty-five biopsies were collected<br />

from 16 patients with metastatic melanoma pretreatment (day<br />

0) and at 10 to 14 days after initiation <strong>of</strong> treatment with either<br />

BRAF inhibitor alone (vemurafenib) or BRAF + MEK inhibition<br />

(dabrafenib + trametinib) and were also taken at time <strong>of</strong><br />

progression. Biopsies were analyzed for melanoma antigens,<br />

T-cell markers, and immunomodulatory cytokines.<br />

Results: Treatment with either BRAF inhibitor alone or BRAF + MEK<br />

inhibitor was associated with an increased expression <strong>of</strong> melanoma<br />

antigens and an increase in CD8+ T-cell infiltrate. This was also<br />

associated with a decrease in immunosuppressive cytokines<br />

[interleukin (IL)-6 and IL-8] and an increase in markers <strong>of</strong> T-cell<br />

cytotoxicity. Interestingly, expression <strong>of</strong> exhaustion markers TIM-3<br />

and PD1 and the immunosuppressive ligand PDL1 was increased<br />

on treatment. A decrease in melanoma antigen expression and CD8<br />

T-cell infiltrate was noted at time <strong>of</strong> progression on BRAF inhibitor<br />

alone and was reversed with combined BRAF and MEK inhibition.<br />

Conclusions: Together, these data suggest that treatment<br />

with BRAF inhibition enhances melanoma antigen expression<br />

and facilitates T-cell cytotoxicity and a more favorable tumor<br />

microenvironment, providing support for potential synergy<br />

<strong>of</strong> BRAF-targeted therapy and immunotherapy. Interestingly,<br />

markers <strong>of</strong> T-cell exhaustion and the immunosuppressive ligand<br />

PDL1 are also increased with BRAF inhibition, further implying<br />

that immune checkpoint blockade may be critical in augmenting<br />

responses to BRAF-targeted therapy in patients with melanoma.<br />

Clin Cancer Res; 19(5); 1225–31. ©2013 <strong>AACR</strong>.<br />

Clin Cancer Res March 1, 2013 19:5 1225–31; Published OnlineFirst<br />

January 10, 2013; doi:10.1158/1078-0432.CCR-12-1630<br />

Research Article<br />

Analysis <strong>of</strong> Tumor Specimens at the Time <strong>of</strong> Acquired Resistance to<br />

EGFR-TKI Therapy in 155 Patients with EGFR-Mutant Lung Cancers<br />

Helena A. Yu, Maria E. Arcila, Natasha Rekhtman, Camelia S. Sima, Maureen F. Zakowski, William Pao, Mark G. Kris,<br />

Vincent A. Miller, Marc Ladanyi, and Gregory J. Riely<br />

Gregory J. Riely<br />

Abstract<br />

Purpose: All patients with EGF receptor (EGFR)–mutant lung<br />

cancers eventually develop acquired resistance to EGFR tyrosine<br />

kinase inhibitors (TKI). Smaller series have identified various<br />

mechanisms <strong>of</strong> resistance, but systematic evaluation <strong>of</strong> a large<br />

number <strong>of</strong> patients to definitively establish the frequency <strong>of</strong><br />

various mechanisms has not been conducted.<br />

Experimental Design: Patients with lung adenocarcinomas<br />

and acquired resistance to erlotinib or gefitinib enrolled onto a<br />

prospective biopsy protocol and underwent a rebiopsy after the<br />

development <strong>of</strong> acquired resistance. Histology was reviewed.<br />

Samples underwent genotyping for mutations in EGFR, AKT1, BRAF,<br />

ERBB2, KRAS, MEK1, NRAS and PIK3CA, and FISH for MET and HER2.<br />

Results: Adequate tumor samples for molecular analysis were obtained<br />

in 155 patients. Ninety-eight had second-site EGFR T790M mutations<br />

[63%; 95% confidence interval (CI), 55%–70%] and four had small cell<br />

transformation (3%, 95% CI, 0%–6%). MET amplification was seen in 4<br />

<strong>of</strong> 75 (5%; 95% CI, 1%–13%). HER2 amplification was seen in 3 <strong>of</strong> 24 (13%;<br />

95% CI, 3%–32%). We did not detect any acquired mutations in PIK3CA,<br />

AKT1, BRAF, ERBB2, KRAS, MEK1, or NRAS (0 <strong>of</strong> 88, 0%; 95% CI, 0%–4%).<br />

Overlap among mechanisms <strong>of</strong> acquired resistance was seen in 4%.<br />

Conclusions: This is the largest series reporting mechanisms <strong>of</strong><br />

acquired resistance to EGFR-TKI therapy. We identified EGFR T790M<br />

as the most common mechanism <strong>of</strong> acquired resistance, whereas<br />

MET amplification, HER2 amplification, and small cell histologic<br />

transformation occur less frequently. More comprehensive methods<br />

to characterize molecular alterations in this setting are needed to<br />

improve our understanding <strong>of</strong> acquired resistance to EGFR-TKIs.<br />

Clin Cancer Res; 19(8); 2240–7. ©2013 <strong>AACR</strong>.<br />

Clin Cancer Res April 15, 2013 19:8 2240–7; Published OnlineFirst<br />

March 7, 2013; doi:10.1158/1078-0432.CCR-12-2246<br />

The <strong>Best</strong> <strong>of</strong> the <strong>AACR</strong> <strong>Journals</strong> 17


Clinical Cancer Research<br />

Research Article<br />

Durable Cancer Regression Off-Treatment and Effective<br />

Reinduction Therapy with an Anti-PD-1 Antibody<br />

Evan J. Lipson, William H. Sharfman, Charles G. Drake, Ira Wollner, Janis M. Taube, Robert A. Anders, Haiying Xu,<br />

Sheng Yao, Alice Pons, Lieping Chen, Drew M. Pardoll, Julie R. Brahmer, and Suzanne L. Topalian<br />

Abstract<br />

Purpose: Results from the first-in-human phase I trial <strong>of</strong> the<br />

anti–programmed death-1 (PD-1) antibody BMS-936558 in<br />

patients with treatment-refractory solid tumors, including safety,<br />

tolerability, pharmacodynamics, and immunologic correlates,<br />

have been previously reported. Here, we provide long-term followup<br />

on three patients from that trial who sustained objective tumor<br />

regressions <strong>of</strong>f therapy, and test the hypothesis that reinduction<br />

therapy for late tumor recurrence can be effective.<br />

Experimental Design: Three patients with colorectal cancer, renal<br />

cell cancer, and melanoma achieved objective responses on an<br />

intermittent dosing regimen <strong>of</strong> BMS-936558. Following cessation<br />

<strong>of</strong> therapy, patients were followed for more than 3 years. A patient<br />

with melanoma who experienced a prolonged partial regression<br />

followed by tumor recurrence received reinduction therapy.<br />

Suzanne L. Topalian<br />

renal cell cancer experienced a partial response lasting 3 years<br />

<strong>of</strong>f therapy, which converted to a complete response, which<br />

is ongoing at 12 months. A patient with melanoma achieved<br />

a partial response that was stable for 16 months <strong>of</strong>f therapy;<br />

recurrent disease was successfully treated with reinduction<br />

anti-PD-1 therapy.<br />

Conclusion: These data represent the most prolonged<br />

observation to date <strong>of</strong> patients with solid tumors responding<br />

to anti-PD-1 immunotherapy and the first report <strong>of</strong> successful<br />

reinduction therapy following delayed tumor progression.<br />

They underscore the potential for immune checkpoint<br />

blockade with anti-PD-1 to reset the equilibrium between<br />

tumor and the host immune system. Clin Cancer Res; 19(2);<br />

462–8. ©2012 <strong>AACR</strong>.<br />

Results: A patient with colorectal cancer experienced a<br />

complete response, which is ongoing after 3 years. A patient with<br />

Clin Cancer Res January 15, 2013 19:2 462–8; Published OnlineFirst<br />

November 20, 2012; doi:10.1158/1078-0432.CCR-12-2625<br />

Research Article<br />

An Epithelial–Mesenchymal Transition Gene Signature<br />

Predicts Resistance to EGFR and PI3K Inhibitors and<br />

Identifies Axl as a Therapeutic Target for Overcoming<br />

EGFR Inhibitor Resistance<br />

Lauren Averett Byers<br />

John V. Heymach<br />

Lauren Averett Byers, Lixia Diao, Jing Wang, Pierre Saintigny, Luc Girard, Michael Peyton, Li Shen, Youhong Fan, Uma Giri,<br />

Praveen K. Tumula, Monique B. Nilsson, Jayanthi Gudikote, Hai Tran, Robert J.G. Cardnell, David J. Bearss, Steven L. Warner,<br />

Jason M. Foulks, Steven B. Kanner, Varsha Gandhi, Nancy Krett, Steven T. Rosen, Edward S. Kim, Roy S. Herbst, George R. Blumenschein,<br />

J. Jack Lee, Scott M. Lippman, K. Kian Ang, Gordon B. Mills, Waun K. Hong, John N. Weinstein, Ignacio I. Wistuba, Kevin R. Coombes,<br />

John D. Minna, and John V. Heymach<br />

Abstract<br />

Purpose: Epithelial–mesenchymal transition (EMT) has been<br />

associated with metastatic spread and EGF receptor (EGFR)<br />

inhibitor resistance. We developed and validated a robust 76-<br />

gene EMT signature using gene expression pr<strong>of</strong>iles from four<br />

platforms using non–small cell lung carcinoma (NSCLC) cell lines<br />

and patients treated in the Biomarker-Integrated Approaches <strong>of</strong><br />

Targeted Therapy for Lung Cancer Elimination (BATTLE) study.<br />

Experimental Design: We conducted an integrated gene expression,<br />

proteomic, and drug response analysis using cell lines and tumors<br />

from patients with NSCLC. A 76-gene EMT signature was developed<br />

and validated using gene expression pr<strong>of</strong>iles from four microarray<br />

platforms <strong>of</strong> NSCLC cell lines and patients treated in the BATTLE study,<br />

and potential therapeutic targets associated with EMT were identified.<br />

Results: Compared with epithelial cells, mesenchymal cells<br />

showed significantly greater resistance to EGFR and PI3K/Akt<br />

pathway inhibitors, independent <strong>of</strong> EGFR mutation status, but<br />

more sensitivity to certain chemotherapies. Mesenchymal cells<br />

also expressed increased levels <strong>of</strong> the receptor tyrosine kinase Axl<br />

and showed a trend toward greater sensitivity to the Axl inhibitor<br />

SGI-7079, whereas the combination <strong>of</strong> SGI-7079 with erlotinib<br />

reversed erlotinib resistance in mesenchymal lines expressing<br />

Axl and in a xenograft model <strong>of</strong> mesenchymal NSCLC. In patients<br />

with NSCLC, the EMT signature predicted 8-week disease control<br />

in patients receiving erlotinib but not other therapies.<br />

Conclusion: We have developed a robust EMT signature that predicts<br />

resistance to EGFR and PI3K/Akt inhibitors, highlights different<br />

patterns <strong>of</strong> drug responsiveness for epithelial and mesenchymal cells,<br />

and identifies Axl as a potential therapeutic target for overcoming<br />

EGFR inhibitor resistance associated with the mesenchymal<br />

phenotype. Clin Cancer Res; 19(1); 279–90. ©2012 <strong>AACR</strong>.<br />

Clin Cancer Res January 1, 2013 19:1 279–90; Published OnlineFirst<br />

October 22, 2012; doi:10.1158/1078-0432.CCR-12-1558<br />

18<br />

aacrjournals.org


Molecular<br />

Cancer<br />

Research<br />

Molecular Cancer Research<br />

Karen E. Knudsen, PhD<br />

Editor-in-Chief<br />

Scope<br />

Molecular Cancer Research publishes articles describing novel basic<br />

cancer research discoveries <strong>of</strong> broad interest to the field. Studies<br />

must be <strong>of</strong> demonstrated significance, and the journal prioritizes<br />

analyses performed at the molecular and cellular level that reveal novel<br />

mechanistic insight into pathways and processes linked to cancer<br />

risk, development, and/or progression. Areas <strong>of</strong> emphasis include all<br />

cancer-associated pathways (including cell-cycle regulation; cell death;<br />

chromatin regulation; DNA damage and repair; gene and RNA regulation;<br />

genomics; oncogenes and tumor suppressors; and signal transduction),<br />

in addition to studies describing new molecular mechanisms and<br />

interactions that support cancer phenotypes.<br />

Review Article<br />

Radiation Survivors: Understanding and Exploiting the<br />

Phenotype following Fractionated Radiation Therapy<br />

Adeola Y. Makinde, Molykutty John-Aryankalayil, Sanjeewani T. Palayoor, David Cerna, and C. Norman Coleman<br />

Abstract<br />

Radiation oncology modalities such as intensity-modulated and<br />

image-guided radiation therapy can reduce the high dose to normal<br />

tissue and deliver a heterogeneous dose to tumors, focusing on areas<br />

deemed at highest risk for tumor persistence. Clinical radiation<br />

oncology produces daily doses ranging from 1 to 20 Gy, with<br />

tissues being exposed to 30 or more daily fractions. Hypothesizing<br />

the cells that survive fractionated radiation therapy have a<br />

substantially different phenotype than the untreated cells, which<br />

might be exploitable for targeting with molecular therapeutics or<br />

immunotherapy, three prostate cancer cell lines (PC3, DU145, and<br />

LNCaP) and normal endothelial cells were studied to understand<br />

the biology <strong>of</strong> differential effects <strong>of</strong> multifraction (MF) radiation<br />

<strong>of</strong> 0.5, 1, and/or 2 Gy fraction to 10 Gy total dose, and a single<br />

dose <strong>of</strong> 5 and 10 Gy. The resulting changes in mRNA, miRNA, and<br />

phosphoproteome were analyzed. Significant differences were<br />

Adeola Y. Makinde<br />

observed in the MF radiation exposures including those from the<br />

0.5 Gy MF that produces little cell killing. As expected, p53 function<br />

played a major role in response. Pathways modified by MF include<br />

immune response, DNA damage, cell-cycle arrest, TGF-β, survival,<br />

and apoptotic signal transduction. The radiation-induced stress<br />

response will set forth a unique platform for exploiting the effects<br />

<strong>of</strong> radiation therapy as “focused biology” for cancer treatment in<br />

conjunction with molecular targeted or immunologically directed<br />

therapy. Given that more normal tissue is treated, albeit to lower<br />

doses with these newer techniques, the response <strong>of</strong> the normal<br />

tissue may also influence long-term treatment outcome. Mol<br />

Cancer Res; 11(1); 5–12. ©2012 <strong>AACR</strong>.<br />

Mol Cancer Res January 2013 11:5–12; Published OnlineFirst<br />

November 21, 2012; doi:10.1158/1541-7786.MCR-12-0492<br />

The <strong>Best</strong> <strong>of</strong> the <strong>AACR</strong> <strong>Journals</strong> 19


Molecular Cancer Research<br />

Research Article<br />

A Modified HSP70 Inhibitor Shows Broad Activity<br />

as an Anticancer Agent<br />

Gregor M. Balaburski, Julia I.-Ju Leu, Neil Beeharry, Seth Hayik, Mark D. Andrake, Gao Zhang, Meenhard Herlyn,<br />

Jessie Villanueva, Roland L. Dunbrack, Tim Yen, Donna L. George, and Maureen E. Murphy<br />

Maureen E. Murphy<br />

Abstract<br />

The stress-induced HSP70 is an ATP-dependent molecular<br />

chaperone that plays a key role in refolding misfolded proteins<br />

and promoting cell survival following stress. HSP70 is marginally<br />

expressed in nontransformed cells, but is greatly overexpressed<br />

in tumor cells. Silencing HSP70 is uniformly cytotoxic to tumor<br />

but not normal cells; therefore, there has been great interest in<br />

the development <strong>of</strong> HSP70 inhibitors for cancer therapy. Here, we<br />

report that the HSP70 inhibitor 2-phenylethynesulfonamide (PES)<br />

binds to the substrate-binding domain <strong>of</strong> HSP70 and requires the<br />

C-terminal helical “lid” <strong>of</strong> this protein (amino acids 573–616) to<br />

bind. Using molecular modeling and in silico docking, we have<br />

identified a candidate binding site for PES in this region <strong>of</strong> HSP70,<br />

and we identify point mutants that fail to interact with PES. A<br />

preliminary structure–activity relationship analysis has revealed<br />

a derivative <strong>of</strong> PES, 2-(3-chlorophenyl) ethynesulfonamide (PES-<br />

Cl), which shows increased cytotoxicity and ability to inhibit<br />

autophagy, along with significantly improved ability to extend<br />

the life <strong>of</strong> mice with pre-B-cell lymphoma, compared with the<br />

parent compound (P = 0.015). Interestingly, we also show that<br />

these HSP70 inhibitors impair the activity <strong>of</strong> the anaphase<br />

promoting complex/cyclosome (APC/C) in cell-free extracts, and<br />

induce G 2 –M arrest and genomic instability in cancer cells. PES-<br />

Cl is thus a promising new anticancer compound with several<br />

notable mechanisms <strong>of</strong> action. Mol Cancer Res; 11(3); 219–29.<br />

©2013 <strong>AACR</strong>.<br />

Mol Cancer Res March 2013 11:219–229; Published OnlineFirst<br />

January 9, 2013; doi:10.1158/1541-7786.MCR-12-0547-T<br />

Research Article<br />

The Heterochronic microRNA let-7 Inhibits Cell Motility<br />

by Regulating the Genes in the Actin Cytoskeleton Pathway<br />

in Breast Cancer<br />

Xiaowen Hu, Jinyi Guo, Lan Zheng, Chunsheng Li, Tim M. Zheng, Janos L. Tanyi, Shun Liang, Chiara Benedetto,<br />

Marco Mitidieri, Dionyssios Katsaros, Xia Zhao, Youcheng Zhang, Qihong Huang, and Lin Zhang<br />

Lin Zhang<br />

Abstract<br />

The heterochronic gene let-7 serves as a tumor suppressor<br />

microRNA by targeting various oncogenic pathways in cancer<br />

cells. Considerable evidence indicates that reduced expression<br />

<strong>of</strong> let-7 might be associated with poor clinical outcome in<br />

patients with cancer. Here, we report that the expression levels<br />

<strong>of</strong> three let-7 family members, let-7a, let-7b, and let-7g, were<br />

significantly decreased in the patients with breast cancer with<br />

lymph node metastasis compared with those without lymph<br />

node metastasis. Enforced expression <strong>of</strong> let-7b significantly<br />

inhibits breast cancer cell motility and affects actin dynamics.<br />

Using bioinformatic and experimental approaches, four genes in<br />

the actin cytoskeleton pathway, including PAK1, DIAPH2, RDX,<br />

and ITGB8, were identified as let-7 direct targets. Blocking the<br />

expression <strong>of</strong> PAK1, DIAPH2, and RDX significantly inhibits<br />

breast cancer cell migration induced by let-7b repression.<br />

Our results indicate that reconstitution <strong>of</strong> let-7 expression in<br />

tumor cells could provide a novel therapeutic strategy for the<br />

treatment <strong>of</strong> metastatic disease. Mol Cancer Res; 11(3); 240–50.<br />

©2013 <strong>AACR</strong>.<br />

Mol Cancer Res March 2013 11:240–250; Published OnlineFirst<br />

January 21, 2013; doi:10.1158/1541-7786.MCR-12-0432<br />

20<br />

aacrjournals.org


Molecular Cancer Research<br />

Research Article<br />

Procollagen Lysyl Hydroxylase 2 Is Essential for<br />

Hypoxia-Induced Breast Cancer Metastasis<br />

Daniele M. Gilkes, Saumendra Bajpai, Carmen C. Wong, Pallavi Chaturvedi, Maimon E. Hubbi,<br />

Denis Wirtz, and Gregg L. Semenza<br />

Gregg L. Semenza<br />

Abstract<br />

Metastasis is the leading cause <strong>of</strong> death among patients who have<br />

breast cancer. Understanding the role <strong>of</strong> the extracellular matrix<br />

(ECM) in the metastatic process may lead to the development <strong>of</strong><br />

improved therapies to treat patients with cancer. Intratumoral<br />

hypoxia, found in the majority <strong>of</strong> breast cancers, is associated<br />

with an increased risk <strong>of</strong> metastasis and mortality. We found<br />

that in hypoxic breast cancer cells, hypoxia-inducible factor 1<br />

(HIF-1) activates transcription <strong>of</strong> the PLOD1 and PLOD2 genes<br />

encoding procollagen lysyl hydroxylases that are required for the<br />

biogenesis <strong>of</strong> collagen, which is a major constituent <strong>of</strong> the ECM.<br />

High PLOD2 expression in breast cancer biopsies is associated<br />

with increased risk <strong>of</strong> mortality. We show that PLOD2 is critical<br />

for fibrillar collagen formation by breast cancer cells, increases<br />

tumor stiffness, and is required for metastasis to lymph nodes<br />

and lungs. Mol Cancer Res; 11(5); 456–66. ©2013 <strong>AACR</strong>.<br />

Mol Cancer Res May 2013 11:456–466; Published OnlineFirst<br />

February 1, 2013; doi:10.1158/1541-7786.MCR-12-0629<br />

Research Article<br />

Activation <strong>of</strong> the FGF2-FGFR1 Autocrine Pathway: A Novel<br />

Mechanism <strong>of</strong> Acquired Resistance to Gefitinib in NSCLC<br />

Hideki Terai, Kenzo Soejima, Hiroyuki Yasuda, Sohei Nakayama, Junko Hamamoto, Daisuke Arai, Kota Ishioka,<br />

Keiko Ohgino, Shinnosuke Ikemura, Takashi Sato, Satoshi Yoda, Ryosuke Satomi, Katsuhiko Naoki, and<br />

Tomoko Betsuyaku<br />

Kenzo Soejima<br />

Abstract<br />

Patients with non-small cell lung cancer (NSCLC) that harbors<br />

epidermal growth factor receptor (EGFR) mutations initially<br />

respond to EGFR-tyrosine kinase inhibitors (TKI) but eventually<br />

experience relapse. Acquired resistance to EGFR-TKIs is<br />

strongly associated with patient mortality. Thus, elucidation <strong>of</strong><br />

the mechanism <strong>of</strong> acquired resistance to EGFR-TKIs is <strong>of</strong> great<br />

importance. In this study, gefitinib-resistant cell line models<br />

were established by long-term exposure to gefitinib using the<br />

gefitinib-sensitive lung cancer cell lines, PC9 and HCC827.<br />

Expression analyses indicated that both FGFR1 and FGF2 were<br />

increased in PC9 gefitinib-resistant (PC9 GR) cells as compared<br />

with PC9 naïve (PC9 na) cells. Importantly, proliferation <strong>of</strong><br />

gefitinib-resistant cells was dependent on the FGF2-FGFR1<br />

pathway. Mechanistically, inhibition <strong>of</strong> either FGF2 or FGFR1<br />

by siRNA or FGFR inhibitor (PD173074) restored gefitinib<br />

sensitivity in PC9 GR cells. These data suggest that FGF2-FGFR1<br />

activation through an autocrine loop is a novel mechanism <strong>of</strong><br />

acquired resistance to EGFR-TKIs. Mol Cancer Res; 11(7); 759–67.<br />

©2013 <strong>AACR</strong>.<br />

Mol Cancer Res July 2013 11:759–767; Published OnlineFirst March<br />

27, 2013; doi:10.1158/1541-7786.MCR-12-0652<br />

The <strong>Best</strong> <strong>of</strong> the <strong>AACR</strong> <strong>Journals</strong> 21


Molecular<br />

Cancer<br />

Therapeutics<br />

Molecular Cancer Therapeutics<br />

Napoleone Ferrara, MD<br />

Editor-in-Chief<br />

Scope<br />

Molecular Cancer Therapeutics strives to be the top choice for publishing<br />

the best science in the discovery and preclinical development <strong>of</strong><br />

novel therapeutic agents for oncology, preclinical studies <strong>of</strong> approved<br />

therapeutics, mechanisms <strong>of</strong> drug action, mechanism <strong>of</strong> drug resistance,<br />

biomarkers <strong>of</strong> drug response, novel models and technologies, and<br />

occasional drug toxicity mechanisms. While the journal’s main focus is<br />

on small molecule and protein drugs, other molecular entities may be<br />

considered.<br />

Review Article<br />

FOX(M1) News—It Is Cancer<br />

Marianna Halasi and Andrei L. Gartel<br />

Abstract<br />

FOXM1 is an oncogenic transcription factor <strong>of</strong> the Forkhead<br />

family and it has a well-defined role in cell proliferation and<br />

cell-cycle progression. Expression <strong>of</strong> FOXM1 is excluded in<br />

quiescent or differentiated cells, but its level is highly elevated<br />

in proliferating and malignant cells. Overexpression <strong>of</strong> FOXM1<br />

has been reported in more than 20 types <strong>of</strong> human cancer. In<br />

recent years, FOXM1 has been implicated in diverse cellular<br />

processes and also a growing body <strong>of</strong> experimental data has<br />

underlined the relevance <strong>of</strong> FOXM1 in tumorigenesis. Although<br />

FOXM1 is under the control <strong>of</strong> three major tumor suppressors<br />

Andrei L. Gartel<br />

(RB, p53, and p19 ARF ), it is still active in the majority <strong>of</strong> human<br />

cancers. The oncogenic potential <strong>of</strong> FOXM1 is mainly based on<br />

its ability to transcriptionally activate genes that are involved<br />

in different facets <strong>of</strong> cancer development. In this review, the<br />

contribution <strong>of</strong> FOXM1 to each <strong>of</strong> the hallmarks <strong>of</strong> cancer will<br />

be summarized and discussed. Mol Cancer Ther; 12(3); 245–54.<br />

©2012 <strong>AACR</strong>.<br />

Mol Cancer Ther March 2013 12:245–54; Published OnlineFirst<br />

February 26, 2013; doi:10.1158/1535-7163.MCT-12-0712<br />

22<br />

aacrjournals.org


Molecular Cancer Therapeutics<br />

Research Article<br />

Genome and Transcriptome Sequencing in Prospective Metastatic<br />

Triple-Negative Breast Cancer Uncovers Therapeutic Vulnerabilities<br />

David W. Craig, Joyce A. O’Shaughnessy, Jeffrey A. Kiefer, Jessica Aldrich, Shripad Sinari, Tracy M. Moses, Shukmei Wong,<br />

Jennifer Dinh, Alexis Christ<strong>of</strong>orides, Joanne L. Blum, Cristi L. Aitelli, Cynthia R. Osborne, Tyler Izatt, Ahmet Kurdoglu,<br />

Angela Baker, Julie Koeman, Catalin Barbacioru, Onur Sakarya, Francisco M. De La Vega, Asim Siddiqui, Linh Hoang,<br />

Paul R. Billings, Bodour Salhia, Anthony W. Tolcher, Jeffrey M. Trent, Spyro Mousses, Daniel Von H<strong>of</strong>f, and John D. Carpten<br />

John D. Carpten<br />

Abstract<br />

Triple-negative breast cancer (TNBC) is characterized by<br />

the absence <strong>of</strong> expression <strong>of</strong> estrogen receptor, progesterone<br />

receptor, and HER-2. Thirty percent <strong>of</strong> patients recur after<br />

first-line treatment, and metastatic TNBC (mTNBC) has a poor<br />

prognosis with median survival <strong>of</strong> one year. Here, we present<br />

initial analyses <strong>of</strong> whole genome and transcriptome sequencing<br />

data from 14 prospective mTNBC. We have cataloged the<br />

collection <strong>of</strong> somatic genomic alterations in these advanced<br />

tumors, particularly those that may inform targeted therapies.<br />

Genes mutated in multiple tumors included TP53, LRP1B,<br />

HERC1, CDH5, RB1, and NF1. Notable genes involved in focal<br />

structural events were CTNNA1, PTEN, FBXW7, BRCA2, WT1,<br />

FGFR1, KRAS, HRAS, ARAF, BRAF, and PGCP. Homozygous<br />

deletion <strong>of</strong> CTNNA1 was detected in 2 <strong>of</strong> 6 African Americans.<br />

RNA sequencing revealed consistent overexpression <strong>of</strong> the<br />

FOXM1 gene when tumor gene expression was compared<br />

with nonmalignant breast samples. Using an outlier analysis<br />

<strong>of</strong> gene expression comparing one cancer with all the others,<br />

we detected expression patterns unique to each patient’s<br />

tumor. Integrative DNA/RNA analysis provided evidence for<br />

deregulation <strong>of</strong> mutated genes, including the monoallelic<br />

expression <strong>of</strong> TP53 mutations. Finally, molecular alterations in<br />

several cancers supported targeted therapeutic intervention on<br />

clinical trials with known inhibitors, particularly for alterations<br />

in the RAS/RAF/MEK/ERK and PI3K/AKT/mTOR pathways.<br />

In conclusion, whole genome and transcriptome pr<strong>of</strong>iling <strong>of</strong><br />

mTNBC have provided insights into somatic events occurring<br />

in this difficult to treat cancer. These genomic data have<br />

guided patients to investigational treatment trials and provide<br />

hypotheses for future trials in this irremediable cancer. Mol<br />

Cancer Ther; 12(1); 104–16. ©2012 <strong>AACR</strong>.<br />

Mol Cancer Ther January 2013 12:104–16; Published OnlineFirst<br />

November 19, 2012; doi:10.1158/1535-7163.MCT-12-0781<br />

Research Article<br />

EGFR Exon 20 Insertion Mutations in Lung Adenocarcinomas:<br />

Prevalence, Molecular Heterogeneity, and Clinicopathologic<br />

Characteristics<br />

Maria E. Arcila, Khedoudja Nafa, Jamie E. Chaft, Natasha Rekhtman, Christopher Lau, Boris A. Reva,<br />

Maureen F. Zakowski, Mark G. Kris, and Marc Ladanyi<br />

Maria E. Arcila<br />

Abstract<br />

In contrast to other primary epidermal growth factor receptor<br />

(EGFR) mutations in lung adenocarcinomas, insertions in exon<br />

20 <strong>of</strong> EGFR have been generally associated with resistance to<br />

EGFR-tyrosine kinase inhibitors. Their molecular spectrum,<br />

clinicopathologic characteristics, and prevalence are not<br />

well established. Tumors harboring EGFR exon 20 insertions<br />

were identified through an algorithmic screen <strong>of</strong> 1,500 lung<br />

adenocarcinomas. Cases were first tested for common mutations<br />

in EGFR (exons 19 and 21) and KRAS (exon 2) and, if negative,<br />

further analyzed for EGFR exon 20 insertions. All samples<br />

underwent extended genotyping for other driver mutations in<br />

EGFR, KRAS, BRAF, ERBB2/HER2, NRAS, PIK3CA, MEK1, and<br />

AKT by mass spectrometry; a subset was evaluated for ALK<br />

rearrangements. We identified 33 EGFR exon 20 insertion cases<br />

[2.2%, 95% confidence interval (CI), 1.6–3.1], all mutually exclusive<br />

with mutations in the other genes tested (except PIK3CA). They<br />

were more common among never-smokers (P < 0.0001). There<br />

was no association with age, sex, race, or stage. Morphologically,<br />

tumors were similar to those with common EGFR mutations but<br />

with frequent solid histology. Insertions were highly variable<br />

in position and size, ranging from 3 to 12 bp, resulting in 13<br />

different insertions, which, by molecular modeling, are predicted<br />

to have potentially different effects on erlotinib binding. EGFR<br />

exon 20 insertion testing identifies a distinct subset <strong>of</strong> lung<br />

adenocarcinomas, accounting for at least 9% <strong>of</strong> all EGFR-mutated<br />

cases, representing the third most common type <strong>of</strong> EGFR mutation<br />

after exon 19 deletions and L858R. Insertions are structurally<br />

heterogeneous with potential implications for response to EGFR<br />

inhibitors. Mol Cancer Ther; 12(2); 220–9. ©2012 <strong>AACR</strong>.<br />

Mol Cancer Ther February 2013 12:220–9; Published OnlineFirst<br />

January 31, 2013; doi:10.1158/1535-7163.MCT-12-0620<br />

The <strong>Best</strong> <strong>of</strong> the <strong>AACR</strong> <strong>Journals</strong> 23


Molecular Cancer Therapeutics<br />

Research Article<br />

NF-κB2/p52 Induces Resistance to Enzalutamide in<br />

Prostate Cancer: Role <strong>of</strong> Androgen Receptor and Its<br />

Variants<br />

Nagalakshmi Nadiminty, Ramakumar Tummala, Chengfei Liu, Joy Yang, Wei Lou,<br />

Christopher P. Evans, and Allen C. Gao<br />

Nagalakshmi Nadiminty<br />

Allen C. Gao<br />

Abstract<br />

Resistance <strong>of</strong> prostate cancer cells to the next-generation<br />

antiandrogen enzalutamide may be mediated by a multitude<br />

<strong>of</strong> survival signaling pathways. In this study, we tested whether<br />

increased expression <strong>of</strong> NF-κB2/p52 induces prostate cancer<br />

cell resistance to enzalutamide and whether this response is<br />

mediated by aberrant androgen receptor (AR) activation and<br />

AR splice variant production. LNCaP cells stably expressing<br />

NF-κB2/p52 exhibited higher survival rates than controls<br />

when treated with enzalutamide. C4-2B and CWR22Rv1 cells<br />

chronically treated with enzalutamide were found to express<br />

higher levels <strong>of</strong> NF-κB2/p52. Downregulation <strong>of</strong> NF-κB2/p52<br />

in CWR22Rv1 cells chronically treated with enzalutamide<br />

rendered them more sensitive to cell growth inhibition by<br />

enzalutamide. Analysis <strong>of</strong> the expression levels <strong>of</strong> AR splice<br />

variants by quantitative reverse transcription PCR and Western<br />

blotting revealed that LNCaP cells expressing p52 exhibit higher<br />

expression <strong>of</strong> AR splice variants. Downregulation <strong>of</strong> expression<br />

<strong>of</strong> NF-κB2/p52 in VCaP and CWR22Rv1 cells by short hairpin<br />

RNA abolished expression <strong>of</strong> splice variants. Downregulation<br />

<strong>of</strong> expression <strong>of</strong> either full-length AR or the splice variant AR-<br />

V7 led to an increase in sensitivity <strong>of</strong> prostate cancer cells to<br />

enzalutamide. These results collectively demonstrate that<br />

resistance to enzalutamide may be mediated by NF-κB2/p52 via<br />

activation <strong>of</strong> AR and its splice variants. Mol Cancer Ther; 12(8);<br />

1629–37. ©2013 <strong>AACR</strong>.<br />

Mol Cancer Ther August 2013 12:1629–37; Published OnlineFirst<br />

May 22, 2013; doi:10.1158/1535-7163.MCT-13-0027<br />

Research Article<br />

Inhibition <strong>of</strong> Mutant GNAQ Signaling in Uveal Melanoma Induces<br />

AMPK-Dependent Autophagic Cell Death<br />

Grazia Ambrosini, Elgilda Musi, Alan L. Ho, Elisa de Stanchina, and Gary K. Schwartz<br />

Abstract<br />

Grazia Ambrosini<br />

Oncogenic mutations in GNAQ and GNA11 genes are found in 80%<br />

<strong>of</strong> uveal melanoma. These mutations result in the activation <strong>of</strong><br />

the RAF/MEK signaling pathway culminating in the stimulation<br />

<strong>of</strong> ERK1/2 mitogen-activated protein kinases. In this study, using<br />

a siRNA strategy, we show that mutant GNAQ signals to both<br />

MEK and AKT, and that combined inhibition <strong>of</strong> these pathways<br />

with the MEK inhibitor selumetinib (AZD6244) and the AKT<br />

inhibitor MK2206 induced a synergistic decrease in cell viability.<br />

This effect was genotype dependent as autophagic markers like<br />

beclin1 and LC3 were induced in GNAQ-mutant cells, whereas<br />

apoptosis was the mechanism <strong>of</strong> cell death <strong>of</strong> BRAF-mutant cells,<br />

and cells without either mutation underwent cell-cycle arrest.<br />

The inhibition <strong>of</strong> MEK/ATK pathways induced activation <strong>of</strong> AMPactivated<br />

protein kinase (AMPK) in the GNAQ-mutant cells.<br />

The downregulation <strong>of</strong> AMPK by siRNA or its inhibition with<br />

compound C did not rescue the cells from autophagy, rather they<br />

died by apoptosis, defining AMPK as a key regulator <strong>of</strong> mutant<br />

GNAQ signaling and a switch between autophagy and apoptosis.<br />

Furthermore, this combination treatment was effective in<br />

inhibiting tumor growth in xenograft mouse models. These<br />

findings suggest that inhibition <strong>of</strong> MEK and AKT may represent<br />

a promising approach for targeted therapy <strong>of</strong> patients with uveal<br />

melanoma. Mol Cancer Ther; 12(5); 768–76. ©2013 <strong>AACR</strong>.<br />

Mol Cancer Ther May 2013 12:768–76; Published OnlineFirst<br />

February 26, 2013; doi:10.1158/1535-7163.MCT-12-1020<br />

24<br />

aacrjournals.org


Mobile Access:<br />

Take the <strong>AACR</strong> <strong>Journals</strong> with you, wherever you go!<br />

Get access to<br />

<strong>AACR</strong> PUBLICATIONS<br />

on your smartphone and tablet.<br />

Search “<strong>AACR</strong> Publications” in the<br />

iTunes App Store to download the FREE app.<br />

The <strong>AACR</strong> online publications are available in a mobileoptimized<br />

format. The content presented on the mobile site<br />

is in a format designed specifically for the smaller screen size<br />

and functionality <strong>of</strong> a mobile device. This format, coupled<br />

with faster loading capabilities on low-bandwidth networks,<br />

provides easy access to journal articles. The mobile edition<br />

is compatible with most smartphones and mobile devices,<br />

including Blackberry, Droid and iPhone.<br />

iPhone and iPad users can access full-text <strong>AACR</strong> journal<br />

content through apps designed specifically for those devices.<br />

Search for “<strong>AACR</strong> Publications” in the iTunes App Store to<br />

download this FREE app.<br />

Learn more at<br />

<strong>AACR</strong>journals.org

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!