You are on page 1of 2648

Any screen.

Any time.
Anywhere.
Activate the eBook version
of this title at no additional charge.

Expert Consult eBooks give you the power to browse and find content,
view enhanced images, share notes and highlights—both online and offline.

Unlock your eBook today.


1 Visit expertconsult.inkling.com/redeem Scan this QR code to redeem your
eBook through your mobile device:
2 Scratch off your code
3 Type code into “Enter Code” box

4 Click “Redeem”
5 Log in or Sign up
6 Go to “My Library”
Place Peel Off
It’s that easy! Sticker Here

For technical assistance:


email expertconsult.help@elsevier.com
call 1-800-401-9962 (inside the US)
call +1-314-447-8200 (outside the US)
Use of the current edition of the electronic version of this book (eBook) is subject to the terms of the nontransferable, limited license granted on
expertconsult.inkling.com. Access to the eBook is limited to the first individual who redeems the PIN, located on the inside cover of this book,
at expertconsult.inkling.com and may not be transferred to another party by resale, lending, or other means.
2015v1.0
Abeloff’s
CLINICAL
ONCOLOGY
This page intentionally left blank
Abeloff’s
CLINICAL
ONCOLOGY
SIXTH EDITION
JOHN E. NIEDERHUBER, MD
Executive Vice President, Inova Health System
President and CEO, Genomics and Bioinformatics Research Institute
Fairfax, Virginia;
Professor, Department of Public Health Sciences
Member, Center for Public Health Genomics
University of Virginia School of Medicine
Charlottesville, Virginia;
Adjunct Professor, Oncology and Surgery
The Johns Hopkins University School of Medicine
Deputy Director
Johns Hopkins Clinical Research Network
Baltimore, Maryland

JAMES O. ARMITAGE, MD JAMES H. DOROSHOW, MD


Joe Shapiro Professor of Medicine Bethesda, Maryland
University of Nebraska Medical Center
Omaha, Nebraska

MICHAEL B. KASTAN, MD, PhD JOEL E.TEPPER, MD


Executive Director, Duke Cancer Institute Hector MacLean Distinguished Professor of
William and Jane Shingleton Professor, Cancer Research
Pharmacology and Cancer Biology Department of Radiation Oncology
Professor of Pediatrics UNC Lineberger Comprehensive Cancer Center
Duke University School of Medicine University of North Carolina School of Medicine
Durham, North Carolina Chapel Hill, North Carolina
ABELOFF’S CLINICAL ONCOLOGY, SIXTH EDITION ISBN: 978-0-323-47674-4

Copyright © 2020 by Elsevier, Inc. All rights reserved.

No part of this publication may be reproduced or transmitted in any form or by any means, electronic or
mechanical, including photocopying, recording, or any information storage and retrieval system, without
permission in writing from the publisher. Details on how to seek permission, further information about the
Publisher’s permissions policies and our arrangements with organizations such as the Copyright Clearance
Center and the Copyright Licensing Agency, can be found at our website: www.elsevier.com/permissions.

This book and the individual contributions contained in it are protected under copyright by the Publisher
(other than as may be noted herein).

Notices

Knowledge and best practice in this field are constantly changing. As new research and experience
broaden our understanding, changes in research methods, professional practices, or medical treatment
may become necessary.
Practitioners and researchers must always rely on their own experience and knowledge in evaluating
and using any information, methods, compounds, or experiments described herein. In using such
information or methods they should be mindful of their own safety and the safety of others, including
parties for whom they have a professional responsibility.
With respect to any drug or pharmaceutical products identified, readers are advised to check the
most current information provided (i) on procedures featured or (ii) by the manufacturer of each product
to be administered, to verify the recommended dose or formula, the method and duration of
administration, and contraindications. It is the responsibility of practitioners, relying on their own
experience and knowledge of their patients, to make diagnoses, to determine dosages and the best
treatment for each individual patient, and to take all appropriate safety precautions.
To the fullest extent of the law, neither the Publisher nor the authors, contributors, or editors,
assume any liability for any injury and/or damage to persons or property as a matter of products liability,
negligence or otherwise, or from any use or operation of any methods, products, instructions, or ideas
contained in the material herein.

Previous editions copyrighted 2014, 2008, 2004, 2000, and 1995.

Library of Congress Control Number: 2018953655

Executive Content Strategist: Robin Carter


Content Development Manager: Laura Schmidt
Publishing Services Manager: Catherine Jackson
Senior Project Manager: Amanda Mincher
Design Direction: Bridget Hoette

Printed in China

Last digit is the print number: 9 8 7 6 5 4 3 2 1

1600 John F. Kennedy Blvd.


Ste 1600
Philadelphia, PA 19103-2899
To my son, Matthew, and my wife, Kathy, who have and continue to make sacrifices so that I might pursue my passions in
medicine and research. To my colleagues at the National Cancer Institute, University of Virginia, Johns Hopkins, and across the
country, whose selfless dedication to patient care and cancer research is truly an inspiration to all. To the many students who have
trained with me over the years, to my patients, and to my colleagues at the Inova Translational Medicine Institute, who have given
me the opportunity to have this tremendously rewarding career. Lastly, to Tracey, and to Marty, who, in memory, inspire all who
knew them to work a little harder each day toward the elimination of the pain and suffering from this disease.
JOHN E. NIEDERHUBER, MD

To my wife, Nancy, for her love and support over 49 ½ years.


JAMES O. ARMITAGE, MD

To my wife, Robin Winkler Doroshow, MD, my classmate and greatest supporter, for her love, dedication, and commitment and
for the remarkable joy and caring she brings to her patients and to all around her. To my remarkable daughter, Deborah Doroshow,
MD, PhD, who is completing her training for a career in academic oncology; my fondest hope is that you will enjoy sharing with
and learning from those you help as much as I have. To my patients and colleagues at the City of Hope and the National Cancer
Institute who have all contributed so much of themselves to my continuing education as a physician and investigator, please accept
my appreciation and utmost gratitude.
JAMES H. DOROSHOW, MD

To my wife, Kathy, and my sons, Benjamin, Nathaniel, and Jonathan. You are the lights of my life. I also acknowledge all of my
mentors, colleagues, and patients, who have taught me so much. A special note of gratitude goes to Marty Abeloff, a mentor and an
inspiring role model for career and for life.
MICHAEL B. KASTAN, MD, PhD

To my wife, Laurie, who has been my soul mate for many years and has constantly reminded me of life’s priorities. To my family
including my daughters, Miriam and Abigail, and my grandchildren, Zekariah, Zohar, Samuel, Marcelo, Jonah, and Aurelio. They
have been an inspiration. To my many teachers through the years who have helped define and foster my professional career, but
especially Herman Suit and Eli Glatstein.
JOEL E. TEPPER, MD
This page intentionally left blank
Memoriam

Martin D. Abeloff, MD (1942-2007)

Martin D. Abeloff, a founding editor of Clinical Oncology, dedicated that would be as valuable to the practicing oncologist as to the primary
his life to caring for patients with cancer and to teaching his art to care physician and physicians-in-training. The first edition of Clinical
fellows, residents, and students. He was a brilliant and caring clinician, Oncology was published in 1995 to a gratifying response. It is now
an extremely effective leader, and a beloved mentor to many trainees established as a cornerstone reference for those caring for patients
and young faculty. with cancer.
Marty was born on April 4, 1942, in Shenandoah, Pennsylvania. He In the sixth edition, we continue Marty’s vision for an ever better,
received his BA from The Johns Hopkins University in 1963 and his unique, and accessible text so that future generations of oncologists
MD from The Johns Hopkins University School of Medicine in 1966. will remember his inspiration and leadership.
He spent the next year as an intern at the University of Chicago Hospitals The editors again dedicate this text, which is already a recognized
and Clinics. His legacy in medicine was established on his return to tangible aspect of his legacy in medicine, as a living memorial to him.
Baltimore in 1971 as a fellow in clinical oncology. He would spend the Abeloff ’s Clinical Oncology will continue to serve as a reminder to all
rest of his career at The Johns Hopkins Hospital, achieving the rank of its users of this extraordinary person and exemplary physician who
Professor of Medicine in 1990. At various times, he served as the fel- went before them.
lowship training program director, chief of medical oncology, clinical
director of the cancer center, oncologist in chief at The Johns Hopkins John E. Niederhuber, MD
Hospital, and in 1992, was appointed the second director of The Johns James O. Armitage, MD
Hopkins Oncology Center, later renamed, thanks to Marty’s efforts, the James H. Doroshow, MD
Sidney Kimmel Comprehensive Cancer Center. Michael B. Kastan, MD, PhD
It was during his time as cancer center director that Marty brought Joel E. Tepper, MD
to life the idea of a comprehensive, user-friendly textbook of oncology

vii
This page intentionally left blank
Contributors

James L. Abbruzzese, MD, FACP, FASCO, DSc (hon) Dara L. Aisner, MD, PhD
Duke Cancer Institute Distinguished Professor of Medical Oncology Associate Professor of Pathology
Chief, Division of Medical Oncology, Department of Medicine CU Anschutz Medical Campus
Associate Director for Clinical Research, Duke Cancer Institute University of Colorado
Duke University Medical Center Aurora, Colorado
Durham, North Carolina
Michelle Alonso-Basanta, MD, PhD
Omar Abdel-Wahab, MD Helene Blum Assistant Professor
Associate Attending Department of Radiation Oncology
Department of Medicine University of Pennsylvania
Leukemia Service Philadelphia, Pennsylvania
Memorial Sloan Kettering Cancer Center
New York, New York Jesus Anampa, MD, MS
Assistant Professor
Ghassan K. Abou-Alfa, MD Department of Oncology
Attending Physician Montefiore Medical Center
Memorial Sloan Kettering Cancer Center Albert Einstein College of Medicine
Professor of Medicine Bronx, New York
Weill Cornell Medicine
New York, New York Megan E. Anderson, MD
Assistant Professor
Janet L. Abrahm, MD Department of Orthopaedic Surgery
Professor of Medicine Harvard Medical School
Harvard Medical School Attending Orthopedic Surgeon
Member, Division of Palliative Care Department of Orthopedic Surgery
Psychosocial Oncology and Palliative Care Boston Children’s Hospital
Dana-Farber Cancer Institute Attending Orthopedic Surgeon
Boston, Massachusetts Department of Orthopedic Surgery
Beth Israel Deaconess Medical Center
Jeffrey S. Abrams, MD Boston, Massachusetts
Associate Director, Cancer Therapy Evaluation Program
Division of Cancer Treatment and Diagnosis Emmanuel S. Antonarakis, MD
National Cancer Institute Associate Professor of Oncology
Rockville, Maryland Sidney Kimmel Comprehensive Cancer Center
Johns Hopkins University School of Medicine
Jeremy S. Abramson, MD, MMSc Baltimore, Maryland
Director, Center for Lymphoma
Hematology/Oncology Richard Aplenc, MD, PhD
Massachusetts General Hospital Department of Pediatrics
Assistant Professor Section Chief, Hematologic Malignancies
Department of Medicine Chief Clinical Research Officer
Harvard Medical School Children’s Hospital of Philadelphia
Boston, Massachusetts Philadelphia, Pennsylvania

ix
x Contributors

Frederick R. Appelbaum, MD Karen Basen-Engquist, PhD, MPH


Executive Vice President and Deputy Director Professor of Behavioral Science
Fred Hutchinson Cancer Research Center University of Texas MD Anderson Cancer Center
Professor Houston, Texas
Division of Medical Oncology
University of Washington Lynda Kwon Beaupin, MD
Seattle, Washington Director, Adolescent and Young Adult Program
Roswell Park Cancer Institute
Luiz H. Araujo, MD, PhD Buffalo, New York
Scientific Director
COI Institute for Research and Education Ross S. Berkowitz, MD
Brazilian National Cancer Institute William H. Baker Professor of Gynecology
Rio de Janeiro, Brazil Department of Obstetrics and Gynecology
Harvard Medical School
Ammar Asban, MD Director of Gynecologic Oncology
Surgical Resident Department of Obstetrics and Gynecology
Department of Surgery Brigham and Women’s Hospital
University of Alabama at Birmingham Boston, Massachusetts
Birmingham, Alabama
Donald A. Berry, PhD
Edward Ashwood, MD Professor of Biostatistics
President and CEO Department of Biostatistics
ARUP Laboratories The University of Texas MD Anderson Cancer Center
Professor of Pathology Houston, Texas
University of Utah
Salt Lake City, Utah Therese Bevers, MD
Professor of Clinical Cancer Prevention
Farrukh T. Awan, MD, MS Medical Director, Cancer Prevention Center
Associate Professor of Medicine The University of Texas MD Anderson Cancer Center
Hematology Houston, Texas
The Ohio State University
Columbus, Ohio John F. Boggess, MD
Professor of Obstetrics and Gynecology
Juliet L. Aylward, MD University of North Carolina
Associate Professor of Dermatology Chapel Hill, North Carolina
University of Wisconsin School of Medicine and Public Health
Madison, Wisconsin Julie R. Brahmer, MD, MSc
Professor of Oncology
Arjun V. Balar, MD Department of Oncology
Associate Professor of Medicine Johns Hopkins Kimmel Cancer Center
Division of Hematology/Oncology Baltimore, Maryland
Director, Genitourinary Cancers Program
New York University Perlmutter Cancer Center Janet Brown, MD, FRCP, MSc, MBBS, BSc
New York University Langone Medical Center Professor
New York, New York Academic Unit of Clinical Oncology, Oncology, and Metabolism
Weston Park Hospital
Courtney J. Balentine, MD University of Sheffield
Assistant Professor of Surgery Sheffield, United Kingdom
Dallas VA Hospital
University of Texas Southwestern Karen Brown, MD
Dallas, Texas Attending Physician
Memorial Sloan Kettering Cancer Center
Stefan K. Barta, MD, MS, MRCP(UK) Professor of Clinical Radiology
Associate Professor Weill Medical College at Cornell University
Hematology and Oncology New York, New York
Fox Chase Cancer Center
Philadelphia, Pennsylvania Powel Brown, MD, PhD
Professor and Chairman
Nancy Bartlett, MD Clinical Cancer Prevention
Professor of Medical Oncology The University of Texas MD Anderson Cancer Center
Washington University School of Medicine Houston, Texas
St. Louis, Missouri
Contributors xi

Ilene Browner, MD Stephen J. Chanock, MD


Assistant Professor Director
Department of Oncology and Division of Geriatric Medicine Division of Cancer Epidemiology and Genetics
The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins National Cancer Institute
and Johns Hopkins Bayview Bethesda, Maryland
The Johns Hopkins University
Baltimore, Maryland Claudia I. Chapuy, MD
St. Elizabeth’s Medical Center
Paul A. Bunn, MD Dana-Farber Cancer Institute
Distinguished Professor of Medical Oncology Boston, Massachusetts
CU Anschutz Medical Campus
University of Colorado Vikash P. Chauhan, PhD
Aurora, Colorado Massachusetts Institute of Technology
Boston, Massachusetts
William R. Burns, MD
Assistant Professor of Surgery Herbert Chen, MD, FACS
University of Michigan Health System Chairman and Fay Fletcher Kerner Endowed Chair
Ann Arbor, Michigan Department of Surgery
University of Alabama at Birmingham
John C. Byrd, MD Surgeon-in-Chief
Professor of Internal Medicine–Hematology University of Alabama at Birmingham Health System
The Ohio State University Birmingham, Alabama
Columbus, Ohio
Ronald C. Chen, MD, MPH
Karen Cadoo, MD Associate Professor
Attending Medical Oncologist Department of Radiation Oncology
Gynecologic Medical Oncology and Clinical Genetic Services University of North Carolina at Chapel Hill
Memorial Sloan Kettering Cancer Center Chapel Hill, North Carolina
Weill Cornell Medical College
New York, New York Nai-Kong V. Cheung, MD, PhD
Enid A. Haupt Endowed Chair in Pediatric Oncology
David P. Carbone, MD, PhD Department of Pediatrics
Professor of Medicine Memorial Sloan-Kettering Cancer Center
Director, James Thoracic Center New York, New York
James Cancer Center
The Ohio State University Medical Center Jennifer H. Choe, MD, PhD
Columbus, Ohio Medical Instructor
Division of Medical Oncology
H. Ballentine Carter, MD Duke Cancer Institute
Professor of Urology Durham, North Carolina
Johns Hopkins University School of Medicine
Baltimore, Maryland Michaele C. Christian, MD
Cancer Therapy Evaluation Program (Retired)
Jorge J. Castillo, MD National Cancer Institute
Physician Rockville, Maryland
Hematologic Malignancies
Dana-Farber Cancer Institute Paul M. Cinciripini, PhD
Assistant Professor Professor and Chair of Behavioral Science
Harvard Medical School The University of Texas MD Anderson Cancer Center
Boston, Massachusetts Houston, Texas

Alfred E. Chang, MD Michael F. Clarke, MD


Hugh Cabot Professor of Surgery Professor of Medicine
University of Michigan Health System Division of Oncology
Ann Arbor, Michigan Stanford School of Medicine
Palo Alto, California
Eric Chang, MD, FASTRO
Professor and Chair of Radiation Oncology Robert E. Coleman, MBBS, MD
Keck School of Medicine of USC Academic Unit of Clinical Oncology
Los Angeles, California Weston Park Hospital
University of Sheffield
Sheffield, United Kingdom
xii Contributors

Robert L. Coleman, MD Jeffrey Crawford, MD


Professor and Executive Director, Cancer Network Research Professor of Medicine
Department of Gynecologic Oncology and Reproductive Medicine Division of Medical Oncology
The University of Texas MD Anderson Cancer Center Duke Cancer Institute
Houston, Texas Durham, North Carolina

Adriana M. Coletta, PhD, RD Kristy Crooks, PhD


Department of Behavioral Science Assistant Professor of Pathology
Center for Energy Balance in Cancer Prevention and Survivorship CU Anschutz Medical Campus
The University of Texas MD Anderson Cancer Center University of Colorado
Houston, Texas Aurora, Colorado

Jerry M. Collins, PhD Daniel J. Culkin, MD


Associate Director Professor
Division of Cancer Treatment and Diagnosis Department of Urology
National Cancer Institute University of Oklahoma Health Sciences Center
Bethesda, Maryland Oklahoma City, Oklahoma

Jean M. Connors, MD Brian G. Czito, MD


Hematology Division Professor of Radiation Oncology
Brigham and Women’s Hospital Duke University Medical Center
Dana-Farber Cancer Institute Durham, North Carolina
Harvard Medical School
Boston, Massachusetts Piero Dalerba, MD
Assistant Professor of Pathology and Cell Biology
Michael Cools, MD Assistant Professor of Medicine
Department of Neurosurgery Division of Digestive and Liver Diseases
University of North Carolina Columbia University College of Physicians and Surgeons
Chapel Hill, North Carolina New York, New York

Kevin R. Coombes, PhD Josep Dalmau, MD, PhD


Professor of Biomedical Informatics ICREA Research Professor
The Ohio State University Hospital Clínic/Institut d’Investigació Biomèdica August Pi i Sunyer
Columbus, Ohio (IDIBAPS)
Barcelona, Spain
Jorge Cortes, MD Adjunct Professor Neurology
The University of Texas MD Anderson Cancer Center University of Pennsylvania
Houston, Texas Philadelphia, Pennsylvania

Mauro W. Costa, MSc, PhD Mai Dang, MD, PhD


Research Scientist Instructor in Neurology
The Jackson Laboratory Children’s Hospital of Philadelphia
Bar Harbor, Maine Philadelphia, Pennsylvania

Anne Covey, MD Michael D’Angelica, MD


Attending Physician Attending Physician
Memorial Sloan Kettering Cancer Center Memorial Sloan Kettering Cancer Center
Professor of Radiology Professor of Surgery
Weill Medical College at Cornell University Weill Medical College at Cornell University
New York, New York New York, New York

Kenneth H. Cowan, MD, PhD Kurtis D. Davies, PhD


Director, Fred and Pamela Buffett Cancer Center Assistant Professor of Pathology
University of Nebraska Medical Center CU Anschutz Medical Campus
Omaha, Nebraska University of Colorado
Aurora, Colorado
Christopher H. Crane, MD
Vice Chairman
Attending Physician
Memorial Sloan Kettering Cancer Center
New York, New York
Contributors xiii

Myrtle Davis, DVM, PhD James H. Doroshow, MD


Chief, Toxicology and Pharmacology Branch Bethesda, Maryland
Division of Drug Treatment and Diagnosis
National Cancer Institute Jay F. Dorsey, MD, PhD
National Institutes of Health Associate Professor of Radiation Oncology
Bethesda, Maryland University of Pennsylvania
Philadelphia, Pennsylvania
Nicolas Dea, MD, MSc, FRCSC
Spinal Neurosurgeon Marianne Dubard-Gault, MD, MS
Clinical Associate Professor Medical Genetics Fellow
Department of Surgery Department of Medicine
Vancouver General Hospital Memorial Sloan Kettering Cancer Center
University of British Columbia New York, New York
Vancouver, British-Columbia, Canada
Steven G. DuBois, MD, MS
Ana De Jesus-Acosta, MD Associate Professor
Assistant Professor of Oncology Department of Pediatrics
Sidney Kimmel Comprehensive Cancer Center Harvard Medical School
The Johns Hopkins University School of Medicine Attending Physician
Baltimore, Maryland Department of Pediatrics
Boston Children’s Hospital
Angelo M. DeMarzo, MD, PhD Dana Farber Cancer Institute
Professor of Pathology Boston, Massachusetts
Johns Hopkins University School of Medicine
Baltimore, Maryland Dan G. Duda, PhD, DMD
Associate Professor
Theodore L. DeWeese, MD Harvard Medical School
Professor and Director of Radiation Oncology and Molecular Boston, Massachusetts
Radiation Sciences
Johns Hopkins University School of Medicine Malcolm Dunlop, MD
Baltimore, Maryland MRC Institute of Genetics and Molecular Medicine
The University of Edinburgh
Maximilian Diehn, MD, PhD Western General Hospital
Associate Professor of Radiation Oncology Edinburgh, United Kingdom
Stanford University
Palo Alto, California Linda R. Duska, MD
University of Virginia Health System
Subba R. Digumarthy, MD Emily Couric Clinical Cancer Center
Massachusetts General Hospital Charlottesville, Virginia
Boston, Massachusetts
Madeleine Duvic, MD
Angela Dispenzieri, MD Professor and Deputy Chairman
Professor of Medicine and Laboratory Medicine Department of Dermatology
Mayo Clinic The University of Texas MD Anderson Cancer Center
Rochester, Minnesota Houston, Texas

Khanh T. Do, MD Imane El Dika, MD


Assistant Professor of Medicine Assistant Attending Physician
Harvard Medical School Memorial Sloan Kettering Cancer Center
Medical Oncology Instructor of Medicine
Dana-Farber Cancer Institute Weill Medical College at Cornell University
Boston, Massachusetts New York, New York

Konstantin Dobrenkov, MD Hashem El-Serag, MD, MPH


Clinical Director, Oncology Margaret M. and Albert B. Alkek Chair of the Department
Merck & Company, Inc. of Medicine
Kenilworth, New Jersey Professor of Gastroenterology and Hepatology
Baylor College of Medicine
Jeffrey S. Dome, MD, PhD Houston, Texas
Vice President, Center for Cancer and Blood Disorders
Children’s National Medical Center
Washington, D.C.
xiv Contributors

Jeffrey M. Engelmann, PhD Debra L. Friedman, MD, MS


Assistant Professor of Psychiatry and Behavioral Medicine Vanderbilt-Ingram Cancer Center
Medical College of Wisconsin Nashville, Tennessee
Milwaukee, Wisconsin
Arian F. Fuller, Jr., MD
David S. Ettinger, MD, FACP, FCCP Winchester Hospital
Alex Grass Professor of Oncology North Reading Medical
The Sidney Kimmel Comprehensive Cancer Center at Johns North Reading, Massachusetts
Hopkins Hospital
The Johns Hopkins University Lorenzo Galluzzi, PhD
Baltimore, Maryland Assistant Professor of Cell Biology in Radiation Oncology
Weill Cornell Medical College
Lola A. Fashoyin-Aje, MD, MPH New York, New York
Medical Officer
Office of Hematology and Oncology Products Mark C. Gebhardt, MD
Center for Drug Evaluation and Research Frederick W. and Jane M. Ilfeld Professor of Orthopaedic Surgery
U.S. Food and Drug Administration Harvard Medical School
Silver Spring, Maryland Surgeon-in-Chief
Department of Orthopedic Surgery
Eric R. Fearon, MD, PhD Beth Israel Deaconess Medical Center
Maisel Professor of Oncology Orthopedic Surgeon
Professor of Internal Medicine Department of Orthopedics
University of Michigan Medical School Children’s Hospital
Ann Arbor, Michigan Boston, Massachusetts

James M. Ford, MD Daniel J. George, MD


Professor of Medicine, Pediatrics, and Genetics Professor of Medicine
Division of Oncology and Medical Genetics Duke University Medical Center
Stanford University School of Medicine Durham, North Carolina
Stanford, California
Mark B. Geyer, MD
Wilbur A. Franklin, MD Assistant Attending
Professor Emeritus of Pathology Department of Medicine
CU Anschutz Medical Campus Leukemia Service and Cellular Therapeutics Center
University of Colorado Memorial Sloan Kettering Cancer Center
Aurora, Colorado Instructor in Medicine
Joan and Sanford I. Weill Department of Medicine
Phoebe E. Freer, MD Weill Cornell Medical College
Associate Professor New York, New York
Radiology and Imaging Sciences
University of Utah Hospitals/Huntsman Cancer Institute Amato J. Giaccia, PhD
Salt Lake City, Utah Jack, Lulu, and Sam Willson Professor of Cancer Biology
Department of Radiation Oncology
Boris Freidlin, PhD Stanford University School of Medicine
Division of Cancer Treatment and Diagnosis Stanford, California
National Cancer Institute
Bethesda, Maryland Mark R. Gilbert, MD
Senior Investigator and Chief
Alison G. Freifeld, MD Neuro-Oncology Branch
Professor of Internal Medicine National Cancer Institute
Infectious Diseases Division Bethesda, Maryland
University of Nebraska Medical Center
Omaha, Nebraska Whitney Goldner, MD
Associate Professor of Internal Medicine
Terence W. Friedlander, MD Division of Diabetes, Endocrinology, and Metabolism
Associate Clinical Professor University of Nebraska Medical Center
Medicine Omaha, Nebraska
UCSF Medical Center
San Francisco, California
Contributors xv

Donald P. Goldstein, MD Missak Haigentz, MD


Professor of Obstetrics, Gynecology, and Reproductive Biology Montefiore Medical Center
Harvard Medical School Bronx, New York
Senior Scientist
Department of Obstetrics and Gynecology John D. Hainsworth, MD
Brigham and Women’s Hospital Chief Scientific Officer
Boston, Massachusetts Sarah Cannon Research Institute
Nashville, Tennessee
Annekathryn Goodman, MD
Massachusetts General Hospital Benjamin E. Haithcock, MD
Boston, Massachusetts Associate Professor of Surgery
University of North Carolina at Chapel Hill
Karyn A. Goodman, MD, MS Chapel Hill, North Carolina
Professor of Radiation Oncology
Grohne Chair in Clinical Cancer Research Christopher L. Hallemeier, MD
University of Colorado School of Medicine Assistant Professor of Radiation Oncology
Aurora, Colorado Mayo Clinic
Rochester, Minnesota
Kathleen Gordon, MD
Medical Director of Ophthalmology Samir Hanash, MD, PhD
IQVIA Evelyn & Sol Rubenstein Distinguished Chair for Cancer Prevention
Co-Chair Professor of Clinical Cancer Prevention
IQIVA Ophthalmology Center of Excellence The University of Texas MD Anderson Cancer Center
Clinical Associate Professor of Ophthalmology Houston, Texas
University of North Carolina at Chapel Hill
Chapel Hill, North Carolina Aphrothiti J. Hanrahan, PhD
Assistant Lab Member
Laura Graeff-Armas, MD, MS Human Oncology and Pathogenesis Program
Associate Professor of Internal Medicine Memorial Sloan Kettering Cancer Center
Division of Diabetes, Endocrine and Metabolism New York, New York
University of Nebraska Medical Center
Omaha, Nebraska James Harding, MD
Assistant Attending Physician
Alexander J. Greenstein, MD, MPH Memorial Sloan Kettering Cancer Center
Associate Professor of Surgery Assistant Professor of Medicine
Icahn School of Medicine at Mount Sinai Weill Medical College at Cornell University
New York, New York New York, New York

Stuart A. Grossman, MD Michael R. Harrison, MD


Professor of Oncology, Medicine, and Neurosurgery Assistant Professor of Medicine
The Sidney Kimmel Comprehensive Cancer Center at Johns Division of Medical Oncology
Hopkins Medicine Duke Cancer Institute
The Johns Hopkins University Durham, North Carolina
Baltimore, Maryland
Muneer G. Hasham, PhD
Stephan Grupp, MD, PhD Research Scientist
Section Chief, Cellular Therapy and Transplant The Jackson Laboratory
Director, Cancer Immunotherapy Frontier Program Bar Harbor, Maine
CCCR Director of Translational Research
Children’s Hospital of Philadelphia Ernest Hawk, MD, MPH
Philadelphia, Pennsylvania Boone Pickens Distinguished Chair for Early Prevention of Cancer
Vice President and Division Head
Arjun Gupta, MD Division of Cancer Prevention and Population Sciences
Assistant Instructor The University of Texas MD Anderson Cancer Center
Department of Internal Medicine Houston, Texas
University of Texas Southwestern Medical Center
Dallas, Texas Jonathan Hayman, MD
Department of Internal Medicine
Irfanullah Haider, MD, MBA Johns Hopkins Bayview Medical Center
Breast Imaging Baltimore, Maryland
Brigham and Women’s Hospital
Boston, Massachussetts
xvi Contributors

Jonathan E. Heinlen, MD Clifford A. Hudis, MD


Assistant Professor Chief Executive Officer
Department of Urology American Society of Clinical Oncology
University of Oklahoma Health Sciences Center Alexandria, Virginia
Oklahoma City, Oklahoma
Stephen P. Hunger, MD
N. Lynn Henry, MD, PhD Jeffrey E. Perelman Distinguished Chair
Associate Professor Department of Pediatrics
Internal Medicine Chief, Division of Oncology
University of Utah Pediatrics
Salt Lake City, Utah Children’s Hospital of Philadelphia
Philadelphia, Pennsylvania
Joseph Herman, MD †
Professor and Division Head ad-interim Arti Hurria, MD
Department of Radiation Oncology Professor
The University of Texas MD Anderson Cancer Center Department of Medical Oncology and Therapeutics Research
Houston, Texas City of Hope Comprehensive Cancer Center
Duarte, California
Brian P. Hobbs, PhD
Associate Staff David H. Ilson, MD, PhD
Quantitative Health Sciences and The Taussig Cancer Institute Attending Physician
Cleveland Clinic Gastrointestinal Oncology Service
Cleveland, Ohio Department of Medicine
Memorial Sloan-Kettering Cancer Center
Ingunn Holen, BSc, MSc, PhD New York, New York
Oncology
University of Sheffield Annie Im, MD
Sheffield, United Kingdom Assistant Professor of Medicine
Department of Hematology and Oncology
Leora Horn, MD, MSc UPMC Hillman Cancer Center
Associate Professor of Medicine Pittsburgh, Pennsylvania
Medicine–Hematology Oncology
Vanderbilt University
Gopa Iyer, MD
Assistant Attending Physician, Genitourinary Oncology Service
Nashville, Tennessee
Department of Medicine
Memorial Sloan Kettering Cancer Center
Neil S. Horowitz, MD
New York, New York
Department of Obstetrics and Gynecology
Division of Gynecologic Oncology
Elizabeth M. Jaffee, MD
Brigham and Women’s Hospital
The Dana and Albert “Cubby” Broccoli Professor of Oncology
Dana Farber Cancer Institute
Deputy Director, Sidney Kimmel Comprehensive Cancer Center at
Boston, Massachusetts
Johns Hopkins
Johns Hopkins University School of Medicine
Steven M. Horwitz, MD Baltimore, Maryland
Associate Attending
Department of Medicine, Lymphoma Service Reshma Jagsi, MD, DPhil
Memorial Sloan Kettering Cancer Center Professor and Deputy Chair
Assistant Professor of Clinical Medicine Radiation Oncology
Weill-Cornell Medical College University of Michigan
New York, New York Ann Arbor, Michigan

Odette Houghton, MD Rakesh K. Jain, PhD


Associate Professor A.W. Cook Professor of Tumor Biology
Department of Ophthalmology Department of Radiation Oncology
Mayo Clinic Harvard Medical School
Scottsdale, Arizona Director
E.L. Steele Laboratory for Tumor Biology
Scott C. Howard, MD, MSc Department of Radiation Oncology
Professor of Acute and Tertiary Care Massachusetts General Hospital
University of Tennessee Health Sciences Center Boston, Massachusetts
Memphis, Tennessee

Deceased.
Contributors xvii

William Jarnagin, MD, FACS Hagop Kantarjian, MD


Winchester Hospital The University of Texas MD Anderson Cancer Center
North Reading Medical Houston, Texas
North Reading, Massachusetts
Giorgos Karakousis, MD
Aminah Jatoi, MD Assistant Professor of Surgery
Professor of Oncology University of Pennsylvania
Mayo Clinic Philadelphia, Pennsylvania
Rochester, Minnesota
Maher Karam-Hage, MD
Anuja Jhingran, MD Professor of Behavioral Science
The University of Texas MD Anderson Cancer Center The University of Texas MD Anderson Cancer Center
Houston, Texas Houston, Texas

David H. Johnson, MD Nadine M. Kaskas, MD


Chairman, Department of Internal Medicine Resident Physician
University of Texas Southwestern Medical School Department of Dermatology
Dallas, Texas The Warren Alpert Medical School of Brown University
Providence, Rhode Island
Brian Johnston, MD
Royal Victoria Hospital Michael B. Kastan, MD, PhD
Belfast, United Kingdom Executive Director, Duke Cancer Institute
Director, Cancer Immunotherapy Frontier Program

Patrick G. Johnston, MD William and Jane Shingleton Professor, Pharmacology and
Center for Cancer Research and Cell Biology Cancer Biology
School of Medicine, Dentistry, and Biomedical Sciences Professor of Pediatrics
Queen’s University Belfast Duke University School of Medicine
Belfast, United Kingdom Durham, North Carolina

Kevin D. Judy, MD Nora Katabi, MD


Professor of Neurosurgery Department of Pathology
Thomas Jefferson University Memorial Sloan-Kettering Cancer Center
Jefferson Medical College New York, New York
Philadelphia, Pennsylvania
Daniel R. Kaul, MD
Lisa A. Kachnic, MD Associate Professor of Infectious Disease
Professor and Chair of Radiation Oncology University of Michigan
Vanderbilt University Medical Center Ann Arbor, Michigan
Nashville, Tennessee
Scott R. Kelley, MD, FACS, FASCRS
Orit Kaidar-Person, MD Assistant Professor of Surgery
Ramban Medical Center Division of Colon and Rectal Surgery
Haifa, Israel Mayo Clinic
Rochester, Minnesota
Sanjeeva Kalva, MD, RPVI, FSIR
Chief, Interventional Radiology Nancy Kemeny, MD
Associate Professor of Radiology Attending Physician
University of Texas Southwestern Medical Center Memorial Sloan Kettering Cancer Center
Dallas, Texas Professor of Medicine
Weill Medical College at Cornell University
Deborah Y. Kamin, RN, MS, PhD New York, New York
Vice President
Policy and Advocacy Erin E. Kent, PhD, MS
American Society of Clinical Oncology Scientific Advisor
Alexandria, Virginia Outcomes Research Branch
Healthcare Delivery Research Program
Division of Cancer Control and Population Sciences
National Cancer Institute
Rockville, Maryland
ICF, Inc.
Fairfax, Virginia

Deceased.
xviii Contributors

Oliver Kepp, PhD Daniel A. Laheru, MD


Metabolomics and Cell Biology Platforms Ian T. MacMillan Professorship in Clinical Pancreatic Research
Gustave Roussy Cancer Campus Department of Medical Oncology
Villejuif, France The Johns Hopkins University School of Medicine
Baltimore, Maryland
Simon Khagi, MD
Assistant Professor Paul F. Lambert, PhD
University of North Carolina School of Medicine Professor of Oncology
Lineberger Comprehensive Cancer Center University of Wisconsin
Chapel Hill, North Carolina Madison, Wisconsin

Joshua E. Kilgore, MD Mark Lawler, PhD


Division of Gynecologic Oncology Chair in Translational Cancer Genomics
University of North Carolina School of Medicine Centre for Cancer Research and Cell Biology
Chapel Hill, North Carolina School of Medicine, Dentistry and Biomedical Sciences
Queen’s University Belfast
D. Nathan Kim, MD, PhD Belfast, United Kingdom
Associate Professor
Department of Radiation Oncology Jennifer G. Le-Rademacher, PhD
University of Texas Southwestern Medical Center Associate Professor of Biostatistics
Dallas, Texas Health Sciences Research
Associate Professor of Oncology
Bette K. Kleinschmidt-DeMasters, MD Mayo Clinic
Professor of Neurology, Neurosurgery, and Pathology Rochester, Minnesota
CU Anschutz Medical Campus
University of Colorado John Y.K. Lee, MD
Aurora, Colorado Associate Professor of Neurosurgery
University of Pennsylvania
Edward L. Korn, PhD Philadelphia, Pennsylvania
Biometric Research Program
National Cancer Institute Nancy Y. Lee, MD
Bethesda, Maryland Department of Radiation Oncology
Memorial Sloan-Kettering Cancer Center
Guido Kroemer, MD, PhD New York, New York
Team 11, Centre de Recherche des Cordeliers
Paris, France Susanna L. Lee, MD, PhD
Massachusetts General Hospital
Geoffrey Y. Ku, MD Boston, Massachusetts
Assistant Attending Physician
Gastrointestinal Oncology Service Jonathan E. Leeman, MD
Department of Medicine Department of Radiation Oncology
Memorial Sloan Kettering Cancer Center Memorial Sloan-Kettering Cancer Center
New York, New York New York, New York

Shivaani Kummar, MD Andreas Linkermann, MD


Professor of Medicine Department of Internal Medicine III
Director, Phase 1 Clinical Research Program Division of Nephrology
Stanford University University Hospital Carl Gustav Carus at the Technische
Palo Alto, California Universität Dresden
Dresden, Germany
Bonnie Ky, MD, MSCE
Assistant Professor of Medicine and Epidemiology Jinsong Liu, MD, PhD
Division of Cardiovascular Medicine Professor of Pathology
Senior Scholar The University of Texas MD Anderson Cancer Center
Center for Clinical Epidemiology and Biostatistics Houston, Texas
University of Pennsylvania School of Medicine
Philadelphia, Pennsylvania Simon Lo, MD, FACR
Professor and Vice Chair for Strategic Planning
Department of Radiation Oncology
Professor of Neurological Surgery
University of Washington School of Medicine
Seattle, Washington
Contributors xix

Jason W. Locasale, PhD Amit Maity, MD


Associate Professor University of Pennsylvania
Department of Pharmacology and Cancer Biology Philadelphia, Pennsylvania
Duke University School of Medicine
Durham, North Carolina Neil Majithia, MD
Mayo Clinic
Charles L. Loprinzi, MD Rochester, Minnesota
Regis Professor of Breast Cancer Research
Department of Oncology Marcos Malumbres, PhD
Mayo Clinic Group Leader
Rochester, Minnesota Cell Division and Cancer
Spanish National Cancer Research Center (CNIO)
Maeve Lowery, MD Madrid, Spain
Professor of Translational Cancer Medicine
Trinity College Karen Colbert Maresso, MPH
Dublin, Ireland Program Director
Division of Cancer Prevention and Population Sciences
Emmy Ludwig, MD The University of Texas MD Anderson Cancer Center
Associate Attending Physician Houston, Texas
Memorial Sloan Kettering Cancer Center
Associate Professor of Medicine John D. Martin, PhD
Weill Medical College at Cornell University The University of Tokyo
New York, New York Tokyo, Japan

Matthew A. Lunning, DO Koji Matsuo, MD, PhD


Associate Professor of Internal Medicine Assistant Professor of Obstetrics and Gynecology
University of Nebraska Medical Center University of Southern California
Omaha, Nebraska Los Angeles, California

Robert A. Lustig, MD Natalie H. Matthews, MD


Professor of Clinical Radiation Oncology Department of Dermatology
University of Pennsylvania The Warren Alpert Medical School of Brown University
Philadelphia, Pennsylvania Providence, Rhode Island

Mitchell Machtay, MD Lauren Mauro, MD


University Hospitals Cleveland Medical Center Assistant Professor of Medicine
Case-Western Reserve University School of Medicine George Washington University School of Medicine
Cleveland, Ohio Washington, D.C.

Crystal Mackall, MD R. Samuel Mayer, MD, MEHP


Endowed Professor of Pediatrics and Medicine Associate Professor and Vice Chair for Education
Stanford University Physical Medicine and Rehabilitation
Director The Johns Hopkins University School of Medicine
Stanford Center for Cancer Cell Therapy Medical Director, Cancer Rehabilitation Program
Director Physical Medicine and Rehabilitation
Parker Institute for Cancer Immunotherapy at Stanford The Johns Hopkins Hospital
Associate Director Baltimore, Maryland
Stanford Cancer Institute
Stanford, California Worta McCaskill-Stevens, MD
Chief, Community Oncology and Prevention Trials Research Group
David A. Mahvi, MD Division of Cancer Prevention
General Surgery Resident National Cancer Institute
Brigham and Women’s Hospital Rockville, Maryland
Boston, Massachusetts
Megan A. McNamara, MD
David M. Mahvi, MD Assistant Professor of Medicine
Professor of Surgery Department of Medical Oncology
Chief, Surgical Oncology Duke University Medical Center
Medical University of South Carolina Durham, North Carolina
Charleston, South Carolina
xx Contributors

Neha Mehta-Shah, MD Jarushka Naidoo, MB, BCH


Assistant Professor Assistant Professor of Oncology
Washington University The Sidney Kimmel Comprehensive Cancer Center at Johns
St. Louis, Missouri Hopkins Hospital
Baltimore, Maryland
Robert E. Merritt, MD
Director, Thoracic Surgery Amol Narang, MD
James Cancer Center Assistant Professor of Radiation Oncology
The Ohio State University Medical Johns Hopkins University School of Medicine
Columbus, Ohio Baltimore, Maryland

Matthew I. Milowsky, MD Heidi Nelson, MD, FACS, FASCRS


Professor of Medicine Professor of Surgery
Division of Hematology/Oncology Division of Colon and Rectal Surgery
UNC Lineberger Comprehensive Cancer Center Mayo Clinic
Chapel Hill, North Carolina Rochester, Minnesota

Lori M. Minasian, MD William G. Nelson, MD, PhD


Deputy Director Professor and Director
Division of Cancer Prevention Sidney Kimmel Comprehensive Cancer Center
National Cancer Institute Johns Hopkins University School of Medicine
National Institutes of Health Baltimore, Maryland
Bethesda, Maryland
Suzanne Nesbit, PharmD, BCPS, CPE
Tara C. Mitchell, MD Clinical Pharmacy Specialist, Pain Management
Assistant Professor of Medicine Research Associate, Department of Oncology
University of Pennsylvania The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins
Philadelphia, Pennsylvania The Johns Hopkins Hospital
Baltimore, Maryland
Demytra Mitsis, MD
Medical Oncology and Hematology Fellow Mark Niglas, MD, FRCPC
Department of Medicine Clinical Fellow
Roswell Park Cancer Institute Department of Radiation Oncology
Buffalo, New York Sunnybrook Health Sciences Centre
Toronto, Ontario, Canada
Michelle Mollica, PhD, MPH, RN
Program Director Tracey O’Connor, MD
Division of Cancer Control and Population Sciences Associate Professor of Oncology
Healthcare Delivery Research Program Department of Medicine
National Cancer Institute Roswell Park Cancer Institute
Bethesda, Maryland Buffalo, New York

Margaret Mooney, MD Kenneth Offit, MD, MPH


Branch Chief, Clinical Investigations Branch Chief, Clinical Genetics Service
Cancer Therapy Evaluation Program Robert and Kate Niehaus Chair in Inherited Cancer Genomics
Division of Cancer Treatment and Diagnosis Memorial Sloan Kettering Cancer Center
National Cancer Institute New York, New York
Rockville, Maryland
Mihaela Onciu, MD
Farah Moustafa, MD Medical Director
Department of Dermatology OncoMetrix Laboratories
The Warren Alpert Medical School of Brown University Poplar Healthcare
Providence, Rhode Island Memphis, Tennessee

Lida Nabati, MD
Instructor in Medicine
Harvard Medical School
Senior Physician
Dana-Farber Cancer Institute
Boston, Massachusetts
Contributors xxi

Eileen M. O’Reilly, MD Steven Z. Pavletic, MD, MS


Winthrop Rockefeller Chair in Medical Oncology Head, Graft-Versus-Host Disease and Autoimmunity Section
Section Head Hepatopancreaticobiliary & Neuroendocrine Cancers Experimental Transplantation and Immunology Branch
Gastrointestinal Oncology Service National Cancer Institute
Associate Director Bethesda, Maryland
David M. Rubenstein Center for Pancreatic Cancer Research
Attending Physician, Member Peter C. Phillips, MD
Memorial Sloan Kettering Cancer Center Professor of Neurology and Oncology
Professor of Medicine The Children’s Hospital of Philadelphia
Weill Medical College at Cornell University Philadelphia, Pennsylvania
New York, New York
Miriam D. Post, MD
Elaine A. Ostrander, PhD Associate Professor of Pathology
Chief and Distinguished Investigator CU Anschutz Medical Campus
Cancer Genetics and Comparative Genomics Branch University of Colorado
National Human Genome Research Institute Aurora, Colorado
Bethesda, Maryland
Amy A. Pruitt, MD
Lisa Pappas-Taffer, MD University of Pennsylvania
Assistant Professor of Clinical Dermatology Philadelphia, Pennsylvania
University of Pennsylvania
Philadelphia, Pennsylvania Christiane Querfeld, MD, PhD
Chief, Division of Dermatology
Drew Pardoll, MD, PhD Director, Cutaneous Lymphoma Program
Director, Bloomberg~Kimmel Institute for Cancer Immunotherapy Assistant Professor of Dermatology
Sidney Kimmel Comprehensive Cancer Center City of Hope
Johns Hopkins School of Medicine Duarte, California
Baltimore, Maryland
Vance A. Rabius, PhD
Jae H. Park, MD Research Director, Tobacco Treatment Program
Assistant Attending The University of Texas MD Anderson Cancer Center
Department of Medicine Houston, Texas
Leukemia Service and Cellular Therapeutics Center
Memorial Sloan Kettering Cancer Center S. Vincent Rajkumar, MD
Assistant Professor of Medicine Edward W. and Betty Knight Scripps Professor of Medicine
Joan and Sanford I. Weill Department of Medicine Division of Hematology
Weill Cornell Medical College Mayo Clinic
New York, New York Rochester, Minnesota

Anery Patel, MD Mohammad O. Ramadan, MD


Clinical Instructor Assistant Professor
Department of Internal Medicine Department of Urology
Division of Diabetes, Endocrine, and Metabolism University of Oklahoma Health Sciences Center
University of Nebraska Medical Center Oklahoma City, Oklahoma
Omaha, Nebraska
Erinn B. Rankin, PhD
Anish J. Patel, MD Assistant Professor of Radiation Oncology, Obstetrics,
Assistant Professor of Endocrinology and Gynecology
Department of Endocrinology Stanford University School of Medicine
University of Alabama at Birmingham Stanford, California
Birmingham, Alabama
Sushanth Reddy, MD
Steven R. Patierno, PhD Assistant Professor of Surgery
Deputy Director Department of Surgery
Duke Cancer Institute University of Alabama at Birmingham
Professor of Medicine Birmingham, Alabama
Professor of Pharmacology and Cancer Biology
Professor of Community and Family Medicine
Duke University Medical Center
Durham, North Carolina
xxii Contributors

Michael A. Reid, PhD Nadia Rosenthal, PhD


Postdoctoral Fellow Scientific Director
Department of Pharmacology and Cancer Biology The Jackson Laboratory
Duke University School of Medicine Bar Harbor, Maine
Durham, North Carolina Chair, Cardiovascular Science
National Heart and Lung Institute
Scott Reznik, MD Imperial College London
Associate Professor London, United Kingdom
Department of Cardiothoracic Surgery
University of Texas Southwestern Medical Center Meredith Ross, MD
Dallas, Texas Fellow
Department of Internal Medicine
Tina Rizack, MD, MPH Division of Diabetes, Endocrinology, and Metabolism
Hematologist/Oncologist University of Nebraska Medical Center
South County Health Omaha, Nebraska
Clinical Assistant Professor of Internal Medicine and Obstetrics
& Gynecology Julia H. Rowland, PhD
Alpert Medical School of Brown University Director, Office of Cancer Survivorship
Providence, Rhode Island Division of Cancer Control and Population Sciences
National Cancer Institute
Jason D. Robinson, PhD Rockville, Maryland
Associate Professor of Behavioral Science
The University of Texas MD Anderson Cancer Center Anthony H. Russell, MD
Houston, Texas Massachusetts General Hospital
Boston, Massachusetts
Leslie Robinson-Bostom, MD
Senior Attending Michael S. Sabel, MD, FACS
Department of Dermatology Associate Professor
The Warren Alpert Medical School of Brown University Surgery
Providence, Rhode Island University of Michigan
Ann Arbor, Michigan
Carlos Rodriguez-Galindo, MD
Departments of Global Pediatric Medicine and Oncology Arjun Sahgal, MD, FRCPC
St. Jude Children’s Research Hospital Professor of Radiation Oncology and Surgery
Memphis, Tennessee Deputy Chief, Department of Radiation Oncology
Sunnybrook Health Sciences Center
Paul B. Romesser, MD University of Toronto Faculty of Medicine
Department of Radiation Oncology Toronto, Ontario
Memorial Sloan-Kettering Cancer Center
New York, New York Ryan D. Salinas, MD
Resident Physician
Steven T. Rosen, MD Department of Neurosurgery
Provost & Chief Scientific Officer University of Pennsylvania
Director, Comprehensive Cancer Center and Beckman Philadelphia, Pennsylvania
Research Institute
Irell & Manella Cancer Center Director’s Distinguished Chair Erin E. Salo-Mullen, MS, MPH, CGC
Helen & Morgan Chu Director’s Chair, Beckman Research Institute Senior Genetic Counselor
City of Hope Clinical Genetics Service
Duarte, California Department of Medicine
Memorial Sloan Kettering Cancer Center
Myrna R. Rosenfeld, MD, PhD New York, New York
Senior Investigator, Neuroimmunology
Institut d’Investigació Biomèdica August Pi i Sunyer (IDIBAPS) Manuel Salto-Tellez, MD
Barcelona, Spain Center for Cancer Research and Cell Biology
Adjunct Professor Neurology School of Medicine, Dentistry, and Biomedical Sciences
University of Pennsylvania Queen’s University Belfast
Philadelphia, Pennsylvania Belfast, United Kingdom
Contributors xxiii

Sydney M. Sanderson, BS Konstantin Shilo, MD


PhD Candidate Department of Pathology
Department of Pharmacology and Cancer Biology James Cancer Center
Duke University School of Medicine The Ohio State University Medical
Durham, North Carolina Columbus, Ohio

John T. Sandlund, MD Eric Small, MD


Member Professor of Medicine
Department of Oncology University of California, San Francisco
St. Jude Children’s Research Hospital San Francisco, California
Professor of Pediatrics
University of Tennessee College of Medicine Angela B. Smith, MD, MS, FACS
Memphis, Tennessee Associate Professor of Urology
Department of Urology
Victor M. Santana, MD University of North Carolina
Department of Oncology Chapel Hill, North Carolina
St. Jude Children’s Research Hospital
Memphis, Tennessee Stephen N. Snow, MD
Professor of Dermatology
Michelle Savage, MD Northwest Permanente
Department of Clinical Cancer Prevention Portland, Oregon
The University of Texas MD Anderson Cancer Center
Houston, Texas David B. Solit, MD
Geoffrey Beene Chair
Eric C. Schreiber, PhD Director, Center for Molecular Oncology
Associate Professor Member, Human Oncology and Pathogenesis Program
Department of Radiation Oncology Attending Physician, Genitourinary Oncology Service, Department
University of North Carolina School of Medicine of Medicine
Chapel Hill, North Carolina Memorial Sloan Kettering Cancer Center
New York, New York
Lynn Schuchter, MD
C. Willard Robinson Professor of Hematology and Oncology Anil K. Sood, MD
Professor of Medicine Professor and Vice Chair
University of Pennsylvania Department of Gynecologic Oncology and Reproductive Medicine
Philadelphia, Pennsylvania The University of Texas MD Anderson Cancer Center
Houston, Texas
Liora Schultz, MD
Department of Pediatric Oncology, Division of Oncology Enrique Soto-Perez-de-Celis, MD
Stanford University International Fellow
Stanford, California Department of Medical Oncology and Therapeutics Research
City of Hope
Michael V. Seiden, MD, PhD Duarte, California
Texas Oncology Researcher in Medical Science
The Woodlands, Texas Cancer Care in the Elderly Clinic
Department of Geriatrics
Morgan M. Sellers, MD, MS Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran
Icahn School of Medicine at Mount Sinai Mexico City, Mexico
New York, New York
Joseph A. Sparano, MD
Payal D. Shah, MD Associate Chairman
Assistant Professor Department of Oncology
Medicine Montefiore Medical Center
University of Pennsylvania Professor of Medicine and Women’s Health
Philadelphia, Pennsylvania Department of Medicine and Oncology
Albert Einstein College of Medicine
Jinru Shia, MD Bronx, New York
Member and Attending Pathologist
Memorial Sloan Kettering Cancer Center Vladimir S. Spiegelman, MD, PhD
Professor of Pathology and Laboratory Medicine Professor of Pediatrics and Pharmacology
Weill Medical College at Cornell University Department of Pediatrics
New York, New York Pennsylvania State University College of Medicine
Hershey, Pennsylvania
xxiv Contributors

Sheri L. Spunt, MD, MBA James E. Talmadge, PhD


Endowed Professor of Pediatric Cancer Professor of Pathology and Microbiology
Department of Pediatrics University of Nebraska Medical Center
Division of Hematology/Oncology Omaha, Nebraska
Stanford University School of Medicine
Stanford, California David T. Teachey, MD
Department of Pediatrics
Zsofia K. Stadler, MD Divisions of Hematology and Oncology
Assistant Attending Physician Children’s Hospital of Philadelphia
Department of Medicine Philadelphia, Pennsylvania
Memorial Sloan Kettering Cancer Center
Assistant Professor of Medicine Catalina V. Teba, MD
Weill Cornell Medical College University Hospitals Cleveland Medical Center
New York, New York Case-Western Reserve University School of Medicine
Cleveland, Ohio
David P. Steensma, MD
Institute Physician Ayalew Tefferi, MD
Department of Medical Oncology Department of Hematology
Dana-Farber Cancer Institute Mayo Clinic
Associate Professor of Medicine Rochester, Minnesota
Harvard Medical School
Boston, Massachusetts Bin Tean Teh, MD, PhD
Professor
Richard M. Stone, MD Division of Medical Sciences
Professor of Medicine National Cancer Centre Singapore
Harvard Medical School Professor, Cancer and Stem Cell Biology Program
Chief of Staff Duke–NUS Medical School
Department of Medical Oncology Singapore
Dana-Farber Cancer Institute
Boston, Massachusetts Joyce M.C. Teng, MD, PhD
Associate Professor of Dermatology and Pediatrics
Steven Kent Stranne, MD, JD Stanford of Medicine
Polsinelli PC Stanford University
Washington, D.C. Palo Alto, California

Kelly Stratton, MD Joel E. Tepper, MD


Assistant Professor Hector MacLean Distinguished Professor of Cancer Research
Department of Urology Department of Radiation Oncology
University of Oklahoma Health Sciences Center University of North Carolina School of Medicine
Oklahoma City, Oklahoma University of North Carolina Lineberger Comprehensive
Cancer Center
Bill Sugden, PhD Chapel Hill, North Carolina
Professor of Oncology
University of Wisconsin Premal H. Thaker, MD
Madison, Wisconsin Professor in Gynecologic Oncology
Division of Obstetrics and Gynecology
Andrew M. Swanson, MD Washington University School of Medicine
Assistant Professor of Dermatology St. Louis, Missouri
University of Wisconsin School of Medicine and Public Health
Madison, Wisconsin Aaron P. Thrift, PhD
Assistant Professor
Martin S. Tallman, MD Dan L. Duncan Comprehensive Cancer Center
Chief, Leukemia Service Department of Medicine, Gastroenterology Section
Memorial Sloan-Kettering Cancer Center Baylor College of Medicine
Professor of Medicine Houston, Texas
Joan and Sanford I. Weill Department of Medicine
Weill Cornell Medical College Arthur-Quan Tran, MD
New York, New York Division of Gynecologic Oncology
University of North Carolina School of Medicine
Chapel Hill, North Carolina
Contributors xxv

Grace Triska, MS Richard L. Wahl, MD


Washington University School of Medicine Elizabeth Mallinckrodt Professor and Director
St. Louis, Missouri Mallinckrodt Institute of Radiology
Washington University School of Medicine
Donald Trump, MD, FACP St. Louis, Missouri
Chief Executive Officer and Executive Director
Inova Schar Cancer Institute Michael F. Walsh, MD, FAAP, FACMG, DABMG
Falls Church, Virginia Assistant Member
Department of Pediatrics and Medicine
Kenneth Tsai, MD, PhD Divisions of Solid Tumor and Clinical Genetics
Associate Member Memorial Sloan Kettering Cancer Center
Anatomic Pathology and Tumor Biology New York City, New York
H. Lee Moffitt Cancer Center and Research Institute
Tampa, Florida Thomas Wang, MD
Professor of Surgery
Chia-Lin Tseng, MD, FRCPC Department of Surgery
Assistant Professor University of Alabama at Birmingham
Radiation Oncologist Birmingham, Alabama
Sunnybrook Health Sciences Centre
Toronto, Ontario, Canada Jared Weiss, MD
Associate Professor of Medicine
Diane Tseng, MD, PhD Section Chief of Thoracic and Head/Neck Oncology
Department of Medicine Division of Hematology and Oncology
Division of Oncology University of North Carolina at Chapel Hill
Stanford University Chapel Hill, North Carolina
Stanford, California
Irving L. Weissman, MD
Sandra Van Schaeybroeck, MD Director, Institute for Stem Cell Biology and Regenerative Medicine
Center for Cancer Research and Cell Biology Director, Stanford Ludwig Center for Cancer Stem Cell Research
School of Medicine, Dentistry, and Biomedical Sciences and Medicine
Queen’s University Belfast Stanford University
Belfast, United Kingdom Palo Alto, California

Brian A. Van Tine, MD, PhD Shannon N. Westin, MD, MPH


Associate Professor Associate Professor of Gynecologic Oncology and
Internal Medicine Reproductive Medicine
Washington University in Saint Louis The University of Texas MD Anderson Cancer Center
St. Louis, Missouri Houston, Texas

Erin R. Vanness, MD Jeffrey D. White, MD


Associate Professor of Dermatology Associate Director, Office of Cancer Complementary and
University of Wisconsin School of Medicine and Public Health Alternative Medicine
Madison, Wisconsin Division of Cancer Treatment and Diagnosis
National Cancer Institute
Gauri Varadhachary, MD Bethesda, Maryland
Professor
Medical Director, Gastrointestinal Center Richard Wilson, MD
Executive Medical Director, Ambulatory Operations Center for Cancer Research and Cell Biology
Department of Gastrointestinal Medical Oncology School of Medicine, Dentistry, and Biomedical Sciences
The University of Texas MD Anderson Cancer Center Queen’s University Belfast
Houston, Texas Belfast, United Kingdom

Marileila Varella-Garcia, PhD Richard J. Wong, MD


Professor of Medicine and Medical Oncology Department of Surgery
CU Anschutz Medical Campus Memorial Sloan-Kettering Cancer Center
University of Colorado New York, New York
Aurora, Colorado
xxvi Contributors

Gary S. Wood, MD Timothy Zagar, MD


Professor and Chair of Dermatology Northeastern Radiation Oncology
University of Wisconsin School of Medicine and Public Health Glens Falls, New York
Middleton VA Medical Center
Madison, Wisconsin Elaine M. Zeman, PhD
Associate Professor
Yaohui G. Xu, MD, PhD Department of Radiation Oncology
Associate Professor of Dermatology University of North Carolina School of Medicine
University of Wisconsin School of Medicine and Public Health Chapel Hill, North Carolina
Madison, Wisconsin
Tian Zhang, MD
Meng Xu-Welliver, MD, PhD Assistant Professor of Medicine
Associate Professor of Radiation Oncology Duke University Medical Center
James Cancer Center Durham, North Carolina
The Ohio State University Medical Center
Columbus, Ohio James A. Zwiebel, MD
Cancer Therapy Evaluation Program (Retired)
Shlomit Yust-Katz, MD National Cancer Institute
Professor Rockville, Maryland
Davidoff Cancer Center
Rabin Medical Center
Petah Tikva, Israel
Preface

New insights into whole genome sequence variations and the genomic useful to students and trainees, experts in the various disciplines of
structural alterations associated with cancer, including their downstream oncology, and as a reference text for physicians from other medical
effects on protein structure and function, are helping us to define specific disciplines and the various staff who regularly care for patients with
communication pathway changes that drive cancer initiation, progression, cancer. It is our hope that readers will find this scholarly textbook
metastasis, and resistance. We have learned that each individual and properly balanced between the disciplines of science, clinical medicine,
each tumor may be unique. Individual physiognomies in terms of path and humanism and that it will serve them well in their efforts to prevent,
of progression and unique cellular communication pathway alterations diagnose, and effectively treat their patients suffering from cancer.
are continuing to define the nature of specific cancers and offer greater The multidisciplinary nature of cancer care is, and will continue to
opportunities for the development of highly prescriptive intervention(s). be, reflected in our editors. Experts in cancer biology, surgical oncology,
In addition, we have a much greater understanding of the relationship pediatric oncology, radiation oncology, medical oncology, and hema-
of the host’s tissues, the patient’s immune system, and the broad tumor tologic malignancies directed the development of the content. Reflecting
microenvironment, to the process of tumor development and progression the multispecialty approach necessary for optimal care of patients, the
and their impact on tumor control. This new body of knowledge on majority of chapters are the joint product of several of these disciplines.
how the body’s immune system and the tumor’s microenvironment Engaging the very best subject matter authorities was a guiding principle
are altered to support disease growth, invasion, and distant spread is for the editors and we are deeply indebted to our outstanding authors
providing opportunities for the development of novel therapeutic who, in a most diligent and thoughtful way, have brought their knowledge
interventions. There is exciting new evidence to support the presence and skills to the sixth edition of Abeloff ’s Clinical Oncology.
of a special subclass of cells within the tumor that has properties of
“stemness,” which places them in the key role of maintaining tumor
ACKNOWLEDGMENTS
growth and tumor spread. The cumulative effect of these advances—
where certain cancers can be prevented and where others will be detected This sixth edition represents a highly collaborative and dynamic effort
earlier and controlled—promises to be transformative in our effort to between the editors and Elsevier. We are greatly indebted to Laura
conquer cancer. Schmidt, Kathleen Schlom, and Kristi Batchelor for their creative input
The sixth edition of Abeloff ’s Clinical Oncology incorporates the and guidance and for turning the principles behind this text into a
exciting advances in basic, translational, clinical, and epidemiologic reality. Finally, we want to express our gratitude to our many contributing
oncology. Each chapter begins with a summary highlighting the key authors for their dedication to this project, their generosity of time,
points and comprises a critical analysis of the literature and updated and, of course, their very valuable friendship.
clinical studies—authors present their own opinions in specially identified
boxes and algorithms. John E. Niederhuber, MD
Despite significant progress, the diagnosis of cancer remains devastat- James O. Armitage, MD
ing to patients and their families. Our goal is to provide a reference James H. Doroshow, MD
textbook that is the most useful, understandable, attractive, and thorough Michael B. Kastan, MD, PhD
in presenting the principles of clinical oncology. It is meant to be equally Joel E. Tepper, MD

xxvii
This page intentionally left blank
Contents

12 Viruses and Human Cancer,  165


Part I Science and Clinical Oncology Paul F. Lambert and Bill Sugden
Section A Biology and Cancer
13 Genetic Factors: Hereditary Cancer Predisposition
Syndromes, 180
1 Molecular Tools in Cancer Research,  2 Michael F. Walsh, Karen Cadoo,
Mauro W. Costa, Muneer G. Hasham, Erin E. Salo-Mullen, Marianne Dubard-Gault,
and Nadia Rosenthal Zsofia K. Stadler, and Kenneth Offit

2 Intracellular Signaling, 24 14 Genetic and Epigenetic Alterations in


Aphrothiti J. Hanrahan, Gopa Iyer, Cancer, 209
and David B. Solit Bin Tean Teh and Eric R. Fearon

3 Cellular Microenvironment and Metastases,  47 Section C Diagnosis of Cancer


Erinn B. Rankin and Amato J. Giaccia
15 Pathology, Biomarkers, and Molecular
4 Control of the Cell Cycle,  56 Diagnostics, 225
Marcos Malumbres Wilbur A. Franklin, Dara L. Aisner, Kurtis D. Davies,
Kristy Crooks, Miriam D. Post, Bette K.
5 Pathophysiology of Cancer Cell Death,  74 Kleinschmidt-DeMasters, Edward Ashwood,
Lorenzo Galluzzi, Andreas Linkermann, Oliver Kepp, Paul A. Bunn, and Marileila Varella-Garcia
and Guido Kroemer
16 Imaging, 254
6 Cancer Immunology, 84 Richard L. Wahl
Diane Tseng, Liora Schultz, Drew Pardoll,
and Crystal Mackall Section D Clinical Trials

7 Stem Cells, Cell Differentiation, and Cancer,  97 17 Biostatistics and Bioinformatics in Clinical
Piero Dalerba, Maximilian Diehn, Irving L. Weissman, Trials, 284
and Michael F. Clarke Brian P. Hobbs, Donald A. Berry,
and Kevin R. Coombes
8 Tumor Microenvironment: Vascular and
Extravascular Compartment,  108 18 Clinical Trial Designs in Oncology,  296
Rakesh K. Jain, John D. Martin, Vikash P. Chauhan, Edward L. Korn and Boris Freidlin
and Dan G. Duda 19 Structures Supporting Cancer Clinical Trials,  308
Jeffrey S. Abrams, Margaret Mooney,
9 Cancer Metabolism, 127 James A. Zwiebel, Worta McCaskill-Stevens,
Michael A. Reid, Sydney M. Sanderson, Michaele C. Christian, and James H. Doroshow
and Jason W. Locasale
20 Oncology and Health Care Policy,  317
Section B Genesis of Cancer Steven Kent Stranne, Clifford A. Hudis,
and Deborah Y. Kamin
10 Environmental Factors,  139
Steven R. Patierno Section E Prevention and Early Detection
11 DNA Damage Response Pathways and 21 Discovery and Characterization of Cancer Genetic
Cancer, 154 Susceptibility Alleles,  323
James M. Ford and Michael B. Kastan Stephen J. Chanock and Elaine A. Ostrander

xxix
xxx Contents

22 Lifestyle and Cancer Prevention,  337 Section B Symptom Management


Karen Basen-Engquist, Powel Brown,
Adriana M. Coletta, Michelle Savage, 35 Hypercalcemia, 565
Karen Colbert Maresso, and Ernest Hawk Anery Patel, Laura Graeff-Armas, Meredith Ross,
and Whitney Goldner
23 Screening and Early Detection,  375
Therese Bevers, Hashem El-Serag, Samir Hanash, 36 Tumor Lysis Syndrome,  572
Aaron P. Thrift, Kenneth Tsai, Scott C. Howard
Karen Colbert Maresso, and Ernest Hawk
37 Cancer-Related Pain,  581
24 Nicotine Dependence: Current Treatments and Suzanne Nesbit, Ilene Browner,
Future Directions,  399 and Stuart A. Grossman
Jeffrey M. Engelmann, Maher Karam-Hage,
Vance A. Rabius, Jason D. Robinson, 38 Cancer Cachexia,  593
and Paul M. Cinciripini Jennifer G. Le-Rademacher and Aminah Jatoi

39 Nausea and Vomiting,  598


Section F Treatment John D. Hainsworth
25 Cancer Pharmacology,  411 Section C Treatment Complications
Jerry M. Collins
40 Oral Complications,  607
26 Therapeutic Targeting of Cancer Cells: Era of Neil Majithia, Christopher L. Hallemeier,
Molecularly Targeted Agents,  420 and Charles L. Loprinzi
Khanh T. Do and Shivaani Kummar
41 Dermatologic Toxicities of Anticancer
27 Basics of Radiation Therapy,  431 Therapy, 621
Elaine M. Zeman, Eric C. Schreiber, Natalie H. Matthews, Farah Moustafa,
and Joel E. Tepper Nadine M. Kaskas, Leslie Robinson-Bostom,
and Lisa Pappas-Taffer
28 Hematopoietic Stem Cell Transplantation,  461
Annie Im and Steven Z. Pavletic 42 Cardiovascular Effects of Cancer Therapy,  649
Lori M. Minasian, Myrtle Davis, and Bonnie Ky
29 Gene Therapy in Oncology,  470
James E. Talmadge and Kenneth H. Cowan 43 Reproductive Complications,  665
Demytra Mitsis, Lynda Kwon Beaupin,
30 Therapeutic Antibodies and Immunologic and Tracey O’Connor
Conjugates, 486
Konstantin Dobrenkov and Nai-Kong V. Cheung 44 Paraneoplastic Neurologic Syndromes,  676
Josep Dalmau and Myrna R. Rosenfeld
31 Complementary and Alternative Medicine,  500
Jeffrey D. White 45 Neurologic Complications,  688
Shlomit Yust-Katz, Simon Khagi,
and Mark R. Gilbert

46 Endocrine Complications,  707


Part II Problems Common to Cancer Donald Trump
and Therapy 47 Pulmonary Complications of Anticancer
Section A Hematologic Problems and Infections Treatment, 715
Mitchell Machtay and Catalina V. Teba

32 Disorders of Blood Cell Production in Clinical Section D Posttreatment Considerations


Oncology, 514
Jennifer H. Choe and Jeffrey Crawford 48 Rehabilitation of Individuals With Cancer,  725
R. Samuel Mayer
33 Diagnosis, Treatment, and Prevention of
Cancer-Associated Thrombosis,  523 49 Survivorship, 732
Claudia I. Chapuy and Jean M. Connors Julia H. Rowland, Michelle Mollica, and Erin E. Kent

34 Infection in the Patient With Cancer,  544 50 Second Malignant Neoplasms,  741
Alison G. Freifeld and Daniel R. Kaul Debra L. Friedman
Contents xxxi

51 Caring for Patients at the End of Life,  751 Section B Head, Neck, and Eye
Lida Nabati and Janet L. Abrahm
64 Ocular Tumors,  968
Section E Local Effects of Cancer and Its Metastasis Odette Houghton and Kathleen Gordon

52 Acute Abdomen, Bowel Obstruction, 65 Cancer of the Head and Neck,  999
and Fistula,  764 Jonathan E. Leeman, Nora Katabi,
William R. Burns and Alfred E. Chang Richard J. Wong, Nancy Y. Lee,
and Paul B. Romesser
53 Superior Vena Cava Syndrome,  775
Arjun Gupta, D. Nathan Kim, Sanjeeva Kalva, Section C Skin
Scott Reznik, and David H. Johnson
66 Melanoma, 1034
54 Spinal Cord Compression,  786 Tara C. Mitchell, Giorgos Karakousis,
Mark Niglas, Chia-Lin Tseng, Nicolas Dea,
and Lynn Schuchter
Eric Chang, Simon Lo, and Arjun Sahgal

55 Brain Metastases and Neoplastic Meningitis,  794 67 Nonmelanoma Skin Cancers: Basal Cell and
Orit Kaidar-Person, Michael Cools, Squamous Cell Carcinomas,  1052
and Timothy Zagar Yaohui G. Xu, Juliet L. Aylward,
Andrew M. Swanson, Vladimir S. Spiegelman,
56 Bone Metastases,  809 Erin R. Vanness, Joyce M.C. Teng,
Robert E. Coleman, Janet Brown, and Ingunn Holen Stephen N. Snow, and Gary S. Wood

57 Lung Metastases,  831 Section D Endocrine


Jonathan Hayman, Jarushka Naidoo,
and David S. Ettinger 68 Cancer of the Endocrine System,  1074
Ammar Asban, Anish J. Patel, Sushanth Reddy,
58 Liver Metastases,  846 Thomas Wang, Courtney J. Balentine,
David A. Mahvi and David M. Mahvi and Herbert Chen

59 Malignancy-Related Effusions,  863


Lola A. Fashoyin-Aje and Julie R. Brahmer
Section E Thoracic

69 Cancer of the Lung: Non–Small Cell Lung Cancer


Section F Special Populations and Small Cell Lung Cancer,  1108
Luiz H. Araujo, Leora Horn, Robert E. Merritt,
60 Cancer in the Elderly: Biology, Prevention, Konstantin Shilo, Meng Xu-Welliver,
and Treatment,  874 and David P. Carbone
Enrique Soto-Perez-de-Celis and Arti Hurria†

61 Special Issues in Pregnancy,  882 70 Diseases of the Pleura and Mediastinum,  1159
Orit Kaidar-Person, Timothy Zagar,
Tina Rizack and Jorge J. Castillo
Benjamin E. Haithcock, and Jared Weiss
62 Human Immunodeficiency Virus (HIV) Infection
and Cancer,  894 71 Cancer of the Esophagus,  1174
Jesus Anampa, Stefan K. Barta, Missak Haigentz, Geoffrey Y. Ku and David H. Ilson
and Joseph A. Sparano
Section F Gastrointestinal

Part III Specific Malignancies 72 Cancer of the Stomach,  1197


Section A Central Nervous System Geoffrey Y. Ku and David H. Ilson

73 Cancer of the Small Bowel,  1211


63 Cancer of the Central Nervous System,  906 Morgan M. Sellers and Alexander J. Greenstein
Jay F. Dorsey, Ryan D. Salinas, Mai Dang,
Michelle Alonso-Basanta, Kevin D. Judy, 74 Colorectal Cancer,  1219
Amit Maity, Robert A. Lustig, John Y.K. Lee, Mark Lawler, Brian Johnston,
Peter C. Phillips, and Amy A. Pruitt Sandra Van Schaeybroeck, Manuel Salto-Tellez,
Richard Wilson, Malcolm Dunlop,

Deceased. and †Patrick G. Johnston
xxxii Contents

75 Cancer of the Rectum,  1281 87 Gestational Trophoblastic Disease,  1544


Scott R. Kelley and Heidi Nelson Donald P. Goldstein, Ross S. Berkowitz,
and Neil S. Horowitz
76 Cancer of the Anal Canal,  1300
Karyn A. Goodman, Lisa A. Kachnic, 88 Cancer of the Breast,  1560
and Brian G. Czito N. Lynn Henry, Payal D. Shah, Irfanullah Haider,
Phoebe E. Freer, Reshma Jagsi,
77 Liver and Bile Duct Cancer,  1314 and Michael S. Sabel
Ghassan K. Abou-Alfa, William Jarnagin,
Imane El Dika, Michael D’Angelica, Maeve Lowery, Section I Sarcomas
Karen Brown, Emmy Ludwig, Nancy Kemeny,
Anne Covey, Christopher H. Crane, James Harding, 89 Sarcomas of Bone,  1604
Jinru Shia, and Eileen M. O’Reilly Megan E. Anderson, Steven G. DuBois,
and Mark C. Gebhardt
78 Carcinoma of the Pancreas,  1342
Ana De Jesus-Acosta, Amol Narang, 90 Sarcomas of Soft Tissue,  1655
Lauren Mauro, Joseph Herman, Brian A. Van Tine
Elizabeth M. Jaffee, and Daniel A. Laheru
Section J Cancer of Undefined Site of Origin
Section G Genitourinary
91 Carcinoma of Unknown Primary,  1694
79 Cancer of the Kidney,  1361 Gauri Varadhachary and James L. Abbruzzese
Megan A. McNamara, Tian Zhang,
Michael R. Harrison, and Daniel J. George Section K Pediatrics
80 Carcinoma of the Bladder,  1382 92 Pediatric Solid Tumors,  1703
Angela B. Smith, Arjun V. Balar, Jeffrey S. Dome, Carlos Rodriguez-Galindo,
Matthew I. Milowsky, and Ronald C. Chen Sheri L. Spunt, and Victor M. Santana

81 Prostate Cancer,  1401 93 Childhood Leukemia,  1748


William G. Nelson, Emmanuel S. Stephen P. Hunger, David T. Teachey,
Antonarakis, H. Ballentine Carter, Stephan Grupp, and Richard Aplenc
Angelo M. DeMarzo and Theodore L. DeWeese
94 Childhood Lymphoma,  1765
82 Cancer of the Penis,  1433 John T. Sandlund and Mihaela Onciu
Jonathan E. Heinlen, Mohammad O. Ramadan,
Kelly Stratton, and Daniel J. Culkin Section L Hematological

83 Testicular Cancer,  1442 95 Acute Leukemias in Adults,  1783


Terence W. Friedlander and Eric Small Frederick R. Appelbaum

Section H Gynecological 96 Myelodysplastic Syndromes,  1798


David P. Steensma and Richard M. Stone
84 Cancers of the Cervix, Vulva, and Vagina,  1468 97 Myeloproliferative Neoplasms,  1821
Anuja Jhingran, Anthony H. Russell, Ayalew Tefferi
Michael V. Seiden, Linda R. Duska,
Annekathryn Goodman, Susanna L. Lee, 98 Chronic Myeloid Leukemia,  1836
Subba R. Digumarthy, and Arlan F. Fuller, Jr. Hagop Kantarjian and Jorge Cortes

85 Uterine Cancer,  1508 99 Chronic Lymphocytic Leukemia,  1850


John F. Boggess, Joshua E. Kilgore, Farrukh T. Awan and John C. Byrd
and Arthur-Quan Tran
100 Hairy Cell Leukemia,  1872
86 Carcinoma of the Ovaries and Fallopian Mark B. Geyer, Omar Abdel-Wahab,
Tubes, 1525 Martin S. Tallman, and Jae H. Park
Robert L. Coleman, Jinsong Liu, Koji Matsuo,
Premal H. Thaker, Shannon N. Westin, 101 Multiple Myeloma and Related Disorders,  1884
and Anil K. Sood S. Vincent Rajkumar and Angela Dispenzieri
Contents xxxiii

102 Hodgkin Lymphoma,  1911 105 Adult T-Cell Leukemia/Lymphoma,  1965


Nancy Bartlett and Grace Triska Matthew A. Lunning, Neha Mehta-Shah,
and Steven M. Horwitz
103 Non-Hodgkin Lymphomas,  1926
Jeremy S. Abramson Index 1975

104 Cutaneous T-Cell Lymphoma and Cutaneous


B-Cell Lymphoma,  1948
Christiane Querfeld, Steven T. Rosen,
and Madeleine Duvic
This page intentionally left blank
P A R T
I

SCIENCE AND CLINICAL ONCOLOGY


A. BIOLOGY AND CANCER 13. Genetic Factors: Hereditary 22. Lifestyle and Cancer Prevention
Cancer Predisposition
1. Molecular Tools in Cancer 23. Screening and Early Detection
Syndromes
Research
24. Nicotine Dependence: Current
14. Genetic and Epigenetic
2. Intracellular Signaling Treatments and Future
Alterations in Cancer
Directions
3. Cellular Microenvironment and
Metastases C. DIAGNOSIS OF CANCER
F. TREATMENT
15. Pathology, Biomarkers, and
4. Control of the Cell Cycle 25. Cancer Pharmacology
Molecular Diagnostics
5. Pathophysiology of Cancer 26. Therapeutic Targeting of
16. Imaging
Cell Death Cancer Cells: Era of
D. CLINICAL TRIALS Molecularly Targeted Agents
6. Cancer Immunology
17. Biostatistics and Bioinformatics 27. Basics of Radiation Therapy
7. Stem Cells, Cell Differentiation, in Clinical Trials
and Cancer 28. Hematopoietic Stem Cell
18. Clinical Trial Designs in Transplantation
8. Tumor Microenvironment: Oncology
Vascular and Extravascular 29. Gene Therapy in Oncology
Compartment 19. Structures Supporting Cancer
Clinical Trials 30. Therapeutic Antibodies and
9. Cancer Metabolism Immunologic Conjugates
20. Oncology and Health Care
B. GENESIS OF CANCER Policy 31. Complementary and Alternative
Medicine
10. Environmental Factors
E. PREVENTION AND EARLY
11. DNA Damage Response DETECTION
Pathways and Cancer 21. Discovery and Characterization
12. Viruses and Human of Cancer Genetic
Cancer Susceptibility Alleles
A. BIOLOGY AND CANCER
1  Molecular Tools in Cancer Research
Mauro W. Costa, Muneer G. Hasham, and Nadia Rosenthal

S UMMARY OF K EY P OI N T S
• Our understanding and treatment of growth and differentiation has techniques, including whole-genome
cancer have always relied heavily on revolutionized the diagnosis, analysis, expression profiling, and
parallel developments in biologic prognosis, and treatment of refined genetic manipulation in and
research. Molecular biology provides malignant disorders. use of genetically diverse animal
the basic tools to study genes • This introductory chapter relates models, providing the conceptual
involved with cancer growth patterns basic principles of molecular biology and technical background necessary
and tumor suppression. An to emerging perspectives on the to grasp the central principles and
advanced understanding of the origin and progression of cancer and new methods of current cancer
molecular processes governing cell explains newly developed laboratory research.

Since the last edition of this book was published, advances in our genetic changes confer selective advantages on tumor cell clones by
understanding of the basic mechanisms of cancer have continued to disrupting control of cell proliferation. The identification of specific
inform and refine clinical approaches to prevention and therapy. New mutations that characterize a tumor cell has proved invaluable for
prognostic and predictive markers derived from molecular biology analyzing the neoplastic progression and remission of the disease. The
can now pinpoint specific genetic changes in particular tumors or emergence of cancer cells is a byproduct of the necessity for continuous
detect occult malignant cells in normal tissues, leading to improved cell division and DNA replication to maintain organ functionality
technologies for tumor screening and early detection. Diagnostic throughout the life cycle.
approaches have expanded from morphologic criteria and single-gene The highly heterogeneous nature of tumors, each composed of
analysis to whole-genome technologies and single-cell genomics multiple cell types, led to the formulation of the “cancer stem cell”
imported from other biologic disciplines. A new systemic vision of hypothesis, which posits that only a subpopulation of cancer cells is
cancer is emerging, in which the importance of individual mutation able to maintain self-renewal, unlimited growth, and capacity for
has been superseded by an appreciation for higher-order organization differentiation into other, more specialized cancer cell types. Cancer
and individual genetic background, disrupted by complex interactions stem cells display bona fide stem cell markers, in contrast to other
of disease-associated factors and gene-environmental parameters that cancer cells present in the tumor, which do not have tumorigenic
affect tumor cell behavior. Results from these cross-disciplinary potential. In fact, fewer than 1 in 10,000 cells present in human acute
investigations underscore the complexity of carcinogenesis and have myeloid leukemia are capable of reinitiating a new tumor when
profoundly influenced the design of strategies for both cancer prevention transplanted into animals. Cancer stem cells have been identified in
and advanced cancer therapy. many solid tumors in the brain, colon, ovaries, prostate, and pancreas,
This overview will serve as a foundation of conceptual and technical suggesting that more effective cancer therapies would target these
information for understanding the exciting new advances in cancer self-renewing cells, rather than the tumor as a whole. The cancer stem
research described in subsequent chapters. Since the discovery of cell concept differs from the original clonal evolution hypothesis,
oncogenes, which provided the first concrete evidence of cancer’s which states that every cell in a tumor mass is capable of self-renewal
genetic basis, applications of advanced molecular techniques and and differentiation, and suggests that detecting and targeting subtle
instrumentation have yielded new insights into normal cell biology. genetic and epigenetic differences that distinguish cancer stem cells
A basic fluency in molecular biology and genetics has become a necessary may provide a more effective avenue to intervention in disease
prerequisite for clinical oncologists because many of the new diagnostic progression.
and prognostic tools currently in use rely on these fundamental Heterogeneity can also arise as a result of stochastic mutational
principles of gene, protein, and cell function. events that lead to cancer progression. Clastogenic insults to the genome,
or genomic instability due to aberrant gene regulation, could lead to
OUR UNSTABLE HEREDITY loss of heterozygosity (LOH) of tumor suppressor genes such as TP53,
RB1, or BRCA, and can also lead to tumor heterogeneity and change
Cancer genetics has classically relied on the candidate-gene approach, in disease progression. Furthermore, activation of DNA or RNA editing
detecting acquired or inherited changes in specific genetic loci accu- enzymes in tumors could lead to kataegis, a DNA hypermutation
mulated in a single cell, which then proliferates to produce a tumor process, and increase tumor heterogeneity. Although there are molecular
composed of its identical clonal progeny. During the early steps of biology tools currently available to detect aberrant but stable genomes,
tumor formation, mutations that lead to an intrinsic genetic instability the later processes that lead to genomic instability make diagnosis
allow additional deleterious genetic alterations to accumulate. These and prognosis more challenging.

2
Molecular Tools in Cancer Research  •  CHAPTER 1 3

DETECTING CANCER MUTATIONS The functional unit of inherited information in DNA, the gene,
is most often represented by a discrete section of sequence necessary
Methods for mutation detection all rely on the manipulation of DNA, to encode a particular protein structure. Gene expression is initiated
the basic building block of heredity in the cell. DNA consists of two by forming a copy of the gene, messenger RNA (mRNA), constructed
long strands of polynucleotides that twist around each other clockwise base by base from the DNA template by a polymerase enzyme. Once
in a double helix (Fig. 1.1). Nucleic acid bases attached to the sugar transcribed, an mRNA transcript is modified and the processed product
groups of each strand face each other within the helix, perpendicular is transported out of the nucleus. In the cytoplasm, proteins are
to its axis. These comprise only four bases: the purines adenine and then synthesized, or translated, in macromolecular complexes called
guanine (A and G) and the pyrimidines cytosine and thymine (C and ribosomes that read the mRNA sequence and convert the nucleic acid
T). During assembly of the double helix, stable pairings of nucleotides code, based on three-base segments or codons, into a 20–amino acid
from either strand are made between A and T, or between G and C. code to form the corresponding protein.
Each base pair forms one of the billions of rungs in the long, unbroken Although these canonic processes drive gene expression in all normal
ladder of DNA forming a chromosome. cells, cancer cells defy the rules. For instance, uracils, which are found

Genes Cell nucleus


containing
23 pairs of
chromosomes

DNA
strand
Chromosomes
Sugar

Cytosine Bases
thymine

Bases

Adenine and
guanine
Phosphate
group P

H
H
C H
H
H C
H
P
H
H
P
H
C
H
H CH2
P P
H
CH2
H
CH2
H
P P
H
CH2
H CH2

Figure 1.1  •  DNA structure. Deoxyribonucleic acid (DNA) is the cell’s genetic material, contained in single compacted strands comprising chromosomes
within the cell nucleus. In the DNA double helix, the two intertwined components of its backbone, composed of sugar (deoxyribose) and phosphate molecules,
are connected by pairs of molecules called bases. The sequence of four bases (guanine, adenine, thymine, and cytosine) in the DNA helix determines the specificity
of genetic information. The bases face inward from the sugar-phosphate backbone and form pairs with complementary bases on the opposing strand for
specific recognition. The arrangement of chemical groups is unique for each base pair, allowing base pairs to be specifically targeted by transcription factors,
polymerases, restriction enzymes, and other DNA-binding proteins. (From http://www.terrapsych.com/dna.jpg.)
4 Part I: Science and Clinical Oncology

on RNA, can be detected in the DNA of cancer cells because of their is shared by all members of a species, the recent sequencing of thousands
high mutation rates. Paradoxically, these deviations from the norm of individual human genomes has given rise to the new field of
allow the development of molecular biology tools to better diagnose genomics, providing us with new tools to reveal the more subtle
and predict tumor progression. variations that arise between individuals. These variations are critical,
both as a natural engine driving heterogeneity within a species, and
GENERATING DIVERSITY WITH as a source of predisposition to cancer types. The most common forms
ALTERNATE SPLICING of human genetic variations, or alleles, arise as single-nucleotide
polymorphisms (SNPs). Because these allelic dissimilarities are abundant,
In higher organisms, most protein coding gene sequences are interrupted inherited, and dispersed throughout the genome, SNPs can be used
by stretches of noncoding DNA sequences, called introns. In the to track racial diversity, personal traits, and susceptibility to common
nucleus, these introns are removed after mRNA transcription to produce forms of cancer (Fig. 1.3). Commercial entities have developed tools
a continuous chain of coding sequences, or exons, that subsequently that can detect thousands of SNPs with relatively little sample material.
undergo translation into protein. The splicing process requires absolute Platforms such as MegaMUGA or GigaMUGA can allow mammalian
precision because the deletion or addition of a single nucleotide at genetic mapping that can aid in a number of diagnoses and can
the splice junction would throw the three-base coding sequence out distinguish between predictive and prognostic markers.
of frame, or lead to exon skipping or addition, creating abnormal How do SNPs arise between individuals? One source of variation
proteins. in DNA sequence derives from deviations in the strict base-pairing
The dramatic increase in genetic complexity conferred by alternate rule underlying the structure, storage, retrieval, and transfer of genetic
RNA splicing is underscored by the multiple splice patterns of many information. The duplicated genetic information in the two strands
medically relevant genes, in which different combinations of exons of DNA not only permits the repair of a damaged coding sequence
are chosen for the final mRNA transcript, such that one gene can but also forms the basis for the replication of DNA. During cell
encode many different proteins (Fig. 1.2). The choice of protein isoform division, polymerase enzymes unwind the DNA strands and copy
to be expressed from a gene with multiple splicing possibilities is a them, using the base sequences as a template for constructing a new
decision that can be perturbed in disease. Errors in splicing mechanisms helix so that the dividing cell passes its entire genetic content on to
have been associated with a large group of cancers. These include its progeny. Errors in this process are rare, and person-to-person
mutations in the oncogene p53 in more then 12 different types of differences comprise only about 0.1% of the human genome. SNPs
cancer, mutL homolog 1 protein (MLH1) mutation in hereditary are inherited if they occur in the germline. Many genetically inherited
nonpolyposis colorectal cancer, and several transcription factors and variations occur in regions that do not encode protein or alter the
cell signaling and membrane proteins. When mutations in the splicing regulation of nearby genes. Given the disruptive effects even subtle
site lead to insertion of novel sequences in the mRNA, the encoded genetic changes may have on cell function, it is important to distinguish
protein can be used as a potential clinical marker, as seen for the SNPs that represent true mutations from benign polymorphisms.
transcription factor NSFR in small cell lung cancer. Owing to their Our ability to monitor hundreds of thousands of SNPs simultane-
unique expression in cancer cells, these markers can be further explored ously is one of the most important advances in modern medical genetics.
as new cancer-specific therapeutic targets. Relatively simple genotyping technologies for SNP detection rely largely
on the polymerase chain reaction (PCR). In procedures that use this
GENOMICS OF CANCER reaction, two chemically synthesized single-stranded DNA fragments,
or primers, are designed to match chromosomal DNA sequences
The complete set of DNA sequences carried on all the chromosomes flanking the segment in which an SNP is positioned. With the addition
is known as the genome. Although the general map of the genome of nucleotide building blocks and a heat-stable DNA polymerase, the

Exon Exon Exon Exon Exon


Gene 1 2 3 4 5

1 2 3 4 5
RNA

Alternative splicing

RNA 1 2 3 4 5 1 2 4 5 1 2 3 5

Translation Translation Translation

Protein A Protein B Protein C

Figure 1.2  •  RNA splicing. Alternate splicing produces multiple related proteins, or isoforms, from a single gene. (From Guttmacher AE, Collins F.
Genomic medicine—a primer. N Engl J Med. 2002;347:1512–1520.)
Molecular Tools in Cancer Research  •  CHAPTER 1 5

Primary tumor Metastasis SNP density


11p15.5
11p15.4
11p15.3
11p15.2
11p15.1
11p14.3
11p14.2
11p14.1
11p13
11p12
Tens to hundreds
11p11.2
of changes between >10 million SNPs 11p11.12
primary and secondary between individuals 11p11.11
tumors 11q11
11q12.1
11q12.2
11q12.3
11q13.1
11q13.2
11q13.3
Primary tumor Metastasis 11q13.4
11q13.5
11q14.1
11q14.2
11q14.3
11q21
11q22.1
11q22.2
11q22.3
11q23.1
11q23.2
11q23.3
11q24.1
11q24.2
11q24.3
11q25

Figure 1.3  •  Determining cancer susceptibility with single-nucleotide polymorphisms (SNPs). Millions of SNPs exist between individuals, as depicted by
the red arrows and the SNP density map of human chromosome 11 (right). By contrast, point mutations, deletions, insertions, and rearrangements between
normal tissues and tumors or between primary and secondary tumors probably number in the tens to hundreds (or potentially thousands), as depicted by the
spectral karyotype image at the bottom of the figure. Because the constitutional genetic polymorphisms are present in all the tissues of the body, it might be
possible to distinguish differences in metastatic versus nonmetastatic tumors and in nontumor tissues before they ever happen to develop a solid tumor. (From
Hunter K. Host genetics influence tumour metastasis. Nat Rev Cancer. 2006;6:141–146.)

primer pairs, or amplicons, initiate synthesis of new DNA strands, base sequence that carries unique protein coding information. Other
using the chromosomal material as a template. Each successive copying noncoding DNA sequences are used for directing the transcription
cycle, initiated by “melting” the resulting double-stranded products of neighboring genes, through complex regulatory circuits involving
with heat, doubles the number of DNA segments in the reaction (Fig. protein binding and modification of the DNA itself, or shifting of
1.4). The technique is exceptionally sensitive; millions of identical its chromosomal packaging. Although genomic instability is generally
DNA copies can be generated in a matter of hours with PCR by using considered a consequence of tumor formation rather than the initial
a single DNA molecule as the starting material. trigger of cancer, the loss, gain, or rearrangement of chromosomal
Other novel methods for large-scale SNP detection include single- segments through deletion or translocation is a common form of
nucleotide primer extension, allele-specific hybridization, oligonucleotide neoplastic mutation, as protein coding segments from different genes
ligation assay, and invasive signal amplification, which detect poly- are combined or regulatory sequences are brought into new proximity
morphisms directly from genomic DNA without the requirement of to genes they do not normally control, as seen in chronic myeloid
PCR amplification. The International HapMap Project was established leukemia (CML). In CML, recombination events lead to the fusion
with the objective of identifying those variations (commonly thought of BCR and ABL genes (Philadelphia chromosome). This results in
to be on the order of 10 million in our genome) in the human popula- constitutive activation of the fused gene, leading to loss of proliferative
tion. This project is already in its third phase (HapMap3), now control in myeloid cells and consequently cancer. Gross changes in
including both SNPs and copy number variations observed in 1184 DNA arrangement can be detected by cytogenetic analysis of chro-
samples from 11 different human populations. Regardless of the method mosomal features on metaphase spreads. Although fluorescence in
used to characterize them, the collective SNPs in a selected genomic situ hybridization (FISH) provides greater resolution by localizing
region characterize a haplotype, or specific combination of alleles at specific chromosomal DNA sequences corresponding to fluorescently
multiple linked genetic loci along a chromosome that are inherited labeled probes (Fig. 1.5), and can be used to track specific alterations
together. in chromosomal structure where known genes are involved, spectral
Even when the SNPs within a given haplotype are not directly karyotyping (SKY) is a powerful and more general tool that could
involved in a disease, they provide markers for clonality and for the aid diagnosis of cancer genomes. With each fluorescently labeled
loss or rearrangement of specific chromosomal segments in growing chromosome assigned a specific color, translocations and additions
tumors. In the human nucleus, each of the 23 tightly compacted are revealed as multicolored chromosomes, or large deletions as pieces
chromosomes has a characteristic size and structure, and a distinctive of missing chromosomes.
6 Part I: Science and Clinical Oncology

Cycle 1 Cycle 2 Cycle 3 The plethora of data arising from genome-wide association studies
using currently available techniques poses particular challenges to
cancer researchers. Discerning the causal genetic variants among
genotype-phenotype associations requires extensive replication, control
for underlying genetic differences in population cohorts, and consistent
classification of clinical outcomes. New technologies must be met
Separation of strands

Separation of strands

Separation of strands
with equivalently sophisticated and rigorous analytic methodologies
for the true genetic cause of cancer to be teased out from our variable
and often unstable heredity.
Primers
Sequence
to be BUILDING GENE LIBRARIES
amplified
The engineering of genes by recombinant DNA technology evolved
from methods initially devised to provide sequences in amounts
sufficient for biochemical analysis. The original protocol involves
clipping the desired segment from the surrounding DNA and inserting
it into a bacterial or viral vector, which is then amplified millions of
times in a host bacterium. Using recombinant DNA technology, genetic
Primers Heat Heat Heat engineering can routinely produce industrial quantities of pure, clinically
useful products in a cost-effective way. For diagnostic purposes, it is
easier and faster to amplify a known genomic DNA sequence directly
Figure 1.4  •  Amplification of DNA by polymerase chain reaction (PCR). from a patient sample with PCR, but the classic approach is still
The DNA sequence to be amplified is selected by primers, which are short, applied to the construction of recombinant DNA libraries.
synthetic oligonucleotides that correspond to sequences flanking the DNA to To be useful, a DNA library must be as complete as possible, with
be amplified. After an excess of primers is added to the DNA, together with recombinant members, or clones, sufficiently numerous to include
a heat-stable DNA polymerase, the strands of both the genomic DNA and all the sequences in an individual genome. For certain kinds of gene-
the primers are separated by heating and allowed to cool. A heat-stable
polymerase elongates the primers on either strand, thus generating two new,
linkage analysis that require long, uninterrupted stretches of DNA,
identical double-stranded DNA molecules and doubling the number of DNA special vectors, such as bacterial or yeast artificial chromosomes, can
fragments. Each cycle takes just a few minutes and doubles the number of carry foreign DNA fragments of enormous lengths. Chromosomal
copies of the original DNA fragment. segments represented in genomic DNA libraries can contain the

9 9 22 22

9 der(9) 22 der(22)

Figure 1.5  •  Detection of chromosomal translocations. Fluorescence in situ hybridization (FISH) technology uses a labeled DNA segment as a probe to
search homologous sequences in interphase chromosomes for the t(9;22)(q34;q11) translocation, associated with chronic myeloid leukemia. On the left, patient
nuclei were hybridized with probes for chromosome 9 (labeled with SpectrumRed fluorophore) and chromosome 22 (labeled with SpectrumGreen). (Modified
from Varella-Garcia M. Molecular cytogenetics in solid tumors: laboratorial tool for diagnosis, prognosis, and therapy. Oncologist. 2003;8:45–58.)
Molecular Tools in Cancer Research  •  CHAPTER 1 7

structure of an entire gene, including the information that regulates The interaction between protein and DNA is increasingly
its expression, and formed the starting material for sequencing of the used to identify transcription factor binding sites in a regulatory
human genome. region. Whereas electrophoretic mobility shift assays (EMSAs), or
Many cancer-associated genes were originally identified through DNA footprinting, were once standard techniques for determining
use of partial DNA libraries, which contain only the DNA sequences protein-DNA interactions, emerging genome-wide technologies, such
transcribed by a particular tissue or type of cell. The starting mate- as chromatin immunoprecipitation on microarray chip (ChIP-chip)
rial in this case is mRNA. For cloning purposes, the enzyme reverse and chromatin immunoprecipitation on sequencing (ChIP-seq), are
transcriptase can convert mRNA into complementary DNA (cDNA). revolutionizing the way in which we see the interaction of a transcription
The number of clones in a cDNA library is much smaller than factor complex with virtually all of its potential genomic targets in
in a genomic library because a cDNA library represents only the a particular cell state. These strategies involve the use of candidate
genes expressed by the tissue of interest and contains exclusively the protein–specific antibodies to pull down DNA targets regulated by
coding portion of genes. For this particular reason, this technique them. These targets are further identified with the use of microarray
has now become obsolete for organisms whose genome has now ChIP-chip or next generation sequencing ChIP-seq technologies
been fully sequenced. New advances in PCR chemistry allowed (see Fig. 1.14).
for the direct cloning of increasingly larger cDNA fragments with Our appreciation of oncogenic perturbations, by mutation of
high specificity and low error rates. Highly accurate PCR technol- regulatory protein coding genes or by loss of controlled signaling by
ogy, coupled with the constantly evolving generation of genomic cell cycle switches or in the target sequences these proteins recognize,
sequence maps in humans and model organisms, has exponentially has recently extended to include posttranslational modifications that
expanded the availability of candidate genes to be tested in cancer control protein activity, such as phosphorylation, ubiquitylation, and
biology. SUMOylation. Tumor-associated changes in these modifications
underscore the multiple levels of control necessary to ensure correct
LOSING CONTROL OF THE GENOME gene expression that is so central to the normal function of the cell.

Mutations that lead to oncogenic transformation of a cell invariably EPIGENETICS AND CANCER
affect the expression of its genetic information that specifies functional
products, either RNA molecules or proteins used for various cellular Epigenetics refers to general control of gene expression that is inherited
functions. The primary level of gene control is the transcription of during cell division, although not part of the DNA sequence itself.
DNA into RNA. Gene regulation, or the control of RNA synthesis, Epigenetic regulation involves changes in chromatin, a higher-order
represents a complex process that is itself a frequent target of neoplastic building block of chromosomes that wraps DNA into coils with
mutation. scaffolding proteins such as histones. Histones are a necessary com-
DNA regulatory sequences do not encode a product. However, ponent of chromosomal compaction, but also play a critical role in
without them a cell could not coordinate the expression of the hundreds gene accessibility (Fig. 1.7). Active genetic loci are associated with
of thousands of genes in its nucleus, select only certain genes for loosely configured euchromatin, whereas silent loci are condensed in
expression, and activate or repress them in response to precise internal heterochromatin. The state of chromatin configuration (euchromatin
or external signals. These control centers of the genome contain binding or heterochromatin) both controls and is controlled by patterns of
sites for multiple proteins, called transcription factors, which interact histone modifications such as methylation and acetylation on specific
to form regulatory networks controlling gene transcription. Their DNA sequences. This pattern relates the underlying genetic information
function can be altered by signals that induce modifications such as to its higher-order structure that determines whether a particular gene
phosphorylation, or by interactions with other regulators such as steroid regulatory element is available to transcription factors (on or off status).
hormones. Many of the cell’s responses to a wide variety of external These epigenetic modifications of the nuclear environment that
stimuli, such as neurotransmitters, antigens, cytokines, and growth determine the accessibility of a gene can persist during cell division,
factors, are mediated through transcription factors binding to DNA because inherited epigenetic patterns provide permanent marks for
regulatory sequences. altered chromatin configuration in daughter cells. The pattern of
Certain regulatory DNA sequences common to many genes are modifications generated by the epigenetic code rivals the complexity
positioned upstream of the transcription start site (Fig. 1.6). Collectively of the DNA code itself.
called the “promoter” of a gene, these proximal sequences comprise The accessibility of genomic regions can favor mutations. Enzymes
binding sites for the RNA polymerase and its numerous cofactors. such as the APOBEC family exploit this accessibility to induce C to
Whereas the position of the promoter with regard to the transcription U mutation, which is then converted to T or staggered single-strand
start site is relatively inflexible, other DNA regulatory elements, known breaks. If not rectified, these point mutations or breaks can lead to
as enhancers, occur in unpredictable locations, often at a considerable hypermutations. Kataegis is an example wherein such hypermutation
distance from the genes they control. Some transcription factors bind occurs on the BRCA locus, generating neoplasia.
to particular regions of enhancers and drive their associated genes in Research has linked rearrangement of chromatin and associated
many types of cells, whereas others, active in only a limited variety DNA methylation with the inactivation of tumor suppressor genes and
of cells, maintain a tissue-specific pattern of gene expression. Enhancers neoplastic transformation. Defects that could lead to cancer involve
are often responsible for the aberrant expression of genes induced by perturbations in the “epigenotype” of a particular locus, through the
chromosomal translocation associated with specific forms of cancer: silencing of normally active genes or activation of normally silent
a normally quiescent gene promoting cell growth that is dislocated genes, associated with changes in DNA methylation, histone modifica-
to a position near a strong enhancer may be activated inappropriately, tion, and chromatin proteins (Fig. 1.8). Changes in the number or
resulting in loss of growth control. density of heterochromatin proteins associated with cancer-related
Enhancers and promoters have been assigned specific roles by means genes such as EZH2, or of euchromatic proteins such as trithorax in
of cell culture assays or in transgenic animals in which putative regula- leukemia, can also be associated with abnormal patterns of methyla-
tory DNA sequences are linked to test or “reporter” genes, and are tion in gene promoter regions and with higher-order chromosomal
examined for their ability to activate expression of the reporter gene structures that are only beginning to be understood. Finally, it is
in response to the appropriate signals. Through assessment of the increasingly evident that interactions among the “epigenome,” the
effects of deleting, adding, or changing DNA sequences within the genome, and the environment are common targets for mutation
regulatory element, the precise nucleotides that are critical for recogni- and can have profound effects on the gene expression readout of a
tion by transcription factors can be determined. cancer cell.
8 Part I: Science and Clinical Oncology

Gene structure
Exon 1 Exon 2 Exon 3
Enhancer Promoter Intron 1 Intron 2

TATAA GT AG GT AG AATAAA

Transcription Gene expression


factors

RNA polymerase
Exon 1 Exon 2 Exon 3

Transcription
Transcription-initiation pre-
complex 5' 3' mRNA
Transcript processing

RNA-clipping enzyme

AAUAAA

5' cap

PolyA tail
AAAA...

Adenosine-adding
Intron lariat enzyme (terminal
transferase)
Splicing
Nucleus AAAA...

Spliceosome

Processed
transcript mRNA
Cytoplasm AAAA...

Translation into protein

Figure 1.6  •  Mammalian gene structure and expression. The DNA sequences that are transcribed as RNA are collectively called the gene and include exons
(expressed sequences) and introns (intervening sequences). Introns invariably begin with the nucleotide sequence GT and end with AG. An AT-rich sequence
in the last exon forms a signal for processing the end of the RNA transcript. Regulatory sequences that make up the promoter and include the TATA box
occur close to the site where transcription starts. Enhancer sequences are located at variable distances from the gene. Gene expression begins with the binding
of multiple protein factors to enhancer sequences and promoter sequences. These factors help form the transcription-initiation complex, which includes the
enzyme RNA polymerase and multiple polymerase-associated proteins. The primary transcript (pre-mRNA) includes both exon and intron sequences. Post-
transcriptional processing begins with changes at both ends of the RNA transcript. At the 5′ end, enzymes add a special nucleotide cap; at the 3′ end, an
enzyme clips the pre-mRNA about 30 base pairs (bp) after the AAUAAA sequence in the last exon. Another enzyme adds a polyA tail, which consists of up
to 200 adenine nucleotides. Next, spliceosomes remove the introns by cutting the RNA at the boundaries between exons and introns. The process of excision
forms lariats of the intron sequences. The spliced mRNA is now mature and can leave the nucleus for protein translation in the cytoplasm. (From Rosenthal
N. Regulation of gene expression. N Engl J Med. 1994;331:931–932.)
Molecular Tools in Cancer Research  •  CHAPTER 1 9

Nucleosome

DNA

The solenoid

Figure 1.7  •  Chromatin packaging of DNA. The 4 meters of DNA in every human cell must be compressed in the nucleus, reaching compaction ratios
of 1 : 400,000. This is achieved by wrapping the DNA (blue) around histone protein complexes (green), forming nucleosomes connected by a thread of free
linker DNA. Each nucleosome, together with its linker, packages about 200 bp (66 nm) of DNA. The nucleosomes are then coiled into chromatin, a rope of
nucleoprotein about 30 nm thick (bottom left electron micrograph). To allow DNA to be accessed by transcription and replication apparatus, chromatin is relaxed
(bottom right electron micrograph). (Courtesy Jakob Waterborg. www.umkc.edu/sbs/waterborg/chromat/chromatn.html. Copyright 1998 Jakob Waterborg.)

Gene X Gene X
X

Gene Y
X

Gene Y

A Normal B Epigenetic lesions


Figure 1.8  •  Gene accessibility through epigenetics. Illustration depicts known and possible defects in the epigenome that could lead to disease. (A) Gene
X is a transcriptionally active gene with sparse DNA methylation (brown circles), an open chromatin structure, interaction with euchromatin proteins (green
protein complex), and histone modifications such as H3K9 acetylation and H3K4 methylation (green circles). Gene Y is a transcriptionally silent gene with
dense DNA methylation, a closed chromatin structure, interaction with heterochromatin proteins (red protein complex), and histone modifications such as
H3K27 methylation (pink circles). (B) The abnormal cell could switch its epigenotype through the silencing of normally active genes or activation of normally
silent genes, with the attendant changes in DNA methylation, histone modification, and chromatin proteins. In addition, the epigenetic lesion could include
a change in the number or density of heterochromatin proteins in gene X (such as EZH2 in cancer) or euchromatic proteins in gene Y (such as trithorax in
leukemia). There may also be an abnormally dense pattern of methylation in gene promoters (shown in gene X ), and an overall reduction in DNA methylation
(shown in gene Y ) in cancer. The insets show that the higher-order loop configuration may be altered, although such structures are currently only beginning
to be understood.
10 Part I: Science and Clinical Oncology

PROFILING TUMORS underlying tumor progression by following the changes in a tumor


cell’s transcriptional landscape.
Monitoring global gene expression patterns of cells represents one of With reliance on two-color fluorescence-based microarray technology
the latest breakthroughs in developing a molecular taxonomy of cancer. (DNA microarray), simultaneous evaluation of thousands of gene
Although classic blotting and probe hybridization techniques (Northern transcripts and their relative expression can provide a snapshot of the
blot) are still a reliable way to monitor expression of individual genes, “transcriptome,” the full complement of RNA transcripts produced
these techniques have limitations, such as unequal hybridization at a specific time during the progression of malignancy.
efficiency of individual probes, sensitivity for low copy or small Transcriptional profiling with microarrays typically involves screens
transcripts, and difficulty in detecting multiple RNAs simultaneously of mRNA expression from two sources (such as tumor and normal
or in simultaneously analyzing a large number of targets. For cancer cells), using cDNA or oligonucleotide libraries that are arranged in
studies, it is important to be able to compare the expression pattern extremely high density on microchips. These are probed with a mixture
of all known RNAs, including noncoding RNAs, between cancer cells of fluorescently tagged cDNAs generated from the tumor and normal
and normal cells. Thus new genome-wide analytic techniques are the samples, which results in differential staining of each gene spot. The
state-of-the-art choice to detect mRNA expression profiles at a single relative intensity of the two different colors reflects the RNA expres-
point in time or cell state. Genome-wide profiling of gene expression sion level of each gene; this is analyzed with a laser confocal scanner
in tumors delivers an unprecedented view into the biologic processes (Fig. 1.9). With microarrays, single genes that constitute diagnostic,

Tumors
Reference Tumor
RNA RNA

Genes

Statistical
cDNA
analysis
Hybridization
of probe to
microarray Multidimensional-scaling plot

A B C

Donor Recipient
paraffin block paraffin block
D E Tissue microarray

Figure 1.9  •  Microarray-based expression profiling of tumor tissue. (A) Reference RNA and tumor RNA are labeled by reverse transcription with different
fluorescent dyes (green for the reference cells and red for the tumor cells) and hybridized to a cDNA microarray containing robotically printed cDNA clones.
(B) The slides are scanned with a confocal laser scanning microscope, and color images are generated with RNA from the tumor and reference cells for each
hybridization. Genes upregulated in the tumors appear red, whereas those with decreased expression appear green. Genes with similar levels of expression in
the two samples appear yellow. Genes of interest are selected on the basis of the differences in the level of expression by known tumor classes (e.g., BRCA1-
mutation–positive and BRCA2-mutation–positive). Statistical analysis determines whether these differences in the gene expression profiles are greater than
would be expected to occur by chance. (C) The differences in the patterns of gene expression between tumor classes can be portrayed in the form of a
color-coded plot, and the relations between tumors can be portrayed in the form of a multidimensional-scaling plot. Tumors with similar gene-expression
profiles cluster close to one another in the multidimensional-scaling plot. (D) Particular genes of interest can be further studied through the use of a large
number of arrayed, paraffin-embedded tumor specimens, referred to as tissue microarrays. (E) Immunohistochemical analyses of hundreds or thousands of
these arrayed biopsy specimens can be performed in order to extend the microarray findings. (From Hedenfalk I, Duggan D, Chen Y, et al. Gene expression
profiles in hereditary breast cancer. N Engl J Med. 2001;344:539–548.)
Molecular Tools in Cancer Research  •  CHAPTER 1 11

prognostic, or therapeutically relevant markers can be systematically technique relies on the generation of small fragments of cDNA from
monitored. Alternatively, the entire set of expressed genes can be any RNA sample, followed by sequencing of these expressed tags from
collectively analyzed through use of powerful statistical methods to one end (single-end sequencing) or both ends (pair-end sequencing),
classify tumors according to their transcriptional profile. Microarray resulting in fragments of 30 to 400 base pairs (bp). The resulting
analysis has already dramatically improved our ability to explore the sequences can be then mapped against the known reference genome
genetic changes associated with cancer etiology and development and or transcriptome of a certain species. Unlike microarray analysis of
is providing new tools for disease diagnosis and prognostic assessment. preselected gene sets, RNA-seq allows the unbiased identification of
For example, DNA microarray analysis of multiple primary breast all genes, or even the presence of different isoforms, expressed in the
tumor transcriptomes has revealed reproducible signature expression of sample, allowing a comprehensive comparison of transcript levels
70 associated genes. These markers have been recently cleared by the between normal and cancer cells.
US Food and Drug Administration (FDA) for PCR–based diagnostics The technologies just described can also be applied to the analysis
showing that expression analysis of a relative small gene group can of noncoding RNA species. In addition to the 20,000 protein coding
predict the prognosis of early stage breast cancers. When applied on a transcripts used to classify a wide variety of human tumors, hundreds
larger scale, these assays can predict response to chemotherapy, or opti- if not thousands of small, noncoding interference RNA species, with
mize pharmaceutical intervention by targeting therapeutic approaches critical functions in multiple biologic processes, have been discovered;
to specific patient populations and ultimately to individualized therapy. many of these RNA species are directly or indirectly involved in the
A novel high-throughput approach for global transcriptome analysis control of cell proliferation. Known as microRNAs (miRNAs), these
has been made possible by advances in strategies that allow mass short transcripts arise from primary genome-encoded transcripts of
sequencing of DNA fragments. With this technique, called RNA-seq, variable sizes that are processed into 70- to 100-nucleotide hairpin-
it is now possible to obtain a comprehensive and unbiased analysis shaped precursors, which are processed into mature miRNAs of 21 to
of all mRNA transcripts present in cells or tissues. (Fig. 1.10). The 23 bp RNA molecules (Fig. 1.11). miRNAs function by base-pairing

2x poly (A) RPKM RPKM


selection
Brain 0.0 0.0

Liver 0.0 0.0

25-bp
reads
Add standards
and shatter RNA Muscle 0.5 50.1

Make cDNA
and sequence Muscle splices

1 kb
Map 25-bp tags RNA-Seq
onto genome graph
Myf5
Myf6
Conservation

25-bpm Uniquely mappable


Calculate splices
transcript
Repeating elements by RepeatMasker
prevalence
Myf6
Conservation 20 kb
Uniquely map.
2 RPKM 1 RPKM 1 RPKM RepeatMasker
A B C

Figure 1.10  •  Methods for high-throughput transcriptome analyses. (A) Schematics of regular protocol for RNA-seq sample preparation, showing poly-A
tail specific mRNA isolation followed by fragmentation of RNA into smaller regions, further used for cDNA conversion. Polymerase chain reaction (PCR)
fragments are then tethered by adaptors, sequenced by synthesis, and aligned to the reference genome or transcriptome to calculate relative prevalence of
mRNAs (RPKM). (B) Target fragments can be used to map exon-intron boundaries and thus infer present and quantify different mRNA isoforms in the
sample of interest, as shown for the muscle specific gene Myf6 in this example. (C) Data generated with this method can also be compared with analysis of
other tissues or samples, allowing assessment of relative quantification of targets, as exemplified here for a highly specific gene (red peaks) for muscle samples.
(From Mortazavi A, Williams BA, et al. Mapping and quantifying mammalian transcriptomes by RNA-seq. Nat Methods. 2008;5:621–628.)
12 Part I: Science and Clinical Oncology

Protein-coding gene MicroRNA gene

Transcription of pri-microRNA
Transcription
of mRNA

Pri-microRNA
OR

Drosha

DGCR8
Processing
Nucleus
of pri-microRNAs
into pre-microRNA

Pre-microRNA Ran-GTP

Exportin 5
Transport of
pre-microRNAs into
the cytoplasm

Processing of
Dicer pre-microRNA into
small RNA duplexes
Loqs/TRBP
Cytoplasm
RISC

||||||||||||||||||||
||||
|||
|||
|||
||| ||||||||||||||||||||||||||||| An
||| |||||||||
||| ||||||
||| |||| |||||
|||| |||||
|||||||||

Delivery of
RISC-microRNA
complex

mRNA degradation Translational repression

Figure 1.11  •  MicroRNA production and gene regulation in animal cells. Mature functional microRNAs of approximately 22 nucleotides are generated
from long primary microRNA (pri-microRNA) transcripts. First, the pri-microRNAs, which usually contain a few hundred to a few thousand base pairs, are
processed in the nucleus into stem-loop precursors (pre-microRNA) of approximately 70 nucleotides by the RNase III endonuclease Drosha and DiGeorge
syndrome critical region gene 8 (DGCR8). The pre-microRNAs are then actively transported into the cytoplasm by exportin 5 and Ran-GTP and further
processed into small RNA duplexes of approximately 22 nucleotides by the Dicer RNase III enzyme and its partner Loqacious (Loqs), a homologue of the
human immunodeficiency virus transactivating response RNA-binding protein. The functional strand of the microRNA duplex is then loaded into the
RNA-induced silencing complex (RISC). Finally, the microRNA guides the RISC to the target messenger RNA (mRNA) target for translational repression or
degradation of mRNA. (Modified from Chen CZ. New Eng J Med. 2005;353:1768–71.)

with target mRNAs to inhibit translation and/or promote mRNA suppression. The usefulness of monitoring the expression of miRNAs
degradation. In the context of cancer, miRNAs may act in concert with in human cancer is just now being explored, but preliminary findings
other effectors such as p53 to inhibit inappropriate cell proliferation. reveal an extraordinary level of diversity in miRNA expression across
A global decrease in miRNA levels is often observed in human cancers, cancers, and the large amount of diagnostic information encoded
indicating that small RNAs may have an intrinsic function in tumor in a relatively small number of miRNAs. Significant technologic
Molecular Tools in Cancer Research  •  CHAPTER 1 13

advances facilitating the profiling of the miRNA expression patterns CANCER PROTEOME
in normal and cancer tissues hint at the unexpected greater reliability of
miRNA expression signatures than the respective signatures of protein The term proteome describes the entire complement of proteins expressed
coding genes in classifying cancer types. Along with their potential by the genome of a cell, tissue, or organism. More specifically, it is
diagnostic value, miRNAs are also being tested for their prognostic used to describe the set of all the expressed proteins at a given time
use in predicting clinical behaviors of cancer patients. point in a defined setting, such as a tumor. Like RNA transcription,
Because probe specificity in miRNA microarray analysis is prob- the synthesis of proteins is a highly regulated process that contributes
lematic owing to the small target size, hybridization can be performed to the specific proteome of a particular cell and can be perturbed in
first in solution, and then quantified with multicolor flow sorting. diseases such as cancer.
Real-time PCR can also be used to quantify specific miRNA sets, or Advances in protein analytic techniques over the last decades have
to capture a more detailed picture of their changing expression profiles progressed to the point that even small numbers of specific proteins
in tumor progression. Identification of the miRNAs involved in tumor expressed in tissues can be used to predict the prognosis of a cancer.
pathogenesis and elucidation of their action in a specific cancer will The improvement of protein-based assays has made it possible to
be the next necessary steps for their manipulation in a therapeutic identify and examine the expression of most proteins, and to envision
setting. large-scale protein analysis on the level of gene-based screens. Various
Advances in this field have revealed that miRNAs are also involved systematic methodologies have contributed to the current explosion
in cancer initiation and progression, and specific modulation of of information on the proteome. These are now being compared for
such RNAs may serve as a therapeutic strategy. Inhibition of key their suitability as platforms for the generation of databases on protein
miRNAs using antagomirs (a class of chemically modified anti-miRNA structural features, interaction maps, activity profiles, and regulatory
oligonucleotides) has been effective in suppressing tumor growth in modifications.
mouse models. It remains to be seen if these results can be extended to The yeast two-hybrid system has been a popular genetics-based
treatment of cancer in the clinic, but interference with miRNA function approach for detecting protein-protein interactions inside a cell (Fig.
is an attractive new tool for the development of cancer therapies. 1.12). One protein fused to the DNA binding domain (bait) and a

DNA-binding domain
fused to protein A A

A Promoter Reporter gene

Activator region fused to


protein B
B

B Promoter Reporter gene


Figure 1.12  •  Exploring protein-protein interactions with the yeast two-hybrid
system. Two-hybrid technology exploits the fact that transcriptional activators are Activator region fused to
modular in nature. Two physically distinct functional domains are necessary to get protein B
transcription: a DNA-binding domain that binds to the DNA of the promoter and DNA-binding domain
an activation domain that binds to the basal transcription apparatus and activates fused to protein A A B Transcription
transcription. (A) The known gene encoding protein A is cloned into the “bait”
vector, fused to the gene encoding a DNA-binding domain from some transcription
factor. When placed into a yeast system with a reporter gene, this fusion protein
can bind to the reporter gene promoter, but it cannot activate transcription. C Promoter Reporter gene
(B) Separately, a second gene (or a library of cDNA fragments encoding potential
interactors), protein B, is cloned into the “prey” vector, fused to an activation domain
of a different transcription factor. When placed into a yeast strain containing the 96 Bait strains 1 Prey strain
reporter gene, it cannot activate transcription because it has no DNA-binding domain.
(C) When the two vectors are placed into the same yeast, a transcription factor is
formed that can activate the reporter gene if protein B, made by the second plasmid,
binds to protein A. (D) Screening a yeast two-hybrid library. The plate on the left
holds 96 different yeast strains in patches (or colonies), each of which expresses a
different bait protein (top). The plate on the right holds 96 patches, each of the
same yeast strain (prey strain) that expresses a protein fused to an activation domain
(prey). The plate of bait strains and the plate of prey strains are pressed to the same
replica velvet, and the impression is lifted with a plate containing yeast extract
peptone dextrose (YPD) medium. After 1 day of growth on the YPD plate, during
which time the two strains mate to form diploids, the YPD plate is pressed to a Replicate velvet
new replica velvet, and the impression is lifted with a plate containing diploid Diploids
selection medium and an indicator such as X-Gal. Blue patches (dark spots) on the
X-Gal plate indicate that the lacZ reporter is transcribed, suggesting that the prey YPD
interacts with the bait at that location. (C from http://www.nature.com/…/journal/
v403/n6770/full/403601a0_r.html. D from Bartel PL, Fields S, eds. The Yeast Replicate velvet
Two-Hybrid System. New York: Oxford University Press; 1997; Finley RL Jr, Brent
R: Two-hybrid analysis of genetic regulatory networks. Retrieved from http://www. D X-Gal
genetics.wayne.edu/finlab/YTHnetworks.html.)
14 Part I: Science and Clinical Oncology

different protein fused to the activation domain of a transcriptional as phosphorylation, SUMOylation, or ubiquitination. These LC-MS/
activator (prey) are expressed together in yeast cells. If the bait MS systems, such as the iTRAQ, allow for a more precise and indi-
and prey interact, transcription of a reported gene is induced and vidualized diagnosis of cancer.
detected typically by a color reaction that reflects the transactiva- Monoclonal antibodies (mAbs) have been a cornerstone of protein
tion of the reporter gene, and by proxy, the interaction of the two analysis in cancer research, and more recently have risen to prominence
test proteins. The method can also be used for large-scale protein as cancer therapeutics based on their exquisite specificity for protein
interactions, determination of RNA-protein interactions, and protein- targets and their potent interference with protein function. Novel
ligand binding. strategies have been developed that target not only antigens highly
As a complementary proteomics tool, mass spectrometry (MS) is expressed in cancer cells but also to enhance the innate immune
an accurate mass measurement of charged peptides isolated by two- response against cancer cells. These antibodies can act via several
dimensional gel electrophoresis, producing a mass-to-charge ratio of mechanisms, including antibody-dependent cellular cytotoxicity
charged samples under vacuum that can be used to determine the (ADCC), complement-mediated cytotoxicity (CMC), and antibody-
sequence identity of peptides. Combined with a specific proteolytic dependent cellular phagocytosis (ADCP) (Fig. 1.13). Laboratory mice
cleavage step, mass spectroscopy can be used for peptide mass mapping. have been the animal model of choice for generating a ready source
Automation of this process has made mass spectroscopy the analytic of diverse, high-affinity and high-specificity mAbs; however, the use
tool of choice for many proteomics projects. For diagnostic purposes, of rodent antibodies as therapeutic agents has been restricted by the
liquid chromatography and mass spectrometry (LC-MS/MS) have inherent immunogenicity of mouse proteins in a human setting. The
been combined to detect not only a single–amino acid change in the more recent application of transgenic mouse technology to introduce
whole proteome, but also posttranslational protein modification such variable regions encoded by human sequences into the corresponding

Signal Bispecific
BiTE
perturbation Toxin
cytotoxicity

cytotoxic cells
Direct

Nonrestricted
activation of
Radionuclide

Drug
TrioMab

Tumor cell
CMC death
MAC

inhibitory signaling
Fc-mediated immune
effector engagement

Blockade of
ADCC

Phagocytosis

ADCP

APC Helper T cell Cytotoxic T cell


IC uptake MHC class II presentation MHC class I cross-presentation
Introduction of adaptive immune responses

Antigen BiTE MHC TCR T cell


class I Phagocytic
APC
Monoclonal Immunotoxin MHC Fc receptor Tumor cell
antibody class II
Perforin and
Bispecific Compliment Innate granzymes
CD3 KIR
antibody (C1q) effector

Figure 1.13  •  Mechanisms for antibody-based therapies used against cancer cells. Multiple current approaches involve direct cytotoxicity, Fc-mediated
immune effector engagement, nonrestricted activation of cytotoxic T cells, and blockade of inhibitory signaling. The diverse spectrum of action of these therapies
will allow the inclusion of various anticancer targets in the near future (From Weiner LM, Murray JC, Shuptrine CW. Antibody-based immunotherapy of
cancer. Cell. 2012;148:1081–1084.)
Molecular Tools in Cancer Research  •  CHAPTER 1 15

mouse immunoglobulin genes has enabled the generation of “human- being developed that have increased specificity toward individualized
ized” therapeutic mAbs with reduced immunogenicity. In addition, cancers.
bispecific antibodies (bsAbs) with dual affinity for tumor antigens, From an epigenetic perspective, new techniques are enabling the
such as TriomAb, have been shown to effectively kill tumor cells by genome-wide characterization of protein-DNA interactions that can
inducing memory T-cell protective immunity. In addition to the uncover novel transcription factor targets, histone modifications,
expected use of mAbs directed at extracellular epitopes (protein regions and DNA methylation patterns within a cancer cell. Combining
recognized by the antibody), evidence from mouse models has raised chromatin immunoprecipitation (ChIP) with microarray (ChIP-chip)
the possibility of using antibodies targeting intracellular epitopes for allows genome-wide screening for the binding position of protein
anticancer therapies. Targeting such antigens would enrich immuno- factors to their gene targets. In ChIP-chip assays or ChIP-seq, a
therapy, allowing the use of tumor-specific intracellular mediators of cross-linking reagent is applied in vivo to proteins associated with
cell survival and proliferation. Numerous mAb-based agents are currently DNA in the nucleus, which then can be coimmunoprecipitated
in trial or in use as therapeutics for cancer, and the potential for with specific antibodies to the protein under analysis. The bound
further optimization of mAbs through genetic engineering promises DNA and appropriate controls are then fluorescently labeled and
to open new avenues for in vivo therapy. applied to microscopic slides for microarray analysis, or directly
A recent advancement in mAb-based cancer therapy is the generation sequenced, rendering a simultaneous profile of all the binding posi-
of chimeric antigen receptor (CAR) T lymphocytes to target tumors tions of specific proteins in the cancer cell’s genome (Fig. 1.14). The
in vivo. These are effector T-lymphocytes engineered to express a mAb global profiling of promoter occupancy of specific cancers, wherein
that recognizes specific groups of cancer cells. The receptors are chimeric, protein-DNA interaction profiles discriminate patients with tumors
composed of engineered molecules from diverse sources. The first from those presenting different clinical outcomes, is a promising
generation of CAR-modified T cells (CAR T cells) showed success in predictive method.
preclinical trials and have entered phase I clinical trials in ovarian After a decade of development, proteomics is still primarily a
cancer, neuroblastoma, and various types of leukemia and lymphoma. basic research activity, yet in the near future this technology is likely
Newer generations of these therapeutic lymphocytes are currently to have a profound impact on medicine. By defining the collective

Analysis and visualization Peak calling Alignment

Chr1:13456-13486
Chr1:24323-24293
2
Chr1:45678-45708 Sequencing
Chr1:54321-54351
Information content

1.5
Chr1:55679-55709
1
etc...
0.5
ChlP-Seq 0
1 2 3 4 5 6 7 8 9 10
Position

Library
construction

Antibody
Binding sites

TF TF

105
4
P < 0.001
log2 enrichment

20Kb

104
Cy3 intensity

0
–1
1000
ChlP-on-chip 200bp
Binding sites

ChlP replicate 1
100
Peak

ChlP replicate 2
Peak
10
10 100 1000 104 105
Cy5 intensity

Binding site identification Array data analysis Genomic arrays

Figure 1.14  •  Methods for unbiased identification of transcription factor binding sites. Chromatin immunoprecipitation on sequencing (ChIP-seq) and
chromatin immunoprecipitation on microarray chip (ChIP-chip) can provide location, isolation, and identification of the DNA sequences occupied by specific
DNA-binding proteins in cells. Proteins capable of DNA interactions are targeted with specific antibodies. DNA and the associated proteins are cross-linked;
DNA is fragmented into 150 to 500 bp and immunoprecipitated. After reversion of the cross-link, DNA is isolated and either mass-sequenced (ChIP-seq)
or used as probes in a genomic array (ChIP-chip), and binding sites occupied by the proteins can be identified in the genome. These binding sites may indicate
functions of various transcriptional regulators and help identify their target genes during development and disease progression. The types of functional elements
identified with these techniques include promoters, enhancers, repressor and silencing elements, insulators, boundary elements, and sequences that control
DNA replication. (From Kim TH, Ren B. Annu Rev Genomics Hum Genet. 2006;7:81–102 and Liu et al. BMC Biol. 2010;8:56.)
16 Part I: Science and Clinical Oncology

Human Drosophila Yeast C. elegans Arabidopsis

Transcriptional Virus-host Metabolic Protein-protein Disease


regulatory network network network interaction network
Alzheimer’s
disease

Hypertension
Pseudohypo-
Atherosclerosis aldosteronism
Asthma

Figure 1.15  •  Interactome networks and human disease. Networks are integrated sources of information obtained from biochemical, molecular, proteomic,
and other high-throughput analyses. Different networks can be obtained for each organism, organ, or cell. In the first instance, central regulatory “nodes”
identify important components in the network. These networks and their data can then be integrated and compared with healthy and disease models, allowing
an integrative view of events that is much more powerful than isolated networks. (Modified from Vidal M, Cusick ME, Barabási A. Interactome networks
and human disease. Cell. 2011;144:986–998.)

protein-protein interactions in a cancer cell (its “interactome”), recently, PDX models have been further optimized with the use of
functional relationships between disease-promoting genes may be humanized host mice that are modified to contain human immune
revealed that provide novel candidates for intervention (Fig. 1.15). systems.
Networks of disorder-gene associations are already being built that
offer a platform for describing all known phenotype and disease-gene TRANSGENIC MODELS OF CANCER
associations, often indicating the common genetic origin of many
diseases. A precise diagnosis of cancer through use of proteomics Integrating an oncogene that causes malignancy into the genome of
can be envisioned, based on highly discriminating patterns of a mouse without altering the mouse’s own genes generates a transgenic,
proteins in easily accessible patient samples. Proteomics informa- cancer-prone mouse that transmits this trait to its offspring with a
tion also promises to provide sophisticated mathematical models of dominant pattern of inheritance. The technology for producing
the molecular events underlying a process as complex as neoplastic transgenic mice joins recombinant DNA methodology with standard
transformation, which will capture the dynamics of the disease with techniques that are used today by in vitro fertilization clinics, relying
unprecedented power. on the understanding of mammalian reproduction and the development
of protocols to harvest, manipulate, and reimplant eggs and early
MODELING CANCER IN VIVO embryos (Fig. 1.17). The transgene is constructed so that the gene
product will be expressed under appropriate spatial and temporal
Once the mechanistic underpinnings of a particular cancer have been control. In addition to all the standard signals necessary for efficient
described, creating an animal model to test that mechanism becomes transcription and translation of the gene, transgenes contain a promoter,
critical to understanding the pathophysiology and to design therapeutic or regulatory region, that drives transcription in either a ubiquitous
strategies for treatment. Advances in manipulation of the mouse genome or a tissue-restricted pattern. This requires an extensive knowledge of
have resulted in more sophisticated models of human cancer. These genetic regulation in the target cells. A recent advance that circumvents
methodologies can circumvent embryonic death by targeted alteration this requirement involves embedding the transgene inside another
of gene expression only after a critical period in development, and gene locus that is expressed in the desired pattern. Held in a bacterial
reduce the complexity of gene functional analysis by restricting its artificial chromosome (BAC) for easier manipulation, this long stretch
pattern of activation. Inducible gene expression or silencing also allows of DNA surrounding the host gene is likely to carry all the necessary
acute, as opposed to chronic, effects to be assessed. Although species regulatory information to guarantee a predictable expression pattern
differences in tumor susceptibility and disease remission exist between of the introduced transgene.
mouse and man, the tools for genetic manipulation in mouse are The transgene DNA is then injected into the male pronucleus of
superior to those in other mammals, and useful information about a fertilized mouse egg, obtained from a female mouse in which
the function of oncogenes can be gained by targeted expression of hyperovulation has been hormonally induced. The injected eggs are
mutant protein products in mouse tissues. cultured to the two-cell stage and then implanted in the oviduct of
A major hurdle in generation of clinically relevant mouse models another recipient female mouse. Transgenic pups are identified by the
to develop cancer treatments stems from the lack of patient tailoring. presence of the transgene in their genomic DNA (obtained from the
Cancer cells present a highly heterogeneous population that varies tip of the tail and analyzed with PCR assay). Typically, several copies
with the genetic makeup of the individual patient. This shortcoming of the transgene are incorporated in a head-to-tail orientation into a
has been addressed with the advent of patient-specific avatars, also single random site in the mouse genome. About 30% percent of the
known as personalized mouse models or patient-derived xenograft resulting pups will have integrated the transgene into their germline
(PDX) models (Fig. 1.16). Implanting patient biopsy specimens into DNA and constitute the founders of the transgenic lines. RNA analysis
immunodeficient mice allows growth of the tumor, generating in vivo of their progeny determines the level of transgene expression, and
precision models without further in vitro manipulation of the tumor whether the transgene is being expressed in the desired location or at
tissue. These models show great promise for designing treatment and the appropriate time. Given the variability in transgene number and
drug tests that should best target the patient-specific tumor. Most chromosomal location, transgene expression patterns and levels can
Molecular Tools in Cancer Research  •  CHAPTER 1 17

A B
Figure 1.16  •  Mouse avatar (PDX) models. (A) Patient-derived xenograft (PDX) mice are generated by implanting patient tumors into immunodeficient/
humanized mice. The tumors can then be propagated for several passages in fresh mice for a number of generations. (B) Usually, after the third generation
the tumors can be isolated and characterized for further study. These mice can potentially be used for patient drug-specific testing and molecular characterization,
therefore allowing for personalization of cancer treatment. (From http://www.the-scientist.com/?articles.view/articleNo/42470/title/My-Mighty-Mouse/.)

diverge considerably among different founder lines carrying the same The general scheme involves two mouse lines, one carrying the
transgene. recombinase either as a transgene driven by inducible regulatory
In general, transgenesis is optimal for modeling oncogenic mutations elements or knocked into one allele of a gene expressed in the desired
that cause a gain of function, producing disease even when they occur tissue. The other mouse line harbors a modified gene target including
in only one of a gene’s two alleles. For example, an activating mutation recognition sequences. Mating the two lines results in progeny carrying
in a growth factor that causes abnormal cell proliferation can be both the target gene and the recombinase, which interacts with the
mimicked by introducing a transgenic version of the mutated growth target gene only in the desired tissue.
factor gene under the control of an appropriate regulatory sequence A popular conditional methodology is based on the activation of
for expression in the tissue of interest. The relative susceptibility of nuclear hormone receptors to control gene expression. Two current
such a transgenic mouse to tumorigenesis can help distinguish between systems involve activation of a mammalian estrogen receptor, estrogen
a primary and secondary role of the mutant factor, and established analogue 4-hydroxy-tamoxifen, or an insect hormone receptor with
lines of these animals can be used for testing new therapeutic the corresponding ligand ecdysone. Although several variations on
protocols. these hormone-receptor systems are currently in use, the underlying
principle is the same. The Cre recombinase gene, or another regulatory
CONDITIONAL CONTROL OF protein, such as a transcription factor, is fused with the ligand-binding
ONCOGENE ACTIVATION domain (LBD) from a nuclear hormone receptor protein. The resulting
chimeric transgene is placed under the control of a promoter that
The genetic construction of cancer-prone transgenic mice with the directs expression to the tissue of interest, and transgenic animals are
capacity to induce oncogene expression in vivo provides a new avenue generated. In the absence of the hormone or an analogue, the fusion
to modeling the role of oncogenes in tumor generation and mainte- protein accumulates in the desired tissue but is rendered inactive
nance. This technology relies on conditional mutagenesis. Producing through its association with resident heat shock proteins. Hormone,
conditional mutations in mice requires a DNA recombinase enzyme administered either systemically or topically, binds to the LBD moiety
that does not recognize any mouse sequence, but rather targets short, of the fusion protein, dissociates it from the heat shock protein, and
foreign recognition sequences to catalyze recombination between them. allows the transcriptional regulatory component to find its natural
By strategic placement of these recognition sequences in appropriate DNA targets and promote lox-P mediated recombination, or in the
orientations either beside or within a mouse gene, the recombination case of an inducible transcription factor, activate expression of the
results in deletion, insertion, inversion, or translocation of associated corresponding genes. If the LBD is fused to a recombinase, administra-
genomic DNA (Fig. 1.18). Two recombinase systems are currently in tion of hormone leads to the rearrangement of target sequences. This
use: the Cre-loxP system from bacteriophage P1, and the Flp-FRT reaction is not reversible, but lends additional temporal control over
system from yeast. The 34 bp loxP or FRT recognition sequences do recombinase-based mutation. If the LBD is fused to a transcription
not occur in the mouse genome, and both Cre and Flp recombinases factor, removal of hormone leads to inactivation of the fusion protein
function autonomously, without the need for cofactors. Cre- or Flp- and gene downregulation.
mediated recombination is not distance or cell-type dependent, and Another inducible method in use is the tetracycline (tet) regula-
can occur in proliferating or differentiated tissues. tory system. In the classic design (tTA or tet-off ), a fusion protein
18 Part I: Science and Clinical Oncology

Figure 1.17  •  Generation of transgenic mice. The transgene containing


3' Flanking the DNA sequences necessary for the expression of a functional protein is
Promoter 5' UT Coding region 2' UT
region injected into the male (larger) pronucleus of uncleaved fertilized eggs through
Transgene
a micropipette. The early embryos are then transferred into the reproductive
tract of a mouse rendered “pseudopregnant” by hormonal therapy. The resulting
pups (founders) are tested for incorporation of the transgene by assaying
genomic DNA from their tails. Founder animals that have incorporated the
transgene (+) are mated with nontransgenic mice, and their offspring are mated
with each other to confirm germline integration and to establish a line of
homozygous transgenic mice. Several transgenic lines that have incorporated
Collection of fertilized eggs from different numbers of transgenes at different integration sites (and thus express
a superovulated donor mouse various amounts of the protein of interest) are usually studied. UT, Untranslated.
(From Schuldiner AR. Transgenic animals. N Engl J Med. 1996;334:653–655.)

Cell type specific


promoter Cre loxP Target gene loxP

X
Injection
of transgene into
male pronucleus of
uncleaved fertilized egg

Transfer of early embryos into reproductive


tract of a pseudopregnant mouse Cre Cre

A Special cell type All other cells

CMV-β actin
– promoter
– βgeo 3PA EGFP

+ loxP loxP
+ Cre

CMV-β actin
promoter
+ EGFP
Assay of genomic DNA from tails of founder
animals for incorporation of the transgene B loxP

Figure 1.18  •  Conditional mutagenesis schemes. (A) Two mouse lines are
required for conditional gene deletion: first, a conventional transgenic mouse
line with Cre targeted to a specific tissue or cell type; and second, a mouse
strain that embodies a target gene (endogenous gene or transgene) flanked by
two loxP sites in a direct orientation (“floxed gene”). Recombination (excision
and consequently inactivation of the target gene) occurs only in those cells
expressing Cre recombinase. Hence, the target gene remains active in all cells
and tissues that do not express the Cre recombinase. (B) The Z/EG double
reporter system. These transgenic mice constitutively express lacZ under the
Sequential matings to determine control of the cytomegalovirus enhancer/chicken actin promoter. Expression
germline integration is widespread, with notable exceptions being liver and lung tissue. Expression
is observed throughout all embryonic and adult stages. When crossed with a
Cre recombinase-expressing strain, lacZ expression is replaced with enhanced
Study of phenotype green fluorescent protein expression in tissues expressing Cre. This double
reporter system makes it possible to distinguish a lack of reporter expression
from a lack of Cre recombinase expression while providing a means to assess
Cre excision activity in live animals and cells. (A Courtesy Kay-Uwe Wagner,
National Institutes of Health; B from Novak A, Guo C, Yang W, Nagy A,
Lobe CG. Z/EG, a double reporter mouse line that expresses enhanced green
fluorescent protein upon Cre-mediated excision. Genesis. 2000;28:147–155.)
Molecular Tools in Cancer Research  •  CHAPTER 1 19

combining a bacterial tet repressor and a viral transactivation domain Overlapping genetic functions can also be defined by crossbreeding
drives expression of the target transgene by binding to upstream tet mice with mutations in different genes. In this way it is possible to
operator sequences flanking the transgene transcription start site. In the study the combinatorial effects of oncogene and tumor suppressor
presence of the antibiotic inducer, the fusion protein is dissociated from gene mutations.
the operator sequences, inactivating the transgene. In a complementary Several caveats are important in considering the use of knockout
design, called reverse tetracycline-controlled transactivator (rtTA or technology in modeling cancer. Most knockouts generate loss-of-
tet-on), structural modification of the tet repressor makes the antibiotic function (null) germline mutations. Inactivation of widely expressed
an active requirement for binding of the fusion protein to the operator genes with multiple functions may have complex phenotypes. Con-
sequences, such that its administration activates transgene expression at versely, if the functions of two genes overlap, a mutation in one of
any time during the life span of the mouse, whereas withdrawal results in the genes may not produce an abnormal phenotype, owing to compensa-
downregulation of the gene. It is important that the transgene integrate tion by the unaltered partner.
into a genomic locus that permits proper tTA or rtTA regulation so that Perhaps the greatest drawback of conventional knockout technology
the system exhibits minimal “intrinsic leakiness” and good antibiotic derives from the disruption of gene function at the earliest stage
responsiveness. of its expression. If the gene has a vital developmental role, the
Conditional expression systems have already been developed to identification of functions later in development can be occluded.
generate hematopoietic, leukemogenic, and lymphomagenic mutations Therefore, although the generation of a null mutation is an excel-
in the mouse, as well as solid tumors. These inducible cancer models lent starting point for analysis, it is far from being functionally
can be exploited to identify oncogenic signals that influence host-tumor exhaustive. For these reasons, conditional mutagenesis is the emerg-
interactions, to establish the role of a given oncogenic lesion in advanced ing method of choice for the elucidation of the gene functions
tumors, and to evaluate therapies targeted toward cancer-causing that exert pleiotropic effects in a variety of cell types and tissues
mutations. Potential clinical application of inducible systems include throughout the life of the animal, which is particularly relevant
targeting virally delivered transgene expression to malignant tissues for the generation of mouse models of adult-onset diseases such
by the use of specific inducible regulatory elements, restricting the as cancer.
expression of transgenes exclusively to affected tissues, and increasing Use of recombinase-mediated gene mutation as described earlier
the therapeutic index of the vectors, particularly in the context of for conditional transgenesis, conditional knockout mutations can be
solid tumors. In all cases, a basic knowledge of the specific mutations designed to disrupt the function of a target gene in a specific tissue
involved in the molecular genetics of malignancies is required because (spatial control) and/or life stage (temporal control). Depending on
it is often unclear that the causal mutation underlying the genesis of the design of the experiment, recombinase action can delete an entire
neoplasia continues to play a central role in the progression to the gene, remove blocking sequences to induce gene expression, or rearrange
fully transformed state. This is particularly important in modeling chromosomal segments. With the advent of recent internationally
cancers characterized by genetic plasticity, wherein drug resistance can coordinated systematic mutagenesis programs aiming to place a
arise subsequent to primary tumor formation. conditional inactivating mutation in each of the 20,000 genes in the
mouse genome, the possibilities for modeling cancer are limited only
MODELS OF RECESSIVE GENE MUTATIONS by a researcher’s choice of the gene loci to test. The constantly evolving
IN CANCER techniques for gene manipulation in vivo constitute a major advance
in cancer research.
In contrast to dominantly acting oncogenes, recessive genetic disorders, Genetically modified mice are of great value in dissecting the
such as loss-of-function mutations in tumor suppressor genes, require pathogenesis of many tumor types. In some knockout studies, the
both copies (alleles) of a gene to be inactivated. The methods needed phenotype of the mutated gene is anticipated by prior knowledge
to produce animal models of recessive genetic disease differ from those of the gene’s function. However, unexpected mutant phenotypes
used in studying dominant traits. Gene knockout technology has been may help clarify the mechanism of the underlying neoplasia.
developed to generate mice wherein one allele of an endogenous gene Pharmacologic manipulation of transgenic, knockout, diversified
is removed or altered in a heritable pattern (Fig. 1.19). Gene disruption animal models of cancer will prove useful in screening therapeutic
or replacement is first engineered in pluripotential cells, termed agents with potential for study in clinical trials. Therapy involving
embryonic stem cells (ESCs), which are genetically altered by introduc- gene or cell replacement can be also tested in genetically engineered
tion of a replacement gene that is inactive or mutant. disease models.
To reduce random integration of the foreign DNA, the replacement
gene is embedded into a long stretch DNA from its native locus in EXPLOITING MOUSE DIVERSITY FOR
the mouse, which targets the recombination event to the homologous CANCER RESEARCH
position in the ESC genome. Inclusion of selectable markers along
with the replacement gene allows selection of the cells in which A novel in vivo tool has emerged that aids in understanding the etiology
homologous recombination has taken place. Site-specific recombinase of cancers, by more accurately reflecting the broad genetic variability
systems combined with gene targeting techniques in ESCs can also in the human population. Cancer research performed with mice has
be used to induce recessive single point mutations or site-specific largely focused on a few individual highly inbred strains with limited
chromosomal rearrangements in a tissue- and time-restricted pattern. genetic diversity, which would equate to single individuals in the
In a variation on this theme called knockin, a foreign gene, such as population. Yet drugs designed to treat one individual are often not
one encoding a marker or a mutated gene, can be placed in the locus effective in other patients. The Collaborative Cross (CC) was created
of an endogenous gene. The engineered ESCs are then microinjected to provide mouse models that better represent the diversity seen in
into the cavity of an intact mouse blastocyst sufficiently early in gestation natural human populations while still retaining the broad power of
that they can, in principle, populate all the tissues of the developing genetic analysis seen in mice. The CC resource is a large panel of
chimeric embryo. This is rarely the case, so contribution of ESCs to recombinant inbred (RI) strains generated by randomly mixing the
the resulting animal is most often assessed with use of ESCs and genetic diversity of eight extant inbred mouse lines, and can be used
blastocysts whose genes for coat color differ. to test the impact of treatments in a diverse genetic pool akin to the
If the ESCs contribute to the germ cells of the founder mouse, human population (Fig. 1.20). A related resource, the Diversity Outcross
their entire haploid genome can be passed on to subsequent generations. (DO), offers higher mapping resolution by randomized outcrossing
Through mating together of subsequent progeny of the founder mouse, of partially inbred CC strains, which segregates the same allelic variants
both alleles of the mutated gene can be passed to a single animal. but embeds them in a distinct population architecture in which each
20 Part I: Science and Clinical Oncology

Embryonic stem cell

Tumor suppressor gene

5' Homologous 3' Homologous


region region
Intron
Cellular
gene

Embryonic stem
Plasmid
cell culture pgk-neo pgk-tk DNA
Knockout
vector
Homologous
recombination
Cellular gene
replaced

Injection of embryonic stem Implantation of chimeric


cells into host blastocyst blastocyst in foster mother
Selection by neomycin and ganciclovir

Germline offspring Chimeric offspring

Figure 1.19  •  Gene knockout strategies. Embryonic stem cells (upper left panel) contain the tumor suppressor cellular gene (upper right panel), which
consists of exon 1 (olive green, a 5′ noncoding region), an intron, and exon 2 (red, a protein coding region, and yellow, a 3′ noncoding region). A knockout
vector—consisting of a collinear assembly of a DNA flanking segment 5′ to the cellular gene (blue), the phosphoglycerate kinase–bacterial neomycin gene
(pgk-neo, violet), a 3′ segment of the cellular gene (yellow), a DNA flanking segment 3′ to the cellular gene (green), and the phosphoglycerate kinase–viral
thymidine kinase gene (pgk-tk, orange)—is created and introduced into the embryonic–stem cell culture. Double recombination occurs between the cellular gene
and the knockout vector in the 5′ homologous regions and the 3′ homologous regions (dashed lines), resulting in the incorporation of the inactive knockout
vector, including pgk-neo but not pgk-tk, into the cellular genomic locus of the embryonic stem cell. The presence of pgk-neo and the absence of pgk-tk in
these replaced genes will allow survival of these embryonic stem cells after positive-negative selection with neomycin and ganciclovir. The clone of mutant
embryonic stem cells is injected into a host blastocyst, which is implanted into a pseudopregnant foster mother and subsequently develops into a chimeric
offspring (bottom). The contribution of the embryonic stem cells to the germ cells of the chimeric mouse results in germline transmission of the embryonic
stem cell genome to offspring that are heterozygous for the mutated tumor suppressor allele. The heterozygotes are mated to produce mutant, cancer-prone
mice homozygous for tumor suppressor deficiency. (Modified from Mazjoub JA, Muglia LJ. Knockout mice. N Engl J Med. 1996;334:904–906.)
Molecular Tools in Cancer Research  •  CHAPTER 1 21

Founder strains Diversity Outbred (DO) formation and holds great promise for improved treatment of
A/J
human cancer.
Chemotherapy still has numerous side effects. As a number of
C57BL/6J
Outbreeding oncologists may testify, patients sometimes forgo treatment because
129S1/SvImJ of its toxic nature. In a number of cases of chronic lymphocytic
leukemia, if the symptoms of the disease are “under control,” treatment
NOD/ShiLtJ is not prescribed. Therefore it is important to find alleviation strategies
NZO/H1LtJ Collaborative Cross (CC) and cures that minimize the side effects. The goal should be to cure
Inbreeding the patient without devastating the person. One future avenue, apart
CAST/EiJ from finding treatment regimens that reduce side effects, is the discovery
PWK/PhJ of chemotherapies with minimal side effects. The nature of several
generations of chemotherapy was to attack cellular processes, such as
WSB/EiJ DNA replication, metabolism, and cell division, with brute force, and
A by trial and error find a balance between alleviating the neoplastic
growth and not interfering with the normal processes. It may be time
to rethink that premise. We now see examples wherein a less potent
chemotherapy elicits a similar effect on a cancer as a potent one,
although the less potent compound may take longer to achieve its
effect. However, because of its low potency, the side effects are minimal.
Therefore, in the future, the success of a therapy needs to be measured
B not only in terms of how quickly a patient is cancer free, but how
well the patient is during and after the treatment. With emerging
Figure 1.20  •  Generation and characteristics of Collaborative Cross (CC) technologies we will be able to fine-tune current successful therapies
and Diversity Outbred (DO) mice. (A) Each CC line originates from intercrosses so that treatment is not so burdensome.
obtained from eight founder lines. Individual unique independent line is In any field of medicine, resistance to the therapy occurs. Evolution-
inbred for at least 15 generations, creating individual CC lines. Together, those ary processes show that predation leads to natural selection of species
lines represent a much broader genetic variation when compared with the
parental lines and therefore are a better representation of natural populations with mutations that avoid their elimination. Similarly, in cancers,
but contain a high degree of homozygosity. Random mating from early 144 during the predation—chemotherapy—clones arise that become
CC crosses leads to a highly genetic heterogeneous outbred population that resistant to the therapy. With emerging technologies such as single-cell
more closely resembles the diversity found in human populations. (B) Images deep sequencing, we will be able to not only detect the rare subclone
of mice representing CC lines. (From Centre for Diabetes Research, Harry that could give rise to resistance, but also predict the probability of
Perkins Institute of Medical Research, Nedlands 6009, W.A Australia” and the development of resistance by sequencing certain markers. For
“School of Medical and Health Sciences, Edith Cowan University, Joondalup instance, clones with mutations in DNA repair pathways have a higher
W.A. 6027 Australia.) probability for genomic instability, which is a precursor for the rise
of resistant clones.
While we pursue new technologies to diagnose patients and
understand the molecular nature of the cancers, an emerging trend
will be to reexamine previous formulated hypotheses or treatments
that could not be tackled before because of the lack of technologies
animal has a high degree of heterozygosity and carries a unique or resources. For instance, it has been long thought that genetic variation
combination of alleles. plays an important role in cancer treatment. This premise was observed
These diversified mice have been shown to be a powerful tool in in human clinical trials, wherein conditions in some populations were
determining the etiology of cancers. In a recent study, analysis of mice refractory to certain treatments. In the future, we would be able to
generated by crossing CC strains with a mouse line carrying a mutated predict the extent to which a treatment would either work or fail in
tumor suppressor APCmin showed that colon cancer frequency and certain population by using diverse mice, and to map alleles that
spectrum varied predictably with genetic background. Identification confer resistance or susceptibility of a tumor before reaching human
of these genetic modifiers of cancer genesis or suppression would clinical trials. Another example is processes that lead to neoplasticity,
inform the design of novel mouse cancer models, potentially yielding such as LOH, which have been studied in cell lines that are homo-
genetic markers that can predict human cancers. geneous in nature. Attempts to use primary cells were not possible
because of polyclonality of cells and the short life span of tumor cells
FUTURE VIEW in vitro.
With all the aforementioned technologies, we now can, and
Recent discoveries that cancer stem cells share essential signaling nodes need to, reexamine older hypotheses with the appropriate type of
with normal stem cells suggest that targeting critical steps in these primary cells. We will probably uncover novel processes of cancer
pathways may lead to improved alternative cancer therapies. However, that were masked, and resolve decades-old controversies and
every human tumor is subtly different. We are now fast approaching competing hypotheses, leading to better understanding and cures
a new era in medicine: creating “tailored” treatments to individual for cancer.
tumors by obtaining integrative “personal omics profiles” (Fig. 1.21). In the future, fields of medicine will continue to marry and
The generation of novel mouse strains that represent the diversity merge; this has been exemplified in numerous examples, some
seen in human populations will likely lead to a more tailored approach mentioned in this chapter—for instance, the use of viruses to deliver
in understanding cancer diversity and therefore will allow the develop- chemotherapy, or the use of engineered T cells to attack cancer. As
ment of more efficient treatments. emerging molecular tools uncover novel processes in different fields
Understanding of underlying molecular biologic principles of of medicine, the cross talk between oncology and these other fields
malignancy, with pathophysiologic consequences, will generate an will continue, enabling discovery of new avenues to alleviate or even
invaluable resource for resolution of complex genetics of tumor cure cancers.
22 Part I: Science and Clinical Oncology

SAMPLE TYPE METHOD ANALYSES

Whole-genome
Variant calling/phasing
sequencing
Heteroallelic and variant
expression
Whole-transcriptome
PBMC sequencing RNA editing
(mRNA and miRNA)
Quantitative differential

Integrated personal OMICS


expression & dynamics

Variant confirmation in
Proteome profiling
RNA and protein

Untargeted proteome Quantitative differential


profiling expression and dynamics

Targeted proteome
Quantitative expression
profiling (cytokines)

Serum Metabolome profiling Dynamics

AutoAntibodyome Differential reactivity


profiling

Medical/lab tests Glucose, HbA1c, CRP,


Telomere length
A

3 4

5
2
Serpina 1
E366K
RNA edits
6

Heteroallelic SNVs
1

Protein-downregulated
7

(HRV vs healthy)
Protein-upregulated
Y

(HRV vs healthy)
RNA-downregulated
8

(HRV vs healthy)
X

RNA-upregulated
(HRV vs healthy)
Indels
9
22

SV-duplications
21

SV-deletions
20

10

Chr. ideogram
19

18
11
17 14 Chr. number
16 12
B 15
14 13

Figure 1.21  •  Integrative personal omics profiles (iPOP). (A) Integrative analysis of iPOP experimental design, indicating tissues and techniques analyzed
in healthy and diseased individuals. (B) Circos plot summarizing iPOP. From outer to inner rings: chromosome ideogram; genomic data (pale blue ring),
structural variants (deletions [blue tiles], duplications [red tiles]), indels (green triangles); transcriptomic data (yellow ring); proteomic data (light purple ring).
(Modified from Chen R, Mias GI, et al. Personal omics profiling reveals dynamic molecular and medical phenotypes. Cell. 2012;148:1293–1307.)
Molecular Tools in Cancer Research  •  CHAPTER 1 23

SUGGESTED READINGS
Alberts B, Johnson A, Lewis J, Raff M, Roberts K, Walter Hunter K. Host genetics influence tumour metastasis. The Complex Trait Consortium. The Collaborative
P. Molecular Biology of the Cell. 4th ed. London, UK: Nat Rev Cancer. 2006;6:141–146. Cross, a community resource for the genetic analysis
Taylor and Francis Group; 2002. Malaney P, Nicosia SV, Davé V. One mouse, one patient of complex traits. Nat Genet. 2004;36:1133–1137.
Chen CZ. MicroRNAs as oncogenes and tumor suppres- paradigm: new avatars of personalized cancer therapy. Weinberg RA. Biology of Cancer. Garland Science; 2006.
sors. New Eng. J. Med. 2005;353:1768–1771. Cancer Lett. 2014;344:1–12. Rosenthal N, Brown S. The mouse ascending: per-
Feinberg AP. Phenotypic plasticity and the epigenetics of Pecorino L. Molecular Biology of Cancer: Mechanisms, spectives for human-disease models. Nat Cell Biol.
human disease. Nature. 2007;447:433–440. Targets, and Therapeutics. USA: Oxford University 2007;9:993–999.
Frese KK, Tuveson DA. Maximizing mouse cancer models. Press; 2005. Wu J, Smith LT, Plass C, Huang TH. ChIP-chip comes
Nat Rev Cancer. 2007;7:654–658. Svenson KL, Gatti DM, Valdar W, Welsh CE, Cheng R, of age for genome-wide functional analysis. Cancer
Goh KI, Cusick ME, Valle D, Childs B, Vidal M, Barabasi Chelser EJ, et al. High resolution genetic mapping Res. 2006;66:6899–6902.
AL. The human disease network. Proc Natl Acad Sci using the mouse diversity outbred population. Genetics.
USA. 2007;104:8685–8690. 2012;190:437–447.
2  Intracellular Signaling
Aphrothiti J. Hanrahan, Gopa Iyer, and David B. Solit

S UMMARY OF K EY P OI N T S
• Ligand binding and activation of growth and survival, a phenomenon the current inability to directly inhibit
cell surface and internal receptors known as oncogene addiction. some oncogenic proteins (i.e.,
trigger the activation and/or • Drugs that selectively target mutated mutant KRAS), the development of
suppression of signaling cascades proteins critical for the maintenance drug resistance, technical hurdles
that regulate diverse cellular of the transformed phenotype have posed by limited tissue availability
processes including cell growth, shown unprecedented clinical for prospective molecular
proliferation, survival, and invasion, activity in genetically defined cancer characterizing, and intratumoral and
among others. subsets. lesion-to-lesion genomic
• Multiple nodes within these • Precision medicine refers to the use heterogeneity.
intracellular signaling networks are of genetic and epigenetic information • Routine genomic analysis of tumors
genetically and epigenetically altered unique to an individual cancer or tumor-derived cell free DNA in
in human cancers, leading to patient to develop treatment plasma is now a component of
constitutive pathway activation or regimens that target the driver standard care in an increasing
suppression. oncogenes and tumor suppressors number of cancer types, with the
• Some cancers are dependent on responsible for tumor progression. results used to guide treatment
genomic alterations in oncogenes or Potential challenges to the selection.
tumor suppressor genes for their application of this approach include

The underlying basis of the cancer phenotype is deregulated cell RECEPTOR TYROSINE KINASE SIGNALING
growth, which stems from two main hallmarks of cancer: uncon-
trolled proliferation, and loss of programmed cell death (enhanced The receptor tyrosine kinases (RTKs) comprise a family of
survival). In normal cells, these processes are tightly controlled transmembrane (TM) cell surface receptors that transduce
through integration of signaling cascades that translate extracellular extracellular signals internally to promote growth and survival and/
and intracellular cues into specific output responses. These signaling or to regulate other cellular phenotypes.2,3 Members of this protein
pathways are often initiated on binding of ligand to the extracellular family share a similar modular domain structure. Growth factors
domain of a receptor, followed by recruitment of adaptor proteins bind to the extracellular ligand-binding domain of RTKs and
or kinases that activate an intracellular cascading network of protein induce dimerization of two receptor monomers, juxtaposing the
and lipid intermediaries that ultimately produce a cellular response. intracellular tyrosine kinase domains of each monomer.4 This results
In normal cells, the specificity, amplitude, and duration of signaling in transphosphorylation of tyrosine residues within the cytoplasmic
are tightly regulated, and these constraints are often abrogated in domains of the RTK dimer. Following transphosphorylation, a variety
human cancers. of intracellular proteins are recruited to the activated RTK through
Investigation of the signal transduction pathways that regulate normal Src homology 2 (SH2) domains that recognize the phosphotyrosine
cellular functions has revealed that key components of these networks plus a specific amino acid sequence motif C-terminal to the tyrosine
are commonly altered in cancer cells by mutation, amplification and residues.5,6
deletion, chromosomal translocation, overexpression, or epigenetic Over 117 SH2 domains have been characterized, each with unique
silencing. These alterations lead to activation or suppression of phosphotyrosine sequence specificities.7 Each domain is part of a larger
signaling cascades that underlie the various hallmarks of the cancer adaptor protein involved in transducing extracellular signals to activate,
phenotype. This chapter reviews the major signal transduction cascades, or in some cases suppress, specific intracellular signaling cascades. Thus
with a focus on those that are frequently altered in human cancers. the complement of signaling pathways that a given RTK regulates is
Individual sections highlight signaling intermediaries that have been dictated by the profile of phosphorylated tyrosine residues plus flanking
validated as drug targets in patients with cancer. Table 2.1 summarizes amino acids within their intracellular domains.8,9 However, more than
actionable gene-level and mutation-level alterations in cancer and one adaptor protein can often recognize individual context-dependent
the drugs that are currently approved by the US Food and Drug phosphotyrosine motifs within an RTK, underscoring how this system
Administration (FDA) for treatment, that are recommended standard is designed to provide both specificity and diversity of intracellular
of care biomarkers, or that have promising clinical or preclinical signaling.
efficacy.1 Text continued on p. 29

24
Intracellular Signaling  •  CHAPTER 2 25

Table 2.1  Targeted Therapy for Disease-Specific Alterations of Actionable Oncogenes in Cancer
Gene Variant Cancer Type Drug Evidencea
ABL1 BCR-ABL1 fusion ALL Dasatinib 1
Imatinib 1
CML Dasatinib 1
Imatinib 1
Nilotinib 1
AKT1 E17K Breast AZD5363 3
Ovarian AZD5363 3
All Tumors ARQ 751 4
ALK Fusions NSCLC Alectinib 1
Ceritinib 1
Crizotinib 1
Oncogenic mutations NSCLC Brigatinib 1
Fusions Soft tissue sarcoma Ceritinib 2
Crizotinib 2
L1196M, L1196Q NSCLC Brigatinib 3
R1275Q Embryonal tumor Crizotinib 4
ARAF S214A Histiocytosis Sorafenib 3
S214C NSCLC Sorafenib 3
ATM N2875K, R3008C, truncating mutations Prostate cancer Olaparib 4
BRAF V600D, V600E, V600G, V600K, V600M, V600R Melanoma Cobimetinib + vemurafenib 1
Dabrafenib 1
Dabrafenib + trametinib 1
Vemurafenib 1
Histiocytosis Vemurafenib 2
NSCLC Dabrafenib 2
Dabrafenib + trametinib 2
Vemurafenib 2
Colorectal Binimetinib + cetuximab + 3
encorafenib
Panitumumab + vemurafenib 3
Colorectal Fluorouracil + radiation + 4
trametinib
V600E, V600K Melanoma Trametinib 1
Fusions Ovarian Paclitaxel + selumetinib 3
K601E Melanoma Trametinib 3
L597Q, L597R, L597S, L597V Melanoma Trametinib 3
D594E, D594N, G466V, G469A, G469V, G596C Melanoma Trametinib 4
KIAA1549-BRAF Fusion Soft tissue sarcoma Sorafenib + temsirolimus 4
L597Q, L597V Melanoma BGB659 4
BRCA1 Oncogenic mutations Ovarian Niraparib 1
Rucaparib 1
Olaparib 2
BRCA2 Oncogenic mutations Ovarian Niraparib 1
Rucaparib 1
Olaparib 2
CDK4 Amplification Dedifferentiated liposarcoma Abemaciclib, palbociclib 2
Well-differentiated Abemaciclib, palbociclib 2
liposarcoma
CDKN2A Oncogenic mutations Breast Letrozole + palbociclib 4
Esophagogastric Palbociclib 4
EGFR 729_761del, 729_761indel, L858R NSCLC Afatinib 1
Erlotinib 1
Gefitinib 1
Osimertinib 4
E709_T710delinsD, E709K, G719A, G719C, G719D, G719S, NSCLC Afatinib, erlotinib, gefitinib 1
A763_Y764insFQEA, L747P, A750P, A763_Y764insFQEA,
L833V, L861Q, L861R, S768I, EGFR-KDD
T790M NSCLC Osimertinib 1
762_823ins NSCLC EGF816 4
762_823ins, G719A, L861R, S768I NSCLC AP32788 4

Continued
26 Part I: Science and Clinical Oncology

Table 2.1  Targeted Therapy for Disease-Specific Alterations of Actionable Oncogenes in Cancer—cont’d
Gene Variant Cancer Type Drug Evidencea
ERBB2 Amplification Breast Ado-trastuzumab emtansine 1
Lapatinib 1
Lapatinib + trastuzumab 1
Pertuzumab + trastuzumab 1
Trastuzumab 1
Esophagogastric Trastuzumab 1
Breast Neratinib 3
Oncogenic mutations Breast Neratinib 3
V659E NSCLC Lapatinib 3
E770_K831indel, E770_K831ins NSCLC AP32788 4
ERCC2 Oncogenic mutations Bladder Cisplatin 3
ESR1 Oncogenic mutations Breast AZD9496, fulvestrant 3
D538G, Y537S Breast GDC-0810 4
EZH2 Oncogenic mutations Diffuse large B-cell GSK126 4
lymphoma Tazemetostat 4
FGFR1 Amplification Lung squamous cell AZD4547 3
carcinoma Debio1347 3
BCR-FGFR1 fusion Leukemia Ponatinib 4
FGFR2 Fusions Adrenocortical carcinoma Debio1347 3
JNJ-42756493 3
Bladder Debio1347 3
JNJ-42756493 3
Cholangiocarcinoma BGJ398 3
Debio1347 3
Endometrial Debio1347 3
JNJ-42756493 3
FGFR3 Fusions Adrenocortical carcinoma Debio1347 3
JNJ-42756493 3
Bladder Debio1347 3
JNJ-42756493 3
Glioma Debio1347 3
JNJ-42756493 3
G370C, G380R, K650E, K650M, K650N, K650Q, K650R, K650T, Bladder Debio1347 3
R248C, S249C, S371C, Y373C JNJ-42756493 3
Breast Debio1347 4
FLT3 Y572_Y630ins AML Sorafenib 3
IDH1 R132C, R132G, R132H, R132Q, R132S AML AG-120 3
All tumors BAY1436032 4
CB-839 4
IDH2 R140Q, R172G, R172K, R172M, R172S All liquid tumors AG-221 3
JAK2 PCM1-JAK2 fusion Leukemia Ruxolitinib 3
KIT 449_514mut, 550_592mut, A502_Y503dup, D579del, D820G, GIST Sunitinib 1
E554_K558del, H697Y, K550_W557del, K558delinsNP, L576P, Thymic tumor Sunitinib 2
P551_M552del, V555_L576del, V560D, V560del, V654A
449_514mut, 550_592mut, D419del, D579del, E554_I571del, GIST Imatinib 1
E554_K558del, E554_V559del, F522C, I563_L576del, I653T,
K550_W557del, K558N, K558_E562del, K558_V559del,
K558delinsNP, K642E, L576P, M541L, M552_W557del,
N564_Y578del, N822H, N822Y, P573_D579del, P577_
W582delinsPYD, P838L, Q556_K558del, T417_D419delinsI,
T417_D419delinsRG, T574insTQLPYD, V530I, V555_L576del,
V555_V559del, V559C, V559D, V559G, V559_V560del,
V559del, V560D, V560G, V560del, V569_L576del, W557G,
W557R, W557_K558del, Y553N, Y553_K558del, Y570H, Y578C
449_514mut, 550_592mut, D820G, D820Y, K550_W557del, GIST Regorafenib 1
K558delinsNP, N822K, V560D
D816F, D816Y, D820G, D820Y, L576P, N822I, V559D, V560G, GIST Dasatinib 2
W557_K558del
D816V, D820A, D820G, D820Y, K642E, L576P, V555_L576del, GIST Nilotinib 2
V559C, V559D, V654A, W557_K558del
D820A, D820E, D820G, D820Y, K642E, N505I, P577_D579del, GIST Sorafenib 2
V559D, W557_K558del Thymic tumor Sorafenib 2
K642E, L576P, V559A Melanoma Imatinib 2
Intracellular Signaling  •  CHAPTER 2 27

Table 2.1  Targeted Therapy for Disease-Specific Alterations of Actionable Oncogenes in Cancer—cont’d
Gene Variant Cancer Type Drug Evidencea
KRAS Wild type Colorectal Cetuximab 1
Panitumumab 1
Regorafenib 1
Cabozantinib + 4
panitumumab
Panitumumab + regorafenib 4
Pembrolizumab 4
Oncogenic mutations All tumors alpelisib + binimetinib 4
Cobimetinib + GDC-0994 4
Colorectal Atezolizumab + cobimetinib 4
Fluorouracil + radiation 4
therapy + trametinib
NSCLC Abemaciclib, PD0325901 + 4
palbociclib, palbociclib,
ribociclib, ribociclib +
trametinib
Binimetinib + erlotinib 4
Binimetinib, selumetinib, 4
trametinib
Docetaxel + trametinib 4
MAP2K1 Oncogenic mutations Histiocytic disorder Cobimetinib, selumetinib, 3
trametinib
Low-grade serous ovarian Cobimetinib, selumetinib, 3
trametinib
Melanoma Cobimetinib, selumetinib, 3
trametinib
NSCLC Cobimetinib, selumetinib, 3
trametinib
MDM2 Amplification Liposarcoma DS-3032b 3
RG7112 3
SAR405838 4
MET 963_D1010splice, 981_1028splice, X1006_splice, X1007_ NSCLC Crizotinib 2
splice, X1008_splice, X1009_splice, X1010_splice, X963_ Cabozantinib 3
splice Capmatinib 3
Amplification NSCLC Crizotinib 2
RCC Cabozantinib 2
D1010H, D1010N, D1010Y NSCLC Crizotinib 2
Cabozatinib 3
Capmatinib 3
MTOR E2014K Bladder Everolimus 3
C1483F, F1888L, L2230V, S2215F, T1977K RCC (clear cell) Everolimus, rapamycin, 4
temsirolimus
NF1 Oncogenic mutations Glioblastoma Trametinib 4
Melanoma Trametinib 4
Neurofibroma Binimetinib 4
Neurofibroma PLX3397 4
NRAS Oncogenic mutations Colorectal Atezolizumab + cobimetinib 3
Melanoma Binimetinib 3
Binimetinib + ribociclib 3
Thyroid Radioiodine uptake therapy 3
+ selumetinib
Colorectal Fluorouracil + radiation 4
therapy + trametinib
NTRK1 Fusions All tumors Larotrectinib 3
Salivary gland Entrectinib 3
NTRK2 Fusions Salivary gland Entrectinib 3
Larotrectinib 3
NTRK3 Fusions Salivary gland Entrectinib 3
Larotrectinib 3

Continued
28 Part I: Science and Clinical Oncology

Table 2.1  Targeted Therapy for Disease-Specific Alterations of Actionable Oncogenes in Cancer—cont’d
Gene Variant Cancer Type Drug Evidencea
PDGFRA FIP1L1-PDGFRA fusion Leukemia Imatinib 1
Fusions Myelodysplasia Imatinib 1
Myeloproliferative Neoplasm Imatinib 1
560_561insER, A633T, C450_K451insMIEWMI, C456_N468del, GIST Imatinib 2
C456_R481del, D568N, D842I, D842_H845del, D842_
M844del, D846Y, E311_K312del, G853D, H650Q, H845Y,
H845_N848delinsP, I843del, N659K, N659R, N659S, N848K,
P577S, Q579R, R748G, R841K, S566_E571delinsR, S584L,
V469A, V536E, V544_L545insAVLVLLVIVIISLI, V561A, V561D,
V561_I562insER, V658A, W559_R560del, Y375_K455del,
Y555C, Y849C, Y849S
D842V GIST Dasatinib 2
PDGFRB Fusions Dermatofibrosarcoma Imatinib 1
Protuberans
Myelodysplasia Imatinib 1
Myeloproliferative neoplasm Imatinib 1
PIK3CA Oncogenic mutations Breast Alpelisib 3
Alpelisib + fulvestrant 3
Buparlisib 3
Buparlisib + fulvestrant 3
Copanlisib 3
Fulvestrant + taselisib 3
GDC-0077 3
Serabelisib 3
Taselisib 3
Alpelisib + everolimus 4
Alpelisib + letrozole, Alpelisib 4
+ letrozole + ribociclib
Alpelisib + LJM716 + 4
trastuzumab
Alpelisib + olaparib, 4
buparlisib + olaparib
AZD5363 + fulvestrant 4
AZD8835 + fulvestrant 4
MLN0128 + serabelisib 4
All tumors ARQ 751 4
AZD5363 + olaparib 4
GDC-0077 4
Endometrial Alpelisib + fulvestrant 4
Buparlisib + fulvestrant 4
Fulvestrant + taselisib 4
Ovarian Alpelisib + fulvestrant 4
Buparlisib + fulvestrant 4
Fulvestrant + taselisib 4
PTCH1 Truncating mutations Embryonal tumor Sonidegib 3
Skin cancer, nonmelanoma Sonidegib 3
Vismodegib 3
PTEN Oncogenic mutations All tumors ARQ 751 4
AZD5363 + olaparib 4
AZD8186 4
Gedatolisib + palbociclib 4
GSK2636771 4
LY3023414 4
Endometrial Olaparib 4
Prostate Enzalutamide + LY3023414 4
RAF1 S257L Lung adenocarcinoma Sorafenib 4
RET Fusions NSCLC Cabozantinib 2
Vandetanib 3
ROS1 Fusions NSCLC Crizotinib 1
D2033N Cabozantinib 3
Intracellular Signaling  •  CHAPTER 2 29

Table 2.1  Targeted Therapy for Disease-Specific Alterations of Actionable Oncogenes in Cancer—cont’d
Gene Variant Cancer Type Drug Evidencea
TSC1 Oncogenic mutations CNS Everolimus 2
RCC Everolimus 2
TSC2 Oncogenic mutations CNS Everolimus 2

a
Levels of evidence:
• Level 1: FDA-recognized biomarker predictive of response to an FDA-approved drug in this indication
• Level 2: Standard care biomarker predictive of response to an FDA-approved drug in this indication or another indication; including those recommended by NCCN, but
not FDA recognized as standard of care
• Level 3: Evidence of clinical activity in this indication, or another indication
• Level 4: Preclinical or biologic evidence of activity
ALL, Acute lymphoblastic leukemia; AML, acute myeloid leukemia; CML, chronic myelogenous leukemia; CNS, central nervous system; FDA, US Food and Drug Administration;
GIST, gastrointestinal stromal tumor; NCCN, National Comprehensive Cancer Network; NSCLC, non-small cell lung cancer; RCC, renal cell carcinoma.
Data modified from oncokb.org1; Chakravarty D, Gao J, Phillips S Kundra R, Zhang H, Wang J. OncoKB: a precision oncology knowledge base. JCO Precis Oncol. Published
online May 16, 2017.

Recruitment of signaling intermediaries to the plasma membrane epigen.19–21 Growth factor binding promotes either homodimerization
facilitates their interaction with membrane-bound proteins responsible or heterodimerization with other HER family members, followed by
for stimulating a diverse array of downstream pathways (Fig. 2.1). As transphosphorylation.22 A ligand for HER2 has not yet been identified;
an example, the lipid kinase phosphatidylinositol 3-kinase (PI3 kinase), instead, HER2 is activated through heterodimer formation with one
described in more detail in a later section, recognizes and binds to a of the other three ligand-bound receptors.23 Notably, HER2 is the
pattern of phosphorylated tyrosine residues present within multiple preferred dimerization partner for EGFR, and EGFR-HER2 heterodi-
activated RTKs through the SH2 domain located in its p85 regulatory mers are more stable than EGFR homodimers, remaining at the cell
subunit. Binding of the p85 regulatory subunit in turn results in surface for a longer duration and undergoing endocytosis at a lower
activation of its kinase activity. Approximately 20 classes of RTKs rate than EGFR homodimers.24,25 Furthermore, HER2 reduces the
have been defined based on growth factor specificity. This section will dissociation rate of EGF from EGFR, allowing for a more sustained
focus on those RTK classes for which specific cancer therapies exist period of EGF-induced signaling.19 The EGFR and HER2 components
or are in development. of the EGFR-HER2 heterodimer are also more likely to be recycled
back to the cell surface than EGFR homodimers, which are more
Epidermal Growth Factor Receptor Signaling readily targeted for degradation.26 In addition, HER2-HER3 het-
erodimers possess the most potent mitogenic activity among the
Historically, the growth factors that stimulate RTKs were first heterodimer and homodimer HER kinase combinations.27 In contrast
discovered, followed by the structural and functional characterization to the other HER kinase family members, HER3 does not have intrinsic
of the RTKs themselves.10,11 Epidermal growth factor (EGF) was kinase activity and preferentially forms heterodimers with HER2.28
initially purified from mouse submaxillary glands in 1962 by Stanley The ligands for HER3 and HER4 are the neuregulins, including
Cohen and was found to stimulate premature eyelid opening and heregulin.
incisor eruption, phenotypes that suggested a role for EGF in the A number of tumor types frequently exhibit alterations within the
regulation of cellular proliferation.12 In 1978, the epidermal growth EGFR family of RTKs.21 Sustained activation of these pathways can
factor receptor (EGFR) was identified as the cell surface binding result in oncogene- or pathway-addicted tumors, and selective HER
site for EGF.13 Over the next several years, tyrosine phosphorylation kinase inhibitors are now a component of the standard treatment of
was identified in cells, followed by the discovery that the viral Src several malignancies. Alterations that affect RTK activity include
oncogene, which induces transformation of cells in vitro, is itself a mutations that result in constitutive activation of the tyrosine kinase;
tyrosine kinase, underscoring the potential importance of tyrosine overexpression of the receptor, often due to gene amplification; and
phosphorylation for oncogenesis.14,15 Once the complete sequence elevated levels of RTK ligands that stimulate signaling. EGFR mutations
of the EGFR protein was elucidated in the 1980s,16 the amino acid are found in 10% to 25% of non–small cell lung cancers (NSCLCs),
sequence of the receptor cytoplasmic domain was found to be similar with variation in the frequency of such alterations influenced by
to Src, suggesting that EGFR also possessed tyrosine phosphorylation ethnicity and geographic location; in-frame microdeletions in exon
activity. The connection between RTK activation and oncogenesis was 19 and point mutations in exon 21 (most commonly L858R or L861Q)
further solidified when the amino acid sequence of EGFR was found or exon 18 (G719X) comprise over 80% of these alterations.29–31 In
to be homologous to the avian erythroblastosis virus erbB oncogene, glioblastoma multiform (GBM), EGFR mutations, indels (including
which, when infected into chicken red blood cell precursors, is sufficient the EGFRvIII variant in which exons 2 to 7 of the extracellular domain
to induce erythroleukemia.17,18 The erbB oncogene encodes a TM are deleted, generating a ligand-independent, activated protein),
protein that lacks the extracellular ligand binding domain of EGFR amplification, splice variants, and rearrangements occur in 57% of
but possesses a cytoplasmic kinase domain that, when expressed in cells, tumors.32–34 However, because of the heterogeneity of GBM tumors,
can signal in a growth factor–independent manner. Subsequent studies targeting EGFR is complicated; EGFR alteration is often concurrent
have since identified within human cancers numerous alterations of with amplification or mutation of another RTK such as PDGFR,
EGFR and other RTKs that enhance proliferation without the need MET, or FGFR, or the presence of EGFRvIII on extrachromosomal
for growth factor stimulation. DNA, or activation of IDH1.35–38 Overexpression of wild-type EGFR
The EGFR class of RTKs comprises four receptor proteins encoded as a result of gene amplification has been observed in NSCLC and
by four genes (in parentheses): EGFR (ERBB1), HER2/Neu (ERBB2), breast, gastric, colorectal, and head and neck cancers, and less commonly
HER3 (ERBB3), and HER4 (ERBB4). EGFR binds to and is activated in other tumor types.39–41
by a number of ligands, including EGF, transforming growth factor–α Up to 30% of breast cancers display overexpression of HER2,
(TGF-α), HB-EGF, amphiregulin, betacellulin, epiregulin, and which is an unfavorable prognostic factor, and therapy for these
30 Part I: Science and Clinical Oncology

Growth Factor Ligand oncogene


tumor suppressor
Receptor Tyrosine Kinase

P Shc
Ras-GDP NF1
P Grb2 Trafficking/
Sos RalGDS RalA/B
proliferation
Ras-GTP PLCε
Vermurafenib
Dabrafenib
Raf p85
P P TIAM1
MEKK1/NF-κB PIK3CA-
p110α
Trametinib P P
Selumetinib MEK
Cobimetinib
CDC42/RAC AKT

SCH772984 P P
DUSPs NF- κB/actin
BVD-523 ERK
DEL-22379 mTORC1
P
p90RSK
translation
NF- κB, Myt-1, GSK3, PP-1

P
Fos/Jun/etc. Proliferation/growth

Negative feedback

Figure 2.1  •  Canonical Ras/MAPK signaling pathway. Ras proteins cycle between GDP-bound inactive and GTP-bound active states. Ras is often activated
in response to ligand-specific binding to its cognate receptor. Ras can also be activated via intracellular cross talk. This schematic depicts the classic Ras/MAPK
signal transduction cascade. Growth factor stimulation induces receptor tyrosine kinase (RTK) dimerization and autophosphorylation of tyrosine residues
located within the intracellular domain of the receptor. These phosphorylated tyrosine residues serve as docking sites for scaffold proteins that facilitate activation
of intracellular signaling cascades. For example, the adaptor protein Grb2, via its SH3 domain, recruits the Ras GEF (guanine-nucleotide exchange factor)
SOS. Colocalization of SOS and Ras facilitates substitution of GTP for GDP and thus Ras activation. Active GTP-bound Ras binds and recruits the Raf (A-,
B-, and C-Raf ) serine/threonine kinases to the plasma membrane and facilitates their activation. Active Raf in turn phosphorylates and activates MEK, which
in turn phosphorylates and activates ERK. ERK phosphorylates substrates in the cytoplasm (p90RSK) and in the nucleus (Jun, Fos, Ets-2, Elk-1, CREB1,
AP-1, ATF-2, among others), which regulate cell proliferation and survival. Ras interacts with more than 20 effector proteins, including the p110α subunit
of PI3 kinase, RasGDS, PLCε, and TIAM1, which in sum control transcription, translation, vesicular trafficking, cell cycle progression, cytoskeletal changes,
metabolic processes, immune inflammatory responses, and survival. Induction of Ras signaling also upregulates negative feedback elements that inhibit the
pathway (e.g., DUSPs and SPROUTYs/SPREDs). Several proteins within the Ras signaling cascade are proto-oncogenes (green) and tumor suppressors (red)
that are mutated, amplified, or deleted in many cancers. A number of selective inhibitors of Ras effectors have been tested as anticancer therapies. Examples
include kinase inhibitors, which selectively target B-Raf and its downstream effectors MEK and ERK (see red boxes).

ERBB2-amplified breast cancers is now distinct from that of breast of KRAS wild-type colorectal and head and neck cancers.51–56 Pani-
cancers with normal HER2 expression levels.21,42 ERBB2 amplification tumumab, another human monoclonal anti-EGFR antibody, is also
is also a driving event in gastric and to a lesser extent in bladder, approved for KRAS wild-type metastatic colorectal cancer.56
endometrial, and cervical cancer.43–45 More recently, activating mutations The first-generation reversible EGFR tyrosine kinase inhibitors
and in-frame insertions/indels in ERBB2 were found to occur in 1% gefitinib and erlotinib, as well as the second-generation irreversible
to 2% of all cancer patients, most commonly in patients with bladder inhibitor afatinib, are FDA approved for the treatment of NSCLC,
cancer.44 Mutations in ERBB2 localize to either the extracellular domain, with greatest efficacy in patients with EGFR mutations or in-frame
where they are presumed to promote dimer formation, or the kinase deletions.57–60 A second site mutation in EGFR (T790M) is a common
domain.46,47 In addition, activating mutations in ERBB3 have also mechanism of acquired resistance to first generation EGFR inhibitors.
been identified in bladder, colon, and gastric cancers.48,49 Osimertinib (AZD9291), a third-generation EGFR inhibitor, is highly
Numerous targeted agents have been developed that selectively active in patients with NSCLC in which resistance is mediated by the
inhibit EGFR-induced signaling (see Table 2.1 and Fig. 2.2).21,50 EGFR T790M mutation and is now FDA approved for this indica-
Cetuximab, a chimeric monoclonal antibody that binds to the tion.61,62 The development of osimertinib and the fourth-generation
extracellular domain of EGFR and competitively inhibits ligand binding, EGFR inhibitor EAI04563 highlights how studies of acquired resistance
thereby preventing receptor activation, is approved for the treatment can lead to the rational development of more effective kinase inhibitors.
Intracellular Signaling  •  CHAPTER 2 31

XL147
Buparlisib Growth Factor Ligand
Alpelisib (α-specific) EGFR:Cetuximab
AZD5363 AZD8186 (β-specific) EGFR/HER2:Lapa­nib,
Copanlisib (α/δ-specific) Receptor
Afureser­b Trastuzumab
Idelalisib (δ-specific) Tyrosine
Kinase

EGFR:gefi­nib, erlo­nib,
PIP2/3 P
PIP2 P osimer­nib
AKT P PI3K-
p85
IRS1 EGFR/HER2:lapa­nib,
P P P
PTEN p110α afa­nib, nera­nib
P
Other: ima­nib, alec­nib,
PIP3 Ras-
IκB PDK1 larotrec­nib, etc
P GTP
mTORC2 SGK CDC42/RAC
Bad
P Dual PI3K-mTOR inhibitors:
P RHEB-GDP NF-κB/ac­n
NF-κB dactolisib, voxtalisib
GSK3β apoptosis TSC1
TSC2

RHEB-GTP
P P
Cyclin
D1 mTOR inhibitors:
PRAS40 mTORC1 rapamycin, everolimus,
temsirolimus, AZD8055, RapaLink
degrada­on
P P
4EBP1 p70S6K
P
Protein transla­on S6

FOXO1/4 Arrest/Apoptosis
oncogene
tumor suppressor

prolifera­on and survival


Figure 2.2  •  PI3K/mTOR signaling pathway. The PI3 kinase family proteins are lipid kinases that transduce signals from receptor tyrosine kinases (RTK)
and G protein–coupled receptors to intracellular cascades that control proliferation, survival, and other cellular phenotypes. As an example, growth factor
binding causes receptor dimerization and subsequent phosphorylation of tyrosine residues in the intracellular domain of the receptor. These tyrosine phosphoryla-
tion sites serve as docking sites for the p85 regulatory subunit of PI3 kinase and adaptor proteins such as IRS-1 in the case of signaling induced by the insulin/
IGF1 receptors. This results in allosteric activation of the catalytic subunit of PI3 kinase, which converts PIP2 to PIP3. PIP3 recruits PDK1 and AKT to the
membrane via their pleckstrin homology (PH) domains. Colocalization of PDK1 and AKT results in phosphorylation (on threonine 308) and activation of
AKT. Phosphorylation of AKT on Ser473 by mTORC2 is required for full activation of AKT. Activated AKT phosphorylates several effectors, including
GSK3β, Bad, PRAS40, IκB, the FOXO1/4 transcription factors, and TSC2. AKT phosphorylation of TSC2, which is bound to TSC1, inhibits the GTPase
function of this complex, thereby allowing activation of Rheb and subsequent activation of mTORC1. In turn, mTORC1 phosphorylates p70S6 kinase
(p70S6K) and 4EBP1, an inhibitor of the eIF4E component of the cap-dependent translation initiation complex. p70S6K and mTOR also function to negatively
regulate the pathway by initiating the phosphorylation and inhibition of IRS-1. PI3 kinases can also signal to other effectors such as Rac/CDC42 and the
serum-glucocorticoid kinase (SGK) family to promote cellular survival, motility, and cytoskeletal rearrangement. Many components of the PI3 kinase signaling
pathway are mutationally altered in cancer (oncogenes in green, tumor suppressors in red). A variety of compounds have been developed that selectively inhibit
PI3 kinase signaling components. US Food and Drug Administration (FDA)–approved drugs and novel inhibitors in clinical testing that target RTKs, PI3
kinase, mTOR kinase, and AKT are highlighted in the red boxes.

ERBB2 amplification strongly correlates with HER2 protein Although the introduction of trastuzumab has resulted in a sig-
overexpression, and the presence of either marker predicts for trastu- nificant improvement in the survival of patients with HER2-
zumab response in certain cancers.64 Trastuzumab, a humanized antibody overexpressing breast cancers, drug resistance remains a major clinical
that binds to the extracellular domain of HER2, has been FDA approved problem. Potential resistance mechanisms include concomitant
for the treatment of breast65–71 and esophagogastric72 cancers displaying overexpression of other HER kinase family members and/or ligands,
HER2 overexpression. In patients with breast and gastric cancers, PTEN loss, and the expression of a truncated HER2 protein lacking
trastuzumab has modest activity when administered as single- the extracellular antibody binding site.79 Additional HER2-directed
agent therapy and is most commonly used in combination with agents include the tyrosine kinase inhibitors lapatinib and neratinib
chemotherapy.72–74 The combination of docetaxel, trastuzumab, and (see Table 2.1). Lapatinib is FDA approved for use in combination
pertuzumab,75 an antibody that binds to a different HER2 epitope with capecitabine in patients with HER2-overexpressing advanced or
(the dimerization domain) than trastuzumab and results in impaired metastatic breast cancer that has progressed on prior therapy with
dimer formation, is also approved for breast cancer.76–78 trastuzumab and certain classes of chemotherapy.80 The combination
32 Part I: Science and Clinical Oncology

of lapatinib and trastuzumab is also FDA approved in HER2-amplified alectinib and ceritinib that are either more potent or more selective
breast cancer.81–83 Lapatinib also received accelerated approval for use for ALK have significant clinical activity in patients with acquired
in combination with the aromatase inhibitor letrozole.84 Clinical efficacy resistance to crizotinib and are FDA approved for this indication.116,117
has been reported with lapatinib in HER2-mutant NSCLC.85,86 The In addition, brigatinib, a dual inhibitor of ALK and EGFR, was
irreversible pan-HER kinase inhibitor neratinib has shown promising granted accelerated FDA approval in 2017 for patients with metastatic
clinical activity in patients with ERBB2-amplified and ERBB2-mutant NSCLC and in patients with ALK alterations who progressed on
breast tumors, but also other cancer types.46,87–93 crizotinib.118

Insulin, Insulin-Like Growth Factor-1 Receptor Platelet-Derived Growth Factor Receptor, KIT,
Signaling, ALK, and ROS1 and FLT-3 Signaling
The insulin and insulin-like growth factor 1 (IGF1) receptor family Platelet-derived growth factor (PDGF) is the ligand for PDGFRs,
is dysregulated in multiple malignancies.94 The insulin receptor exists which stimulate the proliferation and migration of mesenchymal cells,
as two isoforms encoded by splice variants of the same gene.95 Each such as oligodendrocyte precursors, vascular smooth muscle cells, and
isoform can dimerize with the other (forming hybrid dimers) or with pericytes during embryonic development.119 PDGF signaling is also
itself.96 The IGF1 receptor (IGF1R) can dimerize with either of the implicated in organ development, including lung and intestinal epithelial
insulin receptor isoforms or with itself, resulting in six different dimer folding and glomerular capillary tuft formation. Furthermore, PDGFs
combinations.96 The insulin receptor is stimulated by insulin or promote angiogenesis, wound healing, and erythropoiesis.120 Aberrations
insulin-like growth factor 2 (IGF2), whereas IGF1R can be activated in the PDGFR pathway result in uncontrolled proliferation and
by either IGF1 or IGF2. Both of these latter ligands can stimulate enhanced angiogenesis.
IGF1R in an autocrine fashion or can be elaborated from distant Four isoforms of PDGF have been identified: PDGFA, PDGFB,
sites.97,98 Circulating IGF binding proteins have a similar affinity for PDGFC, and PDGFD.121 These isoforms are activated by proteolytic
IGF1 and 2 as IGF1R does and therefore compete for binding to cleavage and assemble into five homodimeric or heterodimeric com-
both ligands, thus titrating the amount of free ligand available for binations that bind to and stimulate either PDGFRα or PDGFRβ.
IGF1R stimulation.99 IGF binding protein proteases provide an PDGFRα homodimers inhibit chemotaxis, whereas PDGFRβ
additional mechanism for controlling ligand levels by increasing the homodimers and α/β heterodimers stimulate chemotaxis within
half-life of free ligand available for receptor binding.100 fibroblasts and smooth muscle cells.122 Angiogenic endothelial cells
After ligand binding, IGF1R dimerizes and undergoes transphos- recruit PDGFRβ-positive pericytes to cover blood channels and aid
phorylation, leading to activation of downstream signaling pathways, in their maturation and stabilization through secretion of PDGFβ.123
including both the Ras-Raf-MAPK and the PI3 kinase-AKT-mTOR Following dimerization and transphosphorylation, PDGFRs activate
cascades (see individual sections later in the chapter; see also Fig. signal transduction pathways through recruitment of adaptor proteins
2.1).101 Specifically, insulin receptor substrate 1 (IRS1) binds to a containing SH2 domains, most notably the Grb2 protein, which in
phosphotyrosine motif on IGF1R via its SH2 domains and is phos- turn binds the guanine nucleotide exchange factor (GEF) Sos, which
phorylated by IGF1R.102 It subsequently recruits PI3 kinase to the subsequently activates Ras.124,125 In addition, phosphorylated tyrosine
plasma membrane, which converts phosphatidylinositol-4,5-bisphosphate residues serve as docking sites for SH2 domain–containing kinases,
(PIP2) to phosphatidylinositol-3,4,5-triphosphate (PIP3), subsequently including PI3 kinase, phospholipase-Cγ, and Src, as well as the tyrosine
resulting in AKT and mTOR pathway activation. phosphatase SHP2 and the STAT transcription factor family.125
Activating mutations of IGF1R do not appear to be common in Alterations in PDGFR signaling in cancer include excess autocrine
human cancer. However, amplification of the IGF1R gene locus has secretion of PDGF (glioblastoma, sarcomas), gain-of-function mutations
been identified in some colon, pancreas, and lung cancers. Sarcomas that cause constitutive tyrosine kinase activation (GISTs),126 transloca-
often exhibit either increased expression of the IGF1 and IGF2 ligands tion of either the PDGF or PDGFR gene (dermatofibrosarcoma
or decreased IGFBP-3 expression (Ewing sarcoma), which results in protruberans, chronic myelomonocytic leukemia, hypereosinophilic
increased IGF1 levels in the tumor microenvironment.103 Gastrointestinal syndrome),127–129 and PDGFR gene amplification (glioblastoma).130
stromal tumors (GISTs) lacking c-KIT and platelet-derived growth PDGFRα mutations are found in approximately 10% of KIT wild-type
factor receptor (PDGFR) mutations also commonly harbor IGF1R GISTs and are sensitive to imatinib, a tyrosine kinase inhibitor of
amplification.104 AMG479, a monoclonal human antibody targeting KIT, BCR-ABL, and PDGFRs, which is standard of care in this setting
IGF1R, has shown promising antitumor activity in patients with (see Table 2.1).122,126,131 The D842V mutation comprises approximately
Ewing sarcoma.103 two-thirds of PDGFRα activating mutations, and confers resistance
Activating kinase domain point mutations and gene rearrange- to imatinib. Notably, the second-generation inhibitor dasatinib is
ments of the insulin receptor family members anaplastic lymphoma effective in preclinical models of imatinib-resistant GIST.126,132,133
kinase (ALK) and ROS1 play driving roles in many cancers, most Dermatofibrosarcoma protuberans is a rare, low-grade cutaneous
notably lymphomas, neuroblastoma, NSCLC, and thyroid cancer.105–108 sarcoma that harbors a chromosome 17;22 translocation that fuses
Chromosomal translocations involving ALK and at least 22 5′ fusion portions of the COL1A1 (collagen 1A1) gene and PDGFB, resulting
partners have been identified,108 which dictate spatial and temporal in overexpression of PDGF-β and subsequent stimulation of PDGFR
expression of the ALK fusions, and likely their function and tumorigenic signaling.122 Twenty other fusions partners have been identified in
potential.105 In NSCLC, the EML4 gene is the preferred translocation PDGFβ rearrangements, including ETV6 and EBF1. Imatinib has
partner, resulting in the expression of an EML4-ALK fusion protein shown significant benefit in patients with recurrent or metastatic
in 4% to 6% of patients.109,110 Notably, EML4-ALK fusions are found dermatofibrosarcoma protuberans, myelodysplasia, and myeloprolifera-
in a mutually exclusive pattern with EGFR kinase domain mutations, tive neoplasms and is FDA approved for these indications.134–138
suggesting that they have overlapping downstream effects. ROS1 gene The KIT gene is a member of the type III RTK family, which
rearrangements are also found in a minority of NSCLC patients with includes PDGFR and FLT3 (see later).139–141 It was first identified as
binding partners including SLC34A2 and CD74.106,111,112 Crizotinib, the human homologue of the viral oncogene v-Kit responsible for the
an inhibitor of the ALK, ROS1, and MET tyrosine kinases (see Table Hardy-Zuckerman IV feline sarcoma virus.142 Mutation of KIT or its
2.1), is now FDA approved for use in NSCLC patients with ALK ligand, stem cell factor (SCF),143–146 in mice induces coat color
or ROS1 fusions (see Table 2.1),113–115 although acquired resistance abnormalities (“white spotting”), anemia, and mast cell deficiencies,
mutations in ALK (of note, C1156Y and the gatekeeper mutation suggesting that it plays a role in hematopoiesis and melanogenesis.147,148
L1196M) commonly develop. Newer ALK inhibitors including Furthermore, the KIT protein was discovered as a cell surface receptor
Intracellular Signaling  •  CHAPTER 2 33

in acute myeloid leukemia (AML).149 KIT expression is mainly restricted factors, including tissue-specific FGF ligand and receptor expression, the
to mast cells, hematopoietic cells, germ cells, melanocytes, and the presence of cell surface molecules that facilitate the interaction between
interstitial cells of Cajal (ICCs) in the gut.150,151 SCF and KIT integrate individual FGF ligands and receptors, and the differential binding
signals that lead to mitogen-activated protein kinase (MAPK), PI3K, capability of the ligands themselves for specific FGFRs.178 Subsequent
and SRC pathway activation, and mediate critical survival and prolifera- stimulation of the tyrosine kinase domain leads to phosphorylation
tion cues to distinct hematopoietic lineages, including the bone marrow and activation of multiple downstream signaling proteins in the same
and progenitor cells. manner as described earlier for other RTKs. Unique to the FGFR
Hot-spot mutations in exons 9 and 11 of KIT have been identified signaling complex is FGFR substrate 2 (FRS2), an adaptor protein
in several tumor types, including GIST, melanomas, and germ cell that binds to specific phosphotyrosines on the intracellular domain
tumors.146,152–156 In GIST, 85% of tumors have activating KIT mutations of active FGFR dimers.179 FRS2 is itself phosphorylated by FGFRs
that drive the transformation of precursors of ICCs.153 Imatinib157–161 and serves as a docking site for the Grb2-Sos adaptor complex, which
and sunitinib158,162,163 inhibit KIT and PDGFR, among other kinases, activates the Ras/Raf/MAPK pathway. Phosphorylated FRS2 also
and are FDA approved for use in patients with KIT mutant GIST recruits Grb2-associated binding protein 1 (GAB1), which activates
(see Table 2.1). Regorafenib is approved for patients with imatinib- or PI3 kinase. In addition, phospholipase-Cγ binds to phosphorylated
sunitinib-refractory GIST.164 Imatinib has also been shown to induce FGFR dimers via an SH2 domain, leading to its activation and the
tumor regression in patients with KIT-mutant or KIT-amplified cleavage of phosphatidylinositol 4,5-bisphosphate (PIP2) to form inositol
melanoma.165,166 Second site mutations in KIT, typically in exon 17, 1,4,5-triphosphate (IP3) and diacylglycerol (DAG).
are a mechanism of acquired resistance to imatinib therapy in patients Germline mutations of the FGFR genes are the basis of a spectrum
with GIST.167 Novel agents that retain activity in the setting of an of skeletal developmental disorders that are thought to derive from
exon 17 KIT mutation are now in clinical testing (clinical trial premature differentiation and growth restriction of chondrocytes
NCT02401815).168 New areas of investigation into mutant KIT resulting from dysregulated FGFR pathway activation.180,181 FGFR
therapies in GIST include blocking mutant KIT subcellular localization signaling is dysregulated in cancer by multiple mechanisms includ-
to the Golgi150 and combination of FGFR3 and KIT inhibitors to ing mutational or translocation-induced activation of FGFRs, gene
quench pathway cross talk.169 amplification of receptors, and abnormal ligand regulation.182 For
The FMS-like tyrosine kinase 3 receptor (FLT3), a third member example, autocrine and paracrine FGF ligand secretion with resultant
of the RTK class that includes PDGFR and KIT, is involved in the pathway activation has been reported to occur in a subset of melanomas
development of normal hematopoietic cells. It contains an extracel- and prostate cancers, respectively.183,184 FGFR1 amplification occurs in
lular region composed of five immunoglobulin (Ig) domains, TM approximately 17% of squamous cell lung cancers and 6% of small cell
and juxtamembrane domains, and two cytoplasmic tyrosine kinase lung cancers (SCLCs).185 Approximately 10% of diffuse-type, aggressive
domains that transmit proliferative signals through the RAS/MAPK, gastric cancers display FGFR2 gene amplification, and cell lines with
PI3K/AKT, and STAT5 pathways.170 Two main FLT3 alterations are this amplification show ligand-independent pathway activation and
common in hematopoietic malignancies, namely in approximately sensitivity to selective FGFR inhibitors.186,187 Whereas FGFR1 mutations
30% of AMLs.171 First, internal tandem duplication (ITD) within are rather rare, FGFR2 mutations are found in approximately 10%
exons 14 and 15 of the FLT3 gene (FLT-ITD) interferes with the of endometrial cancers.188,189 FGFR3 mutations occur in up to 75%
negative regulatory function of the juxtamembrane segment.172 This of non–muscle invasive bladder cancers and 15% of patients with
duplication results in ligand-independent activation of FLT3 and is advanced urothelial tumors.44,179,189,190 Activating mutations within
associated with a poor prognosis in patients with AML. Second, kinase FGFR3 result in constitutive receptor dimerization and subsequent
domain mutations at or near D835 in the activation loop of FLT3 signaling. Unlike EGFR-activating mutations, which predominantly
disrupt autoinhibitory interactions and render the kinase open and affect the tyrosine kinase domain of the receptor, FGFR3 mutations
active.173 The clinical activity of FLT3 inhibitors has been modest to are commonly located within the extracellular domain (R248, S249)
date, although responses appear to be more common in patients with and TM segment (G370, Y373) and promote ligand-independent
FLT3/ITD AML.171 TAK-659, a reversible dual Syk/Flt inhibitor, receptor dimerization through formation of an aberrant disulfide bridge
showed early clinical activity in numerous lymphoma subtypes and between two receptor monomers.191 Chromosomal rearrangement of
AML.174,175 Sorafenib has been shown in preclinical in vitro studies, FGFRs have been identified using next-generation sequencing. Up to
mouse models, and in a phase I study of AML patients to reduce 15% of multiple myelomas harbor an intergenic 4;14 translocation
leukemia burden and block signaling selectively in FLT3-ITD versus between the FGFR3 gene and the Ig heavy chain locus, which places
FLT-wt settings.176 Interesting to note, resistance to FLT3 inhibition in FGFR3 expression under the highly active heavy chain promoter.192,193
such patients is associated with selection for secondary mutations within More recently, translocations involving FGFR2 have been reported
the tyrosine kinase domain of FLT3, suggesting a central role of FLT3 in in cholangiocarcinoma and more rarely in other cancers, whereas
AML pathogenesis.171 FGFR3 fusions are most common in glioblastoma and bladder
cancers but also are found rarely in other solid tumor types.194,195
Fibroblast Growth Factor Receptor Signaling The FGFR3-TACC3 constitutively active fusion protein has been
characterized to induce aneuploidy by disrupting proper chromosomal
Fibroblast growth factor receptors (highly conserved FGFR1, FGFR2, segregation.196
FGFR3, and FGFR4; and FGFRL1/FGFR5, which lacks a kinase Multiple FGFR inhibitors are currently being tested in early-phase
domain) comprise a family of RTKs that regulate cell proliferation, clinical trials, but the majority of these compounds are multitargeted
differentiation, and migration as well as selective apoptosis during tyrosine kinase inhibitors, many of which also potently inhibit members
embryogenesis. The FGFRs are composed of an extracellular ligand- of the VEGFR and PDGFR families. The close structural similarity
binding domain, a hydrophobic TM region, and an intracellular tyrosine between these RTKs has made development of FGFR-selective inhibitors
kinase domain.177 The extracellular domain is organized into three Ig challenging, although several such drugs are now in early clinical testing,
domains; differential splicing of the second half of the third Ig domain such as BGJ398, AZD4547, JNJ-42756493, and Debio1347 (see Table
dictates tissue-specific expression of the receptor. 2.1).179,182,197–200 On-target hyperphosphatemia resulting from FGFR1
Fibroblast growth factors (FGFs) are protein ligands that bind to inhibition is a primary toxicity with this class of agents, suggesting that
the extracellular domain of the FGFRs in combination with specific the development of isoform-selective FGFR inhibitors may be a more
heparan sulfate glycosaminoglycans inducing FGFR dimerization and rational approach for patients whose tumors are driven by mutations
transphosphorylation of intracellular tyrosine residues. Eighteen FGFs or translocations in FGFR2 and FGFR3. FGFRs are located on the
have been identified, and specificity for FGFRs is based on numerous cell surface and thus may also be susceptible to monoclonal antibody
34 Part I: Science and Clinical Oncology

mediated inhibition similar to trastuzumab-mediated inhibition of growth factors 1 and 2.211 VEGF-A has four isoforms produced by
HER2. FGFR ligand traps are also in development.182 alternative gene splicing, with the 165–amino acid length isoform
playing a central role in tumor angiogenesis.212 Specifically, VEGF-A
RET Signaling enhances vascular permeability and stimulates endothelial cell
proliferation, resulting in new blood vessel formation. Vascular
The RET (Rearranged During Transfection) gene encodes three endothelial growth factor receptors (VEGFR1 to VEGFR3) are
alternatively spliced isoforms (RET9, RET43, and RET51) which are RTKs that possess a modular structure consisting of an extracellular
TM RTKs that contain four cadherin-like extracellular repeats important domain with seven Ig-like regions, a TM domain, and an intracellular
for dimerization, a cysteine-rich juxtamembrane region critical for tyrosine kinase domain.211 VEGF-A, VEGF-B, and placental growth
ligand binding and conformation, and an intracellular kinase domain.201 factor all bind VEGFR1 (also known as FLT1), but the exact role
RET9 and RET51 are highly conserved in all vertebrates and play of VEGFR1 in tumor angiogenesis has yet to be fully elucidated. In
important roles in the normal development and maintenance of many some settings it may act as a decoy receptor that prevents ligand-
tissues, including the kidney, spermatogonial stem cells, and the enteric mediated stimulation of VEGFR2 (also known as FLK1/KDR).213
nervous system.201–203 RET is expressed predominantly on the surface VEGFR2 has been implicated in the development of vasculature
of neural crest tissues, and glial-derived neurotrophic factors (GDNFs) during development and is considered the primary receptor through
such as neurturin, artemin, and persephin serve as ligands for RET. which VEGF exerts its angiogenic effects in endothelial cells.213,214
GDNFs initially bind to their cognate coreceptors, the GDNF receptors Binding of ligand to VEGFR2 results in receptor dimerization and
(GFPα1 to GFPα4), on the cell surface, which recruits RET into transphosphorylation followed by activation of multiple mitogenic
lipid raft membrane domains and causes conformational changes via signal transduction cascades.215,216 More recently, VEGF has been
the cadherin-like moieties, dimer formation, and then subsequent implicated in many angiogenesis-independent roles including regula-
transphosphorylation of tyrosine residues and kinase activation.204 tion of immune cells in the tumor microenvironment, fibroblasts
Y1062 is the common docking site for all three RET isoforms and in the tumor stroma, and cancer stem cells.217 VEGF can also bind
serves to recruit many adapters including SHC1, FRS2, IRS1/2, DOK, and signal through a class of TM glycoprotein coreceptors called
and JNK.201 Phosphorylation of Y752 and Y928 binds STAT3, whereas neuropilins (NRP1 and NRP2), which are found on tumor cells
other phosphorylated residues are recognized by Src, resulting in and can signal along many oncogenic axes including Hedgehog
activation of focal adhesion kinase (FAK), which promotes cell migration and JNK.217
and metastatic spread. In addition, the MAP kinase, PI3 kinase/AKT, The complex network of cross talk among VEGFs, VEGFRs, and
and phospholipase-Cγ pathways can be activated by RET to promote canonic oncogenic pathways makes VEGF and VEGFR critical but
cellular proliferation and survival.204 elusive targets in cancer therapy. Targeted therapies that inhibit VEGF
Germline loss-of-function RET mutations occur in Hirschsprung signaling include antibodies that bind circulating ligand and RTK
and CAKUT (congenital anomalies of the kidney and urinary tract) inhibitors. The humanized monoclonal antibody bevacizumab binds
disease, which causes abnormalities of the developing gut and kidneys, to free VEGF, thereby preventing its association with VEGFRs. This
respectively. Conversely, germline activating RET mutations are the antibody has been FDA approved for use in combination with che-
basis for the multiple endocrine neoplasia type 2 (MEN2) syndromes. motherapy for patients with several cancers, including metastatic
Patients with MEN2 develop familial medullary thyroid carcinomas colorectal218 and nonsquamous NSCLCs.211,219 Bevacizumab also has
and other cancers.205 MEN2A is mainly driven by mutations in six activity in patients with glioblastoma220 and metastatic renal cell
cysteine resides in the RET extracellular domain (C609, C611, C618, carcinoma, where it is often used in combination with interferon-α
C620, C630, and C634), whereas the kinase domain mutations M918T (IFN-α).221 In addition, ramucirumab is a VEGFR2-directed antibody
or A883F are associated with MEN2B. Sporadic medullary thyroid that has received FDA approval with or without chemotherapy in
carcinomas are much more common, and up to 60% of such tumors several cancers.222
harbor somatic mutations in RET, notably G691S, which are thought Sorafenib, sunitinib, pazopanib, and axitinib are multitargeted
to be a driver alteration in this disease.206 Furthermore, RET gene tyrosine kinase inhibitors with nanomolar potency for VEGFR2.
rearrangements with numerous fusion partners, including CCDC6 Sunitinib is used in the treatment of patients with metastatic renal
and NCOA4, termed RET-PTC1 and RET-PTC3, respectively, occur cell carcinoma, GISTs, and pancreatic neuroendocrine tumors.223
in 20% to 40% of papillary thyroid carcinomas (PTCs) and often Sorafenib has been approved for the treatment of liver and renal cell
occur as a consequence of high doses of radiation.207 cancers.224,225 Although these agents inhibit multiple kinases, their
RET inhibitors have shown significant antitumor activity in patients antitumor effects have been attributed primarily to their antiangiogenic
with medullary thyroid cancer. Vandetanib, an oral inhibitor of RET, activity. More recently, the tyrosine kinase inhibitor pazopanib was
EGFR, and VEGFR, is FDA approved for the treatment of patients approved for the initial treatment of metastatic renal cell carcinoma
with advanced medullary thyroid cancer.208 A randomized, placebo- and in cytokine-pretreated patients,226 and axitinib227 was approved
controlled phase III study of cabozantinib, an oral, multitargeted TKI in the second-line setting following failure of prior systemic therapy.
that inhibits RET, VEGFR2, and MET, was also recently conducted Lenvatinib, a multitargeted RTK inhibitor that inhibits VEGFR1,
in patients with unresectable, locally advanced, or metastatic medullary VEGFR2, and VEGFR3, has received recent FDA approval for both
thyroid carcinoma.209 This trial documented a statistically significant thyroid cancer (as monotherapy)228 and renal cell carcinoma in combina-
improvement in median progression-free survival with cabozantinib tion with everolimus.229
as compared with placebo (11.2 months versus 4.2 months in placebo Despite widespread activity in preclinical models, antiangiogenic
arm, P < .0001). More recently, cabozantinib was FDA approved for therapies have shown disappointing activity in several tumor types.
the treatment of renal cell carcinomas that progressed on antiangiogenic A number of resistance mechanisms have been hypothesized to
therapy, although the activity of cabozantinib in this context may not explain the lack of broader clinical activity, including the activation
be attributable to its inhibition of RET. Several RET inhibitors have, of redundant signaling pathways that promote angiogenesis; the
however, shown promising clinical activity in patients with NSCLC recruitment by tumors of bone marrow–derived endothelial progenitor
treated with RET fusions (see Table 2.1).210 cells; increased pericyte density around existing blood vessels, which
enhances vascular growth and survival; and the ability of tumor cells
Vascular Endothelial Growth Factor Signaling to invade surrounding stroma to co-opt additional blood supply.230
A better understanding of these resistance mechanisms may lead
Six vascular endothelial growth factor (VEGF) ligands have been to the development of more effective antiangiogenic therapies in
identified: VEGF-A, VEGF-B, VEGF-C, VEGF-D, and placental the future.
Intracellular Signaling  •  CHAPTER 2 35

Hepatocyte Growth Factor Receptor Signaling Tropomyosin Receptor Kinases/Neurotrophic


Tyrosine Kinase
The hepatocyte growth factor receptor (HGFR or MET) is encoded
by the MET gene.231,232 Both MET and its ligand hepatocyte growth First cloned as an oncogenic fusion partner of the tropomyosin receptor
factor/scatter factor (HGF/SF) are expressed as immature precursors kinases and subsequently characterized for their role in neural dif-
that require proteolytic cleavage.233 The MET extracellular domain ferentiation and survival in the peripheral and central nervous systems,
consists of an alpha subunit connected by a disulfide bridge to a TM the TRKA/B/C family of RTKs, encoded by the neurotrophic tyrosine
beta subunit, and contains a Sema domain, a PSI domain, and four kinase (NTRK) genes 1 to 3 (NTRK1/2/3), respectively, integrate
IPT domains.234,235 The intracellular portion of the receptor contains ligand stimulation from nerve growth factor, brain-derived neurotrophic
a juxtamembrane region that harbors a serine residue (Ser 975) that factor, neurotrophins 3 to 6 with downstream activation of PI3K,
inhibits RTK activity on phosphorylation, as well as a tyrosine kinase phospholipase-C (PLC)-gamma, and MAPK signaling.257–261 Chro-
domain with Y1234 and Y1235 acting as key sites of autophosphoryla- mosomal rearrangements involving the tropomyosin receptor kinases
tion required for activation.235 A tyrosine residue at position 1003, (TrkA/NTRK1, TrkB/NTRK2, and TrkC/NTRK3) were recently shown
proximal to the tyrosine kinase domain, serves as an interaction site to occur at high frequency in several rare cancer types including
for the ubiquitin ligase CBL, which marks the receptor for endocytosis mammary secretory carcinoma of the breast and congenital-infantile
and degradation.236,237 C-terminal residues Y1349 and Y1356 represent fibrosarcoma.262–264 NTRK fusions also occur at low frequency in a
docking sites for adaptor proteins.238 On binding of HGF/SF to the broad range of more common adult solid tumors.260,265 These NTRK
extracellular portion of MET, receptor dimerization occurs, followed fusions induce ligand-independent constitutive kinase activity, resulting
by transphosphorylation. A number of adaptor proteins then bind in upregulation of canonic downstream signaling pathways involved
to phosphorylated tyrosine residues, including Grb2 and GAB1, in growth and survival. TPM3, LMNA, MPRIP, TRIM24, ETV6, and
phospholipase-C (PLC), and SRC, which promotes the activation of PPL, among others, have been identified as fusions partners with the
the MAP kinase and PI3 kinase/AKT signaling pathways.239,240 MET NTRK genes.266,267 Dramatic and durable clinical responses have recently
can also activate RAC1/CDC42 and p21-activated kinase (PAK1), both been reported in patients with NTRK fusions, with first- and second-
of which regulate cytoskeletal proteins and integrin expression and generation TRK inhibitors such as larotrectinib (LOXO-101),
activation, and thus cell migration.238,239 MET also plays an important entrectinib, and LOXO-195 (see Table 2.1).268–273 Notably, clinical
role in driving epithelial-to-mesenchymal transition (EMT) cell migra- activity was observed in both adult and pediatric patients and was
tion during embryo development, and organ regeneration.238,241 independent of site of tumor origin.264
Dysregulation of MET signaling can occur through multiple
mechanisms, including activating point mutations (often in the kinase G PROTEIN–COUPLED RECEPTOR SIGNALING
domain; prevalent in lung cancer), exon skipping events, receptor
overexpression, and upregulation of HGF, which can activate MET in G protein–coupled receptors (GPCRs) are seven TM domain–containing
an autocrine and/or paracrine manner.238,240,242–246 Germline mutations proteins that transduce ligand-specific signals across the plasma
of MET are found in patients with hereditary papillary renal cell membrane to mediate numerous physiologic processes including sensory
carcinomas, and MET overexpression is observed in a significant perception, immunologic responses, neurotransmission, weight regula-
proportion of sporadic papillary cancers as well as collecting duct tion, and cardiovascular activity.274 GPCRs also regulate basic cellular
carcinomas.247 Multiple other malignancies exhibit aberrations in functions including growth, motility, differentiation, and gene transcrip-
MET signaling, including lung, breast, pancreatic, colon, and gastric tion. The GPCR family comprises more than 800 receptors, which
cancers. Amplification of MET is associated with a worse prognosis are the targets of over 30% of all FDA-approved drugs, although few
in lung and gastric cancers, whereas expression of MET or HGF is to date have found a role as anticancer therapies.275,276 Given that
an unfavorable prognostic biomarker in liver, kidney, colorectal, and GPCRs activate many of the signaling cascades that are deregulated
gastric cancers.248 in human cancer, it is not surprising that studies have implicated
Recurrent somatic splice site alterations involving MET exon 14 GPCRs in cancer initiation and progression.277–279
(METex14) have been identified in lung cancer. These mutations GPCRs can be categorized into five or six families, depending on
result in exon skipping, loss of the juxtamembrane CBL E3-ubiquitin the nomenclature used.280 In a more recent phylogenetic classification,
ligase-binding site, diminished receptor turnover, and ultimately, the five major families are represented by the acronym GRAFS:
MET activation.243,249,250 These exon 14 MET mutations are mutually Glutamate, Rhodopsin, Adhesion, Frizzled/Taste2, and Secretin.281
exclusive with activating mutations in EGFR and KRAS as well as The Rhodopsin family of receptors (also referred to as class A GPCRs)
ALK, ROS1, and RET fusions, and treatment of patients with exon 14 is the largest class, with more than 670 members. Crystallization of
MET splice variants with a MET kinase inhibitor is now considered the bovine rhodopsin receptor in 2000 provided the first high-resolution
to be a standard treatment option (see Table 2.1).210 Several RTKs insight into the structure of GPCRs.282 In general, Rhodopsin family
have also been shown to activate MET, including EGFR, HER2, and receptors have short N-termini. Included in this family are the α
IGF1R. For example, EGFR activation can stimulate MET signaling, group (histamine, dopamine, serotonin, adrenoceptors, and muscarinic,
and resistance to EGFR inhibitors in some lung cancers has been prostanoid, and cannabinoid receptors), the β group (endothelin,
shown to stem from coactivation of MET in the setting of gene gonadotropin-releasing hormone, and neuropeptide Y), the γ group
amplification.251 (opioid, somatostatin, and angiotensin), and the δ group (P2RYs,
Inhibitors of MET signaling have been in development for a number glycoprotein-binding FSHR/TSHR/LHCGR, PARs, and olfactory
of years. Therapeutic strategies for targeting MET activation in cancer receptors).283
patients include antibodies that target the extracellular domain of the The 15 Secretin receptors (class B) all have conserved cysteines in
receptor, antibodies that bind to and thus sequester circulating HGF, the first and second extracellular loop, and most have three cysteine
and small-molecule tyrosine kinase inhibitors that selectively target bridges in the N-termini. This class includes the calcitonin-like,
MET or are multi-kinase MET inhibitors.248 Durable responses to corticotrophin-releasing hormone, glucagon-like, gastric inhibitory
crizotinib and cabozantinib, multikinase MET inhibitors, have been polypeptide, growth hormone–releasing hormone, adenylate cyclase–
reported in patients with NSCLC with MET splice mutants or MET activating polypeptide, parathyroid hormone, secretin, and vasoactive
amplification (see Table 2.1).252,253 Cabozantinib is also now standard intestinal peptide receptors.284 The Adhesion family (also included in
of care in MET-amplified renal cell carcinoma.254,255 Combination class B, according to another classification system) has distinctly long
therapies to combat MET reactivation after EGFR kinase inhibitor N-termini, and only 3 of 33 receptors in the family have known
therapy suggest an improvement in progression-free survival.256 ligands (epidermal growth factor-like module containing mucin-like
36 Part I: Science and Clinical Oncology

receptors EMR2, EMR, EMR4).285 The Glutamate receptor family stimulates G protein–regulated inwardly rectifying K+ channels adenylyl
(class C) binds ligands in their N-termini, which have a complex cyclase (types II, IV, VII), PLCβ, PI3Kγ, Src, and GPCR kinases
two-domain folded structure bridged by disulfide bonds. Common (GRKs; see later). It inhibits adenylyl cyclase (type I), some Ca+
receptors in this family of 22 include the glutamate, GABAB, calcium- channels, and calmodulin; stabilizes Gα in the GDP-bound, inactive
sensing, sweet and umami taste (TAS1R1–TAS1R3), and GPCR6 state; and helps specify coupling to the proper Gα member.274,295,296,300
receptors.286,287 The Frizzled receptors (FZD1–FZD10 and SMO; see Gα signaling is switched off by a family of GTPase activating
later for in depth discussion of signaling) bind Wnt ligands in the proteins called Regulators of G-protein Signaling, or RGS proteins,
extracellular domain region containing nine conserved cysteine resi- which enhance the intrinsic rate of GTP hydrolysis by greater than
dues.288 The Taste2 receptors (25 members of T2R, bitter taste) have 1000-fold.312,313 Some RGS proteins enhance signaling through
varying sequence homologies that likely allow the sensing of thousands RhoGEFs and act as scaffolds for other signaling cascades (e.g., tethering
of distinct bitter tastes.289,290 Overall, GPCRs share the seven–TM to Raf and MEK2). The GPCR effectors PKA and PKC also contribute
domain structure, but have different regions of conservation. All of to receptor inhibition by phosphorylating their cognate-activated
the families include orphan receptors, which are related by sequence GPCR and thereby uncoupling and inactivating Gαs/Gαq in a classic
and structure but have no identified ligand to date. negative feedback loop.314,315
All GPCRs have seven α-helical domains that weave through the GPCRs can also function via G protein–independent mechanisms
plasma membrane and are interconnected by flexible extracellular and by binding to the arrestin family of cytosolic adapter proteins.316,317
intracellular segments, thus engendering the synonyms serpentine or GPCR-coupled arrestins have pleiotropic cellular roles including (1)
heptahelical receptors. The specificity of biologic response initiated by dampening G-protein signaling by scaffolding enzymes that degrade
each GPCR depends on (1) ligand recognition; (2) the distinct structure G-protein second messengers; (2) desensitizing receptors by binding
of each receptor class and subclass; and (3) ligand-directed binding GPCR kinase (GRK)–phosphorylated GPCRs and sterically blocking
of specific cytosolic enzymes and adapters that initiate a plethora of access to further Gα subunits; (3) mediating GPCR trafficking and
intracellular signaling cascades (general and cancer-specific examples endocytosis to clathrin-coated pits; and (4) acting as a scaffold for
are discussed further later). The GPCR Network was created in 2010 multiple MAP kinase cascades. For example, MEK1 is engaged by a
to tackle the challenge of delineating the structure and function of tethered complex of Raf-1, ERK2, and β-arrestin to facilitate mitogenic
this diverse class of receptors.291 signaling.318,319
Operationally, ligand binding on the GPCR extracellular surface GPCRs are well established drug targets for antihistamine, antacid,
induces a conformational change in the receptor, mainly via TM cardiovascular, and antipsychotic drugs; pain suppressants; and
helices TM3, TM5, and TM6, which creates a deep pocket in the antihypertension therapies.275,276 Although less well appreciated as drug
intracellular face of the receptor.292–294 This cleft enables binding and targets in cancer, there is increasing evidence that dysregulated GPCR
activation of heterotrimeric G proteins, which consist of an inactive, signaling contributes to cancer initiation and progression. For example,
GDP-bound Gα subunit and a Gβγ-subunit dimer, which act as a in 1986 the wild-type MAS1 gene, which encodes the MAS GPCR,
molecular switch.295,296 Activated GPCRs promote GDP for GTP was reported to induce transformation by coupling to the small G
exchange on the Gα nucleotide binding site.297 This GTP-bound Gα protein Rac.320,321 Large-scale deep sequencing efforts have revealed
dissociates from Gβγ and the receptor, and then both activated subunits that GPCR mutations occur in approximately 20% of all cancers.
go on to initiate signaling cascades. There are four members of the Receptors for thyroid-stimulating hormone (TSHR), Hedgehog
Gα family, Gαs, Gαi/o, Gαq/11, and Gα12/13, which can be further (Smoothened receptor [SMO]), glutamate (GRM), the adhesion family,
subtyped and can each stimulate several downstream effectors. Moreover, lysophosphatidic acid (LPA), and sphingosine-1-phosphate (SIP) are
each GPCR can couple to multiple Gα family members, thus generating the most frequent GPCRs altered in cancer.277,312,313,322 G proteins
a complex pattern of intracellular signaling. themselves are also mutated in cancer. In particular, recurrent mutations
Classic GPCR activation of Gαs stimulates adenylyl cyclase, which in GNAS (which encodes Gαs) have been identified in thyroid and
generates the second messenger 3′-5′-cyclic adenosine monophosphate pituitary tumors, as well as mutations in GNAQ and GNA11 in
(cAMP).298,299 cAMP activates multiple downstream effectors including melanoma of the eye and skin, respectively.322 Hot spot resides that
cAMP-gated ion channels; Epac, a GEF for Rap1/2 (which functions disrupt GTPase activity and render the protein constitutively active
in cell adhesion and junction formation); and protein kinase A have been identified in GNAS at positions R201 and Q227, in GNAQ
(PKA).274,295,300–303 cAMP binds to PKA regulatory subunits, releasing at R183, and in GNA11 at Q209. Numerous inhibitors of GPCR
catalytic subunits and triggering the activation of cytosolic and signaling are also being studied in the clinic including BKT-140/
nuclear substrates, including the transcription factor CREB (cAMP BL-8040 in pancreatic adenocarcinoma and blood cancers (target:
response element binding protein), which can induce proliferation and CXCR4 receptor), and CXCR2 ligands (target: CXCR2 receptor).278
differentiation, among other phenotypes, depending on cell origin.304–306 Detailed later are a few specific examples of GPCRs that have been
Gαs also activates the Src tyrosine kinase and the GTPase activity of shown to play a role in cancer initiation and/or progression.
tubulin.295,300 GPCR-coupled Gαi/o typically works in opposition to A connection between GPCRs and EGFR signaling has been
Gαs by inhibiting adenylyl cyclase and decreasing cAMP levels.274,307 established in both normal cell physiology and in colon, lung, breast,
In addition, some Gαi/o isoforms can signal to K+ and Ca+ channels, ovarian, prostate, and head and neck cancer development.323–326
increase cGMP phosphodiesterases, interact with Rap1GAP1, Ligand-bound GPCRs activate Src, PKC, Ca+ channels, and PKA
and cross talk to the MAP kinase pathway (as described later in intermediaries, which stimulate proteolytic cleavage and release of
the chapter).274 membrane-tethered growth factors that bind and thereby transactivate
Members of the Gαq/11 family activate phospholipase-Cβ, which EGFR. Specifically, estrogen binding to the GPCR GRP30 facilitates
catalyzes the hydrolysis of PIP2 to yield IP3 and DAG.308 IP3 mobilizes matrix metalloproteinase–2 (MMP2) and MMP9-mediated cleavage of
calcium from intracellular stores, whereas DAG activates some isoforms the growth factor precursor pro-heparin-binding-EGF (pro-HB-EGF),
of protein kinase C (PKC).309,310 Both Gαq/11 and Gα12/13 activate a thus initiating HB-EGF–mediated EGFR transactivation.324 Through
variety of RhoGEFs (p115-RhoGEF, PDZ-RhoGEF, LARG, Lbc, a related mechanism, LPA-, SIP- and thrombin-activated GPCRs
AKAP-Lbc) and thus regulate Rho activity (mainly RhoA) and its transactivate EGFR in breast cancer cells via growth factor shedding
contribution to actin stress fiber formation, cell shape and polarity, cell of tumor necrosis factor–α (TNF-α) through the action of TACE/
adhesion and migration, gene transcription, and cell cycle progression.311 ADAM17 zinc-dependent proteases.326 Thrombin-mediated N-terminal
In addition, Gα12/13 activates the Na+/H− exchanger, inducible nitric cleavage and activation of proteinase-activated receptor 1 (PAR1), which
oxide synthase, phospholipase D, E-cadherin, radixin, and protein can act through EGFR, has been found to promote metastasis and
phosphatase 5. Gβγ signaling is equally complex, as the active dimer invasiveness in melanoma, breast, colon, and prostate cancers.278,279
Intracellular Signaling  •  CHAPTER 2 37

Intriguingly, MMP1 was found to function similarly to thrombin adenomatous polyposis (FAP).357 Efforts to develop selective inhibitors
in activating PAR1 and promoting breast cancer tumorigenesis and of Wnt signaling are ongoing and will be aided by current endeavors
invasion.327,328 This cross talk between GPCRs and EGFR provides to crystallize members of the Wnt cascade.354,358 Of note, nonsteroidal
a rationale for the combinatorial use of GPCR agonists/antagonists antiinflammatory drugs (NSAIDs) have shown some promise in
and EGFR inhibitors in patients with EGFR-driven lung and modulating Wnt signaling, likely by inhibiting the Wnt-output
colorectal cancers. gene COX-2 or by enhancing E-cadherin signaling.349,359 COX-2
Aberrant GPCR signaling through Gα12/13 also contributes to inhibitors have shown efficacy in reducing the risk of polyps in patients
tumorigenesis by enhancing cancer cell migration, invasion, angio- with FAP.358,360
genesis, and metastasis.329 Ligand-activated LPA, PAR1, SIP, throm-
boxane A2 (TP), CXC chemokine (CXCR4), and prostaglandin E2 CYTOKINE RECEPTOR SIGNALING
(PGE2) receptors couple to Gα12/13 and RhoGEFs to hyperactivate
RhoA (see earlier), which elicits these progression-associated phenotypes Cytokines are protein and glycoprotein ligands secreted by immune
in glioma, melanoma, lung, breast, and ovarian cancers.278,279 Over- cells; they initiate diverse and often opposing effects based on target
expression of Gα12/13 and RhoA in breast, prostate, and colon cancers cell lineage. Processes regulated by cytokine signaling include cell
also promotes metastasis by decreasing cell adhesion.311 RhoGEF proliferation, differentiation, survival, inflammation, angiogenesis,
inhibitors; RhoGTPase inhibitors; inhibitors of prenylation, which antiviral activity, and modulation of immune function. Cytokines
could indirectly impair proper Rho localization (statins, farnesyl/ signal in an autocrine and/or paracrine fashion and can be subclassified
geranylgeranyl transferase inhibitors); and inhibitors of kinases by protein structure into four families, which total over 100 members:
downstream of Rho (ROCK, LIMK, MRCK, PAK) have all been hematopoietins, IFNs, chemokines, and the TNF superfamily.
developed and tested in biochemical, cell line, and mouse experiments. The hematopoietin family consists of interleukins (IL-1 to IL-31),
However, a clear benefit to the use of such compounds in cancer growth hormone, prolactin, erythropoietin, thrombopoietin, leptin,
patients has yet to be established.330 granulocyte colony-stimulating factor and granulocyte-macrophage
Two other prominent examples of dysregulated GPCR signaling colony-stimulating factor, and a few others.361 The majority bind to
in human cancer are the Hedgehog/Smoothened and Wnt/Frizzled either class I or II cytokine receptors, which are single TM glycoproteins
signaling pathways.277,278,331 Both pathways, along with Notch, also that lack kinase activity and diverge in their extracellular domains in
play critical roles in cell fate and have been implicated in the underlying order to specify ligand binding.362,363 Class I receptors function as a
pathway cross talk that is key to cancer stem cells.332 Secreted Hedgehog cluster of two or three subunits that each have two sets of conserved
(Hh) ligands (first identified based on their roles in normal development cysteine pairs and a WSXWS motif in their external domains. Class
and stem cell homeostasis, with Sonic Hedgehog [SHH] being the II receptors lack the WSXWS motif and one of the class I conserved
most ubiquitous) bind to the 12-pass TM receptor Patched (PTCH), cysteine pairs, but contain conserved proline, tryptophan, and an
which relieves its repression of the GPCR Smoothened (SMO).333,334 additional two conserved cysteine residues. Ligand binding causes
Activated Smoothened couples to Gαi and Gα12, which regulate the aggregation of the γ-chain (also called γc, CD132) which is common
glioma-associated oncogene homologue (GLI) transcription factors, to many cytokines, and the β-chain (also known as IL-2Rβ, IL-15Rβ,
which in turn regulate proliferative, survival, and differentiation signals or CD122).364,365 In the case of specific cytokines, such as IL-2,
involving cyclin D1, myc, BCL2 and the Forkhead transcription factors, association with a third subunit, the α chain (also called IL-2Rα,
to name a few.335,336 Mutations in SHH, PTCH, and SMO are found CD25, or Tac antigen), allows for high-affinity ligand binding.366,367
in patients with inherited and sporadic basal cell carcinomas337,338 and The IFN family members are divided into type I, II, and III
ameloblastomas,339 whereas overexpression of Hh ligands has been classes.368,369 Most IFNs are type I and can be further subtyped.370 All
shown to result in hyperactivation of the pathway in breast, colon, type I IFNs bind the type I IFN receptor, which consists of two
prostate, and pancreatic ductal adenocarcinomas331,340 Vismodegib and subunits (IFNAR1 and IFNAR2). The sole type II IFN, IFN-γ, binds
sonidegib, both selective Smoothened receptor inhibitors, are FDA the type II IFN receptor, which is composed of the IFNGR1 and
approved for the treatment of advanced basal cell carcinomas (see IFNGR2 subunits. Both types of IFN receptors belong to the class
Table 2.1).341–344 Several other Hh/SMO inhibitors are currently being II cytokine receptor family. The IFN family also includes a third
tested in patients with cancer, mostly for advanced and/or metastatic branch, the IFN-like molecules, IFN-λ1 (IL-29), IFN-λ2 (IL-28A),
solid tumors.278,345,346 and IFN-λ3 (IL-28B), which display some structural overlap with
The secreted Wnt glycolipoprotein ligands activate the single ILs and the antiviral properties of IFNs, and bind a distinct receptor
TM low-density lipoprotein–related coreceptors LRP5/6 and the made up of IFNLR1/IL-28Rα and IL-10Rβ.371 Given that cytokine
GPCR-like TM protein Frizzled (Fz), which are phosphorylated and receptors are promiscuous and bind multiple cytokine ligands, there
likely couple to Gαq and Gαo, respectively, to activate the cytoplasmic is a high degree of redundancy in the output profiles of individual
scaffold Dishevelled.347–349 Dishevelled in turn inhibits the β-catenin cytokines. This redundancy serves to amplify and sustain signaling
degradation complex (which consists of APC, axin, CKIα and GSK-3β, downstream of these transitory stimuli.
and the E3 ubiquitin ligase β-TrCP).350 This results in accumulation Hematopoietin or IFN binding induces oligomerization of cytokine
of β-catenin, which translocates to the nucleus where it induces TCF/ receptor subunits and autophosphorylation and activation of the JAK
LEF-mediated transcription of genes important for cell differentiation (Janus activated kinase) family of intracellular tyrosine kinases (TYK2,
and proliferation, including myc, cyclin D1, VEGF, FGF4/18, E-cadherin, JAK1–JAK3), which are constitutively bound to box I and box II
COX-2, and members of the Wnt cascade itself.349,351,352 Noncanonic α-helix motifs in the receptor cytoplasmic tail.372 Activated JAK proteins
Wnt/Fz pathways include signaling to the transcription factors phosphorylate tyrosine residues on the cytokine receptor chains, which
NFAT via Gαq/i/Gβγ/PLC/PKC/Ca+ (Wnt-calcium pathway) and classically bind the STAT (signal transducer and activator of transcrip-
AP1 via Rho/Rac/JNK (planar cell polarity pathway).277,353 These tion) family of transcription factors (STAT1–STAT6).361,373 Docking
pathways regulate cell polarity and migration and are implicated in of STATs facilitates their phosphorylation by JAKs, which in turn
cancer metastasis.353 Aberrations in canonical Wnt signaling promote causes STAT dimerization, nuclear translocation, and alterations in
tumorigenesis in melanoma and colon, liver, ovarian, and prostate gene transcription.374 There is also cross talk between STAT signaling
cancers.354,355 Specifically, loss-of-function mutations or truncations and the nuclear factor–κB (NF-κB) and SMAD signaling pathways.375
in APC and AXIN1/2 and gain-of-functions mutations in β-catenin STATs are also activated by RTKs, SRC, and ABL, and STAT activation
are found in almost all colorectal cancers, with APC alteration found also plays a role in transformation initiated by these oncogenes.376
in over 85%.349,356 Germline mutations in the APC gene are also JAK/STAT activity is regulated by several posttranslational modifications
the basis for the inherited cancer predisposition syndrome familial as well as by the SOCS (suppressor of cytokine signaling) and PIAS
38 Part I: Science and Clinical Oncology

(protein inhibitor of activated STAT) proteins.375 In addition to response.383 JNK1 is upregulated in hepatocellular carcinoma and
JAK-STAT signaling, cytokine receptors can transduce messages through prostate cancer, whereas p38α activity is either lost (in hepatocellular
LCK and SYK (Src-family kinases), through BCL-2, and via PI3 carcinoma) or activated in numerous cancers, and targeted inhibitors
kinase/AKT and Ras/Raf mediated upregulation of Fos- and Jun- are being developed.385,397
dependent transcription.376,377
The 29-member TNF superfamily of receptors (TNFSFR1) and SERINE/THREONINE RECEPTOR SIGNALING
their corresponding 19 ligands (TNFSF) induce inflammation in
addition to prosurvival and proapoptotic phenotypes.378,379 TNF ligands Receptor serine/threonine kinases are exemplified by the TGF-β type
are TM proteins that function as membrane-integrated or cleaved, I and II receptors.398,399 Ligands for these receptors include the TGF-β
soluble trimers that bind and activate preformed, single-TM TNF superfamily, which comprises the TGF-β1-3 isoforms, activins, inhibins,
receptor trimers on the cell surface.380 Conserved cysteine residues in Nodal, and Lefty, and the more distantly related bone morphogenic
the TNF receptor external domains dictate ligand binding, and the proteins (BMPs), growth and differentiation factors (GDFs) and
presence of death domains (DDs), TRAF-interacting motifs (TIMs), müllerian inhibitory substance (MIS). TGF-β ligands regulate a diverse
or neither (decoy) dictate downstream signaling. For example, on array of physiologic processes including growth, proliferation, survival,
apoptotic stimuli, ligand activation of the TNF-R1 and DR3 receptors hormone release, and differentiation.398,400 They thus have a central
recruits the adaptor TRADD (TNFR-associated death domain) via role in embryonic patterning, tissue development, and morphogenesis.
DDs, which in turn binds to FADD (Fas-associated protein with Signaling mediated by TGF-β, the namesake and most studied ligand
death domain).378,379,381 FADD binds procaspase-8 and procaspase-10 of the superfamily, will be used to exemplify the general structure of
via death effector domains (DEDs), and induces their cleavage to the serine/threonine kinase cascades.
form active enzymes, which cleave caspase-3 and induce apoptosis. TGF-β is ubiquitously expressed and requires a multistep maturation
TRADD binding is also capable of inducing the intrinsic apoptotic and secretion process to be functional and bioavailable. Initially
cascade (mitochondrial release of ROS, cytochrome C, and Bax, which translated as an immature proprotein, the prodomain (called latency-
leads to caspase-9 and caspase-3 activation and apoptosis).378,382 associated protein [LAP]) is cleaved and noncovalently bound to the
Under proliferative stimuli, TNF-α–dependent activation of remaining mature form of TGF-β.401 Covalently bound mature, active
TNF-R1/TRADD and TNF-R2 converges on the recruitment of dimers are further bound to latent TGF-β binding protein (LTBP)
TRAF2 (TNFR-associated factor 2). TRAF2 binding sequentially such that this complex is sequestered by LTBP binding to the extracel-
recruits the RIP (receptor interacting protein), TAK1 (TGF-β activated lular matrix (ECM) until appropriate signals initiate matrix metal-
kinase 1), and IκB kinase (IKK) trimer; this functions to degrade loproteinase, plasmin, or thrombin-dependent cleavage of LTBP and
IκB-α (inhibitor of NF-κB-α) and in turn facilitates the activation subsequent release of active TGF-β dimers.401,402 TGF-β dimers bind
and translocation of NF-κB.378,379 NF-κB in turn induces mediators constitutively active, single TM, homodimeric TGF-β type II receptors
of inflammation and cytoprotective phenotypes.383 Alternatively, TAK1 (TβRII). Once bound by ligand, TβRII forms a complex with TGF-β
can signal to MKK3/6 (MAP kinase kinase 3/6) and to two MAP type I (TβRI) receptor homodimers, thus creating a receptor hetero-
kinases, ERK (proproliferation) and p38α (to activate the transcription tetramer.398,400 TβRII receptors phosphorylate and activate the intrinsic
factor AP-1 [activator protein-1]).384 Upstream, TRAF2 can additionally kinase activity of TβRI, which in turn phosphorylates serine residues
trigger MEKK1 (MAP/ERK kinase kinase 1), MKK7, and JNK (c-Jun in the C-terminal–SSXS motif of receptor-activated SMAD proteins
activating kinase), the recruitment of which also converges to activate (R-SMAD2 and R-SMAD3 for TGF-β).403 Activated R-SMAD2 and
AP-1 and thus regulate proliferation and survival.385–387 There is an R-SMAD3 then form a heterotrimer with SMAD4, which then localizes
important avenue of cross talk between the TNF-directed NF-κB and to the nucleus, where it both binds DNA and partners with other
JNK pathways, the balance of which ultimately decides cell fate.388 transcription factors (i.e., Forkhead, homeobox, zinc-finger, AP-Ets,
In cancer, dysregulation of cytokine signaling promotes chronic and bHLH family transcription factors) and cofactors (e.g., p300,
inflammatory signals and prevents the immune system from attacking CBP).404,405 These SMAD-containing complexes then target selective
cancer cells. Unfortunately, because of their pleiotropic effects, which promoter elements with high affinity, leading to the induction or
are often cell type and microenvironment specific, therapeutic strategies repression of hundreds of genes, depending on cell context. For example,
that modulate cytokine signaling have demonstrated only modest TGF-β induces the expression of 4EBP1 and the cyclin-dependent
clinical activity in patients with cancer. For example, the recombinant kinase inhibitors INK4B and p21 and represses Myc to elicit growth
human IFN-α2a mimetic, Roferon-A, has been shown to inhibit inhibitory effects.406 It also activates DAPK (death-associated protein
tumor growth in patients with melanoma and hairy cell leukemia, kinase), GADD45β (growth arrest and DNA damage-inducible 45β),
but such treatments have now been supplanted by more active thera- and BIM to promote apoptosis and PDGF in smooth muscle cells
pies.389 Moreover, focal delivery of TNF-α to limbs affected by soft to enhance proliferation, among other effects.407 TGF-β also signals
tissue sarcomas and melanomas through isolated limb perfusion through many non-SMAD effectors including Shc, which can result
techniques has been more effective than systemic use, which is in enhancement of Ras/ERK signaling, and TRAF6, which activates
toxic.390,391 Proapoptotic, anti-TRAIL therapies, such as mapatumumab, the TAK1/MKK3&6/JNK/p38 cascades.408 TGF-β, through indirect
are also in clinical testing.381 mediators, can also activate Src, Rho, and PI3 kinase, and through
Downstream components of the cytokine signaling cascades are pathway cross talk the Wnt, Hedgehog, and Notch cascades.409 The
also being explored as targets for drug development. Gain-of-function outcome of TGF-β–directed transcriptional responses depends on
mutations in the JAK2 (hot spot V617F) and MPL genes, the latter access of TGF-β to particular signaling receptors and SMAD complexes,
of which encodes the thrombopoietin receptor, are common in the availability of transcription factors, and the epigenetic status of
myeloproliferative neoplasms, and the selective JAK1/2 kinase inhibitor the cell.410
ruxolitinib has been approved for this indication (see Table 2.1).392–394 Alterations in TGF-β signaling are common in cancer.407,411,412
STAT3 is frequently hyperactivated in cancer, because it is a downstream For example, mutational inactivation of TβRII is seen in colorectal
effector of both cytokine receptors and mutated and amplified tyrosine cancers with microsatellite instability.413 Mutations and loss of SMAD4
kinases.376,395 Constitutive activation of STAT5 and STAT6 plays a expression are common in colorectal, pancreas, and head and neck
critical role in BCR-Abl–driven chronic myelogenous leukemia (CML), cancers.414,415 Conversely, increased TGF-β expression occurs in breast,
and IL-13–driven lymphomas and leukemias.376 On the basis of these prostate, and colorectal cancers and has been associated with cancer
findings, direct inhibitors of JAK and STAT are currently in develop- progression and the development of metastases.407 TGF-β has also
ment.374,396 Inhibition of NF-κB has been shown to induce apoptosis been shown to play a role in maintaining the tumor-initiating cell
in leukemias and lymphomas and enhance chemotherapy and radiation or cancer stem cell population in gliomas, leukemias, and breast
Intracellular Signaling  •  CHAPTER 2 39

cancer.407 It has been proposed that TGF-β signaling promotes clinical trials.422,434–436 Newer avenues of drug development include
invasion and metastasis by promoting EMT.406 Interesting to note, monoclonal antibodies targeting Notch receptors or ligands.437
in pancreatic ductal adenocarcinoma, wherein loss of SMAD4 is
common, TGF-β can play a novel tumor suppressor role by inducing NUCLEAR HORMONE RECEPTOR SIGNALING
a lethal EMT through the lineage and progenitor regulators Klf5
and Sox4.416 The nuclear hormone superfamily is composed of hormone receptors
Intense efforts are underway to develop therapeutic strategies that and orphan receptors (receptors for which no ligand has yet been
inhibit the tumorigenic properties of TGF-β signaling. These include identified). The nuclear hormone receptors are characterized structurally
the development of selective TGFβRI inhibitors, TGF-β blocking by a ligand binding domain, a DNA binding domain, and a hinge
antibodies, soluble TGF-β antisense therapies, and selective kinase region that connects the ligand and DNA binding domains.438 Nuclear
inhibitors.411,417,418 The development of inhibitors of TGF-β signaling hormone receptors are classified into four subtypes based on ligand
has, however, been confounded by the ability of TGF-β to both promote specificity: steroid, retinoid X receptor (RXR), monomeric or tethered
and suppress tumor progression in a context-specific manner.419,420 orphan receptors, and dimeric orphan receptors.439 The steroid receptor
ligands include estrogen, progesterone, androgen, and growth hormone.
NOTCH RECEPTOR SIGNALING Ligand binding occurs in the cytoplasm and results in receptor
homodimerization followed by nuclear translocation. Once translocated
The mammalian Notch receptors, Notch1 to Notch4, are single-pass into the nucleus, the ligand-bound receptor acts as a transcription
TM receptors that are functionally unique with regard to receptor factor that modulates the expression of several downstream proteins
processing and signal activation and transduction.421–424 Notch receptors through binding to steroid response elements, which are conserved
are translated as immature receptors that undergo S1 cleavage by nucleotide sequences within the regulatory regions of genes.440 In
furin-like convertase during Golgi trafficking.425 This generates two contrast to the steroid receptors, the RXR receptors form heterodimeric
subunits that are retethered at the cell membrane by noncovalent complexes with other partners, including the retinoic acid receptor,
bonds in the heterodimerization domain.423,426 The extracellular domain the thyroid hormone receptor, and vitamin D receptors.
subunit consists of ligand-binding EGF-like repeats, a heterodimer Hormone receptor blockade is a cornerstone in the treatment of
domain, three negative regulatory LIN12 and Notch repeats (LNRs) estrogen receptor (ER)– and progesterone receptor–expressing breast
containing numerous cysteines, and a hydrophobic TM-interacting cancers. The selective estrogen receptor modulator (SERM) tamoxifen
region. The other subunit contains the TM domain and the intracellular competes with estradiol for binding to ER. Notably, tamoxifen binding
domain, which has ankyrin repeats and a RAM domain central to results in ER dimerization, nuclear translocation, and receptor binding
the Notch receptor’s unusual activity as a direct transcription factor, to estrogen response elements in the promoter regions of estradiol-target
and a PEST motif important for the quick termination of Notch genes. It is thought that the ER/tamoxifen complex recruits transcrip-
activity and ubiquitin-mediated degradation. tional corepressors, in contrast to ER/estradiol binding, which recruits
Delta-like ligand (DLL1, DLL3, DLL4) and Jagged (JAG1 and transcriptional coactivators.440,441 Although tamoxifen has antiprolifera-
JAG2) are the five TM-protein ligands for the Notch receptors.427 A tive effects in ER-expressing breast cancer cells, it causes hypertrophy
Notch receptor on one cell binds to DLL or JAG ligand on an adjacent and neoplastic transformation of endometrial tissue, likely as a result
cell. This results in a change in Notch receptor conformation, which of cell type and context-specific recruitment of transcriptional coactiva-
facilitates a series of proteolytic cleavages required for receptor activation. tors that are differentially expressed in these tissues.440 Recently,
First, the ADAM17/TACE (a disintegrin and metalloprotease-17/ mutations in the ESR1 gene, which encodes the ER, have been shown
TNF-α converting enzyme) metalloprotease performs S2 cleavage of to be a common mechanism of acquired resistance to hormonal therapy
the extracellular domain.428,429 This allows subsequent S3 cleavage of in patients with breast cancer.442 Retrospective studies suggest that
the TM domain by the γ-secretase complex and release of the Notch patients with ESR1-mutant breast cancer may have more durable
intracellular domain (NICD).430 The active NICD translocates to the responses to the selective estrogen receptor downregulator (SERD)
nucleus, where it binds to and converts the repressor complex CSL fulvestrant than those treated with the aromatase inhibitor exemestane
(CBF1, Suppressor of Hairless, Lag-1) into a transcriptional activator (see Table 2.1).442,443
that recruits coactivators including the Mastermind-like family proteins Dihydrotestosterone is the primary ligand of the androgen receptor
and p300. NICD target genes include the HES (hairy enhancer of (AR). AR blockade by antiandrogens such as bicalutamide is a com-
split) and HRT/HEY (hair-related transcription factor) transcriptional monly used therapeutic modality for patients with locally advanced
repressors, as well as cyclin D1, myc, p21, NF-κB, and the Notch or metastatic prostate cancer. Bicalutamide competes with dihydrotes-
receptors and ligands themselves, among others.422,426,431 Notch is also tosterone for binding to cytoplasmic AR.444 The mechanism by which
heavily involved in cross talk with other pathways, including RTKs, bicalutamide-bound AR inhibits androgen-dependent gene transcription
PI3 kinase, Ras/ERK, JAK/STAT, Wnt, Hedgehog, TGF-β/SMAD, is unclear but may involve the recruitment of transcriptional corepressors
and p53. Overall, Notch plays a role in proliferation, cell fate determina- as well as histone modifications that lead to tighter chromatin binding
tion in development, and survival. and therefore reduced access to promoter regions by transcription
Notch’s causal role in tumorigenesis was established on its discovery factors. Enzalutamide, a nonsteroidal small-molecule antagonist of
in 1991, when a translocation event in T-cell acute lymphoblastic the AR that inhibits AR nuclear translocation and DNA binding is
leukemia or lymphoma (T-ALL) was identified that fused the T-cell FDA approved for the treatment of patients with metastatic prostate
receptor-β promoter/enhancer elements to Notch, generating a cancer that has progressed following treatment with bicalutamide and
truncated form of Notch1 that was constitutively nuclear and active.432 medical castration (see Table 2.1).445 Abiraterone acetate, an irreversible
Activating NOTCH1 mutations have since been found to occur in inhibitor of CYP17A1 that enables intratumoral and adrenal androgen
approximately 60% of T-ALL patients.433 Subsequent studies have depletion, also has significant clinical activity in patients with castrate-
determined that Notch and its ligands have both oncogenic and tumor resistant prostate cancer.446
suppressor properties in several hematologic and solid tumor
malignancies, including melanoma, glioblastoma, and breast, lung, INTEGRIN RECEPTOR SIGNALING
colorectal, and pancreatic cancers, depending on cellular context.423,426
Thus far, efforts to develop inhibitors of Notch signaling have focused The integrin receptor family regulates cell adhesion, migration, invasion,
primarily on inhibiting cleavage, and thus activation of Notch. and cell survival.447,448 Integrin receptors are heterodimeric molecules
Specifically, selective γ-secretase inhibitors (GSIs), such as MK-0752, consisting of combinations of alpha and beta subunits. Each combina-
PF03084014, and BMS-906024, are currently being tested in early-stage tion dictates the spectrum of ECM components to which these receptors
40 Part I: Science and Clinical Oncology

bind. Once ligated to the ECM, the receptors recruit multiple proteins ubiquitously, whereas the tissue distribution of the latter six is more
to the cell membrane, including cytoskeletal molecules such as paxillin restricted.467 Together, SRC family kinases have pleiotropic roles in
and vinculin that form focal adhesions to ECM components.449 Unlike cellular proliferation, apoptosis, differentiation, motility, adhesion,
the RTK family, integrin receptors do not possess intrinsic kinase angiogenesis, and immunity.468,469
activity but rather promote signaling by facilitating the activation of SRC is by far the most intensively studied family member and
kinases such as Src or FAK.450 Integrins are also unique in that they was the first gene observed to have oncogenic potential.470 Peyton
participate in bidirectional signaling. “Inside-out” signaling occurs Rous was awarded the Nobel Prize for a series of experiments showing
when intracellular adapters, of which talin-1 and kindlin-1/2/3 are that a transmissible factor was present in avian sarcomas capable of
the best known activators, trigger a conformational change of the initiating tumors in recipient birds. Five decades later the viral oncogene
cytoplasmic tails of the alpha and beta subunits, which is transduced v-Src was identified as the oncogenic factor in the Rous sarcoma
to the extracellular component of the receptor, resulting in increased virus.471–473 Bishop and Varmus later showed that v-Src was a mutant
affinity for portions of the ECM.451 Conversely, “outside-in” signaling form of the cellular proto-oncogene c-Src, and Hunter and colleagues
involves binding of ligand to integrins, which stimulate the activation showed that its transformative capacity was dependent on its tyrosine
of multiple intracellular signaling pathways.452 Collectively, the term kinase activity.14,15,474
integrin adhesome is used to describe the site at which integrins initiate SRC is regulated in a number of ways. First, SRC has a myristoylation
contact with the ECM and other cells and recruit all of the underlying site in its N-terminus that is necessary for membrane localization and
intracellular machinery (e.g., cytoskeleton, scaffolds, signaling adapters— that promotes its interaction with nearby membrane-bound effectors.475
over 200 intrinsic and transient components) to generate diverse types The SH2 and SH3 domains facilitate protein-protein interactions and
of adhesions (e.g., focal complexes, focal adhesions, fibrillar adhesions, conformational changes in the protein. Inactive SRC is maintained
podosomes, invadopodia).453 in a closed conformation with phosphorylated Y530 (mediated by
Integrins are expressed on cancer cells and have been shown to CSK, C-terminal SRC, and Csk homology kinases) interacting with
promote disease progression.447,450,454 Integrins are also present on the folded-over SH2 domain.476 The closed, inactive confirmation
stromal cells, including pericytes (which promote endothelial cell of SRC is further stabilized by proline-rich segments of the kinase
growth and proliferation and thus angiogenesis) and fibroblasts, where domain associating with the SH3 domain.477 SRC activation requires
they influence the surrounding microenvironment and thus indirectly dephosphorylation of Y530, likely by PTPα/γ/1β (protein tyrosine
stimulate tumor growth and proliferation. For example, vascular cell phosphatase α/γ/1β) or SHP1/2 (SH-containing phosphatases), which
adhesion molecule 1 (VCAM1) is expressed on pericytes and binds allows the kinase to assume an open conformation.466,478 Autophos-
to the integrin receptor α4β1, which is found on the surface of phorylation of Y419 in the activation loop of the kinase domain also
endothelial cells, resulting in pericyte recruitment to sites of vascular promotes full activity,479 whereas binding of FAK and CRK-associated
maturation.455 Integrin signaling also plays a role in the activation of substrate (CAS) to the SH2 domain induces SRC activation and links
matrix metalloproteinase 2,456 which promotes cell invasion, and has SRC signaling to the regulation of focal adhesion, actin reorganization,
been shown to regulate cyclin D and cyclin-dependent kinase inhibitor and migratory phenotypes.480,481 SRC is a downstream mediator of
expression, thereby controlling cell cycle progression.457 Finally, integrin numerous receptor families including RTKs, integrin receptors, hormone
receptor activation can lead to increased secretion of growth factors, receptors, cytokine receptors, and GCPRs and promotes signaling
which then stimulate tumor invasion through autocrine and paracrine through the PI3 kinase/AKT, Ras/MAP kinase, and JAK/STAT cascades,
mechanisms.456 among others.466,467,469,478,482–486 More than two decades of research
Tumors that express integrin receptors include melanomas, glio- have uncovered numerous SRC substrates, including p85-cortactin,
blastomas, and breast cancers. In melanoma, the αvβ3 and α5β1 p110-AFAP1, p130Cas, p125FAK, and p120-catenin.487
integrin receptors promote vertical growth and metastatic spread to Although mutations in SRC are rare in human cancers, SRC is
lymph nodes.458,459 In glioblastoma, αvβ3 and αvβ5 are expressed frequently activated as a consequence of other mutational events in
mainly at the edge of tumors, suggesting a role in tumor invasion.460 colorectal, breast, esophageal, gastric, pancreatic, hepatocellular, ovarian,
The expression of the α6β4 and αvβ3 integrin receptors in breast and lung cancers.466 In colorectal and hepatocellular carcinomas, SRC
cancer is associated with higher grade and tumor size,461 the development activation occurs in the setting of concomitant loss of CSK.488–490 Newer
of bone metastases,462 and decreased survival.463 To date, drug develop- signaling discoveries have identified roles for SRC in promoting tissue
ment has targeted three integrins—αIIbβ3, α4β1, and α4β7—in the repair after intestinal inflammatory injury, as seen in inflammatory bowel
context of platelet activation and blood clotting, multiple sclerosis, diseases and colorectal cancer, via the IL-6 cytokine coreceptor gp130
and inflammatory bowel disease.451 Clinical trials of monoclonal and YAP.491 Furthermore, norepinephrine-mediated, β-adrenergic/PKA
antibodies that target integrin receptors are underway in several cancer activation of SRC has recently been shown to enhance tumor cell
types. For example, etaracizumab, a humanized monoclonal antibody migration, invasion, and growth.492 The tyrosine kinase inhibitor
targeting αvβ3 integrin, is being tested in solid tumors and has shown dasatinib, which is used in the treatment of CML and Philadelphia
activity in patients with metastatic melanoma.464 Cilengitide, an chromosome–positive acute lymphoblastic leukemia (ALL), inhibits
inhibitor of the αvβ3 and αvβ5 integrins, inhibited angiogenesis and SRC family kinases, in addition to BCR-ABL, KIT, Ephrin A2 receptor,
tumor cell proliferation in preclinical studies; however, a phase III and PDGFR (see Table 2.1).493–495 Additional dual SRC/ABL and SRC
trial in combination with temozolomide and radiation in patients selective inhibitors are approved in CML (bosutinib, ponatinib; see
with glioblastoma did not result in improved outcomes compared later section on ABL signaling) or are in clinical testing, including
with chemoradiation alone.456,465 saracatinib, XL-228, KX2-391, and DCC2036. Most have shown
limited single-agent activity and are being developed as combination
NON–RECEPTOR TYROSINE therapies.469,478,484–486
KINASE SIGNALING
ABL Signaling
SRC Signaling
The ABL gene was first identified in 1980 as the oncogene responsible
The SRC family of intracellular, non-RTK proteins is composed of for driving the Abelson murine leukemia virus, and later was found
11 members (SRC, FYN, YES, Blk, Yrk, Frk/Rak, Fgr, Hck, Lck, to be part of the translocations found in many types of human leu-
Srm, and Lyn).466 They share common structural features including kemias.496 ABL1 and ABL2 isoforms have both overlapping and
the so-called SRC-homology (SH) domains 1 to 4. The SH1 domain divergent functions. ABL is found in an autoinhibited conformation
includes the kinase domain. Only SRC, FYN, and YES are expressed dictated by clamping of the N-terminal hydrophobic region, SH3
Intracellular Signaling  •  CHAPTER 2 41

and SH2 domains onto the C-lobe of the kinase domain.497 Inter- neurofibromin (NF1), GAP1IP4BP, and CAPRI, negatively regulate
molecular interactions with adaptors such as RIN1 or phosphorylation RAS activity and thus function as tumor suppressors.514,518
by SRC, among others, promote stabilization of the open, active form Binding of GTP to RAS induces conformational changes in the
of ABL 23842626. The ABL tyrosine kinase is found in both the switch I (loop 2 residues 30–38) and switch II (helix 2 and loop 4
cytoplasm and the nucleus, and its function varies based on subcellular residues 60–76) domains, which facilitate the association of RAS with
localization.498 Cytoplasmic ABL has been implicated in G1/S check- regulators and downstream effectors.531,532 RAS directly interacts with
point regulation and interaction with actin on C-terminal binding over 20 effector proteins, of which the RAF kinases, PI3 kinases, and
sites,499 whereas nuclear ABL inhibits binding of the DNA repair RALGDS are the best characterized (see Fig. 2.1). The canonic RAS/
protein Rad51 to sites of DNA damage.500,501 Activated ABL kinases RAF/MEK/ERK (classic MAP kinase) cascade is by far the most
are now recognized to play an important role in tumor initiation by extensively characterized RAS effector pathway. This prototype of a
disrupting cell polarity and by promoting invasion and metastasis by three-tiered kinase signaling cascade exemplifies numerous RAS-
regulating invadopodia.502 dependent mitogen-activated protein kinase (MAPK) cascades that
In cancer, translocation of the ABL gene on chromosome 9 with respond to diverse signals, including cell stress and cytokine signaling
the breakpoint cluster region (BCR) gene on chromosome 22 results (see section on cytokines for details of the JNK and p38 pathways).533
in the expression of a BCR-ABL fusion protein.503 This translocation, The RAF protein family (which represent the top-tier MAPK kinase
the Philadelphia chromosome, is found in almost all CMLs and kinases [MAPKKKs], or MEK kinases [MEKKs]) is composed of
represents the pathognomonic molecular lesion in this disease. three differentially expressed isoforms, A-RAF, B-RAF, and C-RAF
BCR-ABL translocations are also found in approximately one-third (RAF-1).534,535 RAF, via its RAS binding domain (RBD) and cysteine
of acute lymphoblastic leukemias (ALLs).504,505 In addition, at least rich domain (CRD), interacts with GTP-bound Ras.536–539 Binding
eight other fusion partners of ABL have been discovered.502 The of RAF to GTP-bound RAS results in RAF localization to the plasma
realization that the proliferation and survival of CML cells is critically membrane and its subsequent phosphorylation and activation.540 The
dependent on the ABL fusion protein led to the development of mechanisms of RAF-1 and B-RAF phosphorylation and activation
imatinib, an inhibitor of the ABL tyrosine kinase.505 To date, there are distinct and are derived from the summation of signaling inputs
are five FDA-approved inhibitors for CML: imatinib, dasatinib, from small G proteins (RAS, RAC, CDC42, RAP-1), kinases (activating
bosutinib, nilotinib, and ponatinib (see Table 2.1).506 Moreover, imatinib inputs: SRC/PAK/PKC, inhibitory inputs: PKA/AKT/SGK), isoform
and dasatinib are approved for treatment of ALL.507–511 A secondary homodimerization and heterodimerization, phosphatases (PP2, PP2A),
mutation in the gatekeeper site T315I is the most common reason scaffolding proteins (KSR, RKIP, HSP90, and so on) and cofactors
for acquired resistance to ABL kinase inhibitors. Combination of (14-3-3), with phosphorylation events regulating critical aspects of
ATP-competitive and allosteric inhibitors such as GNF-5 may be a RAF activation.535,541 In brief, RAS binding to RAF-1 releases 14-3-3,
novel strategy to combat resistance.512 a negative regulator that binds to basally phosphorylated residues
S259 and S261 and sequesters RAF-1 in the cytosol in a closed,
RAS/MAP KINASE PATHWAY SIGNALING inactive conformation. Liberation from 14-3-3 exposes the PKA/
AKT/SGK-dependent inhibitory phosphorylation on S259, facilitating
First identified over 30 years ago as the oncogenes responsible for the its dephosphorylation by protein phosphatase 2A.542–546 Loss of this
transforming potential of the Harvey and Kirsten murine sarcoma inhibitory phosphorylation primes RAF-1 for RAS, PAK, SRC, growth
retroviruses (Ha-MSV and Ki-MSV), RAS proteins are guanine factor, and integrin-stimulated activating phosphorylations on S338,
nucleotide binding proteins.513–517 Ras proteins have intrinsic GTPase Y341, T491, and S494.547–551 A-RAF is activated similarly to RAF-1.
activity and cycle between inactive GDP- and active GTP-bound Notably, B-RAF activation requires fewer steps owing to its constitutive
states. GDP/GTP exchange thus allows Ras proteins to function as phosphorylation at S445, a site analogous to S338 in RAF-1.541,552
binary molecular switches. Binding to RAS-GTP stimulates B-RAF phosphorylation at critical
In the human genome, there are three RAS genes, which encode residues in its activation loop, T599 and S602 (analogous to T491
four homologous proteins (HRas, NRas, and the alternative splice and S494 in RAF-1).553,554 The B-RAF isoform is the most potent
variants KRAS4A and KRAS4B) with highly conserved N-terminal activator of ERK pathway output.535,552,554
and variable C-terminal regions. Following stimulation of cells by Activated Raf proteins bind and phosphorylate MEK1 and MEK2
serum growth factors, cytokines, hormones, and neurotransmitters, (mitogen-activated protein kinase/extracellular signal-regulated kinase
Ras undergoes a series of C-terminal (C186AAX) posttranslational kinases 1 and 2, or MAPKK, or MAP2K) on serines 217 and 221.555
modifications that result in its localization to specific membrane Activated MEK, a dual-specificity threonine/tyrosine kinase, in turn
microdomains.518 Membrane localization is required for the transforming phosphorylates MAPK/ERK-1/2 (mitogen-activated protein kinases/
properties of Ras, because mutation of Ras at C186 results in cytosolic extracellular signal-regulated kinases 1 and 2) on threonine 183 and
localization and protein inactivation whereas Ras activity can be rescued tyrosine 185, inducing a conformational change, activation, and disso-
by myristoylation, which promotes membrane localization.519–521 GEFs ciation from MEK.556 Activated ERK then phosphorylates substrates in
promote Ras activation by binding to GDP-bound Ras and facilitating the cytosol (e.g., p90RSK) and in the nucleus (such as the transcription
the release of GDP and the binding of GTP. SOS1, RAS-GRF (dual factors ELK-1, ETS-2, FOS, JUN, ATF-2, AP-1, MYC, CREB1), which
specificity GEFs for RAS and RAC), and RAS-GRP (stimulated by promote proliferation and survival.557,558 Raf-1 has also been shown
DAG/phorbol esters and Ca+) are the mostly highly characterized to mediate suppression of apoptosis through non–MEK-dependent
RAS-GEFs.522–524 Using RTK stimulation as an example, ligand binding interactions with ASK1, MST2, and MEKK1/NF-κB.559
induces RTK dimerization and autophosphorylation of tyrosine residues RAS/RAF signaling triggers numerous regulatory mechanisms,
in the receptor cytoplasmic tail. Phosphotyrosine docking sites recruit including classic negative feedback loops, which serve to attenuate
scaffold proteins such as SHC and permit interaction with the SH2 pathway output.560 The Sprouty and Sprouty-related EVH1-domain-
domains of adaptor proteins such as Grb2.525 Grb2 in turn recruits containing protein (Spred) proteins (encoded by the SPRY1–4 and
SOS1 via its SRC homology 3 (SH3) domain, thereby positioning SPRED1–4 genes, respectively) inhibit the cascade at the level of RAS
SOS near membrane-anchored RAS (see Fig. 2.1).525–529 SOS1 in turn and RAF activation.561–564 The dual-specificity phosphatases (MKPs/
activates RAS via its CDC25 homology (RASGEF) domain and DUSPs) dephosphorylate MAP kinases, including ERK.565,566 In
N-terminal RAS exchanger motif (REM or RASGEFN domain). addition, increased pathway activity induces ERK-dependent negative
Ras inactivation is catalyzed by GTPase-activating proteins phosphorylations on B-RAF (at S151, S750, T401 and T753) and
(GAPs), which enhance the intrinsic GTPase activity of Ras proteins on RAF-1 (at S29, S43, S289, S296, S301 and S642) that abrogate
by 100,000-fold.530 RAS GAPs, which include p120-RASGAP, interactions with RAS and homodimer and heterodimer formation.567,568
42 Part I: Science and Clinical Oncology

RAS signaling is further regulated by RKIP (RAF kinase-inhibitory emerged as a mechanism of acquired resistance to RAF inhibition.594,595
protein), which disrupts the RAF-1/MEK interaction and IMP (impedes Hot spots include mutations at MEK1 residues F53, K57, and P124
mitogenic-signal propagation), which represses KSR-dependent scaf- and at MEK2 residue F57, which is paralogous to MEK1 F53. ERK
folding of RAF/MEK, among other functions.534,569–571 amplification and mutation, although equally rare (approximately 1%
In its active, GTP-bound state, RAS alternatively binds to the of all tumors), may also emerge as mechanisms of resistance to upstream
p110α catalytic subunit of class I PI3 kinases,572,573 causing activation MAPK inhibition, including alteration at the hot spot residue E322.596
of its lipid kinase activity and thereby generating PIP3, which in turn Given the high prevalence of RAS pathway alterations in human
stimulates the proproliferative and prosurvival kinases PDK1 and cancers, significant attention has been directed toward the development
AKT (see section on PI3 kinase signaling for details). RAS-dependent of selective inhibitors of this pathway.560,580,597,598 To date, clinically
activation of PI3 kinase can further stimulate RAC, a RHO family effective direct inhibitors of oncogenic RAS have yet to be identified.
GTPase involved in regulation of actin and NF-κB.513,574,575 A third The inability to directly target RAS has been attributed to the high
major class of RAS effectors is the group of GEFs for RALA and RALB, affinity of the RAS-GTP interaction. Extensive efforts were thus directed
namely RALGDS, RGL, and RGL2.576–578 These RAL exchange factors toward inhibiting the posttranslation modifications required for RAS
stimulate phospholipase D and the CDC42/RAC-GAP RAL binding activation. Specifically, inhibitors of the enzyme farnesyltransferase,
protein 1 (RALBP1) and inhibit Forkhead transcription factors to which regulates RAS localization, were tested in randomized phase III
regulate transcription, vesicular trafficking, and cell cycle progression. trials but were found to be inactive.599 The failure of farnesyltransferase
Several additional RAS effectors have been identified. These include inhibitors in KRAS-mutant tumors was predicted by the preclinical
(1) PLCε, which generates IP3 and DAG and regulates calcium release observation that geranylgeranyl modification can substitute for farne-
and PKC activation; (2) T-cell lymphoma invasion and metastasis-1 sylation in targeting KRAS and NRAS to the plasma membrane.600,601
(TIAM1), which facilitates actin reorganization via RAC; (3) AF6, Small molecules that irreversibly bind to the mutant cysteine in tumors
which contributes to cytoskeletal changes; (4) RIN1, which regulates with G12C K-RAS have been developed.602 Because this class of
endocytosis; and (5) RASSF and NORE1, which have been shown compounds is selective for the G12C mutant, they are a promising
to regulate apoptosis and cell cycle progression.513,518 Many other novel approach for a subset of patients with KRAS-mutant tumors.
nondirect connections allow RAS to affect the cellular microenviron- As an alternative, extensive efforts have focused on the development
ment, metabolic signaling, autophagy, inflammation, and immune of selective inhibitors of key kinase effectors of RAS transformation.
responses. Overall, the complex effects of RAS activation result in a The selective RAF inhibitors vemurafenib and dabrafenib induce tumor
diversity of context-dependent phenotypes ranging from cell prolifera- regressions in most patients with BRAF V600E mutant melanoma
tion to cell death that are influenced by a wide variety of extracellular and are FDA approved for this indication (see Table 2.1).603–612 Notably,
stimuli and intricately woven layers of regulation. these agents selectively inhibit RAF signaling in BRAF-mutant tumors
The potent oncogenic effects of RAS signaling are highlighted by and are thus inactive in RAS-mutant tumors.613 Several mechanisms
the high prevalence of mutations in the RAS genes and its proximal of resistance to RAF inhibitors have emerged, including BRAF V600E
downstream effectors.518,579–581 RAS mutations are found in approxi- splice variants or amplification; loss or mutation of NF1; parallel
mately 30% of all human tumors, the majority of which (85%) occur pathway activation of RTKs; mutation of PI3K/AKT components;
in the KRAS isoform. KRAS is frequently mutated in pancreatic cancers mutations in NRAS, RAF1, and MEK1/2; and amplification of
(58%), colorectal and biliary tree cancers (33 and 31%), and non–small MITF.592,614 Sorafenib, a multikinase inhibitor of RAF, VEGFR2, and
cell lung adenocarcinomas (17%). Mutations in HRAS are most PDGFRβ, has been shown to have some clinical efficacy in ARAF-
common in low-grade bladder cancers (11%), whereas mutation in mutant histiocytosis and NSCLC.588,589,615
NRAS is a frequent event in melanoma (18%) and biliary tree cancers The selective MEK inhibitor trametinib is also FDA approved
(11%). Mutations that lock RAS in its GTP-bound state confer for use in melanomas with BRAF mutation (see Table 2.1).605,616–619
oncogenic potential. Point (missense) mutations in residues 12, 13, Furthermore, the combination of a RAF and a MEK inhibitor has
61 (exons 2 and 3) generate a constitutively active RAS oncoprotein been shown to have greater activity than Raf inhibitor monotherapy
by abrogating intrinsic GTPase activity and inhibiting GAP binding.582 in both preclinical models and in patients with melanoma, including
Other mutations, including those at residues 117 and 146, contribute the combinations of vemurafenib plus cobimetinib620 and dabrafenib
to RAS activation by increasing GDP-to-GTP exchange.583,584 Heritable plus trametinib.605,619,621,622 MEK inhibition is also emerging as a
germline mutations of several RAS/MAPK pathway components have strategy to target NRAS-mutant melanoma,623 colorectal624 and thyroid
been shown to be the underlying cause of the so-called RASopathies cancers,625 NF1-mutant melanoma,587 GBM,626 and neuroblastoma.627
neurofibromatosis type 1 (NF1); Noonan, Costello, and cardiofacio- MEK inhibition in combination with immunotherapy, chemotherapy,
cutaneous syndromes; and other developmental disorders.579 More radiation, and PI3K and CDK4/6 inhibitors is being investigated
recently, deep sequencing efforts have identified recurrent somatic in KRAS-mutant tumors including colorectal and lung cancer (see
mutations in the RAS-GAP NF1 in glioblastoma585 and melanoma,586,587 Table 2.1).624,628 Furthermore, clinical evidence of activity of the MEK
as well as in A-RAF (hot spot at S214) in histiocytosis and lung inhibitors cobimetinib, selumetinib, and trametinib has been revealed
cancer588–590 and in RAF-1 (hot spot S257).588 Mutations in BRAF in cases and preclinical reports of MEK1-mutant histiocytosis,589
are also common in human cancer and typically occur in a mutually low-grade serous ovarian cancer,629 NSCLC,594 and melanoma.630
exclusive pattern with RAS mutations. BRAF mutations have been Alternative strategies for inhibiting MAP kinase signaling include
identified in approximately 8% of all cancers, most notably in melanoma HSP90 inhibitors that induce the degradation of RAF1 and mutant
(50%–60%) and in papillary thyroid (30%–50%), biliary tree (14%), BRAF.631 Drug development of kinase- and dimerization-targeted ERK
colorectal (10%), ovarian (12%), and lung cancers (3%) and hairy inhibitors (SCH772984, BVD-523, DEL-22379) is also underway and
cell leukemia (100%).534,535,559,579,591,592 A single valine to glutamic acid may provide a downstream alternative when upstream RAF or MEK
substitution at residue 600 (V600E) accounts for more than 80% of inhibitors fail.632–635 Combinatorial approaches are also being actively
all BRAF mutations and renders BRAF an active monomer in settings pursued, given the modest activity of MEK inhibitors in patients
of low RAS activity, that is sensitive to RAF inhibition. Other non- with RAS-mutant tumors and the frequent co-occurrence of RAS and
V600E BRAF mutants have been recently identified and characterized PI3 kinase pathway alterations in multiple cancer types.636–638 Given
to function as RAS-independent dimers that are insensitive to current the recent success of immunotherapy in many cancers, preclinical
RAF inhibitors, such as vemurafenib, that only effectively inhibit evidence supports the triple combination of BRAF and MEK inhibitors
mutant monomers.593 Activating mutations in MEK1 (MAP2K1) and with immunotherapy in BRAF V600E mutant melanoma, despite
MEK2 (MAP2K2) are also present in approximately 1% of human substantial liver toxicities described with initial trials with combined
tumors, more prominently in melanoma and lung cancer, and have treatment of vemurafenib and ipilimumab.639 In addition, elevated
Intracellular Signaling  •  CHAPTER 2 43

antitumor immune responses in mouse models of triple negative AKT-mediated antiapoptotic effects occur through phosphorylation
breast cancer following combined MEK inhibition and immuno- and inhibition of Bad, which negatively regulates the antiapoptotic
therapy suggest that hyperactivate MEK signaling may contribute to protein Bcl-xL; caspase-9, a proapoptotic protease; and the FOXO1
immune evasion.640 transcription factor which regulates the expression of proapoptotic
Given the prominent role of activated ERK in initiating the genes.660–662 AKT also controls cell survival by upregulating NF-κB
transcription and translation of cell cycle components such as cyclin activity through phosphorylation and activation of Iκb kinase, which
D1, inhibitors of cell cycle kinase components have emerged as marks Iκb, an inhibitor of NF-κB, for degradation.663,664 Once released
an alternative strategy to abrogate the output of MAPK signaling. from IκB, NF-κB translocates into the nucleus, where it regulates a
CDK4, a critical serine/threonine kinase that functions in an active multitude of genes involved in cell survival. AKT also phosphorylates
complex with cyclin D to phosphorylate RB and stimulate E2F and activates Mdm2, an E3 ubiquitin ligase that binds to the pro-
release and S phase entry, is amplified in liposarcoma. Palbociclib, apoptotic tumor suppressor p53 and directs it for proteasomal
an inhibitor of CDK4, is FDA approved for use in postmenopausal degradation.665
women with ER+, HER2− metastatic breast cancer; a second drug, AKT-independent effectors of PI3 kinase activation have also been
abemaciclib, has also show efficacy in this same patient population identified and likely play important roles in the development and
(see Table 2.1).641–644 progression of some cancers. These include CDC42 and Rac1, which
are involved in motility and reorganization of the cytoskeleton, and
PI3 KINASE/AKT/MTOR PATHWAY SIGNALING the serum glucocorticoid kinase (SGK) family of serine/threonine
kinases, which promote cell survival.666,667
The PI3 kinase/AKT/mTOR pathway is a key regulator of growth Many of the canonic functions of AKT with regard to cell growth
factor–mediated proliferation and survival.645 Several extracellular and proliferation are mediated through the mTOR pathway. mTOR
growth factors stimulate PI3 kinase by binding to their cognate RTKs is a serine/threonine kinase and a member of the phosphatidylino-
or GPCRs.646 Activated PI3 kinase phosphorylates the 3-OH group sitol kinase-related kinase family.668 mTOR is a component of two
of the inositol ring of phosphatidylinositol, catalyzing the conversion complexes, the rapamycin-sensitive mTORC1 and the rapamycin-
of PIP2 to PIP3. PIP3 then binds to the pleckstrin homology domain insensitive mTORC2 complexes.669 Within mTORC1, mTOR
(PH domain) of multiple proteins, facilitating their recruitment to the associates with Raptor (regulatory associated protein of mTOR) and
plasma membrane and thus regulating their function (see Fig. 2.2). mLST8, whereas the mTORC2 complex consists of mTOR, Rictor
PI3 kinases are grouped into class I, II, and III kinases based on (rapamycin insensitive companion of mTOR), SIN1, and mLST8.670
their structure and substrate preferences, although class II and III In addition to its role in activating AKT as described earlier, mTORC2
kinases have been much less studied. Class I PI3 kinases are subdivided also controls cytoskeletal changes through regulation of paxillin, Rho,
into two groups, IA and IB. Class IA comprises the PIK3CA, PIK3CB, Rac, and PKCα.671
and PIK3CD genes, which encode the catalytic p110α, p110β, and mTORC1 activation is regulated in part by AKT phosphorylation
p110δ subunits, respectively. These p110 components heterodimerize of TSC2. TSC2 forms a heterodimeric complex with TSC1 that acts
with a regulatory subunit (p85), of which there are five isoforms and as a GTPase-activating protein (GAP) for the small GTPase Rheb,
splice variants encoded by the PIK3R1-3 genes. Together, the p110/ causing accumulation of inactive Rheb-GDP.672,673 TSC2 phosphoryla-
p85 complex functions primarily in the generation of PIP3.647,648 The tion results in suppression of this GAP activity and subsequent activation
PIK3CG gene encodes the class IB p110γ isoform, which couples and accumulation of Rheb-GTP, which then activates mTORC1.
with p101 (PIK3R5) or p87 (PIK3R6) regulatory subunits. Notably, mTORC1 serves as a central nexus for integration of multiple extracel-
p110δ/γ catalytic subunits have specialized expression patterns mainly lular signals, including oxygen and amino acid levels, growth factors,
in leukocytes, whereas p110α/β are ubiquitous.649 Analogous to the and stress. Based on these input signals, mTORC1 activity influences
activation of the Ras pathway detailed previously, the p85 regulatory cellular growth, metabolism, protein synthesis, and cell cycle progres-
subunit of PI3 kinase associates with phosphorylated tyrosine residues sion. One such example is the energy sensing mechanism of the cell
located on the intracellular domains of RTKs through an SH2 domain, comprised of LKB1 and AMPK.674 Increasing levels of AMP, a marker
resulting in allosteric activation of the p110 catalytic subunit. PI3 of decreased nutrient levels, results in AMPK phosphorylation and
kinases can also be activated indirectly through the adaptor protein activation by LKB1. AMPK phosphorylates TSC2, which, as described
Grb2 following its association with the scaffolding protein GAB1. earlier, inhibits mTORC1 activity, leading to downregulation of protein
PI3 kinase pathway activity is negatively regulated by the tumor synthesis and cell growth in response to low nutrient levels.675 In part,
suppressor PTEN (phosphatase and tensin homolog), which is a dual mTORC1 activation regulates these phenotypes via phosphorylation
lipid and protein phosphatase that dephosphorylates PIP3, converting and activation of p70S6 kinase and inhibition of 4EBP1. The former
it back to PIP2.650,651 protein stimulates mRNA translation, whereas the latter inhibits
The best characterized effectors of PI3 kinase are the three members translation of mRNA transcripts with a 5′ cap.676
of the serine/threonine kinase AKT family (AKT1, AKT2, and AKT3). Both mTORC1 and S6 kinase also participate in a negative feedback
Both AKT and phosphoinositide-dependent kinase 1 (PDK1) are loop in which both proteins activate insulin receptor substrate 1 (IRS1),
recruited by PIP3 to the plasma membrane via their PH domain, which results in inhibition of insulin-mediated PI3 kinase activation.677
where AKT is phosphorylated at Thr308 by PDK1, resulting in its AKT can also activate mTORC1 in a TSC2-independent manner via
activation.652–654 Phosphorylation at a second residue, Ser473, by mTOR phosphorylation of PRAS40, a protein that interacts with mTORC1.
complex 2 (mTORC2) further enhances AKT activity.655 Activated The mechanisms responsible for PI3 kinase pathway activation in
AKT promotes cellular proliferation, survival, and other phenotypes cancer are diverse and include activating mutations and amplification
through activation of multiple downstream effectors. Proliferative of PIK3CA, AKT1, AKT2, and AKT3, and mTOR; deletion or loss
effects are regulated through phosphorylation and inhibition of GSK3β, of PTEN expression or function; mutation in PIK3R1; loss of TSC1
which phosphorylates cyclin D1 and marks it for degradation; FOXO4, or TSC2 function; RAS mutation; and dysregulated growth factor
a transcription factor that regulates the expression of the CDK inhibitor receptor and integrin activation, as outlined later.645,648,678 In human
p27; and p21, a second CDK inhibitor that, on phosphorylation, tumors, activation of PI3 kinase is frequently a direct consequence
translocates from the nucleus to the cytoplasm, where it regulates cell of dysregulated RTK signaling secondary to mutation, amplification,
survival.656–659 In sum, these actions promote the expression of cyclin or ligand overexpression. For example, ERBB2 amplification in breast
D1, which drives progression of cells through the cell cycle and the and gastric cancer results in AKT activation.27,678 Similarly, kinase
downregulation of cyclin-dependent kinase inhibitors that inhibit cell domain mutations of EGFR induce constitutive AKT activation in
cycle progression. lung cancers and glioblastomas, and AKT activity in these tumors
44 Part I: Science and Clinical Oncology

is critical for EGFR-mediating transformation.637,679 Oncogenic Temsirolimus and everolimus, analogues of rapamycin that inhibit
Ras mutations also activate PI3 kinase, and PI3 kinase activation is the mTORC1 complex, are FDA approved for use in patients with
required for Ras-mediated tumorigenesis in genetically engineered renal cell carcinomas, and everolimus is FDA approved for the treatment
mouse models.572,680–682 of patients with pancreatic neuroendocrine tumors (see Table 2.1).705–708
Mutations in the PIK3CA gene, which encodes the p110α catalytic Everolimus also has significant clinical activity in patients with sub-
subunit, are frequently observed in tumors of the colon, breast, brain, ependymal giant cell astrocytomas that arise in the setting of tuberous
stomach, and ovary and other cancers.648,678,683 The most frequent are sclerosis, an inherited cancer-predisposition syndrome resulting from
E542K and E545K, located in the helical domain (exon 9), and germline mutations in the TSC1 and TSC2 genes.709,710 In metastatic
H1047R (exon 20), located in the kinase domain.684 All three mutants renal cell carcinoma, patients that benefited from treatment with
demonstrate increased lipid kinase activity, induce phosphorylation everolimus more commonly harbored TSC1/2 or mTOR mutations
of AKT and its downstream effectors, and can transform chicken (see Table 2.1).711
embryo fibroblasts.685 Exon 9 helical domain mutations block the A complete response to everolimus has been reported in a patient
inhibitory interaction between the p85 regulatory subunit and the with metastatic bladder cancer that harbored loss-of-function mutations
p110 subunit and result in constitutive kinase activation.686 Exon 20 in TSC1 and NF2, suggesting that such agents can induce significant
catalytic domain mutations result in constitutive kinase activation.678 antitumor responses in genetically selected patients.712 Additional
PIK3CA mutations commonly co-occur with KRAS mutations, and responses to everolimus were observed in TSC1-mutant tumors but
ERBB2 amplification in colon and breast cancers, respectively, and not to the degree of the complete responder, indicating that coaltered
expression of mutant PI3 kinase in breast cell lines as well as fibroblasts genes likely confer resistance to mTOR inhibitory therapy even in
causes neoplastic transformation.687 Unlike wild-type p110α which the setting of a canonic predictive biomarker of response. In sum,
lacks oncogenic potential, wild-type overexpression of the other three mTOR inhibitors are likely most effective when used in genetically
isoforms can induce transformation of cultured cells.688 PIK3R1 encodes defined patient populations, with the majority of patients requiring
the p85 regulatory subunit of PI3 kinase. Alterations in within this combination therapies because of the presence of coaltered genes. As
gene have also been reported in multiple cancers, including glioblas- an example of the latter, everolimus in combination with exemestane
tomas and colorectal, endometrial, and ovarian cancers.585,689 A recurrent was FDA approved for the treatment of advanced hormone receptor–
PIK3R1 mutation, PIK3R1(R348∗), has been shown to have neo- positive, HER2− breast cancer.713
morphic functions resulting in activation of MEK and JNK and Resistance to mTOR inhibition is thought to derive from multiple
sensitivity to inhibitors of these effector kinases.690 Furthermore, partial mechanisms, including activation of parallel signaling networks such
loss of the PIK3R1 gene product p85α was demonstrated to increase as the MAP kinase pathway.714 Furthermore, inhibiting mTORC1
the proportion of p110α/p85 heterodimers bound to active receptors, can preferentially lead to an increase in mTORC2 signaling, and, as
indicating that targeting p110α-selective inhibitors may be effective described earlier, mTORC2 enhances AKT activation via phosphoryla-
in the setting of PIK3R1 loss.691 tion of the serine 473 residue.715 As an alternative approach, potent
Loss of PTEN function is the most frequently observed PI3 kinase/ mTOR kinase inhibitors are being developed, including RapaLink-1,
AKT pathway alteration in human malignancies and is common in and have shown significant preclinical activity in cell lines resistant
tumors of the prostate, breast, ovary, lung, colon, and bladder as well to rapamycin and in in vivo models of glioblastoma.716,717
as melanomas and glioblastomas.678 Loss of PTEN function in tumors
is mediated by a diversity of mechanisms including mutation, deletion, TRANSLATIONAL IMPLICATIONS
posttranslational modification, and promoter hypermethylation.678
Dysregulated expression of microRNAs that target the 3′-untranslated As outlined earlier, mutational and epigenetic alterations induce
region of PTEN has also been shown to induce cell survival and constitutive activation of a broad array of signaling pathways in human
cisplatin resistance.692 tumors. In some instances, the growth and survival of tumor cells
As AKT activation enhances proliferation and suppresses apoptosis, have been shown to be dependent on a single signaling pathway
its activation would be predicted to have strong oncogenic func- activated by a mutated oncogene or tumor suppressor, a phenomenon
tion. Indeed, AKT was initially identified as a proto-oncogene in referred to as oncogene addiction.718 In such instances, targeted inhibitors
the mouse leukemia virus AKT8.693 A recurring hot spot mutation of such pathways have demonstrated unprecedented clinical activity
in the PH-domain of AKT1 (E17K) occurs with low frequency in in molecularly defined subsets of patients (see Table 2.1). Examples
breast, colorectal, bladder, endometrial, and ovarian cancers.694–696 include imatinib in patients with CML, erlotinib in patients with
This mutation results in constitutive localization to the plasma EGFR-mutant NSCLC, and vemurafenib in patients with BRAF-mutant
membrane without the need for PIP3 recruitment.684 Amplification melanoma.30,505,511,603,612,719 Despite these dramatic successes, the majority
of the AKT2 gene has been reported in ovarian and pancreatic cancers, of cancer patients have yet to benefit from this approach. Potential
and gain-of-function AKT3 mutations have been reported to occur in explanations for this lack of benefit include the redundant regulation
melanoma.697 of key downstream mediators of transformation by multiple signaling
Given the significant proportion of malignancies that harbor muta- pathways, the lack of specificity of the drug for the driver alteration,
tions in the PI3 kinase/AKT/mTOR pathway, a concerted effort is and intrinsic and acquired drug resistance due to second site mutations
ongoing to identify selective inhibitors of various PI3 kinase pathway in the target gene or by other mechanisms. Progress in this field has
components. These agents can be categorized as pan-selective or selec- also been delayed by the continued practice of performing clinical
tive PI3 kinase inhibitors, dual PI3 kinase/mTOR kinase inhibitors, trials of targeted inhibitors in unselected patient populations.
AKT inhibitors, and mTOR inhibitors. Both isoform-specific and Recent advances in sequencing methodology have made it feasible
pan-selective inhibitors of PI3 kinase are being explored in several to now prospectively sequence all patients with advanced cancer with
clinical trials across cancers. Most notably, the PI3Kδ inhibitor idelalisib the goal of identifying potentially “actionable” genomic alterations.720
is FDA approved for non-Hodgkin lymphoma and certain types of Such prospective sequencing efforts have highlighted several challenges
leukemia.698,699 Promising clinical responses have also been reported that have slowed the application of targeted inhibitors in cancer patients.
with α-selective PI3 kinase inhibitors in patients with several cancer As one example, most mutations, even in well-characterized cancer
types, most notably breast cancer (see Table 2.1).700–703 Although clinical genes, are likely inert passenger mutations. Furthermore, not all
activity with AKT inhibitors has been modest in patients, a pan-cancer activating mutations respond similarly to targeted inhibitors. For
basket trial of the ATP-competitive pan-AKT kinase inhibitor AZD5363 example, exon 19 EGFR deletions are sensitive to the EGFR kinase
demonstrated significant clinical responses in patients with AKT1 E17K inhibitor erlotinib, whereas exon 20 insertions are intrinsically
mutant tumors.704 resistant.721,722 Given the large number of somatic mutations present
Intracellular Signaling  •  CHAPTER 2 45

in each human tumor, and the variable biologic and clinical significance respond to vemurafenib, whereas colorectal tumors with the same
of individual mutant alleles, there is an urgent need for clinical support mutation are intrinsically resistant. Resistance in the latter results from
tools that will aid clinicians in interpreting molecular tumor profiling rapid adaptation of the cancer cell resulting in activation of EGFR.724
and guiding treatment selection. This phenomenon of adaptive resistance, defined as a rapid reactivation
A second challenge is that many oncogenic alterations are rare. To of parallel signaling pathways after relief of negative feedback signals,
address this challenge, novel clinical trial designs have been formulated likely abrogates the effects of selective pathway inhibitors in many
to test the efficacy of targeted inhibitors in patients with defined cancer types.592,725,726 Because significant drug development efforts are
molecular events independent of the primary site of disease. Eligibility currently focused on the development of targeted inhibitors of
for these “basket” studies is based on the presence of a particular oncogene-activated signaling pathways, a detailed understanding of
molecular alteration (e.g., BRAF V600E, AKT1 E17K, or an NTRK normal physiologic pathways and their dysregulation in cancer will
fusion) rather than site of tumor origin.268,273,704,723 Although promising be critical for the optimal development and clinical application of
results from such trials highlight the importance of tumor mutational targeted kinase inhibitors.
profile in dictating drug sensitivity, lineage-specific differences also
play a role in determining clinical response to inhibitors of activated The complete reference list is available online at
signaling pathways. As an example, most BRAF V600E melanomas ExpertConsult.com.

KEY REFERENCES
1. Chakravarty D, et al. OncoKB: A Precision Oncol- 196. Singh D, et al. Transforming fusions of FGFR 442. Toy W, et al. Activating ESR1 Mutations differen-
ogy Knowledge Base. JCO Precis Oncol. 2017; and TACC genes in human glioblastoma. Science. tially affect the efficacy of ER antagonists. Cancer
1–16. 2012;337:1231–1235. Discov. 2017;7:277–287.
21. Yarden Y, Pines G. The ERBB network: at last, 210. Jordan EJ, et al. Prospective comprehensive molecular 445. Scher HI, et al. Increased survival with enzalutamide
cancer therapy meets systems biology. Nat Rev characterization of lung adenocarcinomas for effi- in prostate cancer after chemotherapy. N Engl J Med.
Cancer. 2012;12:553–563. cient patient matching to approved and emerging 2012;367:1187–1197.
34. Brennan CW, et al. The somatic genomic landscape therapies. Cancer Discov. 2017;7:596–609. 465. Stupp R, et al. Cilengitide combined with stan-
of glioblastoma. Cell. 2013;155:462–477. 217. Goel HL, Mercurio AM. VEGF targets the tumour dard treatment for patients with newly diagnosed
38. Nathanson DA, et al. Targeted therapy resistance cell. Nat Rev Cancer. 2013;13:871–882. glioblastoma with methylated MGMT promoter
mediated by dynamic regulation of extrachromo- 249. Frampton GM, et al. Activation of MET via diverse (CENTRIC EORTC 26071-22072 study): a
somal mutant EGFR DNA. Science. 2014;343: exon 14 splicing alterations occurs in multiple tumor multicentre, randomised, open-label, phase 3 trial.
72–76. types and confers clinical sensitivity to MET inhibi- Lancet Oncol. 2014;15:1100–1108.
43. Cristescu R, et al. Molecular analysis of gastric cancer tors. Cancer Discov. 2015;5:850–859. 492. Armaiz-Pena GN, et al. Src activation by beta-
identifies subtypes associated with distinct clinical 252. Paik PK, et al. Response to MET inhibitors in adrenoreceptors is a key switch for tumour metastasis.
outcomes. Nat Med. 2015;21:449–456. patients with stage IV lung adenocarcinomas Nat Commun. 2013;4:1403.
44. Cancer Genome Atlas Research, N. Comprehensive harboring MET mutations causing exon 14 skipping. 506. Hantschel O, Grebien F, Superti-Furga G. The
molecular characterization of urothelial bladder Cancer Discov. 2015;5:842–849. growing arsenal of ATP-competitive and allosteric
carcinoma. Nature. 2014;507:315–322. 266. Stransky N, Cerami E, Schalm S, Kim JL, Lengauer inhibitors of BCR-ABL. Cancer Res. 2012;72:
46. Bose R, et al. Activating HER2 mutations in HER2 C. The landscape of kinase fusions in cancer. Nat 4890–4895.
gene amplification negative breast cancer. Cancer Commun. 2014;5:4846. 586. Cancer Genome Atlas, N. Genomic classification
Discov. 2013;3:224–237. 268. Drilon A, et al. A Next-generation TRK kinase of cutaneous melanoma. Cell. 2015;161:1681–
50. Tebbutt N, Pedersen MW, Johns TG. Targeting the inhibitor overcomes acquired resistance to prior 1696.
ERBB family in cancer: couples therapy. Nat Rev TRK kinase inhibition in patients with TRK 589. Diamond EL, et al. Diverse and targetable kinase
Cancer. 2013;13:663–673. fusion-positive solid tumors. Cancer Discov. 2017. alterations drive histiocytic neoplasms. Cancer Discov.
62. Janne PA, et al. AZD9291 in EGFR inhibitor- 278. Lappano R, Maggiolini M. G protein-coupled 2016;6:154–165.
resistant non-small-cell lung cancer. N Engl J Med. receptors: novel targets for drug discovery in cancer. 592. Lito P, Rosen N, Solit DB. Tumor adaptation
2015;372:1689–1699. Nat Rev Drug Discov. 2011;10:47–60. and resistance to RAF inhibitors. Nat Med.
78. Swain SM, et al. Pertuzumab, trastuzumab, and 322. O’Hayre M, et al. The emerging mutational land- 2013;19:1401–1409.
docetaxel for HER2-positive metastatic breast cancer scape of G proteins and G-protein-coupled receptors 593. Yao Z, et al. BRAF mutants evade ERK-dependent
(CLEOPATRA study): overall survival results from a in cancer. Nat Rev Cancer. 2013;13:412–424. feedback by different mechanisms that determine
randomised, double-blind, placebo-controlled, phase 334. Briscoe J, Therond PP. The mechanisms of Hedgehog their sensitivity to pharmacologic inhibition. Cancer
3 study. Lancet Oncol. 2013;14:461–471. signalling and its roles in development and disease. Cell. 2015;28:370–383.
103. Gombos A, Metzger-Filho O, Dal Lago L, Awada- Nat Rev Mol Cell Biol. 2013;14:416–429. 595. Van Allen EM, et al. The genetic landscape of clinical
Hussein A. Clinical development of insulin-like 344. Migden MR, et al. Treatment with two different resistance to RAF inhibition in metastatic melanoma.
growth factor receptor-1 (IGF-1R) inhibitors: doses of sonidegib in patients with locally advanced Cancer Discov. 2014;4:94–109.
At the crossroad? Invest New Drugs. 2012;30: or metastatic basal cell carcinoma (BOLT): a mul- 602. Ostrem JM, Peters U, Sos ML, Wells JA, Shokat
2433–2442. ticentre, randomised, double-blind phase 2 trial. KM. K-Ras(G12C) inhibitors allosterically control
106. Takeuchi K, et al. RET, ROS1 and ALK fusions in Lancet Oncol. 2015;16:716–728. GTP affinity and effector interactions. Nature.
lung cancer. Nat Med. 2012;18:378–381. 394. Verstovsek S, et al. A double-blind, placebo- 2013;503:548–551.
117. Shaw AT, et  al. Alectinib in ALK-positive, controlled trial of ruxolitinib for myelofibrosis. 619. Long GV, et al. Combined BRAF and MEK inhibi-
crizotinib-resistant, non-small-cell lung cancer: a N Engl J Med. 2012;366:799–807. tion versus BRAF inhibition alone in melanoma.
single-group, multicentre, phase 2 trial. Lancet Oncol. 396. O’Shea JJ, Holland SM, Staudt LM. JAKs and STATs N Engl J Med. 2014;371:1877–1888.
2016;17:234–242. in immunity, immunodeficiency, and cancer. N Engl 632. Herrero A, et al. Small molecule inhibition of ERK
150. Obata Y, et al. Oncogenic signaling by Kit tyrosine J Med. 2013;368:161–170. dimerization prevents tumorigenesis by RAS-ERK
kinase occurs selectively on the Golgi apparatus 416. David CJ, et al. TGF-beta tumor suppression through pathway oncogenes. Cancer Cell. 2015;28:170–182.
in gastrointestinal stromal tumors. Oncogene. a lethal EMT. Cell. 2016;164:1015–1030. 635. Li BT, et al. First-in-class oral ERK1/2 inhibitor
2017;36:3661–3672. 421. Takebe N, Nguyen D, Yang SX. Targeting notch ulixertinib (BVD-523) in patients with advanced
169. Javidi-Sharifi N, et al. Crosstalk between KIT signaling pathway in cancer: clinical development solid tumors: final results of a phase I dose escala-
and FGFR3 promotes gastrointestinal stromal advances and challenges. Pharmacol Ther. 2014;141: tion and expansion study. J Clin Oncol. 2017;35:
tumor cell growth and drug resistance. Cancer Res. 140–149. 2508.
2015;75:880–891. 437. Yen WC, et al. Targeting Notch signaling with a 639. Hu-Lieskovan S, et al. Improved antitumor activity
182. Babina IS, Turner NC. Advances and challenges Notch2/Notch3 antagonist (tarextumab) inhibits of immunotherapy with BRAF and MEK inhibi-
in targeting FGFR signalling in cancer. Nat Rev tumor growth and decreases tumor-initiating cell tors in BRAF(V600E) melanoma. Sci Transl Med.
Cancer. 2017;17:318–332. frequency. Clin Cancer Res. 2015;21:2084–2095. 2015;7:279ra241.
46 Part I: Science and Clinical Oncology

644. Beaver JA, et al. FDA approval: palbociclib for the 691. Thorpe LM, et al. PI3K-p110alpha mediates the 717. Rodrik-Outmezguine VS, et al. Overcoming mTOR
treatment of postmenopausal patients with estrogen oncogenic activity induced by loss of the novel tumor resistance mutations with a new-generation mTOR
receptor-positive, HER2-negative metastatic breast suppressor PI3K-p85alpha. Proc Natl Acad Sci USA. inhibitor. Nature. 2016;534:272–276.
cancer. Clin Cancer Res. 2015;21:4760–4766. 2017;114:7095–7100. 720. Zehir A, et al. Mutational landscape of metastatic
690. Cheung LW, et al. Naturally occurring neomorphic 704. Hyman DM, et al. AKT Inhibition in solid tumors cancer revealed from prospective clinical sequenc-
PIK3R1 mutations activate the MAPK pathway, with AKT1 mutations. J Clin Oncol. 2017;35: ing of 10,000 patients. Nat Med. 2017;23:703–
dictating therapeutic response to MAPK pathway 2251–2259. 713.
inhibitors. Cancer Cell. 2014;26:479–494.
Intracellular Signaling  •  CHAPTER 2 46.e1
46.e1

REFERENCES
1. Chakravarty D, et al. OncoKB: A precision oncology 25. Zaczek A, Brandt B, Bielawski KP. The diverse signal- 46. Bose R, et al. Activating HER2 mutations in HER2
knowledge base. JCO Precis Oncol. 2017;1–16. ing network of EGFR, HER2, HER3 and HER4 gene amplification negative breast cancer. Cancer
2. Robinson DR, Wu YM, Lin SF. The protein tyrosine tyrosine kinase receptors and the consequences for Discov. 2013;3:224–237.
kinase family of the human genome. Oncogene. therapeutic approaches. Histol Histopathol. 2005;20: 47. Greulich H, et al. Functional analysis of receptor
2000;19:5548–5557. 1005–1015. tyrosine kinase mutations in lung cancer identifies
3. Schlessinger J. Cell signaling by receptor tyrosine 26. Worthylake R, Opresko LK, Wiley HS. ErbB-2 oncogenic extracellular domain mutations of ERBB2.
kinases. Cell. 2000;103:211–225. amplification inhibits down-regulation and induces Proc Natl Acad Sci USA. 2012;109:14476–14481.
4. Hubbard SR, Till JH. Protein tyrosine kinase constitutive activation of both ErbB-2 and epidermal 48. Jaiswal BS, et al. Oncogenic ERBB3 mutations in
structure and function. Annu Rev Biochem. 2000;69: growth factor receptors. J Biol Chem. 1999;274: human cancers. Cancer Cell. 2013;23:603–617.
373–398. 8865–8874. 49. Hanrahan AJ, Hyman DM, Sfakianos J, Jones
5. Kuriyan J, Cowburn D. Modular peptide recognition 27. Okines A, Cunningham D, Chau I. Targeting the A, Ramirez R, Johnsen H, et al. Abstract 1101:
domains in eukaryotic signaling. Annu Rev Biophys human EGFR family in esophagogastric cancer. Nat Functional genomics of HER2 and HER3 muta-
Biomol Struct. 1997;26:259–288. Rev Clin Oncol. 2011;8:492–503. tions and response to neratinib. AACR Cancer Res.
6. Pawson T. Protein modules and signalling networks. 28. Pinkas-Kramarski R, et al. Diversification of Neu 2015;75:1101.
Nature. 1995;373:573–580. differentiation factor and epidermal growth factor 50. Tebbutt N, Pedersen MW, Johns TG. Targeting the
7. Liu BA, et al. The human and mouse complement signaling by combinatorial receptor interactions. ERBB family in cancer: couples therapy. Nat Rev
of SH2 domain proteins-establishing the boundaries EMBO J. 1996;15:2452–2467. Cancer. 2013;13:663–673.
of phosphotyrosine signaling. Mol Cell. 2006;22: 29. Janne PA, Engelman JA, Johnson BE. Epidermal 51. Cunningham D, et al. Cetuximab monotherapy
851–868. growth factor receptor mutations in non-small-cell and cetuximab plus irinotecan in irinotecan-
8. Pawson T. Specificity in signal transduction: from lung cancer: implications for treatment and tumor refractory metastatic colorectal cancer. N Engl J
phosphotyrosine-SH2 domain interactions to biology. J Clin Oncol. 2005;23:3227–3234. Med. 2004;351:337–345.
complex cellular systems. Cell. 2004;116:191–203. 30. Pao W, et al. EGF receptor gene mutations are 52. Saltz LB, et al. Phase II trial of cetuximab in patients
9. Pawson T, Nash P. Assembly of cell regulatory common in lung cancers from “never smokers” and with refractory colorectal cancer that expresses the epi-
systems through protein interaction domains. Science. are associated with sensitivity of tumors to gefitinib dermal growth factor receptor. J Clin Oncol. 2004;22:
2003;300:445–452. and erlotinib. Proc Natl Acad Sci USA. 2004;101: 1201–1208.
10. Levi-Montalcini R. Effects of mouse tumor trans- 13306–13311. 53. Bonner JA, et al. Radiotherapy plus cetuximab for
plantation on the nervous system. Ann N Y Acad 31. Chen Z, Fillmore CM, Hammerman PS, Kim CF, squamous-cell carcinoma of the head and neck. N
Sci. 1952;55:330–344. Wong KK. Non-small-cell lung cancers: a hetero- Engl J Med. 2006;354:567–578.
11. Gschwind A, Fischer OM, Ullrich A. The discovery geneous set of diseases. Nat Rev Cancer. 2014;14: 54. Chung KY, et al. Cetuximab shows activity in
of receptor tyrosine kinases: targets for cancer 535–546. colorectal cancer patients with tumors that do
therapy. Nat Rev Cancer. 2004;4:361–370. 32. Wong AJ, et al. Structural alterations of the epidermal not express the epidermal growth factor receptor
12. Cohen S. Isolation of a mouse submaxillary gland growth factor receptor gene in human gliomas. Proc by immunohistochemistry. J Clin Oncol. 2005;23:
protein accelerating incisor eruption and eyelid opening Natl Acad Sci USA. 1992;89:2965–2969. 1803–1810.
in the new-born animal. J Biol Chem. 1962;237: 33. Li B, et al. Mutant epidermal growth factor receptor 55. Van Cutsem E, et al. Cetuximab and chemotherapy
1555–1562. displays increased signaling through the phospha- as initial treatment for metastatic colorectal cancer.
13. Carpenter G, King L Jr, Cohen S. Epidermal growth tidylinositol-3 kinase/AKT pathway and promotes N Engl J Med. 2009;360:1408–1417.
factor stimulates phosphorylation in membrane radioresistance in cells of astrocytic origin. Oncogene. 56. Price TJ, et al. Panitumumab versus cetuximab
preparations in vitro. Nature. 1978;276:409–410. 2004;23:4594–4602. in patients with chemotherapy-refractory wild-
14. Eckhart W, Hutchinson MA, Hunter T. An activity 34. Brennan CW, et al. The somatic genomic landscape type KRAS exon 2 metastatic colorectal cancer
phosphorylating tyrosine in polyoma T antigen of glioblastoma. Cell. 2013;155:462–477. (ASPECCT): a randomised, multicentre, open-label,
immunoprecipitates. Cell. 1979;18:925–933. 35. Furnari FB, Cloughesy TF, Cavenee WK, Mischel PS. non-inferiority phase 3 study. Lancet Oncol. 2014;15:
15. Hunter T, Sefton BM. Transforming gene product Heterogeneity of epidermal growth factor receptor 569–579.
of Rous sarcoma virus phosphorylates tyrosine. Proc signalling networks in glioblastoma. Nat Rev Cancer. 57. Lynch TJ, et al. Activating mutations in the epider-
Natl Acad Sci USA. 1980;77:1311–1315. 2015;15:302–310. mal growth factor receptor underlying responsiveness
16. Ullrich A, et al. Human epidermal growth factor 36. Tanaka S, Louis DN, Curry WT, Batchelor TT, of non-small-cell lung cancer to gefitinib. N Engl J
receptor cDNA sequence and aberrant expression of Dietrich J. Diagnostic and therapeutic avenues for Med. 2004;350:2129–2139.
the amplified gene in A431 epidermoid carcinoma glioblastoma: no longer a dead end? Nat Rev Clin 58. Yang JC, et al. Afatinib versus cisplatin-based
cells. Nature. 1984;309:418–425. Oncol. 2013;10:14–26. chemotherapy for EGFR mutation-positive lung
17. Yamamoto T, Hihara H, Nishida T, Kawai S, 37. Francis JM, et al. EGFR variant heterogeneity adenocarcinoma (LUX-Lung 3 and LUX-Lung 6):
Toyoshima K. A new avian erythroblastosis virus, in glioblastoma resolved through single-nucleus analysis of overall survival data from two randomised,
AEV-H, carries erbB gene responsible for the sequencing. Cancer Discov. 2014;4:956–971. phase 3 trials. Lancet Oncol. 2015;16:141–151.
induction of both erythroblastosis and sarcomas. 38. Nathanson DA, et al. Targeted therapy resistance 59. Rosell R, et al. Erlotinib versus standard che-
Cell. 1983;34:225–232. mediated by dynamic regulation of extrachromosomal motherapy as first-line treatment for European
18. Downward J, et al. Close similarity of epidermal mutant EGFR DNA. Science. 2014;343:72–76. patients with advanced EGFR mutation-positive
growth factor receptor and v-erb-B oncogene protein 39. Krause DS, Van Etten RA. Tyrosine kinases as targets non-small-cell lung cancer (EURTAC): a multicentre,
sequences. Nature. 1984;307:521–527. for cancer therapy. N Engl J Med. 2005;353:172–187. open-label, randomised phase 3 trial. Lancet Oncol.
19. Yarden Y, Sliwkowski MX. Untangling the ErbB 40. Hirsch FR, et al. Epidermal growth factor receptor in 2012;13:239–246.
signalling network. Nat Rev Mol Cell Biol. 2001;2: non-small-cell lung carcinomas: correlation between 60. Maemondo M, et al. Gefitinib or chemotherapy for
127–137. gene copy number and protein expression and non-small-cell lung cancer with mutated EGFR.
20. Jones JT, Akita RW, Sliwkowski MX. Binding impact on prognosis. J Clin Oncol. 2003;21:3798– N Engl J Med. 2010;362:2380–2388.
specificities and affinities of egf domains for ErbB 3807. 61. Skoulidis F, Papadimitrakopoulou VA. Targeting
receptors. FEBS Lett. 1999;447:227–231. 41. Cancer Genome Atlas Research, N. et al. Integrated the gatekeeper: osimertinib in EGFR T790M
21. Yarden Y, Pines G. The ERBB network: at last, genomic characterization of oesophageal carcinoma. mutation-positive non-small cell lung cancer. Clin
cancer therapy meets systems biology. Nat Rev Nature. 2017;541:169–175. Cancer Res. 2017;23:618–622.
Cancer. 2012;12:553–563. 42. Slamon DJ, et al. Human breast cancer: correlation 62. Janne PA, et al. AZD9291 in EGFR inhibitor-
22. Marshall J. Clinical implications of the mechanism of of relapse and survival with amplification of the resistant non-small-cell lung cancer. N Engl J Med.
epidermal growth factor receptor inhibitors. Cancer. HER-2/neu oncogene. Science. 1987;235:177–182. 2015;372:1689–1699.
2006;107:1207–1218. 43. Cristescu R, et al. Molecular analysis of gastric cancer 63. Jia Y, et al. Overcoming EGFR(T790M) and
23. Klapper LN, et al. The ErbB-2/HER2 oncoprotein identifies subtypes associated with distinct clinical EGFR(C797S) resistance with mutant-selective
of human carcinomas may function solely as a shared outcomes. Nat Med. 2015;21:449–456. allosteric inhibitors. Nature. 2016;534:129–132.
coreceptor for multiple stroma-derived growth 44. Cancer Genome Atlas Research, N. Comprehensive 64. Valabrega G, Montemurro F, Aglietta M. Trastu-
factors. Proc Natl Acad Sci USA. 1999;96:4995–5000. molecular characterization of urothelial bladder zumab: mechanism of action, resistance and future
24. Graus-Porta D, Beerli RR, Daly JM, Hynes NE. carcinoma. Nature. 2014;507:315–322. perspectives in HER2-overexpressing breast cancer.
ErbB-2, the preferred heterodimerization partner of 45. Cancer Genome Atlas Research, N. Comprehensive Ann Oncol. 2007;18:977–984.
all ErbB receptors, is a mediator of lateral signaling. molecular characterization of gastric adenocarcinoma. 65. Cobleigh MA, et al. Multinational study of the effi-
EMBO J. 1997;16:1647–1655. Nature. 2014;513:202–209. cacy and safety of humanized anti-HER2 monoclonal
46.e2 Part I: Science and Clinical Oncology

antibody in women who have HER2-overexpressing postmenopausal hormone receptor-positive metastatic stromal tumors. Proc Natl Acad Sci USA. 2008;105:
metastatic breast cancer that has progressed after breast cancer. J Clin Oncol. 2009;27:5538–5546. 8387–8392.
chemotherapy for metastatic disease. J Clin Oncol. 85. Frampton JE. Lapatinib: a review of its use in the 105. Hallberg B, Palmer RH. Mechanistic insight into
1999;17:2639–2648. treatment of HER2-overexpressing, trastuzumab- ALK receptor tyrosine kinase in human cancer
66. Slamon DJ, et al. Use of chemotherapy plus a refractory, advanced or metastatic breast cancer. biology. Nat Rev Cancer. 2013;13:685–700.
monoclonal antibody against HER2 for metastatic Drugs. 2009;69:2125–2148. 106. Takeuchi K, et al. RET, ROS1 and ALK fusions in
breast cancer that overexpresses HER2. N Engl J 86. Serra V, et al. Clinical response to a lapatinib-based lung cancer. Nat Med. 2012;18:378–381.
Med. 2001;344:783–792. therapy for a Li-Fraumeni syndrome patient with 107. Sonnenberg-Riethmacher E, Walter B, Riethmacher
67. Piccart-Gebhart MJ, et al. Trastuzumab after adjuvant a novel HER2V659E mutation. Cancer Discov. D, Godecke S, Birchmeier C. The c-ros tyrosine
chemotherapy in HER2-positive breast cancer. N 2013;3:1238–1244. kinase receptor controls regionalization and dif-
Engl J Med. 2005;353:1659–1672. 87. Wong KK, et al. A phase I study with neratinib ferentiation of epithelial cells in the epididymis.
68. Perez EA, et al. Four-year follow-up of trastuzumab (HKI-272), an irreversible pan ErbB receptor Genes Dev. 1996;10:1184–1193.
plus adjuvant chemotherapy for operable human tyrosine kinase inhibitor, in patients with solid 108. Camidge DR, Doebele RC. Treating ALK-positive
epidermal growth factor receptor 2-positive breast tumors. Clin Cancer Res. 2009;15:2552–2558. lung cancer—early successes and future challenges.
cancer: joint analysis of data from NCCTG N9831 88. Burstein HJ, et al. Neratinib, an irreversible ErbB Nat Rev Clin Oncol. 2012;9:268–277.
and NSABP B-31. J Clin Oncol. 2011;29:3366– receptor tyrosine kinase inhibitor, in patients with 109. Soda M, et al. Identification of the transforming
3373. advanced ErbB2-positive breast cancer. J Clin Oncol. EML4-ALK fusion gene in non-small-cell lung
69. Goldhirsch A, et al. 2 years versus 1 year of adjuvant 2010;28:1301–1307. cancer. Nature. 2007;448:561–566.
trastuzumab for HER2-positive breast cancer 89. Awada A, et al. Safety and efficacy of neratinib 110. Takeuchi K, et al. Multiplex reverse transcription-
(HERA): an open-label, randomised controlled (HKI-272) plus vinorelbine in the treatment of PCR screening for EML4-ALK fusion transcripts.
trial. Lancet. 2013;382:1021–1028. patients with ErbB2-positive metastatic breast Clin Cancer Res. 2008;14:6618–6624.
70. Junttila TT, Li G, Parsons K, Phillips GL, Sliwkowski cancer pretreated with anti-HER2 therapy. Ann 111. Bergethon K, et al. ROS1 rearrangements define
MX. Trastuzumab-DM1 (T-DM1) retains all the Oncol. 2013;24:109–116. a unique molecular class of lung cancers. J Clin
mechanisms of action of trastuzumab and efficiently 90. Chow LW, et al. Combination neratinib (HKI-272) Oncol. 2012;30:863–870.
inhibits growth of lapatinib insensitive breast cancer. and paclitaxel therapy in patients with HER2-positive 112. Davies KD, et al. Identifying and targeting ROS1
Breast Cancer Res Treat. 2011;128:347–356. metastatic breast cancer. Br J Cancer. 2013;108: gene fusions in non-small cell lung cancer. Clin
71. Verma S, et al. Trastuzumab emtansine for HER2-pos- 1985–1993. Cancer Res. 2012;18:4570–4579.
itive advanced breast cancer. N Engl J Med. 2012;367: 91. Jankowitz RC, et al. Safety and efficacy of neratinib 113. Kwak EL, et al. Anaplastic lymphoma kinase inhibi-
1783–1791. in combination with weekly paclitaxel and trastu- tion in non-small-cell lung cancer. N Engl J Med. 2010;
72. Bang YJ, et al. Trastuzumab in combination with zumab in women with metastatic HER2positive 363:1693–1703.
chemotherapy versus chemotherapy alone for breast cancer: an NSABP Foundation Research 114. Shaw AT, et al. Crizotinib in ROS1-rearranged
treatment of HER2-positive advanced gastric or Program phase I study. Cancer Chemother Pharmacol. non-small-cell lung cancer. N Engl J Med. 2014;371:
gastro-oesophageal junction cancer (ToGA): a phase 2013;72:1205–1212. 1963–1971.
3, open-label, randomised controlled trial. Lancet. 92. Saura C, et al. Safety and efficacy of neratinib in com- 115. Shaw AT, et al. Crizotinib versus chemotherapy in
2010;376:687–697. bination with capecitabine in patients with metastatic advanced ALK-positive lung cancer. N Engl J Med.
73. Romond EH, et al. Trastuzumab plus adjuvant human epidermal growth factor receptor 2-positive 2013;368:2385–2394.
chemotherapy for operable HER2-positive breast breast cancer. J Clin Oncol. 2014;32:3626–3633. 116. Ou SH, et al. Alectinib in crizotinib-refractory
cancer. N Engl J Med. 2005;353:1673–1684. 93. Hyman D, et al. Abstract PD5-05: Neratinib for ALK-rearranged non-small-cell lung cancer: a phase
74. Nahta R, Esteva FJ. HER-2-targeted therapy: ERBB2 mutant, HER2 non-amplified, metastatic II global study. J Clin Oncol. 2016;34:661–668.
lessons learned and future directions. Clin Cancer breast cancer: Preliminary analysis from a multi- 117. Shaw AT, et  al. Alectinib in ALK-positive,
Res. 2003;9:5078–5084. center, open-label, multi-histology phase II basket crizotinib-resistant, non-small-cell lung cancer: a
75. Franklin MC, et al. Insights into ErbB signaling from trial. Cancer Res. 2016;76:PD5-05-PD05-05. single-group, multicentre, phase 2 trial. Lancet Oncol.
the structure of the ErbB2-pertuzumab complex. 94. Baserga R, Peruzzi F, Reiss K. The IGF-1 receptor 2016;17:234–242.
Cancer Cell. 2004;5:317–328. in cancer biology. Int J Cancer. 2003;107:873–877. 118. Gettinger SN, et al. Activity and safety of brigatinib
76. Baselga J, et al. Pertuzumab plus trastuzumab plus 95. Belfiore A, Frasca F, Pandini G, Sciacca L, Vigneri in ALK-rearranged non-small-cell lung cancer and
docetaxel for metastatic breast cancer. N Engl J Med. R. Insulin receptor isoforms and insulin receptor/ other malignancies: a single-arm, open-label, phase
2012;366:109–119. insulin-like growth factor receptor hybrids in physiol- 1/2 trial. Lancet Oncol. 2016;17:1683–1696.
77. Gianni L, et al. Efficacy and safety of neoadjuvant ogy and disease. Endocr Rev. 2009;30:586–623. 119. Hoch RV, Soriano P. Roles of PDGF in animal
pertuzumab and trastuzumab in women with 96. Benyoucef S, Surinya KH, Hadaschik D, Siddle K. development. Development. 2003;130:4769–4784.
locally advanced, inflammatory, or early HER2- Characterization of insulin/IGF hybrid receptors: 120. Heldin CH, Westermark B. Mechanism of action
positive breast cancer (NeoSphere): a randomised contributions of the insulin receptor L2 and Fn1 and in vivo role of platelet-derived growth factor.
multicentre, open-label, phase 2 trial. Lancet Oncol. domains and the alternatively spliced exon 11 Physiol Rev. 1999;79:1283–1316.
2012;13:25–32. sequence to ligand binding and receptor activation. 121. Heldin CH, Eriksson U, Ostman A. New members of
78. Swain SM, et al. Pertuzumab, trastuzumab, and Biochem J. 2007;403:603–613. the platelet-derived growth factor family of mitogens.
docetaxel for HER2-positive metastatic breast cancer 97. Jones JI, Clemmons DR. Insulin-like growth factors Arch Biochem Biophys. 2002;398:284–290.
(CLEOPATRA study): overall survival results from a and their binding proteins: biological actions. Endocr 122. Ostman A, Heldin CH. PDGF receptors as targets in
randomised, double-blind, placebo-controlled, phase Rev. 1995;16:3–34. tumor treatment. Adv Cancer Res. 2007;97:247–274.
3 study. Lancet Oncol. 2013;14:461–471. 98. De Meyts P, Whittaker J. Structural biology of insulin 123. Carmeliet P, Jain RK. Molecular mechanisms
79. Shojaei S, Gardaneh M, Rahimi Shamabadi A. Target and IGF1 receptors: implications for drug design. and clinical applications of angiogenesis. Nature.
points in trastuzumab resistance. Int J Breast Cancer. Nat Rev Drug Discov. 2002;1:769–783. 2011;473:298–307.
2012;2012:761917. 99. Firth SM, Baxter RC. Cellular actions of the insulin- 124. Seger R, Krebs EG. The MAPK signaling cascade.
80. Geyer CE, et al. Lapatinib plus capecitabine for like growth factor binding proteins. Endocr Rev. FASEB J. 1995;9:726–735.
HER2-positive advanced breast cancer. N Engl J 2002;23:824–854. 125. Andrae J, Gallini R, Betsholtz C. Role of platelet-
Med. 2006;355:2733–2743. 100. Cohen P, et al. Prostate-specific antigen (PSA) is an derived growth factors in physiology and medicine.
81. Xia W, et al. Combining lapatinib (GW572016), a insulin-like growth factor binding protein-3 protease Genes Dev. 2008;22:1276–1312.
small molecule inhibitor of ErbB1 and ErbB2 tyro- found in seminal plasma. J Clin Endocrinol Metab. 126. Corless CL, et  al. PDGFRA mutations in
sine kinases, with therapeutic anti-ErbB2 antibodies 1992;75:1046–1053. gastrointestinal stromal tumors: frequency, spectrum
enhances apoptosis of ErbB2-overexpressing breast 101. Nakae J, Kido Y, Accili D. Distinct and overlapping and in vitro sensitivity to imatinib. J Clin Oncol.
cancer cells. Oncogene. 2005;24:6213–6221. functions of insulin and IGF-I receptors. Endocr 2005;23:5357–5364.
82. Scaltriti M, et al. Lapatinib, a HER2 tyrosine kinase Rev. 2001;22:818–835. 127. Carroll M, Tomasson MH, Barker GF, Golub TR,
inhibitor, induces stabilization and accumulation of 102. Pollak MN, Schernhammer ES, Hankinson SE. Gilliland DG. The TEL/platelet-derived growth
HER2 and potentiates trastuzumab-dependent cell Insulin-like growth factors and neoplasia. Nat Rev factor beta receptor (PDGF beta R) fusion in
cytotoxicity. Oncogene. 2009;28:803–814. Cancer. 2004;4:505–518. chronic myelomonocytic leukemia is a transforming
83. Baselga J, et al. Lapatinib with trastuzumab for 103. Gombos A, Metzger-Filho O, Dal Lago L, Awada- protein that self-associates and activates PDGF beta
HER2-positive early breast cancer (NeoALTTO): Hussein A. Clinical development of insulin-like R kinase-dependent signaling pathways. Proc Natl
a randomised, open-label, multicentre, phase 3 trial. growth factor receptor-1 (IGF-1R) inhibitors: At the Acad Sci USA. 1996;93:14845–14850.
Lancet. 2012;379:633–640. crossroad? Invest New Drugs. 2012;30:2433–2442. 128. McArthur GA. Dermatofibrosarcoma protuberans:
84. Johnston S, et al. Lapatinib combined with letrozole 104. Tarn C, et al. Insulin-like growth factor 1 receptor a surgical disease with a molecular savior. Curr Opin
versus letrozole and placebo as first-line therapy for is a potential therapeutic target for gastrointestinal Oncol. 2006;18:341–346.
Intracellular Signaling  •  CHAPTER 2 46.e3
46.e3

129. Cools J, et al. A tyrosine kinase created by fusion gastrointestinal stromal tumors. Oncogene. 2017;36: 173. Antar A, et al. Inhibition of FLT3 in AML: a focus
of the PDGFRA and FIP1L1 genes as a therapeutic 3661–3672. on sorafenib. Bone Marrow Transplant. 2017;52:
target of imatinib in idiopathic hypereosinophilic 151. Thomsen L, et al. Interstitial cells of Cajal gener- 344–351.
syndrome. N Engl J Med. 2003;348:1201–1214. ate a rhythmic pacemaker current. Nat Med. 174. Yu J, et al. Anti-tumor activity of TAK-659, a dual
130. Fleming TP, et al. Amplification and/or overexpres- 1998;4:848–851. inhibitor of SYK and FLT-3 kinases, in AML models.
sion of platelet-derived growth factor receptors and 152. Corless CL, Barnett CM, Heinrich MC. Gastro- J Clin Oncol. 2016;34:e14091.
epidermal growth factor receptor in human glial intestinal stromal tumours: origin and molecular 175. Kaplan J, et al. TAK-659, An investigational revers-
tumors. Cancer Res. 1992;52:4550–4553. oncology. Nat Rev Cancer. 2011;11:865–878. ible dual SYK/FLT-3 inhibitor, in patients with
131. Iqbal N, Iqbal N. Imatinib: a breakthrough of targeted 153. Hirota S, et al. Gain-of-function mutations of c-kit lymphoma: updated results from dose-escalation
therapy in cancer. Chemother Res Pract. 2014;2014: in human gastrointestinal stromal tumors. Science. and expansion cohorts of a phase 1 study. Hematol
357027. 1998;279:577–580. Oncol. 2017;35:72–74.
132. von Mehren M. Beyond imatinib: second gen- 154. Kindblom LG, Remotti HE, Aldenborg F, Meis- 176. Zhang W, et al. Mutant FLT3: a direct target of
eration c-KIT inhibitors for the management of Kindblom JM. Gastrointestinal pacemaker cell tumor sorafenib in acute myelogenous leukemia. J Natl
gastrointestinal stromal tumors. Clin Colorectal (GIPACT): gastrointestinal stromal tumors show Cancer Inst. 2008;100:184–198.
Cancer. 2006;6(suppl 1):S30–S34. phenotypic characteristics of the interstitial cells of 177. Knowles MA. Role of FGFR3 in urothelial cell
133. Dewaele B, et al. Activity of dasatinib, a dual SRC/ Cajal. Am J Pathol. 1998;152:1259–1269. carcinoma: biomarker and potential therapeutic
ABL kinase inhibitor, and IPI-504, a heat shock 155. Fletcher JA, Rubin BP. KIT mutations in GIST. target. World J Urol. 2007;25:581–593.
protein 90 inhibitor, against gastrointestinal stromal Curr Opin Genet Dev. 2007;17:3–7. 178. Ornitz DM, et al. Receptor specificity of the fibro-
tumor-associated PDGFRAD842V mutation. Clin 156. Bagrodia A, et al. Genetic determinants of cisplatin blast growth factor family. J Biol Chem. 1996;271:
Cancer Res. 2008;14:5749–5758. resistance in patients with advanced germ cell tumors. 15292–15297.
134. McArthur GA, et  al. Molecular and clinical J Clin Oncol. 2016;34:4000–4007. 179. Turner N, Grose R. Fibroblast growth factor signal-
analysis of locally advanced dermatofibrosarcoma 157. Demetri GD, et al. Efficacy and safety of imatinib ling: from development to cancer. Nat Rev Cancer.
protuberans treated with imatinib: Imatinib Target mesylate in advanced gastrointestinal stromal tumors. 2010;10:116–129.
Exploration Consortium Study B2225. J Clin Oncol. N Engl J Med. 2002;347:472–480. 180. Ornitz DM, Marie PJ. Fibroblast growth factor
2005;23:866–873. 158. Demetri GD, et al. Efficacy and safety of sunitinib signaling in skeletal development and disease. Genes
135. Kerob D, et al. Imatinib mesylate as a preopera- in patients with advanced gastrointestinal stromal Dev. 2015;29:1463–1486.
tive therapy in dermatofibrosarcoma: results of a tumour after failure of imatinib: a randomised 181. Iyer G, Milowsky MI. Fibroblast growth factor
multicenter phase II study on 25 patients. Clin controlled trial. Lancet. 2006;368:1329–1338. receptor-3 in urothelial tumorigenesis. Urol Oncol.
Cancer Res. 2010;16:3288–3295. 159. Verweij J, et al. Progression-free survival in gastro- 2012.
136. Apperley JF, et al. Response to imatinib mesylate in intestinal stromal tumours with high-dose imatinib: 182. Babina IS, Turner NC. Advances and challenges
patients with chronic myeloproliferative diseases with randomised trial. Lancet. 2004;364:1127–1134. in targeting FGFR signalling in cancer. Nat Rev
rearrangements of the platelet-derived growth factor 160. Zalcberg JR, et al. Outcome of patients with Cancer. 2017;17:318–332.
receptor beta. N Engl J Med. 2002;347:481–487. advanced gastro-intestinal stromal tumours crossing 183. Wang Y, Becker D. Antisense targeting of basic
137. Cheah CY, et al. Patients with myeloid malignancies over to a daily imatinib dose of 800 mg after progres- fibroblast growth factor and fibroblast growth
bearing PDGFRB fusion genes achieve durable sion on 400 mg. Eur J Cancer. 2005;41:1751–1757. factor receptor-1 in human melanomas blocks
long-term remissions with imatinib. Blood. 161. Blanke CD, et al. Long-term results from a random- intratumoral angiogenesis and tumor growth. Nat
2014;123:3574–3577. ized phase II trial of standard- versus higher-dose Med. 1997;3:887–893.
138. Roberts KG, et al. Genetic alterations activating imatinib mesylate for patients with unresectable or 184. Memarzadeh S, et al. Enhanced paracrine FGF10
kinase and cytokine receptor signaling in high- metastatic gastrointestinal stromal tumors expressing expression promotes formation of multifocal pros-
risk acute lymphoblastic leukemia. Cancer Cell. KIT. J Clin Oncol. 2008;26:620–625. tate adenocarcinoma and an increase in epithelial
2012;22:153–166. 162. George S, et al. Clinical evaluation of continuous androgen receptor. Cancer Cell. 2007;12:572–585.
139. Andre C, et al. Sequence analysis of two genomic daily dosing of sunitinib malate in patients with 185. Weiss J, et al. Frequent and focal FGFR1 amplifica-
regions containing the KIT and the FMS receptor advanced gastrointestinal stromal tumour after tion associates with therapeutically tractable FGFR1
tyrosine kinase genes. Genomics. 1997;39:216–226. imatinib failure. Eur J Cancer. 2009;45:1959–1968. dependency in squamous cell lung cancer. Sci Transl
140. Yarden Y, et al. Human proto-oncogene c-kit: 163. Reichardt P, et al. Clinical outcomes of patients Med. 2010;2:62ra93.
a new cell surface receptor tyrosine kinase for with advanced gastrointestinal stromal tumors: 186. Takeda M, et al. AZD2171 shows potent antitumor
an unidentified ligand. EMBO J. 1987;6:3341– safety and efficacy in a worldwide treatment-use activity against gastric cancer over-expressing fibro-
3351. trial of sunitinib. Cancer. 2015;121:1405–1413. blast growth factor receptor 2/keratinocyte growth
141. Ashman LK. The biology of stem cell factor and 164. Demetri GD, et al. Efficacy and safety of regorafenib factor receptor. Clin Cancer Res. 2007;13:3051–
its receptor C-kit. Int J Biochem Cell Biol. 1999;31: for advanced gastrointestinal stromal tumours 3057.
1037–1051. after failure of imatinib and sunitinib (GRID): an 187. Matsumoto K, et al. FGFR2 gene amplification and
142. Besmer P, et al. A new acute transforming feline international, multicentre, randomised, placebo- clinicopathological features in gastric cancer. Br J
retrovirus and relationship of its oncogene v-kit controlled, phase 3 trial. Lancet. 2013;381:295–302. Cancer. 2012;106:727–732.
with the protein kinase gene family. Nature. 165. Carvajal RD, et al. KIT as a therapeutic target in 188. Dutt A, et al. Drug-sensitive FGFR2 mutations in
1986;320:415–421. metastatic melanoma. JAMA. 2011;305:2327–2334. endometrial carcinoma. Proc Natl Acad Sci USA.
143. Huang E, et al. The hematopoietic growth factor 166. Hodi FS, et al. Imatinib for melanomas harboring 2008;105:8713–8717.
KL is encoded by the Sl locus and is the ligand of mutationally activated or amplified KIT arising on 189. Helsten T, et al. The FGFR landscape in cancer:
the c-kit receptor, the gene product of the W locus. mucosal, acral, and chronically sun-damaged skin. analysis of 4,853 tumors by next-generation sequenc-
Cell. 1990;63:225–233. J Clin Oncol. 2013;31:3182–3190. ing. Clin Cancer Res. 2016;22:259–267.
144. Mol CD, et al. Structural basis for the autoinhibition 167. Nishida T, et al. Secondary mutations in the 190. Iyer G, et al. Prevalence and co-occurrence of
and STI-571 inhibition of c-Kit tyrosine kinase. kinase domain of the KIT gene are predominant actionable genomic alterations in high-grade bladder
J Biol Chem. 2004;279:31655–31663. in imatinib-resistant gastrointestinal stromal tumor. cancer. J Clin Oncol. 2013;31:3133–3140.
145. Lev S, Yarden Y, Givol D. Dimerization and Cancer Sci. 2008;99:799–804. 191. Bernard-Pierrot I, et al. Oncogenic properties of the
activation of the kit receptor by monovalent and 168. Bollag G. Abstract IA32: Optimizing kinase inhibi- mutated forms of fibroblast growth factor receptor
bivalent binding of the stem cell factor. J Biol Chem. tors to treat cancer. Cancer Res. 2016;76:IA32. 3b. Carcinogenesis. 2006;27:740–747.
1992;267:15970–15977. 169. Javidi-Sharifi N, et al. Crosstalk between KIT and 192. Sibley K, et  al. A molecular study of the
146. Antonescu CR. The GIST paradigm: lessons for other FGFR3 promotes gastrointestinal stromal tumor cell t(4;14) in multiple myeloma. Br J Haematol.
kinase-driven cancers. J Pathol. 2011;223:251–261. growth and drug resistance. Cancer Res. 2015;75: 2002;118:514–520.
147. Russell ES. Hereditary anemias of the mouse: a 880–891. 193. Wu YM, et al. Identification of targetable FGFR
review for geneticists. Adv Genet. 1979;20:357–459. 170. Stirewalt DL, Radich JP. The role of FLT3 in hae- gene fusions in diverse cancers. Cancer Discov.
148. Dexter TM, Moore MA. In vitro duplication and matopoietic malignancies. Nat Rev Cancer. 2003;3: 2013;3:636–647.
“cure” of haemopoietic defects in genetically anaemic 650–665. 194. Ang C. Role of the fibroblast growth factor receptor
mice. Nature. 1977;269:412–414. 171. Leung AY, Man CH, Kwong YL. FLT3 inhibition: axis in cholangiocarcinoma. J Gastroenterol Hepatol.
149. Gadd SJ, Ashman LK. A murine monoclonal a moving and evolving target in acute myeloid 2015;30:1116–1122.
antibody specific for a cell-surface antigen expressed leukaemia. Leukemia. 2012. 195. Costa R, et al. FGFR3-TACC3 fusion in solid tumors:
by a subgroup of human myeloid leukaemias. Leuk 172. Cancer Genome Atlas Research, N. et al. Genomic mini review. Oncotarget. 2016;7:55924–55938.
Res. 1985;9:1329–1336. and epigenomic landscapes of adult de novo acute 196. Singh D, et al. Transforming fusions of FGFR
150. Obata Y, et al. Oncogenic signaling by Kit tyrosine myeloid leukemia. N Engl J Med. 2013;368: and TACC genes in human glioblastoma. Science.
kinase occurs selectively on the Golgi apparatus in 2059–2074. 2012;337:1231–1235.
46.e4 Part I: Science and Clinical Oncology

197. Nogova L, et al. Evaluation of BGJ398, a fibroblast 219. Sandler A, et al. Paclitaxel-carboplatin alone or with 241. Bladt F, Riethmacher D, Isenmann S, Aguzzi A,
growth factor receptor 1-3 kinase inhibitor, in patients bevacizumab for non-small-cell lung cancer. N Engl Birchmeier C. Essential role for the c-met receptor
with advanced solid tumors harboring genetic altera- J Med. 2006;355:2542–2550. in the migration of myogenic precursor cells into
tions in fibroblast growth factor receptors: results of 220. Vredenburgh JJ, et al. Bevacizumab plus irinotecan the limb bud. Nature. 1995;376:768–771.
a global phase I, dose-escalation and dose-expansion in recurrent glioblastoma multiforme. J Clin Oncol. 242. Appleman LJ. MET signaling pathway: a rational
study. J Clin Oncol. 2017;35:157–165. 2007;25:4722–4729. target for cancer therapy. J Clin Oncol. 2011;29:
198. Paik PK, et al. A Phase 1b open label multicentre 221. Escudier B, et al. Bevacizumab plus interferon alfa-2a 4837–4838.
study of AZD4547 in patients with advanced for treatment of metastatic renal cell carcinoma: a 243. Awad MM, et al. MET exon 14 mutations in non-
squamous cell lung cancers. Clin Cancer Res. 2017. randomised, double-blind phase III trial. Lancet. small-cell lung cancer are associated with advanced
199. Tabernero J, et al. Phase I dose-escalation study of 2007;370:2103–2111. age and stage-dependent MET genomic amplification
JNJ-42756493, an oral pan-fibroblast growth factor 222. Tabernero J, et al. Ramucirumab versus placebo and c-Met overexpression. J Clin Oncol. 2016;34:
receptor inhibitor, in patients with advanced solid in combination with second-line FOLFIRI in 721–730.
tumors. J Clin Oncol. 2015;33:3401–3408. patients with metastatic colorectal carcinoma that 244. Ma PC, et al. Functional expression and mutations of
200. Martin Henner Voss CH, Heist RS, Cleary JM, progressed during or after first-line therapy with c-Met and its therapeutic inhibition with SU11274
Meric-Bernstam F, Gandhi L, Ishii N, et al. 1347, an bevacizumab, oxaliplatin, and a fluoropyrimidine and small interfering RNA in non-small cell lung
oral FGFR inhibitor: results from a first-in-human, (RAISE): a randomised, double-blind, multicentre, cancer. Cancer Res. 2005;65:1479–1488.
phase I dose-escalation study in patients with FGFR phase 3 study. Lancet Oncol. 2015;16:499–508. 245. Ma PC, et al. Expression and mutational analysis of
genomically activated advanced solid tumors. J Clin 223. Chow LQ, Eckhardt SG. Sunitinib: from MET in human solid cancers. Genes Chromosomes
Oncol. 2017;35(suppl; abstr 2500). rational design to clinical efficacy. J Clin Oncol. Cancer. 2008;47:1025–1037.
201. Mulligan LM. RET revisited: expanding the 2007;25:884–896. 246. Liu X, et al. Next-generation sequencing of pulmo-
oncogenic portfolio. Nat Rev Cancer. 2014;14:173– 224. Kane RC, et al. Sorafenib for the treatment of nary sarcomatoid carcinoma reveals high frequency
186. unresectable hepatocellular carcinoma. Oncologist. of actionable MET gene mutations. J Clin Oncol.
202. Costantini F, Shakya R. GDNF/Ret signaling 2009;14:95–100. 2016;34:794–802.
and the development of the kidney. Bioessays. 225. Escudier B, et al. Sorafenib for treatment of renal 247. Schmidt L, et al. Germline and somatic mutations
2006;28:117–127. cell carcinoma: final efficacy and safety results of the in the tyrosine kinase domain of the MET proto-
203. Taraviras S, et al. Signalling by the RET receptor phase III treatment approaches in renal cancer global oncogene in papillary renal carcinomas. Nat Genet.
tyrosine kinase and its role in the development of evaluation trial. J Clin Oncol. 2009;27:3312–3318. 1997;16:68–73.
the mammalian enteric nervous system. Development. 226. Sternberg CN, et al. Pazopanib in locally advanced 248. Peters S, Adjei AA. MET: a promising anticancer
1999;126:2785–2797. or metastatic renal cell carcinoma: results of a therapeutic target. Nat Rev Clin Oncol. 2012;9:
204. Wells SA Jr, Santoro M. Targeting the RET randomized phase III trial. J Clin Oncol. 2010;28: 314–326.
pathway in thyroid cancer. Clin Cancer Res. 1061–1068. 249. Frampton GM, et al. Activation of MET via diverse
2009;15:7119–7123. 227. Rini BI, et al. Comparative effectiveness of axitinib exon 14 splicing alterations occurs in multiple tumor
205. Mulligan LM, et al. Germ-line mutations of the versus sorafenib in advanced renal cell carcinoma types and confers clinical sensitivity to MET inhibi-
RET proto-oncogene in multiple endocrine neoplasia (AXIS): a randomised phase 3 trial. Lancet. 2011;378: tors. Cancer Discov. 2015;5:850–859.
type 2A. Nature. 1993;363:458–460. 1931–1939. 250. Kong-Beltran M, et al. Somatic mutations lead to
206. Lantieri F, Caroli F, Ceccherini I, Griseri P. The 228. Yeung KT, Cohen EE. Lenvatinib in advanced, an oncogenic deletion of met in lung cancer. Cancer
involvement of the RET variant G691S in medullary radioactive iodine-refractory, differentiated thyroid Res. 2006;66:283–289.
thyroid carcinoma enlightened by a meta-analysis carcinoma. Clin Cancer Res. 2015;21:5420–5426. 251. Engelman JA, et al. MET amplification leads to
study. Int J Cancer. 2013;132:2808–2819. 229. Motzer RJ, et al. Lenvatinib, everolimus, and the gefitinib resistance in lung cancer by activating
207. Romei C, Elisei R. RET/PTC translocations and combination in patients with metastatic renal cell ERBB3 signaling. Science. 2007;316:1039–1043.
clinico-pathological features in human papillary carcinoma: a randomised, phase 2, open-label, 252. Paik PK, et al. Response to MET inhibitors in
thyroid carcinoma. Front Endocrinol (Lausanne). multicentre trial. Lancet Oncol. 2015;16:1473–1482. patients with stage IV lung adenocarcinomas
2012;3:54. 230. Loges S, Schmidt T, Carmeliet P. Mechanisms of harboring MET mutations causing exon 14 skipping.
208. Wells SA Jr, et al. Vandetanib in patients with locally resistance to anti-angiogenic therapy and develop- Cancer Discov. 2015;5:842–849.
advanced or metastatic medullary thyroid cancer: ment of third-generation anti-angiogenic drug 253. Ou SH, et al. Activity of crizotinib (PF02341066),
a randomized, double-blind phase III trial. J Clin candidates. Genes Cancer. 2010;1:12–25. a dual mesenchymal-epithelial transition (MET) and
Oncol. 2012;30:134–141. 231. Bottaro DP, et al. Identification of the hepatocyte anaplastic lymphoma kinase (ALK) inhibitor, in a
209. Kurzrock R, et al. Activity of XL184 (Cabozantinib), growth factor receptor as the c-met proto-oncogene non-small cell lung cancer patient with de novo MET
an oral tyrosine kinase inhibitor, in patients with product. Science. 1991;251:802–804. amplification. J Thorac Oncol. 2011;6:942–946.
medullary thyroid cancer. J Clin Oncol. 2011;29: 232. Cooper CS, et al. Molecular cloning of a new 254. Choueiri TK, et al. Cabozantinib versus everolimus
2660–2666. transforming gene from a chemically transformed in advanced renal cell carcinoma (METEOR): final
210. Jordan EJ, et al. Prospective comprehensive molecular human cell line. Nature. 1984;311:29–33. results from a randomised, open-label, phase 3 trial.
characterization of lung adenocarcinomas for effi- 233. Hanna JA, Bordeaux J, Rimm DL, Agarwal S. The Lancet Oncol. 2016;17:917–927.
cient patient matching to approved and emerging function, proteolytic processing, and histopathology 255. Grassi P, et al. Cabozantinib in the treatment of
therapies. Cancer Discov. 2017;7:596–609. of Met in cancer. Adv Cancer Res. 2009;103:1–23. advanced renal cell carcinoma: design, development,
211. Ferrara N, Gerber HP, LeCouter J. The biology of 234. Birchmeier C, Birchmeier W, Gherardi E, Vande and potential place in the therapy. Drug Des Devel
VEGF and its receptors. Nat Med. 2003;9:669–676. Woude GF. Met, metastasis, motility and more. Ther. 2016;10:2167–2172.
212. Leung DW, Cachianes G, Kuang WJ, Goeddel Nat Rev Mol Cell Biol. 2003;4:915–925. 256. Spigel D, Ervin TJ, Ramlau R, et al. Final efficacy
DV, Ferrara N. Vascular endothelial growth 235. Ho-Yen CM, Jones JL, Kermorgant S. The clinical results from OAM4558g, a randomized phase II
factor is a secreted angiogenic mitogen. Science. and functional significance of c-Met in breast cancer: study evaluating MetMAb or placebo in combination
1989;246:1306–1309. a review. Breast Cancer Res. 2015;17:52. with erlotinib in advanced NSCLC. J Clin Oncol.
213. Ferrara N. Vascular endothelial growth factor: basic 236. Peschard P, et al. Mutation of the c-Cbl TKB domain 2011;29.
science and clinical progress. Endocr Rev. 2004;25: binding site on the Met receptor tyrosine kinase 257. Klein R, Jing SQ, Nanduri V, O’Rourke E, Barbacid
581–611. converts it into a transforming protein. Mol Cell. M. The trk proto-oncogene encodes a receptor for
214. Kowanetz M, Ferrara N. Vascular endothelial growth 2001;8:995–1004. nerve growth factor. Cell. 1991;65:189–197.
factor signaling pathways: therapeutic perspective. 237. Abella JV, et al. Met/Hepatocyte growth factor 258. Hempstead BL, Martin-Zanca D, Kaplan DR, Parada
Clin Cancer Res. 2006;12:5018–5022. receptor ubiquitination suppresses transformation LF, Chao MV. High-affinity NGF binding requires
215. Olsson AK, Dimberg A, Kreuger J, Claesson-Welsh and is required for Hrs phosphorylation. Mol Cell coexpression of the trk proto-oncogene and the
L. VEGF receptor signalling - in control of vascular Biol. 2005;25:9632–9645. low-affinity NGF receptor. Nature. 1991;350:678–
function. Nat Rev Mol Cell Biol. 2006;7:359–371. 238. Trusolino L, Bertotti A, Comoglio PM. MET 683.
216. Simons M, Gordon E, Claesson-Welsh L. Mecha- signalling: principles and functions in development, 259. Rolfo C, et al. Entrectinib: a potent new TRK,
nisms and regulation of endothelial VEGF receptor organ regeneration and cancer. Nat Rev Mol Cell ROS1, and ALK inhibitor. Expert Opin Investig
signalling. Nat Rev Mol Cell Biol. 2016;17:611– Biol. 2010;11:834–848. Drugs. 2015;24:1493–1500.
625. 239. Furge KA, Zhang YW, Vande Woude GF. Met 260. Brodeur GM, et al. Trk receptor expression and
217. Goel HL, Mercurio AM. VEGF targets the tumour receptor tyrosine kinase: enhanced signaling through inhibition in neuroblastomas. Clin Cancer Res.
cell. Nat Rev Cancer. 2013;13:871–882. adapter proteins. Oncogene. 2000;19:5582–5589. 2009;15:3244–3250.
218. Hurwitz H, et al. Bevacizumab plus irinotecan, 240. Gherardi E, Birchmeier W, Birchmeier C, Vande 261. Nakagawara A. Trk receptor tyrosine kinases: a bridge
fluorouracil, and leucovorin for metastatic colorectal Woude G. Targeting MET in cancer: rationale and between cancer and neural development. Cancer
cancer. N Engl J Med. 2004;350:2335–2342. progress. Nat Rev Cancer. 2012;12:89–103. Lett. 2001;169:107–114.
Intracellular Signaling  •  CHAPTER 2 46.e5
46.e5

262. Euhus DM, Timmons CF, Tomlinson GE. ETV6- 286. Kunishima N, et al. Structural basis of glutamate changes induced by vasopressin in isolated and
NTRK3—Trk-ing the primary event in human recognition by a dimeric metabotropic glutamate cultured hepatocytes. J Biol Chem. 1983;258:
secretory breast cancer. Cancer Cell. 2002;2:347–348. receptor. Nature. 2000;407:971–977. 5716–5725.
263. Davis JL, et  al. Infantile NTRK-associated 287. Muto T, Tsuchiya D, Morikawa K, Jingami H. 311. Siehler S. Regulation of RhoGEF proteins by
mesenchymal tumors. Pediatr Dev Pathol. 2017; Structures of the extracellular regions of the group G12/13-coupled receptors. Br J Pharmacol.
1093526617712639. II/III metabotropic glutamate receptors. Proc Natl 2009;158:41–49.
264. Nagasubramanian R, et al. Infantile fibrosarcoma Acad Sci USA. 2007;104:3759–3764. 312. Sjogren B, Blazer LL, Neubig RR. Regulators of
with NTRK3-ETV6 fusion successfully treated with 288. Dann CE, et al. Insights into Wnt binding and G protein signaling proteins as targets for drug
the tropomyosin-related kinase inhibitor LOXO-101. signalling from the structures of two Frizzled discovery. Prog Mol Biol Transl Sci. 2010;91:81–119.
Pediatr Blood Cancer. 2016;63:1468–1470. cysteine-rich domains. Nature. 2001;412:86–90. 313. Hurst JH, Hooks SB. Regulator of G-protein
265. Tabbo F, Pizzi M, Kyriakides PW, Ruggeri B, Inghi- 289. Adler E, et al. A novel family of mammalian taste signaling (RGS) proteins in cancer biology. Biochem
rami G. Oncogenic kinase fusions: an evolving arena receptors. Cell. 2000;100:693–702. Pharmacol. 2009;78:1289–1297.
with innovative clinical opportunities. Oncotarget. 290. Matsunami H, Montmayeur JP, Buck LB. A 314. Daaka Y, Luttrell LM, Lefkowitz RJ. Switching of
2016;7:25064–25086. family of candidate taste receptors in human and the coupling of the beta2-adrenergic receptor to
266. Stransky N, Cerami E, Schalm S, Kim JL, Lengauer mouse. Nature. 2000;404:601–604. different G proteins by protein kinase A. Nature.
C. The landscape of kinase fusions in cancer. Nat 291. Stevens RC, et al. The GPCR Network: a large-scale 1997;390:88–91.
Commun. 2014;5:4846. collaboration to determine human GPCR structure 315. Pitcher JA, Freedman NJ, Lefkowitz RJ. G protein-
267. Amatu A, Sartore-Bianchi A, Siena S. NTRK gene and function. Nat Rev Drug Discov. 2013;12:25–34. coupled receptor kinases. Annu Rev Biochem.
fusions as novel targets of cancer therapy across mul- 292. Scheerer P, et al. Crystal structure of opsin in 1998;67:653–692.
tiple tumour types. ESMO Open. 2016;1:e000023. its G-protein-interacting conformation. Nature. 316. Gurevich EV, Gurevich VV. Arrestins: ubiquitous
268. Drilon A, et al. A Next-generation TRK kinase 2008;455:497–502. regulators of cellular signaling pathways. Genome
inhibitor overcomes acquired resistance to prior 293. Rasmussen SG, et al. Crystal structure of the beta2 Biol. 2006;7:236.
TRK kinase inhibition in patients with TRK adrenergic receptor-Gs protein complex. Nature. 317. Shenoy SK, Lefkowitz RJ. beta-Arrestin-mediated
fusion-positive solid tumors. Cancer Discov. 2017. 2011;477:549–555. receptor trafficking and signal transduction. Trends
269. Doebele RC, et al. An oncogenic NTRK fusion in 294. Audet M, Bouvier M. Restructuring g-protein- Pharmacol Sci. 2011;32:521–533.
a patient with soft-tissue sarcoma with response to coupled receptor activation. Cell. 2012;151:14–23. 318. Luttrell LM, et al. Activation and targeting of extra-
the tropomyosin-related kinase inhibitor LOXO-101. 295. Cabrera-Vera TM, et al. Insights into G protein cellular signal-regulated kinases by beta-arrestin scaf-
Cancer Discov. 2015;5:1049–1057. structure, function, and regulation. Endocr Rev. folds. Proc Natl Acad Sci USA. 2001;98:2449–2454.
270. American Association for Cancer Research. TRK 2003;24:765–781. 319. DeFea KA, et al. Beta-arrestin-dependent endocytosis
inhibitor shows early promise. Cancer Discov. 2016;6: 296. Milligan G, Kostenis E. Heterotrimeric G-proteins: of proteinase-activated receptor 2 is required for
OF4. a short history. Br J Pharmacol. 2006;147(suppl intracellular targeting of activated ERK1/2. J Cell
271. Farago AF, et al. Durable clinical response to 1):S46–S55. Biol. 2000;148:1267–1281.
entrectinib in NTRK1-rearranged non-small cell 297. Oldham WM, Hamm HE. Heterotrimeric G protein 320. Young D, Waitches G, Birchmeier C, Fasano O,
lung cancer. J Thorac Oncol. 2015;10:1670–1674. activation by G-protein-coupled receptors. Nat Rev Wigler M. Isolation and characterization of a
272. Drilon A, et al. What hides behind the MASC: clini- Mol Cell Biol. 2008;9:60–71. new cellular oncogene encoding a protein with
cal response and acquired resistance to entrectinib 298. Rodbell M, Birnbaumer L, Pohl SL, Krans HM. The multiple potential transmembrane domains. Cell.
after ETV6-NTRK3 identification in a mammary glucagon-sensitive adenyl cyclase system in plasma 1986;45:711–719.
analogue secretory carcinoma (MASC). Ann Oncol. membranes of rat liver. V. An obligatory role of 321. Zohn IE, Symons M, Chrzanowska-Wodnicka M,
2016;27:920–926. guanylnucleotides in glucagon action. J Biol Chem. Westwick JK, Der CJ. Mas oncogene signaling
273. Hyman DM, et al. The efficacy of larotrectinib 1971;246:1877–1882. and transformation require the small GTP-binding
(LOXO-101), a selective tropomyosin receptor kinase 299. Hayes JS, Brunton LL, Mayer SE. Selective activation protein Rac. Mol Cell Biol. 1998;18:1225–1235.
(TRK) inhibitor, in adult and pediatric TRK fusion of particulate cAMP-dependent protein kinase by 322. O’Hayre M, et al. The emerging mutational land-
cancers. J Clin Oncol. 2017;35:LBA2501. isoproterenol and prostaglandin E1. J Biol Chem. scape of G proteins and G-protein-coupled receptors
274. Pierce KL, Premont RT, Lefkowitz RJ. Seven- 1980;255:5113–5119. in cancer. Nat Rev Cancer. 2013;13:412–424.
transmembrane receptors. Nat Rev Mol Cell Biol. 300. Denis C, Sauliere A, Galandrin S, Senard JM, Gales 323. Daub H, Wallasch C, Lankenau A, Herrlich A, Ullrich
2002;3:639–650. C. Probing heterotrimeric G protein activation: A. Signal characteristics of G protein-transactivated
275. Jacoby E, Bouhelal R, Gerspacher M, Seuwen K. applications to biased ligands. Curr Pharm Des. EGF receptor. EMBO J. 1997;16:7032–7044.
The 7 TM G-protein-coupled receptor target family. 2012;18:128–144. 324. Filardo EJ, Quinn JA, Bland KI, Frackelton AR
ChemMedChem. 2006;1:761–782. 301. Edwards HV, Christian F, Baillie GS. cAMP: novel Jr. Estrogen-induced activation of Erk-1 and Erk-2
276. Tyndall JD, Sandilya R. GPCR agonists and antago- concepts in compartmentalised signalling. Semin requires the G protein-coupled receptor homolog,
nists in the clinic. Med Chem. 2005;1:405–421. Cell Dev Biol. 2012;23:181–190. GPR30, and occurs via trans-activation of the
277. Dorsam RT, Gutkind JS. G-protein-coupled receptors 302. Metrich M, et al. Role of the cAMP-binding protein epidermal growth factor receptor through release
and cancer. Nat Rev Cancer. 2007;7:79–94. Epac in cardiovascular physiology and pathophysiol- of HB-EGF. Mol Endocrinol. 2000;14:1649–
278. Lappano R, Maggiolini M. G protein-coupled ogy. Pflugers Arch. 2010;459:535–546. 1660.
receptors: novel targets for drug discovery in cancer. 303. Kopperud R, Krakstad C, Selheim F, Doskeland SO. 325. Pierce KL, Luttrell LM, Lefkowitz RJ. New
Nat Rev Drug Discov. 2011;10:47–60. cAMP effector mechanisms. Novel twists for an ‘old’ mechanisms in heptahelical receptor signaling to
279. Spiegelberg BD, Hamm HE. Roles of G-protein- signaling system. FEBS Lett. 2003;546:121–126. mitogen activated protein kinase cascades. Oncogene.
coupled receptor signaling in cancer biology and gene 304. Walsh DA, Perkins JP, Krebs EG. An adenosine 2001;20:1532–1539.
transcription. Curr Opin Genet Dev. 2007;17:40–44. 3′,5′-monophosphate-dependent protein kinase 326. Hart S, et al. GPCR-induced migration of breast
280. Lagerstrom MC, Schioth HB. Structural diversity from rabbit skeletal muscle. J Biol Chem. 1968;243: carcinoma cells depends on both EGFR signal
of G protein-coupled receptors and significance 3763–3765. transactivation and EGFR-independent pathways.
for drug discovery. Nat Rev Drug Discov. 2008;7: 305. Altarejos JY, Montminy M. CREB and the CRTC Biol Chem. 2005;386:845–855.
339–357. co-activators: sensors for hormonal and metabolic 327. Boire A, et al. PAR1 is a matrix metalloprotease-1
281. Fredriksson R, Lagerstrom MC, Lundin LG, Schioth signals. Nat Rev Mol Cell Biol. 2011;12:141–151. receptor that promotes invasion and tumorigenesis
HB. The G-protein-coupled receptors in the human 306. Beene DL, Scott JD. A-kinase anchoring proteins of breast cancer cells. Cell. 2005;120:303–313.
genome form five main families. Phylogenetic take shape. Curr Opin Cell Biol. 2007;19:192–198. 328. Trivedi V, et al. Platelet matrix metalloprotease-1
analysis, paralogon groups, and fingerprints. Mol 307. Sunahara RK, Dessauer CW, Gilman AG. Complex- mediates thrombogenesis by activating PAR1 at a
Pharmacol. 2003;63:1256–1272. ity and diversity of mammalian adenylyl cyclases. cryptic ligand site. Cell. 2009;137:332–343.
282. Palczewski K, et al. Crystal structure of rhodopsin: Annu Rev Pharmacol Toxicol. 1996;36:461–480. 329. Sahai E, Marshall CJ. RHO-GTPases and cancer.
A G protein-coupled receptor. Science. 2000;289: 308. Birnbaumer L. Expansion of signal transduction by Nat Rev Cancer. 2002;2:133–142.
739–745. G proteins. The second 15 years or so: from 3 to 330. Mardilovich K, Olson MF, Baugh M. Targeting Rho
283. Gloriam DE, Fredriksson R, Schioth HB. The 16 alpha subunits plus betagamma dimers. Biochim GTPase signaling for cancer therapy. Future Oncol.
G protein-coupled receptor subset of the rat Biophys Acta. 2007;1768:772–793. 2012;8:165–177.
genome. BMC Genomics. 2007;8:338. 309. Streb H, Irvine RF, Berridge MJ, Schulz I. Release of 331. Morris JPT, Wang SC, Hebrok M. KRAS, Hedgehog,
284. Harmar AJ. Family-B G-protein-coupled receptors. Ca2+ from a nonmitochondrial intracellular store in Wnt and the twisted developmental biology of
Genome Biol. 2001;2:REVIEWS3013. pancreatic acinar cells by inositol-1,4,5-trisphosphate. pancreatic ductal adenocarcinoma. Nat Rev Cancer.
285. Bjarnadottir TK, et al. The human and mouse Nature. 1983;306:67–69. 2010;10:683–695.
repertoire of the adhesion family of G-protein- 310. Thomas AP, Marks JS, Coll KE, Williamson JR. 332. Takebe N, Harris PJ, Warren RQ, Ivy SP. Targeting
coupled receptors. Genomics. 2004;84:23–33. Quantitation and early kinetics of inositol lipid cancer stem cells by inhibiting Wnt, Notch, and
46.e6 Part I: Science and Clinical Oncology

Hedgehog pathways. Nat Rev Clin Oncol. 2011;8: 358. Barker N, Clevers H. Mining the Wnt pathway for 382. Morgan MJ, Liu ZG. Reactive oxygen species in
97–106. cancer therapeutics. Nat Rev Drug Discov. 2006;5: TNFalpha-induced signaling and cell death. Mol
333. Rubin LL, de Sauvage FJ. Targeting the Hedgehog 997–1014. Cells. 2010;30:1–12.
pathway in cancer. Nat Rev Drug Discov. 2006;5: 359. Dihlmann S, von Knebel Doeberitz M. Wnt/beta- 383. Lin A, Karin M. NF-kappaB in cancer: a marked
1026–1033. catenin-pathway as a molecular target for future target. Semin Cancer Biol. 2003;13:107–114.
334. Briscoe J, Therond PP. The mechanisms of Hedgehog anti-cancer therapeutics. Int J Cancer. 2005;113: 384. Coulthard LR, White DE, Jones DL, McDermott
signalling and its roles in development and disease. 515–524. MF, Burchill SA. p38(MAPK): stress responses from
Nat Rev Mol Cell Biol. 2013;14:416–429. 360. Smigel K. Arthritis drug approved for polyp preven- molecular mechanisms to therapeutics. Trends Mol
335. Kasai K, et al. The G12 family of heterotrimeric G tion blazes trail for other prevention trials. J Natl Med. 2009;15:369–379.
proteins and Rho GTPase mediate Sonic hedgehog Cancer Inst. 2000;92:297–299. 385. Wagner EF, Nebreda AR. Signal integration by JNK
signalling. Genes Cells. 2004;9:49–58. 361. Haque SJ, Sharma P. Interleukins and STAT signal- and p38 MAPK pathways in cancer development.
336. Riobo NA, Saucy B, Dilizio C, Manning DR. Activa- ing. Vitam Horm. 2006;74:165–206. Nat Rev Cancer. 2009;9:537–549.
tion of heterotrimeric G proteins by Smoothened. 362. Bazan JF. Structural design and molecular evolution 386. Sabapathy K. Role of the JNK pathway in human
Proc Natl Acad Sci USA. 2006;103:12607–12612. of a cytokine receptor superfamily. Proc Natl Acad diseases. Prog Mol Biol Transl Sci. 2012;106:145–
337. Hahn H, et al. Mutations of the human homolog of Sci USA. 1990;87:6934–6938. 169.
Drosophila patched in the nevoid basal cell carcinoma 363. Bazan JF. Shared architecture of hormone binding 387. Varfolomeev EE, Ashkenazi A. Tumor necrosis factor:
syndrome. Cell. 1996;85:841–851. domains in type I and II interferon receptors. Cell. an apoptosis JuNKie? Cell. 2004;116:491–497.
338. Xie J, et al. Activating Smoothened mutations in spo- 1990;61:753–754. 388. Wullaert A, Heyninck K, Beyaert R. Mechanisms
radic basal-cell carcinoma. Nature. 1998;391:90–92. 364. Kim HP, Imbert J, Leonard WJ. Both integrated of crosstalk between TNF-induced NF-kappaB and
339. Sweeney RT, et al. Identification of recurrent SMO and differential regulation of components of the JNK activation in hepatocytes. Biochem Pharmacol.
and BRAF mutations in ameloblastomas. Nat Genet. IL-2/IL-2 receptor system. Cytokine Growth Factor 2006;72:1090–1101.
2014;46:722–725. Rev. 2006;17:349–366. 389. Kuroki M, et al. Biological response modifiers
340. Thayer SP, et al. Hedgehog is an early and late 365. Liao W, Lin JX, Leonard WJ. IL-2 family cytokines: used in cancer biotherapy. Anticancer Res. 2012;32:
mediator of pancreatic cancer tumorigenesis. Nature. new insights into the complex roles of IL-2 as a 2229–2233.
2003;425:851–856. broad regulator of T helper cell differentiation. Curr 390. Deroose JP, et al. 20 years experience of TNF-based
341. Von Hoff DD, et al. Inhibition of the hedgehog Opin Immunol. 2011;23:598–604. isolated limb perfusion for in-transit melanoma
pathway in advanced basal-cell carcinoma. N Engl 366. Waldmann TA. The biology of interleukin-2 and metastases: TNF dose matters. Ann Surg Oncol.
J Med. 2009;361:1164–1172. interleukin-15: implications for cancer therapy and 2012;19:627–635.
342. Sekulic A, et al. Efficacy and safety of vismodegib vaccine design. Nat Rev Immunol. 2006;6:595–601. 391. Deroose JP, et al. Long-term results of tumor necrosis
in advanced basal-cell carcinoma. N Engl J Med. 367. Malek TR, Castro I. Interleukin-2 receptor signaling: factor alpha- and melphalan-based isolated limb
2012;366:2171–2179. at the interface between tolerance and immunity. perfusion in locally advanced extremity soft tissue
343. Casey D, et al. FDA approval summary: sonidegib Immunity. 2010;33:153–165. sarcomas. J Clin Oncol. 2011;29:4036–4044.
for locally advanced basal cell carcinoma. Clin Cancer 368. Gonzalez-Navajas JM, Lee J, David M, Raz E. 392. Deisseroth A, et al. U.S. Food and Drug Administra-
Res. 2017;23:2377–2381. Immunomodulatory functions of type I interferons. tion approval: ruxolitinib for the treatment of patients
344. Migden MR, et al. Treatment with two different Nat Rev Immunol. 2012;12:125–135. with intermediate and high-risk myelofibrosis. Clin
doses of sonidegib in patients with locally advanced 369. Platanias LC. Mechanisms of type-I- and type-II- Cancer Res. 2012;18:3212–3217.
or metastatic basal cell carcinoma (BOLT): a mul- interferon-mediated signalling. Nat Rev Immunol. 393. Mascarenhas J, Hoffman R. Ruxolitinib: the
ticentre, randomised, double-blind phase 2 trial. 2005;5:375–386. first FDA approved therapy for the treatment of
Lancet Oncol. 2015;16:716–728. 370. Pestka S, Krause CD, Walter MR. Interferons, myelofibrosis. Clin Cancer Res. 2012;18:3008–
345. Rimkus TK, Carpenter RL, Qasem S, Chan M, interferon-like cytokines, and their receptors. 3014.
Lo HW. Targeting the Sonic Hedgehog signaling Immunol Rev. 2004;202:8–32. 394. Verstovsek S, et al. A double-blind, placebo-
pathway: review of Smoothened and GLI inhibitors. 371. Gad HH, et al. Interferon-lambda is function- controlled trial of ruxolitinib for myelofibrosis. N
Cancers (Basel). 2016;8. ally an interferon but structurally related to the Engl J Med. 2012;366:799–807.
346. Guha M. Hedgehog inhibitor gets landmark skin interleukin-10 family. J Biol Chem. 2009;284:20869– 395. Yu H, Lee H, Herrmann A, Buettner R, Jove R.
cancer approval, but questions remain for wider 20875. Revisiting STAT3 signalling in cancer: new and
potential. Nat Rev Drug Discov. 2012;11:257–258. 372. Stark GR, Darnell JE Jr. The JAK-STAT pathway unexpected biological functions. Nat Rev Cancer.
347. Clevers H. Wnt/beta-catenin signaling in develop- at twenty. Immunity. 2012;36:503–514. 2014;14:736–746.
ment and disease. Cell. 2006;127:469–480. 373. Yu H, Pardoll D, Jove R. STATs in cancer inflam- 396. O’Shea JJ, Holland SM, Staudt LM. JAKs and STATs
348. Liu T, Liu X, Wang H, Moon RT, Malbon CC. mation and immunity: a leading role for STAT3. in immunity, immunodeficiency, and cancer. N Engl
Activation of rat frizzled-1 promotes Wnt signaling Nat Rev Cancer. 2009;9:798–809. J Med. 2013;368:161–170.
and differentiation of mouse F9 teratocarcinoma cells 374. O’Shea JJ, et al. The JAK-STAT pathway: impact on 397. Bubici C, Papa S. JNK signalling in cancer: in need
via pathways that require Galpha(q) and Galpha(o) human disease and therapeutic intervention. Annu of new, smarter therapeutic targets. Br J Pharmacol.
function. J Biol Chem. 1999;274:33539–33544. Rev Med. 2015;66:311–328. 2014;171:24–37.
349. Klaus A, Birchmeier W. Wnt signalling and its 375. Shuai K, Liu B. Regulation of JAK-STAT signal- 398. Hinck AP. Structural studies of the TGF-betas and
impact on development and cancer. Nat Rev Cancer. ling in the immune system. Nat Rev Immunol. their receptors - insights into evolution of the TGF-
2008;8:387–398. 2003;3:900–911. beta superfamily. FEBS Lett. 2012;586:1860–1870.
350. MacDonald BT, Tamai K, He X. Wnt/beta- 376. Yu H, Kortylewski M, Pardoll D. Crosstalk between 399. Shi Y, Massague J. Mechanisms of TGF-beta
catenin signaling: components, mechanisms, and cancer and immune cells: role of STAT3 in the signaling from cell membrane to the nucleus. Cell.
diseases. Dev Cell. 2009;17:9–26. tumour microenvironment. Nat Rev Immunol. 2003;113:685–700.
351. He TC, et al. Identification of c-MYC as a target of 2007;7:41–51. 400. Zi Z, Chapnick DA, Liu X. Dynamics of TGF-beta/
the APC pathway. Science. 1998;281:1509–1512. 377. Miyazaki T, et al. Three distinct IL-2 signaling Smad signaling. FEBS Lett. 2012;586:1921–1928.
352. Riese J, et al. LEF-1, a nuclear factor coordinating pathways mediated by bcl-2, c-myc, and lck 401. Annes JP, Munger JS, Rifkin DB. Making
signaling inputs from wingless and decapentaplegic. cooperate in hematopoietic cell proliferation. Cell. sense of latent TGFbeta activation. J Cell Sci.
Cell. 1997;88:777–787. 1995;81:223–231. 2003;116:217–224.
353. Lai SL, Chien AJ, Moon RT. Wnt/Fz signaling and 378. Aggarwal BB, Gupta SC, Kim JH. Historical 402. Rifkin DB. Latent transforming growth factor-beta
the cytoskeleton: potential roles in tumorigenesis. perspectives on tumor necrosis factor and its (TGF-beta) binding proteins: orchestrators of TGF-
Cell Res. 2009;19:532–545. superfamily: 25 years later, a golden journey. Blood. beta availability. J Biol Chem. 2005;280:7409–7412.
354. Anastas JN, Moon RT. WNT signalling pathways 2012;119:651–665. 403. Xu P, Liu J, Derynck R. Post-translational regulation
as therapeutic targets in cancer. Nat Rev Cancer. 379. Dempsey PW, Doyle SE, He JQ, Cheng G. The of TGF-beta receptor and Smad signaling. FEBS
2013;13:11–26. signaling adaptors and pathways activated by TNF Lett. 2012;586:1871–1884.
355. Behrens J, Lustig B. The Wnt connection to superfamily. Cytokine Growth Factor Rev. 2003;14: 404. Derynck R, Zhang YE. Smad-dependent and
tumorigenesis. Int J Dev Biol. 2004;48:477–487. 193–209. Smad-independent pathways in TGF-beta family
356. Ashton-Rickardt PG, et al. High frequency of APC 380. De Paepe B, Creus KK, De Bleecker JL. The signalling. Nature. 2003;425:577–584.
loss in sporadic colorectal carcinoma due to breaks tumor necrosis factor superfamily of cytokines in 405. Itoh S, ten Dijke P. Negative regulation of TGF-beta
clustered in 5q21-22. Oncogene. 1989;4:1169– the inflammatory myopathies: potential targets for receptor/Smad signal transduction. Curr Opin Cell
1174. therapy. Clin Dev Immunol. 2012;2012:369432. Biol. 2007;19:176–184.
357. Groden J, et al. Identification and characterization 381. Mahmood Z, Shukla Y. Death receptors: targets 406. Heldin CH, Vanlandewijck M, Moustakas A.
of the familial adenomatous polyposis coli gene. for cancer therapy. Exp Cell Res. 2010;316:887– Regulation of EMT by TGFbeta in cancer. FEBS
Cell. 1991;66:589–600. 899. Lett. 2012;586:1959–1970.
Intracellular Signaling  •  CHAPTER 2 46.e7
46.e7

407. Ikushima H, Miyazono K. TGFbeta signalling: a 433. Weng AP, et al. Activating mutations of NOTCH1 in cells is required for blood vessel maturation. J Clin
complex web in cancer progression. Nat Rev Cancer. human T cell acute lymphoblastic leukemia. Science. Invest. 2005;115:1542–1551.
2010;10:415–424. 2004;306:269–271. 456. Desgrosellier JS, Cheresh DA. Integrins in cancer:
408. Moustakas A, Heldin CH. The regulation of 434. Krop I, et al. Phase I Pharmacologic and pharma- biological implications and therapeutic opportuni-
TGFbeta signal transduction. Development. codynamic study of the gamma secretase (Notch) ties. Nat Rev Cancer. 2010;10:9–22.
2009;136:3699–3714. inhibitor MK-0752 in adult patients with advanced 457. Fournier AK, et al. Rac-dependent cyclin D1 gene
409. Javelaud D, Pierrat MJ, Mauviel A. Crosstalk between solid tumors. J Clin Oncol. 2012;30:2307–2313. expression regulated by cadherin- and integrin-
TGF-beta and hedgehog signaling in cancer. FEBS 435. Schott AF, et al. Preclinical and clinical studies of mediated adhesion. J Cell Sci. 2008;121:226–233.
Lett. 2012;586:2016–2025. gamma secretase inhibitors with docetaxel on human 458. Hsu MY, et al. Adenoviral gene transfer of beta3
410. Massague J. TGFbeta signalling in context. Nat Rev breast tumors. Clin Cancer Res. 2013;19:1512–1524. integrin subunit induces conversion from radial to
Mol Cell Biol. 2012;13:616–630. 436. Gavai AV, et al. Discovery of clinical candidate vertical growth phase in primary human melanoma.
411. Neuzillet C, et al. Targeting the TGFbeta pathway BMS-906024: a potent pan-Notch inhibitor for Am J Pathol. 1998;153:1435–1442.
for cancer therapy. Pharmacol Ther. 2015;147:22–31. the treatment of leukemia and solid tumors. ACS 459. Nip J, Shibata H, Loskutoff DJ, Cheresh DA,
412. Massague J. TGFbeta in Cancer. Cell. 2008;134: Med Chem Lett. 2015;6:523–527. Brodt P. Human melanoma cells derived from
215–230. 437. Yen WC, et al. Targeting Notch signaling with a lymphatic metastases use integrin alpha v beta 3
413. Markowitz S, et al. Inactivation of the type II TGF- Notch2/Notch3 antagonist (tarextumab) inhibits to adhere to lymph node vitronectin. J Clin Invest.
beta receptor in colon cancer cells with microsatellite tumor growth and decreases tumor-initiating cell 1992;90:1406–1413.
instability. Science. 1995;268:1336–1338. frequency. Clin Cancer Res. 2015;21:2084–2095. 460. Bello L, et al. Alpha(v)beta3 and alpha(v)beta5
414. Bornstein S, et al. Smad4 loss in mice causes 438. Kumar R, Thompson EB. The structure of the nuclear integrin expression in glioma periphery. Neurosurgery.
spontaneous head and neck cancer with increased hormone receptors. Steroids. 1999;64:310–319. 2001;49:380–389, discussion 390.
genomic instability and inflammation. J Clin Invest. 439. Mangelsdorf DJ, et al. The nuclear receptor 461. Diaz LK, et al. Beta4 integrin subunit gene expression
2009;119:3408–3419. superfamily: the second decade. Cell. 1995;83:835– correlates with tumor size and nuclear grade in early
415. Levy L, Hill CS. Alterations in components of the 839. breast cancer. Mod Pathol. 2005;18:1165–1175.
TGF-beta superfamily signaling pathways in human 440. Gronemeyer H, Gustafsson JA, Laudet V. Principles 462. Felding-Habermann B, et al. Integrin activation
cancer. Cytokine Growth Factor Rev. 2006;17:41–58. for modulation of the nuclear receptor superfamily. controls metastasis in human breast cancer. Proc
416. David CJ, et al. TGF-beta tumor suppression through Nat Rev Drug Discov. 2004;3:950–964. Natl Acad Sci USA. 2001;98:1853–1858.
a lethal EMT. Cell. 2016;164:1015–1030. 441. Shang Y, Hu X, DiRenzo J, Lazar MA, Brown M. 463. Friedrichs K, et al. High expression level of alpha
417. Yingling JM, Blanchard KL, Sawyer JS. Development Cofactor dynamics and sufficiency in estrogen recep- 6 integrin in human breast carcinoma is correlated
of TGF-beta signalling inhibitors for cancer therapy. tor-regulated transcription. Cell. 2000;103:843–852. with reduced survival. Cancer Res. 1995;55:901–906.
Nat Rev Drug Discov. 2004;3:1011–1022. 442. Toy W, et al. Activating ESR1 mutations differentially 464. Hersey P, et al. A phase II, randomized, open-label
418. Schlingensiepen KH, et al. Targeted tumor therapy affect the efficacy of ER antagonists. Cancer Discov. study evaluating the antitumor activity of MEDI-
with the TGF-beta 2 antisense compound AP 12009. 2017;7:277–287. 522, a humanized monoclonal antibody directed
Cytokine Growth Factor Rev. 2006;17:129–139. 443. Fribbens C, et al. Plasma ESR1 mutations and the against the human alpha v beta 3 (avb3) integrin,
419. Roberts AB, Wakefield LM. The two faces of treatment of estrogen receptor-positive advanced ± dacarbazine (DTIC) in patients with metastatic
transforming growth factor beta in carcinogenesis. breast cancer. J Clin Oncol. 2016;34:2961–2968. melanoma (MM). J Clin Oncol. 2005;23.
Proc Natl Acad Sci USA. 2003;100:8621–8623. 444. Wakeling AE. Steroid antagonists as nuclear receptor 465. Stupp R, et al. Cilengitide combined with stan-
420. Bierie B, Moses HL. Tumour microenvironment: blockers. Cancer Surv. 1992;14:71–85. dard treatment for patients with newly diagnosed
TGFbeta: the molecular Jekyll and Hyde of cancer. 445. Scher HI, et al. Increased survival with enzalutamide glioblastoma with methylated MGMT promoter
Nat Rev Cancer. 2006;6:506–520. in prostate cancer after chemotherapy. N Engl J Med. (CENTRIC EORTC 26071-22072 study): a
421. Takebe N, Nguyen D, Yang SX. Targeting notch 2012;367:1187–1197. multicentre, randomised, open-label, phase 3 trial.
signaling pathway in cancer: clinical develop- 446. Ryan CJ, et al. Abiraterone acetate plus prednisone Lancet Oncol. 2014;15:1100–1108.
ment advances and challenges. Pharmacol Ther. versus placebo plus prednisone in chemotherapy- 466. Yeatman TJ. A renaissance for SRC. Nat Rev Cancer.
2014;141:140–149. naive men with metastatic castration-resistant 2004;4:470–480.
422. Yin L, Velazquez OC, Liu ZJ. Notch signaling: prostate cancer (COU-AA-302): final overall 467. Thomas SM, Brugge JS. Cellular functions regulated
emerging molecular targets for cancer therapy. survival analysis of a randomised, double-blind, by Src family kinases. Annu Rev Cell Dev Biol.
Biochem Pharmacol. 2010;80:690–701. placebo-controlled phase 3 study. Lancet Oncol. 1997;13:513–609.
423. Ranganathan P, Weaver KL, Capobianco AJ. 2015;16:152–160. 468. Playford MP, Schaller MD. The interplay between
Notch signalling in solid tumours: a little bit of 447. Mitra SK, Schlaepfer DD. Integrin-regulated Src and integrins in normal and tumor biology.
everything but not all the time. Nat Rev Cancer. FAK-Src signaling in normal and cancer cells. Curr Oncogene. 2004;23:7928–7946.
2011;11:338–351. Opin Cell Biol. 2006;18:516–523. 469. Kim LC, Song L, Haura EB. Src kinases as
424. Roy M, Pear WS, Aster JC. The multifaceted 448. Assoian RK, Klein EA. Growth control by intracel- therapeutic targets for cancer. Nat Rev Clin Oncol.
role of Notch in cancer. Curr Opin Genet Dev. lular tension and extracellular stiffness. Trends Cell 2009;6:587–595.
2007;17:52–59. Biol. 2008;18:347–352. 470. Vogt PK. Retroviral oncogenes: a historical primer.
425. Blaumueller CM, Qi H, Zagouras P, Artavanis- 449. Pytela R, Pierschbacher MD, Ruoslahti E. Iden- Nat Rev Cancer. 2012;12:639–648.
Tsakonas S. Intracellular cleavage of Notch leads tification and isolation of a 140 kd cell surface 471. Martin GS. Rous sarcoma virus: a function required
to a heterodimeric receptor on the plasma membrane. glycoprotein with properties expected of a fibronectin for the maintenance of the transformed state. Nature.
Cell. 1997;90:281–291. receptor. Cell. 1985;40:191–198. 1970;227:1021–1023.
426. Shao H, Huang Q, Liu ZJ. Targeting notch signaling 450. Guo W, Giancotti FG. Integrin signalling during 472. Rous P. A sarcoma of the fowl transmissible by an
for cancer therapeutic intervention. Adv Pharmacol. tumour progression. Nat Rev Mol Cell Biol. agent separable from the tumor cells. J Exp Med.
2012;65:191–234. 2004;5:816–826. 1911;13:397–411.
427. Bray SJ. Notch signalling: a simple pathway becomes 451. Ley K, Rivera-Nieves J, Sandborn WJ, Shattil 473. Rubin H. Quantitative relations between causative
complex. Nat Rev Mol Cell Biol. 2006;7:678–689. S. Integrin-based therapeutics: biological basis, virus and cell in the Rous no. 1 chicken sarcoma.
428. Brou C, et al. A novel proteolytic cleavage involved clinical use and new drugs. Nat Rev Drug Discov. Virology. 1955;1:445–473.
in Notch signaling: the role of the disintegrin- 2016;15:173–183. 474. Stehelin D, Varmus HE, Bishop JM, Vogt PK. DNA
metalloprotease TACE. Mol Cell. 2000;5:207–216. 452. Legate KR, Wickstrom SA, Fassler R. Genetic and cell related to the transforming gene(s) of avian sarcoma
429. Mumm JS, et al. A ligand-induced extracellular biological analysis of integrin outside-in signaling. viruses is present in normal avian DNA. Nature.
cleavage regulates gamma-secretase-like proteolytic Genes Dev. 2009;23:397–418. 1976;260:170–173.
activation of Notch1. Mol Cell. 2000;5:197–206. 453. Winograd-Katz SE, Fassler R, Geiger B, Legate 475. Aleshin A, Finn RS. SRC: a century of science
430. Schroeter EH, Kisslinger JA, Kopan R. Notch-1 KR. The integrin adhesome: from genes and brought to the clinic. Neoplasia. 2010;12:599–607.
signalling requires ligand-induced proteolytic release proteins to human disease. Nat Rev Mol Cell Biol. 476. Cartwright CA, Eckhart W, Simon S, Kaplan PL.
of intracellular domain. Nature. 1998;393:382–386. 2014;15:273–288. Cell transformation by pp60c-src mutated in the
431. Aster JC, Blacklow SC, Pear WS. Notch signal- 454. Han S, Khuri FR, Roman J. Fibronectin stimulates carboxy-terminal regulatory domain. Cell. 1987;49:
ling in T-cell lymphoblastic leukaemia/lymphoma non-small cell lung carcinoma cell growth through 83–91.
and other haematological malignancies. J Pathol. activation of Akt/mammalian target of rapamycin/ 477. Yu H, et al. Solution structure of the SH3 domain
2011;223:262–273. S6 kinase and inactivation of LKB1/AMP-activated of Src and identification of its ligand-binding site.
432. Ellisen LW, et al. TAN-1, the human homolog of the protein kinase signal pathways. Cancer Res. 2006;66: Science. 1992;258:1665–1668.
Drosophila notch gene, is broken by chromosomal 315–323. 478. Sen B, Johnson FM. Regulation of SRC family kinases
translocations in T lymphoblastic neoplasms. Cell. 455. Garmy-Susini B, et al. Integrin alpha4beta1-VCAM- in human cancers. J Signal Transduct. 2011;2011:
1991;66:649–661. 1-mediated adhesion between endothelial and mural 865819.
46.e8 Part I: Science and Clinical Oncology

479. Xu W, Harrison SC, Eck MJ. Three-dimensional leukaemia to solid tumours. Nat Rev Cancer. 2013;13: 525. Gale NW, Kaplan S, Lowenstein EJ, Schlessinger
structure of the tyrosine kinase c-Src. Nature. 1997; 559–571. J, Bar-Sagi D. Grb2 mediates the EGF-dependent
385:595–602. 503. Ren R. Mechanisms of BCR-ABL in the pathogenesis activation of guanine nucleotide exchange on Ras.
480. Sakai R, et al. A novel signaling molecule, p130, of chronic myelogenous leukaemia. Nat Rev Cancer. Nature. 1993;363:88–92.
forms stable complexes in vivo with v-Crk and v-Src 2005;5:172–183. 526. Buday L, Downward J. Epidermal growth factor
in a tyrosine phosphorylation-dependent manner. 504. O’Hare T, Zabriskie MS, Eiring AM, Deininger regulates p21ras through the formation of a complex
EMBO J. 1994;13:3748–3756. MW. Pushing the limits of targeted therapy in of receptor, Grb2 adapter protein, and Sos nucleotide
481. Westhoff MA, Serrels B, Fincham VJ, Frame MC, chronic myeloid leukaemia. Nat Rev Cancer. exchange factor. Cell. 1993;73:611–620.
Carragher NO. SRC-mediated phosphorylation of 2012;12:513–526. 527. Egan SE, et al. Association of Sos Ras exchange
focal adhesion kinase couples actin and adhesion 505. Druker BJ, et al. Effects of a selective inhibitor of protein with Grb2 is implicated in tyrosine kinase
dynamics to survival signaling. Mol Cell Biol. 2004;24: the Abl tyrosine kinase on the growth of Bcr-Abl signal transduction and transformation. Nature.
8113–8133. positive cells. Nat Med. 1996;2:561–566. 1993;363:45–51.
482. Parsons JT, Parsons SJ. Src family protein tyrosine 506. Hantschel O, Grebien F, Superti-Furga G. The 528. Li N, et al. Guanine-nucleotide-releasing factor hSos1
kinases: cooperating with growth factor and adhesion growing arsenal of ATP-competitive and allosteric binds to Grb2 and links receptor tyrosine kinases
signaling pathways. Curr Opin Cell Biol. 1997;9: inhibitors of BCR-ABL. Cancer Res. 2012;72: to Ras signalling. Nature. 1993;363:85–88.
187–192. 4890–4895. 529. Rozakis-Adcock M, Fernley R, Wade J, Pawson T,
483. Frame MC. Src in cancer: deregulation and conse- 507. Ottmann O, et  al. Dasatinib induces rapid Bowtell D. The SH2 and SH3 domains of mam-
quences for cell behaviour. Biochim Biophys Acta. hematologic and cytogenetic responses in adult malian Grb2 couple the EGF receptor to the Ras
2002;1602:114–130. patients with Philadelphia chromosome positive activator mSos1. Nature. 1993;363:83–85.
484. Zhang S, Yu D. Targeting Src family kinases in acute lymphoblastic leukemia with resistance or 530. Donovan S, Shannon KM, Bollag G. GTPase
anti-cancer therapies: turning promise into triumph. intolerance to imatinib: interim results of a phase activating proteins: critical regulators of intracellular
Trends Pharmacol Sci. 2012;33:122–128. 2 study. Blood. 2007;110:2309–2315. signaling. Biochim Biophys Acta. 2002;1602:23–45.
485. Montero JC, Seoane S, Ocana A, Pandiella A. Inhibi- 508. Lilly MB, et al. Dasatinib 140 mg once daily versus 531. Wittinghofer A, Pai EF. The structure of Ras protein:
tion of SRC family kinases and receptor tyrosine 70 mg twice daily in patients with Ph-positive acute a model for a universal molecular switch. Trends
kinases by dasatinib: possible combinations in solid lymphoblastic leukemia who failed imatinib: Results Biochem Sci. 1991;16:382–387.
tumors. Clin Cancer Res. 2011;17:5546–5552. from a phase 3 study. Am J Hematol. 2010;85: 532. Ma J, Karplus M. Molecular switch in signal
486. Puls LN, Eadens M, Messersmith W. Current 164–170. transduction: reaction paths of the conformational
status of SRC inhibitors in solid tumor malignan- 509. Druker BJ, et al. Activity of a specific inhibitor of changes in ras p21. Proc Natl Acad Sci USA. 1997;94:
cies. Oncologist. 2011;16:566–578. the BCR-ABL tyrosine kinase in the blast crisis of 11905–11910.
487. Reynolds AB, et al. SRChing for the substrates of chronic myeloid leukemia and acute lymphoblastic 533. Chang L, Karin M. Mammalian MAP kinase
Src. Oncogene. 2014;33:4537–4547. leukemia with the Philadelphia chromosome. N Engl signalling cascades. Nature. 2001;410:37–40.
488. Masaki T, et al. pp60c-src activation in hepatocellular J Med. 2001;344:1038–1042. 534. Osborne JK, Zaganjor E, Cobb MH. Signal control
carcinoma of humans and LEC rats. Hepatology. 510. Ottmann OG, et al. A phase 2 study of imatinib through Raf: in sickness and in health. Cell Res.
1998;27:1257–1264. in patients with relapsed or refractory Philadelphia 2012;22:14–22.
489. Masaki T, et al. Reduced C-terminal Src kinase (Csk) chromosome-positive acute lymphoid leukemias. 535. Beeram M, Patnaik A, Rowinsky EK. Raf: a strategic
activities in hepatocellular carcinoma. Hepatology. Blood. 2002;100:1965–1971. target for therapeutic development against cancer.
1999;29:379–384. 511. Druker BJ, et al. Five-year follow-up of patients J Clin Oncol. 2005;23:6771–6790.
490. Cam WR, et al. Reduced C-terminal Src kinase activ- receiving imatinib for chronic myeloid leukemia. 536. Moodie SA, Willumsen BM, Weber MJ, Wolfman
ity is correlated inversely with pp60(c-src) activity N Engl J Med. 2006;355:2408–2417. A. Complexes of Ras.GTP with Raf-1 and mitogen-
in colorectal carcinoma. Cancer. 2001;92:61–70. 512. Zhang J, et al. Targeting Bcr-Abl by combining activated protein kinase kinase. Science. 1993;260:
491. Taniguchi K, et al. A gp130-Src-YAP module links allosteric with ATP-binding-site inhibitors. Nature. 1658–1661.
inflammation to epithelial regeneration. Nature. 2010;463:501–506. 537. Vojtek AB, Hollenberg SM, Cooper JA. Mammalian
2015;519:57–62. 513. Downward J. Targeting RAS signalling pathways Ras interacts directly with the serine/threonine kinase
492. Armaiz-Pena GN, et al. Src activation by beta- in cancer therapy. Nat Rev Cancer. 2003;3:11–22. Raf. Cell. 1993;74:205–214.
adrenoreceptors is a key switch for tumour metastasis. 514. Malumbres M, Barbacid M. RAS oncogenes: the 538. Warne PH, Viciana PR, Downward J. Direct
Nat Commun. 2013;4:1403. first 30 years. Nat Rev Cancer. 2003;3:459–465. interaction of Ras and the amino-terminal region
493. Lindauer M, Hochhaus A. Dasatinib. Recent Results 515. Harvey JJ. An unidentified virus which causes of Raf-1 in vitro. Nature. 1993;364:352–355.
Cancer Res. 2010;184:83–102. the rapid production of tumours in mice. Nature. 539. Zhang XF, et al. Normal and oncogenic p21ras
494. Lombardo LJ, et al. Discovery of N-(2-chloro- 1964;204:1104–1105. proteins bind to the amino-terminal regulatory
6-methyl-phenyl)-2-(6-(4-(2-hydroxyethyl)- 516. Kirsten WH, Mayer LA. Morphologic responses to domain of c-Raf-1. Nature. 1993;364:308–313.
piperazin-1-yl)-2-methylpyrimidin-4-ylamino) a murine erythroblastosis virus. J Natl Cancer Inst. 540. Avruch J, et al. Ras activation of the Raf kinase:
thiazole-5-carboxamide (BMS-354825), a dual Src/ 1967;39:311–335. tyrosine kinase recruitment of the MAP kinase
Abl kinase inhibitor with potent antitumor activity in 517. Shih TY, Papageorge AG, Stokes PE, Weeks MO, cascade. Recent Prog Horm Res. 2001;56:127–155.
preclinical assays. J Med Chem. 2004;47:6658–6661. Scolnick EM. Guanine nucleotide-binding and 541. Marais R, Light Y, Paterson HF, Mason CS, Marshall
495. Chang Q, Jorgensen C, Pawson T, Hedley DW. autophosphorylating activities associated with the CJ. Differential regulation of Raf-1, A-Raf, and
Effects of dasatinib on EphA2 receptor tyrosine p21src protein of Harvey murine sarcoma virus. B-Raf by oncogenic ras and tyrosine kinases. J Biol
kinase activity and downstream signalling in Nature. 1980;287:686–691. Chem. 1997;272:4378–4383.
pancreatic cancer. Br J Cancer. 2008;99:1074–1082. 518. Karnoub AE, Weinberg RA. Ras oncogenes: split 542. Kubicek M, et al. Dephosphorylation of Ser-259
496. Goff SP, Gilboa E, Witte ON, Baltimore D. Structure personalities. Nat Rev Mol Cell Biol. 2008;9:517–531. regulates Raf-1 membrane association. J Biol Chem.
of the Abelson murine leukemia virus genome and 519. Lacal PM, Pennington CY, Lacal JC. Transforming 2002;277:7913–7919.
the homologous cellular gene: studies with cloned activity of ras proteins translocated to the plasma 543. Zimmermann S, Moelling K. Phosphorylation and
viral DNA. Cell. 1980;22:777–785. membrane by a myristoylation sequence from the regulation of Raf by Akt (protein kinase B). Science.
497. Hantschel O, et al. A myristoyl/phosphotyrosine src gene product. Oncogene. 1988;2:533–537. 1999;286:1741–1744.
switch regulates c-Abl. Cell. 2003;112:845–857. 520. Mor A, Philips MR. Compartmentalized Ras/MAPK 544. Dhillon AS, et al. A Raf-1 mutant that dissociates
498. Schlatterer SD, Acker CM, Davies P. c-Abl signaling. Annu Rev Immunol. 2006;24:771–800. MEK/extracellular signal-regulated kinase activation
in neurodegenerative disease. J Mol Neurosci. 521. Willumsen BM, Christensen A, Hubbert NL, from malignant transformation and differentiation
2011;45:445–452. Papageorge AG, Lowy DR. The p21 ras C-terminus but not proliferation. Mol Cell Biol. 2003;23:
499. Daniel R, Cai Y, Wong PM, Chung SW. Deregula- is required for transformation and membrane associa- 1983–1993.
tion of c-abl mediated cell growth after retroviral tion. Nature. 1984;310:583–586. 545. Dhillon AS, Meikle S, Yazici Z, Eulitz M, Kolch
transfer and expression of antisense sequences. 522. Mitin N, Rossman KL, Der CJ. Signaling W. Regulation of Raf-1 activation and signalling
Oncogene. 1995;10:1607–1614. interplay in Ras superfamily function. Curr Biol. by dephosphorylation. EMBO J. 2002;21:64–
500. Yuan ZM, et al. Regulation of Rad51 function by 2005;15:R563–R574. 71.
c-Abl in response to DNA damage. J Biol Chem. 523. Vigil D, Cherfils J, Rossman KL, Der CJ. Ras 546. Zhang BH, et al. Serum- and glucocorticoid-inducible
1998;273:3799–3802. superfamily GEFs and GAPs: validated and trac- kinase SGK phosphorylates and negatively regulates
501. Sirvent A, Benistant C, Roche S. Cytoplasmic table targets for cancer therapy? Nat Rev Cancer. B-Raf. J Biol Chem. 2001;276:31620–31626.
signalling by the c-Abl tyrosine kinase in normal 2010;10:842–857. 547. Morrison DK, Heidecker G, Rapp UR,
and cancer cells. Biol Cell. 2008;100:617–631. 524. Bourne HR, Sanders DA, McCormick F. The Copeland TD. Identification of the major phos-
502. Greuber EK, Smith-Pearson P, Wang J, Pendergast GTPase superfamily: a conserved switch for diverse phorylation sites of the Raf-1 kinase. J Biol Chem.
AM. Role of ABL family kinases in cancer: from cell functions. Nature. 1990;348:125–132. 1993;268:17309–17316.
Intracellular Signaling  •  CHAPTER 2 46.e9
46.e9

548. Diaz B, et al. Phosphorylation of Raf-1 serine 573. Sjolander A, Yamamoto K, Huber BE, Lapetina 596. Brenan L, et al. Phenotypic characterization of
338-serine 339 is an essential regulatory event for EG. Association of p21ras with phosphatidylino- a comprehensive set of MAPK1/ERK2 missense
Ras-dependent activation and biological signaling. sitol 3-kinase. Proc Natl Acad Sci USA. 1991;88: mutants. Cell Rep. 2016;17:1171–1183.
Mol Cell Biol. 1997;17:4509–4516. 7908–7912. 597. Montagut C, Settleman J. Targeting the RAF-
549. King AJ, et al. The protein kinase Pak3 positively 574. Gille H, Downward J. Multiple ras effector pathways MEK-ERK pathway in cancer therapy. Cancer Lett.
regulates Raf-1 activity through phosphorylation of contribute to G(1) cell cycle progression. J Biol 2009;283:125–134.
serine 338. Nature. 1998;396:180–183. Chem. 1999;274:22033–22040. 598. Santarpia L, Lippman SM, El-Naggar AK. Targeting
550. Fabian JR, Daar IO, Morrison DK. Critical tyrosine 575. Rodriguez-Viciana P, et al. Role of phosphoinositide the MAPK-RAS-RAF signaling pathway in cancer
residues regulate the enzymatic and biological activity 3-OH kinase in cell transformation and control of the therapy. Expert Opin Ther Targets. 2012;16:103–119.
of Raf-1 kinase. Mol Cell Biol. 1993;13:7170–7179. actin cytoskeleton by Ras. Cell. 1997;89:457–467. 599. Rao S, et al. Phase III double-blind placebo-
551. Marais R, Light Y, Paterson HF, Marshall CJ. 576. Hofer F, Fields S, Schneider C, Martin GS. Activated controlled study of farnesyl transferase inhibitor
Ras recruits Raf-1 to the plasma membrane for Ras interacts with the Ral guanine nucleotide R115777 in patients with refractory advanced
activation by tyrosine phosphorylation. EMBO J. dissociation stimulator. Proc Natl Acad Sci USA. colorectal cancer. J Clin Oncol. 2004;22:3950–3957.
1995;14:3136–3145. 1994;91:11089–11093. 600. Rowell CA, Kowalczyk JJ, Lewis MD, Garcia
552. Mason CS, et al. Serine and tyrosine phosphoryla- 577. Kikuchi A, Demo SD, Ye ZH, Chen YW, Williams AM. Direct demonstration of geranylgeranylation
tions cooperate in Raf-1, but not B-Raf activation. LT. ralGDS family members interact with the effector and farnesylation of Ki-Ras in vivo. J Biol Chem.
EMBO J. 1999;18:2137–2148. loop of ras. Mol Cell Biol. 1994;14(7483–7491):p21. 1997;272:14093–14097.
553. Chong H, Lee J, Guan KL. Positive and negative 578. Spaargaren M, Bischoff JR. Identification of 601. Whyte DB, et al. K- and N-Ras are geranylgeranyl-
regulation of Raf kinase activity and function by the guanine nucleotide dissociation stimulator ated in cells treated with farnesyl protein transferase
phosphorylation. EMBO J. 2001;20:3716–3727. for Ral as a putative effector molecule of R-ras, inhibitors. J Biol Chem. 1997;272:14459–14464.
554. Zhang BH, Guan KL. Activation of B-Raf kinase H-ras, K-ras, and Rap. Proc Natl Acad Sci USA. 602. Ostrem JM, Peters U, Sos ML, Wells JA, Shokat
requires phosphorylation of the conserved residues 1994;91:12609–12613. KM. K-Ras(G12C) inhibitors allosterically control
Thr598 and Ser601. EMBO J. 2000;19:5429–5439. 579. Schubbert S, Shannon K, Bollag G. Hyperactive GTP affinity and effector interactions. Nature. 2013;
555. Dent P, et al. Activation of mitogen-activated protein Ras in developmental disorders and cancer. Nat Rev 503:548–551.
kinase kinase by v-Raf in NIH 3T3 cells and in Cancer. 2007;7:295–308. 603. Flaherty KT, et al. Inhibition of mutated, activated
vitro. Science. 1992;257:1404–1407. 580. Vakiani E, Solit DB. KRAS and BRAF: drug targets BRAF in metastatic melanoma. N Engl J Med.
556. Khokhlatchev AV, et al. Phosphorylation of the MAP and predictive biomarkers. J Pathol. 2011;223: 2010;363:809–819.
kinase ERK2 promotes its homodimerization and 219–229. 604. McArthur GA, et al. Safety and efficacy of vemu-
nuclear translocation. Cell. 1998;93:605–615. 581. Pylayeva-Gupta Y, Grabocka E, Bar-Sagi D. RAS rafenib in BRAF(V600E) and BRAF(V600K)
557. Yoon S, Seger R. The extracellular signal-regulated oncogenes: weaving a tumorigenic web. Nat Rev mutation-positive melanoma (BRIM-3): extended
kinase: multiple substrates regulate diverse cellular Cancer. 2011;11:761–774. follow-up of a phase 3, randomised, open-label study.
functions. Growth Factors. 2006;24:21–44. 582. Malumbres M, Pellicer A. RAS pathways to cell Lancet Oncol. 2014;15:323–332.
558. Robinson MJ, Cobb MH. Mitogen-activated protein cycle control and cell transformation. Front Biosci. 605. Robert C, et al. Improved overall survival in mela-
kinase pathways. Curr Opin Cell Biol. 1997;9: 1998;3:d887–d912. noma with combined dabrafenib and trametinib.
180–186. 583. Feig LA, Cooper GM. Relationship among guanine N Engl J Med. 2015;372:30–39.
559. Pratilas CA, Solit DB. Therapeutic strategies for nucleotide exchange, GTP hydrolysis, and transform- 606. Long GV, et al. Dabrafenib in patients with
targeting BRAF in human cancer. Rev Recent Clin ing potential of mutated ras proteins. Mol Cell Biol. Val600Glu or Val600Lys BRAF-mutant melanoma
Trials. 2007;2:121–134. 1988;8:2472–2478. metastatic to the brain (BREAK-MB): a multi-
560. Pratilas CA, Solit DB. Targeting the mitogen- 584. Chen SY, Huff SY, Lai CC, Der CJ, Powers S. centre, open-label, phase 2 trial. Lancet Oncol.
activated protein kinase pathway: physiological feed- Ras-15A protein shares highly similar dominant- 2012;13:1087–1095.
back and drug response. Clin Cancer Res. 2010;16: negative biological properties with Ras-17N 607. Ascierto PA, et al. Phase II trial (BREAK-2) of
3329–3334. and forms a stable, guanine-nucleotide resistant the BRAF inhibitor dabrafenib (GSK2118436) in
561. Kim HJ, Bar-Sagi D. Modulation of signalling by complex with CDC25 exchange factor. Oncogene. patients with metastatic melanoma. J Clin Oncol.
Sprouty: a developing story. Nat Rev Mol Cell Biol. 1994;9:2691–2698. 2013;31:3205–3211.
2004;5:441–450. 585. Cancer Genome Atlas Research Network. Compre- 608. McGettigan S. Dabrafenib: A new therapy for use
562. Sasaki A, et al. Mammalian Sprouty4 suppresses hensive genomic characterization defines human glio- in BRAF-mutated metastatic melanoma. J Adv Pract
Ras-independent ERK activation by binding to Raf1. blastoma genes and core pathways. Nature. 2008;455: Oncol. 2014;5:211–215.
Nat Cell Biol. 2003;5:427–432. 1061–1068. 609. Falchook GS, et al. Dabrafenib in patients with
563. Hanafusa H, Torii S, Yasunaga T, Nishida E. 586. Cancer Genome Atlas Network. Genomic clas- melanoma, untreated brain metastases, and other
Sprouty1 and Sprouty2 provide a control mechanism sification of cutaneous melanoma. Cell. 2015;161: solid tumours: a phase 1 dose-escalation trial. Lancet.
for the Ras/MAPK signalling pathway. Nat Cell Biol. 1681–1696. 2012;379:1893–1901.
2002;4:850–858. 587. Nissan MH, et al. Loss of NF1 in cutaneous 610. Hauschild A, et al. Dabrafenib in BRAF-mutated
564. Wakioka T, et al. Spred is a Sprouty-related suppres- melanoma is associated with RAS activation and metastatic melanoma: a multicentre, open-label, phase
sor of Ras signalling. Nature. 2001;412:647–651. MEK dependence. Cancer Res. 2014;74:2340–2350. 3 randomised controlled trial. Lancet. 2012;380:
565. Owens DM, Keyse SM. Differential regulation of 588. Imielinski M, et al. Oncogenic and sorafenib-sensitive 358–365.
MAP kinase signalling by dual-specificity protein ARAF mutations in lung adenocarcinoma. J Clin 611. Sosman JA, et al. Survival in BRAF V600-mutant
phosphatases. Oncogene. 2007;26:3203–3213. Invest. 2014;124:1582–1586. advanced melanoma treated with vemurafenib.
566. Keyse SM. Dual-specificity MAP kinase phospha- 589. Diamond EL, et al. Diverse and targetable kinase N Engl J Med. 2012;366:707–714.
tases (MKPs) and cancer. Cancer Metastasis Rev. alterations drive histiocytic neoplasms. Cancer Discov. 612. Chapman PB, et al. Improved survival with vemu-
2008;27:253–261. 2016;6:154–165. rafenib in melanoma with BRAF V600E mutation.
567. Ritt DA, Monson DM, Specht SI, Morrison DK. 590. Nelson DS, et  al. Somatic activating ARAF N Engl J Med. 2011;364:2507–2516.
Impact of feedback phosphorylation and Raf mutations in Langerhans cell histiocytosis. Blood. 613. Joseph EW, et al. The RAF inhibitor PLX4032
heterodimerization on normal and mutant B-Raf 2014;123:3152–3155. inhibits ERK signaling and tumor cell proliferation
signaling. Mol Cell Biol. 2010;30:806–819. 591. Tiacci E, et al. BRAF mutations in hairy-cell in a V600E BRAF-selective manner. Proc Natl Acad
568. Dougherty MK, et al. Regulation of Raf-1 by direct leukemia. N Engl J Med. 2011;364:2305–2315. Sci USA. 2010;107:14903–14908.
feedback phosphorylation. Mol Cell. 2005;17: 592. Lito P, Rosen N, Solit DB. Tumor adaptation 614. Solit DB, Rosen N. Towards a unified model of
215–224. and resistance to RAF inhibitors. Nat Med. RAF inhibitor resistance. Cancer Discov. 2014;4:27–
569. Matheny SA, et al. Ras regulates assembly of 2013;19:1401–1409. 30.
mitogenic signalling complexes through the effector 593. Yao Z, et al. BRAF mutants evade ERK-dependent 615. Wilhelm S, et al. Discovery and development of
protein IMP. Nature. 2004;427:256–260. feedback by different mechanisms that determine sorafenib: a multikinase inhibitor for treating cancer.
570. Therrien M, et al. KSR, a novel protein kinase their sensitivity to pharmacologic inhibition. Cancer Nat Rev Drug Discov. 2006;5:835–844.
required for RAS signal transduction. Cell. 1995;83: Cell. 2015;28:370–383. 616. Infante JR, et al. Safety and efficacy results from
879–888. 594. Arcila ME, et al. MAP2K1 (MEK1) mutations define the first-time-in-human study of the oral MEK
571. Yeung K, et al. Suppression of Raf-1 kinase activ- a distinct subset of lung adenocarcinoma associated 1/2 inhibitor GSK1120212. Journal of Clinical
ity and MAP kinase signalling by RKIP. Nature. with smoking. Clin Cancer Res. 2015;21:1935– Oncology, 2010 ASCO Annual Meeting Proceedings;
1999;401:173–177. 1943. 2010:28.
572. Rodriguez-Viciana P, et al. Phosphatidylinositol-3-OH 595. Van Allen EM, et al. The genetic landscape of clinical 617. Flaherty KT, et al. Improved survival with MEK
kinase as a direct target of Ras. Nature. 1994;370: resistance to RAF inhibition in metastatic melanoma. inhibition in BRAF-mutated melanoma. N Engl J
527–532. Cancer Discov. 2014;4:94–109. Med. 2012;367:107–114.
46.e10 Part I: Science and Clinical Oncology

618. Wright CJ, McCormack PL. Trametinib: first global in BRAF(V600E) melanoma. Sci Transl Med. 2015;7: 661. Cardone MH, et al. Regulation of cell death
approval. Drugs. 2013;73:1245–1254. 279ra241. protease caspase-9 by phosphorylation. Science.
619. Long GV, et al. Combined BRAF and MEK inhibi- 640. Loi S, et al. RAS/MAPK activation is associated 1998;282:1318–1321.
tion versus BRAF inhibition alone in melanoma. with reduced tumor-infiltrating lymphocytes in 662. Brunet A, et al. Akt promotes cell survival by phos-
N Engl J Med. 2014;371:1877–1888. triple-negative breast cancer: therapeutic cooperation phorylating and inhibiting a Forkhead transcription
620. Larkin J, et  al. Combined vemurafenib and between MEK and PD-1/PD-L1 immune checkpoint factor. Cell. 1999;96:857–868.
cobimetinib in BRAF-mutated melanoma. N Engl inhibitors. Clin Cancer Res. 2016;22:1499–1509. 663. Romashkova JA, Makarov SS. NF-kappaB is a target
J Med. 2014;371:1867–1876. 641. Barretina J, et al. Subtype-specific genomic alterations of AKT in anti-apoptotic PDGF signalling. Nature.
621. Flaherty KT, et al. Combined BRAF and MEK define new targets for soft-tissue sarcoma therapy. 1999;401:86–90.
Inhibition in Melanoma with BRAF V600 Muta- Nat Genet. 2010;42:715–721. 664. Kane LP, Shapiro VS, Stokoe D, Weiss A. Induction
tions. N Engl J Med. 2012. 642. Schwartz GK, et al. Phase I study of PD 0332991, of NF-kappaB by the Akt/PKB kinase. Curr Biol.
622. Johnson DB, et al. Combined BRAF (dabrafenib) a cyclin-dependent kinase inhibitor, administered 1999;9:601–604.
and MEK inhibition (trametinib) in patients with in 3-week cycles (Schedule 2/1). Br J Cancer. 665. Mayo LD, Donner DB. A phosphatidylinositol
BRAFV600-mutant melanoma experiencing progres- 2011;104:1862–1868. 3-kinase/Akt pathway promotes translocation of
sion with single-agent BRAF inhibitor. J Clin Oncol. 643. Dickson MA, et al. Phase II trial of the CDK4 Mdm2 from the cytoplasm to the nucleus. Proc
2014;32:3697–3704. inhibitor PD0332991 in patients with advanced Natl Acad Sci USA. 2001;98:11598–11603.
623. Ascierto PA, et al. MEK162 for patients with CDK4-amplified well-differentiated or dedifferen- 666. Han J, et al. Role of substrates and products of
advanced melanoma harbouring NRAS or Val600 tiated liposarcoma. J Clin Oncol. 2013;31:2024– PI 3-kinase in regulating activation of Rac-related
BRAF mutations: a non-randomised, open-label 2028. guanosine triphosphatases by Vav. Science. 1998;279:
phase 2 study. Lancet Oncol. 2013;14:249–256. 644. Beaver JA, et al. FDA Approval: Palbociclib for the 558–560.
624. Bendell JC, et al. Clinical activity and safety of treatment of postmenopausal patients with estrogen 667. Brunet A, et al. Protein kinase SGK mediates
cobimetinib (cobi) and atezolizumab in colorectal receptor-positive, HER2-negative metastatic breast survival signals by phosphorylating the forkhead
cancer (CRC). J Clin Oncol. 2016;34:3502. cancer. Clin Cancer Res. 2015;21:4760–4766. transcription factor FKHRL1 (FOXO3a). Mol Cell
625. Ho AL, et al. Selumetinib-enhanced radioiodine 645. Thorpe LM, Yuzugullu H, Zhao JJ. PI3K in cancer: Biol. 2001;21:952–965.
uptake in advanced thyroid cancer. N Engl J Med. divergent roles of isoforms, modes of activation and 668. Zoncu R, Efeyan A, Sabatini DM. mTOR: from
2013;368:623–632. therapeutic targeting. Nat Rev Cancer. 2015;15:7–24. growth signal integration to cancer, diabetes and
626. Ameratunga M, McArthur G, Gan H, Cher L. 646. Vanhaesebroeck B, Stephens L, Hawkins P. PI3K ageing. Nat Rev Mol Cell Biol. 2011;12:21–35.
Prolonged disease control with MEK inhibitor in signalling: the path to discovery and understanding. 669. Sabatini DM. mTOR and cancer: insights into
neurofibromatosis type I-associated glioblastoma. Nat Rev Mol Cell Biol. 2012;13:195–203. a complex relationship. Nat Rev Cancer. 2006;6:
J Clin Pharm Ther. 2016;41:357–359. 647. Hiles ID, et al. Phosphatidylinositol 3-kinase: 729–734.
627. Woodfield SE, Zhang L, Scorsone KA, Liu Y, Zage structure and expression of the 110 kd catalytic 670. Loewith R, et al. Two TOR complexes, only one
PE. Binimetinib inhibits MEK and is effective against subunit. Cell. 1992;70:419–429. of which is rapamycin sensitive, have distinct roles
neuroblastoma tumor cells with low NF1 expression. 648. Hennessy BT, Smith DL, Ram PT, Lu Y, Mills in cell growth control. Mol Cell. 2002;10:457–468.
BMC Cancer. 2016;16:172. GB. Exploiting the PI3K/AKT pathway for cancer 671. Laplante M, Sabatini DM. mTOR signaling in
628. Tao Z, et al. Coadministration of trametinib and drug discovery. Nat Rev Drug Discov. 2005;4:988– growth control and disease. Cell. 2012;149:274–293.
palbociclib radiosensitizes KRAS-mutant non-small 1004. 672. Gao X, et al. Tsc tumour suppressor proteins
cell lung cancers in vitro and in vivo. Clin Cancer 649. Okkenhaug K, Vanhaesebroeck B. PI3K in lympho- antagonize amino-acid-TOR signalling. Nat Cell
Res. 2016;22:122–133. cyte development, differentiation and activation. Biol. 2002;4:699–704.
629. Grisham RN, et al. Extreme outlier analysis identifies Nat Rev Immunol. 2003;3:317–330. 673. Zhang Y, et al. Rheb is a direct target of the tuberous
occult mitogen-activated protein kinase pathway 650. Vivanco I, Sawyers CL. The phosphatidylinositol sclerosis tumour suppressor proteins. Nat Cell Biol.
mutations in patients with low-grade serous ovarian 3-kinase AKT pathway in human cancer. Nat Rev 2003;5:578–581.
cancer. J Clin Oncol. 2015;33:4099–4105. Cancer. 2002;2:489–501. 674. Faubert B, Vincent EE, Poffenberger MC, Jones
630. Shi H, et al. Preexisting MEK1 exon 3 mutations in 651. Maehama T, Dixon JE. The tumor suppressor, RG. The AMP-activated protein kinase (AMPK)
V600E/KBRAF melanomas do not confer resistance PTEN/MMAC1, dephosphorylates the lipid second and cancer: many faces of a metabolic regulator.
to BRAF inhibitors. Cancer Discov. 2012;2:414–424. messenger, phosphatidylinositol 3,4,5-trisphosphate. Cancer Lett. 2015;356:165–170.
631. Grbovic OM, et al. V600E B-Raf requires the J Biol Chem. 1998;273:13375–13378. 675. Shaw RJ. LKB1 and AMP-activated protein kinase
Hsp90 chaperone for stability and is degraded in 652. Alessi DR, et  al. Characterization of a 3- control of mTOR signalling and growth. Acta Physiol
response to Hsp90 inhibitors. Proc Natl Acad Sci phosphoinositide-dependent protein kinase which (Oxf ). 2009;196:65–80.
USA. 2006;103:57–62. phosphorylates and activates protein kinase Balpha. 676. Hay N, Sonenberg N. Upstream and downstream
632. Herrero A, et al. Small molecule inhibition of ERK Curr Biol. 1997;7:261–269. of mTOR. Genes Dev. 2004;18:1926–1945.
dimerization prevents tumorigenesis by RAS-ERK 653. Currie RA, et al. Role of phosphatidylinositol 677. Harrington LS, et al. The TSC1-2 tumor suppressor
pathway oncogenes. Cancer Cell. 2015;28:170– 3,4,5-trisphosphate in regulating the activity and controls insulin-PI3K signaling via regulation of IRS
182. localization of 3-phosphoinositide-dependent protein proteins. J Cell Biol. 2004;166:213–223.
633. Morris EJ, et al. Discovery of a novel ERK inhibi- kinase-1. Biochem J. 1999;337(Pt 3):575–583. 678. Courtney KD, Corcoran RB, Engelman JA. The
tor with activity in models of acquired resistance 654. Stokoe D, et al. Dual role of phosphatidylinosi- PI3K pathway as drug target in human cancer.
to BRAF and MEK inhibitors. Cancer Discov. tol-3,4,5-trisphosphate in the activation of protein J Clin Oncol. 2010;28:1075–1083.
2013;3:742–750. kinase B. Science. 1997;277:567–570. 679. Mellinghoff IK, et al. Molecular determinants of the
634. Germann U, et al. Abstract 4693: The selective ERK 655. Bellacosa A, et al. Akt activation by growth factors is response of glioblastomas to EGFR kinase inhibitors.
inhibitor BVD-523 is active in models of MAPK a multiple-step process: the role of the PH domain. N Engl J Med. 2005;353:2012–2024.
pathway-dependent cancers, including those with Oncogene. 1998;17:313–325. 680. Almoguera C, et al. Most human carcinomas of the
intrinsic and acquired drug resistance. Cancer Res. 656. Diehl JA, Cheng M, Roussel MF, Sherr CJ. exocrine pancreas contain mutant c-K-ras genes.
2015;75:4693. Glycogen synthase kinase-3beta regulates cyclin Cell. 1988;53:549–554.
635. Li BT, et al. First-in-class oral ERK1/2 inhibitor D1 proteolysis and subcellular localization. Genes 681. Rodriguez-Viciana P, Warne PH, Vanhaesebroeck B,
Ulixertinib (BVD-523) in patients with advanced Dev. 1998;12:3499–3511. Waterfield MD, Downward J. Activation of phos-
solid tumors: final results of a phase I dose escalation 657. Medema RH, Kops GJ, Bos JL, Burgering BM. phoinositide 3-kinase by interaction with Ras and
and expansion study. J Clin Oncol. 2017;35:2508. AFX-like Forkhead transcription factors mediate by point mutation. EMBO J. 1996;15:2442–2451.
636. Dankort D, et al. Braf(V600E) cooperates with Pten cell-cycle regulation by Ras and PKB through 682. Gupta S, et al. Binding of ras to phosphoinositide
loss to induce metastatic melanoma. Nat Genet. p27kip1. Nature. 2000;404:782–787. 3-kinase p110alpha is required for ras-driven
2009;41:544–552. 658. Lawlor MA, Rotwein P. Insulin-like growth factor- tumorigenesis in mice. Cell. 2007;129:957–968.
637. Engelman JA, et al. Effective use of PI3K and mediated muscle cell survival: central roles for Akt 683. Samuels Y, Velculescu VE. Oncogenic muta-
MEK inhibitors to treat mutant Kras G12D and and cyclin-dependent kinase inhibitor p21. Mol Cell tions of PIK3CA in human cancers. Cell Cycle.
PIK3CA H1047R murine lung cancers. Nat Med. Biol. 2000;20:8983–8995. 2004;3:1221–1224.
2008;14:1351–1356. 659. Rossig L, et al. Akt-dependent phosphorylation 684. Engelman JA. Targeting PI3K signalling in cancer:
638. Halilovic E, et al. PIK3CA mutation uncouples of p21(Cip1) regulates PCNA binding and opportunities, challenges and limitations. Nat Rev
tumor growth and cyclin D1 regulation from proliferation of endothelial cells. Mol Cell Biol. Cancer. 2009;9:550–562.
MEK/ERK and mutant KRAS signaling. Cancer 2001;21:5644–5657. 685. Aoki M, Jiang H, Vogt PK. Proteasomal degrada-
Res. 2010;70:6804–6814. 660. Datta SR, et al. Akt phosphorylation of BAD couples tion of the FoxO1 transcriptional regulator in cells
639. Hu-Lieskovan S, et al. Improved antitumor activity survival signals to the cell-intrinsic death machinery. transformed by the P3k and Akt oncoproteins. Proc
of immunotherapy with BRAF and MEK inhibitors Cell. 1997;91:231–241. Natl Acad Sci USA. 2004;101:13613–13617.
Intracellular Signaling  •  CHAPTER 2 46.e11
46.e11

686. Miled N, et al. Mechanism of two classes of cancer 700. Davies BR, et al. Preclinical pharmacology of 713. Baselga J, et al. Everolimus in postmenopausal
mutations in the phosphoinositide 3-kinase catalytic AZD5363, an inhibitor of AKT: pharmacodynamics, hormone-receptor-positive advanced breast cancer.
subunit. Science. 2007;317:239–242. antitumor activity, and correlation of monotherapy N Engl J Med. 2012;366:520–529.
687. Oda K, et al. PIK3CA cooperates with other activity with genetic background. Mol Cancer Ther. 714. Carracedo A, et al. Inhibition of mTORC1 leads
phosphatidylinositol 3’-kinase pathway mutations to 2012;11:873–887. to MAPK pathway activation through a PI3K-
effect oncogenic transformation. Cancer Res. 2008;68: 701. Addie M, et  al. Discovery of 4-amino-N- dependent feedback loop in human cancer. J Clin
8127–8136. [(1S)-1-(4-chlorophenyl)-3-hydroxypropyl]-1- Invest. 2008;118:3065–3074.
688. Kang S, Denley A, Vanhaesebroeck B, Vogt (7H-pyrrolo[2,3-d]pyrimidin-4-yl)piperidine- 715. Breuleux M, et al. Increased AKT S473 phosphoryla-
PK. Oncogenic transformation induced by the 4-carboxamide (AZD5363), an orally bioavailable, tion after mTORC1 inhibition is rictor dependent
p110beta, -gamma, and -delta isoforms of class I potent inhibitor of Akt kinases. J Med Chem. and does not predict tumor cell response to PI3K/
phosphoinositide 3-kinase. Proc Natl Acad Sci USA. 2013;56:2059–2073. mTOR inhibition. Mol Cancer Ther. 2009;8:
2006;103:1289–1294. 702. Davies BR, et al. Tumors with AKT1E17K mutations 742–753.
689. Philp AJ, et al. The phosphatidylinositol 3′-kinase are rational targets for single agent or combination 716. Fan Q, et al. A kinase inhibitor targeted to mTORC1
p85alpha gene is an oncogene in human ovarian therapy with AKT INhibitors. Mol Cancer Ther. drives regression in glioblastoma. Cancer Cell.
and colon tumors. Cancer Res. 2001;61:7426–7429. 2015;14:2441–2451. 2017;31:424–435.
690. Cheung LW, et al. Naturally occurring neomorphic 703. Hyman DM, et al. Abstract B109: AZD5363, a cata- 717. Rodrik-Outmezguine VS, et al. Overcoming mTOR
PIK3R1 mutations activate the MAPK pathway, lytic pan-Akt inhibitor, in Akt1 E17K mutation posi- resistance mutations with a new-generation mTOR
dictating therapeutic response to MAPK pathway tive advanced solid tumors. Mol Cancer Ther. 2015;14: inhibitor. Nature. 2016;534:272–276.
inhibitors. Cancer Cell. 2014;26:479–494. B109. 718. Weinstein IB, Joe A. Oncogene addiction. Cancer
691. Thorpe LM, et al. PI3K-p110alpha mediates the 704. Hyman DM, et al. AKT inhibition in solid Res. 2008;68:3077–3080, discussion 3080.
oncogenic activity induced by loss of the novel tumor tumors with AKT1 mutations. J Clin Oncol. 719. Eberhard DA, et al. Mutations in the epidermal
suppressor PI3K-p85alpha. Proc Natl Acad Sci USA. 2017;35:2251–2259. growth factor receptor and in KRAS are predictive
2017;114:7095–7100. 705. Wander SA, Hennessy BT, Slingerland JM. Next- and prognostic indicators in patients with non-small-
692. Yang H, et al. MicroRNA expression profiling in generation mTOR inhibitors in clinical oncology: cell lung cancer treated with chemotherapy alone
human ovarian cancer: miR-214 induces cell survival how pathway complexity informs therapeutic and in combination with erlotinib. J Clin Oncol.
and cisplatin resistance by targeting PTEN. Cancer strategy. J Clin Invest. 2011;121:1231–1241. 2005;23:5900–5909.
Res. 2008;68:425–433. 706. Hudes G, et al. Temsirolimus, interferon alfa, or 720. Zehir A, et al. Mutational landscape of metastatic
693. Staal SP, Hartley JW. Thymic lymphoma induc- both for advanced renal-cell carcinoma. N Engl J cancer revealed from prospective clinical sequencing
tion by the AKT8 murine retrovirus. J Exp Med. Med. 2007;356:2271–2281. of 10,000 patients. Nat Med. 2017;23:703–713.
1988;167:1259–1264. 707. Motzer RJ, et al. Efficacy of everolimus in advanced 721. Wu JY, et al. Lung cancer with epidermal growth
694. Carpten JD, et al. A transforming mutation in the renal cell carcinoma: a double-blind, randomised, factor receptor exon 20 mutations is associated with
pleckstrin homology domain of AKT1 in cancer. placebo-controlled phase III trial. Lancet. 2008;372: poor gefitinib treatment response. Clin Cancer Res.
Nature. 2007;448:439–444. 449–456. 2008;14:4877–4882.
695. Cohen Y, et al. AKT1 pleckstrin homology domain 708. Yao JC, et al. Everolimus for advanced pancreatic 722. Yasuda H, Kobayashi S, Costa DB. EGFR exon 20
E17K activating mutation in endometrial carcinoma. neuroendocrine tumors. N Engl J Med. 2011; insertion mutations in non-small-cell lung cancer:
Gynecol Oncol. 2010;116:88–91. 364:514–523. preclinical data and clinical implications. Lancet
696. Zilberman DE, et al. AKT1 E17 K pleckstrin 709. Krueger DA, et al. Everolimus for subependymal Oncol. 2012;13:e23–e31.
homology domain mutation in urothelial carcinoma. giant-cell astrocytomas in tuberous sclerosis. N Engl 723. Hyman DM, et al. Vemurafenib in multiple
Cancer Genet Cytogenet. 2009;191:34–37. J Med. 2010;363:1801–1811. nonmelanoma cancers with BRAF V600 mutations.
697. Madhunapantula SV, Robertson GP. Therapeutic 710. Franz DN, et al. Efficacy and safety of everolimus for N Engl J Med. 2015;373:726–736.
implications of targeting AKT signaling in mela- subependymal giant cell astrocytomas associated with 724. Prahallad A, et al. Unresponsiveness of colon cancer
noma. Enzyme Res. 2011;2011:327923. tuberous sclerosis complex (EXIST-1): a multicentre, to BRAF(V600E) inhibition through feedback
698. Rosenthal A. Small molecule inhibitors in chronic randomised, placebo-controlled phase 3 trial. Lancet. activation of EGFR. Nature. 2012;483:100–103.
lymphocytic lymphoma and B cell non-Hodgkin 2013;381:125–132. 725. Camidge DR, Pao W, Sequist LV. Acquired resistance
lymphoma. Curr Hematol Malig Rep. 2017;12: 711. Kwiatkowski DJ, et al. Mutations in TSC1, TSC2, to TKIs in solid tumours: learning from lung cancer.
207–216. and MTOR are associated with response to rapalogs Nat Rev Clin Oncol. 2014;11:473–481.
699. Miller BW, et al. FDA approval: idelalisib mono- in patients with metastatic renal cell carcinoma. Clin 726. Holohan C, Van Schaeybroeck S, Longley DB,
therapy for the treatment of patients with follicular Cancer Res. 2016;22:2445–2452. Johnston PG. Cancer drug resistance: an evolv-
lymphoma and small lymphocytic lymphoma. Clin 712. Iyer G, et al. Genome sequencing identifies a basis ing paradigm. Nat Rev Cancer. 2013;13:714–
Cancer Res. 2015;21:1525–1529. for everolimus sensitivity. Science. 2012;338:221. 726.
46.e12 Part I: Science and Clinical Oncology

SELF-ASSESSMENT REVIEW QUESTIONS ANSWERS


1. Next-generation sequencing has helped reveal numerous unusual 1. (b) PTEN is a tumor suppressor of the AKT pathway and
genomic events, including chromosomal translocations. Which would not be a likely candidate fusion partner, because
of the following genes is NOT found as oncogenic fusion truncating mutations or deletions confer loss of PTEN function.
partners in chromosomal re-arrangements? All of the other genes listed have been found in patients with a
a. ABL variety of fusions partners. BCR-ABL, the Philadelphia
b. PTEN chromosome, which is found in nearly all cases of chronic
c. PDGFRβ myelogenous leukemias (CML) and one-third of ALLs, is
d. ALK the classic example of an oncogenic fusion driving cancer.
e. NTRK EML4-ALK fusions play a driving role in non–small cell lung
2. Which of the following is true of Hedgehog (SHH) signaling cancer (NSCLC). COL1A1-PDGFRβ fusions occur in
and targeted therapies? dermatofibrosarcoma protruberans, a rare sarcoma.
a. Secreted hedgehog ligands bind Smoothened receptors, which TPM3-TRKA fusions were first identified in colon cancer.
causes receptor dimerization and autophosphorylation of 2. (c)  The Smoothened receptor is a GPCR, not a receptor
intracellular tyrosine residues. tyrosine kinase, and therefore it does not get activated by
b. Erlotinib and gefitinib are FDA approved Smoothened dimerization and transphosphorylation. Vismodegib and
receptor inhibitors. sonidegib are FDA-approved smoothened receptor inhibitors.
c. Sonic hedgehog (SHH) binds to the receptor Patched Mutations in SHH, PTCH, and SMO are found in patients
(PTCH), which relieves its repression of Smoothened with inherited and sporadic basal cell carcinomas. Smoothened
(SMO). receptors couple to the G proteins Gαi and Gα12.
d. Mutations in SHH, PTCH, and SMO are found in patients 3. (d)  No direct inhibitors of Ras are available for clinical use.
with small cell lung cancer. Farnesyltransferase inhibitors were tested but were found to be
e. Smoothened receptors couple directly to actin. ineffective in tumors in which Ras mutations are common,
3. The Ras/MAP kinase and PI3 kinase/mTOR signaling cascades likely because of the ability of geranylgeranyl modifications to
are parallel but interconnected pathways that are frequently substitute for farnesylation in effectively targeting Ras to the
mutated in human cancer. Which of the following has NOT membrane. New drug development of allosteric inhibitors that
been an effective approach in targeting these pathways? specifically target KRAS G12C is underway. Trametinib is an
a. Allosteric inhibitors of MEK kinase FDA-approved allosteric inhibitor of MEK. Rapamycin and its
b. Small-molecule inhibitors and kinase of mTOR analogues, as well as novel kinase inhibitors such as RapaLink,
c. Monoclonal antibodies that bind to EGFR are efficacious mTOR inhibitors. Cetuximab and panitumumab
d. Farnesyltransferase inhibitors of Ras are anti-EGFR monoclonal antibody therapies. Vemurafenib,
e. Kinase inhibitors of mutant BRAF an ATP-competitive inhibitor of BRAF, is approved for the
4. Acquired resistance commonly occurs after treatment with treatment of patients with BRAF V600E mutant melanoma.
targeted inhibitors. Which of the following are known 4. (e)  Several mechanisms of resistance to kinase inhibitors have
mechanisms of resistance? emerged. For example, patients with BRAF V600E mutations
a. Parallel pathway upregulation that are treated with the specific RAF inhibitor vemurafenib
b. Gatekeeper mutation often develop resistance mediated by BRAF V600E splice
c. Amplification of specific RTKs variants or amplification; loss or mutation of NF1; parallel
d. Alternative splicing or amplification of the driver oncogene pathway activation of RTKs; mutation of PI3K/AKT
e. All of the above components; mutations in NRAS, RAF1 and MAP2K1/2
5. Which of the following statements is FALSE? (MEK1/2); and amplification of MITF.
a. Chromosomal rearrangement of BCR and ALK to generate 5. (a)  Translocation of the ABL gene on chromosome 9 with the
the Philadelphia chromosome is the driving event in CML. breakpoint cluster region (BCR) gene on chromosome 22 results
b. Recurrent somatic splice site alterations involving MET exon in the expression of a BCR-ABL fusion protein, called the
14 (METex14) have been identified in lung cancer. Philadelphia chromosome. This event is the pathognomonic
c. Integrin receptors perform inside-out and outside-in signaling molecular lesion in almost all chronic myelogenous leukemias
at the integrin adhesome. (CML). It is also found in approximately one-third of acute
d. ESR1 mutations are a common mechanism of acquired lymphoblastic leukemias (ALLs). The ABL tyrosine kinase
resistance to hormonal therapy in patients with breast cancer. inhibitor imatinib, as well as four other ABL inhibitors, are
e. Extracellular and transmembrane FGFR3 mutations are FDA approved in CML; imatinib and dasatinib are approved
recurrent in noninvasive and high-grade invasive bladder for ALL.
cancer.
Cellular Microenvironment and Metastases 3 
Erinn B. Rankin and Amato J. Giaccia

S UMMARY OF K EY P OI NT S
• Metastases are responsible for more the distant tissue; dormancy; and blood flow and by tissue-specific
than 90% of all cancer-related reactivation and proliferation in the factors.
deaths. new tissue microenvironment. • Primary tumors possess stem cells
• Gene dysregulation, the tumor • Colonization of metastatic tumor that can recapitulate the tumor from
microenvironment, and host cells cells requires the ability to a single cell, and a subset of these
drive the metastatic spread of tumor metabolically adapt, develop cancer stem cells may inherently
cells. angiogenesis, overcome dormancy, possess altered gene expression
• Metastasis can be subdivided into and proliferate in a foreign tissue. changes with increased metastatic
invasion and migration from the • The formation of a premetastatic potential.
primary tumor; intravasation into niche is essential for the growth of • Antimetastatic therapy will likely
the vasculature; dissemination extravasating metastatic tumor require the targeted inhibition of
and survival in the circulation; cells. many pathways that control
extravasation from the vasculature; • Organ specificity of tumor proliferation, invasion, angiogenesis,
survival and metabolic adaptation in metastases is determined both by and immune evasion.

One of the most important challenges in clinical oncology is the the tumor microenvironment such as hypoxia and extracellular vesicles
prevention and treatment of metastatic disease. With advances in promote metastatic phenotypes within these cell types.
surgical techniques and conventional and targeted therapies, localized
disease is effectively managed in the clinic. However, metastatic disease Cancer Stem Cells
is the primary cause of cancer-related deaths. Tumor metastasis involves
tumor cell invasion and migration from the primary tumor, intravasation The traditional view of tumors with a relatively homogeneous
into the vasculature, dissemination and survival in the circulation, population of tumorigenic cells has been significantly revised with
extravasation into distant tissues, survival and metabolic adaptation the isolation and characterization of cancer stem cells. Stem cells are
in the distant tissue, dormancy, reactivation, and overt colonization primal cells that retain the ability to renew themselves through cell
to form a new macroscopic tumor at a distant site.1 This process is division and can differentiate into a wide range of specialized cell
highly inefficient; it has been estimated that less than 0.01% of tumor types. Cells with stem cell properties have been identified in a variety
cells that enter the circulation develop into metastases. Despite this of solid tumors including colon, breast, head and neck, and pancreatic
inefficiency, metastases are responsible for more than 90% of all tumors, glioblastomas, medulloblastomas, and melanoma.3 This rare
cancer-related deaths. Understanding the biology and vulnerabilities subpopulation of tumor cells exhibits enhanced tumor-initiating
of metastatic tumor cells is of critical importance to improve overall potential: the ability to self-renew and differentiate into multiple
survival rates in cancer patients. With little evidence to support cell types.
mutations in “metastasis genes” as drivers of metastasis, current data The origin of cancer stem cells remains unclear and may differ
suggest that microenvironmental factors as well as epigenetic changes among tumor types. It has been hypothesized that cancer stem cells
may play key roles in metastasis. This chapter describes the cellular arise from either transformed resident stem and/or progenitor cells,
and molecular traits driving tumor metastasis and addresses how the or may represent a dedifferentiated epithelial tumor cell. In intestinal
tumor microenvironment influences this process. Most important, it cancer, mouse modeling studies have identified intestinal stem cells as
discusses how this knowledge can be translated into current and future the cells of origin. For example, intestinal stem cell–specific deletion
cancer therapies. of adenomatous polyposis coli (APC) leads to rapid cellular transforma-
tion with uncontrolled cellular growth and solid tumor formation.
TUMOR MICROENVIRONMENT AND In contrast, deletion of APC in short-lived transit-amplifying cells
METASTASIS was not sufficient to induce long-term tumor growth.4 Stem cell
properties can also be acquired by tumors cells through epithelial-
Although cellular intrinsic traits acquired by tumor cells are required mesenchymal transition (EMT). Induction of EMT in immortalized
for successful metastatic colonization, cellular and molecular factors human mammary epithelial cells was sufficient to induce the expression
within the tumor microenvironment significantly contribute to meta- of stem cell markers, enhance self-renewal, and increase the number
static progression. The tumor microenvironment contains a number of tumor-initiating cells.5 Although the molecular mechanisms that
of cell types that promote tumor progression, including cancer stem drive the cancer stem cell phenotype in tumor cells remains largely
cells, angiogenic vascular cells, infiltrating immune cells, and cancer- unknown, a study showed that the transcription factors Slug and
associated fibroblasts (CAFs).2 In addition, extracellular factors within Sox9 drive EMT and the cancer stem cell phenotype in breast cancer

47
48 Part I: Science and Clinical Oncology

cells.6 Future studies are eagerly awaited to further delineate the


intracellular signaling pathways driving the cancer stem cell phenotype I, TSP-1
ICAM IL-3
ctin, 3, C
in vivo. sel
e D2
, P- io n 48
The role of cancer stem cells in multistage tumor progression, B3 sat n Intr
KF ava atio ava
particularly with respect to metastasis, is poorly defined. Given that PF Intr avas cy sat
tr n ion
metastasis is an infrequent event that is achieved by only a small Ex orma
D
portion of cancer cells that reach distant sites, it has been hypothesized

GF
that cancer stem cells may be responsible for metastatic disease. Evidence Endothelial Pericyte

s, VEGF, CCLI8, E

MMP
to support this hypothesis has been generated in models of breast and

Extravasation

Premetastatic nic
Intravasation
pancreatic cancer. In the MMTV-PyMT model of breast cancer,

s, ALOX5, HGF, T
Invasion

Intravasation
Malanchi and colleagues observed that only the CD90+ CD24+

Invasion
population of cancer stem cells isolated from primary breast tumors Macrophage TME Neutrophil

contained cells with the ability to form metastases in the lung when

MMP
introduced into the tail vein.7 Furthermore, this study identified the

he
extracellular matrix protein, periostin, produced by fibroblasts and Platelet Fibroblast

GF
stromal tumor cells, as a critical factor to maintain the cancer stem on

-B
lati Pre
me
ircu on
cell phenotype and metastatic potential in these cells.7 In human n c
al i sat
i Intr tasta
tic
breast cancers, single-cell analysis of early-stage metastatic cells
av
rviv ava n rin
s CX Inv asati niche
Su Extr vasio asi on
demonstrated that these samples express distinct stemlike, EMT, eg
In t CL on
,in I2,
tin
prosurvival, and dormancy-associated gene expression signatures
GM
lec -CS
-se F, P
compared with metastatic cells from high-burden tissues.8 Moreover,
, P
TGF-B OSTN

transplanted stemlike metastatic cells from low-burden tissues exhibited


tumor-initiating capacity and were able to differentiate into luminal-like Figure 3.1  •  Multiple cellular components of the tumor microenvironment
cancer cells.8 Similarly in pancreatic cancer, lineage-tracing studies (TME) contribute to metastasis. Key cell types and mechanisms of action are
showed that circulating pancreatic tumor cells exhibit EMT and cancer shown.
stem cell properties, and initiate tumor formation. A critical role for
inflammation to maintain the ability of cancer stem cells to metastasize
was observed in this study.9 In addition, the extracellular protein
tenascin C (TNC), expressed by stem cell niches and cancer cells, Adhesion to the endothelium is an initial step in extravasation
was also found to promote stem cell signaling and lung metastases in that is followed by transendothelial cell migration.13 Tumor cell adhesion
breast cancer cells.10 Collectively, these data strongly implicate the to the endothelium requires the expression of cognate ligands and
tumor microenvironment in promoting the cancer stem cell phenotype receptors by cancer cells and endothelial cells.13 A variety of ligand-
and the initiation of tumor metastasis. receptor interactions have been shown to contribute to tumor
extravasation, including interactions with selectins, integrins, cadherins,
Angiogenic Vascular Cells CD44, and immunoglobulin (Ig) superfamily receptors (for a review,
see Reymond and colleagues13). For example, endothelial cell P- and
Tumor angiogenesis facilitates the hematogenous spread of metastatic E-selectins bind to tumor cells through cell-cell adhesion molecules
tumors. The aberrant production of proangiogenic factors by tumor such as integrins and CD44.14–19 Studies have suggested a role for the
cells results in malformed and irregular tumor blood vessels that often immunosuppressive cytokine interleukin (IL)-35 in facilitating cancer
contain breaks in their lining that facilitate tumor cell intravasation cell adhesion to endothelial cells. IL-35 was found to be highly expressed
and dissemination. Endothelial cells and pericytes are important by human pancreatic cancer cells in which it promoted ICAM1
structural and functional components of the tumor and distant tissue expression to facilitate endothelial cell adhesion and transendothelial
vasculature, and both have been considered as potential targets in migration via an ICAM1-fibrogen-ICAM1 bridge.20
metastatic therapy.11 Finally, endothelial cells within in the perivascular niche of the
bone marrow have been implicated in promoting tumor cell dormancy.
Endothelial Cells Similar to hematopoietic stem cells, dormant cancer cells have been
Endothelial cells play an active role in tumor cell intravasation, found to be localized within perivascular niches of the bone marrow
extravasation, and dormancy (Fig. 3.1). Studies have shown that microenvironment.21 Within stable microvasculature niches, endothelial
metabolic reprogramming of tumor endothelial cells toward a glycolytic cells promote tumor cell quiescence through the production of
phenotype contributes to the early stages of metastasis by promoting thrombospondin-1 (TSP1)21. In contrast, endothelial cells isolated
an abnormal tumor vasculature, tumor intravasation, and dissemination. from sprouting neovasculature promote tumor proliferation through
Cantelmo and colleagues performed mRNA sequencing analysis of the production of tumor-promoting factors including transforming
tumor-associated endothelial cells and discovered that tumor endothelial growth factor–β1 (TGF-β1) and periostin from sprouting endothelial
cells expressed a hyperglycolytic signature compared with normal cell tips.21
endothelial cells.12 The glycolytic phenotype within tumor-associated
endothelial cells could be inhibited through genetic and pharmacologic Pericytes
inhibition of the glycolytic enzyme 6-phosphofructo-2-kinase/fructose- Similar to endothelial cells, pericytes are thought to play a protumori-
2,6-bisphosphatase-3 (PFKFB3). Moreover, PFKFB3 inhibition in genic role by supporting blood vessel maturation and function (see
endothelial cells was sufficient to normalize the tumor vasculature Fig. 3.1). Pericytes promote endothelial cell survival and stabilize the
and significantly reduce tumor cell intravasation and metastasis.12 tumor vasculature.22 In addition, pericytes can directly facilitate distant
There are multiple mechanisms by which PFKFB3 inhibition in metastasis by promoting tumor cell intravasation through the upregula-
endothelial cells may have impaired metastatic dissemination, including tion of the transmembrane receptor endosialin (CD248).23 Studies
enhancing the integrity of the endothelial cell barrier, improving vessel have demonstrated that pericytes can also promote tumor cell intravasa-
maturation and pericyte coverage, and inhibiting cancer cell adhesion tion and metastasis through the IL-33–dependent recruitment of
molecules. These findings suggest that targeting glucose metabolism macrophages.24 Therefore it has been proposed that targeting pericytes
in tumor endothelial cells may offer therapeutic benefit for anticancer alone or in combination with endothelial cells may be an effective
therapy. strategy in the treatment of cancer.24,25 However, clinical data indicate
Cellular Microenvironment and Metastases  •  CHAPTER 3 49

that low pericyte coverage is associated with metastasis and poor circulating tumor cells within DNA meshes at distant tissues, (2)
prognosis in a number of cancers.25 A recent study demonstrated that promoting vascular permeability and the premetastatic niche, and (3)
depletion of pericytes suppressed tumor growth and enhanced metastasis stimulating tumor cell invasion and proliferation at distant tissue
of murine models of breast cancer.25 Enhanced metastasis was associated sites.36–38 Important to note, digesting NETs with DNase I is sufficient
with increased levels of tumor hypoxia, EMT, and Met receptor to reduce metastasis in multiple metastatic tumor models, suggesting
activation.25 These findings indicate that pericyte coverage may be that therapeutic targeting of NETs may prevent metastasis.36,37
important to stabilize the tumor vasculature and prevent the hypoxic
selection of aggressive tumor cells (see later discussion of hypoxia and Platelets
metastasis). These findings raise concerns when considering antipericyte Platelets are the second most abundant cell type in the blood and
treatments in cancer therapy, especially in the setting of metastasis. play an important role in hemostasis, thrombosis, inflammation, and
vascular biology.39 Platelets are among the first cell types that interact
Immune Cells with cancer cells in the circulation.20 Tumor cells release platelet
coagulation factors such as thrombin to promote platelet aggregation
Most solid tumors contain immune infiltrates derived from both the around circulating tumor cells. Indeed, increased blood coagulation
myeloid and lymphoid lineages that can have both positive and negative is often observed in cancer patients as a result of elevated levels of
effects on metastasis.2 This section focuses on immune cells that thromboplastin, procoagulant A, and phosphatidylserine produced
stimulate metastatic progression. by tumor cells.40,41 Platelets facilitate metastasis through multiple
mechanisms (see Fig. 3.1). Platelets are thought to form a physical
Macrophages barrier around disseminated cancer cells, protecting them from shear
Studies have linked a unique subset of macrophages, termed tumor- stress and lysis mediated by natural killer cells.42 In addition, platelets
associated macrophages (TAMs), to tumor progression and metastasis. can facilitate tumor cell extravasation by enhancing tumor cell adhesion
Clinically, the presence of excessive macrophages is associated with to endothelial cells and by promoting endothelial cell permeability.43–46
poor prognosis in the majority of patients with solid tumors including Finally, platelets have been shown to directly stimulate cancer cell
breast, prostate, cervix, bladder, endometrial, and kidney cancer.26–28 EMT and invasion through the release of TGF-β.47 Therapeutic
TAMs express a variety of growth factors and cytokines that promote inhibition of platelet binding to cancer cells through the disruption
tumor angiogenesis, invasion, intravasation, metastasis, and immune of α6β1 integrin/ADAM9 receptor may be an effective therapeutic
suppression (see Fig. 3.1)28,29. For example, macrophages within the strategy to target platelet-mediated metastasis in vivo.48
primary tumor contribute to tumor cell invasion and extravasation
by releasing a variety of proteases including serine, cysteine, and Fibroblasts and Mesenchymal Stem Cells
metalloproteases that remodel the extracellular matrix to enhance
tumor cell invasion and migration (for a review, see Cho and col- Accumulating evidence supports a critical role for CAFs in tumor
leagues2). Perivascular macrophages are thought to play a particularly progression and metastasis (see Fig. 3.1). Clinically it was observed
important role in tumor cell intravasation by promoting vascular that epigenetic changes and mutations commonly found in cancer
permeability. In preclinical models of breast cancer, genetic ablation cells, such as p53 and PTEN mutations, can also be found in cancer-
of vascular endothelial growth factor (VEGF) from macrophages was associated stroma.49,50 These studies provided early evidence to suggest
sufficient to reduce transient vascular permeability, leading to reduced that alterations in the stroma may contribute to tumor progression.
numbers of circulating tumor cells.30 Moreover, macrophages can Gene expression profiling studies of CAFs in human specimens and
promote tumor cell invasion through the release of chemokines and murine tumor models revealed an “activated” proinflammatory gene
cytokines including CCL18 and epidermal growth factor (EGF; for signature.51 Among the cytokines produced by CAFs, the chemokine
a review, see Ruffell and colleagues28). CXCL12 is of particular interest, given its role in driving tumor cell
Many questions remain regarding the factors that control the biologic migration and recruitment of endothelial progenitor cells expressing
activities of TAMs within the tumor microenvironment. TAMs localize the CXCR4 receptor. Consistent with these data, an important role
to regions of hypoxia, and thus it has been hypothesized that the for CAFs in promoting tumor invasion has been observed in several
hypoxic microenvironment may influence the unique characteristics murine models of cancer.51 In breast cancer, Hu and colleagues identified
and gene expression profiles of TAMs. Indeed, TAMs express high a role for CAFs in the transition of mammary ductal carcinoma in
levels of the hypoxia inducible transcription factor (HIF) HIF-2, which situ (DCIS) to invasive carcinoma. Coinjection of fibroblasts with
has been found to be an independent prognostic factor of outcome.31 DCIS cells resulted in invasive carcinomas, whereas injection of DCIS
When exposed to hypoxia, macrophages upregulate the expression of cells alone was only sufficient to induce mammary DCIS.52 CAFs
mitogenic and proangiogenic cytokines.32 Experimentally, deletion of promote tumor cell invasion by establishing invasion-permissive tracks
myeloid HIF-2 reduced TAM infiltration into tumors and impeded in the extracellular matrix through the secretion of factors that induce
tumor proliferation and growth.32 Thus, targeting TAMs and CD11b matrix remodeling.51,53 CAFs can also support tumor cell survival and
cells may be a viable mechanism for antimetastatic therapies. migration through the release of extracellular vesicles carrying annexin
A6/LDL receptor–related protein/thrombospondin 1 (ANXA6/LRP1/
Neutrophils TSP1) complexes.54 In addition to promotion of tumor invasion,
Neutrophils represent 50% to 75% of the total circulating leukocytes supportive roles for CAFs in the maintenance of cancer stem cell
in blood. They play an important role in inflammation and are early signaling and the establishment of the premetastatic niche have been
responders to pathogens including bacteria, fungi, and viruses. found.51 Further characterization of the distinct subsets of CAFs relevant
Neutrophils mediate direct antimicrobial activities through the release to metastasis in various tumor types is needed. In addition, further
of enzymes and toxic factors, the generation of reactive oxygen species, elucidation of the interplay between tumors and the stroma is warranted
and the release of nuclear material into extracellular traps called and may reveal novel strategies in the treatment of metastatic disease.
neutrophil extracellular traps (NETs).33 Studies have suggested an important role for a subset of CAFs,
Although neutrophils can inhibit the metastatic seeding of dis- mesenchymal stem cells (MSCs), in tumor progression and metastasis.
seminated tumor cells under some conditions, accumulating evidence Although MSCs are commonly isolated in CAF preparations, MSCs
suggests a prometastatic role for neutrophils (see Fig. 3.1).34–37 Study are functionally distinct in that they are able to undergo multipotent
findings have suggested that that metastatic cancer cells promote the differentiation. MSCs have been identified as important components
formation of NETs through the release of G-CSF.37 NETs may promote of the tumor stroma that promote the progression of ovarian, colon,
metastasis through multiple mechanisms including (1) trapping and pancreatic cancers.55–57 Cancer-associated MSCs have recently
50 Part I: Science and Clinical Oncology

been shown to promote pancreatic cancer cell proliferation, inva- exosome content and shedding. The shedding of exosomes from tumor
sion, and transendothelial cell migration through the production cells is increased under hypoxic conditions. In breast cancer cells,
of the cytokine granulocyte-macrophage colony-stimulating factor hypoxia promotes microvesicle shedding through the HIF-dependent
(GM-CSF).55 These findings suggest that inhibition of tumor- regulation of the small GTPase RAB22A, a protein that is localized
MSC cross talk may be a therapeutic strategy for the treatment of to budding microvesicles and promotes metastasis.74 Hypoxia also
pancreatic cancer. plays an important role in regulating exosome content and function.
Clinically, the levels of hypoxia-regulated proteins in plasma exosomes
Extracellular Vesicles are significantly higher in glioblastoma multiforme (GBM) patients
compared with healthy control patients.75 Moreover, exosomes derived
Extracellular vesicles are emerging as key factors that promote metastasis. from hypoxic GBM cells are potent inducers of angiogenesis compared
Studies have shown that tumor and stromal cells secrete small vesicles with exosomes isolated from normoxic GBM cells.75 Given that
that contain a variety of bioactive molecules such as proteins, lipids, extracellular vesicles can be detected in patient biologic fluids and
RNA, and DNA that can promote intercellular signaling and tumor contribute to cancer progression, there is great interest in use of
progression. Extracellular vesicles can be derived from either the exosomes as diagnostic biomarkers and therapeutic targets in cancer.
endosome (exosome) or the plasma membrane (microvesicle) to mediate
intercellular signaling with tumor and stromal cells within the local Hypoxia
and distant microenvironments. Clinically, extracellular vesicles and
their cargo have been associated with tumor progression. The concentra- Hypoxia is a potent microenvironmental factor driving metastatic
tions of exosomes have been found to be increased in the peripheral tumor progression. Clinically, hypoxia is associated with metastasis
blood of patients with ovarian, breast, and pancreatic cancers compared and poor survival in variety of cancer patients.76–79 Hypoxia selects
with healthy control patients (for a review, see Becker and colleagues58). for cells with low apoptotic potential and increases genomic instability,
Specific nucleic acid and protein expression signatures within exosomes allowing for rapid mutational adaptations.80–82 In addition, hypoxia
have also been associated with tumor progression and metastasis. For directly increases the expression of genes involved in glycolysis,
example, exosomal miR-141 expression in the serum of prostate cancer angiogenesis, cell survival, invasion, immune suppression, the cancer
patients is significantly higher in metastatic prostate patients compared stem cell phenotype, and metastasis. All of these changes allow cells
with patients with localized disease.59 An exosomal protein signature to adapt to oxygen-deprived conditions and permit cells to escape
containing the melanoma-specific protein tyrosinase-related protein-2 these conditions by establishing new blood supplies or by physically
(TYRP2), very late antigen 4 (VLA-4), HSP-70, and the MET moving from an oxygen-poor environment to an oxygen-rich
oncoprotein has been associated with metastasis in melanoma patients.60 environment.
In pancreatic cancer, the level of circulating glypican-1–positive The primary molecular mediators of hypoxic signaling are HIF-1
exosomes correlates with poor patient survival.61 and HIF-2. In the presence of oxygen, the alpha subunits of HIF-1
Functionally, extracellular vesicles have been shown to play an and HIF-2 are rapidly degraded through the cooperative actions of
important role in the tumor microenvironment and promote metastasis prolyl hydroxylase (PHD) enzymes PHD1, PHD2, and PHD3, and
through a variety of mechanisms including tumor immune suppression, the E3 ubiquitin ligase substrate recognition component VHL.83,84
invasion, angiogenesis, and the premetastatic niche (for a review, see Under hypoxia, HIF-1 and HIF-2 are stabilized and activate the
Kalluri and colleagues62). For example, melanoma cells promote immune expression of genes containing hypoxia response elements (HREs).76
evasion through the release of FasL-bearing microvesicles that trigger Over 200 genes that allow cells to survive and adapt to low oxygen
Fas-dependent apoptosis of lymphoid cells.63 Tumor-derived exosomes tensions are activated in response to HIF-1 and HIF-2.
can also directly promote the immunosuppressive phenotype of Consistent with the association between hypoxia and metastasis,
myeloid-derived suppressor cells through the activation of STAT3 HIF-1 and HIF-2 are highly expressed in primary tumors and
signaling in these cells.64 Tumor-derived exosomes promote an invasive metastases. Immunohistochemical analysis of human cancer specimens
phenotype by activating stromal cells to produce matrix metallo­ indicates that the majority of primary tumors and metastases have
proteinases (MMPs) such as MMP1.65 In addition, tumor-derived increased HIF-1 and/or HIF-2 protein compared with the normal
extracellular vesicles can directly promote the invasive capacity of adjacent tissue.76–78 Moreover, increased HIF expression is often
nonmetastatic cells at local and distant sites by transferring mRNAs associated with increased patient mortality.85 Experimentally, overexpres-
involved in migration and metastasis.66 Conversely, stromal microvesicles sion of HIF in tumor cells promotes metastasis,86 whereas inactivation
have also been shown to promote tumor cell invasion and metastasis. of HIF significantly decreases the metastatic potential of metastatic
Fibroblast-secreted exosomes promote breast cancer cell migration tumor cells.87–89 These clinical and experimental findings indicate an
through the activation of planar cell polarity (PCP) signaling.67 important role for HIF in metastatic tumor progression.
Astrocyte-mediated transfer of exosomal PTEN-targeting microRNAs HIFs influence multiple steps within the metastatic cascade
leads to PTEN loss in brain metastatic tumor cells that can support (Fig. 3.2). HIF-1 and HIF-2 activate the early stages of metastasis in
metastatic outgrowth through mechanisms involving the increased the primary tumor by promoting EMT, the cancer stem cell phenotype,
expression of the chemokine CCL2 and adaptive metastatic outgrowth.68 invasion, and migration (for a review, see Rankin and Giaccia79). HIFs
Exosome-mediated transfer of miR-105 by breast cancer cells promotes promote invasion and migration through multiple mechanisms. First,
vascular leakiness and metastasis by disrupting endothelial cell tight HIF regulates the E- to N-cadherin switch during EMT phenotype
junctions.69 Tumor-derived exosomes may also play a key role in through the direct activation of E-cadherin repressors including Twist1,
establishing a premetastatic niche at distant sites by activating signaling Zeb1/2, Snail.86,90,91 HIF signaling has also been shown to indirectly
within organ-specific cells to recruit bone marrow–derived macrophages, promote EMT through the activation of Notch, TGF-β, integrin-linked
activate Src phosphorylation and proinflammatory S100 gene expression, kinase (ILK), and the receptor tyrosine kinase AXL (for a review, see
and increase nutrient availability.70–72 Rankin and Giaccia79). Second, HIF directly upregulates the expression
The factors that regulate extracellular vesicle formation and content of multiple factors driving cellular invasion and migration. Most
in cancer remain poorly understood. Ostrowski and colleagues used notably, HIF drives the expression of the extracellular matrix protein
an RNA interference screen to identify factors important in exosome LOX.92 Increased LOX expression correlates with decreased distant
secretion in cancer cells. These studies revealed an important role for metastasis-free survival and overall survival in patients with breast and
the Rab GTPases Rab27a and Rab27b and the Rab27 effectors Slp4 head and neck cancer.92 In addition, LOX activation promotes the
and Slac2b in exosome secretion.73 Studies have implicated hypoxia invasive and metastatic potential of breast cancer cells. Erler and
as an important microenvironmental factor that can influence both colleagues demonstrated that genetic and pharmacologic inhibition
Cellular Microenvironment and Metastases  •  CHAPTER 3 51

Intravasation/
Invasion/migration Premetastatic niche Distant growth
extravasation

BMDC

Fibroblast
SNAIL AXL ANGPTL44 LOX
VEGF HK1/2
TWIST CDCP1 L1CAM LOXL2
ANGPT1/2 LDHA
ZEB1/2 ZEB1/2 VEGF LOXL4
PDFG PDK1
MMPs MET UPAR SDF-1
PIGF CKB
LOX PLOD1/2 MMPS VEGF
PAI-1 ENO1
CTGF AKAP12 SDF-1/CXCR4 CXCL12
TIE-2 ALDHA
CCR5 RAB222 VCAM1 CCL2
PGK1 GLUT-1/3
PGF PLUAR ICAM1 EXOSOMES

Figure 3.2  •  Hypoxia inducible transcription factor (HIF) signaling regulates multiple steps within the metastatic cascade. The key steps of the metastatic
cascade and target genes by which HIF signaling regulates these processes are shown.

of LOX in metastatic breast cancer is sufficient to prevent hypoxia- proliferation of endothelial cells and supports pericytes.29 VEGF-A is
induced cell invasion and metastasis. These findings indicate that a well-established HIF target and is significantly induced by HIF
LOX is a promising therapeutic target for the treatment of metastatic signaling in both primary tumors and metastases.102 In summary, HIF
disease. affects multiple aspects of tumor metastasis, indicating that therapeutic
HIF signaling also facilitates tumor cell intravasation and extravasa- targeting of HIF may be an effective strategy to selectively inhibit
tion from the vasculature. HIF activity in tumor cells results in the multiple aspects of metastasis.
release of factors that modulate endothelial-endothelial cell and
endothelial-tumor cell interactions. The upregulation of angiopoietin-
like 4 (ANGPTL4) by HIF promotes tumor cell motility and PATTERNS OF METASTASIS
intravasation of tumor cells through blood vessels.88 Simultaneously, Seed and Soil Hypothesis
HIF strengthens tumor cell–endothelial cell interactions through the
activation of L1 cell adhesion molecule (L1CAM).88 Another mechanism The patterns of colonization cannot solely be explained by the circula-
by which HIF promotes tumor cell intravasation and extravasation tory, lymphatic, and transcelomic routes described earlier. The propensity
is through the activation of genes that control vascular permeability. for certain tumors to seed in particular organs was first discussed as
Hypoxic induction of VEGF, angiopoietin 2, MMPs, and UPAR the “seed and soil” theory by Paget in 1889.103 For example, prostate
cooperatively act to destabilize the vascular wall and allow for tumor cancer often metastasizes to the bones, colon cancer has a tendency
cell entry.93 to metastasize to the liver, and the primary site for ovarian metastasis
Third, HIF activity in the primary tumor is involved in the forma- is the omentum.104 Colonization is an extremely inefficient process
tion of the metastatic niche. As mentioned earlier, priming the pre- that is heavily dependent on the interactions between “seeding” tumor
metastatic site for the recruitment and survival of metastatic tumor cells and the “soil” microenvironment of the secondary site. Many
cells is a critical step in successful metastatic colonization. In breast factors including formation of a premetastatic niche and interactions
cancer cells, HIF activity results in the upregulation and release of between circulating tumor cells and the distant microenvironment
LOX and LOX-like proteins (LOXL2 and LOXL4). These proteins determine patterns of colonization.
recruit bone marrow–derived cells (BMDCs) into the lung and prime
the lung for metastatic colonization.87,92,94 BMDCs produce chemokines Premetastatic Niche
that recruit tumor cells and stimulate blood vessel invasion.95–98 Studies
have also demonstrated a role for LOX in the establishment of the Over the past decade, studies have convincingly shown that formation
premetastatic niche in the bone wherein tumor-secreted LOX is both of a premetastatic niche is essential for the growth of extravasating
necessary and sufficient to induce osteolytic bone lesions and cortical metastatic tumor cells.98 Soluble factors and BMDCs are recruited to
bone loss in mice before the arrival of tumor cells.99 Another mechanism the distant site before the arrival of tumor cells to establish a permissive
by which HIF signaling controls the directional migration of metastatic environment for malignant colonization. The mechanisms by which
tumor cell sites is through the upregulation of SDF1/CXCR4 signal- the premetastatic niche is formed remain largely unknown. However,
ing.100,101 Stromal cells within target tissue sites produce stromal derived recent studies have shown that factors secreted by the primary tumor
factor-1 (SDF1), which recruits cancer cells expressing the receptor are directly involved in establishing a permissive ECM environment
CXCR4.29 Although these studies have indicated an important cellular and in recruiting BMDCs to the distant site.
intrinsic role for hypoxia and HIF signaling in the primary tumor, BMDCs expressing VEGFR1, c-kit, CD133, and CD134 have been
future studies are eagerly awaited to elucidate the role of HIF signaling detected at distant sites and increase angiogenesis at the premetastatic
in tumor support cells. sites. Targeted inhibition of VEGFR1 prevented niche formation and
Finally, HIF promotes the late stages of metastasis at a distant site subsequent metastatic progression. This tissue preconditioning may thus
by stimulating angiogenesis. As in the primary tumor, angiogenesis represent a key step that could be targeted therapeutically, although
is a critical step for metastatic tumor growth. VEGF-A is a proangiogenic studies with anti-VEGF therapy have failed to show significant benefit
factor produced by tumor cells that stimulates the recruitment and in preventing metastatic growth for long periods of time. The role of
52 Part I: Science and Clinical Oncology

hematopoietic progenitor cells and other BMDCs in tumor progression demonstrated different abilities to metastasize to the bone, lung, or
is reviewed by Kaplan and colleagues.105 adrenal medulla. These studies indicated that there are particular
An additional function of the premetastatic niche is to guide requirements for circulating tumor cells to colonize specific organs.
metastases to specific organs. Kaplan and coworkers demonstrated that The factors contributing to tissue specific colonization include cellular
injection of secreted factors collected from cancer cells that metastasize intrinsic factors within the disseminated tumor cells and specific factors
to multiple organs could permit cancer cells that only metastasize within the tissue microenvironment. Some of the key cellular intrinsic
to the lung when grown as subcutaneous tumors in mice to display molecules determining organ-specific metastasis have been identified
widespread metastasis through governing BMDC accumulation.98 and are briefly discussed in the following section. In addition, we are
Elevated fibronectin expression by fibroblasts and fibroblast-like cells beginning to unravel the mechanisms by which infiltrating tumor
resident at premetastatic sites seems to be a critical factor in the develop- cells “educate” the normal tissue stroma at distant sites to support
ment of the premetastatic niche. The key tumor-secreted factors that metastatic growth. Elucidation of the complex signaling networks that
determine metastatic sites and mediate premetastatic niche formation exist between tumor cells and their tissue microenvironment offers
have yet to be identified, although a role for tumor necrosis factor–α new opportunities for targeted therapy against cancer.
(TNF-α), TGF-β, and VEGF-α pathways has been demonstrated.106
MMPs may also play an important role in this process. For example, Metastases to the Bone
VEGFR1 signaling has been shown to be required for premetastatic
induction of MMP-9 expression in endothelial cells and macrophages There are two types of bone metastases: osteoblastic and osteolytic.112
of the lungs by distant primary tumors.107 This is thought to make Osteoblastic metastases are observed in patients with advanced prostate
the lung microenvironment more compliant for invasion of metastasiz- cancer. Both the differentiation of osteoblastic precursors and the
ing cells. This concept is supported by the finding that pericyte activity of osteoblast cells are stimulated by tumor and microenviron-
recruitment and angiogenesis are not observed in tumor-bearing mice mental signals such as bone morphogenetic protein (BMP), fibroblast
with MMP-9 knockout bone marrow cells.108 Furthermore, stromal- growth factor receptors (FGFRs), and insulin-like growth factor 1
derived MMP-2 and MMP-9 have also been shown to contribute to receptor (IGF1R).113 Runx-2 is a key transcription factor that regulates
establishment and growth of metastases.109 Thus, whereas there is the differentiation of osteoblasts and osteoblastic precursor cells, and
evidence that MMPs play multiple roles in metastases, clinical trials represents a potential new target for inhibiting osteoblastic metastases
with MMP inhibitors have failed to show significant efficacy. In large by preventing osteoblastic precursor differentiation.114 In contrast,
part, this has been due to unexpected normal tissue toxicities and osteolytic metastases are observed in patients with breast cancer or
conflicting roles in metastases. multiple myeloma, and in these patients interactions between tumor
cells and the bone microenvironment result in bone resorption and
Organ Specificity metastatic growth as a result of the unique interplay between osteoblasts
and osteoclasts (Fig. 3.3).112,115 Parathyroid hormone–related peptide
The organ distribution of metastases from a primary tumor is not (PTHrP) secreted by the tumor cells stimulates osteoblasts to produce
random. Minn and colleagues used bioluminescence imaging to receptor activator of nuclear factor–κB (RANK) ligand (RANKL).
reveal patterns of metastasis formation by human breast cancer cells Consequently, bone-resorbing osteoclast cells are activated by RANKL
in mice.110 They also showed that individual cells from the pleural when it binds to the RANK receptor. Activated osteoclasts upregulate
effusion of a breast cancer patient showed distinct patterns of organ- MMPs, which degrade the bone matrix–releasing growth factors such
specific metastasis.111 Single-cell progenies derived from this population as TGF-β, IGFs, platelet-derived growth factor (PDGF), fibroblast

37+U3
5$1./
5$1./
7XPRUFHOO 2VWHREODVW

Bone
5$1. marrow
niche
2VWHRFODVW

003V
7*)β
%03
,*)V
3'*)
)*)V
Figure 3.3  •  The vicious cycle of osteoclastic bone metastasis. Interactions between the tumor cells and the bone microenvironment create a vicious cycle
of osteolytic metastatic lesion development. Parathyroid hormone–related peptide (PTHrP), secreted by the tumor cells, stimulates osteoblasts to produce
receptor activator of nuclear factor–κB (RANK) ligand (RANKL). Bone-resorbing osteoclast cells are activated by RANKL when it binds to the RANK receptor.
The activated osteoclasts upregulate matrix metalloproteinases (MMPs) that degrade the bone matrix–releasing growth factors such as transforming growth
factor–β (TGF-β), insulin-like growth factors (IGFs), platelet-derived growth factor (PDGF), fibroblast growth factors (FGFs), and bone morphogenetic proteins
(BMPs). These factors stimulate tumor cells to release PTHrP, thus completing the vicious cycle. (Modified from Steeg PS. Tumor metastasis: mechanistic
insights and clinical challenges. Nat Med. 2006;12:895–904.)
Cellular Microenvironment and Metastases  •  CHAPTER 3 53

growth factors (FGFs), and BMP.112,116,117 These factors stimulate tumor that interactions between breast cancer stem cells and the lung stroma
cells to release PTHrP, thus restarting this pathway of bone resorption. are necessary for metastatic colonization. The ECM components TNC
Gene profiling has identified other important mediators of osteoclastic and periostin (POSTN) have recently been shown to be essential
bone metastases including CXCR4, MMP-1, CTGF, and osteopontin.118 factors required for tumor initiating cells to form metastases in the
Tumor cells additionally induce osteoclast formation by overexpressing lung.7,10,142 Malanchi and colleagues showed that tumor-initiating cells
ILs such as IL-8 and IL-11, and by downregulating macrophage induce POSTN expression in lung myofibroblasts to initiate coloniza-
colony-stimulating factor.119,120 All of these factors represent new targets tion and maintain their stem cell phenotype.7 Similarly, Oskarsson
for metastases, although the importance of each factor in osteoclastic and colleagues demonstrated a role for the extracellular protein TNC
bone metastases requires further clarification. in supporting the breast cancer cell stem cell phenotype and metastases
in the lung.10 Interesting to note, previous studies showed that TNC
Metastases to the Brain and POSTN form complexes together with collagen type I and
fibronectin in the ECM. It was shown that POSTN promotes the
Brain metastases are most commonly observed in patients with breast incorporation of TNC into the ECM to strengthen the ECM archi-
cancer, lung cancer, and melanoma. Vascular access to the brain is tecture, suggesting that these factors may act through similar pathways
strictly regulated by the blood-brain barrier, an endothelial layer in promoting cancer stem cell metastasis.143 In support of this notion,
surrounding the brain, connected by tight junctions and further lined maintenance of the cancer stem cell phenotype by TNC and POSTN
by a basement membrane, pericytes, and astrocytes.121 Macromolecules was mediated at least in part through the induction of WNT signaling.
are not usually able to traverse the blood-brain barrier, and it remains These studies suggest that targeting signaling pathways mediated
unclear how tumor cells are able to penetrate the blood-brain barrier. through the ECM may be an effective strategy against cancer stem
However, once the tumor cells are within the brain parenchyma, glial cell–driven metastasis.
cells permit establishment and growth of metastases by secreting
chemokines, cytokines, and growth factors.122 Other neurotransmitter Metastases to the Liver
hormones in the brain such as norepinephrine have also been reported
to increase tumor cell motility and metastatic spread.123 Liver metastases are observed in patients with breast, lung, and
Little is known about the key factors that determine colonization pancreatic cancers. However, liver metastases are most commonly
of the brain, mostly because there is a lack of good in vivo models of found in patients with metastatic colorectal cancer, because the liver
brain metastasis. Overexpression of Stat3 increases melanoma metastasis is the first capillary bed encountered by the metastasizing cells. The
to the brain and increases invasion of the melanoma cells and angio- circulatory system of the liver, in particular the liver sinusoids, does
genesis, although the pathways modulated by Stat3 signaling require not have a barrier limiting macromolecule flux, and it is well perfused
elucidation.124 The dependence of brain metastases on VEGF has and highly permeable, permitting metastasizing cancer cells to establish
been demonstrated experimentally in animals through inhibition studies themselves and grow. Thus, tumor cell invasion and survival are key
in which VEGF neutralization reduced brain metastases.125,126 determinants in metastatic colonization of the liver.144
In general, patients with brain metastases have an extremely poor Tumor cell survival within the liver is dependent on the cells’
prognosis. It is of concern that there has been an increase in the ability to metabolically adapt to the local tissue microenvironment.145
incidence of brain metastases in patients whose systemic disease is The liver is characterized by regions of hypoxia and highly glycolytic
well controlled.127–129 For example, patients with breast tumors that hepatocytes.146 Preclinical studies have revealed that disseminated colon
overexpress Her2 and who are treated with Her2 targeting trastuzumab cancer cells experience acute hypoxia and competition for glycolytic
(see later discussion) have an incidence of brain metastases twice that substrates early after hepatic dissemination.147 To survive, metastatic
of breast cancer patients who are treated with other agents.128 This is colorectal cells release the enzyme creatine kinase brain-type (CKB)
thought to be because the brain offers a sanctuary for metastatic breast into the extracellular space.147 CKB controls the amount of rapidly
tumor cells when systemic disease is being controlled.130 The develop- mobilized high-energy phosphates by catalyzing the transfer of a
ment of drugs that can cross the blood-brain barrier and target brain high-energy phosphate group from adenosine triphosphate (ATP) to
metastases is of paramount importance in the development of new the metabolite creatine, producing phosphocreatine.148 Under hypoxia,
targeted therapies to tackle this problem. Currently, the best treatment tumor cells import phosphocreatine and use it as a source of high-energy
for oligometastases to the brain is radiosurgery. phosphate that can be transferred to adenosine diphosphate (ADP)
to generate ATP.148 These findings suggest that hepatic hypoxia is a
Metastases to the Lung barrier to disseminated tumor cell survival and that disseminated
tumor cells metabolically adapt to overcome metabolic stress associated
Pulmonary metastases are frequently observed in patients with sarcoma, with hypoxia. Consistent with these findings, Dupuy and colleagues
breast cancer, melanoma, gastrointestinal cancer, and kidney cancer. observed that disseminated breast cancer cells in the liver are dependent
Because cardiac output from the pulmonary artery circulates through on glycolysis for survival. Glycolytic reprogramming in metastatic
the lungs, a high incidence of pulmonary metastases in cancer patients breast cancer cells was mediated by the HIF target gene pyruvate
can be expected on the basis of blood flow alone. Metastases therefore dehydrogenase kinase (PDK1).149 Under hypoxic conditions, PDK1
often initiate in pulmonary arterioles and later traverse the basement upregulation represses mitochondrial function by reducing pyruvate
membrane into the lung parenchyma. TGF-β and NF-κB facilitate entry into the TCA cycle and promoting the conversion of pyruvate
this process in breast cancer, as does osteopontin in hepatocellular to lactate, thereby supporting glycolysis through the regeneration of
cancer, and ezrin in osteosarcoma and breast cancer.131–136 In vivo regenerate NAD.150 Together, these findings highlight the importance
studies have identified a gene expression signature for lung metastasis of metabolic plasticity in promoting tumor cell survival within foreign
including several membrane-localized and secreted proteins that has tissue microenvironments.
been validated in breast cancer patients.110 Interesting to note, this
group of genes was able to induce lung metastasis when expressed CLINICAL RELEVANCE AND APPLICATIONS
together but not individually, suggesting essential cooperation between
proteins. Increased expression of antiapoptotic proteins such as Bcl-2 The future of cancer therapy lies in the ability to effectively prevent
and Bcl-xL is also observed in lung metastases, facilitating survival and treat metastatic disease. Metastases are largely resistant to current
and resulting in resistance to therapy.137–141 cytotoxic therapies and as a result are responsible for more than 90%
Adding to the complexity of signaling interactions needed for of all cancer-related deaths. Over the past decade, many of the cellular
successful metastatic colonization in the lung, studies have revealed and molecular constituents that drive metastatic tumor progression
54 Part I: Science and Clinical Oncology

have been defined. Based on these findings, a number of therapeutic protein that exerts a number of prometastatic activities including
agents targeting key components of the metastatic cascade have been migration, invasion, angiogenesis, and anoikis.157 Both genetic and
developed and tested in preclinical as well as clinical settings. Many therapeutic inhibition of CTGF have shown efficacy in primary and
of the initial targeted therapies developed for tumor metastasis inhibit metastatic tumor growth inhibition of preclinical models of pancreatic
factors driving tumor cell invasion and migration. Preclinical studies cancer.158,159 The results of clinical trials targeting LOX and CTGF
have demonstrated that agents that inhibit MMPs, the receptor tyrosine are eagerly awaited.
kinase AXL, miR-10b, fascin, and exosomes are effective in blocking Among the therapies targeting the cellular components of the
the initiation and/or progression of metastatic tumors in mice (see tumor microenvironment, those that target endothelial cells are the
reviews by Valastyan and Weinberg151 and Rankin and colleagues152). most advanced and have shown success within the clinic.29 Targeting
Of all of these agents, MMP inhibitors have been the most tested in VEGF signaling on vascular and lymphatic endothelial cells has shown
clinical trials. Disappointingly, these inhibitors failed to increase survival clinical benefits in patients with metastatic cancer.160 A variety of
in patients with advanced cancer and were associated with adverse VEGF inhibitors have been approved by the US Food and Drug
side effects.153 Important lessons regarding clinical trial design were Administration (FDA) for the treatment of metastasis. The humanized
learned from these trials. Because MMP inhibitors and other inhibitors anti-VEGF-A monoclonal antibody bevacizumab has been approved
of invasion are likely to function at the early stages of metastasis, it as a first-line therapy in combination with 5-fluorouracil for metastatic
is important to thoughtfully identify patient populations that may colon cancer. In addition, in patients with metastatic non–small cell
benefit most from these types of therapies. In addition, it is likely lung cancer, bevacizumab increased survival in combination with
that the combination of targeted agents controlling distinct aspects chemotherapy.161 In addition to biologic therapies, small-molecule
of metastasis may prove to be the most effective when targeting VEGF receptor inhibitors have also been developed for the treatment
metastatic cancer. of metastasis. Both sorafenib and sunitinib have been approved by
An emerging strategy in the treatment of metastasis involves the the FDA for the treatment of metastatic renal cancer. Phase III clinical
therapeutic targeting of cellular and molecular constituents within trials demonstrated that sorafenib monotherapy resulted in a significant
the tumor microenvironment. As mentioned earlier, hypoxia and HIF increase in progression-free survival in this patient population. The
signaling are critical drivers of both the tumorigenic and metastatic results of future clinical trials targeting additional key cellular com-
phenotypes. HIF and hypoxia-induced proteins represent therapeutic ponents of the tumor microenvironment, such as TAMs, are eagerly
targets that have the potential to be tumor specific because these awaited.
proteins are highly elevated in both the primary tumor and metastases
in comparison with normal tissue.154 In addition, targeting HIF is an CONCLUSION
attractive strategy for the treatment of metastatic disease because HIF
controls multiple aspects of metastasis including EMT, invasion, Metastatic disease, not the primary tumor, kills the majority of cancer
migration, metastatic niche formation, and metastatic tumor growth. patients. For such an important determinant of long-term survival,
In this regard, a number of small-molecule HIF inhibitors including progress in understanding the crucial genes and pathways that drive
digoxin and acriflavine have been identified and have shown success metastatic progression has been slow. The reasons for this slow progress
in preclinical studies by preventing lung metastasis in mice bearing have been numerous, including inadequate animal models that reflect
primary breast tumors.155 Although the efficacy of these inhibitors in the metastatic process in humans, failure to identify genes that specifi-
treating established metastatic disease remains unknown, these cally affect metastatic tumor growth, the complexity of host and
compounds do inhibit metastatic xenograft growth after tumor metastatic tumor interactions, and premature clinical trials focusing
implantation.88 In addition to targeting HIF directly, a number of on “attractive” gene targets. Recent studies have elucidated many of
HIF-induced proteins have been targeted for metastatic therapy. One the cellular and molecular factors driving metastatic progression. Based
very promising target is LOX, a hypoxia-induced secreted protein on the findings of these studies, new opportunities to target metastatic
involved in multiple stages of metastasis.92 LOX contributes to tumor disease have been identified. The future of metastatic therapy looks
cell invasion by cross-linking collagens in the ECM, which stimulates very promising, in large part because we understand the mistakes of
integrin-mediated cell-matrix adhesion and activation of FAK, and the past and are using multiple genomic and proteomic approaches
in addition provides a route (“highway”) by which tumor cells may to target what has for so long seemed to be an intractable problem.
travel. Furthermore, LOX is involved in the formation and maintenance It is only when we are able to attack the problem of metastases that
of the metastatic niche, allowing metastatic dissemination and growth. we will make significant inroads in our war against cancer.
Targeting secreted LOX through antibody or small-molecule inhibition
significantly reduced formation and growth of metastases to the lung,
liver, bone, and brain, in several preclinical studies. Another promising The complete reference list is available online at
hypoxia-induced target is CTGF.156 CTGF is an extracellular matrix ExpertConsult.com.

KEY REFERENCES
1. Oskarsson T, Batlle E, Massague J. Metastatic stem 13. Reymond N, d’Agua BB, Ridley AJ. Crossing the by met signaling pathway. Cancer Cell. 2012;21(1):
cells: sources, niches, and vital pathways. Cell Stem endothelial barrier during metastasis. Nat Rev Cancer. 66–81.
Cell. 2014;14(3):306–321. 2013;13(12):858–870. 28. Ruffell B, Coussens LM. Macrophages and thera-
2. Hanahan D, Coussens LM. Accessories to the crime: 21. Ghajar CM, Peinado H, Mori H, et al. The peri- peutic resistance in cancer. Cancer Cell. 2015;27(4):
functions of cells recruited to the tumor microen- vascular niche regulates breast tumour dormancy. 462–472.
vironment. Cancer Cell. 2012;21(3):309–322. Nat Cell Biol. 2013;15(7):807–817. 29. Joyce JA, Pollard JW. Microenvironmental regulation
7. Malanchi I, Santamaria-Martinez A, Susanto E, 24. Yang Y, Andersson P, Hosaka K, et al. The PDGF- of metastasis. Nat Rev Cancer. 2009;9(4):239–252.
et al. Interactions between cancer stem cells and BB-SOX7 axis-modulated IL-33 in pericytes and 30. Harney AS, Arwert EN, Entenberg D, et al.
their niche govern metastatic colonization. Nature. stromal cells promotes metastasis through tumour- Real-time imaging reveals local, transient vascular
2012;481(7379):85–89. associated macrophages. Nat Commun. 2016;7: permeability, and tumor cell intravasation stimulated
8. Lawson DA, Bhakta NR, Kessenbrock K, et al. 11385. by TIE2hi macrophage-derived VEGFA. Cancer
Single-cell analysis reveals a stem-cell program 25. Cooke VG, LeBleu VS, Keskin D, et al. Pericyte Discov. 2015;5(9):932–943.
in human metastatic breast cancer cells. Nature. depletion results in hypoxia-associated epithelial- 37. Park J, Wysocki RW, Amoozgar Z, et al. Cancer
2015;526(7571):131–135. to-mesenchymal transition and metastasis mediated cells induce metastasis-supporting neutrophil
Cellular Microenvironment and Metastases  •  CHAPTER 3 55

extracellular DNA traps. Sci Transl Med. 2016;8(361): 92. Erler JT, Bennewith KL, Nicolau M, et al. Lysyl 134. Ye QH, Qin LX, Forgues M, et al. Predicting hepatitis
361ra138. oxidase is essential for hypoxia-induced metastasis. B virus-positive metastatic hepatocellular carcinomas
47. Labelle M, Begum S, Hynes RO. Direct signal- Nature. 2006;440(7088):1222–1226. using gene expression profiling and supervised
ing between platelets and cancer cells induces an 93. De Bock K, Mazzone M, Carmeliet P. Antiangiogenic machine learning. Nat Med. 2003;9(4):416–423.
epithelial-mesenchymal-like transition and promotes therapy, hypoxia, and metastasis: risky liaisons, or 135. Khanna C, Wan X, Bose S, et al. The membrane-
metastasis. Cancer Cell. 2011;20(5):576–590. not? Nat Rev Clin Oncol. 2011;8(7):393–404. cytoskeleton linker ezrin is necessary for osteosarcoma
54. Leca J, Martinez S, Lac S, et al. Cancer-associated 94. Erler JT, Bennewith KL, Cox TR, et al. Hypoxia- metastasis. Nat Med. 2004;10(2):182–186.
fibroblast-derived annexin A6+ extracellular vesicles induced lysyl oxidase is a critical mediator of bone 140. Inbal B, Cohen O, Polak-Charcon S, et al. DAP
support pancreatic cancer aggressiveness. J Clin marrow cell recruitment to form the premetastatic kinase links the control of apoptosis to metastasis.
Invest. 2016;126(11):4140–4156. niche. Cancer Cell. 2009;15(1):35–44. Nature. 1997;390(6656):180–184.
58. Becker A, Thakur BK, Weiss JM, Kim HS, Peinado 95. Yang L, DeBusk LM, Fukuda K, et al. Expansion 147. Loo JM, Scherl A, Nguyen A, et al. Extracellular
H, Lyden D. Extracellular vesicles in cancer: cell-to- of myeloid immune suppressor Gr+CD11b+ cells metabolic energetics can promote cancer progression.
cell mediators of metastasis. Cancer Cell. 2016;30(6): in tumor-bearing host directly promotes tumor Cell. 2015;160(3):393–406.
836–848. angiogenesis. Cancer Cell. 2004;6(4):409–421. 148. Wyss M, Kaddurah-Daouk R. Creatine and creatinine
60. Peinado H, Aleckovic M, Lavotshkin S, et al. Mela- 96. Lyden D, Hattori K, Dias S, et al. Impaired recruit- metabolism. Physiol Rev. 2000;80(3):1107–1213.
noma exosomes educate bone marrow progenitor ment of bone-marrow-derived endothelial and hema- 149. Dupuy F, Tabaries S, Andrzejewski S, et al. PDK1-
cells toward a pro-metastatic phenotype through topoietic precursor cells blocks tumor angiogenesis dependent metabolic reprogramming dictates meta-
MET. Nat Med. 2012;18(6):883–891. and growth. Nat Med. 2001;7(11):1194–1201. static potential in breast cancer. Cell Metab. 2015;22(4):
66. Zomer A, Maynard C, Verweij FJ, et al. In vivo 97. Gao D, Nolan DJ, Mellick AS, Bambino K, 577–589.
imaging reveals extracellular vesicle-mediated phe- McDonnell K, Mittal V. Endothelial progenitor 150. Papandreou I, Cairns RA, Fontana L, Lim AL,
nocopying of metastatic behavior. Cell. 2015;161(5): cells control the angiogenic switch in mouse lung Denko NC. HIF-1 mediates adaptation to hypoxia
1046–1057. metastasis. Science. 2008;319(5860):195–198. by actively downregulating mitochondrial oxygen
69. Zhou W, Fong MY, Min Y, et al. Cancer-secreted 98. Kaplan RN, Riba RD, Zacharoulis S, et al. consumption. Cell Metab. 2006;3(3):187–197.
miR-105 destroys vascular endothelial barriers VEGFR1-positive haematopoietic bone marrow 151. Valastyan S, Weinberg RA. Tumor metastasis:
to promote metastasis. Cancer Cell. 2014;25(4): progenitors initiate the pre-metastatic niche. Nature. molecular insights and evolving paradigms. Cell.
501–515. 2005;438(7069):820–827. 2011;147(2):275–292.
70. Costa-Silva B, Aiello NM, Ocean AJ, et al. Pancreatic 101. Ceradini DJ, Kulkarni AR, Callaghan MJ, et al. 152. Rankin EB, Fuh KC, Taylor TE, et al. AXL is an
cancer exosomes initiate pre-metastatic niche forma- Progenitor cell trafficking is regulated by hypoxic essential factor and therapeutic target for metastatic
tion in the liver. Nat Cell Biol. 2015;17(6):816–826. gradients through HIF-1 induction of SDF-1. Nat ovarian cancer. Cancer Res. 2010;70(19):7570–
71. Hoshino A, Costa-Silva B, Shen TL, et al. Tumour Med. 2004;10(8):858–864. 7579.
exosome integrins determine organotropic metastasis. 104. Nieman KM, Kenny HA, Penicka CV, et al. 153. Coussens LM, Fingleton B, Matrisian LM. Matrix
Nature. 2015;527(7578):329–335. Adipocytes promote ovarian cancer metastasis and metalloproteinase inhibitors and cancer: trials
72. Fong MY, Zhou W, Liu L, et al. Breast-cancer- provide energy for rapid tumor growth. Nat Med. and tribulations. Science. 2002;295(5564):2387–
secreted miR-122 reprograms glucose metabolism 2011;17(11):1498–1503. 2392.
in premetastatic niche to promote metastasis. Nat 111. Minn AJ, Kang Y, Serganova I, et al. Distinct 154. Harris AL. Hypoxia—a key regulatory factor in
Cell Biol. 2015;17(2):183–194. organ-specific metastatic potential of individual tumour growth. Nat Rev Cancer. 2002;2(1):38–47.
79. Rankin EB, Giaccia AJ. Hypoxic control of metas- breast cancer cells and primary tumors. J Clin Invest. 158. Dornhofer N, Spong S, Bennewith K, et al. Con-
tasis. Science. 2016;352(6282):175–180. 2005;115(1):44–55. nective tissue growth factor-specific monoclonal
83. Jaakkola P, Mole DR, Tian YM, et al. Targeting of 112. Mundy GR. Metastasis to bone: causes, consequences antibody therapy inhibits pancreatic tumor growth
HIF-alpha to the von Hippel-Lindau ubiquitylation and therapeutic opportunities. Nat Rev Cancer. and metastasis. Cancer Res. 2006;66(11):5816–
complex by O2-regulated prolyl hydroxylation. 2002;2(8):584–593. 5827.
Science. 2001;292(5516):468–472. 118. Kang Y, Siegel PM, Shu W, et al. A multigenic 159. Aikawa T, Gunn J, Spong SM, Klaus SJ, Korc
84. Ivan M, Kondo K, Yang H, et al. HIFalpha program mediating breast cancer metastasis to bone. M. Connective tissue growth factor-specific
targeted for VHL-mediated destruction by proline Cancer Cell. 2003;3(6):537–549. antibody attenuates tumor growth, metastasis,
hydroxylation: implications for O2 sensing. Science. 121. Abbott NJ, Ronnback L, Hansson E. Astrocyte- and angiogenesis in an orthotopic mouse model of
2001;292(5516):464–468. endothelial interactions at the blood-brain barrier. pancreatic cancer. Mol Cancer Ther. 2006;5(5):1108–
85. Semenza GL. Defining the role of hypoxia-inducible Nat Rev Neurosci. 2006;7(1):41–53. 1116.
factor 1 in cancer biology and therapeutics. Oncogene. 130. Steeg PS. Tumor metastasis: mechanistic insights and
2010;29(5):625–634. clinical challenges. Nat Med. 2006;12(8):895–904.
Cellular Microenvironment and Metastases  •  CHAPTER 3 55.e1
55.e1

REFERENCES
1. Oskarsson T, Batlle E, Massague J. Metastatic stem promote metastatic dissemination. Cancer Res. 44. Karpatkin S, Pearlstein E, Ambrogio C, Coller BS.
cells: sources, niches, and vital pathways. Cell Stem 2016;76(18):5313–5325. Role of adhesive proteins in platelet tumor interac-
Cell. 2014;14(3):306–321. 24. Yang Y, Andersson P, Hosaka K, et al. The PDGF- tion in vitro and metastasis formation in vivo. J Clin
2. Hanahan D, Coussens LM. Accessories to the crime: BB-SOX7 axis-modulated IL-33 in pericytes and Invest. 1988;81(4):1012–1019.
functions of cells recruited to the tumor microen- stromal cells promotes metastasis through tumour- 45. Coupland LA, Chong BH, Parish CR. Platelets
vironment. Cancer Cell. 2012;21(3):309–322. associated macrophages. Nat Commun. 2016;7: and P-selectin control tumor cell metastasis in an
3. Cho RW, Clarke MF. Recent advances in cancer 11385. organ-specific manner and independently of NK
stem cells. Curr Opin Genet Dev. 2008;18(1):48–53. 25. Cooke VG, LeBleu VS, Keskin D, et al. Pericyte cells. Cancer Res. 2012;72(18):4662–4671.
4. Barker N, Ridgway RA, van Es JH, et al. Crypt depletion results in hypoxia-associated epithelial- 46. Schumacher D, Strilic B, Sivaraj KK, Wettschureck
stem cells as the cells-of-origin of intestinal cancer. to-mesenchymal transition and metastasis mediated N, Offermanns S. Platelet-derived nucleotides
Nature. 2009;457(7229):608–611. by met signaling pathway. Cancer Cell. 2012;21(1): promote tumor-cell transendothelial migration and
5. Mani SA, Guo W, Liao MJ, et al. The epithelial- 66–81. metastasis via P2Y2 receptor. Cancer Cell. 2013;24(1):
mesenchymal transition generates cells with proper- 26. Lewis CE, Pollard JW. Distinct role of macrophages 130–137.
ties of stem cells. Cell. 2008;133(4):704–715. in different tumor microenvironments. Cancer Res. 47. Labelle M, Begum S, Hynes RO. Direct signal-
6. Guo W, Keckesova Z, Donaher JL, et al. Slug and 2006;66(2):605–612. ing between platelets and cancer cells induces an
Sox9 cooperatively determine the mammary stem 27. Talmadge JE, Donkor M, Scholar E. Inflammatory epithelial-mesenchymal-like transition and promotes
cell state. Cell. 2012;148(5):1015–1028. cell infiltration of tumors: Jekyll or Hyde. Cancer metastasis. Cancer Cell. 2011;20(5):576–590.
7. Malanchi I, Santamaria-Martinez A, Susanto E, Metastasis Rev. 2007;26(3–4):373–400. 48. Mammadova-Bach E, Zigrino P, Brucker C, et al.
et al. Interactions between cancer stem cells and 28. Ruffell B, Coussens LM. Macrophages and thera- Platelet integrin alpha6beta1 controls lung metas-
their niche govern metastatic colonization. Nature. peutic resistance in cancer. Cancer Cell. 2015;27(4): tasis through direct binding to cancer cell-derived
2012;481(7379):85–89. 462–472. ADAM9. JCI insight. 2016;1(14):e88245.
8. Lawson DA, Bhakta NR, Kessenbrock K, et al. 29. Joyce JA, Pollard JW. Microenvironmental regulation 49. Hu M, Yao J, Cai L, et al. Distinct epigenetic changes
Single-cell analysis reveals a stem-cell program of metastasis. Nat Rev Cancer. 2009;9(4):239–252. in the stromal cells of breast cancers. Nat Genet.
in human metastatic breast cancer cells. Nature. 30. Harney AS, Arwert EN, Entenberg D, et al. 2005;37(8):899–905.
2015;526(7571):131–135. Real-time imaging reveals local, transient vascular 50. Kurose K, Gilley K, Matsumoto S, Watson PH, Zhou
9. Rhim AD, Mirek ET, Aiello NM, et al. EMT and permeability, and tumor cell intravasation stimulated XP, Eng C. Frequent somatic mutations in PTEN
dissemination precede pancreatic tumor formation. by TIE2hi macrophage-derived VEGFA. Cancer and TP53 are mutually exclusive in the stroma of
Cell. 2012;148(1–2):349–361. Discov. 2015;5(9):932–943. breast carcinomas. Nat Genet. 2002;32(3):355–357.
10. Oskarsson T, Acharyya S, Zhang XH, et al. Breast 31. Talks KL, Turley H, Gatter KC, et al. The expression 51. Strell C, Rundqvist H, Ostman A. Fibroblasts—a
cancer cells produce tenascin C as a metastatic and distribution of the hypoxia-inducible factors key host cell type in tumor initiation, progression,
niche component to colonize the lungs. Nat Med. HIF-1alpha and HIF-2alpha in normal human and metastasis. Ups J Med Sci. 2012;117(2):187–195.
2011;17(7):867–874. tissues, cancers, and tumor-associated macrophages. 52. Hu M, Yao J, Carroll DK, et al. Regulation of in
11. Bergers G, Song S, Meyer-Morse N, Bergsland E, Am J Pathol. 2000;157(2):411–421. situ to invasive breast carcinoma transition. Cancer
Hanahan D. Benefits of targeting both pericytes and 32. Imtiyaz HZ, Williams EP, Hickey MM, et al. Cell. 2008;13(5):394–406.
endothelial cells in the tumor vasculature with kinase Hypoxia-inducible factor 2alpha regulates macro- 53. Gaggioli C, Hooper S, Hidalgo-Carcedo C, et al.
inhibitors. J Clin Invest. 2003;111(9):1287–1295. phage function in mouse models of acute and tumor Fibroblast-led collective invasion of carcinoma cells
12. Cantelmo AR, Conradi LC, Brajic A, et al. inflammation. J Clin Invest. 2010;120(8):2699–2714. with differing roles for RhoGTPases in leading and
Inhibition of the glycolytic activator PFKFB3 in 33. Branzk N, Papayannopoulos V. Molecular mecha- following cells. Nat Cell Biol. 2007;9(12):1392–1400.
endothelium induces tumor vessel normalization, nisms regulating NETosis in infection and disease. 54. Leca J, Martinez S, Lac S, et al. Cancer-associated
impairs metastasis, and improves chemotherapy. Semin Immunopathol. 2013;35(4):513–530. fibroblast-derived annexin A6+ extracellular vesicles
Cancer Cell. 2016;30(6):968–985. 34. Granot Z, Henke E, Comen EA, King TA, Norton support pancreatic cancer aggressiveness. J Clin
13. Reymond N, d’Agua BB, Ridley AJ. Crossing the L, Benezra R. Tumor entrained neutrophils inhibit Invest. 2016;126(11):4140–4156.
endothelial barrier during metastasis. Nat Rev Cancer. seeding in the premetastatic lung. Cancer Cell. 55. Waghray M, Yalamanchili M, Dziubinski M,
2013;13(12):858–870. 2011;20(3):300–314. et al. GM-CSF mediates mesenchymal-epithelial
14. Mannori G, Santoro D, Carter L, Corless C, Nelson 35. Spiegel A, Brooks MW, Houshyar S, et al. Neu- cross-talk in pancreatic cancer. Cancer Discov.
RM, Bevilacqua MP. Inhibition of colon carcinoma trophils suppress intraluminal NK cell-mediated 2016;6(8):886–899.
cell lung colony formation by a soluble form of tumor cell clearance and enhance extravasation 56. Lin JT, Wang JY, Chen MK, et al. Colon cancer
E-selectin. Am J Pathol. 1997;151(1):233–243. of disseminated carcinoma cells. Cancer Discov. mesenchymal stem cells modulate the tumorigenicity
15. Kim YJ, Borsig L, Varki NM, Varki A. P-selectin 2016;6(6):630–649. of colon cancer through interleukin 6. Exp Cell Res.
deficiency attenuates tumor growth and metastasis. 36. Cools-Lartigue J, Spicer J, McDonald B, et al. 2013;319(14):2216–2229.
Proc Natl Acad Sci USA. 1998;95(16):9325–9330. Neutrophil extracellular traps sequester circulating 57. McLean K, Gong Y, Choi Y, et al. Human ovarian
16. Nesbit M, Herlyn M. Adhesion receptors in tumor cells and promote metastasis. J Clin Invest. carcinoma-associated mesenchymal stem cells regu-
human melanoma progression. Invasion Metastasis. 2013 Jul 01. late cancer stem cells and tumorigenesis via altered
1994;14(1–6):131–146. 37. Park J, Wysocki RW, Amoozgar Z, et al. Cancer cells BMP production. J Clin Invest. 2011;121(8):3206–
17. Wang H, Fu W, Im JH, et al. Tumor cell alpha3beta1 induce metastasis-supporting neutrophil extracellular 3219.
integrin and vascular laminin-5 mediate pulmonary DNA traps. Sci Transl Med. 2016;8(361):361ra138. 58. Becker A, Thakur BK, Weiss JM, Kim HS, Peinado
arrest and metastasis. J Cell Biol. 2004;164(6): 38. Kolaczkowska E, Jenne CN, Surewaard BG, et al. H, Lyden D. Extracellular vesicles in cancer:
935–941. Molecular mechanisms of NET formation and cell-to-cell mediators of metastasis. Cancer Cell.
18. Ruoslahti E. Fibronectin and its alpha 5 beta 1 degradation revealed by intravital imaging in the 2016;30(6):836–848.
integrin receptor in malignancy. Invasion Metastasis. liver vasculature. Nat Commun. 2015;6:6673. 59. Li Z, Ma YY, Wang J, et al. Exosomal microRNA-141
1994;14(1–6):87–97. 39. Kaushansky K. Historical review: megakaryopoiesis is upregulated in the serum of prostate cancer
19. Birch M, Mitchell S, Hart IR. Isolation and and thrombopoiesis. Blood. 2008;111(3):981– patients. Onco Targets Ther. 2016;9:139–148.
characterization of human melanoma cell variants 986. 60. Peinado H, Aleckovic M, Lavotshkin S, et al. Mela-
expressing high and low levels of CD44. Cancer 40. Cliffton EE, Grossi CE. The rationale of anticoagu- noma exosomes educate bone marrow progenitor
Res. 1991;51(24):6660–6667. lants in the treatment of cancer. J Med. 1974;5(1): cells toward a pro-metastatic phenotype through
20. Huang C, Li N, Li Z, et al. Tumour-derived Interleu- 107–113. MET. Nat Med. 2012;18(6):883–891.
kin 35 promotes pancreatic ductal adenocarcinoma 41. Fidler IJ. Macrophages and metastasis—a bio- 61. Melo SA, Luecke LB, Kahlert C, et al. Glypican-1
cell extravasation and metastasis by inducing ICAM1 logical approach to cancer therapy. Cancer Res. identifies cancer exosomes and detects early pan-
expression. Nat Commun. 2017;8:14035. 1985;45(10):4714–4726. creatic cancer. Nature. 2015;523(7559):177–182.
21. Ghajar CM, Peinado H, Mori H, et al. The peri- 42. Nieswandt B, Hafner M, Echtenacher B, Mannel 62. Kalluri R. The biology and function of exosomes
vascular niche regulates breast tumour dormancy. DN. Lysis of tumor cells by natural killer cells in cancer. J Clin Invest. 2016;126(4):1208–1215.
Nat Cell Biol. 2013;15(7):807–817. in mice is impeded by platelets. Cancer Res. 63. Andreola G, Rivoltini L, Castelli C, et al. Induction
22. Raza A, Franklin MJ, Dudek AZ. Pericytes and 1999;59(6):1295–1300. of lymphocyte apoptosis by tumor cell secre-
vessel maturation during tumor angiogenesis and 43. Camerer E, Qazi AA, Duong DN, Cornelissen tion of FasL-bearing microvesicles. J Exp Med.
metastasis. Am J Hematol. 2010;85(8):593–598. I, Advincula R, Coughlin SR. Platelets, protease- 2002;195(10):1303–1316.
23. Viski C, Konig C, Kijewska M, Mogler C, Isacke activated receptors, and fibrinogen in hematogenous 64. Chalmin F, Ladoire S, Mignot G, et al. Membrane-
CM, Augustin HG. Endosialin-expressing pericytes metastasis. Blood. 2004;104(2):397–401. associated Hsp72 from tumor-derived exosomes
55.e2 Part I: Science and Clinical Oncology

mediates STAT3-dependent immunosuppressive 85. Semenza GL. Defining the role of hypoxia-inducible 105. Kaplan RN, Psaila B, Lyden D. Bone marrow cells in
function of mouse and human myeloid-derived factor 1 in cancer biology and therapeutics. Oncogene. the ‘pre-metastatic niche’: within bone and beyond.
suppressor cells. J Clin Invest. 2010;120(2):457–471. 2010;29(5):625–634. Cancer Metastasis Rev. 2006;25(4):521–529.
65. Atay S, Banskota S, Crow J, Sethi G, Rink L, Godwin 86. Yang MH, Wu MZ, Chiou SH, et al. Direct regula- 106. Hiratsuka S, Watanabe A, Aburatani H, Maru Y.
AK. Oncogenic KIT-containing exosomes increase tion of TWIST by HIF-1alpha promotes metastasis. Tumour-mediated upregulation of chemoattractants
gastrointestinal stromal tumor cell invasion. Proc Nat Cell Biol. 2008;10(3):295–305. and recruitment of myeloid cells predetermines lung
Natl Acad Sci USA. 2014;111(2):711–716. 87. Wong CC, Gilkes DM, Zhang H, et al. Hypoxia- metastasis. Nat Cell Biol. 2006;8(12):1369–1375.
66. Zomer A, Maynard C, Verweij FJ, et al. In vivo inducible factor 1 is a master regulator of breast 107. Hiratsuka S, Nakamura K, Iwai S, et al. MMP9
imaging reveals extracellular vesicle-mediated phe- cancer metastatic niche formation. Proc Natl Acad induction by vascular endothelial growth factor
nocopying of metastatic behavior. Cell. 2015;161(5): Sci USA. 2011;108(39):16369–16374. receptor-1 is involved in lung-specific metastasis.
1046–1057. 88. Zhang H, Wong CC, Wei H, et al. HIF-1-dependent Cancer Cell. 2002;2(4):289–300.
67. Luga V, Zhang L, Viloria-Petit AM, et al. Exosomes expression of angiopoietin-like 4 and L1CAM 108. Chantrain CF, Shimada H, Jodele S, et al. Stromal
mediate stromal mobilization of autocrine Wnt-PCP mediates vascular metastasis of hypoxic breast cancer matrix metalloproteinase-9 regulates the vascular
signaling in breast cancer cell migration. Cell. cells to the lungs. Oncogene. 2012;31(14):1757– architecture in neuroblastoma by promoting
2012;151(7):1542–1556. 1770. pericyte recruitment. Cancer Res. 2004;64(5):1675–
68. Zhang L, Zhang S, Yao J, et al. Microenvironment- 89. Liao D, Corle C, Seagroves TN, Johnson RS. 1686.
induced PTEN loss by exosomal microRNA primes Hypoxia-inducible factor-1alpha is a key regulator of 109. Masson V, de la Ballina LR, Munaut C, et al. Con-
brain metastasis outgrowth. Nature. 2015;527(7576): metastasis in a transgenic model of cancer initiation tribution of host MMP-2 and MMP-9 to promote
100–104. and progression. Cancer Res. 2007;67(2):563–572. tumor vascularization and invasion of malignant
69. Zhou W, Fong MY, Min Y, et al. Cancer-secreted 90. Krishnamachary B, Zagzag D, Nagasawa H, et al. keratinocytes. FASEB J. 2005;19(2):234–236.
miR-105 destroys vascular endothelial barriers Hypoxia-inducible factor-1-dependent repres- 110. Minn AJ, Gupta GP, Siegel PM, et al. Genes that
to promote metastasis. Cancer Cell. 2014;25(4): sion of E-cadherin in von Hippel-Lindau tumor mediate breast cancer metastasis to lung. Nature.
501–515. suppressor-null renal cell carcinoma mediated 2005;436(7050):518–524.
70. Costa-Silva B, Aiello NM, Ocean AJ, et al. Pancreatic by TCF3, ZFHX1A, and ZFHX1B. Cancer Res. 111. Minn AJ, Kang Y, Serganova I, et al. Distinct
cancer exosomes initiate pre-metastatic niche forma- 2006;66(5):2725–2731. organ-specific metastatic potential of individual
tion in the liver. Nat Cell Biol. 2015;17(6):816–826. 91. Imai T, Horiuchi A, Wang C, et al. Hypoxia attenu- breast cancer cells and primary tumors. J Clin Invest.
71. Hoshino A, Costa-Silva B, Shen TL, et al. Tumour ates the expression of E-cadherin via up-regulation 2005;115(1):44–55.
exosome integrins determine organotropic metastasis. of SNAIL in ovarian carcinoma cells. Am J Pathol. 112. Mundy GR. Metastasis to bone: causes, consequences
Nature. 2015;527(7578):329–335. 2003;163(4):1437–1447. and therapeutic opportunities. Nat Rev Cancer.
72. Fong MY, Zhou W, Liu L, et al. Breast-cancer- 92. Erler JT, Bennewith KL, Nicolau M, et al. Lysyl 2002;2(8):584–593.
secreted miR-122 reprograms glucose metabolism oxidase is essential for hypoxia-induced metastasis. 113. Logothetis CJ, Lin SH. Osteoblasts in prostate cancer
in premetastatic niche to promote metastasis. Nat Nature. 2006;440(7088):1222–1226. metastasis to bone. Nat Rev Cancer. 2005;5(1):21–28.
Cell Biol. 2015;17(2):183–194. 93. De Bock K, Mazzone M, Carmeliet P. Antiangiogenic 114. Harada S, Rodan GA. Control of osteoblast
73. Ostrowski M, Carmo NB, Krumeich S, et al. Rab27a therapy, hypoxia, and metastasis: risky liaisons, or function and regulation of bone mass. Nature.
and Rab27b control different steps of the exosome not? Nat Rev Clin Oncol. 2011;8(7):393–404. 2003;423(6937):349–355.
secretion pathway. Nat Cell Biol. 2010;12(1):19–30, 94. Erler JT, Bennewith KL, Cox TR, et al. Hypoxia- 115. Kozlow W, Guise TA. Breast cancer metastasis to
sup pp 1–13. induced lysyl oxidase is a critical mediator of bone bone: mechanisms of osteolysis and implications
74. Wang T, Gilkes DM, Takano N, et al. Hypoxia- marrow cell recruitment to form the premetastatic for therapy. J Mammary Gland Biol Neoplasia.
inducible factors and RAB22A mediate formation niche. Cancer Cell. 2009;15(1):35–44. 2005;10(2):169–180.
of microvesicles that stimulate breast cancer 95. Yang L, DeBusk LM, Fukuda K, et al. Expansion 116. Kang Y, He W, Tulley S, et al. Breast cancer
invasion and metastasis. Proc Natl Acad Sci USA. of myeloid immune suppressor Gr+CD11b+ cells bone metastasis mediated by the Smad tumor
2014;111(31):E3234–E3242. in tumor-bearing host directly promotes tumor suppressor pathway. Proc Natl Acad Sci USA.
75. Kucharzewska P, Christianson HC, Welch JE, et al. angiogenesis. Cancer Cell. 2004;6(4):409–421. 2005;102(39):13909–13914.
Exosomes reflect the hypoxic status of glioma cells 96. Lyden D, Hattori K, Dias S, et al. Impaired recruit- 117. Yin JJ, Selander K, Chirgwin JM, et al. TGF-beta
and mediate hypoxia-dependent activation of vascular ment of bone-marrow-derived endothelial and hema- signaling blockade inhibits PTHrP secretion by
cells during tumor development. Proc Natl Acad Sci topoietic precursor cells blocks tumor angiogenesis breast cancer cells and bone metastases development.
USA. 2013;110(18):7312–7317. and growth. Nat Med. 2001;7(11):1194–1201. J Clin Invest. 1999;103(2):197–206.
76. Rankin EB, Giaccia AJ. The role of hypoxia- 97. Gao D, Nolan DJ, Mellick AS, Bambino K, 118. Kang Y, Siegel PM, Shu W, et al. A multigenic
inducible factors in tumorigenesis. Cell Death Differ. McDonnell K, Mittal V. Endothelial progenitor program mediating breast cancer metastasis to bone.
2008;15(4):678–685. cells control the angiogenic switch in mouse lung Cancer Cell. 2003;3(6):537–549.
77. Schindl M, Schoppmann SF, Samonigg H, et al. metastasis. Science. 2008;319(5860):195–198. 119. Morgan H, Tumber A, Hill PA. Breast cancer cells
Overexpression of hypoxia-inducible factor 1alpha 98. Kaplan RN, Riba RD, Zacharoulis S, et al. induce osteoclast formation by stimulating host
is associated with an unfavorable prognosis in VEGFR1-positive haematopoietic bone marrow IL-11 production and downregulating granulocyte/
lymph node-positive breast cancer. Clin Cancer progenitors initiate the pre-metastatic niche. Nature. macrophage colony-stimulating factor. Int J Cancer.
Res. 2002;8(6):1831–1837. 2005;438(7069):820–827. 2004;109(5):653–660.
78. Yamamoto Y, Ibusuki M, Okumura Y, et al. 99. Cox TR, Rumney RM, Schoof EM, et al. The hypoxic 120. Boyle WJ, Simonet WS, Lacey DL. Osteo-
Hypoxia-inducible factor 1alpha is closely linked cancer secretome induces pre-metastatic bone lesions clast differentiation and activation. Nature.
to an aggressive phenotype in breast cancer. Breast through lysyl oxidase. Nature. 2015;522(7554): 2003;423(6937):337–342.
Cancer Res Treat. 2008;110(3):465–475. 106–110. 121. Abbott NJ, Ronnback L, Hansson E. Astrocyte-
79. Rankin EB, Giaccia AJ. Hypoxic control of metas- 100. Staller P, Sulitkova J, Lisztwan J, Moch H, Oakeley endothelial interactions at the blood-brain barrier.
tasis. Science. 2016;352(6282):175–180. EJ, Krek W. Chemokine receptor CXCR4 down- Nat Rev Neurosci. 2006;7(1):41–53.
80. Knowles HJ, Harris AL. Hypoxia and oxidative stress regulated by von Hippel-Lindau tumour suppressor 122. Lassman AB, DeAngelis LM. Brain metastases. Neurol
in breast cancer. Hypoxia and tumourigenesis. Breast pVHL. Nature. 2003;425(6955):307–311. Clin. 2003;21(1):1–23, vii.
Cancer Res. 2001;3(5):318–322. 101. Ceradini DJ, Kulkarni AR, Callaghan MJ, et al. 123. Entschladen F, Drell TL, Lang K, Joseph J, Zaenker
81. Reynolds TY, Rockwell S, Glazer PM. Genetic Progenitor cell trafficking is regulated by hypoxic KS. Neurotransmitters and chemokines regulate
instability induced by the tumor microenvironment. gradients through HIF-1 induction of SDF-1. Nat tumor cell migration: potential for a new pharma-
Cancer Res. 1996;56(24):5754–5757. Med. 2004;10(8):858–864. cological approach to inhibit invasion and metastasis
82. Kim CY, Tsai MH, Osmanian C, et al. Selection of 102. Maxwell PH, Dachs GU, Gleadle JM, et al. development. Curr Pharm Des. 2005;11(3):403–411.
human cervical epithelial cells that possess reduced Hypoxia-inducible factor-1 modulates gene 124. Xie TX, Huang FJ, Aldape KD, et al. Activation of
apoptotic potential to low-oxygen conditions. Cancer expression in solid tumors and influences both stat3 in human melanoma promotes brain metastasis.
Res. 1997;57(19):4200–4204. angiogenesis and tumor growth. Proc Natl Acad Cancer Res. 2006;66(6):3188–3196.
83. Jaakkola P, Mole DR, Tian YM, et al. Targeting of Sci USA. 1997;94(15):8104–8109. 125. Kim LS, Huang S, Lu W, Lev DC, Price JE. Vascular
HIF-alpha to the von Hippel-Lindau ubiquitylation 103. Paget S. The distribution of secondary growths in endothelial growth factor expression promotes the
complex by O2-regulated prolyl hydroxylation. cancer of the breast. 1889. Cancer Metastasis Rev. growth of breast cancer brain metastases in nude
Science. 2001;292(5516):468–472. 1989;8(2):98–101. mice. Clin Exp Metastasis. 2004;21(2):107–118.
84. Ivan M, Kondo K, Yang H, et al. HIFalpha 104. Nieman KM, Kenny HA, Penicka CV, et al. 126. Yano S, Shinohara H, Herbst RS, et al. Expression
targeted for VHL-mediated destruction by proline Adipocytes promote ovarian cancer metastasis and of vascular endothelial growth factor is necessary but
hydroxylation: implications for O2 sensing. Science. provide energy for rapid tumor growth. Nat Med. not sufficient for production and growth of brain
2001;292(5516):464–468. 2011;17(11):1498–1503. metastasis. Cancer Res. 2000;60(17):4959–4967.
Cellular Microenvironment and Metastases  •  CHAPTER 3 55.e3
55.e3

127. Omuro AM, Kris MG, Miller VA, et al. High cells is prevented by soluble factors present in the 150. Papandreou I, Cairns RA, Fontana L, Lim AL,
incidence of disease recurrence in the brain and target organ of metastasis. Breast Cancer Res Treat. Denko NC. HIF-1 mediates adaptation to hypoxia
leptomeninges in patients with nonsmall cell lung 2001;69(1):39–51. by actively downregulating mitochondrial oxygen
carcinoma after response to gefitinib. Cancer. 2005; 138. Martin SS, Ridgeway AG, Pinkas J, et al. A cyto- consumption. Cell Metab. 2006;3(3):187–197.
103(11):2344–2348. skeleton-based functional genetic screen identifies 151. Valastyan S, Weinberg RA. Tumor metastasis:
128. Clayton AJ, Danson S, Jolly S, et al. Incidence of cere- Bcl-xL as an enhancer of metastasis, but not primary molecular insights and evolving paradigms. Cell.
bral metastases in patients treated with trastuzumab tumor growth. Oncogene. 2004;23(26):4641–4645. 2011;147(2):275–292.
for metastatic breast cancer. Br J Cancer. 2004;91(4): 139. Pinkas J, Martin SS, Leder P. Bcl-2-mediated 152. Rankin EB, Fuh KC, Taylor TE, et al. AXL is an
639–643. cell survival promotes metastasis of EpH4 beta­ essential factor and therapeutic target for metastatic
129. Bendell JC, Domchek SM, Burstein HJ, et al. Central MEKDD mammary epithelial cells. Mol Cancer ovarian cancer. Cancer Res. 2010;70(19):7570–7579.
nervous system metastases in women who receive Res. 2004;2(10):551–556. 153. Coussens LM, Fingleton B, Matrisian LM. Matrix
trastuzumab-based therapy for metastatic breast 140. Inbal B, Cohen O, Polak-Charcon S, et al. DAP metalloproteinase inhibitors and cancer: trials
carcinoma. Cancer. 2003;97(12):2972–2977. kinase links the control of apoptosis to metastasis. and tribulations. Science. 2002;295(5564):2387–
130. Steeg PS. Tumor metastasis: mechanistic insights and Nature. 1997;390(6656):180–184. 2392.
clinical challenges. Nat Med. 2006;12(8):895–904. 141. Wong CW, Lee A, Shientag L, et al. Apoptosis: an 154. Harris AL. Hypoxia—a key regulatory factor in
131. Yu Q, Stamenkovic I. Transforming growth factor- early event in metastatic inefficiency. Cancer Res. tumour growth. Nat Rev Cancer. 2002;2(1):38–47.
beta facilitates breast carcinoma metastasis by 2001;61(1):333–338. 155. Wong CC, Zhang H, Gilkes DM, et al. Inhibitors
promoting tumor cell survival. Clin Exp Metastasis. 142. O’Connell JT, Sugimoto H, Cooke VG, et al. VEGF-A of hypoxia-inducible factor 1 block breast cancer
2004;21(3):235–242. and tenascin-C produced by S100A4+ stromal cells metastatic niche formation and lung metastasis.
132. Siegel PM, Shu W, Cardiff RD, Muller WJ, are important for metastatic colonization. Proc Natl J Mol Med. 2012;90(7):803–815.
Massague J. Transforming growth factor beta signal- Acad Sci USA. 2011;108(38):16002–16007. 156. Higgins DF, Biju MP, Akai Y, Wutz A, Johnson RS,
ing impairs Neu-induced mammary tumorigenesis 143. Kii I, Nishiyama T, Li M, et al. Incorporation of Haase VH. Hypoxic induction of Ctgf is directly
while promoting pulmonary metastasis. Proc Natl tenascin-C into the extracellular matrix by periostin mediated by Hif-1. Am J Physiol Renal Physiol.
Acad Sci USA. 2003;100(14):8430–8435. underlies an extracellular meshwork architecture. J 2004;287(6):F1223–F1232.
133. Luo JL, Maeda S, Hsu LC, Yagita H, Karin M. Biol Chem. 2010;285(3):2028–2039. 157. Chu CY, Chang CC, Prakash E, Kuo ML. Con-
Inhibition of NF-kappaB in cancer cells converts 144. Chambers AF, Groom AC, MacDonald IC. Dis- nective tissue growth factor (CTGF) and cancer
inflammation-induced tumor growth mediated by semination and growth of cancer cells in metastatic progression. J Biomed Sci. 2008;15(6):675–685.
TNFalpha to TRAIL-mediated tumor regression. sites. Nat Rev Cancer. 2002;2(8):563–572. 158. Dornhofer N, Spong S, Bennewith K, et al. Con-
Cancer Cell. 2004;6(3):297–305. 145. Lehuede C, Dupuy F, Rabinovitch R, Jones RG, nective tissue growth factor-specific monoclonal
134. Ye QH, Qin LX, Forgues M, et al. Predicting hepatitis Siegel PM. Metabolic plasticity as a determinant antibody therapy inhibits pancreatic tumor growth
B virus-positive metastatic hepatocellular carcinomas of tumor growth and metastasis. Cancer Res. and metastasis. Cancer Res. 2006;66(11):5816–
using gene expression profiling and supervised 2016;76(18):5201–5208. 5827.
machine learning. Nat Med. 2003;9(4):416–423. 146. Jungermann K. Metabolic zonation of liver paren- 159. Aikawa T, Gunn J, Spong SM, Klaus SJ, Korc
135. Khanna C, Wan X, Bose S, et al. The membrane- chyma. Semin Liver Dis. 1988;8(4):329–341. M. Connective tissue growth factor-specific
cytoskeleton linker ezrin is necessary for osteosarcoma 147. Loo JM, Scherl A, Nguyen A, et al. Extracellular antibody attenuates tumor growth, metastasis,
metastasis. Nat Med. 2004;10(2):182–186. metabolic energetics can promote cancer progression. and angiogenesis in an orthotopic mouse model of
136. Sarrio D, Rodriguez-Pinilla SM, Dotor A, Calero F, Cell. 2015;160(3):393–406. pancreatic cancer. Mol Cancer Ther. 2006;5(5):1108–
Hardisson D, Palacios J. Abnormal ezrin localiza- 148. Wyss M, Kaddurah-Daouk R. Creatine and creatinine 1116.
tion is associated with clinicopathological features metabolism. Physiol Rev. 2000;80(3):1107–1213. 160. Sleeman J, Schmid A, Thiele W. Tumor lymphatics.
in invasive breast carcinomas. Breast Cancer Res Treat. 149. Dupuy F, Tabaries S, Andrzejewski S, et al. PDK1- Semin Cancer Biol. 2009;19(5):285–297.
2006;98(1):71–79. dependent metabolic reprogramming dictates 161. Jain RK. Antiangiogenesis strategies revisited: from
137. Ladeda V, Adam AP, Puricelli L, Bal de Kier Joffe E. metastatic potential in breast cancer. Cell Metab. starving tumors to alleviating hypoxia. Cancer Cell.
Apoptotic cell death in mammary adenocarcinoma 2015;22(4):577–589. 2014;26(5):605–622.
4  Control of the Cell Cycle
Marcos Malumbres

S UMMARY OF K EY P OI N T S
• Most cells in postnatal tissues are intrinsic factors, such as growth cycle arrest, senescence, or death in
quiescent. Exceptions include factor or cytokine exposure, cell-to- response to cellular stress.
abundant cells of the hematopoietic cell contact, and metabolic • Checkpoints minimize replication
system, skin, and gastrointestinal constraints. and segregation of damaged DNA,
mucosa, as well as other minor • The internal cell cycle machinery is or the abnormal segregation of
progenitor populations in other controlled largely by oscillating levels chromosomes to daughter cells, thus
tissues. of cyclin proteins and by modulation protecting cells against genome
• Many quiescent cells can reenter of cyclin-dependent kinase (Cdk) instability.
into the cell cycle with the activity. One way in which growth • Disruption of cell cycle controls is a
appropriate stimuli, and the control factors regulate cell cycle hallmark of all malignant cells.
of this process is essential for tissue progression is by affecting the levels Frequent tumor-associated
homeostasis. of the D-type cyclins, Cdk activity, alterations include aberrations in
• The key challenges for proliferating and the function of the growth factor signaling pathways,
cells are to make an accurate copy retinoblastoma protein. dysregulation of the core cell cycle
of the 3 billion bases of DNA • Cell cycle checkpoints are machinery, and/or disruption of cell
(S phase) and to segregate the surveillance mechanisms that link the cycle checkpoint controls.
duplicated chromosomes equally rate of cell cycle transitions to the • Because cell cycle control is
into daughter cells (mitosis). timely and accurate completion of disrupted in virtually all tumor types,
• Progression through the cell cycle is prior dependent events; p53 is a the cell cycle machinery provides
dependent on both extrinsic and checkpoint protein that induces cell multiple therapeutic opportunities.

Most cells in the adult body are quiescent—that is, they are biochemi- exposed to external agents (e.g., reactive chemicals and ultraviolet
cally and functionally active but do not divide to generate daughter light) and to internal agents (e.g., byproducts of normal intracellular
cells. However, specific populations retain the ability to proliferate metabolism, such as reactive oxygen intermediates) that can induce
throughout the adult life span, which is essential for proper tissue DNA damage. A major goal of specific cell cycle checkpoints is to
homeostasis. For example, cells of the hematopoietic compartment detect DNA damage and activate cell cycle arrest and DNA repair
and the gut have a high rate of turnover, and active proliferation is mechanisms, thereby maintaining genomic integrity.
essential for the maintenance of these tissues. On average, about 2 Anything that disrupts proper cell cycle progression can lead to
trillion cell divisions occur in an adult human every 24 hours (about either the reduction or the expansion of a particular cell population.
25 million per second). The decision about whether to proliferate is It is now clear that such changes are a hallmark of tumor cells, which
tightly regulated.1,2 It is influenced by a variety of exogenous signals, carry mutations that impair signaling pathways that suppress prolifera-
including nutrients and growth factors, as well as inhibitory factors, tion and/or activate pathways that promote proliferation. In addition,
and the interaction of the cell with its neighbors and with the underlying most (if not all) human tumor cells have mutations within key
extracellular matrix. Each of these factors stimulates intracellular signal- components of both the cell cycle machinery and checkpoint path-
ing pathways that can either promote or suppress proliferation. The ways.1,5,6 This characteristic has important clinical implications, because
cell integrates all of these signals, and if the balance is favorable, the the presence of these defects can modulate cellular sensitivity to
cell will initiate a series of processes, collectively known as the cell chemotherapeutic regimens that induce DNA damage or mitotic
cycle, that lead to cell division into two daughter cells. catastrophe. This chapter focuses on the mechanics of the cell cycle
During the past four decades, extensive effort has been placed on and checkpoint signaling pathways and discusses how this knowledge
unraveling the basic molecular events that control the cell division can lead to the efficient use of current anticancer therapies and to the
cycle. Studies in a variety of organisms have identified evolutionarily development of novel agents.
conserved machinery that regulates eukaryotic cell cycle transitions
through the action of key enzymes, including cyclin-dependent kinases
(Cdks) and other kinases.3 It is essential that proliferating cells copy CELL DIVISION CYCLE
their genomes and segregate them to the daughter cells with high Overview of the Cell Cycle Machinery
fidelity. Eukaryotic cells therefore have evolved a series of surveillance
pathways, termed cell cycle checkpoints, that monitor for potential Cell division proceeds through a well-defined series of stages
problems during the cell cycle process.4 Human cells are continuously (Fig. 4.1). First, the cell moves from the nonproliferative, quiescent

56
Control of the Cell Cycle  •  CHAPTER 4 57

G1/S Intra-S G2/M SAC

Microtubule nucleation
DNA replication & repair Cytokinesis
spindle

Cell cycle entry Centrosome duplication Chromosome Chromosome


G1 and separation condensation segregation

G0 G1 S G2 M M
Cyclin D
Cyclin E Cyclin A Cyclin B

Restriction point
Figure 4.1  •  The cell division cycle. One round of cell division requires high-fidelity duplication of deoxyribonucleic acid during the S phase of the cell
cycle and proper segregation of duplicated chromosomes during mitosis, or the M phase. Before and after the S phase and M phase, the cell transits through
“gap” phases, termed G1 and G2. Quiescent (G0) cells require the appropriate mitogenic stimuli for reentry into the cell cycle by inducing D-type cyclins.
These stimuli are required up to a specific moment, known as the restriction point, at which time the cell cycle becomes independent of the mitogenic signals.
The cellular processes required for transition through the different cell cycle phases are controlled by the action of regulatory pathways and mostly are driven
by the expression of specific cyclins and the activation of cyclin-dependent kinases. These transitions are monitored by signaling pathways known as the cell
cycle checkpoints (represented by blue columns) that function to arrest the cell cycle at different phases (G1/S, intra–S phase, G2/M) in the presence of DNA
damage. In addition, the spindle assembly checkpoint (SAC) delays the exit from mitosis in the presence of defective chromosome alignment by inhibiting
the degradation of cyclin B1 and the subsequent inactivation of cyclin-dependent kinases.

(also known as G0) state into the first gap phase, or G1, in which (originally cdc2), was cloned by virtue of its ability to complement a
the cell essentially is readying itself for the cell division process. This mutant cdc2 yeast strain.12 Subsequent studies identified additional
process involves a dramatic upregulation of both transcriptional and human Cdks and determined that they regulate distinct cell cycle
translational programs, not only to yield the proteins required to stages; for example, Cdk4 and Cdk6 regulate cell cycle entry, whereas
regulate cell division but also to essentially double the complement Cdk2 may have specific roles during the G1-to-S transition and S
of macromolecules so that one cell can give rise to two cells without a phase. Cdk1 is essential in the control of G2 and mitosis and also
loss of cell size. Not surprisingly, this process takes a significant amount may play additional roles in earlier stages. The human genome encodes
of time (from 8 to 30 hours in cultured cells) and energy.7 Studies about 15 additional Cdks, although the functional relevance of many
with cultured cells show that mitogenic growth factors are essential for of them is still unknown.3,8–11
continued passage through the G1 phase. Specifically, if growth factors The activity of these kinases is controlled by multiple regulatory
are withdrawn at any point during this phase, the cell will not divide. mechanisms. Cdks act in association with a cyclin subunit that binds
However, as it nears the end of the G1 phase, the cell passes through a to the conserved PSTAIRE helix within the kinase.13 Cyclin binding
key transition point, called the restriction point, whereupon it becomes causes a reorientation of residues within the active sites that is essential
growth factor independent and is fully committed to undergoing cell for kinase activity.8,13 The associated cyclin also determines the substrate
division.1 The cell then enters the DNA synthesis phase, or S phase, specificity of the resulting cyclin/Cdk complex. The cyclins are quite
in which each of the chromosomes is replicated once and only once. divergent, especially in their N-terminal sequences, but they all share
This is followed by a second gap phase, called G2, which lasts 3 to a highly conserved 100–amino acid sequence, called the cyclin box,
5 hours, and the cell then initiates mitosis, or the M phase, a rapid that mediates Cdk binding and activation.9 As their name implies,
phase (lasting about 1 hour) in which the chromosomes are segregated. cyclins originally were identified as proteins whose expression was
On completion of mitosis, the daughter cells can enter quiescence restricted to a particular stage of the cell cycle14 because of cell
or initiate a second round of cell division, depending on the milieu. cycle–dependent regulation of both cyclin gene transcription and
Progression throughout the different phases of the cell cycle depends protein degradation. The human genome encodes more than 25
on the activity of key molecules that drive transcription, translation, cyclin-like proteins, yet only four distinct subclasses—D-, E-, A-, and
or the structural changes required for cell division (Table 4.1). A large B-type cyclins—are thought to play key roles in cell cycle regulation
number of these changes are modulated by protein phosphorylation (see Fig. 4.1).9 Each of these classes has a few paralogs (e.g., cyclin
and dephosphorylation, but other molecular processes such as D1, D2, and D3; cyclin E1 and E2; cyclin A1 and A2; and cyclin
SUMOylation, acetylation, or ubiquitin-dependent protein degradation B1, B2, and B3). The relative roles of these paralogs are not completely
are crucial for ordered cell cycle progression. Many of these cell cycle clear in most cases. Although some functional redundancy may exist,
regulators have been involved in tumor development or may be attractive published evidence suggests differences in regulation, expression pattern,
targets for cancer therapy and will be introduced in the following and substrate specificity.9,15
sections. The activation of cyclin/Cdk complexes requires considerable
posttranslational regulation.8,9,16 First, kinase activation is dependent
Cyclin-Dependent Kinases and Their Regulators on phosphorylation of a threonine residue that is adjacent to the
The Cdks constitute a large subfamily of highly conserved Ser/Thr active site (Thr160 in Cdk2). This phosphorylation is catalyzed by a
kinases that are defined by their dependence on a regulatory subunit, kinase, called Cdk-activating kinase (CAK).17,18 In mammalian cells,
called a cyclin.8–11 The first identified human Cdk, called Cdk1 phosphorylation occurs after cyclin binding. Although it appears that
58 Part I: Science and Clinical Oncology

Table 4.1  Representative Molecules Involved in Cell Cycle Regulation


Protein Family/
Complex Representative Members Function
KINASES
Cyclin-dependent Heterodimeric complexes formed of a cyclin (A, B, Phosphorylation of multiple proteins to drive progression
kinases D, and E types) and a Cdk (Cdk1, Cdk2, Cdk4, Cdk6); throughout the different phases of the cell cycle
Cdk7 functions as a Cdk-activating kinase
Wee1/Myt1 Wee1, Myt1 Inactivation of Cdks
Aurora-A holoenzyme Aurora-A and its nonkinase activator, Tpx2 Spindle dynamics, chromosome segregation, and cytokinesis
Chromosome passenger Aurora-B (Aurora-C?), Incenp, survivin, borealin Chromosome segregation
complex (CPC)
Polo-like kinases Plk1–Plk5 Centrosome function, chromosome segregation, and cytokinesis
NIMA-related kinases Nek1–Nek11 Centrosome function and mitosis
Haspin Haspin Phosphorylates histone H3 to recruit the CPC
Mastl Mastl Inhibition of PP2A phosphatases
CDK INHIBITORS
INK4 proteins p16INK4a, p15INK4b, p18INK4c, p19INK4d Inhibition of Cdk4 and Cdk6 during G1 progression
Cip/Kip inhibitors p21Cip1, p27Kip1, p57Kip2 Cdk inhibition and other roles in transcription or the cytoskeleton
TRANSCRIPTIONAL CONTROL
Retinoblastoma family pRB, p107, p130 Repression of the transcription of genes required for the cell cycle
E2F transcription factors E2F1–E2F8 Transcription of genes encoding S-phase and mitotic regulators
PHOSPHATASES
Cdc14 Cdc14a, Cdc14b Control of transcription and cell cycle progression
Cdc25 Cdc25a, Cdc25b, and Cdc25c Cdk activation and cell cycle progression
PP1 Multiple complexes with different regulatory Protein dephosphorylation
subunits
PP2A Multiple complexes with different regulatory Protein dephosphorylation; major Cdk-counteracting phosphatase
subunits
UBIQUITIN LIGASES
SCF E3 ubiquitin ligase formed of Rbx1, Cul1, Skp1, and Targets multiple cell cycle regulators (e.g., p27Kip1 or cyclin E) for
an F-box protein (e.g., Skp2 or βTrCP) ubiquitin-dependent degradation during interphase
APC/C E3 ubiquitin ligase composed for multiple subunits Targets multiple cell cycle regulators for ubiquitin-dependent
including Cdc20 or Cdh1 as coactivator molecules degradation during mitosis (cyclin B, securin) and G1 (e.g.,
Aurora-A, Plk1, or Tpx2)
OTHER FUNCTIONS
Kinesins More than 600 proteins including Eg5, CenpE, Microtubule-based motor proteins that hydrolyze ATP to generate
MCAK energy for movement along microtubule fibers
Cohesins and condensins Smc family (1–4), Rad21, Pds5, and SA (Stag) Structure and regulation of DNA
proteins, among others
Kinetochore proteins More than 100 proteins including the CENP Linking the chromosomes to microtubules and regulation of
(centromere-binding proteins) family, and the Knl1, microtubule dynamics
Mis12, Ndc80, and Dam1 complexes, among many
others
Mitotic checkpoint Mad2, Bub3, BubR1, and Cdc20 Inhibit the APC/C until complete bipolar attachment of
complex (MCC) chromosomes to the mitotic spindle

APC/C, Anaphase-promoting complex/cyclosome; ATP, adenosine triphosphate; Cdk, cyclin-dependent kinase; CPC, chromosomal passenger complex; NIMA, never in mitosis,
gene A; SCF, Skp1–Cullin1–F-box.

at least two mammalian CAKs exist, the major CAK is a trimolecular Activation of the cyclin/Cdk complex is then dependent on the action
complex composed of Cdk7, cyclin H, and Mat1. The Cdk7/cyclinH/ of a dual-specificity phosphatase called Cdc25. Mammalian cells have
Mat1 complex also is required for the control of basal transcription three different Cdc25 proteins, called Cdc25a, Cdc25b, and Cdc25c,
via regulation of RNA polymerase II function.11,18 Second, when it which show some specificity for different cyclin/Cdk complexes and
is first formed, the cyclin/Cdk complex frequently is subject to inhibi- cell cycle stages.20
tory phosphorylation of Thr14 and Tyr15 residues within the Cdk’s Cdks are modulated by a series of Cdk inhibitors (CKIs) that play
active site by the Wee1 (Tyr15) and Myt1 (Thr14 and Tyr15) kinases.9,19 a key role in restricting the activity of the cyclin/Cdk complexes in
Control of the Cell Cycle  •  CHAPTER 4 59

response to either external signals or internal stresses.1,21 The CKIs regulates E2F through two distinct mechanisms. First, its association
can be divided into two distinct families based on their biological with E2F is sufficient to block the transcriptional activity of E2F.
properties. The first CKI family is named INK4, based on their roles Second, the pRB/E2F complex can recruit histone deacetylases to the
as INhibitors of Cdk4. The INK4 family has four members called promoters of E2F-responsive genes and thereby actively repress their
p16INK4a, p15INK4b, p18INK4c, and p19INK4d (encoded by the CDKN2A-D transcription. Cell cycle entry requires the phosphorylation of pRB
genes in humans). These INK4 proteins specifically prevent the binding by cyclin/Cdk complexes and the consequent dissociation of pRB
of cyclins to monomeric Cdk4 and Cdk6 but do not inhibit other from E2F.1,8,33
Cdks.13,21 The second CKI family is named Cip/Kip and includes Studies have identified eight E2F genes that encode nine different
three members: p21Cip1 (also called p21Waf1), p27Kip1, and p57Kip2 E2F proteins.34,35 Pocket proteins can regulate a subset of these factors:
(encoded by the CDKN1A-C genes in humans).21 These Cip/Kip E2F1, E2F2, E2F3a, E2F3b, E2F4, and E2F5. These E2F proteins
proteins have two major activities. First, they do not bind to monomeric associate with a dimerization partner, called DP, and the resulting
Cdks but associate with and inhibit the activity of cyclin/Cdk complexes complexes function primarily as either activators (E2F1, E2F2, and
already formed. Second, Cip/Kip proteins may promote the assembly E2F3a) or repressors (E2F4 and E2F5) of transcription under the
of cyclin D/Cdk4/6 complexes without dramatically perturbing its direction of the pocket proteins. Observations have suggested that
kinase activity.21,22 This activity is modulated by phosphorylation of several of these factors may act either as positive or negative regulators
Cip/Kip proteins by Src, Jak2, and Akt kinases,23–26 directly linking of transcription, depending on the cell type or the differentiation
Cdk regulation with the activity of these upstream mitogenic pathways. state.34,36–38 Most classic E2F target genes are regulated by the coor-
In addition to regulating the cell cycle, Cip/Kip proteins play important dinated action of these repressor and activator E2Fs.
roles in apoptosis, transcriptional regulation, cell fate determination,
cell migration, and cytoskeletal dynamics.27,28 Ubiquitin-Dependent Protein Degradation
The original observation that cyclin levels are tightly regulated during
Retinoblastoma Proteins and E2F Transcription Factors the cell cycle implies that these proteins are regulated not only at the
The retinoblastoma protein (pRB) was originally identified by virtue transcriptional level but also at the protein level. It is now evident
of its association with hereditary retinoblastoma.29 It behaves as a that ubiquitin-mediated protein degradation is a major regulatory
classic tumor suppressor: affected persons inherit a germline mutation mechanism to ensure ordered transition through the different phases
within one allele of the pRB-encoding gene, RB1, and loss of hetero- of the cell division cycle. Ubiquitylation depends on an enzymatic
zygosity is seen in all of the tumors. Subsequent studies showed that cascade, in which ubiquitin ligases recruit specific substrates for
the transforming ability of small DNA tumor viruses, including human modification. About 600 ubiquitin ligases are encoded by the human
papillomavirus, adenovirus, and simian virus, was dependent on the genome. Among them, the Skp1–Cullin1–F-box (SCF) and the
ability of virally encoded oncoproteins (E7, E1A, and SV40, respec- anaphase-promoting complex/cyclosome (APC/C) are known for
tively) to bind and inhibit pRB.30–32 Moreover, the RB1 gene is driving the degradation of cell cycle regulators to accomplish irreversible
inactivated in approximately one-third of all sporadic human tumors. cell cycle transitions.39–41 SCF has three core components: a RING
pRB and the pRB-related proteins p107 and p130, collectively finger protein, called Rbx1, which recruits the E2-ubiquitin conjugate;
known as the pocket proteins, are transcriptional repressors whose a cullin (Cul1); and Skp1 (Fig. 4.2). Skp1 acts to recruit a family of
major function is to inhibit the expression of cell cycle–related proteins proteins, called F-box proteins, which determine the target specificity
(see Table 4.1).33 This suppressive activity is largely dependent on the of the SCF complex. Once SCF binds its substrate, it transfers a
ability to prevent cell cycle entry through inhibition of the E2F ubiquitin molecule to lysine residues within the target protein to
transcription factors.33,34 The E2F proteins regulate the cell cycle– create a polyubiquitin chain, which targets the substrate to the protea-
dependent transcription of numerous targets, including core components some for degradation.42
of the cell cycle control (e.g., cyclin E and cyclin A) and DNA replica- The APC/C is a much larger complex, but it also contains a RING
tion (e.g., Cdc6, Cdt1, and the Mcm proteins) machineries.33–35 pRB finger protein, called Apc11, to recruit the E2-ubiquitin conjugate,

Apc11 E2
E2
Rbx1
Ubiquitin
Ubiquitin
Substrate
Substrate Apc2
P
Cul1 P
Activator
(Cdc20
F-box or Cdh1)
protein

Skp2

SCF ubiquitin ligase APC/C ubiquitin ligase

Figure 4.2  •  Ubiquitin ligases. The Skp1–Cullin1–F-box (SCF) and anaphase-promoting complex/cyclosome (APC/C) ubiquitin ligases play a key role in
enabling forward passage through key cell cycle transitions. These ligases are both large complexes that include three core components: a scaffolding protein
called a cullin in SCF, a protein that recruits the E2 and its associated ubiquitin molecule, and a specificity factor (called the F-box protein in SCF and the
activator in APC/C) that recruits the substrate. SCF and APC/C catalyze polyubiquitination of their substrates, which acts as a signal for substrate degradation
by the 26S proteasome. SCF has numerous substrates whose degradation promotes passage through the early stages of the cell cycle, including the cyclin-
dependent kinase inhibitor p27Kip1, cyclin E, E2F-1, and Cdt1. APC/C is essential for mitotic progression (by promoting degradation of securin and the
mitotic cyclins) and for cell cycle exit (by promoting degradation of multiple cell cycle regulators).
60 Part I: Science and Clinical Oncology

and a core cullin subunit (Apc2). In addition, APC/C is activated by centromeric region of chromosomes is determined by a complex
a cofactor that, in a manner comparable with that of the F-box proteins epigenetic, DNA sequence–independent mechanism.72–74 Kinetochores
of SCF, establishes substrate specificity (see Fig. 4.2).39 Cdc20 is the regulate the proper bipolar attachment between microtubules and
mitotic cofactor of APC/C, and it targets several cell cycle regulators chromosomes in order to distribute the replicated genome from a
(A- and B-type cyclins, Nek2, and securin) during mitotic entry and mother cell to its daughters.71,75 Given the relevance of the kinetochore
the metaphase-to-anaphase transition. Cdh1 (also known as FZR1 in in genome integrity, the composition of the kinetochore and the
mammals) replaces Cdc20 during mitotic exit and is the cofactor identification of various physical and functional modules within its
responsible for the elimination of many cell cycle regulators in the substructure is under deep investigation.76,77
G0 or G1 phase to prevent unscheduled DNA replication.43 Recent
studies have provided new insights into the intricate relationship Cell Cycle Phosphatases
between ubiquitylation and the cell division apparatus, including new Cdc14 phosphatases are the major Cdk-counteracting proteins in
roles for atypical ubiquitin chains, new mechanisms of regulation, yeast. Although two family members exist in mammals, their relevance
and extensive cross talk between ubiquitylation enzymes.44–49 in the cell cycle is not well understood.78,79 In eukaryotes, two major
complexes, PP1 and PP2A, account for more than 90% of protein
Mitotic Spindle and Mitotic Kinases and Kinesins phosphatase activity. In fact, these enzymes correspond to hundreds
In addition to the central role of Cdks in the cell cycle, many other of phosphatase complexes assembled from a few catalytic subunits
kinases play critical roles in cell cycle progression.3 Aurora and Polo (PP1α, PP1β/δ, and PP1γ1/2 for PP1, and the Cα and Cβ isoforms
kinases were first identified in genetic studies in flies as a result of for PP2A) and a diverse array of regulatory subunits.80 Recent evidence
their essential role in mitotic progression.50–52 Each of these families suggests that these protein families cooperate in the dephosphorylation
of kinases is represented by a single member in yeast, whereas three of most cell cycle kinase targets, including the retinoblastoma family
Aurora kinases (Aurora-A, Aurora-B, and Aurora-C) and five Polo-like or mitotic phosphoproteins.78,81–83 PP1 and PP2A are major phospha-
kinases (Plk1 through Plk5) exist in humans (see Table 4.1).52,53 tases responsible for pRB dephosphorylation during mitotic exit,
Aurora-A participates in several processes required for building a bipolar although the relative roles of these complexes or the particular
spindle, including centrosome separation and microtubule dynamics. holoenzymes involved are not clear.81,82 Similarly, both PP1 and PP2A
Aurora-A is activated by Tpx2, and the Aurora-A–Tpx2 holoenzyme are required for dephosphorylation of hundreds of mitotic proteins
may have critical implications in tumor development.51,52,54 Aurora-B, that are phosphorylated by Cdk1, as well as the other mitotic kinases.83,84
on the other hand, is part of the chromosome passenger complex Thus it has been suggested that the cell cycle ultimately is regulated
(CPC) that localizes to the kinetochores from prophase to metaphase by the dynamic equilibrium between Cdks (and partially by the other
and to the central spindle and midbody in cytokinesis. Other com- mitotic kinases) and PP1/PP2A activity. In the absence of Cdk activity,
ponents of the CPC include INCENP, survivin, and borealin, and the balance tilts in favor of the phosphatases. When Cdks are activated,
this complex regulates proper microtubule-kinetochore attachment phosphatase activity is overtaken.
and promotes biorientation during mitosis.51 Aurora-C may play similar Cdk1 is able to directly inhibit PP1 by direct phosphorylation of
roles to Aurora-B, although it is mostly expressed in germ cells and the catalytic subunit. The Cdk-dependent inhibition of PP2A, on the
may play a specific role in meiosis and during the first embryonic other hand, is not direct; rather, it is mediated by a new kinase known
cycles.55–57 Plk1 functions as a major regulator of centrosome matura- as Greatwall in flies and Xenopus or Mastl in mammals.85–87 Cdk1
tion, mitotic entry, and cytokinesis, whereas Plk4 is a critical regulator phosphorylates and activates Mastl, which in turn phosphorylates
of centriole duplication.50,58–60 The other members of the family, Plk2, Arpp-19 and Ensa, two highly related proteins that function as inhibitors
Plk3, and Plk5, are mostly involved in stress responses during interphase of a particular PP2A holoenzyme encompassing a regulatory subunit
or in neuron biology.3,53 of the B55 family.85–89 Reactivation of PP1 and PP2A phosphatases
A different group of additional cell cycle kinases with potential is a mandatory step for the exit from mitosis and the transition to
interest in tumor biology is the never in mitosis, gene A (NIMA)–related interphase.90–92
kinase family (Nek1 through Nek11). Four of these proteins, Nek2,
Nek6, Nek7, and Nek9, are involved in cell cycle progression, whereas Entry Into the Cell Cycle
all other family members are likely to play critical roles in cilia and
centrioles.3,61–63 Nek2, the closest relative to the Aspergillus NIMA Because most adult cells are quiescent, the mechanisms that determine
kinase, localizes to the centrosome and plays a role in establishing the their quiescent state and their reentry into the cell cycle with the
bipolar spindle by initiating the separation of centrosomes and appropriate stimuli are key determinants of tissue homeostasis. In
contributing to microtubule organization at the G2/M transition by quiescent cells, several DP/E2F complexes associate with the promoters
phosphorylating several centrosomal substrates. Nek2 also may play of E2F-responsive genes and recruit pRB-family members, along with
additional roles in chromosome condensation and the mitotic check- their associated histone deacetylases, to actively repress their transcrip-
point. Nek9, Nek6, and Nek7 function in a kinase cascade that tion.93,94 This repression machinery therefore prevents the expression
participates in centrosome separation and the formation and/or of proteins required for DNA synthesis and chromosome segregation.
maintenance of the mitotic spindle.61,63 In addition, CKIs normally are expressed in quiescent cells, preventing
A structurally different kinase, Haspin, is involved in the phos- the activation of Cdks.21 D-type cyclins are present at very low levels
phorylation of histone H3 on threonine 3 (H3T3).64,65 This kinase in most quiescent cells, in large part because they are phosphorylated
is activated by Cdk1 and Plk1,66 and phosphorylation of H3T3 is by an abundant kinase called Gsk3β and then exported to the cytoplasm
required for proper Aurora-B localization and activity and coupling for degradation.95
of mitotic chromosome structure with transcription.64,65,67 The transcription of D-type cyclins is induced in response to a
The activity of all these kinases is functionally linked to many wide variety of mitogenic stimuli.96,97 In addition, Gsk3β is inhibited
other proteins associated with chromosomes, microtubules, or different by mitogens, thus preventing the degradation of these cyclins. D-type
organelles, whose activity is essential for the structural changes associated cyclins then associate with Cdk4 and Cdk6, and the resulting complexes
with cell cycle progression and chromosome segregation.68 Among phosphorylate pRB proteins, partially inactivating their transcriptional
them, microtubule-associated proteins such as kinesin motor proteins suppressor function (Fig. 4.3).98–101 pRB inactivation causes the release
determine the dynamic behavior of the mitotic spindle required for of its associated DP-E2Fs. Repressive E2Fs such as E2F4 and E2F5
chromosome movement during mitosis.69,70 During mitosis, micro- dissociate from the DNA, and the free E2F complexes—DP-E2F1,
tubules associate with chromosomes through a large protein assembly DP-E2F2, and DP-E2F3—now occupy the promoters and activate
known as the kinetochore.71 The position of kinetochores in the their transcription. DP-E2F targets include many genes encoding
Control of the Cell Cycle  •  CHAPTER 4 61

Inhibitory Mitogens
growth factors

P P
Cip/Kip p107
INK4 p130 P
P P P
pRb pRb
P
E2F P
1,2,3 E2F
Cdk4/6 Cdk2 4,5
CycD CycE
p107 HDAC
p130 DNA synthesis
E2F E2F and mitotic
4,5 1,2,3
regulators
G0 /G1 G1/S
Transcriptional Transcriptional
repression activation

Figure 4.3  •  Entry into the cell cycle. The pocket proteins—pRB, p107, and p130—regulate a subset of the E2F family of proteins among many other
transcription factors. The pocket proteins bind to these E2Fs during the G0/early G1 phases, block their transcriptional activity, and recruit histone deacetylase
(HDAC), which comprises repressive complexes. Mitogenic signaling leads to the induction of D-type cyclins and the activation of interphase cyclin-dependent
kinase (Cdk) complexes, which phosphorylate the pocket proteins and release their associated E2Fs. This process allows the activating E2Fs to induce the
transcription of genes required for DNA synthesis and mitosis.

essential proteins required for DNA synthesis as well as mitosis.33–35


Through the control of pRB proteins, cyclin D-Cdk4/6 activity is a MCM MCM
central mediator of the reentry of cells into the cell cycle. Not surpris-
ingly, Cdk4/6 activity is frequently hyperactivated in cancer cells, as Cdc6 Cdt1
described later. Recent data suggest that Cdk6 may have kinase-
independent, transcriptional functions, although the exact mechanisms
behind these functions and their relevance in tumor development ORC
G1
deserve further investigation.102,103
Cyclin E is itself an E2F-responsive gene, and this regulation creates
a strong feed-forward loop. Cyclin E binds to Cdk2, and this complex
may promote further pRB inactivation.104,105 Cyclin E–Cdk2 also Cdc6
phosphorylates the CKI p27Kip1 on Thr187.106,107 This action creates MCM MCM
a high-affinity binding site for the SCF ubiquitin ligase bound to the Cdt1
F-box protein Skp2, leading to p27Kip1 degradation during the G1/S
transition.108 Finally, cyclin E-Cdk2 phosphorylates itself on multiple
DDK + Cdk
sites, creating a recognition site for SCF-Fbw7/Cdc4 and thereby dependent
ensuring its own destruction.109,110 The fact that lack of Cdk2 in the S
ORC
mouse does not result in defective mitotic cycles suggests that the
activity of this protein overlaps with other Cdks, with Cdk1 being
the best candidate.111–113 Indeed, Cdk1 is able to bind interphase
cyclins such as cyclin D and cyclin E, and it is sufficient for G1/S
C t
transition, at least in the absence of other interphase Cdks.114–115 Cdk2 1
is however an essential role in meiosis, although the molecular basis d
for this requirement is not fully understood.112,116
Figure 4.4  •  Origin licensing and firing. The origin replication complex
(ORC) associates with replication origins. During the G1 phase, Cdc6 and
DNA Replication Cdt1 are loaded on chromatin, and they in turn load the mini chromosome
maintenance (MCM) complex on chromatin, at which point licensing is
The DNA replication machinery is optimized to ensure that the genome considered complete, and the multiprotein complex is called the prereplication
is copied once—and only once—in each cell cycle.117 This optimization complex (pre-RC). Once cells pass the G1-to-S transition, this complex is
is achieved through a two-step process that first establishes a prereplica- activated to form the preinitiation complex (pre-IC), and DNA replication
tion complex (pre-RC) at each origin of replication, a process that is is initiated. Activation requires both cyclin-dependent kinase (Cdk) and
frequently referred to as origin licensing, and subsequently transforms Ddf4-dependent kinase (DDK) activity. It results in recruitment of numerous
pre-RCs into the preinitiation complex (pre-IC) that activates DNA proteins and activation of the MCM complex, which unwinds the DNA.
Subsequently, core components of the replication machinery, including DNA
replication (Fig. 4.4). These two steps occur at distinct stages of the polymerase α and DNA polymerase ε, are recruited to initiation sites. The
cell cycle to ensure that origins are licensed only once per cell cycle transition from pre-RC to pre-IC results in inhibition of Cdt1 by ubiquitin-
and rereplication cannot occur. Pre-RC formation takes place in the mediated degradation and geminin binding. Origin licensing cannot occur
initial steps of the cell cycle. The first event in this process is the again until activation of anaphase-promoting complex/cyclosome at the end
recruitment of the multiprotein complex called the origin recognition of mitosis allows accumulation of Cdt1.
62 Part I: Science and Clinical Oncology

complex to the origin DNA.118 The origin recognition complex recruits Mitosis
additional proteins including Cdc6, Cdt1, and finally the mini
chromosome maintenance (MCM) complex, a helicase that is required The mitotic machinery is optimized to ensure that the replicated
to unwind the DNA strands to form the pre-RC. Once cells enter S chromosomes are faithfully segregated to the daughter cells. This
phase, the transformation of the pre-RC to the pre-IC requires the segregation is achieved through the use of a specialized microtubule-
activity of two kinases: a Cdk and the Ddf4-dependent kinase, which based structure, the mitotic spindle, on which the original chromosomes
is composed of the Dbf4 regulatory subunit and the Cdc7 kinase.119,120 and their newly replicated copies, called sister chromatids, align and
The action of these kinases allows numerous additional proteins to then are partitioned to opposite poles of the cell. The mitotic spindle
associate with the pre-RC and form the pre-IC.121 Assembly of the is a highly dynamic structure that is maintained by many protein
pre-IC is thought to trigger DNA unwinding by the MCM complex, families, including motor molecules and other microtubule-associated
recruitment of the DNA polymerases, and initiation of the replication proteins.69,70,136,137 The appropriate side-by-side alignment of the sister
process, frequently called “origin firing.” chromatids, termed biorientation, is facilitated by the physical tethering
The transformation of the pre-RC to the pre-IC can occur at of the sister chromatids to one another. This process, called cohesion,
different time points in S phase, depending on whether the origin actually occurs in S phase in a manner that is coordinated with the
fires early or late.117 The system can tolerate this heterogeneity because replication process.138–140 Cohesin is mediated by four proteins that
the pre-RC is disassembled after firing and cannot reform until the together make up the cohesin complex. Two of these proteins, Smc1
subsequent cell cycle. This process occurs through several mechanisms. and Smc3, have a long coiled structure with a dimerization domain
The MCM complex travels with the replication fork in its role as the at one end that allows them to heterodimerize to form a V-like structure.
DNA helicase. Some evidence also indicates that phosphorylation of Important to note, the remaining ends of Smc1 and Smc3 can associate
Orc1 reduces its ability to bind to origins. Finally, and most important, with each another to form a functional adenosine triphosphate (ATP)
Cdt1 is prevented from participating in pre-RC formation outside of domain. This domain acts in an ATP-dependent manner to recruit
the G1 phase in two distinct ways. First, Cdt1 is marked for destruction two additional proteins, Scc1 and Scc3, which form a closed-ring
by ubiquitination.122 This process is mediated by SCF-Skp2 and structure that most likely encircles the chromosomes.141–143 Cohesin
particularly by an E4 ubiquitin ligase that includes Rbx1 (to recruit loading onto chromosomes, catalyzed by a separate complex called
the E2-ubiquitin), a cullin (Cul4), Ddb1, and Dtl/Cdt2 (the substrate kollerin, is thought to be mediated by the entry of DNA into cohesin
specificity factor).123–125 Important to note, this Cul4-Ddb1-Dtl/Cdt2 rings, whereas dissociation, catalyzed by Wapl and several other cohesin
complex functions independently of Cdt1 phosphorylation. Instead, subunits, is mediated by the subsequent exit of DNA.144 Increasing
Cdt1 is targeted only when proliferative cell nuclear antigen is present evidence indicates that cohesin participates in other cellular processes
on the DNA, which occurs primarily as a consequence of the initiation that involve DNA looping such as transcriptional regulation. Interesting
of DNA replication.126 Second, cells possess a protein called geminin to note, mutations in genes encoding cohesin subunits and other
that sequesters Cdt1 and prevents it from participating in pre-RC regulators of the complex have been identified in several tumor types.145
formation. Geminin is present specifically in S-, G2-, and early M-phase The related family of chromosomal proteins, condensins (condensin
cells. The APC/C ubiquitinates geminin and thereby triggers its I and condensin II), are formed from a conserved pair of Smc proteins
destruction during mitotic exit. This action creates a window between (Smc2 and Smc4) and distinct sets of non-SMC regulatory subunits.146
late mitosis and the end of G1 phase (when APC/C-Cdh1 is inactivated) Condensins participate in a diverse array of chromosomal functions
in which geminin is absent, and therefore Cdt1 is free to participate including chromosomal organization in interphase or the assembly
in pre-RC formation.127 of mitotic chromosomes.
The A-type cyclins are first transcribed late during the G1 phase
under the control of the E2F transcription factors in a similar manner Mitotic Entry
to that of cyclin E. Cyclin A associates with both Cdk2 and Cdk1 In addition to its role in DNA replication as discussed earlier, cyclin A/
and acts both during S-phase and G2/M.128,129 At the start of S Cdk complexes are critical mediators of the changes that occur during
phase, cyclin A–Cdk2 enters the nucleus and is specifically localized the G2 phase in preparation for mitosis.128 Here they are thought to
at nuclear replication foci, where it is thought to be actively involved play a key role in initiating the condensation of chromatin and also
in the firing of replication origins.130 As was described previously, might participate in the activation of the cyclin B/Cdk1 complexes.
cyclin A–Cdk2 also is required to phosphorylate E2F-1 and mediate Data have suggested that cyclin A2 is also crucial for loading of kinesins
its degradation, which is required to prevent E2F1 from triggering to microtubules, thus contributing to the formation of a functional
apoptosis.131,132 spindle.134 Cyclin A2 is destroyed at nuclear envelop breakdown in
Given the redundancy between different cyclin and Cdk family an APC/C-Cdc20–dependent manner.147 A-type cyclins are then
members, the specific role of cyclin A in DNA synthesis is unclear. substituted with B-type cyclins. Cyclin B1 protein accumulates steadily
Studies in mouse models identified a partially overlapping role between through the G2 phase and associates mainly with Cdk1, although it also
E- and A-type cyclins in the control of DNA synthesis in a cell-type may associate with Cdk2.129 The resulting cyclin B1/Cdk1 complex is
specific manner.133 Fibroblasts lacking both cyclin A1 and cyclin A2 mostly sequestered in the cytoplasm, and it is retained in an inactive
are able to proceed to S phase, and this is abrogated if E-type cyclins form throughout the G2 phase via the inhibitory phosphorylation
are deleted. However, A-type cyclins are essential in hematopoietic of Thr14 and Tyr15 in Cdk1’s active site by the Myt1 and Wee1
stem cells, suggesting cell-type differences probably caused by the kinases.148–150
levels of expression of the encoding genes. Although these observations Activation of cyclin B1–Cdk1 occurs in a highly synchronous
suggest overlapping functions for E- and A-type cyclins in the activation manner during G2-prophase transition (see Fig. 4.1).151 This activation
of Cdks, a new kinase-independent role as an RNA-binding protein is mediated by two changes. First, the activities of Myt1 and Wee1
has been recently proposed for cyclin A2.134 Cyclin A2 binds directly are downregulated at the transition between the G2 and M phases.
to the 3′ UTR of Mre11 mRNA in a Cdk-independent manner to Second, a dramatic increase in the activity of the Cdc25 phosphatases
promote its translation. Mre11 is a component of the MRN complex, occurs that relieves the inhibitory phosphorylation of Thr14 and
composed of Mre11, Rad50, and Nbs1, which plays a central role in Tyr15. These activity changes are triggered by the phosphorylation
DSB repair and replication fork restart.135 Depletion of cyclin A2 of Myt1, Wee1, Cdc25a, and Cdc25c. Three different kinases are
results in a reduction in Mre11 levels and defective repair of replication thought to contribute to this phosphorylation: Polo-like kinase
errors. Thus cyclin A2 participates in DNA synthesis in a Cdk- (Plk1), cyclin A–Cdk1, and cyclin B1–Cdk1 itself. The involvement
dependent manner while reinforcing stabilization of the key machinery of cyclin B1–Cdk1 creates a powerful feed-forward loop; once a
responsible for repairing DNA lesions caused by replication errors.134 small amount of cyclin B1–Cdk1 is activated, it simultaneously
Control of the Cell Cycle  •  CHAPTER 4 63

inactivates its own inhibitors and activates its activators, enabling the attachments of microtubules to the chromosomes, and possibly
a rapid transformation of the entire cyclin B1–Cdk1 pool from the the tension that is generated by microtubules on the kinetochores
inactive state to the active state. Once active, cyclin B1–Cdk1 phos- ensures that the sister chromatids are properly aligned at the metaphase
phorylates components of the centrosomes and initiates a process called plate.159,160 This process is the basis of one of several cell cycle check-
centrosome separation, in which the centrosomes move to opposing points, called the mitotic spindle assembly checkpoint (SAC), which
poles of the nascent spindle, an event essential for formation of the is described in more detail in the following sections.
mitotic spindle.68,152
The activation of mitotic kinases leads to dramatic changes in Anaphase
DNA structure. Largely on the basis of these morphologic changes, Anaphase is characterized by the segregation of the chromosomes.161
mitosis is divided into five different stages—prophase, prometaphase, This event is controlled by the mitotic ubiquitin ligase APC/C-Cdc20.
metaphase, anaphase, and telophase (Fig. 4.5)—before separation of APC/C-Cdc20 ubiquitinates, and thereby triggers the degradation of,
the daughter cells or cytokinesis. cyclin B1 and a protein called securin.39 Both securin and cyclin B1/
Cdk1 complexes are able to bind and inhibit a protease called sepa-
Prophase rase.162,163 APC/C-Cdc20 activity results in the degradation of cyclin
Prophase is characterized by condensation of sister chromatids— B and securin and the subsequent separase activation. Once released,
essentially packaging into a more compact chromatin structure. This separase cleaves the Scc1 component of the cohesin complex, which
process involves condensins I and II, which require phosphorylation opens the cohesin ring, unlinking the sister chromatids and allowing
by mitotic Cdks.145 During prophase, the nuclear envelope is still them to be pulled to opposite poles (see Fig. 4.5). The spindle poles
intact; consequently, differences in subcellular localization of the then move farther apart to ensure that the chromosomes are fully
condensin and Cdk complexes allow only condensin II and cyclin segregated. The separase-dependent cleavage of Scc1 also is essential
A-Cdk1 (nuclear), and not condensin I and cyclin B-Cdk1 (cytoplas- to link segregation of chromatids with the separation of centrioles
mic), to initiate condensation. In a parallel process called resolution, during mitotic exit.163 Cyclin B degradation results in the parallel
the sister chromatids are untangled via the action of topoisomerase inhibition of Cdk1 activity, thereby releasing the inhibitory mechanism
II.141,142 Resolution requires removal of the chromosome arm cohesin that limit PP1 and PP2A activity during the earlier phases of
through phosphorylation of Scc3 by Plk1 and histone H3 by the mitosis.84,90,92 The reactivation of these phosphatases results in the
Aurora-B kinase. Important to note, the cohesin complex at the massive dephosphorylation of mitotic phosphoproteins and results in
centromere is somehow protected from this modification by a protein the disassembly of the mitotic spindle, chromosome decondensation,
called shugosin (Sgo).153–155 and the reformation of the nuclear envelope.161
The second major event in prophase is the translocation of the During anaphase, Cdh1, which is inhibited by Cdk-dependent
cytoplasmic cyclin B1–Cdk1 to the nucleus, where it phosphorylates phosphorylation during mitosis, is dephosphorylated and replaces
components of the nuclear envelope and triggers its breakdown,68,156 Cdc20 as the main APC/C activator.39 APC/C-Cdh1 is responsible
which defines the transition from prophase to prometaphase. To prevent for the degradation of multiple cell cycle regulators, including Cdc20.
the activity of phosphatases induced by the mixture of cytoplasmic APC/C-Cdh1 also activates the ubiquitination and degradation of
and nuclear compartments, Mastl (which is mostly nuclear in prophase) geminin, allowing accumulation of Cdt1 for origin relicensing in the
is exported to the cytoplasm, thus inhibiting the Cdk-counteracting subsequent G1 phase, and the mitotic cyclins, allowing loss of Cdk
phosphatase PP2A-B55.68,157 Cdk1 phosphorylates more than 100 kinase activity. Loss of Cdh1 does not result in major abnormalities
proteins and participates in such activities as chromosome condensation, during mitotic exit but results in earlier entry into the following S
nuclear envelope breakdown, modifications in the Golgi apparatus, phase because of increased Cdk activity and DNA damage.164,165 Cdh1
and formation and dynamics of the mitotic spindle.9,68 Cdk1-deficient therefore is required to prevent unscheduled entry into S phase and
mouse embryos arrest during the first embryonic divisions, indicating genomic instability.43
that this kinase is absolutely required for mitosis and cannot be
compensated by other mammalian Cdks.115,158 Telophase
The reactivation of phosphatases during mitotic exit leads to the
Prometaphase dismantling of the mitotic spindle, leaving two discrete sets of
During prometaphase, the condensation process is accelerated because chromosomes with each nascent daughter cell.161 The elimination
condensin I and cyclin B-Cdk1 now have access to the DNA. The of mitotic phosphoresidues by these phosphatases also results in
sister chromatids become attached to spindle microtubules through DNA decondensation, and the nuclear envelope reforms around
the kinetochore, which is assembled onto centromeric DNA.71,72,74–76 the segregated chromosomes to create two new nuclei, an event that
Microtubules nucleated from the centrosomes attach to the kinetochore defines telophase.166
through a process called search and capture, in which individual
microtubules grow and shrink until they contact and bind the kin- Cytokinesis
teochore.73 Typically, one sister chromatid of the pair attaches first,
and this attachment is further stabilized through the recruitment of Finally, the cell undergoes cytokinesis, or cytoplasmic division. This
additional microtubules from the same pole of the mitotic spindle to process involves formation of a structure containing actin and myosin,
create a kinetochore fiber—that is, highly bundled microtubules bound called the contractile ring, on the inner face of the cell membrane.
to the kinetochore. The sister chromatids oscillate in the cell until The position of the contractile ring is carefully controlled. In cultured
the second sister chromatid is captured by microtubules emanating cells, the ring typically begins to form in anaphase, and its position
from the other pole. These oscillations continue until all of the is established by the position of the metaphase plate. As the membrane
chromosomes are properly aligned on the metaphase plate. grows, the contractile ring contracts steadily to form a constriction,
termed the “cleavage furrow,” which ultimately separates the two nuclei
Metaphase and forms the two daughter cells.167,168 This process partially depends
Metaphase is defined as the point at which all of the chromosome on several mitotic kinases such as Aurora-B and Plk1, which are
pairs are fully condensed, attached to the mitotic spindle, and aligned located at the midbody.51,52,58,169 Plk1 controls the activity of RhoA
at the center—termed the metaphase plate. The pulling of the kineto- guanosine triphosphatases, which are major regulators of the actomyosin
chore fibers toward the poles creates tension through the cohesin ring required for abscission.58,170 Aurora-B also ensures that abscission
complex at the kinetochores, which indicates that the sister chromatids does not occur in the presence of DNA bridges to avoid DNA damage
have achieved appropriate biorientation. The cell constantly monitors in a process known as the abscission checkpoint.168,171,172
64 Part I: Science and Clinical Oncology

Cyclin A/B-Cdk1 activity


Interphase

Chromatin begins to condense


Centrosomes move to poles
and mitotic spindle starts to form

NE Chromosomes attach to
breakdown microtubules of spindle
Prophase

Prometaphase

Chromosomes align at metaphase plate

Sister chromatids separate


Chromatin expands
Cytoplasm divides

APC
Metaphase

Cytokinesis
Anaphase

Telophase

Figure 4.5  •  Key stages of mitosis. As the parent cell enters prophase, the chromosomes begin to condense, and multiple proteins associate to form the
kinetochores. The centrosomes segregate to the poles to begin formation of the mitotic spindle. Nuclear envelope (NE) breakdown denotes the start of pro-
metaphase. In this phase, the sister chromatids continue to condense, and they attach to spindle microtubules via their kinetochores. During metaphase, the
sister chromatids align at the metaphase plate and eventually achieve appropriate biorientation. At the onset of anaphase, the sister chromatids separate and
move toward the poles of the spindle. During telophase, the two daughter nuclei are reformed and the daughter cells are finally separated by cytokinesis.
APC/C, Anaphase-promoting complex/cyclosome.
Control of the Cell Cycle  •  CHAPTER 4 65

CELL CYCLE CHECKPOINTS G1/S Checkpoint


At key transitions during eukaryotic cell cycle progression, signaling The molecular pathway that determines cell cycle entry with the
pathways monitor the successful completion of events in one phase appropriate mitogenic stimuli (previously described) is not considered
of the cell cycle before proceeding to the next phase. These regulatory a cell cycle checkpoint, strictly speaking. However, several of its
pathways are commonly referred to as cell cycle checkpoints.4 In a components also are used by a checkpoint that monitors DNA altera-
broader context, cell cycle checkpoints are signal transduction pathways tions before replication. In G1 cells, double-stranded DNA breaks
that link the rate of cell cycle phase transitions to the timely and (DSBs) are the most common and most deleterious type of DNA
accurate completion of prior dependent events. Checkpoint surveillance damage. The central components of the DNA damage response (DDR)
functions are not confined to monitoring normal cell cycle progression; are two members of the phosphoinositide 3-kinase–related kinase
they also are activated by both external and internal stress signals. family: ataxia telangiectasia mutated (ATM) and ATM- and rad3-related
The checkpoint pathways include sensor proteins that detect these (ATR).173 ATM originally was identified by virtue of its mutation in
lesions and simultaneously trigger two processes: they recruit additional a hereditary syndrome, ataxia-telangiectasia, which is associated with
effector complexes to correct the problems and activate signaling radiation hypersensitivity and cancer predisposition.174 ATR also is
pathways that induce a temporary cell cycle arrest. In certain situations, associated with a hereditary syndrome called Seckel syndrome.175 Early
which are determined by the cell type and the degree of damage, the studies suggested that ATM and ATR played distinct roles in the
checkpoint pathways eventually can induce permanent cell cycle arrest response to DSBs (ATM) versus replicative defects and single-stranded
(a process called senescence) or apoptosis. breaks (ATR). However, we now know that the regulation is more
To minimize the possibility of errors, checkpoints exist at the complex; considerable cross talk occurs between ATM and ATR, and
four different phases of the cell cycle: G1 (to prevent entry into S they share many mediators and effectors, but the precise composition
phase), intra S, G2 (to prevent mitosis), and M (to avoid mitotic and role of the DDR complexes vary depending on both the type of
exit with abnormal segregation of chromosomes) (see Fig. 4.1). In damage and the stage of the cell cycle (Fig. 4.6).176
general, these checkpoints monitor the status and structure of DNA These DSBs are recognized by the multifunctional Mre11-Rad50-
during cell cycle progression. In particular, cells scan the chromatin Nbs1 (MRN) complex.135,173 This complex recruits ATM to the site
for partially replicated DNA, as well as DNA strand breaks and other of damage. The active ATM then recruits proteins to modify the
DNA lesions that can result from both extrinsic (e.g., chemicals, chromatin at the region of the break and activate repair and signaling.
ionizing or ultraviolet radiation) and intrinsic (e.g., byproducts As a first step in this process, ATM phosphorylates histone H2AX to
of intracellular metabolism) DNA-damaging agents. In addition, form γH2AX, which helps hold the damaged ends together and acts
checkpoints also monitor the proper structure of chromosomes and as a binding platform for additional factors, including Mdc1, 53BP1,
their biorientation to ensure equal distribution between the two and Brca1, as well as more MRN and ATM. In contrast to the S and
daughter cells. G2 response, no recruitment of ATR to DSB in G1 cells occurs, and

G1 Intra S G2/M

DNA damage Replication- DNA damage


associated
error
DSB DSB

ssDNA
MRN RPA MRN

ATM ATR ATM

γH2AX ATM γH2AX ATR γH2AX

Mediators Mediators Mediators


repair repair repair
machinery machinery machinery

Chk2P Chk2P Chk1P Chk1P Chk2P

Figure 4.6  •  Ataxia telangiectasia mutated/ATM- and rad3-related (ATM/ATR) signaling is activated by DNA damage and replication stress. The cell
constantly monitors the chromatin for lesions, using complex signal transduction pathways that center on the ATM and ATR kinases. The precise mechanism
of response varies according to the type of DNA damage and the cell cycle stage. Double-stranded breaks (DSBs) are the most deleterious form of DNA
damage. DSBs are recognized by the Mre11-Rad50-Nbs1 (MRN) complex that consists of Mre11, Rad50, and Nbs1. This complex recruits ATM to the site
of damage. ATM phosphorylates histone H2AX to form γH2AX, which creates a binding platform for additional proteins that propagate the DNA damage
response and activate repair. For S and G2 phase cells, but not G1 phase cells, ATR also is recruited to the damage site. ATR and/or ATM signal to their
effector kinases (Chk1 and Chk2, respectively) to influence cell cycle progression as described in Fig. 4.7. Errors in DNA replication also can activate the
DNA damage response machinery through the presence of single-stranded DNA (ssDNA), which is a hallmark of the replication fork. The ssDNA is coated
with replication protein A (RPA) and bound by ATR. Active ATR then recruits the DNA damage and repair machinery, including ATM, leading to the
sequential activation of Chk1 and then Chk2.
66 Part I: Science and Clinical Oncology

G1, Intra S, G2/M Intra S G1/S


DNA Replicative Oncogenic
damage stress stress

P P p14ARF
Chk1 and/or Chk1 and
P P
Chk2 Chk2

Phosphorylation of Hdm2
all three Cdc25 proteins
Ubiquitination
P P Degradation
Cdc25a
p53 p53

P Bound and
Cdc25b Oligomerizes to
P inhibited
Cdc25c form active
by 14-3-3
transcription factor

Cdk activation p21Cip1 Pro-apoptotic


genes
Cdk activity

Figure 4.7  •  DNA damage, replicative stress, and oncogenic stress induce cell cycle arrest. DNA damage and replication stress lead to the rapid phosphorylation
and activation of the Chk1 and/or Chk2 kinases. These kinases enforce cell cycle arrest through two mechanisms. Chk1 and Chk2 both phosphorylate the
Cdc25 phosphatases, which triggers their ubiquitination and degradation (Cdc25a) or binding and inhibition by 14-3-3 (Cdc25b and Cdc25c), thereby preventing
activation of cyclin/Cdk kinase complexes. Chk1 and Chk2 also phosphorylate p53 and prevent it from being targeted by Hdm2 for ubiquitin-mediated degrada-
tion. As a result, p53 accumulates and activates transcription of p21Cip1, inhibiting Cdk2 and Cdk1 kinase complexes, or proapoptotic genes. Oncogenic stress
also leads to cell cycle arrest by activating replicative stress and/or inducing transcription or the p14ARF tumor suppressor and suppressing Hdm2-mediated
inhibition of p53. Cdk, Cyclin-dependent kinase.

thus ATM is solely responsible for checkpoint activation. The recruit- Important to note, p53 also is activated by other stress signals (see
ment of additional ATM amplifies the signal, and ATM acts via Fig. 4.7). In particular, it is now well established that numerous
phosphorylation and activation of the effector kinase Chk2.177,178 oncogenes trigger a stress response (called oncogene-induced stress)
Chk2 influences the G1 cell cycle arrest via two mechanisms (Fig. that leads to the activation of p53.188,189 The emerging view is that
4.7). First, it phosphorylates all three members of the Cdc25 family. this process occurs through two distinct mechanisms. First, oncogene
Phospho-Cdc25a is ubiquitinated by SCF-TrCPβ and degraded, whereas activation is thought to yield replicative stress that activates p53 via
phospho-Cdc25b and phospho-Cdc25c are bound and sequestered activation of Chk kinases and phosphorylation of Hdm2/Mdm2 as
by a cytoplasmic protein called 14-3-3.20,179 This process is a rapid just described.190,191 Second, many oncogenes activate transcription
response that can take effect within minutes after DNA damage, and of cell cycle inhibitors such as p16INK4a, p15INK4a, p21CIP1, or p19ARF
it has a widespread effect on cell cycle progression by preventing in a p53-independent manner.188,192–195 The protein p19ARF is encoded
activation of Cdks. Second, Chk2 phosphorylates p53, a critical regula- by the INK4A/ARF (CDKN2A) locus, and it actually shares two
tor of cell cycle checkpoints.180 In normal, nonstressed cells, p53 coding exons, which are read in alternate reading frames (hence the
protein is maintained at low steady state levels because it has a very name ARF) with the p16INKa tumor suppressor.194 The ARF protein
short half-life. This half-life is a result of rapid ubiquitination of p53 product, called p14ARF in humans and p19ARF in mice, binds to Hdm2/
by Hdm2 (the human ortholog of murine Mdm2 protein) and its Mdm2 and prevents it from regulating p53.196–199 As with the DDR,
consequent degradation.181,182 The importance of Mdm2 for mainte- this mechanism frees p53 to activate the transcription or proarrest or
nance of appropriate p53 levels in vivo is highlighted by the fact that proapoptotic targets.
absence of Mdm2 in knockout mice results in early embryonic lethality As an additional DDR in G1 cells, genotoxic agents also inhibit
that is rescued by a dual knockout of Mdm2 and p53.183,184 Phosphoryla- origin licensing by way of an ATM/ATR-independent process that is
tion of p53 by Chk2 is sufficient to prevent its association with Hdm2/ achieved through regulation of Cdt1.200 As described previously, Cdt1
Mdm2,183 which leads to an accumulation of p53, which functions is required for pre-RC formation. In an undamaged cell, Cdt1 is
as a transcriptional activator; p53 induces expression of many genes available during the G1 phase but is inhibited after origin firing by
involved in cell cycle arrest, including the CKI p21Cip1.185,186 This degradation (mediated by the SCF-Skp2 and Cul4-Ddb1-Dtl/Cdt2
p53-mediated arrest takes longer to develop than does the Cdc25 ubiquitin ligases) and geminin binding. As a key feature of this regula-
response (because it requires transcription and protein synthesis), but tory system, Cdt1 is completely resistant to Cul4-Ddb1-Dtl/Cdt2 in
it appears to be much more robust. Moreover, in addition to inducing the G1 phase. However, DNA damage allows the Cul4-Ddb1-Dtl/
cell cycle arrest, p53 has the capacity to induce apoptosis through the Cdt2 complex to ubiquitinate Cdt1 and induce its degradation.
transcriptional activation of proapoptotic regulators (e.g., the BH3-only Important to note, the degradation of Cdt1 is extremely rapid, occurring
proteins Puma and Noxa).187 within minutes of the DNA damage. Both Cdt1 and Cdt2 are
Control of the Cell Cycle  •  CHAPTER 4 67

phosphorylated in response to DNA damage, which results in the localize to the outer kinetochore and, in the absence of biorientation,
ubiquitination of Cdt1. This process depends on the activity of the prevent the Cdc20 activator from binding to the APC/C.
p97 AAA+-ATPase and its cofactor Ufd1, a complex required for the The kinetochore is made of approximately 30 scaffold proteins
extraction of ubiquinated proteins from the chromatin.124,201,202 As a that link chromatin and the mitotic spindle.71,72,75–77 Additional regula-
result, origin licensing is completely blocked until the damage is repaired tory elements include sensors of microtubule-kinetochore attachment
and Cdt1 is resynthesized. and a complex signaling pathway that modulates APC/C-Cdc20 activity.
Lack of tension or lack of attachment at the kinetochore results in
Intra–S Phase Checkpoint stable Mad1/Mad2 complexes that convert an inactive open-Mad2
conformation into a closed-Mad2 conformation that is able to bind
One of the major goals of cell cycle checkpoints is to prevent the to Cdc20.159,213,214 The Mad2-Cdc20 association triggers the recruitment
deleterious consequences of replicating damaged DNA. Therefore of BubR1-Bub3 into an APC/C-inhibitory complex (the mitotic
S-phase cells must respond virtually instantaneously to DNA damage checkpoint complex [MCC]). Because the closed-Mad2 signal is
to halt initiation of new replication forks throughout the S phase.203 diffusible, a single unattached kinetochore is sufficient to form these
The most deleterious type of damage is DSBs. DSBs can occur through complexes and inhibit APC/C-Cdc20 activity. As a result, separase is
the action of DNA-damaging agents (from either extrinsic or intrinsic inhibited by the high levels of securin and cyclin B/Cdk1 complexes,
sources) or as a consequence of the replication process itself—for being unable to cleave the centromeric cohesin (see Fig. 4.8). Once
example, if the replication fork passes through nicked DNA or if all chromosomes are bipolarly attached to the mitotic spindle, the
replication stalls at sites of DNA damage.204 The cell senses the damage SAC is satisfied and the Mad1/Mad2 complex is removed from the
in different ways, depending on whether the lesion is associated with kinetochores.71,77 How the checkpoint signaling is inhibited currently
replication. Ultimately, both ATM and ATR are recruited to the site is unclear. Cdc20 is now released from the MCC complex and activates
of damage, but the order of binding is different.176,203 Replication-linked the APC/C, leading to the rapid ubiquitination and degradation of
DSBs are distinguished by the presence of single-stranded DNA cyclin B and securin. Inactivation of these two proteins results in two
(ssDNA), a hallmark of the replication process. The ssDNA is coated major processes. First, lack of securin and cyclin B results in the
by replication protein A (RPA) and bound by ATR and its regulator activation of separase, a caspase-like protease that cleaves cohesin, the
subunit ATRIP, even during the normal replication process. In response molecule that holds sister chromatids together at the centromere.
to DNA damage, the ATR kinase is activated, and it then recruits a Second, inhibition of Cdk1, due to the lack of cyclin B, leads to the
variety of complexes that mediate both repair and checkpoint activation, activation of mitotic phosphatases such as PP1 and PP2A, triggering
including ATM. In contrast, nonreplication-associated DSBs initially mitotic exit as described earlier.161
recruit and activate ATM through the MRN-dependent process
described previously for the G1/S checkpoint. However, in S-phase CELL CYCLE DEREGULATION IN
cells, DSB resection causes the formation of ssDNA (through the HUMAN CANCERS
action of the MRN endonuclease), which is then bound by RPA and
ATR/ATRIP.203 Thus in S-phase cells, ATR and ATM jointly orchestrate The central relevance of cell cycle regulation in cancer is underscored
the DDR. ATR contributes to the checkpoint response in a similar by the finding that virtually all human tumors carry mutations in (1)
manner to ATM: it activates Chk1, which also can phosphorylate the the basic cell cycle machinery that controls entry into the cell cycle
Cdc25 proteins and p53.205–208 and (2) checkpoint regulators such as the p53 pathway (Table 4.2)
and by the observation that most human tumors display aberrant
G2 Checkpoint chromosome numbers.1,5,6,173,215 Together, the pRB and p53 pathways
are critical gatekeepers of cell cycle progression and stress response,
Whereas the G1/S and intra–S phase checkpoints prevent cells from and their function has crucial implications in the maintenance of
unfaithful replication, the G2 checkpoint is required to prevent the genome integrity at the level of both structural and numeric aberrations
passage of DNA lesions to the two daughter cells during mitosis.173,209,210 in chromosomes.216,217
DSBs are detected exactly as described previously for the S-phase
nonreplication-associated DSBs. Similarly, the ATR/Chk1 and ATM/ Unscheduled Cell Cycle Entry in Cancer
Chk2 pathways enforce arrest through inhibition of G2 and mitotic
Cdk complexes via the rapid removal of the Cdc25 phosphates and The alterations in the cell cycle machinery that occur most frequently
the p53-dependent induction of p21CIP1 to inhibit mitotic Cdk include loss or mutation of the pRB tumor suppressor; overexpression
complexes (cyclin A/B in combination with Cdk1/2 kinases).208,210,211 of cyclins, Cdks, and Cdc25 phosphatases; and loss of expression of
Ubiquitin ligases also are involved in this process. SCF-βTrCP regulates CKIs.1 Mutations that affect the pRB pathway have been identified
the levels of Cdc25, Claspin, and Wee1, whereas APC/C-Cdh1 is in most human cancers.1,29,217 The RB1 gene originally was identified
critical for the elimination of Plk1, a kinase essential for checkpoint by virtue of its mutation in both familial and sporadic retinoblastoma,
recovery.211,212 but it is defective in many other tumor types, especially osteosarcoma
and lung cancer.
Spindle Assembly Checkpoint In tumors that lack RB1 mutations, alterations in other components
of the signaling pathways that regulate pRB frequently are found,
The SAC acts to ensure that appropriate partitioning of the chromo- including cyclin overexpression or loss of CKIs. Nearly 50% of inva-
somes occurs during mitosis.159,160 The concept that chromosome sive breast cancers have elevated cyclin D expression compared with
segregation is prevented until all condensed sister chromatid pairs are surrounding normal breast epithelium, and in transgenic mice with
aligned at the metaphase plate with the appropriate biorientation has overexpression of human cyclin D1 or cyclin E in mammary gland
already been introduced. This process actually is controlled by a signal- cells, mammary adenocarcinomas develop.218–220 Similarly, Cdk4 and
ing network that constitutes the SAC (Fig. 4.8). The core components Cdk6 gene amplification occurs in breast cancers, sarcomas, gliomas,
of this checkpoint—called Mad1, Mad2, BubR1, Bub1, and Bub3 and melanomas, and specific translocations lead to cyclin D or Cdk6
in humans—originally were identified through screens in yeast for overexpression in hematopoietic disorders.1,221 Modifications of CKIs
“mitotic arrest deficient” (MAD) and “budding uninhibited by that act upstream of pRB activity also commonly are found in human
benzimidazole” (BUB) mutants.159 Other components of the checkpoint tumors. The CKI p27Kip1 often is aberrantly expressed in human
include the Mps1 kinase and the three subunits of the Rod, Zwich, breast cancer, and reduced p27Kip1 protein levels are correlated with
and ZW10 (RZZ) complex. During prometaphase, these proteins more aggressive breast tumors.222–224 Likewise, decreased expression
68 Part I: Science and Clinical Oncology

Prometaphase

No attachment
Pole
“Wait” Cohesion
Unattached
kinetochore Spindle
Low checkpoint
Mad2 proteins Cdc20
Metaphase High
Mad2

Attachment
Active Inactive
APC/C APC/C
Securin
ubiquitination
+ degradation
Anaphase
Securin
Inactive
Active separase Separase

Cohesion
(by Scc1
cleavage)

Figure 4.8  •  The spindle assembly checkpoint (SAC). Improper chromosome alignment on the mitotic spindle, disruption of microtubule dynamics, or
unattached kinetochores maintains the SAC in an active state. Checkpoint signaling is mediated by the Bub1, Bub3, BubR1, and Mad2 proteins, which
localize to kinetochores. These core spindle checkpoint regulators prevent Cdc20 from activating the anaphase-promoting complex/cyclosome (APC/C) and
therefore protect securin and cyclin B, two major APC/C-Cdc20 targets, from ubiquitin-mediated degradation. As a result, securin and cyclin B1-Cdk1 remain
bound to separase, which prevents cleavage of Scc1 and loss of centromeric cohesin. The “wait” signal is inhibited at the end of the metaphase by the appropriate
biorientation of the sister chromatids at the metaphase plate. The sensing mechanism involves detecting attachment or tension at the kinetochores that is
created by the pulling of the spindle fibers toward the poles. Mad2 then dissociates from the attached kinetochore, which allows Cdc20 to activate APC/C,
targeting securin and cyclin B for degradation. This process leads to the inactivation of Cdk1 and the activation of separase, triggering sister chromatid segregation
and mitotic exit.

of the CKI p57Kip2 is found in human bladder cancers. Although Mutations in p53 and Checkpoint Regulators
p21Cip1 is not commonly mutated in human cancer, its expression
is strongly reduced in multiple tumors as a consequence of defective The most frequently altered cell cycle checkpoint signaling molecule
p53 signaling.225 The CKI most frequently affected appears to be is the p53 tumor suppressor. The importance of p53-dependent signal-
p16INK4a; it was identified as a tumor suppressor that is associated ing in tumor suppression is underscored by the frequency of mutation
with familial melanoma, and it is inactivated by point mutation, (~50%) in sporadic tumors235 and the finding that germline mutations
deletion, and/or promoter methylation in approximately 30% of all of p53 result in Li-Fraumeni syndrome, a highly penetrant familial
human tumors.221,226 In contrast, point mutations in p15INK4b, p18INK4c, cancer syndrome that is associated with significantly increased rates
and p19INK4d are rare, but promoter methylation and reduced protein of brain tumors, breast cancers, and sarcomas.236 In human tumors
expression of some of these inhibitors have been seen in a variety that lack p53 gene mutation, p53 function may be disrupted by
of tumor types.1,221,227–229 Germline mutations in p16INK4a predispose alterations in cellular proteins that modulate the levels, localization,
individuals to melanoma, whereas deletion of p15INK4b and p16INK4a is and biochemical activity of p53. For example, in some tumors with
linked to the pathogenesis of lymphomas, mesotheliomas, and pancreatic wild-type p53 alleles, Mdm2 gene amplification occurs, resulting in
cancers.1,221 Although point mutations in Cdks are not common in Mdm2 protein overexpression and subsequent p53 inactivation.237 In
human tumors, a specific mutation of Cdk4 (R24C) that prevents its human papillomavirus–induced cervical carcinoma, p53 typically is
inhibition by INK4 proteins has been found in persons with familial not mutated; however, the human papillomavirus E6 protein binds
melanoma.230 Knockin mice harboring this mutation show not only p53 and targets it for degradation, abrogating p53-dependent
an increased susceptibility to melanoma but also the development signaling.238
of multiple tumor types, indicating the relevance of this regulatory Proteins that reside upstream of p53 (including ATM and Chk2)
circuit in human tumors.231,232 Both Cdc25a and Cdc25b phosphatases also are targeted for mutation in human tumors, and their discovery
also are overexpressed in more than 30% of primary breast tumors, and analysis have greatly deepened our insight into DDR signaling
40% to 60% of non–small cell lung cancers, 50% of head and neck pathways. ATM mutations occur in ataxia-telangiectasia, a disorder
tumors, and a significant fraction of non-Hodgkin lymphomas.20,233,234 in which patients have increased sensitivity to radiation and an
All these events can result in increased activation of Cdks, defec- elevated incidence of leukemias, lymphomas, and breast cancer.174,239
tive pRB signaling, unscheduled cell proliferation, and override of ATM-null mice exhibit growth retardation, neurologic dysfunction,
checkpoint arrest. infertility, defective lymphocyte maturation, and sensitivity to ionizing
Control of the Cell Cycle  •  CHAPTER 4 69

Table 4.2  Alteration of Cell Cycle Regulators in Human Tumorsa


Tumors Associated With Mutations or Hereditary Syndromes Associated With
Protein (Gene) Altered Expression Germline Mutations
ATM Breast carcinomas, lymphomas, leukemias Ataxia-telangiectasia
Aurora-A (AURKA) Wide array of tumors NR
Bub1 Colon, lung, and pancreatic cancer NR
BubR1 (BUB1B) Wilms tumor, rhabdomyosarcoma, and acute leukemia Mosaic variegated aneuploidy and premature chromatid
separation syndrome
Brca1/2 Breast and ovarian carcinoma Familial breast and ovarian cancer
Cdc25a Carcinomas of the breast, lung, head, and neck and lymphoma NR
Cdc25b Carcinomas of the breast, lung, head, and neck and lymphoma NR
Cdk4 Wide array of cancers Familial melanoma
Cdk6 Wide array of cancers NR
Chk1 Colorectal and endometrial carcinomas NR
Chk2 Carcinomas of the breast, lung, colon, urogenital tract, and testis Li-Fraumeni syndrome
Cyclin D1 Wide array of cancers NR
(CCND1)
Cyclin D2 Lymphoma and carcinomas of the colon, testis, and ovary NR
(CCND2)
Cyclin D3 Lymphoma, pancreatic carcinoma NR
(CCND3)
Cyclin E Wide array of cancers NR
(CCNE1/2)
Mdm2 (HDM2) Soft tissue tumors, osteosarcomas, esophageal carcinomas NR
Mps1 (TTK) Lung, gastric and bladder cancer NR
Mre11 Lymphoma Ataxia-telangiectasia–like disorder
Nbs1 Lymphomas, leukemias Nijmegen breakage syndrome
p15INK4b (CDKN2B) Wide array of cancers NR
p16INK4a (CDKN2A) Wide array of cancers Familial melanoma
p27Kip1 (CDKN1B) Wide array of cancers NR
p53 (TP53) Wide array of cancers Li-Fraumeni syndrome
p57Kip2 (CDKN1C) Bladder carcinomas NR
p130 (RBL2) Wide array of cancers NR
pRB (RB1) Wide array of cancers Familial retinoblastoma
SA1 (STAG2) Bladder, colorectal, glioblastoma, melanoma and Ewing’s sarcoma NR
Tpx2 Lung, bone, and pancreatic cancer NR

a
Only genetic alterations or defects that are present in more than 10% of primary tumors are represented.
NR, Not reported.

radiation.240,241 The DNA double-strand break repair gene Mre11 also for Nijmegen breakage syndrome, a rare autosomal-recessive disorder
is mutated in persons with an ataxia-telangiectasia–like disorder.242 characterized by chromosome instability, hypersensitivity to ionizing
Mutations of Chk2 and Chk1 also arise in human cancers. Chk2 radiation, and high susceptibility to the development of tumors.135,247
mutations have been reported in several cancers, including lung cancer,
whereas Chk1 mutations have been observed in human colon and Aneuploidy and Chromosomal Instability
endometrial cancers.243,244 In addition, heterozygous alteration of Chk2
occurs in a subset of persons with Li-Fraumeni syndrome who lack Abnormal chromosome numbers, called aneuploidy, is a frequent feature
p53 gene mutations.245 These findings support the theory that in of cancer cells.6,215,248 In addition, many cancer cells also display
human tumors in which p53 is intact, the function of this signaling chromosomal instability or an aberrantly high change in the karyotype.
pathway might be disrupted by alterations in cellular proteins that Chromosomal instability may lead to aneuploidy, but not all aneuploidy
modulate the levels or activity of p53. In addition, the breast cancer cells display chromosomal instability, because many cancer cells exhibit
susceptibility tumor suppressors Brca1 and Brca2 are known to stable aneuploidy karyotypes. Aneuploidy has a high rate of frequency
participate in the DDR and repair.246 Similarly, Fanconi anemia proteins, (more than 75%) in human tumors, and about one-quarter of the
which originally were identified by virtue of their association with a genome is affected by whole-arm or whole-chromosome number
recessive development disorder called Fanconi anemia, which is associ- alterations.249 Yet it is not clear whether aneuploidy is a cause or
ated with increased cancer predisposition (particularly acute myeloid consequence of cancer, and the relative level of chromosomal instability
leukemia), also function in the DDR.246 Mutations in the Nbs1 gene, may have either oncogenic or tumor suppressor properties during
a component of the MRN complex that sensors damage, are responsible tumor development.6,250–252
70 Part I: Science and Clinical Oncology

In vitro estimates suggest that normal cells missegregate a chromo- THERAPEUTIC MANIPULATION OF CELL
some every hundred cell divisions.215 This rate is dramatically increased CYCLE CONTROLS
in cells that display chromosomal instability, which missegregate a
chromosome in every one to three cell divisions in vitro.253 These Research during the past several decades has shown that alterations
defects occur for multiple reasons, including aberrant centrosome in cell cycle machinery and checkpoint signaling lead to tumorigenesis.
numbers, abnormal microtubule-kinetochore attachments, and These findings have important implications for the optimization of
imperfect SAC. In particular, there has been considerable speculation current therapeutic regimens and for the selection of novel cell cycle
that disruption of the SAC could occur during tumor progres- targets for the future development of anticancer agents. A leading
sion.252,254,255 Notably, inactivating mutations in BUB1 have been goal in cancer research is to identify compounds that will target key
identified in human colon carcinoma cell lines, which are known to cell cycle controls in a tumor-specific manner.
have a high degree of aneuploidy.256 These mutations facilitate the
transformation of cells that lack the breast cancer susceptibility gene Targeting Cyclin-Dependent Kinase Activity
BRCA2.257 The gene encoding BubR1, BUB1B, is also mutated in
persons with mosaic variegated aneuploidy and the premature chromatid Considerable debate has occurred about whether inhibition of Cdk
separation syndrome. Both BUB1 and BUB1B also are silenced by activity is a rational strategy for anticancer therapies.267 Cdk activity
promoter hypermethylation in specific tumors.258 Additional mutations frequently is elevated in human tumors, but it also is required to
and epigenetic alterations have been found in the SAC components maintain specific cell populations in adults (e.g., the hematopoietic
Mad1 and Mad2.258 Most of these alterations are loss-of-function compartment and gut) that are essential for viability.3 Thus the key
mutations, suggesting that the SAC is not functional in a variety of issue is whether tumor cells may require different Cdks for proliferation
tumors. Moreover, haploinsufficiency of Mad2 has been shown to or whether sufficient difference in the Cdk activity exists to create a
cause elevated rates of lung tumor development in mice.259 Interesting therapeutic window. During the last few years, the analysis of Cdk
to note, overexpression of Mad2 also results in increased tumor and cyclin mouse models has yielded considerable insight into this
susceptibility and frequent relapses of chromosomally unstable tumors.260 question but also has raised additional questions.268 These mouse
In fact, Mad2 is also frequently overexpressed in human tumors. models show that the cell cycle machinery is extremely robust; it
Several other mitotic regulators such as separase, securin, condensins, adapts easily to the loss of Cdks or cyclins by using other Cdks or
Cdc20, or Aurora kinases, as well as the SAC component Mps1, are cyclins to substitute for the missing activity. For example, cells are
included in the overexpression signature that marks chromosomally able to proliferate without specific interphase Cdks because novel
unstable cancers.258,261 All these data, together with the recent evidence cyclin/Cdk complexes can be formed that allow the cell cycle to
gathered in mouse models, suggest that small aberrations, either progress.111–115 This ability raises the possibility that tumor cells will
overexpression or downregulation, in the levels of mitotic and SAC develop resistance to Cdk-inhibitory drugs rapidly simply by adapting
regulators may contribute to aneuploidy and/or chromosomal instability their cell cycle machinery. On the positive side, studies in cell lines
in human tumors.6,255 Because functional disruption of SAC proteins and mouse models clearly show that tumors can be more dependent
results in the abrogation of the checkpoint and a failure to arrest in on Cdk activity, or at least a specific Cdk activity, than are normal
mitosis in the presence of microtubule poisons such as taxol and tissues. In parallel to these biologic studies, numerous small-molecule
nocodazole, these aberrations also may play a role in the response to inhibitors have been developed with different specificity profiles against
mitotic poisons currently used in the clinic.262 Cdks (Table 4.3).268–270
Multiple cell cycle aberrations, including pRB or p53 inactivation, Which Cdk should be targeted in each tumor type? Pioneering
lead to aberrant levels of mitotic regulators resulting in “oncogene- studies in the mouse showed that loss of D-type Cdk had little or no
induced mitotic stress.”216 The consequences of this aberration in effect on the development and maintenance of many tissues but can
cancer cell proliferation are possibly limited by additional alterations greatly suppress the development of certain tumor types, depending
in other mitotic regulators. For instance, the cancer-associated upregula- on the tissue and the identity of the initiating oncogenic lesions.271–274
tion of the SAC component Mad2 can be balanced by upregulation Mammary gland tumor proliferation dramatically depends on cyclin
of its target Cdc20, thus maintaining the balance between SAC and D1/Cdk4 complexes, whereas the absence of these molecules does
APC/C activity required for genomic stability.216 In the same line, not alter normal mammary gland development. In a pioneering study
mutations in the APC/C component Cdc27 may slow down APC/C in mouse models, cyclin D1–Cdk4 was shown to be required for
activity, thereby facilitating the correction of chromosome segregation Her2 (ErbB2)- or HRas-driven tumors, whereas it was dispensable
errors in the presence of the weak SAC frequently found in tumor for Myc- or Wnt-induced neoplasias, suggesting that the efficacy of
cells.263 Tumor cells also accumulate mutations in other genes whose CKIs may have a strong dependence on the genetic background of
deregulation may induce chromosomal instability, such as cohesin target tumors.272,275,276 Similarly, Cdk4 is required for KRAS-mutant
subunits and their regulators. However, whether chromosome misseg- lung tumors, but it seems dispensable for similar lung tumors with
regation is the major contribution of cohesin mutations to cancer wild-type KRAS alleles.274 On the other hand, cyclin D3/Cdk6
progression is not clear at present.146 complexes are critical for Notch1-mutant T-cell leukemias.277 These
Chromosome missegregation not only results in whole-chromosome studies and parallel efforts in human cancer cell lines led to multiple
aneuploidy but also may generate chromosome breaks and rearrange- clinical trials to test the effect of specific Cdk4/6 inhibitors in a wide
ments. A possible mechanism for DNA damage caused by chromosome spectrum of tumors.278 The success of some of these trials resulted in
missegregation is based on the defective DNA replication of the the rapid approval of palbociclib for the treatment of hormone-positive
micronuclei present in cells affected by aneuploidy.264 These micronuclei breast cancer, and other Cdk4/6 inhibitors such as abemaciclib and
undergo asynchronous DNA replication, resulting in DNA damage ribociclib have showed significant efficacy in advanced clinical trials
and pulverization of chromosomes. This process may provide, at least in multiple tumor subtypes.279–281
partially, an explanation for “chromothripsis,” a situation in which
chromosomes undergo massive local DNA breakage and rearrange- Targeting DNA Damage Response Proteins
ments.264,265 In addition, chromosomes that missegregate may be
damaged during cytokinesis, resulting in DNA double-strand breaks In the past decade, there has been a growing appreciation that many
that can lead to unbalanced translocations in the daughter cells.266 tumor cells carry mutations that disrupt their DDR. This characteristic
All these processes provide additional mechanisms by which defective is a major factor in establishing the resistance of tumors to chemo-
chromosome segregation may induce genomic aberrations during therapeutic agents, many of which work by causing DNA damage
tumor development. and triggering apoptosis through induction of DNA damage pathways.
Control of the Cell Cycle  •  CHAPTER 4 71

Table 4.3  Cell Cycle Regulators With Therapeutic Interest


Target Representative Small-Molecule Inhibitors Preclinical or Clinical Data
MITOTIC SPINDLE
Tubulin Stabilizing: taxanes, eleutherobins, epothilones, laulimalide, In clinical use for solid and hematopoietic tumors
sarcodictyins, and discodermolide; polymerization inhibitors:
vinca-alkaloids, cryptophycins, halichondrins, estramustine
Eg5 (KSP) ARRY-520, AZD4877, MK-0731, SB-715992 (ispinesib), Clinical trials in advanced solid tumors, lymphoma, and
SB-743921 taxane-resistant cancer
KINASES
ATR AZD6738, VX-970, ATR-101 Clinical trials in monotherapy or combination with
radiotherapy in solid tumors
Aurora kinases AT9283, CYC116, GSK1070916A, MLN8237, PF-03814735, VX-680 Clinical trials in solid tumors and hematopoietic malignancies
Cdks (1, 2, 4, 6) AG-024322, EM-1421, LEE-011 (ribociclib), LY2835219 Clinical trials in solid tumors and leukemias; Cdk4/6 inhibitors
(abemaciclib), P276-00, PD-0332991 (palbociclib) approved for hormone-positive breast cancer
Chk1 LY2606368, UCN01, CCT245737 Clinical trials in combination with DNA damaging agents
Plk1 BI2536, BI6727 (volasertib), GSK461364, NMS-1286937, Clinical trials in solid tumors and hematopoietic malignancies
TKM-080301
Mps1 (TTK) NMS-P153, BAY1161909, NTRC 0066-0 Clinical trials in metastatic solid tumors
PHOSPHATASES
Cdc25 ARQ-501, IRC 083864 Preclinical studies and clinical trials in advanced solid tumors
DNA REPAIR
PARP ABT-888, AZD-2281 (olaparib), AG014699, BMN-673, CEP 9722, Clinical trials in solid tumors and hematopoietic malignancies;
MK-4827 (niraparib) olaparib and rucaparib are approved for treating ovarian
cancer
UBIQUITIN LIGASES
APC/C TAME, APCIN Preclinical studies

APC/C, Anaphase-promoting complex/cyclosome; Cdk, cyclin-dependent kinase; ATR, ATM- and rad3-related; PARP, poly (ADP-ribose) polymerase.

Therefore considerable attention has focused on designing cancer Targeting the Mitotic Spindle
treatments that would be effective in cells with an impaired DDR.
Because it is difficult to restore the function of mutant or missing Microtubule poisons, such as taxol and vinblastine, kill cancer cells
proteins, the prevailing strategy is to identify drugs that would synergize at least partially by exploiting their effects on the mitotic spindle.292
with the defective DDR to selectively kill the tumor cells and not the Taxanes, such as docetaxel or paclitaxel, are microtubule-stabilizing
normal cells. For example, inhibitors of poly (adenosine diphosphate drugs widely used to treat breast and ovarian tumors, non–small cell
[ADP]-ribose) polymerase (PARP) selectively kill cells that lack either lung cancer, and Kaposi sarcoma. Vinca alkaloids, such as vinblastine
Brca1 or Brca2.282,283 The rationale for this action is that these proteins or vincristine, are microtubule-destabilizing compounds and have
provide two alternative repair mechanisms in response to DNA damage: shown clinical efficacy against a broad range of hematologic malignan-
homologous recombination (Brca1 and Brca2) and base excision repair cies. Both classes of drugs bind tubulin and inhibit microtubule
(PARP). Therefore loss of one but not both of these pathways can be dynamics, impairing the formation of a functional spindle. The SAC
tolerated. As a second example, inhibition of Chk1 sensitizes p53 senses lack of proper attachment of kinetochores and arrests cells in
mutant cells to DNA damage.284 Because p53 is mutated in approxi- prometaphase in the presence of these compounds. Thus inhibition
mately half of all human tumors and the absence of p53 is a major of spindle dynamics results in abnormal chromosome segregation that
predictor of poor response to classic chemotherapeutic agents, consider- frequently results in either aneuploidy or cell death. The low mitotic
able efforts are being made to develop small-molecular inhibitors of index in human tumors suggests that microtubule poisons may have
Chk1. Although the initial model proposed that this effect was due therapeutic potential in a mitotic-independent manner although this
to the simultaneous abrogation of the G2 (Chk1) and G1 (p53) deserves further exploration.293 About 30 microtubule poisons are
checkpoints, new evidence suggests that the toxicity of Chk1 inhibitors currently in clinical development.
may be due to the generation of replicative stress (RS) in cells, with The success of these molecules in the clinic led to the search for
the less restrictive S-phase entry due to the lack of p53.285 Chk1 additional compounds that target specific regulators of microtubule
inhibitors therefore might synergize with other mutations that promote dynamics rather than tubulin itself. In a pioneer screening, monastrol
a promiscuous S-phase entry. A similar rationale applies to other was identified as an inhibitor of the kinesin Eg5, a protein required
molecules that target the RS response, such as inhibitors of the Chk1- for centrosome separation and the formation of a bipolar spindle.294
activating kinase ATR. Indeed, ATR inhibitors display a selective New drugs have been characterized that result in similar defects by
effect in Myc-driven tumors that display high levels of RS as a con- inhibiting CenpE, another kinesin with critical roles in microtubule-
sequence of the overexpression of Myc.286 This strategy can be general- to-kinetochore attachment.295–297 Inhibition of Aurora-A or Plk1 also
ized to multiple oncogenic alterations that result in cell cycle defects, may be considered to be an antispindle strategy because these kinases,
causing an increased reliance on ATR checkpoint activity, as recently although involved in other mitotic processes, are essential for centrosome
shown for different tumor types.287–291 maturation and separation and the formation of a bipolar spindle.52,295
72 Part I: Science and Clinical Oncology

In general, the inhibition of all these molecules results in an SAC- prevents mitotic arrest in the presence of microtubule poisons and
dependent arrest that impairs proliferation or viability of targeted therefore generates aberrant cells.52,305,306 It has been shown that the
cells. The Plk1 inhibitor volasertib has shown considerable promise reduction in these checkpoint proteins makes tumor cells more sensitive
in clinical studies, having reached phase III trials and been granted than untransformed cells to low doses of spindle poisons.307 The efficacy
the breakthrough therapy designation for its effects in acute myeloid of several Aurora or Mps1 small-molecule inhibitors currently is being
leukemia.298 tested in preclinical or clinical assays (see Table 4.3).52,295,306,308
Aneuploidy is a hallmark of cancer and is now considered as a
Targeting Mitotic Entry and Exit highly attractive therapeutic target.215,309 Most tumor cells are aneuploid,
whereas this abnormality is infrequent (although this has not been
As indicated in the earlier sections of this chapter, multiple enzymatic precisely established) in wild-type cells. This specificity is especially
activities are required for mitosis. Cdk1 is the major engine in this interesting given the problems that most therapeutic strategies that
process, and its activity is essential for mitosis. However, it has not target mitosis have in specifically inhibiting tumor cells. Aneuploid
been considered as a major target because its inhibition may have cells display specific defects in cell cycle kinetics, growth rate, metabo-
strong undesired effects.270 Yet some evidence suggests that it may be lism, and the response to specific stresses.310,311 These defects can be
an interesting target in specific situations. Cdk1 is specifically required exploited by targeting specific cellular pathways that protect cells from
in cells transformed with Myc but not in cells transformed by other the deleterious effects of aneuploidy. For instance, inhibiting the
oncogenes. Inhibition of Cdk1 rapidly downregulates survivin expres- proteotoxic and metabolic stress pathways specifically reduces the
sion, a protein required to avoid apoptosis in the presence of an excess viability of aneuploidy cells because of the unbalanced load of proteins
of Myc. Cdk1 inhibition therefore results in Myc-dependent apoptosis in these cells.312 As previously described, targeting the mitotic checkpoint
and regression of Myc-dependent lymphoma and hepatoblastoma may increase the levels of aneuploidy in the tumor cells, which opens
tumors.299 Cdk1 also is implicated in DNA repair by homologous the opportunity to combine different types of antimitotic strategies
recombination, and its inhibition results in impaired Brca1 activity. to further induce chromosomal instability and aneuploidy and inhibit
Cdk1 inhibition synergizes with PARP inhibitors, and partial inhibition the pathways that cancer cells use to tolerate this abnormal state.309
of Cdk1 therefore may sensitize Brca1/2-proficient cancer cells to
inhibit PARP, suggesting specific applications of Cdk1-targeting SUMMARY
compounds in cancer cells.300 Cdk1 activators such as Cdc25 phos-
phatases are additional targets whose inhibition results in impaired During the past several decades, investigators have uncovered a wealth
Cdk activity and cell cycle arrest.20 The therapeutic usefulness of of information about the proteins that control the division of human
inhibition of additional mitotic kinases such as Mastl, Nek proteins, cells. A key finding is that deregulation of the cell cycle machinery and/
or Haspin, among others, is currently undergoing preclinical or its checkpoints is a universal alteration in human cancer.1,2 Because
evaluation.3,295,301 the basic machinery that controls the cell cycle is similar in all cell
Inhibiting mitotic entry or progression or preventing spindle types, the hope is that common strategies will be developed against
dynamics may result in different outcomes, including cell death or a wide variety of cancers. Even though several of the currently used
the exit from the cell cycle without chromosome segregation.295 In anticancer therapies target nonselective and non–mechanism-based
fact, a rapid exit from mitosis is a major resistance mechanism that targets, their effectiveness, albeit limited in many cases, is likely due
generates viable cells in the presence of mitotic poisons. This finding to the fact that they ultimately target cell cycle regulatory or DDR
led to the evaluation of mitotic exit pathways as new targets for signaling pathways, the status of which is different in normal cells
therapy.302 In fact, genetic ablation of APC/C-Cdc20 completely versus tumor cells. Identification of all the components of the cellular
prevents mitotic exit, and these mutant cells arrest in mitosis until machinery that control the cell cycle both positively and negatively
they die.90 This action results in complete tumor repression in mouse is vital to the continued development of anticancer agents that can
models, and a first generation of APC/C inhibitors is currently available preferentially eliminate cancer cells and minimize the toxicity to normal
to test this strategy in preclinical studies (see Table 4.3).90,303,304 tissues. The complexity of the human genome makes this task difficult
because many members of the major protein families that regulate the
Targeting the Spindle Assembly Checkpoint cell cycle have not been studied thus far. In addition, our knowledge of
and Aneuploidy the in vivo relevance of these proteins in different tissues is limited. It is
obvious from the mouse models studied that the efficacy of inhibiting
Similarly to what was discussed for the DNA damage checkpoint, a specific cell cycle targets depends on the tumor type and the oncogenic
different concept is provided by the use of SAC inhibitors (checkpoint environment in each specific tumor. As our understanding of cell cycle
abrogation) to increase instability in cancer cells. Checkpoint kinases regulation and checkpoint signaling improves, the goal is to use this
such as Mps1 (also known as TTK) and Aurora-B (not considered to knowledge in the design of mechanism-based therapeutics that will
be a “core” checkpoint protein but involved in an error-correction bring anticancer therapy to a new level. There can be little doubt of
mechanism) are required for the proper bipolar spindle attachment the value of targeting the cell cycle in drug discovery.
of chromosomes.3 Inhibition of these kinases results in rapid exit from
mitosis without chromosome segregation, generating tetraploid or The complete reference list is available online at
aneuploid cells. Important to note, inhibition of these molecules ExpertConsult.com.

KEY REFERENCES
1. Malumbres M, Barbacid M. To cycle or not to 3. Malumbres M. Physiological relevance of cell cycle 5. Kastan MB, Bartek J. Cell-cycle checkpoints and
cycle: a critical decision in cancer. Nat Rev Cancer. kinases. Physiol Rev. 2011;91:973–1007. cancer. Nature. 2004;432:316–323.
2001;1:222–231. 4. Hartwell LH, Weinert TA. Checkpoints: controls 6. Holland AJ, Cleveland DW. Boveri revisited: chro-
2. Hartwell LH, Kastan MB. Cell cycle control and that ensure the order of cell cycle events. Science. mosomal instability, aneuploidy and tumorigenesis.
cancer. Science. 1994;266:1821–1828. 1989;246:629–634. Nat Rev Mol Cell Biol. 2009;10:478–487.
Control of the Cell Cycle  •  CHAPTER 4 73

8. Morgan DO. Cyclin-dependent kinases: engines, 115. Santamaria D, Barriere C, Cerqueira A, et al. Cdk1 is 209. Branzei D, Foiani M. Regulation of DNA repair
clocks, and microprocessors. Annu Rev Cell Dev Biol. sufficient to drive the mammalian cell cycle. Nature. throughout the cell cycle. Nat Rev Mol Cell Biol.
1997;13:261–291. 2007;448:811–815. 2008;9:297–308.
9. Malumbres M, Barbacid M. Mammalian 117. Bell SP, Dutta A. DNA replication in eukaryotic 210. Lobrich M, Jeggo PA. The impact of a negligent
cyclin-dependent kinases. Trends Biochem Sci. cells. Annu Rev Biochem. 2002;71:333–374. G2/M checkpoint on genomic instability and cancer
2005;30:630–641. 144. Nasmyth K. Cohesin: a catenase with separate entry induction. Nat Rev Cancer. 2007;7:861–869.
20. Boutros R, Lobjois V, Ducommun B. CDC25 and exit gates? Nat Cell Biol. 2011;13:1170–1177. 215. Gordon DJ, Resio B, Pellman D. Causes and
phosphatases in cancer cells: key players? Good 145. Losada A. Cohesin in cancer: chromosome segrega- consequences of aneuploidy in cancer. Nat Rev
targets? Nat Rev Cancer. 2007;7:495–507. tion and beyond. Nat Rev Cancer. 2014;14:389– Genet. 2012;13:189–203.
32. Dyson NJ. RB1: a prototype tumor suppressor and 393. 246. Wang W. Emergence of a DNA-damage response
an enigma. Genes Dev. 2016;30:1492–1502. 152. Lindqvist A, Rodríguez-Bravo V, Medema RH. network consisting of Fanconi anaemia and BRCA
34. Chen HZ, Tsai SY, Leone G. Emerging roles of The decision to enter mitosis: feedback and proteins. Nat Rev Genet. 2007;8:735–748.
E2Fs in cancer: an exit from cell cycle control. Nat redundancy in the mitotic entry network. J Cell 248. Kops GJ, Weaver BA, Cleveland DW. On the road to
Rev Cancer. 2009;9:785–797. Biol. 2009;185:193–202. cancer: aneuploidy and the mitotic checkpoint. Nat
39. Peters JM. The anaphase promoting complex/ 159. Musacchio A, Salmon ED. The spindle-assembly Rev Cancer. 2005;5:773–785.
cyclosome: a machine designed to destroy. Nat Rev checkpoint in space and time. Nat Rev Mol Cell 265. Stephens PJ, Greenman CD, Fu B, et al. Massive
Mol Cell Biol. 2006;7:644–656. Biol. 2007;8:379–393. genomic rearrangement acquired in a single
40. Silverman JS, Skaar JR, Pagano M. SCF ubiquitin 160. Sacristan C, Kops GJ. Joined at the hip: kinetochores, catastrophic event during cancer development. Cell.
ligases in the maintenance of genome stability. Trends microtubules, and spindle assembly checkpoint 2011;144:27–40.
Biochem Sci. 2012;37:66–73. signaling. Trends Cell Biol. 2015;25:21–28. 268. Malumbres M, Barbacid M. Cell cycle, CDKs
45. Alfieri C, Chang L, Zhang Z, Yang J, Maslen S, 161. Sullivan M, Morgan DO. Finishing mitosis, one and cancer: a changing paradigm. Nat Rev Cancer.
Skehel M, et al. Molecular basis of APC/C regula- step at a time. Nat Rev Mol Cell Biol. 2007;8:894– 2009;9:153–166.
tion by the spindle assembly checkpoint. Nature. 903. 279. Asghar U, Witkiewicz AK, Turner NC, Knudsen ES.
2016;536:431–436. 168. Mierzwa B, Gerlich DW. Cytokinetic abscission: The history and future of targeting cyclin-dependent
52. Lens SM, Voest EE, Medema RH. Shared and molecular mechanisms and temporal control. Dev kinases in cancer therapy. Nat Rev Drug Discov.
separate functions of polo-like kinases and aurora Cell. 2014;31:525–538. 2015;14:130–146.
kinases in cancer. Nat Rev Cancer. 2010;10:825– 172. Nähse V, Christ L, Stenmark H, Campsteijn C. 280. Finn RS, Martin M, Rugo HS, Jones S, Im
841. The abscission checkpoint: making it to the final SA, Gelmon K, et al. Palbociclib and Letrozole
70. Cross RA, McAinsh A. Prime movers: the mecha- cut. Trends Cell Biol. 2017;27:1–11. in Advanced Breast Cancer. N Engl J Med.
nochemistry of mitotic kinesins. Nat Rev Mol Cell 173. Bartek J, Lukas J. DNA damage checkpoints: from 2016;375:1925–1936.
Biol. 2014;15:257–271. initiation to recovery or adaptation. Curr Opin Cell 283. Bryant HE, Schultz N, Thomas HD, et al.
76. Etemad B, Kops GJ. Attachment issues: kinetochore Biol. 2007;19:238–245. Specific killing of BRCA2-deficient tumours with
transformations and spindle checkpoint silencing. 189. Collado M, Serrano M. Senescence in tumours: inhibitors of poly(ADP-ribose) polymerase. Nature.
Curr Opin Cell Biol. 2016;39:101–108. evidence from mice and humans. Nat Rev Cancer. 2005;434:913–917.
77. Weir JR, Faesen AC, Klare K, Petrovic A, Basilico 2010;10:51–57. 284. Garrett MD, Collins I. Anticancer therapy with
F, Fischböck J, et al. Insights from biochemical 190. Bartkova J, Rezaei N, Liontos M, et al. Oncogene- checkpoint inhibitors: what, where and when? Trends
reconstitution into the architecture of human induced senescence is part of the tumorigenesis Pharmacol Sci. 2011;32:308–316.
kinetochores. Nature. 2016;537:249–253. barrier imposed by DNA damage checkpoints. 292. Dumontet C, Jordan MA. Microtubule-binding
78. Wurzenberger C, Gerlich DW. Phosphatases: Nature. 2006;444:633–637. agents: a dynamic field of cancer therapeutics. Nat
providing safe passage through mitotic exit. Nat 191. Di Micco R, Fumagalli M, Cicalese A, et al. Rev Drug Discov. 2010;9:790–803.
Rev Mol Cell Biol. 2011;12:469–482. Oncogene-induced senescence is a DNA damage 306. Dominguez-Brauer C, Thu KL, Mason JM, Blaser
83. Qian J, Winkler C, Bollen M. 4D-networking response triggered by DNA hyper-replication. H, Bray MR, Mak TW. Targeting Mitosis in Cancer:
by mitotic phosphatases. Curr Opin Cell Biol. Nature. 2006;444:638–642. Emerging Strategies. Mol Cell. 2015;60:524–536.
2013;25:697–703. 204. Muñoz S, Méndez J. DNA replication stress: 312. Sheltzer JM, Amon A. The aneuploidy paradox:
87. Glover DM. The overlooked greatwall: a new perspec- from molecular mechanisms to human disease. costs and benefits of an incorrect karyotype. Trends
tive on mitotic control. Open Biol. 2012;2:120023. Chromosoma. 2016;doi:10.1007/s00412-016-0573-x. Genet. 2011;27:446–453.

ADDITIONAL RESOURCES
Chromosome aberrations in cancer: http://cgap.nci.nih.gov/
Chromosomes/Mitelman.
Clinical trials: http://www.clinicaltrials.gov/.
p53 Databases: http://www-p53.iarc.fr/; http://p53.free.fr/
Database/p53_database.html.
Control of the Cell Cycle  •  CHAPTER 4 73.e1
73.e1

REFERENCES
1. Malumbres M, Barbacid M. To cycle or not to 27. Besson A, Assoian RK, Roberts JM. Regulation of 51. Carmena M, Earnshaw WC. The cellular geog-
cycle: a critical decision in cancer. Nat Rev Cancer. the cytoskeleton: an oncogenic function for CDK raphy of aurora kinases. Nat Rev Mol Cell Biol.
2001;1:222–231. inhibitors? Nat Rev Cancer. 2004;4:948–955. 2003;4:842–854.
2. Hartwell LH, Kastan MB. Cell cycle control and 28. Besson A, Dowdy SF, Roberts JM. CDK inhibi- 52. Lens SM, Voest EE, Medema RH. Shared and
cancer. Science. 1994;266:1821–1828. tors: cell cycle regulators and beyond. Dev Cell. separate functions of polo-like kinases and aurora
3. Malumbres M. Physiological relevance of cell cycle 2008;14:159–169. kinases in cancer. Nat Rev Cancer. 2010;10:825–841.
kinases. Physiol Rev. 2011;91:973–1007. 29. Weinberg RA. The retinoblastoma gene and gene 53. de Carcer G, Manning G, Malumbres M. From Plk1
4. Hartwell LH, Weinert TA. Checkpoints: controls product. Cancer Surv. 1992;12:43–57. to Plk5: functional evolution of polo-like kinases.
that ensure the order of cell cycle events. Science. 30. Helt A-M, Galloway DA. Mechanisms by which Cell Cycle. 2011;10:2255–2262.
1989;246:629–634. DNA tumor virus oncoproteins target the Rb family 54. Asteriti IA, Rensen WM, Lindon C, et al. The
5. Kastan MB, Bartek J. Cell-cycle checkpoints and of pocket proteins. Carcinogenesis. 2003;24:159–169. Aurora-A/TPX2 complex: a novel oncogenic holo-
cancer. Nature. 2004;432:316–323. 31. DeCaprio JA. How the Rb tumor suppressor enzyme? Biochim Biophys Acta. 2010;1806:230–239.
6. Holland AJ, Cleveland DW. Boveri revisited: chro- structure and function was revealed by the study of 55. Yang KT, Li SK, Chang CC, et al. Aurora-C kinase
mosomal instability, aneuploidy and tumorigenesis. Adenovirus and SV40. Virology. 2009;384:274–284. deficiency causes cytokinesis failure in meiosis I and
Nat Rev Mol Cell Biol. 2009;10:478–487. 32. Dyson NJ. RB1: a prototype tumor suppressor and production of large polyploid oocytes in mice. Mol
7. Salazar-Roa M, Malumbres M. Fueling the cell an enigma. Genes Dev. 2016;30:1492–1502. Biol Cell. 2010;21:2371–2383.
division cycle. Trends Cell Biol. 2017;27:69–81. 33. van den Heuvel S, Dyson NJ. Conserved functions 56. Fernandez-Miranda G, Trakala M, Martin J, et al.
8. Morgan DO. Cyclin-dependent kinases: engines, of the pRB and E2F families. Nat Rev Mol Cell Biol. Genetic disruption of aurora B uncovers an essential
clocks, and microprocessors. Annu Rev Cell Dev Biol. 2008;9:713–724. role for aurora C during early mammalian develop-
1997;13:261–291. 34. Chen HZ, Tsai SY, Leone G. Emerging roles of ment. Development. 2011;138:2661–2672.
9. Malumbres M, Barbacid M. Mammalian cyclin- E2Fs in cancer: an exit from cell cycle control. Nat 57. Avo Santos M, van de Werken C, de Vries M, et al.
dependent kinases. Trends Biochem Sci. 2005;30: Rev Cancer. 2009;9:785–797. A role for Aurora C in the chromosomal passenger
630–641. 35. Trimarchi JM, Lees JA. Sibling rivalry in the E2F complex during human preimplantation embryo
10. Malumbres M, Harlow E, Hunt T, et al. Cyclin- family. Nat Rev Mol Cell Biol. 2002;3:11–20. development. Hum Reprod. 2011;26:1868–1881.
dependent kinases: a family portrait. Nat Cell Biol. 36. Chong JL, Wenzel PL, Saenz-Robles MT, et al. 58. Petronczki M, Lenart P, Peters JM. Polo on the
2009;11:1275–1276. E2f1-3 switch from activators in progenitor rise—from mitotic entry to cytokinesis with
11. Malumbres M. Cyclin-dependent kinases. Genome cells to repressors in differentiating cells. Nature. Plk1. Dev Cell. 2008;14:646–659.
Biol. 2014;15:122. 2009;462:930–934. 59. Bettencourt-Dias M, Rodrigues-Martins A,
12. Lee MG, Nurse P. Complementation used to clone 37. Iglesias A, Murga M, Laresgoiti U, et al. Dia- Carpenter L, et al. SAK/PLK4 is required for
a human homologue of the fission yeast cell cycle betes and exocrine pancreatic insufficiency in centriole duplication and flagella development.
control gene cdc2. Nature. 1987;327:31–35. E2F1/E2F2 double-mutant mice. J Clin Invest. Curr Biol. 2005;15:2199–2207.
13. Pavletich NP. Mechanisms of cyclin-dependent 2004;113:1398–1407. 60. Rodrigues-Martins A, Riparbelli M, Callaini G,
kinase regulation: structures of Cdks, their cyclin 38. Infante A, Laresgoiti U, Fernandez-Rueda J, et al. et al. Revisiting the role of the mother centriole in
activators, and Cip and INK4 inhibitors. J Mol Biol. E2F2 represses cell cycle regulators to maintain centriole biogenesis. Science. 2007;316:1046–1050.
1999;287:821–828. quiescence. Cell Cycle. 2008;7:3915–3927. 61. Fry AM, O’Regan L, Sabir SR, Bayliss R. Cell cycle
14. Evans T, Rosenthal ET, Youngblom J, et al. Cyclin: 39. Peters JM. The anaphase promoting complex/cyclo- regulation by the NEK family of protein kinases.
a protein specified by maternal mRNA in sea urchin some: a machine designed to destroy. Nat Rev Mol J Cell Sci. 2012;125:4423–4433.
eggs that is destroyed at each cleavage division. Cell. Cell Biol. 2006;7:644–656. 62. Quarmby LM, Mahjoub MR. Caught Nek-ing: cilia
1983;33:389–396. 40. Silverman JS, Skaar JR, Pagano M. SCF ubiquitin and centrioles. J Cell Sci. 2005;118:5161–5169.
15. Deshpande A, Sicinski P, Hinds PW. Cyclins and cdks ligases in the maintenance of genome stability. Trends 63. Sdelci S, Bertran MT, Roig J. Nek9, Nek6, Nek7
in development and cancer: a perspective. Oncogene. Biochem Sci. 2012;37:66–73. and the separation of centrosomes. Cell Cycle.
2005;24:2909–2915. 41. Skaar JR, Pagano M. Control of cell growth by the 2011;10:3816–3817.
16. Russo AA, Jeffrey PD, Pavletich NP. Structural SCF and APC/C ubiquitin ligases. Curr Opin Cell 64. Wang F, Dai J, Daum JR, Niedzialkowska E,
basis of cyclin-dependent kinase activation by Biol. 2009;21:816–824. Banerjee B, Stukenberg PT, et al. Histone H3 Thr-3
phosphorylation. Nat Struct Biol. 1996;3:696–700. 42. Mocciaro A, Rape M. Emerging regulatory mecha- phosphorylation by Haspin positions Aurora B at
17. Lolli G, Johnson LN. CAK-cyclin-dependent nisms in ubiquitin-dependent cell cycle control. centromeres in mitosis. Science. 2010;330:231–235.
activating kinase: a key kinase in cell cycle control J Cell Sci. 2012;125:255–263. 65. Kelly AE, Ghenoiu C, Xue JZ, Zierhut C, Kimura H,
and a target for drugs? Cell Cycle. 2005;4:572–577. 43. Eguren M, Manchado E, Malumbres M. Non-mitotic Funabiki H. Survivin reads phosphorylated histone
18. Fisher RP. Secrets of a double agent: CDK7 in functions of the Anaphase-Promoting Complex. H3 threonine 3 to activate the mitotic kinase Aurora
cell-cycle control and transcription. J Cell Sci. Semin Cell Dev Biol. 2011;22:572–578. B. Science. 2010;330:235–239.
2005;118:5171–5180. 44. Choudhury R, Bonacci T, Arceci A, Lahiri D, Mills 66. Moutinho-Santos T, Maiato H. Plk1 puts a (Has)
19. Kellogg DR. Wee1-dependent mechanisms required CA, Kernan JL, et al. APC/C and SCF(cyclin F) pin on the mitotic histone code. EMBO Rep.
for coordination of cell growth and cell division. constitute a reciprocal feedback circuit controlling 2014;15:203–204.
J Cell Sci. 2003;116:4883–4890. S-phase entry. Cell Rep. 2016;16:3359–3372. 67. Varier RA, Outchkourov NS, de Graaf P, van
20. Boutros R, Lobjois V, Ducommun B. CDC25 45. Alfieri C, Chang L, Zhang Z, Yang J, Maslen S, Schaik FM, Ensing HJ, Wang F, et al. A phospho/
phosphatases in cancer cells: key players? Good Skehel M, et al. Molecular basis of APC/C regula- methyl switch at histone H3 regulates TFIID
targets? Nat Rev Cancer. 2007;7:495–507. tion by the spindle assembly checkpoint. Nature. association with mitotic chromosomes. EMBO J.
21. Sherr CJ, Roberts JM. CDK inhibitors: positive 2016;536:431–436. 2010;29:3967–3978.
and negative regulators of G1-phase progression. 46. Zhang S, Chang L, Alfieri C, Zhang Z, Yang J, 68. Alvarez-Fernández M, Malumbres M. Preparing a
Genes Dev. 1999;13:1501–1512. Maslen S, et al. Molecular mechanism of APC/C cell for nuclear envelope breakdown: spatio-temporal
22. LaBaer J, Garrett MD, Stevenson LF, et al. New activation by mitotic phosphorylation. Nature. control of phosphorylation during mitotic entry.
functional activities for the p21 family of CDK 2016;533:260–264. Bioessays. 2014;36:757–765.
inhibitors. Genes Dev. 1997;11:847–862. 47. Brown NG, VanderLinden R, Watson ER, 69. Wordeman L. How kinesin motor proteins drive
23. Chu I, Sun J, Arnaout A, et al. p27 phosphorylation Weissmann F, Ordureau A, Wu KP, et al. E3 mitotic spindle function: lessons from molecular
by Src regulates inhibition of cyclin E-Cdk2. Cell. architectures regulate multiubiquitination and ubiq- assays. Semin Cell Dev Biol. 2010;21:260–268.
2007;128:281–294. uitin chain elongation by APC/C. Cell. 2016;165: 70. Cross RA, McAinsh A. Prime movers: the mecha-
24. Grimmler M, Wang Y, Mund T, et al. Cdk- 1440–1453. nochemistry of mitotic kinesins. Nat Rev Mol Cell
inhibitory activity and stability of p27Kip1 are 48. Qiao R, Weissmann F, Yamaguchi M, Brown NG, Biol. 2014;15:257–271.
directly regulated by oncogenic tyrosine kinases. VanderLinden R, Imre R, et al. Mechanism of APC/ 71. Foley EA, Kapoor TM. Microtubule attachment
Cell. 2007;128:269–280. CCDC20 activation by mitotic phosphorylation. and spindle assembly checkpoint signalling at the
25. Larrea MD, Liang J, Da Silva T, et al. Phosphoryla- Proc Natl Acad Sci USA. 2016;113:E2570–E2578. kinetochore. Nat Rev Mol Cell Biol. 2013;14:25–37.
tion of p27Kip1 regulates assembly and activation of 49. Fujimitsu K, Grimaldi M, Yamano H. Cyclin- 72. Fukagawa T, Earnshaw WC. The centromere:
cyclin D1-Cdk4. Mol Cell Biol. 2008;28:6462–6472. dependent kinase 1-dependent activation of APC/C chromatin foundation for the kinetochore machinery.
26. Jakel H, Weinl C, Hengst L. Phosphorylation ubiquitin ligase. Science. 2016;352:1121–1124. Dev Cell. 2014;30:496–508.
of p27Kip1 by JAK2 directly links cytokine 50. Archambault V, Glover DM. Polo-like kinases: 73. Bloom KS. Centromeric heterochromatin: the
receptor signaling to cell cycle control. Oncogene. conservation and divergence in their functions and primordial segregation machine. Annu Rev Genet.
2011;30:3502–3512. regulation. Nat Rev Mol Cell Biol. 2009;10:265–275. 2014;48:457–484.
73.e2 Part I: Science and Clinical Oncology

74. McKinley KL, Cheeseman IM. The molecular basis D1 proteolysis and subcellular localization. Genes multiprotein complex. Nature. 1992;357:128–
for centromere identity and function. Nat Rev Mol Dev. 1998;12:3499–3511. 134.
Cell Biol. 2016;17:16–29. 96. Klein EA, Assoian RK. Transcriptional regulation of 119. Nougarede R, Della Seta F, Zarzov P, Schwob E.
75. Godek KM, Kabeche L, Compton DA. Regulation the cyclin D1 gene at a glance. J Cell Sci. 2008;121: Hierarchy of S-phase-promoting factors: yeast Dbf4-
of kinetochore-microtubule attachments through 3853–3857. Cdc7 kinase requires prior S-phase cyclin-dependent
homeostatic control during mitosis. Nat Rev Mol 97. Yu Q, Ciemerych MA, Sicinski P. Ras and Myc can kinase activation. Mol Cell Biol. 2000;20:3795–3806.
Cell Biol. 2015;16:57–64. drive oncogenic cell proliferation through individual 120. Zou L, Stillman B. Assembly of a complex contain-
76. Etemad B, Kops GJ. Attachment issues: kinetochore D-cyclins. Oncogene. 2005;24:7114–7119. ing Cdc45p, replication protein A, and Mcm2p
transformations and spindle checkpoint silencing. 98. Ewen ME, Sluss HK, Sherr CJ, et al. Functional at replication origins controlled by S-phase cyclin-
Curr Opin Cell Biol. 2016;39:101–108. interactions of the retinoblastoma protein with dependent kinases and Cdc7p-Dbf4p kinase. Mol
77. Weir JR, Faesen AC, Klare K, Petrovic A, Basilico mammalian D-type cyclins. Cell. 1993;73:487–497. Cell Biol. 2000;20:3086–3096.
F, Fischböck J, et al. Insights from biochemical 99. Connell-Crowley L, Harper JW, Goodrich 121. Walter J, Newport J. Initiation of eukaryotic
reconstitution into the architecture of human DW. Cyclin D1/Cdk4 regulates retinoblastoma DNA replication: origin unwinding and sequential
kinetochores. Nature. 2016;537:249–253. protein-mediated cell cycle arrest by site-specific chromatin association of Cdc45, RPA, and DNA
78. Wurzenberger C, Gerlich DW. Phosphatases: phosphorylation. Mol Biol Cell. 1997;8:287–301. polymerase alpha. Mol Cell. 2000;5:617–627.
providing safe passage through mitotic exit. Nat 100. Connell-Crowley L, Elledge SJ, Harper JW. G1 122. Arias EE, Walter JC. Replication-dependent destruc-
Rev Mol Cell Biol. 2011;12:469–482. cyclin-dependent kinases are sufficient to initiate tion of Cdt1 limits DNA replication to a single
79. Mocciaro A, Schiebel E. Cdc14: a highly conserved DNA synthesis in quiescent human fibroblasts. Curr round per cell cycle in Xenopus egg extracts. Genes
family of phosphatases with non-conserved func- Biol. 1998;8:65–68. Dev. 2005;19:114–126.
tions? J Cell Sci. 2010;123:2867–2876. 101. Kato J, Matsushime H, Hiebert SW, et al. Direct 123. Higa LA, Banks D, Wu M, et al. L2DTL/CDT2
80. Virshup DM, Shenolikar S. From promiscuity to binding of cyclin D to the retinoblastoma gene interacts with the CUL4/DDB1 complex and PCNA
precision: protein phosphatases get a makeover. Mol product (pRb) and pRb phosphorylation by the and regulates CDT1 proteolysis in response to DNA
Cell. 2009;33:537–545. cyclin D-dependent kinase CDK4. Genes Dev. damage. Cell Cycle. 2006;5:1675–1680.
81. Kurimchak A, Grana X. PP2A holoenzymes nega- 1993;7:331–342. 124. Jin J, Arias EE, Chen J, et al. A family of diverse
tively and positively regulate cell cycle progression 102. Kollmann K, Heller G, Schneckenleithner C, Warsch Cul4-Ddb1-interacting proteins includes Cdt2,
by dephosphorylating pocket proteins and multiple W, Scheicher R, Ott RG, et al. A kinase-independent which is required for S phase destruction of the
CDK substrates. Gene. 2012;499:1–7. function of CDK6 links the cell cycle to tumor replication factor Cdt1. Mol Cell. 2006;23:709–721.
82. Kolupaeva V, Janssens V. PP1 and PP2A phos- angiogenesis. Cancer Cell. 2013;24:167–181. 125. Sansam CL, Shepard JL, Lai K, et al. DTL/CDT2
phatases–cooperating partners in modulating 103. Tigan AS, Bellutti F, Kollmann K, Tebb G, Sexl is essential for both CDT1 regulation and the early
retinoblastoma protein activation. FEBS J. 2013;280: V. CDK6-a review of the past and a glimpse into G2/M checkpoint. Genes Dev. 2006;20:3117–3129.
627–643. the future: from cell-cycle control to transcriptional 126. Arias EE, Walter JC. PCNA functions as a molecular
83. Qian J, Winkler C, Bollen M. 4D-networking regulation. Oncogene. 2016;35:3083–3091. platform to trigger Cdt1 destruction and prevent
by mitotic phosphatases. Curr Opin Cell Biol. 104. Ohtani K, DeGregori J, Nevins JR. Regulation of re-replication. Nat Cell Biol. 2006;8:84–90.
2013;25:697–703. the cyclin E gene by transcription factor E2F1. Proc 127. Sivaprasad U, Machida YJ, Dutta A. APC/C—the
84. Grallert A, Boke E, Hagting A, Hodgson B, Natl Acad Sci USA. 1995;92:12146–12150. master controller of origin licensing? Cell Div.
Connolly Y, Griffiths JR, et al. A PP1-PP2A 105. Geng Y, Eaton EN, Picon M, et al. Regulation of 2007;2:8.
phosphatase relay controls mitotic progression. cyclin E transcription by E2Fs and retinoblastoma 128. Pagano M, Pepperkok R, Verde F, et al. Cyclin A
Nature. 2015;517:94–98. protein. Oncogene. 1996;12:1173–1180. is required at two points in the human cell cycle.
85. Castilho PV, Williams BC, Mochida S, et al. 106. Montagnoli A, Fiore F, Eytan E, et al. Ubiquitination EMBO J. 1992;11:961–971.
The M phase kinase Greatwall (Gwl) promotes of p27 is regulated by Cdk-dependent phosphoryla- 129. Draetta G, Luca F, Westendorf J, et al. Cdc2 protein
inactivation of PP2A/B55delta, a phosphatase tion and trimeric complex formation. Genes Dev. kinase is complexed with both cyclin A and B:
directed against CDK phosphosites. Mol Biol Cell. 1999;13:1181–1189. evidence for proteolytic inactivation of MPF. Cell.
2009;20:4777–4789. 107. Sheaff RJ, Groudine M, Gordon M, et al. Cyclin 1989;56:829–838.
86. Vigneron S, Brioudes E, Burgess A, et al. Greatwall E-CDK2 is a regulator of p27Kip1. Genes Dev. 130. Cardoso MC, Leonhardt H, Nadal-Ginard B.
maintains mitosis through regulation of PP2A. 1997;11:1464–1478. Reversal of terminal differentiation and control of
EMBO J. 2009;28:2786–2793. 108. Carrano AC, Eytan E, Hershko A, Pagano M. SKP2 DNA replication: cyclin A and Cdk2 specifically
87. Glover DM. The overlooked greatwall: a new perspec- is required for ubiquitin-mediated degradation of the localize at subnuclear sites of DNA replication. Cell.
tive on mitotic control. Open Biol. 2012;2:120023. CDK inhibitor p27. Nat Cell Biol. 1999;1:193–199. 1993;74:979–992.
88. Mochida S, Maslen SL, Skehel M, Hunt T. 109. Koepp DM, Schaefer LK, Ye X, et al. Phos- 131. Dynlacht BD, Flores O, Lees JA, Harlow E. Dif-
Greatwall phosphorylates an inhibitor of protein phorylation-dependent ubiquitination of cyclin ferential regulation of E2F transactivation by cyclin/
phosphatase 2A that is essential for mitosis. Science. E by the SCFFbw7 ubiquitin ligase. Science. cdk2 complexes. Genes Dev. 1994;8:1772–1786.
2010;330:1670–1673. 2001;294:173–177. 132. Krek W, Xu G, Livingston DM. Cyclin A-kinase
89. Gharbi-Ayachi A, Labbe JC, Burgess A, et al. The 110. Clurman BE, Sheaff RJ, Thress K, et al. Turnover regulation of E2F-1 DNA binding function
substrate of Greatwall kinase, Arpp19, controls of cyclin E by the ubiquitin-proteasome pathway is underlies suppression of an S phase checkpoint.
mitosis by inhibiting protein phosphatase 2A. Science. regulated by cdk2 binding and cyclin phosphoryla- Cell. 1995;83:1149–1158.
2010;330:1673–1677. tion. Genes Dev. 1996;10:1979–1990. 133. Kalaszczynska I, Geng Y, Iino T, Mizuno S, Choi
90. Manchado E, Guillamot M, de Carcer G, et al. 111. Berthet C, Aleem E, Coppola V, et al. Cdk2 knock- Y, Kondratiuk I, et al. Cyclin A is redundant in
Targeting mitotic exit leads to tumor regression out mice are viable. Curr Biol. 2003;13:1775–1785. fibroblasts but essential in hematopoietic and
in vivo: modulation by Cdk1, Mastl, and the 112. Ortega S, Prieto I, Odajima J, et al. Cyclin-dependent embryonic stem cells. Cell. 2009;138:352–365.
PP2A/B55alpha,delta phosphatase. Cancer Cell. kinase 2 is essential for meiosis but not for mitotic 134. Kanakkanthara A, Jeganathan KB, Limzerwala JF,
2010;18:641–654. cell division in mice. Nat Genet. 2003;35:25–31. Baker DJ, Hamada M, Nam HJ, et al. Cyclin A2
91. Schmitz MH, Held M, Janssens V, et al. Live-cell 113. Martin A, Odajima J, Hunt SL, et al. Cdk2 is is an RNA binding protein that controls Mre11
imaging RNAi screen identifies PP2A-B55alpha dispensable for cell cycle inhibition and tumor mRNA translation. Science. 2016;353:1549–1552.
and importin-beta1 as key mitotic exit regulators suppression mediated by p27(Kip1) and p21(Cip1). 135. Stracker TH, Petrini JH. The MRE11 complex:
in human cells. Nat Cell Biol. 2010;12:886–893. Cancer Cell. 2005;7:591–598. starting from the ends. Nat Rev Mol Cell Biol.
92. Wu JQ, Guo JY, Tang W, et al. PP1-mediated 114. Aleem E, Kiyokawa H, Kaldis P. Cdc2-cyclin E 2011;12:90–103.
dephosphorylation of phosphoproteins at mitotic exit complexes regulate the G1/S phase transition. Nat 136. Glotzer M. The 3Ms of central spindle assembly:
is controlled by inhibitor-1 and PP1 phosphoryla- Cell Biol. 2005;7(8):831–836. microtubules, motors and MAPs. Nat Rev Mol Cell
tion. Nat Cell Biol. 2009;11:644–651. 115. Santamaria D, Barriere C, Cerqueira A, et al. Cdk1 is Biol. 2009;10:9–20.
93. Takahashi Y, Rayman JB, Dynlacht BD. Analysis of sufficient to drive the mammalian cell cycle. Nature. 137. Hornick JE, Karanjeet K, Collins ES, Hinchcliffe
promoter binding by the E2F and pRB families in 2007;448:811–815. EH. Kinesins to the core: the role of microtubule-
vivo: distinct E2F proteins mediate activation and 116. Viera A, Rufas JS, Martinez I, et al. CDK2 is required based motor proteins in building the mitotic spindle
repression. Genes Dev. 2000;14:804–816. for proper homologous pairing, recombination and midzone. Semin Cell Dev Biol. 2010;21:290–299.
94. Rayman JB, Takahashi Y, Indjeian VB, et al. E2F sex-body formation during male mouse meiosis. 138. Nasmyth K, Haering CH. The structure and function
mediates cell cycle-dependent transcriptional repres- J Cell Sci. 2009;122:2149–2159. of SMC and kleisin complexes. Annu Rev Biochem.
sion in vivo by recruitment of an HDAC1/mSin3B 117. Bell SP, Dutta A. DNA replication in eukaryotic 2005;74:595–648.
corepressor complex. Genes Dev. 2002;16:933–947. cells. Annu Rev Biochem. 2002;71:333–374. 139. Wood AJ, Severson AF, Meyer BJ. Condensin and
95. Diehl JA, Cheng M, Roussel MF, Sherr CJ. 118. Bell SP, Stillman B. ATP-dependent recognition cohesin complexity: the expanding repertoire of
Glycogen synthase kinase-3beta regulates cyclin of eukaryotic origins of DNA replication by a functions. Nat Rev Genet. 2010;11:391–404.
Control of the Cell Cycle  •  CHAPTER 4 73.e3
73.e3

140. Sherwood R, Takahashi TS, Jallepalli PV. Sister 165. Li M, Shin YH, Hou L, et al. The adaptor protein of 190. Bartkova J, Rezaei N, Liontos M, et al. Oncogene-
acts: coordinating DNA replication and cohesion the anaphase promoting complex Cdh1 is essential induced senescence is part of the tumorigenesis
establishment. Genes Dev. 2010;24:2723–2731. in maintaining replicative lifespan and in learning barrier imposed by DNA damage checkpoints.
141. Jeppsson K, Kanno T, Shirahige K, Sjögren C. The and memory. Nat Cell Biol. 2008;10:1083–1089. Nature. 2006;444:633–637.
maintenance of chromosome structure: positioning 166. Vagnarelli P, Earnshaw WC. Repo-Man-PP1: a link 191. Di Micco R, Fumagalli M, Cicalese A, et al.
and functioning of SMC complexes. Nat Rev Mol between chromatin remodelling and nuclear envelope Oncogene-induced senescence is a DNA damage
Cell Biol. 2014;15:601–614. reassembly. Nucleus. 2012;3:138–142. response triggered by DNA hyper-replication.
142. Nasmyth K, Haering CH. Cohesin: its roles and 167. Green RA, Paluch E, Oegema K. Cytokinesis in Nature. 2006;444:638–642.
mechanisms. Annu Rev Genet. 2009;43:525–558. animal cells. Annu Rev Cell Dev Biol. 2012;28:29–58. 192. Palmero I, Pantoja C, Serrano M. p19ARF
143. Gligoris T, Löwe J. Structural insights into ring 168. Mierzwa B, Gerlich DW. Cytokinetic abscission: links the tumour suppressor p53 to Ras. Nature.
formation of cohesin and related Smc complexes. molecular mechanisms and temporal control. Dev 1998;395:125–126.
Trends Cell Biol. 2016;26:680–693. Cell. 2014;31:525–538. 193. Malumbres M, Perez De Castro I, Hernandez MI,
144. Nasmyth K. Cohesin: a catenase with separate entry 169. D’Avino PP, Capalbo L. Regulation of midbody et al. Cellular response to oncogenic ras involves
and exit gates? Nat Cell Biol. 2011;13:1170–1177. formation and function by mitotic kinases. Semin induction of the Cdk4 and Cdk6 inhibitor
145. Losada A. Cohesin in cancer: chromosome segrega- Cell Dev Biol. 2016;53:57–63. p15(INK4b). Mol Cell Biol. 2000;20:2915–2925.
tion and beyond. Nat Rev Cancer. 2014;14:389–393. 170. Takaki T, Trenz K, Costanzo V, Petronczki M. Polo- 194. Sherr CJ. The INK4a/ARF network in tumour
146. Hirano T. Condensin-based chromosome organiza- like kinase 1 reaches beyond mitosis–cytokinesis, suppression. Nat Rev Mol Cell Biol. 2001;2:731–737.
tion from bacteria to vertebrates. Cell. 2016;164: DNA damage response, and development. Curr 195. Dimri GP, Itahana K, Acosta M, Campisi J. Regula-
847–857. Opin Cell Biol. 2008;20:650–660. tion of a senescence checkpoint response by the
147. Di Fiore B, Pines J. How cyclin A destruction 171. Steigemann P, Wurzenberger C, Schmitz MH, et al. E2F1 transcription factor and p14(ARF) tumor
escapes the spindle assembly checkpoint. J Cell Aurora B-mediated abscission checkpoint protects suppressor. Mol Cell Biol. 2000;20:273–285.
Biol. 2010;190:501–509. against tetraploidization. Cell. 2009;136:473–484. 196. Kamijo T, Weber JD, Zambetti G, et al. Functional
148. Lundgren K, Walworth N, Booher R, et al. mik1 172. Nähse V, Christ L, Stenmark H, Campsteijn C. and physical interactions of the ARF tumor sup-
and wee1 cooperate in the inhibitory tyrosine The abscission checkpoint: making it to the final pressor with p53 and Mdm2. Proc Natl Acad Sci
phosphorylation of cdc2. Cell. 1991;64:1111–1122. cut. Trends Cell Biol. 2017;27:1–11. USA. 1998;95:8292–8297.
149. Parker LL, Piwnica-Worms H. Inactivation of the 173. Bartek J, Lukas J. DNA damage checkpoints: from 197. Honda R, Yasuda H. Association of p19(ARF) with
p34cdc2-cyclin B complex by the human WEE1 initiation to recovery or adaptation. Curr Opin Cell Mdm2 inhibits ubiquitin ligase activity of Mdm2 for
tyrosine kinase. Science. 1992;257:1955–1957. Biol. 2007;19:238–245. tumor suppressor p53. EMBO J. 1999;18:22–27.
150. Heald R, McLoughlin M, McKeon F. Human wee1 174. Taylor AM, Harnden DG, Arlett CF, et al. Ataxia 198. Pomerantz J, Schreiber-Agus N, Liegeois NJ, et al.
maintains mitotic timing by protecting the nucleus telangiectasia: a human mutation with abnormal The Ink4a tumor suppressor gene product, p19Arf,
from cytoplasmically activated Cdc2 kinase. Cell. radiation sensitivity. Nature. 1975;258:427–429. interacts with MDM2 and neutralizes MDM2’s
1993;74:463–474. 175. Klingseisen A, Jackson AP. Mechanisms and pathways inhibition of p53. Cell. 1998;92:713–723.
151. Gavet O, Pines J. Activation of cyclin B1-Cdk1 of growth failure in primordial dwarfism. Genes Dev. 199. Weber JD, Taylor LJ, Roussel MF, et al. Nucleolar
synchronizes events in the nucleus and the cytoplasm 2011;25:2011–2024. Arf sequesters Mdm2 and activates p53. Nat Cell
at mitosis. J Cell Biol. 2010;189:247–259. 176. Cuadrado M, Martinez-Pastor B, Murga M, Biol. 1999;1:20–26.
152. Lindqvist A, Rodríguez-Bravo V, Medema RH. et al. ATM regulates ATR chromatin loading in 200. Higa LAA, Mihaylov IS, Banks DP, et al. Radiation-
The decision to enter mitosis: feedback and response to DNA double-strand breaks. J Exp Med. mediated proteolysis of CDT1 by CUL4-ROC1 and
redundancy in the mitotic entry network. J Cell 2006;203:297–303. CSN complexes constitutes a new checkpoint. Nat
Biol. 2009;185:193–202. 177. Matsuoka S, Huang M, Elledge SJ. Linkage of ATM Cell Biol. 2003;5:1008–1015.
153. Salic A, Waters JC, Mitchison TJ. Vertebrate to cell cycle regulation by the Chk2 protein kinase. 201. O’Connell BC, Harper JW. Ubiquitin proteasome
shugoshin links sister centromere cohesion and Science. 1998;282:1893–1897. system (UPS): what can chromatin do for you? Curr
kinetochore microtubule stability in mitosis. Cell. 178. Matsuoka S, Rotman G, Ogawa A, et al. Ataxia Opin Cell Biol. 2007;19:206–214.
2004;118:567–578. telangiectasia-mutated phosphorylates Chk2 202. Raman M, Havens CG, Walter JC, Harper JW. A
154. Rivera T, Losada A. Shugoshin and PP2A, shared in vivo and in vitro. Proc Natl Acad Sci USA. genome-wide screen identifies p97 as an essential
duties at the centromere. Bioessays. 2006;28:775–779. 2000;97:10389–10394. regulator of DNA damage-dependent CDT1
155. Watanabe Y. Shugoshin: guardian spirit at the 179. Busino L, Donzelli M, Chiesa M, et al. Degradation destruction. Mol Cell. 2011;44:72–84.
centromere. Curr Opin Cell Biol. 2005;17:590–595. of Cdc25A by beta-TrCP during S phase and in 203. Gottifredi V, Prives C. The S phase checkpoint:
156. Champion L, Linder MI, Kutay U. Cellular response to DNA damage. Nature. 2003;426:87–91. when the crowd meets at the fork. Semin Cell Dev
reorganization during mitotic entry. Trends Cell 180. Hirao A, Kong YY, Matsuoka S, et al. DNA damage- Biol. 2005;16:355–368.
Biol. 2017;27:26–41. induced activation of p53 by the checkpoint kinase 204. Muñoz S, Méndez J. DNA replication stress:
157. Álvarez-Fernández M, Sánchez-Martínez R, Sanz- Chk2. Science. 2000;287:1824–1827. from molecular mechanisms to human disease.
Castillo B, Gan PP, Sanz-Flores M, Trakala M, et al. 181. Manfredi JJ. The Mdm2-p53 relationship evolves: Chromosoma. 2016;doi:10.1007/s00412-016-0573-x.
Greatwall is essential to prevent mitotic collapse after Mdm2 swings both ways as an oncogene and a 205. Furnari B, Blasina A, Boddy MN, et al. Cdc25
nuclear envelope breakdown in mammals. Proc Natl tumor suppressor. Genes Dev. 2010;24:1580–1589. inhibited in vivo and in vitro by checkpoint kinases
Acad Sci USA. 2013;110:17374–17379. 182. Brown CJ, Cheok CF, Verma CS, Lane DP. Reactiva- Cds1 and Chk1. Mol Biol Cell. 1999;10:833–
158. Diril MK, Ratnacaram CK, Padmakumar VC, Du T, tion of p53: from peptides to small molecules. Trends 845.
Wasser M, Coppola V, et al. Cyclin-dependent kinase Pharmacol Sci. 2011;32:53–62. 206. Liu Q, Guntuku S, Cui XS, et al. Chk1 is an essential
1 (Cdk1) is essential for cell division and suppression 183. Montes de Oca Luna R, Wagner DS, Lozano G. kinase that is regulated by Atr and required for
of DNA re-replication but not for liver regeneration. Rescue of early embryonic lethality in mdm2- the G(2)/M DNA damage checkpoint. Genes Dev.
Proc Natl Acad Sci USA. 2012;109:3826–3831. deficient mice by deletion of p53. Nature. 1995;378: 2000;14:1448–1459.
159. Musacchio A, Salmon ED. The spindle-assembly 203–206. 207. Flynn RL, Zou L. ATR: a master conductor of
checkpoint in space and time. Nat Rev Mol Cell 184. Jones SN, Roe AE, Donehower LA, Bradley A. cellular responses to DNA replication stress. Trends
Biol. 2007;8:379–393. Rescue of embryonic lethality in Mdm2-deficient Biochem Sci. 2011;36:133–140.
160. Sacristan C, Kops GJ. Joined at the hip: kinetochores, mice by absence of p53. Nature. 1995;378:206–208. 208. Smith J, Tho LM, Xu N, Gillespie DA. The
microtubules, and spindle assembly checkpoint 185. Waldman T, Kinzler KW, Vogelstein B. p21 is ATM-Chk2 and ATR-Chk1 pathways in DNA
signaling. Trends Cell Biol. 2015;25:21–28. necessary for the p53-mediated G1 arrest in human damage signaling and cancer. Adv Cancer Res.
161. Sullivan M, Morgan DO. Finishing mitosis, one step cancer cells. Cancer Res. 1995;55:5187–5190. 2010;108:73–112.
at a time. Nat Rev Mol Cell Biol. 2007;8:894–903. 186. el-Deiry WS, Tokino T, Velculescu VE, et al. WAF1, 209. Branzei D, Foiani M. Regulation of DNA repair
162. Gorr IH, Boos D, Stemmann O. Mutual inhibition a potential mediator of p53 tumor suppression. Cell. throughout the cell cycle. Nat Rev Mol Cell Biol.
of separase and Cdk1 by two-step complex forma- 1993;75:817–825. 2008;9:297–308.
tion. Mol Cell. 2005;19:135–141. 187. Vousden KH, Lu X. Live or let die: the cell’s response 210. Lobrich M, Jeggo PA. The impact of a negligent
163. Schockel L, Mockel M, Mayer B, et al. Cleavage of to p53. Nat Rev Cancer. 2002;2:594–604. G2/M checkpoint on genomic instability and cancer
cohesin rings coordinates the separation of centrioles 188. Serrano M, Lin AW, McCurrach ME, et al. induction. Nat Rev Cancer. 2007;7:861–869.
and chromatids. Nat Cell Biol. 2011;13:966– Oncogenic ras provokes premature cell senescence 211. Bassermann F, Pagano M. Dissecting the role
972. associated with accumulation of p53 and p16INK4a. of ubiquitylation in the DNA damage response
164. Garcia-Higuera I, Manchado E, Dubus P, et al. Cell. 1997;88:593–602. checkpoint in G2. Cell Death Differ. 2010;17:78–85.
Genomic stability and tumour suppression by the 189. Collado M, Serrano M. Senescence in tumours: 212. van Vugt MA, Medema RH. Getting in and out of
APC/C cofactor Cdh1. Nat Cell Biol. 2008;10: evidence from mice and humans. Nat Rev Cancer. mitosis with Polo-like kinase-1. Oncogene. 2005;24:
802–811. 2010;10:51–57. 2844–2859.
73.e4 Part I: Science and Clinical Oncology

213. Mapelli M, Massimiliano L, Santaguida S, Musacchio 235. Edlund K, Larsson O, Ameur A, et al. Data-driven 258. Perez de Castro I, de Carcer G, Malumbres M. A
A. The Mad2 conformational dimer: structure and unbiased curation of the TP53 tumor suppressor census of mitotic cancer genes: new insights into
implications for the spindle assembly checkpoint. gene mutation database and validation by ultradeep tumor cell biology and cancer therapy. Carcinogenesis.
Cell. 2007;131:730–743. sequencing of human tumors. Proc Natl Acad Sci 2007;28:899–912.
214. Maresca TJ, Salmon ED. Welcome to a new kind USA. 2012;109:9551–9556. 259. Michel LS, Liberal V, Chatterjee A, et al. MAD2
of tension: translating kinetochore mechanics into a 236. Nigro JM, Baker SJ, Preisinger AC, et al. Mutations haplo-insufficiency causes premature anaphase and
wait-anaphase signal. J Cell Sci. 2010;123:825–835. in the p53 gene occur in diverse human tumour chromosome instability in mammalian cells. Nature.
215. Gordon DJ, Resio B, Pellman D. Causes and types. Nature. 1989;342:705–708. 2001;409:355–359.
consequences of aneuploidy in cancer. Nat Rev 237. Momand J, Jung D, Wilczynski S, Niland J. The 260. Sotillo R, Schvartzman JM, Socci ND, Benezra R.
Genet. 2012;13:189–203. MDM2 gene amplification database. Nucleic Acids Mad2-induced chromosome instability leads to lung
216. Malumbres M. Oncogene-induced mitotic stress: p53 Res. 1998;26:3453–3459. tumour relapse after oncogene withdrawal. Nature.
and pRb get mad too. Cancer Cell. 2011;19:691–692. 238. Scheffner M, Werness BA, Huibregtse JM, et al. The 2010;464:436–440.
217. Manning AL, Dyson NJ. RB: mitotic implica- E6 oncoprotein encoded by human papillomavirus 261. Carter SL, Eklund AC, Kohane IS, et al. A signature
tions of a tumour suppressor. Nat Rev Cancer. types 16 and 18 promotes the degradation of p53. of chromosomal instability inferred from gene expres-
2012;12:220–226. Cell. 1990;63:1129–1136. sion profiles predicts clinical outcome in multiple
218. Weinstat-Saslow D, Merino MJ, Manrow RE, et al. 239. Khanna KK. Cancer risk and the ATM gene: a con- human cancers. Nat Genet. 2006;38:1043–1048.
Overexpression of cyclin D mRNA distinguishes tinuing debate. J Natl Cancer Inst. 2000;92:795–802. 262. Yamada HY, Rao CV. Genes that modulate the sen-
invasive and in situ breast carcinomas from non- 240. Barlow C, Hirotsune S, Paylor R, et al. Atm-deficient sitivity for anti-microtubule drug-mediated chemo-
malignant lesions. Nat Med. 1995;1:1257–1260. mice: a paradigm of ataxia telangiectasia. Cell. therapy. Curr Cancer Drug Targets. 2010;10:623–633.
219. Wang TC, Cardiff RD, Zukerberg L, et al. Mammary 1996;86:159–171. 263. Sansregret L, Patterson JO, Dewhurst S, López-
hyperplasia and carcinoma in MMTV-cyclin D1 241. Xu Y, Ashley T, Brainerd EE, et al. Targeted García C, Koch A, McGranahan N, et al. APC/C
transgenic mice. Nature. 1994;369:669–671. disruption of ATM leads to growth retardation, dysfunction limits excessive cancer chromosomal
220. Bortner DM, Rosenberg MP. Induction of mammary chromosomal fragmentation during meiosis, instability. Cancer Discov. 2017;doi:10.1158/2159-
gland hyperplasia and carcinomas in transgenic immune defects, and thymic lymphoma. Genes 8290.CD-16-0645.
mice expressing human cyclin E. Mol Cell Biol. Dev. 1996;10:2411–2422. 264. Crasta K, Ganem NJ, Dagher R, et al. DNA
1997;17:453–459. 242. Petrini JH. The Mre11 complex and ATM: col- breaks and chromosome pulverization from errors
221. Ortega S, Malumbres M, Barbacid M. Cyclin laborating to navigate S phase. Curr Opin Cell Biol. in mitosis. Nature. 2012;482:53–58.
D-dependent kinases, INK4 inhibitors and cancer. 2000;12:293–296. 265. Stephens PJ, Greenman CD, Fu B, et al. Massive
Biochim Biophys Acta. 2002;1602:73–87. 243. Bertoni F, Codegoni AM, Furlan D, et al. CHK1 genomic rearrangement acquired in a single
222. Catzavelos C, Bhattacharya N, Ung YC, et al. frameshift mutations in genetically unstable colorec- catastrophic event during cancer development. Cell.
Decreased levels of the cell-cycle inhibitor p27Kip1 tal and endometrial cancers. Genes Chromosomes 2011;144:27–40.
protein: prognostic implications in primary breast Cancer. 1999;26:176–180. 266. Janssen A, van der Burg M, Szuhai K, et al. Chromo-
cancer. Nat Med. 1997;3:227–230. 244. Matsuoka S, Nakagawa T, Masuda A, et al. Reduced some segregation errors as a cause of DNA damage
223. Porter PL, Malone KE, Heagerty PJ, et al. Expression expression and impaired kinase activity of a Chk2 and structural chromosome aberrations. Science.
of cell-cycle regulators p27Kip1 and cyclin E, alone mutant identified in human lung cancer. Cancer 2011;333:1895–1898.
and in combination, correlate with survival in young Res. 2001;61:5362–5365. 267. Hunt T. You never know: Cdk inhibitors as anti-
breast cancer patients. Nat Med. 1997;3:222–225. 245. Bell DW, Varley JM, Szydlo TE, et al. Heterozygous cancer drugs. Cell Cycle. 2008;7:3789–3790.
224. Chu IM, Hengst L, Slingerland JM. The Cdk germ line hCHK2 mutations in Li-Fraumeni 268. Malumbres M, Barbacid M. Cell cycle, CDKs
inhibitor p27 in human cancer: prognostic potential syndrome. Science. 1999;286:2528–2531. and cancer: a changing paradigm. Nat Rev Cancer.
and relevance to anticancer therapy. Nat Rev Cancer. 246. Wang W. Emergence of a DNA-damage response 2009;9:153–166.
2008;8:253–267. network consisting of Fanconi anaemia and BRCA 269. Shapiro GI. Cyclin-dependent kinase pathways
225. Malumbres M, Carnero A. Cell cycle deregulation: proteins. Nat Rev Genet. 2007;8:735–748. as targets for cancer treatment. J Clin Oncol.
a common motif in cancer. Prog Cell Cycle Res. 247. di Masi A, Antoccia A. NBS1 Heterozygosity 2006;24:1770–1783.
2003;5:5–18. and Cancer Risk. Curr Genomics. 2008;9:275– 270. Malumbres M, Pevarello P, Barbacid M, Bischoff
226. Gil J, Peters G. Regulation of the INK4b-ARF- 281. JR. CDK inhibitors in cancer therapy: what is next?
INK4a tumour suppressor locus: all for one or one 248. Kops GJ, Weaver BA, Cleveland DW. On the road to Trends Pharmacol Sci. 2008;29:16–21.
for all. Nat Rev Mol Cell Biol. 2006;7:667–677. cancer: aneuploidy and the mitotic checkpoint. Nat 271. Robles AI, Rodriguez-Puebla ML, Glick AB,
227. Bartkova J, Thullberg M, Rajpert-De Meyts E, Rev Cancer. 2005;5:773–785. et al. Reduced skin tumor development in cyclin
et al. Cell cycle regulators in testicular cancer: loss 249. Beroukhim R, Mermel CH, Porter D, et al. The D1-deficient mice highlights the oncogenic ras
of p18INK4C marks progression from carcinoma landscape of somatic copy-number alteration across pathway in vivo. Genes Dev. 1998;12:2469–2474.
in situ to invasive germ cell tumours. Int J Cancer. human cancers. Nature. 2010;463:899–905. 272. Yu Q, Geng Y, Sicinski P. Specific protection
2000;85:370–375. 250. Weaver BA, Cleveland DW. Aneuploidy: instiga- against breast cancers by cyclin D1 ablation. Nature.
228. Sanchez-Aguilera A, Delgado J, Camacho FI, et al. tor and inhibitor of tumorigenesis. Cancer Res. 2001;411:1017–1021.
Silencing of the p18INK4c gene by promoter 2007;67:10103–10105. 273. Campaner S, Doni M, Hydbring P, et al. Cdk2
hypermethylation in Reed-Sternberg cells in Hodgkin 251. Funk LC, Zasadil LM, Weaver BA. Living in suppresses cellular senescence induced by the c-myc
lymphomas. Blood. 2004;103:2351–2357. CIN: mitotic infidelity and its consequences for oncogene. Nat Cell Biol. 2010;12:54–59.
229. Morishita A, Masaki T, Yoshiji H, et al. Reduced expres- tumor promotion and suppression. Dev Cell. 274. Puyol M, Martin A, Dubus P, et al. A synthetic
sion of cell cycle regulator p18(INK4C) in human 2016;39:638–652. lethal interaction between K-Ras oncogenes and
hepatocellular carcinoma. Hepatology. 2004;40: 252. Sansregret L, Swanton C. The role of aneuploidy Cdk4 unveils a therapeutic strategy for non-small
677–686. in cancer evolution. Cold Spring Harb Perspect Med. cell lung carcinoma. Cancer Cell. 2010;18:63–73.
230. Wolfel T, Hauer M, Schneider J, et al. A p16INK4a- 2017;7:pii: a028373. doi:10.1101/cshperspect. 275. Landis MW, Pawlyk BS, Li T, et al. Cyclin
insensitive CDK4 mutant targeted by cytolytic a028373. D1-dependent kinase activity in murine develop-
T lymphocytes in a human melanoma. Science. 253. Thompson SL, Compton DA. Examining the link ment and mammary tumorigenesis. Cancer Cell.
1995;269:1281–1284. between chromosomal instability and aneuploidy in 2006;9:13–22.
231. Sotillo R, Garcia JF, Ortega S, et al. Invasive human cells. J Cell Biol. 2008;180:665–672. 276. Yu Q, Sicinska E, Geng Y, et al. Requirement for
melanoma in Cdk4-targeted mice. Proc Natl Acad 254. Ricke RM, van Ree JH, van Deursen JM. Whole CDK4 kinase function in breast cancer. Cancer Cell.
Sci USA. 2001;98:13312–13317. chromosome instability and cancer: a complex 2006;9:23–32.
232. Sotillo R, Dubus P, Martin J, et al. Wide spectrum relationship. Trends Genet. 2008;24:457–466. 277. Sawai CM, Freund J, Oh P, Ndiaye-Lobry D, Bretz
of tumors in knock-in mice carrying a Cdk4 255. Schvartzman JM, Sotillo R, Benezra R. Mitotic JC, Strikoudis A, et al. Therapeutic targeting of the
protein insensitive to INK4 inhibitors. EMBO J. chromosomal instability and cancer: mouse cyclin D3:CDK4/6 complex in T cell leukemia.
2001;20:6637–6647. modelling of the human disease. Nat Rev Cancer. Cancer Cell. 2012;22:452–465.
233. Gasparotto D, Maestro R, Piccinin S, et al. Over- 2010;10:102–115. 278. Malumbres M. Cell cycle-based therapies move
expression of CDC25A and CDC25B in head and 256. Cahill DP, Lengauer C, Yu J, et al. Mutations of forward. Cancer Cell. 2012;22:419–420.
neck cancers. Cancer Res. 1997;57:2366–2368. mitotic checkpoint genes in human cancers. Nature. 279. Asghar U, Witkiewicz AK, Turner NC, Knudsen ES.
234. Wu W, Fan YH, Kemp BL, et al. Overexpression 1998;392:300–303. The history and future of targeting cyclin-dependent
of cdc25A and cdc25B is frequent in primary non- 257. Lee H, Trainer AH, Friedman LS, et al. Mitotic kinases in cancer therapy. Nat Rev Drug Discov.
small cell lung cancer but is not associated with checkpoint inactivation fosters transformation in 2015;14:130–146.
overexpression of c-myc. Cancer Res. 1998;58:4082– cells lacking the breast cancer susceptibility gene, 280. Finn RS, Martin M, Rugo HS, Jones S, Im SA,
4085. Brca2. Mol Cell. 1999;4:1–10. Gelmon K, et al. Palbociclib and Letrozole in
Control of the Cell Cycle  •  CHAPTER 4 73.e5
73.e5

Advanced Breast Cancer. N Engl J Med. 2016;375: 290. Morgado-Palacin I, Day A, Murga M, Lafarga V, 301. Higgins JM. Haspin: a newly discovered regulator
1925–1936. Anton ME, Tubbs A, et al. Targeting the kinase of mitotic chromosome behavior. Chromosoma.
281. Patnaik A, Rosen LS, Tolaney SM, Tolcher AW, activities of ATR and ATM exhibits antitumoral 2010;119:137–147.
Goldman JW, Gandhi L, et al. Efficacy and safety activity in mouse models of MLL-rearranged AML. 302. Huang HC, Shi J, Orth JD, Mitchison TJ.
of abemaciclib, an inhibitor of CDK4 and CDK6, Sci Signal. 2016;9:ra91. Evidence that mitotic exit is a better cancer thera-
for patients with breast cancer, non-small cell lung 291. Karnitz LM, Zou L. Molecular pathways: target- peutic target than spindle assembly. Cancer Cell.
cancer, and other solid tumors. Cancer Discov. ing ATR in cancer therapy. Clin Cancer Res. 2009;16:347–358.
2016;6:740–753. 2015;21:4780–4785. 303. Zeng X, Sigoillot F, Gaur S, et al. Pharmacologic
282. Farmer H, McCabe N, Lord CJ, et al. Targeting 292. Dumontet C, Jordan MA. Microtubule-binding inhibition of the anaphase-promoting complex
the DNA repair defect in BRCA mutant cells as agents: a dynamic field of cancer therapeutics. Nat induces a spindle checkpoint-dependent mitotic
a therapeutic strategy. Nature. 2005;434:917– Rev Drug Discov. 2010;9:790–803. arrest in the absence of spindle damage. Cancer
921. 293. Komlodi-Pasztor E, Sackett D, Wilkerson J, Fojo Cell. 2010;18:382–395.
283. Bryant HE, Schultz N, Thomas HD, et al. T. Mitosis is not a key target of microtubule 304. Sackton KL, Dimova N, Zeng X, Tian W, Zhang M,
Specific killing of BRCA2-deficient tumours with agents in patient tumors. Nat Rev Clin Oncol. Sackton TB, et al. Synergistic blockade of mitotic exit
inhibitors of poly(ADP-ribose) polymerase. Nature. 2011;8:244–250. by two chemical inhibitors of the APC/C. Nature.
2005;434:913–917. 294. Mayer TU, Kapoor TM, Haggarty SJ, et al. Small 2014;514:646–649.
284. Garrett MD, Collins I. Anticancer therapy with molecule inhibitor of mitotic spindle bipolarity 305. Liu X, Winey M. The MPS1 family of protein
checkpoint inhibitors: what, where and when? Trends identified in a phenotype-based screen. Science. kinases. Annu Rev Biochem. 2012;81:561–585.
Pharmacol Sci. 2011;32:308–316. 1999;286:971–974. 306. Dominguez-Brauer C, Thu KL, Mason JM, Blaser
285. Toledo LI, Murga M, Fernandez-Capetillo O. Target- 295. Manchado E, Guillamot M, Malumbres M. H, Bray MR, Mak TW. Targeting mitosis in cancer:
ing ATR and Chk1 kinases for cancer treatment: Killing cells by targeting mitosis. Cell Death Differ. emerging strategies. Mol Cell. 2015;60:524–536.
a new model for new (and old) drugs. Mol Oncol. 2012;19:369–377. 307. Janssen A, Kops GJ, Medema RH. Elevating the
2011;5:368–373. 296. Wood KW, Chua P, Sutton D, Jackson JR. frequency of chromosome mis-segregation as a
286. Murga M, Campaner S, Lopez-Contreras AJ, et al. Centromere-associated protein E: a motor that puts strategy to kill tumor cells. Proc Natl Acad Sci USA.
Exploiting oncogene-induced replicative stress for the the brakes on the mitotic checkpoint. Clin Cancer 2009;106:19108–19113.
selective killing of Myc-driven tumors. Nat Struct Res. 2008;14:7588–7592. 308. Koch A, Maia A, Janssen A, Medema RH. Molecular
Mol Biol. 2011;18:1331–1335. 297. Wood KW, Lad L, Luo L, et al. Antitumor basis underlying resistance to Mps1/TTK inhibitors.
287. Cottini F, Hideshima T, Suzuki R, Tai YT, Bianchini activity of an allosteric inhibitor of centromere- Oncogene. 2016;35:2518–2528.
G, Richardson PG, et al. Synthetic Lethal Approaches associated protein-E. Proc Natl Acad Sci USA. 309. Manchado E, Malumbres M. Targeting aneuploidy
Exploiting DNA Damage in Aggressive Myeloma. 2010;107:5839–5844. for cancer therapy. Cell. 2011;144:465–466.
Cancer Discov. 2015;5:972–987. 298. Gjertsen BT, Schöffski P. Discovery and development 310. Williams BR, Prabhu VR, Hunter KE, et al.
288. Kwok M, Davies N, Agathanggelou A, Smith E, of the Polo-like kinase inhibitor volasertib in cancer Aneuploidy affects proliferation and spontaneous
Petermann E, Yates E, et al. Synthetic lethality in therapy. Leukemia. 2015;29:11–19. immortalization in mammalian cells. Science.
chronic lymphocytic leukaemia with DNA damage 299. Goga A, Yang D, Tward AD, et al. Inhibition of 2008;322:703–709.
response defects by targeting the ATR pathway. CDK1 as a potential therapy for tumors over- 311. Sheltzer JM, Amon A. The aneuploidy paradox:
Lancet. 2015;385(suppl 1):S58. expressing MYC. Nat Med. 2007;13:820–827. costs and benefits of an incorrect karyotype. Trends
289. Williamson CT, Miller R, Pemberton HN, Jones 300. Johnson N, Li YC, Walton ZE, et al. Compromised Genet. 2011;27:446–453.
SE, Campbell J, Konde A, et al. ATR inhibitors as CDK1 activity sensitizes BRCA-proficient cancers 312. Tang YC, Williams BR, Siegel JJ, Amon A. Iden-
a synthetic lethal therapy for tumours deficient in to PARP inhibition. Nat Med. 2011;17:875– tification of aneuploidy-selective antiproliferation
ARID1A. Nat Commun. 2016;7:13837. 882. compounds. Cell. 2011;144:499–512.
5  Pathophysiology of Cancer Cell Death
Lorenzo Galluzzi, Andreas Linkermann, Oliver Kepp, and Guido Kroemer

S UMMARY OF K EY P OI N T S
• Regulated cell death mainly occurs • Oncogenesis results from multiple fashion, the molecular machinery for
via extrinsic apoptosis, intrinsic molecular alterations, one of which apoptotic or necrotic cell death.
apoptosis, necroptosis, and frequently impairs the ability of • Targeting deregulated cell death
mitochondrial permeability transition cancer cells to die in response to signaling pathways in cancer
(MPT)–driven regulated necrosis. exogenous or endogenous signals. constitutes a clinical reality and
Autophagy operates as a bona fide • Several oncoproteins and underlies promising approaches for
cell death mechanism in a few, oncosuppressor proteins regulate, the development of novel anticancer
mostly developmental, settings. either directly or in an indirect regimens.

Perhaps biased by their focus on the cell’s vital functions, biologists hallmarks of cancer, irrespective of the histologic origin of malignant
have disregarded the existence of programmed cell death (PCD; see cells.16 During the subsequent two decades, it rapidly became clear
later for a definition) for a long time. Sporadic observations of PCD that defects in the signaling pathways leading to cell death not only
were made throughout the 19th century by scientists such as Carl contribute to oncogenesis and tumor progression, but also determine,
Vogt, August Weismann, Ludwig Stieda, Élie Metchnikoff, Walther at least in some instances, the resistance of neoplastic cells to chemo-
Flemming, Sigmund Mayer, and John Beard.1 The concept of PCD therapy and radiotherapy.17
was first theorized in the 1960s, thanks to the work of Sir Richard Our understanding of cell death is constantly advancing, and this
Lockshin.2 In 1972 John Kerr, Alastair Currie, and Andrew Wyllie knowledge has already been translated into multiple therapeutic
introduced the term apoptosis (from Greek apo, “from, off, without,” successes. Nevertheless, future investigations will have to provide deeper
and ptosis, “falling”) to describe one type of cell death that manifests insights into the cancer-associated defects of cell death, in all its forms.
with peculiar morphologic traits.3 At that time and for the subsequent
30 years, apoptosis was thought to be the only form of PCD, an FUNDAMENTAL SCIENCE: MECHANISMS OF
oversimplistic notion that has been invalidated only in this century.4,5 CELL DEATH
In the middle of the first decade of the century, it indeed became
clear that other subroutines of cell death, notably necrosis, can occur In cell death research, the attribute “programmed” is used to highlight
in a regulated fashion and account for some instances of PCD.6–8 the implication of one particular instance of cell death in developmental
Regulated cell death (RCD) constitutes a conserved mechanism programs (and hence its physiologic relevance), whereas the adjective
whose usefulness trespasses evolutionistic barriers.9,10 For instance, in “regulated” is employed to stress the notion that one particular instance
unicellular organisms that grow in colonies (such as yeast), the controlled of cell death can be inhibited by specific pharmacologic or genetic
demise of old and damaged cells increases the probability that fit interventions (and hence is mediated by genetically encoded molecular
individuals will survive adverse environmental conditions, and hence mechanisms).4 Thus all instances of PCD are regulated by definition,
will perpetuate their genes.11 Conversely, in metazoans (including but not vice versa. Additional recommendations on how to define
humans), PCD is critical for embryonic and postembryonic develop- specific cell death subroutines based on morphologic5 or biochemical4
ment, as well as for the maintenance of adult tissue homeostasis.12 In parameters have been formulated in the past few years, and will be
line with this notion, defects in the molecular mechanisms that mediate respected throughout this chapter. The Nomenclature Committee on
cell death contribute to the development of a wide array of human Cell Death (NCCD) has expressed doubts regarding the causal
diseases. On one hand, the excessive demise of postmitotic cells implication of specific biochemical processes in RCD.18 Indeed,
decisively contributes to the pathogenesis of diseases encompassing inhibiting the mechanisms that until now were considered to be the
ischemia (of the heart, brain, and kidney) and neurodegeneration. cause of RCD (in all its instances) generally delays, but does not
On the other hand, insufficient rates of cell death have been associated prevent, cell death (at least in mammalian organisms).18 This has
with autoimmune disorders and cancer.13 far-reaching conceptual and therapeutic implications, especially for
The first hint that the molecular machinery for cell death is involved the development of cytoprotective clinical interventions.18
in oncogenesis came in the mid-1980s, when it was found that a According to currently accepted models, there are two main types
translocation between chromosomes 14 and 18 (t14;18), which is of cell death: apoptosis and necrosis.19–23 Additional cell death sub-
common in lymphoma patients, leads to the overexpression of the routines with very specific biochemical traits have been described,
protein BCL2.14 Subsequent work clarified that BCL2 promotes although they often constitute particular cases of apoptosis and
lymphomagenesis by inhibiting the programmed demise of excessive necrosis.4,24,25 Macroautophagy (hereafter referred to as autophagy)
B cells rather than by stimulating their proliferation.15 This was the has also been implicated as a potential mechanism of cell death, a
first demonstration that disabled cell death constitutes one of the notion that remains highly debated (see later).26–28

74
Pathophysiology of Cancer Cell Death  •  CHAPTER 5 75

Apoptosis component of the mitochondrial respiratory chain), diablo IAP-binding


mitochondrial protein (DIABLO), and HtrA serine peptidase 2
For a long time, instances of cell death have been catalogued as (HTRA2).38–40 Once in the cytosol, CYCS—together with dATP and
apoptotic based on purely morphologic manifestations, including the adaptor protein apoptotic peptidase activating factor 1 (APAF1)—
cytoplasmic and nuclear shrinkage (pyknosis), nuclear breakdown drives the assembly of the apoptosome, a supramolecular platform
(karyorrhexis), and plasma membrane blebbing.5,29 This morphologic for the activation of CASP9.40 DIABLO and HTRA2 also stimulate
definition, reflecting the original observations by Kerr, Currie, and caspase activation, although indirectly, by sequestering and/or degrading
Wyllie,3 has been abandoned in favor of a biochemical one.4 Thus, cytosolic caspase inhibitors of the IAP family.38,39 Other mitochondrial
the term apoptosis is now used to define an instance of RCD that is proteins with cytotoxic potential are released in the cytosol during
precipitated by the activation of caspase 3 (CASP3), implying that it the course of intrinsic apoptosis, including apoptosis inducing factor,
can be delayed by chemical CASP3 inhibitors as well as by specific mitochondria associated 1 (AIFM1, which normally contributes to
genetic interventions that inactivate CASP3.18 Apoptosis can be triggered the stability and function of respiratory complex I) and endonuclease
by the activation of either of two distinct but not mutually exclusive G (ENDOG).41,42 Although both AIFM1 and ENDOG can translocate
signaling pathways. Thus, whereas extrinsic apoptosis is initiated by an to the nucleus and mediate large-scale DNA degradation independently
extracellular stimulus acting on plasma membrane receptors, intrinsic of caspases,42,43 their implication in intrinsic apoptosis has been
apoptosis follows the permeabilization of mitochondrial membranes dismissed.44,45
driven by an intracellular stimulus.4 Of note, the enzymatic activity According to current models, mitochondrial outer membrane
of CASP3, caspase 6 (CASP6), and caspase 7 (CASP7)—which are permeabilization (MOMP) is initiated at the mitochondrial outer
cumulatively known as executioner caspases—is responsible for multiple membrane (OM) by the pore-forming activity of proapoptotic
(but not all) classic morphologic and biochemical manifestations of multidomain proteins of the Bcl-2 family.19 These proteins, such as
apoptosis, including karyorrhexis, internucleosomal DNA degradation, BCL2 associated X, apoptosis regulator (BAX), and BCL2 antagonist/
and the exposure of the phospholipid phosphatidylserine on the outer killer 1 (BAK1), contain several Bcl-2 homology (BH) domains as
surface of the plasma membrane.30 well as a transmembrane domain that allow for their constitutive or
Extrinsic apoptosis is frequently elicited by the ligand-induced inducible insertion into the OM. MOMP execution by BAK1 and
activation of plasma membrane proteins of the death receptor family, BAX is regulated by other members of the same protein family. In
such as Fas cell surface death receptor (FAS) and the tumor necrosis particular, antiapoptotic proteins such as BCL2, apoptosis regulator
factor (TNF) superfamily member 1A (TNFRSF1A, best known as (BCL2), BCL2-like 1 (BCL2L1, best known as BCL-xL), and MCL1,
TNFR1).31 Alternatively, apoptotic signals can be transduced by the BCL2 family apoptosis regulator (MCL1) inhibit MOMP by binding
so-called dependence receptors (such as Patched 1, PTCH1) when to BAX and BAK1 and hence by maintaining them in an inactive
the extracellular concentration of trophic factors, such as Sonic conformation.46 Conversely, the so-called BH3-only proteins (small
Hedgehog (SHH), falls below a critical threshold level. This means members of the Bcl-2 family that often contain only the BH3 domain)
that, at odds with death receptors, dependence receptors deliver can promote the pore-forming activity of BAK1 and BAX by two
proapoptotic signals in the absence rather than in the presence of different mechanisms. Thus, BH3-only proteins can either stimulate
their ligands.32 Death receptors undergo spontaneous trimerization the conformational activation of BAX and BAK1 in a direct fashion
owing to the so-called preligand assembly domain (PLAD).33 Ligand or competitively displace BAX, BAK1 or other BH3-only proteins
binding stabilizes this configuration and hence allows for the recruitment from inhibitory interactions with BCL2, BCL-xL and MCL1. BH3-only
of several proteins at the cytoplasmic tails of death receptors, including proteins can be regulated at the transcriptional level as well as by
(but in some instances not limited to) pro-caspase 8 (pro-CASP8), rapid posttranslational modifications (e.g., phosphorylation, proteolytic
receptor interacting serine/threonine kinase 1 (RIPK1, also known as processing), de facto constituting sensors of intracellular stress that
RIP1), FAS-associated protein with a death domain (FADD), and directly impinge on the regulation of intrinsic apoptosis.47
cellular inhibitor of apoptosis proteins (cIAP1 and cIAP2, which are Of note, the signaling pathways leading to extrinsic and intrinsic
E3 ubiquitin ligases that also inhibit apoptosis owing to their ability apoptosis exhibit some degree of cross talk. Thus, whereas in some
to interfere with caspase activation).34 This multiprotein platform is cells including lymphocytes (type I cells) the activation of death
known as death-inducing signaling complex (DISC) and stimulates receptors leads to pro-CASP3 processing and apoptosis without any
the conversion of pro-CASP8 into caspase 8 (CASP8), which proteoliti- mitochondrial involvement,48 in other cells such as hepatocytes (type
cally activates CASP3 and hence initiates the degradative cascade that II cells), CASP8 not only activates CASP3 but also mediates the
precipitates extrinsic apoptosis.35 Of note, the components of the proteolytic cleavage of the BH3-only protein BH3 interacting domain
signaling pathways that emanate from specific death receptors exhibit death agonist (BID), generating a MOMP-inducing fragment.49 Thus,
some degree of variation, yet all converge (in apoptosis-permissive in type II cells, MOMP functions as an amplifier of apoptotic signaling
conditions; see later) to the activation of CASP8. The molecular by eliciting the CASP9-mediated activation of CASP3. Interesting to
mechanisms that link dependence receptors to CASP3 activation are note, whether cells respond to death receptor ligation in a type I- or
not precisely understood, yet appear to involve the adaptor protein type II-like manner depends on the expression levels of the cIAP-like
DRAL and CASP9 (Fig. 5.1).36 protein X-linked inhibitor of apoptosis (XIAP).50 The cross talk between
Intrinsic apoptosis (also known as mitochondrial apoptosis) can be extrinsic and intrinsic apoptosis is pathophysiologically relevant in
activated by a plethora of stimuli, including intracellular damage (to vivo, as demonstrated by the fact that the hepatocytes of Bid−/− mice
virtually any of the subcellular compartments) and oncogenic stress are partially protected from FAS-induced apoptosis (see Fig. 5.1).51
(see later). Cells are equipped with a heterogeneous set of intracellular
sensors that respond to microenvironmental perturbations by activating Necrosis
signaling pathways for the reestablishment of homeostasis and the
repair of damage, and then, if damage is irreparable, by igniting intrinsic Classically, necrosis was defined as an instance of cell death lacking
apoptosis.37 The central step in this cascade is regulated by the integrity the peculiar morphological manifestations of apoptosis as well as the
of mitochondrial structure and function. Indeed, if proapoptotic signals accumulation of cytoplasmic vacuoles that characterize autophagic
are predominant, mitochondrial membranes get permeabilized, resulting cells. Somehow, this was in line with the belief that necrosis would
in the abrupt cessation of adenosine triphosphate (ATP) synthesis and always proceed in an unregulated fashion and would only terminate
other metabolic functions, as well as in the spillage of several proteins accidental instances of cell death.5 In the 1990s, Vandenabeele’s and
that are normally confined in the mitochondrial intermembrane space.37 Schulze-Osthoff ’s groups demonstrated that engagement of FAS does
These proteins include cytochrome c, somatic (CYCS, a semisoluble not always lead to cell death via extrinsic apoptosis.52–54 This observation
76 Part I: Science and Clinical Oncology

Figure 5.1  •  Apoptosis. Extrinsic apoptosis is often ignited by the ligation of death receptors such as FAS. This allows for the stabilization of receptor
trimers and for recruitment at their intracellular tails of a multiprotein complex known as death-inducing signaling complex (DISC). Within the DISC,
pro-CASP8 undergoes spatial-proximity induced autoactivation, hence becoming able to cleave multiple substrates including BID and pro-CASP3. As an
alternative, extrinsic apoptosis can be initiated by so-called “dependence receptors,” such as PTCH1, when the concentrations of their ligands (e.g., SHH)
falls below a specific threshold. In this case, the activation of CASP3 proceeds via a molecular mechanism involving CASP9 and the adaptor proteins DRAL
and TUCAN. Intrinsic apoptosis is activated in response to intracellular stress conditions (e.g., DNA damage) and involves a central step of mitochondrial
regulation. Thus, if proapoptotic signals (often relayed by BH3-only proteins) predominate over antiapoptotic ones, mitochondrial membranes lose their
structural integrity because of the pore-forming activity of Bcl-2 family members such as BAK1 and BAX. Consequent mitochondrial outer membrane per-
meabilization (MOMP) allows for the release of mitochondrial proteins into the cytosol, including direct activators of caspases, such as CYCS, as well as
proteins that indirectly facilitate caspase activation, such as DIABLO and HTRA2. Cytosolic CYCS directs the assembly of an APAF1-containing, dATP-dependent
supramolecular platform that catalyzes CASP9 activation, hence initiating a proteolytic cascade that culminates in CASP3 (CASP6 and CASP7) activation.
Extrinsic apoptosis and intrinsic apoptosis exhibit some degree of cross talk. Indeed, in some cell types, CASP8 can convert the BH3-only protein BID into
a MOMP-promoting fragment, further accelerating caspase activation and apoptosis. FADD, FAS-associated protein with a death domain; FASLG, FAS ligand;
IAPs, inhibitor of apoptosis proteins; tBID, truncated BID.

instilled in some researchers the suspicion that, similar to apoptosis, conditions in which CASP8 is inhibited (either by pharmacologic or
necrosis also might be orchestrated by a refined molecular machinery; by genetic interventions). In this context, DISC-bound RIPK1 does
this ignited an intense wave of research that has not yet come to an not get degraded by CASP8 as it occurs during extrinsic apoptosis,
end.6 Important to note, regulated necrosis occurs during mammalian but recruits and functionally interacts with its homolog receptor
development, in particular at the bone growth plate (i.e., the zone of interacting serine/threonine kinase 3 (RIPK3) and mixed-lineage kinase
the bone that controls its length), as well as during adult tissue like (MLKL), generating the so-called necrosome.58–60 Although PGAM
homeostasis, for instance in the lower regions of intestinal crypts.55,56 family member 5, mitochondrial serine/threonine protein phosphatase
Moreover, multiple instances of necrotic RCD have been involved in (PGAM5) has been suggested to contribute to necroptosis downstream
the pathophysiology of diseases including viral infection, neurodegenera- of RIPK1 and RIPK3 by virtue of its capacity to promote mitochondrial
tion, ischemia, and others.8,22,57 fragmentation,61,62 such a possibility has now been discarded.63,64 Rather,
it is now clear that phosphorylated MLKL can form oligomers that
Necroptosis relocalize to the inner leaflet of the plasma membrane and compromise
The best-characterized pathway of regulated necrosis, which is known its stability, hence precipitating RCD.65–71 Whether phosphorylated
as necroptosis, can be elicited by the ligation of death receptors in MLKL is sufficient to permeabilize the plasma membrane or whether
Pathophysiology of Cancer Cell Death  •  CHAPTER 5 77

additional factors are required for necroptosis execution remains a dissipation of the mitochondrial transmembrane potential (Δψm), rapid
matter of debate. RIPK3 activation is tonically inhibited by a het- cessation of all Δψm-dependent mitochondrial activities (including
erodimer composed of CASP8 and the long isoform of CASP8 and ATP synthesis and protein import), and osmotic breakdown of the
FADD-like apoptosis regulator (CFLAR), best known as FLIPL, but organelle.77,78 MPT has been ascribed to the activity of a multiprotein
the underlying molecular mechanisms remain to be elucidated.72 protein complex that is assembled at the juxtaposition sites between the
Moreover, RIPK3 oligomerization and consequent phosphorylation OM and the IM, the so-called permeability transition pore complex
of MLKL is controlled by the RIP homotypic interacting motif (RHIM) (PTPC).79 The main components of the PTPC, including members of
of RIPK1, the absence of which results in spontaneous necroptosis.73,74 the voltage-dependent anion channel (VDAC) family (located in the
Important to note, stimuli other than death receptor ligands (e.g., OM) and the adenine nucleotide translocase (ANT) family (located
alkylating DNA damage, viral infection) trigger bona fide RIPK3- and in the IM), as well as peptidylprolyl isomerase F (PPIF, a protein of
MLKL-dependent necroptosis (Fig. 5.2).75,76 the mitochondrial matrix best known as CYPD), normally mediate
physiologic functions. For instance, ANT catalyzes the exchange of
Mitochondrial Permeability Transition–Driven adenosine diphosphate (ADP) and ATP between the cytosol and the
mitochondrial matrix. In response to cytosolic Ca2+ overload oxidative
Regulated Necrosis stress, the PTPC has been proposed to assume a high-conductance
In conditions of intense oxidative stress and/or in the presence of conformation leading to rapid mitochondrial breakdown.37 So far,
high cytosolic Ca2+ concentrations, mitochondria experience an mouse knockout experiments failed to attribute to specific compo-
abrupt increase in the permeability to ions and small solutes of the nents of the PTPC a critical role in the regulation of MPT, perhaps
mitochondrial inner membrane (IM), a process known as mitochondrial because of the existence of multiple and (at least partially) redundant
permeability transition (MPT).77,78 MPT results in virtually immediate isoforms of these proteins.80–82 One notable exception is represented by
CYPD, whose absence has been shown to limit pathologic cell death
in multiple circumstances, in vitro and in vivo.83,84 Thus MPT-driven
necrosis can be defined biochemically as a form of RCD that can be
retarded by pharmacologic or genetic inhibition of CYPD.18 Recent
findings suggest that the c subunit of the F1FO ATPase (which in
humans is encoded by ATP5G1, ATP5G2, and ATP5G3) may constitute
the long-sought pore-forming component of the PTPC,85 but robust
genetic evidence in support of this hypothesis is missing (Fig. 5.3).77,78

Other Forms of Regulated Cell Death


Several other specific instances of apoptotic and necrotic RCD have
been described throughout the past two decades, most of which
eventually impinge on the core molecular machinery of apoptosis
(CASP3 activation), necroptosis (RIPK3 and MLKL activation), or
MPT-driven regulated necrosis (CYPD activation).23 Of all such specific
instances, we find of particular relevance (because of their peculiar
mechanistic aspects and pathophysiologic implications) ferroptosis,
pyroptosis, and parthanatos.

Figure 5.2  •  Necroptosis. Necroptosis can be triggered by the ligation of


TNFR1 when caspases (notably CASP8) are inactive (for instance, owing to
the presence of the pan-caspase inhibitor Z-VAD-fmk). In these conditions, Figure 5.3  •  MPT-driven regulated necrosis. In response to oxidative stress
RIPK1 is deubiquitinated and engages in physical and functional interactions or cytosolic Ca2+ overload, the so-called permeability transition pore complex
with its homolog RIPK3 as well as with MLKL in the context of a supramo- (PTPC) assumes a high-conductance conformation that allows for the
lecular entity called necrosome. Phosphorylated MLKL forms oligomers that unregulated entry of solutes and water into the mitochondrial matrix. This
precipitate necroptosis by relocalizing at the inner leaflet of the plasma membrane CYPD-dependent process causes a structural and functional breakdown of
and favoring its irreversible permeabilization. CYLD, CYLD lysine 63 deu- the mitochondrial network that rapidly seals the cell fate. Δψm, Mitochondrial
biquitinase; FADD, FAS-associated protein with a death domain; CFLARL, transmembrane potential; MPT, mitochondrial permeability transition; IM,
CASP8 and FADD like apoptosis regulator, long isoform; Ub, ubiquitin, inner mitochondrial membrane; OM, outer mitochondrial membrane; RN,
TNF, tumor necrosis factor; TRADD, TNFRSF1A associated via death domain. regulated necrosis.
78 Part I: Science and Clinical Oncology

inadvertently suggests a cause-effect relationship between these two


Ferroptosis phenomena.26 Thus far, autophagy has been demonstrated to mediate
Ferroptosis is an iron-dependent form of RCD that involves the cell death in several developmental scenarios, notably during the
depletion of intracellular antioxidants including reduced glutathione metamorphosis of insects.111,112 Moreover, at least in some instances,
(GSH), and consequent lethal lipid peroxidation.86–88 Ferroptosis is autophagy appears to contribute to the execution of human cancer
tonically inhibited by glutathione peroxidase 4 (GPX4), a major cells that succumb to specific experimental cell death inducers in
gatekeeper of intracellular redox balance.89,90 Moreover, various vitro.113 These observations de facto justify the use of the expression
molecules cumulatively known as ferrostatins have been shown to autophagic cell death under highly selected circumstances.4 Defects of
retard ferroptosis in vitro and in vivo, presumably as they inhibit lipid the autophagic machinery have been associated with a plethora of
peroxidation by lipoxygenases.86,91 Ferroptosis appears to be particularly human pathophysiologic conditions, including accelerated aging,
relevant for the synchronized demise of renal tubules in the context neurodegeneration, and cancer.114,115 However, this appears to be more
of acute kidney injury.92,93 strictly related to the role that autophagy exerts in the regulation of
intracellular homeostasis rather than as a bona fide cell death mecha-
Pyroptosis nism28,107 and hence is not discussed here in further detail.
For a long time, pyroptosis was defined as a cell death modality
associated with the secretion of interleukin (IL)-1B and IL-18,94 two FUNDAMENTAL SCIENCE: CELL DEATH
key mediators of systemic inflammation.95 Thus, pyroptosis was thought AND CANCER
to originate from, or at least to be associated with, the activation of
so-called inflammatory caspases, that is, caspase 1 (CASP1), caspase According to classic models, single molecular alterations are per se
4 (CASP4), caspase 5 (CASP5), or mouse caspase 11 (Casp11, the unable to fully transform normal cells into highly aggressive cancer
murine orthologue of human CASP4 and CASP5).96 Study findings cells. Rather, oncogenesis seems to proceed along with a progressive
have demonstrated that inflammatory caspases proteolytically cleave increase in genetic instability and with the accumulation of several
gasdermin D (GSDMD) to generate an N-terminal fragment that molecular defects. Often, if not always, one of these alterations consists
precipitates pyroptosis by forming pores in the plasma membrane on in the interruption of the signaling cascades that ensure the homeostatic
interaction with phosphoinositides.97–101 Thus pyroptosis should now death of continuously proliferating cells.116 As they evolve, premalignant
be defined as a GSDMD-dependent instance of RCD.102 and malignant cells are indeed subjected to elevated levels of stress,
in part as a result of the overactivation of cell-intrinsic oncogenic
Parthanatos signaling pathways (so-called oncogenic stress), and in part due to
Alkylating DNA damage activates repair mechanisms involving the microenvironmental conditions, which are often characterized by
NAD+-dependent enzyme poly (ADP-ribose) polymerase 1 (PARP1).103 hypoxia and nutrient shortage (especially in rapidly proliferating
Catalytically active PARP1 recruits multiple components of the DNA neoplasms).117 Thus, in virtually all scenarios, carcinogenesis requires
repair machinery, hence facilitating the recovery of genetic homeo- the (at least partial) suppression of cell death signaling pathways. This
stasis.103 However, in the context of irreparable alkylating DNA damage, can result from loss-of-function mutations in proteins that transduce
PARP1 hyperactivation has lethal consequences for the cell because lethal signals or execute cell death (as many oncosuppressor proteins),
(1) it favors a nonrecoverable NAD+ depletion that culminates in but also from gain-of-function alterations in molecules that normally
irreversible bioenergetic crisis, and (2) it promotes the selective release deliver pro-survival signals (as several oncoproteins). Defects in the
of AIFM1 from mitochondria, thereby unleashing its concealed molecular pathways that regulate cell death also are instrumental to
nucleolytic activity.104,105 Parthanatos can be defined as a PARP1- tumors when it comes to resistance to chemotherapy and radiotherapy.17
dependent form of RCD. Important to note, intrinsic alterations are generally insufficient for
a healthy cell to form a clinically manifest aggressive neoplasm, owing
Autophagy to the existence of a systemic layer of control on cellular homeostasis
mediated by the immune system (so-called “cancer immunosurveil-
Autophagy entails the engulfment of intracellular structures (including lance”).118 For the sake of simplicity, however, the cell-extrinsic regula-
organelles, protein aggregates, and portions of cytoplasm) by double- tion of oncogenesis and tumor progression is not discussed in further
membraned vacuoles called autophagosomes.106 Autophagosomes detail here.
normally fuse with lysosomes, leading to the degradation of their
content by lysosomal hydrolases. Baseline levels of autophagy contribute Oncogenes and Cell Death Regulation
to the maintenance of intracellular homeostasis by ensuring the removal
of old and damaged (and hence potentially dangerous) organelles, Oncogenes—that is, genes that stimulate malignant transformation—
notably mitochondria.107,108 In addition, autophagy is upregulated in were originally identified in tumorigenic viruses and then were shown
response to a wide array of stressful conditions, including nutrient to exist as inactive variants (or proto-oncogenes), also in the human
deprivation, hypoxia, and the presence of xenobiotics, including multiple genome.119 Proto-oncogenes including MYC and NRAS are involved
anticancer agents.109 in the regulation of mitogenic signals and hence play a critical role
For a while, it was thought that the continuative activation of in the control of tissue homeostasis. Per se, proto-oncogenes are not
autophagy would eventually lead to the exhaustion of cellular resources tumorigenic and must acquire gain-of-function alterations to become
and cell death. Such an “autophagic cell death” was defined morphologi- so. These alterations can be as gross as chromosomal translocations
cally by an extensive vacuolization of the cytoplasm, representative of that bring proto-oncogene coding sequences in the proximity of strong
an elevated number of autophagosomes and autolysosomes (the transcriptional regulators (as in the case of MYC, which is often rear-
organelles that are generated by the autophagosomal-lysosomal ranged in lymphoma patients), or as specific as point mutations that
fusion).5,26 However, the association between the accumulation of render proto-oncogene products constitutively active (as in the case
autophagosomes and cell death has rarely if ever been proved to be of NRAS, which is affected by hyperactivating mutations in 20%
causal, in particular in settings of stress-induced cancer cell death. to 25% of all cancers).120,121 By transducing strong and persistent
Indeed, the inhibition of autophagy by pharmacologic or genetic mitogenic signals, constitutively active MYC and NRAS, in addi-
means often accelerates (rather than inhibits) cell death, suggesting tion to other oncoproteins such as epidermal growth factor receptor
that autophagy constitutes a stress response mechanism that attempts (EGFR, which is often hyperactivated in lung and colon cancer)
(but fails) to avoid the cellular demise.110 These results cast doubts and Abelson tyrosine kinase (ABL, which is fused to BCR on the
on the appropriateness of the term autophagic cell death, which t(9;22) translocation in chronic myelogenous leukemia), facilitate the
Pathophysiology of Cancer Cell Death  •  CHAPTER 5 79

escape of malignant precursors from the growth control that usually their proapoptotic counterparts into inactive complexes, in addition
guarantees tissue homeostasis. As mentioned earlier, however, one to Ca2+-related and metabolic effects (Fig. 5.4).46,122,123 After the
single alteration of this kind is insufficient to convert normal cells discovery that the overexpression of BCL2 as a result of the t(14;18)
into their malignant counterparts. The hyperactivation of mitogenic translocation underlies multiple instances of lymphomagenesis,14
pathways causes consistent functional alterations and drives cells into preclinical and clinical evidence has accumulated to demonstrate that
a state of constant stress featuring significant metabolic rewiring and not only BCL2 but also its antiapoptotic homologues BCL-xL and
increased generation of reactive oxygen species. Because oncogenic MCL1 exert bona fide oncogenic functions.124 The overexpression of
stress would normally lead to cell death, only premalignant cells in BCL2 shortens the latency period required for transgenic mice
which the cell death signaling pathways are blocked can progress overexpressing MYC in B cells to develop lymphomas.15 Along similar
toward tumorigenesis.116 lines, both BCL-xL and MCL1 have been shown to cooperate with
Premalignant cells resist oncogenic stress by activating one (or MYC in murine models of hematopoietic malignant transforma-
more) of several pathways that (1) directly counteract the execution tion.125,126 However, consistent with the notion of multistep oncogenesis
of cell death (because of the deregulation of additional proto-oncogenes) described earlier, mice engineered for the overexpression of BCL2,
or (2) facilitate the management of intracellular stress (because of the BCL-xL, or MCL1 in hematopoietic cells exhibit perturbed myelopoieis
elicitation of per se nononcogenic stress response pathways). In general, or lymphopoieis but are only slightly more prone to the development
this leads to two distinct phenomena of dependence, commonly referred of age-related lymphomas than their normal counterparts.125,127,128 Of
to as “oncogene addiction” and “non–oncogene addiction.”117 In the note, the protein expression levels of BCL2, BCL-xL, and MCL1 are
former scenario, not only the tumorigenic phenotype but also the elevated in a wide range of human tumors.129
survival of cancer cells becomes dependent on the hyperactivation of NF-κB is a transcription factor that participates in the cellular
one or more proto-oncogenes. This has provided a rationale for the response to a wide array of conditions, including (but not limited to)
use of multiple oncogene-targeted agents in anticancer therapy, some cytokine stimulation, infection, and oxidative stress. Under normal
of which (such as the BCR-ABL–targeting compound imatinib) have circumstances, NF-κB subunits such as p65RELA and p50NFKB1 are held
impressive rates of therapeutic responses.116 In the latter scenario, in check in the cytoplasm by inhibitory interactions with IκBα. In
cancer cells become dependent on the activation of per se nononcogenic the presence of NF-κB–inducing conditions, however, IκBα is
mechanisms of intracellular stress management, such as those centered phosphorylated by the so-called IκB kinase (IKK, a multiprotein
around molecular chaperones of the heat-shock protein (HSP) family.117 complex including two catalytic subunits [IKKα and IKKβ] and one
Major examples of prosurvival systems that often get hyperactivated regulatory component [IKKγ/NEMO]), resulting in its proteosomal
throughout (and contribute to) oncogenesis are provided by antiapop- degradation and in the nuclear translocation of NF-κB.130 NF-κB
totic Bcl-2 family members, the nuclear factor–κB (NF-κB) pathway, homodimers and heterodimers control the expression of more than
and AKT1. 150 target genes, including genes that code for mitogens, other
Antiapoptotic Bcl-2 family members have been shown to regulate transcription factors (including MYC), inhibitors of extrinsic apoptosis
cell death by multiple mechanisms, including the sequestration of (e.g., the so-called FLIPs), IAPs, and BCL2 and BCL-xL (Fig. 5.5).131

Figure 5.4  •  Bcl-2 family proteins. Human Bcl-2 family proteins. Official gene names (within brackets) and MW are reported. BH, Bcl-2 homology
domain; TM, transmembrane domain.
80 Part I: Science and Clinical Oncology

Figure 5.5  •  The nuclear factor–κB (NF-κB) canonic pathway. In the


canonic activation pathway (a “noncanonic” pathway exists but is not described
here for the sake of simplicity), the IκB kinase (IKK) complex (comprising
one regulatory subunit, known as IKKγ or NEMO, and two catalytic subunits,
IKKα and IKKβ) responds to specific signals (including the ligation of death Figure 5.6  •  AKT1 signaling. Several growth factor receptors are
receptors) or nonspecific stress by phosphorylating IκB, thereby targeting it coupled to phosphoinositide-3-kinase (PI3-K), catalyzing the conversion
for proteosomal degradation. The degradation of IκB unmasks a nuclear of membrane-bound phosphatidylinositol-4,5-diphosphate (PIP2) into
localization signal that allows preformed NF-κB homodimers and heterodimers phosphatidylinositol-3,4,5-triphosphate (PIP3). By binding to a pleckstrin
to enter the nucleus and bind DNA. In the nucleus, NF-κB dimers control homology (PH) domain, PIP3 recruits AKT1 at the intracellular leaflet of the
the expression of genes involved in several distinct pathophysiologic processes, plasma membrane, hence allowing for its phosphorylation-dependent activation
including innate and adaptive immunity, inflammation, proliferation, and by phosphoinositide-dependent kinase 1 (PDK1). Active AKT1 phosphorylates
cell death and survival. P, Phosphate; Ub, ubiquitin. multiple substrates, thereby transducing multipronged prosurvival signals. For
instance, AKT1 inhibits the BH3-only protein BAD by facilitating its sequestra-
tion by 14-3-3 proteins, stimulates the degradation of the oncosuppressive
transcription factor p53, promotes glucose uptake (hence favoring the
Thus, the activation of NF-κB orchestrates the response of cells to maintenance of bioenergetic such as FKHRL1. Moreover, AKT1 indirectly
stress while delivering a strong prosurvival (and in some cases mitogenic) activates the mechanistic target of rapamycin (MTOR), hence stimulating
signal. Multiple lines of evidence demonstrate that the NF-κB system cell growth and proliferation while inhibiting autophagy. By catalyzing the
exerts bona fide oncogenic functions. First, REL genes are homologues dephosphorylation of PIP3, phosphatase and tensin homolog (PTEN) function-
of the avian reticuloendotheliosis virus v-rel oncogene, which is strictly ally antagonizes PI3-K, de facto inhibiting AKT1 activation. P, Phosphate.
required for virus-induced malignant transformation.132,133 Second,
the human leukemia/lymphoma virus type I (HTLV1) can transform
target cells via a mechanism that depends, at least in part, on the
activation of cellular IKK by the viral protein Tax.134 Finally, several substrates, AKT1 transduces prosurvival signals via distinct mechanisms.
components of the NF-κB pathway get overexpressed or hyperactivated Among others, AKT1 inhibits the BH3-only protein BCL2-associated
during malignant transformation. For instance, REL genes are amplified agonist of cell death (BAD) by facilitating its sequestration by chap-
or affected by gain-of-function mutations in a constituent proportion erones of the 14-3-3 protein family, stimulates the degradation of the
of B-cell lymphomas,135 and the IκB-related protein BCL3 (which oncosuppressor tumor protein p53 (TP53, best known as p53; see
stimulates the nuclear translocation and activity of NF-κB dimers)136 later), promotes glucose uptake and hence favors the maintenance of
is involved in a chromosomal rearrangement, t(14;19), that is found bioenergetic homeostasis, and blocks the nuclear translocation of
in some hematologic malignancies.137 Of note, the NF-κB system proapoptotic Forkhead transcription factors such as Forkhead box O3
appears to play a critical role in the handling of oncogenic stress, as (FOXO3).141 Moreover, AKT1 indirectly activates mechanistic target
demonstrated by the fact that multiple oncoproteins including RAS of rapamycin (MTOR), thus stimulating cell growth and proliferation
and BCR-ABL stimulate NF-κB nuclear translocation and transcrip- while inhibiting autophagy (Fig. 5.6).142 Several preclinical and clinical
tional activity.138,139 studies have demonstrated the importance of AKT1 in tumorigenesis.
One of the hallmarks of cancer cells as originally formulated by Transgenic mice expressing constitutively active Akt1 under the control
Hanahan and Weinberg in 2000 consists of their ability to circumvent of a T cell–restricted promoter develop lymphoma early in life.143
the absence of extracellular growth factors and de facto emancipate Amplification of AKT1 or gain-of-function AKT1 mutations have
from homeostatic tissue regulation.140 Several growth factors, including been detected in several neoplasms, including (but not limited to)
IL-2, platelet-derived growth factor (PDGF), and insulin-like growth breast and gastric cancers. In addition, loss-of-function of the oncosup-
factor 1 (IGF1), promote proliferation and survival via transmembrane pressor PTEN (the phosphatase that directly antagonizes PI3-K and
receptors that activate phosphoinositide-3-kinases (PI3-Ks, also known hence inhibits AKT1, see later) is common across a wide array of
as phosphatidylinositol-3 kinases). Active PI3-K generates plasma tumors.144 Of note, the inhibition of MTOR limits tumor formation
membrane-bound phosphatidylinositol-3,4,5-triphosphate, in turn in Pten+/− mice,145 demonstrating that AKT1-driven oncogenesis relies,
stimulating the activation of the serine/threonine kinase AKT1 (also at least in part, on MTOR hyperactivation. This is particularly interest-
known as protein kinase B [PKB]). By phosphorylating multiple ing in view of the fact the MTOR inhibition stimulates autophagy,
Pathophysiology of Cancer Cell Death  •  CHAPTER 5 81

which is considered as a bona fide oncosuppressor mechanism during p53 was originally identified in 1979 by two methodologically
early tumorigenesis.146 distinct approaches: as a cellular protein coimmunoprecipitating with
the large-T antigen in SV40-transformed cells,161,162 and as the target
Oncosuppressors and Cell Death Regulation of antibodies isolated from the serum of immunodeficient mice bearing
human tumors.163,164 Since then, p53 has been subjected to an intense
The existence of tumor suppressor genes (also known as oncosuppressor wave of investigation, leading to the discovery that p53 regulates an
genes)—that is, genes whose products actively counteract tumorigenesis ever-growing list of cellular processes.165 Historically, the first function
—was first suspected in the 1960s, when Harris and colleagues observed ascribed to p53 was that of a stress-responsive transcription factor. In
that highly malignant Ehrlich ascites cells lose the ability to form physiologic conditions, the intracellular levels of p53 are regulated by
tumors on fusion with virtually nontumorigenic cells.147 The molecular MDM2, an E3 ubiquitin ligase that polyubiquitinates p53 and hence
characterization of the first tumor suppressor gene, however, lagged targets it to proteosomal degradation.166 In response to a variety of
until the 1980s, when the cDNA encoding the protein product of adverse conditions, including DNA damage and oncogenic stress,
the RB1 gene (a negative regulator of cell cycle progression involved p53 escapes MDM2-mediated degradation and accumulates in the
in the development of pediatric retinoblastoma) was isolated and cytoplasm. This can be due to posttranslational modifications of p53
sequenced.148 Since then, several other oncosuppressor proteins have that reduce its affinity for MDM2 or to the expression of MDM2
been identified, encompassing negative regulators of mitogenic signals, inhibitors such as p14ARF.167 Irrespective of the underlying molecular
such as PTEN; proapoptotic Bcl-2 family members such as BAX; and mechanisms, stabilized p53 assembles into transcriptionally proficient
proteins that couple the management of cellular stress with cell death tetramers that can regulate the expression of distinct sets of genes,
induction, such as ATM and p53. Oncosuppressor genes are inactivated leading to highly diverse functional outcomes.168 Among the most
during oncogenesis not only with loss-of-function mutations and common p53-regulated responses are reversible cell cycle arrest,
deletions, but also after epigenetic gene silencing. Thus, promoter senescence, and cell death. Cell death is accomplished via the upregula-
hypermethylation at CpG islands or histone deacetylation can turn tion of multiple genes involved in the execution of apoptosis, including
off oncosuppressor gene expression, owing to local chromatin remodel- APAF1, BAX, FAS, and the BH3-only protein BCL2 binding component
ing and impaired access by essential transcription factors.149 These 3 (BBC3, best known as PUMA),168–171 as well as via transcription-
instances are exemplified by the loss of CASP8 expression in pediatric independent mechanisms whereby cytoplasmic p53 stimulates MOMP
neuroblastomas or by the loss of p14ARF (an upstream activator of by interacting with both proapoptotic and antiapoptotic members of
p53; see later) expression in multiple tumor types.150,151 Irrespective the Bcl-2 family or MPT by interacting with CYPD (Fig. 5.7).172–176
of the underlying mechanisms, both alleles of one tumor suppressor The inactivation of p53, be it mutational, due to the overexpression
gene must be inactivated for the establishment of tumor-permissive or overactivation of p53 negative regulators such as MDM2, or resulting
conditions. In this context, germline mutations in oncosuppressor from the inactivation of upstream p53-activating factors such as p14ARF,
genes are associated with familial tumor syndromes (as one copy of is the most common molecular alteration in human cancer, affecting
the gene is lost in the whole organism), whereas somatic mutations more than 50% of neoplasms, all confounded. In addition, the p53
are frequently detected in sporadic tumors (in this case, both alleles status has been shown to influence prognosis and/or therapeutic
have been inactivated during tumorigenesis).152 outcome in multiple oncologic settings, including (but not limited
As mentioned earlier, the protein product of PTEN (phosphatase to) breast, lung, and colorectal cancer.177 These clinical data reflect a
and tensin homolog) functions as a phosphatase, catalyzing the conver- huge number of preclinical observations demonstrating that p53 exerts
sion of phosphatidylinositol-3,4,5-triphosphate into phosphatidylinositol- bona fide oncosuppression in vivo.178 Of note, recent research has
4,5-diphosphate and de facto antagonizing the enzymatic activity of focused on physiologic aspects of the p53 biology, unveiling a critical
PI3-Ks and negatively regulating AKT1 (see Fig. 5.6).153 PTEN was role for baseline p53 levels in the maintenance of bioenergetic, redox,
discovered in the search for a tumor suppressor on chromosome 10 and genomic homeostasis.179–182 Thus, p53 appears to exert oncosup-
that is often lost in glioblastoma and breast, endometrial, and prostate pressive functions both as it regulates intracellular homeostasis, thereby
cancer.154 Subsequent preclinical and clinical observations supported preventing malignant transformation, and as it orchestrates the
the notion that PTEN loss of function sustains tumorigenesis in many elimination of potentially tumorigenic cells.
different organs.155,156 The oncosuppressive functions of PTEN have
been shown to be particularly sensitive to gene dosage, even beyond CLINICAL RELEVANCE AND APPLICATIONS
the classic concept of haploinsufficiency.157 Of note, the frequency of
PTEN inactivation in human tumors is exceeded only by that of p53 As introduced earlier, genetic, epigenetic, and functional alterations
(see later).144 of the mechanisms that underlie RCD (1) are highly prevalent in
Pro-apoptotic BCL-2 family members, in particular BAX and BAK1, human cancer, (2) are often required for oncogenesis and/or tumor
exert crucial oncosuppressive functions as they precipitate RCD in progression, and (3) underlie many instances of cancer chemoresistance
response to adverse conditions, including oncogenic stress (see Fig. and radioresistance. The clinical relevance of these alterations is therefore
5.4). Results from multiple murine models support the notion that dual. On one hand, the characterization of the genotype and functional
BAX and BAK1 operate in vivo to counteract oncogenesis independent phenotype of specific neoplasms can provide prognostic and/or predic-
of p53. For instance, murine cells expressing adenoviral E1A (a prolifera- tive information. This allows for the stratification of patients into
tive factor) and dominant negative p53 are unable to form tumors in precise risk groups and—at least in some instances—for the implementa-
mice unless Bax and Bak1 are lost.158 In addition, loss-of-function tion of personalized therapeutic regimens.183 On the other hand, a
mutations in BAX and BAK1 have been detected in many hematopoietic great number of anticancer agents that specifically target alterations
and solid malignancies.129,159 By acting as stress sensors that activate in cell death–regulating signaling pathways have been developed, and
intrinsic apoptosis, BH3-only proteins could also exert, at least theoreti- some of them have successfully entered clinical routines. At odds with
cally, prominent oncosuppressive functions. This said, evidence conventional chemotherapeutics, which frequently kill tumor cells
implicating them as bona fide tumor suppressors is scant, and only a because of their elevated proliferative potential, targeted anticancer
few articles report their loss in cancer patients.160 The reasons underlying agents act on cancer cell–specific defects and therefore are generally
this apparent discrepancy have not yet been clarified, but perhaps are associated with a reduced incidence and severity of side effects.184
linked to the fact that whereas BAX and BAK1 operate at the con- These agents include compounds that interrupt oncogene or nonon-
vergence of multiple lethal signaling pathways, single BH3-only proteins cogene addiction as well as strategies that attempt to reconstitute the
are involved in highly specific signaling cascades and perhaps are rela- lost function of tumor suppressors. In most cases, these approaches
tively redundant among themselves. lead to the apoptotic or necrotic demise of cancer cells, although in
82 Part I: Science and Clinical Oncology

The list of successful approaches that target deregulated cell death


signaling in cancer cells is continuously growing and encompasses,
among others, the proteosomal inhibitor bortezomib, which blocks
NF-κB activation and is approved by the US Food and Drug Admin-
istration (FDA) for the treatment of multiple myeloma and mantle
cell lymphoma189; so-called BH3 mimetics, that is, small molecules
that inhibit antiapoptotic members of the Bcl-2 protein family, such
as venetoclax (an orally available agent licensed by the FDA for the
treatment of chronic lymphocytic leukemia)190; rapamycin, an MTOR
inhibitor that is approved by the FDA for transplant rejection and is
now being evaluated in combination therapies against multiple
neoplasms191; and p53-reconstituting strategies, aimed at either
reintroducing wild-type p53 in p53-deficient cancer cells (by gene
therapy) or at blocking MDM2 in p53-proficient cells (by pharma-
cologic inhibitors).192,193 Of note, a recombinant adenovirus engineered
to express wild-type p53 (known as gendicine) is the first gene therapy
product approved for clinical use in humans.194 In addition to targeted
approaches, such as the aforementioned, promising results have been
obtained with epigenetic regulators such as suberoylanilide hydroxamic
acid (SAHA, also known as vorinostat), a histone deacetylase inhibitor
that is currently approved by the FDA for the treatment of cutaneous
T-cell lymphoma,195 and azacitidine or decitabine, methyltransferase
inhibitors that are currently used for the therapy of myelodysplastic
syndromes.196

WHAT THE FUTURE HOLDS


Finely manipulating cell death mechanisms in cancer, a strategy that
20 years ago appeared as a distant and hardly reachable goal, now
Figure 5.7  •  The p53 system. In physiologic settings, p53 levels are constitutes a reality with crucial clinical implications. Future investiga-
maintained under control by MDM2, an E3 ubiquitin ligase that, on polyu- tions will have to provide further insights into the molecular cascades
biquitination, targets it to proteosomal destruction. In response to multiple that underlie cell death in both physiologic (homeostatic cell death)
stress stimuli (e.g., DNA damage and oncogene activation), p53 is subjected and pathologic (in response to oncogenic stress) scenarios, and will
to posttranslational modifications that reduce its affinity for MDM2. Alter- have to unravel the mechanisms through which these are disabled
natively, stressful conditions result in the upregulation of MDM2 inhibitors during oncogenesis and tumor progression. This will be instrumental
such as p14ARF. In both cases, the cytoplasmic levels of p53 rise, allowing p53 not only for the discovery of novel drug targets and hence for the
to assemble into tetramers that enter the nucleus and regulate a plethora of development of new anticancer agents, but also for the precise identifica-
transcriptional responses. Furthermore, a pool of monoubiquitinated p53 tion of patients who may respond to particular therapeutic regimens.
persists into the cytoplasm, where it can facilitate apoptosis by directly activating
the proapoptotic protein BAX and/or by inhibiting antiapoptotic members Furthermore, strategies for the therapeutic induction of nonapoptotic
of the Bcl-2 family, or it can promote mitochondrial permeability transition RCD modes, including necroptosis and MPT-driven necrosis, or
(MPT)-driven regulating necrosis by interacting with CYPD upon entering oncosuppressive signaling networks that operate in multiple ways,
mitochondria. Ac, Acetyl; Me, methyl; Ne, Nedd8; P, phosphate; Su, SUMO; such as mitotic catastrophe,179 should be elaborated. These approaches
Ub, ubiquitin. will be particularly relevant for the treatment of chemoresistant and
radioresistant cancers, which nearly always exhibit alterations in the
molecular machinery that initiates or executes apoptotic cell death.
Finally, it will be indispensable to understand how distinct cell death
some cases the therapeutic benefit results from the activation of cellular subroutines cross talk (i.e., to which extent they can substitute for
senescence (an irreversible arrest in cell cycle progression).185 Cell each other in scenarios in which one is specifically disabled by
death mechanisms have also attracted attention for the development tumorigenic alterations) and to what extent the immune system
of strategies for chemosensitization and radiosensitization. In this case, contributes to the therapeutic efficacy of currently used anticancer
restoring the proficiency of cancer cells to undergo stress-induced cell regimens. In this context, the induction of immunogenic cell death,
death is not therapeutic per se but restores the tumor sensitivity to which converts dying tumor cells into a vaccine that is capable of
conventional chemotherapeutics, which is often lost during tumor eliciting a tumor-specific immune response,197–199 may constitute a
progression.186 Of note, the success of targeted anticancer agents appears particularly interesting therapeutic goal.
to depend, at least in part, on the activation of immune effector
mechanisms,187 which has been demonstrated in multiple settings, in The complete reference list is available online at
vitro and in vivo, including models of pure oncogene addiction.188 ExpertConsult.com.
Pathophysiology of Cancer Cell Death  •  CHAPTER 5 83

KEY REFERENCES
2. Lockshin RA, Williams CM. Programmed cell death 51. Luo X, Budihardjo I, Zou H, Slaughter C, Wang 111. Berry DL, Baehrecke EH. Growth arrest and
II. Endocrine potentiation of the breakdown of the X. Bid, a Bcl2 interacting protein, mediates cyto- autophagy are required for salivary gland cell degra-
intersegmental muscles of silkmoths. J Insect Physiol. chrome c release from mitochondria in response dation in Drosophila. Cell. 2007;131(6):1137–1148.
1964;10:643–649. to activation of cell surface death receptors. Cell. 117. Luo J, Solimini NL, Elledge SJ. Principles of cancer
3. Kerr JF, Wyllie AH, Currie AR. Apoptosis: a basic 1998;94(4):481–490. therapy: oncogene and non-oncogene addiction. Cell.
biological phenomenon with wide-ranging implica- 53. Vercammen D, Brouckaert G, Denecker G, et al. 2009;136(5):823–837.
tions in tissue kinetics. Br J Cancer. 1972;26(4): Dual signaling of the Fas receptor: initiation of both 146. Galluzzi L, Pietrocola F, Bravo-San Pedro JM, et al.
239–257. apoptotic and necrotic cell death pathways. J Exp Autophagy in malignant transformation and cancer
4. Galluzzi L, Vitale I, Abrams JM, et al. Molecular Med. 1998;188(5):919–930. progression. EMBO J. 2015;34(7):856–880.
definitions of cell death subroutines: recommenda- 54. Schulze-Osthoff K, Krammer PH, Droge W. Diver- 161. Lane DP, Crawford LV. T antigen is bound to a
tions of the Nomenclature Committee on Cell Death gent signalling via APO-1/Fas and the TNF receptor, host protein in SV40-transformed cells. Nature.
2012. Cell Death Differ. 2012;19(1):107–120. two homologous molecules involved in physiological 1979;278(5701):261–263.
14. Tsujimoto Y, Finger LR, Yunis J, Nowell PC, Croce cell death. EMBO J. 1994;13(19):4587–4596. 162. Linzer DI, Levine AJ. Characterization of a 54K
CM. Cloning of the chromosome breakpoint of 57. Linkermann A, Brasen JH, Darding M, et al. Two dalton cellular SV40 tumor antigen present in
neoplastic B cells with the t(14;18) chromosome independent pathways of regulated necrosis mediate SV40-transformed cells and uninfected embryonal
translocation. Science. 1984;226(4678):1097–1099. ischemia-reperfusion injury. Proc Natl Acad Sci USA. carcinoma cells. Cell. 1979;17(1):43–52.
16. Hanahan D, Weinberg RA. Hallmarks of cancer: 2013;110(29):12024–12029. 163. Kress M, May E, Cassingena R, May P. Simian virus
the next generation. Cell. 2011;144(5):646–674. 72. Oberst A, Dillon CP, Weinlich R, et  al. 40-transformed cells express new species of proteins
18. Galluzzi L, Bravo-San Pedro JM, Vitale I, et al. Catalytic activity of the caspase-8-FLIP(L) complex precipitable by anti-simian virus 40 tumor serum.
Essential versus accessory aspects of cell death: inhibits RIPK3-dependent necrosis. Nature. J Virol. 1979;31(2):472–483.
recommendations of the NCCD 2015. Cell Death 2011;471(7338):363–367. 172. Caelles C, Helmberg A, Karin M. p53-dependent
Differ. 2015;22(1):58–73. 74. Dillon CP, Weinlich R, Rodriguez DA, et al. RIPK1 apoptosis in the absence of transcriptional activation of
19. Tait SW, Green DR. Mitochondria and cell death: blocks early postnatal lethality mediated by caspase-8 p53-target genes. Nature. 1994;370(6486):220–223.
outer membrane permeabilization and beyond. Nat and RIPK3. Cell. 2014;157(5):1189–1202. 175. Mihara M, Erster S, Zaika A, et al. p53 has a direct
Rev Mol Cell Biol. 2010;11(9):621–632. 83. Baines CP, Kaiser RA, Purcell NH, et al. Loss of apoptogenic role at the mitochondria. Mol Cell.
21. Wallach D, Kang TB, Dillon CP, Green DR. cyclophilin D reveals a critical role for mitochon- 2003;11(3):577–590.
Programmed necrosis in inflammation: toward drial permeability transition in cell death. Nature. 176. Vaseva AV, Marchenko ND, Ji K, Tsirka SE,
identification of the effector molecules. Science. 2016; 2005;434(7033):658–662. Holzmann S, Moll UM. p53 opens the mitochon-
352(6281):aaf2154. 87. Dixon SJ, Lemberg KM, Lamprecht MR, et al. drial permeability transition pore to trigger necrosis.
22. Conrad M, Angeli JP, Vandenabeele P, Stockwell Ferroptosis: an iron-dependent form of nonapoptotic Cell. 2012;149(7):1536–1548.
BR. Regulated necrosis: disease relevance and cell death. Cell. 2012;149(5):1060–1072. 182. Matoba S, Kang JG, Patino WD, et al. p53
therapeutic opportunities. Nat Rev Drug Discov. 93. Linkermann A, Skouta R, Himmerkus N, et al. regulates mitochondrial respiration. Science. 2006;
2016;15(5):348–366. Synchronized renal tubular cell death involves fer- 312(5780):1650–1653.
23. Vanden Berghe T, Linkermann A, Jouan-Lanhouet S, roptosis. Proc Natl Acad Sci USA. 2014;111(47): 187. Galluzzi L, Buque A, Kepp O, Zitvogel L, Kroemer
Walczak H, Vandenabeele P. Regulated necrosis: the 16836–16841. G. Immunological effects of conventional chemo-
expanding network of non-apoptotic cell death path- 100. Kayagaki N, Stowe IB, Lee BL, et al. Caspase-11 therapy and targeted anticancer agents. Cancer Cell.
ways. Nat Rev Mol Cell Biol. 2014;15(2):135–147. cleaves gasdermin D for non-canonical inflamma- 2015;28(6):690–714.
40. Li P, Nijhawan D, Budihardjo I, et al. Cytochrome c some signalling. Nature. 2015;526(7575):666– 197. Kroemer G, Galluzzi L, Kepp O, Zitvogel L.
and dATP-dependent formation of Apaf-1/caspase-9 671. Immunogenic cell death in cancer therapy. Annu
complex initiates an apoptotic protease cascade. Cell. 101. Shi J, Zhao Y, Wang K, et al. Cleavage of GSDMD Rev Immunol. 2013;31:51–72.
1997;91(4):479–489. by inflammatory caspases determines pyroptotic cell 198. Linkermann A, Stockwell BR, Krautwald S, Anders
44. Pospisilik JA, Knauf C, Joza N, et al. Targeted death. Nature. 2015;526(7575):660–665. HJ. Regulated cell death and inflammation: an
deletion of AIF decreases mitochondrial oxidative 110. Boya P, Gonzalez-Polo RA, Casares N, et al. Inhibi- auto-amplification loop causes organ failure. Nat
phosphorylation and protects from obesity and tion of macroautophagy triggers apoptosis. Mol Cell Rev Immunol. 2014;14(11):759–767.
diabetes. Cell. 2007;131(3):476–491. Biol. 2005;25(3):1025–1040.
Pathophysiology of Cancer Cell Death  •  CHAPTER 5 83.e1
83.e1

REFERENCES
1. Clarke PG, Clarke S. Nineteenth century research 24. Green DR, Galluzzi L, Kroemer G. Cell 47. Willis SN, Adams JM. Life in the balance: how
on naturally occurring cell death and related phe- biology. Metabolic control of cell death. Science. BH3-only proteins induce apoptosis. Curr Opin
nomena. Anat Embryol (Berl). 1996;193(2):81–99. 2014;345(6203):1250256. Cell Biol. 2005;17(6):617–625.
2. Lockshin RA, Williams CM. Programmed cell death 25. Galluzzi L, Bravo-San Pedro JM, Kroemer G. 48. Barnhart BC, Alappat EC, Peter ME. The CD95
II. Endocrine potentiation of the breakdown of the Organelle-specific initiation of cell death. Nat Cell type I/type II model. Semin Immunol. 2003;15(3):
intersegmental muscles of silkmoths. J Insect Physiol. Biol. 2014;16(8):728–736. 185–193.
1964;10:643–649. 26. Kroemer G, Levine B. Autophagic cell death: 49. Li H, Zhu H, Xu CJ, Yuan J. Cleavage of BID by
3. Kerr JF, Wyllie AH, Currie AR. Apoptosis: a basic bio- the story of a misnomer. Nat Rev Mol Cell Biol. caspase 8 mediates the mitochondrial damage in the
logical phenomenon with wide-ranging implications 2008;9(12):1004–1010. Fas pathway of apoptosis. Cell. 1998;94(4):491–501.
in tissue kinetics. Br J Cancer. 1972;26(4):239–257. 27. Galluzzi L, Pietrocola F, Levine B, Kroemer G. 50. Jost PJ, Grabow S, Gray D, et al. XIAP discriminates
4. Galluzzi L, Vitale I, Abrams JM, et al. Molecular Metabolic control of autophagy. Cell. 2014;159(6): between type I and type II FAS-induced apoptosis.
definitions of cell death subroutines: recommenda- 1263–1276. Nature. 2009;460(7258):1035–1039.
tions of the Nomenclature Committee on Cell Death 28. Sica V, Galluzzi L, Bravo-San Pedro JM, Izzo V, 51. Luo X, Budihardjo I, Zou H, Slaughter C, Wang
2012. Cell Death Differ. 2012;19(1):107–120. Maiuri MC, Kroemer G. Organelle-specific initiation X. Bid, a Bcl2 interacting protein, mediates cyto-
5. Kroemer G, Galluzzi L, Vandenabeele P, et al. of autophagy. Mol Cell. 2015;59(4):522–539. chrome c release from mitochondria in response
Classification of cell death: recommendations of 29. Galluzzi L, Maiuri MC, Vitale I, et al. Cell to activation of cell surface death receptors. Cell.
the Nomenclature Committee on Cell Death 2009. death modalities: classification and pathophysi- 1998;94(4):481–490.
Cell Death Differ. 2009;16(1):3–11. ological implications. Cell Death Differ. 2007;14(7): 52. Vercammen D, Beyaert R, Denecker G, et al.
6. Vandenabeele P, Galluzzi L, Vanden Berghe T, 1237–1243. Inhibition of caspases increases the sensitivity of
Kroemer G. Molecular mechanisms of necroptosis: 30. Sakahira H, Enari M, Nagata S. Cleavage of CAD L929 cells to necrosis mediated by tumor necrosis
an ordered cellular explosion. Nat Rev Mol Cell Biol. inhibitor in CAD activation and DNA degradation factor. J Exp Med. 1998;187(9):1477–1485.
2010;11(10):700–714. during apoptosis. Nature. 1998;391(6662):96–99. 53. Vercammen D, Brouckaert G, Denecker G, et al.
7. Galluzzi L, Kepp O, Krautwald S, Kroemer G, 31. Wajant H. The Fas signaling pathway: more than Dual signaling of the Fas receptor: initiation of both
Linkermann A. Molecular mechanisms of regulated a paradigm. Science. 2002;296(5573):1635–1636. apoptotic and necrotic cell death pathways. J Exp
necrosis. Semin Cell Dev Biol. 2014;35:24–32. 32. Mehlen P, Bredesen DE. Dependence receptors: Med. 1998;188(5):919–930.
8. Galluzzi L, Vanden Berghe T, Vanlangenakker N, from basic research to drug development. Sci Signal. 54. Schulze-Osthoff K, Krammer PH, Droge W. Diver-
et al. Programmed necrosis from molecules to health 2011;4(157):mr2. gent signalling via APO-1/Fas and the TNF receptor,
and disease. Int Rev Cell Mol Biol. 2011;289:1–35. 33. Siegel RM, Frederiksen JK, Zacharias DA, et al. two homologous molecules involved in physiological
9. Bender CE, Fitzgerald P, Tait SW, et al. Mitochon- Fas preassociation required for apoptosis signaling cell death. EMBO J. 1994;13(19):4587–4596.
drial pathway of apoptosis is ancestral in metazoans. and dominant inhibition by pathogenic mutations. 55. Barkla DH, Gibson PR. The fate of epithelial
Proc Natl Acad Sci USA. 2012;109(13):4904–4909. Science. 2000;288(5475):2354–2357. cells in the human large intestine. Pathology.
10. Galluzzi L, Kepp O, Kroemer G. Mitochondrial 34. Schulze-Osthoff K, Ferrari D, Los M, Wesselborg S, 1999;31(3):230–238.
regulation of cell death: a phylogenetically conserved Peter ME. Apoptosis signaling by death receptors. 56. Roach HI, Clarke NM. Physiological cell death of
control. Microb Cell. 2016;3(3):101–108. Eur J Biochem. 1998;254(3):439–459. chondrocytes in vivo is not confined to apoptosis.
11. Carmona-Gutierrez D, Ruckenstuhl C, Bauer MA, 35. Cohen GM. Caspases: the executioners of apoptosis. New observations on the mammalian growth plate.
Eisenberg T, Buttner S, Madeo F. Cell death in yeast: Biochem J. 1997;326(Pt 1):1–16. J Bone Joint Surg Br. 2000;82(4):601–613.
growing applications of a dying buddy. Cell Death 36. Mille F, Thibert C, Fombonne J, et al. The 57. Linkermann A, Brasen JH, Darding M, et al. Two
Differ. 2010;17(5):733–734. Patched dependence receptor triggers apoptosis independent pathways of regulated necrosis mediate
12. Meier P, Finch A, Evan G. Apoptosis in development. through a DRAL-caspase-9 complex. Nat Cell Biol. ischemia-reperfusion injury. Proc Natl Acad Sci USA.
Nature. 2000;407(6805):796–801. 2009;11(6):739–746. 2013;110(29):12024–12029.
13. Green DR, Kroemer G. The pathophysiology of 37. Kroemer G, Galluzzi L, Brenner C. Mitochondrial 58. Cho YS, Challa S, Moquin D, et al. Phosphorylation-
mitochondrial cell death. Science. 2004;305(5684): membrane permeabilization in cell death. Physiol driven assembly of the RIP1-RIP3 complex regulates
626–629. Rev. 2007;87(1):99–163. programmed necrosis and virus-induced inflamma-
14. Tsujimoto Y, Finger LR, Yunis J, Nowell PC, Croce 38. Chai J, Du C, Wu JW, Kyin S, Wang X, Shi Y. Struc- tion. Cell. 2009;137(6):1112–1123.
CM. Cloning of the chromosome breakpoint of tural and biochemical basis of apoptotic activation 59. He S, Wang L, Miao L, et al. Receptor interacting
neoplastic B cells with the t(14;18) chromosome by Smac/DIABLO. Nature. 2000;406(6798):855– protein kinase-3 determines cellular necrotic response
translocation. Science. 1984;226(4678):1097–1099. 862. to TNF-alpha. Cell. 2009;137(6):1100–1111.
15. Vaux DL, Cory S, Adams JM. Bcl-2 gene pro- 39. Hegde R, Srinivasula SM, Zhang Z, et  al. 60. Zhang DW, Shao J, Lin J, et al. RIP3, an energy
motes haemopoietic cell survival and cooperates Identification of Omi/HtrA2 as a mitochondrial metabolism regulator that switches TNF-induced
with c-myc to immortalize pre-B cells. Nature. apoptotic serine protease that disrupts inhibitor of cell death from apoptosis to necrosis. Science. 2009;
1988;335(6189):440–442. apoptosis protein-caspase interaction. J Biol Chem. 325(5938):332–336.
16. Hanahan D, Weinberg RA. Hallmarks of cancer: 2002;277(1):432–438. 61. Sun L, Wang H, Wang Z, et al. Mixed lineage kinase
the next generation. Cell. 2011;144(5):646–674. 40. Li P, Nijhawan D, Budihardjo I, et al. Cytochrome c domain-like protein mediates necrosis signaling
17. Igney FH, Krammer PH. Death and anti-death: tumour and dATP-dependent formation of Apaf-1/caspase-9 downstream of RIP3 kinase. Cell. 2012;148(1–2):
resistance to apoptosis. Nat Rev Cancer. 2002;2(4): complex initiates an apoptotic protease cascade. Cell. 213–227.
277–288. 1997;91(4):479–489. 62. Wang Z, Jiang H, Chen S, Du F, Wang X. The
18. Galluzzi L, Bravo-San Pedro JM, Vitale I, et al. 41. Galluzzi L, Joza N, Tasdemir E, et al. No death mitochondrial phosphatase PGAM5 functions at
Essential versus accessory aspects of cell death: without life: vital functions of apoptotic effectors. the convergence point of multiple necrotic death
recommendations of the NCCD 2015. Cell Death Cell Death Differ. 2008;15(7):1113–1123. pathways. Cell. 2012;148(1–2):228–243.
Differ. 2015;22(1):58–73. 42. Li LY, Luo X, Wang X. Endonuclease G is an 63. Tait SW, Oberst A, Quarato G, et al. Widespread
19. Tait SW, Green DR. Mitochondria and cell death: apoptotic DNase when released from mitochondria. mitochondrial depletion via mitophagy does not
outer membrane permeabilization and beyond. Nat Nature. 2001;412(6842):95–99. compromise necroptosis. Cell Rep. 2013;5(4):878–
Rev Mol Cell Biol. 2010;11(9):621–632. 43. Joza N, Susin SA, Daugas E, et al. Essential role 885.
20. Tait SW, Green DR. Mitochondrial regulation of of the mitochondrial apoptosis-inducing factor in 64. Moriwaki K, Farias Luz N, Balaji S, et al. The mito-
cell death. Cold Spring Harb Perspect Biol. 2013;5(9). programmed cell death. Nature. 2001;410(6828): chondrial phosphatase PGAM5 is dispensable for
21. Wallach D, Kang TB, Dillon CP, Green DR. 549–554. necroptosis but promotes inflammasome activation
Programmed necrosis in inflammation: toward 44. Pospisilik JA, Knauf C, Joza N, et al. Targeted in macrophages. J Immunol. 2016;196(1):407–415.
identification of the effector molecules. Science. deletion of AIF decreases mitochondrial oxidative 65. Xia B, Fang S, Chen X, et al. MLKL forms cation
2016;352(6281):aaf2154. phosphorylation and protects from obesity and channels. Cell Res. 2016;26(5):517–528.
22. Conrad M, Angeli JP, Vandenabeele P, Stockwell BR. diabetes. Cell. 2007;131(3):476–491. 66. Quarato G, Guy CS, Grace CR, et al. Sequential
Regulated necrosis: disease relevance and therapeutic 45. David KK, Sasaki M, Yu SW, Dawson TM, Dawson engagement of distinct MLKL phosphatidylino-
opportunities. Nat Rev Drug Discov. 2016;15(5): VL. EndoG is dispensable in embryogenesis and sitol-binding sites executes necroptosis. Mol Cell.
348–366. apoptosis. Cell Death Differ. 2006;13(7):1147– 2016;61(4):589–601.
23. Vanden Berghe T, Linkermann A, Jouan-Lanhouet S, 1155. 67. Cai Z, Jitkaew S, Zhao J, et al. Plasma membrane
Walczak H, Vandenabeele P. Regulated necrosis: the 46. Youle RJ, Strasser A. The BCL-2 protein family: translocation of trimerized MLKL protein is
expanding network of non-apoptotic cell death path- opposing activities that mediate cell death. Nat Rev required for TNF-induced necroptosis. Nat Cell
ways. Nat Rev Mol Cell Biol. 2014;15(2):135–147. Mol Cell Biol. 2008;9(1):47–59. Biol. 2014;16(1):55–65.
83.e2 Part I: Science and Clinical Oncology

68. Chen X, Li W, Ren J, et al. Translocation of mixed 88. Dixon SJ, Patel DN, Welsch M, et al. Pharmacologi- 112. Denton D, Shravage B, Simin R, et al. Autophagy,
lineage kinase domain-like protein to plasma mem- cal inhibition of cystine-glutamate exchange induces not apoptosis, is essential for midgut cell death in
brane leads to necrotic cell death. Cell Res. 2014;24(1): endoplasmic reticulum stress and ferroptosis. Elife. Drosophila. Curr Biol. 2009;19(20):1741–1746.
105–121. 2014;3:e02523. 113. Grander D, Kharaziha P, Laane E, Pokrovskaja K,
69. Hildebrand JM, Tanzer MC, Lucet IS, et al. 89. Friedmann Angeli JP, Schneider M, Proneth B, Panaretakis T. Autophagy as the main means of
Activation of the pseudokinase MLKL unleashes et al. Inactivation of the ferroptosis regulator Gpx4 cytotoxicity by glucocorticoids in hematological
the four-helix bundle domain to induce membrane triggers acute renal failure in mice. Nat Cell Biol. malignancies. Autophagy. 2009;5(8):1198–1200.
localization and necroptotic cell death. Proc Natl 2014;16(12):1180–1191. 114. Levine B, Kroemer G. Autophagy in the pathogenesis
Acad Sci USA. 2014;111(42):15072–15077. 90. Yang WS, SriRamaratnam R, Welsch ME, et al. of disease. Cell. 2008;132(1):27–42.
70. Dondelinger Y, Declercq W, Montessuit S, et al. Regulation of ferroptotic cancer cell death by GPX4. 115. Levine B, Kroemer G. Autophagy in aging, disease
MLKL compromises plasma membrane integrity Cell. 2014;156(1–2):317–331. and death: the true identity of a cell death impostor.
by binding to phosphatidylinositol phosphates. Cell 91. Skouta R, Dixon SJ, Wang J, et al. Ferrostatins inhibit Cell Death Differ. 2009;16(1):1–2.
Rep. 2014;7(4):971–981. oxidative lipid damage and cell death in diverse 116. Serrano M, Lin AW, McCurrach ME, Beach D,
71. Wang H, Sun L, Su L, et al. Mixed lineage kinase disease models. J Am Chem Soc. 2014;136(12): Lowe SW. Oncogenic ras provokes premature cell
domain-like protein MLKL causes necrotic mem- 4551–4556. senescence associated with accumulation of p53 and
brane disruption upon phosphorylation by RIP3. 92. Linkermann A. Nonapoptotic cell death in acute p16INK4a. Cell. 1997;88(5):593–602.
Mol Cell. 2014;54(1):133–146. kidney injury and transplantation. Kidney Int. 117. Luo J, Solimini NL, Elledge SJ. Principles of cancer
72. Oberst A, Dillon CP, Weinlich R, et al. Catalytic 2016;89(1):46–57. therapy: oncogene and non-oncogene addiction. Cell.
activity of the caspase-8-FLIP(L) complex inhibits 93. Linkermann A, Skouta R, Himmerkus N, et al. 2009;136(5):823–837.
RIPK3-dependent necrosis. Nature. 2011;471(7338): Synchronized renal tubular cell death involves fer- 118. Kroemer G, Senovilla L, Galluzzi L, Andre F, Zitvogel
363–367. roptosis. Proc Natl Acad Sci USA. 2014;111(47): L. Natural and therapy-induced immunosurveillance
73. Orozco S, Yatim N, Werner MR, et al. RIPK1 16836–16841. in breast cancer. Nat Med. 2015;21(10):1128–1138.
both positively and negatively regulates RIPK3 94. Kepp O, Galluzzi L, Zitvogel L, Kroemer G. 119. Heisterkamp N, Groffen J, Stephenson JR,
oligomerization and necroptosis. Cell Death Differ. Pyroptosis—a cell death modality of its kind? Eur et al. Chromosomal localization of human cel-
2014;21(10):1511–1521. J Immunol. 2010;40(3):627–630. lular homologues of two viral oncogenes. Nature.
74. Dillon CP, Weinlich R, Rodriguez DA, et al. RIPK1 95. Zitvogel L, Kepp O, Galluzzi L, Kroemer G. Inflam- 1982;299(5885):747–749.
blocks early postnatal lethality mediated by caspase-8 masomes in carcinogenesis and anticancer immune 120. Meyer N, Penn LZ. Reflecting on 25 years with
and RIPK3. Cell. 2014;157(5):1189–1202. responses. Nat Immunol. 2012;13(4):343–351. MYC. Nat Rev Cancer. 2008;8(12):976–990.
75. Zong WX, Ditsworth D, Bauer DE, Wang ZQ, 96. Galluzzi L, Lopez-Soto A, Kumar S, Kroemer G. 121. Schubbert S, Shannon K, Bollag G. Hyperactive
Thompson CB. Alkylating DNA damage stimulates Caspases connect cell-death signaling to organismal Ras in developmental disorders and cancer. Nat Rev
a regulated form of necrotic cell death. Genes Dev. homeostasis. Immunity. 2016;44(2):221–231. Cancer. 2007;7(4):295–308.
2004;18(11):1272–1282. 97. Sborgi L, Ruhl S, Mulvihill E, et al. GSDMD 122. Rong Y, Distelhorst CW. Bcl-2 protein family
76. Nogusa S, Thapa RJ, Dillon CP, et al. RIPK3 membrane pore formation constitutes the mechanism members: versatile regulators of calcium signaling in
activates parallel pathways of MLKL-driven of pyroptotic cell death. EMBO J. 2016. cell survival and apoptosis. Annu Rev Physiol. 2008;70:
necroptosis and FADD-mediated apoptosis to 98. Liu X, Zhang Z, Ruan J, et al. Inflammasome- 73–91.
protect against influenza A virus. Cell Host Microbe. activated gasdermin D causes pyroptosis by forming 123. Vander Heiden MG, Chandel NS, Schumacker PT,
2016;20(1):13–24. membrane pores. Nature. 2016;535(7610):153–158. Thompson CB. Bcl-xL prevents cell death following
77. Bonora M, Wieckowski MR, Chinopoulos C, et al. 99. Aglietti RA, Estevez A, Gupta A, et al. GsdmD growth factor withdrawal by facilitating mitochon-
Molecular mechanisms of cell death: central implica- p30 elicited by caspase-11 during pyroptosis forms drial ATP/ADP exchange. Mol Cell. 1999;3(2):
tion of ATP synthase in mitochondrial permeability pores in membranes. Proc Natl Acad Sci USA. 159–167.
transition. Oncogene. 2015;34(12):1475–1486. 2016;113(28):7858–7863. 124. Rassidakis GZ, Jones D, Lai R, et al. BCL-2 family
78. Izzo V, Bravo-San Pedro JM, Sica V, Kroemer G, 100. Kayagaki N, Stowe IB, Lee BL, et al. Caspase-11 proteins in peripheral T-cell lymphomas: correlation
Galluzzi L. Mitochondrial permeability transition: cleaves gasdermin D for non-canonical inflamma- with tumour apoptosis and proliferation. J Pathol.
new findings and persisting uncertainties. Trends some signalling. Nature. 2015;526(7575):666–671. 2003;200(2):240–248.
Cell Biol. 2016. 101. Shi J, Zhao Y, Wang K, et al. Cleavage of GSDMD 125. Linden M, Kirchhof N, Carlson C, Van Ness B.
79. Brenner C, Grimm S. The permeability transition pore by inflammatory caspases determines pyroptotic cell Targeted overexpression of Bcl-XL in B-lymphoid
complex in cancer cell death. Oncogene. 2006;25(34): death. Nature. 2015;526(7575):660–665. cells results in lymphoproliferative disease and
4744–4756. 102. Lim Y, Kumar S. A single cut to pyroptosis. plasma cell malignancies. Blood. 2004;103(7):2779–
80. Baines CP, Kaiser RA, Sheiko T, Craigen WJ, Oncotarget. 2015;6(35):36926–36927. 2786.
Molkentin JD. Voltage-dependent anion channels 103. Gibson BA, Kraus WL. New insights into the 126. Brunelle JK, Ryan J, Yecies D, Opferman JT, Letai
are dispensable for mitochondrial-dependent cell molecular and cellular functions of poly(ADP-ribose) A. MCL-1-dependent leukemia cells are more
death. Nat Cell Biol. 2007;9(5):550–555. and PARPs. Nat Rev Mol Cell Biol. 2012;13(7):411– sensitive to chemotherapy than BCL-2-dependent
81. Galluzzi L, Kroemer G. Mitochondrial apoptosis 424. counterparts. J Cell Biol. 2009;187(3):429–442.
without VDAC. Nat Cell Biol. 2007;9(5):487– 104. Andrabi SA, Umanah GK, Chang C, et al. Poly(ADP- 127. Strasser A, Harris AW, Cory S. E mu-bcl-2 transgene
489. ribose) polymerase-dependent energy depletion facilitates spontaneous transformation of early pre-B
82. Kokoszka JE, Waymire KG, Levy SE, et al. The occurs through inhibition of glycolysis. Proc Natl and immunoglobulin-secreting cells but not T cells.
ADP/ATP translocator is not essential for the Acad Sci USA. 2014;111(28):10209–10214. Oncogene. 1993;8(1):1–9.
mitochondrial permeability transition pore. Nature. 105. Wang Y, Kim NS, Haince JF, et al. Poly(ADP-ribose) 128. Campbell KJ, Bath ML, Turner ML, et al. Elevated
2004;427(6973):461–465. (PAR) binding to apoptosis-inducing factor is Mcl-1 perturbs lymphopoiesis, promotes transforma-
83. Baines CP, Kaiser RA, Purcell NH, et al. Loss of critical for PAR polymerase-1-dependent cell death tion of hematopoietic stem/progenitor cells, and
cyclophilin D reveals a critical role for mitochon- (parthanatos). Sci Signal. 2011;4(167):ra20. enhances drug resistance. Blood. 2010;116(17):
drial permeability transition in cell death. Nature. 106. Galluzzi L, Bravo-San Pedro JM, Blomgren K, 3197–3207.
2005;434(7033):658–662. Kroemer G. Autophagy in acute brain injury. Nat 129. Galluzzi L, Morselli E, Kepp O, et al. Mito-
84. Schinzel AC, Takeuchi O, Huang Z, et al. Rev Neurosci. 2016;17(8):467–484. chondrial gateways to cancer. Mol Aspects Med.
Cyclophilin D is a component of mitochondrial 107. Green DR, Galluzzi L, Kroemer G. Mitochondria and 2010;31(1):1–20.
permeability transition and mediates neuronal cell the autophagy-inflammation-cell death axis in organ- 130. Perkins ND. Integrating cell-signalling pathways
death after focal cerebral ischemia. Proc Natl Acad ismal aging. Science. 2011;333(6046):1109–1112. with NF-kappaB and IKK function. Nat Rev Mol
Sci USA. 2005;102(34):12005–12010. 108. Mizushima N, Levine B, Cuervo AM, Klionsky Cell Biol. 2007;8(1):49–62.
85. Bonora M, Bononi A, De Marchi E, et al. Role of the DJ. Autophagy fights disease through cellular self- 131. Pahl HL. Activators and target genes of Rel/NF-
c subunit of the FO ATP synthase in mitochondrial digestion. Nature. 2008;451(7182):1069–1075. kappaB transcription factors. Oncogene. 1999;18(49):
permeability transition. Cell Cycle. 2013;12(4): 109. Galluzzi L, Bravo-San Pedro JM, Kepp O, Kroemer 6853–6866.
674–683. G. Regulated cell death and adaptive stress responses. 132. Chen IS, Temin HM. Substitution of 5′ helper virus
86. Yang WS, Kim KJ, Gaschler MM, Patel M, Cell Mol Life Sci. 2016;73(11–12):2405–2410. sequences into non-rel portion of reticuloendothe-
Shchepinov MS, Stockwell BR. Peroxidation of 110. Boya P, Gonzalez-Polo RA, Casares N, et al. Inhibi- liosis virus strain T suppresses transformation of
polyunsaturated fatty acids by lipoxygenases drives tion of macroautophagy triggers apoptosis. Mol Cell chicken spleen cells. Cell. 1982;31(1):111–120.
ferroptosis. Proc Natl Acad Sci USA. 2016. Biol. 2005;25(3):1025–1040. 133. Carrasco D, Rizzo CA, Dorfman K, Bravo R.
87. Dixon SJ, Lemberg KM, Lamprecht MR, et al. 111. Berry DL, Baehrecke EH. Growth arrest and The v-rel oncogene promotes malignant T-cell
Ferroptosis: an iron-dependent form of nonapoptotic autophagy are required for salivary gland cell degra- leukemia/lymphoma in transgenic mice. EMBO J.
cell death. Cell. 2012;149(5):1060–1072. dation in Drosophila. Cell. 2007;131(6):1137–1148. 1996;15(14):3640–3650.
Pathophysiology of Cancer Cell Death  •  CHAPTER 5 83.e3
83.e3

134. Chu ZL, DiDonato JA, Hawiger J, Ballard DW. The 154. Li J, Yen C, Liaw D, et al. PTEN, a putative 176. Vaseva AV, Marchenko ND, Ji K, Tsirka SE,
tax oncoprotein of human T-cell leukemia virus type protein tyrosine phosphatase gene mutated in Holzmann S, Moll UM. p53 opens the mitochon-
1 associates with and persistently activates IkappaB human brain, breast, and prostate cancer. Science. drial permeability transition pore to trigger necrosis.
kinases containing IKKalpha and IKKbeta. J Biol 1997;275(5308):1943–1947. Cell. 2012;149(7):1536–1548.
Chem. 1998;273(26):15891–15894. 155. Podsypanina K, Ellenson LH, Nemes A, et al. 177. Cheok CF, Verma CS, Baselga J, Lane DP. Trans-
135. Rayet B, Gelinas C. Aberrant rel/nfkb genes and Mutation of Pten/Mmac1 in mice causes neoplasia lating p53 into the clinic. Nat Rev Clin Oncol.
activity in human cancer. Oncogene. 1999;18(49): in multiple organ systems. Proc Natl Acad Sci USA. 2011;8(1):25–37.
6938–6947. 1999;96(4):1563–1568. 178. Donehower LA, Harvey M, Slagle BL, et al. Mice
136. Fujita T, Nolan GP, Liou HC, Scott ML, Baltimore 156. Di Cristofano A, Pesce B, Cordon-Cardo C, Pandolfi deficient for p53 are developmentally normal
D. The candidate proto-oncogene bcl-3 encodes a PP. Pten is essential for embryonic development but susceptible to spontaneous tumours. Nature.
transcriptional coactivator that activates through NF- and tumour suppression. Nat Genet. 1998;19(4): 1992;356(6366):215–221.
kappa B p50 homodimers. Genes Dev. 1993;7(7B): 348–355. 179. Vitale I, Galluzzi L, Castedo M, Kroemer G. Mitotic
1354–1363. 157. Alimonti A, Carracedo A, Clohessy JG, et al. Subtle catastrophe: a mechanism for avoiding genomic insta-
137. Michaux L, Mecucci C, Stul M, et al. BCL3 variations in Pten dose determine cancer susceptibil- bility. Nat Rev Mol Cell Biol. 2011;12(6):385–392.
rearrangement and t(14;19)(q32;q13) in lympho- ity. Nat Genet. 2010;42(5):454–458. 180. Sablina AA, Budanov AV, Ilyinskaya GV, Agapova
proliferative disorders. Genes Chromosomes Cancer. 158. Degenhardt K, Chen G, Lindsten T, White E. BAX LS, Kravchenko JE, Chumakov PM. The antioxidant
1996;15(1):38–47. and BAK mediate p53-independent suppression of function of the p53 tumor suppressor. Nat Med.
138. Finco TS, Westwick JK, Norris JL, Beg AA, Der CJ, tumorigenesis. Cancer Cell. 2002;2(3):193–203. 2005;11(12):1306–1313.
Baldwin AS Jr. Oncogenic Ha-Ras-induced signaling 159. Rampino N, Yamamoto H, Ionov Y, et al. Somatic 181. Vousden KH, Ryan KM. p53 and metabolism. Nat
activates NF-kappaB transcriptional activity, which frameshift mutations in the BAX gene in colon Rev Cancer. 2009;9(10):691–700.
is required for cellular transformation. J Biol Chem. cancers of the microsatellite mutator phenotype. 182. Matoba S, Kang JG, Patino WD, et al. p53
1997;272(39):24113–24116. Science. 1997;275(5302):967–969. regulates mitochondrial respiration. Science.
139. Reuther JY, Reuther GW, Cortez D, Pendergast 160. Sturm I, Stephan C, Gillissen B, et al. Loss of the 2006;312(5780):1650–1653.
AM, Baldwin AS Jr. A requirement for NF-kappaB tissue-specific proapoptotic BH3-only protein Nbk/ 183. Diamandis M, White NM, Yousef GM. Personalized
activation in Bcr-Abl-mediated transformation. Genes Bik is a unifying feature of renal cell carcinoma. Cell medicine: marking a new epoch in cancer patient
Dev. 1998;12(7):968–981. Death Differ. 2006;13(4):619–627. management. Mol Cancer Res. 2010;8(9):1175–1187.
140. Hanahan D, Weinberg RA. The hallmarks of cancer. 161. Lane DP, Crawford LV. T antigen is bound to a 184. Dancey JE, Chen HX. Strategies for optimizing
Cell. 2000;100(1):57–70. host protein in SV40-transformed cells. Nature. combinations of molecularly targeted anticancer
141. Manning BD, Cantley LC. AKT/PKB signaling: 1979;278(5701):261–263. agents. Nat Rev Drug Discov. 2006;5(8):649–659.
navigating downstream. Cell. 2007;129(7):1261– 162. Linzer DI, Levine AJ. Characterization of a 54K 185. Nardella C, Clohessy JG, Alimonti A, Pandolfi PP.
1274. dalton cellular SV40 tumor antigen present in Pro-senescence therapy for cancer treatment. Nat
142. Manning BD, Tee AR, Logsdon MN, Blenis J, SV40-transformed cells and uninfected embryonal Rev Cancer. 2011;11(7):503–511.
Cantley LC. Identification of the tuberous sclerosis carcinoma cells. Cell. 1979;17(1):43–52. 186. Shabbits JA, Hu Y, Mayer LD. Tumor chemosensi-
complex-2 tumor suppressor gene product tuberin 163. Kress M, May E, Cassingena R, May P. Simian virus tization strategies based on apoptosis manipulations.
as a target of the phosphoinositide 3-kinase/akt 40-transformed cells express new species of proteins Mol Cancer Ther. 2003;2(8):805–813.
pathway. Mol Cell. 2002;10(1):151–162. precipitable by anti-simian virus 40 tumor serum. 187. Galluzzi L, Buque A, Kepp O, Zitvogel L, Kroemer
143. Malstrom S, Tili E, Kappes D, Ceci JD, Tsichlis J Virol. 1979;31(2):472–483. G. Immunological effects of conventional chemo-
PN. Tumor induction by an Lck-MyrAkt transgene 164. Rotter V, Witte ON, Coffman R, Baltimore D. therapy and targeted anticancer agents. Cancer Cell.
is delayed by mechanisms controlling the size of Abelson murine leukemia virus-induced tumors elicit 2015;28(6):690–714.
the thymus. Proc Natl Acad Sci USA. 2001;98(26): antibodies against a host cell protein, P50. J Virol. 188. Rakhra K, Bachireddy P, Zabuawala T, et al.
14967–14972. 1980;36(2):547–555. CD4(+) T cells contribute to the remodeling of
144. Hollander MC, Blumenthal GM, Dennis PA. PTEN 165. Vousden KH, Prives C. Blinded by the light: the the microenvironment required for sustained tumor
loss in the continuum of common cancers, rare growing complexity of p53. Cell. 2009;137(3): regression upon oncogene inactivation. Cancer Cell.
syndromes and mouse models. Nat Rev Cancer. 413–431. 2010;18(5):485–498.
2011;11(4):289–301. 166. Li M, Brooks CL, Wu-Baer F, Chen D, Baer R, 189. Sanchez-Serrano I. Success in translational research:
145. Podsypanina K, Lee RT, Politis C, et al. An inhibitor Gu W. Mono- versus polyubiquitination: dif- lessons from the development of bortezomib. Nat
of mTOR reduces neoplasia and normalizes p70/ ferential control of p53 fate by Mdm2. Science. Rev Drug Discov. 2006;5(2):107–114.
S6 kinase activity in Pten+/− mice. Proc Natl Acad 2003;302(5652):1972–1975. 190. Roberts AW, Davids MS, Pagel JM, et al. Targeting
Sci USA. 2001;98(18):10320–10325. 167. Kruse JP, Gu W. Modes of p53 regulation. Cell. BCL2 with venetoclax in relapsed chronic lympho-
146. Galluzzi L, Pietrocola F, Bravo-San Pedro JM, et al. 2009;137(4):609–622. cytic leukemia. N Engl J Med. 2016;374(4):311–322.
Autophagy in malignant transformation and cancer 168. Riley T, Sontag E, Chen P, Levine A. Transcriptional 191. Dancey J. mTOR signaling and drug development
progression. EMBO J. 2015;34(7):856–880. control of human p53-regulated genes. Nat Rev Mol in cancer. Nat Rev Clin Oncol. 2010;7(4):209–219.
147. Harris H, Miller OJ, Klein G, Worst P, Tachibana Cell Biol. 2008;9(5):402–412. 192. Vassilev LT, Vu BT, Graves B, et al. In vivo activation
T. Suppression of malignancy by cell fusion. Nature. 169. Miyashita T, Krajewski S, Krajewska M, et al. of the p53 pathway by small-molecule antagonists
1969;223(5204):363–368. Tumor suppressor p53 is a regulator of bcl-2 and of MDM2. Science. 2004;303(5659):844–848.
148. Friend SH, Bernards R, Rogelj S, et al. A human bax gene expression in vitro and in vivo. Oncogene. 193. Brown CJ, Lain S, Verma CS, Fersht AR, Lane
DNA segment with properties of the gene that 1994;9(6):1799–1805. DP. Awakening guardian angels: drugging the p53
predisposes to retinoblastoma and osteosarcoma. 170. Moroni MC, Hickman ES, Lazzerini Denchi E, pathway. Nat Rev Cancer. 2009;9(12):862–873.
Nature. 1986;323(6089):643–646. et al. Apaf-1 is a transcriptional target for E2F and 194. Wilson JM. Gendicine: the first commercial gene
149. Baylin SB, Ohm JE. Epigenetic gene silencing in p53. Nat Cell Biol. 2001;3(6):552–558. therapy product. Hum Gene Ther. 2005;16(9):
cancer—a mechanism for early oncogenic pathway 171. Nakano K, Vousden KH. PUMA, a novel 1014–1015.
addiction? Nat Rev Cancer. 2006;6(2):107–116. proapoptotic gene, is induced by p53. Mol Cell. 195. Grant S, Easley C, Kirkpatrick P. Vorinostat. Nat
150. Esteller M, Cordon-Cardo C, Corn PG, et al. 2001;7(3):683–694. Rev Drug Discov. 2007;6(1):21–22.
p14ARF silencing by promoter hypermethylation 172. Caelles C, Helmberg A, Karin M. p53-dependent 196. Quintas-Cardama A, Santos FP, Garcia-Manero G.
mediates abnormal intracellular localization of apoptosis in the absence of transcriptional activation of Therapy with azanucleosides for myelodysplastic
MDM2. Cancer Res. 2001;61(7):2816–2821. p53-target genes. Nature. 1994;370(6486):220–223. syndromes. Nat Rev Clin Oncol. 2010;7(8):433–444.
151. Teitz T, Wei T, Valentine MB, et al. Caspase 8 173. Chipuk JE, Bouchier-Hayes L, Kuwana T, Newmeyer 197. Kroemer G, Galluzzi L, Kepp O, Zitvogel L.
is deleted or silenced preferentially in childhood DD, Green DR. PUMA couples the nuclear and Immunogenic cell death in cancer therapy. Annu
neuroblastomas with amplification of MYCN. Nat cytoplasmic proapoptotic function of p53. Science. Rev Immunol. 2013;31:51–72.
Med. 2000;6(5):529–535. 2005;309(5741):1732–1735. 198. Linkermann A, Stockwell BR, Krautwald S, Anders
152. Hansen MF, Cavenee WK. Tumor suppressors: reces- 174. Chipuk JE, Kuwana T, Bouchier-Hayes L, et al. HJ. Regulated cell death and inflammation: an
sive mutations that lead to cancer. Cell. 1988;53(2): Direct activation of Bax by p53 mediates mito- auto-amplification loop causes organ failure. Nat
173–174. chondrial membrane permeabilization and apoptosis. Rev Immunol. 2014;14(11):759–767.
153. Stambolic V, Suzuki A, de la Pompa JL, et al. Science. 2004;303(5660):1010–1014. 199. Land WG, Agostinis P, Gasser S, Garg AD,
Negative regulation of PKB/Akt-dependent cell 175. Mihara M, Erster S, Zaika A, et al. p53 has a direct Linkermann A. Transplantation and damage associ-
survival by the tumor suppressor PTEN. Cell. apoptogenic role at the mitochondria. Mol Cell. ated molecular patterns (DAMPs). Am J Transplant.
1998;95(1):29–39. 2003;11(3):577–590. 2016.
83.e4 Part I: Science and Clinical Oncology

SELF-ASSESSMENT REVIEW QUESTIONS ANSWERS


1. Which one of the following molecular alterations may be 1. (c)  BAX overexpression is expected to counteract oncogenesis,
common among lymphoma patients? similar to inactivating mutations in AKT1. Conversely, the
a. A translocation between chromosome 14 and chromosome amplification of MDM2 is relatively common among tumors,
19, resulting in the overexpression of BAX leading to the functional inactivation of the p53 system.
b. A loss-of-function point mutation in AKT1 2. (c)  Increasing the levels of mutant p53 by employing MDM2
c. An amplification in the chromosomal region 12q13-14, inhibitors is expected to be useless in the presence of
encompassing MDM2 inactivating TP53 mutations. Along similar lines, the use of
2. The detection of a point mutation in TP53 that negatively DNA-damaging agents would presumably yield poor results,
affects p53-regulated gene transactivation would prompt for: because these compounds most often function by activating
a. The administration of a hypothetical FDA-approved MDM2 p53. Conversely, gendicine would lead to the restoration of a
inhibitor functional p53 system, perhaps exerting direct anticancer effects
b. The administration of DNA damaging agents or rendering cancer cells responsive to chemotherapy.
c. The use of gendicine 3. (a)  Inhibition of BAX or caspases appears to be therapeutically
3. Which one of the following may constitute the basis for the useful in conditions featuring excessive cell death, such as
development of novel anticancer regimens? ischemia or neurodegeneration. Conversely, novel activators of
a. The screening of a combinatorial chemical library for the MLKL that would selectively trigger the necrotic demise of
identification of MLKL activators cancer cells might be very promising, because cancer cells are
b. The discovery of a new inhibitor of caspases often intrinsically resistant to the therapeutic induction of
c. The identification of a new inhibitor of BAX apoptosis.
4. Which one of the following statements is best supported by 4. (b)  Although both death receptors and dependence receptors
experimental evidence? trigger extrinsic apoptosis, not only do they respond to different
a. Death receptors and dependence receptors activate the same conditions, but also they engage distinct signaling cascades. Still,
signaling pathways. both death receptors and dependence receptors eventually lead
b. Death receptors and dependence receptors lead to CASP3 to the proteolytic activation of CASP3.
activation.
c. Death receptors and dependence receptors operate in similar
settings.
6  Cancer Immunology
Diane Tseng, Liora Schultz, Drew Pardoll, and Crystal Mackall

S UMMARY OF K EY P OI N T S
• Cancer is characterized by genetic immune system, through a process demonstrated impressive effects in
and epigenetic instability leading termed editing. Multiple cells and many cancers, including melanoma,
to both unique and sometimes molecular interactions in the tumor non–small cell lung cancer, renal
common mutations and “ectopic” microenvironment also inhibit cell carcinoma, bladder carcinoma,
overexpression of genes not antitumor immune responses, head and neck cancer, Hodgkin
normally expressed in the tissue of including regulatory T cells and lymphoma, Merkel cell carcinoma,
origin. Cancer-specific proteins myeloid-derived suppressor cells. and others.
arising as a result of genetic Tumor-induced immunosuppression • Synthetic biology can be used to
mutations appear to provide the is also mediated by signaling CTLA-4 engineer immunotherapies toward
most potent antigens visible to and PD-1, inhibitory receptors on antigens differentially expressed on
the T-cell arm of the immune T cells. cancer versus normal tissues, and
system. Because the number of • Inhibition of CTLA-4 and/or PD-1 such therapeutics do not require
cancer-specific antigens expressed signaling on T cells enhances inherent tumor immunogenicity to
varies greatly among individual antitumor immunity by unleashing be effective. Examples of such
patients and among cancer naturally induced adaptive antitumor therapeutics are monoclonal
histologies, cancer immunogenicity immune responses that have antibodies, bispecific antibodies,
varies substantially. undergone active suppression. and T cells engineered to express
• As cancers develop, they are Clinical trials using antibodies that chimeric antigen receptors. T cells
sculpted by “immune pressure” to block CTLA-4 have demonstrated expressing a chimeric antigen
eliminate antigens, or diminish the impressive effects in melanoma, and receptor targeting CD19 have
degree to which antigenic peptides clinical trials using antibodies that demonstrated impressive effects
are processed or presented to the block PD-1 signaling have against B-cell malignancies.

OVERVIEW developments in T-cell checkpoint-blocking antibodies, adoptive


therapies with engineered T cells, and tumor vaccines.
At least two features of the immune system make it a unique therapeutic
tool against cancer. First, the diversity of receptors in the adaptive
immune system (T-cell receptor [TCR] for T cells and antibodies THE ANTIGENIC PROFILE THAT
made by B cells) offers unparalleled capacity for target specificity, far DISTINGUISHES TUMORS FROM
greater than any synthetic drug library. Second, diverse cell-killing NORMAL TISSUES
weaponry from both the innate immune system and cytotoxic T cells
offers the potential to kill any cell, once appropriately recognized. Genetic instability, a hallmark of cancer, is a primary generator of
Central to the concept of successful cancer immunotherapy are the tumor-specific antigens. On average, cancers express between 50 and
dual tenets that tumor cells express an antigenic target distinct from 1000 missense mutations in coding regions, roughly 20% of which
their normal cellular counterparts and that the immune system is create neoantigenic peptides presented by at least one of the individual’s
capable of recognizing these antigenic differences. human leukocyte antigen (HLA) alleles and thus recognized by T
The notion that the immune system can be used as an anticancer cells.6–14 In addition, deletions, amplifications, and chromosomal
therapy emerged from experiments in animal models of carcinogen- rearrangements can result in new genetic sequences resulting from
induced cancer. It was demonstrated that a number of experimentally juxtaposition of coding sequences not normally contiguous in untrans-
induced tumors could be rejected on transplantation into syngeneic formed cells. The vast majority of these mutations occur in intracellular
immunocompetent animals.1–4 Extensive studies by Prehn on the proteins, and therefore the “neoantigens” they encode would not be
phenomenon of tumor rejection suggested that the most potent tumor readily targeted by antibodies. However, the major histocompatibility
rejection antigens were unique to the individual tumor.5 These studies complex (MHC) presentation system for T-cell recognition renders
led to the hypothesis that the immune system may be harnessed to peptides derived from all cellular proteins available on the cell surface
eliminate cancer cells while sparing normal tissue. This chapter reviews as peptide MHC complexes capable of being recognized by T cells.
the biology of tumor–immune system interactions and discusses how In accordance with the original findings of Prehn,5 the vast majority
scientific insights from immunology have translated into novel strategies of tumor-specific antigens derived from genetic mutations are unique
for harnessing the immune response to treat cancer, highlighting recent to individual tumors. As a result, antigen-specific immunotherapies

84
Cancer Immunology  •  CHAPTER 6 85

Immune
surveillance
ELIMINATION
Normal Genetic Tumor Resistance
cell alterations cell mechanisms
SURVIVAL
Transformation
progression x
x
x
x Tolerance induction
SURVIVAL

Figure 6.1  •  The balance among immune surveillance, resistance, and tolerance. Transformation of normal cells to cancer cells involves the creation
of neoantigens as a result of mutation as well as upregulation of self-antigens. Successful immune surveillance of tumors based on recognition of these
tumor-specific antigens would lead to tumor elimination at early stages. Clinically relevant tumor survival and progression require that tumors develop resistance
mechanisms that inhibit tumor-specific immune responses to kill tumor cells. Alternatively, if the tumor develops mechanisms to induce immune tolerance
to its antigens, antitumor effector responses do not develop. Finally, tumors could respond to immune pressure from T cells by eliminating mutations that
are efficiently presented by the tumor’s major histocompatibility complex, a process termed immune editing. Evidence is accumulating that tumors actively
develop immune resistance mechanisms as well as immune tolerance mechanisms in order to survive despite displaying antigens capable of recognition by the
immune system. Evidence for immune editing has been demonstrated in experimental carcinogen-induced tumors in mice but not yet in human cancers.

targeted at truly tumor-specific antigens are most commonly patient progressing cancers in humans) fail to be eliminated because they
specific. However, some tumor-specific mutations are shared (e.g., develop active mechanisms of either immune escape or resistance
KRAS codon 12 G->A in colon and pancreatic cancers; BRAF V600E (Fig. 6.1).
found in melanomas; p53 codon 249 G->T mutation found in Genetically manipulated mice have provided clear evidence that
hepatocellular carcinomas)15–18 and could serve as potential immuno- the immune system can eliminate both carcinogen-induced and
therapy targets. spontaneously arising cancers.32–34 When profoundly immunodeficient
Tumors also manifest global alterations in DNA methylation and mice were treated with carcinogens or crossed onto a cancer-prone
chromatin structure resulting in dramatic shifts in gene expression.19 p53 knockout, the incidence of cancers was modestly but significantly
Tumors often overexpress hundreds of genes relative to their normal increased relative to nonimmunodeficient counterparts when observed
counterparts, including genes that are normally completely silent in over an extended period (greater than 1 year). Further, tumors that
their normal cellular counterparts. Overexpressed genes in tumor cells arise in immunodeficient animals behave as regressor tumors when
are attractive targets for both antibody- and cell-based immunotherapies, transplanted into immunocompetent animals. These findings are
because overexpressed genes are shared among many tumors of a given consistent with a model wherein tumors that arise in immunodeficient
tissue origin or sometimes multiple tumor types. For example, animals would have been eliminated had they arisen in immunocom-
mesothelin, which is targeted by T cells from vaccinated pancreatic petent animals.
cancer patients,20 is highly expressed in virtually all pancreatic cancers, Epidemiologic studies of patients with heritable immunodeficiencies
mesotheliomas, and most ovarian cancers.21,22 Whereas mesothelin is have also confirmed a significantly increased risk of certain cancers
expressed at low to moderate levels in the pleural mesothelium, it is that are distinct from the epithelial cancers commonly observed in
not expressed at all in normal pancreatic or ovarian ductal epithelial normal immunocompetent adults.35–37 Although many cancers observed
cells. Another example of tumor-selective expression of epigenetically in immunodeficient individuals are associated with viral infections,
altered genes is the so-called cancer-testis antigens.23 These genes are (e.g., Epstein-Barr virus [EBV]–associated lymphomas,38 Kaposi
expressed almost exclusively in germ cells of the testis and ovaries, sarcoma–associated herpesvirus [KSHV]–associated Kaposi sarcoma,39
and some appear to encode proteins associated with meiosis.24–26 Many and human papillomavirus [HPV])–associated cervical cancer40,41),
cancer-testis antigens are recognized by T cells from nonvaccinated immunodeficient individuals also demonstrate an increased incidence
and vaccinated cancer patients.23 A final category of tumor antigen of non–pathogen-associated cancers, particularly melanoma.42
consists of tissue-specific differentiation antigens shared by tumors of
similar histologic origin. Commonly generated melanoma-reactive T IMMUNE HALLMARKS OF CANCER:
cells from melanoma patients recognize melanocyte antigens,27 including AVOIDING IMMUNE DESTRUCTION AND
tyrosinase, a melanocyte-specific protein required for melanin syn-
thesis.28,29 Although tissue-specific antigens are not truly tumor specific, TUMOR-PROMOTING INFLAMMATION
they are nonetheless potentially effective targets when the tissue is Avoiding Immune Destruction
dispensable (e.g., prostate cancer or melanoma).
Evidence from murine and human tumors demonstrates the capacity
IMMUNE SURVEILLANCE HYPOTHESIS for tumors to induce T-cell tolerance to their antigens.43–46 Tolerance
OF CANCER induction among tumor antigen–specific T cells can be an active
process involving direct antigen recognition or can be associated with
The immune surveillance hypothesis, first conceived nearly a half failure of antigen recognition by T cells—that is, the immune system
century ago,30,31 proposed that a fundamental role of the immune “ignores” the tumor.47,48 However, tumors do not uniformly tolerize
system is to survey the body for tumors as it does for infection with T cells. Ohashi and colleagues observed that lymphocytic choriomen-
pathogens, recognizing and eliminating them based on their expression ingitis virus (LCMV) GP33–specific TCR transgenic CD8 T cells
of tumor-associated antigens (TAAs). In animal models, carcinogen- adoptively transferred into mice expressing pancreatic islet cell tumors
induced tumors can be divided into those that grow progressively that express GP3349 manifested evidence for CD8 T-cell activation
(termed progressor tumors) and those that are rejected after an initial as a result of antigen cross-presentation in the draining lymph nodes.
period of growth (termed regressor tumors).2,3 The phenomenon of Despite the activation of tumor-specific T cells, the tumors grew
regressor tumors was explained based on ongoing immune surveillance progressively, indicating that the degree of immune activation induced
of cancer. A corollary to the original immune surveillance hypothesis by tumor growth was insufficient to ultimately eliminate the tumors.
is that progressor tumors in animals (presumed to represent clinically These results suggest that in some cases T-cell responses are induced
86 Part I: Science and Clinical Oncology

to developing tumors, but if the level of immune activation ultimately pathway such as ERAP1 and ERAP2, and epigenetic regulation of
does not “keep up” with tumor progression, the ultimate result is TAP genes.53–55
tumor outgrowth. In the case of the LCMV GP33-specific TCR
transgenic mice, because neither anergic nor deletional tolerance was Tumor-Promoting Inflammation in the Tumor
observed, animals treated with the dendritic cell (DC) stimulatory Microenvironment
anti-CD40 antibody demonstrated significant slowing of tumor growth.
Thus depending on the mechanism of immune escape at play, agents The tumor microenvironment is replete with suppressive mechanisms
that affect the overall activation state of either antigen-presenting cells that dampen antitumor immune immunity (Fig. 6.2). The inhibitory
or T cells could be used to shift the balance between tumor immune cells, molecules, and signaling pathways found in the tumor micro-
evasion and tumor immune recognition. environment are not unique to tumors, but rather compose an array
T cells retrieved from patients with cancer tend either to be of of physiologic mechanisms evolved to regulate immune responses to
low affinity for their cognate antigen or to recognize antigens that self-antigens and to downmodulate immune and inflammatory responses
bind poorly to their presenting HLA (human MHC) molecule, resulting to foreign antigens so that collateral tissue damage is limited. Tumors
in inefficient recognition by T cells. Furthermore, tumors commonly co-opt and upregulate mechanisms for resistance to immune attack,
acquire defects in MHC class I expression to avoid immune destruction. and much activity in the field of immuno-oncology is focused on
MHC class I proteins display small peptide antigens on the surface delineating these pathways and developing therapeutics capable of
of cells and are key for immune recognition by CD8 T cells. For reversing the effects of these mediators.
example, activated HRAS leads to reduced levels of messenger RNA
(mRNA) transcripts of the transporter associated with antigen processing Regulatory T Cells and Cancer
(TAP), a key protein in the antigen-processing pathway.50 Repression Regulatory T cells (Tregs) maintain tolerance to self-antigens, down-
of proteins involved in the antigen presentation pathway are linked regulate immune responses to pathogens,56,57 and induce tolerance
to overexpressed epidermal growth factor receptor (EGFR) or HPV to tumor antigens.58,59 CD4+ Tregs are characterized by expression
E7 oncoprotein.51,52 MHC class I downregulation can also result from of Foxp3, a central master regulatory transcription factor.60,61 Other
MHC gene mutations, defects in other genes in the antigen presentation Tregs produce inhibitory cytokines such as interleukin (IL)-10 and

Maturation
Inhibitory cytokines ↑STAT3
Tumor (VEGF, IL-10, TGF-β) DC/MΦ

PD-L1
PD-L1/2

B7-H3 LAG-3
PD-1
IDO B7-H4
↑STAT3
TDO CD4 T cell
Adenosine
Treg
A2aR IFNγ
PD-L1
IL-10, TGF-β
PD-L1
PD-1

LAG-3
Antitumor
↑STAT3 CD8 killer cell Tim-3
PD-L2
MDSC
IDO BTLA

Figure 6.2  •  The immune microenvironment of the tumor is generally inhibitory to antitumor immune responses. Activation of oncogenic pathways
such as STAT3 in the tumor leads to a cascade of molecular and cellular processes in the tumor microenvironment that block the killing function of innate
immune effectors such as natural killer (NK) cells and granulocytes and block dendritic cell (DC) maturation and activation/effector function of Th1 T cells
and CD8 cytotoxic T cells. This inhibitory microenvironment is organized by both cytokines, such as interleukin (IL)-10, IL-6, and transforming growth
factor–β (TGF-β), and multiple cell membrane coinhibitory molecules such as PD-L1, PD-L2, B7-H3, and B7-H4 that interact with their cognate checkpoint
receptors expressed at high levels on T cells in the microenvironment. Myeloid-derived suppressor cells (MDSC) produce nitric oxide (NO) that inhibits T
cells, and both tumor cells and myeloid cells produce tryptophan dioxygenase (TDO) and indoleamine 2,3-dioxygenase (IDO), which deplete tryptophan.
Regulatory T cells (Treg) also accumulate in the tumor microenvironment, further blunting antitumor T-cell responses via production of inhibitory cytokines.
Tumors also produce adenosine as a byproduct of cell death, which inhibits immune responses and enhances local Treg generation and suppressive function
via interaction with the adenosine A2a receptor. BTLA, B- and T-lymphocyte attenuator; IFN-γ, interferon-γ; VEGF, vascular endothelial growth factor.
Cancer Immunology  •  CHAPTER 6 87

transforming growth factor–β (TGF-β). In addition, a recently described II expression (Fig. 6.3). Immature DCs can present antigens to T
IL-12 family “hybrid” cytokine, IL-35, consisting of the alpha subunit of cells, but without costimulatory molecules, induce T-cell tolerance,81,82
IL-12 and the beta subunit of IL-27, is produced by Tregs and suppresses whereas in the context of microbial ligands or endogenous “danger
antitumor immunity.62 Numerous murine studies have demonstrated signals,” DCs are activated to present antigen together with costimula-
that Tregs expand in animals with cancer and limit the potency of tory signals that result in an effector T-cell phenotype. A major
antitumor immune responses.63 It is now appreciated that treatment with inhibitory signaling pathway induced in tumor infiltrating DC is the
low-dose cyclophosphamide is a relatively simple and reasonably effective STAT3 pathway, which, when activated, strongly antagonizes Toll-like
way to temporarily eliminate cycling Tregs.64–67 In addition, antibodies receptor (TLR) and CD40-mediated DC activation. Tumor-derived
neutralizing IL-35 limited tumor growth in multiple preclinical mouse factors such as IL-10, IL-6, and vascular endothelial growth factor
models, but their use alone was not sufficient to eliminate tumors.62 (VEGF) (in part induced by STAT3 signaling in the tumor cell) can
As new cell membrane molecules that define Tregs are identified, the induce STAT3 activation in DCs, and such mechanisms shift tumor-
capacity to block regulatory T-cell activity with antibodies to these specific T cells from a state of activation to tolerance (Fig. 6.4).
molecules presents new opportunities for immunotherapeutic strategies Numerous cancer immunotherapies seek to induce activation and
to break tolerance to tumor antigens. Evidence for a role of Tregs maturation of DCs, such as engaging CD40.
in suppressing antitumor immunity in humans includes an extensive
study correlating Treg number in resected ovarian cancers with clinical Immune Inhibitory Molecules Expressed in the Tumor
outcome. Patients with greater numbers of CD4+CD25hiFoxp3+ cells
had a worse outcome.58 A number of clinical trials have been performed Microenvironment
using a toxin-conjugated IL-2 reagent (denileukin diftitox) that would A large number of immune inhibitory molecules are expressed in the
bind CD25 and selectively kill Tregs. Clinical efficacy of this agent was microenvironment of tumors, spanning enzymes that metabolize certain
described in phase III studies of recurrent or refractory cutaneous T-cell amino acids on which lymphocytes are highly dependent, enzymes
lymphoma, and the drug was approved by the US Food and Drug that produce immune-inhibitory products, cytokines that inhibit
Administration (FDA) for this indication, but the agent is currently not antitumor immune responses by acting on immune effector cells to
available owing to the manufacturer’s decision to prioritize development either inhibit their tumor-lytic activity, and membrane-bound ligands
of a newer improved formulation of the drug.68 that bind to inhibitory receptors on lymphocytes (so-called checkpoints).
Myeloid cells in the tumor microenvironment express a number of
Immature Myeloid Cells and Tumor-Associated Macrophages enzymes whose metabolic activity ultimately results in inhibition of
Immature myeloid cells (iMCs)69,70 are a heterogeneous class of cells T-cell responses. These include production of reactive oxygen species
that include myeloid-derived suppressor cells (MDSCs)71–74 as well as (ROS) and reactive nitrogen species (RNS). NO production by iMCs
tumor-associated macrophages (TAMs). In mice, iMCs and MDSCs and MDSCs as a result of arginase and inducible nitric oxide synthase
are characterized by coexpression of CD11b (considered a macrophage (iNOS) activity has been well documented, and inhibition of this
marker) and Gr1 (considered a granulocyte marker) while expressing pathway with a number of drugs can mitigate the inhibitory effects
low or no MHC class II or the CD86 costimulatory molecule. In of iMCs and MDSCs. ROS, including H2O2, have been reported to
humans, MDSCs are most commonly described as CD33+ but lack block T-cell function associated with the downmodulation of the zeta
MHC class II expression, as well as markers of mature macrophages, (ζ) chain of the TCR signaling complex,83 a phenomenon well rec-
DCs, or granulocytes. A number of molecular species produced by ognized in T cells from cancer patients and associated with generalized
tumors tend to drive iMC and MDSC accumulation, including IL-6, T-cell unresponsiveness.
colony stimulating factor 1 (CSF-1), IL-10, and gangliosides. IL-6 Another mediator of T-cell unresponsiveness associated with cancer
and IL-10 are potent inducers of STAT3 signaling, which has been is the production of indoleamine 2,3-dioxygenase (IDO).84 IDO appears
shown to be important in iMC and MDSC persistence and activity. to be produced by DCs either within tumors or in tumor-draining
Macrophages have been divided into M1 type (Th1/interferon [IFN] lymph nodes. It is interesting to note that IDO in DCs has been
instructed) and M2 type (Th2/IL-4 and IL-13 instructed). M1 reported to be induced via backward signaling by B7-1/2 on ligation
macrophages are characterized by expression of genes encoding the with CTLA-4.85,86 The major IDO-producing DC is thought to be
nitric oxide (NO)–producing iNOS-2 enzyme and the cytokine IL-12, either a plasmacytoid dendritic cell (PDC) or a PDC-related cell that
which amplifies Th1 responses. M1 macrophage activation is thought is B220+87; however, IDO has been subsequently shown to be expressed
to be one of the mediators of Th1-orchestrated antitumor immunity, by multiple cell types in the immune microenvironment, including
whereas M2 macrophages are thought to be tumor promoting. tumor cells themselves.88 IDO appears to inhibit T-cell responses through
Macrophage infiltration into tumors has been largely associated catabolism of tryptophan. Activated T cells are highly dependent on
with poor patient prognosis in multiple tumor types. Therapeutic tryptophan and are therefore sensitive to tryptophan depletion. Thus
strategies for targeting TAMs include inhibition of macrophage Munn and Mellor have proposed a bystander mechanism, whereby
recruitment to the tumor or promotion of their antitumor properties DCs in the local environment deplete tryptophan via IDO upregulation,
or effector function. Strategies for preventing macrophage accumulation thereby inducing metabolic apoptosis in locally activated T cells.84 IDO
in tumors include disruption of the CCL2/CCR2 axis in phase I has two isoforms, IDO-1 and IDO-2, encoded by distinct genes. The
clinical trials in patients with advanced-phase solid cancers.75,76 The role of IDO-2 in human cancer is still unclear; a major IDO-2 poly-
CSF-1/CSF-1R pathway is also being evaluated as a therapeutic strategy morphism in humans encodes an inactive enzyme. Epacadostat is an
to target tumor TAMs, and agents targeting these pathways have been oral inhibitor of IDO-1 and is being tested in combination with
shown to reduce TAM accumulation in tumors or repolarize them pembrolizumab (anti-PD-1) in patients with advanced melanoma,
into an antitumor phenotype in early-phase clinical trials.77 Strategies which has demonstrated promising results in phase I/II clinical trials.89
to promote phagocytosis and antigen presentation by TAMs are very
promising—for example, by disrupting the CD47-SIRPalpha axis, Transforming Growth Factor–β: A Major Inhibitory Cytokine
which has been shown to be effective in preclinical models and is
being tested in ongoing clinical trials.78–80 in the Tumor Microenvironment
TGF-β is a major inhibitory cytokine implicated in blunting antitumor
Immature Dendritic Cells immune responses. TGF-β is produced by a variety of cell types,
DCs found within the tumor microenvironment typically have an including tumor cells, and has pleiotropic physiologic effects. For
immature, unactivated phenotype characterized by low levels of most normal epithelial cells, TGF-β is a potent inhibitor of cell prolifera-
proinflammatory cytokine production, CD86, and surface MHC class tion, causing cell cycle arrest in the G1 stage.90 In many cancer cells,
88 Part I: Science and Clinical Oncology

GM-CSF

IL-4, FLT-3L

Bone marrow Dendritic cell


progenitor progenitor
Antigen uptake/Processing
Microbial infection
No danger danger signals
signals
Exogenous Endogenous
LPS TNF-
CpG CD40L
Tolerizing DC Activated DC

Moderate MHC II
Costimulatory molecules

High MHC II
Costimulatory molecules

Figure 6.3  •  A dendritic cell (DC) can either activate adaptive immunity or tolerize T cells depending on its state of maturation. DC progenitors
develop from hematopoietic (bone marrow–derived) progenitors under the influence of various cytokines, particularly granulocyte-macrophage colony-stimulating
factor (GM-CSF). Under circumstances of microbial infection, specific pathogen-associated molecular patterns (PAMPs) engage pattern recognition receptors
(PRRs), as well as endogenous proinflammatory cytokines and “danger”-associated molecules (DAMPs), inducing DC maturation. PAMPs include unmethylated
CpG tracks of DNA characteristic of DNA viruses and bacteria; uncapped RNA, characteristic of RNA viruses; flagellin, the major protein component of
bacterial flagella; and lipopolysaccharide (LPS), a major bacterial cell wall component. DC maturation leads to upregulation of costimulatory molecules, major
histocompatibility complex (MHC), and certain cytokines, as well as decreased expression of coinhibitory molecules, allowing them to efficiently activate
antigen-specific T cells to effector cells (right side of figure). In the absence of these danger signals, DCs follow a default pathway (left side of figure) in which
they become tolerizing DCs that present antigen to T cells in the absence of costimulatory signals and with an excess of coinhibitory signals. This represents
a steady state pathway for continuous presentation of self-antigens. The consequence is that these T cells are turned off (anergy) or deleted, inducing tolerance.
IL-4, Interleukin-4; TNF-α, tumor necrosis factor–α.

LN
Activation
Activated of tumor-
DCs specific
T cells
Danger

A
Inhibition of
danger LN Anergy/
signals
Tolerizing Deletion of
DCs tumor-
specific
Danger T cells

B
Figure 6.4  •  Inhibition of dendritic cell (DC) activation in the tumor microenvironment can shift tumor-specific immune responses from activation
to tolerance. Based on the scenario presented in Fig. 6.3, if a tumor is able to produce factors that inhibit local DCs from becoming activated in response to
the endogenous danger signals associated with tissue invasion, it could shift tumor-specific T cells from a state of activation (A) to one of tumor-specific tolerance
(B). LN, Lymph node.
Cancer Immunology  •  CHAPTER 6 89

however, mutations in the TGF-β pathway confer resistance to cell CTLA-4 Checkpoint: A Global Regulator of
cycle inhibition, allowing uncontrolled proliferation, and promote T-Cell Activation
tissue invasion through induction of matrix metalloproteinases. In
vivo, TGF-β directly stimulates angiogenesis; this stimulation can be CTLA-4 is expressed exclusively on T cells, where it primarily regulates
blocked by anti-TGF-β antibodies.91 Elevated serum TGF-β levels the amplitude of the early stages of T-cell activation, by counteracting
are associated with poor prognosis in a number of malignancies, the activity of the T-cell costimulatory receptor CD28.97–99 CD28
including prostate cancer,92 lung cancer,93 gastric cancer,94 and bladder does not affect T-cell activation unless the TCR is first engaged by
cancer.92 Several inhibitors of the TGF-β pathway have been tested cognate antigen. Once antigen recognition occurs, CD28 signaling
in phase I clinical trials, including fresolimumab, a human strongly amplifies the TCR signal to activate T cells. CD28 and CTLA-4
anti-TGF-β1/2/3 monoclonal antibody (mAb), and TβM1, a human share identical ligands: CD80 (B7-1) and CD86 (B7-2).100–104 Because
anti-TGFβ1 mAb. These molecules were shown to be safe, but data CTLA-4 has higher affinity for both ligands, its expression on the
on efficacy have been limited or not yet reported.95,96 surface of T cells dampens the activation of T cells both by outcompet-
ing CD28 in binding CD80 and CD86 and by actively delivering
COINHIBITORY LIGANDS AND RECEPTORS inhibitory signals to the T cell.105–110 The specific signaling pathways
THAT DOWNMODULATE TUMOR IMMUNITY by which CTLA-4 blocks T-cell activation are still under investigation,
although a number of studies have suggested that activation of the
Major molecules successfully targeted in clinical cancer immunotherapy phosphatases SHP2 and PP2A are important in counteracting kinase
are the growing class of ligand-receptor pairs, commonly referred to signals induced by TCR and CD28.98 However, CTLA-4 also confers
as immune checkpoints. In considering the mechanism(s) of action of “signaling-independent” T-cell inhibition through sequestration of
inhibitors of various checkpoints, it is critical to appreciate the diversity CD80 and CD86 from CD28 engagement, as well as active removal
of immune functions that they regulate. For example, the two immune from the antigen-presenting cell surface.111 The central role of CTLA-4
checkpoint receptors that were first targeted in the context of clinical in maintaining T-cell activation in check is dramatically demonstrated
cancer immunotherapy, CTLA-4 (CD152) and PD-1 (CD279), regulate by the systemic immune hyperactivation phenotype of CTLA-4
immune responses at very different levels and by very different knockout mice.112,113
mechanisms (Fig. 6.5). The clinical activity of blocking antibodies Even though CTLA-4 is expressed by activated CD8 killer T cells,
for each of these receptors implies that antitumor immunity can be the major physiologic role of CTLA-4 appears to be through distinct
enhanced at multiple levels and that combinatorial strategies can be effects on the two major subsets of CD4 T cells: downmodulation of
intelligently designed, guided by mechanistic considerations and helper T-cell activity and enhancement of regulatory T-cell suppressive
preclinical models. This section focuses particular attention on the activity.97,114,115 CTLA-4 blockade results in a broad enhancement of
CTLA-4 and PD-1 pathways because they were the two checkpoints immune responses dependent on helper T cells, and conversely, CTLA-4
whose inhibition has revolutionized clinical cancer immunotherapy. engagement on Tregs enhances their suppressive function. CTLA-4

B7-1/2 CD28 B7-1/2 CD28


T cell still in
+ secondary +
APC APC
lymphoid tissue

Signal 1 Signal 1

CTLA-4
Activation of naïve Antigen-experienced
or resting T cells T cell

B7-1/2 CD28
Tissue
+ Traffic to or
DC
periphery tumor
Signal 1 Signal 1

PD-L1 PD-1
Activation of naïve Antigen-experienced
or resting T cells Inflammation T cell

Figure 6.5  •  CTLA-4 and PD-1 checkpoints act to regulate different elements of the T-cell response. Naïve T cells and resting T cells express little
CTLA-4 or PD-1 on their surface. On initial T-cell activation via triggering of the T-cell receptor (TCR) by cognate peptide/major histocompatibility complex
(MHC) complexes together with engagement of CD28 by B7-1 and/or B7-2, CTLA-4, which is stored preformed in intracellular vesicles, rapidly migrates
to the cell surface while the T cell is still engaged with its antigen-presenting cell (APC; in general, a dendritic cell), usually in the secondary lymphoid tissue.
The greater the TCR stimulus, the more CTLA-4 is expressed on the surface. Inhibitory interaction of CTLA-4 with B7-1 and B7-2 results in a counterregulatory
signal that downmodulates the ultimate amplitude of T-cell activation (top row). In contrast, the PD-1 checkpoint primarily operates in the periphery
(bottom row). PD-1 is induced more slowly than CTLA-4 when T cells become activated. Inflammatory signals, such as interferon-γ, at sites of effector T-cell
function (in the tissue or in tumors) induce PD-L1 (and PD-L2) expression, which downmodulates tissue immune responses, thereby protecting tissues from
collateral damage.
90 Part I: Science and Clinical Oncology

is a target gene of the transcription factor Foxp3,116,117 the expression to the CIITA locus, which is highly transcriptionally active in B-cell
of which determines the Treg lineage,118,119 and Tregs therefore express lymphomas.153
CTLA-4 constitutively. Although the mechanism by which CTLA-4 Two general mechanisms for regulation of B7-H1/PD-L1 expression
enhances the inhibitory function of Tregs is not known, Treg-specific have emerged: innate (tumor cell intrinsic) and adaptive (tumor cell
CTLA-4 knockout or blockade significantly inhibits their ability to extrinsic) (Fig. 6.6). In the innate (tumor cell intrinsic) mechanism,
regulate both autoimmunity and antitumor immunity.114,115 Thus in oncogenes drive expression of PD-L1. For some tumors such as
considering the mechanism of action for CTLA-4 blockade, both glioblastoma, deletion or silencing of phosphatase and tensin homolog
enhancement of effector CD4 T-cell activity and inhibition of Treg- (PTEN) has been shown to drive PD-L1 expression, implicating the
dependent immune suppression are likely important factors. PI3K-AKT pathway.154 In addition, constitutive anaplastic lymphoma
kinase (ALK) signaling, observed in certain lymphomas and occasionally
PD-1 Checkpoint: A Pathway That Functions Within in lung cancer, has been reported to drive PD-L1/B7-H1 expression
the Tumor Microenvironment via STAT3 signaling.155 In Hodgkin disease, genomic gains in chromo-
some 9p24 are frequently observed, a region containing the oncogene
In contrast to CTLA-4, the major role of PD-1 is to limit the activity JAK2, as well as PD-L1 and PD-L2. PD-L1 expression in tumors can
of T cells in the peripheral tissues at the time of an inflammatory also be driven by IFNs, which illustrates an adaptive mechanism
response to infection and to limit autoimmunity (see Fig. 6.5).120–126 (tumor cell extrinsic) for its regulation. This hypothesis is supported
This translates to a major immune resistance mechanism within the by work from studies in melanoma demonstrating a high correlation
tumor microenvironment.127–129 PD-1 expression is induced when T between cell surface PD-L1/B7-H1 expression on tumor cells and
cells become activated.121 When engaged by one of its ligands, PD-1 lymphocytic infiltration and intratumoral IFN-γ expression. This
inhibits kinases involved in T-cell activation via the phosphatase correlation was seen not only among tumors but within individual
SHP2.120 Because PD-1 engagement inhibits the TCR stop signal, B7-H1/PD-L1+ tumors at the regional level, in which regions of
this pathway may also modify the duration of T-cell/APC or T-cell/ lymphocyte infiltration were exactly the regions where B7-H1/PD-L1
target cell contact.130 PD-1 is expressed on a large proportion of was expressed on both tumor cells and infiltrating leukocytes.129
tumor-infiltrating lymphocytes (TILs) from many different tumor
types.131,132 Similar to CTLA-4, PD-1 is highly expressed on Tregs, Additional Checkpoints Participate in Tumor
where it may enhance their proliferation in the presence of ligand.133 Immune Resistance and Tolerance
Because many tumors are highly infiltrated with Tregs, PD-1 pathway
blockade may also enhance antitumor responses by diminishing the Successful clinical outcomes of CTLA-4 and PD-1 pathway targeting
number and/or suppressive activity of intratumoral Tregs. Increased have garnered interest in a number of additional checkpoints
PD-1 expression on CD8 TILs may reflect an anergic or exhausted (Fig. 6.7). In addition to defined lymphocyte inhibitory receptors, a
state, as has been suggested by decreased cytokine production by number of B7 family inhibitory ligands—in particular B7-H3 (CD276)
PD-1+ versus PD-1− TILs from melanomas.131 PD-1 is also expressed and B7-H4—do not yet have defined receptors, but murine knockout
on other non–T lymphocyte subsets, including B cells, natural killer experiments support an inhibitory role for both these molecules,156
(NK) cells, and macrophages.134,135 Thus whereas PD-1 blockade is and they are upregulated on tumor cells or tumor-infiltrating cells.157
typically viewed as enhancing the activity of effector T cells in tissues Lymphocyte activation gene-3 (Lag-3), T cell immunoglobulin-3 (Tim-3),
and in the tumor microenvironment, it likely also enhances NK activity and adenosine A2a receptor (A2aR) are also associated with inhibition
in tumors and tissues and may also enhance antibody production of lymphocyte activity. Antibody targeting of these receptors, either
either indirectly or through direct effects on PD-1+ B cells.136 PD-1 alone or in combination with a second immune checkpoint blocker,
has also been shown to be a marker of dysfunctional macrophages has been shown to enhance antitumor immunity in animal models
and monocytes.137 of cancer. Because many tumors express multiple inhibitory ligands
The two ligands for PD-1 are B7-H1/PD-L1 (CD274) and B7-DC/ and TILs express multiple inhibitory receptors, dual or triple blockade
PD-L2 (CD273).120,138–140 These B7 family members share 37% of immune checkpoints could further augment antitumor reactivity.
sequence homology and arose via gene duplication, positioning them Human blocking antibodies specific for a number of these “second-
within 100 kb of each other in the genome.140 An unexpected molecular generation” inhibitory receptors are under development.
interaction between PD-L1 and CD80 was discovered,141 whereby Lag-3 was first discovered as a CD4 homologue expressed on
CD80 expressed on T cells (and possibly antigen-presenting cells) can activated CD4 T cells, CD8 T cells, and a subset of NK cells.158
potentially behave as a receptor rather than a ligand, delivering inhibi- Lag-3 has been shown to play a role in enhancing Treg function,159,160
tory signals when engaged by B7-H1142,143; the relevance of this and subsequently to inhibit CD8 effector function.161 Lag-3 binds to
interaction in tumor immune resistance has not yet been determined. MHCII, which is upregulated on some epithelial cancers (typically
PD-L1 has also been proposed to function as a receptor to transmit in response to IFN-γ) but is also expressed on tumor-infiltrating
survival signals to cancer cells and T cells, although the downstream macrophages and DCs. Another Lag-3 ligand that has been recently
pathways mediating this function are not understood.144,145 Understand- proposed is LSECtin, a member of the DC-SIGN family.162 Lag-3 is one
ing the role of these various interactions in given cancer settings is of a number of immune checkpoint receptors coordinately upregulated
highly relevant for selection of both antibodies and recombinant ligands on both Tregs and anergic T cells, and simultaneous blockade can result
for use in the clinic. in enhanced reversal of this anergic state relative to blockade of either
Just as PD-1 is highly expressed on TILs from many cancers, receptor. In particular, PD-1 and Lag-3 are commonly coexpressed on
PD-1 ligands are commonly upregulated on many different human anergic or exhausted T cells.163,164 Dual blockade of Lag-3 and PD-1
tumors.128,146 On solid tumors, the major PD-1 ligand to be expressed could synergize to reverse exhaustion among tumor-specific CD8 T
is B7-H1/PD-L1.128,147,148 In addition to tumor cells, B7-H1/PD-L1 cells. Phase I/IIb clinical trials are underway examining the safety
is commonly expressed on myeloid cells in the tumor microenviron- and efficacy of anti-Lag-3 antibodies alone or in combination with
ment such as macrophages and DCs.149–151 Although most of the nivolumab for the treatment of B-cell malignancies or solid tumors.
analyses of PD-1 ligand expression has focused on B7-H1/PD-L1, Tim-3, first discovered to be expressed on IFN-γ expressing CD4
B7-DC/PD-L2 has also been reported to be upregulated on a number cells and CD8 T cells, binds to the ligand galectin-9, which is
of tumors. B7-DC/PD-L2 is highly upregulated on certain B-cell upregulated in a number of cancer types such as breast cancer.
lymphomas such as primary mediastinal, follicular cell B-cell lym- Additional ligands for Tim-3 have also been identified, including
phoma and Hodgkin disease.152 Upregulation in these lymphomas CEACAM1, HMGB1, and phosphatidyl serine.165–167 In preclinical
is commonly associated with gene amplification or rearrangement models, Tim-3 is often coexpressed with PD-1 on a subset of T cells
Cancer Immunology  •  CHAPTER 6 91

Innate (tumor cell intrinsic) resistance

MHC-pep

TCR

Constitutive tumor signaling


induces PD-L1 on tumor cells
Oncogenic
pathway
(STAT3, Akt) PD-L1 PD-1 T Cell

Tumor

Adaptive resistance
T cell-induced
PD-L1 upregulation

T Cell T Cell

Tumor Tumor IFN-γ

Figure 6.6  •  Two mechanisms for PD-L1 induction on tumors: innate and adaptive. PD-L1 can be constitutively expressed on tumors as a consequence
of oncogene-driven transcriptional activation. Alternatively, PD-L1 can be induced in an adaptive fashion when there are the right inflammatory cytokines in
the tumor microenvironment consequent to an ongoing immune response to the tumor. This mechanism of tumor resistance to immune attack is co-opted
from physiologic PD-L1 expression for tissue protection in the setting of antimicrobial immune responses. Innate and adaptive mechanisms of PD-L1 expression
on tumors can coexist. The adaptive resistance mechanism implies that PD-L1 expression is a “marker” of endogenous antitumor immunity. IFN-γ, Interferon-γ;
MHC, major histocompatibility complex; TCR, T-cell receptor.

that are highly dysfunctional,168 and blocking Tim-3 with anti-Tim-3 non–small cell lung cancer as monotherapy and in combination with
antibodies has been shown to enhance antitumor immunity.157 the PD-1 antibody PDR001 (ClinicalTrial.gov ID: NCT02403193).
Coordinate blockade of PD-1 and Tim3 in preclinical models was
reported to enhance antitumor responses and tumor rejection under IMPLICATIONS FOR CANCER
circumstances in which only modest effects from blockade of each IMMUNOTHERAPY
individual molecule were observed.169–171 It is interesting to note that
Tim-3 has been shown to be upregulated after anti-PD-1 therapy, Recent successes in clinical immunotherapy of cancer have emerged
both in preclinical mouse models of lung cancer and in patients, directly from a basic understanding of molecular and cellular immunol-
supporting upregulated Tim-3 as a mechanism of resistance to anti-PD-1 ogy applied to the cancer setting. Recent advances have been made
therapy.172 Anti-Tim-3 antibodies are currently being studied in phase in three major areas of cellular cancer immunotherapy that seek to
I clinical trials for patients with advanced solid tumors. use T cells as the antitumor weapon: checkpoint blockade, cellular
A2aR binds adenosine and inhibits T-cell responses, in part by therapy, and cancer vaccination.
driving CD4 T cells to express Foxp3 and develop into Tregs.173
Knockout of this receptor results in enhanced and sometimes pathologic Checkpoint Blockade
inflammatory responses to infection. This receptor is particularly
relevant in tumor immunity because the rate of cell death in tumors Therapeutic strategies targeting the CTLA-4 and PD-1 immune
from cell turnover is high and dying cells release adenosine. In addition, checkpoints have changed paradigms in cancer therapy by emphasizing
Tregs express high levels of the exoenzymes CD39, which converts the importance of targeting the immune system rather than the tumor
extracellular adenosine triphosphate (ATP) to adenosine monophosphate and offering the possibility of cure in some patients. CTLA-4 was
(AMP), and CD73, which converts AMP to adenosine.174 A2aR the first immune checkpoint receptor to be clinically targeted in human
engagement by adenosine to drive T cells to become Tregs can produce metastatic melanoma.176 Antibodies targeting PD-1 or its ligand PD-L1
a self-amplifying loop within the tumor, as illustrated by the evidence have subsequently revolutionized treatment of a variety of human
that tumors grow more slowly in A2aR knockout mice and tumor cancers, including lung cancer, melanoma, kidney and bladder cancer,
vaccines are more effective against established tumors in these mice.175 Hodgkin disease, head and neck cancer, mismatch-repair deficient
A2aR can be inhibited either by antibodies that block adenosine cancers, and Merkel cell cancers.177–183
binding or by adenosine analogues, some of which are fairly specific Encouraged by murine studies demonstrating antitumor effects of
for A2aR. Currently, an oral small-molecule inhibitor of A2aR CPI-444 CTLA-4 blockade, human anti-CTLA-4 antibodies were developed
is being tested in advanced cancers as monotherapy and in combination and one antibody, ipilimumab, was shown to produce a roughly 10%
with atezolizumab in phase I clinical trials (ClinicalTrials.gov ID: objective response rate in melanoma according to standard RECIST
NCT02655822). Another oral small molecule inhibitor of A2aR, (Response Evaluation Criteria in Solid Tumors) clinical criteria.184
PBF-509, is being tested in phase I clinical trials for patients with Anti-CTLA-4 produced significant immune-related toxicity in 25%
92 Part I: Science and Clinical Oncology

Antigen-presenting cell
or tumor cell T Cell
CD28
+

B7.1/B7.2 CTLA-4

?
+
PD-L1/PD/L2 PD-1

ICOS-L ICOS
+

B7-H3 ?

B7-H4 ?

LIGHT

HVEM BTLA
+ –

MHC/pep TCR Signal 1

LAG-3

4-1BBL 4-1BB
+
OX40L OX40
+
LIGHT LIGHT-R
+
PS/galectin9 Tim-1/ Tim-3

CD200R CD200

CD40 CD40L
+

Cytokines (IL-1, IL-6, IL-10, IL-12, IL-18)

Figure 6.7  •  Multiple costimulatory and coinhibitory ligand-receptor interactions ultimately determine the amplitude of T-cell activation and the
potency of effector T-cell responses in tissue and tumor. B7 family ligands and CD28 family receptors are shown in purple and TNF/TNFR family
ligand-receptor pairs and shown in blue. There are additional inhibitory ligand-receptor pairs that do not fit into either of these families. Some of the receptors
for B7 family members are not yet discovered. Although TNF/TNFR interactions are usually one-on-one pairs, B7 family ligands often interact with multiple
receptors. HVEM is a TNFR family member; in addition to its interaction with the TNF family member LIGHT, it also interacts with the inhibitory receptor
BTLA, which is a member of the CD28 family. Additional signals of activation or inhibition are contributed by cytokines. BTLA, B- and T-lymphocyte
attenuator; HVEM, herpesvirus entry mediator; ICOS, inducible T-Cell COStimulator; IL, interleukin; MHC, major histocompatibility complex; PS, phos-
phatidylserine; TCR, T-cell receptor; TNF, tumor necrosis factor; TNFR, tumor necrosis factor receptor.

to 30% of patients, commonly involving the skin, liver, or colon. in the demonstration of major clinical efficacy in multiple tumor
Subsequent clinical development of ipilimumab culminated in two types.189,190 The durability of the responses, even after cessation of
successful phase III trials in patients with advanced melanoma and treatment, far exceeds that of chemotherapy or tyrosine kinase inhibitors.
FDA approval in 2011.185,186 Since these studies, ipilimumab has also These findings reinforce the notion that a “reeducated” immune system
been shown to prolong survival in patients with stage III melanoma not only has memory, but also can adapt to maneuvers by the tumor
in the adjuvant setting following surgery.187 In addition, ipilimumab to develop resistance. Just as predicted from the mouse knockout
is being investigated in the neoadjuvant setting in the context of studies, toxicity from anti-PD-1 or anti-PD-L1 therapy is lower than
regionally advanced but surgically operable melanoma.188 from anti-CTLA-4 therapy and also of different distribution than
In the case of PD-1, the basic discoveries that this pathway anti-CTLA-4 therapy (colitis is most common with anti-CTLA-4
downmodulated immune responses in the tissue, together with therapy, and pneumonitis is more common with anti-PD-1).
upregulation of PD-1 ligands in human tumors, motivated clinical A number of interesting clinical insights have emerged from the
testing in the mid-2000s. Despite the fact that most of the patients clinical experience with checkpoint inhibitors, which call for novel
in the initial phase I trial had end-stage disease, a number of dramatic approaches to monitoring the safety and efficacy of these drugs. The
clinical responses were observed in multiple cancer types. This led to ipilimumab experience illustrated that the kinetics of clinical responses
larger clinical trials of both anti-PD-1 and anti-PD-L1, culminating to anti-CTLA-4 (i.e., tumor shrinkage) tends to be slower than
Cancer Immunology  •  CHAPTER 6 93

chemotherapy or tyrosine kinase inhibitors, and in some patients demonstrate higher numbers of intratumoral CD8 T cells and a
tumor regression is proceeded by apparent progression as assessed more clonal TCR repertoire in their pretreatment tumor samples.204
with computed tomography (CT) or magnetic resonance imaging T-cell clonotype was also examined after anti-CTLA-4 treatment
(MRI) scans. This finding suggests a mechanism of action in which in patients with metastatic castrate-resistant prostate cancer and in
ultimate tumor regression is preceded by immune activation and tumor metastatic melanoma. This study demonstrated that maintenance of
infiltration of activated lymphocytes. In addition, although formal high-frequency T-cell clones over the course of treatment, but not
regressions are induced in a relatively small proportion of patients the absolute number of T-cell clonotypes, was associated with clinical
and complete responses as assessed with CT or MRI are rare (<5%), response to anti-CTLA-4 blockade.205 These studies demonstrate the
roughly 20% of patients treated with only four doses of ipilimumab importance of T-cell infiltration and clonal expansion, but illustrate
remained alive more than 4 years after therapy (as opposed to approxi- that there is still much to be learned regarding qualitative aspects
mately 5% in the control group). These clinical findings led to the of T-cell responsiveness, such as antigen specificity and downstream
development of “immune response criteria” to help clinicians evaluate programs associated with an antitumor response. Recently, response
the efficacy of this new class of immunotherapy in their patients.191 to anti-PD-1 therapy in melanoma patients was found to correlate
These agents also changed paradigms for dose selection. For example, with a measure of T-cell reinvigoration in the peripheral blood after
efficacy and toxicity do not always correlate, so immunotherapies may correcting for tumor burden.206 This may be a simple blood-based
not achieve a maximum tolerated dose, and this measurement may immunologic metric that can be assessed as soon as 3 to 6 weeks
be less relevant for selecting a recommended dose in phase I trials to after treatment, but it remains to be seen whether this is predictive,
be used in phase II trials.192 These novel clinical paradigms as well as reproducible, and relevant to other tumor types.
the unique immune toxicity profiles encountered with these agents Recently several studies have examined tumor intrinsic mechanisms
highlight the importance of immune monitoring of patients on clinical of poor T-cell infiltration in cancer. Epigenetic silencing has been
trials to advance the understanding of the mechanistic basis of response shown to be a mechanism for suppressing Th1 chemokines CXCL9
and resistance and to pave the way for improving these therapies. and CXCL10 in human ovarian cancer. Epigenetic reprogramming
Since 2010, there has been tremendous energy focused on under- targeting EZH2 or DNMT1 can lead to upregulation of T cell–
standing the molecular basis of response and resistance to CTLA-4 attracting chemokines and increase efficacy of anti-PD-1 therapy.207
and PD-1 checkpoint pathway blockade. Because PD-L1 was the In addition to epigenetic silencing, another tumor-intrinsic mechanism
major ligand for PD-1 and mediated T-cell suppression, it was associated with poor T-cell trafficking to tumors is the WNT-β-catenin
hypothesized that PD-L1 expression in tumor cells or tumor antigen- pathway. In melanoma, activation of WNT/β-catenin is associated
presenting cells may predict clinical responsiveness to PD-1 blockade. with the absence of a T-cell gene expression signature. In mice, β-catenin
Consistent with this, PD-L1 expression in tumor tissue in melanoma, was found to negatively regulate CCL4 expression, limiting DC
non–small cell lung cancer, and renal cell carcinoma correlates with recruitment and T-cell activation. Intratumoral injection of DCs in
response to anti-PD1 therapy.193–196 Expression of PD-L1 in tumor- this model was effective in overcoming resistance to anti-CTLA-4
infiltrating immune cells also correlated with response to anti-PD-L1 and anti-PD-1 therapy.208 Understanding the tumor-intrinsic mecha-
therapy.197 However, PD-L1 is an imperfect biomarker given its nisms responsible for poor T-cell recruitment to tumors may guide
heterogeneous and dynamic expression combined with the observation rationally designed strategies for overcoming resistance.
that tumors with negative PD-L1 expression may still respond to Genomic and transcriptomic characteristics of response have been
therapy. In patients with melanoma treated with ipilimumab (anti- examined in detail in metastatic melanoma. This approach found that
CTLA4) and nivolumab (anti-PD-1), clinical responses were observed BRCA2 mutations were enriched in patients responding to anti-PD-1
in patients with both high and low PD-L1 expression.198,199 This may therapies. A transcriptional signature of innate PD-1 resistance (IPRES)
be due to variability in PD-L1 expression in different parts of the was described, demonstrating that the upregulation of genes involved
tumor as well as the dynamic nature of PD-L1 expression. in mesenchymal transition, angiogenesis, and wound healing correlated
The burden of cancer neoantigens has also been examined as a with lack of response to anti-PD-1 therapy.209 This raises the hypothesis
predictor of CTLA-4 and PD-1 checkpoint pathway blockade. This that modulation of pathways involved in the IPRES signature may
was initially described in a preclinical mouse sarcoma model, which be a strategy for overcoming resistance to anti-PD-1 therapy. Beyond
demonstrated that tumor neoantigen-specific T cells were activated molecular correlates of response described in the tumor and tumor micro-
by treatment with anti-CTLA-4 and anti-PD-1.200 In patients with environment, the gut microbiome has also been shown to shape response
non–small cell lung cancer treated with pembrolizumab (anti-PD-1), to checkpoint blockade. In murine melanoma studies, administration of
higher nonsynonymous mutation burden and higher neoantigen burden Bifidobacterium improved tumor response to anti-PD-L1 therapy,210 and
were associated with clinical response.201 In patients with melanoma response to anti-CTLA-4 treatment to melanoma correlated with the
treated with CTLA-4 blockade, a neoantigen signature was identified presence of B. thetaiotaomicron or B. fragilis.211 These results demonstrate
that correlated with response to therapy.202 Similarly, colorectal cancers that both host and environmental factors shape responses to CTLA-4
with mismatch-repair deficiency had higher objective response rates to and PD-1 checkpoint pathway blockade in patients.
pembrolizumab (anti-PD-1) treatment compared with mismatch-repair Additional insights from cancer immunology are paving the way
proficient colorectal cancers, and colorectal cancers with mismatch-repair to combinatorial therapies—for instance, combining checkpoint
deficiency also manifested higher somatic mutation loads compared with blockade with additional strategies that improve the efficacy of this
mismatch-repair proficient colorectal cancers.182 Neoantigen intratumoral approach. Combination of anti-PD-1 blockade (nivolumab) with anti-
heterogeneity has also demonstrated that response to PD-1 blockade in CTLA-4 blockade (ipilimumab) in metastatic melanoma has already
lung cancer and CTLA-4 blockade in melanoma were associated with demonstrated improved response rates, but was also associated with
higher clonal neoantigens.203 Together, these data support the notion increased toxicity.199 In the future, a comprehensive personalized assay
that tumor neoantigen burden correlates with response to checkpoint incorporating genetic information from the tumor with information on
blockade. It is important to note, however, that there is broad overlap in relevant target molecules in the tumor immune microenvironment may
mutational and neoantigen load between responders and nonresponders, enable biomarker-based prediction of which immune pathways should
limiting the clinically applicability of these measures as a predictor of be targeted in a specific patient. At this point, multiple strategies for
response. Moreover, quantifying mutational burden in tumors may not improving clinical responses to checkpoint immunotherapy are being
necessarily correlate with immunogenic neoantigen burden. investigated, including strategies targeting the tumor or the tumor
The role of tumor-infiltrating T cells in shaping response to check- microenvironment or other factors such as the microbiome. Examples
point blockade therapy is an active area of investigation. In metastatic of tumor-targeting strategies include immune-stimulating radiation
melanoma, patients responding to pembrolizumab (anti-PD-1) typically strategies, chemotherapeutic agents that activate the IFN signaling
94 Part I: Science and Clinical Oncology

pathway or recruit antigen-presenting cells to the tumor, receptor surface MHC complex. Binding of a specific TCR to its cognate
tyrosine kinase inhibitors that increase T-cell accumulation in the antigen initiates T-cell activation and function.225 In cancer patients,
tumor,212 antibody-drug conjugates that promote antigen presentation T cells expressing tumor-specific TCRs comprise only a small fraction
in tumors,213 or epigenetic modifiers that promote T-cell trafficking to of the total T-cell pool and are often tolerized, or demonstrate low-
tumors.207 Examples of tumor microenvironment–targeting strategies affinity binding to tumor-derived peptides. T cells with higher affinity
include activation of DC and antigen presentation (e.g., with STING TCRs specific to TAAs can be isolated directly from tumor-bearing
agonist, CpG vaccine, neoantigen vaccine, oncolytic virus), inhibition patients or from humanized mice immunized against tumor-specific
of TAMs or MDSCs (e.g., targeting phosphoinositide 3-kinase gamma antigen. On isolation and identification of tumor-specific TCRs and
[PI3Kγ] in myeloid cells,214 inhibiting their recruitment to tumors, pro- their respective variable alpha (α) and beta (β) chains, T cells can be
moting phagocytosis and antigen presentation), inhibition of Tregs (e.g., genetically engineered to clonally express a tumor-specific TCR.226
with denileukin diftitox, neutralizing antibodies to IL-35, or low-dose Genetic engineering can efficiently express a transgenic TCR in T
cyclophosphamide), or activation of effector T cells (such as targeting cells with high frequency and allow for mutagenesis to derive high-
additional immune-suppressive checkpoints or activating tumor necrosis affinity variants of native tumor-reactive TCRs.227–229 In addition,
factor [TNF] receptors). The historical segregation of immunotherapy non-TCR elements can be coexpressed within the T cell to enhance
and small-molecule “tumor-targeted” therapy is being replaced by functionality beyond what could be accomplished by a nonengineered
integrative therapeutic approaches that leverage the effects of signaling T cell. Adoptive cellular immunotherapy with transgenic TCRs seeks
modulators directly on the immune system apart from their effects on the to provide the host with a larger pool of functional T cells capable of
tumor itself. efficiently recognizing tumor-specific antigens and eradicating tumor.
One practical challenge of TCR therapy is MHC restriction of
Immunotherapy Using Adoptive T-Cell Strategies these receptors, which permits targeting of intracellular antigens but
also restricts binding of any particular TCR to a specific HLA allele.
Tumor-Infiltrating Lymphocytes Most clinical trials to date have focused on antigens presented within
Adoptive cell transfer employs ex vivo manipulation and transfer of HLA-A*02, an allele expressed in approximately 40% of Caucasians, to
naturally occurring or genetically engineered immune effector cells. demonstrate proof of principle for the approach, leaving most patients
TILs are a naturally occurring, polyclonal lymphocyte population ineligible for studies involving engineered TCRs because of the absence
found within the tumor environment, primarily composed of T cells, of the HLA-A*02 allele. Because each TCR represents a novel receptor,
a fraction of which express TCRs targeting unique or shared TAAs. targeting the same antigen in patients with different MHC alleles requires
TILs can be isolated from neoplastic lesions and the respective sup- substantial additional preclinical and clinical development.
pressive microenvironment, selected for tumor specificity and expanded Clinical experience with engineered TCRs was initiated in malignant
ex vivo. After lymphodepletion, reinfusion of expanded TILs has yielded melanoma, but applications have since been expanded broadly to both
promising results with tumor-specific cytolysis and T-cell persistence hematologic and solid malignancies. The first clinical trial that
in malignant melanoma.215 One benefit of TIL therapy is the broad demonstrated efficacy of TCR therapy used an HLA-A*02 restricted
repertoire of TCRs that target both defined and undefined tumor TCR targeting the melanoma-associated antigen MART-1, administered
antigens. This contrasts with TCR and chimeric antigen receptor after lymphodepletion.230 Fourteen of 15 patients achieved cellular
(CAR) methods in which tumor targeting is focused on a single defined engraftment at 1 month, with two patients demonstrating objective
tumor antigen.215 Unfortunately, in settings beyond malignant mela- tumor regression and prolonged persistence of modified cells for more
noma, TIL therapy has demonstrated limited clinical efficacy. Alternative than 1 year.230 The response rate in 2 of 15 patients (13%) was inferior
adoptive T-cell therapy strategies that rely on genetic modifications to response rates with use of alternative therapeutic modalities but
of patient-derived T cells driving tumor specificity and enhancing supported further exploration and optimization.230 In contrast to the
T-cell cytotoxicity are coming into favor. experience with MART-1, early clinical trials using a TCR with
specificity for NY-ESO-1, a cancer-testis antigen, demonstrated robust
Genetically Engineered Adoptive T-Cell Strategies clinical responses in synovial sarcoma and melanoma.231,232 A human-
Adoptive immunotherapy using genetically engineered T cells seeks derived affinity-enhanced TCR targeting the NY-ESO–derived antigen
to induce expression of novel genes in cytotoxic T cells that facilitate presented within HLA-A*02 yielded objective clinical responses in
tumor recognition, enhance T-cell activation, induce tumor-specific 61% of patients (11 of 18) with synovial sarcoma and 55% of patients
cytotoxicity, and/or augment immune memory. The two most common (11 of 20) with melanoma. This promising work supported the potential
genetic modifications used are expression of transgenes encoding a of TCR therapy in solid tumors and led the FDA to grant breakthrough
tumor-specific TCR or a CAR. To achieve stable, reliable, prolonged therapy designation for affinity-enhanced TCRs targeting NY-ESO-1
transgene expression, most commonly lentiviral or retroviral transduc- in synovial sarcoma in February 2016. This same NY-ESO-1–specific
tion methods are used.216,217 Electroporation or nanoparticle-mediated TCR was studied in advanced multiple myeloma following autologous
delivery of transposon and transposases can also achieve stable transgene stem cell transplantation and yielded an 80% clinical response rate
expression at lower cost with nanoparticle technology, introducing and demonstrated that T cells, lentivirally transduced to express TCR,
the possibility for in vivo CAR delivery.218–220 Delivery of RNA can expanded and persisted in vivo, trafficked to the bone marrow, and
alternatively be used if transient transgene expression is desirable.221 exhibited tumor-specific cytotoxicity. Currently, promising TCR product
New platforms such as megaTAL222 and CRISPR/Cas9223 are available candidates directed at other TAAs including HPV-16 E6 and E7
to permit gene editing, gene deletion, or more precise gene integration. oncoproteins for the treatment of HPV-associated epithelial malignan-
In contrast to past experiences with genetic manipulation of hema- cies and the PRAME cancer testes antigen for the treatment of acute
topoietic stem cells, retroviral-mediated insertional oncogenesis of T myeloid leukemia (AML)233,234 are under study.
cells has not occurred,224 although it remains a theoretical risk that Important lessons regarding the potential for TCR-mediated toxicity
may vary among different delivery systems. were garnered from early studies of this approach. A phase I/II study
of TCR therapy targeting the MAGE-A3/A9 cancer testes antigens
Genetically Modified T-Cell Receptors for Adoptive demonstrated unexpected fatal neurotoxicity. The transgenic TCR
was later found to broadly bind to peptides derived from MAGE-A2,
Cellular Therapy MAGE-A6, and MAGE-A12, and MAGE-A12 expression was identified
T cells are naturally armed with a widely variable repertoire of TCRs on brain parenchyma.235 A study of MAGE-A3 targeting TCRs in
designed to recognize short contiguous amino acid sequences derived patients with myeloma and melanoma revealed unanticipated fatal
from processed intracellular proteins, presented within the target cell cardiac toxicity236,237 in the first two treated patients. Autopsy revealed
Cancer Immunology  •  CHAPTER 6 95

gross myocardial damage and evidence of histopathologic T-cell infil- B-cell acute lymphoblastic leukemia (B-ALL) that demonstrated not
tration. Although MAGE-A3 expression was not found on cardiac only a high feasibility of generating CAR T cells in this setting but
myocytes, unanticipated cross-reactivity of the engineered TCR was also that B-ALL is exquisitely sensitive to CD19-CAR T-cell therapy.
found to peptides derived from titin, an unrelated protein found on Several groups from different institutions have now demonstrated
striated muscle. These experiences highlight the risks of first-in-human that CD19-targeting CARs can induce striking remissions in adults
targeted therapies and remind us of the experimental nature of the field. and children with relapsed or refractory B-ALL despite significant
interinstitutional heterogeneity in CAR constructs, preparative regimens,
Chimeric Antigen Receptors for Adoptive Cellular Therapy and vector platforms.238,240,249–251 All groups report remarkable complete
CARs are artificial tumor-specific receptors generated most often from remission rates ranging from 70% to 90% in B-ALL, which are
the fusion of an antibody-derived binding domain to T-cell–derived markedly superior to historic complete remission rates of 20% to
signaling domains.238 In contrast to TCRs, antibody binding does not 30% when standard salvage chemotherapy is used in patients with
require antigen presentation within an endogenous MHC complex, relapsed or refractory ALL.251 This work led to FDA approval of
and CARs can therefore recognize surface antigens in the face of CTL019, CD19-targeted CAR on August 2017.
tumor MHC downregulation. Antibodies have variable regions Several insights have been gained from parallel yet varying strategies
composed of a heavy and a light chain that dictate antigen specificity. pursued by the different institutions. It is now clear that in vivo CAR
Genetically crafting a receptor to include a single-chain variable T-cell expansion correlates with clinical response250 and that diminished
fragment (scFv) coding for both heavy and light chain variable fragments persistence of CAR T cells is associated with a higher rate of disease
allows CARs to preserve the antigen specificity of the parental antibody. relapse.252 CAR costimulatory domains were shown to critically affect
T cells require both an activation signal and a costimulatory signal the T-cell capacity for long-term persistence, with endogenous 4-1BB
to induce activation. On encountering a cell that solely stimulates mediating superior persistence as compared with CAR T cells with
CD3ζ in the absence of costimulation, the T cell will become anergic CD28 costimulation.240,251 Different relapse patterns following CAR
to that cell and will be inert upon subsequent encounters. First- therapy were identified with CD19 negative relapse due to tumor
generation CARs linked scFvs to the CD3ζ activation signal alone antigen remodeling under the pressure of targeted CAR being the
without the inclusion of a costimulatory domain and did not achieve most common.240,249,250,253 Additional to surface antigen remodeling,
therapeutic benefit.216 Because most tumors do not express ample tumors can undergo lineage reprogramming and relapse with myeloid
levels of costimulation signals, second-generation CARs have been disease.254
developed to include additional costimulatory domains (most commonly Beyond the anticipated on-target–off-tumor B-cell aplasia expe-
CD28 or 4-1BB)238 and have demonstrated remarkable efficacy. It is rienced following CD19-CAR therapy, cytokine release syndrome
now evident that costimulatory signals are not equivalent and influence (CRS) and neurotoxicity are the two most common ominous toxici-
the persistence and efficacy of the CAR T-cell product. Inclusion of ties observed. CRS is a clinical syndrome characterized by fever and
CD28 potentially confers greater initial cytotoxicity; however, CARs hypotension in context of high levels of proinflammatory cytokines
incorporating CD28 signaling endodomains do not demonstrate that typically develops within 1 to 2 weeks after treatment and can
long-term, high-level CAR persistence.239 In contrast, CARs with range from a mild syndrome with constitutional symptoms to a
4-1BB costimulatory signals often demonstrate persistence, and such severe life-threatening syndrome necessitating intensive care unit–level
CARs are not infrequently found in patients more than 2 years after support. Dramatic cytokine elevations including IL-6, IL-10, IFN-γ,
CAR infusion.240 and TNF-α correlate with CRS severity.255 IL-6 has been implicated
Third-generation CARs, which incorporate multiple costimulatory in propagating the clinical syndrome, and treatment with tocilizumab,
signals, have yet to confer superiority over second-generation CARs.241 an anti-IL6R mAb, has been shown to effectively and promptly reverse
The inclusion of more costimulation may in fact influence the exhaus- the syndrome.256 Corticosteroids provide an alternative treatment for
tion status of the T cell, thereby restricting maximal long-term immune CRS, although uncontrolled trials suggest that they may constrain the
effect. Numerous additional genetic modifications are undergoing antitumor immune response more than IL-6 neutralizing strategies.257
testing in CAR T cells, including expression of modifiers designed to Neurologic toxicities following CAR therapy can occur in the
protect the T cell from the tumor inhibitory microenvironment, such presence or absence of CRS, suggesting that the mechanisms may
as modifying CARs to express molecules designed to prevent signaling differ and cytokine dysregulation alone may not fully explain CAR-
via PD-1, or to augment signaling of activating cytokine transgenes mediated neurotoxicity.256 Symptoms include headaches, confusion,
or chemokine receptors.242 The elegance of the CAR is that on binding delirium, hallucinations, facial nerve palsies, apraxia, ataxia, dysmetria,
of a single scFv, multiple signals can be directly stimulated, facilitating dysphagia, seizures, and altered consciousness that can require airway
maximal activation and tumor-specific cytolysis.243 An additional benefit protection with ventilator support.258 Neurotoxicity has not been shown
of the CAR platform is the potential generalizability to any tumor to directly correlate with central nervous system disease, but CAR T cells
for which an antibody targeting a tumor-specific antigen has been and elevated cytokine levels have been identified in the cerebrospinal
developed. In practice, the efficacy of CARs derived from mAbs varies fluid of patients experiencing neurotoxicity.258 This syndrome is most
significantly, and therefore substantial preclinical optimization is commonly reversible; however, rarely patients have developed rapidly
typically required to generate an effective CAR for a particular target. fatal progression of neurotoxicity and cerebral edema following use of
CD19-CARs, with the precise pathophysiology not fully elucidated at
Clinical Translation of Chimeric Antigen Receptor Therapy this time. Preclinical primate studies259 and clinical scrutiny are critical
Hematologic malignancies to understanding and preventing this devastating neurologic sequela.
The first clinical report demonstrating CAR efficacy described dramatic
tumor regression in a patient with advanced follicular lymphoma Solid tumors
following administration of a second-generation CD19-28z CAR.244 Despite several clinical trials targeting numerous antigens in a variety
Subsequently, three patients with chronic lymphocytic leukemia (CLL) of solid cancers, outcomes using CAR therapy in solid tumors have
treated with CD19-BB.z-CARs achieved 1000-fold CAR T-cell expan- been less encouraging than in hematologic tumors. Barriers implicated
sion and CAR persistence for at least 6 months, some of which conferred in restraining outcomes include a dearth of antigens with conserved,
a memory phenotype by flow cytometry.245–247 Subsequent work in specific tumor expression that spare vital tissues, impaired T-cell
CLL demonstrated the importance of lymphodepletion before adoptive trafficking, immunosuppression within the tumor microenvironment,260
transfer of CAR T cells, and the researchers concluded that disease and challenges related to CAR engineering that affect the potency
response and persistence were inversely correlated with tumor burden.248 and exhaustion propensity of some CARs versus others.239 The most
This early work in lymphoma paved the way for CAR T-cell trials in advanced published solid tumor experiences have been with GD2-CARs
96 Part I: Science and Clinical Oncology

in neuroblastoma and sarcoma and Her2.28.z-CARs in Her2+ sarcomas. in progression-free survival or objective response rate (ORR). The
First-generation GD2-CARs mediated long-term complete responses survival benefit of sipuleucel-T ultimately led to FDA approval in
in 2 of 11 patients with neuroblastoma.261,262 Subsequent investigation 2010. Recently there has been a revival of interest in the field of
with a third-generation GD2-CAR263 resulted in early signs of exhaus- therapeutic cancer vaccinations, and efforts to develop personalized
tion attributed to tonic CAR signaling and limited expansion, per- neoantigen vaccines are underway.
sistence, or clinical response.239 Recent work using a Her2.28.z-CAR
in Her2+ sarcomas resulted in 4 of 17 treated patients achieving stable CONCLUSIONS
disease for 12 weeks to 14 months, and CAR T cells persisted at least
6 weeks at low levels in the peripheral blood.264 A recent case report In this chapter we have discussed several key principles of cancer immu-
of a patient with metastatic glioblastoma multiforme who had a dramatic nology, including tumor associated antigens, the immune surveillance
response to regional delivery of a CAR targeting IL13Rα2265 provides hypothesis, and the immune hallmarks of cancer. We also introduced
additional data suggesting that IL13Rα2 may be a promising target multiple mechanisms by which cancer and the immune system shape
for CAR-based targeting of brain tumors.265 Although results have each other. These include downregulation of tumor antigen presentation,
not been robust in solid tumors to date, these early demonstrations development of a tumor microenvironment suppressing antitumor
of efficacy support the potential of CARs to change outcomes in immunity, and expression of coinhibitory ligands and receptors that
otherwise devastating solid tumors. negatively regulate antitumor immunity. In the last section, we review
three classes of cancer immunotherapy: checkpoint blockade, cellular
Cancer Vaccines therapy, and cancer vaccination. These strategies are currently being
tested across cancer types that vary in their cancer immunogenic-
Cancer vaccines include preventive and treatment strategies. Preventive ity, which likely reflects quantitative neoantigen burden as well as
strategies such as the routine administration of HPV vaccination to qualitative properties such as antigen binding to HLA, the nature of
prevent HPV-driven cervical cancer and hepatitis B vaccination to T-cell recognition of these antigen/HLA complexes, and qualitative
prevent hepatocellular carcinoma have proven highly effective, safe, distinctions within the immunosuppressive tumor microenvironment.
and feasible. The first successful treatment vaccine to yield clinical Current models hold that checkpoint blockade is more effective for
benefit in a randomized phase III cancer vaccine trial was a putative immunogenic tumors, whereas engineered immunotherapeutics such
DC vaccine, sipuleucel-T, used in patients with advanced hormone- as T cells engineered to express CARs may mediate similar antitumor
resistant prostate cancer.266 This vaccine is based on the concept that effects regardless of the level of inherent tumor immunogenicity. Cancer
optimal T-cell activation requires antigen processing and presentation vaccines offer a promising approach for shaping and focusing an
by DCs, with the capacity to concomitantly deliver strong costimulatory antitumor immune response, but there is still much to be understood
signals in the form of membrane ligands and secreted cytokines. regarding how to identify the types of antigens to target, the number
Sipuleucel-T is a patient-specific vaccine produced by transiently of antigens, and how to produce the product in a streamlined manner.
incubating the patient’s own peripheral blood mononuclear cells with In reality, there is broad overlap of tumor immunogenicity across
a fusion protein consisting of prostatic acid phosphatase (PAP; a tumor types, and tumors of the same histologic type also vary in
prostate/prostate cancer–specific antigen) linked to the DC growth their immunogenicity. Therefore advances in clinical immunotherapy
and differentiation factor GM-CSF. A 4-month overall survival benefit aimed at augmenting clinical responses while minimizing toxicity
relative to the control arm (uncultured peripheral blood mononuclear requires a deeper understanding of the molecular determinants of
cells without PAP/GM-CSF fusion protein) in the absence of objective antitumor immunity.
tumor regressions or effect on time to progression emphasizes a
developing clinical paradigm in immunotherapy—that immunotherapy The complete reference list is available online at
can potentially provide overall survival benefits that are not reflected ExpertConsult.com.

KEY REFERENCES
177. Reck M, Rodriguez-Abreu D, Robinson AG, 200. Gubin MM, Zhang X, Schuster H, et al. Checkpoint A3-directed T cells. Sci Transl Med. 2013;5(197):
et al. Pembrolizumab versus chemotherapy for blockade cancer immunotherapy targets tumour- 197ra103.
PD-L1-positive non-small-cell lung cancer. N Engl specific mutant antigens. Nature. 2014;515(7528): 239. Long AH, Haso WM, Shern JF, et al. 4-1BB
J Med. 2016;375(19):1823–1833. 577–581. costimulation ameliorates T cell exhaustion induced
179. Motzer RJ, Escudier B, McDermott DF, et al. 201. Rizvi NA, Hellmann MD, Snyder A, et al. Cancer by tonic signaling of chimeric antigen receptors. Nat
Nivolumab versus everolimus in advanced renal-cell immunology. Mutational landscape determines Med. 2015;21(6):581–590.
carcinoma. N Engl J Med. 2015;373(19):1803–1813. sensitivity to PD-1 blockade in non-small cell lung 240. Maude SL, Frey N, Shaw PA, et al. Chimeric antigen
181. Ferris RL, Blumenschein G Jr, Fayette J, et al. cancer. Science. 2015;348(6230):124–128. receptor T cells for sustained remissions in leukemia.
Nivolumab for recurrent squamous-cell carcinoma 202. Snyder A, Makarov V, Merghoub T, et al. Genetic N Engl J Med. 2014;371(16):1507–1517.
of the head and neck. N Engl J Med. 2016;375(19): basis for clinical response to CTLA-4 blockade 244. Kochenderfer JN, Wilson WH, Janik JE, et al.
1856–1867. in melanoma. N Engl J Med. 2014;371(23): Eradication of B-lineage cells and regression of
182. Le DT, Uram JN, Wang H, et al. PD-1 Blockade 2189–2199. lymphoma in a patient treated with autologous
in tumors with mismatch-repair deficiency. N Engl 204. Tumeh PC, Harview CL, Yearley JH, et al. PD-1 T cells genetically engineered to recognize CD19.
J Med. 2015;372(26):2509–2520. blockade induces responses by inhibiting adaptive Blood. 2010;116(20):4099–4102.
186. Robert C, Thomas L, Bondarenko I, et al. Ipili- immune resistance. Nature. 2014;515(7528):568– 250. Lee DW, Kochenderfer JN, Stetler-Stevenson M,
mumab plus dacarbazine for previously untreated 571. et al. T cells expressing CD19 chimeric antigen
metastatic melanoma. N Engl J Med. 2011;364(26): 207. Peng D, Kryczek I, Nagarsheth N, et al. Epigenetic receptors for acute lymphoblastic leukaemia in
2517–2526. silencing of TH1-type chemokines shapes tumour children and young adults: a phase 1 dose-escalation
190. Topalian SL, Hodi FS, Brahmer JR, et al. Safety, immunity and immunotherapy. Nature. 2015; trial. Lancet. 2015;385(9967):517–528.
activity, and immune correlates of anti-PD-1 527(7577):249–253. 257. Davila ML, Riviere I, Wang X, et al. Efficacy and
antibody in cancer. N Engl J Med. 2012;366(26): 231. Robbins PF, Morgan RA, Feldman SA, et al. Tumor toxicity management of 19-28z CAR T cell therapy
2443–2454. regression in patients with metastatic synovial cell in B cell acute lymphoblastic leukemia. Sci Transl
195. Garon EB, Rizvi NA, Hui R, et al. Pembrolizumab sarcoma and melanoma using genetically engineered Med. 2014;6(224):224ra225.
for the treatment of non-small-cell lung cancer. N lymphocytes reactive with NY-ESO-1. J Clin Oncol. 266. Kantoff PW, Higano CS, Shore ND, et al. Sipuleucel-
Engl J Med. 2015;372(21):2018–2028. 2011;29(7):917–924. T immunotherapy for castration-resistant prostate
198. Wolchok JD, Kluger H, Callahan MK, et al. 236. Cameron BJ, Gerry AB, Dukes J, et al. Identifica- cancer. N Engl J Med. 2010;363(5):411–422.
Nivolumab plus ipilimumab in advanced melanoma. tion of a Titin-derived HLA-A1-presented peptide
N Engl J Med. 2013;369(2):122–133. as a cross-reactive target for engineered MAGE
Cancer Immunology  •  CHAPTER 6 96.e1

REFERENCES
1. Baldwin RW. Immunity to methylcholanthrene- analysis of gene expression (SAGE). Clin Cancer 41. Boshart M, Gissmann L, Ikenberg H, Kleinheinz
induced tumours in inbred rats following atrophy Res. 2001;7(12):3862–3868. A, Scheurlen W, zur Hausen H. A new type of
and regression of the implanted tumours. Br J 22. Chang K, Pastan I. Molecular cloning of mesothelin, papillomavirus DNA, its presence in genital cancer
Cancer. 1955;9(4):652–657. a differentiation antigen present on mesothelium, biopsies and in cell lines derived from cervical cancer.
2. Foley EJ. Antigenic properties of methylcholanthrene- mesotheliomas, and ovarian cancers. Proc Natl Acad EMBO J. 1984;3(5):1151–1157.
induced tumors in mice of the strain of origin. Sci USA. 1996;93(1):136–140. 42. Euvrard S, Kanitakis J, Pouteil-Noble C, Claudy
Cancer Res. 1953;13(12):835–837. 23. Van Der Bruggen P, Zhang Y, Chaux P, et al. A, Touraine JL. Skin cancers in organ transplant
3. Gross L. Intradermal immunization of C3H mice Tumor-specific shared antigenic peptides recog- recipients. Ann Transplant. 1997;2(4):28–32.
against a sarcoma that originated in an animal of nized by human T cells. Immunol Rev. 2002;188: 43. Bogen B. Peripheral T cell tolerance as a tumor escape
the same line. Cancer Res. 1943;3(5):326–333. 51–64. mechanism: deletion of CD4+ T cells specific for
4. Old LJ, Carswell EA, Boyse EA, Clarke DA. 24. Madsen B, Tarsounas M, Burchell JM, Hall D, a monoclonal immunoglobulin idiotype secreted
Antigenic properties of chemically induced tumors. Poulsom R, Taylor-Papadimitriou J. PLU-1, a tran- by a plasmacytoma. Eur J Immunol. 1996;26(11):
Ann N Y Acad Sci. 1962;101(1):80–&. scriptional repressor and putative testis-cancer antigen, 2671–2679.
5. Prehn RT, Main JM. Immunity to methylcho- has a specific expression and localisation pattern 44. Bogen B, Munthe L, Sollien A, et al. Naive CD4+
lanthrene-induced sarcomas. J Natl Cancer Inst. during meiosis. Chromosoma. 2003;112(3):124– T cells confer idiotype-specific tumor resistance in
1957;18(6):769–778. 132. the absence of antibodies. Eur J Immunol. 1995;
6. Bronner CE, Baker SM, Morrison PT, et al. Muta- 25. Osterlund C, Tohonen V, Forslund KO, Nordqvist K. 25(11):3079–3086.
tion in the DNA mismatch repair gene homologue Mage-b4, a novel melanoma antigen (MAGE) gene 45. Sotomayor EM, Borrello I, Rattis FM, et al. Cross-
hMLH1 is associated with hereditary non-polyposis specifically expressed during germ cell differentiation. presentation of tumor antigens by bone marrow-
colon cancer. Nature. 1994;368(6468):258–261. Cancer Res. 2000;60(4):1054–1061. derived antigen-presenting cells is the dominant
7. Fearon ER, Vogelstein B. A genetic model for 26. Tureci O, Sahin U, Zwick C, Koslowski M, Seitz G, mechanism in the induction of T-cell tolerance
colorectal tumorigenesis. Cell. 1990;61(5):759– Pfreundschuh M. Identification of a meiosis-specific during B-cell lymphoma progression. Blood. 2001;
767. protein as a member of the class of cancer/testis 98(4):1070–1077.
8. Fishel R, Lescoe MK, Rao MRS, et al. The human antigens. Proc Natl Acad Sci USA. 1998;95(9): 46. Staveley-O’Carroll K, Sotomayor E, Montgomery
mutator gene homolog Msh2 and its association 5211–5216. J, et al. Induction of antigen-specific T cell anergy:
with hereditary nonpolyposis colon-cancer. Cell. 27. Kawakami Y, Robbins PF, Wang RF, Parkhurst M, an early event in the course of tumor progression.
1993;75(5):1027–1038. Kang XQ, Rosenberg SA. The use of melanosomal Proc Natl Acad Sci USA. 1998;95(3):1178–1183.
9. Gowen LC, Avrutskaya AV, Latour AM, Koller proteins in the immunotherapy of melanoma. 47. Speiser DE, Miranda R, Zakarian A, et al. Self
BH, Leadon SA. BRCA1 required for transcription- J Immunother. 1998;21(4):237–246. antigens expressed by solid tumors Do not efficiently
coupled repair of oxidative DNA damage (Retracted 28. Brichard V, Vanpel A, Wolfel T, et al. The Tyrosinase stimulate naive or activated T cells: implications
article. See vol 300, pg 1657, June 13 2003). Science. gene codes for an antigen recognized by autologous for immunotherapy. J Exp Med. 1997;186(5):645–
1998;281(5379):1009–1012. cytolytic T-lymphocytes on HLA-A2 melanomas. 653.
10. Leach FS, Nicolaides NC, Papadopoulos N, et al. J Exp Med. 1993;178(2):489–495. 48. Wick M, Dubey P, Koeppen H, et al. Antigenic
Mutations of a Muts homolog in hereditary non- 29. Topalian SL, Rivoltini L, Mancini M, et al. Human cancer cells grow progressively in immune hosts
polyposis colorectal-cancer. Cell. 1993;75(6):1215– CD4+ T cells specifically recognize a shared mela- without evidence for T cell exhaustion or systemic
1225. noma-associated antigen encoded by the tyrosinase anergy. J Exp Med. 1997;186(2):229–238.
11. Lu X, Lane DP. Differential induction of tran- gene. Proc Natl Acad Sci USA. 1994;91(20):9461– 49. Nguyen LT, Elford AR, Murakami K, et al. Tumor
scriptionally active P53 following UV or ionizing- 9465. growth enhances cross-presentation leading to limited
radiation—defects in chromosome instability 30. Burnet FM. The concept of immunological surveil- T cell activation without tolerance. J Exp Med.
syndromes. Cell. 1993;75(4):765–778. lance. Prog Exp Tumor Res. 1970;13:1–27. 2002;195(4):423–435.
12. Papadopoulos N, Nicolaides NC, Wei YF, et al. 31. Thomas L. In: Lawrence HS, ed. Discussion of 50. Seliger B, Harders C, Lohmann S, et al. Down-
Mutation of a mutL homolog in hereditary colon Cellular and Humoral Aspects of the Hypersensitive regulation of the MHC class I antigen-processing
cancer. Science. 1994;263(5153):1625–1629. States. New York: Hoeber-Harper; 1959. machinery after oncogenic transformation of murine
13. Sharan SK, Morimatsu M, Albrecht U, et al. 32. Dunn GP, Koebel CM, Schreiber RD. Interferons, fibroblasts. Eur J Immunol. 1998;28(1):122–133.
Embryonic lethality and radiation hypersensitivity immunity and cancer immunoediting. Nat Rev 51. Concha-Benavente F, Srivastava RM, Ferrone S,
mediated by Rad51 in mice lacking Brca2. Nature. Immunol. 2006;6(11):836–848. Ferris RL. EGFR-mediated tumor immunoescape:
1997;386(6627):804–810. 33. Fenner JE, Starr R, Cornish AL, et al. Suppressor the imbalance between phosphorylated STAT1 and
14. Sjoblom T, Jones S, Wood LD, et al. The consensus of cytokine signaling 1 regulates the immune phosphorylated STAT3. Oncoimmunology. 2013;
coding sequences of human breast and colorectal response to infection by a unique inhibition of 2(12):e27215.
cancers. Science. 2006;314(5797):268–274. type I interferon activity. Nat Immunol. 2006;7(1): 52. Georgopoulos NT, Proffitt JL, Blair GE. Transcrip-
15. Almoguera C, Shibata D, Forrester K, Martin J, 33–39. tional regulation of the major histocompatibility
Arnheim N, Perucho M. Most human carcinomas 34. Shankaran V, Ikeda H, Bruce AT, et al. IFNgamma complex (MHC) class I heavy chain, TAP1 and
of the exocrine pancreas contain mutant c-K-ras and lymphocytes prevent primary tumour develop- LMP2 genes by the human papillomavirus (HPV)
genes. Cell. 1988;53(4):549–554. ment and shape tumour immunogenicity. Nature. type 6b, 16 and 18 E7 oncoproteins. Oncogene.
16. Bressac B, Kew M, Wands J, Ozturk M. Selective 2001;410(6832):1107–1111. 2000;19(42):4930–4935.
G to T mutations of p53 gene in hepatocellular 35. Gaidano G, Dalla-Favera R. Biologic aspects of 53. Setiadi AF, Omilusik K, David MD, et al. Epigenetic
carcinoma from southern Africa. Nature. 1991; human immunodeficiency virus-related lymphoma. enhancement of antigen processing and presentation
350(6317):429–431. Curr Opin Oncol. 1992;4(5):900–906. promotes immune recognition of tumors. Cancer
17. Davies H, Bignell GR, Cox C, et al. Mutations of 36. Penn I. Tumors of the immunocompromised patient. Res. 2008;68(23):9601–9607.
the BRAF gene in human cancer. Nature. 2002; Annu Rev Med. 1988;39:63–73. 54. Khan AN, Gregorie CJ, Tomasi TB. Histone
417(6892):949–954. 37. Knowles DM. In: Frizzera G, ed. Neoplastic deacetylase inhibitors induce TAP, LMP, Tapasin
18. Forrester K, Almoguera C, Han K, Grizzle WE, Hematopathology. Baltimore: Williams & Wilkins; genes and MHC class I antigen presentation by
Perucho M. Detection of high incidence of K-ras 1992:459–495. melanoma cells. Cancer Immunol Immunother.
oncogenes during human colon tumorigenesis. 38. Heslop HE, Ng CY, Li C, et al. Long-term 2008;57(5):647–654.
Nature. 1987;327(6120):298–303. restoration of immunity against Epstein-Barr virus 55. Fruci D, Giacomini P, Nicotra MR, et al. Altered
19. Jones PA, Baylin SB. The epigenomics of cancer. infection by adoptive transfer of gene-modified expression of endoplasmic reticulum aminopeptidases
Cell. 2007;128(4):683–692. virus-specific T lymphocytes. Nat Med. 1996;2(5): ERAP1 and ERAP2 in transformed non-lymphoid
20. Thomas AM, Santarsiero LM, Lutz ER, et al. 551–555. human tissues. J Cell Physiol. 2008;216(3):742–
Mesothelin-specific CD8(+) T cell responses 39. Mesri EA, Cesarman E, Arvanitakis L, et al. 749.
provide evidence of in vivo cross-priming by Human herpesvirus-8/Kaposi’s sarcoma–associated 56. Sakaguchi S, Ono M, Setoguchi R, et al. Foxp3+
antigen-presenting cells in vaccinated pancreatic herpesvirus is a new transmissible virus that infects CD25+ CD4+ natural regulatory T cells in dominant
cancer patients. J Exp Med. 2004;200(3):297–306. B cells. J Exp Med. 1996;183(5):2385–2390. self-tolerance and autoimmune disease. Immunol
21. Argani P, Iacobuzio-Donahue C, Ryu B, et al. 40. Beaudenon S, Kremsdorf D, Obalek S, et al. Plurality Rev. 2006;212:8–27.
Mesothelin is overexpressed in the vast majority of of genital human papillomaviruses: characterization 57. Sakaguchi S, Sakaguchi N, Asano M, Itoh M, Toda
ductal adenocarcinomas of the pancreas: identifica- of two new types with distinct biological properties. M. Immunologic self-tolerance maintained by acti-
tion of a new pancreatic cancer marker by serial Virology. 1987;161(2):374–384. vated T cells expressing IL-2 receptor alpha-chains
96.e2 Part I: Science and Clinical Oncology

(CD25). Breakdown of a single mechanism of 76. Nywening TM, Wang-Gillam A, Sanford DE, 93. Hasegawa Y, Takanashi S, Kanehira Y, Tsushima T,
self-tolerance causes various autoimmune diseases. et al. Targeting tumour-associated macrophages Imai T, Okumura K. Transforming growth factor-
J Immunol. 1995;155(3):1151–1164. with CCR2 inhibition in combination with FOL- beta1 level correlates with angiogenesis, tumor
58. Curiel TJ, Coukos G, Zou L, et al. Specific recruit- FIRINOX in patients with borderline resectable progression, and prognosis in patients with nonsmall
ment of regulatory T cells in ovarian carcinoma and locally advanced pancreatic cancer: a single- cell lung carcinoma. Cancer. 2001;91(5):964–
fosters immune privilege and predicts reduced centre, open-label, dose-finding, non-randomised, 971.
survival. Nat Med. 2004;10(9):942–949. phase 1b trial. Lancet Oncol. 2016;17(5):651– 94. Saito H, Tsujitani S, Oka S, et al. The expression
59. Liyanage UK, Moore TT, Joo HG, et al. Prevalence 662. of transforming growth factor-beta1 is significantly
of regulatory T cells is increased in peripheral blood 77. Ries CH, Cannarile MA, Hoves S, et al. Targeting correlated with the expression of vascular endothelial
and tumor microenvironment of patients with tumor-associated macrophages with anti-CSF-1R growth factor and poor prognosis of patients with
pancreas or breast adenocarcinoma. J Immunol. antibody reveals a strategy for cancer therapy. Cancer advanced gastric carcinoma. Cancer. 1999;86(8):
2002;169(5):2756–2761. Cell. 2014;25(6):846–859. 1455–1462.
60. Williams LM, Rudensky AY. Maintenance of the 78. Tseng D, Volkmer JP, Willingham SB, et al. Anti- 95. Morris JC, Tan AR, Olencki TE, et al. Phase
Foxp3-dependent developmental program in mature CD47 antibody-mediated phagocytosis of cancer by I study of GC1008 (fresolimumab): a human
regulatory T cells requires continued expression of macrophages primes an effective antitumor T-cell anti-transforming growth factor-beta (TGFbeta)
Foxp3. Nat Immunol. 2007;8(3):277–284. response. Proc Natl Acad Sci USA. 2013;110(27): monoclonal antibody in patients with advanced
61. Zheng Y, Rudensky AY. Foxp3 in control of 11103–11108. malignant melanoma or renal cell carcinoma. PLoS
the regulatory T cell lineage. Nat Immunol. 79. Willingham SB, Volkmer JP, Gentles AJ, et al. ONE. 2014;9(3):e90353.
2007;8(5):457–462. The CD47-signal regulatory protein alpha (SIRPa) 96. Cohn A, Lahn MM, Williams KE, et al. A phase
62. Turnis ME, Sawant DV, Szymczak-Workman AL, interaction is a therapeutic target for human solid I dose-escalation study to a predefined dose of a
et al. Interleukin-35 limits anti-tumor immu- tumors. Proc Natl Acad Sci USA. 2012;109(17): transforming growth factor-beta1 monoclonal
nity. Immunity. 2016;44(2):316–329. 6662–6667. antibody (TbetaM1) in patients with metastatic
63. Sutmuller RP, van Duivenvoorde LM, van Elsas 80. Majeti R, Chao MP, Alizadeh AA, et al. CD47 is an cancer. Int J Oncol. 2014;45(6):2221–2231.
A, et al. Synergism of cytotoxic T lymphocyte- adverse prognostic factor and therapeutic antibody 97. Lenschow DJ, Walunas TL, Bluestone JA. CD28/B7
associated antigen 4 blockade and depletion of target on human acute myeloid leukemia stem cells. system of T cell costimulation. Annu Rev Immunol.
CD25(+) regulatory T cells in antitumor therapy Cell. 2009;138(2):286–299. 1996;14:233–258.
reveals alternative pathways for suppression of 81. Bonifaz L, Bonnyay D, Mahnke K, Rivera M, 98. Rudd CE, Taylor A, Schneider H. CD28 and
autoreactive cytotoxic T lymphocyte responses. Nussenzweig MC, Steinman RM. Efficient CTLA-4 coreceptor expression and signal transduc-
J Exp Med. 2001;194(6):823–832. targeting of protein antigen to the dendritic cell tion. Immunol Rev. 2009;229(1):12–26.
64. Berd D, Maguire HC Jr, Mastrangelo MJ. Induc- receptor DEC-205 in the steady state leads to 99. Schwartz RH. Costimulation of T lymphocytes: the
tion of cell-mediated immunity to autologous antigen presentation on major histocompatibility role of CD28, CTLA-4, and B7/BB1 in interleukin-2
melanoma cells and regression of metastases after complex class I products and peripheral CD8+ T production and immunotherapy. Cell. 1992;71(7):
treatment with a melanoma cell vaccine preceded by cell tolerance. J Exp Med. 2002;196(12):1627–1638. 1065–1068.
cyclophosphamide. Cancer Res. 1986;46(5):2572– 82. Steinman RM, Hawiger D, Nussenzweig MC. 100. Azuma M, Ito D, Yagita H, et al. B70 antigen is
2577. Tolerogenic dendritic cells. Annu Rev Immunol. a second ligand for CTLA-4 and CD28. Nature.
65. Berd D, Mastrangelo MJ, Engstrom PF, Paul A, 2003;21:685–711. 1993;366(6450):76–79.
Maguire H. Augmentation of the human immune 83. Schmielau J, Finn OJ. Activated granulocytes and 101. Freeman GJ, Gribben JG, Boussiotis VA, et al.
response by cyclophosphamide. Cancer Res. 1982; granulocyte-derived hydrogen peroxide are the Cloning of B7-2: a CTLA-4 counter-receptor that
42(11):4862–4866. underlying mechanism of suppression of t-cell costimulates human T cell proliferation. Science.
66. Ercolini AM, Ladle BH, Manning EA, et al. Recruit- function in advanced cancer patients. Cancer Res. 1993;262(5135):909–911.
ment of latent pools of high-avidity CD8(+) T cells 2001;61(12):4756–4760. 102. Hathcock KS, Laszlo G, Dickler HB, Bradshaw J,
to the antitumor immune response. J Exp Med. 84. Munn DH, Sharma MD, Lee JR, et al. Potential Linsley P, Hodes RJ. Identification of an alternative
2005;201(10):1591–1602. regulatory function of human dendritic cells express- CTLA-4 ligand costimulatory for T cell activation.
67. North RJ. Cyclophosphamide-facilitated adoptive ing indoleamine 2,3-dioxygenase. Science. 2002;297 Science. 1993;262(5135):905–907.
immunotherapy of an established tumor depends (5588):1867–1870. 103. Linsley PS, Brady W, Urnes M, Grosmaire LS, Damle
on elimination of tumor-induced suppressor T cells. 85. Baban B, Hansen AM, Chandler PR, et al. A minor NK, Ledbetter JA. CTLA-4 is a second receptor
J Exp Med. 1982;155(4):1063–1074. population of splenic dendritic cells expressing CD19 for the B cell activation antigen B7. J Exp Med.
68. Prince HM, Duvic M, Martin A, et al. Phase III mediates IDO-dependent T cell suppression via type 1991;174(3):561–569.
placebo-controlled trial of denileukin diftitox for I IFN signaling following B7 ligation. Int Immunol. 104. Linsley PS, Clark EA, Ledbetter JA. T-cell antigen
patients with cutaneous T-cell lymphoma. J Clin 2005;17(7):909–919. CD28 mediates adhesion with B cells by interacting
Oncol. 2010;28(11):1870–1877. 86. Mellor AL, Chandler P, Baban B, et al. Specific with activation antigen B7/BB-1. Proc Natl Acad Sci
69. Kusmartsev S, Gabrilovich DI. Role of immature subsets of murine dendritic cells acquire potent T USA. 1990;87(13):5031–5035.
myeloid cells in mechanisms of immune evasion in cell regulatory functions following CTLA4-mediated 105. Egen JG, Allison JP. Cytotoxic T lymphocyte
cancer. Cancer Immunol Immunother. 2006;55(3):237– induction of indoleamine 2,3 dioxygenase. Int antigen-4 accumulation in the immunological
245. Immunol. 2004;16(10):1391–1401. synapse is regulated by TCR signal strength.
70. Young MR, Petruzzelli GJ, Kolesiak K, Achille N, 87. Munn DH, Sharma MD, Hou D, et al. Expression Immunity. 2002;16(1):23–35.
Lathers DM, Gabrilovich DI. Human squamous of indoleamine 2,3-dioxygenase by plasmacytoid 106. Linsley PS, Greene JL, Brady W, Bajorath J, Ledbet-
cell carcinomas of the head and neck chemoattract dendritic cells in tumor-draining lymph nodes. ter JA, Peach R. Human B7-1 (CD80) and B7-2
immune suppressive CD34(+) progenitor cells. Hum J Clin Invest. 2004;114(2):280–290. (CD86) bind with similar avidities but distinct
Immunol. 2001;62(4):332–341. 88. Uyttenhove C, Pilotte L, Theate I, et al. Evidence kinetics to CD28 and CTLA-4 receptors. Immunity.
71. Bronte V, Apolloni E, Cabrelle A, et al. Identification for a tumoral immune resistance mechanism 1994;1(9):793–801.
of a CD11b(+)/Gr-1(+)/CD31(+) myeloid progeni- based on tryptophan degradation by indoleamine 107. Parry RV, Chemnitz JM, Frauwirth KA, et al.
tor capable of activating or suppressing CD8(+) T 2,3-dioxygenase. Nat Med. 2003;9(10):1269– CTLA-4 and PD-1 receptors inhibit T-cell activation
cells. Blood. 2000;96(12):3838–3846. 1274. by distinct mechanisms. Mol Cell Biol. 2005;25(21):
72. Bronte V, Serafini P, De Santo C, et al. IL-4-induced 89. Gangadhar T. Epacadostat plus pembrolizumab in 9543–9553.
arginase 1 suppresses alloreactive T cells in tumor- patients with advanced melanoma and select solid 108. Riley JL, Mao M, Kobayashi S, et al. Modulation
bearing mice. J Immunol. 2003;170(1):270–278. tumors: updated phase 1 results from ECHO-202 of TCR-induced transcriptional profiles by ligation
73. Mazzoni A, Bronte V, Visintin A, et al. Myeloid / KEYNOTE-037. Ann Oncol. 2016;27(suppl 6). of CD28, ICOS, and CTLA-4 receptors. Proc Natl
suppressor lines inhibit T cell responses by 90. Blobe GC, Schiemann WP, Lodish HF. Role of Acad Sci USA. 2002;99(18):11790–11795.
an NO-dependent mechanism. J Immunol. transforming growth factor beta in human disease. 109. Schneider H, Downey J, Smith A, et al. Reversal
2002;168(2):689–695. N Engl J Med. 2000;342(18):1350–1358. of the TCR stop signal by CTLA-4. Science.
74. Zea AH, Rodriguez PC, Atkins MB, et al. Arginase- 91. Pepper MS. Transforming growth factor-beta: 2006;313(5795):1972–1975.
producing myeloid suppressor cells in renal cell vasculogenesis, angiogenesis, and vessel wall integrity. 110. Schneider H, Mandelbrot DA, Greenwald RJ,
carcinoma patients: a mechanism of tumor evasion. Cytokine Growth Factor Rev. 1997;8(1):21–43. et al. Cutting edge: CTLA-4 (CD152) differen-
Cancer Res. 2005;65(8):3044–3048. 92. Shariat SF, Kim JH, Andrews B, et al. Preoperative tially regulates mitogen-activated protein kinases
75. Moisan F, Francisco EB, Brozovic A, et al. Enhance- plasma levels of transforming growth factor beta(1) (extracellular signal-regulated kinase and c-Jun
ment of paclitaxel and carboplatin therapies by CCL2 strongly predict clinical outcome in patients with N-terminal kinase) in CD4+ T cells from receptor/
blockade in ovarian cancers. Mol Oncol. 2014;8(7): bladder carcinoma. Cancer. 2001;92(12):2985– ligand-deficient mice. J Immunol. 2002;169(7):3475–
1231–1239. 2992. 3479.
Cancer Immunology  •  CHAPTER 6 96.e3

111. Qureshi OS, Zheng Y, Nakamura K, et al. Trans- blocking the TCR-induced stop signal. Nat Immunol. carcinoma foster immune privilege and disease pro-
endocytosis of CD80 and CD86: a molecular basis 2009;10(11):1185–1192. gression through PD-L1. J Exp Med. 2009;206(6):
for the cell-extrinsic function of CTLA-4. Science. 131. Ahmadzadeh M, Johnson LA, Heemskerk B, et al. 1327–1337.
2011;332(6029):600–603. Tumor antigen-specific CD8 T cells infiltrating the 151. Liu Y, Zeng B, Zhang Z, Zhang Y, Yang R. B7-H1
112. Tivol EA, Borriello F, Schweitzer AN, Lynch WP, tumor express high levels of PD-1 and are function- on myeloid-derived suppressor cells in immune
Bluestone JA, Sharpe AH. Loss of CTLA-4 leads to ally impaired. Blood. 2009;114(8):1537–1544. suppression by a mouse model of ovarian cancer.
massive lymphoproliferation and fatal multiorgan 132. Sfanos KS, Bruno TC, Meeker AK, De Marzo AM, Clin Immunol. 2008;129(3):471–481.
tissue destruction, revealing a critical negative regula- Isaacs WB, Drake CG. Human prostate-infiltrating 152. Hino R, Kabashima K, Kato Y, et al. Tumor cell
tory role of CTLA-4. Immunity. 1995;3(5):541– CD8+ T lymphocytes are oligoclonal and PD-1. expression of programmed cell death-1 ligand 1 is a
547. Prostate. 2009;69(15):1694–1703. prognostic factor for malignant melanoma. Cancer.
113. Waterhouse P, Penninger JM, Timms E, et al. 133. Francisco LM, Salinas VH, Brown KE, et al. PD- 2010;116(7):1757–1766.
Lymphoproliferative disorders with early lethality L1 regulates the development, maintenance, and 153. Steidl C, Shah SP, Woolcock BW, et al. MHC class
in mice deficient in Ctla-4. Science. 1995;270(5238): function of induced regulatory T cells. J Exp Med. II transactivator CIITA is a recurrent gene fusion
985–988. 2009;206(13):3015–3029. partner in lymphoid cancers. Nature. 2011;471
114. Peggs KS, Quezada SA, Chambers CA, Korman AJ, 134. Fanoni D, Tavecchio S, Recalcati S, et al. New mono- (7338):377–381.
Allison JP. Blockade of CTLA-4 on both effector clonal antibodies against B-cell antigens: possible 154. Parsa AT, Waldron JS, Panner A, et al. Loss of
and regulatory T cell compartments contributes to new strategies for diagnosis of primary cutaneous tumor suppressor PTEN function increases B7-H1
the antitumor activity of anti-CTLA-4 antibodies. B-cell lymphomas. Immunol Lett. 2011;134(2):157– expression and immunoresistance in glioma. Nat
J Exp Med. 2009;206(8):1717–1725. 160. Med. 2007;13(1):84–88.
115. Wing K, Onishi Y, Prieto-Martin P, et al. CTLA-4 135. Terme M, Ullrich E, Aymeric L, et al. IL-18 induces 155. Marzec M, Zhang Q, Goradia A, et al. Oncogenic
control over Foxp3+ regulatory T cell function. PD-1-dependent immunosuppression in cancer. kinase NPM/ALK induces through STAT3 expres-
Science. 2008;322(5899):271–275. Cancer Res. 2011;71(16):5393–5399. sion of immunosuppressive protein CD274 (PD-L1,
116. Fontenot JD, Gavin MA, Rudensky AY. Foxp3 136. Velu V, Titanji K, Zhu B, et al. Enhancing SIV- B7-H1). Proc Natl Acad Sci USA. 2008;105(52):
programs the development and function of specific immunity in vivo by PD-1 blockade. Nature. 20852–20857.
CD4+CD25+ regulatory T cells. Nat Immunol. 2009;458(7235):206–210. 156. Yi KH, Chen L. Fine tuning the immune response
2003;4(4):330–336. 137. Huang X, Venet F, Wang YL, et al. PD-1 expression through B7-H3 and B7-H4. Immunol Rev.
117. Hori S, Nomura T, Sakaguchi S. Control of regula- by macrophages plays a pathologic role in altering 2009;229(1):145–151.
tory T cell development by the transcription factor microbial clearance and the innate inflamma- 157. Ngiow SF, von Scheidt B, Akiba H, Yagita H, Teng
Foxp3. Science. 2003;299(5609):1057–1061. tory response to sepsis. Proc Natl Acad Sci USA. MW, Smyth MJ. Anti-TIM3 antibody promotes T
118. Gavin MA, Rasmussen JP, Fontenot JD, et al. 2009;106(15):6303–6308. cell IFN-gamma-mediated antitumor immunity and
Foxp3-dependent programme of regulatory T- 138. Dong H, Zhu G, Tamada K, Chen L. B7-H1, a suppresses established tumors. Cancer Res. 2011;71
cell differentiation. Nature. 2007;445(7129):771– third member of the B7 family, co-stimulates T-cell (10):3540–3551.
775. proliferation and interleukin-10 secretion. Nat Med. 158. Triebel F, Jitsukawa S, Baixeras E, et al. LAG-3, a
119. Hill JA, Feuerer M, Tash K, et  al. Foxp3 1999;5(12):1365–1369. novel lymphocyte activation gene closely related to
transcription-factor-dependent and -independent 139. Latchman Y, Wood CR, Chernova T, et al. PD-L2 CD4. J Exp Med. 1990;171(5):1393–1405.
regulation of the regulatory T cell transcriptional is a second ligand for PD-1 and inhibits T cell 159. Goldberg MV, Drake CG. LAG-3 in Cancer
signature. Immunity. 2007;27(5):786–800. activation. Nat Immunol. 2001;2(3):261–268. Immunotherapy. Curr Top Microbiol Immunol.
120. Freeman GJ, Long AJ, Iwai Y, et al. Engagement of 140. Tseng SY, Otsuji M, Gorski K, et al. B7-DC, a new 2011;344:269–278.
the PD-1 immunoinhibitory receptor by a novel B7 dendritic cell molecule with potent costimulatory 160. Huang CT, Workman CJ, Flies D, et al. Role of
family member leads to negative regulation of lym- properties for T cells. J Exp Med. 2001;193(7): LAG-3 in regulatory T cells. Immunity. 2004;21(4):
phocyte activation. J Exp Med. 2000;192(7):1027– 839–846. 503–513.
1034. 141. Butte MJ, Keir ME, Phamduy TB, Sharpe AH, 161. Grosso JF, Kelleher CC, Harris TJ, et al. LAG-3
121. Ishida Y, Agata Y, Shibahara K, Honjo T. Induced Freeman GJ. Programmed death-1 ligand 1 interacts regulates CD8+ T cell accumulation and
expression of PD-1, a novel member of the immu- specifically with the B7-1 costimulatory molecule effector function in murine self- and tumor-
noglobulin gene superfamily, upon programmed cell to inhibit T cell responses. Immunity. 2007;27(1): tolerance systems. J Clin Invest. 2007;117(11):3383–
death. EMBO J. 1992;11(11):3887–3895. 111–122. 3392.
122. Keir ME, Butte MJ, Freeman GJ, Sharpe AH. PD-1 142. Park JJ, Omiya R, Matsumura Y, et al. B7-H1/ 162. Xu F, Liu J, Liu D, et al. LSECtin expressed on
and its ligands in tolerance and immunity. Annu CD80 interaction is required for the induction and melanoma cells promotes tumor progression by
Rev Immunol. 2008;26:677–704. maintenance of peripheral T-cell tolerance. Blood. inhibiting antitumor T-cell responses. Cancer Res.
123. Keir ME, Liang SC, Guleria I, et al. Tissue expression 2010;116(8):1291–1298. 2014;74(13):3418–3428.
of PD-L1 mediates peripheral T cell tolerance. J Exp 143. Paterson AM, Brown KE, Keir ME, et al. The 163. Blackburn SD, Shin H, Haining WN, et al.
Med. 2006;203(4):883–895. programmed death-1 ligand 1:B7-1 pathway restrains Coregulation of CD8+ T cell exhaustion by multiple
124. Nishimura H, Nose M, Hiai H, Minato N, Honjo diabetogenic effector T cells in vivo. J Immunol. inhibitory receptors during chronic viral infection.
T. Development of lupus-like autoimmune diseases 2011;187(3):1097–1105. Nat Immunol. 2009;10(1):29–37.
by disruption of the PD-1 gene encoding an 144. Dong H, Strome SE, Matteson EL, et  al. 164. Grosso JF, Goldberg MV, Getnet D, et al. Function-
ITIM motif-carrying immunoreceptor. Immunity. Costimulating aberrant T cell responses by B7-H1 ally distinct LAG-3 and PD-1 subsets on activated
1999;11(2):141–151. autoantibodies in rheumatoid arthritis. J Clin Invest. and chronically stimulated CD8 T cells. J Immunol.
125. Nishimura H, Okazaki T, Tanaka Y, et  al. 2003;111(3):363–370. 2009;182(11):6659–6669.
Autoimmune dilated cardiomyopathy in PD-1 145. Azuma T, Yao S, Zhu G, Flies AS, Flies SJ, Chen 165. Santiago C, Ballesteros A, Tami C, Martinez-Munoz
receptor-deficient mice. Science. 2001;291(5502): L. B7-H1 is a ubiquitous antiapoptotic receptor on L, Kaplan GG, Casasnovas JM. Structures of T Cell
319–322. cancer cells. Blood. 2008;111(7):3635–3643. immunoglobulin mucin receptors 1 and 2 reveal
126. Okazaki T, Honjo T. PD-1 and PD-1 ligands: 146. Zou W, Chen L. Inhibitory B7-family molecules in mechanisms for regulation of immune responses
from discovery to clinical application. Int Immunol. the tumour microenvironment. Nat Rev Immunol. by the TIM receptor family. Immunity. 2007;26(3):
2007;19(7):813–824. 2008;8(6):467–477. 299–310.
127. Blank C, Brown I, Peterson AC, et al. PD-L1/B7H-1 147. Brown JA, Dorfman DM, Ma FR, et al. Blockade 166. Chiba S, Baghdadi M, Akiba H, et al. Tumor-
inhibits the effector phase of tumor rejection by T of programmed death-1 ligands on dendritic cells infiltrating DCs suppress nucleic acid-mediated
cell receptor (TCR) transgenic CD8+ T cells. Cancer enhances T cell activation and cytokine production. innate immune responses through interactions
Res. 2004;64(3):1140–1145. J Immunol. 2003;170(3):1257–1266. between the receptor TIM-3 and the alarmin
128. Dong H, Strome SE, Salomao DR, et al. Tumor- 148. Konishi J, Yamazaki K, Azuma M, Kinoshita I, HMGB1. Nat Immunol. 2012;13(9):832–842.
associated B7-H1 promotes T-cell apoptosis: a Dosaka-Akita H, Nishimura M. B7-H1 expression 167. Huang YH, Zhu C, Kondo Y, et al. CEACAM1
potential mechanism of immune evasion. Nat Med. on non-small cell lung cancer cells and its relation- regulates TIM-3-mediated tolerance and exhaus-
2002;8(8):793–800. ship with tumor-infiltrating lymphocytes and their tion. Nature. 2015;517(7534):386–390.
129. Taube JMAR, Xu H, Sharma R, McMiller T, PD-1 expression. Clin Cancer Res. 2004;10(15): 168. Zhu C, Anderson AC, Schubart A, et al. The Tim-3
Klein A, Pardoll DM, et al. B7-H1 expression 5094–5100. ligand galectin-9 negatively regulates T helper type
co-localizes with inflammatory infiltrates in benign 149. Curiel TJ, Wei S, Dong H, et al. Blockade of B7-H1 1 immunity. Nat Immunol. 2005;6(12):1245–
and malignant melanocytic lesions: implications for improves myeloid dendritic cell-mediated antitumor 1252.
immunotherapy. Sci Transl Med. 2012;in press. immunity. Nat Med. 2003;9(5):562–567. 169. Baitsch L, Legat A, Barba L, et al. Extended
130. Fife BT, Pauken KE, Eagar TN, et al. Interactions 150. Kuang DM, Zhao Q, Peng C, et al. Activated co-expression of inhibitory receptors by human
between PD-1 and PD-L1 promote tolerance by monocytes in peritumoral stroma of hepatocellular CD8 T-cells depending on differentiation,
96.e4 Part I: Science and Clinical Oncology

antigen-specificity and anatomical localization. patients with advanced cancer. N Engl J Med. 211. Vetizou M, Pitt JM, Daillere R, et al. Anticancer
PLoS ONE. 2012;7(2):e30852. 2012;366(26):2455–2465. immunotherapy by CTLA-4 blockade relies on the
170. Fourcade J, Sun Z, Benallaoua M, et  al. 190. Topalian SL, Hodi FS, Brahmer JR, et al. Safety, gut microbiota. Science. 2015;350(6264):1079–1084.
Upregulation of Tim-3 and PD-1 expression is activity, and immune correlates of anti-PD-1 antibody 212. Ebert PJ, Cheung J, Yang Y, et al. MAP kinase
associated with tumor antigen-specific CD8+ T in cancer. N Engl J Med. 2012;366(26):2443–2454. inhibition promotes T cell and anti-tumor activity
cell dysfunction in melanoma patients. J Exp Med. 191. Hoos A, Eggermont AM, Janetzki S, et al. Improved in combination with PD-L1 checkpoint blockade.
2010;207(10):2175–2186. endpoints for cancer immunotherapy trials. J Natl Immunity. 2016;44(3):609–621.
171. Sakuishi K, Apetoh L, Sullivan JM, Blazar BR, Cancer Inst. 2010;102(18):1388–1397. 213. Muller P, Kreuzaler M, Khan T, et al. Trastuzumab
Kuchroo VK, Anderson AC. Targeting Tim-3 192. Kohrt HE, Tumeh PC, Benson D, et al. Immuno- emtansine (T-DM1) renders HER2+ breast cancer
and PD-1 pathways to reverse T cell exhaustion dynamics: a cancer immunotherapy trials network highly susceptible to CTLA-4/PD-1 blockade. Sci
and restore anti-tumor immunity. J Exp Med. review of immune monitoring in immuno-oncology Transl Med. 2015;7(315):315ra188.
2010;207(10):2187–2194. clinical trials. J Immunother Cancer. 2016;4:15. 214. De Henau O, Rausch M, Winkler D, et al. Over-
172. Koyama S, Akbay EA, Li YY, et al. Adaptive resistance 193. Brahmer JR, Tykodi SS, Chow LQ, et  al. coming resistance to checkpoint blockade therapy
to therapeutic PD-1 blockade is associated with Safety and activity of anti-PD-L1 antibody in by targeting PI3Kgamma in myeloid cells. Nature.
upregulation of alternative immune checkpoints. patients with advanced cancer. N Engl J Med. 2016;539(7629):443–447.
Nat Commun. 2016;7:10501. 2012;366(26):2455–2465. 215. Wu R, Forget MA, Chacon J, et al. Adoptive
173. Zarek PE, Huang CT, Lutz ER, et al. A2A receptor 194. Topalian SL, Hodi FS, Brahmer JR, et al. Safety, T-cell therapy using autologous tumor-infiltrating
signaling promotes peripheral tolerance by inducing activity, and immune correlates of anti-PD-1 antibody lymphocytes for metastatic melanoma: current status
T-cell anergy and the generation of adaptive regula- in cancer. N Engl J Med. 2012;366(26):2443–2454. and future outlook. Cancer J. 2012;18(2):160–175.
tory T cells. Blood. 2008;111(1):251–259. 195. Garon EB, Rizvi NA, Hui R, et al. Pembrolizumab 216. Sadelain M, Riviere I, Brentjens R. Targeting tumours
174. Deaglio S, Dwyer KM, Gao W, et al. Adenosine for the treatment of non-small-cell lung cancer. with genetically enhanced T lymphocytes. Nat Rev
generation catalyzed by CD39 and CD73 expressed N Engl J Med. 2015;372(21):2018–2028. Cancer. 2003;3(1):35–45.
on regulatory T cells mediates immune suppression. 196. Taube JM, Klein A, Brahmer JR, et al. Association 217. Thomas S, Hart DP, Xue SA, Cesco-Gaspere M,
J Exp Med. 2007;204(6):1257–1265. of PD-1, PD-1 ligands, and other features of the Stauss HJ. T-cell receptor gene therapy for cancer:
175. Waickman AT, Alme A, Senaldi L, Zarek PE, Horton tumor immune microenvironment with response to the progress to date and future objectives. Expert
M, Powell JD. Enhancement of tumor immuno- anti-PD-1 therapy. Clin Cancer Res. 2014;20(19): Opin Biol Ther. 2007;7(8):1207–1218.
therapy by deletion of the A2A adenosine receptor. 5064–5074. 218. Kebriaei P, Singh H, Huls MH, et al. Phase I trials
Cancer Immunol Immunother. 2012;61(6):917–926. 197. Herbst RS, Soria JC, Kowanetz M, et al. Predic- using Sleeping Beauty to generate CD19-specific
176. Hodi FS, O’Day SJ, McDermott DF, et al. Improved tive correlates of response to the anti-PD-L1 CAR T cells. J Clin Invest. 2016;126(9):3363–3376.
survival with ipilimumab in patients with metastatic antibody MPDL3280A in cancer patients. Nature. 219. Singh H, Huls H, Kebriaei P, Cooper LJ. A new
melanoma. N Engl J Med. 2010;363(8):711–723. 2014;515(7528):563–567. approach to gene therapy using Sleeping Beauty to
177. Reck M, Rodriguez-Abreu D, Robinson AG, 198. Wolchok JD, Kluger H, Callahan MK, et al. genetically modify clinical-grade T cells to target
et al. Pembrolizumab versus chemotherapy for Nivolumab plus ipilimumab in advanced melanoma. CD19. Immunol Rev. 2014;257(1):181–190.
PD-L1-positive non-small-cell lung cancer. N Engl N Engl J Med. 2013;369(2):122–133. 220. Smith TT, Stephan SB, Moffett HF, et al. In situ
J Med. 2016;375(19):1823–1833. 199. Postow MA, Chesney J, Pavlick AC, et al. Nivolumab programming of leukaemia-specific T cells using
178. Robert C, Schachter J, Long GV, et al. Pembroli- and ipilimumab versus ipilimumab in untreated synthetic DNA nanocarriers. Nat Nanotechnol. Apr
zumab versus ipilimumab in advanced melanoma. melanoma. N Engl J Med. 2015;372(21):2006– 17 2017.
N Engl J Med. 2015;372(26):2521–2532. 2017. 221. Schutsky K, Song DG, Lynn R, et al. Rigorous
179. Motzer RJ, Escudier B, McDermott DF, et al. 200. Gubin MM, Zhang X, Schuster H, et  al. optimization and validation of potent RNA CAR
Nivolumab versus everolimus in advanced renal-cell Checkpoint blockade cancer immunotherapy T cell therapy for the treatment of common epithelial
carcinoma. N Engl J Med. 2015;373(19):1803–1813. targets tumour-specific mutant antigens. Nature. cancers expressing folate receptor. Oncotarget.
180. Rosenberg JE, Hoffman-Censits J, Powles T, et al. 2014;515(7528):577–581. 2015;6(30):28911–28928.
Atezolizumab in patients with locally advanced 201. Rizvi NA, Hellmann MD, Snyder A, et al. Cancer 222. Sather BD, Romano Ibarra GS, Sommer K,
and metastatic urothelial carcinoma who have immunology. Mutational landscape determines et al. Efficient modification of CCR5 in primary
progressed following treatment with platinum-based sensitivity to PD-1 blockade in non-small cell lung human hematopoietic cells using a megaTAL
chemotherapy: a single-arm, multicentre, phase 2 cancer. Science. 2015;348(6230):124–128. nuclease and AAV donor template. Sci Transl Med.
trial. Lancet. 2016;387(10031):1909–1920. 202. Snyder A, Makarov V, Merghoub T, et al. Genetic 2015;7(307):307ra156.
181. Ferris RL, Blumenschein G Jr, Fayette J, basis for clinical response to CTLA-4 blockade in 223. Ren J, Liu X, Fang C, Jiang S, June CH, Zhao Y.
et al. Nivolumab for recurrent squamous-cell melanoma. N Engl J Med. 2014;371(23):2189–2199. Multiplex genome editing to generate universal CAR
carcinoma of the head and neck. N Engl J Med. 203. McGranahan N, Furness AJ, Rosenthal R, et al. T cells resistant to PD1 inhibition. Clin Cancer Res.
2016;375(19):1856–1867. Clonal neoantigens elicit T cell immunoreactivity Nov 04 2016.
182. Le DT, Uram JN, Wang H, et al. PD-1 Blockade and sensitivity to immune checkpoint blockade. 224. Scholler J, Brady TL, Binder-Scholl G, et al. Decade-
in tumors with mismatch-repair deficiency. N Engl Science. 2016;351(6280):1463–1469. long safety and function of retroviral-modified
J Med. 2015;372(26):2509–2520. 204. Tumeh PC, Harview CL, Yearley JH, et al. PD-1 chimeric antigen receptor T cells. Sci Transl Med.
183. Nghiem PT, Bhatia S, Lipson EJ, et al. PD-1 Block- blockade induces responses by inhibiting adaptive 2012;4(132):132ra153.
ade with pembrolizumab in advanced Merkel-cell immune resistance. Nature. 2014;515(7528): 225. Han A, Glanville J, Hansmann L, Davis MM.
carcinoma. N Engl J Med. 2016;374(26):2542–2552. 568–571. Linking T-cell receptor sequence to functional
184. Phan GQ, Yang JC, Sherry RM, et al. Cancer 205. Cha E, Klinger M, Hou Y, et al. Improved survival phenotype at the single-cell level. Nat Biotechnol.
regression and autoimmunity induced by cytotoxic T with T cell clonotype stability after anti-CTLA-4 treat- 2014;32(7):684–692.
lymphocyte-associated antigen 4 blockade in patients ment in cancer patients. Sci Transl Med. 2014;6(238): 226. Schmitt TM, Ragnarsson GB, Greenberg PD. T
with metastatic melanoma. Proc Natl Acad Sci USA. 238ra270. cell receptor gene therapy for cancer. Hum Gene
2003;100(14):8372–8377. 206. Huang AC, Postow MA, Orlowski RJ, et al. T-cell Ther. 2009;20(11):1240–1248.
185. Hodi FS, O’Day SJ, McDermott DF, et al. Improved invigoration to tumour burden ratio associated with 227. Zhao Y, Bennett AD, Zheng Z, et al. High-affinity
survival with ipilimumab in patients with metastatic anti-PD-1 response. Nature. Apr 10 2017. TCRs generated by phage display provide CD4+
melanoma. N Engl J Med. 2010;363(8):711–723. 207. Peng D, Kryczek I, Nagarsheth N, et al. Epigen- T cells with the ability to recognize and kill tumor
186. Robert C, Thomas L, Bondarenko I, et al. Ipilimumab etic silencing of TH1-type chemokines shapes cell lines. J Immunol. 2007;179(9):5845–5854.
plus dacarbazine for previously untreated metastatic tumour immunity and immunotherapy. Nature. 228. Varela-Rohena A, Molloy PE, Dunn SM, et al.
melanoma. N Engl J Med. 2011;364(26):2517–2526. 2015;527(7577):249–253. Control of HIV-1 immune escape by CD8 T cells
187. Eggermont AM, Chiarion-Sileni V, Grob JJ, 208. Spranger S, Bao R, Gajewski TF. Melanoma-intrinsic expressing enhanced T-cell receptor. Nat Med.
et al. Prolonged survival in stage III melanoma beta-catenin signalling prevents anti-tumour immu- 2008;14(12):1390–1395.
with ipilimumab adjuvant therapy. N Engl J Med. nity. Nature. 2015;523(7559):231–235. 229. Amir AL, van der Steen DM, van Loenen MM,
2016;375(19):1845–1855. 209. Hugo W, Zaretsky JM, Sun L, et al. Genomic et al. PRAME-specific Allo-HLA-restricted T
188. Tarhini AA, Edington H, Butterfield LH, et al. and transcriptomic features of response to anti- cells with potent antitumor reactivity useful for
Immune monitoring of the circulation and the PD-1 therapy in metastatic melanoma. Cell. therapeutic T-cell receptor gene transfer. Clin Cancer
tumor microenvironment in patients with region- 2016;165(1):35–44. Res. 2011;17(17):5615–5625.
ally advanced melanoma receiving neoadjuvant 210. Sivan A, Corrales L, Hubert N, et al. Commensal Bifi- 230. Morgan RA, Dudley ME, Wunderlich JR, et al. Cancer
ipilimumab. PLoS ONE. 2014;9(2):e87705. dobacterium promotes antitumor immunity and facil- regression in patients after transfer of genetically
189. Brahmer JR, Tykodi SS, Chow LQ, et  al. itates anti-PD-L1 efficacy. Science. 2015;350(6264): engineered lymphocytes. Science. 2006;314(5796):
Safety and activity of anti-PD-L1 antibody in 1084–1089. 126–129.
Cancer Immunology  •  CHAPTER 6 96.e5

231. Robbins PF, Morgan RA, Feldman SA, et al. Tumor 243. Brentjens RJ, Curran KJ. Novel cellular therapies inherent leukaemic plasticity. Nat Commun. 2016;7:
regression in patients with metastatic synovial cell for leukemia: CAR-modified T cells targeted to the 12320.
sarcoma and melanoma using genetically engineered CD19 antigen. Hematology Am Soc Hematol Educ 255. Bonifant CL, Jackson HJ, Brentjens RJ, Curran KJ.
lymphocytes reactive with NY-ESO-1. J Clin Oncol. Program. 2012;2012:143–151. Toxicity and management in CAR T-cell therapy.
2011;29(7):917–924. 244. Kochenderfer JN, Wilson WH, Janik JE, et al. Mol Ther Oncolytics. 2016;3:16011.
232. Robbins PF, Kassim SH, Tran TL, et al. A pilot Eradication of B-lineage cells and regression of 256. Lee DW, Gardner R, Porter DL, et al. Current con-
trial using lymphocytes genetically engineered with lymphoma in a patient treated with autologous cepts in the diagnosis and management of cytokine
an NY-ESO-1-reactive T-cell receptor: long-term T cells genetically engineered to recognize CD19. release syndrome. Blood. 2014;124(2):188–195.
follow-up and correlates with response. Clin Cancer Blood. 2010;116(20):4099–4102. 257. Davila ML, Riviere I, Wang X, et al. Efficacy and
Res. 2015;21(5):1019–1027. 245. Porter DL, Levine BL, Kalos M, Bagg A, June toxicity management of 19-28z CAR T cell therapy
233. Ding K, Wang XM, Fu R, Ruan EB, Liu H, Shao CH. Chimeric antigen receptor-modified T cells in B cell acute lymphoblastic leukemia. Sci Transl
ZH. PRAME gene expression in acute leukemia in chronic lymphoid leukemia. N Engl J Med. Med. 2014;6(224):224ra225.
and its clinical significance. Cancer Biol Med. 2011;365(8):725–733. 258. Brudno JN, Kochenderfer JN. Toxicities of chimeric
2012;9(1):73–76. 246. Kalos M, Levine BL, Porter DL, et al. T cells antigen receptor T cells: recognition and manage-
234. Goswami M, Hensel N, Smith BD, et al. Expres- with chimeric antigen receptors have potent ment. Blood. 2016;127(26):3321–3330.
sion of putative targets of immunotherapy in acute antitumor effects and can establish memory in 259. Taraseviciute A, Kean L, Jensen M. Creation of
myeloid leukemia and healthy tissues. Leukemia. patients with advanced leukemia. Sci Transl Med. the first non-human primate model that faithfully
2014;28(5):1167–1170. 2011;3(95):95ra73. recapitulates chimeric antigen receptor (CAR) T
235. Morgan RA, Chinnasamy N, Abate-Daga D, et al. 247. Porter DL, Kalos M, Zheng Z, Levine B, June cell-mediated cytokine release syndrome (CRS) and
Cancer regression and neurological toxicity following C. Chimeric antigen receptor therapy for B-cell neurologic toxicity following B cell-directed CAR-T
anti-MAGE-A3 TCR gene therapy. J Immunother. malignancies. J Cancer. 2011;2:331–332. cell therapy. paper presented at: Blood 2016.
2013;36(2):133–151. 248. Brentjens RJ, Riviere I, Park JH, et al. Safety and 260. Newick K, Moon E, Albelda SM. Chimeric antigen
236. Cameron BJ, Gerry AB, Dukes J, et al. Identifica- persistence of adoptively transferred autologous receptor T-cell therapy for solid tumors. Mol Ther
tion of a Titin-derived HLA-A1-presented peptide CD19-targeted T cells in patients with relapsed or Oncolytics. 2016;3:16006.
as a cross-reactive target for engineered MAGE chemotherapy refractory B-cell leukemias. Blood. 261. Pule MA, Savoldo B, Myers GD, et al. Virus-
A3-directed T cells. Sci Transl Med. 2013;5(197): 2011;118(18):4817–4828. specific T cells engineered to coexpress tumor-
197ra103. 249. Grupp SA, Kalos M, Barrett D, et al. Chimeric specific receptors: persistence and antitumor
237. Linette GP, Stadtmauer EA, Maus MV, et al. antigen receptor-modified T cells for acute lymphoid activity in individuals with neuroblastoma. Nat
Cardiovascular toxicity and titin cross-reactivity of leukemia. N Engl J Med. 2013;368(16):1509– Med. 2008;14(11):1264–1270.
affinity-enhanced T cells in myeloma and melanoma. 1518. 262. Louis CU, Savoldo B, Dotti G, et al. Antitumor
Blood. 2013;122(6):863–871. 250. Lee DW, Kochenderfer JN, Stetler-Stevenson M, activity and long-term fate of chimeric antigen
238. Brentjens RJ, Santos E, Nikhamin Y, et al. et al. T cells expressing CD19 chimeric antigen receptor-positive T cells in patients with neuro-
Genetically targeted T cells eradicate systemic acute receptors for acute lymphoblastic leukaemia in blastoma. Blood. 2011;118(23):6050–6056.
lymphoblastic leukemia xenografts. Clin Cancer Res. children and young adults: a phase 1 dose-escalation 263. Savoldo B, Ramos CA, Liu E, et al. CD28 costimula-
2007;13(18 Pt 1):5426–5435. trial. Lancet. 2015;385(9967):517–528. tion improves expansion and persistence of chimeric
239. Long AH, Haso WM, Shern JF, et al. 4-1BB 251. Davila ML, Sauter C, Brentjens R. CD19-targeted T antigen receptor-modified T cells in lymphoma
costimulation ameliorates T cell exhaustion induced cells for hematologic malignancies: clinical experience patients. J Clin Invest. 2011;121(5):1822–1826.
by tonic signaling of chimeric antigen receptors. Nat to date. Cancer J. 2015;21(6):470–474. 264. Ahmed N, Brawley VS, Hegde M, et al. Human
Med. 2015;21(6):581–590. 252. Ghoneim HE, Zamora AE, Thomas PG, Youngblood epidermal growth factor receptor 2 (HER2)–specific
240. Maude SL, Frey N, Shaw PA, et al. Chimeric antigen BA. Cell-intrinsic barriers of T cell-based immuno- chimeric antigen receptor-modified T cells for the
receptor T cells for sustained remissions in leukemia. therapy. Trends Mol Med. Nov 4 2016. immunotherapy of HER2-positive sarcoma. J Clin
N Engl J Med. 2014;371(16):1507–1517. 253. Sotillo E, Barrett DM, Black KL, et al. Convergence Oncol. 2015;33(15):1688–1696.
241. Sadelain M, Brentjens R, Riviere I. The basic of acquired mutations and alternative splicing of 265. Brown CE, Alizadeh D, Starr R, et al. Regression of
principles of chimeric antigen receptor design. CD19 enables resistance to CART-19 immuno- glioblastoma after chimeric antigen receptor T-cell
Cancer Discov. 2013;3(4):388–398. therapy. Cancer Discov. 2015;5(12):1282–1295. therapy. N Engl J Med. 2016;375(26):2561–2569.
242. Pegram HJ, Park JH, Brentjens RJ. CD28z CARs 254. Jacoby E, Nguyen SM, Fountaine TJ, et al. CD19 266. Kantoff PW, Higano CS, Shore ND, et al. Sipuleucel-
and armored CARs. Cancer J. 2014;20(2):127– CAR immune pressure induces B-precursor acute T immunotherapy for castration-resistant prostate
133. lymphoblastic leukaemia lineage switch exposing cancer. N Engl J Med. 2010;363(5):411–422.
Stem Cells, Cell Differentiation, and Cancer 7 
Piero Dalerba, Maximilian Diehn, Irving L. Weissman, and Michael F. Clarke

S UMMARY OF K EY P OI NT S
• Many tumors originate in organs and • In many tissues, stem cells are the leukemias, brain gliomas, breast,
tissues that undergo a continuous only long-lived cell population. This colon, head and neck, bladder, and
process of cell turnover, which is observation suggests that early prostate carcinomas), only a specific,
sustained by a minority population of transforming events, either genetic phenotypically distinct, subset of
stem cells (e.g., the colon, breast, mutations or epigenetic cancer cells is able to form tumors
lung, prostate, brain, and bone modifications, are likely to when serially transplanted in mice.
marrow). accumulate in stem cells. • To result in cure, therapies must
• Stem cells have four fundamental • In addition to oncogenes that control eradicate self-renewing cancer cells
properties: the ability to give rise to cell survival and proliferation, there is (cancer stem cells). The ability to
new stem cells with intact and a class of oncogenes that regulates identify these cells should allow the
unlimited growth potential self-renewal. In some cancers, tumor identification of new diagnostic
(self-renewal), the ability to give rise growth might be sustained by markers and therapeutic targets.
to a progeny of specialized cells progenitor cells, which naturally do • Studies on the gene-expression
(differentiation), the ability to migrate not self-renew but have aberrantly profile of cancer stem cell
into new tissue locations and acquired this ability during disease populations helped identify tumor
establish tissue growth (migration progression, as a result of mutations subtypes with differential response
and tissue repair), and the ability to that either activate genes required to antitumor drugs, providing first
balance the previous three properties for self-renewal, or inactivate genes evidence for the clinical usefulness
according to a genetic program that that disable it. of cancer stem cell research,
places constraints on their numeric • Experimental data suggest that in especially in guiding treatment
expansion (homeostatic control). many forms of human cancer (e.g., decisions.

Many tumors originate from tissues that naturally undergo a continuous the only long-lived cells of normal adult tissues, they also represent
process of cell turnover. In such tissues, cell maturation is arranged the most likely target population in which transforming events, either
according to a hierarchic system, in which a minority population of genetic mutations or epigenetic modifications, progressively accumulate,
stem cells is able to perpetuate itself through a process called self-renewal at least in the initial phases of the disease.2 Third, increasing evidence
while also giving rise to several stages of intermediate progenitors and, suggests that many tumor tissues, akin to normal ones, contain a
eventually, to terminally differentiated cells (Fig. 7.1). As opposed to minority “cancer stem cell” population with extensive proliferative
stem cells, intermediate progenitors and terminally differentiated cells potential, which drives tumor growth and metastasis, and which can
have a limited expansion potential and are unable to self-renew.1,2 undergo differentiation, sustaining the formation of a heterogeneous
Because stem cells are frequently found in small numbers, they must progeny of specialized cell types.18–21 Finally, many physiologic properties
be isolated and tested prospectively to define their molecular and of stem cells are mirrored in cancer cells and contribute to define
biochemical properties. Among the stem cell populations that have their behavior. For example, because stem cells are long-lived and
been best characterized are those that give rise to the lymphohema- necessary to maintain tissue homeostasis over the lifetime of an organ-
topoietic system, known as hematopoietic stem cells (HSCs), which ism, they are frequently endowed with high levels of enzymatic systems
have been purified from both mice and humans.3–7 HSCs have that protect them from environmental hazards. These very same
important applications in cancer therapy, especially in clinical settings enzymatic systems endow specific subsets of cancer cells with resistance
where bone marrow transplantation is used to regenerate the hema- to antitumor agents and might contribute to treatment failure.
topoietic system after myeloablative treatments.6 The ability to isolate
HSCs with high degrees of purity enables the execution of tumor-free PROPERTIES OF NORMAL STEM CELLS
autologous bone-marrow transplants in cancer patients.8,9
Understanding the biology of the normal tissues from which tumors HSCs are the most studied and best understood type of adult stem
originate, and especially of their stem cell populations, can provide cells and serve as a model for stem cells in other tissues.22,23 Hema-
important insights into cancer biology. Several aspects of stem cell topoiesis is a tightly regulated process in which a small pool of HSCs
biology are relevant to cancer.10,11 First, many cancer cells share with give rise to an increasingly diversified population of oligolineage
normal stem cells the capacity to self-renew, and emerging evidence intermediates, which in turn form the full repertoire of mature elements
suggests that similar molecular pathways underpin this property in of the lymphohematopoietic system (e.g., erythrocytes, platelets,
the two cell types.12–17 Second, because stem cells are long-lived, often granulocytes, macrophages, and lymphocytes; see Fig. 7.1).24,25 These

97
98 Part I: Science and Clinical Oncology

HSC Self-renewal Phenotype in humans:


CD34+ CD38- CD90+
Differentiation

MPPs
Phenotype in humans:
CD34+ CD38- CD90-

Common
myeloid
progenitor
CMP
Megakaryocyte
Common
erythroid Granulocyte lymphoid
progenitor macrophage progenitor
progenitor CLP
MEP GMP

Erythrocyte Granulocyte Monocyte Dendritic cell NK-cell T-cell B-cell


Megakaryocyte progenitors progenitors progenitors progenitors progenitors progenitors progenitors

Platelets Erythrocytes Macrophages NK cells B-cells


Granulocytes Dendritic cells T-cells

Figure 7.1  •  Developmental hierarchy of the hematopoietic system. All mature blood cell types arise from a small subset of immature and multipotent
cells, which contains two distinct cell types: the hematopoietic stem cells (HSCs) and the multipotent progenitors (MPPs). Both HSCs and MPPs are capable
of multilineage reconstitution of the hematopoietic tissues in lethally irradiated mice, but only the HSCs are capable of self-renewal for the lifetime of the
animals. In contrast, MPPs, although able to give rise to large numbers of mature blood cells, can do so for only a limited time, usually a few months. (Diagram
modified from Seita J, Weissman IL. Hematopoietic stem cell: self-renewal versus differentiation. Wiley Interdiscip Rev Syst Biol Med. 2010;2:640–653.)

mature cell types perform a diverse set of functions, ensuring tissue the ability to self-renew. Only HSCs can reconstitute hematopoiesis
oxygenation, blood coagulation, and immunity. HSCs have four in the long-term, for the lifetime of the animal, whereas MPPs can
fundamental properties: self-renewal, differentiation, migration, and reconstitute hematopoiesis for a short period of time, usually less than
homeostatic control. First, HSCs need to self-renew to maintain the 16 weeks.31
stem cell pool. Self-renewal is not synonymous with proliferation. A similar hierarchy is mirrored also in human blood tissues, wherein
Self-renewal is a cell division in which one or both of the daughter HSCs and MPPs are characterized by a CD34+CD38− phenotype and
cells remain undifferentiated and maintain the identity of a stem cell, by the lack of mature blood cell lineage markers (Lin−), but can be
endowed with unlimited growth potential. Second, HSCs must undergo separated based on the differential expression of CD90 in HSCs (see
differentiation: they must generate a progeny of cells that expand and Fig. 7.1).32 More generally, the hierarchic organization of hematopoietic
progressively specialize in order to replenish the various populations tissues can be successfully applied to model the cellular composition
of mature cells that are damaged, aged, or otherwise lost. Third, HSCs and developmental dynamics of many adult mammalian tissues, such
are able to enter the circulation and migrate from one blood-forming as the brain, mammary gland, skin, and intestinal epithelium.33–37
site to another (e.g., from the marrow of one bone to that of another)
to replace HSCs that have been lost and maintain a constant output GENETIC REGULATION OF SELF-RENEWAL
of blood cells.26 Finally, HSCs must balance self-renewal, differentiation,
migration, and tissue repair according to a strict genetic program, The long-term survival of a tissue, either normal or neoplastic, is
which ensures homeostatic control of their numbers and which regulates dependent on its capacity to self-renew, whereas its overall size is
their expansion in response to various types of stress, such as bleeding determined by the balance between the rates of cell proliferation and
or infection.27–29 cell death (or removal) across its various components.38 In normal
In mice, HSCs represent less than 0.05% of bone marrow cells and tissues, stem cell numbers are under tight genetic regulation, resulting
are admixed with a heterogeneous pool of other immature progeni- in the long-term maintenance of a constant tissue size.27–29 In contrast,
tors. This pool includes a specific subset of immature multipotent tumor tissues have escaped this homeostatic regulation. Within a
progenitors (MPPs), which are able to differentiate into the full variety tumor, the number of cells with the ability to self-renew is constantly
of hematopoietic lineages but lack self-renewal capacity.30 These popula- expanding, resulting in continuous tissue growth. It is not surprising
tions form a hierarchy in which HSCs give rise to MPPs, which in therefore that many known oncogenes are able to expand stem cell
turn give rise to oligolineage progenitors and the various mature cell numbers, either by protecting them from apoptosis or by inhibiting
types of the blood and lymphoid tissues (see Fig. 7.1).6,25 As HSCs their differentiation. For example, enforced expression of Bcl2 results
mature to become MPPs, they increase their proliferation rate but lose in an expansion of HSCs,39 whereas enforced expression of c-myb and
Stem Cells, Cell Differentiation, and Cancer  •  CHAPTER 7 99

c-myc prevents hematopoietic cell differentiation along the erythroid The Wnt pathway was first implicated in a mouse model of breast
lineage.40,41 cancer, in which aberrant expression of Wnt1, caused by insertion of
Because the size of both normal and cancer tissues is dependent the mouse mammary tumor virus (MMTV) close to the Wnt1 gene,
on the number of cells able to self-renew, it is also not surprising that resulted in mammary tumors.49,50 Wnt1 belongs to a large family
a specific subset of oncogenes and/or tumor suppressor genes might of secreted proteins that bind to receptors of both the Frizzled and
directly activate and/or disable self-renewal pathways.10,42 Indeed, the low-density lipoprotein receptor–related protein (Lrp) families, result-
basic molecular machinery that ensures the unlimited growth capacity ing in activation of β-catenin.51 In the absence of Wnt stimulation,
of most cancer cells (e.g., the telomerase enzymatic complex) is β-catenin is degraded by the adenomatous polyposis coli (Apc), glycogen
fundamental to ensure self-renewal of most normal stem cells.12–14 synthase kinase-3β (Gsk3β), and Axin protein complex.52 Wnt/β-catenin
The capacity to modulate self-renewal, however, is not a necessary signaling plays a pivotal role in the self-renewal of many normal
attribute of all oncogenes and/or tumor suppressor genes: enforced stem cells.53 Activation of β-catenin signaling by Wnt proteins allows
expression of Bcl2, for instance, although able to expand the number expansion of stem/progenitor cells, both in vitro and in vivo, and
of self-renewing HSCs, is unable per se to endow self-renewal properties across different tissues, including the bone marrow, skin, mammary
on more mature cell types, such as MPPs.39 gland, and small intestinal epithelium.54–57 Constitutive activation
Among cancer genes with direct control over self-renewal func- of the β-catenin pathway is oncogenic58,59 and is almost invariably
tions, the best example is probably the Bmi1 oncogene, which can observed in colon cancer, most frequently by inactivating mutations
cooperate with c-myc to induce B-cell lymphomas.43 Bmi1 is a member of human APC.60 Current evidence suggests that β-catenin signaling is
of the Polycomb repressor complex 1 (PRC1), a multiprotein tran- key for cancer cells to maintain a stem/progenitor cell phenotype.20,61
scriptional repression complex involved in the epigenetic regulation In colon cancer cells, inhibition of the β-catenin pathway induces
of developmental and differentiation processes. In the hematopoietic expression of p21cip1/waf1, a cell-cycle inhibitor, followed by prolifera-
system, Bmi1 expression is highest in HSCs and gradually decreases tion arrest and acquisition of a differentiated phenotype.61 Enforced
as they differentiate into MPPs and oligolineage progenitors.44 In expression of c-myc, an oncogene whose transcription is activated by
mice genetically deficient for Bmi1 (Bmi1−/−) the number of HSCs β-catenin, inhibits p21cip1/waf1 expression and allows cancer cells to
is markedly reduced at birth, and their transplantation in lethally continue proliferating in the absence of β-catenin signaling, revealing
irradiated animals is able to reconstitute hematopoiesis, but only in a yet another role for a classic oncogene, c-myc, in the regulation of cell
transient and self-limiting fashion, indicating a cell autonomous defect differentiation.61
of self-renewal.15 Lack of Bmi1 expression causes similar phenotypes The Notch family of receptors, first identified as regulators of wing
across many types of adult stem cells, including neural and mammary patterning in the Drosophila fruit fly, controls development and dif-
stem cells,45,46 and can be phenocopied by the overexpression of PRC1 ferentiation in many tissues, and across many animal species.62 In
inhibitors, such as the histone deubiquitinase Usp16.47 Important to vitro stimulation of HSCs with selected Notch ligands (i.e., Jagged-1,
note, lack of Bmi1 expression is also associated with upregulation of Delta) transiently increases the activity of stem/progenitor cells, both
tumor suppressors, such as the cyclin-dependent kinase inhibitors in vitro and in vivo, suggesting that Notch activation promotes
encoded by the alternative transcripts of the Cdkn2a locus (i.e., the expansion of either HSCs or MPPs.63,64 Constitutive activation of the
p16Ink4a and p19Arf proteins), and with overexpression of transcription Notch pathway is endowed with powerful oncogenic effects in both
factors that promote differentiation, such as selected members of the mouse and human cells. The mouse oncogene int-3 encodes a con-
Hox family.15 This suggests that, in stem cells, the function of Bmi1 stitutively active, truncated variant of the Notch-4 receptor.65 In humans,
is to prevent the expression of a cascade of genes whose coordinated aberrant activation of NOTCH1, either by point mutation or chro-
activity can lead to loss of self-renewal capacity. Remarkably, in a mosomal translocation, is a common occurrence in T-cell acute
mouse model of leukemia initiated in Bmi1−/− mice, leukemic cells lymphoblastic leukemia (T-ALL).66,67 Important to note, inhibition
are initially able to expand and cause the death of their primary hosts, of NOTCH-1 signaling can induce apoptosis in T-ALL cell lines, and
but are unable to sustain long-term growth when transplanted into is now being explored as a promising therapeutic strategy.68
secondary syngeneic animals, where they eventually arrest, differentiate, The SHH pathway provides yet another example of a pathway with
and undergo apoptosis.16 Infection of Bmi1−/− leukemic cells with a key roles in tissue development, stem cell homeostasis, and oncogenesis.
retrovirus encoding for Bmi1 completely rescues this defect, allowing Like the Wnt factors, SHH is a secreted molecule and a powerful
leukemic cells to grow indefinitely and be serially passaged in mice.16 morphogen.69 SHH acts as a ligand for receptors encoded by members
Similar observations have also been made in human breast cancer, of the Patched gene family69 and activates signaling circuitries that
where downregulation of BMI1 by means of enforced expression of regulate self-renewal in selected types of stem cells, such as bladder
microRNA-200c associates with reduced expansion and impaired stem cells.70 Germline mutations in the SHH gene cause aberrations
tumorigenicity of cancer cells.17 Once more, this defect can be rescued of embryo development in Drosophila and holoprosencephaly in
by enforced expression of a modified version of the BMI1 mRNA, humans.42 With regard to human cancer, germline mutations in
insensitive to the inhibitory action of microRNA-200c.17 Interesting to Patched-1 (PTCH1) cause Gorlin or basal cell nevus syndrome (BCNS),
note, human BMI1 appears to induce telomerase activity.48 Again, in whereas sporadic mutations in PTCH1 are observed in the majority
many human model systems, cancer cells lacking telomerase, although of sporadic basal cell carcinomas (BCCs) and a substantial fraction of
capable of substantial short-term proliferation and numeric expansion, medulloblastomas.42 Most importantly, pharmacologic inhibition of the
progressively exhaust their growth capacity and irreversibly arrest.14 SHH pathway has shown powerful antitumor activity in clinical trials,
These studies reinforce the concept that in order to endow cancer cells against both human BCCs and medulloblastomas harboring PTCH1
with unlimited growth potential, a property frequently referred to as mutations, and is rapidly changing treatment guidelines for these
“immortality,”38 constitutive activation of proliferation pathways is not diseases.71–74
sufficient. It is also necessary to ensure the activation of self-renewal In conclusion, mouse genetic studies have contributed to the
pathways, either directly (i.e., by activating genes that positively discovery of several gene families involved in either the positive or
regulate them) or indirectly (i.e., by inactivating genes that negatively negative regulation of self-renewal properties in normal stem cell
regulate them). populations. Normal stem cells are characterized by the high expression
Many other signaling pathways implicated in oncogenesis are known of genes required to enable self renewal, and by the low expression
to play central roles in stem cell biology, tissue morphogenesis and of genes known to disable it. The observation that, in cancer cells,
development. Classic examples among them are the Wnt, Notch, and transforming mutations often target the same gene families, either by
Sonic hedgehog (SHH) pathways, all of which appear to regulate self- constitutively activating genes that promote self-renewal (Bmi1, Wnt,
renewal functions in many normal tissues. Notch, SHH) or by inactivating genes that contribute to restrict it
100 Part I: Science and Clinical Oncology

(Cdkn2a), suggests that stem cells and cancer cells depend on a common life span, it is unlikely that the full repertoire of mutations necessary
set of signaling pathways to control their numbers and stimulate their to achieve malignant transformation might occur during their relatively
growth. The therapeutic success obtained with SHH inhibitors short life. Second, many tumors arise in tissues that are known to
demonstrates how the study of stem cells and their self-renewal pathways contain stem cell populations, intrinsically endowed with the ability
can lead to novel and powerful antitumor treatments. to self-renew. Because cancer cells must undergo self-renewal to achieve
unlimited growth, stem cells might be more easily poised to undergo
TARGET CELLS FOR MALIGNANT early malignant transformation steps as compared with more mature
TRANSFORMATION cells, which naturally lack this fundamental property and would need
to aberrantly activate self-renewal pathways to become malignant.
If oncogenic mutations often target signaling pathways that regulate However, even if the first set of initiating events is likely to target a
self-renewal, then are stem cells the targets of neoplastic transformation? stem cell population, it is possible that, as part of disease progression,
Several lines of evidence suggest that stem cells might be involved in the aberrant progeny of mutated stem cells might acquire additional
the initial phases of cancer development. First, the fact that multiple changes and eventually gain the capacity to self-renew. In this scenario,
and sequential mutations are necessary for a cell to become cancerous75,76 the malignant clone would acquire a second population of self-renewing
suggests that, in many cases, mutations might need, at first, to cells, whose phenotypic identity might resemble that of a progenitor
accumulate in a stem cell. Because mature cells often have a limited rather than a stem cell (Fig. 7.2).2

Mature cells
Normal stem cell Progenitor cells

Loss of homeostatic controls due to:


“Early” mutations - inhibition of apoptosis
- increased proliferation
1-5 - escape from innate immune surveillance
- escape from adaptive immune surveillance
Aberrant and/or incomplete
differentiation
Pre-cancerous
lesion 1-5
1-2 1-5
1 1-5 1-5
1-5
1-4 1-5 1-5
1-3 1-5 1-5
Early stage
disease 1-5 1-5
1-5
(e.g., chronic phase
of CML)

“Late” mutations Aberrant acquisition of Blast transformation and


6-7 self-renewal capacity by accelerated disease
progenitor cells
disease progression
1-7
1-7
1-6 1-7 1-7
1-7
1-7 1-7
1-7 1-7 1-7
1-7 1-7
1-7

Figure 7.2  •  Target cells for neoplastic transformation. In many tissues, stem cells are the only cells capable of self-renewal, which confers them a long
life-span and unlimited expansion capacity. As a result of this unique property, stem cells are ideal targets for neoplastic transformation, because they can
accumulate several mutations over time and are naturally endowed with one of the hallmark features of cancer cells (i.e., immortality). On the other hand,
for other cell types to become the cells of origin of a malignant tumor, they must first acquire the ability to self-renew. Current evidence suggests that during
the natural history of human tumors such as chronic myelogenous leukemia (CML), both events can take place. During the initial stages of the disease, a first
set of “early” mutations (1–5) accumulates in the stem-cells, causing loss of normal homeostatic controls on tissue expansion. Subsequently, during the advanced
stages of the disease, a second set of “late” mutations (6–7) causes aberrant acquisition of self-renewal by more mature progenitor compartments of the
neoplastic clone, resulting in the onset of a second population of cancer cells with unlimited growth potential. (Diagram modified from Rossi DJ, Jamieson
CH, Weissman IL. Stem cells and the pathways to aging and cancer. Cell. 2008;132:681–696.)
Stem Cells, Cell Differentiation, and Cancer  •  CHAPTER 7 101

Experimental evidence in support of a stem cell origin of cancer, subsequent mutations in either the stem cells or their progeny might
at least during early initiation phases, is provided by elegant experiments lead to overt leukemia.2 Indeed, recent studies indicate that, within
in a mouse model of glioblastoma, caused by conditional deletion of the same AML patient, cells that appear to behave as phenotypically
the Neurofibromatosis-1 (Nf1), Tp53, and Pten tumor suppressor genes.77 normal HSCs are often carriers of a subset of the somatic mutations
In this model, combined deletion of the three genes in brain tissues found in the leukemic blasts.80 Interesting to note, such shared muta-
leads to rapid development of high-grade gliomas with 100% penetrance. tions often target genes encoding for epigenetic modifiers involved
This effect, however, can be observed only when the deletion is specifi- in the regulation of DNA methylation and hydroxymethylation
cally targeted to cell compartments that include stem cell populations, (DNMT3A, TET2), whereas mutations restricted to leukemic blasts
either by conditional knockout of the tumor suppressor genes in cells often target classic oncogenes (FLT3, KRAS).81
expressing Nestin (a marker associated with neural stem or progenitor A similar scenario is observed in patients with CML, wherein
cells), or by somatic ablation of the tumor suppressor genes in the leukemic cells are characterized by the t(9;22) BCR/ABL chromosomal
mouse brain’s subventricular zone (SVZ), where neural stem cells translocation. In the initial “chronic phase” of human CML, the
reside, achieved by stereotactic injection of an adenovirus encoding leukemic population undergoes a multilineage differentiation process,
the Cre recombinase in mice genetically engineered to carry Nf1, similar to that sustained by normal HSCs.87,88 At this initial stage,
Tp53, and Pten alleles between loxP sites. Most remarkably, somatic β-catenin signaling is active in HSCs but not in their descendant
ablation of the same tumor suppressors in other areas of the brain, progenitor cells. However, when CML progresses to “myeloid blast
such as the striatum, is unable to generate any tumor. Moreover, crisis”, patients develop an AML-like disease. In this second stage,
tumors generated from adenovirus injection in the SVZ are able to the leukemic clone acquires a second, phenotypically distinct self-
disseminate throughout the brain parenchyma, following natural renewing population, whose surface marker profile corresponds to
migration pathways of neural stem cells, and exhibiting phenotypic that of a granulocyte-macrophage progenitor cell (GMP) and which
hallmarks of multilineage differentiation along the astrocytic, neuronal, is able to transplant the disease to immunodeficient mice.89 This
and oligodendrocytic lineages. Conceptually similar observations have second population is characterized by de novo aberrant activation of
also been made in mouse models of intestinal cancer.78 the β-catenin pathway, often secondary to a pathologic missplicing
Although it is likely that a first set of early-transforming mutations of the mRNA encoding for the GSK3β inhibitory enzyme.90 Thus
might accumulate in the stem cell compartment, it is also possible that disease progression in human CML appears to result from aberrant
more mature, downstream progenitors that originated as the progeny activation of self-renewal pathways in a progenitor cell population or,
of mutated stem cells might acquire a second set of late mutations, more likely, failure to shut down these pathways during differentiation
which might constitute the ultimate transforming event giving rise processes. Once again, these studies suggest that, in human CML, early
to cancer.79 In this scenario, mutated stem cells might represent a mutational events (e.g., BCR/ABL) accumulate in HSCs during the
“reservoir” population of precancerous cells, and fully transformed chronic phase of the disease, whereas during blast crisis a second set
progenitors might sustain the growth of the full-blown neoplastic of mutations leads to the rise of a second self-renewing population,
mass (see Fig. 7.2).80,81 This concept is supported by studies on normal aberrantly originated from a progenitor cell, not an HSC (see Fig. 7.2).
hematopoietic development and hematologic malignancies, in which Most remarkably, a conceptually analogous picture is also emerging
the developmental hierarchy of the different cellular compartments is with regard to different forms of human lymphoid malignancy.91 In
best understood, and in which phenotypic differences between HSCs the case of chronic lymphocytic leukemia (CLL), for example, a subset
and MPPs allow a careful dissection of their relative contributions. of the somatic mutations associated with the transformed B-cell clone
For example, in mice, simultaneous deletion of the Trp53 gene and are frequently found in the CD34+CD19− (i.e., stem/progenitor)
the Cdkn2a locus, which encodes for the p16Ink4a and p19Arf tumor fraction of circulating hematopoietic cells.92 This finding is theoretically
suppressor proteins, leads to abnormal and selective acquisition of important because it introduces the possibility that therapies aimed
self-renewal properties by MPPs, which remain capable of producing at eradicating B-cell populations (e.g., anti-CD20 monoclonal antibod-
the various differentiated cell types, but also acquire self-renewal and ies, anti-CD19 bispecific antibodies, T cells engineered with anti-CD19
become capable of supporting the production of blood tissues on serial chimeric antigen receptors), although capable of inducing profound
transplantation, in essence behaving as stem cells.82 This observation clinical responses, might be unable to fully eradicate the preneoplastic
indicates that genetic mutations commonly observed in human cancer cells that give rise to such disorders. In this scenario, treated patients
might be responsible for a progressive and pathologic “unleashing” might develop late recurrences, in which both the mutational repertoire
of self-renewal properties in cells that are usually not endowed and phenotypic traits of the relapsed clone could share only a limited
with them. overlap with that of the original disease, as commonly observed in
Similar observations have been made also for different forms of the case of follicular lymphomas (FLs) that “transform” into diffuse
human leukemia, such as acute myelogenous leukemia (AML) and large B-cell lymphomas (DLBCLs).93
chronic myelogenous leukemia (CML). One of the most frequent Taken together, these observations support the notion that “early”
mutations in AML is the t(8;21) translocation, which results in the oncogenic mutations might accumulate in stem cells and that during
expression of a chimeric AML1/ETO transcript.83 Marrow samples disease progression additional “late” mutations can accumulate in
from patients with t(8;21) AML treated at Hiroshima Hospital, when their transformed progenies, leading to aberrant acquisition of self-
collected at clinical remission, contained HSCs (CD34+CD90+ renewal in more differentiated cell types.
CD38−Lin−) that had up to 90% incidence of the chimeric AML1-ETO
transcript.84 When these mutated HSCs were analyzed in vitro for EVIDENCE FOR CANCER STEM CELLS
their differentiation capacity, they gave rise to normal myeloerythroid
progenies, showing that the mutation, alone and by itself, was insuf- It has long been known that tumor tissues contain heterogeneous
ficient to confer a fully transformed phenotype. Remarkably, marrow populations of cancer cells, characterized by different phenotypes.94
samples from the same patients, when collected at relapse, contained This diversity can arise in part from accumulation of sequential and/
high numbers of cells with an MPP phenotype (CD34+CD90− or divergent mutations caused by genetic instability, and in part by
CD38−Lin−) that gave rise to leukemic “blast” colonies in vitro.84 This the variable action of environmental factors (Fig. 7.3A). In addition,
observation is common to many forms of AML85 and is associated however, a tumor can also be viewed as an aberrant organ, sustained in
with the expansion of a pathologic population of MPPs that have its growth by a pathologic stem cell population, which originates from
abnormally gained the capacity to self-renew.86 Taken together, these genetic mutations and/or epigenetic modifications leading to increased
observations suggest that, in human AML, a first set of mutations cell proliferation and tissue expansion. It is also possible that, in this
(e.g., AML1/ETO) might initially accumulate in stem cells, and that aberrant organ, the pathologic stem cell population could undergo
102 Part I: Science and Clinical Oncology

Genetic
mutation

on
ati
Genetic

nti
mutation

e
fer
Dif
Environmental Environmental
modulation modulation

A B

Figure 7.3  •  Two models of cancer cell heterogeneity. (A) In the traditional model, heterogeneity is explained by differences in the repertoire of genetic
mutations (magenta cells) and by the variable effects of microenvironmental factors (red cells). In this model, all cancer cells are assumed to have the intrinsic
ability to self-renew and form new tumors (circular arrows). (B) In the “cancer stem cell” model, cell heterogeneity is caused not only by genetic mutations
and environmental factors, but also by differentiation. In this case, only a subset of cancer cells, the cancer stem cells (CSCs), are capable of self-renewal and
the formation of new tumors (orange cells). As observed for normal stem cells in healthy tissues, CSCs can give rise to more CSCs with the capacity to self-renew,
as well as differentiated cancer cells that lack this ability (various shades of green cells). CSCs can also acquire additional mutations (magenta cells) and are subject
to the action of environmental stimuli, both of which add to the tumor tissue’s diversity (red cells). Genetic mutations can sometimes perturb differentiation
processes, leading to aberrant differentiated phenotypes (blue cells). (Diagram modified from Reya T, Morrison SJ, Clarke MF, Weissman IL. Stem cells, cancer,
and cancer stem cells. Nature 2001;414:105-11.)

an abnormal multilineage differentiation process that is reminiscent clonogenic assays showed that only a minority of cancer cells, ranging
(a “caricature”) of the one observed in normal tissues, giving rise to from 1 : 100 to 1 : 10,000, were able to form colonies.98 These results
multiple, phenotypically diverse populations of cancer cells, some of were reproduced across many tumor types and suggested the existence
which, although genetically identical, might differ in their epigenetic of minority populations of stemlike cancer cells, but researchers were
status and their ability to self-renew (Fig. 7.3B).10,11 Several lines of unable to formally prove this hypothesis because they could not rule
evidence indicate that, indeed, multilineage differentiation does occur out stochastic effects in cell survival and engraftment.100
in cancer tissues and contributes substantially to cellular heterogeneity,95 To prove that tumor tissues contain a specific subset of cancer cells
in addition to genetic instability and environmental factors, which that is preferentially responsible for sustaining tumor growth, it is
undoubtedly contribute to the observed variability.96 Indeed, it is well necessary to isolate different populations of cancer cells in parallel
documented that the cell composition of cancer tissues frequently and compare their properties side by side, in a prospective assay—for
mirrors that of their normal counterparts in terms of differentiated example, a transplantation assay in which cancer cells are tested for
cell types.95 A classic example is the frequent presence of abundant their tumorigenic capacity.101 This type of experiment was first per-
mucus-producing gobletlike cells in human colon carcinomas (Fig. formed in human AML, where it was shown that, in some cases, the
7.4A–B). Perhaps one of the most striking examples of abnormal “tumor-initiating” capacity was enriched in a phenotypic subset of
differentiation in cancer is observed in some germ cell tumors, such as the leukemic clone. In these studies, the subset of leukemic cells that
teratomas and teratocarcinomas, in which mature organlike structures, displayed the highest capacity for establishing human AML in
such as teeth, skin, and hair, can be found simultaneously admixed immunodeficient mice was the population characterized by the
within the tumor mass. Remarkably, within many of these tumors, CD34+CD38− phenotype.86,102,103 As previously discussed, the human
only a minority of cancer cells express immature cell markers such CD34+CD38− population corresponds to a very immature subset of
as α-fetoprotein (Fig. 7.4D). The terminally differentiated cancer hematopoietic cells that contains HSCs (i.e., CD90+CD34+CD38−)
cells that form the teeth and hair in the tumors are, most likely, and MPPs (i.e., CD90−CD34+CD38−),32 both of which are involved
unable to proliferate and therefore contribute substantially to in the natural history of these leukemic disorders.84,86 Although
the long term expansion and metastasis of the malignant tissues. characterized by the highest frequency of tumorigenic cells, the
On the contrary, the minority population of immature cancer cells CD34+CD38− population is not the only self-renewing population
expressing α-fetoprotein is likely to be the source of long-term tumor observed across all cases of AML.104 As observed for CML during
growth, as well as represent the progenitor population from which the blast crisis, a second tumorigenic population, characterized by a more
other, more mature populations arise. Preliminary data on teratoma mature, GMP-like phenotype (CD34+CD38+CD110−CD45RA+), can
formation by human embryonic stem cells (hESCs) lend support to be frequently detected also in AML. This second population is hierarchi-
this working model.97 cally related to the first (i.e., originates as the progeny of CD34+CD38−
If a tumor is viewed as an abnormal organ, then the principles of cells, but not vice versa) and is characterized by a much lower frequency
stem cell biology can be applied to model its physiology and clinical of tumorigenic cells.86
behavior.10,11 Historically, this concept was first explored using clono- Tumorigenic and nontumorigenic subsets of cancer cells have been
genic assays, in which single-cell suspensions of cancer cells were tested isolated also from epithelial solid tumors, such as breast and colon
in limiting dilution for their capacity to form colonies, either in vitro carcinomas.18,19 As in the case of leukemias, tumorigenic cancer cells
or in vivo.98 Indeed, in vivo clonogenic assays had been previously could be distinguished from nontumorigenic ones based on their dif-
used to provide the first experimental evidence of HSCs, because they ferential expression of surface markers. In breast cancer, the tumorigenic
showed that only a minority of normal bone marrow cells, when cell population was characterized as CD44+CD24−/low,18 whereas in colon
transplanted in irradiated mice, were able to engraft and give rise to cancer it was characterized as EpCAMhighCD44+CD166+.19 In both
blood-forming colonies in the spleen.99 When applied to tumors, experimental systems, as few as 100 to 200 cells from the tumorigenic
Stem Cells, Cell Differentiation, and Cancer  •  CHAPTER 7 103

A B

C D

E F

Figure 7.4  •  Clinical evidence in support of the “cancer stem cell” (CSC) model. When analyzed histologically, cancer tissues frequently recapitulate the
cell composition of their normal counterparts, including the presence of mature, differentiated cells. (A) Normal colon epithelia contain large numbers of
differentiated mucus-secreting goblet cells, which can be easily visualized by immunohistochemistry against the Mucin-2 (MUC2) antigen (brown staining).
(B) Colon cancer tissues frequently display a cellular composition similar to that of normal epithelia, characterized by large numbers of abnormal goblet cells.
Similar observations can been made in other forms of solid cancer, including teratocarcinomas. (C) A pretreatment abdominal computed tomography (CT)
scan of a patient with metastatic teratocarcinoma of the testis demonstrates a large retroperitoneal mass (arrowheads). (D) A biopsy specimen of the testicular
tumor analyzed with immunohistochemistry reveals expression of the immature marker α-fetoprotein by only rare cancer cells (brown cells, arrows). (E) A CT
scan after four cycles of platinum-based chemotherapy reveals persistence of a large retroperitoneal mass (arrowhead). (F) A biopsy of the residual mass reveals
only mature teratoma tissues and no cells expressing α-fetoprotein. This patient remained disease-free for more than 10 years, suggesting that, in some patients,
therapies that selectively eliminate a minority CSC population could be curative.

population were able to consistently form tumors when injected in including MUC2+ goblet cells.95 These experiments provide formal
immunodeficient mice, whereas as many as 104 cells from other cell proof that (1) nontumorigenic populations originate as the progeny of
populations within the same tumors repeatedly failed to do so. In both tumorigenic ones and (2) tumorigenic cells can undergo multilineage
cases, cancer tissues originating from the tumorigenic subset contained differentiation processes, akin to those that allow normal stem cells to
the full repertoire of cell populations observed in the parent tissues, sustain the generation of complex and diverse tissues. Conceptually
including the tumorigenic subset itself and other nontumorigenic similar results have also been obtained from lineage-tracing experiments
populations. Important to note, these cancer tissues could be serially in transgenic mouse cancer models105,106 and from single-cell RNA-
passaged in mice, each time with similar results. Lineage-tracing sequencing experiments in human primary oligodendrogliomas.107
experiments performed in colon cancer showed that monoclonal Taken together, these data indicate the presence of a cellular
cancer tissues, originated by injection of a single tumorigenic cancer hierarchy within many cancer tissues, in which only a fraction of the
cell bearing the EpCAMhighCD44+ phenotype and infected with a cells have the ability to engraft in mice. These cells can expand
reporter lentivirus encoding for the enhanced green fluorescence protein indefinitely and differentiate into multiple lineages of specialized cell
(EGFP), reproduced the full cellular diversity of the parent tissues, types, thus showing two hallmark properties of stem cells: self-renewal
including the presence of EGFP+EpCAMlowCD44− nontumorigenic and differentiation. Based on the fulfillment of these two properties,
cancer cells, and a variety of multiple EGFP+ differentiated cell types, the tumorigenic subset of cancer cells is often called the “cancer stem
104 Part I: Science and Clinical Oncology

Box 7.1. ALTERNATIVE MODELS OF CANCER CELL HETEROGENEITY


An alternative explanation for the fact that different subsets of cancer case of mouse tumors, transplantation experiments are performed into
cells display different tumorigenic capacity when transplanted in syngeneic hosts, thus ruling out confounding effects from cross-species
immunodeficient mice could be that all cancer cells are actually barriers, either biologic or immunologic; lineage-tracing experiments
tumorigenic, but only a specific subset, with a specific phenotype, is are performed in genetically engineered mice, in which primary tumors
able to engraft in the mouse. The ability of purified cell populations to can be studied in situ, in the original location where they arise, thus
engraft in immunodeficient mice is dependent on many external ruling out transplantation artifacts. Among the advantages of
variables, such as the purification and transplantation procedures, the lineage-tracing experiments performed in transgenic mouse models is
degree of mouse immunodeficiency, the capacity of the cells to resist the possibility to perform intravital imaging on primary tumors and
nutrient deprivation and induce neoangiogenesis, and the degree of follow the clonal growth dynamics of individual cancer cells in a
cross talk between secreted growth factors and their receptors across time-lapse fashion. This is usually achieved by performing experiments
mice and humans.145 Despite this, multiple lines of evidence argue in in genetically engineered mice that enable the “tagging” of individual
support of the existence of CSCs across many forms of cancer. For cancer cells with distinct fluorescent reporters. The results emerging
example, lineage-tracing experiments have shown that human tumors from this generation of in vivo imaging experiments show that, within
engrafted in mice do recapitulate the cell composition and phenotypic the same tumor, some cells are able to grow indefinitely, whereas
diversity of the primary tumors from which they have been derived, others appear to gradually exhaust their proliferation capacity.146
and contain both tumorigenic and nontumorigenic populations.95 This Finally, the differences observed among cancer cells in the expression
finding indicates that the mouse environment is compatible with of lineage-restricted differentiation antigens do not appear to be
multilineage differentiation processes and with survival of human caused by clonal differences in their repertoire of genetic mutations, as
nontumorigenic cancer cells. Most importantly, identical results can be demonstrated by single-cell RNA-sequencing experiments in human
reproduced using mouse-in-mouse tumors, with regard to in vivo primary oligodendrogliomas.107 Taken together, these findings support
lineage tracing experiments105,106,146 and the existence of both the idea that, as predicted by CSC models, tumors can retain elements
tumorigenic and nontumorigenic cancer cell populations.20,147 In the of the hierarchic organization of their parent tissues.

cell” subset. It is important to remember, however, that this is an poor outcomes, likely reflecting a higher CSC content and a more
operational definition, introduced to identify a subgroup of cancer immature differentiation state. This was first shown for breast cancer,
cells characterized by unique functional properties, but which, as using a 186-gene signature called the “invasiveness gene signature”
observed in leukemia, might not always necessarily correspond to the (IGS), obtained by comparing the gene expression profile of breast
stem cell population of their respective normal tissues. In colon CSCs with that of normal breast epithelial cells.116 The IGS was used
carcinomas, for instance, CSCs display a surface marker phenotype to stratify breast cancer patients with early-stage disease in different
(EpCAMhighCD44+CD166+) that is characteristic of cells found at the groups, based on the similarity of their whole tumor’s gene expression
bottom of normal colon crypts, and that defines a cell compartment profile with the IGS. Remarkably, high similarity to the IGS was
probably encompassing both stem and MPP subsets.95,108–110 Because associated with reduced overall and metastasis-free survival. Conceptu-
of our limited understanding of differentiation hierarchies in both ally similar findings have also been observed across a variety of other
normal breast and normal colon epithelia, it is currently impossible cancer types, including leukemia, head and neck, bladder, and colon
to dissect with precision whether in breast and colon cancer CSC cancer.95,104,117–120
activity is found in stem cells, in downstream MPPs, or in both, and More recently, a new generation of studies explored whether the
this remains the subject of active investigations. gene-expression profile of CSCs can be used to guide the discovery of
CSCs have also been identified in many other human tumors, clinically actionable biomarkers (e.g., markers able to predict tumor
including glioblastomas and several types of epithelial cancer, such as response to chemotherapy and guide treatment decisions). In colon
head and neck, pancreas, prostate, and bladder carcinomas.111–115 CD44 cancer, for example, a bioinformatics analysis designed to discover
can be used for the isolation of CSCs across many epithelial carcinomas. biomarkers of colon epithelial differentiation (i.e., markers whose
This is in agreement with the notion that CD44 is often expressed expression was inversely related to that of CSC markers, such as
heterogeneously within normal epithelia, usually in a gradient, with CD166) led to the identification of a novel subtype of clinically
the highest levels being observed at the most basal layers, where normal aggressive, poorly differentiated intestinal malignancies, characterized
stem or progenitor cells are usually thought to reside. Important to by the lack of expression of the caudal-type homeobox transcription
note, histologic analysis of well-differentiated head-and-neck carcinomas factor 2 (CDX2), a master transcription factor that is necessary for
showed that, in these tumors, CD44 expression is often inversely the correct multilineage differentiation of colon epithelial cells.121
correlated to expression of mature cell markers, such as involucrin Human CDX2neg colon tumors are associated with an increased risk
(IVL), and positively associated with expression of BMI1, a key element of relapse and reduced survival rates, irrespective of their clinical
of the self-renewal machinery in many types of stem cells.112 Box 7.1 stage or pathologic grade. Most importantly, however, patients with
discusses alternative models for cancer cell heterogeneity. CDX2neg colon tumors also appear to derive substantial benefit from
early treatment with adjuvant chemotherapy, in both stage II and stage
CLINICAL IMPLICATIONS OF CANCER III disease.121 This observation is very important because, although
STEM CELLS colon cancer patients with stage III disease are routinely treated with
adjuvant chemotherapy, patients with stage II disease are usually not,
The existence of CSCs has profound implications for clinical oncology. as most of them are cured by surgery alone, and clinicians do not
One of them is concerned with the prediction of patient outcomes have a reliable way to identify the small patient subgroup in which
at the time of diagnosis. If tumor tissues vary in CSC content, then the hazards of chemotherapy can be offset by a reduction in the risk
tumors containing a higher fraction of CSCs might be characterized of relapse. The identification of CDX2 as a predictive biomarker
by a more aggressive biology and worse clinical outcomes. Indeed, a of benefit from adjuvant chemotherapy therefore has the potential
number of studies have shown that tumors whose bulk gene expression to change the treatment algorithms for stage II colon cancer, and
profiles are similar to those of purified CSCs are associated with represents the first example of a finding related to CSC biology that
Stem Cells, Cell Differentiation, and Cancer  •  CHAPTER 7 105

can be translated into clinical practice. A similar set of findings has also as powerful antitumor drugs, future studies will need to address to
been made in the case of AML, wherein the gene-expression profile what extent their efficacy is related to their capacity to enable T-cell
of leukemic cells with CSC properties was used to generate a 17-gene elimination of CSCs.132
“leukemia stem cell” (LSC17) signature that can be used to identify Overall, preclinical studies have suggested that CSCs might hold
AML patients who are unlikely to benefit from standard treatments, the key to understanding many aspects of cancer resistance to antitumor
such as induction chemotherapy followed by allogeneic hematopoietic therapies in human patients. However, to formally address this question,
stem cell transplantation (HSCT), and who should be prioritized CSCs must be enumerated before and after therapy, thus requiring
for enrollment in clinical studies aimed at testing new therapeutic repeated in vivo tumor sampling. This kind of analysis can be performed
approaches.122 Taken together, these studies highlight the prognostic and in the case of patients undergoing neoadjuvant treatment regimens,
predictive significance of CSCs in the natural history of human tumors, because they receive chemotherapy and/or radiotherapy immediately
and underscore their relevance in the clinical management of cancer before surgery. Remarkably, neoadjuvant treatment of breast cancer
patients. with standard chemotherapy has been shown to cause an increase in
The existence of CSCs has important implications also for the the percentage content of CSCs in vivo.133 Given the variety of resistance
clinical evaluation of antitumor therapies. Historically, most antitumor mechanisms displayed by CSCs across different tumor types and
agents have been developed based on their ability to reduce tumor individual patients, characterization of the dominant resistance
size. Tumor shrinkage is dependent on the elimination of large numbers mechanisms in a tumor’s CSCs may become integral to the future
of cancer cells, but not necessarily of the CSCs, which can constitute development of personalized therapies.
a minority of the cancer tissue. This implies that CSCs might be Therapies specifically designed to target CSCs are still in their
spared even when tumors appear to be responding. Indeed, in diseases infancy but are making rapid progress. One promising avenue of
such as metastatic lung or breast cancer, although standard treatments research is targeting stem cell pathways, such as the wnt, Notch, and
frequently shrink tumors, tumors rapidly recur and only limited impact SHH pathways. The remarkable clinical results observed in BCCs
on patient survival is usually observed.123,124 Because anticancer therapies and medulloblastomas with inhibition of the SHH pathway support
are often screened based on their ability to shrink tumors rapidly, the use of this approach.
agents that selectively target CSCs could be overlooked. Indeed, in Another promising target is CD47, a cell surface receptor that is
the initial phases of the screen, CSC-specific agents might cause only overexpressed by CSCs in many tumor types.134 In normal physiology,
a modest reduction in tumor growth. However, if complete elimination CD47 mediates a “don’t eat me” signal that prevents normal cells,
of CSCs could be achieved, these treatments could permanently such as healthy red blood cells, from being eliminated by macro-
eradicate tumor tissues. Perhaps the best clinical evidence for this phages.135 CD47 thus acts as an inhibitor of the normal physiologic
concept comes from patients with teratocarcinoma. Platinum-based pathways that mediate the removal of aged and/or damaged cells by
chemotherapy is curative in the majority of these patients.125 Many macrophage scavenging, a process also known as “programmed cell
patients, however, are left with residual masses (see Fig. 7.4E), which removal”.136,137 CD47 is upregulated by HSCs when they enter the
usually undergo surgical resection. In most cases, histologic analysis bloodstream to recirculate, thus protecting them from phagocytosis.138
of residual tumor tissues reveals that cancer cells expressing α-fetoprotein Targeted inhibition of CD47 leads to eradication of established tumors
have been eliminated, leaving only differentiated cancer cells in a in many in vivo experimental models, including leukemias, lymphomas,
mature teratoma (see Fig. 7.4F). Patients with mature teratomas rarely and various forms of solid cancer.134,139–141
develop metastases and frequently achieve long-term cure, suggesting These observations suggest that, as part of neoplastic transformation,
that selective elimination of immature cancer cell populations can be cancer cells commonly disable pathways that are meant to ensure
sufficient to permanently abolish tumor growth. Conceptually similar homeostatic control of cell numbers by means of active cell elimination.
observations have also been made in experimental mouse models of These pathways include “suicide” or “programmed cell death” pathways
glioblastoma.126 (i.e., elimination by apoptosis) and “programmed cell removal” pathways
The lack of strong associations between tumor shrinkage and tumor (i.e., elimination by phagocytosis).136,137 Although several molecular
eradication led to the hypothesis that CSCs may be intrinsically resistant mechanisms are known to mediate the escape of cancer cells from
to chemotherapy and radiotherapy. This hypothesis is supported by apoptosis (e.g., Bcl2 overexpression, p53 inactivation), only one is
the observation that normal adult stem cells, such as HSCs, are fre- currently known to inhibit programmed cell removal: overexpression
quently found in a G0 quiescent state, which reduces their sensitivity of CD47. Future investigations will shed new light on the molecular
to antiproliferative agents.127 If the same is true for CSCs, then these regulation of CD47 and reveal other molecules involved in programmed
cells may be significantly more resistant to cytotoxic agents than their cell removal, such as those mediating positive “eat me” (i.e., opsoniza-
nontumorigenic progeny. In support of this concept, for instance, is tion) signals.142,143 These investigations will provide new molecular
the observation that the antitumor drug cytosine arabinoside (ara-C), targets for therapeutic interventions.
although capable of killing the majority of AML blast cells, selectively In summary, a new generation of antitumor therapies specifically
spares the CD34+CD38−CD123+ subset of leukemic CSCs.127 Similarly, targeting CSCs is currently under development and is about to enter
it has been shown that CSCs can be resistant to both chemotherapy clinical experimentation. Ultimately, well-designed trials of CSC-
and radiotherapy in many types of solid tumors, and several mechanisms directed therapies will provide the final testing ground for the CSC
mediating this resistance have been uncovered. For example, glioblas- hypothesis and its promise to improve patient lives.
toma CSCs have been shown to be radioresistant compared with their
nontumorigenic counterparts.128 This resistance is mediated, in part, FUTURE IMPLICATIONS OF CANCER
by enhanced activation of DNA repair pathways and rapid repair of STEM CELLS
radiation-induced DNA damage. Similar observations have been made
also for breast CSCs, in this case as the result of enhanced free radical The identification of CSCs across many tumor types is key to the
scavenging, which prevents radiation-induced DNA damage.129,130 In future development of clinical oncology, especially for the discovery
some tumors, CSCs might also be able to selectively evade elimination of new predictive biomarkers and the design of novel antitumor agents.
by cytotoxic T-cell effectors of the adaptive immune system. In head The study of CSCs, however, is technically challenging. CSCs often
and neck squamous cell carcinomas (HNSCCs), for example, CD44+ represent a minority of the cancer cell populations and need to be
cancer cells, which are enriched in CSCs, can sometimes over-express purified from primary tissues if their functional properties are to be
PD-L1, a costimulatory molecule able to dampen the activation of evaluated in the context of their original tissue microenvironment.
cytotoxic T cells by engaging a cognate receptor (PD-1) on their Future studies will need to devise new experimental platforms to
surface.131 Because inhibitors of the PD-1/PD-L1 pathway have emerged address this challenge, such as single-cell genomics technologies, able
106 Part I: Science and Clinical Oncology

to perform precise measurements on small biopsy samples and with Research, the Thomas and Stacey Siebel Foundation, the Damon
single-cell resolution.95 Runyon Cancer Research Foundation (Island Outreach Foundation),
The ability to identify and quantify CSCs should improve our the Schaefer Research Scholars Program of Columbia University, the
ability to evaluate the curative potential of both traditional and Breast Cancer Research Foundation (BCRF), the Adenoid Cystic
investigational therapies. Although cancer cell lines are useful experi- Carcinoma Research Foundation (ACCRF), the Edward Mallinckrodt
mental tools, especially suited for the in vitro dissection of individual Jr. Foundation, the Sydney Kimmel Foundation for Cancer Research,
biochemical pathways, they have often proven unreliable when used the Doris Duke Charitable Foundation, and the Ellison Medical
to model the clinical efficacy of antitumor drugs.144 Human xenografts Foundation.
established in immunodeficient mice recapitulate more accurately the P.D., M.D., I.L.W and M.F.C. are listed as a co-inventors on
morphology and phenotypic diversity of the patients’ original tumors, several patents and patent applications related to the contents of this
including their three-dimensional architecture and the presence of chapter. Such patents are owned by academic institutions (University
both tumorigenic and nontumorigenic cell populations.19 Xenograft of Michigan, Stanford University, Columbia University) and some of
models may help evaluate the differential effect of antitumor drugs them have been licensed to various biotechnology companies that
on different cancer cell populations. New antitumor agents could develop anti-tumor treatments, including: OncoMed Pharmaceuticals
therefore be tested for their ability to eliminate CSCs in vivo, across Inc., Quanticel Pharmaceuticals Inc. (now a fully owned subsidiary
multiple xenografts, and those with the greatest activity could be of Celgene) and Forty Seven Inc. As a result of the licensing agreements
selected for evaluation in clinical trials, hopefully paving the way to negotiated by the aforementioned academic institutions, the authors
a new generation of more effective cancer therapies. have received royalties from and/or stock in the aforementioned
companies. M.D. received research support from Varian Medical
ACKNOWLEDGMENTS Systems. I.L.W. is a founder and member of the board of directors
of Forty Seven Inc.
The authors have been supported by grants and scholarships from
the National Institutes of Health (NIH), the National Cancer Institute
(NCI), the Department of Defense (DoD), the California Institute The complete reference list is available online at
for Regenerative Medicine (CIRM), the Ludwig Institute for Cancer ExpertConsult.com.

KEY REFERENCES
1. Weissman IL. Stem cells: units of development, 17. Shimono Y, Zabala M, Cho RW, et al. Downregula- 81. Corces-Zimmerman MR, Hong WJ, Weissman IL,
units of regeneration, and units in evolution. Cell. tion of miRNA-200c links breast cancer stem cells Medeiros BC, Majeti R. Preleukemic mutations in
2000;100(1):157–168. with normal stem cells. Cell. 2009;138(3):592–603. human acute myeloid leukemia affect epigenetic
2. Weissman I. Stem cell research: paths to cancer thera- 18. Al-Hajj M, Wicha MS, Benito-Hernandez A, regulators and persist in remission. Proc Natl Acad
pies and regenerative medicine. JAMA. 2005;294(11): Morrison SJ, Clarke MF. Prospective identification Sci USA. 2014;111(7):2548–2553.
1359–1366. of tumorigenic breast cancer cells. Proc Natl Acad 82. Akala OO, Park IK, Qian D, Pihalja M, Becker MW,
3. Spangrude GJ, Heimfeld S, Weissman IL. Purifica- Sci USA. 2003;100(7):3983–3988. Clarke MF. Long-term haematopoietic reconstitution
tion and characterization of mouse hematopoietic 19. Dalerba P, Dylla SJ, Park IK, et al. Phenotypic charac- by Trp53−/−p16Ink4a−/−p19Arf−/− multipotent
stem cells. Science. 1988;241(4861):58–62. terization of human colorectal cancer stem cells. Proc progenitors. Nature. 2008;453(7192):228–232.
4. Baum CM, Weissman IL, Tsukamoto AS, Buckle Natl Acad Sci USA. 2007;104(24):10158–10163. 84. Miyamoto T, Weissman IL, Akashi K. AML1/
AM, Peault B. Isolation of a candidate human 20. Malanchi I, Peinado H, Kassen D, et al. Cutaneous ETO-expressing nonleukemic stem cells in acute
hematopoietic stem-cell population. Proc Natl Acad cancer stem cell maintenance is dependent on beta- myelogenous leukemia with 8;21 chromosomal
Sci USA. 1992;89(7):2804–2808. catenin signalling. Nature. 2008;452(7187):650– translocation. Proc Natl Acad Sci USA. 2000;97(13):
6. Weissman IL, Shizuru JA. The origins of the 653. 7521–7526.
identification and isolation of hematopoietic stem 26. Wright DE, Wagers AJ, Gulati AP, Johnson 86. Goardon N, Marchi E, Atzberger A, et al. Coex-
cells, and their capability to induce donor-specific FL, Weissman IL. Physiological migration of istence of LMPP-like and GMP-like leukemia
transplantation tolerance and treat autoimmune hematopoietic stem and progenitor cells. Science. stem cells in acute myeloid leukemia. Cancer Cell.
diseases. Blood. 2008;112(9):3543–3553. 2001;294(5548):1933–1936. 2011;19(1):138–152.
7. Chen JY, Miyanishi M, Wang SK, et al. Hoxb5 30. Morrison SJ, Wandycz AM, Hemmati HD, Wright 87. Fialkow PJ, Jacobson RJ, Papayannopoulou T.
marks long-term haematopoietic stem cells and DE, Weissman IL. Identification of a lineage of Chronic myelocytic leukemia: clonal origin in a
reveals a homogenous perivascular niche. Nature. multipotent hematopoietic progenitors. Development. stem cell common to the granulocyte, erythrocyte,
2016;530(7589):223–227. 1997;124(10):1929–1939. platelet and monocyte/macrophage. Am J Med.
10. Reya T, Morrison SJ, Clarke MF, Weissman IL. 32. Majeti R, Park CY, Weissman IL. Identification of a 1977;63(1):125–130.
Stem cells, cancer, and cancer stem cells. Nature. hierarchy of multipotent hematopoietic progenitors 89. Jamieson CH, Ailles LE, Dylla SJ, et al. Granulocyte-
2001;414(6859):105–111. in human cord blood. Cell Stem Cell. 2007;1(6): macrophage progenitors as candidate leukemic stem
11. Dalerba P, Cho RW, Clarke MF. Cancer stem cells: 635–645. cells in blast-crisis CML. N Engl J Med. 2004;351(7):
models and concepts. Annu Rev Med. 2007;58: 33. Uchida N, Buck DW, He D, et al. Direct isolation 657–667.
267–284. of human central nervous system stem cells. Proc 90. Abrahamsson AE, Geron I, Gotlib J, et al. Glycogen
12. Morrison SJ, Prowse KR, Ho P, Weissman IL. Telom- Natl Acad Sci USA. 2000;97(26):14720–14725. synthase kinase 3beta missplicing contributes to
erase activity in hematopoietic cells is associated 54. Zeng YA, Nusse R. Wnt proteins are self-renewal leukemia stem cell generation. Proc Natl Acad Sci
with self-renewal potential. Immunity. 1996;5(3): factors for mammary stem cells and promote their USA. 2009;106(10):3925–3929.
207–216. long-term expansion in culture. Cell Stem Cell. 91. Kikushige Y, Ishikawa F, Miyamoto T, et al. Self-
14. Dalerba P, Guiducci C, Poliani PL, et al. Recon- 2010;6(6):568–577. renewing hematopoietic stem cell is the primary
stitution of human telomerase reverse transcriptase 71. Rudin CM, Hann CL, Laterra J, et al. Treatment target in pathogenesis of human chronic lymphocytic
expression rescues colorectal carcinoma cells from of medulloblastoma with hedgehog pathway leukemia. Cancer Cell. 2011;20(2):246–259.
in vitro senescence: evidence against immortality inhibitor GDC-0449. N Engl J Med. 2009;361(12): 92. Damm F, Mylonas E, Cosson A, et al. Acquired
as a constitutive trait of tumor cells. Cancer Res. 1173–1178. initiating mutations in early hematopoietic cells
2005;65(6):2321–2329. 73. Sekulic A, Migden MR, Oro AE, et al. Efficacy of CLL patients. Cancer Discov. 2014;4(9):1088–
15. Park IK, Qian D, Kiel M, et al. Bmi-1 is required for and safety of vismodegib in advanced basal-cell 1101.
maintenance of adult self-renewing haematopoietic carcinoma. N Engl J Med. 2012;366(23):2171–2179. 95. Dalerba P, Kalisky T, Sahoo D, et al. Single-cell dissec-
stem cells. Nature. 2003;423(6937):302–305. 77. Alcantara Llaguno S, Chen J, Kwon CH, et al. tion of transcriptional heterogeneity in human colon
16. Lessard J, Sauvageau G. Bmi-1 determines the Malignant astrocytomas originate from neural stem/ tumors. Nat Biotechnol. 2011;29(12):1120–1127.
proliferative capacity of normal and leukaemic stem progenitor cells in a somatic tumor suppressor mouse 97. Tang C, Lee AS, Volkmer JP, et al. An antibody
cells. Nature. 2003;423(6937):255–260. model. Cancer Cell. 2009;15(1):45–56. against SSEA-5 glycan on human pluripotent stem
Stem Cells, Cell Differentiation, and Cancer  •  CHAPTER 7 107

cells enables removal of teratoma-forming cells. Nat stem cell properties in head and neck squamous cell 130. Diehn M, Cho RW, Lobo NA, et al. Association of
Biotechnol. 2011;29(9):829–834. carcinoma. Proc Natl Acad Sci USA. 2007;104(3): reactive oxygen species levels and radioresistance in
103. Bonnet D, Dick JE. Human acute myeloid leu- 973–978. cancer stem cells. Nature. 2009;458(7239):780–783.
kemia is organized as a hierarchy that originates 116. Liu R, Wang X, Chen GY, et al. The prognostic role 131. Lee Y, Shin JH, Longmire M, et al. CD44+ cells in
from a primitive hematopoietic cell. Nat Med. of a gene signature from tumorigenic breast-cancer head and neck squamous cell carcinoma suppress
1997;3(7):730–737. cells. N Engl J Med. 2007;356(3):217–226. T-cell-mediated immunity by selective constitutive
105. Driessens G, Beck B, Caauwe A, Simons BD, 121. Dalerba P, Sahoo D, Paik S, et al. CDX2 as a and inducible expression of PD-L1. Clin Cancer Res.
Blanpain C. Defining the mode of tumour growth Prognostic Biomarker in Stage II and Stage III 2016;22(14):3571–3581.
by clonal analysis. Nature. 2012;488(7412):527– Colon Cancer. N Engl J Med. 2016;374(3):211–222. 134. Willingham SB, Volkmer JP, Gentles AJ, et al. The
530. 122. Ng SW, Mitchell A, Kennedy JA, et al. A 17-gene CD47-signal regulatory protein alpha (SIRPa) inter-
106. Schepers AG, Snippert HJ, Stange DE, et al. Lineage stemness score for rapid determination of risk in action is a therapeutic target for human solid tumors.
tracing reveals Lgr5+ stem cell activity in mouse acute leukaemia. Nature. 2016;540(7633):433– Proc Natl Acad Sci USA. 2012;109(17):6662–6667.
intestinal adenomas. Science. 2012;337(6095):730– 437. 138. Jaiswal S, Jamieson CH, Pang WW, et al. CD47
735. 126. Chen J, Li Y, Yu TS, et al. A restricted cell population is upregulated on circulating hematopoietic stem
107. Tirosh I, Venteicher AS, Hebert C, et al. Single-cell propagates glioblastoma growth after chemotherapy. cells and leukemia cells to avoid phagocytosis. Cell.
RNA-seq supports a developmental hierarchy in Nature. 2012;488(7412):522–526. 2009;138(2):271–285.
human oligodendroglioma. Nature. 2016;539(7628): 128. Bao S, Wu Q, McLendon RE, et al. Glioma 146. Zomer A, Ellenbroek SI, Ritsma L, Beerling E,
309–313. stem cells promote radioresistance by preferential Vrisekoop N, Van Rheenen J. Intravital imaging
112. Prince ME, Sivanandan R, Kaczorowski A, et al. activation of the DNA damage response. Nature. of cancer stem cell plasticity in mammary tumors.
Identification of a subpopulation of cells with cancer 2006;444(7120):756–760. Stem Cells. 2013;31(3):602–606.
Stem Cells, Cell Differentiation, and Cancer  •  CHAPTER 7 107.e1
107.e1

REFERENCES
1. Weissman IL. Stem cells: units of development, 21. Liu H, Patel MR, Prescher JA, et al. Cancer stem 42. Taipale J, Beachy PA. The Hedgehog and Wnt signal-
units of regeneration, and units in evolution. Cell. cells from human breast tumors are involved in ling pathways in cancer. Nature. 2001;411(6835):
2000;100(1):157–168. spontaneous metastases in orthotopic mouse models. 349–354.
2. Weissman I. Stem cell research: paths to cancer thera- Proc Natl Acad Sci USA. 2010;107(42):18115– 43. van Lohuizen M, Verbeek S, Scheijen B, Wientjens
pies and regenerative medicine. JAMA. 2005;294(11): 18120. E, van der Gulden H, Berns A. Identification of
1359–1366. 22. Weissman IL. Translating stem and progenitor cell cooperating oncogenes in E mu-myc transgenic mice
3. Spangrude GJ, Heimfeld S, Weissman IL. Purifica- biology to the clinic: barriers and opportunities. by provirus tagging. Cell. 1991;65(5):737–752.
tion and characterization of mouse hematopoietic Science. 2000;287(5457):1442–1446. 44. Akashi K, He X, Chen J, et al. Transcriptional
stem cells. Science. 1988;241(4861):58–62. 23. Terskikh AV, Easterday MC, Li L, et al. From accessibility for genes of multiple tissues and hema-
4. Baum CM, Weissman IL, Tsukamoto AS, Buckle hematopoiesis to neuropoiesis: evidence of overlap- topoietic lineages is hierarchically controlled during
AM, Peault B. Isolation of a candidate human ping genetic programs. Proc Natl Acad Sci USA. early hematopoiesis. Blood. 2003;101(2):383–389.
hematopoietic stem-cell population. Proc Natl Acad 2001;98(14):7934–7939. 45. Molofsky AV, Pardal R, Iwashita T, Park IK, Clarke
Sci USA. 1992;89(7):2804–2808. 24. Kondo M, Wagers AJ, Manz MG, et al. Biology of MF, Morrison SJ. Bmi-1 dependence distinguishes
5. Morrison SJ, Weissman IL. The long-term hematopoietic stem cells and progenitors: implica- neural stem cell self-renewal from progenitor
repopulating subset of hematopoietic stem cells is tions for clinical application. Annu Rev Immunol. proliferation. Nature. 2003;425(6961):962–967.
deterministic and isolatable by phenotype. Immunity. 2003;21:759–806. 46. Pietersen AM, Evers B, Prasad AA, et al. Bmi1 regu-
1994;1(8):661–673. 25. Seita J, Weissman IL. Hematopoietic stem cell: lates stem cells and proliferation and differentiation
6. Weissman IL, Shizuru JA. The origins of the self-renewal versus differentiation. Wiley Interdiscip of committed cells in mammary epithelium. Curr
identification and isolation of hematopoietic stem Rev Syst Biol Med. 2010;2(6):640–653. Biol. 2008;18(14):1094–1099.
cells, and their capability to induce donor-specific 26. Wright DE, Wagers AJ, Gulati AP, Johnson 47. Adorno M, Sikandar S, Mitra SS, et al. Usp16
transplantation tolerance and treat autoimmune FL, Weissman IL. Physiological migration of contributes to somatic stem-cell defects in Down’s
diseases. Blood. 2008;112(9):3543–3553. hematopoietic stem and progenitor cells. Science. syndrome. Nature. 2013;501(7467):380–384.
7. Chen JY, Miyanishi M, Wang SK, et al. Hoxb5 2001;294(5548):1933–1936. 48. Dimri GP, Martinez JL, Jacobs JJ, et al. The Bmi-1
marks long-term haematopoietic stem cells and 27. Phillips RL, Reinhart AJ, Van Zant G. Genetic oncogene induces telomerase activity and immortal-
reveals a homogenous perivascular niche. Nature. control of murine hematopoietic stem cell pool izes human mammary epithelial cells. Cancer Res.
2016;530(7589):223–227. sizes and cycling kinetics. Proc Natl Acad Sci USA. 2002;62(16):4736–4745.
8. Negrin RS, Atkinson K, Leemhuis T, et al. Trans- 1992;89(23):11607–11611. 49. Nusse R, Brown A, Papkoff J, et al. A new nomen-
plantation of highly purified CD34+Thy-1+ hema- 28. Muller-Sieburg CE, Cho RH, Sieburg HB, clature for int-1 and related genes: the Wnt gene
topoietic stem cells in patients with metastatic breast Kupriyanov S, Riblet R. Genetic control of hema- family. Cell. 1991;64(2):231.
cancer. Biol Blood Marrow Transplant. 2000;6(3): topoietic stem cell frequency in mice is mostly cell 50. Tsukamoto AS, Grosschedl R, Guzman RC, Parslow
262–271. autonomous. Blood. 2000;95(7):2446–2448. T, Varmus HE. Expression of the int-1 gene in
9. Muller AM, Kohrt HE, Cha S, et al. Long-term 29. Morrison SJ, Qian D, Jerabek L, et al. A genetic transgenic mice is associated with mammary gland
outcome of patients with metastatic breast cancer determinant that specifically regulates the frequency hyperplasia and adenocarcinomas in male and female
treated with high-dose chemotherapy and transplan- of hematopoietic stem cells. J Immunol. 2002;168(2): mice. Cell. 1988;55(4):619–625.
tation of purified autologous hematopoietic stem 635–642. 51. Cadigan KM, Nusse R. Wnt signaling: a common
cells. Biol Blood Marrow Transplant. 2012;18(1): 30. Morrison SJ, Wandycz AM, Hemmati HD, Wright theme in animal development. Genes Dev. 1997;11(24):
125–133. DE, Weissman IL. Identification of a lineage of 3286–3305.
10. Reya T, Morrison SJ, Clarke MF, Weissman IL. multipotent hematopoietic progenitors. Development. 52. Spink KE, Polakis P, Weis WI. Structural basis of
Stem cells, cancer, and cancer stem cells. Nature. 1997;124(10):1929–1939. the Axin-adenomatous polyposis coli interaction.
2001;414(6859):105–111. 31. Kiel MJ, Yilmaz OH, Iwashita T, Terhorst C, EMBO J. 2000;19(10):2270–2279.
11. Dalerba P, Cho RW, Clarke MF. Cancer stem cells: Morrison SJ. SLAM family receptors distinguish 53. Reya T, Clevers H. Wnt signalling in stem cells and
models and concepts. Annu Rev Med. 2007;58: hematopoietic stem and progenitor cells and reveal cancer. Nature. 2005;434(7035):843–850.
267–284. endothelial niches for stem cells. Cell. 2005;121(7): 54. Zeng YA, Nusse R. Wnt proteins are self-renewal
12. Morrison SJ, Prowse KR, Ho P, Weissman IL. 1109–1121. factors for mammary stem cells and promote their long-
Telomerase activity in hematopoietic cells is 32. Majeti R, Park CY, Weissman IL. Identification term expansion in culture. Cell Stem Cell. 2010;6(6):
associated with self-renewal potential. Immunity. of a hierarchy of multipotent hematopoietic 568–577.
1996;5(3):207–216. progenitors in human cord blood. Cell Stem Cell. 55. Reya T, Duncan AW, Ailles L, et al. A role for Wnt
13. Allsopp RC, Morin GB, DePinho R, Harley CB, 2007;1(6):635–645. signalling in self-renewal of haematopoietic stem
Weissman IL. Telomerase is required to slow telo- 33. Uchida N, Buck DW, He D, et al. Direct isolation cells. Nature. 2003;423(6938):409–414.
mere shortening and extend replicative lifespan of of human central nervous system stem cells. Proc 56. Korinek V, Barker N, Moerer P, et al. Deple-
HSCs during serial transplantation. Blood. 2003; Natl Acad Sci USA. 2000;97(26):14720–14725. tion of epithelial stem-cell compartments in the
102(2):517–520. 34. Stingl J, Eirew P, Ricketson I, et al. Purification and small intestine of mice lacking Tcf-4. Nat Genet.
14. Dalerba P, Guiducci C, Poliani PL, et al. Recon- unique properties of mammary epithelial stem cells. 1998;19(4):379–383.
stitution of human telomerase reverse transcriptase Nature. 2006;439(7079):993–997. 57. Kishimoto J, Burgeson RE, Morgan BA. Wnt
expression rescues colorectal carcinoma cells from 35. Shackleton M, Vaillant F, Simpson KJ, et al. Genera- signaling maintains the hair-inducing activity of the
in vitro senescence: evidence against immortality tion of a functional mammary gland from a single dermal papilla. Genes Dev. 2000;14(10):1181–1185.
as a constitutive trait of tumor cells. Cancer Res. stem cell. Nature. 2006;439(7072):84–88. 58. Gat U, DasGupta R, Degenstein L, Fuchs E. De
2005;65(6):2321–2329. 36. Yui S, Nakamura T, Sato T, et al. Functional engraft- Novo hair follicle morphogenesis and hair tumors
15. Park IK, Qian D, Kiel M, et al. Bmi-1 is required for ment of colon epithelium expanded in vitro from a in mice expressing a truncated beta-catenin in skin.
maintenance of adult self-renewing haematopoietic single adult Lgr5(+) stem cell. Nat Med. 2012;18(4): Cell. 1998;95(5):605–614.
stem cells. Nature. 2003;423(6937):302–305. 618–623. 59. Chan EF, Gat U, McNiff JM, Fuchs E. A common
16. Lessard J, Sauvageau G. Bmi-1 determines the 37. Fuchs E. Skin stem cells: rising to the surface. J Cell human skin tumour is caused by activating mutations
proliferative capacity of normal and leukaemic stem Biol. 2008;180(2):273–284. in beta-catenin. Nat Genet. 1999;21(4):410–413.
cells. Nature. 2003;423(6937):255–260. 38. Hanahan D, Weinberg RA. Hallmarks of cancer: 60. Segditsas S, Tomlinson I. Colorectal cancer and
17. Shimono Y, Zabala M, Cho RW, et al. Downregula- the next generation. Cell. 2011;144(5):646–674. genetic alterations in the Wnt pathway. Oncogene.
tion of miRNA-200c links breast cancer stem cells 39. Domen J, Cheshier SH, Weissman IL. The role of 2006;25(57):7531–7537.
with normal stem cells. Cell. 2009;138(3):592–603. apoptosis in the regulation of hematopoietic stem 61. van de Wetering M, Sancho E, Verweij C, et al.
18. Al-Hajj M, Wicha MS, Benito-Hernandez A, cells: Overexpression of Bcl-2 increases both their The beta-catenin/TCF-4 complex imposes a crypt
Morrison SJ, Clarke MF. Prospective identification number and repopulation potential. J Exp Med. progenitor phenotype on colorectal cancer cells. Cell.
of tumorigenic breast cancer cells. Proc Natl Acad 2000;191(2):253–264. 2002;111(2):241–250.
Sci USA. 2003;100(7):3983–3988. 40. Prochownik EV, Kukowska J. Deregulated expression 62. Artavanis-Tsakonas S, Rand MD, Lake RJ. Notch
19. Dalerba P, Dylla SJ, Park IK, et al. Phenotypic charac- of c-myc by murine erythroleukaemia cells prevents signaling: cell fate control and signal integration in
terization of human colorectal cancer stem cells. Proc differentiation. Nature. 1986;322(6082):848–850. development. Science. 1999;284(5415):770–776.
Natl Acad Sci USA. 2007;104(24):10158–10163. 41. Clarke MF, Kukowska-Latallo JF, Westin E, Smith M, 63. Varnum-Finney B, Xu L, Brashem-Stein C, et al.
20. Malanchi I, Peinado H, Kassen D, et al. Cutaneous Prochownik EV. Constitutive expression of a c-myb Pluripotent, cytokine-dependent, hematopoietic
cancer stem cell maintenance is dependent on beta- cDNA blocks Friend murine erythroleukemia cell stem cells are immortalized by constitutive Notch1
catenin signalling. Nature. 2008;452(7187):650–653. differentiation. Mol Cell Biol. 1988;8(2):884–892. signaling. Nat Med. 2000;6(11):1278–1281.
107.e2 Part I: Science and Clinical Oncology

64. Duncan AW, Rattis FM, DiMascio LN, et al. on acute myeloid leukemia cells with long-term prolif- 107. Tirosh I, Venteicher AS, Hebert C, et al. Single-cell
Integration of Notch and Wnt signaling in hema- erative ability in vitro and in vivo. Blood. 1997;89(9): RNA-seq supports a developmental hierarchy in
topoietic stem cell maintenance. Nat Immunol. 3104–3112. human oligodendroglioma. Nature. 2016;539(7628):
2005;6(3):314–322. 86. Goardon N, Marchi E, Atzberger A, et al. Coex- 309–313.
65. Gallahan D, Callahan R. The mouse mammary istence of LMPP-like and GMP-like leukemia 108. Wielenga VJ, Smits R, Korinek V, et al. Expression of
tumor associated gene INT3 is a unique member stem cells in acute myeloid leukemia. Cancer Cell. CD44 in Apc and Tcf mutant mice implies regulation
of the NOTCH gene family (NOTCH4). Oncogene. 2011;19(1):138–152. by the WNT pathway. Am J Pathol. 1999;154(2):
1997;14(16):1883–1890. 87. Fialkow PJ, Jacobson RJ, Papayannopoulou T. 515–523.
66. Ellisen LW, Bird J, West DC, et al. TAN-1, the Chronic myelocytic leukemia: clonal origin in a 109. Jiao YF, Nakamura S, Sugai T, Yamada N, Habano
human homolog of the Drosophila notch gene, stem cell common to the granulocyte, erythrocyte, W. Serrated adenoma of the colorectum undergoes
is broken by chromosomal translocations in T platelet and monocyte/macrophage. Am J Med. a proliferation versus differentiation process: new
lymphoblastic neoplasms. Cell. 1991;66(4):649–661. 1977;63(1):125–130. conceptual interpretation of morphogenesis. Oncol-
67. Weng AP, Ferrando AA, Lee W, et al. Activating muta- 88. Takahashi N, Miura I, Saitoh K, Miura AB. Lineage ogy. 2008;74(3–4):127–134.
tions of NOTCH1 in human T cell acute lympho- involvement of stem cells bearing the Philadelphia 110. Levin TG, Powell AE, Davies PS, et al. Charac-
blastic leukemia. Science. 2004;306(5694):269–271. chromosome in chronic myeloid leukemia in terization of the intestinal cancer stem cell marker
68. Samon JB, Castillo-Martin M, Hadler M, et al. the chronic phase as shown by a combination of CD166 in the human and mouse gastrointestinal
Preclinical analysis of the gamma-secretase inhibitor fluorescence-activated cell sorting and fluorescence in tract. Gastroenterology. 2010;139(6):2072–2082,
PF-03084014 in combination with glucocorticoids situ hybridization. Blood. 1998;92(12):4758–4763. e2075.
in T-cell acute lymphoblastic leukemia. Mol Cancer 89. Jamieson CH, Ailles LE, Dylla SJ, et al. Granulocyte- 111. Singh SK, Hawkins C, Clarke ID, et al. Identification
Ther. 2012;11(7):1565–1575. macrophage progenitors as candidate leukemic stem of human brain tumour initiating cells. Nature.
69. Lum L, Beachy PA. The Hedgehog response cells in blast-crisis CML. N Engl J Med. 2004;351(7): 2004;432(7015):396–401.
network: sensors, switches, and routers. Science. 657–667. 112. Prince ME, Sivanandan R, Kaczorowski A, et al.
2004;304(5678):1755–1759. 90. Abrahamsson AE, Geron I, Gotlib J, et al. Glycogen Identification of a subpopulation of cells with cancer
70. Shin K, Lee J, Guo N, et al. Hedgehog/Wnt feedback synthase kinase 3beta missplicing contributes to stem cell properties in head and neck squamous cell
supports regenerative proliferation of epithelial stem leukemia stem cell generation. Proc Natl Acad Sci carcinoma. Proc Natl Acad Sci USA. 2007;104(3):
cells in bladder. Nature. 2011;472(7341):110–114. USA. 2009;106(10):3925–3929. 973–978.
71. Rudin CM, Hann CL, Laterra J, et al. Treat- 91. Kikushige Y, Ishikawa F, Miyamoto T, et al. Self- 113. Chan KS, Espinosa I, Chao M, et al. Identification,
ment of medulloblastoma with hedgehog renewing hematopoietic stem cell is the primary molecular characterization, clinical prognosis, and
pathway inhibitor GDC-0449. N Engl J Med. target in pathogenesis of human chronic lymphocytic therapeutic targeting of human bladder tumor-
2009;361(12):1173–1178. leukemia. Cancer Cell. 2011;20(2):246–259. initiating cells. Proc Natl Acad Sci USA. 2009;106(33):
72. Von Hoff DD, LoRusso PM, Rudin CM, et al. Inhi- 92. Damm F, Mylonas E, Cosson A, et al. Acquired 14016–14021.
bition of the hedgehog pathway in advanced basal-cell initiating mutations in early hematopoietic cells of 114. Li C, Heidt DG, Dalerba P, et al. Identifica-
carcinoma. N Engl J Med. 2009;361(12):1164–1172. CLL patients. Cancer Discov. 2014;4(9):1088–1101. tion of pancreatic cancer stem cells. Cancer Res.
73. Sekulic A, Migden MR, Oro AE, et al. Efficacy 93. Pasqualucci L, Khiabanian H, Fangazio M, et al. 2007;67(3):1030–1037.
and safety of vismodegib in advanced basal-cell Genetics of follicular lymphoma transformation. 115. Patrawala L, Calhoun T, Schneider-Broussard
carcinoma. N Engl J Med. 2012;366(23):2171–2179. Cell Rep. 2014;6(1):130–140. R, et al. Highly purified CD44+ prostate cancer
74. Tang JY, Mackay-Wiggan JM, Aszterbaum M, 94. Heppner GH. Tumor heterogeneity. Cancer Res. cells from xenograft human tumors are enriched
et al. Inhibiting the hedgehog pathway in patients 1984;44(6):2259–2265. in tumorigenic and metastatic progenitor cells.
with the basal-cell nevus syndrome. N Engl J Med. 95. Dalerba P, Kalisky T, Sahoo D, et al. Single-cell dissec- Oncogene. 2006;25(12):1696–1708.
2012;366(23):2180–2188. tion of transcriptional heterogeneity in human colon 116. Liu R, Wang X, Chen GY, et al. The prognostic role
75. Knudson AG Jr, Strong LC, Anderson DE. Hered- tumors. Nat Biotechnol. 2011;29(12):1120–1127. of a gene signature from tumorigenic breast-cancer
ity and cancer in man. Prog Med Genet. 1973;9: 96. Gerlinger M, Rowan AJ, Horswell S, et al. cells. N Engl J Med. 2007;356(3):217–226.
113–158. Intratumor heterogeneity and branched evolution 117. Gentles AJ, Plevritis SK, Majeti R, Alizadeh AA.
76. Fearon ER, Vogelstein B. A genetic model for revealed by multiregion sequencing. N Engl J Med. Association of a leukemic stem cell gene expression
colorectal tumorigenesis. Cell. 1990;61(5):759–767. 2012;366(10):883–892. signature with clinical outcomes in acute myeloid
77. Alcantara Llaguno S, Chen J, Kwon CH, et al. 97. Tang C, Lee AS, Volkmer JP, et al. An antibody leukemia. JAMA. 2010;304(24):2706–2715.
Malignant astrocytomas originate from neural stem/ against SSEA-5 glycan on human pluripotent stem 118. Joshua B, Kaplan MJ, Doweck I, et al. Frequency of
progenitor cells in a somatic tumor suppressor mouse cells enables removal of teratoma-forming cells. Nat cells expressing CD44, a head and neck cancer stem
model. Cancer Cell. 2009;15(1):45–56. Biotechnol. 2011;29(9):829–834. cell marker: correlation with tumor aggressiveness.
78. Barker N, Ridgway RA, van Es JH, et al. Crypt 98. Hamburger AW, Salmon SE. Primary bioassay of Head Neck. 2012;34(1):42–49.
stem cells as the cells-of-origin of intestinal cancer. human tumor stem cells. Science. 1977;197(4302): 119. Merlos-Suarez A, Barriga FM, Jung P, et al. The
Nature. 2009;457(7229):608–611. 461–463. intestinal stem cell signature identifies colorectal
79. Rossi DJ, Jamieson CH, Weissman IL. Stems 99. Till JE, Mc CE. A direct measurement of the radia- cancer stem cells and predicts disease relapse. Cell
cells and the pathways to aging and cancer. Cell. tion sensitivity of normal mouse bone marrow cells. Stem Cell. 2011;8:511–524.
2008;132(4):681–696. Radiat Res. 1961;14:213–222. 120. Volkmer JP, Sahoo D, Chin RK, et al. Three
80. Jan M, Snyder TM, Corces-Zimmerman MR, et al. 100. Weisenthal LM, Lippman ME. Clonogenic and differentiation states risk-stratify bladder cancer
Clonal evolution of preleukemic hematopoietic stem nonclonogenic in vitro chemosensitivity assays. into distinct subtypes. Proc Natl Acad Sci USA.
cells precedes human acute myeloid leukemia. Sci Cancer Treat Rep. 1985;69(6):615–632. 2012;109(6):2078–2083.
Transl Med. 2012;4(149):149ra118. 101. Clarke MF. A self-renewal assay for cancer stem 121. Dalerba P, Sahoo D, Paik S, et al. CDX2 as a
81. Corces-Zimmerman MR, Hong WJ, Weissman IL, cells. Cancer Chemother Pharmacol. 2005;56(suppl 1): prognostic biomarker in stage II and stage III colon
Medeiros BC, Majeti R. Preleukemic mutations in 64–68. cancer. N Engl J Med. 2016;374(3):211–222.
human acute myeloid leukemia affect epigenetic 102. Lapidot T, Sirard C, Vormoor J, et al. A cell initiating 122. Ng SW, Mitchell A, Kennedy JA, et al. A 17-gene
regulators and persist in remission. Proc Natl Acad human acute myeloid leukaemia after transplantation stemness score for rapid determination of risk in
Sci USA. 2014;111(7):2548–2553. into SCID mice. Nature. 1994;367(6464):645–648. acute leukaemia. Nature. 2016;540(7633):433–437.
82. Akala OO, Park IK, Qian D, Pihalja M, Becker MW, 103. Bonnet D, Dick JE. Human acute myeloid leukemia 123. Maemondo M, Inoue A, Kobayashi K, et al. Gefitinib
Clarke MF. Long-term haematopoietic reconstitution is organized as a hierarchy that originates from a or chemotherapy for non–small-cell lung cancer
by Trp53−/−p16Ink4a−/−p19Arf−/− multipotent primitive hematopoietic cell. Nat Med. 1997;3(7): with mutated EGFR. N Engl J Med. 2010;362(25):
progenitors. Nature. 2008;453(7192):228–232. 730–737. 2380–2388.
83. Peterson LF, Boyapati A, Ahn EY, et al. Acute 104. Eppert K, Takenaka K, Lechman ER, et al. 124. O’Shaughnessy J, Osborne C, Pippen JE, et al. Ini-
myeloid leukemia with the 8q22;21q22 transloca- Stem cell gene expression programs influence parib plus chemotherapy in metastatic triple-negative
tion: secondary mutational events and alternative clinical outcome in human leukemia. Nat Med. breast cancer. N Engl J Med. 2011;364(3):205–214.
t(8;21) transcripts. Blood. 2007;110(3):799–805. 2011;17(9):1086–1093. 125. Williams SD, Birch R, Einhorn LH, Irwin L,
84. Miyamoto T, Weissman IL, Akashi K. AML1/ 105. Driessens G, Beck B, Caauwe A, Simons BD, Greco FA, Loehrer PJ. Treatment of disseminated
ETO-expressing nonleukemic stem cells in acute Blanpain C. Defining the mode of tumour growth germ-cell tumors with cisplatin, bleomycin, and
myelogenous leukemia with 8;21 chromosomal by clonal analysis. Nature. 2012;488(7412):527– either vinblastine or etoposide. N Engl J Med.
translocation. Proc Natl Acad Sci USA. 2000;97(13): 530. 1987;316(23):1435–1440.
7521–7526. 106. Schepers AG, Snippert HJ, Stange DE, et al. Lineage 126. Chen J, Li Y, Yu TS, et al. A restricted cell population
85. Blair A, Hogge DE, Ailles LE, Lansdorp PM, tracing reveals Lgr5+ stem cell activity in mouse intes- propagates glioblastoma growth after chemotherapy.
Sutherland HJ. Lack of expression of Thy-1 (CD90) tinal adenomas. Science. 2012;337(6095):730–735. Nature. 2012;488(7412):522–526.
Stem Cells, Cell Differentiation, and Cancer  •  CHAPTER 7 107.e3
107.e3

127. Guzman ML, Neering SJ, Upchurch D, et al. Nuclear 134. Willingham SB, Volkmer JP, Gentles AJ, et al. The 141. Chao MP, Alizadeh AA, Tang C, et al. Anti-CD47
factor-kappaB is constitutively activated in primitive CD47-signal regulatory protein alpha (SIRPa) inter- antibody synergizes with rituximab to promote
human acute myelogenous leukemia cells. Blood. action is a therapeutic target for human solid tumors. phagocytosis and eradicate non-Hodgkin lymphoma.
2001;98(8):2301–2307. Proc Natl Acad Sci USA. 2012;109(17):6662– Cell. 2010;142(5):699–713.
128. Bao S, Wu Q, McLendon RE, et al. Glioma 6667. 142. Chao MP, Jaiswal S, Weissman-Tsukamoto R, et al.
stem cells promote radioresistance by preferential 135. Oldenborg PA, Zheleznyak A, Fang YF, Lagenaur Calreticulin is the dominant pro-phagocytic signal
activation of the DNA damage response. Nature. CF, Gresham HD, Lindberg FP. Role of CD47 on multiple human cancers and is counterbalanced
2006;444(7120):756–760. as a marker of self on red blood cells. Science. by CD47. Sci Transl Med. 2010;2(63):63ra94.
129. Phillips TM, McBride WH, Pajonk F. The response 2000;288(5473):2051–2054. 143. Feng M, Chen JY, Weissman-Tsukamoto R, et al.
of CD24(−/low)/CD44+ breast cancer-initiating 136. Lagasse E, Weissman IL. bcl-2 inhibits apoptosis Macrophages eat cancer cells using their own
cells to radiation. J Natl Cancer Inst. 2006;98(24): of neutrophils but not their engulfment by mac- calreticulin as a guide: roles of TLR and Btk. Proc
1777–1785. rophages. J Exp Med. 1994;179(3):1047–1052. Natl Acad Sci USA. 2015;112(7):2145–2150.
130. Diehn M, Cho RW, Lobo NA, et al. Association of 137. Chao MP, Majeti R, Weissman IL. Programmed cell 144. Brown JM. NCI’s anticancer drug screening program
reactive oxygen species levels and radioresistance in removal: a new obstacle in the road to developing may not be selecting for clinically active compounds.
cancer stem cells. Nature. 2009;458(7239):780–783. cancer. Nat Rev Cancer. 2011;12(1):58–67. Oncol Res. 1997;9(5):213–215.
131. Lee Y, Shin JH, Longmire M, et al. CD44+ cells in 138. Jaiswal S, Jamieson CH, Pang WW, et al. CD47 145. Quintana E, Shackleton M, Sabel MS, Fullen
head and neck squamous cell carcinoma suppress is upregulated on circulating hematopoietic stem DR, Johnson TM, Morrison SJ. Efficient tumour
T-cell-mediated immunity by selective constitutive cells and leukemia cells to avoid phagocytosis. Cell. formation by single human melanoma cells. Nature.
and inducible expression of PD-L1. Clin Cancer Res. 2009;138(2):271–285. 2008;456(7222):593–598.
2016;22(14):3571–3581. 139. Majeti R, Chao MP, Alizadeh AA, et al. CD47 is an 146. Zomer A, Ellenbroek SI, Ritsma L, Beerling E,
132. Topalian SL, Taube JM, Anders RA, Pardoll DM. adverse prognostic factor and therapeutic antibody Vrisekoop N, Van Rheenen J. Intravital imaging
Mechanism-driven biomarkers to guide immune target on human acute myeloid leukemia stem cells. of cancer stem cell plasticity in mammary tumors.
checkpoint blockade in cancer therapy. Nat Rev Cell. 2009;138(2):286–299. Stem Cells. 2013;31(3):602–606.
Cancer. 2016;16(5):275–287. 140. Chao MP, Alizadeh AA, Tang C, et al. Thera- 147. Cho RW, Wang X, Diehn M, et al. Isolation and
133. Li X, Lewis MT, Huang J, et al. Intrinsic resistance peutic antibody targeting of CD47 eliminates molecular characterization of cancer stem cells in
of tumorigenic breast cancer cells to chemotherapy. human acute lymphoblastic leukemia. Cancer Res. MMTV-Wnt-1 murine breast tumors. Stem Cells.
J Natl Cancer Inst. 2008;100(9):672–679. 2011;71(4):1374–1384. 2008;26(2):364–371.
107.e4 Part I: Science and Clinical Oncology

SELF-ASSESSMENT REVIEW QUESTIONS c. The development of a diagnostic bone scan test, based on
immunoscintigraphy using anti-CD44 monoclonal
1. What is self-renewal? antibodies labeled with radioactive isotopes, could aid in the
a. The capacity of selected cells to naturally undergo a process early identification of bone metastases.
of full epigenetic reprogramming, which causes loss of d. The development of a diagnostic assay based on PCR
differentiation identity and reversal to a totipotent stem (polymerase chain reaction) and aimed at measuring CD44
cell state mRNA expression could aid in the early identification of
b. A fundamental property of stem cells, which consists of the lymph node metastases.
capacity to divide and give rise to functionally identical stem 5. Which important implication might cancer stem cells have for
cells, with intact long-term expansion, proliferation, and the future development of antitumor drugs?
differentiation potential a. The development of novel antitumor drugs could be based
c. The capacity possessed by stem cells to induce, when on finding agents able to arrest differentiation processes
damaged, the dedifferentiation of neighboring daughter cells, within the tumor mass.
thus creating new stem cells and renewing the stem cell pool b. To overcome tumor resistance to radiotherapy, new
d. A unique property possessed by mesenchymal stem cells, radiosensitizing agents should be able to selectively protect
defined by the capacity to actively migrate out of a specific nontumorigenic cancer cells during irradiation, thus creating
tissue and relocate into a new one, where they can change a darwinian competition between cancer stem cells and their
their epigenetic identity and acquire the function of other nontumorigenic progeny.
types of resident stem cells c. The development of novel antitumor drugs could be based
2. What is the definition of cancer stem cells? exclusively on in vitro experiments performed on cell lines
a. Normal hematopoietic stem cells that have migrated inside similar to cancer stem cells, thus eliminating the need for in
the tumor mass as a result of the strong inflammatory vivo animal models.
processes usually observed in cancer d. To permanently eradicate cancer tissues and abolish the risk
b. Embryonic stem cells that have been artificially transformed of tumor relapse, novel antitumor agents must be able to
in vitro by means of transduction with retroviral vectors eliminate self-renewing cancer stem cells.
encoding the NRAS oncogene and endowed with high
tumorigenic capacity in immunodeficient mice
c. A subpopulation of cancer cells, often a minority subset,
identified by a specific surface marker phenotype and ANSWERS
defined by the capacity to self-renew and differentiate,
as assessed with serial transplantation experiments in 1. (b)  Self-renewal is a defining and intrinsic property of all types
immunodeficient mice of stem cells. As opposed to most other cell types, which
d. Fusion hybrids between mesenchymal stem cells and cancer progressively exhaust their long-term expansion potential during
cells, endowed with tumorigenic capacity and high sequential rounds of cell division (i.e., they undergo senescence),
metastatic potential in immunodeficient mice stem cells are able to maintain their capacity for unlimited
3. To what kind of normal cell type do cancer stem cells proliferation (i.e., they self-renew). The capacity to self-renew is
correspond, in terms of surface marker phenotype? dependent on the capacity of stem cells to retain over time a
a. They are always characterized by a surface marker profile specific epigenetic identity, rather than to change or transform it.
found exclusively in normal stem cells. 2. (c)  The term cancer stem cells (CSCs) is an operational
b. They are always characterized by a surface marker profile definition (i.e., it is based on the experimental fulfillment of a
found exclusively in a specific lineage of terminally specific set of functional properties). It identifies a specific
differentiated, effector cells. subset of cancer cells that are able to self-renew (i.e., to form
c. Their surface marker profile is usually typical of immature or tumors that can be serially passaged in vivo) and to differentiate
progenitor cells and can correspond to that of stem cells (i.e., to form tumors that also contain other types of
and/or immature multipotent, oligopotent, or lineage- nontumorigenic cancer cells). These cells are usually identified
restricted progenitors, depending on the tumor subtype and based on a specific repertoire of surface markers and frequently
the disease stage. represent a minority subset of the neoplastic clone. Although
d. Their surface marker profile is usually typical of terminally they display stem cell properties (i.e., self-renewal and
differentiated effector cells and can correspond to that of a differentiation), their molecular and phenotypic identity does
specialized cellular lineage, depending on the tumor subtype not necessarily correspond to that of normal stem cells.
and the disease stage. 3. (c)  It is important to remember that cancer stem cells is an
4. Which important implication might cancer stem cells have for operational definition (see explanation of question 2). As such,
the future development of novel diagnostic and/or prognostic this term does not necessarily define a cell population whose
assays in cancer patients? phenotypic or molecular identity is always the same as that of
a. The analysis by gene-expression array of a bulk tumor’s normal stem cell populations. As a general rule, the surface
transcriptional profile, and the identification of a high degree marker phenotype of cancer stem cells corresponds to that of
of similarity with a cancer stem cell gene-expression immature progenitor cells and can encompass both stem cells
signature, could be used to identify patients characterized by and various types of multipotent, oligopotent, or lineage-
a more aggressive disease, associated with poor prognosis and restricted progenitors, depending on the tumor subtype and the
worse clinical outcomes. stage of the disease.
b. The presence of a low percentage of cancer stem cells 4. (a)  Several studies have shown that malignant tumors
within cancer tissues (<30%), as evaluated with characterized by a gene-expression profile similar to that of
immunohistochemistry for CD44, could be used to cancer stem cells (CSCs) are associated with worse clinical
discriminate between benign and malignant tumors across outcomes. A high degree of similarity between a whole tumor’s
different tissue types (e.g., between a teratoma and a gene-expression profile and that of CSCs is likely the
teratocarcinoma, a benign and a malignant mole, and an consequence of a high CSC content in the tumor. In many
adenoma and a carcinoma). epithelial carcinomas, CD44 can be used, in combination with
Stem Cells, Cell Differentiation, and Cancer  •  CHAPTER 7 107.e5
107.e5

a panel of other markers, to isolate a subpopulation of cancer hold the potential to lead to tumor relapse. To permanently
cells enriched in CSCs. This is likely because, within many eradicate tumor tissues, antitumor therapies must be able to kill
epithelial tissues, CD44 is expressed in a gradient, with the or otherwise eliminate CSCs. From a theoretical perspective, a
highest levels observed in the most basal layers, where immature possible strategy to deplete tumor tissues of CSCs could be to
stem and progenitor cells usually reside. CD44, however, is not promote their differentiation into terminally mature cells, not to
specific to epithelial stem and progenitor cells and is expressed inhibit or prevent it. As a general rule, nontumorigenic cancer
abundantly across many other cell types, including stromal and cells are the progeny of CSCs, not a different genetic clone of
immune cells (e.g., fibroblasts, endothelial cells, monocytes, the cancer tissue. Thus nontumorigenic cancer cells originate
lymphocytes). Expression of CD44 cannot be used to purify from CSCs and are not in “Darwinian competition” with them.
and/or identify CSCs across all tumors, especially nonepithelial The dynamic equilibrium between CSCs and other cellular
tumors. Even within epithelial tumors, expression of CD44 components of cancer tissues is extremely difficult to model in
alone (e.g., without careful exclusion of the contribution of vitro using traditional cell lines grown as two-dimensional
stromal cells) cannot be used to accurately define CSCs. monolayers. Currently, CSCs can be best, if not exclusively,
5. (d)  Being able to self-renew, cancer stem cells (CSCs) are able studied using solid tissue xenografts in animal models.
to propagate themselves indefinitely. If not eliminated, they
8  Tumor Microenvironment: Vascular and
Extravascular Compartment
Rakesh K. Jain, John D. Martin, Vikash P. Chauhan, and Dan G. Duda

S UMMARY OF K EY P OI N T S
• A solid tumor is an organ composed contribute to heterogeneity in proliferating cancer cells. Interstitial
of neoplastic cells and stromal cells vascular permeability, blood flow, hypertension cannot compress leaky
including immune cells nourished by and the microenvironment. vessels, because the intravascular
vasculature made of endothelial • Tumor interstitial matrix is formed of and extravascular fluid pressures
cells—all embedded in an proteins secreted by host and tumor equilibrate.
extracellular matrix. The interactions cells and by those leaked from the • Judicious application of angiogenic
among these cells, their surrounding nascent blood vessels. therapy can normalize the tumor
matrix, and their local • Tumor interstitium is heterogeneous, vessels and make them more
microenvironment control the with some regions fairly permeable efficient for delivery of oxygen (a
expression of various genes. The and others difficult to penetrate. known radiosensitizer) and drugs.
products encoded by these genes, in Modification of collagen and Indeed, clinical studies indicate that
turn, control the pathophysiologic hyaluronan in the matrix can improve antiangiogenic agents can increase
characteristics of the tumor. Tumor penetration of large-molecular-weight perfusion and oxygenation in tumors,
pathophysiology governs not only therapeutics. and that these effects are associated
tumor growth, invasion, and • Solid components of tumors—cancer with improved response.
metastasis but also the response to cells, stromal cells, and matrix • Twelve antiangiogenic agents have
various therapies. molecules—mechanically compress been approved for cancer. Two main
• Tumor vasculature is made of host blood and lymphatic vessels, goals to improve patient outcomes
vessels co-opted by tumor cells and resulting in reduced perfusion that with antiangiogenic agents are the
by new vessels formed by the limits oxygen and drug supply. development of therapies that fortify
processes of vasculogenesis and Reducing these constituents vessels without pruning and
angiogenesis. A constellation of decompresses blood vessels verification of biomarkers predictive
positive and negative regulators of to enhance perfusion and drug of response. Additionally, the
angiogenesis governs the process delivery. potential of combining antiangiogenic
of neovascularization. • Interstitial hypertension results from and solid stress–alleviating therapies
• Tumor vessels are abnormal in terms vessel leakiness, lack of functional with immunotherapies should be
of their organization, structure, and lymphatics, and compression of confirmed in clinical trials.
function. These abnormalities blood and lymphatic vessels by

A solid tumor is an organ composed of neoplastic cells and host VASCULAR COMPARTMENT
stromal cells including resident and transiting immune cells nourished
by the vasculature made of endothelial cells (ECs)—all embedded in Neoplastic cells, like normal cells, need oxygen and other nutrients
an extracellular matrix (Fig. 8.1). The interactions among these cells for their survival and growth. Every normal cell in the human body
and between these cells, their surrounding matrix, and their local is located within 100 to 200 µm of a blood capillary so that it can
microenvironment, control the expression of various genes. The products receive oxygen and other nutrients by the process of diffusion. Likewise,
encoded by these genes, in turn, control the pathophysiologic char- cells undergoing neoplastic transformation depend on nearby capillaries
acteristics of the tumor. Tumor pathophysiology governs not only for growth. These preneoplastic (i.e., hyperplastic or dysplastic) cells
tumor growth, invasion, and metastasis but also the response to various can grow as a spherical or ellipsoidal cellular aggregate. Once the size
therapies. In this chapter we will discuss various pathophysiologic of the cellular aggregate reaches the diffusion limit for critical nutrients
parameters that characterize the vascular and extravascular compartments and oxygen, however, the aggregate as a whole can become dormant.
of a tumor and the mechanisms governing the formation and function Indeed, human tumors can remain dormant for many years because
of these compartments. of a balance between neoplastic cell proliferation and apoptosis.

108
Tumor Microenvironment: Vascular and Extravascular Compartment  •  CHAPTER 8 109

However, once they have access to new blood vessels, they may grow New Vessel Formation
and metastasize. What triggers the growth of new vessels? What
molecular and cellular mechanisms are involved? How do these vessels It has been known for nearly a century that the vascular system is
compare with normal vessels with respect to structure and function? associated with tumor growth in animals and humans.1 Powerful
Can we prevent or delay tumor progression only by interfering with insights into the neovascularization of transplanted tumors using the
the neovascularization process? transparent window techniques were developed in the 1940s.2–5 The
possibility that tumors produce an “angiogenic” substance was suggested
in 1968.6 The hypothesis that blocking angiogenesis should block
tumor growth and metastasis was proposed shortly thereafter in 1971.7
The concept that a tissue acquires angiogenic capacity during neoplastic
transformation—and, by extension, that antiangiogenesis could be
used to prevent cancer—was put forward in 1978.8 The first antian-
Blood vessel giogenic agent approved for cancer patients was bevacizumab, an
antibody specific to vascular endothelial growth factor (VEGF), on
Basement the basis of the increased survival seen in metastatic colorectal cancer
membrane patients with the combination of bevacizumab with standard chemo-
therapy in a pivotal randomized placebo-controlled phase III trial.9
At present, various antiangiogenesis and proangiogenesis strategies are
being evaluated clinically to prevent or treat a large number of diseases,
including cancer.10–13 Both normal and pathologic angiogenic processes
are governed by the net balance between proangiogenic and antian-
giogenic factors.14,15 This balance is spatially and temporally regulated
under physiologic conditions, so that the “angiogenic switch” is “on”
Interstitial matrix and fluid when needed (e.g., during embryonic development, wound healing,
formation of the corpus luteum) and “off ” at other times. During
neoplastic transformation and tumor progression, this regulation is
deranged, and blood vessels form ectopically to support a growing
Cancer cells Host cells tumor mass.
Cellular Mechanisms
Figure 8.1  •  Schematic representation of a solid tumor. The key components
include cancer cells, host cells, and vasculature made of endothelial cells—all At least four cellular mechanisms are involved in the vascularization
embedded in a matrix bathed in interstitial fluid. Arrows indicate interactions of tumors: co-option, intussusception, sprouting (angiogenesis), and
between the components. (Modified from Jain RK. Angiogenesis and lym- vasculogenesis (Fig. 8.2).12 Tumor cells can co-opt and grow around
phangiogenesis in tumors: insights from intravital microscopy. Cold Spring existing vessels to form “perivascular” cuffs. However, as stated earlier,
Harbor Symp Quant Biol [The Cardiovascular System]. 2002;67:239–248.) these cuffs cannot grow beyond the diffusion limit of critical nutrients

Endothelial precursor

Intussusceptive
growth
Angiogenic sprouting

Figure 8.2  •  Cellular mechanisms of vascularization in tumors. At least four mechanisms are involved: (1) intussusception, wherein tumor vessels enlarge
and an interstitial tissue column grows in the enlarged lumen, expanding the network; (2) vasculogenesis, wherein endothelial precursor cells mobilized from
the bone marrow or peripheral blood contribute to the endothelial lining of tumor vessels; (3) “sprouting” angiogenesis, wherein the existing vascular network
expands by forming sprouts or bridges; and (4) co-option (not shown), wherein tumor cells grow around existing vessels to form “perivascular” cuffs. (Modified
from Jain RK, Carmeliet PF. Angiogenesis in cancer and other diseases. Nature. 2000;407:249–257.)
110 Part I: Science and Clinical Oncology

and may actually cause the collapse of the vessels owing to growth metabolic stress (e.g., low Po2, low pH, or hypoglycemia), mechanical
pressure (referred to as “solid stress”). Alternatively, an existing vessel stress (e.g., shear stress, solid stress), immune or inflammatory cells
may enlarge in response to the growth factors released by tumors, and that have infiltrated the tissue, and genetic mutations (e.g., activation
an interstitial tissue column may grow in the enlarged lumen and of oncogenes or deletion of suppressor genes that control the production
partition the lumen to form an expanded vascular network. This mode of angiogenesis regulators).14,15,45–48 These molecules can emanate from
of intussusceptive microvascular growth has been observed during cancer cells, ECs, stromal cells, blood, and extracellular matrix (Fig.
tumor growth, wound healing, and gene therapy.16–19 “Sprouting” 8.3).49–52 Because the normal host cells differ among organs, the detailed
angiogenesis is perhaps the most widely studied mechanism of vessel mechanisms of angiogenesis might depend on the specific host-tumor
formation. During sprouting angiogenesis, the existing vessels become interactions operating within a given tissue.53–60 Furthermore, because
leaky in response to growth factors released by normal cells or cancer the tumor microenvironment is likely to change during tumor growth,
cells; the basement membrane and the interstitial matrix dissolve; regression, and relapse, profiles of proangiogenic and antiangiogenic
pericytes dissociate from the vessel; ECs migrate and proliferate to molecules are likely to change with time and space.61,62 The challenge
form an array or sprout; a lumen is formed in the sprout (a process currently is to develop a unified conceptual framework to describe
referred to as canalization); branches and loops are formed by confluence the temporal and spatial profiles of this increasingly diverse array of
and anastomoses of sprouts to permit blood flow; and finally, these angiogenesis regulators with the aim of developing effective therapeutic
immature vessels are invested with basement membrane and pericytes. strategies.38,63–65
During physiologic angiogenesis, these vessels differentiate into mature
arterioles, capillaries, and venules, whereas in tumors they remain Vascular Architecture
largely immature.5,12,13,20 During embryonic development, a primitive
vascular plexus is formed from endothelial precursor cells (EPCs, also In a normal tissue, blood flows from an artery to arterioles to capillaries
known as angioblasts) by a process referred to as vasculogenesis. In to venules to a vein. Although the tumor vasculature originates from
adults, EPCs—mobilized from bone marrow niches into the peripheral these host vessels and the mechanisms of angiogenesis are similar, its
blood circulation—can also contribute to neovascularization (a process organization may differ dramatically, depending on the tumor type, its
referred to as postnatal vasculogenesis) in tumors and other tissues.21–23 location, and whether it is growing, regressing, or relapsing.16,66–69 In
In addition, reports have demonstrated that a fraction of ECs in tumor general, tumor vessels are dilated, saccular, tortuous, and chaotic in their
vessels may be derived via transdifferentiation from cancer cells,24 or patterns of interconnection.70 For example, whereas normal vasculature
from cancer stem-like cells (e.g., in glioblastomas [GBMs]).12,25–27 In is characterized by dichotomous branching, tumor vasculature has
this larger context, the term vasculogenesis might be invoked to describe many trifurcations and branches with uneven diameters.71,72 The
neovascularization in tumors from tumor stem cells.28 Tumor vascu- fractal dimensions and minimum path lengths of tumor vasculature
logenesis may also occur from EPCs of non–bone marrow origin that are different from those of normal host vasculature.66–68 The molecular
are recruited from adjacent tissues and/or circulation.29,30 The current mechanisms of this abnormal vascular architecture are not understood,
challenge is to discern the relative contribution of each of these but it seems reasonable to hypothesize that the imbalance of VEGF
mechanisms of neovascularization during tumor growth, and during and angiopoietins is a key contributor.20,73 In mice, “normalization”
and after treatment of tumors.28,31 of the tumor vasculature observed during treatment with therapies
that reduce VEGF (e.g., hormone withdrawal from a hormone-
Molecular Mechanisms dependent tumor), interfere with VEGF signaling (e.g., treatment
Various proangiogenic and antiangiogenic molecules that orchestrate with anti-VEGF or anti-VEGFR2 antibody; Fig. 8.4), or mimic an
different steps in vessel formation, along with their functions, are antiangiogenic cocktail (e.g., trastuzumab [Herceptin] treatment of
listed in Table 8.1. VEGF is currently considered the most critical a HER2-overexpressing tumor) is in concert with this molecular
proangiogenic molecule. Originally discovered in 1983 as the vascular hypothesis.62,64,74–77 Mechanical stress generated by proliferating cancer
permeability factor and cloned in 1989, VEGF increases vascular and stromal cells and their excessive interstitial matrix production also
permeability, promotes migration and proliferation of ECs, serves as can lead to the partially compressed or totally collapsed vessels often
an EC survival factor, can mobilize EPC populations from the bone found in tumors (Fig. 8.5A–B).65,78,79 The decompression of blood
marrow, and is known to upregulate leukocyte adhesion molecules vessels observed after induction of apoptosis in perivascular cells or
on ECs.16,23,32–34 During tumor progression, or with treatment, the stromal cells, or enzymatic degradation of matrix hyaluronan, supports
number of distinct angiogenic molecules produced by a tumor can this mechanical hypothesis (Fig. 8.5C).79–82 Perhaps the combination of
increase.35–37 Thus, after VEGF signaling is blocked, a tumor might both molecular and mechanical factors renders the tumor vasculature
rely on other, alternative angiogenic molecules (e.g., basic fibroblast abnormal, and thus both types of factors must be taken into account
growth factor [bFGF], stromal-derived factor 1α [SDF1α], placental- in designing novel strategies for cancer treatment.
derived growth factor [PlGF], or interleukin-8 [IL-8]).38 Other positive
regulators of angiogenesis include the angiopoietins that are involved Blood Flow and Microcirculation
in stabilizing vessels and controlling vascular permeability; various
proteases involved in dissolving and remodeling matrix and releasing Blood flow in a vascular network, whether normal or abnormal, is
growth factors; and organ-specific angiogenic stimulators (e.g., endocrine governed by the arteriovenous pressure difference and flow resistance.
gland VEGF).20,39,40 Angiogenesis inhibitors include endogenous soluble Flow resistance is a function of the vascular architecture (referred to as
receptors of various proangiogenic ligands (e.g., sVEGFR1/sFLT1) geometric resistance) and of the blood viscosity (rheology, referred to
and molecules that downregulate the expression of stimulators (e.g., as viscous resistance).70 Abnormalities in both vasculature and viscosity
interferons) or that interfere with the release of the stimulators or increase the resistance to blood flow in tumors.72,83–85 As a result, overall
binding with their receptors (e.g., platelet factor 4). Thrombospondins perfusion rates (blood flow rate per unit volume) in tumors are lower
are among the first and best characterized endogenous inhibitors that than in many normal tissues.86,87 Both macroscopically and microscopi-
interfere with the growth, adhesion, migration, and survival of ECs.14 cally, tumor blood flow is temporally and spatially chaotic. Macroscopi-
Other endogenous inhibitors include fragments of various plasma or cally, four spatial regions can be recognized in a tumor (Fig. 8.6):
matrix proteins (e.g., angiostatin, a fragment of plasminogen; endostatin,
a fragment of collagen XVIII; tumstatin, a fragment of collagen IV).41–43 1. An avascular necrotic region
Neither the mechanisms of action of the matrix-derived inhibitors 2. A seminecrotic region
nor their physiologic role is well understood.44 The generation of 3. A stabilized microcirculation region
proangiogenic and antiangiogenic molecules can be triggered by 4. An advancing front88,89
Tumor Microenvironment: Vascular and Extravascular Compartment  •  CHAPTER 8 111

Table 8.1  Angiogenesis Activators and Inhibitorsa


Activators Function Inhibitors Function
VEGF family membersb,c Stimulate angiogenesis and VEGFR1; soluble VEGFR1; Sink for VEGF, VEGF-B, PlGF
vasculogenesis, permeability, soluble neuropilin-1 (NRP-1)
leukocyte adhesion
VEGFRc, NRP-1, NRP-2 Integrate angiogenic and survival Ang 2b,c Antagonist of Ang 1
signals
EG-VEGF Stimulate growth of endothelial cells TSP-1,2 Inhibit endothelial migration,
derived from endocrine glands growth, adhesion, and survival
Ang 1 and Tie 2b,c Stabilize vessels Angiostatin and related Inhibit endothelial migration and
plasminogen kringles survival
PDGF-BB and receptors Recruit smooth muscle cells Endostatin (collagen XVIII Inhibit endothelial survival and
fragment) migration
TGF-β1d, endoglin, TGF-β Stimulate extracellular matrix Tumstatin (collagen IV Inhibit endothelial protein
receptors production fragment) synthesis
FGF, HGF, MCP-1 Stimulate angio/arteriogenesis Vasostatin; calreticulin Inhibit endothelial growth
Integrins αvβ3, αvβ5, α5β1 Receptors for matrix macromolecules Platelet factor 4 Inhibit binding of bFGF and VEGF
and proteinases
VE-cadherin; PECAM (CD31) Endothelial junctional molecules Tissue inhibitors of MMP Suppress pathologic angiogenesis
(TIMPs); MMP inhibitors; PEX
Ephrinsc Regulate arterial and venous Meth-1; Meth-2 Inhibitors containing MMP-, TSP-,
specification and disintegrin domains
Plasminogen activators, MMPs Remodel matrix, release growth factor IFN-α, IFN-β, IFN-γ; IP-10, Inhibit endothelial migration;
IL-4, IL-12, IL-18 downregulate bFGF
PAI-1 Stabilize nascent vessels Prothrombin kringle-2; Suppress endothelial growth
antithrombin III fragment
NOS; COX-2 Stimulate angiogenesis and 16 kD-prolactin Inhibit bFGF/VEGF
vasodilation
AC133 Regulate angioblast differentiation VEGI Modulate cell growth
Chemokinesd Pleiotropic role in angiogenesis Fragment of SPARC Inhibit endothelial binding and
activity of VEGF
Id1/Id3 Inhibit differentiation Osteopontin fragment Interfere with integrin signaling
Maspin, canstatin, Protease inhibitor
proliferin-related protein, Mechanisms unknown
restin

a
Selected list updated from Carmeliet P, Jain RK. Molecular mechanisms and clinical applications of angiogenesis. Nature. 2011;473:298–30712; for complete function and
references, see supplementary information (http://steele.mgh.harvard.edu).
b
Also present in or affecting nonendothelial cells.
c
See Jain.20
d
Opposite effect in some contexts.
COX-2, Cyclooxygenase-2; EG-VEGF, endocrine gland–derived vascular endothelial growth factor; bFGF, basic fibroblast growth factor; HGF, hepatocyte growth factor; IFN,
interferon; IL, interleukin; IP, interferon-inducible protein; MCP-1, monocyte chemotactic protein-1; MMP, metalloproteinase; NRP-1, neuropilin-1; NOS, nitric oxide synthase; PAI-1,
plasminogen activator inhibitor-1; PDGF-BB, platelet-derived growth factor-BB; PECAM, platelet endothelial cell adhesion molecule; PEX, hemopexin fragment of MMP-2; PlGF,
placental growth factor; SPARC, secreted protein acidic and rich in cysteine; TGF, transforming growth factor; TIMP, tissue inhibitor of metalloproteinase; TSP, thrombospondin;
VEGF, vascular endothelial growth factor; VEGFR, vascular endothelial growth factor receptor; VEGI, vascular endothelial growth inhibitor. See text for explanation of
abbreviations.

At the microscopic level, in normal tissues, erythrocyte velocity is chronic hypoxia in tumors—a major cause of resistance to radiation
dependent on vessel diameter, but there is no such dependence in and other therapies. Increased perfusion also correlates with a positive
most tumors.55,60,90 Furthermore, the average erythrocyte velocity can response to radiotherapy or chemotherapy and patient survival.95,96
be an order of magnitude lower in some tumors as compared with Considerable effort has gone into increasing tumor blood flow for
that of normal host tissue (Fig. 8.7).60 In a given vessel within a tumor, improving radiation therapy, or decreasing tumor perfusion in the
blood flow fluctuates with time and can reverse its direction.88,90,91 In case of hyperthermia. This has been difficult to achieve reproducibly,
addition to the elevated geometric and viscous (rheologic) resistance, because tumor vasculature consists of both vessels co-opted from the
other molecular and mechanical factors contribute to this spatial and preexisting host vasculature and vessels resulting from the angiogenic
temporal heterogeneity. These include imbalance between proangiogenic response of host vessels to cancer cells. The former are invested in
and antiangiogenic molecules, solid stress generated by proliferating normal contractile perivascular cells, whereas the latter lack these
tumor cells and matrix production, vascular remodeling by intussuscep- perivascular cells or these cells are abnormal.49,70,97 Presumably as a
tion, and coupling between luminal and interstitial fluid pressure result, efforts to increase the tumor blood flow with pharmacologic
(IFP) via hyperpermeability of tumor vessels.6,17,65,66,78–80,92–94 As will or physical agents have not always been reproducible or success-
be discussed later, this heterogeneity contributes to both acute and ful.49,70,86,97 On the other hand, the strategy of decreasing or shutting
112 Part I: Science and Clinical Oncology

A B

C D E
100 µm 100 µm 50 µm

Figure 8.3  •  Tumor induction of host promoter activity in stromal cells. The expression of vascular endothelial growth factor (VEGF) in host cells can
be examined through use of transgenic mice expressing a green fluorescent protein (GFP) under the control of the VEGF promoter. (A) A murine mammary
carcinoma xenograft shows host cell VEGF expression mainly at the periphery of the tumor after 1 week. (B) After 2 weeks, the VEGF-expressing host cells
have infiltrated the tumor. (C) A GFP-expressing layer of host cells can be seen at the tumor-host interface. (D–E) The VEGF-expressing host cells colocalize
with the angiogenic tumor vessels. ([A–B] From Fukumura D, Xavier R, Sugiura T, et al. Tumor induction of VEGF promoter activity in stromal cells. Cell.
1998;94:715–725. [C–E] From Brown EB, Campbell RB, Tsuzuki Y, et al. In vivo measurement of gene expression, angiogenesis and physiological function
in tumors using multiphoton laser scanning microscopy. Nat Med. 2001;7:1069.)

down the tumor blood flow—by “stealing” blood away from the interendothelial junctions, increased numbers of fenestrations, vesicles,
“passive component” of the tumor vasculature with vasodilators, through and vesicovacuolar channels in tumor vessels, and a lack of normal
vascular targeting, or by means of intravascular coagulation—has shown basement membrane and pericytes.16,52,97,103,104 In concert with these
promise in experimental systems.86,87,98–100 It also appears that judiciously ultrastructural findings, both vascular permeability and hydraulic
applied antiangiogenic therapy could “normalize” the abnormal tumor conductivity (a measure of water movement by pressure gradient)
microcirculation by pruning the immature vessels (see Fig. 8.4), of tumors, in general, are significantly higher than those for various
thus rendering the remaining vasculature more responsive to normal tissues.60,105–109 Furthermore, unlike normal vessels in the
vasoactive agents.101 surrounding tissue, tumor vessels partially lose molecular size–based
selectivity in permeability to different molecules.110 Despite increased
VASCULAR PERMEABILITY overall permeability, not all blood vessels of a tumor are leaky (Fig.
8.8A). Even the leaky vessels have a finite pore size that is tumor
Once a blood-borne molecule has reached an exchange vessel, its dependent (Fig. 8.8B), and ultrastructural studies have shown that
extravasation occurs by diffusion, convection, and, to some extent, the larger pore size in tumors represents wide interendothelial junc-
presumably by transcytosis.101,102 The effective permeability, P, of tions.57,104 Thus permeability is inversely related to molecular size
a molecule depends on the size, shape, charge, and flexibility of because of steric and hydrodynamic hindrance from these pores, and
the molecule, and on the size, shape, charge, and dynamics of the very large molecules, such as nanoparticles, do not penetrate tumors
transvascular transport pathway. In normal vessels, these pathways efficiently.111,112 Molecular shape also influences penetration of these
include diffusion along the EC membrane (for lipophilic solutes), pores, such that rod-shaped molecules have a higher permeability than
trans-EC diffusion, interendothelial junctions (<7 nm), open or closed spherical molecules.113 Similarly, positively charged molecules have a
fenestrations (<10 nm), and transendothelial channels (including vesicles higher affinity for the negatively charged glycocalyx in the pores of these
or vesicovacuolar channels).16,103 Some of these anatomic pathways angiogenic tumor vessels, reducing this hindrance and giving them a
may be lined with glycocalyx on EC, thus effectively reducing the higher permeability.114–117 Not only do the vascular permeability and
size of the pathway. A basement membrane may further retard pore size vary from one tumor to the next (see Fig. 8.8A–C), but within
the movement of molecules. Ultrastructure studies show widened the same tumor they vary spatially and temporally, as they do during
Tumor Microenvironment: Vascular and Extravascular Compartment  •  CHAPTER 8 113

extravasation of cells. Very little is known about intravasation except


that tumors might shed more than a million cells per gram per day,
and most of these are not clonogenic.120–123 We have demonstrated
that metastatic cells could bring their own stroma from the primary
tumor in the form of heterotypic cell clumps.124 More is known about
the molecular and cellular mechanisms of extravasation.125 When a
Tumor Normalized cell enters a blood vessel, it can continue to move with the flowing
blood, collide with the vessel wall, adhere transiently or stably, and
finally extravasate. These interactions are governed by both local
hydrodynamic forces and adhesive forces. The former are determined
by the vessel diameter and fluid velocity, and the latter by the expression,
strength, and kinetics of bond formation between adhesion molecules
and by the surface area of contact.126–130 Deformability of cells affects
both types of forces.131 In addition, cancer cells may grow intravascularly
at the distant site (e.g., in the lungs).132
Rolling of endogenous leukocytes is generally low in tumor vessels,
whereas stable adhesion (≥30 seconds) is comparable between normal
vessels and tumor vessels.133 On the other hand, both rolling and
stable adhesion are nearly zero in angiogenic vessels induced in collagen
gels by bFGF or VEGF, two of the most potent angiogenic factors.134
Whether this observation is due to a low flux of leukocytes into
angiogenic vessels and/or to downregulation of adhesion molecules
in these immature vessels is currently not known. The age of the
animal also plays an important role in leukocyte-endothelial interac-
tions.135 Further insight into the types of cells that adhere to tumor
vessels comes from studies on the localization of IL-2–activated natural
Normal killer (A-NK) cells in normal and tumor tissues in mice in which
positron emission tomography was used.136,137 After systemic injection,
these cells localized primarily in the lungs immediately after injection
Inadequate and could not be detected in the tumor.136 Increased rigidity caused
by IL-2 activation might contribute to the mechanical entrapment of
these cells in the lung microcirculation.138,139 Constitutive expression
of certain adhesion molecules in the lung vasculature might also facilitate
their retention in the lungs.125 One approach to reducing lung entrap-
Figure 8.4  •  Normalization of tumor vasculature. Normal vessels are well ment is to reduce the rigidity of these cells.112,117 Alternatively, the
organized with even diameters. In contrast, tumor vessels are tortuous, with lung can be circumvented by injecting A-NK cells directly into the
increased vessel diameter, length, density, and permeability. Antiangiogenic blood supply of tumors. In this case, A-NK cells, both xenogeneic
therapies “normalize” the tumor vascular network and could ultimately reduce and syngeneic, adhered to some blood vessels in three different tumor
the vasculature to the point at which it provides inadequate support for oxygen
and drug delivery. (Modified from Jain RK. Normalizing tumor vasculature
models via CD18 and very large antigen 4 (VLA-4) on the A-NK
with anti-angiogenic therapy: a new paradigm for combination therapy. Nat cells and intercellular adhesion molecule 1 (ICAM-1), vascular cell
Med. 2001;7:987–989; Jain RK. Angiogenesis and lymphangiogenesis in adhesion molecule 1 (VCAM-1), and E-selectin on the activated
tumors: insights from intravital microscopy. Cold Spring Harbor Symp Quant endothelium of angiogenic vessels.34,137,140–143 These molecules can be
Biol [The Cardiovascular System]. 2002;67:239–248; Jain RK, Carmeliet PF. upregulated by tumor necrosis factor-α (TNF-α) and a protein of 90
Vessels of death or life. Sci Am. 2001;285:38.) kd molecular weight (p90) that is secreted by some neoplastic cells
and downregulated by transforming growth factor-β (TGF-β)—also,
presumably, secreted by cancer cells.56,126,144–148 Surprisingly, the
proangiogenic VEGF also can upregulate these molecules, whereas
tumor growth, regression, and relapse.57,61,62 The local microenvironment another proangiogenic molecule, bFGF, can downregulate these
plays an important role in controlling vascular permeability (see Fig. molecules.34,52,62,77,149 On the other hand, inflammatory cells such as
8.8D). For example, a human glioma (HGL21) has fairly leaky vessels monocytes and macrophages or neutrophils are also recruited by VEGF
when grown subcutaneously in immunodeficient mice, but it exhibits and may play important roles in promoting matrix remodeling and
blood-brain barrier properties in the brain.60,73 Such site-dependent angiogenesis.150 The challenge currently is to decrease nonspecific
differences for other tumors have been observed in other orthotopic entrapment of immune cells in normal vessels and to increase their
sites.55,58,59 One possible explanation is that the host-tumor interactions delivery to tumor vessels to improve various cell-based therapies,
control the production and secretion of cytokines associated with including gene therapy.
permeability increase (e.g., VEGF) and decrease (e.g., angiopoietin
1).20,40,73,118,119 A better understanding of the molecular mechanisms
of permeability regulation in tumors is likely to yield strategies for EXTRAVASCULAR COMPARTMENT
improved delivery of molecular medicine to tumors. Composition and Origin

Movement of Cells Across Vessel Walls The extravascular compartment of a solid tumor consists of neoplastic
cells (parenchyma) and host cells (e.g., inflammatory cells, fibroblasts)
Both cancer cells and immune cells frequently move across the walls residing in an interstitial matrix bathed by the interstitial fluid (see
of blood vessels—the former in the process of metastasis, and the Fig. 8.1). Depending on the tumor type and its stage of differentiation,
latter during immune response or cell-based immunotherapy. Both neoplastic cells might be dispersed in the matrix as individual cells
transendothelial (through ECs) and periendothelial (between ECs) (e.g., lymphomas, melanomas) or as clumps, sheets, or nests (e.g.,
pathways have been proposed as a route for intravasation and carcinomas). More than 80% of tumors are carcinomas arising from
114 Part I: Science and Clinical Oncology

DEPLETE CANCER CELLS DEPLETE FIBROBLASTS

Cancer cell
Fibroblast
Open blood vessel
Partially open blood vessel
Collapsed vessel
Collagen
Stretched Collagen
HA
Compressed HA

DEPLETE COLLAGEN DEPLETE HYALURONAN

Intact Deformed

Untreated Solid stress alleviated

C Collagen Perfused vessel

Figure 8.5  •  Causes and remedies of vascular compression in tumors. (A) Middle, In solid tumors, proliferating cancer cells and activated fibroblasts
deform the interstitial matrix, resulting in stretched collagen fibers, compressed hyaluronan, and deformed cells—all storing solid stress, a type of mechanical
force transmitted by solid tissue components. This stress compresses intratumor blood and lymphatic vessels. Potential strategies to alleviate solid stress and
decompress vessels involve depleting these components. Depleting cancer cells (top left) or fibroblasts (top right) relaxes collagen fibers, hyaluronan, and the
remaining cells, alleviating solid stress. Depleting collagen (bottom left) alleviates the stress that was held within these fibers while also relaxing stretched/
activated fibroblasts and compressed cancer cells within nodules. Finally, depleting hyaluronan (bottom right) alleviates the stored compressive stress, allowing
nearby components to decompress. (For simplification of the schematic, other stromal cells, such as pericytes, macrophages, and various immune cells that
might also control production of collagen or hyaluronan, are not shown. Lymphatic vessels are also not shown for the same reason.) (B) Image of an intact
(left) and deformed (right) soft tissue sarcoma freshly excised from a patient. The intact tumor is free of external loads. Cutting through the long axis of the
tumor allows the tissue to deform, which demonstrates the existence of solid stress held within tumor tissue. (C) Intravital microscopy images of a murine
breast tumor before (left) and after (right) treatment to reduce solid stress. The treatment reduces the density of collagen fibers (blue) leading to a homogenous
distribution of perfused vessels (green). ([A–B] From Stylianopoulos T, Martin JD, Chauhan VP, et al. Causes, consequences, and remedies for growth-induced
solid stress in murine and human tumors. Proc Natl Acad Sci U S A. 2012;109:15101–15108. [C] From Chauhan VP, Martin JD, Liu H, et al. Angiotensin
inhibition enhances drug delivery and potentiates chemotherapy by decompressing tumour blood vessels. Nat Commun. 2013;4:2516.)
Tumor Microenvironment: Vascular and Extravascular Compartment  •  CHAPTER 8 115

e
ur

H
ss

rp
re

y
er
n
la
p O ia l p

io

liv
lu
on

at
l

de
ce
tit

er
si

rs
fu

tra

lif

g
te

ru
r

o
2
Tumor region

Pe

Ex

Pr
In

D
Proliferative ++ + ++ <7.4 ++ ++
Quiescent + +++ + ~ 6.5-7 +/– +
Necrotic – +++ – ~ 7-8 – +/–

Figure 8.6  •  The tumor microenvironment is heterogeneous with proliferative, quiescent, and necrotic regions. These regions can be characterized in
terms of various physiologic parameters. Decreasing magnitude of these parameters is indicated as +++, ++, +, +/−. and −. (From Jain RK, Forbes NS. Can
engineered bacteria help control cancer? Proc Natl Acad Sci U S A. 2001;98:14748–14750.)

Vmax (mm/s)
epithelial cells. The remaining include sarcomas arising from mesen-
chymal cells (e.g., bone or muscle cells), lymphomas arising from
Arterioles 35 Venules lymphoid tissue, leukemias arising from hematopoietic cells, and
hemangiomas arising from ECs. In a poorly differentiated carcinoma,
30 the cancer cells might be packed loosely in clumps, whereas in a
well-differentiated carcinoma, the cells might be connected with
25
intercellular junctions and tightly packed in a nest enveloped by a
20 basement membrane. With tumor progression, cancer cells can invade
the basement membrane and spread to other regions.16 These various
15 types of normal host cells must migrate into the tumor from normal
tissue. Inflammatory cells might enter the tumor via blood vessels or
10 might infiltrate from the adjacent tissue or lymphatics.125 Adaptive
immune cells recruit innate immune response cells based on signals
5 from the stroma.151 Macrophages, which are of the innate response,
are plastic in that they can either fight tumor growth or support it
when exposed to certain cytokines.152 Other host cells, such as
50 25 0 25 50 fibroblasts, might proliferate and migrate from the adjacent connective
A Pial vessel diameter (µm) tissue.20,49,153 The interstitial subcompartment of a tumor is bounded
by the walls of the blood vessels on one side and by the membranes
of cancer and stromal cells on the other. In normal tissues, the blood
0.8 vessels are surrounded by a basement membrane, which, as discussed
MCalV U87
0.7
earlier, is defective in tumors.20 In addition, functional lymphatics
might be confined to the tumor margin.154,155 The interstitial space
0.6 of tumors, like that of normal tissues, is composed of a collagen and
RBC velocity (mm/s)

elastin fiber network that provides structural support to the tissue.


0.5 Interdispersed in this cross-linked structure are the interstitial fluid
and macromolecular constituents (polysaccharides, hyaluronan, and
0.4 proteoglycans), which form a hydrophilic gel. Compared with our
understanding of blood vessel formation, our understanding of stroma
0.3 generation is minimal. Dvorak proposed that the extravasated plasma
0.2
protein fibrinogen, a key component of the tumor interstitial fluid,
clots to form fibrin, which serves as a major component of the pro-
0.1 visional stroma. This provisional stroma eventually is replaced by
more mature connective tissue stroma. The tumor interstitial fluid
0.0 also contains several other Arg-Gly-Asp (RGD)–containing proteins,
0 10 20 30 40 50 60 70 0 10 20 30 40 50 60 70 including fibronectin, vitronectin, osteopontin, thrombospondin,
B Tumor vessel diameter (µm)
decorin, and tenacin.16 These proteins are present in both free and
bound forms. Their Arg-Gly-Asp sequence provides a binding site for
Figure 8.7  •  Blood velocity as a function of vessel diameter. (A) Normal adhesion that assists in the migration of various cells, including stromal
pial vessels. (B) MCaIV (mammary carcinoma) and U87 (glioma) tumors cells. In addition to extravasating from the leaky tumor vessels, these
xenografted on the pial surface. The measured tumor blood velocities are an proteins, along with collagen and various proteoglycans, are also
order of magnitude lower than the velocities in the normal host tissue and synthesized by the stromal cells, albeit in a form that differs from that
are not related to the vessel density. (From Yuan F, Salehi HA, Boucher Y, in the plasma or normal tissues.16 Tumor interstitial fluid also can
et al. Vascular permeability and microcirculation of gliomas and mammary contain various growth factors that facilitate stroma formation. For
carcinomas transplanted in rat and mouse cranial windows. Cancer Res. example, in vitro studies have suggested that platelet-derived growth
1994;54:4564–4568.) factor–BB (PDGF-BB) is involved in the recruitment of fibroblasts
116 Part I: Science and Clinical Oncology

2,500
Dorsal window

2,000

Tracer size (nm)


1,500

1,000

500

ST-12

ST-8
Shionogi

HCaI

LS174T

MCaIV
A B

6.0
Cranial window 2,500
MCaIV HCaI
5.0
2,000
Permeability (nm10−7 cm/sec)

Tracer size (nm)

4.0
1,500

3.0
1,000

2.0
500

1.0
0
Cranial

Cranial
Sc

Sc
0.0
C MCaIV U87 HGL21 LS174T D
Figure 8.8  •  Heterogeneous permeability of tumor vessels. Vessel permeability varies spatially within a given tumor, between tumors of identical type
implanted in different host organ environments, and between different tumor types in the same organ environment. (A) Vessels within a given tumor are
leaky in some areas and relatively impermeable in others. (B–C) Tumors of different types grown in the same environment show variations in both vessel pore
size and permeability. Pore sizes are measured by the largest tracer particle able to permeate the vessel wall. (D) MCaIV and HCaI tumors implanted in two
different sites (subcutaneous and cranial) have vessels with different pore sizes. For both tumor types, larger pore sizes are observed in the subcutaneous (Sc)
tumors compared with the cranial tumors. ([A and C] From Yuan F, Leunig M, Huang SK, et al. Microvascular permeability and interstitial penetration of
sterically stabilized [stealth] liposomes in a human tumor xenograft. Cancer Res. 1994;54:3352–3356. [B and D] From Hobbs SK, Monsky WL, Yuan F, et al.
Regulation of transport pathways in tumor vessels: role of tumor type and microenvironment. Proc Natl Acad Sci U S A. 1998;95:4607–4612.)

to tumors, and TGF-β induces the production of collagen and other K (cm2/mm Hg/s) relates the interstitial velocity to the pressure
matrix molecules in tumors.20,156 With the increasing interest in use gradient.134 Values of these transport coefficients are governed by the
of the fragments of matrix constituents for controlling angiogenesis, structure and composition of the interstitial compartment and by the
our understanding of the molecular and cellular mechanisms of stroma physicochemical properties of the solute molecule.158–168 The value of
generation in tumors will increase.44 K for a human colon carcinoma xenograft (LS174T), measured by
means of two different methods, was found to be higher than that
Interstitial Transport of a hepatoma, which, in turn, was higher than that of the normal
liver.168–170 With use of fluorescence recovery after photobleaching,
Once a molecule has extravasated, its movement through the interstitial D of various molecules in tumors was found to be about one-third
space occurs by diffusion and convection.102,157 Diffusion is proportional that in water and higher than the values in the host tissue (Fig.
to the concentration gradient in the interstitium, and convection 8.9A).160,171 Collagen content and structure have a significant effect
is proportional to the interstitial fluid velocity, which, in turn, is on D in tumors.162,166,169,172–176 This is surprising because hyaluronan
proportional to the fluid pressure gradient in the interstitium. Just as and proteoglycans, not collagen, account for most of the resistance
the interstitial diffusion coefficient D (cm2/s) relates the diffusive flux to transport in normal tissues. Because collagen is produced by host
to the concentration gradient, the interstitial hydraulic conductivity cells (e.g., fibroblasts), the penetrability into a tumor depends on the
−5
10

−6
10
2 −1
Diffusion coefficient, D, cm s

−7
10

−8
10

PBS
−9
10 U87 DC
U87 CW
Mu89 CW
Mu89 DC
−10
10

0.1 1 10 100

Radius, RH, nm
A

Control injection Collagenase injection

Initial virus
distribution
Specific antibody

Initial cell
infection

1 sec 10 sec 100 sec


First viral
replication

Secondary
cell infection
B C Nonspecific antibody

Figure 8.9  •  Interstitial transport in tumors. Transport of molecules through a tumor is affected by several factors. (A) Diffusivity of a molecule in a tumor
decreases with increasing molecular weight and is dependent on the host-tumor interaction. The diffusivity of macromolecules is lower in subcutaneous tumors
in dorsal chamber (DC; U87 glioblastoma [purple circles] and Mu89 melanoma [blue circles]) than in the same tumors grown in cranial windows (CW; U87
[yellow squares] and Mu89 [green diamonds]), and both are less than in phosphate-buffered saline (PBS [red triangles]). (B) A representative model of improvement
in oncolytic viral distribution and tumor cell infection by collagenase treatment. After direct intratumor injection, viral spread (red area) is limited by fibrillar
collagen (red lines) and results in a cluster of infected cells (light green). The collagen network also restricts the distribution of subsequent viral progeny, and
tumor cell infection beyond the initial injection site is not achieved. In contrast, injection of virus together with collagenase results in a more diffuse distribution
of viral particles and a greater number of initially infected cells (light green). Viral particles released by these cells have greater access to neighboring uninfected
cells. This process results in more widespread secondary infection (dark green) and ultimately greater therapeutic efficacy. (C) Interstitial transport is also
reduced by binding. The fluorescence of a photobleached spot recovers much more slowly with a specific antibody than with a nonspecific antibody. The
binding of the specific antibody hinders the transport of the molecule into the photobleached spot, slowing the fluorescence recovery. ([A] From Pluen A,
Boucher Y, Ramanujan S, et al. Role of tumor-host interactions in interstitial diffusion of macromolecules: cranial vs. subcutaneous tumors. Proc Natl Acad
Sci U S A. 2001;98:4628–4633. [B] From McKee TD, Grandi P, Mok W, et al. Degradation of fibrillar collagen in a human melanoma xenograft improves
the efficacy of an oncolytic herpes simplex virus vector. Cancer Res. 2006;66:2509–2513. [C] Modified from Berk DA, Yuan F, Leunig M, Jain RK. Direct in
vivo measurement of targeted binding in a human tumor xenograft. Proc Natl Acad Sci U S A. 1997;94:1785–1790.)
118 Part I: Science and Clinical Oncology

host-tumor interaction (see Fig. 8.9A). Thus, agents that interfere with and human tumors (to be discussed shortly). Embryonic lymphatic
collagen synthesis and/or organization (e.g., relaxin, losartan, bacterial vessels originate primarily from blood vessels according to the following
collagenase) might increase interstitial transport in tumors159,172,173 (see process (see Fig. 8.10B).188–190
Fig. 8.9B). The time constant for a molecule with diffusion coefficient
D to diffuse across a distance L is approximately L2/4D. For diffusion 1. In the early embryo, ECs of the cardinal vein express lymphatic
of immunoglobulin G (IgG) in tumors, this time constant is on the vascular endothelial receptor 1 (LYVE1) and VEGFR3, molecules
order of 1 hour for a 100-µm distance, days for a 1-mm distance, observed primarily (but not exclusively) on lymphatic vessels in
and months for a 1-cm distance. So for a 1-mm distance in tumor, normal adult tissues.
diffusional transport would take days, and for a 1-cm distance in tumor, 2. An as yet unknown signal triggers the expression of the homeobox
it would take months. Furthermore, the path through the interstitium gene Prox1 so that the protein is displayed in a polarized fashion
is tortuous over large length scales, greatly increasing the distance a in the ECs of the cardinal vein. This marks the first stage of
molecule must travel.161 If the central vessels have collapsed completely commitment to the lymphatic lineage.
owing to cellular proliferation and interstitial matrix rearrangement, 3. These LYVE1+VEGFR3+Prox1+ cells then start to bud, again in a
the reduced delivery of macromolecules by blood flow would make polarized fashion.
diffusion the primary mechanism of delivery to this necrotic center.6,80 4. At this stage, these early lymphatic ECs start expressing secondary
Binding of a low- or high-molecular-weight drug to plasma proteins lymphoid chemokine and increased levels of VEGFR3, markers
and various tissue components could further retard their transport in of mature lymphatic ECs.
tumors.171,177–183 The role of binding is clearly illustrated in Fig. 8.9C, 5. They then begin to form the lymphatic system.
which compares the rate of fluorescence recovery of a photobleached
spot in tumor tissue injected with a nonspecific versus a specific IgG. Members of the angiopoietin family (Ang-2) and its receptor (Tie-2)
In addition to the heterogeneity of D in tumors, the most unexpected and podoplanin are presumably involved in the maturation and
result of these photobleaching studies was the large extent (30%–40%) patterning of these nascent lymphatic vessels.191 The hematopoietic
of nonspecific binding.171 These results collectively suggest that the signaling pathway, Syk/SLP-76, contributes to the separation of
interstitial compartment of a tumor can be a formidable barrier to lymphatics from blood vessels. The molecules involved in angiogenesis
the uniform delivery of therapeutic macromolecules (e.g., antibodies, are also involved in lymphangiogenesis. For example, VEGF-C and
genes) in tumors, and strategies are needed to modify this barrier. VEGF-D can induce both angiogenesis and lymphangiogenesis and
are associated with lymphogenic metastasis in a variety of tumors.154
Lymphangiogenesis and Lymphatic Transport Their receptor VEGFR3 is present in both lymphatic and selected
vascular endothelium. Nonetheless, sprouting angiogenesis does not
In most normal tissues, extravasated plasma and macromolecules are occur during the initial growth of lymph node metastases.192 As is the
taken up by the lymphatics and returned to the blood circulation. It case with vascular angiogenesis, other positive and negative regulators
is widely accepted that lymphatic vessels are present in the tumor (e.g., angiopoietins) and other receptors (e.g., chemokine receptors
margin and the peritumoral tissue (Fig. 8.10A). Indeed, invasion of and neuropilins) could be involved in lymphangiogenesis, and as
peritumoral lymphatics is considered to be a poor prognostic factor discussed already, mechanisms analogous to co-option, intussusception,
for several tumors (e.g., breast, colorectal, and endometrial cancers), sprouting, and vasculogenesis might operate in lymphatic growth.12,191
and lymphatic metastasis is a major cause of morbidity and mortality Similar to the organ-specific angiogenic molecule (EG-VEGF) and
for others (e.g., melanoma, head and neck cancer, lung cancer, and EPCs, there could be organ-specific lymphangiogenic molecules and
cervical cancer). The hotly debated issue for nearly a century has been lymphatic EPCs that contribute to tumor-associated lymphangiogen-
whether anatomically defined lymphatic vessels are present within esis.23,39,189 Moreover, the proteolytic processing of lymphangiogenic
solid tumors and, if so, whether they function (see Fig. 8.10A).184,185 molecules and the phenotype and function of the resulting lymphatics
Currently available immunohistochemical markers stain for structures might depend not only on the tumor type but also on the host organ
in some tumors that resemble lymphatic vessels. Because many of in which the tumor is growing.77,187–189
these markers lack specificity, however, it is not clear whether they The mechanical and/or molecular signals that could trigger the
stain functional lymphatic vessels, ECs from remnant lymphatic vessels, lymphangiogenic switch are unknown. Because lymphatic vessels help
or some other structures (e.g., preferential fluid channels).155,169,186,187 maintain the balance of fluid in tissues, hydrostatic pressure is a probable
Mechanical solid stress induced by proliferating tumor cells and matrix trigger. Whether the hyperplasia and the increased density of lymphatic
production compresses and impairs lymphatic vessels that are co-opted vessels seen in the tumor margins are a response to elevated hydrostatic
or formed within a tumor.6,79,81 The impaired lymphatic vessels, in pressure in tumors and whether the newly formed lymphatics are able to
turn, contribute to the interstitial hypertension characteristic of animal remain open and carry cancer cells are open questions. Techniques such

Figure 8.10  •  (A) Schematic of lymphatics in tumors. It is widely accepted that peritumoral lymphatics exist and that metastasis can occur via these
lymphatic vessels. Evidence shows that structures within tumors that stain for lymphatic markers are not functional. Lower left inset, Molecular players in
lymphangiogenesis. (B) Mechanisms of lymphatic vessel formation and separation from blood vessels. (C) The tumor is modeled as a sphere of radius R,
embedded in a body fluid (e.g, peritoneal cavity, pleural cavity) or host tissue. The interstitial fluid oozing from the tumor periphery carries therapeutics (e.g,
monoclonal antibodies), growth factors (e.g, vascular endothelial growth factor [VEGF]–A, VEGF-B, VEGF-C, and VEGF-D), and cells (e.g., metastatic
cells) into the surrounding fluid or tissue. The result may be higher fluxes of cancer cells and growth factors from the tumor into the surrounding tissue or
body fluid. The growth factors (including VEGF-A, VEGF-B, VEGF-C, and VEGF-D) can induce angiogenesis and lymphangiogenesis, and the cancer cells
can contribute to metastatic dissemination via the lymphatic or blood vessels. Fluid seeping from the tumor surface can also cause edema (e.g., around brain
tumors) and ascites formation (e.g., ovarian cancer). Antiangiogenic therapy can “normalize” the tumor vasculature, leading to lower interstitial fluid pressure
(IFP) at the center, less steep IFP gradients, and lower fluid flow rates at the tumor margin, thus potentially reducing peritumoral edema, ascites formation,
and lymphatic metastasis. Furthermore, the normalized vessels may be more resistant to cancer cell intravasation, a prerequisite for hematogenous metastasis.
([A] From Jain RK, Fenton BT. Intratumoral lymphatic vessels: a case of mistaken identity or malfunction? J Natl Cancer Inst. 2002;94:417–421. [B] From
Jain RK, Padera TP. Lymphatics make the break. Science. 2003;299:209–210. Illustration: Katherine Sutliff. Reprinted with permission from AAAS. [C] From
Jain RK, Tong RT, Munn LL. Effect of vascular normalization by anti-angiogenic therapy on interstitial hypertension, peritumor edema and lymphatic
metastasis: insights from a mathematical model. Cancer Res. 2007;67:2729–2735).
Tumor Microenvironment: Vascular and Extravascular Compartment  •  CHAPTER 8 119

Mock transduced tumor VEGF-C overexpression

Tumor cell VEGFR-3


Tumor cell

VEGF-C

Normal
lymphatic Increased
VEGFR-3 vessel lymph flow

Dilated
e lymphatic vessel
ad
b lock
R-3
GF
VEGF-C is blocked VE
VEGF-C overexpression
by anti-VEGFR-3 Ab
Increased tumor cell arrival Afferent
in the lymph node lymphatic vessel
Tumor cell VEGF-C
VEGFR-3 Tumor cell Subcapsular sinus

Lymph node
cortex
Apoptotic and
Lymphatic hyperplasia proliferation rate
Lymphocyte
is blocked Anti-VEGFR-3 the same as control
A

Embryonic vein Metastatic cancer cell

Step 1 LYVE-1
VEGFR-3 Lymphatic
Hyperplastic vessel
lymphatic
vessel Tumor-generated growth factors
(VEGF-A, VEGF-C…)

tic Step 2 Blo


ha od
p
Lym

Prox-1

VEGFR-3 Uniformly Gradient


Step 3 high IFP of IFP
except at
VEGFR-3 margin
Prox-1
SLC Early
lymphocytic
endothelial cells
LYVE-1

Step 4 VEGFR-1
VEGFR-2
Prox-1
Tie-2
Tie-2 High IFV Lower IFV
Nrp-2
Nrp-2 at margin at margin
CD31
SLC
CD34
etc...
etc...

IFP
Step 5 Lymphocytes
with Syk and
SLP-76

? C Untreated tumor Normalized tumor

LEPCs ?
?Syk
?SLP-76
B
120 Part I: Science and Clinical Oncology

Table 8.2  Interstitial Fluid Pressure (in Millimeters 12


of Mercury) in Normal and Neoplastic

Interstitial pressure (mm Hg)


Tissues in Patients 10

Tissue Type N Mean Range References 8


Normal skin 5 0.4 −1.0 to 3.0 208 6
Normal breast 8 0.0 −0.5 to 3.0 208
Head and neck 19 13.2 4.0–33.0 207 4
carcinomas
2
Cervical carcinomas 12 15.7 10.0–26.0 205
Cervical carcinomas 102 19.0b −3.0 to 48.0 214 0
Lung carcinomas 26 10.0 1.0–27.0 124 0.0 0.4 1.2 2.0
Metastatic melanomas 10 21.7 0.0–60.0 209 A Depth (mm)
Metastatic melanomas 12 14.3 2.0–41.0 204
Breast carcinomas 13 29.0 5.0–53.0 211
Breast carcinomas 8 15.0 4.0–33.0 208
HER2− breast 70 14.0 0.0–43.0 273
carcinomasa
Brain tumorsa 17 7.0 2.0–15.0 171
Brain tumorsa 11 2.0 −0.5 to 8.0 213
Colorectal liver 8 21.0 6.0–45.0 208
metastasis
Lymphomas 6 4.6 1.0–12.5 209
Renal cell carcinoma 1 38.0 — 208
Pancreatic ductal 4 11.8 6.1–16.6 322
adenocarcinoma P

a
t
Patients were treated with antiedema therapy.
b
Interstitial fluid pressure given is a median value.

B
as microlymphangiography, reagents that block signaling of VEGF-C Figure 8.11  •  Interstitial fluid pressure (IFP) profile in a tumor and its
and VEGF-D, and lymphangiogenic factors yet to be discovered will potential application. (A) IFP is elevated and nearly uniform in the bulk of
allow us to answer these important questions.4,187,189,193–202 the tumor and drops precipitously in the tumor margin. (B) This elevated
pressure can be exploited to determine the location of a tumor precisely. ([A]
Interstitial Hypertension Modified from Boucher Y, Baxter LT, Jain RK. Interstitial pressure gradients
in tissue-isolated and subcutaneous tumors: implications for therapy. Cancer
Unlike normal tissues, in which the IFP is around 0 mm Hg, both Res. 1990;50:4478–4484. [B] From Jain RK, Boucher Y, Stacey-Clear A, et al.
animal and human tumors exhibit interstitial hypertension (Table Method for locating tumors prior to needle biopsy. US patent 19955396897.
8.2).154,157,170,203–214 The tumor IFP begins to increase as soon as the 1995.)
host vessels become leaky in response to permeability factors such as
VEGF, and thus IFP can be lowered by antibodies against VEGF or
VEGFR2.215–217 The IFP increases with tumor size in some tumors
and remains independent of tumor size in others.203–205,207,208 Three arterial resistance, so the MVP becomes closer to arterial pressure;
mechanisms contribute to interstitial hypertension in tumors. In normal and/or the tumor vessels have increased venous resistance because of
tissues, the lymphatics maintain the fluid homeostasis; thus the lack compression and tortuousity, so the whole intratumor vascular network
of functional lymphatics in tumors is a key contributor. Indeed, DiResta is under hypertension. Indirect evidence for the latter comes from
and colleagues were able to lower the IFP by placing “artificial lymphat- the decrease in IFP after decompression of tumor vessels by taxol-
ics” in tumors.218 The second contributor is the high permeability of induced apoptosis of perivascular cells.80
tumor vessels. As a result, the hydrostatic and oncotic (colloid osmotic) The elevated pressure can compromise the tumor microcirculation
pressures become almost equal between the intravascular and extra- and delivery of therapeutics in three ways:
vascular spaces.206,219 At least two pieces of evidence support this
hypothesis. First, reducing permeability by blocking VEGF signaling 1. Reduced transmural pressure gradients due to equilibrium between
lowers IFP.216,217,220–223 Second, IFP increases and decreases with the MVP and IFP reduce convection across tumor vessel walls and
microvascular pressure (MVP) within seconds.224–226 The two mecha- thus compromise the transport of macromolecules.206,225
nisms described thus far can explain interstitial hypertension only up 2. Because IFP is nearly uniform throughout a tumor and drops
to 20 to 30 mm Hg, the MVP of most exchange vessels (the hydrostatic precipitously in the tumor margin, the interstitial fluid oozes out
pressure within the lumina of capillaries) in human bodies, but IFPs of the tumor into the surrounding normal tissue, carrying mac-
as high as 94 mm Hg have been measured in human tumors.205 Because romolecules with it (Fig. 8.11).203,227
MVP is the driving force for IFP in tumors, these tumors must have 3. Finally, transmural coupling between IFP and MVP due to high
a high MVP. Indeed, this is the case.206 There are two possible explana- permeability of tumor vessels can lead to blood flow stasis in tumors
tions for elevated MVP in tumors: the tumor vessels have reduced without physically occluding the vessels.92–94
Tumor Microenvironment: Vascular and Extravascular Compartment  •  CHAPTER 8 121

The enhanced permeability can also facilitate lymphatic metastasis presumably as a result of lack of oxygen.230 This is referred to as
(see Fig. 8.10C). Thus, decreasing vascular permeability might restore “chronic hypoxia” or “diffusion-limited” hypoxia. Although various
the transmural pressure gradients and potentially resume or reestablish hypoxia markers and microelectrodes have suggested these gradients,
blood flow in the nonperfused regions of tumors. Some direct and the first direct measurements of these perivascular gradients, along
indirect antiangiogenic therapies might “normalize” the tumor vas- with Po2 and blood flow rate of the same vessels, became possible only
culature through this mechanism (see Fig. 8.4).101 This normalization with the development of phosphorescence quenching microscopy (Fig.
can also reduce the number of cells shed into peritumor lymphatics 8.12A).231,232 As discussed previously, blood flow in tumor vessels is
and alleviate peritumor edema (see Fig. 8.10C).74,228 intermittent, and thus some regions of a tumor are starved for oxygen
periodically. The resulting hypoxia is referred to as “acute hypoxia” or
“perfusion-limited hypoxia.”233,234 A necessary consequence of intermit-
METABOLIC MICROENVIRONMENT tent blood flow is the resumption of blood flow after shutdown, and
Hypoxia the resulting production of free radicals can lead to “reperfusion injury”
or “reoxygenation injury,” applying additional selection pressure on
A key function of the vasculature is to provide adequate levels of cancer cells.
nutrients and oxygen to the parenchymal cells and to remove waste
products. Based on the anatomy of the capillary bed and a mathemati- Low pH
cal model of oxygen diffusion and consumption, the Nobel laureate Another consequence of the abnormal microcirculation of the tumor
August Krogh introduced the concept of a diffusion limit for oxygen is low extracellular pH. There are at least two sources of H+ ions in
of 100 to 200 µm nearly a century ago.229 This unit of tissue—a tumors—lactic acid and carbonic acid.235 The former results from
single capillary surrounded by a 100- to 200-µm-radius cylinder—is glycolysis and the latter from conversion of CO2 and H2O via carbonic
referred to as a “Krogh cylinder” in physiology. Nearly 50 years later, anhydrase. The intracellular pH of cancer cells remains neutral or
Thomlinson and Gray identified similar “cords” in human lung cancer alkaline (pH 7.4), however, despite the acidic extracellular pH. Because
and found necrotic cells beyond 180 mm away from blood vessels, carbonic anhydrase-9 (CAIX), various glucose transporters (GLUT1,

7.4 14
pH
7.3 pO2 (mm Hg)
12

7.2
10
pO2 (mm Hg)

7.1
pH

8
7.0
6
6.9
4
6.8

6.7 2

6.6 0
0 50 100 150 200 250 300 350 400
A Distance (µm)

30

7.2
pO2 (mm Hg)

20
pH

7.0

pH
pO2 (mm Hg)
6.8 10
0 200 400
B
Figure 8.12  •  (A) pH and Po2 as a function of distance from a blood vessel in a tumor. The tumor environment becomes progressively more hypoxic
and acidic farther away from a blood vessel. (B) Lack of correlation between pH and Po2 in tumors. (From Helmlinger G, Yuan F, Dellian M, et al. Interstitial
pH and Po2 gradients in solid tumors in vivo: high-resolution measurements reveal a lack of correlation. Nat Med. 1997;3:177.)
122 Part I: Science and Clinical Oncology

GLUT3), and enzymes in the glycolytic pathway are upregulated cells that are more malignant, more invasive and genetically unstable,
by hypoxia, one would expect low extracellular pH and hypoxia to and less susceptible to apoptosis, thus rendering them resistant to
track each other and to colocalize with regions of low blood flow.236 various therapies. Therefore several molecules in the hypoxia-induced
Surprisingly, there is a lack of spatial correlation among these parameters pathways are now being targeted in the development of diagnostic
(see Fig. 8.12B), a discovery made possible by developments in optical and therapeutic agents. Nonetheless, the first and second strategies,
techniques that permit the simultaneous high-resolution mapping of although exploiting hypoxia, are dependent on the inadequate vas-
multiple physiologic parameters.231 A potential explanation for this culature for delivery of the therapeutic agents and are thus most
lack of concordance is that some perfused tumor vessels carry hypoxic amenable to tumors featuring intermittent rather than diffusion-limited
blood.231 Thus, although they might not be able to deliver adequate hypoxia. The third strategy involving normalization of tumor vessel
oxygen to the surrounding cells, they may be able to carry away the function by antiangiogenic and anti–solid stress therapy may reduce
waste products (e.g., lactic acid). intermittent and diffusion-limited hypoxia within the tumor micro-
environment and synergize with other strategies.256
Molecular, Cellular, and Therapeutic Consequences Hypoxia-induced pathways include genes involved in the following
processes:
The abnormal microenvironment promotes the malignant tumor
progression. Indeed, hypoxia is a validated independent negative • Oxygen delivery (e.g., heme oxygenase 1, erythropoietin)
prognostic biomarker for many cancers, and endogenous markers of • Glycolysis and glucose uptake (e.g., GLUT1 and GLUT3,
acidosis are potential negative biomarkers.237–240 Hypoxia and acidosis hexokinase-1 and hexokinase-2)
select for aggressive cancer cells and promote a vicious cycle of • pH control (e.g., carbonic anhydrase-9 and carbonic anhydrase-12)
angiogenesis, desmoplasia, and inflammation and immunosuppression • Stress-response pathways (e.g., growth arrest- and DNA damage-
that leads to exacerbated abnormal metabolism.241–244 Increased hypoxia induced gene GADD153)
predicts the likelihood of metastasis.245 • Growth factor signaling (e.g., IL-6, IL-8, insulin-like growth factor-2
Hypoxia and acidosis indicate poor delivery of blood, nutrients, [IGF-2])
and blood-borne therapies to tumors, and they also directly promote • PDGF-β
resistance to radiotherapy, chemotherapy, and immunotherapy. The • Angiogenesis (e.g., VEGF-A, VEGFR1, Ang-2, Tie-2, FGF-3,
presence of oxygen during irradiation makes the damage to DNA TGF-β, nitric oxide synthase [NOS], cyclooxygenase-2 [COX-2],
produced by radiation-induced free radicals permanent, whereas such hepatocyte growth factor [HGF])
damage can be repaired under hypoxic conditions.246 Therefore, hypoxia • Transcription (e.g., HIF-1α and HIF-2α, JUN, FOS, nuclear
in solid tumors significantly reduces their radiation sensitivity. In factor–κB [NF-κB])
addition, hypoxia and low extracellular pH adversely affect the cel- • Apoptosis (e.g., BCL-interacting killer [BIK], annexin V, 19-kd
lular uptake and cytotoxicity of certain therapeutics, although there interacting protein-3 [NIP3], NIP3-like protein X [NIX])
are several drugs whose efficacy increases in such conditions.246–249 • Growth inhibition signaling factors (e.g., p21, p27, GADD153)
Finally, hypoxia induces immune suppression and reduces the efficacy • Invasion and metastasis (e.g., metalloproteinases [MMPs], MMP-13,
of immune therapies.250–253,253a As a result, for nearly half a century plasminogen activator inhibitor-1 [PAI-1])236
considerable preclinical and clinical efforts have been focused on
alleviating hypoxia by improving tumor perfusion with various therapies, Of the various molecules involved in sensing and responding to
including: hypoxia, HIF-1α has received the most attention. This transcription
factor is upregulated in several human tumors.236 Regulated by a
• Mild hyperthermia or drugs
proline hydroxylase, HIF-1α can activate the genes for desmoplasia,
• Increased oxygen content of the blood (e.g., via hyperbaric
angiogenesis, vasodilation, glycolysis, and erythrocyte production by
oxygenation)
binding to the hypoxia-response element.243,257,258 Surprisingly, teratomas
• Increased hemoglobin and hematocrit (e.g., via erythropoietin)
arising from HIF-1α−/− embryonic stem cells grow more rapidly despite
• Radiation sensitizers
lower levels of VEGF and angiogenesis.52,259 This counterintuitive
Unfortunately, these strategies’ success in clinical studies has been finding could be a result of the ability of HIF-1α−/− cells to survive
limited for multiple reasons. One common theme of these strategies under hypoxic conditions instead of undergoing apoptosis.49 Interesting
is that they rely on the abnormal tumor vasculature. As a result, these to note, other HIF-1α−/− cancer cells lead to slowly growing tumors.
strategies are incapable of increasing Po2 in all regions of tumors to As a result, molecular therapies that target HIF-1α or hypoxia-response
optimal levels and/or delivering radiation sensitizers or chemotherapeutic element are under intensive investigation for cancer detection and
drugs to all regions of a tumor at therapeutically effective levels. treatment.260
Currently, three broad strategies are emerging:
CLINICAL RELEVANCE OF APPROACHES TO
1. Use of hypoxia and/or low pH to selectively activate drugs in the ALLEVIATE HYPOXIA
tumor or to attract anaerobic bacteria
2. Dissection of hypoxia-induced pathways to identify novel targets Two major problems currently plague the nonsurgical treatment of
for drug development malignant solid tumors. First, physiologic barriers within tumors
3. Normalization of tumor vasculature and/or reduction of solid stress impede the delivery of therapeutics and oxygen (a key sensitizer to
to alleviate hypoxia254,255 ionizing radiation and other therapies) at effective concentrations to
all cancer cells.261,262 Second, inherent or acquired resistance result-
The first strategy led to the development of agents such as tira- ing from genetic and epigenetic mechanisms, which themselves are
pazamine and to the rejuvenation of interest in bacteriolytic therapy; promoted by hypoxia, reduces the effectiveness of both conventional
both approaches are still being tested in clinical trials.89,246 The second and novel therapies.263,264 Clinical imaging studies are consistent
strategy has revealed several molecular determinants of the physiologic with the hypothesis that patients whose disease responds to therapies
and pathophysiologic responses to hypoxia.236 The balance between with increased perfusion and oxygenation have better outcomes (Fig.
hypoxia-induced apoptosis and necrosis on one hand, and the increased 8.13A–B).96,265–268 Therefore cancer researchers must strive to optimize
resistance to cell death mediated by various hypoxia-induced pathways strategies that decrease hypoxia in tumor lesions. How can we exploit
on the other, determines whether a tumor can survive and even grow the pathophysiology of tumors to overcome hypoxia for better disease
under hypoxic conditions. Ultimately, hypoxia might select for tumor management?
Tumor Microenvironment: Vascular and Extravascular Compartment  •  CHAPTER 8 123

100 1
P = .028 D
80
.8 In ecre
cr
In ea ase
Overall Survival (%)

De cre sed d p

Overall Survival
60

cr ase hy erfu
.6
ea d p
se pe oxi sion
40

Dose
.4
d rfu a
P = 0.035 hy s
20
po ion
.2
∆ MTT ≤ 0.76 (n = 7)
xia
0 0.76 < ∆ MTT ≤ 1.05 (n = 6)
0 200 400 600 800 ∆ MTT > 1.05 (n = 6)
0
Time (days)
No effect on vessels
Perfusion increased (n = 20)
0 6 12 18 24 30 36
Follow-up time (months)
A Perfusion stable or decreased (n = 20)
B C Length of treatment

Mature vessels
Supply large volumes of tissue
Untreated Normalized
Well Normalization Normalization
Vessel dense
perfused including through
vessel vessel
pruning Combination
fortification only
of vessel
fortification
and stromal
High extent of Enha
Enhanced reprogramming
normalization
oxy
xy
yg
oxygenation

Sparse Dense
perfusion perfusion
Ex Leaves Grants
prucess regions all
nin ive without regions
Vessel sparse g Poorly
flow flow
perfused
Solid tumors Stromal
pretreatment reprogramming

Insufficient Dimi
Diminished
vascular oxygenation Proangiogenesis
network or
stromal destruction

Leaky vessels
Supply small volumes of tissue
blood perfusion recruited
pericytes
vessels oxygenation pericytes
Hypoxia Normoxia
D E
Figure 8.13  •  Vascular “normalization” in cancer patients. (A) Glioblastoma patients who responded to antiangiogenic therapy with increased perfusion
and oxygenation (blue) had longer overall survival compared with those who did not (orange). These data support the normalization hypothesis as well as the
need to test whether perfusion and oxygenation are predictive biomarkers of response in large, prospective trials. (B) Non–small cell lung cancer patients who
responded to antiangiogenic therapy with reduced (green) or stable (red) mean transit time—the absolute value of this quantity is inversely proportional to
perfusion—had longer overall survival than patients with increased mean transit time (gray). (C) In patients, a tumor vessel “normalization window” of
enhanced perfusion and oxygenation depends on the dose and the length of treatment. (D) Preclinical and clinical studies support the hypothesis that the
therapeutic effect of antiangiogenic therapies is best when vascular density is high and normalization fortifies rather than prunes vessels. The left quadrants
depict tumor vessels (red) before antiangiogenic therapy. The right quadrants depict possible outcomes of antiangiogenic therapy. Tumors with low pretreatment
vessel density do not respond to antiangiogenic therapy (bottom left to right quadrant), as the increase in vessel function from the recruitment of pericytes (teal)
cannot overcome the paucity of vessels. In contrast, tumors with high baseline vascularity that recruit pericytes have better outcomes (top left quadrant to top
right quadrant) than tumors with excessive pruning (top left quadrant to bottom right quadrant). (E) Combinations of normalizing therapies that increase vessel
maturity without pruning with solid stress alleviating agents that reprogram rather than destroy stroma might be optimal for alleviating hypoxia. Tumors
(black) usually feature both leaky vessels and sparsely perfused regions, thereby leaving the tissue hypoxic (white). Reengineering the tumor microenvironment
could promote normoxia (blue) and lead to improved treatment outcomes. Normalization strategies that induce pruning lead to sparse perfusion (orange),
whereas proangiogenic and stromal destruction strategies increase vessel leakiness (red). Fortification of vessels with pericytes (green) or reprogramming the
stroma to alleviate solid stress (magenta) avoid detrimental effects, and the combination (greenish blue) might be ideal to alleviate hypoxia in most cases. ([A]
From Batchelor TT, Gerstner ER, Emblem KE, et al. Improved tumor oxygenation and survival in glioblastoma patients who show increased blood perfusion
after cediranib and chemoradiation. Proc Natl Acad Sci U S A. 2013;110:19059–19064. [B] From Heist RS, Duda DG, Sahani DV, et al. Improved tumor
vascularization after anti-VEGF therapy with carboplatin and nab-paclitaxel associates with survival in lung cancer. Proc Natl Acad Sci U S A. 2015;112:1547–1552.
[C–E] From Martin JD, Fukumura D, Duda DG, et al. Reengineering the tumor microenvironment to alleviate hypoxia and overcome cancer heterogeneity.
Cold Spring Harb Perspect Med. 2016;6:a027094.)

Vascular Normalization Through types of cancers including some metastatic settings.10 Studies in a
Antiangiogenic Therapy range of these tumor types have largely confirmed the hypotheses that
anti-VEGF/VEGFR therapy has antivascular effects and can induce
The broad success of antiangiogenic therapy in phase III clinical trials vascular normalization.221,223,267–274 How these effects on the vasculature
across various tumor types provides substantial data to generate new relate to hypoxia and patient survival is dependent on the tumor type
hypotheses regarding strategies to eliminate hypoxia. There are 12 and a topic of continued research.
VEGF-blocking antiangiogenic agents (bevacizumab, regorafenib, Tumor types that are highly sensitive to antiangiogenic therapy
ramucirumab, sorafenib, sunitinib, pazopanib, axitinib, vandetanib, are either highly VEGF dependent (i.e., clear cell renal carcinoma,
lenvatinib, nintedanib, carbozantinib, and aflibercept) that are approved ovarian cancer, cervical cancer, and thyroid cancer) or highly vascularized
by either the Food and Drug Administration or the European Medicines (i.e., neuroendocrine cancers and hepatocellular carcinoma [HCC]).10
Agency.10,12,269 These agents have been approved for treatment of 10 These types of tumors have a “tumor vessel” phenotype, which consists
124 Part I: Science and Clinical Oncology

of vessels among cancer cells without significant intervening stroma.275 would be improved. Unlike preclinical models, the phase III bevaci-
Clinical studies in these tumors indicated that blockade of VEGF has zumab experience in metastatic colorectal cancer patients did not
antivascular and normalizing effects, which are identified as potential identify P53, KRAS, or BRAF status, VEGF or thrombospondin-2
biomarkers of response.274,276–278 Preclinical and clinical studies also (TSP2) expression, or microvascular density at baseline as predictive
demonstrated that there is a dose- and treatment duration–dependent markers of response.309,310 Similarly, circulating VEGF levels—although
window in which VEGF blockade increases oxygen delivery by normal- of potential prognostic biomarker value—have unclear predictive
izing before excessively pruning vessels (see Fig. 8.13C).268,272,279,280 biomarker value.38,277 In collaboration with our clinical colleagues, we
Nonetheless, it remains unclear whether these antivascular and normal- have been examining these questions in over 30 multidisciplinary
izing effects lead to increased oxygen delivery or not. Indeed, blocking trials by measuring tissue, circulating, and imaging biomarkers at
VEGF in highly dependent tumors might be sufficient to elicit a various time points during and after treatment, and correlating these
strong antitumor response even while concurrently increasing hypoxia, with treatment outcomes. Several promising biomarkers have emerged.
but increased hypoxia eventually leads to resistance, as several studies One is the level of circulating sVEGFR1/sFLT1 (an endogenous blocker
in liver cancer have demonstrated.281–284 Thus, efforts must be made of VEGF and PlGF) as a biomarker of intrinsic resistance to anti-VEGF
to maintain and improve the high response rates of antiangiogenic therapy. Locally advanced rectal carcinoma patients who had high
therapy in these sensitive tumors while avoiding increasing hypoxia plasma sVEGFR1 levels before treatment were less likely to benefit—or
in order to extend overall survival. experience toxicities—from bevacizumab therapy combined with
Other tumor types that range from partially to mostly resistant to chemoradiation.223,311 Later the same association was found for newly
antiangiogenic therapy, such as colorectal and breast cancer, respectively, diagnosed GBM, triple-negative breast cancer, non–small cell lung
often have a stromal vessel phenotype characterized by dense stroma cancer, HCC, and metastatic colorectal carcinoma patients treated
encapsulating avascular cancer cell nests.275 Pancreatic ductal adeno- with anti-VEGF agents.267,273,312–315 Of interest, a retrospective analysis
carcinoma, which is a highly desmoplastic tumor type, is resistant to in two randomized phase III trials showed that a genetic variation in
antiangiogenic therapy.10 In this phenotype, VEGF blockade destroys the VEGFR1 gene correlates with increased VEGFR1 expression and
the stroma including blood vessels without reducing tumor size even poor outcome of bevacizumab treatment in metastatic renal cell
transiently.275 In the long term, destruction of the stroma could increase carcinoma and pancreatic ductal adenocarcinoma patients.257 A second
disease progression.285–287 Stromal vessel phenotype tumors have a biomarker is the SDF1α/CXCR4 axis as a potential evasive pathway.
lower vessel density than tumor vessel phenotype tumors.275 Less than Circulating levels of the chemokine SDF1α increase in patients at
half of the vessels in desmoplastic preclinical breast and pancreatic the time of evasion from anti-VEGF therapies. This was the case in
tumor models are patent and perfused because of solid stress.288,289 rectal carcinoma patients treated with bevacizumab, in GBM patients
Thus, stromal vessel phenotype tumors are usually hypovascular and with cediranib, in HCC patients with sunitinib, and in sarcoma patients
hypoperfused. Because VEGF blockade can increase existing vessel with sorafenib.220,271,276,314,316 Interesting to note, the sources of SDF1α
function but it cannot create new vessels, it might only be effective are different in each of these diseases, and so is the role of the SDF1α/
in tumors that have a relatively high microvessel density and when CXCR4 pathway.317 For example, in GBM this pathway appears to
the treatment induces pericyte recruitment (vessel maturation) without facilitate invasion of cancer cells and co-option of host vessels by
pruning (see Fig. 8.13D).242,258,273,290–292 In addition to preclinical and invading cancer cells. These biomarker candidates need to be tested
clinical tissue biomarker studies, the findings of clinical imaging studies prospectively.
indicating that patients with hypoxic tumors are unresponsive to VEGF
blockade are consistent with this hypothesis.293–295 A small clinical Toxicity of Antiangiogenic Therapy
study has suggested that a cytotoxic agent that likely reduces solid
stress could be more efficient at increasing oxygenation than VEGF Experimental studies showed that in 11 of the 17 healthy organs
blockade in breast cancer.296 studied, VEGF blockade significantly decreased the number of normal
Mathematical modeling and preliminary clinical studies indicate capillaries.318 In cancer patients, most anti-VEGF agents often induce
that combining antiangiogenic therapies with solid stress–alleviating proteinuria, hypertension, thyroid-stimulating hormone elevation, and
agents in treating stromal vessel phenotype or antiangiogenic therapy– gastrointestinal toxicity, but agent-specific toxicities have also been
resistant tumors might increase oxygen delivery further (see Fig. reported.11,319 In addition, the long-term effects of antiangiogenic
8.13E).297,298 Metronomic chemotherapy alone could normalize vessels therapies in patients with less advanced lesions remain to be established.
by modulating angiogenesis pathways while alleviating solid stress by
killing cancer cells.299 Solid Stress Alleviation Through Stromal
To an even larger extent than in stromal vessel phenotype tumors, Reprogramming
antiangiogenic therapies are ineffective against early metastases.300
Lymph node metastases lack angiogenesis and thus do not respond Solid stress comprises the forces generated by proliferating and contract-
to antiangiogenic therapies.192 Similarly, preclinical evidence indicates ing cancer and stromal cells that are stored as strain energy and applied
that lung metastases co-opt the existing vasculature without relying by the extracellular matrix and surrounding tissue.289,320–322,322a
on sprouting angiogenesis.301 When cancer cells are co-opting existing Hyaluronan and collagen collaborate to transmit solid stress to compress-
vasculature, they are nearly mature and normally functioning vessels, ing vessels, suggesting that vessel compression is a problem in stromal
so normalizing therapy is unlikely to affect tumor progression. Fur- vessel phenotype tumors.288,323 Nonetheless, vessels are compressed in
thermore, these invading cells are insensitive to chemotherapy, so new small dysplastic lesions, suggesting that even vessels in tumor vessel
cytotoxic agents must be developed to target these cells.302–307 phenotype tumors might be compressed.324 Targeting solid stress by
depleting the stroma could aggravate tumor aggressiveness.285,287,325
Biomarkers of Response to Antiangiogenic Therapy Thus, stromal reprogramming is favored, as activated fibroblasts that
produce and maintain desmoplasia are deactivated, leading to vessel
As with many targeted therapies, the survival benefit of cancer patients decompression and increased oxygenation.288,326,327 Retrospective studies
after antiangiogenic therapies remains modest and restricted to as yet each surveying hundreds of patients with non–small cell lung cancer,
unidentified subsets of tumors. Unfortunately, unlike with many pancreatic ductal adenocarcinoma, and renal cell carcinoma have
anticancer targeted therapies, there are no validated biomarkers of supported the hypothesis that repurposing angiotensin system inhibitors
patient selection, response, or resistance for antiangiogenic agents.38,308 from hypertension to reprogramming the stroma of cancer can increase
If biomarkers could be found to identify patients more likely to benefit patient survival.328–332 Preliminary clinical data indicate that angiotensin
from these drugs, the survival benefit in patients on anti-VEGF drugs system inhibition in patients decompresses vessels, which is consistent
Tumor Microenvironment: Vascular and Extravascular Compartment  •  CHAPTER 8 125

with preclinical data.333 In addition, metastases are desmoplastic and • Improved survival has been observed with broad-spectrum multitar-
preclinical data indicate that angiotensin system inhibition could inhibit geted tyrosine kinase inhibitors (e.g., sorafenib, sunitinib, pazopanib,
metastasis through inhibition of monocyte recruitment.334,335 Clinical axitinib, vandetanib, and lenvatinib) when used as monotherapy.
trials are ongoing to test whether stromal reprogramming can increase Until recently, VEGF receptor–selective tyrosine kinase inhibitors
survival.335a Stromal depleting agents have had mixed success clinically; had repeatedly failed to confer an advantage when combined with
agents targeting lysyl oxidase homolog 2 and Hedgehog pathway chemotherapy. However, a study in lung cancer showed increased
signaling have failed, whereas one agent targeting hyaluronidase has overall survival with nintedanib with chemotherapy.
succeeded in a phase II trial.326,336–338 • Anti-VEGF therapy can prune and normalize tumor vascular
structure and function.
Perspective • There is an urgent need to identify biomarkers to guide anti-VEGF
therapy and combination therapies using anti-VEGF agents.
The major directions for the immediate future are developing therapies
that favor pericyte recruitment over pruning, understanding how to Antiangiogenic agents are now expected to make a difference in
combine antiangiogenic therapies with solid stress–alleviating agents, cancer patients with a wide variety of tumor types. With the advent
and identifying the first biomarkers for anti-VEGF therapy.38 Achieving of specific and potent new agents—approved or in the process of
these goals would allow optimization of treatment protocols and being approved—oncologists have a variety of direct and indirect
reduction of the adverse effects, and better integration of these drugs antiangiogenic agents to choose from when designing therapy protocols.
with emerging immunotherapies. Determining whether the regimens used in the successful trials are
First, identifying the vascular “normalization window” in patients optimal, however, and whether antiangiogenic agents will work in
would allow synergistic combinations with chemotherapy, radiotherapy, patients outside the rigorous inclusion criteria used for those trials,
and immunotherapy.253a Second, understanding the mechanisms of will be critical for deciding the standard of care for different malignan-
vessel pruning and cancer cell apoptosis induced by anti-VEGF therapy, cies. Establishing the most advantageous combinations will require a
and tumor escape from it, may allow further sensitization of tumor better understanding of the mechanisms of action of each anti-VEGF
cells to cytotoxic therapies. Third, characterization of the effect of agent and the sensitivity of each tumor type, as well as development
anti-VEGF therapy on bone marrow–derived cells’ contribution to of robust biomarkers and imaging techniques to guide patient selection
tumor growth and relapse would allow judicious and more effective and protocol design. A deeper understanding of the mechanisms of
approaches to therapy involving these cells. Finally, clarification of antitumor activity of specific and multitargeted antiangiogenic agents
other mechanisms involving the immune system or the stromal and in patients, how these agents can best be combined with other treatment
interstitial matrix compartments would contribute to the establishment approaches such as chemotherapy, radiation therapy, and immuno-
of more efficacious combinatorial strategies. therapy, and how optimization of these effects can be monitored
In this respect, new biomarkers and improved imaging techniques clinically should contribute to significantly improved cancer treatment
will play a major role in monitoring effects and stratifying patients, and extend survival of cancer patients in the near future, in addition
with the ultimate goal of individualized therapy. to enhancing the prospects for development of curative treatments
for different cancers in the more distant future.
CONCLUSION
ACKNOWLEDGMENTS
The successes of the anti-VEGF agents and development of solid
stress–alleviating agents have raised great hope and have taught us We would like to thank Kevin Kozak for his input on updating this
important lessons about the significance of the target, timing, and chapter. This chapter is an updated and expanded version of a chapter
dosage of each agent.38,76 by R. K. Jain entitled Molecular Pathophysiology of Tumors (in Perez
CA, Brady LW, Halperin EC, Schmidt-Ullrich R, eds: Principles and
• According to the results of the phase III trials completed to date,
Practice of Radiation Therapy. New York: Lippincott, Williams &
bevacizumab can increase median overall survival in certain cancers
Wilkins; 2003:163–179) and a review article by J. D. Martin, D.
when combined with standard chemotherapy, but not when used
Fukumura, D. G. Duda, Y. Boucher, and R. K. Jain entitled Reengineer-
as monotherapy.
ing the Tumor Microenvironment to Alleviate Hypoxia and Overcome
• Anti-VEGF therapy with bevacizumab can increase overall survival
Cancer Heterogeneity (in Cold Spring Harbor Perspectives in Medicine,
and/or progression-free survival in advanced colorectal carcinoma,
December 2016, Volume 6, Issue 12). The work summarized here
non–small cell lung carcinoma, breast carcinoma, renal cell carci-
has been supported continuously by the National Cancer Institute
noma, ovarian cancer, and cervical cancer when combined with
since 1980 with funding to R. K. Jain.
cytotoxic agents or immunotherapy. Aflibercept (“VEGF Trap,” an
sFLT1 chimera) and ramucirumab combinations with chemotherapy
resulted in improved survival in metastatic colorectal cancer and The complete reference list is available online at
lung cancer (for ramucirumab). ExpertConsult.com.

KEY REFERENCES
4. Jain RK, Munn LL, Fukumura D. Dissecting tumour 9. Hurwitz H, Fehrenbacher L, Novotny W, et al. angiogenesis and a potential target for diagnosis and
pathophysiology using intravital microscopy. Nat Bevacizumab plus irinotecan, fluorouracil, and therapy. J Clin Oncol. 2002;20:4368–4380.
Rev Cancer. 2002;2:266–276. leucovorin for metastatic colorectal cancer. N Engl 20. Jain RK. Molecular regulation of vessel maturation.
6. Helmlinger G, Netti PA, Lichtenbeld HC, et al. J Med. 2004;350:2335–2342. Nat Med. 2003;9:685–693.
Solid stress inhibits the growth of multicellular 12. Carmeliet P, Jain RK. Molecular mechanisms 33. Ferrara N, Gerber HP, LeCouter J. The biology of
tumor spheroids. Nat Biotechnol. 1997;15:778– and clinical applications of angiogenesis. Nature. VEGF and its receptors. Nat Med. 2003;9:669–676.
783. 2011;473:298–307. 34. Melder RJ, Koenig GC, Witwer BP, et al. During
7. Folkman J. Tumor angiogenesis: therapeutic implica- 15. Hanahan D, Weinberg RA. Hallmarks of cancer: angiogenesis, vascular endothelial growth factor
tions. N Engl J Med. 1971;285:1182–1186. the next generation. Cell. 2011;144:646–674. and basic fibroblast growth factor regulate natural
8. Gullino PM. Angiogenesis and oncogenesis. J Natl 16. Dvorak HF. Vascular permeability factor/vascular killer cell adhesion to tumor endothelium. Nat Med.
Cancer Inst. 1978;61:639–643. endothelial growth factor: a critical cytokine in tumor 1996;2:992–997.
126 Part I: Science and Clinical Oncology

36. Fidler IJ. Angiogenic heterogeneity: regulation of 119. Yuan F, Chen Y, Dellian M, et al. Time-dependent 221. Willett CG, Boucher Y, di Tomaso E, et al.
neoplastic angiogenesis by the organ microenviron- vascular regression and permeability changes in Direct evidence that the VEGF-specific antibody
ment. J Natl Cancer Inst. 2001;93:1040–1041. established human tumor xenografts induced by bevacizumab has antivascular effects in human rectal
38. Jain RK, Duda DG, Willett CG, et al. Biomarkers an anti-vascular endothelial growth factor/vascular cancer. Nat Med. 2004;10:145–147.
of response and resistance to antiangiogenic therapy. permeability factor antibody. Proc Natl Acad Sci 222. Willett CG, Boucher Y, Duda DG, et al. Surrogate
Nat Rev Clin Oncol. 2009;6:327–338. USA. 1996;93:14765–14770. markers for antiangiogenic therapy and dose-limiting
49. Brown EB, Campbell RB, Tsuzuki Y, et al. In 124. Duda DG, Duyverman AM, Kohno M, et al. toxicities for bevacizumab with radiation and
vivo measurement of gene expression, angiogen- Malignant cells facilitate lung metastasis by chemotherapy: continued experience of a phase I
esis and physiological function in tumors using bringing their own soil. Proc Natl Acad Sci USA. trial in rectal cancer patients. J Clin Oncol. 2005;23:
multiphoton laser scanning microscopy. Nat Med. 2010;107:21677–21682. 8136–8139.
2001;7:864–868. 150. Hanahan D, Coussens LM. Accessories to the 223. Willett CG, Duda DG, di Tomaso E, et al. Efficacy,
57. Hobbs SK, Monsky WL, Yuan F, et al. Regulation of crime: functions of cells recruited to the tumor safety, and biomarkers of neoadjuvant bevacizumab,
transport pathways in tumor vessels: role of tumor microenvironment. Cancer Cell. 2012;21:309–322. radiation therapy, and fluorouracil in rectal cancer:
type and microenvironment. Proc Natl Acad Sci USA. 153. Fukumura D, Xavier R, Sugiura T, et al. Tumor a multidisciplinary phase II study. J Clin Oncol.
1998;95:4607–4612. induction of VEGF promoter activity in stromal 2009;27:3020–3026.
61. Izumi Y, Xu L, di Tomaso E, et al. Tumour biology: cells. Cell. 1998;94:715–725. 225. Netti PA, Hamberg LM, Babich JW, et al. Enhance-
Herceptin acts as an anti-angiogenic cocktail. Nature. 155. Padera TP, Kadambi A, di Tomaso E, et al. Lymphatic ment of fluid filtration across tumor vessels: implica-
2002;416:279–280. metastasis in the absence of functional intratumor tion for delivery of macromolecules. Proc Natl Acad
63. Jain RK. Normalization of tumor vasculature: an lymphatics. Science. 2002;296:1883–1886. Sci USA. 1999;96:3137–3142.
emerging concept in antiangiogenic therapy. Science. 157. Jain RK. Transport of molecules in the tumor inter- 229. Krogh A. The Anatomy and Physiology of Capillaries.
2005;307:58–62. stitium: a review. Cancer Res. 1987;47:3039–3051. New York: Yale University Press; 1922.
64. Jain RK. Taming vessels to treat cancer. Sci Am. 159. Brown E, McKee T, diTomaso E, et al. Dynamic 230. Thomlinson RH, Gray LH. The histological structure
2008;298:56–63. imaging of collagen and its modulation in tumors of some human lung cancers and the possible
70. Jain RK. Determinants of tumor blood flow: a review. in vivo using second-harmonic generation. Nat Med. implications for radiotherapy. Br J Cancer. 1955;9:
Cancer Res. 1988;48:2641–2658. 2003;9:796–800. 539–549.
74. Goel S, Duda DG, Xu L, et al. Normalization of 165. Nugent LJ, Jain RK. Extravascular diffusion 231. Helmlinger G, Yuan F, Dellian M, et al. Interstitial
the vasculature for treatment of cancer and other in normal and neoplastic tissues. Cancer Res. pH and pO2 gradients in solid tumors in vivo:
diseases. Physiol Rev. 2011;91:1071–1121. 1984;44:238–244. high-resolution measurements reveal a lack of
79. Stylianopoulos T, Martin JD, Chauhan VP, et al. 166. Pluen A, Boucher Y, Ramanujan S, et al. Role of correlation. Nat Med. 1997;3:177–182.
Causes, consequences and remedies for growth- tumor-host interactions in interstitial diffusion of 236. Harris AL. Hypoxia: a key regulatory factor in
induced solid stress in murine and human tumors. macromolecules: cranial vs. subcutaneous tumors. tumour growth. Nat Rev Cancer. 2002;2:38–47.
Proc Natl Acad Sci USA. 2012;109(38):15101–15108. Proc Natl Acad Sci USA. 2001;98:4628–4633. 258. Mazzone M, Dettori D, Leite de Oliveira R, et al.
81. Padera TP, Stoll BR, Tooredman JB, et al. Pathology: 172. Diop-Frimpong B, Chauhan VP, Krane S, et al. Heterozygous deficiency of PHD2 restores tumor
cancer cells compress intratumour vessels. Nature. Losartan inhibits collagen I synthesis and improves oxygenation and inhibits metastasis via endothelial
2004;427:695. the distribution and efficacy of nanotherapeutics normalization. Cell. 2009;136:839–851.
96. Sorensen AG, Emblem KE, Polaskova P, et al. in tumors. Proc Natl Acad Sci USA. 2011;108: 288. Chauhan VP, Martin JD, Liu H, et al. Angiotensin
Increased survival of glioblastoma patients who 2909–2914. inhibition enhances drug delivery and potentiates
respond to antiangiogenic therapy with elevated 206. Boucher Y, Jain RK. Microvascular pressure is the chemotherapy by decompressing tumour blood
blood perfusion. Cancer Res. 2012;72:402–407. principal driving force for interstitial hypertension vessels. Nat Commun. 2013;4:2516.
101. Jain RK. Normalizing tumor vasculature with anti- in solid tumors: implications for vascular collapse. 323. Chauhan VP, Boucher Y, Ferrone CR, et al. Compres-
angiogenic therapy: a new paradigm for combination Cancer Res. 1992;52:5110–5114. sion of pancreatic tumor blood vessels by hyaluronan
therapy. Nat Med. 2001;7:987–989. 217. Tong RT, Boucher Y, Kozin SV, et al. Vascular is caused by solid stress and not interstitial fluid
112. Chauhan VP, Stylianopoulos T, Martin JD, et al. normalization by vascular endothelial growth factor pressure. Cancer Cell. 2014;26:14–15.
Normalization of tumour blood vessels improves receptor 2 blockade induces a pressure gradient across
the delivery of nanomedicines in a size-dependent the vasculature and improves drug penetration in
manner. Nat Nanotechnol. 2012;7:383–388. tumors. Cancer Res. 2004;64:3731–3736.
Tumor Microenvironment: Vascular and Extravascular Compartment  •  CHAPTER 8 126.e1
126.e1

REFERENCES
1. Goldman E. The growth of malignant disease in 26. Soda Y, Marumoto T, Friedmann-Morvinski D, 48. Xu L, Fukumura D, Jain RK. Acidic extracellular pH
man and the lower animals with special reference et al. Transdifferentiation of glioblastoma cells into induces vascular endothelial growth factor (VEGF)
to the vascular system. Lancet. 1907;2:1236–1240. vascular endothelial cells. Proc Natl Acad Sci USA. in human glioblastoma cells via ERK1/2 MAPK
2. Algire GH, Chalkley HW. Vascular reactions of 2011;108:4274–4280. signaling pathway: mechanism of low pH-induced
normal and malignant tissues in vivo. I. Vascular 27. Wang R, Chadalavada K, Wilshire J, et al. Glioblas- VEGF. J Biol Chem. 2002;277:11368–11374.
reactions of mice to wounds and to normal and neo- toma stem-like cells give rise to tumour endothelium. 49. Brown EB, Campbell RB, Tsuzuki Y, et al. In
plastic transplants. J Natl Cancer Inst. 1945;6:73–85. Nature. 2010;468:829–833. vivo measurement of gene expression, angiogen-
3. Ide AG, Baker NH, Warren SL. Vascularization of the 28. Kozin SV, Duda DG, Munn LL, et al. Neovascular- esis and physiological function in tumors using
Brown-Pearce rabbit epithelioma transplant as seen ization after irradiation: what is the source of newly multiphoton laser scanning microscopy. Nat Med.
in the transplant ear chamber. Am J Radiol. 1939;42: formed vessels in recurring tumors? J Natl Cancer 2001;7:864–868.
891–899. Inst. 2012;104:899–905. 50. Fukumura D, Jain RK. Role of nitric oxide in
4. Jain RK, Munn LL, Fukumura D. Dissecting tumour 29. Ergun S, Hohn HP, Kilic N, et al. Endothelial and angiogenesis and microcirculation in tumors. Cancer
pathophysiology using intravital microscopy. Nat hematopoietic progenitor cells (EPCs and HPCs): Metastasis Rev. 1998;17:77–89.
Rev Cancer. 2002;2:266–276. hand in hand fate determining partners for cancer 51. Helmlinger G, Endo M, Ferrara N, et  al.
5. Jain RK, Schlenger K, Hockel M, et al. Quantitative cells. Stem Cell Rev. 2008;4:169–177. Formation of endothelial cell networks. Nature.
angiogenesis assays: progress and problems. Nat Med. 30. Aicher A, Rentsch M, Sasaki K, et al. Nonbone 2000;405:139–141.
1997;3:1203–1208. marrow-derived circulating progenitor cells contrib- 52. Tsuzuki Y, Fukumura D, Oosthuyse B, et al. Vascular
6. Helmlinger G, Netti PA, Lichtenbeld HC, et al. ute to postnatal neovascularization following tissue endothelial growth factor (VEGF) modulation by
Solid stress inhibits the growth of multicellular tumor ischemia. Circ Res. 2007;100:581–589. targeting hypoxia-inducible factor-1alpha → hypoxia
spheroids. Nat Biotechnol. 1997;15:778–783. 31. Kozin SV, Duda DG, Munn LL, et al. Is vasculogen- response element → VEGF cascade differentially
7. Folkman J. Tumor angiogenesis: therapeutic implica- esis crucial for the regrowth of irradiated tumours? regulates vascular response and growth rate in
tions. N Engl J Med. 1971;285:1182–1186. Nat Rev Cancer. 2011;11:532. tumors. Cancer Res. 2000;60:6248–6252.
8. Gullino PM. Angiogenesis and oncogenesis. J Natl 32. Dvorak HF, Nagy JA, Feng D, et al. Tumor 53. Blouw B, Song H, Tihan T, et al. The hypoxic
Cancer Inst. 1978;61:639–643. Architecture and Targeted Delivery. In: Abrams PG, response of tumors is dependent on their micro-
9. Hurwitz H, Fehrenbacher L, Novotny W, et al. Fritzberg AR, eds. Radioimmunotherapy of Cancer. environment. Cancer Cell. 2003;4:133–146.
Bevacizumab plus irinotecan, fluorouracil, and New York: Marcel Dekker, Inc.; 2000:107–135. 54. Dellian M, Helmlinger G, Yuan F, et al. Fluorescence
leucovorin for metastatic colorectal cancer. N Engl 33. Ferrara N, Gerber HP, LeCouter J. The biology of ratio imaging of interstitial pH in solid tumours:
J Med. 2004;350:2335–2342. VEGF and its receptors. Nat Med. 2003;9:669–676. effect of glucose on spatial and temporal gradients.
10. Jayson GC, Kerbel R, Ellis LM, et al. Antiangiogenic 34. Melder RJ, Koenig GC, Witwer BP, et al. During Br J Cancer. 1996;74:1206–1215.
therapy in oncology: current status and future direc- angiogenesis, vascular endothelial growth factor 55. Fukumura D, Yuan F, Monsky WL, et al. Effect of host
tions. Lancet. 2016. and basic fibroblast growth factor regulate natural microenvironment on the microcirculation of human
11. Carmeliet P. Angiogenesis in life, disease and killer cell adhesion to tumor endothelium. Nat Med. colon adenocarcinoma. Am J Pathol. 1997;151:
medicine. Nature. 2005;438:932–936. 1996;2:992–997. 679–688.
12. Carmeliet P, Jain RK. Molecular mechanisms 35. Casanovas O, Hicklin DJ, Bergers G, et al. Drug 56. Gohongi T, Fukumura D, Boucher Y, et al. Tumor-
and clinical applications of angiogenesis. Nature. resistance by evasion of antiangiogenic targeting of host interactions in the gallbladder suppress distal
2011;473:298–307. VEGF signaling in late-stage pancreatic islet tumors. angiogenesis and tumor growth: involvement of
13. Jain RK, Carmeliet PF. Vessels of death or life. Sci Cancer Cell. 2005;8:299–309. transforming growth factor beta1. Nat Med. 1999;5:
Am. 2001;285:38–45. 36. Fidler IJ. Angiogenic heterogeneity: regulation of 1203–1208.
14. Bouck N, Stellmach V, Hsu SC. How tumors become neoplastic angiogenesis by the organ microenviron- 57. Hobbs SK, Monsky WL, Yuan F, et al. Regulation of
angiogenic. Adv Cancer Res. 1996;135–174. ment. J Natl Cancer Inst. 2001;93:1040–1041. transport pathways in tumor vessels: role of tumor
15. Hanahan D, Weinberg RA. Hallmarks of cancer: 37. Yoshiji H, Harris SR, Thorgeirsson UP. Vascular type and microenvironment. Proc Natl Acad Sci USA.
the next generation. Cell. 2011;144:646–674. endothelial growth factor is essential for initial but 1998;95:4607–4612.
16. Dvorak HF. Vascular permeability factor/vascular not continued in vivo growth of human breast 58. Monsky WL, Mouta Carreira C, Tsuzuki Y, et al.
endothelial growth factor: a critical cytokine in tumor carcinoma cells. Cancer Res. 1997;57:3924–3928. Role of host microenvironment in angiogenesis and
angiogenesis and a potential target for diagnosis and 38. Jain RK, Duda DG, Willett CG, et al. Biomarkers microvascular functions in human breast cancer
therapy. J Clin Oncol. 2002;20:4368–4380. of response and resistance to antiangiogenic therapy. xenografts: mammary fat pad versus cranial tumors.
17. Patan S, Munn LL, Jain RK. Intussusceptive micro- Nat Rev Clin Oncol. 2009;6:327–338. Clin Cancer Res. 2002;8:1008–1013.
vascular growth in a human colon adenocarcinoma 39. LeCouter J, Ferrara N. EG-VEGF and Bv8: a novel 59. Tsuzuki Y, Mouta Carreira C, Bockhorn M, et al.
xenograft: a novel mechanism of tumor angiogenesis. family of tissue-selective mediators of angiogenesis, Pancreas microenvironment promotes VEGF expres-
Microvasc Res. 1996;51:260–272. endothelial phenotype, and function. Trends Car- sion and tumor growth: novel window models for
18. Patan S, Munn LL, Tanda S, et al. Vascular mor- diovasc Med. 2003;13:276–282. pancreatic tumor angiogenesis and microcirculation.
phogenesis and remodeling in a model of tissue 40. Yancopoulos GD, Davis S, Gale NW, et al. Vascular- Lab Invest. 2001;81:1439–1451.
repair: blood vessel formation and growth in the specific growth factors and blood vessel formation. 60. Yuan F, Salehi HA, Boucher Y, et al. Vascular
ovarian pedicle after ovariectomy. Circ Res. 2001;89: Nature. 2000;407:242–248. permeability and microcirculation of gliomas and
723–731. 41. Maeshima Y, Sudhakar A, Lively JC, et al. Tumstatin, mammary carcinomas transplanted in rat and
19. Patan S, Tanda S, Roberge S, et al. Vascular morpho- an endothelial cell-specific inhibitor of protein mouse cranial windows. Cancer Res. 1994;54:4564–
genesis and remodeling in a human tumor xenograft: synthesis. Science. 2002;295:140–143. 4568.
blood vessel formation and growth after ovariectomy 42. O’Reilly MS, Boehm T, Shing Y, et al. Endostatin: 61. Izumi Y, Xu L, di Tomaso E, et al. Tumour biology:
and tumor implantation. Circ Res. 2001;89:732–739. an endogenous inhibitor of angiogenesis and tumor Herceptin acts as an anti-angiogenic cocktail. Nature.
20. Jain RK. Molecular regulation of vessel maturation. growth. Cell. 1997;88:277–285. 2002;416:279–280.
Nat Med. 2003;9:685–693. 43. O’Reilly MS, Holmgren L, Shing Y, et al. Angio- 62. Jain RK, Safabakhsh N, Sckell A, et al. Endothelial
21. Duda DG, Cohen KS, Kozin SV, et al. Evidence for statin: a novel angiogenesis inhibitor that mediates cell death, angiogenesis, and microvascular function
incorporation of bone marrow-derived endothelial the suppression of metastases by a Lewis lung after castration in an androgen-dependent tumor:
cells into perfused blood vessels in tumors. Blood. carcinoma. Cell. 1994;79:315–328. role of vascular endothelial growth factor. Proc Natl
2006;107:2774–2776. 44. Kalluri R. Basement membranes: structure, assembly Acad Sci USA. 1998;95:10820–10825.
22. Isner JM. Myocardial gene therapy. Nature. and role in tumour angiogenesis. Nat Rev Cancer. 63. Jain RK. Normalization of tumor vasculature: an
2002;415:234. 2003;3:422–433. emerging concept in antiangiogenic therapy. Science.
23. Rafii S, Lyden D, Benezra R, et al. Vascular and 45. Fukumura D, Xu L, Chen Y, et al. Hypoxia and 2005;307:58–62.
haematopoietic stem cells: novel targets for anti- acidosis independently up-regulate vascular endo- 64. Jain RK. Taming vessels to treat cancer. Sci Am.
angiogenesis therapy? Nat Rev Cancer. 2002;2: thelial growth factor transcription in brain tumors 2008;298:56–63.
826–835. in vivo. Cancer Res. 2001;61:6020–6024. 65. Ramanujan S, Koenig GC, Padera TP, et al. Local
24. Sajithlal GB, McGuire TF, Lu J, et al. Endothelial- 46. Garkavtsev I, Kozin SV, Chernova O, et al. The imbalance of proangiogenic and antiangiogenic
like cells derived directly from human tumor candidate tumour suppressor protein ING4 regulates factors: a potential mechanism of focal necrosis
xenografts. Int J Cancer. 2010;127:2268–2278. brain tumour growth and angiogenesis. Nature. and dormancy in tumors. Cancer Res. 2000;60:
25. Ricci-Vitiani L, Pallini R, Biffoni M, et al. Tumour 2004;428:328–332. 1442–1448.
vascularization via endothelial differentiation of glio- 47. Kerbel RS. Tumor angiogenesis. N Engl J Med. 66. Baish JW, Jain RK. Fractals and cancer. Cancer Res.
blastoma stem-like cells. Nature. 2010;468:824–828. 2008;358:2039–2049. 2000;60:3683–3688.
126.e2 Part I: Science and Clinical Oncology

67. Gazit Y, Berk DA, Leunig M, et al. Scale-invariant 88. Endrich B, Reinhold HS, Gross JF, et al. Tissue 110. Yuan F, Dellian M, Fukumura D, et al. Vascular
behavior and vascular network formation in normal perfusion inhomogeneity during early tumor growth permeability in a human tumor xenograft: molecular
and tumor-tissue. Phys Rev Lett. 1995;75:2428– in rats. J Natl Cancer Inst. 1979;62:387–395. size dependence and cutoff size. Cancer Res. 1995;55:
2431. 89. Jain RK, Forbes NS. Can engineered bacteria 3752–3756.
68. Gazit Y, Baish JW, Safabakhsh N, et al. Fractal charac- help control cancer? Proc Natl Acad Sci USA. 111. Dreher MR, Liu W, Michelich CR, et al. Tumor
teristics of tumor vascular architecture during tumor 2001;98:14748–14750. vascular permeability, accumulation, and penetration
growth and regression. Microcirculation. 1997;4: 90. Leunig M, Yuan F, Menger MD, et al. Angiogenesis, of macromolecular drug carriers. J Natl Cancer Inst.
395–402. microvascular architecture, microhemodynamics, 2006;98:335–344.
69. McDonald DM, Choyke PL. Imaging of angio- and interstitial fluid pressure during early growth 112. Chauhan VP, Stylianopoulos T, Martin JD, et al.
genesis: from microscope to clinic. Nat Med. of human adenocarcinoma LS174T in SCID mice. Normalization of tumour blood vessels improves
2003;9:713–725. Cancer Res. 1992;52:6553–6560. the delivery of nanomedicines in a size-dependent
70. Jain RK. Determinants of tumor blood flow: a review. 91. Brizel DM, Klitzman B, Cook JM, et al. A com- manner. Nat Nanotechnol. 2012;7:383–388.
Cancer Res. 1988;48:2641–2658. parison of tumor and normal tissue microvascular 113. Chauhan VP, Popovic Z, Chen O, et al. Fluorescent
71. Less JR, Posner MC, Skalak TC, et al. Geometric hematocrits and red cell fluxes in a rat window nanorods and nanospheres for real-time in vivo
resistance and microvascular network architecture chamber model. Int J Radiat Oncol Biol Phys. 1993;25: probing of nanoparticle shape-dependent tumor
of human colorectal carcinoma. Microcirculation. 269–276. penetration. Angew Chem Int Ed Engl. 2011;50:
1997;4:25–33. 92. Baish JW, Netti PA, Jain RK. Transmural coupling 11417–11420.
72. Less JR, Skalak TC, Sevick EM, et al. Microvascular of fluid flow in microcirculatory network and inter- 114. Campbell RB, Fukumura D, Brown EB, et al. Cat-
architecture in a mammary carcinoma: branch- stitium in tumors. Microvasc Res. 1997;53:128–141. ionic charge determines the distribution of liposomes
ing patterns and vessel dimensions. Cancer Res. 93. Netti PA, Roberge S, Boucher Y, et al. Effect of between the vascular and extravascular compartments
1991;51:265–273. transvascular fluid exchange on pressure-flow of tumors. Cancer Res. 2002;62:6831–6836.
73. Jain RK, Munn LL. Leaky vessels? Call Angl1! Nat relationship in tumors: a proposed mechanism 115. Dellian M, Yuan F, Trubetskoy VS, et al. Vascular
Med. 2000;6:131–132. for tumor blood flow heterogeneity. Microvasc Res. permeability in a human tumour xenograft: molecu-
74. Goel S, Duda DG, Xu L, et al. Normalization of 1996;52:27–46. lar charge dependence. Br J Cancer. 2000;82:1513–
the vasculature for treatment of cancer and other 94. Mollica F, Jain RK, Netti PA. A model for temporal 1518.
diseases. Physiol Rev. 2011;91:1071–1121. heterogeneities of tumor blood flow. Microvasc Res. 116. Stylianopoulos T, Soteriou K, Fukumura D, et al.
75. Goel S, Fukumura D, Jain RK. Normalization of 2003;65:56–60. Cationic nanoparticles have superior transvascular
the tumor vasculature through oncogenic inhibition: 95. Park MS, Klotz E, Kim MJ, et al. Perfusion CT: flux into solid tumors: insights from a mathematical
an emerging paradigm in tumor biology. Proc Natl noninvasive surrogate marker for stratification of model. Ann Biomed Eng. 2012.
Acad Sci USA. 2012;doi:10.1073/pnas.1203794109. pancreatic cancer response to concurrent chemo- 117. Thurston G, McLean JW, Rizen M, et al. Cationic
76. Huang Y, Yuan J, Righi E, et al. Vascular normalizing and radiation therapy. Radiology. 2009;250:110– liposomes target angiogenic endothelial cells in
dose of antiangiogenic treatment reprograms the 117. tumors and chronic inflammation in mice. J Clin
immunosuppressive tumor microenvironment and 96. Sorensen AG, Emblem KE, Polaskova P, et al. Invest. 1998;101:1401–1413.
enhances immunotherapy. Proc Natl Acad Sci USA. Increased survival of glioblastoma patients who 118. Monsky WL, Fukumura D, Gohongi T, et al.
2012;109:17561–17566. respond to antiangiogenic therapy with elevated Augmentation of transvascular transport of macro-
77. Kadambi A, Mouta Carreira C, Yun CO, et al. blood perfusion. Cancer Res. 2012;72:402–407. molecules and nanoparticles in tumors using vascular
Vascular endothelial growth factor (VEGF)-C 97. Morikawa S, Baluk P, Kaidoh T, et al. Abnormalities endothelial growth factor. Cancer Res. 1999;59:
differentially affects tumor vascular function and in pericytes on blood vessels and endothelial sprouts 4129–4135.
leukocyte recruitment: role of VEGF-receptor 2 and in tumors. Am J Pathol. 2002;160:985–1000. 119. Yuan F, Chen Y, Dellian M, et al. Time-dependent
host VEGF-A. Cancer Res. 2001;61:2404–2408. 98. Dolmans DE, Kadambi A, Hill JS, et al. Vascular vascular regression and permeability changes in
78. Koike C, McKee TD, Pluen A, et al. Solid stress accumulation of a novel photosensitizer, MV6401, established human tumor xenografts induced by
facilitates spheroid formation: potential involvement causes selective thrombosis in tumor vessels after pho- an anti-vascular endothelial growth factor/vascular
of hyaluronan. Br J Cancer. 2002;86:947–953. todynamic therapy. Cancer Res. 2002;62:2151–2156. permeability factor antibody. Proc Natl Acad Sci
79. Stylianopoulos T, Martin JD, Chauhan VP, et al. 99. Huang X, Molema G, King S, et al. Tumor infarction USA. 1996;93:14765–14770.
Causes, consequences and remedies for growth- in mice by antibody-directed targeting of tissue factor 120. Bockhorn M, Roberge S, Sousa C, et al. Differential
induced solid stress in murine and human tumors. to tumor vasculature. Science. 1997;275:547–550. gene expression in metastasizing cells shed from
Proc Natl Acad Sci USA. 2012;109(38):15101– 100. Ruoslahti E. Specialization of tumor vasculature. kidney tumors. Cancer Res. 2004;64:2469–2473.
15108. Nat Rev Cancer. 2002;2:83–90. 121. Chang YS, di Tomaso E, McDonald DM, et al.
80. Griffon-Etienne G, Boucher Y, Brekken C, et al. 101. Jain RK. Normalizing tumor vasculature with anti- Mosaic blood vessels in tumors: Frequency of cancer
Taxane-induced apoptosis decompresses blood angiogenic therapy: a new paradigm for combination cells in contact with flowing blood. Proc Natl Acad
vessels and lowers interstitial fluid pressure in solid therapy. Nat Med. 2001;7:987–989. Sci USA. 2000;97:14608–14613.
tumors: clinical implications. Cancer Res. 1999;59: 102. Chauhan VP, Stylianopoulos T, Boucher Y, et al. 122. Gullino PM. Techniques in tumor pathophysiology.
3776–3782. Delivery of molecular and nanoscale medicine to In: Busch H, ed. Methods in Cancer Research. New
81. Padera TP, Stoll BR, Tooredman JB, et al. Pathology: tumors: transport barriers and strategies. Annu Rev York: Academic Press; 1970:45–92.
cancer cells compress intratumour vessels. Nature. Chem Biomol Eng. 2011;2:281–298. 123. Swartz MA, Kristensen CA, Melder RJ, et al. Cells
2004;427:695. 103. Jain RK. Transport of molecules across tumor shed from tumours show reduced clonogenicity,
82. Jacobetz MA, Chan DS, Neesse A, et  al. vasculature. Cancer Metastasis Rev. 1987;6:559–593. resistance to apoptosis, and in vivo tumorigenicity.
Hyaluronan impairs vascular function and drug 104. Hashizume H, Baluk P, Morikawa S, et al. Openings Br J Cancer. 1999;81:756–759.
delivery in a mouse model of pancreatic cancer. between defective endothelial cells explain tumor 124. Duda DG, Duyverman AM, Kohno M, et al.
Gut. 2013;62(1):112–120. vessel leakiness. Am J Pathol. 2000;156:1363–1380. Malignant cells facilitate lung metastasis by bringing
83. Sevick EM, Jain RK. Geometric resistance to blood 105. Endo M, Jain RK, Witwer B, et al. Water channel their own soil. Proc Natl Acad Sci USA. 2010;107:
flow in solid tumors perfused ex vivo: effects of (aquaporin 1) expression and distribution in mammary 21677–21682.
tumor size and perfusion pressure. Cancer Res. carcinomas and glioblastomas. Microvasc Res. 1999; 125. Jain RK, Koenig GC, Dellian M, et al. Leukocyte-
1989;49:3506–3512. 58:89–98. endothelial adhesion and angiogenesis in tumors.
84. Sevick EM, Jain RK. Viscous resistance to blood flow 106. Gerlowski LE, Jain RK. Microvascular permeability Cancer Metastasis Rev. 1996;15:195–204.
in solid tumors: effect of hematocrit on intratumor of normal and neoplastic tissues. Microvasc Res. 126. Melder RJ, Munn LL, Yamada S, et al. Selectin- and
blood viscosity. Cancer Res. 1989;49:3513–3519. 1986;31:288–305. integrin-mediated T-lymphocyte rolling and arrest
85. Sevick EM, Jain RK. Effect of red blood cell rigidity 107. Lichtenbeld HC, Yuan F, Michel CC, et al. Perfu- on TNF-alpha-activated endothelium: augmentation
on tumor blood flow: increase in viscous resistance sion of single tumor microvessels: application to by erythrocytes. Biophys J. 1995;69:2131–2138.
during hyperglycemia. Cancer Res. 1991;51: vascular permeability measurement. Microcirculation. 127. Melder RJ, Yuan J, Munn LL, et al. Erythrocytes
2727–2730. 1996;3:349–357. enhance lymphocyte rolling and arrest in vivo.
86. Jain RK, Ward-Hartley KA: Tumor blood flow: char- 108. Sevick EM, Jain RK. Measurement of capillary Microvasc Res. 2000;59:316–322.
acterization, modifications and role in hyperthermia. filtration coefficient in a solid tumor. Cancer Res. 128. Migliorini C, Qian Y, Chen H, et al. Red blood
IEEE Transactions in Sonics and Ultrasonics; Special 1991;51:1352–1355. cells augment leukocyte rolling in a virtual blood
Issue on Hyperthermia, SU-31:504-526, 1984. 109. Yuan F, Leunig M, Berk DA, et al. Microvascular vessel. Biophys J. 2002;83:1834–1841.
87. Vaupel P, Kallinowski F, Okunieff P. Blood flow, permeability of albumin, vascular surface area, and 129. Munn LL, Melder RJ, Jain RK. Role of erythrocytes
oxygen and nutrient supply, and metabolic micro- vascular volume measured in human adenocarcinoma in leukocyte-endothelial interactions: mathemati-
environment of human tumors: a review. Cancer LS174T using dorsal chamber in SCID mice. cal model and experimental validation. Biophys J.
Res. 1989;49:6449–6465. Microvasc Res. 1993;45:269–289. 1996;71:466–478.
Tumor Microenvironment: Vascular and Extravascular Compartment  •  CHAPTER 8 126.e3
126.e3

130. Yuan J, Melder RJ, Jain RK, et al. Lateral view flow microenvironment. Cancer Cell. 2012;21:309– in tumors. Proc Natl Acad Sci USA. 2011;108:
system for studies of cell adhesion and deformation 322. 2909–2914.
under flow conditions. Biotechniques. 2001;30: 151. DeNardo DG, Andreu P, Coussens LM. Interactions 173. McKee TD, Grandi P, Mok W, et al. Degradation
388–394. between lymphocytes and myeloid cells regulate pro- of fibrillar collagen in a human melanoma xenograft
131. Melder RJ, Kristensen CA, Munn LL, et al. Modula- versus anti-tumor immunity. Cancer and Metastasis improves the efficacy of an oncolytic herpes simplex
tion of A-NK cell rigidity: In vitro characterization Reviews. 2010;29:309–316. virus vector. Cancer Res. 2006;66:2509–2513.
and in vivo implications for cell delivery. Biorheology. 152. Coussens LM, Zitvogel L, Palucka AK. Neutralizing 174. Netti PA, Berk DA, Swartz MA, et al. Role of
2001;38:151–159. tumor-promoting chronic inflammation: a magic extracellular matrix assembly in interstitial transport
132. Al-Mehdi AB, Tozawa K, Fisher AB, et al. Intra- bullet? Science. 2013;339:286–291. in solid tumors. Cancer Res. 2000;60:2497–2503.
vascular origin of metastasis from the proliferation 153. Fukumura D, Xavier R, Sugiura T, et al. Tumor 175. Ramanujan S, Pluen A, McKee TD, et al. Diffusion
of endothelium-attached tumor cells: a new model induction of VEGF promoter activity in stromal and convection in collagen gels: implications for trans-
for metastasis. Nat Med. 2000;6:100–102. cells. Cell. 1998;94:715–725. port in the tumor interstitium. Biophys J. 2002;83:
133. Fukumura D, Salehi HA, Witwer B, et al. Tumor 154. Jain RK, Fenton BT. Intratumoral lymphatic vessels: 1650–1660.
necrosis factor alpha-induced leukocyte adhesion a case of mistaken identity or malfunction? J Natl 176. Stylianopoulos T, Poh MZ, Insin N, et al. Diffusion
in normal and tumor vessels: effect of tumor type, Cancer Inst. 2002;94:417–421. of particles in the extracellular matrix: the effect
transplantation site, and host strain. Cancer Res. 155. Padera TP, Kadambi A, di Tomaso E, et al. Lymphatic of repulsive electrostatic interactions. Biophys J.
1995;55:4824–4829. metastasis in the absence of functional intratumor 2010;99:1342–1349.
134. Dellian M, Witwer BP, Salehi HA, et al. Quantitation lymphatics. Science. 2002;296:1883–1886. 177. Baxter LT, Jain RK. Transport of fluid and macro-
and physiological characterization of angiogenic 156. Elenbaas B, Weinberg RA. Heterotypic signaling molecules in tumors. I. Role of interstitial pressure
vessels in mice—effect of basic fibroblast growth between epithelial tumor cells and fibroblasts in car- and convection. Microvasc Res. 1989;37:77–104.
factor vascular endothelial growth factor vascular cinoma formation. Exp Cell Res. 2001;264:169–184. 178. Baxter LT, Jain RK. Transport of fluid and mac-
permeability factor, and host microenvironment. 157. Jain RK. Transport of molecules in the tumor inter- romolecules in tumors. III. Role of binding and
Am J Pathol. 1996;149:59–71. stitium: a review. Cancer Res. 1987;47:3039–3051. metabolism. Microvasc Res. 1991;41:5–23.
135. Yamada S, Mayadas TN, Yuan F, et al. Rolling in 158. Berk DA, Yuan F, Leunig M, et al. Fluorescence 179. Juweid M, Neumann R, Paik C, et al. Micro-
P-selectin-deficient mice is reduced but not elimi- photobleaching with spatial Fourier analysis: pharmacology of monoclonal antibodies in solid
nated in the dorsal skin. Blood. 1995;86:3487–3492. measurement of diffusion in light-scattering media. tumors: direct experimental evidence for a binding
136. Melder RJ, Brownell AL, Shoup TM, et al. Imaging Biophys J. 1993;65:2428–2436. site barrier. Cancer Res. 1992;52:5144–5153.
of activated natural killer cells in mice by positron 159. Brown E, McKee T, diTomaso E, et al. Dynamic 180. Kaufman EN, Jain RK. Quantification of transport
emission tomography: preferential uptake in tumors. imaging of collagen and its modulation in tumors and binding parameters using fluorescence recovery
Cancer Res. 1993;53:5867–5871. in vivo using second-harmonic generation. Nat Med. after photobleaching. Potential for in vivo applica-
137. Melder RJ, Jain RK. Reduction of rigidity in human 2003;9:796–800. tions. Biophys J. 1990;58:873–885.
activated natural killer cells by thioglycollate treat- 160. Chary SR, Jain RK. Direct measurement of interstitial 181. Kaufman EN, Jain RK. Measurement of mass
ment. J Immunol Methods. 1994;175:69–77. convection and diffusion of albumin in normal and transport and reaction parameters in bulk solution
138. Melder RJ, Jain RK. Kinetics of interleukin-2 induced neoplastic tissues by fluorescence photobleaching. using photobleaching: Reaction limited binding
changes in rigidity of human natural killer cells. Cell Proc Natl Acad Sci USA. 1989;86:5385–5389. regime. Biophys J. 1991;60:596–610.
Biophys. 1992;20:161–176. 161. Chauhan VP, Lanning RM, Diop-Frimpong B, et al. 182. Kaufman EN, Jain RK. Effect of bivalent interaction
139. Sasaki A, Jain RK, Maghazachi AA, et al. Low Multiscale measurements distinguish cellular and upon apparent antibody affinity: experimental confir-
deformability of lymphokine-activated killer cells interstitial hindrances to diffusion in vivo. Biophys mation of theory using fluorescence photobleaching
as a possible determinant of in vivo distribution. J. 2009;97:330–336. and implications for antibody binding assays. Cancer
Cancer Res. 1989;49:3742–3746. 162. Davies CL, Berk DA, Pluen A, et al. Comparison of Res. 1992;52:4157–4167.
140. Melder RJ, Salehi HA, Jain RK. Interaction of IgG diffusion and extracellular matrix composition 183. Kaufman EN, Jain RK. In vitro measurement and
activated natural killer cells with normal and tumor in rhabdomyosarcomas grown in mice versus in vitro screening of monoclonal antibody affinity using
vessels in cranial windows in mice. Microvasc Res. as spheroids reveals the role of host stromal cells. fluorescence photobleaching. J Immunol Methods.
1995;50:35–44. Br J Cancer. 2002;86:1639–1644. 1992;155:1–17.
141. Munn LL, Melder RJ, Jain RK. Analysis of cell flux 163. Johnson EM, Berk DA, Jain RK, et al. Diffusion 184. Gullino PM. Extracellular compartments of solid
in the parallel plate flow chamber: implications for and partitioning of proteins in charged agarose gels. tumors. In: Becker FF, ed. Cancer. New York:
cell capture studies. Biophys J. 1994;67:889–895. Biophys J. 1995;68:1561–1568. Plenum; 1975:327–354.
142. Munn LL, Koenig GC, Jain RK, et al. Kinetics of adhe- 164. Johnson EM, Berk DA, Jain RK, et al. Hindered 185. Leu AJ, Berk DA, Lymboussaki A, et al. Absence
sion molecule expression and spatial organization using diffusion in agarose gels: test of effective medium of functional lymphatics within a murine sarcoma:
targeted sampling fluorometry. Biotechniques. 1995; model. Biophys J. 1996;70:1017–1023. a molecular and functional evaluation. Cancer Res.
19:622–631. 165. Nugent LJ, Jain RK. Extravascular diffusion in 2000;60:4324–4327.
143. Sasaki A, Melder RJ, Whiteside TL, et al. Preferential normal and neoplastic tissues. Cancer Res. 1984;44: 186. Carreira CM, Nasser SM, di Tomaso E, et al. LYVE-1
localization of human adherent lymphokine-activated 238–244. is not restricted to the lymph vessels: Expression in
killer cells in tumor microcirculation. J Natl Cancer 166. Pluen A, Boucher Y, Ramanujan S, et al. Role of normal liver blood sinusoids and down-regulation
Inst. 1991;83:433–437. tumor-host interactions in interstitial diffusion of in human liver cancer and cirrhosis. Cancer Res.
144. Melder RJ, Koenig GC, Munn LL, et al. Adhesion macromolecules: cranial vs. subcutaneous tumors. 2001;61:8079–8084.
of activated natural killer cells to tumor necrosis Proc Natl Acad Sci USA. 2001;98:4628–4633. 187. Hoshida T, Isaka N, Hagendoorn J, et al. Imaging
factor-alpha-treated endothelium under physiological 167. Pluen A, Netti PA, Jain RK, et al. Diffusion of steps of lymphatic metastasis reveals that vascular
flow conditions. Nat Immun. 1996;15:154–163. macromolecules in agarose gels: comparison of linear endothelial growth factor-C increases metastasis
145. Gamble JR, Khew-Goodall Y, Vadas MA. Trans- and globular configurations. Biophys J. 1999;77: by increasing delivery of cancer cells to lymph
forming growth factor-beta inhibits E-selectin 542–552. nodes: therapeutic implications. Cancer Res.
expression on human endothelial cells. J Immunol. 168. Swabb EA, Wei J, Gullino PM. Diffusion and 2006;66:8065–8075.
1993;150:4494–4503. convection in normal and neoplastic tissues. Cancer 188. Alitalo K, Carmeliet P. Molecular mechanisms of
146. Gamble JR, Vadas MA. Endothelial adhesiveness Res. 1974;34:2814–2822. lymphangiogenesis in health and disease. Cancer
for blood neutrophils is inhibited by transforming 169. Boucher Y, Brekken C, Netti PA, et al. Intratumoral Cell. 2002;1:219–227.
growth factor-beta. Science. 1988;242:97–99. infusion of fluid: estimation of hydraulic conductivity 189. Jain RK, Padera TP. Development. Lymphatics make
147. Gamble JR, Vadas MA. Endothelial cell adhesiveness and implications for the delivery of therapeutic the break. Science. 2003;299:209–210.
for human T lymphocytes is inhibited by transform- agents. Br J Cancer. 1998;78:1442–1448. 190. Oliver G, Detmar M. The rediscovery of the
ing growth factor-beta 1. J Immunol. 1991;146: 170. Znati CA, Rosenstein M, McKee TD, et al. Irradia- lymphatic system: old and new insights into the
1149–1154. tion reduces interstitial fluid transport and increases development and biological function of the lym-
148. Jallal B, Powell J, Zachwieja J, et al. Suppression of the collagen content in tumors. Clin Cancer Res. phatic vasculature. Genes Dev. 2002;16:773–783.
tumor growth in vivo by local and systemic 90K 2003;9:5508–5513. 191. Gale NW, Thurston G, Hackett SF, et al. Angio-
level increase. Cancer Res. 1995;55:3223–3227. 171. Berk DA, Yuan F, Leunig M, et al. Direct in vivo poietin-2 is required for postnatal angiogenesis and
149. Detmar M, Brown LF, Schon MP, et al. Increased measurement of targeted binding in a human lymphatic patterning, and only the latter role is
microvascular density and enhanced leukocyte rolling tumor xenograft. Proc Natl Acad Sci USA. 1997;94: rescued by angiopoietin-1. Development Cell. 2002;3:
and adhesion in the skin of VEGF transgenic mice. 1785–1790. 411–423.
J Invest Dermatol. 1998;111:1–6. 172. Diop-Frimpong B, Chauhan VP, Krane S, et al. 192. Jeong HS, Jones D, Liao S, et al. Investigation of the
150. Hanahan D, Coussens LM. Accessories to the Losartan inhibits collagen I synthesis and improves lack of angiogenesis in the formation of lymph node
crime: functions of cells recruited to the tumor the distribution and efficacy of nanotherapeutics metastases. J Natl Cancer Inst. 2015;107:djv155.
126.e4 Part I: Science and Clinical Oncology

193. Jain RK, Padera TP. Prevention and treatment 214. Milosevic M, Fyles A, Hedley D, et al. Interstitial 233. Brown JM, Giaccia AJ. The unique physiology of
of lymphatic metastasis by antilymphangiogenic fluid pressure predicts survival in patients with cervix solid tumors: opportunities (and problems) for cancer
therapy. J Natl Cancer Inst. 2002;94:785–787. cancer independent of clinical prognostic factors and therapy. Cancer Res. 1998;58:1408–1416.
194. Padera TP, Stoll BR, So PT, et al. Conventional tumor: oxygen measurements. Cancer Res. 2001; 234. Dewhirst MW. Concepts of oxygen transport at
and high-speed intravital multiphoton laser scanning 61:6400–6405. the microcirculatory level. Semin Radiat Oncol.
microscopy of microvasculature, lymphatics, and leu- 215. Boucher Y, Leunig M, Jain RK. Tumor angio- 1998;8:143–150.
kocyte-endothelial interactions. Mol Imaging. 2002; genesis and interstitial hypertension. Cancer Res. 235. Helmlinger G, Sckell A, Dellian M, et al. Acid
1:9–15. 1996;56:4264–4266. production in glycolysis-impaired tumors provides
195. Leu AJ, Berk DA, Yuan F, et al. Flow velocity in 216. Lee CG, Heijn M, di Tomaso E, et al. Anti-vascular new insights into tumor metabolism. Clin Cancer
the superficial lymphatic network of the mouse tail. endothelial growth factor treatment augments tumor Res. 2002;8:1284–1291.
Am J Physiol. 1994;267:H1507–H1513. radiation response under normoxic or hypoxic 236. Harris AL. Hypoxia: a key regulatory factor in
196. Berk DA, Swartz MA, Leu AJ, et al. Transport conditions. Cancer Res. 2000;60:5565–5570. tumour growth. Nat Rev Cancer. 2002;2:38–
in lymphatic capillaries. II. Microscopic velocity 217. Tong RT, Boucher Y, Kozin SV, et al. Vascular 47.
measurement with fluorescence photobleaching. Am normalization by vascular endothelial growth factor 237. Nordsmark M, Bentzen SM, Rudat V, et al. Prog-
J Physiol. 1996;270:H330–H337. receptor 2 blockade induces a pressure gradient across nostic value of tumor oxygenation in 397 head and
197. Swartz MA, Berk DA, Jain RK. Transport in lym- the vasculature and improves drug penetration in neck tumors after primary radiation therapy. An
phatic capillaries. I. Macroscopic measurements using tumors. Cancer Res. 2004;64:3731–3736. international multi-center study. Radiother Oncol.
residence time distribution theory. Am J Physiol. 218. DiResta GR, Lee J, Healey JH, et al. Artificial 2005;77:18–24.
1996;270:H324–H329. lymphatic system”: a new approach to reduce inter- 238. Giatromanolaki A, Koukourakis MI, Sivridis E,
198. Jeltsch M, Kaipainen A, Joukov V, et al. Hyperplasia stitial hypertension and increase blood flow, pH and et al. Expression of hypoxia-inducible carbonic
of lymphatic vessels in VEGF-C transgenic mice. pO(2) in solid tumors. Ann Biomed Eng. 2000;28: anhydrase-9 relates to angiogenic pathways and
Science. 1997;276:1423–1425. 543–555. independently to poor outcome in non-small cell
199. Isaka N, Padera TP, Hagendoorn J, et al. Peritumor 219. Stohrer M, Boucher Y, Stangassinger M, et al. lung cancer. Cancer Res. 2001;61:7992–7998.
lymphatics induced by vascular endothelial growth fac- Oncotic pressure in solid tumors is elevated. Cancer 239. Koukourakis M, Giatromanolaki A, Sivridis E, et al.
tor-C exhibit abnormal function. Cancer Res. 2004;64: Res. 2000;60:4251–4255. Lactate dehydrogenase-5 (LDH-5) overexpression
4400–4404. 220. Raut CP, Boucher Y, Duda DG, et al. Effects of in non-small-cell lung cancer tissues is linked to
200. Fukumura D, Duda DG, Munn LL, et al. Tumor sorafenib on intra-tumoral interstitial fluid pressure tumour hypoxia, angiogenic factor production and
microvasculature and microenvironment: novel and circulating biomarkers in patients with refrac- poor prognosis. Br J Cancer. 2003;89:877–885.
insights through intravital imaging in pre-clinical tory sarcomas (NCI protocol 6948). PLoS ONE. 240. Vaupel P, Mayer A. Hypoxia in tumors: pathogenesis-
models. Microcirculation. 2010;17:206–225. 2012;7:e26331. related classification, characterization of hypoxia
201. Liao S, Cheng G, Conner DA, et al. Impaired 221. Willett CG, Boucher Y, di Tomaso E, et al. subtypes, and associated biological and clinical impli-
lymphatic contraction associated with immuno- Direct evidence that the VEGF-specific antibody cations. Oxygen Transport to Tissue XXXVI. 2014;
suppression. Proc Natl Acad Sci USA. 2011;108: bevacizumab has antivascular effects in human rectal (Springer):19–24.
18784–18789. cancer. Nat Med. 2004;10:145–147. 241. Estrella V, Chen T, Lloyd M, et al. Acidity generated
202. Munn LL, Padera TP. Imaging the lymphatic system. 222. Willett CG, Boucher Y, Duda DG, et al. Surrogate by the tumor microenvironment drives local invasion.
Microvasc Res. 2014;96:55–63. markers for antiangiogenic therapy and dose-limiting Cancer Res. 2013;73:1524–1535.
203. Boucher Y, Baxter LT, Jain RK. Interstitial pressure toxicities for bevacizumab with radiation and 242. Martin JD, Fukumura D, Duda DG, et al.
gradients in tissue-isolated and subcutaneous tumors: chemotherapy: continued experience of a phase I Reengineering the tumor microenvironment to
implications for therapy. Cancer Res. 1990;50: trial in rectal cancer patients. J Clin Oncol. 2005;23: alleviate hypoxia and overcome cancer heterogeneity.
4478–4484. 8136–8139. Cold Spring Harbor Perspectives in Medicine. 2016;
204. Boucher Y, Kirkwood JM, Opacic D, et al. Interstitial 223. Willett CG, Duda DG, di Tomaso E, et al. Efficacy, a027094.
hypertension in superficial metastatic melanomas in safety, and biomarkers of neoadjuvant bevacizumab, 243. Spivak-Kroizman TR, Hostetter G, Posner R, et al.
humans. Cancer Res. 1991;51:6691–6694. radiation therapy, and fluorouracil in rectal cancer: Hypoxia triggers hedgehog-mediated tumor-stromal
205. Roh HD, Boucher Y, Kalnicki S, et al. Interstitial a multidisciplinary phase II study. J Clin Oncol. interactions in pancreatic cancer. Cancer Res. 2013;73:
hypertension in carcinoma of uterine cervix in 2009;27:3020–3026. 3235–3247.
patients: possible correlation with tumor oxygenation 224. Netti PA, Baxter LT, Boucher Y, et al. Time- 244. Wenes M, Shang M, Di Matteo M, et al. Macrophage
and radiation response. Cancer Res. 1991;51:6695– dependent behavior of interstitial fluid pressure in metabolism controls tumor blood vessel morphogen-
6698. solid tumors: implications for drug delivery. Cancer esis and metastasis. Cell Metab. 2016;24:701–715.
206. Boucher Y, Jain RK. Microvascular pressure is the Res. 1995;55:5451–5458. 245. Brizel DM, Scully SP, Harrelson JM, et al. Tumor
principal driving force for interstitial hypertension 225. Netti PA, Hamberg LM, Babich JW, et al. Enhance- oxygenation predicts for the likelihood of distant
in solid tumors: implications for vascular collapse. ment of fluid filtration across tumor vessels: implica- metastases in human soft tissue sarcoma. Cancer
Cancer Res. 1992;52:5110–5114. tion for delivery of macromolecules. Proc Natl Acad Res. 1996;56:941–943.
207. Gutmann R, Leunig M, Feyh J, et al. Interstitial Sci USA. 1999;96:3137–3142. 246. Brown JM. The hypoxic cell: a target for selective
hypertension in head and neck tumors in 226. Zlotecki RA, Boucher Y, Lee I, et al. Effect of angio- cancer therapy. Eighteenth Bruce F. Cain Memorial
patients: correlation with tumor size. Cancer Res. tensin II induced hypertension on tumor blood flow Award lecture. Cancer Res. 1999;59:5863–5870.
1992;52:1993–1995. and interstitial fluid pressure. Cancer Res. 1993;53: 247. Cowan DS, Tannock IF. Factors that influence the
208. Less JR, Posner MC, Boucher Y, et al. Interstitial 2466–2468. penetration of methotrexate through solid tissue.
hypertension in human breast and colorectal tumors. 227. Butler TP, Grantham FH, Gullino PM. Bulk transfer Int J Cancer. 2001;91:120–125.
Cancer Res. 1992;52:6371–6374. of fluid in interstitial compartment of mammary 248. Kozin SV, Boucher Y, Hicklin DJ, et al. Vascular
209. Curti BD, Urba WJ, Alvord WG, et al. Interstitial tumors. Cancer Res. 1975;35:3084–3088. endothelial growth factor receptor-2-blocking anti-
pressure of subcutaneous nodules in melanoma 228. Jain RK, Tong RT, Munn LL. Effect of vascular body potentiates radiation-induced long-term control
and lymphoma patients: changes during treatment. normalization by antiangiogenic therapy on of human tumor xenografts. Cancer Res. 2001;61:
Cancer Res. 1993;53:2204–2207. interstitial hypertension, peritumor edema, and 39–44.
210. Arbit E, Lee J, DiResta GR. Interstitial hypertension lymphatic metastasis: insights from a mathematical 249. Vukovic V, Tannock IF. Influence of low pH
in human brain tumors: possible role in peritumoral model. Cancer Res. 2007;67:2729–2735. on cytotoxicity of paclitaxel, mitoxantrone and
edema formulation. In: Nagai H, Kamiya K, Ishi 229. Krogh A. The Anatomy and Physiology of Capillaries. topotecan. Br J Cancer. 1997;75:1167–1172.
S, eds. Intracranial Pressure. Tokyo: Spirnger-Verlag; New York: Yale University Press; 1922. 250. Clever D, Roychoudhuri R, Constantinides MG,
1994:604–619. 230. Thomlinson RH, Gray LH. The histological et al. Oxygen sensing by T cells establishes an
211. Nathanson SD, Nelson L. Interstitial fluid pressure structure of some human lung cancers and the immunologically tolerant metastatic niche. Cell.
in breast-cancer, benign breast conditions, and possible implications for radiotherapy. Br J Cancer. 2016;166:1117–1131, e14.
breast parenchyma. Ann Surg Oncol. 1994;1:333– 1955;9:539–549. 251. Noman MZ, Desantis G, Janji B, et al. PD-L1 is
338. 231. Helmlinger G, Yuan F, Dellian M, et al. Interstitial a novel direct target of HIF-1α, and its blockade
212. Boucher Y, Lee I, Jain RK. Lack of general correla- pH and pO2 gradients in solid tumors in vivo: under hypoxia enhanced MDSC-mediated T cell
tion between interstitial fluid pressure and oxygen high-resolution measurements reveal a lack of activation. J Exp Med. 2014;211:781–790.
partial pressure in solid tumors. Microvasc Res. correlation. Nat Med. 1997;3:177–182. 252. El-Kenawi AE, Ibrahim-Hashim AA, Luddy KA,
1995;50:175–182. 232. Torres-Filho IP, Leunig M, Yuan F, et al. Noninvasive et al. Extracellular acidosis alters polarization of
213. Boucher Y, Salehi H, Witwer B, et al. Interstitial measurement of microvascular and interstitial oxygen macrophages. Cancer Res. 2015;75:3213.
fluid pressure in intracranial tumours in patients profiles in a human tumor in SCID mice. Proc Natl 253. Noman MZ, Hasmim M, Messai Y, et al. Hypoxia: a
and in rodents. Br J Cancer. 1997;75:829–836. Acad Sci USA. 1994;91:2081–2085. key player in antitumor immune response. A review
Tumor Microenvironment: Vascular and Extravascular Compartment  •  CHAPTER 8 126.e5
126.e5

in the theme: cellular responses to hypoxia. Am J neoadjuvant bevacizumab and chemotherapy in breast 292. Patenaude A, Woerher S, Umlandt P, et al. A novel
Physiol Cell Physiol. 2015;309:C569–C579. cancer patients. Proc Natl Acad Sci USA. 2015;112: population of local pericyte precursor cells in tumor
253a.  Fukumura D, Kloepper J, Amoozgar Z, Duda DG, 14325–14330. stroma that require Notch signaling for differentia-
Jain RK. Enhancing cancer immunotherapy using 274. Vasudev NS, Reynolds AR. Anti-angiogenic therapy tion. Microvasc Res. 2015;101:38–47.
antiangiogenics: opportunities and challenges. Nat for cancer: current progress, unresolved questions and 293. Ueda S, Roblyer D, Cerussi A, et al. Baseline tumor
Rev Clin Oncol. 2018;15:325–340. future directions. Angiogenesis. 2014;17:471–494. oxygen saturation correlates with a pathologic com-
254. Martin JD, Fukumura D, Duda DG, et al. Reen- 275. Smith NR, Baker D, Farren M, et al. Tumor plete response in breast cancer patients undergoing
gineering the tumor microenvironment to alleviate stromal architecture can define the intrinsic tumor neoadjuvant chemotherapy. Cancer Res. 2012;72:
hypoxia and overcome cancer heterogeneity. Cold response to VEGF-targeted therapy. Clin Cancer 4318–4328.
Spring Harbor Perspectives in Medicine. 2016;6: Res. 2013;19:6943–6956. 294. Ueda S, Kuji I, Shigekawa T, et al. Optical imaging
a027094. 276. Zhu AX, Sahani DV, Duda DG, et al. Efficacy, safety, for monitoring tumor oxygenation response after
255. Jain RK. Normalizing tumor microenvironment to and potential biomarkers of sunitinib monotherapy initiation of single-agent bevacizumab followed by
treat cancer: bench to bedside to biomarkers. J Clin in advanced hepatocellular carcinoma: a phase II cytotoxic chemotherapy in breast cancer patients.
Oncol. 2013;31:2205–2218. study. J Clin Oncol. 2009;27:3027–3035. PLoS ONE. 2014;9:e98715.
256. Winkler F, Kozin SV, Tong RT, et al. Kinetics of 277. Zhu AX, Duda DG, Sahani DV, et al. HCC and 295. Quintela-Fandino M, Lluch A, Manso LM, et al.
vascular normalization by VEGFR2 blockade governs angiogenesis: possible targets and future directions. 18F-fluoromisonidazole PET and activity of neo-
brain tumor response to radiation: role of oxygen- Nat Rev Clin Oncol. 2011;8:292–301. adjuvant nintedanib in early HER2-negative breast
ation, angiopoietin-1, and matrix metalloproteinases. 278. Vasudev N, Goh V, Juttla J, et al. Changes in tumour cancer: a window-of-opportunity randomized trial.
Cancer Cell. 2004;6:553–563. vessel density upon treatment with anti-angiogenic American Association for Cancer Research:clincanres.
257. Leite de Oliveira R, Deschoemaeker S, Henze AT, agents: relationship with response and resistance to 2016;0738.2016.
et al. Gene-targeting of phd2 improves tumor therapy. Br J Cancer. 2013;109:1230–1242. 296. Ueda S, Saeki T, Takeuchi H, et al. In vivo imaging
response to chemotherapy and prevents side-toxicity. 279. Huang Y, Yuan J, Righi E, et al. Vascular normalizing of eribulin-induced reoxygenation in advanced breast
Cancer Cell. 2012;22:263–277. doses of antiangiogenic treatment reprogram the cancer patients: a comparison to bevacizumab. Br J
258. Mazzone M, Dettori D, Leite de Oliveira R, et al. immunosuppressive tumor microenvironment and Cancer. 2016.
Heterozygous deficiency of PHD2 restores tumor enhance immunotherapy. Proc Natl Acad Sci USA. 297. Stylianopoulos T, Jain RK. Combining two strate-
oxygenation and inhibits metastasis via endothelial 2012;109:17561–17566. gies to improve perfusion and drug delivery in
normalization. Cell. 2009;136:839–851. 280. Batchelor TT, Duda DG, di Tomaso E, et al. Phase solid tumors. Proc Natl Acad Sci USA. 2013;110:
259. Carmeliet P, Dor Y, Herbert JM, et al. Role of HIF- II study of cediranib, an oral pan-vascular endothelial 18632–18637.
1alpha in hypoxia-mediated apoptosis, cell prolifera- growth factor receptor tyrosine kinase inhibitor, in 298. Emblem KE, Gerstner ER, Sorensen AG, et al.
tion and tumour angiogenesis. Nature. 1998;394: patients with recurrent glioblastoma. J Clin Oncol. Matrix-depleting anti-hypertensives decompress
485–490. 2010;28:2817–2823. tumor blood vessels and improve perfusion in
260. Harris AL. Hypoxia—a key regulatory factor in 281. Frentzas S, Simoneau E, Bridgeman VL, et al. Vessel patients with glioblastomas receiving anti-angiogenic
tumour growth. Nat Rev Cancer. 2002;2:38–47. co-option mediates resistance to anti-angiogenic therapy. AACR Annual Meeting:abstr. 2016;3975.
261. Jain RK. Barriers to drug delivery in solid tumors. therapy in liver metastases. Nat Med. 2016. 299. Mpekris F, Baish JW, Stylianopoulos T, et al. Role of
Sci Am. 1994;271:58–65. 282. Chen Y, Huang Y, Reiberger T, et al. Differential vascular normalization in benefit from metronomic
262. Jain RK. The next frontier of molecular medicine: effects of sorafenib on liver versus tumor fibrosis chemotherapy: insights from a mathematical
delivery of therapeutics. Nat Med. 1998;4:655–657. mediated by stromal-derived factor 1 alpha/C-X-C model. Proc Natl Acad Sci USA. 2017;114:1994–
263. Thienpont B, Steinbacher J, Zhao H, et al. Tumour receptor type 4 axis and myeloid differentiation 1999.
hypoxia causes DNA hypermethylation by reducing antigen-positive myeloid cell infiltration in mice. 300. Sledge GW. Anti-vascular endothelial growth factor
TET activity. Nature. 2016. Hepatology. 2014;59:1435–1447. therapy in breast cancer: game over? J Clin Oncol.
264. McCormick F. New-age drug meets resistance. 283. Chen Y, Ramjiawan RR, Reiberger T, et al. CXCR4 2015;33:133–135.
Nature. 2001;412:281–282. inhibition in tumor microenvironment facilitates 301. Szabo V, Bugyik E, Dezso K, et al. Mechanism
265. Batchelor TT, Gerstner ER, Emblem KE, et al. anti-programmed death receptor-1 immunotherapy of tumour vascularization in experimental lung
Improved tumor oxygenation and survival in in sorafenib-treated hepatocellular carcinoma in mice. metastases. J Pathol. 2015;235:384–396.
glioblastoma patients who show increased blood Hepatology. 2015;61:1591–1602. 302. Matus DQ, Lohmer LL, Kelley LC, et al. Invasive
perfusion after cediranib and chemoradiation. Proc 284. Rahbari NN, Kedrin D, Incio J, et al. Anti-VEGF cell fate requires G1 cell-cycle arrest and histone
Natl Acad Sci USA. 2013;110:19059–19064. therapy induces ECM remodeling and mechani- deacetylase-mediated changes in gene expression.
266. Emblem KE, Mouridsen K, Bjornerud A, et al. cal barriers to therapy in colorectal cancer liver Dev Cell. 2015;35:162–174.
Vessel architectural imaging identifies cancer patient metastases. Sci Transl Med. 2016;8:360ra135. 303. Cheng G, Tse J, Jain RK, et al. Micro-environmental
responders to anti-angiogenic therapy. Nat Med. 285. Rhim AD, Oberstein PE, Thomas DH, et al. mechanical stress controls tumor spheroid size and
2013;19:1178–1183. Stromal elements act to restrain, rather than support, morphology by suppressing proliferation and induc-
267. Heist RS, Duda DG, Sahani DV, et al. Improved pancreatic ductal adenocarcinoma. Cancer Cell. ing apoptosis in cancer cells. PLoS ONE. 2009;4:
tumor vascularization after anti-VEGF therapy 2014;25:735–747. e4632.
with carboplatin and nab-paclitaxel associates with 286. Ozdemir BC, Pentcheva-Hoang T, Carstens JL, et al. 304. Desmaison A, Frongia C, Grenier K, et al. Mechani-
survival in lung cancer. Proc Natl Acad Sci USA. Depletion of carcinoma-associated fibroblasts and cal stress impairs mitosis progression in multi-cellular
2015;112:1547–1552. fibrosis induces immunosuppression and accelerates tumor spheroids. PLoS ONE. 2013;8:e80447.
268. Sorensen AG, Batchelor TT, Zhang WT, et al. pancreas cancer with reduced survival. Cancer Cell. 305. Delarue M, Montel F, Vignjevic D, et al. Com-
A “vascular normalization index” as potential 2014;25:719–734. pressive stress inhibits proliferation in tumor
mechanistic biomarker to predict survival after a 287. Cooke VG, LeBleu VS, Keskin D, et al. Pericyte spheroids through a volume limitation. Biophys J.
single dose of cediranib in recurrent glioblastoma depletion results in hypoxia-associated epithelial- 2014;107:1821–1828.
patients. Cancer Res. 2009;69:5296–5300. to-mesenchymal transition and metastasis mediated 306. Fischer KR, Durrans A, Lee S, et al. Epithelial-to-
269. Jain RK, Duda DG, Clark JW, et al. Lessons from by met signaling pathway. Cancer Cell. 2012;21: mesenchymal transition is not required for lung
phase III clinical trials on anti-VEGF therapy for 66–81. metastasis but contributes to chemoresistance.
cancer. Nat Clin Pract Oncol. 2006;3:24–40. 288. Chauhan VP, Martin JD, Liu H, et al. Angiotensin Nature. 2015;527:472–476.
270. Carmeliet P, Jain RK. Principles and mechanisms of inhibition enhances drug delivery and potentiates 307. Zheng X, Carstens JL, Kim J, et al. Epithelial-to-
vessel normalization for cancer and other angiogenic chemotherapy by decompressing tumour blood mesenchymal transition is dispensable for metastasis
diseases. Nat Rev Drug Discov. 2011;10:417–427. vessels. Nat Commun. 2013;4:2516. but induces chemoresistance in pancreatic cancer.
271. Batchelor TT, Duda DG, di Tomaso E, et al. Phase 289. Stylianopoulos T, Martin JD, Chauhan VP, et al. Nature. 2015;527:525–530.
II study of cediranib, an oral pan-vascular endothelial Causes, consequences, and remedies for growth- 308. Lambrechts D, Lenz HJ, de Haas S, et al. Markers of
growth factor receptor tyrosine kinase inhibitor, in induced solid stress in murine and human tumors. response for the antiangiogenic agent bevacizumab.
patients with recurrent glioblastoma. J Clin Oncol. Proc Natl Acad Sci USA. 2012;109:15101–15108. J Clin Oncol. 2013;31:1219–1230.
2010;28:2817–2823. 290. Goel S, Gupta N, Walcott BP, et al. Effects of 309. Ince WL, Jubb AM, Holden SN, et al. Association
272. Batchelor TT, Sorensen AG, di Tomaso E, et al. vascular-endothelial protein tyrosine phosphatase of k-ras, b-raf, and p53 status with the treatment
AZD2171, a pan-VEGF receptor tyrosine kinase inhibition on breast cancer vasculature and metastatic effect of bevacizumab. J Natl Cancer Inst. 2005;97:
inhibitor, normalizes tumor vasculature and alleviates progression. J Natl Cancer Inst. 2013;105:1188– 981–989.
edema in glioblastoma patients. Cancer Cell. 2007;11: 1201. 310. Jubb AM, Hurwitz HI, Bai W, et al. Impact of
83–95. 291. Maes H, Kuchnio A, Peric A, et al. Tumor vessel vascular endothelial growth factor-A expression,
273. Tolaney SM, Boucher Y, Duda DG, et al. Role of normalization by chloroquine independent of thrombospondin-2 expression, and microvessel
vascular density and normalization in response to autophagy. Cancer Cell. 2014;26:190–206. density on the treatment effect of bevacizumab
126.e6 Part I: Science and Clinical Oncology

in metastatic colorectal cancer. J Clin Oncol. pressure in tumors during progression: implications advanced pancreatic cancer receiving gemcitabine.
2006;24:217–227. for vascular collapse. Cancer Res. 2013;73:3833– Br J Cancer. 2010;103:1644–1648.
311. Duda DG, Willett CG, Ancukiewicz M, et al. Plasma 3841. 331. Nakai Y, Isayama H, Sasaki T, et al. The inhibition
soluble VEGFR-1 is a potential dual biomarker 321. Jain RK, Martin JD. Stylianopoulos T: The role of of renin-angiotensin system in advanced pancreatic
of response and toxicity for bevacizumab with mechanical forces in tumor growth and therapy. cancer: an exploratory analysis in 349 patients.
chemoradiation in locally advanced rectal cancer. Annu Rev Biomed Eng. 2014;16:321–346. J Cancer Res Clin Oncol. 2015;141:933–939.
Oncologist. 2010;15:577–583. 322. Nia HT, Liu H, Seano G, et al. Solid stress and elastic 332. Wilop S, von Hobe S, Crysandt M, et al. Impact
312. Gerstner ER, Emblem KE, Chi AS, et al. Effects of energy as measures of tumour mechanopathology. of angiotensin I converting enzyme inhibitors and
cediranib, a VEGF signaling inhibitor, in combina- Nature Biomedical Engineering. 2016;1:0004. angiotensin II type 1 receptor blockers on survival
tion with chemoradiation on tumor blood flow and 322a.  Stylianopoulos T, Munn LL, Jain RK. Reengineering in patients with advanced non-small-cell lung
survival in newly diagnosed glioblastoma. J Clin the physical microenvironment of tumors to improve cancer undergoing first-line platinum-based che-
Oncol. 2012;30(suppl; abstr 2009). drug delivery and efficacy: from math modeling to motherapy. J Cancer Res Clin Oncol. 2009;135:1429–
313. Meyerhardt JA, Ancukiewicz M, Abrams TA, bench to bedside. Trends Cancer. 2018;4:292–319. 1435.
et al. Phase I study of cetuximab, irinotecan, and 323. Chauhan VP, Boucher Y, Ferrone CR, et al. Compres- 333. Emblem KE, Gerstner ER, Sorensen G, et al.
vandetanib (ZD6474) as therapy for patients with sion of pancreatic tumor blood vessels by hyaluronan Matrix-depleting anti-hypertensives decompress
previously treated metastatic colorectal cancer. PLoS is caused by solid stress and not interstitial fluid tumor blood vessels and improve perfusion in
ONE. 2012;7:e38231. pressure. Cancer Cell. 2014;26:14–15. patients with glioblastomas receiving anti-angiogenic
314. Tolaney SM, Duda DG, Boucher Y, et al. A phase 324. Hagendoorn J, Tong R, Fukumura D, et al. Onset therapy. Cancer Res. 2016;76:3975.
II study of preoperative (preop) bevacizumab (bev) of abnormal blood and lymphatic vessel function 334. Regan DP, Guth AM, Hansen RJ, et al. Abstract
followed by dose-dense (dd) doxorubicin (A)/ and interstitial hypertension in early stages of A161: Angiotensin II type 1 receptor antagonism
cyclophosphamide (C)/paclitaxel (T) in combination carcinogenesis. Cancer Res. 2006;66:3360–3364. suppresses tumor metastasis through inhibition of
with bev in HER2-negative operable breast cancer 325. Lee JJ, Perera RM, Wang H, et al. Stromal CCL2-CCR2 mediated monocyte recruitment.
(BC). J Clin Oncol. 2012;30(suppl; abstr 1026). response to Hedgehog signaling restrains pancre- Cancer Immunol Res. 2016;4:A161.
315. Zhu AX, Ancukiewicz M, Supko JG, et al. Clinical, atic cancer progression. Proc Natl Acad Sci USA. 335. Whatcott CJ, Diep CH, Jiang P, et al. Desmoplasia in
pharmacodynamic (PD), and pharmacokinetic (PK) 2014;111:E3091–E3100. primary tumors and metastatic lesions of pancreatic
evaluation of cediranib in advanced hepatocellular 326. Whatcott CJ, Han H, Von Hoff DD. Orchestrating cancer. Clin Cancer Res. 2015;21:3561–3568.
carcinoma (HCC): A phase II study (CTEP 7147). the tumor microenvironment to improve survival 335a.  Murphy JE, Wo JYL, Ryan DP, et al. Potentially
J Clin Oncol. 2012;30(suppl; abstr 4112). for patients with pancreatic cancer: normalization, curative combination of TGF-b1 inhibitor losartan
316. Xu L, Duda DG, di Tomaso E, et al. Direct evi- not destruction. The Cancer Journal. 2015;21:299– and FOLFIRINOX (FFX) for locally advanced
dence that bevacizumab, an anti-VEGF antibody, 306. pancreatic cancer (LAPC): R0 resection rates and
up-regulates SDF1alpha, CXCR4, CXCL6, and 327. Sherman MH, Yu RT, Engle DD, et al. Vitamin D preliminary survival data from a prospective phase
neuropilin 1 in tumors from patients with rectal receptor-mediated stromal reprogramming suppresses II study. ASCO. 2018;Abstract #222259 (2018).
cancer. Cancer Res. 2009;69:7905–7910. pancreatitis and enhances pancreatic cancer therapy. 336. Benson A, Bendell J, Wainberg Z, et al. 618PDA
317. Duda DG, Kozin SV, Kirkpatrick ND, et al. Cell. 2014;159:80–93. phase 2 randomized, double-blind, placebo controlled
CXCL12 (SDF1alpha)-CXCR4/CXCR7 pathway 328. Menter AR, Carroll N, Delate T, et al: Effect of study of simtuzumab or placebo in combination with
inhibition: an emerging sensitizer for anticancer angiotensin system inhibitors on survival in patients gemcitabine for the first line treatment of pancreatic
therapies? Clin Cancer Res. 2011;17:2074–2080. receiving chemotherapy for advanced non-small adenocarcinoma. Ann Oncol. 2014;25:iv211.
318. Kamba T, Tam BY, Hashizume H, et al. VEGF- cell lung cancer, 50th Annual Meeting of ASCO. 337. Catenacci DV, Junttila MR, Karrison T, et al.
dependent plasticity of fenestrated capillaries in the Chicago, IL, 2014. Randomized Phase Ib/II study of gemcitabine
normal adult microvasculature. Am J Physiol Heart 329. Keizman D, Huang P, Eisenberger MA, et al. plus placebo or vismodegib, a hedgehog pathway
Circ Physiol. 2006;290:H560–H576. Angiotensin system inhibitors and outcome of inhibitor, in patients with metastatic pancreatic
319. Verheul HM, Pinedo HM. Possible molecular sunitinib treatment in patients with metastatic renal cancer. J Clin Oncol. 2015;33:4284–4292.
mechanisms involved in the toxicity of angiogenesis cell carcinoma: a retrospective examination. Eur J 338. Hingorani SR, Harris WP, Beck JT, et al. Phase
inhibition. Nat Rev Cancer. 2007;7:475–485. Cancer. 2011;47:1955–1961. Ib study of PEGylated recombinant human hyal-
320. Stylianopoulos T, Martin JD, Snuderl M, et al. 330. Nakai Y, Isayama H, Ijichi H, et al. Inhibition uronidase and gemcitabine in patients with advanced
Coevolution of solid stress and interstitial fluid of renin-angiotensin system affects prognosis of pancreatic cancer. Clin Cancer Res. 2016.
Tumor Microenvironment: Vascular and Extravascular Compartment  •  CHAPTER 8 126.e7
126.e7

SELF-ASSESSMENT QUESTIONS ANSWERS


1. Malignant tumors are 1. (c) Malignant tumors are organlike structures composed of
a. conglomerates of genetically mutated cancer cells. malignant cells and host-derived stromal cells (recruited from
b. conglomerates of genetically mutated cancer cells and surrounding tissue and from blood circulation) in variable
stromal cells recruited from blood circulation. proportions depending on tumor type, stage, and site.
c. organlike structures composed of extracellular matrices 2. (e) Solid components of tumors, including cancer cells, stromal
containing genetically mutated cancer cells, host-derived cells, and matrix molecules, lead to elevated mechanical solid
stromal cells recruited from surrounding tissues and from stress. These solid tissue forces can greatly exceed the
blood circulation and extracellular matrices. microvascular pressure, and thus solid components of tumors
2. Vascular compression in tumors limits perfusion and is caused compress blood vessels. Interstitial hypertension is caused by the
by leakiness of tumor blood vessels, as in physiological edema
a. interstitial hypertension associated with uid retained in during in inflammation. Thus interstitial fluid pressure (fluid
tumors. stress) in tumors cannot exceed the microvascular pressure and
b. solid stress produced from cancer and stromal cells as they these fluid pressures completely oppose each other, such that
proliferate. interstitial hypertension cannot lead to vascular compression.
c. transmission of solid stress by matrix molecules. 3. (b) sVEGFR1/sFLT1 is an endogenous blocker of VEGF and
d. a and b placental-derived growth factor and has been proposed as a
e. b and c predictive biomarker of intrinsic resistance to anti-VEGF
3. Plasma soluble VEGFR1 is an endogenous vascular endothelial therapy. Our group reported that patients with locally advanced
growth factor (VEGF) inhibitor and is currently explored in rectal carcinoma who had high plasma sVEGFR1 levels prior to
trials of antiangiogenic agents as a treatment were less likely to benefit—or experience toxicities—
a. pharmacodynamic biomarker. from bevacizumab therapy combined with chemoradiation. We
b. predictive biomarker. later found the same association for newly diagnosed patients
c. prognostic biomarker. with glioblastoma multiforme, triple-negative breast cancer,
d. surrogate biomarker. hepatocellular carcinoma, and metastatic colorectal carcinoma
4. Interstitial hypertension is a hallmark of solid tumors and who were treated with anti-VEGF agents.
results from 4. (d) The blood vasculature of malignant tumors is abnormal and
a. vessel leakiness. hyperpermeable, and the lymphatic (drainage) system is
b. lack of functional lymphatics. disrupted within tumors. Along with the pressure created by
c. compression of vessels. proliferating cancer cells, these factors result to an increase in
d. all of the above. interstitial fluid pressure from approximately 0 mm Hg
e. none of the above. (normal) to levels that can reach 94 mm Hg.
5. Low vascular perfusion in solid tumors leads to 5. (e) Reduced perfusion, caused by vascular compression and
a. hypoxia. interstitial hypertension, limits the supply of oxygen and drugs
b. poor response to chemotherapy. to tumors. This limitation promotes hypoxia and inadequate
c. reduced metastasis. drug delivery in tumors, both leading to drug resistance. Higher
d. all of the above. perfusion correlates with lengthened survival in patients, and
e. a and b. agents in preclinical development that improve perfusion lead to
enhanced therapeutic outcomes. Metastasis thus far is not
affected by agents that increase perfusion.
Cancer Metabolism 9 
Michael A. Reid, Sydney M. Sanderson, and Jason W. Locasale

S UMMARY OF K EY P OI NT S
• Metabolism supports biosynthesis, exploited clinically for diagnostics their environment, germline genetics,
energetics, and cell signaling and therapeutics. liver physiology, the microbiome,
including chromatin state and • Old and new therapies target cancer and other influences including diet
epigenetics. metabolism, and the repurposing of and exercise.
• A common feature of each element metabolic drugs for cancer therapy
of malignancy is the necessity to is attractive and ongoing.
adapt metabolic functions. • Future work in the area of cancer
• Altered metabolism in key metabolic metabolism will incorporate the
pathways and networks is being interaction between tumor cells and

Cancers are diseases in which cells acquire mutations that result in lead to this altered metabolism and the specific requirements that
proliferation that occurs outside the context of normal tissue develop- result. Particular attention will be paid to key metabolic pathways
ment. These somatic mutations interact with their environment to and networks and to areas that have direct clinical application such
result in cell transformation. This environment is determined by as current antimetabolite chemotherapy agents and ongoing drug
numerous factors including, but not exclusively, host genetics, lifestyle development efforts. Our hope is to give the reader a broad overview
including diet, host physiology, the state of the immune system, and of this rapidly progressing area of cancer biology and medical
the microenvironment of the tumor cells. At the nexus of this interaction oncology.
between genes and environment is cellular metabolism.
Metabolism involves a set of interconnected chemical reactions
mediated by enzymes. At the cellular level, nutrients including glucose, FUNDAMENTAL SCIENCE
lipids, and amino acids that are derived from their constituent dietary Warburg Effect
macronutrients—carbohydrates, fats, and proteins—are taken up and
then transformed through a series of interconnected chemical reactions In conditions with sufficient oxygen, terminally differentiated cells
that comprise the metabolic network (Fig. 9.1). These nutrients are usually metabolize glucose through three highly regulated sequences
stored in their electron-rich reduced forms and through a process of enzymatic processes: glycolysis, the tricarboxylic acid (TCA) cycle,
known as catabolism are oxidized. The release of the electrons from and the electron transport chain (ETC).1,2 On complete oxidation,
these oxidative processes generates energy that is used to sustain all one molecule of glucose can ultimately generate approximately 36
elements of life. Metabolism also allows for the conversion of intermedi- units of the energy-storing molecule adenosine 5′-triphosphate (ATP)
ate products of catabolism to materials used as structural components and other energy-rich metabolic currency such as reduced nicotinamide
or building blocks for the production of proteins, nucleic acids, and adenine dinucleotide (NADH).1,2 These processes are compartmental-
cell membranes. This is termed anabolism or anabolic metabolism. ized: glycolysis in the cytosol, and the TCA cycle and ETC in the
Metabolism also can communicate directly to other elements of cellular mitochondria. Under conditions of oxygen limitation such as exercise
machinery by generating molecules that directly react with and alter in muscle, the end product of glycolysis (pyruvate) is diverted away
the activity of proteins and nucleic acids. from the mitochondria to produce the fermentation product lactate.2
A common feature of each element of malignancy is the necessity This is known as anaerobic glycolysis and generates only two net ATP
to adapt each of these metabolic functions to support the demands molecules per molecule of glucose while consuming NADH (produces
of uncontrolled proliferation. Indeed, differential metabolic require- oxidized nicotinamide adenine dinucleotide [NAD+]), and is a common
ments manifest during each cancer-associated process such as anoikis, form of glucose metabolism for unicellular organisms such as certain
invasion, and metastasis. This metabolic programming comes from yeasts and parasites.3,4 Although this may seem less efficient for mam-
both the oncogenic signaling that drives distinct metabolic processes malian cells, the rate of ATP production via glycolysis can be much
and the environmental factors such as the tissue vasculature and the faster than the time it takes for mitochondrial respiration; therefore
nutrient content of serum. Furthermore, metabolic enzymes, by virtue in some cases greater amounts of ATP can be generated from glycolysis
of their design and particularly their binding pocket, which encapsulates in a given amount of time in the presence of abundant glucose.5,6
small molecules, are readily druggable. In addition, metabolism may One of the first documented points of evidence for an altered
be manipulated by lifestyle factors such as diet and exercise. Thus molecular property in tumors was the seminal observation made by
there is tremendous opportunity to exploit metabolism for cancer Otto Warburg and colleagues in 1924 regarding the tumor cell–specific
prevention and therapy. fate of glucose (Fig. 9.2). The essential features of what came to be
This chapter discusses the fundamentals of metabolic reprogramming defined as the “Warburg effect” were the observations that neoplastic
and adaptation in cancer cells and outlines both the principles that cells consumed large amounts of glucose and fermented it into lactate.7

127
128 Part I: Science and Clinical Oncology

and fibroblasts use aerobic glycolysis for normal function.12,13 Nonethe-


less, the Warburg effect in tumors is generally broad and has been
exploited clinically for many years through imaging of glucose uptake
Dietary sources Roles (discussed later in further detail) for diagnosis and monitoring of
–Carbohydrates 1 Energy treatment progress in numerous cancers including lymphomas, breast
–Fats – Oxidation cancer, non–small cell lung cancer (NSCLC), esophageal cancer, and
–Protein 2 Biosynthesis colorectal cancer.14
– Reduction Several hypotheses as to why cancer cells favor what seems like a
wasteful process have been proposed.15 Warburg and colleagues eventu-
3 Signalling
Digestion ally concluded it was due to defective mitochondria,16 but many cancer
cells in fact use diverse mitochondrial metabolic functions for growth
advantages, which are discussed in detail later. One proposal for why
cancer cells undergo the Warburg effect is the need for rapid production
Macronutrients of ATP, and as mentioned previously this can be achieved by enhanced
–Glucose glycolysis. In support of this, it has been demonstrated that cells
–Lipids upregulate aerobic glycolysis when challenged with conditions that
–Amino acids require generation of large amounts of ATP while oxidative phosphoryla-
tion remains constant.17 However, mathematical calculations indicate
that the amount of ATP required for cell growth and division may
Cellular be less than the amount required for normal cell survival, suggesting
uptake
that ATP may not be a limiting factor in cancer cell proliferation.18
Another model hypothesizes that the Warburg effect is important for
Central carbon generating biomass in the form of donating carbons for biosynthetic
metabolism processes. For example, the enzyme phosphoglycerate dehydrogenase
(PHGDH) diverts glucose carbons away from glycolysis into the serine
biosynthetic pathway,19 which is important for one-carbon metabolism
Figure 9.1  •  Overview of macronutrient metabolism. Macronutrients, and nucleotide synthesis (discussed later), but it remains unclear if
carbohydrates, fats, and proteins are digested into constituent macronutrients or how this would promote biomass production because the majority
glucose, lipids, and amino acids. The cellar uptake of these macronutrients serves of glucose carbons are excreted as lactate during the Warburg effect.20
three main roles: energy production, biosynthesis, and cellular signaling. Energy
production is generally oxidative, and biosynthesis is generally reductive. An intriguing hypothesis suggests that the acidic microenvironment
created by lactate secretion promotes tumorigenesis. This is supported
by studies showing that increased acidity promotes cancer cell invasive-
ness,21,22 but the Warburg effect occurs at an early time point during
18FDG-PET Glucose tumorigenesis before invasion, which calls into question whether this
can be the main function. Another model reasons that cancer cells
undergo aerobic glycolysis for the purpose of cellular signaling such
as the opening of chromatin, although it is difficult to conduct experi-
ments that conclusively demonstrate this.15
Although the exact reason why cells favor the Warburg effect remains
elusive, there has been considerable effort toward exploiting it for
± Oxygen cancer treatment by generating new therapeutic targets. It is clearly
required for certain tumors, and furthermore, glucose uptake and
catabolism are driven by enzymes encoded by oncogenes commonly
mutated or overexpressed in many tumor types such as PI3K, KRas,
and HIF1α, suggesting that tumors select for aerobic glycolysis.23
Future work will focus on elucidating exactly how the Warburg effect
promotes tumorigenesis and better defining the specificity of targetable
Liver metastasis nodes for new cancer therapeutic strategies.
(Source: Wikipedia)

Amino Acid Metabolism


Lactate
In addition to glucose, cancer cells rely on other nutrients to meet
increased metabolic demands. Several amino acids have been shown
Figure 9.2  •  The Warburg effect. In the presence or absence of oxygen, to play important roles for various cancer cell functions. Similar to
cancer cells take in large amounts of glucose and preferentially ferment the glucose, amino acid uptake through cell membrane transport channels
final product of glycolysis, pyruvate, into lactate. Fluorine-18 fluorodeoxy- and catabolism can be driven by oncogenes.23–26 Cancer cells can also
glucose–positron emission tomography (18F-FDG–PET) is commonly used obtain amino acids via de novo synthesis, breakdown of the serum
as an imaging modality to monitor tumors. peptide albumin after micropinocytosis,27 and autophagic digestion
of cellular proteins.28,29 Of the 20 common amino acids, nine are
essential—meaning that they are obtained from dietary sources.
It was also found that this occurred even in the presence of abundant However, certain nonessential amino acids are considered to be
oxygen; thus this phenomenon is commonly referred to as aerobic conditionally essential in cases in which rapid catabolism outcompetes
glycolysis, and Warburg hypothesized that killing cancer cells would the local rate of generation.30
require depleting both glucose and oxygen.8 These initial findings There have been substantial efforts in studying cancer cell gluta-
have been conclusively validated by genetic and pharmacologic mine metabolism as both a carbon and a nitrogen source. Glutamine
studies,9,10 but not all cancers undergo or require aerobic glycolysis.11 levels in serum are the highest of all amino acids,31 and glutamine is
Moreover, nontransformed proliferative cell types such as lymphocytes important for several cellular functions. Glutamine maintains redox
Cancer Metabolism  •  CHAPTER 9 129

balance by contributing to glutathione (GSH) synthesis and participates


in anabolic metabolism such as lipid biosynthesis through reductive Glucose Lactate
carboxylation.32,33 Glutamine also maintains nitrogen pools for the
synthesis of nonessential amino acids and nucleotides.34 Of particular Pyruvate
importance is that glutamine-derived glutamate is converted into
α-ketoglutarate (αKG), which can maintain the flow of the TCA Pyruvate
cycle leading to ATP production via the ETC.35 This is known as TCA
Acetyl-coA cycle
anaplerosis and is critical for continuing mitochondrial respiration.
There is strong evidence that this occurs in cell culture, but reports Energy
using stable isotope labeling in mice have indicated that glutamine may Lipid (Oxidative
not be a major source of αKG in all tumors.36 However, numerous Amino acids phosphorylation)
studies have demonstrated the importance of glutamine in certain TCA Biosynthesis
tumors; inhibition of glutamine metabolism prevents tumor growth cycle (Nucleic acids,
in models of breast cancer,37 liver cancer,38 kidney cancer,39 and T-cell amino acids)
acute lymphoblastic leukemia.40
Signalling
Another fundamental metabolic process is one-carbon metabolism, (e.g. Reactive oxygen
which involves the amino acids serine, glycine, and methionine. Epigenetics
(Histone/DNA species, HIFs)
One-carbon metabolism that consists of the folate and methionine methylation)
cycles is a master integrator of nutritional status. Conversion of serine
to glycine via serine hydroxymethyltransferase (SHMT) generates
one-carbon molecules that are passed to folate moieties.41 These folate Figure 9.3  •  Mitochondrial metabolism. Mitochondria serve at the center
moieties undergo further chemical reactions that lead to the transfer of macronutrient metabolism. Glucose, lipids, and amino acids enter the
mitochondria and are oxidized through the tricarboxylic acid (TCA) cycle.
of carbon atoms to downstream metabolic pathways. A major output The TCA cycle provides several functions, noted on the right. HIF, Hypoxia-
of the folate cycle is the generation of nucleotides, which are essential inducible factor.
for proliferating cells. Serine can be taken up by cells through transport-
ers or synthesized de novo from glucose or gluconeogenic intermedi-
ates.42 In addition to contributing to the generation of nucleotides,
serine coupled to the folate cycle is necessary for other biomass such
as phospholipids and the generation of redox factors such as NADPH, generation of reactive oxygen species (ROS). This led to the proposal
a molecule used for reductive biosynthesis and restoration of gluta- that mitochondria may promote DNA damage via high amounts of
thionine, which is used to protect against oxidative stress.43 Serine ROS accumulation, which could contribute to genome instability.56
further contributes to redox balance by ligating with homocysteine Although potentially damaging to cells at high levels, at lower concentra-
to begin the transsulfuration pathway for cysteine synthesis, which is tions ROS have been shown to act as signaling molecules that can
required for GSH production.44 Highlighting the importance of serine promote proliferation and may be essential for tumorigenicity.57 In
biosynthesis for certain cancers was the discovery that PHGDH, the all, mitochondria promote cell growth through generation of energy,
gene encoding the enzyme that catalyzes the committed step, is amplified biomass, and signaling cascades (Fig. 9.3).
in melanoma and breast cancer.19 Moreover, PHGDH inhibitors have Warburg and others originally hypothesized that tumor cells
shown promise in reducing tumor growth in mouse models.45–47 Also harbored dysfunctional mitochondria, which is how it was reasoned
of note, one-carbon units are used to methylate homocysteine to that the TCA cycle and ETC were not used for tumor cell glucose
generate methionine, which can have a major effect on protein regula- metabolism.16 However, subsequent studies found that tumor cells
tion and epigenetics (discussed in more detail later).48 In many cases, could undergo oxidative metabolism, and in fact the mitochondria
the majority of one-carbon units used to recycle methionine come play a critical role in cancer cell metabolism. For example, it was
from serine.49 In all, carbon units derived from serine, glycine, and reported in the 1950s that oxidation of the fatty acid (FA) palmitate
other one-carbon donors such as choline, betaine, sarcosine, and occurred in a manner similar to that in healthy liver cells.58 In addition,
histidine are used to produce biosynthetic components, maintain redox enzymes of the TCA cycle such as isocitrate dehydrogenase (IDH)
status, and provide the substrates for methylation reactions.43,44 have similar expression levels in cancer cell and normal cell mitochon-
Other amino acids important for tumorigenesis include arginine, dria.59 Numerous studies have corroborated these findings, and several
tryptophan, and the branched-chain amino acids leucine, isoleucine, reports have provided genetic evidence that mitochondrial metabolism
and valine. Arginine can act as nitrogen donor for nitric oxide (NO) is necessary for tumorigenesis.57,60 Indeed, the current paradigm in
signaling, as a fuel for the urea cycle, or to maintain pools of nonessential the cancer metabolism field is that cancer cells actively use and require
amino acids, and tumors lacking the argininosuccinate synthase enzyme both glycolysis and mitochondrial metabolism.61
highly depend on exogenous arginine for growth.50 Tryptophan There is developing interest in targeting tumor mitochondrial
catabolism by tumors has been shown to create an immunosuppressive processes for cancer therapy.61 As mentioned, the Warburg effect
tumor microenvironment.51,52 Branched-chain amino acids contribute can generate sufficient amounts of ATP to fuel cancer cells in the
to biosynthesis of alanine and glutamine, and high levels are found absence of mitochondrial metabolism. Thus targeting mitochondrial
in the serum of patients with early-stage pancreatic cancer.53 In addition, ATP production may not be a viable therapeutic strategy. However,
leucine and arginine are potent activators of mammalian target of certain subtypes of cancer have been shown to be highly reliant on
rapamycin (mTOR), which is a regulator of protein translation and mitochondrial-dependent ATP production via oxidative phosphoryla-
metabolism and cell growth and often dysregulated in cancers.54 tion.62,63 Moreover, the tumor microenvironment may have limited
glucose availability, which would reduce the glycolytic capacity of
Mitochondrial Metabolism the cell and make it more dependent on mitochondrial metabolism
(discussed later in detail). A major pitfall for targeting these processes
Mitochondria are organelles that play an important role in cell survival is the fact that a majority of normal cells require functional mito-
and proliferation.1,2 Mitochondrial metabolism is important for the chondrial metabolism for survival. Thus tumor-specific delivery and
function of the TCA cycle; ETC, fatty acid oxidation (FAO); synthesis uptake of therapeutics or the targeting of biosynthetic precursors
of amino acids, lipids, and nucleotides; and production of ATP and selectively required in cancer cells would be essential for reducing
antioxidants.55 One consequence of mitochondrial metabolism is the healthy cell toxicity. Potential candidates include the antidiabetes
130 Part I: Science and Clinical Oncology

drug metformin and certain glutaminase inhibitors (discussed later been observed in preclinical models.81 Other enzymes of targetable
in detail). interest include CPT1, ACLY, and ACC, with results similar to those
with FASN inhibition.82 An interesting strategy for using cancer cell
Lipid Metabolism lipid dependence against tumors is loading drugs into liposomal
nanoparticles, which has been shown to increase tumor-specific drug
Although given less attention than glucose or amino acids, lipid delivery.83 As is the case with targeting most metabolic processes, the
metabolism provides important signaling intermediates and biomass inherent challenge is overcoming toxic side effects to normal cells.
and energy resources to cancer cells. There are numerous species of
lipids, or FAs, and all contain a hydrocarbon chain and terminal Metabolism and Epigenetics
carboxyl group.1,2 The synthesis and oxidation of FAs contribute to
numerous cellular growth and survival functions such as membrane Epigenetics, or more specifically chromatin state, encompasses changes
formation, signaling molecules, and energy sources. Similar to other in gene expression that are not the result of mutations in DNA
nutrients that support proliferation, lipid metabolism is upregulated sequences.84 Chromatin accessibility can in part determine gene
in cancer cells.64 This is largely because cells must double their lipid expression and is regulated by posttranslational modifications of the
membrane in order to divide, resulting in a massive demand for chromatin-bound histone proteins including methylation, acetylation,
lipid and phospholipid precursors. Building blocks for membrane ubiquitination, and phosphorylation in addition to methylation of
production and lipid energy sources come from exogenous sources the DNA base cytosine.85 Changes in epigenetics and chromatin have
such as diet and/or are synthesized de novo by cells. Cancer cells emerged as drivers of cancer initiation and progression86,87 and can
preferentially synthesize lipids locally, and early studies revealed a shift function similarly to the amplification of oncogenes or the deletion
toward FA synthesis in neoplastic cells.65 of tumor suppressor genes.88,89 From a therapeutic perspective, targeting
De novo synthesis of FAs is coupled to glucose and glutamine epigenetics is appealing for several reasons, including the druggability
metabolism. The TCA cycle intermediate citrate is the starting material of the enzymes that carry out modifications to chromatin and DNA
for a majority of local FA production.66 As mentioned, citrate can be and the fact that many of the effects are reversible, as opposed to the
generated by glucose-derived pyruvate or, in high–Warburg effect irreversible changes of genetic mutations.
cells, glutamine-derived αKG. Several enzymes including ATP citrate Histone acetylation and DNA and histone methylation are among
lyase (ACLY), acetyl-coenzyme A (CoA) carboxylase (ACC), and fatty the best studied modifications. DNA methylation occurs at the fifth
acid synthase (FASN) are required to metabolize citrate into bioactive position of cytosine bases and is commonly observed in promoter
FAs.67 ACLY converts citrate to oxaloacetate and acetyl-CoA, and regions of genes.86 Histone acetylation and methylation occur on
acetyl-CoA is the major precursor for all lipids in the cell. The lysine (K) residues. Acetylation generally leads to active chromatin,
importance of ACLY for tumor growth is demonstrated by studies whereas histone methylation is associated with both active and inactive
showing that loss of ACLY reduced tumor growth in mouse models.68–70 chromatin.90 For example, methylation of histone H3K4 leads to
ACC catalyzes the committed step in FA synthesis via carboxylation enhanced gene expression, whereas methylation of H3K9 or H3K27
of acetyl-CoA to malonyl-CoA. ACC is a highly regulated enzyme, is associated with gene suppression. Underlying many epigenetic states
and its activity is negatively regulated by the tumor suppressor LKB1 is cellular metabolism because certain metabolites provide substrates
via AMPK-dependent phosphorylation.71 ACC has been shown to be for posttranslational modifications and essential cofactors for epigenetic
required for cancer cell growth and survival,72–74 but additional complex- machinery.91,92
ity comes from other studies that have indicated a tumor suppressive Methionine-derived S-adenosyl methionine (SAM) is a methyl
role for ACC.75 FASN is commonly upregulated in several cancer donor for several methyl transferase enzymes, including DNA and
types, and its expression correlates with poor prognosis.76 FASN histone methyl transferases (Fig. 9.4A). Therefore changes in one-carbon
condensates one acetyl-CoA and seven malonyl-CoA molecules to metabolism can have dramatic effects on epigenetics.93 Indeed, dietary
generate the initial product of FA synthesis, palmitate, which is then restriction of methionine reduces levels of H3K4 methylation.48
chemically altered to fuel pools of a diverse array of lipids including Moreover, limiting glycine input reduces SAM levels and has a similar
those involved in membrane formation and signaling molecules.76 effect on H3K4 methylation.94 Similarly, SAM levels also affect DNA
In addition to relying on lipids for biomass, cancer cells can oxidize methylation during osteoclast differentiation.95 Acetyl-CoA derived
FAs for energy production. The synthesis and oxidation of FAs are from glycolysis and β-oxidation is the primary acetyl donor for histone
mutually exclusive, and several mechanisms for this regulation have acetylation reactions carried out by histone acetyltransferases (Fig.
been described in cancer cells. In a process known as β-oxidation, 9.4B). Previous studies have demonstrated that acetyl-CoA availability
cellular FAs such as palmitate are catabolized in peroxisomes and largely regulates histone acetylation,96 and histone acetylation depends
mitochondria to generate energy and reducing molecules. Carnitine on ACLY activity.97 Histone deacetylases remove acetyl groups from
palmitoyltransferase 1 (CPT1) catalyzes the rate-limiting step for histones, and some require NAD+ as a cofactor.98 Therefore the NAD+/
importing FAs into the mitochondria, where it is catabolized to generate NADH ratio in addition to the levels of acetyl-CoA, and NAD+,
acetyl-CoA. The 16-carbon palmitate ultimately generates eight which can be increased in high Warburg cells, may have an effect on
acetyl-CoA molecules, which condense with oxaloacetate to drive the epigenetics and gene expression.
TCA cycle.77 Thus one molecule of palmitate can produce more energy A major breakthrough in understanding of the connection between
than fully oxidized glucose. The brain isoform of CPT1 (CPT1C) cancer metabolism and epigenetics was the discovery of mutations
was identified as a potential oncogene for its role in promoting cancer in the IDH genes IDH1 and IDH2 in leukemias and gliomas.99,100
cell survival under metabolic stress.78 β-Oxidation has been shown to The enzymes IDH1 and IDH2 normally convert citrate to αKG
be particularly important for cancer cells that have lost attachment in the mitochondria and cytosol, respectively. Mutations in IDH1
to the basal membrane,79 and targeting β-oxidation was sufficient to and IHD2 occur in the active site, and loss of αKG production by
reduce tumor growth in a patient-derived triple-negative breast cancer IDH-mutant enzymes was originally attributed to loss-of-function.101
(TNBC) xenograft model.80 Subsequent studies found the IDH mutants to be gain-of-function
Because cancer cells may selectively rely on de novo FA synthesis, mutations, where αKG was converted to 2-hydroxyglutarate (2HG)
many efforts have been made toward targeting these pathways. Owing at high levels, resulting in a neomorphic enzymatic activity.102 Because
to its upregulation in numerous cancers, FASN in particular has been αKG is an essential cofactor and 2HG is an inhibitor of many DNA
a popular target for drug development efforts, with several inhibitors and histone demethylases, IDH-mutant cells show broad DNA and
evaluated in preclinical studies showing strong antitumor efficacy.76 histone hypermethylation, and supplementation with αKG attenuates
Unfortunately, harsh side effects such as detrimental weight loss have these changes.103,104 Of note, pharmaceutical targeting of IDH1 and
Cancer Metabolism  •  CHAPTER 9 131

Methionine Glutamine Glucose

Succinate
SAM Fumarate
2HG
αKG

H/D MT H/D DM Mutant


Methyl IDH1/2
K K Methyl K K K K
Methyl

C C C

Figure 9.4  •  Metabolism and epigenetics. (A) Methionine- and S-adenosyl methionine (SAM)–derived methyl moieties are attached to lysine (K) residues
of DNA binding histone proteins and/or cytosine (C) DNA bases by histone or DNA methyltransferases (H/D MT). Cytosine methylation generally attenuates
transcription, whereas histone methylation may activate or suppress transcription. Removal of methyl groups is carried out by histone or DNA demethylase
(H/D DM), and some require glutamine and/or glucose-derived α-ketoglutarate (αKG) as a cofactor. H/D DM can be inhibited by loss of αKG, by accumulation
of succinate and fumarate, or by mutant IDH–produced 2-hydroxyglutarate (2HG). (B) Acetyl-CoA generated from glucose and/or fatty acids such as palmitate
is attached to histone lysine residues by histone acetyltransferase (HAT) enzymes to enhance transcription. HATs require oxidized nicotinamide adenine
dinucleotide (NAD+) as a cofactor.

IDH2 has shown promising results in leukemia and glioma models.105,106 For example, the hypoxia-inducible factor (HIF) proteins are activated
Similarly, the loss of glutamine, which can produce αKG, results in by hypoxia and promote downstream transcriptional events to adapt
increased histone methylation of stem cells107 and alters drug responses to limited oxygen availability.114,115 This promotes survival in response
in tumors; targeting the methyltransferases or supplementing with to therapeutically induced stress such as radiation, and targeting HIF
αKG resensitized cancer cells to the drug.108 Other metabolites besides in combination with radiation therapy has shown promise.116 HIF
mutant IDH–derived 2HG can inhibit demethylase activity. Cancer proteins have also been implicated in maintaining cancer initiating
cells with loss-of-function mutations in the TCA cycle enzymes suc- or cancer stem cells that can repopulate tumors, and these cells are
cinate dehydrogenase and fumarate hydratase accumulate succinate commonly found in hypoxic niches.117,118
and fumarate, respectively, and display global DNA and histone In addition to oxygen, tumor cells often encounter low levels of
hypermethylation109 (Fig. 9.4A). Thus future cancer treatments may nutrients in the microenvironment (see Fig. 9.5). A study conducted
target epigenetic machinery or use dietary supplements in combination with paired pancreatic cancer biopsies found pancreatic tumors are
with establish chemotherapeutics and new agents that target cancer generally nutrient poor compared with adjacent normal tissues.119 It
metabolism. was shown that many amino acids were depleted, including glutamine
and serine. Studies have found that cancer cells can survive glutamine
Nutrient Heterogeneity and Tumor limitation through induction of cell cycle arrest or alteration in glucose
Microenvironment usage.120–122 Similarly, cells adapt to serine deprivation by inhibiting
cell division.123 In addition, a subtype of breast and melanoma cells
Many studies conducted in cell culture are performed on single cell with amplifications of PHGDH can compensate for the loss of
types in the presence of abundant nutrients. In physiologic environ- exogenous serine by upregulating de novo serine biosynthesis from
ments, tumor cells are surrounded by numerous other cell types glucose.19 Yet glucose availability itself is reduced in the microenviron-
including fibroblasts, endothelial cells, and lymphocytes, resulting in ment of some tumors.119 Further research will undoubtedly identify
competition for local nutrients110 (Fig. 9.5). In addition, tumors often other survival pathways and redundancies used by tumors to survive.
contain incomplete and unorganized vasculature, which can lead to One goal of understanding these mechanisms is to target them in
regions of nutrient and oxygen limitation.111 Although tumor cells combination with established therapeutics.
require an abundance of nutrients to maintain rapid growth and The tumor microenvironment can play a major role in metabolism of
proliferation, they paradoxically persist in these harsh environments. tumors, depending on the tissue of origin. Highlighting this is a study
Consequently, cells that adapted to low nutrients and oxygen are that showed metabolism within the same tumor types was drastically
among the most difficult to kill and present major challenges for different depending on the surrounding tissue.36 Heterogeneity of
many current cancer therapies.108 Thus in order to develop better nutrient availability and use can occur within a single tumor. For
therapeutic strategies, it is important to understand the tumor example, nutrient availability drastically differs in core regions of solid
microenvironment and mechanisms of cellular adaptation to nutrient- tumors compared with the peripheral regions.108 Furthermore, a study
deficient environments. of a non–small cell lung carcinoma patient cohort in which stable-
Hypoxia, or low oxygen, is one of the most studied properties of isotope labeling was used found evidence of Warburg and non-Warburg
the tumor microenvironment. Oxygen levels in healthy tissue are utilizing cells within a single tumor, and these metabolic phenotypes
around 5%, whereas regions of solid tumors can have oxygen levels largely differed from patient to patient.124 These results indicate that
below 0.5%.112 The ETC can function in oxygen levels as low as multiple types of therapeutics targeting metabolism may be necessary
0.5%,113 allowing tumor cells to mostly undergo normal cellular for treatment of a single tumor.
functions, but they may gain survival advantages based on signaling Tumor cell metabolism can alter the microenvironment to promote
changes in response to oxygen stress due to changes in redox balance. tumorigenesis and reduce immune activity. As mentioned previously,
132 Part I: Science and Clinical Oncology

Tumor
Epithelial
cells

Amino
acids Tumor cells

Glucose

Fibroblasts
Suppresive
macrophage

Lactate
Blood
FAs Macrophage vessel

Blood
vessel
Epithelial
cells Active Inactive
T-cells T-cells

Nutrient and oxygen availability Nutrient and oxygen availability


Figure 9.5  •  Nutrient heterogeneity and tumor microenvironment. Tumors contain numerous cell types including tumor cells, fibroblasts, epithelial cells,
macrophages, and T cells, which all compete for local nutrients. Tumor cell metabolism such as enhanced lactate secretion and glucose consumption can
suppress tumor-infiltrating immune cells. Cells furthest from the vasculature encounter nutrient- and oxygen-poor environments, which also alters their
characteristics and metabolism. FAs, Fatty acids.

highly glycolytic cells convert the majority of their glucose carbons Obesity and Cancer
to lactate and secrete those carbons out of the cells. Lactate secretion
is coupled to the cotransport of H+, and CO2 generated by the ETC Obesity is a health crisis in the developed world, with estimates of
perfuses out of the cell before conversion to H+ and HCO3− in the over 30% of adults in the United States classified as obese. This is
extracellular space.125 Consequently, the accumulation of H+ reduces thought to be a major if not the leading source of human mortality.137
the local pH, creating an acidic microenvironment around tumors. Although associated more commonly with type II diabetes and car-
Furthermore, studies have shown that acidified environments activate diovascular disease, accumulating evidence demonstrates that obese
matrix metalloproteases, which degrade the extracellular matrix and individuals are more likely to develop cancer. Obesity has been shown
promote tumor invasiveness.126,127 Moreover, lactate polarizes tumor- to increase risk of numerous cancers including breast, prostate,
associated macrophages into an immunosuppressive state.128 In addition pancreatic, and colon cancers,138 with some estimates concluding that
to innate immune cells, adaptive immune cells such as T cells require up to one in five cancers are directly attributable to obesity.139 Obesity
specific metabolic functions in order to facilitate their protective is characterized by excessive amounts of adipose tissue resulting in
effects. The ability of T cells to transition from a naïve (inactive) excessive nutrient storage in the form of lipids. Adipose tissue is
state to an effector (active) state to a memory state is determined by important for energy storage, cell signaling through hormone secretion,
their metabolism.12 Circulating T cells are normally in nutrient- and and inflammatory responses through cytokine secretion, all of which
oxygen-rich environments. Upon entering tumors, T cells are met are altered in the obese state.140 It has also been shown in ovarian
with challenging metabolic conditions that can affect their activity cancer that FAs produced by local adipose tissue fueled tumor cell
(see Fig. 9.5). T cells require large amounts of glucose for activation, metastasis.141 Thus it is likely that fat depots in themselves can promote
and undergo the Warburg effect to a similar degree as many cancer tumorigenesis.142
cells.12 The absence of abundant glucose can lead to T-cell inaction The major regulator of glucose homeostasis is the hormone insulin,
or death.129,130 Indeed, much of the immunosuppression found in which mediates insulin receptor (IR)–initiated cell signaling cascades.
the tumor microenvironment is attributed to the inability of T cells Insulin is released by pancreatic β-cells in response to higher blood
to obtain sufficient nutrients.131 In addition, tumor cells upregulate glucose levels and signals the uptake of glucose by peripheral tissues.
indolamine 2′3′-dioxygenase (IDO) to promote tryptophan catabolism, On activation, IR promotes the activation of signaling pathways
which induces cell death in effector T cells.132 In addition to nutrient including PI3K, AKT, and mTOR, which promote glucose uptake,
stress, oxygen availability also plays a major role in affecting T-cell anabolic metabolism, and cell growth.143,144 Because increased insulin
function. Hypoxia-induced HIF activation transcriptionally upregulates levels and insulin resistance are hallmarks of obesity and the PI3K/
PD-1 and CTLA4, which act to suppress immune activity.133,134 HIF also AKT/mTOR signaling axis is the most frequently altered signaling
promotes expression of T regulatory cells (Tregs), which downregulate cascade, it is possible that tumor cells may use insulin signaling resulting
the immune response.135 Because of this, T cells can be tumor suppres- from obesity-associated hyperinsulinemia to enhance their own glucose
sive or tumor promoting.136 Immunotherapy focusing on ex vivo and uptake and growth. In support of this are studies demonstrating
in vivo stimulation of T cells is a promising avenue for management of increased IR expression in certain liver, breast, and lung cancers.145–147
tumors, but overcoming the tumor microenvironment will be critical to Similar yet distinct hormones upregulated during obesity are insulin-like
its success. growth factors 1 and 2 (IGF1 and IGF2). IGF1 binds the insulin-like
Cancer Metabolism  •  CHAPTER 9 133

growth factor receptor (IGFR), leading to similar downstream signaling used chemotherapy agents. Methotrexate, a potent and well-characterized
cascades as insulin and IR. High levels of IGF1 have been shown to polyglutamatable antifolate compound, mimics folic acid but has an
increase the risk of breast cancer.148 On the other hand, IGF2 can additional methyl group at the N10 position; this allows the compound
bind both IR and IGFR149 and reportedly promotes tumor growth to have an exceptionally high affinity for DHFR, but the subtle
in models of breast cancer.150 Other secreted factors altered in response structural methyl modification antagonizes the enzyme’s ability to
to obesity that may contribute to tumorigenesis include adiponectin reduce DHF155,156 (Fig. 9.6). However, methotrexate can exhibit a
and leptin.151–153 Overall, obesity can lead to changes in circulating toxicity profile against normal proliferating tissues such as bone marrow
signaling molecules that directly enhance the tumorigenic capabilities and intestinal tissue157; to increase the therapeutic window, usually
of neoplastic tissues. The mechanisms, however, remain controversial high doses of methotrexate are administered followed by low doses
and complicated, with multifactorial effects occurring through signaling, of leucovorin (5-formyl-THF) to competitively inhibit intracellular
inflammation, and likely changes in tumor cell autonomous metabolism, transport of methotrexate in tissues with close proximity to blood
although this effect has not yet been well established. vessels (referred to as the “leucovorin rescue”).158,159 Methotrexate
remains a primary treatment option for lymphoma, osteosarcoma,
and leukemias.155 Other polyglutamatable antifolates that inhibit DHFR
CLINICAL RELEVANCE AND APPLICATIONS activity include pralatrexate and pemetrexed; the nonpolyglutamatable
antifolates that act on DHFR include trimetrexate, piritrexim, and
Antimetabolite Chemotherapy talotrexin.160 Pemetrexed remains the first-line treatment for NSCLC
as a dual treatment with cisplatin (a DNA cross-linking compound),161
Antifolates and remains under clinical investigation for the treatment of various
Folates, commonly supplemented in the diet in their more stable other cancers.162
oxidized form as folic acid, are B9 vitamins that are essential for major
biosynthetic reactions such as amino acid metabolism and cell prolifera- Nucleotide Synthesis Inhibitors
tion.44 For instance, 5-methyl-tetrahydrofolate (5-methyl-THF) is used Another class of antimetabolites includes compounds that directly
along with vitamin B12 to generate methionine, as was previously interfere with DNA or RNA synthesis. The original and most well-
discussed.154 In these folate-mediated reactions, dihydrofolate (DHF) known of these compounds is 5-fluorouracil (5-FU), a pyrimidine
is produced and is then reduced by dihydrofolate reductase (DHFR) analogue that bears structural similarity to uracil but with a fluoride
to regenerate THF cofactor pools. atom at the 5C position of its ring.163 The enzyme thymidylate synthase
There is a selective dependence on folates for cancer cell proliferation, (TS) converts uracil to thymine via a methylation reaction at the 5C
and a class of drugs known as antifolates is a group of commonly carbon position but is unable to execute this methylation of 5-FU

O BASE O O O

HO P O
Riboneucleotide O P O 5' Thymidylate H 3C
BASE
O
reductase OH O NH synthase NH
O- 4' 1'

3' 2' N O N O
OH OH H H
O OH
Riboneucleotide Uracil Thymine
Deoxyribonucleotide Cell membrane

NH2 F
N O
N O HN NH
HO
O F Nucleus
O 5-FU
Gemcitabine
OH F
NH2
Mitochondria
O COOH N N

NH2 N HO N O
H O
N
N N COOH
CH3 Cytosol OH
Decitabine
H 2N N N
Methotrexate
NH2 NH2
CH3
N N

N O Methyltransferase N O
Dihydrofolate Tetrahydrofolate H H
Dihydrofolate
reductase Cytosine 5-methyl cytosine

Figure 9.6  •  Antimetabolite therapies. Current antimetabolite therapies inhibit synthesis of essential compounds needed for efficient DNA, RNA, and
protein synthesis. Methotrexate differs from folic acid by a key methyl group (circled in red) and acts on the metabolic enzyme dihydrofolate reductase (DHFR)
to inhibit the production of tetrahydrofolate pools. Gemcitabine inhibits the activity of ribonucleotide reductase, and 5-fluorouracil (5-FU) inhibits the production
of thymine by thymidylate synthase because of the presence of a fluoride atom (circled in red); both of these inhibitory activities interfere with DNA synthesis
and elongation. Decitabine inhibits the methylation activity of DNA methyltransferases (DNMTs), thereby preventing DNA methylation and subsequent
gene repression.
134 Part I: Science and Clinical Oncology

owing to the presence of the fluoride atom, effectively inhibiting TS. compound, CB-839, has been shown to exhibit antiproliferative activity
5-FU remains a commonly used agent clinically; it is regularly used against TNBC cell lines and in xenograft murine models,37 and has
as a frontline agent for the treatment of colorectal cancer and is successfully moved through phase I clinical trials for both TNBC and
approved for other tissue types such as breast cancer. renal cell carcinoma.170 Of note, this small-molecule agent does not
Gemcitabine (2′,2′-difluorodeoxycytidine [dFdC]) is another popular act on the glutaminase isoform found in the liver, which is the main
pyrimidine analogue used as a chemotherapeutic agent. Gemcitabine isoform believed to be responsible for circulating glutamate,173 allowing
is a deoxycytidine analogue that has fluoride atoms in place of the for a better toxicity profile.
hydrogen atoms at the 2C position of its ring.164 Gemcitabine (both
alone and in combination with other agents such as cisplatin) has Fatty Acid Synthase Inhibitors
been approved for the treatment of numerous cancers including breast, FASN plays an essential role in lipogenesis by catalyzing the NADPH-
ovarian, and bladder cancers, NSCLC, and pancreatic cancer, and it dependent conversion of acetyl-CoA and malonyl-CoA to palmitic
is being investigated as an adjuvant to 5-FU therapy for metastatic acid, which acts as a precursor to long-chain FAs174 (Fig. 9.7). Cancer
colorectal cancer.165 cells have been shown to be dependent on FA synthesis for processes
Finally, the pyrimidine analogue decitabine (also referred to as including membrane biosynthesis and energy metabolism and typically
aza-dCyd) is converted by deoxycytidine kinase to aza-dCTP and is exhibit a high expression of FASN.175 A small-molecule inhibitor of
rapidly incorporated into DNA. This analogue prevents the epigenetic FASN, TVB-1366, was found to induce apoptosis in breast and prostate
modification of DNA methylation. Normally, cytosine residues of cancer cell lines, which was shown to be mediated by the disruption
DNA can be methylated to prevent transcription; aza-dCTP inhibits of membrane lipid distribution and localization of membrane-bound
DNA methyltransferases, thereby inducing enhanced gene expression.166 proteins.176 More recently, another small-molecule FASN inhibitor
Decitabine is approved for the treatment of myelodysplastic syndromes, (TVB-2640) has been investigated in phase I clinical trials for the
including acute myeloid leukemia (AML) in elderly cancer patients.167 treatment of KRAS-positive lung cancer, and as a combinatorial
treatment in breast cancer170; however, as previously mentioned, toxicity
Current Metabolic Drug Targets appears to be a limiting factor in its further development.
IDH1/2 Indolamine 2,3-Dioxygenase Inhibitors
As mentioned, IDH enzymes frequently exhibit heterozygous gain- IDO regulates metabolism of the amino acid tryptophan via the
of-function mutations in cancer, particularly in glioma (70%)100 and kynurenine pathway.177 Interesting to note, IDO was found to mediate
AML (15%–20%),168 resulting in an ability of the IDH enzymes to the ability of tumors to escape recognition by immune cells by sup-
further catalyze the conversion of αKG to 2HG.102,169 Two IDH pressing T cells, activating Treg cells, and promoting inflammatory
inhibitors, AG-221 and AG-120, have been clinically investigated for and angiogenic processes.178–180 IDO expression was also shown to be
the treatment of AML.170 The initial phase I and II clinical trials associated with resistance to paclitaxel in patients with serous ovarian
successfully demonstrated limited toxicity in addition to proof of cancer.181 Numerous IDO inhibitors are under preclinical investigation
concept, with nearly 40% of patients responding to treatment (Table as novel cancer treatments. 1-Methyl-dl-tryptophan (both the d and
9.1). Accelerated approval by the US Food and Drug Administration the l isomers) acts as a tryptophan mimetic and has been shown to
(FDA) of these compounds was completed in early 2017. effectively inhibit IDO activity in human dendritic cells.182 In addition,
D-1MT (but not L-1MT) was shown to induce T-cell activation and
Glutaminase Inhibitors to prolong survival in combination with paclitaxel in a murine model
Glutaminase is an enzyme that catalyzes the production of glutamate of breast cancer; this difference in efficacy was found to be due to
from the amino acid glutamine, which then feeds into the TCA cycle.171 D-1MT’s activity on the IDO2 isomer, which is expressed in only a
This enzyme was initially targeted with the small-molecule inhibitor subset of tissues that express IDO1.183 D-1MT (also known as
6-diazo-5-oxy-l-norleucine (DON).172 However, a small-molecule indoximod) is one of four IDO inhibitors under clinical investigation;
compound has been found to selectively inhibit glutaminase; this the others include INCB024360 (a hydroxyamidine small-molecule

Table 9.1  Metabolic Drugs Currently in Clinical Trials as of Late 2016


Company and Location Agent Target Indications Status
Agios and Celgene AG-221, AG-120, AG-881 IDH1 and IDH2 AML, MDS, solid tumors Phase III
Polaris Group, San Diego, CA ADI-PEG 20 Pegylated arginine deiminase HCC, others Phase III in HCC missed
primary endpoint
Cornerstone Pharmaceuticals, CPI-613 Pyruvate dehydrogenase AML, MDS, solid tumors Phase II
Cranbury, NJ
Calithera Biosciences CB-839 Glutaminase 1 RCC, breast cancer Phase I/II
3-V Biosciences TVB-2640 Fatty acid synthase Ovarian, breast, lung Phase II pending
Novartis, Basel, Switzerland IDH305 IDH1 Advanced cancers Phase I
Forma Therapeutics, Watertown, MA FT-2102 IDH1 AML, MDS Phase I
Bayer, Leverkusen, Germany BAY-1436032 IDH1 Solid tumors Phase I
Advanced Cancer Therapeutics, PFK-158 PFKFB3 Solid tumors Phase I
Louisville, KY
Aeglea BioTherapeutics, Austin, TX AEB-1102 Modified human arginase AML, MDS, solid tumors Phase I

Listed are examples of metabolic drug targets currently under clinical trial investigation, some of which are discussed in this text.
AML, Acute myeloid leukemia; HCC, hepatocellular carcinoma; IDH, isocitrate dehydrogenase; MDS, myelodysplastic syndrome; PFKFB3, 6-phosphofructo-2-kinase; RCC, renal
cell carcinoma.
Modified from Garber K. (2016) Nature Biotechnology, ref. 170. Pending adaptation approval.
Cancer Metabolism  •  CHAPTER 9 135

H+
H +
Gln Lactate Glucose H+ H+ H+ H2O Folic acid Ser Asn Arg Choline
H+ + +
HCO3 CO2
MCT1 MCT4 GLUT1/GLUT4 NEH1 CAs

Gln Glucose Na+ Folate/nucleotide Ser Asn Arg


GLS1 metabolism
HKs PPP
TKTL1 Folic acid Choline
Glu G6P F6P X5P R5P Gln
Glycolysis CK
Nicotinamide PFKFB3
F6P F2.6BP P-choline
NAMPT 5.10-CH2 THF
F1.6BP IMP UMP TYMS
Amino acid Sphingomyelin
NADH metabolism
GAPDH RNR THF DHF
Fatty acids PHGDH
3PG 3PHP SER dNTPs DHFR Isoprenoids
PGAM1
Asp Cholesterol
Fatty acyl-CoA 2PG Lipidogenesis
HMGCR
OAA Acetyl-CoA HMG-CoA Mevalonate
CPT1 PEP
ACLY
PKM2 OAA ACC
Fatty acyl- LDHA
carnitine Lactate Pyruvate Malate Citrate Malonyl-CoA FASN Fatty acids Glycerol
MGLL

MAG
NADH
Cl H+
OAA PC Pyruvate Malate H+ H + H+
Cytosol
PDK1 NAD+
Fatty acyl- Q10
carnitine
Acetyl-CoA
H+

Pol
OAA Citrate H+ H+
OXPHOS H+ Nucleus
β-oxidation
Malate Cyt c
D-isocitrate H+
Krebs cycle H+ H+
IDH ADP H+
ADP ADP
α-KG Glu
H+ H + H+
H+
GLUD1 ATP F1FO
ATP ATPase
ATP
Mitochondrial
matrix
Intermembrane space

Figure 9.7  •  Current metabolic targets in cancer therapy. Current drug targets in cancer therapy include numerous metabolic targets involved in multiple
mitochondrial and cytosolic energetic pathways. Targets labeled in red are under preclinical investigation, targets labeled in blue are in early stages of clinical
trials, and targets labeled in green are in advanced stages of clinical trial investigation. This diagram depicts targets described in this text and additional targets
not covered in this chapter (for detailed review, see ref. 193). ACC, Acetyl-CoA carboxylase; ACLY, ATP citrate lyase; ADP, adenosine diphosphate; ATP, adenosine
triphosphate; αKG, α-ketoglutarate; Arg, arginine; Asn, asparagine; Asp, aspartate; CA, carbonic anhydrase; 5,10-CH2-THF, 5,10-methylene tetrahydrofolate;
CI, complex I; CK, choline kinase; CPT1, carnitine O-palmitoyltransferase 1; Cyt c, cytochrome c; DHF, dihydrofolate; DHFR, dihydrofolate reductase; dNTP,
deoxynucleotide triphosphate; F1,6BP, fructose-1,6-bisphosphate; F2,6BP, fructose-2,6-bisphosphate; F6P, fructose-6-phosphate; FASN, fatty acid synthase;
GAPDH, glyceraldehyde-3-phosphate dehydrogenase; Gln, glutamine; GLS1, glutaminase 1; Glu, glutamate; GLUD1, glutamate dehydrogenase 1; GLUT,
glucose transporter; G6P, glucose-6-phosphate; HK, hexokinase; HMG-CoA, 3-hydroxy-3-methyl-glutaryl coenzyme A; HMGCR, HMG-CoA reductase; IDH,
isocitrate dehydrogenase; IMP, inosine monophosphate; LDHA, lactate dehydrogenase A; MAG, monoacylglycerol; MCT, monocarboxylate transporter; MGLL,
monoglyceride lipase; NAD+, nicotinamide adenine dinucleotide (oxidixed); NADH, nicotinamide adenine dinucleotide (reduced); NAMPT, nicotinamide
phosphoribosyltransferase; NHE1, Na+/H+ exchanger 1; OAA, oxaloacetate; OXPHOS, oxidative phosphorylation; PC, pyruvate carboxylase; PDK1, pyruvate
dehydrogenase kinase 1; PEP, phosphoenolpyruvate; PFKFB3, 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3; 2PG, 2-phosphoglycerate; 3PG,
3-phosphoglycerate; PGAM1, phosphoglycerate mutase 1; PHGDH, phosphoglycerate dehydrogenase; 3PHP, 3-phosphohydroxypyruvate; PKM2, pyruvate
kinase, muscle, M2 isoform; Pol, DNA polymerase; PPP, pentose phosphate pathway; Q10, coenzyme Q10; R5P, ribose-5-phosphate; RNR, ribonucleotide
reductase; Ser, serine; THF, tetrahydrofolate; TKTL1, transketolase-like protein 1; TYMS, thymidylate synthase; UMP, uridine monophosphate; X5P, xylulose
5-phosphate. (Modified from Galluzi L. (2013) Nature Reviews: Drug Discovery.)
136 Part I: Science and Clinical Oncology

compound), NLG919, and an IDO peptide vaccine. All of these metformin accumulation within tumors was found to correlate with
compounds have shown limited toxicity thus far and are being pursued the degree of mitochondrial metabolic dysfunction including accumula-
both as monotherapies and in combination with other leading tion of both NADH and ROS.206
chemotherapies.184,185 Both preclinical and epidemiologic studies have demonstrated the
antitumor potential of metformin. Some meta-analyses have found
Ornithine Decarboxylase Inhibitors an approximately 30% reduced cancer incidence among type II diabetic
Ornithine decarboxylase (ODC) catalyzes the initiation of de novo metformin users compared with nonusers.207 Dozens of clinical trials
polyamine synthesis and is frequently upregulated in cancer as a examining metformin as either a monotherapy or in combination
consequence of events such as Myc activation,186 ultraviolet (UV) with various chemotherapies are currently ongoing, especially as a
radiation exposure,187 and methylthioadenosine phosphorylase (MTAP) repurposed treatment for breast, endometrial, pancreatic, and prostate
deletion.188 This increase in ODC activity provides an abundant cancers.208
polyamine pool for cell proliferation,189 and promotes signal transduc-
tion by the activation of MAP kinases.190 An FDA-approved ODC Statins
inhibitor, difluoromethylornithine (DFMO, or eflornithine), has been Inhibitors of hydroxymethylglutaryl coenzyme A (HMG-CoA)
shown to effectively reduce polyamine levels, ultimately resulting in reductase, an enzyme in the mevalonate pathway—commonly referred
reduced tumor vascularity191 and metastasis192 in transgenic murine to as statins—are a class of cholesterol-lowering drugs that are used
models. DFMO has been evaluated for the prevention of numerous clinically as a preventative measure against and as a treatment for
cancers193 and is currently under clinical investigation for the treatment cardiovascular disease.209,210 A handful of statins are currently FDA
of Myc-deregulated cancers such as childhood neuroblastoma194; approved, including lovastatin (the most commonly prescribed),
however, early clinical trials investigating the efficacy of DFMO as a simvastatin, fluvastatin, atorvastatin, rosuvastatin, and pravastatin.
general chemotherapeutic agent failed to demonstrate a significant Statins are known to inhibit lipogenesis via inhibition of the mevalonate
outcome, and the drug was found to induce hearing loss in some pathway.211 Overall, preclinical studies have demonstrated that statins
patients.195 Nevertheless, newfound understanding of methionine are able to effectively induce cell death in various types of human
metabolism in cancer may provide better rationale for further investigat- cancer cells.211 Numerous clinical trials have investigated the efficacy
ing these compounds. of statins in the prevention and/or treatment of various types of cancer,
yet the majority of these trials have shown inconclusive results.212 A
Repurposing Common Metabolic Agents meta-analysis found that statin treatment was negatively correlated
with cancer incidence, but this effect was mostly limited to lung and
Non–cancer-treating drugs that have been traditionally used to treat prostate cancer and was concluded to be nonsignificant.213 It is believed
various disorders are currently being investigated as repurposed che- that the inconclusive results found in the large number of observational
motherapeutic agents owing to their relatively reduced toxicity, increased investigations are due to multiple confounding factors that differed
tolerability, and lower cost.196,197 In addition, these FDA-approved among studies, including timing of when treatment was initiated,
compounds have the potential to enter phase III clinical trials more dosage regimen, and heterogeneity of other risk factors (such as obesity
readily than novel drugs because their safety profiles have already been and smoking). However, the use of statins in the prevention and
well established. The following section summarizes some of the current treatment of cancer still remains an attractive prospect and continues
drugs that are being investigated as repurposed cancer treatments. to be under investigation.
Metformin Aspirin
Metformin is the leading therapy prescribed for the management of The nonsteroidal antiinflammatory drug (NSAID) aspirin, a cyclo-
type II diabetes.198 This compound has been shown to reduce hepatic oxygenase (COX) inhibitor that reduces prostaglandin and thromboxane
gluconeogenesis199 and increase insulin sensitivity,200 thereby increasing production, is an over-the-counter medication used to relieve pain
peripheral glucose uptake and reducing circulating levels of glucose. and reduce fever. Aspirin is also commonly recommended by physicians
Although the exact mechanism of action that mediates its efficacy to reduce the risk of heart attack and stroke and the recurrence of
remains unknown,201 metformin has been observed to inhibit complex pulmonary embolism and deep vein thrombosis, because of its ability
I (NADH dehydrogenase) of the mitochondrial respiratory chain,202 to reduce blood clotting.214 Interesting to note, evidence appears to
inhibit mitochondrial glycerophosphate dehydrogenase,199 and activate also link long-term aspirin use to protection against cancer development
AMPK signaling through these mechanisms in certain settings.203 and mortality, particularly colorectal cancer.215 An observational study
Metformin also inhibits insulin production and IGF signaling,204 which investigated aspirin use in 135,965 women and men over the course
can also result in antineoplastic effects. of 32 years and found that regular low-dose (weekly administration
Substantial evidence for the direct effect of metformin on tumor of 0.5 to 1.5 standard tablets) aspirin over the course of 6 years reduced
cell autonomous metabolism is also apparent. Because, as mentioned, overall cancer risk by 3%, with a 15% reduction in gastrointestinal
mitochondria have pleiotropic functions in maintaining tumorigenesis, cancers.216 Although it has been demonstrated that aspirin can activate
disruptions to mitochondria by metformin have been observed to AMPK signaling,217 it is believed that aspirin may exert its antitumor
have global effects, altering mitochondrial metabolism in tumor cells. effects via both off-target (COX-independent) and on-target (COX-
For instance, although it was originally believed that metformin exerted dependent) mechanisms.218 For instance, COX enzymes have been
its antitumor effects primarily via its ability to activate AMPK signaling, found to play a significant role in the development of intestinal tumors
it was found that cancer cell lines that demonstrated high sensitivity in animal models.219 Aspirin may also reduce the incidence of tumor
to metformin tended to exhibit mitochondrial mutations in complex metastasis via its antiplatelet activity220 because platelets have been
I genes; this sensitivity was abolished by the exogenous expression of shown to promote secondary lesion development by shielding tumor
an enzyme (NDI1, derived from yeast) that enables cells to undergo cells from immune responses.221 Nevertheless, more work is needed
oxidative phosphorylation without a functional complex I,205 implying to understand the molecular mechanisms that may contribute to the
an AMPK-independent antineoplastic mechanism. Similar results were antitumor effects of aspirin.
found in murine models, in which tumors of NDI1 transgenic mice
failed to show sensitivity to metformin compared with tumors harbored Vitamins C and D
by mice that did not express NDH1.202 This is also consistent with Evidence suggests that some commonly used compounds contained
metabolic profiles that are observed in the biopsies of ovarian tumors in the diet and used as nutritional supplements, particularly vitamins
obtained from patients taking metformin, in which the level of C and D, may play a much more substantial role in combating cancer
Cancer Metabolism  •  CHAPTER 9 137

progression than was originally believed. For instance, a recently Fluorine-18 fluorodeoxyglucose–positron emission tomography
published preclinical study found that deletion of the vitamin D (18F-FDG–PET) is a technique in which a radioactively labeled glucose
receptor accelerated tumor growth and promoted metastasis in a murine analogue is used to measure glucose uptake, allowing for the anatomic
model of breast cancer,222 implying that vitamin D could act as a localization of tumors when coupled with computed tomography
negative regulator of tumor progression. Another study found that in (CT). The glucose analogue 18F-FDG is phosphorylated by hexokinase
colorectal cancer cell lines exhibiting mutations in either BRAF or similarly to glucose, but is not further metabolized, resulting in the
KRAS, vitamin C was able to induce oxidative stress in cells that accumulation of 18F-FDG in regions with high glucose uptake rates
could inactivate GAPDH (among many other effects), thereby prevent- such as the liver, the brain, and the majority of tumors.241 This allows
ing cancer cells from undergoing glycolysis; this finding extended to not only for the initial identification of tumors, but also for tumor
mouse models, in which vitamin C administration significantly impaired monitoring.
tumor growth.223 Interesting to note, studies have also demonstrated However, this technique exhibits variable sensitivity in detecting
that vitamin C can boost the efficacy of DNA methyltransferase primary tumors localized to different regions; for instance, 18F-FDG–
inhibitors, such as 5-azacytidine and 5-aza-2′-deoxycytidine, by PET has relatively low sensitivity, about 68%, for primary breast
enhancing the catalytic activity of the TET family of DNA demethylases, tumors that are less than 2 cm wide,242 making it an insufficient
which oxidize methylated cytosine residues to hydroxymethyl cyto- technique for initial breast cancer detection. Interesting to note, this
sine.224,225 Given the tolerability of these vitamins, these preclinical sensitivity jumps to almost 90% for recurrent breast tumors, and is
findings are an exciting new field of investigation, and these compounds nearly 100% for metastatic tumors originating from breast tumors,
remain under clinical investigation as an adjuvant therapy.226 so it is frequently used for screening cancer recurrence and metastasis
after initial remission.243 It has also become an established standard
Diet and Exercise in the initial detection of numerous types of cancer including lym-
phoma,244 cervical cancer,245 and lung cancer.246 Important to note,
An emerging topic of investigation is the role of environmental factors there does not appear to be a substantial benefit of using 18F-FDG–PET
that mediate metabolism, such as diet and exercise, in the develop- in the characterization of urologic cancers owing to high signal-to-noise
ment and progression of cancer. It has been estimated that diet could ratios.247 Another limitation of this technique that warrants consid-
account for roughly 35% of the risk factors associated with cancer eration is that inflammatory processes that involve the activation of
development,227 making dietary intervention an attractive approach macrophages and fibroblasts can also induce an increase in the
to cancer prevention. Dietary intervention as an adjuvant therapy FDG-PET signal.248
in cancer treatment has also gained considerable attention. Because
of the differential nutrient requirements of cancer cells, there is a Other Positron Emission Tomography Agents
possibility of inducing antitumor effects through dietary manipulation. Various radiolabeled tracers other than 18F-FDG have been investigated
For instance, under fasting conditions, the brain is capable of using for use in PET imaging in cancer diagnostics for tumors in which
18
ketone bodies in the absence of sufficient glucose, whereas tumors F-FDG–PET imaging is insufficient. For example, acetate is produced
(such as glioblastoma multiforme) remain reliant on an abundant as a result of FA synthesis, and the FA synthase enzyme has been
glucose supply.228,229 This led to the investigation of a high-fat, shown to be upregulated in malignant prostate tumors.249 Therefore
low-carbohydrate (“ketogenic”) diet as a viable adjuvant treatment carbon-11 (11C)–labeled acetate has been explored as a potential
in glioma, in which increased ketone bodies and reduced glucose in PET tracer in the detection of recurrent prostate cancer, with an
circulation leads to increased FAO and ultimately induces a “fasting” observed sensitivity of roughly 75%.250 Similarly, 11C-choline was
state in the patient.230,231 It has been shown in preclinical studies that found to exhibit relatively little accumulation in urine, making it a
a ketogenic diet combined with the targeted therapy temozolomide more advantageous tracer for prostate cancer251; choline is essential for
significantly prolonged survival in a murine model of glioblastoma cellular membrane synthesis and has also been shown to accumulate
multiforme.232 After a handful of case study reports investigating the in malignant tumors.252 As mentioned earlier, another hallmark of
use of a ketogenic diet to treat glioma,233,234 there are a number of tumor biology is the occurrence of hypoxia because proximity to the
ongoing phase II clinical trials examining the efficacy of this dietary vasculature decreases with increasing tumor density.253 The radiotracer
18
intervention as an adjuvant therapy (NCT01754350, NCT01535911, F-fluoromisonidazole (18FMISO) is reduced in hypoxic conditions,
NCT01865162). Similarly, periods of caloric restriction or fasting have resulting in intracellular accumulation; this PET tracer has been investi-
been shown to slow tumor growth and sensitize tumors originating gated in the imaging of breast cancer, glioma, lung cancer, and head and
from various organs to targeted therapies.235,236 Evidence also sug- neck cancers.254
gests that deprivation of certain nutrients has antitumor properties;
one preclinical study found that depletion of the amino acid serine CONCLUSIONS AND FUTURE OUTLOOK
was sufficient to induce the activation of stress pathways and reduce
proliferation in tumors lacking p53.123 Over recent years, the metabolic reprogramming and adaptation that
As detailed previously, obesity has been shown to be a prominent occur in cancer cells to support uncontrolled proliferation have been
risk factor in the development of cancer.237 A significant field of research extensively studied, and their pervasiveness well established. As a result,
is currently investigating the role of energy expenditure in reducing new drug targets and other therapeutic strategies that affect this
tumor development and recurrence. Physical activity has been linked programming are actively being pursued. Challenges in defining the
to prolonged disease-free survival and quality of life among cancer cancer contexts (i.e., which patients to give these drugs to) and thera-
patients,238 and studies have observed a positive effect of exercise on peutic windows limit further translation into the clinic of these advances
the reduction of proangiogenic239 and hypoxic240 factors, resulting in in basic science. In addition, agents that target metabolism such as
reduced tumor vascularity. A role for physical activity in inhibiting or pemetrexed and 5-FU are widely used clinically as effective chemo-
reversing tumor growth is a promising current area of investigation. therapeutic agents; however, in these cases the specificity of these
compounds for particular patients, although apparent clinically, is
Metabolic Biomarkers and Diagnostics poorly understood. That is, standard chemotherapy given within a
Fluorine-18 Fluorodeoxyglucose–Positron cancer patient population results in widely disparate clinical outcomes
ranging from complete resistance to full objective responses. Now
Emission Tomography with newfound insight into the molecular and cellular basis of metabolic
With the discovery of the Warburg effect, it became possible for reprogramming and adaptation, it may be possible to obtain further
glucose uptake to be used as a type of biomarker for tumor location. specificity in precisely defining a priori for these outcomes.
138 Part I: Science and Clinical Oncology

Although much of the recent surge of interest in cancer metabolism such as caloric or total protein intake can readily be controlled if it
has focused on the cell autonomous alterations to metabolic pathways is understood how these factors could influence cancer outcomes in
in cancer cells, it is now more appreciated that these tumor cells ways similar to chemotherapies. Future work in the area of cancer
interact with their environment, which includes their microenviron- metabolism will undoubtedly need to incorporate these factors into
ment, tissue of origin, germline genetics, liver physiology, microbiome, future study design.
and other influences including diet and exercise. Whether and how
variation in these factors can affect metabolic pathways in tumors The complete reference list is available online at
remains a highly active area of research. For example, dietary variables ExpertConsult.com.

KEY REFERENCES
8. Warburg O, Wind F, Negelein E. The metabolism 63. Haq R, Shoag J, Andreu-Perez P, et al. Oncogenic 138. Bhaskaran K, Douglas I, Forbes H, dos-Santos-
of tumors in the body. J Gen Physiol. 1927;8(6): BRAF regulates oxidative metabolism via PGC1alpha Silva I, Leon DA, Smeeth L. Body-mass index
519–530. and MITF. Cancer Cell. 2013;23(3):302–315. and risk of 22 specific cancers: a population-based
9. Fantin VR, St-Pierre J, Leder P. Attenuation 69. Hatzivassiliou G, Zhao F, Bauer DE, et al. ATP cohort study of 5.24 million UK adults. Lancet.
of LDH-A expression uncovers a link between citrate lyase inhibition can suppress tumor cell 2014;384(9945):755–765.
glycolysis, mitochondrial physiology, and tumor growth. Cancer Cell. 2005;8(4):311–321. 141. Nieman KM, Kenny HA, Penicka CV, et al.
maintenance. Cancer Cell. 2006;9(6):425–434. 71. Shaw RJ, Kosmatka M, Bardeesy N, et al. The tumor Adipocytes promote ovarian cancer metastasis and
10. Shim H, Chun YS, Lewis BC, Dang CV. A unique suppressor LKB1 kinase directly activates AMP- provide energy for rapid tumor growth. Nat Med.
glucose-dependent apoptotic pathway induced activated kinase and regulates apoptosis in response 2011;17(11):1498–1503.
by c-Myc. Proc Natl Acad Sci USA. 1998;95(4): to energy stress. Proc Natl Acad Sci USA. 2004; 144. Ruderman NB, Kapeller R, White MF, Cantley LC.
1511–1516. 101(10):3329–3335. Activation of phosphatidylinositol 3-kinase by insulin.
15. Liberti MV, Locasale JW. The Warburg effect: 77. Carracedo A, Cantley LC, Pandolfi PP. Cancer Proc Natl Acad Sci USA. 1990;87(4):1411–1415.
how does it benefit cancer cells? Trends Biochem metabolism: fatty acid oxidation in the limelight. Nat 160. Gonen N, Assaraf YG. Antifolates in cancer therapy:
Sci. 2016;41(3):211–218. Rev Cancer. 2013;13(4):227–232. structure, activity and mechanisms of drug resistance.
18. Locasale JW, Cantley LC. Metabolic flux and the 91. Gao X, Reid MA, Kong M, Locasale JW. Metabolic Drug Resist Updat. 2012;15(4):183–210.
regulation of mammalian cell growth. Cell Metab. interactions with cancer epigenetics. Mol Aspects Med. 170. Garber K. Cancer anabolic metabolism inhibitors
2011;14(4):443–451. 2016. move into clinic. Nat Biotechnol. 2016;34(8):794–
19. Locasale JW, Grassian AR, Melman T, et al. 97. Wellen KE, Hatzivassiliou G, Sachdeva UM, Bui 795.
Phosphoglycerate dehydrogenase diverts glycolytic TV, Cross JR, Thompson CB. ATP-citrate lyase links 185. Galluzzi L, Kepp O, Vander Heiden MG, Kroemer
flux and contributes to oncogenesis. Nat Genet. cellular metabolism to histone acetylation. Science. G. Metabolic targets for cancer therapy. Nat Rev
2011;43(9):869–874. 2009;324(5930):1076–1080. Drug Discov. 2013;12(11):829–846.
23. Boroughs LK, DeBerardinis RJ. Metabolic pathways 100. Yan H, Parsons DW, Jin G, et al. IDH1 and IDH2 193. Gerner EW, Meyskens FL Jr. Polyamines and cancer:
promoting cancer cell survival and growth. Nat Cell mutations in gliomas. N Engl J Med. 2009;360(8): old molecules, new understanding. Nat Rev Cancer.
Biol. 2015;17(4):351–359. 765–773. 2004;4(10):781–792.
24. Wise DR, DeBerardinis RJ, Mancuso A, et al. 102. Dang L, White DW, Gross S, et al. Cancer-associated 197. Ashburn TT, Thor KB. Drug repositioning: identify-
Myc regulates a transcriptional program that IDH1 mutations produce 2-hydroxyglutarate. Nature. ing and developing new uses for existing drugs. Nat
stimulates mitochondrial glutaminolysis and leads 2009;462(7274):739–744. Rev Drug Discov. 2004;3(8):673–683.
to glutamine addiction. Proc Natl Acad Sci USA. 104. Lu C, Ward PS, Kapoor GS, et al. IDH mutation 202. Wheaton WW, Weinberg SE, Hamanaka RB, et al.
2008;105(48):18782–18787. impairs histone demethylation and results in a block Metformin inhibits mitochondrial complex I of
27. Commisso C, Davidson SM, Soydaner-Azeloglu to cell differentiation. Nature. 2012;483(7390): cancer cells to reduce tumorigenesis. Elife. 2014;3:
RG, et al. Macropinocytosis of protein is an amino 474–478. e02242.
acid supply route in Ras-transformed cells. Nature. 108. Pan M, Reid MA, Lowman XH, et al. Regional 206. Liu X, Romero IL, Litchfield LM, Lengyel E,
2013;497(7451):633–637. glutamine deficiency in tumours promotes dedif- Locasale JW. Metformin targets central carbon
28. Degenhardt K, Mathew R, Beaudoin B, et al. ferentiation through inhibition of histone demeth- metabolism and reveals mitochondrial requirements
Autophagy promotes tumor cell survival and restricts ylation. Nat Cell Biol. 2016;18(10):1090–1101. in human cancers. Cell Metab. 2016;24(5):728–739.
necrosis, inflammation, and tumorigenesis. Cancer 109. Xiao M, Yang H, Xu W, et al. Inhibition of alpha- 217. Hawley SA, Fullerton MD, Ross FA, et al. The ancient
Cell. 2006;10(1):51–64. KG-dependent histone and DNA demethylases drug salicylate directly activates AMP-activated
30. Altman BJ, Stine ZE, Dang CV. From Krebs to by fumarate and succinate that are accumulated protein kinase. Science. 2012;336(6083):918–922.
clinic: glutamine metabolism to cancer therapy. Nat in mutations of FH and SDH tumor suppressors. 220. Rothwell PM, Wilson M, Price JF, Belch JF,
Rev Cancer. 2016;16(10):619–634. Genes Dev. 2012;26(12):1326–1338. Meade TW, Mehta Z. Effect of daily aspirin on
34. DeBerardinis RJ, Mancuso A, Daikhin E, et al. 115. Nakazawa MS, Keith B, Simon MC. Oxygen avail- risk of cancer metastasis: a study of incident cancers
Beyond aerobic glycolysis: transformed cells can ability and metabolic adaptations. Nat Rev Cancer. during randomised controlled trials. Lancet. 2012;
engage in glutamine metabolism that exceeds the 2016;16(10):663–673. 379(9826):1591–1601.
requirement for protein and nucleotide synthesis. Proc 119. Kamphorst JJ, Nofal M, Commisso C, et al. Human 235. Lee C, Raffaghello L, Brandhorst S, et al. Fasting cycles
Natl Acad Sci USA. 2007;104(49):19345–19350. pancreatic cancer tumors are nutrient poor and retard growth of tumors and sensitize a range of cancer
36. Davidson SM, Papagiannakopoulos T, Olenchock tumor cells actively scavenge extracellular protein. cell types to chemotherapy. Sci Transl Med. 2012;
BA, et al. Environment impacts the metabolic Cancer Res. 2015;75(3):544–553. 4(124):124ra127.
dependencies of Ras-driven non–small cell lung 120. Reid MA, Wang WI, Rosales KR, Welliver MX, Pan 236. Levine ME, Suarez JA, Brandhorst S, et al. Low
cancer. Cell Metab. 2016;23(3):517–528. M, Kong M. The B55alpha subunit of PP2A drives protein intake is associated with a major reduction
38. Xiang Y, Stine ZE, Xia J, et al. Targeted inhibition a p53-dependent metabolic adaptation to glutamine in IGF-1, cancer, and overall mortality in the 65
of tumor-specific glutaminase diminishes cell- deprivation. Mol Cell. 2013;50(2):200–211. and younger but not older population. Cell Metab.
autonomous tumorigenesis. J Clin Invest. 2015;125(6): 123. Maddocks OD, Berkers CR, Mason SM, et al. Serine 2014;19(3):407–417.
2293–2306. starvation induces stress and p53-dependent metabolic 237. Khandekar MJ, Cohen P, Spiegelman BM. Molecular
44. Locasale JW. Serine, glycine and one-carbon units: remodelling in cancer cells. Nature. 2013;493(7433): mechanisms of cancer development in obesity. Nat
cancer metabolism in full circle. Nat Rev Cancer. 2013; 542–546. Rev Cancer. 2011;11(12):886–895.
13(8):572–583. 124. Hensley CT, Faubert B, Yuan Q, et al. Metabolic het- 240. Betof AS, Lascola CD, Weitzel D, et al. Modulation
48. Mentch SJ, Mehrmohamadi M, Huang L, et al. erogeneity in human lung tumors. Cell. 2016;164(4): of murine breast tumor vascularity, hypoxia and
Histone methylation dynamics and gene regulation 681–694. chemotherapeutic response by exercise. J Natl Cancer
occur through the sensing of one-carbon metabolism. 128. Colegio OR, Chu NQ, Szabo AL, et al. Functional Inst. 2015;107(5).
Cell Metab. 2015;22(5):861–873. polarization of tumour-associated macrophages by
54. Laplante M, Sabatini DM. mTOR signaling in growth tumour-derived lactic acid. Nature. 2014;513(7519):
control and disease. Cell. 2012;149(2):274–293. 559–563.
Cancer Metabolism  •  CHAPTER 9 138.e1
138.e1

REFERENCES
1. Berg JM, Tymoczko JL, Stryer L, Stryer L. Biochem- to glutamine addiction. Proc Natl Acad Sci USA. 44. Locasale JW. Serine, glycine and one-carbon units:
istry. 6th ed. New York: WH Freeman; 2007. 2008;105(48):18782–18787. cancer metabolism in full circle. Nat Rev Cancer.
2. Lehninger AL, Nelson DL, Cox MM. Lehninger 25. Gao P, Tchernyshyov I, Chang TC, et al. c-Myc 2013;13(8):572–583.
Principles of Biochemistry. 6th ed. New York: W.H. suppression of miR-23a/b enhances mitochondrial 45. Mullarky E, Lucki NC, Beheshti Zavareh R, et al.
Freeman; 2013. glutaminase expression and glutamine metabolism. Identification of a small molecule inhibitor of
3. Rolland F, Winderickx J, Thevelein JM. Glucose- Nature. 2009;458(7239):762–765. 3-phosphoglycerate dehydrogenase to target serine
sensing and -signalling mechanisms in yeast. FEMS 26. Son J, Lyssiotis CA, Ying H, et al. Glutamine supports biosynthesis in cancers. Proc Natl Acad Sci USA.
Yeast Res. 2002;2(2):183–201. pancreatic cancer growth through a KRAS-regulated 2016;113(7):1778–1783.
4. Sherman IW. Malaria: Parasite Biology, Pathogenesis, metabolic pathway. Nature. 2013;496(7443): 46. Pacold ME, Brimacombe KR, Chan SH, et al. A
and Protection. Washington, DC: ASM Press; 1998. 101–105. PHGDH inhibitor reveals coordination of serine
5. Pfeiffer T, Schuster S, Bonhoeffer S. Cooperation 27. Commisso C, Davidson SM, Soydaner-Azeloglu synthesis and one-carbon unit fate. Nat Chem Biol.
and competition in the evolution of ATP-producing RG, et al. Macropinocytosis of protein is an amino 2016;12(6):452–458.
pathways. Science. 2001;292(5516):504–507. acid supply route in Ras-transformed cells. Nature. 47. Wang Q, Liberti MV, Liu P, et al. Rational design
6. Shestov AA, Liu X, Ser Z, et al. Quantitative 2013;497(7451):633–637. of selective allosteric inhibitors of PHGDH and
determinants of aerobic glycolysis identify flux 28. Degenhardt K, Mathew R, Beaudoin B, et al. serine synthesis with anti-tumor activity. Cell Chem
through the enzyme GAPDH as a limiting step. Elife. Autophagy promotes tumor cell survival and restricts Biol. 2017;24(1):55–65.
2014;3. necrosis, inflammation, and tumorigenesis. Cancer 48. Mentch SJ, Mehrmohamadi M, Huang L, et al.
7. Warburg O, Posener K, Negelein E. On the Cell. 2006;10(1):51–64. Histone methylation dynamics and gene regulation
metabolism of carcinoma cells. Biochem Z. 1924;152: 29. Yang S, Wang X, Contino G, et al. Pancreatic cancers occur through the sensing of one-carbon metabolism.
309–344. require autophagy for tumor growth. Genes Dev. 2011; Cell Metab. 2015;22(5):861–873.
8. Warburg O, Wind F, Negelein E. The metabolism 25(7):717–729. 49. Davis SR, Stacpoole PW, Williamson J, et al. Tracer-
of tumors in the body. J Gen Physiol. 1927;8(6): 30. Altman BJ, Stine ZE, Dang CV. From Krebs to derived total and folate-dependent homocysteine
519–530. clinic: glutamine metabolism to cancer therapy. Nat remethylation and synthesis rates in humans indicate
9. Fantin VR, St-Pierre J, Leder P. Attenuation of Rev Cancer. 2016;16(10):619–634. that serine is the main one-carbon donor. Am J
LDH-A expression uncovers a link between glycolysis, 31. Bergstrom J, Furst P, Noree LO, Vinnars E. Intracel- Physiol Endocrinol Metab. 2004;286(2):E272–E279.
mitochondrial physiology, and tumor maintenance. lular free amino acid concentration in human muscle 50. Dillon BJ, Prieto VG, Curley SA, et al. Incidence
Cancer Cell. 2006;9(6):425–434. tissue. J Appl Physiol. 1974;36(6):693–697. and distribution of argininosuccinate synthetase
10. Shim H, Chun YS, Lewis BC, Dang CV. A unique 32. Welbourne TC. Ammonia production and glutamine deficiency in human cancers: a method for identify-
glucose-dependent apoptotic pathway induced incorporation into glutathione in the functioning ing cancers sensitive to arginine deprivation. Cancer.
by c-Myc. Proc Natl Acad Sci USA. 1998;95(4): rat kidney. Can J Biochem. 1979;57(3):233– 2004;100(4):826–833.
1511–1516. 237. 51. Grohmann U, Bronte V. Control of immune
11. Le A, Stine ZE, Nguyen C, et al. Tumorigenicity 33. Jiang L, Shestov AA, Swain P, et al. Reductive carbox- response by amino acid metabolism. Immunol Rev.
of hypoxic respiring cancer cells revealed by a ylation supports redox homeostasis during anchorage- 2010;236:243–264.
hypoxia-cell cycle dual reporter. Proc Natl Acad Sci independent growth. Nature. 2016;532(7598): 52. Mellor AL, Munn DH. IDO expression by dendritic
USA. 2014;111(34):12486–12491. 255–258. cells: tolerance and tryptophan catabolism. Nat Rev
12. Gerriets VA, Kishton RJ, Nichols AG, et al. Metabolic 34. DeBerardinis RJ, Mancuso A, Daikhin E, et al. Immunol. 2004;4(10):762–774.
programming and PDHK1 control CD4+ T cell Beyond aerobic glycolysis: transformed cells can 53. Mayers JR, Wu C, Clish CB, et al. Elevation of
subsets and inflammation. J Clin Invest. 2015;125(1): engage in glutamine metabolism that exceeds the circulating branched-chain amino acids is an early
194–207. requirement for protein and nucleotide synthesis. event in human pancreatic adenocarcinoma develop-
13. Vincent AS, Phan TT, Mukhopadhyay A, Lim Proc Natl Acad Sci USA. 2007;104(49):19345– ment. Nat Med. 2014;20(10):1193–1198.
HY, Halliwell B, Wong KP. Human skin keloid 19350. 54. Laplante M, Sabatini DM. mTOR signaling in growth
fibroblasts display bioenergetics of cancer cells. J 35. DeBerardinis RJ, Lum JJ, Hatzivassiliou G, Thompson control and disease. Cell. 2012;149(2):274–293.
Invest Dermatol. 2008;128(3):702–709. CB. The biology of cancer: metabolic reprogramming 55. Wallace DC. Mitochondria and cancer. Nat Rev
14. Ben-Haim S, Ell P. 18F-FDG PET and PET/CT fuels cell growth and proliferation. Cell Metab. 2008; Cancer. 2012;12(10):685–698.
in the evaluation of cancer treatment response. J 7(1):11–20. 56. Ames BN, Shigenaga MK, Hagen TM. Oxidants, anti-
Nucl Med. 2009;50(1):88–99. 36. Davidson SM, Papagiannakopoulos T, Olenchock oxidants, and the degenerative diseases of aging. Proc
15. Liberti MV, Locasale JW. The Warburg effect: BA, et al. Environment impacts the metabolic Natl Acad Sci USA. 1993;90(17):7915–7922.
how does it benefit cancer cells? Trends Biochem dependencies of ras-driven non–small cell lung 57. Weinberg F, Hamanaka R, Wheaton WW, et al.
Sci. 2016;41(3):211–218. cancer. Cell Metab. 2016;23(3):517–528. Mitochondrial metabolism and ROS generation are
16. Warburg O. On the origin of cancer cells. Science. 37. Gross MI, Demo SD, Dennison JB, et al. Antitumor essential for Kras-mediated tumorigenicity. Proc Natl
1956;123(3191):309–314. activity of the glutaminase inhibitor CB-839 in triple- Acad Sci USA. 2010;107(19):8788–8793.
17. Slavov N, Budnik BA, Schwab D, Airoldi EM, negative breast cancer. Mol Cancer Ther. 2014;13(4): 58. Weinhouse S, Millington RH, Wenner CE.
van Oudenaarden A. Constant growth rate can be 890–901. Metabolism of neoplastic tissue. I. The oxidation of
supported by decreasing energy flux and increasing 38. Xiang Y, Stine ZE, Xia J, et al. Targeted inhibition carbohydrate and fatty acids in transplanted tumors.
aerobic glycolysis. Cell Rep. 2014;7(3):705–714. of tumor-specific glutaminase diminishes cell- Cancer Res. 1951;11(11):845–850.
18. Locasale JW, Cantley LC. Metabolic flux and the autonomous tumorigenesis. J Clin Invest. 2015;125(6): 59. Wenner CE, Spirtes MA, Weinhouse S. Metabolism
regulation of mammalian cell growth. Cell Metab. 2293–2306. of neoplastic tissue. II. A survey of enzymes of the
2011;14(4):443–451. 39. Kung HN, Marks JR, Chi JT. Glutamine synthe- citric acid cycle in transplanted tumors. Cancer Res.
19. Locasale JW, Grassian AR, Melman T, et al. Phos- tase is a genetic determinant of cell type-specific 1952;12(1):44–49.
phoglycerate dehydrogenase diverts glycolytic flux glutamine independence in breast epithelia. PLoS 60. Fogal V, Richardson AD, Karmali PP, Scheffler
and contributes to oncogenesis. Nat Genet. 2011; Genet. 2011;7(8):e1002229. IE, Smith JW, Ruoslahti E. Mitochondrial p32
43(9):869–874. 40. Herranz D, Ambesi-Impiombato A, Sudderth J, et al. protein is a critical regulator of tumor metabolism
20. Lunt SY, Vander Heiden MG. Aerobic glycolysis: Metabolic reprogramming induces resistance to anti- via maintenance of oxidative phosphorylation. Mol
meeting the metabolic requirements of cell prolifera- NOTCH1 therapies in T cell acute lymphoblastic Cell Biol. 2010;30(6):1303–1318.
tion. Annu Rev Cell Dev Biol. 2011;27:441–464. leukemia. Nat Med. 2015;21(10):1182–1189. 61. Weinberg SE, Chandel NS. Targeting mitochondria
21. Gatenby RA, Gawlinski ET. A reaction-diffusion 41. MacFarlane AJ, Liu X, Perry CA, et al. Cytoplasmic metabolism for cancer therapy. Nat Chem Biol.
model of cancer invasion. Cancer Res. 1996;56(24): serine hydroxymethyltransferase regulates the meta- 2015;11(1):9–15.
5745–5753. bolic partitioning of methylenetetrahydrofolate but 62. Vazquez F, Lim JH, Chim H, et al. PGC1alpha
22. Estrella V, Chen T, Lloyd M, et al. Acidity generated is not essential in mice. J Biol Chem. 2008;283(38): expression defines a subset of human melanoma
by the tumor microenvironment drives local invasion. 25846–25853. tumors with increased mitochondrial capacity
Cancer Res. 2013;73(5):1524–1535. 42. Kalhan SC, Uppal SO, Moorman JL, et al. Metabolic and resistance to oxidative stress. Cancer Cell.
23. Boroughs LK, DeBerardinis RJ. Metabolic pathways and genomic response to dietary isocaloric protein 2013;23(3):287–301.
promoting cancer cell survival and growth. Nat Cell restriction in the rat. J Biol Chem. 2011;286(7): 63. Haq R, Shoag J, Andreu-Perez P, et al. Oncogenic
Biol. 2015;17(4):351–359. 5266–5277. BRAF regulates oxidative metabolism via PGC1alpha
24. Wise DR, DeBerardinis RJ, Mancuso A, et al. 43. Yang M, Vousden KH. Serine and one-carbon and MITF. Cancer Cell. 2013;23(3):302–315.
Myc regulates a transcriptional program that metabolism in cancer. Nat Rev Cancer. 2016;16(10): 64. Ookhtens M, Kannan R, Lyon I, Baker N. Liver and
stimulates mitochondrial glutaminolysis and leads 650–662. adipose tissue contributions to newly formed fatty acids
138.e2 Part I: Science and Clinical Oncology

in an ascites tumor. Am J Physiol. 1984;247(1 Pt 2): 86. Sharma S, Kelly TK, Jones PA. Epigenetics in cancer. demethylation. Nat Cell Biol. 2016;18(10):1090–
R146–R153. Carcinogenesis. 2010;31(1):27–36. 1101.
65. Medes G, Thomas A, Weinhouse S. Metabolism of 87. Suva ML, Riggi N, Bernstein BE. Epigenetic 109. Xiao M, Yang H, Xu W, et al. Inhibition of alpha-
neoplastic tissue. IV. A study of lipid synthesis in neo- reprogramming in cancer. Science. 2013;339(6127): KG-dependent histone and DNA demethylases
plastic tissue slices in vitro. Cancer Res. 1953;13(1): 1567–1570. by fumarate and succinate that are accumulated
27–29. 88. Ptashne M. How eukaryotic transcriptional activators in mutations of FH and SDH tumor suppressors.
66. Wakil SJ, Stoops JK, Joshi VC. Fatty acid synthesis work. Nature. 1988;335(6192):683–689. Genes Dev. 2012;26(12):1326–1338.
and its regulation. Annu Rev Biochem. 1983;52: 89. Ullrich A, Schlessinger J. Signal transduction 110. Hanahan D, Coussens LM. Accessories to the crime:
537–579. by receptors with tyrosine kinase activity. Cell. functions of cells recruited to the tumor microen-
67. Currie E, Schulze A, Zechner R, Walther TC, Farese 1990;61(2):203–212. vironment. Cancer Cell. 2012;21(3):309–322.
RV Jr. Cellular fatty acid metabolism and cancer. 90. Yun J, Johnson JL, Hanigan CL, Locasale JW. 111. Less JR, Skalak TC, Sevick EM, Jain RK. Micro-
Cell Metab. 2013;18(2):153–161. Interactions between epigenetics and metabolism vascular architecture in a mammary carcinoma:
68. Bauer DE, Hatzivassiliou G, Zhao F, Andreadis C, in cancers. Front Oncol. 2012;2:163. branching patterns and vessel dimensions. Cancer
Thompson CB. ATP citrate lyase is an important 91. Gao X, Reid MA, Kong M, Locasale JW. Metabolic Res. 1991;51(1):265–273.
component of cell growth and transformation. interactions with cancer epigenetics. Mol Aspects Med. 112. McKeown SR. Defining normoxia, physoxia and
Oncogene. 2005;24(41):6314–6322. 2016. hypoxia in tumours-implications for treatment
69. Hatzivassiliou G, Zhao F, Bauer DE, et al. ATP 92. Kinnaird A, Zhao S, Wellen KE, Michelakis ED. response. Br J Radiol. 2014;87(1035):20130676.
citrate lyase inhibition can suppress tumor cell Metabolic control of epigenetics in cancer. Nat Rev 113. Rumsey WL, Schlosser C, Nuutinen EM, Robiolio
growth. Cancer Cell. 2005;8(4):311–321. Cancer. 2016;16(11):694–707. M, Wilson DF. Cellular energetics and the oxygen
70. Migita T, Narita T, Nomura K, et al. ATP citrate 93. Mentch SJ, Locasale JW. One-carbon metabolism dependence of respiration in cardiac myocytes
lyase: activation and therapeutic implications in and epigenetics: understanding the specificity. Ann isolated from adult rat. J Biol Chem. 1990;265(26):
non–small cell lung cancer. Cancer Res. 2008; N Y Acad Sci. 2016;1363:91–98. 15392–15402.
68(20):8547–8554. 94. Shyh-Chang N, Locasale JW, Lyssiotis CA, 114. Rankin EB, Giaccia AJ. The role of hypoxia-
71. Shaw RJ, Kosmatka M, Bardeesy N, et al. The et al. Influence of threonine metabolism on S- inducible factors in tumorigenesis. Cell Death Differ.
tumor suppressor LKB1 kinase directly activates adenosylmethionine and histone methylation. Science. 2008;15(4):678–685.
AMP-activated kinase and regulates apoptosis in 2013;339(6116):222–226. 115. Nakazawa MS, Keith B, Simon MC. Oxygen avail-
response to energy stress. Proc Natl Acad Sci USA. 95. Nishikawa K, Iwamoto Y, Kobayashi Y, et al. ability and metabolic adaptations. Nat Rev Cancer.
2004;101(10):3329–3335. DNA methyltransferase 3a regulates osteoclast 2016;16(10):663–673.
72. Chajes V, Cambot M, Moreau K, Lenoir GM, differentiation by coupling to an S-adenosylme- 116. Semenza GL. Targeting HIF-1 for cancer therapy.
Joulin V. Acetyl-CoA carboxylase alpha is essential to thionine-producing metabolic pathway. Nat Med. Nat Rev Cancer. 2003;3(10):721–732.
breast cancer cell survival. Cancer Res. 2006;66(10): 2015;21(3):281–287. 117. Zeng W, Wan R, Zheng Y, Singh SR, Wei Y.
5287–5294. 96. Cai L, Sutter BM, Li B, Tu BP. Acetyl-CoA induces cell Hypoxia, stem cells and bone tumor. Cancer Lett.
73. Beckers A, Organe S, Timmermans L, et al. Chemical growth and proliferation by promoting the acetylation 2011;313(2):129–136.
inhibition of acetyl-CoA carboxylase induces growth of histones at growth genes. Mol Cell. 2011;42(4): 118. Lee KE, Simon MC. From stem cells to cancer
arrest and cytotoxicity selectively in cancer cells. 426–437. stem cells: HIF takes the stage. Curr Opin Cell Biol.
Cancer Res. 2007;67(17):8180–8187. 97. Wellen KE, Hatzivassiliou G, Sachdeva UM, Bui 2012;24(2):232–235.
74. Jeon SM, Chandel NS, Hay N. AMPK regulates TV, Cross JR, Thompson CB. ATP-citrate lyase links 119. Kamphorst JJ, Nofal M, Commisso C, et al. Human
NADPH homeostasis to promote tumour cell sur- cellular metabolism to histone acetylation. Science. pancreatic cancer tumors are nutrient poor and
vival during energy stress. Nature. 2012;485(7400): 2009;324(5930):1076–1080. tumor cells actively scavenge extracellular protein.
661–665. 98. Chalkiadaki A, Guarente L. The multifaceted func- Cancer Res. 2015;75(3):544–553.
75. Pizer ES, Thupari J, Han WF, et al. Malonyl- tions of sirtuins in cancer. Nat Rev Cancer. 2015; 120. Reid MA, Wang WI, Rosales KR, Welliver MX, Pan
coenzyme-A is a potential mediator of cytotoxicity 15(10):608–624. M, Kong M. The B55alpha subunit of PP2A drives
induced by fatty-acid synthase inhibition in human 99. Mardis ER, Ding L, Dooling DJ, et al. Recurring a p53-dependent metabolic adaptation to glutamine
breast cancer cells and xenografts. Cancer Res. mutations found by sequencing an acute myeloid deprivation. Mol Cell. 2013;50(2):200–211.
2000;60(2):213–218. leukemia genome. N Engl J Med. 2009;361(11): 121. Reid MA, Lowman XH, Pan M, et al. IKKbeta
76. Menendez JA, Lupu R. Fatty acid synthase and the 1058–1066. promotes metabolic adaptation to glutamine
lipogenic phenotype in cancer pathogenesis. Nat Rev 100. Yan H, Parsons DW, Jin G, et al. IDH1 and IDH2 deprivation via phosphorylation and inhibition of
Cancer. 2007;7(10):763–777. mutations in gliomas. N Engl J Med. 2009;360(8): PFKFB3. Genes Dev. 2016;30(16):1837–1851.
77. Carracedo A, Cantley LC, Pandolfi PP. Cancer 765–773. 122. Tran TQ, Lowman XH, Reid MA, et al. Tumor-
metabolism: fatty acid oxidation in the limelight. Nat 101. Zhao S, Lin Y, Xu W, et al. Glioma-derived mutations associated mutant p53 promotes cancer cell survival
Rev Cancer. 2013;13(4):227–232. in IDH1 dominantly inhibit IDH1 catalytic activity upon glutamine deprivation through p21 induction.
78. Zaugg K, Yao Y, Reilly PT, et al. Carnitine palmi- and induce HIF-1alpha. Science. 2009;324(5924): Oncogene. 2016.
toyltransferase 1C promotes cell survival and tumor 261–265. 123. Maddocks OD, Berkers CR, Mason SM, et al.
growth under conditions of metabolic stress. Genes 102. Dang L, White DW, Gross S, et al. Cancer-associated Serine starvation induces stress and p53-dependent
Dev. 2011;25(10):1041–1051. IDH1 mutations produce 2-hydroxyglutarate. Nature. metabolic remodelling in cancer cells. Nature.
79. Schafer ZT, Grassian AR, Song L, et al. Anti- 2009;462(7274):739–744. 2013;493(7433):542–546.
oxidant and oncogene rescue of metabolic defects 103. Xu W, Yang H, Liu Y, et al. Oncometabolite 124. Hensley CT, Faubert B, Yuan Q, et al. Metabolic het-
caused by loss of matrix attachment. Nature. 2-hydroxyglutarate is a competitive inhibitor of erogeneity in human lung tumors. Cell. 2016;164(4):
2009;461(7260):109–113. alpha-ketoglutarate-dependent dioxygenases. Cancer 681–694.
80. Camarda R, Zhou AY, Kohnz RA, et al. Inhibition Cell. 2011;19(1):17–30. 125. Swietach P, Vaughan-Jones RD, Harris AL. Regula-
of fatty acid oxidation as a therapy for MYC- 104. Lu C, Ward PS, Kapoor GS, et al. IDH mutation tion of tumor pH and the role of carbonic anhydrase
overexpressing triple-negative breast cancer. Nat Med. impairs histone demethylation and results in a block 9. Cancer Metastasis Rev. 2007;26(2):299–310.
2016;22(4):427–432. to cell differentiation. Nature. 2012;483(7390): 126. Martinez-Zaguilan R, Seftor EA, Seftor RE, Chu
81. Loftus TM, Jaworsky DE, Frehywot GL, et al. 474–478. YW, Gillies RJ, Hendrix MJ. Acidic pH enhances
Reduced food intake and body weight in mice 105. Wang F, Travins J, DeLaBarre B, et al. Targeted the invasive behavior of human melanoma cells.
treated with fatty acid synthase inhibitors. Science. inhibition of mutant IDH2 in leukemia cells induces Clin Exp Metastasis. 1996;14(2):176–186.
2000;288(5475):2379–2381. cellular differentiation. Science. 2013;340(6132): 127. Rothberg JM, Bailey KM, Wojtkowiak JW, et al.
82. Rohrig F, Schulze A. The multifaceted roles of fatty 622–626. Acid-mediated tumor proteolysis: contribution of cys-
acid synthesis in cancer. Nat Rev Cancer. 2016;16(11): 106. Rohle D, Popovici-Muller J, Palaskas N, et al. teine cathepsins. Neoplasia. 2013;15(10):1125–1137.
732–749. An inhibitor of mutant IDH1 delays growth and 128. Colegio OR, Chu NQ, Szabo AL, et al. Functional
83. Graeser R, Bornmann C, Esser N, et al. Anti- promotes differentiation of glioma cells. Science. polarization of tumour-associated macrophages by
metastatic effects of liposomal gemcitabine and 2013;340(6132):626–630. tumour-derived lactic acid. Nature. 2014;513(7519):
empty liposomes in an orthotopic mouse model 107. Carey BW, Finley LW, Cross JR, Allis CD, Thompson 559–563.
of pancreatic cancer. Pancreas. 2009;38(3):330–337. CB. Intracellular alpha-ketoglutarate maintains 129. Wahl DR, Byersdorfer CA, Ferrara JL, Opipari
84. Morgan HD, Santos F, Green K, Dean W, Reik W. the pluripotency of embryonic stem cells. Nature. AW Jr, Glick GD. Distinct metabolic programs in
Epigenetic reprogramming in mammals. Hum Mol 2015;518(7539):413–416. activated T cells: opportunities for selective immu-
Genet. 2005;14(Spec1):R47–R58. 108. Pan M, Reid MA, Lowman XH, et al. Regional nomodulation. Immunol Rev. 2012;249(1):104–115.
85. Dawson MA, Kouzarides T. Cancer epigenetics: from glutamine deficiency in tumours promotes 130. Ron-Harel N, Sharpe AH, Haigis MC. Mitochon-
mechanism to therapy. Cell. 2012;150(1):12–27. dedifferentiation through inhibition of histone drial metabolism in T cell activation and senescence:
Cancer Metabolism  •  CHAPTER 9 138.e3
138.e3

a mini-review. Gerontology. 2015;61(2):131– 151. Dalamaga M, Diakopoulos KN, Mantzoros CS. The 173. Aledo JC, Gomez-Fabre PM, Olalla L, Marquez J.
138. role of adiponectin in cancer: a review of current Identification of two human glutaminase loci and
131. Mellor AL, Munn DH. Creating immune privilege: evidence. Endocr Rev. 2012;33(4):547–594. tissue-specific expression of the two related genes.
active local suppression that benefits friends, but 152. Uddin S, Hussain AR, Siraj AK, Khan OS, Bavi PP, Mamm Genome. 2000;11(12):1107–1110.
protects foes. Nat Rev Immunol. 2008;8(1):74–80. Al-Kuraya KS. Role of leptin and its receptors in 174. Maier T, Jenni S, Ban N. Architecture of mammalian
132. Fallarino F, Grohmann U, Vacca C, et al. T cell the pathogenesis of thyroid cancer. Int J Clin Exp fatty acid synthase at 4.5 A resolution. Science.
apoptosis by tryptophan catabolism. Cell Death Pathol. 2011;4(7):637–643. 2006;311(5765):1258–1262.
Differ. 2002;9(10):1069–1077. 153. Garofalo C, Surmacz E. Leptin and cancer. J Cell 175. Sebastiani V, Botti C, Di Tondo U, et al. Tissue
133. Fooksman DR, Vardhana S, Vasiliver-Shamis G, et al. Physiol. 2006;207(1):12–22. microarray analysis of FAS, Bcl-2, Bcl-x, ER, PgR,
Functional anatomy of T cell activation and synapse 154. Fox JT, Stover PJ. Folate-mediated one-carbon Hsp60, p53 and Her2-neu in breast carcinoma.
formation. Annu Rev Immunol. 2010;28:79–105. metabolism. Vitam Horm. 2008;79:1–44. Anticancer Res. 2006;26(4B):2983–2987.
134. Patsoukis N, Bardhan K, Chatterjee P, et al. PD-1 155. Visentin M, Zhao R, Goldman ID. The antifolates. 176. Ventura R, Mordec K, Waszczuk J, et al. Inhibition
alters T-cell metabolic reprogramming by inhibiting Hematol Oncol Clin North Am. 2012;26(3):629– of de novo palmitate synthesis by fatty acid synthase
glycolysis and promoting lipolysis and fatty acid 648, ix. induces apoptosis in tumor cells by remodeling cell
oxidation. Nat Commun. 2015;6:6692. 156. Fabre I, Fabre G, Goldman ID. Polyglutamylation, membranes, inhibiting signaling pathways, and repro-
135. Clambey ET, McNamee EN, Westrich JA, et al. an important element in methotrexate cytotoxicity gramming gene expression. EBioMedicine. 2015;2(8):
Hypoxia-inducible factor-1 alpha-dependent induc- and selectivity in tumor versus murine granulocytic 808–824.
tion of FoxP3 drives regulatory T-cell abundance progenitor cells in vitro. Cancer Res. 1984;44(8): 177. Moffett JR, Namboodiri MA. Tryptophan and the
and function during inflammatory hypoxia of the 3190–3195. immune response. Immunol Cell Biol. 2003;81(4):
mucosa. Proc Natl Acad Sci USA. 2012;109(41): 157. Koizumi S, Ueno Y, Ohno I, Ichihara T, Tamaru Y, 247–265.
E2784–E2793. Matsukawa H. Reversal of methotrexate cytotoxicity 178. Prendergast GC, Smith C, Thomas S, et al.
136. Kouidhi S, Noman MZ, Kieda C, Elgaaied AB, to human bone marrow cells and leukemic K562 cells Indoleamine 2,3-dioxygenase pathways of pathogenic
Chouaib S. Intrinsic and tumor microenvironment- by leucovorin: methotrexate polyglutamates forma- inflammation and immune escape in cancer. Cancer
induced metabolism adaptations of t cells and impact tion as a possible important factor. Jpn J Cancer Res. Immunol Immunother. 2014;63(7):721–735.
on their differentiation and function. Front Immunol. 1990;81(11):1162–1167. 179. Uyttenhove C, Pilotte L, Theate I, et al. Evidence
2016;7:114. 158. Pratt CB, Roberts D, Shanks EC, Warmath EL. for a tumoral immune resistance mechanism
137. Ogden CL, Carroll MD, Kit BK, Flegal KM. Preva- Clinical trials and pharmacokinetics of intermit- based on tryptophan degradation by indoleamine
lence of childhood and adult obesity in the United tent high-dose methotrexate—“leucovorin rescue” 2,3-dioxygenase. Nat Med. 2003;9(10):1269–1274.
States, 2011-2012. JAMA. 2014;311(8):806–814. for children with malignant tumors. Cancer Res. 180. Munn DH. Indoleamine 2,3-dioxygenase, Tregs and
138. Bhaskaran K, Douglas I, Forbes H, dos Santos- 1974;34(12):3326–3331. cancer. Curr Med Chem. 2011;18(15):2240–2246.
Silva I, Leon DA, Smeeth L. Body-mass index 159. Howard SC, McCormick J, Pui CH, Buddington 181. Okamoto A, Nikaido T, Ochiai K, et al. Indoleamine
and risk of 22 specific cancers: a population-based RK, Harvey RD. Preventing and managing 2,3-dioxygenase serves as a marker of poor prognosis
cohort study of 5.24 million UK adults. Lancet. toxicities of high-dose methotrexate. Oncologist. in gene expression profiles of serous ovarian cancer
2014;384(9945):755–765. 2016;21(12):1471–1482. cells. Clin Cancer Res. 2005;11(16):6030–6039.
139. Calle EE, Kaaks R. Overweight, obesity and cancer: 160. Gonen N, Assaraf YG. Antifolates in cancer therapy: 182. Hou DY, Muller AJ, Sharma MD, et al. Inhibition
epidemiological evidence and proposed mechanisms. structure, activity and mechanisms of drug resistance. of indoleamine 2,3-dioxygenase in dendritic cells
Nat Rev Cancer. 2004;4(8):579–591. Drug Resist Updat. 2012;15(4):183–210. by stereoisomers of 1-methyl-tryptophan correlates
140. Font-Burgada J, Sun B, Karin M. Obesity and 161. Cohen MH, Justice R, Pazdur R. Approval summary: with antitumor responses. Cancer Res. 2007;67(2):
cancer: the oil that feeds the flame. Cell Metab. pemetrexed in the initial treatment of advanced/ 792–801.
2016;23(1):48–62. metastatic non–small cell lung cancer. Oncologist. 183. Metz R, Duhadaway JB, Kamasani U, Laury-Kleintop
141. Nieman KM, Kenny HA, Penicka CV, et al. 2009;14(9):930–935. L, Muller AJ, Prendergast GC. Novel tryptophan
Adipocytes promote ovarian cancer metastasis and 162. Jarmula A. Antifolate inhibitors of thymidylate catabolic enzyme IDO2 is the preferred biochemical
provide energy for rapid tumor growth. Nat Med. synthase as anticancer drugs. Mini Rev Med Chem. target of the antitumor indoleamine 2,3-dioxygenase
2011;17(11):1498–1503. 2010;10(13):1211–1222. inhibitory compound D-1-methyl-tryptophan.
142. Nieman KM, Romero IL, Van Houten B, 163. Longley DB, Harkin DP, Johnston PG. 5- Cancer Res. 2007;67(15):7082–7087.
Lengyel E. Adipose tissue and adipocytes support Fluorouracil: mechanisms of action and clinical 184. Moon YW, Hajjar J, Hwu P, Naing A. Targeting the
tumorigenesis and metastasis. Biochim Biophys Acta. strategies. Nat Rev Cancer. 2003;3(5):330–338. indoleamine 2,3-dioxygenase pathway in cancer. J
2013;1831(10):1533–1541. 164. Ewald B, Sampath D, Plunkett W. Nucleoside Immunother Cancer. 2015;3:51.
143. White MF, Kahn CR. The insulin signaling system. analogs: molecular mechanisms signaling cell 185. Galluzzi L, Kepp O, Vander Heiden MG, Kroemer
J Biol Chem. 1994;269(1):1–4. death. Oncogene. 2008;27(50):6522–6537. G. Metabolic targets for cancer therapy. Nat Rev
144. Ruderman NB, Kapeller R, White MF, Cantley LC. 165. Elnaggar M, Giovannetti E, Peters GJ. Molecular Drug Discov. 2013;12(11):829–846.
Activation of phosphatidylinositol 3-kinase by insulin. targets of gemcitabine action: rationale for develop- 186. Wagner JP, Seidler FJ, Slotkin TA. Presynaptic input
Proc Natl Acad Sci USA. 1990;87(4):1411–1415. ment of novel drugs and drug combinations. Curr regulates development of beta-adrenergic control of
145. Flannery CA, Saleh FL, Choe GH, et al. Differential Pharm Des. 2012;18(19):2811–2829. rat brain ornithine decarboxylase: effects of 6-hydroxy-
expression of IR-A, IR-B and IGF-1R in endometrial 166. Stresemann C, Lyko F. Modes of action of the dopamine or propranolol. Brain Res Bull. 1991;26(6):
physiology and distinct signature in adenocarcinoma. DNA methyltransferase inhibitors azacytidine and 885–890.
J Clin Endocrinol Metab. 2016;101(7):2883–2891. decitabine. Int J Cancer. 2008;123(1):8–13. 187. Ahmad N, Gilliam AC, Katiyar SK, O’Brien
146. Huang J, Morehouse C, Streicher K, et al. Altered 167. Malik P, Cashen AF. Decitabine in the treatment of TG, Mukhtar H. A definitive role of ornithine
expression of insulin receptor isoforms in breast acute myeloid leukemia in elderly patients. Cancer decarboxylase in photocarcinogenesis. Am J Pathol.
cancer. PLoS ONE. 2011;6(10):e26177. Manag Res. 2014;6:53–61. 2001;159(3):885–892.
147. Jiang L, Zhu W, Streicher K, et al. Increased IR-A/ 168. Abbas S, Lugthart S, Kavelaars FG, et al. Acquired 188. Subhi AL, Diegelman P, Porter CW, et al. Methyl-
IR-B ratio in non–small cell lung cancers associ- mutations in the genes encoding IDH1 and IDH2 thioadenosine phosphorylase regulates ornithine
ates with lower epithelial-mesenchymal transition both are recurrent aberrations in acute myeloid decarboxylase by production of downstream metabo-
signature and longer survival in squamous cell lung leukemia: prevalence and prognostic value. Blood. lites. J Biol Chem. 2003;278(50):49868–49873.
carcinoma. BMC Cancer. 2014;14:131. 2010;116(12):2122–2126. 189. Oredsson SM. Polyamine dependence of normal
148. Renehan AG, Egger M, Minder C, O’Dwyer 169. Ward PS, Patel J, Wise DR, et al. The common cell-cycle progression. Biochem Soc Trans. 2003;31(2):
ST, Shalet SM, Zwahlen M. IGF-I, IGF binding feature of leukemia-associated IDH1 and IDH2 366–370.
protein-3 and breast cancer risk: comparison of 3 mutations is a neomorphic enzyme activity convert- 190. Feith DJ, Bol DK, Carboni JM, et al. Induction of
meta-analyses. Int J Cancer. 2005;115(6):1006–1007, ing alpha-ketoglutarate to 2-hydroxyglutarate. Cancer ornithine decarboxylase activity is a necessary step
author reply 1008. Cell. 2010;17(3):225–234. for mitogen-activated protein kinase kinase-induced
149. Alvino CL, Ong SC, McNeil KA, et al. Understand- 170. Garber K. Cancer anabolic metabolism inhibitors skin tumorigenesis. Cancer Res. 2005;65(2):572–578.
ing the mechanism of insulin and insulin-like growth move into clinic. Nat Biotechnol. 2016;34(8): 191. Lan L, Trempus C, Gilmour SK. Inhibition of
factor (IGF) receptor activation by IGF-II. PLoS 794–795. ornithine decarboxylase (ODC) decreases tumor
ONE. 2011;6(11):e27488. 171. Wang JB, Erickson JW, Fuji R, et al. Targeting vascularization and reverses spontaneous tumors in
150. Pierre-Eugene C, Pagesy P, Nguyen TT, et al. Effect mitochondrial glutaminase activity inhibits onco- ODC/Ras transgenic mice. Cancer Res. 2000;60(20):
of insulin analogues on insulin/IGF1 hybrid recep- genic transformation. Cancer Cell. 2010;18(3):207– 5696–5703.
tors: increased activation by glargine but not by its 219. 192. Wheeler DL, Ness KJ, Oberley TD, Verma AK.
metabolites M1 and M2. PLoS ONE. 2012;7(7): 172. Pinkus LM. Glutamine binding sites. Methods Inhibition of the development of metastatic squa-
e41992. Enzymol. 1977;46:414–427. mous cell carcinoma in protein kinase C epsilon
138.e4 Part I: Science and Clinical Oncology

transgenic mice by alpha-difluoromethylornithine 212. Boudreau DM, Yu O, Johnson J. Statin use and 233. Nebeling LC, Miraldi F, Shurin SB, Lerner E.
accompanied by marked hair follicle degeneration cancer risk: a comprehensive review. Expert Opin Effects of a ketogenic diet on tumor metabolism
and hair loss. Cancer Res. 2003;63(12):3037–3042. Drug Saf. 2010;9(4):603–621. and nutritional status in pediatric oncology patients:
193. Gerner EW, Meyskens FL Jr. Polyamines and cancer: 213. Ahn J, Lim U, Weinstein SJ, et al. Prediagnostic two case reports. J Am Coll Nutr. 1995;14(2):202–
old molecules, new understanding. Nat Rev Cancer. total and high-density lipoprotein cholesterol and 208.
2004;4(10):781–792. risk of cancer. Cancer Epidemiol Biomarkers Prev. 234. Zuccoli G, Marcello N, Pisanello A, et al. Metabolic
194. Bassiri H, Benavides A, Haber M, Gilmour SK, 2009;18(11):2814–2821. management of glioblastoma multiforme using
Norris MD, Hogarty MD. Translational development 214. Jankowski J, Attwood S, deCaestecker J, Barr H. standard therapy together with a restricted ketogenic
of difluoromethylornithine (DFMO) for the treat- Aspirin in the primary prevention of vascular disease. diet: Case Report. Nutr Metab (Lond). 2010;7:33.
ment of neuroblastoma. Transl Pediatr. 2015;4(3): Lancet. 2009;374(9693):877–878, author reply 879. 235. Lee C, Raffaghello L, Brandhorst S, et al. Fasting
226–238. 215. Rothwell PM, Price JF, Fowkes FG, et al. Short-term cycles retard growth of tumors and sensitize a range
195. Schweitzer VG. Ototoxicity of chemotherapeutic effects of daily aspirin on cancer incidence, mortality, of cancer cell types to chemotherapy. Sci Transl Med.
agents. Otolaryngol Clin North Am. 1993;26(5): and non-vascular death: analysis of the time course 2012;4(124):124ra127.
759–789. of risks and benefits in 51 randomised controlled 236. Levine ME, Suarez JA, Brandhorst S, et al. Low
196. Jiao M, Liu G, Xue Y, Ding C. Computational drug trials. Lancet. 2012;379(9826):1602–1612. protein intake is associated with a major reduction
repositioning for cancer therapeutics. Curr Top Med 216. Cao Y, Nishihara R, Wu K, et al. Population-wide in IGF-1, cancer, and overall mortality in the 65
Chem. 2015;15(8):767–775. impact of long-term use of aspirin and the risk for and younger but not older population. Cell Metab.
197. Ashburn TT, Thor KB. Drug repositioning: identify- cancer. JAMA Oncol. 2016;2(6):762–769. 2014;19(3):407–417.
ing and developing new uses for existing drugs. Nat 217. Hawley SA, Fullerton MD, Ross FA, et al. The ancient 237. Khandekar MJ, Cohen P, Spiegelman BM. Molecular
Rev Drug Discov. 2004;3(8):673–683. drug salicylate directly activates AMP-activated mechanisms of cancer development in obesity. Nat
198. Inzucchi SE, Bergenstal RM, Buse JB, et al. Manage- protein kinase. Science. 2012;336(6083):918–922. Rev Cancer. 2011;11(12):886–895.
ment of hyperglycaemia in type 2 diabetes, 2015: 218. Thun MJ, Jacobs EJ, Patrono C. The role of aspirin 238. Courneya KS. Exercise in cancer survivors: an over-
a patient-centred approach. Update to a position in cancer prevention. Nat Rev Clin Oncol. 2012;9(5): view of research. Med Sci Sports Exerc. 2003;35(11):
statement of the American Diabetes Association and 259–267. 1846–1852.
the European Association for the Study of Diabetes. 219. Oshima M, Taketo MM. COX selectivity and 239. Jones LW, Fels DR, West M, et al. Modulation of
Diabetologia. 2015;58(3):429–442. animal models for colon cancer. Curr Pharm Des. circulating angiogenic factors and tumor biology by
199. Madiraju AK, Erion DM, Rahimi Y, et al. Met- 2002;8(12):1021–1034. aerobic training in breast cancer patients receiving
formin suppresses gluconeogenesis by inhibiting 220. Rothwell PM, Wilson M, Price JF, Belch JF, Meade neoadjuvant chemotherapy. Cancer Prev Res (Phila).
mitochondrial glycerophosphate dehydrogenase. TW, Mehta Z. Effect of daily aspirin on risk of cancer 2013;6(9):925–937.
Nature. 2014;510(7506):542–546. metastasis: a study of incident cancers during ran- 240. Betof AS, Lascola CD, Weitzel D, et al. Modulation
200. Matthaei S, Greten H. Evidence that metformin domised controlled trials. Lancet. 2012;379(9826): of murine breast tumor vascularity, hypoxia and
ameliorates cellular insulin-resistance by potentiating 1591–1601. chemotherapeutic response by exercise. J Natl Cancer
insulin-induced translocation of glucose transporters 221. Gay LJ, Felding-Habermann B. Contribution of plate- Inst. 2015;107(5).
to the plasma membrane. Diabete Metab. 1991;17(1 lets to tumour metastasis. Nat Rev Cancer. 2011;11(2): 241. Berger A. How does it work? Positron emission
Pt 2):150–158. 123–134. tomography. BMJ. 2003;326(7404):1449.
201. Pernicova I, Korbonits M. Metformin—mode of 222. Williams JD, Aggarwal A, Swami S, et al. Tumor 242. Yang SK, Cho N, Moon WK. The role of PET/
action and clinical implications for diabetes and autonomous effects of vitamin D deficiency promote CT for evaluating breast cancer. Korean J Radiol.
cancer. Nat Rev Endocrinol. 2014;10(3):143– breast cancer metastasis. Endocrinology. 2016;157(4): 2007;8(5):429–437.
156. 1341–1347. 243. Eubank WB, Mankoff DA. Evolving role of positron
202. Wheaton WW, Weinberg SE, Hamanaka RB, 223. Yun J, Mullarky E, Lu C, et al. Vitamin C selectively emission tomography in breast cancer imaging. Semin
et al. Metformin inhibits mitochondrial complex kills KRAS and BRAF mutant colorectal cancer cells Nucl Med. 2005;35(2):84–99.
I of cancer cells to reduce tumorigenesis. Elife. by targeting GAPDH. Science. 2015;350(6266): 244. D’Souza M, Jaimini A, Bansal A, et al. FDG-PET/CT
2014;3:e02242. 1391–1396. in lymphoma. Indian J Radiol Imaging. 2013;23(4):
203. Zhou G, Myers R, Li Y, et al. Role of AMP-activated 224. Sajadian SO, Tripura C, Samani FS, et al. Vitamin 354–365.
protein kinase in mechanism of metformin action. C enhances epigenetic modifications induced by 5- 245. Nakamoto Y, Saga T, Fujii S. Positron emission
J Clin Invest. 2001;108(8):1167–1174. azacytidine and cell cycle arrest in the hepatocellular tomography application for gynecologic tumors.
204. Xie Y, Wang JL, Ji M, et al. Regulation of insulin-like carcinoma cell lines HLE and Huh7. Clin Epigenetics. Int J Gynecol Cancer. 2005;15(5):701–709.
growth factor signaling by metformin in endometrial 2016;8:46. 246. Ferreira D, Oliveira A, Barroso A, Conde A, Parente
cancer cells. Oncol Lett. 2014;8(5):1993–1999. 225. Liu M, Ohtani H, Zhou W, et al. Vitamin C increases B. Positron emission tomography: indications in lung
205. Birsoy K, Possemato R, Lorbeer FK, et al. Metabolic viral mimicry induced by 5-aza-2′-deoxycytidine. Proc cancer - prospective experience of a department. Rev
determinants of cancer cell sensitivity to glucose Natl Acad Sci USA. 2016;113(37):10238–10244. Port Pneumol. 2007;13(1):35–51.
limitation and biguanides. Nature. 2014;508(7494): 226. High-Dose Vitamin C (PDQ(R)): Health Profes- 247. Bouchelouche K, Oehr P. Positron emission tomogra-
108–112. sional Version. PDQ Cancer Information Summaries. phy and positron emission tomography/computerized
206. Liu X, Romero IL, Litchfield LM, Lengyel E, Bethesda (MD) 2002. tomography of urological malignancies: an update
Locasale JW. Metformin targets central carbon 227. Irigaray P, Newby JA, Clapp R, et al. Lifestyle-related review. J Urol. 2008;179(1):34–45.
metabolism and reveals mitochondrial requirements factors and environmental agents causing cancer: 248. Wu C, Li F, Niu G, Chen X. PET imaging of inflam-
in human cancers. Cell Metab. 2016;24(5):728–739. an overview. Biomed Pharmacother. 2007;61(10): mation biomarkers. Theranostics. 2013;3(7):448–466.
207. Libby G, Donnelly LA, Donnan PT, Alessi DR, 640–658. 249. Liu Y. Fatty acid oxidation is a dominant bioenergetic
Morris AD, Evans JM. New users of metformin 228. Morris AA. Cerebral ketone body metabolism. pathway in prostate cancer. Prostate Cancer Prostatic
are at low risk of incident cancer: a cohort study J Inherit Metab Dis. 2005;28(2):109–121. Dis. 2006;9(3):230–234.
among people with type 2 diabetes. Diabetes Care. 229. Maurer GD, Brucker DP, Bahr O, et al. Differential 250. Oyama N, Akino H, Kanamaru H, et al. 11C-acetate
2009;32(9):1620–1625. utilization of ketone bodies by neurons and glioma PET imaging of prostate cancer. J Nucl Med. 2002;
208. Chae YK, Arya A, Malecek MK, et al. Repurposing cell lines: a rationale for ketogenic diet as experi- 43(2):181–186.
metformin for cancer treatment: current clinical mental glioma therapy. BMC Cancer. 2011;11:315. 251. Farsad M, Schiavina R, Castellucci P, et al. Detection
studies. Oncotarget. 2016;7(26):40767–40780. 230. Veech RL, Chance B, Kashiwaya Y, Lardy HA, Cahill and localization of prostate cancer: correlation of (11)
209. Newby LK, Kristinsson A, Bhapkar MV, et al. Early GF Jr. Ketone bodies, potential therapeutic uses. C-choline PET/CT with histopathologic step-section
statin initiation and outcomes in patients with IUBMB Life. 2001;51(4):241–247. analysis. J Nucl Med. 2005;46(10):1642–1649.
acute coronary syndromes. JAMA. 2002;287(23): 231. Lee C, Longo VD. Fasting vs dietary restriction 252. Janardhan S, Srivani P, Sastry GN. Choline kinase:
3087–3095. in cellular protection and cancer treatment: from an important target for cancer. Curr Med Chem.
210. Nissen SE, Nicholls SJ, Sipahi I, et al. Effect of model organisms to patients. Oncogene. 2011;30(30): 2006;13(10):1169–1186.
very high-intensity statin therapy on regression 3305–3316. 253. Brown JM. Tumor hypoxia in cancer therapy. Methods
of coronary atherosclerosis: the ASTEROID trial. 232. Scheck AC, Abdelwahab MG, Fenton KE, Stafford Enzymol. 2007;435:297–321.
JAMA. 2006;295(13):1556–1565. P. The ketogenic diet for the treatment of glioma: 254. Laking G, Price P. Radionuclide imaging of perfu-
211. Chan KK, Oza AM, Siu LL. The statins as anticancer insights from genetic profiling. Epilepsy Res. 2012; sion and hypoxia. Eur J Nucl Med Mol Imaging.
agents. Clin Cancer Res. 2003;9(1):10–19. 100(3):327–337. 2010;37(suppl 1):S20–S29.
B. GENESIS OF CANCER
Environmental Factors 10 
Steven R. Patierno

S UMMARY OF K EY P OI NT S
Current Concepts in classifications are blurred by include specific forms of arsenic,
Carcinogenesis examples of complex interplay nickel, and hexavalent chromium.
• A key paradigm in environmental among all three types of exposure. • Radiation carcinogens include
carcinogenesis—that of • Carcinogen identification has evolved ultraviolet radiation, ionizing
gene-environment interactions, from historical observational studies, radiation, and radon.
a concept used to describe the to occupational cohort epidemiology, • Fibers such as asbestos and certain
complex interplay between individual to broad-scale screening and testing dusts are etiologic agents in lung
or population genetics and in experimental animals. cancers and mesothelioma.
responses to chemical agents— • Next-generation approaches to • Many components in the diet can
has undergone an expansive carcinogen identification use influence the development of
transformation into a more exposome approaches of integrating cancer through carcinogenic or
contemporary understanding of the sophisticated individual exposure anticarcinogenic mechanisms.
human “exposome.” The exposome monitoring and biosensing with Exposure Biomarkers,
is the cumulative lifelong burden of highly sensitive “omics” approaches Susceptibility Factors, and
disease-contributing stressors, to detect early perturbation of Prevention
including exogenous and disease-relevant signaling pathways. • The identification of molecular
endogenous agents of all types, as • Most human cancers probably result biologic markers of exposure, effect,
well as an individual’s genetic from the interaction of several and susceptibility (reflecting early
susceptibility and “omics” response exogenous carcinogenic influences events that link exposure to adverse
profile (genomics, epigenomics, (often unidentified, but many are outcome, but before detection of
transcriptomics, proteomics, dietary or lifestyle related in origin) clinical disease) will help further our
metabolomics) and their microbiome. along with intrinsic factors (e.g., understanding of human
• A classic sequential model of inherited genes, hormones, carcinogenesis.
initiation, promotion, conversion, and inflammation and oxidative stress, • In addition to genetic polymorphisms
progression has increasingly evolved immune status, energy balance). indicative of susceptibility,
into less linear models that may no Role of Environmental Agents in biomarkers of disease-relevant
longer classify carcinogens as the Etiology of Human Cancer exposure will increasingly rely on
initiators or promoters per se. It may • Although the causes of most highly sensitive, multiplatform, and
be more important to understand the individual human cancers are not multidimensional analysis of the
contribution of environmental agents identifiable, there is incontrovertible impact of potential carcinogenic
to the various hallmarks of cancer: evidence that certain chemical agents on epigenomics,
genomic instability, resisting cell agents, radiation, and certain transcriptomics, proteomics,
death, deregulating cellular biologic agents are contributors to metabolomics, and the microbiome.
energetics, sustaining proliferative the overall incidence of human • Primary, secondary, and tertiary
signals, evading growth suppressors, cancer. cancer prevention approaches will
avoiding immune destruction, • Consumption of tobacco products, be greatly facilitated by the
enabling replicative immortality, especially cigarette smoking, is development of noninvasive
promoting tumor inflammation, responsible for nearly 30% of all biomarkers that identify high-risk
activating invasion and metastasis, cancers. individuals and by identification of
and inducing angiogenesis. • Chemical carcinogens include the disease-consequential
Identification of Human polycyclic aromatic hydrocarbons perturbations in molecular signaling
Carcinogens (PAHs), aromatic amines, benzene, that may also be targets for
• A traditional paradigm classified aflatoxins, tobacco chemicals, and chemoprevention or cancer
carcinogenic agents as chemotherapeutic agents. Metal interception.
environmental, lifestyle related, or carcinogens are largely associated
occupational in origin, but these with occupational exposures and

139
140 Part I: Science and Clinical Oncology

Direct causes of most individual human cancers will not be identifiable, intervention beginning with avoidance of disease-consequential exposure
but there is incontrovertible evidence that certain chemical agents, and chemoprevention (primary prevention), early diagnosis and cancer
radiation, and certain biologic agents are contributors to the overall interception (secondary prevention), and prevention of relapse and
incidence of cancer. A traditional paradigm has classified carcinogenic second cancers (tertiary prevention). It also helps give temporal perspec-
agents as environmental or lifestyle related or occupational in origin, tive to the carcinogenic “lag” period, the presumptive period of time
but these classifications are blurred by examples of complex interplay between an initiation event and clinical detection of a tumor.
among all three “sources” of exposure.1 For example, cigarette smoking A consequence of this sequential model is that it tends to narrowly
is likely responsible for 25% to 30% of human cancer: to smoke or relegate environmental carcinogens as mutagenic “initiating” agents,
not to smoke can be thought of as a lifestyle choice, but the act of and certain other chemical agents as nonmutagenic “promoting” agents,
smoking also gives rise to secondhand (environmental) tobacco smoke and does not account for the increasing weight of evidence that the
exposure, and smoking is pervasive in some occupational settings. process of early carcinogenesis at the cellular level is highly nonlinear
Obesity is now also recognized as a major risk factor for certain cancers.2 and better understood as a perturbation of molecular homeostasis
Obesity is etiologically related to energy balance (lifestyle) and genetics, resulting in accelerated evolution toward acquisition of any of the 10
but certain environmental agents may act as “obesogens” and may hallmarks of cancer.12,13 As shown in Fig. 10.2, these hallmarks are
indirectly contribute to cancer risk. Certain occupational chemical best graphically portrayed in circular fashion because they do not
agents may also be released into the general environment, or unknow- necessarily follow a temporal sequence. This model elaborates our
ingly carried home to affect lifestyle, and occupation itself can be understanding of oncogenesis as a disruption of highly interconnected
considered a lifestyle choice in some parts of the globe. A more recent and complex signaling networks that intersect cytostasis and differentia-
paradigm in environmental carcinogenesis—gene-environment interac- tion circuitry, cell viability circuitry, proliferation circuitry, and motility
tions,3 a concept used to describe the complex interplay between circuitry. It necessitates broadening our understanding of mechanisms
individual or population genetics and responses to chemical agents—has of action in environmental carcinogenesis to include the potential
also undergone an expansive transformation into a more contemporary ability of chemical carcinogens to provoke genomic instability, sustained
understanding of the human “exposome.”4–6 The exposome is the proliferative signaling, acquisition of death resistance, evasion of growth
cumulative lifelong burden of disease-contributing stressors including suppressors, enabling of replicative immortality, and/or activated cellular
exogenous and endogenous agents of all types, as well as an individual’s motility and invasiveness.
“omics” profile (genomics, epigenomics, transcriptomics, proteomics, The “hallmarks of cancer” model also underscores the possible
metabolomics), and his or her microbiome. Yet another traditional impact of environmental agents on deregulation of cellular energetics,
model of environmental carcinogenesis, that of the classic sequential avoidance of immune destruction by emerging tumor cells, promotion
model of initiation, promotion, conversion, and progression (history of tumor inflammation, and provocation of nonneoplastic additional
reviewed succinctly in Weiss7), has also increasingly been supplanted cells in the tumor microenvironment, such as stromal cells, to contribute
by less linear models that no longer classify carcinogens as initiators to the survival, development, and emergence of an early precancerous
or promoters per se. Instead, the concepts of epigenomic and tran- cell. One illustrative example of this complexity emerges from increased
scriptional reprogramming, including alternative RNA splicing, in understanding of the role of cell death in cancer and carcinogene-
response to microenvironmental shifts in allostatic load (cellular sis.10,11,13–15 Studies of genotoxin-treated cells in culture have drawn
physiologic stress) are increasingly understood as drivers of early-stage attention to the importance of an overlooked outcome in earlier
carcinogenesis through the expansion of death-resistant cells.8–11 Thus attempts to measure mutagenesis as a surrogate marker for carcinogenic
a more apt approach may be to understand the contribution of chemical potential: the frequent emergence of death-resistant subclones of cells
agents to the various hallmarks of cancer12,13: genomic instability, out of which an occasional cell carrying a mutated selectable gene
resisting cell death, deregulating cellular energetics, sustaining prolifera- could be detected as a low-frequency event.14,16 This carcinogen-
tive signals, evading growth suppressors, avoiding immune destruction, provoked early acquisition of resistance to apoptosis has challenged
enabling replicative immortality, promoting tumor inflammation, the perspective that active cell death processes necessarily protect against
activating invasion and metastasis, and inducing angiogenesis. These cancer and carcinogenesis. To the contrary, the facile ability of cells
evolving new thought paradigms hold promise for improved exposure to circumvent cell death when under stress actually renders the existence
monitoring, more accurate identification and earlier detection of of such cell death signaling pathways as a risk factor for carcinogenesis.15
disease-consequential exposures, and increasingly effective primary, Indeed, it is now appreciated that additional modes of cell death,
secondary, and tertiary public health and clinical cancer prevention such as autophagy, can mediate either tumor cell survival or death;
and interception measures. and even necrosis, often a consequence of toxic exposure, has been
shown to be potentially proinflammatory and tumor promoting.15,17
EVOLVING MODELS FOR CHEMICAL In 2015 a series of review articles were published by an international
CARCINOGENESIS consortium of scientists, collectively called the Halifax Project, which
conducted detailed assessments of the potential of environmental
Fig. 10.1 gives a linear representation of the traditional paradigm for chemicals to contribute causatively to the process of carcinogenesis
progressive carcinogenesis. This model is useful in that it depicts what by affecting each “hallmark” of cancer.18–25
is thought to be the monoclonal nature of cancer (originating from
a single cell) and the concept of clonal evolution and expansion. It
evokes a chemical- or radiation-provoked initiating event, followed HISTORY OF ENVIRONMENTAL
by promotion of clonal expansion to form a preneoplastic lesion. In CARCINOGENESIS AND SUPPORT FOR
this model, it is thought that a single cell of a preneoplastic lesion THE ROLE OF ENVIRONMENTAL AGENTS IN
undergoes “conversion,” which theoretically equates to progeny cells THE ETIOLOGY OF HUMAN CANCERS
acquiring enough cumulative genotypic and phenotypic alterations
to express a malignant phenotype. Malignant cells, defined pathologi- The methods and concepts for identifying chemical agents potentially
cally as “invasive,” can then undergo progression (volume expansion carcinogenic to humans have evolved over time and continue to
due to increased net cell proliferation) to a clinically relevant cancer evolve as both the general public and regulatory agencies grapple
with metastatic potential. This pictorial sequence is often punctuated with the fact that humans live in a chemical-laden society, which
with reference to mutagenic activation of oncogenes, inactivation of generally contributes to improved global health and increased longev-
tumor suppressor genes, and, more recently, epigenetic alterations. ity. In addition to consumption of and exposure to tobacco smoke,
Such a linear model is particularly illustrative of protective points of itself containing more than 30 potentially carcinogenic chemicals26–28
Environmental Factors  •  CHAPTER 10 141

Sequence Leading to Neoplasia Interventions and Strategies

Viruses
Radiation
Chemicals
Procarcinogens Direct carcinogens Avoid Primary
exposure prevention

Initiation

Ultimate carcinogens Prevent


formation of
Genetic and
carcinogens
epigenetic
changes

Initiated Block
Chemo
cell interactions
prevention
with genome
Selective
Promotion clonal
expansion
Preneoplastic Suppress
lesion growth

Genetic and
epigenetic
Conversion changes
Early Secondary
diagnosis prevention
Malignant
tumor

Surgery
Radiation
Cell Therapy
therapy
heterogeneity Chemotherapy

Progression
Prevent relapse
Clinical Tertiary
and secondary
cancer prevention
tumors

Figure 10.1  •  Linear representation of a sequence leading to neoplasia and points of public health intervention strategies for prevention of multistage
carcinogenesis.

(Box 10.1) delivered directly by inhalation of a chronically injurious The influences of occupation and lifestyle in cancer occurrence trace
toxic fume, humans are awash in a sea of low-level exposure to both back at least to the 16th century when Ramazzini, in 1700, noted
natural and anthropomorphic chemicals. For example, polycyclic that nuns showed a higher frequency of breast cancer than was observed
aromatic hydrocarbons (PAHs), a broad class of chemicals including among other women. Also in that century, Paracelsus and Agricola
some with carcinogenic potential, are released into the environment described Bergkrankheiten (lung cancer) in German miners, probably
by both anthropomorphic fuel combustion and natural volcanic caused by uranium and its decay product radon. In 1761, Hill
eruptions and forest fires, as well as consumed through smoking observationally associated the use of tobacco snuff with cancer in the
and eating flame-cooked meat. Such exposure complexity neces- nasal passage, and in 1775 Pott noted the occurrence of soot-related
sitates improved differentiation of “any” exposure from “disease- scrotal cancer among chimney sweeps. In 1895, Rehn published
consequential” exposures, which itself represents a next-generation evidence that occupational exposure to aromatic amines was associated
evolution of carcinogen identification from historical observational with bladder cancer, and Unna in 1894 associated sunlight exposure
studies, to occupational cohort epidemiology, to broad-scale screen- with skin cancer.29–34
ing and testing in experimental animals, to exposome approaches of These observational associations gradually gave way to more proac-
integrating sophisticated individual exposure monitoring with highly tive approaches to attempt to identify potential carcinogens both
sensitive “omics” approaches to detect perturbation of disease-relevant computationally (retrospective epidemiology, usually of occupationally
signaling pathways. exposed or catastrophe-exposed cohorts) and by animal experimentation.
The carcinogenic effects of a sizable number of environmental or However, it was not until the early 20th century that animal models
industrial chemicals were first described observationally in humans. for chemical carcinogenesis were developed, beginning in 1915 when
142 Part I: Science and Clinical Oncology

Box 10.1.  HEALTH EFFECTS AND CONTROL OF


SMOKING
A series of reports from the US Surgeon General since 1964 have
argued that cigarette smoking is the most significant source of
preventable morbidity and premature mortality in high-income
countries. An estimated annual excess mortality of 400,000 is attributed
to cigarette smoking in the United States. These deaths are the result of
coronary heart disease, cancer, and various respiratory diseases.40
Cancers associated with smoking or smokeless tobacco use include
those of the lung, oral cavity, esophagus, pharynx, larynx, and bladder.
Additional associations have been reported for cancers of the pancreas,
kidney, stomach, nasopharynx, and cervix.12 The overall increase in risk
of disease among smokers compared with nonsmokers is about
tenfold for lung cancer, sixfold for chronic obstructive pulmonary
disease, and twofold for myocardial infarction. The combined effect of
smoking-related diseases on the average life expectancy of smokers is
a reduction of 5 to 8 years. On a worldwide basis, an estimated 6
million deaths per year are attributed to tobacco use, with upward of
80% of these deaths in low- and middle-income countries. If current
use trends continue unabated, estimates as high as 8 million deaths
due to tobacco use annually could be expected by the year 2030, and a
total of 1 billion this century.41
The addictive properties of tobacco smoking, due primarily to its
nicotine content, cause both physiologic and psychologic dependence.
Figure 10.2  •  Hallmarks of cancer and points of potential carcinogen Withdrawal symptoms can be severe and include irritability,
impact. aggressiveness, hostility, depression, difficulty concentrating, and a
craving for tobacco. These pharmacologic factors are reinforced by
social factors such as peer pressure, emulation of family role models,
and cultural influences. Because most smokers begin smoking, and
Yamagiwa and Ichikawa demonstrated the production of skin tumors often form lifelong smoking habits, in their teenage years, preventive
after topical application of crude coal tar to the ears of rabbits.35 These educational measures should be focused on this age group.
early studies gradually led to the standardization of animal carcinogenesis Smoking control measures use various strategies: cessation
bioassays, with the goal of testing chemical class prototypes for car- programs, clinical or community interventions, governmental or
cinogenic potential, ostensibly before human exposure. private-sector regulations, taxation of tobacco products, warning labels,
Contrary to experiences in earlier centuries, with the advent of and smoking prevention programs. Although the majority of
animal bioassay programs, evidence of carcinogenicity in experimental ex-smokers have achieved abstinence without extensive personal
animals has sometimes preceded evidence obtained from epidemiologic assistance from organized cessation programs, other control measures
studies or case reports. Although the term carcinogen means “giving (e.g., national health education programs, physician counseling, and
rise to carcinomas,” loosely referring to malignancies in general, more indoor smoking regulations) and family pressure are important
operational definitions are used for carcinogens in animal bioassays. influences contributing to smoking cessation. Smoking prevention
In this context, a carcinogen may be defined as an agent whose programs, particularly in schools, and government taxation have been
administration to previously untreated cells or animals leads to a statisti- somewhat effective in reducing the initiation of smoking among
cally significant increased incidence of malignant neoplasms, compared children and adolescents in developing countries. These approaches
with the incidence in appropriate untreated control cells or animals. will need to be applied more vigorously, especially in low- to middle-
Data from such studies, combined with epidemiologic and mechanistic income countries, to stem the expansion of smoking worldwide. New
studies, are employed by regulatory agencies to try to protect humans challenges also are arising with the creation of new and emerging
tobacco products such as dissolvable tobacco products, e-cigarettes,
from known chemical, radiation, and infectious hazards. From a
and hookah tobacco. The Family Smoking Prevention and Tobacco
mechanism of action (MOA) perspective, the regulatory thrust has
Control Act (2009) in the United States has given the US Food and
been to determine whether a carcinogenic agent was mutagenic or
Drug Administration broad authority to regulate the manufacturing,
nonmutagenic; the latter involved recognition that chronic tissue
distribution, and marketing of these and more traditional tobacco
toxicity itself, in the absence of DNA damage, was potentially carci-
products to protect public health.
nogenic. Synthetic and naturally occurring chemicals comprise the
largest group of known human carcinogens. More than 100 chemicals,
chemical mixtures, biologic agents, physical agents, or industrial
processes have been classified as Class 1 human carcinogens (Table
10.1) by the International Agency for Research on Cancer (IARC), underpinning the role of the environment in cancer risk relates to the
and more than 300 chemicals have been designated as probable (Class impact of geography on lifestyle adoption and adaptation and cancer
2A) or possible (Class 2B) human carcinogens.36 For perspective, these susceptibility, and derives from the following series of epidemiologic
numbers of potential carcinogenic hazards derive from an environmental observations:
milieu of nearly 10 million chemicals.
Substantial growth has occurred in our understanding of how these • Although the overall incidence of cancer development is reasonably
extrinsic factors (chemicals, radiation, infectious agents) interact with constant among countries, incidences of specific cancer types can
intrinsic factors (e.g., genetics of inherited genes, “omics” profile, diet, vary up to several hundredfold.
body mass index (BMI), hormones, immune status, and microbiome) • Large differences in tumor incidence exist within populations of a
to determine overall susceptibility and risk. Indeed, a key concept single country.
Environmental Factors  •  CHAPTER 10 143

Table 10.1  Agents and Processes Considered Carcinogenic in Humans (Class 1) by the International
Agency for Research on Cancer
Common Organ or Tissue Common Organ or Tissue
Agent or Process Sites of Cancer Agent or Process Sites of Cancer
AMBIENT AND DIETARY EXPOSURE Mineral oils (untreated and Skin, scrotum
Acetaldehyde (from alcoholic Upper digestive tract mildly treated)
beverages) Mustard gas Lung, larynx and pharynx
Aflatoxins Liver 2-Naphthylamine Urinary bladder
Areca nut Oral cavity Nickel and nickel compounds Lung, nasal sinus
Aristolochic acid (from plants) Renal pelvis and ureter Painting Lung
Arsenic and arsenic compounds Lung, skin 2,3,4,7,8-Pentachlorodibenzofuran Soft tissue sarcoma, non-Hodgkin
Erionite Pleura, peritoneum lymphoma, cancer of the lung
Polychlorinated biphenyl (PCB-126) Liver, bile duct, lymphoid tissue
CULTURAL HABITS
Rubber industry Urinary bladder, leukemia
Alcoholic beverages Oral cavity, pharynx, larynx,
Shale oils Skin, scrotum
esophagus, liver
Silica, crystalline Lung
Betel quid with tobacco Oral cavity
Soots Skin, scrotum, lung
Tobacco products, smokeless Oral cavity
Strong inorganic acid mists Larynx
Tobacco smoke Respiratory tract, urinary bladder,
renal pelvis, pancreas Talc containing asbestiform fibers Lung
Salted fish, Chinese style Nasopharynx Toluidine Urinary bladder
Solar radiation Skin Underground mining with Lung
exposure to radon
OCCUPATIONAL EXPOSURES Vinyl chloride Liver, lung, gastrointestinal tract,
Aluminum production Lung, urinary bladder brain
4-Aminobiphenyl Urinary bladder Wood dust Nasal cavities, paranasal sinuses
Asbestos Lung, pleura, peritoneum, larynx, THERAPEUTIC AGENTS
gastrointestinal tract
Analgesics mixtures containing Renal, urinary bladder
Auramine production Urinary bladder
phenacetin
Benzene Leukemia
Azathioprine Leukemia
Benzidine Urinary bladder
N,N-bis(2-chloroethyl)-2- Urinary bladder
Benzo(a)pyrene Lung naphthylamine
Beryllium Lung Busulphan Lymphohematopoietic tissue
Boot and shoe manufacture Nasal sinus 1,4-Butanediol dimethanesulfonate Leukemia
and repair
Chlorambucil Leukemia
Cadmium Lung
Chlornaphazine Urinary bladder
Chromium (VI) compounds Lung
Cyclophosphamide Urinary bladder, leukemia
Coal combustion (indoors) Lung
Cyclosporin Lymphoma
Coal gasification Lung, urinary bladder, scrotum
Diethylstilbestrol Breast, cervix
Coal-tar pitches Skin, scrotum, lung
Estrogen replacement therapy Endometrium, breast
Coal tars Skin, lung
Estrogens, nonsteroidal Cervix and vagina, breast,
Coke production Skin, scrotum, lung, urinary endometrium, testes
bladder
Estrogens, steroidal Endometrium, breast
Dioxin Multiple sites
Etoposide Lymphohematopoietic tissue
Ethylene oxide Lymphatic, hematopoietic
Melphalan Leukemia
Fission products Multiple sites
8-Methoxypsoralen plus UV Skin
Formaldehyde Liver radiation
Furniture and cabinet making Nasal sinus MOCA (Methylene Urinary bladder
Hematite mining Lung bis[2-chloroaniline])
Ionizing radiation (all types) Multiple sites MOPP combination therapy Leukemia
Iron and steel founding Lung Oral contraceptives (combined) Liver
Isopropyl alcohol manufacture Nasal sinus Oral contraceptives (sequential) Endometrium
(strong acid process) Phenacetin Renal pelvis, ureter
Leather dust Nasal sinus Semustine (methyl-CCNU) Leukemia
Magenta, manufacture of Urinary bladder Sulfur mustard Lung
Methyl ether Lung Tamoxifen Endometrium

Continued
144 Part I: Science and Clinical Oncology

Table 10.1  Agents and Processes Considered Carcinogenic in Humans (Class 1) by the International
Agency for Research on Cancer—cont’d
Common Organ or Tissue Common Organ or Tissue
Agent or Process Sites of Cancer Agent or Process Sites of Cancer
Thiotepa Leukemia Human immunodeficiency Kaposi sarcoma
Treosulfan Leukemia virus type 1
INFECTIOUS AGENTS Human papillomavirus types Cervix
16, 18, others
Clonorchis sinensis Liver, bile duct
Human T-cell lymphotropic Adult T-cell leukemia/lymphoma
Epstein-Barr virus Lymphoma
virus type I
Helicobacter pylori Stomach
Kaposi sarcoma herpesvirus Kaposi sarcoma
Hepatitis B virus Liver
Opisthorchis viverrini Liver (cholangiocarcinoma)
Hepatitis C virus Liver
Schistosoma haematobium Urinary bladder

Data from the International Agency for Research on Cancer (IARC). IARC Monographs on the Evaluation of Carcinogenic Risk to Humans. Vols. 1–104. Lyon, France: IARC;
1970–2012. An updated listing of the overall evaluation of carcinogenicity to humans can be accessed at http://monographs.iarc.fr under the heading “Classifications.” (Note: For
examples of carcinogenic ionizing radiations, see Table 10.3.)

• Migrant populations assume the cancer incidence of their new Chapter 11). These must be overcome before toxic and carcinogenic
environment within one to two generations. properties are manifest, underscoring the importance of dose to
• Cancer rates within a population can change rapidly.37 toxicology.
Once formed intracellularly, the reactive intermediates can inter-
It should be noted, however, that a major aspect of geography- act with both protective and vulnerable molecular targets related
influenced carcinogenesis is related to exposure to unique dietary and to carcinogenesis. As described earlier, in classic linear models of
lifestyle behaviors and infectious agents, and the possible additive or carcinogenesis, mutagenicity was considered a necessary property
synergistic interactions between infectious agents and diet. For example, of carcinogenic “initiators.” DNA was considered the principal
the incidence of cancer of the esophagus, the sixth most common molecular target of carcinogens, and it was presumed that nearly
cause of death from cancer worldwide30 but the third most common all carcinogens interacted with DNA to produce genetic lesions
in developing countries after stomach and lung cancer, varies from (DNA damage) that can result in mutation of critical cellular genes,
less than 5 per 100,000 persons per year in Central America and including oncogenes and tumor suppressor genes. More recently, the
Western Africa to more than 50 per 100,000 persons per year in parts potential interactions of reactive chemicals with signaling pathways
of China and central Asia. Putative causes for such “hot spots” have governing the various hallmarks of cancer, and the cell’s molecular
been identified, such as consumption of hot yerba maté in Uruguay, responses to those interactions, have come to be recognized as part
Brazil, and northeast Argentina; however, the causative factors in other of the early carcinogenic process. One such example is the potential
high-risk areas remain unknown. Other cancers demonstrating unique connection between compounds known as environmental estrogens,
geographic distribution include biliary cancer (cholangiocarcinoma) chronic receptor-mediated growth signaling, and hormonally related
associated with liver fluke infection in some Asian (especially Thailand) cancers42–44 (Box 10.2). Other critical examples, discussed later, include
and African countries; gastric adenocarcinoma associated with virulent the nonmutagenic impact of chemically reactive species on gene
strains of Helicobacter pylori infection in South Korea, Japan, and regulation through epigenomic, metabolomic, and/or transcriptional
parts of China; and hepatocellular carcinoma (HCC) associated with reprogramming.
hepatitis B virus (HBV) and aflatoxin exposure in parts of western
Africa and eastern China.38–40 Other connections between geography EXPOSURE BIOMARKERS AND ASSESSING
and lifestyle/environment are documented by studies of shifting cancer HUMAN EXPOSURE
organ-site rates over generations following immigration, such as the
decrease in stomach cancer rates and increase in prostate cancer rates Increased understanding of the mechanistic basis of carcinogenesis
in offspring of Japanese immigrants to North America.41 provides opportunities for the identification of molecular biologic
markers that reflect both disease-relevant exposure and/or events
Chemical Biology of Carcinogenesis occurring between exposure and clinical disease. It is hoped that the
use of biologic markers will help define the roles of environmental
Chemical carcinogens comprise a diverse array of chemical structures, agents (particularly at low levels and in complex mixtures) in the
including both organic and inorganic compounds, the one common etiology of human cancer. These molecular biologic markers can be
characteristic being that either the chemical itself, or its derivatives, classified into three major categories:
are reactive species capable of interacting with biomolecules. Because
the innate reactivity of such compounds also tends to make them 1. Markers of exposure reflecting a biologically effective dose of a
unstable, relatively few carcinogens are direct acting. Instead, most carcinogen
carcinogens require metabolic activation to reactive species, often inside 2. Markers of effect indicating a disease-relevant biologic response to
of the target cells. Metabolic pathways can be strongly influenced by an exposure
a variety of extrinsic and intrinsic factors and are important determinants 3. Markers of susceptibility that characterize the inherent susceptibility
of both interindividual and target organ susceptibilities to carcinogens. of an individual to a carcinogenic agent45–49
Because the mammalian species, including humans, developed in a
complex milieu of naturally occurring toxic agents, a panoply of innate The detectable persistent interaction of a carcinogen with macro-
mechanisms of extracellular and intracellular resistance to such toxic molecules was first demonstrated by Miller and Miller in 1947, who
insults developed, ranging from circulating and intracellular sulfhydryl- showed azo dye residues bound to liver proteins of treated rats. Later
rich peptides and antioxidants to extensive DNA repair pathways (see studies indicated the importance of carcinogen modification of DNA
Environmental Factors  •  CHAPTER 10 145

and several other aflatoxin metabolites in urine were shown to be


Box 10.2.  ENVIRONMENTAL ESTROGENS AND predictive of the development of liver cancer.
BREAST CANCER Carcinogen-protein adducts have also been studied as biomarkers
of human exposure. Alkylation damage in hemoglobin has been used
Women with a high lifetime exposure to estrogen, such as that as an exposure index in risk assessment analyses of persons occupation-
occurring with early onset of menarche and late menopause, may be at ally exposed to ethylene oxide. It has been found that 4-aminobiphenyl
higher risk for breast cancer. In obese menopausal women, adipose adducts in hemoglobin are highly specific (and sensitive) markers of
tissue becomes the major source of estrogens, and higher blood exposure to cigarette smoke. The level of 4-aminobiphenyl hemoglobin
estrogen levels among postmenopausal women have shown that adducts is related to the number of cigarettes smoked, the type of
higher levels are associated with increased risk of subsequent breast tobacco smoked, and the metabolic phenotype of the smoker. The
cancer.42 These observations provoke an important question: do levels of these adducts drop markedly after smoking cessation.37
xenoestrogens (synthetic chemicals that can act like estrogens) or Unreacted carcinogen metabolites excreted in urine also are
other environmental factors such as phytoestrogens (plant estrogens) used to monitor human exposure to and uptake of carcinogens and
play a role in the risk for breast cancer? There is clear evidence that related compounds. Concentrations of hydroxylated PAHs (e.g.,
these molecules may interfere with the interactions of human 1-hydroxypyrene) in urine are elevated in smokers, in patients after
estrogens with their receptors, and perturb signaling pathways that not topical treatment with coal tar, in road pavers, in coke oven workers,
only promote proliferative signaling, but possibly even early resistance in aluminum plant workers, and in persons ingesting PAHs from food.
to antiestrogens.63 However, thus far no epidemiologic “smoking gun” In occupational settings, the concentration of urinary 1-hydroxypyrene
has been identified. The Long Island Breast Cancer Study was unable to is highly correlated with estimated or measured concentrations of
identify any environmental factors that could be responsible for the airborne PAHs.
elevated incidence of breast cancer on Long Island or other locations43 These noninvasive approaches to exposure assessment have potential
and no evidence emerged for organochlorine pesticides, which have applications as biomonitoring tools, but at present they have major
long been known to have weak estrogenic activities. Bisphenol A, an limitations, and by themselves do not address the key question of
industrial chemical present in some polycarbonate plastics used in parsing out disease-relevant exposure. In all of these studies, significant
bottles and infant food packaging, has come under recent scrutiny as a differences in adduct or metabolite levels are often observed between
xenoestrogen.44 Consumer advocacy and industry action rather than persons with similar carcinogen exposure, likely due to individual
regulatory mandates seem to be reducing exposures to bisphenol biologic variability, exposure misclassification, and confounding variables
A at this time. Some studies have shown that women who have diets
such as diet, physical activity, smoking, or personal environment. It
high in phytoestrogens have lower rates of breast cancer. These
is hoped that the exposome approach, at the intersection between
phytoestrogens may actually oppose the actions of natural estrogen
high-sensitivity biomarker detection and perturbation of disease-relevant
and sometimes lead to lower serum estrogen levels. The role of early
signaling pathways, may improve capabilities for detection of meaningful
life exposures as risk factors for late-in-life events, potentially via
exposures as a function of individual susceptibility.
epigenomic effects, is emerging as a key area of investigation in
environmental carcinogenesis.42
Carcin-Omics and the Exposome Approach to
Environmental Carcinogenesis
The concept of the cumulative lifelong human exposome is the most
recent integrative approach to understanding the complex interplay
(either directly or after metabolic activation) in the cancer process. among natural and anthropogenic chemicals, the environment-lifestyle-
The measurement of carcinogen metabolites, carcinogen-DNA adducts, occupational continuum of exposures, and the response of host cells
or carcinogen-protein adducts in human tissues or fluids provides the to exposure.4–6 This includes the intersection with diet and energy
basis for development and application of individual exposure monitor- balance, the interplay between extrinsic and intrinsic exposures in the
ing, molecular dosimetry research, and the rapidly expanding field of context of cellular physiologic stress (illustrated by the conundrum of
“molecular” cancer epidemiology. The potential advantage of this the necessity of oxygen for life in juxtaposition with the toxic conse-
approach is that more accurate assessments of individual or group quences of oxidative stress), and the concept of disease-consequential
dose may be achieved than through estimates of carcinogen exposure exposure, especially in the context of dose. The importance of dose
in the environment or workplace. is critical, a steady holdover from the 16th century alchemist and
Carcinogen-DNA and carcinogen-protein adducts have been father of toxicology, Paracelsus: “All things are poison and there is
detected in tissues from a variety of human populations with known nothing without poison; the right dose differentiates a poison from
or suspected carcinogen exposure. PAH-DNA adducts are elevated a remedy” or “the dose makes the poison.” Herein lies the central
in white blood cells from persons occupationally exposed to airborne conceptual challenge of exposome research: to drive innovation in
PAHs, including coke oven workers, foundry workers, and aluminum increasingly sensitive methods to monitor and quantify individual
plant workers, and in lung tissue from heavy smokers. DNA isolated environmental and internal human exposures, and to combine those
from exfoliated bladder epithelial cells of cigarette smokers has been data with leading-edge methodologies to detect subclinical perturba-
shown to contain 4-aminobiphenyl adducts. Alkylation damage is tions in disease-relevant signaling pathways that directly contribute to
detected in DNA from esophageal tissue of persons with documented disease evolution.
dietary exposure to nitrosamines. Aflatoxin adducts are found in hepatic In traditional risk assessment paradigms, exposure science focused
DNA after dietary exposure to this mycotoxin. Cisplatin-DNA on providing an exposure estimate that would then be benchmarked
intrastrand adducts have been measured in white blood cells of patients against algorithm-driven regulatory guidance values in order to
undergoing cancer chemotherapy. maximally protect public health. More recently, exposure science has
Carcinogen-DNA adducts excreted in urine provide a potentially shifted toward development and application of advanced analytic
noninvasive means for quantifying DNA damage. Urinary concentration methods including remote and on-person sensors, mechanistic and
of aflatoxin-guanine adducts is strongly correlated with dietary aflatoxin computational exposure biology, and advanced “systems-omics” analysis
intake. In addition to their use as indicators of previous carcinogen as next-generation biomarkers for meaningful exposure. These
exposure, DNA adducts have the potential for use as direct indicators approaches may revolutionize conventional testing strategies and risk
of future cancer risk. The first prospective test of this application assessment, particularly as larger numbers of chemicals are being
appeared in 1992, when detectable levels of aflatoxin-guanine adducts detected in biologic samples at lower and lower levels. Systems level
146 Part I: Science and Clinical Oncology

“omics” approaches to biomarkers are shedding light on early biologic growth factor receptor (EGFR) pathway.63 Likewise, exposure of the
effects of external and intrinsic stressors on adverse health outcomes, developing prostate to endocrine-disrupting chemicals can increase
including tobacco, diet, occupational exposures, and environmental risk of adult prostate cancer, and this has been associated with epig-
pollutants. As described by Escher and colleagues,50 “a mechanistic enomic disruption via phosphatidylinositol 3-kinase (PI3-K) effects
understanding of the causal links between exposure and adverse effects on histone methyltransferase.55
on human health and the environment can be improved by integrating A complex interplay between genetic (mutational) and epigenetic
the exposome approach with the adverse outcome pathway (AOP) contributions to carcinogenesis is highlighted by multidimensional
concept that structures and organizes the sequence of biologic events analysis of the genome, epigenome, and transcriptome of glioblastoma.53
from an initial molecular interaction of a chemical with a biologic Mutated EGFR appears to promote epigenomic remodeling and
target to an adverse outcome. Complementing exposome research transcriptome reprogramming in EGFR-dependent glioblastoma
with the AOP concept may facilitate a mechanistic understanding of multiforme (GBM) pathogenesis. Interesting to note, the MOA of
stress-induced adverse effects, examine the relative contributions from carcinogenic forms of arsenic and nickel seems to involve epigenetic
various components of the exposome, determine the primary risk modification of chromatin structure and function,58,59 in addition to
drivers in complex mixtures, and promote an integrative assessment genotoxic and actions. Epigenetic dysregulation through changes in
of chemical risks for both human and environmental health.” Similarly, the methylome and histone modification has also been observed in
additional model constructs for understanding the potential impact virus-associated neoplasms.54
of environmental carcinogens point out that different chemicals in Important to note, the epigenome is also a potential target for
chemical mixtures might affect different target cells (somatic or stem chemoprevention; for example, inhibitors of bromodomain proteins
cell) with different but complementary procarcinogenic effects on that serve as epigenetic “readers” inhibit in vitro neoplastic transforma-
signaling pathways governing the nexus between cell senescence and tion by 12-O-tetradecanoylphorbol-13-acetate (TPA), ostensibly by
immortality.18 inhibiting TPA-induced expression of prosurvival genes.57 Likewise,
Some of these concepts were also recently highlighted by a Working a number of anticarcinogenic phytochemicals have a profound
Group convened by the National Institute of Environmental Health effect on DNA methylation, histone modification, and regulation of
Sciences (NIEHS) to give guidance on addressing the challenges of noncoding RNAs.60
“low-dose” carcinogenesis.51 The Working Group concluded, “The
scientific community will need to acknowledge limitations of animal- Transcriptomic Exposure Profiling
based models in predicting human responses; evaluate biologic events Next-generation gene-chip and deep RNA sequencing technologies,
leading to carcinogenesis both spatially and temporally; examine the deployed against both in vivo and in vitro exposures for cross-platform
overlap between measurable cancer hallmarks and characteristics of concordance, are providing insights into early molecular changes
carcinogens; incorporate epigenetic biomarkers, in silico modelling, induced by carcinogens, which could contribute to the development
high-performance computing and high-resolution imaging, microbiome, of biomarkers for detection of disease-relevant exposure, early disease
metabolomics, and transcriptomics into future research efforts; and detection, and prevention.64–66 For example, (S)-N′-nitrosonornicotine
build molecular annotations of network perturbations. The restructuring ([S]-NNN), the major form of NNN in tobacco products, is an oral
of many existing regulatory frameworks will require adequate testing cavity and esophageal carcinogen in rats. Differences in the expression
of relevant environmental mixtures to build a critical mass of evidence of 40 to 70 genes involved in immune regulation, inflammation, and
on which to base policy decisions.” cancer were detected in treated rats and cultured oral keratinocytes,
and interrogation of TCGA data sets showed that several of the genes
Epigenomic Exposure Profiling that were deregulated by (S)-NNN in rat tissues are also altered in
The epigenome is a principal determinant of cellular transcriptional esophageal and head and neck tumors. Likewise, changes in global
programming, and perturbations in the epigenomic are increasingly gene expression were detected, after carcinogen treatment and before
understood as instrumental in cancer development.52–60 In general, the appearance of squamous cell carcinoma tumors, in a mouse
“epigenetics” refers to heritable changes in gene expression that are tongue model of human oral cavity and esophageal squamous cell
not caused by alterations in the primary DNA sequence but are carcinoma through use of the carcinogen 4-nitroquinoline 1-oxide
associated with altered DNA methylation, multiple histone modifica- (4-NQO). Gene ontology and pathway analyses have shown that
tions, and tight regulation of multiple species of noncoding RNAs. transcriptional networks affecting cell cycle progression, migration
These factors, functionally characterized as epigenetic chromatin writers, and invasion, and cellular energetics were early events after expo-
readers, or erasers, govern the dynamics of chromatin conformation sure and that changes in these pathways were also present in the
and transcription, and their complex net function can be perturbed actual tumors.
by both genotoxic and nongenotoxin carcinogens. A number of studies Another study illustrates the potential application of transcriptomics
have identified carcinogen-induced, rapid alterations in epigenomic to parse out the individual effects of constituents of complex mixtures,
“marks,” some of which are also present in tumors from that tissue, or even metabolites of the same parent carcinogen. Benzo(a)pyrene
and smoking-associated DNA methylation changes in buccal cells (BaP) is produced from incomplete combustion of organic compounds
have been associated with DNA methylation changes in smoking-related and also is present in cigarette smoke. The transcriptomic biologic
epithelial cancers. effects of BaP, its genotoxic metabolite BPDE, and an array of intermedi-
Studies have shown that environmental estrogens can disrupt ary metabolites were studied in Hep2 cells, revealing activation, at
estrogen receptor (ER) signaling, alter DNA and histone methylation, very early exposure time points, of several transcription factor networks
and reprogram the epigenome.61,62 Some of this epigenomic reprogram- affecting metabolism, oxidative stress, cell proliferation, cell energetics,
ming may occur as a result of adverse exposure to environmental DNA repair, and apoptotic pathways. These gene expression pattern
estrogens during development, and possible disruption of normal changes were compared with a signature set of approximately 9000
mammary gland development, resulting in changes in disease susceptibil- gene expressions derived from HCC patients. This approach showed
ity across the life course. The mechanism of disruption can involve that exposure of cells to BaP, but not TCDD, deregulated metastatic
perturbation of genomic signaling through altered binding of ligand- markers via nongenotoxic and genotoxic mechanisms, activated
associated ER with its DNA binding site, and/or nongenomic signaling inflammatory pathways, and repressed expression of genes involved
of membrane-bound ER resulting in activation of growth-regulating in cholesterol and fatty acid biosynthesis. Other studies on critical
or growth-deregulating kinase cascades. A study suggested that RNA regulatory functions have suggested that disruption of cancer-
environmental estrogens such as bisphenol A activates growth-promoting related microRNA-mRNA pairings may be an early sentinel of
proliferation and resistance to therapeutic drugs that target the epidermal disease-related exposure. Taken together, such studies suggest that a
Environmental Factors  •  CHAPTER 10 147

transcriptomics approach may identify pathways that change as a tar were PAHs,31 with most of the carcinogenic activity attributed to
result of exposure, which may also contribute to the evolution of key the PAH BaP.
hallmarks of cancer. Humans are exposed to PAHs from a variety of sources that include
occupation, smoking, diet, and air.32 PAHs are readily absorbed into
Metabolomic Exposure Profiling the body through the skin, lungs, and gastrointestinal tract. Occupa-
Metabolite profiling is being increasingly employed in the study of tional and medicinal exposures constitute the highest levels of human
carcinogenesis as a systems-level approach and means of identifying PAH exposure (small groups within the population), whereas diet
biomarkers for exposure that are predictive of risk and diagnostic of and smoking are the major sources of exposure to PAHs in the general
meaningful, disease-relevant exposure.67–71 This includes metabolomic population. An excess of lung cancer has been demonstrated among
profiling of exposed target cells (in vitro and in vivo) and profiling persons with substantial inhalation exposure to PAHs, including roofers
of plasma and urine of exposed experimental animals and humans. and pavers, coke oven workers, certain steel and iron manufacturing
For example, smoking-related biomarkers for lung cancer risk and workers, and aluminum production workers.33 In addition, several
early-stage lung cancer are needed to enhance early detection strategies studies have suggested that workers highly exposed through inhalation
and to provide a science base for tobacco product regulation. In an also might be at increased risk of cancer at sites other than skin and
untargeted metabolomics approach using ultra-high-performance lung, with suggestive increases in risk in the bladder, pancreas, and
liquid chromatography–quadrupole time-of-flight mass spectrometry upper gastrointestinal tract.
(UHPLC-Q-TOF MS), blood was collected from human subjects Several biochemical pathways are involved in the metabolism of
before and after they smoked a single cigarette. Sex- and race-related PAHs and of BaP in particular.31 One major pathway results in highly
dynamic metabolomic changes were detected in plasma, and novel reactive 7,8-dihydrodiol-9,10-epoxide-BaP, thought to produce DNA
biomarkers affected by cigarette smoking were identified, including adducts leading to cancers of the esophagus, forestomach, intestine,
a network of 12 molecules involved in cancer and carcinogenesis. lungs, and mammary gland in rodents.
Similarly, a nuclear magnetic resonance (NMR)-based metabolomic
study of sera from a rat model of gastric carcinogenesis revealed Aromatic Amines
early perturbation of metabolic networks associated with oxidative The occurrence of bladder cancer among dye industry workers was
stress. Choline phosphorylation, fatty acid degradation, amino acid reported in 1895 by the German physician Ludwig Rehn, who suggested
metabolism, and glycosis were disturbed during gastric carcinogenesis. a causal relationship. With the rapid expansion of the chemical industry
Likewise, a series of metabolomics case-control studies of prostate cancer during and after World War I, increased risk of bladder cancer was
revealed patterns predictive of disease risk and progression, including observed among workers employed in chemical manufacturing and
early disruption of pathways associated with abnormal cell growth, textile dyeing.34 An industry-wide study of workers exposed to dyes
intensive cell proliferation, and dysregulation of lipid metabolism. in England and Wales demonstrated strong increased risks of bladder
cancer among men exposed to 1-naphthylamine, benzidine, 2-
Microbiome naphthylamine, or mixed dyes, with 2-napthyamine implicated at the
One of the most surprising recent developments related to carcinogenesis highest hazard level.36 Additional studies in the dye industry identified
is the emerging importance of the microbiome as a major environmental auramine O and magenta as human bladder carcinogens. Increased
factor influencing cancer susceptibility and the carcinogenic process.72–78 risk of bladder cancer among rubber workers and in the electric cable
An individual’s microbiome is a complex function of genetics, lifestyle, industry has been attributed to the naphthylamine added to rubber
and culture including diet, and other exogenous factors including as an antioxidant.
antibiotic and antiinflammatory therapy. The relationship between the Aromatic amines are metabolized and excreted through a process
microbiome and host health is complex and can include prevention or involving acetylation by N-acetyltransferase.8 Genetic variation in one
promotion of carcinogenesis, modulation of the inflammatory response of the genes, NAT2, encoding this enzyme produces either rapid or
of the host, influence on the production of anticarcinogenic metabolic slow metabolic phenotypes in humans. Analysis of the NAT2 phenotypes
products such as butyrate, or even the production of carcinogenic of patients with bladder cancer from the dye industry indicates that
metabolic products or of toxins that disrupt the cell cycle. Although persons showing the slow phenotype could be more susceptible to
many of the studies conducted thus far have addressed the role of the bladder cancer caused by aromatic amines. This finding is consistent
microbiome in gastrointestinal and colorectal cancers, in which protein with a meta-analysis indicating that NAT2 slows acetylation status is
residues and fat-stimulated bile acids are metabolized by the microbiota associated with an increased risk of bladder cancer in the general
to inflammatory and/or carcinogenic metabolites, there is increasing population.79,80
evidence for its role in modulating the evolution of pancreatic, lung,
colorectal cancer, oral squamous cell carcinoma, and HCC. Benzene
Exposure to benzene was suspected to be the cause of leukemia in a
Chemicals number of individual cases and case series reported worldwide between
1928 and 1976.29 Case-control studies indicated increased risks of
Following are brief depictions of several chemical and physical agents nonlymphocytic leukemia among workers in Sweden exposed to
of environmental concern, which are illustrative of the roles of these petroleum products containing benzene and of lymphomas among
agents in the etiology of human cancers. A deeper understanding of workers in New York State who were exposed to benzene. Prospective
the mechanistic basis for the actions of these carcinogenic agents will studies conducted in the rubber industry provide the most convincing
allow for more effective means to identify other carcinogens in our evidence for an association between benzene exposure and leukemia.
environment and to develop preventive strategies to interrupt, block, Most of the excess leukemia in this industry is found among rubber
intercept, or reverse the neoplastic process. workers exposed to solvents, including benzene.
Polycyclic Aromatic Hydrocarbons Aflatoxins
The English surgeon Percivall Pott was among the first to document The hepatotoxic effect of aflatoxins was first recognized when aflatoxin-
the association of an environmental agent with cancer.29,30 During the contaminated feed was inadvertently fed to poultry. Subsequent animal
late 18th century, he determined that the unusually high incidence of studies demonstrated the carcinogenic potential of the aflatoxins,
scrotal cancer among chimney sweeps was due to their occupational particularly aflatoxin B1. The aflatoxins are produced by the fungal
exposure to soot and tar. It was not until the 20th century that strains Aspergillus flavus and Aspergillus parasiticus. Grains and foodstuffs
animal testing showed that the active carcinogens in soot and coal for human consumption such as corn, peanuts, and rice can become
148 Part I: Science and Clinical Oncology

contaminated with aflatoxin during growth or storage. The considerable although combustion enhances the carcinogenic properties of tobacco,
variation in levels of human exposure to aflatoxin worldwide is it is not required for cancer induction.
determined by climate and by the preventive measures used to protect Although the carcinogenic properties of tobacco tar were first
susceptible foods from mold contamination and growth.81 demonstrated experimentally during the 1920s, evidence of a human
Dietary aflatoxin is correlated with high liver cancer rates in cancer risk from the use of tobacco did not appear until 1939, when
sub-Saharan Africa, the Philippines, Mozambique, and Asia. The Muller and colleagues, as reviewed by Doll,26 reported an association
cocarcinogenic role of HBV infection and dietary aflatoxin in liver between tobacco use and lung carcinoma in Germany. Subsequent
cancer has been the focus of several studies. In a prospective study epidemiologic studies conducted in the United States and the United
conducted in Guangxi Province, China, the incidence of liver cancer Kingdom confirmed this causal relationship but were met with consider-
in regions of high and low aflatoxin contamination were compared, able societal resistance because tobacco use was considered a pleasurable
and HBV infection status was determined.82 A strong interaction personal habit. At that time the addictive characteristics of nicotine, a
between aflatoxin exposure and HBV-positive status was observed major constituent of tobacco, was unrecognized. Societal acceptance
for relative risk of liver cancer. Among HBV-positive individuals, the of a causal association with lung cancer was advanced by the first reports
incidence of liver cancer was 649 per 100,000 in the high-aflatoxin from the Royal College of Physicians in the United Kingdom (1962)
region and 66 per 100,000 in the low-aflatoxin region, whereas and from the Surgeon General in the United States (1964) regarding
among HBV-negative individuals, the incidence of liver cancer was the risks of tobacco use. By contrast, the tobacco industry has steadfastly
99 per 100,000 and less than 1 per 100,000 in high- or low-aflatoxin resisted attempts to educate the public regarding the health hazards of
regions, respectively. The association of urinary aflatoxin-DNA adducts tobacco use and has continued to market cigarettes aggressively, par-
with risk of liver cancer also has been demonstrated in a prospective ticularly in developing countries. Epidemic increases in the incidence
epidemiologic study.45 of lung cancer can be anticipated in countries where tobacco use is
surging. It is estimated that more than 1 million new cases per year of
Tobacco Chemicals lung cancer will occur in China in the 21st century.26–28,81,83
Tobacco use causes more cancer deaths worldwide than any other More than 3000 chemicals have been identified in cigarette smoke,
human activity. Cigarette smoking is associated with cancers of the of which at least 30 are known to be carcinogenic in animals (Table
lung, oral cavity, pharynx, larynx, esophagus, bladder, renal pelvis, 10.2). The gas phase of tobacco smoke contains several carcinogenic
and pancreas (see Box 10.1). The use of smokeless tobacco (chewing or tumor-promoting compounds, including dimethylnitrosamine,
tobacco or snuff ) leads to cancer of the oral cavity, indicating that dialkylnitrosamines, vinyl chloride, acrolein, and benzene. The

Table 10.2  Potential Carcinogenic Agents in Tobacco Smoke


Mainstream Smoke Mainstream Smoke
Compounds Compounds (Per Cigarette) Compounds Compounds (Per Cigarette)
POLYCYCLIC AROMATIC HYDROCARBONS 4-(Methylnitrosamine)-1- 0.08–0.77 µg
Benzo(a)anthracene 20–70 ng (3-pyridyl)-1-butanone
Benzo(b)fluoranthene 4–22 ng N-nitrosoanabasine 0.14–4.6 µg
Benzo(f)fluoranthene 6–21 ng AROMATIC AMINES
Benzo(k)fluoranthene 6–12 ng 2-Toluidine 30–200 ng
Benzo(a)pyrene 20–40 ng 2-Naphthylamine 1–22 ng
Chrysene 40–60 ng 4-Aminobiphenyl 2–5 ng
Dibenz(a,h)anthracene 4 ng ALDEHYDES
Dibenzo(a,i)pyrene 1.7–3.2 ng
Formaldehyde 70–100 µg
Dibenzo(a-1)pyrene Detectable
Acetaldehyde 18–1400 ng
Indenol(1,2,3-c,d)pyrene 4–20 ng
Crotonaldehyde 10–20 µg
5-Methylchrysene 0.6 ng
MISCELLANEOUS ORGANIC COMPOUNDS
AZA-ARENES
Benzene 12–48 µg
Quinoline 1–2 µg
Acrylonitrile 3.2–15 µg
Dibenz(a,h)acridine 0.1 µg
2-Nitropropane 0.73–1.21 µg
Dibenz(a,j)acridine 3–10 ng
Ethylcarbamate 20–38 ng
7H-dibenzo(c,g)carbazole 0.7 ng
Vinyl chloride 1–16 ng
N-NITROSAMINES INORGANIC COMPOUNDS
N-nitrosodimethylamine 0.1–180 ng
Hydrazine 24–43 ng
N-nitrosoethylmethylamine 3–13 ng
Arsenic 40–120 ng
N-nitrosodiethylamine 0–25 ng
Nickel 0–600 ng
N-nitrosopyrrolidine 1.5–110 ng
Chromium 4–70 ng
N-nitrosodiethanolamine 0–36 ng
Cadmium 41–62 ng
N-nitrosonornicotine 0.12–2.7 µg
Polonium-210 0.03–1.0 pCi

From U.S. Department of Health and Human Services. Reducing the Health Consequences of Smoking: 25 Years of Progress. Washington, DC: US Department of Health and
Human Services; 1989.
Environmental Factors  •  CHAPTER 10 149

particulate phase contains carcinogenic and cocarcinogenic PAHs, pigmentation of the skin, either constitutive or induced (as in tanning),
methylated PAHs, heterocyclic hydrocarbons, chlorinated hydrocarbons, plays an important role in protecting skin from the carcinogenic
phenols, catechols, and metals. There is a strong interactive effect effects of UV radiation.
observed when certain mixtures of these compounds are assayed for Increased levels of DNA photodamage are detected in the normal
carcinogenic potential. epidermis of individuals after exposure to solar UV radiation, and
Secondhand smoke contains many of the same carcinogens analysis of mutational spectra in human nonmelanoma skin cancer
that are inhaled by smokers. It is now known that secondhand DNA shows that mutations specific for UV radiation (dipyrimidine
smoke causes lung cancer in adults who do not themselves smoke.13 mutations) often are present. Additional potential contributing effects
Nonsmokers exposed to secondhand smoke at home or work are at include immune suppression and the molecular and cellular conse-
a 20% to 30% increased risk for the development of lung cancer.28 quences of chronic tissue damage and repair. Animal studies confirm
the carcinogenic effects of UV radiation and indicate that the ultraviolet
Chemotherapeutic Agents B (UVB) portion (280 to 320 nm) of the solar spectrum is primarily
responsible for the carcinogenic properties of sunlight. This wave
The systemic toxicity of sulfur mustard gas among soldiers exposed band encompasses the long-wavelength end of the absorbance spectrum
during World War I led to investigations of the cytotoxic MOA of of DNA and has been shown to cause mutations in mammalian cells
nitrogen mustard compounds and subsequent use as antineoplastic (reviewed in D’Orazio and colleagues85 and Shah and colleagues86).
drugs in the 1940s. Other types of drugs were also developed at that
time for use in the treatment of cancer, including the antibiotic Ionizing Radiation
actinomycin A and the antimetabolite methotrexate. In later decades The discovery and manipulation of ionizing radiation in the early
a variety of alkylating agents were introduced (e.g., chlorambucil, 20th century led to detrimental health effects among many researchers.
cyclophosphamide, bis-chloroethylnitrosourea, busulfan, and cisplatin), Toxicity, radiation burns, and cancer were observed among handlers
along with antimetabolites (e.g., 5-fluorouracil and 6-mercaptopurine), of radioactive materials. The deaths of Marie Curie and Thomas Edison’s
antibiotics (e.g., Adriamycin, bleomycin, and daunomycin), and mitotic assistant from cancer have been attributed to severe radiation exposure.
inhibitors (e.g., vincristine and vinblastine) as antineoplastics. The use of radium in luminous paint during the 1930s led to a high
As early as 1948, the carcinogenic properties of the anticancer incidence of osteosarcoma among dial painters who inadvertently
drug 4-aminostilbene and its metabolites were reported, leading to the ingested radium when shaping their brush tips with the tongue.1 By
institution of carcinogenesis bioassays for new anticancer drugs under the 1940s, an elevated incidence of leukemia was observed among
development by the National Cancer Institute. The appearance of radiologists. After World War II, excessive rates of leukemia were
frank second malignancies among patients treated with chemotherapy observed among atomic bomb survivors and among patients treated
was reported during the 1970s. The successful treatment of Hodgkin with x-rays.
disease with multiagent chemotherapy is associated with the long-term Epidemiologic studies of populations exposed to high doses of
complication of acute myeloid leukemia (AML) and non-Hodgkin ionizing radiation have indicated increased risks for a variety of cancers,
lymphoma. Increased risk of AML among patients treated for non- depending on the type of radiation and route of exposure (Table
Hodgkin lymphoma, ovarian cancer, multiple myeloma, or small 10.3). Among atomic bomb blast survivors, excessive rates of leukemias
cell carcinoma of the lung also has been attributed to antineoplastic appeared within several years of exposure, whereas excessive rates of
therapy. The risk of AML is most strongly associated with the alkyl- cancers of the breast, lung, esophagus, thyroid, colon, bladder, and
ating antineoplastics—particularly cyclophosphamide, melphalan, ovary, as well as multiple myeloma, appeared only 20 to 25 years later.
busulfan, treosulfan, and semustine (methyl-CCNU)—or with In contrast, populations exposed to nuclear weapon fallout show only
combination chemotherapies that include alkylating agents. Ovarian an excessive risk of thyroid cancer due to radioactive iodine. Almost
cancer patients receiving chemotherapy alone (including melphalan, all data on carcinogenic risks of ionizing radiation derive from extremely
thiotepa, chlorambucil, cyclophosphamide, Adriamycin, cisplatinum, high-exposure scenarios, making it difficult to extrapolate risk estimates
alone or in combination) had a relative risk of 12.0 for leukemia to low-dose or ambient exposure levels.
compared with patients treated with surgery alone (reviewed in De Vita Heavy exposure to x-rays for diagnostic or therapeutic procedures
and Chu84). has been associated with increased risk of the following types of cancers:
leukemia after in utero exposure; breast cancer after repeated chest
Radiation Carcinogenesis exposure; leukemia, lung, stomach, and esophagus after spinal exposure;
and thyroid, skin, and neck after scalp or thymus exposure.5 The use
Ultraviolet Radiation of cobalt-60 x-ray treatment for cervical cancer is associated with
Solar ultraviolet (UV) radiation is the major physical carcinogen in leukemia and cancers of the stomach, rectum, bladder, vagina, buccal
our environment and the primary cause of skin cancer in humans. cavity, nasopharynx, and lung (reviewed in Robertson and colleagues87).
New basal cell carcinoma or squamous cell carcinoma of the skin will
develop in more than 800,000 persons each year in the United States, Radon
making nonmelanoma skin cancer the most common cancer.7 The Radon gas is encountered in hard rock mining for iron, tin, fluorspar,
incidence of both nonmelanoma and melanoma skin cancer among and uranium. The radioactive decay of radon and its products produces
light-skinned individuals is increasing at a rate of 3% to 5% per year alpha particles. The earliest reports defining an association between
in the United States. This increase has been attributed to changing lung cancer and mining described the high rate of lung cancers among
lifestyle and leisure habits during the past five decades, resulting in uranium miners in the Schneeberg region of Czechoslovakia in the
an increase in the number of people receiving greater exposure to late 19th century. Many potential causes were proposed, including
sunlight. radon inhalation. Studies of lung cancer mortality among Colorado
Geography plays a role in the incidence of nonmelanoma skin uranium miners demonstrated dose-related increases in lung cancer
cancer, which shows a generally increasing trend with decreasing latitude risk in miners with protracted exposure to radon. In addition, small
among persons with similar skin types.7 Nonmelanoma skin cancers cell undifferentiated carcinomas predominated in miners with high
and the premalignant skin neoplasm actinic keratosis, particularly on exposure to radon, in contrast to the typical distribution of pulmonary
solar-exposed skin areas, are also associated with cumulative lifetime cancer pathology in the general US population. An elevated risk of
UV exposure estimated from outdoor occupations and activities. Light lung cancer also has been reported for iron ore miners in England,
skin complexion, ease of sun burning (skin type), and light hair color France, and Sweden; however, the proportion of risk attributable to
are known to enhance the risk of nonmelanoma skin cancer, whereas radon in these populations is more difficult to assess.
150 Part I: Science and Clinical Oncology

Table 10.3  Examples of Radiation-Induced Cancers


Sources of Exposure Exposure Circumstances Cancer Types
EXPLOSIONS OF NUCLEAR WEAPONS
Blast Atomic bombing survivors in Hiroshima and Nagasaki Leukemia, breast, lung, thyroid, stomach, colon, multiple
myeloma, esophagus, ovary
Fallout Populations exposed through atmospheric testing, Thyroid
including Marshall Islanders, veterans in the Pacific,
general population in Nevada, Utah
DIAGNOSTIC PROCEDURES
X-rays Children exposed in utero Leukemia
Thorotrast Cerebral and limb angiography; of biliary passages Liver
Fluoroscopic x-rays Monitoring of lung infections in patients with tuberculosis Breast
THERAPEUTIC PROCEDURES
X-rays Postpartum mastitis Breast
X-rays Ankylosing spondylitis Leukemia, lung, stomach, esophagus
Cobalt-60 Treatment for cancer of the cervix Leukemia, stomach, rectum, bladder, vagina, female genital,
lung, buccal cavity, nasopharynx, esophagus
X-rays Treatment of benign head and neck conditions Thyroid, skin, central nervous system
Radium-224 Ankylosing spondylitis, bone tuberculosis Bone sarcoma
PROFESSIONAL EXPOSURES
X-rays Early radiologists Skin, leukemia
Radon Uranium, hard-rock miners Lung cancer
X-rays, γ-rays, neutrons Nuclear industry Multiple myeloma
Radium isotopes Radium dial painters Bone, head sarcoma

Modified from Higginson J, Muir CS, Munoz N. Human Cancer: Epidemiology and Environmental Causes. Cambridge Monographs on Cancer Research. Cambridge: Cambridge
University Press; 1992.

Analysis of lung cancer mortality and smoking in Colorado uranium sheep dip in Wales found an excess of skin and lung cancers, particularly
miners suggests a greater than additive mortality rate for cumulative among persons directly involved in the chemical processes. Case-control
radon exposure and cumulative cigarette smoking. In other words, studies in two US plants manufacturing arsenical pesticides found
the increased risk of lung cancer among miners compared with nonmin- arsenic dose–related increases in the risk of lung cancer. Reports of
ers is larger when comparing smokers than when comparing nonsmok- skin and lung cancers among vineyard workers with exposure to arsenic
ers. Interesting to note, among atomic bomb survivors, cigarette fungicides and pesticides appeared during the late 1950s. An autopsy
smoking and radiation exposure have an additive effect only for lung series of 82 vineyard workers exposed in Germany found 61 deaths
cancer risk. This anomaly has been attributed to the different exposure from cancer, including 44 respiratory tract cancers; many skin cancers
patterns experienced by atomic bomb survivors (acute) and uranium and Bowen disease also were reported.
miners (chronic) (reviewed in Robertson and colleagues87 and Jostes88). The most common route of exposure to arsenic worldwide is through
drinking water, with estimates of more than 50 million people using
Metals well water containing excess arsenic levels—20 million in Bangladesh
alone. Chronic arsenic exposure has been linked to cancer of the
Arsenic bladder, lungs, skin, and kidney (reviewed in IARC Working Group
Medicinal use of inorganic arsenic was associated with skin cancers on the Evaluation of Carcinogenic Risks to Humans89 and Bustaffa
in the early 20th century. More recently, excessive rates of skin cancer and colleagues90).
have been observed in populations exposed to drinking water con-
taminated with arsenic, whereas excessive rates of lung cancer have Nickel and Chromium
been found in populations with occupational exposure to inorganic Carcinogenic associations for nickel and chromium are generally limited
arsenic compounds. An increased risk of lung cancer of 6- to 14-fold to occupational inhalation exposure to either specific particulate forms
was reported in gold miners in Rhodesia, where the ore contains or acid mists of these metals. The nickel refining industry was established
arsenic. Chronic arsenism also was prevalent among these miners. in South Wales around 1900. During the subsequent 40 years, evidence
Several studies in Japan, Sweden, and the United States have docu- accumulated for increased rates of nasal cancer and, later, lung cancer
mented excessive rates of lung cancers among workers involved in among workers in nickel refineries, particularly among process workers
copper smelting. Inorganic arsenic is a byproduct of the smelting in the refinery. Cancer risk for nickel began to decline when occupa-
process and also is used as a hardener. Two large retrospective studies tional safety measures were introduced in the industry from the 1930s
of copper smelters have shown that lung cancer mortality is related to 1950s.91,92
to estimated arsenic exposure. Several reports of lung cancer in chromate industry workers appeared
Another source of occupational and potential environmental in Germany during the 1930s. Subsequent extensive epidemiologic
exposure is the manufacture and use of arsenical pesticides. An early investigations examined this association in workers involved in chromate
study of mortality among workers at a factory manufacturing arsenical production, chromate pigment manufacture, and chrome plating in
Environmental Factors  •  CHAPTER 10 151

England and Baltimore. These studies suggest that inhalation of specific suggests that overall cancer mortality increases by 10% for each 5 kg/
forms of particulate hexavalent chromium compounds, or of acid m2 increase in BMI. The IARC concluded that the existing evidence
mists, were related to excessive rates of lung cancer. Experimental base supports an association between obesity and cancers of the liver,
investigations indicate that highly toxic or cytotoxic doses of hexavalent colon, postmenopausal breast, endometrium, kidney, and esophagus.36
chromium salts of chromium are genotoxic and carcinogenic. There A recent meta-analysis of prospective studies found strong associations
is evidence of a potential high-dose threshold effect related to the between increased BMI and esophageal adenocarcinoma, thyroid cancer,
high capacity of body fluids to reduce hexavalent chromium to trivalent kidney cancer, and colon cancer among men and associations between
chromium. In contrast to hexavalent chromium, trivalent chromium increased BMI and endometrial cancer, gallbladder cancer, kidney
is a human essential element and is not carcinogenic (reviewed in cancer, and esophageal cancer among women.
Nickens and colleagues14 and IARC Working Group on the Evaluation The mechanism by which obesity increases cancer risk is unclear
of Carcinogenic Risks to Humans91). at this time, but many hypotheses have been advanced, including
increased storage and release of lipophilic carcinogens in and from
Fibers fatty tissue; lack of excretion of carcinogens due to decreased physical
activity; and altered gene expression related to inflammation or disrup-
Asbestos tion in hormonal homeostasis and energy balance. A molecular link
The appearance of lung cancer in patients exposed to asbestosis was has been found between cholesterol metabolism and breast cancer
first reported in the 1930s. Early epidemiologic studies were limited by risk, related to increased levels of circulating estrogenic metabolites.94–97
various deficiencies in exposure assessment, but gradually studies showed The risk of colon cancer is strongly associated with consumption
increased lung cancer risk in workers with potential exposure to asbestos of red meat and animal fat. This association has been observed in
during mining, milling, manufacturing, insulating, and shipbuilding. several international correlative studies and case-control studies. Prospec-
The association between asbestos exposure and mesothelioma of the tive studies of colon cancer risk demonstrate an association with
lung, a relatively rare cancer, was reported in the early 1960s and consumption of red meat and animal fat (but not with vegetable fat),
was confirmed among insulation workers, asbestos manufacturing independent of total energy intake. The increased risk associated with
workers, and other populations exposed through their occupations. animal fat primarily is due to meat intake as opposed to dairy product
Further studies showed that the risk of mesothelioma was not limited intake. Other studies that have addressed cooking practices have found
to the workers but extended to members of their household and to that consumption of fried foods and barbecued, broiled, or smoked
residents living in the vicinity of asbestos-related industries. There is meats is associated with an increased risk of colorectal cancer. Several
also evidence of a synergistic effect of asbestos and smoking for lung hypotheses have been proposed to explain the strong association of
cancer (reviewed in O’Reilly and colleagues93). colon cancer risk with red meat and animal fat. Diets high in meat
fat increase the incidence of colon cancers in rats and also increase
DIETARY FACTORS IN HUMAN the excretion of primary bile acids, which are converted to secondary
CARCINOGENESIS bile acids by bacterial metabolism. Some secondary bile acids (e.g.,
deoxycholic and lithocholic acid) are colon tumor promoters in animal
Most of the known human carcinogens discussed in the foregoing models. Moreover, the cooking of meat, particularly by broiling or
sections have been identified from occupational and iatrogenic exposures frying and pyrolysis of amino acids and proteins, produces at least
at high doses to small cohorts; however, with the exception of tobacco, two major classes of carcinogens—PAHs and heterocyclic aromatic
such agents are rather minor contributors to the current overall cancer amines—that are capable of producing genotoxic and mutagenic
burden. This fuels an overarching assumption that most human cancers damage. Heterocyclic amines, including quinolines, quinoxalines,
may result from interactions of several or more carcinogenic influ- pyridines, and carbolines, cause colon cancer, mammary cancer, liver
ences, with many modifying factors, the most significant thought to cancer, prostate cancer, and lymphoma in experimental animals.
be diet and other lifestyle habits. Many epidemiologic studies have Case-control studies of heterocyclic amine intake have found an
indicated that general increases in consumption of fiber-rich cereals, increased risk for colon cancer among those who had the highest
fruits, and vegetables and decreased consumption of fat-rich foods and levels of heterocyclic amine intake.98–100
excessive alcohol will serve as prudent approaches to reducing overall Numerous studies have identified many other dietary components
cancer risk. as having either carcinogenic or opposing anticarcinogenic potential.23
Two major dietary influences are thought be fat and calorie con- Such food-derived anticarcinogens consist of both nutrient (e.g.,
sumption. Fat consumption is most strongly associated with the vitamins and minerals) and nonnutrient components, many of which
hormone-dependent cancers (i.e., breast [see Box 10.2], ovary, and function as antioxidants. Inverse epidemiologic associations between
endometrium in women and prostate in men) and gastrointestinal risk of cancer at several sites and ingestion of fruits and vegetables
cancers (i.e., gallbladder, colon, and rectum in both sexes). It is not have been observed.24 These associations might be linked to β-carotene,
clear to what degree these relationships are causal or if they relate to other carotenoids, folate, fiber, vitamin C, and other antioxidants,
the type of fat (saturated, unsaturated, polyunsaturated) or the overall and to other components of the fruits and vegetables. A deeper
caloric content of the diet. Fats can promote tumor development understanding of the role of dietary factors in human carcinogenesis,
directly and also are a major source of calories. Animals fed high-fat coupled with effective behavioral adaptations, will likely have a sig-
diets consistently demonstrate enhanced tumorigenic outcomes. nificant impact on disease incidence.49,101
Conversely, it has been recognized for decades that caloric restriction
has a very powerful, general inhibitory effect on carcinogenesis in GENETIC POLYMORPHISMS AND
many induced and spontaneous laboratory animal tumor models. The HUMAN SUSCEPTIBILITY
human behavioral changes required to effect comparable population-
wide reductions in fat and/or calorie consumption, however, pose Another major area of research in molecular cancer epidemiology is
formidable challenges. the study of individual and population-wide genetic polymorphisms
The increasing prevalence of overweight and obese individuals that affect cancer susceptibility, particularly as they affect metabolic
worldwide, combined with mounting evidence for a clear association and DNA repair.102 Many cytochrome P450 metabolic enzymes are
between obesity and cancer risk, presents a major public health concern. known to be involved in the activation of specific human carcinogens,
Analysis has indicated that more than 900 million adults are overweight and some have been linked to increased cancer risk. Inducible CYP1A1
and almost 400 million are obese (BMI ≥30 kg/m2).16 In addition to activity is higher in cultured lymphocytes from persons with lung
the noncancer health risks associated with obesity, current evidence cancer than in control subjects. Genetic polymorphisms in the CYP1A1
152 Part I: Science and Clinical Oncology

structural gene have been associated with lung cancer risk in Asian Cancer chemoprevention could be especially valuable in populations
populations. Hepatic arylamine N-acetyltransferase correlates with at high risk of certain neoplasms, particularly as improved molecular
individual differences in susceptibility to bladder cancer in humans.28 techniques allow for the identification of such high-risk individuals.
A series of genetic polymorphisms of this enzyme exists in humans, Preventive strategies can involve interventions with specific nutrients,
resulting in slow- and rapid-acetylator phenotypes. The rapid-acetylator nonnutrients, and antiinflammatory drugs in select populations at
phenotype seems to protect aromatic amine-exposed individuals from high risk of cancer, and lifestyle changes that include altered nutrition
bladder cancer. These results are attributed to the competition of and social habits.105–108 Finally, increased attention is currently being
N-acetylation of arylamines against the formation of reactive arylamine applied to cancer “interception,” which is focused on identifying the
metabolites that reach the bladder. earliest stages of carcinogenesis and applying therapeutic interventions
An important mechanism that protects the genome from genotoxic to prevent that early cancer from progressing.109
effects of carcinogens is the repair of cellular DNA. The rare inherited
disorder xeroderma pigmentosum (XP) is characterized by various SUMMARY
levels of DNA repair deficiencies, and persons with XP show an
unusually high incidence of multiple skin cancers and rarely live beyond A key paradigm in environmental carcinogenesis—gene-environment
early adulthood in the absence of protective measures against sunlight.103 interactions, a concept used to describe the complex interplay between
The median age at onset for nonmelanoma skin cancers among persons individual or population genetics and responses to chemical agents—has
with XP is approximately 8 years, compared with about 60 years in also undergone an expansive transformation into a more contemporary
the general population. understanding of the human exposome. The exposome is the cumulative
Several other inherited diseases show altered cellular response to lifelong burden of disease-contributing stressors including exogenous
DNA damage. Ataxia telangiectasia, Bloom syndrome, and Fanconi and endogenous agents of all types, in addition to an individual’s
anemia are autosomal-recessive genetic disorders characterized by “omics” profile (genomics, epigenomics, transcriptomics, proteomics,
chromosomal instability, with malignancies developing in patients metabolomics) and his or her microbiome. A classic sequential model
more frequently and at a younger age than occurs in the general of initiation, promotion, conversion, and progression has increasingly
population. Heterozygous relatives of persons with ataxia telangiectasia evolved into less linear models that no longer classify carcinogens as
and those with Fanconi anemia also are at moderately increased risk initiators or promoters per se. It may be more important to understand
of cancer compared with unrelated persons.104 the contribution of chemical agents to the various hallmarks of cancer:
Although these are rare diseases, they suggest that moderate or genomic instability, resisting cell death, deregulating cellular energetics,
even small deficiencies in DNA repair may affect susceptibility to sustaining proliferative signals, evading growth suppressors, avoiding
cancer, and many studies have documented significant variability in immune destruction, enabling replicative immortality, promoting
DNA repair proficiency among disease-free individuals. tumor inflammation, activating invasion and metastasis, and inducing
angiogenesis.
PUBLIC HEALTH APPROACHES TO CANCER A traditional paradigm has classified carcinogenic agents as envi-
PREVENTION AND INTERCEPTION ronmental or lifestyle related or occupational in origin, but these
classifications are blurred by many examples of complex interplay
The optimal way for dealing with virtually all diseases, including among all three types of exposure. Carcinogen identification has
cancer, is prevention. Thus the challenge to public health professionals evolved from historic observational studies, to occupational cohort
is to devise and implement preventive measures against cancer. As epidemiology, to broad-scale screening and testing in experimental
presented in Fig. 10.1, the prevention of cancer can take several forms. animals. Next-generation approaches to carcinogen identification use
Primary prevention targets healthy persons and can be achieved by exposome approaches of integrating sophisticated individual exposure
avoiding exposure to risk factors. Another approach is to stimulate monitoring and biosensing with highly sensitive “omics” approaches
the defense mechanisms of the host to interfere with the carcinogenic to detect early perturbation of disease-relevant signaling pathways.
process. Secondary prevention measures use early detection and Most human cancers probably result from the interaction of several
intervention (cancer interception) before pathologic conditions are carcinogenic influences (often unidentified, but many dietary or lifestyle
clinically apparent. The success of these strategies will be greatly related in origin) along with intrinsic factors (e.g., inherited genes,
facilitated by exposome approaches (see earlier) to the development hormones, inflammation and oxidative stress, immune status, energy
of noninvasive biomarkers that identify disease-relevant exposures, as balance).
well as high-risk individuals. Finally, tertiary prevention is aimed at Although the causes of most individual human cancers are not
minimizing the effect of existing disease and consequent disability by identifiable, there is incontrovertible evidence that certain chemical
avoiding development of new cancers and complications or relapses agents, radiation, and certain biologic agents are contributors to the
after therapy for initial malignancies. overall incidence of human cancer. Consumption of tobacco products,
The use of chemical or dietary interventions to alter the susceptibility especially cigarette smoking, is responsible for nearly 30% of all cancers.
of humans to the actions of carcinogens and to retard, block, or reverse Chemical carcinogens include aromatic amines, benzene, aflatoxins,
carcinogenesis can be applied to all levels of prevention and has been tobacco chemicals, and chemotherapeutic agents. Metal carcinogens
termed chemoprevention. These strategies have proved to be extremely include specific forms of arsenic, nickel, and hexavalent chromium,
effective in laboratory animals, but increasing evidence indicates that largely associated with occupational exposures. Radiation carcinogens
chemoprevention against cancer also works in humans. The field of include UV radiation, ionizing radiation, and radon. Fibers such as
cancer chemoprevention did not receive significant attention until asbestos and certain dusts are etiologic agents in lung cancers and
the early 1970s, when Wattenberg demonstrated that dietary antioxi- mesothelioma. Many components in the diet can influence the
dants could protect against tumor formation. The experimental development of cancer through carcinogenic or anticarcinogenic
observations that seemingly innocuous preservatives found in the mechanisms.
Western diet could dramatically protect against diverse carcinogens The identification of molecular biologic markers of exposure,
at distal sites sparked the development of chemoprevention as a viable effect, and susceptibility (reflecting early events that link exposure
strategy for the reduction of human cancers. More than 20 classes of to adverse outcome, but before clinical disease) will help further our
discrete chemicals have been shown to be effective inhibitors of understanding of human carcinogenesis. In addition to genetic poly-
experimental carcinogenesis. Investigations have established significant morphisms indicative of susceptibility, biomarkers of disease-relevant
site- and agent-specific effects in the prevention of invasive skin, upper exposure will increasingly rely on highly sensitive, multiplatform, and
aerodigestive tract, colorectal, and breast cancers in high-risk groups. multidimensional analysis of the impact of potential carcinogenic
Environmental Factors  •  CHAPTER 10 153

agents on epigenomics, transcriptomics, proteomics, metabolomics, signaling that may also be targets for chemoprevention or cancer
and the microbiome. Primary, secondary, and tertiary cancer preven- interception.
tion approaches will be greatly facilitated by the development of
noninvasive biomarkers that identify high-risk individuals, and by The complete reference list is available online at
identification of the disease-consequential perturbations in molecular ExpertConsult.com.

KEY REFERENCES
1. Doll R, Peto R. The causes of cancer: quantitative 22. Casey SC, et al. The effect of environmental chemi- cancer link. Endocr Relat Cancer. 2014;21:T33–
estimates of avoidable risks of cancer in the United cal on tumor microenvironment. Carcinogenesis. T55.
States today. J Natl Cancer Inst. 1981;66:1191–1308. 2015;36(suppl 1):S160–S183. 71. Hsu PC, Lan RS, Brasky TM, Marian C,
3. Hunter DJ. Gene-environment interactions in 23. Thompson PA. Environmental immune disrup- Cheema AK, Ressom HW, et al. Metabolomic
human diseases. Nat Rev Genet. 2005;6:287–298. tors, inflammation and cancer risk. Carcinogenesis. profiles of current cigarette smokers. Mol Carcinog.
4. Wild CP. Complementing the genome with an 2015;36(suppl 1):S232–S253. 2017;56(2):594–606. doi:10.1002/mc.22519. [Epub
“exposome”: the outstanding challenge of envi- 24. Robey RB, et al. Metabolic reprogramming and dysreg- 2016 Aug 22].
ronmental exposure measurement in molecular ulated metabolism: cause, consequence and/or enabler 72. Pope JL, Tomkovich S, Yang Y, Jobin C. Microbiota
epidemiology. Cancer Epidemiol Biomarkers Prev. of environmental carcinogenesis? Carcinogenesis. as a mediator of cancer progression and therapy.
2005;14:1847–1850. 2015;36(suppl):S203–S231. Transl Res. 2017;179:139–154.
5. Rappaport SM. Implications of the exposome for 25. Langie SA, et al. Causes of genomic instability: the 73. Spor A, Koren O, Ley R. Unravelling the effects
exposure science. J Exp Sci Environ Epidemiol. effect of low dose chemical exposures in modern of the environment and host genotype on the gut
2011;21:5–9. society. Carcinogenesis. 2015;36(suppl):S61–S88. microbiome. Nat Rev Microbiol. 2011;9:279–290.
7. Weiss RA. Multistage carcinogenesis. Br J Cancer. 26. Doll R. Uncovering the effects of smoking: historical 77. Pevsner-Fischer M, Tuganbaev T, Meijer M, et al.
2004;91:1981–1982. perspective. Stat Methods Med Res. 1998;7:87–117. Role of the microbiome in non-gastrointestinal
8. Chappell G, Pogribny IP, Guyton KZ, Rusyn 28. Centers for Disease Control and Prevention. Smoking- cancers. World J Clin Oncol. 2016;73:200–213.
I. Epigenetic alterations induced by genotoxic attributable mortality, years of potential life lost, 78. Schwabe RF, Jobin C. The microbiome and cancer.
occupational and environmental human chemical and productivity losses—United States, 2000–2004. Nat Rev Cancer. 2013;13:800–812.
carcinogens: a systematic review. Mutat Res Rev Mutat MMWR Morb Mortal Wkly Rep. 2008;57(45): 82. Kensler T, Qian GS, Chen JG, Groopman JD.
Res. 2016;768:27–45. 1226–1228. Translational strategies for cancer prevention in
9. Shkreta L, Bell B, Revil T, Venables JP, Prinos P, 29. Alderson M. Occupational Cancer. London: liver. Nat Rev Cancer. 2003;3:321–329.
Elela SA, et al. Cancer-associated perturbations in Butterworth; 1986. 83. World Health Organization. WHO Report on the
alternative pre-messenger RNA splicing. Cancer Treat 33. Higginson J, Muir CS, Munoz N. Human Cancer: Global Tobacco Epidemic, 2011: Warning About the
Res. 2013;158:41–94. Epidemiology and Environmental Causes. Cambridge Dangers of Tobacco. Geneva: World Health Organiza-
10. Reed JC. Dysregulation of apoptosis in cancer. Monographs on Cancer Research. Cambridge: tion; 2011.
J Clin Oncol. 1999;17:2941053. Cambridge University Press; 1992. 84. DeVita VT Jr, Chu E. A history of cancer chemo-
13. Hanahan D, Weinberg RA. Hallmarks of cancer: 36. International Agency for Research on Cancer. IARC therapy. Cancer Res. 2008;21:8643–8653.
the next generation. Cell. 2011;144:646–674. Monographs on the Evaluation of Carcinogenic Risk 85. D’Orazio J, Jarrett S, Amaro-Ortiz A, Scott
14. Nickens KP, Patierno SR, Ceryak S. Chromium to Humans. Vol. 1–104. Lyon, France: IARC Press; T. UV Radiation and the Skin. Int J Mol Sci.
genotoxicity: a double-edged sword. Chem Biol 1970–2012. 2013;14:12222–12248.
Interact. 2010;188:276–288. 37. Rothman N, Hainaut P, Schulte PA, eds. Molecular 86. Shah DJ, Sachs RK, Wilson DJ. Radiation-induced
15. Liu X, He Y, Li F, Huang Q, Kato TA, Hall RP, Epidemiology: Principles and Practices. IARC Sci Publ cancer: a modern view. Br J Radiol. 2012;85:
et al. Redefining the roles of apoptotic factors in No. 163. Lyon: IARC Press; 2012. 1166–1173.
carcinogenesis. Mol Cell Oncol. 2015;3:e1054550. 38. Parkin DM, Bray F, Ferlay J, Pisani P. Global cancer 87. Robertson A, Allen J, Laney R, Curnow A. The
16. Pritchard DE, Ceryak S, Ramsey KE, O’Brien TJ, statistics, 2002. CA Cancer J Clin. 2005;55:74–108. cellular and molecular carcinogenic effects of radon
Ha J, Fornsaglio JL, et al. Resistance to apoptosis, 40. Munoz N, Day N. Esophageal Cancer. In: exposure: a review. Int J Mol Sci. 2013;14:14024–
increased growth potential, and altered gene Schottenfeld D, Fraumeni JF, eds. Cancer Epidemiol- 14063.
expression in cell that survived genotoxic hexavalent ogy and Prevention. New York: Oxford University 89. IARC Working Group on the Evaluation of
chromium exposure. Mol Cell Biochem. 2005;279: Press; 1996:681–706. Carcinogenic Risks to Humans. Arsenic, metals,
169–181. 42. Institute of Medicine. Breast Cancer and the Environ- fibres and dusts. IARC Monogr Eval Carcinog Risks
18. Carnero A, et al. Disruptive chemicals, senescence ment: A Life Course Approach. Washington, DC: Hum. 2012;100:11–465.
and immortality. Carcinogenesis. 2015;36:S19–S37. Institute of Medicine; 2011. 91. IARC Working Group on the Evaluation of Carci-
19. Goodson WH 3rd, et al. Assessing the carcinogenic 49. World Cancer Research Fund/American Institute for nogenic Risks to Humans. Chromium, nickel and
potential of low-dose exposures to chemical mixtures Cancer Research. Food, Nutrition and the Prevention welding. IARC Monogr Eval Carcinog Risks Hum.
in the environment: the challenge ahead. Carcino- of Cancer: A Global Perspective. Washington, DC: 1990;49:1–648.
genesis. 2015;36:S254–S296. AICR; 1997. 94. Basen-Engquit K, Chang M. Obesity and cancer
20. Narayanan KB, et al. Disruptive environmental 50. Escher BI, et al. From the exposome to a mechanistic risk: recent review and evidence. Curr Oncol Rep.
chemicals and cellular mechanisms that confer understanding of chemical-induced adverse effects. 2011;13:71–76.
resistance to cell death. Carcinogenesis. 2015;36(suppl Environ Int. 2017;99:97–106. 101. Ames BN. Dietary carcinogens and anticarcinogens.
1):S89–S110. 51. Miller MF, et al. Low-dose mixture hypothesis of Science. 1983;221:1256–1264.
21. Hu Z, et al. Assessing the carcinogenic potential carcinogenesis workshop: scientific underpinning and 102. Vineis P, ed. Metabolic Polymorphisms and Susceptibil-
of low-dose exposure to chemical mixtures in the research recommendations. Environ Health Perspect. ity in Cancer. IARC Sci Publ No. 148. Lyon: IARC
environment: focus on the cancer hallmark of 2017;125(2):163–169. Press; 1999.
tumor angiogenesis. Carcinogenesis. 2015;36(suppl 62. Knower KC, To SQ, Leung YK, Ho SM, Clyne CD. 109. Blackburn EH. Cancer Interception. Cancer Prev
1):S184–S202. Endocrine disruption of the epigenome: a breast Res (Phila). 2011;4:787–792.
Environmental Factors  •  CHAPTER 10 153.e1

REFERENCES
1. Doll R, Peto R. The causes of cancer: quantitative 24. Robey RB, et al. Metabolic reprogramming and dysreg- 46. Groopman JD, Kensler TW. The light at the end of
estimates of avoidable risks of cancer in the United ulated metabolism: cause, consequence and/or enabler the tunnel for chemical-specific biomarkers: daylight
States today. J Natl Cancer Inst. 1981;66:1191–1308. of environmental carcinogenesis? Carcinogenesis. or headlight? Carcinogenesis. 1999;20:1–11.
2. Arnold M, Leitzmann M, Freisling H, Bray F, 2015;36(suppl):S203–S231. 47. Strickland P, Kang D, Sithisarankul P. Polycyclic
Romieu I, Renehan A, et al. Obesity and cancer: 25. Langie SA, et al. Causes of genomic instability: the aromatic hydrocarbon metabolites in urine as
an update of the global impact. Cancer Epidemiol. effect of low dose chemical exposures in modern biomarkers of exposure and effect. Environ Health
2016;41:8–15. society. Carcinogenesis. 2015;36(suppl):S61–S88. Perspect. 1996;104(suppl 5):927–932.
3. Hunter DJ. Gene-environment interactions in 26. Doll R. Uncovering the effects of smoking: historical 48. Huff J. Chemicals causally associated with cancers
human diseases. Nat Rev Genet. 2005;6:287–298. perspective. Stat Methods Med Res. 1998;7:87– in humans and in laboratory animals. In: Waalkes
4. Wild CP. Complementing the genome with an “expo- 117. MP, Ward JM, eds. Carcinogenesis. New York: Raven
some”: the outstanding challenge of environmental 27. U.S. Department of Health and Human Services. Press; 1994.
exposure measurement in molecular epidemiology. The Health Consequences of Involuntary Exposure 49. World Cancer Research Fund/American Institute for
Cancer Epidemiol Biomarkers Prev. 2005;14: to Tobacco Smoke: A Report of the Surgeon General. Cancer Research. Food, Nutrition and the Prevention
1847–1850. Atlanta: U.S. Department of Health and Human of Cancer: A Global Perspective. Washington, DC:
5. Rappaport SM. Implications of the exposome for Services, Centers for Disease Control and Preven- AICR; 1997.
exposure science. J Exp Sci Environ Epidemiol. tion, Coordinating Center for Health Promotion, 50. Escher BI, et al. From the exposome to a mechanistic
2011;21:5–9. National Center for Chronic Disease Prevention and understanding of chemical-induced adverse effects.
6. Wild CP, Scalbert A, Herceg Z. Measuring the expo- Health Promotion, Office on Smoking and Health; Environ Int. 2017;99:97–106.
some: a powerful basis for evaluating environmental 2006. 51. Miller MF, et al. Low-dose mixture hypothesis of
exposures and cancer risk. Environ Mol Mutagen. 28. Centers for Disease Control and Prevention. carcinogenesis workshop: scientific underpinning and
2013;54:480–499. Smoking-attributable mortality, years of potential life research recommendations. Environ Health Perspect.
7. Weiss RA. Multistage carcinogenesis. Br J Cancer. lost, and productivity losses—United States, 2000– 2017;125(2):163–169.
2004;91:1981–1982. 2004. MMWR Morb Mortal Wkly Rep. 2008;57(45): 52. Brien GL, Valerio DG, Armstrong SA. Exploiting
8. Chappell G, Pogribny IP, Guyton KZ, Rusyn 1226–1228. the epigenome to control cancer-promoting gene-
I. Epigenetic alterations induced by genotoxic 29. Alderson M. Occupational Cancer. London: expression programs. Cancer Cell. 2016;29:464–476.
occupational and environmental human chemical Butterworth; 1986. 53. Liu F, Hon GC, Villa GR, Turner KM, Ikegama S,
carcinogens: a systematic review. Mutat Res Rev Mutat 30. Waldron HA. A brief history of scrotal cancer. Br Yang H, et al. EGFR Mutation promotes glioblas-
Res. 2016;768:27–45. J Ind Med. 1983;40:390–401. toma through epigenome and transcription factor
9. Shkreta L, Bell B, Revil T, Venables JP, Prinos P, 31. Dipple A, Moschel RC, Bigger CAH. Polynuclear network remodeling. Mol Cell. 2015;307–318.
Elela SA, et al. Cancer-associated perturbations in aromatic carcinogens. In: Searle CE, ed. Chemical 54. Minaroits J, Demcsak A, Banati F, Niller HH. Epi-
alternative pre-messenger RNA splicing. Cancer Treat Carcinogens, Vol. 2 (ACS Monograph 182). Wash- genetic dysregulation in virus-associated neoplasm.
Res. 2013;158:41–94. ington, DC: American Chemical Society; 1984:41. Adv Exp Med Biol. 2016;879:71–90.
10. Reed JC. Dysregulation of apoptosis in cancer. 32. Sontag JM. Carcinogens in Industry and the Environ- 55. Wang Q, Trevino LS, Wong RL, Medvedovic M,
J Clin Oncol. 1999;17:2941053. ment. New York: Marcel Dekker; 1981. Chen J, Ho SM, et al. Reprogramming of the
11. Tang D, Lotze MT, Kang R, Zeh HJ. Apop- 33. Higginson J, Muir CS, Munoz N. Human Cancer: epigenome by MLL1 links environmental exposures
tosis promotes early tumorigenesis. Oncogene. Epidemiology and Environmental Causes. Cambridge to prostate cancer risk. Mol Endocrinol. 2016;30:
2011;30:1851–1854. Monographs on Cancer Research. Cambridge: 856–871.
12. Hanahan D, Weinberg RA. Hallmarks of cancer. Cambridge University Press; 1992. 56. Teschendorff AE, Yang Z, Wong A, Pipinikas CP,
Cell. 2000;100:57–70. 34. Case RAM, Hosker ME, McDonald DB, Pearson JT. Jones A, Anjum S, et al. Correlation of smoking-
13. Hanahan D, Weinberg RA. Hallmarks of cancer: Tumours of the urinary bladder in workers engaged associated DNA methylation changes in buccal cells
the next generation. Cell. 2011;144:646–674. in the manufacture and use of certain dyestuff with DNA methylation changes in epithelial cancer.
14. Nickens KP, Patierno SR, Ceryak S. Chromium intermediates in the British chemical industry. JAMA Oncol. 2015;1:476–485.
genotoxicity: a double-edged sword. Chem Biol Br J Prev Soc Med. 1954;11:75. 57. Zhang C, Su ZY, Wang L, Shu L, Yang Y, Guo Y,
Interact. 2010;188:276–288. 35. Fujiki H. Gist of Dr. Katsusaburo Yamagiwa’s papers et al. Epigenetic blockade of neoplastic transforma-
15. Liu X, He Y, Li F, Huang Q, Kato TA, Hall RP, entitled “Experimental study on the pathogenesis tion by bromodomain and extra-terminal (BET)
et al. Redefining the roles of apoptotic factors in of epithelial tumors” (I to VI reports). Cancer Sci. domain protein inhibitor JQ-1. Biochem Pharmacol.
carcinogenesis. Mol Cell Oncol. 2015;3:e1054550. 2014;105:143–149. 2016;117:35–45.
16. Pritchard DE, Ceryak S, Ramsey KE, O’Brien 36. International Agency for Research on Cancer. IARC 58. Eckstein M, Eleazer R, Rea M, Fondufe-Mittendorf
TJ, Ha J, Fornsaglio JL, et al. Resistance to Monographs on the Evaluation of Carcinogenic Risk Y. Epigenomic reprogramming in inorganic
apoptosis, increased growth potential, and altered to Humans. Vol. 1–104. Lyon, France: IARC Press; arsenic-mediated gene expression patterns during
gene expression in cell that survived genotoxic 1970–2012. carcinogenesis. Rev Environ Health. 2016; Oct 4.
hexavalent chromium exposure. Mol Cell Biochem. 37. Rothman N, Hainaut P, Schulte PA, eds. Molecular 59. Sun H, Shamy M, Costa M. Nickel and epigenetic
2005;279:169–181. Epidemiology: Principles and Practices. IARC Sci Publ silencing. Genes (Basel). 2013;25:583–595.
17. Mathew R, Karantza-Wadsworth V, White E. No. 163. Lyon: IARC Press; 2012. 60. Li W, Guo Y, Zhang C, Wu R, Yang AY, Gaspar
Role of Autophagy in cancer. Nat Rev Cancer. 38. Parkin DM, Bray F, Ferlay J, Pisani P. Global cancer J, et al. Dietary phytochemicals and cancer che-
2007;7:961–967. statistics, 2002. CA Cancer J Clin. 2005;55:74–108. moprevention: a perspective on oxidative stress,
18. Carnero A, et al. Disruptive chemicals, senescence 39. Islami F, Kamangar F, Aghcheli K, et al. Epidemio- inflammation and epigenetics. Chem Res Toxicol.
and immortality. Carcinogenesis. 2015;36:S19–S37. logic features of upper gastrointestinal tract cancers in 2016;29:2071–2095.
19. Goodson WH 3rd, et al. Assessing the carcinogenic northeastern Iran. Br J Cancer. 2004;5:1402–1406. 61. Trevino LS, Wang Q, Walker CL. Hypothesis: activa-
potential of low-dose exposures to chemical mixtures 40. Munoz N, Day N. Esophageal cancer. In: tion of rapid signaling by environmental estrogens
in the environment: the challenge ahead. Carcino- Schottenfeld D, Fraumeni JF, eds. Cancer Epidemiol- and epigenetic reprogramming in breast cancer.
genesis. 2015;36:S254–S296. ogy and Prevention. New York: Oxford University Reprod Toxicol. 2015;54:136–140.
20. Narayanan KB, et al. Disruptive environmental Press; 1996:681–706. 62. Knower KC, To SQ, Leung YK, Ho SM, Clyne CD.
chemicals and cellular mechanisms that confer resis- 41. Tominaga S. Cancer incidence in Japanese in Japan, Endocrine disruption of the epigenome: a breast
tance to cell death. Carcinogenesis. 2015;36(suppl1): Hawaii, and western United States. Natl Cancer Inst cancer link. Endocr Relat Cancer. 2014;21:T33–T55.
S89–S110. Monogr. 1985;69:83–92. 63. Sauer SJ, Tapley M, Shah M, Save AV, Lyerly HK,
21. Hu Z, et al. Assessing the carcinogenic potential 42. Institute of Medicine. Breast Cancer and the Environ- Patierno SR, et al. Bisphenol A activates EGFR and
of low-dose exposure to chemical mixtures in the ment: A Life Course Approach. Washington, DC: ERK promoting proliferation, tumor spheroid forma-
environment: focus on the cancer hallmark of Institute of Medicine; 2011. tion and resistance to EGFR pathway inhibition
tumor angiogenesis. Carcinogenesis. 2015;36(suppl): 43. Winn DM. Science and society: the long island in estrogen receptor-negative inflammatory breast
S184–S202. breast cancer study project. Nat Rev Cancer. 2005;5: cancer cells. Carcinogenesis. 2017;38(3):252–260.
22. Casey SC, et al. The effect of environmental chemi- 986–994. 64. Tang XH, et al. Gene-expression profiling signatures
cal on tumor microenvironment. Carcinogenesis. 44. Fernandez SV, Russo J. Estrogen and xenoestrogens for the diagnosis and prevention of oral cavity car-
2015;36(suppl):S160–S183. in breast cancer. Toxicol Pathol. 2010;38:1–10. cinogenesis – genome-wide analysis using RNA-seq
23. Thompson PA. Environmental immune disrup- 45. Ross RK, Yuan JM, Yu MC, et al. Urinary aflatoxin technology. Oncotarget. 2015;6:24424–24435.
tors, inflammation and cancer risk. Carcinogenesis. biomarkers and risk of hepatocellular carcinoma. 65. Souza T, et al. New insights into BaP-induced toxic-
2015;36(suppl):S232–S253. Lancet. 1992;339:943–946. ity: role of major metabolites in transcriptomics and
153.e2 Part I: Science and Clinical Oncology

contribution to hepatocarcinogenesis. Arch Toxicol. 79. Hein DW. Molecular genetics and function of NAT1 94. Basen-Engquit K, Chang M. Obesity and cancer
2016;90:1449–1458. and NAT2: role in aromatic amine metabolism and risk: recent review and evidence. Curr Oncol Rep.
66. Shi J, et al. Distinct response of the hepatic transcrip- carcinogenesis. Mutat Res. 2002;506(507):65– 2011;13:71–76.
tome to Aflatoxin B1 hepatocellular carcinogenesis 77. 95. Vainio H, Bianchini F, eds. IARC Handbook of Cancer
and resistance in rats. Sci Rep. 2016;6:31898. 80. Marcus PM, Vineis P, Rothman N. NAT2 slow Prevention. Vol. 6. Weight control and physical
67. Cook JA, et al. Mass spectrometry–based metabo- acetylation and bladder cancer risk: a meta-analysis activity. Lyon, France: IARC Press; 2002.
lomics identifies longitudinal urinary metabolite of 22 case-control studies conducted in the general 96. Renehan AG, Tyson M, Egger M, et al. Body-mass
profiles predictive of radiation-induced cancer. population. Pharmacogenetics. 2000;10:115–122. index and incidence of cancer: a systematic review
Cancer Res. 2016;76:1569–1577. 81. Peto R, Lopez AD. Future worldwide health effects of and meta-analysis of prospective observational
68. Xie G, et al. Dysregulated hepatic bile acids current smoking patterns. In: Koop CE, Pearson CE, studies. Lancet. 2008;371:569–578.
collaboratively promote liver carcinogenesis. Int Schwartz MR, eds. Critical Issues in Global Health. 97. Roberts DL, Dive C, Renehan AG. Biological
J Cancer. 2016;139(8):1764–1775. doi:10.1002/ San Francisco: Jossey-Bass; 2001. mechanisms linking obesity and cancer risk: new
ijc.30219. [Epub 2016 Jun 17]. 82. Kensler T, Qian GS, Chen JG, Groopman JD. perspectives. Annu Rev Med. 2010;61:301–316.
69. Gu J, et al. Metabolomic analysis reveals altered Translational strategies for cancer prevention in 98. Willett WC, Stampfer MJ, Colditz GA, et al.
metabolic pathways in a rat model of gastric car- liver. Nat Rev Cancer. 2003;3:321–329. Relation of meat, fat, and fiber intake to risk of
cinogenesis. Oncotarget. 2016;7(37):60053–60073. 83. World Health Organization. WHO Report on the colon cancer in a prospective study of women. N
doi:10.18632/oncotarget.11049. Global Tobacco Epidemic, 2011: Warning About the Engl J Med. 1990;323:1664–1672.
70. Kuligowski J, Sanjuan-Herráez D, Vázquez-Sánchez Dangers of Tobacco. Geneva: World Health Organiza- 99. Knize MG, Kulp KS, Salmon CP, et al. Factors affect-
MA, Brunet-Vega A, Pericay C, Ramírez-Lázaro MJ, tion; 2011. ing human heterocyclic amine intake and the metab-
et al. Metabolomic Analysis of gastric cancer progres- 84. DeVita VT Jr, Chu E. A history of cancer chemo- olism of PhIP. Mutat Res. 2002;506(507):153–162.
sion within the Correa’s cascade using ultraperfor- therapy. Cancer Res. 2008;21:8643–8653. 100. Butler LM, Sinha R, Millikan RC, et al. Heterocyclic
mance liquid chromatography–mass spectrometry. 85. D’Orazio J, Jarrett S, Amaro-Ortiz A, Scott amines, meat intake, and association with colon
J Proteome Res. 2016;15(8):2729–2738. doi:10.1021/ T. UV radiation and the skin. Int J Mol Sci. cancer in a population-based study. Am J Epidemiol.
acs.jproteome.6b00281. [Epub 2016 Jul 20]. 2013;14:12222–12248. 2003;157:434–445.
71. Hsu PC, Lan RS, Brasky TM, Marian C, 86. Shah DJ, Sachs RK, Wilson DJ. Radiation-induced 101. Ames BN. Dietary carcinogens and anticarcinogens.
Cheema AK, Ressom HW, et al. Metabolomic cancer: a modern view. Br J Radiol. 2012;85: Science. 1983;221:1256–1264.
profiles of current cigarette smokers. Mol Carcinog. 1166–1173. 102. Vineis P, ed. Metabolic Polymorphisms and Susceptibil-
2017;56(2):594–606. doi:10.1002/mc.22519. [Epub 87. Robertson A, Allen J, Laney R, Curnow A. The ity in Cancer. IARC Sci Publ No. Lyon: IARC Press;
2016 Aug 22]. cellular and molecular carcinogenic effects of radon 1999:148.
72. Pope JL, Tomkovich S, Yang Y, Jobin C. Microbiota exposure: a review. Int J Mol Sci. 2013;14:14024– 103. Moriwaki S, Kraemer KH. Xeroderma pig-
as a mediator of cancer progression and therapy. 14063. mentosum—bridging a gap between clinic and
Transl Res. 2017;179:139–154. 88. Jostes RF. Genetic, cytogenetic, and carcinogenic laboratory. Photodermatol Photoimmunol Photomed.
73. Spor A, Koren O, Ley R. Unravelling the effects effects of radon: a review. Mutat Res. 1996;340: 2001;17:47–54.
of the environment and host genotype on the gut 125–139. 104. Friedberg EC, Walker GC, Siede W. DNA Repair
microbiome. Nat Rev Microbiol. 2011;9:279–290. 89. IARC Working Group on the Evaluation of and Mutagenesis. Washington, DC: ASM Press; 1995.
74. Ussar S, Griffin NW, Bezy O, et al. Interactions Carcinogenic Risks to Humans. Arsenic, metals, 105. Kelloff GJ, Sigman CC, Greenwald P. Cancer
between gut microbiota, host genetics and diet fibres and dusts. IARC Monogr Eval Carcinog Risks chemoprevention: progress and promise. Eur J
modulate the predisposition to obesity and metabolic Hum. 2012;100:11–465. Cancer. 1999;35:2031–2038.
syndrome. Cell Metab. 2015;22:516–530. 90. Bustaffa E, Stoccoro A, Biachi F, Migliore L. 106. Lippman SM, Hong WK. Cancer prevention by
75. Borges-Canha M, Portela-Cidade JP, Dinis-Ribeiro Genotoxic and epigenetic mechanisms in arsenic delay (commentary). Clin Cancer Res. 2002;8:305–
M, Leite-Moreira AF, Pimental-Nunes P. Role of carcinogenicity. Arch Toxicol. 2004;88:1043–1067. 313.
colonic microbiota in colorectal carcinogenesis: a 91. IARC Working Group on the Evaluation of Carci- 107. Kelloff GJ, Lippman SM, Dannenberg AJ, et al.
systematic review. Rev Esp Enferm Dig. 2015;107: nogenic Risks to Humans. Chromium, nickel and Progress in chemoprevention drug development: the
659–671. welding. IARC Monogr Eval Carcinog Risks Hum. promise of molecular biomarkers for prevention of
76. Jobin C. Colorectal cancer: looking for answers in 1990;49:1–648. intraepithelial neoplasia and cancer—a plan to move
the microbiota. Cancer Discov. 2013;3:384–387. 92. Lu H, Shi X, Costa M, Huang C. Carcinogenic forward. Clin Cancer Res. 2006;12:3661–3697.
77. Pevsner-Fischer M, Tuganbaev T, Meijer M, et al. effect of nickel compounds. Mol Cell Biochem. 108. Colditz GS, Wolin KY, Gehlert S. Applying what
Role of the microbiome in non-gastrointestinal 2005;279:45–67. we know to accelerate cancer prevention. Sci Transl
cancers. World J Clin Oncol. 2016;73:200–213. 93. O’Reilly KM, McLaughlin AM, Beckett WS, Sime Med. 2012;4(127):127rv4.
78. Schwabe RF, Jobin C. The microbiome and cancer. PJ. Asbestos-related lung disease. Am Fam Physician. 109. Blackburn EH. Cancer interception. Cancer Prev
Nat Rev Cancer. 2013;13:800–812. 2007;75:683–688. Res (Phila). 2011;4:787–792.
11  DNA Damage Response Pathways
and Cancer
James M. Ford and Michael B. Kastan

S UMMARY OF K EY P OI N T S
• DNA repair and the cellular response • Inherited defects in base excision mutations of the BRCA1 and BRCA2
to DNA damage are critical for repair can result in colorectal cancer genes, exhibit defects in multiple
maintaining genomic stability. susceptibility. DNA repair and DNA damage
• Defects in DNA repair or the • Inherited defects in mismatch repair response pathways.
response to DNA damage result in Lynch syndrome (hereditary • Because most cancer therapeutic
encountered from endogenous or nonpolyposis colorectal cancer agents that are currently used
external sources results in an syndrome), leading to increased damage DNA, understanding how
increased rate of genetic mutations, incidence of gastrointestinal cancers, normal cells and tumor cells respond
often leading to the development of endometrial cancer, and other to and repair DNA damage is an
cancer. malignancies. important aspect of understanding
• Inherited mutations in DNA damage • Biallelic inherited defects in DNA cancer therapeutic responses and
response pathway genes often result double-strand break repair and toxicities of treatment.
in cancer susceptibility. response pathways underlie a • The presence of mutations in DNA
• The major active pathways for number of rare, recessive damage response and repair
DNA repair in humans are nucleotide cancer-prone disorders, including pathways in tumors presents
excision repair, base excision ataxia-telangiectasia, Nijmegen opportunities for developing
repair, mismatch DNA repair, breakage syndrome, Bloom therapeutics that target alternative
translesional DNA synthesis, and syndrome, Werner syndrome, damage response pathways and can
homologous recombination or Rothmund-Thomson syndrome, and be selectively lethal to the mutated
nonhomologous end joining Fanconi anemia. tumor cells, an approach called
processes for double-strand break • Persons with autosomal dominant synthetic lethality.
repair. Li-Fraumeni syndrome, who are • Genomic instability, particularly that
• Inherited defects in nucleotide highly prone to cancer because of conferred by underlying DNA repair
excision repair lead to the skin inherited monoallelic p53 mutations, defects, can lead to enhanced
cancer–prone syndrome xeroderma and with breast-ovarian cancer responses to immune checkpoint
pigmentosum, Cockayne syndrome, syndrome, who are highly prone to inhibitors owing to increased
and trichothiodystrophy. cancer because of inherited expression of mutant neoantigens.

Cancer is a disease caused by the accumulation over time of changes human tumorigenesis remains a controversial issue,6,7 but cellular
to the normal DNA sequence resulting in alterations, loss, amplification, responses to DNA damage are clearly important determinants of both
or changes in expression of genes important for normal cellular functions cancer development and outcomes following cancer treatments.
and growth properties, including proto-oncogenes and tumor suppressor Cancer cells must be able to tolerate increased amounts of unrepaired
genes. Nearly all cancers are clonal in origin—that is, they originate DNA damage associated with genomic instability, and therefore
from a single progenitor cell rather than a group of cells. The develop- inactivate DNA damage–inducible signaling and checkpoint pathways.4
ment of cancer in a particular cell type or tissue is caused by a series Therefore, DNA repair and the DNA damage response are essential
of specific mutations, each of which could be caused by DNA replication not only for the basic processes of transcription and replication necessary
errors or unrepaired endogenous or exogenous DNA damage or could for cellular survival, but also for maintaining genomic stability and
be the result of inherited mutations. For the most common cancers, avoiding the development of malignancies. This chapter reviews the
multiple genetic events occur in many different genes during the major DNA repair mechanisms active in mammalian cells, and our
process of carcinogenesis, suggesting that an early and perhaps necessary understanding of DNA damage signaling pathways that integrate with
event in the cancer process is an underlying defect in mechanisms to other cellular processes regulating transcription, replication, cell division
maintain genomic stability.1–4 In fact, alterations in the specific genes and apoptosis in response to DNA damage. The relevance of these
required for recognizing, processing, and responding to DNA damage mechanisms to cancer is explored by focusing on several human cancer
may result in an enhanced rate of accumulation of additional mutations, predisposition syndromes caused by underlying defects in DNA damage
recombinational events, chromosomal abnormalities, and gene amplifica- processing. Dysregulation of the DNA damage response is also associated
tion.5 Whether such a “mutator phenotype” is a required event for with increased sensitivity or resistance of tumors to various classes of

154
DNA Damage Response Pathways and Cancer  •  CHAPTER 11 155

oxygen species), whereas others, including ionizing radiation and


Table 11.1  Human DNA Repair Pathways ultraviolet (UV) light, are threats from the external environment (Fig.
Pathway Type of Approximate 11.1). One pronounced example is exposure to genotoxic compounds
DNA Repair DNA Damage No. of Genes in cigarette smoke, which currently are responsible for the most common
cancers in Western countries. Most active chemotherapeutic agents
Nucleotide Bulky or helix-distorting 37 function by damaging DNA through alkylation, cross-linking, and
excision repair DNA adducts, for example, other means, and mechanisms to repair these lesions determine the
ultraviolet photoproducts sensitivity and resultant resistance of a tumor to such treatments.
and carcinogen adducts
Damage to DNA can cause genetic mutations, and these mutations
Base excision Oxidative DNA damage; 40 can lead to the development of cancer. A complex set of cellular surveil-
repair spontaneous depurination lance and repair mechanisms has evolved to reverse or limit potentially
Mismatch repair Mispaired nucleotides 26 deleterious DNA damage. Some of these DNA repair systems are so
1–15 nucleotide important that life cannot be sustained without them. An increasing
insertion-deletion loops number of human hereditary diseases that are characterized by severe
Homologous Double-strand DNA 20 developmental problems or a predisposition to cancer have been linked
recombination breaks, DNA cross-links to deficiencies in DNA repair (see Table 11.2).
Nonhomologous Double-strand DNA 10
end joining breaks CONSEQUENCES OF DNA DAMAGE
The results of DNA damage are diverse and usually adverse. Acute
effects arise from disturbed DNA metabolism, triggering cell cycle
arrest or cell death. Long-term effects result from irreversible mutations
therapeutics, and can be further exploited. Finally, new opportunities contributing to oncogenesis and inherited genetic disorders. Many
for cancer treatment will be discussed based on targeting DNA repair lesions block transcription, which has elicited the development of a
or DNA repair deficiencies and genomic instability. dedicated repair system, transcription-coupled repair (TCR), which
Studies of cancer genetics have defined three general groups of displaces or removes the stalled RNA polymerase and ensures preferential
genes involved in the development of human cancers: oncogenes, repair of lesions within the transcribed strand of expressed genes.11–13
tumor suppressor genes, and DNA damage repair and response genes. Transcriptional stress due to DNA lesions that block RNA polymerase,
The latter set of genes is particularly important to hereditary cancer and DNA strand breaks caused by DNA damage or stalled replication
susceptibility owing to their direct involvement in maintaining genomic forks, constitute two major signals for DNA damage–inducible
stability. Much of what we know regarding cancer genes in sporadic responses, including apoptosis,14–16 through both p53-dependent and
tumors comes from the study of relatively rare inherited cancer p53-independent mechanisms.17
syndromes caused by mutations passed along in the germline DNA Lesions also may interfere with DNA replication. A class of more
of families and predisposing to the development of cancers, often at than 17 specialized DNA polymerases that are devoted to overcoming
a very young age and at a high incidence, in affected carriers. Individuals damage-induced replication stress has been discovered.18–20 These special
who inherit a germline mutation in genes involved in or required for polymerases take over temporarily from the stalled replicative DNA
DNA repair usually are at increased risk for the development of cancer polymerases. Although translesion polymerases protect the genome,
because of the enhanced frequency of mutations and increased genomic this solution to replication blocks comes at the expense of a higher
instability. Susceptibility to cancer may also be affected by environmental replicative error rate, and mutations in some of these polymerases
factors and multiple low-penetrance modifier genes. cause cancer susceptibility.10 Therefore, detection of DNA lesions may
Many converging lines of experimental evidence reveal the complex- occur by blocked transcription, replication, or specialized sensors.
ity of the cellular responses to DNA damage and their role in malignant
transformation.8 A number of interrelated biochemical pathways exist DNA DAMAGE RESPONSE PATHWAYS
that influence the following actions: (1) the metabolism of potentially
mutagenic or carcinogenic agents, (2) the efficiency and manner by DNA damage checkpoints initially were defined as regulatory pathways
which damaged DNA is recognized and repaired, (3) cell cycle progres- that control the ability of cells to arrest the cell cycle in response to
sion and the coordination of DNA replication and cell division relative DNA damage, perhaps allowing time for repair or other cellular
to the repair of lesions, and (4) the decision point determining survival functions.21 However, in addition to controlling cell cycle arrest, proteins
or the active induction of programmed death of cells carrying different involved in these pathways have been shown to control the activation
types and amounts of DNA damage. Many cellular pathways have of DNA repair pathways,22–26 the movement of DNA repair proteins
evolved that require hundreds of gene products for the repair of DNA to sites of DNA damage,27–32 and activation of transcriptional
damage, and involve excision of damaged DNA bases, joining of responses.33–35 When damage is too significant, a cell may opt for the
broken DNA strands, or replication bypass of DNA lesions (Table ultimate mode of rescue by initiating apoptosis to benefit the organism
11.1). The central role of DNA damage responses in neoplastic at the expense of losing a cell.36–38 As the DNA damage response
transformation has been highlighted by the discovery that mutations pathway has been better defined at a molecular level, it has been seen
in numerous classes of genes required for DNA repair and the as a network of interacting pathways that together execute the response.4
maintenance of genomic integrity result in a predisposition to the Initial recognition of DNA damage occurs by a variety of damage-
development of many malignancies.9 In fact, a number of rare, inherited specific DNA binding proteins that, either by themselves or together
disorders have been described that appear to be caused by defects in with complexes of associated proteins not directly involved in DNA
the repair of DNA lesions (Table 11.2), and many of these are associated repair, signal the DNA damage response.39 Initiation of DNA damage
with an increased risk of the development of cancers.10 signaling pathways often is carried out by the phosphoinositide-3-kinase-
related proteins which include ataxia-telangiectasia mutated (ATM)
TYPES OF DNA DAMAGE and ATM-Rad3-related (ATR) proteins, DNA-PK, the checkpoint
kinases CHEK1 and CHEK2, and others.8,40–45 ATM plays a critical
DNA undergoes many types of spontaneous modifications, and it role in DNA damage signaling originating at double-strand breaks
also can react with physical and chemical agents, some of which are (DSBs), whereas ATR responds to single-strand DNA regions. Many
endogenous products of normal cellular metabolism (e.g., reactive of these protein kinases are themselves targets for phosphorylation
156 Part I: Science and Clinical Oncology

Table 11.2  Human Genetic Diseases Involving Defects in DNA Damage Response Pathways
Syndrome Gene(s) Biologic Functions Clinical Features Hypersensitivities
Xeroderma pigmentosum XPA-XPG Nucleotide excision repair Sunlight hypersensitivity UVR, chemical carcinogens
XPV Translesional DNA synthesis Neurologic defects; skin cancers
Cockayne syndrome CSA, CSB Transcription-coupled repair Growth retardation UVR, chemical carcinogens
XPB, XPD Mental retardation
Reactive oxygen species
XPG Premature aging; sunlight
hypersensitivity
Trichothiodystrophy XPB, XPD, Nucleotide excision repair; Sulfur-deficient brittle hair; dry, UVR
TTDA transcription scaly skin; mental and physical
retardation; sunlight sensitivity
Hereditary nonpolyposis MLH1, MSH2, Mismatch repair 6-Thioguanine and cisplatin
colorectal cancer (Lynch MSH6, PMS2 resistance. Colorectal, endometrial,
syndrome) gastric, bile duct cancers
Ataxia-telangiectasia ATM DNA damage-responsive Cerebellar ataxia; telangiectasia; Ionizing radiation
kinase immunodeficiency; lymphomas
Ataxia-telangiectasia–like MRE11 Double-strand break repair Similar to ataxia-telangiectasia
disease
Nijmegen breakage NBS1 Double-strand break repair Microcephaly; immunodeficiency; Ionizing radiation
syndrome lymphomas; neuroblastoma;
rhabdomyosarcoma
Bloom syndrome BLM DNA helicase; homologous Sunlight hypersensitivity; growth UVR, hydroxyurea
recombination at stalled retardation; leukemias, lymphomas;
replication forks? breast and intestinal cancers
Werner syndrome WRN DNA helicase; homologous Premature aging; atherosclerosis; 4-NQO, camptothecin;
recombination?; translesional soft tissue sarcomas; melanoma, hydroxyurea
synthesis? thyroid cancer
Rothmund-Thomson RECQL4 DNA helicase Growth deficiency; sunlight UVR
syndrome sensitivity; osteogenic sarcomas;
squamous cell carcinomas
Fanconi anemia FANCA–G Interstrand cross-link repair Growth retardation Bifunctional alkylating agents
BRCA2 Homologous recombination Anatomic defects; bone marrow Ionizing radiation
failure; myeloid leukemia;
squamous cell cancers
Li-Fraumeni syndrome p53 Apoptosis; cell cycle Breast cancer; brain cancers; ?
checkpoints; nucleotide adrenocortical carcinoma; leukemia;
excision repair bone and soft tissue sarcomas
Li-Fraumeni–like syndrome CHEK2 DNA damage responsive kinase Similar to Li-Fraumeni syndrome
Breast-ovarian cancer BRCA1, Double-strand break repair Breast cancer, ovarian cancer Ionizing radiation, cisplatin,
syndrome BRCA2 PARP inhibitors

PARP, Poly (ADP-ribose) polymerase; UVR, ultraviolet radiation.

and activation; they then further target downstream gene products at any level of these pathways can alter repair and result in carcinogenesis
critical to oncogenesis such as p53 and BRCA1.40,46–48 The ultimate (Fig. 11.2).
targets of this highly regulated DNA damage response include
mechanisms for DNA repair, and although much of DNA repair is TYPES OF DNA REPAIR AND THEIR
constitutive, a number of regulatory connections between the DNA CONTRIBUTION TO CANCER
damage response pathway and DNA repair have emerged.8 In mammals,
a large number of genes involved in DNA repair are transcriptionally DNA repair may be defined as those cellular responses associated with the
upregulated in response to DNA damage, suggesting that many facets restoration of the normal nucleotide sequence after events that damage
of repair are inducible. In fact, the p53 tumor suppressor gene is a or alter the genome.50 Given the wide variety of DNA damage a cell
central mediator of the DNA damage–inducible transcriptional response encounters, it is not surprising that there is an equally large number of
in humans, and p53-mutant mammalian cells are deficient in several repair systems available to handle these insults. Indeed, many are broadly
aspects of DNA repair.22–26,35,49 Therefore the mammalian DNA overlapping and interacting, with several sharing certain strategies and
damage–inducible response pathway is highly regulated, and fine-tuned even specific gene products. In humans, a great deal has been learned
to determine if a particular cell type proceeds to a cell cycle checkpoint regarding DNA repair from the often rare, autosomal recessive hereditary
and DNA repair, or cell death, after a significant damage insult. Defects syndromes associated with defects in DNA repair genes.10
DNA Damage Response Pathways and Cancer  •  CHAPTER 11 157

DNA damage type Endogenous Environmental


Spontaneous base changes Chemical mutagens
Replication errors Cytotoxic agents
Oxygen radicals UV and ionizing radiation

DNA lesions Base-pair mismatches Abasic sites DNA adducts Pyrimidine dimers Strand breaks
Insertion and deletions Oxidized DNA Cross-links
Strand breaks

Signaling pathways Protein kinases


p53 BRCA1 BRCA2 Fanconi anemia
ATM, ATR, Chk1, Chk2 complex

DNA repair pathways Mismatch Base excision Nucleotide excision Double-strand


repair repair repair break repair

Cellular responses to Cell cycle Upregulated Apoptosis Translesional


persisting damage checkpoints repair synthesis

Mutation
Consequences Cell survival Cell death
Malignant transformation

Figure 11.1  •  Cellular responses to DNA damage. Different types of DNA damage cause a variety of different types of lesions, and these in turn are dealt
with by a variety of DNA repair mechanisms and signal various cellular response pathways. The outcome of DNA damage may be cell survival of a normal
cell, cell death, or mutagenesis, possibly leading toward malignant transformation.

Nucleotide Excision Repair A unique problem arises if a bulky lesion is encountered by a


translocating RNA polymerase making messenger RNA before repair
The most versatile and ubiquitous mechanisms for DNA repair are enzymes have removed the damage and restored intact DNA. The
those in which the damaged or incorrect part of a DNA strand is polymerase may be arrested at the site of the lesion and prevent access
excised and the resulting gap is filled through repair replication with to the damage by repair enzymes. Furthermore, the arrest of transcrip-
use of the complementary strand as a template. The redundancy of tion in human cells is a strong signal for p53 activation and can trigger
genetic information provided by the duplex DNA structure is essential apoptosis.17 In this situation, a dedicated excision repair pathway
to the maintenance of the genome by this “cut and patch” mode known as transcription-coupled repair comes to the rescue to displace
known as excision repair. Each DNA strand can serve as a template the RNA polymerase, and then efficiently repairs the blocking lesion
for replication-based repair of the other strand. Nucleotide excision so that transcription may resume and the cell may survive.55
repair (NER) functions to remove many types of lesions, including It has become apparent that the GGR subpathway of NER is
bulky base adducts of chemical carcinogens, intrastrand cross-links damage inducible and highly regulated by both transcriptional and
(ICLs), and UV-induced cyclobutane pyrimidine dimers (CPDs) and posttranslational mechanisms after DNA damage, in concert with
6-4 photoproducts.51 Such lesions may serve as structural blocks to damage-inducible cell cycle checkpoints and apoptosis.8,49 In fact, the
transcription and replication due to distortion of the helical conforma- p53 gene, central to maintaining genomic stability in human cells, is
tion of DNA, and they also may result in mutations if translesional required for efficient GGR of UV light– and carcinogen–induced
replication occurs or if they are not repaired correctly. The sequential DNA damage, and functions as a DNA damage–activated transcription
steps for NER are (1) recognition of the damaged site, (2) incision factor that directly regulates the expression of several NER damage
of the damaged DNA strand near the site of the defect, (3) removal recognition genes.24–26,35 Similarly, several other important cancer-related
of a stretch of the affected strand containing the lesion, (4) repair genes have been shown to transcriptionally regulate the DNA damage
replication to replace the excised region with a corresponding stretch recognition NER genes XPC and DDB2, including BRCA1 and
of normal nucleotides with use of the complementary strand as a E2F1.56–58 Therefore the GGR pathway of NER appears relevant to
template, and (5) ligation to join the repair patch at its 3′ end to the suppressing DNA damage–induced malignancy, and highly regulated
contiguous parental DNA strand.52,53 This excision repair pathway by genes involved in tumor suppression.
can remove DNA damage from sites throughout the genome and is Despite the plethora of mechanisms for DNA repair described
termed global genomic repair (GGR). The majority of human NER herein, it is likely that the cellular replication machinery will nevertheless
genes have been identified and cloned, and many have been shown to encounter lesions in the DNA template strand that block replication
be mutated in hereditary NER-deficient, cancer-prone diseases.26,49,54 during each cell cycle. The solution adapted by cells is DNA damage
158 Part I: Science and Clinical Oncology

that the cancer-prone hereditary disease XP involved a defect in the


Structure distortion 3' 5'
repair of DNA lesions produced by UV light.63 Since then, at least
four syndromes have been attributed to inborn errors in NER: XP,
Cockayne syndrome (CS), trichothiodystrophy (TTD), and ultraviolet-
Damage recognition XPC/hHR23B XPE(p48/p127) sensitive syndrome (UVSS), all characterized by exquisite sun sensitivity.
XP is a rare, autosomal recessive disease in which homozygous
individuals display several characteristics: (1) extreme sensitivity of
the skin to sun exposure evident by 1 year of age, (2) pigmentation
RPA XPA abnormalities and premalignant lesions in sun-exposed skin, (3) increases
up to 4000-fold in incidence of skin cancers (predominantly squamous
and basal cell carcinomas, but also melanomas) and ocular neoplasms,
Incision occurring three to five decades earlier than in the general population,
TFIIH
ERCC1 and (4) a 10- to 20-fold increased incidence of internal cancers in
XPG XPF
non–sun-exposed sites.10,64,65 Overall, lifespan is reduced by approxi-
mately 30 years in patients with XP, and many die due to malignancies.66
Approximately 20% of patients with XP also display progressive
RPA XPA neurologic degeneration, characterized by peripheral neuropathy,
sensorineural deafness, progressive mental retardation, and cerebellar
Excision and pyramidal tract involvement.67 XP occurs worldwide, in all ethnic
groups, and with a frequency varying from 1 to 10 patients per million.
27-29 n The biochemical defect in cells from most individuals with XP is
in NER,63 although in a small number of cases (termed XP-variants),
excision repair appears normal, and a defect exists in bypass replication
Repair replication RFC at unrepaired lesions as a result of a mutation in the pol η TLS gene
PCNA PoI (XPV).68 Complementation analysis via fusion of cells from different
RPA
patients has demonstrated genetic heterogeneity within XP and provided
evidence for the existence of at least seven excision-deficient comple-
mentation groups, termed XP-A to XP-G, in addition to XP-variant.10
Rejoining Ligase I CS is an autosomal recessive disease that is associated with defec-
tive TCR of UV-damaged and oxidative-damaged DNA.69–71 It is
characterized by cutaneous photosensitivity, cachectic dwarfism,
skeletal abnormalities, retinal degeneration, cataracts, severe mental
retardation, and neurologic degeneration characterized by primary
Figure 11.2  •  Mechanism for human nucleotide excision repair (NER). demyelination.67,72 In contrast to patients with XP, those with CS
Ultraviolet irradiation–induced adducts in genomic DNA are recognized by are not at increased risk for developing skin cancers. The average
the XPE and XPC/hHR23B protein heteroduplexes that recruit the XPA/ lifespan of individuals with CS is only 12 years, with most patients
RPA complex and the larger TFIIH protein complex. Dimers that occur in dying from infectious or renal complications rather than cancer.73 CS
the transcribed strand of an expressed gene result in a blocked RNA polymerase is characterized by the existence of at least three complementation
II molecule, which, together with the CSA and CSB gene products, serves to groups. Several patients have been described in XP groups B, D,
recruit the downstream repair machinery. The TFIIH complex contains helicases and G who share the DNA repair defects and clinical features of CS
including XPB and XPD that unwind the DNA and allow the other repair together with the cutaneous manifestations of XP.74,75 One model for
proteins access for incision and excision of the damaged DNA oligonucleotide. the severe developmental problems in CS is that endogenous oxidative
After excision, repair replication based on the normal DNA template and
ligation of the newly synthesized DNA sequence occurs. In total, more than
damage in metabolically active cells, including neurons, is blocking
25 proteins participate in NER. transcription. The lack of TCR could then lead to apoptosis of these
essential cells. Alternatively, CS could be a “transcription disease”
caused by defects in TFIIH-associated gene products and resulting
in inadequate gene expression.
tolerance, in which DNA is synthesized past the damaged bases and TTD is an autosomal recessive condition sharing many of the
subsequently excised after the replication fork has passed—a process signs and symptoms of CS, with the additional hallmark of brittle,
known as translesional synthesis (TLS) and carried out by a class of sulfur-deficient hair and nails, due to reduced cysteine content in the
specialized error-prone DNA polymerases.18,19,59,60 These Y-family DNA component proteins. As with CS, several of the responsible genes
polymerases take over temporarily from the stalled replicative DNA implicated are XPB and XPD, but there is a third complementation
polymerases. They have more flexible base-pairing properties that group, TTD-A, in which mutations of a small 8-kDa TFIIH stabilizing
permit translesion DNA synthesis, with each polymerase probably factor GRF2H5 has been identified.76
designed for a specific category of injury. Although translesion Analysis of the specific abnormalities in NER displayed by the
polymerases protect the genome, this solution to replication blocks various genetic complementation groups of XP, CS, TTD, and UVSS
comes at the expense of a higher error rate. For instance, inherited allow correlations to be drawn with their heterogeneous clinical features.
defects in polymerase eta (pol η), encoded for by the XPV/POLH/ Specifically, only those subgroups of patients who display a defect in
RAD30 gene, which specializes in relatively error-free bypassing of GGR are at significantly increased risk for developing UV-induced
UV-induced CPDs, cause a variant form of the skin cancer–prone malignancies. In contrast, the neurologic symptoms and developmental
disorder Xeroderma pigmentosum (XP).61,62 abnormalities associated with other complementation groups of XP
and CS are found only in those groups that are defective in TCR.
Human Nucleotide Excision Repair–Deficient The fact that the TFIIH complex, containing the XPB and XPD
Syndromes and Cancer proteins, is common to both core NER and transcriptional initiation,
supports the suggestion that the clinical phenotype of patients with
A direct correlation between unrepaired DNA damage and carcino- defects in TCR may actually be due to abnormalities in transcription
genesis in humans was first established when James Cleaver found rather than in repair itself.77
DNA Damage Response Pathways and Cancer  •  CHAPTER 11 159

Although these observations may explain the molecular basis for single-strand DNA break intermediate. In some cases, bifunctional
many of the clinical characteristics of XP and CS, they present an glycosylases, which also cleave the phosphodiester bond adjacent to
apparent paradox with regard to these patients’ cancer risk. Many the damaged base, preclude the need for endonuclease activity of
currently recognized oncogenes and tumor suppressor genes are known APE1 and instead require polynucleotide kinase 3′-phosphatase for
to possess important cellular functions and to be actively expressed single-strand break end-processing. In the short-patch pathway, DNA
in normal cells. Because CS cells are defective in the repair of actively polymerase (POLB) displaces the AP site and adds a nucleotide. Last,
expressed genes, it would be reasonable to expect that these patients ligase (LIG3) forms a phosphodiester bond to complete repair. In the
would acquire mutations in genes leading to transformation more long-patch pathway, DNA polymerase (POLB, POLD, or POLE)
readily than normal patients. However, this is not supported by the displaces and adds more than one nucleotide, flap structure-specific
clinical picture. It has been demonstrated that defects in TCR specifically endonuclease (FEN1) removes the displaced nucleotides, and ligase
activate DNA damage–induced apoptosis,14,15 which may eliminate (LIG1) completes repair. Proliferating cell nuclear antigen (PCNA)
potentially mutagenic, premalignant cells. is required for POLD function and acts as a scaffolding protein. Poly
(ADP-ribose) polymerase (PARP) binds to and recruits essential media-
Base Excision Repair tors to single-strand break intermediates. The major oxidized purine
lesion is 8-oxo-7,8-dihydroguanine (8-oxoG), which is abundant and
A major source of DNA damage to cellular genomes arises from normal has strong mutagenic properties. Oxidized pyrimidines include thymine
metabolism in the cytoplasmic environment through hydrolysis and glycol, 5-hydroxycytosine, and formamidopyrimidines. Oxidized bases,
exposure to reactive metabolites that cause oxidation and alkylation of including both 8-oxoG and thymine glycol, share the property of
DNA. The repair system primarily involved in identifying and removing blocking DNA replication and transcription, and must be repaired
such lesions, as well as in dealing with the spontaneous loss of purines efficiently if genomic stability is to be maintained.84,85
from DNA, is the base excision repair (BER) pathway.78,79 The essential In mammalian cells, the gene functions responsible for the strand
nature of BER for viability is highlighted by the fact that although a incision steps of BER include the glycosylases hNTH1, which removes
number of BER proteins have been discovered, only recently has a single oxidized pyrimidines; hOGG1, which targets oxidized purines; and
human hereditary disease been identified that appears to result from MYH, which removes adenines mispaired with an 8-oxoG, together
a mutation in a gene unique to this pathway.80–83 BER is initiated by with APE1.86 Given the mutagenic and cytotoxic potential of the
one of a set of lesion-specific glycosylases that recognize the altered or classes of DNA damage that are BER substrates, it seems likely that
inappropriate base and cleave it from its sugar moiety in the DNA (Fig. altered activity in these pathways would result in enhanced cancer
11.3). Different DNA glycosylases remove different kinds of damage, risk. The most direct evidence for a role for BER in cancer comes
conferring specificity to the process. AP-endonuclease (APE1) then from the discovery that germline mutations in the MYH gene, involved
cleaves the phosphodiester backbone 5′ to the AP site, resulting in a in processing 8-oxoG lesions, is associated with recessive inheritance
of a predisposition to develop multiple colorectal adenomas (polyposis)
and colon cancers.80–82 Tumors from affected individuals exhibit excess
transversions of a guanine-cytosine pair to a thymine-adenine pair in
BER NER MMR the APC gene, itself associated with colon carcinogenesis and causative
of familial adenomatous polyposis. Therefore, biallelic inherited
mutations in MYH result in a polyposis-like syndrome termed MYH-
Base damage Pyrimidine dimer Mismatch associated polyposis (MAP). Patients with MAP tend to develop tens
to hundreds of polyps by the age of 40, and nearly 50% have colon
cancer at presentation.83,87
A great deal of effort has gone into attempts to identify functional
polymorphic genetic variation in DNA repair genes that modulate
Recognition, incision, excision activity and affect individual cancer risk. To date, genetic epidemiology
studies of single-nucleotide polymorphisms (SNPs), particularly in
BER genes, have been inconsistent.88 However, meta-analyses have
identified modest associated increased lung cancer risk with OGG1
and XRCC1 SNPs.89 Continued advances in SNP maps and high-
Repair patch synthesis throughput genotyping methods will facilitate the analysis of multiple
polymorphisms within multiple genes through use of haplotypes;
with larger sample sizes, perhaps future studies will reveal clearer
risk associations.

Ligation Mismatch Repair


Mismatch repair (MMR) is another example of an excision repair
mechanism that uses a similar strategy for genomic maintenance (see
Fig. 11.3). MMR is a process that corrects mismatched nucleotides
Figure 11.3  •  Excision repair pathways for DNA damage. The three main in the otherwise complementary paired DNA strands, arising from
excision repair pathways in human cells—base excision repair (BER), nucleotide DNA replication errors and recombination, as well as from some
excision repair (NER), and mismatch repair (MMR)—proceed through similar types of base modifications.90–92 This repair mode also can deal with
steps to restore the normal DNA sequence. After recognition of altered DNA small loops of single-stranded DNA at sites of insertions or deletions
bases, using lesion-specific glycosylases for BER, XP proteins for NER, and in the duplex DNA structure. The importance of this repair mechanism
MutS homologs for MMR, incision of DNA is achieved by endonucleases, in maintaining genetic stability is illustrated by the observation that
and displacement or degradation of single-stranded sections of DNA that
contain the damage by enzymes with helicase and exonuclease activity. Repair its absence results in a large increase in the frequency of spontaneously
replication of the resulting DNA gap and strand ligation results in the repaired occurring mutations, particularly in microsatellite sequences of highly
double-stranded DNA molecule. The many repair enzymes involved in each repetitive DNA.93 Some of these spontaneous mutations arise from
specific step are tightly coupled and may be regulated or inducible by DNA mistakes introduced during DNA replication, in spite of the operation
damage response pathways. of a “proofreading” system that also helps to ensure the high fidelity
160 Part I: Science and Clinical Oncology

of replication. In humans, genetic defects in several MMR genes have


been linked to Lynch syndrome (hereditary nonpolyposis colorectal Box 11.1.  GENOMIC INSTABILITY AND
cancer) and to sporadic cancers that exhibit instability in regions of RESPONSE TO IMMUNE CHECKPOINT
DNA containing short repetitive sequences of nucleotides, a feature BLOCKADE
known as microsatellite instability (MSI).
As with other modes of excision repair, four principal steps are One of the most exciting advances in cancer therapeutics has been the
required for MMR: (1) mismatch recognition, (2) recruitment of use of immune checkpoint inhibitors in a wide range of tumors,
additional MMR factors, (3) identification of the newly synthesized including classically “immunogenic” cancers such as melanoma and
DNA strand containing the mismatched nucleotides, followed by renal cell cancer,111 but also many others. For most cancers, predictive
their excision, and (4) resynthesis of the excised tract and ligation. biomarkers to, for example, anti-PD-(L)1 agents remain poorly
The biochemical workings of this pathway are best understood in understood. However, it has recently been appreciated that the overall
bacteria, but a similar set of events occurs in human cells. Based on tumor mutational burden correlates with clinical benefit of anti-PD-1
functional homologies to their bacterial counterparts, and sequence and anti-CTLA4 agents, particularly for patients with conditions
homology to corresponding yeast genes, a number of human genes involving high somatic mutation rates such as non–small cell lung
have been cloned that participate in MMR, including those homologous cancer and melanoma and those with DNA repair deficiencies.112 It has
to the bacterial MutS mismatch recognition protein (hMSH2, hMSH3, been postulated that the load of neoantigens resulting from DNA
and hMSH6) and to the bacterial MutL gene (hMLH1 and hPMS2).94–96 mutations in coding regions leads to immunotherapy responses and
In humans, heterodimers of the MSH2/6 proteins recognize single may be a biomarker.113 A survey of mutational load in various tumors
base-pair mismatches and short insertion-deletion loops, whereas showed that among the very highest are microsatellite instability–high
MSH2/3 dimers recognize longer loops. Heterodimeric complexes of (MSI-H) colorectal cancers and POLE mutant hypermutator colorectal
MLH1/PMS2 and MLH1/PMS1 interact with the MSH complexes and endometrial cancers.114 This is consistent with the long-standing
and replication factors, for strand discrimination and DNA excision. observation that Lynch syndrome tumors exhibit a high level of
Similar to NER and BER, additional proteins are then recruited for tumor-infiltrating lymphocytes (TILs).115 A remarkable clinical study
repair replication based on the original DNA template. showed that DNA mismatch repair–deficient MSI-H colorectal and
The MMR system also may interact with DNA damage because of endometrial cancers achieve excellent and durable responses to
certain alkylators and intercalating agents that assume similar structural immune checkpoint inhibitors, whereas microsatellite-stable (MSS)
alterations in DNA as mismatches and actually result in erroneous or tumors showed no responses.116 Immunohistochemistry showed
futile MMR cycles and ultimately in apoptosis. Thus, intact MMR greater densities of CD8+ TILs and PD-L1 expression in MSI-H than MSS
may confer chemosensitivity to these chemotherapeutic agents, and tumors in this study, and these were associated with responses. Based
MMR-defective tumors may exhibit resistance to certain drugs.97 on these results, the checkpoint inhibitor pembrolizumab received
breakthrough therapy designation for MSI-H advanced colorectal
cancers. Trials examining this approach for adjuvant treatment of early
Human Mismatch Repair Deficiency and Cancer colorectal cancer and any cancers with an MSI phenotype are
Lynch syndrome is the most common hereditary colorectal cancer underway. Ultimately, there appears to be a clear link among DNA
predisposition syndrome.98,99 Lynch syndrome is an autosomal domi- repair deficiency, mutational landscape, predicted neoantigen load, and
nant inherited condition with an incidence of 1 : 1000 in the general clinical activity of immune checkpoint inhibitors. The role of somatic
population. It accounts for approximately 3% of all colorectal cancers tumor mutational load and spectrum in priming the immune system in
patients; patients with Lynch syndrome are also at elevated risk for tumors is a fascinating new approach to linking personalized cancer
cancers of the endometrium, ovary, stomach, small bowel, and other genomic profiling and immunotherapy.
sites. Patients with Lynch syndrome have an 80% lifetime risk for
colorectal cancer and a 50% lifetime risk for endometrial cancer. The
discovery of MSI—that is, the frequent alteration in the tract lengths
of certain short repetitive nucleotide sequences—in some hereditary associated with defects in MMR results in the genotype of tumors
colorectal cancers provided the first indication that the etiology of these that arise because of these defects.104 Intriguingly, the survival of patients
cancers might involve a problem in the MMR system.100 The finding with MSI-associated colorectal cancer is better than in those with
of germline MMR gene defects in patients with Lynch syndrome more typical tumors exhibiting chromosomal instability.105–107 These
established that these defects are the cause of the enhanced incidence tumors also demonstrate different sources of genomic instability,
of cancer.94–96 Germline mutations in MLH1 and MSH2 together resulting in distinct biologic and pathologic characteristics.108 Whether
account for more than half of all cases of Lynch syndrome. Defects their more favorable outcome reflects differences in clinical behavior,
in MSH6 cause a late-onset Lynch syndrome phenotype, and less responsiveness to therapy, or both remains to be fully determined.
penetrant mutations in PMS2 can also result in Lynch syndrome. No Given the readily available phenotypic assays for Lynch syndrome
strong genotype-phenotype correlations have been observed to date, (MSI and immunohistochemistry for the four causative gene products),
but mutations in the MSH2 gene do appear to be associated with more “universal” screening is now performed on all colon and endometrial
extracolonic manifestations than are mutations in the MLH1 gene. cancers at many centers, leading to cost-effective identification of
MSI has been identified as a source of the genomic instability Lynch syndrome families and hopefully improved clinical outcomes.109,110
driving tumorigenesis in a number of sporadic tumor types, in addition Recently, very interesting findings have been reported regarding
to those that arise in the context of inherited germline mutations of treatment of MSI tumors that targets their genomic instability phe-
MMR genes.100,101 For example, up to 20% of sporadic colon cancers notype (Box 11.1).
exhibit MSI, particularly in the ascending colon and in individuals
younger than 50 years, the majority due to epigenetic silencing of Double-Strand Break Repair
MLH1 gene expression by promoter hypermethylation.102,103 Whether
through genetic or epigenetic inactivation, loss of MMR results in DSBs in DNA induced by ionizing radiation, chemicals, and replication
the elevated rate of mutations, particularly at microsatellite sequences, across single-strand breaks and during repair of interstrand DNA cross-
several of which occur in the coding sequences of other genes often links usually are dealt with through the recombination machinery. An
found mutated in cancers, including TGF beta type II receptor, BAX, unrepaired DSB is a highly lethal event, and even a single occurrence
and the MMR genes MSH3 and MSH6 themselves. Therefore, clear in the entire genome is thought to be sufficient to signal cell cycle
genetic evidence demonstrates that the phenotype of genetic instability checkpoints that prevent attempted DNA synthesis or cell division
DNA Damage Response Pathways and Cancer  •  CHAPTER 11 161

Nonhomologous Homologous
end-joining recombination

DSB DSB

RAD51
MRE11
End alignment Resection
NBS1
Ku80 Ku80 RAD52

DNA-PKcs

DNA-PKcs
Ku70 Ku70 RAD52
BRCA1
XRCC4 RAD51 Strand invasion
BRCA2 Branch migration
Ligase IV
RAD54

RAD51

Ligation
Junction resolution

Figure 11.4  •  Mechanisms for DNA double-strand break (DSB) repair. The repair of DNA DSBs is carried out by two mechanisms. Left, The rapid, but
error-prone, nonhomologous end-joining that directly seals breaks but may result in the gain or loss of several nucleotides owing to short areas of microhomologies
used for annealing before ligation. Exposed DNA ends are recognized by the Ku70/80 heterodimer that recruits the DNA-dependent protein kinase catalytic
subunit and other proteins assisting in strand alignment. The XRCC4-ligase IV heteroduplex joins the breaks. Right, The high-fidelity, homologous recombination
of sister chromatids at sites of DSBs is the less prominent mode in mammalian cells. This DNA repair pathway is mediated by RAD51-associated proteins,
including RAD52, which recognizes single-strand DNA ends and together with other proteins results in short nuclease-mediated resection. RAD51 then forms
a nucleoprotein filament on the exposed strand, and probably with BRCA2 and other proteins, promotes strand invasion and displacement at homologous
sequences. Thus, the undamaged sister molecule acts as a template for the resynthesis of the missing nucleotides.

until repair has been completed, or apoptosis, if improperly repaired. (Nijmegen breakage syndrome), BRCA1 and BRCA2 (breast-ovarian
DSBs also pose problems during mitosis because intact chromosomes cancer syndrome), and the RecQ-like helicases (Werner, Bloom, and
are a prerequisite for proper chromosome segregation during cell divi- Rothmund-Thomson syndromes).124
sion. Thus these lesions often induce various sorts of chromosomal
aberrations, including aneuploidy, deletions, and chromosomal Ataxia-Telangiectasia
translocations—all of which are associated with carcinogenesis. Genetic
recombination is the principal mechanism for dealing with DSBs Ataxia-telangiectasia (AT) is a disease of pleiotropic abnormalities,
that involve homologous stretches of nucleotides at the ends to be including cerebellar degeneration, cancer predisposition, progressive
joined. If no such homology is present, however, another system exists pulmonary dysfunction, and profound radiosensitivity. A single gene,
for nonhomologous end joining (NHEJ), which is more error-prone ATM, is responsible for the multiple and surprisingly diverse symptoms
(Fig. 11.4). Whereas NHEJ is the more predominant mechanism for of this disease. Over 10% of AT patients develop cancer at an early
DSBR in humans, homologous recombination (HR) is specifically age, primarily leukemias and lymphomas. AT is an autosomal recessive
required for repair of DSBs formed at collapsed replication forks, disease with an incidence of nearly 1 in 100,000 live births. Persons
and for ICL repair together with NER proteins. Recent studies have who are heterozygous for AT mutations, about 1% of the general
identified a cascade of protein kinases involved in signaling cellular population, may have an increased predisposition to cancer, in particular
processes in response to DSBs. Many of these have been found to breast cancers, especially in those individuals expressing an ATM with
be defective in cancer-prone disorders exhibiting genomic instability, missense mutations resulting in a dominant-negative effect on their
such as the ATM protein45 and the CHEK2 protein kinase.117–119 A wild-type ATM gene.125–127 The ATM gene product is a central signaling
major target for these kinase activities is the p53 tumor suppressor protein in the DNA damage response, and cells lacking ATM fail to
gene. This protein, when phosphorylated, is activated and involved execute many critical cellular responses to DNA damage. For example,
in G1 arrest and apoptosis after ionizing radiation40,41,120–123 and, the ATM gene product is a key element in delaying the initiation of
when found mutated in the germline, results in the Li-Fraumeni DNA replication after DNA damage resulting in strand breaks. In
cancer susceptibility syndrome. Many other enzymes involved in the fact, lack of ATM function leads to abnormalities in all major DNA
actual DNA transactions necessary for DSB repair have been found damage–induced cell cycle checkpoints, G1, S, and G2/M.41 In addition,
in cancer-prone disorders, including MRE11 (AT-like disorder), NBS1 ATM directly phosphorylates the NBS1 protein, resulting in its
162 Part I: Science and Clinical Oncology

association with the MRE11 and RAD50 gene products, which together BRCA1 and BRCA2, in addition to rare cases caused by mutations
are required for both NHEJ and HR of DSBs.128 Inherited germline in the p53 gene in persons with LFS, the PTEN gene in persons with
mutations of the NBS1 and MRE11 genes, themselves, result in clinical Cowden disease, and carriers with mutations in ATM, CHEK2, PALB2,
variants of AT, termed Nijmegen breakage syndrome and AT-like disorder, and others.144,145
respectively.128,129 Therefore, ATM, NBS1, and MRE11 are central to The BRCA1 and BRCA2 genes are large and complex. Many
DNA damage response pathways critical for regulating recombination hundreds of different germline mutations have been detected in each.
and repair following DSBs. Defects in many of the component proteins Strong evidence suggests that BRCA1 and BRCA2 are centrally involved
in the pathway result in genomic instability and a predisposition to in various aspects of DNA repair and DNA damage response pathways.
cancer. Interesting to note, ATM has also recently been implicated in For example, BRCA1 is phosphorylated after exposure to DNA-
cellular processes other than DNA damage responses, including insulin damaging agents by ATM, ATR, and Chk2; associates with a number
signaling and mitochondrial function.130,131 Thus, although only a of DNA repair proteins including ATM, RAD51, and the RAD50-
single gene is mutated in AT, loss of ATM results in a pleiotropic MRE11-NBS1 protein complex following DNA damage; and localizes
disease phenotype in patients because of its multifaceted functions. to nuclear foci with these proteins after treatment with ionizing
radiation.29,146,147 The association of BRCA1 with RAD51, an enzyme
p53 Gene and Li-Fraumeni Syndrome involved in the coordination of recombination, suggests its involvement
in DSB repair and implicates BRCA1 in HR.148,149 Other studies
The discovery of the p53 tumor suppressor gene over 35 years ago suggest that BRCA1 may regulate cellular processes through tran-
inspired widespread investigations with the aim of understanding the scriptional coactivation. BRCA1 has been shown to transcriptionally
basic biology behind its role in maintaining genomic stability and the regulate the NER genes XPC and DDB2 and affect GGR of UV- and
cellular response to DNA damage. The p53 gene is one of the most cisplatin-induced DNA damage,56,150,151 and to transcriptionally regulate
commonly mutated genes in human cancers,132 and its product is a BER genes and affect BER of oxidative DNA damage151–153 Chromo-
multifunctional protein that regulates many physiologic processes, somal instability also is characteristic of breast tumors that harbor
including cell cycle checkpoints, apoptosis, and DNA repair.26,121,133–135 BRCA2 mutations, probably because of defective recombination-
The primary role of p53 in tumor suppression has been attributed to mediated DSB repair. BRCA2 has been shown to bind to the RAD51
its function as a transcription factor, regulating expression of several protein, an enzyme involved in the coordination of recombination,
hundred different cellular genes,136 although it also exhibits transcription- and together they colocalize to nuclear sites containing DNA strand
independent activities. Indeed, p53 appears to act as a central “node” breaks caused by ionizing radiation. Structural studies of BRCA2
that lies at the intersection of upstream signaling cascades induced DNA binding domains suggest that it may facilitate interactions of
by DNA damage and cellular stress responses, and downstream DNA RAD51 with single-stranded DNA during recombination.154,155 The
repair and DNA damage response pathways. In response to a variety genomic instability associated with mutations of BRCA1 and BRCA2,
of genotoxic stimuli, p53 protein is induced by both increased mRNA therefore, may be due in part to the intact but error-prone NHEJ
translation137,138 and protein stabilization through a series of post- repair pathway.156 See Box 11.2 for a discussion of DNA repair and
translational modifications.136 The induced protein then binds to DNA cancer treatment.
in a sequence-specific manner, resulting in transcriptional regulation
of downstream target genes that contain a consensus p53 response Fanconi Anemia, Cancer, and Interstrand
element in their promoter or intronic segments. These p53 target Cross-link Repair
genes include those important for cell cycle checkpoints, such as p21139;
apoptosis, such as BAX and PERP140; and DNA repair, such as the The centrality and interwoven nature of DNA repair pathways for
DDB2 and XPC genes required for NER.24,25,35 genomic stability have been highlighted by findings regarding Fanconi
Patients with the rare autosomal dominant Li-Fraumeni syndrome anemia, a rare, autosomal recessive disease that confers an increased
(LFS) are at increased risk for developing a number of common tumors risk of acute myeloid leukemia, squamous-cell carcinomas of the head
at an early age because of an inherited germline defect in one allele and neck and esophagus, gynecologic carcinomas, and liver tumors
of the p53 gene, including soft tissue sarcomas and osteosarcomas, at a young age.175,176 At least 19 subtypes of Fanconi anemia have
breast cancer, brain tumors, lymphomas, leukemia, and adrenocortical been determined by complementation analyses, and germline mutations
carcinomas. Mutations in the p53 tumor suppressor gene account for in genes have been identified for most (FANCA to FANCT).177,178,179
70% to 85% of classic LFS cases.141–143 Although the heterozygote The proteins encoded by these genes all cooperate in a common DNA
carriers of a defective p53 allele do not appear to have clinical problems repair pathway involved in interstrand cross-link repair and processing
or somatic DNA repair defects, when the second allele has been mutated of arrested replication forks.180,181 Germline homozygous inactivating
or lost, the absence of functional p53 results in severe problems for mutations of the BRCA2 gene result in the FANCD1 group.182 A
the cell and substantial cancer susceptibility for the host. First, the “core complex” of these proteins consists of subunits of a nuclear E3
p53-controlled pathway of apoptosis is disengaged, so severely damaged ligase, required for the monoubiquitination of the downstream
cells will survive and be at risk for carcinogenic transformation because FANCD2 protein, which itself links Fanconi anemia proteins to BRCA1
of their genomic instability. That genomic instability derives from the in the response to DNA damage by colocalizing to nuclear sites occupied
fact that p53 is also an important regulator of cell cycle checkpoints. by RAD51 and BRCA2.183 It has long been appreciated that cells
Finally, p53 serves an important regulatory function in DNA repair from Fanconi anemia patients are hypersensitive to DNA cross-linking
and recombination, and in its absence some important mutagenic agents, such as mitomycin C and cisplatin, in addition to being modestly
lesions are simply not repaired, leading to genomic instability and the sensitive to ionizing radiation. Therefore the Fanconi anemia proteins
consequent development of tumors. appear to function at an interface between several DNA repair and
DNA damage response pathways. Monoallelic germline mutation of
BRCA1, BRCA2, and Breast-Ovarian many FANC genes (such as PALB2/FANCN, RAD51C/FANCO, BRIP1/
Cancer Susceptibility FANCJ) has been shown to result in a modest increase in breast,
ovarian, and pancreatic cancers.
Hereditary breast cancer includes a broad group of hereditary predisposi-
tion conditions in which breast cancer is a component tumor and CONCLUSIONS AND FUTURE DIRECTIONS
accounts for approximately 5% to 10% of all breast cancer cases.
Hereditary syndromes of breast and ovarian cancer susceptibility have Molecular biology and genetic research have provided ample evidence
been particularly associated with germline mutations of two genes, to support the long-standing prediction that genomic instability is
DNA Damage Response Pathways and Cancer  •  CHAPTER 11 163

Box 11.2.  TARGETING DNA REPAIR FOR CANCER TREATMENT


As discussed in this chapter, many genes implicated in the cancer are currently under investigation; the first is the use of PARP
development of cancer play roles in DNA repair and in maintenance inhibitors as sensitizers to DNA-damaging chemotherapy or radiation,
of genomic stability.4 The mechanism of action for most cancer and the second approach exploits specific genetic characteristics of
chemotherapeutic drugs, as well as radiation therapy, is thought to be certain cancers that leave them vulnerable to DNA damage via the
through DNA damage. Therefore, developing inhibitors of DNA damage mechanism of chemical “synthetic lethality.”165 Because many effective
response pathways is an attractive approach for making tumor cells cytotoxic chemotherapies and ionizing radiotherapy exert their
more sensitive to chemotherapy or radiation therapy. Small-molecule antitumor effect through production of DNA damage, it was
inhibitors of the ATM, ATR, and DNA-PK kinases are currently being hypothesized that interference with cellular DNA repair through use of
developed and studied for their ability to enhance therapeutic PARP inhibitors may mitigate repair of this injury, resulting in decreased
responses.157 Furthermore, it seems logical that cancers with germline treatment resistance. Indeed, preclinical and clinical studies have
or acquired somatic defects in DNA repair occurring during the demonstrated potentiation or synergy of PARP inhibitors with
tumorigenic process would also be particularly susceptible to the DNA-damaging radiotherapy or chemotherapeutic drugs such as
cytotoxic effects of DNA damaging therapeutic agents. However, for methylating agents, platinums, and topoisomerase inhibitors.58,151,166
most common cancers, the clinical experience suggests otherwise. Even more compelling was the application of the idea of synthetic
It is likely that the frequently concurrent inactivation of cell cycle lethality from basic experimental genetics to cancer therapeutics.167–169
checkpoints and apoptotic processes during tumorigenesis obscures A synthetic lethal interaction describes the scenario in which a
the effect of DNA repair defects; therefore the response of clinical mutation in either of two genes alone has no phenotypic effect, but
tumors to various treatments remains very difficult to predict even the combination of both mutations results in death of the cell. Tumors
with genetic information. Nevertheless, several examples have emerged arising in patients with a germline BRCA1/2 mutation are associated
in which a detailed understanding of the DNA repair defects present with reduced DNA repair capacity owing to the loss of BRCA function
in a particular tumor type allows for the rational selection of certain and are hypothesized to rely more heavily on alternate compensatory
treatment approaches likely to be more effective.158 DNA repair processes for survival. PARP is required for the repair of
A primary example relates to the function of the BRCA1 gene, which oxidative DNA damage–associated single-strand DNA breaks in its
is involved in several types of DNA repair, including DSB repair, NER, central role in base excision repair, and if PARP is inhibited, repair-
BER, and DNA cross-link repair.49,56,153,159 Germline mutations in the associated breaks result in replication fork-mediated DSB formation,
BRCA1 gene predispose individuals to a very high risk for developing which require BRCA1/2-associated recombination to resolve. Thus, in
breast and ovarian cancers, as well as to a lesser degree pancreatic and tumors lacking intact BRCA function owing to loss of its second allele,
prostate cancers, and somatic inactivation of BRCA1 activity has also inhibition of PARP with a small molecule is postulated to be the second
been observed in sporadic breast and ovarian cancers because of hit that renders the cell incapable of survival and mimics the situation
promoter methylation. Clinical and experimental data suggest that of genetic absence of PARP. It has also become evident that PARP
breast and ovarian tumors deficient in BRCA1 function are particularly trapping on DNA single-strand breaks causes replication fork stalling
sensitive to the chemotherapeutic drug cisplatin, which causes DNA and collapse and leads to DSBs.170 However, in the host’s somatic
damage repaired through the cross-link repair pathways, and ionizing tissues heterozygous for a BRCA mutation and maintaining normal
radiation, which causes double-strand breaks (DSBs).158,160 Indeed, BRCA protein function, PARP inhibitors are thought to have little or no
tumors beyond those associated with BRCA1/2 mutations exhibiting effect. This concept of chemical synthetic lethality with use of PARP
defective homologous recombination (HRD), as determined by various inhibitors in DNA repair–deficient cancers has generated tremendous
DNA-based measures of genomic instability such as loss of enthusiasm and fueled the rapid development of clinical investigation
heterozygosity (LOH), telomeric allelic imbalance, and large-scale state in this area, particularly in BRCA mutation–associated breast and
transitions, also are relatively more sensitive to platinum agents and ovarian cancer, and phenotypically related triple-negative breast
potentially other DNA repair–targeted agents as discussed later.161 cancer.165,171 Early clinical trials of several PARP inhibitors showed
Strategies to target the three major proteins that trigger the DNA durable antitumor responses in patients with advanced germline
damage response signaling pathways—ATM, ATR, and DNA-PK—have BRCA1/2-mutated ovarian, breast, and castration-resistant prostate
identified small-molecule inhibitors with provocative preclinical activity cancer, resulting in clinical registration of olaparib (Lynparza;
in a variety of tumor types, particularly when in combination with AstraZeneca) and rucaparib (Clovis).157,172 Additional potent and
classic DNA-damaging chemotherapy or ionizing radiation. A number selective PARP inhibitors are in late-phase monotherapy and
of these are currently in phase I trials.157 combination clinical trial development, including niraparib (Tesaro),
Another very exciting finding relates to the selective activity of talazoparib (Pfizer), and veliparib (AbbVie). Of particular interest will be
inhibitors of poly (ADP-ribose) polymerase (PARP) in tumors deficient the activity of these agents in tumors with mutations in DSB repair
for BRCA1/2.162,163 PARP1 is the first described member of a large family pathway genes other than BRCA1/2 (e.g., ATM, PALB2, BRIP1) or even
of enzymes that can detect and repair DNA base damage. After DNA HRD without identified causative gene mutations, being discovered
damage, PARP1 is recruited and binds to the damaged DNA with a through increasing tumor genomic profiling efforts. Finally, as with all
subsequent increase in catalytic activity that results in the formation new therapeutic advances in cancer, tumor resistance soon arises. A
of polymer chains through use of the substrate NAD+, resulting in fascinating mechanism for resistance to PARP inhibitors for BRCA1/2-
recruitment of the BER machinery to the site of the DNA damage and mutant tumors is genetic reversion events that restore the normal
relaxation of the chromatin structure to facilitate repair.164 Two primary coding sequence, often through small deletions of the mutated
therapeutic strategies employing PARP inhibitors in the treatment of sequence, and result in improved DNA repair.173,174
164 Part I: Science and Clinical Oncology

a major factor driving the onset and progression of carcinogenesis.4 opportunities for development of more selective and more effective
Overlapping and interacting mechanisms for DNA repair and the anticancer therapies. Continued exploration of the DNA damage
cellular response to DNA damage are critical components for the response will prove important for our improved understanding of cancer
maintenance of genomic stability. Alterations in these pathways often etiology, prevention, genetic susceptibility, diagnosis, and treatment.
are early events in the multistep acquisition of genetic mutations
leading to cancer development. In addition, mutations in DNA damage The complete reference list is available online at
repair and signaling pathways are common in human tumors and offer ExpertConsult.com.

KEY REFERENCES
4. Hanahan D, Weinberg RA. Hallmarks of cancer: 37. Lowe SW, Ruley HE, Jacks T, Housman DE. 99. Lynch HT, de la Chapelle A. Hereditary colorectal
the next generation. Cell. 2011;144:646–674. p53-dependent apoptosis modulates the cytotoxicity cancer. N Engl J Med. 2003;348:919–932.
6. Loeb LA, Bielas JH, Beckman RA. Cancers exhibit of anticancer agents. Cell. 1993;74:957–967. 105. Gryfe R, Kim H, Hsieh ET, et al. Tumor micro-
a mutator phenotype: clinical implications. Cancer 40. Canman CE, Lim DS, Cimprich KA, et al. Activation satellite instability and clinical outcome in young
Res. 2008;68:3551–3557, discussion 7. of the ATM kinase by ionizing radiation and phos- patients with colorectal cancer. N Engl J Med. 2000;
7. Bodmer W, Bielas JH, Beckman RA. Genetic phorylation of p53. Science. 1998;281:1677–1679. 342:69–77.
instability is not a requirement for tumor develop- 41. Kastan MB, Lim DS. The many substrates and 109. Hampel H, Frankel WL, Martin E, et al. Feasibility
ment. Cancer Res. 2008;68:3558–3560, discussion functions of ATM. Nat Rev Mol Cell Biol. 2000;1: of screening for Lynch syndrome among patients with
60–1. 179–186. colorectal cancer. J Clin Oncol. 2008;26:5783–5788.
8. Ciccia A, Elledge SJ. The DNA damage response: 47. Xu B, O’Donnell AH, Kim ST, Kastan MB. Phos- 110. Ladabaum U, Wang G, Terdiman J, et al. Strategies
making it safe to play with knives. Mol Cell. 2010;40: phorylation of serine 1387 in Brca1 is specifically to identify the Lynch syndrome among patients with
179–204. required for the Atm-mediated S-phase checkpoint colorectal cancer: a cost-effectiveness analysis. Ann
9. Hoeijmakers JH. DNA damage, aging, and cancer. after ionizing irradiation. Cancer Res. 2002;62: Intern Med. 2011;155:69–79.
N Engl J Med. 2009;361:1475–1485. 4588–4591. 113. Lee V, Murphy A, Le DT, Diaz LA Jr. Mismatch
10. Ford JM, Hanawalt PC. Role of DNA excision 48. Bakkenist CJ, Kastan MB. DNA damage activates repair deficiency and response to immune checkpoint
repair gene defects in the etiology of cancer. Curr ATM through intermolecular autophosphorylation blockade. Oncologist. 2016;21:1200–1211.
Top Microbiol Immunol. 1997;221:47–70. and dimer dissociation. Nature. 2003;421:499–506. 114. Vogelstein B, Papadopoulos N, Velculescu VE,
13. Mellon I, Spivak G, Hanawalt PC. Selective removal 51. Spivak G. Nucleotide excision repair in humans. Zhou S, Diaz LA Jr, Kinzler KW. Cancer genome
of transcription-blocking DNA damage from the DNA Repair (Amst). 2015;36:13–18. landscapes. Science. 2013;339:1546–1558.
transcribed strand of the mammalian DHFR gene. 53. de Laat WL, Jaspers NG, Hoeijmakers JH. Molecular 116. Le DT, Uram JN, Wang H, et al. PD-1 blockade
Cell. 1987;51:241–249. mechanism of nucleotide excision repair. Genes Dev. in tumors with mismatch-repair deficiency. N Engl
15. Ljungman M, Zhang F. Blockage of RNA polymerase 1999;13:768–785. J Med. 2015;372:2509–2520.
as a possible trigger for UV light-induced apoptosis. 55. Spivak G, Ganesan AK. The complex choreography 132. Hollstein M, Sidranksky D, Vogelstein B, Harris
Oncogene. 1996;13:823–831. of transcription-coupled repair. DNA Repair (Amst). CC. p53 mutations in human cancers. Science. 1991;
16. Nelson WG, Kastan MB. DNA strand breaks: the DNA 2014;19:64–70. 253:49–53.
template alterations that trigger p53-dependent DNA 56. Hartman AR, Ford JM. BRCA1 induces DNA 135. Sengupta S, Harris CC. p53: traffic cop at the
damage response pathways. Mol Cell Biol. 1994;14: damage recognition factors and enhances nucleotide crossroads of DNA repair and recombination. Nat
1815–1823. excision repair. Nat Genet. 2002;32:180–184. Rev Mol Cell Biol. 2005;6:44–55.
18. Sale JE, Lehmann AR, Woodgate R. Y-family DNA 61. Masutani C, Kusumoto R, Yamada A, et al. The 144. Kurian AW, Hare EE, Mills MA, et al. Clinical
polymerases and their role in tolerance of cellular XPV (xeroderma pigmentosum variant) gene encodes evaluation of a multiple-gene sequencing panel
DNA damage. Nat Rev Mol Cell Biol. 2012;13: human DNA polymerase eta. Nature. 1999;399: for hereditary cancer risk assessment. J Clin Oncol.
141–152. 700–704. 2014;32:2001–2009.
21. Hartwell LH, Kastan MB. Cell cycle control and 63. Cleaver JE. Defective repair replication of DNA 146. Xu B, Kim S, Kastan MB. Involvement of Brca1 in
cancer. Science. 1994;266:1821–1828. in xeroderma pigmentosum. Nature. 1968;218: S-phase and G(2)-phase checkpoints after ionizing
22. Ford JM, Hanawalt PC. Li-Fraumeni syndrome 652–656. irradiation. Mol Cell Biol. 2001;21:3445–3450.
fibroblasts homozygous for p53 mutations are 80. Al-Tassan N, Chmiel NH, Maynard J, et al. 153. Alli E, Sharma VB, Sunderesakumar P, Ford JM.
deficient in global DNA repair but exhibit normal Inherited variants of MYH associated with somatic Defective repair of oxidative DNA damage in
transcription-coupled repair and enhanced UV resis- G:C→T:A mutations in colorectal tumors. Nat triple-negative breast cancer confers sensitivity to
tance. Proc Natl Acad Sci USA. 1995;92:8876–8880. Genet. 2002;30:227–232. inhibition of poly(ADP-ribose) polymerase. Cancer
25. Adimoolam S, Ford JM. p53 and DNA damage- 81. Jones S, Emmerson P, Maynard J, et al. Biallelic Res. 2009;69:3589–3596.
inducible expression of the xeroderma pigmentosum germline mutations in MYH predispose to multiple 157. Brown JS, O’Carrigan B, Jackson SP, Yap TA.
group C gene. Proc Natl Acad Sci USA. 2002;19: colorectal adenoma and somatic G:C→T:A muta- Targeting DNA repair in cancer: beyond PARP
12985–12990. tions. Hum Mol Genet. 2002;11:2961–2967. inhibitors. Cancer Discov. 2017;7:20–37.
27. Scully R, Chen J, Ochs RL, et al. Dynamic 83. Cheadle JP, Sampson JR. MUTYH-associated 158. Lord CJ, Ashworth A. The DNA damage response
changes of BRCA1 subnuclear location and phos- polyposis—from defect in base excision repair to and cancer therapy. Nature. 2012;481:287–294.
phorylation state are initiated by DNA damage. clinical genetic testing. DNA Repair (Amst). 2007;6: 163. Farmer H, McCabe N, Lord CJ, et al. Targeting
Cell. 1997;90:425–435. 274–279. the DNA repair defect in BRCA mutant cells as a
30. Lim DS, Kim ST, Xu B, et al. ATM phosphorylates 85. Bohr VA. Repair of oxidative DNA damage in nuclear therapeutic strategy. Nature. 2005;434:917–921.
p95/nbs1 in an S-phase checkpoint pathway. Nature. and mitochondrial DNA, and some changes with aging 177. D’Andrea AD. Susceptibility pathways in Fanconi’s
2000;404:613–617. in mammalian cells. Free Radic Biol Med. 2002;32: anemia and breast cancer. N Engl J Med. 2010;
32. Fitch ME, Nakajima S, Yasui A, Ford JM. In vivo 804–812. 362:1909–1919.
recruitment of XPC to UV-induced cyclobutane 96. Papadopoulos N, Nicolaides NC, Wei YF, et al.
pyrimidine dimers by the DDB2 gene product. Mutation of a mutL homolog in hereditary colon
J Biol Chem. 2003;278:46906–46910. cancer. Science. 1994;263:1625–1629.
DNA Damage Response Pathways and Cancer  •  CHAPTER 11 164.e1
164.e1

REFERENCES
1. Gray J, Druker B. Genomics: the breast cancer p53-dependent and is involved in global genomic 46. Tibbetts RS, Brumbaugh KM, Williams JM, et al.
landscape. Nature. 2012;486:328–329. repair. Proc Natl Acad Sci USA. 1999;96:424–428. A role for ATR in the DNA damage-induced
2. Comprehensive molecular characterization of human 25. Adimoolam S, Ford JM. p53 and DNA damage- phosphorylation of. Genes Dev. 1999;13(152–7):
colon and rectal cancer. Nature. 2012;487:330–337. inducible expression of the xeroderma pigmen- p53.
3. Nik-Zainal S, Davies H, Staaf J, et al. Landscape tosum group C gene. Proc Natl Acad Sci USA. 47. Xu B, O’Donnell AH, Kim ST, Kastan MB.
of somatic mutations in 560 breast cancer whole- 2002;19:12985–12990. Phosphorylation of serine 1387 in Brca1 is spe-
genome sequences. Nature. 2016;534:47–54. 26. Adimoolam S, Ford JM. p53 and regulation of DNA cifically required for the Atm-mediated S-phase
4. Hanahan D, Weinberg RA. Hallmarks of cancer: damage recognition during nucleotide excision repair. checkpoint after ionizing irradiation. Cancer Res.
the next generation. Cell. 2011;144:646–674. DNA Repair (Amst). 2003;2:947–954. 2002;62:4588–4591.
5. Loeb LA, Loeb KR, Anderson JP. Multiple mutations 27. Scully R, Chen J, Ochs RL, et al. Dynamic 48. Bakkenist CJ, Kastan MB. DNA damage activates
and cancer. Proc Natl Acad Sci USA. 2003;100: changes of BRCA1 subnuclear location and phos- ATM through intermolecular autophosphorylation
776–781. phorylation state are initiated by DNA damage. and dimer dissociation. Nature. 2003;421:499–506.
6. Loeb LA, Bielas JH, Beckman RA. Cancers exhibit Cell. 1997;90:425–435. 49. Ford JM. Regulation of DNA damage recognition
a mutator phenotype: clinical implications. Cancer 28. Scully R, Chen J, Plug A, et al. Association of and nucleotide excision repair: another role for p53.
Res. 2008;68:3551–3557, discussion 7. BRCA1 with Rad51 in mitotic and meiotic cells. Mutat Res. 2005;577:195–202.
7. Bodmer W, Bielas JH, Beckman RA. Genetic Cell. 1997;88:265–275. 50. Friedberg EC, Walker G, Siede W, Wood RD, Schultz
instability is not a requirement for tumor develop- 29. Cortez D, Wang Y, Qin J, Elledge SJ. Requirement R, Ellenburger T. DNA Repair and Mutagenesis. New
ment. Cancer Res. 2008;68:3558–3560, discussion of ATM-dependent phosphorylation of brca1 in York: ASM Press; 2006.
60–1. the DNA damage response to double-strand breaks. 51. Spivak G. Nucleotide excision repair in humans.
8. Ciccia A, Elledge SJ. The DNA damage response: Science. 1999;286:1162–1166. DNA Repair (Amst). 2015;36:13–18.
making it safe to play with knives. Mol Cell. 2010;40: 30. Lim DS, Kim ST, Xu B, et al. ATM phosphorylates 52. Wood RD. Nucleotide excision repair in mammalian
179–204. p95/nbs1 in an S-phase checkpoint pathway. Nature. cells. J Biol Chem. 1997;272:23465–23468.
9. Hoeijmakers JH. DNA damage, aging, and cancer. 2000;404:613–617. 53. de Laat WL, Jaspers NG, Hoeijmakers JH. Molecular
N Engl J Med. 2009;361:1475–1485. 31. Fitch ME, Cross IV, Ford JM. p53 responsive mechanism of nucleotide excision repair. Genes Dev.
10. Ford JM, Hanawalt PC. Role of DNA excision nucleotide excision repair gene products p48 and 1999;13:768–785.
repair gene defects in the etiology of cancer. Curr XPC, but not p53, localize to sites of UV-irradiation- 54. Wood RD, Mitchell M, Sgouros J, Lindahl T. Human
Top Microbiol Immunol. 1997;221:47–70. induced DNA damage, in vivo. Carcinogenesis. DNA repair genes. Science. 2001;291:1284–1289.
11. Bohr VA, Smith CA, Okumoto DS, Hanawalt PC. 2003;24:843–850. 55. Spivak G, Ganesan AK. The complex choreography
DNA repair in an active gene: removal of pyrimidine 32. Fitch ME, Nakajima S, Yasui A, Ford JM. In vivo of transcription-coupled repair. DNA Repair (Amst).
dimers from the DHFR gene of CHO cells is much recruitment of XPC to UV-induced cyclobutane 2014;19:64–70.
more efficient than in the genome overall. Cell. pyrimidine dimers by the DDB2 gene product. 56. Hartman AR, Ford JM. BRCA1 induces DNA
1985;40:359–369. J Biol Chem. 2003;278:46906–46910. damage recognition factors and enhances nucleotide
12. Mellon I, Bohr VA, Smith CA, Hanawalt PC. 33. Smith ML, Fornace AJ Jr. Mammalian DNA damage- excision repair. Nat Genet. 2002;32:180–184.
Preferential DNA repair of an active gene in human inducible genes associated with growth arrest and 57. Lin PS, McPherson LA, Chen AY, Sage J, Ford
cells. Proc Natl Acad Sci USA. 1986;83:8878–8882. apoptosis. Mutat Res. 1996;340:109–124. JM. The role of the retinoblastoma/E2F1 tumor
13. Mellon I, Spivak G, Hanawalt PC. Selective removal 34. Fornace AJ Jr, Amundson SA, Bittner M, et al. The suppressor pathway in the lesion recognition step
of transcription-blocking DNA damage from the complexity of radiation stress responses: analysis by of nucleotide excision repair. DNA Repair (Amst).
transcribed strand of the mammalian DHFR gene. informatics and functional genomics approaches. 2009;8:795–802.
Cell. 1987;51:241–249. Gene Expr. 1999;7:387–400. 58. Alli E, Sharma VB, Hartman AR, Lin PS, McPherson
14. Yamaizumi M, Sugano T. UV-induced nuclear 35. Hastak K, Adimoolam S, Trinklein ND, Myers L, Ford JM. Enhanced sensitivity to cisplatin and
accumulation of p53 is evoked through DNA RM, Ford JM. Identification of a functional in gemcitabine in Brca1-deficient murine mammary
damage of actively transcribed genes independent vivo p53 response element in the coding sequence epithelial cells. BMC Pharmacol. 2011;11:7.
of the cell cycle. Oncogene. 1994;9:2775–2784. of the xeroderma pigmentosum group C gene. Genes 59. Friedberg EC, Wagner R, Radman M. Specialized
15. Ljungman M, Zhang F. Blockage of RNA polymerase Cancer. 2012;2:1–10. DNA polymerases, cellular survival, and the genesis
as a possible trigger for UV light-induced apoptosis. 36. Lowe SW, Schmitt EM, Smith SW, Osborne BA, of mutations. Science. 2002;296:1627–1630.
Oncogene. 1996;13:823–831. Jacks T. p53 is required for radiation-induced apopto- 60. Kunkel TA. Considering the cancer consequences
16. Nelson WG, Kastan MB. DNA strand breaks: the sis in mouse thymocytes. Nature. 1993;362:847–849. of altered DNA polymerase function. Cancer Cell.
DNA template alterations that trigger p53-dependent 37. Lowe SW, Ruley HE, Jacks T, Housman DE. 2003;3:105–110.
DNA damage response pathways. Mol Cell Biol. p53-dependent apoptosis modulates the cytotoxicity 61. Masutani C, Kusumoto R, Yamada A, et al. The
1994;14:1815–1823. of anticancer agents. Cell. 1993;74:957–967. XPV (xeroderma pigmentosum variant) gene encodes
17. Ljungman M, Lane DP. Transcription—guarding the 38. Clarke AR, Purdie CA, Harrison DJ, et al. Thy- human DNA polymerase eta. Nature. 1999;399:
genome by sensing DNA damage. Nat Rev Cancer. mocyte apoptosis induced by p53-dependent and 700–704.
2004;4:727–737. independent pathways. Nature. 1993;362:849–852. 62. Johnson RE, Kondratick CM, Prakash S, Prakash L.
18. Sale JE, Lehmann AR, Woodgate R. Y-family DNA 39. Cline SD, Hanawalt PC. Who’s on first in the cellular hRAD30 mutations in the variant form of xeroderma
polymerases and their role in tolerance of cellular response to DNA damage? Nat Rev Mol Cell Biol. pigmentosum. Science. 1999;285:263–265.
DNA damage. Nat Rev Mol Cell Biol. 2012;13: 2003;4:361–373. 63. Cleaver JE. Defective repair replication of DNA
141–152. 40. Canman CE, Lim DS, Cimprich KA, et al. Activation in xeroderma pigmentosum. Nature. 1968;218:
19. Wickramasinghe CM, Arzouk H, Frey A, Maiter of the ATM kinase by ionizing radiation and phos- 652–656.
A, Sale JE. Contributions of the specialised DNA phorylation of p53. Science. 1998;281:1677–1679. 64. Kraemer KH, Lee MM, Scotto J. DNA repair
polymerases to replication of structured DNA. DNA 41. Kastan MB, Lim DS. The many substrates and protects against cutaneous and internal neoplasia:
Repair (Amst). 2015;29:83–90. functions of ATM. Nat Rev Mol Cell Biol. 2000;1: evidence from xeroderma pigmentosum. Carcino-
20. Wood RD, Doublie S. DNA polymerase theta 179–186. genesis. 1984;5:511–514.
(POLQ), double-strand break repair, and cancer. 42. Shieh SY, Ahn J, Tamai K, Taya Y, Prives C. 65. Cleaver JE, Kraemer KH. Xeroderma pigmentosum.
DNA Repair (Amst). 2016;44:22–32. The human homologs of checkpoint kinases In: Scriver CR, Beudet AL, Sly WS, Valle D, eds.
21. Hartwell LH, Kastan MB. Cell cycle control and Chk1 and Cds1 (Chk2) phosphorylate p53 at Metabolic Basis of Inherited Disease. New York:
cancer. Science. 1994;266:1821–1828. multiple DNA damage-inducible sites. Genes Dev. McGraw-Hill; 1989:2949–2971.
22. Ford JM, Hanawalt PC. Li-Fraumeni syndrome 2000;14:289–300. 66. Kraemer KH, Slor H. Xeroderma pigmentosum.
fibroblasts homozygous for p53 mutations are 43. Matsuoka S, Huang M, Elledge SJ. Linkage of ATM Clin Dermatol. 1985;3:33–69.
deficient in global DNA repair but exhibit normal to cell cycle regulation by the Chk2 protein kinase. 67. Robbins JH. Xeroderma pigmentosum: defective
transcription-coupled repair and enhanced UV resis- Science. 1998;282:1893–1897. DNA repair causes skin cancer and neurodegenera-
tance. Proc Natl Acad Sci USA. 1995;92:8876–8880. 44. Liu Q, Guntuku S, Cui XS, et al. Chk1 is an essential tion. JAMA. 1988;260:384–388.
23. Ford JM, Hanawalt PC. Expression of wild-type p53 kinase that is regulated by Atr and required for 68. Wang YC, Maher VM, Mitchell DL, McCormick
is required for efficient global genomic nucleotide the G(2)/M DNA damage checkpoint. Genes Dev. JJ. Evidence from mutation spectra that the UV
excision repair in UV-irradiated human fibroblasts. 2000;14:1448–1459. hypermutability of xeroderma pigmentosum variant
J Biol Chem. 1997;272:28073–28080. 45. Shiloh Y. ATM and related protein kinases: safe- cells reflects abnormal, error-prone replication on a
24. Hwang BJ, Ford JM, Hanawalt PC, Chu G. Expres- guarding genome integrity. Nat Rev Cancer. 2003; template containing photoproducts. Mol Cell Biol.
sion of the p48 xeroderma pigmentosum gene is 3:155–168. 1993;13:4276–4283.
164.e2 Part I: Science and Clinical Oncology

69. Schmickel RD, Chu EHY, Trosko JE. Cockayne 90. Kolodner R. Biochemistry and genetics of eukaryotic 113. Lee V, Murphy A, Le DT, Diaz LA Jr. Mismatch
syndrome: a cellular sensitivity to ultraviolet light. mismatch repair. Genes Dev. 1996;10:1433–1442. repair deficiency and response to immune checkpoint
Pediatrics. 1977;60:135–139. 91. Kolodner RD, Marsischky GT. Eukaryotic DNA blockade. Oncologist. 2016;21:1200–1211.
70. Venema J, Mullenders LHF, Natarajan AT, Van mismatch repair. Curr Opin Genet Dev. 1999;9: 114. Vogelstein B, Papadopoulos N, Velculescu VE,
Zeeland AA, Mayne LV. The genetic defect in Cock- 89–96. Zhou S, Diaz LA Jr, Kinzler KW. Cancer genome
ayne syndrome is associated with a defect in repair of 92. Jiricny J, Nystrom-Lahti M. Mismatch repair landscapes. Science. 2013;339:1546–1558.
UV-induced DNA damage in transcriptionally active defects in cancer. Curr Opin Genet Dev. 2000;10: 115. Kim H, Jen J, Vogelstein B, Hamilton SR. Clinical
DNA. Proc Natl Acad Sci USA. 1990;87:4707–4711. 157–161. and pathological characteristics of sporadic colorectal
71. Cooper PK, Nouspikel T, Clarkson SG, Leadon SA. 93. Peltomaki P. Role of DNA mismatch repair defects carcinomas with DNA replication errors in micro-
Defective transcription-coupled repair of oxidative in the pathogenesis of human cancer. J Clin Oncol. satellite sequences. Am J Pathol. 1994;145:148–156.
base damage in Cockayne syndrome patients from 2003;21:1174–1179. 116. Le DT, Uram JN, Wang H, et al. PD-1 Blockade
XP group G. Science. 1997;275:990–993. 94. Fishel R, Lescoe MK, Rao MRS, et al. The human in tumors with mismatch-repair deficiency. N Engl
72. Timme TL, Moses RE. Review: diseases with mutator gene homolog MSH2 and its association J Med. 2015;372:2509–2520.
DNA damage-processing defects. Am J Med Sci. with hereditary nonpolyposis colon cancer. Cell. 117. Bell DW, Varley JM, Szydlo TE, et al. Heterozygous
1988;295:40–48. 1993;75:1027–1038. germ line hCHK2 mutations in Li-Fraumeni
73. Nance MA, Berry SA. Cockayne Syndrome: review 95. Hemminki A, Peltomaki P, Mecklin JP, et al. syndrome. Science. 1999;286:2528–2531.
of 140 cases. Am J Med Gen. 1992;42:68–84. Loss of the wild type MLH1 gene is a feature of 118. Meijers-Heijboer H, van den Ouweland A, Klijn J,
74. Robbins JH, Kraemer KH, Lutzner MA, Festoff BW, hereditary nonpolyposis colorectal cancer. Nat Genet. et al. Low-penetrance susceptibility to breast cancer
Coon HG. Xeroderma pigmentosum: an inherited 1994;8:405–410. due to CHEK2(*)1100delC in noncarriers of BRCA1
disease with sun sensitivity, multiple cutaneous 96. Papadopoulos N, Nicolaides NC, Wei YF, et al. or BRCA2 mutations. Nat Genet. 2002;31:55–59.
neoplasms and abnormal repair. Ann Intern Med. Mutation of a mutL homolog in hereditary colon 119. Vahteristo P, Bartkova J, Eerola H, et al. A CHEK2
1974;80:221–228. cancer. Science. 1994;263:1625–1629. genetic variant contributing to a substantial frac-
75. Vermeulen W, Scott RJ, Rodgers S, et al. Clinical 97. Karran P, Bignami M. DNA damage tolerance, tion of familial breast cancer. Am J Hum Genet.
heterogeneity within xeroderma pigmentosum mismatch repair and genome instability. Bioessays. 2002;71:432–438.
associated with mutations in the DNA repair and 1994;16:833–839. 120. Kastan MB, Onyekwere O, Sidransky D, Vogelstein
trasncription gene ERCC3. Am J Hum Genet. 98. Lynch HT, Smyrk T. Hereditary nonpolyposis B, Craig RW. Participation of p53 protein in the
1994;54:191–200. colorectal cancer (Lynch syndrome). An updated cellular response to DNA damage. Cancer Res.
76. Cleaver JE, Lam ET, Revet I. Disorders of nucleotide review. Cancer. 1996;78:1149–1167. 1991;51:6304–6311.
excision repair: the genetic and molecular basis of 99. Lynch HT, de la Chapelle A. Hereditary colorectal 121. Kuerbitz SJ, Plunkett BS, Walsh WV, Kastan MB.
heterogeneity. Nat Rev Genet. 2009;10:756–768. cancer. N Engl J Med. 2003;348:919–932. Wild-type p53 is a cell cycle checkpoint determinant
77. Lehmann AR. The xeroderma pigmentosum group D 100. Parsons R, Li GM, Longley MJ, et al. Hypermutabil- following irradiation. Proc Natl Acad Sci USA.
(XPD) gene: one gene, two functions, three diseases. ity and mismatch repair deficiency in RER+ tumor 1992;89:7491–7495.
Genes Dev. 2001;15:15–23. cells. Cell. 1993;75:1227–1236. 122. Khanna KK, Keating KE, Kozlov S, et al. ATM
78. Demple B, Harrison L. Repair of oxidative damage to 101. Thibodeau SN, Bren G, Schaid D. Microsatellite associates with and phosphorylates p53: mapping the
DNA: enzymology and biology. Annu Rev Biochem. instability in cancer of the proximal colon. Science. region of interaction. Nat Genet. 1998;20:398–400.
1994;63:915–948. 1993;260:816–819. 123. Banin S, Moyal L, Shieh S, et al. Enhanced phos-
79. Seeberg E, Eide L, Bjoras M. The base excision repair 102. Kuismanen SA, Holmberg MT, Salovaara R, et al. phorylation of p53 by ATM in response to DNA
pathway. Trends Biochem Sci. 1995;20:391–397. Epigenetic phenotypes distinguish microsatellite- damage. Science. 1998;281:1674–1677.
80. Al-Tassan N, Chmiel NH, Maynard J, et al. stable and -unstable colorectal cancers. Proc Natl 124. O’Driscoll M, Jeggo PA. The role of double-strand
Inherited variants of MYH associated with somatic Acad Sci USA. 1999;96:12661–12666. break repair—insights from human genetics. Nat
G:C→T:A mutations in colorectal tumors. Nat 103. Nakagawa H, Nuovo GJ, Zervos EE, et al. Rev Genet. 2006;7:45–54.
Genet. 2002;30:227–232. Age-related hypermethylation of the 5′ region of 125. Gatti RA, Tward A, Concannon P. Cancer risk
81. Jones S, Emmerson P, Maynard J, et al. Biallelic MLH1 in normal colonic mucosa is associated with in ATM heterozygotes: a model of phenotypic
germline mutations in MYH predispose to multiple microsatellite-unstable colorectal cancer develop- and mechanistic differences between missense
colorectal adenoma and somatic G:C→T:A muta- ment. Cancer Res. 2001;61:6991–6995. and truncating mutations. Mol Genet Metab.
tions. Hum Mol Genet. 2002;11:2961–2967. 104. Ford JM, Whittemore AS. Predicting and 1999;68:419–423.
82. Halford SE, Rowan AJ, Lipton L, et al. Germline preventing hereditary colorectal cancer. JAMA. 126. Dork T, Bendix R, Bremer M, et al. Spectrum of
mutations but not somatic changes at the MYH locus 2006;296:1521–1523. ATM gene mutations in a hospital-based series
contribute to the pathogenesis of unselected colorec- 105. Gryfe R, Kim H, Hsieh ET, et al. Tumor micro- of unselected breast cancer patients. Cancer Res.
tal cancers. Am J Pathol. 2003;162:1545–1548. satellite instability and clinical outcome in young 2001;61:7608–7615.
83. Cheadle JP, Sampson JR. MUTYH-associated patients with colorectal cancer. N Engl J Med. 127. Scott SP, Bendix R, Chen P, Clark R, Dork T, Lavin
polyposis—from defect in base excision repair 2000;342:69–77. MF. Missense mutations but not allelic variants alter
to clinical genetic testing. DNA Repair (Amst). 106. Hemminki A, Mecklin JP, Jarvinen H, Aaltonen LA, the function of ATM by dominant interference in
2007;6:274–279. Joensuu H. Microsatellite instability is a favorable patients with breast cancer. Proc Natl Acad Sci USA.
84. Le Page F, Kwoh EE, Avrutskaya A, et al. Transcrip- prognostic indicator in patients with colorectal 2002;99:925–930.
tion-coupled repair of 8-oxoguanine: requirement cancer receiving chemotherapy. Gastroenterology. 128. Carney JP, Maser RS, Olivares H, et al. The
for XPG, TFIIH, and CSB and implications for 2000;119:921–928. hMre11/hRad50 protein complex and Nijmegen
Cockayne syndrome. Cell. 2000;101:159–171. 107. Samowitz WS, Curtin K, Ma KN, et al. Microsatellite breakage syndrome: linkage of double-strand break
85. Bohr VA. Repair of oxidative DNA damage in instability in sporadic colon cancer is associated with repair to the cellular DNA damage response. Cell.
nuclear and mitochondrial DNA, and some changes an improved prognosis at the population level. Cancer 1998;93:477–486.
with aging in mammalian cells. Free Radic Biol Med. Epidemiol Biomarkers Prev. 2001;10:917–923. 129. Stewart GS, Maser RS, Stankovic T, et al. The DNA
2002;32:804–812. 108. Ji H, Kumm J, Zhang M, et al. Molecular inver- double-strand break repair gene hMRE11 is mutated
86. Morland I, Rolseth V, Luna L, Rognes T, Bjoras sion probe analysis of gene copy alterations reveals in individuals with an ataxia-telangiectasia-like
M, Seeberg E. Human DNA glycosylases of the distinct categories of colorectal carcinoma. Cancer disorder. Cell. 1999;99:577–587.
bacterial Fpg/MutM superfamily: an alternative Res. 2006;66:7910–7919. 130. Schneider JG, Finck BN, Ren J, et al. ATM-
pathway for the repair of 8-oxoguanine and other 109. Hampel H, Frankel WL, Martin E, et al. Feasibility dependent suppression of stress signaling reduces
oxidation products in DNA. Nucleic Acids Res. of screening for Lynch syndrome among patients with vascular disease in metabolic syndrome. Cell Metab.
2002;30:4926–4936. colorectal cancer. J Clin Oncol. 2008;26:5783–5788. 2006;4:377–389.
87. Goodenberger M, Lindor NM. Lynch syndrome 110. Ladabaum U, Wang G, Terdiman J, et al. Strategies 131. Valentin-Vega YA, Maclean KH, Tait-Mulder J, et al.
and MYH-associated polyposis: review and testing to identify the Lynch syndrome among patients with Mitochondrial dysfunction in ataxia-telangiectasia.
strategy. J Clin Gastroenterol. 2011;45:488–500. colorectal cancer: a cost-effectiveness analysis. Ann Blood. 2012;119:1490–1500.
88. Kiyohara C, Takayama K, Nakanishi Y. Association Intern Med. 2011;155:69–79. 132. Hollstein M, Sidranksky D, Vogelstein B, Harris
of genetic polymorphisms in the base excision repair 111. Hodi FS, O’Day SJ, McDermott DF, et al. Improved CC. p53 mutations in human cancers. Science.
pathway with lung cancer risk: a meta-analysis. Lung survival with ipilimumab in patients with metastatic 1991;253:49–53.
Cancer. 2006;54:267–283. melanoma. N Engl J Med. 2010;363:711–723. 133. Kastan MB, Radin AI, Kuerbitz SJ, et al. Levels of
89. Hung RJ, Hall J, Brennan P, Boffetta P. Genetic 112. Chabanon RM, Pedrero M, Lefebvre C, Marabelle p53 protein increase with maturation in human
polymorphisms in the base excision repair pathway A, Soria JC, Postel-Vinay S. Mutational landscape hematopoietic cells. Cancer Res. 1991;51:4279–4286.
and cancer risk: a HuGE review. Am J Epidemiol. and sensitivity to immune checkpoint blockers. Clin 134. Kastan MB, Zhan Q, El-Deiry WS, et al. A mam-
2005;162:925–942. Cancer Res. 2016;22:4309–4321. malian cell cycle checkpoint pathway utilizing p53
DNA Damage Response Pathways and Cancer  •  CHAPTER 11 164.e3
164.e3

and GADD45 is defective in ataxia-telangiectasia. 151. Hastak K, Alli E, Ford JM. Synergistic chemo- 167. Lucchesi JC. Synthetic lethality and semi-lethality
Cell. 1992;71:587–597. sensitivity of triple-negative breast cancer cell among functionally related mutants of Drosophila
135. Sengupta S, Harris CC. p53: traffic cop at the lines to poly(ADP-Ribose) polymerase inhibition, melanogaster. Genetics. 1968;59:37–44.
crossroads of DNA repair and recombination. Nat gemcitabine, and cisplatin. Cancer Res. 2010;70: 168. Hartwell LH, Szankasi P, Roberts CJ, Murray
Rev Mol Cell Biol. 2005;6:44–55. 7970–7980. AW, Friend SH. Integrating genetic approaches
136. Oren M. Regulation of the p53 tumor suppressor 152. Saha T, Rih JK, Roy R, Ballal R, Rosen EM. into the discovery of anticancer drugs. Science.
protein. J Biol Chem. 1999;274:36031–36034. Transcriptional regulation of the base excision 1997;278:1064–1068.
137. Takagi M, Absalon MJ, McLure KG, Kastan MB. repair pathway by BRCA1. J Biol Chem. 2010;285: 169. Kaelin WG Jr. The concept of synthetic lethality in
Regulation of p53 translation and induction after 19092–19105. the context of anticancer therapy. Nat Rev Cancer.
DNA damage by ribosomal protein L26 and 153. Alli E, Sharma VB, Sunderesakumar P, Ford JM. 2005;5:689–698.
nucleolin. Cell. 2005;123:49–63. Defective repair of oxidative DNA damage in 170. Murai J, Huang SY, Das BB, et al. Trapping of
138. Chen J, Kastan MB. 5′-3′-UTR interactions regulate triple-negative breast cancer confers sensitivity to PARP1 and PARP2 by Clinical PARP Inhibi-
p53 mRNA translation and provide a target for inhibition of poly(ADP-ribose) polymerase. Cancer tors. Cancer Res. 2012;72:5588–5599.
modulating p53 induction after DNA damage. Genes Res. 2009;69:3589–3596. 171. Fong PC, Boss DS, Yap TA, et al. Inhibition of
Dev. 2010;24:2146–2156. 154. Pellegrini L, Yu DS, Lo T, et al. Insights into DNA poly(ADP-ribose) polymerase in tumors from BRCA
139. El-Deiry WS, Tokino T, Velculescu VE, et al. WAF1, recombination from the structure of a RAD51- mutation carriers. N Engl J Med. 2009;361:123–134.
a potential mediator of p53 tumor suppression. Cell. BRCA2 complex. Nature. 2002;420:287–293. 172. Kaufman B, Shapira-Frommer R, Schmutzler
1993;75:817–825. 155. Yang H, Jeffrey PD, Miller J, et al. BRCA2 func- RK, et al. Olaparib monotherapy in patients with
140. Vousden KH, Lu X. Live or let die: the cell’s response tion in DNA binding and recombination from a advanced cancer and a germline BRCA1/2 mutation.
to p53. Nat Rev Cancer. 2002;2:594–604. BRCA2-DSS1-ssDNA structure. Science. 2002;297: J Clin Oncol. 2015;33:244–250.
141. Malkin D, Li FP, Strong LC, et al. Germ line 1837–1848. 173. Bouwman P, Jonkers J. Molecular pathways: how can
p53 mutations in a familial syndrome of breast 156. Venkitaraman AR. Cancer susceptibility and the BRCA-mutated tumors become resistant to PARP
cancer, sarcomas and other neoplasms. Science. functions of BRCA1 and BRCA2. Cell. 2002;108: inhibitors? Clin Cancer Res. 2014;20:540–547.
1990;250:1233–1238. 171–182. 174. Afghahi A, Timms KM, Vinayak S, et al. Tumor
142. Srivastava S, Zou ZQ, Pirollo K, Blattner W, Chang 157. Brown JS, O’Carrigan B, Jackson SP, Yap TA. BRCA1 reversion mutation arising during neoadju-
EH. Germ-line transmission of a mutated p53 gene Targeting DNA Repair in Cancer: Beyond PARP vant platinum-based chemotherapy in triple-negative
in a cancer-prone family with Li-Fraumeni syndrome. Inhibitors. Cancer Discov. 2017;7:20–37. breast cancer is associated with therapy resistance.
Nature. 1990;348:747–749. 158. Lord CJ, Ashworth A. The DNA damage response Clin Cancer Res. 2017;23(13):3365–3370.
143. Frebourg T, Barbier N, Yan YX, et al. Germ-line and cancer therapy. Nature. 2012;481:287–294. 175. Alter BP. Cancer in Fanconi anemia, 1927-2001.
p53 mutations in 15 families with Li-Fraumeni 159. Hartman AR, Ford JM. BRCA1 and p53: compensa- Cancer. 2003;97:425–440.
syndrome. Am J Hum Genet. 1995;56:608–615. tory roles in DNA repair. J Mol Med. 2003. 176. D’Andrea AD, Grompe M. The Fanconi anaemia/
144. Kurian AW, Hare EE, Mills MA, et al. Clinical 160. Telli ML, Jensen KC, Vinayak S, et al. Phase II BRCA pathway. Nat Rev Cancer. 2003;3:23–34.
evaluation of a multiple-gene sequencing panel Study of gemcitabine, carboplatin, and iniparib 177. D’Andrea AD. Susceptibility pathways in Fanconi’s
for hereditary cancer risk assessment. J Clin Oncol. as neoadjuvant therapy for triple-negative and anemia and breast cancer. N Engl J Med. 2010;
2014;32:2001–2009. BRCA1/2 mutation-associated breast cancer with 362:1909–1919.
145. Kurian AW, Kingham KE, Ford JM. Next-generation assessment of a tumor-based measure of genomic 178. Kennedy RD, D’Andrea AD. DNA repair pathways
sequencing for hereditary breast and gynecologic instability: PrECOG 0105. J Clin Oncol. 2015;33: in clinical practice: lessons from pediatric cancer
cancer risk assessment. Curr Opin Obstet Gynecol. 1895–1901. susceptibility syndromes. J Clin Oncol. 2006;24:
2015;27:23–33. 161. Telli ML, Timms KM, Reid J, et al. Homologous 3799–3808.
146. Xu B, Kim S, Kastan MB. Involvement of Brca1 recombination deficiency (HRD) score predicts 179. Ceccaldi R, Sarangi P, D’Andrea AD. The Fanconi
in S-phase and G(2)-phase checkpoints after response to platinum-containing neoadjuvant anaemia pathway: new players and new functions.
ionizing irradiation. Mol Cell Biol. 2001;21: chemotherapy in patients with triple-negative breast Nat Rev Mol Cell Biol. 2016;17:337–349.
3445–3450. cancer. Clin Cancer Res. 2016;22:3764–3773. 180. Moldovan GL, D’Andrea AD. To the rescue: the
147. Wang Y, Cortez D, Yazdi P, Neff N, Elledge SJ, Qin 162. Bryant HE, Schultz N, Thomas HD, et al. Specific Fanconi anemia genome stability pathway salvages
J. BASC, a super complex of BRCA1-associated killing of BRCA2-deficient tumours with inhibitors replication forks. Cancer Cell. 2012;22:5–6.
proteins involved in the recognition and repair of poly(ADP-ribose) polymerase. Nature. 2005;434: 181. Kim H, D’Andrea AD. Regulation of DNA cross-link
of aberrant DNA structures. Genes Dev. 2000; 913–917. repair by the Fanconi anemia/BRCA pathway. Genes
14:927–939. 163. Farmer H, McCabe N, Lord CJ, et al. Targeting Dev. 2012;26:1393–1408.
148. Moynahan ME, Chiu JW, Koller BH, Jasin M. Brca1 the DNA repair defect in BRCA mutant cells 182. Howlett NG, Taniguchi T, Olson S, et al. Biallelic
controls homology-directed DNA repair. Mol Cell. as a therapeutic strategy. Nature. 2005;434:917–921. inactivation of BRCA2 in Fanconi anemia. Science.
1999;4:511–518. 164. Durkacz BW, Omidiji O, Gray DA, Shall S. (ADP- 2002;297:606–609.
149. Roy R, Chun J, Powell SN. BRCA1 and BRCA2: ribose)n participates in DNA excision repair. Nature. 183. Kim H, Yang K, Dejsuphong D, D’Andrea AD.
different roles in a common pathway of genome 1980;283:593–596. Regulation of Rev1 by the Fanconi anemia core
protection. Nat Rev Cancer. 2012;12:68–78. 165. Telli ML, Ford JM. PARP inhibitors in breast cancer. complex. Nat Struct Mol Biol. 2012;19:164–170.
150. Harkin DP, Bean JM, Miklos D, et al. Induction Clin Adv Hematol Oncol. 2010;8:629–635.
of GADD45 and JNK/SAPK-dependent apoptosis 166. Javle M, Curtin NJ. The role of PARP in DNA
following inducible expression of BRCA1. Cell. repair and its therapeutic exploitation. Br J Cancer.
1999;97:575–586. 2011;105:1114–1122.
164.e4 Part I: Science and Clinical Oncology

SELF-ASSESSMENT REVIEW QUESTIONS ANSWERS


1. True or false? Hereditary defects in DNA repair genes are 1. (a)
common in cancer. 2. (d)
a. True 3. (d)
b. False 4. (b)
2. DNA mismatch repair targets which lesions? 5. (e)
a. UV photoproducts 6. (e)
b. Oxidative base damage 7. (d)
c. DNA strand breaks
d. Replication errors
3. Lynch syndrome is associated with enhanced risk for which of
the following malignancies?
a. Colorectal cancer
b. Endometrial cancer
c. Cervical cancer
d. Colorectal and endometrial cancers
e. Colorectal, endometrial, and cervical cancers
4. How common are BRCA1 and BRCA2 mutations in the
general population?
a. 1 in 10,000
b. 1 in 400
c. 1 in 40
d. 1 in 4
5. Microsatellite instability is a feature of which malignancies?
a. All colon cancers in patients with Lynch syndrome
b. Twenty percent of all colon cancers
c. Endometrial cancers in patients with Lynch syndrome
d. Half of breast cancers in patients with Lynch syndrome
e. All of the above
6. Breast cancers that arise in BRCA1 mutation carriers are
specifically sensitive to which therapies?
a. Cisplatin
b. PARP inhibitors
c. Taxanes
d. Bevacizumab
e. a and b
f. All of the above
7. MSI in colon cancer is:
a. A good prognostic marker
b. A poor prognostic marker
c. A predictive marker for drug response
d. a and c
e. b and c
f. None of the above
Viruses and Human Cancer 12 
Paul F. Lambert and Bill Sugden

S UMMARY OF K EY P OI NT S
• More than 15% of human cancers cancer indirectly by causing chronic virus is no longer able to replicate in
are known today to be caused by destruction of the target organ from the cancer.
viruses. which the cancer arises. • Virally induced cancers can be
• A hallmark of virally induced cancers • The etiologic role of viruses in prevented through the use of
is that they are associated with cancer is established through a effective prophylactic vaccines.
persistent viral infections. combination of epidemiologic and • Viral genes expressed in associated
• Although some viruses encode molecular evidence. cancers represent targets for
oncogenes that directly contribute to • In many cases, the cancers caused therapeutic vaccines and
the cancers they cause, other by the virus represent dead-end viral-specific anticancer drugs.
viruses are thought to result in streets for the virus—that is, the

Viruses cause cancers in people. More than 15% of all human cancers cancers rapidly; often 15 to 50 years will elapse between the primary
are thought to have a viral etiology,1 and this fraction is likely to grow infection and tumor development. Nor do human tumor viruses express
as we investigate additional cancers for a potential viral cause and cellularly derived oncogenes; rather, some of them have evolved to
identify new human viruses. Identifying a human cancer as having inhibit cellular tumor suppressor genes. These differences between
a viral etiology has substantive consequences both for its treatment highly oncogenic animal viruses and human tumor viruses probably
and its prevention. The known virally caused human cancers often have contributed to the delay in our recognition that viruses do cause
express virally encoded products in the tumor cells. These viral cancers in people.
products are potential targets for antiviral, tumor-specific therapies. A second obstacle in our recognition that viruses can be tumorigenic
Some viral infections can now be prevented with vaccines; some now in their human host is that we lack convincing animal models in
can be cured with antiviral drugs; therefore it may be possible to which to test these viruses directly. For all practical purposes, all
eliminate the human cancers that require viral contributions for their known human tumor viruses infect only people. In addition, we now
development. know that human viruses found not to be tumorigenic in people
For example, we now can realistically look forward to the elimination are tumorigenic when experimentally introduced into test animals.
of many or most cases of primary hepatocellular carcinoma (HCC) For example, human adenovirus 12, which causes only respiratory
and to most cases of cervical carcinoma. This happy eventuality is infections in people, is highly oncogenic when inoculated into newborn
thanks to the development and eventual use of vaccines effective hamsters.7 The lack of an experimentally tractable animal host for
against, respectively, hepatitis B virus (HBV) and most tumorigenic human tumor viruses has required multiple lines of evidence to affirm
strains of human papillomaviruses (HPVs), and potent antiviral drugs that a given virus can contribute to a given human cancer. In particular,
active against hepatitis C virus (HCV). epidemiologic findings have been combined with genetic and molecular
The search for human tumor viruses has been propelled by a long analyses in cell culture to identify human tumor viruses. Experi-
appreciation that viruses can cause cancers in birds and rodents. Viruses ments with mice transgenic for viral genes also have supported these
were isolated as filterable extracts from avian tumors in the first decade identifications.
of the last century and were shown to induce tumors in susceptible This chapter introduces the seven known viruses that cause human
animals.2,3 Parallel findings were made in mice in the 1940s.4 These tumors, the tumors with which they are associated, data that support
animal tumor viruses were in the retrovirus family and led researchers these associations, and models to explain the viral contributions to
to look for retroviruses as human tumor viruses in the 1960s and these tumors. These viruses are presented in the order of their discovery,
1970s. However, most of the human tumor viruses subsequently because early findings often have provided insights for analyses of
identified are in different virus families and do not conform to some subsequently identified viruses. Finally, the chapter outlines the types
of the expectations derived from the study of highly oncogenic animal of virus-specific therapies that are possible to develop and the likelihood
retroviruses. For example, highly oncogenic tumor viruses induce of developing vaccines for human tumor viruses in order to limit or
tumors in animals rapidly. The inoculation of Rous sarcoma virus eliminate specific human cancers.
into the wing web of newborn chicks can induce fatal sarcomas within
2 weeks in 100% of susceptible animals.5 In addition, highly oncogenic EPSTEIN-BARR VIRUS
tumor viruses are oncogenic because they have acquired and express
potent derivatives of cellular proto-oncogenes. In fact, many of the Epstein-Barr virus (EBV) was identified through the insight and
known human proto-oncogenes were first identified as homologues advocacy of Denis Burkitt. As a young surgeon, having identified
of the oncogenes transduced by the highly oncogenic animal retrovi- Burkitt lymphoma as a new disease entity, he analyzed the geographic
ruses.6 Known human tumor viruses, however, usually do not induce and climatic distribution of this childhood lymphoma, determined

165
166 Part I: Science and Clinical Oncology

that it overlapped with that of malaria, and postulated that it had an The association of EBV with Burkitt lymphoma and NPC and
infectious etiology.8 At his urging and with his proffered biopsy samples, the demonstration that EBV causes the bulk of infectious mononucleosis
Tony Epstein and colleagues identified EBV in Burkitt lymphoma– have led researchers to consider other diseases with which EBV might
derived cells.9 To do so, they developed the expertise to propagate be associated. During the past 20 years, EBV also has been linked to
these cells in culture.10 Cell lines derived from EBV-positive Burkitt posttransplant lymphoproliferative disease (PTLD)17; oral hairy leu-
lymphomas proved to be powerful tools in associating EBV with koplakia18; approximately one-third to one-half of cases of Hodgkin
different human diseases. Different EBV-positive cell lines express disease19,20; one-third of AIDS-related diffuse large B-cell lymphomas
different viral antigens and thereby have served as test samples for (DLBCLs)21; and one-tenth of gastric carcinomas.22 These linkages
patients’ expression of antibodies to EBV-encoded antigens. have been made not only through serologic findings but also by
The analyses of these antibodies by serologic methods led the molecular genetic analyses that render the linkages more robust. The
Henles in Philadelphia to propose EBV as the cause of infectious latter analyses have been made possible by the elucidation of the
mononucleosis.11,12 A colleague in their laboratory who had lacked molecular virology of EBV in cell culture.23
antibodies to EBV-encoded antigens had developed those antibodies EBV is a herpesvirus; it has a double-stranded DNA of 165,000
on presentation with infectious mononucleosis. The etiologic role for to 170,000 base pairs24 and encodes more than 100 genes,25 with 25
EBV in this “self-limiting lymphoproliferation” was subsequently of these being pre-microRNAs (miRNAs), which encode up to 50
established by careful, prospective epidemiologic studies in which miRNAs26–29 (Fig. 12.1). As with other herpesviruses, EBV has two
serologic analysis was used to demonstrate that only immunologically distinct phases to its life cycle. It can infect cells, express a small subset
naïve people were at risk of the development of infectious mononucleo- of its genes (see Fig. 12.1), and cohabit with the cell without killing
sis; with its development, they would first express antibodies to it (its “latent” phase). EBV also can emerge from its latency, express
EBV-encoded antigens of the immunoglobulin (Ig) M class, and only all or most of its genes, amplify its DNA, assemble progeny virions,
later to those of the IgG class.13 Thus about 85% of infectious and kill its host cell by lysis (its “lytic” phase). Unlike neurotropic
mononucleosis cases were shown to arise from a primary infection herpesviruses such as herpes simplex virus type 1 (HSV-1) and varicella-
with EBV. Serologic studies also allowed the Henles to propose that zoster virus, EBV in its latent phase need not be maintained in a
nasopharyngeal carcinoma (NPC) might be caused by EBV because nonproliferating host cell. Rather, it has the capacity to both initiate
patients with NPC were characterized by having atypically high titers and maintain proliferation in at least the B lymphocytes it infects in
to EBV-associated antigens.14 However, the data that linked EBV cell culture and at early stages of primary infection in vivo.30,31 Although
causally to Burkitt lymphoma and NPC by the early 1970s were only it is the ability of EBV to support proliferation and survival of its
“guilt by association.” Although EBV caused most infectious mono- infected host cell that likely renders it oncogenic, the first step in its
nucleosis on primary infection, serologic findings also had demonstrated life cycle is to infect a host’s B cells successfully. As with other her-
that children in the parts of Africa in which Burkitt lymphoma is pesviruses, EBV has evolved a powerful mechanism to inhibit newly
endemic and adults in the parts of China in which NPC is prevalent infected B cells from being recognized by the host’s immune response.
all had been infected with EBV—that is, they were “EBV seropositive” EBV encodes at least 44 miRNAs that can target a suite of cellular
long before these cancers developed. mRNAs for degradation. Studies using newly infected B cells and
The serologic analyses of EBV in the 1960s and 1970s illustrate autologous T cells have shown that viral miRNAs target mRNAs
a conundrum for viruses and human cancers: “How can many people encoding IL12B, resulting in suppression of type 1 helper T cell (Th1)
be infected with a given virus, and yet how can that virus contribute differentiation. These viral miRNAs also control gene expression of
to tumor development in only a few infected subjects after long HLA class II and three lysosomal enzymes important for proteolysis
periods?” This apparent paradox applies, in fact, to most cancers and epitope presentation to CD4+ T cells.32 Parallel analyses have
associated with human tumor viruses and explains a major reluctance shown that the EBV’s miRNAs also inhibit recognition of infected
to consider viruses as etiologic agents for human cancers. The World cells by cytotoxic CD8+ T cells.33
Health Organization (WHO), without resolving this conundrum, Clearly EBV’s success as a pathogen reflects, in part, its ability
sponsored a prospective epidemiologic survey in Uganda to assay to inhibit its host from recognizing and killing the cells that the
42,000 youngsters serologically for evidence for or against the contribu- virus infects.
tion of EBV causally to Burkitt lymphoma. The region studied had EBV efficiently induces and maintains proliferation of infected B
a high incidence of this cancer. Samples of blood were collected from cells. In genetic experiments in which two viral genes, EBNA2 and
children, their serum was stored, and for children later identified as LMP1 (see Fig. 12.1), are expressed conditionally within the context
having Burkitt lymphoma, blood samples were collected again and of the virus, each gene product when assayed alone needs to function
their titers to EBV antigens were determined. In this prospective for infected B cells to continue to proliferate.34,35 These observations
survey, Burkitt lymphoma developed in 14 youngsters during the 5 are particularly telling because they help to explain the multistep
years they were monitored, and those in whom the lymphoma developed evolution of Burkitt lymphoma. Many other genetic analyses have
had, before tumor development, on average a 3.4-fold higher titer of shown that four additional viral genes—EBNA1, EBNA3a, EBNA3b,
antibodies to one class of EBV antigens than did the children in and EBNA3c (see Fig. 12.1)—affect some facet of cell proliferation
whom the lymphoma did not develop.15 That is, for children in the or survival.36 EBNA1 is essential for the viral genome to be maintained
portions of the world in which EBV-associated Burkitt lymphoma is as a plasmid in cells.37 Its inhibition leads to the loss of the viral DNA
endemic, a high titer of antibodies to a given set of EBV-encoded from tumor cells and to their death.38 EBNA3a acts at the stage of
antigens represents a 30-fold risk factor for the development of Burkitt initiation of proliferation.39 EBNA3b is a tumor suppressor.40 EBNA3a
lymphoma.15 and EBNA3c inhibit p16INK4 and p14ARF through complex, distinct
Do these findings prove that EBV causes Burkitt lymphoma? No; binding to promoter elements41,42 A seventh viral gene, LMP2a, can
proof in such cases for which direct experiments are not feasible substitute for the signaling provided by a functional B-cell receptor
ultimately comes from the accretion of supporting findings in the (BCR) to support the survival of murine and human B cells that lack
absence of confounding data. However, the WHO study did make functional BCRs.43–45 This activity of LMP2a likely underlies one
it unlikely that EBV is a passenger virus that merely replicates well contribution of EBV to the tumor cells in Hodgkin disease that fail
in tumor cells, because the antibody titers were elevated 7 to 54 to express functional BCRs.46 Two additional genes of EBV are expressed
months before tumor detection.15 Similar prospective surveys were as nontranslated, small RNAs, termed “EBERs.” They contribute to
carried out in China, which identified antibodies of the IgA class to the efficiency with which EBV induces and maintains proliferation
the same set of EBV antigens as a risk factor for the development of B lymphocytes infected in culture47; they also induce type I
of NPC.16 interferon48 and mediate other activities that potentially contribute
Viruses and Human Cancer  •  CHAPTER 12 167

BARF0

14
RT

m R- BA RT T2 3
RT2

1
m iR- -BA
m iR- -BA AR RT
-BA

m iR- BAR RT 19 0
miR

iR

iR iR-B BA
m
HI/A
BAM

12
m miR R-B AR RT 1
T1
-B -BA RT T9 0
m
F5

1
BAL BALF
4

m iR- BA
i

T7 8
BALF3

B
A
AR R
BamHI Cp

18
i

T
2
LF

iR
1
LF

58
F1 LMP2A/B
BIL

B9
BamHI Wp
i n LMP-1p
ed
ART17
ART6

ART16

let
ART5
ART15

De

N
miR-B
miR-B

ART1
ART4
miR-B

A C
ART3
miR-B

EBERs
miR-B

W
LF3
miR-B
miR-B

W
miR-B

I TR W
d oriP W
V
W EBNA-LP
X
T W
b W
c W

D W
W
EBV W
G 165 kbp oriLyt
Y
H
EBNA-2
B
F

BHLF1
K Q
R U Qp
Z e P
EBNA-1 O
E

-1
L M a

F1
S

HR
-B
iR
m
2
EBNA-3C LF
BF
EBNA-3A
EBNA-3B LF1 -2 F1
BF HR
F1 R
R -B F 1-3 BH
mi R
-BH
R
mi

Green pre-miRNAs: Encode miRNA on one arm of the pre-miRNA


Purple pre-miRNAs: Encode miRNAs on both arms of pre-miRNA
(as shown in miRBase release 22)
Figure 12.1  •  Map of the Epstein-Barr virus (EBV) genome. The genome of the B95-8 strain of EBV in its circular double-stranded DNA form of
165 kbp is depicted as it is found in latently infected B cells. The genome is a linear DNA within the viral particle and is circularized on infection at its
terminal repeat (TR) elements found at the 5′ and 3′ ends of the linear molecule. The EBV genome encodes approximately 100 genes. Shown as white boxes
are the exons for coding segments of the viral genes expressed in B cells infected in vitro, including EBNA1, EBNA2, EBNA3A/B/C, EBNA-LP, LMP1, and
LMPA/B. The EBERs denoted by the white box are two small RNAs encoded by EBV. Dashed lines represent primary transcripts originating from viral promoters
denoted with a lowercase p. Also shown are the positions of the origins of replication, oriP and oriLyt, that support the latent and lytic replication of the viral
genome, respectively. See reference 23 for details of EBV’s genome and life cycle.

to tumor phenotypes.49 The EBERs usually are highly expressed in proliferate and detectably express only LMP2a (see Fig. 12.1), a viral
EBV-positive tumor cells and thus provide a convenient, sensitive gene product not directly required for cellular proliferation.56,57
assay for identifying these tumor cells.50,51 A failure of this robust immune response to EBV’s transforming
All of the transforming genes of EBV that are expressed as proteins proteins contributes to PTLD. The infected proliferating B cells in
are recognized by the host’s T-cell response.52 EBNA1, which can be the these immunosuppressed patients are of donor origin and express all
only viral protein detected in some EBV-positive tumors,53 is recognized of EBV’s transforming proteins, as well as the EBERs.58 Two types of
in an atypical response involving CD4+ T cells and its endogenous successful treatments for PTLD demonstrate the critical role of the
major histocompatibility complex class II processing.54,55 The host’s patient’s immune response in failing to limit this “iatrogenic” tumor.
cytotoxic response is sufficiently robust that patients recovering from First, if immunosuppression can be reduced for the patient such that
infectious mononucleosis lack B cells that detectably express RNAs the transplant is still tolerated, PTLD may regress. Second, several
that encode these transforming genes. The surviving EBV-infected investigative groups have amplified the donor’s T cells that are cytotoxic
B cells are in distinct, differentiated states in which they no longer for EBV’s transforming proteins before bone marrow transplantation.
168 Part I: Science and Clinical Oncology

Treatment of patients with PTLD who have these syngenic, specific EBV in maintaining its associated tumors and the importance of the
T-killer cells has cured the disease.59 These encouraging findings miRNAs it encodes in oncogenesis.
underscore the important role of the immune response in limiting Treatments for Burkitt lymphoma have evolved to favor high-dose,
survival of EBV-infected cells. The need to immunosuppress recipients short-term chemotherapies that can yield greater than 90% 5-year
of solid organ transplants increases their risk of developing PTLD. A survival rates for children with localized disease.72 This high rate has
controversy has arisen about the efficacy of treating recipients with not been achieved, however, in some centers in Africa, where the rate
drugs that inhibit EBV’s lytic cycle to prevent PTLD. If the virus of event-free survival over 12 months has been found to be only 33%
that causes PTLD is derived from its release following transplantation, for patients at all stages of the disease.73
then blocking that release could diminish the incidence of this All EBV-associated cancers contain EBV DNA and express EBNA1,
lymphoproliferation. A large retrospective study of patients at high EBERs, and all or a subset of EBV’s miRNAs (see Fig. 12.1).58 We
risk, those who are EBV seronegative and receive organs from seroposi- know less about the genesis of these other tumors, but findings with
tive donors, has shown that prophylaxis with drugs that target viral NPC provide evidence for an unexpected contribution of EBV to its
DNA synthesis during the lytic cycle (acyclovir, ganciclovir, valacyclovir, etiology. Chan and colleagues have shown that EBV infects cells that
and valganciclovir) do not decrease the incidence of PTLD, which already can be distinguished as being “preneoplastic” in the evolution
ranges from 1% to 8%.60 It is therefore essential to monitor such of NPC.74 This finding might lead one to think that EBV is merely
high-risk recipients for the potential development of PTLD after a passenger in this tumor. However, the fact that 100% of NPC
transplantation. tumors are infected with EBV and that most tumor cells retain viral
Two startling features of tumor cells freshly isolated from Burkitt plasmid genomes means that EBV must provide these tumor cells
lymphoma biopsy specimens help to explain the evolution of this with selective advantages so that the cells with EBV outgrow those
tumor when considered in the context of EBV’s transforming genes that inevitably lose the plasmid. It is not known what this selective
and the immune response to them. These tumor cells express only advantage is, but it is reasonable to hypothesize that EBV could provide
EBNA1 among the required viral transforming proteins, along with these cells with proliferative or survival signals, as it does with Burkitt
the EBERs and miRNAs, yet they proliferate.53 They also display a lymphoma cells. Viral gene expression in NPC biopsies correlates
chromosomal translocation between one of three human Ig loci and with decreased expression of host cell genes involved in antigen display,
the c-myc proto-oncogene.61,62 These translocations are likely fostered which likely facilitates the growth of these cells in vivo as well.75 Some
by expression of the activation-induced cytidine deaminase gene, which of EBV’s miRNAs are particularly well expressed in biopsies of NPCs
is essential for Ig class switching.63 The juxtaposition of an Ig locus relative to their levels in Burkitt lymphomas and cells infected in
to c-myc drives expression of the proto-oncogene in these B cells as vitro.76,77 The viral miRNAs are thus likely candidates for providing
it does in murine plasmacytomas that display similar translocations.64 NPC tumor cells with selective advantages that act in vivo.
These observations can be arranged to provide a satisfying though The many clinical and basic scientific studies of EBV and its
necessarily speculative model for the genesis of Burkitt lymphoma. associated cancers can be extracted to yield some lessons for tumor
First, EBV infects young children living in regions of central Africa viruses in general. First, the viral genomic nucleic acid remains in
in which malaria is endemic.65 Malaria is a T-cell immunosuppressive tumor cells and expresses one or more viral genes. Second, the virus
disease that decreases the children’s ability to limit proliferation of contributes information to infected cells, which provides them with
infected cells66 and increases their viral loads.67 Although the youngsters a selective advantage both in evolving toward and sustaining tumor
with severe EBV infections have increased antibody titers to viral cells. However, this information is not sufficient for tumor formation.
antigens, they have more proliferating B cells and are at increased risk Additional, multiple, rare events must occur in the infected cells for
for the chromosomal translocations, fostered by the recombination them to evolve into tumors. These additional essential events explain
mechanism, that occur in B cells and use signals at Ig loci.68 A rare both why the associated tumors develop in only a fraction of the
Ig/c-myc translocation provides the cell some undefined selective people infected with a given tumor and why the tumors usually develop
advantages. In addition, multiple viral genes including LMP1, EBNA2, only after long delays. That tumor viruses are retained in their associated
EBNA3a, and EBNA3c are shut off and the cell proliferates, acquires tumors and help sustain these tumors indicates that targeting the
additional mutations (often mutations inactivating p53), and evolves transforming genes of tumor viruses is a rational approach to the
rapidly into a Burkitt lymphoma.69 treatment of these tumors therapeutically.
Two recent findings affect this speculative model for Burkitt
lymphoma and have implications for the other EBV-associated cancers HEPATITIS B VIRUS
as well. Studies of the replication of EBV’s plasmid genome have
revealed unexpectedly that only 84% of the viral DNAs on average HBV was identified by virtue of being recognized as an antigen in
are synthesized each cell cycle.70 This defect in DNA synthesis occurs the sera of donors detected by antibodies in sera of other infected
in all cell types tested and indicates that EBV DNA must be lost from donors.78 Thoughtful analyses by Blumberg correlated the presence
proliferating cells.70 Proliferating populations of cells can remain EBV of the antigen with hepatitis, a correlation strengthened by the
positive only if the daughter cells that retain EBV genomes are provided seroconversion of a laboratory member who contracted hepatitis.78
with selective advantages, such that they outgrow their sisters that By the late 1960s, blood donated to blood banks was screened for
lose EBV. All EBV-associated tumors maintain EBV DNA as plasmids the antigen, positive samples were removed, and only negative samples
in most of the tumor cells; therefore EBV must provide all of its were used for transfusions. This early insightful intervention led to a
associated tumors one or more selective advantages. This profound significant reduction in transfusion-associated hepatitis.79 Blumberg
insight means that EBV provides lymphomas such as AIDS-related also demonstrated a striking association between HBV antibodies to
DLBCL with selective advantages, even though the EBV-negative and its antigens, and HCC.78 These early findings have been built on to
EBV-positive forms of the disease appear similar,21 or that EBV provides demonstrate that HBV does cause HCC, which is either the fifth or
selective advantages to NPC tumors in vivo even though these tumors sixth most common cancer in people today. Of the approximately
typically lose EBV on explantation into cell culture. EBV, when found 500,000 new cases of HCC in the world each year, HBV is estimated
as a plasmid in tumor cells, cannot merely be a passenger. to cause between 50% and 70% of them.80,81 Most of the rest of these
This insight has been tested with two forms of Burkitt lymphomas cases are attributable to HCV, a member of the Flaviviridae family.
and PTLD. Tumor cells were engineered to force the loss of their Two types of data have established HBV’s causal role in HCC. A
EBV plasmids. All of these tumors died by apoptosis on the loss of prospective survey of 22,707 male civil servants in Taiwan was initiated
EBV.38 Viral miRNAs rescued the Burkitt lymphomas that expressed at the end of 1975.82 Of these subjects, 3454 were found to be positive
the fewest viral genes.71 These observations underscore the role of for HBV’s surface antigen (HBsAg), indicating that they were
Viruses and Human Cancer  •  CHAPTER 12 169

chronically infected with HBV. The entire group of 22,707 members


was observed on average for 8.9 years. By the end of 1986, HCC had Pre-S1
Pre-S2
developed in 152 of the 3454 HBsAg-positive men, whereas it had
developed in only 9 of the 19,253 HBsAg-negative men. The relative
risk of the HBsAg-positive cohort for the development of HCC was
therefore 100 times greater than that for the HBsAg-negative group.83
This prospective epidemiologic study provides robust data that the
presence of HBV is strongly associated with the development of HCC.
The second type of data demonstrating that HBV can cause HCC S gene
has been derived by removing HBV from a population and determining HBV
whether the incidence of HCC declines. Taiwan began vaccinating 3.2 kbp
children in 1984, first with a plasma-derived antigen and eventually
with a recombinant antigen. Between 1984 and 1994, the incidence
of infection as monitored by the presence of HBsAg had dropped C gene
from 9.8% to 1.3% among children 12 years or younger.84 Similar
findings have been made in the Gambia, where children vaccinated P gene
during their first year develop into only 10% as many chronically
infected 9-year-olds as do unvaccinated children.85 HCC is a cancer Pre-C
that peaks between 50 and 60 years of age and rarely occurs in children
from 6 to 14 years of age. The incidence of HCC in this latter popula-
tion in Taiwan dropped from 0.64 per 100,000 per year when averaged X gene
from 1981 to 1990 to 0.36 per 100,000 per year when averaged
between 1990 and 1994, a finding that is statistically significant (P Figure 12.2  •  Map of the hepatitis B virus (HBV) genome. The 3200 bp
genome of HBV is a circular double-stranded DNA in infected cells (shown
< .01).86 This decline presumably reflects the eightfold reduction of in black). The genome in the viral particle is partially double-stranded DNA
chronic HBV infection in children at risk for the development of because of an incomplete extension of the plus strand by the viral DNA
HCC. Removal of a virus from a population with a subsequent decrease polymerase P. Shown as colored boxes are the coding segments for the structural
in an associated cancer in that population provides compelling evidence (blue) and nonstructural (orange) viral genes. The arrowheads represent the
that the virus contributes causally to the cancer. We can expect that sites at which translation of the viral proteins initiates. These viral proteins
a decline of HCC in the vaccinated adult population will be more are translated from multiple messenger RNAs: one for Pre-S1, Pre-S2, and S;
striking in decades to come. one for Pre-S2 and S; one for core and P; and one for x. See reference 378
Although it is clear that HBV causes HCC in people, it is not for details of HBV’s genome and life cycle.
clear how it does so. Researchers now invoke two distinct contributions,
direct or indirect, to explain HBV’s oncogenesis. HBV encodes one
gene, pX (Fig. 12.2), which can affect viral and cellular transcription in HCC biopsy specimens,89 and between 30% and 90% of such
and has been proposed to contribute directly to oncogenesis. HCC biopsy specimens have mutations in p53.88,89 The viral protein pX in
in general evolves in patients with marked liver cirrhosis. HBV can studies in cell culture can bind one subunit of RNA polymerase II,
contribute to that cirrhosis by providing targets for T-cell killing and as well as transcription factor IIB.91,92 It also associates with Smad4,
thereby could contribute indirectly to oncogenesis. an integral member of the transforming growth factor–β (TGF-β)
Molecular virology studies of HBV have illuminated the viral life signaling pathway, to foster this pathway’s signaling.93 How these
cycle but have yet to identify its mode of oncogenesis.87 HBV is a different transcriptional activities of pX might contribute to the evolu-
small, enveloped virus with a double-stranded DNA genome, one tion of HCC is unclear. A study has reported that pX can induce
strand of which is incomplete. The complete viral duplex DNA is markers for “stemness” in hepatocytes by activating β-catenin and
3.2 kilobase pairs (kbp) in length (see Fig. 12.2), serves as a template epigenetic upregulation of miR-181,94 leading to the hypothesis
for transcription by RNA polymerase II, and is replicated via reverse that pX may contribute to the induction of cancer stem cells in the
transcription of a greater than full-length RNA transcript of approxi- context of HCC.
mately 3.4 kb. All members of the Hepadnaviridae family preferentially HBV is often integrated in HCC tumors,95 and it has been proposed
infect hepatocytes. This tropism is apparently mediated by a cellular that integration of the viral DNA could affect transcription of nearby
receptor expressed in hepatocytes and by viral transcription that is cellular genes. This suggestion has been strengthened by the recognition
controlled in part by cellular transcription factors principally expressed that the woodchuck member of the Hepadnaviridae family contributes
in hepatocytes. Unlike most DNA viruses, HBV undergoes its complete to HCC via insertional mutagenesis.96 However, HBV has not been
life cycle to yield progeny virions, which exit from hepatocytes via found to integrate at sites that can be interpreted to affect its onco-
secretory pathways without killing the host cell. This anomalous genesis. In addition, HBV DNAs cloned from HCC biopsy specimens
behavior of hepadnaviruses means that, in the absence of an exogenous have been tested and found not to score as enhancer sequences in
function of the host, an infected hepatocyte could survive, carry out hepatoma cells in culture.97
its normal functions, and release large amounts of infectious HBV The hypothesis that HBV contributes to the development of HCC
for long periods. Accordingly, some chronically infected people have indirectly by inducing rounds of cirrhosis and subsequent liver
large amounts of HBV in their sera. regeneration is appealing. HBV infection can be acute or chronic; it
Mammalian hepadnaviruses encode pX, which is not found in the appears that acute infection correlates with a robust cytotoxic T-cell
avian species; only the mammalian members are known to cause HCC response to all viral antigens, whereas chronic infection correlates with
in their hosts. This correlation has focused interest on pX as being likely a weak T-cell response.98 These cytotoxic responses do lead to the
to contribute to the oncogenesis of mammalian hepadnaviruses. It is death of hepatocytes; however, experiments in mice transgenic for
difficult to gauge pX’s potential role in the oncogenesis of HBV; the HBV genes and in infected chimpanzees indicate that a potent
literature includes much information about it, yet no ready synthesis noncytotoxic mechanism also exists for limiting viral expression in
of this information explains such a role. Most HCC tumor biopsies infected hepatocytes.99,100 In these experiments, immune cells release
retain viral sequences encoding pX,88 but few express the protein γ-interferon, which by some means inhibits viral gene expression and
detectably.89 It has been proposed that pX associates with p53 and promotes loss of viral DNA from infected cells.99,100 The administration
inhibits its activation of apoptosis90; however, pX is not detected of interleukin (IL)-18 limits viral replication efficiently in a transgenic
170 Part I: Science and Clinical Oncology

mouse model by inducing production of both type 1 and type 2 The study of papillomaviruses in the laboratory began in earnest
interferons.101 Chronic but not acute infection correlates with the in the late 1970s when Doug Lowy, Peter Howley, and their colleagues
eventual development of HCC. How these two modes of eliminating at the National Institutes of Health in Bethesda discovered that BPV-1
infected cells would be balanced to yield long-term or chronic infection, infects and transforms a mouse fibroblast cell line, C127, in cell
the resulting cirrhosis, and the concomitant hepatocellular regeneration culture.114–116 The parental C127 cells, although immortalized, are
required for the accumulation of mutations predisposing to HCC is contact inhibited. Infection by BPV-1 or transfection of C127 cells
not known. with a bacterial recombinant plasmid containing the entire BPV-1
A role for cytotoxic T cells in the evolution of HCC has been genome yields foci of cells that are no longer contact inhibited.
modeled in mice transgenic for HBV surface proteins (see Fig. 12.2). Transformed C127 cells harbor the viral genome as a nuclear plasmid
These mice are tolerant to these viral antigens, but when the mice and express early viral genes. The viral gene products E5 (E referring
are reconstituted with syngeneic, nontransgenic bone marrow and to early and 5 referring to the fifth largest translational open reading
subsequently challenged with syngeneic, immune, nontransgenic frame [ORF]), E6, and E7117–122 contribute to this transformation
splenocytes, cirrhosis develops, and they maintain cytotoxic T cells (Fig. 12.3). Thus by the time HPVs were recognized as potential etio-
specific for HBsAg.102 These animals have long-term liver damage, logic agents in human cervical cancers in the mid-1980s, a wealth of
and by 18 to 20 months of age, HCC develops. information pointing to the transforming potential of animal papil-
Although vaccination against HBV is effective at preventing infection lomaviruses in tissue culture had been established.
and subsequent disease, there are still approximately 250 million people The seminal studies in the early to mid-1980s of zur Hausen and
in the world chronically infected with it and at risk for developing colleagues, who identified HPV DNA in cell lines derived from cervical
cirrhosis and HCC. This risk translates into approximately 700,000 cancers, checkmated a long-argued role for HSV-2 in human cervical
people dying each year, so 15% to 25% of the chronically infected cancer. The posited role for HSV-2 in cervical cancer arose from
will eventually die from it.103 Current treatments for the chronically findings that patients with cervical cancer often had antibodies to
infected include long-term use of pegylated interferon-α and nucleoside HSV-2, another sexually transmitted agent. However, their tumor
analogues such as tenofovir.104 cells lacked HSV-2 DNA, and today it is accepted that HSV-2 does
These treatments are not cures, and reduce detection of viral antigens not contribute causally to cervical cancer. This early error provides
in only 10% of patients. Important to note, they do not eliminate an important lesson to researchers and epidemiologists trying to identify
the viral DNA resident in hepatocyte nuclei, which sustains the chronic biologic agents that contribute to cancer. Proof by today’s standards
infection. Much necessary work is now focused on developing effective requires a “smoking gun” (in this case, the gun includes the viral
treatments for eliminating HBV from infected people. genome and its expression of viral genes in cancer cells).

HUMAN PAPILLOMAVIRUSES
Cervical cancer is caused by HPVs. Approximately 200 genotypes of
HPVs have been identified to date. Most HPV genotypes infect LCR
squamous epithelia lining the skin; a subset are mucosotropic and E6
infect stratified, squamous epithelia lining the anogenital tract and
oral cavity. A subset of these mucosotropic HPVs, the so-called “high-
risk” HPVs, are associated with more than 99% of human cervical L1
cancers, other anogenital cancers, and a subset of squamous carcinomas E7
of the head and neck, particularly those of the oropharynx. HPV-16
and HPV-18, the high-risk HPVs most common in cancers, are present
in more than 85% of human cervical carcinomas. The mucosotropic HPV
HPVs are transmitted sexually and represent the most frequently 8 kbp
detected sexually transmitted disease. The association of specific
E1
mucosotropic HPVs with human cancers was first recognized in the
1980s when zur Hausen and associates at the German Cancer Research
Institute in Heidelberg detected the presence of then-novel HPV
genotypes in human cervical cancers and in cell lines derived from L2 E2
such cancers.105,106 In these cell lines, which include HeLa cells, the
HPV DNA often is integrated into the host genome, and only a
subset of viral genes, E6 and E7, is expressed.107,108 This discovery led
E5 E4
to the hypothesis that HPV E6 and E7 genes contribute to cervical
cancer, a premise now well supported by experimental research.
An association of papillomaviruses with cancer was first demon- Figure 12.3  •  Map of the human papillomavirus (HPV) genome.
Indicated by the circle is the approximately 7900 bp circular double-stranded
strated in the early 1930s with the recognition that a subcellular, DNA genome of HPV-16 as found in viral particles and infected cells. Shown
transmittable (i.e., infectious) agent causes squamous carcinomas in by the boxes outside the circle are the various translational open reading frames
cottontail rabbits.109 The agent was later identified to be a virus, (ORFs) that encode the viral proteins, including the early (E) and late (L)
cottontail rabbit papillomavirus (CRPV), that induces warts in the ORFs. Most early ORFs (those highlighted in orange) are found expressed
rabbits. In a subset of these infected rabbits, cancers develop at the throughout the viral life cycle within stratified squamous epithelia, whereas
original site of the CRPV infection. Other animal papillomaviruses the late ORFs, which encode the capsid proteins (highlighted in blue), and
induce frank cancer. Bovine papillomaviruses (BPVs), which represent E4 (highlighted in green), are selectively expressed in the productively infected,
a class of papillomaviruses that induce fibropapillomas and are character- terminally differentiated epithelial cells. Note that among the early ORFs are
ized by hyperplasia of both the dermal fibroblasts and epidermal E6 and E7, the two ORFs encoding like-named oncoproteins commonly
found expressed in HPV-associated anogenital and oral cancers. RNA synthesis
epithelial cells, can induce epithelial tumors of the alimentary canal of papillomaviruses is complex, yielding many potential messenger RNAs, all
in cows.110 Such tumors are thought to arise when animals ingest of which terminate at polyadenylation sites located at the end of the E5 and
bracken fern, which contains a potent chemical carcinogen, L1 open reading frames. The long control region (LCR) encodes multiple
quercetin.111–113 Thus papillomaviruses and chemical carcinogens act cis-acting elements that regulate viral transcription and synthesis of viral DNA.
together to induce tumors in cattle. See reference 379 for details of the HPV genome and life cycle.
Viruses and Human Cancer  •  CHAPTER 12 171

In 1985, both the zur Hausen and Howley laboratories reported that E6 and E7 both can bind additional cellular factors, and these
that HPV DNAs (see Fig. 12.3) were integrated in chromosomal interactions also may contribute to HPV-associated carcinogenesis.
DNAs in cell lines derived from cervical cancers.107,108 This finding Studies in HPV-16 transgenic mice strongly support this premise.164–167
initially led to speculation that HPVs contribute to cervical cancer Which of these many interactions contribute to their oncogenic
by integrating in or nearby to disrupt the function of cellular genes potential largely remains to be determined.
that protect against cancer (i.e., tumor suppressor genes) or activate E6 proteins from multiple papillomaviruses bind to cellular proteins
the cellular genes that can promote cancers (i.e., proto-oncogenes), other than p53 and E6AP. These other interacting partners include
akin to the mechanism of oncogenesis by certain oncogenic avian and p300168,169; paxillin170,171; E6 target protein 1 (E6TP1)172; interferon
rodent retroviruses. However, a role of HPV as an insertional mutagen regulatory factor 3 (IRF3)173; E6 binding protein 1 (E6BP1)174; Bak175;
in cancer is not consistent with its different sites of integration in protein kinase PKN176; myc177; the mammalian homologue of Drosophila
different cancers, which rarely are found to be near known or suspected discs large tumor suppressor gene product (DLG)178,179; Scribble180;
tumor suppressor genes or proto-oncogenes. Rather, it is likely that MAGI1181; and MUPP1.182 In addition, E6 can induce expression of
the integration of the HPV genome leads to the selective, and in some telomerase activity at least in part through its interaction with multiple
cases, upregulated expression of two viral genes, E6 and E7 (see Fig. isoforms of NFX1,183–186 a property that correlates with the immortal-
12.3), which encode gene products that directly contribute to cancer. izing potential of E6. In many cases, including but not limited to
The mechanism for this upregulation remains poorly understood but p53, the proteins E6 interacts with are targeted for proteasomal degrada-
may reflect (1) derepression of E6 and E7 expression from the viral tion largely through E6’s recruitment of a ubiquitin ligase, E6AP. The
promoter resulting from the disruption of a viral transcription factor, importance of E6AP in E6-mediated cervical carcinogenesis has been
E2, that can repress their transcription123; (2) an increase in the stability clearly demonstrated.187
of the E6 and E7 mRNAs resulting from the disruption on integration In addition to binding and degrading pRB, E7 can bind to other
of an mRNA instability element present in the 3′ end of the E6 and cellular proteins, p107 and p130, that are related to pRB protein188
E7 mRNAs124; or (3) increased transcriptional initiation from the and interact with different members of the E2F family of transcription
viral promoter directing expression of E6 and E7 after integration of factors.189,190 The inactivation of the pocket proteins drives carcino-
the viral DNA. The recognition of the increased expression of E6 and genesis in the head and neck region191 but is insufficient to do so in
E7 in cervical carcinomas, coupled with the knowledge that E6 and the cervix.192 E7 also is argued to associate with cyclins188,193–195 and
E7 contribute to the transforming potential of BPV-1 in mouse to inactivate the cyclin-associated kinase inhibitors p21 and p27.196–198
fibroblasts, provided the impetus to examine the tumorigenic activities A role for E7’s inactivation of p21 in cervical carcinogenesis has been
of these two viral genes. demonstrated.166 Thus E7 can associate with and/or alter the activities
Evidence for a critical role of increased expression of HPV E6 and of multiple cellular factors that themselves interact normally and thereby
E7 in the genesis of cervical cancers comes from multiple studies: (1) contribute to the normal regulation of the cell cycle. Still other
cervical epithelial cells harboring integrated HPV-16 DNA have a interactions have been identified between E7 and cellular factors,
selective growth advantage over cells harboring normal extrachromo- including the S4 subunit of the 26 S proteasome199; Mi2beta, a
somal viral genomes, and this growth advantage correlates with the component of the NuRD histone deacetylase (HDAC) complex200;
increased expression of E6 and E7125; (2) E6 and E7 bind and inactivate the Forkhead domain transcription factor MPP2201; the transcription
the tumor suppressor gene products p53 and retinoblastoma protein factor AP1202; insulin-like growth factor-binding protein 3203; TATA
(pRB), respectively126,127; (3) p53 and pRB are wild type in cell lines box-binding protein (TBP)204,205; TBP-associated factor-110206; and
derived from HPV-positive cervical cancers, whereas they are mutated a novel human DnaJ protein, hTid1.207 Recently, E7 has been shown
in HPV-negative cervical cancer–derived cell lines128; and (4) the to alter proteins that modify chromatin,200,208–210 raising the possibility
expression of the E6 and E7 viral genes is required for survival of that E7 causes global changes in host gene expression. It remains
cervical cancer–derived cell lines,129–134 and in the case of E7, main- unclear whether any of these targets of E7 contribute to E7’s oncogenic
tenance of a tumor phenotype in the context of mouse models for potential.
HPV-driven cervical carcinogenesis is required.135,136 Together, these Appreciation of the role of HPV is growing, not only in anogenital
observations strongly support the hypothesis that E6 and E7 contribute cancers such as cervical cancer, but also in head and neck cancers of
causally to human cervical cancers at least by blocking the functions the oral cavity and in skin cancers.211–214 The multiple interactions of
of cellular tumor suppressors. E6 and E7 with cellular proteins that have regulatory functions is
The E6 and E7 genes from the high-risk HPVs are transforming consistent with the contribution of E6 and E7 to cancers through
in tissue culture. They act independently or synergistically to immortal- multiple mechanisms. Studies in tissue culture strongly support the
ize multiple cell types, including human foreskin keratinocytes, cervical hypothesis that continued expression of E6 and E7 is required for the
epithelial cells, or mammary epithelial cells.137–142 In addition, E7 continued growth of cervical cancer cells.129–133 Studies in mouse models
cooperates with an activated ras to transform baby rat kidney or human also support this premise.135,136 These studies point to the potential
cervical epithelial cells.143–145 The oncogenic properties of high-risk value of developing drugs that interfere with the expression or function
HPV E6 and E7 in vivo have been validated through the characterization of these viral oncogenes as means for treating HPV-associated cancers.
of HPV transgenic mice in which expression of E6 and E7, as well Cervical cancers take decades to arise in most patients after initial
as a third viral oncogene, E5, predisposes animals to epithelial cancers infection with high-risk HPVs. During this time, the HPV must
of the skin, cervix, anus, and head and neck region.146–156 persist in the patient. Strategies that can interfere with viral persistence
E6 and E7 are best known for their ability to associate with the may prove effective in preventing the development of cancer. E6 and
cellular tumor suppressors p53 and pRB, respectively.126,127 The discovery E7 are important to the replicative phase of the HPV life cycle and
of these interactions represents a major advance in our understanding therefore for viral persistence, indicating that they are also appropriate
of the mechanisms of oncogenesis: the inhibition of tumor suppressors targets for antiviral and antitumor drug development. Recent studies
predisposes cells to evolve into tumors. E6 induces degradation of have used organotypic tissue culturing to recapitulate the life cycle of
p53 via recruitment of a ubiquitin ligase, E6AP.157,158 E6 inhibits p53 HPVs in fully differentiating, stratified squamous epithelial cells. These
protein’s transcriptional regulatory activities in tissue culture cells.159,160 studies have implicated E7 in reprogramming cells within the terminally
Association of E7 with pRB also promotes the degradation of pRB161,162 differentiating compartment of the epithelia to support the amplification
and disrupts the capacity of pRB to bind and functionally inactivate of the viral DNA genome, likely through its inactivation of pRB.215
the cellular E2F transcription factors.145,163 Although these abilities of The inactivation of p53 by E6 may be necessary for viral replication
E6 and E7 to inactivate p53 and pRB, respectively, likely play an by inhibiting cellular stress responses elicited by E7’s inactivation of
important role in their oncogenic potentials, it is important to recognize pRB.216 At least two more HPV proteins, E1 and E2 (see Fig. 12.3),
172 Part I: Science and Clinical Oncology

contribute to the replication of the viral genome.217 E1 and E2 bind


5' LTR 3' LTR
to an origin-of-DNA replication on the viral genome.218 E1 is a DNA
helicase that unwinds the viral double-stranded DNA genome at its
origin and, together with E2, recruits cellular DNA replication proteins
gag pol
that then synthesize the viral DNA.218–220 Thus E1 and E2 both represent
potentially useful targets for intervening in the viral life cycle.
So far, we have focused on the role of certain mucosotropic HPVs pro env
in causing human cancer. There is a growing appreciation that certain tax
cutaneous HPVs may also give rise to skin cancers. This is most clearly
established for patients with the genetic disease epidermodysplasia rex
verruciformis (EV). EV patients, who carry homozygous mutations
in one of two genes, EVER1 and EVER2, identified so far, are highly
susceptible to warts caused by certain cutaneous HPVs (e.g., HPV-5, Figure 12.4  •  Map of the human T-cell leukemia virus I (HTLV-I)
HPV-8), and these warts can progress to squamous cell carcinoma, genome. Indicated by the line drawing is the approximately 9000 bp DNA
particularly at sun-exposed sites.221 A recently discovered mouse proviral form of the HTLV-I genome as it is found integrated into the host
genome. The genome as present in the viral particle consists of two copies of
papillomavirus that causes cutaneous warts in laboratory mice has a single-stranded positive-strand RNA. Shown are the long terminal repeats
been shown to induce warts and squamous cell carcinoma in mice (LTRs, in green) flanking the unique region that contains the translational
treated with ultraviolet irradiation).222 Interesting to note, the E6 open reading frames (boxes) for structural (blue) and nonstructural (orange)
protein of this virus shares the ability of EV-associated HPV-8 E6 viral proteins. The LTRs contain cis-acting elements required for transcription
proteins to inactivate the Notch and TGF-β signaling pathways.1,223 and replication of the viral genome. The structural genes are transcribed late
This mouse papillomavirus may therefore provide an informative in the viral life cycle from the 5′ LTR, whereas the nonstructural proteins are
animal model for understanding the role of cutaneous HPVs in skin transcribed early from the 5′ LTR from different, spliced transcripts. See reference
cancer, the most common human cancer. 380 for details of the genome and life cycle of HTLV-I.
One desirable long-term public health strategy for dealing with
this and other human tumor viruses is the generation of an effective
prophylactic vaccine to prevent initial infection. Such vaccines are
now available and have been shown not only to efficiently prevent is a complex retrovirus that encodes multiple ORFs in addition to
HPV infection but also to reduce significantly the risk of developing the gag, pol, and env genes common to simple retroviruses (Fig. 12.4).
HPV-associated neoplastic disease. However, none of these additional viral genes is obviously related to
known proto-oncogenes, and all are thought to affect the viral life
HUMAN T-CELL LEUKEMIA VIRUS TYPE I cycle either directly or indirectly by affecting the host cell.230,231 It is
accepted that one viral protein, Tax (see Fig. 12.4), which regulates
Adult T-cell leukemia or lymphoma (ATLL), a tumor of CD4+ T both viral and cellular gene expression, is a major contribution of
cells, is caused by human T-cell leukemia virus type I (HTLV-I), the HTLV-I to leukemogenesis.232 It is also evident that viral infection
only retrovirus now accepted as being oncogenic in people. HTLV-I precedes onset of ATLL by 50 years or more, that many cells are
is found worldwide, with approximately 10 to 20 million people infected, and that this clonal tumor develops in only a minority of
estimated to be infected today. This number is likely an overestimate infected people. These combined observations indicate that multiple
because of a failure to distinguish serologically between HTLV-I and rare events in an HTLV-I–infected CD4+ T cell must occur for that
HTLV-II.224 HTLV-I is particularly prevalent in restricted sites, cell to evolve into ATLL.
including southern Japan, the Caribbean, and West Central Africa. Multiple approaches demonstrate that Tax can transform cells in
ATLL is prevalent in areas in which HTLV-I is common, and it is culture and be oncogenic in animal models. The introduction of a
estimated that ATLL will develop in as many as 5% of HTLV-I–positive vector expressing Tax into established, adherent rodent cells can
carriers in their lifetime.225 HTLV-I also causes a progressive, paralytic transform them to grow in an anchorage-independent fashion.231 Strains
myelopathy termed HTLV-I–associated myelopathy or tropical spastic of rodent cells can be transformed with Tax in combination with the
paraparesis. This neurologic disorder appears to arise preferentially in ras oncogene to yield cells tumorigenic in nude mice.233 Tax also has
patients whose human leukocyte antigen (HLA) haplotypes fail to been recombined into herpesvirus saimiri and introduced into resting
limit viral load.226 human T cells. These infected cells can proliferate and yield infected,
The data that link HTLV-I causally to ATLL are varied. ATLL immortalized progeny, whereas the Tax-negative parental virus cannot
can occur in familial clusters. It is characterized by an average age at do so.234 In accordance with these data, variants of HTLV-I from
onset of 56 years and proves rapidly fatal, with 50% of patients dying which the Tax gene has been deleted no longer can immortalize human
within 6 months of diagnosis.225 In general, patients have antibodies T cells in culture.235 These results in culture are paralleled and bolstered
to HTLV-I–encoded proteins,227 and in 88 of 88 primary biopsy by others in transgenic animal models. Expression of Tax from the
specimens of ATLL examined, all had single copies of integrated HTLV-I long terminal repeat (LTR; the viral promoter) in transgenic
HTLV-I proviruses.228 The presence of the provirus of HTLV-I in all mice leads to mesenchymal tumors.236 When its expression is directed
of the tumors makes it likely that infection with the virus is an early, to lymphoid cells, leukemias develop in the transgenic animals.237
contributing event in the evolution of the tumor. The presence of However, the exact means by which Tax transforms cells in culture
ATLL in familial clusters is consistent with the routes of transmission or is oncogenic in animal models is not obvious.
of HTLV-I. HTLV-I is passed from male to female via semen and Tax can be considered a paradigm for viral proteins that affect
from mother to child by breast milk. The latter route has been host cells in that it has multiple, distinct functions not found in any
demonstrated prospectively. Encouraging carrier mothers to refrain one cellular protein. Apparently HTLV-I during its evolution has
from breastfeeding has decreased transmission of HTLV-I to their assimilated multiple cellular activities in this one gene product. Tax
children by 80%.229 It appears highly likely that this form of public can be viewed as having at least three kinds of activities: it activates
health intervention will lead to a corresponding decrease in ATLL in transcription via nuclear factor–κB (NF-κB) and CBP, the cyclic
Japan in the future. Such a decrease would constitute formal proof adenosine monophosphate response element-binding (CREB) protein;
of the oncogenic role of HTLV-I in ATLL. it inhibits transcription, perhaps through binding HDAC1, and it
HTLV-I clearly differs from the highly oncogenic animal retroviruses inhibits several tumor suppressor gene products.231,238–242 Tax potently
that encode oncogenes derived from cellular proto-oncogenes. HTLV-I activates transcription from HTLV-I’s own LTR243 and activates the
Viruses and Human Cancer  •  CHAPTER 12 173

promoters for IL-2, IL-2 receptor α chain, and c-fos.244–246 This originally was discovered to inhibit expression of the sense mRNAs,
transcriptional activation obviously could contribute to proliferation including those that encode Tax,260 through its interaction with CREB
of an infected T cell. It also can activate the promoter for Bcl-xL and proteins261 and p300/CBP.262 Of relevance to ATLL, knockdown of
thereby help to inhibit apoptosis.247 Tax can positively regulate transcrip- expression of HBZ in ATLL-derived cell lines led to suppression of
tion by binding CREB and CBP, as well as some members of the proliferation.259 Overexpression of the spliced HBZ could induce the
NF-κB family.248,249 Tax not only binds members of the NF-κB family proliferation of ATLL cells, whereas overexpression of usHBZ could
directly, but it also can activate NF-κB’s homing to the nucleus by not induce such proliferation.258 In the context of transgenic mice,
binding to IκBα and promoting its degradation.250,251 Some of Tax’s expression of the HBZ gene from the CD4 promoter/enhancer led to
protein-protein associations have been functionally validated through increases in the frequency of CD4+ T cells, systemic inflammation,
chromatin immunoprecipitations that have documented the binding and the spontaneous development of T-cell lymphomas after a long
of Tax, CREB, and CBP to HTLV-I’s LTR in intact, HTLV-I–trans- latent period in more than a third of the mice,263 all hallmarks of
formed T cells.252 human patients infected with HTLV-I. The HBZ protein has recently
Tax also can inhibit transcription. It has been shown to inhibit been found to activate E2F transcription factor 1 (E2F1)–dependent
both expression of the β-DNA polymerase gene239 and some promoters transcription by targeting pRb/(E2F1) leading to dysregulation of the
regulated by CBP/p300.253 It has been proposed that the latter inhibition cell cycle. This activity is much like that of the HPV E7 oncoprotein
occurs by Tax’s binding to CBP and effectively sequestering CBP such complex which activates the transcription of genes under E2F1
that it is unavailable to bind other DNA-binding transcriptional factors. control that are critical for DNA replication and cell cycle progres-
It also has been shown that Tax binds HDAC1 as measured by co- sion.264 These studies highlight the potential importance of antisense
immunoprecipitations.236 Were Tax to tether HDAC1 to a promoter, transcripts arising from human retroviruses, contributing to their
the resulting localized histone deacetylation presumably would lead pathogenicity.
to its decreased support of transcription.254 A genome-wide analysis of ATLL patients revealed reactivating
A third activity of Tax is its inhibition of some cellular tumor mutations in several cellular genes including phospholipase-Cγ1, protein
suppressors. Tax has been shown to bind to and inhibit the function kinase Cβ, caspase recruitment domain-containing protein 11, discs
of p16INK4A.242 p16INK4A binds cyclin-dependent kinase 4 (CDK4), a large homolog 1, vav guanine nucleotide exchange factor 1, and cluster
kinase that, when activated, can phosphorylate pRb, yielding release of differentiation 28, in that order of prevalence, and show functional
of E2F transcription factors and promotion of the G1 to S transition overlap with the Tax interactome, perhaps explaining how ATLL
of the cell cycle. Tax, on binding p16INK4A, can increase the kinase becomes independent of Tax.265
activity of CDK4 kinase.242 p16INK4A often is found to be mutationally
inactivated in tumors. Consistent with Tax’s functionally inactivating HUMAN HEPATITIS C VIRUS
p16INK4A, p16INK4A was shown to be wild type in sequence in two
HTLV-I–infected T-cell lines in which Tax is expressed, but deleted Human HCV is accepted as an etiologic agent for HCC along with
in four uninfected T-cell lines.242 Tax also appears to bind cyclin D3 HBV. Approximately half of the cases in the United States266 and 25%
and thereby may foster, by a second means, the activity of CDK4 globally267 are ascribed to HCV. Infection with HCV constitutes a
and CDK6.240 Tax’s binding to p16INK4A and cyclin D3 would inhibit twentyfold risk for men in Taiwan of developing HCC.268 HCV
control of the cell cycle and promote cell proliferation. Tax also can represents the most common chronic viral infection among bloodborne
inhibit the transcriptional activity of p53 by an NF-κB–dependent pathogens in the United States, where the rate of infection during
mechanism that culminates in the phosphorylation of p53 as certain the period from 1988 to 1994 was estimated to be 1.8%.269 The
residues.241 Such an inhibition of p53 is likely to limit its induction WHO estimates the worldwide infection rate at 3%, yielding more
of apoptosis and increase the rate of survival of HTLV-I–infected, than 170 million infected persons.270 The rates of infection vary widely,
proliferating T cells. with rates as low as 0.6% in Germany to as high as 22% in Egypt.269
The multiple activities of Tax likely contribute to HTLV-I’s associ- Infection is thought to arise primarily from the use of shared needles
ated leukemogenesis but must be insufficient for the development of among intravenous drug users, blood transfusions, and contaminated
ATLL. Multiple T cells are initially infected by HTLV-I, but over the syringes in medical settings in developing countries.269 The establishment
course of the 50 to 60 years of its development, only one infected of sensitive tests for identifying contaminated blood and blood products
cell and its progeny give rise to the tumor. The additional genetic and fortunately has now greatly reduced the rate of infection in the general
epigenetic events necessary for this evolution are not known. It also population in developed countries. As with people chronically infected
is clear that the expression of viral genes in infected cells is surprisingly with HBV, HCC in HCV-positive individuals correlates with chronic
low; measurements of RNAs encoding Tax indicate that between 0.1% hepatitis and cirrhosis. Di Bisceglie271 has estimated that cirrhosis
and 10% of freshly harvested, infected T cells express any Tax message develops in 20% of people chronically infected with HCV per decade,
in vivo.255 The fact that Tax expression is absent in approximately and by two decades, HCC develops in 2% to 7% of chronically
60% of ATLL indicates that a role of this viral gene in cancer develop- infected people. The hyperproliferative state induced by chronic hepatitis
ment must be transient in nature,256 and this finding has led to and cirrhosis is argued to lead to an accumulation of genetic changes
additional studies, described later, that have identified another HTLV-I and contribute to the onset of liver cancer. What specifically HCV
gene that is potentially as important, if not more important, in ATLL. contributes to this scheme is not known. Fortunately, potent antiviral
One of the hallmarks of ATLL is that, whereas the 5′ untranslated drugs that can clear infection have been developed, so the incidence
region (UTR) of HTLV-I that drives expression of Tax is often disrupted of HCV-caused HCC should decrease rapidly.
in ATLL, the 3′ UTR is not disrupted.256 Analysis of HTLV-I transcripts HCV is a flavivirus, indicating that it is unique among the human
led to the realization that the viral genome gives rise to antisense tumor viruses for having RNA as its only genetic material (Fig. 12.5).
transcripts,257 one arising from the 3′ UTR that is spliced, and the It was identified in 1989 during a search for the causal agent of non-A,
other arising from within the Tax gene that is unspliced.258 Both non-B hepatitis.272 This flavivirus contains a 9.6-kb, positive-stranded
encode for highly related proteins referred to as HTLV-I bZIP factor RNA as its genome, which encodes a single translation product of
(HBZ). Important to note, HBZ is found expressed in all ATLLs.259 approximately 3000 amino acids (see Fig. 12.5). This polyprotein is
The two forms of HBZ, which differ by only nine amino acids at cleaved by both cellularly and virally encoded proteases to yield at
their N-terminus, contain three domains: an N-terminal activation least 10 proteins.273 Inhibitors of these viral proteases are being
domain, a central domain, and basic ZIP domain in the C-terminus. developed, which do reduce the levels of HCV RNA in the plasma
The version of HBZ arising from the spliced mRNA, sHBZ, is more of treated patients for short times.274,275 The study of HCV has been
abundant than that arising from the unspliced mRNA, usHBZ. HBZ daunting for at least two reasons. Its sequence varies in infected people
174 Part I: Science and Clinical Oncology

and enter cells have not yet been fully elucidated, it is clear that HCV
2A 4A 5C
particles on binding serum molecules such as high-density lipoprotein
p22 gp35 gp70 p21 p70 p27 p56 p66
become more infectious.284 This binding would lower their buoyant
C E1 E2/NS 1 2B NS3 4B 5A 5B
5' 3' density and mediate binding to a cellular receptor such as the human
scavenger receptor class B type I.284,285 Virus grown in cell culture
Figure 12.5  Map of the hepatitis C virus (HCV) genome. The would not be exposed to these serum molecules and thus would be
9500-nucleotide-long positive-strand RNA of HCV is shown in black. It less infectious.
encodes a single 3000–amino acid polyprotein that is proteolytically cleaved One promising path to study HCV infections in cell culture is to
into mature proteins (indicated on the map by labeled boxes with alternative generate hepatocytes by inducing their differentiation from either
names indicated above the boxes) by virally encoded proteases (NS3/4A and human embryonic stem cells (hESCs) or induced pluripotent stem
possibly 2B). At the 5′ and 3′ ends are short noncoding regions (NCRs) that cells (IPSCs). Wu and colleagues286 have generated hepatocytes through
contain signals for replication of the viral genome. The 5′ NCR also contains differentiation of both types of stem cells in culture and infected these
an internal ribosome entry site (IRES). Structural proteins are shown in blue. hepatocyte-like cells with derivatives of HCV isolated from cell culture
Nonstructural proteins, including proteases, helicases, and RNA polymerase,
are shown in orange. See reference 381 for details of HCV’s genome and and with serum isolates. Both forms of the virus yielded successful
life cycle. infections, each of which secreted core antigens.286 The success of
infecting these hepatocyte-like cells with serum isolates of genotypes
1A and 1B is particularly desirable because other culture systems do
not support infection by serum isolates in general and those of genotypes
such that there are six recognized genotypes with multiple subtypes 1A and 1B in particular. The general application of hESCs and IPSCs
among patients and a spectrum of quasispecies within any one infected after their differentiation to hepatocyte-like cells for study of infection
individual.276 The different genotypes vary substantially in the success by HCV will depend both on the efficiency of this differentiation
with which they can be treated.277 Members of these quasispecies and the practicality of carrying out this differentiation with large
within one patient vary by 1% to 2% in their sequence.278 In addition, numbers of precursors.
until quite recently, no cell culture for HCV has been available. Evidence for a direct role of HCV in HCC has come from studies
Researchers have made heroic efforts to overcome this hurdle and of the HCV Core protein. The HCV Core protein can cooperate
have met with partial success. with an activated form of the ras oncogene to transform primary
In 1999 Lohmann and colleagues279 described the replication of a rodent cells in tissue culture.287 HCC develops in mice transgenic for
subgenomic derivative of HCV in a cell line derived from a human the HCV Core protein.288 Several potential mechanisms have been
HCC. These subgenomic replicons were difficult to establish, but once invoked to explain the transforming potential of HCV Core protein.
established, they exhibited bona fide characteristics of flaviviral nucleic It has been found to enhance cell proliferation through the stimulation
acid replication. In each cell, 1000 to 5000 molecules of plus-strand of mitogen-activated protein kinase.289,290 In addition, HCV Core
RNA were present; minus-strand RNA was present at 10% to 20% protein can inactivate a transcription factor, lZIP, and this inactivation
of the level of the plus-strand RNA; and this viral RNA replication correlates with transformation in rodent cells.291 More recently, the
was insensitive to treatment of cells with actinomycin D.279 This mode Core protein has been found to activate STAT3, and this activation
of nucleic acid replication in cells would allow the elucidation of the may contribute to its transforming potential.292 Whether the HCV
cis and trans elements it requires, but with an unexpected twist. The Core protein contributes directly to human HCC is uncertain. Neither
efficient replication of the subgenomic derivatives of HCV requires replication of chimeric HCV in cell culture nor infection of chimeric
mutations in at least two viral genes that enhance replication in cells mice populated with human hepatocytes will allow ready testing of
but abrogate infectivity of intact HCV RNA.280 Chimpanzees are the the possible role of the Core protein in HCV’s oncogenesis.
only nonhuman host for HCV. The parental strain of HCV used to HCV infection alone is not sufficient to give rise to HCC, indicating
derive the subgenomic replicons is infectious in this primate host, that other events must occur that contribute to this cancer. Exome
whereas a derivative of it, which contains the mutations required for sequence analyses have begun to identify changes in cellular genes
the efficient replication of the subgenomic replicons, is not infectious that contribute to HCC. One such study293 identified mutations in
in this primate host.280 This finding must limit the conclusions to be a number of known cancer-related genes such as p53 and CTNNB1,
drawn from the analyses of replication of these mutated subgenomic which previously had been identified as being mutated in HCC, as
replicons in cells. This finding also indicates that the generation well as mutations in ARID2, DMXL1, and NLRP1. The mutations
of the many sequence variants that constitute the quasispecies in ARID2 were all found to disrupt the coding for its gene product,
within any one infected patient may contribute to the successful which is consistent with ARID2 being a tumor suppressor in HCC.
replication of HCV. ARID2 is a subunit of the polybromo- and BRG1-associated factor
Earlier attempts at developing cell cultures that support the chromatin remodeling complex, which facilitates ligand-dependent
replication of HCV have been substantively advanced by the transcriptional activation by nuclear receptors. Interesting to note,
identification of an isolate of HCV genotype 2a, JFH1, which replicates mutations in ARID2 selectively arise in HCV-associated HCC and
in a human HCC–derived cell line after transfection of its RNA.281,282 are infrequently observed in HBV-associated HCC.293 The significance
A chimeric HCV genome consisting of a portion of this isolate and of this finding remains unclear.
a portion of a different isolate of genotype 2a strain has been found Potent inhibitors of HCV have been developed that target different
to be infectious in cell culture, and the virus derived in cell culture essential steps in viral replication. The first generation of these drugs
has been found to be infectious in chimpanzees and in mice reconstituted inhibits a protease encoded by HCV and is effective in patients at
with human hepatocytes.283 These findings will now pave the way to picomolar concentrations.294 This and other new inhibitors achieve
dissect HCV’s replicative functions molecularly. They have facilitated sustained virologic responses, defined as “undetectable HCV RNA in
the identification and testing of drugs targeted to inhibit HCV’s the serum at a 24-week follow-up,” in greater than 95% of patients
replication, such as the inhibitors of its encoded proteases. and are active against most or all genotypes of the virus.294 In the
A second important insight has been confirmed from the analyses United States, approximately 10,000 deaths are attributed to HCV
of HCV propagated in cell culture and animal models. Virus isolated each year, and today infection with HCV is the leading indication
from infected animals is more infectious on a “per-particle basis” than for liver transplantation.269 It is likely that these new treatments will
virus isolated from cell culture; this increased specific infectivity limit or abrogate HCV in patients with HCC before they are recipients
correlates with a lower buoyant density of the virus isolated from of liver transplants. Such treatment should increase the long-term
animals.283 Although the cellular receptors used by HCV to bind to success of this transplant therapy.
Viruses and Human Cancer  •  CHAPTER 12 175

KAPOSI SARCOMA HERPESVIRUS it must be providing the tumor cells with one or more selective
advantages.
The identification of Kaposi sarcoma herpesvirus (KSHV) represents KSHV encodes many genes with clear homologies to cellular genes,
the culmination of much scientific detective work. Kaposi sarcoma some of which are candidates for contributing to viral oncogenesis
(KS) was known before the AIDS epidemic,296,297 but it increased (Fig. 12.6). Three categories of these cellular homologues are particularly
markedly among HIV-positive people. Researchers therefore looked likely to be important for tumor development: cyclins; inhibitors of
for molecular evidence of an infectious agent present in KS lesions. apoptosis; and cytokines and receptors. KSHV encodes its own cyclin.
In 1994, Chang and colleagues298 used a newly developed enrichment Cellular cyclins bind specific, dependent kinases (CdKs) whose activities
procedure based on polymerase chain reaction to identify DNA promote progression through the cell cycle and are controlled by
sequences present in KS but absent in normal cells. They identified cellular inhibitors, including p16INK4A, p21CIP1, and p27Kip1. The
a new herpesvirus, KSHV (also termed human herpesvirus 8 [HHV-8]), KSHV-encoded cyclin can promote cellular proliferation in part by
which is related to EBV and herpesvirus saimiri.298 Retrospective studies overcoming these cellular inhibitors in its complex with CdK6.316
indicate that KSHV was prevalent in different parts of the world KSHV also encodes its own inhibitors of apoptosis, viral Bcl-2
before the spread of HIV and that in Africa, for example, the prevalence and viral FLIP (an inhibitor of cellular FLICE, a protein that mediates
of KSHV has not changed. What has changed there is the incidence Fas ligand-induced cell death) (see Fig. 12.6). The viral Bcl-2 shares
of KS, so that in regions where it was formerly infrequent, the its limited sequence homology with cellular Bcl-2 in the regions critical
incidence of KS rose threefold between 1988 and 1996.299 Similarly, for inhibiting apoptosis and fails to dimerize with cellular Bcl-2 and
the frequency of infection with KSHV of certain cohorts in the United Bcl-xL, thus avoiding regulation by potential cell-binding partners.317
States has not altered with the advent of HIV,300 but the incidence It can inhibit the apoptosis induced by efficient expression of KSHV’s
of KS has changed.301 KSHV also has been detected in one class of cyclin.318 Viral FLIP can inhibit apoptosis mediated by the Fas pathway
lymphomas termed body cavity–based lymphomas or primary effusion and induced by cytotoxic T lymphocytes.319 Viral FLIP also activates
lymphomas (PELs)302 and in an atypical lymphoproliferative disorder NF-κB signaling by binding to its inhibitor, IKKγ,320,321 which fosters
termed multicentric Castleman disease.303 expression of cytokines in infected cells.316
KSHV can be transmitted sexually, as evidenced by its prevalence Finally, KSHV encodes homologues of cytokines, cytokine receptors,
in a population being related to the number of sex partners of its and an intriguing protein that stabilizes cellular cytokine mRNAs (see
members.304 Its most frequent route of transmission is via saliva.305 Fig. 12.6). Its viral IL-6 may promote proliferation of PEL cells,
KSHV also is transmitted among intravenous drug users, with 30% of although they appear to be dependent on cellular IL-6 and not viral
this group being infected in parts of China.306 Its prevalence worldwide IL-6 for their continued growth.322 This dependence may be facilitated
is clearly nonuniform, with a low prevalence in the United States and by KSHV’s Kaposin gene, which is synthesized by an atypical trans-
Northern Europe, intermediate levels around the Mediterranean, and lational initiation to yield a protein formed from 23 residue repeats.
high levels in the Middle East and in sub-Saharan Africa.306 Although Kaposin activates a protein kinase to stabilize mRNAs that encode
KS was a rather rare tumor before the AIDS epidemic, it has evolved to cellular cytokines, including IL-6.323 KSHV also encodes a G protein–
become common in areas of the world in which people are coinfected coupled receptor, viral GPCR, which is likely to be pivotal for the
with KSHV and HIV. The immunosuppressive role of HIV makes it development of KS. The expression of viral GPCR in endothelial cells
likely that immunosuppression, along with KSHV, contributes to the in mice leads to KS-like tumors, whereas the individual expression of
development of KS.307 Treating HIV-positive people with highly active KSHV cyclin, KSHV Bcl-2, or KSHV FLIP does not lead to such
antiretroviral therapy (HAART) not only decreases a person’s load of tumors.324 What is particularly exciting is that cells inoculated into
HIV but also decreases the incidence of KS and, importantly, leads to mice that express viral GPCR promote tumor formation by coinoculated
a regression of the tumor in patients with KS.308 These observations cells that individually express viral cyclin or viral FLIP, or both.324
indicate that immunosuppression not only likely contributes to the These findings support a model in which viral GPCR contributes to
development but also the maintenance of KS, along with infection the development of KS by affecting neighboring cells not infected by
with KSHV. HAART also has led to a decrease in the incidence of KSHV. The model is also supported by the findings that viral GPCR
overall AIDS-related non-Hodgkin lymphomas, as well as an increased induces expression of the cellular vascular endothelial growth factor
tolerance for their treatment with chemotherapy.308,309 (VEGF) receptor 2 in endothelial cells that proliferate in the presence
KSHV is now accepted as contributing causally to KS, PEL, and of VEGF and that viral GPCR induces expression of VEGF.325 Clearly,
multicentric Castleman disease. For example, a prospective study in these observations define an autostimulatory loop in which KSHV
HIV-infected adults has identified a significant elevation in titers GPCR alone can maintain proliferation of endothelial cells. They are
of antibodies against KSHV in those in whom KS develops.310 KS consistent with findings in which mouse bone marrow cells transfected
and PEL also display intriguing features likely to reflect their viral with recombinant KSHV grow as “KS-like” tumors in nude mice.326
etiology. Single-cell assays of early KS lesions have detected KSHV in The inhibition of expression of viral GPCR in these KSHV-positive
a minority of cells that surround their vascular spaces, whereas in the cells inhibits tumor formation when the engineered cells are transplanted
more advanced, nodular lesions, more than 90% of the spindle cells into recipients.326 These data indicate that although the viral GPCR
characteristic of these lesions are KSHV-antigen positive.311 The viral may be expressed only in a minority of infected endothelial cells, it
antigen-positive cells also stain with antibodies against VEGF receptor makes an essential contribution to oncogenicity by KSHV.
3, indicating that they are lymphatic or proliferating endothelial cells.311 KSHV limits its host’s immune response. The virus suppresses
That only a subset of the cells characteristic of early KS lesions appear Toll-like receptor 4 (TLR4), which decreases the infected cell’s innate
to be infected with KSHV likely indicates that infected cells affect immune response.327 It inhibits expression of HLA-1 molecules, which
the development of their neighbors. This possibility is supported by would help to avoid recognition of infected cells by cytotoxic T cells328
the multiple cellular homologues of cytokines and receptors encoded but would increase the likelihood of their being recognized by NK
by KSHV that may allow infected cells to interact with adjacent, cells. KSHV infection also limits expression of receptors on NK cells
uninfected cells.312,313 PEL cells are B cells in origin, and in six of seven needed to recognize their infected targets,329 showing that it uses
cases examined, they have nongermline Ig mRNAs, indicating that multiple avenues to evade the immune response. This immune evasion
they are likely derived from B cells that have encountered antigen.314 also likely contributes to its oncogenicity.
However, they often fail to express B-cell activation antigens.315 PEL Recent studies of cell lines derived from PEL and human endothelial
cells usually but not always are coinfected with EBV.302,315 The role cells are providing the foundation for understanding infections by
of EBV in the etiology of this lymphoma is not yet determined, KSHV in vivo. PEL cells maintain KSHV DNA extrachromosomally
but for cases in which EBV is retained as a plasmid in the PELs, and consistently express the viral protein LANA-1 (see Fig. 12.6),
176 Part I: Science and Clinical Oncology

TR

GPCR

LANA1

vCYC vIL-6

miR-K12-9a/b

miR -K122-11
vFLIP

miR -K1 -8
miR -K1 2-7
miRK12-10
2

mi -K -6
miRR-K112-5
miR-K12-1

miR -K12

-K 2-4
miR -K112-3
-K1 2-2
2-1
K12 -
miR

miR
KSHV
160 kbp

vBcl2

LANA2

ORF50

pre-miRNAs encode miRNAs on both arms of pre-miRNA


(as shown in miRBase release 22)
Figure 12.6  •  Map of the Kaposi sarcoma herpesvirus (KSHV) genome. The genome of KSHV consists of approximately 160 kbp of linear double-
stranded DNA in the viral particle. The viral genome becomes circularized on infection via the terminal repeats found at the 5′ and 3′ ends of the linear
genome. The terminal repeats also contain the origin of plasmid replication used during the latent phase of the viral life cycle. Shown by the white boxes are
positions of the viral genes expressed during latency and/or thought to contribute to oncogenesis by KSHV. The arrows represent the positions from which
the RNAs encoding the labeled proteins are expressed. Several of these primary transcripts are polycistronic. See reference 382 for details of KSHV’s genome
and life cycle.

which is required for viral plasmid replication.330,331 The KSHV genomes lytic cycle may be resolved. A sustained, spontaneous conversion of
are found in clusters and partition randomly to daughter cells.331a 5% to 10% of KSHV-infected cells to support the viral lytic cycle
This unprecedented mechanism provides an appealing target for antiviral might allow enough infected cells to express enough KSHV GPCR
therapy. In general, these cells support the latent phase of KSHV’s to promote the bystander-dependent tumor evolution proposed by
life cycle and also express the viral cyclin and the viral inhibitor of Bais and colleagues.325
cellular FLICE, FLIP, but few other viral proteins. Some PEL cell KS is the most common malignancy among HIV-positive people.
lines support an inefficient spontaneous conversion to the lytic phase Although there are currently no effective antiviral therapies directed
of the viral cycle, which can be further induced by treatment of cells against latent infection with KSHV, multiple trials aimed at inhibiting
with tetradecanoyl phorbol acetate or introduction of a plasmid its lytic cycle appear to have achieved a decrease in the incidence of
including ORF50 (see Fig. 12.6), a viral inducer of the lytic cycle.332 KS.335 Treatment with acyclovir is ineffective, but treatment with
The released virus is infectious in human dermal microvascular ganciclovir reduced the incidence of KS from 40% to 90% in various
endothelial cells (DMVECs).333,334 These DMVEC cultures can be trials.335 Similar treatments also were partially effective in limiting the
passaged to yield populations in which all cells are infected, express incidence of PEL and Castleman disease. Whether this inhibition of
LANA-1, assume a spindle-cell morphology, and can proliferate KSHV’s lytic cycle is limiting disease by preventing infection of cells
indefinitely.333 The spindle shape is characteristic of cells in KS lesions that evolve into tumors or preventing lytically infected cells from
in vivo. Staining of these infected DMVEC-derived spindle cells contributing to a necessary tumor microenvironment is not known.
indicates that 5% to 10% of them spontaneously support early stages
of KSHV’s lytic cycle and 1% to 2% express genes diagnostic of the MERKEL CELL POLYOMAVIRUS
late stages of the viral life cycle.333 If endothelial cells infected in vivo
by KSHV display a similar distribution of cells supporting the latent, The association of viruses with AIDS-related malignancies drove
early, and late lytic phases of the viral life cycle, then the enigma of Chang and colleagues to search for a virus associated with a second
the oncogenic contribution of viral genes expressed only during the AIDS-related malignancy, Merkel cell carcinoma (MCC). This rare but
Viruses and Human Cancer  •  CHAPTER 12 177

highly aggressive skin cancer arises in elderly and in immunosuppressed MCC cells have been engrafted,357 providing a potential new therapeutic
and immunodeficient patients, often in sun-exposed regions of the approach for treating patients with this deadly cancer.
body.336 In the general population, more than a third of patients with Several transgenic mouse models have been developed in which
MCC will die of their cancer337; in immunocompromised patients, the the MCPyV tumor antigens are targeted in their expression to different
percentage rises to greater than 50%. MCC is distinguished by a unique cell types. In one case, the region of MCPyV present in an MCC
pattern of expression of neuroendocrine and epithelial-specific markers encoding for small tumor antigen and a truncated form of large tumor
expressed in Merkel cells, which are specialized mechanoreceptors antigen was placed under the control of the ROSA26 allele carrying,
cells within epithelia that transduce mechanical stimuli from the skin upstream of the MCPyV sequences, a strong transcriptional stop
(reviewed in references 338–341). Feng and colleagues342 used a state of sequence flanked by Lox P sites, allowing for tissue-specific expression
the art “digital transcriptome subtraction” (DTS) approach to identify by crossing to mice expressing Cre in a tissue-specific manner. Once
nonhuman sequences present in MCCs. DTS involves deep sequencing MCPyV genes were turned on in the epidermis, the mice developed
of complementary DNAs followed by in silico subtraction of host hyperplastic epithelium and spontaneously developed papillomas on
transcripts to divulge nonhuman sequences present in a sample.342 Using the rear torso.2,358 Both small tumor antigen and the truncated large
DTS, Feng and colleagues342 identified nonhuman sequences in four tumor antigen were found to be expressed in the epithelium and
MCC samples that had similarity but were not identical to the large tumors. Two other models were generated in which just small tumor
tumor antigen genes of previously identified human polyomaviruses, antigen was expressed in the epidermis. In neither of these models
which led them to a full-length clone of a novel polyomavirus that were MCCs observed.3,359 However, in a more recent study, cellular
they called Merkel cell polyomavirus (MCPyV). MCPyV is present aggregates with histology and marker expression mimicking that of
in approximately 80% of cases of MCC.343–345 Like other human human intraepidermal MCC were observed in embryos when investiga-
polyomaviruses, MCPyV has a circular double-stranded DNA genome tors coexpressed in the epidermis small tumor antigen and the cell
containing a well-conserved replication origin and transcriptional control fate determinant, atonal bHLH transcription factor 1 (Atoh1),
region and translational ORFs encoding for small and large tumor selectively found to be expressed in Merkel cells.4,360,361
antigens on one strand, and capsid proteins VP1, VP2, and VP3 on
the other strand. Serologic studies indicate that most of the human TREATMENT AND PREVENTION OF
population is exposed to MCPyV,346–350 likely in early childhood.346,347 VIRAL TUMORS
The virus has been detected in a number of different cell types, including
non-MCC cells of the skin.351–354 The identification of this ubiquitous Tumors associated causally with viruses are treated variously; surgical
novel human polyomavirus in the skin led other groups to screen for resections, when appropriate, are used in combination with chemo-
other human polyomaviruses in the skin using a technique for cloning therapy and/or radiation therapy. These treatments are disappointing
small circular DNAs called rolling circle amplification. This screening led in that overall survival rates are often low and thus far have not been
to the identification of many additional novel human polyomaviruses, able to capitalize on any specific antiviral therapies. Current survival
most of which have not been linked to any specific disease. rates after different therapies obviously vary with the malignancy, its
In their analyses of MCC, Shuda and colleagues345 made another stage, and the setting in which it is treated. For example, youngsters
important observation, akin to that made by zur Hausen in the context with Burkitt lymphoma have a 4-year overall survival rate of 65%.362
of HPV’s role in cervical cancer: the MCPyV genome is found clon- Approximately 45% of patients with NPC remain disease free after
ally integrated into the host genome in the cancers. Importantly, treatment for 10 years, but this value is highly dependent on the stage
these integrated copies of MCPyV carried mutations in the large at which the NPC was diagnosed.179 In the United States, 26% of
tumor antigen gene that resulted in expression of truncated large HBV-positive patients with HCC that has been surgically resected
tumor antigen gene products (these mutations did not alter the have a 5-year, local disease-free survival rate compared with 38% for
small tumor antigen gene, which is also expressed in MCC). The the analogous HCV-positive group.363 A retrospective study of German
truncated large tumor antigen proteins expressed in MCC retain patients with a mix of HBV- and HCV-associated HCC found a
the domain within the N-terminal half of the full-length protein median survival rate of 7 years.364 The 5-year median survival rate for
that mediates interaction with the cellular tumor suppressor, pRb, patients with cervical carcinoma ranges from 65%365 to 75% to 90%366
but they are missing part or all of the highly conserved adenosine in different studies. The median survival rate for patients who have
triphosphatase/DNA helicase domain required for viral DNA been diagnosed with ATLL is only 10 months.367 The overall survival
replication. This finding led to the hypothesis that in MCC, there rates of patients with KS who are HIV positive have changed with
is selection for replication-defective MCPyV genomes that retain an the introduction of HAARTs. Before this therapy was available, median
ability to inactivate pRb. Consistent with this hypothesis, knockdown overall survival was 13 months; with HAART, it surpasses 28 months.368
of expression of the MCPyV tumor antigens (both large and small The median survival for patients with PEL has been described as
tumor antigens) in MCC-derived cell lines led to cell growth arrest “dismal,” but case reports indicate that some combination therapies
and death.355 Follow-up studies by Shuda and colleagues,356 however, are helpful.369 These actuarial findings indicate that the development
led to the further discovery that specifically knocking down expression of antiviral therapies directed at the viral gene products that maintain
of MCPyV small tumor antigen, although not affecting expression tumor phenotypes is a highly desirable goal. It also obviously is desirable
of MCPyV large tumor antigen, led to growth arrest, although not to develop vaccines to prevent infections by tumor viruses. The
death, of MCPyV-positive MCC cell lines, leading to the revised development of effective vaccines that can prevent an initial infection
hypothesis that both small and large tumor antigens contribute to or induce the elimination of viral persistence is being pursued for all
MCC. MCPyV small tumor antigen was found to transform cells human tumor viruses. Success has been achieved for two of them.
at least in part by acting downstream of the mammalian target of
rapamycin signaling pathway to preserve eukaryotic translation Hepatitis B Virus Vaccine
initiation factor 4E–binding protein 1 hyperphosphorylation, resulting
in dysregulated cap-dependent translation.356 That continued expression Although four serotypes of HBV exist, highly effective vaccines for
of MCPyV large tumor antigen is specifically necessary for the survival HBV have been developed that consist only of its surface antigen,
of MCPyV-positive MCC cells and appears to correlate with large HBsAg. Today this subunit vaccine is usually synthesized with a
tumor antigen, causing an increase in levels of expression of survivin.357 recombinant DNA expressed in yeast. Vaccines for HBV began to
A drug that inhibits survivin was shown to induce programmed cell be used in the early 1980s, and since 2001, 129 countries throughout
death of MCPyV-positive MCC cells in tissue culture and prolong the world have routinely vaccinated infants and/or adolescents against
the survival of immunodeficient mice in which these tumor-forming HBV.370 These vaccinations are effective. The fraction of children
178 Part I: Science and Clinical Oncology

infected with HBV has been reported to have declined between immune responses invoked by VLP-based vaccines for specific HPV
7.5-fold and 10-fold in Taiwan and the Gambia, respectively.84,85 This genotypes. Approximately a dozen HPV genotypes are associated with
decline has been paralleled by a detectable drop in the frequency of anogenital cancers. VLP-based vaccines induce protective immunity
HCC in children in Taiwan, an age group for which this viral tumor that is highly specific for the genotype from which the VLP is generated.
is rare.86 This specificity led to the development of polyvalent vaccines composed
The current HBV vaccines apparently are free of unwanted adverse of a mixture of VLPs generated with L1 proteins from multiple
effects but can select for variants of HBV resistant to the neutralizing mucosotropic HPV genotypes. One of the available vaccines, Gardasil,
antibodies they elicit.370 One domain of the HBsAg in particular is a multivalent vaccine composed of HPV-16, HPV-18, HPV-6, and
elicits neutralizing antibodies efficiently, and mutations in it can allow HPV-11 VLPs. HPV-6 and HPV-11 are low-risk mucosotropic HPVs
for viral escape. The frequency of these escape mutants in populations that induce genital warts but do not contribute to cervical cancer.
of vaccinated children has risen significantly.371 One means to overcome The other available vaccine, Cervarix, is composed of just HPV-16
the selection for such escape mutants would be to generate vaccines and HPV-18 VLPs. It remains to be seen whether, once a population
for HBV that include more than the HBsAg as an immunogen; such is protected from HPV-16– and HPV-18–induced cancers, other HPV
vaccines are now being developed. genotypes arise to induce a greater incidence of cervical cancers than
they currently cause. For this reason, the development of vaccines
Human Papillomavirus Vaccine that provide protection against a wider variety of high-risk HPVs is
now being considered. One approach is to add L1 VLPs specific for
The second family of human tumor viruses for which an effective other genotypes to the current vaccines. Alternatively, incorporation
prophylactic vaccine can now be envisioned are the HPVs associated of the minor capsid protein L2 may be of benefit, because neutralizing
with cervical cancer, other anogenital cancers, and certain head and antibodies induced against L2 tend to be more generally effective at
neck cancers, in particular HPV-16 and HPV-18. These two viruses neutralizing multiple genotypes.377 In this regard, a stand-alone L2
account for approximately 85% of cervical cancers worldwide, and vaccine might be effective. Concern exists about whether the HPVs,
in a recent study, 16% of new infections.372 Two highly effective particularly HPV-16 and HPV-18, will evolve to become resistant to
vaccines that prevent infections by HPV-16 and HPV-18 have been VLP-based vaccines. Assuming the high effectiveness of the vaccines
approved for use in the United States and other countries. These currently under development and their worldwide distribution, and
vaccines are based on the production of viruslike particles (VLPs) given the long latency of cervical cancer, the WHO estimates that
composed of the major capsid protein of HPV-16 and HPV-18, L1, prophylactic HPV vaccines will lead to a reduction of deaths due to
which self-assembles into icosahedrons (see Fig. 12.3). These L1-based cervical cancer no earlier than 2040. Given that HPVs also cause
VLPs induce neutralizing antibodies in vaccinated individuals. Preclini- other anogenital cancers, as well as head and neck cancers that affect
cal studies with CRPV and canine oral papillomavirus demonstrated both men and women, the available vaccines are now being considered
the effectiveness of VLP-based vaccines in protecting animals from for use in vaccinating both young men and young women in the
CRPV and canine oral papillomavirus infection.373,374 Phases I and II United States and elsewhere in the world.
clinical trials likewise demonstrated the effectiveness of HPV-16
VLP-based vaccines in inducing neutralizing antibodies specific for
HVP-16 and in preventing HPV-16 infections.375,376 One issue of The complete reference list is available online at
relevance to the effectiveness of these vaccines is the specificity of ExpertConsult.com.

KEY REFERENCES
1. Parkin DP, Pisani P, Munoz N, Ferlay J. The global virus and Burkitt’s lymphoma from Ugandan 86. Chang MH, Chen CJ, Lai MS, et al. Universal
health burden of infection associated cancers. Cancer prospective study. Nature. 1978;274:756–761. hepatitis B vaccination in Taiwan and the incidence
Surv. 1999;33:5–33. 23. Kieff E, Rickinson AB. Epstein Barr virus and its of hepatocellular carcinoma in children. Taiwan
3. Rous P. A sarcoma of the fowl transmissible by an agent replication. In: Fields Virology. 2001:2511–2573. Childhood Hepatoma Study Group. N Engl J Med.
separable from the tumor cells. J Exp Med. 1911;13: 29. Pfeffer S, Zavolan M, Grasser FA, et al. Identi- 1997;336:1855–1859.
397–411. fication of virus-encoded microRNAs. Science. 89. Henkler F, Waseem N, Golding MH, et al. Mutant
4. Gross L. “Spontaneous” leukemia developing in 2004;304:734–736. p53 but not hepatitis B virus X protein is present
CH3 mice following inoculation in infancy, with 37. Humme S, Reisbach G, Feederle R, et al. The in hepatitis B virus-related human hepatocellular
AK-leukemic extracts, or AK-embryos. Proc Soc Exp EBV nuclear antigen 1 (EBNA1) enhances B cell carcinoma. Cancer Res. 1995;55:6084–6091.
Biol Med. 1951;76:27–32. immortalization several thousandfold. Proc Natl Acad 98. Chisari FV, Ferrari C. Hepatitis B virus immuno-
5. Payne LNAHGP. Diseases of Poultry. 9th ed. Ames, Sci USA. 2003;100:10989–10994. pathogenesis. Annu Rev Immunol. 1995;13:29–60.
Iowa, USA: Iowa State University Press; 1991. 47. Yajima M, Kanda T, Takada K. Critical role of 100. Guidotti LG, Rochford R, Chung J, et al. Viral
6. Cooper GM. Oncogenes. 2nd ed. Boston: Jones and Epstein-Barr virus (EBV)-encoded RNA in efficient clearance without destruction of infected cells
Bartlett Publishers; 1995. EBV-induced B-lymphocyte growth transformation. during acute HBV infection. Science. 1999;284:825–
7. Trentin JJ, Yabe Y, Taylor G. The quest for human J Virol. 2005;79:4298–4307. 829.
cancer viruses. Science. 1962;137:835–849. 61. Taub R, Kirsch I, Morton C, et al. Translocation 106. Durst M, Gissmann L, Ikenberg H, zur Hausen H.
8. Burkitt D. A children’s cancer dependent upon of the c-myc gene into the immunoglobulin heavy A papillomavirus DNA from a cervical carcinoma
climatic factors. Nature. 1962;194:232–234. chain locus in human Burkitt lymphoma and and its prevalence in cancer biopsy samples from
9. Epstein M, Achong BG, Barr YM. Virus particles murine plasmacytoma cells. Proc Natl Acad Sci different geographic regions. Proc Natl Acad Sci USA.
in cultured lymphoblasts from Burkitt’s lymphoma. USA. 1982;79:7837–7841. 1983;80:3812–3815.
Lancet. 1964;1:702–703. 70. Nanbo A, Sugden A, Sugden B. The coupling of 107. Schwarz E, Freese UK, Gissmann L, et al. Structure
10. Epstein M, Barr YM. Cultivation in vitro of human synthesis and partitioning of EBV’s plasmid replicon and transcription of human papillomavirus sequences
lymphoblasts from Burkitt’s lymphoma. Lancet. is revealed in live cells. EMBO J. 2007;26:4252– in cervical carcinoma cells. Nature. 1985;314:
1964;1:252–253. 4262. 111–114.
11. Henle G, Henle W, Diehl V. Relation of Burkitt’s 78. Blumberg BS. Australia antigen and the biology of 109. Shope RE. A transmissible tumor-like condition in
tumor-associated herpes-type virus to infectious hepatitis B. Science. 1977;197:17–25. rabbits. J Exp Med. 1932;56:793–802.
mononucleosis. Proc Natl Acad Sci USA. 1968;59: 82. Beasley RP, Hwang LY, Lin CC, Chien CS. 116. Lowy DR, Dvoretzky I, Shober R, et al. In vitro
94–101. Hepatocellular carcinoma and hepatitis B virus. A tumorigenic transformation by a defined sub-
15. de-The G, Geser A, Day NE. Epidemiological prospective study of 22 707 men in Taiwan. Lancet. genomic fragment of bovine papilloma virus DNA.
evidence for causal relationship between Epstein-Barr 1981;2:1129–1133. Nature. 1980;287:72–74.
Viruses and Human Cancer  •  CHAPTER 12 179

126. Dyson N, Howley PM, Munger K, Harlow E. The leukemia virus in the disease. Proc Natl Acad Sci 345. Shuda M, Feng H, Kwun HJ, et al. T antigen
human papillomavirus-16 E7 oncoprotein is able to USA. 1984;81:2534–2537. mutations are a human tumor-specific signature
bind to the retinoblastoma gene product. Science. 235. Ross TM, Pettiford SM, Green PL. The tax gene of for Merkel cell polyomavirus. Proc Natl Acad Sci
1989;243:934–937. human T-cell leukemia virus type 2 is essential for trans- USA. 2008;105:16272–16277.
127. Werness BA, Levine AJ, Howley PM. Association of formation of human T lymphocytes. J Virol. 1996;70: 351. Feng H, Shuda M, Chang Y, Moore PS. Clonal
human papillomavirus types 16 and 18 E6 proteins 5194–5202. integration of a polyomavirus in human Merkel
with p53. Science. 1990;248:76–79. 256. Matsuoka M, Jeang KT. Human T-cell leukaemia cell carcinoma. Science. 2008;319:1096–1100.
130. Goodwin EC, DiMaio D. Repression of human virus type 1 (HTLV-I) infectivity and cellular 356. Shuda M, Kwun HJ, Feng H, et al. Human Merkel
papillomavirus oncogenes in HeLa cervical carcinoma transformation. Nat Rev Cancer. 2007;7:270–280. cell polyomavirus small T antigen is an oncoprotein
cells causes the orderly reactivation of dormant 272. Choo QL, Kuo G, Weiner AJ, et al. Isolation of a targeting the 4E-BP1 translation regulator. J Clin
tumor suppressor pathways. Proc Natl Acad Sci cDNA clone derived from a blood-borne non-A, non-B Invest. 2011;121:3623–3634.
USA. 2000;97:12513–12518. viral hepatitis genome. Science. 1989;244:359–362. 357. Arora R, Shuda M, Guastafierro A, et al. Survivin
145. Phelps WC, Yee CL, Munger K, Howley PM. The 279. Lohmann V, Korner F, Koch J, et al. Replication of is a therapeutic target in Merkel cell carcinoma.
human papillomavirus type 16 E7 gene encodes subgenomic hepatitis C virus RNAs in a hepatoma Sci Transl Med. 2012;4:133ra156.
transactivation and transformation functions similar cell line. Science. 1999;285:110–113. 373. Christensen ND, Reed CA, Cladel NM, et al. Immu-
to those of adenovirus E1A. Cell. 1988;53:539– 281. Wakita T, Pietschmann T, Kato T, et al. Production nization with viruslike particles induces long-term
547. of infectious hepatitis C virus in tissue culture from protection of rabbits against challenge with cot-
146. Arbeit JM, Howley PM, Hanahan D. Chronic a cloned viral genome. Nat Med. 2005;11:791–796. tontail rabbit papillomavirus. J Virol. 1996;70:960–
estrogen-induced cervical and vaginal squamous 298. Chang Y, Cesarman E, Pessin MS, et al. Identifica- 965.
carcinogenesis in human papillomavirus type 16 tion of herpesvirus-like DNA sequences in AIDS- 374. Suzich JA, Ghim SJ, Palmer-Hill FJ, et al. Systemic
transgenic mice. Proc Natl Acad Sci USA. 1996;93: associated Kaposi’s sarcoma. Science. 1994;266: immunization with papillomavirus L1 protein
2930–2935. 1865–1869. completely prevents the development of viral mucosal
191. Shin MK, Pitot HC, Lambert PF. Pocket proteins 302. Cesarman E, Chang Y, Moore PS, et al. Kaposi’s papillomas. Proc Natl Acad Sci USA. 1995;92:
suppress head and neck cancer. Cancer Res. 2012;72: sarcoma-associated herpesvirus-like DNA sequences 11553–11557.
1280–1289. in AIDS-related body-cavity-based lymphomas. 376. Koutsky LA, Ault KA, Wheeler CM, et al. Proof
228. Yoshida M, Seiki M, Yamaguchi K, Takatsuki K. N Engl J Med. 1995;332:1186–1191. of Principle Study, I. A controlled trial of a human
Monoclonal integration of human T-cell leukemia 313. Moore PS, Chang Y. Kaposi’s sarcoma-associated papillomavirus type 16 vaccine. NEJM. 2002;347:
provirus in all primary tumors of adult T-cell herpesvirus-encoded oncogenes and oncogenesis. 1645–1651.
leukemia suggests causative role of human T-cell J Natl Cancer Inst Monogr. 1998;65–71.
Viruses and Human Cancer  •  CHAPTER 12 179.e1

REFERENCES
1. Parkin DP, Pisani P, Munoz N, Ferlay J. The global 24. Bloss TA, Sugden B. Optimal lengths for DNAs 44. Casola S, Otipoby KL, Alimzhanov M, et al. B cell
health burden of infection associated cancers. Cancer encapsidated by Epstein-Barr virus. J Virol. 1994;68: receptor signal strength determines B cell fate. Nat
Surv. 1999;33:5–33. 8217–8222. Immunol. 2004;5:317–327.
2. Ellerman VaOB. Experimentelle Leukamie bei 25. Baer R, Bankier AT, Biggin MD, et al. DNA 45. Mancao C, Altmann M, Jungnickel B, Hammerschmidt
Huhnern. Zentralbl Bakteriol Parasitenkd Infectionskr sequence and expression of the B95-8 Epstein-Barr W. Rescue of “crippled” germinal center B cells from
Hyg Abt Orig. 1908;46:595–609. virus genome. Nature. 1984;310:207–211. apoptosis by Epstein-Barr virus. Blood. 2005;106:
3. Rous P. A sarcoma of the fowl transmissible by an agent 26. Cai X, Schafer A, Lu S, et al. Epstein-Barr virus 4339–4344.
separable from the tumor cells. J Exp Med. 1911;13: microRNAs are evolutionarily conserved and dif- 46. Kuppers R, Schwering I, Brauninger A, et al. Biology
397–411. ferentially expressed. PLoS Pathog. 2006;2:e23. of Hodgkin’s lymphoma. Ann Oncol. 2002;13(suppl
4. Gross L. “Spontaneous” leukemia developing in 27. Chen SJ, Chen GH, Chen YH, et al. Charac- 1):11–18.
CH3 mice following inoculation in infancy, with terization of Epstein-Barr virus miRNAome in 47. Yajima M, Kanda T, Takada K. Critical role of
AK-leukemic extracts, or AK-embryos. Proc Soc Exp nasopharyngeal carcinoma by deep sequencing. Epstein-Barr virus (EBV)-encoded RNA in efficient
Biol Med. 1951;76:27–32. PLoS ONE. 2010;5(9):pii: e12745. EBV-induced B-lymphocyte growth transformation.
5. Payne LNAHGP. Diseases of Poultry. 9th ed. Ames, 28. Grundhoff A, Sullivan CS, Ganem D. A combined J Virol. 2005;79:4298–4307.
Iowa, USA: Iowa State University Press; 1991. computational and microarray-based approach 48. Samanta M, Iwakiri D, Kanda T, et al. EB virus-
6. Cooper GM. Oncogenes. 2nd ed. Boston: Jones and identifies novel microRNAs encoded by human encoded RNAs are recognized by RIG-I and
Bartlett Publishers; 1995. gamma-herpesviruses. RNA. 2006;12:733–750. activate signaling to induce type I IFN. EMBO J.
7. Trentin JJ, Yabe Y, Taylor G. The quest for human 29. Pfeffer S, Zavolan M, Grasser FA, et al. Identi- 2006;25:4207–4214.
cancer viruses. Science. 1962;137:835–849. fication of virus-encoded microRNAs. Science. 49. Nanbo A, Takada K. The role of Epstein-Barr virus-
8. Burkitt D. A children’s cancer dependent upon 2004;304:734–736. encoded small RNAs (EBERs) in oncogenesis. Rev
climatic factors. Nature. 1962;194:232–234. 30. Henle W, Diehl V, Kohn G, et al. Herpes-type Med Virol. 2002;12:321–326.
9. Epstein M, Achong BG, Barr YM. Virus particles virus and chromosome marker in normal leukocytes 50. Howe JG, Steitz JA. Localization of Epstein-Barr
in cultured lymphoblasts from Burkitt’s lymphoma. after growth with irradiated Burkitt cells. Science. virus-encoded small RNAs by in situ hybridization.
Lancet. 1964;1:702–703. 1967;157:1064–1065. Proc Natl Acad Sci USA. 1986;83:9006–9010.
10. Epstein M, Barr YM. Cultivation in vitro of human lym- 31. Robinson JE, Smith D, Niederman J. Plasmacytic 51. Wu TC, Mann RB, Epstein JI, et al. Abundant
phoblasts from Burkitt’s lymphoma. Lancet. 1964;1: differentiation of circulating Epstein-Barr virus– expression of EBER1 small nuclear RNA in
252–253. infected B lymphocytes during acute infectious nasopharyngeal carcinoma. A morphologically
11. Henle G, Henle W, Diehl V. Relation of Burkitt’s mononucleosis. J Exp Med. 1981;153:235–244. distinctive target for detection of Epstein-Barr virus
tumor-associated herpes-type virus to infectious 32. Tagawa T, Albanese M, Bouvet M, et al. Epstein-Barr in formalin-fixed paraffin-embedded carcinoma
mononucleosis. Proc Natl Acad Sci USA. 1968;59: viral miRNAs inhibit antiviral CD4+ T cell responses specimens. Am J Pathol. 1991;138:1461–1469.
94–101. targeting IL-12 and peptide processing. J Exp Med. 52. Moss DJ, Burrows SR, Silins SL, et al. The immunol-
12. Niederman JC, McCollum RW, Henle G, Henle W. 2016;213(10):2065–2080. ogy of Epstein-Barr virus infection. Philos Trans R
Infectious mononucleosis: clinical manifestations in 33. Albanese M, Tagawa T, Bouvet M, et al. Epstein-Barr Soc Lond B Biol Sci. 2001;356:475–488.
relation to EB virus antibodies. JAMA. 1968;203. virus microRNAs reduce immune surveillance by 53. Rowe M, Rowe DT, Gregory CD, et al. Differences
13. Evans AS, Niederman JC. Epstein Barr virus. In: virus-specific CD8+ T cells. Proc Natl Acad Sci USA. in B cell growth phenotype reflect novel patterns of
Evans AS, ed. Viral Infections of Humans. 3rd ed. 2016;113(42):E6467–E6475. Epstein-Barr virus latent gene expression in Burkitt’s
New York: Plenum Press; 1989:265–292. 34. Kempkes B, Spitkovsky D, Jansen-Durr P, et al. B-cell lymphoma cells. EMBO J. 1987;6:2743–2751.
14. Henle W, Henle G, Ho HC, et al. Antibodies to proliferation and induction of early G1-regulating 54. Heller KN, Upshaw J, Seyoum B, et al. Distinct
Epstein-Barr virus in nasopharyngeal carcinoma, proteins by Epstein-Barr virus mutants conditional memory CD4+ T-cell subsets mediate immune
other head and neck neoplasms, and control groups. for EBNA2. EMBO J. 1995b;14:88–96. recognition of Epstein Barr virus nuclear antigen 1 in
J Natl Cancer Inst. 1970;44:225–231. 35. Kilger E, Kieser A, Baumann M, Hammerschmidt healthy virus carriers. Blood. 2007;109:1138–1146.
15. de-Thé G, Geser A, Day NE. Epidemiological W. Epstein-Barr virus-mediated B-cell proliferation is 55. Paludan C, Schmid D, Landthaler M, et al. Endog-
evidence for causal relationship between Epstein- dependent upon latent membrane protein 1, which enous MHC class II processing of a viral nuclear
Barr virus and Burkitt’s lymphoma from Ugandan simulates an activated CD40 receptor. EMBO J. antigen after autophagy. Science. 2005;307:593–
prospective study. Nature. 1978;274:756–761. 1998;17:1700–1709. 596.
16. Zeng Y, Zhang LG, Wu YC, et al. Prospective studies 36. Bornkamm GW, Hammerschmidt W. Molecular 56. Babcock GJ, Decker LL, Volk M, Thorley-Lawson
on nasopharyngeal carcinoma in Epstein-Barr virus virology of Epstein-Barr virus. Philos Trans R Soc DA. EBV persistence in memory B cells in vivo.
IgA/VCA antibody-positive persons in Wuzhou City, Lond B Biol Sci. 2001;356:437–459. Immunity. 1998;9:395–404.
China. Int J Cancer. 1985;36:545–547. 37. Humme S, Reisbach G, Feederle R, et al. The 57. Qu L, Rowe DT. Epstein-Barr virus latent gene
17. Crawford DH, Thomas JA, Janossy G, et al. Epstein EBV nuclear antigen 1 (EBNA1) enhances B cell expression in uncultured peripheral blood lympho-
Barr virus nuclear antigen positive lymphoma immortalization several thousandfold. Proc Natl Acad cytes. J Virol. 1992;66:3715–3724.
after cyclosporin A treatment in patient with renal Sci USA. 2003;100:10989–10994. 58. Crawford DH. Biology and disease associations of
allograft. Lancet. 1980;1:1355–1356. 38. Vereide DT, Sugden B. Lymphomas differ in their Epstein-Barr virus. Philos Trans R Soc Lond B Biol
18. Greenspan JS, Greenspan D, Lennette ET, et al. dependence on Epstein-Barr virus. Blood. 2011;117: Sci. 2001;356:461–473.
Replication of Epstein-Barr virus within the epithelial 1977–1985. 59. Aguilar LK, Rooney CM, Heslop HE. Lympho-
cells of oral “hairy” leukoplakia, an AIDS-associated 39. Kempkes B, Pich D, Zeidler R, et al. Immortalization proliferative disorders involving Epstein-Barr virus
lesion. N Engl J Med. 1985;313:1564–1571. of human B lymphocytes by a plasmid containing after hemopoietic stem cell transplantation. Curr
19. Evans AS, Gutensohn NM. A population-based 71 kilobase pairs of Epstein-Barr virus DNA. J Virol. Opin Oncol. 1999;11:96–101.
case-control study of EBV and other viral antibodies 1995a;69:231–238. 60. AlDabbagh MA, Gitman MR, Kumar D, et al. The
among persons with Hodgkin’s disease and their 40. White RE, Ramer PC, Naresh KN, et al. EBNA3B- role of antiviral prophylaxis for the prevention of
siblings. Int J Cancer. 1984;34:149–157. deficient EBV promotes B cell lymphomagenesis in Epstein-Barr virus–associated posttransplant lympho-
20. Weiss LM, Strickler JG, Warnke RA, et al. Epstein- humanized mice and is found in human tumors. proliferative disease in solid organ transplant recipi-
Barr viral DNA in tissues of Hodgkin’s disease. Am J Clin Invest. 2012;122:1487–1502. ents: a systematic review. Am J Transplant. 2017;17(3):
J Pathol. 1987;129:86–91. 41. Wang A, Welch R, Zhao B, et al. Epstein-Barr virus 770–781.
21. Chadburn A, Chiu A, Lee JY, et al. Immunophe- nuclear antigen 3 (EBNA3) Proteins regulate EBNA2 61. Taub R, Kirsch I, Morton C, et al. Translocation
notypic analysis of AIDS-related diffuse large B-cell binding to distinct RBPJ genomic sites. J Virol. of the c-myc gene into the immunoglobulin heavy
lymphoma and clinical implications in patients from 2015;90(6):2906–2919. chain locus in human Burkitt lymphoma and murine
AIDS Malignancies Consortium clinical trials 010 42. Maruo S, Zhao B, Johannsen E, et al. Epstein- plasmacytoma cells. Proc Natl Acad Sci USA. 1982;
and 034. J Clin Oncol. 2009;27:5039–5048. Barr virus nuclear antigens 3C and 3A maintain 79:7837–7841.
22. Imai S, Koizumi S, Sugiura M, et al. Gastric lymphoblastoid cell growth by repressing p16INK4A 62. Zech L, Haglund U, Nilsson K, Klein G. Charac-
carcinoma: monoclonal epithelial malignant cells and p14ARF expression. Proc Natl Acad Sci USA. teristic chromosomal abnormalities in biopsies and
expressing Epstein-Barr virus latent infection protein. 2011;108:1919–1924. lymphoid-cell lines from patients with Burkitt and
Proc Natl Acad Sci USA. 1994;91:9131–9135. 43. Caldwell RG, Wilson JB, Anderson SJ, Longnecker non-Burkitt lymphomas. Int J Cancer. 1976;17:
23. Kieff E, Rickinson AB. Epstein Barr virus and its R. Epstein-Barr virus LMP2A drives B cell devel- 47–56.
replication. In: Knipe DM, Howley PM, Griffin opment and survival in the absence of normal 63. Ramiro AR, Nussenzweig MC, Nussenzweig A.
DE, et  al, eds. Fields Virology. Philadelphia: B cell receptor signals. Immunity. 1998;9:405– Switching on chromosomal translocations. Cancer
Lippincott Williams & Wilkins; 2001:2511–2573. 411. Res. 2006;66:7837–7839.
179.e2 Part I: Science and Clinical Oncology

64. Stanton LW, Watt R, Marcu KB. Translocation, of hepatocellular carcinoma in children. Taiwan human cervical carcinoma cell lines. Am J Pathol.
breakage and truncated transcripts of c-myc onco- Childhood Hepatoma Study Group. N Engl J Med. 1985;119:361–366.
gene in murine plasmacytomas. Nature. 1983;303: 1997;336:1855–1859. 109. Shope RE. A Transmissible tumor-like condition
401–406. 87. Seeger C, Mason WS. Hepatitis B virus biology. in rabbits. J Exp Med. 1932;56:793–802.
65. Morrow RH, Kisuule A, Pike MC, Smith PG. Microbiol Mol Biol Rev. 2000;64:51–68. 110. Jarrett WF, Murphy J, O’Neil BW, Laird HM.
Burkitt’s lymphoma in the Mengo Districts of 88. Unsal H, Yakicier C, Marcais C, et al. Genetic Virus-induced papillomas of the alimentary tract
Uganda: epidemiologic features and their relationship heterogeneity of hepatocellular carcinoma. Proc of cattle. Int J Cancer. 1978;22:323–328.
to malaria. J Natl Cancer Inst. 1976;56:479–483. Natl Acad Sci USA. 1994;91:822–826. 111. Campo MS, Moar MH, Sartirana ML, et al. The
66. Whittle HC, Brown J, Marsh K, et al. T-cell control 89. Henkler F, Waseem N, Golding MH, et al. Mutant presence of bovine papillomavirus type 4 DNA is not
of Epstein-Barr virus-infected B cells is lost during p53 but not hepatitis B virus X protein is present required for the progression to, or the maintenance
P. falciparum malaria. Nature. 1984;312:449–450. in hepatitis B virus-related human hepatocellular of, the malignant state in cancers of the alimentary
67. Moormann AM, Chelimo K, Sumba OP, et al. carcinoma. Cancer Res. 1995;55:6084–6091. canal in cattle. EMBO J. 1985;4:1819–1825.
Exposure to holoendemic malaria results in elevated 90. Wang XW, Gibson MK, Vermeulen W, et al. Abroga- 112. Jarrett WF, McNeil PE, Grimshaw WT, et al. High
Epstein-Barr virus loads in children. J Infect Dis. tion of p53-induced apoptosis by the hepatitis B incidence area of cattle cancer with a possible interac-
2005;191:1233–1238. virus X gene. Cancer Res. 1995;55:6012–6016. tion between an environmental carcinogen and a
68. Lam KM, Syed N, Whittle H, Crawford DH. 91. Haviv I, Shamay M, Doitsh G, Shaul Y. Hepatitis B papilloma virus. Nature. 1978;274:215–217.
Circulating Epstein-Barr virus–carrying B cells in virus pX targets TFIIB in transcription coactivation. 113. Pennie WD, Campo MS. Synergism between
acute malaria. Lancet. 1991;337:876–878. Mol Cell Biol. 1998;18:1562–1569. bovine papillomavirus type 4 and the flavonoid
69. Gaidano G, Ballerini P, Gong JZ, et al. p53 92. Lin Y, Nomura T, Cheong J, et al. Hepatitis B quercetin in cell transformation in vitro. Virology.
mutations in human lymphoid malignancies: virus X protein is a transcriptional modulator that 1992;190:861–865.
association with Burkitt lymphoma and chronic communicates with transcription factor IIB and 114. Dvoretzky I, Shober R, Chattopadhyay SK, Lowy
lymphocytic leukemia. Proc Natl Acad Sci USA. the RNA polymerase II subunit 5. J Biol Chem. DR. A quantitative in vitro focus assay for bovine
1991;88:5413–5417. 1997;272:7132–7139. papilloma virus. Virology. 1980;103:369–375.
70. Nanbo A, Sugden A, Sugden B. The coupling of syn- 93. Lee DK, Park SH, Yi Y, et al. The hepatitis B virus 115. Law MF, Lowy DR, Dvoretzky I, Howley PM. Mouse
thesis and partitioning of EBV’s plasmid replicon is encoded oncoprotein pX amplifies TGF-beta family cells transformed by bovine papillomavirus contain
revealed in live cells. EMBO J. 2007;26:4252–4262. signaling through direct interaction with Smad4: only extrachromosomal viral DNA sequences.
71. Vereide DT, Seto E, Chiu YF, et al. Epstein-Barr virus potential mechanism of hepatitis B virus-induced Proc Natl Acad Sci USA. 1981;78:2727–2731.
maintains lymphomas via its miRNAs. Oncogene. liver fibrosis. Genes Dev. 2001;15:455–466. 116. Lowy DR, Dvoretzky I, Shober R, et al. In vitro
2014;33(10):1258–1264. 94. Arzumanyan A, Friedman T, Ng IO, et al. tumorigenic transformation by a defined sub-
72. Ferry JA. Burkitt’s lymphoma: clinicopathologic fea- Does the hepatitis B antigen HBx promote the genomic fragment of bovine papilloma virus DNA.
tures and differential diagnosis. Oncologist. 2006;11: appearance of liver cancer stem cells? Cancer Res. Nature. 1980;287:72–74.
375–383. 2011;71:3701–3708. 117. Androphy EJ, Schiller JT, Lowy DR. Identification of
73. Hesseling P, Broadhead R, Mansvelt E, et al. The 95. Buendia MA. Hepatitis B viruses and hepatocellular the protein encoded by the E6 transforming gene of
2000 Burkitt lymphoma trial in Malawi. Pediatr carcinoma. Adv Cancer Res. 1992;59:167–226. bovine papillomavirus. Science. 1985;230:442–445.
Blood Cancer. 2005;44:245–250. 96. Fourel G, Couturier J, Wei Y, et al. Evidence for 118. Nakabayashi Y, Chattopadhyay SK, Lowy DR. The
74. Chan AS, To KF, Lo KW, et al. High frequency of long-range oncogene activation by hepadnavirus transforming function of bovine papillomavirus
chromosome 3p deletion in histologically normal insertion. EMBO J. 1994;13:2526–2534. DNA. Proc Natl Acad Sci USA. 1983;80:5832–5836.
nasopharyngeal epithelia from southern Chinese. 97. Pineau P, Marchio A, Mattei MG, et al. Extensive 119. Sarver N, Rabson MS, Yang YC, et al. Localiza-
Cancer Res. 2000;60:5365–5370. analysis of duplicated-inverted hepatitis B virus tion and analysis of bovine papillomavirus type
75. Sengupta S, den Boon JA, Chen IH, et al. Genome- integrations in human hepatocellular carcinoma. 1 transforming functions. J Virol. 1984;52:377–
wide expression profiling reveals EBV-associated inhi- J Gen Virol. 1998;79(Pt 3):591–600. 388.
bition of MHC class I expression in nasopharyngeal 98. Chisari FV, Ferrari C. Hepatitis B virus immuno- 120. Schiller JT, Vass WC, Vousden KH, Lowy DR. E5
carcinoma. Cancer Res. 2006;66:7999–8006. pathogenesis. Annu Rev Immunol. 1995;13:29–60. open reading frame of bovine papillomavirus type 1
76. Cosmopoulos K, Pegtel M, Hawkins J, et al. 99. Guidotti LG, Ando K, Hobbs MV, et al. Cytotoxic T encodes a transforming gene. J Virol. 1986;57:1–6.
Comprehensive profiling of Epstein-Barr virus lymphocytes inhibit hepatitis B virus gene expression 121. Schlegel R, Wade GM, Rabson MS, Yang YC. The
microRNAs in nasopharyngeal carcinoma. J Virol. by a noncytolytic mechanism in transgenic mice. E5 transforming gene of bovine papillomavirus
2009;83:2357–2367. Proc Natl Acad Sci USA. 1994;91:3764–3768. encodes a small, hydrophobic polypeptide. Science.
77. Pratt ZL, Kuzembayeva M, Sengupta S, Sugden B. 100. Guidotti LG, Rochford R, Chung J, et al. Viral 1986;233:464–467.
The microRNAs of Epstein-Barr virus are expressed clearance without destruction of infected cells 122. Yang YC, Okayama H, Howley PM. Bovine papil-
at dramatically differing levels among cell lines. during acute HBV infection. Science. 1999;284:825– lomavirus contains multiple transforming genes. Proc
Virology. 2009;386:387–397. 829. Natl Acad Sci USA. 1985;82:1030–1034.
78. Blumberg BS. Australia antigen and the biology of 101. Kimura K, Kakimi K, Wieland S, et al. Interleukin-18 123. Thierry F, Yaniv M. The BPV1-E2 trans-acting
hepatitis B. Science. 1977;197:17–25. inhibits hepatitis B virus replication in the livers of protein can be either an activator or a repressor of
79. Senior JR, Sutnick AI, Goeser E, et al. Reduction of transgenic mice. J Virol. 2002;76:10702–10707. the HPV18 regulatory region. EMBO J. 1987;6:
post-transfusion hepatitis by exclusion of Australia 102. Nakamoto Y, Guidotti LG, Kuhlen CV, et al. 3391–3397.
antigen from donor blood in an urban public Immune pathogenesis of hepatocellular carcinoma. 124. Jeon S, Lambert PF. Integration of human papil-
hospital. Am J Med Sci. 1974;267:171–177. J Exp Med. 1998;188:341–350. lomavirus type 16 DNA into the human genome
80. Monto A, Wright TL. The epidemiology and preven- 103. Nelson NP, Easterbrook PJ, McMahon BJ. Epide- leads to increased stability of E6 and E7 mRNAs:
tion of hepatocellular carcinoma. Semin Oncol. 2001; miology of hepatitis B virus infection and impact of implications for cervical carcinogenesis. Proc Natl
28:441–449. vaccination on disease. Clin Liver Dis. 2016;20(4): Acad Sci USA. 1995;92:1654–1658.
81. Wild CP, Hall AJ. Primary prevention of hepatocel- 607–628. 125. Jeon S, Allen-Hoffmann BL, Lambert PF. Integration
lular carcinoma in developing countries. Mutat Res. 104. Testoni B, Durantel D, Zoulim F. Novel targets for of human papillomavirus type 16 into the human
2000;462:381–393. hepatitis B virus therapy. Liver Int. 2017;37(suppl genome correlates with a selective growth advantage
82. Beasley RP, Hwang LY, Lin CC, Chien CS. 1):33–39. of cells. J Virol. 1995;69:2989–2997.
Hepatocellular carcinoma and hepatitis B virus. A 105. Boshart M, Gissmann L, Ikenberg H, et al. A new 126. Dyson N, Howley PM, Munger K, Harlow E. The
prospective study of 22 707 men in Taiwan. Lancet. type of papillomavirus DNA, its presence in genital human papilloma virus-16 E7 oncoprotein is able
1981;2:1129–1133. cancer biopsies and in cell lines derived from cervical to bind to the retinoblastoma gene product. Science.
83. Beasley RP. Hepatitis B virus. The major etiology cancer. EMBO J. 1984;3:1151–1157. 1989;243:934–937.
of hepatocellular carcinoma. Cancer. 1988;61: 106. Durst M, Gissmann L, Ikenberg H, zur Hausen H. 127. Werness BA, Levine AJ, Howley PM. Association of
1942–1956. A papillomavirus DNA from a cervical carcinoma human papillomavirus types 16 and 18 E6 proteins
84. Chen HL, Chang MH, Ni YH, et al. Seroepide- and its prevalence in cancer biopsy samples from with p53. Science. 1990;248:76–79.
miology of hepatitis B virus infection in children: different geographic regions. Proc Natl Acad Sci USA. 128. Scheffner M, Munger K, Byrne JC, Howley PM.
ten years of mass vaccination in Taiwan. JAMA. 1983;80:3812–3815. The state of the p53 and retinoblastoma genes in
1996;276:906–908. 107. Schwarz E, Freese UK, Gissmann L, et al. Struc- human cervical carcinoma cell lines. Proc Natl Acad
85. Montesano R. Hepatitis B immunization and ture and transcription of human papillomavirus Sci USA. 1991;88:5523–5527.
hepatocellular carcinoma: the Gambia Hepatitis sequences in cervical carcinoma cells. Nature. 129. Francis DA, Schmid SI, Howley PM. Repression
Intervention Study. J Med Virol. 2002;67:444–446. 1985;314:111–114. of the integrated papillomavirus E6/E7 promoter is
86. Chang MH, Chen CJ, Lai MS, et al. Universal 108. Yee C, Krishnan HI, Baker CC, et al. Presence and required for growth suppression of cervical cancer
hepatitis B vaccination in Taiwan and the incidence expression of human papillomavirus sequences in cells. J Virol. 2000;74:2679–2686.
Viruses and Human Cancer  •  CHAPTER 12 179.e3

130. Goodwin EC, DiMaio D. Repression of human papil- and E7 oncogenes in the epidermis develop verrucous 167. Simonson SJ, Difilippantonio MJ, Lambert PF.
lomavirus oncogenes in HeLa cervical carcinoma cells lesions and spontaneous, rasHa-activated papillomas. Two distinct activities contribute to human papil-
causes the orderly reactivation of dormant tumor sup- Cell Growth Differ. 1994;5:667–675. lomavirus 16 E6’s oncogenic potential. Cancer Res.
pressor pathways. Proc Natl Acad Sci USA. 2000;97: 149. Herber R, Liem A, Pitot H, Lambert PF. Squamous 2005;65:8266–8273.
12513–12518. epithelial hyperplasia and carcinoma in mice 168. Patel D, Huang SM, Baglia LA, McCance DJ. The
131. Goodwin EC, Yang E, Lee CJ, et al. Rapid induction transgenic for the human papillomavirus type 16 E6 protein of human papillomavirus type 16 binds
of senescence in human cervical carcinoma cells. E7 oncogene. J Virol. 1996;70:1873–1881. to and inhibits co-activation by CBP and p300.
Proc Natl Acad Sci USA. 2000;97:10978–10983. 150. Lambert PF, Pan H, Pitot HC, et al. Epidermal EMBO J. 1999;18:5061–5072.
132. Hwang ES, Riese DJ 2nd, Settleman J, et al. cancer associated with expression of human papil- 169. Zimmermann H, Degenkolbe R, Bernard HU,
Inhibition of cervical carcinoma cell line prolifera- lomavirus type 16 E6 and E7 oncogenes in the O’Connor MJ. The human papillomavirus type
tion by the introduction of a bovine papillomavirus skin of transgenic mice. Proc Natl Acad Sci USA. 16 E6 oncoprotein can down-regulate p53 activity
regulatory gene. J Virol. 1993;67:3720–3729. 1993;90:5583–5587. by targeting the transcriptional coactivator CBP/
133. Nishimura A, Ono T, Ishimoto A, et al. Mechanisms 151. Pan H, Griep AE. Altered cell cycle regulation in p300. J Virol. 1999;73:6209–6219.
of human papillomavirus E2-mediated repression of the lens of HPV-16 E6 or E7 transgenic mice: 170. Tong X, Howley PM. The bovine papillomavirus
viral oncogene expression and cervical cancer cell implications for tumor suppressor gene function E6 oncoprotein interacts with paxillin and disrupts
growth inhibition. J Virol. 2000;74:3752–3760. in development. Genes Dev. 1994;8:1285–1299. the actin cytoskeleton. Proc Natl Acad Sci USA.
134. Wells SI, Francis DA, Karpova AY, et al. Papil- 152. Song S, Liem A, Miller JA, Lambert PF. Human 1997;94:4412–4417.
lomavirus E2 induces senescence in HPV-positive papillomavirus types 16 E6 and E7 contribute dif- 171. Vande Pol SB, Brown MC, Turner CE. Association
cells via pRB- and p21(CIP)-dependent pathways. ferently to carcinogenesis. Virology. 2000;267:141– of bovine papillomavirus type 1 E6 oncoprotein
EMBO J. 2000;19:5762–5771. 150. with the focal adhesion protein paxillin through a
135. Jabbar SF, Abrams L, Glick A, Lambert PF. Persis- 153. Song S, Pitot HC, Lambert PF. The human papillo- conserved protein interaction motif. Oncogene. 1998;
tence of high-grade cervical dysplasia and cervical mavirus type 16 E6 gene alone is sufficient to induce 16:43–52.
cancer requires the continuous expression of the carcinomas in transgenic animals. J Virol. 1999;73: 172. Gao Q, Srinivasan S, Boyer SN, et al. The E6
human papillomavirus type 16 E7 oncogene. Cancer 5887–5893. oncoproteins of high-risk papillomaviruses bind to
Res. 2009;69:4407–4414. 154. Jabbar S, Strati K, Shin MK, et al. Human papil- a novel putative GAP protein, E6TP1, and target it
136. Jabbar SF, Park S, Schweizer J, et al. Cervical cancers lomavirus type 16 E6 and E7 oncoproteins act for degradation. Mol Cell Biol. 1999;19:733–744.
require the continuous expression of the human synergistically to cause head and neck cancer in 173. Ronco LV, Karpova AY, Vidal M, Howley PM.
papillomavirus type 16 E7 oncoprotein even in the mice. Virology. 2010;407:60–67. Human papillomavirus 16 E6 oncoprotein binds
presence of the viral E6 oncoprotein. Cancer Res. 155. Riley RR, Duensing S, Brake T, et al. Dissection to interferon regulatory factor-3 and inhibits its tran-
2012;72(16):4008–4016. of human papillomavirus E6 and E7 function in scriptional activity. Genes Dev. 1998;12:2061–2072.
137. Band V, Zajchowski D, Kulesa V, Sager R. Human transgenic mouse models of cervical carcinogenesis. 174. Chen JJ, Reid CE, Band V, Androphy EJ. Interaction
papilloma virus DNAs immortalize normal human Cancer Res. 2003;63:4862–4871. of papillomavirus E6 oncoproteins with a putative
mammary epithelial cells and reduce their growth 156. Thomas MK, Pitot HC, Liem A, Lambert PF. calcium-binding protein. Science. 1995;269:529–531.
factor requirements. Proc Natl Acad Sci USA. 1990;87: Dominant role of HPV16 E7 in anal carcinogenesis. 175. Thomas M, Banks L. Human papillomavirus (HPV)
463–467. Virology. 2011;421:114–118. E6 interactions with Bak are conserved amongst E6
138. Durst M, Dzarlieva-Petrusevska RT, Boukamp P, 157. Huibregtse JM, Scheffner M, Howley PM. A cellular proteins from high and low risk HPV types. J Gen
et al. Molecular and cytogenetic analysis of immortal- protein mediates association of p53 with the E6 Virol. 1999;80(Pt 6):1513–1517.
ized human primary keratinocytes obtained after oncoprotein of human papillomavirus types 16 or 176. Gao Q, Kumar A, Srinivasan S, et al. PKN binds
transfection with human papillomavirus type 16 18. EMBO J. 1991;10:4129–4135. and phosphorylates human papillomavirus E6
DNA. Oncogene. 1987;1:251–256. 158. Scheffner M, Huibregtse JM, Vierstra RD, Howley oncoprotein. J Biol Chem. 2000;275:14824–14830.
139. Hawley-Nelson P, Vousden KH, Hubbert NL, PM. The HPV-16 E6 and E6-AP complex functions 177. Gross-Mesilaty S, Reinstein E, Bercovich B,
et al. HPV16 E6 and E7 proteins cooperate to as a ubiquitin-protein ligase in the ubiquitination et al. Basal and human papillomavirus E6
immortalize human foreskin keratinocytes. EMBO of p53. Cell. 1993;75:495–505. oncoprotein-induced degradation of Myc proteins
J. 1989;8:3905–3910. 159. Lechner MS, Mack DH, Finicle AB, et al. Human by the ubiquitin pathway. Proc Natl Acad Sci USA.
140. Schlegel R, Phelps WC, Zhang YL, Barbosa M. papillomavirus E6 proteins bind p53 in vivo and 1998;95:8058–8063.
Quantitative keratinocyte assay detects two biological abrogate p53-mediated repression of transcription. 178. Kiyono T, Hiraiwa A, Fujita M, et al. Binding of
activities of human papillomavirus DNA and identi- EMBO J. 1992;11:3045–3052. high-risk human papillomavirus E6 oncoproteins
fies viral types associated with cervical carcinoma. 160. Mietz JA, Unger T, Huibregtse JM, Howley to the human homologue of the Drosophila discs
EMBO J. 1988;7:3181–3187. PM. The transcriptional transactivation function large tumor suppressor protein. Proc Natl Acad Sci
141. Kaur P, McDougall JK. HPV-18 immortalization of of wild-type p53 is inhibited by SV40 large USA. 1997;94:11612–11616.
human keratinocytes. Virology. 1989;173:302–310. T-antigen and by HPV-16 E6 oncoprotein. EMBO 179. Lee SS, Weiss RS, Javier RT. Binding of human virus
142. Woodworth CD, Bowden PE, Doniger J, et al. Char- J. 1992;11:5013–5020. oncoproteins to hDlg/SAP97, a mammalian homolog
acterization of normal human exocervical epithelial 161. Boyer SN, Wazer DE, Band V. E7 protein of human of the Drosphilia discs large tumor suppressor protein.
cells immortalized in vitro by papillomavirus types papilloma virus-16 induces degradation of retino- Proc Natl Acad Sci U S A. 1997;94:6670–6675.
16 and 18 DNA. Cancer Res. 1988;48:4620–4628. blastoma protein through the ubiquitin-proteasome 180. Nakagawa S, Huibregtse JM. Human scribble
143. Crook T, Storey A, Almond N, et al. Human pathway. Cancer Res. 1996;56:4620–4624. (Vartul) is targeted for ubiquitin-mediated degrada-
papillomavirus type 16 cooperates with activated 162. Jones DL, Thompson DA, Munger K. Destabilization tion by the high-risk papillomavirus E6 proteins and
ras and fos oncogenes in the hormone-dependent of the RB tumor suppressor protein and stabilization the E6AP ubiquitin-protein ligase. Mol Cell Biol.
transformation of primary mouse cells. Proc Natl of p53 contribute to HPV type 16 E7-induced 2000;20:8244–8253.
Acad Sci USA. 1988;85:8820–8824. apoptosis. Virology. 1997;239:97–107. 181. Glaunsinger BA, Lee SS, Thomas M, et al.
144. Matlashewski G, Schneider J, Banks L, et al. 163. Chellappan S, Kraus VB, Kroger B, et al. Adenovirus Interactions of the PDZ-protein MAGI-1 with
Human papillomavirus type 16 DNA cooperates E1A, simian virus 40 tumor antigen, and human adenovirus E4-ORF1 and high-risk papillomavirus
with activated ras in transforming primary cells. papillomavirus E7 protein share the capacity to E6 oncoproteins. Oncogene. 2000;19:5270–5280.
EMBO J. 1987;6:1741–1746. disrupt the interaction between transcription factor 182. Lee SS, Glaunsinger B, Mantovani F, et al. Multi-
145. Phelps WC, Yee CL, Munger K, Howley PM. The E2F and the retinoblastoma gene product. Proc Natl PDZ domain protein MUPP1 is a cellular target
human papillomavirus type 16 E7 gene encodes Acad Sci USA. 1992;89:4549–4553. for both adenovirus E4-ORF1 and high-risk papil-
transactivation and transformation functions similar 164. Balsitis S, Dick F, Dyson N, Lambert PF. Critical lomavirus type 18 E6 oncoproteins. J Virol. 2000;74:
to those of adenovirus E1A. Cell. 1988;53:539–547. roles for non-pRb targets of human papillomavirus 9680–9693.
146. Arbeit JM, Howley PM, Hanahan D. Chronic type 16 E7 in cervical carcinogenesis. Cancer Res. 183. Gewin L, Myers H, Kiyono T, Galloway DA.
estrogen-induced cervical and vaginal squamous 2006;66:9393–9400. Identification of a novel telomerase repressor that
carcinogenesis in human papillomavirus type 16 165. Shai A, Brake T, Somoza C, Lambert PF. The human interacts with the human papillomavirus type-16
transgenic mice. Proc Natl Acad Sci USA. 1996;93: papillomavirus E6 oncogene dysregulates the cell cycle E6/E6-AP complex. Genes Dev. 2004;18:2269–2282.
2930–2935. and contributes to cervical carcinogenesis through 184. Xu M, Luo W, Elzi DJ, et al. NFX1 interacts with
147. Elson DA, Riley RR, Lacey A, et al. Sensitivity two independent activities. Cancer Res. 2007;67: mSin3A/histone deacetylase to repress hTERT
of the cervical transformation zone to estrogen- 1626–1635. transcription in keratinocytes. Mol Cell Biol.
induced squamous carcinogenesis. Cancer Res. 166. Shin MK, Balsitis S, Brake T, Lambert PF. Human 2008;28:4819–4828.
2000;60:1267–1275. papillomavirus E7 oncoprotein overrides the tumor 185. Katzenellenbogen RA, Vliet-Gregg P, Xu M, Galloway
148. Greenhalgh DA, Wang XJ, Rothnagel JA, et al. suppressor activity of p21Cip1 in cervical carcino- DA. NFX1-123 increases hTERT expression and
Transgenic mice expressing targeted HPV-18 E6 genesis. Cancer Res. 2009;69:5656–5663. telomerase activity posttranscriptionally in human
179.e4 Part I: Science and Clinical Oncology

papillomavirus type 16 E6 keratinocytes. J Virol. contributes to E7 transforming activity. J Gen Virol. target the NOTCH and TGF-beta tumor suppressors
2009;83:6446–6456. 1997;78(Pt 10):2607–2613. to inhibit differentiation and sustain keratinocyte
186. Katzenellenbogen RA, Vliet-Gregg P, Xu M, Gal- 205. Phillips AC, Vousden KH. Analysis of the interaction proliferation. PLoS Pathog. 2017;13(1):e1006171.
loway DA. Cytoplasmic poly(A) binding proteins between human papillomavirus type 16 E7 and the doi:10.1371/journal.ppat.1006171. [Epub 2017/01/
regulate telomerase activity and cell growth in human TATA-binding protein, TBP. J Gen Virol. 1997;78(Pt 21]; PubMed PMID 28107544. PMCID 5287491.
papillomavirus type 16 E6-expressing keratinocytes. 4):905–909. 224. Madeleine MM, Wiktor SZ, Goedert JJ, et al.
J Virol. 2010;84:12934–12944. 206. Mazzarelli JM, Atkins GB, Geisberg JV, Ricciardi RP. HTLV-I and HTLV-II world-wide distribution:
187. Shai A, Pitot HC, Lambert PF. E6-associated The viral oncoproteins Ad5 E1A, HPV16 E7 and reanalysis of 4,832 immunoblot results. Int J Cancer.
protein is required for human papillomavirus type SV40 TAg bind a common region of the TBP-asso- 1993;54:255–260.
16 E6 to cause cervical cancer in mice. Cancer Res. ciated factor-110. Oncogene. 1995;11:1859–1864. 225. Tajima K. Malignant lymphomas in Japan: epidemio-
2010;70:5064–5073. 207. Schilling B, De-Medina T, Syken J, et al. A novel logical analysis of adult T-cell leukemia/lymphoma
188. Dyson N, Guida P, Munger K, Harlow E. Homolo- human DnaJ protein, hTid-1, a homolog of the (ATL). Cancer Metastasis Rev. 1988;7:223–241.
gous sequences in adenovirus E1A and human Drosophila tumor suppressor protein Tid56, can 226. Jeffery KJ, Usuku K, Hall SE, et al. HLA
papillomavirus E7 proteins mediate interaction with interact with the human papillomavirus type 16 alleles determine human T-lymphotropic virus-I
the same set of cellular proteins. J Virol. 1992;66: E7 oncoprotein. Virology. 1998;247:74–85. (HTLV-I) proviral load and the risk of HTLV-I-
6893–6902. 208. Davies R, Hicks R, Crook T, et al. Human papil- associated myelopathy. Proc Natl Acad Sci USA.
189. Classon M, Salama S, Gorka C, et al. Combi- lomavirus type 16 E7 associates with a histone H1 1999;96:3848–3853.
natorial roles for pRB, p107, and p130 in E2F- kinase and with p107 through sequences necessary 227. Hinuma Y, Nagata K, Hanaoka M, et al. Adult
mediated cell cycle control. Proc Natl Acad Sci USA. for transformation. J Virol. 1993;67:2521–2528. T-cell leukemia: antigen in an ATL cell line and
2000;97:10820–10825. 209. McLaughlin-Drubin ME, Crum CP, Munger K. detection of antibodies to the antigen in human
190. Dyson N, Dembski M, Fattaey A, et al. Analysis Human papillomavirus E7 oncoprotein induces sera. Proc Natl Acad Sci USA. 1981;78:6476–6480.
of p107-associated proteins: p107 associates with KDM6A and KDM6B histone demethylase expres- 228. Yoshida M, Seiki M, Yamaguchi K, Takatsuki K.
a form of E2F that differs from pRB-associated sion and causes epigenetic reprogramming. Proc Natl Monoclonal integration of human T-cell leukemia
E2F-1. J Virol. 1993;67:7641–7647. Acad Sci USA. 2011;108:2130–2135. provirus in all primary tumors of adult T-cell
191. Shin MK, Pitot HC, Lambert PF. Pocket proteins 210. Zhang B, Laribee RN, Klemsz MJ, Roman A. leukemia suggests causative role of human T-cell
suppress head and neck cancer. Cancer Res. 2012;72: Human papillomavirus type 16 E7 protein increases leukemia virus in the disease. Proc Natl Acad Sci
1280–1289. acetylation of histone H3 in human foreskin USA. 1984;81:2534–2537.
192. Shin MK, Sage J, Lambert PF. Inactivating all three keratinocytes. Virology. 2004;329:189–198. 229. Hino S, Katamine S, Miyata H, et al. Primary
rb family pocket proteins is insufficient to initiate 211. Andl T, Kahn T, Pfuhl A, et al. Etiological involve- prevention of HTLV-I in Japan. Leukemia.
cervical cancer. Cancer Res. 2012;72(20):5418–5427. ment of oncogenic human papillomavirus in tonsillar 1997;11(suppl 3):57–59.
193. Arroyo M, Bagchi S, Raychaudhuri P. Association squamous cell carcinomas lacking retinoblastoma 230. Albrecht B, Lairmore MD. Critical role of human
of the human papillomavirus type 16 E7 protein cell cycle control. Cancer Res. 1998;58:5–13. T-lymphotropic virus type 1 accessory proteins in
with the S-phase-specific E2F-cyclin A complex. 212. Biliris KA, Koumantakis E, Dokianakis DN, et al. viral replication and pathogenesis. Microbiol Mol
Mol Cell Biol. 1993;13:6537–6546. Human papillomavirus infection of non-melanoma Biol Rev. 2002;66:396–406.
194. McIntyre MC, Ruesch MN, Laimins LA. Human skin cancers in immunocompetent hosts. Cancer 231. Yoshida M. Multiple viral strategies of HTLV-I
papillomavirus E7 oncoproteins bind a single form of Lett. 2000;161:83–88. for dysregulation of cell growth control. Annu Rev
cyclin E in a complex with cdk2 and p107. Virology. 213. Gillison ML, Koch WM, Capone RB, et al. Evidence Immunol. 2001;19:475–496.
1996;215:73–82. for a causal association between human papilloma- 232. Tanaka A, Takahashi C, Yamaoka S, et al. Oncogenic
195. Tommasino M, Adamczewski JP, Carlotti F, et al. virus and a subset of head and neck cancers [see transformation by the tax gene of human T-cell
HPV16 E7 protein associates with the protein kinase comments]. J Natl Cancer Inst. 2000;92:709–720. leukemia virus type I in vitro. Proc Natl Acad Sci
p33CDK2 and cyclin A. Oncogene. 1993;8:195–202. 214. Kiviat NB. Papillomaviruses in non-melanoma skin USA. 1990;87:1071–1075.
196. Funk JO, Waga S, Harry JB, et al. Inhibition of CDK cancer: epidemiological aspects. Semin Cancer Biol. 233. Pozzatti R, Vogel J, Jay G. The human T-lympho-
activity and PCNA-dependent DNA replication by 1999;9:397–403. tropic virus type I tax gene can cooperate with the
p21 is blocked by interaction with the HPV-16 E7 215. Flores ER, Allen-Hoffmann BL, Lee D, Lambert PF. ras oncogene to induce neoplastic transformation
oncoprotein. Genes Dev. 1997;11:2090–2100. The human papillomavirus type 16 E7 oncogene of cells. Mol Cell Biol. 1990;10:413–417.
197. Jones DL, Alani RM, Munger K. The human is required for the productive stage of the viral life 234. Grassmann R, Berchtold S, Radant I, et al. Role of
papillomavirus E7 oncoprotein can uncouple cycle. J Virol. 2000;74:6622–6631. human T-cell leukemia virus type 1 X region proteins
cellular differentiation and proliferation in human 216. Park RB, Androphy EJ. Genetic analysis of high-risk in immortalization of primary human lymphocytes
keratinocytes by abrogating p21Cip1-mediated e6 in episomal maintenance of human papillomavirus in culture. J Virol. 1992;66:4570–4575.
inhibition of cdk2. Genes Dev. 1997;11:2101–2111. genomes in primary human keratinocytes. J Virol. 235. Ross TM, Pettiford SM, Green PL. The tax gene of
198. Yan X, Shah W, Jing L, et al. High-risk human 2002;76. human T-cell leukemia virus type 2 is essential for
papillomavirus type 18 E7 caused p27 elevation 217. Ustav M, Stenlund A. Transient replication of transformation of human T lymphocytes. J Virol.
and cytoplasmic localization. Cancer Biol Ther. BPV-1 requires two viral polypeptides encoded 1996;70:5194–5202.
2010;9:728–735. by the E1 and E2 open reading frames. EMBO J. 236. Nerenberg M, Hinrichs SH, Reynolds RK, et al.
199. Berezutskaya E, Bagchi S. The human papillomavirus 1991;10:449–457. The tat gene of human T-lymphotropic virus type
E7 oncoprotein functionally interacts with the S4 218. Mohr IJ, Clark R, Sun S, et al. Targeting the E1 1 induces mesenchymal tumors in transgenic mice.
subunit of the 26 S proteasome. J Biol Chem. replication protein to the papillomavirus origin Science. 1987;237:1324–1329.
1997;272:30135–30140. of replication by complex formation with the E2 237. Grossman WJ, Kimata JT, Wong FH, et al. Develop-
200. Brehm A, Nielsen SJ, Miska EA, et al. The E7 transactivator. Science. 1990;250:1694–1699. ment of leukemia in mice transgenic for the tax
oncoprotein associates with Mi2 and histone 219. Sedman J, Stenlund A. Co-operative interaction gene of human T-cell leukemia virus type I. Proc
deacetylase activity to promote cell growth. EMBO between the initiator E1 and the transcriptional Natl Acad Sci USA. 1995;92:1057–1061.
J. 1999;18:2449–2458. activator E2 is required for replicator specific DNA 238. Ego T, Ariumi Y, Shimotohno K. The interaction of
201. Luscher-Firzlaff JM, Westendorf JM, Zwicker J, et al. replication of bovine papillomavirus in vivo and in HTLV-I Tax with HDAC1 negatively regulates the
Interaction of the fork head domain transcription vitro. EMBO J. 1995;14:6218–6228. viral gene expression. Oncogene. 2002;21:7241–7246.
factor MPP2 with the human papilloma virus 16 220. Yang L, Mohr I, Fouts E, et  al. The E1 239. Jeang KT, Widen SG, Semmes OJT, Wilson SH.
E7 protein: enhancement of transformation and protein of bovine papilloma virus 1 is an ATP- HTLV-I trans-activator protein, tax, is a trans-
transactivation. Oncogene. 1999;18:5620–5630. dependent DNA helicase. Proc Natl Acad Sci USA. repressor of the human beta-polymerase gene.
202. Antinore MJ, Birrer MJ, Patel D, et al. The human 1993;90:5086–5090. Science. 1990;247:1082–1084.
papillomavirus type 16 E7 gene product interacts 221. Akgul B, Cook JC, Storey A. HPV-associated skin 240. Neuveut C, Low KG, Maldarelli F, et al. Human
with and trans-activates the AP1 family of transcrip- disease. J Pathol. 2006;208(2):165–175. doi:10.1002/ T-cell leukemia virus type 1 Tax and cell cycle
tion factors. EMBO J. 1996;15:1950–1960. path.1893. [Epub 2005/12/20]; PubMed PMID progression: role of cyclin D-cdk and p110Rb.
203. Mannhardt B, Weinzimer SA, Wagner M, et al. 16362995. Mol Cell Biol. 1998;18:3620–3632.
Human papillomavirus type 16 E7 oncoprotein 222. Uberoi A, Yoshida S, Frazer IH, Pitot H, Lambert 241. Pise-Masison CA, Mahieux R, Jiang H, et al.
binds and inactivates growth-inhibitory insulin-like PF. Role of ultraviolet radiation in papillomavirus- Inactivation of p53 by human T-cell lymphotropic
growth factor binding protein 3. Mol Cell Biol. induced disease. PLoS Pathog. 2016;12(5):e1005664. virus type 1 Tax requires activation of the NF-kappaB
2000;20:6483–6495. doi:10.1371/journal.ppat.1005664. [Epub 2016/06/ pathway and is dependent on p53 phosphorylation.
204. Massimi P, Pim D, Banks L. Human papillomavirus 01]; PMCID: 4887022. Mol Cell Biol. 2000;20:3377–3386.
type 16 E7 binds to the conserved carboxy-terminal 223. Meyers JM, Uberoi Grace M, Lambert PF, Munger 242. Suzuki T, Kitao S, Matsushime H, Yoshida M.
region of the TATA box binding protein and this K. Cutaneous HPV8 and MmuPV1 E6 proteins HTLV-I Tax protein interacts with cyclin-dependent
Viruses and Human Cancer  •  CHAPTER 12 179.e5

kinase inhibitor p16INK4A and counteracts its factor that down-regulates viral transcription. J Virol. 281. Wakita T, Pietschmann T, Kato T, et al. Production
inhibitory activity towards CDK4. EMBO J. 1996;15: 2002;76:12813–12822. of infectious hepatitis C virus in tissue culture from
1607–1614. 261. Lemasson I, Lewis MR, Polakowski N, et al. Human a cloned viral genome. Nat Med. 2005;11:791–796.
243. Felber BK, Paskalis H, Kleinman-Ewing C, et al. The T-cell leukemia virus type 1 (HTLV-I) bZIP protein 282. Zhong J, Gastaminza P, Cheng G, et al. Robust
pX protein of HTLV-I is a transcriptional activator of interacts with the cellular transcription factor CREB hepatitis C virus infection in vitro. Proc Natl Acad
its long terminal repeats. Science. 1985;229:675–679. to inhibit HTLV-I transcription. J Virol. 2007;81: Sci USA. 2005;102:9294–9299.
244. Cross SL, Feinberg MB, Wolf JB, et al. Regulation 1543–1553. 283. Lindenbach BD, Meuleman P, Ploss A, et al. Cell
of the human interleukin-2 receptor alpha chain 262. Clerc I, Polakowski N, Andre-Arpin C, et al. An culture-grown hepatitis C virus is infectious in vivo
promoter: activation of a nonfunctional promoter interaction between the human T cell leukemia and can be recultured in vitro. Proc Natl Acad Sci
by the transactivator gene of HTLV-I. Cell. 1987;49: virus type 1 basic leucine zipper factor (HBZ) and USA. 2006;103:3805–3809.
47–56. the KIX domain of p300/CBP contributes to the 284. Bartosch B, Cosset FL. Cell entry of hepatitis C
245. Fujii M, Sassone-Corsi P, Verma IM. c-fos promoter down-regulation of tax-dependent viral transcrip- virus. Virology. 2006;348:1–12.
trans-activation by the tax1 protein of human T-cell tion by HBZ. J Biol Chem. 2008;283:23903– 285. Bartosch B, Verney G, Dreux M, et al. An interplay
leukemia virus type I. Proc Natl Acad Sci USA. 1988;85: 23913. between hypervariable region 1 of the hepatitis C
8526–8530. 263. Satou Y, Yasunaga J, Zhao T, et al. HTLV-I bZIP virus E2 glycoprotein, the scavenger receptor BI, and
246. Siekevitz M, Feinberg MB, Holbrook N, et al. factor induces T-cell lymphoma and systemic inflam- high-density lipoprotein promotes both enhancement
Activation of interleukin 2 and interleukin 2 receptor mation in vivo. PLoS Pathog. 2011;7:e1001274. of infection and protection against neutralizing
(Tac) promoter expression by the trans-activator (tat) 264. Kawatsuki A, Yasunaga JI, Mitobe Y, Green PL, antibodies. J Virol. 2005;79:8217–8229.
gene product of human T-cell leukemia virus, type Matsuoka M. HTLV-1 bZIP factor protein targets 286. Wu X, Robotham JM, Lee E, et al. Productive
I. Proc Natl Acad Sci USA. 1987;84:5389–5393. the Rb/E2F-1 pathway to promote prolifera- hepatitis C virus infection of stem cell-derived
247. Tsukahara T, Kannagi M, Ohashi T, et al. Induction tion and apoptosis of primary CD4 (+) T cells. hepatocytes reveals a critical transition to viral
of Bcl-x(L) expression by human T-cell leukemia virus Oncogene. 2016;35(34):4509–4517. doi:10.1038/ permissiveness during differentiation. PLoS Pathog.
type 1 Tax through NF-kappaB in apoptosis-resistant onc.2015.510. [Epub 2016 Jan 25]. 2012;8:e1002617.
T-cell transfectants with Tax. J Virol. 1999;73: 265. Kataoka K, Nagata Y, Kitanaka A, et al. Integrated 287. Ray R, Lagging L, Meyer K, Ray R. Hepatitis C
7981–7987. molecular analysis of adult T cell leukemia/lym- virus core protein cooperates with ras and transforms
248. Kwok RP, Laurance ME, Lundblad JR, et al. Control phoma. Nat Genet. 2015;47:1304–1315. primary rat embryo fibroblasts to tumorigenic
of cAMP-regulated enhancers by the viral transactiva- 266. Yao F, Terrault N. Hepatitis C and hepatocellular phenotype. J Virol. 1996;70:4438–4443.
tor Tax through CREB and the co-activator CBP. carcinoma. Curr Treat Options Oncol. 2001;2:473– 288. Moriya K, Fujie H, Shintani Y, et al. The core protein
Nature. 1996;380:642–646. 483. of hepatitis C virus induces hepatocellular carcinoma
249. Suzuki T, Hirai H, Fujisawa J, et al. A trans-activator 267. Perz JF, Armstrong GL, Farrington LA, et al. The in transgenic mice. Nat Med. 1998;4:1065–1067.
Tax of human T-cell leukemia virus type 1 binds to contributions of hepatitis B virus and hepatitis C 289. Aoki H, Hayashi J, Moriyama M, et al. Hepatitis
NF-kappa B p50 and serum response factor (SRF) virus infections to cirrhosis and primary liver cancer C virus core protein interacts with the 14-3-3
and associates with enhancer DNAs of the NF-kappa worldwide. J Hepatol. 2006;45:529–538. protein and activates the kinase Raf-1. J Virol.
B site and CArG box. Oncogene. 1993;8:2391–2397. 268. Sun CA, Wu DM, Lin CC, et al. Incidence and 2000;74:1736–1741.
250. Maggirwar SB, Harhaj E, Sun SC. Activation of cofactors of hepatitis C virus–related hepatocellular 290. Hayashi J, Aoki H, Kajino K, et al. Hepatitis C virus
NF-kappa B/Rel by Tax involves degradation of carcinoma: a prospective study of 12,008 men in core protein activates the MAPK/ERK cascade syn-
I kappa B alpha and is blocked by a proteasome Taiwan. Am J Epidemiol. 2003;157:674–682. ergistically with tumor promoter TPA, but not with
inhibitor. Oncogene. 1995;11:993–998. 269. Yen T, Keefe EB, Ahmed A. The epidemiology of EGF or TGFalpha. Hepatology. 2000;32:958–961.
251. Suzuki T, Hirai H, Murakami T, Yoshida M. Tax hepatitis C virus infection. J Clin Gastroenterol. 2003; 291. Jin D, Wang H, Zhou Y, et al. Hepatitis C virus core
protein of HTLV-I destabilizes the complexes of NF- 36:47–53. protein-induced loss of LZIP function correlates with
kappa B and I kappa B-alpha and induces nuclear 270. WHO. WHO Global Surveillance and control cellular transformation. EMBO J. 2000;19:729–740.
translocation of NF-kappa B for transcriptional of hepatitis C. Report of a WHO Consultation 292. Yoshida T, Hanada T, Tokuhisa T, et al. Activation
activation. Oncogene. 1995;10:1199–1207. organized in collaboration with the viral Hepatitis of STAT3 by the hepatitis C virus core protein leads
252. Lemasson I, Polakowski NJ, Laybourn PJ, Nyborg Prevention Board. J Viral Hepat. 1999;6:35–47. to cellular transformation. J Exp Med. 2002;196:
JK. Transcription factor binding and histone 271. Di Bisceglie AM. Hepatitis C and hepatocellular 641–653.
modifications on the integrated proviral promoter carcinoma. Hepatology. 1997;26:34S–38S. 293. Li M, Zhao H, Zhang X, et al. Inactivating mutations
in human T-cell leukemia virus-I-infected T-cells. 272. Choo QL, Kuo G, Weiner AJ, et al. Isolation of a of the chromatin remodeling gene ARID2 in hepa-
J Biol Chem. 2002;277:49459–49465. cDNA clone derived from a blood-borne non-A, tocellular carcinoma. Nat Genet. 2011;43:828–829.
253. Suzuki T, Uchida-Toita M, Yoshida M. Tax protein non-B viral hepatitis genome. Science. 1989;244: 294. Schinazi RF, Asselah T. From HCV to HBV Cure.
of HTLV-I inhibits CBP/p300-mediated transcrip- 359–362. Liver Int. 2017;37(suppl 1):73–80.
tion by interfering with recruitment of CBP/p300 273. Rosenberg S. Recent advances in the molecular 295. Deleted in review.
onto DNA element of E-box or p53 binding site. biology of hepatitis C virus. J Mol Biol. 2001;313: 296. Cook-Mozaffari P, Newton R, Beral V, Burkitt DP.
Oncogene. 1999;18:4137–4143. 451–464. The geographical distribution of Kaposi’s sarcoma
254. Knoepfler PS, Eisenman RN. Sin meets NuRD and 274. Lamarre D, Anderson PC, Bailey M, et al. An NS3 and of lymphomas in Africa before the AIDS
other tails of repression. Cell. 1999;99:447–450. protease inhibitor with antiviral effects in humans epidemic. Br J Cancer. 1998;78:1521–1528.
255. Kinoshita T, Shimoyama M, Tobinai K, et al. Detec- infected with hepatitis C virus. Nature. 2003;426: 297. Geddes M, Franceschi S, Barchielli A, et al. Kaposi’s
tion of mRNA for the tax1/rex1 gene of human 186–189. sarcoma in Italy before and after the AIDS epidemic.
T-cell leukemia virus type I in fresh peripheral blood 275. Reiser M, Hinrichsen H, Benhamou Y, et al. Antiviral Br J Cancer. 1994;69:333–336.
mononuclear cells of adult T-cell leukemia patients efficacy of NS3-serine protease inhibitor BILN-2061 298. Chang Y, Cesarman E, Pessin MS, et al. Identification
and viral carriers by using the polymerase chain reac- in patients with chronic genotype 2 and 3 hepatitis of herpesvirus-like DNA sequences in AIDS-associ-
tion. Proc Natl Acad Sci USA. 1989;86:5620–5624. C. Hepatology. 2005;41:832–835. ated Kaposi’s sarcoma. Science. 1994;266:1865–1869.
256. Matsuoka M, Jeang KT. Human T-cell leukaemia 276. Smith DB, Pathirana S, Davidson F, et al. The 299. Dedicoat M, Newton R. Review of the distribution
virus type 1 (HTLV-I) infectivity and cellular origin of hepatitis C virus genotypes. J Gen Virol. of Kaposi’s sarcoma-associated herpesvirus (KSHV)
transformation. Nat Rev Cancer. 2007;7:270–280. 1997;78(Pt 2):321–328. in Africa in relation to the incidence of Kaposi’s
257. Larocca D, Chao LA, Seto MH, Brunck TK. Human 277. Lange CM, Moradpour D, Doehring A, et al. Impact sarcoma. Br J Cancer. 2003;88:1–3.
T-cell leukemia virus minus strand transcription of donor and recipient IL28B rs12979860 genotypes 300. Osmond DH, Buchbinder S, Cheng A, et al. Preva-
in infected T-cells. Biochem Biophys Res Commun. on hepatitis C virus liver graft reinfection. J Hepatol. lence of Kaposi sarcoma-associated herpesvirus infec-
1989;163:1006–1013. 2011;55:322–327. tion in homosexual men at beginning of and during
258. Yoshida M, Satou Y, Yasunaga J, et al. Transcriptional 278. Bukh J, Miller RH, Purcell RH. Genetic heterogene- the HIV epidemic. JAMA. 2002;287:221–225.
control of spliced and unspliced human T-cell ity of hepatitis C virus: quasispecies and genotypes. 301. Jacobson LP, Jenkins FJ, Springer G, et al. Interaction
leukemia virus type 1 bZIP factor (HBZ) gene. Semin Liver Dis. 1995;15:41–63. of human immunodeficiency virus type 1 and human
J Virol. 2008;82:9359–9368. 279. Lohmann V, Korner F, Koch J, et al. Replication of herpesvirus type 8 infections on the incidence of
259. Satou Y, Yasunaga J, Yoshida M, Matsuoka M. subgenomic hepatitis C virus RNAs in a hepatoma Kaposi’s sarcoma. J Infect Dis. 2000;181:1940–1949.
HTLV-I basic leucine zipper factor gene mRNA cell line. Science. 1999;285:110–113. 302. Cesarman E, Chang Y, Moore PS, et al. Kaposi’s
supports proliferation of adult T cell leukemia cells. 280. Bukh J, Pietschmann T, Lohmann V, et al. Muta- sarcoma-associated herpesvirus-like DNA sequences
Proc Natl Acad Sci USA. 2006;103:720–725. tions that permit efficient replication of hepatitis in AIDS-related body-cavity-based lymphomas.
260. Gaudray G, Gachon F, Basbous J, et al. The comple- C virus RNA in Huh-7 cells prevent productive N Engl J Med. 1995;332:1186–1191.
mentary strand of the human T-cell leukemia virus replication in chimpanzees. Proc Natl Acad Sci USA. 303. Soulier J, Grollet L, Oksenhendler E, et al.
type 1 RNA genome encodes a bZIP transcription 2002;99:14416–14421. Kaposi’s sarcoma-associated herpesvirus-like DNA
179.e6 Part I: Science and Clinical Oncology

sequences in multicentric Castleman’s disease. Blood. vGPCR initiates Kaposi’s sarcomagenesis and can carcinoma of European patients. J Invest Dermatol.
1995;86:1276–1280. promote the tumorigenic potential of viral latent 2009;129:248–250.
304. Martin JN. Diagnosis and epidemiology of human genes. Cancer Cell. 2003;3:23–36. 344. Kassem A, Schopflin A, Diaz C, et al. Frequent
herpesvirus 8 infection. Semin Hematol. 2003;40: 325. Bais C, Van Geelen A, Eroles P, et al. Kaposi’s detection of Merkel cell polyomavirus in human
133–142. sarcoma associated herpesvirus G protein-coupled Merkel cell carcinomas and identification of a unique
305. Uldrick TS, Whitby D. Update on KSHV epidemiol- receptor immortalizes human endothelial cells by deletion in the VP1 gene. Cancer Res. 2008;68:
ogy, Kaposi sarcoma pathogenesis, and treatment of activation of the VEGF receptor-2/ KDR. Cancer 5009–5013.
Kaposi sarcoma. Cancer Lett. 2011;305:150–162. Cell. 2003;3:131–143. 345. Shuda M, Feng H, Kwun HJ, et al. T antigen
306. Zhang T, Shao X, Chen Y, et al. Human herpes- 326. Mutlu AD, Cavallin LE, Vincent L, et al. In mutations are a human tumor-specific signature
virus 8 seroprevalence, China. Emerg Infect Dis. vivo-restricted and reversible malignancy induced for Merkel cell polyomavirus. Proc Natl Acad Sci
2012;18:150–152. by human herpesvirus-8 KSHV: a cell and animal USA. 2008;105:16272–16277.
307. Engels EA, Biggar RJ, Hall HI, et al. Cancer risk model of virally induced Kaposi’s sarcoma. Cancer 346. Carter JJ, Paulson KG, Wipf GC, et al. Association
in people infected with human immunodeficiency Cell. 2007;11:245–258. of Merkel cell polyomavirus-specific antibodies
virus in the United States. Int J Cancer. 2008;123: 327. Lagos D, Vart RJ, Gratrix F, et al. Toll-like receptor with Merkel cell carcinoma. J Natl Cancer Inst.
187–194. 4 mediates innate immunity to Kaposi sarcoma 2009;101:1510–1522.
308. Scadden DT. AIDS-related malignancies. Annu Rev herpesvirus. Cell Host Microbe. 2008;4:470–483. 347. Kean JM, Rao S, Wang M, Garcea RL. Seroepide-
Med. 2003;54:285–303. 328. Ishido S, Wang C, Lee BS, et al. Downregulation of miology of human polyomaviruses. PLoS Pathog.
309. Bower M, Palmieri C, Dhillon T. AIDS-related major histocompatibility complex class I molecules 2009;5:e1000363.
malignancies: changing epidemiology and the impact by Kaposi’s sarcoma-associated herpesvirus K3 and 348. Pastrana DV, Tolstov YL, Becker JC, et al. Quantita-
of highly active antiretroviral therapy. Curr Opin K5 proteins. J Virol. 2000;74:5300–5309. tion of human seroresponsiveness to Merkel cell
Infect Dis. 2006;19:14–19. 329. Dupuy S, Lambert M, Zucman D, et al. Human polyomavirus. PLoS Pathog. 2009;5:e1000578.
310. Newton R, Carpenter L, Casabonne D, et al. A Herpesvirus 8 (HHV8) sequentially shapes the NK 349. Tolstov YL, Pastrana DV, Feng H, et al. Human
prospective study of Kaposi’s sarcoma–associated cell repertoire during the course of asymptomatic Merkel cell polyomavirus infection II. MCV is a
herpesvirus and Epstein-Barr virus in adults with infection and Kaposi sarcoma. PLoS Pathog. common human infection that can be detected by
human immunodeficiency virus-1. Br J Cancer. 2012;8:e1002486. conformational capsid epitope immunoassays. Int
2006;94:1504–1509. 330. Ballestas ME, Kaye KM. Kaposi’s sarcoma-associated J Cancer. 2009;125:1250–1256.
311. Dupin N, Fisher C, Kellam P, et al. Distribution herpesvirus latency-associated nuclear antigen 1 350. Touze A, Gaitan J, Arnold F, et al. Generation of
of human herpesvirus-8 latently infected cells in mediates episome persistence through cis-acting Merkel cell polyomavirus (MCV)-like particles and
Kaposi’s sarcoma, multicentric Castleman’s disease, terminal repeat (TR) sequence and specifically binds their application to detection of MCV antibodies.
and primary effusion lymphoma. Proc Natl Acad Sci TR DNA. J Virol. 2001;75:3250–3258. J Clin Microbiol. 2010;48:1767–1770.
USA. 1999;96:4546–4551. 331. Hu J, Garber AC, Renne R. The latency-associated 351. Feng H, Shuda M, Chang Y, Moore PS. Clonal
312. Jenner RG, Boshoff C. The molecular pathology of nuclear antigen of Kaposi’s sarcoma-associated integration of a polyomavirus in human Merkel
Kaposi’s sarcoma–associated herpesvirus. Biochim herpesvirus supports latent DNA replication in cell carcinoma. Science. 2008;319:1096–1100.
Biophys Acta. 2002;1602:1–22. dividing cells. J Virol. 2002;76:11677–11687. 352. Foulongne V, Kluger N, Dereure O, et al. Merkel
313. Moore PS, Chang Y. Kaposi’s sarcoma-associated 331a. Chiu YF, Sugden AU, Fox K, et al. Kaposi’s cell polyomavirus in cutaneous swabs. Emerg Infect
herpesvirus-encoded oncogenes and oncogenesis. sarcoma-associated herpesvirus stably clusters its Dis. 2010;16:685–687.
J Natl Cancer Inst Monogr. 1998;65–71. genomes across generations to maintain itself extra- 353. Schowalter RM, Pastrana DV, Pumphrey KA,
314. Matolcsy A, Nador RG, Cesarman E, Knowles DM. chromosomally. J Cell Biol. 2017;216(9):2745–2758. et al. Merkel cell polyomavirus and two previously
Immunoglobulin VH gene mutational analysis sug- 332. Gradoville L, Gerlach J, Grogan E, et al. Kaposi’s unknown polyomaviruses are chronically shed from
gests that primary effusion lymphomas derive from sarcoma-associated herpesvirus open reading frame human skin. Cell Host Microbe. 2010;7:509–515.
different stages of B cell maturation. Am J Pathol. 50/Rta protein activates the entire viral lytic cycle in 354. Wieland U, Mauch C, Kreuter A, et al. Merkel cell
1998;153:1609–1614. the HH-B2 primary effusion lymphoma cell line. polyomavirus DNA in persons without Merkel cell
315. Nador RG, Cesarman E, Chadburn A, et al. Primary J Virol. 2000;74:6207–6212. carcinoma. Emerg Infect Dis. 2009;15:1496–1498.
effusion lymphoma: a distinct clinicopathologic 333. Ciufo DM, Cannon JS, Poole LJ, et al. Spindle 355. Houben R, Schrama D, Alb M, et al. Comparable
entity associated with the Kaposi’s sarcoma–associ- cell conversion by Kaposi’s sarcoma–associated expression and phosphorylation of the retinoblastoma
ated herpes virus. Blood. 1996;88:645–656. herpesvirus: formation of colonies and plaques protein in Merkel cell polyoma virus-positive and neg-
316. Mesri EA, Cesarman E, Boshoff C. Kaposi’s sarcoma with mixed lytic and latent gene expression in ative Merkel cell carcinoma. Int J Cancer. 2010;126:
and its associated herpesvirus. Nat Rev Cancer. infected primary dermal microvascular endothelial 796–798.
2010;10:707–719. cell cultures. J Virol. 2001;75:5614–5626. 356. Shuda M, Kwun HJ, Feng H, et al. Human Merkel
317. Cheng EH, Nicholas J, Bellows DS, et al. A Bcl-2 334. Moses AV, Fish KN, Ruhl R, et al. Long-term infec- cell polyomavirus small T antigen is an oncoprotein
homolog encoded by Kaposi sarcoma–associated tion and transformation of dermal microvascular targeting the 4E-BP1 translation regulator. J Clin
virus, human herpesvirus 8, inhibits apoptosis but endothelial cells by human herpesvirus 8. J Virol. Invest. 2011;121:3623–3634.
does not heterodimerize with Bax or Bak. Proc Natl 1999;73:6892–6902. 357. Arora R, Shuda M, Guastafierro A, et al. Survivin
Acad Sci USA. 1997;94:690–694. 335. Andrei G, Snoeck R. Kaposi’s sarcoma-associated is a therapeutic target in Merkel cell carcinoma. Sci
318. Ojala PM, Yamamoto K, Castanos-Velez E, et al. The herpesvirus: the role of lytic replication in targeted Transl Med. 2012;4:133ra156.
apoptotic v-cyclin-CDK6 complex phosphorylates therapy. Curr Opin Infect Dis. 2015;28(6):611–624. 358. Spurgeon ME, Cheng J, Bronson RT, Lambert PF,
and inactivates Bcl-2. Nat Cell Biol. 2000;2:819–825. 336. Moll I, Bladt U, Jung EG. Presence of Merkel cells in DeCaprio JA. Tumorigenic activity of merkel cell
319. Djerbi M, Screpanti V, Catrina AI, et al. The inhibi- sun-exposed and not sun-exposed skin: a quantitative polyomavirus T antigens expressed in the stratified
tor of death receptor signaling, FLICE-inhibitory study. Arch Dermatol Res. 1990;282:213–216. epithelium of mice. Cancer Res. 2015;75(6):1068–
protein defines a new class of tumor progression 337. Hodgson NC. Merkel cell carcinoma: changing 1079. doi:10.1158/0008-5472.CAN-14-2425. [Epub
factors. J Exp Med. 1999;190:1025–1032. incidence trends. J Surg Oncol. 2005;89:1–4. 2015/01/18]; PubMed PMID: 25596282. PMCID:
320. Bagneris C, Ageichik AV, Cronin N, et al. Crystal 338. Halata Z, Grim M, Bauman KI. Friedrich Sigmund 4359959.
structure of a vFlip-IKKgamma complex: insights Merkel and his “Merkel cell,” morphology, develop- 359. Verhaegen ME, Mangelberger D, Harms PW,
into viral activation of the IKK signalosome. Mol ment, and physiology: review and new results. Anat Vozheiko TD, Weick JW, Wilbert DM, et al.
Cell. 2008;30:620–631. Rec A Discov Mol Cell Evol Biol. 2003;271:225–239. Merkel cell polyomavirus small T antigen is
321. Liu L, Eby MT, Rathore N, et al. The human 339. Lucarz A, Brand G. Current considerations about oncogenic in transgenic mice. J Invest Dermatol.
herpes virus 8–encoded viral FLICE inhibitory Merkel cells. Eur J Cell Biol. 2007;86:243–251. 2015;135(5):1415–1424. doi:10.1038/jid.2014.446.
protein physically associates with and persistently 340. Moll I, Roessler M, Brandner JM, et al. Human [Epub 2014 Oct 14]; PMID: 25313532.
activates the Ikappa B kinase complex. J Biol Chem. Merkel cells—aspects of cell biology, distribution 360. Shuda M, Guastafierro A, Geng X, Shuda Y, Ostrowski
2002;277:13745–13751. and functions. Eur J Cell Biol. 2005;84:259–271. SM, Lukianov S, et al. Merkel cell polyomavirus
322. Asou H, Said JW, Yang R, et al. Mechanisms of 341. Sidhu GS, Chandra P, Cassai ND. Merkel cells, small T antigen induces cancer and embryonic
growth control of Kaposi’s sarcoma–associated herpes normal and neoplastic: an update. Ultrastruct Pathol. Merkel cell proliferation in a transgenic mouse
virus-associated primary effusion lymphoma cells. 2005;29:287–294. model. PLoS ONE. 2015;10(11):e0142329. doi:
Blood. 1998;91:2475–2481. 342. Feng H, Taylor JL, Benos PV, et al. Human tran- 10.1371/journal.pone.0142329. PMID: 26544690.
323. McCormick C, Ganem D. The kaposin B protein of scriptome subtraction by using short sequence tags to eCollection 2015.
KSHV activates the p38/MK2 pathway and stabilizes search for tumor viruses in conjunctival carcinoma. 361. Verhaegen ME, Mangelberger D, Harms PW, Eberl
cytokine mRNAs. Science. 2005;307:739–741. J Virol. 2007;81:11332–11340. M, Wilbert DM, Meireles J, et al. Merkel cell
324. Montaner S, Sodhi A, Molinolo A, et al. Endothelial 343. Becker JC, Houben R, Ugurel S, et al. MC polyomavirus small T antigen initiates Merkel cell
infection with KSHV genes in vivo reveals that polyomavirus is frequently present in Merkel cell carcinoma-like tumor development in mice. Cancer
Viruses and Human Cancer  •  CHAPTER 12 179.e7

Res. 2017;doi:10.1158/0008-5472.CAN-17-0035. 369. Ghosh SK, Wood C, Boise LH, et al. Potentiation 376. Koutsky LA, Ault KA, Wheeler CM, et al. Proof of
[Epub ahead of print]; PMID: 28512245. of TRAIL-induced apoptosis in primary effusion principle study, I. A controlled trial of a human papillo-
362. Cairo MS, Sposto R, Perkins SL, et al. Burkitt’s and lymphoma through azidothymidine-mediated inhibi- mavirus type 16 vaccine. NEJM. 2002;347:1645–1651.
Burkitt-like lymphoma in children and adolescents: tion of NF-kappa B. Blood. 2003;101:2321–2327. 377. Roden RB, Yutzy WHT, Fallon R, et al. Minor
a review of the Children’s Cancer Group experience. 370. Rizzetto M, Zanetti AR. Progress in the prevention capsid protein of human genital papillomaviruses
Br J Haematol. 2003;120:660–670. and control of viral hepatitis type B: closing remarks. contains subdominant, cross-neutralizing epitopes.
363. Ahmad SA, Bilimoria MM, Wang X, et al. Hepatitis J Med Virol. 2002;67:463–466. Virology. 2000;270:254–257.
B or C virus serology as a prognostic factor in 371. Zuckerman JN, Zuckerman AJ. Current topics in 378. Ganem D, Schneider RJ. Hepadnaviridae: the viruses
patients with hepatocellular carcinoma. J Gastrointest hepatitis B. J Infect. 2000;41:130–136. and their replication. In: Knipe DM, Howley PM,
Surg. 2001;5:468–476. 372. Winer RL, Lee SK, Hughes JP, et al. Genital human Griffin DE, et al, eds. Fields Virology. Philadelphia,
364. Rabe C, Pilz T, Klostermann C, et al. Clinical papillomavirus infection: incidence and risk factors PA: Lippincott Williams & Wilkins; 2001.
characteristics and outcome of a cohort of 101 in a cohort of female university students. Am J 379. Howley PM, Lowy DR. Papillomaviruses and
patients with hepatocellular carcinoma. World J Epidemiol. 2003;157:218–226. their replication. In: Knipe DM, Howley PM, eds.
Gastroenterol. 2001;7:208–215. 373. Christensen ND, Reed CA, Cladel NM, et al. Immu- Fields Virology. 4th ed. Philadelphia, PA: Lippincott
365. Wang H, Chia KS, Du WB, et al. Population-based nization with viruslike particles induces long-term Williams & Wilkins; 2001:2197–2229.
survival for cervical cancer in Singapore, 1968-1992. protection of rabbits against challenge with cot- 380. Green PL, Chen SY. Human T-cell leukemia virus
Am J Obstet Gynecol. 2003;188:324–329. tontail rabbit papillomavirus. J Virol. 1996;70:960– types 1 and 2. In: Knipe DM, Howley PM, eds.
366. Donato DM. Surgical management of stage 965. Fields Virology. 4th ed. Philadelphia, PA: Lippincott
IB-IIA cervical carcinoma. Semin Surg Oncol. 374. Suzich JA, Ghim SJ, Palmer-Hill FJ, et  al. Williams & Wilkins; 2001.
1999;16:232–235. Systemic immunization with papillomavirus L1 381. Major M, Rehermann B, Feinstone SM. Hepatitis
367. Yamaguchi K, Watanabe T. Human T lymphotropic protein completely prevents the development of C viruses. In: Knipe DM, Howley PM, eds. Fields
virus type-I and adult T-cell leukemia in Japan. Int viral mucosal papillomas. Proc Natl Acad Sci USA. Virology. Philadelphia: Lippincott Williams &
J Hematol. 2002;76(suppl 2):240–245. 1995;92:11553–11557. Wilkins.; 2001:1127–1161.
368. Tam HK, Zhang ZF, Jacobson LP, et al. Effect of 375. Harro CD, Pang YY, Roden RB, et al. Safety and 382. Moore P, Chang Y. Kaposi’s sarcoma–associ-
highly active antiretroviral therapy on survival among immunogenicity trial in adult volunteers of a human ated herpesvirus. In: Knipe DM, ed. Virology.
HIV-infected men with Kaposi sarcoma or non- papillomavirus 16 L1 virus-like particle vaccine. Philadelphia, PA: Lippincott Williams and Wilkins;
Hodgkin lymphoma. Int J Cancer. 2002;98:916–922. J Natl Cancer Inst. 2001;93:284–292. 2001:2803–2833.
13  Genetic Factors: Hereditary Cancer
Predisposition Syndromes
Michael F. Walsh, Karen Cadoo, Erin E. Salo-Mullen,
Marianne Dubard-Gault, Zsofia K. Stadler, and Kenneth Offit

S UMMARY OF K EY P OI N T S
• The discovery of inherited mutations syndromes can usually be confirmed to tumors to discover targets for
of genes associated with increased with molecular genetic testing of therapy. Because tumor genomic
risk for cancer provides important patients who have hereditary analysis will also include a
clinical opportunities for early malignancies. Genetic testing can comparison with the germline, the
detection and prevention of common then be extended to relatives as a need for genetic counseling for both
and rare forms of human predictive test to guide their cancer and noncancer disease risks
malignancies. preventive management. will be increased. This chapter
• Syndromes of cancer predisposition • Medical, surgical, and radiation reviews both common and more
often involve multiple organ systems, oncologists, genetic counselors, and recently described familial cancer
affect paired organs with bilateral or allied professionals are playing a syndromes, with an emphasis on the
multifocal tumors, and have onset leading role in the integration of clinical application of cancer genetic
at an earlier age compared with genetic testing into the practice of and genomic analysis in the
nonfamilial tumors. The diagnosis of preventive oncology. Recently, management of patients who have or
particular cancer predisposition genomic analysis has been applied are at risk for cancer.

During the past decade, the availability of clinical testing for inherited or may occur de novo in families in which the history of cancer may
mutations of cancer predisposition genes has had a major impact on be unremarkable. In many instances, the same genes recurrently mutated
the practice of clinical oncology.1–3 As these genes were identified and in specific sporadic cancers, if mutated in the germline, are also
characterized, guidelines for the responsible clinical translation of this susceptibility genes for the same types of tumors. This phenomenon
information were developed by medical and surgical subspecialty reflects the genotype-phenotype relationships that arise as a result of
societies, such as the American Society of Clinical Oncology (updated aberration of particular biologic pathways and thus define the patterns
in 2016),4 the American Association for Cancer Research Pediatric that characterize hereditary cancer syndromes. Increasingly, specific
Cancer Working Group,5–8 and other organizations.9–13 These guidelines pathologic hallmarks have been associated with cancer predisposition
emphasized that in the process of offering a predictive genetic test to syndromes (see Table 13.1). It is now widely accepted that cancer
a patient or family that is affected by cancer, the provider and the pathogenesis derives from inherited or acquired alterations in a set of
individual who is being tested must be prepared to deal with all the “driver” genes that are associated with abnormal cellular function,
medical, psychological, and social consequences of a positive, negative, resulting in uncontrolled cell division and/or loss of the normal fidelity
or ambiguous result. These guidelines define the form and content of DNA repair and replication. This process results in accumulation
of genetic counseling as a component of cancer risk assessment and of further mutations characterized as “passengers” in the neoplastic
management. process which contribute to tumor survival.16 With the advent of
A selected set of syndromes of cancer predisposition is reviewed massively parallel sequencing, progress in the understanding of tumor
in this chapter (Tables 13.1 and 13.2). Detailed discussions of breast, genetics has catalogued these “driver” and “passenger” mutations,
colon, gastric, gynecologic, pancreatic, prostate, and renal cancer illuminating biologic pathways and facilitating development of therapies
susceptibility are found in the chapters that discuss these tumors. targeted toward these pathways. At the same time, the definition of
Whether offered by a physician, genetic counselor, or other health pathways perturbed in malignant transformation and progression has
care professional, genetic testing for inherited cancer risk requires provided novel biomarkers of utility for disease prognosis and, as is
careful informed consent. The elements of informed consent for genetic the focus here, disease susceptibility.
testing are summarized in Box 13.1. With the advent of genomic The syndromes of cancer susceptibility included in this chapter
screening of tumors, gene panel testing, and genomic screening in are those that are most commonly encountered in oncologic
the germline, these elements of informed consent must also include practice, as well as several recently defined entities, some of which
the eventuality of “incidental” findings associated with risk of cancer, have been associated with targeted therapies. The most common of
noncancer diseases, and nonpaternity.14,15 these syndromes, predisposing to cancers of the breast, ovary, colon,
Mutations (i.e., pathogenic variants) in genes whose alterations prostate, and pancreas, affect tens of thousands of Americans who
result in susceptibility to cancer may be inherited, whereby a family are diagnosed with these cancers each year in the United States and
history usually includes multiple individuals diagnosed with cancer, Text continued on p. 194

180
Genetic Factors: Hereditary Cancer Predisposition Syndromes  •  CHAPTER 13 181

Table 13.1  Pathologic Hallmarks of Selected Cancer Predisposition Syndromes


Neoplastic or Preneoplastic Pathologic Conditions Associated Genetic Syndromes (Gene)
Atypical rhabdoid tumor, schwannomatosis Rhabdoid tumor predisposition syndrome (SMARCB1)
Wilms tumor Li-Fraumeni syndrome (TP53), (WT1),(BWS), Simpson-Golabi-
Behmel syndrome, Beckwith-Wiedemann syndrome, Fanconi
anemia compound heterozygotes (see Fanconi anemia)
Ductal breast cancer with estrogen receptor, progesterone receptor, and Hereditary breast and ovarian cancer syndrome (BRCA1)
HER2-neu negativity (“triple negative”)
Lobular breast cancer Hereditary diffuse gastric cancer syndrome (CDH1)
High-grade serous (papillary serous) ovarian cancer Hereditary breast and ovarian cancer syndrome (see Table 13.2)
Hepatoblastoma Familial adenomatous polyposis (APC), Beckwith-Wiedemann
syndrome, Simpson-Golabi-Behmel syndrome, Sotos syndrome
Hereditary paraganglioma-pheochromocytoma Hereditary paraganglioma-pheochromocytoma syndromes (SDHB,
SDHC, SDHD, SDHAF2)
Hypodiploid leukemia Li-Fraumeni syndrome (TP53)
Clear cell or endometrioid ovarian cancers Lynch syndrome (MLH1, MSH2, MSH6, PMS2, EPCAM)
Ovarian sex cord tumors with annular tubules (SCTATs) Peutz-Jeghers syndrome (STK11)
Clear cell or endometrioid endometrial cancers Lynch syndrome (MLH1, MSH2, MSH6, PMS2, EPCAM)
Uterine leiomyomas Hereditary leiomyoma renal cell carcinoma syndrome (FH)
Adenoma malignum of the cervix Peutz-Jeghers syndrome (STK11)
Colorectal cancer with tumor infiltrating lymphocytes, Crohnlike lymphocytic Lynch syndrome (MLH1, MSH2, MSH6, PMS2, EPCAM)
reaction, mucinous or signet ring cells, medullary growth pattern
Lauren diffuse type, signet ring cell, or linitus plastica gastric cancer Hereditary diffuse gastric cancer syndrome (CDH1)
Leiomyosarcoma Li-Fraumeni syndrome (TP53), retinoblastoma (RB1)
Hamartomatous gastrointestinal polyps Peutz-Jeghers syndrome (STK11); juvenile polyposis syndrome
(BMPR1A, SMAD4) ; Cowden syndrome (PTEN)
Hypermutable tumors Constitutional mismatch repair deficiency, Lynch syndrome (MLH1
MSH2 MSH6 PMS2 EPCAM)
Paget breast tumor Li-Fraumeni syndrome (TP53)
Pancreatic neuroendocrine tumors Multiple endocrine neoplasia type 1 (MEN1) ; von Hippel-Lindau
disease (VHL)
Renal papillary carcinoma type I Hereditary papillary renal cancer syndrome (MET)
Renal papillary carcinoma type II, collecting duct, tubulopapillary Hereditary leiomyoma renal cell carcinoma syndrome (FH)
Renal clear cell carcinoma von Hippel-Lindau disease (VHL)
Renal oncocytoma Birt-Hogg-Dube syndrome (FLCN); tuberous sclerosis complex
(TSC1)
Renal angiomyolipoma Tuberous sclerosis complex (TSC1)
Renal chromophobe Birt-Hogg-Dube syndrome (FLCN)
Renal medullary cancer Sickle cell trait
Sertoli cell tumors of the testes Peutz-Jeghers syndrome (STK11);
Small cell carcinoma of the ovary–hypercalcemic type (SCCOHT) Rhabdoid tumor predisposition syndrome (SMARCA4)
Medullary thyroid cancer Multiple endocrine neoplasia type 2 (RET)
Meningioma Neurofibromatosis type 2 (NF2), Gorlin syndrome (PTCH1)
Cribriform-morula variant of papillary thyroid carcinoma Familial adenomatous polyposis (APC)
Follicular thyroid cancer Cowden syndrome (PTEN)
Medulloblastoma Turcot syndrome variant of familial adenomatous polyposis (APC);
Gorlin syndrome (PTCH1), Fanconi anemia (FANC complex genes),
Glioblastoma Turcot syndrome variant of Lynch syndrome (MLH1, MSH2, MSH6,
nPMS2)
Basal cell carcinoma Xeroderma pigmentosum (XPA,B,C,D; DDB2, ERCC4, ERCC5, POLH);
Gorlin syndrome (PTCH1)
Schwannomatosis Neurofibromatosis type 2; rhabdoid predisposition syndrome
(SMARCB1, LZTR1)
Sebaceous adenoma, sebaceous carcinoma, keratoacanthoma Muir Torre syndrome variant of Lynch syndrome (MLH1, MSH2)
Fibrofolliculomas Birt-Hogg-Dube syndrome (FLCN)
Facial tricholemmomas Cowden syndrome (PTEN)
Table 13.2  Cancer Susceptibility Genes, Syndromes, and Management Considerations
Genes and Details Syndrome Penetrance Associated Neoplasms Management Considerations Other Notes
231,233,422,423
APC Familial High Colorectal lesions (adenomatous Colonoscopy and total colectomy based Genotype-phenotype correlations
Tumor suppressor adenomatous polyposis and cancer) on polyp burden between classic FAP and AFAP: Mutations
Dominant inheritance polyposis (FAP) Gastric and duodenal or small bowel Upper endoscopy on the 5′ and 3′ ends of the gene
Attenuated FAP lesions (adenomatous polyps and cancer) Consideration of small bowel visualization correlate with AFAP
(AFAP) Desmoid tumors Thyroid examination and consideration of High de novo mutation rate (~20%–25%)
Variant: Turcot Thyroid (typically papillary) ultrasound examination Extracolonic features: Congenital
182 Part I: Science and Clinical Oncology

syndrome Hepatoblastoma (patients usually ≤5 yr Consideration of childhood liver hypertrophy of the retinal pigment
old) palpation, abdominal ultrasound epithelium (CHRPE); osteomas,
Pancreatic examination, AFP measurement supernumerary teeth; desmoids,
medulloblastoma (Turcot syndrome) (investigational) epidermoid cysts; gastric fundic gland
polyps
Particularly cribriform-morula variant of
papillary thyroid carcinoma
Chemoprevention not a replacement for
colonoscopy and colectomy
APC*I1307K Moderate Colorectal Colonoscopy based on family history or Ashkenazi Jewish founder mutation
(c.3920T>A)265,424,425 similar to first-degree relative risk Not associated with polyposis
ATM5,84,110,421,426–429 Ataxia Moderate Monoallelic carriers: Monoallelic carriers: Breast screening with Monoallelic carriers: Limited evidence for
telangiectasia (AT) (monoallelic/ Breast, possibly pancreas annual mammogram and breast magnetic pancreas or prostate cancer
heterozygous Biallelic carriers: resonance imaging (MRI) starting at age Risk of autosomal recessive AT condition
carriers) Leukemia, lymphoma 40 yr or per family historya in biallelic offspring of heterozygous
Rare: High Biallelic carriers: AT specialist for carriers
(biallelic) multidisciplinary management: screening AT: Childhood cerebellar ataxia,
complete blood count (CBC) with telangiectasias of the conjunctivae,
differential; consider bone marrow biopsy, immunodeficiency, sensitivity to
AFP measurement radiation
Immunology: Monitoring immunoglobulin
levels per immunologist recommendation
Dermatology: annual skin examination
Pulmonary: Baseline and as-needed
pulmonary function tests
Gastroenterology and nutrition: Baseline
and as-needed swallowing function and
nutritional management
Endocrine: Annual diabetes screen
Neurology: Supportive medications
Orthopedics: Annual scoliosis evaluation
Dental: Biannual examination
BAP1411–413,430 BAP1 tumor Undetermined Atypical Spitz tumors Eye examinations Limited evidence for possible other
Possible tumor predisposition Uveal melanoma Dermatologic examinations associated tumors: non–small cell lung
suppressor syndrome Malignant mesothelioma Consideration of abdominal ultrasound adenocarcinoma, breast cancer,
Dominant inheritance Cutaneous melanoma examination and/or MRI of the kidneys cholangiocarcinoma, meningioma,
Clear cell renal cell carcinoma neuroendocrine carcinoma
Basal cell carcinoma
BLM5,431–436 Bloom syndrome Low or Biallelic: Myelodysplasia Biallelic carriers: Bloom syndrome Monoallelic carriers: Limited evidence for
undetermined Variety of epithelial carcinomas specialist for management: risk of colorectal and breast cancer
(monoallelic/ (gastrointestinal, genitourinary), CBC every 3–4 mo, avoidance of radiation, Bloom syndrome: Chromosome breakage
heterozygous lymphoid, hematopoietic, sarcomas, CNS, breast MRI or ultrasound starting at 18 yr syndrome with growth deficiency,
carriers) and others of age, annual colonoscopy starting at erythematous facial skin lesion,
Rare: High age 15 yr, renal ultrasound examination at immunodeficiency, frequent infections
(biallelic) diagnosis and every 3 mo through age Founder mutation in Ashkenazi Jewish
8 yr to assess for Wilms tumor, HPV population
vaccine per AAP guidelines
Dermatology: Annual skin examination,
limit sun exposure
Pulmonary: Pulmonary function tests
Gastroenterology and nutrition: Baseline
and as-needed swallowing function
evaluation and nutritional management
Endocrine: Annual fasting blood sugar
and TSH level
Orthopedics: Annual scoliosis evaluation
Dental: Biannual examination
BMPR1A and Juvenile polyposis High Gastrointestinal lesions (juvenile/ Colonoscopy based on polyp burden “Juvenile” refers to polyp histology, not
SMAD4423,437–443 syndrome hamartomatous polyps and cancer— Upper endoscopy age of onset
SMAD4: Tumor colorectal, stomach, small bowel) Surgical management based on polyp SMAD4 mutations associated with HHT
suppressor burden SMAD4 mutations associated with
Dominant inheritance Hereditary hemorrhagic telangiectasia massive gastric polyposis
(HHT) screening Limited evidence for association with
pancreas
BRCA1 and Hereditary breast High Breast (BRCA1: often triple-negative Breast screening with annual Founder mutations in certain
BRCA25,91,101,127,137,444–449 and ovarian tumor) mammogram and breast MRI starting at populations—Ashkenazi Jewish,
Tumor suppressor cancer syndrome Male breast age 25–30 yr Icelandic, and others
Dominant inheritance (HBOC) Ovarian (epithelial; high-grade serous), Male self- and clinical-breast examination Autosomal recessive Fanconi anemia
fallopian tube, primary peritoneal Optional risk-reducing mastectomy with biallelic germline mutations
Prostate (high Gleason score) Risk-reducing BSO BRCA1/2 heterozygous mutations
Pancreatic (exocrine) PSA measurement and digital rectal detected in childhood through tumor
examination normal sequencing should be reviewed
Consideration of eligibility and pros and with clinical genetics team and treating
cons of chemoprevention with tamoxifen oncology team for importance
Consideration of eligibility for treatment pertaining to care of relatives and
with PARP inhibitors potentially molecularly based therapy
Investigational-based pancreas screening

Continued
Genetic Factors: Hereditary Cancer Predisposition Syndromes  •  CHAPTER 13 183
Table 13.2  Cancer Susceptibility Genes, Syndromes, and Management Considerations—cont’d
Genes and Details Syndrome Penetrance Associated Neoplasms Management Considerations Other Notes
184 Part I: Science and Clinical Oncology

BRIP184,113,450 Moderate Ovarian (heterozygous carriers) Consider risk-reducing BSO at age Risk of autosomal recessive Fanconi
Possible tumor (monoallelic/ 50–55 yr or as per family historyb anemia in biallelic offspring of
suppressor heterozygous heterozygous carriers
carriers)
Rare: High
(biallelic)
CDH1269,271,451 Hereditary diffuse High Diffuse gastric cancer Prophylactic total gastrectomy Upper endoscopy not proven to be an
Tumor suppressor gastric cancer Lobular breast cancer Breast screening with annual effective method of screening or
Dominant inheritance syndrome mammogram and breast MRI detecting diffuse gastric cancer
Optional risk-reducing mastectomy Insufficient evidence for colorectal
cancer
CDKN2A293,452,453 Familial atypical High Melanoma and dysplastic nevi Dermatologic examination Risk of melanoma may be independent
Tumor suppressor multiple mole Pancreas Investigational-based pancreatic of genetic test result
CDK4 oncogene melanoma screening CDKN2a (p16) founder mutation in
Dominant inheritance (FAMMM) Netherlands
syndrome
CHEK25,63,65,84,109,425 Moderate Breast Breast screening with annual Genotype-phenotype: Different risks for
Tumor suppressor Colon mammogram and breast MRI starting at truncating mutations vs missense
Dominant inheritance age 40 yr or per family historya mutations
Colonoscopy based on family history or Limited evidence for association with
similar to first-degree relative risk prostate cancer
DICER1454–459 High Pleuropulmonary blastoma (PPB) No guidelines have been established Other features: Ciliary body
Dominant inheritance Ovarian sex cord-stromal tumors Consideration of annual physical medulloepithelioma; botryoid-type
(Sertoli-Leydig cell tumor, juvenile examination with targeted systems review embryonal rhabdomyosarcoma of the
granulose cell tumor, gynandroblastoma) and possible imaging cervix or other sites; nasal
Cystic nephroma chondromesenchymal hamartoma; renal
Thyroid gland neoplasia (cancer, sarcoma; pituitary blastoma;
multinodular goiter, adenomas) pineoblastoma
Early-onset disease—before age 40 yr
Maternal fetal medicine care when lung
cysts are identified prenatally
Papillary or follicular thyroid cancer
GREM1 (SCG5- Hereditary mixed High Colorectal lesions (polyps of mixed Colonoscopy and surgical management Only a 40-kb duplication upstream of
GREM1)262,460,461 polyposis histologies and cancer) based on polyp burden GREM1 (spanning 3′ end of SCG5 and
Dominant inheritance syndrome region upstream of GREM1) implicated in
disease; identified in Ashkenazi Jewish
population
ETV6/CEBPA/ Familial leukemia/ High ALL, AML, MDS History and physical examination Other syndromes predisposing to
PAX5/RUNX1 MDS Medical history: Prior cytopenias, bleeding leukemia include LFS (TP53), CMMRD
IKAROS/SAMD9/ history, nononcologic manifestations (MLH1, MSH2, MSH6, PMS2), T21, Bloom
SAMD9L462–465 Family history: (BLM), Nijmegen (NBN), ataxia
Dominant Inheritance Review and document types of cancer telangiectasia (ATM), NF1, Noonan:
and leukemia, ages at cancer or leukemia PTPN11, CBL (RAS-activating syndromes),
onset Fanconi anemia (FANCA-E, BRCA, RAD51D
Include history of antecedent cytopenias [autosomal recessive]), telomere
and/or bleeding syndromes (TERT, TERC, DKC), severe
Physical examination: congenital neutropenia (ELANE, HAX1),
Signs of leukemia, lymphoma Diamond-Blackfan syndrome (RPS19,
Other syndrome specific findings, RPLS, RPL11), Shwachman-Diamond
including signs of solid tumors syndrome (SBDS), GATA2, monosomy 7
CBC
Manual differential
Reticulocyte count
Blood smear with morphology
Bone marrow evaluation
Aspirate and biopsy
Morphology
Cytogenetics
Patient and family education about signs
and symptoms of cancer, including
leukemia
HSCT consultation
Consider HLA typing and genetic testing
of potential familial donors
Discuss enrollment in registries or other
research studies

Continued
Genetic Factors: Hereditary Cancer Predisposition Syndromes  •  CHAPTER 13 185
Table 13.2  Cancer Susceptibility Genes, Syndromes, and Management Considerations—cont’d
Genes and Details Syndrome Penetrance Associated Neoplasms Management Considerations Other Notes
Fanconi anemia Fanconi anemia High: Myelodysplastic syndrome Fanconi anemia specialist for Increased chromosome breakage in
(multiple genes—at Biallelic for Acute myeloid leukemia management lymphocytes tested with diepoxybutane
least 20)5,466–472 the recessive Squamous cell carcinoma of head and HSCT and mitomycin C (does not identify
Recessive inheritance genes and neck, esophagus, vulva Early detection and surgical management carriers)
(most genes) monoallelic Genitourinary (cervix, Wilms tumor, of solid tumors: oral/ otolaryngology/ Physical abnormalities (not in all
Dominant inheritance for RAD51, neuroblastoma) ear-nose-throat examination; gynecologic patients): Short stature, abnormal skin
(RAD51 gene) FANCB, BRCA2, Other solid tumors: Liver, brain, skin, examination. pigmentation (café au lait or
X-linked inheritance PALB2, BRCA1 breast, gastrointestinal HPV prevention hypopigmentation or
186 Part I: Science and Clinical Oncology

(FANCB gene) Moderate: Associated cancers for BRCA2, PALB2, Management for BRCA2, PALB2, BRIP1, and hyperpigmentation), skeletal
Monoallelic BRIP1, and RAD51C carriers RAD51C carriers (discussed above and malformations of the upper and lower
for BRIP1, below) limbs (thumbs, radii, hands, ulnae),
RAD51C microcephaly, developmental delay, and
ophthalmic, genitourinary, cardiac,
gastrointestinal, CNS anomalies
Progressive bone marrow failure (not in
all patients): Pancytopenia,
thrombocytopenia, leucopenia
Extreme toxicities from chemotherapy or
radiation
Genes and complement groups:
BRCA2 FA-D1
BRIP1 FA-J
PALB2 FA-N
RAD51 FA-R
RAD51C FA-O
BRCA1 FANCES
XRCC2 FANCU
FH359,374,398,459,473–476 Hereditary High Cutaneous leiomyomata Dermatologic and gynecologic Fumarate hydratase enzyme assay might
Tumor suppressor leiomyomatosis Uterine leiomyomata examination—evaluate for changes be helpful in some situations
Dominant inheritance and renal cell Renal tumors (type 2 papillary, tubule- suggestive of leiomyosarcoma Risk for uterine leiomyosarcoma is
cancer (HLRCC) papillary, collecting duct carcinomas; Medication or resection of leiomyomata unclear
unilateral, solitary lesions) Imaging for and surgical management of Biallelic FH mutations cause a recessive
renal tumors consider starting at age 8 disorder known as fumarate hydratase
deficiency—metabolic disorder, profound
developmental delay, seizures, fumaric
aciduria
FLCN398,477–480 Birt-Hogg-Dube High Cutaneous (fibrofolliculomas, No consensus at this time Fibrofolliculomas are specific to BHD
Tumor suppressor (BHD) syndrome trichodiscomas/ angiofibromas, Dermatologic care Lung cysts are multiple and bilateral
Dominant inheritance perifollicular fibromas, acrochordons) Imaging for and surgical management Spontaneous pneumothorax
Pulmonary cysts (nephron-sparing) of renal tumors Renal tumors are multifocal, bilateral,
Renal tumors (oncocytoma, slow growing
chromophobe, oncocytic hybrid tumors) Other features: Parotid lesions, oral
papules, thyroid lesions
Unclear data regarding colon cancer
Possible genotype-phenotype
correlations
KIT (C-Kit)406,481–484 High Gastrointestinal stromal tumors (GISTs) No consensus at this time Diffuse interstitial cell of Cajal
Oncogene Imaging and endoscopy might be hyperplasia (ICCH)
Dominant inheritance considered Skin hyperpigmentation or
hypopigmentation
Mast-cell disorders
Related gene: PDGFRA
MEN18,485–488 Multiple High Gastroenteropancreatic (GEP) tract well- Biochemical testing Primary hyperparathyroidism,
Tumor suppressor endocrine differentiated endocrine tumors Imaging hypercalcemia, oligomenorrhea or
Dominant inheritance neoplasia type 1 Pituitary tumors (prolactinoma) Surgical management amenorrhea, galactorrhea, Zollinger-
(MEN1) Parathyroid tumors Various medications Ellison syndrome (gastrinoma),
Carcinoid tumors insulinoma, glucagonoma, vasoactive
Adrenocortical tumors intestinal peptide (VIP)–secreting tumor
(VIPomas)
Other features: Skin (angiofibromas,
collagenomas), lipomas, CNS lesions
(meningioma, ependymoma),
leiomyomas
MET (c-Met)398,489,490 Hereditary High Papillary renal cancer (multifocal, Imaging
Oncogene papillary renal bilateral, type 1 papillary) Surgical management
Dominant inheritance carcinoma (HPRC)
MLH1, MSH2, Lynch syndrome High Colorectal Colonoscopy every 1–2 yr starting at age Tumor analyses with
MSH6, PMS2, (formerly: Endometrial 20–25 yr immunohistochemical (IHC) staining,
EPCAM167,173,175,177,180,182, hereditary Ovarian (epithelial) Consideration of prophylactic colectomy microsatellite instability (MSI) analysis,
188,421,491–504
non-polyposis Stomach Hysterectomy and risk-reducing BSO somatic tumor genetic analysis, BRAF
Tumor suppressor colorectal cancer Small bowel Consideration of upper endoscopy V600E somatic analysis, MLH1 promoter
Dominant inheritance [HNPCC]) Upper urinary tract (renal pelvis, ureter) Consideration of urinalysis or urine hypermethylation analysis
Variant: Muir-Torre Pancreas cytology Amsterdam Criteria I and II; Revised
syndrome Hepatobiliary tract Consideration of eligibility for treatment Bethesda Guidelines
Variant: Turcot Sebaceous neoplasms (Muir-Torre with cancer immunotherapy with Recent recommendations for universal
syndrome syndrome) checkpoint blockade screening of colorectal and endometrial
Variant: Glioblastoma (Turcot syndrome) Consideration of aspirin for cancers
Constitutional chemoprevention Genotype-phenotype correlations
mismatch repair CMMR-D recessive syndrome emerging but not significant enough to
deficiency MRI of brain every 6 mo starting at alter management
syndrome diagnosis Limited evidence for moderate risk of
[CMMR-D] Whole-body MRI starting at age 6 once a breast and prostate cancer
(biallelic year Autosomal recessive inheritance—
mutations; CBC every 6 mo biallelic (homozygote or compound
recessive Abdominal ultrasound examination every heterozygote) with childhood onset of
inheritance) 6 mo severe CMMR-D syndrome—café au lait
Upper GI endoscopy, VCE, macules, solid tumors, hematologic
ileocolonoscopy—start at 4–6 yr then cancers
every 12 mo
Gynecologic examination, transvaginal
ultrasound, urine cytology, dipstick
starting at age 20 then every 12 mo

Continued
Genetic Factors: Hereditary Cancer Predisposition Syndromes  •  CHAPTER 13 187
Table 13.2  Cancer Susceptibility Genes, Syndromes, and Management Considerations—cont’d
188 Part I: Science and Clinical Oncology

Genes and Details Syndrome Penetrance Associated Neoplasms Management Considerations Other Notes
MUTYH236,247,505–508 MUTYH-associated High Colorectal lesions (adenomatous Colonoscopy and total colectomy based Phenotypically similar to AFAP
Recessive inheritance polyposis (MAP) polyposis and cancer; serrated and on polyp burden Two Northern European founder
hyperplastic polyps also described) Upper endoscopy mutations
Stomach Controversy regarding risk in
Duodenal lesions (adenomatous polyps, heterozygous carriers (see below)
cancer)
MUTYH425,506,509,510 Moderate Colorectal Colonoscopy based on family history or Controversy regarding risk
Monoallelic/ similar to risk with first-degree relative
heterozygous carrier
NBN5,84,421,429,511,512 Nijmegen Moderate Monoallelic carrier: Monoallelic carriers: Breast screening with Heterozygous carrier breast cancer risk;
breakage (monoallelic/ Breast annual mammogram and breast MRI limited evidence for prostate cancer
syndrome (NBS) heterozygous Biallelic carrier: starting age at age 40 yr or as per family Slavic founder mutation
carriers) Lymphoma historya Risk of autosomal recessive condition in
Rare: High Biallelic carriers: NBS specialist for offspring of heterozygous carriers
(biallelic) multidisciplinary management (NBS)
Pediatric autosomal recessive syndrome NBS: Chromosomal breakage,
Hematology-oncology: History and microcephaly, dysmorphic features,
physical examination, annual CBC, immunodeficiency, lymphomas
metabolic profile and lactate
dehydrogenase, avoid excessive radiation,
HPV vaccine per AAP guidelines
Dermatology: Annual skin examination
Pulmonary: Baseline pulmonary function
tests with follow-up as needed, aggressive
treatment of recurrent infections
Gastroenterology and nutrition: Baseline
and as-needed swallowing function
evaluation and nutritional management
Endocrine: Monitor growth, assess female
patients for ovarian failure
Neurology: Developmental assessment
and early intervention if needed
Ophthalmology: Annual examination
Orthopedics: Baseline assessment for
anomalies and as needed
Dental: Biannual examination
NF1482,513–517 Neurofibromatosis High Cutaneous neurofibromas NF1 specialist for management Café au lait macules, axillary and inguinal
Tumor suppressor type 1 (moderate Plexiform neurofibromas Physical examination; ophthalmologic freckling, iris Lisch nodules, learning
Dominant inheritance risk for breast Optic nerve and other CNS gliomas examination; imaging; surgical disabilities, scoliosis, tibial dysplasia,
cancer) Malignant peripheral nerve sheath management vasculopathy
tumors Breast screening with annual
Breast (moderate risk) mammogram and breast MRI as per
Others: GIST, leukemia family historya
Children with NF1 should have
ophthalmic assessments every 6 to 12 mo
from birth to 8 yr; one baseline
assessment of color vision and visual
fields should be undertaken when the
child is developmentally able
Assess with history and clinical
examination annually for typical signs of
MPNST: any nondermal neurofibroma
with rapid growth, loss of neurologic
function, or increasing pain or change in
consistency
Assess for risk of JMML in NF1 in children
with juvenile xanthogranulomas
Baseline whole-body MRI should be
considered between ages 16 and 20 yr to
assess internal tumor burden to
determine adult follow-up regimen if
signs of MPNST are present
MRI with or without FDG-PET is
recommended
NF2/SMARCB1/ Neurofibromatosis High Acoustic neuromas Annual history and physical examination
LZTR1/SMARCE1518 type 2 Schwannomatosis (including audiology with measurement
Dominant inheritance Meningiomas of pure-tone thresholds and Word
Leukemia Recognition Scores)
Atypical rhabdoid tumor Annual (consider twice yearly in first year
after diagnosis or signs of rapid growth)
brain MRI starting at 10 yr of age;
screening may begin earlier in patients
with high-risk genotypes or symptomatic
diagnoses
If baseline imaging shows no
characteristic sites of involvement, reduce
frequency of screening to every 2 yr
Protocols should include high-resolution
(1- to 3-mm section thickness) imaging
through the internal auditory meatus,
preferably in at least two orthogonal
planes
Surveillance spinal MRI is recommended
at 24- to 36-month intervals beginning at
10 yr of age
Whole-body MRI may be performed

Continued
Genetic Factors: Hereditary Cancer Predisposition Syndromes  •  CHAPTER 13 189
Table 13.2  Cancer Susceptibility Genes, Syndromes, and Management Considerations—cont’d
Genes and Details Syndrome Penetrance Associated Neoplasms Management Considerations Other Notes
PALB217,49–51,84,448,519 High Breast Breast screening with annual Insufficient evidence for ovarian cancer
Tumor suppressor Limited evidence for pancreatic cancer mammogram and breast MRI starting at Risk of autosomal recessive Fanconi
190 Part I: Science and Clinical Oncology

Dominant inheritance age 30 yr or per family historya anemia in biallelic offspring of
Optional risk-reducing mastectomy based heterozygous carriers
on family history
Investigational-based pancreas screening
POLD1, Polymerase High Colorectal lesions (polyps and cancer) Colonoscopy and surgical management Disease-causing mutations occur in the
POLE260,261,520–523 proofreading- based on polyp burden exonuclease domain
Dominant inheritance associated Limited data regarding extracolonic
polyposis (PPAP) cancer risk
PRSS1, SPINK1, Hereditary High (PRSS1) Pancreas Pancreatitis management Pancreatitis—recurrent, acute, chronic
CFTR, CTRC261,524–526 pancreatitis Variable throughout lifetime
Various modes of (SPINK1, CFTR, More severe disease if biallelic mutations
inheritance CTRC) are present
Biallelic (homozygote or compound
heterozygote) CFTR mutations associated
with cystic fibrosis
PTCH, SUFU527–530 Gorlin syndrome/ High Jaw (odontogenic) keratocysts Referral to a specialist for management Other features: Congenital skeletal
Tumor suppressor nevoid basal cell Basal cell carcinomas Physical examination anomalies, cleft lip and palate, cerebral
Dominant inheritance carcinoma Medulloblastoma (PNET—desmoplastic) Sun exposure avoidance and at least and falx calcifications, macrocephaly with
syndrome annual dermatology examination frontal bossing, polydactyly, intellectual
Surgical management disability, lymphomesenteric or pleural
cysts, palmar and plantar pits, cardiac
fibromas, ovarian fibromas, ocular
abnormalities
Related gene: SUFU
PTEN158,459,531–540 Cowden High Breast Breast screening with annual Major and minor criteria (tumors,
Tumor suppressor syndrome/PTEN Thyroid (most often follicular) mammogram and breast MRI physical and dermatologic or
Dominant inheritance hamartoma tumor Endometrial Optional risk-reducing mastectomy mucocutaneous findings, developmental
syndrome Colorectal lesions (hamartomas, Thyroid examination and ultrasound disability)
ganglioneuromas, cancer) Endometrial screening with random Macrocephaly
Genitourinary tumors (renal cell biopsy and ultrasound Bannayan-Riley-Ruvalcaba syndrome
carcinoma) Optional hysterectomy PTEN-related Proteus syndrome
Colonoscopy Autism spectrum disorder
Renal imaging Lhermitte-Duclos disease
RAD51C and Moderate Ovarian Consider risk-reducing BSO at age Risk of autosomal recessive Fanconi
RAD51D43,84,114,541,542 Rare: High 50–55 yr or per family historyb anemia in biallelic RAD51C offspring of
Tumor suppressor (biallelic heterozygous carriers
RAD51C)
RB1543–551 Retinoblastoma High Retinoblastoma (trilateral disease = Ophthalmologic examination and Knudson’s two-hit hypothesis
Tumor suppressor (RB) co-occurrence of pineoblastoma) imaging; care by a multidisciplinary team Usually bilateral or multifocal disease
Dominant inheritance Bone or soft tissue sarcomas Examination schedule for carriers: Mosaic forms, therefore analysis of both
Melanoma Birth to 8 weeks: Non-sedated tumor tissue and blood is important
Others examination every 2 weeks RNA studies may also be recommended
8 weeks to 12 mo: EUA monthly Low-penetrance mutations identified
12–24 mo: EUA every 2 mo Parent-of-origin effect in some families
24–36 mo: EUA every 3 mo
36–48 mo: EUA every 4 mo
48–60 mo: EUA every 6 mo
5–7 yr: Examination under sedation every
6 mo
Surveillance for trilateral RB
Brain MRI at the time of RB diagnosis;
some centers recommend brain MRI every
6 mo until 5 yr old
Surveillance for second primary tumors
Education regarding second primary
tumor risks, and close attention to any
new signs or symptoms
Skin examination by a pediatrician during
well-child visits, to be continued annually
by the primary care physician or
dermatologist for melanoma from age 18
Some consider annual WBMR annually
after age 8, but no consensus
Radiation avoidance
RET8,332,334,552,553 Multiple High Medullary thyroid carcinoma (MTC) Prophylactic thyroidectomy (varies from Genotype-phenotype correlations
Oncogene endocrine (multifocal, bilateral) infancy to 5 yr of age or older depending MEN2A: MTC in early-adulthood
Dominant inheritance neoplasia type 2 and on specific RET pathogenic mutation; FMTC: MTC in middle age
Three subtypes: primary C-cell hyperplasia genotype-phenotype correlations) MEN2B: MTC in early-childhood mucosal
• MEN2A (Sipple PCC (bilateral) (MEN2A and MEN2B) Surgical management of identified neuromas (lips, tongue), dysmorphic
syndrome) Parathyroid adenoma or hyperplasia disease features (large lips, “marfanoid” body
• Familial (hyperparathyroidism, hypercalcemia, Serum calcitonin concentration habitus), gastrointestinal
medullary renal stones) (MEN2A) Consideration of eligibility for treatment ganglioneuromatosis
thyroid with kinase inhibitors RET gene also associated with
carcinoma Biochemical screening and imaging for Hirschsprung disease
(FMTC) pheochromocytoma and parathyroid
• MEN2B abnormalities

Continued
Genetic Factors: Hereditary Cancer Predisposition Syndromes  •  CHAPTER 13 191
Table 13.2  Cancer Susceptibility Genes, Syndromes, and Management Considerations—cont’d
Genes and Details Syndrome Penetrance Associated Neoplasms Management Considerations Other Notes
SDHB, SDHC, Hereditary High Paraganglioma (PGL)—sympathetic and Biochemical screening, physical Secretory and nonsecretory PGL
SDHD, SDHA, paraganglioma- (particularly parasympathetic examination, and imaging High blood pressure, headache, anxiety,
SDHAF2313,482,554–558 PCC syndrome SDHB and PCC Blood pressure at all medical visits profuse sweating, palpitations, pallor
Tumor suppressors SDHD) GISTs starting at age 6–8 SDHB: High risk of malignant
192 Part I: Science and Clinical Oncology

Dominant inheritance Renal clear cell carcinoma Plasma methoxytyramine starting at age transformation
Papillary thyroid carcinoma (unclear data) 6–8 yr then annually SDHD: Parent-of-origin effect—disease
Plasma free or 24-hour fractionated causing when paternally inherited
metanephrines starting at age 6–8 yr then SDHAF2: Possible parent-of-origin effect
annually (paternal inheritance)
Optional serum chromogranin starting at
6–8 yr then annually
Whole-body MRI skull base to pelvis at
6–8 yr then biennially
Optional: Neck MRI with or without
contrast agent starting at age 6–8 then
biennially
CBC with RBC indices start at age 6-8 then
annual
SMARCB1, SMARCA47 Rhabdoid tumor High Rhabdoid tumor Brain MRI every 3 mo to age 5 Ovarian and fallopian tumor removal to
Epigenetic regulator predisposition Small cell carcinoma of the ovary Abdomen: Consider whole-body MRI to be considered in carriers before puberty
Dominant inheritance syndrome hypercalcemic type (SCCOHT) age 5, ultrasound every 3 mo
Schwannomatosis
STK11 (LKB1)438,559–564 Peutz-Jeghers High Gastrointestinal lesions (PJS-type Colonoscopy Mucocutaneous hyperpigmentation of
Tumor suppressor syndrome (PJS) hamartomatous polyps; colorectal, Upper endoscopy with small bowel mouth, lips, nose, eyes, genitalia, fingers;
Dominant inheritance stomach, small bowel cancer) visualization may fade after puberty
Breast Breast screening with annual Females: Sex cord tumors with annular
Pancreas mammogram and breast MRI tubules (SCTAT); adenoma malignum of
Gynecologic (ovarian, cervix, uterus) Investigational-based pancreatic the cervix
Testes (Sertoli cell) screening
Lung Gynecologic examination
Testicular examination
TP53565–573 Li-Fraumeni High Soft tissue and bone sarcomas Breast screening with annual breast MRI Classic LFS criteria
Tumor suppressor syndrome (LFS) Breast and mammogram beginning in the 20s Chompret criteria
Dominant inheritance Brain and 30s, respectively Consider genetic testing in BRCA-
Adrenocortical carcinoma Optional risk-reducing mastectomy negative isolated early-onset breast
Leukemia Physical examination including cancer (age <30)
Others: Gastrointestinal, genitourinary, dermatologic and neurologic Breast cancers more likely to be estrogen,
lung, lymphomas, thyroid, neuroblastoma, examinations progesterone, HER2/neu receptor
skin Colonoscopy positive
Investigational-based whole-body MRI
Consideration of radiation avoidance, if
clinically appropriate
TSC1 and TSC2574–583 Tuberous sclerosis High Kidney: TSC specialist for management Skin (hypomelanotic macules, facial
Tumor suppressor complex (TSC) Renal cell carcinomas, angiomyolipomas Imaging angiofibromas, shagreen patches,
Dominant inheritance (benign and malignant), Dermatologic, dental, ophthalmologic cephalic plaques, ungual fibromas)
Epithelial cysts, examinations; EEG, echocardiogram, ECG Brain (cortical dysplasias, subependymal
oncocytoma (benign adenomatous Consideration of eligibility for treatment nodules and subependymal giant cell
hamartoma) with mTOR inhibitors astrocytomas, seizures, intellectual
Surgical management disability or developmental delay, autism
spectrum disorder, psychiatric illness)
Heart (rhabdomyomas, arrhythmias)
Lungs (lymphangioleiomyomatosis)
Genotype-phenotype correlations:
Higher risk of renal cell carcinoma in
TSC2
Possible association with NETs
VHL379,380,398,558,584–589 von Hippel-Lindau High Clear cell renal cell carcinoma Eye examination including retina starting Hemangioblastomas of the brain, spinal
Tumor suppressor disease PCC, paragangliomas at birth then annually cord, and retina; renal cysts; pancreatic
Dominant inheritance Pancreatic NETs Blood pressure at all medical visits cysts; endolymphatic sac tumors;
starting at age 2 epididymal and broad ligament cysts
Plasma free metanephrines or 24-hour Genotype-phenotype correlations: VHL
urine fractionated metanephrines starting type 1, type 2A, type 2B, type 2C with
at age 2 then annually different risks of PCC or renal cell
Audiogram starting at age 5 then carcinoma
biennially
MRI of brain with and without contrast
agent and MRI of spine with contrast
starting at age 8 then biennially
MRI of abdomen starting at age 10 then
annually (for RCC and pancreatic NET
screen)

Leukemia and lymphoma predisposition syndromes are not listed. Please refer to dedicated chapters.
The majority of childhood onset conditions are not listed. Please refer to Lindor NM, McMaster ML, Lindor CJ. Concise handbook of familial cancer susceptibility syndromes, 2nd ed. J Natl Cancer Inst Monogr. 2008;(38):1-93.
PMID:18559331
a
Earlier initiation of breast cancer surveillance may be warranted in the presence of a significant family history of breast cancer.
b
Earlier consideration of risk-reducing salpingo-oophorectomy may be warranted in the presence of a clear family history of ovarian cancer (>1 case).
AAP, American Academy of Pediatrics; ALL, acute lymphoblastic leukemia; AML, acute myeloblastic leukemia; BSO, bilateral salpingo-oophorectomy; CBC, complete blood count; CNS, central nervous system; ECG,
electroencephalogram; EEG, electroencephalogram; EUA, examination under anesthesia; FDG-PET, fluorodeoxyglucose–positron emission tomography; GI, gastrointestinal; GIST, gastrointestinal stromal tumor; HPV, human
papillomavirus; HSCT, hematopoietic stem cell transplantation; JMML, juvenile myelo monocytic leukemia; MDS, myelodysplasia; MPNST, malignant peripheral nerve sheath tumors; MRI, magnetic resonance imaging; NET,
neuroendocrine tumor; PARP, poly (ADP-ribose) polymerase; PCC, pheochromocytoma; PSA, prostate-specific antigen; RCC, renal cell carcinoma; TSH, thyroid-stimulating hormone; VCE, video capsule endoscopy; WBMR, whole
body MRI.
Genetic Factors: Hereditary Cancer Predisposition Syndromes  •  CHAPTER 13 193
194 Part I: Science and Clinical Oncology

clinical decision making and leads to improved health outcomes, has


Box 13.1.  ELEMENTS OF INFORMED CONSENT been clearly proven.18 In addition to the high- and moderate-penetrance
FOR GENETIC CANCER TESTING cancer genes, for nearly all the common malignancies, hundreds of
additional genetic loci have been identified, largely via genome-wide
1. What the test is intended to do—that is, determine whether a association studies, with each genetic variant, usually single-nucleotide
mutation can be detected in a specific cancer susceptibility gene polymorphisms (SNPs), being associated only a modest increased risk
2. What can be learned from both a positive and negative test result, of cancer (RR, ~1.1–1.5).19 Given the limited clinical validity and
including information on the magnitude of health risks associated utility of SNPs with such small effect sizes, clinical testing for individual
with a positive test result, as well as the risks that may remain even risk loci is currently performed, although research efforts to devise
after a negative test result polygenic risk models are underway20 and have already been incor-
3. The possibility that no additional risk information will be obtained porated into models of risk stratification for BRCA mutation–carrying
after testing or that the test will result in a finding of unknown males and females.21,22 In addition, these frequent but low-penetrance
significance (e.g., a polymorphism) that might require further variants identified through genome-wide association studies23 are
studies thought to explain a portion of the excess familiality of cancer. To
4. The options for approximation of risk without genetic testing—for account for the missing familiarity that remains, sequencing of protein
example, using empiric risk tables for breast cancer given differing coding regions of the genome or the entire genome itself has led to
family histories the discovery of novel cancer susceptibility genes.2,3 Together, the
5. The risk of passing a mutation on to children, including options for knowledge of rare and common variations has the potential to inform
assisted reproduction (e.g., preimplantation genetics, if discussion the clinician about the combined effect of genetic factors that lead
is appropriate) to cancer, extending beyond risks conferred by single genes. Ultimately,
6. The importance of notification of family members that they might to assess a person’s risk of cancer, one will need to combine genetic
share a hereditary risk for cancer, with every effort made to assist in factors within the context of environmental factors, which also may
contacting family members and providing them access to include exposures to cancer therapies.
counseling and testing
7. The medical options and limited proof of efficacy for surveillance and
cancer prevention for individuals with a positive test result, as well as
COMMON SYNDROMES OF CANCER
the accepted recommendations for cancer screening even if PREDISPOSITION
genetic testing results are negative The major syndromes of cancer predisposition that affect adults include
8. The technical accuracy of the test—that is, the sensitivity and breast, ovarian, colon, prostate, gastric, and pancreatic cancer, as well
specificity of the analytic methodology as a number of other less common but equally important cancer
9. The risks of psychologic distress and family disruption whether a predispositions. Some of these syndromes, including multiple endocrine
mutation is found or not found neoplasia type 1 (MEN1), retinoblastoma, melanoma, von Hippel-
10. The risk of employment and/or insurance discrimination after Lindau (VHL) disease, and familial pheochromocytoma and para-
disclosure of genetic test results and the level of confidentiality of ganglioma, are described elsewhere in this text. Comprehensive reviews
results compared with other medical tests and procedures of these cancer predispositions are offered elsewhere, including Li-
11. The risks that nonrelatedness of family members will be discovered Fraumeni syndrome,24 MEN1,25,26 retinoblastoma,27 melanoma,28,29
and how this information will be disclosed (or not disclosed) and VHL disease.30,31 Some syndromes such as neurofibromatosis also
12. The fees and costs of testing, including the laboratory test and the have cancer predispositions.32–34 A detailed outline of the elements of
associated consultation with the health professional who is setting up a comprehensive cancer genetic and genomics counseling
providing pretest education, results disclosure, and follow-up, and service has also been published.1,35
the costs of preventive procedures, which might not be covered by
third-party payers Breast and Ovarian Cancer Syndromes
Modified from Offit K. Clinical Cancer Genetics: Risk Counseling and Management.
New York: Wiley-Liss; 1998, with permission.
Clinical Features
Although only about 18,000 cases of breast cancer each year are
associated with an obvious hereditary predisposition, primary cancers
developed in more than 200,000 breast cancer survivors in the United
States as a result of a hereditary predisposition, and these survivors
result in an increased risk for a second neoplasm for millions of cancer remain at risk for secondary cancers.36,37 Genetic testing has emerged
survivors. as one of the most important indicators of risk factors, pointing to a
Inherited cancer predisposition can be considered as a spectrum, need for intensified screening for breast cancer.38 When detected at
arising from single or combined low-, moderate-, and high-risk genetic an early stage, more than 90% of breast cancers are curable. These
variants for which the timing of disease onset is likely modified by statistics underscore the rationale for the use of genetics in clinical
the type of genetic variant and its effect on normal cellular function oncology. We have previously reviewed in detail the management of
and the responses to environmental factors. In general, highly penetrant women and men who are at hereditary risk for breast cancer,39 and
hereditary cancer syndromes account for about 5% to 10% of most this review is summarized and updated here.
types of cancer and are caused by rare genetic variants. We are now From 1 in 150 to 1 in 800 individuals in the population carry a
also aware of dozens of “moderate-penetrance” genes that in general genetic susceptibility to breast cancer,40–42 and the prevalence is much
confer a modest degree of cancer risk, with a relative risk (RR) ranging higher in certain ethnic groups. Syndromes of breast cancer susceptibility
between 2 and 5 (Table 13.3).17 Although screening for such moderate- are linked to mutations of BRCA1 and BRCA2, in addition to a
penetrance genes was previously not routinely undertaken, with the smaller number of cases with germline mutations of ATM, PALB2,
availability of next-generation sequencing, screening for mutations in PTEN, p53, CHEK2, STK11, CDH1, and rarer syndromes (Tables
many genes simultaneously, often in multigene panels, has become 13.4 and 13.5).43 Cowden syndrome (CS) was initially described as a
readily available. The value of screening for moderate-penetrance genes dominant inheritance of multiple hamartomatous lesions, including
remains controversial because neither the clinical validity, the accuracy papillomas of the lips and mucous membranes and acral keratoses of
with which a genetic test predicts the development of cancer, nor the the skin.44 This disease was ultimately linked to germline mutations of
clinical utility, the degree to which the use of the genetic test informs PTEN and is discussed separately later. In persons with Li-Fraumeni
Table 13.3  Selected Moderate-Penetrance Genes and Associated Malignancies
Breast Ovary Colorectal Pancreas Melanoma Prostate Renal Other
APC*I1307K X
RR, 2.17
(95% CI, 1.6–2.9)a
ATM X X Autosomal recessive
RR, 2.8 (90% CI, 2.2–3.7) Ataxia telangiectasia
BARD1 (ID) (ID)
BRIP1 X Autosomal recessive
RR 11.2 (95% CI, 3.2–34.1) Fanconi anemia
in case control;
RR, 3.41 (95% CI, 2.1–5.5) in
segregation analysis
CHEK2 X (ID) X (X)
Truncating: 1100delC:
RR, 3.0 (90% CI 2.6–3.5); RR, 1.88 (95% CI, 1.3–2.7);
Missense (I157T): I157T:
RR, 1.58 (95% CI, 1.4–1.8) RR, 1.56 (95% CI, 1.3–1.8)
MAX X (Pheochromocytoma,
paraganglioma)
MITF X (X)
MRE11A (ID) (ID)
MUTYH X
(monoallelic) RR, 1.17 (95% CI, 1.0–1.3)
NBN (X) Autosomal recessive Nijmegen
c.657del5: breakage syndrome
RR, 2.7 (90% CI 1.9–3.7)
NF1 X High penetrance
Neurofibromatosis type 1
RAD50 (ID) (ID)
RAD51C, X RAD51C:
RAD51D RAD51C: RR, 5.2 (95% CI, Autosomal recessive Fanconi
1.1–24); RAD51D: RR, 12 Anemia
(95% CI, 1.5–90)
TMEM127 X (Pheochromocytoma)

Risk estimates listed assume no family history of the implicated cancer.


a
Risk estimate for APC*I1307K is based on Ashkenazi Jewish population.
(X): To date, limited evidence to support the association and the strength of the association is unclear.
(ID): Insufficient data. Occurrence documented, but association not clearly supported by evidence.
CI, Confidence interval; RR, average relative risk.
Data from Tung N, Domchek SM, Stadler Z, et al. Counselling framework for moderate-penetrance cancer-susceptibility mutations. Nat Rev Clin Oncol. 2016;13(9):581-8; Ma X, Zhang B, Zheng W. Genetic variants associated
with colorectal cancer risk: comprehensive research synopsis, meta-analysis, and epidemiological evidence. Gut. 2014;63(2):326-36; and Liang J, Lin C, HuF, et al. APC polymorphisms and the risk of colorectal neoplasia: a HuGE
review and meta-analysis. Am J Epidemiol. 2013;177(11):1169-79.
Genetic Factors: Hereditary Cancer Predisposition Syndromes  •  CHAPTER 13 195
196 Part I: Science and Clinical Oncology

Table 13.4  Genes Associated With Hereditary Breast Cancer (BC) Predisposition
Relative Risk BC Risk by
Gene Syndrome of BC Age 80 Yr Associated Cancers
HIGH PENETRANCE
BRCA136,87,590–593 HBOC ~15–30 70% Ovarian, other36,75,444
BRCA236,87,591–593 HBOC ~10–20 70%36,75,444 Ovarian, pancreatic, prostate, other
p53594–596 Li-Fraumeni syndrome 10017 50% by 60 yr597,598 Soft tissue sarcoma, osteosarcoma,
brain tumors, adrenocortical
carcinoma, leukemia, other
PTEN599–601 Cowden syndrome No reliable estimate17 70%–80%158,602 Thyroid (follicular and rarely
papillary) endometrial,
genitourinary, other
Bannayan-Riley-Ruvalcaba
syndrome
Proteus
Proteus-like syndrome
STK1147,289 Peutz-Jeghers syndrome No reliable estimate17 30% by age 6047 Small intestine, colorectal, uterine,
testicular and ovarian sex chord
tumors, other
CDH137,270,603 Hereditary diffuse gastric ~3.25 39% Lobular breast, diffuse gastric,
carcinoma other
LOWER OR MODERATE PENETRANCE
ATM (heterozygote)604–606 Ataxia-telangiectasia in ~3 3084 Undefined in heterozygotes
homozygotes
CHK2 (CHEK2)63,64,607,608 Li-Fraumeni variant 1.5-3 20%–30%84 Undefined

PALB2609 None known 5 ~4049,84 Undefined in heterozygotes

HBOC, Hereditary breast and ovarian cancer syndrome.

Table 13.5  Genes Associated With Hereditary Ovarian Cancer (OC) Predisposition
Relative Risk OC Risk by
Gene Syndrome of OC Age 80 Yr Associated Cancers
HIGH PENETRANCE
BRCA1 Hereditary breast ovarian cancer syndrome ~50 ~40%36,75,444 Breast, other
BRCA2 Hereditary breast ovarian cancer syndrome ~8 11%– Breast, pancreas, prostate, other
26%36,75,444
MLH1 Lynch syndrome ~4 ~20%83,610,611 Colon
MSH2 Uterine
MSH6 Stomach
PMS2 Small intestine
EPCAM Urinary tract
Pancreatic
Possible other sites
LOWER OR MODERATE PENETRANCE
RAD51C None ~5 ~6% Undefined84
Autosomal recessive Fanconi anemia
RAD51D None ~12 ~14% Undefined84
Autosomal recessive Fanconi anemia

syndrome, early-onset breast cancer occurs with soft tissue sarcomas, recently, mutations in PALB2, partner and localizer of BRCA2, have
osteosarcoma, leukemia (particularly hypodiploid acute lymphoblastic been implicated in both familial breast and pancreas cancer risk.48–51
leukemia), brain tumors, adrenal cortical tumors, and other cancers. Early linkage studies suggested that in about 50% of breast cancer
Rarely, atypical breast-ovarian kindred may be found to have a germline kindreds, the cancer was linked to BRCA1; in 30%, it was linked to
p53 mutation.45 Although rare, women with Peutz-Jeghers syndrome, BRCA2; and in the remainder, it was linked to other either identified
associated with germline mutations in the STK11 gene, are at increased or unidentified genes.52 In up to two-thirds of families with both male
risk for breast cancer as well as a variety of other malignancies.46,47 More and female breast cancer, the cancers were due to BRCA2, whereas
Genetic Factors: Hereditary Cancer Predisposition Syndromes  •  CHAPTER 13 197

more than 80% of families with both breast and ovarian cancer harbored As expected, mutations in BRCA1 and BRCA2 accounted for the bulk
mutations in BRCA1.53 BRCA1-linked breast cancers are associated of inherited risk, with mutations in BRIP1, RAD51C, RAD51D, and
with a basal-like subtype and higher mitotic indices.54 They are more the MMR genes accounting for 3% cumulatively. Compared with a
likely to be of higher grade, are more frequently estrogen and pro- population risk for ovarian cancer of approximately 1.5%, mutations
gesterone receptor negative,55,56 and demonstrate a basal-like pheno- in BRCA1 and BRCA2 are associated with a risk of 44% and 17%
type.57 They display a more “aggressive” phenotype, including a higher by age 80, respectively.75 For BRIP1, RAD51C, and RAD51D, the
proportion of cells in S phase, and other indices.54,56,58–60 cumulative lifetime risk by age 80 is 4% to 14%.84
Multiple moderate-penetrance breast cancer susceptibility genes The role of ascertainment and other possible biases in deriving
have also been identified (see Table 13.4).17,62–66 In Northern European cancer risk estimates, as well as risks for cancer of the prostate, colon,
families, specific mutations of CHEK2 are associated with familial pancreas, and other sites in BRCA mutation carriers has been reviewed.87
breast cancer, with pathogenic CHEK2 variants being present in ~1% In addition to Fanconi anemia, persons with compound BRCA2
of Caucasians of European descent.64 Although the common European mutations may experience childhood medulloblastomas.88 Heterozygous
mutation in CHEK2 is rare in persons in North America,67 analysis mutations in BRCA1 and BRCA2 harbored by pediatric cancer patients
of women for CHEK2 founder mutations stratified for family history have been detected through tumor normal sequencing; however, it is
of breast cancer demonstrated that carriers with a positive family not clear at this point if these are cancers associated with the familial
history had a greater than 25% lifetime risk for breast cancer.68 syndrome or simply true and unrelated, and this is an active area of
Important to note, degree of breast cancer risk in women with mutations research.89–91
in CHEK2 appears to be dependent on both the nature of the specific
mutation (truncating versus missense) and the strength of the family Genetics
breast cancer history. Carriers of mutations in ATM, another moderate- BRCA1 and BRCA2 are both important for DNA repair, in particular
penetrance breast cancer susceptibility gene, have an approximately homologous recombination, which enables repair of double-stranded
twofold elevated breast cancer risk.62,69,70 DNA breaks. BRCA1 is a large gene, spanning more than 100,000
Multiple, common, lower-penetrance genes are likely to account bases of genomic DNA with 22 coding and 2 noncoding exons.
for a significant component of currently unexplained familial breast BRCA2 is also large, consisting of 27 exons across 70 kb of genomic
cancer risk.71 A host of putative lower-penetrance gene mutations DNA. Both genes, by coincidence, have a large exon 11. An update
have been identified through whole-genome association studies,72,73 of reported mutations is accessible through the Internet at http://
although the clinical relevance of these associations remains unclear. research.nhgri.nih.gov/bic/.
In a large cohort of breast cancer patients referred for genetic testing, Most BRCA1 mutations cause premature truncation of the peptide
approximately 10% had a related germline mutation.74 by frameshift or nonsense sequence changes. Large germline rear-
Compared with the 10% breast cancer risk for women in the rangements also occur in BRCA1 and BRCA2.92,93 Five percent to
general population, estimates of the breast cancer risk that is conferred 10% of BRCA mutations that are missense are problematic because
by a common high-penetrance susceptibility gene ranged from 67% they are of unknown clinical significance. The proportion of these
to 69% by age 70 years based on epidemiologic analyses.40,41 This was variants that are of unknown significance was as high as 10% to 23%
confirmed in a prospective study that defined breast cancer risk by in some series, posing counseling challenges.94,95 The role of BRCA1
age 80 of 72% and 69% for BRCA1 and BRCA2 carriers, respectively.75 and BRCA2 in DNA damage response and homologous recombination
This study also reported a contralateral breast cancer risk, up to 20 is reviewed elsewhere.96
years after diagnosis, of 40% and 26% for BRCA1 and BRCA2, Founder BRCA mutations have been documented in genetically
respectively.75 In the setting of a mutation in PALB2, the breast cancer isolated populations. In North American families, the most common
risk to age 80 is approximately 45%, whereas for those with mutations founder mutations occur in persons of Ashkenazi Jewish origin. These
in ATM and CHEK2 it is less than 30%.17 mutations include a two-base-pair deletion in codon 23 of BRCA1,
Epithelial ovarian carcinoma can be delineated into six distinct termed 185delAG (c.68_69delAG); another mutation in BRCA1,
histologic subtypes—high-grade serous, carcinosarcoma, clear cell, 5382insC (c.5266dupC); and the 6174delT (c.5946delT) mutation
endometrioid (International Federation of Gynecology and Obstetrics in BRCA2.96–99 About 1 in 40 Ashkenazi Jews harbor one of the
[FIGO] grades 1–3), mucinous, and low-grade serous—with differing common BRCA1 or BRCA2 mutations,98,100,101 a relatively high carrier
manifestations, prognoses, and molecular and immunohistochemistry frequency for an inherited cancer predisposition syndrome. Other
profiles.76–78 The high-grade serous subtype comprises the majority of mutations in BRCA1 and BRCA2 occur in the Ashkenazim; 16 of
cases of ovarian cancer.77 Mutations in BRCA1 and BRCA2 show a 737 Ashkenazi Jews who were tested in a clinic-based ascertainment
strong association with the high-grade serous carcinoma of ovary and had a nonfounder mutation (2%).53 In another study of Ashkenazi
fallopian tube or peritoneal origin, such that germline testing for individuals with a personal history of breast or ovarian cancer who
BRCA1 and BRCA2 together can have up to a 25% detection rate in had previously been shown not to have a founder mutation, 3 of 70
this histologic subtype.79–82 Germline testing for BRCA1 and BRCA2 (4.3%) had a deleterious nonfounder mutation.102 This very low
is standard of care for all patients with a high-grade serous ovarian nonfounder BRCA rate in Ashkenazim was also confirmed again in
cancer diagnosis. Mutations in the mismatch repair (MMR) genes a 2017 study.103 Founder mutations in populations other than the
are also associated with ovarian cancer risk,83 usually endometrioid Ashkenazim have also been observed.104–106
histology; this is further discussed in the later section on Lynch
syndrome (LS). Similar to breast cancer, a number of more moderate- Other Genes
penetrance ovarian cancer susceptibility genes have also been identified. Protein truncating variants in PALB2, CHEK2, and ATM have been
BRIP1, RAD51C, and RAD51D are associated with approximately a associated with increased breast cancer risk as outlined earlier. The risk
5-fold to 12-fold risk of ovarian cancer84 (see Table 13.5). Earlier data associated varies per study and variant. A Finnish founder mutation in
suggested an association with BRIP1 and breast cancer, but a large PALB2 c.1592delT is associated with higher than population risk of
population-based study demonstrated that truncating mutations in breast cancer,107 but risk estimates are lower than those reported with
this gene were not associated with a substantial increase in breast other PALB2 mutations. Most of the data for CHEK2 and increased
cancer risk.85 breast cancer risk relates to the truncating c.1100delC variant found
In a study of close to 2000 women with ovarian cancer, 18% of in Northern Europeans.63,64,108 The missense variant CHEK2 I157T is
patients had a germline mutation in a gene associated with ovarian associated with a very modest increased risk (RR, 1.58; 95% confidence
cancer risk.86 The majority of patients in this study had high-grade interval [CI], 1.42–1.75),109 with some laboratories not reporting
serous histology (78%), but all histologic subtypes were represented. this variant as being pathogenic. The autosomal recessive neurologic
198 Part I: Science and Clinical Oncology

disorder ataxia telangiectasia is associated with compound heterozygote cancer, risk-reducing surgery is also recommended for carriers of the
mutations in ATM, a gene involved with DNA double-strand break more modest-risk mutations in genes BRIP1, RAD51C, and RAD51D.
repair and cell cycle control. Although this is a rare event, monoallelic However, given the later age of onset of ovarian cancer compared
ATM mutations are relatively common, occurring in approximately with BRCA1 and BRCA2, risk-reducing surgery may be delayed until
3% of Caucasians. Women with heterozygous ATM mutations have patients in this group are perimenopausal or postmenopausal.
a moderate, approximately double population risk of breast cancer as Combination oral contraceptives that contain estrogen and high-dose
noted earlier.62,110,111 BRIP1, BRCA1 interacting protein C-terminal progestin result in a time-dependent, protective effect against ovarian
helicase 1 gene, is part of the DNA repair machinery. A founder cancer in some but not all studies of BRCA mutation carriers.142–144
mutation, BRIP1, c.2040_2041insTT was associated with increased There remains the concern of a small increased risk of breast cancer
risk of ovarian cancer in the Icelandic population.112 Subsequently, due to oral contraceptives in this group, particularly in persons with
case-control series reported increased ovarian cancer risk for other BRCA1 mutations.145
truncating BRIP1 mutations.113 RAD51C and RAD51D are part of Germline BRCA1 and BRCA2 mutation–associated tumors have
the Fanconi-BRCA homologous recombination repair pathway and defective DNA repair mechanisms because of the loss of function of
have been associated with a modest increased risk of ovarian cancer.114 the remaining normal copy of the gene. This inability of the tumor
to repair its DNA effectively can be exploited by therapies that disrupt
Clinical Management alternative DNA repair pathways, such that the tumor cell accumulates
Three elements of breast surveillance that are recommended to women so much DNA damage that it can no longer survive.146,147 Inhibitors
with increased risk of breast cancer are self-examination, clinician of the base excision repair enzyme poly (ADP-ribose) polymerase
examination, and imaging. The evidence base underlying these recom- (PARP inhibitors) use this principle of synthetic lethality and are
mendations has been reviewed,39,115 and updated guidelines are FDA approved for women with recurrent ovarian cancer in a number
available.116 of clinical settings.148–150 Although benefit has been demonstrated in
Increasingly, breast cancer screening including mammography and all patients with ovarian cancer, it is substantially greater in patients
magnetic resonance imaging (MRI) together has been shown to have with a germline or somatic BRCA1/2 mutation.148,149 A recent random-
greatest sensitivity.117 For women who are at the highest hereditary ized trial has also demonstrated that patients with metastatic breast
risk for breast cancer, whose breasts are difficult to examine, or who cancer and germline BRCA1/2 mutations had significant progression-
have had biopsy results showing atypia, it might be appropriate to free survival benefit with olaparib.151
discuss the option of removing the healthy breasts as a preventive
measure (i.e., prophylactic mastectomy). Retrospective studies and Cowden Syndrome
reviews document the well-established risk-reducing role of surgery
in high-risk patients.118–120 Prospective and combined consortium Clinical Features
studies have also shown the efficacy of this approach.64,121,122 CS is an autosomal dominant disorder characterized by multiple
Because of their antiestrogen properties, tamoxifen, raloxifene, and hamartomas with a high risk of benign and malignant tumors of the
aromatase inhibitors have been shown to decrease breast cancer rates thyroid, breast, and endometrium. Consensus criteria for CS establish
in persons who are at increased risk.123–125 Two studies on the impact three diagnostic categories.152
of tamoxifen on subsequent breast cancer risk in BRCA1 and BRCA2 Pathognomonic criteria include adult Lhermitte-Duclos disease
mutation carriers have shown conflicting conclusions,126,127 although (LDD) (defined as presence of a cerebellar dysplastic gangliocytoma)153
only one of these studies was sufficiently powered to reach significant and mucocutaneous lesions, facial trichilemmomas, acral keratoses,
results. Tamoxifen was confirmed to decrease contralateral breast cancer papillomatous lesions, and mucosal lesions. Major criteria include breast
risk in a follow-up of one of these studies of BRCA mutation carriers.128 cancer, thyroid cancer (especially follicular histology), macrocephaly,154
Providers are encouraged to discuss breast cancer chemoprevention and endometrial carcinoma. Minor criteria include other thyroid
with women who are at increased risk of breast cancer.129 There are lesions (e.g., goiter), mental retardation, hamartomatous intestinal
no studies demonstrating a mortality benefit for this approach, and polyps, fibrocystic breast disease, lipomas, fibromas, and genitourinary
a careful discussion of potential benefit versus toxicity is required. tumors (e.g., uterine fibroids and renal cell carcinoma) or genitourinary
Small trials have demonstrated the ability of ultrasound with Doppler malformation. The operational diagnosis of CS is made if an individual
and CA125 to reveal early-stage ovarian cancers in BRCA mutation meets any one of the following criteria: pathognomonic mucocutaneous
carriers.130,131 However, the efficacy of this approach has not been proven lesions alone if there are six or more facial papules, of which three
in large, prospective studies,132 and the US Food and Drug Administra- or more must be trichilemmoma, or cutaneous facial papules and
tion (FDA) has recommended against screening for ovarian cancer oral mucosal papillomatosis; or oral mucosal papillomatosis and acral
in all women, including those at high risk.133 Therefore prophylactic keratoses; or six or more palmoplantar keratoses. Alternatively, the
removal of the ovaries and fallopian tubes is recommended to women individual may fulfill two major criteria, but one must include either
with strong family histories of ovarian cancer, in families linked to macrocephaly or LDD. Alternatively, the individual may have one
BRCA1 or BRCA2, or to women who are considering hysterectomy major and three minor criteria or four minor criteria.
in the setting of a germline mutation associated with LS. Studies The palmar and plantar hyperkeratotic pits usually become evident
examining specimens from prophylactic salpingo-oophorectomy in later in childhood. Subcutaneous lipomas and cutaneous hemangiomas
BRCA1 and BRCA2 mutation carriers have implicated the fimbriated are seen in persons with CS with low frequency.155 An increased
part of the fallopian tube as harboring precursor lesions to high-grade risk of early-onset male breast cancer has been noted in mutation
serous epithelial ovarian cancer.84,134–136 Thus prophylactic salpingo- carriers.156
oophorectomy is recommended because such surgeries not only decrease Cumulative lifetime risks for female breast cancer are 81% to
the incidence of subsequent breast and ovarian cancer but also reveal 85.2%157,158; for LDD, 32% (CI, 19%–49%)157; for thyroid cancer,
occult early-stage pelvic neoplasms,136–138 decreasing mortality.139 These 21%–35.2%,157,158; for endometrial cancer, 19%–28.2%157,158; and
studies have also confirmed that high-grade serous carcinoma can still for renal cancer, 15%–33.6% (CI, 6%–32%).157,158 The lifetime risks
occur after prophylactic oophorectomy,140,141 albeit at much reduced for colorectal cancer (CRC) are 9% to 16%,157,158 and the risk for
rates. For women with high-penetrance risk genes, risk-reducing melanoma is 6%.158
prophylactic oophorectomy is recommended when childbearing is
complete or at approximately age 40.11 Given the lack of an adequate Genetics
screening modality, difficulty in diagnosis, and resultant late stage The CS-linked PTEN was mapped to 10q22-23.159 Bannayan-Riley-
at presentation and the associated mortality with invasive ovarian Ruvalcaba syndrome, characterized by macrocephaly, intestinal polyps,
Genetic Factors: Hereditary Cancer Predisposition Syndromes  •  CHAPTER 13 199

lipomas, and pigmented penile macules, is also caused by germline age of diagnosis of 44 years in the proband and 61 years in mutation
mutations in PTEN.160 carrier relatives.168–171 A 40% to 60% endometrial cancer risk by age
PTEN acts as a tumor suppressor by mediating cell cycle arrest, 70 years has been reported,172–175 with a median age at onset from the
apoptosis, or both.161 Full sequencing and deletion/duplication analysis late 40s to early 50s,176 as compared with a 3% risk for endometrial
are available clinically, and promoter analysis is available on a research cancer in the general population. LS also confers a high lifetime risk
basis. Heterozygous germline mutations in PTEN cause most cases for ovarian cancer (12%–15%).177 Five additional tumor sites dem-
of CS. Nonsense, missense, and frameshift mutations that are predicted onstrated increased observed/expected (O/E) ratios in LS kindreds:
to disrupt normal PTEN function have been identified in some cancers of the stomach (O/E = 4.1), small intestine (O/E = 25), ureter
families,159 including mutations that disrupt the protein tyrosine/ (O/E = 22), and kidney (O/E = 3.2).178 More recently, adrenocortical
dual-specificity phosphatase domain. Initially it was shown that PTEN neoplasm have also been implicated in LS and, although still contro-
mutations can be detected in about 80% of patients with CS.160 More versial, some studies also have demonstrated a modest increased risk
recently, the mutation detection rate in persons meeting CS criteria of prostate and breast cancer in patients with LS.179–181
was found to be 34%, raising the possibility that current aspects of At a 1991 meeting in Amsterdam, the International Collabora-
testing criteria are too broad.162 tive Group on LS defined the syndrome as (1) histologically verified
CRC in three or more relatives, including a first-degree relative of
Risk Management Recommendations the other two; (2) CRC involving at least two generations; and (3)
Persons with known germline PTEN mutations should undergo one or more CRCs diagnosed before age 50 years. The subsequent
appropriate cancer screening.116,163 In view of recent work outlining “Amsterdam II criteria” for LS were redefined to include extracolonic
cancer risks in persons with CS, some investigators have suggested LS-associated cancers.182 In 1996, the “Bethesda criteria” delineated
that National Comprehensive Cancer Network (NCCN) management individuals at risk for LS for whom molecular genetic analysis may
guidelines be modified to include renal cancer screening using biannual be considered.183 A revised set of Bethesda Guidelines was developed
renal imaging from age 40 years or 5 years earlier than the earliest to identify subjects who are at high risk of having a germline MMR
kidney cancer diagnosis in the family, and endometrial sampling in gene mutation.184 Multivariate logistic regression risk models using
adulthood or 5 years earlier than the earliest endometrial cancer personal and family medical histories estimate the probability of
diagnosis in the family.158 Female patients with CS should have breast carrying an LS mutation.185–187 In the most common LS-associated
self-examination training and education starting at age 18 years. cancers, including CRC and endometrial cancers, the need for tumor
Beginning at age 25 years, clinical breast examinations should be screening for LS has traditionally been based on the age at cancer
performed every 6 to 12 months, and annual mammography and diagnosis or on the strength of the personal and family history, as
MRI screening should start at age 30 years or 5 years before the earliest outlined in the Revised Bethesda Guidelines.188 However, per the most
age of breast cancer diagnosis in the family.158 Men should perform recent guidelines from EGAPP (Evaluation of Genomic Applications
monthly breast self-examination. Female patients should receive in Practice and Prevention Working Group) and NCCN, screening
endometrial cancer screening beginning around age 30 years or 5 of all colorectal tumors (or at least all colorectal tumors in patients
years before the earliest age of endometrial cancer diagnosis in the younger than 70 years at diagnosis or those older than 70 years who
family.158 Persons with CS should undergo biannual colonoscopy from meet Revised Bethesda Guidelines) and endometrial tumors via MSI
age 40 years.158 Both men and women should have a comprehensive or MMR protein immunohistochemical staining analysis is now
annual physical examination starting at diagnosis with screening for recommended.189,190
skin and thyroid lesions, including a baseline and annual thyroid
ultrasound and dermatologic examination.158 Finally, preimplantation Genetics
genetic testing for CS can be performed if a mutation is described in About 45% to 70% of LS families harbor mutations in one of the
a parent. following four genes: MSH2, MLH1, MSH6, and PMS2.191–195 In
addition, germline deletions of the 3′ region of the EPCAM gene,
Common Colon Cancer Predisposition Syndromes just upstream of MSH2, also result in LS, through silencing of the
MSH2 gene.196 Mutations of MSH2 and MLH1 were far more
High-penetrance cancer susceptibility syndromes account for about frequent than the others, accounting for about 30% each of families
5% of CRC cases. The most common syndrome is LS, with familial meeting Amsterdam criteria for LS. More than 75% of mutations in
adenomatous polyposis (FAP) constituting a rarer familial syndrome. MSH2 and MLH1 were inactivating insertions, deletions, alterations
Genetic epidemiologic analyses suggest a common susceptibility allele in premessenger RNA splicing signals, and nonsense mutations. Of
for both colon cancer and adenomatous polyps that accounts for at 120 mutations surveyed, 23% were missense mutations.197 Muta-
least 15% and possibly half of cases.164,165 tions of MSH6 less frequently result in the “replication error repair”
phenotype but account for a significant number of familial colon cancer
Lynch Syndrome families.198 The replication error repair phenotype is commonly detected
A constellation of colon and endometrial cancers became known as as MSI with use of polymerase chain reaction screening of tumors
Lynch syndrome, formerly referred to as hereditary nonpolyposis colon with microsatellite markers. The MSI phenotype is present in about
cancer (HNPCC).166 LS is an autosomal dominant syndrome caused 80% of LS-associated colon cancers199 and in about 15% of sporadic
by mutations in one of four MMR genes (MLH1, MSh2, MSH6, colon tumors,200 as well as in other tumors associated with LS (e.g.,
PMS2), or a gene located nearby (EPCAM), with resultant errors in uterine and gastric cancers). MSI results in a genome-wide increased
DNA replication yielding a microsatellite instability (MSI) phenotype. mutation rate, causing mutations in oncogenes, tumor suppressors, and
LS accounts for 3% of all CRCs.167 microsatellite regions.188
For patients with an abnormal screening test result (MSI-high or
Clinical features MMR-deficient tumor), various algorithms have been developed to
LS-associated CRC is characterized by an accelerated progression of help guide subsequent evaluations, which may include germline genetic
the adenoma-to-carcinoma sequence, with a predominance of right-sided testing or further tumor analyses, such as MLH1 promoter hyper-
colorectal tumors with pathology demonstrating poorly differentiated methylation and/or BRAF*V600E somatic mutation in MLH1/PMS2
adenocarcinoma with mucinous and signet ring cell features, a Crohnlike protein deficient cases.189,190 In older patients with CRC, hypermeth-
reaction, and tumor-infiltrating lymphocytes, in addition to presentation ylation of the MLH1 promoter, or presence of a BRAF*V600E somatic
with metachronous or synchronous colorectal tumors. Patients with mutation, may account for the lack of MLH1 protein expression.201
LS have a 50% to 80% lifetime risk of colon cancer, with a median This epigenetic (nonhereditary) mechanism of MLH1 promoter
200 Part I: Science and Clinical Oncology

hypermethylation is responsible for most of the remaining patients and other hepatopancreatic tumors.224 Gastric fundic gland polyps
whose tumors are characterized by defective DNA MMR.202 occur and are often dysplastic,225 although an increased risk of gastric
Biallelic mutations in the same MMR genes result in constitutional cancer has yet to be definitely demonstrated in Western populations.226
MMR deficiency, which classically manifests as childhood-onset cancers, Duodenal polyps are also prevalent, with a lifetime risk of duodenal
in particular hematologic malignancies, brain tumors, and early-onset cancer, particularly ampullary cancer, of 5% to 12%.227,228 Other
CRCs and café au lait macules similar to those seen in neurofibromatosis FAP-associated cancers are listed in Table 13.2. Benign extraintestinal
type 1,203,204 although a milder phenotype can exist in persons with manifestations of FAP include desmoid tumors, osteomas of the jaw
biallelic mutations in PMS2 as evidenced by reports of first cancer and dental anomalies, congenital hypertrophy of the retinal pigment
diagnoses in mutation carriers in the third and fourth decades.205,206 epithelium (CHRPE), lipomas, fibromas, sebaceous and epidermoid
cysts, and nasopharyngeal angiofibromas.228
Clinical management Genetics.  FAP is an autosomal dominant syndrome that occurs
For probands diagnosed with LS, observational data have shown that in 1 of every 7000 to 38,000 individuals and arises from germline
surveillance colonoscopy lowers CRC incidence and mortality by more mutations in the APC gene.228–230 Twenty-five percent of mutations
than 50%207 and improves patient survival.208 Persons with LS are are de novo,231 whereas a small fraction of cases result from somatic
advised to undergo colonoscopic surveillance every 1 to 2 years, mosaicism.232 Genotype-phenotype correlations have been described,233
preferably annually, starting at age 20 to 25 years. Some guidelines particularly for CHRPE and the number of polyps observed. An
suggest that for MSH6 mutation carriers, screening colonoscopy may attenuated form of FAP is associated with mutations at the extreme
be delayed to the age 30 years.209,210 A baseline upper endoscopy 5′ and 3′ ends of the gene.234,235 Up to 30% of patients with multiple
should be performed, but the optimal subsequent screening interval adenomas (15–100 adenomas) who test negative for APC mutations
has yet to be established. Prophylactic subtotal colectomy sparing the carry biallelic mutations in MUTYH,236 discussed in detail later in
rectum should be considered after the first CRC diagnosis, given the this chapter.
high rate of metachronous CRCs.211 No benefit has as yet been Clinical management. FAP may manifest as childhood malig-
demonstrated with other screening strategies directed toward the nancy, and thus the diagnosis is important as early as possible in
LS-associated malignancies.190 Endometrial cancer screening entails a proband’s lifetime, even prenatally via preimplantation genetic
endometrial biopsy at age 30 to 35 years. Annual urinalysis with testing if in vitro fertilization was used.237 Children are at risk for
cytologic evaluation or urinalysis for microhematuria may also be hepatoblastoma for the first 5 years of life and are screened accord-
considered, especially in MSH2 carriers, in whom urothelial risk is ingly. Annual flexible sigmoidoscopy for CRC screening begins at
highest, but minimal data support the efficacy of this screening test age 10 to 12 years. Once polyps are identified, annual colonoscopic
in early urothelial tumor detection. A retrospective study of prophylactic surveillance is recommended. Prophylactic proctocolectomy, usually
bilateral oophorectomy and hysterectomy showed protection from in the late teens, is the treatment of choice.190 Sulindac, a nonsteroidal
uterine and endometrial cancer,212 although the estimates were antiinflammatory drug, and selective cyclooxygenase-2 inhibitors
influenced by a retrospective study design.213 Nonetheless, a combination reduce adenoma size and number and may be considered for che-
of risk-reducing bilateral oophorectomy and hysterectomy is a reasonable moprevention to delay but not preclude surgical intervention.238–240
option after childbearing or at the time of colorectal resection for a Patients require lifelong surveillance for extracolonic tumors, including
CRC occurring in women with LS. tumors of the upper gastrointestinal tract and the ileal pouch (if
Identification of LS at the time of CRC diagnosis can affect clinical proctocolectomy is performed).241 The burden and histologic features
management. Subtotal (versus segmental) colectomy can reduce the of duodenal polyps determines the need for endoscopic versus surgical
increased risk of metachronous CRC in persons with LS by 31% for treatment.242 Additional screening measures are directed toward the
every additional 10 cm of bowel removed.214 Tumors that demonstrate FAP-associated cancers listed in Table 13.2.190 Unaffected individu-
the MSI-H phenotype may not benefit from 5-fluorouracil–based als with wild-type APC who have an affected family member with
chemotherapy and have improved stage-independent survival compared a known mutation are screened in the same way as the general
with proficient-MMR CRC.215–218 population.243
More recently, presence of MMR deficiency, a nearly universal
finding in LS-associated tumors, predicted for response to immune Attenuated familial adenomatous polyposis
checkpoint blockade. Specifically, in patients with advanced MMR- Clinical features. Attenuated familial adenomatous polyposis
deficient CRC, a response rate of 62% was seen, as compared with (AFAP) is an FAP variant characterized by oligopolyposis (<100 colonic
none in MMR-proficient tumors, to pembrolizumab, an anti– adenomas) and a CRC onset 10 to 20 years later than in patients
programmed death 1 immune checkpoint inhibitor.219 In further with FAP, although the precise lifetime risk of CRC is not well
evaluation of immune checkpoint blockade across 12 different defined.244 AFAP may display malignant and benign manifestations
MMR-deficient malignancies, a comparable high response rate was similar to those of FAP.245
observed, including response in LS patients.220 Checkpoint blockade Genetics.  Like FAP, AFAP is an autosomal dominant cancer
has also been shown to be of benefit for children with biallelic MMR syndrome caused by germline mutations in the APC gene, with defects
deficiency.221 Future studies are evaluating the role of immunotherapy that tend to localize in the 3′ or 5′ regions of the gene.
in patients with early-stage MMR-deficient tumors. Clinical management. Right-sided colon lesions are frequent,
making colonoscopy the preferred CRC screening modality. Colonoscopy
Polyposis Syndromes begins in the late teens on a 1- to 2-year basis, with surgical intervention
Familial adenomatous polyposis considered with advanced polyp number, size, or histology.190 Surgical
Clinical features.  FAP (adenomatous polyposis coli) manifests with approach (e.g., colectomy versus proctocolectomy) is determined by
hundreds to thousands of adenomatous polyps at a young age. The the specific APC mutation, clinical presentation, patient preference,
hallmark of FAP is the development of more than 100 adenomatous and postulated adherence to postoperative screening.242 Other screening
colonic polyps in the teenage years with a resultant risk of CRC of measures in AFAP are directed toward the associated malignancies
approximately 90% by age 45 years.222,223 Colon cancer is therefore listed in Table 13.2.190
inevitable if the colon is not removed; cancer occurs at an average age
of 39 years. Flexible sigmoidoscopy at an early age establishes the MUTYH-associated polyposis
diagnosis, and prophylactic colectomy is performed in the teen years. Clinical features.  Some AFAP probands have MUTYH-associated
Patients remain at risk for primary adenomas and carcinomas of the polyposis (MAP) on the basis of biallelic germline mutations in the
duodenum and rectum, as well as thyroid carcinoma, hepatoblastoma, base excision repair gene MUTYH.236,246,247 MAP is an oligopolyposis
Genetic Factors: Hereditary Cancer Predisposition Syndromes  •  CHAPTER 13 201

with a CRC risk of at least 50% by age 48 years.232,248,249 Colonic Hereditary Diffuse Gastric Cancer
polyps may be adenomatous, sessile-serrated, and/or hyperplastic.
The full spectrum of disease is still being defined, and extracolonic Clinical Features
manifestations, benign and malignant, may resemble FAP to LS.250 Hereditary diffuse gastric cancer (HDGC) is characterized by autosomal
A search for MUTYH mutations should be pursued in patients with dominant susceptibility to diffuse gastric cancer (DGC) and lobular
polyposis who have tested negative for APC mutations.236 In this breast cancer (LBC). Penetrance for small foci of in situ or invasive
scenario, the detection rate for biallelic MUTYH mutations may be cancer is virtually complete; however, the lifetime risk of clinically
17% to 27%.251 significant gastric cancer is estimated to be 80%.267 The ages of diagnosis
Genetics.  MUTYH mutations have a frequency of 1% to 2% in of DGC in CDH1 mutation carriers ranges from 14 to 85 years of
the general population. Conflicting data exist as to whether monoal- age,37 with an average of 38 years.268 Unfortunately, endoscopic surveil-
lelic MUTYH mutation confers increased risk of CRC.252,253 MAP lance for DGC is inadequate, and therefore germline CDH1 mutation
is considered an autosomal recessive syndrome; parents of affected carriers are advised to undergo prophylactic total gastrectomies to
homozygote or compound heterozygote mutation carriers have been remove their risk of DGC. Female carriers also have a 60% lifetime
reported to be unaffected.254 However, both monoallelic and biallelic risk of LBC.267
germline mutations in MUTYH have been associated with multiple
adenomas.247,254 Biallelic carriers have a higher frequency of CRC than Genetics
do monoallelic patients.255 Patients with multiple adenomas (more HDGC was initially found to be due to germline mutations in CDH1
than 15) without APC mutations are more likely to have biallelic in a large Maori kindred, with multiple family members affected with
MUTYH mutations than are control subjects.247,256 Combining APC DGC.269 Since then, 30% and 50% of HDGC families around the
and MUTYH sequencing increases the yield of diagnosis from 34.4% world and in North America have been found to carry pathogenic
to 41.3% in polyposis CRC.257 The mutations Y165C and G382D germline variants in CDH1, respectively.270–272 These differences likely
are recurring in Caucasian Europeans, making up 86% of biallelic relate to overall increases in gastric cancer in the lower-frequency
mutations in three series.247,254,255 In 358 early-onset polyposis cases areas. Pathology is characterized by pagetoid spread of signet ring cells
that were unselected for APC mutation status, two cases carried bial- beneath a normal mucosa.273 The International Gastric Cancer Linkage
lelic MUTYH mutations, and eight MUTYH mutation heterozygotes Consortium has updated testing criteria for HDGC267 to include (1)
were identified. No MUTYH mutations were detected in 354 control two gastric cancer cases in a family, with one confirmed DGC diagnosed
subjects, leading the authors to conclude that biallelic MUTYH muta- in a person younger than 50 years, (2) three confirmed DGC cases
tions might account for up to 3% of early-onset CRC. Patients in in first- or second-degree relatives independent of age, (3) a case of
this series did not exhibit profuse polyposis, consistent with previous DGC diagnosed before age 40 years, or (4) a personal or family history
reports, and distally located tumors were prevalent.258,259 of DGC and LBC, with one diagnosed before age 50 years. Because
Management.  CRC screening in biallelic mutation carriers includes of the early age at onset, similar to that of FAP, and the estimated
colonoscopy beginning at age 25 to 30 years and repeated every 3 to 10% 5-year survival rate after DGC diagnosis, persons as young as
5 years until polyps are detected, at which point the surveillance 13 years and able to give informed consent or assent should be
interval is decreased to every 1 to 2 years. Prophylactic colectomy is considered for genetic counseling.
undertaken depending on patient age, disease location, and polyp
burden. Other screening measures in persons with MAP are directed Clinical Management
toward the associated malignancies listed in Table 13.2.190 Surgical CDH1 mutation carriers should be referred to a multidisciplinary
options for MUTYH mutation carriers depend on polyp distribution team, including a geneticist, a gastroenterologist, a surgeon, and a
and burden and may include ileorectal anastomosis for younger patients nutritionist experienced in HDGC for discussion and counseling
with few rectal adenomas and a milder family history or with AFAP regarding the recommendation for total prophylactic gastrectomy. If
or total proctocolectomy with the creation of an ileal pouch and anal total prophylactic gastrectomy is refused or delayed, surveillance
anastomosis for more diffuse polyposis. gastroscopies should be undertaken on an annual basis, preferably
in a center experienced with HDGC, where there should be sufficient
Other hereditary predisposition to colon cancer time for inspection, and a minimum of 30 biopsy specimens should
With use of a combination of whole-exome sequencing and linkage be taken from the five gastric zones: the antrum, transitional zone,
analysis in individuals with colorectal adenomas without mutations body, fundus, and cardia.267 Gastrectomy has a significant effect on
in known polyposis genes, germline mutations in DNA polymerase quality of life and should be undertaken only after extensive genetic
ε (POLE) and δ (POLD1) have been identified.260 These genetic changes counseling. The long-term morbidity that can result from gastrectomy
are located in the proofreading (exonuclease) domain of the POLE includes weight loss, lactose intolerance, fat malabsorption and
and POLD1 genes and result in deficient proofreading repair during steatorrhea, dumping syndrome, bacterial overgrowth, postprandial
DNA replication. In addition to multiple adenomas and early-onset fullness, and vitamin deficiencies.274 Female carriers should be
CRC, the phenotypic features probably also include brain tumors referred to a high-risk breast clinic and advised regarding monthly
and, in POLD1 mutation carriers, endometrial cancer.261 breast self-examinations starting at age 35 years, an annual
Hereditary mixed polyposis syndrome is characterized by serrated mammogram and breast MRI, and a biannual clinical breast
adenomas, juvenile polyps, adenomas, and CRC, and maps to chromo- examination. It is currently not well established whether cancer
some 15q13-q14. Recently this syndrome has been associated with develops at any other sites with increased frequency with CDH1
mutations in BMPR1A in a family from Singapore, and in GREM1 mutations.
in an Ashkenazi Jewish family.262,263
Low-penetrance colon cancer susceptibility alleles include the APC Pancreatic Adenocarcinoma
I1307K polymorphism, CHEK2 gene mutations, and monoallelic Predisposition Syndromes
MUTYH (see Table 13.3), each associated with an approximate twofold
increased risk of CRC.264,265 Homozygous mutations in BUB1B Clinical Features
predispose a healthy adult to gastrointestinal polyps and CRC. Approximately 53,000 new cases of pancreatic adenocarcinoma (PAC)
Identification of BUB1B variants requires karyotype analysis of dividing are expected to be diagnosed in the United States in 2017.275 Familial
cells.266 Peutz-Jeghers syndrome (STK11) and juvenile polyposis clustering of PAC was demonstrated in 5% to 10% of these cases,
(SMAD4, BMPR1A) are associated with hamartomatous polyposis as whereas 2% could be attributed to mutations in a highly penetrant
well as an increased CRC risk. mendelian susceptibility gene.276
202 Part I: Science and Clinical Oncology

occur in 14%,298 suggesting that pelvic ultrasound examination may


Genetics be indicated as a part of the evaluation of women with CNC.303
Several hereditary syndromes predispose to PAC (see Table 13.2). The Diagnostic criteria for CNC297 require two or more of the manifesta-
hereditary breast and ovarian cancer susceptibility gene BRCA2 has tions or one major manifestation and one of the minor criteria. Major
been associated with sporadic and familial PAC; 7% of patients with criteria are skin pigmentary abnormalities (multiple lentigines of the
sporadic PAC were identified with mutations in BRCA2.277 A BRCA2 face, blue nevus, or epithelioid blue nevus); myxoma (cutaneous or
gene mutation has been identified in up to 19% of cases with familial mucosal myxomatosis); cardiac myxoma; endocrine tumors or overactiv-
PAC,278,279 and families carrying a BRCA2 gene mutation have a 3.5-fold ity (primary pigmented nodular adrenocortical disease, a micronodular
to 7-fold increased risk of PAC.280,281 BRCA1 gene mutations likely form of adrenal hyperplasia, growth hormone–producing pituitary
also predispose to PAC.282–284 A recent study demonstrated an increased adenoma, large-cell calcifying Sertoli cell tumor, or thyroid adenoma
prevalence of mutations in BRCA2 and BRCA1 in a cohort of Ashkenazi or carcinoma); psammomatous melanotic schwannoma; thyroid
origin with familial PAC.284 In persons with Fanconi anemia, mutations carcinoma or multiple thyroid nodules on ultrasound in a young
in multiple causative genes including PALB2, which complexes with patient; multiple breast ductal adenomas; and osteochondromyxoma.
BRCA2 to become part of the FA-DNA repair pathway, predispose To make the diagnosis, an individual must have two of the components
to PAC.51,276,285–287 listed or one of the manifestations in addition to an affected first-degree
FAP confers an RR of 4.5 for PAC,238 whereas LS probands have relative or have an inactivating mutation of the PRKAR1A gene. A
an approximately ninefold increased risk of PAC.288 Persons with malignant neoplasm is a relatively uncommon finding in affected
Peutz-Jeghers syndrome have an RR of 132 for PAC.289 Familial atypical patients.
multiple mole melanoma syndrome, associated with germline mutations
in CDKN2A/p16, has an RR of 13 to 22 for PAC.290 Hereditary Genetics
pancreatitis (PRSS1 or SPINK1) confers a 53- to 87-fold increased Mutations in PRKAR1A (17q23-q24) are identified in about 40% of
risk for PAC that is compounded by smoking.291,292 persons with CNC.304 PRKAR1A codes for the RI-α subunit of PKA,
a critical cellular component of a number of cyclic nucleotide–dependent
Clinical Management signaling pathways.305 PRKAR1-α frameshift mutations cause haplo-
Screening modalities for PAC include endoscopic ultrasound, magnetic insufficiency of R1-α and manifest as CNC. As predicted by the
resonance cholangiopancreatography (MRCP), computed tomography Knudson “two-hit” hypothesis, loss of heterozygosity of the normal
(CT), and serial serologic monitoring for tumor markers. Screening allele supports the model that R1-α may have tumor suppressor function
studies to date have not demonstrated a decrease in mortality. However, in the target tissues. The most common mutation in patients with
a European study of a large cohort of CDKN2A mutation carriers CNC, a two–base-pair deletion (TGdel) in exon 5 of PRKAR1, has
suggested that surveillance with annual MRI, MRCP, and/or endoscopic also been found de novo in several kindreds, suggestive of a mutational
ultrasound of the pancreas was relatively successful in detecting most hot spot in the gene.306 About 17% of diagnosed individuals harbor
PACs at a resectable stage.293 Participation in clinical trials assessing de novo mutations.
the efficacy of PAC screening in high-risk individuals is encouraged.
Clinical Management
Carney Complex Recommendations include annual echocardiography, annual measure-
ment of urinary free cortisol, testicular sonogram in males at their
Clinical Features initial visit, thyroid ultrasound at the initial visit and as needed
Carney complex (CNC) is a multiple neoplasia syndrome that is thereafter, pelvic ultrasound examination in female patients at their
referred to by acronyms such as NAME294 and LAMB syndrome initial visit, breast imaging, spine MRI, pituitary MRI, and adrenal
in the medical genetics literature.163,295 As described by Carney in CT scan.306 Children should undergo echocardiography during the
1985,163 the complex is characterized by myxomas, skin pigment first 6 months of life and annually thereafter to detect potentially
abnormalities, endocrine tumors, and schwannomas. The median lethal myxomas. Children with large-cell calcifying Sertoli cell tumor
age at diagnosis is 20 years, with spotty skin pigmentation and heart may require monitoring of growth rate and pubertal status. Bone age
myxomas being the most common initial clinical manifestations.296 determination and further laboratory evaluation might be necessary
The skin abnormalities involve the lips, the conjunctiva and inner or if gynecomastia is present.297
outer canthi, and the mucosa of the vagina or penis. Atrial myxomas
are by far of greatest clinical concern, because they usually result in a Hereditary Paraganglioma-Pheochromocytoma
decreased life span and account for the major causes of mortality in Syndromes
affected individuals; cardiac myxoma can cause stroke and death.297
Cardiac myxomas occur in 32% of mutation carriers.298 Ductal breast Clinical Features
adenoma has been described as part of CNC, as well as myxoid Paragangliomas are tumors of the paraganglia that are derived from
fibroadenomas and other findings on breast imaging.299 In one study neuroectodermal and mesodermal origins in the parasympathetic and
of 338 persons diagnosed with CNC, 34 of 194 women (17.5%) had sympathetic axis and thus can exist from the base of the skull to the
breast myxomas, often bilateral.297 Large-cell calcifying Sertoli cell pelvis. These tumors are called chemodectomas in the head and neck
tumor, seen in 41% of male mutation carriers,298 detected as early as and are derived from nonchromaffin chemoreceptors in the carotid,
2 years of age, has been associated with infertility due to hormonal aortic, jugular, or vagal bodies. These tumors are usually benign, and
imbalance.300 also can be symptomatic because of size and compression effects of
Primary pigmented nodular adrenocortical disease occurs most surrounding local structures. Of 30 cases reviewed in one series, about
frequently in association with CNC in 60% to 95% of patients,298,301 half were bilateral, with a family history of chemodectoma in about
and 14% have Cushing syndrome.301 Thus the diagnosis of primary one-third of these cases.307,308 Most of the familial cases presented
pigmented nodular adrenocortical disease should prompt screening with multiple tumors. The remarkable feature about hereditary
for CNC. Other organs that are involved in CNC are the thyroid chemodectomas is the evidence of imprinting; chemodectomas develop
gland and ovaries. Thyroid gland tumors occur in 25% and include in children of affected fathers, but not in the children of affected
follicular hyperplasia and follicular adenoma; thyroid cancer, follicular mothers.309 Pheochromocytomas are paragangliomas that are located
and papillary carcinoma, occurs in 2.5%.298 Ultrasonography is useful in the adrenal medulla and or anywhere along the parasympathetic
for screening and diagnosis of these lesions.302 Ovarian serous cyst- and sympathetic tracts. Pheochromocytomas are usually benign tumors
adenomas have been described in the literature, and ovarian lesions and can be active (secreting hormones) or inactive depending on their
Genetic Factors: Hereditary Cancer Predisposition Syndromes  •  CHAPTER 13 203

sympathetic or parasympathetic origin. The paragangliomas and


pheochromocytomas can be single or multiple; the presence of multiple, Genetics
bilateral, recurrent, and early (in patients younger than 40 years) In 1993 it was observed that RET gene mutations were associated
tumors or a family history of such tumors should prompt early referral with MEN2A, MEN2B, and FMCT.321,322 Specific RET gene mutations
for genetic assessment. have been associated with MEN2A, MEN2B, and FMCT.318,321,323–325
RET testing of sporadic cases of medullary carcinoma of the thyroid
Genetics will yield a relatively low (5%) rate of diagnosis of MEN2A in the
Given the oxygen sensor function of the carotid body, a common site absence of a family history of the disease, C-cell hyperplasia, or
for chemodectomas, in paraganglioma syndromes, loss-of-function multifocality.322,326 Nonetheless, RET testing is more sensitive than
mutation in all four of the subunits of the mitochondrial succinate traditional biochemical screening. Asymptomatic children with RET
dehydrogenase (SDH) enzyme complex encoded by the genes SDHA, mutations and normal plasma calcitonin levels had small foci of
SDHB, SDHC, and SDHD have been implicated,310 in addition to a medullary carcinoma of the thyroid at time of “prophylactic” surgery.327
gene encoding a mitochondrial protein, SDH5.311 Mutations in SDHD These findings were confirmed in a large study in the United States.328
exhibit autosomal dominant inheritance, with an imprinting mecha- A study of 477 MEN2A families showed an association between
nism310; germline mutations in SDHAF2,311 which encodes the codon 634 mutations and pheochromocytoma and between mutations
mitochondrial protein SDH5 that is required for flavination of the at codons 768 and 804 and FMCT, whereas codon 918 mutation is
SDHA subunit of SDH, have also been associated with disease and MEN2B-specific. Rare families with both MEN2 and Hirschsprung
have been shown to have imprinted parent-of-origin effects. In contrast, disease have MEN2-specific codon mutations.329 A 611 codon mutation
SDHB and SDHC mutations do not appear to be imprinted genes. is associated with a mild form of FMCT with slow progression. The
Although maternally derived SDHD cases have been reported, further classic M918T mutation in exon 16 is found in MEN2B, and a less
analysis revealed that a paternal mutation on an 11p15.5 allele was common mutation in RET codon 883 has also been reported.330,331
also necessary to result in paraganglioma.312 SDHD mutations give
rise to more frequent head and neck tumors, whereas SDHB mutations Clinical Management
exhibit a greater propensity for malignancy and also are associated RET testing has been established as the gold standard for MEN2A
with renal cell cancers (RCCs).313 screening, and prophylactic thyroidectomy has been established as
Of 271 paraganglioma and/or pheochromocytoma cases unselected the primary preventive intervention. Guidelines from the American
for family history, germline mutations were identified in 66 (24%), Thyroid Association for the management of medullary thyroid cancer
with SDHD and SDHB mutations accounting for 23 cases. Of the reflect the known genotype-phenotype correlations in relation to the
remaining 43 germline mutation cases, 30 were attributed to VHL ages at which they recommend childhood prophylactic surgeries.332
disease mutations, and 13 were caused by mutations in the RET Heterozygotes for RET mutations in the setting of MEN2A are screened
gene.314 Given the high frequency of germline mutations in paragan- with abdominal ultrasound and CT, as well as 24-hour urine studies
glioma cases, diagnosis should prompt consideration of genetic testing.315 through the adult years, at least to age 35 years. Plasma screening for
Recently a Dutch founder mutation in SDHD, c.274G>T, p.Asp92Tyr, the pheochromocytomas has been suggested (see the section on VHL
was found to account for 69.1% of all Dutch carriers of a mutation disease). The treatment of choice for patients with MEN2A or MEN2B
in an SDH gene; the second most commonly affected gene was with a unilateral pheochromocytoma is unilateral resection, because
SDHAF2, indicating the importance of considering regional differences substantial morbidity and mortality are associated with the addisonian
in mutation frequencies when formulating a testing strategy.316 state after bilateral adrenalectomy.333 RET kinase inhibitors have been
in clinical trials to evaluate their efficacy in treatment of metastatic
Clinical Management medullary thyroid cancer in sporadic and familial cases of medullary
Treatment for paraganglioma is surgical. Minimal morbidity occurs thyroid cancer.334–338 The FDA has approved the use of the RET
with small tumors, but once the size is greater than 5 cm, cranial kinase, vascular endothelial growth factor (VEGF) receptor, and
nerve loss and baroreceptor failure may be postoperative sequelae. epidermal growth factor receptor (EGFR) inhibitor vandetanib to
Screening for pheochromocytoma is by serum and/or urinary meta- treat adult patients with metastatic medullary thyroid cancer. This
nephrines. Screening for renal cancers is unproven, but abdominal/ approval emerged from the results of a randomized, double-blind
pelvic imaging seems appropriate. phase III trial that showed improvement in progression-free survival
after once-daily administration of the oral agent in this setting.334

A SELECTION OF CANCER PREDISPOSITION Gorlin Syndrome and Nevoid Basal Cell


SYNDROMES WITH TARGETED THERAPIES Carcinoma Syndrome
Multiple Endocrine Neoplasia Type 2 Basal cell carcinomas (BCCs) are the most common malignancy in
humans, with three quarters of a million cases each year. In a small
Clinical Features subset of families, BCCs occur at an early age and in great numbers.
Medullary thyroid carcinoma runs in families about 25% of the time, The nevoid basal cell carcinoma syndrome (NBCCS) consists of
as a syndrome of site-specific familial medullary carcinoma of the multiple BCCs and usually manifests after puberty, accompanied by
thyroid (FMCT) or as multiple endocrine neoplasia type 2 (MEN2).317 odontogenic jaw cysts, congenital skeletal abnormalities, ectopic
Cases usually manifest in the third and fourth decades and are often calcification of the falx cerebri, and characteristic pits in the skin of
bilateral and multifocal. MEN2A is characterized by predisposition the palms and soles.339,340
to medullary thyroid carcinoma, pheochromocytoma, and parathyroid A hallmark of the syndrome is the increased susceptibility of the
disease.318 Patients with MEN2B have an earlier age of onset than do skin to the damaging and tumor-inducing effects of ionizing radiation.341
patients with MEN2A and are characterized by enlarged, nodular Multiple BCCs have developed within 6 to 36 months after radiation
lips, a marfanoid habitus, ganglioneuromatosis of the intestine, and therapy. Unlike Bloom syndrome or ataxia telangiectasia, there is no
other abnormalities.319 This phenotype also includes medullary thyroid in vitro evidence of chromosome fragility.
carcinoma, which may be more aggressive, and pheochromocytoma. One estimate of the prevalence of the syndrome, 1 in 57,000,
Parathyroid disease is less common than in MEN2A. comes from a study of a population of 4 million in northwest
Familial papillary thyroid cancer is distinct from FMCT and is England.342 As described by Gorlin339 in 1987, penetrance for the
associated with an increased incidence of CRC.320 syndrome is virtually 100% over a lifetime.340 Although NBCCS
204 Part I: Science and Clinical Oncology

diagnostic criteria are based on the presence of major and/or minor of at-risk children can be used to diagnose medulloblastomas, although
criteria (two major criteria and one minor criterion, or one major it is unclear whether this scanning improves outcome. If no physical
and two minor criteria),343,344 limited information is available regarding or radiographic stigmata are noted by 5 years of age in the child of
the sensitivity and specificity of each criterion. As will be described an affected patient, the chances that the child is a heterozygote are
shortly, a consensus statement from an international colloquium on small.343 As has been mentioned, radiotherapy for large basal cell
basal cell nevus syndrome suggested that a suspected diagnosis should cancers should be avoided, because this treatment can lead to the
be considered based on one major criterion and molecular confirmation; development of thousands of BCCs in the radiation field.340,341 Prenatal
two major criteria; or one major and two minor criteria.345 The testing for NBCCS is possible if the disease-causing mutation has
component tumors that are included in these criteria are also a topic been identified in an affected family member.
of debate. One study showed that in approximately 5% of patients In recent years, the efficacy of an oral small-molecule inhibitor
with Gorlin syndrome, medulloblastoma develops in the first few of Smoothened homologue, GDC-0449 or vismodegib, has been
years of life, and that 10% of patients with medulloblastoma diagnosed demonstrated in persons with metastatic or locally advanced BCC.350
at age 2 years or younger have Gorlin syndrome.346 Another study In January 2012, vismodegib was the first drug approved by the
found that the only histopathologic subtype of medulloblastoma in FDA for use in the treatment of advanced BCC after a phase II trial
patients with NBCCS is the desmoplastic subtype, which occurs in showed a 9.5-month median progression-free survival in patients with
only 20% of sporadic medulloblastoma cases; it was suggested that locally advanced disease and metastatic disease who responded (43%
the occurrence of the desmoplastic subtype in a patient younger than and 30%, respectively).356,357 Other findings have also demonstrated
2 years of age is pathognomic.347 The international consensus group the usefulness of vismodegib in prevention of BCCs in patients
concluded that childhood medulloblastoma (also called primitive with NBCCS.358
neuroectodermal tumor) should be considered a major criterion. Thus
the major criteria include (1) BCC before 20 years of age, or excessive Hereditary Leiomyomatosis Renal Cell
numbers of BCCs out of proportion to prior sun exposure and skin Cancer Syndrome
type; (2) odontogenic keratocyst of the jaw before 20 years of age;
(3) palmar or plantar pitting; (4) lamellar calcification of the falx Clinical Features
cerebri; (5) medulloblastoma, typically desmoplastic; and (6) a first- The autosomal dominant hereditary leiomyomatosis and renal cell
degree relative with basal cell nevus syndrome. The minor criteria cancer syndrome (HLRCC) predisposes to benign cutaneous and
include (1) rib anomalies; (2) other specific skeletal malformations uterine leiomyomas (fibroids), uterine leiomyosarcomas, and papillary
and radiologic changes (i.e., vertebral anomalies, kyphoscoliosis, short type II RCC or renal collecting duct carcinoma.359–362
fourth metacarpals, and postaxial polydactyly); (3) macrocephaly; (4) Cutaneous leiomyomas arise from the erector pili muscles, existing
cleft lip or palate; (5) ovarian or cardiac fibroma; (6) lymphomesenteric as single or multiple lesions. They can be skin-colored or light brown
cysts; (7) ocular abnormalities (i.e., strabismus, hypertelorism, congenital and are sensitive to touch and cold temperature. Cutaneous lesions
cataracts, glaucoma, and coloboma).345 may be diffuse and symmetric or present in segmental bands along
the lines of Blaschko. Of 11 females, 9 had cutaneous leiomyomas
Genetics and 9 had uterine leiomyomas.363,364 Leiomyomas of the uterus are
In 1996 Johnson and coworkers found mutations in exon 15 of the multiple and symptomatic and result in hysterectomy by age 30 years
human homologue of the Drosophila patched gene (called PTH or in more than 44% of women with HLRCC.361 Kidney tumors in
PTCH)348 in 2 of 60 typical NBCCS kindreds. In 86%, the mutations persons with HLRCC are aggressive and often single, small lesions
caused a truncated protein.349 NBCCS is caused by germline loss of of high malignant potential. Within an HLRCC kindred, the age at
function mutations in the gene PTCH mapped to chromosomal locus onset of four cases of kidney cancer ranged from age 33 to 48 years,
9q22.3. The PTCH gene consists of 23 exons and encodes an integral and all were diagnosed in females.359,365 A Finnish series of 98 HLRCC
membrane protein, PTCH1, with 12 transmembrane regions and cases found a standardized increased risk of 71-fold for uterine leio-
two extracellular loops and a putative sterol-sensing domain. When myosarcoma and 6.5-fold for RCC.366 In a separate series of patients
unbound by Hedgehog protein, PTCH1 inhibits Smoothened with HLRCC, renal cancer occurred in 62% of 21 patients, 76% had
homologue, which in turn inhibits the Hedgehog signaling pathway, cutaneous leiomyomas, and uterine leiomyomas occurred in 90% to
a pathway upregulated in BCCs.350 Mutations of PTCH are the only 100% of the women.367 Adrenal gland adenoma and kidney cysts, as
known genetic alterations associated with NBCCS.351 However, several well as breast and bladder carcinoma, have also been reported.366
studies have found no genotype-phenotype correlations,349,352,353 which However, a study of 85 patients with breast cancer with a family
suggests that other factors might add to the development of patients’ history of HLRCC found no increased risk for breast cancer, although
clinical features. For kindreds with diagnostic clinical findings of a considerably larger sample size could be more informative.368
NBCCS that test negative for a mutation by sequence analysis, testing
that identifies deletions and duplications may be performed. About Genetics
70% to 80% of probands have inherited the condition from a parent, The gene associated with hereditary leiomyomatosis and RCC on
and about 20% to 30% of probands have a de novo mutation. 1q42.3-q43 encodes fumarate hydratase (FH), a tricarboxylic acid/
Krebs cycle, mitochondrial enzyme. Dominant mutations in FH cause
Risk Management Recommendations HLRCC in a manner that is consistent with Knudsen’s two-hit tumor
Life expectancy in persons with NBCCS is not significantly different suppressor model. Hypoxia-inducible factor (HIF) levels are increased
from average. The major clinical issues revolve around the cosmetic in clear cell renal carcinomas. FH is critical for aerobic metabolism
effects of treating multiple skin tumors and jaw keratocysts, which and functions to convert fumarate to malate in Krebs cycle. When
can recur. The jaw cysts may also undergo malignant transformation.354 FH is inactivated, fumarate levels build up and competitively inhibit
Consultation with oral and plastic surgeons and dermatologists is HIF prolyl hydroxylase (HPH), a regulator of intracellular HIF. When
important. Because of early-onset disease risk, for example, for medul- HPH is inactivated, HIF levels are upregulated, resulting in transcription
loblastomas, genetic testing of children is appropriate for this condition. of downstream genes.369 Mutations in the FH gene are detected in
Evaluation of members of NBCCS kindreds includes skin examination, about 75% to 100% of patients with multiple cutaneous and uterine
measurement of head circumference, and radiographic examination leiomyomatosis.360,362,367 One mutation, R190H, was detected in 11
of the skull, spine, ribs, and jaws.355 Ophthalmologic and dental of 31 unrelated North American HLRCC kindreds, and the mutation
examination and radiographic monitoring of the jaw cysts (oropan- R58X has also been identified in multiple unrelated kindreds.367 In
tomography) may be performed on affected individuals. MRI scans addition, a mutation, 905-1G>A, was found in four Iranian HLRCC
Genetic Factors: Hereditary Cancer Predisposition Syndromes  •  CHAPTER 13 205

kindreds, possibly representing a founder mutation in this population.370 VEGF, platelet-derived growth factor, EGFR, and transforming
Germline biallelic mutations in FH cause the mitochondrial encepha- growth factor–α. Many of the current therapeutic strategies are
lomyopathy FH deficiency, and obligate carriers have developed based on targeting the receptors of the HIF-regulated genes. Sunitinib,
HLRCC.371–374 Reduced FH activity in lymphoblastoid and fibroblast sorafenib, bevacizumab plus interferon-α, pazopanib, temsirolimus,
cells of HLRCC patients may facilitate confirmation of clinical diagnosis and everolimus are drugs that are currently approved for treating
via enzymatic testing.375 metastatic RCC through targeting the VHL transcription pathway, and
they form the backbone of the treatment of a disease that is otherwise
Clinical Management resistant to other treatment modalities such as radiotherapy and
Screening at regular intervals for renal lesions with CT with contrast chemotherapy.384
agent is indicated in patients with FH mutations. MRI is recommended
for uterine leiomyoma screening, and ultrasonography can also be Birt-Hogg-Dubé Syndrome
used.376 Systemic therapy for HLRCC will likely attempt to prevent
the increase in HIF levels or target the transcription products of Clinical Features
VHL-independent HIF accumulation such as VEGF and EGFR. A Birt-Hogg-Dubé (BHD) cancer predisposition syndrome was first
phase II trial of erlotinib, an oral EGFR tyrosine kinase inhibitor, in described in 1977.385 This rare genodermatosis comprises an unusual
locally advanced and metastatic papillary renal cell carcinoma reported triad of findings: fibrofolliculomas that appear as white or skin-colored
an overall Response Evaluation Criteria in Solid Tumors (RECIST) papules on the face and upper torso, spontaneous pneumothorax, and
response rate of 11%, with an additional 24 patients (53%) experiencing kidney tumors.362 The cutaneous lesions usually appear in the region
stable disease377; another phase II trial of erlotinib and bevacizumab of the head, neck, and upper part of the trunk in the third or fourth
(monoclonal antibody against VEGF) is currently underway.378 decade of life, and renal tumors develop in about 15% to 30% of
patients with cutaneous BHD syndrome. A recent review of 130 solid
von Hippel-Lindau Disease renal tumors resected from 30 patients with BHD in 19 different
families revealed that renal tumors were multiple and bilateral and
Clinical Features were noted at an early age (mean, 50.7 years). The vast majority were
VHL disease is characterized by hemangioblastomas of the retina, hybrid oncocytic neoplasms with areas reminiscent of chromophobe
brain, and spinal cord; pheochromocytomas; renal cysts, and clear renal cell carcinoma and oncocytoma, whereas a significant number
cell renal cell carcinoma; pancreatic endocrine tumor; and endolym- were chromophobe renal cell carcinomas, and a minority were con-
phatic sac tumors.379,380 Diagnosis depends on the presence of a ventional clear cell renal carcinomas,386,387 thus demonstrating the
VHL-associated tumor and a family history consistent with VHL or phenotypic heterogeneity of renal cancer that is unique to this syndrome
alternatively the presence of at least two VHL-associated tumors.380 compared with other hereditary renal cancer predispositions. Chro-
mophobe and hybrid oncocytic tumors comprise 23% and 67% of
Genetics BHD renal tumors, respectively; clear cell tumors comprise 7% and
VHL is an autosomal dominant disease caused by germline mutations oncocytomas comprise 3%, whereas papillary renal cell carcinomas
in VHL, the protein product of which degrades hydroxylated HIF. are rarely noted.386 Persons with BHD syndrome are at markedly
HIF is a transcription factor for angiogenesis genes, upregulated in increased risk of spontaneous pneumothoraces. About a third, 64 of
renal cell carcinoma. In normal conditions of oxygenation, HPH 198 (32%), of BHD-affected individuals had a history of a spontaneous
hydroxylyzes HIF and VHL recognizes the hydrolysed HIF and mediates pneumothorax.388 The syndrome is also associated with a progressive
proteosomal degradation. In hypoxic conditions, HPH does not flecked chorioretinopathy with constricted visual fields.389 There is an
hydrolyze HIF, and therefore HIF is not degraded.369 association with colorectal neoplasia in some families with BHD,390
as well as colorectal polyps in up to half of patients with BHD.390,391
Screening for von Hippel-Lindau Disease Similarities exist between BHD and tuberous sclerosis (TSC), which
Health screening in persons with VHL disease should include annual may be explained by the fact that both BHD and TSC genes function
evaluations starting at 1 year of age. It includes screening for neurologic, in the same pathway, mammalian target of rapamycin (mTOR). In
vision (retinal angiomas), and hearing symptoms and signs and blood this regard, angiomyolipomas have been described in patients with
pressure. Starting at age 5 years, annual blood or urinary fractionated BHD and fibrofolliculomas were described in patients with TSC in
metanephrines (pheochromocytoma) should be evaluated, audiology case reports.392,393
assessment should be performed every 2 to 3 years or annually if the
patient is symptomatic, and MRI should be performed if repeated Genetics
ear infections are present (looking for endolymphatic sac tumors). BHD is inherited in an autosomal dominant manner and is caused
Starting at age 16 years, an annual abdominal ultrasound examination by germline mutations in the gene FLCN located on chromosome
and MRI scan of the abdomen are recommended (with scans of the 17p11.2, which encodes folliculin.394,395 FLCN acts as a tumor sup-
kidney, pancreas, and adrenal glands every other year), along with an pressor protein involved in the regulation of adenosine monophosphate–
MRI of the brain and total spine every 2 years.381 No standardized activated protein kinase and mTOR signaling pathways.396,397 More
guidelines exist for the management of other VHL-associated conditions, than 50 mutations in the FLCN gene have been reported, located in
and thus an individualized approach is required.380 The literature is all translated exons (4–14) and intronic borders, with exon 11 being
growing regarding management of renal cell carcinoma; early surgery considered a mutation hot spot.397 In a large study, 27 of 52 BHD-
for lesions greater than 3 cm is recommended, depending on the size affected families had an insertion/deletion in exon 11 of the gene
and location of the tumor, along with undertaking nephron-sparing (c.1733insC or c.1733delC). Phenotype manifestations among those
procedures382 with preservation of the adrenal gland. with either the insertion or the deletion were similar for fibrofollicu-
lomas and pneumothoraces. However, the incidence of renal tumors
Systemic Therapy in von Hippel-Lindau Disease was significantly higher among those with the C-insertion compared
Understanding the VHL pathway has provided targets for systemic with the C-deletion (33% versus 6%).388
therapy in patients with advanced renal cell carcinoma. The VHL gene
has been vigorously studied, and its product is vital in targeting HIF-1α Risk Management
and HIF-2α for ubiquitin-mediated degradation.383 The transcription Patients with BHD syndrome and their relatives should undergo
products of HIF-1α and HIF-2α are known to regulate a number abdominal CT and renal ultrasound screening for renal tumors. Lung
of downstream genes that are involved in tumorigenesis, including cysts are best seen on CT scans. Dermatologic consultation is advised.
206 Part I: Science and Clinical Oncology

Ophthalmologic examination should be performed in patients with A common theme relating to the pharmacologic treatment and
BHD syndrome because of the high incidence of chorioretinopathy. prevention of hereditary cancers is the development of drugs targeting
Because of the newly discovered association between FLCN and mTOR specific pathways. Cancer predisposition syndromes represent human
activation, there is a potential therapeutic or preventive role of mTOR models of “knockouts” or “knockins” of germline alterations in
inhibitors in patients affected by BHD, which is currently being oncogenic pathways and as such may be amenable to pathway-specific
investigated in sporadic chromophobe tumors.398 approaches. Therefore ongoing advancements in the understanding
of the molecular pathways and the development of targeted agents
Tuberous Sclerosis and Gastrointestinal will have immediate applicability to the treatment and, ultimately,
Stromal Tumor chemoprevention of hereditary cancer.

Other syndromes that have shown the promise of targeted therapy BAP1 INHERITED CANCER SUSCEPTIBILITY
include TSC399 and inherited gastrointestinal stromal tumor (GIST).400
TSC is an autosomal dominant syndrome caused by germline muta- More recently it has been elucidated that germline and sporadic
tions in TSC1 and TSC2. Major features include facial angiofibromas mutations in BAP1, a nuclear-localized, ubiquitin carboxy-terminal
or forehead plaque, nontraumatic ungual or periungual fibromas, hydrolase that binds to the RING finger domain of BRCA1,409 are
hypomelanotic macules (three or more), shagreen patch (connective associated with uveal melanomas and mesothelioma.410–412 Wiesner
tissue nevus), multiple retinal nodular hamartomas, cortical tuber, and colleagues412 identified germline mutations in BAP1 in two
subependymal nodule, subependymal giant cell astrocytoma (SEGA), families, each with a single case of uveal melanoma in addition to
cardiac rhabdomyoma, single or multiple, lymphangiomyomatosis, multiple affected individuals with melanocytic lesions inherited in
and renal angiomyolipoma. Minor features include multiple randomly an autosomal dominant manner. The BAP1-associated melanocytic
distributed pits in dental enamel, hamartomatous rectal polyps, bone neoplasms were described as atypical, with an appearance ranging from
cysts, cerebral white matter radial migration lines, gingival fibromas, epithelioid nevi to atypical melanocytic proliferations. A follow-up
nonrenal hamartoma, retinal achromic patch, “confetti” skin lesions, and study suggests that the frequency of uveal melanomas attributable
multiple renal cysts. Clinical diagnosis determining the probability of to germline mutations in BAP1 is low (1 of 53 probands with uveal
having TSC is based on a combination of major and minor features.399 melanoma, 5 of whom had a family history of uveal melanoma).413
Angiomyolipomas are the dominant renal lesions in TSC, whereas In this same study, lung adenocarcinoma and meningioma were also
RCC has been reported in 1% to 3% of patients with TSC.401 Clinical reported in the proband’s carrier family members, the tumors of
management of angiomyolipomas is by observation, removal, or which, in addition to the uveal melanoma, demonstrated biallelic
embolization, depending on the size and location of the tumor and inactivation, suggestive of tumors associated with a new syndrome.413
its potential for spontaneous bleeding. Recently, medical therapies Testa and colleagues411 found germline mutations in BAP1 in two
targeting the mTOR pathway have been proposed as another treatment families with multiple affected individuals with mesothelioma and
modality. In 2009, the FDA granted everolimus (Afinitor) an orphan showed an increased frequency of germline mutations in sporadic
drug designation to treat renal angiomyolipomas and SEGAs in patients mesothelioma samples (2 of 26 cases). Somatic mutations in BAP1 have
with TSC. (Orphan designations are given to drugs that have shown also been found to define a new class of renal cell carcinoma, whereby
promise in the treatment of a disease or condition affecting fewer a germline variant was also reported.414 Epigenetic EZH2 inhibitors
than 200,000 patients in the United States.) In April 2012, the FDA are being explored in targeted treatment of mesothelioma with BAP1
granted approval to specifically treat noncancerous kidney tumors mutations.415
(renal angiomyolipomas) not requiring immediate surgery in patients
with TSC complex after the results of a phase III randomized controlled TUMOR-NORMAL SEQUENCING
trial (EXIST-2) that enrolled 118 patients at least 18 years of age who
had angiomyolipomas, randomized 2 : 1 to receive oral everolimus Inclusion of “normal” or constitutional DNA as a reference for somatic
(Afinitor) at 10 mg once daily or placebo until tumor progression or (tumor) sequencing adds to the sensitivity of the assay416 and poses
unacceptable toxicity. With only half of the patients receiving less opportunities for targeted (“precision”) prevention.417 Anonymized
than 8 months of treatment, 42% treated with Afinitor showed a retrospective analyses of tumor-normal genomic data from several
substantial reduction in tumor size lasting, on average, more than 5 large centers89,90,416,418–420 Table 13.6) demonstrated 3% to 17.5%
months, whereas none of the patients receiving placebo showed any clinically actionable germline findings. These studies will also allow
tumor shrinkage.402 As part of the FDA approval, Afinitor’s manufacturer for targeting treatment as well as prevention, as already evidenced by
(Novartis) will continue to monitor the patients for at least 4 years the 12% fraction of prostate cases with inherited mutations of DNA
to determine the duration of the response and how it affects the need repair genes.421 Included in the DNA repair genes are DNA homologous
for surgery and the control of the disease.403 repair genes potentially amenable to therapy with PARP inhibitors,
Mesenchymal stromal tumors of the gastrointestinal tract (GISTs) as well as subsets with Lynch-associated mutations, potentially amenable
are associated with germline and sporadic mutations in KIT and to immunotherapy.
PDGFRA,404–407 leading to constitutive activation. Familial cases are These findings have led some institutions to collect tumor-normal
inherited in an autosomal dominant manner; the mean age of diagnosis DNA sequence in the setting of a consent process that allows com-
is in the fifth decade, and multicentric disease generally develops in munication of these findings. This tumor-normal testing will result
the small bowel.400 Neurofibromatosis type 1, Carney-Stratakis, and in a “cascade” of genomic information to unaffected relatives for use
Carney triad are associated with GIST, although these tumors do not in targeted prevention (and even reproductive planning), at the same
tend to demonstrate mutations in c-KIT and PDGFR.400 In 2002 the time as tumor-derived information from the proband is used to target
selective abl, c-kit, and PDGFR tyrosine kinase inhibitor imatinib therapy. This new model of integrated tumor and inherited (germline)
was granted approval by the FDA for the treatment of CD117-positive DNA sequencing poses a challenge for clinical oncology over the next
unresectable or metastatic GIST. In 2006, sunitinib was granted decade.
approval by the FDA for patients whose disease progressed while
taking imatinib or for those who could not tolerate imatinib. Because CONCLUSION
of an overlap between germline and sporadic mutations, it is observed
that the use of imatinib stabilized the esophageal GISTs in a patient The relatively high-penetrance cancer susceptibility alleles associated
harboring a germline mutation in c-KIT, previously reported in the with the syndromes reviewed in this chapter account for a minority
germline and sporadic setting mutation.408 of human cancers. Nonetheless, the number of molecularly defined
Genetic Factors: Hereditary Cancer Predisposition Syndromes  •  CHAPTER 13 207

Table 13.6 Tumor-Normal Sequencing Studies and Proportion of Clinically Actionable


Findings89,90,416,418–420
St. Jude
Memorial Sloan Memorial Sloan Children’s
Institutional Johns Hopkins University Kettering Kettering (Clinically Research Baylor College
Series University of Michigan (Anonymized) Annotated) Hospital of Medicine
Number sequenced 815 91 1566 1040 1120 150
Number of germline 27 9 198 182 95 13
actionable findings
Percentage of 3% 10% 12.6% 17.5% 8.5% 8.6%
germline actionable
findings

cancer syndromes continues to grow and will increasingly include a ACKNOWLEDGMENTS


large number of genomic variants associated with a more modest risk
for disease, as well as measures of polygenic interactions of these This work was supported by the Robert and Kate Niehaus Center
pathways and interactions of these alleles with acquired risk factors. for Inherited Cancer Genomics at Memorial Sloan Kettering Cancer
The emergence of next-generation sequencing applied to diagnostic Center. We are grateful to Carolyn Stewart and Christina Tran for
evaluation of tumor-normal DNA samples offers a major new challenge their help in preparing the manuscript.
to the integration of cancer risk counseling into the “targeted” thera-
peutic management of patients and families affected by inherited The complete reference list is available online at
malignancies. ExpertConsult.com.

KEY REFERENCES
2. Offit K. Decade in review—genomics: a decade of susceptibility: out on the high wire without a net? 150. Kaufman B, Shapira-Frommer R, Schmutzler
discovery in cancer genomics. Nat Rev Clin Oncol. J Clin Oncol. 2013;31(10):1267–1270. RK, et al. Olaparib monotherapy in patients with
2014;11(11):632–634. 19. Stadler ZK, Schrader KA, Vijai J, Robson ME, Offit advanced cancer and a germline BRCA1/2 mutation.
3. Offit K. The future of clinical cancer genomics. K. Cancer genomics and inherited risk. J Clin Oncol. J Clin Oncol. 2015;33(3):244–250.
Semin Oncol. 2016;43(5):615–622. 2014;32(7):687–698. 151. Robson M, Im SA, Senkus E, et al. Olaparib for
4. Robson ME, Bradbury AR, Arun B, et al. American 21. Kuchenbaecker KB, McGuffog L, Barrowdale D, metastatic breast cancer in patients with a germline
Society of Clinical Oncology policy statement et al. Evaluation of polygenic risk scores for breast BRCA mutation. N Engl J Med. 2017.
update: genetic and genomic testing for cancer and ovarian cancer risk prediction in BRCA1 and 167. Hampel H, Frankel WL, Martin E, et  al.
susceptibility. J Clin Oncol. 2015;33(31):3660–3667. BRCA2 mutation carriers. J Natl Cancer Inst. 2017; Screening for the Lynch syndrome (hereditary
5. Walsh MF, Chang VY, Kohlmann WK, et al. 109(7). nonpolyposis colorectal cancer). N Engl J Med.
Recommendations for childhood cancer screening 35. Weitzel JN, Blazer KR, MacDonald DJ, Culver 2005;352(18):1851–1860.
and surveillance in DNA repair disorders. Clin JO, Offit K. Genetics, genomics, and cancer risk 181. Win AK, Young JP, Lindor NM, et al. Colorectal
Cancer Res. 2017;23(11):e23–e31. assessment: state of the art and future directions in and other cancer risks for carriers and noncarriers
6. Villani A, Greer MC, Kalish JM, et al. Recommen- the era of personalized medicine. CA Cancer J Clin. from families with a DNA mismatch repair gene
dations for cancer surveillance in individuals with 2011;61(5):327–359. mutation: a prospective cohort study. J Clin Oncol.
rasopathies and other rare genetic conditions with 39. Robson M, Offit K. Clinical practice. Management 2012;30(9):958–964.
increased cancer risk. Clin Cancer Res. 2017;23(12): of an inherited predisposition to breast cancer. N 185. Barnetson RA, Tenesa A, Farrington SM, et al.
e83–e90. Engl J Med. 2007;357(2):154–162. Identification and survival of carriers of mutations
7. Foulkes WD, Kamihara J, Evans DGR, et al. Cancer 49. Antoniou AC, Casadei S, Heikkinen T, et al. Breast- in DNA mismatch-repair genes in colon cancer.
surveillance in Gorlin syndrome and rhabdoid cancer risk in families with mutations in PALB2. N Engl J Med. 2006;354(26):2751–2763.
tumor predisposition syndrome. Clin Cancer Res. N Engl J Med. 2014;371(6):497–506. 189. Recommendations from the EGAPP Working
2017;23(12):e62–e67. 74. Tung N, Battelli C, Allen B, et al. Frequency Group: genetic testing strategies in newly diagnosed
8. Wasserman JD, Tomlinson GE, Druker H, et al. of mutations in individuals with breast cancer individuals with colorectal cancer aimed at reducing
Multiple endocrine neoplasia and hyperparathyroid- referred for BRCA1 and BRCA2 testing using morbidity and mortality from Lynch syndrome in
jaw tumor syndromes: clinical features, genetics, and next-generation sequencing with a 25-gene panel. relatives. Genet Med. 2009;11(1):35–41.
surveillance recommendations in childhood. Clin Cancer. 2015;121(1):25–33. 190. National Comprehensive Cancer Centers. NCCN
Cancer Res. 2017;23(13):e123–e132. 84. Tung N, Domchek SM, Stadler Z, et al. Counsel- Clinical Practice Guidelines in Oncology, Genetic/
10. Lancaster JM, Powell CB, Chen LM, Richardson ling framework for moderate-penetrance cancer- Familial High-Risk Assessment: Colorectal Version
DL. Society of Gynecologic Oncology statement susceptibility mutations. Nat Rev Clin Oncol. 2016; 1.2017; 2017. https://www.nccn.org/professionals/
on risk assessment for inherited gynecologic cancer 13(9):581–588. physician_gls/pdf/genetics_colon.pdf.
predispositions. Gynecol Oncol. 2015;136(1):3–7. 116. National Comprehensive Cancer Centers. 212. Schmeler KM, Lynch HT, Chen L-m, et al. Pro-
13. Daly MB, Pilarski R, Berry M, et al. NCCN NCCN Clinical Practive Guidelines in Oncol- phylactic surgery to reduce the risk of gynecologic
Guidelines Insights: Genetic/Familial High-Risk ogy, Genetic/Familial High-Risk Assessment: cancers in the Lynch syndrome. N Engl J Med. 2006;
Assessment: Breast and Ovarian, Version 2.2017. Breast and Ovarian Version 2.2017; 2016. http:// 354(3):261–269.
J Natl Compr Canc Netw. 2017;15(1):9–20. www.nccn.org/professionals/physician_gls/pdf/ 220. Le DT, Durham JN, Smith KN, et al. Mismatch-
17. Easton DF, Pharoah PD, Antoniou AC, et al. Gene- genetics_screening.pdf. repair deficiency predicts response of solid tumors
panel sequencing and the prediction of breast-cancer 137. Kauff ND, Satagopan JM, Robson ME, et al. to PD-1 blockade. Science. 2017.
risk. N Engl J Med. 2015;372(23):2243–2257. Risk-reducing salpingo-oophorectomy in women 237. Offit K, Kohut K, Clagett B, et al. Cancer genetic
18. Domchek SM, Bradbury A, Garber JE, Offit K, with a BRCA1 or BRCA2 mutation. N Engl J Med. testing and assisted reproduction. J Clin Oncol.
Robson ME. Multiplex genetic testing for cancer 2002;346(21):1609–1615. 2006;24(29):4775–4782.
208 Part I: Science and Clinical Oncology

242. Guillem JG, Wood WC, Moley JF, et al. ASCO/ 345. Bree AF, Shah MR, Group BC. Consensus statement 421. Pritchard CC, Mateo J, Walsh MF, et al. Inherited
SSO review of current role of risk-reducing surgery from the first international colloquium on basal DNA-repair gene mutations in men with metastatic
in common hereditary cancer syndromes. J Clin cell nevus syndrome (BCNS). Am J Med Genet A. prostate cancer. N Engl J Med. 2016;375(5):443–453.
Oncol. 2006;24(28):4642–4660. 2011;155A(9):2091–2097. 438. Syngal S, Brand RE, Church JM, Giardiello FM,
249. Nielsen M, Morreau H, Vasen HFA, Hes FJ. 350. Von Hoff DD, LoRusso PM, Rudin CM, et al. Inhi- Hampel HL, Burt RW. ACG clinical guideline:
MUTYH-associated polyposis (MAP). Crit Rev bition of the hedgehog pathway in advanced basal-cell genetic testing and management of hereditary
Oncol Hematol. 2011;79(1):1–16. carcinoma. N Engl J Med. 2009;361(12):1164– gastrointestinal cancer syndromes. Am J Gastroenterol.
260. Palles C, Cazier JB, Howarth KM, et al. Germline 1172. 2015;110(2):223–262, quiz 263.
mutations affecting the proofreading domains of 361. Linehan WM, Pinto PA, Srinivasan R, et al. Identi- 455. Foulkes WD, Bahubeshi A, Hamel N, et al.
POLE and POLD1 predispose to colorectal adenomas fication of the genes for kidney cancer: opportunity Extending the phenotypes associated with DICER1
and carcinomas. Nat Genet. 2013;45(2):136–144. for disease-specific targeted therapeutics. Clin Cancer mutations. Hum Mutat. 2011;32(12):1381–1384.
267. Fitzgerald RC, Hardwick R, Huntsman D, et al. Res. 2007;13(2 Pt 2):671s–679s. 478. Hasumi H, Baba M, Hasumi Y, Furuya M, Yao M.
Hereditary diffuse gastric cancer: updated consensus 402. Bissler J, Kingswood JC, Zonnenberg BA, et al. Birt-Hogg-Dube syndrome: clinical and molecular
guidelines for clinical management and directions for Everolimus therapy for angiomyolipoma in patients aspects of recently identified kidney cancer syndrome.
future research. J Med Genet. 2010;47(7):436–444. with tuberous sclerosis complex or sporadic lymphan- Int J Urol. 2016;23(3):204–210.
306. Boikos SA, Stratakis CA. Carney complex: the first gioleiomyomatosis: results from EXIST-2. J Clin 504. Tabori U, Hansford JR, Achatz MI, et al. Clinical
20 years. Curr Opin Oncol. 2007;19(1):24–29. Oncol. 2012;30(suppl 5):356. management and tumor surveillance recommenda-
314. Neumann HPH, Bausch B, McWhinney SR, et al. 417. Bombard Y, Robson M, Offit K. Revealing the tions of inherited mismatch repair deficiency in
Germ-line mutations in nonsyndromic pheochromo- incidentalome when targeting the tumor genome. childhood. Clin Cancer Res. 2017;23(11):e32–
cytoma. N Engl J Med. 2002;346(19):1459–1466. JAMA. 2013;310(8):795–796. e37.
318. Moline J, Eng C. Multiple endocrine neoplasia type 420. Mandelker D, Zhang L, Kemel Y, et al. Mutation 536. Pilarski R, Burt R, Kohlman W, Pho L, Shannon
2: an overview. Genet Med. 2011;13(9):755–764. detection in patients with advanced cancer by univer- KM, Swisher E. Cowden syndrome and the PTEN
336. Lam ET, Ringel MD, Kloos RT, et al. Phase II clini- sal sequencing of cancer-related genes in tumor and hamartoma tumor syndrome: systematic review
cal trial of sorafenib in metastatic medullary thyroid normal DNA compared to guideline-based germline and revised diagnostic criteria. J Natl Cancer Inst.
cancer. J Clin Oncol. 2010;28(14):2323–2330. testing. JAMA. 2017;In Press. 2013;105(21):1607–1616.
Genetic Factors: Hereditary Cancer Predisposition Syndromes  •  CHAPTER 13 208.e1
208.e1

REFERENCES
1. Offit K. Clinical Cancer Genetics: Risk Counseling BRCA2 mutation carriers. J Natl Cancer Inst. 44. Starink TM. Cowden’s disease: analysis of fourteen
and Management. New York: Wiley-Liss; 1998. 2017;109(7). new cases. J Am Acad Dermatol. 1984;11(6):
2. Offit K. Decade in review—genomics: a decade of 22. Lecarpentier J, Silvestri V, Kuchenbaecker KB, 1127–1141.
discovery in cancer genomics. Nat Rev Clin Oncol. et al. Prediction of breast and prostate cancer risks 45. Sidransky D, Tokino T, Helzlsouer K, et al. Inherited
2014;11(11):632–634. in male BRCA1 and BRCA2 mutation carriers p53 gene mutations in breast cancer. Cancer Res.
3. Offit K. The future of clinical cancer genomics. using polygenic risk scores. J Clin Oncol. 2017; 1992;52(10):2984–2986.
Semin Oncol. 2016;43(5):615–622. 35(20):2240–2250. 46. Lim W, Hearle N, Shah B, et al. Further observations
4. Robson ME, Bradbury AR, Arun B, et al. American 23. Stadler ZK, Thom P, Robson ME, et al. Genome- on LKB1/STK11 status and cancer risk in Peutz-
Society of Clinical Oncology Policy statement wide association studies of cancer. J Clin Oncol. Jeghers syndrome. Br J Cancer. 2003;89(2):308–313.
update: genetic and genomic testing for cancer 2010;28(27):4255–4267. 47. Hearle N, Schumacher V, Menko FH, et al. Fre-
susceptibility. J Clin Oncol. 2015;33(31):3660–3667. 24. Malkin D. Li-fraumeni syndrome. Genes Cancer. quency and spectrum of cancers in the Peutz-Jeghers
5. Walsh MF, Chang VY, Kohlmann WK, et al. Recom- 2011;2(4):475–484. syndrome. Clin Cancer Res. 2006;12(10):3209–3215.
mendations for Childhood cancer screening and 25. Lakhani VT, You YN, Wells SA. The multiple 48. Wong MW, Nordfors C, Mossman D, et al. BRIP1,
surveillance in DNA repair disorders. Clin Cancer endocrine neoplasia syndromes. Annu Rev Med. PALB2, and RAD51C mutation analysis reveals
Res. 2017;23(11):e23–e31. 2007;58:253–265. their relative importance as genetic susceptibility
6. Villani A, Greer MC, Kalish JM, et al. Recommen- 26. Marx SJ. Molecular genetics of multiple endo- factors for breast cancer. Breast Cancer Res Treat.
dations for cancer surveillance in individuals with crine neoplasia types 1 and 2. Nat Rev Cancer. 2011;127(3):853–859.
RASopathies and other rare genetic conditions with 2005;5(5):367–375. 49. Antoniou AC, Casadei S, Heikkinen T, et al. Breast-
increased cancer risk. Clin Cancer Res. 2017;23(12): 27. Balmer A, Zografos L, Munier F. Diagnosis and cancer risk in families with mutations in PALB2.
e83–e90. current management of retinoblastoma. Oncogene. N Engl J Med. 2014;371(6):497–506.
7. Foulkes WD, Kamihara J, Evans DGR, et al. Cancer 2006;25(38):5341–5349. 50. Slater EP, Langer P, Niemczyk E, et al. PALB2
surveillance in Gorlin syndrome and rhabdoid tumor 28. Hayward NK. Genetics of melanoma predisposition. mutations in European familial pancreatic cancer
predisposition syndrome. Clin Cancer Res. 2017; Oncogene. 2003;22(20):3053–3062. families. Clin Genet. 2010;78(5):490–494.
23(12):e62–e67. 29. Felsani A, Mileo AM, Maresca V, Picardo M, Paggi 51. Jones S, Hruban RH, Kamiyama M, et al. Exomic
8. Wasserman JD, Tomlinson GE, Druker H, et al. MG. New technologies used in the study of human sequencing identifies PALB2 as a pancreatic cancer
Multiple endocrine neoplasia and hyperparathyroid- melanoma. Int Rev Cytol. 2007;261:247–286. susceptibility gene. Science. 2009;324(5924):217.
jaw tumor syndromes: clinical features, genetics, and 30. Kaelin WG. Molecular basis of the VHL hereditary 52. Ford D, Easton DF, Stratton M, et al. Genetic
surveillance recommendations in childhood. Clin cancer syndrome. Nat Rev Cancer. 2002;2(9): heterogeneity and penetrance analysis of the BRCA1
Cancer Res. 2017;23(13):e123–e132. 673–682. and BRCA2 genes in breast cancer families. The
9. Zon RT, Goss E, Vogel VG, et al. American Society 31. Woodward ER, Maher ER. Von Hippel-Lindau Breast Cancer Linkage Consortium. Am J Hum
of Clinical Oncology policy statement: the role of the disease and endocrine tumour susceptibility. Endocr Genet. 1998;62(3):676–689.
oncologist in cancer prevention and risk assessment. Relat Cancer. 2006;13(2):415–425. 53. Frank TS, Deffenbaugh AM, Reid JE. Clinical
J Clin Oncol. 2009;27(6):986–993. 32. Sudarshan S, Linehan WM. Genetic basis of cancer characteristics of individuals with germline mutations
10. Lancaster JM, Powell CB, Chen LM, Richardson of the kidney. Semin Oncol. 2006;33(5):544– in BRCA1 and of 10,000 individuals. J Clin Oncol.
DL. Society of Gynecologic Oncology statement 551. 2002;20(6):1480–1490.
on risk assessment for inherited gynecologic cancer 33. Ferner RE. Neurofibromatosis 1 and neuro- 54. Pathology of familial breast cancer: Differences
predispositions. Gynecol Oncol. 2015;136(1):3–7. fibromatosis 2: a twenty first century perspective. between breast cancers in carriers of BRCA1 or
11. ACOG Practice Bulletin No. 103: Hereditary Lancet Neurol. 2007;6(4):340–351. BRCA2 mutations and sporadic cases. Breast Cancer
breast and ovarian cancer syndrome. Obstet Gynecol. 34. Lee M-J, Stephenson DA. Recent developments Linkage Consortium. Lancet. 1997;349(9064):
2009;113(4):957–966. in neurofibromatosis type 1. Curr Opin Neurol. 1505–1510.
12. American Gastroenterological Association medical 2007;20(2):135–141. 55. Johannsson OT, Idvall I, Anderson C, et al. Tumour
position statement: hereditary colorectal cancer 35. Weitzel JN, Blazer KR, MacDonald DJ, Culver biological features of BRCA1-induced breast and
and genetic testing. Gastroenterology. 2001;121(1): JO, Offit K. Genetics, genomics, and cancer risk ovarian cancer. Eur J Cancer. 1997;33(3):362–371.
195–197. assessment: state of the art and future directions in 56. Robson M, Rajan P, Rosen PP, et al. BRCA-associated
13. Daly MB, Pilarski R, Berry M, et al. NCCN the era of personalized medicine. CA Cancer J Clin. breast cancer: absence of a characteristic immuno-
Guidelines Insights: Genetic/Familial High-Risk 2011;61(5):327–359. phenotype. Cancer Res. 1998;58(9):1839–1842.
Assessment: Breast and Ovarian, Version 2.2017. 36. Antoniou A, Pharoah PDP, Narod S, et al. Average 57. Foulkes WD, Stefansson IM, Chappuis PO, et al.
J Natl Compr Canc Netw. 2017;15(1):9–20. risks of breast and ovarian cancer associated with Germline BRCA1 mutations and a basal epithelial
14. Johnston JJ, Rubinstein WS, Facio FM, et al. BRCA1 or BRCA2 mutations detected in case series phenotype in breast cancer. J Natl Cancer Inst. 2003;
Secondary variants in individuals undergoing exome unselected for family history: a combined analysis of 95(19):1482–1485.
sequencing: screening of 572 individuals identifies 22 studies. Am J Hum Genet. 2003;72(5):1117–1130. 58. Bignon YJ, Fonck Y, Chassagne MC. Histo-
high-penetrance mutations in cancer-susceptibility 37. Pharoah PD, Guilford P, Caldas C. International prognostic grade in tumours from families with
genes. Am J Hum Genet. 2012;91(1):97–108. Gastric Cancer Linkage Consortium. Incidence hereditary predisposition to breast cancer. Lancet.
15. Wolf SM, Crock BN, Van Ness B, et al. Managing of gastric cancer and breast cancer in CDH1 1995;346(8969):258.
incidental findings and research results in genomic (E-cadherin) mutation carriers from hereditary 59. Jacquemier J, Eisinger F, Birnbaum D, Sobol H.
research involving biobanks and archived data sets. diffuse gastric cancer families. Gastroenterology. Histoprognostic grade in BRCA1-associated breast
Genet Med. 2012;14(4):361–384. 2001;121(6):1348–1353. cancer. Lancet. 1995;345(8963):1503.
16. Pon JR, Marra MA. Driver and passenger mutations 38. Narod SA, Offit K. Prevention and manage- 60. Marcus JN, Watson P, Page DL, et al. Hereditary
in cancer. Annu Rev Pathol. 2015;10:25–50. ment of hereditary breast cancer. J Clin Oncol. breast cancer: pathobiology, prognosis, and BRCA1
17. Easton DF, Pharoah PD, Antoniou AC, et al. Gene- 2005;23(8):1656–1663. and BRCA2 gene linkage. Cancer. 1996;77(4):
panel sequencing and the prediction of breast-cancer 39. Robson M, Offit K. Clinical practice. Management 697–709.
risk. N Engl J Med. 2015;372(23):2243–2257. of an inherited predisposition to breast cancer. N 61. Deleted in review.
18. Domchek SM, Bradbury A, Garber JE, Offit K, Engl J Med. 2007;357(2):154–162. 62. Thompson D, Duedal S, Kirner J, et al. Cancer
Robson ME. Multiplex genetic testing for cancer 40. Claus EB, Risch N, Thompson WD. Genetic analysis risks and mortality in heterozygous ATM mutation
susceptibility: out on the high wire without a net? of breast cancer in the cancer and steroid hormone carriers. J Natl Cancer Inst. 2005;97(11):813–822.
J Clin Oncol. 2013;31(10):1267–1270. study. Am J Hum Genet. 1991;48(2):232–242. 63. CHEK2*1100delC and susceptibility to breast
19. Stadler ZK, Schrader KA, Vijai J, Robson ME, Offit 41. Whittemore AS, Gong G, Itnyre J. Prevalence cancer: a collaborative analysis involving 10,860
K. Cancer genomics and inherited risk. J Clin Oncol. and contribution of BRCA1 mutations in breast breast cancer cases and 9,065 controls from 10
2014;32(7):687–698. cancer and ovarian cancer: results from three U.S. studies. Am J Hum Genet. 2004;74(6):1175–1182.
20. Mavaddat N, Pharoah PD, Michailidou K, et al. population-based case-control studies of ovarian 64. Meijers-Heijboer H, van den Ouweland A, Klijn J,
Prediction of breast cancer risk based on profiling cancer. Am J Hum Genet. 1997;60(3):496–504. et al. Low-penetrance susceptibility to breast cancer
with common genetic variants. J Natl Cancer Inst. 42. Ford D, Easton DF. The genetics of breast and due to CHEK2(*)1100delC in noncarriers of BRCA1
2015;107(5). ovarian cancer. Br J Cancer. 1995;72(4):805–812. or BRCA2 mutations. Nat Genet. 2002;31(1):55–59.
21. Kuchenbaecker KB, McGuffog L, Barrowdale D, 43. Loveday C, Turnbull C, Ramsay E, et al. Germline 65. Weischer M, Nordestgaard BG, Pharoah P, et al.
et al. Evaluation of polygenic risk scores for breast mutations in RAD51D confer susceptibility to CHEK2*1100delC heterozygosity in women with
and ovarian cancer risk prediction in BRCA1 and ovarian cancer. Nat Genet. 2011;43(9):879–882. breast cancer associated with early death, breast
208.e2 Part I: Science and Clinical Oncology

cancer-specific death, and increased risk of a second 87. Offit K. BRCA mutation frequency and penetrance: 107. Erkko H, Xia B, Nikkilä J, et al. A recurrent muta-
breast cancer. J Clin Oncol. 2012;30(35):4308–4316. new data, old debate. J Natl Cancer Inst. 2006;98(23): tion in PALB2 in Finnish cancer families. Nature.
66. Kilpivaara O, Vahteristo P, Falck J, et al. CHEK2 1675–1677. 2007;446(7133):316–319.
variant I157T may be associated with increased breast 88. Offit K, Levran O, Mullaney B, et al. Shared genetic 108. Adank MA, Verhoef S, Oldenburg RA, et al. Excess
cancer risk. Int J Cancer. 2004;111(4):543–547. susceptibility to breast cancer, brain tumors, and breast cancer risk in first degree relatives of CHEK2
67. Offit K, Pierce H, Kirchhoff T, et al. Frequency of Fanconi anemia. J Natl Cancer Inst. 2003;95(20): *1100delC positive familial breast cancer cases. Eur
CHEK2*1100delC in New York breast cancer cases 1548–1551. J Cancer. 2013;49(8):1993–1999.
and controls. BMC Med Genet. 2003;4:1. 89. Zhang J, Walsh MF, Wu G, et al. Germline muta- 109. Han FF, Guo CL, Liu LH. The effect of CHEK2
68. Cybulski C, Wokołorczyk D, Jakubowska A, et al. tions in predisposition genes in pediatric cancer. N variant I157T on cancer susceptibility: evidence from
Risk of breast cancer in women with a CHEK2 Engl J Med. 2015;373(24):2336–2346. a meta-analysis. DNA Cell Biol. 2013;32(6):329–335.
mutation with and without a family history of breast 90. Parsons DW, Roy A, Yang Y, et al. Diagnostic yield of 110. Renwick A, Thompson D, Seal S, et al. ATM
cancer. J Clin Oncol. 2011;29(28):3747–3752. clinical tumor and germline whole-exome sequencing mutations that cause ataxia-telangiectasia are
69. Chenevix-Trench G, Spurdle AB, Gatei M, et al. for children with solid tumors. JAMA Oncol. 2016. breast cancer susceptibility alleles. Nat Genet.
Dominant negative ATM mutations in breast cancer 91. Walsh MF, Kennedy J, Harlan M, et al. Germ line 2006;38(8):873–875.
families. J Natl Cancer Inst. 2002;94(3):205–215. BRCA2 mutations detected in pediatric sequencing 111. Paglia LL, Lauge A, Weber J, et al. ATM germline
70. Ahmed M, Rahman N. ATM and breast cancer studies impact parents’ evaluation and care. Cold mutations in women with familial breast cancer and
susceptibility. Oncogene. 2006;25(43):5906–5911. Spring Harb Mol Case Stud. 2017. a relative with haematological malignancy. Breast
71. Balmain A, Gray J, Ponder B. The genetics and genom- 92. Casilli F, Tournier I, Sinilnikova OM, et al. The con- Cancer Res Treat. 2010;119(2):443–452.
ics of cancer. Nat Genet. 2003;33(suppl):238–244. tribution of germline rearrangements to the spectrum 112. Rafnar T, Gudbjartsson DF, Sulem P, et al. Mutations
72. Hunter DJ, Kraft P, Jacobs KB, et al. A genome- of BRCA2 mutations. J Med Genet. 2006;43(9): in BRIP1 confer high risk of ovarian cancer. Nat
wide association study identifies alleles in FGFR2 e49. Genet. 2011;43(11):1104–1107.
associated with risk of sporadic postmenopausal 93. Woodward AM, Davis TA, Silva AGS, Kirk JA, 113. Ramus SJ, Song H, Dicks E, et al. Germline muta-
breast cancer. Nat Genet. 2007;39(7):870–874. Leary JA, kConFab I. Large genomic rearrangements tions in the BRIP1, BARD1, PALB2, and NBN genes
73. Easton DF, Pooley KA, Dunning AM, et al. Genome- of both BRCA2 and BRCA1 are a feature of the in women with ovarian cancer. J Natl Cancer Inst.
wide association study identifies novel breast inherited breast/ovarian cancer phenotype in selected 2015;107(11).
cancer susceptibility loci. Nature. 2007;447(7148): families. J Med Genet. 2005;42(5):e31. 114. Song H, Dicks E, Ramus SJ, et al. Contribution of
1087–1093. 94. Peshkin BN, DeMarco TA, Brogan BM, Lerman C, germline mutations in the RAD51B, RAD51C, and
74. Tung N, Battelli C, Allen B, et al. Frequency Isaacs C. BRCA1/2 testing: complex themes in result RAD51D genes to ovarian cancer in the population.
of mutations in individuals with breast cancer interpretation. J Clin Oncol. 2001;19(9):2555–2565. J Clin Oncol. 2015;33(26):2901–2907.
referred for BRCA1 and BRCA2 testing using 95. Stoppa-Lyonnet D, Laurent-Puig P, Essioux L, et al. 115. Nelson HD, Huffman LH, Fu R, Harris EL,
next-generation sequencing with a 25-gene panel. BRCA1 sequence variations in 160 individuals referred Force USPST. Genetic risk assessment and BRCA
Cancer. 2015;121(1):25–33. to a breast/ovarian family cancer clinic. Institut Curie mutation testing for breast and ovarian cancer
75. Kuchenbaecker KB, Hopper JL, Barnes DR, et al. Breast Cancer Group. Am J Hum Genet. 1997; susceptibility: systematic evidence review for the
Risks of breast, ovarian, and contralateral breast 60(5):1021–1030. U.S. Preventive Services Task Force. Ann Intern Med.
cancer for BRCA1 and BRCA2 mutation carriers. 96. Venkitaraman AR. Cancer susceptibility and the 2005;143(5):362–379.
JAMA. 2017;317(23):2402–2416. functions of BRCA1 and BRCA2. Cell. 2002;108(2): 116. National Comprehensive Cancer Centers. NCCN
76. Köbel M, Kalloger SE, Boyd N, et al. Ovarian 171–182. Clinical Practive Guidelines in Oncology, Genetic/
carcinoma subtypes are different diseases: implica- 97. Offit K, Gilewski T, McGuire P, et al. Germline Familial High-Risk Assessment: Breast and Ovarian
tions for biomarker studies. PLoS Med. 2008;5(12). BRCA1 185delAG mutations in Jewish women with Version 2.2017; 2016. http://www.nccn.org/profes-
77. Gilks CB, Prat J. Ovarian carcinoma pathology breast cancer. Lancet. 1996;347(9016):1643–1645. sionals/physician_gls/pdf/genetics_screening.pdf.
and genetics: recent advances. Hum Pathol. 98. Roa BB, Boyd AA, Volcik K, Richards CS. Ash- 117. Robson ME, Offit K. Breast MRI for women
2009;40(9):1213–1223. kenazi Jewish population frequencies for common with hereditary cancer risk. JAMA. 2004;292(11):
78. Kalloger SE, Köbel M, Leung S, et al. Calculator for mutations in BRCA1 and BRCA2. Nat Genet. 1368–1370.
ovarian carcinoma subtype prediction. Mod Pathol. 1996;14(2):185–187. 118. Ziegler LD, Kroll SS. Primary breast cancer
2011;24(4):512–521. 99. Neuhausen S, Gilewski T, Norton L, et al. Recurrent after prophylactic mastectomy. Am J Clin Oncol.
79. Alsop K, Fereday S, Meldrum C, et al. BRCA muta- BRCA2 6174delT mutations in Ashkenazi Jewish 1991;14(5):451–454.
tion frequency and patterns of treatment response in women affected by breast cancer. Nat Genet. 1996; 119. Hartmann LC, Sellers TA, Schaid DJ, et al. Efficacy
BRCA mutation-positive women with ovarian cancer: 13(1):126–128. of bilateral prophylactic mastectomy in BRCA1 and
a report from the Australian Ovarian Cancer Study 100. Struewing JP, Abeliovich D, Peretz T, et al. The BRCA2 gene mutation carriers. J Natl Cancer Inst.
Group. J Clin Oncol. 2012;30(21):2654–2663. carrier frequency of the BRCA1 185delAG mutation 2001;93(21):1633–1637.
80. Press JZ, De Luca A, Boyd N, et al. Ovarian is approximately 1 percent in Ashkenazi Jewish 120. Rebbeck TR, Kauff ND, Domchek SM. Meta-
carcinomas with genetic and epigenetic BRCA1 individuals. Nat Genet. 1995;11(2):198–200. analysis of risk reduction estimates associated with
loss have distinct molecular abnormalities. BMC 101. Levy-Lahad E, Catane R, Eisenberg S, et al. Founder risk-reducing salpingo-oophorectomy in BRCA1
Cancer. 2008;8:17. BRCA1 and BRCA2 mutations in Ashkenazi Jews or BRCA2 mutation carriers. J Natl Cancer Inst.
81. McAlpine JN, Porter H, Köbel M, et al. BRCA1 in Israel: frequency and differential penetrance in 2009;101(2):80–87.
and BRCA2 mutations correlate with TP53 abnor- ovarian cancer and in breast-ovarian cancer families. 121. Rebbeck TR, Friebel T, Lynch HT, et al. Bilateral
malities and presence of immune cell infiltrates in Am J Hum Genet. 1997;60(5):1059–1067. prophylactic mastectomy reduces breast cancer risk in
ovarian high-grade serous carcinoma. Mod Pathol. 102. Kauff ND, Perez-Segura P, Robson ME, et al. BRCA1 and BRCA2 mutation carriers: the PROSE
2012;25(5):740–750. Incidence of non-founder BRCA1 and BRCA2 Study Group. J Clin Oncol. 2004;22(6):1055–1062.
82. Schrader KA, Hurlburt J, Kalloger SE, et al. mutations in high risk Ashkenazi breast and ovarian 122. Kauff ND, Domchek SM, Friebel TM, et al.
Germline BRCA1 and BRCA2 mutations in ovarian cancer families. J Med Genet. 2002;39(8):611– Risk-reducing salpingo-oophorectomy for the
cancer: utility of a histology-based referral strategy. 614. prevention of BRCA1- and BRCA2-associated breast
Obstet Gynecol. 2012;120(2 Pt 1):235–240. 103. Walsh T, Mandell JB, Norquist BM, et al. Genetic and gynecologic cancer: a multicenter, prospective
83. Ryan NA, Evans DG, Green K, Crosbie EJ. predisposition to breast cancer due to mutations study. J Clin Oncol. 2008;26(8):1331–1337.
Pathological features and clinical behavior of Lynch other than BRCA1 and BRCA2 founder alleles 123. Vogel VG, Costantino JP, Wickerham DL,
syndrome-associated ovarian cancer. Gynecol Oncol. among Ashkenazi Jewish Women. JAMA Oncol. et al. Effects of tamoxifen vs raloxifene on the
2017;144(3):491–495. 2017. risk of developing invasive breast cancer and
84. Tung N, Domchek SM, Stadler Z, et  al. 104. Szabo CI, King MC. Population genetics of BRCA1 other disease outcomes: the NSABP Study of
Counselling framework for moderate-penetrance and BRCA2. Am J Hum Genet. 1997;60(5): Tamoxifen and Raloxifene (STAR) P-2 trial. JAMA.
cancer-susceptibility mutations. Nat Rev Clin Oncol. 1013–1020. 2006;295(23):2727–2741.
2016;13(9):581–588. 105. Khoo U-S, Chan KYK, Cheung ANY, et al. Recur- 124. Cuzick J, Sestak I, Forbes JF, et al. Anastrozole
85. Easton DF, Lesueur F, Decker B, et al. No evidence rent BRCA1 and BRCA2 germline mutations in for prevention of breast cancer in high-risk post-
that protein truncating variants in BRIP1 are ovarian cancer: a founder mutation of BRCA1 identi- menopausal women (IBIS-II): an international,
associated with breast cancer risk: implications for fied in the Chinese population. Hum Mutat. 2002; double-blind, randomised placebo-controlled trial.
gene panel testing. J Med Genet. 2016;53(5):298– 19(3):307–308. Lancet. 2014;383(9922):1041–1048.
309. 106. Thorlacius S, Sigurdsson S, Bjarnadottir H, et al. 125. Goss PE, Ingle JN, Ales-Martinez JE, et al. Exemes-
86. Norquist BM, Harrell MI, Brady MF, et al. Inherited Study of a single BRCA2 mutation with high carrier tane for breast-cancer prevention in postmenopausal
mutations in women with ovarian carcinoma. JAMA frequency in a small population. Am J Hum Genet. women. N Engl J Med. 2011;364(25):2381–
Oncol. 2016;2(4):482–490. 1997;60(5):1079–1084. 2391.
Genetic Factors: Hereditary Cancer Predisposition Syndromes  •  CHAPTER 13 208.e3
208.e3

126. Narod SA, Brunet JS, Ghadirian P, et al. Tamoxifen Hereditary Ovarian Cancer Clinical Study Group. pigmentation, and endocrine overactivity. Medicine
and risk of contralateral breast cancer in BRCA1 N Engl J Med. 1998;339(7):424–428. (Baltimore). 1985;64(4):270–283.
and BRCA2 mutation carriers: a case-control study. 144. Narod SA, Sun P, Risch HA. Hereditary Ovarian 164. Cannon-Albright LA, Skolnick MH, Bishop DT, Lee
Hereditary Breast Cancer Clinical Study Group. Cancer Clinical Study Group. Ovarian cancer, oral RG, Burt RW. Common inheritance of susceptibil-
Lancet. 2000;356(9245):1876–1881. contraceptives, and BRCA mutations. N Engl J Med. ity to colonic adenomatous polyps and associated
127. King MC, Wieand S, Hale K, et al. Tamoxifen 2001;345(23):1706–1707. colorectal cancers. N Engl J Med. 1988;319(9):
and breast cancer incidence among women with 145. Narod SA, Dubé M-P, Klijn J, et al. Oral contra- 533–537.
inherited mutations in BRCA1 and BRCA2: ceptives and the risk of breast cancer in BRCA1 165. Houlston RS, Collins A, Slack J, Morton NE.
National Surgical Adjuvant Breast and Bowel Project and BRCA2 mutation carriers. J Natl Cancer Inst. Dominant genes for colorectal cancer are not rare.
(NSABP-P1) Breast Cancer Prevention Trial. JAMA. 2002;94(23):1773–1779. Ann Hum Genet. 1992;56(Pt 2):99–103.
2001;286(18):2251–2256. 146. Farmer H, McCabe N, Lord CJ, et al. Targeting the 166. Marra G, Boland CR. Hereditary nonpolyposis
128. Gronwald J, Tung N, Foulkes WD, et al. Tamoxifen DNA repair defect in BRCA mutant cells as a thera- colorectal cancer: the syndrome, the genes, and his-
and contralateral breast cancer in BRCA1 and BRCA2 peutic strategy. Nature. 2005;434(7035):917–921. torical perspectives. J Natl Cancer Inst. 1995;87(15):
carriers: an update. Int J Cancer. 2006;118(9): 147. Bryant HE, Schultz N, Thomas HD, et al. 1114–1125.
2281–2284. Specific killing of BRCA2-deficient tumours with 167. Hampel H, Frankel WL, Martin E, et  al.
129. Visvanathan K, Hurley P, Bantug E, et al. Use of inhibitors of poly(ADP-ribose) polymerase. Nature. Screening for the Lynch syndrome (hereditary
pharmacologic interventions for breast cancer risk 2005;434(7035):913–917. nonpolyposis colorectal cancer). N Engl J Med.
reduction: American Society of Clinical Oncology 148. Swisher EM, Lin KK, Oza AM, et al. Rucaparib in 2005;352(18):1851–1860.
clinical practice guideline. J Clin Oncol. 2013;31(23): relapsed, platinum-sensitive high-grade ovarian carci- 168. Hampel H, Stephens JA, Pukkala E, et al. Cancer
2942–2962. noma (ARIEL2 Part 1): an international, multicentre, risk in hereditary nonpolyposis colorectal cancer
130. Bourne TH, Campbell S, Reynolds KM, et al. open-label, phase 2 trial. Lancet Oncol. 2017;18(1): syndrome: later age of onset. Gastroenterology.
Screening for early familial ovarian cancer with 75–87. 2005;129(2):415–421.
transvaginal ultrasonography and colour blood 149. Mirza MR, Monk BJ, Herrstedt J, et  al. 169. Bailey-Wilson JE, Elston RC, Schuelke GS, et al.
flow imaging. BMJ. 1993;306(6884):1025–1029. Niraparib maintenance therapy in platinum- Segregation analysis of hereditary nonpolyposis
131. Scheuer L, Kauff N, Robson M, et al. Outcome sensitive, recurrent ovarian cancer. N Engl J Med. colorectal cancer. Genet Epidemiol. 1986;3(1):27–38.
of preventive surgery and screening for breast and 2016;375(22):2154–2164. 170. Scapoli C, Ponz De Leon M, Sassatelli R, et al.
ovarian cancer in BRCA mutation carriers. J Clin 150. Kaufman B, Shapira-Frommer R, Schmutzler Genetic epidemiology of hereditary non-polyposis
Oncol. 2002;20(5):1260–1268. RK, et al. Olaparib monotherapy in patients with colorectal cancer syndromes in Modena, Italy: results
132. Menon U, Ryan A, Kalsi J, et al. Risk algorithm advanced cancer and a germline BRCA1/2 mutation. of a complex segregation analysis. Ann Hum Genet.
using serial biomarker measurements doubles the J Clin Oncol. 2015;33(3):244–250. 1994;58(Pt 3):275–295.
number of screen-detected cancers compared with 151. Robson M, Im SA, Senkus E, et al. Olaparib for 171. Vasen HF, Taal BG, Griffioen G, et al. Clinical
a single-threshold rule in the United Kingdom metastatic breast cancer in patients with a germline heterogeneity of familial colorectal cancer and its
Collaborative Trial of Ovarian Cancer Screening. BRCA mutation. N Engl J Med. 2017. influence on screening protocols. Gut. 1994;35(9):
J Clin Oncol. 2015;33(18):2062–2071. 152. Eng C. Will the real Cowden syndrome please 1262–1266.
133. U.S. Food and Drug Administration. The FDA stand up: revised diagnostic criteria. J Med Genet. 172. Dunlop MG, Farrington SM, Carothers AD,
recommends against using screening tests for ovarian 2000;37(11):828–830. et al. Cancer risk associated with germline DNA
cancer screening: FDA Safety Communication; 153. Zhou X-P, Marsh DJ, Morrison CD, et al. Germline mismatch repair gene mutations. Hum Mol Genet.
2016. https://www.fda.gov/MedicalDevices/Safety/ inactivation of PTEN and dysregulation of the 1997;6(1):105–110.
AlertsandNotices/ucm519413.htm. phosphoinositol-3-kinase/Akt pathway cause human 173. Aarnio M, Sankila R, Pukkala E, et al. Cancer risk
134. Medeiros F, Muto MG, Lee Y, et al. The tubal fimbria Lhermitte-Duclos disease in adults. Am J Hum Genet. in mutation carriers of DNA-mismatch-repair genes.
is a preferred site for early adenocarcinoma in women 2003;73(5):1191–1198. Int J Cancer. 1999;81(2):214–218.
with familial ovarian cancer syndrome. Am J Surg 154. Eng C, Murday V, Seal S, et al. Cowden syndrome 174. Watson P, Lynch HT. Cancer risk in mismatch repair
Pathol. 2006;30(2):230–236. and Lhermitte-Duclos disease in a family: a single gene mutation carriers. Fam Cancer. 2001;1(1):
135. Lee Y, Miron A, Drapkin R, et al. A candidate genetic syndrome with pleiotropy? J Med Genet. 57–60.
precursor to serous carcinoma that originates in the 1994;31(6):458–461. 175. Lu KH, Dinh M, Kohlmann W, et al. Gynecologic
distal fallopian tube. J Pathol. 2007;211(1):26–35. 155. Hanssen AM, Fryns JP. Cowden syndrome. J Med cancer as a “sentinel cancer” for women with heredi-
136. Finch A, Shaw P, Rosen B, Murphy J, Narod S, Genet. 1995;32(2):117–119. tary nonpolyposis colorectal cancer syndrome. Obstet
Colgan T. Clinical and pathologic findings of 156. Fackenthal JD, Marsh DJ, Richardson AL, et al. Male Gynecol. 2005;105(3):569–574.
prophylactic salpingo-oophorectomies in 159 breast cancer in Cowden syndrome patients with 176. Watson P, Vasen HF, Mecklin JP, Järvinen H,
BRCA1 and BRCA2 carriers. Gynecol Oncol. germline PTEN mutations. J Med Genet. 2001;38(3): Lynch HT. The risk of endometrial cancer in
2006;100(1):58–64. 159–164. hereditary nonpolyposis colorectal cancer. Am J
137. Kauff ND, Satagopan JM, Robson ME, et al. 157. Riegert-Johnson DL, Gleeson FC, Roberts M, et al. Med. 1994;96(6):516–520.
Risk-reducing salpingo-oophorectomy in women Cancer and Lhermitte-Duclos disease are common 177. Lynch HT, Lynch PM, Lanspa SJ, Snyder CL, Lynch
with a BRCA1 or BRCA2 mutation. N Engl J Med. in Cowden syndrome patients. Hered Cancer Clin JF, Boland CR. Review of the Lynch syndrome:
2002;346(21):1609–1615. Pract. 2010;8(1):6. history, molecular genetics, screening, differential
138. Rebbeck TR, Lynch HT, Neuhausen SL, et al. 158. Tan M-H, Mester JL, Ngeow J, Rybicki LA, Orloff MS, diagnosis, and medicolegal ramifications. Clin Genet.
Prophylactic oophorectomy in carriers of BRCA1 Eng C. Lifetime cancer risks in individuals with germ- 2009;76(1):1–18.
or BRCA2 mutations. N Engl J Med. 2002;346(21): line PTEN mutations. Clin Cancer Res. 2012;18(2): 178. Watson P, Lynch HT. Extracolonic cancer in
1616–1622. 400–407. hereditary nonpolyposis colorectal cancer. Cancer.
139. Domchek SM, Friebel TM, Neuhausen SL, et al. 159. Li J, Yen C, Liaw D, et al. PTEN, a putative 1993;71(3):677–685.
Mortality after bilateral salpingo-oophorectomy in protein tyrosine phosphatase gene mutated in 179. Raymond VM, Everett JN, Furtado LV, et al. Adre-
BRCA1 and BRCA2 mutation carriers: a prospec- human brain, breast, and prostate cancer. Science. nocortical carcinoma is a lynch syndrome-associated
tive cohort study. Lancet Oncol. 2006;7(3):223– 1997;275(5308):1943–1947. cancer. J Clin Oncol. 2013;31(24):3012–3018.
229. 160. Marsh DJ, Kum JB, Lunetta KL, et al. PTEN muta- 180. Raymond VM, Mukherjee B, Wang F, et al.
140. Struewing JP, Watson P, Easton DF, Ponder BA, tion spectrum and genotype-phenotype correlations Elevated risk of prostate cancer among men with
Lynch HT, Tucker MA. Prophylactic oophorectomy in Bannayan-Riley-Ruvalcaba syndrome suggest a Lynch syndrome. J Clin Oncol. 2013;31(14):1713–
in inherited breast/ovarian cancer families. J Natl single entity with Cowden syndrome. Hum Mol 1718.
Cancer Inst. 1995;17:33–35. Genet. 1999;8(8):1461–1472. 181. Win AK, Young JP, Lindor NM, et al. Colorectal
141. Chen KT, Schooley JL, Flam MS. Peritoneal 161. Eng C. Role of PTEN, a lipid phosphatase upstream and other cancer risks for carriers and noncarriers
carcinomatosis after prophylactic oophorectomy in effector of protein kinase B, in epithelial thyroid from families with a DNA mismatch repair gene
familial ovarian cancer syndrome. Obstet Gynecol. carcinogenesis. Ann N Y Acad Sci. 2002;968:213– mutation: a prospective cohort study. J Clin Oncol.
1985;66(suppl 3):93S–94S. 221. 2012;30(9):958–964.
142. Modan B, Hartge P, Hirsh-Yechezkel G, et al. 162. Shiovitz S, Everett J, Huang S-C, Orloff MS, Eng 182. Vasen HF, Watson P, Mecklin JP, Lynch HT. New
Parity, oral contraceptives, and the risk of ovarian C, Gruber SB. Head circumference in the clinical clinical criteria for hereditary nonpolyposis colorectal
cancer among carriers and noncarriers of a BRCA1 detection of PTEN hamartoma tumor syndrome cancer (HNPCC, Lynch syndrome) proposed by
or BRCA2 mutation. N Engl J Med. 2001;345(4): in a clinic population at high-risk of breast cancer. the International Collaborative group on HNPCC.
235–240. Breast Cancer Res Treat. 2010;124(2):459–465. Gastroenterology. 1999;116(6):1453–1456.
143. Narod SA, Risch H, Moslehi R, et al. Oral contra- 163. Carney JA, Gordon H, Carpenter PC, Shenoy 183. Rodriguez-Bigas MA, Boland CR, Hamilton SR,
ceptives and the risk of hereditary ovarian cancer. BV, Go VL. The complex of myxomas, spotty et al. A National Cancer Institute Workshop on
208.e4 Part I: Science and Clinical Oncology

Hereditary Nonpolyposis Colorectal Cancer Syn- in hMLH1-deficient children. Cancer Res. common in familial adenomatous polyposis. Clin
drome: meeting highlights and Bethesda Guidelines. 1999;59(2):294–297. Gastroenterol Hepatol. 2008;6(2):180–185.
J Natl Cancer Inst. 1997;89(23):1758–1762. 205. Van Galen P, Perrier R, Bernier FP. Late presentation 226. Lynch HT, Snyder C, Davies JM, et al. FAP, gastric
184. Umar A, Risinger JI, Hawk ET, Barrett JC. Testing of cancer in compound heterozygote PMS2 mutation cancer, and genetic counseling featuring children
guidelines for hereditary non-polyposis colorectal carrier. Hered Cancer Clin Pract. 2011;9(suppl 1): and young adults: a family study and review. Fam
cancer. Nat Rev Cancer. 2004;4(2):153–158. P38. Cancer. 2010;9(4):581–588.
185. Barnetson RA, Tenesa A, Farrington SM, et al. 206. Durno CA, Sherman PM, Aronson M, et al. Phe- 227. Groves CJ, Saunders BP, Spigelman AD, Phillips
Identification and survival of carriers of mutations notypic and genotypic characterisation of biallelic RKS. Duodenal cancer in patients with familial
in DNA mismatch-repair genes in colon cancer. mismatch repair deficiency (BMMR-D) syndrome. adenomatous polyposis (FAP): results of a 10 year
N Engl J Med. 2006;354(26):2751–2763. Eur J Cancer. 2015;51(8):977–983. prospective study. Gut. 2002;50(5):636–641.
186. Chen S, Wang W, Lee S, et al. Prediction of germline 207. Järvinen HJ, Aarnio M, Mustonen H, et al. Con- 228. Half E, Bercovich D, Rozen P. Familial adenomatous
mutations and cancer risk in the Lynch syndrome. trolled 15-year trial on screening for colorectal cancer polyposis. Orphanet J Rare Dis. 2009;4:22.
JAMA. 2006;296(12):1479–1487. in families with hereditary nonpolyposis colorectal 229. Kinzler KW, Nilbert MC, Su LK, et al. Identification
187. Balmaña J, Stockwell DH, Steyerberg EW, et al. cancer. Gastroenterology. 2000;118(5):829–834. of FAP locus genes from chromosome 5q21. Science.
Prediction of MLH1 and MSH2 mutations in Lynch 208. Renkonen-Sinisalo L, Aarnio M, Mecklin JP, Järvinen 1991;253(5020):661–665.
syndrome. JAMA. 2006;296(12):1469–1478. HJ. Surveillance improves survival of colorectal cancer 230. Vasen HFA, Möslein G, Alonso A, et al. Guidelines
188. Umar A, Boland CR, Terdiman JP, et al. Revised in patients with hereditary nonpolyposis colorectal for the clinical management of familial adenomatous
Bethesda Guidelines for hereditary nonpolyposis cancer. Cancer Detect Prev. 2000;24(2):137–142. polyposis (FAP). Gut. 2008;57(5):704–713.
colorectal cancer (Lynch syndrome) and microsatellite 209. Lindor NM, Petersen GM, Hadley DW, et al. 231. Bisgaard ML, Fenger K, Bülow S, Niebuhr E,
instability. J Natl Cancer Inst. 2004;96(4):261–268. Recommendations for the care of individuals with Mohr J. Familial adenomatous polyposis (FAP):
189. Recommendations from the EGAPP Working an inherited predisposition to Lynch syndrome: a frequency, penetrance, and mutation rate. Hum
Group: genetic testing strategies in newly diagnosed systematic review. JAMA. 2006;296(12):1507–1517. Mutat. 1994;3(2):121–125.
individuals with colorectal cancer aimed at reducing 210. Järvinen HJ, Aarnio M. Surveillance on mutation 232. Aretz S, Stienen D, Friedrichs N, et al. Somatic APC
morbidity and mortality from Lynch syndrome in carriers of DNA mismatch repair genes. Ann Chir mosaicism: a frequent cause of familial adenomatous
relatives. Genet Med. 2009;11(1):35–41. Gynaecol. 2000;89(3):207–210. polyposis (FAP). Hum Mutat. 2007;28(10):985–992.
190. National Comprehensive Cancer Centers. NCCN 211. Syngal S, Weeks JC, Schrag D, Garber JE, Kuntz 233. Nieuwenhuis MH, Vasen HFA. Correlations between
Clinical Practice Guidelines in Oncology, Genetic/ KM. Benefits of colonoscopic surveillance and mutation site in APC and phenotype of familial
Familial High-Risk Assessment: Colorectal Version prophylactic colectomy in patients with hereditary adenomatous polyposis (FAP): a review of the litera-
1.2017; 2017. https://www.nccn.org/professionals/ nonpolyposis colorectal cancer mutations. Ann Intern ture. Crit Rev Oncol Hematol. 2007;61(2):153–161.
physician_gls/pdf/genetics_colon.pdf. Med. 1998;129(10):787–796. 234. Bertario L, Russo A, Sala P, et al. Multiple approach
191. Leach FS, Nicolaides NC, Papadopoulos N, et al. 212. Schmeler KM, Lynch HT, Chen L-M, et al. Prophy- to the exploration of genotype-phenotype correla-
Mutations of a mutS homolog in hereditary nonpol- lactic surgery to reduce the risk of gynecologic cancers tions in familial adenomatous polyposis. J Clin Oncol.
yposis colorectal cancer. Cell. 1993;75(6):1215–1225. in the Lynch syndrome. N Engl J Med. 2006;354(3): 2003;21(9):1698–1707.
192. Bronner CE, Baker SM, Morrison PT, et al. Muta- 261–269. 235. Spirio L, Olschwang S, Groden J, et al. Alleles of the
tion in the DNA mismatch repair gene homologue 213. Offit K, Kauff ND. Reducing the risk of gynecologic APC gene: an attenuated form of familial polyposis.
hMLH1 is associated with hereditary non-polyposis cancer in the Lynch syndrome. N Engl J Med. 2006; Cell. 1993;75(5):951–957.
colon cancer. Nature. 1994;368(6468):258–261. 354(3):293–295. 236. Sieber OM, Lipton L, Crabtree M, et al. Multiple
193. Liu B, Parsons R, Papadopoulos N, et al. Analysis of 214. Parry S, Win AK, Parry B, et al. Metachronous colorectal adenomas, classic adenomatous polyposis,
mismatch repair genes in hereditary non-polyposis colorectal cancer risk for mismatch repair gene and germ-line mutations in MYH. N Engl J Med.
colorectal cancer patients. Nat Med. 1996;2(2): mutation carriers: the advantage of more extensive 2003;348(9):791–799.
169–174. colon surgery. Gut. 2011;60(7):950–957. 237. Offit K, Kohut K, Clagett B, et al. Cancer genetic
194. Wijnen JT, Vasen HF, Khan PM, et al. Clinical 215. Gryfe R, Kim H, Hsieh ET, et al. Tumor microsatel- testing and assisted reproduction. J Clin Oncol.
findings with implications for genetic testing in lite instability and clinical outcome in young patients 2006;24(29):4775–4782.
families with clustering of colorectal cancer. N Engl with colorectal cancer. N Engl J Med. 2000;342(2): 238. Giardiello FM, Hamilton SR, Krush AJ, et al. Treat-
J Med. 1998;339(8):511–518. 69–77. ment of colonic and rectal adenomas with sulindac
195. Liu T, Yan H, Kuismanen S, et al. The role of hPMS1 216. Popat S, Hubner R, Houlston RS. Systematic review in familial adenomatous polyposis. N Engl J Med.
and hPMS2 in predisposing to colorectal cancer. of microsatellite instability and colorectal cancer 1993;328(18):1313–1316.
Cancer Res. 2001;61(21):7798–7802. prognosis. J Clin Oncol. 2005;23(3):609–618. 239. Nugent KP, Farmer KC, Spigelman AD, Williams
196. Ligtenberg MJL, Kuiper RP, Chan TL, et al. 217. Sargent DJ, Marsoni S, Monges G, et al. Defective CB, Phillips RK. Randomized controlled trial
Heritable somatic methylation and inactivation of mismatch repair as a predictive marker for lack of of the effect of sulindac on duodenal and rectal
MSH2 in families with Lynch syndrome due to efficacy of fluorouracil-based adjuvant therapy in polyposis and cell proliferation in patients with
deletion of the 3′ exons of TACSTD1. Nat Genet. colon cancer. J Clin Oncol. 2010;28(20):3219–3226. familial adenomatous polyposis. Br J Surg. 1993;
2009;41(1):112–117. 218. Ribic CM, Sargent DJ, Moore MJ, et al. Tumor 80(12):1618–1619.
197. Papadopoulos N, Lindblom A. Molecular basis of microsatellite-instability status as a predictor of 240. Steinbach G, Lynch PM, Phillips RK, et al. The
HNPCC: mutations of MMR genes. Hum Mutat. benefit from fluorouracil-based adjuvant chemo- effect of celecoxib, a cyclooxygenase-2 inhibitor,
1997;10(2):89–99. therapy for colon cancer. N Engl J Med. 2003;349(3): in familial adenomatous polyposis. N Engl J Med.
198. Offit K. MSH6 mutations in hereditary nonpolyposis 247–257. 2000;342(26):1946–1952.
colon cancer: another slice of the pie. J Clin Oncol. 219. Le DT, Uram JN, Wang H, et al. PD-1 Blockade 241. Parc YR, Olschwang S, Desaint B, Schmitt G, Parc
2004;22(22):4449–4451. in tumors with mismatch-repair deficiency. N Engl RG, Tiret E. Familial adenomatous polyposis: preva-
199. Aaltonen LA, Peltomäki P, Leach FS, et al. Clues J Med. 2015;372(26):2509–2520. lence of adenomas in the ileal pouch after restorative
to the pathogenesis of familial colorectal cancer. 220. Le DT, Durham JN, Smith KN, et al. Mismatch- proctocolectomy. Ann Surg. 2001;233(3):360–364.
Science. 1993;260(5109):812–816. repair deficiency predicts response of solid tumors 242. Guillem JG, Wood WC, Moley JF, et al. ASCO/
200. Thibodeau SN, Bren G, Schaid D. Microsatellite to PD-1 blockade. Science. 2017. SSO review of current role of risk-reducing surgery
instability in cancer of the proximal colon. Science. 221. Bouffet E, Larouche V, Campbell BB, et al. in common hereditary cancer syndromes. J Clin
1993;260(5109):816–819. Immune checkpoint inhibition for hypermutant Oncol. 2006;24(28):4642–4660.
201. Cunningham JM, Christensen ER, Tester DJ, et al. glioblastoma multiforme resulting from germline 243. King JE, Dozois RR, Lindor NM, Ahlquist DA. Care
Hypermethylation of the hMLH1 promoter in colon biallelic mismatch repair deficiency. J Clin Oncol. of patients and their families with familial adenoma-
cancer with microsatellite instability. Cancer Res. 2016;34(19):2206–2211. tous polyposis. Mayo Clin Proc. 2000;75(1):57–67.
1998;58(15):3455–3460. 222. Petersen GM, Slack J, Nakamura Y. Screening 244. Knudsen AL, Bisgaard ML, Bülow S. Attenuated
202. Cunningham JM, Kim CY, Christensen ER, et al. guidelines and premorbid diagnosis of familial familial adenomatous polyposis (AFAP). A review
The frequency of hereditary defective mismatch adenomatous polyposis using linkage. Gastroenterol- of the literature. Fam Cancer. 2003;2(1):43–55.
repair in a prospective series of unselected colorectal ogy. 1991;100(6):1658–1664. 245. Brensinger JD, Laken SJ, Luce MC, et al. Variable
carcinomas. Am J Hum Genet. 2001;69(4):780–790. 223. Galiatsatos P, Foulkes WD. Familial adenomatous phenotype of familial adenomatous polyposis in
203. Ricciardone MD, Ozçelik T, Cevher B, et al. Human polyposis. Am J Gastroenterol. 2006;101(2):385–398. pedigrees with 3′ mutation in the APC gene. Gut.
MLH1 deficiency predisposes to hematological 224. Campbell WJ, Spence RA, Parks TG. Familial adeno- 1998;43(4):548–552.
malignancy and neurofibromatosis type 1. Cancer matous polyposis. Br J Surg. 1994;81(12):1722– 246. Bussey HJ. Familial polyposis coli. Pathol Annu.
Res. 1999;59(2):290–293. 1733. 1979;14(Pt 1):61–81.
204. Wang Q, Lasset C, Desseigne F, et  al. 225. Bianchi LK, Burke CA, Bennett AE, Lopez R, Hasson 247. Al-Tassan N, Chmiel NH, Maynard J, et al.
Neurofibromatosis and early onset of cancers H, Church JM. Fundic gland polyp dysplasia is Inherited variants of MYH associated with somatic
Genetic Factors: Hereditary Cancer Predisposition Syndromes  •  CHAPTER 13 208.e5
208.e5

G:C–>T:A mutations in colorectal tumors. Nat neoplasia. N Engl J Med. 2010;363(27):2628– (FaPaCa): ten years experience. Fam Cancer. 2011;
Genet. 2002;30(2):227–232. 2637. 10(2):323–330.
248. Goodenberger M, Lindor NM. Lynch syndrome 267. Fitzgerald RC, Hardwick R, Huntsman D, et al. 288. Kastrinos F, Mukherjee B, Tayob N, et al. Risk of
and MYH-associated polyposis: review and testing Hereditary diffuse gastric cancer: updated consensus pancreatic cancer in families with Lynch syndrome.
strategy. J Clin Gastroenterol. 2011;45(6):488–500. guidelines for clinical management and directions for JAMA. 2009;302(16):1790–1795.
249. Nielsen M, Morreau H, Vasen HFA, Hes FJ. future research. J Med Genet. 2010;47(7):436–444. 289. Giardiello FM, Brensinger JD, Tersmette AC, et al.
MUTYH-associated polyposis (MAP). Crit Rev 268. Caldas C, Carneiro F, Lynch HT, et al. Familial Very high risk of cancer in familial Peutz-Jeghers
Oncol Hematol. 2011;79(1):1–16. gastric cancer: overview and guidelines for manage- syndrome. Gastroenterology. 2000;119(6):1447–1453.
250. Vogt S, Jones N, Christian D, et al. Expanded ment. J Med Genet. 1999;36(12):873–880. 290. Lynch HT, Fusaro RM, Lynch JF, Brand R. Pancreatic
extracolonic tumor spectrum in MUTYH-associated 269. Guilford P, Hopkins J, Harraway J, et al. E-cadherin cancer and the FAMMM syndrome. Fam Cancer.
polyposis. Gastroenterology. 2009;137(6):1976–1985. germline mutations in familial gastric cancer. Nature. 2008;7(1):103–112.
e1971. 1998;392(6674):402–405. 291. Lowenfels AB, Maisonneuve P, DiMagno EP, et al.
251. Aretz S, Uhlhaas S, Goergens H, et al. MUTYH- 270. Brooks-Wilson AR, Kaurah P, Suriano G, et al. Hereditary pancreatitis and the risk of pancreatic
associated polyposis: 70 of 71 patients with biallelic Germline E-cadherin mutations in hereditary diffuse cancer. International Hereditary Pancreatitis Study
mutations present with an attenuated or atypical gastric cancer: assessment of 42 new families and Group. J Natl Cancer Inst. 1997;89(6):442–446.
phenotype. Int J Cancer. 2006;119(4):807–814. review of genetic screening criteria. J Med Genet. 292. Rebours V, Boutron-Ruault M-C, Schnee M, et al.
252. Balaguer F, Castellví-Bel S, Castells A, et al. Identifi- 2004;41(7):508–517. Risk of pancreatic adenocarcinoma in patients with
cation of MYH mutation carriers in colorectal cancer: 271. Kaurah P, MacMillan A, Boyd N, et al. Founder and hereditary pancreatitis: a national exhaustive series.
a multicenter, case-control, population-based study. recurrent CDH1 mutations in families with heredi- Am J Gastroenterol. 2008;103(1):111–119.
Clin Gastroenterol Hepatol. 2007;5(3):379–387. tary diffuse gastric cancer. JAMA. 2007;297(21): 293. Vasen H, Ibrahim I, Ponce CG, et al. Benefit of
253. Jones N, Vogt S, Nielsen M, et al. Increased 2360–2372. surveillance for pancreatic cancer in high-risk indi-
colorectal cancer incidence in obligate carriers of 272. Oliveira C, Senz J, Kaurah P, et al. Germline CDH1 viduals: outcome of long-term prospective follow-up
heterozygous mutations in MUTYH. Gastroenterol- deletions in hereditary diffuse gastric cancer families. studies from three European expert centers. J Clin
ogy. 2009;137(2):489–494, 494.e481; quiz 725–486. Hum Mol Genet. 2009;18(9):1545–1555. Oncol. 2016;34(17):2010–2019.
254. Jones S, Emmerson P, Maynard J, et al. Biallelic 273. Carneiro F, Oliveira C, Suriano G, Seruca R. 294. Atherton DJ, Pitcher DW, Wells RS, MacDonald
germline mutations in MYH predispose to multiple Molecular pathology of familial gastric cancer, with DM. A syndrome of various cutaneous pigmented
colorectal adenoma and somatic G:C–>T:A muta- an emphasis on hereditary diffuse gastric cancer. lesions, myxoid neurofibromata and atrial myxoma:
tions. Hum Mol Genet. 2002;11(23):2961–2967. J Clin Pathol. 2008;61(1):25–30. the NAME syndrome. Br J Dermatol. 1980;103(4):
255. Russell AM, Zhang J, Luz J, et al. Prevalence of 274. Strong VE, Gholami S, Shah MA, et al. Total 421–429.
MYH germline mutations in Swiss APC mutation- gastrectomy for hereditary diffuse gastric cancer at 295. Rhodes AR, Silverman RA, Harrist TJ, Perez-Atayde
negative polyposis patients. Int J Cancer. 2006; a single center: postsurgical outcomes in 41 patients. AR. Mucocutaneous lentigines, cardiomucocutaneous
118(8):1937–1940. Ann Surg. 2016. myxomas, and multiple blue nevi: the “LAMB”
256. Jo W-S, Bandipalliam P, Shannon KM, et al. 275. Howlader N, Noone AM, Krapcho M, Miller D, syndrome. J Am Acad Dermatol. 1984;10(1):72–82.
Correlation of polyp number and family history Bishop K, Kosary CL, et al, eds. SEER Cancer 296. Stratakis CA, Carney JA, Lin JP, et al. Carney
of colon cancer with germline MYH mutations. Statistics Review, 1975-2014. Bethesda, MD: complex, a familial multiple neoplasia and lentigi-
Clin Gastroenterol Hepatol. 2005;3(10):1022–1028. National Cancer Institute. https://seer.cancer.gov/ nosis syndrome. Analysis of 11 kindreds and linkage
257. Eliason K, Hendrickson BC, Judkins T, et al. The csr/1975_2014/, based on November 2016 SEER to the short arm of chromosome 2. J Clin Invest.
potential for increased clinical sensitivity in genetic data submission, posted to the SEER web site, April 1996;97(3):699–705.
testing for polyposis colorectal cancer through the 2017. 297. Stratakis CA, Kirschner LS, Carney JA. Clinical and
analysis of MYH mutations in North American 276. Klein AP. Genetic susceptibility to pancreatic cancer. molecular features of the Carney complex: diagnostic
patients. J Med Genet. 2005;42(1):95–96. Mol Carcinog. 2012;51(1):14–24. criteria and recommendations for patient evalua-
258. Fleischmann C, Peto J, Cheadle J, Shah B, 277. Goggins M, Schutte M, Lu J, et al. Germline tion. J Clin Endocrinol Metab. 2001;86(9):4041–
Sampson J, Houlston RS. Comprehensive analysis BRCA2 gene mutations in patients with appar- 4046.
of the contribution of germline MYH variation to ently sporadic pancreatic carcinomas. Cancer Res. 298. Bertherat J, Horvath A, Groussin L, et al. Mutations
early-onset colorectal cancer. Int J Cancer. 2004; 1996;56(23):5360–5364. in regulatory subunit type 1A of cyclic adenosine
109(4):554–558. 278. Hahn SA, Greenhalf B, Ellis I, et al. BRCA2 germline 5′-monophosphate-dependent protein kinase
259. Nielsen M, Franken PF, Reinards THCM, et al. mutations in familial pancreatic carcinoma. J Natl (PRKAR1A): phenotype analysis in 353 patients and
Multiplicity in polyp count and extracolonic Cancer Inst. 2003;95(3):214–221. 80 different genotypes. J Clin Endocrinol Metab. 2009;
manifestations in 40 Dutch patients with MYH 279. Murphy KM, Brune KA, Griffin C, et al. Evaluation 94(6):2085–2091.
associated polyposis coli (MAP). J Med Genet. 2005; of candidate genes MAP2K4, MADH4, ACVR1B, 299. Courcoutsakis NA, Chow CK, Shawker TH,
42(9):e54. and BRCA2 in familial pancreatic cancer: del- Carney JA, Stratakis CA. Syndrome of spotty skin
260. Palles C, Cazier JB, Howarth KM, et al. Germline eterious BRCA2 mutations in 17%. Cancer Res. pigmentation, myxomas, endocrine overactivity, and
mutations affecting the proofreading domains of 2002;62(13):3789–3793. schwannomas (Carney complex): breast imaging
POLE and POLD1 predispose to colorectal adenomas 280. Cancer risks in BRCA2 mutation carriers. J Natl findings. Radiology. 1997;205(1):221–227.
and carcinomas. Nat Genet. 2013;45(2):136–144. Cancer Inst. 1999;91(15):1310–1316. 300. Premkumar A, Stratakis CA, Shawker TH, Papa-
261. Bellido F, Pineda M, Aiza G, et al. POLE and 281. Risch HA, McLaughlin JR, Cole DEC, et al. Popula- nicolaou DA, Chrousos GP. Testicular ultrasound
POLD1 mutations in 529 kindred with familial tion BRCA1 and BRCA2 mutation frequencies and in Carney complex: report of three cases. J Clin
colorectal cancer and/or polyposis: review of reported cancer penetrances: a kin-cohort study in Ontario, Ultrasound. 1997;25(4):211–214.
cases and recommendations for genetic testing and Canada. J Natl Cancer Inst. 2006;98(23):1694–1706. 301. Stratakis CA, Sarlis N, Kirschner LS, et al. Paradoxi-
surveillance. Genet Med. 2016;18(4):325–332. 282. Ferrone CR, Levine DA, Tang LH, et al. BRCA cal response to dexamethasone in the diagnosis of
262. Jaeger E, Leedham S, Lewis A, et al. Hereditary germline mutations in Jewish patients with pancreatic primary pigmented nodular adrenocortical disease.
mixed polyposis syndrome is caused by a 40-kb adenocarcinoma. J Clin Oncol. 2009;27(3):433–438. Ann Intern Med. 1999;131(8):585–591.
upstream duplication that leads to increased and 283. Thompson D, Easton DF, Breast Cancer Linkage 302. Stratakis CA, Courcoutsakis NA, Abati A, et al.
ectopic expression of the BMP antagonist GREM1. C. Cancer incidence in BRCA1 mutation carriers. Thyroid gland abnormalities in patients with the
Nat Genet. 2012;44(6):699–703. J Natl Cancer Inst. 2002;94(18):1358–1365. syndrome of spotty skin pigmentation, myxomas,
263. Cao X, Eu KW, Kumarasinghe MP, Li HH, Loi C, 284. Stadler ZK, Salo-Mullen E, Patil SM, et al. Prevalence endocrine overactivity, and schwannomas (Carney
Cheah PY. Mapping of hereditary mixed polyposis of BRCA1 and BRCA2 mutations in Ashkenazi complex). J Clin Endocrinol Metab. 1997;82(7):
syndrome (HMPS) to chromosome 10q23 by Jewish families with breast and pancreatic cancer. 2037–2043.
genomewide high-density single nucleotide polymor- Cancer. 2012;118(2):493–499. 303. Stratakis CA, Papageorgiou T, Premkumar A, et al.
phism (SNP) scan and identification of BMPR1A 285. Rogers CD, van der Heijden MS, Brune K, et al. The Ovarian lesions in Carney complex: clinical genetics
loss of function. J Med Genet. 2006;43(3):e13. genetics of FANCC and FANCG in familial pancre- and possible predisposition to malignancy. J Clin
264. Cybulski C, Górski B, Huzarski T, et al. CHEK2 is atic cancer. Cancer Biol Ther. 2004;3(2):167–169. Endocrinol Metab. 2000;85(11):4359–4366.
a multiorgan cancer susceptibility gene. Am J Hum 286. Tischkowitz MD, Sabbaghian N, Hamel N, et al. 304. Kirschner LS, Sandrini F, Monbo J, Lin JP, Carney
Genet. 2004;75(6):1131–1135. Analysis of the gene coding for the BRCA2- JA, Stratakis CA. Genetic heterogeneity and
265. Laken SJ, Petersen GM, Gruber SB, et al. Familial interacting protein PALB2 in familial and sporadic spectrum of mutations of the PRKAR1A gene in
colorectal cancer in Ashkenazim due to a hypermu- pancreatic cancer. Gastroenterology. 2009;137(3): patients with the carney complex. Hum Mol Genet.
table tract in APC. Nat Genet. 1997;17(1):79–83. 1183–1186. 2000;9(20):3037–3046.
266. Rio Frio T, Lavoie J, Hamel N, et al. Homozygous 287. Schneider R, Slater EP, Sina M, et al. German 305. Casey M, Vaughan CJ, He J, et al. Mutations in
BUB1B mutation and susceptibility to gastrointestinal national case collection for familial pancreatic cancer the protein kinase A R1alpha regulatory subunit
208.e6 Part I: Science and Clinical Oncology

cause familial cardiac myxomas and Carney complex. 326. Decker RA, Peacock ML, Borst MJ, Sweet JD, tumour-suppressor gene in medulloblastoma. Br J
J Clin Invest. 2000;106(5):R31–R38. Thompson NW. Progress in genetic screening of Cancer. 1997;76(2):141–145.
306. Boikos SA, Stratakis CA. Carney complex: the first multiple endocrine neoplasia type 2A: is calcitonin 347. Amlashi SFA, Riffaud L, Brassier G, Morandi X.
20 years. Curr Opin Oncol. 2007;19(1):24–29. testing obsolete? Surgery. 1995;118(2):257–263, Nevoid basal cell carcinoma syndrome: relation
307. Milanesi U, Mangili F, Milanesi I. Flow-cytometric discussion 263. with desmoplastic medulloblastoma in infancy. A
study of familial paragangliomas of the carotid body. 327. Lips CJ, Landsvater RM, Höppener JW, et al. population-based study and review of the literature.
Acta Otorhinolaryngol Ital. 1994;14(4):439–447. Clinical screening as compared with DNA analysis Cancer. 2003;98(3):618–624.
308. Netterville JL, Reilly KM, Robertson D, Reiber ME, in families with multiple endocrine neoplasia type 348. Johnson RL, Rothman AL, Xie J, et  al.
Armstrong WB, Childs P. Carotid body tumors: a 2A. N Engl J Med. 1994;331(13):828–835. Human homolog of patched, a candidate gene
review of 30 patients with 46 tumors. Laryngoscope. 328. Wells SA, Chi DD, Toshima K, et al. Predictive DNA for the basal cell nevus syndrome. Science.
1995;105(2):115–126. testing and prophylactic thyroidectomy in patients 1996;272(5268):1668–1671.
309. van der Mey A, Maaswinkel-Mooy PD, Cornelisse at risk for multiple endocrine neoplasia type 2A. 349. Wicking C, Shanley S, Smyth I, et al. Most germ-
CJ, Schmidt P, van de Kamp J. Genomic imprinting Ann Surg. 1994;220(3):237–247, discussion 247. line mutations in the nevoid basal cell carcinoma
in hereditary glomus tumours: evidence for new 329. Eng C, Clayton D, Schuffenecker I, et al. The rela- syndrome lead to a premature termination of the
genetic theory. Lancet. 1989;2:1291–1294. tionship between specific RET proto-oncogene muta- PATCHED protein, and no genotype-phenotype
310. Baysal BE, Ferrell RE, Willett-Brozick JE, et al. tions and disease phenotype in multiple endocrine correlations are evident. Am J Hum Genet. 1997;
Mutations in SDHD, a mitochondrial complex neoplasia type 2. International RET mutation con- 60(1):21–26.
II gene, in hereditary paraganglioma. Science. sortium analysis. JAMA. 1996;276(19):1575–1579. 350. Von Hoff DD, LoRusso PM, Rudin CM, et al. Inhi-
2000;287(5454):848–851. 330. Leboulleux S, Travagli JP, Caillou B, et al. Medullary bition of the hedgehog pathway in advanced basal-cell
311. Hao H-X, Khalimonchuk O, Schraders M, et al. thyroid carcinoma as part of a multiple endocrine carcinoma. N Engl J Med. 2009;361(12):1164–1172.
SDH5, a gene required for flavination of succinate neoplasia type 2B syndrome: influence of the stage 351. Tate G, Li M, Suzuki T, Mitsuya T. A new germline
dehydrogenase, is mutated in paraganglioma. Science. on the clinical course. Cancer. 2002;94(1):44–50. mutation of the PTCH gene in a Japanese patient
2009;325(5944):1139–1142. 331. Smith DP, Houghton C, Ponder BA. Germline with nevoid basal cell carcinoma syndrome
312. Hensen EF, Jordanova ES, van Minderhout IJHM, mutation of RET codon 883 in two cases of de associated with meningioma. Jpn J Clin Oncol.
et al. Somatic loss of maternal chromosome 11 causes novo MEN 2B. Oncogene. 1997;15(10):1213–1217. 2003;33(1):47–50.
parent-of-origin-dependent inheritance in SDHD- 332. American Thyroid Association Guidelines Task Force, 352. Boutet N, Bignon Y-J, Drouin-Garraud V,
linked paraganglioma and phaeochromocytoma Kloos RT, Eng C, et al. Medullary thyroid cancer: et al. Spectrum of PTCH1 mutations in French
families. Oncogene. 2004;23(23):4076–4083. management guidelines of the American Thyroid patients with Gorlin syndrome. J Invest Dermatol.
313. Neumann HPH, Pawlu C, Peczkowska M, et al. Association. Thyroid. 2009;19(6):565–612. 2003;121(3):478–481.
Distinct clinical features of paraganglioma syndromes 333. Lairmore TC, Ball DW, Baylin SB, Wells SA. 353. Lindstrom E, Shimokawa T, Toftgard R, Zaphi-
associated with SDHB and SDHD gene mutations. Management of pheochromocytomas in patients ropoulos PG. PTCH mutations: distribution and
JAMA. 2004;292(8):943–951. with multiple endocrine neoplasia type 2 syndromes. analyses. Hum Mutat. 2006;27(3):215–219.
314. Neumann HPH, Bausch B, McWhinney SR, et al. Ann Surg. 1993;217(6):595–601, discussion 601. 354. Anand VK, Arrowood JP, Krolls SO. Malignant
Germ-line mutations in nonsyndromic pheochromo- 334. Wells SA, Robinson BG, Gagel RF, et al. Vandetanib potential of the odontogenic keratocyst. Otolaryngol
cytoma. N Engl J Med. 2002;346(19):1459–1466. in patients with locally advanced or metastatic Head Neck Surg. 1994;111(1):124–129.
315. Bryant J, Farmer J, Kessler LJ, Townsend RR, medullary thyroid cancer: a randomized, double- 355. Bitar GJ, Herman CK, Dahman MI, Hoard MA.
Nathanson KL. Pheochromocytoma: the expanding blind phase III trial. J Clin Oncol. 2012;30(2):134– Basal cell nevus syndrome: guidelines for early detec-
genetic differential diagnosis. J Natl Cancer Inst. 141. tion. Am Fam Physician. 2002;65(12):2501–2504.
2003;95(16):1196–1204. 335. Schlumberger MJ, Elisei R, Bastholt L, et al. Phase II 356. National Cancer Institute. FDA approval for vis-
316. Hensen EF, van Duinen N, Jansen JC, et al. High study of safety and efficacy of motesanib in patients modegib; 2012. http://www.cancer.gov/cancertopics/
prevalence of founder mutations of the succinate with progressive or symptomatic, advanced or druginfo/fda-vismodegib.
dehydrogenase genes in the Netherlands. Clin Genet. metastatic medullary thyroid cancer. J Clin Oncol. 357. Food and Drug Administration. Vismodegib; 2012.
2012;81(3):284–288. 2009;27(23):3794–3801. http://www.fda.gov/Drugs/InformationOnDrugs/
317. Farndon JR, Leight GS, Dilley WG, et al. Familial 336. Lam ET, Ringel MD, Kloos RT, et al. Phase II clini- ApprovedDrugs/ucm289571.htm.
medullary thyroid carcinoma without associated endo- cal trial of sorafenib in metastatic medullary thyroid 358. Tang JY, Mackay-Wiggan JM, Aszterbaum M,
crinopathies: a distinct clinical entity. Br J Surg. 1986; cancer. J Clin Oncol. 2010;28(14):2323–2330. et al. Inhibiting the hedgehog pathway in patients
73(4):278–281. 337. Mologni L. Development of RET kinase inhibi- with the basal-cell nevus syndrome. N Engl J Med.
318. Moline J, Eng C. Multiple endocrine neoplasia type tors for targeted cancer therapy. Curr Med Chem. 2012;366(23):2180–2188.
2: an overview. Genet Med. 2011;13(9):755–764. 2011;18(2):162–175. 359. Launonen V, Vierimaa O, Kiuru M, et  al.
319. Vasen HF, van der Feltz M, Raue F, et al. The 338. Phay JE, Shah MH. Targeting RET receptor Inherited susceptibility to uterine leiomyomas
natural course of multiple endocrine neoplasia tyrosine kinase activation in cancer. Clin Cancer and renal cell cancer. Proc Natl Acad Sci USA.
type IIb. A study of 18 cases. Arch Intern Med. Res. 2010;16(24):5936–5941. 2001;98(6):3387–3392.
1992;152(6):1250–1252. 339. Gorlin RJ, Goltz RW. Multiple nevoid basal-cell 360. Alam NA, Rowan AJ, Wortham NC, et al.
320. Stoffer SS, Van Dyke DL, Bach JV, Szpunar W, epithelioma, jaw cysts and bifid rib: a syndrome. Genetic and functional analyses of FH mutations
Weiss L. Familial papillary carcinoma of the thyroid. N Engl J Med. 1960;262:908–912. in multiple cutaneous and uterine leiomyomatosis,
Am J Med Genet. 1986;25(4):775–782. 340. Gorlin RJ. Nevoid basal-cell carcinoma syndrome. hereditary leiomyomatosis and renal cancer, and
321. Mulligan LM, Eng C, Healey CS, et al. Specific Medicine (Baltimore). 1987;66(2):98–113. fumarate hydratase deficiency. Hum Mol Genet.
mutations of the RET proto-oncogene are related 341. Strong LC. Genetic and environmental interactions. 2003;12(11):1241–1252.
to disease phenotype in MEN 2A and FMTC. Nat Cancer. 1977;40(suppl 4):1861–1866. 361. Linehan WM, Pinto PA, Srinivasan R, et al. Identi-
Genet. 1994;6(1):70–74. 342. Evans DG, Farndon PA, Burnell LD, Gattamaneni fication of the genes for kidney cancer: opportunity
322. Eng C, Mulligan LM, Smith DP, et al. Low HR, Birch JM. The incidence of Gorlin syndrome for disease-specific targeted therapeutics. Clin Cancer
frequency of germline mutations in the RET in 173 consecutive cases of medulloblastoma. Br J Res. 2007;13(2 Pt 2):671s–679s.
proto-oncogene in patients with apparently sporadic Cancer. 1991;64(5):959–961. 362. Toro JR, Nickerson ML, Wei M-H, et al. Mutations
medullary thyroid carcinoma. Clin Endocrinol (Oxf). 343. Evans DG, Ladusans EJ, Rimmer S, Burnell in the fumarate hydratase gene cause hereditary
1995;43(1):123–127. LD, Thakker N, Farndon PA. Complications leiomyomatosis and renal cell cancer in families in
323. Mulligan LM, Kwok JB, Healey CS, et al. Germ- of the naevoid basal cell carcinoma syndrome: North America. Am J Hum Genet. 2003;73(1):95–
line mutations of the RET proto-oncogene in results of a population based study. J Med Genet. 106.
multiple endocrine neoplasia type 2A. Nature. 1993; 1993;30(6):460–464. 363. Martinez-Mir A, Glaser B, Chuang GS, et al.
363(6428):458–460. 344. Kimonis VE, Goldstein AM, Pastakia B, et al. Germline fumarate hydratase mutations in families
324. Donis-Keller H, Dou S, Chi D, et al. Mutations in Clinical manifestations in 105 persons with nevoid with multiple cutaneous and uterine leiomyomata.
the RET proto-oncogene are associated with MEN basal cell carcinoma syndrome. Am J Hum Genet. J Invest Dermatol. 2003;121(4):741–744.
2A and FMTC. Hum Mol Genet. 1993;2(7):851– 1997;69(3):299–308. 364. Badeloe S, van Geel M, van Steensel MAM,
856. 345. Bree AF, Shah MR, Group BC. Consensus statement et al. Diffuse and segmental variants of cutaneous
325. Schuffenecker I, Ginet N, Goldgar D, et al. from the first international colloquium on basal leiomyomatosis: novel mutations in the fumarate
Prevalence and parental origin of de novo RET cell nevus syndrome (BCNS). Am J Med Genet A. hydratase gene and review of the literature. Exp
mutations in multiple endocrine neoplasia type 2011;155A(9):2091–2097. Dermatol. 2006;15(9):735–741.
2A and familial medullary thyroid carcinoma. Le 346. Cowan R, Hoban P, Kelsey A, Birch JM, 365. Sudarshan S, Pinto PA, Neckers L, Linehan WM.
Groupe d’Etude des Tumeurs a Calcitonine. Am J Gattamaneni R, Evans DG. The gene for the Mechanisms of disease: hereditary leiomyomatosis
Hum Genet. 1997;60(1):233–237. naevoid basal cell carcinoma syndrome acts as a and renal cell cancer–a distinct form of hereditary
Genetic Factors: Hereditary Cancer Predisposition Syndromes  •  CHAPTER 13 208.e7
208.e7

kidney cancer. Nat Clin Pract Urol. 2007;4(2): 384. Singer EA, Gupta GN, Srinivasan R. Update on factor receptor alpha gene in gastrointestinal stromal
104–110. targeted therapies for clear cell renal cell carcinoma. tumors. Gastroenterology. 2003;125(3):660–667.
366. Lehtonen HJ, Kiuru M, Ylisaukko-Oja SK, et al. Curr Opin Oncol. 2011;23(3):283–289. 406. Nishida T, Hirota S, Taniguchi M, et al. Familial
Increased risk of cancer in patients with fumarate 385. Birt AR, Hogg GR, Dubé WJ. Hereditary multiple gastrointestinal stromal tumours with germline
hydratase germline mutation. J Med Genet. 2006;43(6): fibrofolliculomas with trichodiscomas and acrochor- mutation of the KIT gene. Nat Genet. 1998;
523–526. dons. Arch Dermatol. 1977;113(12):1674–1677. 19(4):323–324.
367. Wei MH, Toure O, Glenn GM, et al. Novel 386. Pavlovich CP, Grubb RL, Hurley K, et al. Evaluation 407. Chompret A, Kannengiesser C, Barrois M, et al.
mutations in FH and expansion of the spectrum and management of renal tumors in the Birt-Hogg- PDGFRA germline mutation in a family with
of phenotypes expressed in families with hereditary Dubé syndrome. J Urol. 2005;173(5):1482–1486. multiple cases of gastrointestinal stromal tumor.
leiomyomatosis and renal cell cancer. J Med Genet. 387. Durrani OH, Ng L, Bihrle W. Chromophobe renal Gastroenterology. 2004;126(1):318–321.
2006;43(1):18–27. cell carcinoma in a patient with the Birt-Hogg-Dube 408. Graham J, Debiec-Rychter M, Corless CL, Reid R,
368. Kiuru M, Lehtonen R, Eerola H, et al. No germline syndrome. J Urol. 2002;168(4 Pt 1):1484–1485. Davidson R, White JD. Imatinib in the management
FH mutations in familial breast cancer patients. Eur 388. Schmidt LS, Nickerson ML, Warren MB, et al. of multiple gastrointestinal stromal tumors associated
J Hum Genet. 2005;13(4):506–509. Germline BHD-mutation spectrum and phenotype with a germline KIT K642E mutation. Arch Pathol
369. Isaacs JS, Jung YJ, Mole DR, et al. HIF overexpression analysis of a large cohort of families with Birt-Hogg- Lab Med. 2007;131(9):1393–1396.
correlates with biallelic loss of fumarate hydratase in Dubé syndrome. Am J Hum Genet. 2005;76(6): 409. Jensen DE, Proctor M, Marquis ST, et al. BAP1:
renal cancer: novel role of fumarate in regulation 1023–1033. a novel ubiquitin hydrolase which binds to the
of HIF stability. Cancer Cell. 2005;8(2):143– 389. Walter P, Kirchhof B, Korge B, Heimann K. Flecked BRCA1 RING finger and enhances BRCA1-
153. chorioretinopathy associated with Birt-Hogg-Dubé mediated cell growth suppression. Oncogene.
370. Chuang GS, Martinez-Mir A, Geyer A, et al. Germ- syndrome. Graefes Arch Clin Exp Ophthalmol. 1997; 1998;16(9):1097–1112.
line fumarate hydratase mutations and evidence for 235(6):359–361. 410. Bott M, Brevet M, Taylor BS, et al. The nuclear
a founder mutation underlying multiple cutaneous 390. Khoo SK, Giraud S, Kahnoski K, et al. Clinical deubiquitinase BAP1 is commonly inactivated by
and uterine leiomyomata. J Am Acad Dermatol. and genetic studies of Birt-Hogg-Dubé syndrome. somatic mutations and 3p21.1 losses in malignant
2005;52(3 Pt 1):410–416. J Med Genet. 2002;39(12):906–912. pleural mesothelioma. Nat Genet. 2011;43(7):
371. Kiuru M, Launonen V, Hietala M, et al. Familial 391. Kluger N, Giraud S, Coupier I, et al. Birt-Hogg-Dubé 668–672.
cutaneous leiomyomatosis is a two-hit condition syndrome: clinical and genetic studies of 10 French 411. Testa JR, Cheung M, Pei J, et al. Germline BAP1
associated with renal cell cancer of characteristic families. Br J Dermatol. 2010;162(3):527–537. mutations predispose to malignant mesothelioma.
histopathology. Am J Pathol. 2001;159(3):825– 392. Byrne M, Mallipeddi R, Pichert G, Whittaker S. Nat Genet. 2011;43(10):1022–1025.
829. Birt-Hogg-Dubé syndrome with a renal angiomyo- 412. Wiesner T, Obenauf AC, Murali R, et al. Germline
372. Zinn AB, Kerr DS, Hoppel CL. Fumarase deficiency: lipoma: further evidence of a relationship between mutations in BAP1 predispose to melanocytic
a new cause of mitochondrial encephalomyopathy. Birt-Hogg-Dubé syndrome and tuberous sclerosis tumors. Nat Genet. 2011;43(10):1018–1021.
N Engl J Med. 1986;315(8):469–475. complex. Australas J Dermatol. 2012;53(2):151–154. 413. Abdel-Rahman MH, Pilarski R, Cebulla CM, et al.
373. Alam NA, Bevan S, Churchman M, et  al. 393. Misago N, Narisawa Y. Fibrofolliculoma in a patient Germline BAP1 mutation predisposes to uveal
Localization of a gene (MCUL1) for multiple with tuberous sclerosis complex. Clin Exp Dermatol. melanoma, lung adenocarcinoma, meningioma, and
cutaneous leiomyomata and uterine fibroids to 2009;34(8):892–894. other cancers. J Med Genet. 2011;48(12):856–859.
chromosome 1q42.3-q43. Am J Hum Genet. 394. Khoo SK, Bradley M, Wong FK, Hedblad MA, 414. Pena-Llopis S, Vega-Rubin-de-Celis S, Liao A, et al.
2001;68(5):1264–1269. Nordenskjöld M, Teh BT. Birt-Hogg-Dubé syn- BAP1 loss defines a new class of renal cell carcinoma.
374. Tomlinson IPM, Alam NA, Rowan AJ, et al. Germline drome: mapping of a novel hereditary neoplasia Nat Genet. 2012;44(7):751–759.
mutations in FH predispose to dominantly inherited gene to chromosome 17p12-q11.2. Oncogene. 2001; 415. Beguelin W, Popovic R, Teater M, et al. EZH2 is
uterine fibroids, skin leiomyomata and papillary renal 20(37):5239–5242. required for germinal center formation and somatic
cell cancer. Nat Genet. 2002;30(4):406–410. 395. Nickerson ML, Warren MB, Toro JR, et al. Muta- EZH2 mutations promote lymphoid transformation.
375. Pithukpakorn M, Wei MH, Toure O, et al. Fumarate tions in a novel gene lead to kidney tumors, lung Cancer Cell. 2013;23(5):677–692.
hydratase enzyme activity in lymphoblastoid cells and wall defects, and benign tumors of the hair follicle 416. Jones S, Anagnostou V, Lytle K, et al. Personalized
fibroblasts of individuals in families with hereditary in patients with the Birt-Hogg-Dubé syndrome. genomic analyses for cancer mutation discovery
leiomyomatosis and renal cell cancer. J Med Genet. Cancer Cell. 2002;2(2):157–164. and interpretation. Sci Transl Med. 2015;7(283):
2006;43(9):755–762. 396. Hartman TR, Nicolas E, Klein-Szanto A, et al. 283ra253.
376. Choyke PL. Imaging of hereditary renal cancer. The role of the Birt-Hogg-Dubé protein in mTOR 417. Bombard Y, Robson M, Offit K. Revealing the
Radiol Clin North Am. 2003;41(5):1037–1051. activation and renal tumorigenesis. Oncogene. incidentalome when targeting the tumor genome.
377. Gordon MS, Hussey M, Nagle RB, et al. Phase II 2009;28(13):1594–1604. JAMA. 2013;310(8):795–796.
study of erlotinib in patients with locally advanced 397. Toro JR, Wei MH, Glenn GM, et al. BHD muta- 418. Schrader KA, Cheng DT, Joseph V, et al. Germline
or metastatic papillary histology renal cell cancer: tions, clinical and molecular genetic investigations variants in targeted tumor sequencing using matched
SWOG S0317. J Clin Oncol. 2009;27(34): of Birt-Hogg-Dube syndrome: a new series of 50 normal DNA. JAMA Oncol. 2016;2(1):104–111.
5788–5793. families and a review of published reports. J Med 419. Mody RJ, Wu YM, Lonigro RJ, et al. Integrative
378. National Institutes of Health Clinical Center Genet. 2008;45(6):321–331. clinical sequencing in the management of refractory
(CC) (National Cancer Institute [NCI]). A phase 398. Barrisford GW, Singer EA, Rosner IL, Linehan or relapsed cancer in youth. JAMA. 2015;314(9):
II study of bevacizumab and erlotinib in subjects WM, Bratslavsky G. Familial renal cancer: molecular 913–925.
with advanced hereditary leiomyomatosis and renal genetics and surgical management. Int J Surg Oncol. 420. Mandelker D, Zhang L, Kemel Y, et al. Mutation
cell cancer (HLRCC) or sporadic papillary renal 2011;(2011):658767. detection in patients with advanced cancer by univer-
cell cancer. Bethesda, MD: National Library of 399. Roach ES, Sparagana SP. Diagnosis of tuberous scle- sal sequencing of cancer-related genes in tumor and
Medicine (US)2000: http://clinicaltrials.gov/show/ rosis complex. J Child Neurol. 2004;19(9):643–649. normal DNA compared to guideline-based germline
NCT01130519. 400. Agarwal R, Robson M. Inherited predisposition to testing. JAMA. 2017;In Press.
379. Lonser RR, Glenn GM, Walther M, et al. von Hippel- gastrointestinal stromal tumor. Hematol Oncol Clin 421. Pritchard CC, Mateo J, Walsh MF, et al. Inherited
Lindau disease. Lancet. 2003;361(9374):2059–2067. North Am. 2009;23(1):1–13. DNA-repair gene mutations in men with metastatic
380. Maher ER, Neumann HP, Richard S. von Hippel- 401. Lane BR, Aydin H, Danforth TL, et al. Clinical prostate cancer. N Engl J Med. 2016;375(5):443–453.
Lindau disease: a clinical and scientific review. Eur correlates of renal angiomyolipoma subtypes in 209 422. Lynch HT, Lynch JF, Lynch PM, Attard T. Hereditary
J Hum Genet. 2011;19(6):617–623. patients: classic, fat poor, tuberous sclerosis associated colorectal cancer syndromes: molecular genetics,
381. Frantzen C, Links TP, Giles RH. Von Hippel-Lindau and epithelioid. J Urol. 2008;180(3):836–843. genetic counseling, diagnosis and management.
disease. In: GeneReviews at GeneTests Medical 402. Bissler J, Kingswood JC, Zonnenberg BA, et al. Fam Cancer. 2008;7(1):27–39.
Genetics Information Resource (database online). Everolimus therapy for angiomyolipoma in patients 423. Achatz MI, Porter CC, Brugieres L, et al. Cancer
Updated June 21, 2012. Copyright University of with tuberous sclerosis complex or sporadic lymphan- screening recommendations and clinical management
Washington, Seattle. 1997-2013. Available at: http:// gioleiomyomatosis: Results from EXIST-2. J Clin of inherited gastrointestinal cancer syndromes in
www.genetests.org. Accessed October 1, 2012. Oncol. 2012;30(suppl 5):356. childhood. Clin Cancer Res. 2017;23(13):e107–
382. National Comprehensive Cancer Centers. NCCN 403. FDA. FDA approves Afinitor for non-cancerous e114.
Clinical Practice Guidelines in Oncology, Kidney kidney tumors caused by rare genetic disease; 2012. 424. Locker GY, Kaul K, Weinberg DS, et al. The
Cancer Version 2.2017; 2016. http://www.nccn.org/ 404. Hirota S, Isozaki K, Moriyama Y, et al. Gain-of- I1307K APC polymorphism in Ashkenazi Jews with
professionals/physician_gls/pdf/kidney.pdf. function mutations of c-kit in human gastrointestinal colorectal cancer: clinical and pathologic features.
383. Brugarolas J, Kaelin WG. Dysregulation of HIF and stromal tumors. Science. 1998;279(5350):577–580. Cancer Genet Cytogenet. 2006;169(1):33–38.
VEGF is a unifying feature of the familial hamartoma 405. Hirota S, Ohashi A, Nishida T, et al. Gain-of- 425. Ma X, Zhang B, Zheng W. Genetic variants asso-
syndromes. Cancer Cell. 2004;6(1):7–10. function mutations of platelet-derived growth ciated with colorectal cancer risk: comprehensive
208.e8 Part I: Science and Clinical Oncology

research synopsis, meta-analysis, and epidemiological 443. Friedl W, Uhlhaas S, Schulmann K, et al. Juvenile polyposis syndrome. Gastroenterology. 1997;112(2):
evidence. Gut. 2014;63(2):326–336. polyposis: massive gastric polyposis is more common 327–334.
426. Gatti R, Perlman S. Ataxia-telangiectasia. In: in MADH4 mutation carriers than in BMPR1A 461. Plesec T, Brown K, Allen C, et al. Clinicopathological
Pagon RA, Adam MP, Ardinger HH, et al, eds. mutation carriers. Hum Genet. 2002;111(1):108–111. features of a kindred with SCG5-GREM1-associated
GeneReviews(R). Seattle (WA): University of 444. Chen S, Parmigiani G. Meta-analysis of BRCA1 hereditary mixed polyposis syndrome. Hum Pathol.
Washington, Seattle University of Washington, and BRCA2 penetrance. J Clin Oncol. 2007;25(11): 2017;60:75–81.
Seattle. GeneReviews is a registered trademark of the 1329–1333. 462. Porter CC, Druley TE, Erez A, et al. Recom-
University of Washington, Seattle. All rights reserved; 445. Gonzalez-Angulo AM, Timms KM, Liu S, et al. Inci- mendations for surveillance for children with
1993. dence and outcome of BRCA mutations in unselected leukemia-predisposing conditions. Clin Cancer Res.
427. Suarez F, Mahlaoui N, Canioni D, et al. Incidence, patients with triple receptor-negative breast cancer. 2017;23(11):e14–e22.
presentation, and prognosis of malignancies in ataxia- Clin Cancer Res. 2011;17(5):1082–1089. 463. Topka S, Vijai J, Walsh MF, et al. Germline ETV6
telangiectasia: a report from the French national 446. Gallagher DJ, Gaudet MM, Pal P, et al. Germline mutations confer susceptibility to acute lymphoblas-
registry of primary immune deficiencies. J Clin BRCA mutations denote a clinicopathologic subset tic leukemia and thrombocytopenia. PLoS Genet.
Oncol. 2015;33(2):202–208. of prostate cancer. Clin Cancer Res. 2010;16(7): 2015;11(6):e1005262.
428. Grant RC, Selander I, Connor AA, et al. Prevalence 2115–2121. 464. Holmfeldt L, Wei L, Diaz-Flores E, et al. The
of germline mutations in cancer predisposition genes 447. Fong PC, Boss DS, Yap TA, et al. Inhibition of genomic landscape of hypodiploid acute lympho-
in patients with pancreatic cancer. Gastroenterology. poly(ADP-ribose) polymerase in tumors from BRCA blastic leukemia. Nat Genet. 2013;45(3):242–252.
2015;148(3):556–564. mutation carriers. N Engl J Med. 2009;361(2): 465. Dang J, Wei L, de Ridder J, et al. PAX5 is a tumor
429. Walsh MF, Nathanson KL, Couch FJ, Offit K. 123–134. suppressor in mouse mutagenesis models of acute
Genomic biomarkers for breast cancer risk. Adv 448. Salo-Mullen EE, O’Reilly EM, Kelsen DP, et al. Iden- lymphoblastic leukemia. Blood. 2015;125(23):
Exp Med Biol. 2016;882:1–32. tification of germline genetic mutations in patients 3609–3617.
430. Pilarski R, Rai K, Cebulla C, Abdel-Rahman M. with pancreatic cancer. Cancer. 2015;121(24): 466. Mehta PA, Tolar J. Fanconi anemia. In: Pagon RA,
BAP1 tumor predisposition syndrome. In: Pagon RA, 4382–4388. Adam MP, Ardinger HH, et al, eds. GeneReviews(R).
Adam MP, Ardinger HH, et al, eds. GeneReviews(R). 449. Meyer S, Tischkowitz M, Chandler K, Gillespie A, Seattle (WA): University of Washington, Seattle
Seattle (WA): University of Washington, Seattle Birch JM, Evans DG. Fanconi anaemia, BRCA2 University of Washington, Seattle. GeneReviews is a
University of Washington, Seattle. GeneReviews is a mutations and childhood cancer: a developmental registered trademark of the University of Washington,
registered trademark of the University of Washington, perspective from clinical and epidemiological obser- Seattle. All rights reserved; 1993.
Seattle. All rights reserved; 1993. vations with implications for genetic counselling. 467. Kutler DI, Singh B, Satagopan J, et al. A 20-year
431. Sanz MM, German J, Cunniff C. Bloom’s syndrome. J Med Genet. 2014;51(2):71–75. perspective on the International Fanconi Anemia
In: Pagon RA, Adam MP, Ardinger HH, et al, 450. Pennington KP, Swisher EM. Hereditary ovarian Registry (IFAR). Blood. 2003;101(4):1249–1256.
eds. GeneReviews(R). Seattle (WA): University of cancer: beyond the usual suspects. Gynecol Oncol. 468. Rosenberg PS, Greene MH, Alter BP. Cancer
Washington, Seattle University of Washington, 2012;124(2):347–353. incidence in persons with Fanconi anemia. Blood.
Seattle. GeneReviews is a registered trademark of 451. van der Post RS, Vogelaar IP, Carneiro F, et al. 2003;101(3):822–826.
the University of Washington, Seattle. All rights Hereditary diffuse gastric cancer: updated clinical 469. Auerbach AD, Rogatko A, Schroeder-Kurth TM.
reserved; 1993. guidelines with an emphasis on germline CDH1 International Fanconi Anemia Registry: relation of
432. Bloom D. Congenital telangiectatic erythema mutation carriers. J Med Genet. 2015;52(6): clinical symptoms to diepoxybutane sensitivity. Blood.
resembling lupus erythematosus in dwarfs; probably 361–374. 1989;73(2):391–396.
a syndrome entity. Am J Dis Child. 1954;88(6): 452. Vasen HF, Gruis NA, Frants RR, van Der Velden 470. Kutler DI, Auerbach AD, Satagopan J, et al. High
754–758. PA, Hille ET, Bergman W. Risk of developing incidence of head and neck squamous cell carcinoma
433. German J, Sanz MM, Ciocci S, Ye TZ, Ellis NA. pancreatic cancer in families with familial atypical in patients with Fanconi anemia. Arch Otolaryngol
Syndrome-causing mutations of the BLM gene in multiple mole melanoma associated with a specific Head Neck Surg. 2003;129(1):106–112.
persons in the Bloom’s Syndrome Registry. Hum 19 deletion of p16 (p16-Leiden). Int J Cancer. 471. Faivre L, Guardiola P, Lewis C, et al. Association of
Mutat. 2007;28(8):743–753. 2000;87(6):809–811. complementation group and mutation type with clin-
434. Cunniff C, Bassetti JA, Ellis NA. Bloom’s syndrome: 453. Goldstein AM, Struewing JP, Chidambaram A, ical outcome in Fanconi anemia. European Fanconi
clinical spectrum, molecular pathogenesis, and cancer Fraser MC, Tucker MA. Genotype-phenotype Anemia Research Group. Blood. 2000;96(13):
predisposition. Mol Syndromol. 2017;8(1):4–23. relationships in U.S. melanoma-prone families with 4064–4070.
435. de Voer RM, Hahn MM, Mensenkamp AR, et al. CDKN2A and CDK4 mutations. J Natl Cancer Inst. 472. Brosh RM Jr, Bellani M, Liu Y, Seidman MM.
Deleterious germline BLM mutations and the risk for 2000;92(12):1006–1010. Fanconi Anemia: A DNA repair disorder character-
early-onset colorectal cancer. Sci Rep. 2015;5:14060. 454. Doros L, Schultz KA, Stewart DR, et al. DICER1- ized by accelerated decline of the hematopoietic
436. Prokofyeva D, Bogdanova N, Dubrowinskaja N, related disorders. In: Pagon RA, Adam MP, Ardinger stem cell compartment and other features of aging.
et al. Nonsense mutation p.Q548X in BLM, the HH, et al, eds. GeneReviews(R). Seattle (WA): Ageing Res Rev. 2017;33:67–75.
gene mutated in Bloom’s syndrome, is associated with University of Washington, Seattle University of 473. Pithukpakorn M, Toro JR. Hereditary leiomyoma-
breast cancer in Slavic populations. Breast Cancer Washington, Seattle. GeneReviews is a registered tosis and renal cell cancer. In: Pagon RA, Adam MP,
Res Treat. 2013;137(2):533–539. trademark of the University of Washington, Seattle. Ardinger HH, et al, eds. GeneReviews(R). Seattle
437. Larsen Haidle J, Howe JR. Juvenile polyposis All rights reserved; 1993. (WA): University of Washington, Seattle University
syndrome. In: Pagon RA, Adam MP, Ardinger HH, 455. Foulkes WD, Bahubeshi A, Hamel N, et al. of Washington, Seattle. GeneReviews is a registered
et al, eds. GeneReviews(R). Seattle (WA): University Extending the phenotypes associated with DICER1 trademark of the University of Washington, Seattle.
of Washington, Seattle University of Washington, mutations. Hum Mutat. 2011;32(12):1381–1384. All rights reserved; 1993.
Seattle. GeneReviews is a registered trademark of 456. Schultz KA, Pacheco MC, Yang J, et al. Ovarian sex 474. Stewart L, Glenn GM, Stratton P, et al. Association
the University of Washington, Seattle. All rights cord-stromal tumors, pleuropulmonary blastoma and of germline mutations in the fumarate hydratase
reserved; 1993. DICER1 mutations: a report from the International gene and uterine fibroids in women with hereditary
438. Syngal S, Brand RE, Church JM, Giardiello FM, Pleuropulmonary Blastoma Registry. Gynecol Oncol. leiomyomatosis and renal cell cancer. Arch Dermatol.
Hampel HL, Burt RW. ACG clinical guideline: 2011;122(2):246–250. 2008;144(12):1584–1592.
genetic testing and management of hereditary 457. Slade I, Bacchelli C, Davies H, et al. DICER1 475. Sanz-Ortega J, Vocke C, Stratton P, Linehan
gastrointestinal cancer syndromes. Am J Gastroenterol. syndrome: clarifying the diagnosis, clinical features WM, Merino MJ. Morphologic and molecular
2015;110(2):223–262, quiz 263. and management implications of a pleiotropic characteristics of uterine leiomyomas in hereditary
439. Brosens LA, Langeveld D, van Hattem WA, Giardi- tumour predisposition syndrome. J Med Genet. leiomyomatosis and renal cancer (HLRCC) syn-
ello FM, Offerhaus GJ. Juvenile polyposis syndrome. 2011;48(4):273–278. drome. Am J Surg Pathol. 2013;37(1):74–80.
World J Gastroenterol. 2011;17(44):4839–4844. 458. Faure A, Atkinson J, Bouty A, et al. DICER1 pleuro- 476. Schmidt LS, Linehan WM. Hereditary leiomyoma-
440. Jass JR. Colorectal polyposes: from phenotype to pulmonary blastoma familial tumour predisposition tosis and renal cell carcinoma. Int J Nephrol Renovasc
diagnosis. Pathol Res Pract. 2008;204(7):431–447. syndrome: what the paediatric urologist needs to Dis. 2014;7:253–260.
441. Chow E, Macrae F. A review of juvenile know. J Pediatr Urol. 2016;12(1):5–10. 477. Toro JR. Birt-Hogg-Dube syndrome. In: Pagon RA,
polyposis syndrome. J Gastroenterol Hepatol. 459. Schultz KAP, Rednam SP, Kamihara J, et al. Adam MP, Ardinger HH, et al, eds. GeneReviews(R).
2005;20(11):1634–1640. PTEN, DICER1, FH, and their associated tumor Seattle (WA): University of Washington, Seattle
442. Iyer NK, Burke CA, Leach BH, Parambil JG. susceptibility syndromes: clinical features, genetics, University of Washington, Seattle. GeneReviews is a
SMAD4 mutation and the combined syndrome of and surveillance recommendations in childhood. registered trademark of the University of Washington,
juvenile polyposis syndrome and hereditary haem- Clin Cancer Res. 2017;23(12):e76–e82. Seattle. All rights reserved; 1993.
orrhagic telangiectasia. Thorax. 2010;65(8):745– 460. Whitelaw SC, Murday VA, Tomlinson IP, et al. Clini- 478. Hasumi H, Baba M, Hasumi Y, Furuya M, Yao M.
746. cal and molecular features of the hereditary mixed Birt-Hogg-Dube syndrome: Clinical and molecular
Genetic Factors: Hereditary Cancer Predisposition Syndromes  •  CHAPTER 13 208.e9
208.e9

aspects of recently identified kidney cancer syndrome. Non-Polyposis Colorectal Cancer (ICG-HNPCC). 517. Evans DGR, Salvador H, Chang VY, et al. Cancer
Int J Urol. 2016;23(3):204–210. Dis Colon Rectum. 1991;34(5):424–425. and central nervous system tumor surveillance in
479. Kuroda N, Furuya M, Nagashima Y, et al. Review 499. Perez-Carbonell L, Ruiz-Ponte C, Guarinos C, et al. pediatric neurofibromatosis 1. Clin Cancer Res. 2017;
of renal tumors associated with Birt-Hogg-Dube Comparison between universal molecular screening 23(12):e46–e53.
syndrome with focus on clinical and pathobiological for Lynch syndrome and Revised Bethesda Guidelines 518. Evans DGR, Salvador H, Chang VY, et al. Cancer
aspects. Pol J Pathol. 2014;65(2):93–99. in a large population-based cohort of patients with and central nervous system tumor surveillance in
480. Furuya M, Nakatani Y. Birt-Hogg-Dube syndrome: colorectal cancer. Gut. 2012;61(6):865–872. pediatric neurofibromatosis 2 and related disorders.
clinicopathological features of the lung. J Clin Pathol. 500. South CD, Hampel H, Comeras I, Westman JA, Clin Cancer Res. 2017;23(12):e54–e61.
2013;66(3):178–186. Frankel WL, de la Chapelle A. The frequency of 519. Stadler ZK, Salo-Mullen E, Sabbaghian N, et al.
481. Forde PM, Cochran RL, Boikos SA, et al. Familial Muir-Torre syndrome among Lynch syndrome Germline PALB2 mutation analysis in breast-pancreas
GI stromal tumor with loss of heterozygosity and families. J Natl Cancer Inst. 2008;100(4):277–281. cancer families. J Med Genet. 2011;48(8):523–525.
amplification of mutant KIT. J Clin Oncol. 2016; 501. Hamilton SR, Liu B, Parsons RE, et al. The 520. Valle L, Hernandez-Illan E, Bellido F, et al. New
34(3):e13–e16. molecular basis of Turcot’s syndrome. N Engl J insights into POLE and POLD1 germline mutations
482. Ricci R. Syndromic gastrointestinal stromal tumors. Med. 1995;332(13):839–847. in familial colorectal cancer and polyposis. Hum
Hered Cancer Clin Pract. 2016;14:15. 502. Bakry D, Aronson M, Durno C, et al. Genetic and Mol Genet. 2014;23(13):3506–3512.
483. Ricci R, Martini M, Cenci T, et al. PDGFRA-mutant clinical determinants of constitutional mismatch 521. Church JM. Polymerase proofreading-associated
syndrome. Mod Pathol. 2015;28(7):954–964. repair deficiency syndrome: report from the con- polyposis: a new, dominantly inherited syndrome
484. Lasota J, Miettinen M. KIT and PDGFRA mutations stitutional mismatch repair deficiency consortium. of hereditary colorectal cancer predisposition. Dis
in gastrointestinal stromal tumors (GISTs). Semin Eur J Cancer. 2014;50(5):987–996. Colon Rectum. 2014;57(3):396–397.
Diagn Pathol. 2006;23(2):91–102. 503. Walsh MD, Buchanan DD, Cummings MC, 522. Elsayed FA, Kets CM, Ruano D, et al. Germline
485. Giusti F, Marini F, Brandi ML. Multiple endocrine et al. Lynch syndrome-associated breast cancers: variants in POLE are associated with early onset
neoplasia type 1. In: Pagon RA, Adam MP, Ardinger clinicopathologic characteristics of a case series mismatch repair deficient colorectal cancer. Eur J
HH, et al, eds. GeneReviews(R). Seattle (WA): from the colon cancer family registry. Clin Cancer Hum Genet. 2015;23(8):1080–1084.
University of Washington, Seattle University of Res. 2010;16(7):2214–2224. 523. Spier I, Holzapfel S, Altmuller J, et al. Frequency
Washington, Seattle. GeneReviews is a registered 504. Tabori U, Hansford JR, Achatz MI, et al. Clinical and phenotypic spectrum of germline mutations
trademark of the University of Washington, Seattle. management and tumor surveillance recommenda- in POLE and seven other polymerase genes in 266
All rights reserved; 1993. tions of inherited mismatch repair deficiency in patients with colorectal adenomas and carcinomas.
486. Machens A, Schaaf L, Karges W, et al. Age-related childhood. Clin Cancer Res. 2017;23(11):e32–e37. Int J Cancer. 2015;137(2):320–331.
penetrance of endocrine tumours in multiple 505. Cleary SP, Cotterchio M, Jenkins MA, et al. 524. Solomon S, Whitcomb DC, LaRusch J. PRSS1-
endocrine neoplasia type 1 (MEN1): a multicentre Germline MutY human homologue mutations related hereditary pancreatitis. In: Pagon RA, Adam
study of 258 gene carriers. Clin Endocrinol (Oxf ). and colorectal cancer: a multisite case-control study. MP, Ardinger HH, et al, eds. GeneReviews(R). Seattle
2007;67(4):613–622. Gastroenterology. 2009;136(4):1251–1260. (WA): University of Washington, Seattle University
487. Lemos MC, Thakker RV. Multiple endocrine 506. Lubbe SJ, Di Bernardo MC, Chandler IP, Houlston of Washington, Seattle. GeneReviews is a registered
neoplasia type 1 (MEN1): analysis of 1336 mutations RS. Clinical implications of the colorectal cancer risk trademark of the University of Washington, Seattle.
reported in the first decade following identification associated with MUTYH mutation. J Clin Oncol. All rights reserved; 1993.
of the gene. Hum Mutat. 2008;29(1):22–32. 2009;27(24):3975–3980. 525. Solomon S, Whitcomb DC. Genetics of pancreatitis:
488. Thakker RV, Newey PJ, Walls GV, et al. Clinical 507. Aretz S, Tricarico R, Papi L, et al. MUTYH- an update for clinicians and genetic counselors. Curr
practice guidelines for multiple endocrine neo- associated polyposis (MAP): evidence for the origin Gastroenterol Rep. 2012;14(2):112–117.
plasia type 1 (MEN1). J Clin Endocrinol Metab. of the common European mutations p.Tyr179Cys 526. Rosendahl J, Landt O, Bernadova J, et al. CFTR,
2012;97(9):2990–3011. and p.Gly396Asp by founder events. Eur J Hum SPINK1, CTRC and PRSS1 variants in chronic
489. Coleman JA, Russo P. Hereditary and familial kidney Genet. 2014;22(7):923–929. pancreatitis: is the role of mutated CFTR overesti-
cancer. Curr Opin Urol. 2009;19(5):478–485. 508. Nielsen M, Poley JW, Verhoef S, et al. Duodenal mated? Gut. 2013;62(4):582–592.
490. Rini BI, Campbell SC, Rathmell WK. Renal cell carcinoma in MUTYH-associated polyposis. J Clin 527. Evans DG, Farndon PA. Nevoid basal cell carcinoma
carcinoma. Curr Opin Oncol. 2006;18(3):289–296. Pathol. 2006;59(11):1212–1215. syndrome. In: Pagon RA, Adam MP, Ardinger HH,
491. de Vos tot Nederveen Cappel WH, Jarvinen 509. Win AK, Cleary SP, Dowty JG, et al. Cancer risks et al, eds. GeneReviews(R). Seattle (WA): University
HJ, Lynch PM, Engel C, Mecklin JP, Vasen HF. for monoallelic MUTYH mutation carriers with of Washington, Seattle University of Washington,
Colorectal surveillance in Lynch syndrome families. a family history of colorectal cancer. Int J Cancer. Seattle. GeneReviews is a registered trademark of
Fam Cancer. 2013;12(2):261–265. 2011;129(9):2256–2262. the University of Washington, Seattle. All rights
492. Bupathi M, Wu C. Biomarkers for immune therapy 510. Jenkins MA, Croitoru ME, Monga N, et al. Risk of reserved; 1993.
in colorectal cancer: mismatch-repair deficiency and colorectal cancer in monoallelic and biallelic carriers 528. Soufir N, Gerard B, Portela M, et al. PTCH
others. J Gastrointest Oncol. 2016;7(5):713–720. of MYH mutations: a population-based case-family mutations and deletions in patients with typical
493. Castro MP, Goldstein N. Mismatch repair deficiency study. Cancer Epidemiol Biomarkers Prev. 2006; nevoid basal cell carcinoma syndrome and in patients
associated with complete remission to combination 15(2):312–314. with a suspected genetic predisposition to basal cell
programmed cell death ligand immune therapy 511. Zhang G, Zeng Y, Liu Z, Wei W. Significant carcinoma: a French study. Br J Cancer. 2006;95(4):
in a patient with sporadic urothelial carcinoma: association between Nijmegen breakage syndrome 548–553.
immunotheranostic considerations. J Immunother 1 657del5 polymorphism and breast cancer risk. 529. Athar M, Li C, Kim AL, Spiegelman VS, Bickers
Cancer. 2015;3:58. Tumor Biol. 2013;34(5):2753–2757. DR. Sonic hedgehog signaling in Basal cell
494. Burn J, Gerdes AM, Macrae F, et al. Long-term 512. Dembowska-Baginska B, Perek D, Brozyna A, et al. nevus syndrome. Cancer Res. 2014;74(18):4967–
effect of aspirin on cancer risk in carriers of Non-Hodgkin lymphoma (NHL) in children with 4975.
hereditary colorectal cancer: an analysis from the Nijmegen Breakage syndrome (NBS). Pediatr Blood 530. Smith MJ, Beetz C, Williams SG, et al. Germline
CAPP2 randomised controlled trial. Lancet. 2011; Cancer. 2009;52(2):186–190. mutations in SUFU cause Gorlin syndrome-
378(9809):2081–2087. 513. Friedman JM. Neurofibromatosis 1. In: Pagon RA, associated childhood medulloblastoma and redefine
495. Shia J. Immunohistochemistry versus microsatellite Adam MP, Ardinger HH, et al, eds. GeneReviews(R). the risk associated with PTCH1 mutations. J Clin
instability testing for screening colorectal cancer Seattle (WA): University of Washington, Seattle Oncol. 2014;32(36):4155–4161.
patients at risk for hereditary nonpolyposis colorectal University of Washington, Seattle. GeneReviews is a 531. Eng C. PTEN Hamartoma tumor syndrome.
cancer syndrome. Part I. The utility of immunohis- registered trademark of the University of Washington, In: Pagon RA, Adam MP, Ardinger HH, et al,
tochemistry. J Mol Diagn. 2008;10(4):293–300. Seattle. All rights reserved; 1993. eds. GeneReviews(R). Seattle (WA): University of
496. Zhang L. Immunohistochemistry versus microsatellite 514. Madanikia SA, Bergner A, Ye X, Blakeley JO. Washington, Seattle University of Washington,
instability testing for screening colorectal cancer Increased risk of breast cancer in women with NF1. Seattle. GeneReviews is a registered trademark of the
patients at risk for hereditary nonpolyposis colorectal Am J Med Genet A. 2012;158a(12):3056–3060. University of Washington, Seattle. All rights reserved;
cancer syndrome. Part II. The utility of microsatellite 515. Seminog OO, Goldacre MJ. Risk of benign tumours 1993.
instability testing. J Mol Diagn. 2008;10(4):301–307. of nervous system, and of malignant neoplasms, 532. Nelen MR, Padberg GW, Peeters EA, et al. Localiza-
497. Bouzourene H, Hutter P, Losi L, Martin P, Benhat- in people with neurofibromatosis: population- tion of the gene for Cowden disease to chromosome
tar J. Selection of patients with germline MLH1 based record-linkage study. Br J Cancer. 2013; 10q22-23. Nat Genet. 1996;13(1):114–116.
mutated Lynch syndrome by determination of 108(1):193–198. 533. Tan MH, Mester J, Peterson C, et al. A clinical
MLH1 methylation and BRAF mutation. Fam 516. Walker L, Thompson D, Easton D, et al. A pro- scoring system for selection of patients for PTEN
Cancer. 2010;9(2):167–172. spective study of neurofibromatosis type 1 cancer mutation testing is proposed on the basis of a
498. Vasen HF, Mecklin JP, Khan PM, Lynch HT. The incidence in the UK. Br J Cancer. 2006;95(2):233– prospective study of 3042 probands. Am J Hum
International Collaborative Group on Hereditary 238. Genet. 2011;88(1):42–56.
208.e10 Part I: Science and Clinical Oncology

534. Heald B, Mester J, Rybicki L, Orloff MS, Burke 553. Eng C. Seminars in medicine of the Beth Israel families with p53 germline mutations. J Clin Oncol.
CA, Eng C. Frequent gastrointestinal polyps and Hospital, Boston. The RET proto-oncogene in mul- 2009;27(8):1250–1256.
colorectal adenocarcinomas in a prospective series tiple endocrine neoplasia type 2 and Hirschsprung’s 571. Bougeard G, Renaux-Petel M, Flaman JM, et al.
of PTEN mutation carriers. Gastroenterology. disease. N Engl J Med. 1996;335(13):943–951. Revisiting Li-Fraumeni syndrome from TP53 muta-
2010;139(6):1927–1933. 554. Kirmani S, Young WF. Hereditary paraganglioma- tion carriers. J Clin Oncol. 2015;33(21):2345–2352.
535. Eng C. PTEN: one gene, many syndromes. Hum pheochromocytoma syndromes. In: Pagon RA, Adam 572. Villani A, Shore A, Wasserman JD, et al. Biochemical
Mutat. 2003;22(3):183–198. MP, Ardinger HH, et al, eds. GeneReviews(R). Seattle and imaging surveillance in germline TP53 muta-
536. Pilarski R, Burt R, Kohlman W, Pho L, Shannon (WA): University of Washington, Seattle University tion carriers with Li-Fraumeni syndrome: 11 year
KM, Swisher E. Cowden syndrome and the PTEN of Washington, Seattle. GeneReviews is a registered follow-up of a prospective observational study. Lancet
hamartoma tumor syndrome: systematic review trademark of the University of Washington, Seattle. Oncol. 2016;17(9):1295–1305.
and revised diagnostic criteria. J Natl Cancer Inst. All rights reserved; 1993. 573. Kratz CP, Achatz MI, Brugieres L, et al. Cancer
2013;105(21):1607–1616. 555. Lenders JW, Duh QY, Eisenhofer G, et al. Pheo- screening recommendations for individuals with Li-
537. McBride KL, Varga EA, Pastore MT, et al. chromocytoma and paraganglioma: an Endocrine Fraumeni syndrome. Clin Cancer Res. 2017;23(11):
Confirmation study of PTEN mutations among Society clinical practice guideline. J Clin Endocrinol e38–e45.
individuals with autism or developmental delays/ Metab. 2014;99(6):1915–1942. 574. Northrup H, Koenig MK, Pearson DA, Au KS.
mental retardation and macrocephaly. Autism Res. 556. Gottlieb E, Tomlinson IP. Mitochondrial tumour Tuberous sclerosis complex. In: Pagon RA, Adam
2010;3(3):137–141. suppressors: a genetic and biochemical update. Nat MP, Ardinger HH, et al, eds. GeneReviews(R). Seattle
538. Haibach H, Burns TW, Carlson HE, Burman Rev Cancer. 2005;5(11):857–866. (WA): University of Washington, Seattle University
KD, Deftos LJ. Multiple hamartoma syndrome 557. Pai R, Manipadam MT, Singh P, Ebenazer A, of Washington, Seattle. GeneReviews is a registered
(Cowden’s disease) associated with renal cell car- Samuel P, Rajaratnam S. Usefulness of Succinate trademark of the University of Washington, Seattle.
cinoma and primary neuroendocrine carcinoma of dehydrogenase B (SDHB) immunohistochemistry All rights reserved; 1993.
the skin (Merkel cell carcinoma). Am J Clin Pathol. in guiding mutational screening among patients 575. Northrup H, Krueger DA. Tuberous sclerosis
1992;97(5):705–712. with pheochromocytoma-paraganglioma syndromes. complex diagnostic criteria update: recommenda-
539. Schrager CA, Schneider D, Gruener AC, Tsou HC, APMIS. 2014;122(11):1130–1135. tions of the 2012 International Tuberous Sclerosis
Peacocke M. Clinical and pathological features of 558. Rednam SP, Erez A, Druker H, et al. Von Complex Consensus Conference. Pediatr Neurol.
breast disease in Cowden’s syndrome: an under- Hippel-Lindau and Hereditary pheochromocytoma/ 2013;49(4):243–254.
recognized syndrome with an increased risk of breast paraganglioma syndromes: clinical features, genetics, 576. Patel U, Simpson E, Kingswood JC, Saggar-Malik
cancer. Hum Pathol. 1998;29(1):47–53. and surveillance recommendations in childhood. AK. Tuberose sclerosis complex: analysis of growth
540. Pilarski R. Cowden syndrome: a critical review of the Clin Cancer Res. 2017;23(12):e68–e75. rates aids differentiation of renal cell carcinoma from
clinical literature. J Genet Couns. 2009;18(1):13–27. 559. McGarrity TJ, Amos CI, Baker MJ. Peutz-Jeghers atypical or minimal-fat-containing angiomyolipoma.
541. Meindl A, Hellebrand H, Wiek C, et al. Germline syndrome. In: Pagon RA, Adam MP, Ardinger HH, Clin Radiol. 2005;60(6):665–673, discussion
mutations in breast and ovarian cancer pedigrees et al, eds. GeneReviews(R). Seattle (WA): University 663–664.
establish RAD51C as a human cancer susceptibility of Washington, Seattle University of Washington, 577. Crino PB, Nathanson KL, Henske EP. The tuberous
gene. Nat Genet. 2010;42(5):410–414. Seattle. GeneReviews is a registered trademark of sclerosis complex. N Engl J Med. 2006;355(13):
542. Loveday C, Turnbull C, Ruark E, et al. Germline the University of Washington, Seattle. All rights 1345–1356.
RAD51C mutations confer susceptibility to ovarian reserved; 1993. 578. Borkowska J, Schwartz RA, Kotulska K, Jozwiak S.
cancer. Nat Genet. 2012;44(5):475–476, author reply 560. Beggs AD, Latchford AR, Vasen HF, et  al. Tuberous sclerosis complex: tumors and tumorigen-
476. Peutz-Jeghers syndrome: a systematic review and esis. Int J Dermatol. 2011;50(1):13–20.
543. Lohmann DR, Gallie BL. Retinoblastoma. In: recommendations for management. Gut. 2010;59(7): 579. Al-Saleem T, Wessner LL, Scheithauer BW, et al.
Pagon RA, Adam MP, Ardinger HH, et al, eds. 975–986. Malignant tumors of the kidney, brain, and soft
GeneReviews(R). Seattle (WA): University of 561. Tomas C, Soyer P, Dohan A, Dray X, Boudiaf M, tissues in children and young adults with the tuberous
Washington, Seattle University of Washington, Hoeffel C. Update on imaging of Peutz-Jeghers sclerosis complex. Cancer. 1998;83(10):2208–2216.
Seattle. GeneReviews is a registered trademark of syndrome. World J Gastroenterol. 2014;20(31): 580. Au KS, Williams AT, Roach ES, et al. Genotype/
the University of Washington, Seattle. All rights 10864–10875. phenotype correlation in 325 individuals referred
reserved; 1993. 562. Giardiello FM, Trimbath JD. Peutz-Jeghers syn- for a diagnosis of tuberous sclerosis complex in the
544. Knudson AG Jr. Mutation and cancer: statistical drome and management recommendations. Clin United States. Genet Med. 2007;9(2):88–100.
study of retinoblastoma. Proc Natl Acad Sci USA. Gastroenterol Hepatol. 2006;4(4):408–415. 581. Qin W, Kozlowski P, Taillon BE, et al. Ultra deep
1971;68(4):820–823. 563. Jasperson K, Burt RW. The genetics of colorectal sequencing detects a low rate of mosaic muta-
545. Abramson DH, Beaverson K, Sangani P, et al. cancer. Surg Oncol Clin N Am. 2015;24(4):683–703. tions in tuberous sclerosis complex. Hum Genet.
Screening for retinoblastoma: presenting signs 564. Stoffel EM, Mangu PB, Gruber SB, et al. Hereditary 2010;127(5):573–582.
as prognosticators of patient and ocular survival. colorectal cancer syndromes: American Society of 582. Spurling Jeste S, Wu JY, Senturk D, et al. Early
Pediatrics. 2003;112(6 Pt 1):1248–1255. Clinical Oncology Clinical Practice Guideline developmental trajectories associated with ASD in
546. Abramson DH, Frank CM. Second nonocular tumors endorsement of the familial risk-colorectal cancer: infants with tuberous sclerosis complex. Neurology.
in survivors of bilateral retinoblastoma: a possible European Society for Medical Oncology Clinical 2014;83(2):160–168.
age effect on radiation-related risk. Ophthalmology. Practice Guidelines. J Clin Oncol. 2015;33(2): 583. Dworakowska D, Grossman AB. Are neuroendocrine
1998;105(4):573–579, discussion 579–580. 209–217. tumours a feature of tuberous sclerosis? A systematic
547. Dimaras H, Kimani K, Dimba EA, et al. Retino- 565. Schneider K, Zelley K, Nichols KE, Garber J. review. Endocr Relat Cancer. 2009;16(1):45–
blastoma. Lancet. 2012;379(9824):1436–1446. Li-Fraumeni syndrome. In: Pagon RA, Adam MP, 58.
548. Kleinerman RA, Yu CL, Little MP, et al. Variation Ardinger HH, et al, eds. GeneReviews(R). Seattle 584. Frantzen C, Klasson TD, Links TP, Giles RH, et al.
of second cancer risk by family history of retino- (WA): University of Washington, Seattle University Von Hippel-Lindau syndrome. In: Pagon RA, Adam
blastoma among long-term survivors. J Clin Oncol. of Washington, Seattle. GeneReviews is a registered MP, Ardinger HH, eds. GeneReviews(R). Seattle
2012;30(9):950–957. trademark of the University of Washington, Seattle. (WA): University of Washington, Seattle University
549. Klutz M, Brockmann D, Lohmann DR. A parent- All rights reserved; 1993. of Washington, Seattle. GeneReviews is a registered
of-origin effect in two families with retinoblastoma 566. Li FP, Fraumeni JF Jr. Soft-tissue sarcomas, trademark of the University of Washington, Seattle.
is associated with a distinct splice mutation in the breast cancer, and other neoplasms. A familial All rights reserved; 1993.
RB1 gene. Am J Hum Genet. 2002;71(1):174–179. syndrome? Ann Intern Med. 1969;71(4):747–752. 585. Binderup ML, Bisgaard ML, Harbud V, et al. Von
550. Price EA, Price K, Kolkiewicz K, et al. Spectrum 567. Li FP, Fraumeni JF Jr, Mulvihill JJ, et al. A cancer Hippel-Lindau disease (vHL). National clinical
of RB1 mutations identified in 403 retinoblastoma family syndrome in twenty-four kindreds. Cancer guideline for diagnosis and surveillance in Denmark.
patients. J Med Genet. 2014;51(3):208–214. Res. 1988;48(18):5358–5362. 3rd edition. Dan Med J. 2013;60(12):B4763.
551. Kamihara J, Bourdeaut F, Foulkes WD, et al. Reti- 568. Srivastava S, Zou ZQ, Pirollo K, Blattner W, Chang 586. Kwon T, Jeong IG, Pak S, et al. Renal tumor size is
noblastoma and neuroblastoma predisposition and EH. Germ-line transmission of a mutated p53 gene an independent prognostic factor for overall survival
surveillance. Clin Cancer Res. 2017;23(13):e98–e106. in a cancer-prone family with Li-Fraumeni syndrome. in von Hippel-Lindau disease. J Cancer Res Clin
552. Marquard J, Eng C. Multiple endocrine neoplasia Nature. 1990;348(6303):747–749. Oncol. 2014;140(7):1171–1177.
type 2. In: Pagon RA, Adam MP, Ardinger HH, 569. Chompret A, Abel A, Stoppa-Lyonnet D, et al. 587. Grubb RL 3rd, Choyke PL, Pinto PA, Linehan
et al, eds. GeneReviews(R). Seattle (WA): University Sensitivity and predictive value of criteria for WM, Walther MM. Management of von Hippel-
of Washington, Seattle University of Washington, p53 germline mutation screening. J Med Genet. Lindau-associated kidney cancer. Nat Clin Pract Urol.
Seattle. GeneReviews is a registered trademark of 2001;38(1):43–47. 2005;2(5):248–255.
the University of Washington, Seattle. All rights 570. Gonzalez KD, Noltner KA, Buzin CH, et al. Beyond 588. Corcos O, Couvelard A, Giraud S, et al. Endocrine
reserved; 1993. Li Fraumeni syndrome: clinical characteristics of pancreatic tumors in von Hippel-Lindau disease:
Genetic Factors: Hereditary Cancer Predisposition Syndromes  •  CHAPTER 13 208.e11
208.e11

clinical, histological, and genetic features. Pancreas. 596. Evans DG, Birch JM, Thorneycroft M, McGown 604. Athma P, Rappaport R, Swift M. Molecular genotyp-
2008;37(1):85–93. G, Lalloo F, Varley JM. Low rate of TP53 germline ing shows that ataxia-telangiectasia heterozygotes are
589. Blansfield JA, Choyke L, Morita SY, et al. Clinical, mutations in breast cancer/sarcoma families not predisposed to breast cancer. Cancer Genet Cytogenet.
genetic and radiographic analysis of 108 patients fulfilling classical criteria for Li-Fraumeni syndrome. 1996;92(2):130–134.
with von Hippel-Lindau disease (VHL) manifested J Med Genet. 2002;39(12):941–944. 605. Berstein JL, Concannon P, Langholz B, et al. Multi-
by pancreatic neuroendocrine neoplasms (PNETs). 597. Masciari S, Dillon DA, Rath M, et al. Breast cancer center screening of mutations in the ATM gene
Surgery. 2007;142(6):814–818, discussion 818. phenotype in women with TP53 germline mutations: among women with breast cancer—the WECARE
e811–818.e812. a Li-Fraumeni syndrome consortium effort. Breast Study. Radiat Res. 2005;163(6):698–699.
590. Ford D, Easton DF, Bishop DT, Narod SA, Cancer Res Treat. 2012;133(3):1125–1130. 606. Bretsky P, Haiman CA, Gilad S, et al. The relation-
Goldgar DE. Risks of cancer in BRCA1-mutation 598. Hwang SJ, Lozano G, Amos CI, Strong LC. ship between twenty missense ATM variants and
carriers. Breast Cancer Linkage Consortium. Lancet. Germline p53 mutations in a cohort with childhood breast cancer risk: the Multiethnic Cohort. Cancer
1994;343(8899):692–695. sarcoma: sex differences in cancer risk. Am J Hum Epidemiol Biomarkers Prev. 2003;12(8):733–738.
591. Satagopan JM, Offit K, Foulkes W, et al. The lifetime Genet. 2003;72(4):975–983. 607. Thompson D, Seal S, Schutte M, et al. A multicenter
risks of breast cancer in Ashkenazi Jewish carriers of 599. Brownstein MH, Wolf M, Bikowski JB. Cowden’s study of cancer incidence in CHEK2 1100delC
BRCA1 and BRCA2 mutations. Cancer Epidemiol disease: a cutaneous marker of breast cancer. Cancer. mutation carriers. Cancer Epidemiol Biomarkers Prev.
Biomarkers Prev. 2001;10(5):467–473. 1978;41(6):2393–2398. 2006;15(12):2542–2545.
592. Lalloo F, Varley J, Ellis D, et al. Prediction of patho- 600. Starink TM, van der Veen JP, Arwert F, et al. The 608. Shaag A, Walsh T, Renbaum P, et al. Functional and
genic mutations in patients with early-onset breast Cowden syndrome: a clinical and genetic study in genomic approaches reveal an ancient CHEK2 allele
cancer by family history. Lancet. 2003;361(9363): 21 patients. Clin Genet. 1986;29(3):222–233. associated with breast cancer in the Ashkenazi Jewish
1101–1102. 601. Zbuk K, Stein JL, Eng C. PTEN Hamartoma tumor population. Hum Mol Genet. 2005;14(4):555–563.
593. Chen S, Iversen ES, Friebel T, et al. Characterization syndrome (PHTS). Gene Reviews at GeneTests: 609. Rahman N, Seal S, Thompson D, et al. PALB2,
of BRCA1 and BRCA2 mutations in a large United Medical Genetics Information Resource; 2013. which encodes a BRCA2-interacting protein, is
States sample. J Clin Oncol. 2006;24(6):863–871. http://www.genetests.org. a breast cancer susceptibility gene. Nat Genet.
594. Birch JM, Alston RD, McNally RJ, et al. Relative 602. Bubien V, Bonnet F, Brouste V, et al. High cumulative 2007;39(2):165–167.
frequency and morphology of cancers in carriers of risks of cancer in patients with PTEN hamartoma 610. Bonadona V, Bonaiti B, Olschwang S, et al. Cancer
germline TP53 mutations. Oncogene. 2001;20(34): tumour syndrome. J Med Genet. 2013;50(4): risks associated with germline mutations in MLH1,
4621–4628. 255–263. MSH2, and MSH6 genes in Lynch syndrome.
595. Chompret A, Brugieres L, Ronsin M, et al. P53 603. Yoon KA, Ku JL, Yang HK, Kim WH, Park SY, JAMA. 2011;305(22):2304–2310.
germline mutations in childhood cancers and cancer Park JG. Germline mutations of E-cadherin gene 611. Watson P, Vasen HF, Mecklin JP, et al. The risk of
risk for carrier individuals. Br J Cancer. 2000;82(12): in Korean familial gastric cancer patients. J Hum extra-colonic, extra-endometrial cancer in the Lynch
1932–1937. Genet. 1999;44(3):177–180. syndrome. Int J Cancer. 2008;123(2):444–449.
Genetic and Epigenetic Alterations in Cancer 14 
Bin Tean Teh and Eric R. Fearon

S UMMARY OF K EY P OI NT S
• A root cause of cancer is the and epigenetic defects present in a inhibitory cues; (3) an increased
accumulation of genetic and cancer cell. Clonal selection is mutation rate to allow for the rapid
epigenetic defects in key cellular essentially an evolutionary process generation of new variant daughter
pathways regulating proliferation, that promotes outgrowth of cells; (4) the ability to attract and
differentiation, and death. The precancerous and cancerous cells support a new blood supply
defects in cancer cells are of two carrying those mutations and gene (angiogenesis); (5) the capacity to
types: gain-of-function alterations expression changes that confer the minimize an immune response and/
affecting oncogenes, and loss-of- most potent proliferative and survival or evade destruction by immune
function alterations affecting tumor properties on the cancer cells in a effector cells; (6) the capacity for
suppressor genes. Regardless of given context. essentially limitless cell division;
whether the defects are genetic or • Although a sizeable and diverse (7) a failure to respect tissue
epigenetic in nature, a common net array of mutations and gene boundaries, allowing for invasion into
consequence is dysregulation of expression changes have been adjacent tissues and organs with
gene expression in cancer cells. This implicated in cancer pathogenesis, microenvironments that are markedly
intimate relationship between genetic the defects appear to affect a more different from the one where the
and epigenetic changes has been limited number of conserved cancer cells arose; (8) the ability to
further confirmed by findings of signaling pathways or networks. escape cell death; (9) altered cell
frequent mutations in chromatin A relatively small collection of metabolism to support uncontrolled
enzymes. oncogenes and tumor suppressor proliferation; and (10) the acquisition
• More recently, genetic alterations in genes is recurrently deranged in of immune cells that promote tumor
noncoding DNA have also been cancer cells of various types and progression.1
reported in cancer tissues. They includes the RAS, PIK3CA, EGFR, • Certain gene defects in cancer cells
contribute to tumorigenesis by RAF, β-catenin, IDH1, and MYC may contribute to a few or perhaps
affecting the regulatory elements oncogene proteins and the p53, even only one of the signature traits.
(e.g., promoters, enhancers) that p16Ink4a, ARF, RB1, PTEN, APC, NF1, However, many of the gene defects
influence gene expression of key and ARID1A tumor suppressor and expression changes might have
cancer-related genes, and some proteins. The proteins that are been selected for in large part
of them may correspond to recurrently targeted by mutations in because they exert pleiotropic
cancer risk genetic variants in cancer likely represent particularly effects on the cancer cell
noncoding regions identified by critical hubs in the cell’s regulatory phenotype.
genome-wide association studies circuitry. • Despite the fact that some gene
(GWASs). • Although cancer represents a very defects may arise early in the
• In addition to genetic mutations and heterogeneous collection of development of certain cancer types,
genomic alterations, it is becoming diseases, the development of all advanced cancer cells might still be
more evident that clinical and cancers, regardless of type, appears critically dependent on the “early
pathologic studies indicate that to be critically dependent on the gene defects” for continued growth,
many cancers arise from preexisting acquisition of certain traits that allow survival, and even metastasis.
benign lesions, and numerous the cancer cells to grow in an Studies have shown that metastatic
cooperating genetic and epigenetic unchecked fashion in their tissue of cancers still harbor the same genetic
defects affecting multiple origin and to grow as metastatic alterations as the primary tumors.
independent signaling pathways lesions in distant sites in the body. Such findings imply that agents that
are likely needed for development Signature traits that are likely to be specifically target key signaling
of most clinically recognizable inherent in the majority of, if not all, pathways and proteins could
cancers. cancers include (1) an enhanced have use in advanced cancers,
• A process termed clonal selection response to proliferative and even if the signaling pathway defect
has a key role in determining the growth-promoting signals; (2) a arose very early in cancer
particular constellation of genetic relative resistance to growth development.
Continued

209
210 Part I: Science and Clinical Oncology

• Genomic and epigenomic • Future studies will further clarify the and more specific strategies for
characterization of organ site cancer role of the diverse array of genetic cancer detection, diagnosis, and
has identified a number of subtypes and epigenetic defects in cancer therapeutic targeting of cancer
with different prognoses and phenotypes, even at the level of a cells.
therapeutic relevance. single cell, allowing more definitive

A genetic basis for human cancer has been recognized for perhaps the term antioncogene was sometimes used with respect to the tumor
more than a century and has been supported by data from familial suppressor gene class. The term suggests that the primary function
and epidemiologic studies and animal studies. However, only during of the genes might be to act in direct opposition to activated oncogenes.
the past three decades has definitive molecular evidence been accu- Although many of the proteins that are encoded by tumor suppressor
mulated to support the view that all cancer types arise from defects genes do in fact bind to and regulate the function of proto-oncogenes
in the structure and/or regulation of genes. Studies from many different or function in pathways that regulate proto-oncogene activity, it is
fields, including tumor virology, chemical carcinogenesis, molecular not by necessity a general principle. Hence, genes that contribute to
biology and biochemistry, somatic cell genetics, developmental biology, cancer by virtue of their inactivation or loss-of-function mutations
and genetic epidemiology, have all played critical contributing roles in human cancers are referred to here as tumor suppressor genes. Similarly
in clarifying the contributions of genetic and epigenetic mechanisms to the proto-oncogenes, the normal functions of tumor suppressor
to cancer development. Although environmental and dietary factors genes are diverse, and the proteins encoded by these genes are found
have substantial roles in cancer development, it is now well established in essentially all compartments of the cell.
that the accumulation of multiple genetic and epigenetic alterations In addition to the well-established role of oncogene and tumor
in a single cell plays a fundamental role in cancer initiation and suppressor gene mutations in cancer, it is now abundantly clear that
progression. epigenetic mechanisms play critical roles in altering the patterns and
The mutations that arise in cancer cells can be divided into two levels of expression of certain proto-oncogenes and tumor suppressor
functionally distinct classes: oncogene and tumor suppressor gene genes in cancer. For instance, in some cancers, altered transcriptional
mutations. In addition to the classic alterations of oncogenes and regulatory mechanisms can lead to markedly increased levels of proto-
tumor suppressor genes, other genetic alterations occur in genes regulat- oncogene expression, akin to the level seen in cancer cells with
ing transcription and translation, as well as in genes responsible for mutational defects that alter the structure or copy number of the
DNA repair. Although localized mutations are commonly observed, proto-oncogene. Conversely, gene-silencing mechanisms can exert
other genetic variations are also important in oncogenesis, such as dramatic effects on the expression of certain tumor suppressor genes
copy number variation, deletion, and translocations. It is becoming in cancer cells, essentially rendering the genes functionally inactive
more evident that these genetic changes could occur in both coding in the absence of any mutations. It is interesting that sequence-based
and noncoding DNA and that the latter may involve cis-regulatory analyses of many different cancer types have revealed that many of
regions affecting gene transcription. All of these genomic and epi­ the mutations in cancer cells directly affect the cancer epigenome.
genomic alterations in the initiation and progression of cancer can Specifically, the oncogenes and tumor suppressor gene mutations in
be broadly considered as oncogenic or “gain of function” and suppressor cancer cells often target transcription factors, chromatin modifying
or “loss of function.” In addition, some of these genetic alterations proteins, and other chromatin-associated proteins, leading to potentially
may be present in an individual’s germline and may predispose to quite dramatic global effects on the structure and activity of chromatin,
particular cancers. Such mutations can also be passed on to future DNA methylation, histone modifications, and patterns of gene expres-
generations. The nature and role of certain germline mutations in sion in cancer cells.
cancer development are of great interest to cancer biologists because Finally, mutations in genes that regulate the recognition and repair
the mutations provide powerful clues about the identity of genes and of DNA damage also play critical roles in tumorigenesis. The DNA
pathways that play particularly critical roles in the malignant conversion damage recognition and repair genes could be considered to constitute
of cells. Nevertheless, germline mutations in oncogenes or tumor a distinct class of cancer genes, because at least some of the DNA
suppressor genes likely have a major contributing role in the develop- repair proteins might have a more passive role in cell proliferation,
ment of only hereditary cancer, representing a small fraction of all differentiation, and cell survival. Their inactivation in tumor cells
cancers, and the vast majority of mutations in cancer are somatic (i.e., might lead predominantly to the acquisition of a “mutator phenotype,”
present only in the tumor cells). with a resultant increased rate of mutations in other cellular genes.
A few of the cellular genes that are recurrently affected by inherited More recently, these tumors with higher mutation load, and therefore
and somatic mutations in human cancer will be discussed in more presumably generating more tumor-related neoantigens, were found
detail later. Brief mention will be made here of some general properties to be more responsive to checkpoint immunotherapy.2,3
of the mutated genes. Oncogenes act in a positive fashion to promote Given the enormous advances during the past two decades, especially
tumorigenesis. Their normally functioning cellular counterparts, termed the recent use of next-generation sequencing (NGS) in profiling and
proto-oncogenes, have been found to be important regulators of many characterizing gene mutations and expression defects in cancer, it will
aspects of cell physiology. The proteins encoded by various proto- not be possible in this chapter to review in a comprehensive fashion
oncogenes can be found in virtually all subcellular compartments. the vast collection of genetic and epigenetic defects that have been
The term proto-oncogene does not imply that genes of this class lie catalogued in human cancers. Nor will it be possible to discuss in
dormant in the cell with the purpose of promoting tumorigenesis. detail the possible contributions of the many different gene defects
Rather, the terminology reflects the fact that mutations in cancer cells to alterations in cell signaling and cell physiology. Rather, the primary
alter the normal structure and/or expression pattern of the proto- aim of this chapter will be to offer a framework for understanding
oncogene, generating oncogenic variants with altered function. In the relationships between genetic and epigenetic defects in cancer
genetic terms, oncogenic alleles have gain-of-function mutations that cells and the impact of the accumulated defects on the cancer cell
confer enhanced or novel functions. phenotype. Although some details on the identity and nature of gene
In contrast to the activating mutations in oncogenes, tumor sup- defects in cancer will be offered here, the emphasis will be on concepts
pressor genes harbor loss-of-function defects in cancer cells. Historically, with biologic and clinical significance.
Genetic and Epigenetic Alterations in Cancer  •  CHAPTER 14 211

RECURRENT MUTATIONAL TARGETS cytoplasmic signal transducers, and nuclear proteins, such as transcrip-
IN CANCER tion factors. Second, although some oncogene mutations may be
unique to cancers of a particular type, such as the specific chromosomal
Oncogenic variant alleles that are present in cancer are generated translocations and resultant fusion proteins that are seen in cancers
from the normal counterpart proto-oncogenes by various mutational of hematopoietic origin (e.g., the BCR-ABL translocation that is seen
mechanisms, including point or localized mutations, gross chromo- in chronic myelogenous leukemia and a subset of acute lymphoid
somal rearrangements, or gene amplification. Some representative leukemias and the PML-RARα translocation that is seen in acute
oncogene mutations in human cancer are summarized in Table 14.1. promyelocytic leukemia), other mutations, such as those affecting the
From a brief review of the data in Table 14.1, some generalizations KRAS, β-catenin, and c-MYC genes, are found in a broad spectrum
can be offered. First, the mutations affect proteins functioning in of different cancer types. Third, oncogene mutations in cancer are
various compartments of the cell, including growth factor receptors, nearly always somatic, because only a very limited number of germline

Table 14.1  Selected Oncogene Mutations in Cancer


Gene Activation Mechanism Protein Properties Tumor Types
KRAS Point mutation Signal transducer Pancreatic, colorectal, lung (adeno), endometrial,
other carcinomas
NRAS Point mutation Signal transducer Myeloid leukemia, colorectal cancer
HRAS Point mutation Signal transducer Bladder carcinoma
EGFR (ERBB) Amplification, mutation Growth factor (EGF) receptor Gliomas, lung (non–small cell) carcinoma
NEU (HER2/ERBB2) Amplification Growth factor receptor Breast, ovarian, gastric, other carcinomas
c-MYC Chromosome translocation Transcription factor Burkitt lymphomas
Amplification Small cell lung carcinoma (SCLC); other carcinomas;
glioblastoma
MYCN Amplification Transcription factor Neuroblastoma, SCLC; glioblastoma
MYCL1 Amplification Transcription factor SCLC, ovarian carcinoma
BCL2 Chromosome translocation Antiapoptosis protein B-cell lymphoma (follicular type)
CCND1 Amplification Cyclin D, cell cycle control Breast and other carcinomas
Chromosome translocation B-cell lymphoma, parathyroid adenoma
BCR-ABL Chromosome translocation Chimeric nonreceptor tyrosine kinase CML, ALL (T cell)
RET Chromosome translocation GDNF receptor tyrosine kinase Thyroid cancer (papillary type)
Point mutation Thyroid cancer (medullary type: germline mutations)
CDK4 Amplification
Point mutation Cyclin-dependent kinase Sarcoma, glioblastoma
MET Point mutation Hepatocyte growth factor (HGF) Renal carcinoma (papillary type: germline mutations)
receptor
SMO Point mutations Transmembrane signaling molecule Basal cell skin cancer
in Sonic Hedgehog pathway
CTNNB1 (β-CAT) Point mutation, in-frame deletion Transcriptional coactivator, links Melanoma; colorectal, endometrial, ovarian,
E-cadherin to cytoskeleton hepatocellular, and other carcinomas,
hepatoblastoma, Wilms tumor
FGF4 Amplification Growth factor (FGF-like) Gastric carcinoma
PML-RARA Chromosome translocation Chimeric transcription factor APL
TCF3-PBX1 Chromosome translocation Chimeric transcription factor Pre-B ALL
MDM2 Amplification p53 binding protein Sarcoma
GLI1 Amplification Transcription factor Sarcoma, glioma
TTG2 Chromosome translocation Transcription factor T-cell ALL
AKT2 Amplification Signal transducer (serine/threonine Pancreatic and ovarian carcinoma
kinase; downstream effector of PI3K)
PIK3CA Amplification Catalytic subunit of PI3K Ovarian carcinoma
AURKA Amplification Centrosome-associated kinase Breast, colon, ovarian, and prostate carcinomas
gliomas
TMPRSS2-ERG Chromosome translocation Transcription factor (ETS family) Prostate cancer
TMPRSS2-ETV1
TMPRSS2-ETV4

ALL, Acute lymphocytic leukemia; APL, acute promyelocytic leukemia; CML, chronic myelogenous leukemia; EGF, epidermal growth factor; FGF, fibroblast growth factor; GDNF,
glial-derived neurotrophic factor; HGF, hepatocyte growth factor; PI3K, phosphatidylinositol 3-kinase; SCLC, small cell lung carcinoma.
212 Part I: Science and Clinical Oncology

mutations in proto-oncogenes have been linked to cancer predisposi-


tion thus far. Fourth, some proto-oncogenes, such as KRAS or BCL2, CANCERS ARISE FROM THE
are somatically altered in cancer by a single mutational mechanism, ACCUMULATION OF MULTIPLE GENETIC
namely, point mutations in the KRAS gene and chromosomal transloca- AND EPIGENETIC DEFECTS
tions affecting the BCL2 gene. In contrast, other proto-oncogenes,
such as c-MYC, may be activated by more than one mechanism in On the basis of a simple consideration that a cancer involves a large
cancer, including chromosomal translocation, gene amplification, number of genetic and epigenetic alterations that arise in normal cells
and, more recently discovered, association with superenhancer. These during the many years of life, it would seem quite unlikely that any
mutational mechanisms lead to increased levels of c-MYC transcripts adult cancer may arise as the result of single gene defect. Even in
and protein. In fact, specific missense mutations at threonine 58 persons who are strongly predisposed to cancer as a result of a germline
of the c-MYC gene in some lymphomas may further enhance mutation in a specific oncogene or tumor suppressor gene, the vast
c-MYC protein levels by abrogating a phosphorylation-ubiquitination majority of cells in a person never develop into cancer or even display
mechanism targeting c-MYC for proteosomal degradation.4 However, definitive morphologic changes akin to those that are seen in benign
enhanced c-MYC protein levels can also result from alterations in tumors. Therefore, any model for cancer must incorporate these data,
c-MYC–specific microRNAs, methylation, and other noncoding which suggest that cancers likely arise as the result of the accumulation
regulatory elements functioning to regulate c-MYC transcription and of multiple genetic and epigenetic defects in an affected cell. Another
translation. issue to consider is that clinical and histopathologic data indicate that
In contrast to the activating mutations that generate oncogenic the development of nearly all cancers, regardless of the organ site, is
alleles from proto-oncogenes, inactivation of the normal function of often, if not invariably, preceded by precancerous phases or stages in
tumor suppressor genes is critical in tumorigenesis. Akin to the proto- which the neoplastic cells manifest increasingly disordered patterns
oncogenes, the functions of tumor suppressor genes are diverse, and of differentiation and morphology.
proteins that are encoded by these genes reside in practically all subcel- Given this background, it would appear that cancers arise from
lular compartments (Table 14.2). Many tumor suppressor genes were accumulated defects affecting multiple genes and pathways, and
identified by virtue of the fact that they are mutated in the germline precancerous (benign) precursor lesions harbor fewer of the key gene
of persons who are affected by a known mendelian cancer syndrome defects than established cancers. One question, therefore, is how many
or who at the very least display a markedly elevated risk of cancer. rate-limiting defects or “hits” are required for cancer development.
The link between a germline-inactivating mutation in a purported Although a definitive answer to this question cannot be given at this
tumor suppressor gene and increased cancer predisposition provides point, some estimates can be offered. Most common cancers show
very persuasive evidence of the functional significance of the gene in dramatically increased incidence with increasing age. On the basis of
the cancer process. Nevertheless, for the vast majority of tumor sup- analysis of the age-specific incidence of a number of common cancers
pressor genes, in terms of their magnitude, somatic inactivating and some straightforward assumptions about the rate of mutations
mutations play a far more significant role in cancer development than and the size of the target cell population, it was argued as early as the
do germline mutations. mid-1950s that most common epithelial cancers arise as the result
Another important point to consider is that much of the attention of four to seven rate-limiting events.5,9 It was inferred that these
for tumor suppressor genes has been focused on demonstrating that rate-limiting events might represent mutational events. Moreover,
cancer cells carry biallelic inactivating mutations. Clearly, a diverse benign lesions harboring fewer gene defects are often shown to have
array of mechanisms can inactivate gene function, including nonsense, an age-incidence distribution that was shifted roughly one to two
frameshift, and nonconservative missense mutations, as well as splicing decades earlier in life than cancers arising in the corresponding organ
mutations and gross deletions of the gene or even the chromosome or tissue site. Nevertheless, confounding the use of age-incidence
region that contains the gene. In a number of cases, studies of the data to model the number of rate-limiting mutations were questions
chromosomal mechanisms associated with tumor suppressor gene about the weight of importance of certain key biologic pathways
inactivation in cancer tissues, such as loss of the parental heterozygosity underlying the multiple hits models. On the other hand, studies of
(i.e., loss of heterozygosity) that is present in normal tissues, have the genetic alterations of pediatric cancers that occur much earlier
even been used to infer the existence of tumor suppressor genes, in and are perhaps less complex in terms of their genetic signature may
particular, chromosomal regions, before the actual identification of offer insights into key cancer target genes (see Chapter 92 on pediatric
the tumor suppressor gene of interest. The emphasis on defining solid tumors).
biallelic inactivating mutations in tumor suppressor genes has been
stimulated in large part by the Knudson hypothesis,5,6 which predicted CLONAL SELECTION AND EVOLUTION
that recessive genetic determinants played a critical role in retinoblas- IN CANCER
toma and many other cancers and that inactivation of both alleles of
a tumor suppressor gene was needed to abrogate tumor suppressor As previously noted, most if not all cancers arise from preexisting
gene activity. Nevertheless, as will be discussed in a bit more detail precancerous populations of cells, and multiple genetic and epigenetic
in the following sections, a variety of observations indicate that epi- defects are likely needed for conversion of a normal cell to a cancerous
genetic (nonmutational) mechanisms might play a prominent role in cell. Molecular studies of various cancer types and their corresponding
inactivating tumor suppressor gene function in sporadic tumors. associated precancerous lesions have yielded some fundamental insights
Furthermore, for certain tumor suppressor genes, inactivation of only into the processes likely to be critical in the emergence of the cancer.
one of the two alleles of a tumor suppressor gene might significantly First, although normal tissues and tissues from noncancerous disease
impair cell growth regulation or programmed cell death. For example, states display polyclonal (balanced) cell populations, the neoplastic
a number of mutant p53 proteins that carry missense mutations likely component that is present in benign lesions and cancers displays a
potently interfere via dominant negative mechanisms with the wild-type clonally related cell population, consistent with the notion that
p53 protein in the cell because p53 functions as a homotetrameric neoplastic transformation of one or a number of different cells within
protein and all subunits must be wild-type for fully intact p53 function a tissue give rise to all daughter cells or subclones that are present in
in transcriptional regulation.7 For other tumor suppressor proteins, the tumor. This is further established by NGS deep sequencing in
such as the cyclin-dependent kinase inhibitory protein p27, reduction studying intratumor heterogeneity.10 Second, in tumors in which it
of protein levels to 50% of the levels present in normal cells might has been possible to analyze both cancer cells and associated precancer-
result in significant detrimental effects on the ability of the cell to ous cell populations, a subset of the somatic gene defects present in
appropriately regulate growth.8 the cancer are clonally represented in the precancerous cell population.
Genetic and Epigenetic Alterations in Cancer  •  CHAPTER 14 213

Table 14.2 Selected Tumor Suppressor Gene Mutations in Hereditary Cancer Syndromes and
Sporadic Cancers
Associated Inherited Cancer Cancers With Somatic
Gene Syndrome Mutations Presumed Function of Protein
RB1 Familial retinoblastoma Retinoblastoma, osteosarcoma, SCLC, Transcriptional regulator; E2F binding
breast, prostate, bladder, pancreas,
esophageal, others
TP53 Li-Fraumeni syndrome Approximately 50% of all cancers Transcription factor; regulates cell
(rare in some types, such as prostate cycle and apoptosis
carcinoma and neuroblastoma)
CDKN2A (p16INK4a protein) Familial melanoma, familial Approximately 25%–30% of many Cyclin-dependent kinase inhibitor (i.e.,
pancreatic carcinoma different cancer types (e.g., breast, CDK4 and CDK6)
lung, pancreatic, bladder)
CDKN2A (ARF protein) Familial melanoma (?) Approximately 15% of many Regulates MDM2 protein stability and
different cancer types hence p53 stability; alternative reading
frame of CDKN2A gene
APC FAP coli, Gardner syndrome, Colorectal carcinomas, desmoid Regulates levels of β-catenin protein in
Turcot syndrome tumors, hepatocellular carcinoma, the cytosol; binding to EB1 and
breast (rare) microtubules
WT1 WAGR, Denys-Drash syndrome Wilms tumor Transcription factor
NF1 Neurofibromatosis type 1 Melanoma, neuroblastoma p21ras-GTPase
NF2 Neurofibromatosis type 2 Schwannoma, meningioma, Juxtamembrane link to cytoskeleton at
ependymoma adherens junction
VHL von Hippel–Lindau syndrome Renal (clear cell type), Regulator of protein stability
hemangioblastoma
BRCA1 Inherited breast and ovarian Ovarian (~10%), rare in breast cancer DNA repair; complexes with Rad 51
cancer and BRCA2; transcriptional regulation
BRCA2 Inherited breast (both female and Rare mutations in pancreatic, DNA repair; complexes with Rad 51
male), pancreatic cancer others (?) and BRCA1
MEN1 Multiple endocrine neoplasia Parathyroid adenoma, pituitary Nuclear protein; unknown function
type 1 adenoma
Endocrine tumors of the pancreas
PTCH1 Gorlin syndrome, hereditary basal Basal cell skin carcinoma, Transmembrane receptor for Sonic
cell carcinoma syndrome medulloblastoma Hedgehog factor; negative regulator of
Smoothened protein
PTEN Cowden syndrome; sporadic cases Glioma, breast, prostate, follicular Phosphoinositide 3-phosphatase;
of juvenile polyposis syndrome thyroid carcinoma, head and neck protein tyrosine phosphatase
squamous carcinoma; many others
SMAD4 Familial juvenile polyposis Pancreatic (~50%), approximately Transcriptional factor in TGF-β
syndrome 10%–15% of colorectal cancers, rare signaling pathway
in others
BMPR1A Familial juvenile polyposis Not known Receptor for bone morphogenetic
syndrome protein
MSH2, MLH1 PMS1, PMS2, Hereditary nonpolyposis Colorectal, gastric, endometrial, DNA mismatch repair
MSH6 colorectal cancer ovarian
CDH1 Familial diffuse-type gastric Gastric (diffuse type), lobular breast E-cadherin cell-cell adhesion molecule
cancer carcinoma, rare in other types (e.g.,
ovarian)
STK11 (LKB1) Peutz-Jeghers syndrome Lung adenocarcinoma; rare pancreas Serine/threonine protein kinase
cancers; absent in most other
cancers
EXT1 Hereditary multiple exostoses Osteochondroma Glycosyltransferase; heparan sulfate
chain elongation
EXT2 Hereditary multiple exostoses Not known Glycosyltransferase; heparan sulfate
chain elongation
TSC1 Tuberous sclerosis Bladder carcinoma; head and neck Hamartin; binds tuberin (TSC2);
squamous cancer; hepatocellular regulates cell size by inhibiting TOR
carcinoma function and protein synthesis
TSC2 Tuberous sclerosis Head and neck squamous cancer Tuberin (see previous row regarding
TSC1)

FAP, Familial adenomatous polyposis; GTPase, guanosine triphosphatase; SCLC, small cell lung carcinoma; TGF, transforming growth factor; TOR, target of rapamycin; WAGR,
Wilms tumor, aniridia, genitourinary anomalies, and mental retardation.
214 Part I: Science and Clinical Oncology

Other somatic gene defects appear to be acquired during progression progression of one cancer type often differs from that in another
from the precancer subclones to the dominant subclones in the cancer. cancer type. As a result, a genetic or epigenetic change that is critical
These molecular findings in benign and malignant tumors are in tumor initiation in one cancer type might contribute to tumor
essentially consistent with a model initially proposed by Foulds11 and progression in another tumor type and vice versa. Second, defects
subsequently advanced by Nowell12 (Fig. 14.1). In brief, the clonal that arise at “early” stages of tumorigenesis might play a vital role not
evolution model predicts that cancers arise as the result of successive only in tumor initiation but also in the aggressive behavior of advanced
expansions of clonally related cell populations. The successive expansions stage cancers. Third, the model predicts that the acquisition of further
are driven by the gradual or punctuated acquisition of mutations and genetic heterogeneity will be a common and important factor in primary
gene expression changes that endow a particular cell and its progeny cancer lesions and metastases. It is becoming more evident that genetic
with a selective advantage over cells that do not harbor the gene heterogeneity plays a significant role in resistance to chemotherapy
defects. In essence, clonal selection is an evolutionary process that and targeted therapy and the emergence of aggressive cell populations
allows the outgrowth of precancerous and cancerous cells that carry in patients with advanced cancer.
mutations and gene expression changes that confer the most potent Recent comprehensive sequence-based analyses of cancer cell
proliferative and survival properties on the cancer cells. However, it populations from individual patients in which the tumor cells under
is important to note that the specific constellation of genetic and study were distinct from one another in location and/or time has
epigenetic changes that are present in precancerous and cancerous begun to reveal that quite dramatic intratumoral genetic heterogeneity
cells is context-dependent and certainly varies considerably from one may be a “rule” in cancer, rather than an exception.13 Certain, perhaps
cancer type to another and even most certainly varies to a significant key, initiating genetic lesions (e.g., truncal mutations) might be shared
degree among patients whose cancers display quite similar clinical among all neoplastic clones, but geographically distinct regions of
and histopathologic features. The clonal evolution model has biologic large primary tumors may have very distinct mutation profiles (e.g.,
and clinical ramifications, just a few of which will be mentioned here. branched mutations) from those in other portions of the primary
First, the clonal gene defects that are present in a cancer can be traced tumor, and the metastatic cell populations may have considerable
in precancerous lesions from the same organ site with a goal of mutational divergence from the nonmetastatic cells. Accordingly, it
attempting to clarify the preferred order in which gene defects arise seems that quite extensively branched evolutionary growth may be
in the natural history of a particular cancer type. The particular order an important feature in both primary tumors and metastatic lesions,
in which defects accumulate during the initiation and subsequent with multiple competing clonal populations evolving through divergent

Normal Competing neoplastic subclones Successful malignant


stem/progenitor subclone
cell population

Time (years to decades)

Mutations and epigenetic defects

Figure 14.1  •  Role of clonal selection in cancer development and progression. Clonal selection is essentially an evolutionary process that promotes outgrowth
of precancerous and cancerous cells that carry the mutations and gene expression changes that confer the most potent proliferative and survival properties on
the cancer cells in a given context. The schematic diagram indicates that the stepwise emergence of benign and malignant cells over time is critically influenced
by mutations and epigenetic defects. Neoplasms most likely arise from a stem cell or progenitor cell population that is capable of additional cell divisions and
acquisition of certain differentiated characteristics. After the accumulation of a particular constellation of mutations and epigenetic defects in oncogenes and
tumor suppressor genes, a successful malignant subclone will outgrow the various competing neoplastic subclones. Further genetic heterogeneity within a
malignant subclone (not depicted) is likely, and the genetic heterogeneity might give rise to new subclones that display increased invasive and metastatic
potential. (Modified from Kern SE. Progressive genetic abnormalities in human neoplasia. In: Mendelsohn J, Howley PM, Israel MA, Liotta LA, eds. The
Molecular Basis of Cancer. 2nd ed. Philadelphia: Saunders; 2001.)
Genetic and Epigenetic Alterations in Cancer  •  CHAPTER 14 215

and convergent mutational mechanisms.13 This more recent view of tissue of origin but also to gain the ability to disseminate into sur-
the potentially quite extensive intratumoral genetic heterogeneity in rounding tissues and organs, lymphatics, and bloodstream and ultimately
any given cancer and the contributions of intratumoral heterogeneity to grow as metastatic lesions in distant sites in the body.1 As indicated
to tumor progression contrasts with some earlier views. Before the in Fig. 14.2, among the signature traits that are likely to be expressed
more recent studies, it was suspected that the cell populations in many in the majority, if not all, of cancers are the following: (1) an increased
primary cancers might be more homogeneous, wherein somatic tendency to manifest a stem cell or progenitor-like phenotype; (2) an
mutations were accumulated in a more stepwise fashion as a result of enhanced response to growth-promoting signals; (3) a relative resistance
multiple sequential clonal sweeps of each variant cell population in to growth-inhibiting cues; (4) an increased mutation rate to allow for
the primary cancer, with metastases perhaps most often arising from the rapid generation of new variant daughter cells; (5) the ability to
the clonally dominant cell population in the primary tumor. attract and support a new blood supply (angiogenesis); (6) the capacity
It is important to note that clonal somatic mutations are often to minimize an immune response and/or evade destruction by immune
presumed to have a causal role in promoting further tumor outgrowth effector cells; (7) the capacity for essentially limitless cell division; (8)
or progression because somatic mutations can become clonal (i.e., a failure to respect tissue boundaries, allowing for invasion into adjacent
present in all neoplastic cells) by only a limited number of mechanisms. tissues and organs as well as blood vessels and lymphatics; and (9)
For instance, the genetic alteration itself could have been selected for the ability to grow in organ sites with microenvironments markedly
because it provided the neoplastic cell with a growth advantage, allowing different from the one where the cancer cells arose. The development
it to become the predominant cell type in the tumor (clonal expansion). of some traits is likely to be associated with certain stages of tumori-
Genes with critical roles in promoting clonal outgrowth in a given genesis (see Fig. 14.2), but acquisition of signature traits in cancers
cancer have been termed drivers.14 Genes that are mutant in a significant is more likely to show a preferred order than an invariant order.
fraction of cancers and for which other lines of evidence link them Furthermore, many of the signature traits of cancer cells represent
to the cancer process can more readily be classified as drivers. Alter- complex biologic capabilities (e.g., angiogenic activity, immune evasion
natively, a somatic mutation, when detected, might have arisen and resistance, and metastatic competence). Therefore, it is likely that
essentially coincident with another, perhaps undetected, alteration substantial changes in many signaling pathways are needed for the
that was the crucial change underlying clonal outgrowth. Somatic cancer cell to manifest these traits.
mutations of this latter type have been termed passengers.14 However, Some of the gene defects seen in cancer cells may contribute
studies have shown that some of these passengers, which serve as predominantly to a few or perhaps even only one of the signature
metabolic and housekeeping genes, can be targeted therapeutically, traits of cancer cells previously listed, but it seems likely that many
leading to tumor suppression.15,16 Therefore, on the basis of extensive of the gene defects and expression changes have been selected for in
data from large-scale sequencing analyses that reveal large numbers large part because they exert pleiotropic effects on the cancer cell
of distinct genes that are each mutated in only a minority of cancers phenotype. Furthermore, defects in certain genes and signaling pathways
of a given type,17–20 sorting out drivers and passengers might not be might be strongly selected for at a certain point in cancer development
entirely straightforward based solely on sequencing data and will likely and progression; perhaps in large part the alterations allow the precancer-
require a significant body of functional studies and data. Indeed, ous or cancerous cells to acquire certain critical phenotypic features.
increasingly even noncoding alterations have been shown to play Gene defects that might be nearly uniformly present in early-stage
important roles in causing and maintaining the cancer phenotypes lesions of one tumor type might preferentially arise in later-stage
by introducing new cancer-causing promoters and enhancers that lead tumors in another organ site. A good example will be mutations of
to altered gene expression (see the later discussion of noncoding the KRAS oncogene, which is commonly present in early-stage pan-
mutations). Given this discussion about the potential uncertainties creatic cancer but appears in late-stage cancer of other types. The data
associated with linking specific somatic mutations to cancer develop- suggest that cellular and tissue context has critical, albeit poorly
ment, it is even more challenging to assign causal significance to any understood, modifying effects on the specific genetic defects that give
given gene expression change in precancerous and cancerous cells. In rise to neoplastic transformation, clonal outgrowth, and tumor progres-
particular, the ambiguities arise when considering whether a gene sion. Despite the elaboration of the so-called hallmarks of cancers,
expression or epigenetic change merely reflects the relative difference which involve not just cancer cells but the whole environment and
between cancer and its matched normal tissue or is truly causally noncancerous cells such as immune, stromal, and blood vessel cells,
involved in the cancer process. Nonetheless, if a specific gene or to fully understand the underlying causal mechanisms of each trait
epigenetic alteration can be shown to promote tumorigenesis or and how to therapeutically target them individually or in combination
neoplastic transformation in a variety of in vitro and in vivo tumor remains very challenging.
models or if the same gene or chromosomal region is recurrently
altered by particular epigenetic changes in tumors, then it might be Alterations in Cancer Target Conserved Signaling
reasonable to infer that the particular defect might indeed have a Pathways and Networks
causal (driver) role in tumorigenesis.
As noted previously and summarized in Tables 14.1 and 14.2, the
CONTRIBUTION OF GENE DEFECTS TO THE protein products of proto-oncogenes and tumor suppressor genes have
SIGNATURE TRAITS OF CANCER CELLS been implicated in diverse cellular processes. In light of the potentially
enormous level of complexity suggested by the vast and diverse array
Cancer represents a highly heterogeneous collection of diseases. Each of gene defects in cancer, it is somewhat reassuring that some general
cancer type has distinct biologic and clinical features and a variable concepts have emerged with respect to the means by which genetic
prognosis. Even cancers that arise at a single organ site, such as the and epigenetic alterations likely contribute to cancer initiation and
ovary, kidney, or lung, may represent a hodgepodge of different diseases progression. A principal overarching theme is that the protein products
at the molecular level. Morphologic features often allow the particular of oncogenes and tumor suppressor genes function in highly conserved
cancer types to be distinguished to some degree from one another. signaling pathways and regulatory networks.
Yet even for patients whose cancers have essentially identical gross The network in which the retinoblastoma protein (pRb) tumor
and microscopic appearances and very similar clinical manifestations, suppressor protein (encoded by the RB1 gene) functions is one of the
there may be vast differences in outcome. In spite of this complexity, more intensively studied oncogene–tumor suppressor gene networks.
the development of all cancers, regardless of type, is likely to be critically The pRb protein regulates cell cycle progression, in large part via its
dependent on the acquisition of certain phenotypic features that allow ability to bind to E2F transcription factor proteins.21,22 In addition
the cancer cells not only to grow in an unchecked fashion in their to its role as a cell cycle regulator, the pRb protein has been implicated
216 Part I: Science and Clinical Oncology

Normal

Stem/progenitor cell phenotype Benign lesion

Enhanced response
to growth-promoting signals
Carcinoma in situ
Resistance to growth-inhibitory
and apoptosis-inducing cues

Increased mutation rate

Immune evasion/resistance Locally invasive carcinoma

Angiogenic activity

Capacity for limitless cell division

Invasive capacity

Metastatic competence
Distant
metastasis

Figure 14.2  •  Acquisition of signature traits in neoplastic cells during cancer progression. Depicted in the figure are representative stages in the development
of a cancer, perhaps a typical epithelial cancer, such as those that typically arise in the lung, colon, breast, or prostate. The schema suggests that most advanced
cancers arise via clonal selection from subclones present in earlier-stage benign and localized lesions (e.g., carcinoma in situ and locally invasive carcinoma).
Some of the properties of advanced cancer cells are depicted by the ability of the cells to enter the bloodstream and to seed and grow in distant organ sites,
such as the liver (bottom). Nine signature traits of cancer cells are listed. The relative time at which neoplastic cells acquire some of the traits is uncertain,
although it seems likely that some traits, such as the expression of a stem/progenitor cell phenotype or enhanced response to growth-promoting signals, may
be acquired earlier in cancer development. Other traits, such as invasive capacity and/or metastatic competence, may be acquired later. Many signature traits
of cancer cells are in fact complicated biologic capabilities (e.g., angiogenic activity, immune evasion and resistance, and metastatic competence) and likely
depend on defects in a number of different factors and signaling pathways.

in regulation of cellular differentiation, survival, and even angiogenesis pRb function and its ability to regulate expression of critical E2F
in certain settings.22 The binding of pRb to E2F proteins allows pRb target genes (see Fig. 14.3).
to silence expression of E2F-regulated or “target” genes, such as those Studies of other proto-oncogenes and tumor suppressor genes have
needed for the DNA synthetic (S) phase of the cell cycle. The ability also supported the existence of conserved regulatory networks in which
of the pRb protein to bind to E2F proteins and to function in multiple different tumor suppressor gene and proto-oncogene protein
transcriptional repression appears to be tightly linked to its phosphoryla- products function. Although the adenomatous polyposis coli (APC)
tion status, with the hyperphosphorylated forms of pRb being incapable tumor suppressor protein may have roles in regulating various processes
of binding to and regulating E2F proteins. The cyclin D1 protein in the cell,23 a key function of APC is to participate in a multiprotein
and its associated protein kinase, cyclin-dependent kinase 4 (CDK4), complex that regulates the levels of the β-catenin protein in the
negatively regulate pRb by phosphorylating it. The p16INK4a tumor cytoplasm and nucleus (see Fig. 14.3). Components of the multiprotein
suppressor protein is a critical inhibitor of the CDK4/cyclin D1 complex complex that regulates β-catenin include the APC protein, another
(Fig. 14.3) and can therefore prevent the inactivation of pRb. As is tumor suppressor protein known as AXIN1, and a kinase known as
noted in Table 14.2, a subset of sporadic cancers of various types has glycogen synthase kinase 3β (GSK3β). Inactivation of APC or AXIN1
inactivating mutations in the RB1 gene. In other cancers, pRb function function in cancer cells appears to lead to an inability to phosphorylate
appears to be critically compromised as a result of mutations in other β-catenin and hence target it for recognition and subsequent ubiquitina-
components of the network or pathway. For example, in many cancers tion by the βTrCP ubiquitin ligase and ultimately its destruction by
that lack RB1 mutations, inactivating mutations in the CDKN2A the proteasome.23,24 As a result, cancer cells with APC or AXIN1
gene have been noted. In other cancers, including some breast cancers, inactivation display increased levels of β-catenin in the cytoplasm and
gene amplification and overexpression of cyclin D1 is found. In yet nucleus and essentially constitutive complexing of β-catenin with
others, such as some glioblastomas and sarcomas, amplification and transcription factors of the T-cell factor (TCF) family, such as TCF4.
overexpression of the CDK4 gene have been seen. The net effect of When bound to TCF4, β-catenin can function as a transcriptional
mutations in the pRb pathway, whether in RB1 itself or in other coactivator, and in cancers with APC inactivation, such as colorectal
genes, such as CDKN2A, CCND1 (cyclin D1), or CDK4, is to inactivate carcinomas, TCF transcriptional activity is clearly deregulated. In a
Genetic and Epigenetic Alterations in Cancer  •  CHAPTER 14 217

pRb pathway APC/β-catenin pathway p53 pathway

p16 Wnt p19ARF

CYC D1 Cdk4 GSK3β MDM2


APC AXIN1

pRb p53
β-CAT TCF-4

E2F DP

Target genes Target genes Target genes


[e.g., Cyclin E, DHFR, [e.g., c-MYC, CYC D1, MMP-7, [e.g., p21CIP1, GADD45, BAX,
TS, DNA Pol α, RNR] survivin, CD44, EphB2/B3] p53AIP1, TSP1, MDM-2]

Figure 14.3  •  Recurrent gene defects in conserved signaling pathways in cancer. Three signaling pathways that are commonly affected by mutations in
various cancers are shown. Selected interactions between components of the pRb (left), APC/β-catenin (middle), and p53 pathways (right) are shown. Tumor
suppressor proteins are indicated with red symbols, oncogene products are indicated by green, and proteins that are not known to be affected by mutational
or epigenetic defects in human cancer are indicated in yellow. Inhibitory interactions between proteins are indicated by perpendicular lines, and activating
effects are indicated by arrows. Presumptive downstream genes whose expression is affected by the pathways are noted. APC, Adenomatous polyposis coli;
AXIN1, axis inhibition protein 1; β-CAT, β-catenin; Cdk4, cyclin-dependent kinase 4; CYC D1, cyclin D1; DHFR, dihydrofolate reductase; DNA Pol α, DNA
polymerase α; GADD45, growth arrest and DNA damage inducible gene 45; GSK3β, glycogen synthase 3β; MMP-7, matrix metalloproteinase 7; p21, p21
CDK-interacting protein 1; p53AIP1, p53-regulated apoptosis-inducing protein 1; RNR, ribonucleotide reductase; TCF-4, T-cell factor-4; TS, thymidylatesynthase;
TSP1, thrombospondin 1.

subset of the colorectal carcinomas that lack APC inactivation and including mutations in RB1, CDKN2A, CCND1 [cyclin D1], and
in a variety of other cancer types (see Table 14.1), activating (oncogenic) CDK4) are seen in the majority of a broad array of cancer types.26,27
mutations in the β-catenin protein have been found.24 These missense Unfortunately, at present, although no compelling mechanistic explana-
and in-frame deletion mutations affect key phosphorylation sites in tion exists for these observations, perhaps a general explanation can
the N-terminus of β-catenin, essentially rendering β-catenin resistant be offered. Specifically, the genetic pathways in which certain oncogenes
to regulation by the APC/AXIN/GSK3β complex. Hence, the mutant and tumor suppressor genes function are not simply linear pathways
β-catenin protein accumulates in the cell and deregulates TCF transcrip- as indicated schematically in Fig. 14.3 but more likely represent much
tion. Transcription of selected proto-oncogenes may be activated directly more complex and branched networks. The branches of the network
by the β-catenin/TCF complex, such as c-MYC.25 may even vary considerably, depending on cell type and developmental
Other tumor suppressor gene regulatory networks have been defined, context, although it seems reasonable to predict that the genes and
including the p53/MDM2/p19Arf pathway (see Fig. 14.3), the PTCH/ the protein products recurrently affected by mutation in human cancer
SMO/GLI pathway, and the MSH2/MLH1/PMS2 DNA mismatch represent particularly critical hubs in the pathways and networks.
recognition and repair pathway. Similar to the situation for the pRb,
APC/β-catenin, and p53 pathways, mutations in cancer cells not Epigenetic Mechanisms of Proto-oncogene
infrequently target the PTCH/SMO/GLI and MSH2/MLH1/PMS2 Activation and Tumor Suppressor Inactivation
pathways, either activating an oncogene within the pathway (e.g.,
SMO or GLI1 for the PTCH/SMO/GLI pathway) or inactivating The preceding discussion largely emphasized the role and significance
one of the key tumor suppressors (e.g., either MLH1 or MSH2 in of somatic and germline mutations in proto-oncogenes and tumor
the mismatch repair [MMR] pathway). suppressor genes in cancer. However, changes in the levels and/or
Although a large collection of genetic and biochemical data support patterns of gene expression, DNA methylation, and chromatin
the proposed protein functions and interactions depicted in Fig. 14.3, modifications have also been proven to contribute to tumorigenesis.
it seems likely that the situation in vivo is far more complex. For Arguably, the most compelling data for assigning a critical and likely
example, on the basis of the regulatory scheme outlined for the pRb causal role to changes in gene expression in the cancer process are for
pathway in Fig. 14.3, it might appear that the phenotypic consequences the genes that have already been well established in prior mutational
of RB1 or CDKN2A inactivation are functionally equivalent. However, and/or function studies to be tumor suppressor genes, particularly in
patients with germline mutations that inactivate RB1 are predisposed the cancer type under study. Hence, the emphasis here is placed on
to retinoblastomas and osteosarcomas, whereas persons with germline illustrating how epigenetic mechanisms have been assigned a causal
defects in CDKN2A are predisposed predominantly to melanoma and role in silencing tumor suppressor genes in cancer. The roles of
pancreatic cancer.26,27 Furthermore, whereas persons with germline superenhancer and other epigenetic mechanisms in proto-oncogene
mutations that affect RB1 or CDKN2A are predisposed to a rather activation, akin to those seen in cancers with high copy amplification
limited spectrum of cancers, somatic defects in the pRb pathway (e.g., of the respective proto-oncogene, are discussed later.
218 Part I: Science and Clinical Oncology

Mut Me
Mutation Hit #1 Epigenetic
silencing

LOH Epigenetic LOH Epigenetic


Hit #2 silencing Hit #2 silencing
Mut Mut Me Me

Me Me

Mutation Mutation Epigenetic silencing Biallelic epigenetic


+ + + silencing
LOH Epigenetic silencing LOH

Figure 14.4  •  Knudson’s two-hit hypothesis revised. In brief, Knudson’s hypothesis predicted that both alleles of a tumor suppressor gene would need
to be inactivated by germline and/or somatic mutations to elicit critical phenotypic alterations associated with cancer development. The revised version of
Knudson’s two-hit hypothesis considers the possibility that tumor suppressor gene inactivation can result from either genetic (mutation) or epigenetic silencing
events. The two functional alleles of a given tumor suppressor gene are indicated by the two purple boxes (top). The first inactivating event affecting one of
the two tumor suppressor gene alleles could be either a mutation, such as the localized defect indicated by the yellow box (left), or transcriptional silencing
associated with or caused by hypermethylation of CpG-rich sequences in the promoter/regulatory region (right). The inactivating event for the second tumor
suppressor gene allele (i.e., “Hit #2”) could be a nondisjunction event resulting in loss of the chromosome containing the wild-type tumor suppressor gene allele
(loss of heterozygosity [LOH]) or epigenetic silencing. (Modified from Jones PA, Laird PW. Cancer epigenetics comes of age. Nat Genet. 1999;21:163–167.)

As is summarized in Table 14.2, somatic inactivation of selected Mutations Affecting DNA Methylation Enzymes
tumor suppressor genes has been well established to result from
mutational (genetic) mechanisms in many cancers. However, a Colorectal cancers (CRCs) were among the cancer types studied initially
robust and growing body of data supports the view that epigenetic in great depth for consistent patterns of epigenetic alterations, and it
mechanisms somatically inactivate selected tumor suppressor genes was long known that whereas most CRCs display modest to moderate
in certain cancer types28,29 (Fig. 14.4). Although data are still emerg- global loss of 5-methylcytosine (5mC), certain subsets of CRC were
ing on the likely diverse transcriptional and chromatin remodeling identified that appear to preferentially inactivate many different genes
mechanisms responsible for epigenetic silencing of particular tumor by promoter hypermethylation—the so-called CpG island methylator
suppressor genes, many studies have demonstrated that increases in the phenotype (CIMP).30 The findings on CIMP-positive CRCs suggested
methylation of CpG-rich sequences (CpG islands) in the regulatory that unidentified alterations in the methylation/demethylation
regions (i.e., promoter/enhancer) of tumor suppressor genes are often machinery might play a role in the development of colorectal and
linked to loss of tumor suppressor gene expression. For instance, perhaps other cancer types.
whereas the VHL gene is inactivated by mutational mechanisms in Indeed, during the past few years, intriguing insights into factors
roughly 80% of renal carcinomas of clear cell type, in the majority and mechanisms regulating DNA methylation state in cancer have
of the clear cell renal carcinomas in which specific VHL mutations emerged. Nearly all DNA methylation in somatic mammalian cells
cannot be detected, loss of VHL gene expression appears to be tightly occurs at the cytosine residue of CpG dinucleotides and is catalyzed
linked to hypermethylation of the VHL promoter.28 For some other by DNA methyltransferases (DNMTs). Whereas DNMT mutations
tumor suppressor genes, including the CDKN2A, BRCA1, and MLH1 have been suggested to play a role in several different cancer types, it
genes, promoter hypermethylation has also been implicated as key is now clear that localized mutations in DNMT3A are present in
mechanism of inactivation.29 In fact, on the basis of studies of genes roughly 20% to 25% of patients with acute myeloid leukemia (AML).31
that display extensive CpG island methylation and decreased or absent Although the DNMT3A mutations are associated with poor prognosis
gene expression in cancer cells of one type or another, it has been in persons with AML, it remains unclear exactly if and how the
suggested that aberrant CpG methylation might play a very broad mutations affect DNA methylation in persons with AML.31 In addition
and important role in silencing tumor suppressor genes in the cancer to mutations in DNMT3A and potentially other DNMT genes, other
process.28,29 Nevertheless, it remains unknown what fraction of the gene defects linked to DNA methylation factors and methylation state
genes whose promoters show increased methylation in cancers actually have been uncovered. A family of Tet methylcytosine dioxygenase
function in vivo as tumor suppressor genes. Promoter hypermethyl- proteins (TET1, TET2, and TET3) was identified in the past few
ation and loss of gene expression coupled with additional data such years; these proteins modify DNA by hydroxylating 5mC to
as restoration of gene expression by treatment with demethylating 5-hydoxymethylcytosine (5hmC) and, as a consequence, result in loss
agents and/or other agents that affect chromatin functional state (e.g., of 5mC in cellular DNA. More significantly for cancer development,
histone deacetylase inhibitors) might be best to establish their tumor inactivation of the TET2 gene was found in upward of 30% of all
suppressive roles. myeloid malignancies, and the TET2-mutant myeloid neoplastic cells
Genetic and Epigenetic Alterations in Cancer  •  CHAPTER 14 219

have reduced levels of 5hmC and display a 5mC hypermethylator


phenotype, with resultant changes in chromatin state and gene expres- Table 14.3 Selected Chromatin Regulators and
sion patterns.32 Remarkably, further connections to altered DNA Modifiers Recurrently Mutated in
methylation in cancer have been revealed though studies of the linkages Cancer
between mutations in the isocitrate dehydrogenase genes IDH1 and
IDH2 and the hypermethylator phenotype in myeloid neoplasms and Protein Tumor Types With Mutations
some other tumors.32 Specifically, a critical cofactor for proper TET2 HISTONE ACETYLTRANSFERASES
function is alpha-ketoglutarate, and the IDH proteins normally generate CBP AML, ALL, DLBCL, TCC
alpha-ketoglutarate from isocitrate. The mutant IDH proteins found p300 AML, ALL, DLBCL, TCC, CRC, BrCA, PanCA
in several cancer types, including myeloid cancers and gliomas, generate
2-hydroxyglutarate instead, which renders the TET2 protein in affected MOZ AML, MDS
cells nonfunctional, with a resultant hypermethylator phenotype similar MORF AML, EndometCA
to that seen in TET2-mutant cancers.32,33 It is expected that further HISTONE ACETYLATION READERS
studies will clarify the key genetic and epigenetic mechanisms underlying
CIMP status in colorectal and other cancer types in which the TET BRD1 ALL
and IDH proteins appear to be functionally intact. PBRM1 RenalCA, BrCA
HISTONE METHYLTRANSFERASES
Mutations in Histones, Histone Modifiers, MLL1 AML, ALL, TCC
and Chromatin Remodelers MLL2 Medulloblastoma, RenalCA, DLBCL, FL
Although the progress in defining mutations that can underlie changes MLL3 Medulloblastoma, TCC, BrCA
in 5mC and 5hmC state and DNA methylation in cancer has been SETD2 RenalCA, BrCA
highly encouraging, truly dramatic progress has been achieved during EZH2 DLBCL, MDS, BrCA, ProstateCA, ALL
the past few years in defining a broad array of mutations that are
related to chromatin, including histones, histone modifiers, and HISTONE DEMETHYLASES
chromatin remodelers in different cancer types.34,35 These illustrate JARID1A AML
the importance of chromatin structures and modifications in the JARID1C RenalCA
pathogenesis of cancer. Chromatin is made up of repetitive fundamental
units called nucleosomes, and each nucleosome consists of 147 base UTX AML, TCC, RenalCA, EsophagealCA, MM
pairs of DNA wrapped around an octamer of histones. Accessibility HISTONE METHYLATION READERS
of chromatin to transcription apparatus is integral to gene expression, TRIM33 ThyroidCA
and alterations in structures of chromatin and their modifications will
play an important role in altered gene expression and tumorigenesis. ING1 Melanoma, BrCA
Notably, one of the variant histones, namely histone H3.3, which is ING4 HNSCC
incorporated during mitosis into actively transcribed regions such as MSH6 CRC
promoters, gene bodies, and telomeres, is found to be mutated in
CHROMATIN REMODELERS
upward of one-third of pediatric glioblastomas (Table 14.3).34,36 These
mutations act by inhibiting polycomb repressive complex 2 (PRC2), BRG1 LungCA, medulloblastoma, BrCA, ProstateCA,
a multiprotein complex responsible for the methylation of H3 at PanCA, HNSCC
lysine 27, therefore leading to global reduction in H3K27 trimethyl- BRM HNSCC
ation. These mutations were shown to be therapeutically targetable ARID1A OvCA, EndometCA, BrCA, RenalCA, CRC, GastricCA,
by using EZH2 inhibitors and BET bromodomain inhibitors.37,38 medulloblastoma, TCC
In addition to the DNA and histone mutations, histone modifiers, ARID1B BrCA
which are responsible for reading, writing, and erasing histone modifica-
BAF60A BrCA
tions (e.g., methylation, acetylation) that directly influence gene
expression, are frequently mutated in cancer. For example, EZH2, a ATRX Glioblastoma, PanNET
histone methyltransferase that catalyzes the repressive H3K27me3 DAXX Glioblastoma, PanNET
histone mark and a subunit of the polycomb repressive complex 2 HISTONES
(PRC2), is mutated in diffuse large B-cell lymphoma and follicular
lymphoma. The active mutation causes increased methyltransferase H3.1 Glioblastoma
activity leading to increased H3K27 trimethylation. Its opposite H3.3 Glioblastoma
counterpart, KDM6A or UTX, which is an H3K27 demethylase, is
frequently inactively mutated in different cancer types including ALL, Acute lymphoblastic leukemia; AML, acute myeloid leukemia; BrCA, breast
multiple myeloma and bladder cancer. Interesting to note, both the carcinoma; CRC, colorectal carcinoma; DLBCL, diffuse large B-cell lymphoma;
EndometCA, endometrial carcinoma; EsophagealCA, esophageal carcinoma; FL, follicular
activating mutations of EZH2 and the inactivating mutation of lymphoma; GastricCA, gastric carcinoma; HNSCC, head and neck squamous cell
KDM6A can be targeted therapeutically by EZH2 inhibitors.39,40 To carcinoma; LungCA, lung carcinoma; MDS, myelodysplastic syndrome; MM, multiple
date, many other histone modifiers have been found to be mutated myeloma; OvCA, ovarian carcinoma; PanCA, pancreatic carcinoma; PanNET, pancreatic
in different cancer types including KMT2A (MLL1), KMT2C (MLL3), neuroendocrine tumor; ProstateCA, prostate carcinoma; RenalCA, renal carcinoma;
SETD2, KDM5C (JARID1C), CREBBP, and EP300. TCC, transitional cell carcinoma of the bladder; ThyroidCA, thyroid carcinoma.
Modified from the findings and literature cited in Dawson MA, Kouzarides T.
Chromatin remodelers can cause sliding, ejection, or insertion Cancer epigenetics: from mechanism to therapy. Cell. 2012;150:12–27.
of nucleosomes along the chromatin. Of the four main classes of
chromatin remodelers, SWI/SNF complex is best characterized and
is collectively mutated in approximately 20% of all human cancers.41 are frequently found in clear cell renal cell carcinoma, and BAF250/
It has many subunits subdivided into enzymatic, core, and accessory ARID1A mutations are found in ovarian cell carcinoma and various
subunits. Mutations in these subunits can be organ specific; for gastrointestinal cancers. Exactly how these mutations contribute to
example, the core subunit SMARCB1/SNF5 is invariably mutated tumorigenesis has not been actively studied, but most likely they
in rhaboid tumors, accessory subunit BAF180/PBRM1 mutations affect or alter expression of cancer-related genes. Recent studies have
220 Part I: Science and Clinical Oncology

shown that, for example, SMARCB1, also known as SNF5, INI1, transcripts into proteins is highly regulated by a novel class of short
and BAF47 and a core subunit of the complex invariably inactivated noncoding RNAs, the so-called microRNAs (miRNAs). The mature
in pediatric rhabdoid tumors, has been shown to play an important forms of miRNAs are 20 to 24 nucleotides in length and are generated
role in the integrity of SWI/SNF complexes, and its loss affects the by successive cleavages by the Drosha and Dicer nucleases from longer
binding of enhancers, particularly those required for differentiation. precursor transcripts that contain characteristic hairpin formations.53
Furthermore, it maintains binding to superenhancers that are essential Recognition of target transcripts occurs principally by binding of the
for rhabdoid tumor survival.42 Animal model studies of ARID1A, the miRNAs in the 3′ untranslated regions (3′UTR). Depending on the
most frequent subunit of the complex, also show that it targets SWI/ degree of homology to their target sequence, miRNAs induce trans-
SNF complexes to enhancers, where they work closely with transcription lational repression or cleavage of mRNAs. Roughly 1000 human
factors, and its loss impairs enhancer-mediated gene regulation that miRNAs have been described, and each miRNA can target hundreds
drives colon cancer tumorigenesis.43 to perhaps even a thousand or more mRNAs.53 This ability allows for
substantial combinatorial complexity and functional redundancy,
Alterations in DNA cis-Regulatory Landscape making the identification of specific functions of miRNAs and their
in Cancer involvement in oncogenic or tumor suppressive networks difficult.
The fact that miRNAs act via base-pairing of the miRNA with the
Although recent large-scale sequencing efforts have focused more 3′UTR also implies that alterations of both miRNA sequence and
attention on the coding sequence of genes, combined whole-genome miRNA target sequence might have functional consequences.
sequencing, DNA chip sequencing, and RNA sequencing technologies Several lines of evidence make it likely that miRNAs do play a
have identified and characterized more and more functionally relevant role in the development of cancers. For instance, global inhibition of
alterations in noncoding DNA regions that underlie the transcriptional miRNA production seems to facilitate the acquisition of a neoplastic
programs involved in unannotated transcription start sites.44 Others phenotype in primary mouse cells, suggesting that the net effect of
have found losses or gains of enhancers in cancer by comparing dif- miRNAs collectively might be a tumor suppressive effect.54 The genomic
ferential H3K4me1Chip-seq levels between tumors and matched location of many miRNAs map close to common chromosomal
normal tissues, leading to identification of variant enhancer loci predic- breakpoints in cancer. In most cases, however, it is still unclear whether
tive of gene expression. More recently, dense clusters of enhancers, the miRNAs are actually involved in conferring the selective advantage
also known as superenhancers or stretch enhancers, have been found to that is gained by these translocations or whether miRNAs for other
play an important role not only in cell lineage and identity, but also reasons are localized in regions of high genomic fragility.55 Compre-
in cancer tumorigenesis.45 Cancer cells can acquire superenhancers hensive analyses of miRNA expression patterns in human cancers
that drive expression of key cancer-related genes through different have revealed that different cancer types have distinct miRNA expression
mechanisms that involve noncoding DNA cis-regulatory regions (Fig. patterns. In fact, in many instances, miRNA expression patterns might
14.5). One mechanism involves somatic gains of transcription factor be more precise in determining the tissue of origin than mRNA
binding sites, either through mutations or microinsertions that can expression profiles.56 Similar to the situation with protein-coding genes,
affect the strength of a promoter or induce a new superenhancer. For in most cases it is unclear which expression changes are causative and
the former, two single nucleotide mutations (C228T and C250T) in which are a result of the development of the tumors.57
the TERT promoter generate an identical 11 bp nucleotide stretch Several miRNAs seem to play a role as part of classic tumor sup-
(5′-CCCCTTCCGGG-3′) serving as a de novo binding site for the pressive or oncogenic signal transduction pathways. The miRNA17-92
ETS transcription factor, leading to increased TERT expression.46,47 locus has been described as a direct transcriptional target of the c-MYC
For the latter, a 12 bp intergenic insertion at the 3′UTR of the TAL1 and E2F oncogenes.58 This polycistron locus encodes for seven miRNAs
gene creates a new MYB binding site adjacent to the core members and has been shown to be genomically amplified and overexpressed
of the TAL1 complex including RUNX1, GATA3, and ETS1.48 Another in some human B-cell lymphomas and lung cancers. When overex-
mechanism involves enhancer hijacking, which basically, through pressed, it can cooperate with c-MYC to accelerate lymphoma
chromosomal deletion or translocation, brings the gene adjacent to development in a murine model system. In addition, miRNA372 and
an active superenhancer, as shown by the cases of GF1 genes in miRNA373 have been implicated as oncogenes in the development
medulloblastoma.49 In other cases, enhancer duplication leading to of germ-cell tumors at least in part by inactivating the p53 tumor
gene overexpression has been reported.50 For example, two distinct suppressor pathway.59 miRNA10a has been suggested to be a driving
focal amplifications of superenhancer 3′ to MYC have resulted in force behind the metastatic progression of breast cancer cell lines.60
MYC overexpression in lung adenocarcinoma and endometrial car- Members of the let-7 family of miRNAs have been proposed as
cinoma, respectively. Similarly, focal amplification at the enhancer, tumor suppressor genes in lung cancer, supposedly in part by their
rather than the coding regions, of KLF5 has also been described in ability to inhibit the translation of the KRAS and HMGA2 oncogenes.61
head and neck cancer.50 Finally, removal of regulatory boundaries The members of the miRNA34 family have been shown to be direct
marked by the binding sites of insulators (e.g., CTCG and cohesion) transcriptional targets of the p53 tumor suppressor gene and may play
can lead to enhancer spreading and enhanced gene expression.45,51 a significant role in mediating the downstream functional activity of
Genomic deletion of CTCF binding sites has been found to cause p53 in some settings.62
overexpression of TAL1 and LMO2. Equally important, DNA Long noncoding RNAs (lncRNAs) represent another important
hypermethylation of the CTCF/cohesin binding sites can disrupt its class of noncoding RNAs in cancer pathogenesis.63 By convention,
binding leading to enhancer spreading.52 These emerging findings of lncRNAs are defined as a group of RNAs greater than 200 nucleotides
noncoding variants and their roles in tumorigenesis have led to the in length that do not appear to encode a protein. Given the range of
realization that many of the disease risk single-nucleotide polymorphisms possible variations on this theme, many subclasses of lncRNAs have
(SNPs) from GWAS findings are indeed located in the noncoding been suggested, often based on their location and orientation relative
regions, and they may well contribute to tumorigenesis through the to other transcribed genes near their chromosome position. There is
aforementioned mechanisms. much circumstantial evidence, largely based on expression data, to
implicate many different lncRNAs in cancer development and progres-
Noncoding RNAs in Cancer—microRNAs and Long sion, and space does not permit a full review of these data.63 For the
Noncoding RNAs most part, relatively few functional data are available at this point
that definitively implicate particular lncRNAs in cancer development
It has been recognized during the past decade that the abundance of or progression. Given the current uncertainties about which lncRNAs
messenger RNA (mRNA) transcripts and the translation of mRNA have critical functional roles in cancer development, some general
Genetic and Epigenetic Alterations in Cancer  •  CHAPTER 14 221

C D
Figure 14.5  •  Various mechanisms by which somatic changes in the noncoding regions can lead to activation of proto-oncogenes. (A) Mutations in the
TERT promoter gene create a de novo binding site for ETS transcription factor. A 12-bp microinsertion in the 3′ untranslated region (UTR) of TAL1 creates
an MYB binding site and activates TAL1 transcriptions. (B) Chromosomal translocation places a superenhancer adjacent to GFI1B or its paralogue, GFI.
(C) Focal amplification of superenhancer by copy number gain induces KLF5 overexpression. (D) Deletion of insulator element at CTCF/cohesion binding
site causes the activation of TAL1 by its neighboring genes. (Modified from Yao X, Xing M, Ooi WF, Tan P, Teh BT. Epigenomic consequences of coding
and noncoding driver mutations. Trends Cancer. 2016;2:585–605.)

concepts are highlighted here instead. lncRNAs may function in a remain to be uncovered and substantiated, but it is possible that given
variety of ways to regulate the expression of various cellular genes, the many structures that RNA can adopt and the catalytic functions
such as by serving as a scaffold for chromatin regulatory factors that of certain RNAs, lncRNAs may have a quite broad array of biochemical
regulate genes near the locus of the lncRNA and/or genes at other functions in cancer.
chromosome locations. Other lncRNAs may contain various miRNA
binding sites and may functionally compete with other coding region Genetic and Epigenetic Alterations and
transcripts for binding to particular miRNAs, thus acting in some Genomic Integrity
settings to quite dramatically modulate the levels and ability of
endogenous miRNAs to regulate transcripts of certain proto-oncogenes A characteristic feature seen in most cancers is genetic instability,
and tumor suppressor genes. Other functions for lncRNAs in cancer including high rates of localized point mutations or small insertion/
222 Part I: Science and Clinical Oncology

deletion mutations in a subset of cancers and a chromosomal instability dramatic shattering of a chromosome or chromosomes and recombina-
in a large fraction of many different cancer types.64 The chromosome tion of the fragments—may occur in 2% to 3% of all cancers and
instability includes numeric abnormalities (aneuploidy), as well as that in certain tumors, such as osteosarcomas, it may occur in up to
simple and complex structural abnormalities. Considerable progress 25% of cases.69 Chromotripsis may represent another mutator
has been made in defining some of the factors and mechanisms that mechanism, because it would allow multiple genetic abnormalities to
contribute to genetic instability in human cancer. arise and be selected for concurrently rather than sequentially.
Roughly 2% to 4% of CRCs arise in persons with hereditary
nonpolyposis colorectal cancer (HNPCC), a familial cancer syndrome Role of Tissue and Context Differences in the
attributable to germline mutation in one allele of an MMR gene (see Contributions of Gene Defects to Cancer
Table 14.2; e.g., MSH2, MLH1, and MSH6).65 The CRCs arising in Cell Phenotype
persons with HNPCC arise in large part as a result of the somatic
inactivation of the remaining wild-type MMR allele in an affected The published literature on the potential contributions of gene defects
individual with HNPCC. The cancer cells manifest a markedly increased to the altered phenotype of cancer cells has offered some suggestions
frequency of point mutations and small insertion and deletion mutations about how to consider the role of the gene defects in cancer patho-
due to defective MMR function, reflected by the so-called high- genesis. For instance, terms such as gatekeeper and caretaker have been
frequency microsatellite instability (MSI-H) phenotype.65 In the other used to classify the contributions of genes to cancer development.70
cancer types arising in individuals and families with HNPCC, such “Gatekeeper” genes have been suggested to be genes that play particu-
as ovarian, endometrial, and gastric cancer, similar MSI-H phenotypes larly critical roles in regulating cell proliferation and inhibiting cancer
are seen. In the setting of HNPCC, there would appear to be more development in certain tissues, such as the APC gene in CRC; the
rapid tumor progression from an initiated clone to frank malignancy tumor suppressive function of the genes must be overcome for cancers
as a result of an MMR-defective mutator phenotype. In addition to to arise in a given tissue or organ site.70 “Caretakers” have been generally
the HNPCC CRCs, about 10% to 12% of apparently sporadic CRCs defined as genes that do not play direct roles in growth control but
manifest the MSI-H phenotype, and epigenetic silencing of MLH1 rather likely play important roles in a number of tissues in maintaining
by DNA hypermethylation of the promoter region and perhaps other the fidelity of the genome via their role in DNA damage recognition
chromatin modifications plays a critical role in the majority of these and repair processes. The MLH1 and MSH2 MMR genes have been
sporadic MSI-H CRC cases.65 proposed to be representative caretaker genes,70 and some researchers
Based on the results of recent comprehensive DNA sequence analyses have suggested that perhaps the BRCA1 and BRCA2 genes also represent
of human cancers, it seems that hundreds to thousands of localized caretaker genes.71
mutations can be detected in most primary cancers, yet the specific The use of terms such as gatekeeper and caretaker might have some
factors and mechanisms responsible for the mutator phenotypes in merit. However, as will be illustrated in the following discussion,
most of these cancers remain to be defined.20 Possible mechanisms given the apparently important role of “gatekeeper” gene defects in
include mutations and alterations of DNA polymerases, including cancers that arise in various organ sites but the quite variable timing
aberrant expression of alternative specialized DNA polymerases known of “gatekeeper” gene defects in the natural history of one cancer type
as translesional polymerases.66 versus another, the term gatekeeper might be more confusing than
Functional studies in cultured cancer cells and in mice provide illuminating. In the case of some presumed “caretaker” genes, the
quite a broad array of mechanisms that could contribute to numerous genes might not be playing the passive role in the cancer process that
chromosomal abnormalities and the associated chromosomal instability has been assigned to them. This view is based on three lines of argument:
(CIN) phenotype in cancer. Such chromosomal abnormalities include (1) the apparent tissue specificity of the tumors that arise in persons
mitotic checkpoint defects, chromatin modification or condensation who harbor germline mutations in the “caretaker” genes, such as in
defects, and chromosome segregation defects due to centrosome persons who are affected by HNPCC and germline MLH1 or MSH2
abnormalities and other possibilities, such as altered sister chromatid mutations72; (2) the likely variable time in cancer development at
cohesion.67 For the most part, although mutations in a few genes that which “caretaker” mutations may arise from one tumor to the next,
might contribute to chromosome instability have been reported, the with sporadic colon tumors not usually manifesting MMR gene
CIN phenotype in the vast majority of human cancers remains inactivation and MSI until the carcinoma stage, despite the fact that
unexplained. A recent article indicates that the X-chromosome gene adenomas in persons with HNPCC often show MSI-H73,74; and (3)
STAG2, a gene encoding a subunit of a sister chromatid separation the evidence that “caretaker” genes might actually play key roles in
complex, is mutated in significant subsets of melanomas, glioblastomas, regulating cell proliferation and promoting apoptosis in certain
and Ewing sarcoma, and that inactivation of STAG2 can promote contexts.56
aneuploidy in cultured cells.68 The findings highlight the possibility In general, persons who harbor a germline mutation in a specific
that further work on STAG2, STAG2-interacting proteins, and the tumor suppressor gene or proto-oncogene are predisposed to a very
other proteins that function in regulating proper segregation of limited spectrum of cancer types. This observation is puzzling for a
chromosomes at mitosis will uncover more insights into the molecular couple of reasons. The majority of genes that are affected by germline
basis of the CIN phenotype. mutations in specific inherited cancer syndromes are essentially
Structural chromosomal alterations are common in many cancer ubiquitously expressed in adult tissues. Furthermore, for a number
types and presumably reflect the consequences of DNA double-strand of the tumor suppressor genes, mutations are often found to inactivate
breaks and nonhomologous end joining to generate chromosomal the gene in a much broader collection of sporadic cancer types than
deletions, inversions, and translocations, with biologic selection acting the types that commonly arise in germline mutation carriers. Children
on the variant cells with the structural alterations. If the structural who carry a germline mutation in the RB1 gene have a very elevated
abnormality activates and/or inactivates a gene or genes that lead to risk of the development of retinoblastoma and a more modest risk of
more robust proliferation and survival properties in a given context, the development of osteosarcoma but no dramatic increase in the risk
cells carrying a given structural alteration will be positively selected of most common adult cancers. Yet somatic defects in RB1 have been
for during cancer development. The fact that certain structural altera- found and are believed to be critical in the development of many
tions are recurrent in cancer offers strong support for this concept. different cancers, such as small cell lung carcinomas, in which the
For the most part, particular structural chromosomal alterations in vast majority have pRb defects.27,75
cancer were thought to be relatively straightforward, resulting in Potential explanations exist for these puzzling observations. For
recombination of noncontiguous sequences, sometimes accompanied instance, whereas pRb might have an essential role in regulating reti-
by deletions. However, recent work indicates that chromotripsis—a noblast cell proliferation and/or differentiation, in other tissues, such
Genetic and Epigenetic Alterations in Cancer  •  CHAPTER 14 223

as lung or breast epithelial cells, pRb might have a redundant role in squamous cell carcinomas (HNSCCs) indicated that NOTCH1 harbors
growth control, perhaps because of the contribution of pRb-related various inactivating mutations, including nonsense and frameshift
proteins, such as p107 and p130.21 Under this scenario, RB1 inactivation mutations, in roughly 10% to 17% of primary HNSCCs.81 Based on
in most cell types might not promote neoplastic growth unless other the well-known function of NOTCH proteins to regulate cell fate
defects, such as those in pRb-related proteins, are also present. An choices during development and adult tissues, it is perhaps not surprising
alternative and perhaps equally likely possibility is that somatic inactiva- that NOTCH1 can function as an oncogene when activated in some
tion of pRb might trigger apoptosis in many cell types unless other contexts and as a tumor suppressor gene when inactivated in other
somatic gene defects have arisen previously and these other defects contexts.81
interfere with the cell’s ability to undergo apoptosis after disruption
of RB1 function. Evidence that pRb inactivation can act in a context- CLINICAL IMPLICATIONS
dependent fashion to promote apoptosis versus neoplastic transforma-
tion has been offered.76,77 The tissue specificity of cancers that are Identification of genetic and epigenetic defects contributing to human
seen in persons who carry germline mutations in inherited cancer cancer reveals significant clinical implications for the present and the
genes is not restricted to the case of pRb. Germline p53 mutations future, and some current directions and possibilities for the field are
predispose primarily to osteosarcoma, soft tissue sarcoma, leukemia, highlighted here. As previously noted, a very great and diverse array
brain tumors, and breast cancer in women, and CDKN2A germline of mutations in oncogenes and tumor suppressor genes and epigenetic
mutations predispose primarily to melanoma and pancreatic cancer.26 changes underlie different forms of cancer. However, because certain
In spite of the relatively limited spectrum of cancer types that are mutations and epigenetic changes either alone or collectively are
seen in people who carry germline p53 or CDKN2A mutations, the preferentially and sometimes even uniquely associated with certain
p53 and p16INK4a genes are very commonly altered in human cancer, cancer types, molecular analyses of cancer specimens and biopsies are
with each of the genes being inactivated in upward of 35% to 50% likely to be an important adjunctive approach in the cancer diagnostic
of many different sporadic cancer types. area. For many hematologic malignancies and a subset of solid tumors,
Some genes with prominent roles in the development of a variety molecular analyses already serve a vital role in the diagnostic arena,
of different cancer types are sometimes presumed to have essentially helping, for instance, to distinguish subsets of tumors that manifest
singular functions in the cancer process in spite of data that suggest similar or overlapping histologies or to aid in diagnosis when the
otherwise. As an example, the E-cadherin protein plays an important primary organ site of a metastatic cancer is uncertain. It seems likely
role in cell-cell adhesion via the ability of its extracellular domain to that cancer diagnostic approaches that incorporate molecular annotation
form adhesive interactions with E-cadherin molecules on opposing will continue to expand during the next 5 to 10 years. Moreover,
cell surfaces and the ability of the E-cadherin cytoplasmic domain to based on recent reports that cell-free tumor-derived DNA can be
link to the actin cortical cytoskeleton via interactions with catenin readily detected in the plasma of patients with cancer and that copy
proteins at the plasma membrane.78 Early functional studies have number changes along with specific nucleotide changes can be detected
suggested that restoration of E-cadherin in cancer cells that had with comprehensive sequencing approaches,82,83 it seems likely that
endogenous E-cadherin defects interfered with the invasive properties molecular interrogation of plasma and/or other body fluids may well
of cancer cells in selected in vitro assays.79 Perhaps in large part because be pursued for improved staging information and to assess for relapse
of these observations, the loss of E-cadherin expression in cancer has in patients with a solid tumor after surgery, radiation, and/or chemo-
often been assigned a role in promoting invasive behavior in advanced therapy. Indeed, a recent early study suggests that molecular interroga-
cancer cells. Although loss of E-cadherin function might indeed tion of circulating tumor-derived DNA in plasma may be useful for
contribute to invasive behavior in cancers arising in vivo, it is worth identifying drug-resistant cancer cells even before the initiation of
bearing in mind that defects in E-cadherin could in fact play a distinct certain therapies.84
role in altering cell growth very early in the neoplastic transformation The genes and the protein products that are recurrently altered by
process in some tumor types, such as the gastric carcinomas that arise mutations and/or epigenetic defects in human cancer more than likely
in patients who carry germline E-cadherin mutations.80 represent particularly critical hubs in the pathways and networks that
A couple of additional examples of context-dependent effects of regulate cell growth, differentiation, and programmed cell death. Hence
mutations in cancer are offered here, in hopes of highlighting some efforts to target the particular proteins and pathways with apparently
of the potential challenges in unambiguously defining certain genes central roles in the pathogenesis of certain cancer types would appear
as oncogene targets or tumor suppressor gene targets in cancer develop- to offer potentially broad impact. Successes with targeted therapy such
ment. The p53 gene represents one such example. As previously as EGFR inhibitors in the setting of EGFR-mutant non–small cell
indicated, the wild-type p53 protein is a transcriptional regulator that lung cancer85 and with ALK inhibitors in the setting of lung cancer
is mutated in roughly 50% of all cancers. A common theme for the harboring translocations activating ALK 86 offer evidence for the notion
majority of the mutations is loss of wild-type p53 function as a transcrip- of targeting critical driver oncogene defects in cancer, despite the fact
tion regulator, owing to the mutant p53 protein’s loss of sequence- that subsets of these patients may develop resistance.
specific DNA binding activity and/or misfolding of the protein.7 In spite of the generally optimistic view that is offered as a result
However, more than 75% of the p53 mutations in human cancer of our rapidly expanding understanding of the nature and contribution
lead to expression of a mutant p53 protein that may have oncogenic of gene defects in cancer pathogenesis, some significant challenges
activity independent of its ability to dominantly interfere with wild-type remain if novel and specific new anticancer therapies are to be achieved
p53.7 Although the gain-of-function properties of mutant p53 in in the near term. In particular, in a number of the cases with dramatic
cancer are clear, the mechanisms underlying the oncogenic activity initial responses to single agents targeting the specific driver oncogene
of mutant p53 are complex and likely vary depending on the particular lesions, drug resistance and cancer recurrence and progression are
mutant p53 protein and cellular context.7 A second example is the seen. In certain other cancer types carrying some of the same driver
NOTCH1 gene, which is rarely mutationally activated by translocations oncogene lesions, such as BRAF-mutant CRCs, the cancers show little
in T-cell acute lymphoblastic leukemia (T-ALL) and much more if any evidence for response to the targeted agents.87 Another issue is
commonly activated in T-ALL by localized mutations in a juxtamem- that for a fair number of the specific signaling pathways that are
brane domain that allows the mutant NOTCH1 protein to be activated commonly disrupted in cancer, it might prove difficult to define readily
in a ligand-independent manner or by mutations in a cytoplasmic tractable targets for therapeutic intervention. For instance, in the case
domain that lead to increased stability of the cleaved cytoplasmic of the p53 pathway, it is unclear how effectively p53 function can be
domain of NOTCH1.81 In contrast to the gain-of-function mutations restored for the proteins that carry missense substitutions that inactivate
in NOTCH1 in T-ALL, sequence-based studies in head and neck p53 tumor suppressor activity and perhaps also generate oncogenic
224 Part I: Science and Clinical Oncology

p53 gain-of-function mutants. In the case of cancers with mutations and future efforts to understand the relationships among gene defects,
leading to β-catenin deregulation, a potential goal might be to define altered cell signaling and physiology, and cancer phenotype have already
agents that specifically interfere with the nuclear function of β-catenin shaped and will continue to shape our views of how best to proceed
in transcriptional activation of β-catenin/TCF-regulated target genes. with novel therapeutic interventions. An optimistic view is that
Although this is not an unreasonable notion, given the rather limited combining various therapeutic approaches including targeted therapy,
successes to date in defining small molecules that specifically affect immunotherapy or cell therapy, and even conventional chemotherapy
transcription factor complexes, it could be challenging to target with new molecular diagnostics efforts that inform about various
β-catenin via conventional pharmacologic approaches. Similar concerns issues—for example, disease subtypes, occult disease spread beyond
could perhaps be raised regarding the merits of attempting to specifically the primary organ site, disease recurrence at earlier time points, and
target other nuclear proteins and transcription factors that are deregu- intratumor heterogeneity and resistance mechanisms—will yield major
lated in cancer cells. With increasing understanding of the role of improvements in patient survival and quality of life for a range of
chromatin biology and its roles in tumorigenesis, we may witness an cancer types within the next decade.
increasing number of chromatin-related or epigenetic drugs being
developed for treatment of cancer.
Although the success of approaches to target cancer cells more The complete reference list is available online at
selectively remains to be broadly established, it is obvious that ongoing ExpertConsult.com.

KEY REFERENCES
1. Hanahan D, Weinberg RA. Hallmarks of cancer: the 32. Cimmino L, Abdel-Wahab O, Levine RL, Aifantis 65. Boland CR, Goel A. Microsatellite instability
next generation. Cell. 2011;144:646–674. I. TET family proteins and their role in stem cell in colorectal cancer. Gastroenterology. 2010;138:
2. Chan TA, Wolchok JD, Snyder A. Genetic basis for differentiation and transformation. Cell Stem Cell. 2073–2087.e3.
clinical response to CTLA-4 blockade in melanoma. 2011;9:193–204. 66. Hoffmann JS, Cazaux C. Aberrant expression of
N Engl J Med. 2015;373:1984. 33. Turcan S, et al. IDH1 mutation is sufficient to alternative DNA polymerases: a source of mutator
3. Le DT, et al. PD-1 Blockade in tumors with establish the glioma hypermethylator phenotype. phenotype as well as replicative stress in cancer. Semin
mismatch-repair deficiency. N Engl J Med. 2015;372: Nature. 2012;483:479–483. Cancer Biol. 2010;20:312–319.
2509–2520. 35. Yao X, Xing M, Ooi WF, Tan P, Teh BT. Epigenomic 67. Janssen A, Medema RH. Genetic instability: tipping
4. Hemann MT, et al. Evasion of the p53 tumour surveil- consequences of coding and noncoding driver muta- the balance. Oncogene. 2013;32:4459–4470.
lance network by tumour-derived MYC mutants. tions. Trends in Cancer. 2016;2:585–605. 68. Solomon DA, et al. Mutational inactivation of
Nature. 2005;436:807–811. 36. Schwartzentruber J, et al. Driver mutations in histone STAG2 causes aneuploidy in human cancer. Science.
5. Renan MJ. How many mutations are required for H3.3 and chromatin remodelling genes in paediatric 2011;333:1039–1043.
tumorigenesis? Implications from human cancer data. glioblastoma. Nature. 2012;482:226–231. 69. Stephens PJ, et al. Massive genomic rearrangement
Mol Carcinog. 1993;7:139–146. 37. Mohammad F, et al. EZH2 is a potential therapeutic acquired in a single catastrophic event during cancer
6. Knudson AG. Mutation and cancer: statistical target for H3K27M-mutant pediatric gliomas. Nat development. Cell. 2011;144:27–40.
study of retinoblastoma. Proc Natl Acad Sci USA. Med. 2017;23:483–492. 70. Kinzler KW, Vogelstein B. Cancer-susceptibility genes.
1971;68:820–823. 38. Piunti A, et al. Therapeutic targeting of polycomb Gatekeepers and caretakers. Nature. 1997;386(761):
7. Muller PAJ, Vousden KH. p53 mutations in cancer. and BET bromodomain proteins in diffuse intrinsic 763.
Nat Cell Biol. 2013;15:2–8. pontine gliomas. Nat Med. 2017;23:493–500. 72. Lynch HT, de la Chapelle A. Genetic susceptibility to
9. Knudson AG. Two genetic hits (more or less) to 39. McCabe MT, et al. EZH2 inhibition as a therapeutic non-polyposis colorectal cancer. J Med Genet. 1999;36:
cancer. Nat Rev Cancer. 2001;1:157–162. strategy for lymphoma with EZH2-activating muta- 801–818.
11. Foulds L. The natural history of cancer. J Chronic tions. Nature. 2012;492:108–112. 77. Morgenbesser SD, Williams BO, Jacks T, DePinho
Dis. 1958;8:2–37. 40. Ler LD, et al. Loss of tumor suppressor KDM6A RA. p53-dependent apoptosis produced by Rb-
12. Nowell PC. The clonal evolution of tumor cell amplifies PRC2-regulated transcriptional repression in deficiency in the developing mouse lens. Nature.
populations. Science. 1976;194:23–28. bladder cancer and can be targeted through inhibition 1994;371:72–74.
13. Swanton C. Intratumor heterogeneity: evolution of EZH2. Sci Transl Med. 2017;9. 78. Gumbiner BM. Regulation of cadherin-mediated
through space and time. Cancer Res. 2012;72: 42. Wang X, et al. SMARCB1-mediated SWI/SNF adhesion in morphogenesis. Nat Rev Mol Cell Biol.
4875–4882. complex function is essential for enhancer regulation. 2005;6:622–634.
14. Haber DA, Settleman J. Cancer: drivers and pas- Nat Genet. 2017;49:289–295. 79. Vleminckx K, Vakaet L, Mareel M, Fiers W, van Roy
sengers. Nature. 2007;446:145–146. 43. Mathur R, et al. ARID1A loss impairs enhancer- F. Genetic manipulation of E-cadherin expression by
15. Muller FL, et al. Passenger deletions generate thera- mediated gene regulation and drives colon cancer epithelial tumor cells reveals an invasion suppressor
peutic vulnerabilities in cancer. Nature. 2012;488: in mice. Nat Genet. 2017;49:296–302. role. Cell. 1991;66:107–119.
337–342. 54. Kumar MS, Lu J, Mercer KL, Golub TR, Jacks 80. Guilford P, et al. E-cadherin germline mutations in
16. Nijhawan D, et al. Cancer vulnerabilities unveiled T. Impaired microRNA processing enhances cel- familial gastric cancer. Nature. 1998;392:402–405.
by genomic loss. Cell. 2012;150:842–854. lular transformation and tumorigenesis. Nat Genet. 81. Lobry C, Oh P, Aifantis I. Oncogenic and tumor
22. Sage J. The retinoblastoma tumor suppressor and 2007;39:673–677. suppressor functions of Notch in cancer: it’s NOTCH
stem cell biology. Genes Dev. 2012;26:1409–1420. 55. Calin GA, Croce CM. Chromosomal rearrangements what you think. J Exp Med. 2011;208:1931–1935.
23. Aoki K, Taketo MM. Adenomatous polyposis coli and microRNAs: a new cancer link with clinical 82. Forshew T, et al. Noninvasive identification and
(APC): a multi-functional tumor suppressor gene. implications. J Clin Invest. 2007;117:2059–2066. monitoring of cancer mutations by targeted deep
J Cell Sci. 2007;120:3327–3335. 58. He L, et al. A microRNA polycistron as a potential sequencing of plasma DNA. Sci Transl Med. 2012;4:
24. Polakis P. The many ways of Wnt in cancer. Curr human oncogene. Nature. 2005;435:828–833. 136ra68.
Opin Genet Dev. 2007;17:45–51. 59. Voorhoeve PM, et al. A genetic screen implicates 83. Leary RJ, et al. Detection of chromosomal alterations
25. Myant K, Sansom OJ. Wnt/Myc interactions in intes- miRNA-372 and miRNA-373 as oncogenes in tes- in the circulation of cancer patients with whole-
tinal cancer: partners in crime. Exp Cell Res. 2011;317: ticular germ cell tumors. Cell. 2006;124:1169–1181. genome sequencing. Sci Transl Med. 2012;4:162ra154.
2725–2731. 60. Ma L, Teruya-Feldstein J, Weinberg RA. Tumour 84. Diaz LA, et al. The molecular evolution of acquired
26. Fearon ER. Human cancer syndromes: clues to invasion and metastasis initiated by microRNA-10b resistance to targeted EGFR blockade in colorectal
the origin and nature of cancer. Science. 1997;278: in breast cancer. Nature. 2007;449:682–688. cancers. Nature. 2012;486:537–540.
1043–1050. 61. Johnson SM, et al. RAS is regulated by the let-7 87. Corcoran RB, et al. EGFR-mediated re-activation
28. Jones PA, Baylin SB. The epigenomics of cancer. microRNA family. Cell. 2005;120:635–647. of MAPK signaling contributes to insensitivity of
Cell. 2007;128:683–692. 62. He L, He X, Lowe SW, Hannon GJ. microRNAs BRAF mutant colorectal cancers to RAF inhibition
30. Issa JP. CpG island methylator phenotype in cancer. join the p53 network–another piece in the tumour- with vemurafenib. Cancer Discov. 2012;2:227–
Nat Rev Cancer. 2004;4:988–993. suppression puzzle. Nat Rev Cancer. 2007;7:819–822. 235.
Genetic and Epigenetic Alterations in Cancer  •  CHAPTER 14 224.e1
224.e1

REFERENCES
1. Hanahan D, Weinberg RA. Hallmarks of cancer: the 32. Cimmino L, Abdel-Wahab O, Levine RL, Aifantis 56. Lu J, et al. MicroRNA expression profiles classify
next generation. Cell. 2011;144:646–674. I. TET family proteins and their role in stem cell human cancers. Nature. 2005;435:834–838.
2. Chan TA, Wolchok JD, Snyder A. Genetic basis for differentiation and transformation. Cell Stem Cell. 57. Garzon R, Fabbri M, Cimmino A, Calin GA, Croce
clinical response to CTLA-4 blockade in melanoma. 2011;9:193–204. CM. MicroRNA expression and function in cancer.
N Engl J Med. 1984;373:2015. 33. Turcan S, et al. IDH1 mutation is sufficient to Trends Mol Med. 2006;12:580–587.
3. Le DT, et al. PD-1 blockade in tumors with mismatch- establish the glioma hypermethylator phenotype. 58. He L, et al. A microRNA polycistron as a potential
repair deficiency. N Engl J Med. 2015;372:2509–2520. Nature. 2012;483:479–483. human oncogene. Nature. 2005;435:828–833.
4. Hemann MT, et al. Evasion of the p53 tumour surveil- 34. Dawson MA, Kouzarides T. Cancer epigenetics: from 59. Voorhoeve PM, et al. A genetic screen implicates
lance network by tumour-derived MYC mutants. mechanism to therapy. Cell. 2012;150:12–27. miRNA-372 and miRNA-373 as oncogenes in tes-
Nature. 2005;436:807–811. 35. Yao X, Xing M, Ooi WF, Tan P, Teh BT. Epigenomic ticular germ cell tumors. Cell. 2006;124:1169–1181.
5. Renan MJ. How many mutations are required for consequences of coding and noncoding driver muta- 60. Ma L, Teruya-Feldstein J, Weinberg RA. Tumour
tumorigenesis? Implications from human cancer data. tions. Trends Cancer. 2016;2:585–605. invasion and metastasis initiated by microRNA-10b
Mol Carcinog. 1993;7:139–146. 36. Schwartzentruber J, et al. Driver mutations in histone in breast cancer. Nature. 2007;449:682–688.
6. Knudson AG. Mutation and cancer: statistical study H3.3 and chromatin remodelling genes in paediatric 61. Johnson SM, et al. RAS is regulated by the let-7
of retinoblastoma. Proc Natl Acad Sci USA. 1971;68: glioblastoma. Nature. 2012;482:226–231. microRNA family. Cell. 2005;120:635–647.
820–823. 37. Mohammad F, et al. EZH2 is a potential therapeutic 62. He L, He X, Lowe SW, Hannon GJ. microRNAs
7. Muller PAJ, Vousden KH. p53 mutations in cancer. target for H3K27M-mutant pediatric gliomas. Nat join the p53 network—another piece in the tumour-
Nat Cell Biol. 2013;15:2–8. Med. 2017;23:483–492. suppression puzzle. Nat Rev Cancer. 2007;7:819–822.
8. Payne SR, Kemp CJ. Tumor suppressor genetics. 38. Piunti A, et al. Therapeutic targeting of polycomb 63. Prensner JR, Chinnaiyan AM. The emergence of
Carcinogenesis. 2005;26:2031–2045. and BET bromodomain proteins in diffuse intrinsic lncRNAs in cancer biology. Cancer Discov. 2011;1:
9. Knudson AG. Two genetic hits (more or less) to pontine gliomas. Nat Med. 2017;23:493–500. 391–407.
cancer. Nat Rev Cancer. 2001;1:157–162. 39. McCabe MT, et al. EZH2 inhibition as a therapeutic 64. Lengauer C, Kinzler KW, Vogelstein B. Genetic insta-
10. Gerlinger M, et al. Intratumor heterogeneity and strategy for lymphoma with EZH2-activating muta- bilities in human cancers. Nature. 1998;396:643–649.
branched evolution revealed by multiregion sequenc- tions. Nature. 2012;492:108–112. 65. Boland CR, Goel A. Microsatellite instability in
ing. N Engl J Med. 2012;366:883–892. 40. Ler LD, et al. Loss of tumor suppressor KDM6A colorectal cancer. Gastroenterology. 2010;138:2073–
11. Foulds L. The natural history of cancer. J Chronic amplifies PRC2-regulated transcriptional repression in 2087.e3.
Dis. 1958;8:2–37. bladder cancer and can be targeted through inhibition 66. Hoffmann J-S, Cazaux C. Aberrant expression of
12. Nowell PC. The clonal evolution of tumor cell of EZH2. Sci Transl Med. 2017;9. alternative DNA polymerases: a source of mutator
populations. Science. 1976;194:23–28. 41. Kadoch C, et al. Proteomic and bioinformatic phenotype as well as replicative stress in cancer. Semin
13. Swanton C. Intratumor heterogeneity: evolution analysis of mammalian SWI/SNF complexes identifies Cancer Biol. 2010;20:312–319.
through space and time. Cancer Res. 2012;72: extensive roles in human malignancy. Nat Genet. 67. Janssen A, Medema RH. Genetic instability: tipping
4875–4882. 2013;45:592–601. the balance. Oncogene. 2013;32:4459–4470.
14. Haber DA, Settleman J. Cancer: drivers and pas- 42. Wang X, et al. SMARCB1-mediated SWI/SNF 68. Solomon DA, et al. Mutational inactivation of
sengers. Nature. 2007;446:145–146. complex function is essential for enhancer regulation. STAG2 causes aneuploidy in human cancer. Science.
15. Muller FL, et al. Passenger deletions generate thera- Nat Genet. 2017;49:289–295. 2011;333:1039–1043.
peutic vulnerabilities in cancer. Nature. 2012;488: 43. Mathur R, et al. ARID1A loss impairs enhancer- 69. Stephens PJ, et al. Massive genomic rearrangement
337–342. mediated gene regulation and drives colon cancer acquired in a single catastrophic event during cancer
16. Nijhawan D, et al. Cancer vulnerabilities unveiled in mice. Nat Genet. 2017;49:296–302. development. Cell. 2011;144:27–40.
by genomic loss. Cell. 2012;150:842–854. 44. Muratani M, et al. Nanoscale chromatin profiling 70. Kinzler KW, Vogelstein B. Cancer-susceptibility genes.
17. Wood LD, et al. The genomic landscapes of human of gastric adenocarcinoma reveals cancer-associated Gatekeepers and caretakers. Nature. 1997;386(761):
breast and colorectal cancers. Science. 2007;318: cryptic promoters and somatically acquired regulatory 763.
1108–1113. elements. Nat Commun. 2014;5:4361. 71. Levitt NC, Hickson ID. Caretaker tumour suppressor
18. Greenman C, et al. Patterns of somatic mutation in 45. Hnisz D, et al. Activation of proto-oncogenes by genes that defend genome integrity. Trends Mol Med.
human cancer genomes. Nature. 2007;446:153–158. disruption of chromosome neighborhoods. Science. 2002;8:179–186.
19. Thomas RK, et al. High-throughput oncogene 2016;351:1454–1458. 72. Lynch HT, de la Chapelle A. Genetic susceptibility to
mutation profiling in human cancer. Nat Genet. 46. Huang FW, et al. Highly recurrent TERT pro- non-polyposis colorectal cancer. J Med Genet. 1999;36:
2007;39:347–351. moter mutations in human melanoma. Science. 801–818.
20. Loeb LA. Human cancers express mutator phenotypes: 2013;339:957–959. 73. Aaltonen LA, et al. Replication errors in benign and
origin, consequences and targeting. Nat Rev Cancer. 47. Horn SS. TERT promoter mutations in familial malignant tumors from hereditary nonpolyposis
2011;11:450–457. and sporadic melanoma. Science. 2013;339:959– colorectal cancer patients. Cancer Res. 1994;54:
21. Henley SA, Dick FA. The retinoblastoma family 961. 1645–1648.
of proteins and their regulatory functions in the 48. Figueroa ME, et al. Leukemic IDH1 and IDH2 74. Young J, et al. Genomic instability occurs in colorectal
mammalian cell division cycle. Cell Div. 2012;7:10. mutations result in a hypermethylation phenotype, carcinomas but not in adenomas. Hum Mutat.
22. Sage J. The retinoblastoma tumor suppressor and disrupt TET2 function, and impair hematopoietic 1993;2:351–354.
stem cell biology. Genes Dev. 2012;26:1409–1420. differentiation. Cancer Cell. 2010;18:553–567. 75. Kaye FJ. RB and cyclin dependent kinase pathways:
23. Aoki K, Taketo MM. Adenomatous polyposis coli 49. Northcott PA, et al. Enhancer hijacking activates defining a distinction between RB and p16 loss in
(APC): a multi-functional tumor suppressor gene. GFI1 family oncogenes in medulloblastoma. Nature. lung cancer. Oncogene. 2002;21:6908–6914.
J Cell Sci. 2007;120:3327–3335. 2014;511:428–434. 76. Khidr L, Chen P-L. RB, the conductor that
24. Polakis P. The many ways of Wnt in cancer. Curr 50. Zhang X, et al. Identification of focally amplified orchestrates life, death and differentiation. Oncogene.
Opin Genet Dev. 2007;17:45–51. lineage-specific super-enhancers in human epithelial 2006;25:5210–5219.
25. Myant K, Sansom OJ. Wnt/Myc interactions in intes- cancers. Nat Genet. 2016;48:176–182. 77. Morgenbesser SD, Williams BO, Jacks T, DePinho
tinal cancer: partners in crime. Exp Cell Res. 2011;317: 51. Katainen R, et al. CTCF/cohesin-binding sites RA. p53-dependent apoptosis produced by Rb-
2725–2731. are frequently mutated in cancer. Nat Genet. deficiency in the developing mouse lens. Nature.
26. Fearon ER. Human cancer syndromes: clues to the 2015;47:818–821. 1994;371:72–74.
origin and nature of cancer. Science. 1997;278:1043– 52. Flavahan WA, et al. Insulator dysfunction and 78. Gumbiner BM. Regulation of cadherin-mediated adhe-
1050. oncogene activation in IDH mutant gliomas. Nature. sion in morphogenesis. Nat Rev Mol Cell Biol. 2005;6:
27. Sellers WR, Kaelin WG. Role of the retinoblastoma 2016;529:110–114. 622–634.
protein in the pathogenesis of human cancer. J Clin 53. Valencia-Sanchez MA, Liu J, Hannon GJ, Parker R. 79. Vleminckx K, Vakaet L, Mareel M, Fiers W, van Roy
Oncol. 1997;15:3301–3312. Control of translation and mRNA degradation by F. Genetic manipulation of E-cadherin expression by
28. Jones PA, Baylin SB. The epigenomics of cancer. miRNAs and siRNAs. Genes Dev. 2006;20:515–524. epithelial tumor cells reveals an invasion suppressor
Cell. 2007;128:683–692. 54. Kumar MS, Lu J, Mercer KL, Golub TR, Jacks role. Cell. 1991;66:107–119.
29. Esteller M. Epigenetics in cancer. N Engl J Med. T. Impaired microRNA processing enhances cel- 80. Guilford P, et al. E-cadherin germline mutations in
2008;358:1148–1159. lular transformation and tumorigenesis. Nat Genet. familial gastric cancer. Nature. 1998;392:402–405.
30. Issa J-P. CpG island methylator phenotype in cancer. 2007;39:673–677. 81. Lobry C, Oh P, Aifantis I. Oncogenic and tumor
Nat Rev Cancer. 2004;4:988–993. 55. Calin GA, Croce CM. Chromosomal rearrangements suppressor functions of Notch in cancer: it’s NOTCH
31. You JS, Jones PA. Cancer genetics and epigenetics: two and microRNAs: a new cancer link with clinical what you think. J Exp Med. 2011;208:1931–
sides of the same coin? Cancer Cell. 2012;22:9–20. implications. J Clin Invest. 2007;117:2059–2066. 1935.
224.e2 Part I: Science and Clinical Oncology

82. Forshew T, et al. Noninvasive identification and 84. Diaz LA, et  al. The molecular evolution of 86. Kwak EL, et al. Anaplastic lymphoma kinase inhibi-
monitoring of cancer mutations by targeted deep acquired resistance to targeted EGFR blockade tion in non-small-cell lung cancer. N Engl J Med.
sequencing of plasma DNA. Sci Transl Med. 2012;4: in colorectal cancers. Nature. 2012;486:537– 2010;363:1693–1703.
136ra68. 540. 87. Corcoran RB, et al. EGFR-mediated re-activation
83. Leary RJ, et al. Detection of chromosomal alterations 85. Mok TS, et al. Osimertinib or platinum-pemetrexed of MAPK signaling contributes to insensitivity of
in the circulation of cancer patients with whole- in EGFR T790M-positive lung cancer. N Engl J Med. BRAF mutant colorectal cancers to RAF inhibition
genome sequencing. Sci Transl Med. 2012;4:162ra154. 2017;376:629–640. with vemurafenib. Cancer Discov. 2012;2:227–235.
C. DIAGNOSIS OF CANCER
Pathology, Biomarkers, and 15 
Molecular Diagnostics
Wilbur A. Franklin, Dara L. Aisner, Kurtis D. Davies, Kristy Crooks,
Miriam D. Post, Bette K. Kleinschmidt-DeMasters, Edward Ashwood,
Paul A. Bunn, and Marileila Varella-Garcia

S UMMARY OF K EY P OI NT S
• Biomarker development has • Early detection using quite different can now be accomplished with
profoundly affected basic approaches has changed the natural accurate use of formalin-fixed
understanding of carcinogenesis and history of cervical, colorectal, and paraffin-embedded tissues.
expanded means of intervention in lung cancer. Progress depends • Molecular testing can provide
human cancers. Biomarkers have on improving our understanding guidance in prioritizing therapies for
been applied with variable success of the biology of site-specific patients most likely to benefit and
in three broad areas that correspond carcinogenesis and using can identify patients who have been
to phases of tumor development and intervention strategies informed by proven not to benefit from a targeted
progression: (1) early detection, (2) basic biology. therapy.
diagnosis, and (3) prediction of • Cancer diagnosis still relies on an • Molecular testing can also provide
clinical outcome (prognosis) and evaluation of tumor tissue histologic clinicians with a rationale to guide
response to targeted treatment. findings, a complex process specific patients toward specific
• For early detection, biomarkers go associated with interindividual clinical trials.
through five stages of development: variation; however, new approaches • As new molecular techniques have
(1) identification of promising to automated quantitative histologic been applied to cancer diagnosis
directions; (2) validation of a clinical evaluation and immunohistochemical and treatment, the necessity of
assay; (3) demonstration that the analyses are under active developing standardized processing
biomarker can detect disease before development. approaches for solid tumor
it is clinically relevant; (4) evaluation • New molecular diagnostic specimens has been appreciated
of the biomarker during prospective approaches that use assessment of and has helped to change tissue
screening; and (5) quantification of quantitative messenger RNA collection and specimen-handling
the effect of screening on reducing signatures, fluorescence in situ routines.
the burden of disease in a hybridization, quantitation of
population. microRNAs, and genetic sequencing

Since the time of Virchow more than 150 years ago, diagnosis and invasive cancers and the broad range of coverage needed to identify
treatment of cancer have depended on the appearances of tumor cells them have come into focus. This chapter reviews concepts of solid
under the microscope and the diverse and often subtle morphologic tumor carcinogenesis and biomarker development for early detection
differences that distinguish benign from malignant cells. Careful study and distinguishes early detection testing from biomarker applications
of cellular and later of molecular features of both hematologic and in diagnosis, prognosis, and treatment of established solid tumors.
solid tumors led to the idea that cancer results from a series of genetic Hematologic biomarkers are described elsewhere in this book.
and phenotypic changes in progenitor cells that occur over a period
of many years. In epithelial tumors, the final histologic changes that
define transformation of premalignant lesions into malignancy are EARLY DETECTION OF CANCER: THREE
the invasion of stromal tissues and the elicitation of a stromal response SUCCESSES IN REDUCING CANCER
including angiogenesis and metastasis to lymph nodes and distant MORTALITY DISCUSSED AS EARLY
sites.1 The clinical implications of this multistep process are best DETECTION MODELS
illustrated in the cervix, colon, and lung, where early intervention has
resulted in reduced mortality from cancer. Once invasive carcinoma Remarkable strides have been made in reducing morbidity and mortality
has occurred, testing becomes more urgent and intervention more through early intervention in several different kinds of solid tumors.
aggressive. Recently, the great diversity of genetic changes that drive The triggers for intervention have until now been largely based on

225
226 Part I: Science and Clinical Oncology

Delineation of molecular carcinogenesis in the cervix (Figs. 15.1


Table 15.1 Pepe’s Phases of Biomarker and 15.2) has provided alternative methods for detection, risk assess-
Development: Early Detection Research ment, and diagnosis. Nearly all cervical carcinomas and their precursor
Network lesions are caused by human papillomavirus (HPV).8 HPV is a DNA
tumor virus shown by Peyton Rous to produce squamous tumors in
Phase Category Descriptor rabbits in 1934.9–11 It was not until 1974 that zur Hausen recognized
1 Preclinical/Exploratory Promising directions identified the association between HPV and cervical cancer.12–14 Persistent infection
2 Clinical Assay and Clinical assay detects established with high-risk strains of HPV, most commonly types 16 and 18, is
Validation disease an essential step in cervical carcinogenesis (reviewed in zur Hausen15).
3 Retrospective/ Biomarker detects disease early, The virus encodes three proteins that stimulate cell proliferation and
Longitudinal before it becomes clinically evident survival, E5, E6, and E7 (reviewed in zur Hausen16). The viral proteins
and a screen-positive rule is defined E6 and E7 interfere with p53 and Rb, respectively17,18 resulting in
4 Prospective Screening Extent and characteristics of disease resistance to apoptosis and chromosomal instability. The HPV genome
detected by the test and the false may be integrated into the host cell genome, accounting for long-term
referral rate are identified persistence of the virus. Integration of E6 and E7 genes19,20 is crucial
5 Cancer Control Impact of screening on reducing
to progression of the infected cell to a malignant state, whereas other
the burden of disease on the
viral genes may be lost.
population quantified Detection of HPV DNA in clinical samples can be accomplished
by a number of methods, with the dominant techniques being based
on PCR or hybridization with signal amplification (reviewed in Leal
and Gulley21). These include the Hybrid Capture 2 method (HC2,
Qiagen), COBAS (Roche Molecular Diagnostics), and Aptima HPV
(HOLOGIC) assays, which have similar sensitivities and specificities22,23;
cytologic evaluation of exfoliated cells or advances in imaging technol- the latter two systems provide genotyping information in addition to
ogy. To date, few molecular markers have been sufficiently sensitive, simple detection information.
specific, or accessible to be useful in population-based studies for early Understanding the molecular oncogenesis of cervical squamous
detection or prevention. carcinoma has provided a standardized biomarker of risk. More than
There are several reasons for this. Sensitive biomarkers for risk 95% of HPV infections clear without further consequences. However,
assessment and early detection must be inexpensive, noninvasive, and persistent infection for months to years with HPV types 16 or 18
easily applied to be useful for population-based screening. However, carries a 40% risk of squamous intraepithelial lesions (SIL)24; untreated
such biomarkers are rarely sufficiently specific for diagnosis, prognosis, high-grade SILs have been estimated to result in invasive carcinoma
or response to treatment. A formal process to guide the development in approximately 30% of individuals over 5 to 10 years after diagno-
of early detection biomarkers has been defined by Pepe and colleagues.2 sis.24,25 Persistent HPV infection thus indicates high risk for eventual
In this construct, biomarkers must pass through five phases of develop- invasive carcinoma and represents an additional biomarker that could
ment that correspond to the strength of evidence that the biomarker be useful in the prevention of invasive carcinoma.
will be a useful population screening tool. These phases of development Testing for HPV DNA has been found to be more sensitive than
are analogous to the structure for therapeutic drug development3 that cytologic screening for detecting high-grade SIL26,27 but somewhat
has been used for many years. Pepe’s phases are listed in Table 15.1 less specific. In view of the number of infections that are self-limited,
and are the framework for the discussion of biomarker development relying on a single HPV test may result in greater numbers of negative
in individual organs later in this chapter. colposcopy examination results. To address this problem, cotesting
As will be seen, biomarker development for early detection has strategies have been evaluated whereby both cytologic evaluation and
been uneven across the various organs sites, and successes and challenges HPV testing are performed on initial examination.28 Negative results
faced for three exemplary organs are placed in context below. These have a high negative predictive value and the rescreening interval can
three organs were chosen because they exemplify organ sites where be extended from 1 to 5 years,29 reducing overall screening expense.
early detection trials have demonstrated reduction in disease morbidity Individuals with a positive viral DNA test result but negative cytologic
and mortality but where success of early detection biomarker develop- findings have been rescreened at 1 year and examined with colposcopy
ment has been variable. The organ site for which biomarker development if results are positive. Whether this or other strategies will be universally
is most advanced is the cervix, wherein a clear viral cause has been adopted for cervical cancer risk management remains to be deter-
identified and where the constituents of that virus can be used for mined.30,31 What is clear is that HPV testing provides a means for
risk assessment, early detection, diagnosis, triggering colposcopic improving sensitivity for detection of early cervical lesions, further
intervention, and vaccination, achieving Pepe’s stage 5 status of changing reducing mortality from invasive cervical carcinoma while potentially
clinical practice as described later. reducing screening expense.
Finally, HPV not only is a biomarker for risk assessment and
Cervix and a Validated Biomarker colposcopic intervention, but also is an immunogen that has been
successfully incorporated into bivalent (types 16 and 18; Cervarix),
Invasive cervical carcinoma is one of the few tumors that results from quadrivalent (types 6, 11, 16, and 18; Gardasil), and nonavalent (types
well-defined premalignant lesions that can be identified and effectively 16, 18, 31, 33, 45, 52, 58, 6; and 11; Gardasil 9) vaccines to prevent
treated, reducing morbidity and mortality from a tumor that is the infection and carcinogenesis (reviewed by Stanley32 and Villa33) in both
third most common in women worldwide.4 More than 60 years ago, female and male patients. In-depth discussion of HPV vaccines for
it was recognized that cellular abnormalities in cervical epithelium tumor prevention is beyond the scope of this chapter. However, the
detectable in exfoliated cells could predict the presence of carcinoma.5 many facets of HPV expression in cervical carcinoma and the emerging
By 1988, premalignant cervical cytology had been codified as the role of HPV in other tumors including vulvar and vaginal condylomata,
Bethesda system for reporting cervical and vaginal cytologic diagnoses.6 certain skin cancers, and head and neck carcinoma illustrate how a
Population-based light microscopic screening for atypical cells in cervical valuable biomarker that is integral to oncogenesis can be used to
smears with surgical removal of detected abnormal epithelium in Great improve the accuracy of estimating tumor risk and the specificity of
Britain is estimated to have reduced the prevalence of invasive squamous treatment. Unfortunately, such clearly defined and easily accessible
carcinoma by 80% after 1988.7 biomarkers are rare. Rather, the more usual case is described here for
Pathology, Biomarkers, and Molecular Diagnostics  •  CHAPTER 15 227

Cellular Carcinogenesis, Cervix

H&E

p16

Normal
cervix
#1
Low grade
(CIN I)
#2
High grade
(CIN III)
#3
Invasive
carcinoma
#4

Vaccination Colposcopy Resection


Severity of Histological Lesion

Figure 15.1  •  Three potentially premalignant lesions are depicted. 1, Normal cervical mucosa at the squamocolumnar junction (not shown) is composed
of a basilar zone from which squamous cells progressively mature and ultimately give way to flattened cells on the mucosal surface. The p16 immunostain in
the lower frame is negative, reflecting the absence of expression of p16 protein by normal cells. 2, Low-grade squamous dysplasia may occur as a consequence
of HPV infection. In this lesion, the basilar zone is expanded through the lower third of the mucosa, and cells have become more disorderly with irregular
spacing between cells, variable nuclear size and density, and clearing of cytoplasm in some of the cells in the upper mucosa (koilocytosis); p16 immunostains
show strong expression in the lower third of the mucosa. 3, Progression to high-grade squamous dysplasia is indicated by the near complete absence of cellular
maturation toward the surface. Cells contain large, misshapen, and irregularly distributed nuclei. In immunostained sections, p16 is strongly expressed through
the full thickness of the epithelium. 4, Finally, invasive carcinoma occurs when precursor cells acquire the capacity to invade the underlying cervical stroma
as shown in this photomicrograph. Invasive cervical squamous cell carcinoma strongly expresses p16 as shown in the lower frame. Vaccination may prevent
the earliest changes in the progression to invasive carcinoma. Low-grade squamous dysplasia (CIN I) usually regresses and does not require ablation. High-grade
in situ dysplastic changes can be effectively treated by excision of dysplastic epithelium, but if invasive carcinoma occurs, more aggressive surgery and/or radiation
therapy may be required. CIN, Cervical intraepithelial neoplasia.

Molecular Carcinogenesis, Cervix


Mucosal
exposure to HPV
Normal E6 integration, p53 inactivation
cervix E7 integration, pRb inactivation
p16 activation, overexpression
Type 16, 18
HPV infection
Chromosomal instability
Viral persistence other unknown factors
Dysplasia (LSIL)
Dysplasia (HSIL)

Viral persistence
Invasive carcinoma
Emergence from
latency
Clearing of Clearing of
virus virus and dysplasia

Severity of Histological Lesion

Figure 15.2  •  This figure depicts molecular changes that follow infection with high-risk subtypes of human papillomavirus (HPV). In most cases virus is
cleared, but persistent infection may lead to dysplasia. Virus may also be cleared from dysplastic epithelium, but in a subset of cases persistent infection
ultimately leads to carcinoma. HSIL, High-grade squamous intraepithelial lesions; LSIL, low-grade squamous intraepithelial lesions.
228 Part I: Science and Clinical Oncology

colon and lung, wherein potential biomarkers are defined but have not 15.3 and 15.4) and provided a rationale for colonoscopic surveillance
found a role in early detection, diagnosis, prevention, or treatment. and polypectomy. In the mid-20th century an association between
between benign polyps and invaive carcinoma was suspected, but at
Colon and Multistep Carcinogenesis With that time there was no direct evidence that polyps undergo transition
Hereditary Components to CRC.38 Suspicions were finally confirmed in the 1980s, when the
clonal nature of both sporadic polyps and CRC was documented.39
Like cervical carcinoma, colonic adenocarcinoma can be effectively Activating mutations in the KRAS oncogene and inactivating mutations
prevented by extirpation of premalignant epithelium, which in colon in several tumor suppressor genes (TSGs) were described.40 The same
is represented by polyps. Several trials have indicated that removal of mutations were demonstrated in both polyps and carcinoma, and the
polyps through colonoscopy34–36 reduces the frequency of colorectal number of mutations was found to increase with histologic grade of
carcinoma (CRC), and consensus opinion is that endoscopic polyp- the adenoma. This suggested a progression to invasive carcinoma
ectomy has a significant impact on the incidence of CRC. Data have through the accumulation of genetic and epigenetic changes. These
also suggested that colonoscopic screening reduces CRC mortality by changes provide a proliferative and survival advantage to mutant cells,
more than 50%36 and that the less extensive sigmoidoscopy reduces which continue to proliferate and mutate until a malignant phenotype
mortality from distal CRC by a nearly equal percentage (50%).37 The is reached. The combined data from these studies led to the proposal
latter sigmoidoscopic study was a large randomized population-based of a multistep model of colon carcinogenesis,41 which continues as
screening trial. Colonoscopic screening for colon polyps beginning at the standard model.42
age 50 has become a standard of care. Further insight into colon carcinogenesis has been provided by
Molecular biomarkers to date have not played a direct role in studies of hereditary forms of colon cancer and the molecular changes
reduction of morbidity and mortality in CRC. However, molecular that underlie them. A detailed discussion of hereditary colon cancer
studies have facilitated understanding of colon carcinogenesis (Figs. is provided elsewhere in this book and is beyond the scope of this

Cellular Carcinogenesis, Colon

Normal mucosa Low-grade sporadic polyp


#1 #2

Adenomatous polyp High-grade


(FAP, AFAP, MUTYH) sporadic polyp
#5 #3

Serrated adenoma
#6

Invasive carcinoma
#4
Juvenile polyp
(juvenile polyposis)
#7

Peutz-Jeghers polyp
(Peutz-Jeghers syndrome)
#8

Age/years Colonoscopy/excision Resection

Figure 15.3  •  This figure depicts progression from 1, normal mucosa through 2, low-grade and 3, high-grade polyps to 4, invasive carcinoma, the last
represented by poorly formed clusters of cells pushing aside the normal intestinal crypts visible on the right of the frame. Sporadic polyps shown in the upper
row of hematoxylin-eosin (H&E)–stained sections are typically pedunculated and composed of proliferating cells extending to the mucosa surface. As cellular
atypia increases, there is an overlapping of cells along the irregular mucosal surface of the high-grade polyp. 5, Polyps of FAP, AFAP, and MUTYH are mor-
phologically similar to sporadic polyps but occur at an earlier age. In 6, Lynch syndrome, polyps are flat and individual crypts are preserved without the convolutions
present in sporadic polyps. 7, Juvenile polyps are characterized by the presence of mucous cysts (visible on the left side of the frame) and an edematous and
inflamed stroma. The surface of the juvenile polyp may be denuded of epithelium, and there may granulation tissue in the mucosa. Finally, 8, Peutz-Jeghers
polyps have a complex lobulated appearance and contain smooth muscle bundles in the stalk. Removal of sporadic polyps has been found to reduce the frequency
of progression to invasive carcinoma. The presence of invasive carcinoma may require vigorous intervention (discussed elsewhere in this book).
Pathology, Biomarkers, and Molecular Diagnostics  •  CHAPTER 15 229

Molecular Carcinogenesis, Colon


MGMT, APC, WNT P16, KRAS, BRAF P53, DCC
Mutation/ Mutation/ Mutation/
methylation/ methylation/ methylation/
rearrangement rearrangement rearrangement

Wild type Low-grade High-grade


polyps polyps

MMR mutation MSI Lynch syndrome


CRC
APC mutation FAP
MUTYH mutation MAP
STK11 mutation PJS “Second hit”:
SMAD4 mutation JPS, HHT • Methylation
• Deletion
BMPR1 mutation JPS
• Mutation, etc.
Other/unknown ??

Age/years

Figure 15.4  •  Accumulation of molecular changes that precede colorectal carcinoma (CRC). Sporadic tumors are initiated by a serial accumulation of
somatic mutations that may eventuate in CRC. The initiating event in these polyposes is the mutation present at birth. Second hits may include a variety
molecular changes including tumor suppressor gene promoter methylation, mutations, and copy number changes. Mismatch repair (MMR) and includes
hMLH1, hMLH2, hMSH2, hPMS2, and EpCAMP genes. FAP, Familial adenomatous polyposis; HHT, hereditary hemorrhagic telangiectasia; JPS, juvenile
polyposis syndrome; MAP, MUTYH-associated polyposis; MSI, microsatellite instability; PJS, Peutz-Jeghers syndrome.

Table 15.2  Genetics of Colonic Polyposis


Syndrome (Abbreviation,
Synonym) Gene(s) Molecular Phenotype Noncolonic Organs Affected
Sporadic adenomatous polyp Unknown Chromosomal instability None known
(aneuploidy)
Familial adenomatous polyposis (FAP) APC Chromosomal instability Duodenum, stomach, pancreas, thyroid, liver,
(aneuploidy) central nervous system
Attenuated familial adenomatous APC Chromosomal instability Duodenum, thyroid, liver, central nervous
polyposis (AFAP) (aneuploidy) system
Hereditary nonpolyposis colon cancer hMLH1, hMSH2, hMSH6, MSI Endometrium, stomach, ovary, biliary and
(HNPCC, Lynch syndrome) hPMS2 urinary tracts, small bowel, central nervous
system
MUTYH-linked adenomatous MUTYH Chromosomal instability Duodenum
polyposis (MAP) (aneuploidy)
Peutz-Jeghers syndrome (PJS) STK1 (70%) Unknown Breast, pancreas, stomach, ovary, lung, small
bowel, uterus, testis
Juvenile polyposis syndrome SMAD4, BMPR1A Unknown Blood vessels (hereditary hemorrhagic
telangiectasia)
Hyperplasic polyposis SMAD4, BMPR1A (40%), PTEN Unknown Stomach, pancreas, small bowel

chapter. However, a brief description of the major forms of hereditary individuals with the formation of invasive carcinomas at an early age.
tumors will provide insight into colon carcinogenesis and perspective Polyps may also sometimes be found in the upper gastrointestinal
on the strengths and limitations of the role of genetic data in early tract. As long ago as 1882, siblings were described with numerous
detection of this prevalent tumor type. and recurring polyps,44 and by the mid-20th century FAP had been
Several forms of hereditary colon cancer are recognized (reviewed established as a hereditary autosomal dominant disorder.45 A gene
by Jasperson and colleagues43) and are listed in Table 15.2. mutation associated with FAP was identified on chromosome 5q by
linkage analysis46,47 and positional cloning studies.48–51 The gene,
Familial Adenomatous Polyposis adenomatous polyposis coli (APC), is now known to encode a tumor
The first hereditary colon carcinoma to be described, familial adeno- suppressor that modulates Wnt signaling through binding and stabiliza-
matous polyposis (FAP), is characterized in its classic form by the tion of a complex of proteins that includes GSK3b, β-catenin, and
occurrence of hundreds to thousands of polyps in the colon of affected axin family members. Mutations in APC truncate the protein at its
230 Part I: Science and Clinical Oncology

carboxyl terminus and disrupt the protein signaling complex. This that is recognized by MUTYH, which removes the mismatched A
results in deregulation of cell cycle control, cell migration, differentia- and restores the correct base, cytosine (C). The damaged G (8-oxoG)
tion, and apoptosis (reviewed by Goss and Groden52). In addition to is then replaced with an intact G. Mutation of MUTYH at specific
being responsible for FAP, APC mutation is present in approximately hot spots hinders recognition of the 8-oxoG:A mispairing, resulting
60% of sporadic colon adenocarcinomas53,54 and an approximately in G:C to A:T transversion at sites of oxidative damage. The mutational
equal number of sporadic polyps,53,55 suggesting that APC mutation hot spots are found in exon 7, resulting in amino acid substitution
is an early event in sporadic carcinogenesis as well as in hereditary Y179C, and in exon 13, resulting in substitution G496D. Germline
disease. An attenuated form of FAP with smaller numbers of tumors mutation at both hot spots (biallelic mutation) occurs in 0.3% of
(<100) has been described.54,55 This condition is referred to as attenuated patients with CRC. Biallelic mutation confers 28-fold relative risk for
familial adenomatous polyposis (AFAP)56 and may be driven by the CRC compared with controls. Monoallelic mutation is more common,
mutations in APC in the 3′ and 5′ ends rather the central portion of with a frequency of approximately 2% in CRC, but also occurs at
the gene that is affected in classic FAP.57 approximately the same frequency in control populations. Most studies
suggest that monoallelic mutation confers little or no increased risk
Hereditary Nonpolyposis Colorectal Cancer of CRC.77,78 Among control patients without polyposis, biallelic
The most common form of hereditary colon cancer is hereditary mutation is rare.78
nonpolyposis colorectal cancer (HNPCC). HNPCC is a familial
syndrome that differs from FAP by involving smaller numbers of Hamartomatous Polyposes
polyps in comparison to FAP. The earliest description of this disorder The fourth hereditary form of colon cancer are the hamartomatous
can again be traced back to the early 20th century,58 but the hereditary polyposes, a category that includes Peutz-Jeghers syndrome (PJS) and
character and syndromic features of the disease were firmly established juvenile polyposis syndrome. First described in the 1890s,80,81 PJS was
by the diligent kindred studies of Lynch and colleagues,59 after whom named for the work of Peutz82 and Jeghers83,84 in 1954 by Bruwer.85
the syndrome is named (Lynch syndrome). In brief, the syndrome PJS is an autosomal dominant condition characterized by multiple
consists of CRC occurring in close relatives, often younger than age histologically distinct polyps (see Fig. 15.3) occurring in the large and
50,60,61 without adenomatous polyposis. HNPCC is also associated small bowel in association with melanin pigmentation of the lips,
with an increased frequency of tumors in the endometrium, stomach, mouth, nose, anus, and extremities.86 The condition is associated with
ovary, hepatobiliary tract, upper urinary tract, small bowel, and central a high risk for cancer in not only the large and small intestine but
nervous system (CNS).43 In contrast to FAP polyps, which are frequently also in the esophagus, stomach, pancreas, breast, uterus, ovary, and
found on the left side of the colon, individuals with HNPCC frequently lung.87–89 Gene mapping has identified a locus on chromosome
have flat lesions on the right side of the colon with a distinct histologic 19.3p1390,91 that encodes a serine/threonine kinase, STK11 (LKB1),
appearance referred to as serrated adenoma (see Fig. 15.3). a gene that is mutated in more than 90% of affected individuals.92–96
The molecular genetic underpinnings of HNPCC were deciphered A variety of mutations including deletions, truncations, and missense
beginning with the linkage to microsatellite markers on chromosome point mutations may occur over nearly the full coding region of the
2p.62 The genetic basis of the disease was established when it was gene.88 STK11 protein is multifunctional, playing a role in cell cycle
found that patients with the syndrome harbored mutations in the arrest, wnt signaling, and the TSC pathway, with effects on mammalian
mismatch repair (MMR) genes hMLH1,63,64 hMSH2,65,66 hMSH6,67 target of rapamycin (mTOR) signaling.86 Mutation, often associated
and hPHS2,68,69 with mutations in hMLH1 and hMSH2 accounting with loss of heterozygosity (LOH),97,98 results in loss of function, and
for over 90% of HNPCC cases. The germline mutations are hetero- the gene is thus considered to be a TSG. The overall frequency of
zygous, and colon carcinogenesis results from silencing of the nonmutant PJS is estimated at 0.5 to 2.0 per 100,000 live births.99
allele by methylation70,71 or by acquired allelic loss.
The phenotypic consequence of gene silencing is loss of MMR Juvenile Polyposis
protein and the resulting inability to repair DNA replication errors, Juvenile polyps are characteristically solitary peculated lesions that
ultimately leading to accumulation of mutations contributing to contain tortuous, often cystically dilated crypts, stromal granulation
carcinogenesis. At a clinical level, protein loss can be demonstrated tissue, and inflammation and denudation of the surface epithelium.100
by immunochemistry and optimally requires four separate stains. The These lesions frequently occur in children, and there is no evidence
inability of affected cells to repair DNA replication errors results that solitary juvenile polyps pose an increased risk for carcinoma.100–102
in insertions or deletions in short tandem repeats (microsatellites) However, multiple juvenile polyps may occur in young individuals
that exist throughout the genome and is referred to as microsatel- and their family members,103–105 and the presence of more than five
lite instability (MSI).72,73 Currently, MSI is documented by testing juvenile polyps or a single juvenile polyp in an individual with a
of a standard set of five microsatellite markers recommended by a family member with juvenile polyposis now defines the condition
consensus panel.74 Results are scored on the basis of the number juvenile polyposis. This form of polyposis carries a lifetime risk of
of unstable loci and are classified as microsatellite instability–high colon carcinoma of 39% and a relative risk of 34.106 Fifty percent to
(MSI-H), microsatellite instability–low (MSI-L), or microsatellite 60% of affected individuals carry germline mutations in SMAD4 or
instability–stable (MSI-S). More than 90% of HNPCC tumors BMPR1A.107–109 Nearly all patients with SMAD4 mutations also have
are MSI-H, but approximately 15% of all sporadic tumors are hereditary hemorrhagic telangiectasia110 (overlap syndrome). Both
also MSI-H.75 SMAD4 and BMPR1A are involved in the transforming growth
factor–β (TGF-β)/bone morphogenetic protein (BMP) pathway.
MUTYH-Linked Polyposis
A third hereditary variant of colon carcinoma is MUTYH-linked Other Polyposes
polyposis (MAP), the newest polyposis syndrome, first described in Finally, a variety of polyposis syndromes with potential genetic linkage
2002.76 This is a disorder similar to AFAP and is characterized by have been described and are best exemplified by hyperplastic polyposis
adenomatous polyps that may number in the hundreds but not in syndrome (serrated polyposis syndrome).111,112 This condition consists
the thousands, usually ranging from 10 to 1000.77–79 Polyps may also of the occurrence of multiple sessile polyps variously referred to as
occur in extracolonic sites, usually the duodenum. Adenomas tend hyperplastic polyps or serrated adenomas, frequently on the right side
to occur in the proximal colon.78 MUTYH is a glycosylase and part of the colon and numbering from 5 to more than 100.113 Although
of the base excision repair (BER) pathway. Oxidative damage to DNA familial associations of the syndrome114 and linkage to high risk of
frequently results in the modification of guanine (G) to 8-oxoG. colon carcinoma115 have been reported, the genetic basis is complex
8-oxoG can pair with adenine (A) during DNA replication, a mismatch and associated with mutations in several driver genes including BRAF
Pathology, Biomarkers, and Molecular Diagnostics  •  CHAPTER 15 231

and KRAS.112 No single genetic mechanism has been linked to the in mortality from colon cancer. Colonoscopy enables direct identifica-
syndrome, and the true frequency is not established. To date, the tion of potential precursor lesions and at the same time provides a
number of reported cases is small. means for ablating them. This is strikingly evident in the new sig-
moidoscopy findings37 indicating that whereas sigmoidoscopy reduces
Biomarkers for Colon Cancer Screening mortality from visible distal colon tumors, mortality from proximal
Genetic tests lesions is unaffected. It seems likely that colonoscopy will be the main
Although the number of highly penetrant mutations that predispose to vehicle for early detection and intervention in colon cancer for the
colon carcinoma is large, the proportion of colon carcinomas that can foreseeable future.
be accounted for by these mutations is approximately 5%43 and has
been overestimated in the past.75 Markers of hereditary predisposition Lung Carcinogenesis and a Known Carcinogen
and molecular progression in sporadic tumors that have been delineated
to date have not been easily converted into an early detection test For several decades lung carcinoma has had the highest mortality of
suitable for broad population-based applications for several reasons. all tumors, accounting for more than twice as many deaths as the
First, the types of accessible specimens that can be used for testing for next most lethal tumor.123,124 Initial large-scale efforts during the 1970s
early detection consist primarily of blood and fecal DNA. Although and 1980s to reduce mortality by early detection using chest radiography
DNA from invasive tumors can be detected in the blood by PCR- and/or sputum cytology were not successful. Chemoprevention
based mutation testing, it is not as useful at earlier polyp stages. In using β-carotene and/or α-tocopherol dietary supplementation also
addition, collection and processing of fecal specimens for a biomarker failed.125–128 However, early detection efforts have begun to yield some
that is expected to be present at low level and heavily contaminated success in reducing mortality even in this problematic tumor. Low–
by nonneoplastic cells is a logistic challenge. Second, some of the radiation dose computed tomography (LDCT) has a higher clinical
genetic abnormalities identified in colon carcinogenesis do not readily sensitivity for detecting smaller tumors than chest radiography. Early
lend themselves to screening assays. Mutations in APC, for example, studies in smokers documented a 2.7% prevalence of malignant disease
consist mainly of truncations and deletions that may occur over a detected by LDCT versus 0.7% in the same patients screened by chest
wide region of the gene, so designing a sensitive clinical assay has radiography.129 In a multicenter study of more than 30,000 smokers,
been difficult. Finally, wide availability of colonoscopy services that LDCT revealed lung cancer in 1.3% at baseline and an additional
combine polyp detection and excision reduces the cost and improves the 0.3% at annual follow-up examinations, for an overall total detection
attractiveness of direct and systematic colonoscopic population-based rate of approximately 1.5%.130 Although this study was not randomized
surveillance. and had no unscreened control arm, it showed a survival advantage
in comparison to historical controls. Finally, a large randomized trial
Other screening biomarkers of computed tomography (CT) versus radiography conducted annually
Biomarkers that have been most successful for colon cancer screening in three rounds found that LDCT enabled detection of carcinoma in
are fecal tests such as the guaiac fecal occult blood test (FOBT), the 2.4% of the screened high-risk population and resulted in a 20%
fecal immunochemical test (FIT), and the stool DNA (sDNA) test. reduction in lung cancer mortality that was not observed in the chest
The greatest experience has been with the FOBT. This test is based radiograph control group.131 The weight of evidence supporting
on a color change whereby α-guaiaconic acid extracted from the wood improved long-term mortality rates in smokers has prompted the
of tropical Guaiacum tree is rapidly oxidized by hydrogen peroxide publication of multisociety consensus guidelines recommending annual
to a blue quinone in the presence of heme derived from blood. There LDCT screening for smokers and former smokers aged 55 to 74 years
is wide variability in results116 as a consequence of the level of compliance with access to appropriate follow-up.132 This study has raised awareness
with collection methods, type or brand of kit, frequency of testing, of possible benefits of early detection of lung cancer but has been
user interpretation, and other factors. Despite a lack of standardization, criticized for possible overdiagnosis,133 a question that is currently
FOBT screening with follow-up colonoscopy is reported to reduce unresolved.
both incidence117 and mortality118,119 from colon carcinoma. The former
is attributed to polyp removal. FIT is a second test for occult blood Premalignant Lesions in the Lung
and is reported to be more sensitive and specific than the guaiac assays. What is clear is that lung carcinogenesis is a multistep process and
It has been shown to have a cancer detection rate similar to colonoscopy that premalignant lesions exist in both central and peripheral airways
but has a lower sensitivity for polyps.120 (Figs. 15.5 and 15.6) before invasive carcinoma occurs.
Occult blood tests measure host response (hypervascularity and
bleeding) to tumor. The sDNA test, however, aims to measure a direct Central airway premalignancy: squamous dysplasia
product of tumor cells. A recent version of the sDNA test measures The predominant premalignant lesion of the central airways is squamous
multiple markers in stool using an array-based platform121 and for dysplasia134 (also referred to as atypia). Squamous dysplasia was docu-
this reason has been referred to as “next generation,” although this mented over 50 years ago in association with carcinoma135 as well as
technology does not employ deep sequencing methods and should in smokers without concurrent carcinoma.136 Central squamous lesions
not be confused with “next-generation sequencing.” The biomarkers are flat, inconspicuous, and only rarely identified by white light
that are incorporated into the platform include four methylation probes, fiberoptic bronchoscopy. They are somewhat better visualized with
as well as mutated KRAS and hemoglobin. Patterns of gene methylation laser-induced fluorescence emission (LIFE) bronchoscopy.137 This
have been identified in colon tumors that have been characterized as method relies on a loss of autofluorescence and signal from dysplasias;
CpG island methylation (CIMP) high and CIMP low (reviewed by it may be mimicked by fluorescence loss caused by mucosal hemorrhage
Kim and colleagues122). When applied to a preliminary cohort of 678 and other reactive changes, so whereas sensitivity of LIFE bronchoscopy
subjects with polyps or CRC, the combination test including methyla- is high, specificity is low.
tion markers, KRAS mutation, and occult blood had a sensitivity of Prospective studies on the power of dysplasia to prospectively predict
85% for carcinoma and 54% for polyps. future central airway malignancy are few and face the forbidding
Despite the availability of these fecal-based tests, the target in early challenges of inaccessibility of the bronchi and the prolonged follow-up
colon cancer intervention appears to be shifting from detection of that is required to achieve significant results in a sufficiently powered
invasive carcinoma to prevention by detection and thorough clearing cohort. Sputum studies indicate that atypia of cells exfoliated from
of adenomatous polyps from the colon. Defining the biology of colon the central airways is associated with elevated risk for incidental
carcinogenesis and confirming that polyps are the precursor lesions carcinoma. Hazard ratio increases with proximity of sputum collection
along with improvements in colonoscopic methods have led to reduction date to lung cancer diagnosis, but sputum atypia even 3 years before
232 Part I: Science and Clinical Oncology

Cellular Carcinogenesis, Lung

Normal bronchiolar
epithelium
#1 Low-grade
squamous dysplasia
#2 High-grade
squamous dysplasia
#3 Invasive
squamous carcinoma
#4

Normal alveolar septum


#5
Atypical adenomatous
hyperplasia
#6 Adenocarcinoma
in situ
Invasive adenocarcinoma
#7
#8

Progression over time (years) LDCT/resection

Figure 15.5  •  Multistep carcinogenesis in the lung is complicated by the heterogeneity of tumor types that arise in separate locations in the respiratory
tract. Squamous tumors typically form in the central airways, where they are thought to arise from 1, the respiratory mucosa. The earliest premalignant lesion,
2, low-grade squamous dysplasia, exhibits a polarity in which dark basaloid cells mature into large cells that flatten along the surface as seen in the cervix (see
Fig. 15.1). 3, High-grade squamous dysplasia exhibits less distinct maturation and a higher level of cytologic diversity. 4, Invasive squamous cell carcinoma
occurs when tumor cells extend in the underlying stroma, sometimes forming a mass visible on low–radiation dose computed tomography (LDCT). Adeno-
carcinomas typically arise peripherally in 5, the alveoli and terminal bronchioles. The best-defined precursor lesion of adenocarcinoma is 6, atypical adenomatous
hyperplasia (AHH). These are small lesions (<5 mm) in which the alveolar architecture is preserved. 7, Adenocarcinoma in situ (AIS) is a further progression
of peripheral airway epithelium toward malignancy. This category represents a redefinition of the low-grade adenocarcinoma previously known as bronchioloalveolar
carcinoma. These lesions are solitary small (≤3 cm) lesions consisting of a single cell layer that spreads in a nondestructive pattern of growth along the alveolar
septae that is referred to as lepidic and is detectable as a ground-glass nodule by LDCT. 8, Invasive adenocarcinoma spreads beyond the alveolar surface,
forming a mass that appears solid on LDCT. The presence of a mass visible with LDCT suggests tumor. However, by the time this occurs, tumor is often
invasive and must be treated aggressively.

Phases of Molecular Carcinogenesis, Lung


Early premalignant
Tobacco smoke exposure Late premalignant
Inflammatory changes Clonal proliferation
Angiogenesis Lateral spread
Angiogenesis
Diploidy Malignant
Bulky adducts, Vertical spread
Tissue invasion
Mitotic recombination
LOH (allelic loss, UPD)
Methylation myc dysregulation
Telomerase expression TP53, KRAS, APC,
Other TSG mutations
Further mitotic recombination
Methylation
Chromosomal instability
Array signature
Aneuploidy, translocation
Resistance to treatment

Age/years

Figure 15.6  •  The molecular pathology of smoking-related lung cancer begins with the direct interaction between metabolites of carcinogenic polyaromatic
hydrocarbons in cigarette smoke with host DNA. This creates bulky adducts that affect DNA repair and create transcription errors. The result is a wide range
of genomic losses, gains, translocations and point mutations that account for the high degree of molecular heterogeneity in lung tumors with diverse physiologic
effects. LOH, Loss of heterozygosity; UPD, uniparental disomy; TSG, tumor suppressor gene.
Pathology, Biomarkers, and Molecular Diagnostics  •  CHAPTER 15 233

lung cancer diagnosis is associated with increased risk of lung been sufficiently informative to guide intervention in potentially affected
carcinoma.138 individuals.148–152
Bronchial biopsy is an alternative way to evaluate the status of the
airway in high-risk smokers. A recent prospective study of 188 patients Tumor suppressor gene methylation
who underwent biopsy at mapped bronchial sites found that the DNA damage in lung premalignancy engenders a number of genetic
persistence of high-grade dysplasia at the same bronchial site was and epigenetic changes that provide a cellular survival or proliferative
associated with future squamous carcinoma but not adenocarcinoma, advantage to affected bronchial epithelial cells. Perhaps the earliest
suggesting that persistent dysplasia may be a marker of progressive molecular events may be epigenetic and include DNA methylation,
field change.139 This study also illustrates the multiyear timeline and histone modification, and alterations in miRNA expression (reviewed
multifocality of premalignant changes. by Jones and Baylin153 and Berger and colleagues154). The greatest
The corollary question of whether ablation of premalignant central experience regarding the use of epigenetic changes in the context of
airway lesions could reduce lung cancer mortality remains an open one. early detection of lung cancer has been obtained with regard to
The findings of a chemoprevention study140 using iloprost, a prosta- methylation. Methylation refers to the conversion of cytosine to
cyclin agonist, have suggested that reversal of premalignant dysplasia 5′-methylcytosine by DNA methyltransferase (DNMT). In lung
morphological features may be possible, but the study was a small carcinogenesis, methylation preferentially occurs in CpG-rich regions
one and the end point was purely histological. The long-term effect (CpG islands) that are present in normally unmethylated promoter
on mortality of agents that may alter the premalignant morphologic regions of critical genes (hypermethylation) and may silence gene
characteristics of central airway epithelium remains unknown. expression. In lung tumors, interest has focused on genes such as
p16,155 which are silenced in tumors but expressed in normal control
Atypical adenomatous hyperplasia, adenocarcinoma in situ, tissue, and are considered to be TSGs. The development of a simple
and lepidic carcinoma and sensitive assay based on the conversion of methyl cytosine to
Atypical adenomatous hyperplasia (AAH), adenocarcinoma in situ uracil in the presence of metabisulfite followed by PCR with primers
(AIS), and pure lepidic carcinomas consist of alveolar epithelial cell specific for the methylated bases, methylation-specific polymerase chain
proliferations that do not invade the alveolar wall or elicit a significant reaction (MSP),156 has greatly facilitated the assessment of methylation
stromal response.141 AAH and AIS are small (<5 mm) solitary lesions status as a risk marker for lung carcinoma.
that are usually recognized incidentally in screening trials or in clinical Methylation of key TSG promoter regions in cells exfoliated from
practice by their ground-glass appearance on CT imaging. AAH and bronchial surfaces into the sputum is nearly universally associated
AIS are characterized by indolent growth rate and 100% survival after with prevalent lung carcinomas157 and frequently also occurs in
resection. Larger solitary ground-glass lesions of pure lepidic carcinomas sputum of smokers without tumor. TSG methylation is not present
as well as some lesions with areas of consolidation and even multifocal in sputum of never smokers. This finding initially led to optimism
ground-glass lesions also are characterized by slow growth and excellent that methylation could be used to screen selected populations at risk
prognosis and may persist for many months to years without changing for lung carcinoma. However, the predictive power of single-gene
size or evincing evidence of alveolar wall invasion.142–145 It appears methylation or methylation of a small number of genes is limited by
that the time to progression of premalignant lesions in both central the ubiquity of methylation in smokers and the consequent high rate
and peripheral airways is long and capricious, complicating assessment of false-positive results. This problem led to the strategy of testing
of prognosis and intervention strategies. promoter regions in large numbers of genes to estimate overall
methylation burden.158 Identification of panels of genes to optimize
Molecular Changes During Lung Carcinogenesis the predictive power of methylation testing is a process that is still
As in other organs, most notably the colon, molecular changes that underway. In receiver operating characteristic (ROC) curve analyses, a
accompany the morphologic changes of dysplasia have been investigated seven-gene methylation profile selected from a 31-gene panel achieved
in considerable detail. These studies are informed by the well-established a predictive accuracy of 71% to 77%. However, with a sensitivity
role of tobacco smoke as a carcinogen in lung cancer. The dominant set at 75%, the false-positive rate was 75%.159 Improved results were
carcinogenic compounds found in cigarette smoke are the polyaromatic obtained in a case-control study of CT-detected nodules by using a
hydrocarbons (PAHs) and nicotine-derived nitrosamine ketone three-gene combination having a sensitivity and specificity of 98% and
(NNK).146 These compounds are metabolized to highly reactive 71%, respectively.160 Although TSG promoter methylation may prove
intermediates that are inactivated largely by mitochondrial glutathione problematic as a general screening tool, it may be useful in specific
transferases (GSTs). However, repair is imperfect and a small proportion niche populations or to supplement other methods of screening such
of the intermediates react directly with DNA, forming bulky adducts. as LDCT, and ongoing studies aim to further evaluate the use of this
Unless removed by the DNA repair system, adducts can create errors approach.
of replication. Over time, molecular alterations accumulate, providing
a selective advantage to mutant cells for growth and survival. Aneuploidy
Components of this pathway have been associated with increased A second molecular consequence of tobacco exposure is chromosomal
risk for future lung cancer. Case control studies indicate that current instability, which is detected as aneuploidy with fluorescence in situ
smokers with tumors have higher levels of adducts in blood or tissue hybridization (FISH) in bronchial epithelial cells in situ and in
than non-tumor controls.147 However, bulky adducts appear to reflect sputum. Aneuploidy is nearly universal in lung carcinoma. It is also
ongoing smoking-related DNA damage rather than biologic changes frequently observed in high-grade dysplastic lesions in the central
specifically associated with malignant transformation. Clinically airways.161 Aneuploidy in sputum has been associated with prevalent
applicable adduct levels predictive of lung cancer have not been carcinoma162,163 and has recently been shown to have a sensitivity of
established. 76% within a detection window of 18 months before the appearance
GST and DNA repair enzyme mutations and polymorphisms of invasive carcinoma.164 However, FISH performed on sputum is
that affect DNA repair activity could represent a risk factor for lung labor-intensive and difficult to perform both in the specimen prepara-
cancer because efficient deactivation of reactive carcinogens and tion and slide interpretation, requiring a high level of expertise at
repair of DNA damage caused by bulky adducts are important in every step. This has led to the development of automated methods
protecting potential precursor cells from critical mutational changes. of FISH screening. However, to date these automated methods have
However, single-nucleotide polymorphism (SNP) association studies not been broadly applied. It seems likely that FISH may most suc-
in many carcinogen-metabolizing and DNA repair enzymes have cessfully be applied in restricted settings in conjunction with other
demonstrated only weak and inconsistent associations and have not technologies.
234 Part I: Science and Clinical Oncology

TP53 mutation refers to destructive, invasive malignant tumors that form solid masses.
Mutations in several tumor-associated genes have been documented The primary tool of the pathologist remains the hematoxylin-and-eosin
in adjacent nonmalignant airways or in subjects at high risk for stained section, but this is now supplemented by an arsenal of
carcinoma but without concurrent malignancy. Chief among these is immunohistochemical stains and other tissue testing procedures that
TP53, which marks clonal populations that may spread laterally across permit much greater specificity of diagnosis than was previously possible.
the bronchial surfaces without invading stroma. Mutation in TP53 Diagnostic immunohistochemical tests are relatively simple and
appears late in lung carcinogenesis; and although it is one of the most inexpensive and can be assessed in conjunction with in situ cellular
commonly mutated gene in lung carcinoma, it is mutated in only architecture, which ensures a high level of specificity and permits an
half of tumors and in fewer benign airways. It seems unlikely that assessment of the relevance of results to the overall clinical and anatomic
single-gene mutation analysis will be sufficiently sensitive for use as features. Interpretation of immunohistochemical data is now assisted
a screening tool. by online databases that provide frequencies of immunohistochemical
results in specific tumors and organ sites. Histologic diagnosis was
High-throughput technologies discussed earlier in the context of early detection and is addressed
High-throughput methodologies have been suggested as possible early elsewhere in this book in relation to individual tumor types.
detection tools. Genome-wide association studies (GWASs) have Although histologic evaluation remains the primary method for
identified an SNP variant at 15q15.2 that is significantly associated diagnosis of solid tumors, molecular methods are increasingly influential
with cancer risk, with an overall odds ratio of 1.15.165 The low odds not only in identification of treatment targets as discussed here, but
ratio suggests that the SNP variant itself will not represent a useful also in tumor classification. In CNS tumors, for example, molecular
predictor. However, this region of the gene is still being explored for findings are such powerful predictors of biologic behavior that molecular
more powerful biomarkers. features have been formally incorporated into the diagnostic classifica-
Gene expression microarrays have identified consistent changes in tion of this complicated group of neoplasms (see later). Both molecular
the transcriptome of epithelium brushed from nonmalignant bronchial and cellular diagnostics continue to depend not only on consistent
surfaces that distinguish smokers from nonsmokers.166 An additional and effective fixation and processing of tissue, but also on reliable
set of changes are reported to identify subjects with cancer from accessioning and informatics.
smokers without cancer with an accuracy of 83%.167 Finally, large-scale
genomic sequencing studies have identified differences in the “genomic Algorithm for Cellular Molecular Testing
landscape” between smokers and nonsmokers.168 It is expected that
manageable numbers of biomarkers and clinically applicable testing Molecular and cellular tissue testing is a multistep process (Fig. 15.7),
platforms will emerge from these exploratory studies that will improve and success requires tight control of collection, accessioning, fixation,
risk assessment and management of high-risk smokers as well as patients dissection, extraction, and storage of tissue as discussed in the following
with already invasive carcinomas. sections.
Accessioning and Informatics
Other Organ Sites
Registering patients and biopsy specimens is a demanding exercise
Premalignant cellular and molecular changes are described in several that is often overlooked and underfunded. Although numerous and
other tumors including breast, prostate, bladder, and esophagus. variable protocols are available for tracking specimens, virtually all
Detailed review of these changes may be found in appropriate chapters tracking systems include a secure electronic database with password-
of this text. However, to date, identification of these changes has not limited access and reliable backup. Appropriate specimen identifiers,
led to mortality reductions. which today are increasingly bar-coded, access to signed consent forms
when associated research studies are underway, and timely data entry
Conclusion: Cells and Molecules in Early Detection are all essential for any well-run laboratory. A detailed discussion of
laboratory information systems for molecular testing is beyond the
The examples described above illustrate the uneven progress that has scope of this chapter but must be a major consideration in the manage-
been made and the challenges that remain in reducing morbidity and ment and quality assurance programs of all clinical molecular testing
mortality from cancer. What emerges from this review is that progress laboratories.
depends on improved understanding of site-specific carcinogenesis
and intervention strategies that are informed by basic biology. Relatively Tissue Collection and Processing
simple technologies such as cytologic evaluation or HPV testing can Tissues obtained for solid tumor molecular testing may come from
profoundly reduce cancer burden. Understanding the biology of surgical resection or biopsies (fine-needle aspirates and cores). Most
premalignancy provides a lead-time advantage that can be translated often, the tissue needed for molecular testing is derived from diagnostic
into greatly reduced cancer morbidity and mortality. Appropriate material, unless special arrangements are made to specifically collect
molecular strategies may supplement and eventually replace morphology tissues for molecular studies. Issues regarding specific specimen types
but will require thorough validation. are discussed in the following sections.
Resection Specimens
DIAGNOSIS AND CLASSIFICATION OF SOLID Resection specimens are specimens obtained as a result of surgical
MALIGNANCIES: HISTOLOGY AND procedures to remove tumors for therapeutic purposes. These specimens
EXPANDING ROLE OF MOLECULAR provide the largest amount of material for molecular testing. Several
DIAGNOSTICS aspects of specimen collection must be borne in mind to optimize
resection specimen quality and yield.
Histology of tumor tissue has been the bedrock on which diagnosis Gross dissection. Identification of tumor tissue in resected
of malignancy is established and is the basis for selecting appropriate specimens requires a high level of morphologic and dissection skill.
testing to guide treatment. Histologic diagnosis, although simple in For this reason, it is essential that a trained pathologist be involved
concept, is extremely complex in practice and crucially depends on in establishing protocols for the collection of specimens for molecular
the expertise of the examining pathologist. In solid tumors, a nearly studies. For DNA-based assays including mutation analysis, timing of
universal feature of malignancy is the capacity of tumor cells to invade the collection procedure can be somewhat flexible because DNA is a
and replace stromal tissue, and nearly all of the following discussion particularly robust analyte. However, efforts should be made to avoid
Pathology, Biomarkers, and Molecular Diagnostics  •  CHAPTER 15 235

Preanalytical
processing
1. Estimate tumor cell content
2. Isolate tumor and/or normal cells
A. Whole sections
B. Coring or
C. Trimming of block
D. Microdissection DNA
a) Scrape extraction
b) Laser capture
1. Deparaffinization
2. Proteinase K digestion
3. Precipitation/capture Library
4. Shear DNA
a) Sonication preparation
b) Enzymatic
1. Target enrichment
a) Hybrid capture Massively parallel
b) Amplicon
2. Quantitation sequencing
3. Pooling
1. Ion torrent
2. Illumina Data
analysis
1. Raw data analysis (base calling)
2. Mapping to genome
3. Variant calling Clinical
4. Variant annotation report
1. Adequacy
2. Depth of coverage
3. Frequency of mut
4. Predicted phenotype
5. Drugs/trials available
6. Referral to genetic
counseling (germline)

Figure 15.7  •  Steps entailed in obtaining optimal results for clinical next-generation sequencing (NGS).

prolonged exposure to formalin fixation, or exposure to harsh acids in increasing reliance on small biopsy specimens for diagnosis and
such as those that can be encountered during decalcification treatments. monitoring of treatment. The primary objective of these tissue acquisi-
Specimen preservation.  Fresh tissue, tissue preserved in transport tion methods has been to obtain tissue for unequivocal diagnosis,
media such as RNAlater, or frozen samples are acceptable starting which can require multiple immunohistochemical stains in addition
materials for molecular testing. However, these samples may be to conventionally stained slices. Meeting this objective can leave little
inconvenient to process in clinical laboratories and have the disadvantage remnant tissue behind for ancillary studies. When tumor is of an
that tumor cell purification by macrodissection or microdissection advanced stage and surgery is not appropriate, remnant tissue from
may be more difficult and complicated than with paraffin sections these diagnostic biopsies may be the only tissue available for molecular
cut from fixed tissue. Formalin-fixed paraffin-embedded (FFPE) testing. As morbidity from biopsy procedures has declined and the
specimens are now routinely used for molecular testing. The most value of information derived from molecular testing has become more
cost-effective fixative is 10% buffered neutral (pH 7.4–7.6) formalin. critical, performing biopsies solely for molecular analysis has gained
Buffering is needed to eliminate possible DNA depurination, which increasing acceptance by clinicians and patients alike. The major
may occur at acidic pH. Phosphate is the preferred buffering salt. limitation of biopsy specimens for molecular testing is small tumor
Formalin works by cross-linking amino groups in proteins, introducing cell yield. Also, testing of DNA extracted from paraffin sections must
a -CH2- linkage. This linkage is stable and permits long-term storage take into account shearing of DNA caused by sectioning. Despite
of tissues in paraffin with minimal degradation. Fixatives such as these challenges, isolation of tumor DNA from small specimens of
acetone and alcohol work by rapidly dehydrating tissues. These fixatives many types including tissue cores, cell block preparations, smears and
may not completely inactivate autolysis and may require modification imprinted slides has been well documented.171–174 Individual laboratories
of testing platforms. Metal-based (e.g., zinc, mercury) fixatives, which must validate testing protocols for each specimen type.
are favored in some contexts because of the excellent histologic detail
they provide, may inactivate polymerases used in molecular analyses. Enriching for Tumor Cells
For this reason, specimens exposed to metal-based fixatives cannot be Because nonmalignant cells invariably accompany (“contaminate”) all
used for molecular biomarker studies. Finally, acid decalcification clinical samples, specimens must be enriched for tumors to a level
causes DNA deterioration and may result in amplification failure. that is consistent with the analytic sensitivity of the platform that will
Decalcification status should be kept in mind in interpretation of be used for testing. As a rule of thumb, a minimum of 100 viable
testing results. tumor cells in a 10-µm section is required for a single PCR-based
assay. Quality of the biopsy specimen may be more important than
Biopsies quantity for critical molecular studies.175,176
Improved imaging and tissue sampling techniques in recent years, Although tumor enrichment methods may vary from laboratory
exemplified by advances in lung sampling methods,169,170 have resulted to laboratory, it is essential that tumor enrichment methodology be
236 Part I: Science and Clinical Oncology

appropriately matched to the testing platform. At least five different appearance of mutations associated with acquired resistance to targeted
procedures may be used for tumor cell enrichment, depending on the therapies, a setting in which serial biopsy may not be feasible. The
amount of contaminating normal tissue that is present in the specimen: identities and methods of testing for these target sequences are discussed
(1) en face sectioning of a trimmed block, (2) coring of paraffin blocks in the following sections.
in regions enriched for tumor cells, (3) macrodissection, (4) manual
microdissection, and (5) laser capture microdissection. Macrodissection TREATMENT TARGETS AND
can refer to the practice of scraping of tumor cells from a glass slide, MOLECULAR ANALYSIS
typically done without direct microscopic visualization. Manual
microdissection, by contrast, involves the use of some form of micro- At the start of the millennium, the sequencing of the human
scopic assistance to identify tumor cells. Manual microdissection is genome192–194 and the discovery that somatic mutation could activate
typically performed by using a dissecting microscope and usually transmembrane receptors and tumorigenicity195 led to anticipation
involves counterstaining of slides to identify regions of interest, which that anticancer drugs affecting this process would be found in abun-
can be removed from the slide with a scalpel, needle, or glass micro- dance.196 The Cancer Genome Atlas (TCGA) program was launched,197
pipette. Laser capture microdissection involves melting of supporting in part, to identify essentially all mutations that might be subject to
matrix with a laser beam created with an epifluorescence lens and external regulation by specific pharmaceuticals. TCGA revealed the
removal of the target cells for extraction and quantitative testing of large mutational burden in many solid tumors,198 particularly in those
DNA and other analytes. Although more precise than manual microdis- that have been associated with a high degree of genetic damage caused
section, laser capture microdissection is slower, is costlier, and generates by physical or chemical agents such as ultraviolet light in melanoma199,200
lower yields of extraction products. and tobacco smoke in lung carcinoma.201,202 TCGA and other deep
sequencing studies have also revealed the complexity, heterogeneity,
Liquid Biopsy and lability of mutations in tumors.203–208
Obtaining tissue for microscopic examination requires invasive pro- Among the most relevant findings of TCGA has been the confirma-
cedures and a significant commitment of resources but assesses only tion that mutation of proteins in the tyrosine kinase signaling pathway
a limited portion of tumor. So-called “liquid biopsy” circumvents is common in human tumors and that many are affected by the
these limitations by testing peripheral blood. Testing can use either increasing number of FDA-approved inhibitors that are now available
isolated circulating tumor cells (CTCs) or extracted cell-free tumor for treatment of both solid (Table 15.3) and liquid tumors (Table 15.4;
DNA (ctDNA) from biologic fluids. discussed elsewhere in this book). Tyrosine kinase receptors (TKRs)
CTCs are produced by shedding of tumor cells into the lympho- are transmembrane immunoglobulin-like molecules209 containing an
vascular system. They are usually found in very low abundance in extracellular ligand-binding region, a transmembrane region, and an
blood (on average ~1 cell per milliliter), and therefore require meticulous intracellular tyrosine kinase region that contains tyrosine residues whose
methods to isolate them from the large numbers of contaminating phosphorylation regulates signal transduction. Specific mutations in
normal blood cells.177 This is typically achieved by using cell-surface tyrosine kinase domains enhance phosphorylation and stimulate
markers specific for cancer cells—for instance, the epithelial cell downstream intracellular signaling. The physiologic result is accelerated
adhesion molecule (EpCAM) that is found on most epithelial-derived cell proliferation, extended cell survival, and increased angiogenesis
tumor cells but not normal blood cells.178 Alternatively, physical and cellular migration, properties that are hallmarks of carcinogenesis.1
differences between CTCs and normal blood cells, such as size, Although mutations cluster around hot spots in the receptor, they
deformability, and bioelectric differences, can be used to achieve isola- may also occur at less common loci. In addition, unexpected tyrosine
tion.177 Regardless of the isolation technique, this approach is hampered kinases and other signaling molecules may drive tumor cells, so it
by the low yield of isolation methods. However, CTC analysis, in is increasingly important that sequencing techniques have broad
particular overall detection of CTCs or quantification of CTCs, has coverage. For this reason and for economies of scale, next-generation
demonstrated benefits in cancer diagnosis, prognosis assessment, and sequencing (NGS) is now widely used to identify treatment targets in
monitoring of treatment response.179–182 solid tumors.
Circulating ctDNA consists of short (~140–170 bp) fragments of
DNA found in blood or urine. It is derived from both normal and DNA: Next-Generation (Massively Parallel)
tumor cells undergoing apoptosis, necrosis, or release from live cells.183 Sequencing as a Biomarker Predicting Response
Fragments are typically short because they are derived from DNA to Treatment
that wraps around the nucleosome and is thus protected from degrada-
tion. The percentage of free DNA derived from tumor varies widely A number of small-molecule tyrosine kinase inhibitors (TKIs) specific
(0.01%–93%) and is influenced not only by the amount of ctDNA for these receptors have been designed to block phosphorylation and
released by cancer cells but also the total load of cell free DNA, which suppress tumor growth and spread. In addition, the extracellular
itself can vary dramatically from person to person.184 To distinguish domains of these molecules have proven to be antigenic, and therefore
tumor from normal DNA, analysis relies on tumor-specific markers the extracellular ligand-binding domain is also a target for therapeutic
(e.g., point mutations). The percentage of ctDNA can be lower than antibody development. Several of these drugs have successfully
the limits of detection of standard DNA sequencing techniques, and transferred to the clinic and the number of such drugs available for
specialized approaches such as allele-specific PCR, digital PCR, and cancer treatment is likely to continue to expand, creating an ongoing
presequencing enrichment of mutant alleles are required to achieve demand for more comprehensive molecular testing of malignancies
significant sensitivity.185–187 Also, specialized bioinformatics measures of virtually all types. The increased demand for sequence coverage is
can be undertaken in next-generation tests to reduce background being filled by NGS.
noise and improve lower limits of detection.188 Despite these efforts,
typical reported sensitivity for detection of ctDNA ranges from 49% Definition
to 78% for localized tumors and 86% to 100% for metastatic tumors, NGS refers to techniques in which millions of nucleotide sequences are
depending on tumor type,189 the yield of positive results is sufficiently deciphered simultaneously (reviewed by Goodwin and colleagues210)
high that the test may provide diagnostic, prognostic, and disease and is often called massively parallel sequencing. The complete sequenc-
burden information in a significant patient fraction without the need ing of the human genome during the early 2000s192–194 engendered
for biopsy.189–191 As ctDNA typically provide sequence data, this a need for relatively inexpensive, high-throughput sequencing
approach can also be used to detect actionable genetic alterations.185 methods, a need that was met by several NGS platforms that were
This is particularly useful in the serial monitoring of blood for the quickly commercialized. NGS platforms have proven to be highly
Pathology, Biomarkers, and Molecular Diagnostics  •  CHAPTER 15 237

Table 15.3  Molecularly Targeted Agents Approved by FDA for Solid Tumors

Tumor(s) Gene(s) or Protein Molecular Lesion(s) Drugs


Breast ER or ER/PR Overexpression Fulvestrant, Tamoxifen, Anastrazole, Exemestane,
Letrazole
Breast CDK4/6 Expression/overexpression Ribociclib, Palbociclib + Fulvestrant or Letrazole
(for ER+, HER2−)
Breast/Stomach HER2/neu Amplification/overexpression Everolimus, Lapatinib, Pertuzumab, Trastuzumab,
Ado-trastuzumab Ematansine, Niratinib
Colon KRAS Absence of mutation Cetuximab, Panitumumab
Colon/NSCLC EGFR Overexpression/increased gene copy number Cetuximab, Panitumumab, Necitmumab
GIST Kit/PDGFRα Activating mutations Imatinib, Midostaurin (PKC412)
Melanoma KIT Activating mutations Imatinib
Melanoma/Colon/Lung BRAF Activating mutations Vemurafenib/cobimetinib, Dabrafinib/trametinib
NSCLC ALK Gene rearrangement (multiple fusion Crizotinib, Ceritinib, Alectinib, Brigatinib
partners)
NSCLC EGFR Activating Mutation (small deletion/point Erlotinib, Gefitinib, Afatinib, Osimertinib
mutation)
NSCLC EGFR T790M Resistance mutation Osimertinib
NSCLC ROS1 Gene rearrangement (multiple fusion Crizotinib
partners)
NSCLC BRAFV600E Activating muation Dabrafinib + Trametinib
Soft tissue sarcoma PDGFR alpha Expression/overexpression Olartumab
Prostate Androgen receptor Expression/overexpression Abiraterone Acetate. Bicalutamide, Cabazitaxel,
Casodex, Degarelix, Enzalutamide, Flutamide.
Ovary BRCA Mutation, PARP inhibition Venatoclax

GIST, Gastrointestinal stromal tumor; NSCLC, non-small cell lung carcinoma.

Table 15.4  FDA Approvals of Targeted Agents for Hematologic Malignancies


Tumor(s) Gene(s) or Protein Molecular Lesion(s) Drugs
ALL B cell CD19, CD3 Expression/overexpression Blinatumomab (bivalent)
ALL B cell CD19-directed genetically Expression/overexpression Tisagenlecleucel
modified autologous T cell
ALL B cell CD22 Expression/overexpression Inotuzumab zogamicin
AML CD33 Expression/overexpression Gemtuzumab ozogamicin
AML FLT3 Activating mutations Midostaurin/lestaurinib
AML IDH2 Activating mutations Enasidenib
B cell lymphoma CD20 antigen Expression/overexpression Rituximab, Tositumomab, Ofatumumab,
Ibritumomab-tiuxetan (radiolabeled)
CLL CDK4/6 Expression/overexpression Idelalisib
CLL/SLL Bruton tyrosine kinase Expression/overexpression Ibrutinib
CLL/SLL CD 52 antigen Expression/overexpression Alemtuzumab
CLL/SLL CD20 antigen Expression/overexpression Obinutuzumab, ofatumumab
CML BCR/ABL Activating mutations Imatinib, Dasatinib, Nilotinib, Bosutinib, Ponatinib
CML, Ph+ ALL c-Kit/PDGFRα Activating mutations Imatinib, Midostaurin, lestaurinib
Follicular lymphoma, relapsed PIK3 Expression/overexpression Copanlisib
Lymphoma CD30 antigen Expression/overexpression Brentuximab-vedotin (antibody-toxin conjugate)
Mantel cell lymphoma Bruton tyrosine kinase Expression/overexpression Ibrutinib
Multiple myeloma CD38 antigen Expression/overexpression Daratumumab
Myelodysplastic syndrome c-Kit/PDGFRα Activating mutations Imatinib
Myelodysplastic syndrome JAK2 Expression/overexpression Ruxolitinib
Peripheral T cell/HTLV Lymphoma CD25 antigen Expression/overexpression Denileukin-difitox (antibody-toxin conjugate)

ALL, Acute lymphoblastic leukemia; AML, acute myeloid leukemia; CLL, chronic lymphocytic leukemia; CML, chronic myeloid leukemia; SLL, small lymphocytic lymphoma.
238 Part I: Science and Clinical Oncology

quantitative211 and adaptable for multiplex marker testing to meet Library construction is a crucial step in testing. At this stage, the
specific clinical needs.210,212,213 These platforms require a stepwise number of genes and specific mutations must be specified, ranging
approach to tissue processing, nucleotide extraction, and library from a few genes to whole exomes. Two techniques are employed in
preparation for successful biomarker testing; this approach is depicted library creation: PCR amplification and hybrid capture or commonly
in Fig. 15.7. a combination of the two. The size of the library determines how
many tests can be performed in a single or multiple flow cells and
Platforms thus determines the cost of the test.
Currently two platforms dominate the field of clinical NGS testing, Platform-specific sequencing protocols must be thoroughly vetted
Ion Torrent (Thermo Fisher Scientific, Waltham, MA) and Illumina by the testing site. Analytic validation or validation of analytic methods
NGS (Ilumina, San Diego, CA).210 Both methods represent the refers to the assessment of sensitivity, specificity, accuracy, precision,
so-called “sequence by synthesis” approach to sequencing, and both repeatability, reproducibility, detection and quantification limits,
require isolation, separation, and amplification of single DNA strands, linearity, range, and robustness for each analyte. This can be a major
creating a vetted library of primary sequences (see later). However, challenge, and synthetic nucleotides have been introduced to address
the platforms differ in their methods of accomplishing these steps. this problem. An additional approach to analytic validation consists
With Ion Torrent (Fig. 15.8), copies of a single nucleotide template of specimen sharing and estimation of concordance among different
are linked through an adapter sequence to a single bead. Beads are then institutions.
placed in wells engineered into a flow cell, and individual nucleotides
are added one base at a time. Each addition of base into a forming Genetic alterations detected
strand releases a hydrogen ion, resulting a pH change in the well that Two types of mutation can be detected with conventional NGS
is measured in each well, thus registering each base incorporated into platforms: small variants, including SNVs and small indels, and large
the elongating sequence. The numbers of beads containing identical structural abnormalities, including copy number abnormalities and
strands provides a measure of the frequency of the sequence in the overall gene fusions that identify translocations. A remarkable number of
template mixture. clinically relevant consistent driver mutations have been described in
With the Ilumina platform (Fig. 15.9), amplified nucleotide libraries solid tumors during the past two decades, as shown in Table 15.3.
are created from a sample template using defined probes. Creation of Several of these specific mutations are described later.
libraries is a crucial step in NGS that imparts coverage and sensitivity
to testing procedures. Customized reagents214,215 or commercial kits Interpretation and significance of next-generation
can be used in this procedure. Regardless of source, libraries must be sequencing results
continuously monitored for consistency, sensitivity, and specificity. Final interpretation of the data must rest on the judgment of experienced
During Illumina library preparation, end adapters are incorporated clinical laboratory staff. To accomplish this, filtered NGS data must
into the DNA template and are used to attach the template to insoluble be accessible through an online database containing flags for quality
matrix in a “flow cell” at a calculated density. The attached single assurance data and for the presence of clinically significant and
strand is then amplified in a step referred to as bridge amplification, insignificant variants. Reporting personnel should assess all of the
in which strands with adapters at both ends can be bent to bond available data presented to review and render an opinion on the likely
with matrix at both ends. Replication can proceed from either end of clinical significance genomics findings. Reporting and interpretation
the strand, thus providing two copies of each strand for each round of NGS data have undergone scrutiny by representatives of several
of PCR. This process results in the formation of clusters of forward involved organizations, and consensus guidelines and recommendations
and reverse strands that can be visualized by incorporating fluorescent have been published.219 These guidelines recognize the wide range of
nucleotides. The fluorescent clusters are precisely mapped in the flow cell biologic effects of mutations that occur in solid tumors, and participat-
and imaged after addition of each nucleotide. Consecutive images are ing experts have proposed a four-tiered system to classify mutations,
used to establish the sequence at each cluster location on the flow cell. as shown in Table 15.5.
Strand length can reach up to 300 bases. Sequences can be assembled
computationally to cover any length required from multiplexed genes
to whole exons to whole genomes.
Table 15.5  Evidence-Based Variant Categorization
Informatics pipeline
A series of computational steps is performed to assess the large quantity Tier Evidence
of raw data generated by NGS that are collectively known as the 1. Variants of strong FDA-approved therapy; included in
informatics pipeline. Each NGS platform generates base calls from clinical significance professional guidelines
proprietary algorithms that emerge in the form of tab delimited text Well-powered studies with consensus from
files.216 Ilumina systems typically provide FASTQ files that contain experts in the field
sequence information in which each base is symbolized by a single 2. Variants of FDA-approved for different tumor type or
letter and that include a quantitative quality score for each base potential clinical consensus in multiple small published studies
(Phred-like or Q score). These files are fed into sequence alignment significance Preclinical trials or few case reports without
software, which, by comparing test sequences with reference sequences, consensus
can detect single nucleotide variants, short indels, large structural 3. Variants of Not observed at a significant allele frequency
alterations, copy number variations, and gene fusions. These data can unknown clinical in the general or specific subpopulation
be visualized with several available tools, such as the Integrative significance databases, or pan-cancer or tumor-specific
Genomics Viewer217 and SAMTools.218 variant databases
No convincing published evidence of cancer
Quality assurance association
NGS testing is a multistep process as diagramed in Fig. 15.7, and 4. Benign or likely Observed at significant allele frequency in
quality must be maintained at each step. The smallest proportion of benign variants the general or specific subpopulation
tumor cells in a cell mixture (analytic sensitivity) that can be confidently databases
detected with a molecular test must be determined by each testing No existing published evidence of cancer
laboratory. Successful testing can be performed with use of as few as association
50 intact tumor cells in formalin-fixed material.
Pathology, Biomarkers, and Molecular Diagnostics  •  CHAPTER 15 239

Adapters dNTPs

polymerase

DNA

An emulsion of water and oil is created. The water


The DNA library is produced by ligating adapters to droplets contain beads with covalently-bound
segments of DNA generated by fragmentation or PCR. oligonucleotides, dNTPs, polymerase, primers, and the
DNA library. The oligonucleotides capture the DNA,
and PCR extends the complimentary strand.

The bead-bound DNA is applied to a semiconductor chip


that has thousands of microscopic wells. The beads settle
When cycling is complete, thousands of identical DNA into the wells.
molecules are bound to the bead.

dNTPs
primers

Primers and polymerase are added, then nucleotides are An ion-sensitive layer underneath each well records the
washed over the wells one at a time, in several-second change in Ph associated with hydrogen release and
intervals. Each time a nucleotide is incorporated, records the corresponding nucleotide sequence.
hydrogen is released.

Figure 15.8  •  Diagrammatic representation of Ion Torrent testing platform. dNTP, Deoxynucleotide triphosphate; PCR, polymerase chain reaction.
240 Part I: Science and Clinical Oncology

Adapters

Adapters

DNA

The DNA library is produced by ligating adapters to The DNA is applied to the flow cell and captured by
segments of DNA generated by fragmentation or PCR. adapters that are covalently bound to its surface. A copy
of the original strand is created by PCR.

The original strand is washed away, leaving the copy,


which is covalently bound to the flow cell. The bound
DNA then forms a bridge with a nearby adapter, and the Bridge amplification results in a cluster of DNA
complementary strand is generated. This process, called comprising both forward and reverse strands. The
bridge amplification, is repeated to generate thousands of reverse strands are cleaved and washed away, leaving a
copies of the original template. cluster of identical DNA strands ready to be sequenced.

ddNTPs

Primers

The sequencing chemistry uses fluorescent ddNTPs,


This process can image thousands of clusters
which cannot be extended. During each cycle, a single
simultaneously and is commonly referred to as
fluorescent ddNTP binds to each strand in the cluster,
massively parallel sequencing
and the flow cell is imaged.

Figure 15.9  •  Diagrammatic representation of Illumina Next Generation Sequencing platform. ddNTP, Dideoxynucleotide triphosphate; PCR, polymerase
chain reaction.
Pathology, Biomarkers, and Molecular Diagnostics  •  CHAPTER 15 241

Assays for Large-Scale Gene and and p23.2 (Fig. 15.10). These two genes originally are separated by
approximately 12.8 MB. The other described ALK fusions in lung
Chromosome Rearrangements cancer, TFG-ALK,221 KIF5B-ALK,222 KLC1-ALK,223 and PTPN3-ALK,224
Large-scale genomic rearrangements are not easily detected with are caused by chromosomal translocations t(2;10)(p23.2; p11.22),
conventional gene sequencing, and specialized methods have been t(2;3)(p23.2;q12.1), t(2;9)(p23.2;q31.3), and t(2;14)(p23.2;q32.1),
used for decades to demonstrate innumerable complex abnormalities respectively.
for which nomenclature is now standardized220 and uniformly applied Currently, FISH is the most commonly used assay for ALK
in searchable databases (http://cgap.nci.nih.gov/Chromosomes/ rearrangements in lung cancer. The standard FISH assay uses a
Mitelman). Both FISH and molecular methods are validated for dual-color break-apart (BA) probe set encompassing the 3′ and the
application to clinical samples to identify complex rearrangements. 5′ sequences immediately adjacent to the breakpoint area in ALK
Each method has its advantages. In FISH, histologic architecture is (intron 19), labeled in orange and in green fluorophores, respec-
maintained and the technique can be accurately applied to small tively.225 Chromogenic in situ hybridization (CISH) BA assay for
samples. It is also widely available in clinical laboratories. The large ALK rearrangements is also commercially available. Despite interesting
number of FISH tests currently in active use cannot be fully addressed results and a high level of concordance with FISH,226,227 the ALK
here owing to space limitations. However, lung cancer provides an CISH assay has not yet been comprehensively explored. The ALK
example of the relevance of discovery and clinical testing for large-scale BA FISH probe was used in the initial clinical studies with crizo-
gene rearrangements to targeted therapy, as discussed later. tinib228,229 and is approved by the US Food and Drug Administration
(FDA) as a companion diagnostic test. The ALK BA FISH assay is
Fluorescence in situ hybridization assays for DNA based and is thus robust and resistant to technical artifacts.
chromosome rearrangement It has multiple other advantages, such as being highly suitable for
Copy number changes and chromosomal rearrangement are ubiquitous FFPE tissue sections, and detects all variants of EML4-ALK as well
in lung carcinoma, but to date only a small proportion of those as any of the reported or uncharacterized partners. On the other
described are recurrent. An example of a now well-established recurrent hand, the FISH platform requires specialized resources (fluorescence
rearrangement is ALK gene inversion and fusion. In this rearrange- microscope, light-protected laboratory space) and highly trained
ment, ALK is the oncogenic driver switched on by the rearrangement. personnel.
The EML4-ALK fusion is generated by a paracentric chromosomal Two other receptor kinase genes have been identified as acti-
inversion in the short arm of chromosome 2, between bands p21 vated in lung cancers by fusions, ROS1221 and RET.230 Integrated

ALK
Normal 2

3 5 5 3
ALK EML4

3 5 5 3
EML4 ALK
ALK EML4

Inv(2)(p21p23.2)
A EML4:ALK

B C D
Figure 15.10  •  The EML-ALK fusion is generated by a paracentric inversion in the short arm of chromosome 2. The break-apart fluorescence in situ
hybridization (FISH) probe includes DNA sequences centromeric to the breakpoint (5′ ALK) labeled in SpectrumGreen and sequences telomeric to the
breakpoint (3′ ALK) labeled in SpectrumOrange (A). Normal cells carry only fused 3′ ALK–5′ ALK signals, indicated by yellow arrows (B). Cells with rear-
rangement in the ALK gene exhibit split 3′ (orange) and 5′ (green) signals (C) or additional copies of single 3′ (orange) signals (D). Red arrows indicate 3′
ALK signals; green arrows indicate 5′ ALK signals.
242 Part I: Science and Clinical Oncology

molecular and histopathology-based screening systems have been large discrepancies between 5′ and 3′ regions can indicate the presence
successful in detecting a large number of fusion partners in lung of a fusion.244
cancer for these two genes, and preclinical studies have confirmed
their tumorigenic potential.222 Clinical trials of targeted agents for Clinical Testing for Predictive Biomarkers by
ROS1 are currently underway, and rapid expansion of this field is Immunohistochemistry
expected.
Protein biomarkers may be detected in situ by simple and inexpensive
Molecular methods for genomic rearrangement immunohistochemistry (IHC). Cellular morphologic features are
The past few years have also witnessed rapid advances in molecular retained in immunohistochemical methods, which adds to specificity
methods for the assessment of large-scale gene rearrangements. and sensitivity over protein measurement in fluids or homogenates
Molecular methods can be readily standardized, automated and of disrupted cells. In all immunologic detection methods, binding
multiplexed, which is now necessary due to the large number of characteristics of the primary antibody used are crucially important,
known actionable gene fusions. and local familiarity with the idiosyncrasies of each antibody is essential
In the majority of cases of pathogenic fusion genes, the genomic in assessment of results. Examples of immunohistochemical tests that
breakpoints occur in intronic regions. Therefore DNA-based detection are used for clinical decision making are described in the following
of gene rearrangements by molecular methods requires analysis of sections.
introns. Because introns are, in general, far greater in size than exons
(thus requiring far more sequencing), NGS-based assays are typically Abnormal Expression of Target Genes
used in this setting. This can be achieved either through whole-genome ER, PR, and HER2 in breast cancer
sequencing231 or by targeted sequencing of selected introns through Immunohistochemical staining for estrogen receptors (ERs) and
hybrid-capture target enrichment (often termed intron “tiling”),232,233 progesterone receptors (PRs) was one of the earliest applications to
with the targeted approach being much more feasible in the clinical find a place in patient management. Until the early 1980s, antibod-
setting. The detection of rearrangements via DNA-based NGS requires ies to these proteins were not widely available. However, with the
bioinformatics approaches beyond those typically applied to mutation development of monoclonal antibody technology, antibodies that could
calling. In general, these can be divided into two distinct categories. In identify hormone receptors were created245,246 and found to reliably label
one, paired sequencing of DNA molecules of known size is performed. receptors even in FFPE sections of breast carcinomas.247,248 Unexpected
If the two paired ends map to the reference genome at locations results of this testing were that these receptors are typically found in
unexpected based on the size of the input DNA, then it can be deduced the cell nucleus and that the intratumoral and intertumoral range
that a rearrangement occurred within that DNA fragment.234,235 In of expression is large. Because of this, semiquantitative scoring that
the other, mapping to the reference genome at a conserved genomic takes into account intensity of staining and percentage of cells stained
location of a DNA sequence (termed “soft-clipping”) is used to guide has been used to establish expression levels that define positive and
rearrangement detection.236 negative results.249,250 These scoring systems continue to prove their
To avoid these two complications, many fusion detection assays prognostic value.251
query the mRNA (or cDNA derived from the mRNA) instead. A second immunohistochemical test that has been successfully
Accordingly, the amount of nucleic acid to be sequenced is far less, applied to the management of breast carcinoma is the test for HER2
and difficult-to-map repetitive sequences are avoided. The traditional (HER2/neu, ErbB2) expression. HER2 is a TKR and a member of
mRNA-based approach is reverse transcriptase polymerase chain reaction the epidermal growth factor receptor (EGFR) family of signaling
(RT-PCR), in which primers to both genes involved in the fusion are molecules. Its role in human carcinogenesis was initially discovered
used. This technique has been used successfully to detect gene fusions through DNA screening, where it was found to be amplified in 30%
in clinical samples.237 However, the ability to multiplex traditional of the tested breast cancers.252 RNA and protein were shown to be
RT-PCR assays is limited, making it a less than ideal approach to query overexpressed in tumors that are amplified as well as in a subset of
for multiple gene fusions simultaneously. Furthermore, RT-PCR requires nonamplified tumors.253 HER2 testing became crucial when it was
knowledge of the fusion partner (to design primers for the assay), and found that HER2 protein overexpression was a determining factor
for some clinically actionable gene fusions, dozens of known fusion for in vitro and xenograft responsiveness to anti-HER2 monoclonal
partners have been described. Therefore fusion events not in the original antibody treatment.254,255 In early clinical trials, anti-HER2 IHC was
design will not be detected, making this method very biased. NGS-based used as the method for determination of HER2 status and determined
assays using mRNA or cDNA input are a less biased approach in this eligibility for treatment with an anti-HER2 antibody drug, Herceptin.256
regard and can be performed on the whole transcriptome238 or in a A standardized approach to HER2 immunohistochemical testing was
targeted fashion by using hybrid capture techniques.239 Amplicon-based established several years later when expert panels agreed on criteria
target enrichment for NGS can also be applied. However, similar to for determining HER2 status (Fig. 15.11).257
RT-PCR, a priori knowledge of the fusion partner is required.240,241 Tests for multiple immunohistochemical markers have been com-
A clever amplicon-based approach for NGS that does not require bined into panels that include ER, PR, HER2, and the proliferation
knowledge of the fusion partner is the anchored multiplex PCR marker Ki-67,258 in addition to several other markers thought to
technique.242 In this assay, an adapter sequence is ligated to the end be of prognostic importance.259 These panels have been found to
of cDNA fragments, and this adapter serves as a primer binding site, provide prognostic information that is comparable to that of multiplex
thus negating the necessity of having a primer specific to the fusion mRNA expression assays discussed later and have the added advan-
partner. Anchored multiplex PCR has been successfully applied to tage of accessibility in local laboratories and ready correlation with
clinical samples to detect gene fusions in a fusion partner–agnostic histologic features.
fashion.243 A substantial subset (~15%) of breast tumors do not express ER,
An alternative mRNA-based approach for fusion gene detection PR, or HER2 and are now referred to as triple-negative tumors.260
relies on quantification of expression of defined regions of a gene These lesions are notable for their aggressive histologic characteristics
(generally at least one region on either side of an expected breakpoint). and poor long-term outcome. This category overlaps but is not identical
This quantification can be achieved by several different methods. In to the basaloid-like breast cancer category defined by microarray-based
theory, if little to no wild-type expression of a gene is expected in the expression profiling261 and includes nonhereditary tumors that frequently
tissue type of interest, the region of the gene involved in the fusion express perturbations of BRCA1 pathway gene expression. The triple-
(i.e., 5′ or 3′) should be expressed at a far greater level than the rest negative category thus has increased the importance of accuracy
of the gene. Thus when measuring expression levels across the gene, in both positive and negative immunohistochemical testing and
Pathology, Biomarkers, and Molecular Diagnostics  •  CHAPTER 15 243

HER2 Immunohistochemical Scoring

1+ 2+ 3+

Figure 15.11  •  Scoring of Her2/neu immunohistochemical predictive marker in breast carcinoma is illustrated in this figure as follows: 1+, faint partial
membrane labeling (brown staining) of more than 10% of tumor cells; 2+, weak to moderate complete membrane labeling of more than 10% of tumor cells;
3+, strong complete membrane labeling of more than 10% of tumor cells.

reemphasizes the need for local validation and controls for accuracy screening test for ALK rearrangement in lung cancer. Positive
of immunohistochemical procedures. results should be confirmed by additional testing, including FISH or
molecular testing.
CDX2 expression in stage II colon carcinoma
The transcription factor CDX2 is a master regulator of intestinal ROS1 and other markers
development that is expressed in the cell nuclei of intestinal epithelium. Preliminary data indicate that immunohistochemical testing for other
A gene expression study has identified this gene as a prognostic marker markers including ROS1 can possibly substitute for FISH testing. In
in early-stage (II) colon adenocarcinoma.262 Widely used and well- a study266 in which lung tumors were tested with both FISH and
described immunohistochemical assays can classify colon tumors as IHC, IHC was 100% sensitive and 92% specific, suggesting that this
CDX2 negative or positive. Disease-free survival is worse for CDX2- infrequent rearrangement may be detected as efficiently with IHC as
negative than for CDX2-positive stage II colon tumors (49% versus by FISH. It seems likely that additional genetic lesions will be detectable
87%), and 5-year disease-free survival is better in patients with by aberrant expression, and this will be a convenient method to screen
CDX2-negative tumors treated with adjuvant chemotherapy than in for unusual lesions in the future.
those with untreated tumors (91% versus 56%).262 CDX2 status has
therefore been suggested as an indication for chemotherapeutic treat- Markers of Immune Response
ment in stage II colon tumors. Immunologic responses to tumor have long been observed and quanti-
fied in experimental systems,267 but immunotherapy as a viable clinical
EGFR expression in lung carcinoma option has only recently emerged with the demonstrated effectiveness
Finally, EGFR is expressed at high levels in most non–small cell lung of anti-CTLA4268 and anti-PD-1 therapy.269–273 A large and rapidly
carcinomas (NSCLCs), including both adenocarcinoma and squamous increasing number of immunologic agents are currently approved for
carcinoma, and expression correlates with gene amplification and copy cancer treatment (Table 15.6).
number. When a scoring system was applied in a phase III trial testing CTLA4 and PD1 are immunologic checkpoints that inhibit the
the effectiveness of the anti-EGFR chimeric (mouse/human) monoclonal host immune response. Suppression of inhibitory checkpoints by
antibody, cetuximab, in advanced NSCLC, only patients with the monoclonal antibodies to CTLA4 and PD-1 or its ligand PD-L1
highest levels of EGFR expression (scores >200) experienced a survival promote cytotoxic T-cell responses to antigens expressed by tumor
benefit.263 This result suggests that EGFR expression levels determined cells.274 These may consist of nonmutated antigens for which tolerance
with IHC may be a reasonable selection criterion for anti-EGFR is incomplete. More commonly, they are neoantigens that are the
antibody treatment. Properly scored EGFR immunohistochemical result of tumor cell mutations encoding novel epitopes that can elicit
expression may prove to be a valuable predictor of response to anti- a T-cell response.275
EGFR monoclonal antibody treatment. Because responses to anti-CTLA4 have been weaker and less durable
than responses to anti-PD-1 or anti-PD-L1 and because less than half
Aberrant Expression of Altered Genes of tumors typically respond to anticheckpoint treatment, efforts to
ALK develop biomarkers that predict response have mainly centered on
The availability of monoclonal antibodies to ALK fusion protein has PD-1 and PD-L1. The markers with greatest clinical potential are
resulted in the development of highly sensitive and specific immu- mutational burden, thought to reflect the level of expression of tumor-
nohistochemical assays for ALK rearrangement, one of which (ALK associated neoantigens; PD-L1 protein expression by tumor cells; and
[D5F3] CDx Assay; Ventana Medical Systems, Oro Valley, AZ) is now quantification of tumor-infiltrating lymphocytes (TILs). Following is
FDA approved as a stand-alone diagnostic for ALK rearrangement and a brief discussion of each of these markers as they relate to clinical
selection for anti-ALK therapy. Numerous studies have documented laboratory practice.
near 100% sensitivity and specificity of ALK immunohistochemical
tests (reviewed by Lantuejoul and colleagues264). Details of testing Mutational burden
procedures and interpretation of test results have been discussed in Deep sequencing of solid tumors has shown sharp differences in the
an International Association for the Study of Lung Cancer (IASLC) number of mutations occurring in specific tumors with histologic
monograph.265 Because of the economies and wide availability of characteristics and consequently variation in the number of predicted
immunohistochemical testing, ALK IHC is now a widely adopted neoantigens harbored by tumors of various histologic types.275 This
244 Part I: Science and Clinical Oncology

IHC is perhaps the most direct measure of expression but to date has
Table 15.6  FDA Approved Immunotherapeutic been a problematic predictor of response to treatment. Although there
Agents is a general association between immunohistochemical expression of
PD-L1 and response to anti-PD-1/PD-L1 antibody treatment,283–285
Molecular
patients with IHC-negative tumors may respond to treatment, and
Tumor(s) Target Drugs
published data indicate that the negative predictive value of IHC
Melanoma, NSCLC, SCCHN, PD1 Nivolumab for PD-L1 may be as low as 58% for nivolumab in melanoma.283
renal, Hodgkin Moreover, standardization and validation of immunohistochemical
Melanoma, NSCLC, SCCHN, PD1 Pembrolizumab tests has been difficult so that different antibodies with similar
urothelial tumors, reactivities286,287 but different cut points are used with each check-
microsatellite instability-high point inhibitor.288 Finally, the focal distribution of the PD-1 and
(MSI-H) or mismatch repair PD-L1 protein within tumors renders small biopsy and cytologic
deficient (dMMR) specimens of limited use for these immunohistochemical tests. These
NSCLC, urothelial tumors PD-L1 Atezolizumab problems have prevented the widespread adoption of immunohisto-
NSCLC PD-L1 Durvalumab chemical testing for prediction of response to checkpoint inhibition
Urothelial tumors PD-L1 Durvalumab and currently limit the use of these tests to research and clinical
trial settings.
Merkel tumor, urothelial PD-L1 Avelumab
tumors
Tumor-associated lymphocytes
Melanoma CTLA4 Ipilumumab Solid tumors are frequently associated with lymphocytic infiltrates
Melanoma CTLA4 Tremilumumab (TILs); it has been suggested that the level of TILs or TIL subsets
Melanoma CTLA4+ PD1 Ipilumumab+Nivolumab may correlate with outcome.289–291 In addition, attempts have been
made to relate response to immune checkpoint inhibition to response
NSCLC, Non–small cell lung cancer; SCCHN, squamous cell carcinoma of the head to treatment (reviewed by Gibney and colleagues283). At the present
and neck. time, however, no validated studies have produced reproducible cut
points that predict survival or response to immunologic or more
conventional treatment. Evaluation of TILs in solid tumors currently
observation has led to the hypothesis that antitumor responses could remains suitable only for research use.
be strongest in tumors with the greatest number of neoantigens.
Whole-exome sequencing has been found feasible in clinical tumors Examples of Tumor- and Organ-Specific
including lung cancer. In two high-mutation tumors, melanoma and Mutational Profiles
lung cancer, whole-exome sequencing has shown associations between
mutation burden of individual tumors and response and longer survival Although there is considerable overlap in mutational profiles among
with anti-CTLA4276 and anti-PD-1,277 respectively. These findings are tumors of variable tumor type, a consistency of mutations occurs
consistent with the suggestion that neoantigen formation may be a within individual tumor types. A summary of mutational profiles by
crucial determinant of antitumor response. Differences in outcomes, tumor type is provided here.
however, were not sufficient to regard mutational burden as a predictive
marker by itself. At the present time, mutational burden must be Gastrointestinal Stromal Tumors
considered developmental, and continued refinement of mutational One of the earliest successes of the mutation-targeted approach to
cut points will be required before whole-exome sequencing can be cancer treatment was the discovery of mutations in the KIT gene in
considered an actionable predictive test validated for clinical practice. gastrointestinal stromal tumor (GIST). GISTs were until recently a
rare and ill-defined neoplasm of somewhat mysterious origin. In the
Mismatch repair deficiency and microsatellite instability early 1990s these tumors were defined as spindle cell malignancies
A possible source of neoantigen formation is MMR deficiency. As that exhibited no evidence of cell of origin.292 The status of these
described earlier, loss of MMR capacity results in high frequency of tumors began to change as it was recognized in the mid-1990s that
mutation in affected tumors.278–280 The proposed explanation is that GISTs express CD34 but not desmin293 and thus displayed a distinct
mutation may lead to expression of altered protein, possibly stimulating immunophenotype. A watershed discovery was reported by Hirota and
an immune response to the neoantigen. MMR deficiency is reflected colleagues in 1998.294 These investigators described nearly uniform
in a high number of deletions and insertions in tandem repeat sequences, expression of KIT (CD117) in GISTs and suggested that they may arise
a finding referred to as microsatellite instability.72,73 As described earlier, from the immunophenotypically similar paraganglionic interstitial cells
MSI is measured by testing of five standard microsatellite markers. of Cajal. Most important, they identified deletions and point mutations
MMR as reflected by MSI is associated with a high objective response in the juxtamembrane portion of the KIT TKR gene. These mutations
rate (40%) and progression-free survival (PFS; 78%) in patients with were shown to activate KIT signaling, and KIT was thus a potential
colon cancer treated with PD-L1 inhibitor.281 Additional tumor types therapeutic target. KIT is a member of the TKR signaling family and
including carcinomas of endometrium, stomach, liver, ampulla of is encoded at chromosome 4q11-q12. In GISTs, mutations in the KIT
Vater, thyroid, skin (sebaceous tumors and melanoma), ovary, cervix, gene (reviewed by Corless and colleagues295; Lasota and Miettinen296;
esophagus, soft tissue, head and neck, kidney, and bone (Ewing sarcoma) and Martin-Broto and colleagues297) are located predominantly in
also have significant frequency of MMR deficiency and thus are sensitive the juxtamembrane domain (exon 11), with two-thirds of tumors
to PD-1 and PD-L1 therapy.282 MSI testing is an appropriate surrogate harboring mutations in this region of the gene. Exon 11 mutations
for neoantigen expression for a wide variety of tumor types being allow adenosine triphosphate (ATP) to phosphorylate receptor in
considered for PD-1 and PD-L1 therapy until and if direct assays for the absence of ligand (stem cell factor [SCF]). Phosphorylation of
tumor neoantigens are developed. the receptor triggers downstream signaling of RAF, MEK, MAPK,
PIK3CA, and STAT3, transforming mutated cells to malignant status.
PD-1 and PD-l1 expression detected Approximately 10% of mutations are found in exon 9, which encodes
with immunohistochemistry the extracellular domain of the molecule. These mutations induce a
Expression of PD-L1 would seem to be a reasonable biomarker for change in receptor conformation mimicking that seen in the presence
predicting response to inhibition of the PD-1/PD-L1 checkpoint. of ligand and resulting in receptor phosphorylation. Exon 9 mutations
Pathology, Biomarkers, and Molecular Diagnostics  •  CHAPTER 15 245

are commonly seen in large and small intestinal tumors but rarely stimuli from cell surface growth factor receptors. On activation, RAS
in gastric neoplasms. Finally, mutations in exons 13 (tyrosine kinase undergoes prenylation (addition of a 15-carbon chain) of a CAAX
domain) and 17 (activation loop) are occasionally found in GISTs. (C, cysteine; A, aliphatic amino acid; X, serine or methionine) motif
The effects of exon 13 and 17 mutations on the receptor are less well by a farnesyl transferase. This makes RAS more hydrophobic and
established, but the net result of all these mutations is receptor activation adherent to the inner aspect of the cytoplasmic membrane, where it
and oncogenic transformation of mutant cells. KIT mutations occur activates transducers in an intracellular signaling cascade involving
in approximately three-fourths of GISTs.295–297 numerous effector molecules including PI3K and MAPK.
Tumors that do not contain a KIT mutation may contain a mutation Consistent mutations in several components of the RAS signaling
in the closely homologous TKR gene platelet-derived growth factor pathway occur in human solid tumors. Mutations in KRAS are par-
receptor alpha polypeptide (PDGFRA).298,299 PDGFR protein has two ticularly common. They permit stimulus-independent activation of
isoforms, α and β, and it is the α form that is mutated. Approximately intracellular signaling. Hydrolysis of GTP bound to RAS must occur
10% of GISTs contain mutations in PDGFRA.295–297 The mutated for RAS to return to an inactive state. Typical mutations in codon
loci in PDGFRA are distributed similarly to those in KIT, but frequencies 12, which normally encodes glycine, result in substitution with an
differ. The most frequent mutation site (exon 18) encodes a portion amino acid that sterically interferes with GTP hydrolysis. This perpetu-
of the tyrosine kinase domain (TK2) and occurs in 6% of all GISTs. ates independent intracellular signaling and permanently stimulates
Juxtamembrane mutations in PDGFRA are relatively infrequent, proliferation and evasion of apoptosis. Because activation of the KRAS
occurring in less than 1% of GISTs. gene is stimulus independent, targeting of the upstream TKRs has
Introduction of TKIs that block signaling by mutant receptor has proven to be ineffective in countering the proliferative and invasive
had a profound effect on the outlook for this tumor. Before 2000, properties of tumors that are driven by mutation in KRAS.
the outlook for patients with GIST was bleak. In a majority of patients, Mutation in KRAS occurs in approximately 40% of colon carci-
disease recurred after surgical resection and the responses to chemo- nomas. The independent effect of mutation in KRAS on outcome in
therapy were poor. Only 10% of patients were disease free after extended colon cancer is still unclear and remains controversial.309 In a series
follow-up.300,301 There was a dramatic reversal of this situation when of 3439 patients with CRC, it was found that of the 12 possible
it was realized that GIST is exquisitely sensitive to tyrosine kinase mutations on codons 12 and 13, only the glycine-to-valine mutation
blockade. The drug successfully used to treat GIST was originally on codon 12 (8.6%) had a statistically significant impact on outcome.310
designed to block signaling by the BCR-ABL fusion protein in chronic Smaller series have shown similar results, but retrospective data from
myelogenous leukemia. Structural similarities between the intended other large randomized studies have failed to consistently demonstrate
target of the drug and KIT led to the discovery that the TKI now a meaningful effect of mutation in KRAS on outcome in CRC. In a
known as imatinib strongly inhibits mutated KIT.302 Subsequent clinical study of a large number of untreated patients, there was no difference
trials demonstrated high response rates and prolonged PFS,303–305 in PFS (7.3 weeks) between mutant and nonmutant tumors.311
leading to FDA approval of the drug as discussed elsewhere in this KRAS as a predictor of treatment response in colorectal carci-
book. KIT testing is now routinely employed to identify individuals noma.  In 2007 an article published in The New England Journal of
likely to respond to the drug. Medicine312 reported that administration of the monoclonal antibody
Mutations in PDGFRA are not equivalent to those in KIT. More cetuximab to patients with advanced CRC expressing immunohis-
than half of mutations in PDGFRA encode a D842V amino acid tochemically detectable EGFR had resulted in improved survival in
substitution that is resistant to imatinib, whereas other mutations in comparison to untreated controls. Both treatment and control arms
PDGFRA are sensitive.306 Alternative drugs that show activity against had been treated with a fluoropyrimidine, irinotecan, and oxaliplatin or
the D842V resistance mutation, such as midostaurin (PKC412),307 had contraindications to treatment with these drugs. The difference in
are under investigation. survival, although not large, suggested that this specific treatment could
be useful in managing CRC and prompted a closer look at responder
Colorectal Carcinoma versus nonresponders. Several subsequent studies have indicated that
Molecular markers now guide treatment for CRC, and comprehen- KRAS mutation status dictates response to anti-EGFR monoclonal
sive guidelines have been published providing recommendations antibody treatment in CRC. In the randomized studies summarized in
on molecular tests that are appropriate for this tumor type.308 Table 15.7,311,313–315 wild-type tumors treated with anti-EGFR antibody
The following discussion is intended to provide perspective on (cetuximab or panitumumab) have a PFS advantage of several months
molecular changes that may be important in the management and a significantly reduced hazard ratio in comparison with mutated
of CRC. tumors in first-line and third-line settings.
It has been suggested that not all mutations are equally predictive
KRAS as a prognostic marker in colorectal carcinoma of poor response to monoclonal anti-EGFR therapy. Patients with
KRAS is a member of a superfamily of guanosine-5-triphosphatase codon 13 mutation (p.G13D) appear to respond to such treatment
(GTPase) proteins that also includes NRAS and HRAS and a number with a significantly longer overall survival and a reduced hazard ratio
of less well-known proteins. The role of these proteins is to transduce in comparison with patients with other KRAS mutations.316

Table 15.7  KRAS Mutation as a Predictive Biomarker in Clinical Trials


KRAS
No. of KRAS Hazard Non-
Investigator Treatment Patients Mutationa Ratio mutateda HR Reference
Amado Panitumumab vs best supportive care (third line) 427 7.4 wk 0.99 12.3 wk 0.45 311
Karapetis Cetuximab versus best supportive care (third line) 572 1.8 mo 0.99 3.7 mo 0.40 313
Van Cutsem (CYRSTAL) FOLFIRI ± cetuximab (first line) 540 7.6 mo 1.07 9.9 mo 0.68 314
Bokemeyer (OPUS) FOLFOX ± cetuximab (first line) 233 5.5 mo 1.83 7.7 mo 0.57 315

a
Progression-free survival.
246 Part I: Science and Clinical Oncology

BRAF mutation and prognosis Mutational profiles of NSCLC separate according to histologic
A second mutation in colon carcinoma of clinical importance is BRAF. type. TKR mutations (discussed later) are far more commonly found
BRAF mutations were first identified in colon cancer through a survey in adenocarcinoma than in squamous carcinoma. Many of the mutations
of BRAF mutations in all human tumors.317 The reported frequency in adenocarcinoma involve tyrosine kinase receptors and their signal
of BRAF mutation in clinical samples ranges from approximately transducers. The first of these receptors to be defined as a therapeutic
5% to 15%.318–321 BRAF mutation is more frequent in tumors on target was epidermal growth factor receptor (EGFR).
the right side of the colon.322–324 Microsatellite instability (MSI-H)
occurs in approximately half of BRAF-mutated tumors.321,324,325 Single-Nucleotide Variants, Small Deletions and Insertions
BRAF mutation is associated with aggressive tumor behavior and Epidermal growth factor receptor
shortened survival, but only in tumors that are MSI stable321,324,326,327; The original isolation of epidermal growth factor (EGF) from mouse
mutation in MSI-H tumors carries no survival disadvantage. MSI in salivary gland in 1962,336 the demonstration of its binding to cell
BRAF-positive tumors is attributed to methylation of MMR rather membrane receptor,337 and sequencing of the receptor338 established
than mutation and appears to be unrelated to HNPCC and germline the role of intercellular signaling in epithelial cell growth and main-
mutation of MMR.328,329 BRAF mutation in colon tumors confers tenance. By the 1990s, overexpression of EGFR had been demonstrated
resistance to anti-EGFR monoclonal antibody treatments,320,330–332 and for squamous carcinoma. Monoclonal antibodies to EGFR that were
BRAF-mutated tumors respond poorly to BRAF inhibitors. Resistance effective in localizing EGFR in situ became available in the late 1990s,
to BRAF inhibitors is thought to be due to activation of EGFR in documenting the broad expression of EGFR in adenocarcinoma.339
response to BRAF inhibition.333 It has been suggested that BRAF- EGFR expression in NSCLC was found to be closely linked to gene
positive tumors may therefore respond to dual inhibition of both EGFR copy number.340
and BRAF. Further studies of EGFR were driven by of the discovery of a
subset of patients who were mostly nonsmokers with adenocarcinoma
Other Mutations who responded dramatically to EGFR TKIs (Fig. 15.12). Simultane-
Additional mutations of predictive importance are NRAS and PIK3CA, ously, two groups reported that mutations in the EGFR TK domain
which occur at frequencies of less than 5%318,320,334 and 10% to were present in a subset of adenocarcinomas and that their presence
15%,320,332,334 respectively, in colon carcinoma. PIK3CA mutations correlated with sensitivity to TK inhibition.341,342 The mutations most
overlap those found in other genes, but NRAS mutations are nonoverlap- commonly consisted of either single base substitution in exon 21
ping with mutations in other genes including KRAS and BRAF. (c.2573G>C, p.L858R) or in-frame deletion within exon 19 ranging
Mutations in either the PIK3CA or the NRAS gene are associated from 9 to 18 base pairs in the region coding for the ATP-binding
with poor response to EGFR monoclonal antibody therapy.320,332 Finally, pocket of the tyrosine kinase domain. These resulted in strong activation
initial results of comprehensive molecular analysis by TCGA have of the mutant receptor.
been reported and identify large numbers of mutations that are likely Activating mutations of EGFR result in tumor susceptibility to
to become targets of specific molecular inhibitors.334 Comprehensive TKI therapy.343 However, responses to therapy are temporary, and
molecular testing is expected to contribute greatly to colon carcinoma hopes of a long-lasting therapy are generally unrealized owing to
management in the near future. resistance to targeted therapy that inevitably arises in treated patients.
Two common mechanisms of resistance to EGFR TKI are MET
Lung Carcinoma—A Heterogeneous Tumor amplification344–346 and a point mutation in exon 20 of EGFR (c.2369
Since the 1920s, lung tumors have been divided into small cell (“oat C>T, p.T790M),347 the effect of which is depicted in Fig. 15.13. As
cell”) lung carcinoma (SCLC) and non-small cell lung carcinoma a result of the point mutation in EGFR exon 20, a bulky methionine
(NSCLC), which differ not only morphologically but by molecular side chain is thought to sterically hinder binding of TKIs. Tumors
profile as well. SCLCs express a number of markers that are also found cells harboring this mutation may exist in low numbers in untreated
on neural cells and, in current classifications, are categorized as one tumors and emerge through selective pressure exerted by targeted
of several different types of “neuroendocrine” carcinomas. SCLC is therapy. In addition, insertions in exon 20 of EGFR are associated
uniformly aggressive and particularly sensitive to mitotic inhibitors. with primary resistance to TKI therapy.348 This also is thought to be
Finally, this tumor type is highly genetically unstable and appears to due to steric hindrance of TKIs. Finally, other mechanisms of resistance
be driven predominantly by seven transmembrane receptors and has including activation of alternative signaling pathways and epithelial-
not been successfully treated with targeted agents. mesenchymal transition or small cell transformation may also be causes
Most NSCLCs do not express neuroendocrine markers but instead of resistance. The mechanism of resistance in individual cases can be
strongly express cell surface TKR. Further, NSCLC is divided into ascertained by349,350 cellular or liquid biopsy of recurrent tumor and
(1) adenocarcinomas that are composed of cuboid or columnar cells is discussed elsewhere in this book.
that often form acinus-like or papillary structures or grow along
pulmonary alveolar surfaces (as illustrated in the discussion of lung KRAS
carcinogenesis earlier), and (2) squamous tumors that form defining KRAS mutations are more common in lung cancer than EGFR
microscopic structures such as keratin pearls and intercellular bridges. mutations and, unlike EGFR mutations, occur predominantly in
These tumor types are also distinguished by variation in expression smokers.351,352 Coexistence of KRAS and EGFR mutation is uncom-
of specific keratin subgroups and other lineage specific markers, most mon but has been reported.353 Because of its position downstream
notably TTF1, which is expressed by adenocarcinoma but not squamous of EGFR signaling, the hypothesis that an activating mutation in
carcinoma. KRAS would render a tumor nonsusceptible to TKI therapy in
The phenotypic heterogeneity of lung carcinoma is reflected in lung tumors has been extensively investigated, with conflicting
the recent revision of the anatomical classification of lung carcinoma.335 results.354
Genomic changes are also highly heterogeneous. Lung cancer is one
of the most highly mutated of tumors198 containing large numbers Large-Scale Rearrangements
of SNVs, chromosomal arrangements, and copy number abnormalities. Large-scale chromosomal rearrangements including translocations,
Lung cancers exhibit a high degree of intratumoral heterogeneity and inversions, duplications, and amplifications are common in solid tumors
frequently contain subclones of mutant cells207,208a that may undergo and are reflected in global methods for chromosomal imaging such
selection in response to treatment or other microenvironmental changes. as chromosome painting or spectral imaging. These rearrangements
Mutation and clonal selection continue during the life of the tumor may create new fusion genes or activate oncogenes, becoming drivers
and may affect response to treatment and overall patient survival.208a of cell proliferation and oncogenesis. Rearrangements that fuse gene
Pathology, Biomarkers, and Molecular Diagnostics  •  CHAPTER 15 247

Positions of Mutations Detected in EGFR Tyrosine Kinase Domain in NSCLC~


EGF ligand binding Tyrosine kinase Autophosphorylation

TM K DFG Y Y Y Y

718 745 776 835 858 861 869 964

GXGXXG K R H DFG L L Y
Exon: 18 19 20 21 22 23 24

719 747-750 858

Paez:

Lynch:

Pao:

Tumor with point mutation (amino acid substitution)


Tumor with in-frame deletion

EGFR
747-750

G719

L858

Activation
loop

B
Figure 15.12  •  (A) Schematic view of epidermal growth factor receptor (EGFR) and its key domains including the extracellular ligand-binding domain,
the transmembrane domain, the tyrosine kinase domain, and the autophosphorylation domain. The tyrosine kinase domain (exons 18 through 24) is expanded,
showing the sites of described point mutations at G719, S752, R776, H835, L858, and L861, and in-frame deletions at L747-A750. (B) Three-dimensional
ribbon structure of the intracellular domain: the positions of the three most common mutations are shown in relation to the activation loop. EGF, Epidermal
growth factor; NSCLC, non–small cell lung carcinoma; TM, transmembrane.

promoters to the TK domain of growth factors, growth factor receptors, fibroblasts.355 Further investigation demonstrated the transcript to be
or transcription factors may result in inappropriate signaling from fusion of a gene called echinoderm microtubule-associated protein-like
the fusion products. 4 (EML4) with the anaplastic lymphoma kinase (ALK) gene (see
Fig. 15.10), which is also rearranged in large cell lymphomas.
ALK This rearrangement has been detected in 4% to 7% of pulmonary
Examples of activation by gene fusion in lung cancer involve ALK, adenocarcinomas, with an increased proportion seen when cohorts are
ROS1, and RET. In 2007, Soda and colleagues discovered a transcript selected based on clinicopathologic features including lack of smoking
derived from a patient with NSCLC that could transform T3T history.356–359 The driver of tumorigenesis in this rearrangement is
248 Part I: Science and Clinical Oncology

Tyrosine Kinase Scenarios


Ligand
Extracellular

Physiological Mutation Mutation


Inactive activation Mutation TKI inhibition TKI resistance
TM

TKI
ATP PP ATP PP
P P
Tyrosine kinase

ATP PP ATP PP TKI ATP PP

Signal P Signal P Signal P


proteins proteins proteins

Signal Signal
proteins proteins

Proliferation
Proliferation Motility
Motility Cell survival
Cell survival Invasion
Metastasis

Figure 15.13  •  Tyrosine kinase receptors of many types including KIT, EGFR, HER2/neu, HGHR (MET), IGFR1, ROS1, RET, and PDGFRA, have
similar molecular structures comprising extracellular, transmembrane (TM), and tyrosine kinase domains. Physiologic activation of receptor by binding of
ligand induces conformational changes in the intracellular tyrosine kinase domain that permits binding of adenosine triphosphate (ATP) and phosphorylation
of signaling proteins, which ultimately may lead to cell proliferation, increased motility, and cell survival. Activating mutations (zipper line) affect the ATP
binding pocket of the tyrosine kinase domain, causing phosphorylation of signaling proteins in the absence of ligand binding. Receptor mutations may
ultimately lead to uncontrolled cell proliferation and tumor formation. Treatment of mutated tumors with tyrosine kinase small molecular inhibitors (TKI)
blocks ATP binding and phosphorylation of signaling proteins. Resistance to TKI may be caused by additional mutation (double zipper line) affecting the
tyrosine kinase domain. Cells carrying the resistance mutations may exist in low numbers in untreated tumors but selectively proliferate in the presence of
inhibitor.

thought to be ALK, which is not typically expressed in adult lung


tissue. Juxtaposition of a 5′ partner with a promoter results in high-level Summary
expression of the kinase domain of ALK and leads to inappropriate Mutations in EGFR, KRAS and ALK underscore the diversity of
signaling by this molecule.355,360 molecular alterations in NSCLC, and with ongoing extensive efforts,
Less than 5 years after the initial discovery of this rearrangement, “driver mutations” have been identified in a significant proportion of
over 10,000 lung cancers have been tested for its presence. More than 13 lung cancers. Molecular testing can provide guidance for prioritizing
molecular variants of EML4-ALK fusion have been detected,357,361 and therapies to those patients who are likely to receive the greatest benefit,
three other rare activating gene partners have been identified: TGF,221 and conversely can identify patients proven to benefit from a targeted
KIF5B,222 and KLC1.223 A novel targeted therapy agent—crizotinib—has therapy. Finally, molecular testing can provide clinicians with a rationale
been found to be effective against tumors driven by this molecular to guide certain patients toward clinical trials of targeted therapeutics.
marker228 and has received approval for treatment of these specific These selected examples of targets are by no means the only ones
tumors by the FDA. of interest in NSCLC, in which a growing number of molecularly
However, as is the case with EGFR, acquired resistance to treatment targeted agents with companion molecular alterations are available and
inevitably occurs in initially sensitive tumor (reviewed by Lin and being developed. These include mutations in BRAF,363,364 HER2,365
colleagues362). The most important resistance mechanism is mutation NTRK,366 and NRAS,367 and rearrangements in RET368 and ROS1,369
in the ALK tyrosine kinase domain, followed by amplification of the among others.370
ALK gene, activation of bypass signaling pathways, change in tumor A graphical summary of the relative frequencies of mutations in
phenotype to, for example epithelial-mesenchymal transition or small an adenocarcinoma population enrolled in a recent Lung Cancer
cell phenotype, and overexpression for the drug efflux pump protein, Mutation Consortium study is illustrated in Fig. 15.14. Preliminary
p-glycoprotein. Analysis of specimens from cellular or liquid biopsy data from this study, which tested an expanded set of predictive markers
of recurrent tumor can identify which of these mechanisms is responsible by NGS, suggest improved outcome for those tumors in which a
for resistance, and treatment strategies can be adjusted accordingly. targetable driver mutation is found.371 Comprehensive guidelines for
Detailed discussion of strategies for management of ALK resistance molecular testing in lung cancer and its clinical ramifications have
is found elsewhere in this book. recently been published.372
Pathology, Biomarkers, and Molecular Diagnostics  •  CHAPTER 15 249

Mutaon Frequency
Lung Cancer Mutaon Consorum II
Adenocarcinoma veBRAF
MET 2%
oEGFR
2% amp
3%
ALKr
sEGFR 4%
10% oBRAF
1%
RETr
doubletons 2%
2%
KRAS
25% ROS1r ERBB2
2% 1%

PIK3CA [CATEGORY
1% NAME]
<1%

[CATEGORY NAME]
<1%

Unknown
44%

Figure 15.14  •  Pie chart showing relative frequencies (N = 875) of specific mutations in a Lung Cancer Mutation Consortium adenocarcinoma cohort
tested by next-generation sequencing (NGS) and fluorescence in situ hybridization (FISH). Prefix and suffix letters are defined as follows: s, sensitizing; o,
other; ve, V600E; r, rearrangement. (Prepared from data presented at American Society of Clinical Oncology [ASCO] annual meeting 2016.371)

studies reinforce the need for accurate molecular characterization of


Melanoma: Targeted Treatment of an Aggressive Tumor melanoma to allow for selection of effective treatment.
Melanoma is also a highly mutated tumor and several targetable
mutations have been found in this tumor in recent years involving KIT
the BRAF, NRAS, and KIT genes (see Table 15.3). Of these, the most A second mutation target in melanoma is KIT (reviewed by Woodman
common is BRAF.373 and colleagues373 and Flaherty and Fisher382; discussed earlier with regard
to GIST). KIT mutations differentially affect subsets of melanoma.
BRAF Mutation frequencies in sun-damaged cutaneous, acral, and mucosal
BRAF protein is a serine/threonine kinase that is encoded on chromo- melanomas range from 2% to 20%, but KIT mutations are absent
some 7q34. It is an important signal transduction molecule mediating from cutaneous tumors arising in non–sun-damaged skin.373,383 In
signals from RAS to MEK, ultimately promoting cell proliferation and addition, a different spectrum of mutations affects melanoma than
mobility. Activating missense mutations in BRAF were first identified GIST.383–385 Whereas KIT mutations in GIST are often short deletions
in a screening of over 500 cell lines from multiple organ sites.317 or insertions in exon 11, the vast majority of mutations in melanoma
Mutations were found in 60% of melanoma lines and tumors at reported to date are point mutations. Mutations in exon 9, which
lower frequencies in carcinomas of colon (15%), lung (3%), breast constitute over 10% of GIST mutations, have not been found in
(3%), and ovary (4%) and in glioma (11%). BRAF mutation is now melanoma. Mutations in exon 13, which encoded the kinase domain
thought to occur in approximately half of primary and metastatic of KIT, are more common in melanoma than in GIST. Finally, KIT
melanoma clinical samples,374–377 with some variation according to gene amplification is frequent in melanoma383–385 but is not described
melanoma subtype. Activating mutations in BRAF largely occur in in GIST.295
exon 15, which encodes the catalytic domain of BRAF. By far the most Therapeutic trials testing the effectiveness of KIT inhibitors on
frequent single base change (c.1799T>A) results in an amino acid KIT-mutated tumors have had limited success. In a phase II trial
change, V600E.317,377 This mutation activates continuous intracellular enrolling 51 metastatic melanoma patients with genetic alterations of
signaling, resulting in cell proliferation, increased tumor cell survival, KIT, durable complete responses to imatinib have been reported in
and enhanced mobility. Two compounds selectively block the kinase two patients with mutations in exon 11 and partial durable responses
activity of mutated BRAF, vemurafenib and dabrafenib. In clinical trials, in four others, for an overall response rate of 16%.385 Another study
both compounds have produced high response rates and improved has reported similar findings (23% overall partial response rate) in
PFS or overall survival in tumors with BRAF mutations.378–381 These tumors with exon 11 and 13 mutations.386 Imatinib blockade of KIT
250 Part I: Science and Clinical Oncology

may be a life-extending alternative for those patients with BRAF differentiation and maintains glial cells in stem cell–like state, prone
wild-type/KIT-mutated metastatic tumors of specific subtype. This to self-renewal and tumorigenesis.399
new clinically relevant development will add urgency to the implementa- IDH1/IDH2 mutations defines new categories of diffuse low-grade
tion of multiplex testing at many institutions that are equipped to glioma as listed in Table 15.8. Today, diffuse gliomas in adults are
perform molecular testing. classified by IDH1/2 mutational status as WHO grade II (diffuse
astrocytoma), III (anaplastic astrocytoma), or IV ([glioblastoma
NRAS multiforme (GBM)]). Across all grades, IDH-mutant tumors display
NRAS, encoded on chromosome 1, is a member of the RAS family a more favorable prognosis than their like-graded IDH–wild-type
of signaling molecules and was originally identified as an oncogene counterparts.400–402 IDH–wild-type grade III tumors are less frequent
in neuroblastoma cells.387,388 Mutation in melanoma was reported than IDH-mutant grade III tumors, but the converse is true in
shortly after discovery of the gene.389 NRAS is altered in approximately glioblastoma (6%–13% mutant403–407). In general, histologic and
20% to 30% of melanomas of all types,373,377 except for uveal mela- cytologic features do not distinguish IDH-mutant from IDH–wild-type
noma. Mutations are restricted mainly to exon 3, codon 61 (>80%), gliomas at any grade. Thus assessment of mutational status is key for
with a smaller proportion occurring at exon 2, codon 13.377 These classification and prognostication. The large majority (>88%) of IDH
mutations are thought to lock NRAS in its activated state. However, mutations in gliomas are substitutions at codon 132 of histidine for
mutation in NRAS by itself may not be sufficient for malignant arginine (R132H). A high-fidelity antibody identifying this mutant
transformation, because both sporadic and congenital nevi of several epitope is available,408 obviating need to perform actual mutational
types have been shown to harbor NRAS mutation without evidence testing if the immunohistochemical test result is positive. Less common
of malignant transformation.390,391 To date, no drugs are known to IDH1 mutation or any IDH2 mutation (the latter more often seen
directly and effectively target NRAS signaling. However, downstream in oligodendroglial lineage tumors) confers the same favorable prognosis
signal transduction has been the subject of ongoing preclinical as the canonic mutation.
investigation373,382
1p/19q codeletion
GNAQ/GNA11/BAP1 Codeletion of recurrent regions in chromosomes 1p and 19q (1p/19q)
Mutations in GNAQ, GNA11, and BAP1 are confined largely to uveal was initially described over two decades ago and occurs in 50% to
melanomas, blue nevi, and meningeal melanoma. The GNAQ and 70% of both low- and high-grade oligodendrogliomas.409–412 Its presence
GNA11 genes encode membrane-bound GTPases that regulate signaling has been found to strongly correlate with biologic behavior and
through seven transmembrane receptors. Activating mutations in either prognosis in these tumors.413–415
GNAQ or GNA11 are reported in over 80% of uveal melanomas and
blue nevi392,393 at codon Q209 and R183 hot spots. BRCA1-associated Histone mutations
protein 1 (BAP1) is encoded on chromosome 3p21.1 and is mutated Histone mutations occur in many tumor types, but in glioma, specific
in over 80% of metastatic uveal melanomas.394 BAP1 mutation has histone mutations are closely associated with morphologic features
also recently been described in a familial syndrome with develop- and biologic behavior and are now incorporated into the WHO glioma
ment of clinically and histologically characteristic tumors with some classification. Histones are protein octamers that organize DNA replica-
features of melanoma. This mutation also occurs in sporadic tumors tion and gene regulation (reviewed by Lee and colleagues416). Mutation
with similar features.395 BAP1 mutations cause protein truncation can have the effect of disrupting the many functions of the protein,
and therefore inactivation of the protein. For this reason, BAP1 is including DNA replication, chromosome condensation, mitosis, DNA
thought to be a TSG. BAP1 mutations are of limited prognostic repair, telomere and centromere maintenance, and gene expression.
value and are not yet therapeutic targets but illustrate the highly The mutation H3.3 K27M occurs in 70% to 80% of high-grade
specific relationships that may exist between morphology and molecular midline pediatric tumors417,418 and is now designated as diffuse midline
pathology. glioma, H3 K27M mutant in the 2016 edition of the WHO classifica-
tion of gliomas. Other histone mutations along with their phenotypic
Central Nervous System: Classification by expression are listed in Table 15.5.
Molecular Diagnostics ATRX
Tumors of the CNS are a heterogeneous group that range from slow- ATRX (α-thalassemia/mental retardation syndrome–X linked) is a
growing low-grade gliomas to rapidly growing and often rapidly fatal transcriptional regulator that is expressed in the nucleus of normal
glioblastoma (glioblastoma multiforme, GBM). Both low-grade396 and cells. Mutation, most often frameshift deletion,396,419,420 results in
high-grade397 tumors have been comprehensively tested through the loss of expression of the encoded protein that can be demonstrated
Cancer Genome Atlas program, and driver mutations are numerous by immunohistochemical methods. ATRX mutation occurs in both
in these tumors. To a remarkable extent, molecular profiles of tumors adult diffuse gliomas and glioblastoma associated with mutation
of the brain correlate with or supplement histologic subclassification, with IDH1/IDH2 and TP53 and G-CIMP, suggesting a role in
so molecular findings in addition to histologic appearances are used chromosomal instability. That the combination of mutations occurs
in the definition of diagnostic categories in the most recent World in both diffuse gliomas and glioblastomas suggests that the latter
Health Organization (WHO) classification of CNS tumors.398 Brain may be the result of mutational progression of preexisting diffuse
tumors have moved further toward a molecular classification than lymphoma.
any of the other solid tumors. Mutations of a single dominant gene
are accompanied by consistent associated genotypic changes, so that TERT promoter
a constellation of genomic changes encode consistent histologic and TERT (telomerase reverse transcriptase) promoter mutations are frequent
biologic phenotypes. in gliomas of many types and have the predicted effect of upregulating
telomerase expression and thus preventing postmitotic apoptosis.
IDH1/IDH2 Mutations of TERT are often associated with a constellation of muta-
IDH1IDH2 encode two isoforms of isoicitrate hydrogenase. The enzyme tions in other genes that together form a gene profile that is expressed
metabolizes isocitrate to α-ketoglutarate (α-KG) in the citric acid as a particular glioma type. For example, TERT promoter mutation
cycle. Mutation of IDH1/IDH2 ablates normal enzymatic function may combine with IDH1/IDH2 mutation and 1p19q codeletion to
and shifts the metabolic product to 2-hydroxyglutarate (2-HG). 2-HG generate phenotypes oligodendroglioma and anaplastic oligodendro-
induces widespread hypermethylation (G-CIMP), which inhibits cellular glioma, as shown in Table 15.8.
Pathology, Biomarkers, and Molecular Diagnostics  •  CHAPTER 15 251

Table 15.8 Genetic Mutations, Histology, and World Health Organization (WHO) Classification

Primary Age Histology


Mutation Associated Genotypes Anatomic Location Group (Phenotype) WHO Classification
IDH1/2 mutation TP53, ATRX mutation Cerebral hemispheres Adult Diffuse Diffuse astrocytoma, IDH
astrocytoma mutant
TP53, ATRX mutation Cerebral hemispheres Pediatric Anaplastic Anaplastic astrocytoma, IDH
or adult astrocytoma mutant
1p/19q codeletion, TERT, CIC, Cerebral hemispheres Adult Oligodendroglioma Oligodendroglioma, IDA
FUBP1 mutations mutant and 1p/19q codeleted
1p/19q codeletion, TERT, CIC, Cerebral hemispheres Adult Anaplastic Anaplastic oligodendroglioma,
FUBP1, TCF12 mutation; oligodendroglioma IDH mutant and 1p/19q
CDKN2A deletion codeleted
TP53, ATRX mutation CDKN2A Cerebral hemispheres Adult Glioblastoma Glioblastoma, IDH mutant
homozygous deletion
IDH1/2 wild-type TERT, PTEN, TP53, PIK3CA, Cerebral hemispheres Adult Glioblastoma Glioblastoma, IDH wild-type
PIK3R1, NF1, H3F3A-G34
mutation; CDKN2A, PTEN
homozygous deletion; EGFR,
PDGFRA, MET, CDK4, CDK6,
MDM2, MDM4 amplification;
EGFRvIII rearrangement
H3-K27M mutant TP53, PPMD1, ACVR1, FGFR1 Midline structures, Pediatric Diffuse midline Diffuse midline glioma,
mutations, PDGFRA, MYC, thalamus, brainstem and glioma H3-K62M-mutant
MYCN, CDK4, CDK6, CCND1-3, spinal cord
ID2, MET amplifications
BRAF mutation or RAF1, NTRK2 gene fusions; Cerebral hemispheres Adult Pilocytic Pilocytic astrocytoma
rearrangement NF1, KRAS, FGFR1, PTPN11 astrocytoma
mutation
CDKN2A/p14ARF homozygous Cerebral hemispheres Pediatric Pleomorphic Pleomorphic
deletion xanthoastrocytoma xanthoastrocytoma
MYB or MYBL FGFR1 duplication Cerebral hemispheres Pediatric Well-differentiated Well-differentiated pediatric
rearrangement pediatric diffuse diffuse glioma
glioma

BRAF include TP53, PTEN, PIK3CA, PIK3R1, NF1, and H3F3A-G34


BRAF is a tyrosine kinase pathway gene that is mutated in a limited mutations; CDKN2A and PTEN homozygous deletions; FUBP1
number of brain tumor types including pleomorphic xanthoastrocytoma, and TCF12 mutations; EGFR, PDGFRA, MET, CDK4, CDK6,
ganglioglioma, extracerebellar pilocytic astrocytoma,421 and epithelioid MDM2, and MDM4 amplifications; EGFRvIII rearrangement; and
glioblastoma.422 BRAF mutations may consist of either point muta- PPMD1, ACVR1, FGFR1, PDGFRA, MYC, MYCN, CDK4, CDK6,
tion, typically V600E, or chromosomal rearrangement resulting in CCND1-3, ID2, and MET amplifications.
the fusion gene KIAA1549–BRAF. Both mutations result in activation
of the gene and tumorigenesis, and both have been treated with TK REGULATORY CONSIDERATIONS
inhibitors.423–428
The wide scope of approaches for biomarker evaluation makes for a
C11orf95-RELA fusions challenging regulatory landscape. Within the United States and around
Ependymoma (reviewed by Wu and colleagues429) represents 1.9% of the world, there have been varying approaches to consideration of
adult and 5.2% of pediatric brain tumors; most adult tumors are biomarker testing and the types of oversight that should be applied.
supratentorial, whereas most pediatric tumors occur in the posterior For the purposes of this text, the regulatory considerations in the
fossa and spinal cord.430 C11orf95-RELA fusions in a region of United States are the focus; however, the framework employed in
chromothripsis on chromosome 11 are present in 70% of supratentorial the United States sometimes forms the underpinnings of systems
ependymomas.431 The RELA component of the fusion is the principal elsewhere.
effector of canonic nuclear factor–κB (NF-κB) signaling and is a Testing performed in the United States for the purposes of clinical
potential treatment target, although no targeted drug has yet passed decision making is mandated to be compliant with the Clinical Labora-
clinical trials. YAP1-MamlD1 fusions occur in those supratentorial tory Improvement Amendments (CLIA) of 1988, and accordingly
tumors that lack the C11orf95-RELA fusion431,432 and are associated there is a Clinical Lab Center within the Centers for Medicare and
with a better prognosis. Medicaid Services (CMS) that maintains the specific regulations (https://
www.cms.gov/Center/Provider-Type/Clinical-Labs-Center.html). CLIA
Other mutations regulations stipulate “quality standards for proficiency testing (PT),
Several other mutations shown in Table 15.8 are found in gliomas but patient test management, quality control, personnel qualifications and
do not uniquely define a tumor type and may be discussed elsewhere quality assurance for laboratories performing moderate and/or high
in this book. Although some of these genes may be therapeutic complexity tests” (https://www.cms.gov/Regulations-and-Guidance/
targets, owing to space limitations they are only mentioned here and Legislation/CLIA/Program_Descriptions_Projects.html).
252 Part I: Science and Clinical Oncology

Almost all laboratories must obtain a CLIA certificate. Many class III. Examples of class III IVDs receiving FDA approval include
laboratories receive accreditation through their state agency or through BrAF V600, BRCA, and NGS oncological panel (see devices with
one of seven organizations with deeming authority (https://www codes PQP, NKF, MVC, and PHP at https://www.accessdata.fda.gov/
.cms.gov/Regulations-and-Guidance/Legislation/CLIA/Accreditation scripts/cdrh/cfdocs/cfPMA/pma.cfm for examples).
_Organizations_and_Exempt_States.html). In some circumstances (e.g., Although high-risk IVDs with FDA approval are available for
in New York and Washington states), laboratories are exempt from purchase, the burdensome regulatory framework limits their availability
CLIA requirements based on equivalent or more stringent requirements to assays that are intended for widespread distribution. In contrast,
at the state level. a substantial proportion of biomarker assays performed for clinical
The FDA regulates companies that sell in-home and laboratory decision making are not FDA approved and instead are developed by
diagnostic tests (in vitro diagnostic devices [IVDs]) to clinical labo- individual laboratories as laboratory developed tests (LDTs). The FDA
ratories. The FDA cites the authority of the Medical Device Amend- defines LDTs as in vitro diagnostic test(s) that are manufactured by
ments of 1976 for this oversight. Although IVDs are not specifically and used within a single laboratory. In November 2016, the FDA
mentioned in the legislation, the FDA considers IVDs to be medical announced it would not immediately finalize its proposed regulatory
devices. Per the 1976 law, medical devices are required to be classified framework for LDTs and issued a subsequent discussion paper sur-
into three categories—low risk (class I), moderate risk (class II), and rounding the issue.
high risk (class III)—according to the level of regulatory control needed The issues surrounding potential regulatory approaches for LDTs
to ensure safety and effectiveness. Low-risk IVDs need only be listed remain controversial, both with various stakeholders and politically.
with the FDA. Moderate-risk IVDs must be submitted for premarket Those in favor of increased regulatory requirements for LDTs argue
clearance following 510(k) regulations. High-risk IVDs must receive that lack of oversight makes for potentially inaccurate testing with
Premarket Approval (PMA) from the FDA. the potential for patient harm, whereas those who oppose such oversight
In 1992 the FDA stated publicly that its authority extended to point out that the regulatory framework suggested will significantly
clinical laboratories using laboratory developed tests (LDTs), but that hamper patient access to innovative testing and stifle iterative improve-
it chose to exercise “enforcement discretion” by not requiring clinical ments of existing LDTs. As of the writing of this chapter, no resolution
laboratories to submit premarket applications. In 2010 the FDA has been identified, although some legislative solutions are being
announced its intent to regulate LDTs, and in 2014 it published a considered. As the technology driving biomarker analysis continues
draft guidance document for comment. As of 2017, no LDT guidance to evolve at a tremendous pace, solutions that account for the pace
or regulation had been finalized. of progress will certainly be paramount.
Biomarker assays use a wide variety of techniques, including IHC,
molecular pathologic methods (e.g., PCR, NGS), and proteomics, The complete reference list is available online at
among others. The FDA would likely classify most of these tests as ExpertConsult.com.

KEY REFERENCES
15. zur Hausen H. Papillomaviruses causing cancer: 122. Kim MS, Lee J, Sidransky D. DNA methylation 198. Kandoth C, McLellan MD, Vandin F, et al. Muta-
evasion from host-cell control in early events in car- markers in colorectal cancer. Cancer Metastasis Rev. tional landscape and significance across 12 major
cinogenesis. J Natl Cancer Inst. 2000;92(9):690–698. 2010;29(1):181–206. cancer types. Nature. 2013;502(7471):333–339.
16. zur Hausen H. Papillomaviruses and cancer: from basic 131. Aberle DR, Adams AM, Berg CD, et al. Reduced 202. Comprehensive molecular portraits of human breast
studies to clinical application. Nat Rev Cancer. 2002; lung-cancer mortality with low-dose computed tumours. Nature. 2012;490(7418):61–70.
2(5):342–350. tomographic screening. N Engl J Med. 2011;365(5): 204. Burrell RA, McGranahan N, Bartek J, Swanton C.
29. Huh WK, Ault KA, Chelmow D, et al. Use of 395–409. The causes and consequences of genetic heterogene-
primary high-risk human papillomavirus testing for 134. Franklin WA, Muller KM, Wistuba I, et al. Squa- ity in cancer evolution. Nature. 2013;501(7467):
cervical cancer screening: interim clinical guidance. mous dysplasia and carcinoma in situ. In: Travis WD, 338–345.
J Low Genit Tract Dis. 2015;19(2):91–96. Brambilla E, Muller-Hermelink HK, Harris CC, eds. 210. Goodwin S, McPherson JD, McCombie WR. Coming
32. Stanley M. Prospects for new human papillomavirus Tumours of the Lung, Pleura, Thymus and Heart. of age: ten years of next-generation sequencing
vaccines. Curr Opin Infect Dis. 2010;23(1):70–75. 2nd ed. Lyon, France: IARC Press; 2004:68–72. technologies. Nat Rev Genet. 2016;17(6):333–351.
33. Villa LL. HPV prophylactic vaccination: The first 140. Keith RL, Blatchford PJ, Kittelson J, et al. Oral 225. Camidge DR, Kono SA, Flacco A, et al. Optimizing
years and what to expect from now. Cancer Lett. 2011; iloprost improves endobronchial dysplasia in former the detection of lung cancer patients harboring ana-
305(2):106–112. smokers. Cancer Prev Res (Phila). 2011;4(6):793– plastic lymphoma kinase (ALK) gene rearrangements
37. Schoen RE, Pinsky PF, Weissfeld JL, et al. Colorectal- 802. potentially suitable for ALK inhibitor treatment.
cancer incidence and mortality with screening flexible 146. Hecht SS. Cigarette smoking and lung cancer: Clin Cancer Res. 2010;16(22):5581–5590.
sigmoidoscopy. N Engl J Med. 2012;366(25): chemical mechanisms and approaches to prevention. 228. Kwak EL, Bang YJ, Camidge DR, et al. Anaplastic
2345–2357. Lancet Oncol. 2002;3(8):461–469. lymphoma kinase inhibition in non-small-cell
43. Jasperson KW, Tuohy TM, Neklason DW, Burt RW. 153. Jones PA, Baylin SB. The epigenomics of cancer. lung cancer. N Engl J Med. 2010;363(18):1693–
Hereditary and familial colon cancer. Gastroenterol- Cell. 2007;128(4):683–692. 1703.
ogy. 2010;138(6):2044–2058. 154. Berger SL, Kouzarides T, Shiekhattar R, Shilatifard 231. Campbell PJ, Stephens PJ, Pleasance ED, et al.
59. Lynch HT, Lynch JF. Lynch syndrome: history and A. An operational definition of epigenetics. Genes Identification of somatically acquired rearrangements
current status. Dis Markers. 2004;20(4–5):181–198. Dev. 2009;23(7):781–783. in cancer using genome-wide massively parallel
73. Boland CR, Thibodeau SN, Hamilton SR, et al. A 168. Govindan R, Ding L, Griffith M, et al. Genomic paired-end sequencing. Nat Genet. 2008;40(6):722–
National Cancer Institute Workshop on Microsat- landscape of non-small cell lung cancer in smokers 729.
ellite Instability for cancer detection and familial and never-smokers. Cell. 2012;150(6):1121–1134. 241. Beadling C, Wald AI, Warrick A, et al. A multiplexed
predisposition: development of international criteria 184. Crowley E, Di Nicolantonio F, Loupakis F, Bardelli amplicon approach for detecting gene fusions by
for the determination of microsatellite instability A. Liquid biopsy: monitoring cancer-genetics in the next-generation sequencing. J Mol Diagn. 2016;18(2):
in colorectal cancer. Cancer Res. 1998;58(22): blood. Nat Rev Clin Oncol. 2013;10(8):472–484. 165–175.
5248–5257. 189. Bettegowda C, Sausen M, Leary RJ, et al. Detection 246. Greene GL, Fitch FW, Jensen EV. Monoclonal anti-
79. Grover S, Kastrinos F, Steyerberg EW, et al. of circulating tumor DNA in early- and late-stage bodies to estrophilin: probes for the study of estrogen
Prevalence and phenotypes of APC and MUTYH human malignancies. Sci Transl Med. 2014;6(224): receptors. Proc Natl Acad Sci USA. 1980;77(1):
mutations in patients with multiple colorectal 224ra224. 157–161.
adenomas. JAMA. 2012;308(5):485–492. 195. Hunter T. The Croonian Lecture 1997. The 258. Cuzick J, Dowsett M, Pineda S, et al. Prognostic
96. Aretz S, Stienen D, Uhlhaas S, et al. High proportion phosphorylation of proteins on tyrosine: its role value of a combined estrogen receptor, progesterone
of large genomic STK11 deletions in Peutz-Jeghers in cell growth and disease. Philos Trans R Soc Lond receptor, Ki-67, and human epidermal growth
syndrome. Hum Mutat. 2005;26(6):513–519. B Biol Sci. 1998;353(1368):583–605. factor receptor 2 immunohistochemical score
Pathology, Biomarkers, and Molecular Diagnostics  •  CHAPTER 15 253

and comparison with the Genomic Health recur- 296. Lasota J, Miettinen M. Clinical significance of onco- 362. Lin JJ, Riely GJ, Shaw AT. Targeting ALK: Precision
rence score in early breast cancer. J Clin Oncol. genic KIT and PDGFRA mutations in gastrointestinal medicine takes on drug resistance. Cancer Discov.
2011;29(32):4273–4278. stromal tumours. Histopathology. 2008;53(3): 2017;7(2):137–155.
263. Pirker R, Pereira JR, von Pawel J, et al. EGFR 245–266. 372. Ettinger DS, Wood DE, Aisner DL, et al. Non-Small
expression as a predictor of survival for first-line 297. Martin-Broto J, Rubio L, Alemany R, Lopez- Cell Lung Cancer, Version 5.2017, NCCN Clinical
chemotherapy plus cetuximab in patients with Guerrero JA. Clinical implications of KIT and Practice Guidelines in Oncology. J Natl Compr Canc
advanced non-small-cell lung cancer: analysis of PDGFRA genotyping in GIST. Clin Transl Oncol. Netw. 2017;15(4):504–535.
data from the phase 3 FLEX study. Lancet Oncol. 2010;12(10):670–676. 373. Woodman SE, Lazar AJ, Aldape KD, Davies MA.
2012;13(1):33–42. 308. Sepulveda AR, Hamilton SR, Allegra CJ, et al. New strategies in melanoma: molecular testing in
264. Lantuejoul S, Varella-Garcia M, Thunnissen E, Yatabe Molecular biomarkers for the evaluation of colorectal advanced disease. Clin Cancer Res. 2012;18(5):
Y. Comparison of different assay platforms of ALK cancer: guideline from the American Society for 1195–1200.
testing. In: Tsao MS, Hirsch FR, Yatabe Y, eds. Clinical Pathology, College of American Pathologists, 382. Flaherty KT, Fisher DE. New strategies in metastatic
IASLC Atlas of ALK and ROS1 Testing in Lung Association for Molecular Pathology, and American melanoma: oncogene-defined taxonomy leads to
Cancer. 2nd ed. North Fort Myers, FL: Editorial Society of Clinical Oncology. Arch Pathol Lab Med. therapeutic advances. Clin Cancer Res. 2011;17(15):
Rx Press; 2016:73–83. 2017;141(5):625–657. 4922–4928.
267. Schreiber RD, Old LJ, Smyth MJ. Cancer 333. Prahallad A, Sun C, Huang S, et al. Unresponsive- 396. Cancer Genome Atlas Research Network, Brat
immunoediting: integrating immunity’s roles ness of colon cancer to BRAF(V600E) inhibition DJ, Verhaak RG, et al. Comprehensive, integrative
in cancer suppression and promotion. Science. through feedback activation of EGFR. Nature. genomic analysis of diffuse lower-grade gliomas.
2011;331(6024):1565–1570. 2012;483(7387):100–103. N Engl J Med. 2015;372(26):2481–2498.
274. Sharma P, Allison JP. Immune checkpoint targeting 341. Paez JG, Janne PA, Lee JC, et al. EGFR mutations 416. Lee J, Solomon DA, Tihan T. The role of histone
in cancer therapy: toward combination strategies in lung cancer: correlation with clinical response to modifications and telomere alterations in the patho-
with curative potential. Cell. 2015;161(2):205–214. gefitinib therapy. Science. 2004;304(5676):1497– genesis of diffuse gliomas in adults and children.
283. Gibney GT, Weiner LM, Atkins MB. Predictive 1500. J Neurooncol. 2017;132(1):1–11.
biomarkers for checkpoint inhibitor-based immu- 342. Lynch TJ, Bell DW, Sordella R, et al. Activating 419. Jiao Y, Killela PJ, Reitman ZJ, et al. Frequent
notherapy. Lancet Oncol. 2016;17(12):e542–e551. mutations in the epidermal growth factor receptor ATRX, CIC, FUBP1 and IDH1 mutations refine
294. Hirota S, Isozaki K, Moriyama Y, et al. Gain-of- underlying responsiveness of non-small-cell lung the classification of malignant gliomas. Oncotarget.
function mutations of c-kit in human gastrointestinal cancer to gefitinib. N Engl J Med. 2004;350(21): 2012;3(7):709–722.
stromal tumors. Science. 1998;279(5350):577–580. 2129–2139. 429. Wu J, Armstrong TS, Gilbert MR. Biology and
295. Corless CL, Barnett CM, Heinrich MC. Gastro- 343. Mok TS, Wu YL, Thongprasert S, et al. Gefitinib or management of ependymomas. Neuro Oncol.
intestinal stromal tumours: origin and molecular carboplatin-paclitaxel in pulmonary adenocarcinoma. 2016;18(7):902–913.
oncology. Nat Rev Cancer. 2011;11(12):865–878. N Engl J Med. 2009;361(10):947–957.
Pathology, Biomarkers, and Molecular Diagnostics  •  CHAPTER 15 253.e1
253.e1

REFERENCES
1. Hanahan D, Weinberg RA. Hallmarks of cancer: in the genomic era. J Mol Diagn. 2017;19(3): 42. Markowitz SD, Bertagnolli MM. Molecular origins
the next generation. Cell. 2011;144(5):646–674. 366–377. of cancer: molecular basis of colorectal cancer.
2. Pepe MS, Etzioni R, Feng Z, et al. Phases of bio- 22. Arbyn M, Roelens J, Cuschieri K, et al. The N Engl J Med. 2009;361(25):2449–2460.
marker development for early detection of cancer. APTIMA HPV assay versus the Hybrid Capture 43. Jasperson KW, Tuohy TM, Neklason DW, Burt RW.
J Natl Cancer Inst. 2001;93(14):1054–1061. 2 test in triage of women with ASC-US or LSIL Hereditary and familial colon cancer. Gastroenterol-
3. ICH Harmonised Tripartite Guideline. Statistical cervical cytology: a meta-analysis of the diagnostic ogy. 2010;138(6):2044–2058.
principles for clinical trials. International Conference accuracy. Int J Cancer. 2013;132(1):101–108. 44. Cripps WH. Two cases of disseminated polypus of
on Harmonisation E9 Expert Working Group. Stat 23. Arbyn M, Snijders PJ, Meijer CJ, et al. Which the rectum. Trans Pathol Soc London. 1882;33(16):
Med. 1999;18(15):1905–1942. high-risk HPV assays fulfil criteria for use in 165–168.
4. Pisani P, Parkin DM, Bray F, Ferlay J. Erratum: primary cervical cancer screening? Clin Microbiol 45. Veale AM. Clinical and genetic problems in familial
Estimates of the worldwide mortality from 25 cancers Infect. 2015;21(9):817–826. in-intestinal polyposis. Gut. 1960;1:285–290.
in 1990. Int. J. Cancer, 83, 18-29 (1999). Int J 24. Schlecht NF, Kulaga S, Robitaille J, et al. Persistent 46. Leppert M, Dobbs M, Scambler P, et al. The gene
Cancer. 1999;83(6):870–873. human papillomavirus infection as a predictor of cer- for familial polyposis coli maps to the long arm of
5. Papanicolaou GN. A survey of the actualities and vical intraepithelial neoplasia. JAMA. 2001;286(24): chromosome 5. Science. 1987;238(4832):1411–1413.
potentialities of exfoliative cytology in cancer 3106–3114. 47. Bodmer WF, Bailey CJ, Bodmer J, et al. Localization
diagnosis. Ann Intern Med. 1949;31(4):661–674. 25. Kinlen LJ, Spriggs AI. Women with positive cervical of the gene for familial adenomatous polyposis on
6. The 1988 Bethesda System for reporting cervical/ smears but without surgical intervention. A follow-up chromosome 5. Nature. 1987;328(6131):614–616.
vaginal cytological diagnoses. National Cancer study. Lancet. 1978;2(8087):463–465. 48. Groden J, Thliveris A, Samowitz W, et al. Identifica-
Institute Workshop. JAMA. 1989;262(7):931–934. 26. Mayrand MH, Duarte-Franco E, Rodrigues I, et al. tion and characterization of the familial adenomatous
7. Peto J, Gilham C, Fletcher O, Matthews FE. The Human papillomavirus DNA versus Papanicolaou polyposis coli gene. Cell. 1991;66(3):589–600.
cervical cancer epidemic that screening has prevented screening tests for cervical cancer. N Engl J Med. 49. Nishisho I, Nakamura Y, Miyoshi Y, et al. Mutations
in the UK. Lancet. 2004;364(9430):249–256. 2007;357(16):1579–1588. of chromosome 5q21 genes in FAP and colorectal
8. Walboomers JM, Jacobs MV, Manos MM, et al. 27. Ronco G, Giorgi-Rossi P, Carozzi F, et al. Efficacy cancer patients. Science. 1991;253(5020):665–669.
Human papillomavirus is a necessary cause of inva- of human papillomavirus testing for the detection of 50. Kinzler KW, Nilbert MC, Su LK, et al. Identification
sive cervical cancer worldwide. J Pathol. 1999;189(1): invasive cervical cancers and cervical intraepithelial of FAP locus genes from chromosome 5q21. Science.
12–19. neoplasia: a randomised controlled trial. Lancet 1991;253(5020):661–665.
9. Beard JW, Rous P. A Virus-induced mammalian Oncol. 2010;11(3):249–257. 51. Joslyn G, Carlson M, Thliveris A, et al. Identification
growth with the characters of a tumor (the Shope 28. Castle PE, Fetterman B, Thomas Cox J, et al. The of deletion mutations and three new genes at the
rabbit papilloma): II. Experimental alterations of the age-specific relationships of abnormal cytology and familial polyposis locus. Cell. 1991;66(3):601–613.
growth on the skin: morphological considerations: the human papillomavirus DNA results to the risk of 52. Goss KH, Groden J. Biology of the adenomatous
phenomena of retrogression. J Exp Med. 1934;60(6): cervical precancer and cancer. Obstet Gynecol. 2010; polyposis coli tumor suppressor. J Clin Oncol. 2000;
723–740. 116(1):76–84. 18(9):1967–1979.
10. Rous P, Beard JW. A virus-induced mammalian 29. Huh WK, Ault KA, Chelmow D, et al. Use of 53. Powell SM, Zilz N, Beazer-Barclay Y, et al. APC
growth with the characters of a tumor (the Shope primary high-risk human papillomavirus testing for mutations occur early during colorectal tumorigen-
rabbit papilloma): III. Further characters of the cervical cancer screening: interim clinical guidance. esis. Nature. 1992;359(6392):235–237.
growth: general discussion. J Exp Med. 1934;60(6): J Low Genit Tract Dis. 2015;19(2):91–96. 54. Samowitz WS, Slattery ML, Sweeney C, Herrick J,
741–766. 30. Schiffman M, Wentzensen N, Wacholder S, Kinney Wolff RK, Albertsen H. APC mutations and other
11. Rous P, Beard JW. A virus-induced mammalian W, Gage JC, Castle PE. Human papillomavirus genetic and epigenetic changes in colon cancer. Mol
growth with the characters of a tumor (the Shope testing in the prevention of cervical cancer. J Natl Cancer Res. 2007;5(2):165–170.
rabbit papilloma): I. The growth on implantation Cancer Inst. 2011;103(5):368–383. 55. De Benedetti L, Sciallero S, Gismondi V, et al.
within favorable hosts. J Exp Med. 1934;60(6): 31. Gravitt PE. The known unknowns of HPV natural Association of APC gene mutations and histological
701–722. history. J Clin Invest. 2011;121(12):4593–4599. characteristics of colorectal adenomas. Cancer Res.
12. zur Hausen H, Meinhof W, Scheiber W, Bornkamm 32. Stanley M. Prospects for new human papillomavirus 1994;54(13):3553–3556.
GW. Attempts to detect virus-specific DNA in vaccines. Curr Opin Infect Dis. 2010;23(1):70– 56. Lynch HT, Smyrk T, McGinn T, et al. Attenuated
human tumors. I. Nucleic acid hybridizations with 75. familial adenomatous polyposis (AFAP). A pheno-
complementary RNA of human wart virus. Int J 33. Villa LL. HPV prophylactic vaccination: The first typically and genotypically distinctive variant of FAP.
Cancer. 1974;13(5):650–656. years and what to expect from now. Cancer Lett. Cancer. 1995;76(12):2427–2433.
13. zur Hausen H. Condylomata acuminata and human 2011;305(2):106–112. 57. Knudsen AL, Bisgaard ML, Bulow S. Attenuated
genital cancer. Cancer Res. 1976;36(2 Pt 2):794. 34. Citarda F, Tomaselli G, Capocaccia R, Barcherini familial adenomatous polyposis (AFAP). A review
14. zur Hausen H. Human papillomaviruses and their S, Crespi M. Efficacy in standard clinical practice of the literature. Fam Cancer. 2003;2(1):43–55.
possible role in squamous cell carcinomas. Curr Top of colonoscopic polypectomy in reducing colorectal 58. Warthin AS. Heredity with reference to carcinoma.
Microbiol Immunol. 1977;78:1–30. cancer incidence. Gut. 2001;48(6):812–815. Arch Intern Med. 1913;12:546–555.
15. zur Hausen H. Papillomaviruses causing cancer: 35. Winawer SJ, Zauber AG, Ho MN, et al. Prevention 59. Lynch HT, Lynch JF. Lynch syndrome: history and
evasion from host-cell control in early events in car- of colorectal cancer by colonoscopic polypectomy. current status. Dis Markers. 2004;20(4–5):181–198.
cinogenesis. J Natl Cancer Inst. 2000;92(9):690–698. The National Polyp Study Workgroup. N Engl J 60. Vasen HF, Mecklin JP, Khan PM, Lynch HT. The
16. zur Hausen H. Papillomaviruses and cancer: from Med. 1993;329(27):1977–1981. International Collaborative Group on Hereditary
basic studies to clinical application. Nat Rev Cancer. 36. Zauber AG, Winawer SJ, O’Brien MJ, et al. Colo- Non-Polyposis Colorectal Cancer (ICG-HNPCC).
2002;2(5):342–350. noscopic polypectomy and long-term prevention Dis Colon Rectum. 1991;34(5):424–425.
17. Werness BA, Levine AJ, Howley PM. Association of of colorectal-cancer deaths. N Engl J Med. 2012; 61. Vasen HF, Watson P, Mecklin JP, Lynch HT. New
human papillomavirus types 16 and 18 E6 proteins 366(8):687–696. clinical criteria for hereditary nonpolyposis colorectal
with p53. Science. 1990;248(4951):76–79. 37. Schoen RE, Pinsky PF, Weissfeld JL, et  al. cancer (HNPCC, Lynch syndrome) proposed by
18. Dyson N, Howley PM, Munger K, Harlow E. The Colorectal-cancer incidence and mortality with the International Collaborative group on HNPCC.
human papilloma virus-16 E7 oncoprotein is able screening flexible sigmoidoscopy. N Engl J Med. Gastroenterology. 1999;116(6):1453–1456.
to bind to the retinoblastoma gene product. Science. 2012;366(25):2345–2357. 62. Peltomaki P, Aaltonen LA, Sistonen P, et al. Genetic
1989;243(4893):934–937. 38. Rider JA, Kirsner JB, Moeller HC, Palmer WL. mapping of a locus predisposing to human colorectal
19. Schwarz E, Freese UK, Gissmann L, et al. Struc- Polyps of the colon and rectum; their incidence and cancer. Science. 1993;260(5109):810–812.
ture and transcription of human papillomavirus relationship to carcinoma. Am J Med. 1954;16(4): 63. Bronner CE, Baker SM, Morrison PT, et al. Muta-
sequences in cervical carcinoma cells. Nature. 555–564. tion in the DNA mismatch repair gene homologue
1985;314(6006):111–114. 39. Fearon ER, Hamilton SR, Vogelstein B. Clonal hMLH1 is associated with hereditary non-polyposis
20. Wentzensen N, Vinokurova S, von Knebel Doeberitz analysis of human colorectal tumors. Science. 1987; colon cancer. Nature. 1994;368(6468):258–261.
M. Systematic review of genomic integration sites 238(4824):193–197. 64. Papadopoulos N, Nicolaides NC, Wei YF, et al.
of human papillomavirus genomes in epithelial 40. Vogelstein B, Fearon ER, Hamilton SR, et al. Genetic Mutation of a mutL homolog in hereditary colon
dysplasia and invasive cancer of the female lower alterations during colorectal-tumor development. N cancer. Science. 1994;263(5153):1625–1629.
genital tract. Cancer Res. 2004;64(11):3878– Engl J Med. 1988;319(9):525–532. 65. Leach FS, Nicolaides NC, Papadopoulos N, et al.
3884. 41. Fearon ER, Vogelstein B. A genetic model for Mutations of a mutS homolog in hereditary non-
21. Leal SM Jr, Gulley ML. Current and emerging molec- colorectal tumorigenesis. Cell. 1990;61(5):759– polyposis colorectal cancer. Cell. 1993;75(6):1215–
ular tests for human papillomavirus-related neoplasia 767. 1225.
253.e2 Part I: Science and Clinical Oncology

66. Fishel R, Lescoe MK, Rao MR, et al. The human mucosa, lips and digits; a syndrome of diagnostic sig- 108. Calva-Cerqueira D, Chinnathambi S, Pechman B,
mutator gene homolog MSH2 and its association nificance. N Engl J Med. 1949;241(26):1031–1036. Bair J, Larsen-Haidle J, Howe JR. The rate of germ-
with hereditary nonpolyposis colon cancer. Cell. 85. Bruwer A, Bargen JA, Kierland RR. Surface line mutations and large deletions of SMAD4 and
1994;77(1):1 p following 166. pigmentation and generalized intestinal polyposis; BMPR1A in juvenile polyposis. Clin Genet. 2009;
67. Miyaki M, Konishi M, Tanaka K, et  al. (Peutz-Jeghers syndrome). Proc Staff Meet Mayo Clin. 75(1):79–85.
Germline mutation of MSH6 as the cause of 1954;29(6):168–171. 109. van Hattem WA, Brosens LA, de Leng WW, et al.
hereditary nonpolyposis colorectal cancer. Nat Genet. 86. Beggs AD, Latchford AR, Vasen HF, et  al. Large genomic deletions of SMAD4, BMPR1A
1997;17(3):271–272. Peutz-Jeghers syndrome: a systematic review and and PTEN in juvenile polyposis. Gut. 2008;57(5):
68. Li GM, Modrich P. Restoration of mismatch repair recommendations for management. Gut. 2010;59(7): 623–627.
to nuclear extracts of H6 colorectal tumor cells by a 975–986. 110. O’Malley M, Laguardia L, Kalady MF, et al. The
heterodimer of human MutL homologs. Proc Natl 87. Mehenni H, Resta N, Park JG, Miyaki M, Guanti prevalence of hereditary hemorrhagic telangiectasia
Acad Sci USA. 1995;92(6):1950–1954. G, Costanza MC. Cancer risks in LKB1 germline in juvenile polyposis syndrome. Dis Colon Rectum.
69. Nakagawa H, Lockman JC, Frankel WL, et al. Mis- mutation carriers. Gut. 2006;55(7):984–990. 2012;55(8):886–892.
match repair gene PMS2: disease-causing germline 88. Hearle N, Schumacher V, Menko FH, et al. Fre- 111. Hyman NH, Anderson P, Blasyk H. Hyperplastic
mutations are frequent in patients whose tumors quency and spectrum of cancers in the Peutz-Jeghers polyposis and the risk of colorectal cancer. Dis Colon
stain negative for PMS2 protein, but paralogous syndrome. Clin Cancer Res. 2006;12(10):3209–3215. Rectum. 2004;47(12):2101–2104.
genes obscure mutation detection and interpretation. 89. Giardiello FM, Brensinger JD, Tersmette AC, et al. 112. Rosty C, Buchanan DD, Walsh MD, et al. Phenotype
Cancer Res. 2004;64(14):4721–4727. Very high risk of cancer in familial Peutz-Jeghers and polyp landscape in serrated polyposis syndrome:
70. Kane MF, Loda M, Gaida GM, et al. Methylation syndrome. Gastroenterology. 2000;119(6):1447–1453. a series of 100 patients from genetics clinics. Am J
of the hMLH1 promoter correlates with lack of 90. Mehenni H, Blouin JL, Radhakrishna U, et al. Surg Pathol. 2012;36(6):876–882.
expression of hMLH1 in sporadic colon tumors Peutz-Jeghers syndrome: confirmation of linkage 113. Young JP, Parry S. Risk factors: Hyperplastic polyposis
and mismatch repair-defective human tumor cell to chromosome 19p13.3 and identification of a syndrome and risk of colorectal cancer. Nat Rev
lines. Cancer Res. 1997;57(5):808–811. potential second locus, on 19q13.4. Am J Hum Gastroenterol Hepatol. 2010;7(11):594–595.
71. Herman JG, Umar A, Polyak K, et al. Incidence Genet. 1997;61(6):1327–1334. 114. Boparai KS, Reitsma JB, Lemmens V, et al. Increased
and functional consequences of hMLH1 promoter 91. Amos CI, Bali D, Thiel TJ, et al. Fine mapping of a colorectal cancer risk in first-degree relatives of
hypermethylation in colorectal carcinoma. Proc Natl genetic locus for Peutz-Jeghers syndrome on chromo- patients with hyperplastic polyposis syndrome.
Acad Sci USA. 1998;95(12):6870–6875. some 19p. Cancer Res. 1997;57(17):3653–3656. Gut. 2010;59(9):1222–1225.
72. Dietmaier W, Wallinger S, Bocker T, Kullmann 92. de Leng WW, Jansen M, Carvalho R, et al. Genetic 115. Boparai KS, Mathus-Vliegen EM, Koornstra JJ, et al.
F, Fishel R, Ruschoff J. Diagnostic microsatellite defects underlying Peutz-Jeghers syndrome (PJS) Increased colorectal cancer risk during follow-up in
instability: definition and correlation with mis- and exclusion of the polarity-associated MARK/ patients with hyperplastic polyposis syndrome: a mul-
match repair protein expression. Cancer Res. 1997; Par1 gene family as potential PJS candidates. Clin ticentre cohort study. Gut. 2010;59(8):1094–1100.
57(21):4749–4756. Genet. 2007;72(6):568–573. 116. Allison JE, Tekawa IS, Ransom LJ, Adrain AL. A
73. Boland CR, Thibodeau SN, Hamilton SR, 93. Volikos E, Robinson J, Aittomaki K, et al. LKB1 comparison of fecal occult-blood tests for colorectal-
et al. A National Cancer Institute Workshop on exonic and whole gene deletions are a common cause cancer screening. N Engl J Med. 1996;334(3):
Microsatellite Instability for cancer detection and of Peutz-Jeghers syndrome. J Med Genet. 2006;43(5): 155–159.
familial predisposition: development of international e18. 117. Mandel JS, Church TR, Bond JH, et al. The effect
criteria for the determination of microsatellite 94. Jenne DE, Reimann H, Nezu J, et al. Peutz-Jeghers of fecal occult-blood screening on the incidence
instability in colorectal cancer. Cancer Res. 1998; syndrome is caused by mutations in a novel serine of colorectal cancer. N Engl J Med. 2000;343(22):
58(22):5248–5257. threonine kinase. Nat Genet. 1998;18(1):38–43. 1603–1607.
74. Umar A, Boland CR, Terdiman JP, et al. Revised 95. Hemminki A, Markie D, Tomlinson I, et al. A serine/ 118. Kronborg O, Fenger C, Olsen J, Jorgensen OD,
Bethesda Guidelines for hereditary nonpolyposis threonine kinase gene defective in Peutz-Jeghers Sondergaard O. Randomised study of screening
colorectal cancer (Lynch syndrome) and micro- syndrome. Nature. 1998;391(6663):184–187. for colorectal cancer with faecal-occult-blood test.
satellite instability. J Natl Cancer Inst. 2004;96(4): 96. Aretz S, Stienen D, Uhlhaas S, et al. High proportion Lancet. 1996;348(9040):1467–1471.
261–268. of large genomic STK11 deletions in Peutz-Jeghers 119. Hardcastle JD, Chamberlain JO, Robinson MH,
75. Samowitz WS, Curtin K, Lin HH, et al. The colon syndrome. Hum Mutat. 2005;26(6):513–519. et al. Randomised controlled trial of faecal-occult-
cancer burden of genetically defined hereditary non- 97. Hemminki A, Tomlinson I, Markie D, et al. blood screening for colorectal cancer. Lancet.
polyposis colon cancer. Gastroenterology. 2001;121(4): Localization of a susceptibility locus for Peutz- 1996;348(9040):1472–1477.
830–838. Jeghers syndrome to 19p using comparative genomic 120. Quintero E, Castells A, Bujanda L, et  al.
76. Al-Tassan N, Chmiel NH, Maynard J, et al. Inherited hybridization and targeted linkage analysis. Nat Colonoscopy versus fecal immunochemical testing
variants of MYH associated with somatic G:C– Genet. 1997;15(1):87–90. in colorectal-cancer screening. N Engl J Med. 2012;
>T:A mutations in colorectal tumors. Nat Genet. 98. Bignell GR, Barfoot R, Seal S, Collins N, Warren W, 366(8):697–706.
2002;30(2):227–232. Stratton MR. Low frequency of somatic mutations in 121. Ahlquist DA, Zou H, Domanico M, et al. Next-
77. Nielsen M, Morreau H, Vasen HF, Hes FJ. MUTYH- the LKB1/Peutz-Jeghers syndrome gene in sporadic generation stool DNA test accurately detects
associated polyposis (MAP). Crit Rev Oncol Hematol. breast cancer. Cancer Res. 1998;58(7):1384–1386. colorectal cancer and large adenomas. Gastroenterol-
2011;79(1):1–16. 99. Giardiello FM, Trimbath JD. Peutz-Jeghers syn- ogy. 2012;142(2):248–256, quiz e225–e246.
78. Lubbe SJ, Di Bernardo MC, Chandler IP, Houlston drome and management recommendations. Clin 122. Kim MS, Lee J, Sidransky D. DNA methylation
RS. Clinical implications of the colorectal cancer risk Gastroenterol Hepatol. 2006;4(4):408–415. markers in colorectal cancer. Cancer Metastasis Rev.
associated with MUTYH mutation. J Clin Oncol. 100. Roth SI, Helwig EB. Juvenile polyps of the colon 2010;29(1):181–206.
2009;27(24):3975–3980. and rectum. Cancer. 1963;16:468–479. 123. Siegel R, Desantis C, Virgo K, et al. Cancer treatment
79. Grover S, Kastrinos F, Steyerberg EW, et al. 101. Giardiello FM, Hamilton SR, Kern SE, et al. and survivorship statistics, 2012. CA Cancer J Clin.
Prevalence and phenotypes of APC and MUTYH Colorectal neoplasia in juvenile polyposis or juvenile 2012.
mutations in patients with multiple colorectal polyps. Arch Dis Child. 1991;66(8):971–975. 124. Siegel R, Naishadham D, Jemal A. Cancer statistics,
adenomas. JAMA. 2012;308(5):485–492. 102. Nugent KP, Talbot IC, Hodgson SV, Phillips RK. 2012. CA Cancer J Clin. 2012;62(1):10–29.
80. Hutchinson J. Pigmentation of lips and mouth. Solitary juvenile polyps: not a marker for subsequent 125. Omenn GS, Goodman GE, Thornquist MD, et al.
Arch Surg. 1896;7:290e291. malignancy. Gastroenterology. 1993;105(3):698–700. Risk factors for lung cancer and for intervention
81. Connor JT. Aesculapian Society of London. Lancet. 103. McColl I, Busxey HJ, Veale AM, Morson BC. Juvenile effects in CARET, the Beta-Carotene and Retinol
1895;2(1):169. polyposis coli. Proc R Soc Med. 1964;57:896–897. Efficacy Trial. J Natl Cancer Inst. 1996;88(21):
82. Peutz J. Very remarkable case of familial polyposis of 104. Jass JR, Williams CB, Bussey HJ, Morson BC. 1550–1559.
mucous membrane of intestinal tract and nasophar- Juvenile polyposis—a precancerous condition. 126. Goodman GE, Thornquist MD, Balmes J, et al.
ynx accompanied by peculiar pigmentation of skin Histopathology. 1988;13(6):619–630. The Beta-Carotene and Retinol Efficacy Trial:
and mucous membrane. Nederl Maandschr Geneesk. 105. Veale AM, McColl I, Bussey HJ, Morson BC. incidence of lung cancer and cardiovascular disease
1921;10:134e146. Juvenile polyposis coli. J Med Genet. 1966;3(1):5–16. mortality during 6-year follow-up after stopping
83. Jeghers H, McKusick KV, Katz KH. Generalized 106. Brosens LA, van Hattem A, Hylind LM, et al. beta-carotene and retinol supplements. J Natl Cancer
intestinal polyposis and melanin spots of the oral Risk of colorectal cancer in juvenile polyposis. Gut. Inst. 2004;96(23):1743–1750.
mucosa, lips and digits; a syndrome of diagnostic 2007;56(7):965–967. 127. Hennekens CH, Buring JE, Manson JE, et al.
significance. N Engl J Med. 1949;241(25):993, illust; 107. Aretz S, Stienen D, Uhlhaas S, et al. High proportion Lack of effect of long-term supplementation
passim. of large genomic deletions and a genotype phenotype with beta carotene on the incidence of malignant
84. Jeghers H, McKusick KV, Katz KH. Generalized update in 80 unrelated families with juvenile polypo- neoplasms and cardiovascular disease. N Engl J Med.
intestinal polyposis and melanin spots of the oral sis syndrome. J Med Genet. 2007;44(11):702–709. 1996;334(18):1145–1149.
Pathology, Biomarkers, and Molecular Diagnostics  •  CHAPTER 15 253.e3
253.e3

128. Heinonen OP, Albanes D. The effect of vitamin E 147. Veglia F, Matullo G, Vineis P. Bulky DNA adducts transcriptome. Proc Natl Acad Sci USA. 2004;101(27):
and beta carotene on the incidence of lung cancer and risk of cancer: a meta-analysis. Cancer Epidemiol 10143–10148.
and other cancers in male smokers. The Alpha- Biomarkers Prev. 2003;12(2):157–160. 167. Spira A, Beane JE, Shah V, et al. Airway epithelial gene
Tocopherol, Beta Carotene Cancer Prevention Study 148. Hung RJ, Christiani DC, Risch A, et al. International expression in the diagnostic evaluation of smokers
Group. N Engl J Med. 1994;330(15):1029–1035. Lung Cancer Consortium: pooled analysis of sequence with suspect lung cancer. Nat Med. 2007;13(3):
129. Henschke CI, McCauley DI, Yankelevitz DF, et al. variants in DNA repair and cell cycle pathways. 361–366.
Early Lung Cancer Action Project: overall design Cancer Epidemiol Biomarkers Prev. 2008;17(11): 168. Govindan R, Ding L, Griffith M, et al. Genomic
and findings from baseline screening. Lancet. 1999; 3081–3089. landscape of non-small cell lung cancer in smokers
354(9173):99–105. 149. Yu H, Zhao H, Wang LE, et al. An analysis of and never-smokers. Cell. 2012;150(6):1121–1134.
130. Henschke CI, Yankelevitz DF, Libby DM, Pasmantier single nucleotide polymorphisms of 125 DNA repair 169. Ahrar K, Wallace M, Javadi S, Gupta S. Mediastinal,
MW, Smith JP, Miettinen OS. Survival of patients genes in the Texas genome-wide association study hilar, and pleural image-guided biopsy: current
with stage I lung cancer detected on CT screening. of lung cancer with a replication for the XRCC4 practice and techniques. Semin Respir Crit Care
N Engl J Med. 2006;355(17):1763–1771. SNPs. DNA Repair (Amst). 2011;10(4):398–407. Med. 2008;29(4):350–360.
131. Aberle DR, Adams AM, Berg CD, et al. Reduced 150. Wei S, Niu J, Zhao H, et al. Association of a novel 170. Cham MD, Lane ME, Henschke CI, Yankelevitz
lung-cancer mortality with low-dose computed functional promoter variant (rs2075533 C>T) in the DF. Lung biopsy: special techniques. Semin Respir
tomographic screening. N Engl J Med. 2011; apoptosis gene TNFSF8 with risk of lung cancer—a Crit Care Med. 2008;29(4):335–349.
365(5):395–409. finding from Texas lung cancer genome-wide associa- 171. Franklin WA, Haney J, Sugita M, Bemis L, Jimeno
132. Bach PB, Mirkin JN, Oliver TK, et al. Benefits and tion study. Carcinogenesis. 2011;32(4):507–515. A, Messersmith WA. KRAS mutation: comparison
harms of CT screening for lung cancer: a systematic 151. Dong LM, Potter JD, White E, Ulrich CM, Cardon of testing methods and tissue sampling techniques
review. JAMA. 2012;307(22):2418–2429. LR, Peters U. Genetic susceptibility to cancer: the in colon cancer. J Mol Diagn. 2010;12(1):43–50.
133. Patz EF Jr, Pinsky P, Gatsonis C, et al. Overdiagnosis role of polymorphisms in candidate genes. JAMA. 172. Malapelle U, de Rosa N, Rocco D, et al. EGFR
in low-dose computed tomography screening for lung 2008;299(20):2423–2436. and KRAS mutations detection on lung cancer
cancer. JAMA Intern Med. 2014;174(2):269–274. 152. Vineis P, Manuguerra M, Kavvoura FK, et al. A liquid-based cytology: a pilot study. J Clin Pathol.
134. Franklin WA, Muller KM, Wistuba I, et al. Squamous field synopsis on low-penetrance variants in DNA 2012;65(1):87–91.
dysplasia and carcinoma in sutu. In: Travis WD, repair genes and cancer susceptibility. J Natl Cancer 173. Malapelle U, Bellevicine C, Zeppa P, Palombini
Brambilla E, Muller-Hermelink HK, Harris CC, Inst. 2009;101(1):24–36. L, Troncone G. Cytology-based gene mutation
eds. Tumours of the Lung, Pleura, Thymus and Heart. 153. Jones PA, Baylin SB. The epigenomics of cancer. tests to predict response to anti-epidermal growth
2nd ed. Lyon, France: IARC Press; 2004:68–72. Cell. 2007;128(4):683–692. factor receptor therapy: a review. Diagn Cytopathol.
135. Auerbach O, Stout AP, Hammond EC, Garfinkel 154. Berger SL, Kouzarides T, Shiekhattar R, Shilatifard 2011;39(9):703–710.
L. Changes in bronchial epithelium in relation to A. An operational definition of epigenetics. Genes 174. Aisner DL, Deshpande C, Baloch Z, et al. Evaluation
cigarette smoking and in relation to lung cancer. Dev. 2009;23(7):781–783. of EGFR mutation status in cytology specimens: an
N Engl J Med. 1961;265:253–267. 155. Merlo A, Herman JG, Mao L, et al. 5′ CpG island institutional experience. Diagn Cytopathol. 2011.
136. Auerbach O, Stout AP, Hammond EC, Garfinkel L. methylation is associated with transcriptional silenc- 175. da Cunha Santos G, Saieg MA, Geddie W, Leighl N.
Changes in bronchial epithelium in relation to sex, ing of the tumour suppressor p16/CDKN2/MTS1 EGFR gene status in cytological samples of nonsmall
age, residence, smoking and pneumonia. N Engl J in human cancers. Nat Med. 1995;1(7):686–692. cell lung carcinoma: controversies and opportunities.
Med. 1962;267:111–119. 156. Herman JG, Graff JR, Myohanen S, Nelkin BD, Cancer Cytopathol. 2011;119(2):80–91.
137. Hirsch FR, Prindiville SA, Miller YE, et al. Fluores- Baylin SB. Methylation-specific PCR: a novel PCR 176. Pirker R, Filipits M. Monoclonal antibodies against
cence versus white-light bronchoscopy for detection assay for methylation status of CpG islands. Proc EGFR in non-small cell lung cancer. Crit Rev Oncol
of preneoplastic lesions: a randomized study. J Natl Natl Acad Sci USA. 1996;93(18):9821–9826. Hematol. 2011;80(1):1–9.
Cancer Inst. 2001;93(18):1385–1391. 157. Palmisano WA, Divine KK, Saccomanno G, et al. 177. Cabel L, Proudhon C, Gortais H, et al. Circulating
138. Byers T, Wolf HJ, Franklin WA, et al. Sputum Predicting lung cancer by detecting aberrant promoter tumor cells: clinical validity and utility. Int J Clin
cytologic atypia predicts incident lung cancer: methylation in sputum. Cancer Res. 2000;60(21): Oncol. 2017.
defining latency and histologic specificity. Cancer 5954–5958. 178. Riethdorf S, Fritsche H, Muller V, et al. Detection of
Epidemiol Biomarkers Prev. 2008;17(1):158–162. 158. Belinsky SA, Liechty KC, Gentry FD, et al. Promoter circulating tumor cells in peripheral blood of patients
139. Merrick DT, Gao D, Miller YE, et al. Persistence of hypermethylation of multiple genes in sputum with metastatic breast cancer: a validation study of
bronchial dysplasia is associated with development precedes lung cancer incidence in a high-risk cohort. the CellSearch system. Clin Cancer Res. 2007;13(3):
of invasive squamous cell carcinoma. Cancer Prev Cancer Res. 2006;66(6):3338–3344. 920–928.
Res (Phila). 2016;9(1):96–104. 159. Leng S, Do K, Yingling CM, et al. Defining a gene 179. Hou JM, Krebs MG, Lancashire L, et al. Clinical
140. Keith RL, Blatchford PJ, Kittelson J, et al. Oral promoter methylation signature in sputum for lung significance and molecular characteristics of circulat-
iloprost improves endobronchial dysplasia in former cancer risk assessment. Clin Cancer Res. 2012;18(12): ing tumor cells and circulating tumor microemboli
smokers. Cancer Prev Res (Phila). 2011;4(6):793–802. 3387–3395. in patients with small-cell lung cancer. J Clin Oncol.
141. Travis WD, Naguchi M, Yatabe Y, et al. Adeno- 160. Hulbert A, Jusue-Torres I, Stark A, et al. Early 2012;30(5):525–532.
carcinoma. In: Travis WD, Brambilla E, Burke AP, detection of lung cancer using DNA promoter 180. Cristofanilli M, Budd GT, Ellis MJ, et  al.
Marx A, Nicholson AG, eds. WHO Classification hypermethylation in plasma and sputum. Clin Circulating tumor cells, disease progression, and
of Tumours of the Lung, Pleura, Thymus and Heart. Cancer Res. 2017;23(8):1998–2005. survival in metastatic breast cancer. N Engl J Med.
4th ed. Lyon: International Agency for REsearch 161. Jonsson S, Varella-Garcia M, Miller YE, et al. 2004;351(8):781–791.
on Cancer (IARC); 2015:26–50. Chromosomal aneusomy in bronchial high-grade 181. Bidard FC, Peeters DJ, Fehm T, et al. Clinical validity
142. Yip R, Yankelevitz DF, Hu M, et al. Lung cancer lesions is associated with invasive lung cancer. Am of circulating tumour cells in patients with metastatic
deaths in the National Lung Screening Trial attrib- J Respir Crit Care Med. 2008;177(3):342–347. breast cancer: a pooled analysis of individual patient
uted to nonsolid nodules. Radiology. 2016;281(2): 162. Varella-Garcia M, Kittelson J, Schulte AP, et al. data. Lancet Oncol. 2014;15(4):406–414.
589–596. Multi-target interphase fluorescence in situ hybridiza- 182. Ilie M, Hofman V, Long-Mira E, et al. Sentinel”
143. Henschke CI, Yip R, Smith JP, et al. CT screen- tion assay increases sensitivity of sputum cytology circulating tumor cells allow early diagnosis of
ing for lung cancer: part-solid nodules in baseline as a predictor of lung cancer. Cancer Detect Prev. lung cancer in patients with chronic obstructive
and annual repeat rounds. AJR Am J Roentgenol. 2004;28(4):244–251. pulmonary disease. PLoS ONE. 2014;9(10):e111597.
2016;207(6):1176–1184. 163. Li R, Todd NW, Qiu Q, et al. Genetic deletions in 183. Lo YM, Chan KC, Sun H, et al. Maternal plasma
144. Yip R, Wolf A, Tam K, et al. Outcomes of lung sputum as diagnostic markers for early detection of DNA sequencing reveals the genome-wide genetic
cancers manifesting as nonsolid nodules. Lung stage I non-small cell lung cancer. Clin Cancer Res. and mutational profile of the fetus. Sci Transl Med.
Cancer. 2016;97:35–42. 2007;13(2 Pt 1):482–487. 2010;2(61):61ra91.
145. Detterbeck FC, Marom EM, Arenberg DA, et al. The 164. Varella-Garcia M, Schulte AP, Wolf HJ, et al. The 184. Crowley E, Di Nicolantonio F, Loupakis F, Bardelli
IASLC Lung Cancer Staging Project: background detection of chromosomal aneusomy by fluorescence A. Liquid biopsy: monitoring cancer-genetics in
data and proposals for the application of TNM in situ hybridization in sputum predicts lung cancer the blood. Nat Rev Clin Oncol. 2013;10(8):472–
staging rules to lung cancer presenting as multiple incidence. Cancer Prev Res (Phila). 2010;3(4): 484.
nodules with ground glass or lepidic features or a 447–453. 185. Kimura H, Kasahara K, Kawaishi M, et al. Detection
pneumonic type of involvement in the forthcoming 165. Rafnar T, Sulem P, Besenbacher S, et al. Genome- of epidermal growth factor receptor mutations in
eighth edition of the TNM Classification. J Thorac wide significant association between a sequence serum as a predictor of the response to gefitinib
Oncol. 2016;11(5):666–680. variant at 15q15.2 and lung cancer risk. Cancer in patients with non-small-cell lung cancer. Clin
146. Hecht SS. Cigarette smoking and lung cancer: Res. 2011;71(4):1356–1361. Cancer Res. 2006;12(13):3915–3921.
chemical mechanisms and approaches to prevention. 166. Spira A, Beane J, Shah V, et al. Effects of ciga- 186. Vogelstein B, Kinzler KW. Digital PCR. Proc Natl
Lancet Oncol. 2002;3(8):461–469. rette smoke on the human airway epithelial cell Acad Sci USA. 1999;96(16):9236–9241.
253.e4 Part I: Science and Clinical Oncology

187. Thompson JD, Shibahara G, Rajan S, Pel J, Marziali 211. Carter SL, Cibulskis K, Helman E, et al. Absolute ALK gene rearrangement: a retrospective analysis.
A. Winnowing DNA for rare sequences: highly quantification of somatic DNA alterations in human Lancet Oncol. 2011;12(11):1004–1012.
specific sequence and methylation based enrichment. cancer. Nat Biotechnol. 2012;30(5):413–421. 230. Ju YS, Lee WC, Shin JY, et al. A transforming
PLoS ONE. 2012;7(2):e31597. 212. Aziz N, Zhao Q, Bry L, et al. College of American KIF5B and RET gene fusion in lung adenocarcinoma
188. Newman AM, Lovejoy AF, Klass DM, et al. Pathologists’ laboratory standards for next-generation revealed from whole-genome and transcriptome
Integrated digital error suppression for improved sequencing clinical tests. Arch Pathol Lab Med. sequencing. Genome Res. 2012.
detection of circulating tumor DNA. Nat Biotechnol. 2015;139(4):481–493. 231. Campbell PJ, Stephens PJ, Pleasance ED, et al. Iden-
2016;34(5):547–555. 213. Horak P, Frohling S, Glimm H. Integrating next- tification of somatically acquired rearrangements in
189. Bettegowda C, Sausen M, Leary RJ, et al. Detection generation sequencing into clinical oncology: strate- cancer using genome-wide massively parallel paired-
of circulating tumor DNA in early- and late-stage gies, promises and pitfalls. ESMO Open. 2016;1(5): end sequencing. Nat Genet. 2008;40(6):722–729.
human malignancies. Sci Transl Med. 2014;6(224): e000094. 232. Lipson D, Capelletti M, Yelensky R, et al. Identifica-
224ra224. 214. Fisher S, Barry A, Abreu J, et al. A scalable, fully tion of new ALK and RET gene fusions from colorec-
190. Maheswaran S, Sequist LV, Nagrath S, et al. Detec- automated process for construction of sequence-ready tal and lung cancer biopsies. Nat Med. 2012;18(3):
tion of mutations in EGFR in circulating lung-cancer human exome targeted capture libraries. Genome 382–384.
cells. N Engl J Med. 2008;359(4):366–377. Biol. 2011;12(1):R1. 233. Drilon A, Wang L, Arcila ME, et al. Broad, hybrid
191. Dawson SJ, Tsui DW, Murtaza M, et al. Analysis of 215. van Dijk EL, Jaszczyszyn Y, Thermes C. Library capture-based next-generation sequencing identifies
circulating tumor DNA to monitor metastatic breast preparation methods for next-generation sequencing: actionable genomic alterations in lung adenocarci-
cancer. N Engl J Med. 2013;368(13):1199–1209. tone down the bias. Exp Cell Res. 2014;322(1):12–20. nomas otherwise negative for such alterations by
192. Collins FS, Morgan M, Patrinos A. The Human 216. Roy S, LaFramboise WA, Nikiforov YE, et al. Next- other genomic testing approaches. Clin Cancer Res.
Genome Project: lessons from large-scale biology. generation sequencing informatics: challenges and 2015;21(16):3631–3639.
Science. 2003;300(5617):286–290. strategies for implementation in a clinical environ- 234. Korbel JO, Urban AE, Affourtit JP, et al. Paired-end
193. Lander ES, Linton LM, Birren B, et  al. ment. Arch Pathol Lab Med. 2016;140(9):958–975. mapping reveals extensive structural variation in the
Initial sequencing and analysis of the human 217. Robinson JT, Thorvaldsdottir H, Winckler W, human genome. Science. 2007;318(5849):420–426.
genome. Nature. 2001;409(6822):860–921. et al. Integrative genomics viewer. Nat Biotechnol. 235. Chen K, Wallis JW, McLellan MD, et al. Break-
194. Venter JC, Adams MD, Myers EW, et al. The 2011;29(1):24–26. Dancer: an algorithm for high-resolution mapping of
sequence of the human genome. Science. 2001; 218. Li H, Handsaker B, Wysoker A, et al. The Sequence genomic structural variation. Nat Methods. 2009;6(9):
291(5507):1304–1351. Alignment/Map format and SAMtools. Bioinformat- 677–681.
195. Hunter T. The Croonian Lecture 1997. The ics. 2009;25(16):2078–2079. 236. Wang J, Mullighan CG, Easton J, et al. CREST
phosphorylation of proteins on tyrosine: its role 219. Li MM, Datto M, Duncavage EJ, et al. Standards maps somatic structural variation in cancer genomes
in cell growth and disease. Philos Trans R Soc Lond and guidelines for the interpretation and reporting with base-pair resolution. Nat Methods. 2011;8(8):
B Biol Sci. 1998;353(1368):583–605. of sequence variants in cancer: a joint consensus 652–654.
196. Hunter T. Signaling—2000 and beyond. Cell. recommendation of the Association for Molecular 237. Li C, Fang R, Sun Y, et al. Spectrum of oncogenic
2000;100(1):113–127. Pathology, American Society of Clinical Oncology, driver mutations in lung adenocarcinomas from East
197. Barker A. Q&A: Anna Barker on the cancer genome and College of American Pathologists. J Mol Diagn. Asian never smokers. PLoS ONE. 2011;6(11):e28204.
atlas. Cancer Discov. 2011;1(3):193. 2017;19(1):4–23. 238. Stransky N, Cerami E, Schalm S, Kim JL, Lengauer
198. Kandoth C, McLellan MD, Vandin F, et al. Muta- 220. Shaffer LG, Slovak ML, Campbell LJ. ISCN 2009: C. The landscape of kinase fusions in cancer. Nat
tional landscape and significance across 12 major an international system for human cytogenetic Commun. 2014;5:4846.
cancer types. Nature. 2013;502(7471):333–339. nomenclature. Karger; 2009. 239. Levin JZ, Berger MF, Adiconis X, et al. Targeted
199. Cancer Genome Atlas Research Network. Genomic clas- 221. Rikova K, Guo A, Zeng Q, et al. Global survey next-generation sequencing of a cancer transcriptome
sification of cutaneous melanoma. Cell. 2015;161(7): of phosphotyrosine signaling identifies oncogenic enhances detection of sequence variants and novel
1681–1696. kinases in lung cancer. Cell. 2007;131(6):1190– fusion transcripts. Genome Biol. 2009;10(10):R115.
200. Gupta S, Artomov M, Goggins W, Daly M, Tsao H. 1203. 240. Pfarr N, Stenzinger A, Penzel R, et al. High-
Gender disparity and mutation burden in metastatic 222. Takeuchi K, Choi YL, Togashi Y, et al. KIF5B-ALK, throughput diagnostic profiling of clinically action-
melanoma. J Natl Cancer Inst. 2015;107(11). a novel fusion oncokinase identified by an immu- able gene fusions in lung cancer. Genes Chromosomes
201. Cancer Genome Atlas Research Network. Compre- nohistochemistry-based diagnostic system for ALK- Cancer. 2016;55(1):30–44.
hensive molecular profiling of lung adenocarcinoma. positive lung cancer. Clin Cancer Res. 2009;15(9): 241. Beadling C, Wald AI, Warrick A, et al. A Multiplexed
Nature. 2014;511(7511):543–550. 3143–3149. amplicon approach for detecting gene fusions by
202. Comprehensive molecular portraits of human breast 223. Togashi Y, Soda M, Sakata S, et al. KLC1-ALK: next-generation sequencing. J Mol Diagn. 2016;18(2):
tumours. Nature. 2012;490(7418):61–70. a novel fusion in lung cancer identified using a 165–175.
203. Yates LR, Campbell PJ. Evolution of the cancer formalin-fixed paraffin-embedded tissue only. PLoS 242. Zheng Z, Liebers M, Zhelyazkova B, et al. Anchored
genome. Nat Rev Genet. 2012;13(11):795–806. ONE. 2012;7(2):e31323. multiplex PCR for targeted next-generation sequenc-
204. Burrell RA, McGranahan N, Bartek J, Swanton C. 224. Jung Y, Kim P, Keum J, et al. Discovery of ALK- ing. Nat Med. 2014;20(12):1479–1484.
The causes and consequences of genetic heterogene- PTPN3 gene fusion from human non-small cell 243. Shim HS, Kenudson M, Zheng Z, et al. Unique
ity in cancer evolution. Nature. 2013;501(7467): lung carcinoma cell line using next generation RNA genetic and survival characteristics of invasive
338–345. sequencing. Genes Chromosomes Cancer. 2012;51(6): mucinous adenocarcinoma of the lung. J Thorac
205. McGranahan N, Swanton C. Biological and 590–597. Oncol. 2015;10(8):1156–1162.
therapeutic impact of intratumor heterogeneity in 225. Camidge DR, Kono SA, Flacco A, et al. Optimizing 244. Suehara Y, Arcila M, Wang L, et al. Identification
cancer evolution. Cancer Cell. 2015;27(1):15–26. the detection of lung cancer patients harboring ana- of KIF5B-RET and GOPC-ROS1 fusions in lung
206. de Bruin EC, McGranahan N, Mitter R, et al. plastic lymphoma kinase (ALK) gene rearrangements adenocarcinomas through a comprehensive mRNA-
Spatial and temporal diversity in genomic instability potentially suitable for ALK inhibitor treatment. based screen for tyrosine kinase fusions. Clin Cancer
processes defines lung cancer evolution. Science. Clin Cancer Res. 2010;16(22):5581–5590. Res. 2012;18(24):6599–6608.
2014;346(6206):251–256. 226. Kim H, Yoo SB, Choe JY, et al. Detection of ALK 245. Greene GL, Nolan C, Engler JP, Jensen EV. Mono-
207. Zhang J, Fujimoto J, Zhang J, et al. Intratumor gene rearrangement in non-small cell lung cancer: clonal antibodies to human estrogen receptor. Proc
heterogeneity in localized lung adenocarcinomas a comparison of fluorescence in situ hybridization Natl Acad Sci USA. 1980;77(9):5115–5119.
delineated by multiregion sequencing. Science. and chromogenic in situ hybridization with cor- 246. Greene GL, Fitch FW, Jensen EV. Monoclonal anti-
2014;346(6206):256–259. relation of ALK protein expression. J Thorac Oncol. bodies to estrophilin: probes for the study of estrogen
208. de Bruin EC, McGranahan N, Swanton C. Analysis 2011;6(8):1359–1366. receptors. Proc Natl Acad Sci USA. 1980;77(1):
of intratumor heterogeneity unravels lung cancer 227. Yoshida A, Tsuta K, Nitta H, et al. Bright-field 157–161.
evolution. Mol Cell Oncol. 2015;2(3):e985549. dual-color chromogenic in situ hybridization for 247. Shimada A, Kimura S, Abe K, et al. Immunocyto-
208a. Jamal-Hanjani M, Wilson GA, McGranahan diagnosing echinoderm microtubule-associated chemical staining of estrogen receptor in paraffin
N, et al. Tracking the evolution of non-small-cell protein-like 4-anaplastic lymphoma kinase-positive sections of human breast cancer by use of monoclonal
lung cancer. N Engl J Med. 2017;376(22):2109– lung adenocarcinomas. J Thorac Oncol. 2011;6(10): antibody: comparison with that in frozen sections.
2121. 1677–1686. Proc Natl Acad Sci USA. 1985;82(14):4803–
209. Blume-Jensen P, Hunter T. Oncogenic kinase 228. Kwak EL, Bang YJ, Camidge DR, et al. Anaplastic 4807.
signalling. Nature. 2001;411(6835):355–365. lymphoma kinase inhibition in non-small-cell lung 248. Poulsen HS, Ozzello L, King WJ, Greene GL. The
210. Goodwin S, McPherson JD, McCombie WR. cancer. N Engl J Med. 2010;363(18):1693–1703. use of monoclonal antibodies to estrogen receptors
Coming of age: ten years of next-generation sequenc- 229. Shaw AT, Yeap BY, Solomon BJ, et al. Effect of (ER) for immunoperoxidase detection of ER in
ing technologies. Nat Rev Genet. 2016;17(6):333– crizotinib on overall survival in patients with paraffin sections of human breast cancer tissue.
351. advanced non-small-cell lung cancer harbouring J Histochem Cytochem. 1985;33(2):87–92.
Pathology, Biomarkers, and Molecular Diagnostics  •  CHAPTER 15 253.e5
253.e5

249. Harvey JM, Clark GM, Osborne CK, Allred DC. 267. Schreiber RD, Old LJ, Smyth MJ. Cancer immu- 289. Zeng DQ, Yu YF, Ou QY, et al. Prognostic and
Estrogen receptor status by immunohistochemistry noediting: integrating immunity’s roles in cancer sup- predictive value of tumor-infiltrating lymphocytes
is superior to the ligand-binding assay for predicting pression and promotion. Science. 2011;331(6024): for clinical therapeutic research in patients with
response to adjuvant endocrine therapy in breast 1565–1570. non-small cell lung cancer. Oncotarget. 2016;7(12):
cancer. J Clin Oncol. 1999;17(5):1474–1481. 268. Hodi FS, O’Day SJ, McDermott DF, et al. Improved 13765–13781.
250. McCarty KS Jr, Szabo E, Flowers JL, et al. Use of a survival with ipilimumab in patients with metastatic 290. Thomas NE, Busam KJ, From L, et al. Tumor-
monoclonal anti-estrogen receptor antibody in the melanoma. N Engl J Med. 2010;363(8):711–723. infiltrating lymphocyte grade in primary melanomas
immunohistochemical evaluation of human tumors. 269. Robert C, Schachter J, Long GV, et al. Pembroli- is independently associated with melanoma-specific
Cancer Res. 1986;46(8 suppl):4244s–4248s. zumab versus ipilimumab in advanced melanoma. survival in the population-based genes, environment
251. Bartlett JM, Brookes CL, Robson T, et al. Estrogen N Engl J Med. 2015;372(26):2521–2532. and melanoma study. J Clin Oncol. 2013;31(33):
receptor and progesterone receptor as predictive 270. Larkin J, Chiarion-Sileni V, Gonzalez R, et al. Com- 4252–4259.
biomarkers of response to endocrine therapy: a bined nivolumab and ipilimumab or monotherapy 291. Huh JW, Lee JH, Kim HR. Prognostic significance
prospectively powered pathology study in the in untreated melanoma. N Engl J Med. 2015;373(1): of tumor-infiltrating lymphocytes for patients with
Tamoxifen and Exemestane Adjuvant Multinational 23–34. colorectal cancer. Arch Surg. 2012;147(4):366–372.
trial. J Clin Oncol. 2011;29(12):1531–1538. 271. Garon EB, Rizvi NA, Hui R, et al. Pembrolizumab 292. Lewin KJ, Riddell RH, Weinstein WM. Mesen-
252. Slamon DJ, Clark GM, Wong SG, Levin WJ, Ullrich for the treatment of non-small-cell lung cancer. N chymal tumors. In: Gastrointestinal Pathology and
A, McGuire WL. Human breast cancer: correlation of Engl J Med. 2015;372(21):2018–2028. Its Clinical Implications. Vol. 1. 1st ed. New York,
relapse and survival with amplification of the HER-2/ 272. Brahmer J, Reckamp KL, Baas P, et al. Nivolumab Tokyo: Igaku-Shoin; 1992:284–341.
neu oncogene. Science. 1987;235(4785):177–182. versus docetaxel in advanced squamous-cell non- 293. van de Rijn M, Hendrickson MR, Rouse RV. CD34
253. Slamon DJ, Godolphin W, Jones LA, et al. Studies of small-cell lung cancer. N Engl J Med. 2015;373(2): expression by gastrointestinal tract stromal tumors.
the HER-2/neu proto-oncogene in human breast and 123–135. Hum Pathol. 1994;25(8):766–771.
ovarian cancer. Science. 1989;244(4905):707–712. 273. Borghaei H, Paz-Ares L, Horn L, et al. Nivolumab 294. Hirota S, Isozaki K, Moriyama Y, et al. Gain-of-
254. Lewis GD, Figari I, Fendly B, et al. Differential versus docetaxel in advanced nonsquamous non- function mutations of c-kit in human gastrointestinal
responses of human tumor cell lines to anti- small-cell lung cancer. N Engl J Med. 2015;373(17): stromal tumors. Science. 1998;279(5350):577–580.
p185HER2 monoclonal antibodies. Cancer Immunol 1627–1639. 295. Corless CL, Barnett CM, Heinrich MC. Gastro-
Immunother. 1993;37(4):255–263. 274. Sharma P, Allison JP. Immune checkpoint targeting intestinal stromal tumours: origin and molecular
255. Shepard HM, Lewis GD, Sarup JC, et al. Monoclo- in cancer therapy: toward combination strategies oncology. Nat Rev Cancer. 2011;11(12):865–878.
nal antibody therapy of human cancer: taking the with curative potential. Cell. 2015;161(2):205–214. 296. Lasota J, Miettinen M. Clinical significance of onco-
HER2 protooncogene to the clinic. J Clin Immunol. 275. Schumacher TN, Schreiber RD. Neoantigens in cancer genic KIT and PDGFRA mutations in gastrointestinal
1991;11(3):117–127. immunotherapy. Science. 2015;348(6230):69–74. stromal tumours. Histopathology. 2008;53(3):
256. Slamon DJ, Leyland-Jones B, Shak S, et al. Use of 276. Snyder A, Makarov V, Merghoub T, et al. Genetic 245–266.
chemotherapy plus a monoclonal antibody against basis for clinical response to CTLA-4 blockade in 297. Martin-Broto J, Rubio L, Alemany R, Lopez-
HER2 for metastatic breast cancer that overexpresses melanoma. N Engl J Med. 2014;371(23):2189–2199. Guerrero JA. Clinical implications of KIT and
HER2. N Engl J Med. 2001;344(11):783–792. 277. Rizvi NA, Hellmann MD, Snyder A, et al. Cancer PDGFRA genotyping in GIST. Clin Transl Oncol.
257. Wolff AC, Hammond ME, Schwartz JN, et al. immunology. Mutational landscape determines 2010;12(10):670–676.
American Society of Clinical Oncology/College of sensitivity to PD-1 blockade in non-small cell lung 298. van de Rijn M, Lombard CM, Rouse RV. Expression
American Pathologists guideline recommendations cancer. Science. 2015;348(6230):124–128. of CD34 by solitary fibrous tumors of the pleura,
for human epidermal growth factor receptor 2 testing 278. Cancer Genome Atlas N. Comprehensive molecular mediastinum, and lung. Am J Surg Pathol. 1994;18(8):
in breast cancer. J Clin Oncol. 2007;25(1):118–145. characterization of human colon and rectal cancer. 814–820.
258. Cuzick J, Dowsett M, Pineda S, et al. Prognostic Nature. 2012;487(7407):330–337. 299. Hirota S, Ohashi A, Nishida T, et al. Gain-of-
value of a combined estrogen receptor, progesterone 279. Eshleman JR, Lang EZ, Bowerfind GK, et al. function mutations of platelet-derived growth
receptor, Ki-67, and human epidermal growth factor Increased mutation rate at the hprt locus accom- factor receptor alpha gene in gastrointestinal stromal
receptor 2 immunohistochemical score and com- panies microsatellite instability in colon cancer. tumors. Gastroenterology. 2003;125(3):660–667.
parison with the Genomic Health recurrence score Oncogene. 1995;10(1):33–37. 300. Ng EH, Pollock RE, Munsell MF, Atkinson EN,
in early breast cancer. J Clin Oncol. 2011;29(32): 280. Timmermann B, Kerick M, Roehr C, et al. Somatic Romsdahl MM. Prognostic factors influencing
4273–4278. mutation profiles of MSI and MSS colorectal survival in gastrointestinal leiomyosarcomas. Implica-
259. Bartlett JM, Bloom KJ, Piper T, et al. Mammostrat cancer identified by whole exome next generation tions for surgical management and staging. Ann
as an immunohistochemical multigene assay for sequencing and bioinformatics analysis. PLoS ONE. Surg. 1992;215(1):68–77.
prediction of early relapse risk in the tamoxifen 2010;5(12):e15661. 301. Dematteo RP, Heinrich MC, El-Rifai WM, Demetri
versus exemestane adjuvant multicenter trial pathol- 281. Le DT, Uram JN, Wang H, et al. PD-1 blockade G. Clinical management of gastrointestinal stromal
ogy study. J Clin Oncol. 2012;30(36):4477–4484. in tumors with mismatch-repair deficiency. N Engl tumors: before and after STI-571. Hum Pathol. 2002;
260. Foulkes WD, Smith IE, Reis-Filho JS. Triple- J Med. 2015;372(26):2509–2520. 33(5):466–477.
negative breast cancer. N Engl J Med. 2010;363(20): 282. Dudley JC, Lin MT, Le DT, Eshleman JR. Microsat- 302. Heinrich MC, Griffith DJ, Druker BJ, Wait CL, Ott
1938–1948. ellite instability as a biomarker for PD-1 blockade. KA, Zigler AJ. Inhibition of c-kit receptor tyrosine
261. Perou CM, Sorlie T, Eisen MB, et al. Molecular portraits Clin Cancer Res. 2016;22(4):813–820. kinase activity by STI 571, a selective tyrosine kinase
of human breast tumours. Nature. 2000;406(6797): 283. Gibney GT, Weiner LM, Atkins MB. Predictive inhibitor. Blood. 2000;96(3):925–932.
747–752. biomarkers for checkpoint inhibitor-based immu- 303. Verweij J, Casali PG, Zalcberg J, et al. Progression-
262. Dalerba P, Sahoo D, Paik S, et al. CDX2 as a notherapy. Lancet Oncol. 2016;17(12):e542–e551. free survival in gastrointestinal stromal tumours
prognostic biomarker in stage II and stage III colon 284. Sacher AG, Gandhi L. Biomarkers for the clinical with high-dose imatinib: randomised trial. Lancet.
cancer. N Engl J Med. 2016;374(3):211–222. use of PD-1/PD-L1 inhibitors in non-small-cell lung 2004;364(9440):1127–1134.
263. Pirker R, Pereira JR, von Pawel J, et al. EGFR cancer: a review. JAMA Oncol. 2016;2(9):1217– 304. Blanke CD, Rankin C, Demetri GD, et al. Phase
expression as a predictor of survival for first-line che- 1222. III randomized, intergroup trial assessing imatinib
motherapy plus cetuximab in patients with advanced 285. Gandini S, Massi D, Mandala M. PD-L1 expres- mesylate at two dose levels in patients with unresect-
non-small-cell lung cancer: analysis of data from sion in cancer patients receiving anti PD-1/PD-L1 able or metastatic gastrointestinal stromal tumors
the phase 3 FLEX study. Lancet Oncol. 2012;13(1): antibodies: a systematic review and meta-analysis. expressing the kit receptor tyrosine kinase: S0033.
33–42. Crit Rev Oncol Hematol. 2016;100:88–98. J Clin Oncol. 2008;26(4):626–632.
264. Lantuejoul S, Varella-Garcia M, Thunnissen E, 286. Rimm DL, Han G, Taube JM, et al. A prospective, 305. Demetri GD, von Mehren M, Blanke CD, et al.
Yatabe Y. Comparison of different assay platforms multi-institutional, pathologist-based assessment of 4 Efficacy and safety of imatinib mesylate in advanced
of ALK testing. In: Tsao MS, Hirsch FR, Yatabe Y, immunohistochemistry assays for PD-L1 expression gastrointestinal stromal tumors. N Engl J Med. 2002;
eds. IASLC Atlas of ALK and ROS1 Testing in Lung in non-small cell lung cancer. JAMA Oncol. 2017. 347(7):472–480.
Cancer. 2nd ed. North Fort Myers, FL: Editorial 287. Hirsch FR, McElhinny A, Stanforth D, et al. PD-L1 306. Corless CL, Schroeder A, Griffith D, et al. PDGFRA
Rx Press; 2016:73–83. Immunohistochemistry assays for lung cancer: results mutations in gastrointestinal stromal tumors: fre-
265. Tsao MS, Hirsch FR, Yatabe Y. IASLC Atlas of ALK from phase 1 of the Blueprint PD-L1 IHC Assay quency, spectrum and in vitro sensitivity to imatinib.
and ROS1 Testing in Lung Cancer. 2nd ed. North Comparison Project. J Thorac Oncol. 2017;12(2): J Clin Oncol. 2005;23(23):5357–5364.
Fort Myers, FL: Editorial Rx Press; 2016. 208–222. 307. Weisberg E, Wright RD, Jiang J, et al. Effects of
266. Sholl LM, Sun H, Butaney M, et al. ROS1 288. Kerr KM, Tsao MS, Nicholson AG, et al. Pro- PKC412, nilotinib, and imatinib against GIST-
immunohistochemistry for detection of ROS1- grammed death-ligand 1 immunohistochemistry associated PDGFRA mutants with differential
rearranged lung adenocarcinomas. Am J Surg Pathol. in lung cancer: in what state is this art? J Thorac imatinib sensitivity. Gastroenterology. 2006;131(6):
2013;37(9):1441–1449. Oncol. 2015;10(7):985–989. 1734–1742.
253.e6 Part I: Science and Clinical Oncology

308. Sepulveda AR, Hamilton SR, Allegra CJ, et al. 326. Saridaki Z, Papadatos-Pastos D, Tzardi M, et al. BRAF 344. Bean J, Brennan C, Shih JY, et al. MET amplifica-
Molecular biomarkers for the evaluation of colorectal mutations, microsatellite instability status and cyclin tion occurs with or without T790M mutations in
cancer: guideline from the American Society for D1 expression predict metastatic colorectal patients’ EGFR mutant lung tumors with acquired resistance
Clinical Pathology, College of American Pathologists, outcome. Br J Cancer. 2010;102(12):1762–1768. to gefitinib or erlotinib. Proc Natl Acad Sci USA.
Association for Molecular Pathology, and American 327. French AJ, Sargent DJ, Burgart LJ, et al. Prognostic 2007;104(52):20932–20937.
Society of Clinical Oncology. Arch Pathol Lab Med. significance of defective mismatch repair and BRAF 345. Nguyen KS, Kobayashi S, Costa DB. Acquired resis-
2017;141(5):625–657. V600E in patients with colon cancer. Clin Cancer tance to epidermal growth factor receptor tyrosine
309. Jimeno A, Messersmith WA, Hirsch FR, Franklin Res. 2008;14(11):3408–3415. kinase inhibitors in non-small-cell lung cancers
WA, Eckhardt SG. KRAS mutations and sensitivity 328. Weisenberger DJ, Siegmund KD, Campan M, et al. dependent on the epidermal growth factor receptor
to epidermal growth factor receptor inhibitors in CpG island methylator phenotype underlies sporadic pathway. Clin Lung Cancer. 2009;10(4):281–289.
colorectal cancer: practical application of patient microsatellite instability and is tightly associated with 346. Suda K, Murakami I, Katayama T, et al. Reciprocal
selection. J Clin Oncol. 2009;27(7):1130–1136. BRAF mutation in colorectal cancer. Nat Genet. and complementary role of MET amplification and
310. Andreyev HJ, Norman AR, Cunningham D, et al. 2006;38(7):787–793. EGFR T790M mutation in acquired resistance to
Kirsten ras mutations in patients with colorectal cancer: 329. Wang L, Cunningham JM, Winters JL, et al. BRAF kinase inhibitors in lung cancer. Clin Cancer Res.
the ‘RASCAL II’ study. Br J Cancer. 2001;85(5): mutations in colon cancer are not likely attributable 2010;16(22):5489–5498.
692–696. to defective DNA mismatch repair. Cancer Res. 2003; 347. Pao W, Miller VA, Politi KA, et al. Acquired
311. Amado RG, Wolf M, Peeters M, et al. Wild-type 63(17):5209–5212. resistance of lung adenocarcinomas to gefitinib or
KRAS is required for panitumumab efficacy in 330. Di Nicolantonio F, Martini M, Molinari F, et al. erlotinib is associated with a second mutation in the
patients with metastatic colorectal cancer. J Clin Wild-type BRAF is required for response to panitu- EGFR kinase domain. PLoS Med. 2005;2(3):e73.
Oncol. 2008;26(10):1626–1634. mumab or cetuximab in metastatic colorectal cancer. 348. Yasuda H, Kobayashi S, Costa DB. EGFR exon 20
312. Jonker DJ, O’Callaghan CJ, Karapetis CS, et al. J Clin Oncol. 2008;26(35):5705–5712. insertion mutations in non-small-cell lung cancer:
Cetuximab for the treatment of colorectal cancer. 331. Laurent-Puig P, Cayre A, Manceau G, et al. Analysis preclinical data and clinical implications. Lancet
N Engl J Med. 2007;357(20):2040–2048. of PTEN, BRAF, and EGFR status in determining Oncol. 2012;13(1):e23–e31.
313. Karapetis CS, Khambata-Ford S, Jonker DJ, et al. K- benefit from cetuximab therapy in wild-type KRAS 349. Sequist LV, Waltman BA, Dias-Santagata D, et al.
ras mutations and benefit from cetuximab in advanced metastatic colon cancer. J Clin Oncol. 2009;27(35): Genotypic and histological evolution of lung cancers
colorectal cancer. N Engl J Med. 2008;359(17): 5924–5930. acquiring resistance to EGFR inhibitors. Sci Transl
1757–1765. 332. Sartore-Bianchi A, Di Nicolantonio F, Nichelatti Med. 2011;3(75):75ra26.
314. Van Cutsem E, Kohne CH, Hitre E, et al. Cetuximab M, et al. Multi-determinants analysis of molecular 350. Morgillo F, Della Corte CM, Fasano M, Ciardiello
and chemotherapy as initial treatment for metastatic alterations for predicting clinical benefit to EGFR- F. Mechanisms of resistance to EGFR-targeted drugs:
colorectal cancer. N Engl J Med. 2009;360(14): targeted monoclonal antibodies in colorectal cancer. lung cancer. ESMO Open. 2016;1(3):e000060.
1408–1417. PLoS ONE. 2009;4(10):e7287. 351. Ahrendt SA, Decker PA, Alawi EA, et al. Cigarette
315. Bokemeyer C, Bondarenko I, Makhson A, et al. Fluo- 333. Prahallad A, Sun C, Huang S, et al. Unresponsive- smoking is strongly associated with mutation of the
rouracil, leucovorin, and oxaliplatin with and without ness of colon cancer to BRAF(V600E) inhibition K-ras gene in patients with primary adenocarcinoma
cetuximab in the first-line treatment of metastatic through feedback activation of EGFR. Nature. of the lung. Cancer. 2001;92(6):1525–1530.
colorectal cancer. J Clin Oncol. 2009;27(5):663– 2012;483(7387):100–103. 352. Riely GJ, Kris MG, Rosenbaum D, et al. Frequency
671. 334. Comprehensive molecular characterization of human and distinctive spectrum of KRAS mutations in never
316. De Roock W, Jonker DJ, Di Nicolantonio F, et al. colon and rectal cancer. Nature. 2012;487(7407): smokers with lung adenocarcinoma. Clin Cancer
Association of KRAS p.G13D mutation with 330–337. Res. 2008;14(18):5731–5734.
outcome in patients with chemotherapy-refractory 335. Travis WD, Brambilla E, Noguchi M, et al. 353. Benesova L, Minarik M, Jancarikova D, Belsanova B,
metastatic colorectal cancer treated with cetuximab. International Association for the Study of Lung Pesek M. Multiplicity of EGFR and KRAS mutations
JAMA. 2010;304(16):1812–1820. Cancer/American Thoracic Society/European in non-small cell lung cancer (NSCLC) patients
317. Davies H, Bignell GR, Cox C, et al. Mutations Respiratory Society international multidisciplinary treated with tyrosine kinase inhibitors. Anticancer
of the BRAF gene in human cancer. Nature. classification of lung adenocarcinoma. J Thorac Res. 2010;30(5):1667–1671.
2002;417(6892):949–954. Oncol. 2011;6(2):244–285. 354. Roberts PJ, Stinchcombe TE, Der CJ, Socinski MA.
318. Vaughn CP, Zobell SD, Furtado LV, Baker CL, 336. Cohen S. Isolation of a mouse submaxillary gland Personalized medicine in non-small-cell lung cancer:
Samowitz WS. Frequency of KRAS, BRAF, and protein accelerating incisor eruption and eyelid is KRAS a useful marker in selecting patients for
NRAS mutations in colorectal cancer. Genes opening in the new-born animal. J Biol Chem. epidermal growth factor receptor-targeted therapy?
Chromosomes Cancer. 2011;50(5):307–312. 1962;237:1555–1562. J Clin Oncol. 2010;28(31):4769–4777.
319. Roth AD, Tejpar S, Delorenzi M, et al. Prognostic 337. Carpenter G, Lembach KJ, Morrison MM, Cohen 355. Soda M, Choi YL, Enomoto M, et al. Identification
role of KRAS and BRAF in stage II and III resected S. Characterization of the binding of 125-I-labeled of the transforming EML4-ALK fusion gene in
colon cancer: results of the translational study on epidermal growth factor to human fibroblasts. J Biol non-small-cell lung cancer. Nature. 2007;448(7153):
the PETACC-3, EORTC 40993, SAKK 60-00 trial. Chem. 1975;250(11):4297–4304. 561–566.
J Clin Oncol. 2010;28(3):466–474. 338. Ullrich A, Coussens L, Hayflick JS, et al. Human epi- 356. Rodig SJ, Mino-Kenudson M, Dacic S, et al. Unique
320. De Roock W, Claes B, Bernasconi D, et al. Effects dermal growth factor receptor cDNA sequence and clinicopathologic features characterize ALK-rear-
of KRAS, BRAF, NRAS, and PIK3CA mutations aberrant expression of the amplified gene in A431 ranged lung adenocarcinoma in the western popula-
on the efficacy of cetuximab plus chemotherapy in epidermoid carcinoma cells. Nature. 1984;309(5967): tion. Clin Cancer Res. 2009;15(16):5216–5223.
chemotherapy-refractory metastatic colorectal cancer: 418–425. 357. Sasaki T, Rodig SJ, Chirieac LR, Janne PA. The
a retrospective consortium analysis. Lancet Oncol. 339. Franklin WA, Veve R, Hirsch FR, Helfrich BA, biology and treatment of EML4-ALK non-small
2010;11(8):753–762. Bunn PA Jr. Epidermal growth factor receptor family cell lung cancer. Eur J Cancer. 2010;46(10):1773–
321. Ogino S, Shima K, Meyerhardt JA, et al. Predictive in lung cancer and premalignancy. Semin Oncol. 1780.
and prognostic roles of BRAF mutation in stage III 2002;29(1 suppl 4):3–14. 358. Takahashi T, Sonobe M, Kobayashi M, et al.
colon cancer: results from intergroup trial CALGB 340. Hirsch FR, Varella-Garcia M, Bunn PA Jr, et al. Clinicopathologic features of non-small-cell lung
89803. Clin Cancer Res. 2012;18(3):890–900. Epidermal growth factor receptor in non-small- cancer with EML4-ALK fusion gene. Ann Surg
322. Yamauchi M, Lochhead P, Morikawa T, et al. cell lung carcinomas: correlation between gene Oncol. 2010;17(3):889–897.
Colorectal cancer: a tale of two sides or a continuum? copy number and protein expression and impact 359. Pillai RN, Ramalingam SS. The biology and clinical
Gut. 2012;61(6):794–797. on prognosis. J Clin Oncol. 2003;21(20):3798– features of non-small cell lung cancers with EML4-
323. Yamauchi M, Morikawa T, Kuchiba A, et al. Assess- 3807. ALK translocation. Curr Oncol Rep. 2012;14(2):
ment of colorectal cancer molecular features along 341. Paez JG, Janne PA, Lee JC, et al. EGFR mutations 105–110.
bowel subsites challenges the conception of distinct in lung cancer: correlation with clinical response 360. Soda M, Takada S, Takeuchi K, et al. A mouse model
dichotomy of proximal versus distal colorectum. to gefitinib therapy. Science. 2004;304(5676): for EML4-ALK-positive lung cancer. Proc Natl Acad
Gut. 2012;61(6):847–854. 1497–1500. Sci USA. 2008;105(50):19893–19897.
324. Samowitz WS, Sweeney C, Herrick J, et al. Poor 342. Lynch TJ, Bell DW, Sordella R, et al. Activating 361. Sanders HR, Li HR, Bruey JM, et al. Exon scanning
survival associated with the BRAF V600E mutation mutations in the epidermal growth factor receptor by reverse transcriptase-polymerase chain reaction for
in microsatellite-stable colon cancers. Cancer Res. underlying responsiveness of non-small-cell lung detection of known and novel EML4-ALK fusion
2005;65(14):6063–6069. cancer to gefitinib. N Engl J Med. 2004;350(21): variants in non-small cell lung cancer. Cancer Genet.
325. Ogino S, Nosho K, Kirkner GJ, et al. CpG island 2129–2139. 2011;204(1):45–52.
methylator phenotype, microsatellite instability, 343. Mok TS, Wu YL, Thongprasert S, et al. Gefitinib or 362. Lin JJ, Riely GJ, Shaw AT. Targeting ALK: precision
BRAF mutation and clinical outcome in colon carboplatin-paclitaxel in pulmonary adenocarcinoma. medicine takes on drug resistance. Cancer Discov.
cancer. Gut. 2009;58(1):90–96. N Engl J Med. 2009;361(10):947–957. 2017;7(2):137–155.
Pathology, Biomarkers, and Molecular Diagnostics  •  CHAPTER 15 253.e7
253.e7

363. Villaruz LC, Socinski MA, Abberbock S, et al. 383. Curtin JA, Busam K, Pinkel D, Bastian BC. Somatic glioblastoma from a distinct cell of origin. J Clin
Clinicopathologic features and outcomes of patients activation of KIT in distinct subtypes of melanoma. Oncol. 2011;29(34):4482–4490.
with lung adenocarcinomas harboring BRAF muta- J Clin Oncol. 2006;24(26):4340–4346. 405. Ichimura K, Pearson DM, Kocialkowski S, et al.
tions in the Lung Cancer Mutation Consortium. 384. Beadling C, Jacobson-Dunlop E, Hodi FS, et al. IDH1 mutations are present in the majority
Cancer. 2015;121(3):448–456. KIT gene mutations and copy number in melanoma of common adult gliomas but rare in primary
364. Nguyen-Ngoc T, Bouchaab H, Adjei AA, Peters subtypes. Clin Cancer Res. 2008;14(21):6821– glioblastomas. Neuro Oncol. 2009;11(4):341–347.
S. BRAF alterations as therapeutic targets in non- 6828. 406. Balss J, Meyer J, Mueller W, Korshunov A, Hartmann
small-cell lung cancer. J Thorac Oncol. 2015;10(10): 385. Carvajal RD, Antonescu CR, Wolchok JD, et al. C, von Deimling A. Analysis of the IDH1 codon
1396–1403. KIT as a therapeutic target in metastatic melanoma. 132 mutation in brain tumors. Acta Neuropathol.
365. Ou SI, Schrock AB, Bocharov EV, et al. HER2 JAMA. 2011;305(22):2327–2334. 2008;116(6):597–602.
transmembrane domain (TMD) mutations (V659/ 386. Guo J, Si L, Kong Y, et al. Phase II, open-label, 407. Nobusawa S, Watanabe T, Kleihues P, Ohgaki
G660) that stabilize homo- and heterodimerization single-arm trial of imatinib mesylate in patients with H. IDH1 mutations as molecular signature and
are rare oncogenic drivers in lung adenocarcinoma metastatic melanoma harboring c-Kit mutation or predictive factor of secondary glioblastomas. Clin
that respond to afatinib. J Thorac Oncol. 2017; amplification. J Clin Oncol. 2011;29(21):2904–2909. Cancer Res. 2009;15(19):6002–6007.
12(3):446–457. 387. Marshall CJ, Hall A, Weiss RA. A transforming 408. Capper D, Weissert S, Balss J, et al. Characteriza-
366. Vaishnavi A, Capelletti M, Le AT, et al. Oncogenic gene present in human sarcoma cell lines. Nature. tion of R132H mutation-specific IDH1 antibody
and drug-sensitive NTRK1 rearrangements in lung 1982;299(5879):171–173. binding in brain tumors. Brain Pathol. 2010;20(1):
cancer. Nat Med. 2013;19(11):1469–1472. 388. Hall A, Marshall CJ, Spurr NK, Weiss RA. 245–254.
367. Ohashi K, Sequist LV, Arcila ME, et al. Character- Identification of transforming gene in two human 409. von Deimling A, Louis DN, von Ammon K, Petersen
istics of lung cancers harboring NRAS mutations. sarcoma cell lines as a new member of the ras gene I, Wiestler OD, Seizinger BR. Evidence for a tumor
Clin Cancer Res. 2013;19(9):2584–2591. family located on chromosome 1. Nature. 1983; suppressor gene on chromosome 19q associated with
368. Rosell R, Karachaliou N. RET inhibitors for 303(5916):396–400. human astrocytomas, oligodendrogliomas, and mixed
patients with RET fusion-positive and RET wild- 389. Padua RA, Barrass N, Currie GA. A novel transform- gliomas. Cancer Res. 1992;52(15):4277–4279.
type non-small-cell lung cancer. Lancet Oncol. ing gene in a human malignant melanoma cell line. 410. Rosenberg JE, Lisle DK, Burwick JA, et al. Refined
2016;17(12):1623–1625. Nature. 1984;311(5987):671–673. deletion mapping of the chromosome 19q glioma
369. Shaw AT, Ou SH, Bang YJ, et al. Crizotinib in 390. Pollock PM, Harper UL, Hansen KS, et al. High tumor suppressor gene to the D19S412-STD
ROS1-rearranged non-small-cell lung cancer. N Engl frequency of BRAF mutations in nevi. Nat Genet. interval. Oncogene. 1996;13(11):2483–2485.
J Med. 2014;371(21):1963–1971. 2003;33(1):19–20. 411. Kraus JA, Koopmann J, Kaskel P, et al. Shared
370. Pao W, Girard N. New driver mutations in non-small- 391. Bauer J, Curtin JA, Pinkel D, Bastian BC. Congenital allelic losses on chromosomes 1p and 19q suggest
cell lung cancer. Lancet Oncol. 2011;12(2):175–180. melanocytic nevi frequently harbor NRAS muta- a common origin of oligodendroglioma and oligoas-
371. Aisner D, Sholl LM, Berry LD, et al. Effect of tions but no BRAF mutations. J Invest Dermatol. trocytoma. J Neuropathol Exp Neurol. 1995;54(1):
expanded genomic testing in lung adenocarcinoma 2007;127(1):179–182. 91–95.
(LUCA) on survival benefit: the Lung Cancer Muta- 392. Van Raamsdonk CD, Bezrookove V, Green G, et al. 412. Reifenberger J, Reifenberger G, Liu L, James
tion Consortium II (LCMC II) experience. J Clin Frequent somatic mutations of GNAQ in uveal CD, Wechsler W, Collins VP. Molecular genetic
Oncol. 2016;34(15_suppl):11510. melanoma and blue naevi. Nature. 2009;457(7229): analysis of oligodendroglial tumors shows prefer-
372. Ettinger DS, Wood DE, Aisner DL, et al. Non-Small 599–602. ential allelic deletions on 19q and 1p. Am J Pathol.
Cell Lung Cancer, Version 5.2017, NCCN Clinical 393. Van Raamsdonk CD, Griewank KG, Crosby MB, 1994;145(5):1175–1190.
Practice Guidelines in Oncology. J Natl Compr Canc et al. Mutations in GNA11 in uveal melanoma. N 413. Cairncross JG, Ueki K, Zlatescu MC, et al. Specific
Netw. 2017;15(4):504–535. Engl J Med. 2010;363(23):2191–2199. genetic predictors of chemotherapeutic response and
373. Woodman SE, Lazar AJ, Aldape KD, Davies MA. 394. Harbour JW, Onken MD, Roberson ED, et al. survival in patients with anaplastic oligodendroglio-
New strategies in melanoma: molecular testing in Frequent mutation of BAP1 in metastasizing uveal mas. J Natl Cancer Inst. 1998;90(19):1473–1479.
advanced disease. Clin Cancer Res. 2012;18(5): melanomas. Science. 2010;330(6009):1410–1413. 414. Fallon KB, Palmer CA, Roth KA, et al. Prognostic
1195–1200. 395. Wiesner T, Obenauf AC, Murali R, et al. Germline value of 1p, 19q, 9p, 10q, and EGFR-FISH analyses
374. Long GV, Menzies AM, Nagrial AM, et al. Prognostic mutations in BAP1 predispose to melanocytic in recurrent oligodendrogliomas. J Neuropathol Exp
and clinicopathologic associations of oncogenic tumors. Nat Genet. 2011;43(10):1018–1021. Neurol. 2004;63(4):314–322.
BRAF in metastatic melanoma. J Clin Oncol. 2011; 396. Cancer Genome Atlas Research Network, Brat 415. van den Bent MJ, Weller M, Wen PY, Kros JM,
29(10):1239–1246. DJ, Verhaak RG, et al. Comprehensive, integrative Aldape K, Chang S. A clinical perspective on the
375. Shinozaki M, Fujimoto A, Morton DL, Hoon DS. genomic analysis of diffuse lower-grade gliomas. N 2016 WHO brain tumor classification and routine
Incidence of BRAF oncogene mutation and clinical Engl J Med. 2015;372(26):2481–2498. molecular diagnostics. Neuro Oncol. 2017.
relevance for primary cutaneous melanomas. Clin 397. Brennan CW, Verhaak RG, McKenna A, et al. The 416. Lee J, Solomon DA, Tihan T. The role of histone
Cancer Res. 2004;10(5):1753–1757. somatic genomic landscape of glioblastoma. Cell. modifications and telomere alterations in the patho-
376. Edlundh-Rose E, Egyhazi S, Omholt K, et al. NRAS 2013;155(2):462–477. genesis of diffuse gliomas in adults and children.
and BRAF mutations in melanoma tumours in 398. Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, J Neurooncol. 2017;132(1):1–11.
relation to clinical characteristics: a study based on eds. WHO Classification of Tumours of the Central 417. Wu G, Broniscer A, McEachron TA, et al. Somatic
mutation screening by pyrosequencing. Melanoma Nervous System. Vol 4th ed. Lyon, France: IARC; histone H3 alterations in pediatric diffuse intrinsic
Res. 2006;16(6):471–478. 2016. Revised 4th edn 10–122 (IARC, 2016). pontine gliomas and non-brainstem glioblastomas.
377. Hocker T, Tsao H. Ultraviolet radiation and mela- 399. Viswanath P, Chaumeil MM, Ronen SM. Molecular Nat Genet. 2012;44(3):251–253.
noma: a systematic review and analysis of reported Imaging of Metabolic Reprograming in Mutant IDH 418. Khuong-Quang DA, Buczkowicz P, Rakopoulos
sequence variants. Hum Mutat. 2007;28(6):578–588. Cells. Front Oncol. 2016;6:60. P, et al. K27M mutation in histone H3.3 defines
378. Hauschild A, Grob JJ, Demidov LV, et al. Dabrafenib 400. Reuss DE, Sahm F, Schrimpf D, et al. ATRX and clinically and biologically distinct subgroups of
in BRAF-mutated metastatic melanoma: a multi- IDH1-R132H immunohistochemistry with subse- pediatric diffuse intrinsic pontine gliomas. Acta
centre, open-label, phase 3 randomised controlled quent copy number analysis and IDH sequencing Neuropathol. 2012;124(3):439–447.
trial. Lancet. 2012;380(9839):358–365. as a basis for an “integrated” diagnostic approach 419. Jiao Y, Killela PJ, Reitman ZJ, et al. Frequent
379. Sosman JA, Kim KB, Schuchter L, et al. Survival in for adult astrocytoma, oligodendroglioma and glio- ATRX, CIC, FUBP1 and IDH1 mutations refine
BRAF V600-mutant advanced melanoma treated with blastoma. Acta Neuropathol. 2015;129(1):133–146. the classification of malignant gliomas. Oncotarget.
vemurafenib. N Engl J Med. 2012;366(8):707–714. 401. Reuss DE, Mamatjan Y, Schrimpf D, et al. IDH 2012;3(7):709–722.
380. Long GV, Trefzer U, Davies MA, et al. Dabrafenib in mutant diffuse and anaplastic astrocytomas have 420. Liu XY, Gerges N, Korshunov A, et al. Frequent
patients with Val600Glu or Val600Lys BRAF-mutant similar age at presentation and little difference ATRX mutations and loss of expression in adult
melanoma metastatic to the brain (BREAK-MB): a in survival: a grading problem for WHO. Acta diffuse astrocytic tumors carrying IDH1/IDH2 and
multicentre, open-label, phase 2 trial. Lancet Oncol. Neuropathol. 2015;129(6):867–873. TP53 mutations. Acta Neuropathol. 2012;124(5):
2012. 402. Reuss DE, Kratz A, Sahm F, et al. Adult IDH 615–625.
381. Flaherty KT, Infante JR, Daud A, et al. Combined wild type astrocytomas biologically and clinically 421. Schindler G, Capper D, Meyer J, et al. Analysis
BRAF and MEK inhibition in melanoma with BRAF resolve into other tumor entities. Acta Neuropathol. of BRAF V600E mutation in 1,320 nervous
V600 mutations. N Engl J Med. 2012;367(18): 2015;130(3):407–417. system tumors reveals high mutation frequencies
1694–1703. 403. Yan H, Parsons DW, Jin G, et al. IDH1 and in pleomorphic xanthoastrocytoma, ganglioglioma
382. Flaherty KT, Fisher DE. New strategies in IDH2 mutations in gliomas. N Engl J Med. and extra-cerebellar pilocytic astrocytoma. Acta
metastatic melanoma: oncogene-defined taxonomy 2009;360(8):765–773. Neuropathol. 2011;121(3):397–405.
leads to therapeutic advances. Clin Cancer Res. 404. Lai A, Kharbanda S, Pope WB, et al. Evidence for 422. Kleinschmidt-DeMasters BK, Aisner DL, Birks
2011;17(15):4922–4928. sequenced molecular evolution of IDH1 mutant DK, Foreman NK. Epithelioid GBMs show a high
253.e8 Part I: Science and Clinical Oncology

percentage of BRAF V600E mutation. Am J Surg 426. Lassaletta A, Guerreiro Stucklin A, Ramaswamy V, 429. Wu J, Armstrong TS, Gilbert MR. Biology and man-
Pathol. 2013;37(5):685–698. et al. Profound clinical and radiological response to agement of ependymomas. Neuro Oncol. 2016;18(7):
423. Robinson GW, Orr BA, Gajjar A. Complete clini- BRAF inhibition in a 2-month-old diencephalic 902–913.
cal regression of a BRAF V600E-mutant pediatric child with hypothalamic/chiasmatic glioma. Pediatr 430. Ostrom QT, Gittleman H, Liao P, et al. CBTRUS
glioblastoma multiforme after BRAF inhibitor Blood Cancer. 2016;63(11):2038–2041. statistical report: primary brain and central nervous
therapy. BMC Cancer. 2014;14:258. 427. Olow A, Mueller S, Yang X, et al. BRAF Status system tumors diagnosed in the United States in
424. Lee EQ, Ruland S, LeBoeuf NR, Wen PY, Santagata in personalizing treatment approaches for 2007-2011. Neuro Oncol. 2014;16(suppl 4):iv1–iv63.
S. Successful treatment of a progressive BRAF pediatric gliomas. Clin Cancer Res. 2016;22(21): 431. Parker M, Mohankumar KM, Punchihewa C, et al.
V600E-mutated anaplastic pleomorphic xanthoas- 5312–5321. C11orf95-RELA fusions drive oncogenic NF-kappaB
trocytoma with vemurafenib monotherapy. J Clin 428. Miller C, Guillaume D, Dusenbery K, Clark HB, signalling in ependymoma. Nature. 2014;506(7489):
Oncol. 2016;34(10):e87–e89. Moertel C. Report of effective trametinib therapy 451–455.
425. Chamberlain MC. Salvage therapy with BRAF in 2 children with progressive hypothalamic optic 432. Pajtler KW, Witt H, Sill M, et al. Molecular
inhibitors for recurrent pleomorphic xanthoastro- pathway pilocytic astrocytoma: documentation of classification of ependymal tumors across all CNS
cytoma: a retrospective case series. J Neurooncol. volumetric response. J Neurosurg Pediatr. 2017;19(3): compartments, histopathological grades, and age
2013;114(2):237–240. 319–324. groups. Cancer Cell. 2015;27(5):728–743.
16  Imaging
Richard L. Wahl

S UMMARY OF K EY P OI N T S
• Noninvasive medical imaging often is being the main imaging method and complement diffusion contrast
essential to cancer management at clearly proven capable of reducing enhancement (DCE) MRI and appear
multiple times in the course of the cancer deaths when applied in the promising in assessing response to
illness. screening setting. A downside of tumor treatment. Multiparametric
• Imaging currently is used for screening imaging is the small but MRI sequences are being used more
screening to detect cancer, real risk of “overdiagnosis” and frequently to characterize the
characterizing lesions, performing “overtreatment”—detection and prostate to determine if and where
locoregional and systemic staging, aggressive treatment of cancers that biopsies should be performed
providing prognostic information, may not be life-threatening. and to help in conducting active
assessing response during and after • CT remains a cornerstone surveillance, delaying or reducing
therapy, restaging after treatment, technology for most oncologic prostatectomies.
performing follow-up of patients for imaging, and CT technology allowing • Bone scans using single-photon
recurrence, and precisely guiding for rapid-sequence angiography is methods (technetium-99m [99mTc]
biopsies and therapies such as finding new applications, as is methylene diphosphonate) remain
external beam or systemic radiation, three-dimensional reconstruction of the dominant procedure for detecting
brachytherapy, or thermal and other CT data sets. Screening data with suspected bone metastases;
ablations. CT colonography continues to however, the PET agent fluorine-18
• More invasive interventional improve, and in some studies it has (18F) sodium fluoride is increasingly
radiologic procedures also can guide been found to be comparable to applied since its approval by the US
and be used to monitor and deliver traditional colonoscopy for colon Food and Drug Administration (FDA).
vascular or intraluminal delivery of cancer screening. CT scanning for These techniques may be less
treatments such as radioactive lung cancer screening reduces lung sensitive for marrow involvement
microspheres, embolic materials, cancer death rates when applied to than MRI and other PET techniques
radiofrequency ablation or high-risk populations. The radiation for detecting bone metastases of
cryoablation, and therapeutic drugs. dose from CT is a concern, and many tumors.
• Imaging methods range from the major efforts to reduce the dose • PET and PET-CT technology using
18
traditional anatomic methods— from CT scanning have been F-fluorodeoxyglucose (FDG)
radiography, computed tomography implemented in newer CT systems. continues to grow in a wide variety
(CT), and ultrasonography—to the • MRI is the imaging tool of choice for of applications, and its use is
more functional methods of magnetic central nervous system, spinal, and becoming increasingly routine in the
resonance imaging (MRI) and nuclear musculoskeletal neoplasms, as well management of patients with cancer
medicine methods, including as for assessment of vascular and at varying states of the disease
positron emission tomography (PET), some hepatobiliary and pelvic process. PET is used with increasing
single-photon emission computed lesions. MRI also can be used frequency in the staging and
tomography (SPECT), and planar to detect breast cancers, follow-up of lung, colorectal, and
nuclear imaging. Hybrid methods especially in women with dense head and neck cancers, as well as
combining PET and CT, SPECT and breasts. Concerns regarding lymphomas and other types of
CT, and PET and MRI are growing in gadolinium-associated nephrogenic tumors, and is now a routine tool in
importance. Optical imaging is systemic fibrosis (NSF) have led to lymphoma management at several
promising but remains limited, by caution in the use of MRI contrast points in the disease. PET with
limited penetration of light through medium in patients with impaired non-FDG tracers is a promising
tissues, to superficial structures in renal function. There are also minor research area with growing clinical
most cases. concerns regarding gadolinium applications. There has been
• Plain films and mammography accumulation in the brain, although particular progress in imaging of
remain useful techniques, with these are of no known clinical prostate cancer with several imaging
mammography (including digital significance. Newer MRI techniques agents including FDA-approved C-11
mammography and tomosynthesis) such as diffusion imaging choline.

254
Imaging  •  CHAPTER 16 255

• The fusion of anatomic and radiopeptides binding to the fluoroscopy, and innovative MRI
functional images to create hybrid somatostatin receptors, and these systems can guide interventional
“anatomolecular images” with tumors can be treated with procedures such as thermal and
software or dedicated instruments radiopeptides labeled with cryotherapeutic lesion ablations.
such as PET-CT, SPECT-CT, or the lutetium-177 (177Lu), among other Bifunctional methods including a
newer PET-MRI devices also is therapeutic agents. radionuclide probe and an optical
seeing rapid growth in applications • In prostate cancer, available imaging probe may guide diagnosis and
in cancer imaging. Fully diagnostic methods remain suboptimal for surgery.
CT scans coupled with PET imaging detection of primary tumor and early • Highly specific probe-reporter
in the form of PET-CT often provide determination of local or systemic systems are being developed to
valuable composite imaging for tumor spread. MRI nodal contrast allow for optical and radionuclide
cancer management. PET-MRI is an agents are promising but not yet imaging of transfected gene
evolving technology, but fully routinely available, and magnetic biodistribution and function. These
integrated simultaneous systems are resonance spectroscopy has had approaches face major regulatory
being used much more widely. only limited success in the prostate. challenges when being translated to
• Imaging management for staging A variety of MRI sequences including humans.
lung cancer and characterizing T2 images, diffusion images, and • Combined anatomic and functional
solitary pulmonary nodules often DCE MRI appear to improve on information is being applied to allow
includes FDG-PET in addition to CT purely anatomic MRI approaches for for more precise planning of external
when the technology is available lesion detection and detection of beam radiation therapy, including
because PET-CT has higher extracapsular involvement. More intensity-modulated radiation therapy
accuracy in lung cancer standardized reading systems such (IMRT) and conformal therapy,
assessments compared with CT. as Prostate Imaging–Reporting and methods that potentially allow for
• Imaging management of suspected Data System (PI-RADS) may lead to increasing dose escalation and
recurrences of colorectal cancer, more consistent use of this minimization of toxicity to normal
head and neck cancer, lymphoma, technology to help differentiate tissues.
and many other cancers often now low- and high-risk tumors from each • Emerging imaging methods are
includes the use of PET in addition other and inform potential active proving increasingly useful in
to CT. Negative FDG-PET scans of surveillance choices. A variety of providing information on the
the neck after chemoradiotherapy innovative radiotracers for PET show physiology and molecular
can obviate the need for surgery in promise for detecting disease characteristics of lesions. This
patients with head and neck cancer. recurrence, and C-11 choline is now means that a multiparametric
Response criteria for FDG-avid FDA approved in the United States biologic imaging phenotype for
lymphomas are now mainly PET for use in prostate cancer, as is the tumors can be obtained and makes
based, and PET assessments of synthetic amino acid fluciclovine. it possible to display heterogeneities
68
treatment response are increasingly Ga- or 18F-radiolabeled peptides in tumors. Given the complexity and
applied. Use of PET at earlier stages with binding to the prostate cancer multidimensionality of these imaging
in the workup is becoming prostate-specific membrane antigen data, artificial intelligence (AI)
increasingly common, as is the use (PSMA) molecule show great approaches are being applied to
of PET in early assessments of the promise diagnostically and can guide augment radiologist performance.
efficacy of cancer therapies. radiopeptide therapies. “Radiomics” is the emerging field of
Adapting treatments based on the • Visceral angiography for diagnostic linking detailed analysis of
response seen on PET-CT is also purposes has been substantially quantitative images to tumor
increasingly applied with treatment supplanted by CT and MRI methods; genetics and biology. Imaging
deintensification approaches for however, it remains important as a phenotypes can more precisely
rapid responders, and treatment tool for intravascular delivery of guide individualized tumor treatment
intensification for poor responders therapies such as chemotherapy, to yield a higher probability of
being informed by PET. coils, or radioactive microspheres. success without excessive toxicity
Neuroendocrine tumors are now • CT, ultrasonography (including with for treatment of the selected
diagnosed with gallium-68 (68Ga) ultrasonographic contrast media), neoplastic process.

Noninvasive imaging is of fundamental and increasing importance in Specific clinical questions addressed by imaging include screening
the daily management of the patient with cancer. Although physical for the presence of cancer (e.g., breast, lung, or colorectal), characterizing
examination and laboratory diagnosis remain key for planning treat- anatomic lesions as malignant or benign, and staging a neoplasm—that
ment, for solid tumor management, imaging tests represent a major is, determining the size and local extent of a primary lesion and
objective metric of disease presence or absence and activity and may determining whether it is localized or locoregionally or systemically
be used at different times during the course of the disease to monitor metastatic. Such studies are essential for determining whether the
the efficacy (or lack of efficacy) of treatment. Imaging to determine patient is a candidate for surgical resection, identifying the extent of
tumor size is an objective end point in disease management used to the field for radiation therapy, and determining whether systemic
compare different types of cancer treatment and treatment across chemotherapy is appropriate. Initial staging of tumor size and extent
institutions. The use of imaging also is increasing in the drug develop- also can provide important prognostic data. During the course of
ment process and in developing new cancer therapies. treatment, imaging is used to determine response of the cancer. Imaging
256 Part I: Science and Clinical Oncology

Table 16.1 Imaging in Cancer: Key Current Clinical Sensitivity


Uses in Cancer Management Sensitivity describes how often the imaging test would give a “positive
• Screening
result” in a patient with cancer (i.e., true-positive finding). Ideally,
• Lesion characterization: malignant or benign, size, local invasion
the test precisely detects and locates one or multiple cancers in a given
• Tumor staging: locoregional, systemic, at initial presentation or on patient. Thus
retreatment
• Size and extent of tumor: to plan radiation or other local therapy % sensitivity = 100 × (test positive disease present )
• Prognostic information
• Defining sites for biopsy and subsequent analysis by pathology Sensitivity can be calculated on a per-patient basis or a per–
• Guidance of interventional therapy malignant lesion basis. The per-patient basis most commonly is used
• Assessment of response to treatment in screening studies for early diagnosis, whereas the per-lesion basis
• Restaging tumor after treatment may be used in patients expected to have multiple sites of tumor.
• Assessment of normal organ function or status before, during, and Per-lesion detection analyses can be misleading because they can be
after treatment heavily biased by a single patient’s results if that patient has multiple
• Assessment for toxicity or complications of treatment tumor foci.
90% SENSITIVE AND SPECIFIC TEST It can sometimes be difficult to judge how “good” a test is by
reading the literature. Sensitivity is supposed to be substantially
• 50% Prevalence of cancer in the population imaged independent of study population composition, but as discussed in
• 1000 Independent scans the next section, certain imaging tests may be insensitive for some
• 50 False-positive findings in healthy patients (10% of 500)
very early-stage disease, but very, very sensitive for more advanced
• 450 True-positive findings in patients with disease (90% of 500)
• Positive predictive value (disease positive/test positive): 450/500
disease. Each imaging test has a limit of detection threshold below
(90%)
which tumors cannot be detected because they are not distinguishable
• Possible conclusion: an excellent test! from the background tissues. Thus the patient population and very
often the tumor burden and average tumor size can make a difference
90% SENSITIVE AND SPECIFIC TEST in the sensitivity of a test for detection of cancer. Virtually all non-
• 10% Prevalence of cancer in the population imaged invasive imaging tests are less sensitive for small-volume disease than
• 1000 Independent scans for large-volume disease. For example, if an imaging test is used in a
• 90 False-positive findings in healthy patients (10% of 900) patient population in which patients have advanced disease before
• 90 True-positive findings in patients with disease (10% of 900) seeking medical attention (e.g., they are symptomatic at presentation),
• Positive predictive value (disease positive/test positive): 50/100 the imaging test may have far greater sensitivity than if it were used
(50%) in patients with earlier-stage, smaller tumors. For example, positron
• Possible conclusion: a rather lousy test! emission tomography (PET) with Fluorine-18 (18F)–fluorodeoxyglucose
But these are the same test! (FDG) has been reported to be more than 90% sensitive for detecting
metastatic melanoma, but it is less than 20% sensitive in detecting
early (low tumor volume) nodal metastases of melanoma at initial
surgical resection. Mammography has higher sensitivity in women
with more radiolucent than radiodense breasts. A test with high sensitiv-
ity has a low number of false-negative results.2 The false-negative
also is often used to follow patients for recurrence or development of fraction usually is expressed as 1 – sensitivity (in this case, sensitivity
second malignancies. Imaging is being used more often as a method being rated on a 0–1 scale).3
to assist in delivery of minimally invasive therapeutic procedures to
ablate cancers, to guide radiation therapy, and to guide the dosage of Specificity
therapeutic drugs, including radiopharmaceuticals, more precisely.1 Specificity is the frequency with which a test result is negative if
Imaging often is the best means of noninvasively identifying and no disease is present, or the true-negative ratio. As a percentage,
assessing tumors. With information gleaned from imaging studies, specificity is
the prognosis can be established and treatment decisions made with
greater certainty. Before discussing the varying imaging methods 100 × (test negative disease negative)
available for patients with cancer, this chapter considers some general
principles that are applicable to all imaging tests. Specificity can be calculated on a per-patient, per-lesion, or per-
region basis. The per-patient calculations commonly are performed
Tasks for Imaging in the screening setting. They also can be done per region of the body
(e.g., Is the liver free of tumor? Are the draining lymph nodes free of
The major roles of imaging in the current and evolving practice of tumor?). It is technically difficult and sometimes impossible to know
cancer management are shown in Table 16.1. exactly how many tumor foci are present because this depends on the
reference gold standard. It is not possible to perform “whole-body”
biopsies antemortem, so some very small tumor foci may not be
GENERAL CONSIDERATIONS known to be present when disease is diagnosed. Specificity can be
Performance of Imaging Tests affected substantially if the imaging test is used in a population that
has a characteristic that can result in false-positive results for the
Noninvasive imaging is used to perform a wide variety of important imaging test. For example, inflammatory and infectious lung disease,
tasks. Although the best way to determine the medical usefulness of such as active tuberculosis (TB) or sarcoidosis, if present in a patient
a diagnostic test can be argued, a few key concepts are required to population, can result in false-positive findings on PET or computed
understand and compare diagnostic tests. These can be applied to tomography (CT) scans or other imaging methods. In this situation,
one of the most basic tasks (i.e., determining whether tumor is present) the specificity of FDG-PET, and likely of CT, for staging the
and also to the ability of imaging to predict resectability or response mediastinum for cancer would vary. Thus the specificity of PET for
to treatment. assessing mediastinal lymph nodes may be much lower in areas of the
Imaging  •  CHAPTER 16 257

world with endogenous TB than in developed areas without it. Therefore death rates. In addition, strong data also exist to support virtual
an imaging test that is very useful in one part of the world may be colonoscopy for colorectal cancer screening.
far less useful in another part of the world. A highly specific test has
a low frequency of false-positive results (i.e., a low frequency of positive Receiver Operating Characteristic Curves
test results in the patient population that does not have the disease).
The ideal imaging test has both high sensitivity and high specificity, Cancer imaging tests are interpreted by imaging specialists, often
although none of our current imaging tests have perfect sensitivity radiologists. As with all of medicine, there is considerable science
and specificity.2 involved in image interpretation, but the human element, or “art,”
as it is referred to in some settings, also is involved. In developed
Accuracy of Imaging countries, medical specialty boards have been established to ensure
that practitioners have a basal level of training and knowledge, thereby
For detection of disease, a binary, yes-or-no answer as to whether providing some level of uniformity to image interpretations. However,
disease is present or absent is desirable. When such binary answers even with board certification and extensive training, not all imaging
can be provided, it is simple to mathematically provide an accuracy specialists interpret a given imaging study in the same manner. Thus
value for a diagnostic imaging test. Thus accuracy is although the goal of an imaging test often is a simple binary “yes,
there is tumor” or “no, there is not tumor” answer, there are varying
100 × ( TP + TN ) ( TP + FP + TN + FN ) degrees of certainty in the interpretation of an image in most instances.
Some readers read with high sensitivity, whereas others read with high
where TP = true positive, TN = true negative, FP = false positive, specificity. Unless a test is very robust, it is hard to achieve both high
and FN = false negative. sensitivity and high specificity.5
In this case, the number of patients with each finding is recorded. An example of a receiver operating characteristic (ROC) curve is
A highly accurate test is one with a low prevalence of false-positive shown in Fig. 16.1. This set of curves reflects the performance of PET
and false-negative results. imaging in detecting axillary metastases in patients with newly diagnosed
breast cancer. The axes of the curves are the true-positive fraction
Positive and Negative Predictive Values (sensitivity/100), which forms the y-axis, and the false-positive fraction
(1 – specificity/100), which forms the x-axis, on a scale of 0 to 1. A
Sensitivity and specificity define a test reasonably well, but its perfor- perfect diagnostic test would yield no false-positive or false-negative
mance in a specific patient is affected by the characteristics of the results. The greater the area under an ROC curve, the greater the
population from which the patient is drawn. A physician normally accuracy of the test.5
wants to know whether an individual patient has cancer and whether The results shown in Fig. 16.1 are from three readers who graded
the tumor is localized or metastatic (and where). The correct answer PET scans using a five-point certainty scale (i.e., not a simple yes/no,
is binary in most cases, but imaging does not always reveal the true but a continuum from definitely abnormal to definitely normal). The
status of the individual patient. Thus the statistical likelihood of the three readers had similar ROC curves, indicating that they were of
accuracy of the result might be conveyed in the clinical imaging test generally comparable accuracy. For the same test, however, two readers
report. This statistical likelihood will be related to the accuracy of the may be reading at different points on the ROC curve, meaning that
test as well as to the patient population characteristics. Therefore the one is more sensitive and one is more specific, but both are of equal
positive predictive value often is of considerable clinical relevance. accuracy.
For example, the positive predictive value of a test with 90% sensitivity An excellent reader may have a greater area under the ROC curve
and 90% specificity will vary markedly, depending on the frequency (AUC) than a less skilled reader, meaning that the more experienced
of disease in the population. With these test performance characteristics, (and hopefully more capable) reader is both more sensitive and more
the clinician could reach two different conclusions regarding the specific than a less experienced (and presumably less capable) reader.
practical value of the same imaging test (Table 16.1). However, virtually none of our imaging tests is perfect, and varying
A test that is effective in a patient population with a high prevalence
of a disease may be far less valuable in a patient population with a
lower prevalence of the same disease because there would be far too
many false-positive results. The most effective use of imaging technology
is in groups of patients in whom the imaging characteristics are expected 1.0
to be robust enough to allow for predictions in individual patients. 0.9
These challenges are particularly apparent when a test that was developed 0.8
and validated in a patient population with disease is used to evaluate
individuals with a lower prevalence of tumor (i.e., screening), or smaller 0.7
tumors. In this situation, the number of false-positive findings may 0.6
TPF

rise dramatically, sometimes nearly completely negating the value of 0.5


the test.4 0.4
Costs associated with a high false-positive rate can include excessive
0.3
biopsies, with both considerable economic and personal costs, as well
as an increased radiation dose in the population tested. Higher radiation 0.2
Az0.76 Reader 1 Az0.70 Reader 3
doses in a population may lead to a risk of an increased prevalence 0.1 Az0.75 Reader 2 Az0.95 Better test
of cancer. A clear balance must be achieved between when imaging 0.0
tests are applied, especially in patient groups with a low prevalence 0 0.2 0.4 0.6 0.8 1.0
of cancer, to minimize generating risks and maximize disease detection
FPF
with the test. Screening programs may have specific challenges discussed
later in this chapter. They may reveal disease that may be clinically Figure 16.1  •  Receiver operating characteristic (ROC) curves plotting
irrelevant, resulting in “overdiagnosis” and “overtreatment.” However, the true-positive fraction (TPF; sensitivity) versus the false-positive fraction
proving overtreatment is challenging and ultimately requires randomized (FPF; 1 − specificity) for the three independent readers of the entire analysis
trials. That said, high-level evidence exists that screening for breast data set. A hypothetical curve for the test if it had 95% accuracy also is shown.
cancer and lung cancer in appropriate populations reduces cancer Az, Estimated area under the ROC curve.
258 Part I: Science and Clinical Oncology

“cut points” between disease and normalcy often are made, affecting screening mammography programs have been shown to be capable
the overall performance of the test. In this study, the AUC of 0.7 to of saving lives in women older than 50 years. These programs also
0.76 was not viewed as sufficiently good for the task of nodal detection may save lives in women 40 to 50 years of age, but the data are less
of metastatic cancer spread to the axilla.6 Despite this, a very high compelling.9
sensitivity or a very high specificity can be achieved depending on Studies have been initiated in which CT scanning is used in an
which part of the curve one operates in. A higher, hypothetical curve, attempt to detect early lung cancer.10 Screening high-risk populations
with an AUC of 0.9, is shown for a more robust test, such as a with CT imaging was proven to reduce lung cancer–specific mortality
higher-resolution PET system devoted to imaging the axilla. in the National Lung Screening Trial (NLST). This followed results
In practice, sentinel node sampling, often guided by imaging or from the Early Lung Cancer Action Project (ELCAP), a large study
a radionuclide-sensitive probe system, is assuming a very important screening patients at increased risk of lung cancer with low-dose CT,
role in this area of tumor staging.6 Practically, if a rather insensitive which reported promising results in 1999.11 The ELCAP showed that
test has a high positive predictive value, then the test may be of value lung cancers are detected at a smaller size and that patients whose
if the result is positive, but of little value if negative. For example, a cancers are detected by screening live longer following diagnosis than
strongly positive PET scan for axillary metastases may obviate the patients whose tumors are not detected by screening. Whether this
need for a pretreatment axillary dissection in a patient with newly translates into longer-term survival for the screened population remains
diagnosed advanced breast cancer in whom neoadjuvant chemotherapy unclear. The ELCAP study further evaluated 31,567 asymptomatic
could be given. persons at risk for lung cancer with use of low-dose CT from 1993
through 2005, and from 1994 through 2005; 27,456 repeated screenings
Other Approaches to Assessing the Value were performed.12 A diagnosis of lung cancer was made in 484 par-
of Imaging ticipants based on screening. Of particular note, 412 patients (85%)
had clinical stage I lung cancer. Ten-year survival approached 90%
Although sensitivity, specificity, and accuracy commonly are used in this group.11,12 This study demonstrated that annual spiral CT
to characterize the tumor detection process, other metrics may be screening can detect lung cancer that is curable.
of greater importance. For example, some studies have focused on The results of another large randomized trial of lung cancer screen-
how often imaging substantially changes management. This kind ing, the NLST, were reported in 2011.13 In this trial, 53,454 persons
of study is of great practical interest, but the optimal methods to at high risk for lung cancer were enrolled from over 30 US sites. Study
assess such changes in treatment decisions are evolving. Ideally, participants were randomly assigned to undergo three annual screenings
one would like to show that the use of imaging, especially a new with either low-dose CT (26,722 participants) or single-view postero-
imaging technology, when applied randomly to half of the study anterior chest radiography (26,732); 24.2% and 6.9%, respectively,
population provided a reduction in the number of adverse events of the CT-screened and chest radiography–screened groups had positive
in the imaged population, improved survival, or had comparable screening studies at some point. There was a high false-positive screening
outcomes at lower costs than standard treatments. As an example, a rate: 96.4% of the positive screening results in the low-dose CT group
reduction in the number of “futile thoracotomies” has been used as and 94.5% in the radiography group. The incidence of lung cancer
a metric of success for PET versus CT in planning the treatment of was significantly higher (1060 versus 941 cancers) in the low-dose
newly diagnosed lung cancer.7 Ideally, randomization of patients to CT group, as compared with the chest radiography group, There were
imaged versus not imaged groups can be shown to improve survival. 247 deaths from lung cancer per 100,000 person-years in the low-dose
Performance of randomized trials in which a portion of the patients CT group and 309 deaths per 100,000 person-years in the radiography
undergo imaging and the other patients do not (or they obtain differ- group, a relative reduction in mortality from lung cancer with low-dose
ent kinds of imaging), with an end point of survival, will be of great CT screening of 20.0% (95% CI, 6.8–26.7; P = .004). The rate of
interest. Unfortunately, such studies are complex or impossible, because death from any cause was reduced in the low-dose CT group as
management of patients after imaging may be altered markedly based compared with the radiography group by 6.7% (95% CI, 1.2–13.6;
on the imaging results. Therefore it can be difficult to separate the P = .02).
imaging study effect from the treatment effect. Ultimately, however, This exciting trial has raised some controversies: the vast majority
for some imaging studies to be adopted, such evaluations of survival of small pulmonary nodules identified are not malignant, the costs
will be needed. This point is particularly relevant to screening, as of the screening and of the medical care for incidentally detected
discussed later. lesions are substantial, and a substantial radiation burden, which in
Registry data have been applied with substantial benefit to determine principle could be carcinogenic, is delivered to patients undergoing
if planned or actual patient management is altered through the use the screening. However, there is considerable hope that screening
of imaging tests. The National Oncologic PET Registry (NOPR) has programs can achieve a reduction in lung cancer mortality as the
provided a great deal of information on the use of FDG-PET imaging radiation is given to patients later in their lives, when risks of radiation-
in the management of patients with a variety of cancers. The NOPR induced cancers are reduced. Indeed, the evidence supporting lung
collected questionnaire data from referring physicians on intended cancer screening with CT was of sufficiently high quality to be included
patient management before and after PET. After 1 year, the cohort as a covered benefit in the United States under insurance policies
included data from 22,975 studies (83.7% PET-CT) from 1178 centers. consistent with the Affordable Care Act (ACA).
Overall, physicians changed their intended management in 36.5% Based on the findings of the NLST, and the approval by US Medicare
(95% confidence interval [CI], 35.9–37.2) of cases after PET, supporting of screening, many centers have offered lung cancer screening clinics
the usefulness of PET for cancer imaging in registry cases, which are for high-risk patients. A risk, however, for lower-risk patients is that
from a wide range of sources.8 screening may have an unacceptably high false-positive rate. It is
important to note that the promising results of NLST are for a higher-
Screening Concepts and Challenges risk population, and extrapolations of benefit to lower-risk populations
Screening programs for cancer often have taken the form of laboratory may be highly problematic.
tests such as the Papanicolaou (Pap) smear, or, more recently, blood As discussed later in this chapter, CT colonography holds excellent
tests for tumor markers. The success of the Pap smear in reducing promise as a screening method for colon cancer, as an alternative to
mortality rates from cervical cancer is incontrovertible. The use of optical colonoscopy, and is increasingly being paid for by health insurers
imaging in screening for cancer is an example of success and is of based on its accuracy.
considerable interest, but also is a source of considerable controversy. Other areas in which screening by noninvasive imaging has been
As discussed in detail in the chapters on breast cancer and lung cancer, performed include colorectal cancer, where virtual colonoscopy can
Imaging  •  CHAPTER 16 259

be used to look for early colon cancers, and in the pelvis in women biologically relevant at all. If so, the patients with cancers identified
who are at risk for ovarian cancer. More recently, screening CT centers in the screened population could appear to have a better survival than
offering a virtual evaluation of the entire body have become available, the patients with cancers identified in the unscreened population.
and even more recently, magnetic resonance imaging (MRI) and PET This concern exists for prostate cancer screening by prostate-specific
screening have been offered in some locales. The growth of these antigen (PSA), for example, wherein there are major concerns regarding
centers has been driven emotionally and economically; these tests overdiagnosis of slow-growing, and presumed indolent, cancers that
are not yet well founded scientifically in the screening setting. In might not need surgery.14 This concern also exists for breast cancer,
some cultures, screening imaging is done more commonly than in wherein some argue that a small to a more substantial portion of
others. The risk of false positives in patients at low cancer risk is the cancers diagnosed would not have proven harmful to patients
substantial. had they been left undiagnosed and untreated. That said, we do not
Screening carries challenges, risks, and costs that are beyond the fully know which cancers need no treatment, and an issue exist-
scope of this overview chapter. However, several key points apply to ing with “overdiagnosis” is that it really is “overtreatment.” Better
all screening approaches, including those using noninvasive imaging. screening tests that identify biologically relevant cancers would
These points include (1) whether a screening program is reasonable be ideal.
to consider; (2) lead-time bias; (3) length bias; (4) the overall economic A third factor is the selection bias that is difficult to control for
cost implications of screening, especially the costs of investigating in retrospective observational studies: how the patients and their
false-positive results, and (5) the risk that radiation used in screening referring physicians determined to have a scan performed. Selection
may include subsequent cancers. bias occurs when there are unintended differences between the groups
The requirements that a screening program must meet to be observed that, although they are associated with the variable used to
considered “reasonable” may differ substantially based on the specific sort the groups—for example, exposure in case-control studies and
society’s values and a specific individual’s perception of risk. However, in outcome in cohort studies—affect measurement of the study variable.15
general, the following characteristics are important for cancer screening: For instance, in a case-control study of the effects on disease-specific
mortality of a particular screening program, investigators would examine
• The cancer must have a considerable public health effect.
records from patients who have died from the disease in question
• The disease must have an asymptomatic period in which detection
versus those who have not, then determine the rates of the screening
by imaging is possible.
intervention in these two populations.
• A therapeutic intervention that should lead to better survival or
It is possible that those likeliest to have sought screening were the
quality of life must be available.
ones at highest risk for the disease. Results for the screening’s effect
• The prevalence of the disease must be sufficient in the population
on mortality could be underestimated in such a scenario. Selection
being screened to justify screening (especially the cost). Low preva-
bias also can work in the opposite direction, wherein the high-risk or
lence of disease lowers the positive predictive value of positive scans.
poor-prognosis individuals are less likely to seek screening. With the
• Medical, surgical, or other treatment must be available for the
relative absence of large randomized prospective trials, as is commonly
early-stage cancer identified by screening.
the case when evaluating imaging as a screening tool, one may be
• The screening test itself must not cause disease at a significant rate.
tempted to base conclusions on the findings of cohort or case-control
• There must be a high likelihood that the patients in whom early
studies. However, the most accurate conclusions would be drawn
cancer is identified by image-based screening will go on to undergo
from a prospective randomized study design.
a suitable therapeutic intervention.
Collectively, lead-time bias and length bias and, at times, selection
Furthermore, the imaging test itself must be acceptable to patients bias can make screening programs appear to improve the survival of
(in terms of level of discomfort, cost, and radiation burden), and it patients with cancer. Because of these major biases intrinsic to screening,
must be sufficiently sensitive to identify cancer often and sufficiently very large, randomized, studies are required to show that overall
specific to minimize false-positive results. Finally, the costs of the cancer-specific mortality (and ideally mortality from all causes) declines
screening process, and attendant costs related to false-positive results, as a result of screening programs.
must be compatible with the society’s or the individual’s economic All-cause mortality is a critical parameter. If the treatment of a
resources, and the screening procedure must pose little or no risk to presumed tumor discovered by a screening imaging test carries with
the patient.4 it a risk of death or morbidity, the screening program could lower
Another important consideration in screening programs is lead-time cancer-specific mortality but not all-cause mortality. If the screened
bias. This concept, simply stated, indicates that if the natural history population is very young, late adverse effects of screening may be
of a disease is unchanged but the diagnosis is made earlier in the difficult to detect, such as slightly increased risks of cancer due to
course of the illness, the apparent survival will be improved. For radiation.
example, let us assume that a tumor has a 6-year natural history from The advent of imaging and other screening tools that uncover a
its beginning until the death of the patient, and that treatment is tumor long before it is symptomatic also brings up the need for
ineffective. The disease might become clinically detectable after 4 biomarker discovery to help physicians to determine whether to treat
years and lead to death in 6 years, a 2-year survival after diagnosis. what has been discovered through screening. The experience with
With screening, if the tumor is detected 3 years after the onset of PSA screening in prostate cancer serves to illustrate the point that
disease and no improvement in treatment occurs, then the survival not all cancers identified by screening eventually lead to death. This
in the screened population would appear to increase from 2 to 3 years is likely also the case for some early-stage breast cancers of low prolifera-
after diagnosis. This illusion of improved survival in the screened tive rate and potential. Discovering and validating biomarkers that
population is a considerable concern and can lead to inappropriate can help to distinguish reliably between lethal and nonlethal tumor
enthusiasm for screening programs.4 types will be of great help in reducing unnecessary treatment in patients
Another important consideration in screening is the possibility of with less active disease. Identifying and phenotyping tumors with the
length bias. This is a more complex concept, but it may be related greatest risk for progression is of great importance because it is clear
to the types of cancer that can be detected by screening programs. A that not all cancers carry the same risk of mortality if left untreated.
possibility is that very rapidly growing and presumably highly lethal
cancers are less likely to be detected by annual screening programs, Screening Costs
whereas more slowly growing cancers, which have an intrinsically The determination of whether a screening program is valuable to
better prognosis, may be detected more frequently by screening. In society is often, in part, based on its cost and benefit. The concept
fact, some of the early cancers discovered by screening may not be of quality-adjusted life-years (QALYs) often is applied. This concept
260 Part I: Science and Clinical Oncology

is defined as the economic cost to society required to result in 1 Despite the markedly superior sensitivity of histologic methods
additional year of good-quality life for a member of the society. In over noninvasive imaging, there is a major sampling error issue for
many Western countries, a figure of $50,000 has been considered a histologic sampling, and, paradoxically, imaging in some diseases
useful guide (although many affluent countries are willing to spend potentially may be more sensitive than histologic examination for
far more per QALY), with QALYs costing less than this amount cancer. This situation can arise in mammography, in which small
considered cost-effective. Such a guideline, however, does not neces- tumor foci can be seen on the mammogram but can be missed on
sarily apply when individuals make their own determinations as to cytologic sampling, possibly because of sampling error. When tumors
whether to pay for a screening test. For example, it is reasonable are imaged with noninvasive methods, the entire tumor is visual-
to expect that those with greater disposable income would be willing ized, not just a small portion. So, paradoxically, imaging, despite
to pay more per QALY than those with less disposable income. Thus limited resolution, can be more sensitive than cytology or pathol-
it can be difficult to generalize about the cost efficacy of screening ogy. However, if exhaustive and thorough sampling is performed,
procedures.16 with microscopic examination of tissue there usually is greater
Even when people choose to undergo a screening test at their own sensitivity for tumor than there is with current noninvasive imaging
expense, considerable costs can be transferred to society as a result of techniques.
the screening program. For example, if a screening test has a high rate With the advent of serum protein tumor markers, circulating tumor
of false-positive results, a substantial number of follow-up biopsies or cell assays, and DNA- and RNA-based methods, it is possible that
procedures will be performed and can cost a great deal of money. imaging will not always be a primary tool for cancer detection. These
Such costs can dramatically raise the total cost per QALY. Invasive other methods may be more sensitive than imaging, although imaging
procedures also can increase the likelihood of morbidity or death as has the distinct advantage of both detection and physical localization
a result of additional investigations. To determine the true cost per of tumor foci. In addition, noninvasive imaging can detect heterogeneity
QALY associated with screening, these additional costs and risks must in cancers that cannot be identified with circulating cells or DNA.
be considered. Thus screening remains an area of great promise, but Such information is potentially more “actionable” medically than
also of considerable controversy. simply detecting whether tumor is present or absent.
Screening beyond breast imaging must overcome major hurdles
before it is likely to be accepted. There has been considerable Stage Migration
progress for both virtual colonoscopy as an alternative to optical
colonoscopy and for CT scanning of the lungs in patients at high One of the major goals of noninvasive imaging is to stage the tumor
risk for lung cancer. In both these populations, the Centers for precisely to allow the clinician to best choose treatment and determine
Medicare and Medicaid Services (CMS) pays for screening, as do the prognosis. The evolving concepts of tumor staging are discussed
ACA-compatible private insurance policies. It is almost certainly the elsewhere in this text, but improvements in detection technology can
case, however, that in high-risk patient groups, such as those with a change the understanding of the natural history of a given stage of
family history of cancer or major carcinogen exposure with a high disease.
penetrance or early onset of disease, screening may prove of greater Patients with small or microscopic lung cancer metastases to the
value than in the general population. As an example, MRI screening mediastinum are likely to do better than patients with bulky metastases,
in patients at high risk for breast cancer is increasingly accepted as but both may have the same stage of disease. As the sensitivity for
appropriate. detecting small lesions improves, it becomes possible to identify more
patients with small primary tumors and small metastases to the lymph
Size of Detectable Lesions nodes or small systemic foci of metastatic disease.
Noninvasive imaging methods in humans cannot enable detection When primary tumors are detected at ever earlier and less advanced
and localization of a single malignant cell, although flow cytometric stages as imaging and other detection methods improve, patients are
methods are very sensitive for finding a very few cancer cells in a patient’s assigned to a higher stage than was used historically. Their inclusion
blood, and there is a great interest in assays for circulating tumor in the advanced-stage group, as opposed to the localized disease group,
cells and for tumor DNA and RNA in the blood. Imaging methods appears to improve survival and outcome. The outcome of the lower-
are improving, however, and detection of a much smaller number of staged group also may improve because a subset of patients with
cells is possible in small-animal models. It has been estimated that by now-detectable tumors has been removed from that group. The outcome
the time a tumor reaches 3 to 5 mm in diameter, which is the lower of the overall group will not have changed because the numbers of
limit in size for detection by the best current noninvasive methods patients in each group will have been altered.
in humans, the tumor has undergone more than 25 doublings and One has to be very cautious in extrapolating historical survival
contains 0.1 to 1 billion cells, depending on their size.1 In contrast, data in a given advanced cancer stage based on an insensitive
a cytologist on a very good day or with use of flow cytometric staging method to that observed with a more sensitive method,
methods may be able to identify a single cell as malignant by using a which might move some patients to a higher stage, so-called stage
microscope. migration.4
Realistically, even for histologic assessment of malignancy, typically
a group of tumor cells must be present before cancer is diagnosed. MAJOR IMAGING MODALITIES
However, light microscopy and more sensitive techniques such as
immunohistochemistry and polymerase chain reaction studies mean Broadly stated, cancer imaging can be performed with anatomic or
that pathologic techniques potentially will be more sensitive than functional (“molecular”) imaging methods.1 The traditional imaging
imaging methods. One very important proviso is that for light of the patient with cancer, and the most established methods, are
microscopy pathologic methods to be effective, actual examination based on anatomic imaging. However, interest is increasing in more
of the malignant cells is required; the sample containing the tumor functional methods in cancer imaging. Furthermore, several anatomic
must be cut appropriately and viewed under a microscope. This task imaging methods offer functional components that complement the
may be impossible, because the 8-µm sections that are used for anatomic method. Hybrid images, derived from and displaying both
pathologic examination typically are taken only from a small portion functional and anatomic data, also are becoming more widely available,
of a tumor or lymph node, whereas most of the tumor or node will often coming from the same hybrid imaging machine, such as in
be unexamined (e.g., to assess a 1-cm node by using 8-µm-thick PET-CT.17,18 Imaging data are almost exclusively digital or digitized
sections, approximately 125 slices would be required). This large and suitable for postprocessing and image exchange. The major imaging
number of sections is not typically obtained. modalities are discussed in the following section.
Imaging  •  CHAPTER 16 261

Plain-Film Radiographs Computed Tomography


The traditional radiograph remains an important part of cancer imaging. CT is now established as the dominant imaging technique for cancer
It commonly is used to detect bone tumors and can be used to detect detection and follow-up. Currently, tumor response and staging criteria
lung cancers in the thorax. The method displays mainly water, air, very commonly are based on tumor size as measured on CT. CT
fat, and calcium density and is affected by overlapping tissue in front scanners acquire images by using an x-ray source and digital detector
of or behind the lesion. Radiographs have become increasingly digitized elements. The x-ray source rotates rapidly around the patient, usually
in the last few years, with the introduction of film digitization and with a single scan level taken in 0.5 seconds or less. Faster and faster
solid-state image screen capture devices. rotation speeds of the scanners, along with multiple simultaneous
Plain radiographs offer exceptional resolution but provide relatively detectors capable of imaging multiple slice thicknesses in a rapid spiral
little image contrast if there is not much calcium present. The radiation motion, are being used.
dose from a plain film radiograph depends on which portion of the Scanners with 16 and 64 simultaneous slices are commonly in use,
body is being examined. In most centers, plain film radiographs for with some scanners with 256 or more slices now in use. Large–field-
cancer management are being used less often, with CT scanning of-view detectors may allow even more of the body to be evaluated
increasingly replacing radiographs in the abdomen and MRI in the nearly instantaneously. Current fast scanners can potentially scan the
brain and extremities.19 entire body in a fraction of a minute. Although such evaluations
provide key information about lesion size, some lesions may elude
Mammography detection unless contrast medium is given intravenously, orally, or
both. With such devices, it also is possible to capture contrast in
Mammography is a specialized form of plain radiograph. Very arteries or veins to achieve superior visualization of these structures,
high-resolution images of the breast are obtained using specialized which can then be displayed three dimensionally or in a volume-
x-ray sources and devices optimized for breast cancer detection. rendered fashion.
Digital mammography is now available and offers greater flexibility Although more slices in a CT scanner make for faster CT scans,
of image display because of the digital image format. A limitation it is not clear that having more slices always results in a superior
of the digital format, however, is the field of view of the imaging diagnostic-quality scan. More slices and faster scans do mean that
phosphor, which may be too small to fully include some breast tissue. whole-body scans in a single breath hold can be obtained, which is
In the last several years, there has been a major shift toward digital advantageous because it can reduce the frequency of breathing
mammography. artifacts.
The Digital Mammographic Imaging Screening Trial (DMIST), A clear disadvantage of CT is its cost, both technically and in
which included nearly 50,000 women, showed comparable overall terms of the radiation dose. In the past several years, increased concern
accuracy between film-screen and digital mammography in the overall has developed regarding the total radiation dose being delivered by
study. A higher accuracy for cancer detection in women younger than CT, particularly in children, because of the potential risks of carci-
50, women with radiodense breasts, and premenopausal and peri- nogenesis. Great efforts have been undertaken to reduce the dose from
menopausal women was seen with digital mammography as compared CT, especially with use of more advanced software methods for
with film-screen mammography.20 Despite this higher accuracy, the reconstruction.
reported sensitivity for both techniques for cancers was only 41%, Although CT is an exceptional technique, it remains a predominantly
with a positive predictive value of 12%, albeit with 98% specificity. anatomic imaging method. Information can be derived based on the
This is far from ideal performance for a screening test. The move to timing of intravenous CT contrast enhancement, such as the ability
digital mammography has occurred in part to increase diagnostic to estimate tumor blood flow, but it is not easily extracted without a
accuracy but also to allow digital images to be viewed on a picture substantial radiation dose from repeated images. Thus only a limited
archiving and communication system (PACS), as is the case with number of post-intravenous contrast images are obtained with CT,
virtually all other diagnostic imaging methods in more and more to limit radiation dose. All CT images are digital. Another major
imaging centers.20,21 challenge with CT is the large amount of image data generated for
A newer technique, tomosynthesis, is under evaluation; in this analysis, data that can take the radiologist a long time to interpret
technique, a number of “slices” of the breast are generated during a fully. Newer dual-energy or multispectral CT scanners generate
mammographic image acquisition that involves a moving x-ray source. even more data and can provide virtual contrast-enhanced and
This approach offers considerable promise going forward, but is early non–contrast-enhanced images from a post–contrast administration
in its evolution technically. It has received FDA approval and will acquisition. The large amount of data in a series of CT scans is of
likely improve on the performance of mammography.22 Data suggest interest for purposes of further characterization beyond a visual one.
that tomosynthesis may help identify the 15% to 30% of breast cancers Radiomic features can be extracted and analyzed, which may add
not identified by full-field digital mammography (FFDM). The information beyond tumor size and location. In addition, artificial
specificity of tomosynthesis may be better than that of FFDM, as intelligence approaches may be of growing importance to characterize
well. However, there are challenges with the large volumes of data large data sets.24
and the potential failure to detect micrometastatic disease. The cost
efficacy of such an approach remains in evolution, and larger multicenter Angiography
studies will inform medical decisions regarding the precise role of
digital tomosynthesis—for example, whether it can totally replace Historically, angiography has been performed after intravascular inser-
mammography.23 tion of catheters into arteries, followed by rapid injection of iodinated
Two emerging technologies in breast imaging—contrast-enhanced contrast media, along with rapid-sequence filming of the images. The
breast imaging and molecular breast imaging (MBI)—may ultimately improving ability of rapid-sequence CT scanning to show the vascular
play an important role. The former involves intravenous administra- anatomy (CT angiography) has caused it to rapidly replace angiography
tion of contrast agent to identify areas of altered blood vessel density for diagnostic purposes. Angiography still can be used to produce the
and permeability (often seen in cancer). The latter uses a radioactive most precise maps of vascular anatomy before organ transplantation
tracer to identify areas rich in mitochondrial density and flow, such or radical cancer surgery. Most angiography now is performed with
as cancer. Both are showing better performance than mammog- digital image-capture devices, known as digital angiography.
raphy alone in some settings, with performance approaching that Angiographic delivery of therapy is, however, an important area.
of MRI. This form of imaging can allow for therapeutic delivery of embolization
262 Part I: Science and Clinical Oncology

materials such as coils, delivery of chemotherapeutic agents regionally, appear as areas of contrast enhancement. Such studies have shown the
or delivery of radioactive microspheres, for example. higher sensitivity of MRI versus mammography. However, there remain
Angiography has high resolution but typically delivers a high concerns that MRI may achieve this sensitivity with an accompanying
dose of radiation energy to the patient. The use of angiography relatively high false-positive rate, depending on the methods used for
remains essential for studies to evaluate gastrointestinal (GI) bleed- interpretation. Still, MRI probably is our most accurate technique for
ing, but with CT angiography continuing to improve in quality, the detecting and characterizing breast masses.
use of diagnostic angiography has become less frequent in routine MRI also can provide valuable information about tumors in close
clinical practice while cancer therapies by catheter have grown in proximity to vascular structures as well as in the liver and upper
application.25 abdomen. Tumors in the upper abdomen and liver can be degraded
in their appearance on MRI because of respiratory motion artifacts.
Ultrasonography Therefore this approach is applied less commonly than in other situ-
ations, such as in the lower pelvis, where there often is less respiratory
Ultrasonography uses reflected beams of high-frequency sound rather motion artifact. Research currently is working on developing agents
than ionizing radiation to generate images. Ultrasonography provides that can facilitate specific MRI contrast enhancement. More rapid
high resolution and some functional information, specifically about whole-body MRI acquisition methods also are under evaluation, which
the presence and direction of blood flow in tissues. It also provides may broaden the use of MRI.
some information regarding tissue characterization properties and is In magnetic resonance spectroscopy (MRS), tissue characterization
very effective in determining whether a tissue is cystic or solid. The can be achieved by sampling its magnetic spectrum at 1.5 T or higher.
method also is excellent for detecting vascular structures and determin- More and more scanners now provide a 3-T or higher field strength
ing extent of flow. signal for evaluation, offering the possibility of more refined tissue
Ultrasonography provides real-time imaging capability to guide characterization. Opportunities to detect increased content of choline
biopsies and procedures effectively. It is less effective in the evaluation (often increased in tumor foci) versus other substituents can be helpful
of deeper structures and requires access to a sonographic window. in separating tumor from nonmalignant tissues in the brain and
Therefore it has only a modest role in evaluating deep abdominal elsewhere. Spectroscopy also can provide information on lactate
structures. Ultrasonography is used commonly in evaluations of the concentration and pH, among other parameters. A limitation of
pelvis, neck (including the thyroid), and gallbladder and liver areas. spectroscopy is resolution, which typically is not nearly as fine as that
Agents that can enhance the visualization of vessels or can be specifically of anatomic MRI itself. Thus spectroscopy has only limited application
retained in clots are under evaluation, suggesting that ultrasound in most oncologic practices.29
examination can offer some functional information beyond the purely Diffusion MRI has shown promise in tumor response assessment.
anatomic.26 This depends on the freer movement of nuclei in areas of necrosis as
Ultrasound contrast agents are being applied to a limited extent. compared with the motion of nuclei in fully viable tumors. This
Currently these agents are mainly for intravascular use. An ultrasound technique is demonstrating considerable potential for response assess-
contrast agent has been approved for use in assessing the liver to help ment in brain tumors and recently has shown promise in tumors in
determine if liver lesions are malignant or benign. High-energy focused the bones, breast, prostate, and other tissues.30
ultrasound remains under development as a tool to ablate, or at least Nephrogenic systemic fibrosis (NSF) has been linked to gadolinium-
deliver thermal energy to, tumors with therapeutic intent. Ultraso- containing MRI contrast agents.31 This condition has been described
nography coupled with needle aspiration biopsy has been used in in patients with substantially impaired renal function who receive
some settings as a minimally invasive procedure to characterize nodal MRI contrast material intravenously. Black box warning labels now
metastases and primary lesions. Ultrasonography combined with appear on the product inserts for MRI, and we are seeing increased
mammography also is being evaluated as a screening method for breast use of alternative imaging methods to MRI in patients with low
cancers, but results in high-risk patients have shown disappointingly creatinine clearance rates followed by sequential imaging studies.
low detection rates relative to MRI and high false-positive rates for Gadolinium may be responsible in part for this process as it has been
ultrasonography.27 found in the skin of some of such affected patients.32,33 In 2007, black
box warning labels were added to the package inserts related to
Magnetic Resonance Imaging and Magnetic gadolinium-containing MRI contrast (Box 16.1). Gadolinium deposits
Resonance Spectroscopy in the brain have been identified on MRI scans, with the amount of
gadolinium deposited in rough proportion to the number of gadolinium
In many clinical settings, MRI, as applied in imaging for most cancers, doses the patient has received. Although this is an area of active
is used predominantly as an anatomic imaging method that does not controversy, no adverse effects have been identified with the gadolinium
use ionizing radiation. MRI offers superb contrast resolution between deposition. However, the European Medicines Agency (EMA) has
tissues and excellent spatial resolution. MRI also offers a variety of recommended that gadolinium-based contrast agents (GBCAs) be
forms of functional information. However, it does not offer the level used with caution and that linear GBCAs not be used, favoring
of temporal resolution, in general, seen with ultrasound examination, macrocyclic chelators for gadolinium. A list of approved GBCAs is
fluoroscopy, or recent-generation, multislice CT scanners. However, shown at the end of this chapter. For practical purposes, it is probably
MRI technology has moved forward inexorably, and rapid-pulse wisest to use gadolinium contrast only when necessary and in most
sequences allowing gating of images now are available for several types cases to use macrocyclic contrast agents, given the EMA’s recom-
of units. Magnetic resonance images can be of a variety of pulse mendation. In May 2018, the FDA approved new medication guides
sequences, allowing visualization of several parameters. However, for patients receiving gadolinium contrast.
visualization of hydrogen nuclei is the major approach with conventional An exciting area of application of MRI technology is in the field
1.5-T and 3-T (tesla) machines. Visualization of blood, especially of whole-body MRI. Such a test may begin to offer whole-body
with contrast materials such as gadolinium chelates, and of altered cancer surveys with no ionizing radiation delivered to the patient.
vascular permeability is routinely applied.28 Whole-body diffusion imaging methods are quite informative
MRI is the preferred procedure in evaluating neoplasms of the brain because they can detect a variety of tumors, including in bone. Such
and spinal cord regions as well as musculoskeletal tumors. MRI also is applications to date have been less sensitive than whole-body PET
being used with increasing frequency in the evaluation of tumors of imaging, but this area is emerging very rapidly34 (Figs. 16.2 and 16.3).
the extremities, such as sarcomas. With gadolinium contrast enhance- Recently, hyperpolarized C-13 imaging has been investigated. This
ment, MRI also can be useful in locating tumors of the breast, which field is in its infancy but allows some level of magnetic resonance
Imaging  •  CHAPTER 16 263

A B C

R L L R

Anterior Posterior

Figure 16.2  •  Representative 64-year-old patient with metastatic disease. (A) Technetium-99m–methylene diphosphonate (MDP) bone scan obtained
after injection of 21 mCi of the radiopharmaceutical. The bone scan demonstrates multiple areas of increased uptake in the calvaria, sternum, ribs, vertebra,
pelvis, and femur. Degenerative and inflammatory changes are noted in the left knee and left ankle (after fracture due to fall). (B) Axial diffusion-weighted
MRI images (b = 300–600 s/mm2) of corresponding areas of metastatic sites. The sternum metastases are clearly seen, as are those in the rib and femur
(arrows). (C) Sagittal image showing whole-body coverage. (Courtesy Dr. Martin Pomper, Johns Hopkins.)

single-photon and positron-emitting isotopes. Single-photon emitters


Box 16.1.  US FOOD AND DRUG typically have longer half-lives than positron emitters, and decay in
ADMINISTRATION (FDA) BLACK BOX a different fashion, emitting gamma rays. Depending on the ligand
WARNING REGARDING GADOLINIUM attached to the radioactive isotope, a wide variety of processes can be
MAGNETIC RESONANCE IMAGING imaged. For single-photon imaging, the most common isotope is
(MRI) CONTRAST MEDIUM technetium-99m (99mTc), which can be used to image bone (bone
scan with 99mTc–methylene diphosphonate) or thyroid (technetium
Warning: Nephrogenic Systemic Fibrosis (NSF) pertechnetate), for example.
Gadolinium-based contrast agents increase the risk for nephrogenic With positron emitters, the most commonly used tracer is 18F,
systemic fibrosis (NSF) in patients with which is used as the radiolabel for FDG, an agent that images glycolysis
• Acute or chronic severe renal insufficiency (glomerular filtration rate in vivo. Because tumors have increased glycolytic metabolism in general,
<30 mL/min/1.73 m2) use of this agent in tumor imaging is increasing very rapidly, especially
OR in lung and colorectal tumors and lymphomas.36
• Acute renal insufficiency of any severity due to the hepatorenal Both PET and single-photon emission computed tomography
syndrome or in the perioperative liver transplantation period (SPECT) have very high sensitivity for a small number of radioactive
With these patients, avoid the use of gadolinium-based contrast molecules in vivo as well as the ability to quantitate the radioactivity
agents unless the diagnostic information is essential and not available concentration precisely. Thus these methods are important as both
with non–contrast-enhanced MRI. NSF may result in fatal or debilitating clinical and research tools. The intrinsic lack of anatomic resolution
systemic fibrosis affecting the skin, muscle, and internal organs. Screen for PET and SPECT can be partly addressed by fusing the PET or
all patients for renal dysfunction by obtaining a history and/or SPECT images to CT by using computer software. More recently,
performing laboratory tests. When administering a gadolinium-based dedicated hybrid imaging devices, including both PET and CT or
contrast agent, do not exceed the recommended dose, and allow a SPECT and CT in a single device, have been applied to the imaging
sufficient period of time for elimination of the agent from the body practice of cancer.
before any readministration. In the last several years, combined PET-MRI devices have also
been constructed. These can include a PET scanner housed within
an MRI unit performing both PET and MRI studies at the same
time or adjacent PET and MRI scanners in which the patient table is
metabolic imaging in vivo without the need for radioactive tracer precisely aligned to allow for sequential imaging using PET and then
administration.35 MRI imaging with preservation of patient alignment. PET-MRI is
in evolution, and two current limitations are the cost of the systems
Nuclear Medicine and Positron and challenges associated with precise quantitation of the PET
Emission Tomography images for radioactivity concentrations in vivo due to the limitations
intrinsic to MRI-based attenuation correction algorithms.37 In clinical
Radionuclide methods can provide a great deal of functional informa- practice, PET-MRI and PET-CT have shown relatively comparable
tion, but the anatomic resolution often is limited. Broadly, there are outcomes.38
264 Part I: Science and Clinical Oncology

A B
Whole Body–DWI ADC map
b=50 b=300 b=600

B
PET CT PET/CT

Figure 16.3  •  This 72-year-old man had colorectal cancer and metastatic disease in the liver. (A) Diffusion-weighted images of different b-values in the
liver. (B) Corresponding positron emission tomography (PET), computed tomography (CT), and PET-CT images.

Table 16.2  Imaging Methods


Modality Resolution Sensitivity Specificity Functional Imaging Ability
Magnetic resonance imaging 1–2 mm Moderate Moderate Moderate with spectroscopy
Computed tomography 1–2 mm Moderate Moderate Low, except angiography
Radiographs 1–2 mm Low Moderate Very little
Single-photon emission computed tomography 1 cm High Moderate Excellent
Positron emission tomography 5 mm High Relatively high Excellent
Ultrasonography 2 mm Low Low Some, especially with contrast agent
Mammography 1–2 mm Moderate Relatively low None
Angiography 1–2 mm Moderate Moderate Low

Adapted from Bragg DG, Rubin P, Hricak H. Imaging strategies for oncologic diagnosis and multidisciplinary treatment. In Bragg D, Rubin P, Hricak H, eds. Oncologic Imaging.
2nd ed. Philadelphia: WB Saunders; 2002:2–20.

Higher doses of radioactive isotopes also can be therapeutic. For of experimental studies, or for superficial organs such as the breast.
example, iodine-131 (131I) is used for the treatment of thyroid cancer, Similarly, the technique may have greater usefulness in evaluation of
as sodium iodide. The same isotope, conjugated to an anti-CD20 intraoperative procedures.
monoclonal antibody, was approved by the FDA for the treatment The possibility of constructing light-emitting contrast media is a
of low-grade and transformed B-cell non-Hodgkin lymphoma that real one, and optical imaging has the potential to provide remarkable
is considered refractory to standard treatments. The tracer doses are sensitivity and resolution in superficial structures. However, it is not
used to guide the treatment doses in such instances, or at least to routinely applied in cancer imaging, with the exception of visualization
determine if the radioantibody has any unexpected targeting behavior.39 of the interior of the eye, visualization of the cervix, and endoscopy,
bronchoscopy from above and below.40 The combination of light-
Optical Imaging Methods generating acoustic signals, photoacoustic imaging, also has considerable
promise and potential for areas of the body in which light can be
Optical imaging methods are applied in a variety of ways. The external delivered at sufficient intensity. The strengths and weaknesses of the
physical examination of a patient involves visual interrogation of major imaging methods are contrasted in Table 16.2.
reflected light. Infrared and transmitted light also are used to a limited
extent in evaluating small parts of the body. Optical imaging has been Radiation Dose and Imaging
limited in clinical deployment by the limited penetration depth of a
variety of forms of light in the body. Therefore this type of approach Imaging methods that use ionizing radiation are a major source of
may prove of greatest usefulness in evaluation of small animals as part population radiation exposure. CT and nuclear medicine procedures
Imaging  •  CHAPTER 16 265

(notably, cardiac nuclear medicine procedures) are the major sources of in the clinical practice of PET is the increased glucose metabolism
the ionizing radiation.41 The most vulnerable population with regard to present in most cancers. Other PET tracers, such as those targeting
radiation exposure is children, in whom the risk of radiation-induced hypoxia, proliferation, amino acid transport, blood-brain barrier
cancers is higher, with a longer time for manifestation, than seen in permeability, and protein synthesis, are discussed in the following
cancers in older individuals.42 Clearly, caution must be applied when sections. The challenge with all imaging modalities is how best to
choosing an imaging study using ionizing radiation, because it is quite integrate them into clinical practice. The next section addresses
likely that there are some increased risks of cancer associated with a low these issues.
radiation dose distributed over a very large population of individuals.43
It should be noted that many cancer patients have relatively short DISEASE-SPECIFIC IMAGING
life spans because of their underlying disease, and the risks of the RECOMMENDATIONS
known cancer greatly exceed the potential risk of cancer from imaging.
It is also important to realize that lifetime risks of cancer may carry Brief discussions of the role and possible role of imaging in managing
varying significance—for example, a hypothetically “radiation-induced” several common cancers follow. There is great variation in the imaging
cancer developing 40 years after cure of testicular cancer has a less workup of specific types of tumors, depending on the type of therapy
immediate health care impact than a cancer developing within a few planned.
years of radiation.44 Certainly, concerns about radiation are appropriate, In general, the more aggressive and radical the planned treatment,
and every effort should be made to use imaging methods that include the more critical the need is for accurate determination of the location
ionizing radiation only when appropriate, and then at the lowest dose of all foci of tumor through imaging. Similarly, the intensity and
possible consistent with adequate image quality. frequency of follow-up imaging examinations must be guided by the
potential importance of the information gained. For example, if no
effective salvage therapy is available, intensive surveillance for recurrent
ANATOMIC VERSUS FUNCTIONAL IMAGING tumor makes little sense, except to provide reassurance to patients
Limitations of Anatomic Imaging of Cancer when test results are negative. This may seem self-evident, but it is
surprising how practice patterns vary.
Anatomic imaging has been the fundamental approach to cancer The National Comprehensive Cancer Network continues to update
imaging for more than 100 years. Anatomic methods are quite robust, recommendations on imaging approaches in specific cancers. In
as is supported by their daily use in managing individual patients addition, the American College of Radiology and Society of Nuclear
with cancer. Anatomic imaging normally enables detection of a Medicine have provided “appropriateness guidelines” for a variety of
phenotypic alteration that is sometimes, but not invariably, associated cancer therapies. These guidelines inherently lag behind developments
with cancer—a mass. However, with anatomic imaging, we often do in technology but are based on careful analyses of the literature and
not know whether masses are the result of malignant or benign etiolo- expert opinion and thus are worth consulting as they continue to be
gies, as in solitary pulmonary nodules or borderline-size lymph nodes. updated. The CMS in the United States has provided guidance allowing
Similarly, small cancers are undetectable with traditional anatomic PET with FDG to be used in the diagnosis and subsequent management
methods, because they have not yet formed a mass. of nearly all cancers based in substantial measure on the results of the
After surgery, it is even more difficult to assess for the presence of NOPR trial.
recurrent tumor with anatomic methods. Posttreatment scans are
complicated by the need for comparisons with normal anatomy to Lung Cancer
detect altered morphologic findings as a result of cancer. Anatomic
methods do not predict cancer response to treatment and do not The initial diagnosis of lung cancer often is based on incidental detection
quickly document tumors that are responding to therapy.45 Despite of an abnormality on an imaging study, although more advanced lung
these challenges, anatomic images remain routine in cancer manage- cancer can cause hemoptysis, cough, weight loss, hoarseness, infection,
ment. PET, a functional imaging method, helps to address many of or shortness of breath. There has been a great deal of interest in testing
the limitations of anatomic imaging, and when combined with anatomic screening programs for lung cancer in high-risk groups such as heavy
images in fusion images, is emerging as a particularly valuable tool, smokers. Both chest x-ray screening and CT screening have been
providing both anatomic precision and functional information in a evaluated.
single image set.45 Chest x-ray screening has not been proven to be effective at reducing
mortality from lung cancer. CT-based screening programs are of greater
Molecular and Functional Alterations in Cancer interest, and it is clear that these programs can detect lung cancers at
an earlier stage, when they are smaller than lung cancers diagnosed
The molecular bases of neoplasia are increasingly becoming well defined. by other methods, and appear to prolong survival. Patients with such
Mutations in genomic DNA precede the development of overt cancers tend to live longer than patients with cancers diagnosed at a
neoplasia.46 With sufficient alterations in genotype, phenotypic changes more advanced stage. However, detecting additional cancers is
occur. These genotypic and phenotypic changes in cancer antedate dependent on detection of small lung nodules and many small lung
the development of a discrete mass lesion and represent potential nodules, and most of these do not have a malignant etiology. The
targets for innovative imaging agents. The concepts of altered “genome,” costs, both economic and in terms of risks from invasive diagnostic
“proteome,” and “methylome” resulting in alterations in metabolism, procedures, are considerable. In addition, concerns exist that the cancers
consistent with an altered “metabolome,” are increasingly recognized detected by such an approach may be the more slowly growing cancers.
as present in cancers along with a variety of typical cancer “hallmarks.” Results from the NLST support CT screening in a high-risk smoking
PET, because of its superb sensitivity to low signal levels, can detect population. A summary of the results in a format easily understood
signals from tracers, targeting such alterations that are preferentially by patients is included.47
present in cancer. The Fleischner Society has offered guidelines for dealing with
PET has led the growing field of molecular imaging to the clinic, in small, incidentally detected pulmonary nodules, which involve variable
part because of the quantitative capabilities of PET and the sensitivity imaging follow-up for lesions greater than 4 mm in size. Growing
of electronic collimation, but also because of the choice of a proper lesions require additional investigation.48
radiotracer for cancer imaging. Although a wide variety of molecular, Certainly, if a solitary pulmonary nodule 8 mm or larger in diameter
proteomic, and metabolic alterations occur in cancers, and many of is identified on a chest radiograph or on screening CT, the workup
these can or ultimately may be imaged with PET, the most useful target to determine whether it represents lung cancer can take a variety of
266 Part I: Science and Clinical Oncology

forms. Comparison with old anatomic images is essential, but if none useful. PET has been reported to have sensitivity of approximately
are available, a decision must be made whether to perform additional 96% for detecting cancer in solitary pulmonary nodules (predominantly
imaging, perform a biopsy, remove the nodule, or follow the abnormal- ≥1 cm in diameter) in a retrospective meta-analysis,51 with specificity
ity. A variety of factors can be considered, including patient age, of approximately 80%. However, some histologic types are less well
smoking history, lesion size, and history of exposure to potential detected, such as those of adenocarcinoma in situ (bronchioloalveolar
infectious agents. carcinomas), which have lower FDG uptake.52,53 Nonetheless, the
The morphology of the nodule is investigated to determine whether PET scan can help to determine which patients require immediate
it has characteristics that suggest malignancy or benignity. The margin biopsy or excision of a nodule (i.e., a nodule with intense FDG uptake)
and internal density of the lesion are examined. Smooth, well-defined versus those who can be observed (some of those with low or no tracer
margins suggest a benign nodule, but these also are seen in 21% of uptake). The use of PET varies widely, but it can be a valuable tool
malignant nodules.49 A lobulated margin suggests cells of different for helping to determine which patients need invasive procedures for
lines with uneven growth, but can be seen in 25% of benign nodules. pulmonary nodules. Because false-negative findings occur, however,
A spiculated margin is highly suggestive of malignancy. Both benign patients who do not have surgery should be followed up regularly for
and malignant nodules can be homogeneous and can cavitate. A cavity up to 2 years to ensure that there has been no lesion growth. Fig.
with a wall thickness of 4 mm or less is likely benign in 95% of cases, 16.4A shows “hot” and “cold” nodules in the same patient.
a wall thickness of 5 to 15 mm is indeterminate, and a wall thickness Once lung cancer has been diagnosed, an appropriate staging workup
of more than 15 mm is likely malignant in 95% of cases.49 If the should be undertaken. The workup for non–small cell lung cancer
lesion contains fat, it is specific for a hamartoma, and 50% of ham- often is done to determine whether the patient is a candidate for
artomas have fat on CT. surgery. Because FDG-PET imaging is at least 20% more accurate
Benign calcifications are central in the lesion, diffuse and solid, than CT imaging, PET is commonly recommended as a staging
laminated, or popcornlike. Calcification is seen in 6% of lung cancers procedure for the mediastinum and for systemic evaluation for
on CT and tends to be eccentric or amorphous.49 Thus calcification metastases.54 In a prospective randomized trial, PET reduced the
alone does not indicate benignity in a lung nodule. If contrast agent number of futile thoracotomies by half, from 41% to 21%, versus
is administered and the lesion is monitored over 5 minutes, enhance- algorithms in which PET was not performed.55 This occurred, in part,
ment of less than 15 Hounsfield units (HU) suggests a benign lesion because remote foci of metastatic disease that were not identified with
and enhancement of greater than 20 HU suggests a malignant lesion, standard staging methods were identified with PET.
with reported sensitivity of as high as 98%, specificity of 73%, and Consensus is developing on how PET should be used in staging
accuracy of 85%.50 However, these CT criteria rely on very small non–small cell lung cancer, but many centers routinely perform PET-CT
changes in CT attenuation levels. These subtle changes may be insuf- before surgery, with the incremental benefit being most apparent (in
ficient to allow for reliable stratification of nodules as malignant or terms of detecting additional remote disease from the primary lesion)
benign, because timing of the CT bolus also is an important consid- in patients with larger primary lung cancers. It usually is considered
eration. The growth rate of the lesion also can be evaluated, but this prudent to perform a biopsy on FDG-avid tissues to prove that they
is difficult for subcentimeter lesions. Computer programs for nodule represent cancer. Many would argue that mediastinoscopy is no longer
detection are being developed that also provide lesion volume, and essential for patients who have negative mediastinal PET and CT
this may prove useful in follow-up. scans, although in some patients, cancer in nodes is detected only
However, for a significant number of patients, the risk of cancer surgically. An example of a positive PET scan with ipsilateral and
remains intermediate. For such patients, FDG-PET imaging may be contralateral mediastinal tumor involvement is shown in Fig. 16.4B.

A B
Figure 16.4  •  (A) Images obtained with positron emission tomography (PET) and computed tomography (CT). The upper left image is CT, the upper
right image is PET, and the lower left image is a fusion of PET and CT data. The CT scan shows a smaller right apical nodule and a large left upper lung
nodule. PET shows increased uptake of fluorine-18 fluorodeoxyglucose (18F-FDG) in the left apical nodule, consistent with cancer, whereas only minimal
uptake is seen in the right apical lesion, consistent with benign disease or a very low grade malignancy. Fused PET and CT images confirm the location of
increased apical FDG uptake, as in the left lung nodule. (B) Whole-body images from PET scans of the patient shown in (A). The images include coronal,
sagittal, transverse, and “projection” whole-body views, from left to right. They show cancer in the left upper lung, mediastinal involvement, and a right
paratracheal tumor focus. Normal uptake is seen in the heart, brain, and excretory system (bladder).
Imaging  •  CHAPTER 16 267

The role of PET-CT in lung cancer is to provide a thorough whole-body and diagnostic-quality CT used together provide sufficient diagnostic
screening assessment. information that no additional studies are required.
For mediastinal nodes, a short-axis diameter of greater than 1 cm For small cell lung cancer, it must be determined whether the
is considered abnormal on CT. Larger nodes can be reactive, and disease is extensive or localized before the form of therapy can be
nodes smaller than 1 cm can contain tumor, leading to sensitivity of chosen. CT is essential for the chest and upper abdomen. MRI of
40% to 67% and specificity of 79% to 86% for metastatic disease.56 the brain typically is performed, and a bone scan is done to search
The hybrid PET-CT technology is significantly superior to PET for bone metastases. PET scans have been shown to upstage about
alone for the staging of lung cancer.57 The best algorithm for staging 10% of patients with limited-stage small cell lung cancer to extensive
the mediastinum is evolving, but PET and PET-CT are the most disease and to find lesions not identified with CT. FDG-PET identifies
accurate noninvasive methods. Some argue that mediastinoscopy is essentially all lesions detectable with CT.58 Thus many centers are
necessary in each case, however, because PET may produce false- using FDG-PET more routinely for this type of staging, and the CMS
negative results in patients with a low tumor burden, although the has approved FDG-PET for staging of small cell lung cancer.
negative predictive value of a negative PET scan and a negative PET also is useful in the evaluation of mesotheliomas, although
CT scan is approximately 95%. Others, however, would argue that the data are evolving, and CT is the established method for this type
patients with a low tumor burden, below the level of detectability of evaluation. For follow-up of patients with lung cancer, the guidelines
with PET and CT, may be suitable candidates for surgery without are more challenging because the available therapeutic options often
mediastinoscopy. Positive PET scans for metastases usually require tissue are more limited. PET has a growing role because it can better separate
confirmation of the most advanced site of tumor to avoid false-positive residual scarring from viable tumor than can CT.
imaging findings that indicate a tumor is not resectable. Fig. 16.5
illustrates detection of lung cancer on CT and the use of reformatted Evaluation of the Adrenal
virtual images.
MRI with contrast is recommended for imaging the brain if brain Adrenal masses occur in approximately 9% of the population. These
imaging is indicated. It usually is performed if patients have larger masses can be benign adenomas, metastatic disease from primary
primary tumors or any symptoms that suggest central nervous system tumors such as lung cancer, or primary adrenal cortical carcinoma.
involvement, although in some centers, MRI with contrast is performed CT and MRI are used to distinguish adenomas from malignant lesions
in every patient with lung cancer in whom resection for cure is planned. in the adrenals. Adrenal adenomas have low attenuation on noncontrast
Evaluation for bone metastases currently is performed by bone scan. CT as a result of elevated lipid content. If a threshold value of 0 HU
Any patient with bone pain or an elevated alkaline phosphatase level is used, sensitivity is 47% and specificity is 100% for the diagnosis
should undergo this study. In practice, where PET is available, bone of adenoma.59 If a threshold value of 10 HU is used, sensitivity is
scans are increasingly being replaced by PET scans. This area is evolving, 71% and specificity is 98% for the diagnosis of adenoma.
and the bone scan remains the routine procedure in most centers Adenomas take up contrast material, but the washout of contrast
when bone metastases are suspected, but FDG-PET can reveal lung material from an adenoma is faster than from a metastatic lesion. On
cancer metastases to bone that are not detected with CT alone. 10-minute delayed images obtained after contrast injection, a decrease
In institutions where PET is not available, bone scan, MRI of the in the density of the lesion greater than 50% is specific for an adenoma.59
brain, and CT of the chest and abdomen, to include the adrenals, If the lesion is atypical on CT, chemical shift imaging on MRI can
are recommended. The abdominal CT scan should be performed with be used to determine whether it is an adenoma. An adenoma has
contrast agent to best evaluate the liver. Fig. 16.6 shows extensive both lipid and water content, and decreases in signal are seen on
mediastinal disease and liver metastases on CT. In some centers, PET out-of-phase T1-weighted images. PET has a growing role in evaluating

A B
Figure 16.5  •  Lung cancer. (A) Axial computed tomography (CT) image (lower left) shows a mediastinal mass (arrows) narrowing the airway. Coronal
(top left) and sagittal (top right) reconstructed images show narrowing of the airway by a mass (arrows). The virtual bronchoscopy image (lower right) shows
an endoluminal view of the narrowed airway. (B) Coronal reconstruction of CT data in a different patient shows a cavitating mass in the left upper lobe.
(Courtesy Dr. Leo Lawler, The Johns Hopkins University.)
268 Part I: Science and Clinical Oncology

A B
Figure 16.6  •  Lung cancer. (A) Axial computed tomography (CT) scan of the chest with intravenous contrast agent shows a mass obstructing the right
upper lobe bronchus. (B) CT scan of the abdomen shows a metastatic hypodense lesion in the left lobe of the liver.

Although mammography is a reasonably sensitive and specific


procedure, the relatively low prevalence of cancer means that many
image-directed biopsies show no tumor; thus the results of the mam-
mogram were falsely positive. Stereotactic biopsy devices have greatly
facilitated nonsurgical breast biopsies. Although the sensitivity of
mammography is generally considered “good,” an overall sensitivity
of less than 50% was seen for x-ray mammography in DMIST. Even
though this can be improved on through the use of digital rather than
screen-film mammography, these results indicate clearly that mam-
mography is far from perfect as a screening tool.
Early efforts with tomosynthesis of the breasts, a mammo-
graphic method that provides a number of “slices” of the breast
for analysis, are promising.61 Recently, breast tomosynthesis devices
have received FDA approval, and some studies have shown superior
performance of breast tomosynthesis compared with more standard
mammography. Tomosynthesis helps remove overlying structures
from the breast image, potentially improving lesion detection. It
also is possible that use of intravenous contrast agent may enhance
mammographic results.62
Although other techniques can reveal breast cancer, only ultraso-
nography is used fairly commonly in most imaging centers to help
to separate cystic from solid lesions or to help to locate and evaluate
palpable but mammographically negative lesions. MRI with gadolinium
Figure 16.7  •  Positron emission tomography (PET) and computed contrast is used because it is a very sensitive technique and can help
tomography (CT) image panel shows a variety of PET, CT, and fused images to determine whether disease is unifocal or multicentric. An example
that reveal diffusely metastatic lung carcinoma. Notable are adrenal metastases,
best seen on the transverse images, although many metastases are visible in a
of a positive mammogram is shown in Fig. 16.8A.
variety of locations. This figure also shows the multiple types of images available The use of mammography in screening has been established as a
from a PET and CT system. technique that saves lives; however, controversies remain regarding
the method. As a tool that delivers ionizing radiation, it is not without
risks. The benefit of mammography in women younger than 50 years
of age is somewhat less clear because they typically have more radiodense
the adrenals, with accuracy of 90% and greater reported in some breasts and have a lower frequency of breast cancer than older patients.
series. High FDG uptake typically is seen in metastases to the adrenal. The optimal frequency of breast cancer screening has been subject to
Caution is in order, however, in that some adrenal adenomas can have varying recommendations as well, with some guidelines recommending
moderately high FDG uptake. Fig. 16.7 illustrates detection of adrenal screening every 2 years as opposed to annually. It has also been argued
and systemic metastases on CT and PET. that mammography may disproportionately detect slower-growing
breast cancers, which may be less medically relevant and possibly lead
Breast Cancer to “overdiagnosis” and “overtreatment.”
Another concern is that the rate of detection of advanced breast
Mammography is the major imaging tool in breast cancer, allowing cancers has not changed a great deal since the introduction of mam-
for early detection of tumors. When properly used, mammographic mography, and that some of the improvements in outcomes are due
screening programs have been shown to save lives when compared to better overall therapies of breast cancer and not solely to early
with unscreened populations, and these programs are routinely detection related to improved diagnosis.63 Although there are some
implemented in many countries.60 controversies, available data point to regular mammography in women
Imaging  •  CHAPTER 16 269

A B C
Figure 16.8  •  Breast cancer. (A) Mammogram of the right breast shows cancer. (B) Axial computed tomography (CT) image shows an irregular soft tissue
primary mass in the right breast in a different patient. (C) Axial CT image with a lung window shows a metastatic nodule in the right lung. ([A] Courtesy
Dr. Nagi Khouri, The Johns Hopkins University.)

older than 50 and younger than 75 years as a valuable tool for sub- reviewed.67 PET methods occasionally are applied in the breast, but
stantially reducing the risk of death from breast cancer. they are used for diagnosis infrequently owing to the low sensitivity
Another method of breast imaging using radionuclides is molecular of whole-body PET scanners to small tumors. PET is used more
breast imaging (MBI), in which 99mTc-MIBI is injected intravenously, commonly in disseminated disease. FDG-PET appears to be superior
and the breasts are imaged with a high-resolution dual-head or single- to CT for detecting disseminated breast cancer.68
head gamma camera, optimized for breast imaging. This approach Dedicated PET imaging devices for the breast have been increasingly
has been evaluated in a number of settings. The radiodense breast is applied for diagnosis. Such positron emission mammography (PEM)
highly problematic for cancer detection with mammography, but it devices have relatively good resolution, likely superior to that available
appears that the MBI approach is more sensitive. In a study in which with whole-body PET alone. PEM and MRI performed relatively
MBI was offered as a part of screening for radiodense breasts, of 936 comparably. PEM has been directly compared with MRI in a study
women screened, 11 cancers were identified (one with mammography of 388 women with newly diagnosed breast cancer. Additional cancers
only, seven with gamma imaging only, two with both combined, and were found in 21% of women. Thirty-four percent of the 82 cancers
one with neither). Diagnostic yield was 3.2 per 1000 (95% CI, 1.1–9.3) were identified with both PEM and MRI; 26% with MRI only; 17%
for mammography, 9.6 per 1000 (95% CI, 5.1–18.2) for gamma with PEM only; and 8.5% with mammography and ultrasonography.
imaging, and 10.7 per 1000 (95% CI, 5.8–19.6) for both (P = .016 Of 306 breasts without additional cancer, 279 (91.2%) were correctly
versus mammography alone).64 The challenge with this method is assessed with PEM compared with 264 (86.3%) that were correctly
radiation dose. Major efforts to reduce radiation dose are ongoing. assessed with MRI (P = .03). The positive predictive value of biopsy
With dose reduction, these approaches may find a growing role in prompted by PEM findings (47 [66%] of 71 cases) was higher than
breast cancer screening, but they must compete directly with MRI that of biopsy prompted by MRI findings (61 [53%] of 116 cases)
approaches, which do not use ionizing radiation. (P = .016).69 MRI was slightly more sensitive, but PEM more specific
MRI has been evaluated as a tool for characterizing substantially in this group. Thus there is promise for the PEM method, especially
abnormal mammograms or ultrasound scans. The prospective mul- in women who cannot undergo MRI. However, it is probable that
ticenter National Institutes of Health (NIH)–sponsored trial in which tomosynthesis or MBI may provide similar data at a lower radiation
821 patients referred for breast biopsy because of American College dose, although this area continues to evolve.
of Radiology category 4 or 5 mammographic assessment or suspicious Breast cancer often metastasizes first to locoregional lymph nodes.
clinical or ultrasound findings were studied with MRI found an AUC Current noninvasive imaging methods do not evaluate the axillary
of 0.88 in a population with 404 cancers present, with dichotomized nodes effectively. However, imaging can help to define which lymph
data demonstrating sensitivity of 88% and a specificity of 68%. The nodes should be resected for histologic sampling. Sentinel node imaging
positive predictive value was about 72%. The high sensitivity was still or detection through use of a radiation-sensitive probe system is
insufficient to obviate the need for biopsy in these patients, however.65 becoming increasingly more important in axillary assessment of patients
MRI has been found to reveal breast cancer in the contralateral with breast cancer.70 This procedure is discussed in more detail elsewhere
breast of women with newly diagnosed primary breast cancers in in the text, but imaging is used, especially in European centers, to
approximately 3% to 4% of cases.66 These additional cancers are found better localize the axillary nodes for intraoperative assessment and
at a rate of about 25% in the lesions identified in this manner (i.e., determine atypical routes of lymphatic drainage.
75% of the biopsy results are negative). In addition, MRI has been Studies have shown sentinel node sampling procedures to be at
used to screen high-risk women (e.g., those with the BRCA1 or BRCA2 least 90% sensitive—which is considered useful sensitivity—relative to
mutation) for breast cancer to some advantage. This higher sensitivity axillary dissection, and they are virtually 100% specific.71 FDG-PET is
comes at the cost of more false-positive results and challenges in biopsy, not sufficiently sensitive to detect small metastases and has an accuracy
which have limited deployment of the method. However, increased of only about 75% in axillary nodal staging.6,72 Data are emerging
uniformity of reporting and greater similarities in technique are leading that detection of bulky nodal lesions is of greatest importance for
to more use of this methodology, which has been comprehensively outcomes.
270 Part I: Science and Clinical Oncology

other methods, though there has been progress. Although transrectal


Table 16.3 US Food and Drug Administration ultrasound (TRUS) has been used to guide biopsies, up to 40% of
(FDA)–Approved Gadolinium-Based prostate cancers are isoechoic (and thus undetectable) by this method.
Contrast Agents Similarly, the positive predictive value for cancer of hypoechoic lesions
of the prostate typically is well below 50%.
Brand Name Generic Name Structure Ultrasound imaging can be used to detect abnormalities and guide
Ablavar gadofosveset trisodium Linear biopsy of the entire prostate because it does an excellent job of defining
Dotarem gadoterate meglumine Macrocyclic the overall shape, size, and location of the prostate for purposes of
systematic biopsy. A sextant approach often is used to sample the
Eovist gadoxetate disodium Linear
base, middle, and apex of the prostate bilaterally, in addition to taking
Gadavist gadobutrol Macrocyclic biopsy samples of any suspicious areas. Although this technique is
Magnevist gadopentetate Linear less than perfectly accurate for detecting prostate cancers, on ultrasound
dimeglumine examination, tumors in the peripheral zone are more readily visible
MultiHance gadobenate Linear than tumors in the inner gland. The peripheral zone is echogenic,
dimeglumine and tumors that are hypoechoic to it (approximately 60%) can be
Omniscan gadodiamide Linear detected.74 However, hypoechoic lesions also can be caused by inflam-
OptiMARK gadoversetamide Linear
matory processes, and the positive predictive value of ultrasound is
approximately 18% to 52%.74
ProHance gadoteridol Macrocyclic Contrast enhancement of the prostate on ultrasonography has
been used to some advantage to enhance detection rates and positive
yields on biopsies of the prostate.75 Similarly, on MRI, cancers that
The extent of systemic imaging required in the initial workup of a are hypointense on T2-weighted images are seen, but the findings are
patient with breast cancer depends on the size of the primary tumor not specific for tumor. Extension of cancer beyond the prostate capsule
and the status of the axillary nodes at biopsy. It can be argued that is suggested on ultrasound when the capsule margin is irregular or
the likelihood of systemic metastatic disease is higher for patients the seminal vesicles are abnormal in morphology; however, sensitivity
with positive nodes; therefore more intensive evaluation may be more of as low as 20% has been reported. The sensitivity of MRI for
appropriate. Some would suggest a baseline bone scan and CT of extracapsular invasion is approximately 50%, and specificity is 95%.74
the chest and abdomen at this time. The frequency of follow-up is The range of reported accuracies varies widely.
debated, given the improbability of curing recurrent systemic disease, Previously, with 1.5-T magnets, intrarectal coils were sometimes
and some have advocated only limited biochemical follow-up. However, required to produce optimal images of the prostate. However, with
this area is controversial. Examples of primary and metastatic breast newer 1.5-T and 3-T systems, very good images of the prostate can
cancers imaged on CT are shown in Fig. 16.8B–C. PET is a highly be obtained, often without the need for a rectal coil This leads to
effective tool for evaluating soft tissue involvement and quickly better patient acceptance and greater dissemination of this methodology.
assessing treatment response.73 FDG-PET imaging can detect dis- The typical approach to prostate imaging with MRI has been an
seminated cancers; however, the optimal role of PET in imaging is emphasis on T2-weighted images, in which cancers often appear as
evolving. PET is now approved for monitoring treatment response of hypodense; however, a variety of other pulse sequences are now being
breast cancer and often can predict histological outcomes within 2 applied, so imaging of the prostate may include T1 imaging (for
weeks of therapy initiation. detection of neurovascular involvement), T2 imaging for intraprostate
and capsular assessment, and diffusion contrast enhancement (DCE)
Prostate Cancer (Table 16.3) and apparent diffusion coefficient (ADC) images. The DCE images
can be kinetically modeled and a variety of parameters extracted and
The role of imaging in the diagnosis and management of prostate displayed. Thus a multiparametric MRI approach to prostate cancer
cancer is rapidly evolving. The serum PSA test remains controversial imaging is increasingly being applied, and likely is leading to more
but when used in the screening setting has allowed the detection of accurate diagnoses and local staging.
smaller prostate cancers than generally are detectable clinically, along A systematic reporting system for prostate cancer, Prostate Imaging–
with opportunities for earlier interventions. In addition, mortality Reporting and Data System (PI-RADS), has been developed and
from prostate cancer has declined in the past two decades. Many continues to be updated.76 At present, although the DCE portion of
believe these two observations (more PSA testing and fewer deaths the MRI is perhaps the least valuable, it has been reported to have
from prostate cancer) are not random associations, although this is more value in the peripheral zone of the prostate and is usually included
discussed in more detail elsewhere in this book, and is a topic of in multiparametric imaging studies. A prospective multicenter MRI
controversy. Although there is some evidence that early surgical resec- study from the United Kingdom, PROMIS, compared multiparametric
tions are more closely associated with diminished mortality from MRI with grid biopsies of the prostate and TRUS postgrid biopsies
prostate cancer than active surveillance, large prospective trials compar- in 740 men, 576 of whom underwent 1.5-T multiparametric MRI
ing radiation therapy, prostatectomy, and active surveillance (with the followed by both TRUS biopsy and template prostate mapping (TPM)
possibility of intervention if there is progression of disease) have shown biopsy. On TPM biopsy, 408 (71%) of 576 men had cancer, with
similar 10-year survival outcomes. These studies were not image driven 230 (40%) of 576 patients having “clinically significant” cancers. For
at their outset (i.e., there was not extensive imaging of the prostate clinically significant cancer, multiparametric MRI was more sensitive
itself ), but they indicate an opportunity to include advanced imaging (93%; 95% CI, 88%–96%) than postgrid biopsy and TRUS biopsy
in prostate cancer treatment decision making. Imaging in prostate (48%; 95% CI, 42%–55%; P < .0001) and less specific (41%
cancer could play several roles: detecting cancer in the prostate, detecting [36%–46%] for multiparametric MRI versus 96% CI [94%–98%]
the extent of tumor, determining if the tumor is highly likely to be for TRUS biopsy; P < .0001). These authors suggest the potential for
aggressive, and determining whether the tumor has spread beyond multiparametric MRI to identify patients who do not need biopsies
the prostate locally or systemically at diagnosis or recurrence. of the prostate. Of course, some false negatives would occur, which
might be problematic if not subsequently identified at a treatable
Evaluation of the Prostate time. This study also did not optimally evaluate the TRUS biopsy,
Intraprostate carcinoma is not detected particularly well with imaging because these were performed after whole-prostate template biopsy
with ultrasound, and there are limitations to imaging with MRI and sequences. MRI-guided biopsy techniques are also now being applied
Imaging  •  CHAPTER 16 271

in research and clinical settings, for the purpose of diagnosis and also It is not clear that MRI is better than CT for nodal staging;
as a means of guiding locoregional therapies. Such approaches, when however, data on MRI using a node-specific paramagnetic contrast
applied by experts, increase the yield of biopsy results from the prostate agent have shown very high sensitivity and accuracy in the detection
compared with random biopsy approaches.77 This area continues to of nodal metastases.78 There is considerable hope for this or related
evolve, but the precise and optimal method of deploying MRI methods, methods, but MRI- based contrast agents have not yet been approved
and the precise patients who will benefit most from the method, by the FDA for this indication, and use of such agents has not yet
remain in evolution, and practice patterns vary considerably across been widely accepted.80 Given the traditionally low sensitivity of CT
centers, although expert-based guidelines have been developed. It is and MRI to detect nodal metastases, it has been recommended that
clear that biopsy remains essential for findings seen on MRI owing the serum PSA level be at least 20 ng/mL before CT is performed.
to a relatively high false-positive rate of lesion identification on MRI. If the CT findings are positive, then biopsy can be performed of the
It is also commonly the case that MRI-informed biopsies can be enlarged node or nodal sampling can be performed before radical
performed by importing MRI data into an ultrasound-guided system prostatectomy. CT also is the test of choice for visceral metastases.
to increase the yield of biopsies. These approaches can increase the Nuclear medicine methods such as C-11 choline and prostate-specific
yield of “significant” cancers, although there is not full agreement on membrane antigen (PSMA) targeting agents are showing increased
what is a significant cancer. Patients at intermediate risk for invasion, application for nodal staging.
with a PSA level of 10 to 20 ng/mL and a Gleason score of 5 to 7, A 99mTc bone scan is a reasonably sensitive technique relative to
may benefit from MRI staging of local extension.74 radiographs for bone metastases. The results of a bone scan can be
MRI methods are improving. Diffusion MRI appears particularly positive when radiographs of bone are essentially normal. The current
helpful and in some centers is preferred over DCE studies. Some recommendation is that a radionuclide bone scan be performed only
centers use spectroscopy74,78; however, this is not yet a routine part if the serum PSA level at presentation is greater than 10 ng/mL. Only
of management of prostate cancer. 3-T methods are likely superior very rarely is a bone scan positive for tumor at lower levels at the time
to 1.5-T methods for spectroscopy. Increased field strength for MRI of diagnosis. Some argue that for large primary tumors or very high
is now being more systematically assessed and appears promising as Gleason scores a bone scan still may be appropriate at staging. Bone
a tool to separate benign from malignant prostate tissue by enabling scans with 18F-sodium fluoride (18F-NaF) PET imaging or SPECT
detection of tissue with increased ratios of choline to citrate. To date, are likely more sensitive than standard bone scans. The precise role
it has been hard to prove that spectroscopy greatly improves the for NaF PET in evaluating patients with prostate cancer is uncertain,
diagnosis of prostate cancer.79 The role of MRI in prostate cancer but it is increasingly available in the United States with FDA approval
imaging continues to evolve. It is quite possible the MRI will in the of NaF for bone imaging. Typically, more lesions are seen with NaF
future be able to identify patients with small primary tumors who PET than with standard bone scan or bone SPECT imaging. For
may be managed with active surveillance. The role of MRI in follow-up example, in a study of 44 men with high-risk prostate cancer, bone
of active surveillance remains in evolution. scans (99mTc–methylene diphosphonate [MDP]), SPECT bone scans,
CT has no established role in evaluating the prostate itself or and NaF PET scans were compared. Of the 156 18F-fluoride lesions,
local invasion (25% or less sensitivity for capsular invasion), although 81 lesions (52%), including 34 metastases, were overlooked with
tumors sometimes can be seen on CT (Fig. 16.9A). CT is used to normal appearance on planar bone scan. SPECT revealed 62% of the
detect bladder and rectal invasion, adenopathy, and distant metastases, lesions overlooked with planar bone scan. 18F-fluoride PET-CT was
and MRI is used to assess local extent. Similarly, the sensitivity for more sensitive and more specific than bone scan and more specific
detecting nodal metastases has been reported to be as low as 30% than PET alone (P < .001).
with CT. Higher sensitivity can be achieved, but with lower specificity. Thus imaging is used selectively in patients with newly diagnosed
Pathologic proof is desirable before it is concluded that a patient prostate cancer. A bone scan commonly is used for recurrent prostate
has metastatic disease to the lymph nodes, and if metastatic disease cancer whenever the PSA level begins to rise.
is demonstrated, radical prostatectomy is considered inappropriate. Much monitoring of prostate cancer now involves sequential
Large nodal metastases are easily detected with CT imaging, however blood tests to measure the PSA level. When the PSA level rises
(Figs. 16.9B and 16.10). persistently, the tumor has recurred, but determining the site of

A B
Figure 16.9  •  Prostate cancer. (A) Axial computed tomography (CT) image shows an enlarged prostate with an enhancing mass and irregular margins.
(B) A more superior image shows enlarged bilateral pelvic nodes from metastatic disease.
272 Part I: Science and Clinical Oncology

A B C

D E F

G H I
Figure 16.10  •  Prostate cancer of the transition zone in a 52-year-old man with a Gleason score of 3+4 and a prostate-specific antigen level of 19 ng/
mL. Endorectal magnetic resonance imaging was performed at 1.5 T. (A) An axial T2-weighted image (6000/92) shows ill-defined homogeneous dark infiltration
of the central gland (arrows). (B) A sagittal T2-weighted image (3350/92) shows homogeneous dark tissue replacing the central gland (arrows). (C) An axial
color diffusion contrast-enhanced magnetic resonance imaging map shows a large area of high permeability (Ktrans) (red areas) in the transition zone. (D) A
permeability histogram shows a shift toward high permeability values, a finding characteristic of cancer. (E) A kinetic curve (percentage of enhancement over
time) shows a typical washout pattern in the transition zone tumor. (F) The magnetic resonance spectroscopic spectrum from the transition zone tumor shows
a high choline (Ch) peak (arrow) at 3.2 ppm that is above that of citrate (Ci) at 2.64 ppm. Cho + Cr/Ci = 1.31, where Cr = creatine; this value is typical of
prostate cancer. (G) An ex vivo T2-weighted image (4700/42) of the specimen, obtained at 9.4 T, shows highly cellular, compact dark tissue in the central
gland (arrows) surrounding the urethra (U). (H) A photograph of a whole-mount reconstructed histologic section (original magnification ×2; hematoxylin-eosin
[H&E] stain) of the midgland shows a large volume of tumor in the transition zone (outlined in green). Note the excellent correlation with the ex vivo image
in (G) and the in vivo image in (C), which show cancer of high cellular density in the transition zone. (I) A photomicrograph of a histologic section (original
magnification ×40; H&E stain) from the transition zone tumor shows loss of gland units and sheets of cancer cells with randomly scattered lumina. Note the
muscular stroma component between the tumor cells. (From Bonekamp D, Jacobs MA, El-Khouli R, Stoianovici D, Macura KJ. Advancements in MR imaging
of the prostate: from diagnosis to interventions. Radiographics. 2011;31:677–703.)

recurrence often is problematic. Bone scans and CT often are used, the PET scans. In each of the four studies, at least half the patients
with radio­antibody imaging for anti-PSMA antibodies used only who had abnormalities detected on PET scans also had recurrent
infrequently because of limited diagnostic accuracy, although some prostate cancer confirmed by tissue sampling of the abnormal areas.
promising results have been obtained by experienced groups. PET PET scan errors also were reported. Depending on the study, falsely
methods are growing rapidly in prostate cancer; however, FDG-PET positive PET scans were observed in 15% to 47% of the patients.
often is falsely negative in prostate cancer, especially for disease in the These findings underscore the need for confirmatory tissue sampling
earliest stages.62 of abnormalities detected with C-11 choline injection PET scans.
Several more accurate methods of detecting metastatic prostate Other agents are under development for prostate cancer imaging. For
cancer have been developed. C-11 choline PET was approved by the example, 18F anti-FACBC, a synthetic amino acid, shows considerable
FDA in September 2012. C-11 choline provides an important imaging promise.81 When compared with FDA-approved ProstaScint for disease
method to help detect the location of prostate cancer in patients detection in the prostate bed, anti-3-(18)F-FACBC had an accuracy
whose blood test results suggest recurrent cancer when other imaging of 83%. Indium-111 (111In)–capromab pendetide had an accuracy
findings are negative. Per the FDA labeling of the product, the safety of 67%. In the detection of extraprostatic recurrence, anti-3-(18)
and effectiveness of C-11 choline injection were verified by a systematic F-FACBC had an accuracy of 100%. 111In–capromab pendetide had
review of published study reports. an accuracy of 47%. 18F-FACBC now known as fluciclovine, has
Four independent studies examined a total of 98 patients with been FDA approved in the United States for intravenous injection.
elevated blood PSA levels but no sign of recurrent prostate cancer on It is indicated for PET imaging in men with suspected prostate
conventional imaging. After PET imaging with C-11 choline, the cancer recurrence based on elevated blood PSA levels following prior
patients underwent tissue sampling of the abnormalities detected on treatment.
Imaging  •  CHAPTER 16 273

Several radiopeptides that bind to the PSMA target in prostate Evidence supporting the accuracy of CT colonography has continued
cancer are showing great promise in human studies. Gallium-68 (68Ga) to grow. In the United States, a variety of insurance carriers pay for
and 18F peptides have been evaluated. Gallium PSMA agents have virtual colonoscopy screening for colon cancer. There is also evidence
better detection capabilities for nodal metastases than anatomic imaging that in the 65-year-old and older population, virtual colonoscopy
methods.82 In a study of 130 patients, lymph node metastases were performs as well as does optical colonoscopy. However, coverage by
found in 41 of 130 patients (31.5%). On patient-based analysis, the the CMS in the United States for screening has been slow in evolution,
sensitivity, specificity, and accuracy of 68Ga-PSMA-PET were 65.9%, and at the time of writing this chapter, virtual colonoscopy has been
98.9%, and 88.5%, and those of morphologic imaging were 43.9%, approved for diagnostic but not screening purposes by the CMS.
85.4%, and 72.3%, respectively. Direct comparisons of C-11 choline Regardless of the means of detection, the extent of preoperative
and 68Ga PSMA binding peptides show higher frequencies of lesion imaging performed before resection of primary colon cancer is variable,
detection with the PSMA binding ligand.83 In a study of 129 patients based on the institution. In general, the larger the primary tumor,
with biochemical recurrence of prostate cancer measured with serum the more aggressive is the staging procedure required. In most instances,
PSA, and imaged with sequential F choline and, if negative, gallium the primary tumor must be surgically resected for palliation (or cure),
PSMA scanning, the overall detection rates were 85.6% (107 of 125) even if metastatic tumor is present. For staging, the most common
for the sequential imaging approach and 74.4% (93 of 125) for studies include CT scan with contrast enhancement of the abdomen
18
F-choline-PET-CT alone. 68Ga-PSMA-PET-CT detected sites of and pelvis, CT scan of the thorax (or chest radiograph), and bowel
recurrence in 43.8% (14 of 32) of the choline-negative patients. imaging to exclude the presence of a second primary colon cancer.
Detection rates of the sequential imaging approach and 18F-choline- In institutions in which PET imaging is available, PET is used somewhat
PET-CT alone increased with higher serum PSA levels. Subgroup more frequently at presentation; however, it is much more commonly
analysis of 68Ga-PSMA-PET-CT in 18F-choline–negative patients applied in the setting of suspected recurrence.
revealed detection rates of 28.6%, 45.5%, and 71.4% for PSA levels For colorectal cancer, many advocate regular imaging studies after
of 0.2 or greater to less than 1 ng/mL, 1 to 2 ng/mL, and greater surgery for “cure,” because isolated metastases or oligometastases of
than 2 ng/mL, respectively.84 Several 18F-labeled small molecules, such colorectal cancer can be resected from the liver or lungs; in some
as 18F DCFBC, which binds to PSMA, and successor molecules such instances the patient is disease free for a long period. Thus before
as 18F DCFPYL, result in exceptionally high-quality images of metastatic such removal of a limited number of metastases is contemplated, a
prostate cancer.85–87 Thus there is considerable progress in the molecular thorough imaging procedure is performed. This usually includes CT
imaging of prostate cancer, with agents approved and under develop- of the abdomen and pelvis with contrast agent and CT of the thorax
ment that are likely to alter practice patterns in prostate cancer, especially without contrast agent. CT of the thorax may be replaced with a
metastatic disease assessments. chest radiograph, but chest radiographs are less sensitive for pulmonary
metastases.
Colon Cancer PET is applied very often in this setting and in the setting of a
rising carcinoembryonic antigen level after surgery. The precise timing
The role of noninvasive imaging in the diagnosis of the primary lesion of follow-up studies can be variable, but they often are done every 6
in colon cancer has changed over the last several decades. At one time, to 12 months in the early years after surgery. Considerable evidence
barium enema studies were used extensively to search for colorectal supports the idea that PET can be used to detect more metastatic foci
cancer. They have been replaced, in large measure, by fiberoptic than CT in the setting of a rising carcinoembryonic antigen level.90
colonoscopy. However, there is a growing level of interest in virtual An example of a patient initially believed to have only a limited
colonoscopy, which is performed with use of CT scanning and per- number of liver metastases is shown in Fig. 16.11; however, more
rectum insufflation after thorough bowel preparation. This procedure extensive disease was identified (Fig. 16.11B). PET-CT has been shown
is used to a limited extent for screening. The technique faces challenges to offer higher accuracy than PET alone in recurrent colorectal cancer.91
because it requires bowel preparation and the interpretation is quite Although immunoscintigraphy has been used, it has been rendered
time-consuming. Screening for colon cancer is an area of considerable essentially obsolete because of the availability of FDG-PET. FDG-PET
opportunity for noninvasive imaging. Infrequently, these cancers can often does not detect small (<5 mm) tumors, however, and is known
be detected by other methods, such as ultrasonography. to be less sensitive for tumors of mucinous histology, so opportunities
Virtual colonoscopy was directly compared with optical colonoscopy for improvement remain.
in a study of over 6000 patients. Patients were randomized to either Ultrasonography can reveal many liver lesions and is a very useful
an optical or a CT colonography group and were compared for the technique for guiding biopsies of the liver.29 For liver metastases, CT
detection of advanced neoplasia and the total number of harvested is still the most commonly used procedure, but in a meta-analysis,
polyps. Advanced neoplasia was confirmed in 100 of the 3120 patients PET was a more robust test to identify the presence and location of
in the CT colonography group (3.2%) and in 107 of the 3163 patients hepatic metastases of colorectal cancer compared with CT, MRI, or
in the optical colonography group (3.4%). There were seven colonic ultrasound methods.92
perforations in the optical colonography group and none in the CT MRI methods have continued to improve, and MRI likely can
colonography group. Primary CT colonography and optical colonog- reveal more lesions in the liver than CT or PET. Thus for small lesion
raphy screening strategies resulted in similar detection rates for advanced detection, MRI may offer advantages over PET, but at the cost of
neoplasia, although the numbers of polypectomies and complications increased false-positive results.93
were considerably smaller in the CT colonography group. CT colo-
nography also was much less expensive than optical colonoscopy.88 Gynecologic Neoplasms
Interesting to note, the detection rate for polyps requiring biopsy was
statistically identical between the two groups of patients. Screening for gynecologic neoplasms usually is not performed with
In a study of 2531 evaluable participants screened with virtual imaging, except for very limited programs that have evaluated the use
colonoscopy for large adenomas and cancers, the mean with standard of either transabdominal or, more commonly, transvaginal ultrasound
error for per-patient estimates of the AUC for CT colonography was of the pelvis to detect masses that may represent ovarian cancer. These
0.89 ± 0.02. The sensitivity of 0.90 (i.e., 90%) indicates that CT programs have been combined with serum biomarkers. Such programs
colonography failed to detect a lesion measuring 10 mm or more in have not been proven cost-effective and are not widely applied, but
diameter in 10% of patients. The per-polyp sensitivity for large adeno- they warrant further study, because this is a very important health
mas or cancers was 0.84 ± 0.04. The per-patient sensitivity for detecting problem. In women with high genetic risk of ovarian cancer, such as
adenomas that were 6 mm or more in diameter was 0.78.89 those with the BRCA mutation, screening may have greater value.
274 Part I: Science and Clinical Oncology

A B
Figure 16.11  •  Colon cancer. (A) Positron emission tomography (PET) and computed tomography (CT) image display of a patient with two fluorine-18
(18F)–fluorodeoxyglucose (FDG)–avid lesions in the liver. These are seen on the CT scan (upper left), the attenuation-corrected PET scan (upper right), the
non–attenuation-corrected PET scan (lower right), and fused images (lower left). (B) PET and CT images of the pelvis, oriented as in part (A), show increased
FDG uptake in a left external iliac lymph node metastasis.

In cervical carcinoma, screening with use of the Pap smear has a


large effect in terms of lowering mortality rates by enabling detection
of premalignant changes and early-stage disease. Although much of
the staging of cervical carcinoma is performed through physical
examination, for larger primary tumors, imaging has an important
role. Both CT and MRI are used in the pelvis; however, the use of
PET for tumor staging is increasing. As in other tumors, PET appears
to be more sensitive than anatomic imaging methods. Emerging data
show that PET provides better prognostic value than anatomic imaging
in cervical cancer.94–96
In ovarian cancer, no technique is able to reveal microscopic
metastatic disease. Ultrasound examination is the main method by
which ovarian tumors are identified at their earliest stages; however,
the unfortunate fact is that these tumors are usually diagnosed at an
advanced stage. Imaging can be used in an attempt to determine the
extent of the surgical procedure that will be required. Both CT and
MRI are used to assess the extent of ovarian cancer, with CT the
preferred method (Fig. 16.12). PET is not sensitive to tumor foci
smaller than 5 mm, but it is reasonably reliable in detecting larger
tumor foci. For this reason, some advocate the use of PET to determine
whether tumor debulking should be performed. PET has a role in
the setting of a rising CA125 level in patients with normal CT Figure 16.12  •  Axial computed tomography (CT) image shows a dense,
findings.97 calcified mass in the left pelvis, compatible with a metastatic implant from
The use of serum markers and imaging is recommended for surveil- ovarian cancer.
lance after surgery for ovarian carcinoma. The aggressiveness of imaging
follow-up depends on the treatments available. PET has an emerging
role in this setting, although practice patterns vary widely. PET also
has been shown to provide information related to treatment response 20% more lesions than are seen with CT alone.101 PET can demonstrate
in preliminary studies.98,99 disease in the bone marrow and spleen in some instances. In fact,
marrow involvement seen on FDG-PET often is associated with a
Lymphoma negative CT scan. Because the negative predictive value of a negative
PET scan of the marrow in a patient with Hodgkin lymphoma is
For both Hodgkin and non-Hodgkin lymphomas, accurate staging high, omission of bone marrow biopsy is a reasonable consideration
is important. For both types of lymphoma, accurate definition of the in PET-negative patients.102,103
tumor burden is needed for effective treatment planning, especially CT is traditionally used for follow-up of lymphoma, and there are
treatment with external beam radiation. CT was the historically accepted clear response criteria in place to follow lymphoma therapy. One of
method for noninvasive staging of lymphoma, with PET being used the challenges in lymphoma is that large masses often do not normalize
increasingly because many studies have shown it to be capable of in size after treatment, leading to questions in interpretation of a
locating more tumor foci than CT.100 An example of CT imaging of residual mass lesion. Determining whether these lesions contain a
abdominopelvic lymphoma is shown in Fig. 16.13. In most lymphoma viable tumor is important, because it defines whether more treatment
histologic types, FDG-PET is more sensitive than CT, often showing is needed.
Imaging  •  CHAPTER 16 275

A B

C
Figure 16.13  •  (A) Axial computed tomography (CT) image with oral and intravenous contrast agent shows paraaortic adenopathy and infiltration of
the left kidney by lymphoma. (B) Image of the lower abdomen in the same patient shows enlarged mesenteric nodes. (C) Positron emission tomography
(PET)–only images (left to right: coronal, sagittal, transverse, and anterior projections) show focal areas of increased fluorine-18 (18F)–fluorodeoxyglucose
(FDG) uptake in multiple lymph node groups, including the axillary, inguinal, and iliac regions, consistent with disseminated lymphoma.

Data indicate that gallium-67 (67Ga) scintigraphy can be of growing usefulness. A limitation of PET-only methods had been the
effective for assessing the viability of residual Hodgkin lymphoma lack of a standardized set of response criteria. Tracer activity in PET
and intermediate- and high-grade non-Hodgkin lymphoma. However, images typically decreases more rapidly than tumor shrinkage occurs (i.e.,
the results of multiple studies on FDG-PET in this setting now have anatomic changes of treatment lag behind metabolic changes detectable
been published, and this imaging modality has essentially replaced with PET). For these reasons, PET scans may appear normal before
67
Ga scintigraphy in assessing the viability of residual masses of CT scans do. A concern is that there is not strong evidence showing
lymphoma. If a residual mass of lymphoma shows increased FDG that a negative PET scan should be used to truncate the duration
uptake, that usually is indicative of residual viable tumor; however, of lymphoma therapy. For example, if a PET scan became negative
scans also can be falsely positive because of inflammation and falsely after two cycles of treatment, this would not justify, based on current
negative, likely because some tumor foci may be smaller than the data, discontinuation of treatment. However, if a treatment involved a
resolution of PET imaging.104,105 standard of four possible courses of treatment and PET findings became
Positive midtreatment 67Ga and PET scans predict a poor outcome negative after these courses were completed, available data suggest that
from therapy, and positive PET scans at the conclusion of a therapeutic this portends a very good prognosis compared with a positive PET
regimen also indicate a poor prognosis. It is increasingly appreciated scan. New criteria for response including PET have been developed
that PET findings soon after treatment is initiated can be highly for lymphoma (International Workshop Criteria [IWC] + PET). For
predictive of ultimate outcome of the therapy. Indeed, a rapid decline FDG-avid lymphomas, a negative PET scan is required to determine
in FDG uptake to background levels after one cycle of therapy indicates a complete response. The five-point Deauville and Lugano score has
a highly responsive patient.106–108 assumed widespread use in lymphoma imaging. Caution is in order
Challenges associated with PET include reactive lymph nodes and in patients who have received immune checkpoint treatments.110–115
nodes involved with inflammatory processes such as sarcoidosis, which It can be argued that PET is not essential in all patients with
can be very FDG avid. Similarly, uptake of FDG in brown fat in the lymphoma. However, the use of PET and PET-CT is increasingly
neck and thymus can be somewhat confusing.109 becoming the norm in centers where this technology is available.
Although anatomic imaging has been the key for lymphoma assess- Because PET can find some lymphomatous tumors that cannot be
ment, there is a growing trend toward greater use of PET. PET imaging detected with CT, PET is increasingly finding routine application in
and PET-CT imaging, providing additional functional information, are the care of patients with lymphoma (see Fig. 16.13C).
276 Part I: Science and Clinical Oncology

An exciting new opportunity for the use of PET in non-Hodgkin metastases of melanoma, but it is not widely applied and is also less
lymphoma is in the setting of a “response adaptive” approach. In such sensitive than sentinel node imaging.1 However, PET can show most
treatment approaches, patients who are poorly responding can be tumor foci larger than 6 mm and sometimes can reveal smaller tumor
identified with PET performed soon after treatment is initiated. This foci. CT is a more robust technique for small pulmonary nodules
approach can allow segregation of responding from nonresponding than is PET.120
patients. In the responding patients, standard treatment is given, Melanoma metastases, especially if they are localized, can be resected
whereas in the poorly responding patients, alternative approaches such surgically. However, identifying whether only one or two versus many
as stem cell transplants are used. This approach currently is investi- metastases are present is a major challenge. Aggressive surgical pro-
gational, but it is an area of great promise because it allows the potential cedures are not appropriate if there are disseminated metastases.
for tailoring the therapy to the individual patient’s responsiveness to Therefore staging imaging procedures are performed aggressively before
the treatment algorithm.116 Recently, use of such an approach has major surgery is undertaken to resect melanoma metastases. PET
been shown feasible in a clinical comparative trial on Hodgkin commonly is part of such a staging evaluation.
lymphoma.117 For metastatic melanoma, PET has been reported to have sensitivity
The possibility of increasing dose in patients identified as being well over 90%. Thus although anatomic imaging dominates, PET
at high risk of progression and decreasing therapy in low-risk groups has a growing role in melanoma assessment.121 For bone metastases,
is being tested more commonly. In a Hodgkin lymphoma trial involving radionuclide bone scanning also is an important diagnostic procedure.
602 patients, 571 underwent PET scanning posttreatment. The PET Recently, it has been shown that the performance of FDG-PET in
findings were negative in 426 of these patients (74.6%), 420 of whom melanoma is significantly enhanced if the CT scan portion of PET-CT
were randomly assigned to a study group (209 to the radiotherapy is analyzed very carefully. This appears to increase both sensitivity and
group and 211 to no further therapy). The 3-year progression-free specificity of the method, Caution is in order for assessing melanomas
survival rate was 94.6% (95% CI, 91.5–97.7) in the radiotherapy undergoing immune checkpoint inhibitor therapies. Increases in FDG
group and 90.8% (95% CI, 86.9–94.8) in the group that received uptake can be seen soon after treatment is started, yet can be associated
no further therapy. Death rates were comparable. This suggests that with a favorable response. Repeat imaging is often in order122 before
PET can be used to deintensify therapy of Hodgkin disease, as long disease is considered nonresponsive.123
as patients are managed for recurrence, which is slightly more common
in the nonirradiated group. A concern with FDG-PET in lymphoma Bladder Carcinoma
is false-positive results due to inflammation. It is important to note
that both false-positive and false-negative PET study findings can Bladder carcinoma often manifests at an early stage, and no imaging
occur in patients with lymphoma. evaluation is performed to determine whether there are locoregional
PET is changing the therapy of lymphoma, and this field is in or systemic metastases. However, ultrasound examination has been
rapid evolution. The ability to measure total tumor volumes and used to determine the depth of penetration of primary bladder car-
modify therapies based on PET is leading to a wide range of studies cinomas. An important consideration in bladder carcinomas is that
including dose intensification. Some are discussed in more detail in uroepithelial tumors are often multicentric. Thus intravenous pyelogram
the lymphoma therapy chapter elsewhere in this book. PET with examinations to evaluate the entire genitourinary system commonly
FDG is markedly changing and routinely informing treatment of are performed early in the diagnostic algorithm. Although ultraso-
patients with lymphoma. nography can reveal many bladder cancers 5 mm and larger, trans-
urethral sonography is more sensitive; obviously, however, it is invasive.
Melanoma MRI is used more commonly than CT for assessing primary bladder
lesions because of its superior soft tissue contrast characterization
The imaging management of melanoma varies based on the stage of abilities.124
disease. Radiologic imaging has no significant role in the diagnosis For larger primary tumors that are invasive, imaging evaluation
of primary melanomas. Lymphoscintigraphy—the injection of is important to help to determine whether surgery is appropriate.
radiolabeled colloidal material, typically 99mTc-sulfur colloid, into the Metastatic disease to locoregional nodes or systemic metastases indicates
subcutaneous tissues or intradermally to locate lymphatic drainage disease with a poorer prognosis, and tumor invading local structures
routes, and thus lymph nodes with the potential for metastatic or metastatic either to nodes or systemically often is considered
involvement—commonly is performed for primary melanomas of unresectable. CT is used most commonly for local nodal staging,
intermediate thickness. Although practice patterns vary, this method but MRI also can be used. Small nodal metastases usually are not
typically is used for melanomas that are thicker than 1 mm without detected with CT, because they have not enlarged the lymph nodes
other evidence of metastases. If the sentinel node identified at surgery sufficiently to allow the tumor-involved nodes to be detectable.
(often with radionuclide guidance) is involved with tumor, additional CT has a reported sensitivity of 60% to 70%. Biopsy commonly
staging procedures often are done.118 is used to determine whether an enlarged node seen on CT truly
Data suggest that SPECT-CT imaging may improve the accuracy contains tumor.
of the sentinel lymph node procedure and reduce the probability of MRI probably is more sensitive than CT in detecting nodal
recurrence of disease in a lymph node basin119 versus more standard metastases, and new contrast agents that accumulate in normal nodes
sentinel node identification procedures. These procedures most com- are potentially important for enhancing the diagnostic accuracy of
monly include CT of the chest and abdomen, and of the pelvis if the MRI in detecting nodal metastases.125 Initial data with MRI contrast
melanoma affected the lower extremities. If there are systemic metastases, agents support the accuracy of this approach, but also point out that
brain imaging is performed with MRI with and without gadolinium the interpreter’s experience is important.126 Such agents are not yet
contrast enhancement. routinely available or FDA approved.
PET and PET-CT with FDG also are potent methods for detecting FDG-PET has been used and is promising in bladder carcinoma;
metastatic melanoma, often revealing more tumor foci than CT. PET however, images of the pelvis can be degraded by intense 18F activity
can show nodal metastases but is not as sensitive as sentinel node in the bladder. The use of PET in bladder cancer is enhanced by
imaging and is not a replacement for sentinel node imaging and PET-CT and iterative reconstruction methods that make for better
removal. PET is particularly good for soft tissue metastases but cannot assessments of the pelvis with less degradation due to the bladder
reveal microscopic disease. Thus sentinel lymph node biopsy is used radiotracer activity. For bone metastases, radionuclide bone scan remains
in preference to PET for detection of early metastases. There is some the procedure of choice, and MRI also can be sensitive. PET-CT is
evidence that ultrasonography can be used to detect small nodal quite sensitive for metastatic disease beyond the pelvis and is being
Imaging  •  CHAPTER 16 277

applied to a greater extent because of the results of the CMS registry enhancement (Fig. 16.14A). Because MRI is subject to respiratory
in the United States.127 and motion artifacts, CT is used somewhat more commonly in the
Follow-up of patients with bladder carcinoma usually involves the initial staging of these tumors.128
use of CT scans. Follow-up for new or recurrent bladder carcinoma CT and MRI can characterize the extent of primary tumors; however,
within the bladder usually requires direct visualization of the bladder CT is more effective than MRI in assessing the extent of involvement
with cystoscopy. of cartilage or bone. For nodal metastases, current diagnostic schemes
are based mainly on nodal size. Nodal size is an imperfect indicator
Head and Neck Cancer of tumor involvement, however.
Increasingly, FDG-PET is being applied to the assessment of head
Head and neck cancers often are associated with cigarette smoking and and neck cancers. Although several studies have suggested that PET,
alcohol use or abuse. Human papillomavirus also has been linked to MRI, and CT have similar sensitivity, more recent studies have suggested
head and neck cancers, especially in younger women. Most malignancies that PET is more accurate in staging (Fig. 16.14B–C). However, PET
in the head and neck (except for lymphomas, as discussed earlier) are can demonstrate increased glucose uptake in nonmalignantly involved
of squamous cell etiology. A key issue in these lesions is determining inflamed nodes (false-positive findings). These can occur in patients
whether the disease is localized to the head and neck or whether with head and neck or gingival infection or the common cold. Defining
metastatic disease or a second primary lesion is present. Thus imaging of the precise extent of tumor is important to determine whether surgery
the lungs usually is done to exclude a primary or metastatic lung tumor. or radiation therapy should be performed, because more extensive
The physical examination is very important in assessing a primary tumors are less amenable to surgical resection.
lesion in the head and neck, but both CT and MRI are very potent Occasionally, head and neck cancers manifest as isolated nodal
methods. MRI typically is performed before and after gadolinium metastases without the location of the primary tumor being evident.
contrast is administered; CT scanning usually is performed after contrast Imaging has a role in such cases. Often MRI is performed in addition

A B

Figure 16.14  •  Head and neck tumors. (A) Axial computed tomography (CT) image of the neck with intravenous contrast agent shows a mass in the
left piriform sinus. (B) Positron emission tomography (PET) and PET-CT images show right vocal cord cancer with intense tracer uptake. (C) PET and
PET-CT images from the lower level of the neck show nodal tracer uptake consistent with metastases. (Courtesy Dr. David Yousem, The Johns Hopkins
University.)
278 Part I: Science and Clinical Oncology

to extensive inspection and biopsies; however, FDG-PET also has They sometimes are challenging to assess, because the appearance of
been applied. This method can be used to detect as many as 15% to cirrhosis with regenerating nodules and tumors can overlap. Multiphase
30% of primary tumors. CT imaging, CT angiography, and MRI with and without gadolinium
For recurrent tumors, PET appears to be a more robust test than contrast material are commonly used in hepatomas. PET is less reliable,
MRI or CT, especially when timed properly, probably because contrast because approximately half—and sometimes more—of hepatomas are
enhancement can be seen in both postoperative changes (and postradia- not FDG avid. There has been some use of alternative PET tracers
tion tissue) and tumors. In general, FDG uptake is a more reliable such as C-11 acetate to image some hepatocellular cancers, because
predictor of tumor than the anatomic methods. PET is useful in some FDG-negative tumors are C-11 acetate avid.137 Ultrasonography
surveillance for the recurrence of these tumors, but it is not yet also can be used to assess the liver and guide biopsies.
considered the standard of care. Metastatic lesions to the liver also are common, especially in the
An extremely important trial of PET as a tool to inform cancer United States. For most tumors, CT is the initial method used for
management was reported by Mehanna and colleagues.129 They prospec- assessing whether tumor is present. However, FDG-PET is more
tively studied 564 patients with newly diagnosed head and neck cancer sensitive than CT in detecting liver metastases in common cancers
who would typically have been managed by chemoradiation followed such as colorectal cancer.90,92 Thus PET is seeing greater application
by surgery. In this study, patients were randomized (282 patients to in assessing suspected liver metastases, although ultrasonography, CT,
the planned-surgery group and 282 patients to the surveillance group) and MRI also are important methods and still are more commonly
from 37 centers in the United Kingdom. A total of 84% of the patients applied in many centers (Fig. 16.16).
had oropharyngeal cancer, and 75% had tumor specimens that stained
positive for the p16 protein, an indicator that human papillomavirus Kidney Cancer
had a role in the causation of the cancer. The median follow-up was 36
months. The PET-CT–guided surveillance patients (notably those with In the past, renal cancers were detected with intravenous pyelograms;
a negative PET scan after chemoradiotherapy) underwent fewer neck currently, however, the most common method for detection is CT.
dissections than did the patients randomized to the planned dissection Renal cell cancer commonly is detected incidentally because of the
surgery (54 versus 221, respectively); rates of surgical complications widespread use of cross-sectional imaging. Between 25% and 50% of
were similar in the two groups (42% and 38%, respectively). The surgically treated renal cell cancers are discovered incidentally.138 Renal
2-year overall survival rate was 84.9% in the surveillance group and lesions are classified as cysts or solid masses, depending on their
81.5% in the planned-surgery group. The hazard ratio for death slightly characteristics as shown by imaging. Renal cysts are fluid-filled and
favored PET-CT–guided surveillance and indicated noninferiority appear anechoic with increased through-transmission on ultrasound
(upper boundary of the 95% CI for the hazard ratio, <1.50; P = examination. They show water density without enhancement on CT,
.004). Quality of life was similar in the two groups. PET-CT–guided and appear hyperintense on T2-weighted images, also without enhance-
surveillance, as compared with neck dissection, resulted in savings ment, on MRI.
of £1492 (approximately $2190 in US dollars; as reported in 2016) Renal masses typically are evaluated with CT because of its short
per person over the duration of the trial.130 Therefore PET guidance examination times and ease of evaluation, even in patients with a
of treatment in head and neck cancer has strong evidence of “value.” large body habitus, which can make ultrasonography difficult. MRI
For head and neck cancers, MRI offers excellent contrast resolution typically is used for problem solving. Multiphasic scanning is performed
for soft tissues, but images can be degraded substantially by motion. with CT to evaluate the density of lesions before administration of
For this reason, CT with contrast is much more commonly performed. contrast agent and as contrast material filters from the cortex into the
FDG-PET is assuming a growing role in cancer management, especially medulla and collecting system. An increase in density of greater than
for recurrence and for assessment of response to treatment. PET with 20 HU corresponds to lesion enhancement and confirms the presence
CT is being used more often to stage and monitor these tumors during of a solid mass.
and after treatment and may become the standard of care.131 A recent CT is used to stage the tumor by determining the presence of
comparison of PET-CT with MRI and PET alone showed superior renal vein invasion, adenopathy, local extension, and distant metastases.
accuracy of PET-CT versus the other methods.132 The accuracy of CT for staging is 91%.138 For resectable lesions, CT
can provide information on whether the lesion is amenable to nephron-
Pancreatic Carcinoma sparing surgery or partial nephrectomy. Lesions that are smaller than
4 cm, polar, and cortical and do not involve the renal hilum or collecting
The standard of care for the imaging diagnosis of pancreatic cancers system may be candidates for partial nephrectomy or ablation.
is CT scanning. Although MRI can be useful, CT, including CT Although CT, MRI, and ultrasonography can all be used to assess
angiography, is the main method used for staging and assessing tumor renal lesions, CT with contrast is the dominant method (Fig. 16.17).
invasion of vessels (Fig. 16.15). Some studies have shown FDG-PET PET is useful only when the tumor is FDG-avid. However, the normal
to be somewhat more sensitive for detection of tumors and to have excretion of FDG by the kidneys makes evaluation of the kidneys
moderately high accuracy—approximately 85%—in characterizing more challenging than other tissues, and some renal masses are not
pancreatic lesions as malignant or benign.133–135 Ultrasound examination very FDG avid. Thus FDG-PET is not currently recommended for
is used to assess cystic lesions. After treatment, salvage therapy is renal cancers, at least not for reliably characterizing renal masses as
ineffective, and these patients often are less aggressively monitored malignant or benign. Metastatic renal cancer is more accurately imaged
than those with other, more treatable cancers. on FDG-PET than is primary renal cancer.139,140
Neuroendocrine tumors of the pancreas often can be detected with Renal cancers can also be imaged by using iodine-124 (124I)–labeled
use of 111In-pentetreotide scanning (OctreoScan), a SPECT procedure. monoclonal antibodies to carbonic anhydrase IX, which is overexpressed
Positron emitter–labeled analogues of somatostatin have been developed in clear cell renal cancers. A prospective open-label multicenter study
and show encouraging biologic characteristics compared with single- of 124I-girentuximab PET-CT in patients with renal masses who were
photon agents.136 The FDA has approved Ga 68 dotatate (NETSPOT) scheduled for resection was performed and PET-CT and contrast-
for imaging NET. enhanced CT of the abdomen were compared. In 195 patients, complete
data sets (histopathologic diagnosis and PET-CT and contrast-enhanced
Liver Cancer CT results) were available. The average sensitivity was 86.2% for
PET-CT and 75.5% for contrast-enhanced CT (P = .023). The average
Hepatic malignancies, especially hepatomas, are common worldwide. specificity was 85.9% for PET-CT and 46.8% for contrast-enhanced
Both CT and MRI can be very effective in detection of these lesions. CT (P = .005). These data suggest this investigational agent may be
Imaging  •  CHAPTER 16 279

A B

C D
Figure 16.15  •  Pancreatic cancer. (A) Axial computed tomography (CT) image shows a mass in the pancreatic body, encasing the splenic artery.
(B) Coronal volume-rendered three-dimensional CT image shows tumor encasing the splenic and left gastric arteries. (C) Coronal volume-rendered maximum-
intensity projection. (D) Sagittal volume-rendered three-dimensional CT images show a mass in the pancreatic head without invasion of the superior mesenteric
artery or vein. A common bile duct stent also is seen.

111
useful for non-invasive phenotyping of renal masses as malignant In pentetreotide is FDA approved and used in detecting neu-
or benign.141 roendocrine tumors. 68Ga-labeled DOTA-TOC and related compounds
are typically more effective than 111In-labeled compounds and are
Endocrine Tumors available in a variety of settings. These agents are typically more accurate
than 111In OctreoScan. A meta-analysis of the 68Ga peptide imaging
Imaging is used to study several types of endocrine tumors in a variety literature showed the AUC in the task of diagnosing somatostatin
of locations. For adrenal tumors, CT is the procedure of choice, with receptor–expressing neuroendocrine tumors in 16 studies comprising
metaiodobenzyl-guanidine 123I (MIBG) scanning and MRI scanning 567 patients. The pooled AUC was 0.96. Since FDA approved in the
also proving useful for lesion characterization.140 MIBG accumulates United States, these Ga 68 dotatate techniques, where available, should
selectively in pheochromocytomas. Adrenal masses with low HU values be considered as first-line diagnostic imaging methods in patients
(<10 HU) typically are adenomas, which are lipid rich. with suspected neuroendocrine tumors.136,143,144
For the thyroid gland, radioiodine imaging commonly is used. For
non–radioiodine-avid thyroid cancers, FDG-PET is very useful for Brain Tumors
lesion detection and is recommended in the setting of a rising serum
thyroglobulin level with a normal 131I or 23I scan, particularly when The dominant method for the assessment of brain tumors is the MRI
recombinant thyroid-stimulating hormone (TSH) stimulation is used scan, which is the preferred method for initial detection, assessment
(Fig. 16.18).142 For neuroendocrine tumors such as carcinoid tumors, of extent of disease, and assessment of efficacy of therapy. The superior
CT and radiolabeled octreotide analogues are very useful, especially soft tissue contrast provided by MRI makes it superior to CT for
the newer Ga 68 agents. lesion characterization (Fig. 16.19).
280 Part I: Science and Clinical Oncology

A B

C
Figure 16.16  •  (A) Carcinoid metastases to the liver. Axial magnetic resonance image with gadolinium shows multiple enhancing masses in both lobes
of the liver. (B) Hepatoma. Axial magnetic resonance image with gadolinium shows a solid mass in the left lobe of the liver. (C) Intraductal papillary mucinous
tumor of the pancreas. Axial T1-weighted magnetic resonance image of the abdomen shows a low–signal intensity cystic mass in the head of the pancreas.
(Courtesy Dr. Ihab Kamel, The Johns Hopkins University.)

Figure 16.17  •  Axial computed tomography (CT) image of the abdomen


with intravenous contrast agent shows enhancing renal cell carcinoma in the
right kidney. Enhancing tumor invades the right renal vein.
Figure 16.18  •  Positron emission tomography (PET) and computed
tomography (CT) image panel showing an intense fluorine-18 (18F)–fluoro-
deoxyglucose (FDG) uptake focus near clips in the left thyroid bed, consistent
with recurrent thyroid cancer.
Imaging  •  CHAPTER 16 281

wall of the esophagus or involved the periesophageal nodes. PET


scanning is very sensitive in determining whether stage IV disease is
present, and typically is performed as part of the initial staging evalu-
ation, at least for esophageal cancer, to identify patients who are clearly
not candidates for surgery.149 FDG-PET also is being used with
increasing frequency to assess treatment response in patients with
esophageal cancer, because early changes in glycolysis after treatment
appear to be related to treatment efficacy.150

Sarcomas
MRI is the method of choice for detection and assessment of soft
tissue sarcomas. Gadolinium contrast enhancement is also commonly
used. FDG-PET imaging has been useful in assessing primary sarcomas
for aggressiveness and defining possible sites for biopsy in addition
to defining prognosis and grade.151,152 It also has been used to assess
and predict the response of sarcomas to therapy. Early (10 days into
treatment) FDG-PET scans show changes in metabolism predictive
of outcomes, with a rapid decline in FDG uptake associated with a
more favorable survival with some treatments.153,154

Figure 16.19  •  Coronal magnetic resonance image of the brain with


gadolinium shows a mass with peripheral enhancement (arrows), compatible
Gastrointestinal Stromal Tumors
with tumor, and a more hypointense necrotic area. GI stromal tumors are relatively infrequent, but have been studied
extensively with both CT and PET imaging in the past few years.
These tumors often are responsive to imatinib and other tyrosine
Unfortunately, even sophisticated MRI techniques, often relying kinase inhibitors, in part because of their constitutive overactivity of
on tumor enhancement with gadolinium, cannot be used to detect a mutated KIT oncogene. These tumors have been shown to be typically
microscopic disease. MRI findings, although fairly specific for tumor, FDG avid. FDG accumulation declines very rapidly with effective
are not completely specific. Thus infarcts, infections, and foci of treatments, earlier than changes in tumor size. Of interest is that
demyelination occasionally can mimic tumor foci on MRI. FDG-PET revised CT criteria may allow CT to assess response more quickly to
has a very limited role but can be useful in assessing residual tumor treatment in such tumors, with changes in tumor Hounsfield units
after radiation therapy and determining whether residual masses are being one of the characteristic findings of response. Both PET and
caused by tumor or tumor necrosis. It is most accurate in highly CT thus have a role in this somewhat uncommon tumor, especially
aggressive tumors in which brain tumor uptake of FDG is greater for the early assessment of treatment response.155
than that of normal brain tissue. Other tracers, such as FDOPA, FET,
and FLT, have shown results more promising than those for FDG.145,146 TREATMENT RESPONSE ASSESSMENT
MRS also can be useful in evaluating this issue because tumors
typically have high levels of choline and low levels of N-acetylaspartate. The use of imaging to assess treatment response is common. The
Angiography, although a historically useful method, is performed less precise best time to assess response depends on the treatment, its
frequently for diagnostic purposes in brain tumors. MRI, CT, and toxicities, and the alternative therapies available. Typically, the same
PET can guide biopsies. Furthermore, MRI can be used intraoperatively methodology used at baseline is used to follow the response of the
to guide therapy.147 cancer to treatment. Early diagnosis of failure of response is a key
goal of treatment response assessments. However, typically response
Pediatric Tumors is assessed after two or more cycles of treatment when anatomic methods
are used.
CT usually is the method of choice for evaluation of tumors in children. Functional methods such as PET with FDG or possibly MRI with
For tumors of the central nervous system and for sarcomas, MRI is diffusion assessments may provide earlier indices of response than
preferred. FDG has a growing role, especially for lymphomas and seen with size measures alone. Diffusion and DCE MRI are quite
sarcomas. MIBG scanning often is used in neuroblastomas. CT scanning promising methods, but standardization of diffusion MRI is in its
should be performed with a reduced tube current and energy to early phases, although it seems effective in single-center studies,
minimize the radiation dose while preserving the quality of the including in brain tumors.156
diagnostic image. There are systematic response criteria available for anatomic imaging
Multiphase CT should be avoided in children, unless it is clearly (most recently, Response Evaluation Criteria in Solid Tumors [RECIST]
indicated, to minimize the radiation dose to the child.148 PET-CT 1.1) and proposed criteria for PET response, the European Organisation
has shown broad usefulness in pediatric cancers, and the evolving for Research and Treatment of Cancer (EORTC) criteria and the
literature suggests that PET-CT often offers improvements over CT more recent Positron Emission Tomography Response Criteria In Solid
alone. Given the radiation dose and long-term risks of carcinogenesis, Tumors (PERCIST) 1.0.157,158 To use quantitation in imaging, close
it is possible that MRI will have a growing role in pediatric cancer attention to detail is required in the imaging process. CT or MRI
imaging owing to its non-ionizing nature. PET-MRI, a newer technol- can provide tumor sizes. PET can provide a standardized uptake value
ogy, may be particularly attractive in children. (SUV), which can be used to phenotype tumors and to quickly assess
treatment response, but such values may vary from device to device
Esophageal and Gastric Cancer and over time, unless careful standardization is followed. Response
criteria for lymphoma are discussed earlier in this chapter. A challenge
CT scanning is the method of choice for initial staging of esophageal in treatment response is deciphering responses to immunotherapy
and gastric cancer. Endoscopic ultrasonography is the most sensitive with checkpoint inhibitors and other treatments. With these treatments,
and accurate method for determining whether tumor has invaded the transient increases in tumor size, metabolic activity, and lesion number
282 Part I: Science and Clinical Oncology

can be seen before a response or in association with stable disease. Catheter-based delivery systems also are important. For example,
This phenomenon is known as “pseudoprogression.”159 Thus some angiographic catheters can be used to deliver regional chemotherapy,
caution is in order in assessing mild tumor size increases or increased emboli, or radioactive or chemical microspheres to treat tumors.
metabolism in patients on checkpoint inhibitor therapy.160,161 Radioactive microspheres also are assuming a growing role in inter-
ventional oncologic radiology.164
DEFINING NORMAL ORGAN FUNCTION FOR
CANCER THERAPY EMERGING OPPORTUNITIES IN IMAGING
Several tests are used to determine whether a patient is a suitable Functional imaging methods such as PET and various innovative
candidate for aggressive therapy. These tests include myocardial perfu- MRI techniques are being used increasingly to assess treatment response
sion imaging at stress to determine whether ischemia is present, because early after treatment is begun.30 Beyond this, a variety of imaging
if present it could increase the risk for a major surgical procedure. methods are being developed to image key aspects of tumor biology
Echocardiography also is used for this purpose. Myocardial function (Table 16.4). An exciting area for both MRI and PET is in detection
often is evaluated before chemotherapy is given. This may be done of hypoxia, which is common in a broad range of malignancies and
by using a myocardial blood pool study or, less commonly, an echo- which represents a possible target for cancer treatments.165 In addition
cardiogram to determine chamber size and ejection fraction. to hypoxia, tumor perfusion can be imaged with a variety of approaches.
Pulmonary function usually is determined by pulmonary ventilatory Even though the process is difficult, interest remains in gene
function tests; however, split lung function and regional function may therapeutic approaches, which has led to attempts to determine, through
be determined with pulmonary perfusion imaging with 99mTc MAA imaging, whether genes, when delivered, actually reach tumors and,
(macroaggregated albumin; i.e., a quantitative lung scan). Regional more importantly, whether they express in vivo the desired levels of
ventilation also can be assessed quantitatively. Such determinations gene product expected to be required to achieve a therapeutic effect.
help to predict the level of pulmonary function expected after surgery. For example, dopamine receptors have been transfected into cells and
Split assessment of renal function sometimes is performed before imaged with radioligands capable of binding to the D2 dopamine
removal of a renal cancer to ensure that the remaining kidney will be receptor. Similarly, genes have been transfected that express varying
functional. thymidine kinase activities. This agent is suitable for gene therapy
Functional imaging can be used to identify the location of eloquent and can be imaged with radiolabeled substrates such as FMAU.166,167
brain activity and evaluate motor cortex function. It also can help to Similarly, a great deal of interest has been expressed in stem cell
guide brain tumor surgery by avoiding key areas of the brain. biology and the potential for stem cells to allow for regeneration of
tissues. Tracking these stem cells in vivo and determining their bio-
GUIDANCE OF RADIATION THERAPY distribution and ultimate proliferation are exceptional opportunities
for imaging. These goals have been achieved with both nuclear medicine
Radiation therapy can be palliative, or it may be performed with and MRI methods.
curative intent. In general, the goal is to deliver maximum radiation Small-animal imaging devices of a variety of types are now being
to the tumor while minimizing radiation delivery to normal tissues. used to help in drug development. Small-animal PET, SPECT, MRI,
This delicate balance is achieved through increasingly sophisticated and CT scanners have been used to assess treatment response and
dose delivery systems. The anatomic location of a tumor most com- aspects of tumor biology. Such methods are of critical importance for
monly is defined by treatment-planning CT. The CT data are used assessing the response of cancers to treatment with newer agents and
to define tumor and normal tissues with a therapy-planning system. understanding therapeutic effects. Even combined human PET-CT
The planning potentially can be enhanced by better definition of the devices have been used for imaging and can provide useful information
gross tumor volume (or biologic tumor volume) versus anatomic tumor for drug development in cancer.
volume, which are not always the same. Some methods are of tremendous importance in preclinical studies
FDG-PET is beginning to be used to better define the biologic but will be more difficult to extend to human studies. One example
tumor volume, often with data from PET-CT. Although this procedure is optical imaging. Such methods are capable of tremendous sensitivity
is not yet fully the norm, it is clear that imaging is key to the optimal
planning of radiation therapy ports. Substantial potential exists to
target areas of tumor that are not identified on CT (expand port size)
or reduce ports to areas that are not involved with tumor, because Table 16.4  Emerging Uses of Imaging
the goal is to irradiate tumor while not irradiating normal tissues.162
A substantial number of centers now include PET imaging as part of INDIVIDUAL PATIENT MANAGEMENT DECISIONS
their radiation therapy planning in a broad range of diseases.163 Treat- • Screening
ment plans for radiation therapy are discussed in a separate chapter • Phenotyping of the tumor and host
in this book. PET agents identifying areas of tumor hypoxia may be TUMOR
useful in helping guide escalating doses to areas at high risk of treatment
failure. Another area of opportunity is elimination of radiation therapy • Viability, proliferation, extent of necrosis or apoptosis, hypoxia,
in low-risk patients, in whom the incremental benefit of radiation receptor expression prognosis, type of treatment most likely to be
may be less than the risks of radiation. This has been studied in effective based on receptors or presence of imageable pathways
Hodgkin lymphoma, and image-guided elimination of radiation HOST
treatment in low-risk patients is a tenable option. • Individualized assessment of pharmacokinetics of the drug and of
organ function and pharmacodynamics
INTERVENTIONAL PROCEDURES DRUG DEVELOPMENT
Increasingly, imaging is being combined with a therapeutic procedure • Does the tumor have a relevant target?
to provide minimally invasive therapeutics. Examples include the use • Does the drug reach the target?
of CT to guide thermal or cryoablation of liver or lung tumors and • Is the target pathway affected by treatment?
the use of MRI to guide ablation of brain and prostate tumors (through • Is the proper dose of drug being given?
heat and cold). The ability to locate tumors and follow, in near real • Does alteration of the target pathway alter the tumor biology?
time, the response to treatment is of tremendous potential.
Imaging  •  CHAPTER 16 283

and excellent resolution in vivo in small animals. A variety of approaches The functional information provided by MRI, including diffusion
can be used, with emitted light, transmitted light, and reflected light.168 characteristics and, to a substantially lesser extent, MRS, adds to the
Photoacoustic imaging approaches, in which light is transmitted and anatomic signature of the lesions and continues to grow in application.
sound is received, are also being applied.169 Imaging of additional phenotypic alterations of cancers with functional
With bioluminescence approaches, a very small number of cancer imaging methods such as PET adds information that often is clinically
cells can be identified in vivo in small animals. Such approaches, valuable for patient management in many common cancers.
although very potent in vivo in small animals and capable of being Although many molecular alterations are present in cancer, the
combined with radionuclide and other methods, are not as easily one that is by far most exploited in clinical practice and clinical
translated into humans, at least for broad applications, because of the research is the accelerated glucose metabolism present in most cancers.
limited penetration in tissue of light photons. However, in a broad This process is well imaged with the radiotracer FDG. The ability to
range of detection issues the light can reach detectors or targets, both localize the molecular alterations of cancer spatially, through qualitative
intraoperatively and endoscopically for superficial structures, so this cognitive methods performed by the imaging specialist, computer
area is likely to be increasingly translated to practice. Detection of fusion of image sets, or fused “anatomolecular” image sets using
sentinel lymph nodes and resection margins are both areas of consider- dedicated PET-CT, is key to optimal use of the imaging methods.
able promise. Thus small-animal imaging is a key element of progress The newer PET imaging agents for prostate cancer, especially those
allowing for proof of concept and refinement of tumor biologic processes targeting PSMA, are expected to have a great impact on prostate
before translation to human use.170 cancer management.
Increasingly, the ability of PET imaging to quickly assess treatment
SUMMARY response is allowing adaptation of treatments depending on the response
of a specific patient’s tumor to treatment. This response adaptive
Anatomic imaging of cancer using x-rays has been and remains extremely approach is expected to grow in the coming years. It has had great
useful, and in the more modern form of CT is still the dominant impact in lymphoma and in head and neck cancer. Systematic response
approach to imaging the patient with visceral cancer. Anatomic methods, criteria for treatment response with PET have been developed
although very potent, have clear limitations, but continue to improve, (PERCIST 1.0). Functional imaging also is being revisited in the
as evidenced by digital mammography, tomosynthesis, and other approach to radiation therapy treatment planning. Functional
techniques. Screening programs with mammography, CT of the thorax, “molecular” imaging methods such as PET, SPECT, and varying
and CT colonography are now proven methods to detect disease at methods of MRI, in addition to optical, photoacoustic, and ultrasound
earlier stages and improve outcomes. Anatomic methods can detect imaging and technical improvements in CT and interventional
disease early, and such methods reliably show whether a mass is present. techniques, are expected to enhance the care of the patient with cancer
However, they provide limited information regarding the composition in the coming years and will likely continue to change the way oncology
of the mass. In addition, they can be insensitive to the presence of is practiced.
small tumor foci, may be slow to change in response to therapy, are
not predictive of response, and may be more challenging to apply in The complete reference list is available online at
evaluating the postoperative patient. ExpertConsult.com.

KEY REFERENCES
1. Bragg DG, Rubin P, Hricak H. Imaging strategies for nephrogenic systemic fibrosis. Radiology. 2007;242: 104. Spaepen K, Stroobants S, Dupont P, et al. Prognostic
oncologic diagnosis and multidisciplinary treatment. 647–649. value of positron emission tomography (PET) with
In: Bragg D, Rubin P, Hricak H, eds. Oncologic 34. Blomqvist L, Torkzad MR. Whole-body imaging fluorine-18 fluorodeoxyglucose ([18F]FDG) after
Imaging. 2nd ed. Philadelphia: WB Saunders; 2002: with MRI or PET/CT: the future for single-modality first-line chemotherapy in non-Hodgkin’s lymphoma:
3–20. imaging in oncology? JAMA. 2003;290:3248– is [18F]FDG-PET a valid alternative to conventional
3. Bossuyt PM, Cohen JF, Gatsonis CA, Korevaar 3249. diagnostic methods? J Clin Oncol. 2001;19:414–
DA. STARD 2015: updated reporting guidelines 47. Woloshin S, Schwartz LM, Black WC, et al. Cancer 419.
for all diagnostic accuracy studies. Ann Transl Med. screening campaigns—getting past uninformative 105. Schaefer NG, Taverna C, Strobel K, et al. Hodgkin
2016;4(4). persuasion. N Engl J Med. 2012;367:1677– disease: diagnostic value of FDG PET/CT after
5. Hanley JA. Receiver operating characteristic (ROC) 1679. first-line therapy—is biopsy of FDG-avid lesions
methodology: the state of the art. Crit Rev Diagn 51. Gould MK, Maclean CC, Kuschner WG, et al. still needed? Radiology. 2007;244:257–262.
Imaging. 1989;29:307–335. Accuracy of positron emission tomography for 106. Front D, Bar-Shalom R, Mor M, et al. Hodgkin
8. Hillner BE, Siegel BA, Liu D, et al. Impact of diagnosis of pulmonary nodules and mass lesions: disease: prediction of outcome with 67Ga scintig-
positron emission tomography/computed tomog- a meta-analysis. JAMA. 2001;285:914–924. raphy after one cycle of chemotherapy. Radiology.
raphy and positron emission tomography (PET) 55. van Tinteren H, Hoekstra OS, Smit EF, et al. 1999;210:487–491.
alone on expected management of patients with Effectiveness of positron emission tomography 110. Cheson BD, Pfistner B, Juweid ME, et al. Revised
cancer: initial results from the National Oncologic in the preoperative assessment of patients with response criteria for malignant lymphoma. J Clin
PET Registry. J Clin Oncol. 2008;2155–2161. suspected non-small-cell lung cancer: the PLUS mul- Oncol. 2007;25:579–586.
doi:10.1200/JCO.2007.14.5631. ticentre randomised trial. Lancet. 2002;359:1388– 119. Stoffels I, Boy C, Pöppel T, Kuhn J, et al. Associa-
26. DePriest PD, DeSimone CP. Ultrasound screening 1393. tion between sentinel lymph node excision with or
for the early detection of ovarian cancer. J Clin 57. Lardinois D, Weder W, Hany TF, et al. Staging of without preoperative SPECT/CT and metastatic
Oncol. 2003;21(suppl 10):194–199. non-small-cell lung cancer with integrated positron- node detection and disease-free survival in mela-
30. Lee KC, Sud S, Meyer CR, et al. An imaging emission tomography and computed tomography. noma. JAMA. 2012;308(10).
biomarker of early treatment response in prostate N Engl J Med. 2003;348:2500–2507. 172. Tatsumi M, Miller JH, Wahl RL. 18F-FDG PET/
cancer that has metastasized to the bone. Cancer 89. Johnson CD, Mei-Hsiu Chen MH, Alicia Y, CT in evaluating non-CNS pediatric malignancies.
Res. 2007;67:3524–3528. Toledano AY, et al. Accuracy of CT colonography J Nucl Med. 2007;48(12):1923–1931.
32. Kuo PH, Kanal E, Abu-Alfa AK, Cowper SE. for detection of large adenomas and cancers. N Engl
Gadolinium-based MR contrast agents and J Med. 2008;359:1207–1217.
Imaging  •  CHAPTER 16 283.e1
283.e1

REFERENCES
1. Bragg DG, Rubin P, Hricak H. Imaging strategies for mammography for breast-cancer screening. N Engl pediatric CT. AJR Am J Roentgenol. 2001;176(2):289–
oncologic diagnosis and multidisciplinary treatment. J Med. 2005;353:1773–1783. 296.
In: Bragg D, Rubin P, Hricak H, eds. Oncologic 21. Shah AJ, Wang J, Yamada T, Fajardo LL. Digital 43. Brenner DJ, Hall EJ. Computed tomography—an
Imaging. 2nd ed. Philadelphia: WB Saunders; mammography: a review of technical develop- increasing source of radiation exposure. N Engl J
2002:3–20. ment and clinical applications. Clin Breast Cancer. Med. 2007;357:2277–2284.
2. Friedman KP, Wahl RL. Clinical use of positron 2003;4:63–70. 44. Pandharipande PV, Eisenberg JD, Lee RJ, Gilmore
emission tomography in the management of cutane- 22. Svahn TM. Breast tomosynthesis and digital mam- ME, Turan EA, Singh S, et al. Patients with testicular
ous melanoma. Semin Nucl Med. 2004;34:242–253. mography: a comparison of diagnostic accuracy. BJR. cancer undergoing CT surveillance demonstrate a
3. Bossuyt PM, Cohen JF, Gatsonis CA, Korevaar 2012;85:1019, e1074–e1082. pitfall of radiation-induced cancer risk estimates: the
DA. STARD 2015: updated reporting guidelines 23. Gilbert FJ, Tucker L, Young KC. Digital breast timing paradox. Radiology. 2013;266(3):896–904.
for all diagnostic accuracy studies. Ann Transl Med. tomosynthesis (DBT): a review of the evidence for doi:10.1148/radiol.12121015. [Epub 2012 Dec 18].
2016;4(4). use as a screening tool. Clin Radiol. 2016;71(2):141– 45. Wahl RL. Anatomolecular imaging of cancer. Mol
4. Gates TJ. Screening for cancer: evaluating the 150. Imag Biol. 2003;5:49–56.
evidence. Am Fam Physician. 2001;63:513–522. 24. Miles KA. Functional computed tomography in 46. Hanahan D, Weinberg RA. Hallmarks of cancer:
5. Hanley JA. Receiver operating characteristic (ROC) oncology. Eur J Cancer. 2002;38:2079–2084. the next generation. Cell. 2011;144(5):646–674.
methodology: the state of the art. Crit Rev Diagn 25. Singh K. Interventional radiology in the gynaeco- doi:10.1016/j.cell.2011.02.013.
Imaging. 1989;29:307–335. logical oncology patient. Best Pract Res Clin Obstet 47. Woloshin S, Schwartz LM, Black WC, et al. Cancer
6. Wahl RL, Siegel BA, Coleman RE, Gatsonis CG. Gynaecol. 2001;15:279–290. screening campaigns—getting past uninformative
Prospective multicenter study of axillary nodal 26. DePriest PD, DeSimone CP. Ultrasound screening persuasion. N Engl J Med. 2012;367:1677–1679.
staging by positron emission tomography in breast for the early detection of ovarian cancer. J Clin 48. Guidelines for management of small pulmonary
cancer. J Clin Oncol. 2004;22:277–285. Oncol. 2003;21(suppl 10):194–199. nodules detected on CT scans: a statement from
7. Verboom P, Van Tinteren H, Hoekstra OS, et al. 27. Lehman CD, Isaacs C, Schnall MD, et al. Cancer the Fleischner Society. Radiology. 2005;237:395–400.
Cost-effectiveness of FDG-PET in staging non-small yield of mammography, MR, and US in high-risk 49. Erasmus JJ, Connolly JE, McAdams HP, Roggli
cell lung cancer: the PLUS study. Eur J Nucl Med women: prospective multi-institution breast cancer VL. Solitary pulmonary nodules: Part I. Morpho-
Mol Imaging. 2003;30:1444–1449. screening study. Radiology. 2007;244:381–388. logic evaluation for differentiation of benign and
8. Hillner BE, Siegel BA, Liu D, et al. Impact of 28. Laking GR, Price PM, Sculpher MJ. Assessment of malignant lesions. Radiographics. 2000;20:43–58.
positron emission tomography/computed tomog- the technology for functional imaging in cancer. Eur 50. Erasmus JJ, McAdams HP, Connolly JE. Solitary
raphy and positron emission tomography (PET) J Cancer. 2002;38:2194–2199. pulmonary nodules: Part II. Evaluation of the
alone on expected management of patients with 29. Kwock L, Smith JK, Castillo M, et al. Clinical indeterminate nodule. Radiographics. 2000;20:59–66.
cancer: initial results from the National Oncologic applications of proton MR spectroscopy in oncology. 51. Gould MK, Maclean CC, Kuschner WG, et al.
PET Registry. J Clin Oncol. 2008;2155–2161. Technol Cancer Res Treat. 2002;1:17–28. Accuracy of positron emission tomography for
doi:10.1200/JCO.2007.14.5631. 30. Lee KC, Sud S, Meyer CR, et al. An imaging diagnosis of pulmonary nodules and mass lesions:
9. de Koning HJ. Mammographic screening: evidence biomarker of early treatment response in prostate a meta-analysis. JAMA. 2001;285:914–924.
from randomised controlled trials. Ann Oncol. cancer that has metastasized to the bone. Cancer 52. Naidich DP, Bankier AA, MacMahon H. Recommen-
2003;14:1185–1189. Res. 2007;67:3524–3528. dations for the management of subsolid pulmonary
10. Henschke CI, Yankelevitz DF, McCauley DI, 31. Cowper SE, Su LD, Bhawan J, et al. Nephrogenic nodules detected at CT: a statement from the
et al. Guidelines for the use of spiral computed fibrosing dermopathy. Am J Dermatopathol. 2001;23: Fleischner Society. Radiology. 2013;266(1):304–317.
tomography in screening for lung cancer. Eur Respir 383–393. doi:10.1148/radiol.12120628. Published online
J. 2003;39:45s–51s. 32. Kuo PH, Kanal E, Abu-Alfa AK, Cowper SE. Gad- October 15, 2012.
11. Henschke CI, McCauley DI, Yankelevitz DF, et al. olinium-based MR contrast agents and nephrogenic 53. Goudarzi B, Jacene HA, Wahl RL. Diagnosis and
Early Lung Cancer Action Project: overall design and systemic fibrosis. Radiology. 2007;242:647–649. differentiation of bronchioloalveolar carcinoma from
findings from baseline screening. Lancet. 1999;354: 33. High WA, Ayers RA, Chandler J, et al. Gadolinium adenocarcinoma with bronchioloalveolar components
99–105. is detectable within the tissue of patients with with metabolic and anatomic characteristics using
12. Henschke CI, Yankelevitz DF, Libby DM, et al. Sur- nephrogenic systemic fibrosis. J Am Acad Dermatol. PET/CT. J Nucl Med. 2008;49:1585–1592.
vival of patients with stage I lung cancer detected on 2007;56:21–26. 54. Dwamena BA, Sonnad SS, Angobaldo JO, Wahl RL.
CT screening. N Engl J Med. 2006;355:1763–1771. 34. Blomqvist L, Torkzad MR. Whole-body imaging Metastases from non-small cell lung cancer: medias-
13. Aberle DR, Adams AM, Berg CD, et al. National Lung with MRI or PET/CT: the future for single-modality tinal staging in the 1990s. Meta-analytic comparison
Screening Trial Research Team: Reduced lung-cancer imaging in oncology? JAMA. 2003;290:3248– of PET and CT. Radiology. 1999;213:530–536.
mortality with low-dose computed tomographic 3249. 55. van Tinteren H, Hoekstra OS, Smit EF, et al.
screening. N Engl J Med. 2011;365(5):395– 35. Cho A, Lau JY, Geraghty BJ, Cunningham CH, Effectiveness of positron emission tomography in the
409. doi:10.1056/NEJMoa1102873. [Epub 2011 Keshari KR. Hyperpolarized MRI of cancer preoperative assessment of patients with suspected
Jun 29]. metabolism. J Nucl Med. 2017;jnumed-116. non-small-cell lung cancer: the PLUS multicentre
14. Carter HB, Partin AW, Walsh PC, et al. Gleason 36. Jerusalem G, Hustinx R, Beguin Y, Fillet G. randomised trial. Lancet. 2002;359:1388–1393.
score 6 adenocarcinoma: should it be labeled as PET scan imaging in oncology. Eur J Cancer. 56. Beadsmoore CJ, Screaton NJ. Classification,
cancer? J Clin Oncol. 2012;30(35):4294–4296. 2003;39:1525–1534. staging and prognosis of lung cancer. Eur J Radiol.
15. Jepsen P, Johnsen SP, Gillman MW, et  al. 37. Kim JH, Lee JS, Song IC, Lee DS. Comparison of 2003;45:8–17.
Interpretation of observational studies. Heart. segmentation-based attenuation correction methods 57. Lardinois D, Weder W, Hany TF, et al. Staging of
2004;90:956–960. for PET/MRI: evaluation of bone and liver standard- non–small-cell lung cancer with integrated positron-
16. Ubel PA, Hirth RA, Chernew ME, Fendrick ized uptake value with oncologic PET/CT data. emission tomography and computed tomography.
AM. What is the price of life and why doesn’t it J Nucl Med. 2012;53(12):1878–1882. N Engl J Med. 2003;348:2500–2507.
increase at the rate of inflation? Arch Intern Med. 38. Fraum TJ, Fowler KJ, McConathy J. PET/MRI: 58. Bradley JD, Dehdashti F, Mintun MA, et al. Positron
2003;163:1637–1641. Emergency applications of oncology. Acad Radiol. emission tomography in limited-stage small-cell lung
17. Wahl RL, Quint LE, Cieslak RD, et al. “Anatometa- 2016;23(2):220–236. cancer: a prospective study. J Clin Oncol. 2004;22:
bolic” tumor imaging: fusion of FDG PET with 39. Kaminski MS, Zelenetz AD, Press OW, et al. 3248–3254.
CT or MRI to localize foci of increased activity. Pivotal study of iodine I 131 tositumomab for 59. Mayo-Smith WW, Boland GW, Noto RB, Lee MJ. State-
J Nucl Med. 1993;34:1190–1197. chemotherapy-refractory low-grade or transformed of-the-art adrenal imaging. Radiographics. 2001;21:
18. Cohade C, Wahl RL. Applications of positron low-grade B-cell non-Hodgkin’s lymphomas. J Clin 995–1012.
emission tomography/computed tomography image Oncol. 2001;19:3918–3928. 60. Smith RA, Saslow D, Sawyer KA, et al. American
fusion in clinical positron emission tomography: 40. Bremer C, Ntziachristos V, Weissleder R. Cancer Society guidelines for breast cancer screening:
clinical use, interpretation methods, diagnostic Optical-based molecular imaging: contrast agents update 2003. CA Cancer J Clin. 2003;53:141–169.
improvements. Semin Nucl Med. 2003;33:228–237. and potential medical applications. Eur Radiol. 61. Poplack SP, Tosteson TD, Kogel CA, Nagy HM.
19. Gourtsoyiannis N, Grammatikakis J, Prassopoulos P. 2003;13:231–243. Digital breast tomosynthesis: initial experience in
Role of conventional radiology in the diagnosis and 41. Fazel R. Exposure to Low-dose ionizing radiation 98 women with abnormal digital screening mam-
staging of gastrointestinal tract neoplasms. Semin from medical imaging procedures. N Engl J Med. mography. AJR Am J Roentgenol. 2007;189:616–623.
Surg Oncol. 2001;20:91–108. 2009;361:849–857. 62. Chen SC, Carton AK, Albert M, et al. Initial clinical
20. Pisano ED, Gatsonis C, Hendrick E, et al. 42. Brenner DJ, Elliston CD, Hall EJ, et al. Estimated experience with contrast-enhanced digital breast
Diagnostic performance of digital versus film risks of radiation-induced fatal cancer from tomosynthesis. Acad Radiol. 2007;14:229–238.
283.e2 Part I: Science and Clinical Oncology

63. Bleyer A, Welch HG. Effect of three decades of anti-1-amino-3-18F-fluorocyclobutane-1-carboxylic emission tomography in the diagnosis of recurrent
screening mammography on breast-cancer incidence. acid PET/CT and 111In-capromab pendetide ovarian cancer. Gynecol Oncol. 2001;83:310–315.
N Engl J Med. 2012;367:1998–2005. SPECT/CT. Radiology. 2011;259(3):852–861. 98. Avril N, Sassen S, Schmalfeldt B, et al. Prediction
64. Rhodes DJ, Hruska CB, Phillips SW, et al. Dedicated doi:10.1148/radiol.11102023. [Epub 2011 Apr 14]. of response to neoadjuvant chemotherapy by
dual-head gamma imaging for breast cancer screening 82. Maurer T, Gschwend JE, Rauscher I, Souvatzoglou sequential F-18-fluorodeoxyglucose positron emission
in women with mammographically dense breasts. M, Haller B, Weirich G, et al. Diagnostic efficacy tomography in patients with advanced-stage ovarian
Radiology. 2011;258(1):106–118. doi:10.1148/ of 68 gallium-PSMA positron emission tomography cancer. J Clin Oncol. 2005;23:7445–7453.
radiol.10100625. [Epub 2010 Nov 2]. compared to conventional imaging for lymph node 99. Bristow RE, del Carmen MG, Pannu HK, et al.
65. Bluemke DA, Gatsonis CA, Chen MH, et al. staging of 130 consecutive patients with intermediate Clinically occult recurrent ovarian cancer: patient
Magnetic resonance imaging of the breast prior to to high risk prostate cancer. J Urol. 2016;195(5): selection for secondary cytoreductive surgery using
biopsy. JAMA. 2004;292:2735–2742. 1436–1443. combined PET/CT. Gynecol Oncol. 2003;90:
66. Lehman CD, Gatsonis C, Kuhl CK, et al. MRI 83. Schwenck J, Rempp H, Reischl G, Kruck S, Stenzl 519–528.
evaluation of the contralateral breast in women with A, Nikolaou K, et al. Comparison of 68Ga-labelled 100. O’Doherty MJ, Macdonald EA, Barrington SF,
recently diagnosed breast cancer. N Engl J Med. PSMA-11 and 11C-choline in the detection of et al. Positron emission tomography in the manage-
2007;356:1295–1303. prostate cancer metastases by PET/CT. Eur J Nucl ment of lymphomas. Clin Oncol (R Coll Radiol).
67. Kuhl CK. Current status of breast MR imaging. Med Mol Imaging. 2017;44(1):92–101. 2002;14:415–426.
Part 2. Clinical applications. Radiology. 2007;244: 84. Bluemel C, Krebs M, Polat B, Linke F, Eiber M, 101. Nakamoto Y, Tatsumi M, Hammoud D, et al. Normal
672–691. Samnick S, et al. 68Ga-PSMA-PET/CT in patients FDG distribution patterns in the head and neck:
68. Tatsumi M, Cohade C, Mourtzikos KA, et al. Initial with biochemical prostate cancer recurrence and PET/CT evaluation. Radiology. 2005;234:879–885.
experience with FDG-PET/CT in the evaluation negative 18F-choline-PET/CT. Clin Nucl Med. 102. Nakamoto Y, Cohade C, Tatsumi M, et al. CT
of breast cancer. Eur J Nucl Med Mol Imaging. 2016;41(7):515–521. appearance of bone metastases detected with FDG
2006;33(3):254–262. 85. Cho SY, Gage KL, Mease RC, et al. Biodistribu- PET as part of the same PET/CT examination.
69. Berg WA, Madsen KS, Schilling K, et al. Breast tion, tumor detection, and radiation dosimetry of Radiology. 2005;237:627–634.
cancer: comparative effectiveness of positron emission 18F-DCFBC, a low-molecular-weight inhibitor of 103. El-Galaly TC, d’Amore F, Mylam KJ, de Nully Brown
mammography and MR imaging in presurgi- prostate-specific membrane antigen, in patients with P, Bøgsted M, Bukh A, et al. Routine bone marrow
cal planning for the ipsilateral breast. Radiology. metastatic prostate cancer. J Nucl Med. 2012;53(12): biopsy has little or no therapeutic consequence for
2011;258(1):59–72. doi:10.1148/radiol.10100454. 1883–1891. positron emission tomography/computed tomogra-
[Epub 2010 Nov 12]; PMID:21076089. 86. Rowe SP, Gage KL, Faraj SF, Macura KJ, Cornish phy–staged treatment-naive patients with Hodgkin
70. Lyman GH, Giuliano AE, Somerfield MR, et al. TC, Gonzalez-Roibon N, et al. 18F-DCFBC lymphoma. J Clin Oncol. 2012;30(36):4508–4514.
American Society of Clinical Oncology guideline PET/CT for PSMA-based detection and charac- 104. Spaepen K, Stroobants S, Dupont P, et al. Prognostic
recommendations for sentinel lymph node biopsy terization of primary prostate cancer. J Nucl Med. value of positron emission tomography (PET) with
in early-stage breast cancer. J Clin Oncol. 2005;23: 2015;56(7):1003–1010. fluorine-18 fluorodeoxyglucose ([18F]FDG) after
7703–7720. 87. Szabo Z, Mena E, Rowe SP, Plyku D, Nidal R, first-line chemotherapy in non-Hodgkin’s lymphoma:
71. Veronesi U, Paganelli G, Viale G, et al. A randomized Eisenberger MA, et al. Initial evaluation of [18F] is [18F]FDG-PET a valid alternative to conventional
comparison of sentinel-node biopsy with routine DCFPyL for prostate-specific membrane antigen diagnostic methods? J Clin Oncol. 2001;19:414–419.
axillary dissection in breast cancer. N Engl J Med. (PSMA)-targeted PET imaging of prostate cancer. 105. Schaefer NG, Taverna C, Strobel K, et al. Hodgkin
2003;349:546–553. Mol Imaging Biol. 2015;17(4):565–574. disease: diagnostic value of FDG PET/CT after
72. Galimberti V, Cole BF, Viale G, Veronesi P, Vicini 88. Kim DH, Pickhardt PJ, Taylor AJ, et al. CT first-line therapy—is biopsy of FDG-avid lesions
E, Intra M, et al. Axillary dissection versus no colonography versus colonoscopy for the detection still needed? Radiology. 2007;244:257–262.
axillary dissection in patients with breast cancer of advanced neoplasia. N Engl J Med. 2007;357: 106. Front D, Bar-Shalom R, Mor M, et al. Hodgkin
and sentinel-node micrometastases (IBCSG 23-01): 1403–1412. disease: prediction of outcome with 67Ga scintig-
10-year follow-up of a randomised, controlled phase 89. Johnson CD, Mei-Hsiu Chen M-H, Alicia Y, raphy after one cycle of chemotherapy. Radiology.
3 trial. Lancet Oncol. 2018. Toledano AY, et al. Accuracy of CT colonography 1999;210:487–491.
73. Wu D, Gambhir SS. Positron emission tomography for detection of large adenomas and cancers. N Engl 107. Kasamon YL, Jones RJ, Wahl RL. Integrating PET
in diagnosis and management of invasive breast J Med. 2008;359:1207–1217. and PET/CT into the risk-adapted therapy of
cancer: current status and future perspectives. Clin 90. Chin BB, Wahl RL. 18F-Fluoro-2-deoxyglucose lymphoma. J Nucl Med. 2007;48(suppl 1):19S–27S.
Breast Cancer. 2003;4(suppl 1):55–63. positron emission tomography in the evaluation of 108. Hutchings M, Kostakoglu L, Zaucha JM, Mal-
74. Yu KK, Hricak H. Imaging prostate cancer. Radiol gastrointestinal malignancies. Gut. 2003;52(suppl kowski B, Biggi A, Danielewicz I, et al. In vivo
Clin North Am. 2000;38:59–85. 4):23–29. treatment sensitivity testing with positron emission
75. Heijmink SW, Barentsz JO. Contrast-enhanced versus 91. Cohade C, Osman M, Leal J, Wahl RL. Direct tomography/computed tomography after one cycle
systematic transrectal ultrasound-guided prostate comparison of 18-FDG PET and PET/CT in of chemotherapy for Hodgkin lymphoma. J Clin
cancer detection: an overview of techniques and a patients with colorectal carcinoma. J Nucl Med. Oncol. 2014;32(25):2705–2711.
systematic review. Eur J Radiol. 2007;63:310–316. 2003;44:1797–1803. 109. Cohade C, Osman M, Pannu HK, Wahl RL.
76. Weinreb JC, Barentsz JO, Choyke PL, Cornud F, 92. Kinkel K, Lu Y, Both M, et al. Detection of hepatic Uptake in supraclavicular area fat (“USA-Fat”):
Haider MA, Macura KJ, et al. PI-RADS Prostate metastases from cancers of the gastrointestinal tract description on 18F-FDG PET/CT. J Nucl Med.
Imaging–Reporting and Data System: 2015, version by using noninvasive imaging methods (US, CT, 2003;44:170–176.
2. Eur Urol. 2016;69(1):16–40. MR imaging, PET): a meta-analysis. Radiology. 110. Cheson BD, Pfistner B, Juweid ME, et al. Revised
77. Pinto PA, Chung PH, Rastinehad AR, et al. 2002;224:748–756. response criteria for malignant lymphoma. J Clin
Magnetic resonance imaging/ultrasound fusion 93. Niekel MC, Bipat S, Stoker J. Diagnostic imaging Oncol. 2007;25:579–586.
guided prostate biopsy improves cancer detection of colorectal liver metastases with CT, MR imaging, 111. Juweid ME, Cheson BD. Positron-emission
following transrectal ultrasound biopsy and correlates FDG PET, and/or FDG PET/CT: a meta-analysis tomography and assessment of cancer therapy.
with multiparametric magnetic resonance imaging. of prospective studies including patients who have N Engl J Med. 2006;354:496–507.
J Urol. 2011;186(4):1281–1285. doi:10.1016/j. not previously undergone treatment. Radiology. 112. Juweid ME, Wiseman GA, Vose JM, et al. Response
juro.2011.05.078. [Epub 2011 Aug 17]. 2010;257:674–684. assessment of aggressive non-Hodgkin’s lymphoma
78. Harisinghani MG, Barentsz J, Hahn PF, et al. 94. Singh AK, Grigsby PW, Dehdashti F, et al. FDG- by integrated International Workshop Criteria and
Noninvasive detection of clinically occult lymph- PET lymph node staging and survival of patients fluorine-18-fluorodeoxyglucose positron emission
node metastases in prostate cancer. N Engl J Med. with FIGO stage IIIb cervical carcinoma. Int J Radiat tomography. J Clin Oncol. 2005;23:4652–4661.
2003;348:2491–2499. Oncol Biol Phys. 2003;56:489–493. 113. Cheson BD, Fisher RI, Barrington SF, Cavalli F,
79. Scheenen TW, Heijmink SW, Roell SA, et al. Three- 95. Grigsby PW, Siegel BA, Dehdashti F, et al. Post- Schwartz LH, Zucca E, et al. Recommendations for
dimensional proton MR spectroscopy of human therapy [18F] fluorodeoxyglucose positron emission initial evaluation, staging, and response assessment of
prostate at 3 T without endorectal coil: feasibility. tomography in carcinoma of the cervix: response Hodgkin and non-Hodgkin lymphoma: the Lugano
Radiology. 2007;245:507–516. and outcome. J Clin Oncol. 2004;22:2167–2171. classification. J Clin Oncol. 2014;32(27):3059–
80. Barentsz JO, Futterer JJ, Takahashi S. Use of 96. Grigsby PW, Siegel BA, Dehdashti F. Lymph 3067.
ultrasmall superparamagnetic iron oxide in lymph node staging by positron emission tomography in 114. Cheson BD, Ansell S, Schwartz L, Gordon LI,
node MR imaging in prostate cancer patients. Eur patients with carcinoma of the cervix. J Clin Oncol. Advani R, Jacene HA, et al. Refinement of the
J Radiol. 2007;63:369–372. 2001;19:3745–3749. Lugano Classification lymphoma response criteria
81. Schuster DM, Savir-Baruch B, Nieh PT, et al. 97. Zimny M, Siggelkow W, Schroder W, et al. in the era of immunomodulatory therapy. Blood.
Detection of recurrent prostate carcinoma with 2-[Fluorine-18]-fluoro-2-deoxy-d-glucose positron 2016;128(21):2489–2496.
Imaging  •  CHAPTER 16 283.e3
283.e3

115. Gallamini A, Barrington SF, Biggi A, Chauvie S, and neck squamous cell carcinoma. J Clin Oncol. in adenocarcinomas of the esophagogastric junction.
Kostakoglu L, Gregianin M, et al. The predictive 2002;20:4199–4208. J Clin Oncol. 2006;24:4692–4698.
role of interim positron emission tomography for 132. Branstetter BF, Blodgett TM, Zimmer LA, et al. Head 151. Hoffer FA. Primary skeletal neoplasms: osteosarcoma
Hodgkin lymphoma treatment outcome is confirmed and neck malignancy: is PET/CT more accurate and Ewing sarcoma. Top Magn Reson Imaging.
using the interpretation criteria of the Deauville five- than PET or CT alone? Radiology. 2005;235:580– 2002;13:231–239.
point scale. Haematologica. 2014;99(6):1107–1113. 586. 152. Brenner W, Bohuslavizki KH, Eary JF. PET imaging
116. Kasamon YL, Wahl RL, Swinnen LJ. FDG PET and 133. Kalra MK, Maher MM, Boland GW, et al. Correla- of osteosarcoma. J Nucl Med. 2003;44:930–
high-dose therapy for aggressive lymphomas: toward tion of positron emission tomography and CT in 942.
a risk-adapted strategy. Curr Opin Oncol. 2004;16: evaluating pancreatic tumors: technical and clinical 153. Hawkins DS, Schuetze SM, Butrynski JE, et al. [18F]
100–105. implications. Am J Roentgenol. 2003;181:387–393. Fluorodeoxyglucose positron emission tomography
117. Dann EJ, Bar-Shalom R, Tamir A, et al. Risk-adapted 134. Hanbidge AE. Cancer of the pancreas: the best image predicts outcome for Ewing sarcoma family of
BEACOPP regimen can reduce the cumulative dose for early detection. CT, MRI, PET or US? Can J tumors. J Clin Oncol. 2005;23:8828–8834.
of chemotherapy for standard and high-risk Hodgkin Gastroenterol. 2002;16:101–105. 154. Luber BS, Leal JP, Wang H, Bolejack V, Schuetze
lymphoma with no impairment of outcome. Blood. 135. Pakzad F, Groves AM, Ell PJ. The role of positron SM, Schwartz LH, et al. Response to early treatment
2007;109:905–909. emission tomography in the management of pan- evaluated with 18F-FDG PET and PERCIST 1.0
118. Alazraki N, Glass EC, Castronovo F, Olmos RA, creatic cancer. Semin Nucl Med. 2006;36:248–256. predicts survival in patients with Ewing sarcoma
Podoloff D. Procedure guideline for lymphos- 136. Antunes P, Ginj M, Zhang H, et al. Are radiogallium- family of tumors treated with a monoclonal antibody
cintigraphy and the use of intraoperative gamma labelled DOTA-conjugated somatostatin analogues to the insulinlike growth factor 1 receptor. J Nucl
probe for sentinel lymph node localization in superior to those labelled with other radiometals? Med. 2016;57(5):735–740.
melanoma of intermediate thickness 1.0. J Nucl Eur J Nucl Med Mol Imaging. 2007;34:982–993. 155. Choi H, Charnsangavej C, Faria SC, et al. Cor-
Med. 2002;43:1414–1418. 137. Ho CL, Chen S, Yeung DW, Cheng TK. Dual-tracer relation of computed tomography and positron
119. Stoffels I, Boy C, Pöppel T, Kuhn J, et al. Associa- PET/CT imaging in evaluation of metastatic hepa- emission tomography in patients with metastatic
tion between sentinel lymph node excision with or tocellular carcinoma. J Nucl Med. 2007;48:902–909. gastrointestinal stromal tumor treated at a single
without preoperative SPECT/CT and metastatic 138. Sheth S, Scatarige JC, Horton KM, et al. Current institution with imatinib mesylate: proposal of
node detection and disease-free survival in mela- concepts in the diagnosis and management of renal new computed tomography response criteria.
noma. JAMA. 2012;308(10). cell carcinoma: role of multidetector C and three- J Clin Oncol. 2007;25:1753–1759.
120. Cobben DC, Koopal S, Tiebosch AT, et al. New dimensional CT. Radiographics. 2001;21:237–254. 156. Padhani AR, Liu G, Koh DM, et al. Diffusion-
diagnostic techniques in staging in the surgical 139. Aide N, Cappele O, Bottet P, et al. Efficiency of weighted magnetic resonance imaging as a cancer
treatment of cutaneous malignant melanoma. Eur [(18)]FDG PET in characterising renal cancer and biomarker: consensus and recommendations.
J Surg Oncol. 2002;28:692–700. detecting distant metastases: a comparison with CT. Neoplasia. 2009;11(2):102–125, 71.
121. Swetter SM, Carroll LA, Johnson DL, Segall GM. Eur J Nucl Med Mol Imaging. 2003;30:1236–1245. 157. Wahl RL, Jacene H, Kasamon Y, et al. From
Positron emission tomography is superior to com- 140. Pocaro AB, Cavalleri S, Ballista C, et al. Modern RECIST to PERCIST: evolving considerations for
puted tomography for metastatic detection in mela- imaging methods and preoperative management of PET response criteria in solid tumors. J Nucl Med.
noma patients. Ann Surg Oncol. 2002;9:646–653. pheochromocytoma: review of the literature and 2009;50(suppl 1):122S–150S.
122. Cho SY, Lipson EJ, Im HJ, Rowe SP, Mena E, case report. Arch Esp Urol. 2000;53:749–753. 158. Eisenhauer EA, Therasse P, Bogaerts J, et al. New
Blackford A, et al. Prediction of response to immune 141. Divgi CR, Uzzo RG, Gatsonis C, et al. Positron response evaluation criteria in solid tumours: revised
checkpoint inhibitor therapy using early time-point emission tomography/computed tomography RECIST guideline (version 1.1). Eur J Cancer. 2009;
FDG-PET/CT imaging in patients with advanced identification of clear cell renal cell carcinoma: results 45(2):228–247.
melanoma. J Nucl Med. 2017;pp.jnumed-116. from the REDECT Trial. J Clin Oncol. 2013;31(2): 159. Hodi FS, Hwu WJ, Kefford R, Weber JS, Daud
123. Strobel K, Dummer R, Husarik DB, et al. High-risk 187–194. A, Hamid O, et al. Evaluation of immune-related
melanoma: accuracy of FDG PET/CT with added 142. Chin BB, Patel P, Cohade C, et al. Recombinant response criteria and RECIST v1. 1 in patients with
CT morphologic information for detection of human thyrotropin stimulation of fluoro-d-glucose advanced melanoma treated with pembrolizumab.
metastases. Radiology. 2007;244:566–574. positron emission tomography uptake in well- J Clin Oncol. 2016;34(13):1510–1517.
124. Barentsz JO, Witjes JA, Ruijs JH. What is new differentiated thyroid carcinoma. J Clin Endocrinol 160. Shankar LK, Hoffman JM, Bacharach S, et al. Con-
in bladder cancer imaging? Urol Clin North Am. Metab. 2004;89:91–95. sensus recommendations for the use of 18F-FDG
1997;24:583–602. 143. Treglia G, Castaldi P, Rindi G, et al. Diagnostic PET as an indicator of therapeutic response in
125. Lawler LP. MR imaging of the bladder. Radiol Clin performance of gallium-68 somatostatin receptor patients in National Cancer Institute Trials. J Nucl
North Am. 2003;41:161–177. PET and PET/CT in patients with thoracic and Med. 2006;47(6):1059–1066.
126. Harisinghani MG, Saksena MA, Hahn PF, et al. gastroenteropancreatic neuroendocrine tumours: 161. Wolchok JD, Hoos A, O’Day S, Weber JS, Hamid
Ferumoxtran-10-enhanced MR lymphangiography: a meta-analysis. Endocrine. 2012;42(1):80–87. O, Lebbé C, et al. Guidelines for the evaluation
does contrast-enhanced imaging alone suffice for doi:10.1007/s12020-012-9631-1. [Epub 2012 of immune therapy activity in solid tumors:
accurate lymph node characterization? Am J Feb 20]. immune-related response criteria. Clin Cancer Res.
Roentgenol. 2006;186:144–148. 144. Kauhanen S, Seppanen M, Minn H, et al. 2009;15(23):7412–7420.
127. Powles T, Murray I, Brock C, et al. Molecular Fluorine-18-l-dihydroxyphenylalanine (18F-DOPA) 162. Ling CC, Humm J, Larson S, et al. Towards mul-
positron emission tomography and PET/CT imaging positron emission tomography as a tool to localize an tidimensional radiotherapy (MD-CRT): biological
in urological malignancies. Eur Urol. 2007;51: insulinoma or beta-cell hyperplasia in adult patients. imaging and biological conformality. Int J Radiat
1511–1520. J Clin Endocrinol Metab. 2007;92:1237–1244. Oncol Biol Phys. 2000;47:551–560.
128. Zinreich J. Imaging in laryngeal cancer: computed 145. Chen W, Silverman DH, Delaloye S, et al. 18F- 163. Brianzoni E, Rossi G, Ancidei S, et al. Radiotherapy
tomography, magnetic resonance imaging, positron FDOPA PET imaging of brain tumors: comparison planning: PET/CT scanner performances in the
emission tomography. Otolaryngol Clin North Am. study with 18F-FDG PET and evaluation of definition of gross tumour volume and clinical target
2002;35:971–991. diagnostic accuracy. J Nucl Med. 2006;47:904– volume. Eur J Nucl Med Mol Imaging. 2005;32:
129. Mehanna H, McConkey CC, Rahman JK, et al. 911. 1392–1399.
PET-NECK: a multicenter randomized phase III 146. van Waarde A, Jager PL, Ishiwata K, et al. Com- 164. Lin M, Shon IH, Wilson R, et al. Treatment response
non-inferiority trial comparing a positron emission parison of sigma-ligands and metabolic PET tracers in liver metastases following 90Y SIR-spheres:
tomography-computerised tomography-guided for differentiating tumor from inflammation. J Nucl an evaluation with PET. Hepatogastroenterology.
watch-and-wait policy with planned neck dissec- Med. 2006;47:150–154. 2007;54:910–912.
tion in the management of locally advanced (N2/ 147. Jacobs AH, Winkler A, Dittmar C, et al. Molecular 165. van Baardwijk A, Dooms C, van Suylen RJ,
N3) nodal metastases in patients with squamous and functional imaging technology for the develop- et al. The maximum uptake of 18-deoxyglucose
cell head and neck cancer. Health Technol Assess. ment of efficient treatment strategies for gliomas. on positron emission tomography scan correlates
2017;21:1–122. Technol Cancer Res Treat. 2002;1:187–204. with survival, hypoxia inducible factor-1alpha and
130. Mehanna H, Wong WL, McConkey CC, Rahman 148. Frush DP. Pediatric CT: practical approach to GLUT-1 in non–small cell lung cancer. Eur J Cancer.
JK, Robinson M, Hartley AG, et al. PET-CT diminish the radiation dose. Pediatr Radiol. 2002;32: 2007;43:1392–1398.
surveillance versus neck dissection in advanced 714–717. 166. Tjuvajev JG, Avril N, Oku T, et al. Imaging herpes
head and neck cancer. NEJM. 2016;374(15):1444– 149. Reed CE, Eloubeidi MA. New techniques for virus thymidine kinase gene transfer and expres-
1454. staging esophageal cancer. Surg Clin North Am. sion by positron emission tomography. Cancer Res.
131. Wong RJ, Lin DT, Schoder H, et al. Diagnostic 2002;82:697–710. 1998;58:4333–4341.
and prognostic value of [(18)]fluorodeoxyglucose 150. Ott K, Weber WA, Lordick F, et al. Metabolic 167. Yaghoubi SS, Berger F, Gambhir SS. Study-
positron emission tomography for recurrent head imaging predicts response, survival, and recurrence ing the biodistribution of positron emission
283.e4 Part I: Science and Clinical Oncology

tomography reporter probes in mice. Nat Protoc. 169. Akers WJ, Edwards WB, Kim C, et al. Multimodal 170. Emerson DK, Limmer KK, Hall DJ, et al. A
2007;2:1752–1755. sentinel lymph node mapping with single-photon receptor-targeted fluorescent radiopharmaceutical
168. Keereweer S, Hutteman M, Kerrebijn JD, et al. emission computed tomography (SPECT)/computed for multireporter sentinel lymph node imaging.
Translational optical imaging in diagnosis and treat- tomography (CT) and photoacoustic tomography. Radiology. 2012;265(1):186–193.
ment of cancer. Curr Pharm Biotechnol. 2012;13(4): Transl Res. 2012;159(3):175–181.
498–503.
Imaging  •  CHAPTER 16 283.e5
283.e5

SELF-ASSESSMENT REVIEW QUESTIONS ANSWERS


1. In evaluating CT scanning for lung cancer as reported in the 1. (a, b, d)  Note that to achieve a reduction in lung cancer and all
NLST trial, which of the following statement(s) are true? cancer mortality in this study, a high prevalence of false-positive
a. CT screening reduced mortality from lung cancer by 20%. findings was required.
b. Of the CT findings, 95% were false positives. 2. (b)  Mammography and tomosynthesis with modern systems
c. CT screening and chest x-ray screening were equally have comparable radiation doses. Younger patients are more
ineffective in detecting lung cancer. at risk of carcinogenesis than older patients. MRI scans carry
d. CT screening was associated with decreased all-cause no risk of irradiation but carry risks of adverse reactions
mortality in this trial. to IV contrast, especially in patients with impaired renal
2. Which of the following statement(s) are true regarding radiation function (NSF).
dose in imaging? 3. (c)  Ultrasound may be the least expensive study to perform,
a. Mammography results in a much lower dose than breast but it may not be the most accurate in the abdomen; thus it is
tomosynthesis. not the most cost-effective. MRI has low sensitivity for
b. CT and nuclear medicine procedures represent over 75% of detecting contrast agent molecules versus optical and nuclear
the imaging-related radiation dose in the United States. methods. Thus large amounts of MRI contrast material must be
c. Age at the time of irradiation is not a major factor in given to obtain a suitable signal in vivo. PET can be quantified
potential radiation-induced carcinogenesis. for SUV if performed correctly per RSNA QIBA guidelines.
d. MRI scans with contrast agent carry no risk of side effects.
3. Which of the following statement(s) are true about imaging
methods in cancer?
a. Ultrasound is the most cost-effective method in abdominal
imaging.
b. MRI is the most sensitive method for detecting low numbers
of contrast agent molecules compared with nuclear or optical
methods.
c. Radiographs have higher resolution than ultrasound images
or nuclear imaging scans.
d. FDG-PET studies cannot be quantified.
D. CLINICAL TRIALS
17  Biostatistics and Bioinformatics in
Clinical Trials
Brian P. Hobbs, Donald A. Berry, and Kevin R. Coombes

S UMMARY OF K EY P OI N T S
• The process of conducting cancer • Modern technology and advanced normalization procedures and
research must change in the face of analytic methods are directing the rigorous statistical methods to
prohibitive costs and limited patient focus of medical research to subsets account for sample collecting, batch
resources. of disease types and to future trials effects, multiple testing, confounding
• Biostatistics has a tremendous across different types of cancer. covariates, and any other potential
impact on the level of science in • A consequence of the rapidly biases.
cancer research, especially in the changing technology for generating • Best practices in developing
design and conducting of clinical “omics” data is that biologic assays prediction models include public
trials. are often not stable long enough to access to the information, rigorous
• The bayesian statistical approach to discover and validate a model in a validation of the model, and model
clinical trial design and conduct can clinical trial. lockdown before its use in patient
be used to develop more efficient • Bioinformaticians must use care management.
and effective cancer studies. technology-specific data

BIOSTATISTICS APPLIED TO inference being the entire trial rather than, for example, a patient within
CANCER RESEARCH the trial.
For reasons discussed in this chapter, much interest has recently
The modern era of cancer therapy raises major issues regarding statistical been expressed in the bayesian approach to statistics. In the first half
inference and study design. First, as biomarkers become available, of the 20th century there were few bayesian biostatisticians, and few
they divide cancers into ever smaller subsets, with unique biomarker- biostatisticians knew what it meant to be bayesian.
defined combinations of targeted therapies for each subset. Soon every The bayesian approach predates the frequentist approach. Thomas
patient with cancer will have an orphan disease. A related issue is the Bayes developed his treatise on inverse probabilities in the 1750s, and
ever-increasing cost of drug development and the consequent cost of it was published posthumously in 1763 as “An Essay Towards Solving
delivering care to persons with cancer. Unless we modify our approaches a Problem in the Doctrine of Chances.”
to cancer research, we will not be able to afford the therapies we In the 200 years after Bayes, the discipline of statistics was influenced
develop, and innovation will cease. by probability theory and, in particular, games of chance, dating to
Biostatistical philosophy is critical in understanding the state of the early 1700s.1–3 This view focused on probability distributions of
cancer research today and where it is heading. Biostatistics has had outcomes of experiments, assuming a particular value of a parameter.
an immense impact on the level of science in cancer research, especially A simple example is the binomial distribution. This distribution gives
in designing and conducting clinical trials. However, as we make the probabilities of outcomes of a specified number of tosses of a
progress in developing better cancer therapeutics, patients’ prognoses coin with known probability of heads, which is the distribution’s
improve, and clinical trials get larger because they are event driven. parameter. The binomial distribution continues to be important today.
The irony is that despite the development of cancer in increasing For example, it is used in designing cancer clinical trials in which the
numbers of persons in the world, fewer clinical investigations are end point is dichotomous (tumor response or not, say) and assuming
possible because of limited resources, and fewer drugs can be developed a predetermined sample size.
despite the burgeoning number of potential anticancer drugs. False-
positive and false-negative conclusions are well known, but the most CLINICAL TRIALS
frustrating problem in the modern era may be the number of “false
neutrals”—drugs and therapeutic strategies that are never investigated A clinical trial is an experiment involving human subjects with the
because of insufficient resources. goal of evaluating one or more treatments for a disease or condition.
The history of biostatistics is dominated by the so-called frequentist A randomized controlled trial (RCT) compares two or more treatments
approach. An alternative approach, the bayesian approach, was largely in which treatment assignment is determined by chance, such as by
ignored as biostatistics developed into a discipline. In the frequentist rolling a die or tossing a coin.
approach, probabilities are defined as long-run proportions. The The traditional statistical approach is to consider two possible
focus is on the final results of a clinical trial, say, with the unit of values of the unknown parameters, such as the tumor response rate

284
Biostatistics and Bioinformatics in Clinical Trials  •  CHAPTER 17 285

0.25
Probability of number of responses
0.20

0.15

0.10

0.05

0.00
0 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20
Number of responses

Figure 17.1  •  Probabilities for number of responses when the rate of response is r = 20% (red bars) and when r = 50% (blue bars). Like all probability
distributions, the sums of the heights of the red bars and the blue bars both equal 1.

r. For one value of r the treatment has no useful benefit, the so-called The distinction between rate and probability in the aforementioned
null hypothesis. The alternative hypothesis is a value of r that is clinically description is important, and failing to discriminate these terms has
important in the sense of meriting future development. led to much confusion in medical research.4 The type I error rate is a
Consider a single-arm clinical trial with the objective of evaluating probability only when one assumes that the null hypothesis is true.
r. The null value of r is taken to be 20%. The alternative value is r = Probabilities of events requiring the truth of the null hypothesis are
50%. The trial consists of treating n = 20 patients. The exact number not available in the frequentist approach, and indeed this is a principal
of responses is not known in advance, but it is known to be either 0 contrast with the bayesian approach (described later).
or 1 or 2 on up to 20. The relevant probability distribution of the Suppose a trial is conducted and 9 of 20 patients respond. One
outcome is binomial, with one distribution for r = 20% and a second concludes that the null, r = 20%, is rejected because 9 is greater
distribution for r = 50%. These distributions are shown in Fig. 17.1, than the predetermined cut point of 8. However, the results are
with red bars for r = 20% and blue bars for r = 50%. More generally, more convincing than had the result been exactly 8. Such reasoning
there is a different binomial distribution for each possible value of r. led to the convention of reporting a P value, an “observed type I
Both the frequentist and bayesian approaches to clinical trial design error rate.” This is the type I error rate had the predetermined cut
and analysis use distributions such as those shown in Fig. 17.1, but point been set to the observed number. Thus the P value is .0100, as
they use them differently. calculated earlier.
Values of r other than 20% and 50% are possible. The standard
Frequentist Approach frequentist approach to representing the range of possibilities is to
provide a confidence interval, which is the set of values of r that would
The frequentist approach to inference is based on error rates. A type not be rejected as null hypotheses based on the data actually observed:
I error is rejecting a null hypothesis when it is true, and a type II 9 responses out of 20 patients (or 45%). For these data, the values of
error is accepting the null hypothesis when it is false, in which case r that would be rejected are those less than 26%. Because very large
the alternative hypothesis is assumed to be true. It seems reasonable values of r are also inconsistent with the observed rate of 45%, it is
to reject the null, r = 20% (red bars in Fig. 17.1), in favor of the conventional to also exclude large values from the confidence interval.
alternative, r = 50% (blue bars in Fig. 17.1), if the number of responses Assuming type I error rates of 5% for both small and large values
is sufficiently large. Candidate values of “large” might reasonably be of r, the resulting confidence interval is from about 26% to 61%,
those in which the red and blue bars in Fig. 17.1 start to overlap, which is a 90% confidence interval in the sense that 5% is excluded
perhaps 9 or greater, 8 or greater, 7 or greater, or 6 or greater. on both sides. Excluding only 2.5% on each side provides a 95%
The type I error rates for these rejection rules are the respective confidence interval: 23% to 64%. Ninety-five percent confidence
sums of the heights of the red bars in Fig. 17.1. For example, when intervals are commonly used and are always somewhat wider than 90%
the cut point is 9, the type I error rate is the sum of the heights of confidence intervals.
the red bars for 9, 10, 11, and so on, which is 0.007387 + 0.002031
+ 0.000462 + … = 0.0100. When the cut points are 8, 7, and 6, Bayesian Approach
the respective type I error rates are 0.0321, 0.0867, and 0.1958. One
convention is to define the cut point so that the type I error rate is no A major difference between the bayesian and frequentist approaches is
greater than 0.05. The largest of the candidate type I error rates that the use of probability distributions to represent unknown values. In the
is less than 0.05 is 0.0321, the test that rejects the null hypothesis if frequentist approach, probabilities apply only for “data.” Parameters
there are eight or more responses. The type II error rate is calculated that index data distributions (such as r in the aforementioned example)
from the blue bars in Fig. 17.1, wherein the alternative hypothesis is are unknown but are fixed and thus are not subject to probability state-
assumed to be true. The sum of the heights of the blue bars for 0 up ments. In the bayesian approach, all unknowns, including parameters,
to 7 responses is 0.1316. Convention is to consider the complementary have probability distributions.
quantity and call it “statistical power:” 0.8684, which rounds off In the example in which 20 patients yielded 9 responses, suppose
to 87%. that r is assumed to be either 20% or 50%. The bayesian conclusion
286 Part I: Science and Clinical Oncology

is the probability of r = 50%, given the final results. (This is 1 minus of a diagnostic test is the prevalence of the condition or disease in
the probability of r = 20% under the assumption that these are the question. Prior probability depends on other evidence that may be
only two possible values of r.) The calculation uses the Bayes rule. available from previous studies of the same therapy in the same disease,
The method is the same as when finding the positive predictive value or related therapies in the same disease or different diseases. Assessment
of a test as it relates to the condition’s prevalence and the test’s sensitivity may differ depending on the person making the assessment. Subjectivity
and specificity. The Bayes rule is also called the rule of inverse probabilities is present in all of science; the bayesian approach has the advantage
because it relates the probability of some event A, given that event of making subjectivity explicit and open.5
B has occurred, with the probability of event B given that event A When the prior probability equals 0.50, as assumed in Fig. 17.2,
has occurred. the posterior probability of r = 20% is 0.0441. Obviously, this is
The calculation is intuitive when viewed as a tree diagram, as in different from the frequentist P value of 0.0100 calculated earlier.
Fig. 17.2. Fig. 17.2A shows the full set of probabilities. The first Posterior probabilities and P values have very different interpretations.
branching shows the two possible parameters, r = 20% and r = 50%. The P value is the probability of the actual observation, 9 responses,
The probabilities shown in the figure, 0.50 for both, will be discussed plus that of 10 responses, 11 responses, and so on, assuming the null
later. The data are shown in the next branching, with the observed distribution (the red bars in Fig. 17.1). The posterior probability is
data, nine responses (nine resp) on one branch and all other data on computed conditionally with respect to the actual observation of nine
the other. The probability of the data given r, which statisticians call responses and is the probability of the null hypothesis—that the red
the likelihood of r, is the height of the bar in Fig. 17.1 corresponding bars are in fact correct—but assuming that the true response rate is
to nine responses, the red bar for r = 20%, and the blue bar for r = a priori equally likely to be 20% and 50%.
50%. The rightmost column in Fig. 17.2A gives the probability of P values are not intuitive in that they are calculated conditionally
both the data and r along the branch in question. For example, the with respect to a hypothesis that seems unlikely to be true in view of the
probability of r = 20% and “nine resp” is 0.50 multiplied by 0.0074. results (the null hypothesis) and because they depend on probabilities
The probability of r = 20% given the experimental results depends of possible occurrences that were not observed. For example, if there
on the probability of r = 20% without any condition, its so-called are two candidate null distributions with the same probability of
prior probability. The analog in finding the positive predictive value the actual observation, the one with the smaller “tail” of unobserved
values will have a smaller P value. Because they are counterintuitive,
misinterpretations of P values abound. People usually convert a P
value into something they understand but that is wrong, and the
misinterpretation usually has a bayesian ring to it; for example, “The P
Probability Probability
value is the probability that the results could have occurred by chance
Probability Prior of data of data alone.” An objection to bayesian inferences is that they are specific to
of r probability Data given r given r assumed prior probabilities. A sensible type of report that addresses
this concern is the following: “If your prior probability is this, then
9 resp 0.0074 0.0037
your posterior probability is that.”
As an example of such a report, Fig. 17.3 shows the relationship
0.50
r = 20% between the prior and posterior probabilities. The figure indicates
Not 9 0.9926 0.4963 that the posterior probability is moderately sensitive to the prior.
Someone whose prior probability of r = 20% is 0 or 1 will not be
swayed by the data. However, as Fig. 17.3 indicates, the conclusion
9 resp 0.1602 0.0801 that r = 20% has low probability is robust over a broad range of prior
r = 50% probabilities. A conclusion that r = 20% has moderate to high prob-
0.50 ability is possible only for someone who was convinced that r = 20%
in advance of the trial.
A Not 9 0.8398 0.4199 In the example, r was assumed to be either 20% or 50%. It would
be unusual to be certain that r is one of two values and that no other
values are possible. A more realistic example would be to allow r to
9 resp 0.0074 0.0037 have any value between 0 and 1 but to associate weights with different
values depending on the degree to which the available evidence supports
0.50 those values. In such a case, prior probabilities can be represented
r = 20% with a histogram (or density). A common assumption is one that
Not 9 0.9926 0.4963 reflects no prior information that any particular interval of values of
r is more probable than any other interval of the same width. The
9 resp 0.1602 0.0801 corresponding density is flat over the entire interval from 0 to 1 and
r = 50% is shown in red in Fig. 17.4A.
0.50 The probability of the observed results (9 responses and 11
nonresponses) for a given r is proportional to r9(1 − r)11, with the
B Not 9 0.8398 0.4199 likelihood of r based on the observed results. The prior density is
updated by multiplying it by the likelihood. Because the prior density
is constant, this multiplication preserves the shape of the likelihood.
Probability of 9 responses: 0.0037 + 0.0801 = 0.0838 Thus the posterior density equals just the likelihood itself, shown in
Probability that r = 20% given 9 responses: 0.0037/0.0838 = 0.0441 green in Fig. 17.4A.
C Probability that r = 50% given 9 responses: 0.0801/0.0838 = 0.9559
The bayesian analog of the frequentist confidence interval is called
a probability interval or a credibility interval. A 95% credibility interval
Figure 17.2  •  (A) Tree diagram showing probabilities and conditional is shown in Fig. 17.4B: r = 26% to 66%. It is similar to but different
probabilities when there are 20 observations. (B) Modification of (A), restricting from the 95% confidence interval discussed earlier: 23% to 64%.
to the actual number of nine responses (9 resp), with “Not 9” grayed out. Confidence intervals and credibility intervals calculated from flat prior
Possible observations that were not observed are not considered. (C) Calculations densities tend to be similar, and indeed they agree in most circumstances
demonstrating the Bayes rule. when the sample size is large. However, their interpretations differ. A
Biostatistics and Bioinformatics in Clinical Trials  •  CHAPTER 17 287

credible interval has a particular probability that the parameter lies now serves as the prior density for the next trial. Multiplying that
in the interval. Statements involving probability or chance or likelihood density by likelihood number 2 gives the posterior density based on
cannot be made for confidence intervals. the data from both trials, as shown in Fig. 17.5. The order of observation
Any interval is an inadequate summary of a posterior density. For is not important. Updating first on the basis of the second trial gives
example, although r = 45% and r = 65% are both in the 95% credibility the same result. Indeed, multiplying the prior density, likelihood
interval in the aforementioned example (see Fig. 17.4), the posterior number 1, and likelihood number 2 in Fig. 17.5 gives the posterior
density shows the former to be five times as probable as the latter. density shown in the figure.
A characteristic of the bayesian approach is the synthesis of evidence. The calculations of Fig. 17.5 assume that r is the same in both
The prior density incorporates what is known about the parameter trials. This assumption may not be correct. Different trials may well
or parameters in question. For example, suppose another trial is have different response rates, say r1 and r2. In the bayesian approach,
conducted under the same circumstances as the aforementioned example these two parameters have a joint prior density. One way to incorporate
trial, and suppose the second trial yields 15 responses among 40 into the prior distribution the possibility of correlated r1 and r2 is to
patients. Fig. 17.5 shows the prior density and the likelihoods from use a hierarchical model in which r1 and r2 are regarded to be sampled
the first and second trials. Multiplying likelihood number 1 by the from a probability density that is unknown, and therefore this density
prior density gives the posterior density, as shown in Fig. 17.4. This itself has a probability distribution. More generally, there may be
multiple sources of information that are correlated and multiple
parameters that have an unknown probability distribution. A hierarchi-
cal model allows for borrowing strength across the various sources
1
depending in part on the similarity of the results.6–8

0.9 0.82
Posterior probability that r = 20%

0.8
7
0.7
Posterior density
0.6 6 Prior density
0.5 Likelihood 1
5 Likelihood 2
0.4
0.3 0.0441 4

0.2
3
0.1
2
0
0 0.1 0.2 0.3 0.4 0.5 0.6 0.7 0.8 0.9 1
1
Prior probability that r = 20%
0
Figure 17.3  •  Influence of prior probability of rate of response, r = 20%,
on its posterior probability after observing 9 responses among 20 patients. The 0 0.1 0.2 0.3 0.4 0.5 0.6 0.7 0.8 0.9 1
calculation uses the Bayes rule, a different application of the Bayes rule for r
each possible prior probability. The figure highlights the posterior probability
of 0.0441 when the prior probability is 0.50, as calculated in Fig. 17.2C. If Figure 17.5  •  Two trials, conducted under the same circumstances, with
the prior probability is very high—for example, 0.99—then the posterior data indicated by the likelihoods 1 and 2. The posterior density is that for
probability is about 0.82, still high but much reduced from its prior probability. the data from both trials.

4 4

3 3

2 2

1 1
2.5% 2.5%

0 0
A 0 0.1 0.2 0.3 0.4 0.5 0.6 0.7 0.8 0.9 1 B 0 0.1 0.2 0.3 0.4 0.5 0.6 0.7 0.8 0.9 1
r r
Posterior density
Prior density

Figure 17.4  •  Histograms of probability distributions. (A) The horizontal red line shows the prior density, assumed to be flat and hence “noninformative”
in the sense described in the text. The blue curve shows the posterior density on the basis of 9 responses among 20 patients. Because the prior density is flat,
the posterior density is proportional to the probability of the results for a given response rate, r, which is r9(1 − r)11. The proportionality constant makes the
area under the curve equal to 1, the same as the area under the prior density. (B) The area under the posterior density to the left of r = 0.26 equals 2.5%,
and the same is true for that to the right of 0.66. The values between these two limits form a 95% credibility interval for r.
288 Part I: Science and Clinical Oncology

A rather different application of bayesian hierarchical models, represented a response to the treatment—in fact, the trial would have
called a tumor agnostic, is destined to become important in cancer stopped after four responses in four patients.
clinical trials. Consider an agent targeted at a particular mutation. It happens that for this simple adaptive design it is possible to find
There may be subsets of patients who harbor this mutation across a its operating characteristics algebraically, but the calculations are tedious.
broad range of tumor types. The mutation may be rare in some or In more complicated circumstances, algebraic calculations may not
all tumor types. Mustering a compelling clinical trial in any given be possible and simulations are necessary.
tumor type may be impossible. Instead, one might conduct a single Simulations are easy to describe. To address r = 50%, start with a
trial across 10 tumor types, say, regarding response rates r1, r2, …, r10 fair coin, one with probability of heads equal to 50%, and interpret
as a sample from a population. The population distribution may be “heads” to mean response. Whenever a patient is accrued to the trial,
homogeneous, in which case there is substantial “borrowing of strength” toss the coin, observe the result, and update the probability that r =
across the tumor types. This may enable a claim of effectiveness in 50% on the basis of the tosses thus far. Stop the trial if this probability
tumor types that, when left to stand alone, would have wide credibility is greater than 95% and make a mark so indicating. If the number
intervals because of their small sample sizes. On the other hand, if of tosses reaches 20 without having made a mark, then stop the trial
the population of response rates is heterogeneous, then the observed because you have hit the maximal sample size. When the trial stops,
response rates will be highly variable, and borrowing will occur mainly make a note of the total number of tosses, say n, in the trial. Repeat
across neighboring tumor types. the trial a total of 10,000 times. Count the number of marks and
We next apply the bayesian perspective in two important directions divide by 10,000. This is the estimated power of the study assuming
that are the focus of attention in modern cancer research. r = 50%. Tabulate the 10,000 values of n to give an estimate of the
distribution of the final sample size under the alternative hypothesis.
Adaptive Designs of Clinical Trials (You should have a mark for every trial with n <20 and also for some
trials with n = 20.)
Randomization was introduced into scientific experimentation by To find the type I error rate, repeat the aforementioned process
R.A. Fisher in the 1920s and 1930s and applied to clinical trials with another set of 10,000 iterations generated using a device that
by A.B. Hill in the 1940s.9 Hill’s goal was to minimize treatment has probability of heads equal to 20%. (An example device is a regular
assignment bias, and his approach changed medicine in a funda- 20-sided die with “heads” labeled on four of the sides.) The proportion
mentally important way. The RCT is now the gold standard of of marks is the estimated type I error rate. The tabulated values of n
medical research. A mark of its impact is that the RCT has changed are the estimated distribution of the final sample size under the null
little during the past 65 years, except that RCTs have gotten bigger. hypothesis.
Traditional RCTs are simple in design and address a small number of Tossing a coin perhaps 100,000 times will take a while, especially
questions, usually only one. However, progress is slow, not because of because you will have to keep track of which trial you are in and
randomization but because of limitations of traditional RCTs. Trial whether you have achieved the stopping boundary for that trial. The
sample sizes are prespecified. Trial results sometimes make clear that good news is that a simple program running on a modern desktop
the answer was present well before the results became known. The computer can simulate 10,000 trials in a few seconds. The computer
only adaptations considered in most modern RCTs are interim analyses does all the work. Moreover, an additional few seconds gives you
with the goal of stopping the trial on the basis of early evidence of another 10,000 simulated trials assuming another value of r by simply
efficacy or for safety concerns. There are usually few interim analyses, changing the value of r in the program.
and stopping rules are conservative. As a consequence, few trials Fig. 17.6 shows the sample size distribution for 10,000 trials under
stop early. the assumption that r = 50%. The estimated type II error rate is
In traditional RCTs, randomization proportions are fixed through- 0.1987, which is the proportion of these 10,000 trials that reached
out. The possibility that the accumulating data in the trial can influence n = 20 without ever concluding that the posterior probability of r =
randomization probabilities or other aspects of a trial’s course may 50% is at least 95%. The sample size distribution when r = 20% is
affect the trial’s performance characteristics, including its type I error not shown but is easy to describe: 9702 of the trials (97.02%) went
rate and statistical power. Moreover, these effects may be difficult to to the maximum sample size of n = 20 without hitting the posterior
analyze with traditional mathematics. However, modern computers probability boundary and the other 3% stopped early (at various
and software make traditional mathematics unnecessary. Any prospective values of n <20) with an incorrect conclusion that r = 50%. Thus
trial design, however complicated, can be simulated. A prospective 3% is the estimated type I error rate. The “estimated” aspect of these
trial design is an automaton. At any time during the trial and based statements is because there is some error due to simulation. Based on
on the information available from trial participants, the next patient 10,000 iterations, the standard error of the estimated power is small,
is assigned a particular therapy, possibly based on an adaptive randomiza- but positive: 0.4%. The standard error of the type I error rate is less
tion scheme. Any trial that has a prospective design can be simulated. than 0.2%.
Virtual patients can be generated with their outcomes depending on Because the bayesian approach allows for updating knowledge
assumed parameter values and treatment assignment according to the incrementally as data accrue, even after every observation, it is ideally
prospective design. Replicating the trial 10,000 times, say, gives a firm suited for building efficient and informative adaptive clinical trials.10–12
handle on the trial conclusion for the parameter values assumed in The bayesian approach serves as a tool. As evinced in the aforementioned
the simulation. example, simulations enable calculation of traditional frequentist
Consider a simple case, a variant of the earlier example in which measures of type I error rate and power. The Institute of Medicine
the response rate r was assumed to be either 20% or 50%. Set the (IOM) recently advocated for the need to restructure the entire clinical
maximum number of patients in the trial to 20. Stop the trial with trial system to advocate for adaptive designs and to address other
a claim favoring the alternative hypothesis r = 50% whenever the deficiencies that limit the effectiveness and efficiency of trials.13 The
probability of r = 50% versus r = 20% is at least 95% (and therefore initiative was reaffirmed with the 21st Century Cures Act passed by
the probability of r = 20% is less than 5%). the United States House of Representatives in July 2015.
As a check that the reader is following this description: the binomial Innovations in adaptive design have been proposed to address all
distribution assumed earlier is no longer relevant, even if the final aspects of drug development. Despite the extensive use of traditional
sample size happens to be 20. Consider an extreme case. The binomial dose-escalation studies, their limitations with monitoring rules based
distribution gives positive probability to 20 responses regardless of on algorithmic formulations, such as the 3 + 3 design, have been well
the value of r (unless r = 0). However, the adaptive design would have described (see e.g., Wong and colleagues14). Recent innovations have
stopped long before getting to 20 patients when every patient effectuated designs for phase I trials that use the sequential application
Biostatistics and Bioinformatics in Clinical Trials  •  CHAPTER 17 289

2500

Number of simulated studies


2000

1500

1000

500

0
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20
n = Final sample size

Figure 17.6  •  Sample size distribution when the response rate is r = 50% in the adaptive stopping example in the text. Among the 2353 simulated trials
that finished with maximum sample size of n = 20 were 366 that achieved a posterior probability of r = 50% greater than 0.95 at that point; the other 1987
simulated trials reached n = 20 without such a conclusion, and thus 0.1987 is the estimated type II error rate (equivalently, the estimated power is 0.8013).
The mean sample size in the 10,000 simulations was 12.2, with a standard deviation of 5.7. Some sample sizes never occurred. For example, the stopping
bound for the probability of r = 50% requires at least five responses in n = 6 patients. However, both four and five responses also call for stopping at n = 5,
and thus five responses cannot occur at n = 6.

of decision rules derived from formal probability models15 and have biologic molecules: genomics for sequence-based studies of DNA,
facilitated designs that are devised to optimize dose and schedule epigenomics for DNA modifications, transcriptomics for RNA,
conjointly.16,17 Several authors have explored the extent to which proteomics for proteins, and metabolomics for small molecules. Predic-
platform trials18 (which enable study arms to be dropped or added tors that can potentially be used for classification, diagnosis, prognosis,
during the course of enrollment) may yield improvements in efficiency or selection of therapy may be found in any of these classes of molecules.
as well as improve treatment efficacy for trial participants. The role of bioinformatics is to manage and organize the data and
A patient’s experience on receiving a particular therapeutic strategy to make possible the statistical analysis needed to discover and validate
is often a complex synthesis of measures that describe both the extent predictive models based on omics technologies.
of induced harm as well as realized clinical benefit achieved. Thus
“patient response” in itself is often difficult to characterize, especially Challenges
when designing studies to compare the multimodal treatment strategies
that are actually used as matter of routine in clinical oncology. Clinical Numerous challenges must be overcome to incorporate omics data
translation has been limited by statistical testing procedures and designs into clinical oncology. The technology changes so rapidly that it can
that rely on reductive characterizations of a patient’s experience based be difficult to develop an assay that will remain stable long enough
on binary and univariate endpoints. A few authors have put forth to discover and validate a model in a clinical trial. A wide variety of
innovations to address these limitations19–22 and to establish designs omics technologies are available to assay different classes of molecules,
that account for the dynamic nature of cancer care in settings that which requires researchers and analysts to develop a broad range of
require multiple therapies administered in stages over the course of expertise, not only in the individual technologies, but also in statistical
treatment.23,24 methods to integrate the resulting data. Omics data sets often are
Taking an adaptive approach is fruitless when there is no information hampered by batch effects, which can be overcome only by careful
to which to adapt. In particular, for long-term end points there may attention to experimental design and the development of standard
be little information available when making an adaptive decision. protocols. Statistically, the analysis of omics data sets is a struggle
However, early indications of therapeutic effect are sometimes available, against issues of multiple testing and overfitting. The development of
including longitudinal biomarkers and measurements of tumor burden, second-generation sequencing technologies poses additional challenges
for example. These indications can be correlated with long-term clinical from the sheer volume of data and the need for substantial computer
outcomes to enable better interim decisions.7,12 power to interpret the data.
A limitation of adaptive approaches is that they require the ability
to update the outcome database and connect it to the patient assignment Pace of Technologic Change
algorithm. Another limitation is that an adaptive design, although
fully prospective, can be complicated to convey to investigators, patients, The first microarrays that could simultaneously measure the messenger
institutional review boards, and regulators. RNA (mRNA) expression levels of thousands of genes were developed
in the mid 1990s.25,26 Within a few years, they were being used to
BIOINFORMATICS study cancer.27–31 The technology evolved rapidly. For example, one
of the major manufacturers of microarrays, Affymetrix, began with
As a discipline, bioinformatics sits at the interface where biology and HuGeneFL GeneChip arrays that contained 6800 probe sets. They
medicine meet a confluence of quantitative sciences, including computer advanced over the course of little more than a decade through the
science, mathematics, and statistics. Its primary application to oncology U95A (12,000 probe sets), U133A (22,000), U133Plus2.0 (54,000),
is sifting through genome-scale (“omics”) data sets to identify biomarkers Human Gene ST 1.0 and 2.0, and Human Exon ST 1.0 and 2.0
and molecular signatures that can be used to predict clinically relevant microarrays. The typical time between the introductions of successive
outcomes. Each omics technology focuses on a particular class of generations of microarray platforms was 2 or 3 years. Nevertheless,
290 Part I: Science and Clinical Oncology

as that decade ended, researchers were increasingly moving away from data after quantification, summarization, and normalization is often
microarray technology and toward RNA sequencing technology as similar even for radically different technologies, in which case similar
their primary tool to measure mRNA abundance. The technologies statistical methods can be applied. The exact form of the statistical
used to measure DNA copy number alterations went through a similar analysis depends less on the nature of the technology platform and
evolution during this period. more on the design of the experiment. Bioinformaticians must consider
This rapid pace of change poses serious challenges when trying to several factors, such as the kinds of patient samples that are being
apply these technologies in clinical trials, which may take several years contrasted, whether the outcome measures binary, continuous, or
to conduct. Often the technologic platform used to develop a predictive time-to-event data, and the covariates (e.g., age, stage, grade, and
model differs from the one proposed to test the predictions in a clinical smoking status) for which they must account in the analysis.
trial. It is even possible for a manufacturer to discontinue production It is unreasonable to expect manufacturers of scientific instruments
of a platform that researchers are using or proposing to use in a clinical to have the expertise required to program the wide variety of sophis-
trial. Every time the platform changes, the assay must be recomputed ticated statistical methods needed to account for differences in
and revalidated (preferably by biostatisticians and bioinformaticians experimental design. It is surprising, however, that manufacturers do
working with a laboratory that has been certified according to the not always know the best ways to process their own raw data. For
Clinical Laboratory Improvement Amendments [CLIA] of the US example, the first Affymetrix microarrays were designed with use of
Code of Federal Regulations). multiple pairs of “perfect match” and “mismatch” (MM) probes to
target each gene. The idea was that the MM probes could be used to
Breadth of Technologies estimate nonspecific cross-hybridization, and so their initial algorithm
quantified the expression of a gene as the average difference between
At the same time that microarray technologies were expanding to the perfect match and MM probes. Li and Wong43 recognized that
allow the entire transcriptome to be assayed in a single experiment, different probes for the same gene have different affinities, and thus
new technologies were emerging that focused on other biologically their mean expression will vary even within the same sample. They
important molecules. The Cancer Genome Atlas (TCGA) started replaced the simple average with a statistical model that accounted
assaying approximately 500 tumors per cancer type by using a broad for different probe affinities.43 Bolstad and colleagues44 and Irizarry
spectrum of omics technologies.32 Initial plans called for microarray and associates45 showed that the MM probes increased the noise in
approaches to measure mRNA expression, microRNA (miRNA) the summary measurements with no compensating gain in signal
expression, DNA copy number alteration, and methylation. These clarity; they introduced an improved statistical method known as the
plans were supplemented by direct Sanger sequencing of a predefined robust multiarray average (RMA). Most current analyses of Affymetrix
set of cancer-related genes and by proteomic techniques, including gene expression data use RMA.
mass spectrometry and reverse-phase protein lysate arrays. More recently, Many advances in the methods for processing and analyzing
TCGA began applying second-generation sequencing technology to omics data sets have come from academics and are made available as
study DNA (whole-genome or whole-exome sequencing), RNA open source software. Bioconductor (http://www.bioconductor.org)
(RNA-Seq), and methylation (chromatin immunoprecipitation on is the largest repository of such software packages, written for the
sequencing [ChIP-Seq]) in these tumor samples.33,34 R statistical programming environment.46,47 The Bioconductor
In at least one case, an entire class of biologically interesting repository is the equivalent of a hardware store for statisticians and
molecules is itself relatively new. The first miRNA was discovered in bioinformaticians searching for tools with which to analyze their latest
1993 by cloning the lin-4 gene in Caenorhabditis elegans.35 That there data set. An alternative approach is provided by GenePattern48 (http://
were numerous (highly conserved) miRNAs in different species did www.broadinstitute.org/cancer/software/genepattern/). GenePattern is
not become known until the simultaneous publication of three papers a Web service through which data can be uploaded and analyzed by
in 2001.36–38 A year later, Calin and colleagues39 demonstrated that biologists as well as statisticians. Modules can be written and shared
miRNAs played an important role in cancer by showing that the in a variety of programming languages, then assembled into reusable
“tumor suppressor genes” contained in the minimal deleted region of self-documenting pipelines.
a recurrent deletion of chromosome 13q in chronic lymphocytic
leukemia consisted of miRNA-15a and miRNA-16-1. Version 19 of Batch Effects and Experimental Design
the reference database miRBase (http://www.mirbase.org/), maintained
by the Wellcome Trust Sanger Institute, now lists 1600 precursors Batch effects are an unavoidable characteristic of data collected using
and 2042 mature human miRNAs.40–42 In addition, of course, there cutting-edge technologies on research-grade scientific instruments.49
are now microarrays that simultaneously measure (almost) all of them. The instruments are often temperamental, requiring frequent tuning
Each omics technology requires specialized methods for processing and calibration. Reagents change, and new printings of the “same”
the raw data and converting it into a form that can be analyzed to microarray can be subtly different. As a result, a batch of tumor
discover or validate biomarkers and signatures. Often the raw data samples analyzed in November may differ in many ways (although
are fluorescence-based images that must be quantified and summarized. not of biologic interest) from a similar batch analyzed in February.
Each assay requires its own form of normalization that is intended to These technologic effects are often large enough to swamp any interest-
correct for technologic artifacts that may distort the measurements. ing biology, and they can occur on the timescale of days rather than
At its simplest, the process of normalization is rescaling the data to months. Unless accounted for in the experimental design, batch effects
account for differences in starting material between samples; if one can ruin a perfectly good experiment. For example, in 2002, Petricoin
starts an experiment with twice as much total RNA from one sample, and colleagues50 published an article that claimed they had developed
then one would expect all the measurements to be twice as large. a tool that could be used to diagnose ovarian cancer based on proteomic
More complicated normalization schemes, based on sophisticated patterns detected in serum samples. Their results were soon questioned;
statistical models, are common. Bioinformaticians must understand it eventually transpired that the signals they had detected were
enough about how different biologic assays work to select (or develop) technologic artifacts, caused by running all of the controls on their
appropriate methods to process each kind of data. mass spectrometer before all the cases.51–53
Historically, when manufacturers introduced new instruments, they There are several ways to deal with batch effects. First, pay attention
also provided software to quantify and analyze the data that they to experimental design. Apply the basic principles of randomization
produced. Statisticians and bioinformaticians, sometimes with good and blinding to ensure that the contrasts of interest (tumor versus
reason, tend to be leery of the “black box” software packages that normal, or responder versus nonresponder, for example) are not
accompany scientific instruments. For one thing, the structure of the confounded with any batch effects that may be present. Second, if
Biostatistics and Bioinformatics in Clinical Trials  •  CHAPTER 17 291

batch effects are suspected, there are existing statistical methods to used to construct a multivariate computational model that will be
model them and, to some extent, remove them.54–56 able to perfectly predict any desired outcome variable on any given
Additional challenges arise in the context of clinical trials. The set of biologic samples. Because the data are random, it is guaranteed
standard normalization methods for most omics technologies require that any such model is bogus: it “overfits” the existing data and will
analyzing the entire set of data at once, which is impossible when not generalize to new data. Second, the P value correction methods
patients arrive one by one and a decision about how to randomly that work one feature at a time will not help. For example, the number
assign them to treatment arms depends on the results of an individual of 35-feature signatures that can be selected from a set of 10,000
assay. In the context of Affymetrix microarrays, this issue was addressed features is approximately a googol (10100). No molecular signature
by the introduction of “frozen RMA,” which computes the normaliza- will ever survive Bonferroni correction of this magnitude. In addition,
tion parameters from a training set and then applies them to new two different 35-feature signatures that share 30 features in common
arrays one at a time.57 Not all omics technologies have completely will be highly correlated, invalidating most of the methods for estimating
faced this issue. In many cases, as with the comparative CT (or ΔΔCT) the FDR. The solution to this problem, however, is simple: any putative
method for quantitative, real-time, reverse-transcription polymerase predictive model based on a complex signature must be validated on
chain reaction (qPCR),58 the best solution may be to run a normalization an independent data set, preferably one collected prospectively with
or calibration standard alongside every experimental sample. respect to the definition of the predictive model.

Multiple Testing and Overfitting Sequencing


At this point, we can assume that we have settled on the technology The move from hybridization-based microarray assays to second-
platform to assay a particular class of biologic molecules. We have generation sequencing technology poses additional bioinformatics
collected the patient samples, randomized their run order, and corrected challenges. First, there is the sheer volume of data. Even after discarding
any batch effects in the processed data. We are now ready to discover the raw images, sequencing data from one sample takes about 1 terabyte
the new biomarkers or molecular signatures that will revolutionize of disk space. Storing these data, moving them across the network,
the practice of clinical oncology. And we run head-on into an enormous and accessing them efficiently requires improvements in computer
statistical hurdle: multiple testing. hardware and software. Second, raw sequence data require heavy-duty
Each omics technology measures thousands of features simultane- computer processing before they begin to become useful. Millions of
ously. (Feature is the standard term in the machine learning literature sequence “reads” must be mapped to the genome before they can be
for a potential predictor. The first step in building a complex predictive summarized to provide information about individual genes or other
model is feature selection—deciding which features to include and which genomic features. In the hybridization approaches, this computational
to leave out of the model. We use the term here for its neutrality; it burden is front-loaded and performed once when designing probes.
covers genes, proteins, miRNAs, single-nucleotide polymorphisms, In sequencing, a computational burden exists for every sample processed.
enhancers, promoters, or anything else that might help us predict Third, because every individual genome is unique, the mapped reads
who will or will not respond to a particular therapy.) Each feature can must be interpreted to identify single-nucleotide polymorphisms, small
be tested separately for its ability to predict the outcome of interest, insertions and deletions (indels), structural variations (translocations
and a P value can be associated with each feature in the usual way for or gene fusions), and DNA copy number variation, either present in
the statistical test being used. However, those P values do not mean germline DNA or acquired somatically. Generating the raw sequence
what one might think they mean. Suppose, for example, that 20,000 data currently takes about a week; mapping and interpretation of the
features are tested, and it is discovered that only 1000 of them have data may take a month to get through the queue on a high-performance
P values less than the magic number of .05. In that case, a reasonable computing cluster.
conclusion is that nothing that has been measured is actually related
to the outcome the test is trying to predict! The problem is that BEST PRACTICES
1000/20000 = 5%, which is the number of small P values one would
expect to occur by chance when 20,000 statistical tests are performed. In response to problems encountered with the premature use of
The traditional statistical response to the problem of multiple gene-expression signatures in clinical trials at Duke University,65 the
testing is to apply a “Bonferroni correction” to the cutoff used to IOM convened a committee to clarify the steps needed to move
define significance. If one is testing for N features, then the P value omics-based signatures from their initial discovery into clinical trials
should be less than 0.05/N in order for one to claim significance at where they can affect patient management and, ideally, improve patient
the 5% level. With 20,000 features, this requirement translates into outcomes.66 The most important committee recommendation was to
a P value below .0000025. One might think that this requirement draw a bright line after the discovery and validation of a predictive
is extreme, and perhaps it is. The Bonferroni correction is extremely model and before its application in a clinical trial. Before crossing that
conservative. It tries to control the “family-wise error rate”; in other line, the test should already be validated, preferably in an independent
words, it attempts to make sure there are no (type I) errors in what set of samples or, if that method is not available, by cross-validation. A
one calls significant. Continuing the hypothetical experiment, suppose clinical assay should be developed in a CLIA-certified laboratory, and
20,000 features are tested, and it is found that 50 of them have a P the complete computational procedure must be completely specified
value below 2.5 × 10−6. In only 5% of such experiments would one and “locked down.” Any change to the procedure requires revalidation
expect to find any errors in the list of 50 features. A less conservative of the method before it is used to affect patient management. An
approach is to control the false discovery rate (FDR), the expected example is changing the prespecified cutoffs that distinguish patients
fraction of false-positive (FP) calls among all positive calls: FP/(FP with high-risk disease from those with low-risk disease; this warrants
+ TP), where TP is a true-positive call.59 Numerous methods have revalidating the method.
been developed to estimate the FDR in omics experiments.60–64 A The IOM recommendations build on a long history of related
few of these methods also allow one to estimate the number or rate guidelines. For example, the Early Detection Research Network
of false-negative calls. Sometimes called type II errors, false-negative proposed a sequence of phases for the development of biomarkers
calls have an opportunity cost in terms of potential discoveries that intended for cancer screening.67 However, because the requirements
are never made. for a good screening biomarker are different from those for a prognostic
Multiple testing becomes much worse when one is evaluating or predictive biomarker, the Early Detection Research Network
molecular signatures. First, it is easy to show that random data (created biomarker phases are not universally applicable. The minimum
with a random number generator) containing enough features can be information about a microarray experiment (MIAME) standard defines
292 Part I: Science and Clinical Oncology

data structures for making microarray data publicly available.68 Many technology and research-grade scientific instruments does not provide
journals require authors of papers that use microarray data to deposit the stability needed for a clinical assay. One consequence is that the
them in a public repository (such as the Gene Expression Omnibus development of a clinical assay may involve a change of technology.
or ArrayExpress) in a format that adheres to the MIAME standard. Instead of using microarrays or RNA-Seq to measure gene expression,
The MIAME data structures apply directly to a wide variety of technolo- the assay may be converted to use qPCR. Whole-genome sequencing
gies. One weakness of MIAME and related standards, such as the will be replaced by sequencing targeted at a few relevant genes. Tests
minimum information about a proteomics experiment (MIAPE) for DNA copy number alterations may be converted to use fluorescence
standard,69 is that, although they describe the collection of metadata in situ hybridization. The role of the clinical laboratory is to demonstrate
about the technology used in an experiment, they do not include a that the assay is analytically reproducible, and, if necessary, to adjust
structured way to store clinical or demographic data about the patients the locked-down predictive model to use the measurements made on
whose samples were used in the experiments. Other important guidelines the new technology platform.
include the reporting recommendations for tumor marker prognostic
studies (REMARK),70,71 the CONSORT statement for randomized Evaluation of Clinical Utility
trials,72 and the STARD initiative for diagnostic studies.73
After developing the clinical assay to implement the locked-down
Discovery Phase predictive model, researchers can start designing the clinical trial.
At this point, regulatory issues come into play. As with all clinical
The discovery phase lasts from the initial experiments through complete trials, researchers must obtain approval from the institutional review
definition of a locked-down computational model to predict an outcome board. However, additional action may also be required. In response
of interest. During this phase, researchers should: to a proposed test to detect ovarian cancer (OvaCheck) that grew
out of the flawed mass spectrometry experiments on serum, to the
• Make the data and metadata publicly available
clinical trials at Duke, and to similar issues elsewhere, the US Food
• Make the computer code and analysis protocols publicly available
and Drug Administration (FDA) has ruled that complex predictive
• Confirm the model by using an independent, preferably blinded,
models are “medical devices,” the regulation of which falls under
set of samples
their jurisdiction. If those devices will be used in a clinical trial
• Lock down the model, including molecular measurements, com-
to guide patient management in any way, then researchers must
putational procedures, and intended clinical use
obtain an investigational device exemption (IDE). The IDE is the
The recommendations to make the data and code available grew medical device equivalent of the better-known investigational new
out of the problems encountered at Duke University,74,75 but they drug application that is needed for testing a new drug in a clinical
reflect a larger concern with reproducible research in computationally trial. The IOM committee recommends that specific members of the
intensive sciences.76–79 Because it is impossible to fully understand the institutional review board be made responsible for considering the
biologic rationale behind a complex predictive model involving tens need for an investigational new drug application or IDE in proposed
or hundreds of genes, it is critically important to be able to confirm clinical trials.
that the statistical analysis to discover the model was performed both Not every clinical trial evaluating the usefulness of a predictive
sensibly and reproducibly. Checking these results requires access to model needs an IDE; the fundamental criterion is whether the
both the original data and the computer code used to analyze it. molecular signature or assay device is being used to direct patient
The requirement to specify the intended clinical use is also critical. management. A prospective-retrospective82 study design in which
Such information should answer the following questions: In what the signature is measured on archived specimens to determine
group of patients will the signature be tested? Will the result of the whether it would have been useful should not require an IDE. Simi-
test be used to screen patients for early diagnosis? Will it provide larly, a prospective clinical trial in which the signature is passively
prognostic information? Will it help select therapy? Will it be used measured but not used to determine patient care would not need
to monitor minimal residual disease or the possibility of recurrence? an IDE.
Different applications require different performance characteristics to
demonstrate the usefulness of a biomarker or signature and thus require Signatures Are Not Enough
different experimental designs and different types of controls. Research
scientists who have the expertise to develop omics signatures but are In most of the literature, “signature” is a synonym for “list of genes.”
not accustomed to designing or running clinical trials can fail to think As should be clear by this point, having a list of genes is not enough
carefully about these issues. In one example, researchers reported the to make predictions about patient outcomes. For that purpose, you
discovery of peptide patterns with nearly 100% sensitivity and specificity must know exactly how to measure each gene, how to combine those
to detect prostate cancer.80 The problem was that the cancer specimens measurements to produce a numeric score, and how to interpret that
came from a group of men with a mean age of 67 years, whereas the score in a clinical context. Only then do you have a fully specified,
control subjects came from a group composed of 58% women, with locked-down computational model. The good news, however, is that
a mean age of 35 years.81 for statistical purposes the fully specified model can be considered
to be just one thing. Omics-based predictive signatures are a form
Test Validation Phase of complex biomarker. As a result, the statistical designs for clinical
trials that will evaluate one such signature are exactly the same as the
The goal of the test validation phase is to take the locked-down predic- designs for evaluating a single biomarker (such as the mutation status
tive model and turn it into a usable clinical assay. During this phase, of the EGFR gene). The sample size computations are, in principle,
researchers should: identical.
• Use a CLIA-certified laboratory
• Design, optimize, validate, and implement the test using current Clustering Is Not Prediction
laboratory standards
Clustering algorithms, especially in the form of two-way clustered,
• If multiple laboratories will perform the test, ensure that all labo-
colored heat maps, have been ubiquitous in the microarray literature
ratories meet the analytic validation and CLIA requirements
since its inception.83,84 Just as a list of genes (a signature) is not enough
We have touched briefly on the need to standardize the assay in to make predictions, and neither is clustering. There are certainly
our discussion of challenges. The current era of rapidly changing scientific questions that can be appropriately answered by applying a
Biostatistics and Bioinformatics in Clinical Trials  •  CHAPTER 17 293

clustering algorithm; the canonic example involves identifying natural and tumor (such as histopathology and cytopathology), genetic variants
biologic subtypes within a larger class of cancer samples. It is, however, and somatic mutations present in malignant cells, or predisposition
possible to identify subtypes with different response profiles. The most to mutation based on identified by simple sequence repeats DNA.89a
likely way to find subtypes related to outcome is to start by performing Although the evolution of informatics technology has the potential
feature-by-feature statistical tests that identify individual predictors to generate massive databases for quantitative-based interrogations of
of outcome. After selection of the best predictors, the resulting list of many informatics sources, the success of precision medicine will depend
genes (signature!) can be used to cluster the samples. A statistical test on the extent to which these rich databases may be used to advance
can then be performed to determine if the resulting “cluster member- understanding of the disease profiles and ultimately integrated for
ship” variable is a good predictor of outcome. treatment selection, necessitating robust quantitative methodology
The literature distinguishes between supervised and unsupervised for dimension reduction and appropriate trial design. Specifically, the
analyses. Supervised methods use the outcome during the analysis; effectiveness with which a treatment’s usefulness in a given type of
the simplest example is a t-test to identify genes that are differentially tumor or patient may be estimated from data inherently depends on
expressed between responders and nonresponders. By contrast, clustering the effectiveness with which one may characterize the relative contribu-
is the classic example of an unsupervised approach, because it uses the tions of determinants that are predictive of treatment effectiveness
gene expression measurements only during clustering. However, the versus those features that are prognostic of the disease extent.90 Consider-
procedure described in the previous paragraph is not unsupervised; ing the oncology context, predictive features could characterize correlates
the process of selecting the best predictive features is a supervised of the particular mechanisms by which the tumor evades apoptosis,
(and essential) part of the procedure. and prognostic features determine the likelihood of response with an
Two questions illustrate why this clustering procedure, by itself, is established standard-of-care strategy.
inadequate for prediction or classification. First, what happens when Although several statistical methods have been proposed to address
you get the data for one new patient? Can you tell to which cluster challenges arising with the high-dimensionality of omics-type data,
he or she belongs? Second, how do you validate the clustering results these methods have largely emphasized the manner in which associations
on an independent data set? In both cases, clustering is unable to may be identified with clinical outcomes when genomic features have
tell us how to react to new data. Again, this problem has a simple been acquired from a relatively limited number of patients. These
solution. The clustering results must be supplemented by developing methods, although useful for identifying associations deriving from
a completely specified, locked-down predictive model that assigns prognostic biomarkers, are often limited for discovering predictive
new samples to the existing clusters. Not surprisingly, this model biomarkers that interact with treatment and fail to elucidate the predic-
will probably use the same features that were selected to perform tive power of subpopulations. Ma and colleagues91 conducted a survey
clustering. After this model is constructed, the predictions can be of statistical methods for establishing personalized treatment selection
validated on new data sets and used in prospective clinical trials to rules that are currently available in the statistical literature. Bayesian
classify patients one at a time. approaches that predict the usefulness of personalized treatment for
a given individual’s tumor on the basis of measures of interpatient
PRECISION MEDICINE molecular similarity92,93 have been established.
The potential benefits of such biomarker-guided therapy are
Precision medicine endeavors to understand the mechanisms of seemingly substantial, but discovery of prognostic and predictive
pharmacogenetics and ultimately to achieve conformality between markers from signal in the presence of many noise covariates remains
therapeutic interventions and the individuals being treated. With a challenge.94 Several aspects of the feature inputs, such as process or
advancements in the understanding of molecular cancer biology, several interobserver reproducibility, need to be considered carefully before
treatment strategies that target specific biomarkers, such as crizotinib the proposed methods are used for personalized selection with large-scale
for non–small cell lung cancer (NSCLC) characterized by an anaplastic genomic, imaging, and clinical data. A checklist of criteria has been
lymphoma kinase (ALK) gene rearrangement, have been discovered developed by the US National Cancer Institute to address issues of
and translated into clinical practice.85 Early successes of molecularly specimens, assays, and clinical trial design.95
targeted interventions have to some extent effectuated precision
medicine. For example, antiestrogen agents, such as tamoxifen, are Biomarker-Driven Adaptive Clinical Trials and
recommended for patients with breast tumors that harbor receptors Case Studies
for estrogen.86 Melanomas with BRAF V600E mutations have been
shown to benefit from vemurafenib.87 In addition, therapy may be Precision medicine challenges the traditional paradigms of clinical
guided by signatures that comprise multiple markers, such as the translation, for which estimates of population-averaged effects from
Oncotype DX recurrence score (RS). Based on 21 genes, Oncotype large randomized trials are used as the basis for demonstrating improve-
DX is used to predict recurrence and response to adjuvant chemotherapy ments in comparative effectiveness. Moreover, successes for targeted
for estrogen receptor–positive (ER+) stage I or II invasive breast cancer.88 therapeutic strategies have been limited. For example, from 2003 to
Implicit to the concept of precision medicine is heterogeneity of 2011, 71.7% of investigational agents failed in phase II trials, and
treatment benefit among patients and patient populations. Its successful only 10.5% attained final market approval from the FDA.96 The low
implementation relies on our understanding of distinct molecular success rates may be attributable to reliance on inadequate trial designs
profiles and their biomarkers, which can be used as targets to devise for characterizing treatment benefit and on analytic approaches that
treatment strategies that exploit current understanding of the biologic are inadequate for characterizing selection rules based on the joint
mechanisms of the disease. The concept of personalizing clinical care, effects of multiple candidate biomarkers or environmental exposures.97
although a topic of recent emphasis, is not entirely new. Evaluations Despite innovations in trial design for precision medicines, statistical
of individual-level characteristics have long been used to inform methods for validating predictive biomarkers remain less established
treatment selection.89 Advances in informatics technologies, however, than for prognostic biomarkers.98 Strategies such as enrichment, all-
have provided access to enormous databases yielding inputs that facilitate comers, and hybrid designs99,100 have been proposed for predictive
interrogation of therapeutic options in the presence of diverse types biomarker validation. Moreover, current validation designs predomi-
of molecular and clinical information. nately consider validation strategies for binary markers arising from
In oncology settings, biomarkers arising from diverse sources of a single source. Most validation approaches rely on linear models that
patient-level informatics are being interrogated for their utility to use interaction terms to characterize the partial effects of receipt of a
inform treatment decisions and treatment monitoring. Biomarkers type of targeted therapy given observed values of candidate predictive
used in cancer may derive from clinical characteristics of the patient biomarkers.101,102
294 Part I: Science and Clinical Oncology

Examining interactions in ongoing clinical trials has many advan- but they help improve the design’s performance. Bayesian modeling
tages.12 One is that requiring biomarker information for randomization appropriately accounts for the uncertainty involved in this prediction
means that information will be available for all patients, thus avoiding process.
the “data missingness” problem that plagues retrospective biomarker
studies.103 Another advantage is that when patients in a definable Case Studies
biomarker subset are not benefiting from a particular therapy, that
subset can be dropped from the trial. In a multiarmed trial, biomarker Oncotype DX
subtypes that do not respond to a particular treatment can be excluded The series of studies performed by Genomic Health to establish its
from that treatment arm, possibly gradually, through use of adaptive Oncotype DX assay followed most of the procedures recommended
randomization.11 A consequence of excluding nonresponders is that by the IOM committee. First, the researchers asked a well-defined
focusing on responders means trials can become smaller. And of course, clinical question about a well-defined patient population. Their goal
excluding patients who do not benefit reduces the extent of overtreat- was to predict the risk of distant recurrence after tamoxifen treatment
ment of trial participants. of women with node-negative, ER+ breast cancer. Second, they used
An adaptive biomarker-driven clinical trial can begin by including four existing microarray data sets from the published literature to
all the patients who meet the enrollment criteria for the trial, but select 250 candidate genes. Third, they developed a new assay using
then can continue by restricting enrollment to individuals with qPCR to measure the expression levels of those 250 genes in tumor
biomarker profiles that match those of the responding population as samples. Fourth, they validated the assay by obtaining data from tumor
the results accumulate over the course of the trial. Biomarker subsets samples that had been collected in three independent clinical trials
can be identified in advance, generated on the basis of the data in the of breast cancer, involving a total of 447 women. They performed
trial, or some combination of the two can be used by incorporating univariate analyses and selected 23 genes that were associated with
historical data via the prior distribution, as previously described. Any the risk of recurrence in at least two of three qPCR data sets. They
approach is subject to multiplicities.4 Basing an assessment on the then constructed multivariate predictive models and further reduced
responding subsets is particularly prone to false-positive conclusions the set of predictors to a panel of 16 cancer-related genes and five
and requires a level of within-trial empiric validation. The extent of reference genes. Their approach was simple but elegant, reducing a
validation is a design characteristic that can be determined prospectively. multidimensional problem into a single number, an RS, which made
False-positive rates and statistical power can be evaluated and controlled, for a straightforward validation process. The algorithm included cutoffs
usually requiring simulations. Comprehensive overviews of recent to separate patients into low-, intermediate-, and high-risk categories
advances in designs used in oncology for studying many agent-and-target and was completely specified during this step. Fifth, they tested the
combinations in parallel, such as platform trials, basket trials, and prospectively defined qPCR assay and RS algorithm for their ability
umbrella trials, can be found.104,105 to predict recurrence in a retrospective set of 668 samples from the
An example of a complicated biomarker-driven, adaptively ran- National Surgical Adjuvant Breast and Bowel Project (NSABP) B14
domized clinical trial is I-SPY212,106,107 (http://clinicaltrials.gov/show/ trial, for which paraffin blocks were available.109 Another study showed
NCT01042379) (http://www.ispy2.org/). The setting is neoadjuvant that RS did not predict recurrence in women with node-negative
treatment for breast cancer in which the end point is pathologic complete breast cancer (regardless of ER status) who had received no adjuvant
response (pathCR), which the FDA has recently determined to be a reg- chemotherapy, which suggests that the signature depends on either
istration end point in high-risk early breast cancer (http://www.fda.gov/ the ER positivity or on the treatment.110 Although the RS was built
downloads/Drugs/GuidanceComplianceRegulatoryInformation/ to be prognostic, samples from the NSABP B20 trial showed that
Guidances/UCM305501.pdf ). Investigational drugs are added to a it helped predict response to chemotherapy for women with node-
taxane in the initial cycles of standard therapy. Similar trials are being negative, ER+ breast cancer, with no apparent benefit for women with
explored in other diseases, including lymphoma.108 low RSs.111
The goal of I-SPY2 is to efficiently pair drugs and combinations
with the patients’ biomarker profiles that characterize the disease subset BATTLE Trial
that responds to that treatment. I-SPY2 is essentially a screening process. The Biomarker-Integrated Approaches of Targeted Therapy for Lung
Through use of bayesian updating, assignment to therapy is adaptively Cancer Elimination (BATTLE) trial was a prospective phase II,
randomized based on the patient’s tumor biomarkers and the drug’s biomarker-based, adaptively randomized study in 255 patients who
performance in patients with similar biomarker profiles. A consequence had been pretreated for NSCLC.112,113 The trial consisted of four
of adaptive randomization is that better-performing drugs move through treatment arms of targeted therapies, each of which was associated
the process faster. Drugs are graduated when they have a sufficiently with a prespecified set of biomarkers that were anticipated to predict
high (bayesian) predictive probability of success in a small, focused the efficacy of the therapy: erlotinib (EGFR), sorafenib (KRAS/BRAF),
phase III trial involving patients who are most likely to benefit from vandetanib (VEGFR2), and bexarotene plus erlotinib (RXR/CCND1).
the drug based on their biomarker profiles (or the molecular signature The primary end point of the trial was 8-week disease control. After
of their disease subtype). an initial equal randomization period, patients were adaptively random-
The biomarker categories for I-SPY2 are set in advance and apply ized to one of the treatment arms, based on the molecular biomarkers
to all therapies. The design considers 10 biomarker profiles, or molecular analyzed in fresh core needle biopsy specimens. Overall results included
signatures, that make both marketing and biologic sense. The categories a 46% 8-week disease control rate, median progression-free survival
and signatures are fixed throughout, and any new therapy that is of 1.9 months (95% confidence interval [CI], 1.8–2.4), and median
inserted into the trial conforms to the predefined set of biomarker overall survival of 8.8 months (95% CI, 6.3–10.6). The results
signatures. In a trial similar to I-SPY, the tumor categories could be confirmed that EGFR mutations predicted better 8-week disease control
determined by the drugs’ targets. with sorafenib.
Even though the I-SPY2 end point is ascertained relatively
quickly at surgery, waiting 6 months to learn whether a drug is CONCLUSIONS
benefiting a particular patient can be a long time. Therefore, mag-
netic resonance imaging was incorporated to assess tumor volume Innovations in cancer research are developing rapidly. The quantitative
after the first cycle of therapy and after the 12 weeks of taxane or aspects of this research, from assessing individual genes and biomarkers
investigational drug cycles. Modeling the longitudinal relationship to evaluating pathways and systems biology, are becoming both more
between pathCR and tumor volume reduction enables prediction of important and more difficult. Assessing which patients benefit from
whether the patient will have a pathCR. Predictions are not perfect, which therapies is the holy grail of cancer research, culminating in
Biostatistics and Bioinformatics in Clinical Trials  •  CHAPTER 17 295

clinical trials to demonstrate benefit, or lack thereof. A single adaptive will be. Our aim has been to describe some ways in which the scientific
clinical trial can be designed to discover responding subsets and validate winds are blowing, preparing the reader to understand and appreciate
the discovery. However, despite the great strides seen in modern cancer the path being followed.
biology, strides of much greater magnitude are yet to come. Advances
in biology lead to ever more and ever smaller subsets. Large clinical
trials will soon be impossible, and traditional statistical approaches The complete reference list is available online at
will have to be replaced. We cannot predict what those replacements ExpertConsult.com.

KEY REFERENCES
4. Berry DA. Multiplicities in cancer research: ubiqui- 49. Leek JT, Scharpf RB, Bravo HC, et al. Tackling 86. Simon R. Advances in clinical trial designs for
tous and necessary evils. J Natl Cancer Inst. 2012;104: the widespread and critical impact of batch effects predictive biomarker discovery and validation.
1125–1133. in high-throughput data. Nat Rev Genet. 2010;11: Current Breast Cancer Reports. 2009;1(4):216–221.
7. Berry DA. Statistical innovations in cancer research. 733–739. 87. Sosman JA, Kim KB, Schuchter L, Gonzalez R,
In: Holland J, Frei T, et al, eds. Cancer Medicine. 50. Petricoin EF, Ardekani AM, Hitt BA, et al. Use Pavlick AC, Weber JS, et al. Survival in BRAF
8th ed. London: BC Decker; 2009. of proteomic patterns in serum to identify ovarian V600-mutant advanced melanoma treated with
8. Berry DA. Bayesian approaches for comparative cancer. Lancet. 2002;359:572–577. vemurafenib. N Engl J Med. 2012;366(8):707–714.
effectiveness research. Clin Trials. 2012;9:37–47. 52. Baggerly KA, Edmonson SR, Morris JS, Coombes 90. Simon R. Clinical trial designs for evaluating the
10. Biswas S, Liu DD, Lee JJ, Berry DA. Bayesian clinical KR. High-resolution serum proteomic patterns medical utility of prognostic and predictive bio­
trials at the University of Texas MD Anderson Cancer for ovarian cancer detection. Endocr Relat Cancer. markers in oncology. Per Med. 2010;7(1):33–47.
Center. Clin Trials. 2009;6:205–216. 2004;11:583–584, author reply 585–7. 91. Ma J, Hobbs BP, Stingo FC. Statistical methods for
12. Berry DA. Adaptive clinical trials in oncology. Nat 53. Baggerly KA, Morris JS, Coombes KR. Reproduc- establishing personalized treatment rules in oncology.
Rev Clin Oncol. 2011;9:199–207. ibility of SELDI-TOF protein patterns in serum: Biomed Res Int. 2015;2015:670691.
13. Nass SJ, Moses HL, Mendelsohn J. Institute of comparing datasets from different experiments. 92. Ma J, Stingo FC, Hobbs BP. Bayesian predictive
Medicine: a national cancer clinical trials system for Bioinformatics. 2004;20:777–785. modeling for genomic based personalized treatment
the 21st century: reinvigorating the NCI Coopera- 54. Luo J, Schumacher M, Scherer A, et al. A comparison selection. Biometrics. 2016;72(2):575–583.
tive Group Program. Washington, DC: National of batch effect removal methods for enhancement of 93. Ma J, Hobbs BP, Stingo FC. Integrating genomic
Academies Press; 2010. prediction performance using MAQC-II microar- signatures for treatment selection with Bayesian
14. Wong KM, Capasso A, Eckhardt SG. The changing ray gene expression data. Pharmacogenomics J. predictive failure time models. Stat Methods Med
landscape of phase I trials in oncology. Nat Rev Clin 2010;10:278–291. Res. 2016;[Epub ahead of print].
Oncol. 2016;13(2):106–117. 55. Chen C, Grennan K, Badner J, et al. Removing batch 95. McShane LM, Cavenagh MM, Lively TG, Eberhard
15. Ji Y, Wang SJ. Modified toxicity probability interval effects in analysis of expression microarray data: an DA, Bigbee WL, Williams PM, et al. Criteria for
design: a safer and more reliable method than the 3 evaluation of six batch adjustment methods. PLoS the use of omics-based predictors in clinical trials.
+ 3 design for practical phase I trials. J Clin Oncol. ONE. 2011;6(2):e17238. Nature. 2013;502(7471):317–320.
2013;31(14):1785–1791. 57. McCall MN, Bolstad BM, Irizarry RA. Frozen 96. Hay M, Thomas DW, Craighead JL, Economides C,
16. Thall PF, Nguyen HQ, Braun TM, Qazilbash MH. robust multiarray analysis (fRMA). Biostatistics. Rosenthal J. Clinical development success rates for
Using joint utilities of the times to response and 2010;11:242–253. investigational drugs. Nat Biotechnol. 2014;32(1):
toxicity to adaptively optimize schedule-dose regimes. 60. Efron B, Tibshirani R. Empirical Bayes methods and 40–51.
Biometrics. 2013;69(3):673–682. false discovery rates for microarrays. Genet Epidemiol. 100. Mandrekar SJ, Sargent DJ. Predictive biomarker
18. Hobbs BP, Chen N, Lee JJ. Controlled multi-arm 2002;23:70–86. validation in practice: lessons from real trials. Clinical
platform design using predictive probability. Stat 62. Storey JD, Tibshirani R. Statistical significance trials. 2010.
Methods Med Res. 2018;27(1):65–78. for genomewide studies. Proc Natl Acad Sci USA. 101. Mandrekar SJ, Sargent DJ. Design of clinical trials for
20. Hobbs BP, Thall PF, Lin SH. Bayesian group 2003;100:9440–9445. biomarker research in oncology. Clinical investigation.
sequential clinical trial design using total toxicity 63. Pounds S, Cheng C. Improving false discovery rate 2011;1(12):1627–1636.
burden and progression-free survival. J R Stat Soc estimation. Bioinformatics. 2004;20:1737–1745. 107. Berry DA, Herbst RS, Rubin EH. Design strategies for
Ser C Appl Stat. 2016;65(2):273–297. 64. Qian HR, Huang S. Comparison of false discovery personalized therapy trials. Clin Cancer Res. 2012;18:
24. Kidwell KM. SMART designs in cancer research: past, rate methods in identifying genes with differential 638–644.
present, and future. Clin Trials. 2014;11(4):445–456. expression. Genomics. 2005;86:495–503. 108. Younes A, Berry DA. From drug discovery to
32. Cancer Genome Atlas Research Network. Com- 65. Baggerly KA, Coombes KR. Deriving chemosen- biomarker-driven clinical trials in lymphoma. Nat
prehensive genomic characterization defines human sitivity from cell lines: forensic bioinformatics and Rev Clin Oncol. 2012;9:643–653.
glioblastoma genes and core pathways. Nature. reproducible research in high-throughput biology. 109. Paik S, Shak S, Tang G, et al. A multigene assay to
2008;455:1061–1068. Ann Appl Stat. 2009;3:1309–1334. predict recurrence of tamoxifen-treated, node-negative
33. Chin L, Andersen JN, Futreal PA. Cancer genomics: 67. Pepe MS, Etzioni R, Feng Z, et al. Phases of bio- breast cancer. N Engl J Med. 2004;351:2817–2826.
from discovery science to personalized medicine. marker development for early detection of cancer. 110. Esteva FJ, Sahin AA, Cristofanilli M, et al. Prognostic
Nat Med. 2011;17:297–303. J Natl Cancer Inst. 2001;93:1054–1061. role of a multigene reverse transcriptase-PCR assay
39. Calin GA, Dumitru CD, Shimizu M, et al. Frequent 74. Baggerly K. Disclose all data in publications. Nature. in patients with node-negative breast cancer not
deletions and down-regulation of micro-RNA 2010;467:401. receiving adjuvant systemic therapy. Clin Cancer
genes miR15 and miR16 at 13q14 in chronic 75. Baggerly KA, Coombes KR. What information Res. 2005;11:3315–3319.
lymphocytic leukemia. Proc Natl Acad Sci USA. should be required to support clinical “omics” 111. Paik S, Tang G, Shak S, et al. Gene expression and
2002;99:15524–15529. publications? Clin Chem. 2011;57:688–690. benefit of chemotherapy in women with node-
42. Kozomara A, Griffiths-Jones S. miRBase: integrating 79. Peng RD. Reproducible research in computational negative, estrogen receptor-positive breast cancer.
microRNA annotation and deep-sequencing data. science. Science. 2011;334:1226–1227. J Clin Oncol. 2006;24:3726–3734.
Nucleic Acids Res. 2011;39:D152–D157. 82. Simon RM, Paik S, Hayes DF. Use of archived 113. Kim ES, Herbst RS, Wistuba II, et al. The BATTLE
48. Reich M, Liefeld T, Gould J, et al. GenePattern specimens in evaluation of prognostic and predictive trial: personalizing therapy for lung cancer. Cancer
2.0. Nat Genet. 2006;38:500–501. biomarkers. J Natl Cancer Inst. 2009;101:1446–1452. Discov. 2011;1:44–53.
Biostatistics and Bioinformatics in Clinical Trials  •  CHAPTER 17 295.e1
295.e1

REFERENCES
1. Bernoulli J. The Art of Conjecturing, Together with 25. Fodor SP, Rava RP, Huang XC, et al. Multiplexed 46. Gentleman RC, Carey VJ, Bates DM, et al.
Letter to a Friend on Sets in Court Tennis (English biochemical assays with biological chips. Nature. Bioconductor: open software development for
translation of the 1713 text by Edith Sylla). Baltimore: 1993;364:555–556. computational biology and bioinformatics. Genome
Johns Hopkins University Press; 2005. 26. Schena M, Shalon D, Davis RW, Brown PO. Biol. 2004;5:R80.
2. Stigler SM. Gauss and the invention of least squares. Quantitative monitoring of gene expression patterns 47. R Core Team. R: A Language and Environment for
Ann Statist. 1981;9:465–474. with a complementary DNA microarray. Science. Statistical Computing. Vienna: R Foundation for
3. Bernoulli D. Exposition of a new theory on 1995;270:467–470. Statistical Computing; 2012.
the measurement of risk [1738]. Econometrica. 27. DeRisi J, Penland L, Brown PO, et al. Use of a 48. Reich M, Liefeld T, Gould J, et al. GenePattern
1954;22:23–36. https://engineering.purdue.edu/ cDNA microarray to analyse gene expression patterns 2.0. Nat Genet. 2006;38:500–501.
~ipollak/ece302/SPRING12/notes/Bernoulli_1738 in human cancer. Nat Genet. 1996;14:457–460. 49. Leek JT, Scharpf RB, Bravo HC, et al. Tackling
.pdf/. Accessed March 15, 2013. 2009. 28. Khan J, Simon R, Bittner M, et al. Gene expression the widespread and critical impact of batch effects
4. Berry DA. Multiplicities in cancer research: profiling of alveolar rhabdomyosarcoma with cDNA in high-throughput data. Nat Rev Genet. 2010;11:
ubiquitous and necessary evils. J Natl Cancer Inst. microarrays. Cancer Res. 1998;58:5009–5013. 733–739.
2012;104:1125–1133. 29. Alon U, Barkai N, Notterman DA, et al. Broad 50. Petricoin EF, Ardekani AM, Hitt BA, et al. Use
5. Berger JO, Berry DA. Statistical analysis and the patterns of gene expression revealed by clustering of proteomic patterns in serum to identify ovarian
illusion of objectivity. Am Sci. 1988;76:159–165. analysis of tumor and normal colon tissues probed cancer. Lancet. 2002;359:572–577.
6. Berry DA, Stangl DK. Bayesian Biostatistics. New by oligonucleotide arrays. Proc Natl Acad Sci USA. 51. Sorace JM, Zhan M. A data review and re-assessment
York: Marcel Dekker; 1996. 1999;96:6745–6750. of ovarian cancer serum proteomic profiling. BMC
7. Berry DA. Statistical innovations in cancer research. 30. Golub TR, Slonim DK, Tamayo P, et al. Molecular Bioinformatics. 2003;4:24.
In: Holland J, Frei T, et al, eds. Cancer Medicine. classification of cancer: class discovery and class 52. Baggerly KA, Edmonson SR, Morris JS, Coombes
8th ed. London: BC Decker; 2009. prediction by gene expression monitoring. Science. KR. High-resolution serum proteomic patterns
8. Berry DA. Bayesian approaches for comparative 1999;286:531–537. for ovarian cancer detection. Endocr Relat Cancer.
effectiveness research. Clin Trials. 2012;9:37–47. 31. Perou CM, Jeffrey SS, van de Rijn M, et al. Distinc- 2004;11:583–584, author reply 585–7.
9. Yoshioka A. Use of randomisation in the Medical tive gene expression patterns in human mammary 53. Baggerly KA, Morris JS, Coombes KR. Reproduc-
Research Council’s clinical trial of streptomycin epithelial cells and breast cancers. Proc Natl Acad ibility of SELDI-TOF protein patterns in serum:
in pulmonary tuberculosis in the 1940s. BMJ. Sci USA. 1999;96:9212–9217. comparing datasets from different experiments.
1998;317:1220–1223. 32. Cancer Genome Atlas Research Network. Com- Bioinformatics. 2004;20:777–785.
10. Biswas S, Liu DD, Lee JJ, Berry DA. Bayesian clinical prehensive genomic characterization defines human 54. Luo J, Schumacher M, Scherer A, et al. A comparison
trials at the University of Texas MD Anderson Cancer glioblastoma genes and core pathways. Nature. of batch effect removal methods for enhancement of
Center. Clin Trials. 2009;6:205–216. 2008;455:1061–1068. prediction performance using MAQC-II microarray
11. Berry DA. Adaptive clinical trials: the promise and 33. Chin L, Andersen JN, Futreal PA. Cancer genomics: gene expression data. Pharmacogenomics J. 2010;
the caution. J Clin Oncol. 2011;29:606–609. from discovery science to personalized medicine. 10:278–291.
12. Berry DA. Adaptive clinical trials in oncology. Nat Nat Med. 2011;17:297–303. 55. Chen C, Grennan K, Badner J, et al. Removing batch
Rev Clin Oncol. 2011;9:199–207. 34. Chin L, Hahn WC, Getz G, Meyerson M. Making effects in analysis of expression microarray data: an
13. Nass SJ, Moses HL, Mendelsohn J. Institute of sense of cancer genomic data. Genes Dev. 2011;25: evaluation of six batch adjustment methods. PLoS
Medicine: a national cancer clinical trials system for the 534–555. ONE. 2011;6(2):e17238.
21st century: reinvigorating the NCI Cooperative Group 35. Lee RC, Feinbaum RL, Ambros V. The C. elegans 56. Lazar C, Meganck S, Taminau J, et al. Batch effect
Program. Washington, DC: National Academies heterochronic gene lin-4 encodes small RNAs removal methods for microarray gene expression data
Press; 2010. with antisense complementarity to lin-14. Cell. integration: a survey. Brief Bioinform. 2012;Jul 31
14. Wong KM, Capasso A, Eckhardt SG. The changing 1993;75:843–854. [Epub ahead of print].
landscape of phase I trials in oncology. Nat Rev Clin 36. Lagos-Quintana M, Rauhut R, Lendeckel W, Tuschl 57. McCall MN, Bolstad BM, Irizarry RA. Frozen
Oncol. 2016;13(2):106–117. T. Identification of novel genes coding for small robust multiarray analysis (fRMA). Biostatistics.
15. Ji Y, Wang SJ. Modified toxicity probability interval expressed RNAs. Science. 2001;294:853–858. 2010;11:242–253.
design: a safer and more reliable method than the 3 37. Lau NC, Lim LP, Weinstein EG, Bartel DP. 58. Livak KJ, Schmittgen TD. Analysis of relative gene
+ 3 design for practical phase I trials. J Clin Oncol. An abundant class of tiny RNAs with probable expression data using real-time quantitative PCR
2013;31(14):1785–1791. regulatory roles in Caenorhabditis elegans. Science. and the 2(-Delta Delta C(T). Method. Methods.
16. Thall PF, Nguyen HQ, Braun TM, Qazilbash MH. 2001;294:858–862. 2001;25:402–408.
Using joint utilities of the times to response and 38. Lee RC, Ambros V. An extensive class of small 59. Benjamini Y, Hochberg Y. Controlling the false
toxicity to adaptively optimize schedule-dose regimes. RNAs in Caenorhabditis elegans. Science. 2001;294: discovery rate: a practical and powerful approach
Biometrics. 2013;69(3):673–682. 862–864. to multiple testing. J R Stat Soc Series B Stat Methodol.
17. Zhang J, Braun TM. A phase I Bayesian adaptive 39. Calin GA, Dumitru CD, Shimizu M, et al. Frequent 1995;57:289–300.
design to simultaneously optimize dose and schedule deletions and down-regulation of micro-RNA 60. Efron B, Tibshirani R. Empirical Bayes methods and
assignments both between and within patients. genes miR15 and miR16 at 13q14 in chronic false discovery rates for microarrays. Genet Epidemiol.
J American Statistical Assoc. 2013;108:892–901. lymphocytic leukemia. Proc Natl Acad Sci USA. 2002;23:70–86.
18. Hobbs BP, Chen N, Lee JJ. Controlled multi-arm 2002;99:15524–15529. 61. Pounds S, Morris SW. Estimating the occurrence
platform design using predictive probability. Stat 40. Griffiths-Jones S, Grocock RJ, van Dongen S, of false positives and false negatives in microarray
Methods Med Res. 2018;27(1):65–78. et al. miRBase: microRNA sequences, targets and studies by approximating and partitioning the
19. Bekele BN, Thall PF. Dose-finding based on multiple gene nomenclature. Nucleic Acids Res. 2006;34: empirical distribution of p-values. Bioinformatics.
toxicities in a soft tissue sarcoma trial. J. Am.Statist. D140–D144. 2003;19:1236–1242.
Ass. 2004;99:26–34. 41. Griffiths-Jones S, Saini HK, van Dongen S, Enright 62. Storey JD, Tibshirani R. Statistical significance
20. Hobbs BP, Thall PF, Lin SH. Bayesian group AJ. miRBase: tools for microRNA genomics. Nucleic for genomewide studies. Proc Natl Acad Sci USA.
sequential clinical trial design using total toxicity Acids Res. 2008;36:D154–D158. 2003;100:9440–9445.
burden and progression-free survival. J R Stat Soc 42. Kozomara A, Griffiths-Jones S. miRBase: integrating 63. Pounds S, Cheng C. Improving false discovery rate
Ser C Appl Stat. 2016;65(2):273–297. microRNA annotation and deep-sequencing data. estimation. Bioinformatics. 2004;20:1737–1745.
21. Thall PF, Cook JD. Dose-finding based on efficacy- Nucleic Acids Res. 2011;39:D152–D157. 64. Qian HR, Huang S. Comparison of false discovery
toxicity trade-offs. Biometrics. 2004;60(3):684–693. 43. Li C, Wong WH. Model-based analysis of oligo- rate methods in identifying genes with differential
22. Zhang W, Sargent DJ, Mandrekar S. An adaptive nucleotide arrays: expression index computation expression. Genomics. 2005;86:495–503.
dose-finding design incorporating both toxicity and and outlier detection. Proc Natl Acad Sci U SA. 65. Baggerly KA, Coombes KR. Deriving chemosen-
efficacy. Stat Med. 2006;25(14):2365–2383. 2001;98:31–36. sitivity from cell lines: forensic bioinformatics and
23. Wang L, Rotnitzky A, Lin X, Millikan RE, Thall 44. Bolstad BM, Irizarry RA, Astrand M, Speed TP. reproducible research in high-throughput biology.
PF. Evaluation of viable dynamic treatment regimes A comparison of normalization methods for high Ann Appl Stat. 2009;3:1309–1334.
in a sequentially randomized trial of advanced density oligonucleotide array data based on variance 66. Committee on the Review of Omics-Based Tests for
prostate cancer. J Am Stat Assoc. 2012;107(498): and bias. Bioinformatics. 2003;19:185–193. Predicting Patient Outcomes in Clinical Trials. In:
493–508. 45. Irizarry RA, Hobbs B, Collin F, et al. Exploration, Christine MM, Sharly JN, Gilbert SO, eds. Evolution
24. Kidwell KM. SMART designs in cancer research: normalization, and summaries of high density of Translational Omics: Lessons Learned and the Path
past, present, and future. Clin Trials. 2014;11(4): oligonucleotide array probe level data. Biostatistics. Forward. Washington, DC: The National Academies
445–456. 2003;4:249–264. Press; 2012.
295.e2 Part I: Science and Clinical Oncology

67. Pepe MS, Etzioni R, Feng Z, et al. Phases of bio- 84. Eisen MB, Spellman PT, Brown PO, Botstein D. treatment decisions. Ann Intern Med. 2011;154(4):
marker development for early detection of cancer. Cluster analysis and display of genome-wide expres- 253–259.
J Natl Cancer Inst. 2001;93:1054–1061. sion patterns. Proc Natl Acad Sci USA. 1998;95: 99. Mandrekar SJ, Sargent DJ. Clinical trial designs for
68. Brazma A, Hingamp P, Quackenbush J, et al. 14863–14868. predictive biomarker validation: one size does not
Minimum information about a microarray experi- 85. Kelloff GJ, Sigman CC. Cancer biomarkers: selecting fit all. J Biopharm Stat. 2009;19(3):530–542.
ment (MIAME)-toward standards for microarray the right drug for the right patient. Nat Rev Drug 100. Mandrekar SJ, Sargent DJ. Predictive biomarker
data. Nat Genet. 2001;29:365–371. Discov. 2012;11(3):201–214. validation in practice: lessons from real trials. Clinical
69. Orchard S, Hermjakob H, Taylor CF, et al. Further 86. Simon R. Advances in clinical trial designs for trials. 2010.
steps in standardisation. Report of the second annual predictive biomarker discovery and validation. 101. Mandrekar SJ, Sargent DJ. Design of clinical trials for
Proteomics Standards Initiative Spring Workshop Current Breast Cancer Reports. 2009;1(4):216–221. biomarker research in oncology. Clinical investigation.
(Siena, Italy 17–20th April 2005). Proteomics. 2005;5: 87. Sosman JA, Kim KB, Schuchter L, Gonzalez R, 2011;1(12):1627–1636.
3552–3555. Pavlick AC, Weber JS, et al. Survival in BRAF 102. Chen JJ, Lu TP, Chen DT, Wang SJ. Biomarker
70. McShane LM, Altman DG, Sauerbrei W, et al. V600-mutant advanced melanoma treated with adaptive designs in clinical trials. Translational Cancer
Statistics Subcommittee of the NCIEWGoCD. vemurafenib. N Engl J Med. 2012;366(8):707–714. Research. 2014;3(3):279–292.
Reporting recommendations for tumor marker 88. Albain KS, Barlow WE, Shak S, Hortobagyi GN, 103. Rimm DL, Nielsen TO, Jewell SD, et al. Cancer and
prognostic studies (REMARK). J Natl Cancer Inst. Livingston RB, Yeh IT, et al. Prognostic and Leukemia Group B Pathology Committee guidelines
2005;97:1180–1184. predictive value of the 21-gene recurrence score for tissue microarray construction representing
71. Altman DG, McShane LM, Sauerbrei W, Taube assay in postmenopausal women with node-positive, multicenter prospective clinical trial tissues. J Clin
SE. Reporting Recommendations for Tumor Marker oestrogen-receptor-positive breast cancer on chemo- Oncol. 2011;29:2282–2290.
Prognostic Studies (REMARK): explanation and therapy: a retrospective analysis of a randomised 104. Renfro L, Sargent D. Statistical controversies in
elaboration. PLoS Med. 2012;9:e1001216. trial. Lancet Oncol. 2010;11(1):55–65. clinical research: basket trials, umbrella trials, and
72. Moher D, Schulz KF, Altman D, Group C. The 89. Byar DP, Corle DK. Selecting optimal treatment in other master protocols: a review and examples. Ann
CONSORT statement: revised recommendations clinical trials using covariate information. J Chronic Oncol. 2016.
for improving the quality of reports of parallel-group Dis. 1977;30(7):445–459. 105. Berry DA. The brave new world of clinical cancer
randomized trials. JAMA. 2001;285:1987–1991. 89a.  Ellegren H. Microsatellites: simple sequences with research: adaptive biomarker-driven trials integrating
73. Bossuyt PM, Reitsma JB, Bruns DE, et al. Standards complex evolution. Nat Rev Genet. 2004;5(6): clinical practice with clinical research. Mol Oncol.
for Reporting of Diagnostic A. The STARD state- 435–445. 2015;9(5):951–959.
ment for reporting studies of diagnostic accuracy: 90. Simon R. Clinical trial designs for evaluating the 106. Barker AD, Sigman CC, Kelloff GJ, et al. I-SPY2:
explanation and elaboration. Clin Chem. 2003;49: medical utility of prognostic and predictive biomark- an adaptive breast cancer trial design in the setting
7–18. ers in oncology. Per Med. 2010;7(1):33–47. of neoadjuvant chemotherapy. Clin Pharmacol Ther.
74. Baggerly K. Disclose all data in publications. Nature. 91. Ma J, Hobbs BP, Stingo FC. Statistical methods for 2009;86:97–100.
2010;467:401. establishing personalized treatment rules in oncology. 107. Berry DA, Herbst RS, Rubin EH. Design strategies
75. Baggerly KA, Coombes KR. What information Biomed Res Int. 2015;2015:670691. for personalized therapy trials. Clin Cancer Res.
should be required to support clinical “omics” 92. Ma J, Stingo FC, Hobbs BP. Bayesian predictive 2012;18:638–644.
publications? Clin Chem. 2011;57:688–690. modeling for genomic based personalized treatment 108. Younes A, Berry DA. From drug discovery to
76. Buckheit J, Donoho DL. Wavelab and reproducible selection. Biometrics. 2016;72(2):575–583. biomarker-driven clinical trials in lymphoma. Nat
research. In: Antoniadis A, ed. Wavelets and Statistics. 93. Ma J, Hobbs BP, Stingo FC. Francesco C Stingo. Rev Clin Oncol. 2012;9:643–653.
New York: Springer-Verlag; 1995. Integrating genomic signatures for treatment selec- 109. Paik S, Shak S, Tang G, et al. A multigene assay
77. Leisch F, Rossini AJ. Reproducible statistical research. tion with bayesian predictive failure time models. to predict recurrence of tamoxifen-treated, node-
Chance. 2003;16:46–50. Stat Methods Med Res. 2016;[Epub ahead of print]. negative breast cancer. N Engl J Med. 2004;351:
78. Gentleman R. Reproducible research: a bioinformat- 94. Food and Drug Administration. Paving the Way 2817–2826.
ics case study. Stat Appl Genet Mol Biol. 2005;4: for Personalized Medicine: FDA’s Role in a New 110. Esteva FJ, Sahin AA, Cristofanilli M, et al. Prognostic
Article2. Era of Medical Product Development. http://www. role of a multigene reverse transcriptase-PCR assay
79. Peng RD. Reproducible research in computational fda.gov/downloads/ScienceResearch/SpecialTopics/ in patients with node-negative breast cancer not
science. Science. 2011;334:1226–1227. PersonalizedMedicine, 2013. receiving adjuvant systemic therapy. Clin Cancer
80. Villanueva J, Shaffer DR, Philip J, et al. Differential 95. McShane LM, Cavenagh MM, Lively TG, Eberhard Res. 2005;11:3315–3319.
exoprotease activities confer tumor-specific serum DA, Bigbee WL, Williams PM, et al. Criteria for 111. Paik S, Tang G, Shak S, et al. Gene expression and
peptidome patterns. J Clin Invest. 2006;116:271–284. the use of omics-based predictors in clinical trials. benefit of chemotherapy in women with node-
81. Ransohoff DF, Gourlay ML. Sources of bias in Nature. 2013;502(7471):317–320. negative, estrogen receptor-positive breast cancer.
specimens for research about molecular markers 96. Hay M, Thomas DW, Craighead JL, Economides J Clin Oncol. 2006;24:3726–3734.
for cancer. J Clin Oncol. 2010;28:698–704. C, Rosenthal J. Clinical development success 112. Zhou X, Liu S, Kim ES, et al. Bayesian adaptive
82. Simon RM, Paik S, Hayes DF. Use of archived rates for investigational drugs. Nat Biotechnol. design for targeted therapy development in lung
specimens in evaluation of prognostic and predictive 2014;32(1):40–51. cancer—a step toward personalized medicine. Clin
biomarkers. J Natl Cancer Inst. 2009;101:1446–1452. 97. Knox SS. From ‘omics’ to complex disease: a systems Trials. 2008;5:181–193.
83. Weinstein JN, Myers TG, O’Connor PM, et al. An biology approach to gene-environment interactions 113. Kim ES, Herbst RS, Wistuba II, et al. The BATTLE
information-intensive approach to the molecular in cancer. Cancer Cell Int. 2010. trial: personalizing therapy for lung cancer. Cancer
pharmacology of cancer. Science. 1997;275:343– 98. Janes H, Pepe MS, Bossuyt PM, Barlow WE. Discov. 2011;1:44–53.
349. Measuring the performance of markers for guiding
18  Clinical Trial Designs in Oncology
Edward L. Korn and Boris Freidlin

S UMMARY OF K EY P OI N T S
• Phase I trials to determine • Phase II/III trial designs, multiarm patients who benefit from an agent
recommended doses and schedules designs, and the use of master that targets tumors that express a
for further testing of new treatments protocols, which allow treatment particular molecular abnormality.
need to be designed to minimize the arms to be added to an ongoing • Simultaneous screening of patients
number of patients exposed to trial, can speed the development of for multiple, possibly rare,
unacceptable toxicity. new treatments but are not without biomarkers in order to assign them
• The use of formal interim monitoring disadvantages. to different biomarker-restricted
of accruing outcome data in phase II • It is important that the primary end subtrials is a highly efficient way to
and phase III trials is important in point of a trial be chosen so that the assess multiple biomarker-driven
order to allow trials to be stopped as results of the trial will meet its treatment hypotheses.
soon as possible because of positive clinical objectives.
or negative results, while retaining • Trial designs have been developed
the statistical validity of the trial to assess whether a biomarker can
conclusions. be used to identify a subgroup of

Clinical trials are designed to answer specific clinical questions in the specific histologic type, so that the investigators can quickly move on
development of new treatments. In order to address the study question, to testing the therapy for efficacy.
it is important for the trial protocol to describe prospectively how Although there are many possible phase I dose escalation designs,3
patients will be treated and how the resulting data will be analyzed. there are two general approaches to choosing the next dose level to
This chapter begins with a review of the traditional classification of be tested based on the DLTs seen up to that point. One approach
clinical trial designs: phase I trials (to find safe doses and schedules uses only the information on the patients being treated at the current
of new agents), phase II trials (to assess the biologic activity or to dose level (and, if available, at the dose levels immediately above and
offer a preliminary assessment of clinical efficacy), and phase III trials below it). For example, the commonly used 3 + 3 design4 treats cohorts
(to provide definitive evidence of treatment efficacy for changing of three patients and decides the next dose level (escalate one level,
clinical practice). Interim monitoring, which allows trials to be stopped de-escalate one level, or remain the same level) based on the number
early based on the accrual of positive or unpromising results, is discussed of DLTs seen at the current dose: no DLTs in three patients or one
next. Phase II/III trials, which combine the phase II and phase III DLT in six patients, escalate; one DLT in three patients, remain at
trials into one trial, can speed the development of new treatments. the same level; and two or more DLTs in three patients or two or
For all phases of clinical trials, it is important to choose the primary more DLTs in six patients, de-escalate. The recommended dose is the
end point to meet the objectives of the trial; some commonly used highest dose level at which there was one or no DLTs in six patients.
end points are discussed. The chapter ends with a discussion of trial A modification of the 3 + 3 design that is slightly more aggressive,
designs that use and evaluate biomarkers and treatments together, the “rolling six design,”5 is sometimes used when the accrual is rapid
considering both situations in which there is a single biomarker and as compared with the evaluation period. Additional modifications
associated experimental treatment and situations in which there are that accelerate the design with one or two patient cohorts initially
multiple biomarkers that can potentially help choose which among until some (possibly less than DLT) toxicity is seen have also been
many treatments would be best for the patient. The chapter offers a considered.4,6,7 These designs implicitly target a 20–25% DLT rate
brief survey of the important elements of the designs of clinical trials as maximally acceptable; designs that target different DLT rates are
to address various cancer treatment questions. More detailed expositions possible.7a
including reviews of statistical methods are given elsewhere.1,2 The other general approach to phase I dose escalation is based on
a statistical model for the probability of DLT as a function of the
PHASE I DESIGNS dose level. An example of a model-based method is the continual
reassessment method.8 Model-based escalation approaches use the DLT
The purpose of a phase I trial is to find a dose and schedule of a new data from all the current patients treated to determine the dose level
therapy to take forward for further evaluation. This is typically done for the next cohort of patients. For example, the decision to escalate
by sequentially treating small cohorts of patients with advanced disease from dose level 3 to level 4 will partially depend on the proportion
(e.g., three patients), starting at a low dose of the agent(s) that is not of DLTs seen at dose level 1. The benefit of model-based escalations
expected to cause toxicity, and then increasing dose levels until unac- is that by using all the data, the investigators can better choose the
ceptable rates of toxicity (dose-limiting toxicity [DLT]) are seen. Phase next dose level at which to treat patients (if the assumed model is
I trials are designed to be small and are typically not limited to a correct). The problem with the methods is that if the assumed model

296
Clinical Trial Designs in Oncology  •  CHAPTER 18 297

is incorrect, too many patients may be treated at dose levels that are molecular target through use of low exposures of the agent not expected
too high.6 To mitigate this problem, it is useful to specify some to cause any toxicity.16
constraints on model-based approaches—for example, not allowing
the treatment of new patients at (or above) a dose level at which 33% PHASE II DESIGNS
or more of at least four patients have had a DLT. Regardless of whether
one uses a statistical model to guide the dose escalation, one could We first define some statistical parameters used to design clinical trials
use a model to fit the toxicity data (as a function of dose) after the to evaluate treatment effects. The three principal parameters are (1)
trial is over to identify the dose to recommend for further testing. the target treatment effect—the treatment effect of interest, which
the study should have a reasonable chance to identify (referred to
Combinations of Agents as the alternative hypothesis, contrasted with the null hypothesis of
no treatment effect); (2) the false-positive error rate (type I error)—the
For phase I trials involving a combination of agents, the goal is to probability of the study findings being positive when the null hypothesis
identify the doses of each of the agents to be used in the combination. is true; and (3) the false-negative error rate—the probability of the
There may be many dose combinations that have acceptable toxicity. study findings being negative when the alternative hypothesis is true,
For example, for a combination of agents X and Y, both high-dose X that is, the target treatment effect is present. (Note that the study
plus low-dose Y and low-dose X plus high-dose Y may have acceptable power, the probability of a positive study result under the alternative
toxicity (but not high-dose X plus high-dose Y), so the choice between hypothesis, is 1 minus the false-negative error rate.) An appropriate
the two acceptable combinations (and the dose escalation scheme) statistical design is obtained by selecting values for the three parameters
will need to be made based on biologic and clinical considerations. corresponding to the study goal.
If it is known that a certain minimal dose of X is necessary for its Phase II trials are designed to provide preliminary efficacy data to
effectiveness, then the escalation would be of agent Y, with the dose identify therapies that have sufficient activity to be worthy of further
of X fixed at its appropriate level. In addition, the toxicity profiles of testing in definitive (phase III) trials. Because they are preliminary,
the individual agents may suggest dose escalation schemes.9 the designs allow the probability of moving an ineffective therapy
forward to be larger than one would use in a phase III trial (e.g., a
Late Dose-Limiting Toxicities type I error of 10% instead of 2.5%). More important, phase II trials
typically use end points that measure treatment activity (e.g., tumor
For some treatments, some DLTs of concern may occur late—for shrinkage); the effect of the treatment on these end points is expected
example, toxicities occurring up to a year after radiation treatment. to be larger and available more quickly than the effect of the treatment
This can lead to a very long phase I trial if one has to wait a lengthy on a phase III end point that measures direct patient benefit (e.g.,
evaluation period (e.g., 1 year) for each cohort of patients before the survival). Both of these factors allow for a smaller sample size and
next cohort of patients can be treated. One approach to this problem shorter time to completion than a phase III trial.
is to model the occurrence of DLT times, for example, by assuming For a phase II trial of a single agent, the historical approach is a
that DLTs are uniformly likely to occur over the evaluation period. single-arm trial in a specific histologic type designed to assess whether
If this were true, then one could extrapolate from the early toxicity the experimental agent yields sufficient responses (partial or complete
experience of the patients to allow dose escalation before the patients responses as defined by the Response Evaluation Criteria in Solid
have been followed for the full evaluation period. An example of this Tumors [RECIST]17); the denominator for calculating the response
approach is the time-to-event continual reassessment method10 (although rate is typically taken to be all patients who started the agent. For
this particular method has apparently not worked well in practice3). example, one could consider a trial of 32 patients in which the agent
If one designs a trial assuming the uniform occurrence of DLTs over would be deemed worthy of further study if four or more responses
the evaluation period and the assumption in reality does not hold, were seen. This design would have both false-positive and false-negative
then one is at risk of exposing too many patients to DLTs. For example, error probabilities of less than 10% (a typical value chosen) for the
if all the DLTs typically occur near the end of the evaluation period, null and alternative response rates of interest. If the true response rate
then too many patients may be accrued at that dose level and a higher was 5% or less (the null hypothesis, considered too low to be interest-
dose level based on not seeing any early DLTs. Because of this concern, ing), then there would be less than a 10% probability of declaring
additional constraints on these approaches are useful—for example, the agent worth pursuing, and if the true response rate was 20% or
not accruing more than three patients at a dose level until the initial higher (the alternative hypothesis), then there would be less than a
three patients at that dose level have been observed for at least one-half 10% probability of a negative conclusion.
the evaluation period. To minimize the number of patients treated with an inactive agent,
various two-stage designs have been developed that allow for early
Biologic End Points stopping because of negative results.18,19 For example, in a study with
a Simon minimax two-stage design for targeting 5% versus 20%
Phase I designs that escalate to find the highest dose level with acceptable response rates (with 10% error rates), 18 patients are treated at the
toxicity (maximum tolerated dose) are based on the assumptions that first stage. If there is at least one response among these 18 patients,
(1) higher doses are more effective than lower doses, (2) higher doses a second stage of 14 patients is accrued. The agent is considered
are associated with more toxicity than lower doses, and (3) there is a worthy of further study if there are at least four responses seen among
dose with acceptable toxicity that will be effective.11 As opposed to the 32 patients. The required sample size of a single-arm phase II trial
cytotoxic agents, with targeted agents the first two assumptions may (either one-stage or two-stage) depends on the hypothesized targeted
not hold, and with immunologic agents the first assumption may not response rates of interest (e.g., 20% versus 5%), and the error prob-
hold. This suggests the use of a biologic (nontoxicity) end point to abilities (both 10% in the aforementioned examples). The sample size
guide the dose escalation and to choose the dose for further testing. will be smaller when the difference in target response rates is larger
Although attractive in theory,12,13 designs using nontoxicity end points (e.g., 25% versus 5%) or when the error probabilities are larger (e.g.,
have been infrequent in practice.14 Instead, it may be preferable to 15% instead of 10%).
assess the biologic end point at the maximum dose level determined
according to DLT, and possibly at lower dose levels for comparison.15 Randomized Screening Designs
In the context of a targeted agent, “phase 0 trials” are very small
first-in-human trials that are designed not to find the recommended There are two general situations in which a single-arm phase II trial
dose of the new agent, but instead to assess the agent’s effect on its design will not be appropriate because the results would be difficult
298 Part I: Science and Clinical Oncology

Table 18.1  Examples of Required Total Sample Sizes for Phase II and Phase III Randomized Trials
With a Response Rate End Point to Achieve 90% Power for the Designated Target
Response Rates
Response rates Control 20% 20% 20% 40% 40% 40% 60% 60%
Experimental 30% 40% 50% 50% 60% 70% 80% 90%
PHASE II (ONE-SIDED TYPE I ERROR = 10%)
Sample size 530 156 78 688 182 82 156 66
PHASE III (ONE-SIDED TYPE I ERROR = 2.5%)
Sample size 824 236 116 1076 280 126 236 98

Table 18.2  Examples of Required Total Numbers of Events for Phase II and Phase III Randomized Trials
With a Time-to-Event (Survival) End Point to Achieve 90% Power for the Designated Target
Hazard Ratioa
Hazard ratio 0.90 0.8 0.71 0.67 0.6 0.56 0.50 0.40
1/Hazard ratio 1.11 1.25 1.40 1.50 1.67 1.80 2.00 2.50
PHASE II (ONE-SIDED TYPE I ERROR = 10%)
Number of events 2367 528 232 160 101 76 55 31
PHASE III (ONE-SIDED TYPE I ERROR = 2.5%)
Number of events 3786 844 371 256 161 122 87 50

a
For outcomes that are approximately exponentially distributed, the hazard ratio is approximately equal to the ratio of the median survival in the experimental treatment
arm to the median survival in the control arm.

to interpret. The first is when the agent is thought to be primarily screening design: an intent-to-treat analysis, which includes all eligible
cytostatic rather than cytotoxic, so an effective agent might not shrink randomized patients, or only those eligible randomized patients who
tumors in a nonnegligible proportion of patients.20 One could, in started their assigned treatment. Restricting the analysis to patients
theory, use a time-to-event end point such as progression-free survival starting treatment will provide a more sensitive assessment of treatment
(PFS) or overall survival (OS) in this situation. However, because of efficacy but is subject to bias if substantially more patients in one arm
trial-to-trial variability in time-to-event outcomes, it can be difficult drop out of the trial before starting treatment; in the context of a
to identify a benchmark for an experimental treatment to beat in a phase II trial, this is not an issue if the trial is blinded.
single-arm trial.21 The other situation in which a single-arm phase II Unequal randomization is possible in a screening design, whereby,
trial design would not be appropriate is when the treatment being for example, twice as many patients are randomized to the experimental
evaluated is a combination of agents (agent X + agent Z), one of arm as the control arm. Unequal randomization makes the required
which is known to be active (agent Z). The problem is again identifying sample size larger—for example, 13%, 33%, or 278% larger for 2 : 1,
a null benchmark (e.g., response rate), in this case to isolate the 3 : 1, or 9 : 1 randomization, respectively. It is sometimes justified
contribution of agent X; there may be some data on the response rate because it is said that the unequal randomization will make the trial
of agent Z alone, but typically not from enough different trials in more attractive to patients, thereby speeding accrual (although we
comparable settings that one could feel comfortable moving the know of no hard evidence on this). A better justification for unequal
combination therapy forward if its observed response rate was mod- randomization is that in situations with limited experience with use
erately higher than that of the response rate of Z alone. of the experimental agent, it will allow more experience to be obtained
For situations in which a single-arm design is not appropriate, a (e.g., 40 patients instead of 30 in a 60-patient trial using 2 : 1 randomiza-
randomized screening design is an alternative.20,22 In this design, patients tion instead of 1 : 1 randomization). Other issues concerning randomized
are randomized to the experimental treatment versus a control treatment screening designs are discussed later in the section on phase III random-
to demonstrate superior activity with use of a phase II end point. ized designs.
Some examples of sample sizes for a response rate end point and a
time-to-event end point (e.g., PFS, OS) are shown in Tables 18.1 and Randomized Selection Designs
18.2, respectively. Note that for time-to-event end points, in which
the time-to-event (survival) curves are typically compared with a Another type of randomized phase II design is the randomized selection
log-rank statistic, the design can be specified in terms of the hazard design. In this design, the two (or more) treatment arms are experi-
ratio and the required number of events to be observed.23 For example, mental treatments. The goal of the trial is to select which of the
a trial to detect a hazard ratio of 0.5 with 90% power and 10% type treatment arms to take forward to further testing.24 At the end of the
I error would require 54 events. If one were targeting an improvement trial, the treatment arm that has the better efficacy outcomes is selected
to a median PFS of 6 months (for the experimental treatment) from for further development. For example, if response rate was chosen as
a median PFS of 3 months (for the control treatment), then one could the outcome, then the arm with the better response would be selected.
randomize 62 patients over 1 year; the analysis would be performed The sample size for this design is chosen so that there is a high
when 54 events were observed, which would be about 9 months after probability that one will not select a worse treatment arm, if in fact
the last patient was randomized. There is a choice regarding which there is one that is worse by a specified indifference margin. Selection
patients should be included in the primary analysis of a randomized designs require smaller sample sizes than screening designs because
Clinical Trial Designs in Oncology  •  CHAPTER 18 299

the conclusions from the design are much weaker (demonstrating that in both treatment arms together, but there are other alternatives32;
treatment X is not much worse than treatment Y instead of demonstrat- the specific timing of the definitive and interim analyses should be
ing that treatment X is better than treatment Y). When it is possible specified in the trial protocol. The trial protocol should also specify
to evaluate each arm individually for efficacy—for example, when a the analyses that will be performed and which patients will be included
response rate end point is being used for evaluating single agents—it the analyses. Typically, the intent-to-treat principle is used wherein
is possible to embed within the selection design minimum (single-arm) all eligible randomized patients are included in the analyses, and eligibil-
response rates. For example, one could select the treatment arm with ity is determined before randomization or assessed blindly based on
the better response rate provided that the response rate was at least prerandomization patient samples.
15%; otherwise, neither arm would warrant further study. The typical
use of selection designs is for deciding which of several schedules of Randomization and Stratification
a new agent to use for further agent development.
The randomization in a phase III trial is typically 1 : 1 (to avoid the
Designs With Biomarkers inefficiency of an unbalanced randomization), and, after informed
consent has been obtained, is ideally performed right before the
Phase II trials offer an opportunity to assess biomarkers for their experimental treatment is given (to avoid having to include patients
ability to enable identification of a patient population for which the who drop out of the trial before receiving the treatment under study).
experimental treatment will be effective or especially effective.25 Unless The randomization is typically stratified on a small number of variables
there is strong evidence that the treatment will work only in biomarker- thought to be highly associated with outcome to balance the distribution
positive patients, the phase II trial should not restrict enrollment to of these variables across the treatment arms and thus ensure that
such patients.26 Instead, the trial design should accommodate the approximately equal proportions of patients in each arm of the trial
possibility that benefit will be restricted but not assume it. For example, will have relatively good and bad prognoses (as determined according
in a single-arm trial with a response rate end point, two-stage designs to these stratifying variables). A stratified analysis can be used even if
can be used wherein both biomarker-positive and biomarker-negative stratification was not part of the randomization—for example, when
patients are accrued at the first stage; depending on what responses the stratifying variable is defined with a biomarker assay that is per-
are seen at the first stage, the trial is either stopped or more biomarker- formed on a prerandomization sample after the randomization.
positive or unselected patients are accrued.27,28 Stratification is more important in smaller trials than larger ones
When there is strong evidence that a molecularly targeted agent because the chances of a worrying imbalance are larger in a small
will work only in biomarker-positive patients and some evidence that trial. When outcomes have a subjective component, blinding of the
its efficacy may not depend on tumor histologic type, a single phase treatment assignment with placebos (when feasible) can remove a
II trial that pools response data across biomarker-positive patients in concern about potential bias in the trial results.
different histologic groups may be useful. However, in the absence of
reliable evidence that the activity level is uniform across these histologic Multiarm Trials
groups, a separate evaluation may have to be conducted in each group
to allow for a reliable evaluation. Note that the use of Bayesian hierarchi- A trial with multiple experimental arms and a single control arm can be
cal modeling to borrow information across groups does not work in an efficient way to test multiple treatments in a single disease setting.33
this setting,29 although simpler pooled futility rules can be useful.30 For example, a single randomized clinical trial (RCT) with a control arm
In settings in which a randomized screening design is used, one and three experimental arms can be 33% smaller than three separate
would again generally not restrict enrollment to biomarker-positive RCTs evaluating each experimental treatment separately. Multiarm
patients. After enrolling all comers, one could consider in an ad hoc trials can add complexity because they may require multiple placebos,
manner the experimental-versus-control treatment effect in the overall restriction of eligibility to ensure that all the agents can be given safely,
group and the biomarker-positive and biomarker-negative subgroups. agreement among multiple industry partners to participate and agree-
Alternatively, one could use a formal phase II biomarker trial design that ment regarding how the data will be analyzed and shared, complex
directly addresses the relevant drug development question31: Should a funding arrangements, and additional regulatory complexities.33,34
phase III trial be performed, and if so, how should the biomarker be A factorial trial design is a multiarm trial in which the arms represent
incorporated into that design? Phase II designs with multiple biomarkers all possible combinations of the experimental and control treatments.
are discussed later under Designs With Multiple Biomarkers. For example, in a 2 × 2 factorial design evaluating experimental
treatments A and B (relative to no treatment), patients are randomized
PHASE III DESIGNS to one of the following four arms: (1) no treatment (arm C), (2)
treatment A (arm A), (3) treatment B (arm B), or (4) treatment A
Phase III trials are the definitive randomized comparisons of treatments plus treatment B (arm AB). (In some applications, a common backbone
and should provide sufficiently compelling evidence to change clinical treatment will be given in all the arms.) A factorial analysis of a factorial
practice. Accordingly, they are designed with a small false-positive design assumes that the effect of treatment A does not depend on
rate—for example, a one-sided type I error rate of 2.5%, and high whether the patient received treatment B—that is, the benefit of arm
power for detecting truly positive effects (e.g., 80% to 90%). In A over arm C is the same as the benefit of arm AB over arm B. (This
addition to depending on the type I error and power, the required is equivalent to assuming that that effect of treatment B does not
sample size depends inversely on the magnitude of the targeted treat- depend on whether the patient received treatment A.) This assumption,
ment effect between the treatment arms and is typically hundreds or which is referred to as no statistical interaction among the treatments,
thousands of patients (see Tables 18.1 and 18.2). As noted previously, is very strong. When the assumption is satisfied, one can assess the
the required sample size for time-to-event end points is a function efficacy of A and B in a trial with the same sample size as would be
of the expected number of events, rather than the sample size. Therefore required to assess a single agent, a remarkable savings. However, if
the required sample size will be larger for a clinical setting with lower the assumption is not true, then a factorial analysis can incorrectly
event rates than one with higher event rates. For example, with 3 suggest that agents work when they do not, and do not work when
years of accrual and 3 years of follow-up, a setting with 70% 2-year they do.35,36 Even when a factorial analysis is problematic, the factorial
event-free survival will have a required sample size roughly 40% larger trial design with a nonfactorial analysis is an efficient way to study
than that needed for a setting with 50% 2-year event-free survival. two treatments and their possible interaction.36
For time-to-event end points, the definitive analysis and interim analyses When a multiarm trial allows new trial arms to be added as it is
are typically performed when the expected number of events is observed ongoing, it is referred to as a “master” protocol or a “platform” trial.
300 Part I: Science and Clinical Oncology

The idea is that when promising new agents become available they a relatively rare cancer, it may be impractical to perform a large RCT,
can be added (possibly along with corresponding control arms). In but the challenges in using results from nonrandomized trial designs
addition, trial arms are dropped when the questions involving these to change clinical practice should not be ignored.44
arms have been answered. An example of a master protocol is the
STAMPEDE,37 which evaluates various agents in addition to a standard INTERIM MONITORING
hormone therapy for advanced prostate cancer. The advantage of a
master protocol is that it allows testing of agents more quickly because Interim monitoring of accruing outcome data from a clinical trial is
a new protocol does not need to be developed for each new treatment. motivated by both ethical and resource considerations; it allows one
This can be especially important when new treatment arms are suggested to stop a trial as soon as the scientific question is answered. It is the
by the ongoing discovery of new biomarkers and targeted treatments. most important adaptive trial element of a clinical trial.
For any master protocol, one must avoid potential pressure to add
new treatment arms or trial questions that may not be of the highest Stopping for Superiority (Efficacy) in a
priority just to keep the trial ongoing. Randomized Clinical Trial

Noninferiority Trials If it becomes obvious during the course of a trial that the experimental
treatment is much more effective than the standard treatment, then
In a noninferiority trial, one is interested in showing that the new one would want to stop the trial and release the results to the public.
treatment is not inferior to the standard treatment. These trials arise However, an undisciplined examination of accruing outcome data
when the new treatment is expected to be less toxic or more convenient can lead to the release of results that do not have acceptable statistical
than the standard therapy. The sample size considerations are similar validity. For example, if one repeatedly examines accruing outcome
to those of a superiority trial (see Tables 18.1 and 18.2) with one data from a trial and stops it as soon as the (one-sided) P value is
important exception: the targeted treatment effect will typically be below .025, then the chances of incorrectly recommending an ineffective
smaller, leading to much larger trials. The targeted effect is small because treatment (a type I error) can be much higher than 2.5% (e.g., 12.5%
one does not want to recommend a new treatment that is inferior to with 20 examinations). Therefore the study protocol should include
a standard treatment by a nonnegligible amount. In particular, it has a formal interim analysis plan that prospectively designates the times
been suggested that the targeted difference should be no greater than when the interim analyses will be performed, as well as the decision
a fraction of the benefit (e.g., 50%) seen for the standard treatment,38 rules that will be used at each time to decide if the outcome data are
which itself may not be large. In designing a noninferiority trial, the extreme enough to stop.
role of type I error and power are reversed, so that priority is given A large number of formal interim analysis plans are possible.45,46
to minimizing the probability of declaring noninferiority when the The analysis times are typically specified in terms of the “information
new treatment is actually inferior; this probability should be kept very fraction,” which is the proportion of the information available in the
small (e.g., 2.5%), whereas the probability of declaring noninferiority data at the interim analysis time compared with the information
when the treatments are equally effective could be set at 80% to 90%. available at the regularly scheduled end of the trial (100%, by defini-
For noninferiority trials, using the intention-to-treat principle can tion). With a response rate end point, the information fraction is the
be problematic if there are a nonnegligible proportion of patients proportion of the total sample size that has been evaluated for response,
who do not receive their assigned treatments, because this can lead and with a survival end point it is the proportion of events observed
to underestimation of any true loss in efficacy38; to accommodate as compared with the total number of events required at the final
this, an additional analysis of only those patients who received their analysis time. Table 18.3 displays the popular Haybittle-Peto47 and
assigned treatments is typically performed.39 O’Brien-Fleming48 monitoring plans (in terms of P value cutoffs
In some situations, it is expected that the new treatment will required for stopping) for interim analyses at 40% and 70% informa-
actually be modestly better than the standard treatment, but because tion. (Many common interim monitoring designs accommodate flexible
it is less toxic, it would be sufficient to demonstrate that it is noninferior analysis timing using the “spending function approach.”49) Note that
to the standard treatment. In these special situations, the required the P value cutoffs for the final analysis are less than the nominal
sample size can be reduced.40 0.025 type I error of the trial that would be used if no interim analyses
were performed. Because of this, use of interim monitoring necessitates
Nonrandomized Trial Designs a small increase in the sample size to maintain the power of the trial
(e.g., 2% increases in sample size for a trial with 90% power for the
If a treatment works dramatically better than the standard treatment, O’Brien-Fleming plan described in Table 18.3).
then one does not need an RCT to obtain the necessary evidence to
change clinical practice. For example, the hematologic and cytogenetic
responses seen for second-line imatinib in single-arm studies of Phila-
delphia chromosome–positive (Ph+) chronic myelogenous leukemia
led the FDA to grant accelerated approval of this agent in that setting.41
Another example is the benefit of imatinib in pediatric Ph+ acute Table 18.3 One-Sided P Value Cutoffs for
lymphoblastic leukemia (ALL), in which an 80% 3-year event-free Haybittle-Peto and O’Brien-Fleming
survival rate was seen in a Children’s Oncology Group trial compared Interim Monitoring Plans With Interim
with 35% seen in three previous trials without imatinib.42 Care must Analyses at 40% and 70% Information
be taken when drawing conclusions from comparisons with historical for Overall One-Sided Type I Error
experience; positive-appearing results could be due to selection bias of 2.5%
(the patients treated with the experimental treatment may have better
prognoses than those previously treated with the standard treatment), INFORMATION TIME OF ANALYSIS
or improvements in ancillary care may make an experimental treatment Monitoring Plan 40% 70% 100% (Final Analysis)
appear better than a standard treatment. An example of how things
None — — .0250
can go wrong was the widespread use of high-dose chemotherapy
with autologous stem cell rescue for metastatic breast cancer. A series Haybittle-Peto .0010 .0010 .0246
of small single-arm trials had suggested that it was highly beneficial, O’Brien-Fleming .0004 .0073 .0227
but later RCTs demonstrated that it did not work.43 In considering
Clinical Trial Designs in Oncology  •  CHAPTER 18 301

Stopping for Inefficacy (Futility) in a Randomized interim monitoring. There are a number of drawbacks as compared
Clinical Trial with standard interim monitoring that make outcome-adaptive
randomization not useful or appropriate. The first is that any time
Most randomized trials are asking one-sided questions: Does the trends in the prognostic characteristics of the patient population
experimental treatment work better than a standard treatment? Accord- enrolled in the trial will bias the results of the trial, making it
ingly, there is no need to demonstrate that the experimental arm is difficult to interpret the results of a completed trial.62–65 A second
significantly worse that the standard therapy; instead it is sufficient problem is statistical inefficiency due to having an unequal number
to show that it is no better.50 Therefore interim monitoring should of patients in the treatment arms.63 This inefficiency means that
stop the trial for inefficacy as soon as tangible benefit over the control trials using outcome-adaptive randomization will necessarily be
treatment can be ruled out. Formal inefficacy interim analysis plans larger and will result in delay in getting new effective treatments to
allow for such stopping, but they keep a truly effective therapy from the clinical community, and they also will expose more patients to
being dismissed. bad outcomes. This is the same issue as discussed earlier with fixed
In a very simple approach to inefficacy monitoring, the outcome unequal randomization (e.g., 2 : 1 randomization) but can become
data are examined at 50% information, and the trial is stopped if the more problematic as the randomization ratio becomes more extreme
observed treatment effect favors the control arm (by any amount).51 in outcome-adaptive randomization (e.g., 9 : 1). A third problem with
A number of more complex approaches have been suggested (that outcome-adaptive randomization is that even though it will generally
allow for more interim looks).52,53 It has been argued that inefficacy put more patients in the better treatment arm (if there is one), it will
monitoring is so important for protecting patients from ineffective occasionally put a moderately larger proportion of patients on the
therapies that if it is not included in the trial protocol, or if the worse treatment arm.66 Finally, there has been ongoing discussion
specified monitoring plan has not been followed during the trial, then regarding ethical issues (pro and con) involved in outcome-adaptive
the reasons for this should be stated in the trial report.54 Inefficacy randomization.67–74
monitoring is also critical for noninferiority trials, as it allows stopping
a trial early when the new treatment is unlikely to be noninferior to Monitoring for Rare Serious Toxicities
the standard treatment (and may be non-negligibly inferior).54a
Most common toxicities of an experimental treatment are identified
Other Considerations for Efficacy and and quantified in the early testing of the agent. When the toxicity is
Inefficacy Monitoring rare, however, one can monitor for it in a large phase II or phase III
trial to assess its magnitude and see whether the treatment should be
With electronic data capture there is the possibility of easily having stopped or modified. For a rapidly occurring toxicity, one can use a
many interim analysis looks. Once interim monitoring has commenced, continuous monitoring plan that identifies the problem as soon as
there is very little cost in having frequent monitoring from that point the number of patients experiencing the toxicity crosses a defined
onward,55 so one can analyze the data more often and stop earlier boundary.75,76 For example, if a 1% (5%) rate of the toxicity is acceptable
(when appropriate). For example, looking at the data at 40% informa- (unacceptable), then one could use a Pocock boundary with the first
tion and then in 10% increments (six instead of two interim looks) 169 patients treated in the experimental arm that has the following
would require increasing the sample size by 3% for the O’Brien-Fleming properties: If the true toxicity rate is 5%, then the problem will be
monitoring plan. identified 95% of the time at an average sample size of 54. If the true
Potential drawbacks that should be considered when designing an toxicity rate is 1%, then the toxicity rate will be deemed acceptable
interim monitoring plan are that if a trial stops early, then there may 90% of the time.
be very little or no information about the longer-term effects of the
treatments or the effects of treatments on secondary end points, and PHASE II/III DESIGNS
the treatment effects will be estimated less precisely than if the trial
had not been stopped early. Of special concern is when the benefits A phase II/III trial is designed as a phase III trial but with an interim
of the experimental treatment over the control treatment are expected “phase II” go/no-go rule to decide whether the experimental treatment
to be delayed; in these situations, inefficacy monitoring should not is active enough to continue the trial to its phase III sample size.77
begin until the benefits of the experimental treatment are expected The advantages of a phase II/III trial over a phase II trial followed
to be seen.55a Another concern is that trials that stop early for superiority by a phase III trial are efficiency and speed, in that (1) the phase
overestimate the treatment effect.56 However, it has been shown that II patients can be included in the phase III analysis, and (2) one
except for stopping at very early interim analyses (≤25% of information), does not have to wait for the phase III protocol development after
the overestimation is minor.57 Furthermore, a review of the National the phase II results become available. Phase II/III trials are most
Cancer Institute (NCI) Cooperative Group phase III trials that were useful when the phase II end point (e.g., PFS) can be achieved
stopped early for positive results confirmed that mature results were earlier than the definitive phase III end point (e.g., OS). What the
consistent with the early results.58 phase II end point should be, how high the bar (go versus no-go
Interim monitoring is best performed by an independent data threshold) should be set for that end point, and whether there
monitoring committee,59,60 who are the only ones who see accruing should be an accrual suspension while waiting for the phase II data
outcome data (besides the trial statisticians). In some very special to mature are the key design considerations for phase II/III trials.78
circumstances, it may be possible to release preliminary data to the In general, one could choose the phase II end point and could set
public when a trial is in its follow-up stage and the release would not the bar at the same level as would be used in a stand-alone phase
compromise the integrity of the trial.61 II trial. When the same end point needs to be used for both the
phase II and phase III evaluations (e.g., when the treatment is not
Outcome-Adaptive Randomization expected to affect any end points other than OS), the gains achieved
with a phase II/III design will be modest unless there is an accrual
RCT designs generally keep arm assignment probabilities constant suspension.78a
throughout the trial (e.g., 1 : 1). However, there are designs in which There is a close relationship between phase II/III trials and phase
the accruing outcome data are used to adjust the randomization III trials with futility monitoring: in both cases, interim results are
ratio so that a higher proportion of patients are randomized to the used to decide whether to continue to the full phase III sample size.
treatment arm(s) that appear to be doing better (outcome-adaptive However, in a phase II/III trial, the interim results need to be sufficiently
randomization); this can be thought of as a type of continuous promising, whereas for a typical futility analysis the experimental
302 Part I: Science and Clinical Oncology

treatment just needs to be not worse. This difference is reasonable Progression-Free Survival
because in a phase III trial one has promising evidence from previous
phase II trials. In addition, it is possible to conduct a phase II/III trial PFS is defined as the time from randomization to progression or
with multiple experimental arms (along with the control arm), with death. (Time to progression is a related, less-preferred end point wherein
the most promising (or all of the promising) agents continuing to deaths without progression are censored observations rather than
phase III.79 Also, if the experimental treatment is a single agent expected counted as events.) Because PFS requires a subjective reading of patient
to produce responses (if it is effective), then it is possible for the scans, if there is a concern of bias in a trial in which the treatment
interim phase II look to be based solely on the experimental arm assignment is not blinded, a blinded independent central review is
response rate. often considered.87 However, this is not recommended as a general
A potential disadvantage of a phase II/III trial is that one is eliminat- strategy, because a central review can actually add bias to the trial
ing the flexibility of modifying the phase III trial design based on the results.88 In addition, evaluations comparing trial results using investiga-
results of the phase II trial—for example, changing the dose or schedule tor versus independent assessment of PFS have not shown substantial
of the experimental agent, supportive care requirements, or the patient differences.88,89 If investigator bias in evaluating PFS is a concern, one
population.80–82 In addition, committing to the question to be asked can perform an audit comparing investigator assessments versus
in phase III at an earlier time locks in a sponsoring organization that independent assessments of PFS.90 Another potential source of bias
may want to keep its options open longer with regard to what phase with the use of PFS in an unblinded trial is assessment time bias: if
III trials to perform. the assessment times are different between the treatment arms, a
standard PFS time-to-event analysis can give incorrect results.91 If
END POINTS FOR RANDOMIZED TRIALS there is a concern about bias in assessing PFS, it is best to conduct a
double-blind trial when possible.
The choice of a primary end point in an RCT should reflect the Because progression is typically measured according to a specified
study goal. In general, a primary end point for a phase III RCT radiologic increase in tumor size (or a change in blood chemistry
should measure direct clinical benefit or a surrogate for direct clinical values in hematologic cancers), PFS is a measure of biologic activity
benefit. On the other hand, the primary end point in a randomized and does not directly measure clinical benefit.17,86 A sufficiently large
phase II trial need only be able to suggest that the treatment would increase in PFS in a randomized phase II trial of an experimental
show direct clinical benefit in a larger trial or in a different clinical treatment suggests that the treatment should be assessed in a phase
setting. By “direct clinical benefit” we mean a “direct measure of how III trial. Similarly, a sufficiently large increase in PFS in an RCT in
a patient feels, functions, or survives.”83 This section first consid- a metastatic disease setting may suggest that the experimental treatment
ers some commonly used RCT end points, and whether they are will show direct clinical benefit in a less advanced disease setting.
measuring direct clinical benefit. This is followed by a consideration Related alternatives to PFS that capture later events, such as time
of patient-reported outcomes. We end with discussion of surrogate from randomization to second progression or death, have sometimes
end points. been used in trials of maintenance therapies to measure patient benefit
of a therapy in the context of the subsequent therapies.92 Considerations
Overall Survival for PFS as a surrogate end point for clinical benefit are discussed in
a subsequent section.
An experimental treatment that improves OS (death from any cause)
benefits patients. However, there are a number of potential complica- Disease-Free Survival
tions when it is used as a primary end point.84 It can lead to very
large and long trials in settings where the standard therapies (including Disease-free survival (DFS) is defined as the time from randomization
after relapses) work quite well. This is because the required size of the to recurrence of tumor or death, and it is typically used in the adjuvant
trial depends on the number of deaths observed. A related potential treatment setting. It can be considered a direct measure of clinical
difficulty with OS as the primary end point occurs when the trial benefit if the magnitude of its prolongation with an experimental
design crosses patients over to the experimental treatment at relapse therapy outweighs its toxicity87; a consideration here is the proportion
or progression. With crossover as part of the trial design, many patients of patients whose recurrences are symptomatic. For example, if the
in the standard arm will eventually receive the experimental treatment, recurrence of disease is measured with a very sensitive blood assay
potentially lessening the OS differences between the treatment arms. and the cancer is relatively indolent at recurrence, then it could be
If the experimental treatment is a standard second-line treatment years before patients become symptomatic, and it is questionable
being tested for first-line treatment (as is frequently the case when whether improving DFS is offering patients a direct clinical benefit.
agents are tested and approved in advanced disease settings first), then In some settings DFS may also be a surrogate for OS, because patients
this is not a problem; the OS differences observed between the who remain disease free for an extended period of time may be cured.
experimental arm and the control arm (standard treatment followed
by experimental treatment at relapse) exactly captures the clinical Tumor Response Rates
benefit of the experimental treatment.85 In particular, absence of an
improvement in OS means there is no benefit to patients from moving A sufficient improvement in tumor response rates in a randomized
the standard second-line treatment to the first line. If the experimental phase II trial shows activity that could be tested in a phase III trial.
treatment is not a standard second-line treatment, then OS may indeed Durable complete responses might be a surrogate for clinical benefit,
underestimate its benefits when a crossover is part of the trial design. but PFS would be a more efficient end point in a randomized trial.
It could be argued that crossover should not be allowed unless there
is definitive evidence that the experimental treatment is beneficial in Patient-Reported Outcomes
the second line.85,86
In evaluating a treatment in a population with high rates of compet- Patient-reported outcomes (PROs) related to a specific cancer symptom
ing mortality (e.g., an elderly population), noncancer deaths can dilute are sometimes used to provide primary or supporting evidence that
the treatment’s effect on OS, especially in an adjuvant setting. However, a treatment is offering direct clinical benefit (e.g., improvement in
the OS effect seen does represent the benefit to that population. If cancer-related pain).93 A number of trial design issues should be
one was interested in isolating the treatment’s effect on cancer and addressed when PROs are used.94 A PRO instrument that is appropriate
was able to accurately separate cancer from noncancer deaths, one for the trial population should be chosen to collect the patient evalu-
could use cancer-specific survival as the end point.84 ations. The design should specify when in patient courses the PROs
Clinical Trial Designs in Oncology  •  CHAPTER 18 303

100% 100%

75% 75%

Survival
Survival

50% 50%

25% 25%

0% 0%
A Time B Time
Figure 18.1  •  Hypothetical example for an individual-level surrogate that is not a trial-level surrogate (green curves, surrogate-positive patients; red curves,
surrogate-negative patients; black curves, overall) demonstrating individual-level but not trial-level surrogacy. (A) Standard treatment for which 30% of patients
have a positive value of the surrogate immediately after treatment. (B) Experimental treatment for which 58% of the patients have a positive value of the
surrogate immediately after treatment. Note that the treatments are equally effective in terms of overall survival (black curves) even though the proportion that
is positive for the (individual-level) surrogate outcome is higher for the experimental treatment.

will be recorded and whether a baseline PRO will be used to adjust more effective postrelapse treatments have become available and have
the patient responses before comparisons between treatment arms are changed the OS outlook.
made. Ideally, patients should be blinded as to their treatment assign- Trial-level surrogacy is frequently confused with individual-level
ment, and it may be useful to collect data on what treatment patients surrogacy; an individual-level surrogate end point is measured after
think they received to check for unintentional unblinding. The design treatment and predicts which patients will do well versus poorly given
should attempt to minimize missing data. Finally, the analysis strategy a specific treatment. One might think that a good individual-level
of the PRO data should be specified in the protocol, including how surrogate would always be a good trial-level surrogate, but this is not
missing data will be accommodated in the analysis. true.97,99 As a hypothetical example, consider the survival curves in Fig.
In addition to being used to measure treatment efficacy, PROs can 18.1. The binary surrogate variable (S) here is a good individual-level
be used to better understand the toxicities of treatments.95 They can surrogate for survival as seen by the green curves being above the
also be used as primary end points to assess agents used to lessen the red curves for both the experimental treatment (Fig. 18.1A) and the
toxicities of anticancer treatments. standard treatment (Fig. 18.1B). We assume that 70% of the patients
who receive the experimental treatment (X) have a positive S value,
Functional Outcomes whereas only 20% of the patients who receive the standard treatment
(Y) have a positive S value. If S were a good trial-level surrogate for
In some clinical situations the trial end point may be related to how survival, this would mean that treatment X was better than treat-
a patient functions, which is a measure of direct patient benefit. For ment Y. However, as seen in Fig. 18.1, the treatments have the same
example, in a phase III RCT of three different nonsurgical treatment survival (black curves in Fig. 18.1). Heuristically, one can imagine a
strategies for locally advanced laryngeal cancer,96 the primary end situation as depicted in Fig. 18.1 when the experimental treatment
point was laryngectomy-free survival (time from randomization to moves some of the better prognosis surrogate-negative patients into
laryngectomy or death). Another example is a phase II trial of radiation being positive on the surrogate, without affecting their survival. A
and chemotherapy for stage T1 bladder cancer (NCT00981656) in real example of an individual-level surrogate for which there is no
which the primary end point was freedom from radical cystectomy evidence of trial-level surrogacy is pathologic complete response as
at 3 years. a surrogate end point for event-free survival or OS for early-stage
breast cancer.99 An intermediate end points can be useful for drug
Trial-Level Surrogate End Points development even though it has not been demonstrated to be a trial-
level surrogate.99a
A trial-level surrogate end point is one for which the results of the
RCT with this end point can be used to reliably predict what the PHASE III TRIAL DESIGNS WITH A
results of the trial would be with the definitive end point. A trial-level SINGLE BIOMARKER
surrogate end point may allow the results of the trial to be obtained
earlier and with a smaller sample size than if the definitive end point Traditional phase III trials focus on an average treatment effect for
was used. It is, however, challenging to demonstrate trial-level surrogacy; an overall population. However, new targeted agents often benefit
it requires a comparison of the two end points in a series of trials in only a subset of patients whose tumors express the molecular target.
a similar disease setting, with the treatments being tested having similar The use of biomarkers measured before treatment allows one to focus
mechanisms of action.97 For example, Sargent and colleagues98 examined trials on the patients who are most likely to benefit.
the results of 18 RCTs of adjuvant fluorouracil-based therapies for
colon cancer and showed that 3-year DFS was a good surrogate for Prognostic and Predictive Biomarkers
5-year OS. However, this trial-level surrogacy would not necessarily
carry over to a different clinical setting, different types of treatments Clinical trial biomarkers are generally described as prognostic or
(e.g., antiangiogenic agents or immunotherapies), or later times when predictive. A prognostic biomarker categorizes patients into subgroups
304 Part I: Science and Clinical Oncology

with distinct prognoses—that is, patients who will do relatively well Enrichment Designs
versus relatively poorly when treated with no treatment or a standard
treatment. For example, KRAS mutation is a prognostic biomarker When there is convincing evidence that any treatment benefit of the
(for OS) for patients with metastatic colorectal cancer treated with experimental treatment would be limited to biomarker-positive patients
best supportive care, the control arm in an RCT of panitumumab.100 (based on clinical and preclinical data), then the trial should include
For patients receiving best supportive care, the median OS was 7.6 only patients who are biomarker positive (Fig. 18.2A). By identifying
months for patients who had wild-type KRAS tumors and 4.4 months the subpopulation in which the treatment is expected to work well,
for patients who had mutant KRAS tumors. The clinical usefulness one can frequently target a larger treatment effect than would be seen
of a prognostic biomarker is measured according to its ability to in the overall population, thereby reducing the required sample size
improve patient outcome by guiding patient treatment (more treatment of the trial. An example of an enrichment trial is the trial of the BRAF
or experimental treatments for high-risk patients and/or less treatment inhibitor vemurafenib versus dacarbazine in patients with metastatic
for low-risk patients). melanoma carrying the BRAF V600E mutation107; the hazard ratios
A predictive biomarker for an experimental treatment identifies for PFS and OS were 0.26 and 0.37, respectively. However, even if
patients for whom that treatment is especially beneficial—that is, the the sample size required for the randomized trial is small, the number
treatment effect (comparing the experimental treatment and the standard of patients screened to obtain these biomarker-positive patients may
treatment) is larger for the biomarker-positive patients than for the be large, depending on the prevalence of biomarker positivity and the
biomarker-negative patients. An example of a predictive biomarker (for ability of the biomarker to identify treatment-responsive patients.108
PFS) is epidermal growth factor receptor (EGFR) mutation status for Use of an enrichment design does not allow assessment of the treatment
gefitinib for non–small cell lung cancer patients.101 Gefitinib is more effect in the biomarker-negative patients.
effective than chemotherapy for EGFR mutation–positive patients
but less effective than chemotherapy for EGFR mutation–negative Biomarker-Stratified Designs
patients. EGFR mutation status in this case not only is predictive but
also exhibits a “qualitative interaction,” defined by the experimental In a biomarker-stratified design, both biomarker-positive and biomarker-
treatment working better than the standard treatment for biomarker- negative patients are randomized (see Fig. 18.2B). Alternatively, all
positive patients, but being only equally effective (or worse) for the patients are randomized and their biomarker status is determined
biomarker-negative patients.102 Predictive biomarkers—that is, those after randomization. The advantage of the first approach is that one
exhibiting a qualitative interaction—have clinical usefulness because is guaranteed that all the patients will have their biomarker status
they can direct treatment in an obvious manner.103 Biomarkers can determined. The advantage of the second approach is that one will
be both predictive and prognostic. not have to wait for the biomarker evaluation to enroll the patient
From randomized clinical trial data, one can evaluate a biomarker in the trial.109
to see if it is predictive for the experimental treatment and/or prognostic. There are several different analytic approaches for analyzing a
This can be done with a well-defined analysis plan on previously biomarker-stratified design, which involve various strategies for testing
collected trial data if the biomarker status is available.104 For planning the treatment effect in the biomarker-positive, biomarker-negative, and/
a phase III trial, the choice of a biomarker design should depend on or overall populations.105 A good strategy should aim at maximizing
the strength of the available evidence that the biomarker can be the probability that the new treatment is recommended for patients
successfully used to identify patients who will benefit from the who benefit and minimizing the probability that the treatment is
experimental treatment.105 In all cases, before a biomarker is used in recommended for patients who do not benefit. Accordingly, analysis
a phase III trial, its analytic validity should be ensured—that is, the strategies that recommend the treatment for the biomarker-negative
biomarker assay should consistently capture the biologic characteristics subpopulation without assessing the treatment effect in that sub-
it is putatively measuring.106 population should generally be avoided.110,111 Two effective strategies

Evaluate biomarker Evaluate biomarker

Biomarker positive Biomarker negative Biomarker positive Biomarker negative

Randomize Randomize Randomize

New Standard New Standard New Standard


treatment treatment Off study treatment treatment treatment treatment

A B
Figure 18.2  •  Two biomarker randomized clinical trial designs. (A) Enrichment design. (B) Biomarker-stratified design. (From Freidlin B, Korn EL.
Biomarker enrichment strategies: matching trial design to biomarker credentials. Nat Rev Clin Oncol. 2014;11:81–90.)
Clinical Trial Designs in Oncology  •  CHAPTER 18 305

Yes Recommend the treatment


for biomarker-positive and
biomarker-negative patients
Yes Test
biomarker-negative if
significant at level α
Recommend the treatment
Test for biomarker-positive
biomarker-positive if No patients only
significant at level α

STOP Do not recommend treatment


A No

Yes Recommend the treatment


for biomarker-positive and
biomarker-negative patients
Yes Test
biomarker-negative if
significant at level α
Recommend the treatment
for biomarker-positive
No patients only
Test
biomarker-positive if
significant at level α1
Yes Recommend the treatment
for all patients
Test
overall population if
significant at level α−α1
No
Do not recommend treatment
B No

Figure 18.3  •  Two analysis strategies for analyzing a biomarker-stratified design (α is the overall type I error of the design). (A) Sequential subgroup-specific
strategy. The biomarker-positive subgroup is first tested with type I error of α. If this test rejects the null hypothesis of no treatment effect, then the biomarker-
negative subgroup is tested with type I error of α. (B) MaST strategy. The biomarker-positive subgroup is first tested with type I error of α1, a value less than
α. Depending on whether this test rejects the null hypothesis of no treatment effect, the biomarker-negative subgroup or overall randomized population is
tested with type I error of α or α − α1, respectively. (From Freidlin B, Korn EL. Biomarker enrichment strategies: matching trial design to biomarker credentials.
Nat Rev Clin Oncol. 2014;11:81–90.)

for analyzing a biomarker-stratified design are the subgroup-specific Designs With a Continuous Biomarker
sequential strategy (Fig. 18.3A) and the MaST strategy112 (Fig. 18.3B).
Ideally, a cut point separating positive and negative values of a biomarker
Biomarker-Strategy Designs would be determined before its prospective evaluation in a phase III
trial. For example, cut points for the prognostic Oncotype DX test
In a biomarker-strategy design with a single biomarker, patients are were developed from retrospective analyses of tumor specimens from
randomized to a standard therapy versus a “strategy” treatment arm two completed breast cancer trials before the cut points were applied
in which the treatment is chosen based on the biomarker status. If prospectively in the TAILORx trial.113,114 If the cut point of a continuous
the biomarker status is positive, then the patients receive the experi- biomarker is not precisely determined before a planned RCT, sometimes
mental treatment; otherwise they receive the standard treatment. there is a lower value of it for which it is believed the experimental
Although superficially appealing because it directly estimates how well treatment will not work at all, and there is also a higher value that is
the biomarker-guided treatment would improve outcomes in practice, thought to identify patients who will be most helped by the treatment.
this design is inefficient owing to the dilution of the between-arm In these situations, one can randomize only patients who have a value
difference in outcomes as a result of a potentially large proportion of greater than this lower value, but specify the higher value as the
patients being treated with the standard treatment in both arms of cut point for a biomarker-stratified design. For example, in the
the trial. Therefore use of this design (with a single biomarker) is not KEYNOTE-010 trial,115 patients with advanced non–small cell lung
recommended. Instead, one can use a biomarker-stratified design to cancer patients whose PD-L1 expression was at least 1% were random-
estimate the treatment effect in the biomarker-positive and biomarker- ized to pembrolizumab (an antibody against PD-1) versus docetaxel,
negative subgroups, and then use these subgroup-specific estimates with a biomarker-stratified design stratified on PD-L1 expression
to estimate more efficiently how well a biomarker strategy would (1%–49% versus ≥50%). In situations in which little pretrial evidence
compare with a standard treatment.109 for a cutoff is available, there are designs116 that allow a rigorous
306 Part I: Science and Clinical Oncology

assessment of different possible cutoffs in the phase III trial (controlling proportion of patients receiving the standard treatment in the
for the multiple comparisons), but at the cost of lower statistical power biomarker-driven treatment arm. A modified biomarker-strategy design
as compared with a design with a predetermined cutoff. enrolls only patients who are positive for one of the biomarkers, who
are than randomized between a standard treatment arm and the arm
DESIGNS WITH MULTIPLE BIOMARKERS in which the biomarkers are used to determine which experimental
therapy will be recommended; this completely eliminates the problem
The promise of precision medicine is the use of multiple biomarkers of patients receiving a standard therapy in the biomarker-strategy
to select a treatment that is most likely to work in a given patient. treatment arm. An example is the histology-agnostic SHIVA trial,124
Development and validation of these biomarkers and treatment strategies in which patients with alterations within one of three molecular
involves clinical trial designs that may or may not use the biomarkers pathways were randomized to a matched targeted agent or physician’s
to direct treatment. choice of standard cytotoxic therapy. A shortcoming of a biomarker-
strategy design with multiple biomarkers is that it can be hard to
Biomarker-Directed Treatment Designs interpret the results of the trial; a negative result may occur because
enough of the biomarker-directed treatments are not working even
Trials using biomarker-directed treatments can have different designs though others work quite well, and a positive trial result may occur
depending on their objectives.117 In an umbrella trial, patients with because several of the biomarker-directed treatments are working very
a specific tumor histologic type are directed into different subtri- well, thus obscuring absence of benefit for the rest of the treatments.
als based on their biomarkers. For example, the Adjuvant Lung The SHIVA trial results did not show a benefit for the biomarker-driven
Cancer Enrichment Marker Identification and Sequencing Trial strategy, with a possible reason being that the agents did not work
(ALCHEMIST)118 enrolls patients with early-stage nonsquamous for the subset of patients with hormonal molecular alterations.124
non–small cell lung cancer into one of three phase III RCTs: a trial A critical source of the design efficiency of a biomarker-directed
of erlotinib versus no treatment for patients with EGFR-mutant treatment design is the screening component, which allows screening
tumors, a trial of crizotinib versus no treatment for patients with of patients for multiple biomarkers to direct them into the correspond-
ALK-rearranged tumors, and a trial of nivolumab versus no therapy ing biomarker-defined subtrials. Given that many biomarkers have
for patients without EGFR mutations or ALK rearrangements. The low prevalence of positivity, it may not be feasible to perform separate
master protocol FOCUS4 trial119 for patients with advanced or trials for each biomarker-associated treatment.
metastatic colorectal cancer with stable or responding disease after
first-line chemotherapy enrolls patients into separate phase II/III Nonbiomarker Directed Designs
trials based on their biomarkers, with patients without a biomarker
match currently being enrolled in a phase II/III trial of capecitabine In nonbiomarker directed designs the biomarkers of the patients are
versus active monitoring. The master protocol LUNG-MAP120 enrolls recorded but the patients are (initially) randomized to the treatment
patients with previously treated squamous cell lung cancer into arms regardless of the status of their biomarkers, with the efficacy
biomarker-directed single-arm phase II trials, with patients not being assessments being made within biomarker subsets. The biomarkers
positive for any of the specified biomarkers currently being enrolled can also be used in the interim monitoring—for example, to stop the
in a randomized phase II trial of nivolumab versus nivolumab plus randomization to a certain treatment for a certain biomarker subgroup.
ipilimumab. An example is the randomized phase II BATTLE trial,125 in which
A basket trial is similar to an umbrella trial except the patients chemotherapy-refractory non–small cell lung cancer patients were
enrolling are not restricted to a specific tumor histologic type. An randomly assigned to four therapies and were simultaneously categorized
example of a basket trial is the master protocol NCI-MATCH,121 in into five biomarker subgroups; the outcome was 8-week disease control
which patients whose disease has progressed on standard therapies are rates compared with historical controls. Another example is the random-
assigned to a phase II single-arm trial of a targeted agent when they ized phase II BATTLE-2 trial,126 in which advanced non–small cell
have the relevant biomarker target; the trial is expected assess 20 to lung cancer patients were randomized to a control arm or three
30 targeted agents. The raison d’être of a basket trial is the assumption experimental arms; the outcome was 8-week disease control rates
that response to a targeted treatment will depend on the biomarkers compared with the control arm. Trial designs such as these in which
of the patients and not on (or not much on) the histologic type of biomarkers are used for monitoring and assessment are more exploratory
the tumor. If one pools across histologic types, it is feasible to conduct than trials that use biomarkers to direct treatment, and are appropriate
a subtrial of a targeted agent in which the target may be quite rare. when one has little idea about the relationships between the biomarkers
There is the possibility that the assumption is wrong—for example, and the therapies.
BRAF inhibitors work in BRAF-positive melanoma but not BRAF-
positive colon cancer122—so a negative subtrial does not conclusively CONCLUSIONS
prove that the targeted agent would not be effective in some specific
histologic type with the target. It is also possible to design a basket Properly designed clinical trials ensure that patients are treated safely
trial that assesses the outcome within each histologic type without and that the scientific objectives of the trial can be achieved in an
pooling. For example, a basket trial was performed to examine the efficient manner, so that current and future patients can benefit from
response rate of the BRAF inhibitor vemurafenib for BRAF-positive the trial results. Judicious use of important trial design elements, such
patients in seven histologically specific cohorts.123 as interim monitoring and the choice of appropriate end points, can
Biomarker-strategy designs wherein patients are randomized to an ensure that trials definitively answer their scientific objectives as quickly
arm in which the treatment is suggested by the biomarker versus a as possible, be that changing clinical practice or screening a new
standard treatment arm were discussed in the context of a single treatment for further development. The increasing use of biomarkers
biomarker earlier. It was noted that use of biomarker-stratified designs in cancer treatment algorithms has great potential for improving patient
is preferable because biomarker-strategy designs are inefficient—they care, but does offer some challenges to clinical trial designs. Clinical
can result in treating a high proportion of patients in both arms with trial designs have been developed to assess the clinical benefits of
the same treatment (the standard treatment). In the setting of many experimental biomarker-directed agents and to assess the usefulness
biomarkers, each with a small probability of being positive, a biomarker- of multiple biomarkers for guiding patient treatment.
stratified design may not be feasible. In addition, use of many biomark-
ers as targets for experimental treatments increases the probability The complete reference list is available online at
that a patient will be positive for some biomarker, reducing the ExpertConsult.com.
Clinical Trial Designs in Oncology  •  CHAPTER 18 307

KEY REFERENCES
1. Green S, Smith A, Benedetti J, Crowley J. Clinical 44. Korn EL, McShane LM, Freidlin B. Statistical 88. Dodd LE, Korn EL, Freidlin B, et al. Blinded
Trials in Oncology. 3rd ed. New York: CRC Press; challenges in the evaluation of treatments for small independent central review of progression-free
2012. patient populations. Sci Transl Med. 2013;5:178r3. survival in phase III clinical trials: important design
3. Tourneau CL, Lee JJ, Siu LL. Dose escalation 48. O’Brien PC, Fleming TR. A multiple testing element or unnecessary expense? J Clin Oncol.
methods in phase I cancer clinical trials. J Natl procedure for clinical trials. Biometrics. 1979;35:549– 2008;26:379–3796.
Cancer Inst. 2009;101:708–720. 556. 92. Freidlin B, Little RF, Korn EL. Design issues in
8. O’Quigley J, Pepe M, Fisher L. Continual reassess- 49. Lan KKG, DeMets DL. Discrete sequential bound- randomized clinical trials of maintenance therapies.
ment method: a practical design for phase I clinical aries for clinical trials. Biometrika. 1983;70:659– J Natl Cancer Inst. 2015;107(11):djv225.
trials in cancer. Biometrics. 1990;46:33–48. 663. 94. U. S. Food and Drug Administration. Guidance
11. Korn EL. Nontoxicity endpoints in phase I trial 51. Wieand S, Schroeder G, O’Fallon JR. Stopping when for Industry. Patient-Reported Outome Measures:
designs for targeted, non-cytotoxic agents. J Natl the experimental regimen does not appear to help. Use in Medical Product Development to Support
Cancer Inst. 2004;96:977–978. Stat Med. 1994;13:1453–1458. Labeling Claims. Rockville, MD. 2009.
14. Parulekar WR, Eisenhauer EA. Phase 1 trial design for 52. Freidlin B, Korn EL, Gray R. A general inefficacy 97. Korn EL, Albert PS, McShane LM. Assessing sur-
solid tumor studies of targeted, non-cytotoxic agents: interim monitoring rule for randomized clinical rogates as trial endpoints using mixed models. Stat
theory and practice. J Natl Cancer Inst. 2004;96: trials. Clin Trials. 2010;7:197–208. Med. 2005;24:163–182.
990–997. 54a. Korn EL, Freidlin B. Interim monitoring for 98. Sargent DJ, Wieand HS, Haller DG, et al. Disease-
16. Murgo AJ, Kummar S, Rubinstein L, et al. non-inferiority trials: minimizing patient exposure free survival versus overall survival as a primary end
Designing phase 0 clinical trials. Clin Cancer Res. to inferior therapies. Ann Oncol. 2018;29:573–577. point for adjuvant colon cancer studies: individual
2008;14:3675–3682. 58. Korn EL, Freidlin B, Mooney M. Stopping or patients data from 20,898 patients on 18 randomized
17. Eisenhauer EA, Therasse P, Bogaerts J, et al. New reporting early for positive results in randomized trials. J Clin Oncol. 2005;23:8664–8670.
response criteria in solid tumors: revised RECIST clinical trials: the national cancer institute coopera- 99. Korn EL, Sachs MC, McShane LM. Statistical
guideline (version 1.1). Eur J Cancer. 2009;45: tive group experience from 1990 to 2005. J Clin controversies in clinical research: assessing patho-
228–247. Oncol. 2009;27:1712–1721. logic complete response as a trial-level surrogate
18. Simon R. Optimal two-stage designs for phase II 59. Smith MA, Ungerleider RS, Korn EL, Rubinstein end point for early-stage breast cancer. Ann Oncol.
clinical trials. Control Clin Trials. 1989;10:1–10. L, Simon R. Role of independent data-monitoring 2016;27:10–15.
20. Korn EL, Arbuck SG, Pluda JM, et al. Clinical trial committees in randomized clinical trials sponsored 103. Polley MY, Freidlin B, Korn EL, et al. Statistical
designs for cytostatic agents: are new approaches by the national cancer institute. J Clin Oncol. and practical considerations for clinical evalua-
needed? J Clin Oncol. 2001;19:265–272. 1997;15:2736–2743. tion of predictive biomarkers. J Natl Cancer Inst.
21. Korn EL, Liu PY, Lee SJ, et al. Meta-analysis of 60. Ellenberg SS, Fleming TR, DeMets DL. Data 2013;105:1677–1683.
phase II cooperative group trials in metastatic stage Monitoring Committees in Clinical Trials. Chichester, 104. Simon RM, Paik S, Hayes DF. Use of archived
IV melanoma to determine progression-free and England.: Wiley; 2002. specimens in evaluation of prognostic and predictive
overall survival benchmarks for future phase II trials. 63. Korn EL, Freidlin B. Outcome-adaptive random- biomarkers. J Natl Cancer Inst. 2009;21:1446–1452.
J Clin Oncol. 2008;26:527–534. ization: is it useful? J Clin Oncol. 2011;29:771– 105. Freidlin B, Korn EL. Biomarker enrichment strate-
22. Rubinstein LV, Korn EL, Freidlin B, et al. Design 776. gies: matching trial design to biomarker credentials.
issues of randomized phase II trials and a proposal 65. Korn EL, Freidlin B. Adaptive clinical trials: Nat Rev Clin Oncol. 2014;11:81–90.
for phase II screening trials. J Clin Oncol. 2005;23: advantages and disadvantages of various adaptive 106. Clark GM, McShane LM. Biostatistical consider-
7199–7206. design elements. J Natl Cancer Inst. 2017;109(6). ations in development of biomarkers-based tests to
23. Rubinstein LV, Gail MH, Santner TJ. Planning the 66. Thall P, Fox P, Wathen J. Statistical controversies guide treatment decisions. Stat Biopharm Res. 2011;3:
duration of a comparative clinical trial with loss to in clinical research: scientific and ethical problems 549–560.
follow-up and a period of continued observation. with adaptive randomization in comparative clinical 109. Freidlin B, McShane LM, Korn EL. Randomized
J Chronic Dis. 1981;34:469–479. trials. Ann Oncol. 2015;26:1621–1628. clinical trials with biomarkers: design issues. J Natl
29. Freidlin B, Korn EL. Borrowing information across 67. Hey SP, Kimmelman J. Are outcome-adaptive alloca- Cancer Inst. 2010;102:152–160.
subgroups in phase II trials: is it useful? Clin Cancer tion trials ethical? Clin Trials. 2015;12:102–106. 111. Freidlin B, Sun Z, Gray R, Korn EL. Phase III
Res. 2012;19:1–9. 72. Buyse M. Commentary on Hey and Kimmelman. clinical trials that integrate treatment and biomarker
31. Freidlin B, McShane LM, Polley MC, Korn EL. Clin Trials. 2015;12:119–121. evaluation. J Clin Oncol. 2013;31:3158–3161.
Randomized phase II trial designs with biomarkers. 78. Korn EL, Freidlin B, Abrams JS, Halabi S. Design 116. Jiang W, Freidlin B, Simon R. Biomarker-adaptive
J Clin Oncol. 2012;30:3304–3309. issues in randomized phase II/III trials. J Clin Oncol. threshold design: a procedure for evaluating treat-
33. Freidlin B, Korn EL, Gray R, Martin A. Multi-arm 2012;30:667–671. ment with possible biomarker-defined subset effect.
clinical trials of new agents: some design consider- 80. U. S. Food and Drug Administration. Draft Guid- J Natl Cancer Inst. 2007;99:1036–1043.
ations. Clin Cancer Res. 2008;14:4368–4371. ance for Industry—Adaptive Design Clinical Trials 117. Simon R. Genomic alteration-driven clinical trial
35. Green S, Liu PY, O’Sullivan J. Factorial design for Drugs and Biologics. Rockville, MD. 2010. designs in oncology. Ann Intern Med. 2016;165:
considerations. J Clin Oncol. 2002;20:3424–3430. 85. Korn EL, Freidlin B, Abrams J. Overall survival 270–278.
38. U. S. Food and Drug Administration. Guidance for as the outcome for randomized clinical trials 121. Conley BA, Doroshow JH. Molecular analysis for
Industry—Non-Inferiority Clinical Trials to Establish with effective subsequent therapies. J Clin Oncol. therapy choice: NCI MATCH. Semin Oncol. 2014;41:
Effectiveness. Silver Spring, MD. 2016. 2011;29:2439–2442. 297–299.
40. Freidlin B, Korn EL, George SL, Gray R. Random- 87. U. S. Food and Drug Administration. Guidance for
ized clinical trial design for assessing noninferiority Industry. Clinical Trial Endpoints for the Approval
when superiority is expected. J Clin Oncol. 2007;25: of Cancer Drugs and Biologics. Rockville, MD.
5019–5023. 2007.
Clinical Trial Designs in Oncology  •  CHAPTER 18 307.e1
307.e1

REFERENCES
1. Green S, Smith A, Benedetti J, Crowley J. Clinical 22. Rubinstein LV, Korn EL, Freidlin B, et al. Design 44. Korn EL, McShane LM, Freidlin B. Statistical
Trials in Oncology. 3rd ed. New York: CRC Press; issues of randomized phase II trials and a pro- challenges in the evaluation of treatments for small
2012. posal for phase II screening trials. J Clin Oncol. patient populations. Sci Transl Med. 2013;5:178r3.
2. George SL, Wang X, Pang H. Cancer Clinical Trials. 2005;23:7199–7206. 45. Jennison C, Turnbull BW. Group Sequential Methods
Boca Raton; 2016. 23. Rubinstein LV, Gail MH, Santner TJ. Planning the With Applications to Clinical Trials. Boca Raton:
3. Tourneau CL, Lee JJ, Siu LL. Dose escalation duration of a comparative clinical trial with loss to Chapman & Hall/CRC; 2000.
methods in phase I cancer clinical trials. J Natl follow-up and a period of continued observation. 46. Whitehead J. The Design and Analysis of Sequential
Cancer Inst. 2009;101:708–720. J Chronic Dis. 1981;34:469–479. Clinical Trials. Revised. 2nd ed. Chichester: Wiley;
4. Storer BE. Design and analysis of phase I clinical 24. Simon R, Thall PF, Ellenberg SS. New designs for 1997.
trials. Biometrics. 1989;45:925–937. the selection of treatments to be tests in randomized 47. Haybittle JL. Repeated assessment of results in
5. Skolnik JM, Barrett JS, Jayaraman B, Patel D, clinical trials. Stat Med. 1994;13:417–429. clinical trials of cancer treatment. Br J Radiol.
Adamson PC. Shortening the timeline of pediatric 25. McShane LM, Hunsberger S, Adjei AA. Effective 1971;44:793–797.
phase I trials: the rolling six design. J Clin Oncol. incorporation of biomarkers into phase II trials. Clin 48. O’Brien PC, Fleming TR. A multiple testing proce-
2008;26:190–195. Cancer Res. 2009;15:1898–1905. dure for clinical trials. Biometrics. 1979;35:549–556.
6. Korn EL, Midthune D, Chen TT. A comparison 26. Seymour L, Ivy SP, Sargent D, et al. The design of 49. Lan KKG, DeMets DL. Discrete sequential boundar-
of two phase I trial designs. Stat Med. 1994;13: phase II clinical trials testing cancer therapeutics: ies for clinical trials. Biometrika. 1983;70:659–663.
1799–1806. consensus recommendations from the clinical trial 50. Freidlin B, Korn EL. Monitoring for lack of benefit:
7. Simon R, Freidlin B, Rubinstein L, et al. Accelerated design task force of the National Cancer Institute a critical component of a randomized clinical trial.
titration designs for phase I clinical trials in oncology. Investigational Drug Steering Committee. Clin J Clin Oncol. 2009;27:629–633.
J Natl Cancer Inst. 1997;89:1138–1147. Cancer Res. 2010;16:1764–1769. 51. Wieand S, Schroeder G, O’Fallon JR. Stopping when
7a. Yuan Y, Hess KR, Hilsenbeck SG, Gilbert MR. 27. Pusztai L, Anderson K, Hess KR. Pharmacogenomic the experimental regimen does not appear to help.
Bayesian optimal interval design: a simple and predictor discovery in phase II clinical trials for breast Stat Med. 1994;13:1453–1458.
well-performing design for phase I oncology trials. cancer. Clin Cancer Res. 2007;13:6080–6086. 52. Freidlin B, Korn EL, Gray R. A general inefficacy
Clin Cancer Res. 2016;22:4291–4301. 28. Jones CL, Holmgren E. An adaptive Simon two-stage interim monitoring rule for randomized clinical
8. O’Quigley J, Pepe M, Fisher L. Continual reassess- design for phase 2 studies of targeted therapies. trials. Clin Trials. 2010;7:197–208.
ment method: a practical design for phase I clinical Contemp Clin Trials. 2007;28:654–661. 53. Zhang Q, Freidlin B, Korn EL, et al. Comparison of
trials in cancer. Biometrics. 1990;46:33–48. 29. Freidlin B, Korn EL. Borrowing information across futility monitoring guidelines using completed phase
9. Korn EL, Simon R. Using the tolerable-dose diagram subgroups in phase II trials: is it useful? Clin Cancer III oncology trials. Clin Trials. 2017;14(1):48–58.
in the design of phase I combination chemotherapy Res. 2012;19:1–9. 54. Korn EL, Freidlin B. Inefficacy interim monitoring
trials. J Clin Oncol. 1993;11:794–801. 30. LeBlanc M, Rankin C, Crowley J. Multiple histology procedures in randomized clinical trials: the need
10. Cheung YK, Chappell R. Sequential designs for phase phase II trials. Clin Cancer Res. 2009;15:4256–4262. to report. Am J Bioeth. 2011;11:2–10.
I clinical trials with late-onset toxicities. Biometrics. 31. Freidlin B, McShane LM, Polley MC, Korn EL. 54a. Korn EL, Freidlin B. Interim monitoring for
2000;56:1177–1182. Randomized phase II trial designs with biomarkers. non-inferiority trials: minimizing patient exposure
11. Korn EL. Nontoxicity endpoints in phase I trial J Clin Oncol. 2012;30:3304–3309. to inferior therapies. Ann Oncol. 2018;29:573–577.
designs for targeted, non-cytotoxic agents. J Natl 32. Freidlin B, Othis M, Korn EL. Information time 55. Freidlin B, Korn EL, George SL. Data monitoring
Cancer Inst. 2004;96:977–978. scales for interim analysis of randomized clinical committees and interim monitoring guidelines.
12. Hunsberger S, Rubinstein LV, Dancey J, Korn EL. trials. Clin Trials. 2016;13:391–399. Control Clin Trials. 1999;20:395–407.
Dose escalation trial designs based on a molecularly 33. Freidlin B, Korn EL, Gray R, Martin A. Multi-arm 55a.  Korn EL, Freidlin B. Interim futility monitoring
targeted endpoint. Stat Med. 2005;24:2171– clinical trials of new agents: some design consider- assessing immune therapies with a potentially delayed
2181. ations. Clin Cancer Res. 2008;14:4368–4371. treatment effect. J Clin Oncol. 2018; (published
13. Ivy SP, Siu LL, Garrett-Mayer E, Rubinstein L. 34. Redman MW, Allegra CJ. The master protocol online before print).
Approaches to phase 1 clinical trial design focused concept. Semin Oncol. 2015;42:724–730. 56. Bassler D, Briel M, Montori VM, et al. Stopping
on safety, efficiency, and selected patient populations: 35. Green S, Liu PY, O’Sullivan J. Factorial design randomized trials early for benefit and estimation
a report from the clinical design task force of the considerations. J Clin Oncol. 2002;20:3424–3430. of treatment effects. JAMA. 2010;303:1180–1187.
National Cancer Institute Investigational Drug 36. Korn EL, Freidlin B. Non-factorial analyses of two- 57. Freidlin B, Korn EL. Stopping clinical trials early
Steering Committee. Clin Cancer Res. 2010;16: by-two factorial trial designs. Clin Trials. 2016;13: for benefit: impact on estimation. Clin Trials.
1726–1736. 651–659. 2009;6:119–125.
14. Parulekar WR, Eisenhauer EA. Phase 1 trial design 37. James ND, Sydes MR, Clarke NW, et al. Addition 58. Korn EL, Freidlin B, Mooney M. Stopping or
for solid tumor studies of targeted, non-cytotoxic of docetaxel, zoledronic acid, or both to first-line reporting early for positive results in randomized
agents: theory and practice. J Natl Cancer Inst. long-term hormone therapy in prostate cancer clinical trials: the national cancer institute coopera-
2004;96:990–997. (STAMPEDE): survival results from an adap- tive group experience from 1990 to 2005. J Clin
15. Booth CM, Calbert AH, Giaccone G, et al. End- tive, multiarm, multistage, platform randomised Oncol. 2009;27:1712–1721.
points and other considerations in phase I studies of controlled trial. Lancet. 2016;387:1163–1177. 59. Smith MA, Ungerleider RS, Korn EL, Rubinstein
targeted anticancer therapy: recommendations from 38. U. S. Food and Drug Administration. Guidance for L, Simon R. Role of independent data-monitoring
the task force on Methodology for the Development Industry—Non-Inferiority Clinical Trials to Establish committees in randomized clinical trials sponsored by
of Innovative Cancer Therapies (MDICT). Eur J Effectiveness. Silver Spring, MD. 2016. the national cancer institute. J Clin Oncol. 1997;15:
Cancer. 2008;44:19–24. 39. D’Agostino RB, Massaro JM, Sullivan LM. Non- 2736–2743.
16. Murgo AJ, Kummar S, Rubinstein L, et al. inferiority trials: design concepts and issues—the 60. Ellenberg SS, Fleming TR, DeMets DL. Data
Designing phase 0 clinical trials. Clin Cancer Res. encounters of academic consultants in statistics. Stat Monitoring Committees in Clinical Trials. Chichester,
2008;14:3675–3682. Med. 2003;22:169–186. England.: Wiley; 2002.
17. Eisenhauer EA, Therasse P, Bogaerts J, et al. New 40. Freidlin B, Korn EL, George SL, Gray R. Random- 61. Korn EL, Hunsberger S, Freidlin B, Smith MA,
response criteria in solid tumors: revised RECIST ized clinical trial design for assessing noninferior- Abrams JS. Preliminary data release for randomized
guideline (version 1.1). Eur J Cancer. 2009;45: ity when superiority is expected. J Clin Oncol. clinical trials of noninferiority: a new proposal.
228–247. 2007;25:5019–5023. J Clin Oncol. 2005;23:5831–5836.
18. Simon R. Optimal two-stage designs for phase II 41. Johnson JR, Bross P, Cohen M, et al. Approval 62. Byar DP, Simon RM, Friedewald WT, et al. Random-
clinical trials. Control Clin Trials. 1989;10:1–10. summary: imatinib mesylate capsules for treatment ized clinical trials—perspectives on some recent ideas.
19. Jung SH, Lee T, Kim KM, George SL. Admissible of adult patients with newly diagnosed Philadelphia N Engl J Med. 1976;295:74–80.
two-stage designs for phase II cancer clinical trials. chromosome-positive chronic myelogenous leukemia 63. Korn EL, Freidlin B. Outcome-adaptive random-
Stat Med. 2004;23:561–569. in chronic phase. Clin Caner Res. 2003;9:1972–1979. ization: is it useful? J Clin Oncol. 2011;29:771–
20. Korn EL, Arbuck SG, Pluda JM, et al. Clinical trial 42. Schultz KR, Bowman WP, Aledo A, et al. Improved 776.
designs for cytostatic agents: are new approaches early event-free survival with imatinib in Philadelphia 64. Simon R, Simon NR. Using randomization tests
needed? J Clin Oncol. 2001;19:265–272. chromosome-positive acute lymphoblastic leukemia: to preserve type I error with response-adaptive and
21. Korn EL, Liu PY, Lee SJ, et al. Meta-analysis of a Children’s Oncology Group study. J Clin Oncol. covariate-adaptive randomization. Stat Probab Lett.
phase II cooperative group trials in metastatic stage 2009;27:5175–5181. 2011;81:767–772.
IV melanoma to determine progression-free and 43. Hortobagyi GN. What is the role of high-dose 65. Korn EL, Freidlin B. Adaptive clinical trials:
overall survival benchmarks for future phase II trials. chemotherapy in the era of targeted therapies? advantages and disadvantages of various adaptive
J Clin Oncol. 2008;26:527–534. J Clin Oncol. 2004;22:2263–2266. design elements. J Natl Cancer Inst. 2017;109(6).
307.e2 Part I: Science and Clinical Oncology

66. Thall P, Fox P, Wathen J. Statistical controversies 89. Amit O, Mannino F, Stone AM, et al. Blinded 107. Chapman PB, Hauschild A, Robert C, et al. Improved
in clinical research: scientific and ethical problems independent central review of progression in cancer survival with vemurafenib in melanoma with
with adaptive randomization in comparative clinical clinical trials: results from a meta-analysis. Eur J BRAF V600E mutation. N Engl J Med. 2011;364:
trials. Ann Oncol. 2015;26:1621–1628. Cancer. 2011;47:1772–1778. 2507–2516.
67. Hey SP, Kimmelman J. Are outcome-adaptive alloca- 90. Dodd LE, Korn EL, Freidlin B, et al. An audit 108. Simon R, Maitournam A. Evaluating the efficiency
tion trials ethical? Clin Trials. 2015;12:102–106. strategy for progression-free survival. Biometrics. of targeted designs for randomized clinical trials.
68. Berry DA. Commentary on Hey and Kimmelman. 2011;67:1092–1099. Clin Cancer Res. 2004;10:6759–6763.
Clin Trials. 2015;12:107–109. 91. Freidlin B, Korn EL, Hunsberger S, et al. Proposal 109. Freidlin B, McShane LM, Korn EL. Randomized
69. Lee JJ. Commentary on Hey and Kimmelman. Clin for the use of progression-free survival in unblended clinical trials with biomarkers: design issues. J Natl
Trials. 2015;12:110–112. trials. J Clin Oncol. 2007;25:2122–2126. Cancer Inst. 2010;102:152–160.
70. Saxman SB. Commentary on Hey and Kimmelman. 92. Freidlin B, Little RF, Korn EL. Design issues in 110. Rothmann MD, Zhang JJ, Fleming TR. Testing
Clin Trials. 2015;12:113–115. randomized clinical trials of maintenance therapies. in a prespecified subgroup and the intent-to-treat
71. Joffe S, Ellenberg SS. Commentary on Hey and J Natl Cancer Inst. 2015;107(11):djv225. population. Drug Inf J. 2012;46:175–179.
Kimmelman. Clin Trials. 2015;12:116–118. 93. Basch E, Trentacosti AM, Burke LB, et al. Pain 111. Freidlin B, Sun Z, Gray R, Korn EL. Phase III
72. Buyse M. Commentary on Hey and Kimmelman. palliation measurement in cancer clinical trials. clinical trials that integrate treatment and biomarker
Clin Trials. 2015;12:119–121. Cancer. 2014;120:761–767. evaluation. J Clin Oncol. 2013;31:3158–3161.
73. Korn EL, Freidlin B. Commentary on Hey and 94. U. S. Food and Drug Administration. Guidance 112. Freidlin B, Korn EL, Gray R. Marker sequential
Kimmelman. Clin Trials. 2015;12:122–124. for Industry. Patient-Reported Outome Measures: test (MaST) design. Clin Trials. 2014;11:19–27.
74. Hey SP, Kimmelman J. Rejoinder. Clin Trials. Use in Medical Product Development to Support 113. Sparano JA, Paik S. Development of the 21-gene
2015;12:125–127. Labeling Claims. Rockville, MD. 2009. assay and its application in clinical practice and
75. Pocock SJ. Interim analyses for randomized clinical 95. Basch E, Reeve BB, Mitchell SA, et al. Development clinical trials. J Clin Oncol. 2008;26:721–728.
trials: the group sequential approach. Biometrics. of the national cancer institute’s patient-reported 114. Sparano JA, Gray RJ, Makower DF, et al. Prospective
1982;38:153–162. outcomes version of the common terminology validation of a 21-gene expression assay in breast
76. Ivanova A, Qaqish BF, Schell MJ. Continuous criteria for adverse events (PRO-CTCAE). J Natl cancer. N Engl J Med. 2015;373:2005–2014.
toxicity monitoring in phase II trials in oncol- Cancer Inst. 2014;106:dju244. 115. Herbst RS, Baas P, Kim DW, et al. Pembrolizumab
ogy. Biometrics. 2005;61:540–545. 96. Forastiere AA, Zhang Q, Weber RS, Maor MH, versus docetaxel for previously treated, PD-l1-positive,
77. Bretz F, Schmidli H, Konig F, Racine A, Maurer W. Goepfert H, et al. Long-term results of RTOG advanced non-small-cell lung cancer (KEYNOTE-
Confirmatory seamless phase II/III clinical trials with 99-11: a comparison of three nonsurgical treat- 010): a randomized controlled trial. Lancet. 2016;
hypotheses selection at interim: general concepts. ment strategies to preserve the larynx in patients 387:1540–1550.
Biom J. 2006;48:623–634. with locally advanced larynx cancer. J Clin Oncol. 116. Jiang W, Freidlin B, Simon R. Biomarker-adaptive
78. Korn EL, Freidlin B, Abrams JS, Halabi S. Design 2013;31:845–852. threshold design: a procedure for evaluating treat-
issues in randomized phase II/III trials. J Clin Oncol. 97. Korn EL, Albert PS, McShane LM. Assessing sur- ment with possible biomarker-defined subset effect.
2012;30:667–671. rogates as trial endpoints using mixed models. Stat J Natl Cancer Inst. 2007;99:1036–1043.
78a.  Freidlin B, Korn EL, Abrams JS. Bias, operational Med. 2005;24:163–182. 117. Simon R. Genomic alteration-driven clinical trial
bias, and generalizability in phase II/III trials. J Clin 98. Sargent DJ, Wieand HS, Haller DG, et al. Disease- designs in oncology. Ann Intern Med. 2016;165:
Oncol. 2018;36:1902–1904. free survival versus overall survival as a primary end 270–278.
79. Parmar MKB, Barthel FMS, Sydes M, et al. Speeding point for adjuvant colon cancer studies: individual 118. Govindan R, Mandrekar SJ, Gerber DE, et al.
the evaluation of new agents in cancer. J Natl Cancer patients data from 20,898 patients on 18 randomized ALCHEMIST trials: a golden opportunity to
Inst. 2008;100:1204–1214. trials. J Clin Oncol. 2005;23:8664–8670. transform outcomes in early-stage non-small cell
80. U. S. Food and Drug Administration. Draft guidance 99. Korn EL, Sachs MC, McShane LM. Statistical lung cancer. Clin Cancer Res. 2015;21:5439–5444.
for industry—adaptive design clinical trials for drugs controversies in clinical research: assessing pathologic 119. Kaplan R, Maughan T, Crook A, et al. Evaluating
and biologics. Rockville, MD. 2010. complete response as a trial-level surrogate end point many treatments and biomarkers in oncology: a
81. Emerson SS, Fleming TR. Adaptive methods: for early-stage breast cancer. Ann Oncol. 2016;27: new design. J Clin Oncol. 2013;31:4562–4568.
telling “the rest of the story. J Biopharm Stat. 10–15. 120. Herbst RS, Gandara DR, Hirsch FR, et al. Lung
2010;20:1150–1165. 99a.  Korn EL, Freidlin B. Surrogate and intermediate master protocol (Lung-MAP)—a biomarker-driven
82. Cuffe RL, Lawrence D, Stone A, Vandemeulebroecke endpoints in randomized trials: what’s the goal? Clin protocol for accelerating development of therapies
M. When is a seamless study desirable? Case studies Cancer Res. 2018;24:2239–2240. for squamous cell lung cancer: SWOG s1400. Clin
from different pharmaceutical sponsors. Pharm Stat. 100. Amado RG, Wolf M, Peters M, et al. Wild-type Cancer Res. 2015;21:1514–1524.
2014;13:229–237. KRAS is required for panitumumab efficacy in 121. Conley BA, Doroshow JH. Molecular analysis
83. Temple R. Are surrogate markers adequate to assess patients with metastatic colorectal cancer. J Clin for therapy choice: NCI MATCH. Semin Oncol.
cardiovascular disease drugs? JAMA. 1999;282: Oncol. 2008;26:1626–1634. 2014;41:297–299.
290–795. 101. Mok TS, Wu YL, Thongprasert S, et al. Gefitinib or 122. Kopetz S, Desai J, Chan E, et al. PLX4032 in
84. Freidlin B, Abrams J, Korn EL. New challenges carboplatin-paclitaxel in pulmonary adenocarcinoma. metastatic colorectal cancer patients with mutant
for comparative effectiveness in oncology: choice of N Engl J Med. 2009;361:947–957. BRAF tumors. J Clin Oncol. 2010;28:3534. abstract.
primary end points for randomized clinical trials. 102. Peto R. Statistical aspects of clinical trials. In: Halnan 123. Hyman DM, Puzanov I, Subbiah V, et al. Vemu-
J Comp Eff Res. 2013;2:469–481. KE, ed. Treatment of Cancer. London: Chapman rafenib in multiple nonmelanoma cancers with BRAF
85. Korn EL, Freidlin B, Abrams J. Overall survival and Hall; 1982. v600 mutations. N Engl J Med. 2015;373:726–
as the outcome for randomized clinical trials 103. Polley MY, Freidlin B, Korn EL, et al. Statistical 736.
with effective subsequent therapies. J Clin Oncol. and practical considerations for clinical evalua- 124. Le Tourneau C, Delord JP, Goncalves A, et al.
2011;29:2439–2442. tion of predictive biomarkers. J Natl Cancer Inst. Molecularly targeted therapy based on tumour
86. Fleming TR, Rothmann MD, Lu HL. Issues in using 2013;105:1677–1683. molecular profiling versus conventional therapy for
progression-free survival when evaluating oncology 104. Simon RM, Paik S, Hayes DF. Use of archived advanced cancer (SHIVA): a multicenter, open-label,
products. J Clin Oncol. 2009;27:2874–2880. specimens in evaluation of prognostic and predictive proof-of-concept, randomised, controlled phase 2
87. U. S. Food and Drug Administration. Guidance for biomarkers. J Natl Cancer Inst. 2009;21:1446– trial. Lancet Oncol. 2015;16:1324–1334.
Industry. Clinical Trial Endpoints for the Approval 1452. 125. Kim ES, Herbst RS, Wistuba II, et al. The BATTLE
of Cancer Drugs and Biologics. Rockville, MD. 105. Freidlin B, Korn EL. Biomarker enrichment strate- trial: personalizing therapy for lung cancer. Cancer
2007. gies: matching trial design to biomarker credentials. Discov. 2011;1:44–53.
88. Dodd LE, Korn EL, Freidlin B, et al. Blinded inde- Nat Rev Clin Oncol. 2014;11:81–90. 126. Papadimitrakopoulou V, Lee JJ, Wistuba II, et al. The
pendent central review of progression-free survival 106. Clark GM, McShane LM. Biostatistical consider- BATTLE-2 study: a biomarker-integrated targeted
in phase III clinical trials: important design element ations in development of biomarkers-based tests therapy study in previously treated patients with
or unnecessary expense? J Clin Oncol. 2008;26: to guide treatment decisions. Stat Biopharm Res. advanced non-small-cell lung cancer. J Clin Oncol.
379–3796. 2011;3:549–560. 2016;34:3638–3647.
19  Structures Supporting Cancer Clinical Trials
Jeffrey S. Abrams, Margaret Mooney, James A. Zwiebel,
Worta McCaskill-Stevens, Michaele C. Christian, and James H. Doroshow

S UMMARY OF K EY P OI N T S
• Cancer clinical trials provide the • Successful execution of clinical trials ○ Access to an authorized
evidence on which sound oncology includes the following clinical institutional review board
practice is based. components: ○ Access to adequate laboratory
• Providing greater efficiency in ○ The attitude and commitment of facilities to process
implementing clinical trials and the physician leader (medical, protocol-required specimens
achieving enrollment rapidly have surgical, radiation oncologist) ○ Adherence to good clinical
been major goals of the National ○ Sufficient preparation and practices
Cancer Institute (NCI), cancer centers, infrastructure ○ Accurate and timely data reporting
the biopharmaceutical industry, and ○ Trained staff, including at least ○ Proper maintenance of primary
patient advocacy groups. some of the following: clinical source documentation
• Oncology specialists in private research nurse, clinical research ○ Adequate preparation for trial
practice have many opportunities to associate, pharmacist, pathologist, monitoring and on-site audits
participate in clinical trials sponsored radiologist ○ Adequate financial support
by the NCI, private foundations, and/ ○ Affiliation with an institution or • Many organizations now provide
or the biopharmaceutical industry. network that provides scientific and access to clinical trials and/or
• Physicians and patients have online administrative support, such as the provide the necessary training and
access to all US-sponsored clinical following: NCI-supported network certification; relevant Web sites are
trials at ClinicalTrials.gov and have group, cancer center network, included in this chapter.
access to additional information biopharmaceutical industry
about cancer trials at cancer.gov. network, research institution

Modern oncology practice is founded on results from thousands of made participation very challenging. Fortunately, because of intense
clinical trials conducted during the past four decades. Thousands publicity and educational programs by both patient advocacy groups
more clinical trials are ongoing at any given time and provide the and clinical trial organizations and widespread access to clinical trial
evidence base for the rapidly changing therapeutic practices of this information on the Internet, a growing number of patients now expect
specialty. Motivations for the decision by an oncologist to participate that clinical trials will be included in the discussion of options for the
actively in this extensive system of medical and scientific inquiry range treatment of their cancers. The purpose of this chapter is to describe
from the ability to offer patients state-of-the-art treatments, which are some of the requirements, resources, and structures that are available
available through well-designed clinical trials, to the personal satisfaction to enable practicing oncologists to participate in clinical trials and to
and educational benefits that can be achieved from participation in discuss the responsibilities that come with such participation. Numerous
this process. The commitment of time and resources necessary to opportunities now exist for practicing physicians and their patients to
participate effectively in clinical research, and sometimes unfamiliarity participate in cancer clinical trials, including treatment, prevention,
with clinical research requirements and procedures, prevent many and cancer control trials, supported by the National Cancer Institute
oncologists from taking part. It has been estimated that only 3% (NCI), cancer treatment institutions, or the biopharmaceutical industry.
to 5% of adults with cancer in the United States are treated while
taking part in clinical trials, with even lower rates of participation NATIONAL CANCER INSTITUTE–SPONSORED
in many other countries. In stark contrast, approximately two-thirds CLINICAL TRIAL ACTIVITIES
of children with cancer are enrolled in clinical trials. Although this
discrepancy exists for multiple reasons, a major factor relates to the The NCI supports the development of more than 100 agents for
relative rarity of cancer in children (approximately 10,500 cases per year cancer treatment and prevention (often in collaboration with phar-
in children younger than 15 years), resulting in substantial centralization maceutical and biotechnology companies) and has an extensive clinical
of pediatric care at major academic research centers, a culture in which trial system that encompasses treatment, prevention, and cancer control
participation in trials is supported and fostered. Care for adults with studies. More than 800 trials are active at any given time, and several
cancer in the United States is more decentralized, with many adult hundred new trials open each year. In the treatment area, the NCI
oncologists working in private practices that are not tied to academic has programs for early therapeutic development (primarily phase
institutions. For these practitioners, inadequate understanding of the I and II trials), including many sites with grants to complete these
clinical trial process plus pressures on time and reimbursement have early trials.

308
Structures Supporting Cancer Clinical Trials  •  CHAPTER 19 309

support only the late-phase trials,7 the CTSU and CIRB have been
Table 19.1  Barriers to Clinical Trial Participation expanded to now foster efficiency and networking for all of NCI’s
Physician Related Patient Related network early- and late-phase trials, as described subsequently.
Inadequate funding for data The patient’s doctor never
management personnel discussed or offered the option of Cancer Trials Support Unit
participating in a trial The NCI CTSU is designed to facilitate one-stop online access to a
Burdensome regulatory Unaware of trials as an option broad menu of clinical trials and selected international collaborative
requirements Concerns about insurance coverage trials by a national network of investigators who are members of
Institutional review board Fear of receiving placebo NCI-supported clinical trial programs. Network investigators can access
Informed consent Many patients have comorbidities the CTSU menu of treatment trials from a public website (www.ctsu.org).
Conflict of interest The scientific leadership for each study remains within the organization
that developed the trial, but patient enrollment can come from any
Overly strict eligibility criteria
network physician across the country, and all patient enrollment is
Inadequate reimbursement
handled through CTSU’s central registration system, the Oncology
Lack of time Patient Enrollment Network (OPEN). By providing more physicians
Resistance by third-party payers and their patients with the opportunity to choose from a broader
menu of trials, the CTSU promotes faster accrual to individual trials,
allows increased access and additional treatment options to more
For several decades the NCI also supported a large program of patients nationwide, and renders trials involving uncommon cancers
clinical trial cooperative groups to provide a standing mechanism for more feasible. Although the clinical trials menu and centralized patient
performing late-phase, definitive, clinical treatment trials. Although enrollment are the most visible aspects of the CTSU, another major
these cooperative group trials provided a significant proportion of the function of the CTSU is its centralized regulatory database. For all
evidence on which oncology practice is based, public advocacy for physicians in the network, the CTSU maintains important demographic
more rapid progress, increasing fiscal pressures on medical practice, information about their sites or practices, including clinical trial program
and the accelerated pace of drug discovery, especially in the new era affiliation and academic and practice affiliation(s), Office for Human
of precision medicine, caused the NCI to review and restructure aspects Research Protections assurance numbers for their sites, institutional
of its late-phase clinical trial program. Surveys of patients and physicians review board (IRB) approvals for specific protocols, and conflict of
found that the obstacles to accrual in oncology trials were multifactorial interest forms for investigators. This process enables physicians, nurses,
(Table 19.1).1–5 With these barriers in mind, the NCI undertook a and clinical research associates to register once annually instead of
series of comprehensive analyses of how it conducts and funds clinical having to register for each clinical trial program or trial in which they
trials by involving a wide range of stakeholders that included leaders participate. Centralizing regulatory data has reduced the workload
of the Cooperative Group and Cancer Center Programs, patient for investigators in the field, consolidated duplicative work, and allowed
advocates, representatives from the US Food and Drug Administration clinical trial program staff to partially offload this activity to the CTSU
(FDA) and the pharmaceutical industry, and government staff, including and focus instead on protocol development and analysis.
the analysis described in the 2010 Institute of Medicine (IOM) report.6
Based on these reviews and analyses, the NCI transformed its Central Institutional Review Board
long-standing Cooperative Group Clinical Trials program in 2014
into a more highly integrated network. By rebranding the infrastructure In NCI-sponsored multicenter trials, the identical protocol is carried
as the National Clinical Trials Network (NCTN), reducing the number out at many sites, often including as many as 100 different practices;
of groups, and changing the review criteria for funding to emphasize each site requires its own local institutional review board (LIRB) to
collaboration and coordination, the NCI has refocused its research conduct an initial full-board review and subsequent annual reviews,
effort. Competition among the remaining groups has been reduced adverse event reviews, and amendment reviews. These multiple IRB
and replaced with more efficient and rapid development of a menu reviews create a largely redundant, time-consuming workload at these
of Network trials in which all group members are encouraged to sites, compounding the ever-mounting pressures on the nation’s IRB
participate in order to facilitate speedy enrollment. system, which have been well documented.8 To provide an idea of
The development of targeted therapies directed against specific the scope of the duplicative effort, consider that NCI currently has
molecular alterations identified in various cancers and the emergence more than 19,000 registered investigators participating in all its clinical
of successful immunotherapeutic approaches have fundamentally trial programs at more than 2700 sites. Under the former NCI-
changed the approach to cancer treatment and have introduced new sponsored Cooperative Group Program, for example, there were about
challenges to conducting clinical trials. Identification of patient 160 ongoing phase III trials and 30 new trials entering the NCI
populations that can potentially benefit from these new therapies program annually, resulting in approximately 16,000 IRB reviews
often requires clinical trials that screen large numbers of patients for (3000 initial reviews) conducted each year.8 In addition, investigators
specific molecular alterations. With its state-of-the-art clinical trial often mention that the amount of time, paperwork, and funding
infrastructure, the NCTN is well positioned to implement and complete required for them to obtain IRB approval is a serious barrier to opening
trials far more rapidly than in the past. The NCTN has streamlined trials. These factors provided the impetus for the NCI to develop the
administrative and regulatory processes related to the conduct of its CIRB and centralize the approach to human subject protection for
clinical trials, including patient enrollment, data management, ethics its clinical trial programs.
review, and tumor banking processes, and has provided a common In late 2012, the NCI CIRB initiative began to transition from
menu of trials for all academic and community member sites in the its historic “facilitated review” model to an independent model in
NCTN program. which the NCI CIRB is the sole IRB of Record responsible for both
Critical to the achievement of a tightly integrated trials network study review and local context considerations for enrolled institutions.
are the Cancer Trials Support Unit (CTSU) and the Central Institutional The primary reason for this model change was to further streamline
Review Board (CIRB). The CTSU provides an online menu of all the IRB review process, increasing efficiency of implementation and
trials conducted by the various NCTN groups. The CIRB provides conduct of multisite clinical trials and promoting consistency of ethics
a single, expert ethics review of all NCTN trials, avoiding repetitive reviews. On December 13, 2012, the Association for the Accreditation
costly reviews at individual sites. Although originally conceived to of Human Research Protection Programs (AAHRPP) awarded
310 Part I: Science and Clinical Oncology

accreditation to the NCI CIRB under its independent model, the participation by practicing oncologists, advocates, and translational
first National Institutes of Health (NIH) entity to earn this distinction. scientists, a number of other important mechanisms provide additional
With its adoption of the independent model, the NCI CIRB program options for support and participation.
was a forerunner of the new policy of the NIH requiring US centers
participating in NIH-funded nonexempt, multicenter studies to use National Cancer Institute Community Oncology
a single IRB for initial and ongoing ethics review, a policy that will Research Program
be implemented starting in 2018.9
The NCI CIRB program provides a centralized approach to human The NCI Community Oncology Research Program (NCORP) was
subject protection. The program currently consists of four boards: established in 2014 to serve as NCI’s single community-based research
one primarily for late-phase clinical trials, including all adult oncology program. NCORP was developed to build on the progress made in
phase III treatment trials; an early-phase clinical trial board; a pediatric NCI’s previously supported community-based programs, the NCI’s
clinical trial board; and a board covering cancer control and prevention. Community Clinical Oncology Program (CCOP) network, the
The NCI CIRBs are composed of distinguished panels of oncology Minority-Based CCOPs, Research Bases, and the NCI Community
physicians, nurses, and patient representatives and include a pharmacist, Cancer Centers Program (NCCCP). NCI recognized the need to
an ethicist, and a lawyer. Unlike most local IRBs, the NCI CIRBs respond to the IOM’s report6 to address efficiency in the conduct of
are focused exclusively on cancer trials and have sufficient time and clinical trials and to respond to the dynamic health care environment
expertise to review each protocol in detail. This model for human and challenges faced by today’s practicing community oncologists.
subject protection in multicenter trials is now used by more than NCORP is an integrated national network to (1) design and conduct
1700 sites, including NCI-designated Cancer Centers, other university cancer prevention, control, and screening trials, and quality-of-life
medical centers, and community hospitals. studies embedded in treatment trials; (2) expand its research scope to
design and conduct cancer care delivery research; (3) enhance patient
National Cancer Institute National Clinical and provider access to treatment and imaging clinical trials conducted
Trials Network under the reorganized NCTN; and (4) integrate disparity research
questions into clinical trials and cancer care delivery research. The
The NCTN Program was conceived and developed so that it could overarching goal of NCORP is to provide access to clinical trials and
operate as a true network with incentives for collaboration in the cancer care delivery research to individuals in their own communities,
development and conduct of innovative clinical trials. These trials generating evidence that contributes to improved patient outcomes
prioritize precision medicine approaches, rare cancers, rare subsets of and a reduction in cancer disparities. NCORP preserved a three-
more common cancers, multimodality therapies, special populations, component organizational structure, as a partnership between academic
and research questions unlikely to be addressed in the private sector. and community practices. It is composed of 34 community sites, 12
In the NCTN Program, network groups are expected to collaborate minority/underserved sites, and 7 research bases. Community sites
with one another and with NCI to achieve the overall goal of the accrue patients or participants to NCI-approved clinical trials and
Program. Member institutions or sites of any network group are able cancer care delivery research designed by NCORP research bases and
to enroll patients in all adult phase III and phase II/III trials, as well NCI’s NCTN groups. Minority/underserved sites also accrue patients
as most early-phase trials and trials in adolescents and young adults, and participants from catchment areas in which at least 30% of their
irrespective of the specific network group that is leading the trial. The rural resident populations are members of racial or ethnic minorities.
NCTN Program, including each of its six key components, is illustrated Five of the research bases are funded through the NCORP cooperative
in Fig. 19.1. funding mechanism and have the same consolidated configuration as
One of the key aims of the NCTN when it was launched in 2014 the NCTN groups. Two additional NCORP research bases are non-
was to provide for the conduct of precision medicine clinical trials NCTN, academic institutions.
by harnessing the scientific leadership of the NCTN investigators, NCORP is a diverse network of practices ranging from single
leveraging the Network to screen large numbers of patients to identify practices, integrated and nonintegrated health systems, safety-net
those whose tumors exhibit molecular features that may be responsive hospitals, and academic institutions. Fifteen states with greater than
to new, targeted treatments and/or immunotherapy, and fostering 30% rural populations have NCORP components.10 The NCORP
clinical research partnerships with biotechnology and pharmaceutical grants provide up-front funding to enable sites to develop a standing
companies to collaborate on a series of unique precision medicine infrastructure to support patient accrual to network trials. Sites must
trials in oncology. NCI’s Molecular Analysis for Therapy Choice document the ability to enroll at least 80 patients or individuals per
(NCI-MATCH) is the archetype of a precision medicine trial. It is year in cancer control, prevention, screening, and treatment. The
led by ECOG-ACRIN (one of the NCTN groups) and has active program is housed in the NCI Division of Cancer Prevention, but
participation by member sites belonging to any group within the collaborates closely with both the NCI Division of Cancer Control
NCTN. NCI-MATCH opened to patient enrollment in 2015 and and Population Sciences that coordinates the cancer care delivery
includes adult patients with any type of solid tumor or lymphoma research portfolio and the NCI Division of Cancer Treatment and
whose disease is no longer responding to standard therapy. By the Diagnosis that oversees the cancer treatment clinical trials. The NCI
end of 2017, over 6000 patients had their tumors screened for molecular Center to Reduce Health Disparities also plays an important role
alterations and approximately 1000 were “matched” to one of a series across all NCORP programs.
of small phase II trials that target a specific mutation in their tumor. In 2017, 4065 physicians and 421 hospitals participated in NCORP.
Although NCI-MATCH is led by ECOG-ACRIN, investigators from Forty-six community sites and minority/underserved sites have 860
all the NCTN groups lead the individual phase II trials, illustrating associated component and subcomponent sites throughout the United
the collaborative aspect of the NCTN. States and Puerto Rico. NCORP sites accrued more than 12,000
patients and participants to cancer treatment, cancer prevention, and
cancer control trials during the first 2 years of the program.
OTHER NATIONAL CANCER INSTITUTE– With its diverse populations, NCORP’s network is a resource suitable
SPONSORED STRUCTURES SUPPORTING as a laboratory for cancer health disparities research. It can serve as
CLINICAL TRIALS proof-of-principle for the conduct of precision medicine trials and the
infrastructure requirements for success outside of academic institutions;
Although the CTSU, the CIRB, and the restructured NCTN represent and it is a resource for the collection of patient-reported symptom
mechanisms to reduce barriers and facilitate broader clinical trial toxicities from cancer and cancer treatments. The incorporation
Structures Supporting Cancer Clinical Trials  •  CHAPTER 19 311

Figure 19.1  •  National Cancer Institute (NCI) National Clinical Trials Network (NCTN) structure. The key components of the NCTN are completely
integrated into the other initiatives and components of the NCI clinical trial system, including the NCI Cancer Trials Support Unit, NCI Central Institutional
Review Boards, other NCI-funded programs such as the NCI Community Oncology Research Program (NCORP) and the Minority/Underserved NCORP
as well as NCI advisory and review committees (not illustrated in the figure), which help oversee the entire program. The six major components are (1)
Network Group Operations Centers—four adult groups (Alliance, ECOG-ACRIN, NRG Oncology, and SWOG) and one pediatric group (Children’s Oncology
Group [COG])—which provide scientific leadership for developing and implementing multidisciplinary, multiinstitutional trials in a range of diseases and
special populations with specific scientific strategy and goals; (2) Network Group Statistics and Data Management Centers, associated with specific Network
Group Operations Centers, which provide the statistical expertise required to ensure effective scientific design and conduct of clinical trials, in addition to
data management, data analysis, which provide support for leadership and expertise to facilitate incorporation of translational science into network group
clinical trials; (4) Network Lead Academic Participating Sites, which provide scientific leadership in development and conduct of clinical trials in association
with one or more adult network groups, as well as substantial accrual to clinical trials conducted across the entire NCTN; (5) Network Radiotherapy and
Imaging Core Services Center, which provides scientific and technical expertise for incorporation of appropriate, integrated quality assurance and image data
management for applicable clinical trials conducted by the NCTN that require specialized quality assurance or imaging data management and/or assessment;
and the (6) Canadian Collaborating Clinical Trial Network (Canadian Cancer Trials Group), which will be capable of being a full partner with the US Network
in conducting large-scale, multisite clinical trials. In addition, all NCTN Groups use a Common Data Management System to conduct NCTN trials.

of cancer screening and reinvigoration of cancer prevention in the more than 30 NCI-designated US Comprehensive Cancer Centers
community setting has enhanced the partnerships with nononcology and leading institutions in Canada, have enrolled approximately 2000
specialists and primary care physicians both within and affiliated with patients per year in clinical trials, many of which include novel
NCORP.11 The introduction of cancer care delivery research into combinations of investigational agents. A major feature of this network
community-based practices enables investigators to study the provider, is the expertise of the investigators and their institutions in tumor
the system, and the patient influences on cancer care and outcomes, sample acquisition, pharmacokinetic assay development and monitoring,
a timely topic in view of the national effort to improve the quality and the development of biomarker assays.
of routine medical care in the United States. In keeping with the many new scientific opportunities afforded by
Details on NCORP can be found at the NCI Division of Cancer advances in molecular diagnostics, the NCI Experimental Therapeutics
Prevention website at https://ncorp.cancer.gov. Clinical Trials Network (ETCTN) has been restructured to facilitate the
exploration of novel targeted agents in appropriately selected patients.
Phase I and II Early Therapeutic Clinical This restructuring involves a number of changes, including:
Trials Networks
• Integration of NCI’s funded tumor biology and translational science
The NCI has long supported a network of individual academic programs (e.g., the NCI-designated Cancer Centers and the Special-
institutions and academic consortia to conduct phase I and early phase ized Programs of Research Excellence) with clinical investigators
II clinical trials, often performed with pharmacokinetic and correlative managing the ETCTN. This integration has included the formation
pharmacodynamic studies. These institutions, currently comprising of interdisciplinary project teams (see later) for investigational agent
312 Part I: Science and Clinical Oncology

development. These project teams bring together the necessary pharmaceutical industry. In part, this lack of participation was a
expertise to both formulate key questions and execute clinical trials reflection of the complexity of therapeutic development during a
in the development of specific investigational agents. period when an insufficiently detailed understanding of cancer biology
• Support for laboratories with validated assays that will carry out made rational drug development difficult. This relative lack of industry
the molecular characterization of patient tumors both before and involvement, coupled with the significant public health problem
after treatment. presented by cancer, was responsible for the development of the large
• Centralized regulatory, data management, and pharmacologic core NCI-sponsored clinical trial apparatus described in detail in this chapter.
functions for the ETCTN, making multisite studies easier to perform During the past 30 years, however, a remarkable increase in biophar-
even in the earliest stages of drug development. maceutical investment in cancer drug discovery research has occurred,
• Extension of the NCI CIRB to these early-phase multicenter trials along with a parallel increase in both the extent and sophistication
by establishment of a new review board with the requisite expertise of industry sponsorship of clinical trials for the development of new
to focus on early-phase clinical trials. cancer therapeutics. Hundreds of new agents are currently in different
• Integration of phase I and phase II clinical trials under a single grant stages of clinical evaluation by industry, either alone or in cooperation
structure, enabling ETCTN awardees to have the flexibility to expand with the NCI or similar noncommercial organizations in the United
phase I studies quickly on detection of early activity signals. States and in other parts of the world.
• Inclusion of investigators from NCI-designated Cancer Centers
that are not affiliated with the ETCTN who may now submit Purpose and Nature of Industry-Sponsored
clinical trial proposals, and, if approved, receive full ETCTN support Clinical Trials
for the clinical trial.
The primary goal of therapeutic agent investigation by the biophar-
The overall intent of these changes is the creation of an early maceutical industry is the evaluation of promising agents for eventual
clinical trial network that is integrated both scientifically and operation- registration and commercialization. Because registration of a new cancer
ally to pursue the development of novel therapeutics. drug requires the demonstration of safety and efficacy for the new
agent in the context of currently available therapy for the cancer being
National Cancer Institute Drug Development treated, industry tends to focus its efforts on so-called explanatory
Project Teams trials.12 Explanatory trials attempt to isolate the benefits and risks of
the investigational agent(s) under ideal study circumstances (efficacy
The NCI extramural program now has a single pipeline to review studies), whereas so-called pragmatic trials are more often conducted
and approve the development of high-priority agents, originating from by NCI or other non–commercially supported networks. Pragmatic
industry or academia. Once vetted via this single entry process, called trials, which can include multimodality treatments, ask which treatment
NExT (NCI Experimental Therapeutics Program), NCI’s Cancer approach is best in a representative sample of patients (effectiveness
Therapy Evaluation Program (CTEP) convenes a Drug Development studies). However, the distinction between the types of trials is often
Project Team. The Project Team members are selected based on their blurred because a long tradition exists of industry providing investi-
qualifications and the expertise they provide. The team members will gational agents for the conduct of clinical studies through NCI-
determine which clinical trials will be conducted across the NCI sponsored mechanisms, and many new agents or indications have
CTEP clinical trial network sites, and how best to approach critical received FDA approval on the basis of NCI-sponsored clinical trials.
translational studies. Ethical considerations regarding human investigation and expectations
Extramural investigators on the Project Teams may fill one or more of adherence to standards for the conduct of clinical trials do not
of the following roles: differ substantially between NCI-sponsored and industry-sponsored
clinical trials; however, NCI-sponsored studies must adhere to the
• Clinician scientists who will lead the clinical trials recommended
US Code of Federal Regulations (45 CFR Part 46), whereas trials of
by the NCI Drug Development Project Team and who will create
investigational drugs or devices that fall under the FDA’s purview
clinical protocols for execution of these studies.
must comply with the regulations in 21 CFR. When the NCI sponsors
• Translational scientists who will provide guidance on prioritization
trials with new agents or devices, then both federal codes (along with
of biomarkers for the studies under development, including recom-
their respective guidances and policies) are applicable, and any dif-
mendations for technologies and platforms that meet increasingly
ferences between them require the sponsor to adhere to the most
stringent requirements for integral and integrated biomarkers.
stringent aspects of the regulations.
• Basic scientists who will provide scientific guidance for the study
design based on the mechanism of action of the investigational
agent and who will help prioritize the clinical study choices based Particular Characteristics of
on published literature and unpublished data. Basic scientists on Industry-Sponsored Trials
the team will have access to the agents in order to conduct additional
Biopharmaceutical development proceeds in a heavily regulated
laboratory studies in support of the clinical trials.
environment, with detailed regulations from various government
Once convened, the NCI Drug Development Project Team meets agencies covering a spectrum of activities ranging from those related
frequently over a 6- to 8-week period to finalize a comprehensive drug to preparing an agent for first entry into humans, through the years
development plan of biomarker-rich, preclinical and clinical trial of clinical investigation in patients, to postapproval restrictions on
proposals. Once the Project Team’s development plan has been approved, public discussion regarding possible uses of the agent for indications
CTEP will also make the agent available to other qualified investigators, other than those for which the drug was approved. As a result of the
contingent on the company or academic collaborator’s approval. These requirements for working in such an environment, corporate clinical
investigators must submit Letters of Intent (LOIs) to conduct clinical investigations tend to be focused on the “clinical development plan,”
trials, or they can also request agents for preclinical studies. the specific set of clinical trials that will produce an appropriate
evidentiary base to allow for regulatory review of the safety and efficacy
BIOPHARMACEUTICAL INDUSTRY– profile of the agent. During the investigational phase of an agent’s
SPONSORED CANCER CLINICAL TRIALS life cycle, therefore, companies might restrict the general availability
of the agent to individual investigators for clinical study. This perceived
During the first three decades of the development of the field of need for containment and control can lead to tension between the
medical oncology, there was relatively little participation by the investigative community and the industrial sponsor. Other potential
Structures Supporting Cancer Clinical Trials  •  CHAPTER 19 313

sources of tension in the interaction between companies and clinical in the case of academic centers. Clinical trial contract budgets have
investigators relate to the investigators’ perceptions of the need for included direct trial-related costs such as performing additional labora-
independence and objectivity in the conduct of multicenter trials. tory studies that are not being done as part of usual medical care plus
Historically, for example, many companies have generated phase III direct site-related costs associated with the time spent on the trial by
protocols internally, although usually with considerable input from the various participating staff, and indirect costs for institutional
both external advisors and regulatory bodies. These trials would be overhead. Recent years have seen the emergence of consortia of
conceived in whole or part by company personnel, and conduct of investigators (sometimes under the rubric of a site management
the trials would occur in a group of institutions, usually academic or organization) or consortia of institutions presenting themselves to
community-based or both (most often selected on the basis of the companies as clinical trial entities, often linked by a centralized IRB,
sites’ accrual track record), and authorship would be conferred on the offering the advantage of working under a single, negotiated contract.
principal investigator of the largest accruing site. Increasingly, a new In addition, individual academic centers have sometimes formed
model has emerged in which a recognized expert is appointed as the networks of oncologists within their referral area for the purpose of
principal investigator. This individual has a much greater role in the presenting themselves as more efficient entities for interaction.
trial design and the monitoring and eventual analysis and publication
of the trial results than might have been the case historically. Similarly, CHANGING NATURE OF ONCOLOGY TRIALS:
in recent years, the almost universal adoption of independent Data IMPACT ON INFRASTRUCTURE
and Safety Monitoring Committees for late-stage clinical trials has
occurred to oversee safety-related information as it emerges from the The same explosion in our understanding of cancer biology that led
ongoing trial and to make recommendations to company staff about to the increase in the number of new agents under development
appropriate actions. brought with it a realization that the most appropriate tests of those
biologically targeted agents are clinical trials in which the patients
Impact of Globalization on who participate have tumors that are biologically appropriate for the
Pharmaceutical Development agent. For example, imatinib administered to all newly diagnosed
patients with any form of leukemia would have a response rate much
Because pharmaceutical products increasingly are marketed globally, lower than that observed in newly diagnosed patients with chronic
large multinational pharmaceutical companies need to conduct myelogenous leukemia; selection of patients with chronic myelogenous
clinical development from a global perspective. Clinical trials with leukemia with the BCR-ABL mutation allowed for a focused develop-
investigational anticancer agents involving the FDA are being conducted ment program for imatinib that led to rapid initial registration. The
around the world. This tendency toward global development has been same considerations can be extended to matching biologically directed
greatly accelerated by the International Council for Harmonization agents with specific cancer patient populations and argues strongly
process, which facilitated standardization of many of the activities for more complete biologic characterization and monitoring of patients
that are involved in preparing agents for clinical investigation, entering cancer clinical trials. Regardless of whether such characteriza-
conducting those investigations, and then preparing the informa- tion is designed for prospective or retrospective analyses, the increased
tion for registration. A single set of standards now exists for the need for collection of peripheral blood for immune parameters; germline
conduct of industry-sponsored trials worldwide (available at http:// and circulating free tumor DNA, tumor tissue for DNA, RNA, and/
www.fda.gov/ScienceResearch/SpecialTopics/RunningClinicalTrials/ or protein; and the need for specialized functional imaging, have
GuidancesInformationSheetsandNotices/ucm219488.htm). Attempts placed increasing demands on the infrastructure required to conduct
are therefore made to harmonize the development process to produce trials. These requirements have also introduced the necessity for
a globally accepted drug registration package to the greatest extent enhanced quality control on sample collection and storage that in
possible. turn necessitates increased resources. Nonetheless, these clinical trials
are more informative, and the need for sample collection is likely to
Models for the Conduct of Industry Clinical Trials increase in future trials. Numerous examples now exist (e.g., BRAF-
mutated melanoma, EGFR-mutated and ALK-mutated non–small
Biopharmaceutical industry sponsors usually produce the investigational cell lung cancer, and HER2 amplified breast cancer) to support the
agent in their own facilities because they anticipate eventually being contention that classifying tumors by their molecular characteristics,
responsible for the commercial production and distribution of the as opposed to classic histopathology, will result in more rapid drug
agent. They also can conduct the series of required clinical trials development, and more effective and well-tailored treatments.
directly using their own clinical trial personnel, which may include
internal company physicians, statisticians, monitors, data managers, EXPECTATIONS OF CLINICAL
quality assurance auditors, and the rest of the required infrastructure, RESEARCH SITES
such as company standard operating procedures, company information
system support, and drug distribution apparatus. Alternatively, drug Staffing is foremost among the critical components of an effective
sponsors may use a contract research organization to perform the research practice listed in Box 19.1. The number and precise composi-
clinical trials. In fact, for large development programs, it is not unusual tion of the necessary staff depend on the number of patients who are
for large companies to coordinate clinical trial programs using a mixture enrolled in clinical trials and the nature of the practice. In some settings
of both internal and externally acquired resources. Contract research in which the number of patients in studies is small, a single research
organizations are companies in the business of conducting clinical nurse can perform many of the required functions. At more active sites,
trials. During the past two decades, after the explosive growth of research nurses, clinical research associates, and research pharmacists
biopharmaceutical clinical investigation, a large number of such perform separate functions. For budgeting purposes, for example, it
companies were created, some capable of conducting global trials. is often estimated that one full-time clinical research associate can
The actual arrangement—direct or indirect—that a drug sponsor uses handle 25 new patients and up to 50 patients in follow-up in a year,
for a particular clinical trial is important to the investigator and staff although this will undoubtedly vary with the complexity of the trial
at the clinical trial site because it determines the predominant source menu. Some of the structures that support clinical trial participation,
of interaction and contact during the conduct of the trial. such as the NCORP program, provide substantial up-front funding
Different models also exist for investigator participation in clinical to support salaries for the necessary staff in return for a commitment
trials with industry. Traditionally, pharmaceutical sponsors have dealt to substantial accrual. Many others have a hybrid model, including
either with individual investigators or with individual institutions, as the NCTN and some industry-sponsored trials, wherein selected sites
314 Part I: Science and Clinical Oncology

clinical trial procedures at participating sites. NCI audits are typically


Box 19.1.  COMPONENTS OF AN EFFECTIVE conducted at a site every 3 years and review a sample of patients
RESEARCH PRACTICE enrolled in a variety of protocols, whereas industry audits are typically
focused on a single trial. Data quality initially became a concern when
• The presence of committed physicians who are willing to devote the clinical trial participation moved beyond academic sites to community
time and energy necessary to conduct clinical research and to practices. A number of evaluations in the 1980s, however, documented
conscientiously accept the significant responsibility inherent in the the ability of community sites to perform at a level comparable to
conduct of human research that of academic institutions in terms of data quality, protocol adher-
• The availability of suitably trained staff (preferably an experienced ence, and patient outcome. Indeed, approximately 50% of the accrual
research nurse) with enough time to assist in screening patients for to adult NCTN trials now comes from community practices in the
protocol eligibility and for follow-up of patients receiving protocol NCORP and from other community sites that are affiliated with
treatment academic centers that are NCTN group members.
• The availability of suitable staff to administer the required treatments As noted previously, although regulatory standards regarding the
in the protocol-prescribed manner; increasingly, this task might conduct of clinical trials impose similar overall expectations, for its
include administration of a wide range of potential therapeutics own reasons, industry monitoring is both more extensive and frequent
including, but not limited to, more conventional intravenous than is usual with public- or nonprofit-funded trial organizations.
chemotherapy and, in some cases, radiation
The basic tenet of monitoring for both industry-sponsored and NCI-
• Adequate and committed pharmacy capabilities to handle and
sponsored clinical trials is similar: the need to verify data accuracy by
account for investigational agents if they are part of the protocol
comparing case report forms, whether submitted by paper or electroni-
treatment
cally, with source data in the patient’s medical record. Increasingly,
• Adequate data management staff to handle the data-reporting
industry and public- or nonprofit-funded trial organizations are
requirements for patients who are being treated according to
implementing risk-based monitoring by applying metrics and trend
protocols
analysis to review trial data and assess site performance to detect
• Access to an institutional review board with Office for Human
potential problems early so that corrective and preventive measures
Research Protections assurances to approve the protocol and
can be implemented. The sponsor-assigned clinical trial monitor will
monitor the progress of the research
visit the site regularly, educate the involved staff about the goals and
• Access to suitable laboratory facilities to complete the studies
particular details of the protocol, and then track the progress of
required by the protocol
protocol-related activities throughout the conduct of the trial. Monitor-
• Willingness to comply with certain federal regulatory requirements,
ing responsibilities include confirming appropriate CIRB or LIRB
including adequate privacy procedures and training in human
review, investigator registration via completion of the FDA 1572 form,
subjects protection (available as an online course through the
and the existence of timely informed consent documents for each
National Institutes of Health at http://cme.cancer.gov/c01/)
patient; inspecting drug accountability records; confirming timely
and complete submission of serious adverse events reports; and ensuring
appropriate and timely handling of amendments. These activities all
fall within the responsibility of the clinical trial monitor, whether the
receive up-front payments but other sites receive funding only on a monitor is provided directly from the company sponsor or through
“per-case” basis, and thus start-up costs must be borne by the site. a contract research organization. Furthermore, regardless of whether
the clinical trial is conducted directly by its own organization or by
Quality Assurance, Monitoring, and Audits a contract research organization, biopharmaceutical companies often
conduct their own quality assurance audits and monitoring associated
Because the accuracy of the data collected in clinical trials is critical with the trial to further ensure the integrity of the data they submit
to the validity of the conclusions from the trials, all clinical trial to the FDA. The intensity of clinical trial monitoring tends to increase
organizations include quality assurance programs that reinforce “good as clinical trials mature and the data management group prepares to
clinical practice” guidelines by proactively monitoring and auditing officially “lock” the database before the conduct of prespecified analyses
clinical trials. Although these programs are structured somewhat and reporting activities. The intensity of quality assurance auditing
differently depending on the mechanism of participation (industry also increases for a particular clinical trial when it has been identified
studies involve the most frequent and extensive quality assurance), all as part of a new drug application or biologics license application for
such programs have certain features in common. All send queries to drug registration with the FDA. Monitoring and data management
the site when discrepancies or suspected errors are noted in submitted activities are evolving with the widespread introduction of electronic
information, and all compare data that are submitted from the sites data collection and submission systems, which have replaced traditional
against the primary medical or research record for verification by paper-based case report form approaches, and in the future it is likely
either monitoring or audit visits. The deployment of electronic data that less on-site and more central monitoring will occur.
capture systems (EDCs) in clinical trials has enabled the use of built-in
edit checks as an additional safeguard for ensuring data quality. Trial Educational and Training Tools and
monitoring is done in real-time primarily to assess a site’s performance New Federal Guidelines
during the conduct of a trial. Protocol deviations and violations at a
site recognized by monitors allow appropriate corrections to be made. As described previously, participation in clinical trials adds many
Auditors also review for data accuracy and protocol adherence, but complexities to the care of cancer patients and can require that physi-
in addition, informed consents are reviewed, as are adverse event cians, nurses, and other office staff acquire new and different skills.
reporting compliance, pharmacy practices, and timeliness of required Fortunately, because of the widespread interest in clinical trials in the
IRB submissions and approvals. Preparation for audits is time- cancer community, many resources exist for gaining information about
consuming for the sites, because all relevant records, including laboratory clinical trials and for acquiring the necessary skills. Professional societies
studies and films (e.g., computed tomography and magnetic resonance are a good source for educational programs and materials, and some,
imaging), required to document tumor measurements and response such as the Oncology Nursing Society (https://www.ons.org/), the
verification, must be gathered for the audit team. Some participants Society of Clinical Research Associates (http://www.socra.org/), and
consider this work onerous; however, audits fulfill an important the American Society of Health-System Pharmacists (http://
educational role in addition to ensuring the quality of the data and www.ashp.org/), actually offer certification programs that can serve
Structures Supporting Cancer Clinical Trials  •  CHAPTER 19 315

Box 19.2.  USEFUL WEBSITES FOR CLINICAL Table 19.2  New Requirements for National
TRIAL RESOURCES AND Institutes of Health (NIH)–Supported
INFORMATION and US Food and Drug Administration
(FDA)–Regulated Clinical Trials
• National Cancer Institute (NCI): http://www.cancer.gov
• Cancer Trials Support Unit (including links to NCI National Clinical NIH Supported FDA Regulated
Trials Network [NCTN] and NCI Community Oncology Research Good Clinical Practice (GCP) training
Program [NCORP] network groups and research bases): http://www required every 3 years for
.ctsu.org investigators and NIH staff
• Cancer Therapy Evaluation Program: https://ctep.cancer.gov/ Encourage use of a clinical trial Protocol template was
• Community Clinical Oncology Program: http://www3.cancer.gov/ protocol template to ensure that developed via a collaboration
prevention/ccop protocols contain all the information between NIH and FDA that
• Cancer Centers Program: https://cancercenters.cancer.gov/ needed for institutional review board meets the ICH Good Clinical
• NCI Central Institutional Review Board: http://www.ncicirb.org (IRB) and FDA investigational new Practice Guidance
• NCI Coordinating Center for Clinical Trials: http://ccct.cancer.gov/ drug (IND) applications
• National Institutes of Health: http://ClinicalTrials.gov Requirement for single IRB of record Single IRBs for multisite trials
• US Food and Drug Administration: http://www.fda.gov/ for multisite trials are permitted but not required
All NIH-supported clinical trials must Trials of drugs, devices, and
be registered in the ClinicalTrials.gov biologicals, other than phase I
database within 21 days of investigations, of a product
as important career development incentives to office staff. The American enrollment of the first participant subject to FDA regulation
Society of Clinical Oncology (http://www.asco.org/) also has a very must be registered in the
useful website with links to a variety of sites that provide information ClinicalTrials.gov database
about available clinical trials and detailed information about chemo- within 21 days of enrollment
therapy agents for physicians and nurses. NCI-sponsored NCTN and of first participant
NCORP groups and research bases provide regular educational activities All NIH-funded trials, including phase Trials of drugs, devices, and
for physicians, statisticians, nurses, and data managers who participate I, early device, and behavioral biologicals, other than phase I
in their trials. Furthermore, the biopharmaceutical industry sponsors intervention trials, are required to investigations, of a product
a wide range of educational activities that are conducted by both submit summary results to subject to FDA regulation
academic institutions and professional societies (e.g., the American ClinicalTrials.gov database within 12 must be registered in the
Society of Clinical Oncology and the American Association of Cancer months of the prespecified primary ClinicalTrials.gov database
Research) about the clinical trial process in general and about the outcome measures (primary within 12 months of the
responsibilities of the individual clinical investigator. completion date) prespecified primary outcome
The NCI’s home page (http://www.cancer.gov/) provides a gateway measures (primary completion
to the many websites of the NCI and provides links to many other date)
useful sites (Box 19.2). It contains extensive information about cancer
ICH, International Council for Harmonisation.
in general and cancer statistics, as well as detailed information about
clinical trials. A comprehensive listing of all trials conducted in the
United States, whether they are federally or privately sponsored, can improving cancer treatment and prevention. A growing number of
be found at www.ClinicalTrials.gov/. clinical practices have been able to integrate clinical research into their
Recently, the regulations regarding NIH-sponsored clinical trials activities successfully. Online tool kits can be helpful to sites as they
and FDA-regulated trials underwent important changes.9,13 These are prepare for participation in a clinical trial. For example, the CTSU
outlined in Table 19.2. These new procedures were created to meet the provides IRB submission packets, protocol calendars, and trial sum-
challenge of improving the quality of clinical trials while also enabling maries. The availability of a single IRB for multisite trials, as currently
the sharing of clinical trial data more expeditiously. The changes exists for all NCI-supported network trials, and common electronic
focus on enhancing the review, monitoring, and results reporting of data reporting systems also eliminates critical barriers and has made
NIH-sponsored clinical trials, along with new requirements for Good it easier for community physicians to participate. The biopharmaceutical
Clinical Practice (GCP) training and single IRBs for multisite studies. industry continually seeks interested, conscientious physicians to
Implementation of these comprehensive changes began in 2018. participate in its trials. The shortage of such physicians creates a
potentially serious limitation on the rate of development of new
CONCLUSION treatments for cancer. Surveys1 have suggested that the attitude of the
treating physician is perhaps the most critical factor in patient enroll-
Although involvement by oncologists in clinical trials introduces ment in clinical trials. An increasing number of resources and tools
additional complexities to their daily practice, it also provides substantial are now available to enhance access, with the potential to benefit both
benefits to all participants and ultimately contributes to the goals of current and future patients with cancer.

REFERENCES
1. Comis RL, Aldige CR, Stovall EL, et al. A quantitative 3. Sung NS, Crowley WF Jr, Genel M, et al. Central 5. Dilts DM, Sandler AB. Invisible barriers to clini-
survey of public attitudes towards cancer clinical trials. challenges facing the national research enterprise. cal trials: the impact of structural, infrastructural,
Available at: http://www.cancertrialshelp.org/static JAMA. 2003;289:1278–1287. and procedural barriers to opening oncology
_binary/308.pdf. 4. Unger JM, Barlow WE, Martin DP, et  al. clinical trials. J Clin Oncol. 2008;24:4545–
2. Mills EJ, Seely D, Rachis B, et al. Barriers to participa- Comparison of survival outcomes among cancer 4552.
tion in clinical trials of cancer: a meta-analysis and patients treated in and out of clinical trials. J Natl 6. Institute of Medicine. A National Cancer Clinical
systematic review of patient-reported factors. Lancet Cancer Inst. 2014;106(3):dju002. doi:10.1093/jnci/ Trials System for the 21st Century: Reinvigorating
Oncol. 2006;7(2):141–148. dju002. the NCI Cooperative Group Program. Washington,
316 Part I: Science and Clinical Oncology

DC: The National Academies Press; 2010. https:// 10. NCI Division of Cancer Prevention. NCORP Rural industry compared to publicly sponsored random-
doi.org/10.17226/12879. Populations infographic. https://prevention.cancer.gov/ ized controlled trials. PLoS ONE. 2013;8(3):e58711.
7. Christian MC, Goldberg JL, Killen J, et al. Sound- news-and-events/infographics/ncorp%E2%80%99s doi:10.1371/journal.pone.0058711.
ing board: a central institutional review board for -first-year-reviewed. Accessed January 12, 2017. 13. Zarin DA, Tse T, Williams RJ, Carr S. Trial reporting
multi-institutional trials. N Engl J Med. 2002;346: 11. McCaskill-Stevens W, Pearson DC, Kramer BS, Ford in ClinicalTrials.gov—the final rule. N Engl J Med.
1405–1408. LG, Lippman SM. Identifying and creating the next 2016;17(375):1998–2004.
8. Wagner TH, Murray C, Goldberg J, et al. Costs and generation of community-based cancer prevention
benefits of the National Cancer Institute Central studies: summary of a national cancer institute think
Institutional Review Board. J Clin Oncol. 2009;28: tank. Cancer Prev Res (Phila). 2017;10(2):99–107.
662–666. https://www.ncbi.nlm.nih.gov/pubmed/?term=10.11
9. Hudson KL, Lauer MS, Collins FS. Toward a new 58%2F1940230.
era of trust and transparency in clinical trials. JAMA. 12. Djulbegovic B, Kumar A, Miladinovic B, Reljic
2016;316(13):1353–1354. T, Galeb S, et al. Treatment success in cancer:
Oncology and Health Care Policy 20 
Steven Kent Stranne, Clifford A. Hudis, and Deborah Y. Kamin

S UMMARY OF K EY P OI NT S
• Policies promulgated by government relatively flat funding for the National the face of growing economic
at the federal, state, and local levels Cancer Institute since 2003 and pressure, oncologists and other
have profound effects on virtually biomedical inflation. The promise cancer care professionals must
every aspect of the day-to-day offered by new, highly effective remain engaged in helping to inform
practice of oncology. In the face of cancer therapies has never been policy makers and to shape these
growing financial pressures, policy brighter, and as a result the benefits changes. This effort includes working
makers will continue to make we are likely to realize from to ensure that policies designed to
decisions that may have tremendous adequate and consistent research reduce health care expenditures do
impacts on the cancer community. funding have never been greater. not undermine access to care or the
• Federal policies play a critical role • Policies adopted by the Medicare quality of care received by persons
in cancer research. Although the program regarding the prevention, with cancer and that reimbursement
national resources devoted to cancer diagnosis, and treatment of cancer for cancer care is fair and adequate.
research remain substantial ($5.6 have greatly influenced both the • Oncologists and other cancer care
billion in 2017), federal funding has practice of oncology and the specialists have unique insights
not kept pace with inflation or services available to both Medicare involving the care of patients with
increased scientific opportunities, and non-Medicare patients cancer, and it will be increasingly
and the nation is not making all of throughout the United States. Both important to communicate these
the progress it should. Legislation public and private insurers often insights effectively. Policy can have a
enacted in 2015 and 2016 increased rely on the coverage policies, profound impact on practice, making
the federal commitment to cancer reimbursement levels, and coding engagement in the process not only
research but does not overcome used by Medicare as a starting point important but also a professional
the impact of the combination of for establishing their own policies. In responsibility.

Federal and state governments play critical roles in establishing policies university-based cancer care providers and researchers are increasingly
that shape and fund virtually every aspect of cancer care and research linked in ways that may not be transparent to the casual observer.
throughout the United States. Although federal health care reform This chapter does not catalogue every policy initiative that affects
legislation—including the Affordable Care Act (ACA)1 and subsequent the cancer community. Rather, the following discussion provides a
legislative efforts—has attracted headlines for many years, such high- framework for understanding the important roles that federal and
profile legislation is just one aspect of cancer policy. Many important state governments play in regulating, shaping, and funding the oncology
policies arise with much less fanfare. Examples include policies adopted care delivery system and research enterprise.
by the National Cancer Institute (NCI) to guide the funding of research The roles played by federal and state policy makers in cancer
proposals, national coverage determinations for Medicare promulgated include:
by the Centers for Medicare and Medicaid Services (CMS), and policies
established by the US Food and Drug Administration (FDA) regarding • Research. Through the NCI and other federal agencies, the federal
products used for the prevention, diagnosis, and treatment of cancer. government serves a leadership role in and provides significant
For the foreseeable future, cancer policy initiatives must be forged funding for cancer research performed throughout the United
in a political environment beset by growing financial pressure. Potential States, including basic science research, translational research, and
cuts in reimbursement and funding will remain an ongoing threat, clinical trials.
raising the stakes for efforts to advocate for policies that protect and • Health care insurance. Through the ACA and subsequent legislative
promote the needs of persons with cancer, the cancer research enterprise, initiatives, the US Congress is engaged in reforming the health care
and oncology professionals. system, addressing concerns regarding access to health care insurance
As the modalities and therapies available to diagnose and treat cancer and the affordability of health care insurance. In addition, the
have become more successful and more complex, the administrative federal government operates and funds the Medicare program, which
burdens and financial constraints placed on all cancer care specialists covers approximately 60% of all patients with cancer in the United
have become more extreme. Perhaps more than at any other time, States.2 The federal government also oversees public health programs
policy issues confronting the cancer community are most properly managed by the CMS, the Department of Defense, the Veterans
viewed as affecting the oncology community as a whole rather than any Health Administration, and other federal agencies. Coverage,
individual segment. The fates of community-based, hospital-based, and reimbursement, and coding policies established by Medicare have

317
318 Part I: Science and Clinical Oncology

significant influence on the policies adopted by most other public patients and providers, the issue of Medicaid expansion remains
and private health care insurers. Federal and state laws may also complicated. The coverage requirements for the expanded population
create patient safeguards that private insurers must follow, such as created under the ACA differ from traditional Medicaid. Although
the protections established under the ACA to promote patient we do not have much evidence regarding the adequacy of coverage
access to cancer care through clinical research trials. for cancer care under the ACA expansion population, concerns remain
• Cost of care. Through its payment policies for providers and regarding Medicaid-covered cancer patients because some studies have
services, the federal government has a major impact on the care of indicated that, historically, cancer outcomes are not significantly better
patients with cancer. Policy makers have placed increasing emphasis for Medicaid patients in comparison with the uninsured.5–7 The Trump
on promoting quality, cost-effectiveness, and value in the health Administration has refocused federal administration of the Medicaid
care system over the past decade. With strong bipartisan support, program by enhancing opportunities for states to apply for and receive
Congress enacted the Medicare Access and CHIP Reauthorization waivers to redesign the rules for Medicaid program benefits and eligibil-
Act (MACRA) in 2015 to create a new framework for promoting ity, creating the potential that protections could erode for cancer
quality and value. The CMS articulated an aggressive timeline for patients on a state-by-state basis.
reforming its long-standing fee-for-service payment framework in Although some provisions of the ACA will remain highly contro-
favor of a system driven by value. These are some of the most versial, a number of relatively noncontroversial provisions are particularly
significant changes in health care organization and delivery since the important for the cancer community. These provisions include protec-
Medicare program was established in 1965.3 They will also motivate tions for patient access to preventive screening for cancer; protections
significant transformation on the part of oncology practices in the to help vulnerable individuals with cancer secure and retain access to
coming years. health insurance; safeguards for individuals with cancer and other
• Health information technology. Oncology care often involves preexisting conditions; and protections for patient access to clinical
complex diagnostic and treatment regimens delivered by multiple trials.8,9 These patient safeguards, which have bipartisan support,
health care providers, and health information technology holds warrant the cancer community’s ongoing attention and support.
tremendous promise to promote quality, communication, efficiencies, As with the ACA, cancer-related policies of significance at the
and research. Despite a federal investment of over $30 billion to federal level often arise from laws enacted by the US Congress (Table
promote the adoption of health information technology infrastruc- 20.1). However, in virtually every law enacted by Congress, there are
ture, health care providers continue to face significant challenges substantial gaps, conflicts, and ambiguities within the legislation that
in obtaining health care information stored by other health informa- must be resolved as part of the implementation process. As a result,
tion technology systems. Congress enacted provisions to promote the work to influence the final version of a national policy involving
interoperability as part of the 21st Century CURES Act in 2016. cancer rarely ends with the enactment of legislation. Advocates for
the cancer community must typically devote substantial attention to
educate agency officials and influence policies adopted by agency
BACKGROUND officials under both new and preexisting legislative authority. These
dynamics also occur at the state level.
Multiple areas of authority and discretion are relevant to cancer policy For policies relevant to the cancer community at the federal level,
development within federal and state governments. Each of the three the discretion to identify and address the gaps, conflicts, and ambiguities
branches of government—the legislature, the executive branch, and in the statutory language rests with the executive branch, including
the courts—plays a distinct role in a carefully constructed system of the Office of the President and the US Department of Health and
checks and balances. Although the Supreme Court’s reviews of the Human Services. In practice, all but the most politically sensitive
ACA during both 2012 and 2015 provide notable examples of the issues are typically determined by one of the sister agencies within
interplay among these branches of government, most cancer policies the Department of Health and Human Services, including the NCI,
do not involve litigation. National Institutes of Health (NIH), CMS, FDA, Centers for Disease
In 2012 the Supreme Court considered the constitutionality of Control and Prevention, Health Resources and Services Administration,
two major provisions of the ACA—the individual mandate (a require- and Agency for Healthcare Research and Quality.
ment that individuals maintain a minimum level of health care insurance These agencies are charged with providing the expertise to make
for themselves and their tax dependents or pay a tax penalty) and the effective policies in areas involving complex scientific and clinical
ACA’s requirements for states to expand their Medicaid programs. issues. As a result, there often are a substantial number of agency
The architects of the ACA relied heavily on Medicaid as a vehicle for employees who have scientific and clinical backgrounds, although
expanding coverage to the uninsured. In June 2012 the Supreme other agency officials may have limited experience in these areas. In
Court upheld the individual mandate but struck down the ACA’s many instances, agency employees are left with lean staffing to contend
requirement for states to expand their Medicaid programs.4 with some of the most challenging issues facing the US health care
In 2015, the Supreme Court again considered the constitutionality system. This situation heightens the importance of efforts by oncologists
of a major provision in the ACA, this time focusing on the validity to share detailed, practical solutions with federal and state policy
of federal tax credits, which provide financial subsidies for health makers regarding the complex issues facing the cancer community
insurance, in states without state-operated health insurance exchanges. and the health care system.
With annual federal subsidies to 6.4 million Americans at stake in
the 34 states with insurance marketplaces operated by the federal RESEARCH
government, the Supreme Court upheld the ACA subsidies. In 2017,
Congress eliminated the financial penalty for individuals who fail to The federal government plays an integral role in cancer research in
maintain health insurance (effective January 1, 2019). Although the United States, including basic science research, translational research,
Congress did not repeal the requirement for individuals to maintain and clinical trials. Although a number of federal agencies are active
health insurance, the legislation eliminated the ACA’s enforcement in supporting cancer research, the NCI (one of 27 NIH institutes
mechanism used to encourage individuals to obtain health insurance and centers) is the primary agency charged with guiding and funding
coverage. national research initiatives in oncology. Congress established the NCI
Medicaid funding and oversight are a shared responsibility between under the National Cancer Institute Act of 1937.10 The NCI is subject
states and the federal government, and political leaders in many states to oversight by Congress and the administration, but it retains significant
have indicated an initial unwillingness to engage in any significant discretion to establish overarching policies and make case-by-case
expansion of their Medicaid programs. From the perspective of oncology decisions that shape national research efforts.
Oncology and Health Care Policy  •  CHAPTER 20 319

Table 20.1  Selected Federal Laws Relevant to Cancer Policy

Federal Law Summary of Selected Effects on the Cancer Community


National Cancer Institute Act of Established the National Cancer Institute and charged it with conducting, fostering, and coordinating
1937 research and training related to the cause, prevention, diagnosis, and treatment of cancer
National Cancer Act of 1971 Began “War on Cancer,” initiated the National Cancer Program and its various boards, authorized creation of
new cancer centers and training programs, expanded existing research facilities, enhanced collaboration in
cancer research among federal, state, and other entities, and appropriated significant funds for a variety of
research-related purposes
Biomedical Research Training Expanded research on preventing cancer caused by workplace and environmental carcinogens and
Amendments (1978) emphasized cancer education programs initiated within local communities and hospitals
Veterans Health Care Act of 1992 Amended the Public Health Service Act by adding section 340B, which limits the amount certain safety net
providers must pay to manufacturers for covered outpatient drugs
Omnibus Budget Reconciliation Required Medicare to cover off-label uses of anticancer drugs recognized in certain medical compendia
Act of 1993 and required Medicare to cover orally administered anticancer drugs that have the same indications and
active ingredients as covered infused anticancer drugs
Health Insurance Portability and Limited the restrictions that group health plans can place on the coverage of preexisting conditions and
Accountability Act (1996) created security and privacy protections
Balanced Budget Act of 1997 Expanded Medicare coverage for several cancer screening tests, implemented the Sustainable Growth Rate
reimbursement methodology, and provided coverage for antiemetic drugs used as part of anticancer
chemotherapeutic regimens
Medicare Prescription Drug, Created Medicare Part D (prescription drug benefit), changed methodology for calculating relative value of
Improvement, and drug administration services, and instituted new average sales price payment methodology for many drugs
Modernization Act (2003) and biological products covered under Medicare Part B
Genetic Information Prohibited employment decisions made on the basis of genetic information and prohibited group health
Nondiscrimination Act (2008) plans and health insurers from making coverage and premium decisions for healthy persons on the basis
of genetic predispositions for future illnesses
Affordable Care Act (2010) Established safeguards under private health insurance plans to ensure coverage of individuals with
preexisting conditions and for individuals participating in clinical trials, established protections for patient
access to preventive screenings for cancer under Medicare, Medicaid, and private insurance, and
established the Innovation Center within the CMS for evaluating new payment methodologies, including
but not limited to accountable care organizations
Food and Drug Administration Established provisions to address shortages of cancer drugs and other medications, included a notification
Safety and Innovation Act (2012) requirement for manufacturers 6 months in advance of an anticipated drug shortage, directed the Food
and Drug Administration to include biologicals on drug shortage listings, and included fees for generic
drug applications to support agency resources for more rapid reviews of generic drugs, which also may
help avoid or alleviate drug shortages
Medicare Access and CHIP Created a new framework for promoting quality and value within the context of Medicare’s traditional
Reauthorization Act (2015) fee-for-service benefits, replacing the previous formula for physician reimbursement
21st Century Cures Act (2016) Authorized additional federal funding for cancer research and created mandates to promote
interoperability among health information technology systems.
Bipartisan Budget Act of 2018 Repealed the Affordable Care Act’s “individual mandate” tax penalty for failure to maintain health insurance
and made unrelated changes to the Medicare program in a number of ways that benefit the cancer
community, including a correction to avoid reductions to Medicare Part B drug reimbursement.

Most of the NCI’s funds are used to support extramural research welcomed ARRA funding, despite the fact that the ARRA funds
activities in communities located throughout the United States, which did not become part of the permanent baseline for the NCI and
often focus on important scientific questions that industry may have NIH budgets.
little incentive to explore (such as comparisons of competing drugs or In both 2015 and 2016, Congress enacted additional increases in
drug regimens). The NCI also supports intramural research conducted federal funding for cancer research. When finalizing the annual federal
at the NCI’s facilities in Maryland. From 1999 to 2003, the NCI and budget in late 2015 for fiscal year 2016, Congress included an increase
NIH benefitted from strong patient advocacy and a concerted effort in federal research funding of 6.6%, including $5.2 billion in funding
by Congress to double the NIH budget during this 5-year period. for the NCI’s cancer research initiatives. In addition, in December
The NCI budget for fiscal year 2017 exceeded $5.6 billion. 2016, Congress authorized the expenditure of an additional $1.8
Federal policy makers have recognized the importance of harnessing billion in funding over multiple years to support cancer research through
the economic benefits of cancer research and supporting the international the Cancer Moonshot initiative. The initiative, launched by President
leadership position of the United States in this field. For example, as Obama and led by Vice President Biden, was intended to bring public
part of the American Recovery and Reinvestment Act of 2009 (ARRA), attention and renewed focus on addressing barriers—technologic,
Congress provided nearly $1.3 billion in additional funding to the administrative, and clinical—to speedy development of more effective
NCI for use during fiscal years 2009 and 2010.11 Congress required therapies.
that the NCI target the ARRA funding to preserve and create jobs, Despite these positive developments and the substantial national
promote economic recovery, and increase economic efficiency through resources devoted to cancer research, the nation has lost opportunities
technologic advances in science and health. The cancer community and efficiencies because of suboptimal funding. Despite the significant
320 Part I: Science and Clinical Oncology

NCI ANNUAL FUNDING


$6,000,000
NCI funding
$5,000,000 Inflation adjusted NCI funding
Dollars (in thousands)

$4,000,000

$3,000,000

$2,000,000

$1,000,000

$0
2003 2004 2005 2006 2007 2008 2009 2010 2011 2012 2013 2014 2015 2016
Year

Figure 20.1  •  National Cancer Institute (NCI) annual funding 2003–2012. Inflation adjustments based on the National Institutes of Health (NIH)
Biomedical Research and Development Price Index. (From the NCI, Office of Budget and Finance. https://www.cancer.gov/about-nci/budget/fact-book/historical-
trends/funding, and NIH, Office of Budget. https://officeofbudget.od.nih.gov/gbipriceindexes.html. 2012.)

successes achieved in oncology research from the long-standing col- current rate accounts for only one-third to one-half of the actual costs
laboration between the federal government and researchers throughout incurred by providers conducting clinical trials. With current funding
the country, the uncertainty and inconsistency in federal funding constraints, the NCI has said that implementation of such a recom-
for the NCI during recent years has taken a substantial toll. For mendation would require the NCI to scale back plans for new trials
example, the aggregate funding levels for the NCI and NIH were or reduce accrual rates of existing studies.
relatively flat after the doubling of these budgets concluded in 2003 The national commitment to cancer research has continued, even
(Fig. 20.1). This stagnation eroded the effective level of federal support in challenging economic times, since Congress initially authorized
for cancer research by more than 20% between 2003 and 2015 because the NCI in 1937. However, the promise for new, highly effective
of the rate of biomedical research inflation. cancer therapies has never been brighter, and as a result the need to
Our research infrastructure faces ongoing uncertainty in large amplify our commitment to ensure that adequate research funding
part because of the annual nature of federal funding levels. The 21st exists has never been greater.
Century Cures Act, signed into law December 13, 2016, highlights
this concern. Although many advocates urged Congress to guarantee HEALTH CARE INSURANCE
the level of funding for cancer research over a multiyear timeframe,
the final law authorizes but does not guarantee the level of funding Through the ACA and subsequent legislative initiatives, the US
in future years. Ultimately, the funding levels will be determined Congress is engaged in reforming the health care system, including
by Congress through the annual appropriations process. The annual finding ways to help provide health care for those who are uninsured
nature of appropriating funds for federally funded research contributes and underinsured. Although many noneconomic factors influence
to the potential threat of cuts in cancer research funding levels in access to cancer care, lack of health insurance poses a major barrier
the future. to receipt of appropriate treatment. Cancer patients without adequate
The environment of flat budgets and looming cuts is undermining insurance receive less care, receive it later, and often have worse outcomes
and delaying realization of the significant scientific progress made than individuals with insurance. Uninsured and underinsured families
during the past two decades. Without sustained and predictable increases facing a diagnosis of cancer report that they are unable to meet their
in annual funding for the NCI, significant efficiencies are being lost out-of-pocket financial responsibilities and often must forgo cancer
because important studies may be started but not completed, the care in order to pay other bills. The specific direction that the federal
awarding of multiyear research grants is compromised, and established government will take in the future to address health care reform
research teams are destabilized. Oscillations in federal funding levels remains the subject of debate.
and uncertainty have the potential to dissuade highly talented young Policies adopted by the Medicare program regarding the prevention,
people from pursuing careers in cancer research. diagnosis, and treatment of cancer have greatly influenced both the
One issue of particular concern is the need to restructure and practice of oncology and the services available to both Medicare and
improve funding for the NCI Clinical Trials Cooperative Group non-Medicare patients throughout the United States. Medicare is a
Program. The Institute of Medicine published a report in 2010 recom- federal health insurance program that provides health care coverage
mending changes, and the NCI has worked to implement modifications to persons who satisfy eligibility requirements related to age or disability.
designed to both streamline the research process and prioritize clinical The influence of the Medicare program is due in large part to the
trials with the most promise for improving patient outcomes.12 Even significant size of the program, which covers approximately 60% of
with these changes, sustained increases in NCI funding are needed all patients with cancer in the United States.2 However, the influence
to ensure that clinical trials can be conducted in a timely manner to of Medicare’s policies extends well beyond its patient population.
translate recent scientific discoveries into benefits for day-to-day patient Both public and private insurers often rely on coverage policies,
care. For example, the Institute of Medicine report recommended reimbursement levels, and coding used by Medicare as a starting point
that the NCI increase the payment rate for clinical trials because the for establishing their own policies.
Oncology and Health Care Policy  •  CHAPTER 20 321

Cancer care has changed dramatically since Congress initially created In the face of extreme political and economic pressure, major
the Medicare program in 1965, and as Medicare has evolved, the transformations are occurring in both the public and private health
program has played an important role in defining the modern practice insurance sectors. Policy makers are making piecemeal (but significant)
of oncology. As the number of effective anticancer chemotherapy changes to the traditional reimbursement systems at the same time
regimens grew in the 1980s and 1990s, Medicare’s coverage of that new models of reimbursement are being tested by Medicare’s
intravenous drug therapies in outpatient settings enabled hospitals Innovation Center and private payers. Cancer patients are a unique
and physician practices to establish sophisticated, full-service facilities population and require special considerations when evaluating new
that care for patients with cancer within local communities throughout methods for health care delivery to protect and promote patient access
the United States. to high-quality, high-value cancer care. In many instances, a cancer
However, since the late 1990s Congress has enacted changes in diagnosis is both a medical and financial emergency for diagnosed
Medicare reimbursement levels for oncology drugs and professional individuals and their families. Often policy efforts intended to lower
services that have placed significant financial and administrative burdens costs or promote affordability have the consequence of eroding the
on cancer care providers. For example, Congress enacted provisions overall value of health care coverage by providing fewer benefits and
within the Medicare Prescription Drug, Improvement, and Moderniza- making it more expensive for patients to access their benefits. Oncolo-
tion Act of 2003 (commonly referred to as the Medicare Modernization gists and other cancer care professionals must remain engaged in
Act) that changed the reimbursement formula for oncology drugs helping to shape these changes to ensure that efforts to reduce health
administered in outpatient settings.13 Despite multiple steps taken by care expenditures do not undermine the quality of care received by
Congress and the CMS to mitigate adverse effects on oncologists, the persons with cancer, as well as to ensure that fair and adequate
net effect of these changes in reimbursement over time has exacerbated reimbursement exists to permit the ongoing delivery of high-quality,
the financial challenges of providing optimal oncology care. high-value cancer care.
Policy makers often justify the various forms of cuts in Medicare
payments to oncologists and other physicians on the basis that health COST OF CARE
care providers can shift the financial burdens arising from Medicare’s
underreimbursement to private-sector employers and insurers. This Traditional fee-for-service reimbursement rewards the volume of services
rationale is flawed in multiple ways, especially for oncologists who provided, but policy makers have raised concerns that fee-for-service
must serve increasingly large numbers of elderly (i.e., Medicare) patients. reimbursement alone does not promote the delivery of quality or
Congress enacted multiple safeguards over the years to protect cost-effectiveness in health care services. Policy makers have been
persons with cancer. For example, Congress enacted provisions in 1993 working to create incentives and penalties that promote the delivery
requiring Medicare to cover off-label uses of drugs and biologicals if of high-quality, high-value health care services for many decades. To
such uses were supported by clinical evidence reflected in the peer- this end, Congress has encouraged the adoption of managed care
reviewed medical literature or specific compendia.14 This patient under Medicare, Medicaid, and the private insurance sector under
safeguard has been critically important during the past two decades laws such as the Health Maintenance Organization Act of 1973 and
because pharmaceutical companies historically have been unlikely to the Balanced Budget Act of 1997.
seek FDA approval for multiple supplemental indications that support A large portion of Medicare beneficiaries continue to opt for coverage
the most important therapeutic agents for the treatment of cancer. under the traditional fee-for-service benefit of Medicare Part B. As a
Congress enacted a number of changes to Medicare under the result, Congress and agency officials have worked to develop initia-
ACA, including the creation of a new center within Medicare called tives that can mesh with traditional Medicare while promoting both
the Center for Medicare and Medicaid Innovation (the Innovation quality and value. The Physician Quality Reporting System (PQRS),
Center).1 Congress vested broad authority in the Innovation Center formerly known as the Physician Quality Reporting Initiative (PQRI),
to test and expand the use of innovative approaches to reimbursement is one such initiative that the CMS implemented between 2006 and
under Medicare without the need for further action by Congress. In 2016. However, one of the long-standing criticisms of PQRS was the
2016 the Innovation Center enrolled 200 oncology practices in a lack of a robust measure set for oncology and other specialties. To
reimbursement demonstration program called the Oncology Care address the need for improved quality measures in oncology and other
Model (OCM). The OCM operates in concert with Medicare’s tra- medical specialties, Congress enacted a provision under the American
ditional fee-for-service benefit. Central to this initiative, the CMS Taxpayer Relief Act of 2012 that permits use of physician-led registries
recognized the importance of providing additional reimbursement to such as the American Society of Clinical Oncology (ASCO) Quality
medical oncology practices beyond the standard levels available under Oncology Practice Initiative (QOPI) to satisfy requirements related to
the existing codes used for Medicare fee-for-service. The additional Medicare’s PQRS.
payments are triggered at the initiation of anticancer drug regimens, Congress enacted MACRA in 2015 to create a new framework
providing resources so that medical oncology practices can provide for promoting quality and value in conjunction with Medicare Part
additional services to help manage complications, avoid hospitalizations, B’s fee-for-service benefit for physician services. MACRA creates one
and otherwise improve cancer care. Participating oncology practices pathway called the Merit-based Incentive Payment System (MIPS)
are evaluated on the basis of clinical quality measures and cost savings. that incorporates and reconfigures Medicare’s system for measuring
The ACA also established a safeguard to ensure that persons with and creating financial incentives for quality, resource use, and health
cancer and other life-threatening conditions are covered under private information technology use and a new category for clinical quality
insurance if they determine with their physicians that enrolling in a improvement activities. The second MACRA pathway—Advanced
clinical study is in their best interest.1 The legislation requires insurers Alternative Payment Models (Advanced APMSs)—enables providers
to cover the routine costs of care associated with qualifying phase I, to share in potential cost savings achieved as part of their participa-
II, III, and IV clinical trials. Ensuring that patients with cancer can tion in the alternative model. The Advanced APM pathway requires
access clinical trials is not solely focused on promoting research, because providers to develop innovative programs for treating patients while
it is a long-standing and firmly held belief in oncology that the option assuming two-sided risk, which refers to accepting the risk of earning
to participate in a clinical trial is a key component of high-quality either bonuses or penalties based in large part on overall cost savings
cancer care and should be a readily accessible option for any patient achieved compared with benchmarks established by the Medicare
with cancer. In many instances, clinical trials provide persons with program.
cancer with the best chance for a successful outcome. The ACA The reforms established under MACRA hold promise to funda-
safeguard for patients who need access to clinical trials is similar to a mentally transform the Medicare program. The strong bipartisan
protection that already exists for Medicare beneficiaries. support that MACRA received in Congress suggests that the federal
322 Part I: Science and Clinical Oncology

government has identified a general approach to modifying the Medicare report on the technical, operational, and financial barriers to interoper-
fee-for-service program that may endure in the highly politicized health ability. Subsequently, in 2016 Congress enacted additional provisions
policy environment. There remains significant work ahead for oncology to tackle the issue of interoperability in the 21st Century Cures Act.
professionals to help shape the implementation of MACRA in ways Congress mandated the development of an exchange framework and
that support the needs of individuals with cancer and the oncology common agreement to promote interoperability. Although future
community without adding to the administrative burdens placed on adoption of the common standards will be voluntary, the federal
oncology providers. To this end, ASCO has developed cutting-edge government is empowered to encourage adoption by requiring use of
programs that help support the unique needs of cancer patients under the common exchange framework for entities contracting with the
the MIPS and Advanced APM pathways, including the Patient-Centered federal government.
Oncology Payment (PCOP) proposal and the QOPI. Our national interoperability challenges are exacerbated by informa-
tion blocking, which refers to situations in which information technol-
HEALTH INFORMATION TECHNOLOGY ogy vendors, health care providers, or others take proactive steps to
impede the exchange of information between health information
Oncology care often involves complex diagnostic and treatment regi- technology systems. Within the 21st Century Cures Act, Congress
mens delivered by multiple health care providers, and health information established safeguards that are intended to eradicate information
technology holds tremendous promise for oncology to promote quality, blocking, including the authority to pursue civil monetary penalties
efficiencies, and research. Timely and comprehensive access to all against violators.
components of an individual’s medical record provides opportunities In addition to assisting in the treatment of individual patients,
for health care providers to select the most appropriate care, avoid health information technology presents unique opportunities to advance
complications, and minimize needless duplication of diagnostic tests our knowledge base. Big data initiatives such as CancerLinQ provide
and therapeutic services. Although there has been significant progress the potential to collect, aggregate, and interpret data on large numbers
by providers of all types in adopting health information technology of cancer patients, providing observational data that will enable us to
in the United States, we continue to face significant challenges as a learn information from a high percentage of cancer patients—not just
nation with respect to the interoperability of these systems. the small percentage of patients who enroll in traditional randomized
The term interoperability refers to the ability of independent health controlled trials. The federal government is adopting policies that
information technology systems to successfully exchange electronic embrace these initiatives. For example, Congress has enacted safeguards
health information without special efforts by the users. In practice, that expressly extend the federal interoperability protections to
the health information technology systems used by different hospital physician-led registries such as CancerLinQ and that require the FDA
systems and by different physician practices often struggle to com- to evaluate how to incorporate observational data into its drug approval
municate effectively with one another, creating silos of health informa- decisions.
tion rather than a seamless source of information about the health Although issues involving health information technology affect all
care services provided to each patient. medical specialties, there are unique opportunities to harness health
As part of ARRA, Congress enacted a series of provisions known information technology in serving individuals with cancer. The oncology
as the Health Information Technology for Economic and Clinical community has special insights to share with policy makers on how
Health (HITECH) Act. This legislation initiated a multiyear subsidy best to use health information technology as quickly as possible.
program to help support health care providers in purchasing and using
health information technology. Before the enactment of the HITECH CONCLUSION
Act, only approximately 10% of hospitals and 17% of physician offices
had an electronic health records system in place. Between 2011 and Policies promulgated by government at the federal, state, and local
2016, the federal government awarded over $30 billion in incentive levels have profound effects on virtually every aspect of the day-to-day
payments to cover the costs associated with the adoption of health practice of oncology. In the face of significant financial pressures,
information technology infrastructure. By 2015, over 96% percent policy makers will continue to make decisions that have tremendous
of hospitals and over 80% of physician offices had established electronic impact on the cancer community. Oncologists and other cancer care
health records systems that satisfy national certification standards. specialists have unique insights involving the care of patients with
The Medicare program established “meaningful use” requirements cancer, and it will continue to be increasingly important to communicate
intended to determine whether physicians and hospitals are making these insights effectively. Policy can have a profound impact on practice,
use of health information technology. making engagement in the process not only important but a professional
The failure to achieve interoperability has attracted significant responsibility.
attention with many Members of Congress and other policy makers
lamenting this barrier to effective use of the substantial federal invest- The complete reference list is available online at
ment. In 2015, Congress enacted legislation that required a federal ExpertConsult.com.
Oncology and Health Care Policy  •  CHAPTER 20 322.e1

REFERENCES
1. The Patient Protection and Affordable Care Act, Pub. 6. Kelz RR, Gimotty PA, Polsky D, et al. Morbidity 11. American Recovery and Reinvestment Act of 2009.
L No. 111-148, as amended by the Health Care and and mortality of colorectal carcinoma surgery differs Pub. L No. 111-5. http://www.gpo.gov/fdsys/pkg/
Education Reconciliation Act of 2010, Pub. L No. by insurance status. Cancer. 2004;101:2187–2194. PLAW-111publ5/pdf/PLAW-111publ5.pdf.
111-52. http://housedocs.house.gov/energycommerce/ 7. Roetzheim RG, Pal N, Gonzaez EC, et al. Effects of 12. Institute of Medicine of the National Academies.
ppacacon.pdf. health insurance and race on colorectal cancer treat- A National Cancer Clinical Trials System for the
2. Smith BD, Smith GL, Hurria A, et al. Future of cancer ments and outcomes. Am J Public Health. 2000;90: 21st Century: Reinvigorating the NCI Cooperative
incidence in the United States: burdens upon an aging, 1746–1754. Group Program, April 15, 2010. http://iom.edu/
changing nation. J Clin Oncol. 2009;27:2758–2765. 8. Moy B, Polite BN, Halpern MT, et al. American Reports/2010/A-National-Cancer-Clinical-Trials-
3. Health Insurance for the Aged Act. Public Law 89-97. Society of Clinical Oncology Policy Statement: System-for-the-21st-Century-Reinvigorating-the-
July 30, 1965. Opportunities in the Patient Protection and Affordable NCI-Cooperative.aspx..
4. National Federation of Independent Business v. Sebel- Care Act to reduce cancer care disparities. J Clin 13. Medicare Prescription Drug, Improvement, and
ius, No. 11-393, slip op. US June 28, 2012. http://www Oncol. 2011;29:3816–3824. Modernization Act of 2003. Pub. L No. 108-173.
.supremecourt.gov/opinions/11pdf/11-393c3a2.pdf. 9. Stranne SK. An oncology perspective on preventive http://www.gpo.gov/fdsys/pkg/PLAW-108publ173/
5. Roetzheim RG, Gonzalez EC, Ferrante JM, et al. services in the context of US healthcare reform. pdf/PLAW-108publ173.pdf.
Effects of health insurance and race on breast carci- Oncology. 2012;25:1119–1132. 14. Omnibus Budget Reconciliation Act of 1993, Pub. L
noma treatments and outcomes. Cancer. 2000;89: 10. National Cancer Institute Act of 1937. Pub. L No. No. 103-66. http://www.gpo.gov/fdsys/pkg/BILLS-
2202–2213. 75-244. http://legislative.cancer.gov/history/1937. 103 hr2264pp/pdf/BILLS-103 hr2264pp.pdf.
E. PREVENTION AND EARLY DETECTION
Discovery and Characterization of Cancer 21 
Genetic Susceptibility Alleles
Stephen J. Chanock and Elaine A. Ostrander

S UMMARY OF K EY P OI NT S
• The discovery of cancer variation across the human genome in probability; and common variants,
susceptibility regions across the continue to accelerate the pace of which impart a small risk for cancer.
genome provides opportunities to discovery and characterization of • Both association studies and
understand defining events in tumor cancer susceptibility alleles. The family-based studies require
development, specifically identifying conclusive identification of a gene or accurate collection of clinical data by
cellular pathways that contribute to a regulatory region contributes to an clinicians, and both represent an
the complex development of cancer. understanding of defining events in important pillar for precision
• Regions of the genome harboring tumor development. medicine and precision prevention.
susceptibility alleles and genes can • The spectrum of cancer An improved understanding of
be identified in families or unrelated susceptibility alleles includes molecular aspects of cancer and
populations by using association mutations in genes that are highly specifically the use of biomarkers
studies, next-generation sequencing, penetrant, which indicates that such as susceptibility alleles can
and linkage studies. individuals born with a mutant allele inform clinical and public health
• Technologic advances in sequence have a high probability of developing decisions.
analysis in concert with cancer; moderately penetrant
comprehensive annotation of genetic mutations, which confer an increase

For generations, investigators have pursued the heritable contribution large data sets, investigators have forged new collaborations and
to cancer. Seminal studies in families in which members affected with developed computational tools for analyzing ever-enlarging data sets
breast cancer, colorectal cancer, melanoma, or a constellation of cancers in search of new cancer susceptibility alleles. The ability to discover
(e.g., Li-Fraumeni syndrome) have provided evidence for rare mutations and validate cancer susceptibility alleles is highly dependent on sharing
with strong effects.1–3 Generations of family-based and twin studies of data through accessible databases enabling bona fide and approved
have repeatedly shown an excess of familial cancer aggregation for researchers to carry out additional studies.8,9
nearly all types of cancers, although the estimates vary greatly across Cancer susceptibility alleles have been discovered through a spectrum
cancer types. These observations suggested that it would be possible of approaches, yielding a range of inherited genetic variants from rare
to map cancer genes and thus estimate the genetic contribution to mutations with strong effects (i.e., highly penetrant) to common genetic
each molecular type of cancer, even in unrelated populations. Until polymorphisms, each of which confers a small risk for cancer.2 By
the mid-2000s, progress had been slow. However, the pace at which definition, susceptibility alleles increase an individual’s risk of developing
new genetic regions harboring cancer susceptibility alleles have been cancer either within families or across populations, but there are
identified has accelerated substantially because of three converging instances of pleiotropy in which a particular variant may confer
factors. First, a high-quality draft sequence of the human genome was susceptibility to one type of cancer yet protect against another type
produced.4,5 Second, its subsequent annotation resulted in the apprecia- of cancer. It is notable that not all susceptibility alleles for a given
tion of a wide spectrum of variation across the genome.6,7 Third, the gene have equal estimated effects. Consequently, the observed spectrum
development of technical platforms that enable interrogation of genetic of established susceptibility alleles reflects an inverse relationship
variation across the genome has changed the discovery speed of cancer between the effect size and the frequency of the genetic variation (Fig.
susceptibility alleles. In the last decade the scope of studies changed 21.1).10–12 So far, approximately 125 hereditary cancer predisposition
dramatically, expanding from family-based studies to larger population- genes (CPGs) have been identified, and their presence is most evident
based studies of unrelated individuals. These studies have been fueled in studies with familial clustering of one or more cancers.13 Moreover,
by the precipitous drop in price for interrogating large numbers of the majority of the hereditary predisposition genes are known somatic
informative single-nucleotide polymorphisms (SNPs), the most common mutational drivers of cancer, highlighting an important principle first
type of variant in the genome, or massive parallel sequence analysis outlined by Knudson in 1971.14 The first set of CPGs was discovered
of entire or partial genomes, particularly the coding regions of the in family studies using linkage analysis, but now next-generation
genome (known as the exome). To keep pace with new streams of sequencing (NGS) analysis has accelerated the discovery of new genes

323
324 Part I: Science and Clinical Oncology

Effect size
50.0

High Rare alleles


causing
mendelian
disease
3.0

Intermediate Low-frequency
variants with
intermediate effect
1.5
Common
Modest variants
implicated in
1.1 common disease
by GWA
Low

0.001 0.005 0.05


Very rare Rare Low frequency Common
Allele frequency

Figure 21.1  •  Feasibility of identifying genetic variants by risk allele frequency and strength of genetic effect (odds ratio). GWA, Genome-wide
association.

and specific mutations in known CPGs. Susceptibility alleles that indispensable tools for geneticists in identifying cancer susceptibility
have an appreciable frequency in the general population, and smaller alleles (Fig. 21.2).6,7,15–17
effect sizes, can be discovered through association studies in which The search for common alleles is predicated on conclusively finding
the genomes of a set of affected cases are compared with those of “markers” that highlight a region of the genome where a disease
unaffected cancer-free or population-based controls.11 susceptibility allele resides; subsequent mapping and laboratory studies
Genetic mapping of cancer susceptibility genes has shown that are required to understand the link between the susceptibility allele
many signals map to nongenic regions, important in the regulation and its biologic association with disease.18 Sets of markers to test are
of genes or pathways of interacting genes. Although the direct public drawn from dense maps of human genetic variation that are publicly
health impact associated with conclusively establishing a specific available. Although there has been great value in embracing this indirect
cancer susceptibility allele may not be immediately apparent, its approach (Fig. 21.3), it comes at a price—namely, many additional
contribution to understanding tumor development and metastasis steps are needed in order to sort through the correlated variants and
is invaluable, expanding possible pathways and putative targets for then conduct the functional studies that are necessary to illuminate
intervention downstream. Moreover, the possible clinical value of known the underpinnings of the susceptibility allele.11,19
susceptibility alleles will continue to increase as more comprehensive Until whole human genome sequence became available, the field
maps of susceptibility alleles emerge for specific cancers. In this of genetics created maps of variant coordinates based on incomplete
regard, sets of variants tested together show great promise for risk assembly of DNA segments spanning the human genome. Sets of
stratification, important for both the individual and at the population markers can be thought of as molecular street signs, which allowed
level. Roughly 10% to 15% of cancer susceptibility alleles are shared scientists to knowingly navigate their way up or down a region of a
among cancer types, and in some circumstances there are distinct chromosome. Early on, “genetic maps” provided a stable reference for
differences by subtype (e.g., specific alleles for estrogen-negative breast mapping highly penetrant mutations, primarily in families. These
cancer). To define the genetic architecture (see Fig. 21.1), namely were based on empiric evidence of recombination hot spots. The
the constellation of susceptibility alleles that contributes to a specific long-standing value of functional elements, herein recombination
cancer, continued efforts are required to define comprehensive sets frequencies, served adequately for the mapping of disease and traits
of variants, which in turn should emerge as vital tools in both public until genome sequencing emerged as a viable tool. The emergence of
health and individual (known as precision medicine) assessments of a physical map of the human genome that establishes the true order
cancer risk.9,12 of most DNA segments (currently tractable for more than 95% of
the genome) has accelerated the mapping of traits and diseases, as it
produced absolute coordinates, or street signs for variants within the
FUNDAMENTAL SCIENCE genome—that is, the nucleotide location of a given marker or gene
Genetic Variation in the Human Genome in millions of base pairs from the end or terminus of the chromosome
is generally known.
The annotation of sequence variation in the genome has provided The principle of meiotic recombination is critical to understanding
important clues toward elucidation of the genetic history of distinct the relationship between genetic loci, here defined as variants that
populations, the possible impact of environmental or pathogenic assault map to unique coordinates. The correlation between genetic markers
on the genome, and the heterogeneous distribution of human cancers. is fundamental to both association and linkage analysis. In meiosis,
The differences in allele frequency spectrum, as well as types of genetic the cell division leading to gamete formation pairs homologous
variation, from SNPs to large copy number variants, have become chromosomes. Each chromosome consists of two identical strands
Discovery and Characterization of Cancer Genetic Susceptibility Alleles  •  CHAPTER 21 325

Single nucleotide Molecular

Sequence
variation
• Base change – substitution – point mutation genetic
• Insertion-deletions (“indels”) detection
• SNPs – tagSNPs

2 bp to 1,000 bp
• Microsatellites, minisatellites
• Indels
• Inversions
• Di-, tri-, tetranucleotide repeats
• VNTRs

1 kb to submicroscopic
• Copy number variants (CNVs)
• Segmental duplications
• Inversions, translocations
• CNV regions (CNVRs)
Structural variation

• Microdeletions, microduplications

Microscopic to subchromosomal
• Segmental aneusomy
• Chromosomal deletions – losses
• Chromosomal insertions – gains
• Chromosomal inversions
• Intrachromosomal translocations
• Chromosomal abnormality
• Heteromorphisms
• Fragile sites

Whole chromosomal to whole genome


• Interchromosomal translocations
• Ring chromosomes, isochromosomes
• Marker chromosomes
• Aneuploidy
Cytogenetic
• Aneusomy detection

Figure 21.2  •  Spectrum of variation observed in the genome. The figure depicts both the size and scope of variants as a function of their length and
density in the genome. SNPs, Single-nucleotide polymorphisms; VNTRs, variable number tandem repeats.

(chromatids), with each chromosome pairing composed of four strands. these are population private, reflecting population genetics history.20,21
Homologous chromosomes separate from each other during the process The majority of SNPs with an MAF greater than 10% are common
of meiosis except at one or two zones of contact in a process that to most human populations, but the actual frequencies can vary.
leads to genetic recombination (Fig. 21.4). Mendel’s second law, Reported SNPs are cataloged in db-SNP (http://www.ncbi.nlm.nih.gov/
independent assortment, states that alleles of genes at unlinked loci, snp), which is an important reference that is useful for interpreting
such those on different chromosomes or at the ends of a chromosome, variants identified through DNA sequencing.
segregate or assort independently. Deviations from independent A small subset of SNPs are located in exons, of which a fraction
assortment occur when genes are located close to one another, in change the predicted amino acid. SNPs that can alter the coding
which case alleles assort together more than 50% of the time. In this sequence are known as nonsynonymous SNPs, whereas those that are
scenario, the associated loci are “linked.” Distributed throughout the silent are termed synonymous. Although there has been great interest
genome are recombination hot spots, which “divide” the genome in coding SNPs, partly because they appear to be more interpretable,
during meiotic formation of egg and sperm. These can vary by popula- very few of the known associations between a disease and a common
tion genetic history, providing an opportunity to compare groups and SNP marker (MAF > 10%) involve coding SNPs. On the other hand,
use the differences to pinpoint possible susceptibility alleles, especially rare highly penetrant mutations mainly map to coding changes or
if there are substantive differences between populations with respect preterminal stop codons. Many of the reported disease mutations,
to cancer incidence. known as cancer predisposition genes, are cataloged in public databases
The spectrum of human genetic variation varies by the frequency such as Online Mendelian Inheritance in Man (https://www.omim.org/)
of polymorphisms, which is often substantial among populations. or Catalogue of Somatic Mutations in Cancer (COSMIC; http://
The most common sequence variation is the substitution of a single cancer.sanger.ac.uk/cosmic).22
base, known as a single-nucleotide polymorphism, which, by definition, SNP frequencies become fixed in populations over multiple genera-
must be observed in at least 1% in one or more populations. The tions and are generally not inherited independent of the adjacent
minor allele frequency (MAF) refers to the lower allele frequency, often variants. Recombination hot spots can separate sets of highly correlated
varying by population ancestry. A substantially larger fraction of genetic variants, resulting in haplotype blocks (Fig. 21.5).23 These segments of
variation exists for single base substitutions below 1%—and many of a chromosome, usually quite small, are transmitted as a unit from
326 Part I: Science and Clinical Oncology

i
a b c d e f

ii Direct test

a b c d e f

iii Indirect test Indirect test

a b c d e f

iv
1
a c d
Figure 21.4  •  Genetic recombination is the process of exchanging genetic
2 information between two chromatids during meiosis. The recombination events
b f
3 for a single chromosome within a family are illustrated. The father’s homologous
e chromosomes are light and dark purple, and the mother’s are light and dark
green. Recombination events occurring during meiosis create unique parental
chromosomes.
Figure 21.3  •  Direct versus indirect association testing. (i) Six common
single-nucleotide polymorphisms (SNPs) as they would be represented in a
population sample. SNP-c is responsible for conferring a disease phenotype
on carriers. In a direct test (ii), SNP-c would be directly assayed and tested
for association with the disease, perhaps based on prior evidence of structural
or functional consequences of variation at this site. In contrast, the indirect
approach (iii) is agnostic with regard to functional variation. The assayed Unlinked
markers need only be in linkage disequilibrium with the causative variant to
achieve a signal of association. The caveat with this method is that care must
locus 1 AB CC
be taken to type the appropriate markers needed to ensure thorough coverage
of a given region. In the hypothetical example shown, tests of association locus 2 XY ZZ
between disease status and genotype at SNP-b, SNP-e, or SNP-f would prove
nonsignificant. Only SNP-a and SNP-d are indirectly associated with the
disease. The reason (iv) is that SNPs arise on independent haplotypic back-
grounds and that many common haplotypes exist at a given locus (three are
illustrated in the example, but in a true scenario many more are likely to be AC BC AC BC AC BC AC BC
present). If we assume that SNP-c arose on haplotype 1, we can see that XZ YZ YZ XZ XZ XZ YZ YZ
assaying the SNPs that define haplotypes 2 and 3 will not be useful in A NR NR R R NR R R NR
demonstrating an association of this locus with the disease. Instead, to fully
analyze this region, we must assay at least one haplotype “tagging” SNP from
each of the observed haplotypes. Linked

locus 1 AB CC
locus 2 XY ZZ
one generation to the next. The correlation between SNPs is an estimate
of linkage disequilibrium (LD), which is classically defined as the
nonrandom association of alleles at different loci. Individual SNPs
that always track together are said to be in strong LD. This correlation
can be eroded over time by recombination (exchange of genetic material) AC BC AC BC AC BC AC AC
during meiosis, and SNPs can be defined as being in weak LD,24 XZ YZ XZ YZ XZ YZ XZ YZ
signaling a correlation that is not necessarily strong. Measurement of B NR NR NR NR NR NR NR R
LD is with either D′ or r2 statistics.
The concept of LD is important because it enables investigators Figure 21.5  •  Linked and unlinked markers segregating in two families.
to evaluate sets of SNPs and determine proxies for other, untested Below the symbols, the genotypes for both markers are listed. Offspring have
SNPs in a region; this is useful for indirect mapping. If a group of either recombinant (R) or nonrecombinant (NR) haplotypes. The father is
SNPs is in strong LD (e.g., they are inherited together), one can test heterozygous for marker 1 (AB) and marker 2 (XY); and the mother is
for the alleles of just one reference SNP and immediately have informa- homozygous for both markers (CC and ZZ). (A) If the markers were unlinked,
there would be equal numbers of R and NR haplotypes from the father (AX,
tion regarding alleles segregating in a given individual for all adjacent, BY, AY, and BX). (B) There is an excess of NR haplotypes (AX and BY), and
correlated SNPs. By extension, estimates of LD are useful to construct only one R haplotype appears. Therefore these loci are linked.
haplotypes in unrelated subjects. With new reference data sets (e.g.,
1000 Genomes Project), it is possible to reliably determine or “impute”
untested variants using the backbone of stable data sets.25 Computational
efficiencies enable estimation of the correlation between sets of markers
Discovery and Characterization of Cancer Genetic Susceptibility Alleles  •  CHAPTER 21 327

and the construction of haplotypes.25 Still, the most reliable approach from 1.7-fold to 3.7-fold. Younger ages at diagnosis and multiple
is to resolve the phase of haplotypes in multigeneration pedigrees, in affected relatives with the disease tend to be associated with even
which haplotypes can be traced; alternatively, one can infer the relation- higher relative risk (RR).33–35 For example, men with three or more
ship of alleles in unrelated subjects with computational tools.26 Phase first-degree relatives with prostate cancer have an almost 11-fold
refers to the parental (and grandparental) chromosome of origin for increased risk of the disease compared with men who have no known
a set of alleles.27 This information can also be useful for determining family history of the disease.34 For this reason, families ascertained
where in a family a disease allele originates.27 for linkage analysis studies tend to be large, have multiple affected
The annotation of the human genome has revealed a wide spectrum individuals, and feature people who were diagnosed with the disease
of structural variations, which may be either cytologically visible or at a comparatively young age.
detected with either microarray chips or actual sequence analysis (see Familial aggregation describes the occurrence of multiple cases of
Fig. 21.2). Historically, short tandem repeats (STRs) are a class of cancer within a family (Fig. 21.6). Clustering of familial cases can
polymorphisms in which there is a reiteration of a small number of also be due to shared environment, shared alleles of particular genes,
base pairs, such as CACACA. Polymerase chain reaction (PCR) primers or simply chance if the tumor is common in the population. In
are used to define the distinct physical location of one STR from the mapping cancer susceptibility genes for many cancers, particularly for
remaining 50,000 in the genome and to ascertain the number of breast cancer and colon cancer, the most promising pedigrees for
repeats for both chromosomes for any given STR. Also known as hereditary cancer are families with three or more first-degree relatives
microsatellites, they have been used for linkage analysis and forensic with a given cancer, three successive generations with cancer, or at
investigation. Structural variants of all sizes can include deletions, least two siblings with the same cancer detected at a relatively young
insertions, and duplications collectively known as copy number variations age. For common cancers, details regarding tumor stage and grade at
(CNVs). In addition, there are infrequent inversions and translocations diagnosis, histopathology, and response to treatment are key to linkage
of pieces of DNA that vary in size. Some of these are quite common; analysis study design because individuals who share similar disease
chromosome 17 harbors an inversion of 3.5 million base pairs in features likely share common susceptibility alleles. For instance, if a
approximately 20% of the European population.28 CNVs have been subset of affected individuals in the family in Fig. 21.7 all had tumors
shown to influence gene dosage and therefore can contribute to risk of similar stage and grade, the data from this homogenous subset of
for cancer, as demonstrated for a chromosome 1 CNV and risk for individuals could be considered in isolation from the rest of the affected
childhood neuroblastoma.29 There have been formidable technical cases thus reducing heterogeneity and increasing power. For common
challenges in calling CNVs, but with new resources and new sequencing diseases there are many common susceptibility alleles that account
technologies termed next-generation sequencing, it is anticipated that for a substantial fraction of the underlying genetic architecture of
accuracy will continue to improve,30 which in turn should improve cancer susceptibility,11,36 and this approach generally does not work.
the capacity to associate CNVs with disease outcomes. However, it has proven useful in discovery of highly penetrant alleles
for many cancers (e.g., breast and colon cancers).37,38
Principles of Linkage Mapping To identify highly penetrant mutations, success directly correlates
with the identification and collection of high-risk or hereditary families.
Many epidemiologic studies have indicated the presence of a familial To achieve the numbers needed to improve the power to detect a
contribution, such as the observation that family history of a specific disease allele, whether using SNP arrays or NGS, large consortia are
cancer within first-degree relatives is associated with a doubling of often formed, providing an opportunity to increase power through
risk or greater among relatives, particularly in twin registries.31,32 In addition of more families and the chance to define the phenotype—
the case of prostate cancer, for instance, studies of selected hospital-based namely, the required clinical features and family history.39 Large
patient populations, population-based case-control studies, and cohort consortium studies provide an opportunity to conduct segregation
studies all have demonstrated that a family history of disease is correlated analysis, which determines the most likely genetic model that could
with an increase in an individual’s risk. If the affected family members account for the disease (e.g., dominant, codominant, recessive, or
are first-degree relatives (i.e., brothers, fathers, sons), the risk increases sex-linked). Additional informative analyses include an estimate of

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26
00
02 06 82
81 81 03
00 96
63 00
02 00
03 77 02 00 00 61
02 01 00 70 87
21 77 00 77
70 81 97
20 77 70
20 69 97
68 94
26 8 63 81 91
81 83
65 96
32 83
25 83
80

25
25

Figure 21.6  •  Correlation of variants in a linkage disequilibrium plot. A region of the genome is depicted between two recombination hot spots that
shows the relationship between variants based on either D′ or r2 analysis. The red color indicates a high degree of correlation between variants.
328 Part I: Science and Clinical Oncology

DNA samples from appropriate family members can be screened


by using either a set of highly polymorphic markers that span the
76 genome at a sufficiently high density or NGS analysis of the whole
genome or the exome.
Theoretically, a given set of affected individuals within a family
would all carry precisely the same underlying mutation in the relevant
58 68 70 62
susceptibility gene—that is, each member would have inherited not
just a mutated copy of the same gene, but the same deleterious variants
in that gene. Because there are distinct mutations within a gene, each
of which can confer high penetrance, the approach is predicated on
finding a gene and not the specific mutation within a gene. For example,
A 65 80 78 45 50 48 45 there are many mutations and variants of unknown significance across
the BRCA1 gene that can confer an increased risk for breast and/or
ovarian cancer, with measurable differences in penetrance now confirmed
by prospective studies.40,41 This latter point suggests that there are
differential effects of disturbances of key biologic pathways. Moreover,
41
recent genome-wide association studies (GWASs) have begun to identify
secondary, genetic modifiers that further modulate the penetrance of
BRCA1 mutations.42,43
46 47 45 Fig. 21.7 demonstrates two types of seemingly useful families for
linkage mapping studies. Both include a significant number of affected
members. The first family has a large number of affected individuals
(see Fig. 21.5). However, some individuals were affected very early in
life, whereas others were diagnosed at later ages. It is likely that some
B 46 48 50 42 individuals have the disease because they inherited mutated copies of
Figure 21.7  •  Two theoretical breast cancer families. Age at diagnosis is
a particular gene, whereas others have the disease for sporadic reasons
indicated below the symbol; males are indicated by squares, and females by unrelated to the disease allele segregating in the family. Age at onset
circles. (A) The family has many members with breast cancer, but some were provides guidance as to which individuals are more likely to have
given diagnoses relatively early in life (younger than 50 years), whereas others hereditary versus sporadic forms of the disease; but this is not absolute,
were much older at diagnosis (older than 70 years). The usefulness of this and in the case of a disease with age-dependent penetrance, some
family for genetic mapping studies is therefore limiting because it likely contains people will be affected late in life, even though they carry a mutant
individuals with both sporadic and hereditary breast cancer. (B) All individuals allele, and others will be affected early in life for sporadic reasons.
were affected at an early age, but breast cancer, caused by mutations in either The second family shown in Fig. 21.5 appears to be more informative
the same or different genes, is present on both sides of the family. Because for linkage mapping studies because there are several affected individuals
there is no way to distinguish the number of mutant genes, a priori the usefulness
of this family for a genome-wide scan is also somewhat limited.
in the family and all were affected at a relatively early age. However,
the presence of disease segregating on both sides of the family should
be noted. The affected individuals in the youngest generation could
have cancer because they inherited mutant alleles from one or both
sides of their family, and one or multiple genes could be involved.
the frequency and penetrance of the disease allele(s) in the general This scenario happens frequently in studies of common cancers such
population, age-dependent penetrance, and potential number of loci as those of the colon, breast, and prostate. Thus the family is of limited
contributing to the disease. usefulness for mapping studies.
High-risk or hereditary families must be ascertained through use
of appropriate guidelines for working with human subjects to collect Challenges in Finding Cancer Susceptibility Genes
biospecimens, such as germline DNA from blood or buccal materials,
somatic or tumor tissue for DNA or RNA analyses, and other body Traditional approaches have been successful in identifying highly
fluids for determination of biomarkers that could be useful in subse- penetrant mutations in multiply affected families for both common
quent early detection in high-risk settings. Identification of cancer and uncommon cancers. A combination of linkage and candidate
families and collection of critical medical information including family gene analyses revealed mutations in CDKN2A or CDK4 in roughly
history, medical record data, and DNA samples are generally regulated 50% of familial melanomas, although there appears to be heterogeneity
by institutional review boards (IRBs) and require informed consent. in exposure to a strong carcinogen for melanoma, ultraviolet sunrays.44
Families must be identified and approached in a way that is neither Newer approaches involving analysis of smaller family structure together
intrusive nor coercive. Although genetic epidemiologists have success- with functional laboratory data have identified rare, important muta-
fully turned to new approaches, such as social media, to ascertain tions in melanoma. For a rare familial cancer, chordoma, a gene
families, most families are made aware of or referred to studies by duplication of the T (brachyury) gene confers susceptibility.45 Using
their personal physicians. NGS, investigators are revisiting families whose disease was not
To carry out a successful family-based genetic study, medical record understood when traditional methods were used, and are now searching
data must be carefully and systematically extracted into well-protected for sets of susceptibility alleles that can explain an oligogenic risk
databases. Family history data must also be obtained redundantly model—that is, a set of variants with moderate risk that are neither
from multiple family members, with care taken to resolve discrepan- sufficient nor necessary for development of a cancer (e.g., MITF is a
cies, including nonpaternity events. Consent to contact other family moderate-risk gene for melanoma).46,47
members regarding the study is needed, as is permission to obtain The early-onset breast cancer susceptibility genes BRCA1 and
medical records and to recontact study participants years after initial BRCA2 were among the first to be mapped because large and well-
data collection. The protection of individual privacy is paramount, characterized families had been meticulously ascertained. In addition,
and personal identifiers such as names and complete addresses are deleterious alleles were highly penetrant, often at an early age, leaving
rigorously protected, even from most of the scientists participating in little ambiguity as to who in a family should be counted as a “case.”
the study. The presence of ovarian cancer in some families and not others and
Discovery and Characterization of Cancer Genetic Susceptibility Alleles  •  CHAPTER 21 329

the presence of breast cancer in some male carriers allowed for creation in underlying population genetics history). GWASs are pursued free
of data sets enriched for the BRCA1 and BRCA2 genes, respectively. of prior hypotheses, unlike standard linkage analysis.
In turn, the initial identification of the BRCA1 gene and subsequent The GWAS strategy is based on testing indirectly for the actual
removal of BRCA1-linked families from remaining data sets provided genetic variant using surrogate markers in data sets of cases and controls
further useful enrichment for BRCA2-linked families.48,49 For the breast to highlight regions harboring susceptibility alleles.18 Thus the functional
cancer susceptibility genes BRCA1 and BRCA2, founder mutations marker directly responsible for the effect does not have to be actually
have been identified in distinct populations across the globe,50,51 a tested in the scan. Instead, its surrogate, which is in LD as measured
phenomenon most notably observed in Jewish Ashkenazi families.52,53 by a high correlation (r2 > 0.8), can be replicated in subsequent studies,
The three common founder mutations in this population, BRCA1- pointing to the susceptibility allele(s). Hence, substantial effort is
185delAG, BRCA1-5382insC, and BRCA2-6174delT, have a combined required to fine-map the region—that is, to find all of the correlated
population prevalence of 2% to 2.5%. With these observations in variants before choosing which ones to evaluate in follow-up studies.
mind, investigators have frequently sought families for genetic mapping In many regions, variants with lower minor allele frequencies are also
studies from regions of the world where marriage between related highly correlated with the GWAS marker, and on occasion a less
individuals is not discouraged and where geographic barriers have common allele with a stronger effect has been identified, yielding a
restricted gene flow. so-called synthetic association.62 However, to date, the majority of
Locus heterogeneity, the presence of deleterious alleles associated susceptibility alleles cannot be explained by less common variants
with many genes in a population, can be reduced by studying families with stronger effects.63
from isolated or inbred populations. Fewer disease alleles are predicted Patterns of LD vary among populations, both in specific regions
to segregate with a particular phenotype in a population derived from and across the genome.23 This results in major differences in MAF
a limited number of founders. Studies of colon cancer in Finland and among populations on a per-SNP basis, as well as the intervals of LD
breast cancer in Iceland as well as Ashkenazi Jewish populations illustrate defined by recombination hot spots. In rare circumstances, the dif-
this point. In Finland, two variants in the DNA mismatch repair gene ferences between incidences in disease, such as the incidence of prostate
MLH1—mutations 1 and 2—account for 51% of all Finnish families cancer between men of European and of African ancestry, can be
with verified or putative cases of hereditary nonpolyposis colorectal explored by using admixture linkage analysis. Notably, one of the first
cancer.54 Nineteen mutation 1 and six mutation 2 families were further GWAS signals for prostate cancer on 8q24 was also found by admixture
investigated by haplotype analysis using 15 microsatellite markers analysis.64–66
surrounding the MLH1 locus. The presence of two distinct large The basic principle behind an association study is that there is a sta-
conserved disease haplotypes, one in mutation 1 and the other in tistically significant difference in the distribution of one or more alleles
mutation 2 families, indicated that these families are likely to descend between cases and controls, indicating the location of a susceptibility
from two distinct common ancestors born in the 16th and 18th allele that contributes to cancer risk. Although association studies can
centuries, respectively. uncover highly penetrant mutations that are strongly correlated with
cancer risk, much as the family linkage studies discussed earlier, their real
Principles of Association Testing usefulness is in finding common low-penetrant alleles that contribute
to cancer risk. This is reflected in the fact that the estimated odds
In the past, genetic linkage analysis was the workhorse for discovery ratios for alleles found in GWASs are low, almost always estimated at
of CPGs, but the new tools of massively parallel genotyping and less than 1.3, and thus such alleles are not efficiently detected through
sequencing have changed the landscape of discovery, enabling pursuit linkage.12 These low-effect susceptibility alleles are neither necessary
of complex diseases, namely those in which multiple distinct genetic nor sufficient for development of a cancer. Indeed, for most forms
regions plus environmental factors contribute to risk for disease. of cancer we expect that there are many susceptibility alleles, each of
Linkage analysis has been of limited success in studies of common which contributes a small effect to the disease.
and/or complex diseases, primarily because of insufficient power Investigators use one of several commercial SNP microarray chips
to detect association of smaller effect sizes for multiple susceptibil- to scan the genome and compute rank P values for prioritization of
ity alleles and complexities in phenotype assignment. Risch and promising genetic markers for replication studies, which are typically
Merikangas55 pointed out the shortcomings of linkage for complex performed using one or more independent data sets of cases and
disease mapping and made the case for association analyses in populations controls. Because of the daunting challenge of false positives in testing
of unrelated subjects. Their projections have been borne out in the age so many markers, a community-wide standard has emerged, one that
of GWASs.11,56 protects against false-positive findings; this is achieved when studies
In the failed candidate gene era, investigators chose specific variants report markers that surpass the threshold of genome-wide significance,
based on prior hypotheses and analyzed underpowered studies, yielding now generally defined as a trend association test with a P value of 5 ×
a sea of false-positive reports. The most notable examples include 10−8. This can be reported in the primary scan, if large enough, or in
common variants in NAT2 and GSTM1 in bladder cancer and alcohol a combined analysis of the scan with the replication sets. More recently,
dehydrogenase genes (ADH1B and ADH7) in aerodigestive cancers.57–59 large meta-analyses that combine data from several independently
As the annotation of the human genome emerged with first the collected datasets are used for confirmation. Because GWASs discover
International HapMap Project and then the 1000 Genomes Project, loci that are highly associated with specific markers, surpassing this
the approach shifted toward use of surrogate markers across the genome threshold ensures that there is a very small probability of a false-positive
designed to capture the majority of common genetic variation using result. This is particularly important given that extensive follow-up
the GWAS design.60 analyses are required to map and investigate the biologic underpinning
GWASs have been successfully applied across a spectrum of diseases of the susceptibility allele.
and traits, yielding thousands of loci harboring common susceptibility Because GWASs can be effectively scaled to accelerate discovery,
variants, associated with hundreds diseases and traits.56 In 2017 there the major challenge ahead is to determine how to establish the critical
were more than 725 independent cancer susceptibility loci associated connection between the genetic markers of susceptibility alleles and
with at least 30 different cancers, most observed in individuals of carcinogenesis. The rapid pace of discovery with use of the GWAS
European ancestry. The approach is based on an initial scan across approach has not been matched by research to interpret and understand
the genome followed by independent replication.61 The results of the functional significance of different alleles correlated with cancer
scanning hundreds of thousands of SNPs are analyzed with an “agnostic” phenotypes.67 The gap between the number of new independent
statistical approach, using logistic regression analysis that is often, but markers and a biologic understanding of the loci continues to widen
not always, adjusted for critical covariates (e.g., age or subtle differences at an accelerated pace. This is a result of the differences in scientific
330 Part I: Science and Clinical Oncology

22
21
Mosaic Gain

1
20

10
20
30
30
40

40
20

50
10

60
0

70
80

100
40

90

110
0
30

120
20
10

130
140
150
0
60

160
Mosaic Copy Neutral

50
19

170
40
30

180
20

190
10

200
210
0
60

220
50

230
40

240
0 3
Mosaic Loss

20
10
18

0
10
0

20
70

30
60

40
50

50
40

60
30

70
20

80
10

90

2
11 0
0

10
17

0
70

0
12
60

50 0
13 0
14 0
40

30
20 15 0
16 0
17 0
10 0

18 0
19 0
16 60
70
80
20 0
21 0
50 22 0
40 23 0
30 24
20 0
10 10
0 20
100 30
90 40
80 50

15 60
70 60
70
50
40
80
90 3
30 100
110
20
120
10
130
0 140
100 150
90 160
80 170
180
14 70
60 190
50
40 0
30 10
20 20
10 30
0 40
50
110 60
100 70
90 80
80
70
90
4
13
100
60 110
50 120
40 130
30 140
20 150
10 160
0 170
130 180
190
120
110 0
100 10
90 20
80 30

12
70 40
60 50
50 60
70
40
30 80
20
10 10
90
0
5
0 11
12 0
0 0
13 13
0 0
12 14
0 0
11 15
0 16 0
10
90 17 0
0
80 18
70 0

11
0
60 10
50 20
40 30
30 40
20

50
10
0

60 0
7
12 0
13

80
11 0

90 0
10 0

10 0
0

6
11 0
90

12 0
80

13 0
70

14
60

15 0
50

16
10

0
40

17
30

0
0
20

10
10

20
0

30
140

40
50
130

60
120
110

70
100

80
90
90

100
80

110
70

120
60

130
50

140

7
40

150
30

0
20
10

10
0

20
9

140

30
130

40
110
120

50
100

60
70
90
80
8

Figure 21.8  •  Clonal somatic mosaicism of large structural events, based on genome-wide association study data from over 125,000 patients and control
subjects, the distribution of types of events (loss, gain, and copy-neutral loss of heterozygosity) across the autosomal chromosomes. Autosomal events are
depicted for 1315 events greater than 2 Mb (0.75% overall) (Data from reference 118).

approaches; the GWAS approach is scalable if surrogate markers are alcohol, or weight/height) are collected, the latter to study the degree
used across the genome so that with larger sample sets further discovery to which an exposure is associated with disease incidence. Exposure
is possible. However, despite the fact that SNP chips with over 1 is measured at baseline, when the cohort is initially established, and
million markers are available, rarely is the marker also the functional may be updated over the period of follow-up for those exposures
variant. Thus follow-up analyses are required to characterize each that may change over time. The advantages of cohort studies include
susceptibility allele. The patterns of genetic variation vary greatly across minimized information and selection biases and the ability to directly
regions thus requiring a detailed fine-mapping of each region prior to calculate disease incidence in exposed and unexposed groups, and
choosing individual genetic variants for laboratory study (Fig. 21.8). thus the RR and absolute risk (attributable risk, AR). Disadvantages
include the fact that prospective cohort studies are expensive and
Study Design and Association Studies time-consuming and large numbers of study subjects are typically
required for sufficient numbers of outcomes (i.e., cancer cases) to be
For association studies, two primary types of study design are typi- obtained to have adequate power to determine associations. Loss of
cally used: cohort and case-control studies. Recently, however, the subjects to long-term follow-up is also an issue. Cohort studies can
discovery of GWAS regions have come at the expense of epidemiologic be retrospective (i.e., the exposure and subsequent development of
rigor with respect to control selection. In a cohort study, subjects are the disease occur before the study begins).
selected, individuals with the disease of interest (i.e., prevalent cases) Case-control studies differ from cohort studies in that the selection
are excluded, one or more exposures of interest are measured and of subjects is based on disease status, providing an opportunity to
monitored over time, and biospecimens are archived. Cancer and examine multiple risk factors. They are generally either population-based
intermediate outcomes, such as risk factors for cancer (e.g., smoking, or hospital-based. Selection of patients and control subjects should
Discovery and Characterization of Cancer Genetic Susceptibility Alleles  •  CHAPTER 21 331

account for confounding factors such as age, sex, race, and ethnic 1.8, such as in Ewing sarcoma, a rare pediatric cancer, yet for the
background. Controls must be selected from the same underlying adult cancers the norm is low.73 Among the most significant is testicular
population from which the cases were ascertained in order to avoid cancer, known to have a high heritability in families and twin studies;
stratification, either by differences in genetic background or by risk GWASs identified a susceptibility allele with a per-allele effect estimate
or exposure. of greater than 2.5 on chromosome 12 (KITLG).74,75
Population-based case-control studies draw on a well-defined source Initially, the commercial SNP microarrays chips were designed to
population such as a particular geographic region defined by state, efficiently capture SNPs with MAFs greater than 5% to 10%. It is
county, or city for ascertainment of both case patients and unaffected anticipated that new susceptibility alleles in the range of 1% to 10%
control subjects. Control subjects should be selected from the same will be discovered with use of new SNP microarrays with lower MAF
source population by a method designed to randomly sample individu- content. Most of resulting alleles are expected to have small effect
als; historically this has been random digit telephone dialing. Selection sizes. In retrospect, these findings are understandable in light of the
bias, in which selection of case patients, control subjects, or both is power estimates for discovery of new alleles because small effect sizes
influenced by prior exposures, is a particular concern in case-control in less common alleles require larger sample sets. Less than 5% of the
studies. For instance, many studies have shown that nonparticipants susceptibility alleles conclusively established in cancer GWASs have
in such studies are more likely to smoke than individuals who agree led to determination of an actual coding change in a gene. Most
to participate. Therefore there is concern that participants may be regions map to regulatory regions in and around well-recognized genes,
more health conscious than nonparticipants. Also, in recent years the a few of which have been implicated in cancer biology. Nearly one-
lack of a landline phone in many homes has hindered efforts to enroll quarter of markers localize to intergenic regions, namely regions between
randomly selected controls, potentially compromising the unbiased genes in which all of the SNPs in strong LD (r2 > 0.8) fail to localize
nature of control enrollment. in or near a known gene. These findings suggest that a fraction of the
In comparison, hospital-based, case-control studies enlist a sequential genetic contribution to cancer may reside in alterations in the regulation
series of patients who are admitted to the hospital or clinic during a of known or novel pathways.
specific period. Case patients are enrolled because they have the cancer A small fraction of susceptibility alleles have been associated with
of interest, whereas control subjects are determined to be cancer free, more than one distinct cancer type. These are highly informative and
although they may be patients at the same clinic or hospital for unrelated reveal possible common carcinogenic mechanisms underlying distinct
reasons. Depending on the clinic or hospital from which patients are cancers. Included in these results are sometimes alleles within the
drawn, disease presentation, severity, and treatment outcome may be HLA regions, particularly for cancers of the immune system or those
nonrandom. Often cases are drawn from so called “high-risk” clinics, driven by viral infections. Commercial arrays and imputation provide
whereas control subjects may come from nonrandom sampling as inadequate discrimination of the region, particularly because of its
well. Controls from clinics can later be determined to be suboptimal complex structure; hence other technologies will be required to
because they have comorbidities or exposures that were initially thought comprehensively explore this region in cancer susceptibility. The region
to be independent of the disease under study but are later found to flanking the MYC oncogene on 8q24 is also extremely complex; it
be confounders.68–71 Confounders are factors that are associated with harbors at least a dozen independent loci associated with prostate
the exposure as well as the disease. Age is a frequent confounder for cancer, as well as loci associated with multiple other cancers including
cancer studies because the incidence of cancer advances with age, and breast, colon, and bladder cancers and chronic lymphocytic leuke-
often with level of exposures. An important source of bias specific to mia.64,76 The MYC oncogene itself is a plausible candidate gene, and
case-control studies and retrospective cohorts is recall bias, in which work has suggested that allelic differences in enhancers could directly
study subjects inaccurately or incompletely remember information or indirectly interact with MYC, although there is little to suggest
related to disease susceptibility such as environmental or lifestyle factors that coding changes are important.77–79
(e.g., diet, smoking, birth control history, exercise). This introduces A region on 5p15.33 harbors a range of susceptibility alleles for
error into the calculation of the association between the exposure and many cancers, including rare and common SNP alleles. So far, more
the disease because of the inability to statistically adjust for the effects than 10 distinct cancers have been associated with the region by means
of confounders. of GWASs, including both breast and prostate cancer, and there are
Until the age of GWASs, there was a vigorous debate in candidate at least five independent alleles in this region, which contains the
gene studies over the impact of possible differences in underlying telomerase gene (TERT).80,81 Rare mutations in TERT track with
population substructure. The testing of thousands of uncorrelated dyskeratosis congenita (an inherited bone marrow failure syndrome),
SNP markers in GWAS have provided investigators with the opportunity idiopathic pulmonary fibrosis, acute myelogenous leukemia, and chronic
to sift out individuals who differ substantively across the genome. lymphocytic leukemia.82,83 The pleiotropy in this region hints at complex
Several different analytic algorithms can distinguish the degree of gene-gene or gene-environment interactions. For instance, a protective
admixture across chromosomes, based on referential continental allele for one cancer appears to be a susceptibility allele for another
population sets (e.g., International HapMap Project or 1000 Genome cancer; it is remarkable that the same allele has inverse effects for two
Project).72 In fact, this has been used to map a handful of regions that skin cancers, basal cell carcinoma and melanoma.84
appear to be specific to one ancestral group compared with another. The discovery of new susceptibility alleles is now being driven by
the use of imputation, a technique based on computational algorithms
Association Studies in Cancer that infers untested and highly correlated SNPs based on reference
data sets (e.g., International HapMap Project or 1000 Genome
To date, over 725 genomic regions have been conclusively established Project).85 It is predicated on the observed LD between SNPs in refer-
(i.e., achieving the threshold of genome-wide significance, which ence populations drawn from distinct regions. A notable exception is
protects against the likelihood of a false-positive discovery) for more the recent identification of a rare SNP on 8q24 conferring susceptibility
than two dozen distinct types of cancer.12 The majority of susceptibility to prostate cancer, which was discovered and characterized in the
alleles are specific to one cancer, but roughly 10 to 15 are pleiotropic, isolated population of Iceland.86
indicating that the effect can be observed for more than one type of The fine-mapping of susceptibility alleles has led to the discovery
cancer. Nearly all cancer susceptibility alleles discovered by GWASs of independent signals nearby, indicating that more than one allele
have an MAF greater than 10% with a subset in the 5% to 10% contributes to cancer risk. The first prostate cancer susceptibility allele
range as reported in large studies. Overall, the per-allele estimated on 8q24 has now blossomed to more than a dozen separate alleles,
effect size has been small, with odds ratios between 1.1 and 1.3. several of which are distinctive to African Americans. This latter
Several of the alleles reported in pediatric cancers have risks of 1.6 to observation could partially explain the substantial increase in incidence
332 Part I: Science and Clinical Oncology

in prostate cancer in African American men compared with those of Unraveling the Cancer Biology of Cancer
European ancestry.87 An intergenic region of 11q13 harbors several Susceptibility Alleles
independent prostate cancer susceptibility alleles88 but also nearby
distinct alleles for renal and breast cancer, each of which works through One of the major surprises of cancer GWASs is that only a small
separate mechanisms.89,90 fraction of the more than 725 susceptibility alleles map to well-
The formation of numerous international consortia promises to characterized genes already implicated in cancer biology. Notably, the
maintain the pace of discovery of common susceptibility variants, not regions defined by GWAS markers do not harbor genes that are
only in populations of European ancestry but also in those of other significantly mutated, especially as drivers, in large landscape charac-
ancestry, including studies from Asia and Africa.91 To date, the majority terizations of somatic mutations.107 This is in distinct contrast to the
of reported GWASs have been in subjects of European ancestry, but high overlap between CPGs and somatic drivers of cancer.13 Moreover,
progressively more studies have been conducted in subjects of Asian the sets of GWAS signals by specific cancers generally do not correlate
and African ancestry. In a few instances, alleles have been identified with specific patterns or processes fundamental to the cancer. In this
in specific populations with substantially higher frequency. For instance, regard, there is no evidence for enrichment for common variants in
a region of chromosome 17q21 harbors a prostate cancer susceptibility DNA repair pathways that are clearly associated with risk for common
allele in African Americans, for which the best marker has an MAF or rare cancers. New discoveries point toward possibly new biologic
of 5%, whereas in individuals of European background it is below mechanisms underlying cancer susceptibility. Because the interrogation
1%.92 Additional alleles in 8q24 have been reported in men of African of each region is complex and requires extensive bioinformatics,
American background, but they do not fully explain the difference fine-mapping, and laboratory analysis specific to the region of the
in incidence. genome, it is understandable why only three dozen susceptibility alleles
Differences in study design can lead to conflicting conclusions, have been adequately interrogated. Typically each region harboring
including the biologic interpretation of the association. Initially, two one or more susceptibility alleles has a unique local pattern of LD.
distinct GWASs in prostate cancer reported contrary results for alleles It is notable that the breast cancer susceptibility locus BRCA1 was
on chromosome 19q13.33, which harbors the gene responsible for first mapped in 1990 and that more than two decades later the biology
prostate-specific antigen (PSA).93,94 In a GWAS using cohort studies, of BRCA1 and the consequences of germline mutations are still under
the effect appears to be related to PSA levels, whereas in a study using investigation, but specific therapeutic interventions are now clinically
advanced cases and control subjects with low PSA levels, the effect points available.
toward carcinogenesis. Follow-up studies including fine-mapping of the To unravel the biologic underpinnings of cancer susceptibility
KLK3 gene (which encodes PSA) and further replication indicate that regions, investigators are turning to new resources and approaches.67
the locus could be associated with both prostate cancer susceptibility Consortium guidelines have been developed to accelerate the pace of
and PSA levels.95,96 Recently, large GWASs have identified several dozen characterization of cancer susceptibility alleles, beginning with fine-
variants specific to the level of PSA, establishing a foundation for mapping using new tools.108 For instance, progress in understanding
studying the genetics of a complex biomarker for prostate cancer risk.97 regions identified by GWASs has recently received a major boost by
the publication of the ENCODE Project (Encyclopedia of DNA
Genetic Architecture Underlying Elements), a far-reaching project to map the functional genome.109
Cancer Susceptibility The ENCODE papers have begun to shed light on the biology of
the regulation of the genome, specifically cataloging signposts and
The underlying genetic architecture can differ by cancer sites with markers of biologic activity. It has sharpened our vision of the inner
respect to the relative contribution of common variants with small workings of how the genome functions. With ENCODE and its tool
effects through the spectrum to rare variants with strong effects (e.g., packages, it is possible to trace the errors that contribute to cancer
CPGs). As indicated in Fig. 21.1, the emerging picture suggests that susceptibility. It is now possible to look at patterns of regulatory
there are variants of low effect size, commonly seen in the population, variation in individuals and across populations. With integration of
that contribute a fraction of the heritable risk for the cancer. Although experimental data and computational tools, several in silico programs
ongoing studies are expected to generate a more comprehensive catalog should enable assessment of known susceptibility regions and prioritize
of variants in each class (e.g., common and rare), early modeling of variants for further study, with a higher value assigned to those with
empiric data suggests differences among cancers.98–101 In prostate cancer, functional significance.110 New insights into breast cancer susceptibility
rare, highly penetrant mutations account for a very small component, loci identified by GWAs have been generated through use of ENCODE
whereas the known common variants can account for at least one-third insights together with laboratory observations.
of familial risk—namely, the estimated risk of other first-degree relatives The search for functional variants underlying GWAS signals has
of a patient with a newly diagnosed cancer.102 For breast cancer, there uncovered new insights with possible clinical implications. For example,
are more than a dozen highly penetrant CPGs, together accounting investigation of the bladder cancer GWAS signal on 8q24.2, followed
for more than 15% of the familial risk, whereas the common variants by RNA sequencing and genetic and functional analysis, identified a
found by GWASs account for one-third of the familial risk.103 NGS variant that is strongly associated with increased mRNA expression
studies in families and now population-based studies have identified of the prostate stem cell antigen (PSCA) gene.111 Furthermore, this
breast cancer susceptibility alleles with moderate effects (e.g., estimated variant creates an alternative translation start site and leads to increased
RR between 2 and 4).104 It is interesting to note that the majority of expression of PSCA on the cell surface, where it can be subjected to
these susceptibility alleles map to genes in pathways related to BRCA1 immunotherapy with anti-PSCA antibody, an emerging therapy for
activity, including ATM, BRIP1, CHEK2, ERCC2, PALB2, RAD51C, several cancers. Because the genotype of the GWAS variant is predictive
and RAD51D.105,106 The rate of discovery of new mutated genes for of PSCA protein expression, a genetic test could be used to identify
other cancers, both rare and common, continues to accelerate. Cor- patients with bladder cancer who could benefit from the anti-PSCA
roborative laboratory work can supplement analyses in families and therapy.
population-based studies. For instance, a rare variant in the MITF A few of the GWAS susceptibility alleles have been conclusively
gene that has an allele frequency of approximately 1% increases the shown to interact with environmental exposures. An SNP in NAT2
risk for melanoma, a dangerous skin cancer.47 After familial testing is important for bladder cancer susceptibility only in people who have
showed incomplete penetrance and association studies suggested an “ever smoked,” whereas in never-smokers there is no effect.112 The
effect in unrelated populations, laboratory investigation revealed that GWASs of lung cancer, strongly driven by tobacco exposure, have
the mutation resulted in impaired SUMOylation and differentially revealed that select alleles are operative only in smokers whereas others
regulated MITF targets. are observed only in nonsmokers. The strongest signal seen in smoking
Discovery and Characterization of Cancer Genetic Susceptibility Alleles  •  CHAPTER 21 333

lung cancer GWASs on chromosome 15q25 is not evident in nonsmok- studies it is possible to detect somatic events well in advance of the
ing women in Asia, suggesting that the contribution of 15q25 is not diagnosis of chronic lymphocytic leukemia.117 A more refined under-
associated with lung cancer, independent of smoking.113,114 standing of mosaicism in the aging genome promises new insights
Previously, many investigators have attempted to elucidate the into genomic instability as it relates to carcinogenesis, especially in
functional genetic variant and a mechanism underpinning the genetic the hematopoietic system. Large-scale analyses of exome sequenced
association between clearance of hepatitis C virus (HCV), a risk factor subjects have indicated that point mutations frequently arise as mosaic
for liver cancer, and genetic variants upstream of IFNL3, previously events in circulating blood, particularly involving genes critical in
called IL28B on chromosome 19, which were discovered in several hematopoietic diseases, such as DNMT3A, TET2, and TP53. Ongoing
GWASs. However, RNA-sequencing analysis in primary human studies are designed to elucidate the scope and possible risk for one
hepatocytes has uncovered a new gene, IFNL4, which is generated or more cancers related to mosaic events, from single-nucleotide to
by a complex dinucleotide insertion/deletion variant in LD with large structural events involving an entire chromosome.124
associated GWAS markers.115 The deletion allele of this variant causes GWASs in large consortia have also looked at intermediate exposures
a frameshift that leads to a novel protein that induces an interferon-type strongly implicated in cancer risk, such as body mass and other
response. Genetic analysis showed that the IFNL4 genetic variant has quantitative traits (e.g., tobacco use and alcohol consumption), yielding
a strong effect on HCV clearance, especially in individuals of African new insights into biology. The study of height in hundreds of thousands
ancestry. The strength and the effect size of the genetic association in of subjects with GWAS data has uncovered novel pathways and provided
IFNL4 leading to spontaneous and treatment-induced clearance of a more stable assessment of the contribution of common SNPs (MAF
HCV are large enough to warrant pursuit in clinical studies. Use of >5%) to the trait. Polygenic models for many SNPs, each with small
this complex polymorphism is under active study for clinical usefulness effects not yet conclusively discovered by GWAS, define the fraction
in directing therapy and outcomes in HCV treatment. of heritability for important traits such as height, weight, smoking
During the practice of applying stringent quality control metrics behavior, and, more recently, PSA levels and breast density on mam-
to GWAS data, a pattern of unexpected deviations emerged that have mography, all established to be associated with risk of cancer.97
led to the investigation of the detection of genetic mosaicism, defined As the number of individual cancer susceptibility alleles has increased
as two or more distinct karyotypes within an individual.116 It is still with larger GWAS studies, it is now possible to conduct polygenic
not clear whether the presence of large structural mosaicism is associated risk score (PRS) analyses, both in discovery sets and in validation
with risk for nonhematologic cancer, as it is for chronic lymphocytic studies to define the fraction of the genetic architecture explained by
leukemia.117 With use of SNP data, it is possible to estimate that common variants, each contributing only a small fraction.125 Sufficiently
approximately 1% of the adult population harbors one or more large large GWAS efforts have been reported, and models for risk stratification
autosomal somatic events (see Fig. 21.8).118,119 Mosaic events for the can be generated by using empiric data to calibrate the model. PRS
sex chromosomes are even more frequent; roughly 15% to 20% of analyses can also be applied to large population-based studies to identify
men older than 50 years have mosaic loss of the Y chromosome, which the fraction of the population at highest genetic risk. In turn, this
is particularly higher in smokers.120 In fact, the propensity to develop fraction can be targeted in order to reduce modifiable risk factors and
mosaic loss of Y has been mapped by GWAS to nearly 20 distinct decrease the burden of the cancer. Ongoing studies are evaluating the
regions in the genome.121,122 impact of risk reduction programs targeting those at highest risk by
X chromosome mosaicism is observed in women and almost always PRS analysis in order to reduce the incidence of breast cancer, a cancer
involves the inactive X chromosome, comparable to the reported higher with a high absolute risk in the population (Fig. 21.9).98,103,126,127 This
frequency of somatic mutations in the inactive X chromosome of approach is not unique to breast cancer and is being pursued for other
female cancer genomes.119,123 For hematopoietic malignancies, in cohort cancers, such as prostate, colon, and bladder cancers.

Distribution of modifiable risk by deciles of nonmodifiable risk

30 Heavy drinker,
0

smoker, obese/HRT

25
0 0 0

20
Absolute risk (%)

0000 0 0
0 00
0

15
00 0
00 00
00 0 0

10
00 0
0000 0
0000 0

5 Never drinker or
smoker, no HRT use,
healthy weight
0
1 2 3 4 5 6 7 8 9 10
Decile of nonmodifiable risk
Figure 21.9  •  The impact of changes in lifestyle could be targeted at women with higher genetic risk for breast cancer. The distribution of deciles of
women based on the polygenic risk score for known breast cancer susceptibility alleles (n = 77). HRT, Hormone replacement therapy. (Data from reference 98.)
334 Part I: Science and Clinical Oncology

Clinical Implications of Cancer Susceptibility Alleles This transition will accelerate the identification of new potential
variants, and the number of both uncommon and rare variants will
It is clear from the association of many known cancer susceptibility increase. In large-scale sequencing of the exome (defined as the targetable
alleles with more than one cancer type that there may be distinct exons of known genes), there are thousands of novel variants per
alleles that influence the etiology of a cancer and a different set that individual, some reflecting rare and population-private variants.
influence clinical outcomes, such as progression, treatment response, Consequently, the statistical challenge of parsing rare variants in
or metastasis. More productive have been studies that used clinical unrelated population studies is daunting, especially as the MAF decreases
indicators such as high Gleason score as a surrogate for aggressive because the sample sizes required to detect alleles with low to moderate
disease risk. For instance, studies have identified a modest number of effect sizes becomes larger. Large databases and studies should be
genes associated with metastatic disease or death from prostate cancer developed to conduct larger discovery analyses. For the foreseeable
versus indolent disease.128,129 In the childhood cancer neuroblastoma, future, the discovery paradigm has shifted to include correlative labora-
regions have been identified that are associated with more aggressive tory confirmation of promising variants. At the same time, the value
disease.130 of large-scale data sharing, including family history, cannot be
The discipline of pharmacogenomics has been accelerated by the understated, because it will more quickly allow investigators to
same trends that have driven the discovery and characterization of determine the pathogenicity of most variants. This in turn can provide
germline genetic susceptibility alleles.131 Pharmacogenomics seeks to evidence for clinicians to provide state-of-the-art surveillance and
investigate the contribution of germline genetic variation to response interventions, especially in high-risk settings. In the high-risk setting
rates or toxicity associated with therapeutic modalities (e.g., medicinal, of Li-Fraumeni syndrome, early and active surveillance has an important
surgical, or therapeutic radiation). For instance, susceptibility alleles prognostic value.
have been identified for second cancer risk and include both a
highly penetrant mutation of the retinoblastoma gene (RB) leading
to osteogenic sarcoma and common SNPs on chromosome 1q41 CLINICAL RELEVANCE AND APPLICATIONS
associated with breast cancer risk in pediatric survivors after radiation Genetic Counseling and Testing
therapy.132
The effect of highly penetrant germline mutations on cancer In advising patients whether it is appropriate to consider genetic testing,
outcomes and more specifically cancer risk has generated newfound it is important to remember that many currently available tests have
interest in the contribution of the germline mutations to cancer limitations. Clinical validity is the term used to describe the predictive
outcomes across all cancers. In women with invasive ovarian cancer, value of a test for clinical outcomes. It is affected by both the sensitivity
those with germline mutations in BRCA1 or BRCA2 carry an improved and the specificity of the test, as well as a host of factors that are
5-year overall survival.133,134 On the other hand, few of the common beyond laboratory control, such as penetrance of the mutant allele or
susceptibility alleles also map to outcomes, indicating that for the the value of a set of SNPs for a risk profile. Moreover, the degree of
common variant component the contributions to etiology differ from correlation between a variant and cancer risk in any given patients
those related to outcomes. One exception is BRCA2 mutations and can be influenced by genetic background, environmental exposures,
prostate cancer, in which germline susceptibility alleles have been or both. Because nearly all mutations associated with cancer susceptibil-
shown to be associated with early onset, rapid progression, and poor ity genes are not fully penetrant, clinicians will have to continue to
outcomes. educate themselves and their patients concerning the incorporation
of new science and methods that offer improved sensitivity and/or
Next-Generation Sequencing Analysis specificity as a result of modifiers of risk (either genetic or environmental
exposures). In this regard, it is important to help patients understand
With the availability of NGS platforms, investigators are likely to that there may be a small group of individuals who, even if they live
be more successful in their search for less common and rare vari- into their 80s, will not get cancer despite carrying protein-truncating
ants that explain a proportion of heritability of cancer susceptibility. mutations in a gene associated with high risk for a particular cancer.
NGS platforms perform massively parallel sequence analysis of The exponential increase in available information in the community
major fractions of the genome with a high degree of redundancy, and in a patient who undergoes NGS will require new paradigms to
which is needed to minimize the error in calling sequence vari- annotate, present, and revisit previously stored sequence data. It will
ants. These technologies are reshaping the scope of genetic studies, also be critical for clinicians and researchers to be well educated in
in both high-risk families and now in population-based studies. informatics issues related to the privacy of patient sequence data, as
Often, sequencing of tumors is conducted without accompany- well as the capacity to seek guidance and well-annotated information
ing germline material, yet new algorithms can effectively identify that can assist in making informed decisions.
many of the germline variants, especially if the sequence coverage is The National Society of Genetic Counselors defines genetic
sufficiently high. counseling as “the process of helping people understand and adapt
The shift to population-based sequencing, especially in large referral to the medical, psychological, and familial implications of the genetic
cancer centers, has already uncovered a surprising observation: roughly contributions to disease.”136a Therefore the approach should be offered
10% of children and adults with cancer harbor one or more CPGs.135 in consultation with genetic counselors who aim to help patients (1)
In pediatric cancer, the discovery of a CPG could be a sentinel for comprehend the medical facts and risks associated with their disease;
the entire family, particularly as the parents enter the age range of (2) understand potential alternatives for dealing with both risk of
highest risk for many of the CPG-related syndromes. In osteosarcoma, disease and recurrence; (3) choose a clinical course that best meets
the most common pediatric bone tumor, nearly 5% of children without their needs; and (4) when needed, provide support and guidance for
a family history harbor a TP53 mutation, characteristic of the Li- patients experiencing difficulty in dealing with unexpected results.
Fraumeni syndrome.136 The interpretation of new variants is daunting Patients often approach genetic testing and the likelihood that they
and accordingly is stretching the paradigm of genetic counseling, carry a deleterious inherited mutation with strong preconceived notions
which will have to evolve to address the ongoing analyses possible in based simply on intuition, which a trained genetic counselor must
a patient with the entire genome or exome sequenced. Similarly, clini- first overcome.
cians will have an increasing role in discussing genetic findings, Patients will frequently approach clinicians with questions about
determining actionable results, and referring patients to health care genetic testing opportunities for specific cancers, or cancer overall,
professionals who can assist with further testing, monitoring, or effective particularly because the set of targeted genetic tests varies greatly by
lifestyle changes. center and company. Although formal regulations exist for interpretation
Discovery and Characterization of Cancer Genetic Susceptibility Alleles  •  CHAPTER 21 335

of results of tests for highly penetrant mutations, new incidental findings WHAT THE FUTURE HOLDS
will require adequate education and direction in assisting clinicians
in guiding patients to the optimal decisions, which may sometimes The sequence of the human genome has been referred to as an “instruc-
require additional expertise, especially if the incidental findings point tion book for human biology,”137 and it has become clear that there
to nononcologic challenges. Moreover, germline testing can induce are many dynamic factors that interact in regulating and responding to
patients at risk to undergo more vigilant screening, such as frequent the human genome—including environmental, behavioral, and lifestyle
colonoscopy examinations for patients at risk for colon cancer. The factors. Locked within the sequence of each individual’s DNA is the
impact on one’s quality of life is an individual decision based on many genetic code necessary to develop a complete and healthy individual,
factors, and the substantial increase in data available through germline but encoded as well is the sequence-level variation that will determine
sequencing should be carefully weighted in recommending individual each person’s susceptibility to a host of diseases and outcomes.
medical decisions. It is important to note that some patients will The results of the comprehensive sequence analyses of paired sets
strongly wish to retain their “right not to know,” wishing to postpone of genome (e.g., germline and somatically altered cancers) have revealed
any decision making, such as whether to undergo a prophylactic a large spectrum of mutational events and epigenetic alterations.138
oophorectomy in the face of a BRCA1-associated breast cancer, until These newfound insights will provide opportunities to carefully unravel
a cancer diagnosis appears. One final consideration for clinicians and the interrelationship between the germline susceptibility alleles and
genetic counselors is anticipation that the burden that diagnosis of a cancer etiology plus progression. Already we can see somatic alterations
deleterious genetic mutation places on parents, who inevitably struggle that can explain exceptional responses to targeted therapies.139 A more
with factors related to when and if children should be informed of complete understanding of the molecular pathways involved in cancer
their potential risk. susceptibility will suggest avenues for the development of both methods
The identification of cancer susceptibility alleles has prompted of diagnosis and treatments. Identification of specific genes offers the
rapid transition of common SNPs to individualized disease prevention promise of genetic testing to individuals at risk, as well as the hope
and public health policy. With the identification of cancer susceptibility for targeted therapeutics.140 Finally, understanding of sets of specific
SNPs, there has been a debate as to when and how to transition them variations offers great promise for 21st-century precision medicine
into clinical care. Because the cumulative set of SNPs for any one and precision prevention141 in which lifestyle, diet, and preventive
disease now provides a substantial fraction of the population risk for therapies come together to allow patients a full spectrum of choices
a cancer, ongoing studies should provide a sound foundation for for maintaining their personal health.98
developing new paradigms in precision prevention. Eventually it may It is clear that the Human Genome Project has had and will continue
be possible to reclassify high-risk versus low-risk individuals in anticipa- to have a profound effect on human health and biology. What remains
tion of deciding on a preventive or early-detection program. It is also to be seen is the rate at which the successes of the Human Genome
important to keep in mind that SNPs may not be informative for a Project will move from bench to bedside. In a sense, that rate will be
given cancer because differences in the underlying genetic architectures determined by practicing physicians. Knowledge of the underlying
may influence the putative usefulness of introducing sets of independent principles of genetic analysis is fundamental for today’s practicing
SNPs into clinical practice. clinician. The ability to accurately record family history and medical
With advent of NGS technologies introduced into the clinical record data affects the integrity of all subsequent studies for which
venue, the clinician may be faced with interpretation of thousands of those data are used. An understanding by physicians of the findings
variants of unknown significance. For example, which truncating generated through both association studies and family-based studies
mutations in the BRCA1 gene confer high risk for breast and ovarian is key to both moving research forward and discovering what in turn
cancer? There are more than 400 independent missense changes cata- must be tested and carefully integrated in clinical and public health
logued, only a fraction of which (predominately in the RING finger paradigms. The challenge of clinically translating the information
domain and the C-terminal region of the protein) have been conclusively derived from both the germline and cancer genomes will continue to
linked with disease risk. Many, including some amino acid deletions, be daunting as we try to carefully make individual decisions by using
are inconsequential polymorphisms that do not affect protein function, data generated from increasingly larger studies, of often differing study
nor do they likely increase risk for cancer. The data for other genes design, and databases. The way forward for personal health care choices
is sparser or nonexistent, resulting in the daunting challenge of in the 21st century will require communicating what genomic medicine
interpreting variants in sequence data. As studies progress, a fraction has to offer; doing so in a compassionate and accurate way will be
of data will interpretable as clinically significant, moving from required of every health care provider.
the indeterminate category to mutations with evidence for clinical
action. Patience will be required as enough data are acquired to The complete reference list is available online at
examine the rest. ExpertConsult.com.

KEY REFERENCES
2. Malkin D, Garber JE, Strong LC, Friend SH. 13. Rahman N. Realizing the promise of cancer 23. Gabriel SB, Schaffner SF, Nguyen H, et al. The
CANCER. The cancer predisposition revolution. predisposition genes. Nature. 2014;505:302–308. structure of haplotype blocks in the human genome.
Science. 2016;352:1052–1053. 15. Scherer SW, Lee C, Birney E, et al. Challenges Science. 2002;296:2225–2229.
10. Manolio TA, Collins FS, Cox NJ, et al. Finding and standards in integrating surveys of structural 26. Scheet P, Stephens M. A fast and flexible statistical
the missing heritability of complex diseases. Nature. variation. Nat Genet. 2007;39:S7–S15. model for large-scale population genotype data:
2009;461:747–753. 16. Sebat J, Lakshmi B, Troge J, et al. Large-scale copy applications to inferring missing genotypes and hap-
11. Visscher PM, Wray NR, Zhang Q, et al. 10 Years of number polymorphism in the human genome. lotypic phase. Am J Hum Genet. 2006;78:629–644.
GWAS discovery: biology, function, and translation. Science. 2004;305:525–528. 31. Goldgar DE, Easton DF, Cannon-Albright LA,
Am J Hum Genet. 2017;101:5–22. 17. Kidd JM, Cooper GM, Donahue WF, et al. Mapping Skolnick MH. Systematic population-based assess-
12. Chanock S. Cancer biology: genome-wide associa- and sequencing of structural variation from eight ment of cancer risk in first-degree relatives of cancer
tion studies; 2014. World Cancer Report 2014, ed human genomes. Nature. 2008;453:56–64. probands. J Natl Cancer Inst. 1994;86:1600–1608.
Stewart BW WC (International Agency for Research 18. Orr N, Chanock S. Common genetic variation and 32. Lichtenstein P, Holm NV, Verkasalo PK, et al.
on Cancer): pp 193–202. human disease. Adv Genet. 2008;62:1–32. Environmental and heritable factors in the causation
336 Part I: Science and Clinical Oncology

of cancer—analyses of cohorts of twins from 72. Patterson N, Price AL, Reich D. Population structure and bladder cancer risk. Proc Natl Acad Sci USA.
Sweden, Denmark, and Finland. N Engl J Med. and eigenanalysis. PLoS Genet. 2006;2:e190. 2012;109:4974–4979.
2000;343:78–85. 80. Wang Z, Zhu B, Zhang M, et al. Imputation and 118. Machiela MJ, Zhou W, Sampson JN, et al.
36. Fletcher O, Houlston RS. Architecture of inherited subset-based association analysis across different Characterization of large structural genetic mosa-
susceptibility to common cancer. Nat Rev Cancer. cancer types identifies multiple independent risk icism in human autosomes. Am J Hum Genet.
2010;10:353–361. loci in the TERT-CLPTM1L region on chromosome 2015;96:487–497.
40. Rebbeck TR, Mitra N, Wan F, et al. Association of 5p15.33. Hum Mol Genet. 2014;23:6616–6633. 120. Dumanski JP, Rasi C, Lonn M, et al. Mutagenesis.
type and location of BRCA1 and BRCA2 muta- 87. Rebbeck TR, Devesa SS, Chang BL, et al. Global Smoking is associated with mosaic loss of chromo-
tions with risk of breast and ovarian cancer. JAMA. patterns of prostate cancer incidence, aggressiveness, some Y. Science. 2015;347:81–83.
2015;313:1347–1361. and mortality in men of African descent. Prostate 122. Wright DJ, Day FR, Kerrison ND, et al. Genetic
41. Kuchenbaecker KB, Hopper JL, Barnes DR, et al. Cancer. 2013;2013:560857. variants associated with mosaic Y chromosome loss
Risks of breast, ovarian, and contralateral breast 92. Haiman CA, Chen GK, Blot WJ, et al. Genome-wide highlight cell cycle genes and overlap with cancer
cancer for BRCA1 and BRCA2 mutation carriers. association study of prostate cancer in men of African susceptibility. Nat Genet. 2017;49:674–679.
JAMA. 2017;317:2402–2416. ancestry identifies a susceptibility locus at 17q21. 124. Genovese G, Kahler AK, Handsaker RE, et al.
43. Kuchenbaecker KB, McGuffog L, Barrowdale D, Nat Genet. 2011;43:570–573. Clonal hematopoiesis and blood-cancer risk
et al. Evaluation of polygenic risk scores for breast 97. Hoffmann TJ, Passarelli MN, Graff RE, et al. inferred from blood DNA sequence. N Engl J Med.
and ovarian cancer risk prediction in BRCA1 and Genome-wide association study of prostate-specific 2014;371:2477–2487.
BRCA2 mutation carriers. J Natl Cancer Inst. antigen levels identifies novel loci independent of 125. Chatterjee N, Shi J, Garcia-Closas M. Developing
2017;109. prostate cancer. Nat Commun. 2017;8:14248. and evaluating polygenic risk prediction models
48. Miki Y, Swensen J, Shattuck-Eidens D, et al. A 98. Maas P, Barrdahl M, Joshi AD, et al. Breast cancer for stratified disease prevention. Nat Rev Genet.
strong candidate for the breast and ovarian cancer risk from modifiable and non-modifiable risk factors 2016;17:392–406.
susceptibility gene BRCA1. Science. 1994;266:66–71. among white women in the Untied States. JAMA 127. Michailidou K, Beesley J, Lindstrom S, et al.
49. Wooster R, Bignell G, Lancaster J, et al. Identifica- Oncol. 2016;2:1295–1302. Genome-wide association analysis of more than
tion of the breast cancer susceptibility gene BRCA2. 101. Chatterjee N, Wheeler B, Sampson J, Hartge P, 120,000 individuals identifies 15 new susceptibility
Nature. 1995;378:789–792. Chanock SJ, Park J-H. Hidden heritability and risk loci for breast cancer. Nat Genet. 2015;47:373–
55. Risch N, Merikangas K. The future of genetic prediction based on genome-wide association studies. 380.
studies of complex human diseases. Science. Nat Genet. 2013;45:400–405. 133. Bolton KL, Chenevix-Trench G, Goh C, et al.
1996;273:1516–1517. 103. Mavaddat N, Pharoah PD, Michailidou K, et al. Association between BRCA1 and BRCA2 muta-
56. Hindorff LA, Sethupathy P, Junkins HA, et al. Prediction of breast cancer risk based on profiling tions and survival in women with invasive epithelial
Potential etiologic and functional implications of with common genetic variants. J Natl Cancer Inst. ovarian cancer. JAMA. 2012;307:382–390.
genome-wide association loci for human diseases 2015;107. 134. Domchek SM, Friebel TM, Singer CF, et al. Associa-
and traits. Proc Natl Acad Sci USA. 2009;106:9362– 104. Easton DF, Pharoah PD, Antoniou AC, et al. Gene- tion of risk-reducing surgery in BRCA1 or BRCA2
9367. panel sequencing and the prediction of breast-cancer mutation carriers with cancer risk and mortality.
61. Chanock SJ, Manolio T, Boehnke M, et al. Rep- risk. N Engl J Med. 2015;372:2243–2257. JAMA. 2010;304:967–975.
licating genotype-phenotype associations. Nature. 105. Southey MC, Goldgar DE, Winqvist R, et al. PALB2, 135. Zhang J, Walsh MF, Wu G, et al. Germline mutations
2007;447:655–660. CHEK2 and ATM rare variants and cancer risk: in predisposition genes in pediatric cancer. N Engl
62. Dickson SP, Wang K, Krantz I, Hakonarson H, data from COGS. J Med Genet. 2016;53:800–811. J Med. 2015;373:2336–2346.
Goldstein DB. Rare variants create synthetic genome- 107. Machiela MJ, Ho BM, Fisher VA, Hua X, Chanock 136. Mirabello L, Yeager M, Mai PL, et al. Germline
wide associations. PLoS Biol. 2010;8:e1000294. SJ. Limited evidence that cancer susceptibility regions TP53 variants and susceptibility to osteosarcoma.
63. Wray NR, Purcell SM, Visscher PM. Synthetic are preferential targets for somatic mutation. Genome J Natl Cancer Inst. 2015;107.
associations created by rare variants do not Biol. 2015;16:193. 138. Garraway LA, Lander ES. Lessons from the cancer
explain most GWAS results. PLoS Biol. 2011;9: 109. Birney E, Stamatoyannopoulos JA, Dutta A, et al. genome. Cell. 2013;153:17–37.
e1000579. Identification and analysis of functional elements in 140. Collins FS, Varmus H. A new initiative on precision
67. Freedman ML, Monteiro AN, Gayther SA, 1% of the human genome by the ENCODE pilot medicine. N Engl J Med. 2015;372:793–795.
et al. Principles for the post-GWAS functional project. Nature. 2007;447:799–816. 141. Rebbeck TR. Precision prevention of cancer.
characterization of cancer risk loci. Nat Genet. 111. Fu YP, Kohaar I, Rothman N, et al. Common genetic Cancer Epidemiol Biomarkers Prev. 2014;23:2713–
2011;43:513–518. variants in the PSCA gene influence gene expression 2715.
Discovery and Characterization of Cancer Genetic Susceptibility Alleles  •  CHAPTER 21 336.e1

REFERENCES
1. Li FP, Fraumeni JF Jr. Soft-tissue sarcomas, 25. Li Y, Willer C, Sanna S, Abecasis G. Genotype 46. Yokoyama S, Woods SL, Boyle GM, et al. A novel
breast cancer, and other neoplasms. A familial imputation. Annu Rev Genomics Hum Genet. recurrent mutation in MITF predisposes to familial
syndrome? Ann Intern Med. 1969;71:747–752. 2009;10:387–406. and sporadic melanoma. Nature. 2011;480:99–103.
2. Malkin D, Garber JE, Strong LC, Friend SH. 26. Scheet P, Stephens M. A fast and flexible statistical 47. Bertolotto C, Lesueur F, Giuliano S, et al. A
CANCER. The cancer predisposition revolution. model for large-scale population genotype data: SUMOylation-defective MITF germline mutation
Science. 2016;352:1052–1053. applications to inferring missing genotypes and hap- predisposes to melanoma and renal carcinoma.
3. Malkin D, Li FP, Strong LC, et al. Germ line lotypic phase. Am J Hum Genet. 2006;78:629–644. Nature. 2011;480:94–98.
p53 mutations in a familial syndrome of breast 27. Kong A, Frigge ML, Masson G, et al. Rate of de 48. Miki Y, Swensen J, Shattuck-Eidens D, et al. A
cancer, sarcomas, and other neoplasms. Science. novo mutations and the importance of father’s age strong candidate for the breast and ovarian cancer
1990;250:1233–1238. to disease risk. Nature. 2012;488:471–475. susceptibility gene BRCA1. Science. 1994;266:66–71.
4. Lander ES, Linton LM, Birren B, et al. Initial 28. Boettger LM, Handsaker RE, Zody MC, 49. Wooster R, Bignell G, Lancaster J, et al. Identifica-
sequencing and analysis of the human genome. McCarroll SA. Structural haplotypes and recent tion of the breast cancer susceptibility gene BRCA2.
Nature. 2001;409:860–921. evolution of the human 17q21.31 region. Nat Nature. 1995;378:789–792.
5. Venter JC, Adams MD, Myers EW, et  al. Genet. 2012;44:881–885. 50. Kadouri L, Hubert A, Rotenberg Y, et al. Cancer
The sequence of the human genome. Science. 29. Diskin SJ, Hou C, Glessner JT, et al. Copy number risks in carriers of the BRCA1/2 Ashkenazi founder
2001;291:1304–1351. variation at 1q21.1 associated with neuroblastoma. mutations. J Med Genet. 2007;44:467–471.
6. Frazer KA, Ballinger DG, Cox DR, et al. A second Nature. 2009;459:987–991. 51. Tonin P, Weber B, Offit K, et al. Frequency of
generation human haplotype map of over 3.1 million 30. Li W, Xia Y, Wang C, et al. Identifying human recurrent BRCA1 and BRCA2 mutations in
SNPs. Nature. 2007;449:851–861. genome-wide CNV, LOH and UPD by targeted Ashkenazi Jewish breast cancer families. Nat Med.
7. International HapMap C, Altshuler DM, Gibbs RA, sequencing of selected regions. PLoS ONE. 1996;2:1179–1183.
et al. Integrating common and rare genetic variation 2014;10:e0123081. 52. Struewing JP, Abeliovich D, Peretz T, et al. The
in diverse human populations. Nature. 2010;467: 31. Goldgar DE, Easton DF, Cannon-Albright LA, carrier frequency of the BRCA1 185delAG mutation
52–58. Skolnick MH. Systematic population-based assess- is approximately 1 percent in Ashkenazi Jewish
8. Grossman RL, Heath AP, Ferretti V, et al. Toward ment of cancer risk in first-degree relatives of cancer individuals. Nat Genet. 1995;11:198–200.
a shared vision for cancer genomic data. N Engl J probands. J Natl Cancer Inst. 1994;86:1600–1608. 53. Spurdle AB, Couch FJ, Parsons MT, et al. Refined
Med. 2016;375:1109–1112. 32. Lichtenstein P, Holm NV, Verkasalo PK, et al. histopathological predictors of BRCA1 and BRCA2
9. Clinical Cancer Genome Task Team of the Global Environmental and heritable factors in the causa- mutation status: a large-scale analysis of breast
Alliance for Genomics and Health, Lawler M, tion of cancer—analyses of cohorts of twins from cancer characteristics from the BCAC, CIMBA,
et al. Sharing clinical and genomic data on Sweden, Denmark, and Finland. N Engl J Med. and ENIGMA consortia. Breast Cancer Res.
cancer—the need for global solutions. N Engl J 2000;343:78–85. 2014;16:3419.
Med. 2017;376:2006–2009. 33. Spitz MR, Currier RD, Fueger JJ, Babaian RJ, 54. Moisio AL, Sistonen P, Weissenbach J, de la Chapelle
10. Manolio TA, Collins FS, Cox NJ, et al. Finding Newell GR. Familial patterns of prostate cancer: a A, Peltomaki P. Age and origin of two common
the missing heritability of complex diseases. Nature. case-control analysis. J Urol. 1991;146:1305–1307. MLH1 mutations predisposing to hereditary colon
2009;461:747–753. 34. Steinberg GD, Carter BS, Beaty TH, Childs B, cancer. Am J Hum Genet. 1996;59:1243–1251.
11. Visscher PM, Wray NR, Zhang Q, et al. 10 Years of Walsh PC. Family history and the risk of prostate 55. Risch N, Merikangas K. The future of genetic
GWAS discovery: biology, function, and translation. cancer. Prostate. 1990;17:337–347. studies of complex human diseases. Science.
Am J Hum Genet. 2017;101:5–22. 35. Hayes RB, Liff JM, Pottern LM, et al. Prostate cancer 1996;273:1516–1517.
12. Chanock S. Cancer biology: genome-wide associa- risk in US blacks and whites with a family history 56. Hindorff LA, Sethupathy P, Junkins HA, et al.
tion studies; 2014. World Cancer Report 2014, ed of cancer. Int J Cancer. 1995;60:361–364. Potential etiologic and functional implications of
Stewart BW WC (International Agency for Research 36. Fletcher O, Houlston RS. Architecture of inherited genome-wide association loci for human diseases and
on Cancer). pp 193-202. susceptibility to common cancer. Nat Rev Cancer. traits. Proc Natl Acad Sci USA. 2009;106:9362–9367.
13. Rahman N. Realizing the promise of cancer 2010;10:353–361. 57. Garcia-Closas M, Malats N, Silverman D, et al.
predisposition genes. Nature. 2014;505:302–308. 37. Couch FJ, Farid LM, DeShano ML, et al. BRCA2 NAT2 slow acetylation, GSTM1 null genotype,
14. Knudson AG Jr. Mutation and cancer: statistical germline mutations in male breast cancer cases and and risk of bladder cancer: results from the Spanish
study of retinoblastoma. Proc Natl Acad Sci USA. breast cancer families. Nat Genet. 1996;13:123– Bladder Cancer Study and meta-analyses. Lancet.
1971;68:820–823. 125. 2005;366:649–659.
15. Scherer SW, Lee C, Birney E, et al. Challenges 38. Bodmer WF, Bailey CJ, Bodmer J, et al. Localization 58. Garcia-Closas M, Hein DW, Silverman D, et al. A
and standards in integrating surveys of structural of the gene for familial adenomatous polyposis on single nucleotide polymorphism tags variation in
variation. Nat Genet. 2007;39:S7–S15. chromosome 5. Nature. 1987;328:614–616. the arylamine N-acetyltransferase 2 phenotype in
16. Sebat J, Lakshmi B, Troge J, et al. Large-scale copy 39. Hunter DJ, Chanock SJ. Genome-wide association populations of European background. Pharmacogenet
number polymorphism in the human genome. studies and “the art of the soluble.” J Natl Cancer Genomics. 2011;21:231–236.
Science. 2004;305:525–528. Inst. 2010;102:836–837. 59. Hashibe M, McKay JD, Curado MP, et al. Multiple
17. Kidd JM, Cooper GM, Donahue WF, et al. Mapping 40. Rebbeck TR, Mitra N, Wan F, et al. Association of ADH genes are associated with upper aerodigestive
and sequencing of structural variation from eight type and location of BRCA1 and BRCA2 muta- cancers. Nat Genet. 2008;40:707–709.
human genomes. Nature. 2008;453:56–64. tions with risk of breast and ovarian cancer. JAMA. 60. A map of human genome variation from population-
18. Orr N, Chanock S. Common genetic variation and 2015;313:1347–1361. scale sequencing. Nature. 2010;467:1061–1073.
human disease. Adv Genet. 2008;62:1–32. 41. Kuchenbaecker KB, Hopper JL, Barnes DR, et al. 61. Chanock SJ, Manolio T, Boehnke M, et al. Rep-
19. Donnelly P. Progress and challenges in genome- Risks of breast, ovarian, and contralateral breast licating genotype-phenotype associations. Nature.
wide association studies in humans. Nature. cancer for BRCA1 and BRCA2 mutation carriers. 2007;447:655–660.
2008;456:728–731. JAMA. 2017;317:2402–2416. 62. Dickson SP, Wang K, Krantz I, Hakonarson H,
20. 1000 Genomes Project Consortium, Abecasis 42. Antoniou AC, Spurdle AB, Sinilnikova OM, Goldstein DB. Rare variants create synthetic genome-
GR, Altshuler D, et al. A map of human genome et al. Common breast cancer-predisposition alleles wide associations. PLoS Biol. 2010;8:e1000294.
variation from population-scale sequencing. Nature. are associated with breast cancer risk in BRCA1 63. Wray NR, Purcell SM, Visscher PM. Synthetic
2010;467:1061–1073. and BRCA2 mutation carriers. Am J Hum Genet. associations created by rare variants do not explain
21. Genome of the Netherlands Consortium. Whole- 2008;82:937–948. most GWAS results. PLoS Biol. 2011;9:e1000579.
genome sequence variation, population structure 43. Kuchenbaecker KB, McGuffog L, Barrowdale D, 64. Haiman CA, Patterson N, Freedman ML, et al.
and demographic history of the Dutch population. et al. Evaluation of polygenic risk scores for breast Multiple regions within 8q24 independently affect
Nat Genet. 2014;46:818–825. and ovarian cancer risk prediction in BRCA1 and risk for prostate cancer. Nat Genet. 2007;39:638–
22. Forbes SA, Beare D, Boutselakis H, et al. COSMIC: BRCA2 mutation carriers. J Natl Cancer Inst. 644.
somatic cancer genetics at high-resolution. Nucleic 2017;109. 65. Amundadottir LT, Sulem P, Gudmundsson J, et al.
Acids Res. 2017;45:D777–D783. 44. Kamb A, Shattuck-Eidens D, Eeles R, et al. Analysis A common variant associated with prostate cancer
23. Gabriel SB, Schaffner SF, Nguyen H, et al. The of the p16 gene (CDKN2) as a candidate for the in European and African populations. Nat Genet.
structure of haplotype blocks in the human genome. chromosome 9p melanoma susceptibility locus. Nat 2006;38:652–658.
Science. 2002;296:2225–2229. Genet. 1994;8:23–26. 66. Freedman ML, Haiman CA, Patterson N, et al.
24. Hinch AG, Tandon A, Patterson N, et al. The land- 45. Yang XR, Ng D, Alcorta DA, et al. T (brachyury) Admixture mapping identifies 8q24 as a prostate
scape of recombination in African Americans. Nature. gene duplication confers major susceptibility to cancer risk locus in African-American men. Proc
2011;476:170–175. familial chordoma. Nat Genet. 2009;41:1176–1178. Natl Acad Sci USA. 2006;103:14068–14073.
336.e2 Part I: Science and Clinical Oncology

67. Freedman ML, Monteiro AN, Gayther SA, and mortality in men of African descent. Prostate women with breast cancer. Hered Cancer Clin Pract.
et al. Principles for the post-GWAS functional Cancer. 2013;2013:560857. 2016;14:9.
characterization of cancer risk loci. Nat Genet. 88. Johanneson B, Deutsch K, McIntosh L, et al. Sugges- 107. Machiela MJ, Ho BM, Fisher VA, Hua X, Chanock
2011;43:513–518. tive genetic linkage to chromosome 11p11.2-q12.2 SJ. Limited evidence that cancer susceptibility regions
68. Wacholder S. Practical considerations in choosing in hereditary prostate cancer families with primary are preferential targets for somatic mutation. Genome
between the case-cohort and nested case-control kidney cancer. Prostate. 2007;67:732–742. Biol. 2015;16:193.
designs. Epidemiology. 1991;2:155–158. 89. Schodel J, Bardella C, Sciesielski LK, et al. Common 108. Machiela MJ, Chanock SJ. LDlink: a web-based
69. Wacholder S, Silverman DT, McLaughlin JK, genetic variants at the 11q13.3 renal cancer application for exploring population-specific
Mandel JS. Selection of controls in case-control susceptibility locus influence binding of HIF to haplotype structure and linking correlated alleles
studies. III. Design options. Am J Epidemiol. an enhancer of cyclin D1 expression. Nat Genet. of possible functional variants. Bioinformatics.
1992;135:1042–1050. 2012;44:420–425, S421–422. 2015;31:3555–3557.
70. Wacholder S, Silverman DT, McLaughlin JK, 90. French JD, Ghoussaini M, Edwards SL, et al. 109. Birney E, Stamatoyannopoulos JA, Dutta A, et al.
Mandel JS. Selection of controls in case-control Functional variants at the 11q13 risk locus Identification and analysis of functional elements in
studies. II. Types of controls. Am J Epidemiol. for breast cancer regulate cyclin D1 expression 1% of the human genome by the ENCODE pilot
1992;135:1029–1041. through long-range enhancers. Am J Hum Genet. project. Nature. 2007;447:799–816.
71. Wacholder S, McLaughlin JK, Silverman DT, Mandel 2013;92:489–503. 110. Boyle AP, Hong EL, Hariharan M, et al. Annota-
JS. Selection of controls in case-control studies. I. 91. Hunter DJ, Thomas G, Hoover RN, Chanock SJ. tion of functional variation in personal genomes
Principles. Am J Epidemiol. 1992;135:1019–1028. Scanning the horizon: what is the future of genome- using RegulomeDB. Genome Res. 2012;22:1790–
72. Patterson N, Price AL, Reich D. Population structure wide association studies in accelerating discoveries 1797.
and eigenanalysis. PLoS Genet. 2006;2:e190. in cancer etiology and prevention? Cancer Causes 111. Fu YP, Kohaar I, Rothman N, et al. Common genetic
73. Postel-Vinay S, Veron AS, Tirode F, et al. Common Control. 2007;18:479–484. variants in the PSCA gene influence gene expression
variants near TARDBP and EGR2 are associated 92. Haiman CA, Chen GK, Blot WJ, et al. Genome-wide and bladder cancer risk. Proc Natl Acad Sci USA.
with susceptibility to Ewing sarcoma. Nat Genet. association study of prostate cancer in men of African 2012;109:4974–4979.
2012;44:323–327. ancestry identifies a susceptibility locus at 17q21. 112. Garcia-Closas M, Rothman N, Figueroa JD, et al.
74. Rapley EA, Turnbull C, Al Olama AA, et al. A Nat Genet. 2011;43:570–573. Common genetic polymorphisms modify the effect
genome-wide association study of testicular germ 93. Eeles RA, Kote-Jarai Z, Giles GG, et al. Multiple of smoking on absolute risk of bladder cancer. Cancer
cell tumor. Nat Genet. 2009;41:807–810. newly identified loci associated with prostate cancer Res. 2013;73:2211–2220.
75. Kanetsky PA, Mitra N, Vardhanabhuti S, et al. susceptibility. Nat Genet. 2008;40:316–321. 113. McKay JD, Hung RJ, Han Y, et al. Large-scale
Common variation in KITLG and at 5q31.3 94. Ahn J, Berndt SI, Wacholder S, et al. Variation in association analysis identifies new lung cancer
predisposes to testicular germ cell cancer. Nat Genet. KLK genes, prostate-specific antigen and risk of susceptibility loci and heterogeneity in genetic
2009;41:811–815. prostate cancer. Nat Genet. 2008;40:1032–1034, susceptibility across histological subtypes. Nat Genet.
76. Haiman CA, Le Marchand L, Yamamato J, et al. author reply 1035–1036. 2017;49:1126–1132.
A common genetic risk factor for colorectal and 95. Kote-Jarai Z, Amin Al Olama A, Leongamornlert 114. Lan Q, Hsiung CA, Matsuo K, et al. Genome-wide
prostate cancer. Nat Genet. 2007;39:954–956. D, et al. Identification of a novel prostate cancer association analysis identifies new lung cancer
77. Sur IK, Hallikas O, Vaharautio A, et al. Mice susceptibility variant in the KLK3 gene transcript. susceptibility loci in never-smoking women in Asia.
lacking a Myc enhancer that includes human SNP Hum Genet. 2011;129:687–694. Nat Genet. 2012;44:1330–1335.
rs6983267 are resistant to intestinal tumors. Science. 96. Parikh H, Wang Z, Pettigrew KA, et al. Fine mapping 115. Prokunina-Olsson L, Muchmore B, Tang W, et al.
2012;338:1360–1363. the KLK3 locus on chromosome 19q13.33 associated A variant upstream of IFNL3 (IL28B) creating
78. Pomerantz MM, Ahmadiyeh N, Jia L, et al. The with prostate cancer susceptibility and PSA levels. a new interferon gene IFNL4 is associated with
8q24 cancer risk variant rs6983267 shows long- Hum Genet. 2011;129:675–685. impaired clearance of hepatitis C virus. Nat Genet.
range interaction with MYC in colorectal cancer. 97. Hoffmann TJ, Passarelli MN, Graff RE, et al. 2013;45:164–171.
Nat Genet. 2009;41:882–884. Genome-wide association study of prostate-specific 116. Rodriguez-Santiago B, Malats N, Rothman N, et al.
79. Tuupanen S, Turunen M, Lehtonen R, et al. antigen levels identifies novel loci independent of Mosaic uniparental disomies and aneuploidies as
The common colorectal cancer predisposition prostate cancer. Nat Commun. 2017;8:14248. large structural variants of the human genome.
SNP rs6983267 at chromosome 8q24 confers 98. Maas P, Barrdahl M, Joshi AD, et al. Breast cancer Am J Hum Genet. 2010;87:129–138.
potential to enhanced Wnt signaling. Nat Genet. risk from modifiable and non-modifiable risk factors 117. Machiela MJ, Zhou W, Caporaso N, et al. Mosaic
2009;41:885–890. among white women in the Untied States. JAMA 13q14 deletions in peripheral leukocytes of non-
80. Wang Z, Zhu B, Zhang M, et al. Imputation and Oncol. 2016;2:1295–1302. hematologic cancer cases and healthy controls.
subset-based association analysis across different 99. Park JH, Wacholder S, Gail MH, et al. Estimation of J Hum Genet. 2016;61:411–418.
cancer types identifies multiple independent risk effect size distribution from genome-wide association 118. Machiela MJ, Zhou W, Sampson JN, et al.
loci in the TERT-CLPTM1L region on chromosome studies and implications for future discoveries. Nat Characterization of large structural genetic mosa-
5p15.33. Hum Mol Genet. 2014;23:6616–6633. Genet. 2010;42:570–575. icism in human autosomes. Am J Hum Genet.
81. Kote-Jarai Z, Saunders EJ, Leongamornlert DA, et al. 100. Park JH, Gail MH, Weinberg CR, et al. Dis- 2015;96:487–497.
Fine-mapping identifies multiple prostate cancer risk tribution of allele frequencies and effect sizes 119. Machiela MJ, Zhou W, Karlins E, et al. Female
loci at 5p15, one of which associates with TERT and their interrelationships for common genetic chromosome X mosaicism is age-related and
expression. Hum Mol Genet. 2013;22:2520–2528. susceptibility variants. Proc Natl Acad Sci USA. preferentially affects the inactivated X chromosome.
82. Calado RT, Regal JA, Hills M, et al. Constitutional 2011;108:18026–18031. Nat Commun. 2016;7:11843.
hypomorphic telomerase mutations in patients with 101. Chatterjee N, Wheeler B, Sampson J, Hartge P, 120. Dumanski JP, Rasi C, Lonn M, et al. Mutagenesis.
acute myeloid leukemia. Proc Natl Acad Sci USA. Chanock SJ, Park J-H. Hidden heritability and risk Smoking is associated with mosaic loss of chromo-
2009;106:1187–1192. prediction based on genome-wide association studies. some Y. Science. 2015;347:81–83.
83. Savage SA, Stewart BJ, Weksler BB, et al. Mutations Nat Genet. 2013;45:400–405. 121. Zhou W, Machiela MJ, Freedman ND, et al. Mosaic
in the reverse transcriptase component of telomerase 102. Eeles RA, Olama AA, Benlloch S, et al. Identification loss of chromosome Y is associated with common
(TERT) in patients with bone marrow failure. Blood of 23 new prostate cancer susceptibility loci using variation near TCL1A. Nat Genet. 2016;48:563–568.
Cells Mol Dis. 2006;37:134–136. the iCOGS custom genotyping array. Nat Genet. 122. Wright DJ, Day FR, Kerrison ND, et al. Genetic
84. Stacey SN, Gudbjartsson DF, Sulem P, et al. 2013;45:385–391, 391e381–382. variants associated with mosaic Y chromosome loss
Common variants on 1p36 and 1q42 are associ- 103. Mavaddat N, Pharoah PD, Michailidou K, et al. highlight cell cycle genes and overlap with cancer
ated with cutaneous basal cell carcinoma but not Prediction of breast cancer risk based on profiling susceptibility. Nat Genet. 2017;49:674–679.
with melanoma or pigmentation traits. Nat Genet. with common genetic variants. J Natl Cancer Inst. 123. Jager N, Schlesner M, Jones DT, et al. Hypermuta-
2008;40:1313–1318. 2015;107. tion of the inactive X chromosome is a frequent
85. Wang Z. Improved imputation of common and 104. Easton DF, Pharoah PD, Antoniou AC, et al. Gene- event in cancer. Cell. 2013;155:567–581.
uncommon SNPs with a new reference set. Nat panel sequencing and the prediction of breast-cancer 124. Genovese G, Kahler AK, Handsaker RE, et al.
Genet. 2012;44:6–7. risk. N Engl J Med. 2015;372:2243–2257. Clonal hematopoiesis and blood-cancer risk
86. Gudmundsson J, Sulem P, Gudbjartsson DF, et al. 105. Southey MC, Goldgar DE, Winqvist R, et al. PALB2, inferred from blood DNA sequence. N Engl J Med.
A study based on whole-genome sequencing yields a CHEK2 and ATM rare variants and cancer risk: 2014;371:2477–2487.
rare variant at 8q24 associated with prostate cancer. data from COGS. J Med Genet. 2016;53:800– 125. Chatterjee N, Shi J, Garcia-Closas M. Developing
Nat Genet. 2012;44:1326–1329. 811. and evaluating polygenic risk prediction models
87. Rebbeck TR, Devesa SS, Chang BL, et al. Global 106. Southey MC, Winship I, Nguyen-Dumont T. for stratified disease prevention. Nat Rev Genet.
patterns of prostate cancer incidence, aggressiveness, PALB2: research reaching to clinical outcomes for 2016;17:392–406.
Discovery and Characterization of Cancer Genetic Susceptibility Alleles  •  CHAPTER 21 336.e3

126. Morgan TH. Random segregation versus coupling 132. Morton LM, Sampson JN, Armstrong GT, et al. 136a.  National Society of Genetic Counselors’ Definition
in Mendelian inheritance. Science. 1911;34:384. Genome-wide association study identifies susceptibil- Task Force, Resta R, Biesecker BB, et al. A new
127. Michailidou K, Beesley J, Lindstrom S, et al. ity loci that modify radiation-related risk for breast definition of Genetic Counseling: National Society
Genome-wide association analysis of more than cancer after childhood cancer. J Natl Cancer Inst. of Genetic Counselors’ Task Force report. J Genet
120,000 individuals identifies 15 new susceptibility 2017. Couns. 2006;15(2):77–83.
loci for breast cancer. Nat Genet. 2015;47:373–380. 133. Bolton KL, Chenevix-Trench G, Goh C, et al. 137. Collins FS, McKusick VA. Implications of the
128. Helfand BT, Roehl KA, Cooper PR, et al. Associa- Association between BRCA1 and BRCA2 muta- human genome project for medical science. JAMA.
tions of prostate cancer risk variants with disease tions and survival in women with invasive epithelial 2001;285:540–544.
aggressiveness: results of the NCI-SPORE Genetics ovarian cancer. JAMA. 2012;307:382–390. 138. Garraway LA, Lander ES. Lessons from the cancer
Working Group analysis of 18,343 cases. Hum Genet. 134. Domchek SM, Friebel TM, Singer CF, et al. Associa- genome. Cell. 2013;153:17–37.
2015;134:439–450. tion of risk-reducing surgery in BRCA1 or BRCA2 139. Iyer G, Hanrahan AJ, Milowsky MI, et al. Genome
129. Berndt SI, Wang Z, Yeager M, et al. Two susceptibil- mutation carriers with cancer risk and mortality. sequencing identifies a basis for everolimus sensitivity.
ity loci identified for prostate cancer aggressiveness. JAMA. 2010;304:967–975. Science. 2012;338:221.
Nat Commun. 2015;6:6889. 135. Zhang J, Walsh MF, Wu G, et al. Germline mutations 140. Collins FS, Varmus H. A new initiative on precision
130. Maris JM, Mosse YP, Bradfield JP, et al. Chromo- in predisposition genes in pediatric cancer. N Engl medicine. N Engl J Med. 2015;372:793–795.
some 6p22 locus associated with clinically aggressive J Med. 2015;373:2336–2346. 141. Rebbeck TR. Precision prevention of cancer. Cancer
neuroblastoma. N Engl J Med. 2008;358:2585–2593. 136. Mirabello L, Yeager M, Mai PL, et al. Germline Epidemiol Biomarkers Prev. 2014;23:2713–2715.
131. Trevino LR, Yang W, French D, et al. Germline TP53 variants and susceptibility to osteosarcoma.
genomic variants associated with childhood acute lym- J Natl Cancer Inst. 2015;107.
phoblastic leukemia. Nat Genet. 2009;41:1001–1005.
Lifestyle and Cancer Prevention 22 
Karen Basen-Engquist, Powel Brown, Adriana M. Coletta,
Michelle Savage, Karen Colbert Maresso, and Ernest Hawk

S UMMARY OF K EY P OI NT S
• Approximately one-third to • Cancer interception is a type of (squamous cell); (4) colorectal
one-half of cancers can be chemoprevention or molecular (in men only); (5) breast; and
prevented through adoption of prevention that seeks to address (6) liver.
healthy lifestyles and avoidance of those at high risk because of • Both subjective and objective
known risk factors. established precancers. evidence links regular, structured
• The findings of several large • Tobacco use accounts for 30% of all physical activity with reduced risk of
observational studies from the cancer deaths; it is on the rise in chronic disease and certain cancers,
United States and Europe developing countries and remains such as colon and breast cancer.
strongly suggest that following high among several subpopulations • The link between diet and cancer
current cancer prevention within the United States. risk is complex; not all dietary
recommendations from either the • In 2015, electronic nicotine delivery components are linked with cancer
American Cancer Society (ACS) or systems, specifically e-cigarettes, risk, and some components exhibit
the American Institute for Cancer were the most commonly used strong links with cancer risk
Research (AICR) leads to significant tobacco product among both individually whereas others exhibit
health benefits. middle- and high-school students. stronger effects synergistically.
• Advanced cancers are now • Obesity is now linked to 13 different • It is estimated that indoor tanning
understood to be extremely cancers, and approximately 17% of results in 400,000 cases of skin
genetically heterogeneous on children and 36% of adults are cancer in the United States each
numerous levels, so intervening at an classified as obese in the United year, and 6000 of these are
earlier time point when there are States. melanoma.
fewer derangements to contend with, • Alcohol has been shown to • There are 13 agents approved for
at the precancerous lesion stage, convincingly increase the risk of at the treatment of precancerous
offers a rational approach to cancer least six cancers: (1) oral and lesions and cancer risk reduction or
prevention. pharynx; (2) larynx; (3) esophageal prevention.

Cancer prevention is the most cost-effective, long-term strategy for have been theoretically avoided.3 This estimate as to the “preventability”
the control of cancer.1,2 It is now well established that approximately of cancer was based on their approximations for the relative contribution
one-third to one-half of all adult cancers occurring in Western popula- of individual cancer risk factors—30% for tobacco, 2% to 4% for
tions can be prevented through the adoption of “healthy lifestyles.” alcohol, 35% for diet, 1% to 10% for infectious agents, 2% for
A comprehensive view of a healthy lifestyle includes adherence to pollution, and 4% for occupational exposures.3 Subsequent epide-
published cancer prevention recommendations that seek to eliminate or miologic data have largely supported these estimates and have identified
minimize exposure to lifestyle risk factors, and the use of screening and obesity and lack of physical activity as additional cancer risk factors,
early detection tests as well as molecular preventive agents (e.g., aspirin, causing a slight redistribution of the percent of cancer mortality
tamoxifen, human papillomavirus [HPV] vaccine) when warranted. This attributable to the various lifestyle risk factors. A recent review sum-
chapter focuses on the epidemiology of lifestyle risk factors and their marizing updated data on this topic found that obesity accounts for
associated interventions, as well as on the use of molecular preventive 15% to 20% of cancers, physical inactivity accounts for approximately
agents. Screening and early detection are addressed in another chapter. 5%, and the estimate for diet has decreased to 5%, much less than
the initial estimate of 35% by Doll and Peto.4 Although there are
RATIONALE FOR PREVENTION limitations to estimating the relative contributions of individual risk
factors to cancer mortality, and although the estimate of the amount
The rationale for an enhanced focus on prevention as the dominant of cancer that can be prevented may be a best case scenario difficult
strategy by which to reduce the cancer burden is supported by at least to achieve in reality, such studies not only demonstrate a need to
three lines of evidence. better understand the interplay among cancer-associated lifestyle factors,
First, the preventability of cancer was estimated by Doll and Peto but also highlight the potential impact of prevention and underscore
in their 1981 landmark publication on the relative contributions of its urgency.
the avoidable causes of cancer.3 They concluded that three-fourths or Second, several large observational studies from the United States
more of all cancers occurring in the United States during 1970 could and Europe strongly suggest that following current cancer prevention

337
338 Part I: Science and Clinical Oncology

Table 22.1  Current Recommendations for Cancer Prevention From the American Cancer Society (ACS)
and the American Institute for Cancer Research (AICR)
ACS GUIDELINES ON NUTRITION AND PHYSICAL ACTIVITY64a AICR CANCER PREVENTION RECOMMENDATIONS63
Achieve and maintain a healthy weight throughout life. Be as lean as possible without becoming underweight.
• Be as lean as possible throughout life without being underweight. Be physically active for at least 30 minutes every day. Limit
• Avoid excess weight gain at all ages. sedentary habits.
• Get regular physical activity and limit intake of high-calorie food Avoid sugary drinks. Limit consumption of energy-dense foods.
and drinks as keys to help maintain a healthy weight. Eat more of a variety of vegetables, fruits, whole grains, and
Be physically active. legumes such as beans.
• Get at least 150 minutes of moderate intensity or 75 minutes of Limit consumption of red meats (such as beef, pork and lamb)
vigorous intensity activity each week (or a combination of these), and avoid processed meats.
preferably spread throughout the week. If consumed at all, limit alcoholic drinks to 2 per day for men
• Limit sedentary behavior such as sitting, lying down, watching TV, and 1 per day for women.
and other forms of screen-based entertainment. Limit consumption of salty foods and foods processed with salts.
• Doing some physical activity, above usual activities, no matter what Don’t use supplements to protect against cancer.
one’s activity level, can have many health benefits. It is best for mothers to breastfeed exclusively for up to 6
Eat a healthy diet, with an emphasis on plant foods. months and then add other liquids and foods.
• Choose foods and drinks in amounts that help you get to and After treatment, cancer survivors should follow the
maintain a healthy weight. recommendations for cancer prevention.
• Limit how much processed meat and red meat you eat. Do not smoke or chew tobacco.
• Eat at least 2.5 cups of fruits and vegetables each day.
• Choose whole grains instead of refined grain products.
If you drink alcohol, limit your intake.
• Drink no more than 1 drink per day for women or 2 per day for men.

a
Recommendations are in addition to avoiding tobacco, being safe in the sun, and following cancer screening guidelines.

recommendations (Table 22.1) from either the American Cancer Society reverse migration—offers a potential pathway to making this strategy
(ACS) or the American Institute for Cancer Research (AICR) leads a reality. However, the field is currently challenged by a dearth of data
to significant health benefits.5 Significant reductions in both cancer and understanding around the precancerous genome. Gaining a better
risk and cancer mortality, as well as in cardiovascular mortality and understanding of the type, timing, and sequence of molecular aber-
overall mortality, were seen across studies for those individuals who rations underlying precancerous lesions and their development will
adhered to a majority of the recommendations compared with those help drive this strategy forward. Establishing a Precancerous Genome
who adhered to fewer recommendations. Atlas, analogous to The Cancer Genome Atlas (TCGA), is a research
Third, cancer is the last step in a multistep process occurring over priority in the field of cancer prevention.9
decades. This offers time and opportunity to intervene. Carcinogenesis
is characterized by a progression of genetic changes affecting cellular PREVENTION THROUGH LIFESTYLE
identity and growth that culminates in cancer after many years. The
accumulation of these changes at the DNA level is reflected in INTERVENTIONS
cytomorphologic and histopathologic changes termed preinvasive Tobacco
neoplastic lesions, or simply precancerous lesions. Because advanced
cancers are now understood to be extremely genetically heterogeneous Tobacco remains the leading preventable cause of death and disability
on numerous levels—within a primary tumor, between two metastases, in the United States. It has been linked to at least 12 different cancers
within metastatic lesions, and between patients—and because this and accounts for approximately one-third of all cancer deaths, more
heterogeneity negatively affects the response to treatment,6 intervening than any other risk factor. In those who already have cancer, it leads
at an earlier time point when there are fewer derangements to contend to adverse health outcomes, including increasing the risks of second
with, at the precancerous lesion stage, offers a rational approach to primary cancers, cancer-related mortality, and all-cause mortality.10
cancer prevention. Tobacco-using cancer patients may also have an increased risk of
This approach to treating precancerous lesions in order to prevent recurrence, a poorer response to treatment, and increased treatment-
progression to full-blown cancer is being termed cancer interception related toxicity.10 Evidence shows that quitting smoking improves the
and is rapidly gaining traction as a priority strategy to address the prognosis of cancer patients.
cancer burden, in part because of recent advances in understanding The prevalence of cigarette smoking among adults in 2015 was
preinvasive cancer biology. According to Blackburn, cancer interception 15%, a decline from 21% in 2005.11 Yet this prevalence equates to
is “the active way of combatting cancer and carcinogenesis at earlier nearly 37 million adults who still smoke, and disparities in prevalence
and earlier stages.”7 It is “active” in the sense that it involves more persist. Groups with a prevalence higher than the general population
than risk avoidance, such as avoidance of smoking or sun exposure, include males (16.7%); those aged 25 to 44 years (17.7%); American
which is often the domain of primary prevention. Cancer interception Indians/Alaska Natives (21.9%); those with only a General Educational
is a type of chemoprevention or molecular prevention that seeks to Development (GED) credential (34.1%); those living below the
address those at high risk because of established precancers.7,8 A timely poverty level (26.1%); residents of the Midwest (18.7%); those who
example of interception in cancer prevention is the use of aspirin to are uninsured (27.4%) or receiving Medicaid (27.8%); lesbian, gay,
reduce the number of colorectal adenomas (i.e., precancers), which or bisexual individuals (20.6%); and those with serious psychologic
has resulted in significant reductions in colorectal cancer (CRC) distress (40.6%).11
incidence and mortality in randomized trials of use for cardiovascular In addition to these disparities, the growing use of e-cigarettes
event prophylaxis.7 The application of drugs that have been shown among youth is a real concern because it presents a threat to the
to be effective in advanced cancer to precancerous lesions—that is, last half-century of progress in tobacco control. Although traditional
Lifestyle and Cancer Prevention  •  CHAPTER 22 339

cigarette use declined among youth in the 2011–2015 period, there


were substantial increases in e-cigarette use among students during this Table 22.2  The World Health Organization’s
same time period.12 In 2015, e-cigarettes were the most commonly used Framework Convention on Tobacco
tobacco product among both middle- and high-school students, with Control MPOWER Measures
5% and 16% reporting current use, respectively.12 Limited research to
date suggests that e-cigarette users are more likely to take up traditional Monitor tobacco use and prevention policies
tobacco products than are never-users of e-cigarettes.13 However, Protect people from tobacco smoke
regardless of whether e-cigarettes serve as a gateway to traditional Offer help to quit tobacco use
tobacco use, they nevertheless expose youth to nicotine, opening the Warn about the dangers of tobacco
possibility for addiction and altered brain development12 and may pose
unique health threats of their own through their fluid and aerosol Enforce bans on tobacco advertising, promotion, and sponsorship
composition. E-cigarette fluid is known to contain propylene glycol Raise taxes on tobacco
(PG), a US Food and Drug Administration (FDA)–approved food
additive. But the effects of PG when heated and inhaled are unclear,
and e-cigarette aerosol has also been shown to contain numerous
carcinogens, including tobacco-specific nitrosamines, volatile organic women smoke. It is feared that the large number of nonsmoking
compounds, and heavy metals.14 In 2016 the FDA finalized a rule that women in these countries represents a target for tobacco-company
extended its authority to the manufacturing, marketing, and distribution marketing efforts.17 Global tobacco control is tremendously challenging,
of e-cigarettes. Before this, e-cigarettes were completely unregulated, given the varying patterns of use within and between countries and
making it difficult to know what was in them, to study or generate the often less-than-optimal resources available in LMICs. Nevertheless,
data to inform regulations, and to prevent them from being sold to it is essential to reduce the tremendous burden of premature death
minors. With the passing of the new rule, e-cigarette manufacturers, and disability that many of these countries can expect if current trends
retailers, and consumers must now adhere to a set of regulations in tobacco use continue. The World Health Organization (WHO)
meant to protect the health of all Americans. This includes a ban on Framework Convention Alliance for Tobacco Control (FCTC) is a
the sale of e-cigarettes to those younger than age 18. It is hoped that legally binding treaty that requires its parties to implement particular
these regulations will improve the ability of the scientific community evidence-based tobacco control measures. It entered into force in 2005
to more easily study e-cigarettes and their potential health effects. and is meant to assist LMICs in making tobacco control a reality in
Comprehensive tobacco control as established by the Centers for their countries. It provides both supply- and demand-reduction provi-
Disease Control and Prevention (CDC) includes a mix of educational, sions. The WHO’s MPOWER package (Table 22.2) provides technical
clinical, regulatory, economic, and social strategies. The typical goals measures and resources to help countries implement all of the FCTC
of a comprehensive tobacco control program are to (1) prevent initiation demand-reduction provisions (http://www.who.int/tobacco/mpower/
among youth and young adults; (2) promote quitting; (3) eliminate en/). According to a 2017 world tobacco control report done col-
exposure to second-hand smoke; and (4) identify and eliminate laboratively by the WHO and the US National Cancer Institute (NCI),
tobacco-related disparities among population groups.15 The optimal the FCTC has had a galvanizing effect on efforts related to tobacco
mix of strategies to achieve these goals for a particular population control and has resulted in some progress to date, yet the majority of
requires careful consideration. Implementation of any one type of the world’s population remains uncovered by the most effective tobacco
strategy is substantially less effective than the coordinated implementa- control interventions.19 The report calls for “continued research and
tion of some level of all strategies. The combination of these approaches surveillance of the epidemic and implementation of the evidence-based
has been tremendously successful in reducing the number of tobacco strategies set forth in the WHO FCTC, as well as vigilant monitoring
users over the last 50 years.10 According to the CDC, states that have of the tobacco industry’s tactics and strategies to undermine or subvert
made larger investments in tobacco control have seen larger declines tobacco control efforts.”19
in cigarette sales than the United States as a whole, and the prevalence For more on tobacco, particularly as it relates to individual risks
of smoking among adults and youth has declined faster as spending and clinical interventions, the reader is referred to Chapter 24.
for control has risen.15 Consequently, the CDC recommends state-
specific annual funding levels for tobacco control. Unfortunately, most Alcohol
states are not providing funding at their recommended level, missing
a tremendous opportunity to reduce the burden of tobacco use within The acceptability of alcohol in many countries often overshadows the
their populations and across the nation.15 Important to note, with fact that it is a recreational drug with psychoactive properties and the
the emergence and rapid adoption of e-cigarette use among youth, it potential to induce dependence, in addition to being a component
is critical to ensure that these devices are included among all forms cause in approximately 200 different injury conditions and diseases,
of tobacco control and prevention strategies. including cancer.20 There is a complex array of influences on alcohol
Internationally, cigarette consumption is increasing in low- and use at both the societal and the individual levels. The reader is referred
middle-income countries (LMICs) owing to their population and to the WHO Global Status Report on Alcohol and Health for a more
economic growth, but also because of increased social acceptability in-depth review of these factors.20
of smoking and targeted marketing by tobacco companies.16 Of the Based on data from the WHO, more than one-third of the world’s
world’s smokers, 80% reside in LMICs. China is the largest consumer population aged 15 or older are current drinkers.20 In the United
of cigarettes in the world, consuming more than one-third of the States alone, almost 90% of those aged 18 or older report use of
world’s cigarettes,16 and with a population of smokers larger than the alcohol at some point in their lives, and 72% report use within the
entire population of the United States.17 Other countries with high last year. Prevalence of alcohol consumption varies widely around the
rates of consumption include Russia and Greece, and many countries globe, as do drinking patterns. In general, wealthier nations consume
of eastern Europe and in the eastern Mediterranean region.18 Africa more alcohol than do poorer nations. High-income countries have a
represents the greatest risk for future growth in tobacco use as its much higher prevalence of current drinkers and of heavy episodic
population continues to grow and as it continues to develop economi- drinking, 70% and 22%, respectively, than do low-income countries,
cally.18 In general, the prevalence of smoking among men is much where the equivalent rates are 18% and 12%, respectively.20 There
higher than it is among women, especially in Russia and Indonesia, are also large gender differences in the use of alcohol, with women
where at least half of all men smoke, but just 17% and 4% of women more often being lifetime abstainers than men and generally consuming
smoke, respectively.18 In China, 45% of men smoke but just 2% of less alcohol and being less likely to engage in heavy episodic drinking
340 Part I: Science and Clinical Oncology

when they do drink.20 However, alcohol use has been steadily increasing beverage in each study is the one that demonstrates the greatest risk.28
among women, given globalization and the changing roles of women The amount of alcohol consumed appears to be more important
in society.21 This trend could have significant public health impacts, than the type.
as women may be more vulnerable to alcohol-related harm for a given Further supporting the role of alcohol in cancer are studies that
level of use,21 but also because of the known health consequences demonstrate the reversal of risk seen after cessation of drinking. At
associated with alcohol use during pregnancy. Age also affects alcohol least three pooled analyses have suggested significant reductions in risk
consumption, with adolescents having a higher prevalence of monthly of head and neck, esophageal, and liver cancers after alcohol cessation,
heavy episodic drinking than adults.20 Although the most frequently although significant timeframes were required for former drinkers’ risk
consumed type of alcohol varies regionally, spirits constitute half of to equal the risk of never-drinkers. In a 2013 meta-analysis of nine
the total alcohol consumed globally.20 case-control studies examining the effect of cessation on laryngeal
and pharyngeal cancers, a risk reduction of 2% per year was seen, on
The Role of Alcohol in Cancer average, for quitters.29 Although it took over 35 years for the risks of
In 2007, after a week-long review of the evidence by experts, the WHO laryngeal and pharyngeal cancers among quitters to equal the risks
International Agency for Research on Cancer (IARC) concluded that of never-drinkers, 5 years was enough time to see a 15% reduction
cancers of the mouth, pharynx, larynx, esophagus, liver, colorectum, in the alcohol-related elevated risk of these cancers.29 In 2011, Rehm
and breast were causally related to alcohol consumption.22 Bagnardi and colleagues examined the effect of alcohol cessation on risk of
and colleagues provided the most recent relative risk (RR) estimates for esophageal and head and neck cancers in a pooled analysis of 13
the effects of alcohol consumption on these cancers, and others, based studies.30 This report identified significantly increased risks for both
on 572 studies with over 486,000 cancer cases.23 In this meta-analysis, cancers in the 5- to 10-year period immediately following cessation,
light (≤12.5 g of alcohol per day), moderate (≤50 g/day), and heavy with risks then declining but not reaching levels of never-drinkers
(>50 g/day) consumption of alcohol all increased the risk of oral until more than 20 years after quitting.30 Another meta-analysis, by
cavity–pharyngeal and esophageal squamous cell carcinoma (ESCC) in Heckley and colleagues, found a significantly increased risk of liver
a dose-dependent fashion, with heavy drinkers having approximately cancer in the 5 to 10 years after cessation, with a 6% to 7% reduction
five times the risk of these cancers compared with nondrinkers and in risk per year thereafter, with an estimated 23 years required for risk
occasional drinkers (oral cavity and pharynx: RR, 5.13 [95% CI, equivalence between quitters and never-drinkers.31 The results of these
4.31–6.10]; ESCC: RR, 4.95 [95% CI, 3.86–6.34]).23 These were by far studies are consistent with the long latency period of most cancers
the largest RRs detected in the study. Light drinking was not associated but nevertheless demonstrate the reversibility of the increased risk
with cancer of the larynx, although moderate drinking increased the conferred by alcohol. Despite long timeframes required for risk among
risk of this cancer by 44% (RR, 1.44 [95% CI, 1.26–1.66]), and quitters to equal that of never-drinkers, these findings suggest that
heavy drinking more than doubled the risk (RR, 2.65 [95% CI, alcohol cessation efforts are likely to result in a public health benefit
2.19–3.19]).23 Female breast cancer also exhibited a dose-response because significant and often substantial decreases were seen in the
relationship with alcohol, with light drinking being associated with a risks of these alcohol-associated cancers in the short to medium term.
4% increase in risk (RR, 1.04 [95% CI, 1.01–1.07]), moderate drinking Finally, some studies have documented protective effects of low-
conferring a 23% increase in risk (RR, 1.23 [95% CI, 1.19–1.28]), to-moderate alcohol consumption on other health outcomes, notably
and heavy drinking resulting in a 61% risk increase (RR, 1.61 [95% those related to coronary heart disease (CHD) and myocardial infarc-
CI, 1.33–1.94]).23 As with laryngeal cancer, light drinking was not tion.32 However, there is some evidence to suggest that the observed
associated with CRC in either men or women, but moderate and protective effect of alcohol on cardiovascular disease seen in many
heavy drinking significantly increased the risk of this cancer for men by epidemiologic investigations is due to residual or unmeasured confound-
21% (RR, 1.21 [95% CI, 1.11–1.32]) and 53% (RR, 1.53 [95% CI, ing and/or misclassification bias.33 Yet, as reported in a review of
1.30–1.80]), respectively. Cancers for which the risk was elevated only findings from the Nurses’ Health Study, moderate (0.1–14.9 g/day)
for heavy drinkers included cancers of the gallbladder (RR, 2.64 [95% alcohol intake remained associated with a significantly reduced risk
CI, 1.62–4.30]), liver (RR, 2.07 [95% CI, 1.66–2.58]), stomach (RR, of CHD in that cohort even after controlling for critical lifestyle risk
1.21 [95% CI, 1.07–1.36]), pancreas (RR1.19 [95% CI, 1.11–1.28]), factors and incorporating detailed information on amount and frequency
and lung (RR, 1.15 [95% CI, 1.02–1.30]).23 The elevated risk of of consumption.34 The review also notes that the same level of consump-
lung cancer may be due to residual confounding by smoking, as a tion was associated with an increased risk of breast cancer and bone
previous meta-analysis did not show an association between alcohol fractures among the Nurses’ Health Study women.34 Such a compre-
and lung cancer in never smokers.24 Other findings from the 2014 hensive assessment of all the potential health effects of alcohol consump-
meta-analysis include a possible positive association between alcohol tion is challenging, and the balance of risks and benefits is likely to
use and melanoma and prostate cancer; an inverse linear association differ by population and/or other factors. At a more global level, a
with lymphomas; and no associations between alcohol use and ovarian, 2015 prospective cohort study of nearly 115,000 adults from 12
cervical, endometrial, bladder, and brain cancers. With regard to countries across the human development index concluded that current
prostate cancer, a 2016 systematic review and meta-analysis by Zhao drinking was not associated with a net health benefit.35 However, the
and colleagues demonstrated a statistically significant dose-response follow-up time of this study was less than 5 years, and more time is
relationship between level of alcohol consumption and increased risk likely needed to observe the full range of risks and benefits associated
of prostate cancer, with risks being of similar magnitude to the risks with low-to-moderate drinking. More research is needed in this area
detected in the Bagnardi and colleagues meta-analysis.25 Nevertheless, to better understand the balance of risks and benefits of alcohol intake
the literature is mixed on this topic, and the World Cancer Research in particular settings.
Fund (WCRF)/AICR currently states that the evidence is “limited—no In sum, according to the latest consensus of evidence, alcohol has
conclusion” for alcoholic beverages and prostate cancer.26 been shown to convincingly increase the risk of at least six cancers:
An important aspect of alcohol use that has been more difficult to (1) oral and pharyngeal cancer; (2) laryngeal cancer; (3) ESCC; (4)
study, and was not and could not be examined in the comprehensive CRC (in men only); (5) breast cancer; and (6) liver cancer. The risk
meta-analysis by Bagnardi and colleagues, is pattern of use. However, is strongest for oral and pharyngeal cancers, laryngeal cancer, and
in a 2015 study by Cao and colleagues, regularity of drinking and ESCC. A strong, positive dose-response relationship is seen for all of
heavy episodic drinking were not associated with cancer risk, after these cancers with the exception of liver cancer, in which only heavy
adjustment for total alcohol intake, in the Nurses’ Health Study and the consumption appears to increase risk. There is evidence for probable
Health Professionals Follow-Up Study.27 Analyses of beverage type have increased risk for stomach cancer and for CRC in women. There is
not produced consistent findings; and in general the most-consumed no safe level of alcohol use for cancer prevention, because even relatively
Lifestyle and Cancer Prevention  •  CHAPTER 22 341

low levels have been shown to increase risk of particular cancers, and Within the liver, it is likely that oxidative stress plays an important
the amount of alcohol consumed matters more than the type of beverage role in alcohol-related carcinogenesis, because hepatic levels of AA
consumed. And as with smoking, the increased risk conferred by are relatively low after intake of alcohol.45 The CYP2E1 enzyme
alcohol use is reversible, with former drinkers assuming the same level involved in the metabolism of ethanol to AA is capable of producing
of risk as never-drinkers within 20 to 35 years of cessation. reactive oxygen species (ROS) that result in the formation of lipid
Because alcohol use varies across the globe, the attributable frac- peroxidation products (e.g., 4-hydroxynonenal) that are capable of
tions of cancer cases and deaths due to alcohol also vary depending forming highly mutagenic DNA adducts. One such adduct results
on world region and country. According to recent estimates, alcohol in a mutation of TP53 that leads to cells being more resistant to
was responsible for 5.5% of cancer cases and 5.8% of cancer deaths apoptosis and confers a growth advantage.46 Similar to ADH and
globally in 2012.36 By gender, men’s alcohol-attributable fractions were ALDH, CYP2E1 contains polymorphisms that have been associated
twice those of women’s for both overall incidence and mortality.36 By with different levels of enzyme activity. However, results of individual
geographic region, the attributable fractions for incidence and mortality studies examining an association of these variants with liver cancer in
were highest in the Western Pacific region, where alcohol accounted various populations have been inconclusive. A 2017 meta-analysis of
for 7.1% of cancer cases and 7.8% of cancer deaths; but among men 16 studies including 1737 liver cancer cases and 2947 controls found
in this region, the fractions rose to 9.1% and 9.6%, respectively.36 no association between two CYP2E1 variants and risk of liver cancer.47
This region includes China, Japan, Korea, Australia, New Zealand, In addition to CYP2E1-related mechanisms, chronic consumption of
and many other smaller Asian countries and islands of the South alcohol may also result in inflammation-driven oxidative stress, hepatic
Pacific. Unsurprisingly, the lowest attributable fractions were found iron overload, and increased nitric oxide production, all additional
in the Eastern Mediterranean region, which consists largely of Muslim sources of ROS.37,45
countries. In this region, alcohol was responsible for less than 1% of Within the breast, alcohol’s effect on estrogen levels is thought
both cancer cases and deaths.36 Among cancer sites, ESCC had the to be an important mechanism of carcinogenesis, in addition to the
largest number of cases (44.7%) and deaths (43.7%) attributable to aforementioned mechanisms of alcohol metabolism, because breast
alcohol, followed by oral cavity and pharynx (36.7% of cases and 34.2% tissue has the ability to metabolize ethanol at low levels. Estrogens
of deaths due to alcohol) and larynx (26.1% of cases and 24.7% of have a proliferative effect on breast epithelial cells, and it has been
deaths due to alcohol). Less than 10% of colorectal and breast cancer well documented that prolonged exposure to estrogens increases the
cases and deaths were due to alcohol.36 Among women, breast cancer risk for breast cancer.48,49 And alcohol has been shown to increase
was the number one cancer attributable to alcohol, whereas ESCC estrogen and other sex hormone levels in both premenopausal
was number one among men.36 and postmenopausal women. A meta-analysis of eight prospective
studies involving postmenopausal women documented significant
Proposed Mechanisms Linking Alcohol With Cancer increases in all sex hormones in women who consumed 20 g of
The mechanisms behind the carcinogenic effects of alcoholic beverages alcohol per day compared with nondrinkers.50 A randomized trial of
are not entirely clear, although they are likely to be organ specific. 51 postmenopausal women showed that serum estrone sulfate and
Although ethanol itself is considered carcinogenic, it is its metabolite, dehydroepiandrosterone sulfate (DHEAS) were significantly elevated
acetaldehyde (AA), that is likely responsible for much of the carcinogenic by 8% and 5%, respectively, in those who consumed 15 g of alcohol
effect of alcohol consumption, at least within the upper and lower per day;51 and in women who used hormone replacement therapy
gastrointestinal (GI) tracts, where it could come into direct contact (HRT), the effect of alcohol was even more potent, resulting in a
with tissues and where its concentration would be high enough to threefold increase in plasma estradiol among those who drank 15.6 g
exert an effect.37 Ethanol is metabolized into AA by the alcohol of alcohol per week.52 Among premenopausal women, one of the
dehydrogenase (ADH) and cytochrome P450 2E1 enzymes; AA is earliest studies documented a significant increase in plasma estradiol
subsequently degraded to acetate (noncarcinogenic) by the aldehyde levels in healthy women after acute administration of alcohol (0.695 g/
dehydrogenase (ALDH) enzyme. Salivary AA increases as ethanol kg), whereas levels did not change significantly after ingestion of a
ingestion increases, and increased levels of AA have been documented placebo.53 Controlled-diet studies in this population have found similar
in the saliva of those with and without cancer after moderate alcohol results.54,55 Authors of some of these studies have postulated that the
consumption; and experiments in rats and piglets have demonstrated increase in circulating sex hormones may be due to an increase in the
mutagenic levels of colonic AA after administration of ethanol.37 AA hepatic redox state that is associated with the catabolism of ethanol.53
has been shown to induce DNA damage through the formation of Another possible mechanism for the alcohol-mediated increase in
adducts and the inhibition of DNA repair, as well as being shown to circulating sex hormones is the enhanced conversion of testosterone
alter DNA methylation.38 The effect of alcohol and AA has been to estrogen resulting from increased aromatase activity that is seen
shown to be modulated by genetic factors, mainly polymorphisms in after chronic alcohol exposure.56,57 These effects likely operate through
the genes for ADH and ALDH that affect enzymatic activity and the estrogen receptor (ER)–dependent pathways. Epidemiologic studies
amount of AA within saliva and other tissues, and the risk of alcohol- strongly suggest that alcohol use is more strongly associated with ER+
related cancers.28,37 As an example, the ALDH2*2 allele is prevalent tumors than with ER− tumors.58 In addition, experimental studies
among Asians, and those who are homozygous for this allele are unable have shown that ethanol can stimulate proliferation of ER+ breast
to metabolize AA and therefore cannot tolerate alcohol. Those who cancer cells but not ER− ones, and furthermore that ethanol increases
are heterozygous for ALDH2*2 can metabolize only small amounts transcriptional activation of the ER.59 However, the ER pathway alone
of AA and generate threefold higher concentrations of AA in their cannot explain the effects of alcohol on ER+ tumors. Other mechanisms
serum and saliva when they drink, compared with those who are that are currently being investigated include alcohol’s potential ability
homozygous for the normal allele (ALDH2*1).39 A number of studies to promote the epithelial-mesenchymal transition in breast cancer
carried out in the Japanese population have established the mutant cells, to activate stromal cells and influence tumor-stroma interactions,
ALDH2*2 allele as a strong risk factor among drinkers for cancers of and to lead to epigenetic dysregulation by possibly interfering with
the upper aerodigestive tract—most particularly, esophageal cancer.40,41 folate metabolism.59
The mechanism behind the increased risk of carriers of the mutant Within the colorectum, changes in methylation and folate metabo-
ALDH allele may be the formation of DNA adducts, as studies in lism are also believed to be responsible for at least some of the carci-
Japanese drinkers have demonstrated that those with the mutant allele nogenic effects of alcohol.28,37 Numerous epidemiologic studies have
harbor more AA-derived DNA adducts in their peripheral lymphocytes documented a protective effect of folate on risk of CRC and adeno-
than do drinkers without the allele,42 in addition to having more sister matous polyps.60 Alcohol is a known inhibitor of folate metabolism,
chromatid exchanges and micronuclei.43,44 leading to reduced methionine synthesis and subsequently resulting
342 Part I: Science and Clinical Oncology

in abnormal DNA methylation, which is frequently observed in its associated harms. Many of these actions mirror actions used in
colorectal neoplasia.61 Many studies have documented an interaction tobacco control. Taxation, restriction of availability, and regulation
between folate and alcohol, wherein those with low folate intake and of marketing and advertising are common, effective tools not only
high alcohol consumption have a much greater risk of CRC than do for tobacco control, but also for alcohol control.20 Taxes and pricing
those exposed to either risk factor alone.61 Heavy use of alcohol can policies are perhaps some of the more impactful interventions that
lead to reduced intake of foods rich in folate and other B vitamins can be implemented not only to reduce drinking, but also to reduce
and may interfere with intestinal absorption and/or the metabolism alcohol-related mortality. A 2009 meta-analysis of 112 studies providing
of these key nutrients.28 The effect of folate and alcohol on risk of over 1000 estimates of the tax/price–consumption relationship found
CRC may be modified by known polymorphisms in the genes involved that a 10% increase in the price of alcohol resulted in a 5% reduction
in DNA methylation and alcohol and folate metabolism. Perhaps the in drinking; and that price affected drinking of all types of beverages
most extensively studied polymorphisms are those in the MTHFR and across all populations of drinkers, from light to heavy users.66
gene, which is central to the formation of methionine. One polymor- Authors noted that effects were large compared with what is typically
phism in particular, C677T, appears to interact with levels of folate seen for preventive interventions. A subsequent 2010 systematic review
and alcohol intake to influence risk of CRC.28,62 In addition to alcohol’s of 50 studies containing 340 estimates by the same group documented
antifolate effects, other alcohol-related carcinogenic mechanisms are that tax/price policies could significantly reduce alcohol-related
likely at play in the colorectum, including inflammation and the morbidity and mortality, with a 35% reduction in alcohol-related
production of ROS and AA. mortality expected from a doubling of alcohol taxes.67 Because of their
Other mechanisms that may be responsible for some of the car- relatively low cost to implement and their documented large and
cinogenic effects of alcohol include the presence of other carcinogens significant impacts on drinking and related outcomes, tax and pricing
in alcohol in addition to ethanol and AA, such as N-nitrosamines, interventions are strongly recommended by both the US Community
and the ability of alcohol to enhance the absorption or activity of Preventive Services Task Force (https://www.thecommunityguide.org/
other carcinogens (e.g., tobacco).30 topic/excessive-alcohol-consumption) and the WHO for population-
level alcohol control.20
Evidence-Based Interventions for Cancer Prevention Related
to Alcohol Use Obesity
Published cancer prevention recommendations from the ACS and Obesity is now considered an epidemic, with approximately
the AICR/WCRF suggest that if alcohol is consumed, it should be in 17% of children and 36% of adults classified as obese.68 Accord-
moderation, which is defined as up to one drink per day for women ing to the IARC, there is abundant evidence supporting the link
and two per day for men.63,64 Interventions to reduce alcohol consump- between obesity and the following 13 different types of cancers,
tion are available at both the personal (or clinical) and population now considered obesity-related cancers: cancers of the esophagus,
level. At the personal level, the US Preventive Services Task Force gastric cardia, colon and rectum, liver, gallbladder, and pancreas;
(USPSTF) recommends “that clinicians screen adults aged 18 years postmenopausal breast cancer; endometrial, ovarian, and renal cell
or older for alcohol misuse and provide persons engaged in risky or cancers; meningioma; thyroid cancer; and multiple myeloma.69 In
hazardous drinking with brief behavioral counseling interventions addition, a systematic review conducted by Birks and colleagues70
to reduce alcohol misuse.”65 This is a Grade B recommendation, demonstrated a link between deliberate weight loss in overweight or
meaning that there is either high certainty that the net benefit of obese individuals and reduced cancer incidence. This section reviews
this intervention is moderate or that there is moderate certainty the role of obesity in cancer and proposed mechanisms linking obesity
that the net benefit is moderate to substantial. As of the writing with cancer.
of this chapter, this recommendation was in the process of being
updated by the USPSTF. Screening for alcohol can be done in one Role of Obesity in Cancer
of three ways: with (1) the Alcohol Use Disorders Identification Test In obesity, adipose tissue typically accumulates in two different regions:
(AUDIT); (2) the abbreviated AUDIT-Consumption (AUDIT-C) the android region or abdominal area, and the gynoid region, which
screen, or (3) the single-question screen, “How many times in the consists of the hips and thighs. Android obesity differs from gynoid
past year have you had five (for men) or four (for women and all obesity such that it contains two different types of adipose tissue
adults older than 65 years) or more drinks in a day?”65 Each of these deposits, subcutaneous and visceral. The latter, visceral adipose tissue
screening tools has demonstrated good sensitivity and specificity for (VAT), is more metabolically active compared with subcutaneous
detecting alcohol misuse across multiple populations, but the AUDIT adipose tissue, is considered the impetus for metabolic dysfunction,
screen has been the most widely studied in the primary care setting.65 and may contribute to increased cancer risk in obese individuals.71,72–76
After screening, brief interventions that require multiple contacts Therefore, excessive accumulation of VAT is considered to be a major
between provider and patient, with each contact lasting at least 6 contributor to obesity’s role in cancer.
to 15 minutes, demonstrate the strongest evidence for effectiveness; Epidemiologic evidence supports the relationship between VAT
single-contact interventions lasting 5 minutes or less have limited and increased cancer risk. In an observational study conducted by
effect.65 The USPSTF documents that these types of interventions Britton and colleagues,77 adiposity measurements in four different
delivered in the primary care setting can positively affect unhealthy areas (subcutaneous, visceral, periaortic, and pericardial) were compared
drinking behaviors in adults, including reducing the amount of alcohol with incident cancer, cardiovascular disease, and all-cause mortality
consumed weekly, reducing the proportion of persons who engage in over a median 5-year follow-up period. In this study, 3086 middle-aged
episodes of heavy drinking, and increasing long-term adherence to men and women (average age, 50 years ± 10) were included. VAT,
drinking recommendations. It is important to note that the USPSTF but not other adipose areas, was significantly associated with cancer
recommendation distinguishes between those who “misuse” alcohol and incidence (n = 141 incident cancer cases; hazard ratio [HR], 1.43
those who “abuse” alcohol and are dependent on it. There is limited [95% CI, 1.12–1.84]) when the model was adjusted for clinical factors
evidence that these types of brief behavioral interventions are ineffective (e.g., age, sex, blood pressure, body mass index).77 When the sample
in the setting of alcohol abuse or dependence. In such cases the benefits was stratified by sex, there were no significant differences observed
of specialty treatment are well established, and this type of treatment is among adipose areas and cancer incidence; however, HRs were higher
recommended.65 in men compared with women for VAT.77 These findings support the
At the population level, a number of actions can be taken by local, relationship between VAT and cancer incidence in middle-aged men
state, and/or federal authorities to reduce the use of alcohol and prevent and women.
Lifestyle and Cancer Prevention  •  CHAPTER 22 343

Similarly, Murphy and colleagues78 compared the relationship of colon cancer in men.80 Currently proposed mechanisms for these
subcutaneous, visceral, intramuscular, and total-body adiposity with associations vary depending on cancer site. For example, in ovarian,
incident cancer over a 13-year follow-up period in a study that included endometrial, and breast cancers, some studies have shown that cell
2519 older men and women (average age, approximately 74 years). growth is facilitated by leptin binding to ER-α.79 Overall, there is
In considering all incident cancers (n = 617), a significant association evidence to support obesity-induced alterations in adipokines as an
between total adiposity, VAT, and cancer incidence was observed in enabler of tumorigenesis.
women (HR per standard deviation [SD] increase, 1.14 [95% CI, Considering that adiponectin is associated with insulin sensitivity,
1.01–1.30]; and 1.15 [95% CI, 1.02–1.30], respectively) but not in in the obese state a reduction in adiponectin also contributes to a
men when clinical factors were adjusted for.78 Regarding obesity-related reduction in insulin sensitivity and subsequent insulin resistance.81
cancers (n = 224), after adjustment for clinical factors, a significant Insulin resistance facilitates the tumor microenvironment via two
association between total adiposity but not other adiposity measures, interrelated mechanisms. First, increased insulin levels result in a
and obesity-related cancer incidence was observed in women (HR per reduction of insulin-like growth factor–binding proteins 1 and 2
SD increase, 1.23 [95% CI, 1.03–1.46]) and a significant association (IGFBP1, IGFBP2) and subsequent increase in insulin-like growth
between VAT and obesity-related cancer incidence was observed in factor 1 (IGF1), ultimately resulting in increased cellular proliferation
men (HR per SD increase, 1.30 [95% CI, 1.06–1.60]).78 Overall, and decreased apoptosis via the AKT/mTOR pathway.81 Second,
evidence from these prospective cohort studies support the role of increased insulin, IGF1, and TNF-α reduce the amount of sex
obesity, and specifically VAT, in cancer. hormone–binding globulin produced, which increases cancer risk
because of the resultant increase in levels of sex hormones.81 Although
Proposed Mechanisms Linking Obesity With Cancer the mechanisms related to estrogen and cancer risk vary depending
Although the relationship between obesity and cancer is evident from on cancer type, according to Teoh and Das,81 estrogen may cause
observational data, the physiologic mechanisms explaining this relation- replication errors during DNA synthesis and increase cellular prolifera-
ship have recently emerged in the literature.72,79–81 Essentially, the tion; hence the number of mutations accumulates over time, yielding
excessive accumulation of adipose tissue, characterized by adipocyte tumor growth. In addition to insulin serving as a contributing factor
hypertrophy and hyperplasia,80 leads to a state of adipocyte hypoxia for increased estrogen and protumorigenic effects, it is important to
and resultant macrophage activity, and altered adipokine profiles; these note that adipose tissue is a major site for estrogen synthesis in
factors promote an interplay among systemic inflammation, sex postmenopausal women and men; thus, increased adipose tissue leads
hormones, and insulin resistance, all of which elicit conditions that to increased estrogen production, and consequently a greater ratio of
promote tumorigenesis. circulating estrogens to androgens may lead to cancer.81 Taken together,
According to a review conducted by Teoh and Das,81 compared it is evident that excessive accumulation of adipose tissue induces
with lean counterparts, adipocyte samples from obese humans and protumorigenic conditions.
animal models revealed a state of hypoxia within the adipose tissue via
presence of hypoxia-inducible factor (HIF), lower levels of oxygen, and Physical Inactivity
lactate. This demonstrated state of hypoxia causes macrophage recruit-
ment via signaling pathways such as chemokine ligand 2, interleukin Physical activity consists of two subtypes of activity: exercise or
(IL)-1β/C-X-C motif chemokine ligand 12.81 Macrophages remain structured physical activity, which is defined as activity pursued for
in the adipose tissue because of increased expression of netrin-1, a the purpose of improving fitness, and lifestyle physical activity (i.e.,
protein that has been proposed to block migration of macrophages completing house chores, walking to work, taking the stairs). Therefore
and is expressed when fatty acid palmitate is saturated.81 Of note, physical inactivity is identified when one not only does not engage
fatty acid palmitate is saturated within the obese state secondary to in exercise but also has minimal lifestyle activity.
the increase in lipolysis and consequent increase in circulating free
fatty acids. Role of Physical Inactivity in Cancer
Next, the macrophages transition from an M2 prorepair to an M1 Current public health guidelines recommend 150 minutes per week
proinflammatory state, activating nuclear factor-κB (NF-κB) and signal of moderate-intensity or 75 minutes per week of vigorous-intensity
transducer activator of transcription 3 (STAT3).79 NF-κB has been aerobic activity, and at least 2 days per week of strength training
linked with reduction in cellular senescence, facilitation of the epithelial- exercise.82 Compliance with these guidelines may be measured sub-
mesenchymal transition, and an increase in proinflammatory cytokines jectively, through self-report, or objectively, through measurement or
such as tumor necrosis factor–α (TNF-α) and IL-6.81 STAT3 increases estimation of peak oxygen consumption via a maximal or submaximal
expression of cyclins and antiapoptotic proteins, resulting in an increase cardiopulmonary exercise test respectively. Overall, both subjective
in cellular proliferation and decrease in cellular apoptosis.79 These and objective evidence is available linking regular, structured physical
cytokines play different roles depending on cancer type. For example, activity with reduced risk of chronic disease and certain cancers such
in breast cancer, macrophage activation of NF-κB leads to increased as colon and breast cancer.82–88 An inverse relationship has been
expression of cytokines (i.e., IL-1β, TNF-α, prostaglandin E2 [PGE2]) demonstrated between increased physical activity levels and reduction
that increase transcription of CYP19, a transcription factor of aromatase, in cancer risk.82,88 Moreover, a retrospective analysis conducted by Lee
and result in increased bioavailability of estrogen.80 Overall, macrophage and colleagues89 found that individuals who did not engage in regular,
activity secondary to hypoxic conditions from obesity promotes structured activity were 1.32 times more likely to develop colon cancer
tumorigenic conditions. and 1.33 times more likely to develop breast cancer than physically
Along with macrophage activity, adipokine profiles, notably adi- active individuals.
ponectin and leptin, are altered in the obese state. Normal physiologic In addition to structured physical activity, when lifestyle activity
levels of adiponectin are linked with enhancement of insulin sensitivity decreases, it is typically replaced by increased sedentary behavior. A
and reduction in angiogenesis.72,81 However, in the obese state, adi- meta-analysis by Schmid and Leitzmann90 noted that colon cancer
ponectin levels decrease owing to instability of adiponectin mRNA risk increased by 8% and endometrial cancer risk by 10% for every
and subsequent reduction in gene expression.81 In contrast, under 2-hour increase in total sitting time per day. In addition, in a prospective
normal physiologic conditions, leptin promotes satiety and feeding cohort study conducted by Nomura and colleagues,91 a significant
cessation; yet in the obese state, leptin levels increase. The increase association with sitting time and breast cancer incidence was observed
in leptin is associated with increases in cellular proliferation and when individuals spent more than 10 hours sitting per day. Taken
inhibition of apoptosis in breast, ovarian, and cervical cancers.81 together, sedentary behavior is linked with increased risk of colon,
Furthermore, increased leptin has demonstrated an association with breast, and endometrial cancer.
344 Part I: Science and Clinical Oncology

Proposed Mechanisms Linking Physical Activity With with training, yet resting levels remained unchanged, supporting the
role of SPARC as a myokine. In the first animal model, exercised mice
Reduced Cancer Risk
with azoxymethane (AOM)-induced colon tumorigenesis demonstrated
Evidence supports the impact of physical activity on cancer risk a reduction in aberrant crypt foci and aberrant crypts compared with
reduction through mechanisms related to obesity, skeletal muscle AOM nonexercised and control groups.106 These findings were also
cytokine production, and immune function. As mentioned previously, observed in the cell line model and lend support to the role of exercise
obesity-induced insulin resistance promotes tumorigenesis. However, in the reduction of colon tumorigenesis. In addition, the second
physical activity may counteract this by improving insulin sensitivity animal model compared sedentary and exercised SPARC knockout
through alterations in VAT deposition and training adaptations of mice to exercised wild-type mice and demonstrated an increase in
the skeletal muscle. apoptotic cells via caspase-3 and caspase-8 activity in exercised wild-type
Exercise without weight loss or changes in diet has demonstrated mice, with no change in apoptosis observed in either group of the
effective reduction in VAT,92–99 the primary fat depot connected to SPARC knockout mice, supporting SPARC’s role in attenuation of
increased metabolic dysfunction and promotion of tumorigenesis.77,78 tumorigenesis. Similarly, in a review conducted by Sanchis-Gomar and
The proposed mechanism for VAT reduction with exercise alone is colleagues,88 evidence supports SPARC activation of caspase-3 and
attributed to an increase in β-adrenergic receptor activity in adipo- caspase-8 to promote cellular apoptosis and prevent aberrant crypt foci
cytes,100 which results in an increase in lipolysis from abdominal fat formation in colon cancer. Overall, evidence supports SPARC’s role
depots compared with other fat depots during exercise.97,100,101 Moreover, as a myokine and as a contributing factor in reducing colon cancer.
the reduction in VAT results in an increase in adiponectin.102 As Interconnected with myokine activity is the impact of exercise-
mentioned previously, adiponectin is linked with enhancement of induced activation of natural killer (NK) cells in relation to physical
insulin sensitivity and reduction of angiogenesis.72,81 activity and cancer prevention. In a review by Idorn and colleagues109
In addition, in the obese state, impaired glucose tolerance at the a reduction in tumor incidence of greater than 60% was observed
level of the skeletal muscle exacerbates insulin resistance secondary due to exercise-induced activation of NK cells among various mouse
to the increase in circulating free fatty acids and consequent increase models. In humans, NK cells seem to function similarly, such that
in diacylglycerol instead of glucose for substrate utilization.103 However, NK cells recognize and kill cancer cells.110 During exercise the myokine
exercise-induced skeletal muscle contraction prompts increased IL-15 activates NK cells and T cells via transpresentation of IL-15/
translocation of GLUT4 receptors to the T tubules and plasma IL-15 receptor alpha complex.109 In addition, with exercise, the natural
membrane of the working muscles, which improves skeletal muscle increase in epinephrine released from β-adrenergic receptors facilitates
glucose uptake and whole-body insulin sensitivity.104 Therefore, regular the release and mobilization of NK cells.109,111 Moreover, NK cells,
physical activity, even in the obese state, improves insulin sensitivity compared with other lymphocytes, have the greatest density of
and prevents increases in bioavailability of sex hormones, two factors β-adrenergic receptors.109 Taken together, improvements in fat deposi-
that help inhibit tumorigenesis. tion and insulin sensitivity, and the activity of myokines and NK
Along with obesity-related mechanisms, there are also mechanisms cells, are all viable proposed mechanisms explaining the role of physical
unique to skeletal muscle activity—notably, release of cytokines from activity in cancer prevention.
contracting skeletal muscle, commonly referred to as myokines. To date,
identified myokines include IL-6, irisin, IL-15, calprotectin, myonectin, Diet
oncastatin M (OSM), and secreted protein acidic and rich in cysteine
(SPARC).105 According to a review by Goh and colleagues,105 during The link between diet and cancer risk is complex, such that not all
exercise myokines are released from the working skeletal muscles, which dietary components (i.e., types of food, dietary patterns, vitamins,
increases activity of antiinflammatory cytokines (i.e., IL-1 receptor and minerals) are linked with cancer risk, and some components
agonist and IL-10), decreases activity of proinflammatory cytokines, exhibit strong links with cancer risk individually whereas others exhibit
and attenuates migration of M1 macrophages and T cells to areas stronger effects synergistically. This section presents the dietary
with high expression of inflammatory cytokines or inflammation (i.e., components containing the most evidence in relation to increased or
adipose tissue and/or the tumor microenvironment). decreased cancer risk.
Currently, promising evidence is available regarding the roles of
exercise-induced OSM and irisin activity in breast cancer prevention, Dietary Components Linked With Increased Cancer Risk
and SPARC in colon cancer prevention.106–108 Within the context of Red meat and processed meats
breast cancer prevention, Hojman and colleagues 108 treated MCF-7 The majority of evidence from systematic reviews and meta-analyses of
cells (malignant, ER+, breast epithelial cells) with mouse serum collected epidemiologic evidence, randomized trials, and observational studies
immediately after exercise and demonstrated a 52% reduction in cell supports the link between high consumption of red or processed
proliferation and 54% increase in caspase activity. Furthermore, whereas meats and increased risk of CRC.64,112–114 A 15% to 20% increase in
various myokines were upregulated after exercise, OSM demonstrated CRC risk has been detected per 100-g and 50-g daily intake of red
the greatest effects on cellular proliferation, caspase activity, and and processed meats, respectively.64 A meta-analysis of epidemiologic
apoptosis.108 The efficacy of OSM was further supported in a model evidence revealed a summary RR of 1.11 (95% CI, 1.03–1.19) for
in which OSM signaling was blocked.108 CRC when red and processed meats were consumed regularly.112
Similarly, Gannon and colleagues107 assessed the influence of irisin Although significant heterogeneity was observed, the relationship was
activity specifically on malignant breast epithelial cells. Overall, irisin similar and with statistically insignificant heterogeneity when assessed
reduced cellular proliferation in aggressive breast epithelial cells through by subgroups. For example, when stratified by sex, men exhibited a
caspase activity and suppression of NF-κB.107 Taken together, current significant association with regular intake of red or processed meats and
evidence supports OSM and irisin as two contributing myokines CRC risk compared with women (RR, 1.16 [95% CI, 1.02–1.32]; and
connected to breast cancer prevention through their impact on caspase RR, 1.03 [95% CI, 0.91–1.17], respectively).112 Interesting to note,
activity. when comparing North America and Europe, North Americans were
Colon cancer is also linked with myokine activity. Specifically, the at greater risk of CRC (RR, 1.11; 95% CI, 1.03–1.19) than Europeans
myokine SPARC has been linked with reduced risk of colon cancer. (RR, 1.07; 95% CI, 0.98–1.16) when red or processed meats were
Aoi and colleagues 106 assessed SPARC in relation to exercise and colon consumed regularly,112 suggesting that there may be other lifestyle
cancer among a colon-26 carcinoma cell model, two different animal factors unique to North Americans that may contribute to CRC risk.
models, and a human subject exercise intervention. In humans, SPARC Similarly, a systematic review and meta-analysis of case-control
levels released from working muscles during exercise increased linearly and cohort studies115 revealed an 11% increase in pancreatic cancer
Lifestyle and Cancer Prevention  •  CHAPTER 22 345

risk per 100-g daily intake of red meat. When consumed regularly, Vegetarian and vegan dietary pattern
the summary risk for pancreatic cancer was 1.38 (95% CI, 1.05–1.81) In a second meta-analysis of prospective cohort studies conducted by
and 1.62 (95% CI, 1.17–2.26) for red and processed meats, respec- Godos and colleagues,123 different types of vegetarian dietary patterns
tively.115 When assessed by sex, red or processed meat consumption were compared in relation to breast, colorectal, and prostate cancer
put men at significantly greater risk of pancreatic cancer compared risk. The following diets were compared: vegetarian, defined as consump-
with women (men: red meat—RR, 1.21 [95% CI, 1.07–1.31]; tion of meat less than once per month; semivegetarian, defined as
processed meat—RR, 1.18 [95% CI, 1.06–1.31]; women: red consumption of meat more than once per month but less than once
meat—RR, 1.06 [95% CI, 0.85–1.31]; processed meat—RR, 0.99 per week; pescovegetarian, defined as consumption of fish as the only
[95% CI, 0.84–1.16]).115 In addition, a recent systematic review of type of meat, more than once per month; and nonvegetarian, defined
prospective cohort studies revealed a significant increase in gastric as consumption of meat more than once per week. Compared with
cancer risk with regular intake of processed meats (RR, 1.15 [95% the nonvegetarian dietary pattern, significant findings included a
CI, 1.03–1.29]), notably ham, bacon, and/or sausage (RR, 1.21 [95% reduction in CRC risk for semivegetarians (RR, 0.67 [95% CI,
CI, 1.01–1.46]).116 Taken together, evidence supports the link between 0.53–0.83]) and pescovegetarians (RR, 0.86 [95% CI, 0.79–0.94])
regular intake of red or processed meat and risk of colorectal, pancreatic, but not vegetarian (RR, 0.88 [95% CI, 0.74–1.05]), with no significant
and gastric cancer. For colorectal and pancreatic cancer, these effects heterogeneity among studies.123 In a different meta-analysis comparing
seem to be more relevant to men. vegetarian and vegan dietary patterns with nonvegetarian patterns
among 86 cross-sectional and 10 prospective cohort studies,124 a
High-salt foods and salt intake significant reduction in cancer incidence was observed in both the
Unique to processed meats is the high salt content. Regular consumption vegetarian and vegan patterns (RR, 0.92 [95% CI, 0.87–0.98]), with
of high-salt foods (i.e., processed and prepackaged foods) has also greater risk reduction observed in the vegan pattern (RR, 0.85 [95%
been linked with an increased risk of gastric cancer (RR, 1.55 [95% CI, 0.75–0.95]). Taken together, these findings suggest an association
CI, 1.17–2.05]).116 Similar trends were observed for salted fish (RR, between lower intake of meat and cancer risk reduction.
1.25 [95% CI, 1.07–1.47]) and salt added to foods (RR, 1.11 [95%
CI, 1.05–1.16]).116 Furthermore, a 12% increase in gastric cancer risk Mediterranean dietary pattern
has been observed per 5 g/day increase of total dietary salt intake Although there is variability in the definition of the Mediterranean
(RR, 1.12 [95% CI, 1.02–1.23]).116 dietary pattern (MDP), it typically consists of the following dietary
components: fruits, vegetables, nuts, legumes, whole grains, fish, high
Proposed mechanisms of dietary components linked with monounsaturated to saturated fat ratio, and minimal amounts of dairy
increased cancer risk and meat or processed meats.125 In a review by Schwingshackl and
When red meat is prepared at high temperatures (i.e., frying, charcoal Hoffmann125 of randomized controlled trials (RCTs), case-control
broiling or grilling, smoking), the high heat elicits production of studies, and prospective cohort studies, regardless of study design, a
heterocyclic amines and polycyclic hydrocarbons,64,117 which are known significant reduction in breast, colorectal, prostate, gastric, and liver
carcinogenic compounds secondary to their impact on DNA damage.118 cancer was observed among individuals following the MDP compared
These compounds have been linked with increased colorectal and with other patterns. When components of MDP were assessed individu-
pancreatic cancer risk.64,117 In addition, although the high fat content ally in relation to cancer risk, significant effects were not observed,
of certain cuts of red meat is not linked with pancreatic or gastric suggesting a synergistic effect among the components in relation to
cancer, it is associated with CRC.119 This is attributed to excess produc- cancer risk.125 In addition, results from a pooled analysis of case-control
tion of bile acids, resulting in increased concentrations of bile acids studies demonstrated a significant inverse relationship between
in the colon and/or rectum, leading to cellular proliferation and possible components of MDP and endometrial cancer risk, such that as the
tumorigenesis.64,119 number of components consumed increased (i.e., from 0–3 to 7–9),
Moreover, the presence of N-nitroso compounds in nitrite-preserved risk of endometrial cancer decreased.126 The largest number of com-
processed meats may contribute to colorectal, gastric, and pancreatic ponents consumed elicited an RR of 0.43 (95% CI, 0.36–0.68).
cancer risk.64,116,117,119,120 In CRC specifically, the alkaline nature of the Furthermore, a risk reduction of 0.84 (95% CI, 0.80–0.88) was
N-nitroso compounds facilitates DNA damage.119 In gastric cancer, the observed as dietary component consumption increased by one.126
nitrites in processed red meats interact with gastric acids to produce In an RCT comparing a low-fat diet and two different MDPs that
N-nitroso compounds, which enables tumorigenesis.116,117,120 Although manipulated the primary fat source (i.e., MDP with extra virgin olive
processed meats are high in salt, high-salt foods and added salt in oil [EVOO], high in monounsaturated fatty acids, compared with
general have exhibited exacerbation of the carcinogenic effects of MDP with mixed nuts, a mix of saturated, monounsaturated, and
Helicobacter pylori, leading to gastric cancer.116,121,122 In addition, high polyunsaturated fatty acids) on cardiovascular-related health outcomes,
dietary salt intake alone may damage the lining of the stomach, leading breast cancer incidence was assessed as a secondary outcome.127 Overall
to inflammation, cellular proliferation, and possible tumorigenesis.116,121 MDP with EVOO demonstrated the greatest reduction of breast
cancer risk after a median follow-up of 4.8 years. Incidence rates were
Dietary Components Linked With Decreased Cancer Risk the lowest in the EVOO group (1.1 per 100 person-years) compared
Fruits and vegetables with mixed nuts or low-fat diet (1.8 and 2.9 per 100 person-years,
Existing evidence has supported fruit and vegetable intake for risk respectively).127 In addition, the RR of developing breast cancer was
reduction of the following types of cancer: lung, mouth, pharynx, significantly less with EVOO compared with mixed nuts (RR, 0.32
larynx, esophagus, stomach, and colorectal.64,113 Dietary patterns [95% CI, 0.13–0.79]; and RR, 0.59 [95% CI, 0.26–1.35], respec-
emphasizing increased intake of fruit and vegetables have been associated tively).127 These findings suggest that a fat source high in polyunsaturated
with significant reductions in cancer risk. For example, in a systematic fatty acids within the MDP may produce the greatest effects in reducing
review and meta-analysis of observational studies conducted by Godos risk of breast cancer.
and colleagues,114 healthier dietary patterns, defined as high consump-
tion of fruits, vegetables, and whole grains, compared with unhealthier Individual micronutrients
dietary patterns, defined as high consumption of red or processed In assessing individual micronutrient supplementation in relation to
meat, salty or sweet snacks, and refined grains, resulted in a 24% cancer risk, the majority of evidence has demonstrated null or adverse
reduction in CRC risk with statistically insignificant heterogeneity health effects.64,113 In a systematic review and meta-analysis conducted
(RR, 0.81 [95% CI, 0.71–0.94]).114 The specific aspects of diet that by Fang and colleagues,116 the only micronutrient exhibiting cancer
reduce cancer risk remain unclear. prevention benefits was vitamin C, which exhibited a significant
346 Part I: Science and Clinical Oncology

reduction in gastric cancer risk with increased consumption (RR, 0.89


[95% CI, 0.85–0.93]).Yet adequate intake of vitamin C can easily be The Role of Ultraviolet Radiation in Cancer
achieved through consumption of fruits and vegetables. Thus, current Exposure to UVR, whether from the sun or from a tanning device,
recommendations for nutrition and cancer prevention state that unless is a major risk factor for all forms of skin cancer (basal cell carcinoma
an individual has micronutrient deficiencies, which would require [BCC], squamous cell carcinoma [SCC], and melanoma). Despite
supplementation beyond dietary intake, micronutrient consumption being highly preventable, skin cancer is the most common form of
from a diet rich in fruits, vegetables, and whole grains will suffice in cancer in the United States, with 5 billion cases treated annually at a
terms of health benefits related to cancer prevention.113 cost of $8.1 billion.129 Melanoma accounts for a small fraction of
these cases, approximately 63,000, but it is the deadliest form of skin
Proposed mechanisms of dietary components linked with cancer, with nearly 9000 deaths each year.130 Unlike many other
decreased cancer risk common cancers, data suggest that skin cancer incidence is rising,
Increased fruit, vegetable, and whole grain intake increases dietary underscoring the need for a focus on evidence-based preventive actions.
water and fiber content. An increase in dietary water and fiber content The risk of skin cancer varies by skin type, race or ethnicity, the
may increase satiety, prevent excessive energy intake, and replace or pattern of UVR exposure, and the age at which exposure occurs. In
limit poor food choices such as refined grains, added sugars, salty general, lighter-skinned individuals have a higher risk of all skin cancers
snacks, and processed meats.64 In addition, a high-fiber diet may than do darker-skinned individuals. The Fitzpatrick skin type classifica-
decrease transit time in the colon, reducing exposure to potential fecal tion system classifies individuals based on their likelihood of tanning
carcinogens and reducing CRC risk. or burning, with six categories (I–VI) ranging from “always burns
and never tans” to “never burns, deeply pigmented, least sensitive.”
Obesity, Physical Inactivity, and Dietary Recommendations Those who are classified as types I and II have the highest risk of
burns, damage from UVR, and skin cancer.130
and Resources for Cancer Prevention Regarding race, non-Hispanic whites have the highest rates of
Recommendations for cancer prevention63 are shown in Table 22.1. invasive melanoma incidence (24.7 per 100,000) and mortality (3.4
As evidenced by results from the systematic review conducted by Birks per 100,000), whereas African-Americans have the lowest rates (1.0
and colleagues,70 weight loss in overweight or obese individuals is per 100,000 and 0.4 per 100,000, respectively).130
linked with reduction in cancer incidence. The underlying theme in The type of skin cancer for which one is at risk also varies according
weight loss is energy balance—moving more (increasing energy to his or her pattern of UVR exposure. Chronic continuous exposure,
expenditure) and eating less (reducing energy intake). Even small such as that experienced by an outdoor worker, is more strongly
changes can result in large effects over time. associated with SCC, whereas intermittent exposure, such as that
At initiation of an exercise regimen, it is recommended to start experienced by someone vacationing at the beach, is more strongly
with at least 5 days per week of moderate-intensity aerobic exercise associated with BCC and melanoma.130 In a meta-analysis of 34 studies,
(40%–60% heart rate reserve or V̇ o2 reserve) for at least 30 minutes intermittent sun exposure was associated with a significant 61% excess
per exercise session, with eventual progression to the goal of at least risk of melanoma, whereas chronic exposure was slightly inversely
60 minutes per day of aerobic exercise at more vigorous intensities associated with risk, although this was not significant.131 Numerous
(i.e., >60% heart rate reserve or V̇ o2 reserve).82 If an individual is studies have documented the increased risk of skin cancer associated
unable to complete a continuous exercise session, it is recommended with sunburns.132–134 Sunburns during childhood are strongly associated
to break up the exercise time into 10-minute increments.82 Regarding with risk of future melanoma,132,133 although data suggest that sunburns
diet, a caloric reduction of 500 to 1000 kcal/day is typically recom- at any age can significantly increase the risk of melanoma.132,133 A
mended to elicit a 1- to 2-pound weight loss per week. Dietary changes meta-analysis of 26 studies found that the risk of melanoma increases
that are often suggested for weight loss include reducing portion size with increasing number of sunburns during all life periods (i.e.,
and avoiding high–energy-dense foods, especially those with low childhood, adolescence, and adulthood).132–134 Consequently, prevention
nutrient value (e.g., potato chips, sugar-sweetened beverages). efforts should occur across the lifespan.
There are countless resources available to assist individuals with Although the sun is the most common source of UVR exposure,
energy balance to promote weight loss and maintenance of a healthy tanning beds are also an important source, particularly for non-Hispanic
weight. The CDC and healthfinder.gov provide a step-by-step guide white women aged 18 to 25, of whom nearly one-third report tanning
to help individuals with weight loss. This guide includes strategies, indoors.135 It is estimated that indoor tanning results in 400,000 cases
such as goal setting and self-monitoring, to establish healthier dietary of skin cancer in the United States each year, and 6000 of these are
habits in efforts to incorporate the behavior change component into melanoma.130 In general, the risk of skin cancer increases with increasing
weight loss. use, with younger and more frequent users having a steeply elevated
Moreover, the CDC website provides details on the activity risk.136–141 For “ever” indoor tanning, the excess risks of melanoma,
guidelines, tips on how to incorporate structured physical activity BCC, and SCC are 20%, 29%, and 67%, respectively.130,139,140 When
within one’s daily routine and increase lifestyle activity, how to begin “ever” indoor tanning is restricted to those younger than 35 or to those
an exercise program, how to measure exercise intensity, and other considered frequent users, the excess risk dramatically increases.139,140
worthwhile messages. These resources, in coordination with worksite Although the exact magnitude of the increased risk conferred by
and community efforts (e.g., the local gym, YMCA, or community indoor tanning varies quite a bit within the literature (e.g., owing to
recreation center; religious organizations; home exercise videos; wearable differences in study settings and populations), studies consistently show
devices such as an accelerometer or pedometer), aid in promoting significantly elevated risk. Important to note, there is no evidence to
increases in physical activity. In addition, the AICR’s website is also suggest that indoor tanning is safer than tanning outdoors or that it
a useful resource, providing various aids to assist in meeting cancer protects from the effects of future sun exposure.130
prevention guidelines, including a visual aid to assist with portion
control and consumption of a balanced diet. Proposed Mechanism Linking Ultraviolet Radiation to
Skin Cancer
Ultraviolet Radiation
UVR from the sun consists of three different ultraviolet (UV)
Solar radiation, specifically ultraviolet radiation (UVR), and the use wavelengths—UVA (315–400 nm), UVB (280–315 nm) and UVC
of UVR-emitting tanning devices are classified as carcinogenic to humans (100–280 nm). UVC rays do not reach the Earth’s surface owing to
by the IARC.128 stratospheric ozone and so are not considered a source of UVR damage
Lifestyle and Cancer Prevention  •  CHAPTER 22 347

to the skin. The remaining UVR reaching the surface of the earth is carcinogen and an immunosuppressive agent that has been implicated
95% UVA and 5% UVB. UVB rays have more energy but are not in all forms of skin cancer.
absorbed as deeply into the skin as UVA rays, which have less energy
but can penetrate into the dermis. UVB is significantly more carci- Evidence-Based Interventions for Cancer Prevention Related
nogenic than UVA, because it can directly and indirectly damage
DNA, whereas UVA is weakly absorbed by DNA and primarily causes to Ultraviolet Radiation Exposure
DNA damage through indirect photosensitization reactions.142,143 Reducing exposure to UVR can reduce skin cancer risk. There are
UVB directly damages DNA through the formation of DNA many actions that individuals and communities can take to reduce
“photoproducts” or dimers formed by the covalent bonding of two their UVR exposure. Unfortunately, many individuals are not aware
adjacent pyrimidine bases as a result of the absorption of UVR of the risks posed by UVR exposure. Data indicate that one-third
photons.143 A few different types of dimers can form, but the one of US adults have been sunburned in the past year and that most
most frequently induced by UVB is the cyclobutane pyrimidine dimer do not take the recommended preventive actions.147,148 In addition,
(CPD). If these are not remedied by the nucleotide excision repair indoor tanning rates are high in some groups, and skin cancer rates
system, they can result in C to T transitions or CC to TT tandem are increasing. These factors led the Surgeon General to release a
mutations. These types of mutations were once thought to be a signature report in 2014 that serves as a call to action to prevent skin cancer.130
of UVB damage; however, this has been called into question based This report outlines the preventive actions that can be taken today
on data showing that UVA exposure can also result in CPDs, although by various constituencies and at various levels of society (Table 22.3).
it is likely through an indirect mechanism.142,143 C to T mutations in The report also sets five goals to support skin cancer prevention
the TP53 gene are likely an early event in the development of SCC, across the country and discusses several strategies to achieve each goal
whereas they occur later in BCC and melanoma.143 Genes coding for (Table 22.4).
proteins of the Hedgehog signaling pathway, mainly the tumor- The use of sunscreens is a recommended action for skin cancer
suppressor gene PTCH, are frequently mutated in BCC, and many prevention, although there are few studies demonstrating the skin
of these mutations are C to T and CC to TT transitions.143 In mela- cancer–protective effect of these products. Data from the Australian
noma, the PTEN and CDKN2A genes have also been shown to harbor Nambour Skin Cancer Prevention Trial (N = 1621) suggest that the
C to T mutations. However, melanomas from intermittently sun- daily use of sun protective factor (SPF) 15+ sunscreen can reduce
exposed areas also show a high frequency of T:A to A:T mutations, the risk of SCC and melanoma. After 8 years of follow-up, SCC
such as the well-known V600E mutation in BRAF, suggesting another incidence in those randomized to daily sunscreen application was
type of UVR-induced mutation that is not fully understood.143 35% lower (0.65 [95% CI, 0.43–0.98]) than in those randomized
UVA rays primarily cause damage to DNA indirectly. Other to discretionary use (including no use).149 Ten years after cessation
chromophores, such as cytochromes and porphyrins, initially absorb of the trial, overall melanoma risk was reduced by 50% (0.50 [95%
UVA energy and then transfer it to either DNA (a type I photosensitized CI, 0.24–1.02]) and invasive melanoma risk was reduced by 73%
reaction), which results in CPD formation, or to molecular oxygen (0.27 [95% CI, 0.08–0.97]) in the daily sunscreen group compared
(a type II photosensitized reaction), generating ROS that ultimately with the discretionary use group.150 No clear benefit for BCC was
damage the DNA.143 In addition to DNA, oxidative stress also observed. Numerous cohort and case-control studies have examined
damages other cellular components, which can further contribute the relationship between sunscreen use and skin cancer, but results are
to carcinogenesis. conflicting and most have serious methodological limitations relating to
Another mechanism of UVR photocarcinogenesis may relate to measures of sun exposure, sunscreen use, and adjustment for important
the ability of UVA and UVB to induce persistent genomic instability confounders.151 For various reasons, the evidence that sunscreens prevent
through shortening or loss of telomeres as well as through a “bystander skin cancer may remain elusive.152 Nevertheless, studies have shown that
effect,” whereby cells not exposed to UVR are also damaged.142 This sunscreens are capable of preventing or delaying solar keratosis,153–155
may occur through the generation of increasingly damaged progeny a premalignant skin cancer lesion, and the efficacy of sunscreens
of UVR-irradiated cells. to prevent sunburns, an established risk factor for skin cancer, is
Important to note, UVR is a known immunosuppressant. The undisputed.156
ability of UVR to suppress certain parts of the immune system was There is much confusion among the public regarding the type,
first established by Kripke and Fisher in experiments in mice showing amount, and frequency of application when using sunscreens. The
that UVR prevented the rejection of highly immunogenic transplanted American Academy of Dermatology states that sunscreens should
UVR-induced skin tumors.144–146 In addition, UVR is used for its include broad-spectrum protection (protects against UVA and UVB
immunosuppressant effects as local therapy for various skin diseases, rays), should have an SPF of at least 30, and should be water resistant.157
such as psoriasis. Furthermore, it is well known that organ-transplant It further recommends that sunscreen should be applied 15 minutes
recipients who have the immune system suppressed to prevent organ before going outside and then every 2 hours or after swimming or
rejection are susceptible to skin cancer. UVB rays are thought to be sweating, and that approximately 1 ounce of sunscreen should be
primarily responsible for the immunosuppressive capability of UVR, used—enough to fill a shot glass—to cover all exposed skin.157
but UVA rays may also play a role.143 UVR can induce both systemic Although there is currently a great deal of research and controversy
and local immunosuppression, as well as influencing both acquired surrounding the ingredients of sunscreen and their safety, the risk-benefit
and innate immunity. UVB rays suppress the immune system by ratio indicates that sunscreens are appropriate to include as a skin
inducing immunosuppressive mediators, by damaging and triggering cancer preventive intervention.158
the premature migration of antigen-presenting cells required to stimulate
antigen-specific immune responses, by inducing the generation of Family History
suppressor cells, and by inhibiting the activation of effector and memory
T cells.142 Immunosuppression via UVA is less well understood but A family history of cancer among first-degree relatives can significantly
may relate to its ability to produce ROS and reactive nitrogen species increase one’s risk of cancer. Although one’s family history cannot be
as well as to its ability to penetrate more deeply into skin tissue, modified, the increased risk of cancer transmitted through a positive
reaching many more types of cells than UVB, including T cells, mast family history can, nevertheless, be mitigated through awareness and
cells, and granulocytes.142 adherence to cancer prevention and screening recommendations,
In sum, both UVA and UVB are able to induce carcinogenic because there are safe and effective interventions for those known to
changes to DNA and other cellular components, generally through be at high risk of cancer. Important to note, family history often
unique, although at times overlapping, mechanisms. UVR is both a determines when an individual begins cancer screenings, how frequently
348 Part I: Science and Clinical Oncology

Table 22.3  Recommended Actions for Skin Cancer Table 22.4  Goals to Support Skin Cancer
Prevention Prevention and Associated Strategies
FOR THE INDIVIDUAL GOAL 1: INCREASE OPPORTUNITIES FOR SUN PROTECTION
• Wear protective clothing. IN OUTDOOR SETTINGS
• Wear a hat and sunglasses. • Increase shade in outdoor recreational settings
• Seek shade. • Support sun-protective behaviors in outdoor settings
• Avoid times of peak sunlight. • Increase availability of sun protection in educational settings
• Use sunscreen. • Increase availability of sun protection for outdoor workers
• Avoid indoor tanning and sunbathing.
GOAL 2: PROVIDE INDIVIDUALS WITH THE INFORMATION
FOR THE CLINICIAN (IN PRIMARY CARE SETTING) THEY NEED TO MAKE INFORMED, HEALTHY CHOICES
• Counsel patients with fair skin aged 10–24 to minimize their ABOUT EXPOSURE TO ULTRAVIOLET RADIATION (UVR)
ultraviolet light exposure to reduce their risk of skin cancer. • Develop effective messages and interventions for specific
audiences
FOR THE COMMUNITY
• Support skin cancer prevention education in schools
IN OUTDOOR OCCUPATIONAL, RECREATIONAL OR TOURISM • Integrate sun safety into workplace health education and
SETTINGS promotion programs
• Implement interventions to promote sun-protective behaviors that • Partner with health care systems and providers to implement and
include educational approaches, modeling or demonstrating monitor use of recommended preventive services for provider
behaviors, environmental approaches (e.g., providing sun screen), counseling on skin cancer prevention
and policies to support sun protection strategies (e.g., requiring • Establish partnerships between public and private sectors to
hats and/or sun-protective clothing). disseminate effective messages about skin cancer prevention
• Enhance ongoing engagement of federal partners to advance our
IN EDUCATIONAL SETTINGS nation’s skin cancer prevention efforts
• Implement interventions in child care centers and in primary and
middle schools that promote sun-protective behaviors through GOAL 3: PROMOTE POLICIES THAT ADVANCE THE NATIONAL
educational interventions, supportive behavioral interventions, and GOAL OF PREVENTING SKIN CANCER
environmental and policy changes. • Support inclusion of sun protection in school policies, construction
of school facilities, and school curricula
THROUGHOUT COMMUNITY • Promote electronic reporting of reportable skin cancers, and
• Implement multicomponent community-wide interventions that encourage health care systems and providers to use such systems
include individual-directed strategies, mass media campaigns, and • Incorporate sun safety into workplace policies and safety trainings
environmental and policy changes across multiple settings. • Support shade planning in land-use development
FOR THE STATE GOAL 4: REDUCE HARMS FROM INDOOR TANNING
SUN PROTECTION POLICIES AND LEGISLATION • Monitor indoor tanning attitudes, beliefs, and behaviors in the US
• Adopt and support legislation that requires schools to allow population, especially among indoor tanners, youth, and parents
students to wear sun-protective clothing and sunscreen while on • Continue to develop, disseminate, and evaluate tailored messages
campus. to reduce indoor tanning among populations at high risk
• Support organizational policies that discourage indoor tanning by
EDUCATION AND AWARENESS adolescents and young adults
• Adopt and support legislation to mandate instruction on skin • Enforce existing indoor tanning laws and consider adopting
cancer prevention as part of the health education curriculum in additional restrictions
public schools. • Address the risks of indoor tanning with improved warning labels
and updated performance standards
INDOOR TANNING POLICIES AND LEGISLATION
• Adopt and support legislation that prohibits minors from using GOAL 5: STRENGTHEN RESEARCH, SURVEILLANCE,
tanning beds and requires monitoring of use of indoor tanning MONITORING, AND EVALUATION RELATED TO SKIN CANCER
devices; requires warnings to users about health risks associated PREVENTION
with indoor tanning; and enforces such regulations. • Enhance understanding of the burden of skin cancer and its
relationship with UVR
Recommendations are based on the 2014 Surgeon General’s Call to Action to • Evaluate the effect of interventions and policies on behavioral and
Prevent Skin Cancer report.130 health outcomes
• Build on behavioral research and surveillance related to UVR
exposure
• Quantify the prevalence of tanning in unsupervised locations (use
of tanning beds outside of tanning salons)
he or she is screened, and how that screening is performed. Genetic
testing may be warranted in patients with a strong positive family Goals and strategies are drawn from the 2014 Surgeon General’s Call to Action to
history of cancer. In the case of hereditary breast and ovarian cancer Prevent Skin Cancer report.130
syndrome, carriers of BRCA1/2 may be offered prophylactic surgery
as a risk-reducing intervention.159 Because family history is likely to
have potential consequences for multiple family members, families
are encouraged to take the time to collect and share a family health My Family Health Portrait tool (https://familyhistory.hhs.gov/FHH/
history, including the results of any cancer screening and/or genetic html/index.html). These tools are not cancer specific, but are applicable
testing that any family member has undergone. Such results may have to all noncommunicable chronic diseases.
potentially life-saving implications for other members. There are freely For more information related to family history in cancer, the reader
available tools online that enable this, such as the Surgeon General’s is referred to Chapter 13.
Lifestyle and Cancer Prevention  •  CHAPTER 22 349

MOLECULAR PREVENTION be targeted to prevent cancer development; (2) to develop effective,


safe, and tolerable preventive therapies; and (3) to promote the use
Drugs, vaccines, and natural agents represent different strategies for of these safe preventive therapies in at-risk individuals. The ultimate
molecular prevention, with the ultimate goal of inhibiting, reversing, goal of these efforts is to deliver effective, safe, and acceptable interven-
or delaying the onset of carcinogenesis. Advances in our understanding tions to prevent multiple forms of cancer. However, although these
of the mechanistic basis for carcinogenesis, with the identification of strategies have proven effective for preventing specific tumor subtypes,
molecules and signaling pathway alterations critical for tumorigenesis, they are often ineffective or poorly effective in prevention of other
has enabled the development of molecularly targeted therapies to tumor subtypes (e.g., selective estrogen receptor modulators (SERMs)
block tumor development. The novel drugs, signaling inhibitors, and and aromatase inhibitors (AIs) for the prevention of ER+ breast cancer).
vaccines that have been developed and tested as cancer preventive Thus, there is a clear need for the identification and development of
agents are discussed in this section. novel drugs that can more effectively prevent other cancer subtypes
Phase II and III clinical trials have demonstrated the effectiveness (e.g., ER− breast cancer) (Fig. 22.1). Alternative approaches for
of interventions that target oncogenic pathways within the cell for molecular cancer prevention currently under investigation include
the prevention of cancer. Several agents have been FDA approved for cyclooxygenase-2 (COX-2) inhibitors, retinoids, HER2 receptor kinase
treatment of precancerous lesions for cancer risk reduction (Table inhibitors, IGF inhibitors, kinase inhibitors, metformin, poly (ADP-
22.5), although they are often not used. Major challenges are (1) to ribose) polymerase (PARP) inhibitors, statins, and transcription factor
elucidate the critical molecular drivers of early tumorigenesis that can inhibitors, in addition to novel vaccine and inflammation- and
immunoprevention-based approaches.

Lung Cancer
Table 22.5  Agents Approved for the Treatment of
Precancerous Lesions or to Reduce Many phase II and III behavioral and cancer prevention clinical trials
Cancer Risk, 2017 have been conducted in lung cancer. Although nicotine replacement
therapy has been among the leading behavioral treatment strategies,
Disease Intervention preventive therapy using various agents, including vitamins, minerals,
Anal cancer HPV vaccine (Gardasil-9; for cancer caused by drugs, and signal transduction inhibitors, has been tested. Many agents
infection with HPV types 16, 18, 31, 33, 45, 52, have been tested; however, none have been shown to be useful as
and 58 and for precancerous or dysplastic cancer prevention drugs in phase III trials
lesions caused by infection with HPV types 6, 11, Numerous large-scale observational studies have provided results
16, 18, 31, 33, 45, 52, and 58) of patient use of a variety of agents and have helped support the
Bladder cancer Bacillus Calmette-Guérin development of clinical trials. Among these, the Singapore Chinese
(dysplasia) Valrubicin Health Study investigated the effects of the dietary carotenoids and
Breast cancer Tamoxifen risk of lung cancer.160 A total of 63,257 men and women aged 47 to
Raloxifene 75 years participated from 1993 to 1998, with 8 years of follow-up.
The results of this study suggested that dietary β-cryptoxanthin reduces
Cervical cancer HPV vaccine (Cervarix; for cancer caused by
lung cancer risk.
infection with HPV types 16 and 18)
HPV vaccine (Gardasil; for cancer caused by
Several clinical trials testing the effects of vitamins on lung cancer
infection with HPV types 16 and 18) were then conducted. The Alpha-Tocopherol, Beta-Carotene Cancer
HPV vaccine (Gardasil-9; for cancer caused by Prevention Study (ATBC) was a large randomized, placebo-controlled
infection with HPV types 16, 18, 31, 33, 45, 52, phase III cancer prevention trial in which 27,111 male smokers were
and 58 and for precancerous or dysplastic recruited and randomized to receive vitamin E (alpha-tocopherol),
lesions caused by infection with HPV types 6, 11, β-carotene, a combination of these two agents, or placebo.161,162 The
16, 18, 31, 33, 45, 52, and 58) results of this study showed that neither agent alone nor the combination
Colonic adenomas Celecoxiba of vitamin E and β-carotene reduced lung cancer incidence; however,
the study did show that there was a higher incidence of lung cancer
Esophageal cancer Photofrin plus photodynamic therapy (PTD)
in smokers who took β-carotene.163
(dysplasia)
Skin cancer (actinic 5-Aminolevulinic acid plus photodynamic β-Carotene
keratosis) therapy (PTD) Based on previous findings from observational and epidemiologic
Diclofenac sodium
studies that diets rich in β-carotene are associated with reduced cancer
Fluorouracil
risk, two large phase III clinical trials were conducted to test the
Imiquimod
Ingenol mebutate
efficacy of β-carotene for the prevention of lung cancer.
Masoprocol
Alpha-Tocopherol, Beta-Carotene Cancer Trial (ATBC)
Vulvar cancer HPV vaccine (Gardasil-9; for cancer caused by In 1994, the Alpha-Tocopherol, Beta-Carotene (ATBC) Cancer Preven-
infection with HPV types 16, 18, 31, 33, 45, 52,
tion Study Group published the initial findings of their randomized
and 58 and for precancerous or dysplastic
lesions caused by infection with HPV types 6, 11,
double-blind trial of over 29,000 male smokers.161,164 Study participants
16, 18, 31, 33, 45, 52, and 58) received α-tocopherol (20 mg/day), β-carotene (20 mg/day), both
α-tocopherol and β-carotene, or placebo for 5 to 8 years. α-Tocopherol
Vaginal cancer HPV vaccine (Gardasil-9; for cancer caused by
(vitamin E) was not associated with reduced incidence of lung cancer,
infection with HPV types 16, 18, 31, 33, 45, 52,
but was associated with decreased incidence of prostate cancer, whereas
and 58 and for precancerous or dysplastic
lesions caused by infection with HPV types 6, 11,
β-carotene was associated with an 18% higher incidence of lung cancer
16, 18, 31, 33, 45, 52, and 58) in smokers.163

a
US Food and Drug Administration (FDA) labeling voluntarily withdrawn by Pfizer, Beta-Carotene and Retinol Efficacy Trial (CARET)
February 2011. Soon after the ATBC results were released, findings from a second
HPV, Human papillomavirus. phase III lung cancer prevention clinical trial, the Beta-Carotene and
350 Part I: Science and Clinical Oncology

EGFR inhibitors
ErbB1/ErbB2 inhibitors

IGFR IGFR ErbB1 ErbB2 HER2 inhibitors

P P P P
Arachidonic acid
COX-1 EGCG
Cox1/2 & Cox2 inhibitors PI3K, Ras/Raf
COX-2
Prostaglandin G2
B-carotene / Vitamin A PGE2 MAPKs, JNKs, p38, AKT Kinase inhibitors
AMPK Resveratrol
Retinoids / Rexinoids
mTOR mTOR inhibitors
Vitamin D
p70S6K1 & AKT
Growth Statins
Metformin AMP signaling Mevalonate
AMPK
Aromatase
Aromatase inhibitors NR RXR
E2 XXRE
SERMs ER ER

β-catenin
Curcumin
NFκB Tumorigenesis
Growth control genes

PARP inhibitors PARP


Polyamines
Vaccines
ODC Immune response
Immunomodulators
DFMO T-cell
Cell death Checkpoint inhibitors

Figure 22.1  •  Potential strategies for targeting oncogenic pathways in the cell.

Retinol Efficacy Trial (CARET), studying β-carotene for the prevention in RR of lung cancer was no longer statistically significant (RR, 0.70
of lung cancer, were published.165,166 In this study, 18,314 men and [95% CI, 0.40–1.21]; P = .18).171
women at high risk for lung cancer (significant occupational exposure
to asbestos or extensive history of smoking) were randomized to receive Selenium and Vitamin E Cancer Prevention Trial (SELECT)
β-carotene (30 mg/day), retinol (vitamin A, 25,000 IU), combined The Selenium and Vitamin E Cancer Prevention Trial (SELECT) was
treatment with both drugs, or placebo. This study was stopped 21 a large-scale phase III clinical trial testing the efficacy of long-term
months early because of similar findings to the ATBC trial. CARET treatment with vitamin E, selenium, or both for the prevention of
participants who received combination treatment with β-carotene and prostate cancer.172 Incidence of lung, colorectal, and other cancers
retinol developed 28% more lung cancers and had higher all-cause served as secondary end points. A total of 35,533 men were randomized
mortality, cardiovascular mortality, and mortality from lung cancer.167 to receive vitamin E (400 IU/day of all-rac-α-tocopheryl acetate),
These studies of vitamin E and β-carotene demonstrated that vitamins selenium (200 µg/day from l-selenomethionine), both, or placebo.
are not harmless, and they should be used carefully. After 5 years of treatment, although vitamin E was found to increase
risk of prostate cancer (13%; 99% CI, 0.95–1.35), treatment with
Selenium selenium was not associated with decreased incidence of lung, colorectal,
Nutritional Prevention Cancer Trial (NPC) or other cancers. An extended follow-up study through July 2011
After results from geographic and environmental studies suggested showed significantly increased risk of prostate cancer in patients treated
that increased selenium is associated with decreased cancer incidence with vitamin E (17%; 99% CI, 1.004–1.36), selenium (9%; 99%
and mortality for multiple cancer types,168,169 the Nutritional Prevention CI, 0.93–1.27), or vitamin E plus selenium (5%; 99% CI, 0.89–1.22).
Cancer Trial (NPC) was conducted to assess the effects of selenium No significant change was noted from their previously reported findings
versus placebo on risk of skin cancer in patients with a history of for the secondary end points.173
BCC or SCC.170 Secondary end points included diagnosis of other
cancers (not BCC or SCC), cardiovascular disease, and other medical Selenium Supplementation in Patients With Resected Stage I
conditions. A total of 1312 participants with a history of BCC or Non–Small Cell Lung Cancer: ECOG 5597
SCC were randomized to receive selenium (200 µg) or placebo daily. The Selenium Supplementation in Patients With Resected Stage I
Initial results based on 4.5 years of follow-up showed that patients Non–Small Cell Lung Cancer (Eastern Cooperative Oncology Group
treated with selenium had significantly lower incidence of lung cancer [ECOG] 5597) trial accrued and randomized 1561 participants with
(RR, 0.54 [95% CI, 0.30–0.98]; P = .04) than those treated with non–small cell lung cancer (NSCLC) to receive selenized yeast (200 µg/
placebo. However, after an extended follow-up of 7.9 years, the decrease day) or placebo for 48 months.174 Although the interim results suggested
Lifestyle and Cancer Prevention  •  CHAPTER 22 351

increased risk of second primary lung tumors in the selenium arm, specimens was identified in participants treated with enzastaurin
final results from this study confirmed the results of the SELECT compared with those treated with placebo.
study, finding selenium to be safe, but not beneficial over placebo,
for both disease-free survival and the prevention of second primary myo-Inositol
tumors in NSCLC patients. Based on preclinical and early clinical studies that showed a greater
The results of these trials testing β-carotene and selenium failed than 40% decrease in preexisting dysplastic lesions in smokers with
to demonstrate a preventive benefit associated with these compounds. bronchial dysplasia following treatment with myo-inositol,180 Lam and
Moreover, they identified the unexpected potential for significant health colleagues published results from the first phase IIb trial testing myo-
risks following treatment with these compounds. inositol for chemoprevention of lung cancer in smokers with bronchial
dysplasia.181 A total of 74 participants were randomized to receive
Budesonide myo-inositol (9 g/day for 2 weeks, followed by 9 g twice daily for 6
The effects of inhaled budesonide (800 µg twice daily) compared months) or placebo. Treatment with myo-inositol was not found to
with placebo for 1 year was the focus of a phase IIb trial that included have a statistically significant effect on bronchial dysplasia, with
202 current and former smokers with computed tomography (CT) complete response rates of 26.3% (myo-inositol) and 13.9% (placebo)
screen-detected lung nodules.175 This study was nested within the and progressive disease rates of 47.4% (myo-inositol) and 33% (placebo)
large phase III Continuous Observation of Smoking Subjects (P = .76). However, these results, combined with the findings of PI3K
(COSMOS) lung cancer screening trial, which was composed of 5203 activation, significant reductions in bronchoalveolar lavage (BAL) fluid,
participants. Budesonide was found to be well tolerated but not effective IL-6 levels, and statistically insignificant reductions in Ki76 levels,
in reducing lung nodule size compared with placebo in the per-person suggest that future studies of myo-inositol for the chemoprevention
analysis, which was the primary end point (reduction rate: budesonide of lung cancer in a more defined cohort may identify a preventive
2%, placebo 1%). However, budesonide was found to produce sig- benefit.
nificant regression of existing nonsolid and partially solid nodules
(lung nodules most likely representing adenocarcinoma precursors). Retinoids
Long-term follow-up of 5 years showed a significant reduction in Preclinical and epidemiologic studies using retinoids as preventive
nonsolid lung nodule size after treatment with budesonide compared and therapeutic agents for head and neck cancer provided the rationale
with placebo.176 The mean maximum diameter of nonsolid nodules to conduct clinical trials using retinoids in patients with head and
in the budesonide arm dropped from 5.03 mm at baseline to 2.61 mm neck cancer. Results from these studies demonstrated the cancer preven-
after 5 years. Notably, budesonide was not found to reduce the size tive effects of retinoids in this setting. Subsequently, these findings,
of solid nodules. In addition, neither the number of new lesions nor combined with preclinical and early clinical data from use of isotretinoin
the number of lung cancers differed in the patients treated with (13-cis–retinoic acid) in lung cancer, supported the development of
budesonide or placebo. Despite the lack of effect on lung cancer phase III trials for the prevention of lung cancer.
incidence in this phase II trial (which was not powered to detect a
difference in lung cancer incidence), budesonide remains a promising Lung Intergroup Trial (LIT)
agent for lung cancer prevention. The multicenter Lung Intergroup Trial (LIT) randomized 1166 patients
with stage I NSCLC to receive the retinoid isotretinoin (30 mg/day)
Nonsteroidal Antiinflammatory Drugs or placebo for 3 years.182 Patients underwent a 3.5-year follow-up
Sulindac.  Because of the cardiovascular toxicity associated with period, at which time the results showed no benefit in the overall rate
selective COX-2 inhibitors, Limburg and colleagues conducted a study of second primary tumors (primary end point) or in recurrence or
of the nonselective COX-2 inhibitor sulindac for the prevention of mortality (secondary end points). In addition, this study identified a
lung cancer.177 They randomized 61 participants to receive sulindac potential harmful effect of isotretinoin based on smoking status
(150 mg BID) or placebo for 6 months. This study did not find any (non–statistically significant increased recurrence and statistically
statically significant benefit from sulindac treatment on either bronchial significant increased mortality in current smokers compared with never
dysplasia (regression: sulindac arm, 58%; placebo arm, 56%) or mucosal smokers). Updated results were published in 2010 with an extended
proliferation (measured by Ki67 expression: sulindac arm, 30 versus 6.2-year follow-up, which confirmed increased overall and cancer-related
10; placebo arm, 30 versus 5; P = .92). mortality, as well as cardiovascular disease in current smokers treated
with isotretinoin.183 Conversely, never and former smokers exhibited
Iloprost a slight decrease in both overall and cancer-related deaths.
A phase II study of iloprost was conducted to determine its efficacy in
reversing premalignant histologic changes in the bronchial epithelium 13-cis–retinoic acid with or without α-tocopherol
in current and former smokers.178 A total of 152 patients at high Kelly and colleagues randomized 75 subjects at high risk for lung
risk of lung cancer were treated with iloprost (a 50 to 150 mg/ cancer to receive 13-cis–retinoic acid (50 mg/day by mouth [PO])
day dose escalation for 2 months, followed by 150 mg/day for 4 alone or in combination with α-tocopherol (800 mg/day PO) for 12
months) or placebo. Former smokers treated with iloprost versus months to determine its effect on histologic progression.184 Their results
placebo demonstrated a significant improvement in endobronchial were consistent with those of other trials examining the preventive
histologic findings (58.6% versus 28.6%, respectively), whereas current efficacy of 13-cis–retinoic acid in the lung cancer setting, with risk
smokers did not. These results show that iloprost is a promising drug of treatment failure being 50% in the 13-cis–retinoic acid and the
for lung cancer prevention. A future phase III trial will be required 13-cis–retinoic acid plus α-tocopherol treatment arms and 55.6% in the
to conclusively demonstrate the effectiveness of this promising observation arm.
intervention.
Other Agents
Serine/Threonine Kinase Inhibitors Zileuton, a 5-lipoxygenase inhibitor, was studied as a chemopreventive
Based on positive preclinical findings, Gray and colleagues studied agent for lung cancer by Kucuk and colleagues in a phase II study of
the effects of enzastaurin, which inhibits protein kinase C–β activation, 38 patients with bronchial dysplasia (NCT00056004). The trial, which
as a chemopreventive agent for lung cancer.179 In this study, 40 former is investigating the effects of 6 months of treatment with zileuton,
smokers were randomized to receive enzastaurin (500 mg/day) or has been completed, but no results have been released to date. Results
placebo for 6 months. Although enzastaurin treatment proved tolerable, from a second phase I/II study of short-term (6 days) treatment with
no significant reduction in Ki67 labeling index in bronchial biopsy zileuton (1200 mg twice per day) with or without celecoxib (200 mg
352 Part I: Science and Clinical Oncology

twice per day) are also anticipated in the near future (NCT01021215). the planned treatment because of significant toxicities (headache, dry
Preliminary data from this study suggest preventive efficacy associated or cracked skin, and skin rash).
with zileuton, which may be increased when combined with celecoxib. Because toxicity was a major problem with this first trial, sub-
Other novel drugs have been shown to be effective for the treatment sequent trials tested lower doses of isotretinoin. A phase III trial
of lung cancer. These include PARP inhibitors185 and immune testing the efficacy of low-dose isotretinoin (13-cis–retinoic acid;
checkpoint inhibitors.186 With continued development of effective 1–2 mg/kg/day) for prevention of second primary tumor incidence
and safe inhibitors of these novel targets, it will be possible to test and survival in individuals previously diagnosed with early-stage
them in cancer prevention trials. Such agents offer great promise for head and neck squamous cell cancer (HNSCC) was conducted by
prevention of this common cancer. Because lung cancer is the most Hong and colleagues.200 This study showed that isotretinoin did
common cause of cancer-related death, there are continued efforts to not reduce second primary cancer incidence (HR, 1.06 [95% CI,
develop safe and effective strategies for lung cancer prevention. 0.83–1.35]) or mortality (HR, 1.03 [95% CI, 0.81–1.32]) as compared
Decreased incidence of lung cancer in the future will depend on with placebo.
primary prevention strategies (e.g., smoking cessation or avoidance More recently, Hong and colleagues genotyped 9465 single-
and reduced exposure to toxins) and nontoxic preventive therapy. nucleotide polymorphisms (SNPs) in 450 of the participants of the
trial to determine any association between these SNPs and second
Head and Neck Cancers primary cancers.201 They found that 13 loci correlated with significantly
increased risk of second primary tumor incidence or recurrence
Smoking, alcohol, and HPV are all associated with increased risk for (3.33-fold increased risk [95% CI, 1.67–6.67]). In addition, these
head and neck cancers. Thus, as with lung cancer, nicotine replacement investigators identified several loci that were associated with successful
therapy with bupropion or varenicline, smokeless tobacco cessation, and cancer prevention: an SNP in the retinoid X receptor (rs3118570 in
vaccines to induce nicotine binding in the blood have been among the RXRA), an SNP in the CDC25C gene (s6596428), and an SNP in
leading behavioral treatment strategies for the prevention of head and the JAK2 gene (rs1887427). These results suggest that it may be
neck cancer.187,188 In addition, strategies to reduce alcohol use and con- possible to identify host characteristics that predict successful cancer
traction of HPV, including alcohol awareness and intervention programs, prevention with isotretinoin.
also represent key primary interventions, and HPV and Epstein-Barr
virus (EBV) vaccines will be incorporated as immunologic strategies in Eastern Cooperative Oncology Group study
future years.189,190 A study conducted by ECOG examined low-dose 13-cis–retinoic acid
A variety of agents have been investigated for the prevention of for the prevention of second primary tumors in patients with head
head and neck cancer. Types of drugs used in this setting include and neck cancer.202 A total of 189 participants were randomized to
retinoids, peroxisome proliferator-activated receptor (PPAR) agonists, receive 13-cis–retinoic acid (7.5–10 mg) or placebo. After a 3-year
and protease inhibitors (Table 22.6). minimal follow-up, no significant reduction in incidence was observed
between the experimental and control groups.
Retinoids
Clinical trials for the prevention of head and neck cancers have largely Peroxisome Proliferator-Activated Receptor Agonists
investigated the use of retinoids, but to date toxicity of these drugs The PPAR family includes PPAR-α, PPAR-β, and PPAR-γ. PPAR-γ
has impeded their translation to the clinical setting for cancer preven- has been shown to regulate expression of metabolic pathway genes
tion. Consequently, more recent trials have focused on low-dose by heterodimerizing with retinoic acid receptors,203 and to regulate
administration of retinoids or the testing of novel less toxic synthetic differentiation and apoptosis.204 PPAR ligands include pioglitazone,
retinoids for head and neck cancer prevention. rosiglitazone,and troglitazone, which were developed for patients with
type 2 diabetes mellitus.205 Both rosiglitazone and troglitazone are
Retinoid Head and Neck Second Primary (HNSP) trial not currently in use owing to cardiovascular events and hepatotoxicity,
The first demonstration that retinoids could prevent cancer in humans respectively, but pioglitazone is currently used for some individuals
was from a clinical trial conducted by Hong and colleagues testing with type 2 diabetes mellitus.205,206
13-cis–retinoic acid (isotretinoin) for the prevention of second primary A multisite phase IIB clinical trial (NCT00951379) conducted
cancer in individuals with previous squamous cell carcinoma of the through the NCI chemoprevention consortium investigated the effects
head and neck.191 The results of this clinical trial showed that 13-cis– of pioglitazone as a preventive therapy for patients with oral prema-
retinoic acid reduced the incidence of second primary head and neck lignant lesions exhibiting dysplasia or hyperplasia. This trial has
cancers by 83%; however, few of the patients were able to complete completed accrual, and biomarker data are being analyzed. The 52

Table 22.6  Head and Neck Chemoprevention Trials With Outcomes


Phase Patients Agent (Dose) Results Reference
IIB 44 Isotretinoin (13-cis–retinoic acid) (1–2 mg/kg/day) Positive Hong et al191
IIB 103 13-cis–Retinoic acid (50-100 mg/m2/day) Positive Hong et al192
IIB 65 Vitamin A (200,000 IU/wk) Positive Stich et al193
IIB 103 β-Carotene (100,000 IU/wk) + retinol (180 mg/wk) Positive Stich et al194
IIB 31 Retinamide (40 mg/day) Positive Han et al195
IIB 70 Isotretinoin (0.5 mg/day) Positive Lippman et al196
IIB 153 Fenretinide (200 mg/day) Positive Chiesa et al197
IIB 675 β-Carotene (40 mg/day) + retinol (100,000 IU/wk) + vitamin E (8 oz/wk) Positive Zaridze et al198
III 189 cis-Retinoic acid (10 mg/day) Negative Pinto et al199
III 1384 cis-Retinoic acid (30 mg/day) Negative Khuri et al200
Lifestyle and Cancer Prevention  •  CHAPTER 22 353

nondiabetic participants were randomized to receive pioglitazone risk have been assessed, and LOH at specific chromosomal sites
(45 mg/day) or placebo for 24 weeks. Publication of the results from (especially 3p14 and 9p21) in oral premalignant lesions has been
this trial are anticipated in the near future. A second phase II trial, identified as one of the most robust prognostic markers.209 Building
led by Keith at the VA Eastern Colorado Health Care System, has on these results, the first molecularly based chemoprevention study
also completed accrual (NCT00780234). This study is examining has been completed: the EPOC trial.207 In this study, patients with
pioglitazone (30 mg/day) versus placebo for 6 months in 391 non- oral premalignant lesions were tested for LOH, and only those harbor-
diabetic patients at risk for lung cancer. ing high-risk LOH profiles were randomized to receive the EGFR
inhibitor erlotinib versus placebo for 12 months. Although this study
Celecoxib did not demonstrate a reduction in oral CFS in the erlotinib group,
Celecoxib has been investigated in 2 phase II prevention trials for it confirmed, for the first time in a prospective setting within the
head and neck cancers. Of these, Engstrom and colleagues at Fox context of a clinical trial, the role of LOH in predicting cancer risk,
Chase are studying the efficacy of celecoxib in preventing head and initiating an era of personalized prevention. The study also allowed
neck cancer in individuals with oral leukoplakia (NCT00101335). for a rich, clinically annotated biobank to be assembled that could be
A second study of celecoxib in patients with oral leukoplakia and/ interrogated for identification of additional targets for cancer preven-
or head and neck dysplasia is being conducted at Dana Farber by tion. Subsequently, using samples from the EPOC study, William and
Wirth and colleagues (NCT00052611). Although the Fox Chase study colleagues demonstrated that oral premalignant lesions may express
has been completed, results have not yet been released for either of PD-L1, and that PD-L1 expression is higher in LOH-positive lesions
these studies. and independently associated with inferior oral CFS.210 Based on these
findings and approval of PD-1 inhibitors in advanced HNSCC, the
Erlotinib MD Anderson group launched the first PD-1–based Immune Preven-
The epidermal growth factor receptor (EGFR) inhibitor erlotinib was tion of Oral Cancer (IPOC) randomized clinical study, investigating
the focus of the Erlotinib Prevention of Oral Cancer (EPOC) study. the role of pembrolizumab versus observation to improve oral CFS
This phase III clinical trial randomized 150 individuals with high-risk in patients with high-risk, LOH-positive oral premalignant lesions
oral premalignant lesions (defined as loss of heterozygosity [LOH]) (NCT02882282).
to receive erlotinib (150 mg/day) or placebo for 12 months.207 The
median follow-up time was 35 months. Erlotinib did not improve Esophageal Cancers
oral cancer-free survival (CFS; HR, 1.27 [95% CI, 0.68–2.38]).
However, this study demonstrated that LOH is a marker of oral cancer ESCC is the most common type of esophageal carcinoma throughout
risk, with LOH-positive patients having a significantly lower 3-year the majority of the developing world. Although ESCC was the leading
oral CFS compared with LOH-negative patients (HR, 2.19 [95% CI, type of esophageal cancer in the United States in the 1970s, 50% or
1.25–3.83]). LOH positivity was also found to correlate with increased more of esophageal cancer diagnoses in the United States, as well as
EGFR copy number. in a number of other countries, are esophageal adenocarcinoma (EAC).
ESCC and EAC have disparate risk factors, histopathologic charac-
Bowman-Birk Inhibitor Concentrate teristics, and patterns of occurrence, but treatment strategies for both
Following positive results from a phase IIa trial, Armstrong and col- remain fairly similar.
leagues conducted a phase IIb clinical trial of the serine protease
inhibitor Bowman-Birk inhibitor concentrate (BBIC) in patients with Esophageal Squamous Cell Carcinoma
oral leukoplakia.208 A total of 89 subjects received 600 chymotrypsin Although numerous studies of individuals at high risk of ESCC have
inhibitor units of BBIC or placebo for 6 months. The experimental investigated the efficacy of dietary interventions (e.g., herbs, minerals,
and control groups did not demonstrate a significant difference in and vitamins) and have had promising results, these approaches have
clinical response. failed to translate to the clinic.198,211–216 Because the incidence of ESCC
is particularly high in central China, and individuals in that region
Other Agents have a chronically low intake of a number of micronutrients, a series
Other phase II trials are investigating additional strategies for the of these trials have been conducted there. One phase III trial recruited
prevention of head and neck cancer. The kinase inhibitor vandetanib 29,584 40- to 69-year-old men and women who were randomized
is the focus of a study currently recruiting participants that is investigat- to receive one of four nutrient combinations over a 5-year period:
ing its potential as a chemopreventive agent for premalignant lesions retinol plus zinc; riboflavin plus niacin; vitamin C plus molybdenum;
of the head and neck (NCT01414426). Currently underway is a and β-carotene plus vitamin E plus selenium.216 Participants were
phase II trial of metformin, originally designed for diabetic patients, administered doses that were equal to or twice the recommended daily
for preventing oral cancer in individuals with oral premalignant lesion(s) allowance in the United States. Whereas 32% of the 2127 deaths that
(NCT02581137). Accrual to this trial is ongoing, with an estimated occurred during intervention were due to esophageal or stomach cancer,
primary completion date of February 2019. Another study still recruit- mortality was significantly lower among participants receiving β-carotene
ing participants is investigating the efficacy of GL-0817 vaccination plus vitamin E plus selenium (RR, 0.91 [95% CI, 0.84–0.99]).
(10 doses) with cyclophosphamide administered 1 day before the first Moreover, this group exhibited lower incidence of both overall cancer
three vaccinations. GL-0817 is a vaccine against melanoma-associated (RR, 0.87 [95% CI, 0.75–1.00]) and stomach cancer (RR, 0.79 [95%
antigen A3 (MAGEA3). This study started in March 2017 and is CI, 0.64–0.99]). The other three treatment groups did not show
expected to be completed by March 2020. Among the dietary strategies decreased mortality.
being used in studies to prevent recurrence are lyophilized black In another study conducted in this region, Limburg and colleagues
raspberries (NCT01504932), strawberry polyphenols (NCT01514552), examined the effects of selenomethionine (200 µg/day) with or without
and soy isoflavones (NCT020007200). celecoxib (200 mg/day) in 238 individuals with mild to moderate
Despite decades of investigation of chemopreventive agents for esophageal squamous dysplasia.217 Esophageal squamous carcinogenesis
patients with oral premalignant lesions, there is currently no standard- was not decreased after 10 months of intervention with either sele-
of-care drug that has shown consistent and durable benefits in this nomethionine or celecoxib. However, the 115 individuals with mild
setting. One of the greatest challenges of developing a chemopreven- esophageal squamous dysplasia at baseline exhibited decreased dysplasia
tion approach for oral premalignant lesions is the identification of with selenomethionine (P = .02). This protective effect was not observed
a high-risk group that would be expected to benefit the most from with the 123 participants with moderate esophageal squamous dysplasia
candidate agents. Toward this end, molecular markers of oral cancer (P = 1.00).
354 Part I: Science and Clinical Oncology

Unfortunately, translation of these vitamin and mineral combinations versus esomeprazole plus aspirin.239,240 This study has recruited 2500
into clinical use for the prevention of esophageal cancer in high-risk participants to receive esomeprazole (20 mg/day), esomeprazole
populations with low micronutrient intake has not occurred, in part (80 mg/day), esomeprazole (20 mg/day) plus aspirin (300 mg/
because of the difficulty in acquiring and interpreting the data for day), or esomeprazole (80 mg/day) plus aspirin (300 mg/day).241
these strategies, and in identifying those individuals best suited to The results from this trial should provide more information about
receive each treatment. the cancer prevention efficacy of both esomeprazole and aspirin in
this setting.
Esophageal Adenocarcinoma
Preventive strategies for EAC have investigated targeting COX-2, Colorectal Cancer
cytosolic phospholipase, MCM2, and NF-κB via antiinflammatory
drugs, antioxidants, and acid suppression agents because of the associa- Among the strategies that have been investigated for the prevention
tion of this disease with both gastroesophageal reflux disease (GERD) of CRC and have been found to be of some benefit are calcium,
and chronic inflammation.218 eicosapentaenoic acid (EPA), HRT, NSAIDs, and selenium.
Barrett esophagus is a common complication associated with GERD
and results in the replacement of normal squamous esophageal epi- Nonsteroidal Antiinflammatory Drugs
thelium with metaplastic intestinal epithelium.219,220 Approximately NSAIDs have been the primary focus of a large number of observational,
10% to 15% of individuals with GERD develop Barrett esophagus, preclinical, and clinical CRC prevention studies, which have investigated
which then places them at a 30- to 125-fold increased risk of adeno- the use of aspirin, COX-2 inhibitors, and sulindac.242 These studies
carcinoma, which is preceded by dysplasia.221 Although the symptoms have examined both adenoma recurrence and CRC incidence and/or
of acid reflux can be relieved by antireflux therapy, including proton mortality.
pump inhibitors (e.g., lansoprazole),222 antireflux surgery, and endo-
scopic laser ablation,223,224 the effectiveness of these strategies for Selenium
promoting regression of Barrett esophagus and preventing the develop- Epidemiologic and clinical studies have been unable to clearly define
ment of adenocarcinoma remains unclear.225,226 a preventive efficacy of selenium for CRC.243–246
Nutritional prevention of cancer trial (NPCT).  The Nutritional
Nonsteroidal Antiinflammatory Drugs Prevention of Cancer Trial (NPCT) randomized 1312 patients with
Preclinical and early-phase clinical studies suggest that nonsteroidal a history of BCC or SCC of the skin between 1983 and 1991.170
antiinflammatory drugs (NSAIDs) may be effective in preventing Randomization included two arms: selenium (200 µg/day) and placebo.
EAC by reducing proliferation and/or inducing apoptosis.227,228 Although selenium was not effective in decreasing incidence of basal or
Additional findings based on observational studies suggest a potential squamous cell skin cancer, it was associated with decreased incidence of
30% to 40% reduction in risk of EAC associated with NSAID preven- some secondary end points, including incidence of lung cancer, prostate
tive therapy.229,230 cancer, and CRC. Subsequent follow-up results in 2002 demonstrated
Phase II and one phase III clinical trials have examined the efficacy decreased incidence only of total and prostate cancer incidence, and
of aspirin for prevention of esophageal cancer in patients with Barrett not of CRC incidence (HR, 0.46 [95% CI, 0.21–1.02]).247
esophagus. The Chemoprevention for Barrett’s Esophagus Trial (CBET) Selenium and vitamin E cancer prevention trial (SELECT).  SELECT
accrued 222 patients with low- or high-grade Barrett esophagus with was a phase III cancer prevention clinical trial investigating the effects of
dysplasia.231 Participants received celecoxib (200 mg) or placebo twice selenium and vitamin E (α-tocopherol) on the development of prostate
daily for 48 weeks. Celecoxib did not appear to reduce risk of disease cancer. A substudy of this clinical trial (the Adenomatous Colorectal
progression or cancer. However, analysis of the subset of 58 CBET Polyp [APC] study) investigated the effect of these supplements on
participants with usable quantitative endoscopy area measurements the incidence of colorectal adenomas.173 After a 7-year follow-up,
demonstrated that patients in the celecoxib group did exhibit a sig- neither drug (nor their combination) was found to significantly affect
nificant treatment effect measured by quantitative endoscopy before colorectal adenoma occurrence.172 The most recent results from an
and after treatment.232 ancillary study of 6546 participants of the SELECT trial, released
A second phase II study investigated the preventive effects of aspirin in 2017, confirmed these findings, with a 0.96 RR for adenoma
on disease recurrence in 114 patients with Barrett esophagus with no incidence in participants in the selenium group (95% CI, 0.90–1.02;
or low-grade dysplasia after successful elimination of Barrett esophagus P = .194), and 1.03 RR in participants in the vitamin E group (95%
with radiofrequency ablation. This study, led by Limburg at the Mayo CI, 0.96–1.10; P = .38) compared with placebo.248 In 2014, Vinceti
Clinic published results in 2012.233 Participants received short-term and colleagues published their findings of an analysis of data from
(28 days) self-administered treatment with esomeprazole (40 mg twice multiple electronic databases regarding selenium exposure and cancer
daily) plus either placebo (once per day), aspirin (81 mg/day), or risk and the preventive efficacy of selenium for the prevention of
aspirin (325 mg/day). The group treated with esomeprazole plus cancer.249 Their analysis of 55 observational studies did not show
325 mg of aspirin had significantly lower tissue concentrations of a beneficial effect of selenium on cancer risk.250 Results from
PGE2, which appear to be inhibited by NSAIDs.229,234–238 This reduction four additional ancillary studies of the SELECT trial were con-
of PGE2 was not observed in the other two treatment groups. ducted, examining the effect of selenium on preventing Alzheimer
Another phase II trial, led by Bresalier at the University of Texas disease (PREADVISE; NCT00780689), macular degeneration
MD Anderson Cancer Center, is currently recruiting patients with (SEE; NCT00784225), loss of lung function (NCT0063453), and
Barrett esophagus with or without dysplasia who have undergone colorectal polyps (ACP; NCT00706121).248,251
successful elimination of Barrett esophagus via radiofrequency ablation Women’s Health Initiative.  The Women’s Health Initiative (WHI)
(NCT02521285). This multiinstitutional study will investigate the was an observational study within which a nested study of the effects
effect aspirin has on preventing recurrence of Barrett esophagus. A of selenium on risk of CRC was conducted.252 No association was
total of 118 participants will be randomized to receive aspirin (orally found between treatment with selenium and risk of CRC.
once per day) or placebo for 12 months. This study is expected to be Selenium and Celecoxib (Sel/Cel) Trial. This Selenium and
completed in April 2019. Celecoxib (Sel/Cel) Trial, a phase III randomized clinical trial, examined
Jankowski and colleagues at Queen Mary University of London the preventive effects of single and combined treatment with selenized
are conducting the phase III Aspirin Esomeprazole Chemopreven- yeast and celecoxib on colorectal adenoma prevention.253 Following
tion Trial (AspECT), the largest randomized controlled clinical trial reports of toxicity associated with celecoxib by Arber, Bertagnolli, and
to study the long-term chemopreventive efficacy of esomeprazole Solomon,254–256 treatment with celecoxib was prematurely terminated.253
Lifestyle and Cancer Prevention  •  CHAPTER 22 355

Results from this study were published in 2016 and demonstrated CRC Risk study (ASPIRED; NCT02394769).270 This study will use
no overall preventive benefit associated with selenium versus placebo. liquid chromatography–mass spectrometry to assess changes in urinary
The researchers did observe an 18% reduction in adenoma recurrence prostaglandin metabolites to determine the mechanisms aspirin exploits
following treatment with selenium versus placebo in participants to reduce CRC risk.
with advanced adenomas at baseline (P = .01). However, as with Several meta-analyses have been published in recent years, including
Vinceti249 and Stranges,250 increased risk of type 2 diabetes occurred a strategic review conducted by the USPSTF, with findings published
in the selenium treatment group (HR, 1.25 [95% CI, 0.75–2.11]; in 2015.271 This analysis identified a 10-year reduction in risk of CRC
P = .41), and a statistically significant increase was found in older incidence of 20% to 24% after treatment with aspirin (≥75 mg/day)
participants (RR, 2.21 [95% CI, 1.04–4.67]; P = .03).257 Additional versus placebo. In addition, these findings suggest that aspirin may
randomized clinical trials testing selenium are currently being con- be associated with a 33% reduction in risk of CRC mortality within
ducted by Lance (phase III; NCT00078897) and Benya (phase I; 10 years of initiation of treatment versus placebo. A second systematic
NCT01211561). review and meta-analysis, by Lotrionte and colleagues, identified a
significant preventive effect of aspirin (100 mg/day) on cancer incidence
Aspirin and death; this dose was better tolerated than higher doses.272 Finally,
Initial results from both the Women’s Health Study258 and the Physi- Zhao and colleagues conducted a meta-analysis of randomized trials
cian’s Health Study, two observational studies, suggested that aspirin that once again confirmed the preventive effects of aspirin on advanced
does not reduce risk of CRC. Although long-term follow-up reported adenoma recurrence for 1 year (RR, 0.68 [95% CI, 0.49–0.95]; P =
for the Physician’s Health Study supported its earlier findings,259 .582), and on recurrence of any adenoma for longer than 1 year (RR,
the most recent results from the Women’s Health Initiative Study 0.84 [95% CI, 0.72–0.98]; P = .484).273 All these studies provide
reported a significant 42% posttrial reduction in risk of CRC in very strong evidence that aspirin use, particularly for long durations
healthy women treated with aspirin versus placebo (HR, 0.58 [95% (of more than 5 or 10 years) significantly reduces the incidence of
CI, 0.42–0.80]; P < .001), but increased risk of both GI bleeding and CRC. The major questions in this field currently are (1) what is the
peptic ulcers.260 optimal (i.e., effective yet safest) dose of aspirin, and (2) how long a
Two Colorectal Adenoma/Carcinoma Prevention Program (CAPP) duration is necessary for effective cancer prevention. These questions
studies found preventive efficacy associated with aspirin (600 mg/ are currently being addressed in the CAPP3 trial in Lynch syndrome
day), but not resistant starch (30 g/day), compared with placebo for patients.
the prevention of CRC. CAPP1 examined aspirin and resistant starch
in 206 adolescents (10–21 years of age) with familial adenomatous Cyclooxygenase-2 inhibitors
polyposis (FAP), and found that aspirin reduced polyp load (and size) FDA approval of celecoxib in FAP patients was based on the initial
compared with no aspirin, whereas resistant starch did not.261 Subse- results of a study by Steinbach and colleagues, which demonstrated
quently, the CAPP2 study investigated aspirin and resistant starch in that patients with FAP treated with celecoxib (400 mg twice per day)
861 Lynch syndrome carriers. The initial results were reported in exhibited 28% fewer colorectal polyps (P = .003) and a 31% reduced
2008 after 4 years of follow-up and showed no decrease in the number polyp burden (P = .001) versus patients treated with placebo.274
of advanced adenomas or colorectal incidence with use of either aspirin Subsequent results showed that 6 months of treatment with celecoxib
or resistant starch.262 However, a subsequent report in 2011 after 55.7 (400 mg twice per day) was also associated with significantly less
months of follow-up demonstrated that 25 months of treatment with duodenal polyposis compared with placebo.275 Shortly thereafter, both
aspirin was associated with a reduction in CRC incidence (incidence the Adenoma Prevention With Celecoxib255 and Prevention of Colorectal
rate of 0.37 [95% CI, 0.18–0.78]; P = .008) compared with placebo, Sporadic Adenomatous Polyps (PreSAP)254 trials demonstrated a
with no difference in adverse events between these two treatment significant reduction in colorectal adenoma occurrence after treatment
groups.263 The CAPP2 results provide strong rationale to treat patients with celecoxib (400 mg/day) compared with placebo. Unfortunately
with hereditary nonpolyposis colorectal cancer with aspirin for colon both the initial and follow-up results of the Adenoma Prevention
cancer prevention. A subsequent trial, the CAPP3 trial, is now being With Celecoxib and PreSAP studies identified a significant increase
conducted to test several doses of aspirin (100 mg, 300 mg, 600 mg in risk of cardiovascular events associated with celecoxib, particularly
for 2 years, then 75 mg, 300 mg, or 600 mg for 3 more years) to for individuals who have a high baseline risk for cardiovascular
determine the most effective and safest dose. disease.256,276 These results have greatly limited further development
Several trials have studied the use of aspirin for preventing sporadic of COX-2 inhibitors for CRC prevention. However, recent results
adenoma recurrence in patients with a history of adenomas or CRC. from a phase I trial conducted by Lynch and colleagues show that
Studies by Logan,264 Benamouzig,265 and Baron266 have each demon- treatment with short-term (3 months) treatment with celecoxib (16 mg/
strated a significant preventive benefit of aspirin (300 mg/day, 160 or kg/day, a dose that corresponds to 400 mg BID in adults) is well
300 mg/day, and 81 or 325 mg/day, respectively) versus placebo on tolerated and associated with no difference in adverse events from
the prevention of colorectal adenoma recurrence in patients with a placebo in children with FAP.277
history of adenomas. However, the most recent follow-up results from
Benamouzig’s group showed that after 4 years of follow-up the preven- Calcium
tive effects of aspirin were no longer apparent.267 Conversely, Sandler Numerous mechanistic and observational studies have suggested
and colleagues reported reduced incidence of colorectal adenomas the potential benefit for calcium in preventing polyp recurrence
after treatment with aspirin in a study of 517 CRC survivors treated and CRC.278–286
with aspirin (325 mg/day) or placebo.268 In a combined analysis of The Calcium Polyp Prevention and European Cancer Prevention
the British Doctors Aspirin Trial, which tested 5 years of treatment Organization Study Groups have both conducted clinical trials
with aspirin (500 mg/day) versus placebo, and the United Kingdom testing the efficacy of calcium in preventing adenoma recurrence
Transient Ischemic Attack (UK-TIA) Aspirin Trial, which tested 1 in individuals with a history of adenomas.287,288 Their results show
to 7 years of treatment with aspirin (300 or 1200 mg/day) versus 20% to 30% reduced recurrence after calcium supplementation. In
placebo, 5 years of aspirin treatment (≥300 mg/day) was found to contrast, the WHI conducted a trial of 36,282 postmenopausal women
significantly reduce incidence of CRC, particularly after use for from 40 WHI centers and found that supplementation with calcium
≥10 years.269 (500 mg/day) and vitamin D (200 IU/day) did not decrease colorectal
Drew and colleagues are investigating the effects of aspirin (81 or incidence.289 In 2013, updated results 5 years after active interven-
325 mg/day) versus placebo in patients previously diagnosed with tion ended revealed still no decrease in CRC (HR, 0.95 [95% CI,
colorectal adenoma in the Aspirin Intervention for the Reduction of 0.80–1.13]).290
356 Part I: Science and Clinical Oncology

Vitamin D/Calcium Polyp Prevention Study WHO Model List of Essential Medicines.308 Ornithine decarboxylase
The Vitamin D/Calcium Polyp Prevention Study, a phase II/III clinical (ODC) is overexpressed in tumor cells and upregulates both cell growth
trial, tested calcium and vitamin D for the prevention of colorectal and division,309 and is overexpressed in the intestinal mucosa of patients
neoplasia in 2259 participants with recently diagnosed neoplastic with FAP.310 Eflornithine irreversibly inhibits ODC, lowers colorectal
polyp(s) that had been successfully removed. Patients were treated mucosal polyamine levels, and is associated with minimal toxicity.311–314
with vitamin D3 (1000 IU/day) and calcium carbonate (1200 mg/ Several phase II and III clinical trials have studied eflornithine for the
day), alone or together, or placebo. Although the results of this study, prevention of CRC. Meyskens and colleagues conducted a trial testing
published in 2015, included few serious adverse events, the study also eflornithine and sulindac for the prevention of CRC.315 This phase
did not show a significant decrease in risk of colorectal adenomas III trial randomized 375 patients with colon polyps to receive DFMO
after treatment with vitamin D or calcium or both compared with (500 mg/day) and sulindac (150 mg/day) or placebo for 36 months.
placebo over 3 to 5 years.291 However, subsequent findings from this They found that patients treated with DFMO plus sulindac exhibited
study, released in 2017, suggested variable preventive benefit from significantly lower incidence of both colorectal adenoma recurrence
vitamin D supplementation based on SNPs in common variants of (RR, 0.30 [95% CI, 0.18–0.49], P < .001) and advanced adenomas
vitamin D pathway genes.292 Specifically, vitamin D decreased risk of (RR, 0.85 [95% CI, 0.011–0.65], P < .001) with minimal side effects.
advanced adenomas (RR, 0.36 [95% CI, 0.19–0.69]; P = .002) in Three additional trials, a phase II study testing eflornithine plus aspirin
individuals with the vitamin D receptor SNP re7968585 with the (NCT00983580) and two phase III studies testing eflornithine with
AA genotype, but significantly increased risk of advanced adenomas or without sulindac (NCT01483144; NCT01245816), are currently
(RR, 1.41 [95% CI, 0.99–2.00]; P = .05) in individuals with 1 or 2 ongoing. Moreover, Zell and colleagues have reported that the ODC1
G alleles. The researchers found no correlation between the prevention genotype impacts adenoma recurrence in patients treated with eflor-
of advanced colorectal adenomas treated with calcium supplementation nithine and sulindac compared with placebo.316
and SNPs in calcium pathway genes.
In 2016, Bonovas and colleagues published their results from a Eicosapentanoic Acid
meta-analysis of randomized placebo-controlled trials testing calcium Eicosapentanoic acid (EPA) is a naturally occurring omega-3 fatty
for the prevention of adenomas.293 Four trials met the defined study acid that demonstrated variable results in epidemiologic studies and
criteria; calcium was administered daily at a dose of 1200 to 2000 mg. minimal efficacy in reducing risk of CRC in a review of 20 cohorts
Their results show an 11% decrease in adenomas in patients treated from seven countries.317 However, EPA has been shown to decrease
with calcium versus placebo (fixed effects RR, 0.89 [95% CI, polyp count and size by 22% and 30%, respectively, after a treatment
0.82–0.96]). However, no significant decrease was observed in incidence time of 6 months.318 The Seafood Polyp Prevention Trial (AFOP) is
of advanced adenomas (fixed effects RR, 0.92 [95% CI, 0.75–1.13]). currently testing 12 months of treatment with aspirin (300 mg/day)
Collectively, these findings suggest that supplementation with calcium with or without EPA free fatty acid (2 g/day) versus placebo in prevent-
provides minimal prevention of colorectal adenomas, which may persist ing colorectal adenomas in high-risk patients in the English Bowel
for a year and a half to 5 years. Cancer Screening Programme.319
Hormone Replacement Therapy Mesalamine
HRT has been studied extensively as a possible preventive strategy for Gasche and colleagues throughout Europe are currently conducting
CRC. However, although these studies have suggested a preventive the phase II CRC prevention study Mesalamine for Colorectal Cancer
benefit for HRT in risk of CRC,294–297 neither the WHI study298 nor Prevention Program in Lynch Syndrome (MesaCAPP; NCT03070574).
the European Prospective Investigation into Cancer and Nutrition The aminosalicylate antiinflammatory drug mesalamine is used for
(EPIC)299 study, both of which tested estrogen and estrogen plus the treatment of patients with inflammatory bowel disease. This
progestin, demonstrated significantly decreased risk of colorectal study is testing the efficacy of 2 years of treatment with mesalamine
associated with HRT. The results from the subsequently published (1220 or 2400 mg/day) or placebo for the prevention of neoplasia in
follow-up results of the WHI study confirm these results.300–303 However, 540 patients with Lynch syndrome and is estimated to be completed
the most recent results from this study show that treatment with in 2024.
estrogen alone versus placebo is associated with an increased risk of
CRC incidence in postmenopausal women with prior hysterectomy Metformin
and prior colon polyp removal (HR, 13.47 [95% CI, 1.76–103.0], The first trial demonstrating the efficacy of metformin in suppressing
P < .001), and a statistically insignificant increase in CRC deaths colorectal aberrant crypt foci was obtained by Hosono and colleagues.320
(HR, 1.46 [95% CI, 0.86–2.46], P = .16) in postmenopausal The results of a phase III trial conducted by Higurashi and colleagues
women with prior hysterectomy.304 Collectively, these studies do that examined the preventive benefit of metformin for recurrence of
not support the use of estrogen with or without progestin for the metachronous colorectal adenomas and polyps in 151 nondiabetic
prevention of CRC. patients enrolled at five hospitals in Japan were published in 2016.321
These researchers found that 1 year of treatment with metformin
Other Drugs (250 mg/day) significantly decreased incidence of total polyps (hyper-
Curcumin plastic polyps and adenomas; RR, 0.67 [95% CI, 0.47–0.97]) and
Numerous preclinical studies have demonstrated that curcumin adenomas (RR, 0.60 [95% CI, 0.39–0.92]). In addition, minimal
(diferuloylmethane) provides both anticancer and antiinflammatory adverse events were reported, all of which were grade 1, demonstrating
effects305 and is capable of targeting cancer stem cells.306 Giardiello at the safety of metformin in this setting.
Johns Hopkins University has completed a phase II trial testing the
effects of 12 months of treatment with curcumin versus placebo in Vaccines
preventing CRC in 50 patients with FAP (NCT00641147). Results A number of studies are now investigating vaccines for the prevention
from this study, which has variations in polyp count and size as a of CRC. Schoen is leading a phase II study that is investigating the
primary end point, are currently pending. effectiveness of a MUC1–poly-ICLC vaccine on preventing polyp
recurrence and the progression of advanced adenomatous polyps into
Eflornithine colon cancer in individuals with a history of advanced colorectal
Difluoromethylornithine (DFMO, eflornithine) has been used to adenoma(s) (NCT0773097). The MUC1–poly-ICLC vaccine immu-
control facial hair growth and to treat gambiense sleeping sickness nizes against the Muc-1 tumor-associated antigen, a mucin expressed
for well over 20 years,307 and is listed as one of the medicines on the by colonic epithelial cells in the lumen of the large intestine. Initial
Lifestyle and Cancer Prevention  •  CHAPTER 22 357

results published in 2013 demonstrated an immune response in almost The Shandong Intervention Trial, conducted in Linqu County,
44% of vaccinated subjects, with no associated toxicity.322 The results China, randomized 3365 individuals with advanced precancerous
also suggested that lack of immune response in the remaining 66% gastric lesions to receive H. pylori treatment (amoxicillin and omepra-
of subjects was associated with high levels of circulating myeloid- zole) for 2 weeks with or without 7.3 years of vitamin supplements
derived suppressor cells before vaccination. Future results defining (vitamin C, vitamin E, and selenium) and/or garlic supplements.327
the impact of this vaccine on adenoma recurrence will determine its Neither the vitamin nor the garlic supplements for 7.3 years of treatment
efficacy in preventing CRC. Schoen is testing this vaccine in another reduced the prevalence of precancerous gastric lesions or the incidence
multisite phase II trial in 120 patients with newly diagnosed advanced of gastric cancer. However, eradication of H. pylori reduced incidence
colon polyps that is currently ongoing (NCT02134925). Another of precancerous lesions. The 14.7-year follow-up results demonstrated
ongoing early-phase vaccine study is investigating the prevention of a 39% significant reduction in gastric cancer incidence (OR, 0.61
metastases in CRC patients after vaccination with the adenovirus [95% CI, 0.38–0.96]; P = .32), as well as a non–statistically significant
5–human guanylyl cyclase (Ad5-hGCC)–PADRE vaccine (phase I, comparable reduction (33%; HR, 0.67 [95% CI, 0.36–1.28]) in gastric
NCT01972737). cancer mortality.333 In addition, although the vitamin and garlic
supplements demonstrated non–statistically significant decreases in
Gastric Cancer gastric cancer incidence and death, vitamin supplementation was
associated with a significant fully adjusted (for age, sex, alcohol use,
Helicobacter pylori smoking) reduction in esophageal or gastric cancer mortality, which
Risk of both local and distant recurrence of gastric cancer is high, was a secondary study end point (HR, 0.51 [95% CI, 0.29–1.03];
and both chronic gastritis and most gastric cancers are associated with P = .014).
chronic mucosal infection with the gram-negative bacterium H. pylori; The findings of a meta-analysis of the efficacy of adding probiotics
in addition, H. pylori accounts for 5.5% of all cancer incidence.323 A to standard H. pylori triple therapy were published in 2016; data from
recent meta-analysis of the prevalence of H. pylori infection during 30 RCTs with a total of 4302 per-protocol and 4515 intention-to-treat
childhood found that approximately one-third of children worldwide (ITT) patients were analyzed.334 H. pylori triple therapy plus probiotics
were currently or had previously been infected with H. pylori.324 Con- increased eradication of H. pylori by 12.2% in the per-protocol group
sequently, many RCTs investigating the role of H. pylori infection in (RR, 1.122 [95% CI, 1.091–1.153]; P < .001) and 14.1% in the
the development of noncardia gastric cancer have been conducted, with ITT group (RR, 1.141 [95% CI, 1.106–1.175]; P < .001) versus H.
conflicting and/or non–statistically significant initial findings.325–330 pylori triple therapy alone. This increase in eradication was demonstrated
However, extended follow-up results have demonstrated the association regardless of age or status of being Asian. A reduction in risk of
of H. pylori infection and gastric cancer, as well as the efficacy of H. antibiotic-associated side effects was also associated with the addition
pylori eradication in the prevention gastric cancer.331,332 of probiotics.
In 2000 Correa and colleagues released results from a randomized Population-based H. pylori screening and treatment in populations
controlled chemoprevention trial of antioxidant supplements (ascorbic at high risk are currently recommended by a growing number of
acid and β-carotene) and anti–H. pylori triple therapy (combined groups around the world, including the Asian-Pacific Gastric Cancer
treatment with antibiotics and proton pump inhibitors) in 795 Consensus group.335 However, these recommendations are currently
patients with gastric precancerous lesions in a high-risk region of in flux because antibiotic resistance rates of the bacterium vary, and
Colombia.325 After 6 years of follow-up, treatment with anti–H. pylori persistent infection with H. pylori is detrimental for gastric cancer
triple therapy alone (RR, 4.8 [95% CI, 1.6–14.2]) or ascorbic acid and mucosa-associated lymphoid tissue (MALT) lymphoma, among
(RR, 5.0 [95% CI, 1.7–14.4]) or β-carotene (RR, 5.1 [95% CI, other benign diseases, but is potentially beneficial for individuals at
1.7–15.0]) demonstrated significantly increased regression of precur- risk for other diseases, including Barrett esophagus and EAC.336 In
sor lesions, suggesting the potential efficacy of these interventions in addition, geographic variations in the effects of H. pylori infection on
preventing gastric cancer. Combined treatment with anti–H. pylori risk of gastric cancer have been reported, particularly between high-risk
triple therapy and the antioxidant supplements did not significantly Asian populations that show increased risk337 and low-risk Western
improve regression. Patients with atrophy (RR, 8.7 [95% CI, 2.7–28.2]) populations that show no or decreased risk.338–340 It has been suggested
and intestinal metaplasia (RR, 5.4 [95% CI, 1.7–17.6]) demonstrated that these variations in the level of risk for gastric cardia cancer associated
significantly increased rates of regression of precancerous lesions after with H. pylori infection could be a result of etiologically distinct types
eradication of the H. pylori infection.333 After an additional 6 years of gastric cancer.341 This concept is supported by the results of several
of follow-up, the benefits associated with both ascorbic acid and recent meta-analyses, which identified genetic polymorphisms associated
β-carotene were lost; however, eradication of H. pylori continued with increased susceptibility to H. pylori infection, and/or increased
to be associated with precursor lesion regression.331 The most recent risk for H. pylori–mediated gastric cancer. In 2017 alone, study
results from this study with 16 years of follow-up further defined the investigators reported such polymorphisms in (1) DNA repair genes
association between long-term exposure to H. pylori and progression (TP53, BRIP1, ERCC4, and XRCC3)342; (2) IL-1B-31C/T,
of precancerous lesions, and support the use of anti–H. pylori triple IL-1B-511-C/T, and IL-8-251T/A,343 IL-2,344,345 and IL-6 and IL-6R346;
therapy in high-risk individuals.332 The researchers found that 16 (3) the glutathione S-transferases (GSTs) GSTM1, GSTP1, and
years of continuous H. pylori infection increased risk of progression GSTT1347,348; (4) H. pylori349; (5) long noncoding (Lnc) RNA
of precancerous lesions to more advanced diagnoses versus no change rs16901946 (G)350; (6) MUC1351; (7) prostate stem cell antigen (PSCA),
or regression (P = .0001). In addition, higher risk of progression protein kinase AMP-activated catalytic subunit alpha 1 (PRKAA1),
occurred in individuals with incomplete-type intestinal metaplasia solute carrier family 52, member 3 (SLC52A3), and zinc finger and
versus those with complete-type intestinal metaplasia (odds ratio [OR], BTB domain containing 20 (ZBTB20); and (8) Toll-like receptors
11.3 [95% CI, 1.4–91.4]). Continuous H. pylori infection was also (TLRs) TLR1 and TLR10.352 These studies represent just a small
associated with a 0.20 unit/year increase in histopathology scores sample of the results that have been released over the past 5 years but
(95% CI, 0.12–0.28), and individuals with atrophy but not intestinal provide compelling evidence of the existence of etiologically distinct
metaplasia exhibited significantly higher increases in histopathology types of gastric cancer; studies clarifying this issue are currently
scores compared with individuals with intestinal metaplasia (P < .001). underway and will, it is hoped, aid in cancer prevention strategies for
These results demonstrate that risk of progression of precancerous gastric H. pylori–regulated gastric cancer.
lesions is significantly increased with long-term H. pylori exposure, To date, H. pylori screening and treatment have not been adopted
and that individuals with incomplete-type intestinal metaplasia are at by any country.336 However, China is currently recruiting over 180,000
high risk. participants to the largest H. pylori screening and treatment trial.353
358 Part I: Science and Clinical Oncology

The results of this and other long-term studies on the relationship lesions than patients lacking methylation reduction (OR, 1.92 [95%
among H. pylori screening and treatment and the risk of gastric cancer, CI, 1.03–3.60]), suggesting the potential for decreased risk of gastric
antibiotic resistance, and other diseases will determine the impact of cancer due to decreased COX-2 methylation via treatment with
this strategy for the prevention of gastric cancer. celecoxib or anti–H. pylori therapy.
A 2016 meta-analysis that included 47 studies of antiinflammatory
Nonsteroidal Antiinflammatory Drugs drugs for gastric cancer prevention showed decreased risk of gastric
The effectiveness of NSAIDs for the prevention of gastric cancer has cancer with antiinflammatory drug use (RR, 0.78 [95% CI,
been and remains controversial; however, efficacy has been suggested 0.71–0.85]).358 In addition, this analysis showed that use of COX-2
in numerous observational studies. A meta-analysis published in 2010 inhibitors was more effective for the prevention of gastric cancer than
across 14 studies with 5640 diagnoses of gastric cancer was unable to other NSAIDs (RR, 0.45 [95% CI, 0.29–0.70]). Ultimately, additional
demonstrate a statistically significant difference in risk of gastric cancer long-term studies are need to establish the specific populations most
based on aspirin use or nonuse.354 However, on stratification by location likely to benefit from preventive treatment with NSAIDs for the
and infection with H. pylori, aspirin users demonstrated reduced risk prevention of gastric cancer.
of noncardia gastric cancer (OR, 0.62 [95% CI, 0.55–0.69]) and
reduced risk of H. pylori infection (OR, 0.62 [95% CI, 0.42–0.90]) Hepatocellular Cancer
compared with nonusers. Aspirin was also associated with reduced
risk for noncardia gastric cancer (OR, 0.73 [95% CI, 0.62–0.87]) Major risk factors for hepatocellular carcinoma (HCC) include
and H. pylori infection (OR, 0.62 [95% CI, 0.55–0.69]) in Caucasians, viruses, particularly hepatitis B virus (HBV) and hepatitis C virus
suggesting that aspirin may be beneficial in preventing noncardia (HCV),359–361 cirrhosis,362,363 environmental toxins (e.g., ingestion of
gastric cancer in Caucasian and H. pylori–infected patients. Another aflatoxin in parts of Africa),364 diabetes,365 tobacco use, and alcohol
meta-analysis of eight trials with a total of 25,570 participants on the consumption.366
efficacy of aspirin as a preventive strategy for gastric cancer showed Consequently, strategies focused on the prevention of HBV and
a significant decrease in gastric cancer mortality in patients with more HCV,367,368 prevention or diminished progression of cirrhosis,369 reduced
than 10 years of aspirin use.355 Most recently, Huang and colleagues355a exposure to toxins,370 prevention or effective management of diabetes,371
conducted a meta-analysis of 24 studies and identified reductions in tobacco cessation, and alcohol reduction or elimination constitute
risk of gastric cancer following use of any NSAIDs (RR, 0.78 [96% potential techniques for the prevention of liver cancer. In addition,
CI, 0.72–0.85]), aspirin (RR, 0.70 [96% CI, 0.62–0.80]), and as with most cancers, behavioral modifications, including diet and
nonaspirin NSAIDs (RR, 0.86 [96% CI, 0.80–0.94]), particularly exercise, are beneficial preventive interventions.372 Unfortunately, clinical
for noncardia gastric cancer. data have yet to clearly define a preventive benefit associated with
Jankowski is leading the currently ongoing aspirin and esomepra- dietary supplements.373
zole chemoprevention trial AspECT, a national trial in the united
Kingdom that has accrued its goal of 2500 patients (EudraCT Hepatitis B Virus and Hepatitis C Virus Vaccine
2001-003836-77).240 This study, scheduled to publish results in A large-scale study of HBV vaccination of children in Taiwan dem-
2018, is investigating the effects of treatment with the proton pump onstrated the effectiveness of this strategy for the prevention of
inhibitor esomeprazole (20 mg/day or 80 mg/day) with or without HCC.368,374–376 This initiative was implemented in July 1984. Although
aspirin (300 mg/day) in individuals 18 years of age or older with Barrett 82,000 HCC diagnoses were listed in the Taiwan National Cancer
metaplasia in order to determine the effects of aspirin on all-cause Registry from 2003 to 2011, almost all of these were in middle-aged
mortality and EAC. Results from this study on all-cause mortality (50.1%) or elderly (49.1%) individuals. Therefore the launch of this
will, it is hoped, include and demonstrate the effect of individual universal immunization program has resulted in the virtual eradication
and combined treatment with esomeprazole and aspirin on risk of of both HBV and HCC in children, who account for only 0.04% of
gastric cancer. all HCC diagnoses since 2011.377
The Qidong Hepatitis B Intervention Study was conducted in
Cyclooxygenase-2 Inhibitors Qidong, China from 1985 to 1990.378 This population-based RCT
In 2012 the first results of a randomized trial specifically designed to involved the vaccination of 38,366 newborns who completed an HBV
assess the efficacy of treatment with a COX-2 inhibitor (celecoxib, vaccination series and 34,441 newborns who did not receive the
200 mg twice daily for 24 months) with or without prior anti–H. vaccination series. This neonatal HBV regimen demonstrated a sig-
pylori triple therapy (7 days) and anti–H. pylori triple therapy nificant decrease in HBV surface antigen seroprevalence and risk of
alone (7 days) versus placebo for the prevention of gastric cancer primary liver cancer. The long-term preventive efficacy of HBV
were published.356 This study accrued 1024 participants in Linqu immunization for prevention of chronic hepatitis B, liver fibrosis, and
County, Shandong Provence, China, who were 35 to 64 years of cirrhosis have since been evaluated, and protection has been demon-
age and had both H. pylori infection and advanced gastric lesions. strated to extend to marrying age.379 Results from this study have
Whereas individual treatment with both celecoxib and anti–H. pylori shown that the administration of a single HBV vaccine booster at 10
therapy versus placebo was associated with significant regression of to 14 years of age decreases the incidence of HBV infection in high-risk
both gastric lesions (celecoxib: 52.8% versus 41.2%; anti–H. pylori individuals.380
therapy: 59.3% versus 42.1%) and overall risk (celecoxib: OR, 1.72 An effective prophylactic HCV vaccine has yet to be developed.
[95% CI, 1.07–2.76]; anti–H. pylori therapy: OR, 2.19 [95% CI, However, advances in HCV vaccine formulations and modalities381
1.32–3.64]), combined treatment did not improve regression of advanced and preclinical studies in a variety of animal models have demonstrated
gastric lesions. promising candidates for HCV vaccines that may translate to the
More recently, Zhang and colleagues published the results of a clinic in future years.382
trial assessing the relationship between treatment with anti–H. pylori
therapy and/or the COX-2 inhibitor celecoxib and both COX-2 Other Strategies
methylation levels and gastric cancer in 809 participants with gastric Capecitabine
lesions.357 Methylation of COX-2 levels were decreased by treatment A phase II/III trial for the prevention of HCC recurrence after curative
with both celecoxib and anti–H. pylori therapy; however, as with the resection is using capecitabine, which is an FDA-approved drug for
study by Wong and colleagues, no additive or synergetic effects were metastatic colorectal and breast cancers, and gastric and esophageal
associated with combined treatment. Patients with reductions in COX-2 cancers (NCT00561522). Anticipated enrollment is 290, and individu-
methylation demonstrated significantly higher regression of gastric als will be randomized to receive capecitabine (four to six cycles of
Lifestyle and Cancer Prevention  •  CHAPTER 22 359

1250 mg twice daily for 14 days, followed by 7 days without drug)


or no intervention. Selective Estrogen Receptor Modulators
Over the last 25 years there has been a focused effort to develop drugs
Atorvastatin to prevent breast cancer development. The results of clinical trials
Chen is currently conducting a phase IV trial of the lipid-lowering testing antiestrogen drugs for breast cancer prevention are shown in
drug atorvastatin (10 mg/day) versus placebo for the prevention of Table 22.7. The rationale for using antiestrogen drugs for cancer
HCC recurrence in individuals with curative treatment of HCC prevention came from therapeutic trials of antiestrogen drugs that
(NCT03024684). The expected accrual for this study, which opened were tested as adjuvant therapy of early-stage breast cancer. These
in 2017, is 240. clinical trials demonstrated that women taking the antiestrogen drug
tamoxifen developed 50% fewer contralateral primary breast cancers
Interferon than did women taking placebo.393–395 This observation led to the
Interferon (IFN), a cytokine, has been investigated in a randomized development of many phase III breast cancer prevention trials using
clinical trial of 101 male patients with chronic HBV infection in tamoxifen as preventive therapy in women without breast cancer (see
Taiwan.383 Results after long-term follow-up showed a significant Table 22.7). The largest of these trials was the National Surgical
decrease in HCC incidence in the treatment arm (1.5% incidence) Adjuvant Breast and Bowel Project (NSABP)–P1 trial.396 In this trial,
compared with the control arm (12% incidence). Subsequently, a premenopausal and postmenopausal women at moderately high risk
41% reduction in HCC incidence was reported by Yang and colleagues of breast cancer were treated with tamoxifen or placebo for up to 5
in a meta-analysis of RCTs testing IFN therapy in subjects with years.396,397 This study was stopped early when initial results showed
HBV-related cirrhosis.384 IFN has also been tested (although not in that women taking tamoxifen developed 49% fewer breast cancers
a randomized clinical trial) for the prevention of HCC in subjects than did women taking placebo.396 These results led to FDA approval
with chronic HCV.385–389 of tamoxifen for breast cancer risk reduction in high-risk women.
Several similar trials showed similar results with long-term follow-up
Lamivudine (the UK and Italian studies also showed that tamoxifen reduced the
Liaw and colleagues conducted a phase III trial of lamivudine, an risk of breast cancer by at least 30%).398–401 Although tamoxifen
antiretroviral medication for HIV/AIDS and hepatitis B, in the Asian- prevented the development of ER+ breast cancers, the results of each
Pacific region.390 A total of 651 participants were recruited, of whom of these trials showed that tamoxifen did not prevent ER− breast
98% were Asian and 85% were male. Subjects received lamivudine cancers.398,399,401,402 Tamoxifen is FDA approved for breast cancer risk
(n = 436) or placebo (n = 215), and the primary end point was time reduction, but fewer than 1% of eligible women take this medication
to disease progression, which was defined as hepatic decompensation, for breast cancer prevention owing to a concern about the side effects
HCC, spontaneous bacterial peritonitis, bleeding gastroesophageal of tamoxifen (increased risk of hot flashes, vaginal dryness, and blood
varices, or liver disease–related death. Because of the significant protec- clots and a rare increased risk of uterine cancer).
tive effect of lamivudine, the study was terminated prematurely at Another SERM, raloxifene, has been shown to also prevent ER+
32.4 months. Disease progression was reached in 7.8% of lamivudine- breast cancers. Raloxifene was initially developed and later approved
treated participants and 17.7% of placebo-treated participants. In as a drug for the treatment of osteoporosis to prevent bone fractures.
addition, HCC incidence was reduced in the treatment arm (3.9%) However, the MORE (Multiple Outcomes of Raloxifene Evaluation)
compared with the control arm (7.4%). Lamivudine was also the clinical trial tested this drug in postmenopausal women with osteo-
focus of a retrospective study conducted in Japan.391 This study analyzed porosis for its ability to prevent breast cancer.407 The results of the
2795 patients, of whom 657 had received lamivudine and 2138 had MORE study demonstrated that women taking raloxifene had 72%
not. Patients treated with lamivudine (1.1% incidence) had significantly fewer breast cancers than seen in women taking placebo.415 These
fewer cases of HCC compared with those who were not (13.3% promising results led to the development and conduction of the Study
incidence). of Tamoxifen and Raloxifene (STAR) trial, which compared the ability
of tamoxifen or raloxifene to prevent breast cancer in high-risk
Glycyrrhizin postmenopausal women. The initial results of the STAR study showed
Glycyrrhizin, an antiinflammatory agent, was the focus of a retrospective that both tamoxifen and raloxifene had similar preventive activity405
study that examined the effect of glycyrrhizin therapy on HCC in (each prevented approximately 50% of the expected breast cancers);
individuals with IFN-resistant HCV.392 A total of 244 subjects received however, with longer follow-up, raloxifene’s ability to prevent breast
intravenous glycyrrhizin injection, whereas 102 did not; all 346 of cancer eroded over time, with raloxifene being 76% as effective as
the participants had alanine transaminase values that were at least tamoxifen at preventing invasive breast cancer after 7 years. Important
twice the upper limit of normal. Glycyrrhizin vaccination significantly to note, the side effects of raloxifene were reduced compared with
decreased HCC incidence in participants with IFN-resistant HCV tamoxifen. Raloxifene caused fewer side effects, with less frequent hot
and high alanine transaminase values compared with placebo (HR, flashes, episodes of vaginal dryness, and thrombotic events. Raloxifene
0.49 [95% CI, 0.27–0.86]; P = .014). also did not increase the risk of uterine cancer. The promising results
of the STAR trial led to the FDA approval of raloxifene for breast
Breast Cancer cancer prevention in high-risk postmenopausal women. However,
despite the improved side effect profile of raloxifene as compared with
Several strategies for breast cancer prevention have been studied, tamoxifen, less than 5% of eligible women at high risk of breast cancer
including dietary changes, lifestyle changes with increased exer- take this medication for breast cancer prevention, because of concerns
cise, preventive drug therapy, and prophylactic surgery. Of these, about the side effects of this medication.
surgery and preventive drug therapy have been shown to be highly Two other SERMs, lasofoxifene and arzoxifene, have been tested for
effective preventive strategies, but many women do not want to their ability to prevent breast cancer. Like tamoxifen and raloxifene,
experience the side effects of medications or surgical procedures. these drugs reduced the frequency of ER+ breast cancer without
Therefore there has been a major emphasis to find more toler- reducing the frequency of ER− breast cancer. In the PEARL trial,
able, yet effective strategies for breast cancer prevention. Several lasofoxifene (0.25 mg/day and 0.5 mg/day) reduced the incidence of
large studies of diet and exercise are currently ongoing (reviewed all invasive breast cancers by 79%, and reduced ER+ breast cancers
in Chapter 88). This chapter discusses the progress made with use by 83%.412 As with raloxifene, lasofoxifene use was associated with
of hormones, vitamins, targeted drugs, and vaccines to prevent increased thromboembolic events, leg cramps, and hot flushes, but
breast cancer. was not associated with an increased risk of endometrial hyperplasias
360 Part I: Science and Clinical Oncology

Table 22.7  Selective Estrogen Receptor Modulator (SERM) Breast Cancer Prevention Studies
Trial Patient Population Study Design Results Reference
TAMOXIFEN
Royal Marsden 2494 high-risk women Tamoxifen (20 mg/day) or No difference initially; with longer Powles et al, 1998403
30–70 yr of age placebo for 8 yr follow-up, reductions in invasive Powles et al, 2007398
and ER+ breast cancer incidence;
no significant change in ER− or
overall breast cancer incidence
NSABP-P1 (BCPT) 13,388 high-risk women Tamoxifen (20 mg/day) or Reductions in invasive, Fisher et al, 1998396
>35 yr of age placebo for 5 yr noninvasive, and ER+ breast Fisher, 2005397
cancer incidence; no significant Fisher et al, 2005402
change in ER− tumors
Italian 5408 normal-risk women Tamoxifen (20 mg/day) or Reductions in invasive and ER+ Veronesi et al, 1998404
with a hysterectomy placebo for 5 yr breast cancer incidence; no Veronesi et al, 2007399
35–70 yr of age significant change in ER− tumors
IBIS-I 7154 high-risk women Tamoxifen (20 mg/day) or Reductions in invasive, Cuzick et al, 2002400
35–70 yr of age placebo for 5 yr noninvasive (DCIS), ER+, and Cuzick et al, 2007401
overall breast cancer incidence; no
significant change in ER− tumors
NSABP-P2 (STAR) 19,747 high-risk, Tamoxifen (20 mg/day) Reductions in invasive and Vogel et al, 2006405
postmenopausal women or raloxifene (60 mg/day) noninvasive ER+ tumors less Vogel et al, 2010406
>35 yr of age for 5 yr effective (but with decreased
toxicity) with raloxifene than
tamoxifen
RALOXIFENE
MORE 7705 postmenopausal women Raloxifene (60 or 120 mg/ Reductions in invasive, ER+, and Cummings et al, 1999407
with low BMD <80 yr of age day) or placebo for 4 yr overall breast cancer incidence; no Ettinger et al, 1999408
significant change in ER− tumors
CORE 4011 postmenopausal women Raloxifene (60 mg/day) or Reductions in invasive, ER+, and Martino et al, 2004409
with low BMD (reconsented placebo for an additional overall breast cancer incidence; no
from MORE trial) <80 yr of age 4 yr after 4 yr of significant change in noninvasive
raloxifene on MORE trial of ER− tumors
RUTH 10,101 postmenopausal Raloxifene (60 mg/day) or Reduction in invasive ER+ tumors Barrett-Connor et al,
women with CHD placebo for a median of 2006410
>35 yr of age 5.6 yr
LASOFOXIFENE
PEARL 8556 women with low BMD Lasoxifene (0.25 mg or Reduction in invasive ER+ tumors Cummings et al, 2010411
59–80 yr of age 0.5 mg/day) or placebo LaCroix et al, 2010412
ARZOXIFENE
GENERATIONS 9354 women with low BMD Arzoxifene (20 mg/day) Reduction in invasive ER+ tumors Cummings et al, 2010413
60–85 yr of age or placebo for 4 yr Powles et al, 2012414

BMD, Bone mineral density; CHD, coronary heart disease; CORE, Continued Outcomes of Raloxifene Evaluation (CORE); ER, estrogen receptor; IBIS-I, International Breast
Intervention Study; Italian, Italian Randomized Tamoxifen Prevention Trial; MORE, Multiple Outcomes of Raloxifene Evaluation; NSABP-P1, National Surgical Adjuvant Breast and
Bowel Project Breast Cancer Prevention Trial P1; NSABP-P2, National Surgical Adjuvant Breast and Bowel Project Study of Tamoxifen and Raloxifene P2; PEARL, Postmenopausal
Evaluation and Risk Reduction With Lasofoxifene trial; RUTH, Raloxifene Use for the Heart trial; Royal Marsden, Royal Marsden Tamoxifen Prevention Trial.

or endometrial cancers. As with the other SERMs, lasofoxifene had FDA approval for a breast cancer risk reduction indication. Therefore
no effect on the development of ER− breast cancers. In the GEN- these effective drugs are not generally used for breast cancer prevention.
ERATIONS trial, arzoxifene (20 mg/day) was tested for its ability to Cuzick and colleagues published a meta-analysis of studies, showing
prevent bone fractures and breast cancer in postmenopausal women that, overall, clinical trials with SERMs have demonstrated that these
with osteoporosis.413 After a follow-up period of 36 months (for bone antiestrogen drugs can prevent the development of invasive ER+ breast
fracture assessment) or 48 months (for breast cancer assessment), there cancer, but not ER− breast cancer.416 In a consensus statement, Cuzick
were 41% fewer vertebral bone fractures and 56% fewer invasive and colleagues417 suggested that women at high risk of breast cancer,
breast cancers in women taking arzoxifene than in women taking such as those with a strong family history of breast cancer, high
placebo (the specific effect on ER+ and ER− breast cancers was not mammographic density, and atypical hyperplastic premalignant lesions,
described). Arzoxifene caused significantly more thromboembolic events, are good candidates for SERM preventive therapy. A study by Reimers
hot flushes, and vaginal discharge, but did not significantly increase and colleagues suggested that the uptake of these agents may be
uterine cancers or endometrial hyperplasias.413 Despite the significant increasing in the high-risk groups; they observed in the primary care
breast cancer preventive effect and generally tolerable side effect profile setting that 37% of eligible high-risk women took an antiestrogen
of arzoxifene and lasofoxifene, the drug companies have not sought SERM for breast cancer prevention.418
Lifestyle and Cancer Prevention  •  CHAPTER 22 361

overall outcome of women with DCIS). The IBIS-II DCIS trial had
Aromatase Inhibitors a similar design as the NSABP B35 trial, and compared anastrozole
AIs (anastrozole, letrozole, and exemestane) have been shown to be and tamoxifen with regard to their ability to prevent invasive and
more effective than SERMs for the treatment of breast cancer (reviewed contralateral breast cancer in postmenopausal women. The results
in Litton and colleagues419); therefore there has been strong interest showed that both anastrozole and tamoxifen were similar in their
in use of these drugs for breast cancer prevention. The first indication ability to prevent DCIS recurrence. There was no statistically significant
that AI drugs would be effective for breast cancer prevention came difference in their ability to prevent any breast cancer event (HR,
from the ATAC trial, which tested anastrozole, tamoxifen, and the 0.89 [95% CI, 0.64–0.123]; P = .49), invasive breast cancers (HR,
combination for the treatment of early-stage breast cancer.420 Results 0.80 [95% CI, 0.52–1.24]; P = .32), or DCIS recurrences (HR, 0.99
from the ATAC trial showed that anastrozole alone was more effective [95% CI, 0.60–1.65]; P = .98).423 As expected, the side effect profiles
than tamoxifen in preventing the development of second primary for tamoxifen and anastrozole were different, with more muscle spasms,
breast cancers in women with early-stage breast cancer.421 Women uterine cancers, hot flushes, and deep vein thromboses with tamoxifen,
treated with anastrozole had 58% fewer contralateral primary breast and more bone fractures, musculoskeletal events, hypercholesterolemia,
cancers than those treated with tamoxifen when the ATAC trial and strokes with anastrozole.423
was initially reported (OR, 0.42; P = .007).420 With the report of The first of the AI primary prevention studies to be reported was
the 10-year follow-up data, the results showed that women taking the MAP.3 study, which compared the AI exemestane and placebo in
anastrozole developed 32% fewer contralateral breast cancers than healthy postmenopausal women at high risk of breast cancer.428 The
those taking tamoxifen (HR, 0.68; P = .01 for all breast cancers), results showed that women who took exemestane had 65% fewer
and 38% fewer ER+ contralateral breast cancers (HR, 0.62; invasive breast cancers than those who took placebo (HR, 0.35; P =
P = .003).421 .002), and 73% fewer ER+ breast cancers (HR, 0.27; P < .001).
These provocative results led to the development of several clinical Women taking exemestane had significantly more hot flushes, joint
trials testing AIs for their ability to prevent breast cancer in women and muscle pain, and GI symptoms than those taking placebo. However,
with previous ductal carcinoma in situ (DCIS) breast cancer or in there were no significant differences in skeletal fractures, thrombo-
high-risk women with no prior history of breast cancer (Table 22.8). embolic events, cardiovascular events, or other cancers between women
Several trials have tested the ability of anastrozole to prevent invasive taking exemestane and those taking placebo.428 These results indicate
breast cancer, either in postmenopausal women with DCIS breast that exemestane may be particularly useful in preventing ER+ breast
cancer (the NSABP B35 DCIS trial422 or the IBIS-II DCIS trial423), cancers and may be tolerable in high-risk women, particularly those
or in healthy high-risk postmenopausal women (IBIS-II trial).424 In without osteopenia or osteoporosis.
each of these trials, anastrozole was found to reduce the incidence of The IBIS-II prevention trial compared the ability of anastrozole
invasive breast cancer. In the NSABP B35 trial, anastrozole was and placebo to prevent breast cancer in healthy postmenopausal women
compared with tamoxifen for its ability to prevent the development at high risk of breast cancer. These women took anastrozole (or placebo)
of invasive breast cancer in postmenopausal women with DCIS breast for 5 years. After a median follow-up of 5 years, the results demonstrated
cancer. The results from this study showed that anastrozole was slightly that women who took anastrozole had 53% fewer breast cancers (HR,
more effective than tamoxifen at reducing the incidence of breast 0.47 [95% CI, 0.32–0.68]; P < .0001), 50% fewer invasive breast
cancer events (HR, 0.73 [95% CI, 0.56–0.96]; P = .0234) in post- cancers (HR, 0.50 [95% CI, 0.32–0.76]; P = .001), 70% fewer
menopausal women with ER+ DCIS.422 There were also fewer invasive noninvasive breast cancers (HR, 0.30 [95% CI, 0.12–0.74]; P = .009),
breast cancers (HR, 0.62 [95% CI, 0.42–0.90]; P = .0123) and fewer and 58% fewer ER+ invasive breast cancers (HR, 0.42 [95% CI,
DCIS recurrences (although nonsignificant; HR, 0.88 [95% CI, 0.25–0.71]; P = .001), but no difference in ER− invasive breast cancers
0.59–1.30]; P = .52) in women taking anastrozole than in those taking (HR, 0.78 [95% CI, 0.35–1.72]; P = .538).424 Anastrozole use was
tamoxifen.422 In addition, in the women taking anastrozole, there was associated with increased musculoskeletal events, hot flushes, vaginal
a significant reduction in contralateral breast cancers (HR, 0.64 [95% dryness, hypertension, and dry eyes, compared with placebo.424 No
CI, 0.43–0.96]; P = .0322). As with other breast cancer prevention difference was seen in the frequency of bone fractures, stroke, other
trials, there was no detectable difference in survival (given the excellent thrombotic events, or myocardial infarction.424

Table 22.8  Select Aromatase Inhibitor (AI) Breast Cancer Prevention Studies
Trial Patient Population Study Design Primary End Point Reference
DCIS TRIALS
NSABP B-35 3104 women accrued Anastrozole (1 mg/day) vs Ipsilateral/contralateral breast Margolese et al, 2003465
Postmenopausal with tamoxifen (20 mg/day) for 5 yr cancer incidence Richardson et al, 2007425
ER+/PR+ DCIS Margolese et al, 2016422
IBIS-II 2980 women accrued Anastrozole (1 mg/day) vs Ipsilateral/contralateral breast Cuzick, 2003426
(DCIS) Postmenopausal with DCIS tamoxifen (20 mg/day) for 5 yr cancer incidence Cuzick, 2008427
Forbes et al, 2016423
PREVENTION TRIALS
NCIC-MAP.3 4560 women accrued Exemestane (25 mg/day) vs Invasive breast cancer Goss et al, 2011428
Postmenopausal, high-risk placebo for 5 yr incidence
≥35 yr of age
IBIS-II 3864 women accrued Anastrozole (1 mg/day) vs Invasive breast cancer Cuzick, 2003426
Postmenopausal, high-risk placebo incidence Cuzick, 2008427
Cuzick et al, 2014424

DCIS, Ductal carcinoma in situ; IBIS-II, Second International Breast Cancer Intervention Study; NSABP B-35, National Surgical Adjuvant Breast and Bowel Project B-35;
NCIC-MAP.3, NCIC Clinical Trials Group Mammary Prevention.3 trial; PR, progesterone receptor.
362 Part I: Science and Clinical Oncology

Although the results of these phase III AI clinical trials demonstrate the limiting toxicities.445 A phase II clinical trial testing the ability of
that AIs are highly effective in preventing ER+ breast cancers in green tea extract to modulate mammographic density has completed
postmenopausal women, the exact role of AIs in managing breast accrual, but results have not yet been reported. Other studies have
cancer risk remains a debatable topic. It is clear that along with SERMs, tested the anti-HER2 drugs trastuzumab and lapatinib for the ability
the AIs offer a promising strategy to prevent ER+ breast cancer. to suppress the proliferation of HER2+ DCIS breast cancer cells (see
However, the decision to use a SERM, an AI, or no drug therapy at Table 22.9). These trials demonstrate that anti-HER2 drugs can affect
all depends on the physician’s experience and comfort with use of immune response and signaling pathways in HER2+ DCIS cells.
these drugs for breast cancer prevention, the patient’s desire to use These studies suggest that ultimately it may be possible to prevent
drug therapy and willingness to accept risk of side effects, and ultimately HER2+ DCIS from progressing to invasive breast cancer.422
the risk-benefit ratio perceived by both the physician and woman at
risk of breast cancer. Other Strategies to Prevent Breast Cancer
Although traditional targeted drug therapy offers significant potential
Prevention of Estrogen Receptor–Negative Breast Cancer to reduce breast cancer incidence, there is strong interest in developing
SERMs and AIs are very effective at suppressing the development of more effective and safer strategies for breast cancer prevention. To
ER+ breast cancer. However, they do not prevent the 30% to 40% this end, several investigators have developed vaccines for breast cancer
of breast cancers that are ER−. Thus, a major unmet need is the prevention. Peptide vaccines, pulsed dendritic cell vaccines, and
identification of drugs or strategies to prevent these particularly DNA-based vaccines have been developed and are now being tested
aggressive breast cancers. A variety of drugs are being studied in preclini- in clinical trials (Table 22.10). Disis and colleagues have studied peptide
cal models and early clinical trials to determine whether they prevent vaccines against the HER2 oncoprotein. They have conducted several
ER− breast cancer. These agents include drugs targeting the retinoic studies with an HER2 peptide vaccine, showing that it is safe and
acid X receptor, “rexinoids”; vitamin D; the active agent in green tea, that an anti-HER2 immune response can be generated.453–456 Disis
EGCG; metformin; and drugs targeting the HER2 oncogene, in has also tested a DNA-based anti-HER2 vaccine that also induces
addition to drugs being tested for the prevention of “triple-negative” anti-HER2 immunity.457–459 These vaccines are now being tested in
breast cancers. Preclinical studies of bexarotene429–431 and UAB30432 phase II cancer prevention trials. Two other groups have developed
have demonstrated that these RXR-selective retinoids (rexinoids) prevent anti-HER2 vaccines. Peoples and Mittendorf have developed several
the development of ER− and “triple-negative” breast cancer in mouse peptide-based anti-HER2 vaccines, including nelipepimut-S, which
models. These studies supported the early-phase biomarker modulation has been tested in phase I, II, and III trials for the treatment of breast
trials of these drugs in women with or at risk of breast cancer (Table cancer (NCT02636582) and is now being tested in a phase II cancer
22.9). Vitamin D has also been extensively studied as a breast cancer prevention trial in patients with DCIS (see Table 22.10). Czerniecki
prevention agent. These studies were based on epidemiologic studies developed and tested dendritic cell vaccines in which dendritic cells
suggesting that low vitamin D levels are associated with increased risk were pulsed with HER2 peptides and injected either intralesionally
of breast cancer.433–436 Multiple vitamin D trials tested the effect of or intranodally. He showed that this dendritic cell vaccine is safe and
very high doses of vitamin D on mammographic density (as a surrogate can induce anti-HER2 immunity. This anti-HER2 dendritic cell vaccine
end point biomarker for breast cancer; see Table 22.9). These studies is now being tested in a phase II DCIS cancer prevention clinical trial
have shown that high-dose vitamin D is tolerable and safe, but its (see Table 22.10).
effect on mammographic density has not yet been reported. Another Most of these efforts to develop breast cancer vaccines have focused
extensively studied potential breast cancer prevention drug is the on targeting HER2. However, Disis has developed a complex DNA-
antidiabetic drug metformin. The rationale for these studies came based vaccine composed of DNA fragments of three different antigens
from epidemiology studies that showed that diabetic individuals taking present in triple-negative breast cancers (i.e., that are negative for ER,
metformin had a reduced incidence of breast cancer compared with progesterone receptor [PR], and HER2). This STEMVac vaccine is
those taking insulin. Metformin activates AMP kinase, ultimately now being tested in an ongoing phase I clinical trial and is highly
reducing insulin production; in addition, metformin has been shown promising for the prevention of triple-negative breast cancer. The
to inhibit epithelial cell growth at high doses. Multiple early-phase results from these vaccine trials will demonstrate whether breast cancer
cancer prevention trials have been conducted that have demonstrated prevention using vaccines is feasible and effective. Ultimately, it is
the safety of this widely used drug (see Table 22.9). Given the extremely likely that a combination of preventive therapy (behavioral, dietary,
good safety profile of metformin, a phase III adjuvant breast cancer drug, and possibly vaccine interventions) will be required to completely
clinical trial was proposed and opened by the National Cancer Institute prevent all forms of breast cancer.
of Canada to test metformin in women with early-stage breast cancer
after their initial therapy (NCT01101438). This multicenter trial will Cervical Cancer
assess the effect of metformin on disease-free survival (the primary
end point), as well as on overall survival and, important for cancer Interest in the association between cervical cancer and sexual contact was
prevention applications, contralateral breast cancer incidence. This initiated with an observation by Rigoni-Stern in 1842, who observed
very important trial should clearly demonstrate the activity of metformin that cervical cancer–related deaths were more common in women
with regard to breast cancer recurrence as well as breast cancer who were married, widowed, or prostitutes but rare in virgins and
development. nuns.466 It was not until the 1970s that research was designed and
The natural product EGCG, one of the active ingredients in green conducted to define a causal relationship between HPV infection
tea, has been studied both in preclinical and early clinical trials for and the development of cervical cancer.467 Currently, 12 HPV subtypes
its ability to prevent breast cancer. The basis for studying green tea are recognized by the IARC as group 1 carcinogens, including HPV
and its active ingredient comes from epidemiologic studies of cancer types 16, 18, 31, 33, 35, 39, 45, 51, 52, 56, 58 and 59.468 Of these
incidence in China. Several epidemiologic studies have shown that high-risk HPV subtypes, cervical adenocarcinoma has been shown to be
green tea consumption is inversely associated with cancer incidence, caused at particularly high rates by HPV-16 (50.9%), HPV-18 (31.6%),
including breast cancer.437–439 These findings led to testing of EGCG HPV-45 (11.6%),469 HPV-31 (12.6%), and HPV-33 (8.0%).470
in preclinical models, producing results showing that EGCG suppresses
the development of mammary tumors in these animals.440–444 Early-phase Human Papillomavirus Vaccine
clinical trials have been performed to determine the maximum tolerated Three vaccines against HPV infection have been developed: a bivalent
dose (found to be 600 mg twice a day,445 equal to 8 to 10 cups of vaccine for HPV-16 and HPV-18 (bHPV; Cervarix); a quadrivalent
green tea per day), with elevated liver enzymes and GI toxicity being vaccine for HPV types 6, 11, 16, and 18 (qHPV; Gardasil); and a
Lifestyle and Cancer Prevention  •  CHAPTER 22 363

Table 22.9  Breast Cancer Prevention Studies Using Other Novel Agents
Phase Patient Population Study Design Results Reference
REXINOID
Bexarotene 87 high-risk women Bexarotene (200 mg/m2) or Reduction in cyclin D1 RNA Brown et al, 2008446
 II Postmenopausal placebo for 28 days (NCT00055991)
9cUAB30 40 women ≥18 yr of age with early- 9cUAB30 (orally daily for 14–28 Primary end point: changes in Ki67 Krontiras et al
  IB stage invasive breast cancer days before surgery) expression. (NCT02876640)
VITAMIN D
II 20 high-risk premenopausal women Vitamin D (20,000 or 30,000 IU/ Increased vitamin D levels and Crew et al, 2015447
≥21 yr of age with LCIS or DCIS wk) for 1 yr decreased IGF1/IGFBP3 ratio (NCT00976339)
II 20 high-risk postmenopausal women Vitamin D (20,000 or 30,000 IU/ Increased vitamin D levels and Crew et al, 2015447
≥21 yr of age wk) for 1 yr decreased IGF1/IGFBP3 ratio (NCT00859651)
IIB 200 premenopausal women 18–74 yr of Vitamin D (weekly and daily), Primary end point: change Crew et al
age at high risk for breast cancer or placebo (1/wk) and vitamin in mammographic density at (NCT01097278)
D (daily) for 1 yr 12 months
Results not reported
III 250 premenopausal women ≤55 yr of Vitamin D (2000 IU/wk) or Primary end point: change Wood et al
age with breast density ≥25% (scattered placebo for 1 yr in mammographic density at (NCT01224678)
fibroglandular densities or greater) 12 months
Results not yet reported
II 30 premenopausal women ≤55 yr of at Vitamin D (10,000 IU/wk) for Primary end point: change Fabian et al
increased risk for breast cancer 6 months in mammographic density at (NCT01166763)
6 months
Results not yet reported
METFORMIN
I 5 women ≥18 yr of age with operable Metformin (850 mg BID) for Primary end points: changes in Storniolo et al
stage I or II breast cancer 7–21 days Ki67 and caspase-3 expression (NCT00984490)
II 200 nondiabetic women with breast Metformin (850 mg BID) or No difference in Ki67, but different Bonanni et al, 2012448
cancer placebo for 28 days insulin resistance–based effects (in
luminal B tumors)
II 150 premenopausal women 21–54 yr Metformin (850 mg/day for 4 Primary end point: change in Chow et al
of age weeks, then 850 mg BID or mammographic density at 12 mo (NCT02028221)
BMI ≥25 kg/m2 placebo for 48 wk) Results not yet reported
III 3649 women 18–74 yr of age with Metformin (850 mg BID in Primary end points: invasive Goodwin et al
invasive breast cancer weeks 1–4, then BID or placebo disease-free survival (not yet (NCT01101438)
for ≤5 yr) reported)
I 40 women ≥21 yr of age with newly Metformin (1500 mg/day) plus Primary end points: changes in Kalinsky et al
diagnosed invasive breast cancer or atorvastatin (80 mg/day) for 2 Ki67 expression (NCT01980823)
DCIS weeks prior to surgery Results not yet reported
EGCG (ACTIVE AGENT IN GREEN TEA)
I 40 women with a history of stage I to III EGCG (200, 600, 800 mg BID) or The maximum tolerated dose for Crew et al, 2012445
ER− breast cancer placebo for 6 months Poly E (EGCG) is 600 mg, BID (NCT00516243)
IB 40 women 21–65 yr of age with prior EGCG (orally BID) or placebo The maximum tolerated dose for Crew et al, 2012445
hormone receptor–negative stage I–III for 6 months EGCG is 600 mg BID (NCT00516243)
breast cancer
II 1075 postmenopausal women with high EGCG (800 mg/day) or placebo Elevated liver enzymes (6.7%) Samavat et al, 2015449
risk of breast cancer due to dense breast for 12 mo Mammographic density not yet Dostal et al, 2015450
tissue with differing COMT genotypes reported (NCT00917735)
TRASTUZUMAB
Pilot 69 women with DCIS Single dose of trastuzumab Augmented antibody-dependent Kuerer et al, 2011451
(8 mg/kg) cell mediated cytotoxicity (NCT00496808)
LAPATINIB
II 20 women with HER2-positive DCIS Lapatinib (1500 mg) vs placebo Decreased expression of pHER2 Estévez et al, 2014452
for 4 weeks and pERK; no change in Ki67 or
p27 expression
II 40 women with EGFR-positive or Lapatinib (1000 mg) vs placebo Primary end points: changes in Brown et al
HER-positive DCIS for 6 weeks Ki67 expression, safety (NCT00555152)

BMI, Body mass index; COMT, catechol-O-methyltransferase; DCIS, ductal carcinoma in situ; EGCG, epigallocatechin gallate; ER, estrogen receptor; LCIS, lobular carcinoma
in situ.
364 Part I: Science and Clinical Oncology

Table 22.10  Breast Cancer Prevention Studies Using Vaccines


Phase Patient Population Study Design Results Reference
I 22 patients with stage IV breast HER2/neu T-helper Minimal toxicity; prolonged, robust, antigen-specific Disis et al, 2009460
cancer who are receiving peptide-based vaccine immune response (median follow-up of 36 mo from (NCT00194714)
trastuzumab first vaccine)
II 56 patients with stage III/IV Intradermal HER2 ICD Primary end point: determine generation of Salazar et al
breast cancer who completed protein mixture or sterile immunologic memory to the HER2 ICD protein (NCT00363012)
chemotherapy, are receiving water 6 months after
trastuzumab, and completed vaccination with pNGVL3-
HER2 ICD plasmid-based hlCD vaccine
vaccine trial
I/II 23 patients ≥18 yr of age with HER-2/Neu peptide vaccine Primary end points: toxicity of infusing HER2-specific Disis et al
HER2-positive stage IV breast admixed with GM-CSF plus T cells (NCT00791037)
cancer cyclophosphamide and ex Results not yet reported
vivo expanded HER2-
specific T cells
I 30 HER2-negative stage III–IV CD105/Yb-1/SOX2/CDH3/ Primary end points: immunology efficacy (increased Disis et al
breast cancer patients MDM2-polyepitope plasmid Th1 cell immunity) and incidence of toxicity per (NCT02157051)
DNA vaccine (“STEMVac”) cancer therapy evaluation
Results not yet reported
I/II 195 high-risk node-negative HER2 peptide DFS was 94.6% in optimally dosed, 87.1% in Mittendorf et al,
patients with HER2-positive (nelipepimut-S) (E75) suboptimally dosed, versus 80.2% in control patients; 2014461
breast cancer vaccine with booster vaccine is safe (NCT00841399 and
inoculations versus no NCT00584789)
vaccination
II 300 disease-free node-positive Herceptin (6 mg/kg q3wk Primary end point: DFS at 24 and 36 months Peoples et al
and ER−/PR− node-negative for 1 yr) + NeuVax (E75) (NCT01570036)
HER2 1+/2+ breast cancer vaccine (6 vaccinations, 1
patients every 3 weeks)
II 108 high-risk women ≥18 yr of HER2 peptide Primary end point: immune response to the HER2 Mittendorf et al
age with DCIS (nelipepimut-S) plus peptide (NCT02636582)
GM-CSF vaccine, or GM-CSF Results not yet reported
alone (before surgery)
II 298 disease-free node-positive HER2 peptide AE37 vaccine Minimal toxicities; estimated 5-yr DFS in patients Mittendorf et al,
and high-risk node-negative plus GM-CSF vs GM-CSF with IHC 1+/2+ HER2-positive tumors was 77.2% with 2016462
breast cancer patients alone vaccine vs 65.7% with control subjects; estimated (NCT00524277)
5-yr DFS in patients with TNBC was 77.7% with
vaccine vs 49.0% with control subjects
II 180 HLA-A2+, disease-free HER2 peptide GP2 vaccine Interim report: minimal toxicities; estimated 5-yr DFS Mittendorf et al,
node-positive and high-risk plus GM-CSF versus in IHC3+/FISH+ patients was 94% in vaccinated 2016463
node-negative breast cancer GM-CSF alone patients vs 89% in control patients in the intent-to- (NCT02636582)
patients with HER2 (IHC1+/3+)- treat analysis, and was 100% in vaccinated patients
expressing tumors (estimated vs 89% in control patients in the per-treatment
enrollment: 600) analysis
IB 17 disease-free, HLA-A2+/A3+, GP2 vaccine + GM-CSF plus No dose-limiting or grade 3–5 local or systemic Clifton et al, 2017464
HER2-positive breast cancer standard-of-care toxicities; appropriate dose was 1000 µg of GP2 + (NCT03014076)
patients trastuzumab 250 µg of GM-CSF
I 30 women ≥18 yr of age with HER-2/Neu pulsed dendritic HER-2/neu was well-tolerated; vaccine induced Czerniecki et al659
DCIS cell vaccine by intranodal, decline and/or eradication of HER-2/neu expression; (NCT00107211)
intralesional, or both, no DCIS remained in 40% ER− and 5.9% ER+ patients;
vaccination phenotypes shifted after vaccination: 43.8% ER+
HER2+ lumB to ER+ HER2− lumA and 50% ER− HER2+
to ER− HER2−; depressed Th1 response restored with
HER2-directed Th1 immune interventions
I/II 58 women ≥18 yr of age with HER-2/Neu pulsed dendritic Vaccination was well tolerated; vaccination route did Czerniecki et al
HER-2/neu-positive DCIS cell vaccine by intranodal, not affect immune response rate; pCR rate was 28.6% (NCT02061332)
intralesional, or both, in DCIS vs 8.3% in IBC patients; pCR and non-pCR
vaccination DCIS patients had similar peripheral blood anti-HER2
immune responses; all pCR patients had anti-HER2
CD4 immune response in sentinel lymph node
I/II 58 women ≥18 yr of age with HER-2/Neu pulsed dendritic Primary end point: related adverse events (results not Czerniecki et al
HER-2/3-positive DCIS cell vaccine plus yet reported). (NCT02336984)
trastuzumab and
pertuzumab

DCIS, Ductal carcinoma in situ; DFS, disease-free survival; ER, estrogen receptor; FISH, fluorescence in situ hybridization; GM-CSF, granulocyte-macrophage colony-stimulating
factor; HER2, human epidermal growth factor receptor 2; HLA, human leukocyte antigen; IBC, invasive breast cancer; ICD, intracellular domain; IHC, immunohistochemistry; lum,
luminal; pCR, pathologic complete response; PR, progesterone receptor; TNBC, triple-negative breast cancer.
Lifestyle and Cancer Prevention  •  CHAPTER 22 365

nonavalent vaccine for HPV types 6, 11, 16, 18, 31, 33, 45, 52, and and three doses of the HPV-16/18 vaccine.498 Their findings show a
58 (9vHPV; Gardasil 9), which was developed from the quadrivalent 77.0% vaccine efficacy against HPV-16/18 with three doses (95%
vaccine. Although many countries have licensed Cervarix and the CI, 74.7–79.1), 76.0% with two doses (95% CI, 62.0–85.3), and
quadrivalent Gardasil, Gardasil 9 has only recently been licensed in 85.7% with a single dose (95% CI, 70.7–93.7), suggesting that a
some countries. These HPV vaccines provide maximal benefit when similar level of protection is obtained against cervical HPV-16/18
administered in HPV-uninfected individuals. Evidence of the effective- infections with one and two doses of the vaccine. However, cross-
ness of these HPV vaccines in both preventing HPV infection and protective efficacy of the vaccine against incident HPV-31, HPV-33,
decreasing incidence of several cancers, including anal, cervical, and HPV-45 infections showed a high level of dose-dependence: 59.7%
oropharyngeal, penile, vaginal, and vulvar cancers, and in decreasing with three doses (95% CI, 56.0–63.0), 37.7% with two doses (95%
the risk of genital warts and cervical intraepithelial neoplasia (CIN) CI, 12.4–55.9), and 36.6% with one dose (95% CI, −5.4 to 62.2).
grades 2 and 3 precursor lesions has been provided by several studies.467 In addition, analysis of the CVT trial shows that increased protection
However, the effectiveness and immunogenicity of each vaccine vary was obtained in women who received two doses when the second
based on its unique vaccine composition and which high-risk HPV dose was given at 6 months (82.6% [95% CI, 42.3-96.1]) rather than
subtypes are included. 1 month (75.3% [95% CI, 54.2–87.5]).

bHPV (Cervarix) qHPV (Gardasil)


Cervarix, the HP16/18-AS04-adjuvanted vaccine developed by Gardasil, developed by Merck, received FDA approval in 2006 and
GlaxoSmithKline, received FDA approval in 2009 for the prevention was the first HPV vaccine to be licensed for the prevention of (1)
of HPV-16 and HPV-18 infection–related cervical cancer, cervical vulvar and vaginal cancer; (2) HPV-6, HPV-11, HPV-16, and HPV-18
adenocarcinoma in situ, and CIN in girls and women 9 to 25 years infection–related cervical cancer, cervical adenocarcinoma in situ, and
of age.471 Cervarix contains 20 µg of HPV-16 capsid protein and cervical, vaginal, and vulvar intraepithelial neoplasia; and (3) HPV-6
20 µg of HPV-18 capsid protein.467 Numerous randomized phase and HPV-11 infection–related genital warts.499 This approval is for
II/III trials testing the HPV-16/18 vaccine have been conducted, all girls and women 9 to 26 years of age, and applies to boys and men
of which involved administration of three doses to female subjects 9 to 26 years of age only for aforementioned prevention of anal cancer,
15 years of age or older at 0, 1, and 6 months. These trials included anal intraepithelial neoplasia, and genital warts. Gardasil contains
(1) the multinational (Brazil, Canada, and the United States) Study 20 µg of both HPV-6 and HPV-18 capsid protein and 40 µg of both
HPV-001 (NCT00689741)472 and two extensions of this study; HPV-11 and HPV-16 capsid protein, and is the most widely admin-
(2) Study HPV-007 (NCT00120848)473,474; (3) Study HPV-023 istered HPV vaccine around the world.467,500 The initial phase I and
(NCT00518336)475–477; (4) the multinational (14 countries throughout II trials on Gardasil were conducted by Merck501–503 and have been
Asia, Australia, Europe, and North and South America) Study HPV-008, followed by numerous subsequent phase II/III trials, including the
the Papilloma Trial Against Cancer in Young Adults (PATRICIA; phase III Females United to Unilaterally Reduce Endo/Ectocervical
NCT00122681)478–483; (5) Study HPV-009, the Costa Rica HPV Disease (FUTURE) I and II trials, a phase III study by Olsson,504
Vaccine Trial (CVT; NCT00128661)484–491; (6) Study HPV-032 of and postlicensure studies by Brotherton,505 Crowe,506 Kahn,507
Japanese women (NCT00316693)492,493; (7) Study HPV-063, an Herweijer,508 Giuliano,509 Palefsky,510 and Kash and colleagues.511
extension of Study HPV-032 (NCT00929526)494; (8) Study HPV-039 Both the phase II and the FUTURE phase III studies mentioned
of Chinese women, currently ongoing (NCT00779766)495; and (9) previously have reported increased rates of localized adverse events at
Study HPV-015, the multinational (12 countries throughout Australia, the site of injection (3% and 10%, respectively), but no increase in
Europe, North and South America, and Southeast Asia) Human Papil- serious adverse events, in patients receiving the quadrivalent vaccine
lomavirus: Vaccine Immunogenicity and Efficacy trial (VIVIANE; versus those receiving placebo.512–514 In addition, the phase II studies,
NCT00294047).496,497 Prior HPV infection for the participants in which enrolled 2409 and 277 patients, reported a 90% decrease after
these trials is unknown, and aluminum or hepatitis A vaccine was 3 years of follow-up in HPV-6, HPV-11, HPV-16, and HPV-18
used in the control arm. infection in patients receiving the HPV vaccine versus placebo,502
Collectively, these studies have demonstrated that in women with and a 96% decrease in persistent HPV-6, HPV-11, HPV-16, and
no baseline evidence of HPV-16/18 infection, the final efficacy of the HPV-18 infection with 5 years of follow-up.503 The 5-year follow-up
HPV-16/18 vaccine in reducing (6 or 12 month) persistent HPV-16/18 figures also showed that the HPV vaccine conferred 100% preventive
infection is 90.9% to 100% in women 15 to 25 years of age, and efficacy for precancerous cervical dysplasia and genital warts. These
that the interim efficacy for preventing 6-month persistent HPV-16/18 two studies (Protocols 005 and 007) tested slightly different dosage
infection is 77.4% to 93.7% in women older than 25 years (the regimens. Protocol 005 administered vaccinations at months 0, 1,
ongoing VIVIANE study).497 The prevention of cytologic abnormalities and 6 using 0.5 mL of qHPV vaccine (20 µg of HPV-6, 40 µg of
associated with HPV-16/18 infection ranged from 80.6% to 97.1%, HPV-11, 40 µg of HPV-16, and 20 µg of HPV-18) or one of two
and the prevention of CIN ranged from 80.0% to 100% in girls and placebos (225 µg or 450 µg of aluminum adjuvant). Protocol 007
women 15 to 25 years of age and 75.5% to 100% in women older administered vaccinations at months 0, 2, and 6 using low (identical
than 25 years. The HPV-16/18 vaccine proved to be cross-protective to the Protocol 005 vaccine: 20 µg each of HPV-6 and HPV-18, and
at varying levels against infection with other HPV subtypes, including 40 µg each of HPV-11 and HPV-16), medium (40 µg each of HPV-6,
HPV types 31, 33, 35, 39, 45, 51, 52, 56, 58, 59, and 68 and against HPV-11, HPV-16, HPV-18), and high (80 µg each of HPV-6, HPV-11,
CIN associated with these subtypes. Preventive efficacy against the and HPV-18, and 40 µg of HPV-16) doses of the vaccine versus
HPV subtypes that account for most genital warts was reported to one of two placebos. Whereas Protocol 007 demonstrated increased
be moderate for HPV-6 and HPV-11 (34.5%) and HPV-74 (49.5%).483 participants with adverse events in the high-dose group, no serious
In addition, the HPV-16/18 vaccine was effective at preventing oral adverse events relating to the vaccine were reported.502,512 Although
(93.3% [95% CI, 62.5–99.7]),491 anal (83.6% for women HPV-16/18 toxicity did not exclude any of the vaccine doses from moving forward
negative at baseline who received three doses [95% CI, 66.7–92.8]),490 to phase III testing, the comparable efficacy observed in patients
and vulvar (54.1% [95% CI, 4.9–79.1])489 HPV-16/18 infection. treated with the lower and higher doses resulted in selection of the
These preventive effects after administration of the HPV-16/18 vaccine low dose for further development.511
have been demonstrated in all women, independent of age, geographic The phase III FUTURE I trial (n = 5455; conducted in Asia,
location, or sexual experience.497 Australia, Europe, and Latin, North, and South America) and FUTURE
Kreimer and colleagues conducted a combined analysis of the II trial (n = 12,167; conducted in Asia, Europe, and North and South
PATRICIA and CVT trials to compare the effectiveness of one, two, America) enrolled girls and women 16 to 23 years of age for a three-dose
366 Part I: Science and Clinical Oncology

regimen of vaccine or placebo. These studies demonstrated 100% immunobridging efficacy for boys and men 9 to 26 years of age and
efficacy of the vaccine in preventing HPV-16/18 infection–related girls and women 9 to 15 years of age.
CIN grades 2 and 3, adenocarcinoma in situ, and vulvar and vaginal The potential impact of the Gardasil 9 vaccine on increasing the
intraepithelial neoplasia grade 1.515,516 As seen with the bivalent vaccine, prevention of HPV infection–related cervical cancer beyond the
the qHPV vaccine was also shown to be cross-protective by conferring quadrivalent Gardasil and Cervarix vaccines varies globally but is
30%, 48%, and 75% vaccine efficacy against any lesion for cervical, estimated to be 8% higher in Australia (preventing 86.5% of cases),
vaginal, and vulvar intraepithelial neoplasia grade 1, respectively, as 13% in North America and Europe (preventing 92% and 90.9% of
well as 83% (condyloma) preventive efficacy regardless of HPV subtype. cases, respectively), and 18.4% worldwide (preventing 89.9% of
Olsson and colleagues tested three doses of the qHPV vaccine or cases).467,529 Thus, whereas the bivalent and quadrivalent vaccines both
placebo in 552 girls and women 16 to 23 years of age, with 3 years confer protection against approximately 70% of the cancerous and
of follow-up.504 In addition, 241 of the subjects were included in an precancerous cervical conditions associated with HPV infection, the
additional 2 years of follow-up (totaling 5 years of follow-up). This 9vHPV vaccine increases this to roughly 90%.530 However, it is possible
study did not show increased adverse events in the vaccine and placebo that broad-scale vaccination may result in a shift in prevalence of the
arms. In addition, a high level of efficacy and a maintenance of anti- HPV genotypes500; in fact, one study has reported increased prevalence
HPV levels were observed for at least 5 years, as was the induction of HPV subtypes 51, 59, 73, 84, and 89 in school-aged girls vaccinated
of a strong immune memory, suggesting long-term efficacy. Additional with the quadrivalent vaccine through the Australian national vaccina-
post–FDA approval studies have further supported the data provided tion program versus unvaccinated girls.531,532
in these studies, some of which are still ongoing. Future results of epidemiologic data, including the long-term
follow-up results of the studies highlighted earlier, will be critical in
9vHPV (Gardasil 9) better defining the prevalence of HPV infection and the diseases
Gardasil 9, developed by Merck through modification of the quadri- associated with HPV infection, and the effective duration of prevention
valent Gardasil vaccine, received FDA approval in 2014 for the preven- associated with all three HPV vaccines.497 In addition, the global level
tion of (1) HPV type 16, 18, 31, 33, 45, 52, and 58 infection–related of vaccine uptake, the affordability (these vaccines currently cost
cervical, vulvar, vaginal, and anal cancers; (2) HPV type 6, 11, 16, approximately $120 per dose, with three doses recommended500),
18, 31, 33, 45, 52, and 58 infection–related cervical, vaginal, vulvar, global availability, and coverage will all play significant roles in the
and anal intraepithelial neoplasia and cervical adenocarcinoma in situ; ultimate impact of these vaccines on HPV-related diseases and out-
and (3) HPV-6 and HPV-11 infection–related genital warts.517 This comes.528 However, with the prevalence of HPV infection worldwide,
approval is for girls and women 9 to 26 years of age, and applies to although educated modifications in sexual behavior could significantly
boys and men 9 to 26 years of age only for the previously described affect the risk of HPV infection, immunization with the HPV vaccine,
prevention of anal cancer, anal intraepithelial neoplasia, and genital HPV testing, and cervical cancer screening with the Pap test are likely
warts. Gardasil 9 contains 30 µg of HPV-6 capsid protein, 40 µg of to remain critical components for the continued prevention of cervical
both HPV-11 and HPV-18 capsid protein, 60 µg of HPV-16 capsid cancer. Furthermore, despite HPV’s status as the leading sexually
protein, and 20 µg each of HPV-31, HPV-33, HPV-45, HPV-52, transmitted disease in the United States, only 40% of girls 13 to 17
and HPV-58 capsid protein.467 years of age in the United States completed the three-dose HPV series
Eight clinical studies have published results regarding the efficacy, in 2014, which was a slight increase from the 37% who completed
immunogenicity, and toxicity of Gardasil 9.513,518–525 Collectively, these the series in 2013.533
studies demonstrate that Gardasil 9 induces a strong immune response The Advisory Committee on Immunization Practices (ACIP)
and is associated with an almost 100% seroconversion rate after the updated its recommendations for HPV vaccination in December 2016
third dose.467 Maximal benefit in terms of immune response for the to include the nonavalent vaccine.534 The current recommendations
five high-risk HPV subtypes uniquely in Gardasil 9 (HPV types 31, are to vaccinate with two doses (0 and 6–12 months) at age 11 to
33, 45, 52, and 58) appears to be in naïve populations rather than 12, and vaccination can be started at age 9. The ACIP recommends
individuals previously vaccinated with the quadrivalent Gardasil; that girls and women through age 26 and boys and men through age
conversely, lower immune response for all four HPV subtypes in the 21, as well as some high-risk boys and men through age 26, be vac-
quadrivalent vaccine (HPV-6 and HPV-11 and high-risk HPV-16 cinated if they did not previously undergo adequate vaccination (defined
and HPV-18) has been higher in individuals previously vaccinated as initiating vaccination [1] before their 15th birthday and receiving
with the quadrivalent vaccine.467,513,519 Toxicity is minimal, but inci- the two-dose, at 0 and 6–12 months, or three-dose, at 0, 1–2, and 6
dences of local skin reactions have been higher than those associated months, regimens; or [2] on or after their 15th birthday and receiving
with the quadrivalent Gardasil vaccine.519,523 Skin reactions and three doses at 0, 1–2, and 6 months). The three-dose schedule is
headaches have comprised the majority of reported adverse events. recommended for individuals who are immunocompromised. The
Nonetheless, although the HPV vaccinations are being integrated into ACIP does not have a recommendation regarding additional vaccination
the national health programs of more and more countries around the with the nonavalent HPV vaccine for individuals previously adequately
world, Japan withdrew its recommendation for the HPV vaccine vaccinated with the bivalent or quadrivalent vaccines. The ACIP also
following a fearful national response to highly publicized cases of does not recommend restarting interrupted vaccination schedules.
adverse events (complex regional pain syndrome).500,526 In addition,
four cases of premature ovarian failure have been reported to the Prostate Cancer
CDC; these may have been due to an HPV vaccine–triggered auto-
immune or inflammatory response.500,527 Both epidemiologic and experimental data have supported the develop-
Signorelli and colleagues conducted a systematic review of the 16 ment of a broad range of dietary supplements and drug-based interven-
RCTs on 9vHPV listed on RCT registries, eight of which they included tions for prostate cancer prevention. Although some of these have
in their analysis.528 In addition to showing a noninferior immune now been shown to be associated with increased risk of prostate cancer
response following administration of the 9vHPV vaccine compared (e.g., vitamin D),535 numerous clinical trials testing these agents as
with the quadrivalent HPV vaccine for the four HPV subtypes in cancer preventive interventions, including studies of calcium, selenium,
both vaccines (HPV-6, HPV-11, HPV-16, and HPV-18), they found vitamin E, and 5α-reductase inhibitors, have been conducted.
that the efficacy of the 9vHPV vaccine in preventing high-grade cervical,
vulvar, or vaginal diseases associated with the five additional high-risk 5α-Reductase Inhibitors
HPV subtypes was 96.7% for girls and women 16 to 26 years of The conversion of testosterone into 5-dihydrotestosterone, which has
age (95% CI, 80.9–99.8).528 This analysis also demonstrated an stronger androgenic activity, requires the enzyme 5α-reductase.
Lifestyle and Cancer Prevention  •  CHAPTER 22 367

Table 22.11  Prostate Cancer Large Scale (Phase III) Chemoprevention Studies With Outcomes
PROSTATE CANCER
Trial Patients Agent Dose Relative Risk (RR) Absolute Risk (AR) AR: High Grade Reference
PCPT 18,882 Finasteride 5 mg/day 25% decrease 6% decrease 0.6% increase Thompson et al 2003536
REDUCE 8231 Dutasteride 0.5 mg/day 23% decrease 5% decrease ≤0.5 % increase Andriole et al 2010538
SELECT 35,533 Selenium 200 µg/day 9% increase 0.8% increase No known effect Klein et al 2011173
Vitamin E 400 IU/day 17% increase 1.6% increase No known effect
Selenium + 200 µg/day 5% increase 0.4% increase No known effect
vitamin E 400 IU/day
400 IU/day

Antiandrogenic agents that block this conversion by inhibiting hyperplasia in the dutasteride versus the placebo treatment groups
5α-reductase, including finasteride and dutasteride, have been the (OR, 0.47 [95% CI, 0.37–0.59]; P < .001).
focus of several large-scale phase III trials (Table 22.11). Ultimately, the findings from both the PCPT and REDUCE trials,
demonstrating that both finasteride and dutasteride reduce incidence
Prostate Cancer Prevention Trial (PCPT) of low-grade prostate cancer but did not reduce the incidence of
The first large phase III trial for prostate prevention was the PCPT, high-grade prostate cancer, prevented the FDA approval of these
which tested 7 years of treatment with finasteride (5 mg/day) versus 5α-reductase inhibitors as preventive agents for prostate cancer.541
placebo in 18,882 men 55 years of age or older with normal prostate-
specific antigen (PSA) levels (≤3.0 ng/mL) and digital rectal examination Selenium and Vitamin E
findings.536 Incidence of prostate cancer was reduced by 24.8% in Based on positive results of epidemiologic studies and secondary analyses
patients treated with finasteride compared with placebo. However, of randomized trials demonstrating decreased risk of prostate cancer
finasteride was also associated with a 0.6% increase in high-grade risk with selenium and vitamin E, SELECT was developed and
prostate cancer (defined as a Gleason score of 8–10). This increased conducted.172 A total of 35,533 men aged 50 years (black) or 55 years
risk of high-grade prostate cancer appears to be due to an increase in (all others) or older with a PSA of 4.0 ng/mL or lower and normal
detection after finasteride-mediated reduction in prostate size. rectal examination findings in Canada, Puerto Rico, and the United
These results were further clarified by the 18-year follow-up figures, States were randomized to receive treatment with selenium (200 µg/
which demonstrated a 30% decrease in risk of prostate cancer in day of l-selenomethionine) and/or vitamin E (400 IU/day of all-rac-
patients treated with finasteride versus placebo (RR, 1.17 [95% CI, α-tocopherol acetate) or placebo with a follow-up of 7 to 12 years.
0.65–0.76]; P < .001).537 Increased detection of high-grade prostate Interim results after a median of 5.5 years of follow-up revealed increased
cancer (defined as a Gleason score of 7–10) was also seen with the risk of prostate cancer in patients treated with selenium (HR, 1.04
extended follow-up (RR, 1.17 [95% CI, 1.00–1.37]; P = .05). Neither [95% CI, 0.87–1.24]), vitamin E (HR, 1.13 [95% CI, 0.95–1.35]),
overall survival nor survival after diagnosis of prostate cancer was and selenium plus vitamin E (HR, 1.05 [95% CI, 0.88–1.25]). These
significantly different between treatment arms. data suggested no preventive efficacy of selenium and/or vitamin E
supplementation for prevention of prostate cancer.
Reduction by Dutasteride of Prostate Cancer Events Results after a total of 7 years of follow-up indicated a slightly
(REDUCE) trial increased and statistically significant risk compared with the initial
The REDUCE trial compared the efficacy of 4 years of treatment data, revealing that vitamin E (HR, 1.17 [95% CI, 1.004–1.36]; P
with dutasteride (0.5 mg/day) versus placebo for the prevention = .008]) increased risk of prostate cancer versus placebo.173 These
of prostate cancer in 6729 men 50 to 75 years of age with a PSA findings demonstrate an absolute increased risk of prostate cancer in
of 2.5 to 10.0 ng/mL and a negative prostate biopsy finding.538 A healthy men of 1.6 per 1000 person-years for vitamin E.
22.8% reduction in risk of prostate cancer was demonstrated with As with the PCPT, an analysis of a subset of 4934 SELECT
dutasteride compared with placebo (95% CI, 15.2–29.8; P < .001). participants demonstrated an association between vitamin D and
Dutasteride was also shown to reduce the rate of both acute urinary prostate cancer risk.542 This study identified increased risk of prostate
retention (by 77.3%) and adverse events related to benign prostatic cancer with both low and high levels of vitamin D but did show
hyperplasia. However, as with finasteride, increased detection of high- reduced risk of high-grade prostate cancer (Gleason score of 7–10)
grade prostate cancer (Gleason score of 8–10) was associated with in African American men with higher vitamin D concentrations.
dutasteride in years 3 and 4 of treatment (likely because of a shrinkage Ultimately, the results of the SELECT trial suggest that neither
of the prostate gland, facilitating the detection of these high-grade vitamin E nor selenium will reduce the risk of prostate cancer and
prostate cancers). that exceeding the recommended dietary allowance (RDA) of either
A follow-up study examined incidence of newly diagnosed prostate of these supplements could, in fact, increase risk in men older than
cancers in 2715 of the men included in the REDUCE trial during 55 years.543
the 2 years following the 4-year trial described earlier.539 The results
from this study identified no additional high-grade cancers and Aspirin
demonstrated a similar low rate of new prostate cancer diagnoses Several studies are currently testing aspirin alone or in combination
between the two treatment arms. treatment for the prevention of prostate cancer recurrence. Powles and
Another subset of 1617 asymptomatic men with enlarged prostates Cuzick of Queen Mary University of London and their colleagues
in the REDUCE study were included in a post hoc analysis that are conducting a multiinstitutional phase II/III study in the United
investigated the effects of dutasteride on risk of clinical progression Kingdom testing aspirin (100 or 300 mg/day) with or without vitamin
of benign prostatic hyperplasia.540 This study demonstrated a 41% D (4,000 IU/day; Vigantol Oil) or placebo for the prevention of
reduction in RR and a 15% reduction in absolute risk. Furthermore, progression of prostate cancer, with prevention of new lesions as one
a multivariable regression analysis adjusted for covariates demonstrated of the secondary end points (NCT03103152). This study, which
a significant reduction in clinical progression of benign prostatic should be complete in October 2019, is currently recruiting, with
368 Part I: Science and Clinical Oncology

a planned accrual of 104 men 16 to 100 years of age with prostate lesions, and valrubicin has been shown to be effective in patients with
cancer and an estimated life expectancy of over 3 years. Lu at West bladder carcinoma in situ that is refractory to BCG.551
China Hospital is currently recruiting participants for a phase IV
study of aspirin (100 mg QD), the antibiotic levofloxacin (0.5 g QD), Bacillus Calmette-Guérin
both, or neither for the prevention of prostate cancer occurrence Since 1977, post–transurethral resection (TUR) intravesicular treatment
or disease progression (NCT02757365). A total of 100 men aged with BCG has been the standard of care for individuals at high risk
50 to 70 years with benign prostate hyperplasia with lymphocyte for NMIBC. Pinsky and colleagues showed that treatment with standard
infiltration are to be recruited, and the study should be completed therapy (cystoscopy with fulguration) plus 6 weeks of BCG treatment
in April 2019. Finally, the large-scale phase III Add-Aspirin clinical significantly delayed disease progression compared with standard therapy
trial under the direction of Langley at University College, London, alone in high-risk patients (P < .003).552 Long-term follow-up confirmed
is investigating the efficacy of aspirin in preventing recurrence and these findings by demonstrating disease progression in 42% fewer
improving survival in individuals with nonmetastatic breast, prostate, BCG-treated patients than control patients.553 The most recent results
colorectal, or gastroesophageal cancer after standard curative therapy showed 62% and 37% progression-free rates in the BCG plus TUR
(NCT2804815). Accrual is currently underway, with a goal of arm and the control arm (TUR), respectively, and 75% and 55%
11,000 participants, who will be randomized in cancer type–specific 10-year disease-specific survival rates between the experimental and
parallel cohorts to receive aspirin (100 or 300 mg/day) or placebo control arms, respectively (P = .03).554
for 5 years. This study is not scheduled to be completed until Lamm and colleagues conducted a Southwest Oncology Group
October 2026. (SWOG) study of intravesical and percutaneous BCG immunotherapy
A pooled analysis of the National Institutes of Health (NIH)–AARP of BCG induction alone or followed by BCG maintenance for 3
Diet and Health Study (composed of 566,398 US AARP members weeks administered at months 3, 6, 12, 18, 24, and 30.555 They found
aged 50–71 years) and the Prostate, Lung, Colorectal and Ovarian that BCG maintenance therapy significantly increased the efficacy
(PLCO) Cancer Screening Trial (composed of 76,685 and 78,216 US (twofold) of BCG induction therapy alone for recurrence-free survival
men and women, respectively, aged 55–74 years) examined whether (log rank P < .0001). Their results also identified significantly longer
aspirin and other NSAIDs increase survival in men diagnosed with worsening-free survival within the patients receiving BCG maintenance.
prostate cancer.544 Included in this analysis were 221,189 participants Based on these findings, a regimen of 3 years of BCG maintenance
of the NIH-AARP study who responded to a Risk Factor Question- was established.
naire and the male PLCO participants. Although prediagnosis and After these studies, the most effective regimen of inductive and
postdiagnosis use of aspirin and nonaspirin NSAIDs did not affect maintenance BCG therapy has been controversial and the focus of a
prostate cancer–specific mortality, participants with 5 or more years number of clinical trials.556 A variety of doses (e.g., one-third and full
of occasional (defined as less than daily) use of aspirin before diag- dose) of intravesical inductive and maintenance BCG therapy regimens
nosis of prostate cancer demonstrated an 18% reduction in all-cause have been investigated for inductive therapy alone or followed by
mortality (HR, 0.82 [95% CI, 0.75–0.90]), whereas daily aspirin maintenance therapy for timeframes ranging from 1 to 3 years.
users for the 5 years before diagnosis demonstrated a 15% reduction Because of the toxicity-based issues resulting in interruptions
in all-cause mortality (HR, 0.85 [95% CI, 0.66–0.86]) compared and premature discontinuation of BCG maintenance therapy, the
with participants who did not use aspirin during that timeframe. European Organisation for Research and Treatment of Cancer (EORTC)
Thus, although aspirin did not improve prostate cancer–specific conducted a four-arm study of one-third-dose versus full-dose BCG
mortality, long-term or daily or occasional use of aspirin may maintenance therapy for 1 and 3 years.557 A total of 1355 patients
improve overall survival by preventing comorbidity in this patient with intermediate- and high-risk NMIBC were randomized after
population. TUR. Suboptimal efficacy was observed with one-third-dose versus
full-dose treatment for 1 year (HR, 0.75 [95% CI, 0.59–0.94]; P =
Other Agents .01). There was no additional benefit for intermediate-risk patients
A cohort study of a subset of 9457 PCPT participants with 7 years to receive more than 1 year of BCG maintenance therapy. However,
of follow-up showed that statin use is not associated with risk of 3 years of full-dose (but not one-third-dose) BCG maintenance
total, low-grade, or high-grade prostate cancer.545 However, in a therapy did reduce recurrence in high-risk patients (HR, 1.61 [95%
nested case-control analysis of 3377 PCPT participants, higher CI, 1.13–2.30]; P = .009). No difference was observed in tumor
serum 25-hydroxyvitamin D (25[OH]D) levels were associated with recurrence or death in high-risk patients treated for 1 compared
a modest increase in risk of Gleason 2 to 6 disease, but a substantial with 3 years.
reduction in risk of Gleason 8 to 10 prostate cancer, particularly in The French Urological Association Cancer Committee also con-
men 65 years of age or older.546 Preclinical studies of lower doses ducted a phase III multicenter study, titled URO BCG 4, that random-
of some agents (e.g., vitamin D3 in combination form with the soy ized 146 patients with intermediate- or high-risk NMIBC from nine
isoflavone genistein) have been shown to be associated with inhibi- hospitals, in order to investigate the effects of lower dose and fewer
tion of prostate cancer cells,547 although different expression levels instillations of BCG maintenance for the treatment for NMIBC.558
and polymorphisms in the vitamin D receptor may affect preventive Patients were randomized to receive one-third dose of BCG for 3
efficacy.548 The most recent results of the PCPT (2017) further assessed consecutive weeks every 3 (group 1) or 6 (group 2) months. Tumor
the association between vitamin D and prostate cancer risk previously recurrence, muscle invasion, and side effects after 6, 12, and 18 months
observed by Miles and colleagues in 2014.549 This nested case-control of treatment were not significantly different between the two treatment
study of 3387 participants from the PCPT found that above-median groups. Updated figures after 3 years of BCG maintenance therapy
serum 25(OH)D levels in men with higher circulating IGF-2 levels were published in 2017 and revealed 18% tumor recurrence in group
were associated with increased risk of prostate cancer (OR, 1.33 1 and 10% recurrence in group 2 (P = .241). Rates of adverse events
[95% CI, 1.00–1.65]). were slightly lower in group 1 than in group 2 (P = .037), with group
2 accounting for over three times as many major adverse events, which
Bladder Cancer suggests that lower toxicity is associated with the 6-month instillation
schedule.559 However, the most recent meta-analysis of nine of these
Non–muscle-invasive bladder cancer (NMIBC) accounts for 75% to studies reported that extended BCG maintenance therapy (e.g., 3
85% of all bladder cancer diagnoses at presentation and is confined years) is not more effective than shorter-term therapy (e.g., 1 year)
to the mucosa or submucosa.550 Bacillus Calmette-Guérin (BCG) was in decreasing recurrence, but that 3 years of therapy does not increase
initially developed as an adjuvant treatment for patients with preinvasive the incidence of side effects.560
Lifestyle and Cancer Prevention  •  CHAPTER 22 369

Over the last 20 years, several studies have shown BCG to be more receive a 6-week BCG induction course followed by 3 years of
effective than chemotherapy when compared with agents such as maintenance BCG.
doxorubicin and mitomycin C. Induction and maintenance BCG
therapy has also been shown to be significantly more effective than Valrubicin
treatment with epirubicin plus IFN-α2a in both prevention of recur- Although gemcitabine and mitomycin C (with or without hypothermia)
rence (HR, 0.41 [95% CI, 0.28–0.60]; P < .001) and disease-specific have been studied for patients with post-BCG recurrence who were
mortality (HR, 0.20 [95% CI, 0.04–0.91]; P = .04). A meta-analysis unable to undergo cystectomy,574 valrubicin was approved for this
of randomized trials by Shelley and colleagues identified significantly application by the FDA in 1998 and is currently the only drug other
decreased tumor recurrence in high-risk patients treated with BCG than BCG that has received FDA approval for NMIBC.575,576 Steinberg
versus mitomycin C (P < .001).561 Similarly, Böhle and Bock reported and colleagues of the Valrubicin Study Group conducted an open-label,
significantly higher efficacy with BCG maintenance over mitomycin noncomparative study that tested valrubicin in patients with BCG-
C (OR, 0.66 [95% CI, 0.47–0.94]; P = .02).562 However, whereas refractory carcinoma in situ of the bladder.551 Before recruitment, two
intravesicular BCG therapy is more effective than chemotherapy for or more rounds of BCG were administered to 70% of the study
both complete response and disease-free survival, the difference in participants, and one round of BCG followed by one or more rounds
effect on overall survival remains to be determined.563 More recent of another agent were administered to 30% of the study participants.
evidence suggests that BCG should be the standard of care for both In this multiinstitutional trial, intravesical valrubicin (800 mg/wk)
intermediate- and high-risk NMIBC.564 was administered to 90 patients for 6 weeks. Valrubicin was found
Given that recurrence occurs in 30% to 45% of individuals treated to be well tolerated, and after 30 months of follow-up, 21% of the
with intravesicular BCG,565 several phase II trials have attempted to patients were disease-free. Only 2 of the 79 patients who demonstrated
further decrease recurrence by using BCG in combination with other recurrence had advanced tumors (stage T2). Reversible local bladder
agents, including chemotherapeutic agents and IFN.566,567 To date, symptoms accounted for the majority of the reported side effects. In
none of these strategies have proved to significantly improve the preven- 2008, the number of total responders was updated, lowering slightly
tion of recurrence over treatment with BCG alone.568,569 from 21% to 18%.577 In addition, recurrence figures were updated
Two strains of BCG, Connaught and TICE, have been the focus to show that 10 patients developed metastatic or deeply invasive bladder
of several studies to ascertain whether they confer a disparate level of cancer, including four deaths and six cases of stage T3 bladder cancer.
efficacy in preventing recurrence in patients with NMIBC. A retrospec- Steinberg followed this study with the phase II/III A9303 trial,
tive comparison of BCG Connaught and BCG TICE demonstrated another open-label trial, testing 6 versus 9 weeks of valrubicin (800 mg/
that without maintenance therapy Connaught is more effective than wk) in 80 patients with BCG-refractory carcinoma in situ of the
TICE in delaying time to first occurrence (HR, 1.48 [95% CI, bladder who had previously received BCG treatment or two or more
1.20–1.82]; P < .001), but that with maintenance therapy TICE is (39%) or three or more (11%) courses of BCG.576 A total of 78
more effective than Connaught in delaying time to first occurrence patients completed treatment and underwent primary disease evaluation
(HR, 0.66 [95% CI, 0.47–0.93]; P = .019). Conversely, Steinberg at 3 months. The complete response for this study, as with the previous
and colleagues conducted a post hoc analysis of their phase II study study, was 18%. Reversible local bladder adverse events were primarily
testing the BCG Tice and Connaught strains with IFN.570 They found mild to moderate.
that in 901 patients with high-grade NMIBC treated with six weekly
intravesicular BCG instillations and IFN (50 million units), the strain Chemotherapy
of BCG administered did not significant affect recurrence-free survival. Sternberg and colleagues conducted a study of the deoxycytidine
Currently underway is a phase III study that is investigating different gemcitabine in high-risk BCG-refractory NMIBC patients who were
strains of BCG with or without vaccine in patients with high-grade unable or unwilling to undergo cystectomy and found it to be active.578
NMIBC (NCT03091660). Patients will be randomized to receive However, a second phase II trial (SWOG S0353) revealed that although
induction and maintenance therapy with TICE BCG (arm 1), intravesical gemcitabine had activity (2 g in 100 mL of normal saline
Tokyo-172 strain BCG (arm 2), or Tokyo strain BCG with priming weekly for 6 weeks, then monthly to 12 months), only 47% of patients
(vaccine; arm 3). Five-year recurrence and disease-free rates will be with NMIBC were disease free at 3 months, and fewer than 30%
determined for an estimated 969 participants, with a study completion and approximately 20% demonstrated a durable response at 12 and
date of 2025. 24 months, respectively.579 Shelley and colleagues conducted a systematic
The most recent European Association of Urology (EAU) guidelines review of gemcitabine for the prevention or delay of rumor recurrence
for NMIBC were published in 2017.571 They recommend that both after resection in NMIBC patients.580 They found that gemcitabine
patients with low-risk tumors and patients at intermediate risk receive was more effective than mitomycin C in preventing recurrence, and
treatment with one instillation of chemotherapy immediately, and showed potential for patients with BCG refractory NMIBC. However,
that intermediate-risk patients then receive either intravesicular they did note that additional corroborative evidence is needed to
immunotherapy treatment with BCG at full dose for 1 year (level A) confirm these findings.
or a maximum of 1 year of chemotherapy (level B). TUR is recom- Two additional studies of gemcitabine for the prevention of recur-
mended for low-, intermediate-, and high-risk patients. In addition, rence in patients with NMIBC are currently recruiting subjects. A phase
the EAU recommends that intravesicular BCG be administered at II trial is examining gemcitabine plus cisplatin versus no intervention
full dose for 1 to 3 years in individuals with high-risk tumors, and in moderate- to high-risk NMIBC patients who underwent TURBT
that re-TUR should be considered for individuals at highest risk of and had epirubicin instilled 24 hours afterward (NCT02716961);
progression and/or with BCG refractory tumors. The American this trial is scheduled to be completed in 2020. A phase III trial
Urological Association (AUA) updated its recommendations for NMIBC comparing the efficacy of intravesicular gemcitabine (2 g in 100 mL
in 2016,572 and whereas TUR is similarly recommended for low-, of 0.9% sodium chloride) versus intravesicular mitomycin C (40 mg
intermediate-, and high-risk patients, the remaining guidelines vary in 40 mL of 0.9% sodium chloride) versus no intervention in
somewhat from those of the EUA.573 The AUA guidelines now recom- NMIBC patients immediately after TURBT is currently ongoing
mend that one instillation of intravesical chemotherapy (mitomycin (NCT02685771).
C or epirubicin) be considered for individuals with low- or intermediate- A phase II study of pirarubicin for prevention of recurrence after
risk tumors within 24 hours of transurethral resection of bladder nephroureterectomy for upper tract urothelial carcinoma (UTUC) is
tumor (TURBT). They recommend that intermediate-risk patients being conducted by Huang at Renji Hospital (NCT03030157). This
receive 6 weeks of induction chemotherapy or immunotherapy (BCG) trial is comparing long-term versus single intravesical instillation of
and that maintenance therapy be considered, and that high-risk patients pirarubicin (THP; 30 mg in 30 mL of normal saline). The study,
370 Part I: Science and Clinical Oncology

scheduled to be completed in December 2019, has begun accru-


ing, with a goal of 220 participants. Another new phase II study Other Drugs
investigating the effects of prophylactic intravesical chemotherapy of A meta-analysis of randomized controlled clinical trials analyzed the
pharmorubicin versus pirarubicin on the prevention of bladder tumors preventive efficacy of vitamin and antioxidant supplements for bladder
in patients who have undergone nephroureterectomy for UTUC is being cancer, including vitamins A, B6, C, and E, a multivitamin, β-carotene,
conducted by Li at Peking University First Hospital (NCT02547350). fenretinide, selenium, α-tocopherol, and zinc.589 This study analyzed
This trial, which has not yet opened for recruitment, is scheduled 14 clinical trials and found no preventive benefit for risk of bladder
to randomize 200 patients to receive a single or multiple intravesical cancer (RR, 1.04 [95% CI, 0.92–1.17]) associated with any of the
instillations of pharmorubicin (50 mg) or pirarubicin (30 mg). The vitamin and antioxidant supplements, or combinations thereof, and
study is to be completed by 2020. Li has a second study currently a marginal increase in risk of bladder cancer with β-carotene (RR,
accruing participants to receive a single intravesical chemotherapy 1.44 [95% CI, 1.00–2.09]).
instillation of pirarubicin (THP; 40 mg for 30 minutes) immediately Molecular targets for immune-based therapies are currently being
after diagnostic ureteroscopy for UTUC. This study is scheduled to be investigated, including ILs, IFN, TLRs, and TRAIL.565 In addition,
completed by 2021. nanoparticle delivery of BCG cell wall skeleton (BCG-CW) to replace
live BCG and the use of gene and virus therapy are in the early stages
Celecoxib of development.
An Italian study focused on celecoxib for the prevention of recurrence
in NMIBC patients.581 A total of 58 patients were randomized to Skin Cancer
receive celecoxib or intravesical mitomycin C weekly for 4 weeks,
then monthly for 11 months. The results demonstrated improved Nonmelanoma Skin Cancers
efficacy associated with celecoxib versus chemotherapy after 75 months The progression of cutaneous squamous cell carcinoma (cSCC),
of follow-up, evidenced by increased disease-free patients (mitomycin the most prevalent metastatic skin cancer,590–592 is associated with
C, 34.85%; celecoxib, 44.8%). inflammation and has been well characterized, initially manifesting
as a precancerous lesion known as actinic keratosis (AK), that then
Metformin progresses to a cSCC in situ before progressing into an invasive
With the benefits metformin has been shown to have in reducing carcinoma.593 Thus, disruption of the progression of AK to invasive
risk of cancer, including breast, colorectal, pancreatic, and prostate carcinoma represents a strategy with high potential for the prevention of
cancers, it has become the focus of studies to prevent recurrence skin cancer.
in NMIBC patients.582 A meta-analysis of 36 studies found that A variety of strategies have been developed to prevent AK and
patients with diabetes mellitus have a 35% increased risk of bladder superficial BCC progression, including cryosurgery,594 photodynamic
cancer (RR, 1 in 35 [95% CI, 1.17–1.56]; P < .001), with highest therapy,595 electrodissection, curettage, and chemical peels.596 Applica-
risk occurring in patients diagnosed within less than 5 years.583 Seng tion of the FDA-approved topical agents 5-fluorouracil (5-FU) cream
investigated the effects of metformin on risk of bladder cancer in (a cytotoxic agent), diclofenac gel (an NSAID), imiquimod cream
970,708 Taiwanese patients with type 2 diabetes mellitus, including (an immune response modifier), and ingenol mebutate gel (a cell
532,519 never users and 408,189 ever users of metformin.584 He found death inducer) and phytodynamic therapy with δ-aminolevulinic
decreased bladder cancer risk in patients treated with metformin. acid (ALA-PDT; an endogenous 5-carbon aminoketone that is
These findings were reinforced by a retrospective cohort study of the the first compound in the porphyrin synthesis pathway) have
effects of metformin on cancer-specific survival in patients diagnosed been shown to clear AKs and thereby prevent skin cancer, with
with bladder cancer who were undergoing radical cystectomy.585 Of comparable efficacy in phase II trials, although the adverse effects
the 421 patients, 85 had diabetes; 39 of the 85 (46%) were being and extent of cosmesis associated with treatment vary by agent.597
treated with metformin. Metformin improved both recurrence-free The majority of cases of AK and superficial BCC are managed with
survival (HR, 0.54 [95% CI, 0.33–0.88]; P = .013) and bladder cryosurgery because of the low cost, ease of use, acceptable cosmesis,
cancer–specific survival (HR, 0.57 [95% CI, 0.35–0.91]; P = .019). and low rate of recurrence (potentially 7.5%, depending on the
Another retrospective analysis investigating the association of diabetes lesions) versus electrodissection and curettage (7.7%), all non-Mohs
mellitus treated with metformin and oncologic outcomes was conducted modalities (8.7%), radiation therapy (8.7%), and surgical excision
by Rieken and colleagues.586 Of 1117 patients with NMIBC, 125 (10.1%).594
had diabetes mellitus, 43 of whom (3.8% of 1117 patients) were Randomized trials have demonstrated that treatment of AKs,
being treated with metformin. Once again, these researchers found Bowen disease, and both superficial and thin nodular BCC with
increased risk of recurrence (HR, 1.45 [95% CI, 1.09–1.94]; P = .01) δ-aminolevulinic acid phytodynamic therapy is both effective in reduc-
and progression (HR, 2.38 [95% CI, 1.40–4.06]; P = .001), but not ing recurrence and providing superior cosmetic outcomes versus standard
any-cause mortality, in diabetic patients not taking metformin. Finally, therapies.595 Unfortunately, results from long-term follow-up studies
Zhang and colleagues showed in preclinical studies that metformin have reported that although recurrence rates in individuals treated with
suppresses bladder cancer cell proliferation.587 Unfortunately, although δ-aminolevulinic acid phytodynamic therapy are comparable to those
metformin is currently being studied in 33 cancer prevention trials, of other standard therapies for both Bowen disease and superficial
none of these are investigating its efficacy in preventing recurrence of BCC, sustained efficacy is lower in individuals with nodular BCC
bladder cancer. versus surgery.595 5-FU cream (5% twice daily for 2–4 weeks) is the
standard-of-care treatment for AKs, and field therapy with this agent
Toll-Like Receptor Agonist has been shown to be effective for treating AKs.598 Results published in
Based on positive results observed with the TLR agonist imiquimod for 2017 from an RCT titled the Veterans Affairs Keratinocyte Carcinoma
benign and malignant skin cancer in a phase I clinical trial, Donin and Chemoprevention (VAKCC) Trial demonstrated a significant reduction
colleagues studied the efficacy of six doses of intravesical TMX-101, in the development of new AKs after treatment with 5-FU (5% twice
a liquid version of imiquimod, in a phase II pilot study.588 Their daily for 2 to 4 weeks) versus control that persisted for 24 to 36 months
results demonstrate that TMX-101 0.4% (200 mg/50 mL) is well after treatment (6 months: 62% reduction [95% CI, 57–67%]; 24
tolerated and increases urinary cytokines in patients with NMIBC months: 41% reduction [95% CI, 31–49%]; 6 months: 22% reduction
in situ. Future results further investigating the efficacy of TMX-101 [95% CI, 0.54–0.95]).599 RCTs of topical imiquimod (5% applied
in preventing recurrence will determine its efficacy for patients daily 2–3 days per week for 16 weeks) have shown that 45% to 57%
with NMIBC. of patients have a complete response, and 59% to 75% of patients
Lifestyle and Cancer Prevention  •  CHAPTER 22 371

demonstrate a partial response.600–602 Conversely, a study of patients have found no evidence of preventive efficacy for SCC incidence618
treated with diclofenac (3%) in hyaluronan gel (twice daily for 90 or minimal and inconsistent preventive efficacy for SCC and BCC
days) reported a complete response in 50% of participants versus incidence.619 Regardless, a meta-analysis published in 2015 reported
20% of participants treated with vehicle.603 However, the limited a significant reduction in SCC risk in NSAID users compared with
resolution, prolonged treatment period, and adverse effects associated nonusers, which was enhanced in high-risk individuals (reduced risk
with treatment have limited its use. of SCC among users of any NSAIDs: 0.82 [95% CI, 0.71–0.94]).620
Ingenol mebutate, the agent most recently approved by the FDA Clouser and colleagues examined data from the SKICAP-AK trial in
for the treatment of AKs, induces rapid necrosis of lesions and initiates an effort to determine the association of NSAID use and time to first
an immune-mediated reaction that promotes healing.604 A reduction SCC or BCC in aspirin users and nonusers.621 Their results showed a
of 75% or more in existing AKs has been observed in 60% to 68% significant reduction in SCC (HR, 0.49 [95% CI, 0.28–0.87]) and
of participants with face and scalp AKs treated with ingenol mebutate BCC (HR, 0.43 [95% CI, 0.25–0.73]) in participants who were
versus those treated with placebo (7%–8% of participants) in multiple aspirin users for less than the full duration of the study, suggesting
phase III trials.605–606 These studies also show a reduction of 75% or that a shorter duration of NSAID use may be more protective than a
more of AKs on the trunk and extremities with ingenol mebutate longer duration. Most recently, a meta-analysis by Zhu and colleagues
versus placebo (44%–55% of participants versus 7%), and an induction investigated the association between aspirin use and prevention of
of AK necrosis after 2 to 3 days of treatment. A recent randomized skin cancer in individuals from eight case-control and five prospective
clinical trial compared the efficacy of ingenol mebutate (3-day treatment cohort studies.622 Their results demonstrate significantly reduced risk
cycle) and daylight photodynamic with therapy methyl aminolevulinate of skin cancer in individuals taking low-dose (≤150 mg) or higher-
(one treatment), and found similar efficacy for grade I/II AKs, but dose (50–400 mg) aspirin daily, particularly for risk of NMSC (OR,
decreased adverse events and improved cosmetic outcomes with daylight 0.97 [95% CI, 0.95–0.99]; P = .22). Additional RCTs are needed to
photodynamic with therapy methyl aminolevulinate. However, in confirm the potential preventive benefit of aspirin for NMSC. Although
2015 the FDA released a Drug Safety Communication warning of epidemiologic studies and alternative strategies using combined therapy,
severe adverse events associated with ingenol mebutate, including lower doses, and variable treatment times are promising, preventive
severe allergic reactions and herpes zoster (shingles).607 efficacy has not yet been established for naproxen and sulindac623 for
the prevention of NMSC.
Retinoids
Many RCTs have investigated topical and oral retinoids for the Cyclooxygenase-2 inhibitors
prevention of AKs and nonmelanoma skin cancers (NMSCs).608 The Both COX-1 and COX-2 are known to be present in NMSCs624;
retinoid acitretin (30 mg/day) was the focus of a clinical trial of 44 however, studies have shown that the primary COX isoform responsive
renal transplant recipients.609 This study demonstrated a reduction in to UVR in the human epidermis is COX-2.625 Based on accumulating
SCCs (11% versus 47%, χ2 = 6.27, P = .01) and a reduction in AKs evidence from preclinical studies demonstrating the efficacy of COX-2
(13.4% versus 28.2% [95% CI, 11.5–71.7]) in patients treated for 6 inhibitors in the prevention of NMSC, clinical trials have been
months with acitretin versus placebo. The majority of the associated conducted to investigate COX-2 inhibitors in this setting.626 Elmets
side effects were mild mucocutaneous reactions, followed by mild and colleagues conducted a phase III clinical trial of 240 subjects
hair loss; no deterioration in renal function was observed. However, treated with celecoxib (200 mg twice per day) or placebo for 9
conflicting results regarding the efficacy and/or safety of retinoids for months.627 They found that patients treated with celecoxib versus
the prevention of skin cancer have been also reported. Two years of placebo demonstrated no reduction in AKs 9 months after randomiza-
treatment with acitretin (25 mg orally 5 days per week) versus placebo tion, but significant reductions in NMSCs (RR, 0.41 [95% CI, =
was tested in 70 nontransplant high-risk patients with a recent history 0.23–0.72]; P = .002), BCCs (RR, 0.40 [95% CI, 0.18–0.93]; P =
of NMSCs.610 No statistically significant reduction was observed in .032), and SCCs (RR, 0.42 [95% CI, 0.19–0.93]; P = .032) 11
patients treated with acitretin versus placebo, but this may have been months after randomization, with similar cardiovascular severe adverse
a result of low statistical power. Similarly, the Veterans Affairs Topical effects. Regardless, a warning regarding serious or life-threatening
Tretinoin Chemoprevention Trial of high-dose topical tretinoin for adverse events is now required by the FDA for celecoxib owing to
keratinocyte carcinoma prevention, which accrued 1191 participants the potentially fatal side effects associated with the suppression of
for 1.5 to 5.5 years of treatment with high-dose tretinoin (0.1%) or prostaglandin synthesis after COX inhibition.628
vehicle, demonstrated no preventive benefit associated with this retinoid
in preventing BCC, SCC, or AKs, and increased side effects.611 The D,L-α-Difluoromethylornithine
Southwest Skin Cancer Prevention Study (SKICAP-BCC/SCC), one d,l-α-Difluoromethylornithine (DFMO) is an enzyme-activated,
of a set of SKICAP trials, was a large phase III study of oral retinol irreversible inhibitor of ODC and is the first enzyme in polyamine
(25,000 units/day) and isotretinoin (5–10 mg/day) versus placebo in 525 synthesis.629 Based on previous positive clinical results, a phase III
individuals with a history of BCC or SCC.612 No difference in the time clinical trial tested 4 to 5 years of treatment with DFMO (500 mg/
to first new occurrence of either BCC or SCC was observed between m2/day) or placebo in 291 participants with a history of NMSC.630
treatment arms. However, a reanalysis of this study in a dose-response Although DFMO was not associated with reduced incidence of NMSC
format identified an increased risk of SCC development in participants or SCC versus placebo, it was associated with a significant reduction
treated with either retinol or isotretinoin versus placebo in the first in BCC incidence (DFMO, 0.28 BCC per person per year; placebo,
quartile of dose.613 In a second SKICAP study, the SKICAP-AK trial, 0.40 BC per person per /year; P = .03), which was characterized by
2800 high-risk individuals were treated with retinol (25,000 IU) or a significant reduction in ODC activity and minimal adverse events.
placebo for 5 years614; the researchers found that retinol was associated Long-term follow-up of this study demonstrated a persistent but
with a reduction in incidence of first new SCC but not BCC.615 Thus, statistically insignificant reduction in NMSC incidence in individuals
before the incorporation of retinoids in standard of care for NMSC treated with DFMO versus placebo after discontinuation from the
prevention, additional larger-scale clinical trials will be required, with study that was not associated with any difference in latent or cumulative
extended follow-up examining the efficacy and associated toxicity of toxicity.631
these agents in the prevention of AKs and NMSC.608
Vismodegib
Nonsteroidal antiinflammatory drugs In 2012 vismodegib became the first inhibitor of the Hedgehog signaling
Some population-based studies have indicated that NSAIDs are associ- pathway to gain approval from the FDA for individuals with advanced
ated with reduced incidence of AKs and/or SCCs,616,617 but others or metastatic BCC and other cancers, which led to a clinical trial
372 Part I: Science and Clinical Oncology

testing its efficacy in preventing BCC recurrence.632,633 Tang and


colleagues treated 41 individuals with basal cell nevus syndrome with Melanoma
vismodegib (150 mg/day) or placebo for 3 months and found that As with other cancers, melanoma has numerous subtypes (over 30),
vismodegib reduced BCC size (−65% versus −11%) and incidence and approximately 26% of melanomas are histologically associated
of new BCCs (2 versus 29; P < .001) versus placebo; in addition, with dysplastic or other nevi.641–642
83% of patients in the treatment arm had no residual BCC at sites
with previous BCC at 1 month of being on treatment.632 However, Nonsteroidal antiinflammatory drugs
although participants receiving vismodegib commonly demonstrated The efficacy of NSAIDs as a preventive strategy for melanoma has
grade 1 or 2 adverse events, 54% discontinued the drug because of been suggested by both preclinical643–646 and epidemiologic647–650 studies.
adverse events. Based on these findings, Curiel-Lewandrowski and colleagues examined
the effects of 8 weeks of treatment with sulindac (150 twice per day)
Statins versus placebo in 50 individuals with atypical nevi on (1) the bioavail-
Wang and colleagues analyzed data from the Women’s Health Initiative ability of sulindac and metabolites, (2) apoptosis, and (3) vascular
Observational Study and the Women’s Health Initiative Clinical Trial endothelial growth factor A (VEGFA) expression in nevi.623 The findings
to determine whether use of statins was effective in incidence of demonstrated that individuals treated with sulindac had elevated
NMSC.634 A total of 118,357 non-Hispanic white women with no concentrations of the proapoptotic metabolite sulindac sulfone in
history of NMSC at baseline were included in the analysis; with 10.5 benign nevi, but no difference was observed in cleaved caspase-3 or
years of follow-up, individuals who used any statins at baseline VEGFA levels. Although these results demonstrate bioavailability of
demonstrated significantly higher incidence of NMSC, with highest sulindac and sulindac metabolites, further studies are needed to
risk associated with lovastatin (OR, 1.52 [95% CI, 1.08–2.16]), determine its efficacy as a preventive strategy for melanoma.
simvastatin (OR, 1.38 [95% CI, 1.12–1.69]), and lipophilic statins A UCLA retrospective pilot study of 148 patients with melanoma
(OR, 1.39 [95% CI, 1.18–1.64]), suggesting that statins increase risk examined the association between aspirin use (81 mg or 325 mg)
of NMSC in Caucasian postmenopausal women. Conversely, other 1 month or more before diagnosis and the aggressiveness of mela-
studies and meta-analyses have suggested no association or increased noma.651 Aspirin use significantly decreased Breslow depth (95% CI,
risk of NMSC with statin use; additional research is required to 0.297–0.8127; P = .03517), suggesting the potential benefit of aspirin
determine whether there is in fact a causal relationship between cancer for patients with or at high risk of melanoma. In addition, the meta-
risk and use of statins, and to determine the precise biologic mechanisms analysis by Zhu and colleagues, outlined under NMSC earlier, examined
involved.635 aspirin for prevention of skin cancer and found significant reductions
associated with aspirin at both 50 to 400 mg/day (OR, 0.94 [95%
Vitamins and minerals CI, 0.90–0.99]; P = .02) and 100 mg/day or less (OR, 0.95 [95%
A post hoc analysis of the WHI study, in which women were random- CI, 0.90–0.99]; P = .15).622 Although these results are promising,
ized to receive calcium (1000 mg/day) plus vitamin D3 (400 IU/day) the results from large-scale ongoing trials, such as the Aspirin in
or placebo for 7 years, investigated the effects of calcium and vitamin Reducing Events in the Elderly (ASPREE) trial, with an expected
D supplementation on risk of both NMSC and melanoma.636 No accrual of 19,000 participants to be treated with acetylsalicylic acid
difference in incidence of NMSC or melanoma compared with placebo (ASA; 100 mg) or placebo and a secondary end point that includes
was observed; however, patients with a history of NMSC in the melanoma incidence,652 will better define the efficacy of NSAIDs for
treatment arm had lower risk of melanoma compared with those with the prevention of melanoma.
a history of NMSC in the placebo arm (HR, 0.43 [95% CI, 0.21–0.90]).
These findings suggest that women at high risk of NMSC may benefit Statins
from supplementation with calcium and vitamin D. Conversely, the Epidemiologic data have suggested that intake of statins and fibrates,
Physicians’ Health Study of 12 years of treatment with β-carotene both of which are hypolipidemic (lipid-lowering) agents, is associated
(50 mg on alternate days) in 22,071 healthy physicians 40 to 84 years with a 10% and 14%, respectively, decreased risk of melanoma.653
of age found no reduction in risk of NMSC, BCC, or SCC associated Unfortunately, conflicting results regarding the relationship between
with β-carotene.637 statins and melanoma have been reported. A phase II trial of 80
Ferrucci and colleagues investigated the relationship between tea, subjects with multiple atypical nevi examined the impact of 6 months
coffee, and caffeine and early-onset BCC by using data from 767 of treatment with lovastatin versus placebo on biomarkers of melanoma
non-Hispanic white women younger than 40 years listed in Yale’s pathogenesis.654 No significant differences in any of the molecular
Dermatopathology database.638 Women who regularly consumed caf- biomarkers analyzed were apparent between the two study arms,
feinated coffee plus hot tea had reduced incidence of early-onset BCC suggesting that if lovastatin prevents melanoma, the mechanism though
(OR, 0.60 [95% CI, 0.38–0.96]), which was enhanced in women in which it functions is other than reversing precursor atypical nevi.
the highest category of caffeine use (OR, 0.57 [95% CI, 0.34–0.95]; P
trend = .037), versus nonconsumers, suggesting that caffeinated coffee Vitamins, minerals, and dietary factors
plus tea may provide a base level of protection against early-onset BCC. Many studies have examined the effects of vitamins and minerals on
More recently, an 11-year prospective study of black tea consumption risk of melanoma, most with negative or limited results. Numerous
reported no difference in risk of BCC or SCC in individuals who studies have been conducted to determine the potential of vitamin A
drank four or more cups per day and non–black tea drinkers.639 in preventing melanoma. Asgari and colleagues examined dietary and
Another study with positive results was a phase III trial that supplemental vitamin A and carotenoids in 69,635 participants of
investigated vitamin B3 (nicotinamide) for the prevention of mela- the Vitamins And Lifestyle (VITAL) cohort study, and demonstrated
noma.640 A total of 386 participants with a recent history of NMSC a significant reduction in risk of melanoma in individuals with baseline
were randomized to receive nicotinamide (500 mg twice per day) or use of retinol supplements (HR, 0.60 [95% CI, 0.41–0.89]) and
placebo for 12 months. Reduced incidence of NMSC (23% [95% individuals with high-dose (>1200 µg/day) use of retinol supplements
CI, 4–38]; P = .02), BCC (20% [95% CI, −6 to 39]; P = .12), SCC (HR, 0.74 [95% CI, 0.55–1.00]) versus nonusers.655 Neither dietary
(30% [95% CI, 0–51]; P = .05), and AK (13%; P = .001) was or total intake of vitamin A nor carotenoids were found to be associated
demonstrated in individuals treated with nicotinamide compared with with risk of melanoma. Conversely, an analysis published in 2015 of
placebo. Although no difference in adverse events was demonstrated nine cohort and intervention studies found no association between
between arms, the preventive benefit provided by nicotinamide did risk of melanoma and consumption of vitamin A (retinol), vitamin
not persist after discontinuation of use. C, vitamin E (tocopherol), carotenoids, or selenium or high fruit and
Lifestyle and Cancer Prevention  •  CHAPTER 22 373

vegetable intake.639,656 Future studies of different retinoid compounds an inverse association between consumption of caffeinated coffee and
alone or in combination with other agents will be required to make risk of melanoma in men in the highest quartile of consumption
a place for vitamin A in this setting. versus those with no consumption (HR, 0.31 [95% CI, 0.14–0.69]).
As mentioned previously, a post hoc study of the WHI, wherein No statistically significant differences were observed in women, or for
36,282 postmenopausal women were treated with calcium (1000 mg/ tea or decaffeinated coffee in either sex.
day) plus vitamin D3 (400 IU/day) or placebo for 7 years, found no
preventive efficacy on melanoma incidence associated with calcium Other agents
and vitamin D supplementation versus placebo.636 However, reduced Additional agents have been demonstrated to be effective in preventing
incidence of melanoma was observed in high-risk patients previously melanoma in preclinical studies, but positive results in clinical trials
diagnosed with NMSC (HR, 0.43 [95% CI, 0.21–0.90]). Conversely, regarding their efficacy and safety are lacking. Among these are cur-
some studies have suggested efficacy of other vitamin, mineral, or cumin, EGCG, the naturally occurring flavonoid fisetin, resveratrol,
diet-related strategies for reducing risk of melanoma. The large warfarin, and vitamins C, E, and K.658 Currently, although a number
multicenter European Prospective Investigation into Cancer and of agents have been found effective for the chemoprevention of mela-
Nutrition (EPIC) cohort study with more than 500,000 participants noma, none have been approved by the FDA.
aged 25 to 70 years from 10 countries examined the effects of total,
caffeinated, and decaffeinated coffee and tea consumption on melanoma The complete reference list is available online at
incidence.657 With 14.9 years of follow-up, this study demonstrated ExpertConsult.com.

KEY REFERENCES
5. Kohler LN, Garcia DO, Harris RB, Oren E, Roe DJ, loss on cancer incidence and mortality. Obes Rev. other cancers in male smokers. N Engl J Med. 1994;
Jacobs ET. Adherence to diet and physical activity 2012;13(10):868–891. 330(15):1029–1035.
cancer prevention guidelines and cancer outcomes: 77. Britton KA, Massaro JM, Murabito JM, Kreger 165. Omenn GS, Goodman GE, Thornquist MD, et al.
a systematic review. Cancer Epidemiol Biomarkers BE, Hoffmann U, Fox CS. Body fat distribution, Risk factors for lung cancer and for intervention
Prev. 2016;25(7):1018–1028. incident cardiovascular disease, cancer, and all-cause effects in CARET, the Beta-Carotene and Retinol
6. Vogelstein B, Papadopoulos N, Velculescu VE, mortality. J Am Coll Cardiol. 2013;62(10):921– Efficacy Trial. J Natl Cancer Inst. 1996;88(21):
Zhou S, Diaz LA Jr, Kinzler KW. Cancer genome 925. 1550–1559.
landscapes. Science. 2013;339(6127):1546–1558. 79. Donohoe C, Lysaght J, O’Sullivan J, Reynolds J. 172. Lippman SM, Klein EA, Goodman PJ, et al. Effect
8. Sporn MB, Dunlop NM, Newton DL, Smith JM. Emerging concepts linking obesity with the hallmarks of selenium and vitamin E on risk of prostate cancer
Prevention of chemical carcinogenesis by vitamin of cancer. Trends Endocrinol Metab. 2017;28(1): and other cancers: the Selenium and Vitamin E
A and its synthetic analogs (retinoids). Fed Proc. 46–62. Cancer Prevention Trial (SELECT). JAMA. 2009;
1976;35(6):1332–1338. 80. Iyengar NM, Hudis CA, Dannenberg AJ. Obesity 301(1):39–51.
15. Centers for Disease Control and Prevention. and cancer: local and systemic mechanisms. Annu 173. Klein EA, Thompson IM Jr, Tangen CM, et al.
Best Practices for Comprehensive Tobacco Control Rev Med. 2015;66:297–309. Vitamin E and the risk of prostate cancer: the
Programs—2014. Atlanta, GA: Department of 85. Gonçalves A, Dantas Florencio G, Maisonnette de Selenium and Vitamin E Cancer Prevention Trial
Health and Human Services, Centers for Disease Atayde Silva M, Cobucci R, Giraldo P, Cote N. Effect (SELECT). JAMA. 2011;306(14):1549–1556.
Control and Prevention, National Center for Chronic of physical activity on breast cancer prevention: a 191. Hong WK, Endicott J, Itri LM, et al. 13-cis-retinoic
Disease Prevention and Health Promotion, Office systematic review. J Phys Act Health. 2014;11(2): acid in the treatment of oral leukoplakia. N Engl J
on Smoking and Health; 2014. 445–454. Med. 1986;315(24):1501–1505.
18. Eriksen M, Mackay J, Schluger N, Gomeshtapeh 86. Lakoski S, Willis B, Barlow C, et al. Midlife 207. William WN Jr, Papadimitrakopoulou V, Lee JJ, et al.
FI, Drope J. The Tobacco Atlas. 5th ed. Atlanta, GA: cardiorespiratory fitness, incident cancer, and survival Erlotinib and the risk of oral cancer: the Erlotinib
The American Cancer Society; 2015. after cancer in men: the Cooper Center Longitudinal Prevention of Oral Cancer (EPOC) Randomized
23. Bagnardi V, Rota M, Botteri E, et al. Alcohol con- Study. JAMA Oncol. 2015;1(2):231–237. Clinical Trial. JAMA Oncol. 2016;2(2):209–
sumption and site-specific cancer risk: a comprehen- 88. Sanchis-Gomar F, Lucia A, Yvert T, et al. Physical 216.
sive dose-response meta-analysis. Br J Cancer. 2015; inactivity and low fitness deserve more attention 315. Meyskens FL Jr, McLaren CE, Pelot D, et al.
112(3):580–593. to alter cancer risk and prognosis. Cancer Prev Res Difluoromethylornithine plus sulindac for the
26. AICR / WCRF. Continuous Update Project Report: (Phila). 2015;8(2):105–110. prevention of sporadic colorectal adenomas: a
Diet, Nutrition, Physical Activity, and Prostate 89. Lee I, Shiroma E, Lobelo F, et al. Effect of physical inac- randomized placebo-controlled, double-blind trial.
Cancer. World Cancer Research Fund (WCRF) / tivity on major non-communicable disease worldwide: Cancer Prev Res (Phila). 2008;1(1):32–38.
American Institute for Cancer Research (AICR); an analysis of burden of disease and life expectancy. 322. Kimura T, McKolanis JR, Dzubinski LA, et al.
2014. Lancet. 2012;380:219–229. MUC1 vaccine for individuals with advanced
34. Mostofsky E, Mukamal KJ, Giovannucci EL, 97. Ross R, Janssen I, Dawson J, et al. Exercise-induced adenoma of the colon: a cancer immunoprevention
Stampfer MJ, Rimm EB. Key findings on alcohol reduction in obesity and insulin resistance in women: feasibility study. Cancer Prev Res (Phila). 2013;6(1):
consumption and a variety of health outcomes a randomized controlled trial. Obes Res. 2004;12: 18–26.
from the Nurses’ Health Study. Am J Public Health. 789–798. 353. Pan KF, Zhang L, Gerhard M, et al. A large
2016;106(9):1586–1591. 130. US Department of Health and Human Services. randomised controlled intervention trial to prevent
58. Scoccianti C, Lauby-Secretan B, Bello PY, Chajes The Surgeon General’s Call to Action to Prevent gastric cancer by eradication of Helicobacter pylori in
V, Romieu I. Female breast cancer and alcohol Skin Cancer. Washington D.C. 2014. Linqu County, China: baseline results and factors
consumption: a review of the literature. Am J Prev 141. Colantonio S, Bracken MB, Beecker J. The associa- affecting the eradication. Gut. 2016;65(1):9–18.
Med. 2014;46(3 suppl 1):S16–S25. tion of indoor tanning and melanoma in adults: 355. Rothwell PM, Fowkes FG, Belch JF, Ogawa H,
63. AICR / WCRF. Recommendations for Cancer Preven- systematic review and meta-analysis. J Am Acad Warlow CP, Meade TW. Effect of daily aspirin on
tion; 2016. http://www.aicr.org/reduce-your-cancer Dermatol. 2014;70(5):847–857.e841–818. long-term risk of death due to cancer: analysis of
-risk/recommendations-for-cancer-prevention/. 151. Lin JS, Eder M, Weinmann S. Behavioral counseling individual patient data from randomised trials.
64. Kushi L, Doyle C, McCullough M, et al. American to prevent skin cancer: a systematic review for the Lancet. 2011;377(9759):31–41.
Cancer Society Guidelines on nutrition and physical U.S. Preventive Services Task Force. Ann Intern Med. 378. Qu C, Chen T, Fan C, et al. Efficacy of neonatal
activity for cancer prevention: reducing the risk 2011;154(3):190–201. HBV vaccination on liver cancer and other liver
of cancer with healthy food choices and physical 159. Domchek SM, Friebel TM, Singer CF, et al. Associa- diseases over 30-year follow-up of the Qidong
activity. CA Cancer J Clin. 2012;62(1):30–67. tion of risk-reducing surgery in BRCA1 or BRCA2 hepatitis B intervention study: a cluster random-
69. Lauby-Secretan B, Scoccianti C, Loomis D, et al. mutation carriers with cancer risk and mortality. ized controlled trial. PLoS Med. 2014;11(12):
Body fatness and cancer—viewpoint of the IARC JAMA. 2010;304(9):967–975. e1001774.
working group. N Engl J Med. 2016;375(8):794–798. 164. Alpha-Tocopherol, Beta Carotene Cancer Preven- 419. Litton JK, Arun BK, Brown PH, Hortobagyi GN.
70. Birks S, Peeters A, Backholer K, O’Brien P, Brown tion Study Group. The effect of vitamin E and Aromatase inhibitors and breast cancer prevention.
W. A systematic review of the impact of weight beta carotene on the incidence of lung cancer and Expert Opin Pharmacother. 2012;13(3):325–331.
374 Part I: Science and Clinical Oncology

460. Disis ML, Wallace DR, Gooley TA, et al. Concurrent 576. Dinney CP, Greenberg RE, Steinberg GD. 633. Sekulic A, Migden MR, Oro AE, et al. Efficacy
trastuzumab and HER2/neu-specific vaccination in Intravesical valrubicin in patients with bladder and safety of vismodegib in advanced basal-cell
patients with metastatic breast cancer. J Clin Oncol. carcinoma in situ and contraindication to or carcinoma. N Engl J Med. 2012;366(23):2171–
2009;27(28):4685–4692. failure after bacillus Calmette-Guérin. Urol Oncol. 2179.
519. Joura EA, Giuliano AR, Iversen OE, et al. A 9-valent 2013;31(8):1635–1642. 652. ASPREE Investigator Group. Study design of ASPirin
HPV vaccine against infection and intraepithelial 605. Lebwohl M, Swanson N, Anderson LL, Melgaard A, in Reducing Events in the Elderly (ASPREE): a
neoplasia in women. N Engl J Med. 2015;372(8): Xu Z, Berman B. Ingenol mebutate gel for actinic randomized, controlled trial. Contemp Clin Trials.
711–723. keratosis. N Engl J Med. 2012;366(11):1010–1019. 2013;36(2):555–564.
Lifestyle and Cancer Prevention  •  CHAPTER 22 374.e1
374.e1

REFERENCES
1. Union of International Cancer Control (UICC). 20. World Health Organization (WHO). Global Status of acetaldehyde. Alcohol Clin Exp Res. 2000;24(6):
The Economics of Cancer Prevention and Control: Report on Alcohol and Health, 2014. Geneva, 873–877.
Data Digest; 2014. Switzerland, 2014. 40. Yokoyama A, Muramatsu T, Ohmori T, et al.
2. World Health Organization (WHO). Cancer 21. Wilsnack SC, Wilsnack RW, Kantor LW. Focus on: Alcohol-related cancers and aldehyde dehydro-
Prevention. www.who.int/cancer/prevention/en/. women and the costs of alcohol use. Alcohol Res. 2013; genase-2 in Japanese alcoholics. Carcinogenesis.
Accessed February 8, 2015. 35(2):219–228. 1998;19(8):1383–1387.
3. Doll R, Peto R. The causes of cancer: quantita- 22. Baan R, Straif K, Grosse Y, et al. Carcinogenicity 41. Matsuo K, Hamajima N, Shinoda M, et al. Gene-
tive estimates of avoidable risks of cancer in the of alcoholic beverages. Lancet Oncol. 2007;8(4): environment interaction between an aldehyde
United States today. J Natl Cancer Inst. 1981;66(6): 292–293. dehydrogenase-2 (ALDH2) polymorphism and
1191–1308. 23. Bagnardi V, Rota M, Botteri E, et al. Alcohol con- alcohol consumption for the risk of esophageal
4. Colditz GA, Wei EK. Preventability of cancer: the sumption and site-specific cancer risk: a comprehen- cancer. Carcinogenesis. 2001;22(6):913–916.
relative contributions of biologic and social and sive dose-response meta-analysis. Br J Cancer. 2015; 42. Matsuda T, Yabushita H, Kanaly RA, Shibutani S,
physical environmental determinants of cancer 112(3):580–593. Yokoyama A. Increased DNA damage in ALDH2-
mortality. Annu Rev Public Health. 2012;33:137– 24. Bagnardi V, Rota M, Botteri E, et al. Alcohol deficient alcoholics. Chem Res Toxicol. 2006;19(10):
156. consumption and lung cancer risk in never smokers: 1374–1378.
5. Kohler LN, Garcia DO, Harris RB, Oren E, Roe DJ, a meta-analysis. Ann Oncol. 2011;22(12):2631–2639. 43. Morimoto K, Takeshita T. Low Km aldehyde
Jacobs ET. Adherence to diet and physical activity 25. Zhao J, Stockwell T, Roemer A, Chikritzhs T. Is dehydrogenase (ALDH2) polymorphism, alcohol-
cancer prevention guidelines and cancer outcomes: alcohol consumption a risk factor for prostate cancer? drinking behavior, and chromosome alterations in
a systematic review. Cancer Epidemiol Biomarkers A systematic review and meta-analysis. BMC Cancer. peripheral lymphocytes. Environ Health Perspect.
Prev. 2016;25(7):1018–1028. 2016;16(1):845. 1996;104(suppl 3):563–567.
6. Vogelstein B, Papadopoulos N, Velculescu VE, 26. AICR/WCRF. Continuous Update Project Report: 44. Ishikawa H, Yamamoto H, Tian Y, Kawano M,
Zhou S, Diaz LA Jr, Kinzler KW. Cancer genome Diet, Nutrition, Physical Activity, and Prostate Yamauchi T, Yokoyama K. Effects of ALDH2 gene
landscapes. Science. 2013;339(6127):1546–1558. Cancer. World Cancer Research Fund (WCRF)/ polymorphisms and alcohol-drinking behavior on
7. Blackburn EH. Cancer interception. Cancer Prev American Institute for Cancer Research (AICR); micronuclei frequency in non-smokers. Mutat Res.
Res (Phila). 2011;4(6):787–792. 2014. 2003;541(1–2):71–80.
8. Sporn MB, Dunlop NM, Newton DL, Smith JM. 27. Cao Y, Willett WC, Rimm EB, Stampfer MJ, 45. Seitz HK, Stickel F. Risk factors and mechanisms
Prevention of chemical carcinogenesis by vitamin Giovannucci EL. Light to moderate intake of alcohol, of hepatocarcinogenesis with special emphasis on
A and its synthetic analogs (retinoids). Fed Proc. drinking patterns, and risk of cancer: results from alcohol and oxidative stress. Biol Chem. 2006;387(4):
1976;35(6):1332–1338. two prospective US cohort studies. BMJ. 2015; 349–360.
9. Spira A, Yurgelun MB, Alexandrov L, et al. Precancer 351:h4238. 46. Hu W, Feng Z, Eveleigh J, et al. The major lipid per-
atlas to drive precision prevention trials. Cancer Res. 28. Boffetta P, Hashibe M. Alcohol and cancer. Lancet oxidation product, trans-4-hydroxy-2-nonenal, prefer-
2017;77(7):1510–1541. Oncol. 2006;7(2):149–156. entially forms DNA adducts at codon 249 of human
10. U.S. Department of Health and Human Services. 29. Ahmad Kiadaliri A, Jarl J, Gavriilidis G, Gerdtham p53 gene, a unique mutational hotspot in hepa-
The Health Consequences of Smoking: 50 Years of UG. Alcohol drinking cessation and the risk of tocellular carcinoma. Carcinogenesis. 2002;23(11):
Progress. A Report of the Surgeon General. Atlanta, laryngeal and pharyngeal cancers: a systematic review 1781–1789.
GA 2014. and meta-analysis. PLoS ONE. 2013;8(3):e58158. 47. Fu P, Yang F, Li B, et al. Meta-analysis of CYP2E1
11. Jamal A, King BA, Neff LJ, Whitmill J, Babb SD, 30. Rehm J, Patra J, Popova S. Alcohol drinking ces- polymorphisms in liver carcinogenesis. Dig Liver
Graffunder CM. Current cigarette smoking among sation and its effect on esophageal and head and Dis. 2017;49(1):77–83.
adults—United States, 2005–2015. MMWR Morb neck cancers: a pooled analysis. Int J Cancer. 2007; 48. Colditz GA. Relationship between estrogen levels,
Mortal Wkly Rep. 2016;65(44):1205–1211. 121(5):1132–1137. use of hormone replacement therapy, and breast
12. Singh T, Arrazola RA, Corey CG, et al. Tobacco use 31. Heckley GA, Jarl J, Asamoah BO, G-Gerdtham U. cancer. J Natl Cancer Inst. 1998;90(11):814–
among middle and high school students—United How the risk of liver cancer changes after alcohol 823.
States, 2011–2015. MMWR Morb Mortal Wkly Rep. cessation: a review and meta-analysis of the current 49. Key T, Appleby P, Barnes I, Reeves G, Group
2016;65(14):361–367. literature. BMC Cancer. 2011;11:446. EHaBCC. Endogenous sex hormones and breast
13. Barrington-Trimis JL, Urman R, Berhane K, et al. 32. Ronksley PE, Brien SE, Turner BJ, Mukamal KJ, cancer in postmenopausal women: reanalysis
E-cigarettes and future cigarette use. Pediatrics. 2016; Ghali WA. Association of alcohol consumption with of nine prospective studies. J Natl Cancer Inst.
138(1). selected cardiovascular disease outcomes: a systematic 2002;94(8):606–616.
14. U.S. Department of Health and Human Services. review and meta-analysis. BMJ. 2011;342:d671. 50. Key TJ, Appleby PN, Reeves GK, et al. Circulat-
E-Cigarettes Use Among Youth and Young Adults: A 33. Connor J. Alcohol consumption as a cause of cancer. ing sex hormones and breast cancer risk factors in
Report of the Surgeon General. Atlanta, GA: U.S. Addiction. 2017;112(2):222–228. postmenopausal women: reanalysis of 13 studies.
Department of Health and Human Services, Centers 34. Mostofsky E, Mukamal KJ, Giovannucci EL, Br J Cancer. 2011;105(5):709–722.
for Disease Control and Prevention, National Center Stampfer MJ, Rimm EB. Key findings on alcohol 51. Dorgan JF, Baer DJ, Albert PS, et al. Serum hormones
for Chronic Disease Prevention and Health Promo- consumption and a variety of health outcomes and the alcohol-breast cancer association in post-
tion, Office on Smoking and Health; 2016. from the Nurses’ Health Study. Am J Public Health. menopausal women. J Natl Cancer Inst. 2001;93(9):
15. Centers for Disease Control and Prevention. 2016;106(9):1586–1591. 710–715.
Best Practices for Comprehensive Tobacco Control 35. Smyth A, Teo KK, Rangarajan S, et al. Alcohol con- 52. Ginsburg ES, Mello NK, Mendelson JH, et al. Effects
Programs—2014. Atlanta, GA: Department of sumption and cardiovascular disease, cancer, injury, of alcohol ingestion on estrogens in postmenopausal
Health and Human Services, Centers for Disease admission to hospital, and mortality: a prospective women. JAMA. 1996;276(21):1747–1751.
Control and Prevention, National Center for Chronic cohort study. Lancet. 2015;386(10007):1945–1954. 53. Mendelson JH, Lukas SE, Mello NK, Amass L,
Disease Prevention and Health Promotion, Office 36. Praud D, Rota M, Rehm J, et al. Cancer incidence Ellingboe J, Skupny A. Acute alcohol effects on
on Smoking and Health; 2014. and mortality attributable to alcohol consumption. plasma estradiol levels in women. Psychopharmacology
16. Eriksen M, Mackay J, Ross H. The Tobacco Atlas. Int J Cancer. 2016;138(6):1380–1387. (Berl). 1988;94(4):464–467.
4th ed. Atlanta, GA: The American Cancer Society; 37. Seitz HK, Stickel F. Molecular mechanisms of 54. Reichman ME, Judd JT, Longcope C, et al. Effects
2012. alcohol-mediated carcinogenesis. Nat Rev Cancer. of alcohol consumption on plasma and urinary
17. Wipfli H, Samet JM. One hundred years in the 2007;7(8):599–612. hormone concentrations in premenopausal women.
making: the global tobacco epidemic. Annu Rev 38. Seitz HK, Mueller S. Alcohol and cancer: an overview J Natl Cancer Inst. 1993;85(9):722–727.
Public Health. 2016;37:149–166. with special emphasis on the role of acetaldehyde 55. Sarkola T, Mäkisalo H, Fukunaga T, Eriksson
18. Eriksen M, Mackay J, Schluger N, Gomeshtapeh and cytochrome P450 2E1. In: Vasiliou V, Zakhari CJ. Acute effect of alcohol on estradiol, estrone,
FI, Drope J. The Tobacco Atlas. 5th ed. Atlanta, GA: S, Seitz HK, Hoek JB, eds. Biological Basis of Alcohol- progesterone, prolactin, cortisol, and luteinizing
The American Cancer Society; 2015. Induced Cancer, Advances in Experimental Medicine hormone in premenopausal women. Alcohol Clin
19. National Cancer Institute (NCI) and the World and Biology. Switzerland: Springer International Exp Res. 1999;23(6):976–982.
Health Organization (WHO). The Economics of Publishing; 2015. 56. Gavaler JS, Van Thiel DH. The association between
Tobacco and Tobacco Control, National Cancer 39. Väkeväinen S, Tillonen J, Agarwal DP, Srivastava moderate alcoholic beverage consumption and serum
Institute Tobacco Control Monograph 212016. N, Salaspuro M. High salivary acetaldehyde after a estradiol and testosterone levels in normal postmeno-
Located at: NIH Publication No. 16-CA-8029A, moderate dose of alcohol in ALDH2-deficient sub- pausal women: relationship to the literature. Alcohol
Bethesda, MD. jects: strong evidence for the local carcinogenic action Clin Exp Res. 1992;16(1):87–92.
374.e2 Part I: Science and Clinical Oncology

57. Sarkola T, Fukunaga T, Mäkisalo H, Peter Eriksson 79. Donohoe C, Lysaght J, O’Sullivan J, Reynolds in obese individuals without weight loss. Hepatology.
CJ. Acute effect of alcohol on androgens in J. Emerging concepts linking obesity with the 2009;50:1102–1112.
premenopausal women. Alcohol. 2000;35(1):84–90. hallmarks of cancer. Trends Endocrinol Metab. 2017; 100. Arner P, Kriegholm E, Engfeldt P, Bolinder J.
58. Scoccianti C, Lauby-Secretan B, Bello PY, Chajes 28(1):46–62. Adrenergic regulation of lipolysis in situ at rest and
V, Romieu I. Female breast cancer and alcohol 80. Iyengar NM, Hudis CA, Dannenberg AJ. Obesity during exercise. J Clin Invest. 1990;85:893–898.
consumption: a review of the literature. Am J Prev and cancer: local and systemic mechanisms. Annu 101. Horowitz J, Leone T, Feng W, Kelly D, Klein S.
Med. 2014;46(3 suppl 1):S16–S25. Rev Med. 2015;66:297–309. Effect of endurance training on lipid metabolism
59. Liu Y, Nguyen N, Colditz GA. Links between 81. Teoh S, Das S. Tumour biology of obesity-related in women: a potential role for PPAR alpha in
alcohol consumption and breast cancer: a look at the cancers: understanding the molecular concept for the metabolic response to training. Am J Physiol
evidence. Womens Health (Lond). 2015;11(1):65–77. better diagnosis and treatment. Tumor Biol. 2015. Endocrinol Metab. 2000;2079:E348–E355.
60. Mason JB, Tang SY. Folate status and colorectal 82. Pescatello L, Arena R, Riebe D, Thompson P, eds. 102. Sturgeon K, Digiovanni L, Good J, et al. Exercise-
cancer risk: a 2016 update. Mol Aspects Med. 2017;53: ACSM’s Guidelines for Exercise Testing and Prescrip- induced dose-response alterations in adiponectin and
73–79. tion. 9th ed. Baltimore, MD: Lippincott, Williams leptin levels are dependent on body fat changes in
61. Nan H, Lee JE, Rimm EB, Fuchs CS, Giovannucci & Wilkins; 2013. women at risk for breast cancer. Cancer Epidemiol
EL, Cho E. Prospective study of alcohol consumption 83. Abioye AI, Odesanya MO, Abioye AI, Ibrahim N. Biomarkers Prev. 2016;25(8):1195–1200.
and the risk of colorectal cancer before and after Physical activity and risk of gastric cancer: a meta- 103. Attie A, Scherer P. Adipocyte metabolism and obesity.
folic acid fortification in the United States. Ann analysis of observational studies. Br J Sports Med. J Lipid Res. 2009;50:S395–S399.
Epidemiol. 2013;23(9):558–563. 2015;49:224–229. 104. Richter E, Hargreaves M. Exercise, Glut 4, and
62. Kantor ED, Giovannucci EL. Gene-diet interactions 84. Friedenreich C. Physical activity and breast cancer: skeletal muscle glucose uptake. Physiol Rev. 2013;93:
and their impact on colorectal cancer risk. Curr Nutr review of the epidemiologic evidence and biologic 993–1017.
Rep. 2015;4(1):13–21. mechanisms. Recent Results Cancer Res. 2011;188: 105. Goh J, Niksirat N, Campbell K. Exercise training and
63. AICR / WCRF. Recommendations for Cancer Preven- 125–139. immune crosstalk in breast cancer microenvironment:
tion; 2016. http://www.aicr.org/reduce-your-cancer 85. Gonçalves A, Dantas Florencio G, Maisonnette de exploring the paradigms of the exercise-induced
-risk/recommendations-for-cancer-prevention/. Atayde Silva M, Cobucci R, Giraldo P, Cote N. immune modulation and exercise-induced myokines.
64. Kushi L, Doyle C, McCullough M, et al. American Effect of physical activity on breast cancer preven- Am J Transl Res. 2014;6(5):422–438.
Cancer Society Guidelines on nutrition and physical tion: a systematic review. J Phys Act Health. 2014; 106. Aoi W, Naito Y, Takagi T, et al. A novel myokine,
activity for cancer prevention: reducing the risk 11(2):445–454. secreted protein acidic and rich in cysteine (SPARC),
of cancer with healthy food choices and physical 86. Lakoski S, Willis B, Barlow C, et al. Midlife suppresses colon tumorigenesis via regular exercise.
activity. CA Cancer J Clin. 2012;62(1):30–67. cardiorespiratory fitness, incident cancer, and survival Gut. 2013;62(6):882–889.
65. Moyer VA, Force PST. Screening and behavioral after cancer in men: the Cooper Center Longitudinal 107. Gannon N, Vaughan R, Garcia-Smith R, Bisoffi
counseling interventions in primary care to reduce Study. JAMA Oncol. 2015;1(2):231–237. M, Trujillo K. Effects of the exercise-inducible
alcohol misuse: U.S. preventive services task force 87. Wu Y, Zhang D, Kang S. Physical activity and risk of myokine irisin on malignant and non-malignant
recommendation statement. Ann Intern Med. 2013; breast cancer: a meta-analysis of prospective studies. breast epithelial cell behavior in vitro. Int J Cancer.
159(3):210–218. Cancer Res Treat. 2013;137:869–882. 2015;136(4):E197–E202.
66. Wagenaar AC, Salois MJ, Komro KA. Effects of 88. Sanchis-Gomar F, Lucia A, Yvert T, et al. Physical 108. Hojman P, Dethlefsen C, Brandt C, Hansen J, Ped-
beverage alcohol price and tax levels on drinking: inactivity and low fitness deserve more attention ersen L, Pedersen B. Exercise-induced muscle-derived
a meta-analysis of 1003 estimates from 112 studies. to alter cancer risk and prognosis. Cancer Prev Res cytokines inhibit mammary cancer cell growth. Am J
Addiction. 2009;104(2):179–190. (Phila). 2015;8(2):105–110. Physiol Endocrinol Metab. 2011;301(3):E504–E510.
67. Wagenaar AC, Tobler AL, Komro KA. Effects of 89. Lee I, Shiroma E, Lobelo F, et al. Effect of physical 109. Idorn M, Hojman P. Exercise-dependent regulation
alcohol tax and price policies on morbidity and inactivity on major non-communicable disease of NK cells in cancer protection. Trends Mol Med.
mortality: a systematic review. Am J Public Health. worldwide: an analysis of burden of disease and 2016;22(7):565–577.
2010;100(11):2270–2278. life expectancy. Lancet. 2012;380:219–229. 110. Idorn M, Thor Straten P. Exercise: a new role for
68. Ogden C, Carroll M, Kit B, Flegal K. Prevalence of 90. Schmid D, Leitzmann M. Television viewing and an old tool. Mol Cell Oncol. 2016;3(4):e1163005.
childhood and adult obesity in the United States, time spent sedentary in relation to cancer risk: a 111. Pedersen L, Idorn M, Olofsson G, et al. Voluntary
2011-2012. JAMA. 2014;311(8):806–814. meta-analysis. J Natl Cancer Inst. 2014;106(7). running suppresses tumor growth through epineph-
69. Lauby-Secretan B, Scoccianti C, Loomis D, et al. 91. Nomura S, Dash C, Rosenberg L, Palmer J, Adams- rine- and IL-6-dependent NK cell mobilization and
Body fatness and cancer- viewpoint of the IARC Campbell L. Sedentary time and breast cancer redistribution. Cell Metab. 2016;23(3):554–562.
working group. N Engl J Med. 2016;375(8):794–798. incidence in African American women. Cancer 112. Alexander D, Weed D, Miller P, Mohamed M. Red
70. Birks S, Peeters A, Backholer K, O’Brien P, Brown Causes Control. 2016;27:1239–1252. meat and colorectal cancer: a quantitative update
W. A systematic review of the impact of weight 92. Giannopoulou I, Ploutz-Snyder L, Carhart R, on the state of the epidemiologic science. J Am Coll
loss on cancer incidence and mortality. Obes Rev. et al. Exercise is required for visceral fat loss in Nutr. 2015;34(6):521–543.
2012;13(10):868–891. postmenopausal women with type 2 diabetes. 113. Bail J, Meneses K, Demark-Wahnefried W. Nutri-
71. Esposito K, Chiodini P, Colao A, Lenzi A, Giudliano J Clin Endocrinol Metab. 2005;90(3):1511–1518. tional status and diet in cancer prevention. Semin
D. Metabolic syndrome and risk of cancer: a system- 93. Hallsworth K, Thoma C, Hollingsworth K, et al. Oncol Nurs. 2016;32(3):206–214.
atic review and meta analysis. Diabetes Care. 2012; Modified high-intensity interval training reduces liver 114. Godos J, Bella F, Torrisi A, Sciacca S, Galvano F,
35:2402–2411. fat and improves cardiac function in non-alcoholic Grosso G. Dietary patterns and risk of colorectal
72. Micucci C, Valli D, Matacchione G, Catalano A. fatty liver disease: a randomized controlled trial. adenoma: a systematic review and meta-analysis of
Current perspectives between metabolic syndrome Clin Sci. 2015;129(12):1097–1105. observational studies. J Hum Nutr Diet. 2016.
and cancer. Oncotarget. 2016. 94. Ismail I, Keating S, Baker M, Johnson N. A 115. Zhao Z, Yin Z, Pu Z, Zhao Q. Association
73. Uzunlulu M, Ozge T, Oguz A. Association between systematic review and meta-analysis of the effect between consumption of red and processed meat
metabolic syndrome and cancer. Ann Nutr Metab. of aerobic vs. resistance exercise training on visceral and pancreatic cancer risk, a systematic review and
2016;68:173–179. fat. Obes Rev. 2012;13:68–91. meta-analysis. Clin Gastroenterol Hepatol. 2016.
74. Despres J, Lemieux I. Abdominal obesity and meta- 95. Keating S, Hackett D, Parker H, et al. Effect of 116. Fang X, Wei J, He X, et al. Landscape of dietary
bolic syndrome. Nature. 2006;444(14):881–887. aerobic exercise training dose on liver fat and visceral factors associated with risk of gastric cancer: a system-
75. Doyle S, Donohoe C, Lysaght J, Reynolds J. Visceral adiposity. J Hepatol. 2015;63(1):174–182. atic review and dose-response meta-analysis of pro-
obesity, metabolic syndrome, insulin resistance and 96. Lee S, Kuk J, Davidson L, et al. Exercise without spective cohort studies. Eur J Cancer. 2015;51(18):
cancer. Proc Nutr Soc. 2012;71(1):181–189. weight loss is an effective strategy for obesity reduc- 2820–2832.
76. Donohoe C, Doyle S, Reynolds J. Visceral adiposity, tion in obese individuals with and without Type 2 117. Nöthlings U, Wilkens L, Murphy S, Hankin J,
insulin resistance and cancer risk. Diabetol Metab diabetes. J Appl Physiol. 2005;99(3):1220–1225. Henderson B, Kolonel L. Meat and fat intake as
Syndr. 2011;3(12). 97. Ross R, Janssen I, Dawson J, et al. Exercise-induced risk factors for pancreatic cancer: the multiethnic
77. Britton KA, Massaro JM, Murabito JM, Kreger reduction in obesity and insulin resistance in women: cohort study. J Natl Cancer Inst. 2005;97(19):1458–
BE, Hoffmann U, Fox CS. Body fat distribution, a randomized controlled trial. Obes Res. 2004; 1465.
incident cardiovascular disease, cancer, and all-cause 12:789–798. 118. Fahrer J, Kaina B. Impact of DNA repair on
mortality. J Am Coll Cardiol. 2013;62(10):921–925. 98. Thomas E, Brynes A, McCarthy J, et al. Preferen- the dose-response of colorectal cancer formation
78. Murphy RA, Bureyko TF, Miljkovic I, et al. Associa- tial loss of visceral fat following aerobic exercise, induced by dietary carcinogens. Food Chem Toxicol.
tion of total and computed tomographic measures measured by magnetic resonance imaging. Lipids. 2016.
of regional adiposity with incident cancer risk: a 2000;35(7):769–776. 119. Cross A, Sinha R. Meat-related mutagens/carcinogens
prospective population-based study of older adults. 99. Johnson N, Sachinwalla T, Walton D, et al. Aerobic in the etiology of colorectal cancer. Environ Mol
Appl Physiol Nutr Metab. 2014;39(6):987–992. exercise training reduces hepatic and visceral lipids Mutagen. 2004;44(1):44–55.
Lifestyle and Cancer Prevention  •  CHAPTER 22 374.e3
374.e3

120. Song P, Wu L, Guan W. Dietary nitrates, nitrites, and 140. Wehner MR, Shive ML, Chren MM, Han J, Qureshi 160. Yuan JM, Stram DO, Arakawa K, Lee HP, Yu MC.
nitrosamines intake and the risk of gastric cancer: a AA, Linos E. Indoor tanning and non-melanoma Dietary cryptoxanthin and reduced risk of lung
meta-analysis. Nutrients. 2015;7(12):9872–9895. skin cancer: systematic review and meta-analysis. cancer: the Singapore Chinese Health Study. Cancer
121. Cheng X, Lin J, Tu S. Etiology and prevention of BMJ. 2012;345:e5909. Epidemiol Biomarkers Prev. 2003;12(9):890–898.
gastric cancer. Gastrointest Tumors. 2016;3(1):25– 141. Colantonio S, Bracken MB, Beecker J. The associa- 161. Albanes D, Heinonen OP, Taylor PR, et al. Alpha-
36. tion of indoor tanning and melanoma in adults: Tocopherol and beta-carotene supplements and
122. Fox J, Dangler C, Taylor N, King A, Koh T, Wang systematic review and meta-analysis. J Am Acad lung cancer incidence in the Alpha-Tocopherol,
T. High-salt diet induces gastric epithelial hyperplasia Dermatol. 2014;70(5):847–857.e841–818. Beta-Carotene Cancer Prevention Study: effects of
and parietal cell loss, and enhances Helicobacter pylori 142. International Agency for Research on Cancer (IARC). base-line characteristics and study compliance. J Natl
colonization in C57BL/6 mice. Cancer Res. 1999; Biological agents. Volume 100 B. A review of human Cancer Inst. 1996;88(21):1560–1570.
59(19):4823–4828. carcinogens. IARC Monogr Eval Carcinog Risks Hum. 162. Wright ME, Mayne ST, Stolzenberg-Solomon RZ,
123. Godos J, Bella F, Sciacca S, Galvano F, Grosso G. 2012;100(Pt B):1–441. et al. Development of a comprehensive dietary
Vegetarianism and breast, colorectal and prostate 143. Moura Valejo Coelho M, Matos TR, Apetato M. antioxidant index and application to lung cancer
cancer risk: an overview and meta-analysis of cohort The dark side of the light: mechanisms of photo- risk in a cohort of male smokers. Am J Epidemiol.
studies. J Hum Nutr Diet. 2016. carcinogenesis. Clin Dermatol. 2016;34(5):563–570. 2004;160(1):68–76.
124. Dinu M, Abbate R, Gensini G, Casini A, Sofi 144. Kripke ML. Antigenicity of murine skin tumors 163. Albanes D, Heinonen OP, Huttunen JK, et al. Effects
F. Vegetarian, vegan diets and multiple health induced by ultraviolet light. J Natl Cancer Inst. 1974; of alpha-tocopherol and beta-carotene supplements
outcomes: a systematic review with meta-analysis 53(5):1333–1336. on cancer incidence in the Alpha-Tocopherol Beta-
of observational studies. Crit Rev Food Sci Nutr. 145. Fisher MS, Kripke ML. Systemic alteration induced Carotene Cancer Prevention Study. Am J Clin Nutr.
2016;6. in mice by ultraviolet light irradiation and its 1995;62(6 suppl):1427S–1430S.
125. Schwingshackl L, Hoffmann G. Does a relationship to ultraviolet carcinogenesis. Proc Natl 164. Alpha-Tocopherol, Beta Carotene Cancer Preven-
Mediterranean-type diet reduce cancer risk? Curr Acad Sci USA. 1977;74(4):1688–1692. tion Study Group. The effect of vitamin E and
Nutr Rep. 2016;5:9–17. 146. Kripke ML. Immunology and photocarcinogenesis. beta carotene on the incidence of lung cancer and
126. Filomeno M, Bosetti C, Bidoli E, et al. Mediter- New light on an old problem. J Am Acad Dermatol. other cancers in male smokers. N Engl J Med. 1994;
ranean diet and risk of endometrial cancer: a pooled 1986;14(1):149–155. 330(15):1029–1035.
analysis of three Italian case-control studies. Br J 147. Centers for Disease Control and Prevention (CDC). 165. Omenn GS, Goodman GE, Thornquist MD, et al.
Cancer. 2015;112(11):1816–1821. Sunburn and sun protective behaviors among adults Risk factors for lung cancer and for intervention
127. Toledo E, Salas-Salvadó J, Donat-Vargas C, et al. aged 18-29 years—United States, 2000–2010. effects in CARET, the Beta-Carotene and Retinol
Mediterranean Diet and invasive breast cancer risk MMWR Morb Mortal Wkly Rep. 2012;61(18): Efficacy Trial. J Natl Cancer Inst. 1996;88(21):
among women at high cardiovascular risk in the 317–322. 1550–1559.
PREDIMED Trial: a randomized clinical trial. JAMA 148. Buller DB, Cokkinides V, Hall HI, et al. Prevalence 166. Costa A, Formelli F, Chiesa F, Decensi A, De Palo G,
Intern Med. 2015;175(11):1752–1760. of sunburn, sun protection, and indoor tanning Veronesi U. Prospects of chemoprevention of human
128. International Agency for Research on Cancer behaviors among Americans: review from national cancers with the synthetic retinoid fenretinide.
(IARC). IARC Monographs on the Evaluation of surveys and case studies of 3 states. J Am Acad Cancer Res. 1994;54(7 suppl):2032s–2037s.
Carcinogenic Risks to Humans. IARC Monographs. Dermatol. 2011;65(5 suppl 1):S114–S123. 167. Omenn GS, Goodman GE, Thornquist MD, et al.
2009;100(Pt D). 149. van der Pols JC, Williams GM, Pandeya N, Logan Effects of a combination of beta carotene and vitamin
129. Guy GP Jr, Machlin SR, Ekwueme DU, Yabroff V, Green AC. Prolonged prevention of squamous A on lung cancer and cardiovascular disease. N Engl
KR. Prevalence and costs of skin cancer treatment cell carcinoma of the skin by regular sunscreen use. J Med. 1996;334(18):1150–1155.
in the U.S., 2002–2006 and 2007–2011. Am J Prev Cancer Epidemiol Biomarkers Prev. 2006;15(12): 168. Schrauzer GN, White DA, Schneider CJ. Cancer
Med. 2015;48(2):183–187. 2546–2548. mortality correlation studies—III: statistical associa-
130. U.S. Department of Health and Human Services. 150. Green AC, Williams GM, Logan V, Strutton tions with dietary selenium intakes. Bioinorg Chem.
The Surgeon General’s Call to Action to Prevent GM. Reduced melanoma after regular sunscreen 1977;7(1):23–31.
Skin Cancer. Washington D.C. 2014. use: randomized trial follow-up. J Clin Oncol. 169. Clark LC, Cantor KP, Allaway WH. Selenium in
131. Gandini S, Sera F, Cattaruzza MS, et al. Meta-analysis 2011;29(3):257–263. forage crops and cancer mortality in U.S. counties.
of risk factors for cutaneous melanoma: II. Sun 151. Lin JS, Eder M, Weinmann S. Behavioral counseling Arch Environ Health. 1991;46(1):37–42.
exposure. Eur J Cancer. 2005;41(1):45–60. to prevent skin cancer: a systematic review for the 170. Clark LC, Combs GF Jr, Turnbull BW, et al. Effects
132. Dennis LK, Vanbeek MJ, Beane Freeman LE, U.S. Preventive Services Task Force. Ann Intern Med. of selenium supplementation for cancer prevention
Smith BJ, Dawson DV, Coughlin JA. Sunburns 2011;154(3):190–201. in patients with carcinoma of the skin. A randomized
and risk of cutaneous melanoma: does age matter? 152. Diffey B. New sunscreens and the precautionary controlled trial. Nutritional Prevention of Cancer
A comprehensive meta-analysis. Ann Epidemiol. principle. JAMA Dermatol. 2016;152(5):511–512. Study Group. JAMA. 1996;276(24):1957–1963.
2008;18(8):614–627. 153. Thompson SC, Jolley D, Marks R. Reduction of 171. Reid ME, Duffield-Lillico AJ, Garland L, Turnbull
133. Chang YM, Barrett JH, Bishop DT, et al. Sun solar keratoses by regular sunscreen use. N Engl J BW, Clark LC, Marshall JR. Selenium supplementa-
exposure and melanoma risk at different latitudes: Med. 1993;329(16):1147–1151. tion and lung cancer incidence: an update of the
a pooled analysis of 5700 cases and 7216 controls. 154. Naylor MF, Boyd A, Smith DW, Cameron GS, Nutritional Prevention of Cancer Trial. Cancer
Int J Epidemiol. 2009;38(3):814–830. Hubbard D, Neldner KH. High sun protection factor Epidemiol Biomarkers Prev. 2002;11(11):1285–1291.
134. Macbeth AE, Grindlay DJ, Williams HC. What’s sunscreens in the suppression of actinic neoplasia. 172. Lippman SM, Klein EA, Goodman PJ, et al. Effect of
new in skin cancer? An analysis of guidelines and Arch Dermatol. 1995;131(2):170–175. selenium and vitamin E on risk of prostate cancer and
systematic reviews published in 2008-2009. Clin 155. Darlington S, Williams G, Neale R, Frost C, Green other cancers: the Selenium and Vitamin E Cancer
Exp Dermatol. 2011;36(5):453–458. A. A randomized controlled trial to assess sunscreen Prevention Trial (SELECT). JAMA. 2009;301(1):
135. Centers for Disease Control and Prevention (CDC). application and beta carotene supplementation in 39–51.
Use of indoor tanning devices by adults—United the prevention of solar keratoses. Arch Dermatol. 173. Klein EA, Thompson IM Jr, Tangen CM, et al.
States, 2010. MMWR Morb Mortal Wkly Rep. 2003;139(4):451–455. Vitamin E and the risk of prostate cancer: the
2012;61(18):323–326. 156. Green A, Williams G, Neale R, et al. Daily sunscreen Selenium and Vitamin E Cancer Prevention Trial
136. Ferrucci LM, Cartmel B, Molinaro AM, Leffell DJ, application and betacarotene supplementation in (SELECT). JAMA. 2011;306(14):1549–1556.
Bale AE, Mayne ST. Indoor tanning and risk of prevention of basal-cell and squamous-cell carci- 174. Karp DD, Lee SJ, Keller SM, et al. Randomized,
early-onset basal cell carcinoma. J Am Acad Dermatol. nomas of the skin: a randomised controlled trial. double-blind, placebo-controlled, phase III chemo-
2012;67(4):552–562. Lancet. 1999;354(9180):723–729. prevention trial of selenium supplementation in
137. Lazovich D, Vogel RI, Berwick M, Weinstock MA, 157. American Academy of Dermatology. Sunscreen patients with resected stage I non–small-cell lung
Anderson KE, Warshaw EM. Indoor tanning and FAQs; 2017. https://www.aad.org/media/stats/ cancer: ECOG 5597. J Clin Oncol. 2013;31(33):
risk of melanoma: a case-control study in a highly prevention-and-care/sunscreen-faqs. Accessed June 4179–4187.
exposed population. Cancer Epidemiol Biomarkers 22, 2017. 175. Veronesi G, Szabo E, Decensi A, et al. Randomized
Prev. 2010;19(6):1557–1568. 158. Jansen R, Osterwalder U, Wang SQ, Burnett M, phase II trial of inhaled budesonide versus placebo in
138. Gandini S, Stanganelli I, Magi S, et al. Melanoma Lim HW. Photoprotection: part II. Sunscreen: high-risk individuals with CT screen-detected lung
attributable to sunbed use and tan seeking behav- development, efficacy, and controversies. J Am Acad nodules. Cancer Prev Res (Phila). 2011;4(1):34–42.
iours: an Italian survey. Eur J Dermatol. 2014;24(1): Dermatol. 2013;69(6):867.e861–814; quiz 881–862. 176. Veronesi G, Lazzeroni M, Szabo E, et al. Long-term
35–40. 159. Domchek SM, Friebel TM, Singer CF, et al. Associa- effects of inhaled budesonide on screening-detected
139. Boniol M, Autier P, Boyle P, Gandini S. Cutaneous tion of risk-reducing surgery in BRCA1 or BRCA2 lung nodules. Ann Oncol. 2015;26(5):1025–1030.
melanoma attributable to sunbed use: systematic mutation carriers with cancer risk and mortality. 177. Limburg PJ, Mandrekar SJ, Aubry MC, et al.
review and meta-analysis. BMJ. 2012;345:e4757. JAMA. 2010;304(9):967–975. Randomized phase II trial of sulindac for lung
374.e4 Part I: Science and Clinical Oncology

cancer chemoprevention. Lung Cancer. 2013;79(3): cancer chemotherapeutic agent. In Vivo. 1990;4(3): trial in the People’s Republic of China. Cancer Res.
254–261. 153–160. 1988;48(8):2280–2283.
178. Keith RL, Blatchford PJ, Kittelson J, et al. Oral 196. Lippman SM, Batsakis JG, Toth BB, et al. 214. Lin P, Zhang J, Rong Z, et al. Studies on medi-
iloprost improves endobronchial dysplasia in former Comparison of low-dose isotretinoin with beta camentous inhibitory therapy for esophageal
smokers. Cancer Prev Res (Phila). 2011;4(6):793–802. carotene to prevent oral carcinogenesis. N Engl J precancerous lesions—3- and 5-year inhibitory
179. Gray JE, Altiok S, Alexandrow MG, et al. Phase 2 Med. 1993;328(1):15–20. effects of antitumor-B, retinamide and riboflavin.
randomized study of enzastaurin (LY317615) for 197. Chiesa F, Tradati N, Marazza M, et al. Fenretinide Proc Chin Acad Med Sci Peking Union Med Coll.
lung cancer prevention in former smokers. Cancer. (4-HPR) in chemoprevention of oral leukoplakia. 1990;5(3):121–129.
2013;119(5):1023–1032. J Cell Biochem Suppl. 1993;17F:255–261. 215. Li JY, Taylor PR, Li B, et al. Nutrition intervention
180. Lam S, McWilliams A, LeRiche J, MacAulay C, 198. Zaridze D, Evstifeeva T, Boyle P. Chemoprevention trials in Linxian, China: multiple vitamin/mineral
Wattenberg L, Szabo E. A phase I study of myo- of oral leukoplakia and chronic esophagitis in an supplementation, cancer incidence, and disease-
inositol for lung cancer chemoprevention. Cancer area of high incidence of oral and esophageal cancer. specific mortality among adults with esophageal dys-
Epidemiol Biomarkers Prev. 2006;15(8):1526–1531. Ann Epidemiol. 1993;3(3):225–234. plasia. J Natl Cancer Inst. 1993;85(18):1492–1498.
181. Lam S, Mandrekar SJ, Gesthalter Y, et al. A ran- 199. Pinto H, Li Y, Loprinzi C. Phase 3 trial of low-dose 216. Mark SD, Liu SF, Li JY, et al. The effect of vitamin
domized phase IIb trial of myo-inositol in smokers 13-cis-retinoic acid for prevention of second primary and mineral supplementation on esophageal cytology:
with bronchial dysplasia. Cancer Prev Res (Phila). cancers in stage I-II head and neck cancer—an results from the Linxian Dysplasia Trial. Int J Cancer.
2016;9(12):906–914. Eastern Cooperative Oncology Group study. Program 1994;57(2):162–166.
182. Lippman SM, Lee JJ, Karp DD, et al. Randomized and abstracts of the 37th Annual Meeting of the 217. Limburg PJ, Wei W, Ahnen DJ, et al. Randomized,
phase III intergroup trial of isotretinoin to prevent American Society of Clinical Oncology; May 12-15, placebo-controlled, esophageal squamous cell cancer
second primary tumors in stage I non–small-cell lung 2001, 2001; San Francisco, California. chemoprevention trial of selenomethionine and
cancer. J Natl Cancer Inst. 2001;93(8):605–618. 200. Khuri FR, Lee JJ, Lippman SM, et al. Randomized celecoxib. Gastroenterology. 2005;129(3):863–873.
183. Lee JJ, Feng L, Reshef DS, et al. Mortality in the phase III trial of low-dose isotretinoin for preven- 218. Gordon V, Jankowski J. Chemoprevention in Bar-
randomized, controlled lung intergroup trial of tion of second primary tumors in stage I and II rett’s oesophagus. Best Pract Res Clin Gastroenterol.
isotretinoin. Cancer Prev Res (Phila). 2010;3(6): head and neck cancer patients. J Natl Cancer Inst. 2011;25(4–5):569–579.
738–744. 2006;98(7):441–450. 219. Barrett NR. The lower esophagus lined by columnar
184. Kelly K, Kittelson J, Franklin WA, et al. A ran- 201. Lee JJ, Wu X, Hildebrandt MA, et al. Global assess- epithelium. Surgery. 1957;41(6):881–894.
domized phase II chemoprevention trial of 13-CIS ment of genetic variation influencing response to 220. Spechler SJ, Goyal RK. The columnar-lined
retinoic acid with or without alpha tocopherol or retinoid chemoprevention in head and neck cancer esophagus, intestinal metaplasia, and Norman
observation in subjects at high risk for lung cancer. patients. Cancer Prev Res (Phila). 2011;4(2):185–193. Barrett. Gastroenterology. 1996;110(2):614–621.
Cancer Prev Res (Phila). 2009;2(5):440–449. 202. Pinto H, Li Y, Loprinzi C, Phase III. Trial of low-dose 221. Haggitt RC. Barrett’s esophagus, dysplasia, and
185. Cardnell RJ, Feng Y, Diao L, et al. Proteomic markers 13-cis-retinoic acid for prevention of second primary adenocarcinoma. Hum Pathol. 1994;25(10):982–
of DNA repair and PI3K pathway activation predict cancers in stage I-II head and neck cancer—an 993.
response to the PARP inhibitor BMN 673 in small Eastern Cooperative Oncology Group Study. Proc 222. Sampliner RE. Effect of up to 3 years of high-dose
cell lung cancer. Clin Cancer Res. 2013;19(22): Am Soc Clin Oncol. 2001. lansoprazole on Barrett’s esophagus. Am J Gastroen-
6322–6328. 203. Grommes C, Landreth GE, Heneka MT. Antineo- terol. 1994;89(10):1844–1848.
186. Blumenthal GM, Zhang L, Zhang H, et al. Milestone plastic effects of peroxisome proliferator-activated 223. Lightdale CJ. Photodynamic therapy: a new light
analyses of immune checkpoint inhibitors, targeted receptor gamma agonists. Lancet Oncol. 2004;5(7): on Barrett’s esophagus. Gastrointest Endosc. 1995;
therapy, and conventional therapy in metastatic 419–429. 42(1):96–98.
non–small cell lung cancer trials: a meta-analysis. 204. Ondrey F. Peroxisome proliferator-activated recep- 224. Overholt BF, Panjehpour M. Photodynamic therapy
JAMA Oncol. 2017;e171029. tor gamma pathway targeting in carcinogenesis: for Barrett’s esophagus: clinical update. Am J
187. Hieda Y, Keyler DE, Vandevoort JT, et al. Active implications for chemoprevention. Clin Cancer Gastroenterol. 1996;91(9):1719–1723.
immunization alters the plasma nicotine concentra- Res. 2009;15(1):2–8. 225. Ouatu-Lascar R, Fitzgerald RC, Triadafilopoulos
tion in rats. J Pharmacol Exp Ther. 1997;283(3): 205. Yki-Jarvinen H. Thiazolidinediones. N Engl J Med. G. Differentiation and proliferation in Barrett’s
1076–1081. 2004;351(11):1106–1118. esophagus and the effects of acid suppression.
188. Pentel PR, Malin DH, Ennifar S, et al. A nicotine 206. Nissen SE, Nicholls SJ, Wolski K, et al. Comparison Gastroenterology. 1999;117(2):327–335.
conjugate vaccine reduces nicotine distribution of pioglitazone vs glimepiride on progression of 226. Cooper BT, Chapman W, Neumann CS, Gearty
to brain and attenuates its behavioral and cardio- coronary atherosclerosis in patients with type 2 JC. Continuous treatment of Barrett’s oesophagus
vascular effects in rats. Pharmacol Biochem Behav. diabetes: the PERISCOPE randomized controlled patients with proton pump inhibitors up to 13 years:
2000;65(1):191–198. trial. JAMA. 2008;299(13):1561–1573. observations on regression and cancer incidence.
189. Tyagi A, Singh RP, Ramasamy K, et al. Growth 207. William WN Jr, Papadimitrakopoulou V, Lee JJ, et al. Aliment Pharmacol Ther. 2006;23(6):727–733.
inhibition and regression of lung tumors by silibinin: Erlotinib and the risk of oral cancer: the Erlotinib 227. Souza RF, Shewmake K, Beer DG, Cryer B, Spechler
modulation of angiogenesis by macrophage- Prevention of Oral Cancer (EPOC) randomized SJ. Selective inhibition of cyclooxygenase-2 suppresses
associated cytokines and nuclear factor-kappaB and clinical trial. JAMA Oncol. 2016;2(2):209–216. growth and induces apoptosis in human esophageal
signal transducers and activators of transcription 3. 208. Armstrong WB, Taylor TH, Kennedy AR, et al. adenocarcinoma cells. Cancer Res. 2000;60(20):
Cancer Prev Res (Phila). 2009;2(1):74–83. Bowman Birk inhibitor concentrate and oral 5767–5772.
190. Yan Y, Wang Y, Tan Q, Lubet RA, You M. Efficacy leukoplakia: a randomized phase IIb trial. Cancer 228. Kaur BS, Khamnehei N, Iravani M, Namburu
of deguelin and silibinin on benzo(a)pyrene-induced Prev Res (Phila). 2013;6(5):410–418. SS, Lin O, Triadafilopoulos G. Rofecoxib inhibits
lung tumorigenesis in A/J mice. Neoplasia. 2005;7(12): 209. Monteiro de Oliveira Novaes JA, William WN Jr. cyclooxygenase 2 expression and activity and reduces
1053–1057. Prognostic factors, predictive markers and cancer cell proliferation in Barrett’s esophagus. Gastroenterol-
191. Hong WK, Endicott J, Itri LM, et al. 13-cis-retinoic biology: the triad for successful oral cancer chemo- ogy. 2002;123(1):60–67.
acid in the treatment of oral leukoplakia. N Engl J prevention. Future Oncol. 2016;12(20):2379–2386. 229. Corley DA, Kerlikowske K, Verma R, Buffler P.
Med. 1986;315(24):1501–1505. 210. William WN, Uraoka N, Peng SA, et al. Immune Protective association of aspirin/NSAIDs and
192. Hong WK, Lippman SM, Itri LM, et al. Preven- profiling of oral pre-malignant lesions (OPLs): an esophageal cancer: a systematic review and meta-
tion of second primary tumors with isotretinoin in Erlotinib Prevention of Oral Cancer (EPOC) study analysis. Gastroenterology. 2003;124(1):47–56.
squamous-cell carcinoma of the head and neck. N biobank analysis. J Clin Oncol. 2017;2017. 230. Abnet CC, Freedman ND, Kamangar F, Leitzmann
Engl J Med. 1990;323(12):795–801. 211. Munoz N, Wahrendorf J, Bang LJ, et al. No effect MF, Hollenbeck AR, Schatzkin A. Non-steroidal
193. Stich HF, Hornby AP, Mathew B, Sankaranaray- of riboflavine, retinol, and zinc on prevalence of anti-inflammatory drugs and risk of gastric and
anan R, Nair MK. Response of oral leukoplakias precancerous lesions of oesophagus. Randomised oesophageal adenocarcinomas: results from a cohort
to the administration of vitamin A. Cancer Lett. double-blind intervention study in high-risk popula- study and a meta-analysis. Br J Cancer. 2009;100(3):
1988;40(1):93–101. tion of China. Lancet. 1985;2(8447):111–114. 551–557.
194. Stich HF, Rosin MP, Hornby AP, Mathew B, 212. Munoz N, Hayashi M, Bang LJ, Wahrendorf J, 231. Heath EI, Canto MI, Piantadosi S, et al. Second-
Sankaranarayanan R, Nair MK. Remission of Crespi M, Bosch FX. Effect of riboflavin, retinol, and ary chemoprevention of Barrett’s esophagus with
oral leukoplakias and micronuclei in tobacco/ zinc on micronuclei of buccal mucosa and of esopha- celecoxib: results of a randomized trial. J Natl Cancer
betel quid chewers treated with beta-carotene and gus: a randomized double-blind intervention study Inst. 2007;99(7):545–557.
with beta-carotene plus vitamin A. Int J Cancer. in China. J Natl Cancer Inst. 1987;79(4):687–691. 232. Shar AO, Gaudard MA, Heath EI, Forastiere AA,
1988;42(2):195–199. 213. Wahrendorf J, Munoz N, Lu JB, Thurnham DI, Yang VW, Sontag SJ. Modeling using baseline
195. Han J, Jiao L, Lu Y, Sun Z, Gu QM, Scanlon Crespi M, Bosch FX. Blood, retinol and zinc characteristics in a small multicenter clinical trial for
KJ. Evaluation of N-4-(hydroxycarbophenyl) riboflavin status in relation to precancerous lesions of Barrett’s esophagus. Contemp Clin Trials. 2009;30(1):
retinamide as a cancer prevention agent and as a the esophagus: findings from a vitamin intervention 2–7.
Lifestyle and Cancer Prevention  •  CHAPTER 22 374.e5
374.e5

233. Falk GW, Buttar NS, Foster NR, et al. A combination risk of colorectal cancer: primary analysis from the review for the U.S. Preventive Services Task Force.
of esomeprazole and aspirin reduces tissue concentra- Women’s Health Initiative Observational Study and Rockville (MD) 2015.
tions of prostaglandin E(2) in patients with Barrett’s meta-analysis. Cancer Epidemiol Biomarkers Prev. 272. Lotrionte M, Biasucci LM, Peruzzi M, Frati G,
esophagus. Gastroenterology. 2012;143(4):917–926. 2011;20(9):1822–1830. Giordano A, Biondi-Zoccai G. Which aspirin dose
e911. 253. Thompson P, Roe DJ, Fales L, et al. Design and and preparation is best for the long-term prevention
234. Vaughan TL, Dong LM, Blount PL, et al. Non- baseline characteristics of participants in a phase of cardiovascular disease and cancer? Evidence from
steroidal anti-inflammatory drugs and risk of III randomized trial of celecoxib and selenium for a systematic review and network meta-analysis. Prog
neoplastic progression in Barrett’s oesophagus: a pro- colorectal adenoma prevention. Cancer Prev Res Cardiovasc Dis. 2016;58(5):495–504.
spective study. Lancet Oncol. 2005;6(12):945–952. (Phila). 2012;5(12):1381–1393. 273. Zhao TY, Tu J, Wang Y, et al. The efficacy of aspirin
235. Anderson LA, Johnston BT, Watson RG, et al. 254. Arber N, Eagle CJ, Spicak J, et al. Celecoxib for in preventing the recurrence of colorectal adenoma:
Nonsteroidal anti-inflammatory drugs and the the prevention of colorectal adenomatous polyps. a renewed meta-analysis of randomized trials. Asian
esophageal inflammation-metaplasia-adenocarcinoma N Engl J Med. 2006;355(9):885–895. Pac J Cancer Prev. 2016;17(5):2711–2717.
sequence. Cancer Res. 2006;66(9):4975–4982. 255. Bertagnolli MM, Eagle CJ, Zauber AG, et al. 274. Steinbach G, Lynch PM, Phillips RK, et al. The
236. Nguyen DM, Richardson P, El-Serag HB. Medica- Celecoxib for the prevention of sporadic colorectal effect of celecoxib, a cyclooxygenase-2 inhibitor,
tions (NSAIDs, statins, proton pump inhibitors) adenomas. N Engl J Med. 2006;355(9):873–884. in familial adenomatous polyposis. N Engl J Med.
and the risk of esophageal adenocarcinoma in 256. Solomon SD, Wittes J, Finn PV, et al. Cardiovascular 2000;342(26):1946–1952.
patients with Barrett’s esophagus. Gastroenterology. risk of celecoxib in 6 randomized placebo-controlled 275. Phillips RK, Wallace MH, Lynch PM, et al. A
2010;138(7):2260–2266. trials: the cross trial safety analysis. Circulation. randomised, double blind, placebo controlled study
237. Jayaprakash V, Menezes RJ, Javle MM, et al. Regular 2008;117(16):2104–2113. of celecoxib, a selective cyclooxygenase 2 inhibitor,
aspirin use and esophageal cancer risk. Int J Cancer. 257. Thompson PA, Ashbeck EL, Roe DJ, et al. Selenium on duodenal polyposis in familial adenomatous
2006;119(1):202–207. Supplementation for prevention of colorectal polyposis. Gut. 2002;50(6):857–860.
238. Buttar NS, Wang KK, Anderson MA, et al. The adenomas and risk of associated type 2 diabetes. 276. Bertagnolli MM, Eagle CJ, Zauber AG, et al. Five-
effect of selective cyclooxygenase-2 inhibition in J Natl Cancer Inst. 2016;108(12). year efficacy and safety analysis of the Adenoma
Barrett’s esophagus epithelium: an in vitro study. 258. Cook NR, Lee IM, Gaziano JM, et al. Low-dose Prevention with Celecoxib Trial. Cancer Prev Res
J Natl Cancer Inst. 2002;94(6):422–429. aspirin in the primary prevention of cancer: the (Phila). 2009;2(4):310–321.
239. Jankowski JA, Hooper PA. Chemoprevention in Women’s Health Study: a randomized controlled 277. Lynch PM, Ayers GD, Hawk E, et al. The safety and
Barrett’s esophagus: a pill a day? Gastrointest Endosc trial. JAMA. 2005;294(1):47–55. efficacy of celecoxib in children with familial adeno-
Clin N Am. 2011;21(1):155–170. 259. Sturmer T, Glynn RJ, Lee IM, Manson JE, Buring JE, matous polyposis. Am J Gastroenterol. 2010;105(6):
240. Das D, Chilton AP, Jankowski JA. Chemoprevention Hennekens CH. Aspirin use and colorectal cancer: 1437–1443.
of oesophageal cancer and the AspECT trial. Recent post-trial follow-up data from the Physicians’ Health 278. Giovannucci E, Goldin B. The role of fat, fatty acids,
Results Cancer Res. 2009;181:161–169. Study. Ann Intern Med. 1998;128(9):713–720. and total energy intake in the etiology of human
241. Sanders DSA, Grabsch H, Harrison R, et al. Compar- 260. Cook NR, Lee IM, Zhang SM, Moorthy MV, Buring colon cancer. Am J Clin Nutr. 1997;66(6 suppl):
ing virtual with conventional microscopy for the JE. Alternate-day, low-dose aspirin and cancer risk: 1564S–1571S.
consensus diagnosis of Barrett’s neoplasia in the long-term observational follow-up of a randomized 279. Narisawa T, Magadia NE, Weisburger JH, Wynder
AspECT Barrett’s chemoprevention trial pathology trial. Ann Intern Med. 2013;159(2):77–85. EL. Promoting effect of bile acids on colon carci-
audit. Histopathology. 2012;61(5):795–800. 261. Burn J, Bishop DT, Chapman PD, et al. A random- nogenesis after intrarectal instillation of N-methyl-
242. Janakiram NB, Rao CV. The role of inflammation ized placebo-controlled prevention trial of aspirin N′-nitro-N-nitrosoguanidine in rats. J Natl Cancer
in colon cancer. Adv Exp Med Biol. 2014;816:25–52. and/or resistant starch in young people with familial Inst. 1974;53(4):1093–1097.
243. Knekt P, Aromaa A, Maatela J, et al. Serum vitamin adenomatous polyposis. Cancer Prev Res (Phila). 280. Bautista D, Obrador A, Moreno V, et al. Ki-ras muta-
E, serum selenium and the risk of gastrointestinal 2011;4(5):655–665. tion modifies the protective effect of dietary mono-
cancer. Int J Cancer. 1988;42(6):846–850. 262. Burn J, Bishop DT, Mecklin JP, et al. Effect of aspirin unsaturated fat and calcium on sporadic colorectal
244. Knekt P, Aromaa A, Maatela J, et al. Serum or resistant starch on colorectal neoplasia in the Lynch cancer. Cancer Epidemiol Biomarkers Prev. 1997;6(1):
selenium and subsequent risk of cancer among syndrome. N Engl J Med. 2008;359(24):2567–2578. 57–61.
Finnish men and women. J Natl Cancer Inst. 1990; 263. Burn J, Gerdes AM, Macrae F, et al. Long-term 281. Peters U, Chatterjee N, McGlynn KA, et al. Calcium
82(10):864–868. effect of aspirin on cancer risk in carriers of intake and colorectal adenoma in a US colorectal cancer
245. Clark LC, Hixson LJ, Combs GF Jr, Reid ME, hereditary colorectal cancer: an analysis from the early detection program. Am J Clin Nutr. 2004;80(5):
Turnbull BW, Sampliner RE. Plasma selenium CAPP2 randomised controlled trial. Lancet. 2011; 1358–1365.
concentration predicts the prevalence of colorectal 378(9809):2081–2087. 282. Kesse E, Boutron-Ruault MC, Norat T, Riboli E,
adenomatous polyps. Cancer Epidemiol Biomarkers 264. Logan RF, Grainge MJ, Shepherd VC, Armitage Clavel-Chapelon F, E3N Group. Dietary calcium,
Prev. 1993;2(1):41–46. NC, Muir KR. ukCAP Trial Group. Aspirin and phosphorus, vitamin D, dairy products and the risk
246. Lee YW, Klein CB, Kargacin B, et al. Carcinogenic folic acid for the prevention of recurrent colorectal of colorectal adenoma and cancer among French
nickel silences gene expression by chromatin con- adenomas. Gastroenterology. 2008;134(1):29–38. women of the E3N-EPIC prospective study. Int J
densation and DNA methylation: a new model for 265. Benamouzig R, Deyra J, Martin A, et al. Daily Cancer. 2005;117(1):137–144.
epigenetic carcinogens. Mol Cell Biol. 1995;15(5): soluble aspirin and prevention of colorectal adenoma 283. Martinez ME, Willett WC. Calcium, vitamin D,
2547–2557. recurrence: one-year results of the APACC trial. and colorectal cancer: a review of the epidemio-
247. Duffield-Lillico AJ, Reid ME, Turnbull BW, Gastroenterology. 2003;125(2):328–336. logic evidence. Cancer Epidemiol Biomarkers Prev.
et al. Baseline characteristics and the effect of 266. Baron JA, Cole BF, Sandler RS, et al. A random- 1998;7(2):163–168.
selenium supplementation on cancer incidence in ized trial of aspirin to prevent colorectal adenomas. 284. Flood A, Peters U, Chatterjee N, Lacey JV Jr, Schairer
a randomized clinical trial: a summary report of N Engl J Med. 2003;348(10):891–899. C, Schatzkin A. Calcium from diet and supplements
the Nutritional Prevention of Cancer Trial. Cancer 267. Benamouzig R, Uzzan B, Deyra J, et al. Preven- is associated with reduced risk of colorectal cancer
Epidemiol Biomarkers Prev. 2002;11(7):630–639. tion by daily soluble aspirin of colorectal adenoma in a prospective cohort of women. Cancer Epidemiol
248. Lance P, Alberts DS, Thompson PA, et  al. recurrence: 4-year results of the APACC randomised Biomarkers Prev. 2005;14(1):126–132.
Colorectal adenomas in participants of the SELECT trial. Gut. 2012;61(2):255–261. 285. McCullough ML, Robertson AS, Rodriguez C, et al.
randomized trial of selenium and vitamin E for 268. Sandler RS, Halabi S, Baron JA, et al. A randomized Calcium, vitamin D, dairy products, and risk of
prostate cancer prevention. Cancer Prev Res (Phila). trial of aspirin to prevent colorectal adenomas in colorectal cancer in the Cancer Prevention Study
2017;10(1):45–54. patients with previous colorectal cancer. N Engl J II Nutrition Cohort (United States). Cancer Causes
249. Vinceti M, Dennert G, Crespi CM, et al. Selenium Med. 2003;348(10):883–890. Control. 2003;14(1):1–12.
for preventing cancer. Cochrane Database Syst Rev. 269. Flossmann E, Rothwell PM. British Doctors Aspirin 286. Terry P, Baron JA, Bergkvist L, Holmberg L, Wolk
2014;(3):CD005195. Trial and the UK-TIA Aspirin Trial. Effect of aspirin A. Dietary calcium and vitamin D intake and risk
250. Stranges S, Marshall JR, Natarajan R, et al. Effects on long-term risk of colorectal cancer: consistent of colorectal cancer: a prospective cohort study in
of long-term selenium supplementation on the evidence from randomised and observational studies. women. Nutr Cancer. 2002;43(1):39–46.
incidence of type 2 diabetes: a randomized trial. Lancet. 2007;369(9573):1603–1613. 287. Baron JA, Beach M, Mandel JS, et al. Calcium
Ann Intern Med. 2007;147(4):217–223. 270. Drew DA, Chin SM, Gilpin KK, et al. ASPirin supplements for the prevention of colorectal
251. Goodman PJ, Tangen CM, Darke AK, et al. Oppor- Intervention for the REDuction of colorectal cancer adenomas. Calcium Polyp Prevention Study Group.
tunities and challenges in incorporating ancillary risk (ASPIRED): a study protocol for a randomized N Engl J Med. 1999;340(2):101–107.
studies into a cancer prevention randomized clinical controlled trial. Trials. 2017;18(1):50. 288. Bonithon-Kopp C, Kronborg O, Giacosa A, Rath
trial. Trials. 2016;17:400. 271. Chubak J, Kamineni A, Buist DSM, Anderson U, Faivre J. Calcium and fibre supplementation
252. Takata Y, Kristal AR, King IB, et al. Serum ML, Whitlock EP. Aspirin use for the prevention in prevention of colorectal adenoma recurrence: a
selenium, genetic variation in selenoenzymes, and of colorectal cancer: an updated systematic evidence randomised intervention trial. European Cancer
374.e6 Part I: Science and Clinical Oncology

Prevention Organisation Study Group. Lancet. 2000; 307. Sterverding D. Sleeping sickness and nagana disease antioxidant supplements and anti–Helicobacter pylori
356(9238):1300–1306. caused by Trypanosoma brucei. In: Marcondes CB, therapy. J Natl Cancer Inst. 2000;92(23):1881–1888.
289. Wactawski-Wende J, Kotchen JM, Anderson GL, ed. Arthropod Borne Diseases. Switzerland: Springer 326. Wong BC, Lam SK, Wong WM, et al. Helicobacter
et al. Calcium plus vitamin D supplementation and International Publishing; 2017:292. pylori eradication to prevent gastric cancer in a high-
the risk of colorectal cancer. N Engl J Med. 2006; 308. World Health Organization (WHO). WHO Model risk region of China: a randomized controlled trial.
354(7):684–696. List of Essential Medicines; 2015; 19. http://www. JAMA. 2004;291(2):187–194.
290. Cauley JA, Chlebowski RT, Wactawski-Wende J, who.int/medicines/publications/essentialmedicines/ 327. You WC, Brown LM, Zhang L, et al. Randomized
et al. Calcium plus vitamin D supplementation and EML_2015_FINAL_amended_NOV2015.pdf?ua=1. double-blind factorial trial of three treatments to
health outcomes five years after active intervention 309. Raul F. Revival of 2-(difluoromethyl)ornithine reduce the prevalence of precancerous gastric lesions.
ended: the Women’s Health Initiative. J Womens (DFMO), an inhibitor of polyamine biosynthesis, J Natl Cancer Inst. 2006;98(14):974–983.
Health (Larchmt). 2013;22(11):915–929. as a cancer chemopreventive agent. Biochem Soc 328. Sung JJ, Lin SR, Ching JY, et al. Atrophy and
291. Baron JA, Barry EL, Mott LA, et al. A trial of calcium Trans. 2007;35(Pt 2):353–355. intestinal metaplasia one year after cure of H.
and vitamin D for the prevention of colorectal 310. Giardiello FM, Hamilton SR, Hylind LM, Yang pylori infection: a prospective, randomized study.
adenomas. N Engl J Med. 2015;373(16):1519–1530. VW, Tamez P, Casero RA Jr. Ornithine decarboxylase Gastroenterology. 2000;119(1):7–14.
292. Barry EL, Peacock JL, Rees JR, et al. Vitamin D and polyamines in familial adenomatous polyposis. 329. Leung WK, Lin SR, Ching JY, et al. Factors pre-
receptor genotype, vitamin D3 supplementation, and Cancer Res. 1997;57(2):199–201. dicting progression of gastric intestinal metaplasia:
risk of colorectal adenomas: a randomized clinical 311. Gerner EW, Meyskens FL Jr. Polyamines and cancer: results of a randomised trial on Helicobacter pylori
trial. JAMA Oncol. 2017;3(5):628–635. old molecules, new understanding. Nat Rev Cancer. eradication. Gut. 2004;53(9):1244–1249.
293. Bonovas S, Fiorino G, Lytras T, Malesci A, Danese 2004;4(10):781–792. 330. Fukase K, Kato M, Kikuchi S, et al. Effect of eradica-
S. Calcium supplementation for the prevention of 312. Wallace HM, Fraser AV. Inhibitors of poly- tion of Helicobacter pylori on incidence of metachro-
colorectal adenomas: a systematic review and meta- amine metabolism: review article. Amino Acids. nous gastric carcinoma after endoscopic resection
analysis of randomized controlled trials. World J 2004;26(4):353–365. of early gastric cancer: an open-label, randomised
Gastroenterol. 2016;22(18):4594–4603. 313. Bassiri H, Benavides A, Haber M, Gilmour SK, controlled trial. Lancet. 2008;372(9636):392–397.
294. MacLennan SC, MacLennan AH, Ryan P. Colorectal Norris MD, Hogarty MD. Translational development 331. Mera R, Fontham ET, Bravo LE, et al. Long term
cancer and oestrogen replacement therapy. A meta- of difluoromethylornithine (DFMO) for the treat- follow up of patients treated for Helicobacter pylori
analysis of epidemiological studies. Med J Aust. 1995; ment of neuroblastoma. Transl Pediatr. 2015;4(3): infection. Gut. 2005;54(11):1536–1540.
162(9):491–493. 226–238. 332. Mera RM, Bravo LE, Camargo MC, et al. Dynamics
295. Hebert-Croteau N. A meta-analysis of hormone 314. Ye C, Geng Z, Dominguez D, et al. Targeting orni- of Helicobacter pylori infection as a determinant of
replacement therapy and colon cancer in women. thine decarboxylase by alpha-difluoromethylornithine progression of gastric precancerous lesions: 16-year
Cancer Epidemiol Biomarkers Prev. 1998;7(8): inhibits tumor growth by impairing myeloid-derived follow-up of an eradication trial. Gut. 2017.
653–659. suppressor cells. J Immunol. 2016;196(2):915–923. 333. Ma JL, Zhang L, Brown LM, et al. Fifteen-year
296. Grodstein F, Newcomb PA, Stampfer MJ. Postmeno- 315. Meyskens FL Jr, McLaren CE, Pelot D, et al. effects of Helicobacter pylori, garlic, and vitamin
pausal hormone therapy and the risk of colorectal Difluoromethylornithine plus sulindac for the treatments on gastric cancer incidence and mortality.
cancer: a review and meta-analysis. Am J Med. prevention of sporadic colorectal adenomas: a J Natl Cancer Inst. 2012;104(6):488–492.
1999;106(5):574–582. randomized placebo-controlled, double-blind trial. 334. Lau CS, Ward A, Chamberlain RS. Probiotics
297. Nanda K, Bastian LA, Hasselblad V, Simel DL. Cancer Prev Res (Phila). 2008;1(1):32–38. improve the efficacy of standard triple therapy in the
Hormone replacement therapy and the risk of 316. Zell JA, McLaren CE, Chen WP, Thompson eradication of Helicobacter pylori: a meta-analysis.
colorectal cancer: a meta-analysis. Obstet Gynecol. PA, Gerner EW, Meyskens FL. Ornithine Infect Drug Resist. 2016;9:275–289.
1999;93(5 Pt 2):880–888. decarboxylase-1 polymorphism, chemoprevention 335. Fock KM, Talley NJ, Fass R, et al. Asia-Pacific Con-
298. Rossouw JE, Anderson GL, Prentice RL, et al. Risks with eflornithine and sulindac, and outcomes among sensus on the management of gastroesophageal reflux
and benefits of estrogen plus progestin in healthy colorectal adenoma patients. J Natl Cancer Inst. 2010; disease: update. J Gastroenterol Hepatol. 2008;23(1):
postmenopausal women: principal results from the 102(19):1513–1516. 8–22.
Women’s Health Initiative randomized controlled 317. MacLean CH, Newberry SJ, Mojica WA, et al. 336. O’Connor A, O’Morain CA, Ford AC. Popula-
trial. JAMA. 2002;288(3):321–333. Effects of omega-3 fatty acids on cancer risk: a tion screening and treatment of Helicobacter pylori
299. Tsilidis KK, Allen NE, Key TJ, et al. Menopausal systematic review. JAMA. 2006;295(4):403–415. infection. Nat Rev Gastroenterol Hepatol. 2017;14(4):
hormone therapy and risk of colorectal cancer in 318. West NJ, Clark SK, Phillips RK, et al. Eicosa- 230–240.
the European Prospective Investigation into Cancer pentaenoic acid reduces rectal polyp number 337. Dawsey SM, Mark SD, Taylor PR, Limburg PJ.
and Nutrition. Int J Cancer. 2011;128(8):1881– and size in familial adenomatous polyposis. Gut. Gastric cancer and H. pylori. Gut. 2002;51(3):
1889. 2010;59(7):918–925. 457–458.
300. Chlebowski RT, Wactawski-Wende J, Ritenbaugh 319. Hull MA, Sandell AC, Montgomery AA, et al. A 338. Kamangar F, Dawsey SM, Blaser MJ, et al. Oppos-
C, et al. Estrogen plus progestin and colorectal randomized controlled trial of eicosapentaenoic acid ing risks of gastric cardia and noncardia gastric
cancer in postmenopausal women. N Engl J Med. and/or aspirin for colorectal adenoma prevention adenocarcinomas associated with Helicobacter pylori
2004;350(10):991–1004. during colonoscopic surveillance in the NHS Bowel seropositivity. J Natl Cancer Inst. 2006;98(20):1445–
301. Prentice RL, Pettinger M, Beresford SA, et al. Cancer Screening Programme (The seAFOod Polyp 1452.
Colorectal cancer in relation to postmenopausal Prevention Trial): study protocol for a randomized 339. Hansen S, Melby KK, Aase S, Jellum E, Vollset SE.
estrogen and estrogen plus progestin in the controlled trial. Trials. 2013;14:237. Helicobacter pylori infection and risk of cardia cancer
Women’s Health Initiative clinical trial and obser- 320. Hosono K, Endo H, Takahashi H, et al. Metfor- and non-cardia gastric cancer. A nested case-control
vational study. Cancer Epidemiol Biomarkers Prev. min suppresses colorectal aberrant crypt foci in a study. Scand J Gastroenterol. 1999;34(4):353–360.
2009;18(5):1531–1537. short-term clinical trial. Cancer Prev Res (Phila). 340. Helicobacter and Cancer Collaborative Group.
302. Anderson GL, Limacher M, Assaf AR, et al. Effects of 2010;3(9):1077–1083. Gastric cancer and Helicobacter pylori: a combined
conjugated equine estrogen in postmenopausal women 321. Higurashi T, Hosono K, Takahashi H, et al. analysis of 12 case control studies nested within
with hysterectomy: the Women’s Health Initiative Metformin for chemoprevention of metachronous prospective cohorts. Gut. 2001;49(3):347–353.
randomized controlled trial. JAMA. 2004;291(14): colorectal adenoma or polyps in post-polypectomy 341. Cavaleiro-Pinto M, Peleteiro B, Lunet N, Barros
1701–1712. patients without diabetes: a multicentre double- H. Helicobacter pylori infection and gastric cardia
303. Ritenbaugh C, Stanford JL, Wu L, et al. Conjugated blind, placebo-controlled, randomised phase 3 trial. cancer: systematic review and meta-analysis. Cancer
equine estrogens and colorectal cancer incidence and Lancet Oncol. 2016;17(4):475–483. Causes Control. 2011;22(3):375–387.
survival: the Women’s Health Initiative randomized 322. Kimura T, McKolanis JR, Dzubinski LA, et al. 342. Carrera-Lasfuentes P, Lanas A, Bujanda L, et al.
clinical trial. Cancer Epidemiol Biomarkers Prev. MUC1 vaccine for individuals with advanced Relevance of DNA repair gene polymorphisms
2008;17(10):2609–2618. adenoma of the colon: a cancer immunoprevention to gastric cancer risk and phenotype. Oncotarget.
304. Lavasani S, Chlebowski RT, Prentice RL, et al. feasibility study. Cancer Prev Res (Phila). 2013;6(1): 2017;8(22):35848–35862.
Estrogen and colorectal cancer incidence and 18–26. 343. Ma J, Wu D, Hu X, Li J, Cao M, Dong W. Associa-
mortality. Cancer. 2015;121(18):3261–3271. 323. Parkin DM. The global health burden of infection- tions between cytokine gene polymorphisms and
305. Samadi AK, Bilsland A, Georgakilas AG, et al. A associated cancers in the year 2002. Int J Cancer. susceptibility to Helicobacter pylori infection and
multi-targeted approach to suppress tumor-promoting 2006;118(12):3030–3044. Helicobacter pylori related gastric cancer, peptic ulcer
inflammation. Semin Cancer Biol. 2015;35(suppl): 324. Zabala Torres B, Lucero Y, Lagomarcino AJ, et al. disease: a meta-analysis. PLoS ONE. 2017;12(4):
S151–S184. Prevalence and dynamics of Helicobacter pylori e0176463.
306. Li Y, Zhang T. Targeting cancer stem cells by cur- infection during childhood. Helicobacter. 2017. 344. Melchiades JL, Zabaglia LM, Sallas ML, et al.
cumin and clinical applications. Cancer Lett. 2014; 325. Correa P, Fontham ET, Bravo JC, et al. Chemo- Polymorphisms and haplotypes of the interleukin
346(2):197–205. prevention of gastric dysplasia: randomized trial of 2 gene are associated with an increased risk of gastric
Lifestyle and Cancer Prevention  •  CHAPTER 22 374.e7
374.e7

cancer. The possible involvement of Helicobacter 361. Lavanchy D. The global burden of hepatitis C. Liver international symposium on hepatitis C virus and
pylori. Cytokine. 2017;96:203–207. Int. 2009;29(suppl 1):74–81. related viruses. J Hepatol. 2016;64(4):968–973.
345. Chang YW, Oh CH, Kim JW, et al. Combination 362. Benvegnu L, Fattovich G, Noventa F, et al. Concur- 383. Lau DT, Everhart J, Kleiner DE, et al. Long-term
of Helicobacter pylori infection and the interleukin rent hepatitis B and C virus infection and risk of follow-up of patients with chronic hepatitis B treated
8 -251 T > A polymorphism, but not the mannose- hepatocellular carcinoma in cirrhosis. A prospective with interferon alfa. Gastroenterology. 1997;113(5):
binding lectin 2 codon 54 G > A polymorphism, study. Cancer. 1994;74(9):2442–2448. 1660–1667.
might be a risk factor of gastric cancer. BMC Cancer. 363. Chiaramonte M, Stroffolini T, Vian A, et al. 384. Yang YF, Zhao W, Zhong YD, Xia HM, Shen L,
2017;17(1):388. Rate of incidence of hepatocellular carcinoma in Zhang N. Interferon therapy in chronic hepatitis
346. Zhang JZ, Liu CM, Peng HP, Zhang Y. Association patients with compensated viral cirrhosis. Cancer. B reduces progression to cirrhosis and hepatocel-
of genetic variations in IL-6/IL-6R pathway genes 1999;85(10):2132–2137. lular carcinoma: a meta-analysis. J Viral Hepat.
with gastric cancer risk in a Chinese population. 364. Ross RK, Yuan JM, Yu MC, et al. Urinary aflatoxin 2009;16(4):265–271.
Gene. 2017;623:1–4. biomarkers and risk of hepatocellular carcinoma. 385. Yoshida H, Shiratori Y, Moriyama M, et al. Inter-
347. Ribeiro RX, Nascimento C, Silva A. Genotype Lancet. 1992;339(8799):943–946. feron therapy reduces the risk for hepatocellular
association Gstm1 Null and gastric cancer: evidence- 365. Wang P, Kang D, Cao W, Wang Y, Liu Z. Diabetes carcinoma: national surveillance program of cirrhotic
based meta-analysis. Arq Gastroenterol. 2017; mellitus and risk of hepatocellular carcinoma: a and noncirrhotic patients with chronic hepatitis
54(2):101–108. systematic review and meta-analysis. Diabetes Metab C in Japan. IHIT Study Group. Inhibition of
348. Negovan A, Iancu M, Moldovan V, Mocan S, Banescu Res Rev. 2012;28(2):109–122. Hepatocarcinogenesis by Interferon Therapy. Ann
C. The Interaction between GSTT1, GSTM1, 366. Forner A, Llovet JM, Bruix J. Hepatocellular Intern Med. 1999;131(3):174–181.
and GSTP1 Ile105Val gene polymorphisms and carcinoma. Lancet. 2012;379(9822):1245–1255. 386. Imai Y, Kawata S, Tamura S, et al. Relation of
environmental risk factors in premalignant gastric 367. Leung N. HBV and liver cancer. Med J Malaysia. interferon therapy and hepatocellular carcinoma in
lesions risk. Biomed Res Int. 2017;2017:7365080. 2005;60(supplB):63–66. patients with chronic hepatitis C. Osaka Hepatocel-
349. Cherati MR, Shokri-Shirvani J, Karkhah A, Rajabnia 368. Chang MH, Chen CJ, Lai MS, et al. Universal lular Carcinoma Prevention Study Group. Ann Intern
R, Nouri HR. Helicobacter pylori cagL amino acid hepatitis B vaccination in Taiwan and the incidence Med. 1998;129(2):94–99.
polymorphism D58E59 pave the way toward of hepatocellular carcinoma in children. Taiwan 387. Ikeda K, Saitoh S, Arase Y, et al. Effect of inter-
peptic ulcer disease while N58E59 is associated Childhood Hepatoma Study Group. N Engl J Med. feron therapy on hepatocellular carcinogenesis in
with gastric cancer in north of Iran. Microb Pathog. 1997;336(26):1855–1859. patients with chronic hepatitis type C: a long-term
2017;107:413–418. 369. Schuppan D, Afdhal NH. Liver cirrhosis. Lancet. observation study of 1,643 patients using statistical
350. He BS, Sun HL, Xu T, et al. Association of genetic 2008;371(9615):838–851. bias correction with proportional hazard analysis.
polymorphisms in the LncRNAs with gastric cancer 370. Turner PC, Sylla A, Gong YY, et al. Reduction Hepatology. 1999;29(4):1124–1130.
risk in a Chinese population. J Cancer. 2017;8(4): in exposure to carcinogenic aflatoxins by post- 388. Shindo M, Ken A, Okuno T. Varying incidence of
531–536. harvest intervention measures in west Africa: a cirrhosis and hepatocellular carcinoma in patients
351. Giraldi L, Michelazzo MB, Arzani D, Persiani R, community-based intervention study. Lancet. 2005; with chronic hepatitis C responding differently to
Pastorino R, Boccia S. MUC1, MUC5AC, and 365(9475):1950–1956. interferon therapy. Cancer. 1999;85(9):1943–1950.
MUC6 polymorphisms, Helicobacter pylori infec- 371. El-Serag HB, Hampel H, Javadi F. The association 389. Miyake Y, Takaki A, Iwasaki Y, Yamamoto K. Meta-
tion, and gastric cancer: a systematic review and between diabetes and hepatocellular carcinoma: a analysis: interferon-alpha prevents the recurrence
meta-analysis. Eur J Cancer Prev. 2017. systematic review of epidemiologic evidence. Clin after curative treatment of hepatitis C virus–related
352. Tongtawee T, Bartpho T, Kaewpitoon S, et al. Gastroenterol Hepatol. 2006;4(3):369–380. hepatocellular carcinoma. J Viral Hepat. 2010;17(4):
Genetic polymorphisms in TLR1, TLR2, TLR4, 372. Dunn BK, Kramer BS. Cancer prevention: lessons 287–292.
and TLR10 of Helicobacter pylori–associated gastritis: learned and future directions. Trends Cancer. 2016; 390. Liaw YF, Sung JJ, Chow WC, et al. Lamivudine for
a prospective cross-sectional study in Thailand. Eur 2(12):713–722. patients with chronic hepatitis B and advanced liver
J Cancer Prev. 2017. 373. Glauert HP, Calfee-Mason K, Stemm DN, Tharappel disease. N Engl J Med. 2004;351(15):1521–1531.
353. Pan KF, Zhang L, Gerhard M, et al. A large JC, Spear BT. Dietary antioxidants in the prevention 391. Matsumoto A, Tanaka E, Rokuhara A, et al. Effi-
randomised controlled intervention trial to prevent of hepatocarcinogenesis: a review. Mol Nutr Food cacy of lamivudine for preventing hepatocellular
gastric cancer by eradication of Helicobacter pylori in Res. 2010;54(7):875–896. carcinoma in chronic hepatitis B: a multicenter
Linqu County, China: baseline results and factors 374. Chen HL, Chang MH, Ni YH, et al. Seroepidemiol- retrospective study of 2795 patients. Hepatol Res.
affecting the eradication. Gut. 2016;65(1):9–18. ogy of hepatitis B virus infection in children: ten 2005;32(3):173–184.
354. Yang P, Zhou Y, Chen B, et al. Aspirin use and the years of mass vaccination in Taiwan. JAMA. 1996; 392. Ikeda K, Arase Y, Kobayashi M, et al. A long-term
risk of gastric cancer: a meta-analysis. Dig Dis Sci. 276(11):906–908. glycyrrhizin injection therapy reduces hepatocellular
2010;55(6):1533–1539. 375. Lee CL, Hsieh KS, Ko YC. Trends in the incidence of carcinogenesis rate in patients with interferon-
355. Rothwell PM, Fowkes FG, Belch JF, Ogawa H, hepatocellular carcinoma in boys and girls in Taiwan resistant active chronic hepatitis C: a cohort study
Warlow CP, Meade TW. Effect of daily aspirin on after large-scale hepatitis B vaccination. Cancer of 1249 patients. Dig Dis Sci. 2006;51(3):603–609.
long-term risk of death due to cancer: analysis of Epidemiol Biomarkers Prev. 2003;12(1):57–59. 393. Controlled trial of tamoxifen as adjuvant agent in
individual patient data from randomised trials. 376. Chang MH, Shau WY, Chen CJ, et al. Hepatitis B management of early breast cancer. Interim analysis
Lancet. 2011;377(9759):31–41. vaccination and hepatocellular carcinoma rates in at four years by Nolvadex Adjuvant Trial Organisa-
355a. Huang XZ, Chen Y, Wu J, Zhang X, Wu CC, boys and girls. JAMA. 2000;284(23):3040–3042. tion. Lancet. 1983;1(8319):257–261.
Zhang CY, Sun SS, Chen WJ. Aspirin and non- 377. Hung GY, Horng JL, Yen HJ, Lee CY, Lin LY. 394. Early Breast Cancer Trialists’ Collaborative Group.
steroidal anti-inflammatory drugs use reduce Changing incidence patterns of hepatocellular Effects of adjuvant tamoxifen and of cytotoxic
gastric cancer risk: A dose-response meta-analysis. carcinoma among age groups in Taiwan. J Hepatol. therapy on mortality in early breast cancer. An
Oncotarget. 2017;8(3):4781–4795. 2015;63(6):1390–1396. overview of 61 randomized trials among 28,896
356. Wong BC, Zhang L, Ma JL, et al. Effects of 378. Qu C, Chen T, Fan C, et al. Efficacy of neonatal women. N Engl J Med. 1988;319(26):1681–1692.
selective COX-2 inhibitor and Helicobacter pylori HBV vaccination on liver cancer and other liver 395. Cuzick J, Baum M. Tamoxifen and contralateral
eradication on precancerous gastric lesions. Gut. diseases over 30-year follow-up of the Qidong breast cancer. Lancet. 1985;2(8449):282.
2012;61(6):812–818. hepatitis B intervention study: a cluster randomized 396. Fisher B, Costantino JP, Wickerham DL, et al.
357. Zhang Y, Zeng HM, Nie XR, et al. Alterations controlled trial. PLoS Med. 2014;11(12):e1001774. Tamoxifen for prevention of breast cancer: report
of cyclooxygenase-2 methylation levels before and 379. Chen T, Qu C, Yao H, et al. [Long-term efficacy of the National Surgical Adjuvant Breast and Bowel
after intervention trial to prevent gastric cancer of neonatal hepatitis B vaccination against chronic Project P-1 Study. J Natl Cancer Inst. 1998;90(18):
in a Chinese population. Cancer Prev Res (Phila). hepatitis B virus infection and chronic liver disease: 1371–1388.
2016;9(6):484–490. a cross-sectional study based on Qidong Hepatitis 397. Fisher B. Tamoxifen for the prevention of breast
358. Kong P, Wu R, Liu X, et al. The effects of anti- B Intervention Study]. Zhonghua Liu Xing Bing Xue cancer: current status of the National Surgical
inflammatory drug treatment in gastric cancer pre- Za Zhi. 2016;37(1):64–67. Adjuvant Breast and Bowel Project P-1 study. J Natl
vention: an update of a meta-analysis. J Cancer. 2016; 380. Wang Y, Chen T, Lu LL, et al. Adolescent booster with Cancer Inst. 2005;97:1652–1662.
7(15):2247–2257. hepatitis B virus vaccines decreases HBV infection 398. Powles TJ, Ashley S, Tidy A, Smith IE, Dowsett
359. Liu CJ, Jeng YM, Chen CL, et al. Hepatitis B virus in high-risk adults. Vaccine. 2017;35(7):1064–1070. M. Twenty-year follow-up of the Royal Marsden
basal core promoter mutation and DNA load cor- 381. Roohvand F, Kossari N. Advances in hepatitis C randomized, double-blinded tamoxifen breast cancer
relate with expression of hepatitis B core antigen virus vaccines, part two: advances in hepatitis C prevention trial. J Natl Cancer Inst. 2007;99(4):
in patients with chronic hepatitis B. J Infect Dis. virus vaccine formulations and modalities. Expert 283–290.
2009;199(5):742–749. Opin Ther Pat. 2012;22(4):391–415. 399. Veronesi U, Maisonneuve P, Rotmensz N, et al.
360. Marcellin P. Hepatitis B and hepatitis C in. Liver 382. Baumert TF, Schuster C, Cosset FL, et al. Address- Tamoxifen for the prevention of breast cancer:
Int. 2009;29(suppl 1):1–8. ing the next challenges: a summary of the 22nd late results of the Italian Randomized Tamoxifen
374.e8 Part I: Science and Clinical Oncology

Prevention Trial among women with hysterectomy. 417. Cuzick J, DeCensi A, Arun B, et al. Preventive findings in patients from Saudi Arabia. J Steroid
J Natl Cancer Inst. 2007;99(9):727–737. therapy for breast cancer: a consensus statement. Biochem Mol Biol. 2016;164:106–109.
400. Cuzick J, Forbes J, Edwards R, et al. First results Lancet Oncol. 2011;12(5):496–503. 435. Jorde R, Schirmer H, Wilsgaard T, et al. Polymor-
from the International Breast Cancer Intervention 418. Reimers LL, Sivasubramanian PS, Hershman D, et al. phisms related to the serum 25-hydroxyvitamin D
Study (IBIS-I): a randomised prevention trial. Lancet. Breast cancer chemoprevention among high-risk level and risk of myocardial infarction, diabetes,
2002;360(9336):817–824. women and those with ductal carcinoma in situ. cancer and mortality. The Tromso Study. PLoS ONE.
401. Cuzick J, Forbes JF, Sestak I, et al. Long-term results Breast J. 2015;21(4):377–386. 2012;7(5):e37295.
of tamoxifen prophylaxis for breast cancer–96-month 419. Litton JK, Arun BK, Brown PH, Hortobagyi GN. 436. Yao S, Zirpoli G, Bovbjerg DH, et al. Variants in
follow-up of the randomized IBIS-I trial. J Natl Aromatase inhibitors and breast cancer prevention. the vitamin D pathway, serum levels of vitamin D,
Cancer Inst. 2007;99(4):272–282. Expert Opin Pharmacother. 2012;13(3):325–331. and estrogen receptor negative breast cancer among
402. Fisher B, Costantino JP, Wickerham DL, et al. 420. Baum M, Budzar AU, Cuzick J, et al. Anastrozole African-American women: a case-control study. Breast
Tamoxifen for the prevention of breast cancer: alone or in combination with tamoxifen versus Cancer Res. 2012;14(2):R58.
current status of the National Surgical Adjuvant tamoxifen alone for adjuvant treatment of post- 437. Clement Y. Can green tea do that? A literature
Breast and Bowel Project P-1 study. J Natl Cancer menopausal women with early breast cancer: first review of the clinical evidence. Prev Med. 2009;
Inst. 2005;97(22):1652–1662. results of the ATAC randomised trial. Lancet. 2002; 49(2–3):83–87.
403. Powles T, Eeles R, Ashley S, et al. Interim analysis of 359(9324):2131–2139. 438. Nakachi K, Suemasu K, Suga K, Takeo T, Imai K,
the incidence of breast cancer in the Royal Marsden 421. Cuzick J, Sestak I, Baum M, et al. Effect of Higashi Y. Influence of drinking green tea on breast
Hospital tamoxifen randomised chemoprevention anastrozole and tamoxifen as adjuvant treatment cancer malignancy among Japanese patients. Jpn J
trial. Lancet. 1998;352(9122):98–101. for early-stage breast cancer: 10-year analysis of the Cancer Res. 1998;89(3):254–261.
404. Veronesi U, Maisonneuve P, Costa A, et al. Preven- ATAC trial. Lancet Oncol. 2010;11(12):1135–1141. 439. Park OJ, Surh YJ. Chemopreventive potential of
tion of breast cancer with tamoxifen: preliminary 422. Margolese RG, Cecchini RS, Julian TB, et al. epigallocatechin gallate and genistein: evidence from
findings from the Italian randomised trial among Anastrozole versus tamoxifen in postmenopausal epidemiological and laboratory studies. Toxicol Lett.
hysterectomised women. Italian Tamoxifen Preven- women with ductal carcinoma in situ undergoing 2004;150(1):43–56.
tion Study. Lancet. 1998;352(9122):93–97. lumpectomy plus radiotherapy (NSABP B-35): a 440. Sartippour MR, Heber D, Ma J, Lu Q, Go VL,
405. Vogel VG, Costantino JP, Wickerham DL, et al. randomised, double-blind, phase 3 clinical trial. Nguyen M. Green tea and its catechins inhibit breast
Effects of tamoxifen vs raloxifene on the risk of Lancet. 2016;387(10021):849–856. cancer xenografts. Nutr Cancer. 2001;40(2):149–156.
developing invasive breast cancer and other disease 423. Forbes JF, Sestak I, Howell A, et al. Anastrozole 441. Weisburger JH, Chung FL. Mechanisms of chronic
outcomes: the NSABP Study of Tamoxifen and versus tamoxifen for the prevention of locoregional disease causation by nutritional factors and tobacco
Raloxifene (STAR) P-2 trial. JAMA. 2006;295(23): and contralateral breast cancer in postmenopausal products and their prevention by tea polyphenols.
2727–2741. women with locally excised ductal carcinoma in Food Chem Toxicol. 2002;40(8):1145–1154.
406. Vogel VG, Costantino JP, Wickerham DL, et al. situ (IBIS-II DCIS): a double-blind, randomised 442. Baliga MS, Meleth S, Katiyar SK. Growth inhibitory
Update of the National Surgical Adjuvant Breast and controlled trial. Lancet. 2016;387(10021):866–873. and antimetastatic effect of green tea polyphenols
Bowel Project Study of Tamoxifen and Raloxifene 424. Cuzick J, Sestak I, Forbes JF, et al. Anastrozole on metastasis-specific mouse mammary carcinoma
(STAR) P-2 Trial: preventing breast cancer. Cancer for prevention of breast cancer in high-risk post- 4T1 cells in vitro and in vivo systems. Clin Cancer
Prev Res (Phila). 2010;3(6):696–706. menopausal women (IBIS-II): an international, Res. 2005;11(5):1918–1927.
407. Cummings SR, Eckert S, Krueger KA, et al. The double-blind, randomised placebo-controlled trial. 443. Thangapazham RL, Singh AK, Sharma A, Warren J,
effect of raloxifene on risk of breast cancer in Lancet. 2014;383(9922):1041–1048. Gaddipati JP, Maheshwari RK. Green tea polyphenols
postmenopausal women: results from the MORE 425. Richardson H, Johnston D, Pater J, Goss P. The and its constituent epigallocatechin gallate inhibits
randomized trial. Multiple Outcomes of Raloxifene National Cancer Institute of Canada Clinical Trials proliferation of human breast cancer cells in vitro
Evaluation. JAMA. 1999;281(23):2189–2197. Group MAP.3 trial: an international breast cancer and in vivo. Cancer Lett. 2007;245(1–2):232–241.
408. Ettinger B, Black DM, Mitlak BH, et al. Reduction prevention trial. Curr Oncol. 2007;14(3):89–96. 444. Mineva ND, Paulson KE, Naber SP, Yee AS,
of vertebral fracture risk in postmenopausal women 426. Cuzick J. Aromatase inhibitors in prevention–data Sonenshein GE. Epigallocatechin-3-gallate inhibits
with osteoporosis treated with raloxifene: results from from the ATAC (Arimidex, Tamoxifen Alone or in stem-like inflammatory breast cancer cells. PLoS
a 3-year randomized clinical trial. Multiple Outcomes Combination) trial and the design of IBIS-II (the ONE. 2013;8(9):e73464.
of Raloxifene Evaluation (MORE) Investigators. second International Breast Cancer Intervention 445. Crew KD, Brown P, Greenlee H, et al. Phase IB
JAMA. 1999;282(7):637–645. Study). Recent Results Cancer Res. 2003;163:96–103; randomized, double-blinded, placebo-controlled,
409. Martino S, Cauley JA, Barrett-Connor E, et al. discussion 264-106. dose escalation study of polyphenon E in women
Continuing outcomes relevant to Evista: breast 427. Cuzick J. IBIS II: a breast cancer prevention trial with hormone receptor-negative breast cancer. Cancer
cancer incidence in postmenopausal osteoporotic in postmenopausal women using the aromatase Prev Res (Phila). 2012;5(9):1144–1154.
women in a randomized trial of raloxifene. J Natl inhibitor anastrozole. Expert Rev Anticancer Ther. 446. Brown P, Arun B, Miller A, et al. Phase II trial of
Cancer Inst. 2004;96(23):1751–1761. 2008;8(9):1377–1385. bexarotene in women at high risk of breast cancer:
410. Barrett-Connor E, Mosca L, Collins P, et al. Effects of 428. Goss PE, Ingle JN, Ales-Martinez JE, et al. Exemes- comparison of protein and RNA biomarkers. Cancer
raloxifene on cardiovascular events and breast cancer in tane for breast-cancer prevention in postmenopausal Prev Res (Phila Pa). 2008;1(7 suppl):CN4.
postmenopausal women. N Engl J Med. 2006;355(2): women. N Engl J Med. 2011;364(25):2381–2391. 447. Crew KD, Xiao T, Thomas PS, et al. Safety,
125–137. 429. Brown PH, Subbaramaiah K, Salmon AP, et al. Com- feasibility, and biomarker effects of high-dose
411. Cummings SR, Ensrud K, Delmas PD, et al. bination chemoprevention of HER2/neu-induced vitamin D supplementation among women at
Lasofoxifene in postmenopausal women with breast cancer using a cyclooxygenase-2 inhibitor and high risk for breast cancer. Int J Food Sci Nutr Diet.
osteoporosis. N Engl J Med. 2010;362(8):686– a retinoid X receptor-selective retinoid. Cancer Prev 2015;2015(suppl 1):1–16.
696. Res (Phila). 2008;1(3):208–214. 448. Bonanni B, Puntoni M, Cazzaniga M, et al. Dual
412. LaCroix AZ, Powles T, Osborne CK, et al. Breast 430. Li Y, Zhang Y, Hill J, et al. The rexinoid, bexarotene, effect of metformin on breast cancer proliferation in
cancer incidence in the randomized PEARL trial of prevents the development of premalignant lesions a randomized presurgical trial. J Clin Oncol. 2012;
lasofoxifene in postmenopausal osteoporotic women. in MMTV-erbB2 mice. Br J Cancer. 2008;98(8): 30(21):2593–2600.
J Natl Cancer Inst. 2010;102(22):1706–1715. 1380–1388. 449. Samavat H, Dostal AM, Wang R, et al. The Min-
413. Cummings SR, McClung M, Reginster JY, et al. 431. Kim HT, Kong G, Denardo D, et al. Identification nesota Green Tea Trial (MGTT), a randomized
Arzoxifene for prevention of fractures and invasive of biomarkers modulated by the rexinoid LGD1069 controlled trial of the efficacy of green tea extract
breast cancer in postmenopausal women. J Bone (bexarotene) in human breast cells using oligo- on biomarkers of breast cancer risk: study rationale,
Miner Res. 2011;26(2):397–404. nucleotide arrays. Cancer Res. 2006;66(24):12009– design, methods, and participant characteristics.
414. Powles TJ, Diem SJ, Fabian CJ, et al. Breast 12018. Cancer Causes Control. 2015;26(10):1405–1419.
cancer incidence in postmenopausal women with 432. Kapetanovic IM, Horn TL, Johnson WD, Cwik MJ, 450. Dostal AM, Samavat H, Bedell S, et al. The safety
osteoporosis or low bone mass using arzoxifene. Detrisac CJ, McCormick DL. Murine oncogenicity of green tea extract supplementation in postmeno-
Breast Cancer Res Treat. 2012;134(1):299–306. and pharmacokinetics studies of 9-cis-UAB30, an pausal women at risk for breast cancer: results of
415. Johnell O, Cauley JA, Kulkarni PM, Wong M, Stock RXR agonist, for breast cancer chemoprevention. the Minnesota Green Tea Trial. Food Chem Toxicol.
JL. Raloxifene reduces risk of vertebral fractures Int J Toxicol. 2010;29(2):157–164. 2015;83:26–35.
[corrected] in postmenopausal women regardless 433. Thanasitthichai S, Chaiwerawattana A, Prasit- 451. Kuerer HM, Buzdar AU, Mittendorf EA, et al.
of prior hormone therapy. J Fam Pract. 2004; thipayong A. Association of vitamin D level with Biologic and immunologic effects of preoperative
53(10):789–796. clinicopathological features in breast cancer. Asian trastuzumab for ductal carcinoma in situ of the
416. Cuzick J, Powles T, Veronesi U, et al. Overview Pac J Cancer Prev. 2015;16(12):4881–4883. breast. Cancer. 2011;117(1):39–47.
of the main outcomes in breast-cancer prevention 434. Abulkhair O, Saadeddin A, Makram O, et al. 452. Estévez LG, Suarez-Gauthier A, Garcia E, et al.
trials. Lancet. 2003;361(9354):296–300. Vitamin D levels and breast cancer characteristics: Molecular effects of lapatinib in patients with HER2
Lifestyle and Cancer Prevention  •  CHAPTER 22 374.e9
374.e9

positive ductal carcinoma in situ. Breast Cancer Res. 469. Pirog EC, Lloveras B, Molijn A, et al. HPV preva- an unexpected observation. J Infect Dis. 2013;208(9):
2014;16(4):R76. lence and genotypes in different histological subtypes 1391–1396.
453. Knutson KL, Disis ML. Expansion of HER2/ of cervical adenocarcinoma, a worldwide analysis of 484. Hildesheim A, Wacholder S, Catteau G, et al.
neu-specific T cells ex vivo following immunization 760 cases. Mod Pathol. 2014;27(12):1559–1567. Efficacy of the HPV-16/18 vaccine: final according
with a HER2/neu peptide-based vaccine. Clin Breast 470. Wheeler CM, Hunt WC, Joste NE, Key CR, Quint to protocol results from the blinded phase of the
Cancer. 2001;2(1):73–79. WG, Castle PE. Human papillomavirus genotype randomized Costa Rica HPV-16/18 vaccine trial.
454. Schiffman K, Rinn K, Disis ML. Delayed type distributions: implications for vaccination and cancer Vaccine. 2014;32(39):5087–5097.
hypersensitivity response to recall antigens does not screening in the United States. J Natl Cancer Inst. 485. Herrero R, Wacholder S, Rodriguez AC, et al.
accurately reflect immune competence in advanced 2009;101(7):475–487. Prevention of persistent human papillomavirus
stage breast cancer patients. Breast Cancer Res Treat. 471. U.S. Food and Drug Administration. Cervarix infection by an HPV16/18 vaccine: a community-
2002;74(1):17–23. [Human Papillomavirus Bivalent (Types 16 and 18) based randomized clinical trial in Guanacaste, Costa
455. Montgomery RB, Makary E, Schiffman K, Goodell Vaccine, Recombinant]; 2009. https://www.fda.gov/ Rica. Cancer Discov. 2011;1(5):408–419.
V, Disis ML. Endogenous anti-HER2 antibodies BiologicsBloodVaccines/Vaccines/ApprovedProducts/ 486. Rodriguez AC, Solomon D, Herrero R, et al. Impact
block HER2 phosphorylation and signaling through ucm186957.htm. of human papillomavirus vaccination on cervical
extracellular signal-regulated kinase. Cancer Res. 472. Harper DM, Franco EL, Wheeler C, et al. Efficacy of cytology screening, colposcopy, and treatment.
2005;65(2):650–656. a bivalent L1 virus-like particle vaccine in prevention Am J Epidemiol. 2013;178(5):752–760.
456. Phan-Lai V, Dang Y, Gad E, Childs J, Disis ML. of infection with human papillomavirus types 16 487. Lang Kuhs KA, Porras C, Schiller JT, et al. Effect
The antitumor efficacy of IL2/IL21-cultured and 18 in young women: a randomised controlled of different human papillomavirus serological and
polyfunctional neu-specific T cells is TNFalpha/IL17 trial. Lancet. 2004;364(9447):1757–1765. DNA criteria on vaccine efficacy estimates. Am J
dependent. Clin Cancer Res. 2016;22(9):2207–2216. 473. Harper DM, Franco EL, Wheeler CM, et al. Epidemiol. 2014;180(6):599–607.
457. Dols A, Meijer SL, Hu HM, et al. Identification Sustained efficacy up to 4.5 years of a bivalent L1 488. Kreimer AR, Rodriguez AC, Hildesheim A, et al.
of tumor-specific antibodies in patients with breast virus-like particle vaccine against human papilloma- Proof-of-principle evaluation of the efficacy of fewer
cancer vaccinated with gene-modified allogeneic virus types 16 and 18: follow-up from a randomised than three doses of a bivalent HPV16/18 vaccine.
tumor cells. J Immunother. 2003;26(2):163–170. control trial. Lancet. 2006;367(9518):1247–1255. J Natl Cancer Inst. 2011;103(19):1444–1451.
458. Emens LA, Asquith JM, Leatherman JM, et al. Timed 474. GlaxoSmithKline Vaccine HPVSG, Romanowski 489. Lang Kuhs KA, Gonzalez P, Rodriguez AC, et al.
sequential treatment with cyclophosphamide, doxo- B, de Borba PC, et al. Sustained efficacy and Reduced prevalence of vulvar HPV16/18 infection
rubicin, and an allogeneic granulocyte-macrophage immunogenicity of the human papillomavirus among women who received the HPV16/18 bivalent
colony-stimulating factor-secreting breast tumor (HPV)-16/18 AS04-adjuvanted vaccine: analysis of a vaccine: a nested analysis within the Costa Rica
vaccine: a chemotherapy dose-ranging factorial randomised placebo-controlled trial up to 6.4 years. Vaccine Trial. J Infect Dis. 2014;210(12):1890–1899.
study of safety and immune activation. J Clin Oncol. Lancet. 2009;374(9706):1975–1985. 490. Kreimer AR, Gonzalez P, Katki HA, et al. Efficacy
2009;27(35):5911–5918. 475. De Carvalho N, Teixeira J, Roteli-Martins CM, of a bivalent HPV 16/18 vaccine against anal HPV
459. Pathangey LB, McCurry DB, Gendler SJ, et al. Surro- et al. Sustained efficacy and immunogenicity of 16/18 infection among young women: a nested
gate in vitro activation of innate immunity synergizes the HPV-16/18 AS04-adjuvanted vaccine up to 7.3 analysis within the Costa Rica Vaccine Trial. Lancet
with interleukin-7 to unleash rapid antigen-driven years in young adult women. Vaccine. 2010;28(38): Oncol. 2011;12(9):862–870.
outgrowth of CD4+ and CD8+ human peripheral 6247–6255. 491. Herrero R, Quint W, Hildesheim A, et al. Reduced
blood T-cells naturally recognizing MUC1, HER2/ 476. Roteli-Martins CM, Naud P, De Borba P, et al. prevalence of oral human papillomavirus (HPV) 4
neu and other tumor-associated antigens. Oncotarget. Sustained immunogenicity and efficacy of the HPV- years after bivalent HPV vaccination in a randomized
2017;8(7):10785–10808. 16/18 AS04-adjuvanted vaccine: up to 8.4 years clinical trial in Costa Rica. PLoS ONE. 2013;8(7):
460. Disis ML, Wallace DR, Gooley TA, et al. Concurrent of follow-up. Hum Vaccin Immunother. 2012;8(3): e68329.
trastuzumab and HER2/neu-specific vaccination in 390–397. 492. Konno R, Tamura S, Dobbelaere K, Yoshikawa
patients with metastatic breast cancer. J Clin Oncol. 477. Naud PS, Roteli-Martins CM, De Carvalho NS, H. Efficacy of human papillomavirus type 16/18
2009;27(28):4685–4692. et al. Sustained efficacy, immunogenicity, and safety AS04-adjuvanted vaccine in Japanese women aged 20
461. Mittendorf EA, Clifton GT, Holmes JP, et al. Final of the HPV-16/18 AS04-adjuvanted vaccine: final to 25 years: final analysis of a phase 2 double-blind,
report of the phase I/II clinical trial of the E75 analysis of a long-term follow-up study up to 9.4 randomized controlled trial. Int J Gynecol Cancer.
(nelipepimut-S) vaccine with booster inoculations to years post-vaccination. Hum Vaccin Immunother. 2010;20(5):847–855.
prevent disease recurrence in high-risk breast cancer 2014;10(8):2147–2162. 493. Konno R, Tamura S, Dobbelaere K, Yoshikawa H.
patients. Ann Oncol. 2014;25(9):1735–1742. 478. Paavonen J, Jenkins D, Bosch FX, et al. Efficacy of a Efficacy of human papillomavirus 16/18 AS04-
462. Mittendorf EA, Ardavanis A, Symanowski J, et al. prophylactic adjuvanted bivalent L1 virus-like-particle adjuvanted vaccine in Japanese women aged 20 to
Primary analysis of a prospective, randomized, vaccine against infection with human papillomavirus 25 years: interim analysis of a phase 2 double-blind,
single-blinded phase II trial evaluating the HER2 types 16 and 18 in young women: an interim analysis randomized, controlled trial. Int J Gynecol Cancer.
peptide AE37 vaccine in breast cancer patients to of a phase III double-blind, randomised controlled 2010;20(3):404–410.
prevent recurrence. Ann Oncol. 2016;27(7):1241– trial. Lancet. 2007;369(9580):2161–2170. 494. Konno R, Yoshikawa H, Okutani M, et al.
1248. 479. Paavonen J, Naud P, Salmeron J, et al. Efficacy Efficacy of the human papillomavirus (HPV)-
463. Mittendorf EA, Ardavanis A, Litton JK, et al. Primary of human papillomavirus (HPV)-16/18 AS04- 16/18 AS04-adjuvanted vaccine against cervical
analysis of a prospective, randomized, single-blinded adjuvanted vaccine against cervical infection and pre- intraepithelial neoplasia and cervical infection in
phase II trial evaluating the HER2 peptide GP2 cancer caused by oncogenic HPV types (PATRICIA): young Japanese women. Hum Vaccin Immunother.
vaccine in breast cancer patients to prevent recur- final analysis of a double-blind, randomised study in 2014;10(7):1781–1794.
rence. Oncotarget. 2016;7(40):66192–66201. young women. Lancet. 2009;374(9686):301–314. 495. Zhu FC, Chen W, Hu YM, et al. Efficacy, immu-
464. Clifton GT, Litton JK, Arrington K, et al. Results 480. Lehtinen M, Paavonen J, Wheeler CM, et al. nogenicity and safety of the HPV-16/18 AS04-
of a phase Ib trial of combination immunotherapy Overall efficacy of HPV-16/18 AS04-adjuvanted adjuvanted vaccine in healthy Chinese women aged
with a CD8+ T cell eliciting vaccine and trastuzumab vaccine against grade 3 or greater cervical intraepi- 18-25 years: results from a randomized controlled
in breast cancer patients. Ann Surg Oncol. 2017; thelial neoplasia: 4-year end-of-study analysis of the trial. Int J Cancer. 2014;135(11):2612–2622.
24(8):2161–2167. randomised, double-blind PATRICIA trial. Lancet 496. Skinner SR, Szarewski A, Romanowski B, et al.
465. Margolese RG, Vallow LA, Albain KS, Whitworth Oncol. 2012;13(1):89–99. Efficacy, safety, and immunogenicity of the human
PW, Cianfrocca M. National Surgical Adjuvant 481. Wheeler CM, Castellsague X, Garland SM, papillomavirus 16/18 AS04-adjuvanted vaccine
Breast and Bowel Project (NSABP): Anastrozole et al. Cross-protective efficacy of HPV-16/18 in women older than 25 years: 4-year interim
or tamoxifen in treating postmenopausal women AS04-adjuvanted vaccine against cervical infection follow-up of the phase 3, double-blind, randomised
with ductal carcinoma in situ who are undergoing and precancer caused by non-vaccine oncogenic controlled VIVIANE study. Lancet. 2014;384(9961):
lumpectomy and radiation therapy; 2003. HPV types: 4-year end-of-study analysis of the 2213–2227.
466. Rigoni-Stern D. Fatti statistici relative alle mallatie randomised, double-blind PATRICIA trial. Lancet 497. Skinner SR, Apter D, De Carvalho N, et al. Human
cancrosi che servirono de base alla poche cose dette Oncol. 2012;13(1):100–110. papillomavirus (HPV)-16/18 AS04-adjuvanted
dal dott. [Surgical facts, related to cancer diseases]. 482. Szarewski A, Poppe WA, Skinner SR, et al. Efficacy vaccine for the prevention of cervical cancer and
G Servire Progr Path Tera. 1842;2:507–517. of the human papillomavirus (HPV)-16/18 AS04- HPV-related diseases. Expert Rev Vaccines. 2016;15(3):
467. Lopalco PL. Spotlight on the 9-valent HPV vaccine. adjuvanted vaccine in women aged 15-25 years with 367–387.
Drug Des Devel Ther. 2017;11:35–44. and without serological evidence of previous exposure 498. Kreimer AR, Struyf F, Del Rosario-Raymundo MR,
468. International Agency for Research on Cancer to HPV-16/18. Int J Cancer. 2012;131(1):106–116. et al. Efficacy of fewer than three doses of an HPV-
WHO. IARC Monographs on the Evaluation of 483. Szarewski A, Skinner SR, Garland SM, et al. Efficacy 16/18 AS04-adjuvanted vaccine: combined analysis
the Carcinogenic Risks to Human; 2016. http:// of the HPV-16/18 AS04-adjuvanted vaccine against of data from the Costa Rica Vaccine and PATRICIA
monographs.iarc.fr. low-risk HPV types (PATRICIA randomized trial): Trials. Lancet Oncol. 2015;16(7):775–786.
374.e10 Part I: Science and Clinical Oncology

499. U.S. Food and Drug Administration. Gardasil 516. FUTURE II Study Group. Quadrivalent vaccine vaccination—updated recommendations of the Advi-
[Human Papillomavirus Quadrivalent (Types against human papillomavirus to prevent high-grade sory Committee on Immunization Practices. MMWR
6, 11, 16, 18) Vaccine, Recombinant]; 2006. cervical lesions. N Engl J Med. 2007;356(19): Morb Mortal Wkly Rep. 2016;65(49):1405–1408.
https://www.fda.gov/BiologicsBloodVaccines/ 1915–1927. 535. Mondul AM, Weinstein SJ, Layne TM, Albanes D,
Vaccines/ApprovedProducts/ucm094042.htm. 517. U.S. Food and Drug Administration. Gardasil 9 Vitamin D. and cancer risk and mortality: state of
500. Handler NS, Handler MZ, Majewski S, Schwartz [Human Papillomavirus 9-valent Vaccine, Recombi- the science, gaps, and challenges. Epidemiol Rev.
RA. Human papillomavirus vaccine trials and nant]; 2014. https://www.fda.gov/BiologicsBloodVac 2017;39(1):28–48.
tribulations: vaccine efficacy. J Am Acad Dermatol. cines/Vaccines/ApprovedProducts/ucm426445.htm. 536. Thompson IM, Goodman PJ, Tangen CM, et al.
2015;73(5):759–767; quiz 767-758. 518. Castellsague X, Giuliano AR, Goldstone S, et al. The influence of finasteride on the development of
501. U.S. Food and Drug Administration. VRBPAC Immunogenicity and safety of the 9-valent HPV prostate cancer. N Engl J Med. 2003;349(3):215–224.
Background Document: Gardasil HPV Quadrivalent vaccine in men. Vaccine. 2015;33(48):6892–6901. 537. Thompson IM Jr, Goodman PJ, Tangen CM, et al.
Vaccine; 2006. https://www.fda.gov/ohrms/dockets/ 519. Joura EA, Giuliano AR, Iversen OE, et al. A 9-valent Long-term survival of participants in the prostate
ac/06/briefing/2006-4222B3.pdf. HPV vaccine against infection and intraepithelial cancer prevention trial. N Engl J Med. 2013;369(7):
502. Villa LL, Costa RL, Petta CA, et al. Prophylactic neoplasia in women. N Engl J Med. 2015;372(8): 603–610.
quadrivalent human papillomavirus (types 6, 711–723. 538. Andriole GL, Bostwick DG, Brawley OW, et al.
11, 16, and 18) L1 virus-like particle vaccine in 520. Kosalaraksa P, Mehlsen J, Vesikari T, et al. An Effect of dutasteride on the risk of prostate cancer.
young women: a randomised double-blind placebo- open-label, randomized study of a 9-valent human N Engl J Med. 2010;362(13):1192–1202.
controlled multicentre phase II efficacy trial. Lancet papillomavirus vaccine given concomitantly with 539. Grubb RL, Andriole GL, Somerville MC, Mahoney
Oncol. 2005;6(5):271–278. diphtheria, tetanus, pertussis and poliomyelitis C, Manyak MJ, Castro R. The REDUCE follow-up
503. Villa LL, Costa RL, Petta CA, et al. High sustained vaccines to healthy adolescents 11-15 years of age. study: low rate of new prostate cancer diagnoses
efficacy of a prophylactic quadrivalent human papil- Pediatr Infect Dis J. 2015;34(6):627–634. observed during a 2-year, observational, followup
lomavirus types 6/11/16/18 L1 virus-like particle 521. Schilling S, Samuels-Kalow M, Gerber JS, Scribano study of men who participated in the REDUCE
vaccine through 5 years of follow-up. Br J Cancer. PV, French B, Wood JN. Testing and treatment trial. J Urol. 2013;189(3):871–877.
2006;95(11):1459–1466. after adolescent sexual assault in pediatric emergency 540. Toren P, Margel D, Kulkarni G, Finelli A, Zlotta A,
504. Olsson SE, Villa LL, Costa RL, et al. Induction departments. Pediatrics. 2015;136(6):e1495–e1503. Fleshner N. Effect of dutasteride on clinical progres-
of immune memory following administration of 522. Van Damme P, Olsson SE, Block S, et al. Immu- sion of benign prostatic hyperplasia in asymptomatic
a prophylactic quadrivalent human papillomavirus nogenicity and safety of a 9-valent HPV vaccine. men with enlarged prostate: a post hoc analysis of
(HPV) types 6/11/16/18 L1 virus-like particle (VLP) Pediatrics. 2015;136(1):e28–e39. the REDUCE study. BMJ. 2013;346:f2109.
vaccine. Vaccine. 2007;25(26):4931–4939. 523. Vesikari T, Brodszki N, van Damme P, et al. A 541. Theoret MR, Ning YM, Zhang JJ, Justice R,
505. Brotherton JM, Fridman M, May CL, Chappell G, randomized, double-blind, phase III study of the Keegan P, Pazdur R. The risks and benefits of
Saville AM, Gertig DM. Early effect of the HPV immunogenicity and safety of a 9-valent human 5alpha-reductase inhibitors for prostate-cancer
vaccination programme on cervical abnormalities papillomavirus L1 virus-like particle vaccine (V503) prevention. N Engl J Med. 2011;365(2):97–99.
in Victoria, Australia: an ecological study. Lancet. versus Gardasil(R) in 9-15-year-old girls. Pediatr 542. Kristal AR, Till C, Song X, et al. Plasma vitamin D
2011;377(9783):2085–2092. Infect Dis J. 2015;34(9):992–998. and prostate cancer risk: results from the Selenium
506. Crowe E, Pandeya N, Brotherton JM, et al. 524. European Medicines Agency. Gardasil 9. Summary and Vitamin E Cancer Prevention Trial. Cancer
Effectiveness of quadrivalent human papillomavirus of Product Characteristics; 2016. http://www.ema Epidemiol Biomarkers Prev. 2014;23(8):1494–1504.
vaccine for the prevention of cervical abnormali- .europa.eu/docs/en_GB/document_library/EPAR 543. Kristal AR, Darke AK, Morris JS, et al. Baseline
ties: case-control study nested within a population _-_Product_Information/human/003852/ selenium status and effects of selenium and vitamin
based screening programme in Australia. BMJ. WC500189111.pdf. e supplementation on prostate cancer risk. J Natl
2014;348:g1458. 525. Van Damme P, Meijer CJ, Kieninger D, et al. A phase Cancer Inst. 2014;106(3):djt456.
507. Kahn JA, Brown DR, Ding L, et al. Vaccine-type III clinical study to compare the immunogenicity 544. Zhou CK, Daugherty SE, Liao LM, et al. Do aspirin
human papillomavirus and evidence of herd protec- and safety of the 9-valent and quadrivalent HPV and other NSAIDs confer a survival benefit in men
tion after vaccine introduction. Pediatrics. 2012; vaccines in men. Vaccine. 2016;34(35):4205–4212. diagnosed with prostate cancer? a pooled analysis
130(2):e249–e256. 526. Larson HJ, Wilson R, Hanley S, Parys A, Paterson of NIH-AARP and PLCO cohorts. Cancer Prev Res
508. Herweijer E, Leval A, Ploner A, et al. Association P. Tracking the global spread of vaccine sentiments: (Phila). 2017.
of varying number of doses of quadrivalent human the global response to Japan’s suspension of its HPV 545. Platz EA, Tangen CM, Goodman PJ, et al. Statin
papillomavirus vaccine with incidence of condyloma. vaccine recommendation. Hum Vaccin Immunother. drug use is not associated with prostate cancer risk in
JAMA. 2014;311(6):597–603. 2014;10(9):2543–2550. men who are regularly screened. J Urol. 2014;192(2):
509. Giuliano AR, Palefsky JM, Goldstone S, et al. 527. Colafrancesco S, Perricone C, Tomljenovic L, 379–384.
Efficacy of quadrivalent HPV vaccine against Shoenfeld Y. Human papilloma virus vaccine 546. Schenk JM, Till CA, Tangen CM, et al. Serum
HPV Infection and disease in males. N Engl J Med. and primary ovarian failure: another facet of the 25-hydroxyvitamin D concentrations and risk of
2011;364(5):401–411. autoimmune/inflammatory syndrome induced by prostate cancer: results from the Prostate Cancer
510. Palefsky JM, Giuliano AR, Goldstone S, et al. HPV adjuvants. Am J Reprod Immunol. 2013;70(4):309– Prevention Trial. Cancer Epidemiol Biomarkers Prev.
vaccine against anal HPV infection and anal intraepi- 316. 2014;23(8):1484–1493.
thelial neoplasia. N Engl J Med. 2011;365(17): 528. Signorelli C, Odone A, Ciorba V, et al. Human 547. Aghajanpour M, Nazer MR, Obeidavi Z, Akbari M,
1576–1585. papillomavirus 9-valent vaccine for cancer preven- Ezati P, Kor NM. Functional foods and their role in
511. Kash N, Lee MA, Kollipara R, Downing C, Guidry tion: a systematic review of the available evidence. cancer prevention and health promotion: a compre-
J, Tyring SK. Safety and efficacy data on vaccines Epidemiol Infect. 2017;1–21. hensive review. Am J Cancer Res. 2017;7(4):740–769.
and immunization to human papillomavirus. J Clin 529. Zhai L, Tumban E. Gardasil-9: a global survey of 548. Wan L, Chen M, Cao J, et al. The APC/C E3 ligase
Med. 2015;4(4):614–633. projected efficacy. Antiviral Res. 2016;130:101–109. complex activator FZR1 restricts BRAF oncogenic
512. Julius JM, Ramondeta L, Tipton KA, Lal LS, 530. Angioli R, Lopez S, Aloisi A, et al. Ten years of function. Cancer Discov. 2017;7(4):424–441.
Schneider K, Smith JA. Clinical perspectives on the HPV vaccines: state of art and controversies. Crit 549. Miles FL, Goodman PJ, Tangen C, et al. Interactions
role of the human papillomavirus vaccine in the pre- Rev Oncol Hematol. 2016;102:65–72. of the insulin-like growth factor axis and vitamin
vention of cancer. Pharmacotherapy. 2011;31(3):280– 531. Gertig DM, Brotherton JM, Saville M. Measuring D in prostate cancer risk in the Prostate Cancer
297. human papillomavirus (HPV) vaccination coverage Prevention Trial. Nutrients. 2017;9(4).
513. Garland SM, Cheung TH, McNeill S, et al. Safety and the role of the National HPV Vaccination 550. Smith A, Balar AV, Milowsky MI, Chen RC. Bladder
and immunogenicity of a 9-valent HPV vaccine in Program Register, Australia. Sex Health. 2011;8(2): cancer. In: Niederhuber JE, Doroshow JH, Kastan
females 12-26 years of age who previously received 171–178. MB, Tepper JE, eds. Abeloff ’s Clinical Oncology. 5th
the quadrivalent HPV vaccine. Vaccine. 2015;33(48): 532. Osborne SL, Tabrizi SN, Brotherton JM, et al. Assess- ed. Philadelphia, PA: Elsevier; 2014:1445–1462.
6855–6864. ing genital human papillomavirus genoprevalence 551. Steinberg G, Bahnson R, Brosman S, Middleton
514. FUTURE II Study Group. Prophylactic efficacy in young Australian women following the introduc- R, Wajsman Z, Wehle M. Efficacy and safety of
of a quadrivalent human papillomavirus (HPV) tion of a national vaccination program. Vaccine. valrubicin for the treatment of bacillus Calmette-
vaccine in women with virological evidence of HPV 2015;33(1):201–208. Guérin refractory carcinoma in situ of the bladder.
infection. J Infect Dis. 2007;196(10):1438–1446. 533. Reagan-Steiner S, Yankey D, Jeyarajah J, et al. The Valrubicin Study Group. J Urol. 2000;163(3):
515. FUTURE II Study Group, Dillner J, Kjaer SK, National, regional, state, and selected local area 761–767.
et al. Four year efficacy of prophylactic human vaccination coverage among adolescents aged 13-17 552. Pinsky CM, Camacho FJ, Kerr D, et al. Intravesi-
papillomavirus quadrivalent vaccine against low grade years—United States. MMWR Morb Mortal Wkly cal administration of bacillus Calmette-Guérin in
cervical, vulvar, and vaginal intraepithelial neoplasia Rep. 2014;64(29):784–792. patients with recurrent superficial carcinoma of the
and anogenital warts: randomised controlled trial. 534. Meites E, Kempe A, Markowitz LE. Use of urinary bladder: report of a prospective, randomized
BMJ. 2010;341:c3493. a 2-dose schedule for human papillomavirus trial. Cancer Treat Rep. 1985;69(1):47–53.
Lifestyle and Cancer Prevention  •  CHAPTER 22 374.e11
374.e11

553. Herr HW, Laudone VP, Badalament RA, et al. 569. Bazarbashi SN, Azouz HJ, Abu Sabaa AH, Aljubran outcomes of patients with non-muscle-invasive
Bacillus Calmette-Guérin therapy alters the pro- AH, Alzahrani AM, Alotaibi MF. Recurrence and bladder cancer. BJU Int. 2013;112(8):1105–1112.
gression of superficial bladder cancer. J Clin Oncol. progression in nonmuscle invasive transitional cell 587. Zhang T, Guo P, Zhang Y, et al. The antidiabetic drug
1988;6(9):1450–1455. carcinoma of urinary bladder treated with intravesical metformin inhibits the proliferation of bladder cancer
554. Herr HW, Schwalb DM, Zhang ZF, et al. Intravesical bacillus Calmette-Guérin: a single center experi- cells in vitro and in vivo. Int J Mol Sci. 2013;14(12):
bacillus Calmette-Guérin therapy prevents tumor ence and analysis of prognostic factors. Urol Ann. 24603–24618.
progression and death from superficial bladder 2016;8(3):333–337. 588. Donin NM, Chamie K, Lenis AT, et al. A phase
cancer: ten-year follow-up of a prospective random- 570. Steinberg RL, Brooks NA, Thomas LJ, Mott SL, 2 study of TMX-101, intravesical imiquimod, for
ized trial. J Clin Oncol. 1995;13(6):1404–1408. O’Donnell MA. Bacillus Calmette-Guérin strain may the treatment of carcinoma in situ bladder cancer.
555. Lamm DL, Blumenstein BA, Crissman JD, et al. not effect recurrence-free survival when used intra- Urol Oncol. 2017;35(2):39.e31–39.e37.
Maintenance bacillus Calmette-Guérin immuno- vesically with interferon-alpha2b for non-muscle- 589. Park SJ, Myung SK, Lee Y, Lee YJ. Effects of vitamin
therapy for recurrent TA, T1 and carcinoma in invasive bladder cancer. Urol Oncol. 2017;35(5): and antioxidant supplements in prevention of bladder
situ transitional cell carcinoma of the bladder: a 201–207. cancer: a meta-analysis of randomized controlled
randomized Southwest Oncology Group Study. 571. Babjuk M, Bohle A, Burger M, et al. EAU trials. J Korean Med Sci. 2017;32(4):628–635.
J Urol. 2000;163(4):1124–1129. Guidelines on non-muscle-invasive urothelial 590. Diepgen TL, Mahler V. The epidemiology of skin
556. Ehdaie B, Sylvester R, Herr HW. Maintenance carcinoma of the bladder: update 2016. Eur Urol. cancer. Br J Dermatol. 2002;146(suppl 61):1–6.
bacillus Calmette-Guérin treatment of non-muscle- 2017;71(3):447–461. 591. Housman TS, Feldman SR, Williford PM, et al.
invasive bladder cancer: a critical evaluation of the 572. Chang C, Boorjian SA, Chou R, et al. Diagnosis and Skin cancer is among the most costly of all cancers
evidence. Eur Urol. 2013;64(4):579–585. treatment of non-muscle invasive bladder cancer: to treat for the Medicare population. J Am Acad
557. Oddens J, Brausi M, Sylvester R, et al. Final results AUA/SUO Joint Guideline. American Urological Dermatol. 2003;48(3):425–429.
of an EORTC-GU cancers group randomized Association (AUA)/Society of Urologic Oncology 592. Rogers HW, Weinstock MA, Harris AR, et al. Inci-
study of maintenance bacillus Calmette-Guérin in (SUO) Guideline 2016; file:///C:/Users/MISavage/ dence estimate of nonmelanoma skin cancer in the
intermediate- and high-risk Ta, T1 papillary carci- Downloads/Non-Muscle-Invasive-Bladder-Cancer. United States, 2006. Arch Dermatol. 2010;146(3):
noma of the urinary bladder: one-third dose versus pdf. Accessed June 12, 2017. 283–287.
full dose and 1 year versus 3 years of maintenance. 573. Reis LO, Moro JC, Ribeiro LF, Voris BR, Sadi MV. 593. Ratushny V, Gober MD, Hick R, Ridky TW, Seykora
Eur Urol. 2013;63(3):462–472. Are we following the guidelines on non-muscle JT. From keratinocyte to cancer: the pathogenesis
558. Pfister C, Kerkeni W, Rigaud J, et al. Efficacy and invasive bladder cancer? Int Braz J Urol. 2016;42(1): and modeling of cutaneous squamous cell carcinoma.
tolerance of one-third full dose bacillus Calmette- 22–28. J Clin Invest. 2012;122(2):464–472.
Guérin maintenance therapy every 3 months 574. Eifler JB, Scarpato KR, Clark PE. Management 594. Kuflik EG, Gage AA. The five-year cure rate achieved
or 6 months: two-year results of URO-BCG-4 of noninvasive bladder cancers. Curr Opin Oncol. by cryosurgery for skin cancer. J Am Acad Dermatol.
multicenter study. Int J Urol. 2015;22(1):53–60. 2015;27(3):185–190. 1991;24(6 Pt 1):1002–1004.
559. Nouhaud FX, Rigaud J, Saint F, et al. Final results 575. Lammers RJ, Witjes JA, Inman BA, et al. The role of 595. Morton CA, McKenna KE, Rhodes LE, British
of the phase III URO-BCG 4 multicenter study: a combined regimen with intravesical chemotherapy Association of Dermatologists Therapy G, Audit
efficacy and tolerance of one-third dose BCG and hyperthermia in the management of non-muscle- S, the British Photodermatology G. Guidelines
maintenance in nonmuscle invasive bladder cancer. invasive bladder cancer: a systematic review. Eur for topical photodynamic therapy: update. Br J
Anticancer Drugs. 2017;28(3):335–340. Urol. 2011;60(1):81–93. Dermatol. 2008;159(6):1245–1266.
560. Huang Z, Liu H, Wang Y, Zhang C, Xu T. Deter- 576. Dinney CP, Greenberg RE, Steinberg GD. Intravesi- 596. Rigel DS, Stein Gold LF. The importance of early
mining optimal maintenance schedules for adjuvant cal valrubicin in patients with bladder carcinoma diagnosis and treatment of actinic keratosis. J Am
intravesical bacillus Calmette-Guérin immuno- in situ and contraindication to or failure after Acad Dermatol. 2013;68(1 suppl 1):S20–S27.
therapy in non-muscle-invasive bladder cancer: bacillus Calmette-Guérin. Urol Oncol. 2013;31(8): 597. Gupta AK, Paquet M, Villanueva E, Brintnell W.
a systematic review and network meta-analysis. 1635–1642. Interventions for actinic keratoses. Cochrane Database
Curr Med Res Opin. 2017;1–9. 577. Steinberg G. Valstar (valrubicin): Sterile Solution Syst Rev. 2012;(12):CD004415.
561. Shelley MD, Court JB, Kynaston H, Wilt TJ, Coles for Intravesical Instillation; 2008. https://www 598. Krawtchenko N, Roewert-Huber J, Ulrich M, Mann
B, Mason M. Intravesical bacillus Calmette-Guérin .accessdata.fda.gov/drugsatfda_docs/label/ I, Sterry W, Stockfleth E. A randomised study of
versus mitomycin C for Ta and T1 bladder cancer. 2011/020892s013lbl.pdf. topical 5% imiquimod vs. topical 5-fluorouracil vs.
Cochrane Database Syst Rev. 2003;(3):CD003231. 578. Sternberg IA, Dalbagni G, Chen LY, Donat SM, cryosurgery in immunocompetent patients with
562. Böhle A, Bock PR. Intravesical bacille Calmette- Bochner BH, Herr HW. Intravesical gemcitabine actinic keratoses: a comparison of clinical and
Guérin versus mitomycin C in superficial bladder for high risk, nonmuscle invasive bladder cancer histological outcomes including 1-year follow-up.
cancer: formal meta-analysis of comparative studies after bacillus Calmette-Guérin treatment failure. Br J Dermatol. 2007;157(suppl 2):34–40.
on tumor progression. Urology. 2004;63(4):682–686; J Urol. 2013;190(5):1686–1691. 599. Walker JL, Siegel JA, Sachar M, et al. 5-Fluorouracil
discussion 686–687. 579. Skinner EC, Goldman B, Sakr WA, et al. SWOG for actinic keratosis treatment and chemoprevention:
563. Shelley MD, Mason MD, Kynaston H. Intravesical S0353: phase II trial of intravesical gemcitabine in a randomized controlled trial. J Invest Dermatol.
therapy for superficial bladder cancer: a systematic patients with nonmuscle invasive bladder cancer and 2017;137(6):1367–1370.
review of randomised trials and meta-analyses. Cancer recurrence after 2 prior courses of intravesical bacillus 600. Lebwohl M, Dinehart S, Whiting D, et al. Imiqui-
Treat Rev. 2010;36(3):195–205. Calmette-Guérin. J Urol. 2013;190(4):1200–1204. mod 5% cream for the treatment of actinic keratosis:
564. Lamm D, Persad R, Colombel M, Brausi M. 580. Shelley MD, Jones G, Cleves A, Wilt TJ, Mason results from two phase III, randomized, double-blind,
Maintenance bacillus Calmette-Guérin: the standard MD, Kynaston HG. Intravesical gemcitabine therapy parallel group, vehicle-controlled trials. J Am Acad
of care for the prophylaxis and management of for non-muscle invasive bladder cancer (NMIBC): Dermatol. 2004;50(5):714–721.
intermediate- and high-risk non-muscle-invasive a systematic review. BJU Int. 2012;109(4):496– 601. Szeimies RM, Gerritsen MJ, Gupta G, et al.
bladder cancer. Eur Urol Suppl. 2010;9(9):715–734. 505. Imiquimod 5% cream for the treatment of actinic
565. Fuge O, Vasdev N, Allchorne P, Green JS. Immu- 581. Pagliarulo V, Ancona P, Martines I, et al. Celecoxib keratosis: results from a phase III, randomized,
notherapy for bladder cancer. Res Rep Urol. 2015;7: for the prevention of nonmuscle invasive bladder double-blind, vehicle-controlled, clinical trial with
65–79. cancer: results from a matched control study. Ther histology. J Am Acad Dermatol. 2004;51(4):547–555.
566. Bazarbashi S, Raja MA, El Sayed A, et al. Prospec- Adv Urol. 2015;7(6):303–311. 602. Korman N, Moy R, Ling M, et al. Dosing with 5%
tive phase II trial of alternating intravesical Bacillus 582. El-Arabey AA. New insight for metformin against imiquimod cream 3 times per week for the treatment
Calmette-Guérin (BCG) and interferon alpha IIB bladder cancer. Genes Environ. 2017;39:13. of actinic keratosis: results of two phase 3, random-
in the treatment and prevention of superficial tran- 583. Zhu Z, Wang X, Shen Z, Lu Y, Zhong S, Xu C. Risk ized, double-blind, parallel-group, vehicle-controlled
sitional cell carcinoma of the urinary bladder: pre- of bladder cancer in patients with diabetes mellitus: trials. Arch Dermatol. 2005;141(4):467–473.
liminary results. J Surg Oncol. 2000;74(3):181–184. an updated meta-analysis of 36 observational studies. 603. Wolf JE Jr, Taylor JR, Tschen E, Kang S. Topical 3.0%
567. Bazarbashi S, Soudy H, Abdelsalam M, et al. BMC Cancer. 2013;13:310. diclofenac in 2.5% hyaluronan gel in the treatment
Co-administration of intravesical bacillus Calmette- 584. Tseng CH. Metformin may reduce bladder cancer of actinic keratoses. Int J Dermatol. 2001;40(11):
Guérin and interferon alpha-2B as first line in risk in Taiwanese patients with type 2 diabetes. Acta 709–713.
treating superficial transitional cell carcinoma of the Diabetol. 2014;51(2):295–303. 604. Rosen RH, Gupta AK, Tyring SK. Dual mechanism
urinary bladder. BJU Int. 2011;108(7):1115–1118. 585. Nayan M, Bhindi B, Yu JL, et al. The effect of of action of ingenol mebutate gel for topical treat-
568. Joudi FN, Smith BJ, O’Donnell MA, National metformin on cancer-specific survival outcomes in ment of actinic keratoses: rapid lesion necrosis
BCGIPIG. Final results from a national multicenter diabetic patients undergoing radical cystectomy for followed by lesion-specific immune response. J Am
phase II trial of combination bacillus Calmette- urothelial carcinoma of the bladder. Urol Oncol. Acad Dermatol. 2012;66(3):486–493.
Guérin plus interferon alpha-2B for reducing 2015;33(9):386.e7–386.e13. 605. Lebwohl M, Swanson N, Anderson LL, Melgaard A,
recurrence of superficial bladder cancer. Urol Oncol. 586. Rieken M, Xylinas E, Kluth L, et al. Association of Xu Z, Berman B. Ingenol mebutate gel for actinic
2006;24(4):344–348. diabetes mellitus and metformin use with oncological keratosis. N Engl J Med. 2012;366(11):1010–1019.
374.e12 Part I: Science and Clinical Oncology

606. Inc LP. FDA Approves Picato; 2012. http://www 623. Curiel-Lewandrowski C, Swetter SM, Einspahr JG, 642. Bevona C, Goggins W, Quinn T, Fullerton J, Tsao
.prnewswire.com/news-releases/fda-approves-picato et al. Randomized, double-blind, placebo-controlled H. Cutaneous melanomas associated with nevi. Arch
-ingenol-mebutate-gel-the-first-and-only-topical trial of sulindac in individuals at risk for melanoma: Dermatol. 2003;139(12):1620–1624; discussion
-actinic-keratosis-ak-therapy-with-2-or-3- evaluation of potential chemopreventive activity. 1624.
consecutive-days-of-once-daily-dosing-138033448 Cancer. 2012;118(23):5848–5856. 643. Chiu LC, Tong KF, Ooi VE. Cytostatic and cytotoxic
.html. 624. Thomas RM, Worswick S, Aleshin M. Retinoic acid effects of cyclooxygenase inhibitors and their synergy
607. US Food and Drug Administration. FDA Drug for treatment of systemic sclerosis and morphea: a with docosahexaenoic acid on the growth of human
Safety Communication: FDA warns of severe adverse literature review. Dermatol Ther. 2017;30(2). skin melanoma A-375 cells. Biomed Pharmacother.
events with application of Picato (ingenol mebutate) 625. Muller-Decker K. Cyclooxygenase-dependent 2005;59(suppl 2):S293–S297.
gel for skin condition; requires label changes; 2015. signaling is causally linked to non-melanoma skin 644. Ottino P, Duncan JR. Effect of vitamin E and
https://www.fda.gov/Drugs/DrugSafety/ucm459142. carcinogenesis: pharmacological, genetic, and clini- indomethacin treatment on adenylate cyclase activity,
htm. cal evidence. Cancer Metastasis Rev. 2011;30(3–4): PGE2 and cAMP levels in murine melanoma cells.
608. Ianhez M, Fleury LF Jr, Miot HA, Bagatin E. 343–361. Prostaglandins Leukot Essent Fatty Acids. 1997;56(2):
Retinoids for prevention and treatment of actinic 626. Elmets CA, Ledet JJ, Athar M. Cyclooxygenases: medi- 143–149.
keratosis. An Bras Dermatol. 2013;88(4):585–593. ators of UV-induced skin cancer and potential targets 645. Duff M, Stapleton PP, Mestre JR, et  al.
609. Bavinck JN, Tieben LM, Van der Woude FJ, et al. for prevention. J Invest Dermatol. 2014;134(10): Cyclooxygenase-2 inhibition improves macrophage
Prevention of skin cancer and reduction of keratotic 2497–2502. function in melanoma and increases the antineoplas-
skin lesions during acitretin therapy in renal trans- 627. Elmets CA, Viner JL, Pentland AP, et al. Chemopre- tic activity of interferon gamma. Ann Surg Oncol.
plant recipients: a double-blind, placebo-controlled vention of nonmelanoma skin cancer with celecoxib: 2003;10(3):305–313.
study. J Clin Oncol. 1995;13(8):1933–1938. a randomized, double-blind, placebo-controlled trial. 646. Nakata K, Kashimoto S, Yoshida H, Oku T,
610. Kadakia KC, Barton DL, Loprinzi CL, et al. J Natl Cancer Inst. 2010;102(24):1835–1844. Nakamura S. Augmented antitumor effect of recom-
Randomized controlled trial of acitretin versus 628. Gurpinar E, Grizzle WE, Piazza GA. NSAIDs binant human interleukin-1 alpha by indomethacin.
placebo in patients at high-risk for basal cell or inhibit tumorigenesis, but how? Clin Cancer Res. Cancer Res. 1988;48(3):584–588.
squamous cell carcinoma of the skin (North Central 2014;20(5):1104–1113. 647. Harris RE, Beebe-Donk J, Namboodiri KK. Inverse
Cancer Treatment Group Study 969251). Cancer. 629. Meyskens FL Jr, Gerner EW. Development of difluo- association of non-steroidal anti-inflammatory drugs
2012;118(8):2128–2137. romethylornithine (DFMO) as a chemoprevention and malignant melanoma among women. Oncol
611. Weinstock MA, Bingham SF, Digiovanna JJ, agent. Clin Cancer Res. 1999;5(5):945–951. Rep. 2001;8(3):655–657.
et al. Tretinoin and the prevention of keratinocyte 630. Bailey HH, Kim K, Verma AK, et al. A randomized, 648. Ramirez CC, Ma F, Federman DG, Kirsner RS. Use
carcinoma (basal and squamous cell carcinoma of the double-blind, placebo-controlled phase 3 skin cancer of cyclooxygenase inhibitors and risk of melanoma in
skin): a veterans affairs randomized chemoprevention prevention study of {alpha}-difluoromethylornithine high-risk patients. Dermatol Surg. 2005;31(7 Pt 1):
trial. J Invest Dermatol. 2012;132(6):1583–1590. in subjects with previous history of skin cancer. 748–752.
612. Levine N, Moon TE, Cartmel B, et al. Trial of retinol Cancer Prev Res (Phila). 2010;3(1):35–47. 649. Joosse A, Koomen ER, Casparie MK, Herings
and isotretinoin in skin cancer prevention: a random- 631. Kreul SM, Havighurst T, Kim K, et al. A phase III RM, Guchelaar HJ, Nijsten T. Non-steroidal
ized, double-blind, controlled trial. Southwest Skin skin cancer chemoprevention study of DFMO: long- anti-inflammatory drugs and melanoma risk: large
Cancer Prevention Study Group. Cancer Epidemiol term follow-up of skin cancer events and toxicity. Dutch population-based case-control study. J Invest
Biomarkers Prev. 1997;6(11):957–961. Cancer Prev Res (Phila). 2012;5(12):1368–1374. Dermatol. 2009;129(11):2620–2627.
613. Clouser MC, Roe DJ, Foote JA, Harris RB, Alberts 632. Tang JY, Mackay-Wiggan JM, Aszterbaum M, 650. Curiel-Lewandrowski C, Nijsten T, Gomez ML,
DS. Dose response of retinol and isotretinoin in the et al. Inhibiting the hedgehog pathway in patients Hollestein LM, Atkins MB, Stern RS. Long-term use
prevention of nonmelanoma skin cancer recurrence. with the basal-cell nevus syndrome. N Engl J Med. of nonsteroidal anti-inflammatory drugs decreases the
Nutr Cancer. 2010;62(8):1058–1066. 2012;366(23):2180–2188. risk of cutaneous melanoma: results of a United States
614. Moon TE, Levine N, Cartmel B, et al. Design and 633. Sekulic A, Migden MR, Oro AE, et al. Efficacy case-control study. J Invest Dermatol. 2011;131(7):
recruitment for retinoid skin cancer prevention and safety of vismodegib in advanced basal-cell 1460–1468.
(SKICAP) trials. The Southwest Skin Cancer Preven- carcinoma. N Engl J Med. 2012;366(23):2171–2179. 651. Famenini S, Duan L, Young L. Aspirin use and
tion Study Group. Cancer Epidemiol Biomarkers Prev. 634. Wang A, Stefanick ML, Kapphahn K, et al. Relation melanoma: a UCLA pilot study. J Am Acad Dermatol.
1995;4(6):661–669. of statin use with non-melanoma skin cancer: pro- 2015;72(6):1094–1095.
615. Moon TE, Levine N, Cartmel B, Bangert JL. spective results from the Women’s Health Initiative. 652. ASPREE Investigator Group. Study design of ASPirin
Retinoids in prevention of skin cancer. Cancer Lett. Br J Cancer. 2016;114(3):314–320. in Reducing Events in the Elderly (ASPREE): a
1997;114(1–2):203–205. 635. Wang A, Tang JY, Stefanick ML. Relation of statin randomized, controlled trial. Contemp Clin Trials.
616. Butler GJ, Neale R, Green AC, Pandeya N, use with non-melanoma skin cancer: prospective 2013;36(2):555–564.
Whiteman DC. Nonsteroidal anti-inflammatory drugs results from the Women’s Health Initiative. Womens 653. Uzarska M, Czajkowski R, Schwartz RA, Bajek
and the risk of actinic keratoses and squamous cell Health (Lond). 2016;12(5):453–455. A, Zegarska B, Drewa T. Chemoprevention of
cancers of the skin. J Am Acad Dermatol. 2005;53(6): 636. Tang JY, Fu T, Leblanc E, et al. Calcium plus vitamin skin melanoma: facts and myths. Melanoma Res.
966–972. D supplementation and the risk of nonmelanoma 2013;23(6):426–433.
617. Johannesdottir SA, Chang ET, Mehnert F, Schmidt and melanoma skin cancer: post hoc analyses of 654. Linden KG, Leachman SA, Zager JS, et al. A
M, Olesen AB, Sorensen HT. Nonsteroidal anti- the women’s health initiative randomized controlled randomized, double-blind, placebo-controlled phase
inflammatory drugs and the risk of skin cancer: a trial. J Clin Oncol. 2011;29(22):3078–3084. II clinical trial of lovastatin for various endpoints
population-based case-control study. Cancer. 2012; 637. Frieling UM, Schaumberg DA, Kupper TS, of melanoma pathobiology. Cancer Prev Res (Phila).
118(19):4768–4776. Muntwyler J, Hennekens CH. A randomized, 2014;7(5):496–504.
618. Asgari MM, Chren MM, Warton EM, Friedman 12-year primary-prevention trial of beta carotene 655. Asgari MM, Brasky TM, White E. Association of
GD, White E. Association between nonsteroidal anti- supplementation for nonmelanoma skin cancer in the vitamin A and carotenoid intake with melanoma
inflammatory drug use and cutaneous squamous cell physician’s health study. Arch Dermatol. 2000;136(2): risk in a large prospective cohort. J Invest Dermatol.
carcinoma. Arch Dermatol. 2010;146(4):388–395. 179–184. 2012;132(6):1573–1582.
619. Grau MV, Baron JA, Langholz B, et al. Effect of 638. Ferrucci LM, Cartmel B, Molinaro AM, Leffell DJ, 656. Miura K, Green AC. Dietary antioxidants and
NSAIDs on the recurrence of nonmelanoma skin Bale AE, Mayne ST. Tea, coffee, and caffeine and melanoma: evidence from cohort and intervention
cancer. Int J Cancer. 2006;119(3):682–686. early-onset basal cell carcinoma in a case-control studies. Nutr Cancer. 2015;67(6):867–876.
620. Muranushi C, Olsen CM, Pandeya N, Green AC. study. Eur J Cancer Prev. 2014;23(4):296–302. 657. Caini S, Masala G, Saieva C, et al. Coffee, tea
Aspirin and nonsteroidal anti-inflammatory drugs 639. Miura K, Hughes MC, Arovah NI, van der Pols JC, and melanoma risk: findings from the European
can prevent cutaneous squamous cell carcinoma: Green AC. Black tea consumption and risk of skin Prospective Investigation into Cancer and Nutrition.
a systematic review and meta-analysis. J Invest cancer: an 11-year prospective study. Nutr Cancer. Int J Cancer. 2017;140(10):2246–2255.
Dermatol. 2015;135(4):975–983. 2015;67(7):1049–1055. 658. Chhabra G, Ndiaye MA, Garcia-Peterson LM,
621. Clouser MC, Roe DJ, Foote JA, Harris RB. Effect 640. Chen AC, Martin AJ, Choy B, et al. A Phase 3 Ahmad N. Melanoma chemoprevention: current
of non-steroidal anti-inflammatory drugs on non- Randomized trial of nicotinamide for skin-cancer status and future prospects. Photochem Photobiol.
melanoma skin cancer incidence in the SKICAP- chemoprevention. N Engl J Med. 2015;373(17):1618– 2017.
AK trial. Pharmacoepidemiol Drug Saf. 2009; 1626. 659. Sharma A, Koldovsky U, Xu S, et al. HER-2 pulsed
18(4):276–283. 641. Marks R, Dorevitch AP, Mason G. Do all melanomas dendritic cell vaccine can eliminate HER-2 expres-
622. Zhu Y, Cheng Y, Luo RC, Li AM. Aspirin for the come from “moles”? A study of the histological asso- sion and impact ductal carcinoma in situ. Cancer.
primary prevention of skin cancer: a meta-analysis. ciation between melanocytic naevi and melanoma. 2012;118(17):4354–4362.
Oncol Lett. 2015;9(3):1073–1080. Australas J Dermatol. 1990;31(2):77–80.
Screening and Early Detection 23 
Therese Bevers, Hashem El-Serag, Samir Hanash, Aaron P. Thrift,
Kenneth Tsai, Karen Colbert Maresso, and Ernest Hawk

S UMMARY OF K EY P OI NT S
• Cancer incidence is on the rise in the risk-stratified screening for common need for routine cervical cancer
United States and worldwide. cancers to improve the effectiveness screening according to age and
Beyond therapeutics, substantial of screening. clinical history.
reduction in cancer mortality • Overdiagnosis is finding cancers that • Because most risk factors for liver
necessitates improved understanding would never become clinically cancer in the United States and
of cancer risk, implementation of relevant in a person’s lifetime. This other developed countries are
effective preventive intervention has been an issue mainly in breast associated with cirrhosis, patients
strategies, and early detection of and prostate cancer screening. with cirrhosis are the target of
cancers that are likely to progress. • Systematic reviews of randomized cancer prevention and surveillance
• Genomic- and proteomic-based controlled trials (RCTs) of screening efforts for hepatocellular carcinoma.
approaches have the potential to mammography have demonstrated • An effective screening test for
refine risk assessment and an approximate 20% mortality ovarian cancer has not been
stratification, allowing for more reduction in women ages 40 to identified. The two tests commonly
tailored screening and early 74 years. investigated for ovarian cancer
detection strategies. However, the • Guaiac-based fecal occult blood screening are CA125 and
use of such approaches is still in its tests (gFOBTs) have the strongest transvaginal ultrasound.
early stages. direct evidence supporting their use • Randomized clinical trials for skin
• In the United States, population- as a colorectal cancer screening cancer screening are unlikely to be
based screening tests are currently modality, with a 32% reduction in practical. The US Preventive
recommended for breast, cervical, colorectal cancer mortality with Services Task Force (USPSTF)
colon, and lung cancers. annual use. statement on skin cancer is
Individualized decision making after • Lung cancer screening with forward-looking because it opens the
a discussion with a clinician is low-dose computed tomography door to case-control designs that are
recommended for men in the case of resulted in an approximate 20% more practical, while acknowledging
prostate cancer screening. reduction in lung cancer mortality in that strong evidence of benefits and
• Discordant professional society the National Lung Screening Trial. harms is lacking, including for
recommendations for screening have • Human papillomavirus (HPV) nonmelanoma skin cancer.
accentuated the need to implement vaccination does not remove the

Although substantial progress is currently being made in the develop- for at least 13 different cancers. Elucidation of metabolic, immune,
ment of novel and more effective therapeutics, cancer remains a largely and other molecular profiles that contribute to the increased risk
unsolved clinical problem with high mortality. Cancer incidence is would allow for more focused screening strategies for persons with
on the rise in the United States and worldwide. Between 2005 and these risk profiles and would allow implementation of targeted preven-
2015, the number of cancer cases increased by 33%, owing in part tion strategies aimed at the cancer(s) for which they are at risk.3,4
to population aging, which contributed 16%, and population growth
and changes in age-specific rates, which contributed the remainder.1 RISK ASSESSMENT
Cancer incidence and the societal cancer burden are expected to increase
further.2 Beyond therapeutics, substantial reduction in cancer mortality Guidelines are currently available for risk assessment in the clinical
necessitates improved understanding of cancer risk, implementation setting through several organizations. Guidelines allow a determination
of effective preventive intervention strategies, and early detection of of a subject’s risk for the common cancers and the associated options
cancers that are likely to progress to avoid the problem of overdiagnosis, for screening. Organizations creating guidelines include the National
all of which represent substantial challenges that can be overcome. Comprehensive Cancer Network (NCCN; https://www.nccn.org/),
Identifying individuals at increased risk of developing a particular the American Cancer Society (ACS; https://www.acs.org/), and other
cancer type would allow for a range of preventive interventions to be advocacy and professional societies. Family history and other patient
implemented, from altered lifestyle and health behaviors to the use characteristics are often important determinants of risk. In general,
of vaccines, and early detection of particular cancer(s) for which these individuals are first defined as being at either average risk or increased
persons are at risk. A case in point is obesity, which is a risk factor risk for a particular cancer. For example, the NCCN Version 2.2016

375
376 Part I: Science and Clinical Oncology

colorectal cancer (CRC) screening guidelines identify a person as


being at “average risk” based on age, negative family history, and no Table 23.1  Performance Characteristics of
prior personal history of inflammatory bowel disease, adenoma, or Screening Tests
sessile serrated polyp or CRC. Risk is increased with a positive family
Performance Practical
or personal history of cancer or precancer. High-risk syndromes include
Characteristic Definition Formula Application
Lynch syndrome and a number of other syndromes. Screening guidelines
for CRC and the modalities used for screening vary based on the risk Accuracy Proportion of TPs + Have disease and
profile. Other common cancers for which risk assessment and screening correct test results TNs/TPs have positive test
guidelines are available include breast, cervical, lung, and prostate. + TNs + result + do not
Given the widespread use of the prostate-specific antigen (PSA) test FPs + FNs have disease and
for prostate cancer screening, the NCCN guidelines for early detection have negative
test result/all test
of prostate cancer stratify men based on their age and PSA levels.
results
There is currently substantial interest in bolstering traditional
guidelines by adding genomic and other “omic” profiles to other Sensitivity Ability of the test TPs/TPs + Have disease and
personal characteristics and family history to determine the need for to correctly identify FNs have positive test
and frequency of screening.5,6 The field is still in its early stages.7 In someone with the results/true
disease as positive disease
the case of breast cancer, susceptibility genes include highly penetrant
mutations in the BRCA1 and BRCA2 genes, the testing for which is Specificity Ability of the test TNs/TNs Do not have
indicated by a positive family history of breast cancer. Several other to correctly identify + FPs disease and have
moderately penetrant mutations in other genes and more common someone without negative test
genomic variants with a modest increase in risk have been identified.8 the disease as results/true no
negative disease
These may very well influence risk assessments and guide screening
in the near future. Positive Probability that TPs/TPs + Have disease and
predictive someone with a FPs have positive test
value (PPV) positive test result results/all
SCREENING AND EARLY DETECTION actually has the positive test
disease results
Although advances in screening and early detection are essential for
progress in both the prevention and treatment of cancer, the incorpora- Negative Probability that TNs/TNs Do not have
tion of such advances into the clinic is challenging and demands a predictive someone with a + FNs disease and have
careful weighing of all potential risks and benefits. At least three value (NPV) negative test result negative test
does not have the results/all
criteria must be fulfilled for a cancer screening test to be useful9:
disease negative test
results
1. The test must detect the disease earlier than routine methods.
2. Earlier treatment must lead to improved outcomes. FN, False negative; FP, false positive; TN, true negatives; TP, true positive.
3. The benefits of screening must be greater than the risks of any
subsequent diagnostic and therapeutic treatments.

A screening test is assessed by its performance characteristics, which disease, given the test result. The PPV is influenced by the prevalence
include its sensitivity and specificity, its accuracy, and its positive of the disease in the population screened, such that the PPV will be
predictive value (PPV) and negative predictive value (NPV) (Table higher at higher disease prevalence. In addition, when the disease in
23.1). Sensitivity can be considered the “true-positive” rate, and question is relatively rare within a population, as is the case with most
specificity can be considered the “true-negative” rate. If a screening cancers, the specificity of the test being used also matters greatly
test were 100% sensitive, every individual with the disease in question because most people who will be screened will not have the disease.
within the population would be identified as such; and if it were In these patients, increasing the specificity of a screening test will
100% specific, it would identify every healthy individual as not having improve its PPV.
the disease. In reality, however, there is no screening test that achieves Observational data supporting the use of screening tests do not
this ideal. Sensitivity and specificity are inversely related, such that rule out the potential for these tests to mislead as a result of at least
increasing the sensitivity of a test will result in more false positives, two important potential biases: lead-time and length biases (Fig. 23.1).
whereas increasing the specificity will result in more false negatives.9 To minimize the influence of these biases, the development of effective
Striking an acceptable balance between these two performance screening tests should ideally culminate in well-conceived and well-
characteristics is challenging and depends on the disease in question. conducted randomized controlled trials (RCTs) that assess a cancer-
In the case of many types of cancers that are curable only in the earlier related, if not overall, mortality end point.
stages of the disease, it may be desirable to minimize false negatives, Implementation of screening programs for common cancers, notably
because these could result in a patient’s delayed diagnosis and cancer- cervical, breast, lung, and colon cancers, has been shown to reduce
specific death, and accept more false positives (i.e., higher sensitivity the mortality associated with these cancers. However, whenever there
than specificity). However, where the balance is struck ultimately is a screenable disease that is prevalent but potentially indolent, there
depends on the nature and severity of the specific disease being screened is the potential for overdiagnosis and overtreatment, resulting in a
for and the effectiveness of downstream interventions and treatments range of personal and social costs that may ultimately outweigh the
for that disease. intended benefits. Overdiagnosis is the identification by screening of
The sensitivity and specificity of a screening test are critical to a cancer that would never have caused symptoms or adversely influenced
consider in the design of any population-based screening program. the health of an individual during his or her lifetime; and overtreatment
However, at least at the clinical level, it is also useful to appreciate is the treatment of such an identified cancer. Breast and prostate
the PPV and NPV of the screening test. The PPV is the proportion cancer screening are two examples in which these issues have challenged
of patients who test positive who actually have the disease, and the screening efforts. The use of the PSA test for prostate cancer screening
NPV is the proportion of those who test negative who truly do not has been particularly controversial.10 The American Urological Associa-
have the disease. Thus these values provide a clinician with an estimate tion recommended shared decision making for men ages 55 to 69
of the probability that his or her patient does or does not have the years considering PSA-based screening, a target age group for whom
Screening and Early Detection  •  CHAPTER 23 377

women ages 45 to 54 years should be screened annually, although


Lead-time bias
women ages 40 to 44 should have the opportunity to begin screening
No screen Sx-Dx if they so choose, and women age 55 years and older should transition
Death to biennial screening or have the opportunity to continue screening
annually. Screening should continue as long as a woman’s overall
health is good and she has a life expectancy of 10 years or longer.
Clearly, guidelines for cancer screening are continually evolving.
Survival
Discordant professional society recommendations for screening have
Screen CT-Dx accentuated the need to implement risk-stratified screening for common
Death
cancers to improve the effectiveness of screening.16 Risk stratification
may be based on an individual’s genotype or other types of molecular
markers in addition to subject characteristics. For example, a novel
approach to risk-based breast cancer screening has been proposed that
Lead-time integrates clinical risk factors, breast density, a polygenic risk score
With screening, the lead time in diagnosis prolongs representing the cumulative effects of genetic variants, and sequencing
survival even if death is not delayed. for moderate- and high-penetrance germline mutations.17 The added
costs versus benefits of molecular testing to guide screening for common
cancers requires further evaluation.
Length bias Table 23.2 presents a summary of the USPSTF classification of the
evidence for various cancer screening tests in average-risk individuals.
Screening
Indolent Symptoms
cancer begins and Dx
Death BREAST CANCER
Breast cancer is the most commonly occurring cancer in women and
the second leading cause of cancer death. Since 1989, the number of
Detectable deaths from breast cancers has decreased owing to improvements in
preclinical phase treatment as well as screening.18 Screening provides an opportunity
Aggressive
for the early detection of breast cancer, which has been shown to
Sx and Dx reduce not only breast cancer mortality but also treatment morbidity.
cancer begins Death Screening tends to Nonmodifiable risk factors include older age, a personal or
detect more indolent
cancers.
family history of breast or ovarian cancer including women with a
genetic mutation for breast cancer, a history of premalignant breast
lesions such as atypical hyperplasia (AH) or lobular carcinoma in situ
(LCIS), or a history of radiation exposure between the ages of 10
Figure 23.1  •  Schematic depictions of lead-time and length biases. Lead
time refers to the amount of time between screen-detected diagnosis and and 30 years.18
symptom-detected diagnosis, and it appears to prolong survival in screened Modifiable or potentially modifiable risk factors for breast cancer
individuals, although death is not actually delayed. Length bias refers to the include increased breast density, moderate to heavy alcohol use, weight
fact that screening is more likely to detect indolent or slow-growing cancers gain after the age of 18, being overweight or obese (for postmenopausal
than it is to detect more aggressive forms. Persons who are screened will breast cancer), physical inactivity, and use of exogenous estrogen plus
therefore have better survival rates than persons who are not screened; however, progesterone (e.g., in postmenopausal hormone therapy).18
these better rates are not actually a result of the screening itself, but rather of Reproductive factors that increase breast cancer risk include a long
the more indolent nature of cancers that are detected with screening. (From menstrual history (early age of menarche or late age of menopause)
National Cancer Institute. https://www.cancer.gov/about-cancer/screening/ and nulliparity or late age of first pregnancy.18 These endocrine risk
research/what-screening-statistics-mean.)
factors demonstrate the responsiveness of breast cancer to antiestrogen
drugs and have led to the use of antiestrogen strategies for the prevention
benefits may outweigh harms.11 On the other hand, the US Preventive of breast cancer.
Services Task Force (USPSTF) has discouraged (i.e., assigned a D
recommendation to) the use of the PSA for prostate cancer screening, Risk Modeling and Assessment
although the organization is now in the process of updating this recom-
mendation. Nevertheless, it is viewed that the USPSTF decision affected Breast cancer risk assessment is relatively sophisticated compared with
the number and distribution of prostate cancer diagnoses in the United other cancers. The Gail model, a multivariable model used to assess
States.12 Mammography screening for breast cancer has been widely risk of breast cancer based on age, family history of breast cancer in
adopted in many Western countries. However, there has been growing first-degree relatives (FDRs), breast biopsy history, and the presence
debate and concern about the benefits and harms of screening with of AH and various other endocrine features,19,20 is often used to
mammography.13 Guidelines and recommendations for screening determine candidates for preventive therapy with antiestrogen drugs
mammography have varied among countries. A working group such as tamoxifen or raloxifene. Although this model is generally
assembled by the International Agency for Research on Cancer (IARC) accurate for populations, its ability to predict if an individual woman
assessed the cancer-preventive and adverse effects of different methods will get breast cancer is limited. Newer risk assessment models such
of screening for breast cancer.14 The group recognized that the relevance as the Breast Cancer Surveillance Consortium model also consider
of RCTs conducted more than 20 years ago should be questioned. mammographic density in determining the risk of development of
After a careful evaluation of the balance between the benefits and breast cancer.21–23
adverse effects of mammographic screening, the group concluded that For women with a strong family history of breast cancer (with or
there is a net benefit from screening women ages 50 to 69 years of without a family history of ovarian cancer), other models may be
age. However, the merits of mammographic screening have been more accurate. The Tyrer-Cuzick model estimates a woman’s risk of
questioned by others.15 The ACS updated its 2003 breast cancer developing breast cancer through use of many of the variables used
screening guidelines for women at average risk of breast cancer and in the Gail model but includes a more expansive family history. It
issued qualified recommendations. The guidelines of 2015 state that also estimates her risk of a BRCA mutation and may be used to
378 Part I: Science and Clinical Oncology

Table 23.2  US Preventive Services Task Force (USPSTF) Grade Definitions and Associated Cancer
Screening Tests in Average-Risk Individuals as of June 2017a,b
Grade Definition Suggestions for Practice Current Screening Test Recommendations
A The USPSTF recommends the service. Offer or provide this service. Cervical cancer screening
There is high certainty that the net benefit for women ages 21–65 with pap smear
is substantial. or
pap smear with HPV-testing in women ages 30–65
Colorectal cancer screening in adults ages 50–75
B The USPSTF recommends the service. Offer or provide this service. Breast cancer screening (biennial) for women ages 50–74
There is high certainty that the net benefit Lung cancer screening in adults ages 55–80 with a
is moderate or there is moderate certainty history of smoking
that the net benefit is moderate to
substantial.
C The USPSTF recommends selectively Offer or provide this service for Breast cancer screening for women ages 40–49
offering or providing this service to selected patients depending Colorectal cancer screening in adults ages 76–85
individual patients based on professional on individual circumstances. Prostate cancer screening in men ages 55–69c
judgment and patient preferences. There
is at least moderate certainty that the net
benefit is small.
D The USPSTF recommends against the Discourage the use of this Cervical cancer screening in women younger than age 30
service. There is moderate or high service. with HPV testing alone or in combination with cytologic
certainty that the service has no net assessment
benefit or that the harms outweigh the Cervical cancer screening in women younger than age 21
benefits. Cervical cancer screening in women age 65+ who have
had adequate prior screening
Ovarian cancer screening
Pancreatic cancer screening
Prostate cancer screening in men age 70+c
I The USPSTF concludes that the current Read the clinical considerations Breast cancer screening in women age 75+
evidence is insufficient to assess the section of USPSTF Oral cancer screening
balance of benefits and harms of the Recommendation Statement. If Skin cancer screening
service. Evidence is lacking, of poor quality, the service is offered, patients
or conflicting, and the balance of benefits should understand the
and harms cannot be determined. uncertainty about the balance
of benefits and harms.

a
Based on grade definitions after July 2012.
b
The USPSTF defines certainty as “likelihood that the USPSTF assessment of the net benefit of a preventive service is correct.” The net benefit is defined as benefit minus
harm of the preventive service as implemented in a general, primary care population. The USPSTF assigns a certainty level based on the nature of the overall evidence available
to assess the net benefit of a preventive service.
c
Draft recommendation as of June 2017.
HPV, Human papillomavirus
Adapted with permission of the U.S Preventive Services Task Force.

determine if genetic testing is recommended.24 Other models developed BSE arm received intensive instruction in BSE technique. Compliance
for the assessment of a genetic mutation in a specific woman include was encouraged through feedback and reinforcement sessions as well
the following: Berry-Parmigiani-Aguilar (BRCA-Pro),25–27 Claus,28 as monthly reminders. After 10 to 11 years of follow-up, with 135
Couch,29 and the Breast and Ovarian Analysis of Disease Incidence breast cancer deaths in the instruction group and 131 in the control
and Carrier Estimation Algorithm (BOADICEA).30–32 Geneticists and group, there was no statistically significant difference between the two
physicians specializing in cancer risk assessment commonly use these arms in breast cancer mortality (RR, 1.04; 95% confidence interval
models to guide genetic testing decisions as well as recommendations [CI], 0.82–1.33; P = .72). In addition, no statistically significant
for breast screening. difference in breast cancer incidence or stage was seen. However, the
BSE group had a higher rate of false positives.33,34
Screening Nonetheless, it is recognized that women are the most likely person
to find a palpable breast cancer, with most being found during normal
Breast Awareness activities of daily living (e.g., showering, dressing). For this reason,
Breast self-examination (BSE) was a mainstay for decades in breast BSE has been replaced with the concept of breast awareness, which
cancer screening recommendations, but this recommendation changed recommends that women be familiar with their breasts and promptly
for most organizations in the early 2000s when findings from a large report any change. Differing from BSE, breast awareness does not
RCT of BSE in women in Shanghai, China noted no difference in involve formalized instruction in the clinical setting or reminders
breast cancer mortality between women performing BSE versus controls. (such as shower cards).
This trial randomized 266,064 Chinese women to receive instruction
on BSE or a topic unrelated to breast cancer. During the time this Clinical Breast Examination
study was conducted, women in China had access to mammography There are limited data on the effectiveness of clinical breast examination
only for diagnostic evaluation of a clinical finding. Women in the (CBE). Randomized trials comparing CBE versus no screening have
Screening and Early Detection  •  CHAPTER 23 379

not been performed. A review of controlled trials and case-control may also result in lower morbidity because surgery is often less extensive
studies that included CBE as part of the screening modality found and toxic systemic chemotherapy may be less frequently needed.42–45
sensitivity of CBE to be 54% and specificity, 94%.35 Systematic reviews of RCTs of screening mammography have
Whereas the ACS recommends against CBE, the NCCN recom- demonstrated an approximate 20% mortality reduction in women
mends a clinical encounter noting that it provides the opportunity ages 40 to -74 years.37,38 However, all but the Age Trial46 were initiated
to perform a number of important clinical activities that may not in the early 1980s or earlier. Although RCTs have demonstrated a
otherwise be done if a woman only obtains a screening mammogram mortality reduction, they are limited by the quality of mammography
(Table 23.3). In addition to performing breast cancer risk assessment, available at the time the study was conducted, with many cancers
advising on risk reduction strategies, including healthy lifestyle having to be 1 cm or greater to be detected.47 A meta-analysis of
interventions and risk-based screening recommendations, CBE is a observational case-control studies showed a significant 48% mortality
part of this encounter.36 reduction with modern screening mammography.48 Although obser-
vational data are limited by bias, these data are believed by many to
Screening Mammography better reflect contemporary screening practices in which the detection
The primary benefit of screening mammography is a reduction in death of subcentimeter breast cancers is commonplace. Modeling studies
from breast cancer.36–41 Early detection with screening mammography have demonstrated a 29% to 54% mortality reduction with annual
mammographic screening, providing further support for a larger
mortality reduction than seen in the RCTs.49
Various guidelines from the ACS, USPSTF, and NCCN and recom-
Table 23.3  Clinical Encounter Activities Related to mendations from professional organizations, including the American
Breast Cancer Screeninga College of Radiology, American College of Obstetrics and Gynecology,
1. Assess breast cancer risk and refer to a genetic counselor if a American College of Physicians, and American Academy of Family
significant family history is identified. Physicians, are currently available to guide breast cancer screening in
2. Educate about lifestyles that reduce the risk of breast cancer— women at average risk (Table 23.4). A number of organizations recom-
specifically, the importance of exercise, maintaining a healthy body mend annual mammographic screening beginning in the 40s,36,40,50,51
weight, and limiting alcohol intake. whereas others recommend biennial screening beginning at age 50.41,52
3. For women at increased risk of breast cancer, counsel regarding All recommending organizations agree that screening beginning at
preventive therapies such as tamoxifen, raloxifene, or aromatase age 40 results in a mortality reduction and that annual screening leads
inhibitors.b to fewer breast cancer deaths than does biennial screening. Among
4. Provide risk-based breast cancer screening recommendations, women ages 40 to 49 years, the reduction in mortality ranges from
which may include screening breast magnetic resonance imaging 15% in RCTs to 47% in observational studies.38–40,53 Early detection
for women at increased risk. and successful treatment results in significantly more life-years gained
5. Perform clinical breast examination. for this decade than for any other decade.54 Organizations that recom-
a
mend against annual mammographic screening and delaying initiation
As recommended by the National Comprehensive Cancer Network (NCCN) of mammographic screening to the 50s cite concerns that the false
Clinical Practice Guidelines in Oncology: Breast Cancer Screening and Diagnosis
(Version 2.2016).36
positives and overdiagnosis of screening may not balance the benefits
b
Not approved by the US Food and Drug Administration for this indication. of screening for women in their 40s.41 However, the USPSTF analysis
Data from NCCN Clinical Practice Guidelines in Oncology: Breast Cancer Screening shows that the false-positive rate for the same screening interval (annual
and Diagnosis (Version 2.2016)36. or biennial) is equivalent for women in their 40s and 50s and that the

Table 23.4  Breast Cancer Screening Recommendations by Organization


Organization When to Initiate Screening Frequency of Screening When to Stop Screening
American Academy of Follow US Preventive Services Task Follow USPSTF recommendations Follow USPSTF recommendations
Family Physicians (AAFP) Force (USPSTF) recommendations
American Cancer Society Opportunity to begin screening at Annually from age 45–54 Continue screening mammography as
(ACS) ages 40–44 Biennially starting at age 55 with long as overall health is good and life
Regular screening starting at age 45 opportunity to continue annually expectancy is 10 yr or longer
American College of Annual screening starting at age 40 Annually Not specified
Obstetricians and
Gynecologists (ACOG)
American College of Individualized for women ages 40–49 Biennially Age 75 yr or older
Physicians (ACP) Regular screening starting at age 50 Women of any age with life expectancy
<10 yr
American College of annual screening starting at age 40 Annually Should be considered as long as the
Radiology (ACR) patient is in good health and is willing to
undergo additional testing if an
abnormality is detected
National Comprehensive Annual screening starting at age 40 Annually Upper age limit is not yet established
Cancer Network (NCCN) Consider comorbid conditions limiting
life expectancy (e.g., ≤10 yr) and whether
therapeutic interventions are planned
US Preventive Services Individualized for women ages 40–49 Biennially Insufficient evidence to recommend for
Task Force (USPSTF) Regular screening beginning at age 50 or against screening at age 75 or older
380 Part I: Science and Clinical Oncology

biopsy rate is actually higher for women in their 50s.41 In addition, it each year through a healthy diet, weight management, and physical
is recognized that women undergoing their first screening mammogram activity. Nonmodifiable risk factors include a personal or family history
have a higher recall rate owing to a lack of prior mammograms for of CRC or adenomatous polyps, including the inherited conditions
comparison. Moving the initiation of screening to age 50 would further of familial adenomatous polyposis (FAP) and hereditary nonpolyposis
increase the incidence of false-positive mammograms at this age. colorectal cancer (HNPCC), and a personal history of chronic inflam-
Most clinicians recognize overdiagnosis of breast cancer as less an matory bowel disease. In addition, the presence of adenomas at screening
issue of screening and more of a problem related to overtreatment. It is a strong predictor of CRC risk.
is clear that we need to understand which “breast cancers” do not Globally, the incidence of CRC varies greatly, with as much as a
require treatment. Clinical trials are opening to begin to investigate 10-fold variation in both sexes, with the highest rates found in Australia/
this question. Having said this, it is important to recognize that the New Zealand and the lowest in Western Africa.63
number of overdiagnosed breast cancers is not necessarily influenced Mortality rates vary somewhat less than do incidence rates, with
by the age of initiation of screening mammography or the interval in mortality being the highest in Central and Eastern Europe, and the
which screening occurs. It has been shown that breast cancers that lowest in Western Africa.63 Although more than half of new CRC
do not progress (i.e., an overdiagnosed breast cancer) also do not cases occur in more developed regions of the world, more than half
spontaneously resolve.55 These cancers will be diagnosed at the first of the deaths due to CRC occur in less developed regions.63
screening after their development, whether that is at age 40 or 50 (or
somewhere in between) and whether the screening occurs annually Risk Modeling and Assessment
or biennially.
Full-field digital mammography (FFDM) is the current standard Despite the development of numerous risk prediction models for
of care for breast cancer screening. Early data have shown that different populations, there is no validated tool with high discrimina-
tomosynthesis increases the detection of breast cancer and reduces tory power that can be easily applied in various settings to estimate
the recall rate.56–59 This suggests that this modality has the potential risk of colorectal neoplasia. Nevertheless, the Freedman risk model/
to provide a more favorable balance of benefits and risks of screening NCI’s Colorectal Cancer Risk Assessment tool, developed in 2009,
mammography. has been validated in an independent cohort (unlike many of the
Women who have a high risk of breast cancer because of a known other proposed models) and is relevant to a large proportion of the
inherited mutation, a strong family history with a greater than 20% US population that is eligible for CRC screening.64,65 Although the
lifetime risk of breast cancer, or prior exposure to therapeutic radiation model was developed and validated based on non-Hispanic white
between the ages 10 and 30 years should be more aggressively screened men and women, and its generalizability to other racial and ethnic
with supplemental breast magnetic resonance imaging (MRI) in addition groups is uncertain, it is believed that the variables that contribute to
to annual screening mammography.36,60,61 It is suggested that women the model have deep penetration into the US population, regardless
with high-risk lesions such as atypical ductal or lobular hyperplasia of race and ethnicity, and that it generalizes to nonwhite populations
or LCIS should also be considered for supplemental screening with within the United States better than do non-US models, of which
breast MRI based on emerging evidence.36 NCCN outlines screening there are many.66 The Freedman model originally predicted the future
strategies for these high-risk women, including age of initiation and 10- and 20-year absolute risk of CRC, with an area under the curve
frequency of screening.36 Although the optimal frequency and timing (AUC) of 0.61 (95% CI, 0.60–0.62) for men and 0.61 (95% CI,
of breast MRI scans is not well established, it is frequent practice to 0.59–0.62) for women, comparable to other cancer risk prediction
perform annual screening mammogram and annual screening breast models.65 In its current form, the model now predicts 5-year, 10-year,
MRI scans staggered every 6 months. This paradigm provides the and lifetime absolute CRC risk. Imperiale and colleagues tested the
opportunity for the detection of “interval” cancers (defined as cancers ability of the model to predict current risk of advanced neoplasia,
detected between scheduled screening examinations), which may occur reasoning that knowledge of current risk may more directly affect
more frequently in high-risk women. Breast MRI scanning is more short-term decisions regarding screening than knowledge of future
sensitive in detecting breast masses in dense breasts (such as in young risk, and that this could lead to increased adoption of CRC screening
women), whereas mammograms are more sensitive in detecting breast and to tailored screening strategies wherein those at higher risk for
calcifications. Therefore both are used to obtain optimal breast screening advanced neoplasia would be directed toward colonoscopy as the
in high-risk women. Other screening tests, such as breast ultrasonog- preferred method of screening and those with a lower risk of such a
raphy, are not routinely recommended for breast cancer screening. finding toward less invasive screening methods.66 Results of the study
Over half of US states now mandate that women be informed about demonstrated that the model’s discriminatory power for predicting
the increased risk of breast cancer associated with dense breasts and current advanced neoplasia was in the moderate-to-good range, with
the option of supplemental screening, but specific tests to recommend an AUC of 0.71 (95% CI, 0.68–0.73) in the 5-year analysis, which
in these women remains an area of investigation. is better than the AUC of the model in its original validation cohort
Research is being done to identify even more effective screening as well as the Gail model when applied to an independent cohort to
technologies and approaches. In addition to enhanced imaging modali- estimate 5-year breast cancer risk. The authors concluded that the tool
ties, these involve the evaluation of biomarkers in serum, breast duct can be used to estimate current risk of advanced neoplasia, making it
fluid, saliva, and tears. These strategies offer great hope for improving potentially useful for tailoring and improving CRC screening efficiency
breast cancer early detection. It is anticipated that in the future, among those at average-risk of CRC.66 The Cleveland Clinic Score
blood- or biofluid-based early detection tests will be used in conjunction Against Colon Cancer risk model is another tool that is available
with or in place of more standard imaging screening tests. online and is similar to the Freedman/NCI model in terms of the
variables it incorporates. However, it is unclear if it has been evaluated
COLORECTAL CANCER for publication in a peer-reviewed journal.

Despite being largely preventable through lifestyle modifications, an Screening


estimated 140,250 new cases of CRC will be diagnosed in the United
States during 2018, and an estimated 50,630 lives will be lost to The biology of CRC makes it an ideal candidate for early detection
CRC.62 Modifiable risk factors for CRC are obesity, smoking, alcohol, and preventive interventions. The colon is a relatively accessible organ,
and red or processed meat consumption. The World Cancer Research and the vast majority of CRCs develop from adenomas, a precancerous
Fund/American Institute for Cancer Research (WCRF/AICR) has stage that can last as long as 20 to 30 years, providing ample time
estimated that 47% of CRC cases, or 63,652 cases, could be prevented for its detection and removal before it evolves into cancer.
Screening and Early Detection  •  CHAPTER 23 381

The USPSTF currently recommends that average-risk individuals abdominal pain, myocardial infarction, angina, arrhythmias, congestive
be screened for CRC beginning at age 50 and continuing until age heart failure, respiratory arrest, syncope, hypotension, or shock) in a
75. For average-risk adults between 76 and 85 years of age, the decision population of 1000 screened individuals to be 10 to 11 with annual
to screen should be an individual one, considering a patient’s past FIT and 9 to 10 with FIT-DNA at 3-year intervals.70,71
screening history and overall health. The USPSTF does not recommend Direct visualization tests offer not only the ability to detect CRC
a particular screening test. Multiple CRC screening examinations exist, early, but also to prevent it through detection and subsequent removal
although they differ with respect to the level of evidence supporting of precancerous lesions. These tests include flexible sigmoidoscopy
their use, their test performance to detect cancer and adenomas, and (FS), colonoscopy, and computed tomography (CT) colonography
their risk of harms. There has been no assessment of the relative (CTC). In the case of CTC, follow-up endoscopy is required for
benefits and harms among the tests, because comparative studies are removal of polyps detected during the screen.
currently limited in their study design and power to detect cancers, Meta-analysis of four RCTs of FS in over 450,000 individuals
any mortality benefits, and associated harms.67 No screening test has demonstrated a 27% reduction (incidence rate ratio [IRR], 0.73; 95%
been shown to reduce all-cause mortality. CI, 0.66–0.82) in CRC mortality at 11 to 12 years of follow-up (find-
Screening options include stool tests, direct visualization tests, and ings limited to distal CRC).67 A single RCT from Norway demonstrated
a US Food and Drug Administration (FDA)–approved blood test (Epi a 38% reduction in CRC mortality when FS was combined with
proColon). However, because the blood test has very limited data on FIT.72 Although use of FS in the United States is uncommon,73 the
its performance, and because the data that are available suggest that 2016 USPSTF guideline recommends a 5-year screening interval for
its ability to detect CRC is worse than that of other noninvasive tests, FS alone and a 10-year interval if combined with annual FIT testing.
this section will discuss only the stool and direct visualization tests. Despite a lack of RCT evidence supporting the use of colonoscopy,
Guaiac-based fecal occult blood tests (gFOBTs), fecal immuno- it remains the gold standard of screening because of its high sensitivity
histochemical tests (FITs), and a FIT combined wit stool DNA testing and specificity for the identification of polyps and CRC and it ability
(Cologuard) are the three options for stool-based testing. Because to concomitantly diagnose and reduce risks associated with biopsied
stool-based tests do not reliably predict adenomas, these tests are early precancerous lesions. Observational evidence supporting the use of
detection tests and cannot prevent CRC from occurring. The gFOBTs colonoscopy comes from a large prospective analysis of the Nurses’
have the strongest direct evidence supporting their use as a CRC Health Study and the Health Professionals Follow-Up Study, which
screening modality, with biennial screening reducing CRC mortality demonstrated a 68% (hazard ratio [HR], 0.32 [95% CI, 0.24–0.45])
by 9% to 22% and annual screening reducing CRC mortality by 32% reduction in CRC mortality in those who self-reported screening
after 11 to 30 years of follow-up compared with no screening.67 The colonoscopy compared with those who never underwent screening
gFOBTs are noninvasive and inexpensive, although they can have a endoscopy.74 In this study, statistically significant reductions were seen
high false-positive rate and patient compliance is often low because for both distal and proximal CRC, although the mortality reduction was
of the test’s dietary restrictions. greater in the distal colon. There are currently a number of RCTs under-
FITs are rapidly replacing gFOBTs because they do not have dietary way for colonoscopy. At least two of them are comparing colonoscopy
restrictions and because they offer at least equal and likely better and FIT (NCT01239082 and NCT00906997); the Nordic-European
detection of CRC. Different FITs use different assay methods, so Initiative on Colorectal Cancer (NordICC) is comparing colonoscopy
FIT sensitivity has varied widely across studies. In the largest studies and usual care (NCT00883792)75; and one trial in Sweden is comparing
of the USPSTF evidence review for its 2016 recommendation, the colonoscopy with FIT or no screening (NCT02078804).
sensitivity was 73.8% (95% CI, 62.3–83.3) for the quantitative OC Drawbacks of lower endoscopy include its invasiveness, risk of
FIT-CHEK and 78.6% (95% CI, 61.0–90.5) for the qualitative OC- serious complications, expense, and operator dependence. The estimated
Light, based on single stool samples.67 Sensitivity could be increased number of complications from the aforementioned CISNET modeling
by using three stool samples or by lowering the assay cutoff value; study is 14 to 15 with colonoscopy at 10-year intervals and 9 to 12
however, specificity decreased with increasing sensitivity.67 Data are with FS at 5-year intervals.70
lacking regarding the impact of FITs on CRC mortality. Finally, FITs As with colonoscopy, there is currently no direct evidence establish-
are generally inexpensive, with a Centers for Medicare and Medicaid ing the effectiveness of CTC in reducing CRC incidence and mortality.
Services (CMS) reimbursement of $23 and a mean commercial However, a number of studies have examined the diagnostic accuracy
reimbursement of $21.68 of CTCs and are summarized in the 2016 USPSTF evidence review.67
The newest stool-based screen combines a FIT with DNA marker In studies in which bowel preparation was performed before the CTC,
analysis. Sensitivity of one-time FIT-DNA testing for CRC was shown per-person sensitivity and specificity to detect adenomas 10 mm or
to be 92.3% in one large study, significantly better than the sensitivity larger ranged from 66.7% to 93.5% and 86.0% to 97.9%, respectively.
of one-time FIT alone (73.8%); yet, specificity was lower (86.6% For detection of adenomas 6 mm or larger, sensitivity and specificity
versus 94.9).69 In other studies, the sensitivity of FIT was superior were 72.7% to 98.0% and 79.6% to 93.1%, respectively. The sensitivity
when multiple samples or lower assay cutoff values were used; and to detect advanced adenomas ranged from 87.5% to 100%. Limited
specificity for the FIT-DNA test is lower than for all FIT assays, data suggest that CTC without bowel preparation results in lower
resulting in this test having the highest false-positive rate among FIT sensitivity to detect adenomas 10 mm or larger and 6 mm or larger
screens.67 Data regarding a possible CRC mortality benefit for the in addition to advanced adenomas.
FIT-DNA test are lacking. Finally, this is the most expensive stool Serious harms of CTC in asymptomatic individuals appear to be
test, with a CMS reimbursement rate of $493.67 uncommon. The risk of perforation is less than 2 per 100,000 examina-
There are few adverse outcomes associated with stool-based tests tions. Data are lacking to estimate any serious adverse events from
directly, aside from the risk of missed cancers. However, serious adverse follow-up colonoscopy after CTC. Exposure to radiation is perhaps
events can occur during follow-up colonoscopy after positive stool test the most important potential harm from CTC; however, the evidence
results, and some data suggest that the rate of perforations in these to date suggests that the benefits of CTC screening every 5 years far
colonoscopies may be higher than for those performed in average-risk outweigh the potential radiation risks.76 Finally, although this test is
screening populations. The pooled estimate from the USPSTF evidence relatively noninvasive, it can result in the identification of extracolonic
review was 8 (95% CI, 2–32) perforations per 10,000 diagnostic findings that may or may not be clinically important. Estimates are
colonoscopies.67 A modeling study from the Cancer Intervention and that extracolonic findings occur in 41% to 69% of CTC procedures,
Surveillance Modeling Network (CISNET) Colorectal Cancer Working but only 5% to 37% of CTC procedures have extracolonic findings
Group estimated the number of complications (defined as perforations, that require actual diagnostic follow-up, and an even smaller percentage
gastrointestinal bleeding, nausea and vomiting, ileus, dehydration, result in findings that require any definitive treatment.67
382 Part I: Science and Clinical Oncology

CERVICAL CANCER cervical cancer related to DES exposure in utero has been shown to
persist in DES daughters above the age of 40.86
Cervical cancer provides the “perfect” paradigm for cancer prevention,
having a vaccine to reduce the incidence of infection from the sole Cervical Cancer Screening
causative agent, human papillomavirus (HPV), and screening tests to
identify women infected with HPV or cytologic findings that indicate The Pap test is considered to be one of medicine’s most successful
precancerous conditions that can be successfully managed to prevent cancer screening tests. Although there are no RCTs that have evalu-
the development of invasive disease. The incidence and mortality of ated the efficacy of the Pap test, there is overwhelming observational
cervical cancer in the United States has decreased by over 75% since evidence that cervical cancer incidence and mortality have decreased
widespread screening with the Papanicolaou (Pap) test and treatment since Pap testing was integrated into routine clinical practice in the
of precancerous lesions began decades ago.77 United States.77
Infection with high-risk HPV is a necessary, although not sufficient, Cervical cancer is characterized by a long sojourn time, with precur-
cause of cervical cancer. Most HPV is transient and poses little risk sor lesions identifiable long before the development of invasive disease.
of persistent infection.78 The natural history of cervical cancer involves Cytologic screening for these lesions (e.g., cervical intraepithelial
(1) HPV infection; (2) persistent HPV infection; (3) development of neoplasia [CIN]) with the Pap test is effective in reducing both the
a precancerous cervical lesion; and (4) progression to invasive cervical incidence and mortality from cervical cancer because these premalignant
cancer.79 Opportunities for interventions exist throughout the con- lesions are generally amenable to interventions that prevent the progres-
tinuum, including education about HPV transmission, HPV vaccina- sion to invasive disease.77 HPV testing shows whether a person has a
tion, screening with HPV and Pap tests, and treatment of precancerous current HPV infection. It does not provide information on previous
or cancerous lesions of the cervix. Interventions at steps 1 to 3 provide HPV infections. Genotype testing is now available for HPV types 16
the greatest opportunity to reduce the incidence and mortality from and 18 and can be used to aid in the clinical management of precursor
cervical cancer. lesions, but is not currently used in cervical cancer screening.
HPV is the most common sexually transmitted infection in the Studies of HPV cervical cancer screening have found that HPV
United States, with over 80% of sexually active individuals acquiring testing alone was more sensitive but less specific than cytologic evalu-
an HPV infection by age 45.80 A number of other cancers have been ation alone. For CIN3+ outcomes, sensitivity ranged from 86% to
associated with HPV infection, including oropharyngeal (largely in 97% for HPV testing versus 46% to 50% for cytologic evaluation at
men), vaginal, vulvar, penile, and anal (mostly in men who have sex a colposcopy referral threshold of “atypical squamous cells of unde-
with men). Although most HPV is transient in nature, factors that termined significance” (ASCUS). For CIN2+ outcomes, sensitivity
determine which HPV infections will persist and progress are not ranged from 63% to 98% for HPV testing versus 38% to 65% for
completely understood. The HPV genotype appears to be the most cytology. However, specificity for CIN2+ and CIN3+ was consistently
important determinant. HPV-16 has the highest carcinogenic potential, 3% to 5% lower for HPV testing than for cytology.87
causing approximately 55% to 60% of all cases of cervical cancer A 2012 study identified that a negative HPV test result provided
worldwide. HPV-18 is the next most carcinogenic genotype, accounting greater reassurance against CIN3+ over an 18-year follow-up than
for 10% to 15% of cervical cancers. Approximately 12 other genotypes did a normal Pap result.88 Although baseline Pap and HPV tests
are associated with the remainder of cases of cervical cancer.81,82 equally predicted who would develop CIN3+ within the first 2 years
The risk of HPV infection is greatest among teens and women in of follow-up, only HPV testing predicted who would develop CIN3+
their early 20s and decreases with increasing age. Most infections with 10 to 18 years later. These data support extending the screening interval
high-risk HPV do not result in persistent infections or cancer and in women who have negative cotesting results.
are very likely to regress among women with both normal and abnormal
cytology results. However, persistence of HPV infection appears to Current Cervical Cancer Screening: Cytologic
increase with age, with HPV infections detected in women older than Assessment With or Without Human
30 years being more likely to reflect a persistent infection.83 Identifying Papillomavirus Testing
women with a persistent HPV infection is one of the best ways to
distinguish women at increased risk of developing cervical dysplasia Updated screening guidelines were released in 2012 jointly by the
who require closer monitoring. ACS, the American Society for Colposcopy and Cervical Pathology
With the recognition of HPV as the primary etiologic factor in (ASCCP) and the American Society for Cervical Pathology (ASCP)
cervical cancer, variables previously identified as risk factors are now (Table 23.5) and separately by the USPSTF.89,90 These guidelines apply
better classified as cofactors that either increase the risk of HPV infection to women at average risk of cervical cancer who have a cervix.
or increase the likelihood that an infection will be persistent, thus Increasing evidence and an improved understanding of the natural
increasing the risk of cervical neoplasia. These include age at first history of cervical cancer led to the recognition that previous recom-
coitus, number of sexual partners for a woman or her partner, history mendations for annual screening were excessive and resulted in an
of sexually transmitted disease including HPV infection, absence of increased rate of false positives. Annual screening leads to a very small
HPV vaccination, immunosuppression (human immunodeficiency increment in cancers prevented, at the cost of a very large excess of
virus [HIV]–positive individuals, transplant recipients), younger age unnecessary procedures and treatments due to the high prevalence of
at first pregnancy, high parity, long-term oral contraceptive (OC) use, transient, benign HPV infections and associated lesions, most of which
poor nutritional status, tobacco use, race, or ethnicity [Hispanic, will regress within a year or two. Of those that do not regress, on
especially if not English speaking, or African American women), average, they are many years from causing cancer. Women at average
socioeconomic status, and infrequent Pap smear screening.83 risk at any age should not be screened annually by any screening
Diethylstilbestrol (DES) exposure in utero is largely a historical method. Recommended screening intervals for women at average risk
risk factor for the development of cervical cancer. DES is a synthetic are based on age and clinical history.89
form of the hormone estrogen that was prescribed to pregnant women Women ages 21 through 30 should be screened every 3 years with
between 1940 and 1971 to prevent miscarriage and premature labor.84 the Pap test. HPV testing is not recommended in women under age
It has been well defined that the daughters of women who took DES 30 because the risk of HPV infection is high, but the likelihood of a
while pregnant (so-called “DES daughters”) have a 40-fold increased persistent infection is extremely low. Cotesting every 5 years with
risk of developing clear cell adenocarcinoma of the vagina and cervix cytologic evaluation and HPV testing provides benefits similar to a
compared with unexposed women. However, this type of cancer remains Pap test alone done every 3 years for women 30 to 65 years of age.
rare, with only 1 in 1000 DES daughters developing it.85 Risk of Therefore, in women age 30 to 65 years who want to lengthen the
Screening and Early Detection  •  CHAPTER 23 383

Table 23.5  American Cancer Society, American Emerging Tests for Cervical Cancer Screening:
Society for Colposcopy and Cervical Primary Human Papillomavirus Screening
Pathology, and American Society for As previously noted, studies have found that HPV testing alone was
Clinical Pathology Cervical Cancer more sensitive than cytologic evaluation alone. In 2014, the FDA
Screening Recommendations for approved a currently marketed HPV test to include the additional
Women at Average Riska indication of primary cervical cancer screening.
Although HPV testing alone is more sensitive than cytologic
Age Screening Recommendationb evaluation alone, it is also less specific. Strategies that maximize detection
Aged <21 yr No screeningc of CIN3+ by immediate referral to colposcopy with follow-up testing
Aged 21–29 yr Cytology alone every 3 yrc
and triage of women at intermediate risk of CIN maximizes the benefits
of cervical cancer screening while decreasing the potential harm.91
Aged 30–65 yr Human papillomavirus (HPV) and cytology The Addressing the Need for Advanced HPV Diagnostics (ATHENA
“cotesting” every 5 yr (preferred)
HPV) Study evaluated the role of primary HPV screening in women
OR
Cytology alone every 3 yr (acceptable)
25 years of age and older and validated an effective triage algorithm.
In this trial, HPV-positive specimens underwent genotyping for HPV-16
Age >65 yr No screening following adequate negative and HPV-18. If a specimen was positive for HPV-16 or HPV-18,
prior screening colposcopy was performed. If a specimen was negative for HPV-16
a
or HPV-18, cytologic evaluation was performed on the specimen. If
The following are not considered to be at average risk: women with a history of
CIN2+ or cervical cancer in the past 20 years, diethylstilbestrol (DES) daughters, and
the cytologic evaluation results were abnormal, colposcopy was per-
those with human immunodeficiency virus (HIV) infection or immunosuppression formed. If both HPV genotyping and cytologic findings were normal,
due to other factors (e.g., transplant patients). repeat cotesting was performed in 1 year. The trial concluded that
b

c
Average-risk women should not be screened annually at any age by any method. primary HPV screening in women 25 years of age or older is as
HPV testing should not be used for screening in this age group. effective as a screening strategy that uses cytologic evaluation in those
d
If the hysterectomy was supracervical and the cervix is intact, screening would
be the same as in average-risk women.
25 to 29 years old and cotesting in those 30 years of age or older. In
CIN, Cervical intraepithelial neoplasia. addition, this paradigm requires fewer screening tests.92
Modified from Saslow D, Solomon D, Lawson HW, et al. American Cancer Society, In 2015, the ASCCP and the Society of Gynecologic Oncology
American Society for Colposcopy and Cervical Pathology, and American Society for (SGO) provided interim guidance for the use of the FDA-approved
Clinical Pathology screening guidelines for the prevention and early detection of HPV test for primary cervical cancer screening. They concluded that,
cervical cancer. CA Cancer J Clin. 2012;62(3):147–172. because of its equivalent or superior effectiveness in women 25 years
of age or older, the FDA-approved primary HPV screening test can
screening interval, cotesting every 5 years with Pap and HPV is recom- be considered an alternative to current cytology-based cervical cancer
mended. ACS, ASCCP, and ASCP preferred cotesting over the Pap screening methods.93 However, primary HPV testing is not currently
test alone, whereas the USPSTF deemed both cotesting and Pap test considered a standard of care.
alone as acceptable.89,90 Prevention of cervical cancer through education about HPV
Certain low-risk groups should no longer be screened. These include transmission, HPV vaccination, and screening with cytologic evaluation
women under age 21, low-risk women over age 65, and women who with or without HPV testing has the potential to eliminate invasive
have undergone a hysterectomy.89,90 Evidence has shown that screening cervical cancer as a disease of concern for women. Primary HPV
women age 21 and younger does not reduce cervical cancer incidence screening is emerging as a new screening modality.
and mortality but is associated with significant harms. Treatment of
CIN in women younger than age 21 has been shown to increase the LUNG CANCER
risk of adverse pregnancy outcomes, including cervical incompetence
and preterm delivery.89,90 Women age 65 and older who have had the Lung cancer is the most common cause of cancer-related death
recommended screening in the past 10 years and are considered to worldwide and the second most diagnosed cancer in the United States
be at low risk of cervical cancer do not need to continue screening for both men and women.94 Estimated figures for 2016 in the United
beyond age 65. Women who are not low risk include those with States exceed 224,000 new cases and 158,000 deaths as reported by
immunosuppression (e.g., HIV or transplant patients), DES daughters, the ACS. In China alone, estimated figures for 2015 are 733,000 new
and those with a prior history of a high-grade precancerous lesion of cases and 610,000 deaths.95 Lung cancer incidence and death rates
the cervix or cervical cancer within the past 20 years. It is important in the United States have been declining slightly for men and women,
to note that older women who have not had recent screening are a with the magnitude of the decrease varying by race and ethnicity.94
population at risk for the development of cervical cancer. Although
cervical cancer incidence rates are on the decline, 50% of women Risk Factors
diagnosed with cervical cancer have not been screened 3 to 5 years
prior to diagnosis.83,89,90 Women who have had hysterectomy do not Smoking continues to represent the major risk factor for lung cancer,
need to have cervical cancer screening unless the hysterectomy was with a dose-response relationship between pack-years of smoking and
done for the management of CIN2+ or cervical cancer, in which case risk.96 Exposure to environmental tobacco smoke among nonsmokers
they should follow the recommended surveillance guidelines. When a increases lung cancer risk by approximately 20%. Additional risk
supracervical hysterectomy is performed (i.e., leaving the cervix intact), factors associated with smoking include age at smoking onset, type
screening recommendations should be based on cervical cancer risk.89,90 of tobacco product smoked, and depth of inhalation. Risks due to
At this time, women who receive the HPV vaccination should marijuana, hookah use, and e-cigarettes remain to be determined.97
continue to follow current cervical cancer screening recommendations, Other risk factors are related to exposure to other toxins and damaging
because HPV vaccination does not protect against all types of oncogenic agents including asbestos, radon, air pollution, and ionizing radiation.98
HPV. In addition, the HPV vaccination rate is far from optimal at Host factors associated with lung cancer risk include history of chronic
this time, and immunization registries do not identify who has received obstructive pulmonary disease or infections and a family history of
the recommended series of vaccinations. Although HPV vaccination lung cancer.99 Substantial effort has focused on susceptibility genes
is an important step toward cervical cancer prevention, it does not for lung cancer, which have yielded significant associations with
remove the need for routine cervical cancer screening. chromosomal regions including 15q25 and 5p15.100
384 Part I: Science and Clinical Oncology

Risk Modeling in lung cancer mortality and a 6.7% reduction in all-cause mortality.
A striking finding from the NLST was the high rate of false positives,
At present, there is not a currently recommended risk prediction which was 27% at baseline and 28% at 1-year follow-up. After the
model for lung cancer for general clinical use, though several models completion of the NLST, the International Early Lung Cancer Action
have been proposed with varied complexity.101–103 Risk estimates from Program retrospectively analyzed the outcomes of more than 21,000
predictive models have utility in determining who needs screening prospectively enrolled patients who underwent lung cancer screen-
and in counseling individuals at risk, particularly in encouraging ing.116 An increase in size threshold for nodule diameter resulted in
smoking cessation.104 Application of a model to ever-smokers in the an increase in cancer diagnosis rates. An increase in the size threshold
Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial (PLCO) from 5 mm to 6 mm could reduce the workup by 36%, and up to 75%
cohort intervention arm led to improved sensitivity (80% versus 71%) for 9 mm.
and specificity (66% versus 63%) compared with NLST screening Concerns about the high percentage of false positives and potential
criteria alone.105 health hazards resulting from exposure to radiation with repeat LDCT
The addition of pulmonary function has been shown to improve scans provide justification for the application of more informative
risk prediction for lung cancer.106 A recently reported lung cancer risk prediction models to identify individuals at high risk who would
risk-prediction model that incorporated lung function was based on benefit from LDCT screening for early detection, to minimize false
the UK Biobank prospective cohort study.107 Flexible parametric survival positives. An alternative to screening with LDCT is through further
models were used to estimate the 2-year probability of lung cancer, reduction in radiation dose with ultra-low-dose CT without loss of
accounting for the competing risk of death. Models included sex, image quality.117 A Japanese multicenter study compared performance
variables related to smoking history and nicotine addiction, medical of ultra-low-dose CT with LDCT for the detection of lung nodules.118
history, family history of lung cancer, and lung function measured A total of 161 nodules and 60 ground glass opacities were identified,
with spirometry (forced expiratory volume in 1 second [FEV1]). A with comparable performance between the two modalities.
risk prediction model that included lung function had strong predictive Guidelines for the management of patients with nodules detected
ability and had better discrimination than standard lung cancer through CT screening are also evolving. The NCCN provides guidelines
screening eligibility criteria. A potentially important improvement in that avoid invasive follow-up for small nodules with low probably of
risk prediction models will likely result from incorporation of molecular malignancy. The guidelines incorporate recommendations based on
markers. Cotinine, a circulating metabolite of nicotine, may improve the NLST findings, the International Early Lung and Cardiac Action
model discrimination among current smokers. Cotinine has been Program protocols, and the Fleischner Society guidelines. A reporting
found to be associated with lung cancer risk108 and could be used as system has been suggested that classifies nodules observed with screening
a more sensitive assessment of smoking intensity than self-reporting CT based on the risk of malignancy, which is linked to suggested
and thus may improve risk modeling further. A protein, prosurfactant follow-up.119 The goal is to improve communication with patients
protein B (pro-SFTPB), which is a target of the oncogene NKX2-1 and clinicians. Subjects with nodules smaller than 5 mm or with
implicated in lung tumor development, has been shown to be a risk perifissural or long-term stable nodules would require no further workup
marker for lung cancer.109 Using pro-SFTPB as an anchor marker, before the next routine screening CT scan. Subjects with larger nodules
the addition of the metabolite diacetylspermine resulted in a statistically would require CT follow-up at an interval dependent on nodule size,
significant increase in predictive power of the model.110 Comprehensive varying from small (5–9 mm) to large (≥10 mm). Other nodules
models that incorporate demographic data and smoking information, would be considered to have a high likelihood of malignancy based
lung function, and molecular risk markers thus have great potential on screening CT features.
to improve lung cancer risk stratification. Potential improvements in
risk prediction through lab-based tests have to be weighed against PROSTATE CANCER
increased costs and practicality.
Prostate cancer is the most commonly occurring cancer among men
Screening (excluding nonmelanoma skin cancers [NMSCs]) and was the second
most common cause of cancer death among men in 2014.62 Compared
A major cause of high mortality in lung cancer is that most cases are with non-Hispanic whites, African Americans experience higher
diagnosed at advanced stages of the disease, when current therapeutic incidence (203.5 versus 121.9 per 100,000 men) and mortality (44.1
regimens are relatively ineffective.111 There has been long-standing versus 19.1 per 100,000 men). The prostate cancer incidence rate has
interest in detecting cancer at an early stage with imaging modalities. declined sharply, 10% annually from 2010 to 2013, owing to reduced
The concept of screening for lung cancer originated in the 1950s with use of the PSA test after the 2012 USPSTF recommendation against
the Philadelphia Neoplasm Research Project, which performed periodic routine screening with the test.62 The prostate cancer mortality rate
screening on some 6000 males with fluoroscopy, but with no effect on has dropped 51% from 1993 to 2014.62 Local- or regional-stage
outcome.112 In the following decades, several randomized trials were diagnosis is now typical for over 90% of prostate cases, resulting in
conducted to assess the benefits of chest radiography, with similar 5-year survival rates just under 100%, having improved 30% over
conclusions with respect to outcome. These studies reported finding the last several decades. However, 5-year survival is just 29% for those
more early-stage cancers in the screened group, and more surgical with distant metastases.120 The etiology of prostate cancer is incompletely
procedures were performed, but with no impact on cancer-related understood but involves both modifiable risk factors (diet, tobacco
mortality.113 Interest in screening picked up again with the introduc- use, obesity) and nonmodifiable risk factors (age, race or ethnicity,
tion of CT scans and with reports of better detection of early-stage genetics).121 Prostate cancer progression is typically slow, and clinically
cancers with CT scans compared with chest radiographs.114 The lack significant progression may not occur during a man’s lifetime.121
of control groups in the early trials made it difficult to determine
whether screening could decrease lung cancer mortality. The National Risk Modeling and Assessment
Lung Screening Trial (NLST), an RCT comparing CT screening with
chest radiography and with lung cancer mortality as the end point Risk prediction models offer the possibility of tailoring PSA screening
followed.115 The NLST enrolled 53,000 individuals aged 55 to 74 to those who are most likely to benefit from it, thereby reducing
years with a 30–pack-year smoking history. Participants were randomly unnecessary biopsies and maximizing the number of clinically significant
assigned to chest radiography or low-dose computed tomography prostate cancers detected. Hundreds of risk models have been published,
(LDCT) and screened at baseline with two annual follow-up scans. although just a handful have been validated in various external
Follow-up lasted up to 7 years. The LDCT group had a 20% reduction populations.122
Screening and Early Detection  •  CHAPTER 23 385

One such model is the Prostate Cancer Risk Calculator, developed situations.127 More research is needed to determine if these types of
in 2006 with data from the Prostate Cancer Prevention Trial (PCPT) adjunctive tests improve the balance of benefits and harms of PSA-based
and recently updated in 2014 with an expanded set of PCPT biopsy prostate cancer screening.
findings and now reporting on three outcomes (negative biopsy result,
low-grade and high-grade cancer) rather than the initial two (cancer Screening
versus no cancer).123 This 2.0 model was externally validated in 10
international biopsy cohorts and a reference biopsy set from NCI’s The ability of the PSA test to reduce prostate cancer–specific mortality
Early Detection Research Network (EDRN). In these cohorts, the when used in a screening context has been examined in the PLCO
AUC ranged from 0.62 to 0.88, with a median of 0.75, for the and ERSPC. Results of the US-based PLCO128,129 did not identify
detection of clinically significant prostate cancer.123 In another nine any significant benefit from screening on prostate cancer mortality at
studies, the AUC ranged from 0.51 to 0.79, with a median AUC of either 7 or 13 years of follow-up.129,130 However, results of this trial
0.69.121 The AUC of PSA alone in these studies ranged from 0.56 to have been called into question, given the extremely high proportion
0.71 with a median of 0.65.121 The PCPT 2.0 includes age and race, of the control arm that reported having at least one PSA test during
family history, PSA level, digital rectal examination (DRE) result, and or before the trial.131 By contrast, the ERSPC,132 a collaborative effort
prior biopsy findings, and also considers percent-free PSA, if available. involving eight European countries (Belgium, Finland, France, Italy,
A systematic evidence review conducted for the USPSTF’s 2017 update the Netherlands, Spain, Sweden, and Switzerland), identified a sig-
of its prostate cancer screening recommendation states that although nificant 20% reduction in prostate cancer mortality as a result of
the calculator improves discrimination between men with and without PSA-based screening. However, these results demonstrated a high risk
high-grade prostate cancer, both its predictive accuracy and calibration of overdiagnosis of prostate cancer in individuals lacking clinical
vary substantially across samples.121 The model is freely available online symptoms. The 12- to 14-year follow-up results from the Swedish
at http://myprostatecancerrisk.com/. and Netherlands sites of the ERSPC trial found enhanced benefit of
A second commonly used risk prediction model is derived from PSA screening, with prostate cancer mortality reduced by almost half
the Rotterdam data of the European Randomized Study of Screen- in Sweden and by 20% in the Netherlands, although the reduction
ing for Prostate Cancer (ERSPC).122,124 This tool includes PSA level, was as great as 32% in those ages 55 to 69.133,134 Conversely, the
DRE result, prior biopsy, and prostate volume. In the development 12- to 15-year follow-up results from the Finland and Spanish sites
of the model, prostate volume was based on transrectal ultrasound of the ERSPC study failed to demonstrate significant effects of PSA-
(TRUS). But because TRUS is invasive and generally performed during based screening on prostate cancer mortality.135,136 Limitations of the
a biopsy, the model allows for an estimate of prostate volume based ERSPC trial include substantial variation in methods across sites as
on DRE to be used in place of TRUS. The model with the DRE well as unexplained postrandomization treatment differences, which
substitution was validated in 322 men referred for biopsy during may partly explain the benefit of screening observed in this trial.121
later screening rounds of the ERSPC, wherein its AUC was 0.78 for Neither the PLCO nor the ERSPC documented evidence for a reduction
the detection of high-grade cancer.125 The AUC of the PSA test in in overall mortality; and neither trial provides insight into any potential
these men was 0.68.125 In four external validation studies, the AUC benefits of screening that may be specific to men with a family history
of the ERSPC model ranged from 0.69 to 0.78, with the AUC of of prostate cancer or who are African American.
PSA alone ranging from 0.61 to 0.65.121 This model also allows the The previous USPSTF prostate cancer screening guideline (2012)
Prostate Health Index (PHI) to be included, if available. The PHI recommended against (grade D) routine use of the PSA test in average-
slightly increases the calculator’s predictive capability.126 This tool is also risk populations because it concluded that the benefits of PSA testing
freely available online at http://www.prostatecancer-riskcalculator.com/ did not outweigh the potential risks. However, the Task Force is now
seven-prostate-cancer-risk-calculators. in the process of issuing a new prostate cancer screening guideline
In a comparison of the PCPT and ERSPC models, Foley and and, as of this writing, it is recommending that clinicians inform men
colleagues demonstrated that the ERSPC significantly outperforms ages 55 to 69 of the potential benefits and harms of PSA testing and
PCPT 2.0 in the prediction of prostate cancer, with an AUC of 0.71 that the decision to screen should be an individual one (grade C).121
(versus 0.64), and an AUC of 0.74 (versus 0.69) for the prediction This change in recommendation is due, in part, to new evidence
of significant prostate cancer in 2001 men from six tertiary referral supporting the benefits of screening that comes from longer-term
centers in Ireland.126 A meta-analysis of six different risk prediction follow-up, and new reports from subsites, of the ERSPC trial. According
tools, including the PCPT 2.0 and ERSPC models, found that all to the latest evidence, PSA-based screening programs in men ages 55
risk models improved the predictive accuracy of PSA testing to detect to 69 may prevent up to one to two deaths from prostate cancer and
prostate cancer, with the exception of the PCPT 2.0 model.122 up to three metastatic prostate cancers per 1000 men screened over
Important to note, no RCTs have been performed to test the use 13 years.121 The change in recommendation is also driven by new
of a risk prediction model versus no model on outcomes such as data on the use of active surveillance, which may reduce the risk of
clinical decision making, biopsy accuracy, or long-term incidence of overtreatment. The rate of active surveillance in the United States
advanced prostate cancer.121 Few studies have addressed the implementa- among men diagnosed with low-risk prostate cancer has increased
tion of risk calculators into clinical practice and decision making. sharply, from 14.3% in 2009 to 40.4% in 2013. Results from the
Limitations to current prostate cancer risk models include variable British Prostate Testing for Cancer and Treatment (ProtecT) trial
calibration among populations and/or often poorly reported calibration demonstrate no significant difference in either prostate cancer or overall
statistics. mortality among men randomized to active surveillance, radical
In addition to risk models, serum and urine tests and multipara- prostatectomy, or radiotherapy after 10 years of follow-up.137 However,
metric MRI are also being used in an attempt to improve PSA screening, men randomized to active surveillance developed more metastases
although none of them have been evaluated in RCTs. For some men and experienced higher rates of disease progression than did men in
with mildly elevated PSA levels (3–10 ng/dL) at screening, current either the surgery or radiation groups.137 The very high 10-year survival
NCCN guidelines suggest consideration of free PSA, the 4Kscore, or rate (98%) across all three groups may have limited the trial’s power
the PHI to assist in decision making about whether to proceed to to detect differences in mortality among the groups. Longer follow-up
biopsy or whether to repeat biopsy if the result of the first biopsy is is needed to clarify these results.
negative.121,127 The PCA3 (noncoding mRNA overexpressed in prostate The USPSTF also assessed harms related to screening and, because
cancer tissue) test is FDA approved for use in men with a prior negative the benefit of screening can be achieved only through treatment, it
biopsy result to assist in decision making regarding a repeat biopsy, also assessed harms related to the various treatment modalities for
and consideration of its use is recommended by the NCCN in such prostate cancer (i.e., radical prostatectomy, radiotherapy, and active
386 Part I: Science and Clinical Oncology

Table 23.6  Benefits and Harms of Prostate Cancer Screening per 1000 Men Screened
Trial PLCO ERSPC (Core Age Group) Goteborg, Sweden Notes
Screening population Men 55–74 yr; Men 55–69 yr; screened Men 50–64 yr; invited Data for PLCO and Goteborg
screened annually for every 4 yr (biennially in biennially up to 70 yr; PSA derived from reports with 13.0
6 yr; PSA threshold Sweden); PSA threshold threshold 3.0–3.5 ng/mL and 14.0 yr of median follow-up
for biopsy, 4.0 ng/mL most commonly 3.0 ng/mL before 2005 and 2.5 ng/mL as more recent reports with 14.0
from 2005 to 2008 and 18.0 yr median follow-up lack
Median follow-up, yr 13.0 13.0 14.0 some required data
Outcome Number of men affected
Men invited to screening 1000 1000 1000
≥1 positive screen 282 274 248 PLCO, ERSPC, Goteborg trial
≥1 biopsy 130 244 231 reports
Additional men 1.7 3.2 3.0 ProbE cohort study (1.3%
hospitalized for biopsy hospitalized due to biopsy
complications complications)
Additional prostate 11.3 34.8 42.0 Absolute cumulative incidence
cancer diagnosed differences and numbers needed
because of screeninga to invite in trial reports
Additional men with ED 1.8 6.8 8.3 Men with cancer allocated to
due to treatment primary treatments as in trial
Additional men with UI 0.8 2.4 3.3 screening arms. For treatment
after RP harms, number needed to harm is
from EPC pooled meta-analyses
Men treated with radical NC 23.9 25.8 Number computed by
prostatectomy or subtracting number of men with
radiotherapy without prostate cancer death or
benefits metastasis averted from number
of men actively treated
Metastatic cases averted NR 2.9 3.5 ERSPC core age group absolute
risk reduction in metastatic
disease cumulative incidence
(and number need to diagnose)
Prostate cancer deaths 0 1.3 3.4 Absolute risk reduction in
averted prostate cancer deaths (and
numbers needed to diagnose)

a
Based on trial data, 99.5, 68.3, and 72.1 would be diagnosed with prostate cancer among 1000 men not invited to screening in the PLCO, ERSPC, and Goteborg, Sweden
settings, respectively.
ED, Erectile dysfunction; EPC, evidence-based practice center; ERSPC, European Randomized Study of Screening for Prostate Cancer; NC, not calculable; NR, not reported;
PLCO, Prostate, Lung, Colorectal and Ovarian Cancer Screening Trial; ProbE, Prostate Biopsy Effects cohort study; PSA, prostate-specific antigen; UI, urinary incontinence.
Adapted with permission of the U.S Agency for Healthcare Research and Quality.

surveillance). Harms from screening include false-positive results and an interval of every 2 or 4 years, rather than annually, was found to
the potential pain, bleeding, and infection from biopsy. The conclusion offer a reasonable tradeoff between a reduction in overdiagnosis and
of the Task Force regarding harms of screening is that they are “at a small reduction in mortality benefit.141 Evidence was deemed insuf-
least small.”138 Harms related to treatment include urinary and fecal ficient to support the use of alternative PSA screening modalities,
incontinence, erectile dysfunction or long-term sexual impotence, such as single- and adjusted-threshold testing, PSA velocity, and
and, in the case of radical prostatectomy, serious surgical complications double-timing.139
and possible death. The conclusion of the Task Force regarding the Given the new data on the potential benefits of screening (i.e.,
harms of overdiagnosis and treatment is that they are “at least moder- reduced risk of dying from prostate cancer and reduced risk of metastatic
ate.”139 Table 23.6 presents a summary of the benefits and harms of prostate cancer), the USPSTF has stated that the benefits and harms
PSA screening and subsequent treatment, as reported in the USPSTF of PSA screening are more closely balanced than previously estimated.
evidence review and draft recommendation. Therefore it is the intention of the Task Force that, after discussion
Finally, the USPSTF assessed whether changing the screening of these benefits and harms with a health care provider, each man’s
interval or lowering the threshold of PSA testing for diagnostic biopsy particular values and preferences tip the balance in favor of either a
might increase the benefit of screening while simultaneously reducing net benefit or net harm.
harms. An optimal cutoff value for the PSA test that is associated Evidence does not currently support different recommendations
with both high specificity and sensitivity has not yet been established.140 for African American men or men with a family history of the disease,
RCT data suggest that while a greater mortality benefit may be derived although many clinicians would choose to screen these men earlier
from screening every other year, as opposed to longer intervals, and and/or more often. With regard to PSA screening in men 70 of age
from using a lower PSA threshold, harms are also substantially increased, or older, the USPSTF has concluded that there is moderate certainty
including overdiagnosis.139 Nevertheless, in a review of modeling studies that the potential benefits of screening do not outweigh the expected
Screening and Early Detection  •  CHAPTER 23 387

harms, and so it recommends against screening in this group (grade


D). The 2017 guideline brought the USPSTF into line with the recom- Table 23.7  Nongenetic and Genetic Risk Factors
mendations of most other physician and patient advocacy groups. for Pancreatic Cancer
Variable Risk Increase
PANCREATIC CANCER NONGENETIC RISK FACTORS
Pancreatic cancer is the fourth leading cause of cancer death in Western Chronic pancreatitis 13.3
countries, with an overall 5-year survival rate of around 7% and an New onset type 2 diabetes 7.9
average survival of 6 months.142 The median age at diagnosis is 71
Long-standing diabetes mellitus 2.0
years. The annual incidence and mortality rates closely mirror each
other. Pancreatic cancer is estimated to become the second most Obesity 2.0
common cause of cancer-related deaths by 2030.143 A classification Smoking 1.8
system based on gene expression signatures derived from either tumor Alcohol abuse 1.2
or stromal cells has been proposed that predicts patient outcome.144 Non-O blood group 1.3
A model for progression to pancreatic adenocarcinoma has been
established with occurrence of signature mutations in premalignant GENETIC SYNDROME AND ASSOCIATED GENES
lesions.145 There is a stepwise progression of pancreatic intraepithelial Familial pancreatic cancer (unknown gene)
neoplasia from low grade to high grade in types 1, 2, and 3 with One first-degree relative 9
accumulating genetic alterations and occurrence of KRAS mutations
Three first-degree relatives 32
in the vast majority of pancreatic intraepithelial neoplasia of all grades.146
Low-grade pancreatic intraepithelial lesions are frequently detected Familial adenomatous polyposis (APC) 4.5–6
in autopsies in nondiseased pancreas, whereas advanced lesions are Breast and ovarian cancer syndrome (BRCA1, BRCA2, 2–3.5
usually detected adjacent to adenocarcinoma.147 PALB2)
Intraductal papillary mucinous neoplasm (IPMN) is a heterogeneous Peutz-Jeghers syndrome (STK11/LKB1) 132
entity that represents another type of precursor lesion to pancreatic Hereditary pancreatitis (PRSS1, SPINK1) 69
cancer.148 The prevalence of IPMN lesions can be as high as 20% and Familial atypical multiple mole melanoma 47
increases with age.149,150 Risk stratification with regard to cancer can pancreatic carcinoma syndrome (P16INK4A/CDKN2A)
be accomplished according to high-risk characteristics.148,151 Serous
cystic neoplasm (SCN) represents another type of lesion for which Lynch syndrome (MLH1, MSH2, MSH6, PMS2, EPCAM) 8.6
our understanding of the natural history is rather limited. A retrospective Cystic fibrosis (CFTR) 3.5
follow-up study of patients with SCN over a 3-year period revealed Ataxia-telangiectasia (ATM) Unknown
clinically relevant symptoms in a very small proportion of patients
GENETIC POLYMORPHISMS
and a slow increase in size in fewer than half of the patients.152
ABO 1.3
Risk Factors and Inherited Syndromes NR5A2 1.3
TERT 1.2
Some 10% of all patients with pancreatic cancer have a positive family PDX1 1.2
history for pancreatic cancer or for related genetic syndromes that
justify screening.147 The genetic basis for familial aggregation has not BCAR1, CTRB1, CTRB2 1.5
been identified in a large proportion of cases.153 Genetic syndromes ZNRF3 1.2
include Lynch syndrome, FAP, hereditary breast-ovarian cancer syn- LINC00673 1.3
drome, hereditary pancreatitis, Peutz-Jeghers syndrome, and familial ETAA1 1.1
atypical multiple mole melanoma syndrome. In addition, a multitude TP63 1.1
of polymorphisms have been associated with a slightly increased risk
of pancreatic cancer (Table 23.7). SUGCT 1.1
Risk factors associated with pancreatic cancer development in the
general population include aging, tobacco use, heavy alcohol use,
diabetes, and obesity. Tobacco use is estimated to double the risk of
pancreatic cancer, with population estimates of attributable risk and precursor lesions and likelihood of progression among the latter.
estimated to be as high as 25%.154 Heavy alcohol use is associated As a result, at present, efforts in early detection are largely limited to
with a 22% increased risk of pancreatic cancer.155 There is also a individuals at high risk.
significant association between body mass index (BMI), type 1 dia- Screening recommendations of the International Cancer of the
betes,156 long-standing type 2 diabetes,157 and abnormal glucose toler- Pancreas Screening Consortium concern high-risk subjects—namely,
ance158 and pancreatic cancer risk. individuals from a familial pancreatic cancer kindred with at least two
affected FDRs; patients with Peutz-Jeghers syndrome; and p16 or
Screening BRCA1/2 mutation carriers.159,160 Screening is largely based on imaging,
notably MRI or endoscopic ultrasonography. A prospective observational
Although survival rates are dismal, a diagnosis of pancreatic cancer study was done involving 40 consecutive patients with a genetic risk
at a resectable stage carries a better prognosis, with a 5-year survival for developing pancreatic cancer who were referred to Karolinska
rate of 30%, substantiating the potential merits of early detection. University Hospital for MRI-based surveillance.161 During a median
The relatively long timeline for progression provides an opportunity follow-up of approximately 1 year, pancreatic lesions were detected
for preventive intervention and early detection. However, the low in 40% of the patients, of whom five underwent surgery—three for
prevalence of pancreatic cancer, varying from 9 per 100,000 in the pancreatic ductal adenocarcinoma and two for intraductal papillary
general population to 68 per 100,000 persons older than age 55 years, mucinous neoplasia.
and the common occurrence of early precursor lesions in the elderly Screening of individuals at high risk is recommended starting at
population create challenges for implementing early detection and 40 years of age or 10 years earlier than the age at which the youngest
for development of noninvasive tools to distinguish between benign relative was diagnosed with pancreatic cancer.162 In general, screening
388 Part I: Science and Clinical Oncology

examinations are conducted annually until an abnormality is identified, infected with HCV have 15- to 20-fold higher risk of developing
at which time, intervals may shorten to every 3 to 6 months. For HCC. HCV leads to the formation of cirrhosis (i.e., advanced liver
IPMNs, an international consensus panel recommended screening fibrosis), which is present in about 90% of HCC patients at diagnosis
every 3 to 6 months.163 Ideally, prophylactic resection would be recom- with cancer.173,174 Once HCV-related cirrhosis is established, HCC
mended in patients with PanIN3, IPMN, mucinous cystic neoplasms develops at an estimated rate of 3% per year.175 Studies have identified
with high-grade atypia, or minimally invasive cancer.164,165 a number of risk factors for HCC in HCV-infected individuals,
The development of biomarkers to assist in screening or to determine including older age, male sex, Hispanic ethnicity, HCV genotype 3,176
the malignant potential of pancreatic lesions is currently of great HBV or HIV coinfection, diabetes, obesity, and prolonged, heavy
interest. At present, CA19-9 is the marker primarily being evaluated, alcohol drinking.177 HCV viremia of any level is a strong risk factor
in addition to imaging.166 There is potentially a vast array of circulating for HCC compared with no viremia. Virologic cure of HCV, which
markers that could be explored for such purposes, from nucleic acids is becoming very common with the advent of directly acting antivirals,
to proteins, metabolites, autoantibodies, and microvesicles. A highly although resulting in a considerable reduction in HCC risk, does not
performing biomarker panel with the prerequisite performance for eliminate this risk, especially in those who have already developed
screening, demonstrated through prospective blinded validation studies, cirrhosis.
has yet to emerge. The metabolic syndrome and its components have been associated
with higher risk of HCC. Epidemiologic studies have consistently
LIVER CANCER reported increased risk of HCC in patients with type 2 diabetes.
Multiple meta-analyses on this topic show a 2.5-fold higher risk of
Worldwide, primary liver cancer is the sixth most frequent cancer and HCC associated with diabetes. The association did not vary by study
the second leading cause of death from cancer. An estimated 782,500 design (in both case-control and cohort studies) and was independent
new cases of liver cancer were diagnosed and 745,500 deaths from of viral hepatitis and alcohol use.178,179 Obesity, particularly abdominal
liver cancer occurred worldwide during 2012.167 However, incidence obesity and obesity at an earlier age, is associated with increased risk
and mortality rates vary greatly around the world and by sex, such for HCC.180–182
that liver cancer is the second most common cancer among men in Epidemiologic studies show that NAFLD, the hepatic manifestation
developing countries.167 of the metabolic syndrome, is associated with a modest increase in
In the United States, liver cancer rates have more than doubled over the risk of developing HCC; however, the few population-based cohort
the past two decades, and liver cancer is currently the fastest growing studies of patients with NAFLD have been limited by the low numbers
cause of cancer-related death.168 In 2018, as estimated 42,220 new of HCC cases, and the higher risk is largely limited to those NAFLD
cases of liver and intrahepatic bile duct cancer will be diagnosed; and patients who develop cirrhosis.183 Genomic-wide association studies
about 30,200 will die from the disease in the United States.62 Because report an association between variation in the patatin-like phospholipase
of the growing number of patients with advanced hepatitis C virus domain-containing 3 (PNPLA3) gene and risk of NAFLD.184 PNPLA3
(HCV) and/or nonalcoholic steatohepatitis (NASH), it is anticipated may also determine individual susceptibility to HCC development
that hepatocellular carcinoma (HCC) incidence will continue to rise and poor prognosis.185
over the next 20 years. At its current rate of increase, HCC is projected
to surpass breast cancer and CRC to become the third leading cause Screening and Surveillance
of cancer-related death in the United States by 2030.143
Of all primary liver and intrahepatic cancers diagnosed in the Because most risk factors for HCC in the United States and other
United States, more than 70% are classified as HCC.169 HCC incidence developed countries are associated with cirrhosis, patients with cirrhosis
rates among men are twice as high as the incidence rates among are the current target of cancer prevention and surveillance efforts for
women. HCC is rare among persons aged younger than 40 years. HCC.186 However, there are multiple points of failure in this surveillance
HCC incidence rates reach a peak at approximately 70 years of age. process. For example, in the United States, underrecognition of cirrhosis
Despite improvement in prevention and treatment, currently less than (e.g., >20% of HCC patients have unrecognized cirrhosis at diagnosis)
half of all HCC cases are diagnosed at an early stage, and most patients is one of the main reasons for the underuse of HCC surveillance.187
do not receive curative therapy.170 Overall 5-year survival rates in This is especially important given that patients with NASH have the
HCC remain less than 20%.169 highest rates of unrecognized liver disease,188 and the fraction of HCC
patients missed through surveillance of cirrhosis is likely to increase
Risk Modeling and Assessment as NAFLD becomes the leading cause of HCC in the United States.
Uptake of surveillance as well as the sensitivity of the tools used will
Major risk factors for HCC include chronic infection with hepatitis determine the effectiveness of HCC surveillance. A modeling study
B virus (HBV) or HCV, alcoholic liver disease, and nonalcoholic fatty found that percentages of surveillance use and sensitivity must exceed
liver disease (NAFLD). Approximately 350 million people worldwide 34% and 42%, respectively, for HCC surveillance to be associated
have HBV infection, and HBV accounts for more than 50% of all with a survival benefit.189
cases of HCC worldwide. Among active HBV carriers, the risk of Results from a large RCT among patients in China with chronic
developing HCC is approximately 0.02 to 0.2 per 100 person-years.171 HBV infection provide the best evidence to date for HCC surveil-
HCC risk is substantially higher among HBV patients with compensated lance.190 The study randomly assigned over 9000 HBV carriers to a
cirrhosis (2.2–3.7 per 100 person-years) compared with those with surveillance with ultrasound and α-fetoprotein (AFP) group or a no
chronic HBV infection without cirrhosis (0.3–0.63 per 100 person- surveillance group and showed that the HCC mortality rate was
years).171 Furthermore, HCC risk in patients with chronic HBV significantly lower in the screened group (83.2 per 100,000 person-years)
infection varies according to sex, age, and race or ethnicity, as well as compared with the nonscreened group (131.5 per 100,000 person-years)
by a number of viral factors (higher levels of HBV DNA and hepatitis (HR, 0.63 [95% CI, 0.41–0.98]).190 The improved outcomes in the
B surface antigen; hepatitis B e antigen [HBeAg] positivity; HBV surveillance group included a significantly higher proportion of patients
genotype; duration of infection; and coinfection with HCV, HIV, or with HCC who were diagnosed with early-stage tumors (61% versus
hepatitis D virus [HDV]) and environmental factors (heavy intake none) and received curative therapy (47% versus 8%).
of alcohol and possibly tobacco, exposure to aflatoxin).171 However, in the United States where surveillance targets high-risk
In the United States, most of the burden of HCC is due to chronic patients with cirrhosis and where methods for cancer detection and
HCV infection (approximately 70% of HCCs in the United States therapy differ from those for HBV patients, these data may be less
are attributed to HCV).172 Compared with those not infected, persons applicable.191 Currently, the only evidence around efficacy of HCC
Screening and Early Detection  •  CHAPTER 23 389

surveillance in patients with cirrhosis comes from observational studies, similarly, gastric cancer is more frequent in Hispanics, African
with their inherent limitations of lead-time, length-time, and selection Americans, and Native Indians than whites.209
bias. In their meta-analysis of 47 studies, Singal and colleagues reported The epidemiology of gastric cancer has changed over recent
that HCC surveillance in patients with cirrhosis was associated with decades.212 Data from the European Cancer Observatory for cancer
earlier stage at diagnosis, receipt of curative therapy, and overall registries from 26 European countries confirm a decline in the incidence
survival.192 Although an RCT among patients in cirrhosis would provide of gastric cancer from 1988 to 2008.213 Studies have shown that this
the best evidence, it may be unethical to withhold surveillance from decline is due to decreases in Helicobacter pylori infection, cigarette
some patients, given the data from the observational studies.193,194 smoking, and salt intake, and increases in use of refrigerated foods
Nonetheless, as has been the case in other settings (e.g., colonoscopy and availability of fresh produce.214 However, in the United States
is widely accepted for CRC screening without data from randomized between 1977 and 2006, incidence rates for distal gastric cancer have
trials), a lack of RCT data supporting HCC surveillance for patients increased among whites aged 25 to 39 years; the cause of this increase
with cirrhosis does not mean that surveillance is ineffective. remains unknown.212
Currently, liver ultrasound is recommended as the backbone of
HCC surveillance testing. Semiannual surveillance with abdominal Risk Modeling and Assessment
ultrasound has been shown to be efficacious for early tumor detec-
tion,195–197 safe, and cost-effective.198–201 Alternative imaging approaches Chronic infection with H. pylori is the main cause of gastric cancer,
(e.g., CT and MRI) have been proposed to increase sensitivity for accounting for approximately 89% of distal gastric cancer cases
each HCC; however, there are limited data on their efficacy in HCC worldwide.215–217 In 1994 the IARC classified H. pylori as a class I
surveillance. In the only randomized trial to date, ultrasound and carcinogen; it reconfirmed this classification in 2009.218 H. pylori is
annual CT detected similar proportions of early HCCs (6.0% versus a Gram-negative bacterial pathogen that selectively colonizes the human
6.3%), but biannual ultrasound was less costly per HCC detected gastric mucosa and modulates the host’s immune response.219 Most
($17,000 versus $57,000).202 of H. pylori infection is acquired during childhood; once established,
Finally, although on one hand we should continue to focus on infection usually persists for life in the absence of treatment. The
providing a platform to successfully implement HCC surveillance,203 prevalence of H. pylori infection in adults exceeds 50% in many
there is also a clear and urgent need for interventions to address its industrialized countries; however, there is substantial regional variation
shortcomings. For example, interventions could be aimed at the provider in prevalence.220 Prevalence of H. pylori infection is higher in Central
or system level. Although patients are engaging with HCC surveil- and South America and in parts of Asia and Eastern Europe, in
lance,204 providers continue to report difficulty in identifying at-risk comparison with North America, Australia, and Western Europe.220
patients with cirrhosis, suboptimal levels of knowledge about HCC Other, less common causes account for up to 10% of gastric cancers
surveillance guidelines, and time constraints, as barriers to effective and include infection with the Epstein-Barr virus (EBV), autoimmune
HCC surveillance.193 Similarly, focus should be on biomarkers for gastritis, and Ménétrier disease. An international pooled analysis of
improved risk stratification. AFP is one biomarker examined for use 15 studies and involving 5081 gastric cancer cases reported that
in the early detection of HCC. Studies using assays for AFP in combina- approximately 8% of gastric cancers harbor EBV,221 but there is
tion with age, platelet count, and ALT have shown potential for insufficient epidemiologic evidence of a clear etiologic role for EBV
clinical use.205 New biomarkers, including AFPL3 and DCP (FDA in gastric carcinogenesis.218
approved), GP73, and osteopontin, are being evaluated for early HCC Whereas most gastric cancers are sporadic, they can be hereditary,
detection in phase III biomarker studies, including the EDRN.206 associated with specific mutational profiles. Risk of gastric cancer is up
HCC has become the fastest rising cause of cancer-related deaths to 10 times higher in persons with a family history of gastric cancer.215
in the United States. Despite screening and surveillance efforts and Gastric cancer may also develop as part of a familial cancer syndrome,
improved treatments, overall survival for patients diagnosed with HCC such as hereditary diffuse gastric cancer syndrome, Lynch syndrome
remains poor. We have identified actionable information that can be (in particular in patients with an MLH1 or MSH2 mutation222), FAP,
used to reduce the HCC burden; however, multiple challenges at the Peutz-Jeghers syndrome, and Li-Fraumeni syndrome.223
patient, provider, and system levels need to be addressed in order Other factors associated with increased risk for gastric cardia and
improve on current efforts. noncardia cancers include tobacco smoking, low socioeconomic status,
low level of physical activity, and radiation exposure; obesity and
GASTRIC CANCER gastroesophageal reflux disease are associated with increased risk of
gastric cardia cancer only.224 A report on dietary factors and cancer
Globally, gastric cancer is one of the most common cancers. In 2012, prevention published by the WCRF/AICR stated that nonstarchy
952,000 new cases of gastric cancer were diagnosed worldwide vegetables and fruits probably protect against gastric cancer.225 A
(representing 6.8% of all cancers diagnosed in 2012).207 It is the third high-salt diet has been associated with a higher risk for gastric cancer,225
most common cause of cancer-related deaths, accounting for 8.8% and salt may act synergistically with H. pylori infection to confer
of all cancer-related deaths in 2012.207 Gastric cancer is rare among especially high risk for gastric cancer.226,227 Diets high in nitroso
young persons, with incidence increasing with age and reaching a compounds may also be associated with increased risk for gastric
plateau between 55 and 80 years, and rates are twofold higher in men cancer.225 Conversely, use of aspirin and nonsteroidal antiinflammatory
than in women.208 The highest incidence rates of gastric cancer in drugs and statins may lower risk for gastric cancer.224
men are seen in Eastern Asia (Korea, Mongolia, Japan, and China,
with incidence rates between 40 and 60 per 100,000), Eastern Europe Screening and Surveillance
(35 per 100,000), and Central America and the Andean Region (with
incidence rates between 20 and 30 per 100,000).209,210 The highest Even in countries where gastric cancer is relatively common, mass
mortality rates for gastric cancer are seen in Eastern Asia (24 per screening for gastric cancer is generally not included in national
100,000 in men; 9.8 per 100,000 in women), and the lowest in strategies for cancer prevention. There are a number of reasons for
Northern America (2.8 and 1.5, respectively). However, high mortality this, including high cost, decreasing incidence, and a lack of data on
rates are also seen for men and women in Central and Eastern Europe, whom, when, and how to screen. Furthermore, screening services are
and in Central and South America. Notably, the incidence rates for complex, with success hinging on the underlying processes and efforts
gastric cancer also vary among different ethnic groups within a particular to ensure its overall quality.228
country. For example, in the United States, annual incidence among Globally, various types of tests have been proposed and used for
Asians is twice as high as in whites (15.6 versus 7.4 per 100,000);211 gastric cancer screening, with the implementation of endoscopic
390 Part I: Science and Clinical Oncology

methods limited to high-risk populations. Several countries including population-based mass screening programs aimed at reducing the
Venezuela, Chile, and East Asian countries such as China and Japan incidence of gastric cancer. Although mass screening strategies could
have implemented a variety of screening programs. For example, in be beneficial, current modalities are not yet readily implementable in
Korea, the Korean Gastric Cancer Association and National Cancer organized screening settings. Conversely, since the population risk
Center established national guidelines for gastric cancer screening in (based on histology) can change rapidly and H. pylori eradication
2001. These guidelines recommend biennial gastric cancer screening eliminates the problem, population-based programs of screening and
for men and women aged 40 years or older via endoscopy or upper treatment for H. pylori may hold the greatest promise for reducing
gastrointestinal series.229 As a result, a nationwide gastric cancer the burden of gastric cancer.239
screening program in Korea was started in 2002. Other screening
methods for gastric cancer include serum pepsinogen, serum gastrin-17, ESOPHAGEAL CANCER
and H. pylori antibody testing. In 2013 the Japanese government
approved insurance coverage for a gastric cancer prevention program When considering screening and early detection of esophageal cancer,
that includes H. pylori screening and treatment (primary prevention), it is essential to take into account the changing epidemiology of the
as well as posttreatment surveillance (secondary prevention for people disease. Esophageal cancer is relatively uncommon; however, it is a
with atrophic gastritis).230,231 highly fatal malignancy. Worldwide, esophageal cancer is the eighth
Upper gastrointestinal endoscopy studies have been shown to have most common cancer (456,000 new cases in 2012) and the sixth
the highest detection rates among the screening tests and have been most common cause of cancer-related death (400,000 deaths in 2012).207
universally regarded as the gold standard for the diagnosis of gastric The highest incidence rates of esophageal cancer are seen along two
cancer.224 Although the available evidence shows that endoscopic geographic belts: one from north central China through the central
population screening is cost-effective in areas with high gastric cancer Asian republics to northern Iran, and one from eastern to southern
burden,232 further studies are needed to evaluate the impact of this Africa. In the United States, 16,940 new cases of esophageal cancer
technique before its broad use as a primary screening test can be recom- and 15,690 deaths from esophageal cancer are expected to occur in
mended.233 Upper gastrointestinal series has continuously been analyzed 2017.62 Although treatment effectiveness has improved during the
as a rivaling modality for cancer detection but has not shown clear past decade, survival rates for esophageal cancer remain poor. In the
benefits over endoscopy.234 United States, the overall 5-year survival rate for patients diagnosed
There is increasing attention given to identifying higher-risk patients with esophageal cancer is less than 20%.242 There are two main histologic
(e.g., patients with advanced precancerous lesions) for gastric cancer subtypes of esophageal cancer: esophageal adenocarcinoma and
surveillance. In a cohort study conducted among patients in the Dutch esophageal squamous cell carcinoma. There has been a dramatic shift
nationwide histopathology registry, the 10-year gastric cancer risk in in the epidemiology of esophageal cancer in Western populations,
patients with mild to moderate dysplasia was 4%, and for severe such that esophageal adenocarcinoma, which typically affects the lower
gastric dysplasia was 33%.235 In a cohort of high-risk Chinese dysplastic third of the esophagus, has become the predominant subtype of
patients, after 5 years of follow-up, these rates were 3% and 7%, esophageal cancer in North America, Australia, and Europe.243–245
respectively.236 These patients are the ideal target for gastric cancer Among white men in the United States, the annual incidence of
surveillance; however, no evidence-based guidelines for follow-up esophageal adenocarcinoma has increased 10-fold (0.6 per 100,000
strategies have been established, and the few recommendations available to 6.0 per 100,000) since the early 1970s.246 In contrast, esophageal
stem mostly from expert opinions.237 squamous cell carcinoma is now less common than esophageal adeno-
However, there are some data among healthy asymptomatic carcinoma in the Unites States. To stem the increasing incidence and
infected Asians that show that eradicating H. pylori reduces the to improve outcomes for patients diagnosed with esophageal cancer,
incidence of gastric cancer. A systematic review and meta-analysis it is crucial to develop new strategies of screening and surveillance.
of six international RCTs compared risk of incident gastric cancer in
adults who had tested positive for H. pylori and received eradication Risk Modeling and Assessment
therapy or placebo or no therapy.238 The risk of developing incident
gastric cancer was 34% lower among the 3294 individuals who Esophageal Squamous Cell Carcinoma
received eradication therapy compared with the 3203 control subjects Unlike the situation in the United States, elsewhere esophageal
(relative risk [RR], 0.66 [95% CI, 0.46–0.95]). This corresponds squamous cell carcinoma is the most common histologic subtype of
to a number needed to treat to prevent one gastric cancer of 124 esophageal cancer.207 Although the incidence of esophageal adeno-
(95% CI, 78–843).238 carcinoma has increased, the incidence of esophageal squamous cell
The potential effect of a gastric cancer prevention program that carcinoma has decreased in the United States.244 Alcohol consump-
includes H. pylori screening and treatment is dependent on a patient’s tion and tobacco smoking are the main risk factors for esophageal
level of cancer risk at the time that H. pylori is eradicated (e.g., consider- squamous cell carcinoma, such that more than 75% of the esophageal
ing the twofold higher risk of gastric cancer between the most and squamous cell carcinoma burden in men could be attributed to smokers
the least virulent strains). Under various assumptions about both with heavy alcohol consumption.247 The decrease in incidence of
effectiveness and costs, performance of population-based screening esophageal squamous cell carcinoma in the United States is thought
for H. pylori and eradication of the infection has also been shown to to be due to the decreasing consumption of alcohol and tobacco.
be cost-effective.232,239 A phase III RCT in school-aged children reported Squamous cell carcinoma is believed to develop through progres-
the success of a prophylactic oral H. pylori vaccine,240 and a large sion through a premalignant dysplastic phase before development of
community-based intervention trial in China demonstrated that H. carcinoma.248
pylori eradication is feasible and acceptable; however, they also identified
several risk factors for the failure of eradication therapy to prevent Barrett Esophagus and Esophageal Adenocarcinoma
gastric cancer.241 Studies have shown that under conditions of low Epidemiologic studies have implicated gastroesophageal reflux
bacterial burden and acute infection, H. pylori infection may be disease, obesity, and cigarette smoking as the main risk factors for
prevented, and additional follow-up of these patients should provide esophageal adenocarcinoma. Together, these three risk factors account
promising data on the preventive effect of eradication on the incidence for over 70% of all cases of esophageal adenocarcinoma in Western
of gastric cancer. populations.249,250 Barrett esophagus, a condition in which the normal
Further improvements in screening, treatment, and early diagnosis squamous mucosa of the esophagus is replaced by columnar intestinal
are needed for gastric cancer, which remains the third most common epithelium (i.e., metaplasia), is the only known precursor lesion for
cause of cancer-related death worldwide. In general, there are no esophageal adenocarcinoma, likely because of progression through
Screening and Early Detection  •  CHAPTER 23 391

dysplasia. Barrett esophagus is present in up to 15% of individuals and acceptable image quality, and biopsy specimens, although small in
with frequent symptoms of gastroesophageal reflux disease and in size, are still sufficient for acceptable histologic analysis, demonstrating
1% to 2% of the general adult population.251 Compared with the its potential.267,268 However, although this modality is generally preferred
general population, patients with Barrett esophagus have at least a by patients and is associated with less patient anxiety than conventional
10-fold higher risk for esophageal adenocarcinoma.252 Among those sedated endoscopy, acceptance and use among primary care providers
with nondysplastic Barrett esophagus, 0.33% per year progress to have been limited owing to concerns about patient tolerance of an
esophageal adenocarcinoma253; patients with Barrett esophagus who unsedated procedure and uncertainties regarding the safety, time, train-
have low- and high-grade dysplasia progress at rates of 1% and 6% ing, and facilities required for this procedure. For gastroenterologists
per year, respectively.254 and other endoscopists, there is an additional financial disincentive
for performing an office-based procedure: lower reimbursement rates
Screening and Surveillance than for standard upper endoscopy with biopsies.269
Molecular screening methods might soon be practical for Barrett
Owing to the low incidence of esophageal squamous cell carcinoma esophagus. Esophageal capsule cytology (Cytosponge; Medical Research
in the United States, screening for this disease in the general US Council, London, UK) is a new technique that has the potential to
population is not recommended. Screening may be considered for provide a noninvasive, nonendoscopic method of screening for Barrett
specific higher-risk subgroups (e.g., patients with oral and oropharyngeal esophagus.270 The device, which can be used in the office setting
cancer, patients with tylosis). Likewise, based on the available evidence, without sedation, is a sponge within a capsule covered in a gelatin
population-wide mass screening for Barrett esophagus and esophageal layer, which dissolves after swallowing. After that the sponge is released,
adenocarcinoma is seen as not being cost-effective.255 However, despite and esophageal epithelial cells are retrieved.271 Immunohistochemistry
the lack of high-level evidence and remaining questions around whom, is then performed on these cells to analyze for markers such as trefoil
when, and how often to screen, endoscopic screening (and subsequent factor 3, which can differentiate the cells of Barrett esophagus from
surveillance) in high-risk patients is advised, based on expert opinion.256 other columnar cells found in the normal gastric cardia and upper
For example, the 2016 guidelines from the American College of airway.271 Depending on the length of the Barrett esophagus segment,
Gastroenterology recommend endoscopic screening for Barrett prospective cohort studies conducted in the United Kingdom have
esophagus in men with chronic (>5 years) and/or frequent (weekly shown that this technique has a sensitivity of 80% to 90% with
or more) symptoms of gastroesophageal reflux disease and two or a specificity of approximately 92%.272 Additional clinical trials to
more risk factors for Barrett esophagus and esophageal adenocarcinoma further validate this technique (and the use of other biomarkers) are
(e.g., age above 50 years, white race, obesity, history of cigarette ongoing.
smoking, and history of Barrett esophagus or esophageal adenocarci- The incidence of esophageal cancer, especially esophageal adeno-
noma in FDRs).257 carcinoma, is increasing, and overall survival for patients diagnosed
Multiple professional societies have advocated for endoscopic with esophageal cancer remains poor. The ability to increase survival
surveillance in patients with known Barrett esophagus, with the goal in this disease will depend on earlier detection through screening and
of detecting early neoplastic changes (e.g., dysplasia) and early-stage surveillance; however, screening for Barrett esophagus has several
esophageal adenocarcinoma that can be cured with endoscopic or limitations. Because patients are often asymptomatic, it is difficult to
surgical resection. For patients with nondysplastic Barrett esophagus, identify the entire at-risk group and therefore risk-based screening
periodic endoscopic surveillance is recommended every 2 to 3 years may miss a considerable proportion of patients with Barrett esophagus
and requires obtaining random biopsy specimens from the Barrett and esophageal adenocarcinoma. For example, several studies have
region(s) as well as targeted biopsy specimens from nodules or ulcers. shown that about 40% of patients diagnosed with esophageal adeno-
Although evidence from randomized trials is lacking (and will be very carcinoma report no history of gastroesophageal reflux symptoms.273,274
difficult to obtain, given logistic challenges), there is a growing body Furthermore, only 7% of patients with esophageal adenocarcinoma
of evidence from observational studies that having a Barrett esophagus have a prior diagnosis of Barrett esophagus.258 Therefore, current
diagnosed is associated with better neoplasia-associated outcomes.258,259 approaches to esophageal cancer control are missing 93% of cases
These outcomes include earlier stage and greater chances for receipt owing to an emphasis on symptoms. If we are to prevent esophageal
of curative treatment, and importantly reduced cancer-related mortality. adenocarcinoma, much more work needs to be done around screening
Endoscopic surveillance in patients with known Barrett esophagus strategies for Barrett esophagus.
may be cost-effective where curative therapy is available and patients
have limited comorbidities.260,261 Furthermore, continuous endoscopic HEAD AND NECK SQUAMOUS
screening and surveillance have been fortified by the success associated CELL CARCINOMA
with endoscopic radiofrequency ablation in patients with Barrett
esophagus (including resolution of Barrett esophagus and dysplasia, Head and neck squamous cell carcinomas (HNSCCs) can occur on
and durability).262–264 As this technology improves in efficacy and the lip, in the oral cavity, and in the pharynx (nasopharynx, oropharynx,
safety, endoscopic radiofrequency ablation may be an alternative for and hypopharynx) and larynx. Worldwide, approximately 600,000
nondysplastic Barrett esophagus, at least for those at high risk for cases of HNSCCs occur annually, with oral cancer comprising at least
progression (e.g., long segment, severe reflux, family history of Barrett half of the cases.275 Incidence of HNSCC varies greatly around the
esophagus or adenocarcinoma).265 globe because of differences in the prevalence of its primary risk
Upper endoscopy remains the gold standard for Barrett esophagus factors: alcohol, tobacco, betel quid with or without added tobacco
screening, but it has several limitations as a screening tool, including (mainly in Asia), and, more recently identified, HPV. The ACS estimates
its cost (i.e., related to endoscopy, histopathology examination, and that nearly 50,000 cancers of the oral cavity and pharynx will occur
sedation), low NPV for short segments of Barrett esophagus (high in the United States in 2017, with an additional 13,000 laryngeal
NPV is necessary for endoscopy to be a valid screening tool), and cancers.18 Incidence and mortality are generally higher among men,
risk to the patient. Consequently, less invasive screening that can be with each being nearly three times higher in US men than in US
performed without the need for sedation (unsedated endoscopy) has women. Outcomes are dependent on the anatomic site and stage of
been proposed and shown to be tolerable and accurate compared disease at diagnosis. Two-thirds of individuals with oral and pharyngeal
with conventional sedated endoscopy.266 Transnasal endoscopy is one cancers have regional or distant metastases at the time of diagnosis
particular screening modality that can be performed in an office setting and therefore have 5-year survival rates of 64.2% and 38.5%, respec-
without sedation. Transnasal esophageal endoscopy has been assessed for tively.276 HNSCCs are notable for their high rate of recurrence and
feasibility of Barrett esophagus screening. It has relatively high accuracy second primary tumors.
392 Part I: Science and Clinical Oncology

Risk Modeling and Assessment oropharyngeal cancer. The HPV vaccine has the potential to reduce
oral HPV infections. Herrero and colleagues demonstrated that vac-
Risk assessment is generally based on an individual’s current or past cination against HPV strains 16 and 18 prevented more than 90%
history of exposure to one of the primary factors. Numerous epide- of these oral infections within 4 years of vaccination.289 However, it
miologic studies have documented the increased risk associated with is still too early to assess whether this will lead to a reduced risk of
each factor. HPV-associated oropharyngeal cancer later in life.
All forms of tobacco, including smokeless tobacco, are associated
with an increased risk of HNSCC.277 Current cigarette use increases Screening
the risk of oral and oropharyngeal cancers by approximately tenfold
for men and fivefold for women, compared with lifetime nonsmokers.277 Currently, no organized screening programs for any HNSCC exist
Alcohol increases the risk fivefold for those consuming five or more because there is a lack of RCT data to support their use. Screening
drinks per day relative to those who do not drink; and an increased for oral cancer can be carried out through visual and tactile inspection
risk has been observed in nonsmokers and in studies that control for of the oral cavity by a health care provider or by self-examination.
confounding by smoking, demonstrating a direct effect of alcohol.278,279 The mouth is easily accessible, and there are known premalignant
In combination, these two risk factors confer a risk that is two to lesions associated with oral cancer development. However, to date,
three times greater than the risk expected if one were to assume a just one RCT has examined the potential benefits of oral cancer
simple multiplicative effect.280 Individuals who smoke two or more screening.290,291 This was a cluster randomized trial of 13 districts (N
packs per day along with consuming four or more alcoholic drinks = 191,873) in India, which has some of the highest rates of tobacco-
per day have an approximate 35-fold increased risk of oral or oro- associated oral cancer in the world, and used advanced health workers
pharyngeal cancers in comparison with those who neither smoke nor to perform the screening during home visits.292 There was no statistically
drink. In Asia, the use of betel quid and gutka (betel quid combined significant difference in either oral cancer mortality or incidence between
with tobacco) is common. An Indian meta-analysis has suggested that screened and unscreened groups. However, a stage shift and an increase
betel quid doubles the risk of both oral and oropharyngeal cancers in survival were observed in the screened group. Of oral cancer cases
and that gutka confers an eightfold increased risk for oral cancer and in the screened districts, 41% were stage I or II, whereas just 23% of
a fourfold increased risk for oropharyngeal cancer.281 cases were stage I or II in the unscreened districts (P = .004); and
Tobacco cessation significantly reduces the risk of both oral and 5-year survival was 50% in the screened areas versus just 34% in
oropharyngeal cancers, with an approximate 50% risk reduction unscreened areas (P = .009). In a post hoc subgroup analysis of high-risk
observed after 5 to 9 years of quitting and a return to risk comparable individuals (i.e., those using tobacco, alcohol, or both; N = 84,600),
to never-smokers within two decades.282 The data are less strong for a significant 34% reduction in oral cancer mortality was observed in
cessation of alcohol. Nevertheless, given the dose-response relationship the screened group (RR, 0.66 [95% CI, 0.45–0.95]). Although these
of alcohol with oral cancer, it is believed that cessation or a reduction results suggest that screening may be beneficial among high-risk groups,
in consumption would result in reduced risk. A meta-analysis of studies the generalizability of these results to the United States and to other
by the International Head and Neck Caner Epidemiology Consortium Western populations is unknown, given differences in disease prevalence,
suggests a statistically significant 40% reduction in HNSCC risk risk factor distribution, and health care systems. In addition, the trial
(compared with current drinkers) in those who quit drinking at least suffers from a number of flaws, including possible lead-time and
20 years ago.282 By subsite, a 34% risk reduction was seen for oral length-time bias, low compliance with follow-up, and an imbalance
cancer after 10 to 19 years of cessation; and a 31% risk reduction in baseline risk factors.290
was observed at 20 or more years of cessation for laryngeal cancer. In addition to this single RCT, a number of diagnostic accuracy
No statistically significant reduction was observed for oropharyngeal studies have been conducted for conventional oral cancer screening
or hypopharyngeal cancers. Less is known regarding the effect of (visual and tactile inspection by a health care worker), mouth self-
cessation of betel quid and gutka on risk of HNSCC. A 2016 study examination, light-based detection, and vital rinsing (e.g., toluidine
of tobacco-free betel quid in Taiwan demonstrated that every year of blue). These have been conducted in both apparently healthy individuals
cessation was associated with a 2.9% risk reduction for HNSCC, and in those with clinically evident lesions. A review of 10 studies
although risk among those who quit never returned to the level of conducted in apparently healthy populations demonstrated great
nonusers, even after 20 years.283 More studies with long-term follow-up variability in the sensitivity of conventional oral examination (COE),
are needed in areas of betel quid and gutka use to more thoroughly ranging from 0.50 (95% CI, 0.07–0.93) to 0.99 (95% CI, 0.97–1.00)
assess the effect of cessation on subsequent risk of HNSCCs. depending on the prevalence of oral cancer and premalignant lesions,
In the last 15 years, the recognition that HPV is causally linked but found consistently high specificity of around 0.98 (95% CI,
to HNSCCs, primarily to oropharyngeal cancer, has led to a paradigm 0.97–1.00).293 Two studies of mouth self-examination demonstrated
shift in the prevention, detection, and treatment of these cancers. The much lower estimates of sensitivity, 0.18 (95% CI, 0.13–0.24) to
fraction of HNSCCs attributable to HPV varies greatly by geographic 0.33 (95% CI, 0.10–0.65), and variable specificity, 0.54 (95% CI,
region, gender, and age at diagnosis. Studies have suggested worldwide 0.37–0.69) to 1.00 (95% CI, 1.00–1.00). One RCT found a higher
attributable fractions in the range of 18.5% to 39.8% of oropharyngeal rate of detection of oral cancer in the arm of conventional oral
cancers, 7.5% to 16.1% of unspecified pharyngeal cancers, 1.1% to examination plus toluidine blue versus conventional oral examination
5.9% of nasopharyngeal cancers, 2.2% to 16.3% of oral cancers, 1.5% alone, but results were not significant and the overall risk of bias in
to 8.6% of laryngeal cancers, and 2.4% of hypopharyngeal cancers.284–286 this study was judged as unclear; In addition, there was concern among
However, within the United States, data support an HPV-etiologic the reviewers regarding the overall applicability of the RCT, given its
fraction of at least 72% for oropharyngeal cancer.287 The variability method of patient selection. A review of 41 studies of individuals
in HPV attributable fractions originates from differences in incidence with clinically evident lesions found them to be generally of poor
of HPV infection and patterns of behavior, such as sexual practices quality and that none of the adjunctive tests could be recommended
and tobacco use, around the world and across time. Data show that to replace scalpel biopsy and histologic assessment, although cytologic
oral HPV infection is the major risk factor for HPV-positive oropha- evaluation showed promise with both high sensitivity (0.91 [95% CI,
ryngeal cancer, and that more than 90% of these oral infections are 0.91–0.96]) and specificity (0.91 995% CI, 0.91–0.950).294 Neither
sexually acquired.287 Indeed, lifetime number of oral sex partners is review found eligible studies of blood or salivary sample (biomarker)
strongly, consistently, and specifically associated with oropharyngeal analysis.293,294
cancer.287,288 Current studies are examining the risk of oral HPV Given this limited evidence, the USPSTF concluded in 2013 that
infection and cancer in partners of patients with HPV-positive evidence was insufficient to assess the balance of benefits and harms
Screening and Early Detection  •  CHAPTER 23 393

of oral cancer screening by primary care physicians. The American studies showed a 72% increase in risk of ovarian cancer in premeno-
The American Dental Association has recently published updated pausal women who were obese (BMI of 30 or higher) compared
evidence-based clinical practice guidelines for the evaluation of with women of a healthy weight (BMI of 18.5–23). The same
potentially malignant disorders in the oral cavity.295 comparison in postmenopausal women did not identify a link between
The emergence of HPV as a risk factor for oropharyngeal cancer a high BMI and ovarian cancer risk.306 However, results from the
suggests the possibility of screening for oral HPV infection as a means European Prospective Investigation Into Cancer and Nutrition (EPIC)
of prevention and/or early detection, but there is currently no FDA- suggest that being obese after menopause may also increase the risk
approved screening test for oral HPV infection.296 The use of cytologic of ovarian cancer.307
evaluation and HPV DNA testing, as in the cervix, offers two potential An interesting paradox exists with regard to hormones and ovarian
approaches. However, cytologic assessment may be limited by the cancer risk. Although risk of ovarian cancer appears to be increased
anatomic location and properties of oropharyngeal cancer and the with use of postmenopausal hormones, studies have suggested that
tonsillar epithelium, and HPV DNA testing faces technical challenges, use of OCs decreases the risk of ovarian cancer. A systematic review
including how best to obtain a sample.297 Noninvasive sampling strate- of published case-control cohort studies and randomized trials revealed
gies that could be applied in the context of large-scale population-based that use of estrogen-only therapy for 5 years increased the risk of
screening are oral rinses and gargles; however, these strategies are not ovarian cancer by 22%, significantly more than the 10% risk increase
specific to the tonsillar epithelium and may overestimate the actual with use of estrogen-progestin hormone therapy (EPT).308 The UK
rate of tonsillar HPV infection.297 In addition, the lack of knowledge Million Women Study reported that current users of postmenopausal
regarding the natural history of oral HPV infections and the likelihood hormones had a 20% greater risk of developing ovarian cancer (RR,
of a high-risk HPV infection to persist and progress to cancer challenges 1.20 [95% CI, 1.09–1.32]; P = .0002]) and a 23% higher risk of
the use of an HPV DNA testing approach to screening. Blood tests dying from the disease (RR, 1.23 [95% CI, 1.09–1.38]; P = .0006]).
are a third potential screening strategy for HPV-driven HNSCCs, Although incidence of ovarian cancer was higher with increased duration
and recent data suggest that this may be a promising, noninvasive of use for current users, it did not appear to differ significantly by
approach.298 Finally, oropharyngeal examination with narrow band type of hormonal therapy (estrogen-only therapy or EPT). Past users
imaging299 and transcervical and intraoral ultrasonography300–302 hold were not at increased risk of ovarian cancer.309
potential for diagnostic purposes in individuals with a positive screening In contrast, long-term use of OCs has been associated with a
test result. reduced incidence of ovarian cancer in ever-users compared with
never-users (odds ratio [OR], 0.73 [95% CI, 0.66–0.81]). A significant
OVARIAN CANCER duration-response relationship was seen, with a reduction in incidence
of more than 50% among women using OCs for 10 or more years.310
Ovarian cancer is the deadliest of the gynecologic malignancies and In an analysis of 45 epidemiologic studies, this reduction in risk
is the fifth leading cause of cancer mortality among American women.18 persisted for more than 30 years after OC use had ceased but became
The high mortality rate is a result of the typically late presentation somewhat attenuated over time. The proportional risk reductions per
of the disease, largely because of vague, nonspecific, or even lack of 5 years of use were 29% (95% CI, 23–34%) for use that had ceased
early symptoms. For women at high risk of ovarian cancer due to a less than 10 years previously, 19% (95% CI, 14%–24%) for use that
BRCA mutation, prevention through risk-reducing surgery and early had ceased 10–19 years previously, and 15% (95% CI, 9%–21%) for
detection with screening has the potential to improve the outcomes use that had ceased 20 to 29 years previously.311 Use of OCs has also
of this disease.303 been shown to reduce the risk of ovarian cancer in carriers of BRCA1
mutations (OR, 0.56 [95% CI, 0.45–0.71]; P < .0001) and carriers
Risk Factors and Etiology of BRCA2 mutations (OR, 0.39 [95% CI, 0.23–0.66]; P = .0004).312
In another twist on hormones and ovarian cancer risk, use of the
The majority of ovarian cancers are sporadic; only 5% to 10% of fertility drug clomiphene citrate for longer than 1 year may increase
ovarian cancers are related to genetic mutations.304 Absent other relevant the risk of low malignant potential tumors. However, it should be
family history, a single FDR with ovarian cancer is not suggestive of noted that infertile women may be at higher risk of ovarian cancer
an inherited predisposition. Women having one FDR with the disease even if they do not use fertility drugs. Some of this risk may be due
have a threefold to fourfold increased risk of the disease.305 Although to the fact that they have not carried a pregnancy to term or used
they are at increased risk of ovarian cancer compared with average birth control pills long term, both of which are protective.304
risk women, they do not meet the threshold for ovarian cancer screening With regard to hormonal and reproductive factors, incessant
or prophylactic surgery. The strongest known risk factor for ovarian ovulation has been hypothesized to increase the number of spontaneous
cancer is hereditary breast and ovarian cancer syndrome due to muta- mutations and thus the risk of ovarian cancer.313 This may be the
tions in the BRCA1 and BRCA2 genes.304 In addition, women with mechanism by which the risk of ovarian cancer is reduced by OC
a family history suggestive of a BRCA mutation but who are untested use. In line with this hypothesis, ovarian cancer risk is reduced by
are considered at high risk until testing definitively proves otherwise. factors that interrupt ovulation such as pregnancy and breastfeeding.
BRCA1 mutation carriers have a reported 39% to 63% cumulative Tubal ligation and hysterectomy are associated with a protective effect
risk of ovarian cancer by age 70, whereas the risk for BRCA2 mutation of 30% to 40%.313
carriers is 11% to 31%, depending on whether one is using data from Genital talcum powder has been suggested to be associated with
population-based case series or multicase family data (the latter sug- an increased risk of ovarian cancer. A 2006 meta-analysis of 21 studies
gesting higher risk).305 Ovarian cancers are less commonly the result reported an approximately 35% (95% CI, 1.26–1.46) increase in risk
of other inherited genetic syndromes such as Lynch syndrome, Cowden with genital exposure to talc,314 similar to findings from a 2003 meta-
disease, or Li-Fraumeni syndrome. Genetic testing for inherited analysis.315 In a 2006 review by the IARC, talc was classified as possibly
mutations should be discussed with women who have a significant carcinogenic to humans (group 2B) on the basis that most of the 20
family history of ovarian and/or breast cancer. epidemiologic studies on talc and ovarian cancer consistently show a
Advancing age is associated with an increased risk of epithelial 30% to 60% increased risk associated with talc use.316 Before the
ovarian cancer, with half of all cases of ovarian cancer occurring in mid-1970s, contamination of talc with asbestos fibers was known to
women over the age of 63.304 Obese women have a higher risk of occur, and guidelines were introduced to prevent this in 1975. One
ovarian cancer.304 Some studies have suggested that the effect of BMI study that examined talc use by year showed that use before 1975
on ovarian cancer risk is greater in premenopausal women than in was associated with an increase in risk, whereas use after 1975 was
postmenopausal women. A pooled analysis of 12 prospective cohort not.317 However, use after 1975 has been shown in other studies to
394 Part I: Science and Clinical Oncology

be associated with an increased risk of ovarian cancer similar to that in 3 months was 5.8%, and the average annual rate of TVUS and
seen before 1975.318,319 gynecologic oncologist referral was 0.9%. Ten women subsequently
underwent surgery based on the TVUS findings and referral, revealing
Screening four invasive ovarian cancers (one stage IA, two stage IC, and one
stage IIB), two ovarian tumors of low malignant potential (both stage
To date, an effective screening test for ovarian cancer has not been IA), one endometrial cancer (stage I), and three benign ovarian tumors,
identified. The two tests commonly investigated for ovarian cancer providing a PPV of 40% (95% CI, 12.2%–73.8%) for detection of
screening are CA125 and transvaginal ultrasound (TVUS). CA125 invasive ovarian cancer. The combined specificity of ROCA followed
is elevated in 90% of advanced ovarian cancers but in only 50% to by TVUS for referral to surgery is 99.9% (95% CI, 99.7%–100%).
60% of early ovarian cancers.320 Any process that disrupts the epithelial All four women with invasive ovarian cancer had been enrolled in
lining of the peritoneum has the potential to raise the CA125 level. the study for at least 3 years with low-risk annual CA125 levels before
As a result, CA125 has been identified as being elevated in a number a rising CA125 level.324
of benign conditions including menstruation, pregnancy, endometrio- The UK Collaborative Trial of Ovarian Cancer Screening
sis, adenomyosis, pelvic inflammatory disease, uterine leiomyoma, (UKCTOCS) multicenter RCT enrolled 202,638 postmenopausal
pancreatitis, hepatitis, peritonitis, renal failure, and congestive heart women age 50 to 74 women who were not at increased risk of familial
failure. The low incidence of an elevated CA125 level in early-stage ovarian cancer. Participants were randomly allocated to annual
ovarian cancers and the fact that it is elevated in a number of normal multimodal screening (MMS) with the ROCA to interpret serial CA125
conditions significantly affects its usefulness in ovarian cancer screen- results and TVUS as a second-line test, annual TVUS only, or no
ing. TVUS attempts to discriminate between benign and malignant screening. The ROCA doubled the number of screen-detected cancers
adnexal masses by using information on morphology, color flow imaging compared with a single-threshold rule, detecting 86.4% of the invasive
based on the neovascularity of neoplasms, and measurements of epithelial ovarian and tubal cancers, whereas use of annual serum
pulsatility indices. CA125 fixed cutoffs of more than 35, more than 30, and more than
In 2011, initial results from PLCO were published regarding ovarian 22 U/mL enabled identification of only 41.3%, 48.4%, and 66.5%,
cancer screening in 78,216 women aged 55 to 74 years who were respectively.325 The overall sensitivity for detection of ovarian cancer,
randomized to receive usual care or annual screening for 6 years with diagnosed within a year of screening, was 84% (95% CI, 79%–88%)
CA125 and 4 years with TVUS.321 Although screening identified more for MMS and 73% (95% CI, 66%–79%) for TVUS. A higher propor-
ovarian cancers—212 compared with 176 found in women who tion of invasive epithelial ovarian and peritoneal cancer diagnosed
received usual care (RR, 1.21 [95% CI, 0.99–1.48])—the majority with low-volume disease (stage I, II, and IIIa) was seen in the MMS
of the cancers in both groups were stage III or IV tumors. With a group (40%; P < .0001) than in the no screening group (26%), but
median of 12.4 years of follow-up, there were 118 deaths caused by not in the TVUS group (24%; P = .57). With a median of 11.1 years
ovarian cancer in the intervention group and 100 deaths in the usual of follow-up, the relative mortality reduction was 15% in the MMS
care group (RR, 1.18 [95% CI, 0.82–1.71]). Although no mortality group and 11% in the TVUS group, but these reductions were not
reduction was observed, significant harms were identified when significant in the primary prespecified Cox analysis. However, the
subsequent surgery was considered in screened women with false-positive prespecified secondary subgroup analysis with exclusion of prevalent
results. Of 3285 women with false-positive results, one-third (1080 cases in the MMS group was significant, suggesting that the long-term
women) underwent surgical follow-up; 15% (163 women) of these effect of an MMS screening program is about a 28% mortality reduction
women experienced at least one serious complication. Of the 10% of after 7 years of screening.326 Follow-up continues to determine if the
women in the PLCO’s intervention group who had a false-positive observed stage shift will translate into a significant mortality reduction,
result, one-third had an oophorectomy—33% more oophorectomies with the next analysis planned to be performed in 2019.
than in the control group—and 15% had a serious complication, In the United Kingdom Familial Ovarian Cancer Screening Study
such as infection, surgical complication, or cardiovascular or pulmonary (UK FOCSS), all participants had a BRCA1 or BRCA2 mutation or
event. Extended follow-up of the PLCO has continued to demonstrate an estimated lifetime risk of ovarian cancer greater than 10% on the
a lack of ovarian cancer mortality reduction (RR, 1.06 [95% CI, basis of personal and family history of breast or ovarian cancer. In
0.87–1.30]) after a median of 15 years.322 the phase I portion of the trial, annual CA125 screening with a single
Retrospective studies have shown that rising levels of CA125 within cut point and TVUS had a sensitivity of greater than 80% but did
the normal range may be indicative of ovarian cancer, whereas stable not lead to an appreciable stage shift.327 In the phase II portion of
CA125 levels, even if somewhat elevated, were not predictive of the trial, women not undergoing risk-reducing salpingo-oophorectomy
malignancy. Hypothesizing that changes in CA125 over time may (RRSO) had CA125, triaged according to the ROCA, every 4 months.
predict ovarian cancer, Skates and colleagues developed a risk of ovarian In addition to its use as a second-line test in the ROCA, annual TVUS
cancer algorithm (ROCA). They applied linear regression analyses to was performed if ROCA results were normal. With a median follow-up
longitudinal CA125 measurements from 9233 postmenopausal, of 4.8 years, 19 invasive ovarian or tubal cancers were diagnosed out
average-risk women who had two or more serial samples and a 6.8-year of 4348 women who underwent 13,728 woman-years of screening.
median follow-up. Risk was assigned through use of a baseline CA125 Thirteen were screen-detected cancers (1 prevalent, 12 incident); the
and changes in CA125 over time combined with age and other known other 6 were occult cancers discovered at RRSO. All women were
risk factors. The ROCA was shown to improve sensitivity for detection asymptomatic at cancer detection; only 37% had high-volume/
of preclinical disease from 62% to 86%.323 The algorithm is currently advanced-stage (IIIB or IIIC) disease at diagnosis. Of the occult cancers
in clinical trials in the United States and the United Kingdom. found at RRSO, five of the six were stage I or II. However, four of
The US, single-arm, prospective, multicenter screening study the five had normal ROCA findings, as did the occult stage IIIa cancer.
enrolled postmenopausal women age 50 to 74 with no significant All but one of the screen-detected cancers had intermediate or elevated
family history of breast or ovarian cancer. Participants underwent a ROCA findings; the one incident screen-detected cancer with normal
CA125 blood test annually. Based on the ROCA result, women were ROCA findings was found on the annual TVUS. However, only 5
triaged to the next annual CA125 test (low risk), repeat CA125 test of the 12 incident screen-detected cancers (42%) were stage I or II,
in 3 months (intermediate risk), or TVUS and referral to a gynecologic making the main stage shift reported less impressive.328
oncologist (high risk). Based on clinical findings and TVUS, the Currently, there is insufficient evidence to support routine ovarian
gynecologic oncologist made the decision regarding whether to proceed cancer screening with CA125 and TVUS in women at average risk
with surgery. The US study enrolled 4051 women over an 11-year of ovarian cancer. The USPSTF recommends against ovarian cancer
period. The average annual rate of referral for CA125 measurement screening in women at average risk (D recommendation).329 The ROCA
Screening and Early Detection  •  CHAPTER 23 395

screening test was marketed in the United States after publication Among all cancers, increasing BMI and obesity are most strongly
of the UKCTOCS findings in December 2015. In 2016, the FDA associated with endometrial cancer incidence and death.336 In a meta-
issued a Safety Communication, recommending against the use of analysis, Renehan and colleagues found that each increase in BMI of
ovarian cancer screening tests in the general population of women 5 kg/m2 significantly increased a woman’s risk of endometrial cancer by
and noting that testing higher-risk asymptomatic women for ovarian 59% (95% CI, 1.50–1.68).337 A large study of patients who underwent
cancer has no proven benefit and is not a substitute for preventive gastric bypass surgery revealed a 78% reduced risk of endometrial
actions that may reduce their risk.330 The ROCA test was volun- cancer (HR, 0.22 [95% CI, 0.13–0.40]), underscoring the profound
tarily withdrawn in the United States but remains available in the role of obesity in the development of this disease.338 Obesity has been
United Kingdom. associated with several factors known to increase endometrial cancer risk,
Women with a BRCA mutation or a family history suggestive of including upper body or central adiposity, PCOS, physical inactivity,
a mutation but as yet untested should be identified and counseled and a diet high in saturated fat.339 It has long been presumed that
regarding their ovarian cancer risk, surgical strategies to reduce their the peripheral conversion of androstenedione to estrone in adipose
risk, and screening options. Although there are no validated screening tissue resulted in excess circulating estrogen and subsequent risk of
methods for women at high risk of ovarian cancer, mutation carriers endometrial cancer in obese women. However, more recently, other
who have not undergone RRSO should be counseled about screening mechanisms have been suggested, including elevated serum insulin
with pelvic examination, TVUS, and CA125 every 6 months beginning levels and low adiponectin levels, a marker of insulin resistance that
at age 30 years or 5 to 10 years before the earliest age of diagnosis in has also been linked to endometrial cancer risk.336
the family.331 Screening is not a substitute for preventive surgery and In a meta-analysis of 33 studies, the average endometrial cancer
should continue only until RRSO is performed. Because these women risk reduction associated with high versus low physical activity was
are also at increased risk of breast cancer, they should be counseled 20%.340 There is some evidence that the association between physical
regarding breast cancer surgical risk reduction strategies and high-risk activity and endometrial cancer risk may reflect the effect of physical
breast cancer screening recommendations (see section on breast activity on obesity, a known risk factor for endometrial cancer.340–342
cancer screening). The aforementioned findings suggest an opportunity to reduce the
incidence of endometrial cancer by obesity management and increasing
ENDOMETRIAL CANCER physical activity.
In the National Surgical Adjuvant Breast and Bowel Project Breast
Endometrial cancer is the most commonly diagnosed gynecologic Cancer Prevention Trial, endometrial cancer risk was increased with
malignancy in women in the United States.18 Although most endo- use of tamoxifen.343 The annual rate was 2.3 per 1000 for women on
metrial cancers diagnosed in the United States are localized, suggesting tamoxifen versus 0.91 for those on placebo. Women older than 50
a limited value to screening, approximately one in four cases are years experienced the greatest effect. Raloxifene, a second-generation
advanced at the time of diagnosis. selective estrogen receptor modulator, does not have this association.344
Although Lynch syndrome (also called hereditary nonpolyposis
Risk Factors and Etiology colorectal cancer), typically manifests as familial clustering of early-onset
colon cancer, there is also an increased incidence of endometrial cancer
Conditions associated with prolonged progesterone deficiencies promote in women with Lynch syndrome. About 3% to 5% of endometrial
endometrial proliferation and an increased risk of endometrial cancers are attributable to the inherited mutations in the DNA
hyperplasia and cancer. Estrogen drives endometrial epithelial prolifera- mismatch repair (MMR) genes that cause Lynch syndrome.345 Lifetime
tion. Progesterone inhibits growth and causes cell differentiation. risk of endometrial cancer in women with MLH1 or MSH2 mutations
Progesterone has been described as the ultimate endometrial cancer is approximately 40%, with diagnosis at a median age of 49. Women
tumor suppressor.332 The importance of progesterone as a key inhibitor with MSH6 mutations have a similar risk of endometrial cancer but
of carcinogenesis is reflected by the observation that women who a later age of diagnosis.346 In a large series of women with Lynch
regularly ovulate and produce progesterone rarely get endometrial syndrome who developed both colorectal and gynecologic cancers,
cancer. In addition, studies have demonstrated regression of endometrial endometrial and/or ovarian cancer was, in half of the cases, the “sentinel
hyperplasia (both usual and atypical) by 80% to 90% after treatment cancer” preceding the development of colon cancer.347
with progestin.332 However, in premenopausal obese women, lack of
progesterone due to anovulation, such as that observed in polycystic Screening
ovarian syndrome (PCOS), increases a woman’s risk of endometrial
cancer. An Australian case-control study of women younger than 50 There are no RCTs that have demonstrated that screening with
years showed that women with PCOS had a fourfold greater risk of endometrial biopsy (EMB) and/or TVUS is superior to simply
endometrial cancer compared with women without PCOS. When performing a biopsy if abnormal uterine bleeding occurs. Given that
adjusted for BMI, PCOS remained an independent risk factor and most endometrial cancers are found at an early stage based on the
was associated with a greater than twofold increased risk of endometrial development of symptoms (e.g., abnormal uterine bleeding) and that
cancer.333 In addition, the use of unopposed estrogen is a well-established survival rates are high, the efficacy of endometrial cancer screening is
risk factor for the development of endometrial cancer. Addition of questionable. Studies of screening for endometrial cancer with EMB
progesterone to estrogen therapy negates this risk. and/or TVUS in asymptomatic women receiving tamoxifen or with
A review of 15 case-control studies and four large cohort studies Lynch syndrome348–351 reported disappointing results and associated
demonstrated a decrease in the risk of endometrial cancer of about harms including unnecessary EMBs in women with suspicious findings
50% for ever-use of OCs. An increasing protective effect with longer on TVUS, uterine perforations, and an increase in operative procedures.
duration of OC use has been found in most studies. This protective At this time, no organization recommends routine screening for
effect persisted for as long as 20 years after cessation of use.334 Increased endometrial cancer in average-risk women. In addition, screening is
parity has been shown to decrease risk of endometrial cancer. In not recommended for obese women, women with PCOS, women
EPIC, a 35% decreased risk of endometrial cancer was observed in taking tamoxifen or unopposed estrogen, or other women at increased
parous women (HR, 0.65 [95% CI, 0.54–0.77]) compared with risk. However, they should be informed about the risks and symptoms
nulliparous women, with a trend of decreasing risk with increasing of endometrial cancer and strongly encouraged to report any unexpected
number of full-term pregnancies. A shorter menstrual lifespan (late bleeding or spotting to their physicians.
menarche, early menopause) has been associated with a reduced risk of Although there are no validated screening methods for patients at
endometrial cancer.335 high risk of endometrial cancer, women with known or suspected
396 Part I: Science and Clinical Oncology

Lynch syndrome may be offered annual EMB beginning at age 30 to unquantifiable impact of self-selection both into SCREEN and for
35, pending definitive management with total hysterectomy with subsequent examinations, which were not mandated or tracked in
BSO.352,353 It has been shown that EMB performed at the time of the study.364
colonoscopy is a patient-centered option that decreases pain associated The trend of increased survival was short-lived and based on routine
with the biopsy and increases patient satisfaction.354 cause-of-death statistics in Schleswig-Holstein, rather than individual-
level data of screened versus unscreened individuals. This has been
SKIN CANCER questioned as a potential source of poor-quality data,365,366 and the
inability to link outcomes to individual-level data precludes firm
Skin cancer screening has traditionally been done by dermatologists establishment of causal relationships between increased screening and
and is considered a major distinguishing professional responsibility increased survival. In addition, the lack of TNM-based stage-specific
within the specialty. The accessibility of skin and the substantial burden data or subgroup data (e.g., in older men) makes it difficult to hone
of skin disease make it an ideal clinical setting for cancer screening in on the high-risk groups mostly likely to benefit quickly from
and prevention. That said, there is significant controversy regarding screening, although it presents opportunities for guiding future
how effective total body skin examination (TBSE) is in terms of studies.365,366 Finally, it is difficult, if not impossible, to deconvolute
improving survival rates in skin cancer.355,356 This is largely a result the potential for increased public awareness during the pre-SCREEN
of the difficulty with which evidence to support its widespread use advertising campaign to have increased self-selection into the ranks
in the general population can be gathered and is reflected in the recent of those screened during the study.367 Ultimately the absolute magnitude
USPSTF recommendation of I, or insufficient evidence, a reiteration of reduced mortality was modest, only 19% of the general population
of its previous rating from 2009.357 It is very important to recognize was screened, and a substantial number of patients were lost to follow-up
that this rating pertains to evidence for the benefit of screening (37%).361 Nevertheless, SCREEN was performed only for 1 year and
asymptomatic adults with no elevated risk of skin cancer and does mortality tracked for a 10-year period before and after the intervention.
not address individuals who have had a prior history of skin cancer It is possible that extending this effort over time would have yielded
or precancerous lesions.357,358 a more significant, larger, or sustained effect.367
The evidence that TBSE is effective in reducing skin cancer mortality Nevertheless, as a result of the SCREEN study, Germany adopted
is largely derived from two ecologic studies: one focused on employees the Screen Cancer Screening program for all adults older than age
at Lawrence Livermore National Laboratory (LLNL),359 and one 35, who since 2008 may undergo TBSE every 2 years.361,367 This
population-based study in Northern Germany.360,361 Understandably, nationwide screening effort has not shown a sustained decrease in
the focus has been on melanoma, which accounts for over 75% of melanoma mortality, and although direct comparisons have been made
deaths attributed to skin cancer, and the consensus is that melanoma to the initial SCREEN effort, there are important differences, including
survival is enhanced when lesions are diagnosed earlier. The end point older age of eligibility, lack of tracking of referral patterns, and lack
of skin cancer screening is often the selection of appropriate lesions of a public information campaign.367,368 The SCREEN study was
for biopsy, and measures of efficacy other than survival or mortality monumental in its scope, longevity, and cost and generated considerable
include the number of lesions that require biopsy per melanoma discussion in how such efforts should be studied in the future, especially
diagnosis, changes in cancer incidence, earlier stage of diagnoses, and with regard to who should be screened, evidentiary standards of risk
cost-effectiveness of avoiding management of more advanced disease. and benefits, and economic impact.
However, these end points are not used in formulating USPSTF
recommendations. Patient Populations

German SCREEN Study Even within the SCREEN study, it is clear that women and younger
individuals participated the most. This reduced men as a major source
The results of the landmark Skin Cancer Research to Provide Evidence of higher-risk disease, because there is good evidence that survival of
for Effectiveness of Screening in Northern Germany (SCREEN) study men with melanoma, even when controlled for stage, is significantly
have garnered significant attention, and this study is perhaps the inferior to that of women. This is thought to be partially due to less
one cited most frequently in the assessment of TBSE.360–362 In 2003, favorable clinicopathologic factors and older age at diagnosis, suggesting
98% of dermatologists and 64% of general practitioners and other that there may be a particularly positive impact of screening on men.62,369
specialists in the state of Schleswig-Holstein, Germany participated in an A comparison between TBSE and lesion-directed skin (LDS)
8-hour training course on TBSE; from July 1, 2003 to June 30, 2004, examination conducted in two areas of Belgium suggested that detection
360,288 adults aged 20 years or older (19% of the population) were rates for skin cancer were similar (2.3% for TBSE versus 3.2% for
screened for melanoma by TBSE in the state of Schleswig-Holstein.360,361 LDS examination), although TBSE identified more lesions overall.370
Initially, 84.5% were screened by nondermatologists, and 12.9% of The authors concluded that LDS examination might be a more practical
total participants were ultimately triaged for further evaluation by way to increase skin cancer surveillance without burdening a system
dermatologists. A total of 9.3% of screened individuals had skin lesions with a population-based effort.
suspicious for skin cancers, and ultimately 585 melanomas, 1961 basal Another model for this might be patterned on what has been
cell carcinomas, and 293 squamous cell carcinomas were identified recently reported in Australia, in which a high-risk melanoma clinic
through biopsy.360,361 was formed for biannual surveillance of patients with a personal or
In the period from 1980 to 2002, melanoma mortality in Schleswig- strong family history of melanoma, including those with high-penetrance
Holstein was significantly higher than the German average by over 24%, germline mutations. This study identified significant economic and
but in 2002, this decreased significantly. In the 5-year period following quality-adjusted life-year (QALY) benefit as compared with similar
SCREEN, from 2003 to 2008, a 50% reduction in mortality was patients receiving standard care in the community, but did not assess
observed, corresponding to a decrease of 0.8 death per 100,000.360,361 survival.371
Additional analyses demonstrated that melanoma incidence increased
shortly after SCREEN, as did incidences of NMSCs.363 Analysis of Evidentiary Standards and End Points Other
interval melanomas between a negative examination result at SCREEN Than Survival
and a subsequent regular screening in over 350,000 individuals
showed a moderate 29% reduction in invasive melanomas, but a There are several reasons why cancer screening may not translate into
61% increase in in situ melanomas. Possible reasons for this increase a meaningful increase in overall survival, although there is evidence
are difficult to identify, given that overall incidences rose and the that diagnoses of melanoma at earlier stages confers improved cancer-free
Screening and Early Detection  •  CHAPTER 23 397

survival.372 It has also been argued that improved survival is not the Given that cutaneous SCCs have an appreciable metastatic rate and
only meaningful end point of screening. Screening may decrease NMSCs account for over 3.5 million cases of cancer in 2.2 million
morbidity by virtue of permitting less extensive management (e.g., individuals with an annual treatment cost of over $4.8 billion in the
avoiding sentinel lymph node biopsy). Screening also may have collateral United States alone,373,374 the promise of ameliorating the overall burden
benefits of increased education and awareness about avoidable risk of disease would seem to be of benefit. Viewing the increased diagnosis
factors among patients and their family members.372 and presumably earlier treatment of these diseases as a potential harm
The costs of screening for melanoma and economic benefits of precludes a more nuanced perspective on what could be regarded as
treating earlier disease are not often studied. Although the Belgian a collateral benefit.
experience with LDS examination had calculated a cost of $8000 per
melanoma diagnosis,370 analysis of QALYs and years of life saved have The Way Forward
been conducted for melanoma screening.372 Many of these studies
have suggested that focusing on high-risk populations is more The general consensus appears to be that a randomized clinical trial
cost-effective.372 for skin cancer screening is unlikely to be practical. In this regard,
Although major therapeutic gains have been made for advanced the USPTF statement on skin cancer is forward-looking because
disease, the economic burden of targeted and immune-therapies is it opens the door to case-control designs that are more practical,
likely to increase enormously, making it impractical to ignore potential while acknowledging that strong evidence of benefit and harms are
savings of screening versus therapy, especially since these burdens can lacking, including for NMSC. The people most likely to benefit from
be devastating for patients. skin cancer screening are those at high risk by virtue of familial
syndromes or, more commonly, a history of skin cancer or precancerous
Overdiagnosis and Harms of Screening lesions, and the evidence review panel for the USPSTF cited this as
an appropriate area of focus in the future.375 Although the panel
The major harms cited by the panel were overdiagnosis and the risk recognized, for example, that individuals with high numbers of nevi
of poor cosmetic outcomes, although the latter issue is poorly studied. are at elevated risk for melanoma, the way to detect these individuals
The trend of increasing melanoma incidence has been reported to be is through TBSE.
matched by the increase in biopsy rates. This trend was interpreted The SCREEN study, among others, demonstrated the importance
as due to overdiagnosis by the Task Force, which then raised the of a standardized curriculum to boost the sensitivity and specificity
concern that a screening effort would exacerbate this problem.357 Factors of screeners, particularly nondermatologists, who perform TBSE,376
such as pressure to detect lesions earlier and diagnostic drift have been and the Belgian experience suggests that a combination of self-directed
suggested and may well represent nonmedical (e.g., medicolegal) initial screenings370 may be an effective way of balancing the high cost
concerns that do not necessarily reflect an improved understanding of population-based screening with access to specialists. Ultimately,
of the underlying biology of cancer. Conversely, the expected increase screening must fit into a comprehensive cost-effective medical system,
in cancer incidence as a result of screening may only be temporary and benefits that include decreased morbidity, increased QALYs, and
because most individuals will be of average (low) risk and are unlikely treatment savings should be considered alongside survival.
to be continuously screened at close intervals.
An additional harm that was suggested in the 2009 statement but The complete reference list is available online at
not in the 2016 one is the diagnosis of often nonlethal NMSCs.367 ExpertConsult.com.

KEY REFERENCES
4. Iyengar NM, Gucalp A, Dannenberg AJ, Hudis management of IPMN and MCN of the pancreas. pylori vaccine in children in China: a randomised,
CA. Obesity and cancer mechanisms: tumor Pancreatology. 2012;12(3):183–197. double-blind, placebo-controlled, phase 3 trial.
microenvironment and inflammation. J Clin Oncol. 175. El-Serag HB, Kanwal F, Davila JA, Kramer J, Lancet. 2015;386(10002):1457–1464.
2016;34(35):4270–4276. Richardson P. A new laboratory-based algorithm to 246. Vaughan TL, Fitzgerald RC. Precision prevention of
19. Gail MH, Brinton LA, Byar DP, et al. Projecting indi- predict development of hepatocellular carcinoma in oesophageal adenocarcinoma. Nat Rev Gastroenterol
vidualized probabilities of developing breast cancer patients with hepatitis C and cirrhosis. Gastroenterol- Hepatol. 2015;12(4):243–248.
for white females who are being examined annually. ogy. 2014;146(5):1249–1255. 248. Taylor PR, Abnet CC, Dawsey SM. Squamous
J Natl Cancer Inst. 1989;81(24):1879–1886. 188. Blais P, Husain N, Kramer JR, Kowalkowski M, El- dysplasia—the precursor lesion for esophageal squa-
69. Imperiale TF, Ransohoff DF, Itzkowitz SH, et al. Serag H, Kanwal F. Nonalcoholic fatty liver disease mous cell carcinoma. Cancer Epidemiol Biomarkers
Multitarget stool DNA testing for colorectal-cancer is underrecognized in the primary care setting. Am Prev. 2013;22(4):540–552.
screening. N Engl J Med. 2014;370(14):1287–1297. J Gastroenterol. 2015;110(1):10–14. 259. El-Serag HB, Naik AD, Duan Z, et al. Surveillance
89. Saslow D, Solomon D, Lawson HW, et al. American 217. Plummer M, Franceschi S, Vignat J, Forman D, endoscopy is associated with improved outcomes
Cancer Society, American Society for Colposcopy de Martel C. Global burden of gastric cancer of oesophageal adenocarcinoma detected in patients
and Cervical Pathology, and American Society for attributable to Helicobacter pylori. Int J Cancer. 2015; with Barrett’s oesophagus. Gut. 2016;65(8):1252–
Clinical Pathology screening guidelines for the 136(2):487–490. 1260.
prevention and early detection of cervical cancer. 225. World Cancer Research Fund/American Institute for 275. Ferlay J, Soerjomataram I, Dikshit R, et al. Cancer
CA Cancer J Clin. 2012;62(3):147–172. Cancer Research (WCRF/AICR). Food, Nutrition, incidence and mortality worldwide: sources, methods
121. Fenton J, Weyrich M, Durbin S, Liu Y, Bang Physical Activity, and the Prevention of Cancer: a and major patterns in GLOBOCAN 2012. Int J
H, Melnikow J. Prostate-Specific Antigen-Based Global Perspective. Washington DC: AICR 2007. Cancer. 2015;136(5):E359–E386.
Screening for Prostate Cancer: A Systematic Evidence 231. Asaka M, Kato M, Sakamoto N. Roadmap to 280. Blot WJ, McLaughlin JK, Winn DM, et al. Smoking
Review for the US Preventive Services Task Force. eliminate gastric cancer with Helicobacter pylori and drinking in relation to oral and pharyngeal
Rockville, MD: Agency for Healthcare Research and eradication and consecutive surveillance in Japan. cancer. Cancer Res. 1988;48(11):3282–3287.
Quality; 2017. J Gastroenterol. 2014;49(1):1–8. 284. Castellsagué X, Alemany L, Quer M, et al. HPV
130. Andriole GL, Crawford ED, Grubb RL, et al. 238. Ford AC, Forman D, Hunt RH, Yuan Y, Moayyedi involvement in head and neck cancers: compre-
Prostate cancer screening in the randomized Prostate, P. Helicobacter pylori eradication therapy to prevent hensive assessment of biomarkers in 3680 patients.
Lung, Colorectal, and Ovarian Cancer Screening gastric cancer in healthy asymptomatic infected J Natl Cancer Inst. 2016;108(6):djv403.
Trial: mortality results after 13 years of follow-up. individuals: systematic review and meta-analysis of 303. Domchek SM, Friebel TM, Singer CF, et al. Associa-
J Natl Cancer Inst. 2012;104(2):125–132. randomised controlled trials. BMJ. 2014;348:g3174. tion of risk-reducing surgery in BRCA1 or BRCA2
148. Tanaka M, Fernandez-del Castillo C, Adsay V, et al. 240. Zeng M, Mao XH, Li JX, et al. Efficacy, safety, and mutation carriers with cancer risk and mortality.
International consensus guidelines 2012 for the immunogenicity of an oral recombinant Helicobacter JAMA. 2010;304(9):967–975.
398 Part I: Science and Clinical Oncology

322. Pinsky PF, Yu K, Kramer BS, et al. Extended mortal- 343. Fisher B, Costantino JP, Wickerham DL, et al. 361. Breitbart EW, Waldmann A, Nolte S, et al. Systematic
ity results for ovarian cancer screening in the PLCO Tamoxifen for prevention of breast cancer: report skin cancer screening in Northern Germany. J Am
trial with median 15 years follow-up. Gynecol Oncol. of the National Surgical Adjuvant Breast and Bowel Acad Dermatol. 2012;66(2):201–211.
2016;143(2):270–275. Project P-1 Study. J Natl Cancer Inst. 1998;90(18): 366. Stang A, Jockel KH. Does skin cancer screening
337. Renehan AG, Tyson M, Egger M, Heller RF, Zwahlen 1371–1388. save lives? A detailed analysis of mortality time
M. Body-mass index and incidence of cancer: a 344. Vogel VG, Costantino JP, Wickerham DL, et al. trends in Schleswig-Holstein and Germany. Cancer.
systematic review and meta-analysis of prospective Effects of tamoxifen vs raloxifene on the risk of 2016;122(3):432–437.
observational studies. Lancet. 2008;371(9612): developing invasive breast cancer and other disease 370. Hoorens I, Vossaert K, Pil L, et al. Total-body
569–578. outcomes: the NSABP Study of Tamoxifen and examination vs lesion-directed skin cancer screening.
338. Adams TD, Stroup AM, Gress RE, et al. Cancer Raloxifene (STAR) P-2 trial. JAMA. 2006;295(23): JAMA Dermatol. 2016;152(1):27–34.
incidence and mortality after gastric bypass surgery. 2727–2741.
Obesity (Silver Spring). 2009;17(4):796–802.
Screening and Early Detection  •  CHAPTER 23 398.e1
398.e1

REFERENCES
1. Global Burden of Disease Cancer Collaboration, 22. Tice JA, O’Meara ES, Weaver DL, Vachon C, Ballard- guideline update from the American Cancer Society.
Fitzmaurice C, Allen C, et al. Global, regional, and Barbash R, Kerlikowske K. Benign breast disease, JAMA. 2015;314(15):1599–1614.
national cancer incidence, mortality, years of life lost, mammographic breast density, and the risk of breast 41. Siu AL, Force USPST. Screening for breast cancer:
years lived with disability, and disability-adjusted cancer. J Natl Cancer Inst. 2013;105(14):1043–1049. U.S. Preventive Services Task Force recommendation
life-years for 32 cancer groups, 1990 to 2015: a 23. Tice JA, Miglioretti DL, Li CS, Vachon CM, statement. Ann Intern Med. 2016;164(4):279–296.
systematic analysis for the Global Burden of Disease Gard CC, Kerlikowske K. Breast density and 42. Barth RJ, Gibson GR, Carney PA, Mott LA,
Study. JAMA Oncol. 2016. benign breast disease: risk assessment to identify Becher RD, Poplack SP. Detection of breast cancer
2. Levin PB, B. International Agency for Research on women at high risk of breast cancer. J Clin Oncol. on screening mammography allows patients to be
Cancer. World Cancer Report, 2008. Lyon, France: 2015;33(28):3137–3143. treated with less-toxic therapy. AJR Am J Roentgenol.
International Agency for Research on Cancer; 2008. 24. Tyrer J, Duffy SW, Cuzick J. A breast cancer predic- 2005;184(1):324–329.
ISBN 978 92 832 04237. tion model incorporating familial and personal risk 43. Zorzi M, Puliti D, Vettorazzi M, et al. Mastectomy
3. Nimptsch K, Pischon T. Obesity Biomarkers, metab- factors. Stat Med. 2004;23(7):1111–1130. rates are decreasing in the era of service screening:
olism and risk of cancer: an epidemiological perspec- 25. Berry DA, Parmigiani G, Sanchez J, Schildkraut a population-based study in Italy (1997-2001). Br
tive. Recent Results Cancer Res. 2016;208:199–217. J, Winer E. Probability of carrying a mutation of J Cancer. 2006;95(9):1265–1268.
4. Iyengar NM, Gucalp A, Dannenberg AJ, Hudis breast-ovarian cancer gene BRCA1 based on family 44. Malmgren JA, Parikh J, Atwood MK, Kaplan HG.
CA. Obesity and cancer mechanisms: tumor history. J Natl Cancer Inst. 1997;89(3):227–238. Impact of mammography detection on the course of
microenvironment and inflammation. J Clin Oncol. 26. Parmigiani G, Berry D, Aguilar O. Determining breast cancer in women aged 40-49 years. Radiology.
2016;34(35):4270–4276. carrier probabilities for breast cancer-susceptibility 2012;262(3):797–806.
5. Meerzaman D, Dunn BK, Lee M, Chen Q, Yan C, genes BRCA1 and BRCA2. Am J Hum Genet. 1998; 45. Malmgren JA, Parikh J, Atwood MK, Kaplan
Ross S. The promise of omics-based approaches to 62(1):145–158. HG. Improved prognosis of women aged 75 and
cancer prevention. Semin Oncol. 2016;43(1):36–48. 27. Berry DA, Iversen ES, Gudbjartsson DF, et al. older with mammography-detected breast cancer.
6. Foulkes WD, Knoppers BM, Turnbull C. Population BRCAPRO validation, sensitivity of genetic Radiology. 2014;273(3):686–694.
genetic testing for cancer susceptibility: founder muta- testing of BRCA1/BRCA2, and prevalence of other 46. Moss SM, Wale C, Smith R, Evans A, Cuckle H,
tions to genomes. Nat Rev Clin Oncol. 2016;13(1): breast cancer susceptibility genes. J Clin Oncol. Duffy SW. Effect of mammographic screening
41–54. 2002;20(11):2701–2712. from age 40 years on breast cancer mortality in the
7. Bombard Y, Bach PB, Offit K. Translating genomics 28. Claus EB, Risch N, Thompson WD. Autosomal UK Age trial at 17 years’ follow-up: a randomised
in cancer care. J Natl Compr Canc Netw. 2013; dominant inheritance of early-onset breast cancer. controlled trial. Lancet Oncol. 2015;16(9):1123–
11(11):1343–1353. Implications for risk prediction. Cancer. 1994;73(3): 1132.
8. Walsh MF, Nathanson KL, Couch FJ, Offit K. 643–651. 47. Miller AB, Wall C, Baines CJ, Sun P, To T, Narod
Genomic biomarkers for breast cancer risk. Adv 29. Couch FJ, DeShano ML, Blackwood MA, et al. SA. Twenty five year follow-up for breast cancer
Exp Med Biol. 2016;882:1–32. BRCA1 mutations in women attending clinics that incidence and mortality of the Canadian National
9. Wilson J, Jungner G. Principles and Practice of evaluate the risk of breast cancer. N Engl J Med. Breast Screening Study: randomised screening trial.
Screening for Disease. In. Geneva, Switzerland: 1997;336(20):1409–1415. BMJ. 2014;348:g366.
World Health Organization; 1968. 30. Antoniou AC, Pharoah PD, McMullan G, et al. 48. Broeders M, Moss S, Nyström L, et al. The impact of
10. Gill H, Wu J. Prostate specific antigen: the past, A comprehensive model for familial breast cancer mammographic screening on breast cancer mortality
present and future. Curr Urol. 2013;6(4):175–178. incorporating BRCA1, BRCA2 and other genes. Br in Europe: a review of observational studies. J Med
11. Carter HB, Albertsen PC, Barry MJ, et al. Early J Cancer. 2002;86(1):76–83. Screen. 2012;19(suppl 1):14–25.
detection of prostate cancer: AUA Guideline. J Urol. 31. Antoniou AC, Pharoah PP, Smith P, Easton DF. 49. Mandelblatt JS, Cronin KA, Bailey S, et al. Effects
2013;190(2):419–426. The BOADICEA model of genetic susceptibility to of mammography screening under different screening
12. Barocas DA, Mallin K, Graves AJ, et al. Effect of breast and ovarian cancer. Br J Cancer. 2004;91(8): schedules: model estimates of potential benefits and
the USPSTF Grade D Recommendation against 1580–1590. harms. Ann Intern Med. 2009;151(10):738–747.
screening for prostate cancer on incident prostate 32. Antoniou AC, Cunningham AP, Peto J, et al. The 50. The American Congress of Obstetricians and
cancer diagnoses in the United States. J Urol. 2015; BOADICEA model of genetic susceptibility to breast Gynecologists (ACOG). ACOG Statement on Breast
194(6):1587–1593. and ovarian cancers: updates and extensions. Br J Cancer Screening Guidelines; 2016. www.acog.org/
13. Kearney AJ. Viewpoint: it is time to reconsider policy Cancer. 2008;98(8):1457–1466. About-ACOG/News-Room/Statements/2016/
for population-based mammography screening. 33. Thomas DB, Gao DL, Self SG, et al. Random- ACOG-Statement-on-Breast-Cancer-Screening
J Public Health Policy. 2015;36(3):259–269. ized trial of breast self-examination in Shanghai: -Guidelines.
14. Lauby-Secretan B, Scoccianti C, Loomis D, et al. methodology and preliminary results. J Natl Cancer 51. American College of Radiology. American College
Breast-cancer screening—viewpoint of the IARC Inst. 1997;89(5):355–365. of Radiology Position Statement on Screening
Working Group. N Engl J Med. 2015;372(24): 34. Thomas DB, Gao DL, Ray RM, et al. Randomized Mammography and Health Care Coverage. In: 2016.
2353–2358. trial of breast self-examination in Shanghai: final 52. Wilt TJ, Harris RP, Qaseem A, Physicians HVCT-
15. Wise J. Breast screening benefits have been overstated, results. J Natl Cancer Inst. 2002;94(19):1445–1457. FotACo. Screening for cancer: advice for high-value
Danish study finds. BMJ. 2017;356:j94. 35. Barton MB, Harris R, Fletcher SW. The rational care from the American College of Physicians. Ann
16. Hall AE, Chowdhury S, Hallowell N, et al. Imple- clinical examination. Does this patient have breast Intern Med. 2015;162(10):718–725.
menting risk-stratified screening for common cancers: cancer? The screening clinical breast examination: 53. Roder D, Houssami N, Farshid G, et al. Population
a review of potential ethical, legal and social issues. should it be done? How? JAMA. 1999;282(13): screening and intensity of screening are associated
J Public Health (Oxf ). 2014;36(2):285–291. 1270–1280. with reduced breast cancer mortality: evidence of
17. Shieh Y, Eklund M, Madlensky L, et al. Breast cancer 36. National Comprehensive Cancer Network (NCCN). efficacy of mammography screening in Australia.
screening in the precision medicine era: risk-based The NCCN Clinical Practice Guidelines in Oncol- Breast Cancer Res Treat. 2008;108(3):409–416.
screening in a population-based trial. J Natl Cancer ogy: Breast Cancer Screening and Diagnosis (Version 54. Mandelblatt JS, Stout NK, Schechter CB, et al.
Inst. 2017;109(5). 2.2016). In: National Comprehensive Cancer Collaborative modeling of the benefits and harms
18. American Cancer Society (ACS). Cancer Facts & Network; 2016. associated with different U.S. breast cancer screening
Figures; 2017. Atlanta, GA2017. 37. Marmot MG, Altman DG, Cameron DA, Dewar strategies. Ann Intern Med. 2016;164(4):215–225.
19. Gail MH, Brinton LA, Byar DP, et al. Projecting JA, Thompson SG, Wilcox M. The benefits and 55. Arleo EK, Monticciolo DL, Monsees B, McGinty G,
individualized probabilities of developing breast harms of breast cancer screening: an independent Sickles EA. Persistent untreated screening-detected
cancer for white females who are being examined review. Br J Cancer. 2013;108(11):2205–2240. breast cancer: an argument against delaying screening
annually. J Natl Cancer Inst. 1989;81(24):1879–1886. 38. Myers ER, Moorman P, Gierisch JM, et al. Benefits or increasing the interval between screenings. J Am
20. Costantino JP, Gail MH, Pee D, et al. Validation and harms of breast cancer screening: a systematic Coll Radiol. 2017.
studies for models projecting the risk of invasive review. JAMA. 2015;314(15):1615–1634. 56. Ciatto S, Houssami N, Bernardi D, et al. Integration
and total breast cancer incidence. J Natl Cancer 39. Nelson H, Cantor A, Humphrey L, et al. Screening of 3D digital mammography with tomosynthesis
Inst. 1999;91(18):1541–1548. for Breast Cancer: A Systematic Review to Update for population breast-cancer screening (STORM):
21. Tice JA, Cummings SR, Smith-Bindman R, the 2009 U.S. Preventive Services Task Force a prospective comparison study. Lancet Oncol.
Ichikawa L, Barlow WE, Kerlikowske K. Using Recommendation. Evidence Synthesis No. 124. 2013;14(7):583–589.
clinical factors and mammographic breast density Rockville, MD: Agency for Healthcare Research 57. Skaane P, Bandos AI, Gullien R, et al. Comparison
to estimate breast cancer risk: development and and Quality;2016. of digital mammography alone and digital mam-
validation of a new predictive model. Ann Intern 40. Oeffinger KC, Fontham ET, Etzioni R, et al. Breast mography plus tomosynthesis in a population-based
Med. 2008;148(5):337–347. cancer screening for women at average risk: 2015 screening program. Radiology. 2013;267(1):47–56.
398.e2 Part I: Science and Clinical Oncology

58. Friedewald SM, Rafferty EA, Rose SL, et al. Breast 79. Schiffman M, Castle PE, Jeronimo J, Rodriguez AC, diseases in China: time for urgent action. Lancet.
cancer screening using tomosynthesis in combina- Wacholder S. Human papillomavirus and cervical 2016;388(10054):1939–1951.
tion with digital mammography. JAMA. 2014; cancer. Lancet. 2007;370(9590):890–907. 99. Dai J, Yang P, Cox A, Jiang G. Lung cancer and
311(24):2499–2507. 80. Chesson HW, Dunne EF, Hariri S, Markowitz LE. chronic obstructive pulmonary disease: from a
59. Destounis S, Arieno A, Morgan R. Initial experience The estimated lifetime probability of acquiring clinical perspective. Oncotarget. 2017.
with combination digital breast tomosynthesis plus human papillomavirus in the United States. Sex 100. Marshall AL, Christiani DC. Genetic susceptibility
full field digital mammography or full field digital Transm Dis. 2014;41(11):660–664. to lung cancer—light at the end of the tunnel?
mammography alone in the screening environment. 81. Wheeler CM, Hunt WC, Joste NE, Key CR, Quint Carcinogenesis. 2013;34(3):487–502.
J Clin Imaging Sci. 2014;4:9. WG, Castle PE. Human papillomavirus genotype 101. Tammemagi MC. Application of risk prediction
60. Saslow D, Boetes C, Burke W, et al. American Cancer distributions: implications for vaccination and cancer models to lung cancer screening: a review. J Thorac
Society guidelines for breast screening with MRI as screening in the United States. J Natl Cancer Inst. Imaging. 2015;30(2):88–100.
an adjunct to mammography. CA Cancer J Clin. 2009;101(7):475–487. 102. Spitz MR, Etzel CJ, Dong Q, et al. An expanded
2007;57(2):75–89. 82. Bosch FX, de Sanjosé S. Chapter 1: human risk prediction model for lung cancer. Cancer Prev
61. National Comprehensive Cancer Network (NCCN). papillomavirus and cervical cancer—burden and Res (Phila). 2008;1(4):250–254.
The NCCN Clinical Practice Guidelines in Oncol- assessment of causality. J Natl Cancer Inst Monogr. 103. Marcus MW, Chen Y, Raji OY, Duffy SW, Field
ogy: Genetic/Familial High-Risk Assessment: 2003;31:3–13. JK. LLPi: Liverpool Lung Project Risk Prediction
Breast and Ovarian (Version 2.2017). In: National 83. Vesco KK, Whitlock EP, Eder M, Burda BU, Senger Model for Lung Cancer Incidence. Cancer Prev Res
Comprehensive Cancer Network; 2017. CA, Lutz K. Risk factors and other epidemiologic (Phila). 2015;8(6):570–575.
62. American Cancer Society. Cancer Facts & Figures considerations for cervical cancer screening: a nar- 104. Sherratt FC, Marcus MW, Robinson J, Field
2018. Atlanta: American Cancer Society; 2018. rative review for the U.S. Preventive Services Task JK. Utilizing lung cancer risk prediction models
63. World Health Organization, International Agency Force. Ann Intern Med. 2011;155(10):698–705, to promote smoking cessation: two randomized
for Research on Cancer(IARC). Globocan; 2012. W216. controlled trials. Am J Health Promot. 2016.
64. Freedman AN, Slattery ML, Ballard-Barbash R, et al. 84. Exposure in utero to diethylstilbestrol and related 105. Tammemagi MC, Church TR, Hocking WG,
Colorectal cancer risk prediction tool for white men synthetic hormones. Association with vaginal and et al. Evaluation of the lung cancer risks at which
and women without known susceptibility. J Clin cervical cancers and other abnormalities. JAMA. to screen ever- and never-smokers: screening rules
Oncol. 2009;27(5):686–693. 1976;236(10):1107–1109. applied to the PLCO and NLST cohorts. PLoS
65. Park Y, Freedman AN, Gail MH, et al. Validation 85. National Cancer Institute(NCI). Diethylstilbes- Med. 2014;11(12):e1001764.
of a colorectal cancer risk prediction model among trol (DES) and Cancer; 2011. www.cancer.gov/ 106. Tammemagi CM, Pinsky PF, Caporaso NE, et al.
white patients age 50 years and older. J Clin Oncol. about-cancer/causes-prevention/risk/hormones/ Lung cancer risk prediction: prostate, lung, colorectal
2009;27(5):694–698. des-fact-sheet. Accessed March, 2017. and ovarian cancer screening trial models and valida-
66. Imperiale TF, Yu M, Monahan PO, et al. Risk 86. Verloop J, van Leeuwen FE, Helmerhorst TJM, van tion. J Natl Cancer Inst. 2011;103(13):1058–1068.
of advanced neoplasia using the National Cancer Boven HH, Rookus MA. Cancer risk in DES daugh- 107. Muller DC, Johansson M, Brennan P. Lung
Institute’s Colorectal Cancer Risk Assessment Tool. ters. Cancer Causes Control. 2010;21(7):999–991007. cancer risk prediction model incorporating lung
J Natl Cancer Inst. 2017;109(1). 87. Whitlock EP, Vesco KK, Eder M, Lin JS, Senger function: development and validation in the UK
67. Lin J, Piper M, Perdue L, et al. Screening for CA, Burda BU. Liquid-based cytology and human Biobank Prospective Cohort Study. J Clin Oncol.
Colorectal Cancer: A Systematic Review for the U.S. papillomavirus testing to screen for cervical cancer: 2017;JCO2016692467.
Preventive Services Task Force. Evidence Synthesis a systematic review for the U.S. Preventive Services 108. Timofeeva MN, McKay JD, Smith GD, et al. Genetic
No. 135. Rockville, MD: Agency for Healthcare Task Force. Ann Intern Med. 2011;155(10):687–697, polymorphisms in 15q25 and 19q13 loci, cotinine
Research and Quality;2016. W214–W685. levels, and risk of lung cancer in EPIC. Cancer
68. Ladabaum U, Levin Z, Mannalithara A, Brill JV, 88. Castle PE, Glass AG, Rush BB, et al. Clinical human Epidemiol Biomarkers Prev. 2011;20(10):2250–2261.
Bundorf MK. Colorectal testing utilization and papillomavirus detection forecasts cervical cancer 109. Sin DD, Tammemagi CM, Lam S, et al. Pro-
payments in a large cohort of commercially insured risk in women over 18 years of follow-up. J Clin surfactant protein B as a biomarker for lung cancer
US adults. Am J Gastroenterol. 2014;109(10): Oncol. 2012;30(25):3044–3050. prediction. J Clin Oncol. 2013;31(36):4536–4543.
1513–1525. 89. Saslow D, Solomon D, Lawson HW, et al. American 110. Wikoff WR, Hanash S, DeFelice B, et al. Diacetyl-
69. Imperiale TF, Ransohoff DF, Itzkowitz SH, et al. Cancer Society, American Society for Colposcopy spermine is a novel prediagnostic serum biomarker
Multitarget stool DNA testing for colorectal-cancer and Cervical Pathology, and American Society for for non-small-cell lung cancer and has additive
screening. N Engl J Med. 2014;370(14):1287–1297. Clinical Pathology screening guidelines for the performance with pro-surfactant protein B. J Clin
70. Zauber A, Knudsen A, Rutter CM, Lansdorp- prevention and early detection of cervical cancer. Oncol. 2015;33(33):3880–3886.
Vogelaar I, Kuntz KM. Evaluating the Benefits and CA Cancer J Clin. 2012;62(3):147–172. 111. Facts ACSC, Figures ACS, Atlanta. American Cancer
Harms of Colorectal Cancer Screening Strategies: A 90. Moyer VA. Screening for cervical cancer: U.S. Society, Atlanta (2015). American Cancer Society;
Collaborative Modeling Approach. Rockville, MD: Preventive Services Task Force recommendation 2015.
Agency for Healthcare Research and Quality; 2015. statement. Ann Intern Med. 2012;156(12):880–891. 112. Boucot KR, Weiss W. Is curable lung cancer detected
71. Inadomi JM. Screening for colorectal neoplasia. N 91. Cox JT, Castle PE, Behrens CM, et al. Comparison by semiannual screening? JAMA. 1973;224(10):
Engl J Med. 2017;376(2):149–156. of cervical cancer screening strategies incorporating 1361–1365.
72. Holme Ø, Løberg M, Kalager M, et al. Effect of different combinations of cytology, HPV testing, 113. Marcus PM, Bergstralh EJ, Fagerstrom RM, et al.
flexible sigmoidoscopy screening on colorectal cancer and genotyping for HPV 16/18: results from Lung cancer mortality in the Mayo Lung Project:
incidence and mortality: a randomized clinical trial. the ATHENA HPV study. Am J Obstet Gynecol. impact of extended follow-up. J Natl Cancer Inst.
JAMA. 2014;312(6):606–615. 2013;208(3):184.e1–184.e11. 2000;92(16):1308–1316.
73. Centers for Disease Control and Prevention(CDC). 92. Wright TC, Stoler MH, Behrens CM, Sharma A, 114. Henschke CI, Yankelevitz DF, Libby DM, et al.
Vital signs: colorectal cancer screening test use— Zhang G, Wright TL. Primary cervical cancer screen- Early lung cancer action project: annual screening
United States, 2012. MMWR Morb Mortal Wkly ing with human papillomavirus: end of study results using single-slice helical CT. Ann N Y Acad Sci.
Rep. 2013;62(44):881–888. from the ATHENA study using HPV as the first-line 2001;952:124–134.
74. Nishihara R, Wu K, Lochhead P, et al. Long-term screening test. Gynecol Oncol. 2015;136(2):189–197. 115. Hillman BJ, ACRIN. Economic, legal, and ethical
colorectal-cancer incidence and mortality after lower 93. Huh WK, Ault KA, Chelmow D, et al. Use of rationales for the ACRIN national lung screening
endoscopy. N Engl J Med. 2013;369(12):1095–1105. primary high-risk human papillomavirus testing for trial of CT screening for lung cancer. Acad Radiol.
75. Kaminski MF, Bretthauer M, Zauber AG, et al. cervical cancer screening: interim clinical guidance. 2003;10(3):349–350.
The NordICC Study: rationale and design of a Obstet Gynecol. 2015;125(2):330–337. 116. Henschke CI, Yip R, Yankelevitz DF, Smith JP.
randomized trial on colonoscopy screening for 94. Torre LA, Siegel RL, Jemal A. Lung cancer statistics. International Early Lung Cancer Action Program
colorectal cancer. Endoscopy. 2012;44(7):695–702. Adv Exp Med Biol. 2016;893:1–19. I. Definition of a positive test result in computed
76. Berrington de González A, Kim KP, Knudsen 95. Chen W, Zheng R, Baade PD, et al. Cancer statistics tomography screening for lung cancer: a cohort
AB, et al. Radiation-related cancer risks from CT in China, 2015. CA Cancer J Clin. 2016;66(2): study. Ann Intern Med. 2013;158(4):246–252.
colonography screening: a risk-benefit analysis. AJR 115–132. 117. Sui X, Meinel FG, Song W, et al. Detection and size
Am J Roentgenol. 2011;196(4):816–823. 96. Malhotra J, Malvezzi M, Negri E, La Vecchia C, measurements of pulmonary nodules in ultra-low-
77. Scarinci IC, Garcia FAR, Kobetz E, et al. Cervical Boffetta P. Risk factors for lung cancer worldwide. dose CT with iterative reconstruction compared to
cancer prevention: new tools and old barriers. Cancer. Eur Respir J. 2016;48(3):889–902. low dose CT. Eur J Radiol. 2016;85(3):564–570.
2010;116(11):2531–2542. 97. Schwartz AG, Cote ML. Epidemiology of lung 118. Nagatani Y, Takahashi M, Murata K, et al. Lung
78. Trottier H, Franco EL. The epidemiology of cancer. Adv Exp Med Biol. 2016;893:21–41. nodule detection performance in five observers on
genital human papillomavirus infection. Vaccine. 98. Guan WJ, Zheng XY, Chung KF, Zhong NS. Impact computed tomography (CT) with adaptive iterative
2006;24(suppl 1):S1–S15. of air pollution on the burden of chronic respiratory dose reduction using three-dimensional processing
Screening and Early Detection  •  CHAPTER 23 398.e3
398.e3

(AIDR 3D) in a Japanese multicenter study: com- 137. Hamdy FC, Donovan JL, Lane JA, et al. 10-Year 156. Stevens RJ, Roddam AW, Beral V. Pancreatic cancer
parison between ultra-low-dose CT and low-dose outcomes after monitoring, surgery, or radio- in type 1 and young-onset diabetes: systematic review
CT by receiver-operating characteristic analysis. Eur therapy for localized prostate cancer. N Engl J Med. and meta-analysis. Br J Cancer. 2007;96(3):507–509.
J Radiol. 2015;84(7):1401–1412. 2016;375(15):1415–1424. 157. Huxley R, Ansary-Moghaddam A, Berrington de
119. Manos D, Seely JM, Taylor J, Borgaonkar J, Roberts 138. Routine aspirin or nonsteroidal anti-inflammatory Gonzalez A, Barzi F, Woodward M. Type-II diabetes
HC, Mayo JR. The Lung Reporting and Data System drugs for the primary prevention of colorectal cancer: and pancreatic cancer: a meta-analysis of 36 studies.
(LU-RADS): a proposal for computed tomography U.S. Preventive Services Task Force recommendation Br J Cancer. 2005;92(11):2076–2083.
screening. Can Assoc Radiol J. 2014;65(2):121–134. statement. Ann Intern Med. 2007;146(5):361–364. 158. Gapstur SM, Gann PH, Lowe W, Liu K, Colangelo L,
120. National Cancer Institute (NCI). SEER Cancer 139. U.S. Preventive Services Task Force (USPSTF). Dyer A. Abnormal glucose metabolism and pancreatic
Statistics Fact Sheet: Prostate Cancer. Accessed Draft Recommendation Statement: Prostate Cancer: cancer mortality. JAMA. 2000;283(19):2552–2558.
April 25, 2017. Screening. 2017; 2017. www.uspreventiveservices 159. Talamonti MS. Screening strategies for pancreatic
121. Fenton J, Weyrich M, Durbin S, Liu Y, Bang taskforce.org/Page/Document/Recommendations cancer in high-risk patients: opportunities to make
H, Melnikow J. Prostate-Specific Antigen-Based StatementDraft/prostate-cancer-screening1. Accessed a real impact but many questions and challenges
Screening for Prostate Cancer: A Systematic Evidence April 12, 2017. still ahead. JAMA Surg. 2015;150(6):518–519.
Review for the U.S. Preventive Services Task Force. 140. Thompson IM, Ankerst DP, Chi C, et al. Operating 160. Poruk KE, Firpo MA, Adler DG, Mulvihill SJ.
Rockville, MD: Agency for Healthcare Research and characteristics of prostate-specific antigen in men Screening for pancreatic cancer: why, how, and
Quality; 2017. with an initial PSA level of 3.0 ng/ml or lower. who? Ann Surg. 2013;257(1):17–26.
122. Louie KS, Seigneurin A, Cathcart P, Sasieni P. Do JAMA. 2005;294(1):66–70. 161. Del Chiaro M, Verbeke CS, Kartalis N, et al.
prostate cancer risk models improve the predictive 141. Petitti D, Lin J, Burda B. Overdiagnosis in Prostate Short-term results of a magnetic resonance imaging-
accuracy of PSA screening? A meta-analysis. Ann Cancer Screening Decision Models: A Contextual based Swedish screening program for individuals
Oncol. 2015;26(5):848–864. Review for the U.S. Preventive Services Task Force. at risk for pancreatic cancer. JAMA Surg. 2015;
123. Ankerst DP, Hoefler J, Bock S, et al. Prostate Rockville, MD: Agency for Healthcare Research and 150(6):512–518.
Cancer Prevention Trial risk calculator 2.0 for the Quality; 2017. 162. Canto MI, Goggins M, Hruban RH, et al. Screening
prediction of low- vs high-grade prostate cancer. 142. Siegel RL, Miller KD, Jemal A. Cancer statistics, for early pancreatic neoplasia in high-risk individuals:
Urology. 2014;83(6):1362–1367. 2016. CA Cancer J Clin. 2016;66(1):7–30. a prospective controlled study. Clin Gastroenterol
124. Roobol MJ, Steyerberg EW, Kranse R, et al. A 143. Rahib L, Smith BD, Aizenberg R, Rosenzweig AB, Hepatol. 2006;4(6):766–781, quiz 665.
risk-based strategy improves prostate-specific Fleshman JM, Matrisian LM. Projecting cancer 163. Tanaka M, Chari S, Adsay V, et al. International
antigen-driven detection of prostate cancer. Eur incidence and deaths to 2030: the unexpected burden consensus guidelines for management of intraductal
Urol. 2010;57(1):79–85. of thyroid, liver, and pancreas cancers in the United papillary mucinous neoplasms and mucinous cystic
125. Roobol MJ, van Vugt HA, Loeb S, et al. Prediction States. Cancer Res. 2014;74(11):2913–2921. neoplasms of the pancreas. Pancreatology. 2006;
of prostate cancer risk: the role of prostate volume 144. Moffitt RA, Marayati R, Flate EL, et al. Virtual 6(1–2):17–32.
and digital rectal examination in the ERSPC risk microdissection identifies distinct tumor- and 164. Davis B, Lowy AM. Surgical management of
calculators. Eur Urol. 2012;61(3):577–583. stroma-specific subtypes of pancreatic ductal adeno- hereditary pancreatic cancer. Med Clin North Am.
126. Foley RW, Maweni RM, Gorman L, et al. European carcinoma. Nat Genet. 2015;47(10):1168–1178. 2000;84(3):749–759.
Randomised Study of Screening for Prostate Cancer 145. Hruban RH, Maitra A, Schulick R, et al. Emerging 165. Goggins M, Canto M, Hruban R. Can we screen
(ERSPC) risk calculators significantly outperform the molecular biology of pancreatic cancer. Gastrointest high-risk individuals to detect early pancreatic
Prostate Cancer Prevention Trial (PCPT) 2.0 in the Cancer Res. 2008;2(suppl 4):10–15. carcinoma? J Surg Oncol. 2000;74(4):243–248.
prediction of prostate cancer: a multi-institutional 146. Ryan DP, Hong TS, Bardeesy N. Pancreatic adeno- 166. Langer P, Kann PH, Fendrich V, et al. Five years
study. BJU Int. 2016;118(5):706–713. carcinoma. N Engl J Med. 2014;371(11):1039–1049. of prospective screening of high-risk individuals
127. Carroll PR, Parsons JK, Andriole G, et al. NCCN 147. Canto MI, Hruban RH, Fishman EK, et al. from families with familial pancreatic cancer. Gut.
guidelines insights: prostate cancer early detec- Frequent detection of pancreatic lesions in 2009;58(10):1410–1418.
tion, version 2.2016. J Natl Compr Canc Netw. asymptomatic high-risk individuals. Gastroenterology. 167. Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent
2016;14(5):509–519. 2012;142(4):796–804, quiz e714–e795. J, Jemal A. Global cancer statistics, 2012. CA Cancer
128. Prorok PC, Andriole GL, Bresalier RS, et al. Design 148. Tanaka M, Fernandez-del Castillo C, Adsay V, et al. J Clin. 2015;65(2):87–108.
of the Prostate, Lung, Colorectal and Ovarian International consensus guidelines 2012 for the 168. Ryerson AB, Eheman CR, Altekruse SF, et al.
(PLCO) Cancer Screening Trial. Control Clin Trials. management of IPMN and MCN of the pancreas. Annual report to the nation on the status of cancer,
2000;21(suppl 6):273S–309S. Pancreatology. 2012;12(3):183–197. 1975–2012, featuring the increasing incidence of
129. Andriole GL, Crawford ED, Grubb RL, et al. Mortal- 149. Zhang XM, Mitchell DG, Dohke M, Holland liver cancer. Cancer. 2016;122(9):1312–1337.
ity results from a randomized prostate-cancer screen- GA, Parker L. Pancreatic cysts: depiction on 169. Altekruse SF, Henley SJ, Cucinelli JE, McGlynn
ing trial. N Engl J Med. 2009;360(13):1310–1319. single-shot fast spin-echo MR images. Radiology. KA. Changing hepatocellular carcinoma incidence
130. Andriole GL, Crawford ED, Grubb RL, et al. 2002;223(2):547–553. and liver cancer mortality rates in the United States.
Prostate cancer screening in the randomized Prostate, 150. de Jong K, Nio CY, Hermans JJ, et al. High Am J Gastroenterol. 2014;109(4):542–553.
Lung, Colorectal, and Ovarian Cancer Screening prevalence of pancreatic cysts detected by screen- 170. Njei B, Rotman Y, Ditah I, Lim JK. Emerging trends
Trial: mortality results after 13 years of follow-up. ing magnetic resonance imaging examinations. Clin in hepatocellular carcinoma incidence and mortality.
J Natl Cancer Inst. 2012;104(2):125–132. Gastroenterol Hepatol. 2010;8(9):806–811. Hepatology. 2015;61(1):191–199.
131. Shoag JE, Mittal S, Hu JC. Reevaluating PSA 151. Del Chiaro M, Ateeb Z, Hansson MR, et al. Survival 171. El-Serag HB. Epidemiology of viral hepatitis
testing rates in the PLCO Trial. N Engl J Med. analysis and risk for progression of intraductal papil- and hepatocellular carcinoma. Gastroenterology.
2016;374(18):1795–1796. lary mucinous neoplasia of the pancreas (IPMN) 2012;142(6):1264–1273.e1261.
132. Schröder FH, Hugosson J, Roobol MJ, et al. under surveillance: a single-institution experience. 172. Beste LA, Leipertz SL, Green PK, Dominitz JA,
Screening and prostate-cancer mortality in a Ann Surg Oncol. 2016. Ross D, Ioannou GN. Trends in burden of cirrhosis
randomized European study. N Engl J Med. 2009; 152. Jais B, Rebours V, Malleo G, et al. Serous cystic and hepatocellular carcinoma by underlying liver
360(13):1320–1328. neoplasm of the pancreas: a multinational study of disease in US veterans, 2001-2013. Gastroenterology.
133. Hugosson J, Carlsson S, Aus G, et al. Mortality 2622 patients under the auspices of the International 2015;149(6):1471–1482.e1475, quiz e1417–e1478.
results from the Göteborg randomised population- Association of Pancreatology and European Pancre- 173. Lok AS, Seeff LB, Morgan TR, et al. Incidence
based prostate-cancer screening trial. Lancet Oncol. atic Club (European Study Group on Cystic Tumors of hepatocellular carcinoma and associated risk
2010;11(8):725–732. of the Pancreas). Gut. 2016;65(2):305–312. factors in hepatitis C–related advanced liver disease.
134. Roobol MJ, Kranse R, Bangma CH, et al. Screening 153. Klein AP, Brune KA, Petersen GM, et al. Prospective Gastroenterology. 2009;136(1):138–148.
for prostate cancer: results of the Rotterdam section risk of pancreatic cancer in familial pancreatic cancer 174. Yang JD, Kim WR, Coelho R, et al. Cirrhosis
of the European Randomized Study of Screening kindreds. Cancer Res. 2004;64(7):2634–2638. is present in most patients with hepatitis B and
for Prostate Cancer. Eur Urol. 2013;64(4):530–539. 154. Vrieling A, Bueno-de-Mesquita HB, Boshuizen HC, hepatocellular carcinoma. Clin Gastroenterol Hepatol.
135. Luján M, Páez A, Angulo JC, et al. Prostate cancer et al. Cigarette smoking, environmental tobacco 2011;9(1):64–70.
incidence and mortality in the Spanish section of smoke exposure and pancreatic cancer risk in the 175. El-Serag HB, Kanwal F, Davila JA, Kramer J,
the European Randomized Study of Screening for European Prospective Investigation into Cancer Richardson P. A new laboratory-based algorithm to
Prostate Cancer (ERSPC). Prostate Cancer Prostatic and Nutrition. Int J Cancer. 2010;126(10):2394– predict development of hepatocellular carcinoma in
Dis. 2014;17(2):187–191. 2403. patients with hepatitis C and cirrhosis. Gastroenterol-
136. Kilpeläinen TP, Tammela TL, Malila N, et al. 155. Tramacere I, Scotti L, Jenab M, et al. Alcohol drink- ogy. 2014;146(5):1249–1255.
Prostate cancer mortality in the Finnish Randomized ing and pancreatic cancer risk: a meta-analysis of 176. Kanwal F, Kramer JR, Ilyas J, Duan Z, El-Serag HB.
Screening Trial. J Natl Cancer Inst. 2013;105(10): the dose-risk relation. Int J Cancer. 2010;126(6): HCV genotype 3 is associated with an increased risk
719–725. 1474–1486. of cirrhosis and hepatocellular cancer in a national
398.e4 Part I: Science and Clinical Oncology

sample of U.S. Veterans with HCV. Hepatology. hepatocellular carcinoma in patients with cirrhosis. 215. Gonzalez CA, Agudo A. Carcinogenesis, prevention
2014;60(1):98–105. Aliment Pharmacol Ther. 2009;30(1):37–47. and early detection of gastric cancer: where we
177. Chang KC, Wu YY, Hung CH, et al. Clinical-guide 196. Santi V, Trevisani F, Gramenzi A, et al. Semiannual are and where we should go. Int J Cancer. 2012;
risk prediction of hepatocellular carcinoma develop- surveillance is superior to annual surveillance for 130(4):745–753.
ment in chronic hepatitis C patients after interferon- the detection of early hepatocellular carcinoma and 216. Lochhead P, El-Omar EM. Helicobacter pylori
based therapy. Br J Cancer. 2013;109(9):2481–2488. patient survival. J Hepatol. 2010;53(2):291–297. infection and gastric cancer. Best Pract Res Clin
178. El-Serag HB, Hampel H, Javadi F. The association 197. Trinchet JC, Chaffaut C, Bourcier V, et al. Ultraso- Gastroenterol. 2007;21(2):281–297.
between diabetes and hepatocellular carcinoma: a nographic surveillance of hepatocellular carcinoma 217. Plummer M, Franceschi S, Vignat J, Forman D,
systematic review of epidemiologic evidence. Clin in cirrhosis: a randomized trial comparing 3- and de Martel C. Global burden of gastric cancer
Gastroenterol Hepatol. 2006;4(3):369–380. 6-month periodicities. Hepatology. 2011;54(6): attributable to Helicobacter pylori. Int J Cancer. 2015;
179. Wang P, Kang D, Cao W, Wang Y, Liu Z. Diabetes 1987–1997. 136(2):487–490.
mellitus and risk of hepatocellular carcinoma: a 198. Andersson KL, Salomon JA, Goldie SJ, Chung RT. 218. International Agency for Research on Cancer (IARC).
systematic review and meta-analysis. Diabetes Metab Cost effectiveness of alternative surveillance strate- Biological agents. Volume 100 B. A review of human
Res Rev. 2012;28(2):109–122. gies for hepatocellular carcinoma in patients with carcinogens. IARC Monogr Eval Carcinog Risks Hum.
180. Saunders D, Seidel D, Allison M, Lyratzopoulos G. cirrhosis. Clin Gastroenterol Hepatol. 2008;6(12): 2012;100(Pt B):1–441.
Systematic review: the association between obesity 1418–1424. 219. Cover TL, Blaser MJ. Helicobacter pylori in health and
and hepatocellular carcinoma—epidemiological 199. Arguedas MR, Chen VK, Eloubeidi MA, Fallon MB. disease. Gastroenterology. 2009;136(6):1863–1873.
evidence. Aliment Pharmacol Ther. 2010;31(10): Screening for hepatocellular carcinoma in patients 220. Peleteiro B, Bastos A, Ferro A, Lunet N. Prevalence of
1051–1063. with hepatitis C cirrhosis: a cost-utility analysis. Am Helicobacter pylori infection worldwide: a systematic
181. Schlesinger S, Aleksandrova K, Pischon T, et al. J Gastroenterol. 2003;98(3):679–690. review of studies with national coverage. Dig Dis
Abdominal obesity, weight gain during adulthood 200. Patel D, Terrault NA, Yao FY, Bass NM, Ladabaum Sci. 2014;59(8):1698–1709.
and risk of liver and biliary tract cancer in a European U. Cost-effectiveness of hepatocellular carcinoma 221. Camargo MC, Murphy G, Koriyama C, et al.
cohort. Int J Cancer. 2013;132(3):645–657. surveillance in patients with hepatitis C virus–related Determinants of Epstein-Barr virus–positive gastric
182. Hassan MM, Abdel-Wahab R, Kaseb A, et al. cirrhosis. Clin Gastroenterol Hepatol. 2005;3(1): cancer: an international pooled analysis. Br J Cancer.
Obesity early in adulthood increases risk but does 75–84. 2011;105(1):38–43.
not affect outcomes of hepatocellular carcinoma. 201. Thompson Coon J, Rogers G, Hewson P, et al. 222. Capelle LG, Van Grieken NC, Lingsma HF, et al.
Gastroenterology. 2015;149(1):119–129. Surveillance of cirrhosis for hepatocellular carcinoma: Risk and epidemiological time trends of gastric
183. White DL, Kanwal F, El-Serag HB. Association a cost-utility analysis. Br J Cancer. 2008;98(7): cancer in Lynch syndrome carriers in the Nether-
between nonalcoholic fatty liver disease and risk 1166–1175. lands. Gastroenterology. 2010;138(2):487–492.
for hepatocellular cancer, based on systematic review. 202. Pocha C, Dieperink E, McMaken KA, Knott A, 223. Oliveira C, Seruca R, Carneiro F. Genetics, pathology,
Clin Gastroenterol Hepatol. 2012;10(12):1342–1359. Thuras P, Ho SB. Surveillance for hepatocellular and clinics of familial gastric cancer. Int J Surg Pathol.
e1342. cancer with ultrasonography vs. computed tomogra- 2006;14(1):21–33.
184. Romeo S, Kozlitina J, Xing C, et al. Genetic variation phy—a randomised study. Aliment Pharmacol Ther. 224. Karimi P, Islami F, Anandasabapathy S, Freedman
in PNPLA3 confers susceptibility to nonalcoholic 2013;38(3):303–312. ND, Kamangar F. Gastric cancer: descriptive epi-
fatty liver disease. Nat Genet. 2008;40(12):1461– 203. Kanwal F, El-Serag HB, Ross D. Surveillance for hepa- demiology, risk factors, screening, and prevention.
1465. tocellular carcinoma: can we focus on the mission? Cancer Epidemiol Biomarkers Prev. 2014;23(5):
185. Hassan MM, Kaseb A, Etzel CJ, et al. Genetic Clin Gastroenterol Hepatol. 2015;13(4):805–807. 700–713.
variation in the PNPLA3 gene and hepatocellular 204. Singal AG, Volk ML, Rakoski MO, et al. Patient 225. World Cancer Research Fund/American Institute for
carcinoma in USA: risk and prognosis prediction. involvement in healthcare is associated with higher Cancer Research (WCRF/AICR). Food, Nutrition,
Mol Carcinog. 2013;52(suppl 1):E139–E147. rates of surveillance for hepatocellular carcinoma. Physical Activity, and the Prevention of Cancer: a
186. Bruix J, Sherman M. Management of hepatocellular J Clin Gastroenterol. 2011;45(8):727–732. Global Perspective. Washington DC: AICR 2007.
carcinoma: an update. Hepatology. 2011;53(3):1020– 205. El-Serag HB, Kanwal F. Epidemiology of hepato- 226. Toyoda T, Tsukamoto T, Hirano N, et al. Synergistic
1022. cellular carcinoma in the United States: where are upregulation of inducible nitric oxide synthase and
187. Singal AG, Yopp AC, Gupta S, et al. Failure rates we? Where do we go? Hepatology. 2014;60(5): cyclooxygenase-2 in gastric mucosa of Mongolian
in the hepatocellular carcinoma surveillance process. 1767–1775. gerbils by a high-salt diet and Helicobacter pylori
Cancer Prev Res (Phila). 2012;5(9):1124–1130. 206. Marrero JA, Feng Z, Wang Y, et al. Alpha-fetoprotein, infection. Histol Histopathol. 2008;23(5):593–599.
188. Blais P, Husain N, Kramer JR, Kowalkowski M, El- des-gamma carboxyprothrombin, and lectin-bound 227. Loh JT, Torres VJ, Cover TL. Regulation of Heli-
Serag H, Kanwal F. Nonalcoholic fatty liver disease alpha-fetoprotein in early hepatocellular carcinoma. cobacter pylori cagA expression in response to salt.
is underrecognized in the primary care setting. Am Gastroenterology. 2009;137(1):110–118. Cancer Res. 2007;67(10):4709–4715.
J Gastroenterol. 2015;110(1):10–14. 207. Ferlay J, Soerjomataram I, Ervik M, et al. GLOBO- 228. Leung WK, Wu MS, Kakugawa Y, et al. Screening for
189. Mourad A, Deuffic-Burban S, Ganne-Carrie CAN 2012 v1.1, Cancer Incidence and Mortality gastric cancer in Asia: current evidence and practice.
N, et al. Hepatocellular carcinoma screening in Worldwide: IARC CancerBase No. 11. Lyon, France: Lancet Oncol. 2008;9(3):279–287.
patients with compensated hepatitis C virus (HCV)- International Agency for Research on Cancer; 2014. 229. Choi KS, Suh M. Screening for gastric cancer: the
related cirrhosis aware of their HCV status improves 208. Bosman FT, World Health Organization, International usefulness of endoscopy. Clin Endosc. 2014;47(6):
survival: a modeling approach. Hepatology. 2014; Agency for Research on Cancer. WHO Classification 490–496.
59(4):1471–1481. of Tumours of the Digestive System. Lyon: IARC Press; 230. Asaka M. A new approach for elimination of gastric
190. Zhang BH, Yang BH, Tang ZY. Randomized con- 2010. cancer deaths in Japan. Int J Cancer. 2013;132(6):
trolled trial of screening for hepatocellular carcinoma. 209. Correa P. Gastric cancer: overview. Gastroenterol Clin 1272–1276.
J Cancer Res Clin Oncol. 2004;130(7):417–422. North Am. 2013;42(2):211–217. 231. Asaka M, Kato M, Sakamoto N. Roadmap to
191. Finberg HJ. Whither (wither?) the ultrasound special- 210. Tsukanov VV, Butorin NN, Maady AS, et al. Heli- eliminate gastric cancer with Helicobacter pylori
ist? J Ultrasound Med. 2004;23(12):1543–1547. cobacter pylori infection, intestinal metaplasia, and eradication and consecutive surveillance in Japan.
192. Singal AG, Pillai A, Tiro J. Early detection, cura- gastric cancer risk in Eastern Siberia. Helicobacter. J Gastroenterol. 2014;49(1):1–8.
tive treatment, and survival rates for hepatocellular 2011;16(2):107–112. 232. Areia M, Carvalho R, Cadime AT, Rocha Goncalves
carcinoma surveillance in patients with cirrhosis: a 211. Lui FH, Tuan B, Swenson SL, Wong RJ. Ethnic F, Dinis-Ribeiro M. Screening for gastric cancer and
meta-analysis. PLoS Med. 2014;11(4):e1001624. disparities in gastric cancer incidence and survival in surveillance of premalignant lesions: a systematic
193. Dalton-Fitzgerald E, Tiro J, Kandunoori P, Halm EA, the USA: an updated analysis of 1992–2009 SEER review of cost-effectiveness studies. Helicobacter.
Yopp A, Singal AG. Practice patterns and attitudes data. Dig Dis Sci. 2014;59(12):3027–3034. 2013;18(5):325–337.
of primary care providers and barriers to surveillance 212. Anderson WF, Camargo MC, Fraumeni JF Jr, Correa 233. Choi KS, Jun JK, Park EC, et al. Performance
of hepatocellular carcinoma in patients with cir- P, Rosenberg PS, Rabkin CS. Age-specific trends in of different gastric cancer screening methods in
rhosis. Clin Gastroenterol Hepatol. 2015;13(4):791– incidence of noncardia gastric cancer in US adults. Korea: a population-based study. PLoS ONE. 2012;
798.e791. JAMA. 2010;303(17):1723–1728. 7(11):e50041.
194. Poustchi H, Farrell GC, Strasser SI, Lee AU, 213. Arnold M, Karim-Kos HE, Coebergh JW, et al. 234. Tashiro A, Sano M, Kinameri K, Fujita K, Takeuchi
McCaughan GW, George J. Feasibility of con- Recent trends in incidence of five common cancers Y. Comparing mass screening techniques for gastric
ducting a randomized control trial for liver cancer in 26 European countries since 1988: analysis of cancer in Japan. World J Gastroenterol. 2006;12(30):
screening: is a randomized controlled trial for liver the European Cancer Observatory. Eur J Cancer. 4873–4874.
cancer screening feasible or still needed? Hepatology. 2015;51(9):1164–1187. 235. de Vries AC, van Grieken NC, Looman CW, et al.
2011;54(6):1998–2004. 214. Bertuccio P, Chatenoud L, Levi F, et al. Recent Gastric cancer risk in patients with premalignant
195. Singal A, Volk ML, Waljee A, et al. Meta-analysis: patterns in gastric cancer: a global overview. Int J gastric lesions: a nationwide cohort study in the
surveillance with ultrasound for early-stage Cancer. 2009;125(3):666–673. Netherlands. Gastroenterology. 2008;134(4):945–952.
Screening and Early Detection  •  CHAPTER 23 398.e5
398.e5

236. You WC, Li JY, Blot WJ, et al. Evolution of 256. Spechler SJ, Sharma P, Souza RF, Inadomi JM, symptoms of gastro-oesophageal reflux. Aliment
precancerous lesions in a rural Chinese popula- Shaheen NJ. American Gastroenterological Associa- Pharmacol Ther. 2010;32(10):1222–1227.
tion at high risk of gastric cancer. Int J Cancer. tion medical position statement on the management 275. Ferlay J, Soerjomataram I, Dikshit R, et al. Cancer
1999;83(5):615–619. of Barrett’s esophagus. Gastroenterology. 2011;140(3): incidence and mortality worldwide: sources, methods
237. Choi IJ. Endoscopic gastric cancer screening and sur- 1084–1091. and major patterns in GLOBOCAN 2012. Int J
veillance in high-risk groups. Clin Endosc. 2014;47(6): 257. Shaheen NJ, Falk GW, Iyer PG, Gerson LB. Cancer. 2015;136(5):E359–E386.
497–503. ACG Clinical Guideline: Diagnosis and manage- 276. National Cancer Institute (NCI). SEER Stat Fact
238. Ford AC, Forman D, Hunt RH, Yuan Y, Moayyedi ment of Barrett’s esophagus. Am J Gastroenterol. Sheets: Oral Cavity and Pharynx Cancer. Bethesda,
P. Helicobacter pylori eradication therapy to prevent 2016;111(1):30–50. MD SEER 18 2007-2013.
gastric cancer in healthy asymptomatic infected 258. Bhat SK, McManus DT, Coleman HG, et al. 277. U.S. Department of Health and Human Services
individuals: systematic review and meta-analysis of Oesophageal adenocarcinoma and prior diagnosis (DHHS). The Health Consequences of Smoking:
randomised controlled trials. BMJ. 2014;348:g3174. of Barrett’s oesophagus: a population-based study. A Report of the Surgeon General. Atlanta, GA:
239. Helicobacter pylori Eradication as a Strategy for Gut. 2015;64:20–25. Department of Health and Human Services, Centers
Preventing Gastric Cancer. Lyon, France: International 259. El-Serag HB, Naik AD, Duan Z, et al. Surveillance for Disease Control and Prevention, National Center
Agency for Research on Cancer (IARC Working endoscopy is associated with improved outcomes of for Chronic Disease Prevention and Health Promo-
Group Reports, No. 8). 2014. oesophageal adenocarcinoma detected in patients with tion, Office on Smoking and Health; 2004.
240. Zeng M, Mao XH, Li JX, et al. Efficacy, safety, and Barrett’s oesophagus. Gut. 2016;65(8):1252–1260. 278. Goldstein BY, Chang SC, Hashibe M, La Vecchia
immunogenicity of an oral recombinant Helicobacter 260. Hur C, Choi SE, Rubenstein JH, et al. The cost C, Zhang ZF. Alcohol consumption and cancers of
pylori vaccine in children in China: a randomised, effectiveness of radiofrequency ablation for Barrett’s the oral cavity and pharynx from 1988 to 2009: an
double-blind, placebo-controlled, phase 3 trial. esophagus. Gastroenterology. 2012;143(3):567–575. update. Eur J Cancer Prev. 2010;19(6):431–465.
Lancet. 2015;386(10002):1457–1464. 261. Gordon LG, Mayne GC, Hirst NG, Bright T, 279. Huber MA, Tantiwongkosi B. Oral and oropharyngeal
241. Pan KF, Zhang L, Gerhard M, et al. A large Whiteman DC, Watson DI. Cost-effectiveness of cancer. Med Clin North Am. 2014;98(6):1299–1321.
randomised controlled intervention trial to prevent endoscopic surveillance of non-dysplastic Barrett’s 280. Blot WJ, McLaughlin JK, Winn DM, et al. Smoking
gastric cancer by eradication of Helicobacter pylori in esophagus. Gastrointest Endosc. 2014;79(2):242– and drinking in relation to oral and pharyngeal
Linqu County, China: baseline results and factors 256.e246. cancer. Cancer Res. 1988;48(11):3282–3287.
affecting the eradication. Gut. 2016;65(1):9–18. 262. Rees JR, Lao-Sirieix P, Wong A, Fitzgerald RC. Treat- 281. Guha N, Warnakulasuriya S, Vlaanderen J, Straif
242. Thrift AP. The epidemic of oesophageal carcinoma: ment for Barrett’s oesophagus. Cochrane Database K. Betel quid chewing and the risk of oral and
where are we now? Cancer Epidemiol. 2016;41:88–95. Syst Rev. 2010;(1):Cd004060. oropharyngeal cancers: a meta-analysis with implica-
243. Pohl H, Welch HG. The role of overdiagnosis and 263. Gupta M, Iyer PG, Lutzke L, et al. Recurrence of tions for cancer control. Int J Cancer. 2014;135(6):
reclassification in the marked increase of esophageal esophageal intestinal metaplasia after endoscopic 1433–1443.
adenocarcinoma incidence. J Natl Cancer Inst. 2005; mucosal resection and radiofrequency ablation 282. Marron M, Boffetta P, Zhang ZF, et al. Cessation of
97(2):142–146. of Barrett’s esophagus: results from a US Multicenter alcohol drinking, tobacco smoking and the reversal
244. Thrift AP, Whiteman DC. The incidence of esopha- Consortium. Gastroenterology. 2013;145(1):79– of head and neck cancer risk. Int J Epidemiol.
geal adenocarcinoma continues to rise: analysis of 86.e71. 2010;39(1):182–196.
period and birth cohort effects on recent trends. 264. Orman ES, Li N, Shaheen NJ. Efficacy and 283. Wu YH, Yen CJ, Hsiao JR, et al. A Comprehensive
Ann Oncol. 2012;23(12):3155–3162. durability of radiofrequency ablation for Barrett’s analysis on the association between tobacco-free betel
245. Edgren G, Adami HO, Weiderpass E, Nyren O. A Esophagus: systematic review and meta-analysis. Clin quid and risk of head and neck cancer in Taiwanese
global assessment of the oesophageal adenocarcinoma Gastroenterol Hepatol. 2013;11(10):1245–1255. men. PLoS ONE. 2016;11(10):e0164937.
epidemic. Gut. 2013;62(10):1406–1414. 265. El-Serag HB, Graham DY. Routine polypectomy for 284. Castellsagué X, Alemany L, Quer M, et al. HPV
246. Vaughan TL, Fitzgerald RC. Precision prevention of colorectal polyps and ablation for Barrett’s esopha- involvement in head and neck cancers: compre-
oesophageal adenocarcinoma. Nat Rev Gastroenterol gus are intellectually the same. Gastroenterology. hensive assessment of biomarkers in 3680 patients.
Hepatol. 2015;12(4):243–248. 2011;140(2):386–388. J Natl Cancer Inst. 2016;108(6):djv403.
247. Pandeya N, Olsen CM, Whiteman DC. Sex dif- 266. Sorbi D, Gostout CJ, Henry J, Lindor KD. Unsedated 285. de Martel C, Plummer M, Vignat J, Franceschi S.
ferences in the proportion of esophageal squamous small-caliber esophagogastroduodenoscopy (EGD) Worldwide burden of cancer attributable to HPV
cell carcinoma cases attributable to tobacco smoking versus conventional EGD: a comparative study. by site, country and HPV type. Int J Cancer. 2017.
and alcohol consumption. Cancer Epidemiol. 2013; Gastroenterology. 1999;117(6):1301–1307. 286. Ndiaye C, Mena M, Alemany L, et al. HPV DNA,
37(5):579–584. 267. Saeian K, Staff DM, Vasilopoulos S, et al. Unsedated E6/E7 mRNA, and p16INK4a detection in head and
248. Taylor PR, Abnet CC, Dawsey SM. Squamous dys- transnasal endoscopy accurately detects Barrett’s neck cancers: a systematic review and meta-analysis.
plasia—the precursor lesion for esophageal squamous metaplasia and dysplasia. Gastrointest Endosc. 2002; Lancet Oncol. 2014;15(12):1319–1331.
cell carcinoma. Cancer Epidemiol Biomarkers Prev. 56(4):472–478. 287. Gillison ML, Chaturvedi AK, Anderson WF, Fakhry
2013;22(4):540–552. 268. Shariff MK, Bird-Lieberman EL, O’Donovan C. Epidemiology of human papillomavirus–positive
249. Engel LS, Chow WH, Vaughan TL, et al. Population M, et al. Randomized crossover study comparing head and neck squamous cell carcinoma. J Clin
attributable risks of esophageal and gastric cancers. efficacy of transnasal endoscopy with that of standard Oncol. 2015;33(29):3235–3242.
J Natl Cancer Inst. 2003;95(18):1404–1413. endoscopy to detect Barrett’s esophagus. Gastrointest 288. Heck JE, Berthiller J, Vaccarella S, et al. Sexual
250. Olsen CM, Pandeya N, Green AC, Webb PM, Endosc. 2012;75(5):954–961. behaviours and the risk of head and neck cancers:
Whiteman DC. Population attributable fractions 269. Peery AF, Hoppo T, Garman KS, et al. Feasibility, a pooled analysis in the International Head and Neck
of adenocarcinoma of the esophagus and gastro- safety, acceptability, and yield of office-based, Cancer Epidemiology (INHANCE) consortium. Int
esophageal junction. Am J Epidemiol. 2011;174(5): screening transnasal esophagoscopy (with video). J Epidemiol. 2010;39(1):166–181.
582–590. Gastrointest Endosc. 2012;75(5):945–953.e942. 289. Herrero R, Quint W, Hildesheim A, et al. Reduced
251. Ronkainen J, Aro P, Storskrubb T, et al. Prevalence 270. Lao-Sirieix P, Rous B, O’Donovan M, Hardwick prevalence of oral human papillomavirus (HPV)
of Barrett’s esophagus in the general population: RH, Debiram I, Fitzgerald RC. Non-endoscopic 4 years after bivalent HPV vaccination in a ran-
an endoscopic study. Gastroenterology. 2005;129(6): immunocytological screening test for Barrett’s domized clinical trial in Costa Rica. PLoS ONE.
1825–1831. oesophagus. Gut. 2007;56(7):1033–1034. 2013;8(7):e68329.
252. Hvid-Jensen F, Pedersen L, Drewes AM, Sorensen 271. Kadri SR, Lao-Sirieix P, O’Donovan M, et al. 290. Olson C, Burda B, Beil T, Whitlock E. Screening for
HT, Funch-Jensen P. Incidence of adenocarcinoma Acceptability and accuracy of a non-endoscopic Oral Cancer: A Targeted Evidence Update for the
among patients with Barrett’s esophagus. N Engl J screening test for Barrett’s oesophagus in primary U.S. Preventive Services Task Force. In. Rockville,
Med. 2011;365(15):1375–1383. care: cohort study. BMJ. 2010;341:c4372. MD: Agency for Healthcare Research and Quality;
253. Desai TK, Krishnan K, Samala N, et al. The 272. Ross-Innes CS, Debiram-Beecham I, O’Donovan 2013.
incidence of oesophageal adenocarcinoma in non- M, et al. Evaluation of a minimally invasive 291. Brocklehurst P, Kujan O, O’Malley L, Ogden G,
dysplastic Barrett’s oesophagus: a meta-analysis. Gut. cell sampling device coupled with assessment of Shepherd S, Glenny A. Screening programmes for
2012;61(7):970–976. trefoil factor 3 expression for diagnosing Barrett’s the early detection and prevention of oral cancer
254. Rastogi A, Puli S, El-Serag HB, Bansal A, Wani S, esophagus: a multi-center case-control study. PLoS (Review). Cochrane Database Syst Rev. 2013;(11).
Sharma P. Incidence of esophageal adenocarcinoma Med. 2015;12(1):e1001780. 292. Sankaranarayanan R, Ramadas K, Thomas G, et al.
in patients with Barrett’s esophagus and high-grade 273. Lagergren J, Bergstrom R, Lindgren A, Nyren O. Effect of screening on oral cancer mortality in Kerala,
dysplasia: a meta-analysis. Gastrointest Endosc. 2008; Symptomatic gastroesophageal reflux as a risk factor India: a cluster-randomised controlled trial. Lancet.
67(3):394–398. for esophageal adenocarcinoma. N Engl J Med. 2005;365(9475):1927–1933.
255. di Pietro M, Chan D, Fitzgerald RC, Wang KK. 1999;340(11):825–831. 293. Walsh T, Liu J, Brocklehurst P, et al. Clinical assess-
Screening for Barrett’s esophagus. Gastroenterology. 274. Rubenstein JH, Taylor JB. Meta-analysis: the ment to screen for the detection of oral cavity cancer
2015;148(5):912–923. association of oesophageal adenocarcinoma with and potentially malignant disorders in apparently
398.e6 Part I: Science and Clinical Oncology

healthy adults. In. Cochrane Database of Systematic 312. McLaughlin JR, Risch HA, Lubinski J, et al. Repro- Safety/AltersandNotices/ucm519413.htm. Accessed
Reviews, The Cochrane Collaboration: John Wiley ductive risk factors for ovarian cancer in carriers of June 6, 2017.
& Sons, Ltd.; 2013. BRCA1 or BRCA2 mutations: a case-control study. 331. Isaacs C, Peshkin B. Management of Patients at
294. Macey R, Walsh T, Brocklehurst P, et al. Diagnostic Lancet Oncol. 2007;8(1):26–34. High-Risk for Breast and Ovarian Cancer. UpTo-
tests for oral cancer and potentially malignant 313. Reid BM, Permuth JB, Sellers TA. Epidemiology Date. 2016. https://www.uptodate.com/contents/
disorders in patients presenting with clinically of ovarian cancer: a review. Cancer Biol Med. 2017; management-of-patients-at-high-risk-for-breast-and
evident lesions. In. Cochrane Database of Systematic 14(1):9–32. -ovarian-cancer. Accessed June 9, 2017.
Reviews, The Cochrane Collaboration: John Wiley 314. Langseth H, Hankinson SE, Siemiatycki J, Weider- 332. Yang S, Thiel KW, Leslie KK. Progesterone: the
& Sons, Ltd.; 2015. pass E. Perineal use of talc and risk of ovarian cancer. ultimate endometrial tumor suppressor. Trends
295. Lingen MW, et al. Evidence-based clinical practice J Epidemiol Community Health. 2008;62(4):358–360. Endocrinol Metab. 2011;22(4):145–152.
guideline for the evaluation of potentially malignant 315. Huncharek M, Geschwind JF, Kupelnick B. Perineal 333. Fearnley EJ, Marquart L, Spurdle AB, Weinstein
disorders in the oral cavity: a report of the American application of cosmetic talc and risk of invasive P, Webb PM, Group AOCSGaANECS. Polycystic
Dental Association. J Am Dent Assoc. 2017;148: epithelial ovarian cancer: a meta-analysis of 11,933 ovary syndrome increases the risk of endometrial
712–727. subjects from sixteen observational studies. Anticancer cancer in women aged less than 50 years: an Aus-
296. Moyer VA, Force USPST. Screening for oral cancer: Res. 2003;23(2C):1955–1960. tralian case-control study. Cancer Causes Control.
U.S. Preventive Services Task Force recommendation 316. Baan R, Straif K, Grosse Y, et al. Carcinogenicity 2010;21(12):2303–2308.
statement. Ann Intern Med. 2014;160(1):55–60. of carbon black, titanium dioxide, and talc. Lancet 334. Mueck AO, Seeger H, Rabe T. Hormonal contracep-
297. Mirghani H, Jung AC, Fakhry C. Primary, secondary Oncol. 2006;7(4):295–296. tion and risk of endometrial cancer: a systematic
and tertiary prevention of human papillomavirus- 317. Wu AH, Pearce CL, Tseng CC, Templeman C, review. Endocr Relat Cancer. 2010;17(4):263–271.
driven head and neck cancers. Eur J Cancer. 2017; Pike MC. Markers of inflammation and risk of 335. Dossus L, Allen N, Kaaks R, et al. Reproductive
78:105–115. ovarian cancer in Los Angeles County. Int J Cancer. risk factors and endometrial cancer: the European
298. Kreimer AR, Johansson M, Waterboer T, et al. 2009;124(6):1409–1415. Prospective Investigation into Cancer and Nutrition.
Evaluation of human papillomavirus antibodies 318. Ness RB, Grisso JA, Cottreau C, et al. Factors related Int J Cancer. 2010;127(2):442–451.
and risk of subsequent head and neck cancer. to inflammation of the ovarian epithelium and risk of 336. Schmandt RE, Iglesias DA, Co NN, Lu KH.
J Clin Oncol. 2013;31(21):2708–2715. ovarian cancer. Epidemiology. 2000;11(2):111–117. Understanding obesity and endometrial cancer risk:
299. Ebisumoto K, Okami K, Sakai A, Sugimoto R, 319. Mills PK, Riordan DG, Cress RD, Young HA. opportunities for prevention. Am J Obstet Gynecol.
Iida M. Successful detection of a minute tonsillar Perineal talc exposure and epithelial ovarian cancer 2011;205(6):518–525.
cancer lesion on transoral examination with narrow risk in the Central Valley of California. Int J Cancer. 337. Renehan AG, Tyson M, Egger M, Heller RF, Zwahlen
band imaging: a report of 2 cases. Head Neck. 2004;112(3):458–464. M. Body-mass index and incidence of cancer: a
2016;38(suppl 1):E2421–E2424. 320. Bast RC, Badgwell D, Lu Z, et al. New tumor systematic review and meta-analysis of prospective
300. Coquia SF, Hamper UM, Holman ME, et al. markers: CA125 and beyond. Int J Gynecol Cancer. observational studies. Lancet. 2008;371(9612):
Visualization of the oropharynx with transcervical 2005;15(suppl 3):274–281. 569–578.
ultrasound. AJR Am J Roentgenol. 2015;205(6): 321. Buys SS, Partridge E, Black A, et al. Effect of 338. Adams TD, Stroup AM, Gress RE, et al. Cancer
1288–1294. screening on ovarian cancer mortality: the Prostate, incidence and mortality after gastric bypass surgery.
301. Fakhry C, Agrawal N, Califano J, et al. The use Lung, Colorectal and Ovarian (PLCO) Cancer Obesity (Silver Spring). 2009;17(4):796–802.
of ultrasound in the search for the primary site of Screening Randomized Controlled Trial. JAMA. 339. Troisi R, Potischman N, Hoover RN, Siiteri P,
unknown primary head and neck squamous cell 2011;305(22):2295–2303. Brinton LA. Insulin and endometrial cancer. Am
cancers. Oral Oncol. 2014;50(7):640–645. 322. Pinsky PF, Yu K, Kramer BS, et al. Extended mortal- J Epidemiol. 1997;146(6):476–482.
302. Blanco RG, Califano J, Messing B, et al. Transcervical ity results for ovarian cancer screening in the PLCO 340. Schmid D, Behrens G, Keimling M, Jochem C,
ultrasonography is feasible to visualize and evaluate trial with median 15 years follow-up. Gynecol Oncol. Ricci C, Leitzmann M. A systematic review and
base of tongue cancers. PLoS ONE. 2014;9(1): 2016;143(2):270–275. meta-analysis of physical activity and endometrial
e87565. 323. Skates SJ, Menon U, MacDonald N, et al. Calcula- cancer risk. Eur J Epidemiol. 2015;30(5):397–412.
303. Domchek SM, Friebel TM, Singer CF, et al. Associa- tion of the risk of ovarian cancer from serial CA-125 341. Du M, Kraft P, Eliassen AH, Giovannucci E,
tion of risk-reducing surgery in BRCA1 or BRCA2 values for preclinical detection in postmenopausal Hankinson SE, De Vivo I. Physical activity and risk
mutation carriers with cancer risk and mortality. women. J Clin Oncol. 2003;21(suppl 10):206s–210s. of endometrial adenocarcinoma in the Nurses’ Health
JAMA. 2010;304(9):967–975. 324. Lu KH, Skates S, Hernandez MA, et al. A 2-stage Study. Int J Cancer. 2014;134(11):2707–2716.
304. American Cancer Society (ACS). Ovarian Cancer: ovarian cancer screening strategy using the Risk of 342. Friedenreich C, Cust A, Lahmann PH, et al. Physical
Causes, Risk Factors and Prevention; 2017. https:// Ovarian Cancer Algorithm (ROCA) identifies early- activity and risk of endometrial cancer: the European
www.cancer.org/cancer/ovarian-cancer/causes-risks stage incident cancers and demonstrates high positive prospective investigation into cancer and nutrition.
-prevention/risk-factors.html. Accessed May 12, 2017. predictive value. Cancer. 2013;119(19):3454–3461. Int J Cancer. 2007;121(2):347–355.
305. Gayther SA, Pharoah PD. The inherited genetics of 325. Menon U, Ryan A, Kalsi J, et al. Risk algorithm 343. Fisher B, Costantino JP, Wickerham DL, et al.
ovarian and endometrial cancer. Curr Opin Genet using serial biomarker measurements doubles the Tamoxifen for prevention of breast cancer: report
Dev. 2010;20(3):231–238. number of screen-detected cancers compared with of the National Surgical Adjuvant Breast and Bowel
306. Schouten LJ, Rivera C, Hunter DJ, et al. Height, a single-threshold rule in the United Kingdom Project P-1 Study. J Natl Cancer Inst. 1998;90(18):
body mass index, and ovarian cancer: a pooled Collaborative Trial of Ovarian Cancer Screening. 1371–1388.
analysis of 12 cohort studies. Cancer Epidemiol J Clin Oncol. 2015;33(18):2062–2071. 344. Vogel VG, Costantino JP, Wickerham DL, et al.
Biomarkers Prev. 2008;17(4):902–912. 326. Jacobs IJ, Menon U, Ryan A, et al. Ovarian cancer Effects of tamoxifen vs raloxifene on the risk of
307. Lahmann PH, Cust AE, Friedenreich CM, et al. screening and mortality in the UK Collaborative developing invasive breast cancer and other disease
Anthropometric measures and epithelial ovarian Trial of Ovarian Cancer Screening (UKCTOCS): outcomes: the NSABP Study of Tamoxifen and
cancer risk in the European Prospective Investiga- a randomised controlled trial. Lancet. 2016;387 Raloxifene (STAR) P-2 trial. JAMA. 2006;295(23):
tion into Cancer and Nutrition. Int J Cancer. (10022):945–956. 2727–2741.
2010;126(10):2404–2415. 327. Rosenthal AN, Fraser L, Manchanda R, et al. 345. Watson P, Vasen HF, Mecklin JP, Järvinen H, Lynch
308. Pearce CL, Chung K, Pike MC, Wu AH. Increased Results of annual screening in phase I of the United HT. The risk of endometrial cancer in hereditary non-
ovarian cancer risk associated with menopausal Kingdom familial ovarian cancer screening study polyposis colorectal cancer. Am J Med. 1994;96(6):
estrogen therapy is reduced by adding a progestin. highlight the need for strict adherence to screening 516–520.
Cancer. 2009;115(3):531–539. schedule. J Clin Oncol. 2013;31(1):49–57. 346. Lu KH, Daniels M. Endometrial and ovarian cancer
309. Beral V, Bull D, Green J, Reeves G. Million Women 328. Rosenthal AN, Fraser LSM, Philpott S, et al. Evi- in women with Lynch syndrome: update in screening
Study Collaborators. Ovarian cancer and hormone dence of stage shift in women diagnosed with ovarian and prevention. Fam Cancer. 2013;12(2):273–277.
replacement therapy in the Million Women Study. cancer during phase ii of the United Kingdom 347. Lu KH, Dinh M, Kohlmann W, et al. Gynecologic
Lancet. 2007;369(9574):1703–1710. Familial Ovarian Cancer Screening Study. J Clin cancer as a “sentinel cancer” for women with heredi-
310. Havrilesky LJ, Moorman PG, Lowery WJ, et al. Oncol. 2017;35(13):1411–1420. tary nonpolyposis colorectal cancer syndrome. Obstet
Oral contraceptive pills as primary prevention for 329. Moyer VA, Force USPST. Screening for ovarian Gynecol. 2005;105(3):569–574.
ovarian cancer: a systematic review and meta-analysis. cancer: U.S. Preventive Services Task Force reaf- 348. Barakat RR, Gilewski TA, Almadrones L, et al.
Obstet Gynecol. 2013;122(1):139–147. firmation recommendation statement. Ann Intern Effect of adjuvant tamoxifen on the endometrium
311. Beral V, Doll R, Hermon C, Peto R, Reeves G. Med. 2012;157(12):900–904. in women with breast cancer: a prospective study
Ovarian cancer and oral contraceptives: collaborative 330. U.S. Food and Drug Administration (FDA). The using office endometrial biopsy. J Clin Oncol. 2000;
reanalysis of data from 45 epidemiological studies FDA Recommends Against Using Screening Tests for 18(20):3459–3463.
including 23,257 women with ovarian cancer and Ovarian Cancer Screening: FDA Safety Communica- 349. Gerber B, Krause A, Muller H, et al. Effects
87,303 controls. Lancet. 2008;371(9609):303–314. tion; 2016. http://www.fda.gov/MedicalDevices/ of adjuvant tamoxifen on the endometrium in
Screening and Early Detection  •  CHAPTER 23 398.e7
398.e7

postmenopausal women with breast cancer: a Force recommendation statement. JAMA. 2016; and screening to reduce melanoma incidence and
prospective long-term study using transvaginal 316(4):429–435. mortality: The time is now. CA Cancer J Clin. 2016.
ultrasound. J Clin Oncol. 2000;18(20):3464–3470. 358. Tsao H, Weinstock MA. Visual inspection and the US 368. Boniol M, Autier P, Gandini S. Melanoma mortality
350. Rijcken FE, Mourits MJ, Kleibeuker JH, Hollema Preventive Services Task Force recommendation on following skin cancer screening in Germany. BMJ
H, van der Zee AG. Gynecologic screening in skin cancer screening. JAMA. 2016;316(4):398–400. Open. 2015;5(9):e008158.
hereditary nonpolyposis colorectal cancer. Gynecol 359. Schneider JS, Moore DH, Mendelsohn ML. Screen- 369. Scoggins CR, Ross MI, Reintgen DS, et al. Gender-
Oncol. 2003;91(1):74–80. ing program reduced melanoma mortality at the related differences in outcome for melanoma
351. Dove-Edwin I, Boks D, Goff S, et al. The outcome Lawrence Livermore National Laboratory, 1984 to patients. Ann Surg. 2006;243(5):693–698, discussion
of endometrial carcinoma surveillance by ultrasound 1996. J Am Acad Dermatol. 2008;58(5):741–749. 698–700.
scan in women at risk of hereditary nonpolyposis 360. Katalinic A, Waldmann A, Weinstock MA, et al. 370. Hoorens I, Vossaert K, Pil L, et al. Total-body
colorectal carcinoma and familial colorectal carci- Does skin cancer screening save lives?: an obser- examination vs lesion-directed skin cancer screening.
noma. Cancer. 2002;94(6):1708–1712. vational study comparing trends in melanoma JAMA Dermatol. 2016;152(1):27–34.
352. Lindor NM, Petersen GM, Hadley DW, et al. mortality in regions with and without screening. 371. Watts CG, Cust AE, Menzies SW, Mann GJ,
Recommendations for the care of individuals Cancer. 2012;118(21):5395–5402. Morton RL. Cost-effectiveness of skin surveillance
with an inherited predisposition to Lynch syndrome: 361. Breitbart EW, Waldmann A, Nolte S, et al. Systematic through a specialized clinic for patients at high risk
a systematic review. JAMA. 2006;296(12): skin cancer screening in Northern Germany. J Am of melanoma. J Clin Oncol. 2017;35(1):63–71.
1507–1517. Acad Dermatol. 2012;66(2):201–211. 372. Curiel-Lewandrowski C, Kim CC, Swetter SM,
353. American Cancer Society (ACS). Endometrial 362. Breitbart EW, Choudhury K, Anders MP, et al. et al. Survival is not the only valuable end point in
Cancer: Early Detection, Diagnosis, and Staging; Benefits and risks of skin cancer screening. Oncol melanoma screening. J Invest Dermatol. 2012;132(5):
2017. https://www.cancer.org/cancer/endometrial- Res Treat. 2014;37(suppl 3):38–47. 1332–1337.
cancer/detection-diagnosis-staging.html. Accessed 363. Eisemann N, Waldmann A, Geller AC, et al. Non- 373. Guy GP Jr, Machlin SR, Ekwueme DU, Yabroff
June 16, 2017. melanoma skin cancer incidence and impact of skin KR. Prevalence and costs of skin cancer treatment
354. Huang M, Sun C, Boyd-Rogers S, et al. Prospective cancer screening on incidence. J Invest Dermatol. in the U.S., 2002–2006 and 2007–2011. Am J Prev
study of combined colon and endometrial cancer 2014;134(1):43–50. Med. 2015;48(2):183–187.
screening in women with lynch syndrome: a patient- 364. Hubner J, Waldmann A, Geller AC, et al. Interval 374. Rogers HW, Weinstock MA, Feldman SR, Coldiron
centered approach. J Oncol Pract. 2011;7(1):43–47. cancers after skin cancer screening: incidence, BM. Incidence estimate of nonmelanoma skin cancer
355. Tripp MK, Gershenwald JE, Davies MA, et al. tumour characteristics and risk factors for cutaneous (keratinocyte carcinomas) in the U.S. population,
Assessment of compliance with Texas legislation melanoma. Br J Cancer. 2017;116(2):253–259. 2012. JAMA Dermatol. 2015;151(10):1081–
banning indoor UV tanning by minors. JAMA 365. Stang A, Garbe C, Autier P, Jockel KH. The many 1086.
Dermatol. 2016. unanswered questions related to the German skin 375. Wernli KJ, Henrikson NB, Morrison CC, Nguyen
356. Brunssen A, Waldmann A, Eisemann N, Katalinic cancer screening programme. Eur J Cancer. 2016; M, Pocobelli G, Blasi PR. Screening for skin cancer
A. Impact of skin cancer screening and secondary 64:83–88. in adults: updated evidence report and systematic
prevention campaigns on skin cancer incidence and 366. Stang A, Jockel KH. Does skin cancer screening review for the US Preventive Services Task Force.
mortality: a systematic review. J Am Acad Dermatol. save lives? A detailed analysis of mortality time JAMA. 2016;316(4):436–447.
2016. trends in Schleswig-Holstein and Germany. Cancer. 376. Shaikh WR, Geller A, Alexander G, et al. Developing
357. US Preventive Services Task Force (USPSTF), 2016;122(3):432–437. an interactive web-based learning program on skin
Bibbins-Domingo K, Grossman DC, et al. Screen- 367. Tripp MK, Watson M, Balk SJ, Swetter SM, cancer: the learning experiences of clinical educators.
ing for skin cancer: US Preventive Services Task Gershenwald JE. State of the science on prevention J Cancer Educ. 2012;27(4):709–716.
Nicotine Dependence: Current Treatments 24 
and Future Directions
Jeffrey M. Engelmann, Maher Karam-Hage, Vance A. Rabius,
Jason D. Robinson, and Paul M. Cinciripini

S UMMARY OF K EY P OI NT S
Prevalence of Tobacco Use and (e.g., identifying triggers and number of randomized controlled
Nicotine Dependence in Patients managing withdrawal), quitlines, and trials of smoking cessation
With Cancer self-help material (e.g., booklets and treatments have been conducted
• Approximately one in six American videos). among patients with cancer. These
adults is a current smoker. • Nonpharmacologic approaches are reports suggest that a combination
• Smoking accounts for one-third of all popular but yield relatively low quit of medications and a behavioral
cancer deaths. rates if used alone. approach are needed to make a
• In general, patients with cancer have • Pharmacologic treatments approved difference.
a higher dependence on nicotine and by the US Food and Drug • Patients with cancer who use
are more likely to be smokers or Administration, including nicotine tobacco should be treated according
ex-smokers. replacement therapies (e.g., to evidence-based treatment
• Approximately 15.1% of adult cancer transdermal patch, gum, nasal spray, guidelines, with particular attention
survivors are current smokers. inhaler, and lozenge) and bupropion, paid to tailoring education about
Biologic Characteristics and have been shown to double smoking their disease-tobacco link,
Genetics cessation rates compared with pharmacotherapy, comorbid
• The reward pathway and dopamine placebo. Varenicline has efficacy medical and psychiatric disorders,
are involved in the use and abuse superior to that of nicotine and family and household tobacco
of and dependence on all such replacement therapies and use.
substances, including nicotine. bupropion. Barriers to Cessation Treatment
• Pharmacogenetics research has the Smoking Among Cancer Patients in the Oncology Setting
premise to tailor treatment to • About half of patients with cancer • Health care providers have limited
enhance efficacy and reduce continue to smoke after diagnosis, time and expertise to address
toxicity. even though tobacco use smoking among patients with
Diagnosis and Evaluation complicates cancer treatment, cancer, and patients may have
• Several instruments measure reduces survivorship rates, comorbid substance use or
nicotine dependence among increases the risk for a second dependence or other emotional and
cigarette smokers; among them are primary tumor, and diminishes mental disorders that undermine
the Fagerström Test for Nicotine quality of life. their ability to quit smoking.
Dependence (FTND) and Heaviness • Few studies have examined • Systems-level changes and tailored
of Smoking Index (HSI). However, predictors of continued smoking treatment approaches are needed to
the evaluation and final among patients with cancer, but identify all tobacco users, lower the
recommendation are clinical, and some studies have reported on such rate of persistent tobacco use, and
the treatment plan must be factors as nonsmoking-related reduce recidivism among patients
individualized. cancers, comorbid depression, and with cancer.
poor prognosis.
Current Treatment • Several retrospective studies have
Recommendations shown the detrimental effect of
• Nonpharmacologic treatments continuing to smoke on cancer
include behavioral counseling treatment outcomes. A limited

399
400 Part I: Science and Clinical Oncology

Tobacco use has become a worldwide epidemic, and cigarettes are the a diagnosis of mental health or substance use disorders, those who
most common form of tobacco consumed. Smoking results in some are under the poverty level of income, and the unemployed.16 It is
of the deadliest yet most preventable diseases, such as respiratory of importance to note that level of education is inversely related to
diseases, cardiovascular diseases, and cancer.1 Tobacco use accounts smoking rates.17
for at least 30% of all cancer deaths in the United States, and smoking Although the trend for smoking among patients older than 45 years
has been causally linked to 18 different cancers, including lung, head who are recovering from cancer has decreased to about the same as for
and neck, esophageal, pancreatic, bladder, kidney, cervical, endometrial, the general population, the prevalence among survivors in the younger
and gastric cancers and acute myeloid leukemia.2,3 The scare of a age group is still high. Around 40.4% of persons 18 to 44 years of
diagnosis of cancer often impels smokers to quit, and although some age in the cancer survivors cohort still smoke cigarettes,18 compared
people resume use of tobacco once their cancer is in remission, others with 24.4% of persons 18 to 24 and 24.1% of persons 25 to 44
manage to avoid a relapse. years of age in the general population.13 Nevertheless, one potential
The high recidivism rates could be attributed to comorbid conditions confounding factor contributing to these statistics is the high mortal-
and to the neurobiology of tobacco addiction. Tobacco addiction is ity rate in patients with smoking-related tumors. Therefore smokers
a complex phenomenon. Nicotine receptors are spread throughout who would survive less lethal cancers are counted within the younger
most areas of the brain, and nicotine consumption activates the reward age group.19
pathway, as do all other substances of addiction.4 In addition, several Smoking cessation rates in the cancer population seem highly
other neurotransmitter systems are implicated in the process of dependent on the disease (tumor) site and its relationship to smoking.20
establishing an addiction to nicotine.5 After years of tobacco consump- As an example, around 80% of patients with lung cancer and 65%
tion, complex neuronal involvements and adaptations develop, rendering of patients with head and neck cancer were reported to have stopped
the extinguishing of tobacco addiction a challenging task. However, smoking in the year after their treatment21,22 compared with 31% of
pharmacologic and behavioral treatments have proven to be essential patients with bladder cancer.23 Other factors increase the likelihood
and highly effective in comparison with other medical treatments of of smoking cessation among patients with cancer, including having
chronic diseases.6 In recent years, individualized treatment has become a good prognosis, having spent a long time in the hospital (a smoke-free
the gold standard, with hopes that advancement in genetics will allow environment), and having treatment-related adverse effects. In particular,
us to eventually match the best treatment to each individual profile.7 adverse effects of treatment such as nausea, difficulty breathing, facial
Unfortunately, multiple hurdles and obstacles remain in extending surgery, or difficulty swallowing may make it harder or more difficult
basic treatment to all smokers and tobacco users, including those to smoke. Consequently, physicians should pay attention to the
diagnosed with cancer and cancer survivors.8 This chapter summarizes particular smoking-cessation pattern in patients with cancer. Typically,
these aspects of tobacco use disorder and their implication for patients cancer survivors have an increased chance of a delayed relapse to
with cancer. For the purpose of this chapter, the newer term tobacco smoking 1 to 6 months after treatment,24 in contrast to persons in
use disorder is used interchangeably with older ones such as nicotine the general population, who generally relapse within the first week
dependence (ND) and tobacco addiction. or so of cessation. Unfortunately, it has been reported that some
patients with lung cancer who have already abstained from smoking
EPIDEMIOLOGY AND TOBACCO USE IN before their diagnosis tend to return to smoking once they are informed
PATIENTS WITH CANCER of their cancer.25 Many studies have been published on the benefits
of smoking cessation in patients with cancer, with many focusing on
Tobacco use, in particular smoking cigarettes, began an upward trend cancer treatment–related outcomes (Table 24.1). Unfortunately, almost
around the beginning of the 20th century. This trend has been mostly all published studies to that effect have methodological limitations,
attributed to James Buchanan “Buck” Duke, who introduced the such as use of retrospective analyses, nonstandardized tobacco assess-
mechanized manufacturing of cigarettes and used aggressive marketing ment, no biochemical confirmation, and no tracking of behavior after
techniques,9 in addition to other societal forces and circumstances diagnosis. Addressing smoking behavior at the time of diagnosis and
such as the free cigarettes provided with daily rations to all enlisted throughout treatment and recovery is thus essential for persons with
American soldiers during World Wars I and II.10 That upward trend cancer, constituting a continuum of potential teachable moments (i.e.,
continued until a pivotal moment: the release of the landmark first health events that might motivate individuals to adopt risk-reducing
Surgeon General’s report on smoking and health in 1964,11 at which behaviors).26
point the evidence from more than 7000 scientific publications Another opportunity for teachable moments arises during lung
on harm from tobacco smoke was put together and received the cancer screening using low-dose computed tomography (LDCT). The
endorsement of Surgeon General Luther Terry, the highest officer US Preventive Services Task Force recommends LDCT screening in
for public health within the US government. That report had an individuals 55 to 80 years old with a 30 pack-year smoking history
enduring effect on public beliefs and attitudes about tobacco. As an who currently smoke or who have quit within the past 15 years.27
example, a Gallup survey conducted in 1958 found that only 44% A systematic review of two randomized controlled trials (RCTs)
of Americans believed smoking caused cancer, whereas 70% believed and three observational studies found that although LDCT itself
so by 1969, 5 years after the report was published. The belief that does not influence smoking behavior, positive results (i.e., finding
smoking causes cancer continued to rise, and the rate of belief reached a nodule or a tumor) were associated with increased abstinence.28
94% by 1990.12 Furthermore, since 1964 a gradual and almost annual A more recent analysis of 18,840 current and former smokers from
decrease in smoking rates has occurred in the United States, with the the National Lung Screening Trial found that false-positive screening
rate reaching 15.2% in 2015.13 This lower rate nonetheless encompasses results were associated with modest increases in abstinence among
45 million Americans who, more than 50 years after the landmark smokers over the course of a 5-year follow-up period.29 These studies
report, continue to use tobacco despite the clear knowledge of harm have similar limitations to other studies about smoking cessation in
from its consumption. Other Surgeon General reports subsequently cancer patients, including nonstandardized assessment of smoking
solidified the case against tobacco consumption, and to date 37 of behavior and no biochemical verification of abstinence. Therefore more
those reports have been dedicated to tobacco (most recently the 50th research is needed on the effectiveness of lung cancer screening as a
anniversary report14), focusing on specific topics such as nicotine addic- teachable moment for smoking cessation. It will also be important to
tion, secondhand smoke, minorities, women, and youth.15 Although study the effectiveness of offering evidence-based smoking cessation
these efforts have improved public health, disparities in health care interventions at the time of lung cancer screening, which have the
unfortunately extend to tobacco consumption, as evidenced by higher potential to further motivate smokers to quit beyond lung cancer
smoking rates among American Indians/Alaskan Natives, persons with screening itself.30
Nicotine Dependence: Current Treatments and Future Directions  •  CHAPTER 24 401

Table 24.1 Continued Smoking Versus Cessation as It Relates to Cancer Treatment Outcomes in


Retrospective Studies
Sample
Authors (Journal) Year Cancer Outcome if Continued Smoking Size Tumor Site
Fortin et al242 (Int J Radiat Oncol Biol Phys) 2009 Smoking and drinking at baseline were associated with poor 1871 Head and neck
outcomes
Rades et al243 (Int J Radiat Oncol Biol Phys) 2008 Smoking during radiation had a significant effect on 181 Lung
locoregional control
Marin et al244 (Plast Reconstr Surg) 2008 Serum cotinine concentration greater than 10 ng/mL may 89 Head and neck
predict an increased risk of wound complication
Chen et al245 (BJU Int) 2007 Higher recurrence rate for patients with non–muscle- 297 Bladder
invasive bladder cancer
Tammemagi et al246 (Chest) 2004 Worse survival in smokers 1155 Lung
Garces et al228 (Chest) 2004 Smoking after a lung cancer diagnosis negatively affects 1506 Lung
quality-of-life scores
Pytynia et al247 (J Clin Oncol) 2004 Threefold increases in risk for overall death, death as a result 100 Head and neck
of disease, and recurrence of disease
Fox et al248 (Lung Cancer) 2004 Poorer prognosis for survival after radiation therapy of non– 237 Lung
small cell cancer
Nordquist et al249 (Chest) 2004 Worse survival 654 Lung
Videtic et al250 (J Clin Oncol) 2003 Poor survival rates if patients continued to smoke during 215 Lung
radiation and chemotherapy
Chelghoum et al251 (Ann Oncol) 2002 Lower survival in patients with acute myeloid leukemia by 643 Leukemia
shortening complete remission duration and subsequent
survival; severe infections during aplasia
Kreuzer et al252 (Br J Cancer) 2000 Higher risk of developing lung cancer among men than 9792 Lung
among women
Marshak et al253 (Int J Radiat Oncol Biol Phys) 1999 Smoking correlated with decreased local control 207 Glottis
Fleshner et al254 (Cancer) 1999 Diminished recurrence-free survival 286 Bladder
Daniell et al255 (J Urol) 1995 Greater 5-yr tumor-specific mortality rate with stage D2 359 Prostate
disease
Daniell et al256 (Am J Clin Pathol) 1993 More often advanced-stage disease (stages II–IV) 157 Endometrium
Risch et al257 (Am J Epidemiol) 1993 In both sexes, a greatly elevated risk of developing lung 845 Lung
cancer, and an appreciably stronger risk for females than for
males
Rugg et al258 (Br J Radiol) 1990 A highly significant correlation of cancer during and/or after 41 Head and neck
treatment
Archimbaud et al259 (Cancer) 1989 Associated with early blast crisis and short survival 173 Leukemia
Daniell260 (Cancer) 1988 Tobacco and obesity potentiate the early spread of 623 Breast
malignant disease
Daniell261 (Cancer) 1986 More advanced stage of tumors 392 Colon
Phillips et al262 (Cancer) 1985 Marked depression in natural killer activity that was 68 Breast
comparable with that of patients with carcinoma of the lung
Stevens et al263 (Arch Otolaryngol) 1983 Smoking after diagnosis had a fourfold increase in the 200 Head and neck
recurrence rate
Daniell264 (N Engl J Med) 1980 Fared less well than nonsmokers with breast cancer 78 Breast
Hinds et al265 (J Natl Cancer Inst) 1982 Greater probability of dying during the 12 mo after 223 Lung
diagnosis
Johnston-Early et al266 (JAMA) 1980 Decreased survival 112 Lung

REWARD PATHWAY AND BIOLOGIC dopamine release in the nucleus accumbens and the ventral tegmental
area. This stimulation typically occurs within 10 seconds of smoking
CHARACTERISTICS OF ADDICTION a cigarette.31,32 It has been established that natural rewards such as
Reward Pathway food consumption, social affiliation, and sexual activity, which are
linked to survival of the individual or species, normally activate these
Like most drugs associated with abuse and dependence, nicotine two central areas of the reward pathway within the brain. Furthermore,
stimulates a variety of receptors in the brain, in particular nicotinic the reward pathway projects from the nucleus accumbens and ventral
acetylcholine receptors (nAChRs). These nAChRs are spread throughout tegmental area to the prefrontal cortex, the amygdala, and the olfactory
most areas of the brain. Nicotine use leads to a rapid increase in tubercle (Fig. 24.1), leading to a complex interaction among the
402 Part I: Science and Clinical Oncology

GENETICS
VTA Genes Associated With Smoking, Nicotine
Dependence, and Lung Cancer
Prefontal
cortex Several recent genome-wide association (GWA) and candidate gene
studies have revealed significant associations among smoking, ND,
and lung cancer in a region of interest on the long arm of chromosome
15 (15q24/15q25.1), encompassing the nAChR CHRNA3-A5-B4
subunit cluster. Neuronal nAChRs are pentameric ligand-gated channels
Nucleus
composed of alpha and beta subunits,41 several of which contribute
accumbens
to nicotine-stimulated dopamine release in the striatum, a region
associated with reward learning and vital to the development of
substance dependence.42 The CHRNA3-A5-B4 subunit cluster dem-
onstrates extensive linkage disequilibrium, and the single nucleotide
polymorphisms on that cluster are frequently correlated. Some of the
Figure 24.1  •  The reward pathway, consisting of the nucleus accumbens statistically strongest associations with ND in persons of European
and ventral tegmental area (VTA) with projections to the prefrontal cortex. or African ancestry have been obtained for the nonsynonymous
(Modified from National Institute on Drug Abuse. The Neurobiology of Drug CHRNA5 single-nucleotide polymorphism rs16969968.43–51 Other
Addiction. Available at http://www.nida.nih.gov/pubs/teaching/Teaching2/ gene variants in this region (CHRNA3-A5-B4) identified by GWA
Teaching.html.) and candidate gene studies as being significant contributors to the
risk of smoking, ND, and lung cancer include CHRNA3 rs1051730,48,52–55
CHRNA3 rs578776,43,45,50 and AGPHD1 rs8034191.43,52,54 Finally,
candidate gene studies have identified associations between ND and
genetic markers associated with dopamine expression (ANKK1,54,56–58
reward, its saliency, and the ability to inhibit the intake of a drug. In DRD2,58,59 and SLC6A359–62), serotonin expression (SLC6A4),63–65 and
addition to dopamine, several other brain systems (neurotransmitters, nicotine metabolism (CYP2A6).55,66
receptors, and pathways) have been implicated in addiction, including
the γ-aminobutyric acid, glutamate, serotonin, norepinephrine, can- Genes Predicting Treatment Outcome
nabinoid, opioid, and cholinergic systems.33–35 It is important to note
that these major neurotransmitter systems in the brain are in constant A growing body of genome-wide association studies (GWASs) and
and dynamic interplay, possibly resulting in different types of neuro- candidate gene studies have examined genetic predictors of responsive-
adaptations that lead to and maintain the addiction to a substance in ness to smoking-cessation therapies (see references 51 and 67–69 for
a given individual.5 reviews). Cessation outcome for nicotine replacement therapy (NRT)
has been found to be associated with several polymorphisms in the
Neuronal Adaptation D2 receptor gene (DRD2), including C957T rs6277,70 −141C ins/del
rs1799732,70 and ANKK1 (rs1800497).71–73 Other markers that influence
A pleasurable sensation from the activation of the reward pathway dopamine reuptake and expression, including COMT rs468074,75 and
is associated with the acute use of an addictive substance such as a variable number tandem repeat (VNTR) in DRD4 C-521T,76 have
nicotine. However, repeated administration of a substance, in this been shown to predict cessation outcome for NRT. Genes associated
case nicotine, can lead to increased tolerance, which in turn pro- with the opioid (OPRM1),77–79 serotonergic (SLC6A4),77,80,81 canna-
duces a state of withdrawal in the absence of nicotine. Tolerance binoid,82,83 and CHRNA3-A5-B484 pathways have also been associated
and withdrawal are the physiologic hallmarks of dependence and are with NRT outcome.
thought to be the result of neuroadaptive effects occurring within Similarly, variants of many of these same markers have been associ-
the brain.36 It is interesting to note that the chronic use of drugs ated with successful smoking cessation treatment using bupropion,
of abuse and dependence, including nicotine, seems to result in a including ANKK1,85–87 DRD2 −141C ins/del,70 COMT,88 DRD4,89
generalized decrease in dopaminergic neurotransmission. Although SLC6A3,90 CYP2B6,91 and the CHRNA3-A5-B4 subunit cluster.92–94
this decrease can be a prodrome for vulnerability to addiction, it is Varenicline, the most recently approved smoking cessation
likely to be a homeostatic response to the intermittent yet repetitive pharmacotherapy, is the next target for pharmacogenetic inquiry. An
increases in dopamine induced by the frequent and sustained use of analysis of 1175 smokers who received varenicline, bupropion, or
such drugs.37 placebo found that abstinence was associated with multiple CHRNB2,
Sensitization and counteradaptation are two of the major neu- CHRNA7, and CHRNA4 subunit genes for persons given varenicline
roadaptive models that have been proposed to explain how changes and with CYP2B6 for persons given bupropion.7 However, a more
that start in the reward pathway may result in the development of recent analysis of 1026 smokers found that CHRNA5-A3-B4 genetic
substance dependence. Sensitization can be thought of as the gradual variants were not associated with smoking cessation outcome regard-
increase in “wanting” a drug after intermittent but repeated use, which less of treatment (placebo, NRT, or varenicline).50 Thus additional
then can facilitate the transition from occasional use to chronic use research is needed to identify reliable genetic predictors of varenicline
and tolerance.37,38 The counteradaptation model postulates that the treatment.
initial positive feelings of reward (resulting from the use of a drug) Recently, a genetically informed biomarker has been shown to
are followed by an opposing rather than synchronous development be predictive of smoking cessation.95 The nicotine metabolite ratio
of tolerance, as the brain’s homeostatic (counterbalance) response (NMR) is the ratio of 3′-hydroxycotinine to cotinine, which reflects
to the exposure. Other models of nicotine addiction are based on the activity of CYP2A6, the primary enzyme responsible for nicotine
mechanisms associated with capacity for or the lack of cognitive control metabolism. Retrospective studies have shown that slow metabolizers
and the development of reinforcement learning,39 in particular the of nicotine (i.e., those with NMRs that fall in the lowest quartile)
negative reinforcement associated with withdrawal from nicotine, have higher smoking cessation rates than normal metabolizers,96–98 but
which builds up with the reduction in negative effect from smoking that normal metabolizers are more likely to benefit from bupropion
a cigarette.40 treatment than slow metabolizers.99 In a large prospective study,100
Nicotine Dependence: Current Treatments and Future Directions  •  CHAPTER 24 403

1246 smokers were randomly assigned to receive varenicline, NRT, or PATIENT ASSESSMENT
placebo, stratified by NMR. Among normal metabolizers, varenicline
resulted in significantly higher abstinence rates than NRT, but among In the absence of specific biological markers, the diagnosis of nicotine
slow metabolizers, varenicline was not more efficacious than NRT. addiction remains a clinical one. The Diagnostic and Statistical Manual
Also, slow metabolizers reported significantly more side effects while on of Mental Disorders, fifth edition (DSM-5)119 uses universal criteria
varenicline versus placebo. This suggests that treating slow metabolizers for all substance use disorders (traditionally referred to as addiction,
with NRT and normal metabolizers with varenicline may maximize and referred to as dependence in previous editions of the DSM120),
the likelihood of smoking cessation while minimizing the risk of including nicotine. Because of their universality, the DSM-5 criteria
side effects. are not particularly specific to nicotine and therefore do not capture
In summary, GWASs and candidate gene studies point to a variety many of the peculiar aspects of tobacco use and ND (addiction). This
of genes that may be related to one or more smoking phenotypes, lack of specificity has led to the development of specific scales to
including ND, initiation, cessation, and possibly nicotine withdrawal. quantify ND. Traditionally, the six-item Fagerström Test for Nicotine
In all but a few studies, these relations have been studied in smokers Dependence (FTND)121 has been used, and more recently the two-item
of European decent. The areas of interest include nicotine cholinergic Heaviness of Smoking Index (HSI),122 which was derived from the
receptor genes (CHRNA3-A5-B4; CHRNA2, CHRNA4, CHRNA6, first two items of the FTND.
CHRNB1, CHRNB2, and CHRNB3), nicotine and drug metabolism Despite their limitations, the DSM-5 criteria offer ease of use for
genes (CYP2A6 and CYP2B6) and associated biomarkers (NMR), clinicians because of their universality for all substances of dependence.
and dopaminergic signaling genes (DRD2-ANKK1, DRD4, COMT, According to the DSM-5, the diagnosis is made when someone meets
SLC6A3, and DBH). two or more of the 11 criteria for at least a year (the more criteria
that are met, the more severe is the disorder). As previously mentioned,
COMORBID CONDITIONS some criteria are more pertinent than others to nicotine. For instance,
after prolonged smoking the user develops nicotine tolerance and
The smoking rates among persons with no mental illness, past-month begins to exhibit withdrawal symptoms when nicotine is absent, often
mental illness, and lifetime mental illness have been reported to be overnight, largely because nicotine’s half-life in blood is 1 to 2 hours.123
22%, 34%, and 41%, respectively, which means that having a current Having tolerance and/or withdrawal used to be referred to as physiologic
or past mental disorder effectively doubles the likelihood of being a dependence in the DSM-IV-TR. In addition to tolerance and with-
smoker. Furthermore, in a nationally representative sample, it was drawal, nicotine may also be responsible for four other criteria for
reported that smokers with a mental disorder in the past month consume dependence in DSM-5: craving for cigarettes, loss of control over
44% of all cigarettes smoked.101 The evidence that smoking is more smoking (e.g., not being able to reduce or stop smoking, or smoking
prevalent in and closely linked with several psychiatric comorbidities, more than intended), compulsive smoking (e.g., smoking all day long
including abuse of other substances, suggests a shared biologic pathway and giving up important events or activities if there is no place to
between ND and these psychiatric and substance use conditions. For smoke), and continued smoking despite adverse consequences (e.g.,
example, several studies have demonstrated a positive correlation heart attack, emphysema, or cancer).
between smoking and alcohol use, abuse, or dependence, substance The FTND (Table 24.2) is one of the most widely recognized and
abuse or dependence, and other psychiatric disorders.101–108 Conversely, used scales for nicotine dependence. A person with three points or
the prevalence of lifetime alcohol dependence and/or drug abuse with
the adult smoker population is estimated to be 23% to 30%.109,110
Similarly, among current smokers, the lifetime rates of mood and
anxiety disorders have been reported to range from 33% to 46% and
from 12% to 26%, respectively, regardless of nicotine dependence.109 Table 24.2 Items and Scoring for Fagerström Test
Furthermore, among current smokers who are tobacco dependent, for Nicotine Dependence
even the 12-month prevalence of any mood or anxiety disorder is
reported to be 22%, a relatively high proportion.111 Adding support Question Answer Points
to the idea of a possible causal link between smoking and mental 1. How soon after you wake up do Within 5 min 3
health disorders is the fact that smokers also have an elevated risk of you smoke your first cigarette? 6–30 min 2
a first onset of major depression, panic disorder, or generalized anxiety 31–60 min 1
disorder.112–116 Cognitive dysfunction has also been highly linked to After 60 min 0
smoking, as evidenced by the odds ratio comparing persons who have 2. Do you find it difficult to refrain Yes 1
ever smoked with persons who have never smoked, and smoking from smoking in places where it is No 0
is positively related to the number of attention-deficit/hyperactivity forbidden (e.g., in church, at the
disorder (ADHD) symptoms. There seems to be an effect of early library, in a cinema)?
onset on the severity of ADHD or vice versa, because lifetime regular 3. Which cigarette would you hate The first one in 1
smoking has an inverse relationship between the number of ADHD most to give up? the morning
symptoms and the age at onset. In addition, a positive relationship All others 0
between the number of symptoms and the number of cigarettes smoked 4. How many cigarettes per day do ≤10 0
has been observed.117 The aforementioned evidence of co-occurrence you smoke? 11–20 1
supports the importance of screening for and treating mental health 21–30 2
disorders among smokers, whether the relationship is causal or a ≥31 3
simple correlation. Treating these disorders may increase the ability 5. Do you smoke more frequently Yes 1
to focus on quitting smoking, or at least reduce the impact of nega- during the first hours after waking No 0
tive affect or comorbid mental disorders on a person’s ability to quit than during the rest of the day?
using tobacco, and may have an impact on his or her resilience with 6. Do you smoke if you are so ill that Yes 1
regard to maintaining tobacco abstinence. Treating these disorders you are in bed most of the day? No 0
is of particular importance among patients with cancer and cancer
survivors, because higher levels of confidence about the ability to quit From Heatherton TF, Kozlowski LT, Frecker RC, Fagerström KO. The Fagerström Test
smoking were found to be related to lower depression scores and lower for Nicotine Dependence: a revision of the Fagerström Tolerance Questionnaire. Br J
tumor stage.118 Addiction. 1991;86(9):1119-1127.
404 Part I: Science and Clinical Oncology

more out of a possible maximum of 10 points on the FTND is review of all tobacco treatment–related literature, was last updated
customarily considered to be nicotine dependent.124,125 The HSI consists in 2008.6 The key points are presented in an executive summary
of two items from the FTND (“How soon after you wake up do you in Box 24.1. These approaches are summarized and discussed in
smoke your first cigarette?” and “How many cigarettes do you smoke the following section, specifically their applicability to the cancer
per day?”).122 These tools measure physiologic dependence (i.e., tolerance population.
and withdrawal) reliably well. However, they do not capture the
psychosocial and behavioral dimensions of ND.126 Pharmacologic Interventions

TREATMENT OF TOBACCO USE Some persons are not interested in or lack the motivation to quit
smoking, and some persons are not ready for change. Even in these
Tobacco treatment among patients with cancer needs to be as intensive instances, pharmacologic treatments have been shown to increase the
and tailored as possible. Hence, if treatment programs are to be odds of reducing the number of cigarettes per day by half.6 This
comprehensive and to make a difference, they ought to provide at a decrease in cigarettes smoked can in return positively influence patients
minimum both behavioral- and medication-based interventions.127 by increasing their self-efficacy and motivation to quit.139
Comprehensive treatments, by definition, would need to address The FDA has approved the use of several NRTs, among them the
the biologic, psychologic, and social aspects of ND. Moreover, these patch, nasal spray, buccal inhaler, gum, and lozenge. The abstinence
approaches ought to be on a long-term basis, because tobacco addiction results for NRTs are compelling and largely encouraging based on
is a chronic disease that is often characterized by multiple relapses before hundreds of studies.139 Despite that, recent debate over their long-term
full remission is reached.128 The main components of a biopsychosocial efficacy is taking place, in particular now that they are available over
treatment are behavioral therapies and social support in conjunc- the counter.140 However, data on using a combination of a patch with
tion with medications. Among the various therapies, motivational other intermittent NRTs show that combining NRTs is more effective
interviewing, skill-building, problem-solving, and cognitive behavioral than use of single NRT.141 Longer-term NRT use can be effective; a
therapies have been empirically supported.6 Although the main first-line recent study demonstrated the efficacy of NRTs in sustaining abstinence
medications that have been approved by the US Food and Drug when used continuously and up to 24 weeks.142
Administration (FDA) are NRTs, bupropion (Zyban), and varenicline Nicotine is the key ingredient in NRTs, with the premise that
(Chantix), the second-line medications clonidine and nortriptyline have replacing the addictive substance in cigarettes helps the user to quit.
been used off-label and tested in clinical trials with similar positive Nicotine in NRTs is at a much lower dose and is delivered through
results, although with increased side effect profiles. the venous system, compared with a higher dose from smoking
Among several other medications that have been or are being a cigarette that has an arterial (direct) delivery to the brain. The
researched, three have shown promising preliminary results but have intent with NRTs is to reduce cravings and the withdrawal effects of
faced different challenges: topiramate, rimonabant, and nicotine tobacco cessation. Interesting to note, and despite some controversies,
vaccines. Although a care provider may prescribe any available medica- NRTs have been found to be effective with and without the aid
tions off-label, to date, none of the aforementioned three medications of a therapeutic support.143 However, one should not forget that
is FDA approved for smoking cessation purposes, and only topiramate management of expectations and patient education before initiation
is available in the United States. However, their importance lies in of pharmacologic treatment are key to the success of smoking cessation
their mechanisms of action, which may lead to improved or similar interventions. In addition, NRTs are relatively safe to use regardless of
agents in the future. Topiramate, a voltage-dependent sodium channel the individual’s smoking status and may be used while the individual
blocker, augments γ-aminobutyric acid, antagonizes glutamate, and is still smoking.144 Most NRT studies did not identify any noteworthy
inhibits carbonic anhydrase activity, and has FDA label indication as nicotine toxicity or adverse events; however, the results of other studies
an adjunct treatment for seizure disorders and migraine headaches.129 showed that NRT used before quitting helped reduce the number of
Topiramate is reported to have a beneficial effect in the treatment of cigarettes per day by half even in persons who were not interested
alcohol dependence and ND treated independently130,131 but does not in quitting.145–148 Some smoker characteristics (e.g., the metabolism
seem to have an effect on nicotine dependence among abstinent rate of nicotine) can affect response to NRTs, suggesting that the
alcohol-dependent patients during their first 6 months of abstinence tailored use of NRT for subgroups of smokers could improve NRT
from alcohol132; furthermore, it has several adverse effects that limit efficacy.149 For example, the level of nicotine dependence affects the
its use.133 Rimonabant is a selective CB1 endocannabinoid receptor response to NRTs, with more dependent smokers showing higher rates
antagonist reported to have beneficial effects for the treatment of of quitting when they receive higher doses of NRTs.150,151 NRTs are
obesity134 and ND.135 It was shown to be relatively efficacious for an important treatment option for patients with cancer and have been
smoking cessation compared with placebo (odds ratio 1.6)136 in large reported to have similar efficacy to that among other populations of
multinational trials. It was available in Europe from mid-2006 until smokers.127 However, some animal and laboratory cell culture studies
January 2009; then the European Medicines Agency recommended in normal and cancer tissue have shown that nicotine promotes tumor
its withdrawal because of neuropsychiatric adverse effects (i.e., depres- growth, whereas others report it to also have a tumor-promoting
sion and several reports of suicides).137 Although rimonabant is currently effect.152,153 Thus far, human studies have not found a significant causal
available in other countries, it did not receive FDA approval in the relationship between NRTs and cancer genesis,154 even after 5 years of
United States because of similar concerns about neuropsychiatric adverse NRT use.155,156
effects. Several vaccines against nicotine have been under study for
some years and have shown initial promising results in animal and Bupropion
early human trials. The most advanced stage III clinical multisite Bupropion was first introduced on the market as an antidepressant
human trial on one of the nicotine vaccines did not find it to be (Wellbutrin) before being found to be effective in the treatment of
different from placebo as a treatment for smoking cessation. However, tobacco dependence. This medication was consequently approved for
several newer methods to develop better vaccines are still being explored. and is currently used by smokers with or without depression (Zyban).157
The vaccine concept is important and merits further investigation To date, bupropion is the only antidepressant approved for smoking
because it may have a role in relapse prevention and as part of mul- cessation on the market; however, no controlled studies have been
tifaceted approach to smoking cessation that includes behavioral and published that examine the role of depression as a moderator of the
pharmacologic intervention.138 response to bupropion in depressed smokers. Bupropion is a relatively
The Clinical Practice Guideline for Treating Tobacco Use and well-tolerated medication, with dry mouth, insomnia, and fine tremor
Dependence (CPG-TTUD), which provides a comprehensive being the most common adverse effects. A meta-analysis of 44 controlled
Nicotine Dependence: Current Treatments and Future Directions  •  CHAPTER 24 405

Box 24.1.  TEN KEY GUIDELINE RECOMMENDATIONS FROM THE CLINICAL PRACTICE GUIDELINE
ON TREATING TOBACCO USE AND DEPENDENCE
The overarching goal of these recommendations is that clinicians contraindicated or with specific populations for which there is
strongly recommend the use of effective tobacco dependence insufficient evidence of effectiveness (i.e., pregnant women,
counseling and medication treatments to their patients who use smokeless tobacco users, light smokers, and adolescents).
tobacco and that health systems, insurers, and purchasers assist • Seven first-line medications (five nicotine and two non–nicotine-
clinicians in making such effective treatment available. based medications) reliably increase long-term smoking
1. Tobacco dependence is a chronic disease that often requires abstinence rates: bupropion sustained-release (SR), nicotine gum,
repeated intervention and multiple attempts to quit. However, nicotine inhaler, nicotine lozenge, nicotine nasal spray, nicotine
effective treatments exist that can significantly increase rates of patch, and varenicline.
long-term abstinence. • Clinicians also should consider the use of certain combinations
2. It is essential that clinicians and health care delivery systems of medications identified as effective in this guideline.
consistently identify and document tobacco use status and treat 7. Counseling and medication are effective when used by themselves
every tobacco user seen in a health care setting. for treating tobacco dependence. The combination of counseling
3. Tobacco dependence treatments are effective across a broad range and medication, however, is more effective than either alone. Thus
of populations. Clinicians should encourage every patient willing clinicians should encourage all individuals making a quit attempt
to make a quit attempt to use the counseling treatments and to use both counseling and medication.
medications recommended in this guideline. 8. Telephone quitline counseling is effective with diverse populations
4. Brief tobacco dependence treatment is effective. Clinicians should and has broad reach. Therefore both clinicians and health care
offer every patient who uses tobacco at least the brief treatments delivery systems should both ensure patient access to quitlines
shown to be effective in this guideline. and promote quitline use.
5. Individual, group, and telephone counseling are effective, and their 9. If a tobacco user currently is unwilling to make a quit attempt,
effectiveness increases with treatment intensity. Two components clinicians should use the motivational treatments shown in this
of counseling are especially effective, and clinicians should use guideline to be effective in increasing future quit attempts.
these when counseling patients making a quit attempt: 10. Tobacco dependence treatments are both clinically effective and
• Practical counseling (problem solving, skills training) highly cost-effective relative to interventions for other clinical
• Social support delivered as part of treatment disorders. Providing coverage for these treatments increases quit
6. Numerous effective medications are available for tobacco rates. Insurers and purchasers should ensure that all insurance
dependence, and clinicians should encourage their use by all plans include the counseling and medication identified as effective
patients attempting to quit smoking—except when medically in this guideline as covered benefits.
Reprinted from Fiore MC, Jaen CR, Baker TB, et al. Treating tobacco use and dependence: 2008 Update U.S. Public Health Service Clinical Practice Guideline executive
summary. 2008;53(9):1217-22. Updated March 4, 2015.

clinical trials showed bupropion to be twice as effective as placebo in twice per day, it was twice as effective as bupropion and three times
reaching end-of-treatment abstinence (8 weeks), as well as in long-term more effective than placebo at the end of treatment (12 weeks) and
cessation (pooled risk ratio = 1.62).158 at 1-year follow-up.163–165 An additional 12 weeks of randomized
Unfortunately, certain patients with cancer may be unable to use varenicline or placebo therapy (for a total of 24 weeks) was provided
bupropion because it is known to lower the seizure threshold and is in a double-blind design to patients who had abstained from smoking
contraindicated in persons with active seizures or a history of seizures, during the first 3 months of open-label varenicline treatment; 70%
head trauma, elevated liver enzymes, bulimia, or anorexia nervosa. It of those randomly assigned to receive varenicline stayed abstinent at
would be considered risky to give bupropion to patients with cancer the end of the continuation phase of 3 months (a total of 6 months
who have a lower threshold for seizures, such as those with metastases of treatment with varenicline) compared with 50% of persons randomly
to the brain or with low appetite or body weight as a result of cancer assigned to placebo. Furthermore, the 1-year follow-up abstinence
treatment. However, bupropion has been reported to be effective for rate (after medication treatment) was double that of patients who had
smoking cessation including in populations with mental disorders received only 3 months of varenicline (25% and 12%, respectively).166
such as schizophrenia,159 depression,160 and posttraumatic stress dis- Subsequent clinical trials have confirmed that varenicline is more
order.161 Moreover, research has shown that bupropion limits the weight efficacious for smoking cessation than bupropion or placebo,167–170
gain commonly associated with smoking cessation162 and restores sexual but a recent open-label study found no differences in efficacy among
functioning.161 These added advantages can increase its usefulness varenicline, nicotine patch, or combined NRT (nicotine patch plus
among cancer survivors with fatigue from chemotherapy or radiation nicotine lozenge), although adherence to treatment was very low,
or those with comorbid disorders, in addition to persons who are which may be the reason for the lack of difference in this particular
overweight or have sexual dysfunction. study.171 A recent open-label study of varenicline for smoking cessation
in 132 cancer patients found similar abstinence rates at end of treatment
Varenicline (40.2%) and a similar side effect profile to what is seen in the general
Varenicline, marketed as Chantix in the United States and Champix population.172 Although further study is needed, the findings of this
in other countries, is a non–nicotine-based medication for smoking study support the use of varenicline for smoking cessation in cancer
cessation that was introduced in 2006. Varenicline’s unique properties patients.
as a partial nAChR agonist (traditionally referred to as a mixed agonist- The most common adverse event reported was nausea, which is
antagonist) provide some relief from withdrawal symptoms (agonist reported by almost 30% of the persons using varenicline.165,163 Although
property) usually associated with tobacco cessation, while diminishing the nausea was mild or moderate in the majority of cases, this adverse
the pleasurable sensation from smoking (antagonist property). Initial effect limits the use of this drug by patients with cancer undergoing
clinical trials of varenicline showed that when it was used at 1 mg treatment because nausea is a common side effect of cancer treatments.
406 Part I: Science and Clinical Oncology

Other commonly reported adverse effects were vivid dreams and bupropion has gained attention as another option. This combination
flatulence. Less common were the neuropsychiatric events among the is of particular interest because it combines two effective medications
general population reported in the postmarketing phase to the FDA that have different mechanisms of action, with the possibility of
via the voluntary reporting tool MedWatch, with these events consisting potentiating each other. A particular advantage of this combination
mainly of difficulty with coordination, depressive symptoms, aggression, might be the beneficial effect of bupropion as an antidepressant in
irritability, and (more rarely) suicidal ideations.173 In light of these the mitigation of negative affect and neuropsychiatric effects resulting
reports, the FDA issued a black box warning for varenicline regarding from smoking cessation. A single-arm, open-label pilot study tested
neuropsychiatric events and commissioned the manufacturer to deepen the effectiveness of varenicline-bupropion combination in 38 smokers.
its data analyses and conduct postmarketing studies to directly compare The point prevalence smoking abstinence rates were as high as 70% at
psychiatric versus nonpsychiatric populations to determine the 3-month follow-up and almost 60% at 6-month follow-up.186 Three
magnitude of these adverse events and their potential link to varenicline. randomized clinical trials followed, and their results seem to support
The findings of three of those studies were recently published. The the efficacy of combination therapy among heavy smokers (highly
first study reported on varenicline’s efficacy and adverse effect profile addicted to nicotine), and in particular among male heavy smokers.
among stable patients with schizophrenia. Although the quit rates The first study of 506 smokers randomized to varenicline-bupropion
were somewhat lower than expected (19% with varenicline versus 5% combination versus varenicline monotherapy found that smokers given
with placebo), no exacerbation of the schizophrenia symptoms occurred, combination therapy had significantly higher prolonged abstinence
and varenicline was well tolerated.174 The second study was a placebo- rates at the end of treatment (53.0% for combination versus 43.2%
controlled study of the safety and efficacy of varenicline in those who for monotherapy) and at 6-month follow-up (36.6% for combination
had a history of or currently stable depression; no difference was versus 27.6% for monotherapy), but not at 1-year follow-up (30.9%
found between the groups in terms of new occurrence of neuropsy- for combination versus 24.5% for monotherapy).187 The second RCT,
chiatric adverse events.175 The third and largest study to date was a in which pretreatment nicotine patch nonresponders were randomized
placebo-controlled study of the safety and efficacy of varenicline, to receive the varenicline-bupropion combination versus varenicline
bupropion, or nicotine patch in 4116 smokers with psychiatric disorders monotherapy, reported increased abstinence rates in the combination
and 4028 smokers without psychiatric disorders. The study did not group of up to 6 months among male and highly dependent smokers.188
show a significant increase in neuropsychiatric adverse events attributable The third study consisted of 122 male heavy smokers and consolidated
to varenicline, bupropion, or nicotine patch compared with placebo further the findings of the second study, showing a significance of
but did find, as prior studies have shown, that varenicline was more efficacy for the combination in this group.189 Other controlled trials are
effective for smoking cessation than bupropion, nicotine patch, or underway to further test the efficacy and safety of this combination.
placebo.170 Before these studies, two meta-analyses found no evidence
of increased neuropsychiatric side effects in smokers taking vareni- Second-Line Medications
cline.176,177 As a result of these studies, in December 2016 the FDA Smokers who cannot use or tolerate first-line medications are usually
removed the black box warning regarding neuropsychiatric adverse referred to second-line medications such as clonidine or nortripty-
effects. Furthermore, varenicline has been reported to decrease alcohol line.158,190,191 Although several clinical trials have already proven their
consumption among quitting smokers,178 which is of particular efficacy, these agents do not have FDA approval for treatment of
importance given that alcohol and tobacco use disorders are common tobacco use because they are older medications already available in
comorbidities among patients with head and neck cancer, with decreased generic forms. Specifically, clonidine has shown superiority to placebo
risk after cessation.179,180 in two meta-analyses, with odds ratios ranging between 2.0 and 2.4.190
A Cochrane review of partial agonists on nicotine receptors con- Nevertheless, it can be administered three times per day and has a
sidered a number of studies. Two studies found that cytisine (a product considerable adverse effect profile (dry mouth, dizziness, somnolence,
based on natural ingredients) was effective for smoking cessation and orthostatic hypotension); therefore the use of clonidine remains
(pooled relative risk [RR], 3.98), whereas another trial found that narrow even as a second-line treatment. Clinical trials of nortriptyline
dianicline (a synthetic product similar to varenicline) was not effective found it to be effective, with an odds ratio of 2.1 compared with
(RR, 1.2). Fifteen trials found that varenicline at the standard dose placebo.192 However, the potential lethality in an overdose, as well as
(2 mg/day) increased the chances of quitting more than twofold anticholinergic and cardiac adverse effects similar to those of clonidine,
compared with placebo (RR, 2.27). However, low-dose varenicline limit its use in the general and cancer populations.
(1 mg/day) roughly doubled the chances of quitting (RR, 2.09) and
reduced the number and severity of adverse effects. In the same review, Psychosocial Interventions
varenicline was reported to be better than bupropion in three com-
parison trials (pooled RR, 1.52), but two other trials did not show a The Centers for Disease Control and Prevention emphasized the need
clear benefit of varenicline over the nicotine patch.181 to screen all patients at all visits in all health care settings for tobacco
use, and to advise smokers to quit, and refer them to or offer them
Combinations of First-Line Medications treatment.193 Gritz and colleagues194 postulated that the cancer popula-
Combining medications is becoming a trend, in particular for such tion is at the heart of the concern. Smoking rates in certain patients
traditionally hard-to-treat diseases as tuberculosis, gastric ulcers, with cancer are higher than average, and mounting evidence links
human immunodeficiency virus, and others. Tobacco dependence is smoking to outcomes related to cancer treatment success, survival,
not an exception, as combination therapy is reported to be better and quality of life.195–200 In patients receiving advanced treatments
than monotherapy for smoking cessation (Table 24.3).182 It has been such as stem cell transplantation, those who never smoked or recent
found that all smokers (except light smokers and those who live with quitters were reported to have almost double survivorship rates and
a smoker) may respond better to combination pharmacotherapy than about half as many hospitalization days in comparison with smokers.201
to monotherapy.183 Before varenicline was available, the options for Therefore particular attention must be given to the oncology
combination were to add NRTs to bupropion or to use a patch with setting.22,202–204 In general, patients with cancer were found to be more
episodic NRTs (e.g., lozenges and gums).184 Studies designed to compare motivated to quit smoking than were persons in the general popula-
these smoking cessation medications alone or in combination reported tion.22,205 This phenomenon can be seen as an advantage and an
that certain combinations were superior: specifically, bupropion plus opportunity for care providers to intervene and promote smoking
lozenges and the patch plus lozenges resulted in higher cessation cessation while the tobacco user is motivated by the immediacy of
rates than did monotherapy bupropion, patch, or lozenges.144,184,185 the consequences of continued use on his or her disease treatment
Now that varenicline is available, combining this medication with and recovery.
Nicotine Dependence: Current Treatments and Future Directions  •  CHAPTER 24 407

Table 24.3 Meta-analysis (in CPG-TTUD 2008)a: Effectiveness and Abstinence Rates for Various
Medications and Medication Combinations Compared With Placebo 6 Months After Quitting
(N = 83 Studies)
No. of Estimated Odds Ratio Estimated Abstinence
Medication Arms (95% CI) Rate (95% CI)
Placebo 80 1.0 13.8
MONOTHERAPIES
Varenicline (2 mg/day) 5 3.1 (2.5–3.8) 33.2 (28.9–37.8)
Nicotine nasal spray 4 2.3 (1.7–3.0) 26.7 (21.5–32.7)
High-dose nicotine patch (>25 mg) (these patches included both 4 2.3 (1.7–3.0) 26.5 (21.3–32.5)
standard and long-term duration)
Long-term nicotine gum (>14 wk) 6 2.2 (1.5–3.2) 26.1 (19.7–33.6)
Varenicline (1 mg/day) 3 2.1 (1.5–3.0) 25.4 (19.6–32.2)
Nicotine inhaler 6 2.1 (1.5–2.9) 24.8 (19.1–31.6)
Clonidine 3 2.1 (1.2–3.7) 25.0 (15.7–37.3)
Bupropion sustained release (SR) 26 2.0 (1.8–2.2) 24.2 (22.2–26.4)
Nicotine patch (6–14 wk) 32 1.9 (1.7–2.2) 23.4 (21.3–25.8)
Long-term nicotine patch (>14 wk) 10 1.9 (1.7–2.3) 23.7 (21.0–26.6)
Nortriptyline 5 1.8 (1.3–2.6) 22.5 (16.8–29.4)
Nicotine gum (6–14 wk) 15 1.5 (1.2–1.7) 19.0 (16.5–21.9)
COMBINATION THERAPIES
Patch (long-term; >14 wk) + ad lib NRT (gum or spray) 3 3.6 (2.5–5.2) 36.5 (28.6–45.3)
Patch + bupropion SR 3 2.5 (1.9–3.4) 28.9 (23.5–35.1)
Patch + nortriptyline 2 2.3 (1.3–4.2) 27.3 (17.2–40.4)
Patch + inhaler 2 2.2 (1.3–3.6) 25.8 (17.4–36.5)
Patch + second generation antidepressants (e.g., paroxetine, venlafaxine) 3 2.0 (1.2–3.4) 24.3 (16.1–35.0)
MEDICATIONS NOT SHOWN TO BE EFFECTIVE
Selective serotonin reuptake inhibitors 3 1.0 (0.7–1.4) 13.7 (10.2–18.0)
Naltrexone 2 0.5 (0.2–1.2) 7.3 (3.1–16.2)

a
The articles in this meta-analysis can be found at ww.surgeongeneral.gov/tobacco/gdlnrefs.htm.
CCP-TTUD, Clinical Practice Guideline for Treating Tobacco Use and Dependence; CI, confidence interval; NRT, nicotine replacement therapy.
From Fiore MC, Jaen CR, Baker TB, et al. Treating Tobacco Use and Dependence: 2008 Update. U.S. Public Health Service Clinical Practice Guideline. 2008.

Certain issues are particular to the application of smoking cessation Quitlines


programs among patients with cancer. On the one hand, patients
with cancer may be susceptible to guilt and blame attributions206; on Quitlines provide physicians with an alternative easy-access treatment,
the other hand, the clinician must be knowledgeable and alert to which can be especially helpful for physicians who are not affiliated
comorbid disorders, oncologic treatment adverse effects, and medical with hospitals or work in hospitals that do not have a tobacco treatment
contraindications for medications when treating patients undergoing program (TTP). Quitlines are a good option for patients who are
treatment for cancer. The CPG-TTUD, in 2000 and again in 2008, unable or unwilling to travel to have an in-person appointment. In
recommended that all physicians and health care providers advise addition, quitlines provide telephone-based, individual counseling for
their patients to quit.207 This guideline included the five necessary smoking cessation and are recommended as effective in the CPG-
steps in tobacco-dependence treatment, also known as the five As: TTUD6 and a Cochrane review.211 Quitlines have been referred to as
ask, advise, assess, assist, and arrange. Fiore and colleagues208 suggested an underrecognized success story212 and a tool that has been effective
adding smoking to the vital signs checkup of every patient to ensure for both young adult smokers and older smokers213 and for African
proper documentation, systematic screening, and ongoing monitoring Americans as well as white smokers.214
of tobacco use status. Furthermore, electronic health records can be In the United States, each state offers telephone-based quit-
used to increase the identification of smokers and facilitate referrals line counseling for smoking cessation. Many quitlines also offer
to treatment.209,210 Interesting to note, research has shown that even Internet-based assistance. Tobacco users can access telephone quitline
short interventions (3 minutes or less) would have an effect on counseling in all 50 states, the District of Columbia, Guam, and
abstinence. However, as the number of sessions increases, in particular Puerto Rico by calling the national quitline portal at 1-800-QUIT-
for face-to-face interventions, abstinence rates rise as well.6 Therefore NOW. Callers are routed to their appropriate state quitline based
the CPG-TTUD recommends at least a minimal intervention of brief on the area code of the landline they are calling from or the loca-
support in addition to medications, in particular if the patient does tion of the cell phone tower that routes the call. For more general
not have access to or will not ultimately receive a full tobacco treatment information about quitlines, see http://www.naquitline.org/?page
intervention. =whatisquitline.
408 Part I: Science and Clinical Oncology

Most state quitlines also have health care provider referral programs, Other studies examined nurse-delivered interventions in smoking
wherein patients can be referred to quitline services by their physician cessation across various cancer diagnoses. A single-session intervention
or other health care provider via fax or e-mail, or the referral can be had lower cessation rates and multisession interventions had the highest
integrated into an electronic health record.215 Quitlines then make rates.226,227 Although the samples differed greatly in size and composi-
proactive outbound calls to referred tobacco users to enroll them in tion, these authors believed that these results were promising and that
quitline services and provide feedback to the referring clinic. Research nurse-based interventions should be explored further. Moreover, a
indicates that patients referred to a quitline by their health care provider retrospective study by Garces and colleagues228 emphasized the need
are more likely to quit smoking successfully than are those who for early interventions. The analysis showed higher abstinence rates
self-refer.216 among patients with cancer who were treated within 3 months of
Once enrolled in quitline services, tobacco users also have access to diagnosis versus those treated more than 3 months after diagnosis.
free NRTs, when a state program’s budget allows that expenditure, in Finally, a trial assessed peer-based counseling versus self-help interven-
70% of states and in a few cases prescription medications (bupropion tion in young adults who had survived childhood cancer.229 Higher
or varenicline). Information about each state quitline, including specific rates of smoking cessation were found in the counseling group at
services offered, hours of operations, and contact information for fax 12-month follow-up. Another study comparing a mixed group of
and e-referral programs, can be found on the North American Quitline patients with cancer who received motivational interviewing–based
Association website at http://map.naquitline.org. The National Cancer intervention versus those who received usual care consisting of brief
Institute also provides cessation assistance that includes online and smoking cessation advice did not find the results to be significantly
telephone-based support. Telephone counseling and texted support are different.230
available at https://www.smokefree.gov or by calling 1-877-44U-QUIT. In general, the combination of psychosocial therapies and medication
Pharmacotherapy is encouraged but not provided with those innovative doubles the odds that patients will quit smoking, despite the multiple
programs. types and levels of intensity for the psychosocial therapies (see Table
24.3). A review of controlled trials among patients with cancer suggested
Self-Help Materials that the combination of medication and therapy is needed to make
a significant difference in smoking cessation in the oncology setting.127
Self-help smoking cessation interventions are significantly better than Furthermore, the American Society of Addiction Medicine has described
no intervention at all; they are reported to produce quit rates in the several possible levels of care and different types of treatment programs
range of 4% to 11%.217 An important new vehicle is the Internet, for all addictions, with those levels of care and types of programs
through which self-help smoking cessation interventions can be delivered based on whether concurrent psychiatric disorders are diagnosed and
effectively.218,219 Although few studies have evaluated the efficacy of treated.231
Internet-based cessation approaches, in one study a tailored Internet-
based cessation intervention resulted in a quit rate of 24% compared Tobacco Treatment Program at MD Anderson
with 5% in the control condition at 3-month follow-up.220,221 This
finding is consistent with other preliminary work showing the efficacy The TTP at MD Anderson Cancer Center was conceived and built
of Internet-based cessation interventions.222 More controlled trials of as a center of excellence program with a multidisciplinary team. The
Internet-based approaches are needed for this medium to be recom- TTP goal is to tailor tobacco dependence treatment to each person,
mended for the delivery of tobacco and ND treatment. because each individual is thought to be unique in terms of facing
addiction. The treatment is adapted to each patient’s own history,
current level of ND, intensity of distress due to the cancer diagnosis,
CESSATION TREATMENTS FOR PATIENTS and any concurrent psychiatric disorder. The counseling staff includes
WITH CANCER: AVAILABILITY both masters- and doctoral-level counselors who address the psychosocial
AND CHALLENGES aspect of the addiction, and the patient further benefits from the
involvement of a physician assistant, a nurse, and an addiction psy-
Cancer treatments and their contraindications impose certain limitations chiatrist, who focus on the medical and psychiatric aspect of the
for clinicians who treat smokers who wish to quit. Particular attention patient’s treatment. The TTP’s mission is to evaluate and treat all
should be given to the type of pharmacologic or adjuvant treatment patients with cancer and employees at MD Anderson Cancer Center,
given to smokers and any interaction with the current cancer type as well as their household members, when their smoking is an obstacle
and cancer treatment type, in addition to psychiatric comorbidity to the success of the patient with cancer in quitting. Every patient
(e.g., alcohol dependence and depression).197,223 More research is needed who indicates tobacco use in the past 12 months is automatically
to identify proper and tailored smoking cessation treatments for the referred to the TTP via the electronic health record. Referred patients
cancer population.128 are proactively contacted by TTP staff, who provide a motivational
Gritz and colleagues194 have identified several intervention studies interaction and program description. Patients may elect to receive
that addressed smoking cessation among patients with cancer. Two self-help materials or assistance from a counselor by phone or in
studies have assessed the role and efficacy of physician-patient interven- person. Those who choose in-person assistance also receive pharma-
tions.224,225 In the first study, the authors compared patients with cotherapy and psychologic and psychiatric treatment. The TTP has
cancer who were randomly assigned to full physician advice, tailored been expanded to all three Anderson regional care centers currently
booklets, and booster advice sessions versus patients who were offered in operation. Videoconferencing is used to optimize centralized expertise
minimal advice sessions, as a control group. In the second study, the across a diverse infrastructure and to expand treatment as needed
authors compared intensive cognitive behavioral interventions and (with a fast Intranet connection and a new generation of video cameras
patient education and advice. Both studies found no significant dif- and software). In addition, the TTP is currently piloting delivery of
ference between the two conditions; however, in the first study up to follow-up sessions by Web camera to the patient’s home, using special
70% of the participants remained abstinent at 1-year follow-up, and software and a high security standard to safeguard patients’ confidential-
in the latter study about 40% remained abstinent at 3-month follow-up. ity. This approach is in lieu of the usual telephone counseling that
Even though no single treatment was found to be superior in helping currently takes place for follow-up when patients are outside the
patients with cancer to quit smoking, assistance and advice in general metropolitan area or are simply not able to come to the medical center
generated good abstinence rates. The findings thus support incorpora- regularly.
tion of some level of advice that could be tailored to the patient and Currently, over 5000 patients are referred to the TTP each year.
type of cancer diagnosis. Over 80% receive a motivational interaction, and approximately 20%
Nicotine Dependence: Current Treatments and Future Directions  •  CHAPTER 24 409

to 25% choose assistance from a counselor. Since the program’s opening Another area for future study involves the impact that increased
in 2006, TTP counselors have served over 5000 unique patients, with use of electronic nicotine delivery systems (ENDS) such as electronic
36% to 47% reporting abstinence at 9-month follow-up.232 cigarettes (e-cigarettes) have on nicotine dependence and smoking
cessation. ENDS were introduced in the United States in 2007. Public
CHALLENGES AND FUTURE DIRECTIONS awareness among adults had grown to over 75% by 2012, and 88%
of current smokers were aware of them.234 The technology is rapidly
Many challenges remain to be overcome in treating tobacco dependence, changing, and many ENDS are available.235 Most e-cigarette users
in particular in the oncology setting. For example, geographic distance also consume combustible tobacco products.234 Although first-generation
has often been a major obstacle for highly specialized and high-quality e-cigarettes were ineffective at delivering nicotine levels comparable
interventions. However, with advances in videoconferencing and to those obtained from smoking cigarettes,236 e-cigarette technology
telemedicine and their availability in remote locations, this problem is becoming more sophisticated.237 In 2016, the US Surgeon General
can be solved.233 issued his first report on e-cigarettes, which indicates that the rate of
As mentioned in the genetics section, a major challenge and yet e-cigarette use is increasing among youth and young adults and that
an opportunity to advance the current pharmacologic treatments for this population is at risk of transitioning from the use of e-cigarettes
smoking cessation is to match a person’s genetic profile7 both to a to combustible tobacco, although 80% appear to be using nonnicotine
successful pharmacologic intervention and to a lower side effect profile e-cigarettes.238 Although the US Surgeon General does not recommend
within clinical practice and clinical trial guidelines. the use of e-cigarettes for smoking cessation, public health authorities
Single-medication trials have led to around 40% cessation rates in the United Kingdom have stated that the use of e-cigarettes is
by the end of treatment. However, another 40% of participants have preferable to combustible tobacco because of reduced exposure to
a partial response (they reduce their smoking but do not achieve total carcinogens, especially in patients who would otherwise not quit
abstinence). Therefore combining medications is being suggested as smoking.239 The use of e-cigarettes among cancer patients has not
a strategy for all hard-core smokers.183 This approach can be imple- been systemically studied, except in one retrospective study that did
mented by augmentation (i.e., adding another medication or agent not find them to be helpful for cancer patients to quit smoking.240
to the one already in place for partial responders), which is a natural Thus more work is needed to assess the current prevalence of e-cigarette
and logical approach for a patient who is engaged in treatment and use among cancer patients and any impact that e-cigarettes may have
is motivated to quit. Another strategy is prescribing a combination on smoking cessation in this population.
of medications to all or certain smokers from the beginning of cessation In summary, making tobacco use and dependence a major focus
efforts. However, further studies are needed in this area to determine of patient-centered behavioral change during cancer treatment and
whether one strategy is superior. throughout survivorship is a key issue for oncology providers and
The traditional approach and the vast majority of pharmacotherapy cancer care health systems. The field of tobacco dependence treat-
clinical trials ask participants to choose a quit date within 1 to 2 ment is evolving rapidly, with significant increases in cessation rates
weeks of starting medications. However, that practice was born out and the promise of more powerful pharmacotherapy combined
of the practical need to test a hypothesis in a study within a short with greater reach and intensity of treatment. The goal will be to
8- to 12-week period. Allowing patients to choose their own interval integrate state-of-the-art and emerging treatments with ever-advancing
for a quit date or not to set a quit date but rather to reduce gradually oncology care.
and then quit has been suggested in some studies167; however, these
options have not been systematically tested against the strategy of
setting a fixed quit date. Therefore more work is needed to clarify The complete reference list is available online at
this important area. ExpertConsult.com.

KEY REFERENCES
1. World Health Organization. The Top 10 Causes of 14. US Department of Health and Human Services. 32. Pidoplichko VL, DeBiasi M, Williams JT, Dani JA.
Death. www.who.int/mediacentre/factsheets/fs310/ The Health Consequences of Smoking—50 Years of Nicotine activates and desensitizes midbrain dopa-
en/index.html. Updated June 28, 2012. Progress: A Report of the Surgeon General. Atlanta, mine neurons. Nature. 1997;390(6658):401–404.
2. IARC. IARC Monographs on the Evaluation GA; 2014. Surgeon General’s Report. 36. Benowitz NL. Neurobiology of nicotine addiction:
of Carcinogenic Risks to Humans. http:// 17. US Centers for Disease Control and Prevention. implications for smoking cessation treatment. Am
monographs.iarc.fr/ENG/Monographs/vol97/ The CDC Health Disparities and Inequalities J Med. 2008;121(4 suppl 1):S3–S10.
index.php. Updated July 19, 2012. Report—United States. 2011. http://www.cdc.gov/ 37. Volkow ND, Fowler J, Wang G-J. Role of dopamine
4. Pontieri FE, Tanda G, Orzi F, Di CG. Effects minorityhealth/reports/CHDIR11/FactSheets/ in drug reinforcement and addiction in humans:
of nicotine on the nucleus accumbens and Smoking.pdf. Updated June 29, 2012. Results from imaging studies. Behav Pharmacol.
similarity to those of addictive drugs. Nature. 24. Gritz ER, Schacherer C, Koehly L, Nielsen IR, 2002;13:355–366.
1996;382(6588):255–257. Abemayor E. Smoking withdrawal and relapse 38. Robinson TE, Berridge KC. Incentive-sensitization
6. Fiore MC, Jaen CR, Baker TB, et al. Treating in head and neck cancer patients. Head Neck. and addiction. Addiction. 2001;96(1):103–114.
Tobacco Use and Dependence: 2008 Update, 1999;21(5). 48. Tobacco and Genetics Consortium. Genome-wide
Clinical Practice Guideline. PM:18807274. 25. Cooley ME, Sarna L, Kotlerman J, et al. Smoking meta-analyses identify multiple loci associated with
7. King DP, Paciga S, Pickering E, et al. Smoking cessation is challenging even for patients recovering smoking behavior. Nat Genet. 2010;42(5):441–447.
cessation pharmacogenetics: analysis of varenicline from lung cancer surgery with curative intent. Lung 52. Amos CI, Wu X, Broderick P, et al. Genome-wide
and bupropion in placebo-controlled clinical Cancer. 2009;66(2):218–225. association scan of tag SNPs identifies a susceptibil-
trials. Neuropsychopharmacology. 2012;37(3):641– 26. McBride CM, Emmons KM, Lipkus IM. Under- ity locus for lung cancer at 15q25.1. Nat Genet.
650. standing the potential of teachable moments: 2008;40(5):616–622.
8. McBride CM, Ostroff JS. Teachable moments the case of smoking cessation. Health Educ Res. 69. Schnoll RA, Leone FT. Biomarkers to optimize
for promoting smoking cessation: the context 2003;18(2):156–170. the treatment of nicotine dependence. Biomarkers.
of cancer care and survivorship. Cancer Control. 27. Moyer VA. Screening for lung cancer: U.S. Preventive 2011;5(6):745–761.
2003;10(4):325–333. Services Task Force recommendation statement. Ann 100. Lerman C, Schnoll RA, Hawk LW Jr, et al. Use
13. Jamal A, King BA, Neff LJ, Whitmill J, Babb Intern Med. 2014;160(5):330–338. of the nicotine metabolite ratio as a genetically
SD, Graffunder CM. Current Cigarette Smoking 31. Nisell M, Marcus M, Nomikos GG, Svensson TH. informed biomarker of response to nicotine patch
Among Adults—United States, 2005–2015. Differential effects of acute and chronic nicotine on or varenicline for smoking cessation: a randomised,
MMWR Morb Mortal Wkly Rep. 2016;65(44):1205– dopamine output in the core and shell of the rat double-blind placebo-controlled trial. Lancet Respir
1211. nucleus accumbens. J Neural Transm. 1997;104:1–10. Med. 2015;3(2):131–138.
410 Part I: Science and Clinical Oncology

101. Lasser K, Boyd JW, Woolhandler S, Himmelstein MD 2012. http://www.cancer.gov/cancertopics/pdq/ 184. Piper ME, Smith SS, Schlam TR, et al. A random-
DU, McCormick D, Bor DH. Smoking and mental supportivecare/smokingcessation/HealthProfessional. ized placebo-controlled clinical trial of 5 smoking
illness: A population-based prevalence study. JAMA. Updated July 2, 2012. cessation pharmacotherapies. Arch Gen Psychiatry.
2000;284(20):2606–2610. 143. Silagy C, Lancaster T, Stead L, Mant D, 2009;66(11):1253–1262.
102. Breslau N, Johnson EO, Hiripi E, Kessler R. Nicotine Fowler G. Nicotine replacement therapy for 185. Smith SS, McCarthy DE, Japuntich SJ, et al.
dependence in the United States: prevalence, trends, smoking cessation. Cochrane Database Syst Rev. Comparative effectiveness of 5 smoking cessation
and smoking persistence. Arch Gen Psychiatry. 2004;(3):CD000146-CD000146. pharmacotherapies in primary care clinics. Arch
2001;58(9):810–816. 147. Karam-Hage M, Cinciripini PM. Pharmaco- Intern Med. 2009;169(22):2148–2155.
103. Covey LS, Hughes DC, Glassman AH, Blazer therapy for tobacco cessation: nicotine agonists, 194. Gritz ER, Lam CY, Vidrine DJ, Fingeret MC.
DG, George LK. Ever-smoking, quitting, and antagonists, and partial agonists. Curr Oncol Rep. Tobacco dependence and its treatment. In: DeVita V,
psychiatric disorders: Evidence from the Durham, 2007;9(6):509–516. Lawrence T, Rosenberg S, eds. Cancer: Principles and
North Carolina, epidemiologic catchment area. Tob 163. Gonzales D, Rennard SI, Nides M, et al. Var- Practice of Oncology. 9th ed. Philadelphia: Lippincott
Control. 1994;3:222–227. enicline, an alpha4beta2 nicotinic acetylcholine Williams & Wilkins; 2012:529–542.
104. Glassman AH, Helzer JE, Covey LS, et al. Smoking, receptor partial agonist, vs sustained-release 196. Gritz ER, Dresler C, Sarna L. Smoking, the
smoking cessation, and major depression. JAMA. bupropion and placebo for smoking cessation: a missing drug interaction in clinical trials: ignoring
1990;264(12):1546–1549. randomized controlled trial. JAMA. 2006;296(1):47– the obvious. Cancer Epidemiol Biomarkers Prev.
105. Williams JM, Ziedonis D. Addressing tobacco among 55. 2005;14(10):2287–2293.
individuals with a mental illness or an addiction. 164. Gonzales DH, Rennard SI, Billing CB, Reeves K, 197. Gritz ER, Fingeret MC, Vidrine DJ, Lazev AB,
Addict Behav. 2004;29(6):1067–1083. Watsky E, Gong J. A pooled analysis of varenicline, Mehta NV, Reece GP. Successes and failures of the
111. Grant BF, Hasin DS, Chou SP, Stinson FS, Dawson an alpha 4 beta 2 nicotinic receptor partial agonist teachable moment: Smoking cessation in cancer
DA. Nicotine dependence and psychiatric disorders vs bupropion, and placebo for smoking cessation. patients. Cancer. 2006;106(1):17–27.
in the United States: results from the national epide- February; Orlando, FL. 198. Jensen K, Jensen AB, Grau C. Smoking has a nega-
miologic survey on alcohol and related conditions. 165. Jorenby DE, Hays JT, Rigotti NA, et al. Efficacy of tive impact upon health related quality of life after
Arch Gen Psychiatry. 2004;61(11):1107–1115. varenicline, an alpha4beta2 nicotinic acetylcholine treatment for head and neck cancer. Oral Oncol.
115. Johnson RA, Hoffmann JP. Adolescent cigarette receptor partial agonist, vs placebo or sustained- 2007;43(2):187–192.
smoking in U.S. racial/ethnic subgroups: findings release bupropion for smoking cessation: a 206. Gritz ER, Fingeret MC, Vidrine DJ. Tobacco control
from the National Education Longitudinal Study. randomized controlled trial. JAMA. 2006;296(1): in the oncology setting. 2007. Cancer Prevention:
J Health Soc Behav. 2000;41(4):392–407. 56–63. An ASCO Curriculum.
116. Kendler KS, Neale MC, MacLean CJ, Heath 170. Anthenelli RM, Benowitz NL, West R, et al. 212. Lichtenstein E, Zhu SH, Tedeschi GJ. Smoking
AC, Eaves LJ, Kessler RC. Smoking and major Neuropsychiatric safety and efficacy of varenicline, cessation quitlines: an underrecognized intervention
depression: a casual analysis. Arch Gen Psychiatry. bupropion, and nicotine patch in smokers with and success story. Am Psychol. 2010;65(4):252–261.
1993;50(1):36–43. without psychiatric disorders (EAGLES): a double- 222. Walters ST, Wright JA, Shegog R. A review of com-
128. US National Cancer Institute. Smoking Cessation blind, randomised, placebo-controlled clinical trial. puter and Internet-based interventions for smoking
and Continued Risk in Cancer Patients. Bethesda, Lancet. 2016;387(10037):2507–2520. behavior. Addict Behav. 2006;31(2):264–277.
Nicotine Dependence: Current Treatments and Future Directions  •  CHAPTER 24 410.e1
410.e1

REFERENCES
1. World Health Organization. The Top 10 Causes of 20. Schnoll RA, Zhang B, Rue M, et al. Brief 39. Curtin JJ, McCarthy DE, Piper ME, Baker TB.
Death. www.who.int/mediacentre/factsheets/fs310/ physician-initiated quit-smoking strategies for Implicit and explicit drug motivational processes:
en/index.html. Updated June 28, 2012. clinical oncology settings: a trial coordinated by A model of boundary conditions. In: Reinout R,
2. IARC. IARC Monographs on the Evaluation of the Eastern Cooperative Oncology Group. J Clin Stacy A, eds. Handbook of Implicit Cognition and
Carcinogenic Risks to Humans. http://monographs. Oncol. 2003;21(2):355–365. Addiction. Thousand Oaks, CA.: SAGE Publications;
iarc.fr/ENG/Monographs/vol97/index.php. Updated 21. Gritz ER, Nisenbaum R, Elashoff R, Holmes EC. 2006:233–250.
July 19, 2012. Smoking behavior following diagnosis in patients 40. Baker TB, Piper ME, McCarthy DE, Majeskie MR,
3. Jacobs EJ, Newton CC, Carter BD, et al. What with stage I non-small cell lung cancer. Cancer Causes Fiore MC. Addiction motivation reformulated: An
proportion of cancer deaths in the contemporary Control. 1991;2:105–112. affective processing model of negative reinforcement.
United States is attributable to cigarette smoking? 22. Ostroff JS, Jacobsen PB, Moadel AB, et al. Prevalence Psychol Rev. 2004;111(1):33–51.
Ann Epidemiol. 2015;25(3):179–182. and predictors of continued tobacco use after treat- 41. Gotti C, Zoli M, Clementi F. Brain nicotinic acetyl-
4. Pontieri FE, Tanda G, Orzi F, Di CG. Effects ment of patients with head and neck cancer. Cancer. choline receptors: native subtypes and their relevance.
of nicotine on the nucleus accumbens and 1995;75(2):569–576. Trends Pharmacol Sci. 2006;27(9):482–491.
similarity to those of addictive drugs. Nature. 23. Ostroff J, Garland J, Moadel A, et al. Cigarette 42. De Biasi M, Dani JA. Reward, addiction, withdrawal
1996;382(6588):255–257. smoking patterns in patients after treatment of to nicotine. Annu Rev Neurosci. 2011;34:105–
5. Volkow ND, Fowler JS, Wang GJ. The addicted bladder cancer. J Cancer Educ. 2000;15(2):86–90. 130.
human brain viewed in the light of imaging studies: 24. Gritz ER, Schacherer C, Koehly L, Nielsen IR, 43. Conlon MS, Bewick MA. Single nucleotide poly-
Brain circuits and treatment strategies. Neurophar- Abemayor E. Smoking withdrawal and relapse in morphisms in CHRNA5 rs16969968, CHRNA3
macology. 2004;47(suppl 1):3–13. head and neck cancer patients. Head Neck. 1999; rs578776, and LOC123688 rs8034191 are
6. Fiore MC, Jaen CR, Baker TB, et al Treating 21(5). associated with heaviness of smoking in women
tobacco use and dependence: 2008 Update U.S. 25. Cooley ME, Sarna L, Kotlerman J, et al. Smoking in northeastern Ontario, Canada. Nicotine Tob Res.
Public Health Service Clinical Practice Guideline cessation is challenging even for patients recovering 2011;13(6):498–503.
executive summary. Respir Care. 2008;53(9):1217– from lung cancer surgery with curative intent. Lung 44. Saccone NL, Culverhouse RC, Schwantes-An TH,
1222. Cancer. 2009;66(2):218–225. et al. Multiple independent loci at chromosome
7. King DP, Paciga S, Pickering E, et al. Smoking 26. McBride CM, Emmons KM, Lipkus IM. Under- 15q25.1 affect smoking quantity: a meta-analysis
cessation pharmacogenetics: analysis of varenicline standing the potential of teachable moments: and comparison with lung cancer and COPD. PLoS
and bupropion in placebo-controlled clinical trials. the case of smoking cessation. Health Educ Res. Genet. 2010;6(8):1–16.
Neuropsychopharmacology. 2012;37(3):641–650. 2003;18(2):156–170. 45. Bierut LJ, Stitzel JA, Wang JC, et al. Variants in
8. McBride CM, Ostroff JS. Teachable moments 27. Moyer VA. Screening for lung cancer: U.S. Preventive nicotinic receptors and risk for nicotine dependence.
for promoting smoking cessation: the context Services Task Force recommendation statement. Ann Am J Psychiatry. 2008;165(9):1163–1171.
of cancer care and survivorship. Cancer Control. Intern Med. 2014;160(5):330–338. 46. Lips EH, Gaborieau V, McKay JD, et al. Association
2003;10(4):325–333. 28. Slatore CG, Baumann C, Pappas M, Humphrey between a 15q25 gene variant, smoking quantity and
9. Carson T, Bonk M. James Buchanan Duke. Gale LL. Smoking behaviors among patients receiving tobacco-related cancers among 17 000 individuals.
Encyclopedia of U.S. Economic History. Detroit: Gale computed tomography for lung cancer screen- Int J Epidemiol. 2010;39(2):563–577.
Group; 1999. ing. Systematic review in support of the U.S. 47. Liu JZ, Tozzi F, Waterworth DM, et al. Meta-analysis
10. James R. Cigarette Advertising. http://www.time. preventive services task force. Ann Am Thorac Soc. and imputation refines the association of 15q25 with
com/time/magazine/article/0,9171,1905530,00. 2014;11(4):619–627. smoking quantity. Nat Genet. 2010;42(5):436–440.
html. Updated June 29, 2012. 29. Clark MA, Gorelick JJ, Sicks JD, et al. The rela- 48. Tobacco and Genetics Consortium. Genome-wide
11. US Department of Health and Human Services. tions between false positive and negative screens meta-analyses identify multiple loci associated
The health consequences of smoking: a report of and smoking cessation and relapse in the National with smoking behavior. Nature Genet. 2010;
the surgeon general. http://profiles.nlm.nih.gov/ Lung Screening Trial: implications for public health. 42(5):441–447.
NN/B/B/M/Q/. Nicotine Tob Res. 2016;18(1):17–24. 49. Olfson E, Saccone NL, Johnson EO, et al. Rare, low
12. Newport F, Moore DW, Saad L. Long-term Gallup 30. Fucito LM, Czabafy S, Hendricks PS, Kotsen C, frequency and common coding variants in CHRNA5
poll trends: a portrait of American public opinion Richardson D, Toll BA. Pairing smoking-cessation and their contribution to nicotine dependence in
through the century. http://www.gallup.com/ services with lung cancer screening: a clinical European and African Americans. Mol Psychiatry.
poll/3400/longterm-gallup-poll-trends-portrait- guideline from the Association for the Treatment 2016;21(5):601–607.
american-public-opinion.aspx. Updated June 29, of Tobacco Use and Dependence and the Society 50. Tyndale RF, Zhu AZ, George TP, et al. Lack of
2012. for Research on Nicotine and Tobacco. Cancer. associations of CHRNA5-A3-B4 Genetic variants
13. Jamal A, King BA, Neff LJ, Whitmill J, Babb SD, 2016;122(8):1150–1159. with smoking cessation treatment outcomes in
Graffunder CM. Current Cigarette Smoking Among 31. Nisell M, Marcus M, Nomikos GG, Svensson TH. Caucasian smokers despite associations with baseline
Adults—United States, 2005-2015. MMWR Morb Differential effects of acute and chronic nicotine on smoking. PLoS ONE. 2015;10(5):e0128109.
Mortal Wkly Rep. 2016;65(44):1205–1211. dopamine output in the core and shell of the rat 51. Chen L-S, Horton A, Bierut L. Pathways to precision
14. US Department of Health and Human Services. nucleus accumbens. J Neural Transm. 1997;104:1–10. medicine in smoking cessation treatments. Neurosci
The Health Consequences of Smoking—50 Years of 32. Pidoplichko VL, DeBiasi M, Williams JT, Dani JA. Lett. 2016.
Progress: A Report of the Surgeon General. Atlanta, Nicotine activates and desensitizes midbrain dopa- 52. Amos CI, Wu X, Broderick P, et al. Genome-wide
GA; 2014. Surgeon General’s Report. mine neurons. Nature. 1997;390(6658):401–404. association scan of tag SNPs identifies a susceptibil-
15. US Centers for Disease Control and Prevention. 33. Carr LA, Basham JK, York BK, Rowell PP. Inhibition ity locus for lung cancer at 15q25.1. Nat Genet.
Tobacco data statistics Pre-1994. http://www.cdc. of uptake of 1-methyl-4-phenylpyridinium ion and 2008;40(5):616–622.
gov/tobacco/data_statistics/sgr/pre_1994/index.htm. dopamine in striatal synaptosomes by tobacco smoke 53. Thorgeirsson TE, Geller F, Sulem P, et al. A
Updated June 29, 2012. components. Eur J Pharmacol. 1992;215:285–287. variant associated with nicotine dependence, lung
16. Martell BN, Garrett BE, Caraballo RS. Disparities in 34. Le Foll B, Forget B, Aubin H-J, Goldberg SR. Block- cancer and peripheral arterial disease. Nature.
Adult Cigarette Smoking—United States, 2002-2005 ing cannabinoid CB1 receptors for the treatment of 2008;452(7187):638–642.
and 2010-2013. MMWR Morb Mortal Wkly Rep. nicotine dependence: insights from pre-clinical and 54. Huang W, Payne TJ, Ma JZ, et al. Significant
2016;65(30):753–758. clinical studies. Addict Biol. 2008;13(2):239–252. association of ANKK1 and detection of a functional
17. US Centers for Disease Control and Prevention. 35. Lowinson J, Ruiz P, Millman R, Langrod J, eds. polymorphism with nicotine dependence in an
The CDC Health Disparities and Inequalities Substance Abuse A Comprehensive Textbook. Maryland: African-American sample. Neuropsychopharmacology.
Report—United States, 2011. http://www.cdc. Williams & Wilkins; 1992. 2009;34(2):319–330.
gov/minorityhealth/reports/CHDIR11/FactSheets/ 36. Benowitz NL. Neurobiology of nicotine addiction: 55. Thorgeirsson TE, Gudbjartsson DF, Surakka I,
Smoking.pdf. Updated June 29, 2012. implications for smoking cessation treatment. Am et al. Sequence variants at CHRNB3-CHRNA6
18. NCI. Cancer Trends Progress Report—2009/2010 J Med. 2008;121(4 suppl 1):S3–S10. and CYP2A6 affect smoking behavior. Nat Genet.
Update. http://progressreport.cancer.gov/. Updated 37. Volkow ND, Fowler J, Wang G-J. Role of dopamine 2010;42(5):448–453.
June 29, 2012. in drug reinforcement and addiction in humans: 56. Gelernter J, Yu Y, Weiss R, et al. Haplotype spanning
19. Bellizzi KM, Rowland JH, Jeffery DD, McNeel T. Results from imaging studies. Behav Pharmacol. TTC12 and ANKK1, flanked by the DRD2 and
Health behaviors of cancer survivors: examining 2002;13:355–366. NCAM1 loci, is strongly associated to nicotine
opportunities for cancer control intervention. 38. Robinson TE, Berridge KC. Incentive-sensitization dependence in two distinct American populations.
J Clin Oncol. 2005;23(34):8884–8893. and addiction. Addiction. 2001;96(1):103–114. Human Mol Genet. 2006;15(24):3498–3507.
410.e2 Part I: Science and Clinical Oncology

57. Ruyck K, de Nackaerts K, Beels L, et al. Genetic 75. Munafò MR, Johnstone EC, Guo B, Murphy MFG, in smoking cessation therapy. Am J Med Genet B
variation in three candidate genes and nicotine Aveyard P. Association of COMT Val108/158Met Neuropsychiatr Genet. 2008;147B(6):712–719.
dependence, withdrawal and smoking cessation in genotype with smoking cessation. Pharmacogenet 94. Sarginson JE, Killen JD, Lazzeroni LC, et al.
hospitalized patients. Pharmacogenomics. 2010;11(8): Genomics. 2008;18(2):121–128. Markers in the 15q24 nicotinic receptor subunit
1053–1063. 76. David SP, Munafo MR, Murphy MF, Proctor M, gene cluster (CHRNA5-A3-B4) predict severity of
58. Voisey J, Swagell CD, Hughes IP, et al. A DRD2 Walton RT, Johnstone EC. Genetic variation in the nicotine addiction and response to smoking cessation
and ANKK1 haplotype is associated with nicotine dopamine D4 receptor (DRD4) gene and smoking therapy. Am J Med Genet. 2011;156(3):275–284.
dependence. Psychiatry Res. 2012. cessation: follow-up of a randomised clinical trial 95. Allenby CE, Boylan KA, Lerman C, Falcone M.
59. Bergen AW, Conti DV, van Den BD, et al. Dopamine of transdermal nicotine patch. Pharmacogenomics J. Precision medicine for tobacco dependence: devel-
genes and nicotine dependence in treatment-seeking 2008;8(2):122–128. opment and validation of the nicotine metabolite
and community smokers. Neuropsychopharmacology. 77. Lerman C, Wileyto EP, Patterson F. The functional ratio. J Neuroimmune Pharmacol. 2016;11(3):
2009;34(10):2252–2264. mu opioid receptor (OPRM1) Asn40Asp variant 471–483.
60. Wang J, Li MD. Common and unique biological predicts short-term response to nicotine replace- 96. Lerman C, Tyndale R, Patterson F, et al. Nicotine
pathways associated with smoking initiation/progres- ment therapy in a clinical trial. Pharmacogenomics metabolite ratio predicts efficacy of transdermal
sion, nicotine dependence, and smoking cessation. J. 2004;4:184–192. nicotine for smoking cessation. Clin Pharmacol
Neuropsychopharmacology. 2010;35(3):702–719. 78. Munafò MR, Elliot KM, Murphy MF, Walton RT, Ther. 2006;79(6):600–608.
61. Ling D, Niu T, Feng Y, Xing H, Xu X. Association Johnstone EC. Association of the mu-opioid receptor 97. Schnoll RA, Patterson F, Wileyto EP, Tyndale
between polymorphism of the dopamine transporter gene with smoking cessation. Pharmacogenomics J. RF, Benowitz N, Lerman C. Nicotine metabolic
gene and early smoking onset: an interaction 2007;7(5):353–361. rate predicts successful smoking cessation with
risk on nicotine dependence. J Human Genet. 79. Ray R, Jepson C, Patterson F, et al. Association of transdermal nicotine: A validation study. Pharmacol
2004;49:35–39. OPRM1 A118G variant with the relative reinforc- Biochem Behav. 2009;92(1):6–11.
62. Segman RH, Kanyas K, Karni O, et al. Why do ing value of nicotine. Psychopharmacology (Berl). 98. Ho MK, Mwenifumbo JC, Al Koudsi N, et al.
young women smoke? IV. Role of genetic variation 2006;188(3):355–363. Association of nicotine metabolite ratio and CYP2A6
in the dopamine transporter and lifetime traumatic 80. David SP, Munafo MR, Murphy MF, Walton RT, genotype with smoking cessation treatment in
experience. Am J Med Genet B Neuropsychiatr Genet. Johnstone EC. The serotonin transporter 5-HTTLPR African-American light smokers. Clin Pharmacol
2007;144B(4):533–540. polymorphism and treatment response to nicotine Ther. 2009;85(6):635–643.
63. Chu SL, Xiao D, Wang C, Jing H. Association patch: follow-up of a randomized controlled trial. 99. Patterson F, Schnoll RA, Wileyto EP, et al. Toward
between 5-hydroxytryptamine transporter gene-linked Nicotine Tob Res. 2007;9(2):225–231. personalized therapy for smoking cessation: a
polymorphic region and smoking behavior in Chinese 81. Gilbert DG, Zuo Y, Rabinovich NE, Riise H, randomized placebo-controlled trial of bupropion.
males. Chin Med J. 2009;122(12):1365–1368. Needham R, Huggenvik JI. Neurotransmission- Clin Pharmacol Ther. 2008;84(3):320–325.
64. Ishii T, Wakabayashi R, Kurosaki H, Gemma related genetic polymorphisms, negative affectiv- 100. Lerman C, Schnoll RA, Hawk LW Jr, et al. Use
A, Kida K. Association of serotonin transporter ity traits, and gender predict tobacco abstinence of the nicotine metabolite ratio as a genetically
gene variation with smoking, chronic obstructive symptoms across 44 days with and without nicotine informed biomarker of response to nicotine patch
pulmonary disease, and its depressive symptoms. patch. J Abnorm Psychol. 2009;118(2):322–334. or varenicline for smoking cessation: a randomised,
J Human Genet. 2010;56(1):41–46. 82. Bergen AW, Javitz HS, Krasnow R, Nishita D, Michel double-blind placebo-controlled trial. Lancet Respir
65. Lerman C, Caporaso NE, Audrain J, Main D, M, Conti DV, et al. Nicotinic acetylcholine receptor Med. 2015;3(2):131–138.
Boyd NR, Shields PG. Interacting effects of the variation and response to smoking cessation thera- 101. Lasser K, Boyd JW, Woolhandler S, Himmelstein
serotonin transporter gene and neuroticism in pies. Pharmacogenetic Genomics. 2013;32(2):94–103. DU, McCormick D, Bor DH. Smoking and mental
smoking practices and nicotine dependence. Mol 83. Lee W, Bergen AW, Swan GE, et al. Gender-stratified illness: a population-based prevalence study. JAMA.
Psychiatry. 2000;5(2):189–192. gene and gene-treatment interactions in smoking 2000;284(20):2606–2610.
66. Bergen AW, Michel M, Nishita D, et al. Drug cessation. Pharmacogenomics J. 2012;12(6):521–532. 102. Breslau N, Johnson EO, Hiripi E, Kessler R. Nicotine
metabolizing enzyme and transporter gene variation, 84. Munafo M, Johnstone EC, Walther D, Uhl GR, dependence in the United States: prevalence, trends,
nicotine metabolism, prospective abstinence, and Murphy MF, Aveyard P. CHRNA3 rs1051730 and smoking persistence. Arch Gen Psychiatry.
cigarette consumption. PLoS ONE. 2015;10(7): genotype and short-term smoking cessation. Nicotine 2001;58(9):810–816.
e0126113. Tob Res. 2011;13(10):982–988. 103. Covey LS, Hughes DC, Glassman AH, Blazer
67. Kortmann GL, Dobler CJ, Bizarro L, Bau CH. Phar- 85. David SP, Brown RA, Papandonatos GD, et al. DG, George LK. Ever-smoking, quitting, and
macogenetics of smoking cessation therapy. Am J Med Pharmacogenetic clinical trial of sustained-release psychiatric disorders: evidence from the Durham,
Genet B Neuropsychiatr Genet. 2010;153B(1):17–28. bupropion for smoking cessation. Nicotine Tob Res. North Carolina, epidemiologic catchment area. Tob
68. Lessov-Schlaggar CN, Pergadia ML, Khroyan TV, 2007;9(8):821–833. Control. 1994;3:222–227.
Swan GE. Genetics of nicotine dependence and 86. David SP, Strong DR, Munafo MR, et al. Bupropion 104. Glassman AH, Helzer JE, Covey LS, et al. Smoking,
pharmacotherapy. Biochem Pharmacol. 2008;75(1): efficacy for smoking cessation is influenced by the smoking cessation, and major depression. JAMA.
178–195. DRD2 Taq1A polymorphism: analysis of pooled 1990;264(12):1546–1549.
69. Schnoll RA, Leone FT. Biomarkers to optimize data from two clinical trials. Nicotine Tob Res. 105. Williams JM, Ziedonis D. Addressing tobacco among
the treatment of nicotine dependence. Biomarkers. 2007;9(12):1251–1257. individuals with a mental illness or an addiction.
2011;5(6):745–761. 87. Swan GE, Valdes AM, Ring HZ, et al. Dopamine Addict Behav. 2004;29(6):1067–1083.
70. Lerman C, Jepson C, Wileyto EP, et al. Role of receptor DRD2 genotype and smoking cessation 106. Barkley RA, Fischer M, Edelbrock CS, Smallish
functional genetic variation in the dopamine D2 outcome following treatment with bupropion SR. L. The adolescent outcome of hyperactive children
receptor (DRD2) in response to bupropion and Pharmacogenomics J. 2005;5:21–29. diagnosed by research criteria: I. An 8-year pro-
nicotine replacement therapy for tobacco depen- 88. Berrettini WH, Wileyto EP, Epstein L, et al. spective follow-up study. J Am Acad Child Adolesc
dence: results of two randomized clinical trials. Catechol-O-methyltransferase (COMT) gene variants Psychiatry. 1990;29(4):546–557.
Neuropsychopharmacology. 2006;31(1):231–242. predict response to bupropion therapy for tobacco 107. Borland BL, Heckman HK. Hyperactive boys and
71. Breitling LP, Twardella D, Hoffmann MM, Witt dependence. Biol Psychiatry. 2007;61(1):111–118. their brothers. A 25-year follow-up study. Arch Gen
SH, Treutlein J, Brenner H. Prospective association 89. Leventhal AM, David SP, Brightman M, et al. Psychiatry. 1976;33(6):669–675.
of dopamine-related polymorphisms with smoking Dopamine D4 receptor gene variation moderates 108. Hartsough CS, Lambert NM. Pattern and progres-
cessation in general care. Pharmacogenomics. the efficacy of bupropion for smoking cessation. sion of drug use among hyperactives and controls:
2010;11(4):527–536. Pharmacogenomics J. 2012;12(1):86–92. a prospective short-term longitudinal study. J Child
72. Johnstone EC, Yudkin PL, Hey K, et al. Genetic 90. Lerman C, Shields PG, Wileyto EP, et al. Effects of Psychol Psychiatry. 1987;28(4):543–553.
variation in dopaminergic pathways and short-term dopamine transporter and receptor polymorphisms 109. Breslau N. Psychiatric comorbidity of smoking and
effectiveness of the nicotine patch. Pharmacogenetics. on smoking cessation in a bupropion clinical trial. nicotine dependence. Behav Genet. 1995;25(2):
2004;14(2):83–90. Health Psychol. 2003;22(5):541–548. 95–101.
73. Yudkin P, Munafo M, Hey K, et al. Effectiveness of 91. Lee AM, Jepson C, Hoffmann E, et al. CYP2B6 gen- 110. Batel P, Pessione F, Maitre C, Rueff B. Relationship
nicotine patches in relation to genotype in women otype alters abstinence rates in a bupropion smoking between alcohol and tobacco dependencies among
versus men: Randomised controlled trial. BMJ. cessation trial. Biol Psychiatry. 2007;62(6):635–641. alcoholics who smoke. Addiction. 1995;90(7):
2004;328(7446):989–990. 92. Conti DV, Lee W, Li D, et al. Nicotinic acetylcholine 977–980.
74. David SP, Johnstone EC, Churchman M, Aveyard receptor beta2 subunit gene implicated in a systems- 111. Grant BF, Hasin DS, Chou SP, Stinson FS, Dawson
P, Murphy MF, Munafo MR. Pharmacogenetics of based candidate gene study of smoking cessation. DA. Nicotine dependence and psychiatric disorders
smoking cessation in general practice: results from Human Mol Genet. 2008;17(18):2834–2848. in the United States: results from the national epide-
the patch II and patch in practice trials. Nicotine 93. Heitjan DF, Guo M, Ray R, Wileyto EP, Epstein LH, miologic survey on alcohol and related conditions.
Tob Res. 2011;13(3):157–167. Lerman C. Identification of pharmacogenetic markers Arch Gen Psychiatry. 2004;61(11):1107–1115.
Nicotine Dependence: Current Treatments and Future Directions  •  CHAPTER 24 410.e3
410.e3

112. Breslau N, Klein DF. Smoking and panic attacks: 132. Anthenelli RM, Heffner JL, Wong E, et al. A 150. Garvey AJ, Kinnunen T, Doherty K, Vokonas PS.
an epidemiologic investigation. Arch Gen Psychiatry. randomized trial evaluating whether topiramate aids Effects of nicotine gum on smokers differing in
1999;56(12):1141–1147. smoking cessation and prevents alcohol relapse in level of dependence. Paper presented at The Society
113. Breslau N, Peterson EL, Schultz LR, Chilcoat HD, recovering alcohol-dependent men. Alcohol Clin Exp for Research on Nicotine and Tobacco; January 1,
Andreski P. Major depression and stages of smoking: Res. 2017;41(1):197–206. 1996; Washington, D.C.
a longitudinal investigation. Arch Gen Psychiatry. 133. Johnson BA, Rosenthal N, Capece JA, et al. 151. Shiffman S, Dresler CM, Hajek P, Gilburt SJ,
1998;55:161–166. Improvement of physical health and quality of life Targett DA, Strahs KR. Efficacy of a nicotine
114. Brown DC. Smoking cessation in pregnancy. Can of alcohol-dependent individuals with topiramate lozenge for smoking cessation. Arch Intern Med.
Fam Physician. 1996;42:102–105. treatment: US multisite randomized controlled trial. 2002;162(11):1267–1276.
115. Johnson RA, Hoffmann JP. Adolescent cigarette Arch Intern Med. 2008;168(11):1188. 152. Warren GW, Rangnekar V, McGarry R, Arnold
smoking in U.S. racial/ethnic subgroups: findings 134. Gadde KM, Allison DB. Cannabinoid-1 receptor S, Valentino J, Kudrimoti M. Pathways of resis-
from the National Education Longitudinal Study. antagonist, rimonabant, for management of obesity tance: potential effects of nicotine on cancer and
J Health Soc Behav. 2000;41(4):392–407. and related risks. Circulation. 2006;114(9):974– treatment response. Int J Radiat Oncol Biol Phys.
116. Kendler KS, Neale MC, MacLean CJ, Heath 984. 2008;72(1):S715.
AC, Eaves LJ, Kessler RC. Smoking and major 135. Tonstad S. Rimonabant: a cannabinoid receptor 153. Warren GW, Randall M, McGarry R, Kudrimoti
depression: a casual analysis. Arch Gen Psychiatry. blocker for the treatment of metabolic and cardio- M, Rangnekar V. Abstract 1408: Nicotine decreases
1993;50(1):36–43. vascular risk factors. Nutr Metab Cardiovasc Dis. the therapeutic efficacy of radiotherapy and chemo-
117. Kollins SH, McClernon FJ, Fuemmeler BF. 2006;16(2):156–162. radiotherapy in vivo in Proceedings: AACR 101st
Association between smoking and attention-deficit/ 136. US Food and Drug Administration. Briefing Annual Meeting 2010, April 17–21, 2010. Cancer
hyperactivity disorder symptoms in a population- Information for FDA Advisory Committee Meeting Res. 2010;70(suppl):8.
based sample of young adults. Arch Gen Psychiatry. on ZIMULTI (Rimonabant). May 10, 2007. http:// 154. Shields PG. Long-term nicotine replacement therapy:
2005;62(10):1142–1147. www.fda.gov/ohrms/dockets/ac/07/briefing/2007- cancer risk in context. Cancer Prev Res (Phila).
118. Martinez E, Tatum KL, Weber DM, et al. Issues 4306b1-01-sponsor-backgrounder.pdf. Updated 2011;4(11):1719–1723.
related to implementing a smoking cessation clini- April 13, 2016. Accessed April 13, 2016. 155. Murray RP, Connett JE, Zapawa LM. Does nico-
cal trial for cancer patients. Cancer Causes Control. 137. European Medicines Agency. Public statement on tine replacement therapy cause cancer? Evidence
2009;20(1):97–104. Acomplia (rimonabant): withdrawal of the marketing from the Lung Health Study. Nicotine Tob Res.
119. American Psychiatric Association. Diagnostic and authorisation in the European Union. http://www. 2009;11(9):1076–1082.
Statistical Manual of Mental Disorders. 5th ed. ema.europa.eu/docs/en_GB/document_library/ 156. Murray RP, Bailey WC, Daniels K, et al. Safety of
Arlington, VA: American Psychiatric Association; Public_statement/2009/11/WC500012189.pdf. nicotine polacrilex gum used by 3,094 participants in
2013. Updated April 20, 2016. the Lung Health Study. Chest. 1996;109(2):438–445.
120. American Psychiatric Association. Diagnostic and 138. Raupach T, Hoogsteder PHJ, Onno CP. Nicotine 157. Ascher JA, Cole JO, Colin JN, et al. Bupropion: a
Statistical Manual of Mental Disorders. 4th revised. vaccines to assist with smoking cessation: current review of its mechanism of antidepressant activity.
Washington, D.C.: American Psychiatric Association; status of research. Drugs. 2012;72(4):e1–e16. J Clin Psychiatry. 1995;56:395–401.
2000. 139. Moore D, Aveyard P, Connock M, Wang D, 158. Hughes JR, Stead LF, Hartmann-Boyce J, Lancaster
121. Heatherton TF, Kozlowski LT, Frecker RC, Fry-Smith A, Barton P. Effectiveness and safety of T. Antidepressants for smoking cessation. Cochrane
Fagerström KO. The Fagerström Test for Nicotine nicotine replacement therapy assisted reduction to Database Syst Rev. 2014;(1):CD000031.
Dependence: a revision of the Fagerström Tolerance stop smoking: systematic review and meta-analysis. 159. Evins AE, Cather C, Deckersbach T, et al. A
Questionnaire. Br J Addict. 1991;86(9):1119– BMJ. 2009;338:b1024. double-blind placebo-controlled trial of bupropion
1127. 140. Alpert HR, Connolly GN, Biener L. A prospec- sustained-release for smoking cessation in schizophre-
122. Heatherton TF, Kozlowski LT, Frecker RC, Rickert tive cohort study challenging the effectiveness of nia. J Clin Psychopharmacol. 2005;25(3):218–225.
W, Robinson J. Measuring the heaviness of smoking: population-based medical intervention for smoking 160. Beckham JC. Smoking and anxiety in combat
using self-reported time to the first cigarette of the cessation. Tob Control. 2013;22(1):32–37. veterans with chronic posttraumatic stress disorder:
day and number of cigarettes smoked per day. 141. Fagerström KO, Tejding LR, Ake W, Lunell E. Aiding a review. J Psychoactive Drugs. 1999;31(2):103–
Br J Addict. 1989;84(7):791–799. reduction of smoking with nicotine replacement 110.
123. Benowitz NL, Kuyt F, Jacob PI. Circadian blood medications. A strategy for the hopeless? Tob Control. 161. Hertzberg MA, Moore SD, Feldman ME, Beckham
nicotine concentrations during cigarette smoking. 1997;6(4):311–316. JC. A preliminary study of bupropion sustained-
Clin Pharmacol Ther. 1982;32:758–764. 142. Schnoll RA, Goelz PM, Veluz-Wilkins A, et al. release for smoking cessation in patients with chronic
124. Payne TJ, Smith PO, McCracken LM, McSherry Long-term nicotine replacement therapy: a random- posttraumatic stress disorder. J Clin Psychopharmacol.
WC, Antony MM. Assessing nicotine dependence: a ized clinical trial. JAMA Intern Med. 2015;175(4): 2001;21(1):94–98.
comparison of the Fagerström Tolerance Question- 504–511. 162. Mooney ME, Sofuoglu M. Bupropion for the treat-
naire (FTQ) with the Fagerström Test for Nicotine 143. Silagy C, Lancaster T, Stead L, Mant D, ment of nicotine withdrawal and craving. Expert Rev
Dependence (FTND) in a clinical sample. Addict Fowler G. Nicotine replacement therapy for Neurother. 2006;6(7):965–981.
Behav. 1994;19(3):307–317. smoking cessation. Cochrane Database Syst Rev. 163. Gonzales D, Rennard SI, Nides M, et al. Varenicline,
125. Pomerleau CS, Pomerleau OF, Majchrzak MJ, Kloska 2004;(3):CD000146-CD000146. an alpha4beta2 nicotinic acetylcholine receptor
DD, Malakuti R. Relationship between Fagerström 144. Kozlowski LT, Giovino GA, Edwards B, et al. Advice partial agonist, vs sustained-release bupropion
Tolerance Questionnaire scores and plasma cotinine. on using over-the-counter nicotine replacement and placebo for smoking cessation: a randomized
Addict Behav. 1990;15:73–80. therapy-patch, gum or lozenge-to quit smoking. controlled trial. JAMA. 2006;296(1):47–55.
126. Dijkstra A, Tromp D. Is the FTND a measure of Addict Behav. 2007;32(10):2140–2150. 164. Gonzales DH, Rennard SI, Billing CB, Reeves K,
physical as well as psychological tobacco dependence? 145. Bolliger CT, Zellweger JP, Danielsson T, et al. Watsky E, Gong J. A pooled analysis of varenicline,
J Subst Abuse Treat. 2002;23(4):367–374. Smoking reduction with oral nicotine inhalers: an alpha 4 beta 2 nicotinic receptor partial agonist
127. Nayan S, Gupta MK, Sommer DD. Evaluating double blind, randomised clinical trial of efficacy vs bupropion, and placebo for smoking cessation.
smoking cessation interventions and cessation and safety. BMJ. 2000;321(7257):329–333. February; Orlando, FL.
rates in cancer patients: a systematic review and 146. Etter JF, Laszlo E, Zellweger JP, Perrot C, Perneger 165. Jorenby DE, Hays JT, Rigotti NA, et al. Efficacy of
meta-analysis. ISRN Oncol. 2011;2011:849023. TV. Nicotine replacement to reduce cigarette varenicline, an alpha4beta2 nicotinic acetylcholine
128. US National Cancer Institute. Smoking Cessation consumption in smokers who are unwilling to receptor partial agonist, vs placebo or sustained-
and Continued Risk in Cancer Patients. Bethesda, quit: a randomized trial. J Clin Psychopharmacol. release bupropion for smoking cessation: a random-
MD 2012. http://www.cancer.gov/cancertopics/pdq/ 2002;22(5):487–495. ized controlled trial. JAMA. 2006;296(1):56–63.
supportivecare/smokingcessation/HealthProfessional. 147. Karam-Hage M, Cinciripini PM. Pharmaco- 166. Tonstad S, Tonnesen P, Hajek P, Williams KE, Billing
Updated July 2, 2012. therapy for tobacco cessation: nicotine agonists, CB, Reeves KR. Effect of maintenance therapy with
129. PDR Staff. Physician Desk Reference 2012. Montvale: antagonists, and partial agonists. Curr Oncol Rep. varenicline on smoking cessation: a randomized
Thompson Publications; 2012. 2007;9(6):509–516. controlled trial. JAMA. 2006;296(1):64–71.
130. Anthenelli RM, Blom TJ, McElroy SL, Keck PEJ. 148. Wennike P, Danielsson T, Landfeldt B, Westin A, 167. Rennard S, Hughes J, Cinciripini PM, et al. A
Preliminary evidence for gender-specific effects of Tonnesen P. Smoking reduction promotes smoking randomized placebo-controlled trial of varenicline
topiramate as a potential aid to smoking cessation. cessation: results from a double blind, randomized, for smoking cessation allowing flexible quit dates.
Addiction. 2008;103(4):687–694. placebo-controlled trial of nicotine gum with 2-year Nicotine Tob Res. 2012;14(3):343–350.
131. Blodgett JC, Del Re AC, Maisel NC, Finney JW. A follow-up. Addiction. 2003;98(10):1395–1402. 168. Cinciripini PM, Robinson JD, Karam-Hage
meta-analysis of topiramate’s effects for individuals 149. McClure JB, Swan GE. Tailoring nicotine replace- M, et al. Effects of varenicline and bupropion
with alcohol use disorders. Alcohol Clin Exp Res. ment therapy: rationale and potential approaches. sustained-release use plus intensive smoking ces-
2014;38(6):1481–1488. CNS Drugs. 2006;20(4):281–291. sation counseling on prolonged abstinence from
410.e4 Part I: Science and Clinical Oncology

smoking and on depression, negative affect, and 187. Ebbert JO, Hatsukami DK, Croghan IT, et al. Clinical practice guideline. http://rc.rcjournal.com/
other symptoms of nicotine withdrawal. JAMA Combination varenicline and bupropion SR for content/53/9/1217.short. Updated March 4, 2015.
Psychiatry. 2013;70(5):522–533. tobacco-dependence treatment in cigarette smokers: 208. Fiore MC, Jorenby DE, Schensky AE, Smith SS,
169. Ebbert JO, Croghan IT, Hurt RT, Schroeder DR, a randomized trial. JAMA. 2014;311(2):155– Bauer RR, Baker TB. Smoking status as the new
Hays JT. Varenicline for smoking cessation in light 163. vital sign: effect on assessment and intervention
smokers. Nicotine Tob Res. 2016;18(10):2031–2035. 188. Rose JE, Behm FM. Combination treatment with in patients who smoke. Mayo Clinic Proceedings.
170. Anthenelli RM, Benowitz NL, West R, et al. varenicline and bupropion in an adaptive smoking 1995;70:209–213.
Neuropsychiatric safety and efficacy of varenicline, cessation paradigm. Am J Psychiatry. 2014;171(11): 209. Lindholm C, Adsit R, Bain P, et al. A demonstra-
bupropion, and nicotine patch in smokers with and 1199–1205. tion project for using the electronic health record
without psychiatric disorders (EAGLES): a double- 189. Rose JE, Behm FM. Combination varenicline/ to identify and treat tobacco users. Wisc Med J.
blind, randomised, placebo-controlled clinical trial. bupropion treatment benefits highly dependent 2010;109(6):335–340.
Lancet. 2016;387(10037):2507–2520. smokers in an adaptive smoking cessation paradigm. 210. Rigotti NA. Integrating comprehensive tobacco
171. Baker TB, Piper ME, Stein JH, et al. Effects of Nicotine Tob Res. 2017;19(8):999–1002. treatment into the evolving US health care system:
nicotine patch vs varenicline vs combination 190. Gourlay SG, Stead LF, Benowitz NL. Clonidine it’s time to act: comment on “A randomized trial
nicotine replacement therapy on smoking cessation for smoking cessation. Cochrane Database Syst Rev. of Internet and telephone treatment for smoking
at 26 weeks: a randomized clinical trial. JAMA. 2004;(3):CD000058. cessation”. Arch Intern Med. 2011;171(1):53–55.
2016;315(4):371–379. 191. Hughes JR, Stead LF, Lancaster T. Antidepressants 211. Stead LF, Hartmann-Boyce J, Perera R, Lancaster
172. Price S, Hitsman B, Veluz-Wilkins A, et al. The use for smoking cessation. Cochrane Database Syst Rev. T. Telephone counseling for smoking cessation.
of varenicline to treat nicotine dependence among 2003;(2):CD000031. Cochrane Database Syst Rev. 2013;(8):CD002850.
patients with cancer. Psychooncology. 2016. 192. Hughes JR, Stead LF, Lancaster T. Nortriptyline 212. Lichtenstein E, Zhu SH, Tedeschi GJ. Smoking
173. US Food and Drug Administration. Public Health for smoking cessation: A review. Nicotine Tob Res. cessation quitlines: an underrecognized intervention
Advisory, Important Information on Chantix 2005;7(4):491–499. success story. Am Psychol. 2010;65(4):252–261.
(varenicline). Rockville, MD; 2008. 193. US Centers for Disease Control and Prevention. 213. Rabius V, McAlister AL, Geiger A, Huang P,
174. Williams JM, Anthenelli RM, Morris CD, et al. A Cigarette smoking among adults and trends in Todd R. Telephone counseling increases cessation
randomized, double-blind, placebo-controlled study smoking—United States 2008. MMWR Morb Mortal rates among young adult smokers. Health Psychol.
evaluating the safety and efficacy of varenicline for Wkly Rep. 2012;Updated July 3. 2004;23(5):539–541.
smoking cessation in patients with schizophrenia 194. Gritz ER, Lam CY, Vidrine DJ, Fingeret MC. 214. Rabius V, Wiatrek D, McAlister AL. African
or schizoaffective disorder. J Clin Psychiatry. Tobacco dependence and its treatment. In: DeVita V, American participation and success in telephone
2012;73(5):654–660. Lawrence T, Rosenberg S, eds. Cancer: Principles and counseling for smoking cessation. Nicotine Tob Res.
175. Anthenelli RM, Morris C, Ramey TS, et al. Effects Practice of Oncology. 9th ed. Philadelphia: Lippincott 2012;14(2):240–242.
of varenicline on smoking cessation in adults with Williams & Wilkins; 2012:529–542. 215. Karn S, Fernandez A, Grossberg LA, et al. Systemati-
stably treated current or past major depression: a 195. Gritz ER, Carmack CL, de Moor C, et al. First cally improving tobacco cessation patient services
randomized trial. Ann Intern Med. 2013;159(6):390– year after head and neck cancer: quality of life. through electronic medical record integration. Health
400. J Clin Oncol. 1999;17(1):352–360. Promot Pract. 2016;17(4):482–489.
176. Gibbons RD, Mann JJ. Varenicline, smoking ces- 196. Gritz ER, Dresler C, Sarna L. Smoking, the 216. Guy MC, Seltzer RG, Cameron M, Pugmire J,
sation, and neuropsychiatric adverse events. Am J missing drug interaction in clinical trials: Michael S, Leischow SJ. Relationship between
Psychiatry. 2013;170(12):1460–1467. ignoring the obvious. Cancer Epidem Biomar. smokers’ modes of entry into quitlines and treatment
177. Thomas KH, Martin RM, Knipe DW, Higgins JP, 2005;14(10):2287–2293. outcomes. Am J Health Behav. 2012;36(1):3–11.
Gunnell D. Risk of neuropsychiatric adverse events 197. Gritz ER, Fingeret MC, Vidrine DJ, Lazev AB, 217. Lancaster T, Stead LF. Self-help interventions for
associated with varenicline: systematic review and Mehta NV, Reece GP. Successes and failures of the smoking cessation. Cochrane Database Syst Rev.
meta-analysis. BMJ. 2015;350:h1109. teachable moment: smoking cessation in cancer 2005;(3):CD001118.
178. Mitchell JM, Teague CH, Kayser AS, Bartlett SE, patients. Cancer. 2006;106(1):17–27. 218. Graham AL, Cobb NK, Papandonatos GD, et al.
Fields HL. Varenicline decreases alcohol consump- 198. Jensen K, Jensen AB, Grau C. Smoking has a nega- A randomized trial of Internet and telephone
tion in heavy-drinking smokers. Psychopharmacology tive impact upon health related quality of life after treatment for smoking cessation. Arch Intern Med.
(Berl). 2012;223:299–306. treatment for head and neck cancer. Oral Oncol. 2011;171(1):46.
179. Lubin JH, Purdue M, Kelsey K, et al. Total 2007;43(2):187–192. 219. Rabius V, Pike KJ, Wiatrek D, McAlister AL.
exposure and exposure rate effects for alcohol and 199. Park SM, Lim MK, Shin SA, et al. Impact of Comparing internet assistance for smoking cessa-
smoking and risk of head and neck cancer: a pooled prediagnosis smoking, alcohol, obesity, and insulin tion: 13-month follow-up of a six-arm randomized
analysis of case-control studies. Am J Epidemiol. resistance on survival in male cancer patients: controlled trial. J Med Internet Res. 2008;10(5):e45.
2009;170(8):937–947. National Health Insurance Corporation Study. 220. Civljak M, Sheikh A, Stead LF, Car J. Internet-based
180. Marron M, Boffetta P, Zhang Z-F, et al. Cessation of J Clin Oncol. 2006;24(31):5017–5024. interventions for smoking cessation—Updated 2010.
alcohol drinking, tobacco smoking and the reversal 200. Yu GP, Ostroff JS, Zhang ZF, Tang J, Schantz SP. Cochrane Database Syst Rev. 2010;(9):CD007078,
of head and neck cancer risk. Int J Epidemiol. Smoking history and cancer patient survival: a Updated July 5, 2012.
2010;39(1):182–196. hospital cancer registry study. Cancer Detect Prev. 221. Swartz LH, Noell JW, Schroeder SW, Ary DV.
181. Cahill K, Stead LF, Lancaster T. Nicotine receptor 1997;21(6):497–509. A randomised control study of a fully automated
partial agonists for smoking cessation. Cochrane 201. Ehlers SL, Gastineau DA, Patten CA, et al. The internet based smoking cessation programme. Tob
Database Syst Rev. 2011;(2):CD006103-CD006103. impact of smoking on outcomes among patients Control. 2006;15(1):7–12.
182. Shah SD, Wilken LA, Winkler SR, Lin SJ. Sys- undergoing hematopoietic SCT for the treatment 222. Walters ST, Wright JA, Shegog R. A review of com-
tematic review and meta-analysis of combination of acute leukemia. Bone Marrow Transplant. puter and Internet-based interventions for smoking
therapy for smoking cessation. J Am Pharm Assoc. 2011;46(2):285–290. behavior. Addict Behav. 2006;31(2):264–277.
2008;48(5):659–665. 202. Gritz ER. Smoking and smoking cessation in cancer 223. Duffy SA, Ronis DL, Valenstein M, et al. A tailored
183. Loh WY, Piper ME, Schlam TR, et al. Should patients. Br J Addict. 1991;86:549–554. smoking, alcohol, and depression intervention for
all smokers use combination smoking cessation 203. Lippman SM, Lee JJ, Karp DD, et al. Random- head and neck cancer patients. Cancer Epidemiol
pharmacotherapy? Using novel analytic methods ized phase III intergroup trial of isotretinoin to Biomarkers Prev. 2006;15(11).
to detect differential treatment effects over 8 weeks prevent second primary tumors in stage I non- 224. Gritz ER, Carr CR, Rapkin D, et al. Predictors of
of pharmacotherapy. Nicotine Tob Res. 2012;14(2): small-cell lung cancer. National Cancer Institute. long-term smoking cessation in head and neck cancer
131–141. 2001;93(8):605–618. patients. Cancer Epidem Biomar. 1993;2:261–270.
184. Piper ME, Smith SS, Schlam TR, et al. A random- 204. Walker MS, Vidrine DJ, Gritz ER, et al. Smoking 225. Schnoll RA, Rothman RL, Wielt DB, et al. A
ized placebo-controlled clinical trial of 5 smoking relapse during the first year after treatment for randomized pilot study of cognitive-behavioral
cessation pharmacotherapies. Arch Gen Psychiatry. early-stage non-small-cell lung cancer. Cancer Epidem therapy versus basic health education for smoking
2009;66(11):1253–1262. Biomar. 2006;15(12):2370–2377. cessation among cancer patients. Ann Behav Med.
185. Smith SS, McCarthy DE, Japuntich SJ, et al. 205. US Centers for Disease Control and Prevention. 2005;30(1):1–11.
Comparative effectiveness of 5 smoking cessation Tobacco use among adults—United States, 2005. 226. Griebel B, Wewers ME, Baker CA. The effectiveness
pharmacotherapies in primary care clinics. Arch MMWR Morb Mortal Wkly Rep. 2006;55:1145. of a nurse-managed minimal smoking-cessation
Intern Med. 2009;169(22):2148–2155. 206. Gritz ER, Fingeret MC, Vidrine DJ. Tobacco Control intervention among hospitalized patients with cancer.
186. Ebbert JO, Croghan IT, Sood A, Schroeder DR, Hays in the oncology setting. 2007. Cancer Prevention: Oncology Nurses Forum. 1998;25:897–902.
JT, Hurt RD. Vareniclin and bupropion sustained An ASCO Curriculum. 227. Wewers ME, Bowen JM, Stanislaw AE, Desimone
release combination therapy for smoking cessation. 207. Fiore MC, Bailey WC, Cohen SJ, et  al. VB. A nurse-delivered smoking cessation intervention
Nicotine Tob Res. 2009;11(3):234–239. Treating tobacco use and dependence. among hospitalized postoperative patients—influence
Nicotine Dependence: Current Treatments and Future Directions  •  CHAPTER 24 410.e5
410.e5

of a smoking-related diagnosis: a pilot study. Heart cancer: characteristics of electronic cigarette users 252. Kreuzer M, Boffetta P, Whitley E, et al. Gender
Lung. 1994;23(2):151. and their smoking cessation outcomes. Cancer. differences in lung cancer risk by smoking: a
228. Garces YI, Yang P, Parkinson J, et al. The 2014;120(22):3527–3535. multicentre case-control study in Germany and
relationship between cigarette smoking and 241. NIDA. The Neurobiology of Drug Addiction. Italy. Br J Cancer. 2000;82(1):227–233.
quality of life after lung cancer diagnosis. Chest. http://www.nida.nih.gov/pubs/teaching/Teaching2/ 253. Marshak G, Brenner B, Shvero J, et al. Prognostic
2004;126(6):1733–1741. Teaching.html. factors for local control of early glottic cancer:
229. Emmons KM, Puleo E, Park E, et al. Peer-delivered 242. Fortin A, Wang CS, Vigneault E. Influence of the Rabin Medical Center retrospective study
smoking counseling for childhood cancer survivors smoking and alcohol drinking behaviors on treatment on 207 patients. Int J Radiat Oncol Biol Phys.
increases rate of cessation: the partnership for health outcomes of patients with squamous cell carcinomas 1999;43(5):1009–1013.
study. J Clin Oncol. 2005;23(27):6516–6523. of the head and neck. Int J Radiat Oncol Biol Phys. 254. Fleshner N, Garland J, Moadel A, et al. Influence
230. Wakefield M, Olver I, Whitford H, Rosenfeld E. 2009;74(4):1062–1069. of smoking status on the disease-related outcomes
Motivational interviewing as a smoking cessation 243. Rades D, Setter C, Schild SE, Dunst J. Effect of of patients with tobacco-associated superficial
intervention for patients with cancer: randomized smoking during radiotherapy, respiratory insuf- transitional cell carcinoma of the bladder. Cancer.
controlled trial. Nurs Res. 2004;53(6):396. ficiency, and hemoglobin levels on outcome in 1999;86(11):2337–2345.
231. Mee-Lee D, Shulman GD, Fishman M, Gastfriend patients irradiated for non-small-cell lung cancer. 255. Daniell HW. A worse prognosis for smokers with
DR, Griffith JH, eds. ASAM Patient placement criteria Int J Radiat Oncol Biol Phys. 2008;71(4):1134– prostate cancer. J Urol. 1995;154(1):153–157.
for the treatment of substance-related disorders, Second 1142. 256. Daniell HW. More advanced-stage tumors among
Edition-Revised (ASAM PPC-2R). Chevy Chase: 244. Marin VP, Pytynia KB, Langstein HN, Dahl- smokers with endometrial cancer. Am J Clin Pathol.
American Society of Addiction Medicine, Inc; 2001. strom KR, Wei Q, Sturgis EM. Serum cotinine 1993;100(4):439–443.
232. Karam-Hage M, Oughli HA, Rabius V, et al. concentration and wound complications in head 257. Risch HA, Howe GR, Jain M, Burch JD, Holowaty
Tobacco cessation treatment pathways for patients and neck reconstruction. Plast Reconstr Surg. EJ, Miller AB. Are female smokers at higher risk for
with cancer: 10 years in the making. J Natl Compr 2008;121(2):451–457. lung cancer than male smokers? Am J Epidemiol.
Canc Netw. 2016;14(11):1469–1477. http://www. 245. Chen CH, Shun CT, Huang KH, et al. Stop- 1993;138:281–293.
jnccn.org/content/14/11/1469.full. ping smoking might reduce tumour recurrence 258. Rugg T, Saunders MI, Dische S. Smoking and
233. American Telemedicine Association. Telemedicine in nonmuscle-invasive bladder cancer. BJU Int. mucosal reactions to radiotherapy. Br J Radiol.
Standards and Guidelines. http://www.american- 2007;100(2):281–286. 1990;63:554–556.
telemed.org/i4a/pages/index.cfm?pageID=3311. 246. Tammemagi CM, Neslund-Dudas C, Simoff 259. Archimbaud E, Maupas J, Lecluze-Palazzolo C, Fiere
Updated July 3, 2012. M, Kvale P. Smoking and lung cancer survival: D, Viala JJ. Influence of cigarette smoking on the
234. Zhu SH, Gamst A, Lee M, Cummins S, Yin L, Zoref the role of comorbidity and treatment. Chest. presentation and course of chronic myelogenous
L. The use and perception of electronic cigarettes 2004;125(1):27–37. leukemia. Cancer. 1989;63(10):2060–2065.
and snus among the US population. PLoS ONE. 247. Pytynia KB, Grant JR, Etzel CJ, Roberts DB, Wei 260. Daniell HW. Increased lymph node metastases at
2013;8(10):e79332. Q, Sturgis EM. Matched-pair analysis of survival mastectomy for breast cancer associated with host
235. Brown CJ, Cheng JM. Electronic cigarettes: product of never smokers and ever smokers with squamous obesity, cigarette smoking, age, and large tumor
characterisation and design considerations. Tob cell carcinoma of the head and neck. J Clin Oncol. size. Cancer. 1988;62(2):429–435.
Control. 2014;23(suppl 2):ii4–ii10. http://tobac- 2004;22(19):3981–3988. 261. Daniell HW. More advanced colonic cancer among
cocontrol.bmj.com/content/23/suppl_2/ii1.full. 248. Fox JL, Rosenzweig KE, Ostroff JS. The effect smokers. Cancer. 1986;58(3):784–787.
pdf+html. of smoking status on survival following radiation 262. Phillips B, Marshall E, Brown S, Thompson JS. Effect
236. Eissenberg T. Electronic nicotine delivery devices: therapy for non-small cell lung cancer. Lung Cancer. of smoking on human natural killer cell activity.
Ineffective nicotine delivery and craving sup- 2004;44(3):287–293. Cancer. 1985;56:2789–2792.
pression after acute administration. Tob Control. 249. Nordquist LT, Simon GR, Cantor A, Alberts WM, 263. Stevens MH, Gardner JW, Parkin JL, Johnson LP.
2010;19(1):87–88. Bepler G. Improved survival in never-smokers vs Head and neck cancer survival and lifestyle change.
237. Vansickel AR, Eissenberg T. Electronic cigarettes: current smokers with primary adenocarcinoma of Arch Otolaryngol. 1983;109:746–749.
effective nicotine delivery after acute administration. the lung. Chest. 2004;126(2):347–351. 264. Daniell HW. Estrogen receptors, breast cancer,
Nicotine Tob Res. 2013;15(1):267–270. 250. Videtic GM, Stitt LW, Dar AR, et al. Continued and smoking. N Engl J Med. 1980;302(26):
238. US Department of Health and Human Services. cigarette smoking by patients receiving concurrent 1478.
E-cigarette use among youth and young adults: A chemoradiotherapy for limited-stage small-cell lung 265. Hinds MW, Yang HY, Stemmermann G, Lee
report of the Surgeon General. Rockville MD; 2016. cancer is associated with decreased survival. J Clin J, Kolonel LN. Smoking history and lung
239. Dautzenberg B, Garelik D. Patients with lung Oncol. 2003;21(8):1544–1549. cancer survival in women. J Natl Cancer Inst.
cancer: are electronic cigarettes harmful or useful? 251. Chelghoum Y, Danaila C, Belhabri A, et al. 1982;68(3):395–399.
Lung Cancer. 2016. Influence of cigarette smoking on the presentation 266. Johnston-Early A, Cohen MH, Minna JD, et al.
240. Borderud SP, Li Y, Burkhalter JE, Sheffer CE, Ostroff and course of acute myeloid leukemia. Ann Oncol. Smoking abstinence and small cell lung cancer
JS. Electronic cigarette use among patients with 2002;13(10):1621–1627. survival. JAMA. 1980;244:2175–2179.
F. TREATMENT
Cancer Pharmacology 25 
Jerry M. Collins

S UMMARY OF K EY P OI NT S
• Cancer pharmacology encompasses and clinical success has • The era of oral drugs for cancer
the spectrum of therapeutic issues attracted wide corporate treatment is well underway.
from everyday clinical treatment to involvement. • Once the drug is selected to match
the earliest stages of new drug • Regulatory flexibility has made new the tumor, drug delivery must
discovery. therapies available to patients at provide adequate systemic exposure
• Translational research has led to an unprecedented rate and has for tumors while minimizing toxicity
more sophisticated identification of provided a variety of options for and adjusting for concomitant
targets that are relevant for therapy, developmental strategies. therapy.

Cancer pharmacology offers an integrated body of practical knowledge the beginning of the shift from intravenous to oral drug delivery in
that is valuable for the full range of activities related to cancer thera- patients with cancer.
peutics, from everyday clinical treatment decisions, to assisting clinical Although this chapter focuses on small, synthetic molecules, one
trial design in drug development, to discovery and evaluation of of the most exciting developments in cancer pharmacology is the
compounds with potential for advancing to the clinic. successful transition of antibodies from the research stage to full
One of the most obvious metrics for the importance of cancer integration with small molecules in the everyday treatment of patients.
pharmacology is the rapid and continuing expansion in the number There is also a role for toxins that are linked to antibodies, which can
of drugs available to treat cancer. More than 200 agents have been help to guide the toxin to the tumor (see Chapter 30). Although very
approved for marketing in oncology, including 51 that were approved different in terms of their size and manufacturing techniques, the
by the US Food and Drug Administration (FDA) from 2011 to 2015. principles of development for these new therapeutic classes have the
Notably, 15 were approved in 2015 (Table 25.1). As a point of reference, same goals as for small molecules—for example, modulation of specific
at the first meeting of the American Society of Clinical Oncology molecular targets and linkage to a diagnostic test for that target.
(ASCO) in 1964, there were only 12 drugs that had been previously Trastuzumab embodies all of these elements, including its extension
approved for marketing in oncology. into the realm of antibody-drug conjugates with the 2013 FDA approval
When ASCO was founded, the National Cancer Institute (NCI) of ado-trastuzumab emtansine.
was the primary source for new cancer drugs, and the NCI continues The rapid increase in availability of new drugs and new data on
to conduct an extensive program of cancer drug development. NCI how to optimally use drugs, including the ability to avoid drug-drug
also collaborates extensively with industry and other organizations. and drug-food interactions, is an enormously valuable situation for
Over the past 25 years, the pharmaceutical industry has dramatically patients with cancer. How will oncologists keep up with the avalanche
increased the resources invested in cancer pharmacology. Various of information for all of these issues? Everything an oncologist needs
nonprofit organizations also are engaged in the development of cancer to know can no longer be squeezed into a small book that fits in the
drugs, but the private sector is the largest contributor to the effort to pocket of the physician’s white coat. Electronic versions of textbooks
bring new treatments to patients with cancer. and centralized electronic databases help provide the intricate pieces
Beyond the impressive increase in the number of drugs available of data. In this chapter the focus is on the principles of cancer
to treat cancer, other factors are fundamentally changing the way that pharmacology and its ability to help categorize data and organize it
oncologists and drug developers discover, develop, and prescribe drugs into useful information.
for treatment of patients with cancer. In most cases, there is a strong
trend toward narrow indications, as the broad histopathologic categories FUNDAMENTAL SCIENCE
of cancer have become fractionated into many subtypes. There is also
a countertrend: seeking drug approvals based on the target for a drug, The two major disciplines within cancer pharmacology are drug actions
rather than the tissue site. In addition, because effective therapy is (pharmacodynamics [PD]) and drug delivery (pharmacokinetics [PK]).
more widely available for first-line treatment of many cancers, initial Both of these areas are highly dependent on knowledge derived from
approvals are further stratified by the extent of prior treatment. pharmacogenetics and pharmacogenomics.
The approval of imatinib in 2001 heralded several of the most In cancer pharmacology the primary basis for success or failure of
dramatic changes in cancer therapy, foreshadowing entirely new para- drug therapy is the ability to find a treatment that is matched to the
digms that became commonplace during the next 15 years: molecular intrinsic sensitivity of the tumor. No therapeutic benefit will be achieved
targeting, diagnostic tests associated with selection of therapy, and if the target for drug action is not present in the tumor. However,

411
412 Part I: Science and Clinical Oncology

Table 25.1 Cancer Drugs Approved by the Food Box 25.1. STRATEGIC FACTORS FOR DRUG
and Drug Administration in 2015a COMBINATIONS
Drug Route Type • Rationale for target-based combinations
Daratumumab Intravenous Monoclonal antibody • Addition of new drug to established regimen
• Systematic evaluation of all combinations
Dinutuximab Intravenous Monoclonal antibody
• Translation of findings into clinical trials
Elotuzumab Intravenous Monoclonal antibody
Necitumumab Intravenous Monoclonal antibody
Alectinib Oral Small synthetic molecule
Cobimetinib Oral Small synthetic molecule (CML) that is Philadelphia chromosome positive. These early successes
Ixazomib Oral Small synthetic molecule fueled emphasis on other targets. ALK and BRAF are among the
Lenvatinib Oral Small synthetic molecule many descendants of the approach. In 2017, the target became more
Osimertinib Oral Small synthetic molecule
pivotal with the approval for pembrolizumab for treatment of tumors
with tumors that demonstrate mismatch repair or microsatellite
Panobinostat Oral Small synthetic molecule instability, regardless of tumor’s site of origin.
Palbociclib Oral Small synthetic molecule In addition to the obvious attraction of matching drugs to the
Sonidegib Oral Small synthetic molecule molecular characteristics of the tumor, the therapeutic index can also
Trabectedin Intravenous Small synthetic molecule be improved by examining host tissues. In his last major research
Trifluridine/tipiracil Oral Small synthetic molecule
publication,1 Dr. Abeloff was a leader in a large multicenter study of
the pharmacogenetics of cyclophosphamide. The goal was to determine
Uridine triacetate Oral Small synthetic molecule whether the germline DNA of patients altered the toxicity profile of
a
cyclophosphamide. The researchers found a subgroup of women who
Of the 15 new drugs, 4 are monoclonal antibodies delivered intravenously. Of the
11 small molecules, 10 are delivered orally.
had variant GSTP1 alleles that were associated with less susceptibility
to adverse hematologic toxicity in regimens containing cyclophospha-
mide. Subsequent work by large cooperative groups in the United
Kingdom for paclitaxel in breast cancer2 and in the United States for
the perfect match between target and therapy can still fail if adequate docetaxel in prostate cancer3 found other links between germline DNA
systemic exposure of drug is not delivered to the tumor. and sensory neuropathy.
Systemic exposure is pivotal. For a responsive tumor, higher systemic Although these studies were only hypothesis generating, the examples
exposure can be associated with greater benefit but also more adverse illustrate the possible ways that the therapeutic index for a drug can
effects in normal tissue. Lower systemic exposure can have lower be improved with examination of host tissues. Later in this chapter,
toxicity in normal tissues, but at the risk of reduced effect on the in the discussion of drug delivery, the pharmacogenetics of metabolism
tumor. The therapeutic index for a cancer drug is the ratio of its and cellular membrane transporter cell membranes in host tissues are
antitumor activity versus the adverse effects on normal tissues. discussed.
The principles that connect dose, systemic exposure, and toxicity
have been demonstrated regularly for normal tissues in the body, Combinations of Drugs
but the relationships of systemic exposure to antitumor effect are far
more difficult to discern. Understanding the linkage between drug As described in the various chapters relating to specific malignancies
delivery and drug actions continues to be a major challenge in cancer in Part III of this book, it is rare for a drug to be used as a single
pharmacology. agent. The development of combinations would be an extensive topic
in itself. Box 25.1 provides a set of points to consider for combinations.
Principles of Cancer Drug Action Within a combination chemotherapy regimen, the drugs are intended
to interact at the pharmacodynamic level. Originally, a major strategy
Genetics is a fundamental part of the study of cancer research (see was to combine cancer drugs with nonoverlapping toxicities. Increas-
Chapter 1). The range of applications for genetics in cancer includes ingly, more detailed knowledge regarding pathways for cancer drugs
hereditary predisposition and interactions of heredity with nonhereditary permits the design and evaluation of combination strategies that target
factors (see Chapter 13). parallel and/or sequential pathways.
In cancer pharmacology, molecular targets that could be relevant Most combinations are based on a preexisting scientific rationale
to therapy have received the most intense emphasis of pharmacogenom- or by addition of a new agent to an established regimen. However,
ics. This focus includes unique proteins coded by translocations, it would be presumptuous to assume that all possibilities are already
differences in gene expression between tumors and host tissues, and understood. One tool to expand the scope of hypothesis generation
mutations in sequences. The identification of the most attractive is the systematic study of all cancer drugs in combination with one
molecular targets and the ability to monitor target engagement during another or in combination with approved agents outside oncology,
therapy are cornerstones for customized treatment. or the testing of every investigational agent versus all approved cancer
As described in Chapter 26 and elsewhere (e.g., Chapter 8), our drugs.4 Although this screening approach begins as an empiric exercise,
understanding of the physiology and molecular biology of tumors has the successful combinations generate challenges for explaining the
provided a wealth of potential targets for anticancer therapy. Although underlying mechanisms of action. The focus on drugs that are already
the relative intensity has magnified, the concept of molecular targeting approved provides a potentially fast route to clinical implementation.5
and linkage to diagnostics for selection and monitoring of individual
patients has a long history. The selection of hormonal therapies for Molecular Imaging of Cancer Drug Action
patients whose tumors express the estrogen receptor was among the
first successful uses of molecular targeting. With the widespread availability of fluorine-18 fluorodeoxyglucose
The current era of targeting began with approval of trastuzumab (18F-FDG), a consensus was building in the late 1990s that positron
(1998) only in patients with Her2-positive tumors and approval of emission tomography (PET) would emerge as a tool for functional
imatinib (2001) only in patients with chronic myelogenous leukemia assessment, which would complement anatomic imaging modalities.6
Cancer Pharmacology  •  CHAPTER 25 413

Box 25.2.  DRUG DELIVERY CONCEPTS Box 25.3. ROLE OF CLEARANCE IN DOSAGE


ADJUSTMENT
Clearance (dose adjustment)
• Impaired organ function Clearance: summation of all routes of drug elimination from the
• Drug-drug interactions body―renal, hepatic, other
• Metabolic Primary application: guide dose adjustment
• Transport • Low clearance → high exposure (consider reduced dose)
• Pharmacogenetic factors • High clearance → low exposure (consider increased dose)
Bioavailability
• How much drug reaches systemic circulation
• Includes food effects
Half-life (dose interval)
Volume of distribution, protein binding
approved for clinical practice, oncologists do not have a frequent need
to know the specific value for half-life of a cancer drug. Other drug
delivery parameters such as volume of distribution or protein binding
can have value in specific situations, but only on rare occasions.

Since then, FDG has been highly successful as a general probe in Dosage Adjustment Based on Clearance to Achieve
many tumor types, providing additional information to help separate Consistent Systemic Exposure
malignant from benign lesions and to monitor response to therapy.
FDG is now an approved agent for imaging. Its success has spurred Compared with most other medical areas in which fixed doses are
interest in the development of other probes for PET imaging that are prevalent, dosage in cancer pharmacology has long been titrated on
currently in the investigational stage of clinical evaluation, including the basis of each patient’s body weight or body surface area. These
fluorine-18 fluorothymidine (18F-fluorothymidine; FLT). Liu et al. measurements are intended to serve as first approximations for clearance
demonstrated the potential for FLT in a patient with uterine cancer.7 in the absence of data, but their incremental value compared with
At baseline, the FLT image shows major uptake by the tumor, indicative simple use of a standard dose is unfortunately small, and most current
of active proliferation. During treatment with sunitinib, uptake of trials have now adopted fixed doses—that is, “flat dosing.”8
FLT in the tumor decreased, which was interpreted as reduced prolifera- The premise for the comments about dose adjustment in Box 25.3
tion. The image following the withdrawal of treatment exhibited a is that the standard dose was selected on the basis of the needs of
flare—that is, intensity that rebounded above the baseline value. patients with average drug clearance. If a patient has lower clearance
Anatomic imaging by computed tomography (CT) readily identified than the average, then using the standard dose will lead to higher
the tumor and could monitor tumor size over the subsequent weeks than average systemic exposure, with the probability of higher than
or months, but FLT provided the ability to follow molecular pathways average toxicity. Using a dose reduction to match the lower clearance
in real time. will normalize the exposure of this patient relative to the average in
the population. Similarly, a patient with higher than average clearance
Cancer Drug Delivery: Systemic Exposure will have lower systemic drug exposure than will patients with the
average exposure for the population. Although this lower systemic
Once the appropriate drug has been chosen, the most important drug exposure will probably reduce the frequency or severity of toxicity,
everyday question in clinical cancer pharmacology is the selection of it has the potentially lethal consequence of reducing the response of
an appropriate dose. The standard dose determined previously in the tumor.
population studies is a starting point for consideration, but what Information on drug clearance for individual patients is rarely
factors could make this dose too high or too low for individual patients? available. However, the validation of this overall strategy for dosage
For many cancer drugs, evidence-based advice is not available. In adjustment has been elegantly demonstrated for the first scenario in
some cases, guidance for dose adjustment is available based on prior Box 25.2, namely, patients with impaired renal function. The seminal
courses of treatment or age. clinical study of carboplatin by Egorin and colleagues9 in patients
Increasingly, cancer drugs are approved with information about with variable renal function remains the prototype for dosage adjustment
dosage adjustment based on renal function testing, food intake, and based on impaired organ function. For many drugs, renal clearance
the concurrent use of other drugs. All of these factors can produce is not the primary mode of clearance. But for those drugs for which
variability in systemic exposure. Thus the delivery of drugs to both renal clearance does dominate total body clearance, most recent drug
the tumor and normal tissues is affected. In addition to empiric data approvals have included advice in the product label for dosage adjust-
from clinical investigations for specific drugs, what are the principles ment based on renal clearance.
that underlie these decisions based on drug delivery? For the other two areas in Box 25.2 (drug-drug interactions and
Clearance is at the top of the list for drug delivery parameters in pharmacogenetic factors), many practical cases of dosage adjustment
Box 25.2 because it is the underlying factor with the greatest impact exist. Specific examples are discussed in the following section and
on dose adjustment in cancer drug delivery. As stated in Box 25.3, Table 25.2.
clearance is the summation of all mechanisms for a drug to be removed
from the systemic circulation, sometimes called total body clearance. Drug-Drug Interactions
Several specific applications of clearance for dosage adjustment are
covered in the next few sections. Many categories of drug-drug interactions exist. The anticancer drugs
Bioavailability, which is the second concept listed in Box 25.2, is in a chemotherapy regimen, which are generally intended to work at
a measure of how much drug enters the systemic circulation. Because the level of mechanisms of drug action, are only the tip of the iceberg
of the shift toward oral therapy, bioavailability has risen sharply in for drug-drug interactions. Patients with cancer are simultaneously
importance. This concept is described further in the section Oral treated with drugs for various other conditions, including diseases of
Cancer Drugs. the heart, gastrointestinal (GI) tract, and lungs; infections; depression;
Half-life was previously the most discussed parameter for cancer and the relief of pain. All of these permutations in polypharmacy can
drug delivery. However, its role is primarily in the development stage, potentially affect the systemic exposure and thus the delivery of cancer
to help explore frequency of administration. Once a drug has been drugs to the tumor and normal tissues. As a consequence of these
414 Part I: Science and Clinical Oncology

In addition to the interactions of cancer drugs with transporters


Table 25.2 Interaction of Drugs With Enzymes and in the tumor, interactions occur among cancer drugs and cellular
Transporters for Recently Approved transport systems in host tissues that control uptake and elimination
Oral Cancer Drugs of drugs from the GI tract, the liver, and the kidneys. The expression
of transporter function can be modulated by other drugs in a manner
Is This Drug Affected Does This Drug similar to drug-drug metabolic interactions. Indeed, some of the same
by Inducers or Cause Change for inducers of metabolism (phenytoin or rifampin) are also major inducers
Drug Inhibitors? Other Drugs?a of transporters. In contrast, ketoconazole, itraconazole, and certain
Abiraterone No data 2D6 psychopharmacologic agents are inhibitors. This focus on drug transport
Bosutinib 3A No data is important regardless of the route of drug delivery, but similar to
Crizotinib 3A 3A metabolic interactions, additional factors exist that are related to control
of absorption of cancer drugs from the GI tract. Transporters are also
Enzalutamide 2C8, 3A 3A, 2C9, 2C19
the underpinning of the blood-brain barrier and control the entry of
Imatinib 3A 3A, 2D6 cancer drugs into the brain to treat metastases or primary brain tumors.11
Lapatinib 3A 3A, 2D6, ABCB1
Palbociclib 3A 3A Interactions Via Self-Medication
Pazopanib 3A 3A, 2D6, 2C8
The source of drugs for patients with cancer is not restricted to those
Regorafenib 3A No data
prescribed by oncologists, or even medicines prescribed by other medical
Ruxolitinib 3A Not reported practitioners. Self-medication with over-the-counter (OTC) drugs and
Vemurafenib No data 3A, 1A2, 2D6 alternative therapies is prevalent in patients with cancer and includes
Venetoclax 3A, ABCB1 3A, ABCB1 substances that interact with cancer drugs (see Chapter 31).
Far less is known about the interactions of cancer drugs with these
a
The drug metabolism pathways are: 1A2 = CYP1A2; 3A = CYP3A; 2D6 = CYP2D6; substances than is known for prescription drugs. St. John’s wort is a
and various members of the CYP2C family, 2C8, 2C9, 2C19. For transport, the ABCB1 particularly troublesome case. It is marketed OTC for relief of depres-
(MDR1) pathway is cited. sion, which is a major issue for patients with cancer. However, this
Data retrieved from US Food and Drug Administration–approved labeling, found
at http://www.accessdata.fda.gov/scripts/cder/drugsatfda/.
product is a strong inducer of CYP3A enzymes, which inactivate
many newer oral cancer drugs. This situation once again reminds us
of the potentially serious consequences of reduced systemic exposure
and inadequate delivery of cancer drugs to the tumor.

interactions, the standard starting doses of drugs may need to be ORAL CANCER DRUGS
either reduced or increased.
Most drug-drug interactions are assessed as safety issues because Approval of imatinib in 2001 marked a major change in emphasis for
of increased systemic exposure subsequent to reduced clearance. cancer drugs. Oral drugs are overwhelmingly used in essentially all
However, induction (increasing) of clearance can decrease toxicity. other therapeutic areas. In contrast, the intravenous route of administra-
Unfortunately, although this type of interaction is “silent” in terms tion was the dominant form for cancer drugs. Exceptions included
of enhanced toxicity, reduced exposure to drug can also generate the mercaptopurine, some uses of methotrexate, and a few alkylating
worst toxicity of all: decreases in efficacy. agents. In the decade before the approval of imatinib, some attempts
Drug-drug metabolic interactions are possible for parenteral routes were made to convert approved cancer drugs from the intravenous
of drug delivery, but they are far more common for the oral route. route to oral delivery. Although those efforts were mostly unsuccessful,
Because metabolism is the primary determinant of clearance for most capecitabine was approved in 1998 as a prodrug for 5-fluorouracil
drugs, it is the dominant controller for changes in plasma concentra- (5-FU), which had been used intravenously for four decades.
tions. Following the paradigm in Box 25.3, inhibition of drug Parenteral administration will continue to be important, both for
metabolism via a drug-drug interaction will produce lower clearance the many legacy drugs that remain part of regimens with established
and thus higher systemic exposure. A reduction in dose may be necessary therapeutic value and for occasional cases in which an agent cannot
to avoid increased toxicity. Conversely, drugs such as phenytoin can be successfully delivered via the oral route. However, based on the
induce the expression of certain metabolizing enzymes, which would up-front goal to achieve oral delivery for most new cancer drugs under
decrease systemic concentrations for the drugs cleared via those enzymes, development, especially for chronic daily administration, it is easy to
because the rate of their metabolism would be faster. Decreased exposure project that the ratio of oral to intravenous cancer drugs will continue
increases the probability of inadequate delivery of the cancer drug to to rise. As shown in Table 25.1, the FDA approved 15 cancer drugs
the tumor. If safety information is available at higher doses, one option in 2015. Overall, 10 of these drugs are administered orally. Only one
is to increase the dose. Otherwise, it might be necessary to change to synthetic compound is administered intravenously.
a different therapeutic regimen. In the section Oral Cancer Drugs, Contemporary excitement regarding immuno-oncology has been
Table 25.2 provides some examples of the enzymatic pathways that driven by early success with antibodies and therapy with cells that
metabolize cancer drugs. have been reengineered ex vivo. Potential applications for small
molecules in this area are currently in earlier stages of development.
Drug-Drug Transporter Interactions In general, the advantages of oral cancer drugs are compelling, and
the shift from the prior dominance of parenteral routes is irreversible.
In cancer pharmacology, the primary focus for transport of drugs has As a consequence, issues regarding oral drugs have now emerged as
been the flux across the cell membranes of tumors. A major mechanism the aspect of cancer drug delivery that requires the most attention.
of drug resistance has been described for tumors with high expression As indicated in Box 25.4, adherence to the prescribed regimen is
of efflux pumps that reduce the availability of cancer drugs to their one of the first questions to explore. Although patients with cancer are
targets. Investigations into the influx of cancer drugs into tumors have highly motivated to carefully follow their administration instructions,
also been conducted. White and colleagues10 demonstrated that patients it is naïve to expect complete compliance and especially to ignore the
with CML had more long-term benefit from imatinib if expression problems of persistence over a long period. Marin and colleagues12
of its influx pump, OCT-1, was higher. concluded that poor adherence may be the predominant reason for
Cancer Pharmacology  •  CHAPTER 25 415

Box 25.4. ISSUES UNIQUE TO ORAL Table 25.3 Impact of Food on Drug Absorption for
DRUG DELIVERY Recently Approved Oral Oncology
Drugsa
Adherence: Does the patient take the drug?
Variability in absorption due to: Drug Take Drug With Food? Effect of Food
• Degradation in gastrointestinal tract Venetoclax Yes Fourfold increase
• Food effect on drug absorption
Palbociclib Yes ?? (reduces variability)
• Metabolism and transport in liver and intestines
• Drug-drug interactions prior to systemic exposure Imatinib Yes ??
• Emesis before complete absorption Lapatinib No Fourfold increase
• Comorbid difficulties in delivering oral drugs Pazopanib No ??
Ruxolitinib ± NS
Vemurafenib ± NS
Crizotinib ± NS
failure to achieve adequate molecular responses with oral imatinib.
Abiraterone No Tenfold increase
Partridge and colleagues13 examined the complex scenario of elderly
patients with cancer (older than 65 years), a population that has a high Bosutinib Yes ??
frequency of polypharmacy and perhaps a fading memory. For adjuvant Regorafenib Yes ??
therapy of early-stage breast cancer with tamoxifen, 25% of patients Enzalutamide ± NS
took fewer than 80% of the expected doses, which is problematic.
The concept known as bioavailability was introduced in Box 25.2 a
If no preference is provided for taking with food or fasting, ± is indicated, and the
as a measure of the drug delivery to the systemic circulation. Most effect size is not significant (NS). If no advice is provided, ?? is indicated.
discussion in the literature compares the ratios of equal oral and Data retrieved from Food and Drug Administration approved labeling, found at
http://www.accessdata.fda.gov/scripts/cder/drugsatfda/.
intravenous doses:

Bioavailability
of disorders, it is difficult to keep track of the instructions relating to
Systemic drug exposure ratio, at equal doses: food, which are often in conflict. Patients cannot simultaneously be
Systemic drug exposure for oral delivery fasting and eating a full meal. Fortunately, as each new set of cancer
drugs is approved, the quality and quantity of advice regarding the
Systemic drug exposure foor intravenous delivery interaction of drugs with food have improved.
For the representative set of 12 recently approved cancer drugs
Among cancer drugs, imatinib has one of the highest values for described in Table 25.3, the advice for four drugs (ruxolitinib,
oral bioavailability relative to intravenous administration: 98%. By vemurafenib, crizotinib, and enzalutamide) is that the timing of food
definition, the bioavailability of an intravenous drug is 100%. Although intake is not important. For three drugs (abiraterone, pazopanib,
it seems preferable to have high values for relative bioavailability, the lapatinib), the advice is to take while fasting. For five drugs (venetoclax,
most important test for oral administration is whether adequate systemic palbociclib, imatinib, bosutinib, regorafenib), the advice is to take
exposure can be obtained with undue variability, irrespective of its the drug with food.
relative value. For many recently approved oral drugs, there are no If these recommendations are unintentionally reversed, potentially
data for systemic exposure for an intravenous dose in humans. serious issues of reduced efficacy or increased toxicity occur. On the
Although flexibility of oral cancer drug administration is generally other hand, if clinical studies are conducted, there might be potential
a major advantage, it can become a liability when comorbid conditions economic benefit for creating new regimens that exploit increased
make swallowing drugs a major problem (see Chapter 40). Cancer absorption of some drugs with food. The fourfold increase in absorption
chemotherapy can produce emesis (see Chapter 39), which thwarts of lapatinib with food has been the source of substantial public discus-
the reliability of retaining swallowed doses. In addition, the treatment sion,16 which has been extended to cover other oral cancer drugs.17
of these complications of cancer therapy can include the use of For abiraterone, the tenfold difference in absorption is sufficiently
additional drugs to complicate the drug-drug interaction scenarios. large to capture everyone’s attention. Perhaps abiraterone has a wider
Some drugs are impractical for oral administration because they therapeutic index than most cancer drugs, but a tenfold difference in
degrade at the low pH of the stomach. At the other end of the pH exposure seems unlikely to be without consequences.
spectrum, large segments of the population are chronic users of a In addition to the general effects of food on solubility of drugs
variety of OTC agents that reduce acid in the stomach and raise pH. and passage into the body, a few food substances have highly specific
For some drugs, high pH creates a solubility or degradation problem. effects on cancer drug metabolism and cancer drug transporters. The
Unless the drug substance is stable and soluble over a wide range of most notable example is the interaction of grapefruit juice with
pH, the extensive use of acid reducers widens the variability among metabolism via the CYP3A family and the ABC family of transporters.18
patients. The specificity of effects with grapefruit and drugs closely resembles
Capecitabine is a drug for which systemic exposure is reduced at the drug-drug interactions described later. More generally, the whole
high pH, which raises a concern about drug efficacy. Indeed, recent conceptual framework for interactions of drugs with food parallels
clinical studies have demonstrated a negative association between intake the approach for drug-drug interactions.
of proton pump inhibitors and efficacy in colorectal cancer14 and When cancer drugs are administered parenterally, metabolism by
gastroesophageal cancer.15 the luminal portion of the intestine is bypassed, and metabolism by
the liver is diluted because most of the drug is distributed systemically
Effect of Food on Systemic Exposure for Oral before reaching the liver. In contrast, the entire dose of an oral cancer
Drug Delivery drug is exposed to both intestinal and hepatic metabolism and
transporters before any systemic exposure.
The most pragmatic question about oral therapy is whether the drug For the same set of 12 representative oral cancer drugs described
should be taken with food. When patients take medicines for a variety in Table 25.3 for food effects, Table 25.2 provides information from
416 Part I: Science and Clinical Oncology

the FDA website for drug interactions with enzymes and transporters. days. Subcutaneous and intravenous delivery were both approved.
The entries in Table 25.2 indicate that all of these drugs have at least The patient population for myelodysplastic syndromes has a substantial
some information about enzymatic pathways. The largest category of fraction of elderly patients, for whom there could be additional chal-
potential interactions is with the CYP3A family of enzymes, which lenges in obtaining daily intravenous treatment.
are present in both the intestines and the liver and have potential In 2012 the approval of bortezomib was extended from intravenous
interactions with 10 of the 12 drugs. Scattered information is presented delivery to include the subcutaneous route.
for other members of the CYP superfamily: 1A2, 2C8, 2C9, 2C19, The primary limitation for subcutaneous delivery is the low volume
and 2D6. Interest is expanding rapidly in the role of transporters19 of drug solution that can be delivered. The success of chronic subcutane-
interacting with cancer drugs, but only two drugs in this set, lapatinib ous insulin delivery is driven by injection of small volumes, which
and venetoclax, have information about the transporter pathways. eases patient acceptance of injection site issues.
In 2017 the FDA approved rituximab in combination with
CLINICAL RELEVANCE AND APPLICATIONS hyaluronidase for subcutaneous administration.20 Use of hyaluronidase
to facilitate absorption of complex compounds from the subcutaneous
The role of cancer pharmacology for clinical applications is to inform space could widen use of this route of delivery.
or guide oncologists in therapeutic decision making. Cancer pharmacol-
ogy is also a major factor in moving drugs from the laboratory to Regional Cancer Drug Delivery
clinical use and includes studies of formulation, animal model valida-
tion, toxicology, PK, and PD. When cancer is localized between barriers to cancer drug delivery,
A major path forward for advancing our understanding of cancer administering the drug directly to the region of disease is an attractive
drug actions includes the search for biomarkers and evaluation of treat- concept. Nonetheless, many investigations of regional drug delivery
ment effects. In the long term, the identification and implementation of have failed to demonstrate increased benefit compared with systemic
potential predictive biomarkers for selection and monitoring of therapy treatment. The choice of drug is the first consideration. Cyclophos-
is perhaps the most clinically relevant area of cancer pharmacology. phamide and other prodrugs that must be metabolized to their active
Currently, testing is underway to develop strategies to select treat- species are not suitable for regional delivery. The potential advantage
ment for patients based on molecular profiling of their tumors, including of local delivery is lost because the drug must enter the body and be
DNA (especially mutations), RNA (expression arrays and real-time metabolized and then circulate throughout the body as if it had been
polymerase chain reaction), target proteins and their modified states systemically delivered.
(e.g., phosphorylation, enzyme-linked immunosorbent assays, and Intraarterial catheterization has been technically feasible for delivery
Western blots), and functional assays such as enzymatic activity (less of cancer drugs to a variety of places in the body for decades. The
frequently). It is too early to draw conclusions about the ultimate use of an implanted pump for the intraarterial delivery of fluorode-
roles for such studies. It has become more widely appreciated that oxyuridine is approved for the treatment of liver metastases from
each of these end points has various requirements for sample handling colorectal cancer and is the most common application of this approach.20
that are crucial to interpretation of the results. Furthermore, the issue In 2017 a large international study demonstrated a survival advantage
of interlaboratory validation has moved to the top of the list of for this approach.21 Intraperitoneal delivery of drugs for treatment of
requirements for definitive progress in applying these tools. localized ovarian cancer and other intraperitoneal malignancies has
Cancer drug delivery assists in the selection of feasible doses, undergone many evaluations. Three randomized phase III studies
schedules, and routes of administration. The most appealing approach have shown a survival benefit for intraperitoneal delivery compared
to customizing doses for patients would provide advice prior to the with the intravenous route.22 Despite this success, the lack of familiarity
first dose to avoid untoward effects and maximize therapeutic delivery. with the catheters, uncertainty about uniform distribution within the
Earlier sections on impaired organ function and drug-drug interactions peritoneal cavity, and other factors have slowed the penetration of
discussed some modest advances toward this goal. this modality into routine practice.
Once therapy is ongoing, adjustments are frequently required. Intrathecal delivery remains a mainstay for pediatric leukemia. As
Dosage adjustment based on adverse effects is always the most urgent previously mentioned, the transporters that constitute the blood-brain
need. If life-threatening or unbearable toxicity occurs, then the barrier tightly control the passage of drugs into the brain. Direct
antitumor effects will be moot. An adjustment in dosage based on injection of drug into the cerebral spinal fluid produces very high
lack of antitumor activity has its attraction, particularly if the toxicity local concentrations for the areas close to the tissue–cerebrospinal
is less than expected, a potential signal of inadequate exposure. However, fluid interfaces. The major limitation of intrathecal delivery to treat
recognition of the narrow therapeutic index for cancer drugs and solid tumors is the inability of cancer drugs to penetrate deeply into
many other cautionary clinical factors or concerns about liability tend tissue to reach all of the disease.
to discourage escalating a dose higher than the standard, unless adequate The intravesicular route is standard care for some stages of bladder
safety data are available for higher doses. In addition, there is always cancer. Bacillus Calmette-Guérin (BCG) is an approved drug for this
a sense that if the current treatment is not working, switching to an indication, and several other drugs are less commonly used.
alternate therapy is urgent before the tumor becomes even more
resistant. Brand Names, Generics, Biosimilars

Subcutaneous Route of Cancer Drug Delivery When a small synthetic drug is initially approved, oncologists and
patients become accustomed to its brand name. After patents and
The subcutaneous route of delivery has emerged as an occasional exclusivity rights expire, everyone needs to readjust to the generic
solution to difficulties in drug delivery. The two issues that drive name and to additional suppliers. For small synthetic compounds,
consideration of the subcutaneous route are (1) inability of an oral generic drugs contain the same active ingredients as the initial drug.
dose to reliably traverse the GI tract and avoid presystemic metabolism, The manufacturing and quality control standards are identical. For
and (2) impracticality of the intravenous route for relatively frequent oral cancer drugs, there is the additional requirement for bioequivalence,
administration. a term that means that the original drug and the generic drug produce
The approval of 5-azacytidine in 2004 for myelodysplastic syndromes the same range of plasma concentrations. The fundamental assumption
is an ideal example to demonstrate the advantages of subcutaneous is that patients should obtain similar benefits (and toxicity profiles)
drug delivery. In addition to incompatibility with oral delivery, if the same exposure of the active ingredient is delivered. In addition
5-azacytidine requires daily parenteral administration for 7 consecutive to its use for generics, this principle is also applied for changes in the
Cancer Pharmacology  •  CHAPTER 25 417

Box 25.5.  RECENT TRENDS FOR NEW CANCER Box 25.6.  “HISTORICAL” PHASES OF HUMAN
DRUGS EVALUATION FOR CANCER DRUGS
1. Rapid pace of approvals • Phase 0: Target-related study and feasibility of delivery
2. Predominantly oral drugs • Phase I: Safety and early signs of activity
3. Narrow indications • Phase II: Is anticancer activity promising?
4. Links to molecular targeting and diagnostic tests • Phase III: Improvement versus current therapy?
5. Involvement of industry, nonprofit organizations, and government • Phase IV: Postmarketing studies

production of brand name drugs, including changes in suppliers of across phases are always being pushed. It is not possible to assign
bulk ingredients, manufacturing processes, or locations for producing every compound into the same bins. For patients with cancer, this
pills. All of these events occur throughout the marketing history of view of clinical development is continually challenged by the compelling
the drug but are not highly visible to oncologists and their patients. need to provide therapeutic options when no treatments have been
The principles for ensuring similarity or equivalence are the same for established. The boundaries that are calibrated for less serious diseases
all marketed small synthetic drugs. may be a bit more flexible in oncology, just as maximum tolerated dose
A parallel set of procedures has evolved for biologicals and other is a relative term that depends on the medical scenario. Phase I, phase
complex drug substances. The expectations for therapeutic and toxic II, and phase III are long-established terms. Phase IV was created by
effects of biologicals are somewhat different than for small synthetic a formal change in FDA regulations. Phase 0 is a less formal term
compounds, and the approval paths reflect these differences. Most used by some investigators to describe early explorations to seek
important, biologicals are manufactured by living organisms, and the information before deciding to launch full-scale development.
precise characterization of the active ingredient is more difficult to Working backward, phase III clinical studies are the largest and
replicate. The overall program creates a category of drugs known as most expensive periods of clinical development and have the highest
biosimilars. Europe already has 20 biosimilars.23 profile in the oncology community. The results of these studies are
The FDA approved the first biosimilar for the United States in carefully followed and are prominently featured at large national
2015. The important feature for the FDA approach is the designa- meetings. These trials are designated as potentially “practice changing”
tion of two categories: biosimilars and interchangeable biological because of their potential for an immediate improvement in the standard
products. Both categories require extensive preclinical data, and of care for the specific stage and type of cancer. The pivotal hallmark
sufficient clinical data to ensure no meaningful differences from the of phase III trials is a randomized comparison of two or more treatment
reference product. Interchangeable products have additional informa- groups. These trials are undertaken based on preliminary evidence of
tion that demonstrates that patients can switch between the reference anticancer activity, and the key question is simply stated: How does
product and its interchangeable counterpart, with similar tolerance the new therapy compare with established therapy?
and activity. When phase III trials include an investigational agent, the data
In terms of dispensing, the name of the biosimilar product is from these trials are the ordinary basis for the regulatory decision to
specifically written by the prescriber. For products that are interchange- permit an indication for marketing in the population studied in the
able, pharmacists can substitute the interchangeable product when trial. Thus these trials are sponsored by the commercial organizations
the prescription is written for the reference product. that will be seeking marketing approval. If the phase III trial is a
comparison of groups for which all of the drugs are off-patent, then
New Indications and Repurposing (Repositioning) the NCI is usually the only organization with the resources and the
mission to support such studies. Nonprofit organizations could also
Terms such as repurposing and repositioning have become commonplace serve in this role, but they tend to support earlier stages of clinical
as researchers and companies seek new uses for approved drugs. Because studies because of the cost of phase III trials.
the safety of the drug has already been established and the supply of Phase II clinical trials are the setting for determining if an agent
clinical-grade drug is readily available, the road to evaluation in patients or combination of agents has sufficient activity in well-defined groups
is accelerated (Box 25.5). Initial approval for a drug is not the end of patients who have no established treatments.24 In the majority of
of its clinical development. phase II studies, there is only a single treatment group, but randomized
The simplest version of this concept is to search within the same designs of two or more groups have been increasingly used. The goal
therapeutic area. Additional uses for cancer drugs are often actively is to determine if the activity of the agent or combination is sufficient
sought. Once again, imatinib sets the standard. In this case, the approved to move into much larger phase III trials with comparison to standard
indications were extended from CML to GI stromal tumor, and again treatments. Unfortunately, areas within oncology remain for which
to various rare cancers. no standard treatment has been established. In such cases, phase II
When therapeutic boundaries are crossed, potential therapies could studies can be sufficient for regulatory approval.
move from oncology to other diseases or from other diseases into Phase I clinical studies have traditionally been the first-in-human
oncology. Methotrexate is used in juvenile and rheumatoid arthritis. studies for new agents or for combinations of investigational or approved
Cyclophosphamide is used in both children and adults for nephrotic drugs. In diseases other than cancer, these safety studies are generally
syndrome. Paclitaxel is used as a coating for cardiac stents. In the conducted with healthy volunteers. Until recently, phase I clinical
other direction, cancer researchers have a recurring interest in evaluation studies of cancer drugs were conducted only with patients with cancer
of potential anticancer effects of marketed drugs from outside oncology. because of the potential for acute, severe toxicity, as well as longer-term
Recurring examples include metformin and simvastatin. issues such as carcinogenicity or mutagenicity for many classes of
older cancer drugs. As the biologic properties of cancer drugs have
Clinical Phases of Drug Development evolved, some investigational agents have adverse effect profiles that
are similar to those of agents outside oncology. For compounds
As indicated in Box 25.6, the clinical stages of drug development sponsored by the pharmaceutical industry, there has been a trend
have been divided historically into phases that are related to specific toward use of healthy volunteers in phase I clinical studies for certain
goals. These stages also have regulatory significance, but the boundaries classes of cancer drugs. It is likely that both strategies will coexist,
418 Part I: Science and Clinical Oncology

and the approach will be customized for each investigational agent. The fruits of cancer pharmacology are found in the paths that
Phase I studies in healthy volunteers have a track record of lower guide drugs into clinical use and in the adjustment of therapy in
costs, as well as faster and more reliable recruitment of subjects. patients to balance therapeutic and toxic effects. Improving the con-
On the other hand, the unique aspect for inclusion of patients nections between cancer drug delivery and the quantitative aspects
with cancer in phase I is the enormous value for determination of of the activity and toxicity of cancer drugs is essential for further
molecular effects in tumors via biopsies or imaging as well as signals progress. Cancer drug delivery can only provide therapeutic concentra-
of therapeutic benefit. Although expectations are modest for anticancer tions and avoid toxic concentrations if exploitable differences between
effects during initial studies of new agents, favorable responses can tumors and host tissues have been identified and reliable assays are
accelerate development programs. available at the laboratory-clinic interface.
Phase 0 clinical studies are an approach to first-in-human studies
that extends the preclinical search for specific types of information into WHAT THE FUTURE HOLDS
the clinic. Frequently, the question is whether systemic exposure can
be achieved that is considered essential to activity, or whether a specific Three new regulatory actions related to the marketing of pembrolizumab
degree of target engagement, can be reached.25,26 A small number of have stimulated further reexamination of the drug approval process:
analogs might be compared in a phase 0 study before final selection of
the lead candidate. These and other questions can often be explored 1. FDA approval of pembrolizumab for second-line treatment of
with single doses, or short courses. Some of these same questions can metastatic melanoma was based on objective and durable responses
also be answered during phase I studies. There is a smaller package of in a phase I trial.27 Surely, this action validates the recent efforts
preclinical studies required for phase 0, so the choice may be driven to learn as much as possible, as early as possible.
by degree of commitment to full-scale clinical evaluation. 2. For the initial approval of pembrolizumab, FDA and the sponsor
Phase IV was added to the process for clinical development to worked together to rely on amendments to the initial clinical
permit approval when sufficient demonstration of safety and efficacy protocol from phase I throughout the subsequent clinical develop-
has been established, but before all studies are completed. For example, ment.28 FDA staff indicated that this “seamless” approach to
during the review of the marketing application it could be determined oncology drug development has some disadvantages, and is not
that certain subpopulations were not included in the pivotal studies. appropriate for all development programs. Nonetheless, FDA
In return for faster approval, the sponsor of the drug makes a com- convened several public meetings to create dialog on the potential
mitment to conduct “postmarketing” studies to fill any gaps defined advantages and disadvantages. Every development plan will not
by the FDA. Situations include clinical studies of patients with impaired fit into this box, but the concept of an integrated approach to
organ function, drug-drug interactions, or expanded studies of elderly clinical development is surely boosted by this action.
patients. 3. The FDA issued its first “tissue-agnostic” approval, for the use of
The numeric order of the clinical phases provides a logical progres- pembrolizumab based on its target, DNA mismatch repair and
sion for development but should not be considered as universally microsatellite instability, rather than the tissue site.29 The molecular
linear. As previously noted, a new drug is sometimes approved based interrogation of tumor samples to help determine appropriate
on phase II (or even phase I) studies, and phase III trials are not therapy can be viewed as a direct extension of the historical applica-
conducted. At the other extreme, an unanticipated problem with tion of histopathologic classification to guide recommended therapy.
administration during a phase II investigation might prompt the return
to phase I to improve characterization of doses and schedules. Precedents These developments were not foreseen as recently as 5 years ago,
exist for skipping phase II studies and moving directly from phase I and indicate that the “future” is closer than we imagined. Much work
to phase III. Evidence of activity in phase I may seem so compelling remains to be done in the area of acquisition and handling of biopsy
that it warrants immediate comparison with standard of care. This specimens, as well as systematic evaluation using validated methods.
strategy has a high risk in terms of the resources invested in a compound Fortunately, many complementary options are also evolving, such as
without much understanding of the probable activity. Of course, the the collection and analysis of circulating tumor cells, as well as molecular
potential exists for a high reward in terms of bringing new treatments imaging probes that can provide real-time confirmation of target
to patients, as well as financially. Of course, this strategy is feasible engagement in situ.
only if the phase I study is conducted in patients with cancer. All models have limitations, but we certainly need improvement
in preclinical models of cancer to improve the clinical success rate for
new drugs. It is likely that the continued evolution of patient-derived
CANCER PHARMACOLOGY ACROSS xenografts and genetically engineered mice will provide part of the
THE DRUG DISCOVERY AND progress.
DEVELOPMENT PROCESS Another potential model is emerging from investigation and
treatment of spontaneous tumors in dogs.30 Some tumor types have
Applications of the principles of cancer pharmacology are embedded very similar pathologic appearance in dogs and humans. The ability
in the continuum from early drug discovery through preclinical to use a larger mammal with an intact immune system may have some
development, and then bridging to the preapproval stages of clinical advantages in the current era of immune-oncology. There is already
development. a well-organized set of veterinary researchers and clinicians, and the
During the development phases for a cancer drug, it is essential connections to human oncology are growing.
to monitor the relative drug delivery, as assessed by systemic concentra- Finally, with the large and increasing number of drugs available
tion. When comparing activity studies in murine models, safety studies for use (see Appendix),31 we must recognize that it is not feasible to
in various mammals, or early clinical studies, plasma concentrations remember all the details about each one. The widespread availability
can be monitored. Variation across animal models might signal and use of online resources, including FDA databases, supplements
important interspecies differences in factors related to either PK (e.g., traditional textbooks. Assembling the key pieces of data is only the
metabolism) or PD (e.g., inherent host cell sensitivity). first step. Professional interpretation and judgment remain as central
In the same manner, when making comparisons between observa- features.
tions of activity in cell culture versus concentrations that can be safely
achieved in vivo, it is prudent to consider reasons for any discrepancies.
Skepticism is particularly appropriate if “interesting” activity is observed The complete reference list is available online at
in vitro at concentrations much higher than are achievable in vivo. ExpertConsult.com.
Cancer Pharmacology  •  CHAPTER 25 419

REFERENCES
1. Yao S, Barlow WE, Albain KS, et al. Gene polymor- 10. White DL, Dang P, Engler J, et al. Functional activ- 21. Groot Koerkamp B, Sadot E, Kemeny NE, et al.
phisms in cyclophosphamide metabolism pathway, ity of the OCT-1 protein is predictive of long-term Perioperative hepatic arterial infusion pump che-
treatment-related toxicity, and disease-free survival outcome in patients with chronic-phase chronic motherapy is associated with longer survival after
in SWOG 8897 clinical trial for breast cancer. Clin myeloid leukemia treated with imatinib. J Clin Oncol. resection of colorectal liver metastases: a propensity
Cancer Res. 2010;16:6169–6176. 2010;28:2761–2767. score analysis. J Clin Oncol. 2017;35:1938–1944.
2. Abraham JE, Guo Q, Dorling L, et al. Replication 11. Umans RA, Taylor MR. Zerbrafish as a model to 22. Alberts DS, Markman M, Armstrong D, et al.
of genetic polymorphisms reported to be associated study drug transporters at the blood-brain barrier. Intraperitoneal therapy for stage III ovarian cancer:
with taxane-related sensory neuropathy in patients Clin Pharmacol Ther. 2012;92:567–570. a therapy whose time has come! J Clin Oncol.
with early breast cancer treated with paclitaxel. Clin 12. Marin D, Bazeos A, Mahon FX, et al. Adherence is 2002;20:3944–3946.
Cancer Res. 2014;20:2466–2475. the critical factor for achieving molecular responses in 23. Camacho LH. Current status of biosimilars in
3. Hertz DL, Owzar K, Lessans S, et al. Pharmaco- patients with chronic myeloid leukemia who achieve oncology. Drugs. 2017;77:985–997.
genetic discovery in CALGB (Alliance) 90401 and complete cytogenetic responses on imatinib. J Clin 24. Sharma MR, Stadler WM, Ratain MJ. Randomized
mechanistic validation of a VAC14 polymorphism Oncol. 2010;28:2381–2388. phase II trials: a long-term investment with promising
that increases risk of docetaxel-induced neuropathy. 13. Partridge AH, Archer L, Kornblith AB, et al. Adher- returns. J Natl Cancer Inst. 2011;103:1093–1100.
Clin Cancer Res. 2016;22:4890–4900. ence and persistence with oral adjuvant chemotherapy 25. Murgo AJ, Kummar S, Rubinstein L, et al. Design-
4. Kummar S, Chen HX, Wright J, et al. Utilizing in older women with early-stage breast cancer in ing phase 0 cancer clinical trials. Clin Cancer Res.
targeted cancer therapeutic agents in combination: CALGB 49907: adherence companion study 60104. 2008;14:3675–3682.
novel approaches and urgent requirements. Nat Rev J Clin Oncol. 2010;28:2418–2422. 26. Kummar S, Kinders R, Gutierrez ME, et al. Phase
Drug Discov. 2010;9:843–856. 14. Sun J, Ilich AI, Kim CA, et al. Concomitant admin- 0 clinical trial of the poly (ADP-ribose) polymerase
5. Holbeck SL, Camalier R, Crowell JA, et al. The istration of proton pump inhibitors and capecitabine inhibitor ABT-888 in patients with advanced
National Cancer Institute ALMANAC: A comprehen- is associated with increased recurrence risk in early malignancies. J Clin Oncol. 2009;27:2705–2711.
sive screening resource for the detection of anticancer stage colorectal cancer patients. Clin Colorectal Cancer. 27. Chuk MK, Chang JT, Theoret MR, et al. FDA
drug pairs with enhanced therapeutic activity. Cancer 2016;15:257–263. approval summary: accelerated approval of pem-
Res. 2017;77:3564–3576. 15. Chu MP, Hecht JR, Slamon D, et al. Association of brolizumab for second-line treatment of metastatic
6. Niederhuber JE. Future of positron-emission proton pump inhibitors and capecitabine efficacy in melanoma. Clin Cancer Res. 2017 Feb 24. pii:
tomography in oncology. Ann Surg. 1998;227:324– advanced gastroesophageal cancer: secondary analysis clincanres.0663.2016. E-Pub ahead of print (as of
325. of the TRIO-013/LOGiC randomized clinical trial. 11-July-2017).
7. Liu G, Jeraj R, Vanderhoek M, et al. Pharmaco- JAMA Oncol. 2017;3:767–773. 28. Prowell TM, Theoret MR, Pazdur R. Seamless
dynamic study using FLT PET/CT in patients 16. Ratain MJ, Cohen EE. The value meal: how to save oncology-drug development. N Engl J Med.
with renal cell cancer and other solid malignancies $1,700 per month or more on lapatinib. J Clin Oncol. 2016;374:2001–2003.
treated with sunitinib malate. Clin Cancer Res. 2007;25:3397–3398. 29. Poh A. First tissue-agnostic drug approval issued.
2011;17:7634–7644. 17. Ratain MJ. Flushing oral oncology drugs down the Cancer Discov. 2017;7:656.
8. Hartford CM, Desai AA, Janisch L, et al. A phase toilet. J Clin Oncol. 2011;29:3958–3959. 30. LeBlanc AK, Breen M, Choyke P, et al. Perspec-
I trial to determine the safety, tolerability, and 18. Dolton MJ, Roufogalis BD, McLachlan AJ. Fruit tives from man’s best friend: National Academy of
maximum tolerated dose of deforolimus in patients juices as perpetrators of drug interactions: the role of Medicine’s Workshop on Comparative Oncology. Sci
with advanced malignancies. Clin Cancer Res. polypeptides. Clin Pharmacol Ther. 2012;92:622–630. Transl Med. 2016;8:324ps5.
2009;15:1428–1434. 19. Morrissey KM, Wen CC, Johns SJ, et al. The UCSF- 31. Freter C, Perry W. Systemic therapy. In: Abeloff MD,
9. Egorin MJ, Van Echo DA, Tipping SJ, et al. FDA TransPortal: a public drug transporter database. Armitage JO, Niederhuber JE, et al, eds. Abeloff ’s
Pharmacokinetics and dosage reduction of cis- Clin Pharmacol Ther. 2012;92:545–546. Clinical Oncology. 4th ed. Philadelphia: Churchill
diammine(1,1-cyclobutanedicarboxylato) platinum 20. Niederhuber JE, Ensminger W, Gyves J, et al. Regional Livingston; 2008.
in patients with impaired renal function. Cancer chemotherapy of colorectal cancer metastatic to the
Res. 1984;44:5432–5438. liver. Cancer. 1984;53:1336–1343.
Cancer Pharmacology  •  CHAPTER 25 419.e1
419.e1

Appendixa
Drug Class and/or Pharmacokinetics Dosage and
Drug Name Mechanism and Metabolism Toxicity Indications Administration
Abiraterone acetate (Zytiga) Inhibits androgen Do not take with food; Joint swelling or Metastatic prostate 1000 mg QD PO with
biosynthesis inhibits CYP2D6 discomfort, edema; cancer; castration 5 mg prednisone BID
monitor liver enzymes resistant with prior
use of docetaxel
Ado-trastuzumab emtansine Antibody-drug Avoid strong CYP3A Hepatotoxic, cardiotoxic, Her2+ BC metastasis, 3.6 mg/kg IV every
(Kadcyla) conjugate inhibitor fetal harm prior trastuzumab and 3 wk
a taxane
Afatinib (Gilotrif ) Kinase inhibitor Reduce dose if PgP Diarrhea, severe First-line NSLC 40 mg QD PO
inhibitor used cutaneous reaction metastasis with EGFR
exon 19 del or exon
21 sub per FDA test
Aflibercept, ziv-aflibercept Fusion protein No studies for renal, Hemorrhage, GI Metastatic colorectal 4 mg/kg via 1-h
(Zaltrap) binding VEGF and hepatic, or drug-drug perforation, wound cancer resistant or intravenous infusion
related ligands interactions healing problems, fistula, progressed while (not bolus) every
hypertension, arterial taking oxaliplatin 2 wk
thrombosis proteinuria
Alectinib (Alecensa) Kinase inhibitor With food Hepatotoxic—monitor; ALK+ metastasis, 600 mg BID PO
pneumonitis, bradycardia, NSCLC, failed or
myalgia, fetal harm, intolerant to crizotinib
fatigue, constipation,
edema
Alemtuzumab (Campath) Monoclonal antibody Long half-life; no dose Infusion reactions; B-cell CLL Intravenous infusion
against CD52 present adjustments based on prolonged with gradual dose
on B cells and other age myelosuppression, escalation up to
immune cells; hemolytic anemia, 30 mg/kg 3 times
produces ADCC and immune per week with
following binding thrombocytopenia; appropriate
immunosuppression and premedication
infection; cardiac
dysrhythmias
Altretamine (Hexalen), Alkylating agent Well absorbed by Myelosuppression is dose Refractory ovarian 4–12 mg/kg/day in
hexamethylmelamine, HMM mouth, metabolized limiting; leukopenia, carcinoma divided doses for
in the liver; half-life thrombocytopenia, 3–6 wk or 50 mg/m2/
4–13 h; metabolism nausea, vomiting, day for 14 days each
may be slowed by neurologic toxicity are cycle; higher doses
cimetidine or common, including have been used
enhanced by confusion, lethargy,
phenobarbital weakness, and sensory
changes
Amifostine (Ethyol), WR-2721, Cytoprotectant; After intravenous Transient hypotension is Pretreatment with 740 mg/m2
ethiofos free-radical infusion, the drug is dose limiting; nausea, cisplatin; useful as intravenous infusion
scavenger metabolized to a thiol vomiting, and somnolence a bone marrow, over 15 min given
metabolite, which is are common; sneezing, kidney, and nerve 15–30 min before
responsible for its hypocalcemia, and cytoprotectant; useful the cytotoxic agent
beneficial activity flushing can be seen with other alkylators or radiation; lower
as well; also FDA doses and
approved as a subcutaneous
radiation protectant administration have
to reduce xerostomia also been used
Anastrozole (Arimidex) Nonsteroidal Well absorbed from Very well tolerated; Indications: As 1 mg PO every day;
aromatase inhibitor; GI tract, maximum asthenia, headache, and adjuvant therapy of higher doses are no
blocks estrogen plasma levels within hot flashes occur in fewer BC and for treatment more effective
production 2 h; half-life is 50 h; no than 15% of women; of postmenopausal
selectively adjustments needed diarrhea, abdominal pain, women with breast
for abnormal function anorexia, nausea, and carcinoma that has
of these organs vomiting occur in 10% or progressed during
because of the wide fewer; thrombophlebitis treatment with
therapeutic index has been reported tamoxifen

Continued
419.e2 Part I: Science and Clinical Oncology

Appendixa—cont’d
Drug Class and/or Pharmacokinetics Dosage and
Drug Name Mechanism and Metabolism Toxicity Indications Administration
Arsenic trioxide (Trisenox) Novel arsenical Half-life of this Differentiation syndrome is Relapsed acute 0.15 mg/kg/day in
differentiating agent compound is dose limiting—includes promyelocytic 100–250 mL of D5W
unknown; it is leukocytosis, fever, leukemia until remission, not
methylated in the liver dyspnea, chest pain, to exceed 60 doses,
and eliminated in the tachycardia, hypoxia, then up to 25 doses
urine sometimes death; over 5 wk for
corticosteroids seem to consolidation
benefit this syndrome; QT starting 3–6 wk after
prolongation is common; achievement of
also rash, pruritus, remission
headache, arthralgias,
anxiety, bleeding, nausea,
vomiting; liver and renal
toxicity uncommon
L-Asparaginase (Elspar), Naturally occurring After intravenous or Hypersensitivity can be ALL; also used in AML, After a 2-unit
colaspase enzyme derived from intramuscular life-threatening, late-stage CML, CLL, intradermal test
Escherichia coli or injection, the drug is necessitating anaphylaxis and non-Hodgkin dose, an
Erwinia carotovora metabolized precautions and 2-unit lymphomas intramuscular dose
that cleaves intravascularly by test dose; coagulopathy is of 6000–10,000 IU/
asparagine, an proteolysis; common and requires m2 every 3 days for 9
essential amino acid elimination half-life of monitoring; nausea, doses, or 1000 IU/kg/
required by rapidly 8–30 h vomiting, abdominal day IV over 30 min
proliferating cells cramps, anorexia, elevated for 10 days, has been
liver function test results, used
transient renal
insufficiency are common;
lethargy, somnolence,
fatigue, depression,
confusion, pancreatitis,
and fever are seen
Pegaspargase (Oncaspar) Naturally occurring When given by Less immunogenic that ALL, and like 2,500 IU/m2 IM every
enzyme, covalently intramuscular non-PEGylated form, but asparaginase, it is also 14 days with other
linked to injection, it has an hypersensitivity and used for other chemotherapy
polyethylene glycol elimination half-life of anaphylaxis can still occur; leukemias and agents for induction
to reduce approximately 5 days; toxicities similar to those non-Hodgkin or maintenance
immunogenicity, clearance is not of non-PEGylated forms lymphomas
slow metabolism, dependent on renal or including elevated liver
and prolong half-life; hepatic function enzymes, coagulopathy,
the enzyme cleaves hypercholesterolemia,
asparagine, an pancreatitis,
essential amino acid hyperglycemia, fever,
required by rapidly chills, anorexia, lethargy,
proliferating cells confusion, headache,
seizures, and azotemia
Asparaginase Erwinia Substitute for the Not characterized Severe hypersensitivity ALL 25,000 IU IM 3 times
chrysanthemi (Erwinaze) naturally occurring reactions, including per wk for 6 doses
enzyme anaphylaxis per cycle
Atezolizumab (Tecentriq) PD-L1 blocking Potential effect of Pneumonitis, hepatitis, Locally advanced or 1200 mg IV over
antibody renal or hepatic colitis, endocrinopathies, metastatic urothelial 24 h; every 3 wk
impairment unknown myasthenia gravis, ocular cancer following
toxicity, pancreatitis, platinum
infusion reaction, fetal chemotherapy
harm
Avelumab (Bavencio) PD-L1 blocking Premedicate with Fatigue, musculoskeletal Merkel cell carcinoma 10 mg/kg IV over
antibody acetaminophen, effects, diarrhea, infusion metastasis, in adults or 60 min; every 3 wk
antihistamine reaction, rash, nausea, pediatric patients
peripheral edema 12 yr and older
Axitinib (Inlyta) Avoid strong CYP3A Decrease dose 50% Diarrhea, hypertension, Advanced RCC after 5 mg BID PO
inhibitors or reduce for CYP3A inhibitors, fatigue, nausea one treatment failure
dose renal or hepatic
impairment
Cancer Pharmacology  •  CHAPTER 25 419.e3
419.e3

Appendixa—cont’d
Drug Class and/or Pharmacokinetics Dosage and
Drug Name Mechanism and Metabolism Toxicity Indications Administration
Azacitidine (Vidaza) Antimetabolite; Not orally bioavailable; Myelosuppression is dose MDS 75–100 mg/m2 for 7
induces metabolized by the limiting; leukopenia, days; repeat every
hypomethylation of liver and excreted in thrombocytopenia, and 4 wk for 4–6 cycles
DNA, either inducing urine; elimination transient elevation of liver
apoptosis or half-life of 4 h function test results are
restoring normal common; nausea and
function; at higher vomiting and abdominal
doses, acts as a pain are common
cytidine analogue
Bacillus Calmette-Guérin (TICE Immunostimulant, BCG is a live, Urinary symptoms Intravesical instillation 81 mg per treatment,
BCG, TheraCys), BCG vaccine; induces a attenuated bacterial predominate, including for noninvasive in 53 mL total
cellular immune culture; it does not dysuria, hematuria, bladder cancer after volume, given once
response at the site enter the body in hesitancy, urgency, removal of papillary weekly for 6 doses
of instillation viable form; it has frequency, and secondary tumors; also used for and then at 3, 6, 12,
no detectable infection; other toxicities some experimental 18, and 24 mo after
pharmacokinetic fate; include fever, chills, vaccine programs as the induction
in rare cases a clinical malaise, myalgias and an adjuvant to the
infection can result arthralgias, anorexia, vaccine
from treatment, nausea, vomiting, and
indicating invasion of anemia; clinical
the body at site of mycobacterial infection is
administration into rare, usually seen only in
systemic circulation immunocompromised
patients
Belinostat (Beleodaq) Inhibition of HDAC Avoid strong UGT1A1 Nausea, fatigue, pyrexia, Relapsed or refractory 1000 mg/m2 IV over
inhibitors anemia, vomiting, T-cell lymphoma 30 min, days 1–5
decreased platelets and every 21 days
WBCs, infection,
hepatotoxicity, tumor lysis
syndrome, fetal harm
Bendamustine (Treanda) Alkylating agent Reduce dose for Nausea, pyrexia, vomiting, CLL and B-cell CLL: 100 mg/m2 over
hematologic toxicity hematologic non-Hodgkin 60 min, day 1, 2 of
abnormalities lymphoma 28-day cycle; B-cell:
120 mg/m2 over
30 min on day 1, 2 of
21-day cycle
Bevacizumab (Avastin) Recombinant Administered by Asthenia, pain, nausea, Metastatic colorectal 5–15 mg/kg/day IV
humanized intravenous infusion;vomiting, diarrhea, cancer and NSCLC; every 3 wk
monoclonal antibody half-life is 20 days; the
anorexia, stomatitis, RCC
that binds to all fate of parent drug dermatitis, hypertension,
forms of VEGF, and metabolites is proteinuria; infusion-
preventing binding unknown related reactions rare;
to its receptors hemoptysis, hemorrhage,
delayed wound healing, GI
perforations; increased
risk of thromboembolic
events can be severe or
fatal
Bexarotene (Targretin) Synthetic retinoid, Good oral Hyperlipidemia is dose CTCL (mycosis 300 mg/kg/day PO,
differentiating agent bioavailability limiting and should be fungoides) refractory dose adjusted for
increased by a monitored during therapy to at least one prior toxicity
high-fat meal; and treated as therapy
metabolized in the appropriate; pruritus,
liver to oxidative leukopenia, diarrhea,
metabolites by fatigue, headache, and
cytochrome P450 3A4, liver function test result
glucuronidated, elevation can also be dose
eliminated in bile limiting; rash, edema,
fever, chills, and nausea
are uncommon; excessive
bleeding and back or
abdominal pain are rare

Continued
419.e4 Part I: Science and Clinical Oncology

Appendixa—cont’d
Drug Class and/or Pharmacokinetics Dosage and
Drug Name Mechanism and Metabolism Toxicity Indications Administration
Bicalutamide (Casodex) Nonsteroidal Well absorbed orally; Constitutional symptoms Stage D2 prostate 50 mg PO daily, in
antiandrogen highly protein bound; dominate: hot flashes, cancer, in combination combination with an
converted to inactive decreased libido, with an LHRH agonist LHRH agonist agent
metabolites in liver depression, weight gain, agent
via oxidation and edema, gynecomastia,
glucuronidation; early disease-site pain
half-life of several (flare reaction);
days constipation, nausea,
vomiting, anorexia,
diarrhea, dizziness
uncommon; dyspnea,
anemia, fever, and rashes
rare
Bleomycin (Blenoxane) Antitumor antibiotic; Elimination half-life of Pulmonary toxicity, Germ cell tumors, Observe 1–6 h; after
causes DNA strand 3–5 h; incompletely including reversible and Hodgkin disease, and 2-unit intravenous
breaks directly in metabolized by irreversible fibrosis, is dose squamous cell test dose over
normal and intracellular limiting; other common cancers; used off-label15 min, the full dose
neoplastic cells aminopeptidases; toxicities include fever, for melanoma, ovarian can be given; usual
excreted in the kidney chills, rash, exfoliation, cancer, and Kaposi dose is 10–20 units/
as unchanged drug anorexia; nausea, sarcoma; also used as m2 IV, IM, or SC 1–2
and metabolites vomiting, a sclerosing agent for times per wk, or
myelosuppression, malignant pleural or 15–20 units/m2/day
anaphylaxis, mucositis rare pericardial effusions as a continuous
infusion over 3–7
days; as a sclerosing
agent, 60 units
generally used
Blinatumomab (Blincyto) CD19-directed Premedicate with Severe cytokine release Relapsed or refractory Induction: 28 days
antibody to target T dexamethasone syndrome, severe B-cell ALL in adults via continuous
cells neurotoxicity and children intravenous infusion;
variable dosage rates,
see product labeling
Bortezomib (Velcade) Proteasome inhibitor Metabolized in the Cardiovascular; peripheral Multiple myeloma; 1.3 mg/m2 IV or SC
liver by CYP2C19 and neuropathy; skin rash; mantle cell lymphoma days 1, 4, 8, 11, 22, 25,
CYP3A4; reduce dose nausea and diarrhea 29, and 32 of a
for hepatic 42-day treatment
dysfunction cycle for 4 initial
cycles
Bosutinib (Bosulif ) Kinase inhibitor Take with food; reduce GI toxicity should Ph+ CML, resistant or 500 mg/day PO
to 200 mg if hepatic be monitored; intolerant to prior
impaired; CYP3A myelosuppressive therapy
substrate
Brentuximab vedotin CD30-directed Monitor patients Progressive multifocal Hodgkin lymphoma 1.8 mg/kg
(Adcetris) antibody conjugate taking CYP3A inducers leukoencephalopathy; and anaplastic large intravenous infusion
or inhibitors peripheral neuropathy; cell lymphoma over 30 min every
neutropenia 3 wk
Brigatinib (Alunbrig) Kinase inhibitor With or without food Pneumonitis, ALK+ metastasis, 90 mg QD × 7 days
hypertension, bradycardia, NSCLC, failed or PO; increase to
visual effects, enzyme intolerant to crizotinib 180 mg/day if
elevations, hyperglycemia, tolerated
nausea, diarrhea, fatigue,
cough, headache
Buserelin (Suprefact), HOE 766 LHRH agonist; shuts Intravascular and Flare reactions, which can Prostatic cancer 500 µg SC TID for the
off LH and FSH extravascular be prevented; castration first wk, then 200 µg/
secretion, producing proteolysis symptoms such as hot day, or intranasally
chemical castration flashes and decreased 800 µg TID followed
libido common; other by 400 µg TID
nonspecific symptoms
include headache, nausea,
vomiting, diarrhea,
constipation, weakness
Cancer Pharmacology  •  CHAPTER 25 419.e5
419.e5

Appendixa—cont’d
Drug Class and/or Pharmacokinetics Dosage and
Drug Name Mechanism and Metabolism Toxicity Indications Administration
Busulfan (Myleran Oral), Alkylating agent Excellent oral Myelosuppression partly PO: palliative 4–8 mg/day PO
(Busulfex Injection), BSF bioavailability, with chronic and cumulative, is treatment of chronic initially, decreasing
peak levels in serum dose limiting; other myelogenous based on blood
occurring at about common toxicities: (myeloid, myelocytic, counts, with 1–3 mg
1 h; elimination nausea, vomiting, granulocytic) daily used for
half-life of 2.5 h; anorexia, mucositis, leukemia; IV: in maintenance;
metabolized partially hyperpigmentation, combination with intravenous
in liver; parent drug elevated liver function cyclophosphamide as conditioning dose
and metabolites tests (or venoocclusive a conditioning 0.8 mg/kg every 6 h
excreted in the urine; disease of the liver at regimen before for 4 days
also available IV transplant doses); allogeneic
neurologic toxicity, hematopoietic
including blurred vision, progenitor cell
dizziness, and confusion, transplantation for
and ILD are less common CML
Cabazitaxel (Jevtana) Microtubule inhibitor Caution for patients Neutropenia and Metastatic hormone 25 mg/m2 every 3 wk
taking strong CYP3A hypersensitivity refractory prostate 1-h intravenous
inducers or inhibitors cancer when infusion with oral
docetaxel fails prednisone 10 mg
daily
Cabozantinib Inhibits many 55-h half-life; take Perforations, fistulas, and Metastatic medullary 140 mg daily
receptor tyrosine without food hemorrhage thyroid cancer
kinases
Capecitabine (Xeloda) Oral antimetabolite Readily absorbed, Myelosuppression and Metastatic BC and 1250 mg/m2 every 12 
prodrug metabolized in vivo to palmar-plantar metastatic colorectal
h for 14 days every
fluorouracil by erythrodysesthesia are cancer; used also in
21 days; dose
carboxylesterase, dose limiting; diarrhea, head and neck reductions are often
cytidine deaminase, fatigue, stomatitis, and squamous cell cancer
required; treatment
thymidine hyperbilirubinemia are delays are sometimes
phosphorylase uncommon; nausea, required; other doses
vomiting, and rash are and schedules have
rare been used
Carboplatin (Paraplatin), carbo, Atypical alkylator; Rapidly cleared from Hemorrhage; Ovarian cancer and Dosage based on
CBDCA produces intrastrand the bloodstream after thrombocytopenia used extensively in square meters or
and interstrand intravenous infusion, testicular cancer, via formulas that
cross-links in DNA via with a terminal squamous cell cancers take into account
association bonds half-life of 2.5 h; it is of the head and neck renal function and
with the platinum cleared largely as and cervix, and lung desired level of
molecule, leading to unchanged drug by cancer thrombocytopenia
DNA strand breakage the kidneys (such as Calvert’s
during replication formula); typical
doses with normal
renal function are in
300–500 mg/m2
range as intravenous
infusion
Carfilzomib (Kyprolis) Proteosome inhibitor Drug-drug Fatigue, anemia, nausea, Multiple myeloma 20 mg/m2/day IV
interactions unlikely; thrombocytopenia, over 2–10 min, days
no adjustment for dyspnea, diarrhea, and 1, 2, 8, 9, 15, 16; rest
renal impairment; no pyrexia; monitor for period (days 17–28);
experience with cardiac, pulmonary, tumor 27 mg/m2/day other
hepatic impairment lysis, infusion reactions, cycles if tolerated
thrombocytopenia

Continued
419.e6 Part I: Science and Clinical Oncology

Appendixa—cont’d
Drug Class and/or Pharmacokinetics Dosage and
Drug Name Mechanism and Metabolism Toxicity Indications Administration
Carmustine (BiCNU), BCNU, Alkylator agent in the After an intravenous Myelosuppression slow in Melanoma, stomach Single-agent dose is
bischloronitrosourea nitrosourea class; cell infusion, the drug is onset, cumulative and cancer, colon cancer, 150–200 mg/m2
cycle independent rapidly taken up by dose limiting; nausea, and liver cancer; FDA every 6 wk; for
mechanism tissues, including the vomiting common, approved for brain transplant, the dose
CNS; extensively can be severe; tumors, multiple is as high as 600 mg/
metabolized in the hyperpigmentation, renal myeloma, Hodgkin m2, along with other
liver; the serum toxicity can occur; ILD disease, lymphoma; drugs
half-life is only with fibrosis rare but can also used for BC
15–20 min occur with any dose;
transplant doses can
cause severe liver toxicity
and more frequent lung
toxicity
Carmustine impregnated Novel delivery More than 70% of the None Patients with newly Up to 8 wafers are
wafer (Gliadel), polifeprosan 20 mechanism for copolymer degrades diagnosed high-grade placed in the
with carmustine implant classic nitrosourea by 3 wk; minimal malignant glioma as resection cavity at
alkylating agent systemic exposure to an adjunct to surgery the time of
carmustine and radiation; also, craniotomy and
patients with operative resection
recurrent
glioblastoma
multiforme as an
adjunct to surgery
Ceritinib (Zykadia) Kinase inhibitor No food within 2 h Severe GI toxicity: ALK+ metastasis, 750 mg daily PO
diarrhea, nausea, hepatic NSCLC, failed or
enzymes, abdominal pain, intolerant to crizotinib
fatigue, lesser appetite,
constipation
Cetuximab (Erbitux) Recombinant Metabolism poorly Infusion reaction with Metastatic colorectal 400 mg/m2 IV,
humanized understood; half-life rapid-onset dyspnea, fever, cancer, head and neck followed by 250 mg/
monoclonal antibody, 5–7 days with minimal chills, urticaria, flushing, cancer in combination m2 weekly
targeting EGFR; clearance by kidneys angioedema, hypotension with radiation
competitively inhibits or liver is seen in 40%–50%
growth factor of patients; acneiform
binding; inhibits rash is common;
autophosphorylation constitutional symptoms;
and cell signaling hypomagnesemia; ILD
rare
Chlorambucil (Leukeran) Alkylating agent; cell Excellent oral Myelosuppression dose CLL and low-grade 16 mg/m2/day for 5
cycle–independent bioavailability; limiting, universal, lymphomas; also used days every 4 wk, or
maximum plasma can be cumulative; for Waldenström 0.4 mg/kg every
level at 1 h; half-life of nausea, vomiting, diarrhea macroglobulinemia, 2–4 wk, or
1–2 h; extensively mild and uncommon; multiple myeloma, 0.1–0.2 mg/kg/day
metabolized in liver to sterility, alopecia minor hairy cell leukemia, for 3–6 wk
active and inactive occurrence; pulmonary and rarely in some
metabolites fibrosis and neurologic solid tumors
adverse effects quite rare
Cancer Pharmacology  •  CHAPTER 25 419.e7
419.e7

Appendixa—cont’d
Drug Class and/or Pharmacokinetics Dosage and
Drug Name Mechanism and Metabolism Toxicity Indications Administration
Cisplatin (Platinol)—cDDP, Atypical alkylator; Adequate renal Nephrotoxicity dose Used for almost every Cisplatin can be
DDP, cisplatinum, cis- produces intrastrand perfusion and urine limiting for single dose class of solid tumor given all in one
diamminedichloroplatinum (II) and interstrand output are critical for Neurotoxicity, especially and lymphoma; FDA intravenous infusion
cross-links in DNA via minimizing renal painful peripheral approved for testicular or daily as an
association bonds toxicity; therefore neuropathy, dose limiting and ovarian cancer intravenous infusion
with the platinum prehydration and for cumulative doses and transitional cell for several days, (the
molecule, leading to adequate Mild myelosuppression carcinoma latter method is
DNA strand breakage posttreatment Nausea, vomiting somewhat better
during replication hydration are used, common but manageable; tolerated); total dose
usually with normal Anorexia, diarrhea per cycle ranges
saline solution with or common; cumulative from 80–160 mg/m2;
without mannitol, ototoxicity also common; continuous infusion
potassium, and chronic renal magnesium can also be used;
magnesium and potassium wasting dose should be
common, sometimes reduced for
not reversible creatinine clearance
Elevated liver below 60 mL/min
transaminases seen, but along with the
alopecia or cardiac cisplatin; cisplatin
conduction abnormalities 100–200 mg/m2 is
rare also used
intraperitoneally for
ovarian cancer
Cladribine (Leustatin) Purine Dose adjusted for Fatigue, headache; skin AML; CLL; mantle cell IV, dosage regimens
antimetabolite; renal dysfunction rash; nausea; bone lymphoma are disease specific
activated by marrow suppression;
deoxycytidine kinase infection; fever
and incorporated
into DNA, resulting in
DNA strand breaks
Clofarabine (Clofarex, Clolar) Purine Eliminated by the Blood count suppression; ALL in adults at 52 mg/m2, 2-hour IV
antimetabolite; kidney; clearance rash; capillary leak relapse; refractory infusion; QDx5;
inhibits decreased as syndrome; tachycardia adult AML in patients 28-day cycle
ribonucleotide creatinine clearance is and hypotension; nausea younger than 70 yr
reductase affecting reduced or vomiting; diarrhea; Relapsed/refractory
DNA synthesis; also headache; altered liver ALL in patients 1-21
alters mitochondrial function tests; infection years old after at least
membranes and fever 2 prior regiments
enhancing apoptosis
Cobimetinib (Cotellic) Inhibits MEK1 and With or without food; Diarrhea, photosensitivity, Unresectable or 60 mg QD PO for 21
MEK2 in the MAPK avoid strong or new primary malignancy, metastatic melanoma days on 28-day cycle
cascade moderate CYP3A hemorrhage, with BRAF mutation,
inducers or inhibitors cardiomyopathy, retinal, combined with
hepatic, rhabdomyolysis vemurafenib
Crizotinib (Xalkori) ALK kinase inhibitor Avoid strong CYP3A Hepatotoxicity; ALK-positive NSCLC 250 mg BID; reduce
inhibitors, inducers, or pneumonitis; QT interval or interrupt based on
substrates prolonged toxicity
Cyclophosphamide Alkylating agent Metabolized to its Myelosuppression; Hodgkin and 500 mg to several
active alkylating immunosuppression; non-Hodgkin grams IV on a variety
species by mixed hemorrhagic cystitis; lymphoma; leukemias; of schedules;
function oxidases in myocarditis; pneumonitis; ovarian cancer; BC; 1–5 mg/kg PO on an
hepatic microsomes secondary malignancies; retinoblastoma; intermittent
including CYP2A6 and infertility; hyponatremia; neuroblastoma schedule
CYP3A4; decreased venoocclusive disease;
activation to nausea and vomiting;
therapeutic species in mucositis
the face of severe
hepatic dysfunction

Continued
419.e8 Part I: Science and Clinical Oncology

Appendixa—cont’d
Drug Class and/or Pharmacokinetics Dosage and
Drug Name Mechanism and Metabolism Toxicity Indications Administration
Cytarabine Antimetabolite that Metabolized in the Myelosuppression; AML in adults and Intravenous; dose
inhibits DNA liver and excreted by immunosuppression; CNS, children and schedule vary by
polymerase after the kidney cardiac, and pulmonary treatment regimen
activation to its toxicity; infection; fever,
nucleotide bone and chest pain, and
triphosphate, is rash within hours of
incorporated into administration (ara-C
DNA and RNA, and syndrome); nausea,
blocks cell cycle diarrhea; mucositis;
progression from G1 hepatic dysfunction
to S phase
Dabrafenib (Tafinlar) Kinase inhibitor Take 1 h before or 2 h Hyperkeratosis, headache, Unresectable or 150 mg PO BID ±
after food; avoid use hemorrhage, pyrexia, metastatic melanoma 2 mg of trametinib
with strong CYP3A arthralgia, papilloma, with BRAF mutation,
or CYP2C8 inhibitors chills, abdominal pain, single-agent or
or /inducers fatigue, rash, peripheral combined with
Sensitive substrates of edema, constipation, trametinib
CYP3A, CYP2C8, myalgia
CYP2C9, CYP2C19 may
lose efficacy
Daratumumab (Darzalex) CD38-directed Premedicate with Infusion reaction, fatigue,
Multiple myeloma 16 mg/kg IV weekly
antibody corticosteroids, nausea, back pain, pyrexia,

You might also like