Psycho-Oncology Psycho-Oncology FOURTH EDITION EDITED BY William S. Breitbart, MD, FAPOS Phyllis N. Butow, BA(Hons), DipEd, MClinPsych, MPH, PhD Paul B. Jacobsen, PhD Wendy W. T. Lam, RN, PhD, FFPH Mark Lazenby, APRN, PhD Matthew J. Loscalzo, MSW, LCSW, FAPOS 1 3 Oxford University Press is a department of the University of Oxford. It furthers the University’s objective of excellence in research, scholarship, and education by publishing worldwide. Oxford is a registered trade mark of Oxford University Press in the UK and certain other countries. Published in the United States of America by Oxford University Press 198 Madison Avenue, New York, NY 10016, United States of America. © Oxford University Press 2021 All rights reserved. No part of this publication may be reproduced, stored in a retrieval system, or transmitted, in any form or by any means, without the prior permission in writing of Oxford University Press, or as expressly permitted by law, by license, or under terms agreed with the appropriate reproduction rights organization. Inquiries concerning reproduction outside the scope of the above should be sent to the Rights Department, Oxford University Press, at the address above. You must not circulate this work in any other form and you must impose this same condition on any acquirer. Library of Congress Cataloging-in-Publication Data Names: Breitbart, William S., 1951– editor. Title: Psycho-oncology / [edited by] William S. Breitbart, Phyllis N. Butow, Paul B. Jacobsen, Wendy W. T. Lam, Mark Lazenby, Matthew J. Loscalzo ; senior editor, William Breitbart. Other titles: Psycho-Oncology (Holland) Description: 4th edition. | New York, NY : Oxford University Press, [2021] | Includes bibliographical references and index. Identifiers: LCCN 2020029603 (print) | LCCN 2020029604 (ebook) | ISBN 9780190097653 (hardback) | ISBN 9780190097677 (epub) | ISBN 9780190097684 Subjects: MESH: Neoplasms—psychology | Risk Factors | Neoplasms—prevention & control | Neoplasms—therapy Classification: LCC RC262 (print) | LCC RC262 (ebook) | NLM QZ 260 | DDC 616.99/40019—dc23 LC record available at https://lccn.loc.gov/2020029603 LC ebook record available at https://lccn.loc.gov/2020029604 DOI: 10.1093/med/9780190097653.001.0001 This material is not intended to be, and should not be considered, a substitute for medical or other professional advice. Treatment for the conditions described in this material is highly dependent on the individual circumstances. And, while this material is designed to offer accurate information with respect to the subject matter covered and to be current as of the time it was written, research and knowledge about medical and health issues is constantly evolving and dose schedules for medications are being revised continually, with new side effects recognized and accounted for regularly. Readers must therefore always check the product information and clinical procedures with the most up-to-date published product information and data sheets provided by the manufacturers and the most recent codes of conduct and safety regulation. The publisher and the authors make no representations or warranties to readers, express or implied, as to the accuracy or completeness of this material. Without limiting the foregoing, the publisher and the authors make no representations or warranties as to the accuracy or efficacy of the drug dosages mentioned in the material. The authors and the publisher do not accept, and expressly disclaim, any responsibility for any liability, loss, or risk that may be claimed or incurred as a consequence of the use and/or application of any of the contents of this material. 9 8 7 6 5 4 3 2 1 Printed by LSC Communications, United States of America Dedication: Jimmie C. Holland, M.D. (1928–2017) Psycho-Oncology, 4th edition is solemnly dedicated to Professor Jimmie C. Holland, MD (1928–2017), internationally recognized as the founder of the field of psycho-oncology. Dr. Holland, who was affectionately known by her first name, “Jimmie,” had a profound global influence on the fields of psycho-oncology, oncology, supportive care, psychiatry, behavioral medicine, and psychosomatic medicine. At the time of her passing, Dr. Holland was the Attending Psychiatrist and Wayne E. Chapman Chair at Memorial Sloan Kettering Cancer Center (MSK) and Professor of Psychiatry, Weill Medical College of Cornell University in New York. In 1977, Jimmie was appointed Chief of the Psychiatry Service in the Department of Neurology at MSK, by Jerome Posner, MD, then Chairman of Neurology at MSK. The Psychiatry Service at MSK was the first such clinical, research, and training service established in any cancer center in the world. In 1996, Dr. Holland was named the inaugural Chairwoman of the Department of Psychiatry and Behavioral Sciences at MSK—again, the first such department created in any cancer center in the U.S. or the world. Dr. Holland had over a 40-year career at MSK. Jimmie created and nurtured the field of psycho-oncology, established its clinical practice, advanced its clinical research agenda, and, through her pioneering efforts, launched the careers of the leaders of a worldwide field who continue to work in what has become a shared mission to emphasize “care” in cancer care. Dr. Holland founded the International Psycho-Oncology Society (IPOS) in 1984 and the American Psychosocial Oncology Society in 1986. Over 25 years ago, Jimmie founded the international journal Psycho-Oncology and coedited the journal for 30 years. Dr. Holland received many awards recognizing her achievements over the course of her career. Some of her notable awards include the Medal of Honor for Clinical Research from the American Cancer Society, the Clinical Research Award from the American Association of Community Cancer Centers, the American Association for Cancer Research Joseph H. Burchenal Clinical Research Award, the Marie Curie Award from the Government of France, the Margaret L. Kripke Legend Award for contributions to the advancement of women in cancer medicine and cancer science from the MD Anderson Cancer Center, the T. J. Martell Foundation 2015 Women of Influence Award, and the Distinguished Alumnus Award from Baylor College of Medicine in 2016. She served as President of the Academy of Psychosomatic Medicine (APM) in 1996 and was the recipient of the APM’s Hackett Lifetime Achievement Award in 1994. She was the inaugural recipient of the Arthur Sutherland Award for Lifetime Achievement from IPOS. This 4th edition of Psycho-Oncology is the first edition of this text that has not been edited by Dr. Holland. In 1989, Dr. Holland edited the Handbook of Psychooncology: Psychological Care of the Patient with Cancer, the first major textbook in our field. This landmark book was notable for several reasons; it established our “new” field, and it was the first use, in a text, of the term “psychooncology” to name our field (thankfully the hyphen was soon added). Psycho-oncology was thus born and named with the publication of this textbook. Subsequently, Dr. Holland edited, with a group of dedicated coeditors, several editions of what became known as the “Bible” of psycho-oncology or, in many circles, the “Holland Textbook of Psycho-oncology.” The textbook Psycho-Oncology was published in 1998 and represented the most comprehensive, multidisciplinary, and international encyclopedia of a field entering its adolescence. The year 2010 saw the publication of the 2nd edition, followed by the 3rd edition in 2015, both published by Oxford University Press in collaboration with IPOS and APOS. Every card-carrying “psycho-oncologist” in over 60 countries with national psycho-oncology societies around the world had to have the latest edition in their library. For many it represented a valued link to Jimmie Holland. The task of editing this 4th edition of Psycho-Oncology without Jimmie’s firm guidance and wise counsel was daunting for all of us, but we were all deeply inspired to do so because of our loving debt to Jimmie. The torch has been passed. Dedication: Ruth McCorkle, PhD, RN, FAAN (1941–2019) In January 1975, a 33-year-old Ruth McCorkle, a newly minted PhD from the University of Iowa and a new assistant professor at the University of Washington, met Jimmie C. Holland at a conference on the behavioral dimensions of cancer that was organized by the National Cancer Institute in San Antonio, Texas. This meeting began a lifelong friendship and collaboration, not least of which was this book. Ruth McCorkle died on August 17, 2019, in her home in Hamden, CT, from cancer. At the time of her death, she was the Florence Schorske Wald Professor of Nursing Emerita at Yale University. From the earliest days of her career, Ruth was interested in the lived experiences of people diagnosed with cancer, including the effects of touch on the seriously ill and how the attachments and goals of patients undergoing treatment for lung cancer—and their families—changed over time. At the University of Washington, she and Jeanne Quint-Benoliel developed the first multidisciplinary cancer unit in which patients and their families would be seen from the time of diagnosis through the dying experience by an interprofessional team. It was on this unit, in the mid 1970’s, that she developed the first scale that measured the distress cancer patients experienced, the Symptom Distress Scale. As a student of history, she learned of how Sir William Osler had taken field notes on his dying experience, in which he wrote that, because he had “no actual pain,” he felt “singularly free from mental distress” as he was dying. In the early 1970’s, when Ruth had gone to London to study with Dame Cicely Saunders at St. Christopher’s Hospice, she was introduced to the British psychiatrist J. M. Hinton and his now justly famous qualitative work on associations between dying patients’ physical and mental distress. From Saunders and Hinton, and from Osler’s field notes, Ruth began to see that patients’ mental distress could be related to their physical symptoms. She thus became interested in the points at which a physical symptom becomes emotionally unbearable. Hence, her scale measured the presence of a symptom as well as how distressed a patient was by it. The development of the Symptom Distress Scale led to her intervention. She developed and tested in 7 National Institutes of Health-funded clinical trials the Standardized Nursing Intervention Protocol, an intervention in which an advanced practice cancer nurse helped patients and families learn to manage distressing symptoms. In a breakthrough, one of those trials resulted in a 7-month survival benefit. We will read much about distress in this 4th edition of Psycho-Oncology. For the importance of identifying and intervening on the sources of cancer patients’ distress—and even for the presence of the word “distress” in the psycho-oncologic lexicon—we have Ruth—and Jimmie—to thank. Ruth ended the last article she wrote with this: “. . . patients’ physical needs must be addressed before their psychosocial problems are identified. It is not just about taking care of their physical needs first. Rather, it is that we may be creating distress by not doing so.” Over the last 6 weeks of her life, she instructed her hospice care providers on how to manage her physical needs, and her close friends and family provided the physical touch she knew would comfort her emotionally. In this experience, one can find the truth of Ruth’s entire scientific career. In this 4th edition of Psycho-Oncology, you will find this truth woven into the science the book reports on: For Ruth, psycho-oncology was not just about how to support patients and families living with cancer. It was also about enabling them to have deaths “singularly free from mental distress.” It is thus fitting that, along with Jimmie C. Holland, we dedicate this edition to Ruth McCorkle. Contents Section editors xiii Contributors xv Introduction: Our Past, Our Future—New Frontiers in Psycho-Oncology 1 William S. Breitbart (Senior Editor) SECTION I Behavioral and Psychological Factors in Cancer Risk and Prevention Paul B. Jacobsen (Section Editor) 1 Tobacco Use and Cessation 7 Thomas H. Brandon, Vani N. Simmons, Úrsula Martínez, and Patricia Calixte-Civil 8 Cervical Cancer Screening and HPV Vaccination: Multilevel Challenges to Cervical Cancer Prevention 61 Richard Fielding, Samara Perez, Zeev Rosberger, Ovidiu Tatar, and Linda D. L. Wang 9 Breast Cancer Screening 68 Gabriel M. Leung, Irene O. L. Wong, Ava Kwong, and Joseph T. Wu 10 Prostate Cancer Screening 74 Michael A. Diefenbach, Daniel Nethala, Michael Schwartz, and Simon J. Hall 11 Lung Cancer Screening 78 Lisa Carter-Harris and Jamie Ostroff 12 Skin Cancer Screening 87 Jennifer L. Hay and Stephanie N. Christian 2 Diet and Cancer 13 Marian L. Fitzgibbon, Lisa Tussing-Humphreys, Angela Kong, and Alexis Bains 3 Physical Activity, Sedentary Behavior, and Cancer 21 Christine M. Friedenreich, Chelsea R. Stone, and Jessica McNeil 4 Sun Exposure and Cancer Risk 30 Suzanne J. Dobbinson, Afaf Girgis, Bruce K. Armstrong, and Anne E. Cust 5 Psychosocial Factors 36 Anika von Heymann and Christoffer Johansen 6 Viral Cancers and Behavior 43 Susan T. Vadaparampil, Lindsay N. Fuzzell, Shannon M. Christy, Monica L. Kasting, Julie Rathwell, and Anna E. Coghill SECTION III Screening and Testing for Germ Line and Somatic Mutations Paul B. Jacobsen (Section Editor) 13 Psychosocial Issues in Genetic Testing for Breast/Ovarian Cancer 95 Mary Jane Esplen, Jonathan Hunter, and Eveline M. A. Bleiker 14 Psychosocial Issues in Genetic Testing for Hereditary Colorectal Cancer 102 Sukh Makhnoon and Susan K. Peterson 15 Psychosocial Issues in Genomic Testing, Including Genomic Testing for Targeted Therapies 110 Megan Best SECTION II Screening for Cancer in Normal and At-Risk Populations Wendy W. T. Lam (Section Editor) 7 Colorectal Cancer Screening 53 Caitlin C. Murphy and Sally W. Vernon 16 Psychosocial Issues Related to Liquid Biopsy for ctDNA in Individuals at Normal and Elevated Risk 116 Jada G. Hamilton, Amanda Watsula-Morley, and Alicia Latham viii Contents SECTION IV Screening and Assessment in Psychosocial Oncology Wendy W. T. Lam (Section Editor) 17 Screening and Assessment for Distress 121 Alex J. Mitchell 18 Assessment, Screening, and Case Finding for Depression and Anxiety in People with Cancer 130 Kristine A. Donovan and Paul B. Jacobsen 19 Screening for Delirium and Dementia in the Cancer Patient 137 Christian Bjerre-Real, James C. Root, Yesne Alici, Julia A. Kearney, and William S. Breitbart 20 Screening and Assessment for Cognitive Problems 146 Alexandra M. Gaynor, James C. Root, Elizabeth Ryan, and Tim A. Ahles 29 Head and Neck Cancer 215 Loreto Fernández González, Jonathan Irish, and Gary Rodin 30 Central Nervous System Tumors 221 Alan D. Valentine 31 HIV Infection and AIDS-Associated Neoplasms 226 Joanna S. Dognin and Peter A. Selwyn SECTION VI Management of Specific Physical Symptoms William S. Breitbart (Section Editor) 32 Cancer-Related Pain 235 R. Garrett Key, Dustin Liebling, Vivek T. Malhotra, Steven D. Passik, Natalie Moryl, and William S. Breitbart 33 Nausea and Vomiting 255 Laura J. Lundi and Kavitha Ramchandran 34 Cancer-Related Fatigue 265 SECTION V Psychological Issues Related to Site of Cancer Mark Lazenby (Section Editor) 21 Melanoma 155 Nadine A. Kasparian and Iris Bartula 22 Lung Cancer 162 Marianne Davies 23 Breast Cancer 169 M. Tish Knobf and Youri Hwang 24 Colorectal Cancer 176 Anne Miles and Claudia Redeker 25 Prostate Cancer and Genitourinary Malignancies 182 Daniel C. McFarland, Christian Bjerre-Real, Yesne Alici, and William S. Breitbart 35 Sexual Problems and Cancer 276 Jeanne Carter, Ashley Arkema, Andrew J. Roth, Sally Saban, and Christian J. Nelson 36 Neuropsychological Impact of Cancer and Cancer Treatments 283 Alexandra M. Gaynor, James C. Root, and Tim A. Ahles 37 Sleep and Cancer 291 Amy E. Lowery-Allison and E. Devon Eldridge-Smith 38 Weight and Appetite Loss in Cancer 298 Yesne Alici and Victoria Saltz 39 Body Image—An Important Dimension in Cancer Care 303 Mary Jane Esplen and Michelle Cororve Fingeret Andrew J. Roth and Alejandro Gonzalez-Restrepo 26 Gastrointestinal Cancers 189 Daniel C. McFarland and William S. Breitbart 27 Gynecologic Cancers 196 Heidi S. Donovan and Teresa H. Thomas 28 Hematopoietic Dyscrasias and Stem Cell Transplantation/CAR-T Cell Therapy 203 Jesse R. Fann and Nicole Bates SECTION VII Psychiatric Disorders William S. Breitbart (Section Editor) 40 Adjustment Disorders in Cancer 313 Froukje de Vries, Sarah Hales, Gary Rodin, and Madeline Li 41 Depressive Disorders in Cancer 320 Christian Schulz-Quach, Madeline Li, Kimberley Miller, and Gary Rodin Contents 42 Suicide and Medical Aid in Dying 329 Hayley Pessin, Elie Isenberg-Grzeda, Reena Jaiswal, and Monique James 43 Anxiety Disorders 338 Ashley M. Nelson, Chelsea S. Rapoport, Lara Traeger, and Joseph A. Greer 44 Delirium 345 Yesne Alici and William S. Breitbart 45 Substance Use Disorders 355 Sameer Hassamal, Adam Rzetelny, and Steven D. Passik 46 Posttraumatic Stress Disorder Associated with Cancer Diagnosis and Treatment 363 Matthew Doolittle and Katherine N. DuHamel 47 Psychiatric Toxicities of Cancer Therapies: Focus on Immunotherapy and Targeted Therapy 374 Daniel C. McFarland, Mehak Sharma, and Yesne Alici SECTION VIII Evidence-Based Interventions William S. Breitbart and Phyllis N. Butow (Section Editors) Models of Care Delivery 48 Delivering Integrated Psychosocial Oncology Care: The Collaborative Care Model 385 Jesse R. Fann, Julia Ruark, and Michael Sharpe 49 The Engaged Patient: The Cancer Support Community’s Comprehensive Model of Psychosocial Programs, Services, and Research 393 Mitch Golant, Alexandra K. Zaleta, Susan Ash-Lee, Joanne S. Buzaglo, Kevin Stein, M. Claire Saxton, Marcia Donziger, Kim Thiboldeaux, and Linda Bohannon 50 The Role of Implementation Science in Advancing Psychosocial Cancer Care 400 Paul B. Jacobsen and Wynne E. Norton Interventions During Active Treatment 51 Supportive Psychotherapy in Cancer 409 Rosangela Caruso, Maria Giulia Nanni, and Luigi Grassi 52 Cognitive and Behavioral Interventions 416 Barbara L. Andersen, Nicole A. Arrato, and Caroline S. Dorfman 53 Metacognitive Approaches 424 Louise Sharpe and Leah Curran 54 Mindfulness-Based Interventions 429 Linda E. Carlson 55 Acceptance and Commitment Therapy (ACT) for Cancer Patients 438 Nicholas J. Hulbert-Williams, Ray Owen, and Christian J. Nelson 56 Supportive-Expressive and Other Forms of Group Psychotherapy in Cancer Care 445 David W. Kissane 57 Emotion-Focused Therapy 452 Sharon Manne 58 Interpersonal Psychotherapy and Cancer 459 Jennifer Sotsky, Hayley Pessin, and John C. Markowitz 59 Integrative Oncology 470 Santhosshi Narayanan, Gabriel Lopez, Jun J. Mao, Wenli Liu, and Lorenzo Cohen Interventions for Families and Couples 60 Psychosocial Interventions for Couples and Families Coping with Cancer 481 Talia I. Zaider and David W. Kissane Interventions for Advanced Cancer/End of Life/Bereavement 61 Meaning-Centered Psychotherapy 489 Melissa Masterson Duva, Wendy G. Lichtenthal, Allison J. Applebaum, and William S. Breitbart 62 Dignity Therapy 495 Harvey Max Chochinov and Maia S. Kredentser 63 Managing Cancer and Living Meaningfully (CALM) Therapy 502 Sarah Hales and Gary Rodin 64 Bereavement Interventions in the Setting of Cancer Care 509 Wendy G. Lichtenthal, Kailey E. Roberts, Holly G. Prigerson, and David W. Kissane Interventions for Cancer Survivors 65 Meaning-Centered Group Psychotherapy for Cancer Survivors 521 Nadia van der Spek, Wendy G. Lichtenthal, Karen Holtmaat, William S. Breitbart, and Irma M. Verdonck-de Leeuw 66 Physical Activity and Exercise Interventions in Cancer Survivors 528 Chloe Grimmett, Rebecca J. Beeken, and Abigail Fisher ix x Contents Digital Health Interventions 76 Financial Toxicity in Cancer Treatment 616 67 e-Health Interventions for Cancer Prevention and Control 537 77 The Experience of Cancer as an Immigrant 621 Kelly M. Shaffer, Elliot J. Coups,† and Lee M. Ritterband 68 Digital Health Interventions for Psychosocial Distress (Anxiety and Depression) in Cancer 543 Lisa Beatty and Haryana Dhillon 69 e-Health Interventions for Physical Symptom Control 550 Robert Zachariae 70 e-Health Interventions for Tobacco Cessation 561 Chris Kotsen, Jamie Ostroff, and Lisa Carter-Harris Victoria Blinder and Francesca M. Gany Francesca M. Gany and Jennifer Leng 78 Sexual and Gender Minority Health in Psycho-Oncology 627 Charles Kamen and Jennifer M. Jabson Tree SECTION XII Bio-Behavioral Psycho-Oncology William S. Breitbart and Mark Lazenby (Section Editors) 79 Psycho-Oncology, Stress Processes, and Cancer Progression 637 Michael H. Antoni, Jennifer M. Knight, and Susan K. Lutgendorf SECTION IX Psychosocial Issues at the Time of Diagnosis Matthew J. Loscalzo (Section Editor) 71 Treatment Decision Making 573 Allison Marziliano and Michael A. Diefenbach 72 The Family Meeting: Communication across the Continuum of Cancer Care 578 Stefanie N. Mooney and Marinel Olivares 80 Depression, Inflammation, and Cancer 644 Daniel C. McFarland, Leah E. Walsh, and Andrew H. Miller 81 Biobehavioral Psycho-Oncology Interventions 654 Michael A. Hoyt and Frank J. Penedo SECTION XIII Geriatric Psycho-Oncology Matthew J. Loscalzo (Section Editor) SECTION X Palliative and Supportive Care Mark Lazenby (Section Editor) 73 Psychological and Psychiatric Aspects of Palliative and End-of-Life Care: Synergies between Psycho- Oncology and Palliative Care 589 Scott A. Irwin, Nathan Fairman, Chase Samsel, Jeremy M. Hirst, Jason A. Webb, and Manuel Trachsel 74 Prognostic Understanding in Advanced Cancer Patients 599 Laura C. Polacek, Leah E. Walsh, Allison J. Applebaum, and Barry Rosenfeld SECTION XI Diversities in the Experience of Cancer Matthew J. Loscalzo (Section Editor) 75 Cancer, Culture, and Health Disparities 609 Marjorie Kagawa-Singer and Annalyn Valdez-Dadia 82 The Older Cancer Patient 663 Barbara A. Given and Charles W. Given 83 Geriatric Psycho-Oncology Assessment Issues and Interventions 671 Kelly M. Trevino, Rebecca M. Saracino, Andrew J. Roth, Yesne Alici, and Christian J. Nelson 84 Communicating with the Older Adult Cancer Patient 678 Patricia A. Parker, Smita C. Banerjee, and Beatriz Korc-Grodzicki SECTION XIV Pediatric Psycho-Oncology William S. Breitbart (Section Editor) 85 Screening and Assessment in Pediatric Psycho-Oncology 687 Darcy E. Burgers, Sarah J. Tarquini, Anne E. Kazak, and Anna C. Muriel Contents 86 Psychiatric Disorders in Pediatric Psycho- Oncology: Diagnosis and Management 696 Julia A. Kearney, Meredith E. MacGregor, and Maryland Pao 87 Evidence-Based Psychosocial Interventions in Pediatric Psycho-Oncology 703 Lori Wiener, Marie Barnett, Stacy Flowers, Cynthia Fair, and Amanda L. Thompson 88 Adolescent and Young Adult Patients 715 Christabel K. Cheung, Sheila J. Santacroce, and Bradley J. Zebrack SECTION XV Psychological Issues for the Family and Caregivers Phyllis N. Butow (Section Editor) 89 Including Family Members in Caring for the Patient with Cancer: A Family-Centered Approach 723 Douglas S. Rait 90 Couples Facing Cancer 729 Hoda Badr and Courtney Bitz SECTION XVII Building Supportive Care/Psycho-Oncology Teams Phyllis N. Butow (Section Editor) Building Supportive Care Teams: Working Together and Self-Care 96 Integrating Interdisciplinary Supportive Care Programs: Transforming the Culture of Cancer Care 775 Matthew J. Loscalzo, Karen L. Clark, Barry D. Bultz, and Juee Kotwal 97 Occupational Stress in Oncology Staff: Burnout, Resilience, and Interventions 782 Fay J. Hlubocky and Daniel C. McFarland Health Provider/Patient Communication 98 Principles of Communication Skills Training in Cancer Care across the Life Span and Illness Trajectory 791 David W. Kissane and Carma L. Bylund 91 Cancer Caregivers 737 Allison J. Applebaum, Erin Kent, Kristin Litzelman, Betty Ferrell, J. Nicholas Dionne-Odom, and Laurel Northouse 92 Addressing the Needs of Children When a Parent Has Cancer 745 Cynthia W. Moore, Greer J. Dent, and Paula K. Rauch SECTION XVIII Psycho-Oncology in Health Policy Wendy W. T. Lam (Section Editor) 99 Distress, the Sixth Vital Sign: A Catalyst for Standardizing Psychosocial Care Globally 801 SECTION XVI Survivorship Phyllis N. Butow and Wendy W. T. Lam (Section Editors) 93 Fear of Cancer Recurrence 755 Allan B. Smith, Joanna E. Fardell, and Phyllis N. Butow 94 Implementing the Survivorship Care Plan: A Strategy for Improving the Quality of Care for Cancer Survivors 760 Erin E. Hahn and Patricia A. Ganz 95 Adult Survivors of Childhood Cancer 767 Lisa A. Schwartz, Claire E. Wakefield, Jordana K. McLoone, Branlyn Werba DeRosa, and Anne E. Kazak Barry D. Bultz, Matthew J. Loscalzo, Alex J. Mitchell, and Jimmie C. Holland† 100 Implementation of Clinical Practice Guidelines for Psychosocial Cancer Care 806 Jane Turner and Nicole Rankin 101 Emerging International Directions for Psychosocial Care: Perspectives from Asia and Low-Middle-Income Countries 813 Jeff Dunn, Melissa Henry, and Maggie Watson Index 819 xi Section editors William S. Breitbart, MD, FAPOS Wendy W. T. Lam, RN, PhD, FFPH The Jimmie C. Holland Chair in Psychiatric Oncology Chairman Department of Psychiatry and Behavioral Sciences Memorial Sloan Kettering Cancer Center Professor of Clinical Psychiatry Vice-Chairman Department of Psychiatry Weill Cornell Medical College President Emeritus, International Psycho-oncology Society Associate Professor, Head, Division of Behavioural Sciences, School of Public Health Director, Jockey Club Institute of Cancer Care, Li Ka Shing Faculty of Medicine Director, Centre for Psycho-oncology Research and Training (CePORT) The University of Hong Kong Phyllis N. Butow, BA(Hons), DipEd, MClinPsych, MPH, PhD Professor of Psychological Medicine NHMRC Senior Principal Research Fellow Founding Chair, PoCoG School of Psychology University of Sydney Paul B. Jacobsen, PhD Associate Director Healthcare Delivery Research Program Division of Cancer Control and Population Sciences National Cancer Institute Mark Lazenby, APRN, PhD Associate Dean for Faculty and Student Affairs Professor of Nursing and Philosophy University of Connecticut School of Nursing Matthew J. Loscalzo, MSW, LCSW, FAPOS Liliane Elkins Professor in Supportive Care Programs Administrative Director, Sheri and Les Biller Patient and Family Resource Center Executive Director, Department of Supportive Care Medicine Professor, Department of Population Sciences City of Hope National Medical Center Contributors Tim A. Ahles, PhD Hoda Badr, PhD Attending Psychologist Director, Neurocognitive Laboratory Psychiatry Service Department of Psychiatry and Behavioral Sciences Member Memorial Sloan Kettering Cancer Center New York, NY, USA Associate Professor Department of Medicine Baylor College of Medicine Houston, TX, USA Yesne Alici, MD Associate Attending Psychiatrist Clinical Director, Co-Director, Bio-Behavioral Brain Clinic Psychiatry Service Department of Psychiatry and Behavioral Sciences Memorial Sloan Kettering Cancer Center New York, NY, USA Barbara L. Andersen, PhD Distinguished University Professor Department of Psychology Ohio State University Columbus, OH, USA Michael H. Antoni, PhD Professor Department of Psychology University of Miami and Sylvester Comprehensive Cancer Center Miami, FL, USA Allison J. Applebaum, PhD Assistant Attending Psychologist Director, Caregiver’s Clinic Psychiatry Service Department of Psychiatry and Behavioral Sciences Memorial Sloan Kettering Cancer Center New York, NY, USA Ashley Arkema, MS Nurse Practitioner Female Sexual Medicine Brooklyn, NY, USA Bruce K. Armstrong, BMedSci(Hons), MBBS(Hons), DPhil(Oxon), FRACP, FAFPHM Retired School of Population and Global Health The University of Western Australia Perth, WA, Australia Nicole A. Arrato, MA Graduate Research Assistant Department of Psychology Ohio State University Columbus, OH, USA Susan Ash-Lee, MSW, LCSW Vice President Clinical Services Program Cancer Support Community Denver, CO, USA Alexis Bains, BSc Nutrition Research Assistant Department of Kinesiology and Nutrition The University of Illinois at Chicago (UIC) Chicago, IL, USA Smita C. Banerjee, PhD Associate Attending Behavioral Scientist Behavioral Sciences Service Co-Director, Comskils Laboratory Department of Psychiatry and Behavioral Sciences Memorial Sloan Kettering Cancer Center New York, NY, USA Marie Barnett, PhD Assistant Attending Psychologist Psychiatry Service Department of Psychiatry and Behavioral Sciences & Department of Pediatrics Memorial Sloan Kettering Cancer Center New York, NY, USA Iris Bartula, DCP Head of Research Psychology Melanoma Institute Australia Senior Lecturer Northern Sydney Medical School Faculty of Medicine and Health University of Sydney Sydney, NSW, Australia Nicole Bates, MD Acting Assistant Professor Department of Psychiatry and Behavioral Sciences University of Washington Attending Psychiatrist Department of Psychosocial Oncology Seattle Cancer Care Alliance Seattle, WA, USA Lisa Beatty, PhD Senior Research Fellow College of Medicine and Public Health Flinders University Adelaide, SA, Australia Rebecca J. Beeken, PhD Associate Professor of Behavioural Medicine Leeds Institute of Health Sciences University of Leeds Leeds, Yorkshire, UK xvi Contributors Megan Best, PhD, MAAE, BMed(Hons), GradDipQHR Darcy E. Burgers, PhD Senior Lecturer Department of Psycho-Oncology Co-operative Research Group University of Sydney Broadway, NSW, Australia Psychologist Division of Pediatric Psychosocial Oncology Department of Psychosocial Oncology and Palliative Care Dana-Farber Cancer Institute Boston, MA, USA Courtney Bitz, MSW, LCSW, OSW-C Director of Clinical Social Work Department of Supportive Care Medicine City of Hope Duarte, CA, USA Christian Bjerre-Real, MD, MMCI Research Fellow Department of Psychiatry and Behavioral Sciences Memorial Sloan Kettering Cancer Center New York, NY, USA Eveline M. A. Bleiker, PhD Professor Department of Psychosocial Research and Epidemiology Netherlands Cancer Institute Amsterdam, The Netherlands Victoria Blinder, MD, MSc Associate Attending Oncologist Associate Member Immigrant Health and Cancer Disparities Service Department of Psychiatry and Behavioral Sciences Breast Medicine Service Department of Medicine Memorial Sloan Kettering Cancer Center New York, NY, USA Phyllis N. Butow, BA(Hons), DipEd, MClinPsych, MPH, PhD Professor of Psychological Medicine NHMRC Senior Principal Research Fellow, Founding Chair, PoCoG School of Psychology University of Sydney Sydney, NSW, Australia Joanne S. Buzaglo, PhD Executive Director, PRO Solutions Outcomes Sciences ConcertAI Rydal, PA, USA Carma L. Bylund, PhD Professor College of Journalism and Communications University of Florida Newberry, FL, USA Patricia Calixte-Civil, MA Doctoral Student Department of Psychology University of South Florida, Moffitt Cancer Center Tampa, FL, USA Linda E. Carlson, PhD President Global Headquarters Cancer Support Community Washington, DC, USA Professor Department of Oncology University of Calgary Cumming School of Medicine Calgary, AB, Canada Thomas H. Brandon, PhD Jeanne Carter, PhD Department Chair and Program Leader, Health Outcomes and Behavior Moffitt Distinguished Scholar Director, Tobacco Research and Intervention Program Moffitt Cancer Center Professor Departments of Psychology and Oncologic Sciences University of South Florida Tampa, FL, USA Attending Psychologist Director, Female Sexual Health Clinic Gynecology Service Department of Surgery Psychiatry Service Department of Psychiatry and Behavioral Medicine Memorial Sloan Kettering Cancer Center New York, NY, USA William S. Breitbart, MD, FAPOS Associate Attending Behavioral Scientist Behavioral Sciences Service Department of Psychiatry and Behavioral Sciences Memorial Sloan Kettering Cancer Center New York, NY, USA Linda Bohannon, MSM, BSN, RN Jimmie C. Holland Chair in Psychiatric Oncology Chairman Department of Psychiatry and Behavioral Sciences Member Attending Psychiatrist Supportive Care Service Department of Medicine Memorial Sloan Kettering Cancer Center Vice Chairman and Professor of Clinical Psychiatry Department of Psychiatry Weill Cornell Medical College New York, NY, USA Barry D. Bultz, AOE, PhD Professor and Head, Division of Psychosocial Oncology Daniel Family Leadership Chair in Psychosocial Oncology Department of Oncology Cumming School of Medicine Department of Psychosocial Oncology Tom Baker Cancer Center University of Calgary Calgary, AB, Canada Lisa Carter-Harris, PhD, APRN, ANP-C , FAAN Rosangela Caruso, MD, PhD Doctor Biomedical and Specialty Surgical Sciences University of Ferrara Ferrara, Emilia Romagna, Italy Christabel K. Cheung, PhD, MSW Assistant Professor University of Maryland School of Social Work Member University of Maryland Greenbaum Comprehensive Cancer Center Baltimore, MD, USA Harvey Max Chochinov, OM, OC, PhD, MD, FRCPC, FRSC, FCAHS Distinguished Professor Department of Psychiatry University of Manitoba Winnipeg, MB, Canada Contributors Stephanie N. Christian, MPH Haryana Dhillon, BSc, MA(Psych), PhD K. Leroy Irvis Fellow Department of Behavioral and Community Health Sciences University of Pittsburgh Graduate School of Public Health Pittsburgh, PA, USA Associate Professor Centre for Medical Psychology and Evidence-based Decision-making, School of Psychology, Faculty of Science The University of Sydney Camperdown, NSW, Australia Shannon M. Christy, PhD Department of Health Outcomes and Behavior Division of Population Science H. Lee Moffitt Cancer Center and Research Institute Department of Oncologic Sciences Morsani College of Medicine University of South Florida Center for Immunization and Infection Research in Cancer Lee Moffitt Cancer Center and Research Institute Tampa, FL, USA Karen L. Clark, MS Manager of Supportive Care Programs Department of Supportive Care Medicine City of Hope National Medical Center Duarte, CA, USA Anna E. Coghill, PhD, MPH Assistant Member Cancer Epidemiology Moffitt Cancer Center Tampa, FL, USA Lorenzo Cohen, PhD Professor and Director, Integrative Medicine Program Department of Palliative, Rehabilitation and Integrative Medicine The University of Texas MD Anderson Cancer Center Houston, TX, USA Elliot J. Coups, PhD† Member Rutgers Cancer Institute of New Jersey Department of Medicine Rutgers Robert Wood Johnson Medical School Rutgers, The State University of New Jersey New Brunswick, NJ, USA Leah Curran, DCP Clinical Psychologist Department of Psychology The University of Sydney Camperdown, NSW, Australia Anne E. Cust, PhD, MPH(Hons), BSc, BA Professor of Cancer Epidemiology Sydney School of Public Health And the Melanoma Institute Australia The University of Sydney Camperdown, NSW, Australia Marianne Davies, DNP, ACNP, AOCNP Associate Professor Yale School of Nursing Department of Oncology Nurse Practitioner Smilow Cancer Hospital New Haven, CT, USA Greer J. Dent, BA Clinical Research Coordinator Department of Cancer Center Massachusetts General Hospital Boston, MA, USA Branlyn Werba DeRosa, PhD Research Director Department of Research and Training Institute Cancer Support Community Ardmore, PA, USA Michael A. Diefenbach, PhD Professor Departments of Medicine, Urology and Psychiatry Northwell Health Manhasset, NY, USA J. Nicholas Dionne-Odom, PhD, RN, ACHPN Assistant Professor School of Nursing University of Alabama at Birmingham Hoover, AL, USA Suzanne J. Dobbinson, BSc, MSc, PhD Senior Research Fellow Centre for Behavioural Research in Cancer Cancer Council Victoria Kensington, VIC, Australia Joanna S. Dognin, PsyD Psychologist Department of Veterans Affairs NYU Langone Medical Center White Plains, NY, USA Heidi S. Donovan, PhD, RN Professor Co-Director National Rehabilitation Research and Training Center on Family Support Department of Health and Community Systems University of Pittsburgh Pittsburgh, PA, USA Kristine A. Donovan, PhD, MBA Associate Member Department of Supportive Care Medicine Moffitt Cancer Center Tampa, FL, USA Marcia Donziger, BA Vice President Digital Strategy and Business Development Cancer Support Community Denver, CO, USA Matthew Doolittle, MD Assistant Attending Psychiatrist Psychiatry Service Department of Psychiatry and Behavioral Sciences Memorial Sloan Kettering Cancer Center New York, NY, USA Caroline S. Dorfman, PhD Assistant Professor Department of Psychiatry and Behavioral Sciences Duke University Medical Center Durham, NC, USA Katherine N. DuHamel, PhD Director KND Consulting New York, NY, USA Jeff Dunn, PhD, AO Professor Department of Research and Innovation University of Southern Queensland Toowoomba, Queensland, Australia xvii xviii Contributors E. Devon Eldridge-Smith, PhD Marian L. Fitzgibbon, PhD Assistant Professor Department of Medicine National Jewish Health Denver, CO, USA Professor Department of Pediatrics University of Illinois Chicago, IL, USA Mary Jane Esplen, PhD Stacy Flowers, PsyD Professor and Vice-Chair Department of Psychiatry Faculty of Medicine University of Toronto Toronto, ON, Canada Associate Professor Director of Behavioral Science Department of Family Medicine Wright State University Columbus, OH, USA Cynthia Fair, LCSW, MPH, DrPH Christine M. Friedenreich, PhD, FCAHS, FRSC Professor and Department Chair Department of Public Health Studies Elon University Elon, NC, USA Scientific Director Department of Cancer Epidemiology and Prevention Research Alberta Health Services Arnie Charbonneau Cancer Institute Adjunct Professor The Faculties of Medicine and Kinesiology University of Calgary Calgary, AB, Canada Nathan Fairman, MD, MPH Associate Clinical Professor Department of Psychiatry and Behavioral Sciences UC Davis School of Medicine Sacramento, CA, USA Jesse R. Fann, MD, MPH Professor Department of Psychiatry and Behavioral Sciences Adjunct Professor Departments of Rehabilitation Medicine and Epidemiology University of Washington Medical Director Department of Psychosocial Oncology Seattle Cancer Care Alliance Clinical Research Division Fred Hutchinson Cancer Research Center Seattle, Washington Seattle, WA, USA Joanna E. Fardell, PhD, MClinNeuropsych, BSc Research Fellow Department of Behavioural Sciences Unit, Discipline of Paediatrics, School of Women’s and Children’s Health, Faculty of Medicine University of New South Wales Randwick, NSW, Australia Loreto Fernández González, BA, BSc, MPH PhD Student and Connaught Scholar Social and Behavioural Health Sciences Dalla Lana School of Public Health University of Toronto Toronto, ON, Canada Betty Ferrell, RN, PhD, FAAN Professor Nursing Research City of Hope National Medical Center Duarte, CA, USA Richard Fielding, BA(Hons), CPsychol, PhD, FFPH, FHKPsyS Lindsay N. Fuzzell, PhD Applied Research Scientist I Health Outcomes and Behavior Moffitt Cancer Center Tampa, FL, USA Francesca M. Gany, MD, MS Attending Physician Chief, Immigrant Health and Cancer Disparities Service Department of Psychiatry and Behavioral Sciences Member Memorial Sloan Kettering Cancer Center Professor Department of Medicine and Department of Healthcare Policy & Research Weill Cornell Medicine New York, NY, USA Patricia A. Ganz, MD Distinguished Professor Schools of Medicine and Public Health University of California, Los Angeles (UCLA) Los Angeles, CA, USA Alexandra M. Gaynor, PhD Post-Doctoral Neuropsychology Research Fellow Department of Psychiatry and Behavioral Sciences Memorial Sloan Kettering Cancer Center New York, NY, USA Afaf Girgis, PhD, BSc(Hons) Professor Director, Psycho-Oncology Research Group University of New South Wales Sydney, Australia Clinical Lead Jockey Club Institute of Cancer Care, Li Ka Shing Faculty of Medicine Honorary Professor, School of Public Health The University of Hong Kong Hong Kong, China Barbara A. Given, PhD, RN, FAAN Michelle Cororve Fingeret, PhD Charles W. Given, PhD Fingeret Psychology Services Houston, TX, USA Professor Emeritus College of Nursing Michigan State University Okemos, MI, USA Abigail Fisher, PhD Associate Professor Department of Behavioural Science and Health University College London Bloomsbury, London, UK University Distinguished Professor, Associate Dean Emeritus College of Nursing Michigan State University Okemos, MI, USA Mitch Golant, PhD Senior Consultant, Strategic Initiatives Research and Training Institute Cancer Support Community Los Angeles, CA, USA Contributors Alejandro Gonzalez-Restrepo, MD Melissa Henry, PhD Attending Psychiatrist Hartford Hospital/Institute of Living Hartford Healthcare Simsbury, CT, USA Associate Professor Department of Oncology Faculty of Medicine McGill University Montreal, QC, Canada Luigi Grassi, MD Professor and Chair of Psychiatry University of Ferrara Chairman of the Department of Biomedical and Specialty Surgical Sciences University of Ferrara Ferrara, Italy Anika von Heymann, MSc, Psych, PhD Joseph A. Greer, PhD Jeremy M. Hirst, MD Associate Professor of Psychology Department of Psychiatry Harvard Medical School Program Director, Center for Psychiatric Oncology & Behavioral Sciences Massachusetts General Hospital Cancer Center Boston, MA, USA Clinical Professor of Psychiatry; Palliative Care Psychiatry Department of Psychiatry; Palliative Medicine UC San Diego School of Medicine La Jolla, CA, USA Chloe Grimmett, PhD Senior Research Fellow School of Health Sciences University of Southampton Hampshire, UK Erin E. Hahn, PhD, MPH Research Scientist Department of Research and Evaluation Kaiser Permanente Southern California Pasadena, CA, USA Sarah Hales, MD, PhD Assistant Professor Division of Psychosocial Oncology Department Supportive Care Princess Margaret Cancer Centre Centre for Mental Health University Health Network University of Toronto Toronto, ON, Canada Simon J. Hall, MD Zucker Professor of Urologic Oncology Smith Institute of Urology Hofstra Northwell School of Medicine Lake Success, NY, USA Jada G. Hamilton, PhD, MPH Assistant Attending Psychologist Behavioral Sciences Service Department of Psychiatry and Behavioral Sciences; Department of Medicine Assistant Member Memorial Sloan Kettering Cancer Center New York, NY, USA Sameer Hassamal, MD Assistant Professor Department of Psychiatry Arrowhead Regional Medical Center Colton, CA, USA Jennifer L. Hay, PhD Attending Psychologist Behavioral Sciences Service Department of Psychiatry and Behavioral Sciences Member Memorial Sloan Kettering Cancer Center New York, NY, USA Postdoctoral Fellow Department of Oncology Rigshospitalet København, Denmark Fay J. Hlubocky, PhD, MA, CCTP Clinical Health Psychologist Research Project Professor Department of Medicine University of Chicago Medicine Chicago, IL, USA Jimmie C. Holland, MD† Wayne E. Chapman Chair in Psychiatric Oncology Attending Psychiatrist Psychiatry Service Department of Psychiatry and Behavioral Sciences Member Memorial Sloan Kettering Cancer Center New York, NY, USA Karen Holtmaat, MSc, MA Assistant Professor in Psychosocial Oncology Department of Clinical, Neuro-and Developmental Psychology Vrije Universiteit Amsterdam Amsterdam, NH, The Netherlands Michael A. Hoyt, PhD Associate Professor Chao Cancer Center UC Irvine Irvine, CA, USA Nicholas J. Hulbert-Williams, BSc, PhD, CPsychol, APBPsS, FHAE Professor of Behavioural Medicine School of Psychology University of Chester Chester, UK Jonathan Hunter, BSc, MD, FRCPC Professor Department of Psychiatry University of Toronto Toronto, ON, Canada Youri Hwang, MSN, RN, FNP-C PhD Student School of Nursing Yale University New Haven, CT, USA Jonathan Irish, MD, MSc, FRCSC, FACS Professor and Head Division of Head and Neck Oncology and Reconstructive Surgery Department of Otolaryngology-Head and Neck Surgery Princess Margaret Cancer Centre, University of Toronto Toronto, ON, Canada xix xx Contributors Scott A. Irwin, MD, PhD, FACLP, FAPA Anne E. Kazak, PhD, ABPP Professor of Psychiatry and Behavioral Neurosciences Department of Psychiatry and Behavioral Neurosciences Director of Patient and Family Support Program Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Health System Los Angeles, CA, US Editor-in-Chief, American Psychologist Director, Center for Healthcare Delivery Science Nemours Children’s Health System Co-Director Center for Pediatric Traumatic Stress Professor Department of Pediatrics Thomas Jefferson University Wilmington, DE, USA Elie Isenberg-Grzeda, MD, CM, FRCPC Assistant Professor Department of Psychiatry University of Toronto Toronto, ON, Canada Jennifer M. Jabson Tree, PhD, MPH Associate Professor Department of Public Health University of Tennessee Knoxville, TN, USA Paul B. Jacobsen, PhD Associate Director Division of Cancer Control and Population Sciences National Cancer Institute Bethesda, MD, USA Reena Jaiswal, MD Assistant Attending Psychiatrist Psychiatry Service Department of Psychiatry and Behavioral Sciences Memorial Sloan Kettering Cancer Center New York, NY, USA Monique James, MD Assistant Attending Psychiatrist Psychiatry Service Department of Psychiatry and Behavioral Sciences Memorial Sloan Kettering Cancer Center New York, NY, USA Christoffer Johansen, MD, PhD, Dr. Med. Sci. Professor Head, CASTLE—Cancer Late Effect Research Oncology Clinic Department of Oncology Center for Surgery and Cancer Rigshospitalet Copenhagen, Denmark Marjorie Kagawa-Singer, PhD, MA, MN, RN Julia A. Kearney, MD Assistant Attending Psychiatrist Clinical Director, Pediatric Psycho-Oncology Program Department of Psychiatry and Behavioral Sciences & Department of Pediatrics Memorial Sloan Kettering Cancer Center New York, NY, USA Erin Kent, PhD, MS Associate Professor Health Policy and Management University of North Carolina Chapel Hill, NC, USA R. Garrett Key, MD, FAPA, FACLP Assistant Professor Psychiatry and Behavioral Sciences University of Texas at Austin Dell Medical School Austin, TX, USA David W. Kissane, AC, MBBS, MPM, MD, FRANZCP, FAChPM, FACLP UNDA Chair of Palliative Medicine Research Cunningham Centre for Palliative Care, St Vincent’s Sydney University of Notre Dame Australia Head of Szalmuk Family Psycho-oncology Research Unit Department of Palliative Care Cabrini Health, Melbourne, Australia Head of Psycho-Oncology Clinic Monash Medical Centre Monash University Clayton, VIC, Australia Jennifer M. Knight, MD, MS Associate Professor Department of Psychiatry, Medicine, and Microbiology and Immunology Medical College of Wisconsin Shorewood, WI, USA M. Tish Knobf, PhD, RN, FAAN Research Professor Community Health Sciences University of California, Los Angeles (UCLA) Los Angeles, CA, USA Professor Department of Nursing Yale University New Haven, CT, USA Charles Kamen, PhD, MPH Angela Kong, PhD, MPH, RD Assistant Professor Department of Surgery University of Rochester Rochester, NY, USA Assistant Professor Department of Pharmacy Systems, Outcomes, and Policy University of Illinois Chicago Chicago, IL, USA Nadine A. Kasparian, PhD Beatriz Korc-Grodzicki, MD, PhD, FAGS Professor of Pediatrics Director, Cincinnati Children’s Center for Heart Disease and Mental Health Heart Institute and Division of Behavioral Medicine and Clinical Psychology Department of Pediatrics University of Cincinnati College of Medicine Cincinnati, OH, USA Chief, Geriatrics Service Department of Medicine Memorial Sloan-Kettering Cancer Center Professor of Medicine Weill Cornell Medical College New York, NY, USA Monica L. Kasting, PhD Chris Kotsen, PsyD, NCTTP Assistant Professor Department of Public Health Purdue University West Lafayette, IN, USA Associate Attending Psychologist Associate Director, Tobacco Treatment Program Department of Psychiatry and Behavioral Sciences Memorial Sloan Kettering Cancer Center New York, NY, USA Contributors Juee Kotwal, MBS, PMP Kristin Litzelman, PhD Business Manager Department of Supportive Care Medicine City of Hope National Medical Center Duarte, CA, USA Assistant Professor Department of Human Development and Family Studies University of Wisconsin-Madison Madison, WI, USA Maia S. Kredentser, PhD Wenli Liu, MD Research Fellow Clinical Health Psychology University of Manitoba Winnipeg, MB, Canada Associate Professor Department of Palliative, Rehabilitation and Integrative Medicine MD Anderson Cancer Center Houston, TX, USA Ava Kwong, MBBS, BSc, PhD, FRCS, FRCS, FHKAM, FCSHK Gabriel Lopez, MD Clinical Professor Division of Breast Surgery, Department of Surgery The University of Hong Kong Li Ka Shing Faculty of Medicine Pok Fu Lam, Hong Kong, China Associate Professor, Center Medical Director Department of Palliative, Rehabilitation, and Integrative Medicine University of Texas, MD Anderson Cancer Center Houston, TX, USA Wendy W. T. Lam, RN, PhD, FFPH Matthew J. Loscalzo, MSW, LCSW, FAPOS Associate Professor, Head, Division of Behavioural Sciences, School of Public Health Director, Jockey Club Institute of Cancer Care, Li Ka Shing Faculty of Medicine Director, Centre for Psycho-oncology Research and Training (CePORT) The University of Hong Kong Hong Kong, China Alicia Latham, MD Assistant Attending Physician Department of Medicine Memorial Sloan Kettering Cancer Center New York, NY, USA Mark Lazenby, APRN, PhD Professor of Nursing and Philosophy Associate Dean for Faculty and Student Affairs University of Connecticut School of Nursing Storrs, CT, USA Jennifer Leng, MD, MPH Associate Attending Physician Associate Member Immigrant Health and Cancer Disparities Service Department of Psychiatry and Behavioral Sciences Memorial Sloan Kettering Cancer Center New York, NY, USA Gabriel M. Leung, MD Dean of Medicine LKS Faculty of Medicine The University of Hong Kong Pok Fu Lam, Hong Kong Madeline Li, MD, PhD, FRCP(C) Attending Psychiatrist Department of Supportive Care Princess Margaret Cancer Centre University Health Network Associate Professor Department of Psychiatry Faculty of Medicine University of Toronto Toronto, ON, Canada Wendy G. Lichtenthal, PhD, FT Associate Attending Psychologist Director, Bereavement Clinic, Psychiatry Service Department of Psychiatry and Behavioral Sciences Memorial Sloan Kettering Cancer Center New York, NY, USA Dustin Liebling, MD Clinical Fellow Anesthesia Pain Management Service Department of Anesthesiology and Critical Care Medicine Memorial Sloan Kettering Cancer Center New York, NY, USA Liliane Elkins Professor in Supportive Care Programs Administrative Director, Sheri & Les Biller Patient and Family Resource Center Executive Director, Department of Supportive Care Medicine Professor, Department of Population Sciences City of Hope National Medical Center Duarte, CA, USA Amy E. Lowery-Allison, PhD Associate Attending Psychologist Psychiatry Service Department of Psychiatry and Behavioral Sciences Memorial Sloan Kettering Cancer Center New York, NY, USA Laura J. Lundi, BS Administrative Clinical Research Coordinator Department of Medicine Stanford University Stanford, CA, USA Susan K. Lutgendorf, PhD Professor and Starch Faculty Fellow Departments of Psychological and Brain Sciences, Obstetrics and Gynecology, and Urology University of Iowa Iowa City, IA, USA Irma M. Verdonck-de Leeuw, PhD Full Professor in Psychosocial Oncology Department of Otolaryngology/Head and Neck Surgery Vrije University Medical Center/Cancer Center Amsterdam Section of Clinical, Neuro-and Developmental Psychology Vrije Universiteit Amsterdam Amsterdam, The Netherlands Meredith E. MacGregor, MD Assistant Attending Psychiatrist Department of Child and Adolescent Psychiatry and Behavioral Sciences Children’s Hospital of Philadelphia Philadelphia, PA, USA Sukh Makhnoon, PhD, MS Postdoctoral Fellow Department of Behavioral Science University of Texas MD Anderson Cancer Center Houston, TX, USA Vivek T. Malhotra, MD, MPH Chief, Anesthesiology Pain Service Associate Attending Department of Anesthesiology and Critical Care Medicine Memorial Sloan Kettering Cancer Center New York, NY, USA Sharon Manne, PhD Section Chief, Professor of Medicine Department of CINJ, Behavioral Sciences Rutgers, The State University of New Jersey New Brunswick, NJ, USA xxi xxii Contributors Jun J. Mao, MD, MSCE Alex J. Mitchell, MBBS, MSc, MD, MRCPsych Laurance S. Rockefeller Chair in Integrative Medicine Chief, Integrative Medicine Service Attending Physician Department of Medicine Memorial Sloan Kettering Cancer Center, Bendheim Integrative Medicine Center New York, NY, USA Professor Department of Psycho-Oncology and Cancer Care University of Leicester Leicester, UK John C. Markowitz, MD Research Psychiatrist New York State Psychiatric Institute Professor of Clinical Psychiatry Columbia University College of Physicians and Surgeons New York, NY, USA Úrsula Martínez, PhD Applied Research Scientist Department of Health Outcomes and Behavior H. Lee Moffitt Cancer Center and Research Institute Tampa, FL, USA Allison Marziliano, PhD Postdoctoral Fellow Department of Medicine Northwell Health Bethpage, NY, USA Melissa Masterson Duva, PhD Senior Psychologist WTC Health Program Clinical Center of Excellence New York University School of Medicine New York, NY, USA Stefanie N. Mooney, MD Assistant Clinical Professor of Medicine Division of Supportive Medicine Department of Supportive Care Medicine City of Hope National Medical Center Duarte, CA, USA Cynthia W. Moore, PhD Psychologist Department of Child and Adolescent Psychiatry Massachusetts General Hospital Boston, MA, USA Natalie Moryl, MD Associate Attending Physician Supportive Care Service Department of Medicine Memorial Sloan Kettering Cancer Center Associate Professor Department of Medicine Weill Cornell Medical College New York, NY, USA Anna C. Muriel, MD, MPH Research Fellow Department of Psychiatry and Behavioral Sciences Memorial Sloan Kettering Cancer Center New York, NY, USA Chief, Division of Pediatric Psychosocial Oncology Associate Psychiatrist Department of Psychosocial Oncology and Palliative Care Dana Farber Cancer Institute Assistant Professor of Psychiatry Harvard Medical School Boston, MA, USA Jordana K. McLoone, PhD Caitlin C. Murphy, PhD, MPH Daniel C. McFarland, DO Post-Doctoral Research Fellow Women’s and Children’s Health, Faculty of Medicine University of New South Wales NSW, Australia Jessica McNeil, PhD Postdoctoral Fellow Cancer Epidemiology and Prevention Research Alberta Health Services Russell, ON, Canada Anne Miles, BSc, PhD Reader in Psychology Department of Psychological Sciences Birkbeck, University of London Bloomsbury, London, UK Andrew H. Miller, MD William P. Timmie Professor of Psychiatry and Behavioral Sciences Department of Psychiatry and Behavioral Sciences Emory University School of Medicine Atlanta, GA, USA Kimberley Miller, MD, FRCPC Attending Psychiatrist Department of Supportive Care Princess Margaret Cancer Centre, University Health Network Assistant Professor of Psychiatry Department of Psychiatry Faculty of Medicine University of Toronto Toronto, ON, Canada Assistant Professor Department of Population and Data Sciences UT Southwestern Medical Center Dallas, TX, USA Maria Giulia Nanni, MD Associate Professor Department of Biomedical and Specialty Surgical Sciences University of Ferrara Ferrara, Italy Santhosshi Narayanan, MD Assistant Professor Department of PRIM MD Anderson Cancer Center Houston, TX, USA Ashley M. Nelson, PhD Postdoctoral Fellow Department of Psychiatry Massachusetts General Hospital/Harvard Medical School Boston, MA, USA Christian J. Nelson, PhD Chief, Psychiatry Service, Associate Attending Psychologist Department of Psychiatry and Behavioral Sciences Associate Member Memorial Sloan Kettering Cancer Center New York, NY, USA Contributors Daniel Nethala, MD Hayley Pessin, PhD Resident Department of Urology Smith Institute for Urology at Northwell Health Lake Success, NY, USA Psychologist Department of Psychiatry and Behavioral Sciences Memorial Sloan Kettering Cancer Center New York, NY, USA Laurel Northouse, PhD Susan K. Peterson, PhD, MPH Professor Emerita School of Nursing University of Michigan Ann Arbor, MI, USA Professor Department of Behavioral Science The University of Texas MD Anderson Cancer Center Houston, TX, USA Wynne E. Norton, PhD Laura C. Polacek, MA Program Director Division of Cancer Control and Population Sciences National Cancer Institute Bethesda, MD, USA Graduate Student Department of Psychology Fordham University The Bronx, NY, USA Marinel Olivares, LCSW, ACHP-SW Holly G. Prigerson, PhD Clinical Social Worker Adult Hematology/Hematopoietic Cell Transplantation Director, Family Meeting Program Department of Supportive Care Medicine City of Hope National Medical Center Duarte, CA Irving Sherwood Wright Professor in Geriatrics Professor of Sociology in Medicine Director Cornell Center for Research on End-of-Life Care New York, NY, USA Jamie Ostroff, PhD Clinical Professor of Psychiatry and Behavioral Sciences Chief, Couples and Family Therapy Clinic Department of Psychiatry and Behavioral Sciences Stanford University Stanford, CA, USA Chief, Behavioral Sciences Service Vice Chair for Research Member and Attending Psychologist Director, Tobacco Treatment Program Department of Psychiatry and Behavioral Sciences Memorial Sloan Kettering Cancer Center New York, NY, USA Ray Owen, DClinPsych, Cpsychol, FHEA Consultant Clinical Psychologist Health Psychology Department Wye Valley NHS Trust Hereford, UK Maryland Pao, MD Clinical and Deputy Scientific Director Department of NIMH National Institutes of Health (NIH) Bethesda, MD, USA Patricia A. Parker, PhD Attending Psychologist Behavioral Sciences Service Director, Communication Skills Training and Research Program Department of Psychiatry and Behavioral Sciences Member, Memorial Hospital Memorial Sloan-Kettering Cancer Center New York, NY, USA Steven D. Passik, PhD Vice President, Scientific Affairs, Education and Policy Collegium Pharmaceuticals Stoughton, MA, USA Frank J. Penedo, PhD Professor Department of Psychology and Medicine University of Miami Miami, FL, USA Samara Perez, PhD Research Associate Department Lady Davis Institute for Medical Research Jewish General Hospital Clinical Psychologist Psychosocial Oncology Program McGill University Health Center Montreal, QC, Canada Douglas S. Rait, PhD Kavitha Ramchandran, MD Clinical Associate Professor Department of Medicine Stanford University Stanford, CA, USA Nicole Rankin, BA(Hons), MSc, PhD Senior Research Fellow Faculty of Medicine and Health The University of Sydney Camperdown, NSW, Australia Chelsea S. Rapoport, BA Clinical Research Coordinator Department of Psychiatry Massachusetts General Hospital Boston, MA, USA Julie Rathwell, MPH Research Project Specialist, Sr. Department of Cancer Epidemiology Moffitt Cancer Center Tampa, FL, USA Paula K. Rauch, MD Director, Marjorie E. Korff Parenting at a Challenging Time Program Department of Psychiatry Massachusetts General Hospital Boston, MA, USA Claudia Redeker, MSc PhD Student Department of Psychology Birkbeck College Bloomsbury, London, UK Lee M. Ritterband, PhD Professor Department of Psychiatry and Neurobehavioral Sciences University of Virginia Charlottesville, VA, USA xxiii xxiv Contributors Kailey E. Roberts, PhD Sally Saban, BA Research Fellow Department of Psychiatry and Behavioral Sciences Memorial Sloan Kettering Cancer Center New York, NY, USA Clinic Research Coordinator Department of Surgery New York, NY, USA Gary Rodin, MD Student Williams College New York, NY, USA Head of Department Department of Supportive Care Princess Margaret Cancer Centre University Health Network Professor Department of Psychiatry Faculty of Medicine Director Global Institute of Psychosocial, Palliative and End-of-Life Care University of Toronto Toronto, ON, Canada James C. Root, PhD Associate Attending Neuropsychologist Psychiatry Service Department of Psychiatry and Behavioral Sciences Memorial Sloan Kettering Cancer Center New York, NY, USA Zeev Rosberger, PhD Senior Investigator and Associate Professor Departments of Oncology, Psychiatry, and Psychology Lady Davis Institute for Medical Research Institute of Community and Family Psychiatry Jewish General Hospital Gerald Bronfman Department of Oncology McGill University Montréal, QC, Canada Barry Rosenfeld, PhD Professor Chairman Department of Psychology Fordham University Consultant Psychologist Psychiatry Service Department of Psychiatry and Behavioral Sciences Memorial Sloan Kettering Cancer Center New York, NY, USA Andrew J. Roth, MD Attending Psychiatrist Psychiatry Service Department of Psychiatry and Behavioral Sciences Memorial Sloan Kettering Cancer Center Professor of Clinical Psychiatry Weill Cornell Medical College New York, NY, USA Julia Ruark, MD, MPH Attending Psychiatrist Department of Psychosocial Oncology Seattle Cancer Care Alliance Seattle, WA, USA Elizabeth Ryan, PhD, ABPP-CN Associate Attending Neuropsychologist Psychiatry Service Department of Psychiatry and Behavioral Sciences Memorial Sloan Kettering Cancer Center New York, NY, USA Adam Rzetelny, PhD Senior Medical Science Liaison Department of Medical Affairs Collegium Pharmaceuticals Maplewood, NJ, USA Victoria Saltz Chase Samsel, MD Attending Physician Department of Psychiatry Boston Children’s Hospital and Dana-Farber Cancer Institute Boston, MA, USA Sheila J. Santacroce, PhD, RN, CPNP, FAANP Associate Professor School of Nursing University of North Carolina Chapel Hill, NC, USA Rebecca M. Saracino, PhD Assistant Attending Psychologist Psychiatry Service Department of Psychiatry and Behavioral Sciences Memorial Sloan Kettering Cancer Center New York, NY, USA M. Claire Saxton, MBA Vice President Department of Education and Outreach Cancer Support Community Washington, DC, USA Christian Schulz-Quach, MD, MSc, MA Assistant Professor Department of Psychiatry University of Toronto Toronto, ON, Canada Lisa A. Schwartz, PhD Associate Professor Department of Pediatrics/Oncology The Children’s Hospital of Philadelphia Philadelphia, PA, USA Michael Schwartz, MD, FACS Associate Professor Smith Institute for Urology Northwell Health New Hyde Park, NY, USA Peter A. Selwyn, MD, MPH Professor and Chair Department of Family and Social Medicine Director, Palliative Care Program Montefiore Medical Center Albert Einstein College of Medicine The Bronx, NY, USA Kelly M. Shaffer, PhD Assistant Professor Center for Behavioral Health and Technology, Department of Psychiatry and Neurobehavioral Sciences University of Virginia School of Medicine Charlottesville, VA, USA Mehak Sharma, MD Assistant Attending Psychiatrist Psychiatry Service Department of Psychiatry and Behavioral Sciences Memorial Sloan Kettering Cancer Center New York, NY, USA Contributors Louise Sharpe, BA(Hons), MPsychol, PhD Manuel Trachsel, MD, PhD Professor of Clinical Psychology Department of Psychology The University of Sydney Camperdown, NSW, Australia Head Clinical Ethicist Clinical Ethics Unit University Hospital of Basel/University of Zurich Zürich, ZH, Switzerland Michael Sharpe, MA, MD Lara Traeger, PhD Professor of Psychological Medicine Department of Psychiatry University of Oxford Oxford, UK Assistant Professor Department of Psychiatry Massachusetts General Hospital/Harvard Medical School Boston, MA, USA Vani N. Simmons, PhD Kelly M. Trevino, PhD Senior Member Department of Health Outcomes and Behavior Moffitt Cancer Center Tampa, FL, USA Associate Attending Psychologist Psychiatry Service Department of Psychiatry and Behavioral Sciences Memorial Sloan Kettering Cancer Center New York, NY, USA Allan B. Smith, PhD Co-Deputy Director (Policy and Practice) Centre for Oncology Education and Research Translation (CONCERT) Ingham Institute for Applied Medical Research Liverpool, NSW, Australia Jennifer Sotsky, MD, MS Clinical Fellow Department of Psychiatry New York State Psychiatric Institute New York, NY, USA Nadia van der Spek, PhD Assistant Professor Licensed Mental Health Care Psychologist Department of Clinical Psychology Vrije Universiteit Amsterdam Amsterdam, The Netherlands Kevin Stein, PhD, FAPOS Associate Professor (Adjunct) Department of Behavioral Sciences and Health Education Rollins School of Public Health Snellville, GA, USA Chelsea R. Stone, MSc Research Associate Department of Cancer Epidemiology and Prevention Research Alberta Health Services Calgary, AB, Canada Sarah J. Tarquini, PhD Senior Psychologist Department of Psychosocial Oncology and Palliative Care Dana-Farber Boston Children’s Cancer and Blood Disorders Center Boston, MA, USA Ovidiu Tatar, MD, MSc Research Associate Psychosocial Oncology Lady Davis Institute for Medical Research Jewish General Hospital Research Center-Centre Hospitalier de l'Université de Montréal (CRCHUM) Montréal, QC, USA Kim Thiboldeaux, BA Communications CEO Cancer Support Community Washington, DC, USA Teresa H. Thomas, PhD, RN Assistant Professor School of Nursing University of Pittsburgh Pittsburgh, PA, USA Amanda L. Thompson, PhD Chief, Pediatric Psychology Department of Life with Cancer Inova Schar Cancer Institute Fairfax, VA, USA Jane Turner, MBBS, PhD, FRANZCP Professor Department of Psychiatry The University of Queensland Brisbane, Australia Lisa Tussing-Humphreys, PhD, MS, RD Associate Professor Department of Medicine Co-leader, Cancer Prevention and Control Program, Cancer Center University of Illinois Chicago, IL, USA Susan T. Vadaparampil, PhD, MPH Associate Center Director, Community Outreach, Engagement, and Equity Department of Health Outcomes and Behavior Moffitt Cancer Center Tampa, FL, USA Annalyn Valdez-Dadia, DrPH, MPH Assistant Professor Department of Human Services California State University, Dominguez Hills Carson, CA, USA Alan D. Valentine, MD Professor and Chair Department of Psychiatry MD Anderson Cancer Center The University of Texas Houston, TX, USA Sally W. Vernon, MA, PhD Professor Department of Health Promotion and Behavioral Science University of Texas School of Public Health Houston, TX, USA Froukje de Vries, MD, PhD Psychiatrist Department of Supportive Care Princess Margaret Cancer Centre, University Health Network Toronto, ON, Canada Claire E. Wakefield, BPsych(Hons), MPH, PhD Professor School of Women’s and Children’s Health and Kids Cancer Centre UNSW Sydney and Sydney Children’s Hospital North Willoughby, NSW, Australia Leah E. Walsh, MS Doctoral Student Department of Psychology Fordham University The Bronx, NY, USA xxv xxvi Contributors Linda D. L. Wang, PhD Joseph T. Wu, PhD, BS Associate Professor Department of Clinical Medicine Medical College of Yangzhou University Yangzhou, Jiangsu, China Professor School of Public Health The University of Hong Kong Hong Kong, China Maggie Watson, BSc, Dip Clin Psych, PhD Robert Zachariae, DMSc Honorary Professor Research Department of Clinical, Educational and Health Psychology University College London Institute of Cancer Research London, UK Professor Unit for Psycho-oncology and Health Psychology Aarhus University Aarhus, Denmark Amanda Watsula-Morley, MA Clinical Research Supervisor Department of Psychiatry and Behavioral Sciences Memorial Sloan Kettering Cancer Center New York, NY, USA Assistant Attending Psychologist Psychiatry Service Department of Psychiatry and Behavioral Sciences Memorial Sloan Kettering Cancer Center New York, NY, USA Jason A. Webb, MD, FAPA, FAAHPM Alexandra K. Zaleta, PhD Director of Education, Duke Center for Palliative Care Associate Professor Department of Medicine, Department of Psychiatry and Behavioral Sciences Duke University Durham, NC, USA Senior Director of Research Department of Research and Training Institute Cancer Support Community Philadelphia, PA, USA Lori Wiener, PhD, DCSW, FAPOS Bradley J. Zebrack, MSW, MPH, PhD Co-Director, Behavioral Health Care, Director, Psychosocial Support and Research Program Department of Pediatric Oncology Branch, National Cancer Institute, Center for Cancer Research National Institutes of Health Bethesda, MD, USA Professor School of Social Work University of Michigan Ann Arbor, MI, USA Irene O. L. Wong, BSc, MPhil, MMedSci, PhD Scientific Officer School of Public Health The University of Hong Kong Hong Kong, China Talia I. Zaider, PhD Introduction Our Past, Our Future—New Frontiers in Psycho-Oncology William S. Breitbart (Senior Editor) Introduction This textbook, Psycho-Oncology, 4th edition, is the first edition of this series of textbooks, which have defined the field of psycho-oncology, to be edited without Jimmie C. Holland as the senior editor. Jimmie’s imprint on these textbooks and on our international field has resulted in these textbooks often being referred to as the “Holland Textbook of Psycho-Oncology”—that is, when it isn’t, perhaps somewhat sacrilegiously, being referred to as the “Bible of Psycho-Oncology.” Hence, those of us who have taken on the both sacred and significant responsibility of editing this 4th edition of Psycho-Oncology have done so with a sense of both sadness and honor. The field of psycho-oncology is truly interdisciplinary and international, and so it is fitting that the editors and contributors to this 4th edition are indeed truly international and represent multiple disciplines. As senior editor, I have been blessed to have the magnificent talents of the following luminaries in our field as associate editors: Phyllis N. Butow, BA(Hons), DipEd, MClinPsych, MPH, PhD, of the University of Sydney; Paul B. Jacobsen, PhD, of the U.S. National Cancer Institute; Wendy W. T. Lam, RN, PhD, FFPH, of the University of Hong Kong; Mark Lazenby, APRN, PhD, of the University of Connecticut School of Nursing; and Matthew J. Loscalzo, MSW, LCSW, FAPOS, of the City of Hope. Their contributions to the quality of this textbook will be quite evident to the reader. Each of the editors of this textbook has had a direct and often life-changing experience through a long-standing relationship with Jimmie Holland, and our dedication to preserving her legacy is on every page of this text. I do want to take a moment to personally thank Andrea Knobloch, our editor at Oxford University Press, and her team; the personal assistants of many of the editors and contributors; and most significantly Laurie Schulman, who served as managing editor of this textbook and dedicated more than a year of time, energy, and devotion to assisting me in seeing this project through—even through the interruptions of the coronavirus pandemic. Lastly, our deep appreciation goes to the International Psycho-Oncology Society (IPOS), the American Psychosocial Oncology Society (APOS), and the several hundred authors who contributed more than 100 chapters to this textbook. Our Past Jimmie C. Holland, or “Jimmie,” as the world of psycho-oncology knew her, is often credited as being the founder or “mother” of psycho- oncology. Interestingly, Jimmie hated being called the “mother” of psycho-oncology. It particularly irked her when, as she got older, this term changed to “grandmother” of psycho-oncology. Jimmie much preferred these titles/roles to be reserved for her personal life—her family, her beloved children and grandchildren. But after all, she was a “Cicely Saunders–type” figure in a new movement within oncology, and so she was revered and mythologized in ways that did not make her comfortable. As many who knew Jimmie will attest, she was in fact somewhat shy and quite humble, quick to give credit to others, and eager to accelerate the career trajectories of the next generations of clinicians, scientists, and leaders of our field. That isn’t to imply that she didn’t have very strong opinions or have enviable political skills, but she clearly used those skills for the benefit of patients, the field, and the movement. Jimmie and I would often sit in her office at the end of the day and discuss just about everything one can imagine related to work, projects, our field, and politics (and some gossip). There was nothing off limits. Interestingly, we did frequently talk about the origins of psycho-oncology and individuals she felt never quite received enough recognition for their roles in the creation of our field. That is, in part, why she urged the creation of the IPOS Arthur Sutherland Award. Jimmie always felt that she had received too much credit for founding the field of psycho-oncology and was eager for there to be recognition of such pioneers in our field as Margit Von Kerekjarto, Robert Zitoun, Hiroomi Kawano, Steven Greer, Lea Baider, Peter Maguire, Bernard Fox, Juan Ignacio Romero, Avery Weissman, Edwin Cassem, Noemi Fisman, Maria Margarida Carvalho, Morton Bard, and Arthur Sutherland himself. One early pioneer has remained quite obscure and underrecognized for over 50 years, and Jimmie and I became quite fascinated with this figure. I became interested because he, like my parents, was a Holocaust survivor. Jimmie, I believe, became fascinated because he was also someone who had overcome severe physical disabilities (reminding Jimmie of her early days working with polio patients). Loma Feigenberg was a physician at the Karolinska Institute in Stockholm. In the 1950s, as an oncologist and radiotherapist, he noted the lack of attention in addressing the psychological responses of patients with advanced cancer. He later studied psychiatry and began to work with cancer patients at the Karolinska. He made what he called a “friendship contract” with them in which he agreed to “confidentiality” of the content of their “sessions” and made a commitment to ongoing care and support during their cancer treatment. His book, Terminal Care: A Method for Psychological Care 2 Psycho-Oncology of Dying Patients, is a landmark text.1 Feigenberg did finally receive the Distinguished Life Service Award from the IPOS in 1987. He also founded the International Work Group for Death, Dying and Bereavement (IWG), an early beginning of thanatology. However, Dr. Holland’s efforts over the last 43 years, since becoming the founding chief of the Psychiatry Service at Memorial Sloan Kettering Cancer Center, have indeed brought her well- deserved recognition as the founder and past leader of the field of psycho-oncology. Having founded the IPOS in 1984 and the APOS in 1986, Dr. Holland went on to edit the first major textbooks of psycho- oncology for our field. This textbook, Psycho-Oncology, 4th edition, was preceded by four major textbooks that defined our field. In 1989, Dr. Holland edited the Handbook of Psychooncology: Psychological Care of the Patient with Cancer, the first major textbook in our field.2 This landmark book (coedited with Julia Rowland, PhD) established our “new” field and virtually named the field “psycho-oncology.” The follow-up textbook Psycho-Oncology was published in 1998 and represented the most comprehensive, multidisciplinary, and international encyclopedia of a field entering its adolescence.3 The year 2010 saw the publication of the 2nd edition,4 followed by the 3rd edition5 in 2015, both published by Oxford University Press in collaboration with the IPOS and APOS. The field of psycho-oncology was now mature, rich, and filled with talented, creative, and innovative clinicians, scientists, advocates, and global leaders like Maggie Watson, Luigi Grassi, Uwe Koch, David Kissane, Christoffer Johansen, Luzia Travado, Barry Bultz, Maria Die Trill, Gary Rodin, Cristina Bolund, Bill Redd, Anja Mehnert, Francisco Gil, David Spiegel, Joan Bloom, Harvey Chochinov, Barbara Andersen, Jamie Ostroff, Phyllis Butow, Paul Jacobsen, Richard Fielding, Matt Loscalzo, Leslie Fallowfield, Pierre Gagnon, Jeff Dunn, Mitch Golant, Mary Jane Esplen, Sharon Manne, Jane Turner, David Cella, Elisabeth Andritsch, Pat Fobair, Irma Verdonck-de Leeuw, Michael Antoni, James Zabora, and numerous others (apologies to anyone who deserved mention and was omitted unintentionally; noninclusion in this list means you’re not an old-timer and are part of the new wave, the vital leaders of the future of our field). With the publication of this textbook, Psycho-Oncology, 4th edition, we take this moment to both look to our past and start to examine our future as a field. We have a rich legacy given to us by so many of the pioneers of psycho-oncology mentioned earlier. In fact, this textbook is dedicated to the memory of Jimmie C. Holland and we honor her and all the past editors and contributors to the prior editions of this text by moving forward with the creation of what we hope readers will someday view as a milestone textbook itself. Of note, two former associate editors of several of the prior editions of this series of textbooks died in 2019 as this 4th edition was being prepared. We are indebted to and honor the contributions and lives of Ruth McCorkle and Marguerite Lederberg— two remarkable women who were cherished by so many of us, worldwide. Ruth was an editor of several editions of the textbook and so we also dedicate this textbook in her honor as well as in Jimmie’s. Our Future We, the editors of this 4th edition of Psycho-Oncology, undertook a careful examination of the content of the 3rd edition of Psycho- Oncology, as well as the expert authors who contributed their expertise. We were particularly interested in two aspects of the purpose of the Psycho-Oncology textbook’s purpose: (1) to serve as the source textbook that provided the broadest and most multidisciplinary and essential science and practice of the field of psycho-oncology, and (2) to bring to our field the newest and latest innovations and cutting-edge research and clinical practice that would equip our readers with the knowledge and resources to be knowledgeable and to participate in the “new frontiers of psycho- oncology.” We feel we have accomplished this delicate but critical balance in the 4th edition of Psycho-Oncology. We’ve maintained many of the basic but critical aspects of prevention, screening, assessment, and management of basic common psychosocial and psychiatric issues in psycho-oncology, including cancer site–specific psychosocial issues and management. As much as possible, these cancer site–specific chapters also include some basic, updated oncological diagnostic and treatment-related information that is vital for clinicians and clinical researchers in our field. There are, however, a number of new sections that represent new developments in basic psycho-oncology science, breakthroughs in health care delivery, growth in treating special cancer populations, and innovative and novel evidence-based interventions that are changing the landscape of treatment, and a growing international perspective that our field has developed over recent years. Allow me to briefly highlight some of the updates and new sections in the 4th edition of Psycho-Oncology that are designed to prepare psycho-oncologists for the “new frontiers of psycho-oncology”: 1. Evidence-Based Interventions: We have dramatically expanded this section of the textbook and now include a variety of innovative novel interventions with a significant evidence base for efficacy. We’ve divided the interventions into models of care delivery and phases of illness. Models of Care Delivery now includes the following: (a) Collaborative and Integrated Models of Psychosocial Oncology Care, Community- Based Care, and Implementation Science’s Role in Care Delivery (b) Family and Couples Interventions (c) Interventions at various stages of illness including Active Treatment, Advanced Disease, and Survivorship, as well as novel interventions including Cognitive- Behavioral Interventions, Mindfulness-Based Interventions, Acceptance and Commitment Therapy, Interpersonal Therapy, Supportive- Expressive Psychotherapy, and Meaning- Centered Psychotherapy for advanced cancer patients, for bereavement, survivors, and for caregivers, in addition to CALM Therapy, Dignity Therapy, Emotionally Focused Therapy, Metacognitive Approaches, Integrative Oncology Interventions, and Physical Activity Interventions. We had hoped to include Light Therapy, but that was not possible. 2. Digital Health Interventions: We have an expanded section on e-health intervention delivery, which ranges from prevention, smoking cessation, and psychosocial distress to Physical Symptom Control. 3. Biobehavioral Psycho-Oncology: We have included the first ever section on the science of stress and cancer risk and progression. We have wonderful contributions from Mike Antoni and coauthors of Stress Processes and Cancer Progression; Depression, Inflammation, and Cancer from Andrew Miller and Our Past, Our Future 4. 5. 6. 7. 8. 9. 10. coauthors; and Biobehavioral Psycho-Oncology Interventions from Michael Hoyt and Frank Penedo. This somewhat controversial area of psycho-oncology research has now reached a level of maturity and there are evidence-based findings of which all psycho-oncologists must be aware. Geriatric Oncology: This is a growing field in psycho-oncology. This section includes chapters on screening, assessment, interventions, and communications issues specific to managing older cancer patients. Christian Nelson, Andrew Roth, Kelly Trevino, Patricia Parker, Beatriz Korc-Grodzicki, and Yesne Alici were the primary contributors to this section. Their contributions acknowledge the pioneering work of our late friend and colleague Arti Hurria. Pediatric Psycho-Oncology: For the very first time, Pediatric Psycho-Oncology is fully included and represented in the Psycho-Oncology series of textbooks. This section has chapters on pediatric psycho-oncology screening and assessment, management of common psychiatric disorders, evidence- based interventions in pediatric psycho-oncology, and adolescent and young adults with cancer. The contributors to these sections include the leaders of the field—Anne Kazak, Maryland Pao, Julia Kearney, Lori Wiener, Anna Muriel, and Bradley Zebrack. Survivorship: This section has been expanded and has interesting new information on approaches to Fear of Recurrence in Cancer Survivors. Palliative Care and Advanced Planning: These chapters focus on the need to focus on treatment decision making; discussion of advance care planning and care goals at the time of diagnosis—early in the course of life-threatening cancer; and prognostic awareness and the role of the psycho-oncologist in palliative care. The interface of psycho-oncology and palliative care is a critically important one that needs to be navigated with a sense of collaboration and integration. Michael Diefenbach, Stefanie Mooney, Scott Irwin, Barry Rosenfeld, and Allison Applebaum and their coauthors have contributed outstanding chapters. Diversities in the Experience of Cancer: This expanded new section addresses the important issues of cancer and culture; cancer disparities; access to care and food; financial and housing insecurities; cancer and sexual minorities; and the experience of cancer as an immigrant. The contributors to this section include leaders in these areas such as Marjorie Kagawa- Singer, Francesca Gany, Victoria Blinder, Jennifer Leng, and Charles Kamen. Behavioral and Psychological Factors in Cancer Risk; Screening for Cancer in Normal and At-Risk Populations: These sections have been expanded and include a broad international perspective. Contributors include many luminaries such as Christoffer Johansen, Jamie Ostroff, Richard Fielding, Jennifer Hay, Gabriel Leung, and many others. Screening and Testing for Germ Line and Somatic Mutations: With the advent of precision oncology and therapies targeted at actionable tumor mutations, psycho-oncologists have had to learn a great deal about genetics, and now we have begun to explore the various psychosocial sequelae and the need for counseling presented by this revolution in medical oncology. This section has chapters on genetic testing in breast and ovarian cancer, testing in hereditary cancers, genomic testing for targeted therapies, and psychosocial issues related to large- scale liquid biopsy screening for mutations in normal and at- risk populations. Mary Jane Esplen, Susan Peterson, Megan Best, Jada Hamilton, and their coauthors have contributed outstanding chapters. 11. Screening and Assessment in Psychosocial Oncology: We’ve experienced a revolution in screening and brief assessments of patients at risk for distress, anxiety, depression, delirium and cognitive disorders, suicidal ideation, and uncontrolled symptoms. This section addresses many of these issues. Paul Jacobsen, Kristine Donovan, Alex Mitchell, Tim Ahles, James Root, Bill Breitbart, Yesne Alici, and their colleagues have contributed outstanding chapters. 12. Building Supportive Care Teams; Psycho-Oncology in Health Policy: These sections are expanded and have a broad international perspective. Informed by Our Past, Inspired to Create a Better Future We have a great legacy. That is the gift our field has received from its pioneers. We human beings engage in what is termed “cumulative learning.” We build upon the wisdom and knowledge chronicled by those who came before us. Einstein’s work on gravity could not have taken place without building upon our knowledge of the chronicled work of Newton. We are building upon the knowledge accumulated and documented by psycho-oncologists who dedicated their work to establishing and growing a base of research and clinical innovation over the last 50 years. It is our responsibility to contribute to this “cumulative” knowledge base and move our field forward to better “care for the whole person with cancer.” Our hope is that you, the readers of the 4th edition of Psycho-Oncology, feel we have made a valuable contribution to fulfilling the solemn responsibility we have inherited: to create a better future. William S. Breitbart, MD, FAPOS Senior Editor Psycho-Oncology, 4th edition REFERENCES 1. Feigenberg L. Terminal care: Friendship contracts with dying cancer patients. New York, Brunner/Mazel, 1980. 2. Holland JC, Rowland J, Eds: Handbook of Psychooncology. New York, Oxford University Press, 1989. 3. Holland JC, Breitbart WS, Jacobsen PB, Lederberg MS, Loscalzo M, Massie MJ, McCorkle R, Eds: Psycholo-Oncology. New York, Oxford University Press, 1998. 4. Holland JC, Breitbart WS, Jacobsen PB, Lederberg MS, Loscalzo M, McCorkle R, Eds: Psycho-Oncology 2nd Edition. New York, Oxford University Press, 2010. 5. Holland JC, Breitbart WS, Butow PN, Jacobsen PB, Lederberg MS, Loscalzo M, McCorkle R, Eds: Psycho-Oncology 3rd Edition. New York, Oxford University Press, 2015. 3 SECTION I Behavioral and Psychological Factors in Cancer Risk and Prevention Paul B. Jacobsen (Section Editor) 1 Tobacco Use and Cessation 7 4 Thomas H. Brandon, Vani N. Simmons, Úrsula Martínez, and Patricia Calixte-Civil 2 Diet and Cancer 13 Marian L. Fitzgibbon, Lisa Tussing-Humphreys, Angela Kong, and Alexis Bains 3 Physical Activity, Sedentary Behavior, and Cancer 21 Christine M. Friedenreich, Chelsea R. Stone, and Jessica McNeil Sun Exposure and Cancer Risk 30 Suzanne J. Dobbinson, Afaf Girgis, Bruce K. Armstrong, and Anne E. Cust 5 Psychosocial Factors 36 Anika von Heymann and Christoffer Johansen 6 Viral Cancers and Behavior 43 Susan T. Vadaparampil, Lindsay N. Fuzzell, Shannon M. Christy, Monica L. Kasting, Julie Rathwell, and Anna E. Coghill 1 Tobacco Use and Cessation Thomas H. Brandon, Vani N. Simmons, Úrsula Martínez, and Patricia Calixte-Civil Tobacco and Cancer Each year over 7 million people die worldwide due to tobacco use.1 Smoking is responsible for about 22% of all cancer deaths globally. It accounts for over two-thirds of all lung cancer mortalities and contributes significantly to mortality rates for oral cancer, as well as bladder, stomach, liver, pancreas, kidney, cervical, and colorectal cancers.2 In addition to cancer, smoking contributes to coronary heart disease, chronic obstructive pulmonary disease, cardiovascular disease, stroke, and peptic ulcers. Indeed, about 20% of all deaths in the United States can be attributed to smoking. From the individual perspective, a given smoker has about a 50% chance of dying from smoking, with the average smoker living 10 years less than a nonsmoker.3 Although there are risks associated with noncombustible tobacco use (e.g., chew, snuff), this chapter focuses on combustible tobacco use (i.e., smoking) because of its much higher prevalence and relative risk. Nevertheless, patients should be advised to cease all forms of tobacco use. Smoking cessation is associated with decreased mortality and morbidity from cancer and other diseases. Stopping smoking at age 30 restores nine years of life expectancy, whereas stopping at age 60 still restores an expected three years of life, compared to continuing to smoke.3 Thus, great potential for cancer prevention lies with long- term cessation of smoking. In this chapter, we begin by noting the changing demographic profile of current smokers. We then review the evidence-based treatments for tobacco use and dependence, emphasizing primarily qualitative and meta-analytic reviews. We draw upon the 2008 update of the U.S. Public Health Service’s Clinical Practice Guideline, Treating Tobacco Use and Dependence,4 which is based on a review of 8,700 research articles, with treatment recommendations derived from meta-analyses of most treatment modalities. We then discuss special issues of relevance for treating cancer patients. New to this update is a discussion of electronic cigarettes (e-cigarettes), the use of which has grown exponentially over the past decade, including among cancer patients. Evolving Landscape of Tobacco and Nicotine Use Changing Demographics of Tobacco Users Several countries have implemented strategies toward reducing tobacco burden, such as monitoring tobacco use, offering access to smoking cessation interventions, increasing tobacco taxes, or introducing public warnings about the dangers of tobacco use. Although these actions have led to a global reduction in tobacco use in most developed countries, these nations still account for half of all female daily smokers and roughly 75% of male daily smokers. Efforts to reduce tobacco use have been successful in developed countries, and they should be bolstered in developing countries (i.e., countries with historically lower education, per capita income, and life expectancy). Developing countries have shown moderate smoking prevalence across time, but many of these nations are showing increases in the prevalence of smoking and smoking-related mortality that are commensurate with increases in income, decreases in the cost of tobacco, heavier marketing from the tobacco industry, and limited tobacco-related public health policies. As such, developing countries are vulnerable to assuming the burden of the tobacco epidemic.1 Moreover, tobacco use burden is disproportionately affecting certain segments of the population worldwide. For example, tobacco use is rapidly increasing among youth, especially females, in developing countries, and it is highly prevalent among individuals with low income across nations, regardless of per capita gross national income.5 In the U.S., since the seminal 1964 U.S. Surgeon General’s report, the prevalence of smoking among adults has dramatically declined from nearly half of adults to less than one in five.6 The current demographic profile of smokers is markedly different than decades ago because the reduction in smoking prevalence has not been consistent across demographic groups. These differences in prevalence of tobacco use are associated with differential burdens of tobacco- related morbidity and mortality. In particular, a substantial body of evidence demonstrates that lower educational attainment, being below the poverty level, identifying as American Indian/Alaska Native, living in the Midwest or South, working in a blue-collar or service industry, having active military or veteran status, having a disability, having a severe mental illness, and not having health insurance are associated with higher prevalence of smoking in the U.S. There are also more recent changes in the demographics of tobacco users that coincide with shifts in the racial/ethnic composition of the U.S. and/or more inclusive data collection. That is, subgroups who identify as more than one race/ethnicity, as sexual or gender minorities, or as immigrants report greater tobacco use. Regarding race/ethnicity, although Hispanics and non-Hispanic Asians have 8 SECTION I Factors in Cancer Risk and Prevention among the lowest smoking prevalence by race/ethnicity (12.7% and 8%, respectively), there is wide variation in smoking behavior within the subgroups and across gender. Among foreign-born men living in the U.S., 24.8% of Mexicans, 47.7% of Filipinos, and 52.7% of Chinese people reported being current smokers, which is of particular relevance to healthcare in the United States given that Mexico, the Philippines, and China represent three of the top five countries with the largest populations of foreign-born individuals in the U.S. In 2010 alone, 29.3% of all immigrants living in the U.S. were from Mexico. Thus, the distribution of tobacco use and its consequent health and economic burdens are unequal and shifting, requiring attention by both researchers and clinicians. The Emergence of Electronic Cigarettes E-cigarette use has grown dramatically in the last 10 years. E- cigarettes include a battery and heating element that aerosolizes a liquid that typically contains nicotine, flavorants, propylene glycol, and vegetable glycerin. Since their introduction, the available products have expanded and evolved in terms of their ease of use, their sophistication, and their efficiency of nicotine delivery. The newest devices deliver a nicotine dose similar to a combustible cigarette, while also simulating the sensorimotor aspects of smoking (e.g., hand and arm movements, puffing and inhalation behavior, and visible exhalation). Theoretically, these similarities should ease the transition from combustible cigarettes to e-cigarettes. Although there have been regulatory barriers to conducting randomized controlled trials of e-cigarettes for smoking cessation, evidence of their efficacy is now emerging.7 However, e-cigarettes have generated a magnitude of controversy and division never before seen in the tobacco control and research fields. The current scientific consensus is that e-cigarettes are substantially less harmful than combustible cigarettes,8 and therefore complete switching from smoking to “vaping” represents significant harm reduction at the individual and population levels. However, there is growing concern about the recent uptake of vaping by youth and the unknown long-term health outcomes of this behavior. The primary public health challenge related to tobacco use is to develop policy that promotes switching by current smokers while minimizing uptake of vaping by youth who would not have otherwise used nicotine products. Treatment of Tobacco Use and Dependence Tobacco dependence has multiple motivational influences within and across individual smokers.9 Among these are physical dependence on nicotine, operant and classical conditioning processes, environmental and social factors, cognitive expectancies about the benefits of smoking, and desire for weight control. Given the complexity of the factors influencing smoking, it is not surprising that single-treatment approaches have limited success, with the best long-term outcomes obtained from multimodal treatments. In this section, we review pharmacological interventions, followed by social/behavioral interventions, broadly defined, and finally discuss combination treatments. Pharmacotherapy Currently, there are seven pharmacotherapies approved by the U.S. Food and Drug Administration (FDA) for smoking cessation. All of these medications have been found to approximately double the odds of long-term abstinence (with one, varenicline, tripling the odds), and the Clinical Practice Guideline issued by the U.S. Department of Health and Human Services recommends that pharmacotherapy be routinely offered to smokers attempting to quit.4 Nicotine Replacement Therapies Nicotine replacement therapy (NRT) aids smoking cessation by partially replacing plasma nicotine levels, thereby reducing symptoms of nicotine withdrawal (e.g., craving, depression, irritability, difficulty concentrating) and possibly reducing the reinforcement derived from any cigarettes smoked. Five types of NRT have FDA approval: chewing gum, transdermal patch, intranasal spray, inhaler device, and lozenge. In general, NRT is used during the first 8–12 weeks of abstinence, when nicotine withdrawal symptoms are greatest. Of the five NRT delivery methods, the nicotine nasal spray reaches its peak concentration most rapidly, whereas the transdermal patch provides the slowest, but most consistent, serum nicotine levels over the course of a day. Meta-analyses indicate roughly equivalent efficacies for the five NRT products, with odds ratios ranging from 1.5 (for nicotine gum) to 2.3 (for nasal spray) compared to placebo.4 Estimated six-month abstinence rates are approximately 20%–25%. Each product is associated with specific contraindications and cautions, primarily related to its particular mode of drug delivery. Because NRT delivers nicotine without the harmful byproducts of smoked tobacco, it is considered a far safer alternative to smoking. The safety of NRT during pregnancy has not been established. Bupropion SR (Zyban®) Bupropion was the first non-nicotine medication to be approved by the FDA for treating tobacco dependence. Also marketed as an atypical antidepressant (Wellbutrin®), bupropion doubles tobacco abstinence rates compared to placebo, with an average odds ratio of 2.0, and an abstinence rate of approximately 24%.4 It attenuates nicotine withdrawal and cigarette cravings, and can reduce postcessation weight gain. Bupropion’s mechanism of action is not fully understood, but it appears to inhibit the neuronal reuptake of dopamine and norepinephrine—key neurotransmitters in the maintenance of nicotine dependence. It may also have antagonistic effects on nicotinic receptors, attenuating perceived satisfaction from smoking. To reach steady-state blood levels before quitting smoking, the smoker should begin using bupropion SR one week before the target quit date. Contraindications include a history of seizure disorders or factors known to increase the risk of seizures (e.g., bulimia or anorexia nervosa, serious head trauma, alcoholism) and concomitant use of monoamine oxidase (MAO) inhibitors. Because of postmarketing reports of neuropsychiatric adverse events, including suicidality, the FDA required “black box” warnings on both bupropion and varenicline (see later) with respect to possible neuropsychiatric adverse events, including depression, psychosis, aggression, agitation, and anxiety, as well as suicidal ideation or behavior. Although the warning remains, the black box was rescinded in 2016 following additional research that failed to find elevated neuropsychiatric events for varenicline or bupropion compared to NRT or placebo. The safety of bupropion during pregnancy has not been established. CHAPTER 1 Tobacco Use and Cessation Varenicline (Chantix®) Varenicline was the last pharmacotherapy approved for treating nicotine dependence. It is an orally administered partial agonist of α4β2 nicotinic acetylcholine receptors (nAChRs). Varenicline appears to reduce nicotine cravings and withdrawal symptoms, and its agonistic properties appear to attenuate the reinforcing effects of smoking, including perceived satisfaction.10 Similar to bupropion, varenicline use should be initiated one week before the target quit date. Evidence suggests that it has outperformed bupropion in head-to-head studies and is the most effective of the smoking cessation medications, with an average odds ratio of 3.1, producing 33% abstinence.4 The main adverse effect of varenicline is mild to moderate nausea. However, as with bupropion, warnings of neuropsychiatric adverse events are also included in labeling. In addition, there is some evidence that varenicline may increase the risk of major cardiovascular events. Varenicline is not approved for use with pregnant women. Combination Pharmacotherapies Recent research has tested the efficacy of combining different forms of pharmacotherapy. The general model has been to combine a long- acting, relatively stable medication, such as the nicotine patch, with a shorter-acting medication that can be used ad libitum. In this manner, both tonic and phasic nicotine cravings and withdrawal symptoms can be addressed. The combination of nicotine patch with gum, nasal spray, or inhaler has evidence of significant efficacy, as does the combination of the patch and bupropion SR.4 Social/Behavioral Treatments The nonpharmacological therapies described in this section span a wide range of intensity and duration, from minimal self-help interventions to intensive individual counseling. Clinicians should be aware of the availability of these options and should be willing to refer patients for services that they are unable to provide themselves. Self-help Self-help refers to materials that can be provided to smokers, such as pamphlets, booklets, or audiovisual media. Their primary advantages are low cost and ease of distribution. Unfortunately, the efficacy of self-help materials appears to be quite limited, with improved cessation rates of about 1% compared to no-treatment controls.4 However, a self-help intervention that extends over time (i.e., distribution of sets of materials over 12–18 months) has recently demonstrated long-term efficacy.10 Telephone Quitlines Smoking cessation quitlines are available throughout the United States and most of the world. In the United States one number (1-800-QUIT-NOW) serves as a central access point that automatically routes calls to the appropriate state or federal quitline service. Approximately 400,000 smokers in the United States are served annually by state quitlines, with an average utilization rate of about 1%.11 Quitline services differ in the amount and frequency of counseling offered, the provision of ancillary materials, referrals to local smoking cessation agencies, the provision of free or subsidized pharmacotherapies, and whether calls are proactive (call-out), reactive (call-in), or both. Quitlines have the advantage of providing more personal and intensive help than self-help materials, while also having greater potential reach than face-to-face counseling. Meta- analyses show that quitlines are effective, with overall odds ratios of 1.4–1.6 compared to control conditions, which translates into differential long-term abstinence rates of at least 3%–5%.4 Brief Interventions Healthcare providers have the opportunity to deliver relatively brief face-to-face interventions. The U.S. Public Health Service (PHS) Clinical Practice Guideline describes an effective brief smoking cessation intervention model most commonly referred to as the “5’A’s.”4 The five key steps include (1) “Asking” every patient about tobacco use at repeated visits, (2) “Advising” every tobacco user to quit by providing clear and personalized advice to quit, (3) “Assessing” the willingness of patients to quit, (4) “Assisting” patients with quitting, and (5) “Arranging” follow-up cessation support, ideally within a few weeks of the quit attempt. Meta-analyses have indicated that physician advice alone increases abstinence rates by approximately 2.3%–2.5%.4 Because 70% of smokers visit their physician each year, the potential cumulative effect of even this small effect is sizable. Moreover, there is a dose–response relationship between contact time and abstinence outcomes, with minimal counseling (< 3 minutes) yielding 13.4% abstinence, low-intensity counseling (3–10 minutes) yielding 16.0% abstinence, and higher-intensity counseling (> 10 minutes) yielding 22.1% abstinence. Abstinence rates also increase with the number of counseling meetings and/or the number of clinician types delivering the cessation messages.4 Alternative models that reduce provider burden include Ask-Advise-Refer (AAR) and Ask-Advise-Connect (AAC). In both abbreviated models, patients are asked about their smoking and are delivered brief advice to quit. However, in the AAR model, patients are then referred to evidence- based cessation programs for assistance in quitting (e.g., a quitline). Designed to overcome patient barriers that exist with use of a passive referral model, the AAC model directly connects patients to the smoking cessation resource via an automated connection system within the electronic health record (EHR). The AAC method has demonstrated greater impact over the AAR model with respect to a higher proportion of smokers enrolling in treatment.12 Intensive Interventions The most intensive interventions tend to be multisession treatments typically offered through smoking cessation clinics, in either group or individual formats. Of the empirically supported intensive interventions, the most common approach is cognitive-behavioral counseling. Key elements of this approach include patient education regarding tobacco dependence and withdrawal, advice for coping with withdrawal symptoms, identifying high-risk situations (“triggers”) that produce urges to smoke, teaching and practicing cognitive and behavioral responses for coping with urges, discussion of long-term risk factors such as depression and weight gain, and discussion of how to respond in the event of an initial “slip” or “lapse.” It usually involves multiple sessions over several weeks and may begin before the target quit date. Counseling has been found to be effective, with an odds ratio of 1.5 compared to no counseling and an average abstinence rate of 16.2% compared to 11.2%.4 In addition to counseling, the guideline also found evidence for intratreatment social support, and it therefore recommends providing support and encouragement as part of treatment. 9 10 SECTION I Factors in Cancer Risk and Prevention Combining Counseling and Pharmacotherapy A key conclusion of the most recent guideline is that the combination of counseling and medication is more effective than either alone in producing long-term tobacco abstinence. Moreover, as noted earlier, higher abstinence rates tend to be produced with more intensive counseling. Thus, the guideline meta-analysis produced an estimated abstinence rate of approximately 33% when medication was combined with nine or more sessions of counseling, compared to 22% when no more than one counseling session was provided. Conversely, the guideline reported an odds ratio of 1.7 for the combination of medication and counseling, compared to counseling alone.4 Counseling and medication appear to provide complementary benefits. Whereas medication reduces withdrawal symptoms and craving, counseling can teach cognitive and behavioral coping strategies and can provide valuable social support. Therefore, whenever medication is recommended or provided to patients, they should also be offered counseling. Special Issues with Cancer Patients There is a growing body of evidence that smoking following cancer diagnosis has a negative impact on cancer treatment efficacy, treatment-related complications and side effects, cancer recurrence and second malignancies, and overall survival.13 With advances in cancer treatments, the number of cancer survivors is significantly increasing, emphasizing the importance of improving health outcomes and quality of life within this high-risk population. In this section, we will describe the benefits of smoking cessation in cancer patients, review cessation and relapse rates among cancer patients, and summarize the current knowledge regarding cessation interventions for cancer patients. Benefits of Quitting Smoking The last report of the U.S. Surgeon General concluded that continued smoking after cancer diagnosis is causally related to multiple negative consequences, including increased risk of cancer-specific mortality as well as all-cause mortality. Furthermore, persistent smoking after a cancer diagnosis has been strongly associated with cancer recurrence, poor treatment outcomes (e.g., poorer response to treatment, treatment-related toxicities), and higher risk of hospitalization.13 Quitting smoking is associated with fewer medical complications, decreased risk of subsequent malignancies, and increased survival rate.14 Finally, some research indicates that patients who remain smoke-free following cancer treatment report lower levels of depression and fatigue, improving overall quality of life relative to patients who continue to smoke.15 In summary, evidence is accumulating that smoking cessation after a cancer diagnosis improves quality of life, increases survival, and decreases cancer recurrence and psychological distress. Smoking Cessation and Relapse among Cancer Patients Despite the benefits of quitting, over 30% of cancer patients continue to smoke after diagnosis. However, cancer patients who smoke are highly motivated to quit, and many make an attempt to quit at the time of diagnosis.16 Because most quit attempts appear to occur at the time of diagnosis and treatment, the period between cancer diagnosis and end of treatment may represent the optimal window of opportunity for provision of smoking cessation interventions. There is less research on long-term abstinence rates among cancer patients. Estimates of smoking relapse range from 13% to 60%. Unlike the general population of smokers for whom relapse most often occurs within a week after cessation, the majority of relapses among cancer patients occurs within the first few months following a quit attempt, again reflecting the initial motivational impact of a cancer diagnosis. Predictors of both persisting smoking and relapse have included factors such as longer history of smoking, depression, lower desire to quit, and alcohol use.17 Interventions for Cancer Patients Few clinical trials have been conducted on smoking cessation interventions for cancer patients. Interventions tested have included a variety of formats, such as nurse-delivered inpatient counseling, cognitive-behavioral therapy, motivational interviewing, distribution of educational materials, and follow-up phone calls. Several studies have also tested pharmacological cessation treatments (nicotine replacement therapy, varenicline, or bupropion), either alone or combined with counseling. The overall findings have not demonstrated a significant treatment effect.18 More recently, some innovative interventions are being tested. System- based interventions aim at introducing changes in the overall organization to change smoking cessation practices (e.g., automatic referrals using EHRs). Also, interventions using mobile technology have been pilot tested. However, evidence on the effectiveness of these new types of interventions is still limited. Finally, the use of e-cigarettes has increased among cancer patients, paralleling trends in the general population.19 Overall, it seems that cancer patients hold generally positive expectancies regarding e-cigarettes, as compared to both combustible cigarettes and NRT, and find them an attractive way to quit smoking.20 The American Association for Cancer Research (AACR) and the American Society of Clinical Oncology (ASCO) recommend that healthcare providers encourage use of FDA-approved cessation methods, given the lack of definitive data regarding the safety and efficacy of e-cigarettes. However, the American Cancer Society’s 2018 position statement on e-cigarettes recommends harm reduction, including e-cigarettes, for patients who have not otherwise been able to quit smoking. When implementing smoking cessation interventions with cancer patients, clinicians should be mindful of several unique cancer-related issues. For instance, the delay in relapse among cancer patients described earlier may suggest a waning of motivation as patients physically recover and return to their prediagnosis lifestyles. Thus, smoking relapse prevention interventions may be particularly important as patients recover. Another issue relates to potential contraindications with the use of smoking cessation pharmacotherapy. With respect to NRT and e-cigarettes, for example, although nicotine is not itself carcinogenic, preclinical research suggests that it can accelerate tumor growth, inhibit apoptosis induced by several chemotherapy agents, and negatively impact response to radiotherapy. Nevertheless, there is no evidence to date indicating that NRT causes adverse events in cancer patients.14 In addition, NRTs such as nicotine gum, spray, inhaler, or lozenge may not be appropriate for individuals with oral cancers, whereas bupropion is contraindicated for patients with a history of central nervous system (CNS) tumors due to an increased risk of seizures. Hence, clinicians CHAPTER 1 Tobacco Use and Cessation must take extra care in selecting appropriate cessation medications that address cancer patients’ unique needs. Given the growing body of evidence demonstrating the substantial risks of continued smoking among cancer patients, it is not surprising that recognition and support of cessation services are increasing. For example, the ASCO developed updated tobacco guidelines that include recommendations for health professionals to assess tobacco use and integrate cessation services in the oncology setting.21 Similarly, a policy statement by the AACR called for improved documentation of tobacco use among patients, as well as improvements in evidence-based cessation assistance provided to all patients who use tobacco or have recently quit tobacco.22 The National Comprehensive Cancer Network (NCCN) has also developed clinical practice guidelines for smoking cessation that include a thorough assessment of tobacco use and supports the use of evidence-based methods of smoking cessation (i.e., combined pharmacologic and behavior therapy) for every cancer patient throughout the continuum of cancer care.23 Finally, the U.S. National Cancer Institute (NCI), in recognition of the need for system-level smoking cessation services, has provided competitive funding to NCI-designated cancer centers to develop comprehensive plans to identify and offer cessation services to every cancer patient who smokes, via enhanced EHR and other strategies.24 Improved comprehensive documentation of tobacco use would further clinical support for smoking cessation as well as facilitate research on the effects of tobacco on cancer-related outcomes and its impact on treatment response in clinical trials. With the aim of improving the assessment of tobacco use in the oncology patient population, the Cancer Patient Tobacco Use Questionnaire (C-TUQ) was developed.25 This tool comprises up to 22 items for standardized assessment of tobacco use among cancer patients. This is particularly important within clinical trials of cancer therapies, given that tobacco use is a treatment modifier. In summary, the importance of quitting smoking for all cancer patients is clear. Clinicians who treat cancer patients must capitalize on the window of opportunity during cancer diagnosis and treatment to identify smokers and make cessation interventions readily available to these high-risk patients. Cancer patients who stop smoking and remain abstinent after treatment are likely to reap significant benefits, including improved quality of life and prolonged survival. Conclusion Tobacco use by cancer patients appears to be influenced by the same range of biopsychosocial factors as it is in the general population. However, cancer diagnosis and treatment offer a unique and potentially powerful opportunity for healthcare providers to intervene by offering cessation advice and assistance. To date, there is little research to recommend specialized smoking cessation interventions for cancer patients above and beyond the general recommendations of the Clinical Practice Guidelines.7 However, it is likely that targeted treatments that capitalize on the teachable moment could be highly effective, and research has been increasing in this area. Meanwhile, the greatest progress in both cancer prevention and recovery depends on consistent action by all components of the healthcare system to promote tobacco cessation. This includes coverage of smoking cessation interventions by third-party payers, establishment of smoke-free campuses by hospital administrators, and strong cessation advice and assistance by every healthcare provider. Finally, the changing demographics of tobacco users along with evolving noncombustible alternatives to smoking require ongoing monitoring and the updating of policies and clinical practices, as needed. ACKNOWLEDGMENTS Preparation of this chapter was supported by National Cancer Institute grants R01 CA154596, R01 DA037961, R01 CA199143, and R03 CA227044. Disclosure: Dr. Brandon has received research support from Pfizer, Inc., and serves on the advisory board for Hava Health, Inc. REFERENCES 1. World Health Organization. WHO report on the global tobacco epidemic, 2019. Geneva: World Health Organization; 2019. 2. Jemal A, Torre L, Soerjomataram I, Bray F. The cancer atlas. Third Ed. Atlanta, GA: American Cancer Society; 2019. 3. IARC. Reversal of risk after quitting smoking. Lyon, France: IARC Handbooks of Cancer Prevention, Tobacco Control; 2007:11. 4. Fiore MC, Jaén CR, Baker TB, et al. Treating tobacco use and dependence: 2008 update. Clinical Practice Guideline. Rockville, MD: U.S. Department of Health and Human Services, Public Health Service; 2008. 5. Drope J, Schluger N, Cahn Z, et al. The tobacco atlas. Atlanta, GA: American Cancer Society and Vital Strategies; 2018. 6. U.S. National Cancer Institute. A socioecological approach to addressing tobacco- related health disparities. National Cancer Institute Tobacco Control Monograph 22. NIH Publication No. 17-CA-8035A. Bethesda, MD: U.S. Department of Health and Human Services, National Institutes of Health, National Cancer Institute; 2017. 7. Hajek P, Phillips-Waller A, Przulj D, et al. A randomized trial of e-cigarettes versus nicotine-replacement therapy. N Engl J Med. 2019;380(7):629–637. 8. National Academies of Sciences, Engineering, and Medicine. Public health consequences of e- cigarettes. Washington, DC: National Academies Press; 2018. 9. Baker TB, Brandon TH, Chassin L. Motivational influences on cigarette smoking. Annu Rev Psychol. 2004;55:463–491. 10. Brandon TH, Simmons VN, Sutton SK, et al. Extended self-help for smoking cessation: a randomized controlled trial. Am J Prev Med. 2016;51(1):54–62. 11. Cummins SE, Bailey L, Campbell S, Koon-Kirby C, Zhu SH. Tobacco cessation quitlines in North America: a descriptive study. Tob Control. 2007;16:9–15. 12. Vidrine JI, Shete S, Cao Y, et al. Ask-Advise-Connect: a new approach to smoking treatment delivery in health care settings. JAMA Intern Med. 2013;173(6):458–464. 13. U.S. Department of Health and Human Services. The health consequences of smoking—50 years of progress: A report of the Surgeon General. Atlanta, GA: U.S. Department of Health and Human Services, Centers for Disease Control and Prevention, National Center for Chronic Disease Prevention and Health Promotion, Office on Smoking and Health; 2014. 14. Warren GW, Sobus S, Gritz ER. The biological and clinical effects of smoking by patients with cancer and strategies to 11 12 SECTION I Factors in Cancer Risk and Prevention 15. 16. 17. 18. 19. 20. implement evidence- based tobacco cessation support. Lancet Oncol. 2014;15(12):e568–e580. Martínez Ú, Brandon KO, Sutton SK, Brandon TH, Simmons VN. Does smoking abstinence predict cancer patients’ quality of life over time? Psychooncology. 2019;28(8):1702–1711. Westmaas JL, Newton CC, Stevens VL, Flanders WD, Gapstur SM, Jacobs EJ. Does a recent cancer diagnosis predict smoking cessation? An analysis from a large prospective US cohort. J Clin Oncol. 2015;33(15):1647–1652. Chang EHE, Braith A, Hitsman B, Schnoll RA. Treating nicotine dependence and preventing smoking relapse in cancer patients. Expert Rev Qual Life Cancer Care. 2017;2(1):23–39. Sheeran P, Jones K, Avishai A, et al. What works in smoking cessation interventions for cancer survivors? A meta-analysis. Health Psychol. 2019;38(10):855–868. Sanford NN, Sher DJ, Xu X, Aizer AA, Mahal BA. Trends in smoking and e-cigarette use among US patients with cancer, 2014- 2017. JAMA Oncol. 2019;5(3):426–428. Correa JB, Brandon KO, Meltzer LR, et al. Electronic cigarette use among patients with cancer: reasons for use, 21. 22. 23. 24. 25. beliefs, and patient- provider communication. Psychooncology. 2018;27(7):1757–1764. Hanna N, Mulshine J, Wollins DS, Tyne C, Dresler C. Tobacco cessation and control a decade later: American Society of Clinical Oncology policy statement update. J Oncol Pract. 2013;31(25):3147–3157. Toll BA, Brandon TH, Gritz ER, et al. Assessing tobacco use by cancer patients and facilitating cessation: an American Association for Cancer Research policy statement. Clin Cancer Res. 2013;19:1941–1948. National Comprehensive Cancer Network. Smoking cessation guidelines, https://www.nccn.org/professionals/physician_gls/ pdf/smoking.pdf, 2017. Croyle RT, Morgan GD, Fiore MC. Addressing a core gap in cancer care—the NCI Moonshot Program to help oncology patients stop smoking. N Engl J Med. 2019;380(6):512–515. Land SR, Toll BA, Moinpour CM, Mitchell SA, et al. Research priorities, measures, and recommendations for assessment of tobacco use in clinical cancer research. Clin Cancer Res. 2016;22(8):1907–1913. 2 Diet and Cancer Marian L. Fitzgibbon, Lisa Tussing-Humphreys, Angela Kong, and Alexis Bains Overview Research over the past several decades shows that 95% of cancers can be attributed to environmental factors,1 including pollution, infections, radiation, and other external factors as well as tobacco use, alcohol, inactivity, diet, and other lifestyle factors.2 Diet, arguably among the most modifiable of these factors, likely contributes to the development of 30% to 35% of cancers. Substantial shifts in the food landscape in developed countries have contributed to changes in dietary intake, energy balance, increases in body fat, and the development of obesity. Obesity, defined as a body mass index (BMI) ≥ 30 kg/m², is associated with several cancers. Obesity exceeds 30% in both genders and is predicted to reach 51% by 2030 across all adult age groups in the United States (U.S.). Thus, the World Cancer Research Fund (WCRF), the American Institute for Cancer Research (AICR), the American Cancer Society (ACS), and cancer researchers both in the U.S. and globally are devoting significant time and resources to studying the relationship between diet, dietary patterns, lifestyle risk factors, obesity, and cancer.3 Advances in research methodology hold promise for reconciling the complex literature on the role of diet and cancer risk. Prior research focused more often on specific nutrients and foods in isolation rather than examining the effects of dose, timing, exposure, and overall nutritional status. However, more recent studies demonstrate that dietary patterns are key to enhancing our knowledge of the relationship between diet and cancer. The consensus across studies suggests that a healthy dietary pattern includes fruits, vegetables, fish, whole-grain cereals, nuts, legumes, and intake of healthy fats. This is presumably due to the value of these foods in providing a combination of important vitamins, minerals, fiber, protein, and antioxidants associated with reduced cancer risk. An unhealthy dietary pattern, on the other hand, consists of red meat, processed meat, refined sugars and sugar-sweetened beverages, refined flours, alcohol, and high saturated fat intake. While diet is often a major contributor to the energy imbalance that can lead to the development of obesity, physical activity patterns also play a role. Extensive evidence shows increased physical activity may reduce the incidence of and survival from various cancers and that inactivity is associated with many chronic diseases. Strong evidence demonstrates that regular physical activity of all types protects against excessive weight gain and obesity. Thus, the interaction of energy intake (i.e., diet) and energy expenditure (i.e., physical activity) is fundamental to weight management and cancer risk and control. This chapter (1) summarizes the role of dietary factors and cancer risk, (2) highlights the relationship between dietary patterns and cancer, (3) summarizes the role of weight management and energy balance, (4) identifies potential environmental barriers to diet-related cancer risk reduction, and (5) offers areas for future research. Diet-, Physical Activity–, and Body Composition– Related Factors and Cancer Risk This section presents an overview of the best-established associations (i.e., graded as “strong evidence”) reported by the WCRF and the AICR between the leading causes of cancer death worldwide and dietary factors, physical activity, and body fatness4 (summarized in Table 2.1). Lung Cancer. Lung cancer is the most common cause of cancer and cancer death in both sexes combined worldwide. Smoking is the main cause of lung cancer globally, accounting for an estimated 90% of lung cancers among men and 80% in women. Arsenic in drinking water is the most established dietary risk factor for lung cancer. The World Health Organization (WHO) reports that contaminated groundwater is the main source of arsenic.5 Beta-carotene supplements are also associated with increased risk for lung cancer, particularly among smokers. This association was discovered through two large intervention trials, the Beta-carotene and Retinol Efficacy Trial (CARET) and the Alpha-Tocopherol, Beta-Carotene (ATBC) Cancer Prevention Study.6 The CARET study was conducted in the U.S. with male and female smokers and former smokers, as well as men with occupational exposure to asbestos. The ATBC Cancer Prevention Study was conducted in Finland with male smokers. There is only limited evidence of specific foods decreasing (e.g., vegetables, fruits, foods containing carotenoids, etc.) or increasing (e.g., red meat, processed meat, alcohol) lung cancer risk. Liver Cancer. Liver cancer is the fourth most common cause of cancer death worldwide and the fifth most commonly occurring cancer. Established risk factors of liver cancer include cirrhosis of the liver, long-term use of high-dose estrogen and progesterone, chronic 14 SECTION I Factors in Cancer Risk and Prevention Table 2.1. Dietary-, Physical Activity–, and Weight-Related Factors Showing Convincing or Probable Evidence of Association with the Top 10 Causes of Cancer Death Worldwide Cancer Increases Risk Decreases Risk Lung Arsenic in water** High-dose beta-carotene supplements1** Liver Exposure to aflatoxin2** Alcohol (45 grams or 3 drinks a day)** Body fatness (based on BMI)** Coffee* Stomach Body fatness (cardia) (based on BMI)* Alcoholic drinks (45 grams or 3 drinks a day)* Food preserved by salting3* Colorectal Processed meat4** Alcoholic drinks (30 grams or 2 drinks a day)** Body fatness5 Adult attained height6** Red meat (beef, pork, lamb, and goat from domesticated animals)* Physical activity7** Whole grains* Foods containing dietary fiber8* Dairy products9* Calcium supplements* (supplemental dose of 200–1,000 mg/day) * Breast Premenopausal Adult attained height6** Alcoholic drinks (no amount/limit was found)* Greater birthweight10* Postmenopausal Alcoholic drinks (no amount/limit was found)** Body fatness5** Adult weight gain** Adult attained height6** Vigorous physical activity* Body fatness11* Lactation12* Physical activity13* Body fatness in young adulthood11* Lactation12* Esophageal Adenocarcinoma Body fatness5** Squamous Cell Carcinoma Alcoholic drinks** Mate14* Pancreatic Body fatness15** Adult attained height6* Prostate Body fatness (advanced prostate cancer)16* Adult attained height6* ** Indicates convincing evidence. * Indicates probable evidence. Based on the grading criteria reported in the Diet, Nutrition, Physical Activity and Cancer: A Global Perspective, Third Expert Report (2018). ** Convincing (strong evidence): Evidence strong enough to support a judgment of a convincing causal (or protective) relationship, which justifies making recommendations designed to reduce the risk of cancer. The evidence is robust enough to be unlikely to be modifiable in the foreseeable future as new evidence accumulates. * Probable (strong evidence): Evidence strong enough to support a judgment of a probably causal (or protective) relationship, which generally justifies recommendation designed to reduce the risk of cancer. 1 Based on studies testing high-dose supplements on smokers and former smokers (beta carotene: 20 mg/day or retinol: 25,000 IU/day). 2 Grains (cereals), legumes (pulses), seeds, nuts, and some fruits and vegetables are foods that may be contaminated with aflatoxins. 3 Regarding traditionally prepared in East Asia, high-salted foods and foods preserved with salt, such as pickled vegetables and salted or dried fish. 4 Regarding meat preserved by smoking, curing, slating, or adding chemical preservatives. 5 Body fatness represents BMI (body mass index), waist circumference, or waist-hip ratio. 6 Adult attained height is an indicator of genetic, environmental, hormonal, and nutritional growth factors that have an influence on growth from preconception to completion of linear growth. 7 Includes physical activity of all types: occupational, household, transport, and recreational. The panel judges that the evidence for colon cancer is convincing, but no conclusion was drawn for rectal cancer. 8 Both foods that naturally have dietary fiber and foods with dietary fiber added. 9 Evidence from dairy, milk, cheese, and dietary calcium supplements. 10 “The Panel’s conclusion relates to the evidence for overall breast cancer (unspecified). The observed association was in estrogen-receptor-negative (ER–) breast cancer only.” 11 Body fatness represents BMI, waist circumference, or waist-hip ratio. Women 18 to 30 years old included in evidence. For young adults, body fatness represents BMI. 12 “The Panel’s conclusion relates to the evidence for overall breast cancer (unspecified). The evidence for premenopausal and postmenopausal breast cancers separately was less conclusive, but consistent with the overall finding.” 13 “Physical activity including vigorous, occupational, recreational, walking and household activity.” 14 Beverage is consumed scalding hot and through a metal straw (as traditionally consumed in South America). 15 Body fatness referring to/interconnected with fat distribution, BMI, abdominal girth, and adult weight gain. 16 Body fatness represents BMI, waist circumference, or waist-hip ratio. This outcome was seen in advanced cancer only, meaning high-grade and fatal prostate cancers. CHAPTER 2 Diet and Cancer viral hepatitis, and smoking. There is strong evidence that the following diet-and weight-related factors increase liver cancer risk: (1) being overweight or obese (as assessed by BMI), (2) alcoholic drinks (about 3 drinks/day), and (3) exposure to aflatoxins. Aflatoxin, a mold that develops on foods stored in hot, wet conditions, can contaminate foods such as cereals (grains), legumes, seeds, and nuts, and some fruits and vegetables. Coffee consumption is the only diet- related factor that is protective. A dose-response meta-analysis of existing studies conducted by the expert panel suggests that one cup of coffee per day is associated with a 14% decreased risk.4 Stomach (Gastric) Cancer. Stomach cancer is the fourth most common cancer worldwide, with the highest incidence noted among men and in certain regions of Asia, and is the third most common cause of cancer death. Based on the location of the tumor, stomach cancer can be classified as cardia (top part and closest to esophagus) and noncardia (all other regions). Stomach cardia cancers are more common in the U.S. and UK, while noncardia forms of stomach cancer are more prevalent in Asia. However, incidence rates of stomach cancer (particularly noncardia) are declining worldwide due in part to more widespread use of refrigeration to store foods (rather than salting) and a decrease in Helicobacter pylori (H. pylori) infections. Smoking and exposure to industrial chemicals are other established contributors to stomach cancer. Diet-and body composition–related factors that increase the risk of stomach cancer include alcoholic drinks (three drinks/day), high-salt foods, and obesity. Being overweight or obese increases the risk of stomach cardia cancer in particular. Colorectal Cancer. Colorectal cancer is the third most commonly diagnosed cancer and the second most common cause of cancer deaths worldwide. Diet, physical activity, obesity, and alcohol consumption influence risk. The factors with the strongest evidence for increasing risk are (1) processed meat intake, (2) alcoholic drinks (about two drinks/day), (3) body fatness, (4) adult attained height, and (5) red meat. Adult attained height is not a direct risk factor, but rather a marker for factors (e.g., genetic, environmental, hormonal, and nutrition) that could impact growth during the developmental years. Red meat contains the iron-containing protein heme, which can facilitate the formation of potentially carcinogenic compounds. Also, red meat cooked at high temperatures can produce heterocyclic amines and polycyclic aromatic hydrocarbons that may contribute to colon cancer in people with a genetic predisposition. Processed meats (e.g., ham, bacon, sausages, canned meats) are preserved by methods other than freezing, such as smoking, salting, air drying, or heating. Strong evidence of factors decreasing risk include (1) physical activity, (2) whole grains, (3) dietary fiber, (4) dairy products, and (5) calcium supplements. Of these factors, the most convincing evidence is based on studies examining physical activity (e.g., occupational, household, transport, and recreational) and colorectal cancer. Based on a meta-analysis of over 30 studies, a reduced risk of about 14% for colon cancer was observed comparing those in the highest vs. lowest groups for physical activity (risk ratio [RR] = 0.85; 95% confidence interval [CI]: 0.78–0.91).4 For whole- grain consumption there was a reduced risk of 17% per 90 g/day of whole-grain intake (based on six studies consisting of n = 8,320 cases).4 For fiber-containing foods, which include fiber that is added and naturally occurring, there was a reduced risk of 9% per 10 g/ day (based on 15 studies consisting of n = 14,876 cases).4 Calcium and dairy products also appear to reduce colorectal cancer risk, though the effect for milk is, in part, mediated by calcium. Evidence for calcium’s protective effects is based on studies of supplements at doses of 200 to 1,000 mg/day. Breast Cancer. Breast cancer is the most frequently occurring cancer and the most common cause of cancer death for women worldwide. Because it is a hormone-related cancer, risk is most affected by factors that influence exposure to estrogen, including menopausal status. In a recent update by the WCRF/AICR, the following factors were considered strong evidence (convincing) for increasing risk of postmenopausal breast cancer: (1) alcoholic drinks (no amount identified), (2) body fatness, (3) adult weight gain, and (4) adult attained height.4 Adult attained height (a marker for factors affecting growth) and alcohol intake also increase risk for premenopausal breast cancer. Additionally, greater birthweight, which is an indicator of prenatal growth and fetal nutrition, is also recognized as a risk factor for premenopausal women. While body fatness increases breast cancer risk for postmenopausal women, it is actually protective for premenopausal women. Lactation and physical activity decrease risk for both pre-and postmenopausal women. However, evidence is insufficient to confirm protective effects of any specific dietary factors. Esophageal Cancer. Cancer of the esophagus is the sixth most common cause of cancer death and the seventh most common cancer worldwide. There are two main types of esophageal cancer: squamous cell carcinoma (affects the upper part of the esophagus) and adenocarcinoma, which occurs in the region between the esophagus and stomach. Risk factors vary by site. For instance, body fatness increases the risk for esophageal adenocarcinoma but not squamous cell. Squamous cell carcinoma can be impacted by diet-related factors. For instance, intake of alcohol and mate are associated with increased risk of squamous cell carcinoma rather than adenocarcinoma. Mate is a tea-like beverage consumed in parts of South America, usually scalding hot, through a metal straw. Pancreatic Cancer. Pancreatic cancer is the seventh most common cause of cancer deaths. Incidence is higher in men than in women and higher in developed countries. The WCRF/AICR’s continuous update project concluded there is convincing evidence that body fatness and adult attained height increase pancreatic cancer risk.4 No convincing or probable evidence suggests that any dietary factors increase risk, though limited data suggests that red and processed meats, alcohol, high-fructose foods/beverages, and foods containing saturated fatty acids increase risk. Coffee was previously considered a possible risk factor, but the updated report indicates this is unlikely. No food or nutrition factors are identified as decreasing pancreatic cancer risk. Prostate Cancer. Prostate cancer is the second most common cancer and fifth most common cause of cancer death in men. Incidence is much higher in developed countries. The WCRF/ AICR’s continuous update project report suggests there is strong probable evidence that body fatness and adult attained height increase prostate cancer risk. However, insufficient data exists to identify any dietary factor as risk promoting.4 Cervical Cancer. Cervical cancer ranks fourth in both mortality and incidence for women worldwide. The primary risk factor is infection with human papilloma viruses. Food and nutrition do not play a significant role in increasing or decreasing cervical cancer risk.4 15 16 SECTION I Factors in Cancer Risk and Prevention Dietary Patterns and Cancer Risk Single foods and nutrients are not typically consumed in isolation. Because dietary nutrients are consumed in combination, synergistic effects between food and nutrients may create a metabolic milieu that prevents or promotes carcinogenesis. This section presents an overview of dietary patterns and associations with cancer risk and risk of cancer-related mortality as indicated by studies that examined adherence to science-based public health dietary recommendations such as the U.S. government’s Dietary Guidelines for Americans (DGAs) and Mediterranean and vegetarian dietary patterns. The DGAs and a Mediterranean diet (Med Diet) pattern have corresponding index scores that are used to quantify adherence using a standardized approach.7 The DGAs are designed to promote good health and reduce the risk of chronic diseases, including cancer. The guidelines are revised every five years to account for advances in scientific knowledge pertaining to diet and disease relationships (the current DGAs are presented in Table 2.2). The Healthy Eating Index (HEI) is a scoring tool that measures adherence to a given set of DGAs; higher scores are indicative of greater adherence to the guidelines.8 A recent systematic review and meta-analysis of prospective cohort studies examining diet quality, using several metrics including the HEI and various health outcomes, found that individuals consuming the highest-quality diets compared to lowest-quality diets had a 16% reduction in cancer mortality or incidence (RR = 0.84; 95% CI: 0.82–0.87).9 Ecological studies suggest overall cancer risk is lower in Mediterranean countries versus northern Europe, the UK, and the U.S. Many have attributed this distinction to the customary foods consumed by people residing in this region. A Med Diet pattern is one in which vegetables and whole grains feature prominently, fresh fruit is a typical dessert, olive oil is the main fat source, animal-based protein intake is limited, and wine is consumed in moderation, with meals. Mechanistically, it is hypothesized that certain aspects of the Med Diet, including a healthy fatty acid ratio and foods rich in antioxidants and anti-inflammatory nutrients, work synergistically to promote reduced systemic inflammation and down-regulation of pro-carcinogenic pathways. Several research groups have developed scoring indices to operationalize and assess adherence to a Med Diet pattern to relate to disease outcomes. The Alternate Mediterranean Diet (aMED) score is a Med Diet adherence score developed specifically for U.S. populations.10 The aMED has nine components, with one point awarded Table 2.2. Dietary and Lifestyle Recommendations for Good Health and Cancer Prevention 2015–2020 Dietary Guidelines for Americans35 General Mediterranean Diet Characteristics36 American Cancer Society37 American Institute for Cancer Research38 5 overarching guidelines of a healthy eating pattern: • Follow a healthy eating pattern across the lifespan. • Focus on variety, nutrient density, and amount. • Limit calories from added sugars and saturated fats and reduce sodium intake. • Shift to healthier food and beverage choices. • Support healthy eating patterns for all. A healthy eating pattern includes: • A variety of vegetables from all the subgroups—dark green, red and orange, legumes (beans and peas), starchy, and other. • Fruits, especially whole fruits. • Grains, at least half of which are whole grains. • Fat-free or low-fat dairy, including milk, yogurt, cheese, and/or fortified soy beverage. • A variety of protein foods, including seafood, lean meats and poultry, eggs, legumes (beans and peas), and nuts, seeds, and soy products. • Oils. A healthy eating pattern limits: • Saturated fats (<10% calories per day) and trans fats, added sugars (<10% of calories per day), and sodium (<2,300 mg per day). • If alcohol is consumed, it should be consumed in moderation—up to one drink per day for women, and up to two drinks per day for men—and only by adults of legal drinking age. Healthy Eating Patterns Dietary Principles: • An eating pattern represents the totality of all foods and beverages consumed. • Nutritional needs should be met primarily from foods. • Healthy eating patterns are adaptable. Meet the Physical Activity Guidelines for Americans. • Daily abundance of plant- • Achieve and maintain • • • • • Be a healthy weight. • Be physically active. • Eat a diet rich in whole grains, • • • • • based foods including whole grains, vegetables, fruits, and legumes. Olive oil used daily as the principal fat source. Low to moderate daily consumption of low-fat dairy foods. Animal-based protein consumed in low to moderate amounts weekly or monthly. Sweets consumed in low amounts monthly. Wine in moderation with meals. Be physically active. • • • • • a healthy weight throughout life. Be physically active. Limit time spent sitting. Eat a healthy diet, with an emphasis on plant foods. Choose foods and drinks in amounts that help you get to and maintain a healthy weight. Limit how much processed meat and red meat you eat. Eat at least 2½ cups of vegetables and fruits each day. Choose whole grains instead of refined grain products. If you drink alcohol, limit your intake. vegetables, fruits, and beans. • Limit consumption of “fast food” and • • • • • • other processed foods high in fat, starches, or sugars. Limit consumption of red meat and processed meat. Limit consumption of sugar-sweetened beverages. Limit alcohol consumption. Do not use supplements for cancer prevention. For mothers: breastfeed your baby if you can. After a cancer diagnosis: follow our recommendations if you can. CHAPTER 2 Diet and Cancer for scoring higher than the median intake within a given population/cohort for whole grains, fruits, vegetables (except potatoes), nuts, fish, legumes, and monounsaturated versus saturated fat ratio; one point is awarded for red and processed meat below the median; and one point is awarded for consuming one alcoholic beverage daily. In the National Institutes of Health–American Association of Retired Persons (NIH-AARP) Diet and Health observational cohort study, greater adherence to a Med Diet pattern (aMED scores ranging from six to nine points) was associated with decreased risk of cancer-related mortality in both men and women.11 Regarding site-specific cancers, greater adherence to a Med Diet, based on aMED, was associated with lower colorectal cancer risk in men in a combined analysis of the Nurses’ Health Study and Health Professionals Follow-up Study,12 and decreased risk of lung cancer in both men and women in the NIH-AARP cohort, with an even more profound risk reduction in current and former smokers.13 However, not all studies have shown a strong association between Med Diet adherence and decreased cancer risk and mortality. For example, in the French NutriNet-Santé cohort study, greater adherence to a Med Diet, based on the Medi-Lite score, was not associated with decreased risk of breast (women), colorectal, or prostate cancer (men).14 In the Multiethnic Cohort study, greater adherence to a Med Diet, based on aMED, was associated with lower colorectal cancer mortality among African American cancer survivors but not Native Hawaiian, Japanese American, Latino, and white survivors.15 An important issue with the literature examining associations between Med Diet adherence and cancer risk and mortality is the use of different scoring approaches to assess Med Diet adherence. However, in the European Prospective Investigation into Cancer and Nutrition (EPIC) study, researchers investigated three different Med Diet scores (Mediterranean Diet Score [MDS], relative Med Diet Score [rMED], and the Mediterranean Style Dietary Pattern Score [MSDPS]) and associations with overall cancer mortality. Comparing the highest versus lower quartile for each score, higher Med Diet adherence was associated with significantly lower risk of cancer-related mortality irrespective of the scoring approach used.16 The association between a vegetarian dietary pattern and reduced cancer risk stems from studies of the Seventh Day Adventist religious sect whose doctrine advises against eating animal flesh. Seventh Day Adventists adhering to a vegetarian eating pattern had lower rates of cancer overall, lower rates at specific sites such as the prostate and colon, and lower risk of cancer-related mortality compared to the general U.S. population.17 However, Seventh Day Adventists also typically abstain from tobacco and alcohol, which may contribute to the observed health effect. In the EPIC cohort, vegetarianism was associated with lower overall cancer risk and risk for stomach and bladder cancer, but no effect was observed for colorectal and prostate cancer incidence compared to nonvegetarians.18 Studies of breast cancer incidence and mortality have not demonstrated differences between vegetarians and nonvegetarians, although there is some evidence that a vegan diet pattern can reduce breast cancer risk.19 Moreover, a vegan diet pattern was associated with statistically significant protection from overall cancer incidence in the Adventist Health Study-2.20 Diet and Weight Loss Intervention Trials: Effects on Cancer-Related Outcomes This section presents an overview of several large randomized trials designed to examine the effects of dietary factors and weight loss on cancer prevention or control and cancer risk–related biomarkers. Increasing Fiber, Fruits, and Vegetables and Decreasing Total Fat The Women’s Health Initiative (WHI). The WHI was a study of over 45,000 postmenopausal women (1993–2004) that included a clinical trial with three intervention arms, including two that were diet and cancer related. The first of these tested a low-fat eating pattern (less than 20% of total calories; five servings/day of fruits and vegetables; six servings/day of whole grains) on breast cancer and colorectal cancer. Control participants received information consistent with the U.S. Department of Agriculture DGAs. Follow-up at 8.1 years showed no significant reduction in the incidence of breast cancer or colon cancer among women in the intervention group.21 The second arm examined the effects of calcium and vitamin D supplementation on colorectal cancer. Over an average of seven years, no significant difference was observed in colorectal cancer incidence between the intervention and control groups.22 The extended period over which colorectal cancer develops may have led to these null findings. In recent secondary analyses, vitamin D and calcium supplementation were not associated with reduced invasive cancer risk or mortality,23 whereas vitamin B6 and riboflavin intake were associated with lower colorectal cancer risk.23 Women’s Intervention Nutrition Study (WINS). This phase III clinical trial (1994–2001) was designed to examine the relationship between dietary fat intake and breast cancer among 2,437 women with resected, early-stage breast cancer. Women in the intervention group were counseled to reduce dietary fat intake to 15% of calories during a four-month intervention period. The comparison group received no dietary counseling. Interim results at 60 months showed dietary fat intake and body weight were significantly lower in the intervention group compared to the control group.24 Women’s Healthy Eating and Living (WHEL) Study. This randomized trial (1995–2006) assessed whether a significant increase in vegetable, fruit, and fiber intake and a decrease in dietary fat intake could reduce the risk of recurrent and new primary breast cancer and “all cause” mortality among 3,088 survivors of early-stage breast cancer. Women in the intervention were instructed to consume daily five vegetable servings plus 16 ounces of vegetable juice, three fruit servings, 30 grams of fiber, and 15% to 20% of energy intake from fat. Women in the comparison group received written materials consistent with the “5-a-Day” fruits and vegetables message. Although the intervention group did adhere to the prescribed diet, there was no effect on breast cancer events or mortality among early-stage breast cancer survivors.25 Mediterranean Diet Only two studies have tested the effect of a Med Diet on cancer risk in the context of a randomized controlled trial. Lyon Diet and Heart Study. Six hundred and five adult survivors of a first acute myocardial infarction were randomized to a Med Diet–type pattern or control (Step 1 diet of the American Heart 17 18 SECTION I Factors in Cancer Risk and Prevention Association) over a four-to five-year timeframe.26 A secondary outcome of the study was the occurrence of malignant tumors. Seventeen cancers developed in the control group and seven in the Med Diet group (RR = 0.39; 95% CI: 0.15–1.01; p = 0.05). This study demonstrated for the first time in a randomized trial the cancer protective effect of a Med Diet in a non-Mediterranean population. Prevención con Dieta Mediterránea (PREDIMED) Trial. Briefly, the PREDIMED study randomized 7,447 participants (4,282 women) to a Med Diet supplemented with extra-virgin olive oil, Med Diet supplemented with mixed nuts, or control (low-fat diet) intervention with a median follow-up of 4.8 years.27 A secondary outcome of the trial was breast cancer incidence for women without a history of breast cancer (n = 4,152). Breast cancer rates per 1,000 person-years were 1.1 for the Med Diet plus extra-virgin olive oil group, 1.8 for the Med Diet nuts group, and 2.9 for the control group, respectively. Although the results come from a secondary analysis, findings suggest a protective effect of a Med Diet supplemented with olive oil for the primary prevention of breast cancer. Effect of Diet on Premalignant Lesions and Cancer-Related Biomarkers Polyp Prevention Trial (PPT). The PPT28 was a randomized controlled study of the effects of a low-fat (20% of total energy intake), high-fiber (18 g/1,000 calories), high-fruit and -vegetable (five to eight daily servings) diet on the recurrence of colorectal adenomas among individuals who had a polyp removed in the previous six months. At the four-year follow-up, results suggested that adopting a low-fat, high-fiber diet and increasing fruit and vegetable consumption did not affect the risk of recurrence for colorectal adenomas. Controlled Feeding Studies. In a two-week strictly controlled diet exchange study in which native black Africans consumed an animal-based diet and African Americans consumed a plant-based diet, colonic mucosal proliferation and inflammation were significantly lower in the African Americans and significantly higher postdiet in the native black Africans.29 The authors attributed the effect in the African Americans to changes in gut microbial metabolic function (i.e., increased short-chain fatty acid production and decreases in secondary bile acids) that was related to the diet switch. In a crossover feeding trial conducted with relatively healthy men and women, consuming a high (i.e., refined grains and added sugars) and low (i.e., high in whole grains, legumes, fruits, and vegetables) glycemic index diet each for 28 days30 resulted in differing expression of plasma proteins related to carcinogenesis that was dependent on the subject’s baseline body adiposity (high vs. low fat mass). Specifically, in response to the high-glycemic-load diet, those with high fat mass had increased expression of plasma proteins related to cell cycle, DNA repair, and DNA replication that if sustained could lead to carcinogenesis. These findings suggest that obesity’s effect on cancer development may to some extent be tied to biological response to differing dietary patterns. Effect of Weight Loss on Cancer-Related Outcomes Surgically Induced Weight Loss. There is encouraging albeit conflicting evidence regarding the effect of surgical weight loss on cancer risk. In a study of obese patients undergoing laparoscopic gastric banding (n = 327) or medically induced weight loss (n = 681), gastric banding was associated with significantly lower incidence of cancer and cancer-related mortality in the surgical patients31 over a 23-year period. In a large population-based cohort study in the United Kingdom of 8,794 obese patients that underwent bariatric surgery (gastric banding, sleeve gastrectomy, and gastric bypass), decreased risk of hormone-related cancers including breast (odds ratio [OR] = 0.25; 95% CI: 0.19–0.33), endometrium (OR = 0.21; 95% CI: 0.13–0.35), and prostate (OR = 0.37; 95% CI: 0.17–0.76)32 was observed compared to obese patients not undergoing a bariatric procedure that were propensity matched for age, sex, comorbidity, and duration of follow-up. However, in the same study, there was no effect of gastric banding or sleeve gastrectomy on esophageal or colorectal cancer and an increased risk of colorectal cancer in patients receiving gastric bypass. Suggested mechanisms associated with this increase in risk include inflammation and hyperproliferation and gut microbiota changes following the surgical bypass procedure. Weight Management Lifestyle Interventions. Several studies have examined how weight loss through calorie restriction, dietary changes, and increased physical activity affects biological markers related to cancer risk. For example, in the Nutrition and Exercise in Women (NEW) study, 439 overweight and obese postmenopausal women were randomized to aerobic exercise, dietary weight management, or both versus control for 12 months.33 Compared to control, exercise plus diet-induced weight loss was associated with significantly decreased BMI, insulin resistance, systemic inflammation, sex steroid hormones, and genes related to growth factor signaling. Nonetheless, few studies have been able to discern the effect of behavioral weight management interventions on cancer- specific outcomes (e.g., cancer risk, disease-free survival). Challenges to Healthy Eating and Weight Management for Cancer Risk Reduction and Cancer Health Equity As noted earlier and highlighted in consensus reports from leading cancer organizations, modifiable lifestyle behavioral risk factors, including diet and physical inactivity, account for between 30% and 50% of cancers. Often, the combination of less healthful diets and physical inactivity leads to excessive weight gain and obesity, increasing cancer risk. Recent estimates reflect that obesity accounts for 14% to 20% of the attributable cancer risk for U.S. adults and as much as 50% of all cancers for individuals under age 65 years. In the most recent nationally representative survey of adults in the U.S. (2013–2014), the age-adjusted prevalence of obesity was 35.2% among men and 40.4% among women. There were differences, however, across race/ethnicity, with prevalence rates of 38.7%, 57.2%, and 46.6% among non-Hispanic white, Non-Hispanic black, and Hispanic women, respectively.34 The differences in prevalence rates among men were not as striking, with rates of 35.4%, 38.2%, and 38.8% among non-Hispanic white, non-Hispanic black, and Hispanic men, respectively.34 Unfortunately, minorities and low-income individuals are at a significant disadvantage when it comes to making healthier dietary choices, driving obesity rates. For example, the main components of the Med Diet, which is embraced by the scientific community and associated with an inverse association with total mortality incidence of coronary heart disease, stroke, and several cancers, are characterized by a high consumption of vegetables, fruits, whole grains, CHAPTER 2 Diet and Cancer legumes, olive, and fish and a low intake of saturated fats, red meat, and dairy products. However, these foods tend to be more costly than many energy-dense foods that are discouraged in dietary recommendations for cancer prevention. In addition, urban areas that are often predominantly minority and/or low income tend to offer limited access to the foods recommended for cancer prevention, and the built environment may offer fewer opportunities for safe outdoor physical activity and introduce other factors that increase cancer risk (e.g., higher rates of tobacco or alcohol use, increased outlets selling alcohol and tobacco, and exposure to manufacturing chemicals). Conclusions and Future Research Primary prevention is possible through changes in modifiable risk factors, including a healthful diet and regular physical activity, leading to maintenance of a healthy weight. We have reviewed published guidelines from several agencies that made recommendations that have implications especially for the risk of colon, breast, and lung cancer. Increasingly, we are understanding that the overall population’s health is dependent on identifying and intervening on upstream social determinants of health that address social and environmental contextual factors that can drive unhealthful behavior. These factors can impede the ability of individuals to make choices consistent with reduced cancer risk. Further research can provide a lens for population-level action that can reduce lifestyle-related cancer inequities. ACKNOWLEDGMENTS Research reported in this publication was supported, in part, by the University of Illinois Cancer Center, National Institutes of Health’s National Cancer Institute, Grant Numbers U54CA202995, U54CA202997, U54CA203000, and T32CA057699. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health. REFERENCES 1. Anand P, Kunnumakkara AB, Sundaram C, Harikumar KB, Tharakan ST, et al. Cancer is a preventable disease that requires major lifestyle changes. Pharm Res. 2008;25(9):2097–2116. 2. Irigaray P, Newby JA, Clapp R, Hardell L, Howard V, et al. Lifestyle- related factors and environmental agents causing cancer: An overview. Biomed Pharmacother. 2007;61(10):640–658. 3. Kushi LH, Doyle C, McCullough M, Rock CL, Demark-Wahnefried W, et al. American Cancer Society guidelines on nutrition and physical activity for cancer prevention: Reducing the risk of cancer with healthy food choices and physical activity. CA Cancer J Clin. 2012;62(1):30–67. 4. World Cancer Research Fund/ American Institute for Cancer Research; 2018. Diet, nutrition, physical activity and cancer: A global perspective. Available from: dietandcancerreport.org 5. World Health Organization; 2018. Arsenic in drinking water. Available from: https://www.who.int/news-room/fact-sheets/detail/arsenic 6. Greenwald P. Beta-carotene and lung cancer: A lesson for future chemoprevention investigations? J Natl Cancer Inst. 2003;95(1):E1. 7. Chiuve SE, Fung TT, Rimm EB, Hu FB, McCullough ML, et al. Alternative dietary indices both strongly predict risk of chronic disease. J Nutr. 2012;142(6):1009–1018. 8. Krebs-Smith SM, Pannucci TE, Subar AF, Kirkpatrick SI, Lerman JL, et al. Update of the healthy eating index: Hei-2015. J Acad Nutr Diet. 2018;118(9):1591–1602. 9. Schwingshackl L, Bogensberger B, Hoffmann G. Diet quality as assessed by the healthy eating index, alternate healthy eating index, dietary approaches to stop hypertension score, and health outcomes: An updated systematic review and meta-analysis of cohort studies. J Acad Nutr Diet. 2018;118(1):74–100.e11. 10. Fung TT, Chiuve SE, McCullough ML, Rexrode KM, Logroscino G, et al. Adherence to a DASH- style diet and risk of coronary heart disease and stroke in women. Arch Intern Med. 2008;168(7):713–720. 11. Mitrou PN, Kipnis V, Thiebaut AC, Reedy J, Subar AF, et al. Mediterranean dietary pattern and prediction of all-cause mortality in a us population: Results from the NIH-AARP diet and health study. Arch Intern Med. 2007;167(22):2461–2468. 12. Petimar J, Smith-Warner SA, Fung TT, Rosner B, Chan AT, et al. Recommendation-based dietary indexes and risk of colorectal cancer in the nurses’ health study and health professionals follow- up study. Am J Clin Nutr. 2018;108(5):1092–1103. 13. Anic G, Park Y, Subar A, Schap T, Reedy J. Index-based dietary patterns and risk of lung cancer in the NIH–AARP diet and health study. Eur J Clin Nutr. 2016;70(1):123. 14. Lavalette C, Adjibade M, Srour B, Sellem L, Fiolet T, et al. Cancer- specific and general nutritional scores and cancer risk: Results from the prospective Nutrinet- Sante cohort. Cancer Res. 2018;78(15):4427–4435. 15. Jacobs S, Harmon BE, Ollberding NJ, Wilkens LR, Monroe KR, et al. Among 4 diet quality indexes, only the alternate Mediterranean diet score is associated with better colorectal cancer survival and only in African American women in the multiethnic cohort. J Nutr. 2016;146(9):1746–1755. 16. Lassale C, Gunter MJ, Romaguera D, Peelen LM, Van der Schouw YT, et al. Diet quality scores and prediction of all-cause, cardiovascular and cancer mortality in a pan-European cohort study. PLoS One. 2016;11(7):e0159025. 17. Fraser GE. Associations between diet and cancer, ischemic heart disease, and all- cause mortality in non- Hispanic white California Seventh-Day Adventists. Am J Clin Nutr. 1999;70(3 Suppl):532s–538s. 18. Ogden CL, Carroll MD, Kit BK, Flegal KM. Prevalence of childhood and adult obesity in the united states, 2011–2012. JAMA. 2014;311(8):806–814. 19. Penniecook-Sawyers JA, Jaceldo-Siegl K, Fan J, Beeson L, Knutsen S, et al. Vegetarian dietary patterns and the risk of breast cancer in a low-risk population. Br J Nutr. 2016;115(10):1790–1797. 20. Tantamango-Bartley Y, Jaceldo-Siegl K, Fan J, Fraser G. Vegetarian diets and the incidence of cancer in a low-risk population. Cancer Epidemiol Biomarkers Prev. 2013;22(2):286–294. 21. Prentice RL, Caan B, Chlebowski RT, Patterson R, Kuller LH, et al. Low-fat dietary pattern and risk of invasive breast cancer: The women’s health initiative randomized controlled dietary modification trial. JAMA. 2006;295(6):629–642. 22. Wactawski- Wende J, Kotchen JM, Anderson GL, Assaf AR, Brunner RL, et al. Calcium plus vitamin d supplementation and the risk of colorectal cancer. N Engl J Med. 2006;354(7):684–696. 19 20 SECTION I Factors in Cancer Risk and Prevention 23. Brunner RL, Wactawski- Wende J, Caan BJ, Cochrane BB, Chlebowski RT, et al. The effect of calcium plus vitamin d on risk for invasive cancer: Results of the women’s health initiative (WHI) calcium plus vitamin D randomized clinical trial. Nutr Cancer. 2011;63(6):827–841. 24. Chlebowski RT, Blackburn GL, Thomson CA, Nixon DW, Shapiro A, et al. Dietary fat reduction and breast cancer outcome: Interim efficacy results from the women’s intervention nutrition study. J Natl Cancer Inst. 2006;98(24):1767–1776. 25. Pierce JP, Natarajan L, Caan BJ, Parker BA, Greenberg ER, et al. Influence of a diet very high in vegetables, fruit, and fiber and low in fat on prognosis following treatment for breast cancer: The women’s healthy eating and living (WHEL) randomized trial. JAMA. 2007;298(3):289–298. 26. de Lorgeril M, Salen P, Martin J-L, Monjaud I, Boucher P, et al. Mediterranean dietary pattern in a randomized trial: Prolonged survival and possible reduced cancer rate. Arch Intern Med. 1998;158(11):1181–1187. 27. Toledo E, Salas-Salvado J, Donat-Vargas C, Buil-Cosiales P, Estruch R, et al. Mediterranean diet and invasive breast cancer risk among women at high cardiovascular risk in the predimed trial: A randomized clinical trial. JAMA Intern Med. 2015;175(11):1752–1760. 28. Schatzkin A, Lanza E, Freedman LS, Tangrea J, Cooper MR, et al. The polyp prevention trial I: Rationale, design, recruitment, and baseline participant characteristics. Cancer Epidemiol Biomarkers Prev. 1996;5(5):375–383. 29. O’Keefe SJ, Li JV, Lahti L, Ou J, Carbonero F, et al. Fat, fibre and cancer risk in African Americans and rural Africans. Nat Commun. 2015;6:6342. 30. Garrison CB, Zhang Y, Navarro SL, Randolph TW, Hullar MAJ, et al. Proteomic analysis of plasma reveals fat mass 31. 32. 33. 34. 35. 36. 37. 38. influences cancer-related pathways in healthy humans fed controlled diets differing in glycemic load. Cancer Prev Res (Phila). 2019;12(9):567–578. Pontiroli AE, Zakaria AS, Fanchini M, Osio C, Tagliabue E, et al. A 23-year study of mortality and development of co-morbidities in patients with obesity undergoing bariatric surgery (laparoscopic gastric banding) in comparison with medical treatment of obesity. Cardiovasc Diabetol. 2018;17(1):161. Mackenzie H, Markar SR, Askari A, Faiz O, Hull M, et al. Obesity surgery and risk of cancer. Br J Surg. 2018;105(12):1650–1657. Foster-Schubert KE, Alfano CM, Duggan CR, Xiao L, Campbell KL, et al. Effect of diet and exercise, alone or combined, on weight and body composition in overweight-to-obese postmenopausal women. Obesity. 2012;20(8):1628–1638. Flegal KM, Kruszon-Moran D, Carroll MD, Fryar CD, Ogden CL. Trends in obesity among adults in the United States, 2005 to 2014. JAMA. 2016;315(21):2284–2291. Guenther PM, Casavale KO, Reedy J, Kirkpatrick SI, Hiza HA, et al. Update of the healthy eating index: Hei-2010. J Acad Nutr Diet. 2013;113(4):569–580. Willett WC, Sacks F, Trichopoulou A, Drescher G, Ferro-Luzzi A, et al. Mediterranean diet pyramid: A cultural model for healthy eating. Am J Clin Nutr. 1995;61(6 Suppl):1402s–1406s. American Cancer Society; 2017. ACS guidelines on nutrition and physical activity for cancer prevention. Available from: https://www.cancer.org/healthy/eat-healthy-get-active/ acs-guidelines-nutrition-physical-activity-cancer-prevention/ guidelines.html American Institute for Cancer Research; 2020. Cancer prevention recommendations. Available from: https://www.aicr.org/reduce- your-cancer-risk/recommendations-for-cancer-prevention/ 3 Physical Activity, Sedentary Behavior, and Cancer Christine M. Friedenreich, Chelsea R. Stone, and Jessica McNeil Introduction Physical activity includes elements of planned, intentional activity such as exercise but also extends further to include domains of household, occupational, transportation/ commuting, and recreational/leisure activities. While the health benefits associated with regular physical activity for primary prevention of several chronic diseases have been well established, the benefits for cancer prevention have accumulated mainly in the last 30 years. Nearly 500 observational epidemiologic and randomized controlled exercise intervention trials have been conducted worldwide that have examined how physical activity is associated with cancer risk and what the underlying biologic mechanisms are that could explain how physical activity reduces cancer risk. Given this accumulating evidence base, several national and international cancer agencies have adapted physical activity guidelines developed for general health1 to specific guidelines for cancer prevention.2 Emerging observational epidemiologic evidence also suggests that high amounts of sedentary time (e.g., too much sitting) increases the risk of developing cancer, independently of physical activity levels.3 Similar to physical activity, sedentary behavior can be subdivided into different domains including occupational/workplace, transportation/commuting, and recreational/leisure sedentary activities. Although current recommendations for sedentary time are generic (e.g., limit total sitting time), the World Health Organization (WHO) has suggested that the review of the scientific literature on sedentary behavior contributing to disease risk is a priority area for the development of future physical activity guidelines amid the growing amount of research being conducted in this area.1 The World Cancer Research Fund (WCRF) and American Institute for Cancer Research (AICR) also recommend “limiting sedentary habits/sitting less” as part of their physical activity recommendations for cancer prevention.2 In this chapter, we review the current observational epidemiologic evidence on the association between physical activity and sedentary behavior to cancer incidence. In addition, we review how specific types of activity (i.e., aerobic and resistance exercise training) and how cardiorespiratory fitness may play an etiologic role in cancer incidence. We then describe the hypothesized biologic mechanisms that appear to explain the association between physical activity, sedentary behavior, and cancer incidence. We also report on the current burden of cancer that could be attributed to lack of physical activity and sedentary behavior. Finally, we provide an overview of common barriers and facilitators to physical activity participation, as well as recent strategies and recommendations that have been suggested to promote physical activity participation and decrease sedentary time for cancer prevention. Epidemiologic Evidence Physical Activity and Cancer Incidence Literature Review Methods We completed a systematic review of the scientific literature on all English-language articles published on PubMed (National Library of Medicine [NLM], National Institutes of Health [NIH]) through July 1, 2019. Our search strategy combined terms for physical activity (exercise OR motor activity OR physical activity) as well as search terms related to cancer (cancer OR tumor OR neoplasm). All articles were screened via titles and abstracts, with full-text screening occurring for articles related to physical activity and cancer risk/incidence. In instances where articles reported multiple domains of physical activity, we extracted the risk estimates for the most comprehensive domain of physical activity (i.e., total physical activity or recreational physical activity). Additionally, we preferentially extracted overall estimates over stratified results and estimates of reported lifetime physical activity. In the event that lifetime physical activity was not reported or assessed, we utilized estimates for recent physical activity. Physical activity was self-reported in these studies and included a wide range of assessment methods that captured either current activity, activity in the past year, or activity over lifetime with different units of measurement of activity. Since physical activity assessments varied across these studies, the estimates extracted were those that compared the highest versus the lowest levels of activity in each study regardless of how these were defined. Estimates were pooled using DerSimonian-Laird random effects 22 SECTION I Factors in Cancer Risk and Prevention models, and for the purpose of this review, we limit the discussion and presentation of results to cancer sites with 10 or more included articles. Overview of the Evidence Through July 1, 2019, there were over 450 articles published investigating the association between physical activity and cancer risk. Of these 450 papers, we found 10 cancer sites for which at least 10 papers had been published. We estimated the summary risk estimates and extracted information on the evidence of dose-response effects for bladder, breast, colorectal, endometrial, esophageal, kidney, lung, ovarian, pancreatic, and prostate cancers as estimated in the individual studies (Table 3.1). The largest evidence base on the association between physical activity and cancer risk exists for breast, colorectal, and prostate cancers with 132, 107, and 67 studies, respectively, contributing to each estimate. Evidence is also accumulating for associations between physical activity with lung (n = 44 studies), endometrial (n = 31), ovarian (n = 28), and pancreatic cancers (n = 27). For these 10 cancer sites, there is consistent evidence that physical activity is associated with decreased cancer risk. The range of the effect sizes for the associations between physical activity and cancer risk was from 0.57 to 0.94, with the majority between 0.70 and 0.90. The strongest effects were found for esophageal cancer, for which a hazard ratio (HR) of 0.57 (95% confidence interval [CI]: 0.42– 0.78) was estimated from the 13 studies and 14 estimates that were combined for this summary effect estimate. There was considerable consistency across these studies, with 12 of the 14 estimates finding reduced risk of cancer associated with higher levels of physical activity and the two remaining studies finding nonstatistically significant increased risks with higher levels of physical activity. Prostate cancer had the smallest protective effect with a hazard ratio of 0.94 (95% CI: 0.89–0.99), though the summary estimate remained statistically significant. Out of the 67 estimates combined, 18 of the estimates were statistically significant decreased risks (i.e., HR and 95% CI below 1 with increased levels of physical activity). Only five studies found statistically significant increased risk of prostate cancer. The remaining 44 studies reported nonstatistically significant results. Breast cancer had an overall summary estimate of 0.77 (95% CI: 0.75–0.80) from 135 contributing estimates. Since there has been sufficient evidence published on the relation between physical activity and breast cancer incidence, detailed meta-analyses have been published on this topic. One such meta-analysis sought to investigate the association of moderate-to vigorous-intensity recreational physical activity and breast cancer risk, stratified by menopausal status.4 It was determined that estimates were similar for premenopausal and postmenopausal breast cancer risk, with risk estimates of 0.80 (95% CI: 0.74–0.87) and 0.79 (95% CI: 0.74–0.84), respectively. Dose-response analyses were completed for premenopausal and postmenopausal breast cancer risk, for which curvilinear trends were found, reflecting a point of diminishing returns with moderate-to vigorous-intensity recreational activity beyond 20–30 metabolic equivalents of task (MET)-hours/week.4 There were 107 studies (127 estimates) contributing to the summary risk estimate of 0.75 (95% CI: 0.72–0.79) between physical activity and colorectal cancer incidence. Similar to the level of evidence available for breast cancer, there have been targeted systematic reviews and meta-analyses investigating this association. More specifically, a review was completed that investigated the Table 3.1. Summary of Epidemiologic Evidence on the Association between Physical Activity and Cancer Risk through July 1, 2019 Site Bladder a No. of Studies No. of Estimates Pooled Estimate 95% CI I2 Dose-Response Effect (%) WCRF/AICR Classificationa PAGAC Grade 16 19 0.86 0.76–0.97 76.5% 3 of 11 (27%) N/A Strong Breast 132 135 0.77 0.75–0.80 81.4% 58 of 97 (60%) Convincing decreases risk/limited-suggestive decreases risk1 Strong Colorectal 107 127 0.75 0.72–0.79 70.5% 44 of 67 (66%) Convincing decreases risk Strongb Endometrial 31 31 0.74 0.68–0.80 24.4% 15 of 24 (63%) Probable decreases risk Strong Esophageal 13 14 0.57 0.42–0.78 84.4% 4 of 8 (50%) Limited-suggestive decreases risk Strong Kidney 24 30 0.83 0.72–0.95 70.7% 4 of 20 (20%) N/A Strong Lung 36 44 0.75 0.69–0.82 80.3% 10 of 14 (71%) Limited-suggestive decreases risk Moderate Ovarian 28 30 0.87 0.77–0.98 64.7% 10 of 16 (63%) N/A Limited Pancreatic 27 36 0.83 0.76–0.91 35.6% 5 of 17 (29%) N/A Limited Prostate 67 67 0.94 0.89–0.99 68.0% 18 of 43 (42%) N/A Limited WCRF/AICR rates physical activity as convincing decreases risk for premenopausal breast cancer and limited-suggestive decreases risk for postmenopausal breast cancer. Our estimate represents an overall breast cancer risk. b PAGAC grade based on “colon” cancer specifically. CHAPTER 3 Physical Activity, Sedentary Behavior, and Cancer domain-specific association between physical activity and colon and rectal cancer incidence.5 Estimates pertaining to recreational physical activity and cancer risk were 0.80 (95% CI: 0.71–0.89) and 0.87 (95% CI: 0.75–1.01) for colon and rectal cancers, respectively. Occupational physical activity had slightly stronger effects with risk estimates of 0.74 (95% CI: 0.67–0.82) and 0.88 (95% CI: 0.79–0.98) for colon and rectal cancers, respectively. Statistically significant associations were not found for household physical activity and cancer risk for either site.5 Within each included study presented in Table 3.1, we examined whether or not dose-response associations between increasing physical activity volume and reduced cancer risk have been investigated. The most evidence of a dose-response relation between increasing levels of physical activity and decreasing risks for cancer was found for lung, colorectal, endometrial, and ovarian cancers, for which >60% of studies that investigated these trends found statistically significant inverse associations. Evidence of a dose-response effect for these cancer sites provides further support for causal associations. The findings pertaining to physical activity and cancer incidence from the 2018 update of the WCRF/AICR2 as well as the 2018 Physical Activity Guidelines Advisory Committee (PAGAC)6 are also presented in Table 3.1. While our results consistently support the WCRF/AICR findings, there were several cancer sites for which we found considerable evidence for an association between physical activity and cancer risk that were not included in the WCRF/ AICR 2018 report since that report was focused primarily on the evidence from prospective cohort studies, whereas we included all observational epidemiologic research conducted to July 2019. Prostate cancer, for example, has nearly 70 studies that have investigated the association between physical activity and cancer risk; however, it has not been appraised by the WCRF/AICR. Given the evidence that we found in our review, prostate cancer would likely receive a classification of “limited-suggestive decreased risk” since the effect found is statistically significant and the overall magnitude of this effect is small (6% reduction of risk). Other cancers that have yet to be appraised by the WCRF/AICR include bladder (16 studies), kidney (24 studies), ovarian (28 studies), and pancreatic (27 studies). PAGAC grades were similar to our summary estimates. The PAGAC concluded that there was “strong” evidence that greater amounts of physical activity are associated with reduced risks of developing bladder, breast, colon, endometrial, esophageal, and kidney cancers and that there was “moderate” evidence for lung cancer and limited evidence to suggest that increased physical activity was associated with a decreased risk of ovarian, pancreatic, and prostate cancers. Heterogeneity We also examined the level of heterogeneity in the pooled summary estimates presented (Table 3.1), which were high, suggesting that the variations in effects between studies are not due to chance. There are different types of heterogeneity, such as clinical and methodological heterogeneity, that may be contributing to the overall levels found in the pooled summary estimates. Related to clinical heterogeneity, there may have been differences in populations or outcome definitions, though these factors were unlikely to be major sources of heterogeneity given the high degree of certainty in the evidence regarding most cancer diagnoses. Methodological heterogeneity was likely the largest contributor, primarily given the differences in physical activity assessment tools that exist across different studies. Self-reported questionnaires were the primary assessment methods used to capture data on physical activity in these studies; however, the validity and reliability of these questionnaires vary considerably across different instruments. For instance, the domains (i.e., occupational, household, recreational, transport), parameters (frequency, intensity, duration), and time periods (current, past year, lifetime) for physical activity assessment differ by study. Furthermore, different observational epidemiologic study designs have been used to capture physical activity either retrospectively (i.e., in case-control studies) or prospectively (i.e., cohort studies) in relation to cancer diagnosis with consequent differences in the possibility of recall error that can influence the magnitude and precision of the estimates. Sedentary Behavior and Cancer Risk Sedentary behavior is defined as all waking behaviors with an energy expenditure ≤ 1.5 METs in the sitting, reclining, or lying postures.7 Common sedentary behaviors include watching television and video game playing (classified as leisure/recreational sedentary time), sitting and working at a computer (classified as occupational/ workplace sedentary time), and sitting in a car (classified as transportation/commuting sedentary time). Although increases in total physical activity participation (especially ambulatory, spontaneous activities) may lead to reductions in sedentary time over a 24-hour period, it is important to recognize that sedentary behavior and physical activity participation are distinct entities.3 Specifically, high sedentary time is defined as “too much sitting,” whereas physical inactivity is defined as “too little exercise.”3 It is thus possible for an individual to achieve or exceed physical activity recommendations but also spend long, uninterrupted amounts of time sitting at work or at home. Conversely, a person may have a physically demanding job with little sedentary time (e.g., construction worker, cashier), but also no or low recreational physical activity participation. The American Institute of Cancer Research recognized the importance of considering both the amount of time dedicated to physical activity and time spent in sedentary behavior for cancer prevention and developed an educational infographic to illustrate these concepts (Figure 3.1). This infographic illustrates the importance of making time for physical activity and breaking up sedentary time. A meta-analysis published in 2014 that included 17 prospective studies and 18,553 cases reported that the highest levels of sedentary behavior were associated with an increased risk of 1.20 (95% CI: 1.08–1.53) in combined cancers compared to the lowest levels of sedentary behavior.8 Furthermore, a recent narrative review3 summarized the current epidemiologic evidence on the associations between sedentary behavior and the risk of specific cancer sites. This review suggests that sedentary behavior is most strongly associated with colon, endometrial, and lung cancers.3 Specifically, high versus low levels of sedentary time were consistently associated with a range in effect estimates of 1.28–1.44 for colon cancer.3 One meta-analysis9 also specified that the association between sedentary behavior and colon cancer remained after adjusting for physical activity (risk ratio [RR] = 1.31; 95% CI: 1.21–1.42) and stratifying according to sedentary behavior domains (occupational sedentary behavior: RR = 1.30; 95% CI: 1.20–1.40; recreational sedentary behavior: RR = 1.32; 95% CI: 1.17–1.49). A different meta-analysis also conducted a dose-response analysis and observed a 1.08 increased risk of colon cancer (95% CI: 1.04–1.11) for every two-hour increase in sitting time per day.10 Similar to colon cancer, a range in the effect 23 24 SECTION I Factors in Cancer Risk and Prevention Make Time + Break Time = Cancer Protection Daily Activity Cancer Risk Indicators Joe Kim Mike Ann Types of activity: Moderate/Vigorous Break Sedentary Low HIGH Figure 3.1. The American Institute of Cancer Research infographic on making time for physical activity and breaking up sedentary time for cancer prevention. Reprinted with permission from the American Institute for Cancer Research, “Make Time + Break Time,” 2019, https://www.aicr.org/learn-more-about-cancer/infographics/ make-time-break-time.html estimates for endometrial cancer of 1.28–1.36 has been previously reported in those with high versus low levels of sedentary time.3 Dose- response analysis indicated a 1.10 (95% CI: 1.05–1.15) increased risk of endometrial cancer for every two-hour increment in sitting time per day; however, the association between sedentary time and endometrial cancer risk was stronger for recreational sedentary time (RR = 1.66; 95% CI: 1.21–1.68) and not statistically significant for occupational sedentary time (RR = 1.11; 95% CI: 0.88–1.39).10 Lastly, a range in the effect estimates for lung cancer of 1.21–1.27 has been observed in those with high versus low levels of sedentary time,3 with dose-response analysis indicating a 1.06 (95% CI: 1.00– 1.11) increased risk of lung cancer for every two-hour increment in sitting time per day.10 However, these results should be interpreted with caution given potential residual confounding by smoking since smoking has been positively associated with both sedentary behavior and lung cancer risk.3 There has been inconsistent evidence to date regarding the associations between sedentary behavior and rectal, breast, lung, and ovarian cancers.3 There is an overall 1.02–1.15 range in effect estimates for rectal cancer in those with the highest versus lowest levels of sedentary time, with the majority of studies reporting nonstatistically significant associations between sedentary time and rectal cancer.3 Similarly, high levels of sedentary behavior have been associated with a range in effect estimates of 1.08–1.17 for breast cancer, with the majority of studies reporting small effect estimates.3 Conversely, a range in effect estimates of 1.22–1.26 for ovarian cancer in individuals with high levels of sedentary time was reported; however, these effect estimates were not statistically significant.3 Lastly, there is insufficient evidence to currently draw conclusions on the association between sedentary behavior and cancers of the prostate, testes, stomach, and kidneys and non-Hodgkin lymphoma.3 exercise (aerobic versus resistance exercises) influences cancer risk. In addition to the 150 minutes of moderate-intensity aerobic physical activity recommended by the WHO for general health, there are also recommendations to achieve two days or more per week of muscle-strengthening activities involving the major muscle groups.1 This type of physical activity is referred to as resistance training, strength training, weight training, and muscle-strengthening activities in the scientific literature. Unlike aerobic physical activity, there is a paucity of evidence to date on the association between resistance training and cancer risk since few studies have specifically sought to measure resistance exercise training separately from aerobic exercise training. A recent publication by Mazzilli and colleagues investigated the relationship between weight lifting and the risk of 10 common cancer types.11 Using data from the NIH–American Association of Retired Persons (AARP) Diet and Health Study, which included an analytic sample size of 215,122 individuals, this study reported that after adjusting for multiple covariates, including moderate-to vigorous-intensity physical activity participation, there was a statistically significant reduced risk of colon cancer for individuals who weightlifted in the low (5 minutes–1.5 hours) and high (2– 10+ hours) categories compared with those in the no weightlifting category, with hazard ratios of 0.75 (95% CI: 0.66–0.87) and 0.78 (95% CI: 0.61–0.98), respectively. Further analyses by sex for colon cancer indicated that the relation between weightlifting and reduced cancer risk may be modified by sex, with males experiencing a more protective effect than females. There was no association found between weightlifting and the other cancer types investigated (kidney, bladder, breast, lung, non-Hodgkin lymphoma, pancreas, prostate, rectum, or melanoma).11 Resistance Training and Cancer Risk More recently, research has begun to emerge regarding the etiologic role that cardiorespiratory fitness (or physical fitness) has in cancer incidence. An individual’s cardiorespiratory fitness refers to Besides examining the association by domain of physical activity, there is also interest and a need to understand how the type of Cardiorespiratory Fitness and Cancer Risk CHAPTER 3 Physical Activity, Sedentary Behavior, and Cancer the ability of their respiratory and circulatory systems to efficiently supply oxygen to the skeletal muscles and other organs at rest and during sustained bouts of aerobic physical activity.12 Hence, cardiorespiratory fitness can serve as a proxy, but objective, measurement of habitual physical activity, though it is important to recognize that physical activity participation and cardiorespiratory fitness are distinct entities. Being more physically active is often associated with higher cardiorespiratory fitness. However, it is possible for physically active individuals to have low levels of cardiorespiratory fitness, or for individuals with low habitual levels of physical activity to have high cardiorespiratory fitness. This discrepancy between physical activity levels and cardiorespiratory fitness may result from underlying health conditions, performing greater amounts of anaerobic activities (e.g., resistance training), or having a genetic predisposition for high cardiorespiratory fitness/high trainability in response to physical activity. Indeed, a systematic review identified 97 genes that can be used to predict cardiorespiratory fitness trainability, identifying individuals with a certain genetic makeup (i.e., having more positive alleles within these genes) to have the potential for higher increases in cardiorespiratory fitness as a result of physical activity participation.13 Pozuelo-Carrascosa and colleagues completed a systematic review and meta-analysis that investigated the association between cardiorespiratory fitness and site-specific cancer risk in men in 2019.14 This review was restricted to men because, as cited by the authors, no studies retrieved through the literature searches included women. Ten studies were included for qualitative review, while seven studies were carried forward for inclusion in the meta-analysis. Lung (n = 3), colon/rectum (n = 3), prostate (n = 3), skin (n = 2), and combined cancer sites (n = 4) were investigated for cancer risk comparing moderate versus low cardiorespiratory fitness. From these, there was a statistically significant reduced risk of lung, colon/rectum, and combined cancer sites for moderate versus low cardiorespiratory fitness, with hazard ratios of 0.53 (95% CI: 0.39–0.68), 0.74 (95% CI: 0.55–0.93), and 0.86 (95% CI: 0.79–0.93), respectively. Moderate versus low cardiorespiratory fitness was not associated with prostate cancer risk (HR 1.07; 95% CI: 0.92–1.21), and skin cancer did not have enough contributing studies to warrant meta-analyses. In an analysis comparing high versus low cardiorespiratory fitness, the authors investigated lung (n = 4), colon/rectum (n = 4), prostate (n = 4), oral and digestive (n = 2), genitourinary (n = 2), skin (n = 2), and combined cancer sites (n = 5).14 They found a statistically significant reduced risk of lung, colon/rectum, and combined cancer risk for high versus low cardiorespiratory fitness, with hazard ratios of 0.52 (95% CI: 0.42–0.61), 0.77 (95% CI: 0.62–0.92), and 0.81 (95% CI: 0.75–0.87), respectively. There may be an adverse effect of high cardiorespiratory fitness and prostate cancer risk, with an increased incidence of 1.15 (95% CI: 1.00–1.30), and there were not enough contributing studies to warrant meta-analyses for oral and digestive, genitourinary, and skin cancers. Overall, there appears to be a protective effect of higher versus lower cardiorespiratory fitness for all cancers combined, as well as for lung and colon/rectal cancers. It is important to recognize that the body of literature examining the association between cardiorespiratory fitness and cancer risk has limitations given that few studies have been conducted and most of the research has included mainly Caucasian men with high socioeconomic status, which limits the generalizability of these findings. This area of research is of high interest and requires future attention to elucidate the role of cardiorespiratory fitness in cancer etiology. Population Attributable Risk Quantifying the burden of cancer incidence that could be avoided if exposure to a given risk factor was reduced or eliminated can be done by estimating the population attributable risk (PAR). To estimate the PAR of a risk factor such as physical inactivity, the prevalence of exposure among cases and estimated risk of exposure is required (such as those shown in Table 3.1). We recently reported that for Canada in 2015, the total number of cancer cases attributable to moderate or low recreational physical activity participation was 9,247, which was equivalent to 4.9% of all newly diagnosed cancer cases that year (or 10.6% of all cancers associated with physical activity).15 We also estimated that 1.7% of all cancer cases were attributable to leisure-time (nonoccupational) sedentary time (or 5.8% of all cancer cases associated with sedentary behavior), accounting for 3,230 cancer cases.16 PAR estimates for physical inactivity and cancer incidence in other countries have also been provided, including the United States, for which approximately 46,300 cancer cases were attributed to physical inactivity, which accounted for 2.9% of all newly diagnosed cancers.17 This study was restricted to cancers of the corpus uteri, colon (excluding rectum), and breast. Hence, if all cancer sites that are now known to be associated with physical inactivity were included in this estimate, the number of cancer cases that could be prevented with adequate levels of physical activity in the United States would be much higher. Similar analyses have been completed in Australia18 and the United Kingdom,19 with estimates ranging from 1.0–1.6% when considering the proportion of newly diagnosed cancers attributable to physical inactivity in these populations. PAR estimates for sedentary behavior independent of physical inactivity for other countries besides Canada have not yet been published. Biologic Mechanisms Table 3.2 summarizes the biologic mechanisms hypothesized to explain associations of physical activity and sedentary behaviors with cancer risk. The effects of physical inactivity and sedentary behavior on carcinogenesis are likely multifactorial and will vary by cancer site and possibly also by individual factors such as age, gender, ethnicity/ race, body size, body composition, and cardiorespiratory fitness. In addition, the domain, type, duration, frequency, and intensity of physical activity as well as the duration of sedentary behavior are also key factors that will influence how these behaviors influence cancer development. Over the past 20 years, several hypothesized biologic mechanisms that could explain how regular physical activity influences cancer risk have been proposed, and several of these mechanisms have been examined specifically in randomized controlled exercise intervention trials in cancer-free populations.20–22 These trials have targeted biomarkers associated with cancer risk since trials with cancer incidence endpoints have been infeasible given the large sample size (estimates of 35,000–45,000 participants), cost, and long-term follow-up required to observe sufficient numbers of cancer cases. There is now evidence from these trials that aerobic exercise decreases levels of endogenous sex hormones, insulin resistance, inflammation, and obesity, particularly abdominal adiposity.20–22 Higher volumes and more intense levels of aerobic 25 26 SECTION I Factors in Cancer Risk and Prevention Table 3.2. Possible Effects of Physical Activity and/or Sedentary Behavior on Proposed Biologic Mechanisms for the Association with Cancer Risk Proposed Mechanism Possible Effects Strength of Evidence Cancer Sites Lower adiposity levels ↓ anti-inflammatory cytokines (adiponectin) ↓ pro-inflammatory cytokines (leptin, resistin) ↓ estrogen synthesis in fat Strong for physical activity and sedentary behavior Colorectal, postmenopausal breast, endometrial, ovarian, bladder, kidney, pancreatic Lower chronic inflammation and better immune response ↓ pro-inflammatory cytokines (IL-6, TNF-α) ↓ reactive oxygen species, nitrogen species ↑ antioxidant and oxidative damage repair capacity Strong for physical activity and some/limited for sedentary behavior Colorectal, postmenopausal breast, endometrial, prostate, ovarian, lung, esophageal, pancreatic Higher metabolic function ↑ insulin sensitivity ↑ IGF binding proteins ↓ fasting glucose Strong for physical activity and sedentary behavior Colorectal, breast, endometrial, ovarian, prostate, bladder, kidney, pancreatic Lower sex-steroid hormone levels ↓ estrone, estradiol, free estradiol ↓ number of ovulatory cycles ↑ sex hormone–binding globulin Strong for physical activity and some/limited for sedentary behavior Breast, endometrial, ovarian, prostate Lower gastrointestinal transit time ↓ exposure of colon mucosa and carcinogens ↓ concentrations of bile acids ↓ colon segmentation contractions Some/limited evidence for physical activity. None for sedentary time Colorectal Higher lung function ↓ concentration of carcinogenic material in the lungs and shorter duration of agent-airway interaction ↑ pulmonary ventilation and perfusion Some/limited evidence for physical activity. None for sedentary time Lung Higher skeletal muscle function ↑ GLUT-4 concentrations ↑ myokine concentrations ↑ mitochondrial function Some/limited evidence for physical activity and sedentary time Colorectal, breast, endometrial, prostate activity have also been shown to be particularly beneficial in reducing some of these biomarkers and adiposity levels.21–24 As for sedentary behavior, there is some evidence from prospective cohort studies suggesting that higher amounts of sedentary time are associated with an increased risk of obesity/weight gain,25–27 type 2 diabetes,25 and having higher fasting insulin levels.28 A systematic review of imposed uninterrupted sedentary behavior interventions lasting ≤7 days reported that these interventions led to decreases in insulin sensitivity and glucose tolerance, coupled with increases in triglyceride levels.29 There is also some evidence to suggest that short- term (7–14 days), imposed bed rest leads to a pro-inflammatory response through increases in C-reactive protein30 and interleukin-6 levels.30,31 Based on this evidence, it has been hypothesized that standing (rather than sitting) increases postural blood flow, energy expenditure, and muscle contractions, which then improve glucose regulation through the activation of GLUT-4 glucose transporters on skeletal muscles and mitochondrial function to increase energy production.32 To date, no trials have specifically examined associations between sedentary behavior and biomarkers of cancer risk. Furthermore, many studies have used self-reported measurements of TV viewing time or sedentary time, which is subject to recall and reporting biases.3 Therefore, additional studies are also needed to collect both objective and subjective measures of sedentary time to quantify context-specific sedentary behaviors more accurately and to design better randomized controlled trials aimed at reducing sedentary time. There are biological mechanisms that support the associations between cardiorespiratory fitness and cancer risk such as chronic inflammation, innate immunity, DNA repair ability, apoptosis, and cell proliferation, as well as regulations to steroid hormone levels.14 Future intervention trials should address the optimal type of exercise (e.g., aerobic versus resistance exercises) as well as other novel molecular and genetic pathways that could reduce cancer risk through physical activity participation and/or reductions in sedentary time. In addition, mechanisms unique to specific cancer sites (e.g., colon transit time for colon cancer and improved lung function for lung cancer) require targeted research studies. Finally, future observational epidemiologic research will help identify tumor characteristics (e.g., tumor subsite, histologic subtype, hormone receptor status, mutation carrier status) and individual characteristics that may also modify the causal mechanisms relating physical activity to cancer prevention. Barriers and Facilitators to Physical Activity Although the evidence base on the benefits of physical activity for reducing cancer risk is becoming well established, large proportions of the at-risk population fail to achieve recommended levels of physical activity. A pooled analysis of data from 358 population-based surveys done globally with data from 2001 to 2016 revealed that the prevalence of physically inactive individuals has been increasing, particularly in high-income countries.33 More specifically, during this time frame, the prevalence of physical inactivity increased by over five percentage points in high-income Western countries (from 30.9% to 36.8%).33 Understanding the reasons that the general population fails to engage in regular physical activity is challenging since multiple factors influence achieving recommended levels of physical activity. These factors can be broadly categorized as either barriers or facilitators to physical activity participation (Figure 3.2), and extensive research is focused on identifying, developing, and testing interventions that address these barriers and facilitators as a means of increasing physical activity levels and reducing sedentary time.34–37 In general, the social, economic, and built environments have a major impact on determining if individuals can engage in and sustain physical activity CHAPTER 3 Physical Activity, Sedentary Behavior, and Cancer Family friends Weather Lack of time Support Cost Medical professionals External barriers Accountability Access to facilities Desire for health Prevent future illness Location Intrinsic motivation Knowledge Facilitators Barriers Motivation Internal barriers Enjoyment Interest Societal Health Income security Fear Education Comorbid conditions Age Figure 3.2. Model of facilitators and barriers to physical activity. as a regular component of their lives. For example, individuals with lower incomes often have more physically demanding jobs, resulting in higher levels of occupational physical activity, but they may lack the economic resources or time to invest in recreational physical activity participation, which has been most commonly associated with improved health benefits. Typically, those individuals who have higher education and socioeconomic status understand the benefits derived from leading active lifestyles and have the means to be able to engage in such healthy behaviors. It is often the goal of researchers and public health agencies to find ways to reduce inequities to allow larger proportions of the population to access resources that may improve health. Actions have been taken to use publically available educational media campaigns to promote the health benefits of physical activity, and municipalities have been focused on improving the built environment and providing resources to encourage active lifestyles in city planning. Furthermore, employers have been utilizing health benefit plans to promote physical activity participation by allowing employees to claim costs for recreational physical activities under health and wellness spending accounts to provide an incentive to be more physically active. Modifications in workplace designs have also been encouraged to promote physical activity participation (e.g., access to exercise facilities at work and secure bike storage) and reduce sedentary time (e.g., access to sit-stand or cycling desks). Lastly, online platforms, mobile applications, and wearable devices have become increasingly popular to promote increases in physical activity participation coupled with reductions in sedentary time. These digital platforms can promote physical activity behavior change by sending motivational emails or text messages, sharing online educational modules, and providing readily available information on physical activity combined with motivational prompts or “nudges.” Indeed, a systematic review and meta-analysis reported a mean reduction of 40 minutes/day of sedentary time in intervention studies that used technology to promote sedentary behavior change in the workplace, community, and home settings.38 These examples of primary prevention aim to mitigate some of the barriers and foster factors seen as facilitators listed in Figure 3.2 in an effort to ultimately prevent chronic diseases, including cancers, before they ever occur. Public Health Recommendations Cancer and public health agencies worldwide have recognized the benefits of regular physical activity for reducing cancer risk at the population and individual level. As previously mentioned, the WCRF/AICR released its Continuous Update Report 2018, which included the recommendation that individuals be at least moderately active (defined as raising the heart rate to about 60–75% of its maximum) and to follow or exceed national guidelines.2 Specifically, to achieve benefits for cancer prevention and to have a significant impact on weight control, they recommend that 45–60 minutes of moderate-intensity physical activity per day is required. The WHO’s 2010 guidelines for physical activity advise adults to be active daily and to participate in at least 150 minutes of moderate-intensity aerobic physical activity or at least 75 minutes of vigorous-intensity aerobic activity or a combination of these intensities.1 These guidelines are being updated in 2020 and will include specific recommendations for cancer prevention. The recommendations from the WCRF/ 27 28 SECTION I Factors in Cancer Risk and Prevention AICR are also to limit the amount of sedentary time for extended time periods given that increased sedentary behavior is also a cause of weight gain and type 2 diabetes, which are established causes of several cancers. In 2018, the U.S. Department of Health and Human Services released the Physical Activity Guidelines for Americans (PAGA) report in which they recommended that adults should undertake at least 150–300 minutes per week of moderate-intensity or 75–150 minutes per week of vigorous-intensity activity or an equivalent combination of both moderate-and vigorous- intensity aerobic activity.6 In addition, they recommended that adults should also perform muscle-strengthening activities on two or more days per week. For older adults, there were additional recommendations to undertake multicomponent physical activity that includes balance training as well as aerobic and muscle-strengthening activities. The PAGA 2018 report highlighted that the benefits of increased physical activity included lower risk of several cancers including bladder, breast, colon, endometrium, esophagus, kidney, lung, and stomach. While the PAGA 2018 report did not make specific recommendations for physical activity for cancer prevention, it is noteworthy that the guidelines developed by the WCRF/AICR and those from the PAGA committee are both targeting higher levels of activity than those that were previously developed by the WHO. Hence, with the increased evidence base and more precision on the amount, type, and timing of activity required for cancer prevention, it appears that a minimum level of 150 minutes per week of moderate-intensity activity is required, with greater benefit being observed with higher levels of activity (e.g., around 300 minutes weekly). Conclusion The evidence base for the etiologic role of physical activity and sedentary behavior associated with cancer incidence has grown rapidly in the past 30 years, and there is now clear evidence that regular physical activity and decreased sedentary behavior are associated with the risk of 10 or more cancer sites. These associations have been confirmed in nearly 500 studies conducted worldwide using different study designs, study populations, assessment methods, and analytic approaches. These consistent risk associations range from around 10–45% reductions for the highest levels of physical activity in these studies compared to the lowest. There is also evidence for dose-response effects with increasing activity levels and decreasing cancer risk. With respect to type of physical activity, evidence is now emerging that resistance exercise may also be effective in reducing cancer risk in addition to aerobic exercise, for which the evidence base is well established. Cardiorespiratory fitness has more recently been shown in some studies to be an independent risk factor for cancer risk. Sedentary behavior as an independent risk factor for cancer risk has recently emerged, and the risks associated with prolonged sitting are now apparent for at least three cancer sites. The biologic mechanisms for these associations are being elucidated with evidence that physical activity and sedentary behavior influence cancer risk through multiple pathways that include an effect on adiposity, endogenous sex and metabolic hormones, inflammation, and possibly also immune and skeletal muscle functions. The burden of cancer that can be attributed to physical inactivity and sedentary behavior is notable, with up to 10% of all cancers associated with these risk factors. Multiple barriers and facilitators to regular physical activity participation are also being recognized. Interventions and support networks that address these factors are needed to increase the levels of physical activity and reduce sedentary time in the general population since trends of increasing inactivity and sedentary behavior are being reported worldwide. Public health and cancer agencies have recognized the need for clear guidelines on physical activity and sedentary behavior for cancer prevention, and recent updates from these agencies are recommending daily activity with a combination of aerobic and resistance exercises that reaches up to 300 minutes per week of moderate-intensity activity. While considerable progress has been made in elucidating the associations between physical activity, sedentary behavior, and cancer risk, there remain numerous areas that require future research to address the questions regarding the nature of these associations by domain, type, dose, and timing in relation to cancer risk as well as by population subgroups and by cancer types. More behavioral interventions specifically targeted at cancer prevention and focused on reducing sedentary behaviors are also needed with the objective of increasing the prevalence of physically active individuals at the population level. ACKNOWLEDGMENTS The authors acknowledge the assistance of Rebecca Urbat for completing literature searches on resistance training and cancer incidence and also Nathaniel Minichiello for completing literature searches on cardiorespiratory fitness and cancer incidence. REFERENCES 1. World Health Organization. Global recommendations on physical activity for health. World Health Organization; 2010. 2. World Cancer Research Fund/ American Institute for Cancer Research. Continuous Update Project Expert Report 2018: Diet, Nutrition, Physical Activity and Cancer: A Global Perspective. 2018. World Cancer Research Fund/ American Institute for Cancer Research. 3. Jochem C, Wallmann-Sperlich B, Leitzmann MF. The influence of sedentary behavior on cancer risk: Epidemiologic evidence and potential molecular mechanisms. Current Nutrition Reports. 2019;8(3):167–174. 4. Neilson HK, Farris MS, Stone CR, Vaska MM, Brenner DR, Friedenreich CM. Moderate-vigorous recreational physical activity and breast cancer risk, stratified by menopause status: A systematic review and meta-analysis. Menopause. 2017;24(3):322–344. 5. Mahmood S, MacInnis RJ, English DR, Karahalios A, Lynch BM. Domain- specific physical activity and sedentary behaviour in relation to colon and rectal cancer risk: A systematic review and meta- analysis. International Journal of Epidemiology. 2017;46(6):1797–1813. 6. U.S. Department of Health and Human Services. 2018 Physical Activity Guidelines Advisory Committee Scientific Report Washington, DC: U.S. Department of Health and Human Services; 2018. 7. Tremblay MS, Aubert S, Barnes JD, et al. Sedentary Behavior Research Network (SBRN)—Terminology Consensus Project process and outcome. International Journal of Behavioral Nutrition and Physical Activity. 2017;14(1):75. CHAPTER 3 Physical Activity, Sedentary Behavior, and Cancer 8. Shen D, Mao W, Liu T, et al. Sedentary behavior and incident cancer: A meta-analysis of prospective studies. PloS One. 2014;9(8):e105709. 9. Cong YJ, Gan Y, Sun HL, et al. Association of sedentary behaviour with colon and rectal cancer: A meta-analysis of observational studies. British Journal of Cancer. 2014;110(3):817–826. 10. Schmid D, Leitzmann MF. Television viewing and time spent sedentary in relation to cancer risk: A meta-analysis. Journal of the National Cancer Institute. 2014;106(7). 11. Mazzilli KM, Matthews CE, Salerno EA, Moore SC. Weight training and risk of 10 common types of cancer. Medicine & Science inSports & Exercise. 2019;51(9):1845–1851. 12. Lee DC, Artero EG, Sui X, Blair SN. Mortality trends in the general population: The importance of cardiorespiratory fitness. Journal of Psychopharmacology. 2010;24(4 Suppl):27–35. 13. Williams CJ, Williams MG, Eynon N, et al. Genes to predict VO2max trainability: A systematic review. BMC Genomics. 2017;18(Suppl 8):831. 14. Pozuelo-Carrascosa DP, Alvarez-Bueno C, Cavero-Redondo I, Morais S, Lee IM, Martinez-Vizcaino V. Cardiorespiratory fitness and site-specific risk of cancer in men: A systematic review and meta-analysis. European Journal of Cancer. 2019;113:58–68. 15. Friedenreich CM, Barberio AM, Pader J, et al. Estimates of the current and future burden of cancer attributable to lack of physical activity in Canada. Preventive Medicine. 2019;122:65–72. 16. Friedenreich CM, Pader J, Barberio AM, et al. Estimates of the current and future burden of cancer attributable to sedentary behavior in Canada. Preventive Medicine. 2019;122:73–80. 17. Islami F, Goding Sauer A, Miller KD, et al. Proportion and number of cancer cases and deaths attributable to potentially modifiable risk factors in the United States. CA: A Cancer Journal for Clinicians. 2018;68(1):31–54. 18. Olsen CM, Wilson LF, Nagle CM, et al. Cancers in Australia in 2010 attributable to insufficient physical activity. Australian and New Zealand Journal of Public Health. 2015;39(5):458–463. 19. Parkin DM. 9. Cancers attributable to inadequate physical exercise in the UK in 2010. British Journal of Cancer. 2011;105(Suppl 2):S38–S41. 20. Friedenreich CM, Woolcott CG, McTiernan A, et al. Adiposity changes after a 1- year aerobic exercise intervention among postmenopausal women: A randomized controlled trial. International Journal of Obesity (Lond). 2011;35(3):427–435. 21. Campbell KL, Foster-Schubert KE, Alfano CM, et al. Reduced- calorie dietary weight loss, exercise, and sex hormones in postmenopausal women: Randomized controlled trial. Journal of Clinical Oncology. 2012;30(19):2314–2326. 22. Friedenreich CM, Neilson HK, O’Reilly R, et al. Effects of a high vs moderate volume of aerobic exercise on adiposity outcomes in postmenopausal women: A randomized clinical trial. JAMA Oncology. 2015;1(6):766–776. 23. de Roon M, May AM, McTiernan A, et al. Effect of exercise and/ or reduced calorie dietary interventions on breast cancer-related endogenous sex hormones in healthy postmenopausal women. Breast Cancer Research. 2018;20(1):81. 24. Friedenreich CM, Neilson HK, Wang Q, et al. Effects of exercise dose on endogenous estrogens in postmenopausal women: A randomized trial. Endocrine-Related Cancer. 2015;22(5):863–876. 25. Hu FB, Li TY, Colditz GA, Willett WC, Manson JE. Television watching and other sedentary behaviors in relation to risk of obesity and type 2 diabetes mellitus in women. JAMA. 2003;289(14):1785–1791. 26. Blanck HM, McCullough ML, Patel AV, et al. Sedentary behavior, recreational physical activity, and 7- year weight gain among postmenopausal U.S. women. Obesity (Silver Spring, Md). 2007;15(6):1578–1588. 27. Mekary RA, Feskanich D, Malspeis S, Hu FB, Willett WC, Field AE. Physical activity patterns and prevention of weight gain in premenopausal women. International Journal of Obesity (Lond). 2009;33(9):1039–1047. 28. Helmerhorst HJ, Wijndaele K, Brage S, Wareham NJ, Ekelund U. Objectively measured sedentary time may predict insulin resistance independent of moderate-and vigorous-intensity physical activity. Diabetes. 2009;58(8):1776–1779. 29. Saunders TJ, Larouche R, Colley RC, Tremblay MS. Acute sedentary behaviour and markers of cardiometabolic risk: A systematic review of intervention studies. Journal of Nutrition and Metabolism. 2012;2012:712435. 30. Bosutti A, Malaponte G, Zanetti M, et al. Calorie restriction modulates inactivity-induced changes in the inflammatory markers C- reactive protein and pentraxin-3. Journal of Clinical Endocrinology & Metabolism. 2008;93(8):3226–3229. 31. Drummond MJ, Timmerman KL, Markofski MM, et al. Short- term bed rest increases TLR4 and IL-6 expression in skeletal muscle of older adults. American Journal of Physiology-Regulatory, Integrative and Comparative Physiology. 2013;305(3):R216–R223. 32. Kerr J, Anderson C, Lippman SM. Physical activity, sedentary behaviour, diet, and cancer: An update and emerging new evidence. Lancet Oncology. 2017;18(8):e457–e471. 33. Guthold R, Stevens GA, Riley LM, Bull FC. Worldwide trends in insufficient physical activity from 2001 to 2016: A pooled analysis of 358 population-based surveys with 1.9 million participants. Lancet Global Health. 2018;6(10):e1077–e1086. 34. Kuijpers W, Groen WG, Aaronson NK, van Harten WH. A systematic review of web-based interventions for patient empowerment and physical activity in chronic diseases: Relevance for cancer survivors. Journal of Medical Internet Research. 2013;15(2):e37. 35. Hadgraft NT, Brakenridge CL, Dunstan DW, Owen N, Healy GN, Lawler SP. Perceptions of the acceptability and feasibility of reducing occupational sitting: Review and thematic synthesis. International Journal of Behavioral Nutrition and Physical Activity. 2018;15(1):90. 36. Kelly S, Martin S, Kuhn I, Cowan A, Brayne C, Lafortune L. Barriers and facilitators to the uptake and maintenance of healthy behaviours by people at mid-life: A rapid systematic review. PloS One. 2016;11(1):e0145074. 37. Franks H, Hardiker NR, McGrath M, McQuarrie C. Public health interventions and behaviour change: Reviewing the grey literature. Public Health. 2012;126(1):12–17. 38. Stephenson A, McDonough SM, Murphy MH, Nugent CD, Mair JL. Using computer, mobile and wearable technology enhanced interventions to reduce sedentary behaviour: A systematic review and meta-analysis. International Journal of Behavioral Nutrition and Physical Activity. 2017;14(1):105. 29 4 Sun Exposure and Cancer Risk Suzanne J. Dobbinson, Afaf Girgis, Bruce K. Armstrong, and Anne E. Cust Epidemiology Sun Exposure and Cancer Risk Skin Cancer Skin cancers are highly preventable. Exposure to solar ultraviolet (UV) radiation is responsible for the development of some 50–90% of melanomas, 50–90% of basal cell carcinomas (BCCs), and 50–70% of squamous cell carcinomas (SCCs) of skin globally. UV radiation is also linked with ocular cancers and other eye conditions such as cortical cataract and pterygium (a benign growth of the conjunctiva). It is widely accepted that UV radiation causes skin cancer. However, the association of sun exposure with skin cancer appears complex. Sunburn history and other recalled sun exposure that is intermittent in nature, and mainly recreational exposure or holiday exposure, are the UV risk factors most strongly associated with melanoma. More continuous (chronic) sun exposure, typically occupational sun exposure, may not increase melanoma risk. Childhood is the period of greatest susceptibility to sun exposure effects on later melanoma risk; however, risk also accumulates with sun exposure in adulthood. SCCs are mainly associated with total and occupational sun exposure, and BCCs, like melanoma, with early life sun exposure, nonoccupational or recreational sun exposure, and history of sunburn. There is also convincing evidence that indoor tanning is associated with melanoma and keratinocyte skin cancers.1,2 For people who reported ever using indoor tanning devices compared with those who never used them, the relative risks were 1.20 (95% confidence interval [CI]: 1.08–1.34) for melanoma, 1.67 (95% CI: 1.29– 2.17) for SCC, and 1.29 (95% CI: 1.08–1.53) for BCC; these relative risks were higher for people who used indoor tanning devices before the age of 35 years. UV measurements made inside indoor tanning devices show that mean erythema-weighted UV irradiances are higher than those from natural sun, with large variations between devices, and that UVA exposure is particularly high.3 Importantly also, Colantonio et al.2 observed that there was no statistically significant difference in this association between exposure before and after the year 2000, thus suggesting that newer tanning technology is not safer than older technology. Incidence rates for melanoma and other skin cancers continue to rise in predominantly fair-skinned populations in spite of active promotion of sun protection over the past 50 years. There is, though, evidence of a plateau and nascent fall in melanoma incidence rates in some such populations (e.g., Australia, New Zealand, and Denmark), particularly in younger people. These trends could be due to increasing sun protection in recent cohorts born in these countries. There is little evidence of similar trends in incidence of keratinocyte cancers. Other Cancers A systematic review of the epidemiological evidence suggests that there is an inverse association of sun exposure with risk of some internal cancers, namely colorectal, prostate, breast, and non-Hodgkin lymphoma (NHL).4 The evidence was considered strongest for an inverse association between sun exposure and colorectal and prostate cancers; findings were more heterogeneous for the association with breast cancer and NHL. It is not yet possible to say whether these observed associations are causal, since confounding with other lifestyle factors such as diet and physical activity cannot be completely excluded. Paradoxically, a quite strong positive association has been observed between sun exposure and prostate cancer in Australia,4 a much higher UV environment than most countries in which an inverse association has been observed. Possible Cancer Causal or Cancer Protective Mechanisms Current best evidence indicates that sun exposure causes skin cancer by way of UV radiation causing damage to cellular DNA and gene mutation consequent on that damage. Conjunctival melanoma is probably also caused by UV radiation mutation of conjunctival cells, but there is little or no evidence of a similar causal path to uveal (internal ocular) melanoma. Immune-suppressive effects of sun exposure have been offered as a possible explanation for an increase in risk of internal cancer, such as prostate cancer, at higher levels of sun exposure; there is clear evidence that therapeutic immune suppression increases risk of many cancers. DNA damage in circulating lymphocytes has also been observed in people with higher levels of sun exposure, which raises the possibility of other mechanisms, although it is yet to be established that the damage observed in lymphocytes also occurs in solid organs. Anti-carcinogenic effects of vitamin D are the most common mechanism suggested for observed inverse associations between CHAPTER 4 Sun Exposure and Cancer Risk sun exposure and internal cancers. Vitamin D is produced in skin following exposure to solar UVB radiation and is transported in the bloodstream. Many in vitro studies have shown that high concentrations of vitamin D compounds can inhibit cancer cell proliferation and, sometimes, induce differentiation (cell “normalization”).4 A comprehensive review of studies of associations of vitamin D concentration and cancer incidence and mortality concluded, “Higher circulating levels of vitamin D (i.e., 25- hydroxyvitamin D or 25(OH)D) appear to be associated with reduced risk of colorectal and bladder malignancies, but higher risk of prostate and possibly pancreatic cancers, with no clear association for most other organ sites examined.”5 There is inconsistent evidence for a protective effect of vitamin D in the few randomized controlled trials in which the effect on cancer incidence of vitamin D supplementation, with or without supplementary calcium, has been examined (generally not as the primary objective of the study).4,5 Similarly, in a more recent review, Zhang et al.6 found evidence that vitamin D supplementation reduced risk of cancer death, but not all-cause mortality. More substantial such studies are underway (see, e.g., Neale et al.7). Other Health Effects of Sun Exposure There is a growing body of evidence for beneficial effects of sun exposure on health, which could be mediated by way of cutaneous production of vitamin D or by other mechanisms. These effects include reductions in risk of cardiovascular disease, Alzheimer disease or other dementia, myopia, macular degeneration, diabetes, and multiple sclerosis. While not within the scope of this chapter and not yet well reviewed, this evidence may have to be taken into consideration when considering sun protection recommendations and the populations within which they are made. Evidence That Reducing Sun Exposure Can Reduce Risk of Cutaneous and Ocular Cancers There is some evidence of the efficacy or effectiveness of sun exposure reduction in lowering risk of cutaneous or ocular cancer. The strongest evidence is from a handful of trials of the effects of sun protection, principally use of sunscreens, in reducing risk of known skin cancer precursors or skin cancers themselves. The number of cutaneous pigmented nevi is a very strong predictor of cutaneous melanoma risk and a clear precursor to a proportion of cutaneous melanomas. In an individually controlled trial, Vancouver schoolchildren 6–7 or 9–10 years of age were randomly assigned to be sent supplies of sun protective factor (SPF) 30 broad- spectrum sunscreen at the beginning of three consecutive summer vacations. Children given sunscreen had significantly smaller increases in number of pigmented nevi than children not given sunscreen.8 This result has been corroborated by a more broadly based, community intervention trial in Perth, Australia.9 Number of solar keratoses (SKs) is similarly a very strong predictor of keratinocyte cancer, and SKs are precursor lesions to cutaneous SCC. In an individually randomized, placebo-controlled trial of summer use of a broad-spectrum SPF 17 sunscreen in Melbourne, Australia, use of the active sunscreen reduced appearance of new SKs and increased remissions of existing SKs.10 An individually randomized controlled trial of daily use of an SPF 15+ sunscreen in southeastern Australia observed a significant, 39% reduction in risk of cutaneous SCC over 4.5 years of follow-up in those randomized to sunscreen.11 At approximately 15 years of follow-up, the relative risk for melanoma (both in situ and invasive) was 0.50 (95% CI: 0.24–1.02); for invasive melanoma alone it was 0.27 (95% CI: 0.08–0.97).12 Further follow-up for BCC has not been reported. The evidence of a plateau and nascent fall in melanoma incidence rates, particularly in younger people, in some populations with active and long-running sun protection programs (e.g., Australia and New Zealand) suggests that these programs have been effective in preventing melanoma. On balance, there is reasonable evidence that effective protection from exposure to the sun can reduce risk of cutaneous SCC and melanoma. It is also reasonable to conclude that measures demonstrated to provide that protection can also reduce risk of these cancers. Sun Protection Measures Public UV protection messages usually focus on these key points: • Wear clothing that covers your arms and legs. • Use sunscreen with an SPF of 30 or higher, and both UVA and UVB protection. • Wear a hat with a wide brim to shade your face, head, ears, and neck. • Stay in the shade when the UV Index is 3 or more. • Wear sunglasses that wrap around and block both UVA and UVB rays. • Avoid indoor tanning. In Australia, these messages have been framed in a well-known slogan as “Slip” on sun protective clothing; “Slop” on SPF30 or higher broad-spectrum, water-resistant sunscreen 20 minutes before going outdoors and reapply every two hours afterward; “Slap” on a broad- brimmed hat that protects your face, head, neck, and ears; and the more recent addition of “Seek” shade and “Slide” on sunglasses. Recommendations for when and where sun protection is required are complicated by the fact that solar UV radiation varies by latitude, season of the year, and time of day. UV radiation is strongest at solar noon and increases closer to summer solstice and with decreasing latitude. Thick cloud cover can rapidly reduce ambient UV levels; however, modeling of satellite cloud data and ozone levels enables accurate UV forecasts. Real-time UV levels are also available for most capital cities in Australia.13 Education about the UV Index is therefore one means of simplifying the sun protection message for the community. The higher the UV Index value, the greater the potential for damage to one’s skin. Sun protection is recommended when the UV Index is 3 or above.14 Internationally, a range of specific recommendations for sun protection and sun avoidance are made for skin cancer prevention, with some regions incorporating the UV Index while for others it is less of a focus. A more rigorous recommendation for sunscreen application, daily application when the UV Index is 3 or greater, has recently been adopted by skin cancer prevention programs in Australia.15 Sun safety messages in the United States recommend using sun protection and avoiding prolonged exposure to the sun when possible.16 In the United Kingdom, use of multiple sun protection behaviors are recommended when the sun is strong.17 31 32 SECTION I Factors in Cancer Risk and Prevention Psychosocial and Behavioral Research on Sun Exposure and Sun Protection Predictors of Sun Exposure and Sun Protection Behaviors Like most other health-related behaviors, changing personal sun exposure and sun protection behaviors is challenging. Glanz and Rimer18 highlight some key cognitive-behavioral concepts that underlie the complexity of changing personal behavior: • Behavior is mediated by cognitions; that is, what people know and think affects how they act. • Knowledge is necessary for, but not sufficient to produce, most behavior changes. • Perceptions, motivations, skills, and the social environment are key influences on behavior. Knowledge of the dangers of excessive sun exposure and the benefits of sun protection does not necessarily translate into behavior change; it is clear that practical, social, psychological, and environmental barriers must also be addressed. A large cross-sectional online survey conducted by the melanoma genetics consortium (GenoMEL) of 8,178 individuals across Europe (73% of respondents), Australia (12%), the United States (7%), and Israel (2%) found that despite widespread dissemination of sun protection messages, half of all respondents and 27% of those with a previous melanoma reported at least one severe sunburn during the previous 12 months.19 They found the strongest predictors of lack of sun protection behavior were perceived barriers to protection (β = −0.44) and perceiving suntans as “attractive” and “healthy” (β = −0.16). Perceived vulnerability to melanoma, perceived melanoma severity, and worry about melanoma showed moderate positive correlations with sun protection. Sun protection was also higher among women, older people, people with high-risk characteristics such as fair skin, and people living at lower latitudes. Importantly, the pattern of results was similar across the different countries, and the same variables were significant predictors in all regions.19 These demographic predictors of sun protection are also generally consistent with those identified in nationally representative surveys in the United States20 and Australia.21 Young people are a particularly challenging group in whom to motivate risk-reducing behaviors and their protective behaviors remain low. Among adolescents and young adults, suntanning and sun exposure behavior is strongly influenced by social norms, body image concerns, and fashion. Younger people are more likely to perceive a tan as attractive and healthy and to perceive barriers to sun protection. Despite knowing the risks, many adolescents and young adults still desire and actively seek a suntan. Frequent users of indoor tanning often display signs and symptoms consistent with an addictive disorder, suggesting the need to address physiologic effects of tanning as well as psychosocial factors driving tanning behavior. In contrast, young children are generally better protected from the sun than adolescents or adults, while parental attitudes and parental behavior are strongly associated with children’s sun exposure and sun protection behavior. Health Behavior Theories Skin cancer prevention programs are most likely to be effective and sustainable when they are based on theoretical models that underpin a clear understanding of the factors that influence the target population’s behavior, including their personal beliefs and attitudes, and social and physical environments. In that regard, application of theory informs the interpretation of research findings and understanding of the dynamic interactions between behavior and the environmental and social context, hence increasing the likelihood of effective translation of skin cancer prevention strategies into public health policy or clinical practice. Theories and their applications at the individual (intrapersonal), interpersonal, and community levels are relevant to skin cancer prevention. Individual-Level (Intrapersonal) Theories Strategies intended to change people’s behavior can often be derived from individual- level theories such as the Health Belief Model, Stages of Change Model, Theory of Planned Behavior, and Precaution Adoption Process Model. These theories are important for skin cancer prevention campaigns as they are related to individuals’ knowledge, perceptions, attitudes, beliefs, intentions, and decision-making processes about skin cancer prevention behaviors, barriers, and benefits. However, on their own, individual-level theories are usually insufficient to change skin cancer–related behaviors; to be effective they need to be considered within the social and environmental context. Community-Level Theories Some population-based strategies for reducing sun exposure focus on changing the environment and thus draw on community-level theories such as Community Organization, which emphasizes community-driven approaches to assessing and solving health and social problems. An example is the use of shade sails in schools, whereby the health promotion agency, schools, and students work together to identify the problem of lack of shade, decide where best to locate the structures, and raise the funds required for them. Disparities in provision of shade in public parks may also be addressed in a similar manner through advocacy of community groups to local governments.22,23 Mass media campaigns that aim to change social norms, such as tanning desirability, also draw on community- level theories. Institutional factors, such as sun protection policies for outdoor workers and primary school students, have also been successful for reducing childhood sun exposure, as has advocacy aimed at legislative changes, which have been shown to be highly successful in achieving the bans on commercial indoor tanning in Australia and Brazil.14,24 Interpersonal-Level Theories Theories at the interpersonal level, such as Social Cognitive Theory, explore the reciprocal exchanges between individuals and their social environments including family members, friends, health professionals, and others. Interpersonal theories recognize that the social environment influences an individual’s feelings and behaviors. They are particularly useful for examining and understanding behaviors that relate to skin cancer risk, including sun protective behaviors, time spent in the sun, and suntanning, as these behaviors have a strong social influence component. For example, in many societies, having a suntan is seen as being healthy and attractive, and people do not cover up because of peer pressure and fashion trends. As such, strategies based on interpersonal-level theories appear to CHAPTER 4 Sun Exposure and Cancer Risk be the most powerful and effective for skin cancer prevention. The Australian SunSmart program14 is an example of a skin cancer prevention strategy that is based on social cognitive theories of attitudes and behavior change; it targets knowledge, attitudes, and intentions of individuals, along with social and cultural norms and environmental change, to promote sun protection behavior. Interventions and Approaches Table 4.1 summarizes some of the psychosocial and behavioral skin cancer prevention interventions and approaches for different target groups: general population, early childhood, adolescence, and other high-risk groups. This table does not provide an exhaustive list but gives an indication of the multidisciplinary and multidimensional approaches that are needed to reduce skin cancer risk across different community groups. More details of these and other interventions can be found in systematic reviews of these topics.25–31 Given our increasing understanding of the genetic susceptibility to melanoma, advances in genetic technologies, and the increasing use of genomic information for health purposes, future interventions should also consider evaluating the effect of interventions that provide knowledge of personal genetic melanoma risk on motivating risk-reducing behaviors in the general population. Social and behavioral theory suggests that the highly personalized nature of providing results of a genetic risk assessment may increase its motivational potency over standard approaches. Table 4.1. Summary of Psychosocial and Behavioral Skin Cancer Prevention Interventions and Approaches for Different Target Groups Target Group Aims Types of Interventions and Approaches Examples General population - To increase knowledge and awareness - Mass media public education, such as - Slip! Slop! Slap! (Seek! Slide!), - - - - - - - - Early childhood - To reduce children’s sun exposure and - - - - - Adolescence of skin cancer and its causes To improve sun protection behaviors To reduce excessive sun exposure To improve early detection of melanoma To identify high-risk individuals who would benefit from closer follow-up and surveillance To provide information on prevention, early diagnosis, and treatment of skin cancer To reduce the desirability of a tan To increase knowledge of harmful UV levels increase their sun protection behaviors To improve parents’/teachers’/children’s knowledge and awareness of skin cancer, risk factors, and sun protection strategies To improve parents’ and teachers’ self- efficacy, such as confidence in their ability to increase their child/children’s sun protection To reduce perceived barriers to sunscreen use and other forms of sun protection To change attitudes To increase sun protection policies at schools - To change perceptions and attitudes, such as tanning desirability - To increase awareness that young people are susceptible to skin cancer - To reduce sun exposure and increase sun protection intentions and behaviors - To reduce or stop indoor tanning - - - - - Targeted interventions that are implemented in environments such as childcare centers, preschools, and primary schools, such as promoting hat wearing, ensuring adequate shade in the school grounds, and teaching children about sun protection in school - Protective clothing and swimwear designed for children - “Toolkits” for policy development, such as evaluation tools to assess schools’ current sun protection practices and shade availability, aids for curricula training and integration into the classroom, and sun protection policy options - Newsletter information and resources - Mass media campaigns aimed at - - - - - Other high-risk groups - To reduce sun exposure and increase (e.g., genetically sun protection in people at “high risk” of susceptible, high developing skin cancer occupational - To increase early detection among sun exposure, high-risk groups immunosuppressed) - To increase knowledge about skin cancer risk and risk-reducing behaviors advertisements on TV, radio, online/digital media, newspapers, and billboards Protective clothing for swimming, golf, cycling, and gardening and new shade structures for home and beach use Free-of-charge skin examinations for the public Providing shade in public areas1 and at pools/beaches Interventions in recreational or tourism settings Tools to assist with and dissemination of real-time measures of UV for specific locations young adults Increasing shade cover at high schools Use methods such as UV photos, Interventions based on the appearance- damaging effects of UV exposure Policy-based interventions including changes to legislation Coordinated, multi-level, trans-disciplinary approaches to reduce indoor tanning - Genetic screening for people with a strong family history - Workplace policies for outdoor workers - Providing shade in occupational settings - Family-based strategies within melanoma- prone families SunSmart, Euromelanoma prevention campaign, Dear 16- YearOld Me; Wes Bonny testimonial, UV It All Adds Up, Don’t Be A Lobster - Pool Cool, Go SunSmart - Smartphone apps - SunSmart Schools program, SunWise school campaign, Kidskin - SunSmart primary schools members program - Tailored mailed newsletters - Wes Bonny testimonial; Dark Side of Tanning; Tattoo (SunSmart Dear 16YearOld Me - Built shade interventions - Interventions aimed at relatives of people with melanoma - Go Sun Smart Worksite Health Communication Campaign, SUNWISE workplace intervention 33 34 SECTION I Factors in Cancer Risk and Prevention The Vitamin D Conundrum In a sun exposure and cancer risk context, there are four main reasons that vitamin D is important to psycho-oncology. First, vitamin D is essential to healthy bone development and to maintaining bone health. Second, vitamin D is hypothesized to have health benefits that are independent of these skeletal effects. Although evidence is mixed and of variable quality, vitamin D possibly protects against some cancers, infections, autoimmune and cardiovascular diseases, cognitive dysfunction, and depression,32 while a recent high-quality study provides evidence of reduced cancer mortality6. Third, irradiation of skin by solar UVB radiation prompts vitamin D synthesis and makes an important contribution to serum 25-hydroxy vitamin D concentration (the usual measure of vitamin D status) in most populations, with diet (including vitamin D–fortified foods and vitamin D supplements) being the alternative source. Fourth, vitamin D insufficiency (30–<50 nmol/L of 25-hydroxy vitamin D) and frank deficiency (<30 nmol/L) are quite common in fair-skinned as well as dark-skinned populations (who generally have lower average serum 25-hydroxy vitamin D concentrations), even in Australia, where the potential for exposure of a largely fair-skinned population to solar UVB radiation is probably the greatest in the world. Messages about the health benefits of vitamin D and harmful effects of sun exposure are therefore in conflict, and health professionals and their clients are easily confused about how much vitamin D is required, whether it can be safely acquired by prudent sun exposure, and whether more dietary fortification with, or supplemental intake of, vitamin D is a necessary and safer course or whether some beneficial effects of sun exposure can only be obtained from sun exposure itself. The issue has been additionally clouded by some scientists’ promotion of a need for particularly high serum 25-hydoxy vitamin D concentrations to ensure vitamin D’s hypothesized beneficial effects against nonskeletal conditions.33 Additionally, some have raised concern that sunscreens inhibit vitamin D production,34 while application of sunscreen is typically inadequate and is still recommended for use when the UV Index is 3 or above.34 Content analysis of news media reports about sun protection issues showed increasing coverage of vitamin D issues from 2001 to 2012.35 Recent quantitative and qualitative surveys in the community have shown low knowledge about vitamin D and lack of understanding of the “balance message”: that is, “some sunlight exposure each day for adequate vitamin D production, but not so much that would lead to increased skin cancer risk.”36 Moreover, some people reported going out into the sun or intentionally tanning to improve their vitamin D status.37 Publication of findings from vitamin D supplementation trials and studies of health benefits of UV exposure during this period appear to have influenced physicians’ practices with respect to testing of patients’ vitamin D levels and prescribing both sun exposure and vitamin D supplements.38 Physicians surveyed in 2012 also reported a trend toward recommending less sun protection in the winter and, sometimes, in the summer. It is of concern that some doctors endorsed sun exposure during peak UV times of the day, in the summer as well as in the winter, and commonly overestimated the amount of peak UV exposure in the summer that was needed to gain or maintain vitamin D sufficiency. Some doctors also expressed greater concern about vitamin D deficiency than skin cancer in their patients. A number of interested agencies, including national public health agencies, cancer and osteoporosis organizations, and professional organizations of dermatologists, have reviewed the changing evidence about vitamin D and health and considered what, if any, might be an acceptable balance between sun exposure that is sufficient to maintain vitamin D sufficiency and protection against skin cancer due to sun exposure.39 These guidelines inevitably vary depending on the latitude of the population they address; typically they advise sun protection during the middle of the day in the summer, when UVB levels are at their highest, and little or no sun protection at times and in seasons when UVB levels are low (characterized by a UV Index of less than 3). Some, particularly dermatology societies, favor no relaxation in sun protection and increased dietary intake or supplementation to ensure vitamin D sufficiency, while others call for more nuanced messaging to account for duration as well as intensity of UV exposure.40 The research on which these recommendations have been based is very limited, particularly with respect to how much sun exposure will maintain vitamin D sufficiency, and none of the recommendations have been evaluated for their success in reducing vitamin D insufficiency and not increasing skin cancer risk.41 REFERENCES 1. Gandini S, Dore JF, Autier P, Greinert R, and Boniol M. Epidemiological evidence of carcinogenicity of sunbed use and of efficacy of preventive measures. J Eur Acad Dermatol Venereol. 2019; 33(Suppl 2): 57–62. 2. Colantonio S, Bracken MB, and Beecker J. The association of indoor tanning and melanoma in adults: systematic review and meta-analysis. J Am Acad Dermatol. 2014; 70: 847–857, e1–18. 3. Nilsen LT, Hannevik M, and Veierod MB. Ultraviolet exposure from indoor tanning devices: a systematic review. Br J Dermatol. 2016; 174: 730–740. 4. Lucas RM, McMichael AJ, Armstrong BK, and Smith WT. Estimating the global disease burden due to ultraviolet radiation exposure. Int J Epidemiol. 2008; 37: 654–667. 5. Mondul AM, Weinstein SJ, Layne TM, and Albanes D. Vitamin D and cancer risk and mortality: state of the science, gaps, and challenges. Epidemiol Rev. 2017; 39: 28–48. 6. Zhang Y, Fang F, Tang J, et al. Association between vitamin D supplementation and mortality: systematic review and meta-analysis. BMJ. 2019; 366: 14673. 7. Neale RE, Armstrong BK, Baxter C, et al. The D-health trial: a randomized trial of vitamin D for prevention of mortality and cancer. Contemp Clin Trials. 2016; 48: 83–90. 8. Gallagher RP, Rivers JK, Lee TK, Bajdik CD, McLean DI, and Coldman AJ. Broad-spectrum sunscreen use and the development of new nevi in white children: a randomized controlled trial. JAMA. 2000; 283: 2955–2960. 9. Armstrong BK, Vajdic CM, and Cust AE. Melanoma. In: Thun MJ, Linet MS, Cerhan JR, Haiman CA, and Schottenfeld D (eds.). Schottenfeld and Fraumeni Cancer Epidemiology and Prevention. 4th ed. New York: Oxford University Press, 2018, pp. 1061–1087. 10. Thompson SC, Jolley D, and Marks R. Reduction of solar keratoses by regular sunscreen use. N Engl J Med. 1993; 329: 1147–1151. CHAPTER 4 Sun Exposure and Cancer Risk 11. Green A, Williams G, Neale R, et al. Daily sunscreen application and betacarotene supplementation in prevention of basal-cell and squamous-cell carcinomas of the skin: a randomised controlled trial. Lancet. 1999; 354: 723–729. 12. Green AC, Williams GM, Logan V, and Strutton GM. Reduced melanoma after regular sunscreen use: randomized trial follow- up. J Clin Oncol. 2011; 29: 257–263. 13. ARPANSA. Ultraviolet radiation index: realtime UV index. Australian Radiation and Nuclear Safety Protection Agency, 2019, Realtime UV data for Australian cities [cited 2019 Sep 16]: Available from https://www.arpansa.gov.au/our-services/monitoring/ ultraviolet-radiation-monitoring/ultraviolet-radiation-index 14. Sinclair C and Foley P. Skin cancer prevention in Australia. Br J Dermatol. 2009; 161 Suppl 3: 116–123. 15. Whiteman D, Neale RE, Aitken J, et al. When to apply sunscreen: a consensus statement of Australia and New Zealand. Aust N Z J Public Health. 2019; 43: 171–175. 16. Centers for Disease Control and Prevention. Skin Cancer: SunSafety; 2019 [cited 2019 Sep 30]: Available from https://www. cdc.gov/cancer/skin/basic_info/sun-safety.htm 17. Cancer Research UK. Sun, UV and cancer; 2019 [cited 2019 Sep 30]: Available from https://www.cancerresearchuk.org/about- cancer/causes-of-cancer/sun-uv-and-cancer 18. Glanz K and Rimer BK. Theory at a Glance: A Guide for Health Promotion Practice. 2nd ed. US National Cancer Institute, 2005. Bethesda, Maryland, USA. 19. Branstrom R, Kasparian NA, Chang YM, et al. Predictors of sun protection behaviors and severe sunburn in an international online study. Cancer Epidemiol Biomarkers Prev. 2010; 19: 2199–2210. 20. Buller DB, Cokkinides V, Hall HI, et al. Prevalence of sunburn, sun protection, and indoor tanning behaviors among Americans: review from national surveys and case studies of 3 states. J Am Acad Dermatol. 2011; 65: S114–S123. 21. Dobbinson S, Wakefield M, Hill D, et al. Prevalence and determinants of Australian adolescents’ and adults’ weekend sun protection and sunburn, summer 2003-2004. J Am Acad Dermatol. 2008; 59: 602–614. 22. Anderson C, Jackson K, Egger S, Chapman K, and Rock V. Shade in urban playgrounds in Sydney and inequities in availability for those living in lower socioeconomic areas. Aust N Z J Public Health. 2014; 38: 49–53. 23. Buller DB, English DR, Buller MK, et al. Shade sails and passive recreation in public parks of Melbourne and Denver: a randomized intervention. Am J Public Health. 2017; 107: 1869–1875. 24. Gandini S, Sera F, Cattaruzza MS, et al. Meta-analysis of risk factors for cutaneous melanoma: II. Sun exposure. Eur J Cancer (Oxford, Engl. 1990). 2005; 41: 45–60. 25. Horsham C, Auster J, Sendall MC, et al. Interventions to decrease skin cancer risk in outdoor workers: update to a 2007 systematic review. BMC Res Notes. 2014; 7: 10. 26. Saraiya M, Glanz K, Briss P, et al. Interventions to prevent skin cancer by reducing exposure to ultraviolet radiation: a systematic review. Am J Prev Med. 2004; 27: 422–466. 27. Sandhu PK, Elder R, Patel M, et al. Community-wide interventions to prevent skin cancer: two community guide systematic reviews. Am J Prev Med. 2016; 51: 531–539. 28. Persson S, Benn Y, Dhingra K, Clark-Carter D, Owen AL, and Grogan S. Appearance-based interventions to reduce UV exposure: a systematic review. Br J Health Psychol. 2018; 23: 334–351. 29. Stapleton JL, Hillhouse J, Levonyan-Radloff K, and Manne SL. Review of interventions to reduce ultraviolet tanning: need for treatments targeting excessive tanning, an emerging addictive behavior. Psychol Addict Behav. 2017; 31: 962–978. 30. Williams AL, Grogan S, Clark- Carter D, and Buckley E. Appearance-based interventions to reduce ultraviolet exposure and/or increase sun protection intentions and behaviours: a systematic review and meta-analyses. Br J Health Psychol. 2013; 18: 182–217. 31. Holman DM, Fox KA, Glenn JD, et al. Strategies to reduce indoor tanning: current research gaps and future opportunities for prevention. Am J Prev Med. 2013; 44: 672–681. 32. Autier P, Mullie P, Macacu A, et al. Effect of vitamin D supplementation on non-skeletal disorders: a systematic review of meta- analyses and randomised trials. Lancet Diabetes Endocrinol. 2017; 5: 986–1004. 33. Institute of Medicine. Dietary Reference Intakes for Calcium and Vitamin D. Washington, DC: National Academies Press, 2011. 34. Mancebo SE, Hu JY, and Wang SQ. Sunscreens: a review of health benefits, regulations, and controversies. Dermatol Clin. 2014; 32: 427–438. 35. Scully M, Makin J, Maloney S, and Wakefield M. Changes in coverage of sun protection in the news: threats and opportunities from emerging issues. Health Educ Res. 2014; 29: 378–387. 36. Bonevski B, Bryant J, Lambert S, Brozek I, and Rock V. The ABC of vitamin D: a qualitative study of the knowledge and attitudes regarding vitamin D deficiency amongst selected population groups. Nutrients. 2013; 5: 915–927. 37. Youl PH, Janda M, and Kimlin M. Vitamin D and sun protection: the impact of mixed public health messages in Australia. Int J Cancer. 2009; 124: 1963–1970. 38. Bonevski B, Girgis A, Magin P, Horton G, Brozek I, and Armstrong B. Prescribing sunshine: a cross-sectional survey of 500 Australian general practitioners’ practices and attitudes about vitamin D. Int J Cancer. 2012; 130: 2138–2145. 39. Australian and New Zealand Bone and Mineral Society, Australasian College of Dermatologists, Cancer Council Australia, Endocrine Society of Australia, and Osteoporosis Australia. Position Statement—Risks and Benefits of Sun Exposure. Sydney, Australia: Cancer Council Australia, 2016. 40. Lucas RM, Neale RE, Madronich S, and McKenzie RL. Are current guidelines for sun protection optimal for health? Exploring the evidence. Photochem Photobiol Sci. 2018; 17: 1956–1963. 41. Khan SR, Whiteman DC, Kimlin MG, et al. Effect of solar ultraviolet radiation exposure on serum 25(OH)D concentration: a pilot randomised controlled trial. Photochem Photobiol Sci. 2018; 17: 570–577. 35 5 Psychosocial Factors Anika von Heymann and Christoffer Johansen Introduction This topic covers many risk factors, and the scientific literature on their role in cancer causation has a long history. Nevertheless, in scientific studies published during the past 75 years and in reviews of the literature, little is said about cancer causation as such. It is important that this subject be raised to emphasize that the criteria for causation originally stated by Austin Bradford Hill in the early 1960s also apply to psychosocial risk factors. Hill1 suggested that the following issues be considered to distinguish association from causation: temporality, coherence, consistency, specificity, biological gradient, strength of the association, analogy, and biological plausibility. Hill stated, “None of these nine standards can bring indisputable evidence for or against the cause-and-effect hypothesis and none can be required as a sine qua non.”1 This cannot be considered a final list of factors that establish causality, but they must be taken into account in determining the weight of evidence. Hill also warned against overemphasis on statistical significance testing, writing, “The glitter of the t table diverts attention from the inadequacies of the fare.”1 The theory of causality has developed since Hill’s time, increasing the complexity of establishing causality and requiring new levels of sophistication in epidemiology.2 The overwhelming body of the scientific literature on psychosocial risk ignores fundamental methodological issues. Taking this statement further, one can say that psychosocial risk factors can be addressed only in studies designed for that purpose. Recall bias, lack of adjustment for disease-specific factors or comorbidity, use of biased data sources, interviewer bias, and other issues have often been ignored by researchers working in this area. Today, however, more studies in this area fully acknowledge the need for such considerations in the search for possible psychosocial risk factors for cancer. What Is a Risk Factor for Cancer? Researchers on psychosocial risk factors for cancer should refer to the guidelines for cancer causation published by the International Agency for Research in Cancer (IARC). Since 1969, working groups convened by the IARC have assessed the degree of evidence for the carcinogenicity to humans of some 800 biological, physical, chemical, and occupational factors and have ranked risk factors for cancer according to the degree of evidence for causality. Psychological factors have not been reviewed in this context, but a brief review of psychological factors associated with cancer was included in another IARC publication on cancer prevention and control in 1990.3 No conclusion was reached about the degree of evidence, but it was stated that future epidemiological studies should include standardized instruments and data on potential confounders to be credible. The evaluation of psychosocial factors in the causation of cancer must be addressed in the context of certain methodological problems, which are summarized briefly to follow on the basis of the preamble to the IARC Monographs.3 First, the study population, disease, and exposure should have been well defined by the authors. Cases of disease in the study population should have been identified in a way that was independent of the exposure of interest—in this case, the psychological factor. The exposure should have been assessed in a way that was not related to disease status. Second, the authors should have taken into account other variables that can influence the risk for disease and that might have been related to the exposure of interest. Potential confounding by such variables should have been dealt with either in the design of the study, such as by matching, or in the analysis, by statistical adjustment. In cohort studies, comparisons with local rates of disease may or may not be more appropriate than those with national rates. Internal comparisons of frequency of disease among individuals at different levels of exposure are also desirable in cohort studies, as they minimize the potential for confounding related to the difference in risk factors between an external reference group and the study population. Third, the authors should have reported the basic data on which the conclusions are founded, even if sophisticated statistical analyses were employed. At the very least, they should have given the numbers of exposed and unexposed cases and controls in a case–control study and the numbers of cases observed and expected in a cohort study. Further tabulations by time since exposure began and other temporal factors are also important. In a cohort study, data on all cancer sites and all causes of death should have been given, to reveal the possibility of reporting bias. In a case–control study, the effects of investigated factors other than the exposure of interest should have been reported. CHAPTER 5 Psychosocial Factors Finally, the statistical methods used to obtain estimates of relative risk, absolute rates of cancer, confidence intervals, and significance as well as the methods used to adjust for confounding should have been clearly stated by the authors. When an agent is discussed by the independent working groups convened by the IARC, evidence from studies in humans and experimental animals is evaluated together, and the strength of the mechanistic evidence is also characterized. The groups then decide on the strength of the association between the exposure under review and carcinogenicity. An agent is assigned to one of five groups:3 Group 1: The agent is carcinogenic to humans. Group 2A: The agent is probably carcinogenic to humans. Group 2B: The agent is possibly carcinogenic to humans. Group 3: The agent is not classifiable as to its carcinogenicity to humans. Group 4: The agent is probably not carcinogenic to humans. These considerations should be of interest to clinicians and researchers working in the field of psychosocial risk factors for cancer. They stimulate attention to the design and interpretation of scientific studies and form part of the discussion of the current state of the art in this intriguing area of research. Research on Psychosocial Risk Factors for Cancer The three psychosocial factors that have been most rigorously studied in investigations of psychosocial cancer risks are major life events or stress, depression or depressive mood, and personality or personality traits. The commentary in this chapter does not represent a systematic review; it covers only studies conducted as prospective or retrospective cohort studies and case–control studies in which the information on psychosocial variables was collected independently of the outcome, thereby reducing the possibility of selection and recall bias. The findings of some of the best-designed studies published since 2000 are summarized briefly to follow. Major Life Events and Stress Numerous studies have investigated the association between major life events, stress in daily life, or work-related stress and the risk for cancer. Breast cancer has been a focus in this research tradition. With regard to work-related stress, a large Finnish prospective cohort study of 10,519 women aged 18 years or more investigated the relation between stress in daily activities and breast cancer.4 Daily stress was assessed twice, in 1975 and 1981, by a self-administered questionnaire, and study subjects were divided into three groups: no stress (23% of women), some stress (68%), and severe stress (9%). The authors identified 205 incident cases of breast cancer by linkage to the nationwide, population-based cancer registry and observed that the hazard ratio for breast cancer in women with “some stress” was 1.11 (95% confidence interval [CI], 0.78–1.57) when compared with women with “no stress.” For women with “severe stress,” the hazard ratio for breast cancer was 0.96 (95% CI, 0.53–1.73) when compared with those with “no stress.” The analysis included detailed information on reproductive factors, anthropometrics, and lifestyle. Neither shifting the cutoff point for stress nor restricting the analysis to women who reported the same level of stress at the two measurements altered the results.4 The same group investigated the same hypothesis in a cohort of 10,808 women sampled in the Finnish Twins Registry and obtained information on exposure to life events by using a standardized life event inventory.5 They examined the effect of accumulation of life events, placing emphasis on events experienced five years before completion of the questionnaire. They observed that the experience of divorce or the death of a husband was followed by a significant twofold increase in the hazard ratio for breast cancer; the death of a close relative or friend also significantly increased the risk by almost 40%. The analysis included some information on reproductive factors, anthropometrics, and lifestyle. It is notable that the authors investigated the effect of cumulative exposure to life events.5 In the Nurses’ Health Study of 69,886 women aged 46–71 at baseline in the United States, the women answered questions on informal caregiving.6 The authors hypothesized that hours and self-reported levels of stress from informal caregiving would be associated with breast cancer incidence. A total of 1,700 incident cases of breast cancer were identified between 1992 and 2000, in which period the women reported caregiving twice, in 1992 and 1996. The analysis included information on reproductive factors, family history of breast cancer, psychosocial factors such as depressive symptoms, social network and self-reported level of stress, anthropometrics, and lifestyle factors. The authors did not find that stress due to caregiving increased the risk for breast cancer.6 Two reports based on the Nurses’ Health Study in the United States and the Danish Nurse Cohort study of job stress and risk for breast cancer found no increase in the risk of women who reported high levels of strain in their daily working lives.7,8 Both studies were conducted as cohort studies, with morbidity from breast cancer as the outcome and adjustment for a number of factors of relevance for breast cancer risk. The Danish study also investigated the association between stress and stage of disease at diagnosis but did not find that stress affected the prognostic characteristics of disease. More recent studies that included carefully planned follow-up in cohorts with detailed information on other potential risk factors also do not find that stress causes cancer. In a cohort of 84,334 members of the Women’s Health Initiative in the United States, a total of 2,841 invasive breast cancer cases were observed from an average of 7.6 years of follow-up. One stressful life event was associated with breast cancer; however, risk decreased with each additional stressful life event.9 In a cohort of 11,467 women in the United Kingdom, a total of 313 cases of incident breast cancer was observed. The authors reported no association with these cases and the reporting of a summary of social adversities adjusted for well-known risk factors for breast cancer.10 Of interest to this discussion is a study that included members of the Danish resistance movement during the German occupation of Denmark between April 9, 1940, and May 5, 1945. Their resistance included illegal press activities, intelligence services, collection of parachuted containers of weapons, sabotage of factories and railways valuable for Germany, and operation of illegal boats to Sweden (for Danish Jews in October 1943). Between June 1943 and liberation, 1,547 male members of the resistance movement were arrested, interrogated, exposed to severe mental and physical strain, and deported to German concentration camps as political prisoners. The survivors continued to have excess somatic and mental morbidity.11 37 38 SECTION I Factors in Cancer Risk and Prevention Among all male political prisoners (n = 1,547), slightly higher cancer incidence and mortality than national rates were found, driven mainly by increased ratios for smoking-or alcohol-related cancers. The study had complete information on exposure status and follow- up for cancer incidence and death at the individual level for the entire study period.12 In terms of prognosis, a meta-analysis found that stressful life experiences before the diagnosis of cancer were associated with shorter survival among cancer patients, albeit the effect was small (hazard ratio [HR] 1.15; 95% CI, 1.06–1.24).13 After the diagnosis, stress experienced by cancer patients is influenced by numerous factors, stemming from before the diagnosis, over the diagnostics and treatment of cancer, to the immediate and late somatic and psychosocial effects of cancer and its treatment. At any given time, there is likely to be an increased risk for late detection of recurrence and mortality determined by multiple risk factors, if a patient is stressed across all meanings of this word. For several cancers, it is known that clinical depression, which can be detected in almost 20% of cancer patients within the first 5 years after diagnosis, must be connected to the exposures provided by stress associated with the cancer trajectory as such. It is quite difficult to disentangle the multiple sources and determine a causal relationship not confounded residually. Cancer-specific, somatic, and social circumstances play a role in this effect. In light of the results within depression or personality traits as prognostic factors in cancer, stress may also act as a prognosticator. However, not many studies have been published in this area. In summary, very few of the large population-based studies in which information on exposure is collected independently of scientific hypotheses found evidence that work-related stress or major life events are associated with an increased risk for cancer. Although a few well-designed studies have shown an increased risk,5,12,14 the strong consistency among the presumably unbiased studies indicates that explanations for the elevated risk may include selection bias, residual confounding, or chance. This conclusion comes close to the overall conclusion in several reviews.15,16 However, the publication and frequent citation of a review in 2008, which included some debatable criteria, illustrates how much belief and methodological decisions in reviewing scientific literature also influence the judgment of the association between stress and cancer risk and prognosis.13 The review found that stress-related psychosocial factors, but not stressful life experiences or chronic or daily stress, were related to cancer incidence. This extensive review and meta-analysis has been widely cited despite the inclusion of research that has been discredited because the data may be flawed, a number of methodological mistakes in the entire construction of the analytical model, lack of controls in a high number of the studies often underpowered to detect the association, and the use of quite heterogeneous indicators of stress.17 Depression and Depressive Mood Another potential psychosocial risk factor that has been widely investigated is depression or depressive mood. In a nationwide Danish cohort study of the cancer risk of patients hospitalized for depression, all 89,491 adults who had been admitted to a hospital with depression, as defined in the International Classification of Diseases, Eighth Revision,18 between 1969 and 1993 were identified. A total of 9,922 cases of cancer were diagnosed in the cohort, with 9,434.6 expected, yielding a standardized incidence ratio of 1.05 (95% CI, 1.03–1.07). The risk for cancer increased during the first year after hospital admission, with brain cancer in particular occurring more frequently than expected. When the first year of follow-up was excluded, the increase was attributable mainly to an increased risk for tobacco-related cancers, with standardized incidence ratios for non- tobacco-related cancers of 1.00 (95% CI, 0.97–1.03) after 1–9 years of follow-up and 0.99 (95% CI, 0.95–1.02) after 10 or more years of follow-up. These findings provide no support for the hypothesis that depression independently increases the risk for cancer, but they emphasize the deleterious effect that depression can have on lifestyle.18 In a Dutch prospective follow-up study of 5,191 women living in Eindhoven and born in the Netherlands between 1941 and 1947, all the participants answered a questionnaire about the presence of depressive symptoms measured on the Edinburgh Depression Scale.19 The outcome was morbidity from cancer recorded in the regional cancer registry, which reported incident breast cancer cases up to five years after the questionnaire screening. The analyses were adjusted for information on 15 demographic, medical, and lifestyle factors known to be associated with the risk for breast cancer. Breast cancer was diagnosed in 58 women during the follow-up period, yielding an odds ratio of 0.29 (95% CI, 0.09–0.92), which suggested that depressive symptoms may be protective against breast cancer.19 The same design was used in a study in Finland in which 10,892 women aged 48–50 years at the time of inclusion were followed up for breast cancer 6–9 years later.20 The questionnaire included items on depression, personality traits, attitudes toward illness, life events, and health history. The incident cancer cases were obtained from the nationwide Finnish cancer registry, which has almost complete population-based coverage. The multivariate analysis controlled for socioeconomic factors, family history of cancer, parity, and health behavior, and identified a nonsignificant increased risk of 1.15 (95% CI, 1.0–1.28) for breast cancer among women aged 50–59 when compared with the general population. There was no evidence that depression, anxiety, cynical distrust, or coping increased the risk for cancer.20 A large prospective study was conducted of the association between depressive symptoms as measured by the Mental Health Index (MHI) and risks for colorectal cancer and colorectal adenomas.21 Women (who were all part of the Nurses’ Health Study) who scored between 0 and 52 on a 0–100 scale were defined as having significant depressive symptomatology. A total of 33 of the 400 cases of colorectal cancer filled this definition, as did 45 of 680 cases of distal adenoma. The authors created other categories across the range of MHI scores and reported a nonsignificant elevated risk of 1.43 (95% CI, 0.97–2.11) for colorectal cancer in women with the highest score on the index, with a stronger association in overweight women. Depressive symptoms did not increase the risk for adenomas.21 A large population-based record-linkage study in the Oxford National Health Service region in the United Kingdom included people who had been admitted to hospitals for depression or anxiety.22 A reference cohort of 525,436 persons was constructed by selecting records for admission for various other medical and surgical conditions as “controls.” The outcome was identified as either death from cancer or hospital care for any cancer. People who had the cancer at the first recorded admission for the psychiatric disorder or a comparison condition were excluded to avoid misclassification. CHAPTER 5 Psychosocial Factors The authors did not find an overall increase in the risk for cancer in either the 27,818 persons with depression or the 24,292 persons with anxiety. When the first year of follow-up was excluded, the relative risk for lung cancer was significantly increased in both cohorts (1.30; 95% CI, 1.14–1.48 in the depression cohort and 1.21; 95% CI, 1.03–1.36 in the anxiety cohort).22 A study from Taiwan using data from the National Health Research Institute identified all 8,419 newly diagnosed depression patients in the period 2000–2002 and compared the risk for cancer with all 67,352 (case/control ratio, 1/8) other cases claiming insurance but with no diagnosis of depression.23 The authors report no increased risk for cancer in this study adjusting for gender, age, occupation, urbanization, and comorbidity. Another study from Taiwan reported an increased risk for cancer in a cohort of 778 patients hospitalized for depression in the time period 1998–2003, together with 3,890 matched persons from the background population who had undergone minor abdominal surgery.24 The study found 61 cases of cancer among the severely depressed cohort members, finding significantly increased hazard ratios for gastrointestinal, genitourinary, and the group of “other” cancers. The small numbers and relatively broad categories make inference difficult. In terms of whether depression affects prognosis, a study including 45,325 Danish women with early breast cancer found that women who had previously been treated for depression (measured as antidepressant use) were at significantly greater risk of receiving nonguideline treatment (odds ratio [OR] 1.14; 95% CI, 1.03–1.27), as well as significantly greater risk of mortality (overall HR 1.21; 95% CI, 1.14–1.28; breast cancer–specific HR 1.11; 95% CI, 1.03–1.2).25 The analyses controlled for sociodemographics and important clinical factors (comorbidity, menopausal status, tumor size, positive axillary lymph nodes, estrogen receptor status, type of surgery, and treatment received). This study found that a prior depression may affect the cancer treatment received. A meta-analysis of 76 prospective studies found that depression, measured as a diagnosis of depression or depressive symptoms assessed in questionnaires, was a significant predictor of cancer mortality, albeit the effect was relatively small (average effect size 1.17; 95% CI, 1.12–1.22).26 In summary, in most of the studies in which total cancer risk was assessed, no statistically significant increase related to depression was seen, or increases were seen in only some strata. Increased risks were often observed for smoking-associated cancers such as lung cancer. Thus, it is the lifestyle of depressed people that is probably the most straightforward explanation for the positive findings in some studies, although a depressive component cannot be excluded. Further, depression may be associated with survival after a cancer diagnosis. Personality Traits A variety of personality traits have been investigated in relation to cancer incidence and progression. In a large Finnish study based on the aforementioned Twins Cohort, 12,032 women answered questions about life satisfaction and neuroticism in 1975 and 1981. During the 21 years of follow-up, 238 cases of breast cancer were identified in the Finnish Cancer Registry. The authors reported no association between the measures of life satisfaction, neuroticism, and the risk for breast cancer. Subsequent nested case–cohort analyses and analyses that included changes in the levels of neuroticism and life satisfaction at the two times led the authors to conclude that there was no evidence that breast cancer is more likely to occur in unhappy, dissatisfied, anxious women.27 This finding was confirmed in a large study of Swedish and Finnish twin cohorts including close to 60,000 persons. The authors observed a risk close to unity for any cancer (HR 0.99; 95% CI, 0.98–1.01 for extraversion, and HR 1.00; 95% CI, 0.99–1.02 for neuroticism).28 A smaller prospective cohort study in Germany of 5,114 women and men aged 40–65 measured mortality from and incidence of cancer.29 A number of personality scales were included in a questionnaire, and participants were followed up for a median of 8.5 years. During this time, 240 persons developed a cancer or died from the disease. The authors reported no association between the personality measures and cancer occurrence after adjustment for lifestyle, comorbidity, and family history of cancer. A large prospective cohort study investigated the Japanese concept of ikigai (something to live for, the joy and goal of living, or the happiness and benefit of being alive), as well as decisiveness, ease of anger arousal, and perceived stress. In a cohort of 29,098 Japanese women aged 40–79 years, a total of 209 cases of breast cancer were identified from either mortality records or diagnostic information. After a mean follow-up of 12.8 years, none of the traits/factors were associated with breast cancer incidence.30 In a cohort of 9,705 women in Nijmegen, the Netherlands, the authors investigated the association between 10 personality traits as measured with the Self-Assessment Questionnaire-Nijmegen (SAQ- N), a questionnaire developed by this group.31 The participation rate was 34%, which somewhat limits the conclusions. The medical risk factors included in the analysis were family history of breast cancer, parity, age at birth of first child, estrogen use, age at menarche, and body mass index. The authors reported no association between any of the personality traits and the risk for breast cancer. Pooling data from a randomized controlled trial nested in the Whitehall study, with 42 years of follow-up and 219 cancer deaths, neither neuroticism nor extroversion were associated with cancer- specific mortality.32 Analyses were adjusted for health status, health behaviors, and socioeconomic factors. Recently, an individual-participant meta-analysis pooled data from six prospective cohorts of the association between personality traits in the Five Factor Model (extraversion, neuroticism, agreeableness, conscientiousness, and openness to experience). The study investigated both the incidence and mortality as outcomes in 42,843 cancer-free men and women at baseline. A total of 2,156 cancer cases were diagnosed during a mean follow-up of 5.4 years and 421 cancer deaths identified in a subcohort with available cause-specific mortality information. None of the personality traits were associated with the incidence for cancer overall or for six specific cancers (lung, colon, prostate, breast, skin, and leukemia/lymphoma).33 Further, for 21,835 participants with cause-specific mortality information and 421 cancer deaths, none of the traits were associated with survival.33 In summary, these studies do not point to an association between personality traits and the risk for cancer, in particular breast cancer, as well as cancer survival. This statement is based on well- designed prospective studies that covered fairly large populations, 39 40 SECTION I Factors in Cancer Risk and Prevention clear definitions of the “exposure,” an acceptable length of follow- up, and adjustment for some factors that might be confounders. The outcome was based on either information on morbidity or close follow-up of mortality to exclude misclassification. Furthermore, the hypothesis was established independently of the cohort formation, and none of the data sources were biased by interviewers or information to cohort members. The use of administrative sources almost completely excludes selection bias. Socioeconomic Position When discussing the effects of psychosocial factors on cancer, socioeconomic position should also be mentioned. While this is not a factor that per se is thought to cause cancer directly, it is a psychosocial factor that may have substantial indirect effects (e.g., through health-and care-seeking behaviors). In a nationwide register-based study among 3.22 million Danish residents that used individual-level data on socioeconomic position, rather than data collected at the local/area level, individuals with lower education were found to be at significantly increased risk of a range of cancers, particularly those related to lifestyle factors, including tobacco.34 Conversely, individuals with higher education were at significantly increased risk of malignant melanoma, breast, and prostate cancers. However, the same study found that, generally, cancer patients with lower education had lower survival than cancer patients with higher education. Remarkably, these results stem from a country with free universal health care, highlighting that the inequality observed is not based on unequal access to care. A follow- up study revealed that the gap in survival between patients in the lowest compared with the highest income brackets was stable or had increased for most cancers,35 illustrating that the problem remains despite substantial advances in treatment methods. Mechanisms A number of investigations have been conducted of the possible pathways between mind factors and the risk for cancer. These now form a complete research area, named psychoneuroimmunology, which is an important component, with human studies, of a new field that bridges biology, epidemiology, and cancer risk research. It has been hypothesized that psychosocial factors act like stress through the hypothalamic-pituitary-adrenal axis in a complex feedback system that adversely affects overall immune function. It has been further hypothesized that the immune system is involved in eliminating mutated cells, and it is possible that reduced immunity could lead to more rapid development of cancer. Furthermore, psychosocial factors may also act as a promoter of faulty DNA repair and an inhibitor of apoptosis and DNA repair. These systems and changes in function are suggested to be precursors of certain types of cancer, such as hormonal and hematological-lymphatic cancers. Despite the plausibility of these mechanisms, little or no epidemiological evidence has arisen during the past 50 years of research on psychosocial risk factors in cancer. The association between diseases characterized by immune deficiency and breast cancer is weak, although several studies have demonstrated a reverse causation.36,37 In a large Scandinavian case–control study, however, a personal or family history of certain autoimmune conditions was strongly associated with an increased risk for Hodgkin’s lymphoma. The association between both personal and family histories of sarcoidosis and a statistically significantly increased risk for Hodgkin’s lymphoma suggests a shared susceptibility for these conditions.38 This points to the possibility that mechanisms linking psychosocial factors with cancer differ, depending on the cancer site. Alternatively, the mechanism may be driven more by changes in behavior. It is well known that people under severe stress, suffering from depression, or expressing certain personality traits differ from others with regard to health behavior. This is true for smoking, alcohol consumption, diet, and physical activity. Some studies have confirmed this pattern, finding higher risks for cancers associated with these behaviors.18,39,40 Thus, our understanding of psychosocial risk factors might have a larger public health impact if we focused on changes in the lifestyles of people exposed to stress or major life events who exhibit certain personality traits or experience depression or depressive mood. Conclusion During the past 15 years, more than 15 reviews of psychosocial risk factors for cancer have been published. The conclusions reached go in two directions: one group of reviewers find small or putative associations, while the other group find no or only a small etiological fraction reserved for psychosocial risk factors because of methodological limitations in the studies reviewed. Any conclusion must take into account the quality of the studies, whether pro or con. From a methodological viewpoint, it is incorrect to use a case–control design, as such studies have serious problems that are exacerbated in this area of research, such as recall bias. This design can be used only if information on exposure is obtained from administrative sources, in which information is collected for purposes that have nothing to do with the hypothesis of the study, as illustrated by many of the studies cited earlier. In terms of prognosis, the evidence is less clear. We have some evidence that personality traits do not contribute to increased risk of dying from cancer, while depression seems to have some effects on mortality (much like in the general population). For stress, too few studies have yet been conducted for any firm conclusions, but the importance of stress for health behaviors in particular might support the notion that stress during the course of cancer will affect cancer outcomes. Disentangling the effects of prognosis on stress and other psychosocial factors will likely prove a substantial challenge to answering this question. In conclusion, psychosocial factors such as stress, depression, and personality traits do not appear to play a major role in cancer causation. All the studies published so far have limitations, making it difficult to reach a definitive decision about causality. However, the majority of well-designed studies taking into account methodological issues of relevance for the field do not confirm that the mind causes cancer. Future Directions Among the requirements for future research are prospective data, clearer definitions of exposure, and repeated measures during CHAPTER 5 Psychosocial Factors follow-up; we also need longer follow-up to identify more cases and to investigate psychosocial factors with the same rigorous methods used in other areas of research. It has also been noted that this field of research would be more comprehensive if not just one but several psychosocial factors were included.16 We must include information on other well-defined risk factors for the disease under study to find out how they confound or interact with each other in relation to the association investigated. This is being recognized in more and more studies. The socioeconomic aspects of the exposure under study, comorbidity, and health behavior are new aspects of our understanding of the association between mind and cancer risk. We must consider carefully whether health behavior is an intermediate rather than a confounding factor, as this will have profound implications for our understanding of the possible association between psychological factors and the risk for cancer. REFERENCES 1. Hill AB. The environment and disease: Association or causation? Proc Royal Soc Med. 1965;58(5):295– 300. doi:10.1177/ 003591576505800503 2. Greenland S, Pearl J, Robins JM. Causal diagrams for epidemiologic research. Epidemiology. 1999;10(1):37–48. 3. IARC—International Agency for Research on Cancer. https:// www.iarc.fr/. Accessed December 19, 2019. 4. Lillberg K, Verkasalo PK, Kaprio J, Teppo L, Helenius H, Koskenvuo M. Stress of daily activities and risk of breast cancer: A prospective cohort study in Finland. Int J Cancer. 2001;91(6):888–893. 5. Lillberg K, Verkasalo PK, Kaprio J, Teppo L, Helenius H, Koskenvuo M. Stressful life events and risk of breast cancer in 10,808 women: A cohort study. Am J Epidemiol. 2003;157(5):415– 423. doi:10.1093/aje/kwg002 6. Kroenke CH, Hankinson SE, Schernhammer ES, Colditz GA, Kawachi I, Holmes MD. Caregiving stress, endogenous sex steroid hormone levels, and breast cancer incidence. Am J Epidemiol. 2004;159(11):1019–1027. doi:10.1093/aje/kwh148 7. Schernhammer ES, Hankinson SE, Rosner B, et al. Job stress and breast cancer risk: The Nurses’ Health Study. Am J Epidemiol. 2004;160(11):1079–1086. doi:10.1093/aje/kwh327 8. Nielsen NR, Stahlberg C, Strandberg- Larsen K, et al. Are work- related stressors associated with diagnosis of more advanced stages of incident breast cancers? Cancer Causes Control. 2008;19(3):297–303. doi:10.1007/s10552-007-9092-7 9. Michael YL, Carlson NE, Chlebowski RT, et al. Influence of stressors on breast cancer incidence in the Women’s Health Initiative. Health Psychology. 2009;28(2):137–146. doi:10.1037/a0012982 10. Surtees PG, Wainwright NWJ, Luben RN, Khaw K-T, Bingham SA. No evidence that social stress is associated with breast cancer incidence. Breast Cancer Res Treat. 2010;120(1):169–174. doi:10.1007/ s10549-009-0454-6 11. Nielsen H. [1943-79 mortality among members of the Danish resistance movement deported to German concentration camps]. Ugeskr Laeg. 1983;145(5):345–350. 12. Olsen MH, Nielsen H, Dalton SO, Johansen C. Cancer incidence and mortality among members of the Danish resistance movement deported to German concentration camps: 65-Year follow- up. Int J Cancer. 2015;136(10):2476–2480. doi:10.1002/ijc.29288 13. Chida Y, Hamer M, Wardle J, Steptoe A. Do stress-related psychosocial factors contribute to cancer incidence and survival? Nat Clin Practice Oncol. 2008;5(8):466–475. doi:10.1038/ncponc1134 14. Levav I, Kohn R, Iscovich J, Abramson JH, Wei Yann Tsai, Vigdorovich D. Cancer incidence and survival following bereavement. Am J Public Health. 2000;90(10):1601–1607. doi:10.2105/ AJPH.90.10.1601 15. Dalton SO, Boesen EH, Ross L, Schapiro IR, Johansen C. Mind and cancer: Do psychological factors cause cancer? Eur J Cancer. 2002;38(10):1313–1323. doi:10.1016/S0959-8049(02)00099-0 16. Garssen B. Psychological factors and cancer development: Evidence after 30 years of research. Clin Psychology Rev. 2004;24(3):315–338. doi:10.1016/j.cpr.2004.01.002 17. Coyne JC, Ranchor AV, Palmer SC. Meta-analysis of stress-related factors in cancer. Nat Rev Clin Oncol. 2010;7(5):1–2. doi:10.1038/ ncponc1134-c1 18. Dalton SO, Mellemkjær L, Olsen JH, Mortensen PB, Johansen C. Depression and cancer risk: A register-based study of patients hospitalized with affective disorders, Denmark, 1969–1993. Am J Epidemiol. 2002;155(12):1088–1095. doi:10.1093/aje/155.12.1088 19. Nyklíček I, Louwman WJ, Nierop PWMV, Wijnands CJ, Coebergh J-WW, Pop VJ. Depression and the lower risk for breast cancer development in middle-aged women: A prospective study. Psychological Med. 2003;33(6):1111–1117. doi:10.1017/ S0033291703007499 20. Aro AR, Koning HJD, Schreck M, Henriksson M, Anttila A, Pukkala E. Psychological risk factors of incidence of breast cancer: A prospective cohort study in Finland. Psychological Med. 2005;35(10):1515–1521. doi:10.1017/S0033291705005313 21. Kroenke CH, Bennett GG, Fuchs C, et al. Depressive symptoms and prospective incidence of colorectal cancer in women. Am J Epidemiol. 2005;162(9):839–848. doi:10.1093/aje/kwi302 22. Goldacre MJ, Wotton CJ, Yeates D, Seagroatt V, Flint J. Cancer in people with depression or anxiety: Record- linkage study. Soc Psychiat Epidemiol. 2007;42(9):683– 689. doi:10.1007/ s00127-007-0211-2 23. Liang J-A, Sun L-M, Muo C-H, Sung F-C, Chang S-N, Kao C- H. The analysis of depression and subsequent cancer risk in Taiwan. Cancer Epidemiol Biomarkers Prev. 2011;20(3):473–475. doi:10.1158/1055-9965.EPI-10-1280 24. Chen Y-H, Lin H-C. Increased risk of cancer subsequent to severe depression— A nationwide population- based study. J Affective Disord. 2011;131(1–3):200–206. doi:10.1016/j.jad.2010.12.006 25. Suppli NP, Johansen C, Kessing LV, et al. Survival after early-stage breast cancer of women previously treated for depression: A nationwide Danish cohort study. J Clin Oncol. 2017;35(3):334–342. doi:10.1200/JCO.2016.68.8358 26. Pinquart M, Duberstein PR. Depression and cancer mortality: A meta-analysis. Psychological Med. 2010;40(11):1797–1810. doi:10.1017/S0033291709992285 27. Lillberg K, Verkasalo PK, Kaprio J, Teppo L, Helenius H, Koskenvuo M. A prospective study of life satisfaction, neuroticism and breast cancer risk (Finland). Cancer Causes Control. 2002;13(2):191–198. doi:10.1023/A:1014306231709 28. Nakaya N, Bidstrup PE, Saito-Nakaya K, et al. Personality traits and cancer risk and survival based on Finnish and Swedish registry data. Am J Epidemiol. 2010;172(4):377–385. doi:10.1093/aje/ kwq046 29. Stürmer T, Hasselbach P, Amelang M. Personality, lifestyle, and risk of cardiovascular disease and cancer: Follow-up of population based cohort. BMJ. 2006;332(7554):1359. doi:10.1136/ bmj.38833.479560.80 30. Sawada T, Nishiyama T, Kikuchi N, et al. The influence of personality and perceived stress on the development of breast 41 42 SECTION I Factors in Cancer Risk and Prevention 31. 32. 33. 34. 35. cancer: 20-Year follow-up of 29,098 Japanese women. Sci Rep. 2016;6(1):1–7. doi:10.1038/srep32559 Bleiker EMA, Hendriks JHCL, Otten JDM, Verbeek ALM, van der Ploeg HM. Personality factors and breast cancer risk: A 13-year follow-up. J Natl Cancer Inst. 2008;100(3):213–218. doi:10.1093/ jnci/djm280 Batty GD, Russ TC, Stamatakis E, Kivimäki M. Psychological distress in relation to site specific cancer mortality: Pooling of unpublished data from 16 prospective cohort studies. BMJ. January 2017:j108. doi:10.1136/bmj.j108 Jokela M, Batty GD, Hintsa T, Elovainio M, Hakulinen C, Kivimäki M. Is personality associated with cancer incidence and mortality? An individual-participant meta-analysis of 2156 incident cancer cases among 42 843 men and women. Br J Cancer. 2014;110(7):1820–1824. doi:10.1038/bjc.2014.58 Dalton SO, Schüz J, Engholm G, et al. Social inequality in incidence of and survival from cancer in a population-based study in Denmark, 1994–2003: Summary of findings. Eur J Cancer. 2008;44(14):2074–2085. doi:10.1016/j.ejca.2008.06.018 Dalton SO, Olsen MH, Johansen C, Olsen JH, Andersen KK. Socioeconomic inequality in cancer survival—changes over time. 36. 37. 38. 39. 40. A population-based study, Denmark, 1987–2013. Acta Oncol. 2019;58(5):737–744. doi:10.1080/0284186X.2019.1566772 Stewart T, Tsai S-C, Grayson H, Henderson R, Opelz G. Incidence of de-novo breast cancer in women chronically immunosuppressed after organ transplantation. Lancet. 1995;346(8978):796– 798. doi:10.1016/S0140-6736(95)91618-0 Frisch M, Biggar RJ, Engels EA, Goedert JJ, Group for the A- CMRS. Association of cancer with AIDS-related immunosuppression in adults. JAMA. 2001;285(13):1736–1745. doi:10.1001/ jama.285.13.1736 Landgren O, Engels EA, Pfeiffer RM, et al. Autoimmunity and susceptibility to Hodgkin lymphoma: A population-based case– control study in Scandinavia. J Natl Cancer Inst. 2006;98(18):1321– 1330. doi:10.1093/jnci/djj361 Schapiro IR, Nielsen LF, Jørgensen T, Boesen EH, Johansen C. Psychic vulnerability and the associated risk for cancer. Cancer. 2002;94(12):3299–3306. doi:10.1002/cncr.10601 Schapiro IR, Ross-Petersen L, Sælan H, Garde K, Olsen JH, Johansen C. Extroversion and neuroticism and the associated risk of cancer: A Danish cohort study. Am J Epidemiol. 2001;153(8):757–763. doi:10.1093/aje/153.8.757 6 Viral Cancers and Behavior Susan T. Vadaparampil, Lindsay N. Fuzzell, Shannon M. Christy, Monica L. Kasting, Julie Rathwell, and Anna E. Coghill Introduction The International Agency for Research on Cancer (IARC) currently classifies seven different viruses as class I carcinogens, defined as agents with convincing evidence of carcinogenicity in humans. This includes six viruses that directly cause cancer (Figure 6.1):1 human papillomavirus (HPV), hepatitis B virus (HBV), hepatitis C virus (HCV), Epstein-Barr virus (EBV), human herpes virus 8 (HHV8), and human T-cell lymphotropic virus type 1 (HTLV-1), and one virus that indirectly contributes to cancer by impairing the immune system’s ability to fight infections: human immunodeficiency virus (HIV). Notably, approximately one in every six cancers that occur globally is attributable to pathogens, and three of the top four contributors to this infectious cancer burden are viruses, namely HPV, HBV, and HCV. These three agents alone play a role in the development of more than 1.2 million new cancers each year and are responsible for nearly 50% of the infectious cancer burden in the United States (U.S.). We focus on these three viruses, along with HIV, to illustrate the intersection between virus-associated cancers and the field of psycho-oncology. They are highlighted in this chapter not only because of their disproportionate cancer burden but also because lifestyle and health-seeking behaviors are related to both the acquisition VS Human Papillomavirus VT Hepatitis B ST Hepatitis C Epstein-Barr virus 10 00 00 20 00 00 0 10 00 0 HTLV-1 30 00 00 40 00 00 50 00 00 60 00 00 V: vaccination S: screening T: treatment HHV8 Annual case count (Plummer, Lancet 2016) Figure 6.1. Global burden of cancers attributable to viruses and available cancer prevention or treatment options. Figure 6.1 created based on data presented in Plummer M, de Martel C, Vignat J, Ferlay J, Bray F, Franceschi S. Global burden of cancers attributable to infections in 2012: A synthetic analysis. Lancet Global Health. 2016;4(9):e609–e616. of these viruses and the prevention of their associated cancers. For example, HPV is sexually transmitted, and persistent infection status is strongly linked to an individual’s lifetime number of sexual partners. Likewise, one route of transmission for HBV and HCV is the exchange of bodily fluids (e.g., blood), such that risk of infection is greatly increased among injection drug users. In addition to behaviors that can impact rates of viral acquisition, health-seeking practices must also be considered since effective prevention strategies exist for cancers attributable to each of these agents, including (1) primary prevention through vaccination against HBV and HPV, (2) secondary prevention through screening for evidence of HCV in blood or presence of high-risk HPV and related premalignant lesions at the site of infection, and (3) tertiary prevention through appropriate use of anti-viral medication to control hepatitis. Although the correlation between certain high-risk behaviors (e.g., injection drug use) and a lower likelihood of regular health-seeking behavior may complicate cancer prevention efforts, it may also offer opportunities for targeted interventions to impact behavior with the goal of preventing these virus-associated cancers. Each of the virus-attributable cancers listed in Figure 6.1 occurs more frequently in HIV-infected patients because immunosuppression interferes with normal host control of infections. Relevant to this chapter, the high-risk lifestyle behaviors that increase the risk of acquiring HPV, HBV, or HCV, including a high number of lifetime sexual partners and injection drug use, also markedly increase the risk of HIV infection. This overlap in risk factors further contributes to the high rates of hepatitis-related and HPV-associated cancers in the U.S. HIV population, including elevated rates of liver and anal cancers. Human Papillomavirus Epidemiology HPV is the most common sexually transmitted infection both globally and in the U.S. Most sexually active individuals (80%) will be infected with at least one HPV strain—also referred to as HPV type—during their lifetime.2 More than 40 HPV types are transmitted through intimate contact (i.e., vaginal, anal, or oral sex or 44 SECTION I Factors in Cancer Risk and Prevention skin-to-skin genital contact).2 Nearly 80 million individuals in the U.S. have a current HPV infection, with approximately 14 million individuals newly infected each year.2 Natural History Most HPV infections are asymptomatic. HPV types are classified as either low risk or high risk based on their oncogenic potential. Low- risk HPV types can cause anogenital warts. Two low-risk HPV types can also cause recurrent respiratory papillomatosis. Thirteen high- risk HPV types have been identified as carcinogenic to humans; HPV 16 and HPV 18 cause the majority of HPV-related cancers. In the U.S. approximately 34,800 HPV-attributed cancers are diagnosed annually.2 Although most HPV infections clear on their own within two years, persistent infections from oncogenic HPV types over years (and often decades) can progress into oropharyngeal (affecting the throat, tonsils, and/or base of the tongue), cervical, anal, penile, vaginal, and vulvar cancers.2 Indeed, nearly all (99%) cervical cancers, more than 90% of anal, and the majority of oropharyngeal (~70%), vaginal (75%), vulvar (~70%), and penile cancers (>60%) are caused by HPV.3 Prevention The majority of the U.S. HPV-related cancer cases each year (92%) are caused by one of the viruses currently included in the HPV vaccine and could be prevented if males and females 9–26 years old received the HPV vaccine.4 HPV vaccination has been routinely recommended for U.S. females since 2006 and males since 2011. The 9-valent vaccine, the only HPV vaccine currently used in the U.S., prevents infection from nine HPV types (6, 11, 16, 18, 31, 33, 45, 52, and 58), seven of which are oncogenic.4 In the U.S., the 9-valent vaccine is approved for administration between the ages of 9 and 45; two vaccine doses are recommended for individuals aged 9–14, and three for individuals aged 15 and older or who are immunocompromised.2 The HPV vaccine yields the strongest immune response when delivered in early adolescence and prior to sexual experience. The Advisory Committee on Immunization Practices (ACIP) recommends that males and females through age 26 years receive “catch-up” vaccination if they did not receive the vaccine during adolescence as they may not have been previously exposed to all of the HPV types for which the vaccine offers protection.5 The ACIP recommends that individuals between ages 27 and 45 not previously vaccinated against HPV discuss with their health care provider whether they may benefit from vaccination.5 As of 2018, an estimated 68.1% of U.S. adolescents aged 13–17 had initiated the vaccine series and 51.1% were up-to-date with the vaccine series (either two or three doses completed, depending on the age at which the series was started).6 These rates fall well below the Healthy People 2020 goal of 80% vaccination uptake. Provider recommendation remains the strongest predictor of HPV vaccine uptake.6 However, providers often fail to utilize strong, timely, and consistent recommendations. Systems-wide intervention strategies such as the inclusion of electronic medical prompts to remind providers to offer the HPV vaccine to patients, as well as automated reminders to parents/patients to promote next dose receipt, are strategies used successfully in prior interventions.7 Furthermore, studies have demonstrated low levels of HPV-related knowledge in multiple U.S. subgroup populations, including those diagnosed with HPV-related cancers.8–10 Finally, notable disparities in HPV vaccine uptake exist based on geographic location and insurance status.6 Thus, multilevel interventions addressing provider, patient, and systems levels will be needed to improve HPV vaccine series completion rates.7 Sexual activity is associated with HPV infection;11 thus, behavioral interventions to support sexual health may reduce some risk factors associated with HPV infection. In addition, current smokers are at increased risk for higher HPV viral load, cervical precancers, active oral HPV infections, and cervical and oropharyngeal cancer incidence, as are individuals exposed to secondhand smoke (including during childhood). Thus, in addition to vaccination and reducing risk of sexual transmission, reducing tobacco exposure and treating current tobacco use is another important means of preventing the development of the two most common HPV-related cancers. Additional information about tobacco cessation treatment can be found in Section I, Chapter 1, and additional information about HPV vaccination can be found in Section II, Chapter 8. Screening and Early Detection Currently, cervical cancer screening is the only recommended screening for HPV-related cancers. Regardless of HPV vaccination status, cervical cancer screening is recommended beginning at age 21 and includes Pap testing alone or in combination with HPV- DNA testing. In-home HPV self-sampling tests may address logistic (e.g., time, transportation, geographic, cost) and psychosocial (e.g., modesty, discomfort) barriers to in-clinic cervical cancer screening. Appropriate follow-up and treatment following an abnormal Pap or HPV test are critical to cervical cancer prevention or early detection. See Section II, Chapter 8 for additional information about cervical cancer screening and Section XI, Chapter 78 for information about screening for HPV-related cancers within sexual and gender minority populations. Treatment There is currently no treatment to directly combat HPV infection. However, within two years, 90% of HPV infections resolve on their own.2 For those that result in cellular changes, such as cervical dysplasia, or anogenital warts, there are several treatments (e.g., loop electrosurgical excision procedure, topical medications, surgery) to directly combat abnormal cytology discovered by screening. Viral Hepatitis Viral hepatitis includes hepatitis A, B, C, D, and E.12 Although distinct viruses, each impacts the liver. More than 1 million deaths worldwide are caused by infections from viral hepatitis annually. Chronic infections with HBV and HCV lead to prolonged inflammation and cell damage in the liver that increase risk for hepatocellular carcinoma (HCC), a cancer with increasing mortality rates in the U.S. Hepatitis B Virus Epidemiology HBV is a DNA virus that infects an estimated 21,000 people in the U.S. each year.13 Approximately 3–5% of HBV-infected adults and up to 95% of HBV-infected infants go on to develop a chronic infection,13 resulting in 850,000 individuals in the U.S. and 257 million CHAPTER 6 Viral Cancers and Behavior individuals globally living with HBV. Two out of three individuals chronically infected with HBV are unaware of their infection status. Natural History Infection with HBV is generally asymptomatic. Most infections in adults are self-limited and elicit durable immune responses. Acute infections have an incubation period of 4–10 weeks, after which the infection typically resolves with little liver damage. HBV is able to survive for at least seven days outside of the body, and can be transmitted in multiple ways, including sexually, through contact with infected blood (e.g., sharing needles, unsanitary tattooing/ piercing, needle-stick injury, use of contaminated razors), and from mother to child at birth. Although many individuals are asymptomatic, some may experience nausea, vomiting, jaundice, fever, fatigue, loss of appetite, abdominal pain, joint pain, dark urine, and clay-colored stool. Of the less than 5% of adults that develop a chronic infection, 15–25% of those progress to cirrhosis or HCC. The time between HBV infection and HCC development varies but is generally between 10 and 40 years. HBV reactivation or HBV-related disease progression is related to other hepatotoxic factors including alcohol consumption, HCV or HIV coinfection, and immunosuppression. Unfortunately, since 2014, increasing rates of acute HBV infection have been observed; in the U.S. these rates are attributed to increased rates of injection drug use associated with the opioid epidemic. An estimated 14,000 deaths each year in the U.S. are due to chronic HBV infection.13 Worldwide deaths from HBV and subsequent cirrhosis and HCC in 2015 totaled 887,000.14 Prevention and Screening Vaccination remains the best method for preventing HBV infection and the effective, three-dose vaccination schedule for HBV is recommended by the U.S. Centers for Disease Control and Prevention (CDC) for all children beginning at birth.13 In 2018, 92.1% of adolescents between the ages of 13 and 17 in the U.S. had received all three doses.6 Other important prevention methods include use of universal precautions in health care settings, screening of donated blood, screening pregnant women for HBV infection (as children born to infected mothers are at increased risk), consistent condom use, and educating infected individuals on how to prevent HBV transmission to others. HBV screening in the clinic is conducted through a blood test to detect HBV antibodies. Treatment There is currently no effective medication to help resolve acute HBV infection, and treatment involves only supportive care. For those chronically infected, the majority require long-term antiviral treatment to reduce the risk of liver disease and prevent transmission to others. The preferred course of treatment varies by patient; the American Association for the Study of Liver Diseases (AASLD) recommends antiviral therapy for immune-active chronic HBV infection to reduce the risk of liver-related complications.15 However, the AASLD recommends against antiviral therapies for patients with immune-tolerant chronic HBV infection. Although side effects of antiviral therapy vary by medication and dose, they include headaches, fatigue, fever, depression, and, rarely, lactic acidosis and nephropathy.15 Patients who decline antiviral therapies are required to complete regular bloodwork to monitor liver function and disease progression. HBV can reactivate spontaneously and immunosuppressed individuals (e.g., cancer patients receiving chemotherapy or other immunosuppressive therapies, individuals receiving organ and/or bone marrow transplantations, and those with HIV and/or HCV) are at increased risk for HBV infection reactivation. Hepatitis C Virus Epidemiology HCV infects approximately 1.75 million people globally, with 41,000 new infections in the U.S. each year. Unlike HBV, up to 85% of HCV infections become chronic. Approximately 2.4 million people are currently chronically infected in the U.S., 50% of whom are unaware of their infection status. Recent data show that 75% of people infected were born between the years 1945 and 1965 (baby boomers). Chronic infection with HCV results in an increased risk of cirrhosis and HCC. Chronic HCV infection has also been associated with non-Hodgkin lymphoma; however, those cases are rarer manifestations of HCV infection and will not be discussed in this chapter. Natural History of Infection HCV is primarily transmitted through contact with infected blood, although there is a small risk of acquiring an infection through sexual contact. While acute HCV infections are typically asymptomatic, some experience symptoms such as fever, abdominal pain, and jaundice. An infection is considered chronic if a person has detectable HCV RNA at least six months after an acute infection. Chronic infections can persist for approximately 20 years without causing any clinical symptoms, with serious health sequelae developing after approximately 30 years. Among the chronically infected, 10–20% develop cirrhosis, which confers a 1–5% annual risk of developing HCC. Overall, HCV prevalence is higher in males compared to females, and in people born between 1945 and 1965 as compared to other birth cohorts in the U.S. Within the baby boomer cohort, non-Hispanic black males have the highest prevalence. Factors associated with more rapid HCV-related disease progression include older age, male sex, obesity, coinfection with HIV and/or HBV, and alcohol use. Targeted testing and treatment are necessary to reduce the increasing rates of HCV-related morbidity and mortality. Prevention and Screening Prior to 2012, HCV testing was recommended for groups in the U.S. considered high risk, such as persons living with HIV, those who had ever injected illegal drugs, or individuals that had been incarcerated. Other people at risk are those who received clotting factor concentrates produced before 1987 or a blood transfusion or organ transplant prior to 1992 (i.e., before blood products were regularly screened for the virus), healthcare workers exposed to infected blood (e.g., through a needle stick), and children born to HCV-infected mothers. In the U.S. in 2012, testing recommendations were augmented to also include one-time testing for all people born from 1945 to 1965, as baby boomers account for 75% of prevalent HCV cases. Unfortunately, testing rates are low, and a significant proportion of the baby boomer birth cohort remains unaware of their HCV infection status, making it likely that the HCC burden in the U.S. will continue to rise. 45 46 SECTION I Factors in Cancer Risk and Prevention To achieve the World Health Organization goal of eliminating viral hepatitis,12 there needs to be a significant focus on testing and treatment of high-risk populations as recommended by medical and public health organizations. Such strategies include treating persons who inject drugs, as well as implementing harm reduction interventions (e.g., needle and syringe exchange) to halt the spread of the virus. Persons who inject drugs (PWID) are the group most likely to acquire new HCV infections, and this is an ever-growing population, due in large part to the U.S. opioid epidemic. Up to 50% of PWID have been exposed to HCV. This results in a bimodal age distribution of HCV cases, with one peak among people in their mid-20s (PWID) and a second peak among aging adults (baby boomer birth cohort). Other populations with higher-than-average rates of HCV prevalence are incarcerated and unsheltered homeless persons. Despite available screening tests and treatment options, barriers include cost and routine access to health care; this is particularly challenging among patients at highest risk of infection (e.g., homeless, incarcerated, PWID). High-risk persons may not disclose risk behaviors for fear of stigma, and patients who have not engaged in high-risk behavior for decades may not identify themselves as being at risk. Patients often rely on providers to recommend appropriate tests, but providers may not be familiar with the screening guidelines. Finally, HCV testing is a two-step process beginning with an antibody screening, followed by an HCV RNA diagnostic test. Not all patients who receive a positive antibody test return for confirmatory RNA testing. Depending on the population, 50–60% of HCV antibody–positive individuals return for the confirmatory testing. This problem could be ameliorated if health care systems implemented an automatic RNA reflex test if someone screens positive for HCV antibody, but this has not been adopted in all practice settings. Treatment Unlike HBV, there is no vaccine to prevent HCV infection. However, with the development of direct-acting antivirals (DAAs) in recent years, 97–99% of HCV infections can now be cured. Cure is defined as sustained virologic response (SVR). DAAs have fewer side effects than prior interferon-based treatment regimens and can result in SVR with only 8–12 weeks of treatment. However, SVR does not confer protective immunity; thus, there is a risk for reinfection. Reinfection rates are higher among PWID and those with HIV infection. One of the primary barriers to treatment among all populations is the cost of the medications, and policy-level solutions are needed to enable all HCV-infected persons to be treated. Those who have chronic HCV may need support for adherence to treatment regimens and ongoing surveillance. Patients should be tested 12 weeks after completing therapy to determine whether they have achieved SVR. SVR is associated with reduced liver-related morbidity and mortality. Patients with advanced fibrosis require additional surveillance, with screening every six months, as they are at a persistently higher risk for HCC. HIV Epidemiology Approximately 1.1 million individuals in the U.S. are infected with HIV. Unfortunately, this includes a notable percentage (~14%) who are currently undiagnosed. Approximately 38,000 new HIV infections still occur each year in the U.S., even in the era when effective HIV therapy is widely available. These infections are not equitably distributed in the population, with more than 50% of new infections occurring in African Americans and two-thirds in men who have sex with men (MSM). Access to HIV therapy has been remarkably effective at increasing the life expectancy of the U.S. HIV population, with mortality rates falling to less than 5 per 105 patients in the most recent CDC assessment. HIV-associated deaths have dropped sharply since 1995.16 Current variation in HIV mortality patterns is likely a reflection of health care access, with the proportion of U.S. HIV deaths in the South increasing from less than 30% of the national total in the early stages of the HIV epidemic to 53% more recently, in sharp contrast to decreases in mortality rates in the West and Northeast regions of the U.S.17 Natural History HIV does not survive long outside of the body and requires human T-cells to replicate. The virus also can only be transmitted when certain bodily fluids (e.g., blood, breast milk, fluids associated with sexual intercourse) interact with mucous membranes. However, the infection does not spontaneously clear once acquired, persisting for the life of the host in CD4+ T-cells. The most common modes of transmission in the U.S. include receptive anal intercourse and injection drug use, although the likelihood of HIV transmission through these routes is still highly dependent on an individual’s viral load. The risk of HIV transmission is high during the acute phase (i.e., within 2–4 weeks of infection), which is often accompanied by spikes in viral load and flu-like symptoms. Of note, the U.S. HIV epidemic continues to accumulate new diagnoses because those who are infected often are not aware of their HIV status. One French study estimated that enrolling 75% of yet untreated people living with HIV (PLWH) into highly active antiretroviral therapy (HAART) to control viral load would decrease new diagnoses 10- fold.18 After the acute phase, patients persist (even untreated) for an average of 10 years before signs of severe immunosuppression indicative of acquired immunodeficiency syndrome (AIDS) manifest, including opportunistic infections and AIDS-associated malignancies. However, if consistently treated with modern antiretroviral therapy, patients are far more likely to survive without manifestations of AIDS for decades. Infection with HIV increases the risk of cancer. HIV-infected individuals develop AIDS-defining cancers (ADCs) such as Kaposi sarcoma (KS) and non-Hodgkin lymphoma (NHL) at substantially higher rates than HIV-uninfected individuals. Risk is also elevated for several non-AIDS-defining cancers (NADCs) including Hodgkin lymphoma (HL), anal cancer, and lung cancer. The link between HIV and cancer is largely attributable to the effects of immunosuppression. Since antiretroviral therapy to combat HIV and restore patient immunity became widely available in the U.S. (1996), not only has there been a marked decrease in AIDS-related mortality, but also rates of AIDS-associated cancers have decreased sharply. However, even in the HAART era, immunosuppression remains an important risk factor for both ADCs and NADCs. In the most recent (1996–2012) assessment from the HIV/AIDS Cancer Match Study, a national linkage of state HIV and cancer registries, PLWH remained at a higher risk of a range of cancers caused by infections when compared to the general U.S. population.19 This included approximately CHAPTER 6 Viral Cancers and Behavior 3-fold higher rates of liver cancer (associated with HBV and HCV) and up to 19-fold higher rates of anal cancer (associated with HPV). Although the authors of the study acknowledge that risks for these cancers have declined over time in PLWH, likely due to expansion of effective HIV therapy, they also note that the persistence of elevated cancer risk warrants additional efforts at cancer prevention and screening in this high-risk population.19 Prevention The risk of HIV infection is strongly linked to several behavior and/ or lifestyle factors. The most effective way to eliminate risk of HIV is to practice safe sexual behaviors. For example, correct and consistent condom use can result in a 20-fold decrease in HIV infection and is 90–95% effective for preventing transmission of the virus. HIV transmission can also be reduced through avoidance of drug use that involves needle sharing. Cities that have implemented syringe service or needle exchange programs have decreased incidence of HIV annually by 11%. In an occupational context, accidental needle sticks or exposure to blood or other bodily fluids is associated with a low risk of HIV transmission of 0.09–0.30% on average. With proper preventive health care procedures (e.g., personal protective equipment, administrative and work practice controls) and treatment after possible exposure (postexposure prophylaxis [PeP] or HAART), this risk can be effectively managed. Transmission from mother to baby can also occur, but this can be prevented by ensuring the mother’s awareness of her HIV status, having a cesarean rather than vaginal birth, avoidance of breastfeeding, and HAART for the mother and child. For individuals at higher risk of HIV exposure, often in cases where sexual partners are known or suspected to be HIV positive or in PWID, a few treatments can be utilized for prevention. Pre- exposure prophylaxis (PrEP) involves daily medication to prevent HIV in those at high risk of infection. Consistent, daily PrEP use reduces the risk of HIV transmission through sexual contact by 99% and reduces the risk of transmission from injection drug use by 74%. Similarly, PeP can also be used preventively when an individual believes they have recently been exposed to HIV. PeP includes a daily, 28-day course of HAART, begun within 72 hours of potential exposure to HIV. PeP use after occupational exposure to HIV has been associated with an 81% reduction in HIV infection. Effectiveness of PeP after nonoccupational exposure varies by study and is dependent upon factors such as timing of PeP initiation, medication adherence, and continued exposure to HIV through ongoing risk behaviors. For those already infected with HIV, HAART can lower the viral load to undetectable levels and is regarded as a method of preventing HIV transmission to others. Early Detection and Screening Awareness of HIV status can be increased through screening in clinical and nonclinical settings. HIV screening can be performed in a lab via blood test, with rapid testing at home, or in a clinical or nonclinical site via finger prick or oral fluid. Awareness of status can reduce risky HIV sexual transmission behaviors by approximately 68%. The CDC recommends voluntary HIV screening for all adults and adolescents (ages 13–64) as a normal part of medical care.20 Guidelines specific to MSM suggest that those who are sexually active undergo annual screening.21 Other high-risk groups for whom annual screening is recommended include sexual partners of those who are diagnosed with HIV, PWID and their sexual partners, and those who exchange sex for money or drugs. Opt-out testing should be conducted for all pregnant women as part of their routine prenatal care, and repeat screening should be conducted for pregnant women in certain areas. Screening in nonclinical settings may be helpful for those who do not routinely participate in medical care. Common nonclinical settings include mobile testing units, churches, parks, community centers, or syringe service programs. Testing in community settings has successfully increased uptake of screening, is cost effective, and is viewed positively by clients and providers.22 Couples interested in “testing together” may also take advantage of screening in nonclinical or community settings. Treatment Antiretroviral therapy for HIV involves a combination of medications to slow the progression of the virus. HAART should be started immediately upon HIV diagnosis and taken correctly and consistently (daily). Although HAART does not cure HIV, it can reduce the viral load of HIV in the body to undetectable amounts. HAART helps to preserve immune function and can keep patients healthy for many years. However, if HAART treatment is not effectively maintained, viral suppression may not be achieved and progression to AIDS becomes more likely, increasing the likelihood of opportunistic infections and HIV-associated cancers. Medication adherence is a major concern for many PLWH. Barriers include patient beliefs, psychosocial and mental health issues, cognitive barriers and burden in understanding and adhering to complicated treatment regimens, structural barriers (e.g., transportation, insurance, childcare, housing), side effects, and comorbid conditions. In addition to consistent HAART use, HIV treatment includes regular viral load monitoring, regular follow-up care with a provider, and monitoring of potential coinfections. Quality of life while living with HIV is relatively high with correct and consistent HAART use and continued follow-up care. An estimated 22% of patients with a newly diagnosed HIV infection do not receive HIV-specific medical care within at least a month of their diagnosis. This, as well as incomplete retention in medical care for patients who do initiate therapy, contributes to the fact that only 62% of HIV-infected patients achieve viral suppression, which is crucial to reducing the spread of the infection and maintaining adequate levels of immunocompetence in people living with HIV. Implications for Psycho-Oncology Virus-associated cancers may have different considerations with respect to prevention, early detection, treatment, and survivorship compared to other malignancies. For most viruses described in this chapter, transmission is largely attributable to high-risk sexual or drug use–related behaviors. The populations at greatest risk for infection (and subsequent health issues, including cancer) are often from marginalized populations including racial and ethnic minority groups, sexual and gender minority groups, and rural populations. Additionally, there may be stigma associated with reporting risk behaviors to alert providers about possible exposure to a virus that is associated with cancer risk. 47 48 SECTION I Factors in Cancer Risk and Prevention Diagnosis with HPV, HBV, HCV, or HIV may invoke many emotions, often due to feared stigma or social consequence. For example, receipt of a positive HPV-DNA test and/or an abnormal Pap test result can result in distress, anxiety, and fear,23,24 as well as stigma, shame, and self-blame.25 Women with normal Pap test results who tested HPV positive were more anxious than those who tested HPV negative.26 Distress levels may be highest at or near the time of initial diagnosis, and may decrease over time.24,27 Among individuals diagnosed with an HPV-related cancer, the concerns expressed are generally focused on the cancer itself rather than HPV as a cause of the cancer. Because chronic HBV infection requires life-long follow-up, and there is not an available curative treatment, there are significant implications for psychosocial outcomes. Even in the absence of cirrhosis or HCC, patients with chronic HBV infection report various negative psychosocial symptoms related to their condition including feelings of HBV-related isolation, sadness, anger, higher fear and anxiety, and lower vitality.28 While there are few studies on HBV- related stigma, particularly in the U.S., research shows HBV patients frequently report internalized and social stigma.29 Specifically, a systematic review from 2018 found that up to 20% of patients reported believing they were denied health care due to their diagnosis and 30% indicated they faced workplace discrimination due to their HBV diagnosis.29 However, more research is needed to assess the effects of stigma on diverse populations of patients with HBV. Similar to HBV, patients with HCV may be blamed for acquiring the disease due to a history of intravenous drug use, leading to social isolation.30 Patients with HCV can experience decreased muscle strength (asthenia), moderate to severe fatigue, cognitive deficits, depression, and anxiety.30,31 Achieving HCV cure has been shown to improve patients’ quality of life, although the extent of improvement that can be expected is not well established.31 The improvements may depend on the length of time a patient was infected, as well as any underlying psychological issues that the patient had before acquiring the infection. Those living with HIV may feel isolation and depression32 and face perceived or actual discrimination and stigma.33 Isolation and loneliness are common because of the need to disclose HIV status to obtain treatment and social support, and disclosure may come with consequences like judgment, rejection, or discrimination.33 Depression in PLWH negatively impacts treatment outcomes and quality of life, but psychological and antidepressant interventions show promise for addressing these issues.32 Because of increased awareness of certain cancers associated with HIV (e.g., anal cancer in MSM), screening may be more regularly accessed; there may be psychological consequences that accompany more awareness and screening though. For instance, those with HIV who were screened for anal cancer report more anal cancer worry, worse self-rated anal health, and less optimism about the future.34,35 Thus, behavioral interventions to reduce risk or promote prevention and early detection behaviors must consider the unique characteristics, preferences, and values of the target populations. This process may include using formative research methods to develop new, or adapt existing, behavioral interventions. For those already affected with a virus-associated cancer, psychosocial providers and researchers must consider the balance between positive implications for treatment based on infection status (e.g., HPV-positive oropharyngeal cancers have higher disease-free survival), with possible feelings of stigma associated with having a viral cancer diagnosis. Chapters 17 and 18 (Section IV) provide extensive information on techniques to screen for distress, anxiety, and depression. In addition, further information about the psychological needs of gynecological cancer patients and head and neck cancer patients can be found in Section V, Chapters 27 and 29, respectively. Future Directions The field of psycho-oncology is in a unique position to contribute to the prevention of virus-associated cancers. While there are challenges associated with virus-associated cancers, the opportunities for prevention of future cancers are promising. Unlike other tumors for which there is no single direct causal factor for the majority of cases, preventing acquisition of the virus can prevent a majority of these virus-associated cancers. Given the importance of provider and systems/policy-level factors in reducing virus-associated cancers, future behavioral interventions at the patient/client level should also consider addressing issues at the provider, health care system, and/or policy level. For example, health care provider recommendation is a critical factor in promoting uptake of a variety of screening and testing behaviors. However, these recommendations must be made using evidence-based strategies. For health care systems, many prevention and early detection strategies are recommended routinely for a large age group (e.g., HBV vaccination for all infants, routine HPV vaccination for all adolescents ages 11–12, universal screening of baby boomers for HCV infection). As such, it will be important to consider normalizing these recommendations by integrating them into electronic health record reminders, standing orders, or other system-wide approaches to increase uptake. Finally, at the policy level, it will be critical to support local, state, or national policies to facilitate access to screening and treatment (both medical and psychosocial) for those at risk for or affected by virus-associated cancers. REFERENCES 1. Plummer M, de Martel C, Vignat J, Ferlay J, Bray F, Franceschi S. Global burden of cancers attributable to infections in 2012: a synthetic analysis. Lancet Global Health. 2016;4(9):e609–e616. 2. Centers for Disease Control and Prevention. Human Papillomavirus (HPV). 2019; https://www.cdc.gov/hpv/index.html. 3. National Cancer Institute. HPV and Cancer. 2019; https://www. cancer.gov/ a bout- c ancer/ c auses- prevention/ r isk/ i nfectious- agents/hpv-and-cancer. 4. Senkomago V, Henley SJ, Thomas CC, Mix JM, Markowitz LE, Saraiya M. Human papillomavirus–attributable cancers—United States, 2012–2016. Morbidity and Mortality Weekly Report. 2019;68(33):724. 5. Meites E, Szilagyi PG, Chesson HW, Unger ER, Romero JR, Markowitz LE. Human papillomavirus vaccination for adults: updated recommendations of the Advisory Committee on Immunization Practices. MMWR Morbidity and Mortality Weekly Report. 2019;68:698–702. doi:http://dx.doi.org/10.15585/mmwr. mm6832a3external icon 6. Walker TY, Elam-Evans LD, Yankey D, et al. National, regional, state, and selected local area vaccination coverage among adolescents aged 13–17 years—United States, 2018. Morbidity and Mortality Weekly Report. 2019;68(33):718. CHAPTER 6 Viral Cancers and Behavior 7. Smulian EA, Mitchell KR, Stokley S. Interventions to increase HPV vaccination coverage: a systematic review. Human Vaccines & Immunotherapeutics. 2016;12(6):1566–1588. 8. Pruitt SL, Parker PA, Peterson SK, Le T, Follen M, Basen-Engquist K. Knowledge of cervical dysplasia and human papillomavirus among women seen in a colposcopy clinic. Gynecologic Oncology. 2005;99(3):S236–S244. 9. Milbury K, Rosenthal DI, El-Naggar A, Badr H. An exploratory study of the informational and psychosocial needs of patients with human papillomavirus- associated oropharyngeal cancer. Oral Oncology. 2013;49(11):1067–1071. 10. Shelal Z, Cho D, Urbauer DL, et al. Knowledge matters and empowers: HPV vaccine advocacy among HPV-related cancer survivors. Supportive Care in Cancer. 2019:1–7. 11. de Sanjosé S, Brotons M, Pavón MA. The natural history of human papillomavirus infection. Best Practice and Research Clinical Obstetrics & Gynaecology. 2018;47:2–13. 12. World Health Organization. World Hepatitis Day 2019: Invest in Eliminating Hepatitis. 2019; https://www.who.int/campaigns/ world-hepatitis-day/2019. Accessed Sept 12, 2019. 13. Centers for Disease Control and Prevention. Viral Hepatitis. In: Division of Viral Hepatitis NCfHA, Viral Hepatitis, STD, and TB Prevention, ed. Atlanta, GA. 2020; https://www.cdc.gov/hepatitis/index.htm. 14. World Health Organization. Hepatitis B. 2019; https://www.who. int/news-room/fact-sheets/detail/hepatitis-b. 15. Terrault NA, Lok ASF, McMahon BJ, et al. Update on prevention, diagnosis, and treatment of chronic hepatitis B: AASLD 2018 hepatitis B guidance. Hepatology. 2018;67(4):1560–1599. 16. Centers for Disease Control and Prevention. HIV mortality 2017. https://www.cdc.gov/hiv/pdf/library/slidesets/cdc-hiv-surveillance- mortality-2017.pdf 17. Centers for Disease Control and Prevention. Slide Sets | Resource Library | HIV/AIDS | CDC. 2019; https://www.cdc.gov/hiv/library/slidesets/index.html. 18. Jeulin H, Jeanmaire E, Murray JM, et al. Treatment as prevention enrolling at least 75% of individuals on ART will be needed to significantly reduce HIV prevalence in a HIV cohort. Journal of Clinical Virology. 2019;120:27–32. 19. Hernández- Ramírez RU, Shiels MS, Dubrow R, Engels EA. Cancer risk in HIV-infected people in the USA from 1996 to 2012: a population- based, registry- linkage study. Lancet HIV. 2017;4(11):e495–e504. 20. Branson BM, Handsfield HH, Lampe MA, et al. Revised recommendations for hiv testing of adults, adolescents, and pregnant women in health-care settings. Morbidity and Mortality Weekly Report: Recommendations and Reports. 2006;55(14):1–17. 21. DiNenno EA, Prejean K, Irwin K, et al. Recommendations for HIV screening of gay, bisexual, and other men who have sex with 22. 23. 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. 34. 35. men—United States, 2017 | MMWR. Morbidity and Mortality Weekly Report. 2017;66:830–832. Thornton A, Delpech V, Kall M, Nardone A. HIV testing in community settings in resource-rich countries: a systematic review of the evidence—Thornton—2012—HIV Medicine—Wiley Online Library. HIV Medicine. 2012;13:416–426. Maggino T, Casadei D, Panontin E, et al. Impact of an HPV diagnosis on the quality of life in young women. Gynecologic Oncology. 2007;107(1):S175–S179. McBride E, Marlow LA, Forster AS, et al. Anxiety and distress following receipt of results from routine HPV primary testing in cervical screening: the Psychological Impact of Primary Screening (PIPS) study. International Journal of Cancer. 2019;146(8):2113–2121. Perrin KM, Daley EM, Naoom SF, et al. Women’s reactions to HPV diagnosis: insights from in-depth interviews. Women & Health. 2006;43(2):93–110. McCaffery K, Waller J, Forrest S, Cadman L, Szarewski A, Wardle J. Testing positive for human papillomavirus in routine cervical screening: examination of psychosocial impact. BJOG: An International Journal of Obstetrics & Gynaecology. 2004;111(12):1437–1443. Hsu YY, Wang WM, Fetzer SJ, Cheng YM, Hsu KF. Longitudinal psychosocial adjustment of women to human papillomavirus infection. Journal of Advanced Nursing. 2018;74(11):2523–2532. Spiegel BMR, Bolus R, Han S, et al. Development and validation of a disease-targeted quality of life instrument in chronic hepatitis B: the hepatitis B quality of life instrument, version 1.0. Hepatology. 2007;46(1):113–121. Smith-Palmer J, Pollock R, Bonroy K, Sbarigia U, Valentine W, Cerri K. The nature and impact of stigma in patients with chronic hepatitis B: a systematic literature review. Journal of Hepatology. 2018;68:S185. Marinho RT, Barreira DP. Hepatitis C. Stigma and cure. World Journal of Gastroenterology. 2013;19(40):6703–6709. Barreira DP, Marinho RT, Bicho M, Fialho R, Ouakinin SRS. Psychosocial and neurocognitive factors associated with hepatitis C—implications for future health and wellbeing. Frontiers in Psychology. 2018;9:2666. Nanni MG, Caruso R, Mitchell AJ, Meggiolaro E, Grassi L. Depression in HIV infected patients: a review. Current Psychiatry Reports. 2015;17(1):530. Bravo P, Edwards A, Rollnick S, Elwyn G. Tough decisions faced by people living with HIV: a literature review of psychosocial problems. Aids Review. 2010;12(2):76–88. Landstra JM, Ciarrochi J, Deane FP, Botes LP, Hillman RJ. The psychological impact of anal cancer screening on HIV-infected men. Psycho-Oncology. 2013;22(3):614–620. Landstra JM, Deane FP, Ciarrochi J. Psychological consequences of cancer screening in HIV. Current Opinion in Oncology. 2013;25(5):526–531. 49 SECTION II Screening for Cancer in Normal and At-Risk Populations Wendy W. T. Lam (Section Editor) 7 Colorectal Cancer Screening 53 Caitlin C. Murphy and Sally W. Vernon 8 Cervical Cancer Screening and HPV Vaccination: Multilevel Challenges to Cervical Cancer Prevention 61 Richard Fielding, Samara Perez, Zeev Rosberger, Ovidiu Tatar, and Linda D. L. Wang 9 Breast Cancer Screening 68 Gabriel M. Leung, Irene O. L. Wong, Ava Kwong, and Joseph T. Wu 10 Prostate Cancer Screening 74 Michael A. Diefenbach, Daniel Nethala, Michael Schwartz, and Simon J. Hall 11 Lung Cancer Screening 78 Lisa Carter-Harris and Jamie Ostroff 12 Skin Cancer Screening 87 Jennifer L. Hay and Stephanie N. Christian 7 Colorectal Cancer Screening Caitlin C. Murphy and Sally W. Vernon The overall goal of colorectal cancer (CRC) screening is to reduce morbidity and mortality from CRC. CRC is the second leading cause of cancer death in the United States, with estimates of 145,600 new cases and 51,020 deaths in 2019.1 Incidence and mortality rates have declined dramatically in the U.S. since the early 1990s, particularly among older adults. For example, incidence in the 65–69 age group decreased from 210.3 per 100,000 in 1992 to 108.5 per 100,000 in 2016—a 50% decrease. Mortality rates decreased similarly during that period. Improvements in CRC incidence and mortality rates are often attributed to increased uptake of screening.2 Regular screening with guaiac-based fecal occult blood test (gFOBT), fecal immunochemical test (FIT), or sigmoidoscopy facilitates earlier detection of CRC and lowers mortality.2 Screening colonoscopy may also decrease CRC incidence through early detection and removal of precancerous polyps. Most professional organizations agree that CRC screening should begin at age 50 years for those at average risk. In their 2016 update to CRC screening guidelines, the U.S. Preventive Services Task Force recommended screening with colonoscopy every 10 years, annual FIT or high-sensitivity gFOBT, FIT-DNA every 1 or 3 years, sigmoidoscopy every 10 years with annual FIT, flexible sigmoidoscopy every 5 years, or computed tomographic colonography/virtual colonoscopy every 5 years for average-risk, asymptomatic adults aged 50–75 years.3 However, the American Cancer Society issued a new guideline in May 2018 recommending CRC screening begin at age 45 (vs. 50) years for average-risk adults.4 This new guideline was commissioned in response to increasing rates of CRC incidence among younger (age <50 years) adults and is based primarily on simulation modeling. As such, the recommendation to initiate screening at age 45 years is qualified, with some uncertainty about the balance of screening benefits and harms in this age group. Two national sources of prevalence data on CRC screening in the United States are the National Health Interview Survey (NHIS) and the Behavioral Risk Factor Surveillance System (BRFSS). Differences in the methods of data collection have resulted in slightly different estimates of screening prevalence; however, both sources show a gradual increase in overall test use, with some suggestion that use started to plateau after 2010. NHIS data show an increase from 42.5% in 2000 to 58.3% in 2010 for any recent CRC screening (e.g., stool blood test in last year or colonoscopy in last 10 years).5,6 Prevalence slightly increased in 2015 to 62.4%. Most of the increase in screening has been due to increased use of colonoscopy (Figure 7.1). Despite overall improvements in screening uptake, the prevalence of CRC screening in the United States is lower than breast or cervical cancer screening. Moreover, there are disparities in screening prevalence by race/ethnicity and socioeconomic status. For example, NHIS data from 2015 showed that, compared to non-Hispanic whites (63.7%), Hispanics and American Indian/ Alaska Natives had the lowest screening uptake (47.4% and 48.4%, respectively), followed by Asians (52.1%). Screening uptake was also low among adults with less than a high school education (46.7%) and without insurance (25.1%).5 These estimates fall short of the targets set by Healthy People 2020 (70% screened) and the National Colorectal Cancer Roundtable (80% screened). The purpose of this chapter is to review behavioral research on CRC screening in the United States and to identify directions for future research based on this evidence. 60 50 % Screened Introduction 40 30 20 10 0 2000 2003 2005 2010 Any recent CRC test COL past 10 years SIG past 5 years FOBT past year Figure 7.1. Recent colorectal cancer screening test use among respondents age 50 years and older. National Health Interview Survey, 2000, 2003, 2005, 2010, and 2015, National Center for Health Statistics, Centers for Disease Control and Prevention. 2015 54 SECTION II Cancer Screening in Normal and At-Risk CRC Screening Correlates and Predictors Studies of factors associated with CRC screening are used to inform the selection of target populations for intervention and the content of interventions. For example, demographic variables are useful for identifying population subgroups to target interventions. They are less amenable to change and are, therefore, not as useful for informing the content of interventions designed to motivate and enable screening test use. The major categories of factors that have been studied in relation to CRC screening behaviors include sociodemographics, healthcare access, health status, health behavior, and cognitive, psychosocial, and environmental factors. Because CRC screening is unique in that multiple test options are available, the role of test preferences and informed decision-making in CRC screening test use has also been examined. Although numerous studies have identified factors associated with CRC screening, there have been only a few attempts to review this literature.5,6 Most of this literature is based on cross-sectional study designs that can only establish associations. Too few studies have used prospective designs or tested hypotheses about causal pathways of factors influencing CRC screening by examining mediators and moderators. Sociodemographics, Healthcare Access, Health Status, and Health Behavior Factors Studies of sociodemographics, access to healthcare, health status, and health behavior have often used data from national surveys. Although CRC screening rates have increased over time, rates have remained lower among nonwhites, those with less education, those living in rural versus urban areas, and those without a regular source of healthcare or medical insurance.3 CRC screening is the only type of cancer screening that is recommended for both males and females, but few studies have examined whether correlates differ by gender.7 Such differences may inform the use of gender-specific messages or intervention strategies to increase CRC screening test use. The most common reasons for not having a CRC test include lack of awareness or no physician recommendation.7,8 Cross-sectional studies report that a physician’s recommendation for CRC screening is important for patient adherence, but prospective studies suggest that a recommendation may not be sufficient for increasing CRC screening uptake to desired levels.9 Engaging in other preventive health behaviors, such as other cancer screening tests, has been positively associated with CRC screening. Health status has been measured in multiple ways (e.g., self-reported health, comorbidity, functional status), which may explain why the patterns of association with cancer screening behaviors have varied.10 Compared with stool blood tests, the likelihood of completing an endoscopic test is greater among people with a family history of CRC, access to regular healthcare and health insurance, fewer concerns about cost, and higher perceived CRC risk.8 Cognitive and Psychosocial Factors Health behavior theories are used to identify cognitive and psychosocial variables that may influence behavior. In cross-sectional studies, consistent correlates of CRC screening include preventive health orientation, physician recommendation, knowledge of cancer risk factors, perceived benefits (pros) and barriers (cons) to screening, and self-efficacy.6 Most, but not all, studies of perceived risk and severity report a positive association with CRC screening. Fear and worry about CRC or the screening test itself have small, if any, direct effects on CRC screening behavior, possibly due to low levels of worry even among high-risk populations. Specifying the source of worry has shown that worries about cancer are positively associated with screening, whereas worries about the test may act as a barrier to screening. Hypotheses of a curvilinear effect of worry or anxiety have not been supported.11 Relatively few psychosocial constructs have been examined in prospective studies, and fewer have been examined across studies.6 Even when the same constructs were used, they were often defined and operationalized differently, and few measures of psychosocial constructs have been validated. Nevertheless, cumulative evidence based on prospective study designs and multivariable analyses suggest consistent independent associations between CRC screening and perceived benefits and barriers, self-efficacy, and intention. Preferences for specific CRC screening tests vary by screening goals, test characteristics, and patient-level variables. In one prospective study, only about half of the sample received the CRC screening test they preferred.12 Future research should confirm the predictive utility of CRC screening correlates and explore their interrelations in theory-based causal models. Some individual-level variables that warrant examination as predictors in future research include affect, social influence, trait-level personality characteristics such as dispositional optimism or conscientiousness, patient and provider CRC screening test preferences (and their potential mismatch), low perceived need or value for screening, and avoidant or self-exemption tendencies. Conceptual Models Basic causal models are formed when intermediate behavioral outcomes are examined as mediators. Variables that have been examined as intermediate outcomes of intervention efforts to increase CRC screening behavior include intention, awareness, knowledge, risk perceptions, and physician- patient discussion about CRC screening. Intention is a frequently studied intermediate outcome because it is a strong predictor of behavior. Different intervention strategies may be needed to increase CRC screening, depending on individuals’ behavioral intentions. Understanding how to effectively tailor intervention messages or strategies is informed by process evaluation; however, few studies have reported thorough evaluations of participants’ appraisal of CRC screening informational materials or intervention components. Similarly, not all CRC screening discussions lead to CRC screening adherence; therefore, we need to better understand which components of these discussions or which informed decision-making criteria are important for increasing CRC screening uptake. Very few studies have tested hypothesized causal pathways of multiple predictors of CRC screening. Health behavior models that posit only direct effects between predictors and intention and behavior may underestimate the total effects of CRC screening determinants through other mediating and moderating pathways. One study used longitudinal data to examine competing conceptual models of the role of perceived susceptibility on CRC screening intention and behavior.13 That study found that the influence of perceived susceptibility on screening behavior was independent of perceived barriers, moderated changes in perceived benefits and self-efficacy, and was mediated by social influence. Other studies that have examined mediators or moderators generally have focused on evaluating CHAPTER 7 Colorectal Cancer Screening intervention effects rather than testing conceptual models that explain health behaviors. Future studies need to examine different causal models of mediators and moderators to provide new ways to think about the underlying mechanisms relating cognitions and behaviors in order to improve the effectiveness of interventions. Multilevel models can examine the independent and interactive effects of individual-and environmental-level variables on CRC screening. Examining environmental- level variables could potentially identify screening disparities across health systems or geographic areas that could be targeted for interventions or CRC screening programs. An emerging literature has identified multiple environmental-level factors associated with CRC screening, including, among others, several indicators of area socioeconomic status, availability, density, capacity, or features of local healthcare resources, and managed care penetration.14 Future research is needed to further elucidate the mechanisms of influence between environment-level factors and CRC screening. Interventions to Promote CRC Screening Efforts to promote the uptake of CRC screening began in the 1980s and accelerated in 1997 upon publication of consensus screening guidelines. An increasing number of reviews have evaluated the effectiveness of interventions to increase CRC screening. Arguably, the most widely used recommendations for selecting interventions to promote CRC screening are those of the Community Preventive Services Task Force (see more at https://www.thecommunityguide. org/topic/cancer). The Community Guide recommendations were updated in 2016 based on an expanded literature review of studies published through November 2013. The review included 56 intervention studies, and the cumulative evidence demonstrated that multicomponent interventions increase CRC screening by a median of 15%. Multicomponent interventions include approaches within the strategies of increasing community demand (e.g., reminders), increasing community access (e.g., reducing structural barriers), or increasing provider delivery of screening (e.g., provider assessment and feedback), and these interventions may also address needs at the individual, provider, community, and system levels (Table 7.1). The largest effect on screening uptake was observed in multicomponent interventions that combined approaches from each of these three strategies, and those with five or more intervention approaches showed a larger median increase. There was limited evidence on the effect of specific combinations of intervention approaches, as well as the efficacy of interventions among population subgroups (e.g., low health literacy, low income). Recent reviews of the CRC screening intervention literature have focused on mailed outreach, patient navigation, and interventions offering a choice of screening test.7 The field has moved away from patient-level interventions to intervening at the provider or healthcare system levels, and many recent studies were conducted in large, integrated healthcare systems or federally qualified health centers. In these settings, mailed outreach and patient navigation, particularly in the context of multicomponent interventions, are associated with increased CRC screening. Offering patients a choice between a stool blood test and colonoscopy appears to increase screening initiation compared to offering only colonoscopy; some data also suggest better completion with a two-sample FIT compared to a three-sample gFOBT. We highlight briefly some of the key findings from these reviews and selected studies. Interventions Using Mailed Outreach Several studies tested interventions of mailed outreach, or distributing stool blood test kits by mail with postage-paid return envelopes to patients who are not up-to-date with CRC screening.8 Mailed outreach includes two intervention components recommended by the Community Preventive Services Task Force: (1) increasing community demand via client reminders and written education materials and (2) increasing community access by reducing structural barriers through the delivery of a mailed (often free) test kit, eliminating the need for patients to attend a clinic visit to complete screening. Some also consist of telephone or written reminders to complete screening. Importantly, many trials of mailed outreach have been conducted in low-income, uninsured, or minority populations— groups with historically low screening. Mailed outreach in these settings is associated with statistically significant improvements in screening uptake, ranging from 18% to 36% across studies.8 Even in studies not designed to test directly the effect of mailed outreach, Table 7.1. Multicomponent Interventions Include Approaches within the Strategies of Increasing Community Demand, Increasing Community Access, and Increasing Provider Delivery Intervention Strategy Intervention approach Increase Community Demand Increase Community Access Increase Provider Delivery Group education Reduce client out-of-pocket costs Provider reminders One-on-one education Reduce structural barriers Provider incentives Client reminders • Reduce administrative barriers Provider assessment and feedback Client incentives • Provide appointment scheduling assistance Mass media • Use alternative screening sites Small media • Use alternative screening hours • Provide transportation • Provide translation • Provide childcare Adapted from Community Preventive Services Task Force, Increasing Colorectal Cancer Screening: Multicomponent Interventions, Finding and Rationale Statement, August 2016 (see more at https://www.thecommunityguide.org/topic/cancer). 55 56 SECTION II Cancer Screening in Normal and At-Risk this intervention strategy appears to drive screening uptake. For example, tailored telephone counseling increased screening compared to a web-only intervention,9 largely driven by the telephone group’s participants’ ability to request a FIT kit by mail. A systematic review and meta-analysis by Jager et al.8 of seven randomized trials shows a 28% absolute increase in screening when comparing mailed outreach to usual care. Results were consistent in subgroup analyses by test type (FIT vs. gFOBT), setting (underserved/minority vs. all else), and inclusion of telephone reminders (with vs. without). Among studies included in the review were a cluster randomized trial10 at federally qualified health centers in Oregon and California (13.9% mailed outreach vs. 10.4% usual care); comparative effectiveness trial11 in a rural safety-net hospital (40.7% mailed outreach vs. 12.1% usual care); and randomized trial in a community health center network in Chicago (36.7% mailed outreach vs. 14.8% usual care). The 28% absolute increase in screening across these trials compares favorably to or exceeds interventions offering stool blood tests at the time of annual flu vaccination, patient reminders, and educational campaigns. Taken together, the Jager et al. review supports mailed outreach offering either FIT or gFOBT as an effective, evidence-based strategy for increasing CRC screening, particularly in low-income or underserved populations. Observational studies similarly support the efficacy of mailed outreach. For example, in a large, integrated healthcare system, an organized CRC screening program with mailed outreach doubled the proportion of adults up-to-date with screening (from 40% to over 80%).12 The increase in screening was associated with a substantial decrease in CRC incidence and mortality of 26% and 52%, respectively. In the Veteran’s Health Administration (VHA), the largest healthcare provider in the U.S., screening has increased dramatically over time, largely driven by increases in gFOBT13 offered through organized programs. The majority of patients in the VHA who complete CRC screening do so by gFOBT. Finally, many European studies report success with mailed outreach as part of nationwide programs implementing organized screening,14 with up to 60% of participants returning a test kit by mail. These findings suggest that organized screening programs with mailed outreach have the potential to both achieve national screening goals and reduce cancer mortality in the population. Interventions Using Patient Navigation Patient navigation is a barriers-focused intervention, whereby a trained healthcare professional (e.g., nurse, lay health worker) guides a patient through a complex healthcare system and addresses sociocultural, educational, and logistical barriers to screening. A number of CRC screening intervention trials have compared patient navigation of varying levels of intensity to usual care, and with or without mailed outreach. Tested strategies include (1) standard navigation vs. usual care, (2) standard navigation vs. enhanced navigation, and (3) navigation with mailed outreach vs. mailed outreach alone. Ritvo et al.15 conducted a pragmatic trial comparing patient navigation to usual care among more than 5,000 patients in primary care clinics. Patients randomized to patient navigation were contacted by a trained nurse navigator to discuss CRC screening by telephone; interested patients then met with the navigator, who helped them identify and arrange a screening test based on their preferences. Screening was higher in the intervention group (35%) compared to usual care (20%). Green et al.16 evaluated a stepped intervention approach in a large managed care network of primary care clinics, building from an existing system in which usual care involved promotion of preventive services, including CRC screening. Usual care was compared with interventions of increasing intensity, including (1) automatically generated letter and informational pamphlet indicating that patients were due for CRC screening (automated group), (2) telephone assistance from a medical assistant (assisted group), or (3) navigation from a registered nurse who assessed the patient’s CRC and procedural risks; provided motivational counseling to help patients define their intent, select a test, and develop an action plan; assisted with referrals, appointments, and test preparation; and tracked test completion (navigation group). Screening was highest among those randomized to navigation, and there was a stepped increase in screening as the intensity of the interventions increased: 26%, 51%, 58%, and 65% for usual care, automated, assisted, and navigation groups, respectively. Five-year follow-up of this study shows patients assigned to any intervention continued to adhere to screening recommendations.17 Other studies comparing standard navigation to enhanced or culturally tailored navigation show no difference in CRC screening uptake between the two types of navigation (pooled odds ratio [OR]: 1.04; 95% CI: 0.98–1.11).7 Enhanced navigation often included a range of activities, from more frequent contact with patients to address screening barriers to motivational interviewing to language concordance between the patient and navigator. Importantly, many patient navigation interventions also included mail outreach. For example, Myers et al.18 evaluated tailored navigation (i.e., mailed FIT kit and instructions plus a telephone call from a patient navigator) versus standard intervention (mailed FIT kit and instructions) versus usual care. There was no significant difference in screening between the two intervention groups (45% vs. 37%), raising questions about the incremental benefit of patient navigation compared to mailed outreach. However, other studies directly comparing navigation plus mailed outreach to mailed outreach alone demonstrated a small, but statistically significant, benefit of adding navigation.7,19 Indeed, a more recent trial20 compared a decision support and navigation intervention to standard of care, and despite both groups receiving a mailed FIT kit and instructions, those randomized to decision support and navigation were more likely to complete FIT (57%) compared to those randomized to a standard intervention (37%). A challenge in the patient navigation literature has been inconsistent names or labels used to describe similar intervention strategies—ranging from patient management to targeted telephone education. In addition, there are differences across navigation studies in delivery mode (e.g., telephone vs. in-person), dose (e.g., brief telephone call vs. 30-minute educational session), and frequency of contact. Navigation likely involves increasing behavioral capacity and self-efficacy by addressing patient and system barriers to accessing screening. Of the few studies that explicitly state they used theory to inform intervention development, facilitation, cues to action, and tailored messaging are the most common behavior change methods. Patient navigation interventions can be strengthened by a more systematic use of behavioral theory to guide development and implementation. CHAPTER 7 Colorectal Cancer Screening Interventions Offering Choice Several intervention studies have examined the effect of providing patients with a choice of screening test. This approach is consistent with literature on patient preferences for CRC screening, demonstrating that patients often prefer the test they feel most confident in completing. Specifically, patients less interested in getting screened are more likely to choose stool blood tests, which require less planning and preparation and are more convenient and less invasive than colonoscopy; patients exhibiting more readiness to complete screening tend to prefer colonoscopy.21 Incorporating patient preferences in interventions may increase the likelihood that patients initiate screening. For example, Inadomi et al.22 randomized patients to recommendation for screening by FOBT, colonoscopy, or their choice between the two. Participants randomized to colonoscopy were less likely to complete screening (38%) compared to those randomized to FOBT (67%) or a choice (69%). Of the latter group, 31% completed colonoscopy, suggesting that offering only colonoscopy would decrease screening participation. In the 3-year follow-up of this trial, patients offered a choice between FOBT and colonoscopy (42%) continued to have high adherence to screening, but adherence in the FOBT group (14%) fell below the choice and colonoscopy (38%) groups.23 Post hoc analyses of randomized intervention trials have generated compelling evidence supporting the importance of incorporating patient preference in intervention strategies. In a pragmatic trial of mailed outreach, 20% of patients mailed invitations to complete colonoscopy “crossed over” to complete FIT instead (vs. 3% crossover to colonoscopy among those mailed invitations for FIT).24 Similarly, in an intervention trial of patient navigation and mailed outreach, patients were more likely to complete the test they preferred, and the effect was even stronger when combined with navigation.25 Other trials of decision aids26,27 (i.e., structured tool for offering choice to patients) suggest that patients who are able to weigh the pros and cons of CRC screening tests are more likely to complete screening with a test of their choice. We still know relatively little about the importance of patient preference and screening choice in relation to other factors, such as access and navigation that may be more important to test completion. Myers et al. studied 764 African Americans and found that, in an intervention that included a navigation component, patients who expressed a preference for stool testing were much more likely to obtain a stool test than a colonoscopy (41.1% vs. 7.1%).19 In the comparison group without navigation, fewer patients who expressed a preference for stool testing completed the test (12.1% vs. 7.6%), even though a stool kit had been mailed to them. Timely Follow-Up of Abnormal Screening Tests Timely follow-up of abnormal or inconclusive test results achieves maximum benefits from cancer screening because delays in follow- up may contribute to advanced cancers. For example, among patients with a positive FIT, follow-up colonoscopy at or after 7 months is associated with an increased risk of advanced adenoma, any CRC, and advanced-stage disease.28 Yet, as few as 50% of patients with abnormal test results complete colonoscopy within 6 months, compared to the 90% in randomized trials of screening efficacy. Selby et al.29 conducted a systematic review of 23 randomized and nonrandomized studies of interventions to improve follow-up of positive fecal tests, and moderate evidence supported only patient navigation as an effective intervention. Instead, much of the literature is descriptive and focuses on patient characteristics associated with timely follow-up. Evidence concerning provider-and system- level strategies, such as identification, reporting, and resolution of abnormal findings, is scant and insufficient. For example, administrative databases and algorithms that identify the appropriate follow-up needs of individual patients, based on test results, could be linked to communication interventions of increasing intensity, starting with tailored mailed letters or automated phone calls and progressing to personal phone calls and navigation, as was done in one study.16 Implementing standard tracking and reporting procedures in clinical practice may also improve completion of diagnostic colonoscopy, and more intensive outreach may be needed to reduce disparities in follow-up among underserved patients. Longitudinal Adherence to Screening Repeat FOBT Because most studies report one- time use rather than repeat screening outcomes, we know little about interventions designed to promote regular FIT or gFOBT. On-schedule screening is particularly important for these stool-based tests because of the relatively frequent interval over which they need to be performed, and because effectiveness may be reduced when patients do not adhere to a regular schedule. Most European countries, Canada, and Australia recommend stool- based screening every 2 years, while annual screening is recommended in the U.S. and Asian countries.14 Observational studies show a wide range of repeat screening. Prevalence of repeat FIT or gFOBT in community settings varies from 25% to 90% among persons who have previously completed a negative test,30–32 and prevalence generally decreases across screening rounds. Connection to primary care, comorbidity, and self-efficacy appear to be important determinants of repeat screening.30,32–34 Routine screening is most likely to be accomplished in healthcare settings that have systems in place that can monitor patients’ status and can generate automatic reminders, as is the case, for example, in some managed care organizations.20 As healthcare systems adopt a “FIT First” approach to CRC screening,35 data concerning adherence to repeated rounds of screening and over longer time periods will become increasingly relevant. Although few studies have explicitly tested the effects of an intervention to increase repeat or longitudinal adherence to screening, some have examined patient adherence to repeat stool blood tests by extending an intervention trial over at least two rounds of screening. For example, Baker et al.36 examined adherence to repeat FOBT among patients who completed an index test during an intervention trial of mailed outreach; 89% of patients completed repeat screening within 6 months of their second outreach. Long-term follow-up of the Systems of Support to Increase Colorectal Cancer Screening (SOS) Trial shows intervention participants had over 30% more time adherent to screening compared to usual care over a 5-year period, and stool tests accounted for almost all of the difference in covered time between the two groups.17 57 58 SECTION II Cancer Screening in Normal and At-Risk Notably, a recent pragmatic trial conducted by Singal et al.37 suggests that more patients offered mailed outreach for colonoscopy (vs. FIT or gFOBT) complete screening when followed for a longer duration. This trial compared two mailed outreach strategies to usual care in a safety-net hospital over a 3-year period. Patients were randomized to receive a mailed invitation to complete a FIT with a home test kit and postage-paid return envelope; a mailed invitation to schedule a colonoscopy and, if scheduled, a bowel preparation kit and preprocedure instructions; or opportunistic, visit-based screening through usual care. Both outreach groups received telephone reminders from research staff. An important feature of this study was the primary outcome—screening process completion— defined as adherence to colonoscopy, annual testing for a normal FIT, diagnostic colonoscopy after abnormal FIT, or treatment evaluation if CRC was detected. Prior studies of mailed outreach have been limited to 1-year outcomes. During the first year of the trial, screening with FIT was nearly doubled among patients randomized to mailed outreach (58.8%) compared to usual care (29.6%). However, by the end of the 3-year trial period, screening process completion was highest among those randomized to colonoscopy outreach. Notably, screening completion was higher in both outreach groups compared to usual care (38.4% colonoscopy outreach vs. 28.0% FIT outreach vs. 10.7% usual care). FIT may have fewer barriers to one-time completion, but it requires annual screening and diagnostic evaluation of abnormal results. In contrast, colonoscopy is both a screening and diagnostic test, and a single examination can satisfy screening completion for up to 10 years. Under-and Overuse of Colonoscopy Adherence to repeat colonoscopy may be less of a concern because when no polyps are found, the next colonoscopy is not recommended for another 10 years. However, it is important to monitor repeat colonoscopy, particularly in the setting of surveillance colonoscopy administered at more frequent intervals. Deviations from guideline recommendations for follow-up and surveillance colonoscopy include both under-and overuse of colonoscopy. Screening overuse has been identified as a potential risk for overtreatment of benign disease, and overuse of colonoscopy may additionally increase risk of adverse events (e.g., bleeding), increase healthcare costs, and reduce endoscopic capacity in underscreened populations. Among Medicare patients with a negative screening colonoscopy, nearly half underwent a repeat colonoscopy in fewer than 7 years.27 Similarly, a mail survey of gastroenterologists and surgeons performing colonoscopy found that 24% of gastroenterologists and 54% of surgeons recommended repeat colonoscopy at intervals not concordant with guidelines.28 Follow-up of this survey study showed that a nonconcordant recommendation from physicians was associated with screening overuse.38 On the other hand, underuse of colonoscopy remains an ongoing concern, particularly for those at higher risk of CRC (e.g., due to personal history of adenomas). About one-quarter of patients who undergo screening colonoscopy will have adenomas that require removal and a follow-up colonoscopy in 3–5 years. Several recent studies have examined patterns of and factors associated with surveillance colonoscopy, and many of these were conducted in large, integrated healthcare systems. For example, in a multisite study of patients with advanced adenomas, receipt of surveillance colonoscopy at 3 years ranged from 18% to 60% across healthcare systems.39 A study in the VHA found that the majority of patients with advanced adenomas did not receive a follow-up colonoscopy, or received it after the recommended interval.38 Correlates or predictors of adherence to surveillance colonoscopy include perceived barriers, perceived benefits, social deprivation, and cancer worry.40 These observational studies highlight the need for system-level improvements that may facilitate appropriate delivery of repeat colonoscopy. Enhancing existing information systems to systematically track patients by CRC risk could identify patients eligible for surveillance colonoscopy when testing is due. Reminder and recall systems can be set to guideline-recommended intervals that alert both physicians and patients to schedule colonoscopy. Other changes include computer prompts or lock-outs, which prohibit physicians from ordering inappropriate procedures without override. Future Directions Substantial progress has been made in decreasing incidence and mortality from CRC, and this progress is often attributed to increasing use of CRC screening. However, the approximately 65% of adults up-to-date with screening indicates that there is still room for improvement. Moreover, there are important disparities in screening prevalence by race/ethnicity and socioeconomic status. Several systematic reviews focus specifically on interventions in racial/ethnic groups in the United States,16–18 and many of the intervention trials reviewed in this chapter (e.g., mailed outreach, patient navigation) were conducted in low-income or underserved populations. Several points that we raised in an earlier chapter on this topic remain to be addressed. Comparative Effectiveness Research No head-to-head trial has as yet compared screening strategies to determine whether or not some strategies have a greater net benefit than others. In the absence of empirical evidence, the U.S. Preventive Services Task Forces recommends that clinicians engage patients in informed decision-making about the screening strategy that the patient is mostly likely to complete and that reflects the patient’s preferences and local availability.3 An ongoing trial in the VHA will directly address this gap in evidence by comparing colonoscopy versus FIT screening strategies, with a primary outcome of CRC mortality after 10 years of follow-up. The CONFIRM trial (clinicaltrials.gov, NCT01239082) has randomized nearly 50,000 patients to complete annual FITs or to receive colonoscopy consistent with current standards of care. Three ongoing trials in Europe address similar questions. Impact of Healthcare Policy Health insurance is a consistent and strong predictor of CRC screening, and uninsured populations are far less likely to engage in screening. The Affordable Care Act (ACA) has improved insurance coverage and preventive healthcare visits, including uptake of CRC screening, in states expanding Medicaid. Indeed, prevalence of CRC screening among low-income adults increased in Medicaid expansion states, and increases in screening were even higher in states earliest to expand Medicaid.41 Other research suggests that screening has increased among Medicare beneficiaries, driven by the ACA’s mandate to eliminate cost sharing for preventive services.42 CHAPTER 7 Colorectal Cancer Screening As the healthcare landscape continues to evolve in the U.S., it will be important to monitor the impact of policy on screening. Dissemination and Implementation For CRC screening programs to be successful, they must involve more than one-time screening but a series of coordinated steps, from initial screening to diagnostic evaluation to treatment of any detected lesions.43 Patient navigation and mailed outreach have been the most extensively tested interventions and have the strongest evidence base for increasing screening. As noted by Selby et al.,29 patient navigation and clinician reminders have a similar impact on follow-up of abnormal test results, although the strength of evidence is lower by comparison. Broad implementation of either of these strategies, as well as others involving multiple components (see Table 7.1), may bring the current screening prevalence of 65% closer to the national goal of 80%. An important next step is to focus on implementing these approaches in large healthcare systems—or population health management programs—where there can be follow-up to determine effectiveness over the long term, as well as sustainability.44 Future research should move away from efficacy trials and toward a better understanding of how best to implement and scale these strategies. Dissemination and implementation science is a quickly developing field devoted to understanding factors in the adoption of evidence- based interventions and testing different strategies for improving integration of these interventions into practice. REFERENCES 1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA: A Cancer Journal for Clinicians. 2019;69(1):7–34. 2. Murphy CC, Sandler RS, Sanoff HK, Yang YC, Lund JL, Baron JA. Decrease in incidence of colorectal cancer among individuals 50 years or older after recommendations for population-based screening. Clinical Gastroenterology and Hepatology: The Official Clinical Practice Journal of the American Gastroenterological Association. 2017;15(6):903–909.e906. 3. Bibbins-Domingo K, Grossman DC, Curry SJ, et al. Screening for colorectal cancer: US Preventive Services Task Force recommendation statement. JAMA: The Journal of the American Medical Association. 2016;315(23):2564–2575. 4. Wolf AM, Fontham ET, Church TR, et al. Colorectal cancer screening for average-risk adults: 2018 guideline update from the American Cancer Society. CA: A Cancer Journal for Clinicians. 2018;68(4):250–281. 5. White A, Thompson TD, White MC, et al. Cancer screening test use—United States, 2015. MMWR Morbidity and Mortality Weekly Report. 2017;66(8):201–206. 6. de Moor JS, Cohen RA, Shapiro JA, et al. Colorectal cancer screening in the United States: trends from 2008 to 2015 and variation by health insurance coverage. Preventive Medicine. 2018;112:199–206. 7. Dougherty MK, Brenner AT, Crockett SD, et al. Evaluation of interventions intended to increase colorectal cancer screening rates in the United States: a systematic review and meta-analysis. JAMA Internal Medicine. 2018;178(12):1645–1658. 8. Jager M, Demb J, Asghar A, et al. Mailed outreach is superior to usual care alone for colorectal cancer screening in the USA: a systematic review and meta-analysis. Digestive Diseases and Sciences. 2019;64(9):2489–2496. 9. Champion VL, Christy SM, Rakowski W, et al. A randomized trial to compare a tailored web-based intervention and tailored phone counseling to usual care for increasing colorectal cancer screening. Cancer Epidemiology, Biomarkers & Prevention: A Publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology. 2018;27(12):1433–1441. 10. Coronado GD, Golovaty I, Longton G, Levy L, Jimenez R. Effectiveness of a clinic- based colorectal cancer screening promotion program for underserved Hispanics. Cancer. 2011;117(8):1745–1754. 11. Gupta S, Halm EA, Rockey DC, et al. Comparative effectiveness of fecal immunochemical test outreach, colonoscopy outreach, and usual care for boosting colorectal cancer screening among the underserved: a randomized clinical trial. JAMA Internal Medicine. 2013;173(18):1725–1732. 12. Levin TR, Corley DA, Jensen CD, et al. Effects of organized colorectal cancer screening on cancer incidence and mortality in a large community- based population. Gastroenterology. 2018;155(5):1383–1391.e1385. 13. El-Serag HB, Petersen L, Hampel H, Richardson P, Cooper G. The use of screening colonoscopy for patients cared for by the Department of Veterans Affairs. Archives of Internal Medicine. 2006;166(20):2202–2208. 14. Schreuders EH, Ruco A, Rabeneck L, et al. Colorectal cancer screening: a global overview of existing programmes. Gut. 2015;64(10):1637–1649. 15. Ritvo PG, Myers RE, Paszat LF, et al. Personal navigation increases colorectal cancer screening uptake. Cancer Epidemiology, Biomarkers & Prevention: A Publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology. 2015;24(3):506–511. 16. Green BB, Wang CY, Anderson ML, et al. An automated intervention with stepped increases in support to increase uptake of colorectal cancer screening: a randomized trial. Annals of Internal Medicine. 2013;158(5 Pt 1):301–311. 17. Green BB, Anderson ML, Cook AJ, et al. A centralized mailed program with stepped increases of support increases time in compliance with colorectal cancer screening guidelines over 5 years: a randomized trial. Cancer. 2017;123(22):4472–4480. 18. Myers RE, Bittner-Fagan H, Daskalakis C, et al. A randomized controlled trial of a tailored navigation and a standard intervention in colorectal cancer screening. Cancer Epidemiology, Biomarkers & Prevention: A Publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology. 2013;22(1):109–117. 19. Myers RE, Sifri R, Daskalakis C, et al. Increasing colon cancer screening in primary care among African Americans. Journal of the National Cancer Institute. 2014;106(12):dju344. 20. Myers RE, Stello B, Daskalakis C, et al. Decision support and navigation to increase colorectal cancer screening among Hispanic patients. Cancer Epidemiology, Biomarkers & Prevention: A Publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology. 2019;28(2):384–391. 21. Hawley ST, McQueen A, Bartholomew LK, et al. Preferences for colorectal cancer screening tests and screening test use in a large multispecialty primary care practice. Cancer. 2012;118(10):2726–2734. 22. Inadomi JM, Vijan S, Janz NK, et al. Adherence to colorectal cancer screening: a randomized clinical trial of competing strategies. Arch Intern Med. 2012;172(7):575–582. 23. Liang PS, Wheat CL, Abhat A, et al. Adherence to competing strategies for colorectal cancer screening over 3 years. American Journal of Gastroenterology. 2016;111(1):105–114. 59 60 SECTION II Cancer Screening in Normal and At-Risk 24. Murphy CC, Ahn C, Pruitt SL, et al. Screening initiation with FIT or colonoscopy: post-hoc analysis of a pragmatic, randomized trial. Preventive Medicine. 2019;118:332–335. 25. Daskalakis C, Vernon SW, Sifri R, et al. The effects of test preference, test access, and navigation on colorectal cancer screening. Cancer Epidemiology, Biomarkers & Prevention: A Publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology. 2014;23(8):1521–1528. 26. Mehta SJ, Khan T, Guerra C, et al. A randomized controlled trial of opt-in versus opt-out colorectal cancer screening outreach. American Journal of Gastroenterology. 2018;113(12):1848–1854. 27. Reuland DS, Brenner AT, Hoffman R, et al. Effect of combined patient decision aid and patient navigation vs usual care for colorectal cancer screening in a vulnerable patient population: a randomized clinical trial. JAMA Internal Medicine. 2017;177(7):967–974. 28. Corley DA, Jensen CD, Quinn VP, et al. Association between time to colonoscopy after a positive fecal test result and risk of colorectal cancer and cancer stage at diagnosis. JAMA: The Journal of the American Medical Association. 2017;317(16):1631–1641. 29. Selby K, Baumgartner C, Levin TR, et al. Interventions to improve follow-up of positive results on fecal blood tests: a systematic review. Annals of Internal Medicine. 2017;167(8):565–575. 30. Liss DT, Petit-Homme A, Feinglass J, Buchanan DR, Baker DW. Adherence to repeat fecal occult blood testing in an urban community health center network. Journal of Community Health. 2013;38(5):829–833. 31. Gellad ZF, Stechuchak KM, Fisher DA, et al. Longitudinal adherence to fecal occult blood testing impacts colorectal cancer screening quality. American Journal of Gastroenterology. 2011;106(6):1125–1134. 32. Fenton JJ, Elmore JG, Buist DS, Reid RJ, Tancredi DJ, Baldwin LM. Longitudinal adherence with fecal occult blood test screening in community practice. Annals of Family Medicine. 2010;8(5):397–401. 33. Murphy CC, Vernon SW, Haddock NM, Anderson ML, Chubak J, Green BB. Longitudinal predictors of colorectal cancer screening among participants in a randomized controlled trial. Preventive Medicine. 2014;66:123–130. 34. Duncan A, Turnbull D, Wilson C, et al. Behavioural and demographic predictors of adherence to three consecutive faecal occult blood test screening opportunities: a population study. BMC Public Health. 2014;14:238. 35. Jensen CD, Corley DA, Quinn VP, et al. Fecal immunochemical test program performance over 4 rounds of annual screening: a 36. 37. 38. 39. 40. 41. 42. 43. 44. retrospective cohort study. Annals of Internal Medicine. 2016;164(7):456–463. Baker DW, Brown T, Goldman SN, et al. Two-year follow-up of the effectiveness of a multifaceted intervention to improve adherence to annual colorectal cancer screening in community health centers. Cancer Causes & Control: CCC. 2015;26(11):1685–1690. Singal AG, Gupta S, Skinner CS, et al. Effect of colonoscopy outreach vs fecal immunochemical test outreach on colorectal cancer screening completion: a randomized clinical trial. JAMA: The Journal of the American Medical Association. 2017;318(9):806–815. Murphy CC, Sandler RS, Grubber JM, Johnson MR, Fisher DA. Underuse and overuse of colonoscopy for repeat screening and surveillance in the Veterans Health Administration. Clinical Gastroenterology and Hepatology: The Official Clinical Practice Journal of the American Gastroenterological Association. 2016;14(3):436–444.e1. Chubak J, McLerran D, Zheng Y, et al. Receipt of colonoscopy following diagnosis of advanced adenomas: an analysis within integrated healthcare delivery systems. Cancer Epidemiology, Biomarkers & Prevention: A Publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology. 2019;28(1):91–98. Murphy CC, Lewis CL, Golin CE, Sandler RS. Underuse of surveillance colonoscopy in patients at increased risk of colorectal cancer. American Journal of Gastroenterology. 2015;110(5):633–641. Fedewa SA, Yabroff KR, Smith RA, Goding Sauer A, Han X, Jemal A. Changes in breast and colorectal cancer screening after Medicaid expansion under the Affordable Care Act. American Journal of Preventive Medicine. 2019;57(1):3–12. Richman I, Asch SM, Bhattacharya J, Owens DK. Colorectal cancer screening in the era of the Affordable Care Act. Journal of General Internal Medicine. 2016;31(3):315–320. Tiro JA, Kamineni A, Levin TR, et al. The colorectal cancer screening process in community settings: a conceptual model for the population-based research optimizing screening through personalized regimens consortium. Cancer Epidemiology, Biomarkers & Prevention: A Publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology. 2014;23(7):1147–1158. Green BB. Colorectal cancer control: where have we been and where should we go next? JAMA Internal Medicine. 2018;178(12):1658–1660. 8 Cervical Cancer Screening and HPV Vaccination Multilevel Challenges to Cervical Cancer Prevention Richard Fielding, Samara Perez, Zeev Rosberger, Ovidiu Tatar, and Linda D. L. Wang Introduction Although largely preventable, cervical cancer is a disease that affects over half a million women each year. Despite a safe and effective vaccine having been available for 13 years, cervical cancer still causes over 300,000 deaths per year, 90% of which are in low-income (LICs) and lower-middle-income countries (LMICs). Despite monumental advances in prevention, screening, diagnosis, and treatment over the past decade, substantial regional and global disparities in cervical cancer outcomes remain and cervical cancer continues to be a major worldwide health challenge. Epidemiology of Cervical Cancer Cervical cancer ranks as the 10th most prevalent cancer overall and the fourth for both incidence and mortality among females1. Cervical cancer is second only to breast cancer in terms of cumulative risk from birth to age 74 in LICs and LMICs.1 Cervical cancer is often referred to as the “disease of disparity” as age-standardized rates (ASRs) for cervical cancer are highest in the LMICs of Africa and Melanesia. For example, Swaziland ranks #1 with an ASR of 75.3 and Kenya ranks #20 with an ASR of 33.8 cases per 100,000 of the population. The only non-African country in the top 20 is Bolivia, which ranks 16, with an ASR of 38.5. Countries in Southeast Asia, Eastern Europe, the Caribbean, and South America have the next highest rates.2 In contrast, rates are 7–10 times lower in high-income countries (HICs: e.g., US, Canada, New Zealand/Australia, and West Asia).1 Importantly, even in developed countries, disparities remain and disproportionately impact certain women compared to others. For example, in the US, Hispanic and Black American women historically have faced a higher incidence of cervical cancer than other ethnicities, attributable to lower screening rates, lower adherence to medical follow-up recommendations after abnormal screening results, and poorer-quality medical treatment. Most cases of cervical cancer are caused by human papilloma virus (HPV) subtypes. Of the hundreds of types, 12 have been identified as Class 1 carcinogens by the International Agency for Research on Cancer (IARC).1 Despite HPV being the most prevalent sexually transmitted disease, the vast majority of women infected do not develop cancer. Transient infections may cause low-grade cervical lesions, but 90% of HPV infections clear naturally within several months. Other environmental cofactors include (1) early onset of sexual activity and (2) higher number of sexual partners, both of which increase HPV exposure frequency and duration, thereby indirectly affecting risk;2 (3) being overweight;2 (4) smoking, which accounts for around 2% of cervical cancer cases;1,2 and (5) immunosuppression conditions, such as human immunodeficiency virus (HIV).1 Persistent HPV infection is therefore the main risk factor for cervical cancer. Public Health Burden from Cervical Cancers Rates of cervical cancer have declined globally over the past few decades, in part due to increasing socioeconomic development, as well as reduced persistence of HPV infection arising from general improvements in hygiene, reductions in parity (number of births), and declining sexually transmitted disease incidence.1 In virtually all HICs, including the US, Canada, Australia, Germany, the Netherlands, Italy, and Sweden, cervical cytology has played a crucial role in substantially bringing down deaths from cervical cancers. In the absence of effective cervical screening, the reverse trend is found, with premature mortality rates from cervical cancer increasing. In May 2018, the World Health Organization (WHO) declared cervical cancer as “one of the world’s deadliest, but the most easily 62 SECTION II Cancer Screening in Normal and At-Risk preventable forms of cancer for women.”3 The director-general of the WHO called for “coordinated action globally to eliminate cervical cancer,” with the plan to submit a resolution of a global cervical cancer elimination strategy at the World Health Assembly in May 2020.3 The implementation of both cervical cancer screening and HPV vaccination programs faces multiple challenges, particularly in the developing world. Common established barriers include high vaccine prices in countries where no funded vaccination programs exist, poor heath infrastructure, low health literacy, lack of political will, vaccine hesitancy, evolving shifts in screening technologies, and competing public health priorities in countries with limited resources. A further complicating factor is that cervical cancer is exclusively a disease that affects females, and in many countries, patriarchal values place a lower priority on girls and women and their health, limiting arguments for extending preventive care to women’s sexual health. Most recently, the growing anti-vaccine movement has emerged, which adds major challenges to cervical cancer prevention. In Japan and India, for example, unrelated deaths of recently vaccinated young girls were cited as sufficient rationale to lead to a suspension of the national HPV vaccination program in Japan and the continued blockage of HPV vaccine inclusion in the Universal Vaccination Program in India. HPV Vaccines The primary prevention strategy for cervical cancer is HPV vaccination. There are three HPV vaccines available: the bivalent vaccine Cervarix®, the quadrivalent vaccine Gardasil®, and the nonavalent vaccine Gardasil® 9. The primary target of publicly funded HPV vaccination programs is girls aged 9–14 years old with a two-dose schedule. Girls and women over 15 are recommended to receive a three-dose schedule. All three vaccines protect against types 16 and 18, which are the oncogenic high-risk types, and are responsible for about 70% of all cervical cancers. Gardasil® 9 offers protection against five additional high-risk HPV types and can prevent up to 90% of cervical cancers. The HPV vaccines have proven to be safe and highly immunogenic and to induce strong direct and indirect (cross-immunity) protection against HPV. With the development of these vaccines, there is the likelihood of almost completely eliminating cervical cancer among vaccinated women, particularly in the context of continued cervical cancer screening programs and wide population vaccine coverage. HPV Vaccine Availability and Accessibility: Successes and Challenges Over the last 13 years, the HPV vaccine has been approved in 129 countries, and over 270 million doses of the vaccine have been distributed worldwide.4 The proportion of countries that have introduced HPV vaccine by WHO regions are Africa—17%, Americas—69%, Eastern Mediterranean—5%, Europe—64%, Southeast Asia—27%, and Western Pacific—48%.5 These percentages reflect why the title of “disease of disparity” has far from faded. Worldwide, only one- quarter of girls aged 9–14 years live in a country with a national HPV program (~90 programs and 38 pilot programs), with the vast majority of national programs in upper-middle-income countries (UMICs) and HICs and most pilot programs in LICs and LMICs.5,6 The proportion of countries that have introduced HPV vaccination by World Bank categories are LICs—13%, LMICs—20%, UMICs— 42%, and HICs—82%, with 43% globally. Unfortunately, those most at risk of cervical cancer are the least likely to have access to the vaccine, with only 13 LICs having HPV programs.5 The most extensive estimates report a worldwide HPV vaccine coverage of 6.1% (95% confidence interval [CI]: 4.9–7.1) among females aged 10–20 years, with a 33.6% coverage (95% CI: 25.9–41.7) in more developed regions and a mere 2.7% coverage (95% CI: 1.8– 3.6) in less developed regions.5 High, successful coverage (i.e., reaching targets of >80–90%) among the target population has been achieved in 18 countries, for example, Australia, Bhutan, Iceland, Malaysia, Mexico, Rwanda, and Seychelles, typically through school-based programs, complemented and supported by health facility outreach.5 The GAVI Alliance, a public-private partnership working to improve vaccine coverage worldwide, has launched demonstration programs in Ghana, Kenya, Laos, Madagascar, Malawi, Niger, Sierra Leone, and Tanzania providing HPV vaccines to girls 9–13 years old through schools and community health programs since 2013. Through these demonstration programs, an estimated 180,000 or more girls have been protected from HPV. GAVI expects to have more than 30 million girls vaccinated in over 40 countries by 2020.6 However, many LICs do not offer regular health services for girls aged 9–13 and it is difficult to ensure that these programs will reach those at highest risk. Civil unrest in many countries compounds these difficulties. The two countries with the largest populations globally, India and China, currently lack population-based programs for HPV vaccination. India According to data compiled by the HPV Information Center, a collaboration of the Catalan Institute of Oncology and the IARC, India has one of the world’s highest burdens of HPV-related cancer. More than 122,000 Indian women are diagnosed with cervical cancer and around 67,000 die from this disease each year, which is greater than the country’s maternal mortality burden of 45,000.7 Estimates have been made of the cost-effectiveness of various cervical cancer control strategies, namely vaccination prior to 12 years of age and cancer screening among women above 30 years of age living in India. If a bivalent vaccine coverage of 70% was assumed, there would be a 44% mean reduction in the lifetime risk of cervical cancer.7 If HPV testing twice in one’s lifetime was added in the analysis, mean risk reduction increased to 63%.7 The combination of HPV vaccine and screening (either using visual inspection with acetic acid [VIA] or HPV DNA testing) was reported as cost-effective for India if the cost per vaccinated girl was $10 USD (653 INR). Recommendations for a combined vaccination/screening program have been made by medical bodies7 and the Indian Council of Medical Research, which advises government about inclusions into the Universal Vaccination Program. Unfortunately, aggressive objections from nationalist groups, some arising from an earlier failed clinical trial and accusations of associated deaths that on closer investigation proved to be due to unrelated snakebites, drowning, or suicides, have so far blocked the implementation of a national program in India. CHAPTER 8 Cervical Cancer Screening and HPV Vaccination China In July 2016, China’s Food and Health Administration approved the bivalent vaccine, a decade after the first HPV vaccine’s licensing in the US. This delay resulted from no priority-setting system in a lengthy trial registration and almost 8-year-long clinical trials done in the domestic population. The approval lag in mainland China for HPV vaccines prompted thousands of women and girls to seek vaccination in Hong Kong, where HPV vaccines have been available since 2006. The quadrivalent and nonavalent HPV vaccines were recently approved for future use in mainland China. While there is no publicly funded vaccination program, nor even official publicity regarding HPV vaccination, potential vaccine recipients must register for HPV vaccination at their local community health centers. Waiting times for the first vaccination vary from a few months to more than one year. On September 23, 2019, China’s central government released Healthy China Action—Implementation Program for Cancer Prevention and Control (2019–2022). This document presents plans for a strengthened scientific campaign of HPV vaccination and promotion of HPV vaccination uptake among the age-appropriate population; acceleration of the approval process for domestic HPV vaccine production, thereby improving vaccine accessibility; and mechanisms to impel vaccine suppliers to set reasonable prices through price negotiation and centralized purchasing, and it suggests exploring various channels to ensure effective population coverage in poor areas of China. Given the important role of social influence in Chinese vaccination decision-making,8 the central government action plan should help to substantially increase the uptake rate of HPV vaccination and further reduce cervical cancer incidence in this enormous population. High-Income Countries In contrast to India and China and many other LICs and LMICs, HPV vaccination programs are more widely established in UMICs and HICs, yet uptake rates vary considerably— not only from country to country, but also within the same country or jurisdictions, evidencing state/provincial, regional, local, and territorial differences.4 Australia was the first country globally to introduce a nationwide HPV vaccination program for girls in 2007, followed by vaccination for boys in 2013, and by 2017 it had successfully achieved national coverage rates of 80% for females and 76% for males. The UK reports stable and consistent uptake rates of 80–90% in their 2017/2018 school-based HPV vaccination programs for girls, and young boys are now included as of the 2019 academic year. The Netherlands achieves HPV vaccination coverage among girls of ~65% and recently agreed to include boys, along with lowering vaccination age to 9 years. In the US, the HPV vaccine is funded and included nationally by the Vaccines for Children Program and by private insurance, and coverage is mandated by the Affordable Care Act. Importantly, substantial variation has been reported by state, with HPV vaccine coverage as high as 87.5% in Massachusetts and as low as 52.2% in Kentucky in 2018. In Canada, all 10 provinces and three territories have publicly funded school-based HPV vaccination programs for females and males in place. Canadian rates as high as 90% in Newfoundland and Labrador and as low as 40% in the Northwest Territories have been reported.4 From an accessibility standpoint, the HPV vaccine supply is insufficient to meet demand. There are challenges with price and affordability, including high price per vaccine dose; highly variable pricing, with some HICs paying less than the average LMIC prices; and additional delivery operation costs. From an acceptability standpoint, many countries continue to face numerous challenges related to population fears of adverse side effects4 and concerns about safety and growing reactionary attitudes toward vaccination in general. These have significantly impacted and lowered the coverage rate in numerous countries that previously had high levels of vaccine uptake—for example, Japan (1%), Denmark (54%), Columbia (14%), and Ireland (50%), to name a few. These challenges sometimes overshadow the successes of HPV vaccination.4 A recent meta-analysis9 of 65 studies from 14 HICs using pooled follow-up data collected over 9 years on 60 million people found that there have been declines in HPV 16 and 18 infections of 83% (relative risk [RR] 0.17; 95% CI: 0.11–0.25) among vaccinated girls and young women aged 13 to 19 years and 66% (RR 0.34; 95% CI: 0.23–0.49) among women aged 20–24 years, while there was a significant decrease in the prevalence of HPV 31, 33, and 45 infections in 13–19-year-old girls and women (RR 0.46; 95% CI: 0.33–0.66). Taken together, these five HPV strains account for 90% of all cervical cancer cases. Moreover, nonvaccinated populations such as older women, boys, and men have also benefited from vaccination of young girls through reduced rates of infection.9 Thus, the impact of HPV vaccination transcends beyond cervical cancer and also helps prevent other HPV-related cancers such as anal, penile, and head and neck cancers. However, vaccination alone, even if widely implemented, will not eradicate cervical cancer for several reasons. First, over 100 HPV serotypes have been identified, and while the seven most oncogenic strains are covered by existing HPV vaccines, other less oncogenic strains do remain. With the existing HPV vaccines, 70–75% of all cervical cancer cases are preventable given 80–100% population coverage, though such coverage relies on vaccination rates currently underachieved in many parts of the world and assumes a world where people do not travel easily and often. Therefore, it is necessary to retain and more effectively implement cervical cancer screening as a core element of cervical cancer prevention and control. Strong evidence has emerged that high-risk HPV testing has greater sensitivity for cervical cancer risk than does cytology in detecting cervical intraepithelial neoplasia (CIN) in primary cervical cancer screening and is less prone to misinterpretation due to human error. Cost issues, however, may limit uptake LICs and LMICs. Taken together with HPV vaccination, there is a major opportunity for improved screening to drive down long-term rates of advanced disease leading to the virtual elimination of cervical cancer. Cervical Cancer Screening The goal of cervical cancer screening is to identify those at high risk of developing the disease, in this case women having an immediate precursor lesion (CIN). The effective prevention of progression to cervical cancer depends on this early detection. Screening requires three related approaches to be successful: an effective testing protocol associated with a continuous case-finding policy, the willingness of susceptible individuals to attend screening, and affordable and effective follow-up and treatment in those who screened positive. 63 64 SECTION II Cancer Screening in Normal and At-Risk Screening Protocols in High-and Low-Resource Settings Screening may involve a multistep process involving cervical cytology, for example, Papanicolaou (“Pap”) test/smear or liquid cytology, and/or HPV testing, with colposcopy/biopsy where dysplasia is suspected. Cytology requires a sample of cells scraped from the cervix that are later manually or, increasingly, automatically (liquid cytology) scanned for abnormal cells. Generally, cytology is recommended every 3 years, cotesting (cytology + HPV testing) every 5 years, and HPV testing alone every 5 years. Pap smears alone, though, have low sensitivities of around 53–55%, compared to HPV DNA testing, which has sensitivities of ~95%, while both combined give a sensitivity of 100% with 92% specificity.10 However, this approach is both expensive and cumbersome, and unsuitable for most LICs where cervical cancer incidence remains high. Moreover, HPV testing before 25 years old is not indicated as the incidence of HPV infections is high and very rarely associated with dysplasia, and testing has unnecessary financial and psychological costs. In LICs, visual inspection of the cervix (VIC) after application of 3–5% acetic acid or Lugol’s iodine is often used and can achieve sensitivities of 73–85% with 85% specificity.10 This approach has been shown to reduce lifetime risk to women from cervical cancer by around 40%. However, performance remains dependent on the skill of the observer. To avoid this, countries are increasingly moving and progressing toward a single screening HPV test that offers greater sensitivity. There is controversy as to which approach is optimal, and cost-effectiveness is a key issue. Table 8.1 describes programs in a selection of countries, highlighting policy variations (organized vs. opportunistic) and implementation decision strategies. To help decision makers, in 2014, the WHO3 published a manual entitled the Comprehensive Cervical Cancer Control: A Guide to Essential Practice, which offers expert panel guidelines on screening and treatment protocols. As mentioned earlier, with the WHO director-general’s 2018 call to action for the elimination of cervical cancer, many individuals including seven working groups are trying to accelerate the dissemination of up- to-date cervical cancer screening recommendations based on the evolving scientific evidence. Table 8.1. Examples of Routine HPV Vaccination Policy and Primary Cervical Cancer Screening Protocols Country Routine HPV Vaccination Policy Primary Screening for Cervical Cancer a Australia Organized, school-based HPV vaccination program at age 12–13 for girls and boys. Girls and boys up to age 19 can receive HPV vaccine for free. Organized HPV-based testing program from ages 25 to 74 at 5-year intervals Canada*b Organized, school-based HPV vaccination program given at ages 9–15* Organized cytology program in 9 out 13 provinces/territories at 2-or 3-year intervals starting at age 21 or 25 until 65 or 70* UKc Organized, school-based HPV vaccination programs at ages 12–13 for girls Organized HPV-based testing program (with cytology) from ages 25 to 49 at 3-year intervals, or at 5-year intervals for women aged 50–64 years Netherlandsd Organized, school-based HPV vaccination program at ages 12–13 for girls only Organized HPV-based testing program from ages 30 to 60 at 5-year intervals USe Opportunistic HPV vaccination Opportunistic screening with cytology for ages 21–29 at 3-year intervals; opportunistic screening with cytology at 3-year intervals or HPV testing at 5-year intervals or cotesting at 5-year intervals from ages 30 to 65. Women aged 30–65 should discuss with their HCP which of the 3 recommended screening strategies work best for them. No organized HPV vaccination program available Organized HPV-based testing program at ages 30–65 at 5-year intervals India No organized HPV vaccination program. The Indian Academy of Pediatrics Committee on Immunization recommends offering HPV vaccine to all females who can afford the vaccine. No cervical cancer screening program. Screen-and-treat approach using visual inspection with acetic acid, consistent with WHO guidelines for countries that do not have cervical cancer screening programs Chinag No established HPV vaccination program Organized cytology screening program for rural women aged 35–59 years. Opportunistic screening and regular cervical screening benefits offered by urban employers for married female employees Turkeyf g *Variations in grade given dependent on province/territory. European guidelines recommend implementation of HPV-based screening starting at 30–35 years at >5-year intervals up to 60–69 years. WHO guidelines recommendation: HPV screening age 30 and older, every 3–5 years; Cytology acceptable if HPV unavailable and cytology of good quality; visual inspection after application of acetic acid as alternative in low-resource settings. 7 a Australian Government Department of Health. 2018. National Cervical Screening Program. http://www.cancerscreening.gov.au/internet/screening/publishing.nsf/Content/ cervical-screening-1 b Canadian Partnership Against Cancer. 2018. Cervical cancer screening in Canada: Environmental scan. https://www.partnershipagainstcancer.ca/topics/ cervical-cancer-screening-environmental-scan-2018/ c Cancer Research UK. 2019. Cervical cancer screening. https://www.cancerresearchuk.org/about-cancer/cervical-cancer/getting-diagnosed/screening/about d National Institute for Public Health and the Environment. 2019. Cervical cancer screening programme. https://www.rivm.nl/en/cervical-cancer-screening-programme e U.S. Preventive Task Force. 2019. Final Recommendation Statement, Cervical Cancer: Screening. https://www.uspreventiveservicestaskforce.org/Page/Document/ RecommendationStatementFinal/cervical-cancer-screening2 f Turkey Public Health Institution Cancer Control Department. 2016. Turkey Cancer Control Programme. https://www.iccp-portal.org/system/files/plans/Turkiye_Kanser_Kontrol_ Program_English.pdf g Geneva Foundation for Medical Education and Research, Obstetrics and Gynecology Guidelines. Cervical cancer screening, prevention. Accessed on December 13, 2019, from https://www.gfmer.ch/Guidelines/Female_genital_neoplasms/Cervical_cancer_screening.htm CHAPTER 8 Cervical Cancer Screening and HPV Vaccination Changes in Screening Recommendations and Programs Worldwide New recommendations in the US, Europe, and Australia incorporate HPV testing as a primary screen for cervical cancer in women aged between 30 and 65 years, either as a stand-alone test11–13 or with cytology (cotesting).13 Globally, population-based HPV test screening programs that replace Pap testing (cytology) are being implemented in Australia, the Netherlands, Sweden, and Italy, while the UK and Norway started in 2019.14 Other HICs considering such a shift include Canada, France, and Finland.14 Of the existing organized HPV test-based screening programs, there are noted variations, including age of starting (e.g., 25–35 years old) and ending screening (e.g., 60– 74 years old), triage of HPV-positive women (e.g., cytology, colposcopy, repeat HPV testing in 1 year), and longer screening intervals (e.g., 3–5 and possibly up to 10 years).14 As an increasing number of vaccinated females move into the screening target population, ensuring the most appropriate screening protocol to maximize benefits and minimize potential harms for both vaccinated and unvaccinated women becomes even more pressing. Barriers and Facilitators to HPV: Vaccination, Screening, and Testing Central to cervical cancer prevention is the recipient’s or recipient’s parent’s knowledge, beliefs, understanding, and ultimately acceptance of HPV vaccination and/or screening. While HPV vaccination is currently widely and unequivocally backed by the scientific and medical communities, sociocultural and behavioral challenges remain at the individual level,4 as well as accessibility, availability, and cost at the country level, particularly in LICs and LMICs. At the individual level, vaccination decision-making is complex and multifaceted. Currently, parental unwillingness to have their young children vaccinated has led to both lower vaccination coverage and the resurgence of several vaccine-preventable infections, notably measles and pertussis. Novel, nonmandated vaccines particularly generate concerns about safety, “newness” (though HPV vaccines have now been available for over 13 years), and side effects.4 Vaccination hesitancy and general anti-vaccine attitudes are cited as barriers to HPV vaccination for certain subpopulations. Moreover, there are differences in sociocultural characteristics: for example, some studies15 indicate Chinese groups have lower HPV vaccination coverage in the US, while elsewhere lower income, having fewer children, religiosity, and suspicion about physicians’ motivation are associated with higher belief that such vaccination was unnecessary, despite considering vaccination generally to be effective and beneficial. One particular variable that has consistently emerged from studies of Chinese parents is that anticipated affective consequences (asking parents to think about how they might feel if their daughter developed cervical cancer) and social influences (knowing other parents were vaccinating their daughters) influence parents’ plans to vaccinate their daughters. These factors may have wider relevance in other Asian communities. There is also discussion about where and who is best positioned to give the HPV vaccine. In middle-income countries and HICs, the health care provider (HCP), be they doctor, nurse, or pharmacist, can provide key information to motivate an individual to receive the HPV vaccine. Moreover, organized, government-funded programs administered at school are likely to be the most cost-effective and optimal approach, though children not attending school would be missed.7 However, as is the case in India, where it has led to prolonged deferral of program implementation, reactive patriarchal attitudes are often evoked in response, reflecting the wider issue of women’s rights. Barriers to Pap screening uptake include availability, accessibility, affordability, and the lack of women physicians or health workers. However, a self-administered cervical swab can now be used to provide cytology samples for HPV DNA testing, where this is available, removing some of the privacy issues that can discourage women from undergoing vaginal inspection. Self-sampling “represents a new advance in cancer control that is unequivocally empowering to women” and can effectively reach underscreened (and often marginalized) women in which about half of all invasive cervical cancers are diagnosed.16 Overcoming women’s lack of knowledge and anxieties surrounding cervical screening requires both wider education, particularly among older women, those of low socioeconomic status, and those from diverse ethnicities, and interventions aimed to encourage health workers to ask and prompt female clients about cervical screening. HPV and Cervical Cancer Screening Messaging Safe and effective public health programs can fail because of lack of effective communication. The development of the internet and the rise of social media make it clear that a new public health program must be introduced carefully so that the target population’s unique needs are considered in order to increase uptake. This requires the effective education and mobilization of key stakeholders, including not only the young women and girls themselves but also parents, health practitioners/providers, community leaders, religious leaders, teachers, and journalists. With respect to HPV vaccine messaging, high levels of HPV vaccine acceptance are most likely a result of the extensive educational work done in the communities prior to vaccine introduction, a key point described in the PATH HPV Vaccine Lessons Learnt Project, which is part of a comprehensive review of HPV vaccine delivery experiences across 46 low-and middle-income countries.17 Emphasis on the protection against cancer has been critical in many communities that don’t necessarily understand that HPV is the cause of the cancer itself. As such, a key and clear message is that the HPV vaccine prevents cervical cancer, is safe, will not harm future fertility, and is endorsed as safe and effective by public health and other medical professionals, the government, and the WHO. Women’s negative attitudes related to deferring the start of screening using HPV testing to 25 or 30 years and/or increasing the screening interval to 5 years or more, and women’s negative emotions and perceptions related to HPV testing (since it detects a sexually transmitted infection) represent important barriers toward implementation of HPV testing in primary screening.18 Addressing these barriers in a timely manner underscores the importance of effective communication about forthcoming HPV test-based national screening programs. Adequate stakeholder engagement in policy setting appears to be crucial. For example, in Australia, following inadequate community engagement prior to government announcing changes to screening protocols, women’s collective anxiety fueled an organized, grassroots online petition titled “Stop May 1st Changes to Pap Smears—Save Women’s Lives” signed by more than 70,000 women, which forced 65 66 SECTION II Cancer Screening in Normal and At-Risk the Australian Ministry of Health into a 6-month delay of the implementation date to December 2017. Nevertheless, the only informational response to women was a one-paragraph letter from the Ministry of Health summarizing the major points of the change with no apparent input from women. Preliminary reports indicated that, subsequently, initial uptake of HPV testing is suboptimal. Similarly, in the UK, where the new screening program started in 2019, to the best of our knowledge, little information has been disseminated thus far. In the Netherlands, women over age 40 have been expressing concerns about a proposed 10-year screening interval after a negative HPV test. In countries without a national HPV test-based cervical screening program, HCPs’ recommendation for the HPV test is critical.19 In the context of multiple updates of specialty organizations’ guidelines for HPV testing over the last decade and inconsistencies in these guidelines, a recent review has shown that HCPs’ recommendations for HPV testing are suboptimal.19 Improving consistency across guidelines and educational interventions for HCPs is needed to increase HPV test uptake. Psychological Impact of a Positive HPV Test Result after Cervical Cancer Screening Women’s reactions to testing positive for HPV reflects a complex interaction between emotions (feelings), cognitions (thoughts, beliefs, attitudes), and behaviors (actions).20 Receiving a positive HPV test result is associated with elevated state anxiety that declines if women subsequently search for HPV-related information and adopt open and communication-rich interactions with HCPs. Similarly, testing positive for HPV may elicit significant levels of nonspecific or sexual distress around, for example, impact of the result on close relationships or fertility concerns, which persists for at least 6 months. Being diagnosed with high-risk HPV types 16 and 18 can trigger feelings of fear, compounded by the necessity to attend frequent follow-ups after testing positive, that fuel women’s thoughts about the probability of developing cervical cancer at some point.20 Not surprisingly, considering that HPV is a sexually transmitted infection, a positive HPV test result can evoke feelings of shame and disgust in affected women that persist for at least 6 months after diagnosis, along with worries about being sexually rejected by prospective partners. Long- time monogamous relationships can fuel a false sense of security related to sexually transmitted diseases and explain women’s initial emotional reaction of surprise when testing HPV positive and confusion that can be explained by their lack of knowledge related to the asymptomatic nature of HPV infection.20 In some societies a positive test result may even precipitate incorrect accusations of primarily female infidelity and, in extreme cases, marital breakup. Further, testing positive for HPV can be associated with sadness or feelings of depression. Facilitators of coping favorably with a positive HPV test result include acceptance that HPV infections are inevitable when having an active sexual life, receiving reassuring information from HCPs about the natural course of HPV infection and treatment options for precancerous lesions, and their resilience in dealing with health issues. Taken together, HCPs should be aware that HPV-positive test results can have a significant psychological impact on women and should be prepared to address their needs in an open and reassuring way and offer and encourage psychological counseling with knowledgeable professionals. Future Directions and Challenges In contrast with almost all other cancers, we have the tools to eliminate and eradicate this disease. This in itself is likely why cervical cancer is one of the WHO’s top 10 global health priorities for 2020– 2030.3 The WHO’s proposed target is that 90% of girls receive the HPV vaccine by 15 years old, that 70% of women are screened with an HPV test at 35 and 45 years of age, and that 90% of women identified with cervical cancer receive treatment and care;3 these goals are intensive but are within our reach. Studies are underway to confirm if using a single dose of the HPV vaccine offers adequate protection against cervical cancer. A one-dose HPV vaccination schedule would decrease vaccination costs and increase vaccination coverage globally. This possibility, combined with a focus on school settings for optimal delivery as well as government funding, augurs well for high levels of HPV vaccine coverage globally. Moreover, both enhancing demand and simplifying the protocol and implementation of organized HPV testing are key to reaching the WHO goals. However, the policy landscape internationally is vulnerable to cosmic shifts. For example, at the time of completing this chapter, the WHO Strategic Advisory Group of Experts (SAGE) on immunization, in response to a worldwide unexpected shortage of vaccine production capacity to meet demand, has recommended that “all countries should temporarily pause implementation of gender neutral, older age group, and multi-age cohort HPV vaccination strategies until supply allows equitable access by all countries,” until the supply shortfall is met by 2024. While the immediate justification is that available vaccines should be delivered to women most at risk in LMICs, for example, there may be unintended consequences in countries that have already implemented gender-neutral programs. Stopping or delaying gender-neutral programs could provoke a backlash among parents of eligible boys and could inadvertently provide more fodder for the anti-vaxxer movement by suggesting that males are not as “at risk” as has been previously advocated. A better recommendation might have been to invest in screening programs more rapidly in LMICs in the interim, programs that will be critical, in addition to vaccines to ensure effective prevention in the future.21 Differences in infrastructure, cultural barriers, technology, and ability to feasibly address prevention and treatment strategies are key reasons that the epithet “disease of disparity” cannot yet be shed. By using clear policy protocols coupled with appropriate use of vaccines and effective, well-established screening strategies, morbidity from cervical cancers can be drastically reduced and hopefully eliminated in the foreseeable future. REFERENCES 1. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: Can J Clinicians. 2018; 68: 394–424. 2. Continuous Update Project. 2018. https://www.wcrf.org/ dietandcancer/cancer-trends/cervical-cancer-statistics. 3. World Health Organization. Global Strategy Towards the Elimination of Cervical Cancer as a Public Health Problem. [Internet]. 2019. https://www.who.int/docs/default-source/documents/cervical-cancer-elimination-draft-strategy.pdf. Accessed June 3, 2019. CHAPTER 8 Cervical Cancer Screening and HPV Vaccination 4. Perez S, Zimet GD, Tatar O, Stupiansky NW, Fisher WA, Rosberger Z. Human papillomavirus vaccines: successes and future challenges. Drugs. 2018; 78: 1385–1396. 5. Bruni L, Diaz M, Barrionuevo-Rosas L, et al. Global estimates of human papillomavirus vaccination coverage by region and income level: a pooled analysis. Lancet Glob Health. 2016; 4: e453–463. 6. Gallagher KE, LaMontagne DS, Watson-Jones D. Status of HPV vaccine introduction and barriers to country uptake. Vaccine. 2018; 36: 4761–4767. 7. Kaur P, Mehrotra R, Rengaswamy S, et al. Human papillomavirus vaccine for cancer cervix prevention: rationale & recommendations for implementation in India. Ind J Med Res. 2017; 146: 153. 8. Wang LD-L, Lam WWT, Fielding R. Determinants of human papillomavirus vaccination uptake among adolescent girls: a theory- based longitudinal study among Hong Kong Chinese parents. Prevent Med. 2017; 102: 24–30. 9. Drolet M, Bénard É, Pérez N, et al. Population-level impact and herd effects following the introduction of human papillomavirus vaccination programmes: updated systematic review and meta- analysis. Lancet. 2019; 394: 497–509. 10. Brown AJ, Trimble CL. New technologies for cervical cancer screening. Best Pract Res Clin Obstet Gynaecol. 2012;26(2):233–242. 11. von Karsa L, Arbyn M, De Vuyst H, et al. European guidelines for quality assurance in cervical cancer screening. Summary of the supplements on HPV screening and vaccination. Papillomavirus Res. 2015; 1: 22–31. 12. Huh WK, Ault KA, Chelmow D, et al. Use of primary high-risk human papillomavirus testing for cervical cancer screening: interim clinical guidance. Gynecol Oncol. 2015; 136: 178–182. 13. U.S. Preventive Services Task Force. Screening for cervical cancer: US Preventive Services Task Force recommendation statement. JAMA. 2018; 320: 674–686. 14. Wentzensen N, Arbyn M, Berkhof J, et al. Eurogin 2016 roadmap: how HPV knowledge is changing screening practice. Int J Cancer. 2017; 140: 2192–2200. 15. Wang LD-L, Lam WWT, Fielding R. Hong Kong Chinese parental attitudes towards vaccination and associated socio-demographic disparities. Vaccine. 2016; 34: 1426–1429. 16. Franco EL. Self-sampling for cervical cancer screening: empowering women to lead a paradigm change in cancer control. Curr Oncol. 2018; 25: e1–e3. 17. London School of Hygiene & Tropical Medicine & Path. HPV Vaccine Lessons Learnt Project overview. PATH. 2016. https:// www.path.org/resources/hpv-vaccines-lessons-learnt/ 18. Tatar O, Thompson E, Naz A, et al. Factors associated with human papillomavirus (HPV) test acceptability in primary screening for cervical cancer: a mixed methods research synthesis. Prevent Med. 2018; 116: 40–50. 19. Tatar O, Wade K, McBride E, et al. Are health care professionals prepared to implement human papillomavirus testing? A review of psychosocial determinants of human papillomavirus test acceptability in primary cervical cancer screening. J Wom Health. 2019. Epub ahead of print doi.org/10.1089/jwh.2019.7678 20. Bennett KF, Waller J, Ryan M, Bailey JV, Marlow LAV. The psychosexual impact of testing positive for high-risk cervical human papillomavirus (HPV): a systematic review. Psycho-Oncol. 2019; 28: 1959–1970. 21. Arie, S. HPV: WHO calls for countries to suspend vaccination of boys. BMJ. 2019; 367: l6765 doi:10.1136/bmj.l6765 67 9 Breast Cancer Screening Gabriel M. Leung, Irene O. L. Wong, Ava Kwong, and Joseph T. Wu Introduction Globally breast cancer is the most common cancer in women, accounting for an estimated one-quarter of all malignancies.1 While the disease is now very treatable, there remains subgroups of patients who experience dire outcomes, for instance, those with triple- negative disease. Many survivors report significant disability, both physical and psychosocial. Therefore, any effort to ideally prevent incident disease or at least reduce cancer-related morbidity and mortality would be of enormous benefit to the individual patient as well as whole populations at large. While primary prevention by lifestyle modification and environmental improvements are the ultimate determinants of disease in the long run,2 they are not easily amenable to rapid modification given the long latency period and life-course nature of cancer risks. Screening thus represents the direct, expeditious intervention of choice for many women and policymakers. In this chapter, by screening, we mean mass or population-based screening of well women at average risk of disease unless otherwise specified. One should also take care to distinguish between using the same test modality for screening as opposed to diagnosis or postcancer surveillance. Our scope does not include high-risk groups, who may qualify for enhanced screening, intensive lifestyle modification, and/or even primary chemoprevention. Breast Cancer Biology Determines the Comparative Inefficiency of Screening Unlike two other common cancers (i.e., colorectal and cervical) with unequivocally accepted screening protocols, invasive breast malignancies do not exhibit an obligate precursor stage to allow for precancerous detection and thus intervention. The long-held Wilson and Junger screening criteria require that, among other attributes, “the natural history of the condition, including development from latent to declared disease, should be adequately understood.”3 Modern updates additionally stipulate that there should be a detectable preclinical state.3 Specifically applied to the case of breast cancer, over 80% of women with a previous diagnosis of carcinoma in situ (CIS), whether of the ductal (DCIS) or lobular (LCIS) variety, remain free of invasive breast cancer 20 years later.4 Nevertheless, those who have been diagnosed with in situ conditions indeed demonstrate a two-to threefold elevated risk of developing breast cancer.4 Therefore, regardless of the screening modality deployed or design of the screening program, any preventive attempt to reduce breast cancer–related deaths would be handicapped by the biology of the disease per se. Additionally, both cervical and colorectal cancers have precursor lesions, metaplastic intraepithelial lesions and hyperplastic polyps, respectively, with a sufficiently long sojourn time that are amenable to complete excision and thus primary prevention of invasive disease. In contrast, in situ neoplastic lesions of the breast are nonobligate precursors only occasionally, whereas true obligate precursor states remain to be identified. Efficacy and Effectiveness of Screening Mammography Mammography basically refers to an x-ray examination of the breast. It has been the most intensively scrutinized and the only screening modality shown to reduce breast cancer–related deaths. Since the eight original mammography screening trials involving about half a million mostly Caucasian women in the developed West, there have been many systematic reviews of these data (some with longer-term follow-up) as well as observational effectiveness studies of routine screening programs. They consistently converge on a roughly 20% risk reduction of breast cancer–related mortality in women who start biennial screening from age 50 years. Myers et al.,5 in the most comprehensive and most recent systematic review, reported that three major meta-analyses of the trial data, by the UK independent panel (“Marmot review”6), Canadian task force,7 and Cochrane Collaboration8, have yielded almost identical estimates at 18–20% that reach the 0.05 alpha threshold. Observational cohorts tended toward a larger effect size at 25%, as would be anticipated given the uncontrolled or uncontrollable bias inherent in the noninterventional study design. Figure 9.1 presents summary estimates of the pooled effect sizes by study type and age group. CHAPTER 9 Breast Cancer Screening Study 4 systematic reviews (UK Independent Panel, Canadian Task Force, Cochrane, USPSTF) of 8 RCTs Overall (UK Independent Panel) Overall (Canadian Task Force) Overall (Cochrane) Design Age 40–49 Age ≥50 Age 60–69 Age 70–74 1 systematic review including Meta-analysis of 7 cohort studies 95% CI 0.80 0.82 0.81 0.85 0.86 0.69 1.12 (0.73, 0.89) (0.74, 0.94) (0.74, 0.87) (0.75, 0.96) (0.75, 0.99) (0.54, 0.87) (0.73, 1.72) 0.75 0.62 (0.69, 0.81) (0.56, 0.69) 0.52 (0.42, 0.65) Meta-analysis of Cohort studies Age ≥50 (Invited to screen) Age ≥50 (Accepting screening) 1 systematic review including Meta-analysis of 7 studies RR Systematic review and Meta-analysis of RCTs Meta-analysis of Case-control studies Age 50–70 0.25 0.5 0.75 1 1.25 1.5 1.75 Figure 9.1. Summary estimates of pooled effect sizes of mammography screening on breast cancer mortality, by study type and by age group. Data from Myers et al.5 Other Screening Modalities One major caveat concerning the original randomized controlled trials is that mammographic technology has substantially improved since, and thus the protective benefit of regular mammography has been underestimated. The then-prevailing two-dimensional (2D) analog technique has evolved to full-field digital mammography, with some evidence of lower false positivity and greater sensitivity.9 Further, digital breast tomosynthesis or three-dimensional (3D) mammography has been built on digital mammography to provide 3D reconstruction of breast anatomy in thin image slices. It can be deployed alone or in combination with full-field digital mammography, although the latter would necessarily increase radiation dose exposure. Given the recency of these new technologies, there is not yet definitive evidence to directly show substantive patient outcome–relevant improvements.9 In addition to, or more appropriately in conjunction with, mammography, there are other radiologic techniques commonly deployed to screen for breast cancer—namely ultrasound and magnetic resonance imaging (MRI). However, neither modality has been assessed in randomized trials with hard clinical endpoints, and given widespread organized or haphazard screening practices worldwide, the key question of whether either or both modalities can reduce breast cancer–associated mortality will likely remain unanswered. In women with elevated cancer risk and/or those with dense breasts, the latter being particularly prevalent in East Asian women, ultrasonography can be a useful adjunct to mammography.10 Similarly, MRI as a supplemental screening tool could potentially reveal lesions that are otherwise occult to mammography and ultrasonography. It is usually reserved for women with at least a 1 in 5 lifetime risk for invasive disease, who are typically BrCA1/2 susceptibility gene carriers, but does not include those with an otherwise strong family history.11 It should be noted that with the higher sensitivity that ultrasound or MRI examination usually confers, especially for smaller or more hidden lesions, false positivity necessarily increases with the attendant adverse effects (see “Overdiagnosis and Potential Harms of Screening” later). Finally, there are two other nonradiologic strategies that are commonly used—clinical breast examination (CBE) by a trained health care professional and breast self- examination (BSE). The large Shanghai BSE trial conclusively demonstrated no breast cancer– related survival benefit,12 as did a smaller, earlier Russian trial.13 Inferences based on observational data have nevertheless provided tentative evidence of benefit.10 Whereas there has not been a head-to-head trial comparing CBE vs. no CBE, the Canadian National Breast Screening Study showed that breast cancer–related mortality and nodal involvement were similar among those who received CBE alone and those who underwent both CBE and mammography.14 Thus, this suggests good efficacy of CBE, or looked at another way, it infers little added clinical value of routine mammograms. Of note, however, the very high sensitivity achieved under those trial conditions was double that usually achievable in practice.15 Evidence Gap for the Majority of Women Globally The evidence base for or against screening to reduce breast cancer disease and associated mortality is generally weak for non-Caucasian women outside of North America and Western Europe, who form the vast majority of the world’s female population. For instance, although it is the top female cancer among Chinese populations and the incidence has been increasing,16 the lifetime risk of developing breast cancer in Hong Kong, Shanghai, Singapore, Taiwan, and elsewhere in mainland China remains 32–82% lower than that in Western populations.16–18 The epidemiology (e.g., age distribution) is different between Chinese and Western women, thus exposing the potential frailties of wholesale adoption of 69 70 SECTION II Cancer Screening in Normal and At-Risk inferences drawn from the latter group that have so far dominated the literature.19 Therefore, secondary prevention by mass screening mammography in East Asian women remains controversial with limited direct evidence of benefit supporting its population-based deployment.16,20 Despite such an empirical vacuum, several East Asian governments have begun, since the early 2000s, to offer organized population screening (Table 9.1). Additionally, haphazard opportunistic screening in women at average risk has substantially increased in mainland China, which has sustained the fastest growth of mammographic equipment acquisition in the world over the last decade.21 Nevertheless, at the individual level, it is important to offer women an informed choice, especially in places where the private sector thrives, including the newly developed, prosperous Chinese coastal cities. Individual variation in risk is substantial within any given population. For example, although the average lifetime risk of developing invasive cancer was 4.5% for women in Shanghai, the lifetime risk for women at the 90th risk percentile was 9.5%, which was comparable to the population average in the UK and US.22,23 As such, compared to universal screening conventionally adopted in Western populations, risk-based screening that aims to stratify the female population by their remaining lifetime risk and targets only high-risk women for organized screening would be more in keeping Table 9.1. Breast Cancer Screening Practices in Western and East Asian Settings Country Launch Year Screen Age (Years) Frequency Screening Modality Funding Support Notes UKa 1988 50–70 Every 3 years MM NHS Ongoing randomized trial to evaluate whether extend eligible age range to 47–73 years. USb Since early 1980s USPSTF: 50–74 ACS: 40 and older USPSTF: Every 2 years ACS: Every 1 year for ages 40–54 years; every 1–2 years for 55 years or older MM Individual insurance plans ACS recommends that women continue screening if general health is good with remaining life expectancy of 10 years or longer. Greater Chinac Beijing: 2009 Beijing: 35–59 Shanghai: 2008– Shanghai: 35–74 2012 Beijing: Every 2 years Shanghai: Every 2 years Beijing: CBE, US, (MM for suspected positive) Shanghai: MM Social insurance Shanghai: Only lasted 5 years to 2013. Only women with household registered in Minhang district could join. Hong Kong: — — — — CEWG advises that all women at average risk or high risk of breast cancer should be aware of early symptoms of breast cancer and should visit doctors promptly if symptoms appear. Taiwan: 1999 45–69; 40–44 (with second- degree relatives ever diagnosed with breast cancer) Every 2 years MM Social insurance — South Koread 2002 40 and older Every 2 years MM Social insurance — Japane 2000 40 and older Every 2 years MM, CBE Social insurance — 2002 40–69 50–69 years: Every 2 years 40–49 years: Every 1 year MM Medisave — f Singapore MM = mammography; CBE = clinical breast examination; US = ultrasound; ACS = American Cancer Society; USPSTF = US Preventive Services Task Force; NHS = UK National Health Service; CEWG = Cancer Expert Working Group on Cancer Prevention and Screening, the Government of Hong Kong SAR * Websites were accessed in October 2019. Data sources: a UK NHS: https://www.nhs.uk/conditions/breast-cancer-screening/when-its-offered/ b US Preventive Services Task Force: https://www.uspreventiveservicestaskforce.org/Page/Document/RecommendationStatementFinal/breast-cancer-screening1 American Cancer Society: https://www.cancer.org/health-care-professionals/american-cancer-society-prevention-early-detection-guidelines/breast-cancer-screening-guidelines.html c Beijing: http://www.bjwch.cn/zt/lasc/20120815/19537.shtml Shanghai: https://link.springer.com/article/10.1186/2193-1801-2-276 Hong Kong: https://www.chp.gov.hk/files/pdf/breast_ca_en.pdf Taiwan: https://www.hpa.gov.tw/EngPages/Detail.aspx?nodeid=1051&pid=5957 d South Korea: https://ncc.re.kr/main.ncc?uri=english/sub04_ControlPrograms03 e Japan: https://www.mhlw.go.jp/stf/seisakunitsuite/bunya/0000059490.html Hamashima C. Cancer screening guidelines and policy making: 15 years of experience in cancer screening guideline development in Japan. Japanese Journal of Clinical Oncology. 2018;48(3):278–286. f Singapore: https://www.moh.gov.sg/docs/librariesprovider5/licensing-terms-and-conditions/moh-cir-no-08_2019_6mar19_screening.pdf CHAPTER 9 Breast Cancer Screening with precision preventive care. Indeed, some Western countries have already begun to assess the potential benefits of switching from universal to risk-based screening.24 Recent studies in the UK and US have suggested that personalized screening tailored to individuals’ risks and preferences could improve the efficiency and effectiveness of breast cancer screening.25,26 In East Asia, we were only able to identify one study by Sun et al.,23 who evaluated the cost-effectiveness of risk-based screening conducted based on aggregate data of breast cancer epidemiology in mainland China. Their risk stratification model, however, lacked calibration and validation. Another recent cohort study of more than 1.4 million Taiwanese women27 reported that compared with annual clinical breast examination, risk-based biennial mammography screening only provided a modest reduction in breast cancer mortality (hazard ratio [HR] 0.89, 0.75–1.06) compared to its universal counterpart (HR 0.62, 0.50–0.76). This should be interpreted in the context where 45–49% of women enrolled in their risk-based screening were assessed as high risk and referred for mammography and the proportion adherent to these referrals was 58–62%. That is, 26% of the women enrolled in their risk-based screening underwent mammography. Because they used the initial number of enrolled women (i.e., before risk assessment) as the denominator for calculating HR, the HR reduction in risk-based screening (0.11) was approximately 0.26 times that in universal screening (0.38). Of additional note, we recommend caution in understanding their propensity score–based findings, which concluded a relative risk reduction of 38% for universal biennial screening contrasted with virtually the entire corpus of past work consisting of both randomized controlled trials and empirical evidence5 suggesting about only half that quantum at around 20%. Our own model, trained on Hong Kong data and validated on a Shanghai sample, showed that risk-based and conventional universal mammography screening of women aged 44–69 years biennially would provide similar relative reduction in breast cancer–related mortality among screenees, but the former would be far more cost- effective at half the cost per quality-adjusted life-year (QALY) saved, that is US$18,151 vs. $34,953 per QALY, respectively, compared to no screening.28 In fact, most of the difference was driven by the QALY loss due to false-positive mammograms, arising from complications of consequential confirmatory testing as well as psychosocial implications (see next section). Fundamentally, risk-based strategies optimize screening by reducing unnecessary mammography and tissue biopsy among well women. Therefore, one must consider the undesirable adverse effects induced by screening and avoid as far as possible associated iatrogenic harm imposed on otherwise healthy persons. Overdiagnosis and Potential Harms of Screening Overdiagnosis is defined as the “detection of cancers that would never have been found were it not for the screening test.”29 The Independent UK Panel on Breast Cancer Screening30 surveyed the literature for evidence of overdiagnosis and concluded that 11% of cancers identified through population screening would not otherwise be detected, because those lesions would not have progressed to the symptomatic stage or the affected women would have died from another cause. Presented another way, from an individual screenee’s perspective, the likelihood of overdiagnosis if a woman were to receive a breast cancer diagnosis during the screening period is 19%. The foregoing concerns invasive disease, although carcinoma in situ, particularly DCIS, incidence has increased disproportionately in all populations that have implemented organized screening, and is thus strongly suggestive of overdiagnosis. However, because some DCIS indeed progresses to invasive cancer and can affect prognosis, not all such diagnoses should be similarly classified as overdiagnosis. Findings from the ongoing LORIS trial comparing expectant management with annual mammogram vs. immediate surgery for low- grade DCIS may shed new light on this vexed issue.31 Each overdiagnosed invasive or in situ cancer carries with it the attendant risks, cost, and burden of surgery and possible adjuvant therapies and their impact on personal and professional lives.6,32–34 A large part of the harm associated with screening relates to the need for confirmatory testing subsequent to a positive screen. The 10-year cumulative risk of at least one false-positive result with biennial screening has been estimated to be 42%.35 Associated harm would include fine-needle, core, or infrequently excisional biopsy, with or without radiologic guidance or general anaesthesia, and any complications of wound hematoma or infection.36 The probability of a biopsy arising from an initial false-positive screen over 10 years of biennial mammography was 6–10% in the US and breast biopsy had a complication rate of 8–15%.20,37 Taken together, 1.5–4.5% of low-and average-risk women who undergo biennial screening over a lifetime of routine screening would experience at least one episode of complications due to unnecessary biopsy. There is also considerable psychological harm generated by the large number of false positives and their sequelae,35 as well as misplaced reassurance and delayed treatment of the much smaller number of false negatives.38 Future Directions: Precision Screening and Emerging Modalities Given that the benefit-to-harm tradeoff and cost-effectiveness of screening strongly depend on the QALY loss brought about by unnecessary biopsies and potential complications, future efforts should focus on reducing the need to proceed to tissue biopsy for confirmatory workup. For instance, novel noninvasive diagnostics could become the reflex test after a positive mammogram. The prebiopsy odds of disease for those who would subsequently require the invasive procedure could be improved. For instance, recently developed liquid biopsy modalities may hold such promise.39,40 Liquid biopsy harnesses cancer signals embedded in circulating DNA and protein biomarkers for cancer detection.28,41,42 Working further upstream, moving toward risk-based screening with precise stratification before undergoing mammography would better target those more likely to benefit from screening while minimizing overdiagnosis and potential iatrogenic harm for those at lower risk. To this end, improving the predictive power of the risk stratification algorithm would be important. While the conventional risk factors are well rehearsed, single nucleotide polymorphism (SNP)-based polygenic risk scores have already been shown to improve predictive performance.28 Next-generation programs, with increasing affordability and popularity of genetic risk profiling technology, should routinely include such scores into risk assessment tools. 71 72 SECTION II Cancer Screening in Normal and At-Risk Finally, based on the ethos of patient-centered choice and precision preventive care, women’s own risk preferences43 should be explicitly incorporated into algorithms that inform whether a woman would be recommended to undergo screening. This would further modify the usual universally imposed age thresholds for the general population such that each individual woman could input her own risk tolerance into the decision tool.44 REFERENCES 1. Ferlay J, Colombet M, Soerjomataram J, et al: Estimating the global cancer incidence and mortality in 2018: GLOBOCAN sources and methods. International Journal of Cancer 144:1941–1953, 2019. 2. Research WCRFAIfC: Continuous Update Project Expert Report 2018. Diet, nutrition, physical activity and breast cancer, 2018. 3. Dobrow MJ, Hagens V, Chafe R, et al: Consolidated principles for screening based on a systematic review and consensus process. Canadian Medical Association Journal 190:E422–E429, 2018. 4. To T, Wall C, Baines BJ, et al: Is carcinoma in situ a precursor lesion of invasive breast cancer? International Journal of Cancer 135:1646–1652, 2014. 5. Myers ER, Moorman P, Gierisch JM, et al: Benefits and harms of breast cancer screening. A systematic review. JAMA 314:1615–1634, 2015. 6. Marmot MG, Altman DG, Cameron DA, et al: The benefits and harms of breast cancer screening: an independent review. British Journal of Cancer 108:2205–2240, 2013. 7. Fitzpatrick- Lewis D, Hodgson N, Ciliska D, et al: Canadian Task Force on Preventive Health Care. Breast cancer screening. Canadian Task Force on Preventive Health Care website, 2011. https://canadiantaskforce.ca/guidelines/published-guidelines/ breast-cancer/ 8. Gøtzsche PC, Jørgensen KJ: Screening for breast cancer with mammography. Cochrane Database Systematic Review 6:CD001877, 2013. 9. Hodgson R, Heywang- K€obrunner SH, Harvey SC, et al: Systematic review of 3D mammography for breast cancer screening. The Breast 27:52–61, 2016. 10. Shetty MK: Breast Cancer Screening and Diagnosis. Chapter 2: Screening for breast cancer. Springer- Verlag, New York: Springer, 2015. 11. Oeffinger KC, Fontham ETH, Etzioni R, et al: Breast Cancer Screening for Women at Average Risk—2015 guideline update from the American Cancer Society. JAMA 314:1599–1614, 2015. 12. Thomas DB, Gao DL, Ray RM, Wang WW, Allison CJ, Chen FL, Porter P, Hu YW, Zhao GL, Pan LD, Li W, Wu C, Coriaty Z, Evans I, Lin MG, Stalsberg H, Self SG: Randomized trial of breast self- examination in Shanghai: final results. Journal of the National Cancer Institute 94:1445–1457, 2002. 13. Semiglazov VF, Sagaidak VN, Moiseyenko VM, Mikhailov EA: Study of the role of breast self-examination in the reduction of mortality from breast cancer. The Russian Federation/ World Health Organization Study. European Journal of Cancer 29A:2039–2046, 1993. 14. Miller AB, To T, Baines CJ, et al: The Canadian National Breast Screening Study-1: breast cancer mortality after 11 to 16 years of follow-up. A randomized screening trial of mammography in women age 40 to 49 years. Annals of Internal Medicine 137:305–12, 2002. 15. Barton MB, Harris R, Fletcher SW: The rational clinical examination. Does this patient have breast cancer? The screening clinical breast examination: should it be done? How? JAMA 282:1270–1280, 1999. 16. Fan L, Strasser-Weippl K, Li J-J, et al: Breast cancer in China. Lancet Oncology 15:e279–e289, 2014. 17. Bray F, Ferlay J, Soerjomataram I, et al: Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: A Cancer Journal for Clinicians 68:394–424, 2018. 18. Hong Kong Hospital Authority: Hong Kong Cancer Registry, 2019. 19. Wong IOL, Schooling CM, Cowling BJ, et al: Breast cancer incidence and mortality in a transitioning Chinese population: current and future trends. British Journal of Cancer 112:167–170, 2015. 20. Leung GM, Lam TH, Thach TQ, et al: Will screening mammography in the East do more harm than good? American Journal of Public Health 92:1841–1846, 2002. 21. Grand View Research: Breast Imaging Market Size, Share & Trend Analysis Report by Technology (Ionizing Technology, Non- Ionizing Technology), and Segment Forecasts, 2018—2024, 2018. 22. Zheng W, Wen W, Gao Y-T, et al: Genetic and clinical predictors for breast cancer risk assessment and stratification among Chinese women. Journal of the National Cancer Institute 102:972–981, 2010. 23. Sun L, Legood R, Sadique Z, et al: Cost-effectiveness of risk-based breast cancer screening programme, China. Bulletin of the World Health Organization 96:568–577, 2018. 24. Onega T, Beaber EF, Sprague BL, et al: Breast cancer screening in an era of personalized regimens: a conceptual model and National Cancer Institute initiative for risk-based and preference-based approaches at a population level. Cancer 120:2955–2964, 2014. 25. Pashayan N, Morris S, Gilbert FJ, et al: Cost-effectiveness and benefit- to- harm ratio of risk- stratified screening for breast cancer: a life-table model. JAMA Oncology 4:1504–1510, 2018. 26. Ayer T, Alagoz O, Stout NK: OR Forum—a POMDP approach to personalize mammography screening decisions. Operations Research 60:1019–1034, 2012. 27. Yen AM-F, Tsau H-S, Fann JC-Y, et al: Population-based breast cancer screening with risk-based and universal mammography screening compared with clinical breast examination: a propensity score analysis of 1 429 890 Taiwanese women risk-based and universal mammography vs clinical breast examination risk-based and universal mammography vs clinical breast examination. JAMA Oncology 2:915–921, 2016. 28. Leung K, Wu JT, Wong IOL, et al: Characterizing the epidemiology of breast cancer in Chinese women and using risk stratification to optimize personalized mammography screening (Unpublished manuscript under review). 29. International Agency for Research on Cancer: IARC handbooks of cancer prevention (volume 7): breast cancer screening. Lyon: IARC, 2002. 30. Independent UK Panel on Breast Cancer Screening: The benefits and harms of breast cancer screening: an independent review. The Lancet 380:1778–1786, 2012. 31. Cancer Research UK: A trial comparing surgery with active monitoring for low risk DCIS (LORIS). https://www.cancerresearchuk. org/ a bout- c ancer/ f ind- a - c linical- t rial/ a - t rial- c omparing- surgery-with-active-monitoring-for-low-risk-dcis-loris 32. Assad H, Badhwar G, Bhama S, et al: Impact of breast cancer diagnosis and treatment on sexual dysfunction. Journal of Clinical Oncology 32:125–125, 2017. 33. Stanton AL, Wiley JF, Krull JL, et al: Depressive episodes, symptoms, and trajectories in women recently diagnosed with breast cancer. Breast Cancer Research and Treatment 154:105–115, 2015. 34. Ekenga CC, Perez M, Margenthaler JA, et al: Early-stage breast cancer and employment participation after 2 years of follow-up: a comparison with age-matched controls. Cancer 124:2026–2035, 2018. CHAPTER 9 Breast Cancer Screening 35. Nelson HD, Pappas M, Cantor A, et al: Harms of breast cancer screening: systematic review to update the 2009 U.S. Preventive Services Task Force Recommendation. Annals of Internal Medicine 164:256–267, 2016. 36. Le MT, Mothersill CE, Seymour CB, et al: Is the false-positive rate in mammography in North America too high? British Journal of Radiology 89:20160045, 2016. 37. Hofvind S, Ponti A, Patnick J, et al: False-positive results in mammographic screening for breast cancer in Europe: a literature review and survey of service screening programmes. Journal of Medical Screening 19:57–66, 2012. 38. de Gelder R, van As E, Tilanus-Linthorst MM, et al: Breast cancer screening: evidence for false reassurance? International Journal of Cancer 123:680–686, 2008. 39. Cristiano S, Leal A, Phallen J, et al: Genome- wide cell- free DNA fragmentation in patients with cancer. Nature 570:385–389, 2019. 40. Coy JF: EDIM- TKTL1/ Apo10 blood test: an innate immune system based liquid biopsy for the early detection, characterization and targeted treatment of cancer. International Journal of Molecular Science 18(4):878, 2017. 41. Page K, Shaw JA, Guttery DS: The liquid biopsy: towards standardisation in preparation for prime time. Lancet Oncology 20:758–760, 2019. 42. Heitzer E, Haque IS, Roberts CES, et al: Current and future perspectives of liquid biopsies in genomics-driven oncology. Nature Reviews Genetics 20:71–88, 2019. 43. Cohen JD, Li L, Wang Y, et al: Detection and localization of surgically resectable cancers with a multi-analyte blood test. Science 359:926–930, 2018. 44. Puzhko S, Gagnon J, Simard J, et al: Health professionals’ perspectives on breast cancer risk stratification: understanding evaluation of risk versus screening for disease. Public Health Reviews 40:2, 2019. 73 10 Prostate Cancer Screening Michael A. Diefenbach, Daniel Nethala, Michael Schwartz, and Simon J. Hall Introduction It is commonly accepted that cancer morbidity and mortality are lessened or avoided through regular screening programs. Clear epidemiological evidence supports the utility of cancer screening for skin, cervical, colon, and breast cancer on the population level. The recommendations for population-level screenings of other cancers, such as lung or prostate cancer, are less clearly defined. The current chapter describes the rapid and widespread adoption of the prostate- specific antigen, once discovered, as a biomarker for prostate cancer; its impact on prostate cancer detection and treatment rates; the subsequent revision of population-based screening practices; and finally refined screening recommendations, based on the analyses of several large-scale trials. The example of prostate cancer screening can be seen as a case study of how well-intentioned actions have unintended consequences that go beyond the immediate patient- physician encounter and influence how screening recommendations for the public are formulated, evaluated, and communicated. Prostate Cancer Incidence and Risk Factors Prostate cancer is the most frequently diagnosed and second most lethal cancer in men in the United States (US), with an estimated 174,650 new cases detected and 31,620 cancer-related deaths predicted in 2019.1 Approximately one in nine men in the US will be diagnosed with prostate cancer within their lifetime. The risk factors associated with developing prostate cancer include age, being of African American (AA) descent, and having a family history of the disease. Study of age-specific incidence curves show that the risk of developing prostate cancer begins to rise abruptly after the age of 55 and peaks between 70 and 74, with the median age at diagnosis currently being 66.2,3 AA men are at a higher risk of developing and dying from prostate cancer than their Caucasian counterparts. Studies are not clear as to whether this difference in incidence and mortality is explained entirely by socioeconomic limitations and stage at diagnosis or whether an inherent difference exists in the underlying biology of the cancer in these populations. The hereditary risk of developing prostate cancer was delineated originally by extensive cancer pedigrees showing that individuals with first-degree relatives (father or brothers) with prostate cancer carried a two to three times increased risk of developing the disease over families with no history of the disease.4,5 With the advent of sophisticated sequencing technology, numerous single gene alterations have been identified and confirmed that increase the risk of prostate cancer including BRCA1/2, CHEK2, and ATM.6 Controversies in Prostate Cancer Screening The history of prostate cancer screening is storied and centers around one of the most controversial and the most utilized biomarker for the diagnosis and follow-up of any cancer in clinical medicine—the prostate-specific antigen (PSA).7 Prior to the discovery of the PSA in 1979 and US Food and Drug Administration (FDA) approval for its use in the screening of asymptomatic men in 1994, prostate cancer was only detected by digital rectal examination (DRE). Given that the cancer had to be palpable as a nodule or an irregularity of the prostate to be diagnosed, treatment for prostate cancer resulted in a stagnated cancer-related death rate from about 1950 to 1994, likely due to the advanced stage at time of diagnosis.1 In 1987, Stamey et al. launched the PSA as the primary biomarker for screening prostate cancer in men by demonstrating that serum PSA correlated with advancing stages of prostate cancer, was proportional to the estimated volume of tumor, and reached undetectable levels after complete removal of the prostate.8 As a result of numerous studies and the FDA approval, the PSA and DRE became the standard screening protocol for prostate cancer in primary care for asymptomatic men with threshold levels of PSA warranting a biopsy. This protocol led to a staggering increase in prostate cancer incidence and a significant reduction in cancer-related death rates likely due to the increased detection of nonpalpable, clinically localized cancers. The unrestricted usage of the PSA to screen asymptomatic men for prostate cancer was not without harm. Controversy emerged as the widespread screening practices resulted in overdiagnosing clinically insignificant cancers and subsequent treatment of such cancers. Unrestricted screening led to false-positive biopsies; biopsy-related morbidity including pain, bleeding, and infections; psychological harms for the patient undergoing the biopsy; and treatment-related side effects including erectile dysfunction, urinary incontinence, and possible androgen deprivation. CHAPTER 10 Prostate Cancer Screening Three large randomized controlled clinical trials conducted in the US and in Europe were designed to answer the question of whether screening for prostate cancer with PSA truly saved lives. The results published in 2009–2010 showed that in the US, prostate cancer screening did not save lives,9 while data from the European trials showed that screening produced a 27–40% reduction in prostate cancer–specific mortality.10,11 However, all three trials concluded that screening of asymptomatic men with PSA resulted in substantial overdiagnosis and produced unnecessarily high diagnostic and treatment-related morbidity. As a result of these trials, in 2012 the US Preventative Services Task Force (USPSTF) took the position that prostate cancer screening with PSA should not be recommended routinely by providers, giving it a Grade D recommendation. They stated, “Physicians should not offer or order PSA screening unless they are prepared to engage in shared decision making that enables an informed choice by the patients” and that “there is moderate or high certainty that the service has no benefit or that the harms outweigh the benefits.”12 The Grade D recommendation was based on the conclusion that the data for screening with PSA only affected the prostate cancer–specific mortality of patients and not their overall mortality. The scales balancing the risks of screening with the burdens of overdiagnosis and treatment were tipped in the aftermath of this statement, leading to an immediate 12% decline in the incidence of prostate cancer detected in the year following, a durable 28% decline after 1 year, and ultimately a reverse stage migration with an increased incidence of high-risk disease in the years to follow.13 Factoring this data and the further analysis of the original trials, in 2018 the USPSTF amended their 2012 recommendation of not screening asymptomatic men for prostate cancer to a Grade C recommendation for selectively offering screening for men between the ages of 55 and 69 years. Table 10.1 lists the current guidelines for prostate cancer screening.14–16 The Complexity of Prostate Cancer Screening The changing recommendations of the USPSTF and by professional organizations over the past years about prostate cancer screening has been amplified by reports in the popular press. This led to increased confusion among men who are at normal risk (i.e., without any of the traditional risk factors, such as a family history of prostate cancer or being of AA decent). The overall message about prostate cancer screening is made more difficult as scientists and organizations compare different outcomes that are hard to compare. For example, should one focus on preventable cancer deaths, the prevention of metastatic disease, frequent and common side effects of treatment of consequential and nonconsequential cancers, or quality-of-life outcomes of the patient and the impacted family? Given that prostate cancer is a slow-growing tumor, which affects older men who often suffer from other comorbidities, the decision to be screened is now one to be made by the patient with the help of the health care professional within a shared decision-making paradigm. Due to the complexity of the issue, high levels of uncertainty and dependence on patient preferences make it a perfect example of a shared decision-making approach, but the same complexity is a significant barrier for physicians and patients to engage in such an approach, as it takes time and many providers are not adequately trained in this approach. Factors Influencing Screening Decision Making There are a number of factors that influence patients’ decision to obtain prostate cancer screening. Chief among those factors are physician recommendations. A recent study on providers’ attitudes about recommendations for prostate and colon cancer screening indicated Table 10.1. Current Guidelines for Prostate Cancer Screening Source and Year Recommendation for Prostate Cancer Screening Population Guidelines American Cancer Society (ACS) 2019 Asymptomatic AND life expectancy greater than 10 years No PSA screening Average risk AND life expectancy at least 10 years PSA screening starting at 50 years High risk (African American with first-degree relative diagnosed younger than 65 years old) PSA screening starting at 45 years For all patients The ACS recommends patients to make an informed decision with their physician based on their risk level. Yearly PSA testing should be done for men with a PSA level 2.5 ng/mL or greater. Men with a PSA level less 2.5 ng/mL should be tested every 2 years. 40 years of age or younger No PSA screening 40–54 years of age AND average risk No routine PSA screening 55 years of age or younger AND higher risk (i.e., African American, multiple first-degree relatives diagnosed at a younger age) Individualized screening based on the man’s preference and informed discussion with physician 55–69 years of age Shared decision making coinciding with the man’s preference. Routine screening should be every 2 years, but tailored depending on baseline PSA results. 70 years of age or older (or life expectancy less than 10–15 years) No PSA screening, unless in good health 40–54 years of age Not enough evidence on the benefit of screening in this age group 55–69 years of age Shared decision making coinciding with the man’s preference 70 years of age or older No PSA screening American Urological Association (AUA) 2018 US Preventative Services Task Force (USPSTF) 2018 75 76 SECTION II Cancer Screening in Normal and At-Risk that men often receive cancer screenings even if they are of low value to the patient (i.e., when the net harms clearly outweigh the benefits, as is the case for patients older than 70 years of age, according to the USPSTF recommendation). Factors associated with low-value screening behavior are patient requests for screening and, on the provider side, clinical reminders to perform screening, anticipated regret of a later cancer diagnosis and not recommending screening, and worry about lawsuits. Factors that influence patients’ desire to be screened or not to be screened are a family history of prostate cancer, associated higher levels of anxiety, and cognitive factors such as personal experience and anecdotal evidence. Oftentimes men default to an attitude of “it’s cancer, what do I have to lose?”—an attitude that is also reinforced by providers’ attitude of “better safe than sorry.”17 Screening Disparities among High-Risk Groups The evidence is indisputable that AA/Black men are at a higher risk of developing prostate cancer, present with more aggressive disease, and are 2.5 times more likely to die from prostate cancer than White men. AA men are less likely to be screened, although initial efforts from the Affordable Care Act have narrowed the gap in screening rates between AA and White men.18 Because of the higher-risk status of AA men, screening among this population is recommended under the revised USPSTF guidelines. This is supported by an analysis of Surveillance, Epidemiology, and End Results (SEER) data that supported the notion that screening among AA men is associated with a mortality benefit.18 Yet, despite recommendations for a shared decision-making approach, AA men often lack basic knowledge of prostate cancer screening and its pros and cons, and are often unsure about their preferences about screening.17 A recent study underscored this point: in a sample of 414 AA men, prostate cancer screening was discussed by less than 50% (i.e., 45.2%) of men with their physicians, and overall knowledge about prostate cancer was low, but increased with higher education and income,18 but not with more frequent physician contact. Thus, patients cannot rely on health care providers to be the sole source of health information. Having recognized this, researchers, community organizations, and activists have started to find other ways of informing at-risk populations, such as AA men. A promising avenue is the involvement of faith-based organizations and/or places frequented by AA men, such as barbershops, to educate men about recommended screenings and other lifestyle choices.18 Future Directions Prostate cancer screening recommendations have changed radically over the past two decades. The discovery of the PSA and its strong predictive correlate to prostate cancer led to a paradigm change in prostate cancer screening. Routine screening using the PSA test was quickly adopted by physicians and urologists who welcomed a clinical marker to detect early disease. After a few years of routine screening, mounting clinical and epidemiological evidence pointed to the fact that many small and insignificant cancers were detected and treated, which were unlikely to cause harm to the patient. Physicians were urged to involve patients in the screening and subsequent treatment choice decisions. However, as described in this and the shared decision-making chapter, many barriers to shared decision making still exist and this approach is not universally employed. As a consequence, the onus is on the patient and his family to be informed and to ask the “right” questions to the provider about the pros and cons of a screening decision. But how does a patient know which questions to ask and, more importantly, how to explore his preferences regarding screening and subsequent possible follow-up care? Researchers have attempted to answer this challenge by developing various screening decision aids, either in paper or electronic format. A systematic review and meta-analysis of decision aids on screening choice presented mixed evidence of their utility. Although decision aids are possibly associated with improved screening and disease knowledge, and somewhat of a reduction of decisional conflict, they were not found to be associated with an increased discussion of screening in the first place or an increased uptake of screening.17 A second recent systematic review and meta-analysis, in contrast, reported that the use of decision aids was associated with a decrease in screening choice.18 These seemingly contradicting results point to the need for subgroup analyses as other research, discussed earlier, identified disparities in screening knowledge and utilization of at-risk populations. Future efforts need to focus on the development and implementation of targeted and tailored decision aids that meet the needs of at-risk populations. As clinicians struggle with the optimal way of implementing screening in real-world settings, researchers attempt to develop new models to inform and to reach men. These may include targeting individual groups, such as first-degree relatives of affected men or spouses of at-risk men, with the goal to disseminate information and to identify the pros and cons of screening. Other approaches focus on communities and organizations to educate men and their families. Detailed assessments of these resource-intensive approaches need to be performed to assess their long-term utility. Software-and Internet-based solutions to inform patients about screening options have also been developed, yet they face similar problems than more conventional approaches, such as identifying the desired target audience, as well as technology-specific problems, such as access through a still existing “digital divide,” and mistrust of Internet-based information. The issue of screening for prostate cancer has been instructive on many levels. On a public health level, it demonstrated the perils of adopting a screening tool without long-term data; on a communications level, the revisions of screening recommendations affected and complicated health communications to the public. Overall, the experience surrounding the prostate cancer screening recommendations and resulting controversy is a worthwhile case study to improve future public health and communication efforts. REFERENCES 1. Siegel, R.L., K.D. Miller, and A. Jemal, Cancer statistics, 2019. CA Cancer J Clin, 2019. 69(1): p. 7–34. 2. Gann, P.H., Risk factors for prostate cancer. Rev Urol, 2002. 4 Suppl 5: p. S3–S10. 3. NCI, SEER Cancer Stat Facts: Prostate Cancer. 2019. 4. Steinberg, G.D., et al., Family history and the risk of prostate cancer. Prostate, 1990. 17(4): p. 337–347. CHAPTER 10 Prostate Cancer Screening 5. Spitz, M.R., et al., Familial patterns of prostate cancer: a case-control analysis. J Urol, 1991. 146(5): p. 1305–1307. 6. Zhen, J.T., et al., Genetic testing for hereditary prostate cancer: current status and limitations. Cancer, 2018. 124(15): p. 3105–3117. 7. Hernandez, J., and I.M. Thompson, Prostate-specific antigen: a review of the validation of the most commonly used cancer biomarker. Cancer, 2004. 101(5): p. 894–904. 8. Stamey, T.A., et al., Prostate-specific antigen as a serum marker for adenocarcinoma of the prostate. N Engl J Med, 1987. 317(15): p. 909–916. 9. Andriole, G.L., et al., Mortality results from a randomized prostate- cancer screening trial. N Engl J Med, 2009. 360(13): p. 1310–1319. 10. de Koning, H.J., et al., Prostate cancer mortality reduction by screening: power and time frame with complete enrollment in the European Randomised Screening for Prostate Cancer (ERSPC) trial. Int J Cancer, 2002. 98(2): p. 268–273. 11. Hugosson, J., et al., Mortality results from the Goteborg randomised population-based prostate-cancer screening trial. Lancet Oncol, 2010. 11(8): p. 725–732. 12. Moyer, V.A., and U.S.P.S.T. Force, Screening for prostate cancer: U.S. Preventive Services Task Force recommendation statement. Ann Intern Med, 2012. 157(2): p. 120–134. 13. Barocas, D.A., et al., Effect of the USPSTF Grade D Recommendation against Screening for Prostate Cancer on Incident Prostate Cancer Diagnoses in the United States. J Urol, 2015. 194(6): p. 1587–1593. 14. American Cancer Society, American Cancer Society recommendations for prostate cancer early detection, 2019. Retrieved from: https://www.cancer.org/cancer/prostate-cancer/detection- diagnosis-staging/acs-recommendations.html 15. Ballentine Carter, H., et al., Early detection of prostate cancer: AUA guideline. American Urological Association Guideline, 2018. Retrieved from: https://www.auanet.org/guidelines/ prostate-cancer-early-detection-guideline 16. U.S. Preventative Services Task Force, Final recommendation statement: prostate cancer: screening, 2018. Retrieved from: https://www.uspreventiveservicestaskforce.org/Page/Document/ RecommendationStatementFinal/prostate-cancer-screening1 17. Riikonen, J.M. et al., Decision aids for prostate cancer screening choice: a systematic review and meta-analysis. JAMA Intern Med, 2019. 179(8): p. 1072–1082. 18. Ivlev, I., Jerabkova, S., Mishra, M., Cook, L.A., and K.B. Eden, Prostate cancer screening patient decision aids: a systematic review and meta-analysis. Am J Pre Med, 2018. 55(6): p. 896–907. 77 11 Lung Cancer Screening Lisa Carter-Harris and Jamie Ostroff Introduction Lung cancer kills more people worldwide than breast, colorectal, and prostate cancers combined.1,2 Lung cancer screening is intended to identify earlier, more treatable cases of lung cancer—the leading cause of cancer-related deaths, regardless of gender or ethnicity. More than 1.8 million people are expected to die this year from lung cancer.1,2 Most die because they are diagnosed at an advanced stage with limited treatment options and a 1% 5-year relative survival rate.1 Until 2013, an effective screening test to identify lung cancer at an earlier stage did not exist.3 In the United States, the National Lung Screening Trial (NLST) compared screening long-term, heavy smokers with either low-dose computed tomography (LDCT) of the chest or chest radiography. The NLST randomized 53,454 long-term heavy smokers to a series of annual chest LDCT versus chest radiography, finding a significant life-saving benefit for annual chest LDCT with an approximate 20% relative reduction in lung cancer–related mortality and 6.7% relative reduction in all-cause mortality.3 In response to mounting scientific evidence, including the NLST, the U.S. Preventive Services Task Force (USPSTF) issued a Grade B recommendation for annual lung cancer screening for individuals aged 55 to 80 years who are current smokers or former smokers who have quit within the past 15 years.4 Subsequent research has supported the benefit of lung cancer screening with LDCT of the chest including the Dutch-Belgian Lung Cancer Screening trial (commonly known as the NELSON trial), Danish Lung Cancer Screening trial, and UK Lung Cancer Screening trial.5–7 Although screening programs have been implemented in the U.S. over the past 5 years and offer the potential to detect deadly tumors at an earlier stage, resulting in better outcomes, lung cancer screening uptake remains abysmally low in the U.S. Approximately 9 million Americans qualify for lung cancer screening, but less than 4% have been screened annually.8 Despite the fact that lung cancer screening is a covered preventive service benefit for Medicare beneficiaries and those of many other insurers for long-term smokers who meet eligibility requirements, screening uptake is low. Canada is the only other country that currently recommends lung cancer screening with LDCT of the chest.9 This chapter focuses on lung cancer screening and describes risk factors for the development of lung cancer; reviews the lung cancer screening guidelines as well as factors associated with screening uptake, including psychosocial factors and stigma; and discusses shared decision-making, smoking cessation, clinical implications, and suggestions for future work. Risk Factors While a number of factors influence an individual’s risk for developing lung cancer, the two most common risk factors are cumulative exposure to tobacco smoke and increasing age, respectively.10 The incidence of lung cancer increases with age and occurs most commonly in older individuals aged 55 years and older. While it is important to note that the majority of lung cancers are linked to primary tobacco use and secondhand smoke exposure, approximately 15% of patients are never smokers and the second leading cause of lung cancer is exposure to radon followed by exposure to other environmental toxins as well as a genetic predisposition. The greatest risk factor—tobacco smoking—is a modifiable risk factor on which there is opportunity to intervene, which is why lung cancer screening has the potential to capitalize on a potentially teachable moment for smoking cessation interventions among current smokers who present for screening. Controversy and Consensus Lung cancer screening has experienced a complex journey toward implementation in the U.S. since the issuance of the USPSTF’s Grade B recommendation. This is primarily attributed to the fact that lung cancer screening produces potential risks such as false-positive results, overdiagnosis, radiation exposure, and psychological distress in the presence of frequently noted indeterminate results that require a “wait and watch” approach.3 The following section will review the USPSTF recommendation and discuss eligibility criteria, integration of tobacco treatment into lung cancer screening, and the importance of adherence to an annual screening program for screening eligible persons. USPSTF Recommendation In December 2013, the USPSTF issued the first official guidelines for lung cancer screening (Grade B) in response to the findings of the NLST.4 Prior to this recommendation, there were no CHAPTER 11 Lung Cancer Screening Table 11.1. Differences between Lung Cancer Screening Recommendations Organization Age Parameters Minimum Pack-Year History U.S. Preventive Services Task Force4 55–80 years 30 American Cancer Society1 55–74 years 30 National Comprehensive Cancer Network (Group 1)81 55–77 years 30 National Comprehensive Cancer Network (Group 2)81 50 years or older 20 + one other risk factor Centers for Medicare & Medicaid Services11 55–77 years 30 Canadian Task Force on Preventive Health Care9 55–74 years 30 official recommendations for lung cancer screening. The guidelines target the population of asymptomatic adults with a long history of smoking.4 The population targeted for lung cancer screening is based primarily on the strong evidence from the NLST finding that age, total cumulative exposure to tobacco smoke, and years since quitting smoking are the most important risk factors for the development of lung cancer. LDCT of the chest has high sensitivity and acceptable specificity for detecting lung cancer in high-risk individuals and is the only recommended screening test for lung cancer. In addition to the parameters of age and smoking history, the USPSTF lung cancer screening guidelines also include recommendations on counseling and interventions for tobacco cessation among current smokers. In 2015, the Centers for Medicare and Medicaid Services (CMS) went further by mandating a shared decision-making and counseling visit as a requirement for reimbursement of lung cancer screening in their National Coverage Determination.11 While the USPSTF has issued an official recommendation, there are additional organizations that have issued guidelines that slightly differ, primarily with regard to the upper age limit of who is eligible for screening. These differences as well as the guidelines issued by the Canadian Task Force are summarized in Table 11.1. Eligibility Although the upper age limit varies across professional organization recommendations, based on the USPSTF criteria, individuals are eligible for lung cancer screening if they are aged 55 to 80 years, have a 30 pack-year smoking history, and are a current smoker or a former smoker who quit within the past 15 years.4 Pack-year is calculated by multiplication of the total number of packs of cigarettes smoked per days by the total number of years smoked. This population is recommended to screen annually with LDCT of the chest and to discontinue screening when the individual has either not smoked for 15 years or passes the age threshold. Psychosocial Influences on Lung Cancer Screening Behavior Psychosocial factors can influence lung cancer screening behavior. Unlike other types of cancer screening, having a smoking history adds a layer of psychosocial complexity to the decision-making process for lung cancer screening not present in other types of screening.12 Smoking is a unique health status characteristic that has the potential to negatively impact health behavior primarily due to perceived stigma.13 However, other psychosocial factors such as medical mistrust, lung cancer fatalism, lung cancer worry, and lung cancer fear are also important barriers to screening. To date, the Conceptual Model for Lung Cancer Screening Participation is the only conceptual model specific to lung cancer screening from the perspective of the individual making the decision to screen, or not, for lung cancer.12 See Figure 11.1. Lung Cancer Screening Health Beliefs Health beliefs are concepts that have been used widely to explain the etiology of individual preventive actions, efforts for early detection through screening for certain illnesses including cancer, and individual actions to minimize illness.14 Health beliefs have been documented in lung and other types of cancer screening as potentially modifiable intervention targets on which efforts to increase cancer screening participation may be successful; a subset of health beliefs have been found to mediate the relationship between key antecedent variables and the decision to screen, or not, for lung cancer in preliminary research. Perceived risk, perceived benefits, perceived barriers, and self- efficacy are health beliefs that reflect individual beliefs about lung and other cancer screening behaviors.15,16 Health Belief Model constructs have been reported as important in qualitative research exploring lung cancer screening participation13,15,17,18 and predictive in one study thus far.12 In the context of lung cancer, perceived risk is conceptually defined as individuals’ belief in the likelihood they will develop lung cancer and has been shown to predict intention to screen for lung cancer.17 However, early evidence does not support that perceived risk is correlated with screening behavior,19 which is consistent with research in other areas noting that intention does not necessarily equate to behavior. Perceived benefit is conceptually defined as the belief in the efficacy of an advised course of action to reduce risk through early detection.14 In the context of lung cancer, perceived benefits are individuals’ beliefs about the positive outcomes associated with lung cancer screening participation and potential outcomes from early detection. Higher perceived benefits of lung cancer screening have been associated with increased willingness to participate in lung cancer screening with 209 military veterans.17 In addition, focus group participants (N = 26) reported finding lung cancer early, giving peace of mind, and providing a motivation to quit smoking as three perceived benefits of lung cancer screening.13 Conceptually, perceived barriers are an individual’s belief about the costs (both tangible and psychological) of an advised course of action such as cancer screening.14 In the context of lung cancer, perceived barriers are a person’s estimation of the level of challenge associated with lung cancer screening participation. In lung cancer screening, there are multiple notable perceived barriers to screening such as fear, age, inconvenience, mistrust, and stigma. Fear and the belief that one is too old to benefit from screening as well as inconvenience have been identified as barriers to lung cancer screening participation among eligible individuals.13 Stigma and mistrust will be discussed in more detail in the following section. Self-efficacy is the confidence individuals have in their ability to take action.14 In the 79 80 SECTION II Cancer Screening in Normal and At-Risk Mediators Proximal Outcome Lung Cancer Screening Health Beliefs (perceived risk, perceived benefits, perceived barriers, self-efficacy) Shared DecisionMaking Outcomes Antecedents Psychological Variables (perceived smoking-related stigma, medical mistrust, cancer fatalism, lung cancer worry, lung cancer fear) Demographic and Health Status Characteristics (age, gender, race/ethnicity, income, insurance status, education, smoking status, family history of lung cancer) Cognitive Variables (knowledge: lung cancer and lung cancer screening) Healthcare Provider Recommendation Social and Environmental Variables (social influence, media exposure) Distal Outcome Lung Cancer Screening Participation Stage of Adoption (unaware, unengaged, undecided, decided not to act, decided to act, action, maintenance) Preference-Concordant Decision Reached (to be screened, not to be screened, undecided/delayed decision) Shared Decision-Making Process (patient-provider discussion including healthcare provider recommendation, quality of communication, time, discussion of risks vs benefits, and quality of the patient-provider relationship) Figure 11.1. Conceptual model for lung cancer screening participation. Copyright 2016. Dr. Lisa Carter-Harris. Reprinted with Permission. context of lung cancer, self-efficacy is the confidence that one has the ability to perform all tasks related to arranging and completing lung cancer screening. Self-efficacy has a positive association with lung cancer screening behavior,20 with higher levels of self-efficacy correlated with positive screening follow-through. Further, the link between screening behavior and self-efficacy for lung cancer screening appeared to be fully mediated by cancer fatalism, lung cancer fear, positive family history of lung cancer, knowledge of lung cancer risk and screening, higher annual income, receiving a healthcare clinician recommendation to screen, and social influence (p < 0.05).18 Finally, it should be noted that perceived severity was not included in the Conceptual Model for Lung Cancer Screening Participation secondary to numerous studies reporting that perceived severity is not useful in explaining cancer screening behavior because cancer is universally perceived to be severe, and studies have found ceiling effects when examining this construct in this context. Stigma The reality of lung cancer stigma is tangible and has a profound impact on people living with this disease.21 Similarly, lung cancer stigma has the potential to negatively impact individuals at risk for lung cancer who are considering the option of screening. The stigma of lung cancer is perpetuated by the public’s perception that this is a “smoker’s only” disease when in fact 20% to 30% of individuals diagnosed with lung cancer have never smoked. The prevailing perspective is one of blame and highlighting a lifestyle choice fueled by addiction (i.e., smoking). A recent study conducted by the Lung Cancer Alliance in 2018 to examine the prevalence of lung cancer stigma 10 years after the original study found increased levels of perceived stigma among individuals at risk for the development of lung cancer. Despite best efforts, lung cancer–related stigma has actually increased.22 Ten years ago, there were no approved methods to screen for lung cancer. Now that annual LDCT of the chest is an official recommendation for lung cancer screening by the USPSTF, it is concerning that lung cancer–related stigma is trending upward. Stigma has been noted to be a barrier among individuals at risk for the development of lung cancer. If individuals feel stigmatized, they are less likely to engage in a conversation about the potential for screening as well as actually screening. In a recent study exploring the decision to opt out of screening among screening-eligible patients who had recently received a recommendation to screen for lung cancer from their primary care clinician, some reported they would agree to screening just to conclude the conversation with the clinician as quickly as possible, fearing shame and blame from continued discussion, only to cancel or not show to the scheduled screening.23 Mistrust Medical mistrust is the belief that the healthcare system itself and/ or those working within it are untrustworthy. Medical mistrust has been identified as a barrier to lung cancer screening. Individuals have described mistrust of the healthcare system, tobacco industry, CHAPTER 11 Lung Cancer Screening and government overall as linked with their uncertainty of the value of screening, comparing “new machines to screen to a scam.”13 Because medical mistrust has been associated with late-stage lung cancer presentation among minorities,18 an individual’s mistrust of the healthcare system and/or those working within it is concerning in its potential to impede lung cancer screening participation. Lung Cancer Fatalism Lung cancer fatalism is the belief that being diagnosed with lung cancer will result in death.18 Fatalism is significantly associated with avoidance of lung cancer screening.24 Building upon prior research in colorectal and breast cancer screening that found an association between fatalism and lack of screening,25 early qualitative research suggested a similar relationship in lung cancer screening.18 In a population-based survey of 1,007 adults in Wales, Smits et al. found that low perceived effectiveness of lung cancer screening was significantly associated with cancer fatalism (adjusted odds ratio [aOR] = 6.4; 95% CI = 3.5–11.7; p ≤ 0.001).24 Fatalism often reflects individuals’ desire to not know if they have lung cancer and may be reflective of their fear of treatment or death. Psychological Distress, Anxiety, and Health-Related Quality of Life Given that LDCT of the chest is very sensitive to detecting abnormalities and identifies both cancerous and benign noncalcified nodules, Bach and colleagues26 have cautioned about the potential harms of lung cancer screening including false-positive findings, overdiagnosis, complications of diagnostic procedures, and radiation exposure. In addition to the potential physical risks, these potential harms also include psychological risks such as lung cancer worry, anxiety, and fear, particularly for those with true-positive (lung cancer diagnosis) and false-positive findings. Using NLST data from a large ancillary study, Black and colleagues27 examined the psychosocial impact of undergoing lung cancer screening by examining health-related quality of life (HQOL) and anxious mood (state anxiety). Data were collected at 1 month and 6 months following receipt of screening results using psychometrically sound, patient- reported measurement tools. Global HQOL and anxiety outcomes were compared for individuals receiving one of four screening results: true positives (lung cancer diagnosis), false positives (benign noncancerous nodule), significant incidental findings (e.g., emphysema), and negative screening results. They found no significant differences in HQOL or state anxiety at 1 and 6 months post–lung cancer screening result disclosure between participants with negative screening findings (normal scans) compared to those who either received false-positive or had significant incidental findings. Not surprisingly, those who were found to have lung cancer (true positives) reported lower HQOL and higher state anxiety at both follow-up time points. Clark and colleagues conducted a parallel study in the UK and also found no statistically significant differences in lung cancer worry and health anxiety regardless of LDCT scan results.28 Similarly, no adverse psychological consequences were observed in the Pan-Canadian Early Detection of Lung Cancer. A multicenter Veteran’s Administration (VA) study reported that 25% of patients with incidental pulmonary nodules experienced clinically significant distress.29,30 However, no baseline (prescan) data were collected, thereby limiting the ability to determine whether certain individuals may be more or less prone to postscreening emotional distress because of pre-existing anxiety.29,30 These limitations underscore the importance of examining demographic and psychosocial history factors that may moderate the psychological impact of lung cancer screening. Overall, these findings provide evidence that undergoing lung cancer screening in the context of a high-quality screening and follow-up protocol poses little or no psychosocial harm to screening participants, regardless of scan result. Concern about the emotional impact of undergoing cancer screening, particularly for those who receive abnormal or false- positive results, is neither new nor specific to lung cancer screening.31 The findings reported on the psychosocial impact of undergoing lung cancer screening are generally consistent with the existing literature that has examined the psychological consequences of other types of cancer screening.32 For instance, HQOL and cancer-specific distress outcomes were assessed in the Prostate, Lung, Colorectal and Ovarian Screening Trial.33 No short-or intermediate-term differences by screening results on global HQOL were found; however, participants with abnormal findings reported higher short-term cancer-specific distress than those who received normal results. The overwhelming majority of published studies and systematic reviews have focused on the psychological impact of undergoing routine breast cancer screening and the effect of false-positive mammograms.34,35 Similar results have been observed in the context of colorectal cancer screening.36 Overall, the relevant breast cancer and colon cancer screening literature supports the notion that, beyond the transient anxiety and cancer-specific worry experienced by those with abnormal (including false-positive) findings needing further work-up, undergoing cancer screening per se does not generally result in adverse psychosocial outcomes. It is expected that individuals will experience short-term worry and cancer-specific anxiety when there is a suspicion of lung cancer and they are waiting and hoping for reassurance that they do not have cancer.5 Consistent with recommendations for informing individuals about the risks and benefits of undergoing cancer screening, individuals seeking lung cancer screening should be informed about the sensitivity of LDCT of the chest and the likelihood of needing further work-up because of this test’s high sensitivity. A pilot study reported a promising video intervention to reduce screening anxiety and promote preparedness for lung cancer screening. Screening programs need to be aware of the potential for increased short-term, lung cancer–specific anxiety and worry and provide timely disclosure of results, education, and support, as needed.37 Decision Support and Screening Considering the multiple potential benefits and harms associated with lung cancer screening, multiple professional organizations advocate for shared decision-making in lung cancer screening. In an unprecedented move, the CMS mandated shared decision-making documentation for screening reimbursement. Lung cancer screening is the first cancer screening modality to have shared decision- making mandated by the CMS for reimbursement. Among other components, the specific language of the national coverage determination notes that “shared decision-making, including the use of one or more decision aids, to include benefits and harms of screening, follow-up diagnostic testing, over-diagnosis, false positive rate, and 81 82 SECTION II Cancer Screening in Normal and At-Risk total radiation exposure,”11 is required for reimbursement. As a result, a number of decision support tools including patient decision aids, risk prediction tools, and patient education materials have been developed. The next section will discuss shared decision-making specific to lung cancer screening. Shared Decision-Making in the Context of Lung Cancer Screening Shared decision-making is viewed as the pinnacle of patient-centered care and resonates with the ethical imperative of respect for patient autonomy and engagement.38 Epstein and Street’s summary of the evidence on patient-centered communication posits that clinical encounters between a patient and clinician should be based on collaboration and deliberation.39 Shared decision-making first appeared in the literature in 1982.40 Over the past three decades, shared decision- making has become somewhat synonymous with patient decision aids. The prevailing belief that information exchange via patient decision aids would enhance the patient-clinician collaborative process has resulted in a proliferation of studies that target the effects of decision aids. The decision to screen for lung cancer is not straightforward and is the epitome of a preference-sensitive cancer screening decision best made in the context of shared decision-making. The National Academy of Medicine identified patient-centered care as a critical component of healthcare quality.38 Patient-centered care is defined as “respectful of and responsive to individual preferences, needs, and values, and [ensures] that patient values guide all clinical decisions.”38 This principle emphasizes shared decision-making and enabling patients to become active participants in making healthcare decisions when there is no clear, evidence-based “right” choice. Shared decision-making involves weighing benefits against harms with the premise that shared decision-making can result in a more informed decision and better quality of care.41 Shared decision- making is now considered a critical component of patient-centered care, which has contributed to the recent paradigm shift in health communication from a paternalistic authority toward a communication process shared between the clinician and the patient with an emphasis on decision support to promote good medical decision making.”38,41 Many investigators have attempted to enhance shared decision-making by developing and implementing patient decision aids in clinical care without truly understanding the components that lead to positive patient decisional outcomes (i.e., high decision quality) or behavioral outcomes. This has led to inconsistencies reported in the literature such as variable decision aid effects,42 lack of evidence of associations between shared decision-making empirical measures and patient behavioral and health outcomes,43 and lack of knowledge regarding the relationship between communication quality and cancer screening.44 Two studies noted that the decision to screen, or not, for lung cancer was not associated with knowledge or values clarification; rather, it was associated with patient-clinician communication quality.23,45 To date, psychosocial factors that influence the shared decision-making process in lung cancer screening has not been explored. For patients who chose not to screen, most (72%) described a short or limited discussion with their clinician that, from their perspective, focused on the fact that they were eligible and did not include any shared discussion.23 In addition to low knowledge levels about screening benefits and potential harms, patients reported encounters with clinicians where opportunities to fully consider the benefits versus risks of lung cancer screening were not provided nor did they perceive they had made an informed decision. For patients who chose to be screened, knowledge levels also were low, and while participants perceived benefits to detecting lung cancer early, they reported having no discussion about potential screening harms. Both studies highlight the importance of the patient- clinician discussion and communication quality in shared decision-making, and the current knowledge gap about which shared decision-making components lead to positive patient decisional and behavioral outcomes. Integration of Tobacco Treatment in Lung Cancer Screening Widely regarded as a potential teachable moment for tobacco cessation advice and treatment, lung cancer screening offers a critical opportunity to promote smoking cessation and reduce further lung cancer morbidity and mortality46 and the cost-effectiveness of lung cancer screening.27 Despite the potential for reaching high-risk, vulnerable populations, there is strong consensus from systematic reviews and meta-analysis that merely undergoing lung cancer screening itself in the absence of delivery of evidence-based tobacco treatment has limited sustained effect on smoking cessation.47 Quit rates from several international lung cancer screening trials have shown mixed results with regard to the impact of undergoing screening on smoking behavior. Data from the Dutch-Belgian Lung Cancer Screening trial (commonly known as the NELSON trial) suggested a possible negative impact on smoking as screening was associated with a lower prolonged abstinence rate compared to the control group (14.5% vs. 19.1%; odds ratio [OR] = 1.40; 95% CI = 1.01–1.92; p < 0.05).48 The VA Demonstration Project49 and the Danish Lung Cancer Screening trial showed no significant impact of screening on smoking cessation,6 whereas data from the UK Lung Cancer Screening trial demonstrated a positive impact of screening with quit rates of 14% vs. 8% observed at 1 year and 24% vs. 21% at 2 years in the screening vs. control arms.7 The impact was especially strong among those with a positive LDCT result. An ancillary study of the NLST reported that 37% of smokers had quit at their last follow-up assessment and that likelihood of quitting was greater among participants reporting higher perceived severity of smoking-related diseases, greater self-efficacy for quitting, and fewer perceived barriers to quitting.50 In the U.S., the CMS requires that smoking cessation is offered to all current smokers seeking lung cancer screening and virtually all professional health organizations and societies recommend smoking cessation services as an essential aspect of high-quality lung cancer screening.51 Despite the strong endorsement that high-quality lung cancer screening should include integration of evidence-based tobacco treatment, the optimal approach for delivering feasible, cost- effective, and sustainable cessation interventions in the context of lung cancer screening remains largely unknown.52 There is considerable interest in developing and testing the effectiveness of various types and doses of tobacco treatment delivery,53 such as telephone or in-person counseling, pharmacotherapy, and other cessation support strategies. The U.S. National Cancer Institute has established the Smoking Cessation at Lung Examination (SCALE) collaboration,54 a network of randomized clinical trials testing the cost-effectiveness of various tobacco treatment models in the context of lung cancer screening, and several SCALE and other study protocols have been published.55,56 Results from systematic reviews of prior studies examining the effectiveness of integrating smoking cessation within the context of CHAPTER 11 Lung Cancer Screening lung cancer screening have yielded mixed yet promising findings.57– 60 One pilot study showed promise in offering screening participants telephone counseling;60 however, a larger trial did not demonstrate that an opt-out offer of telephone counseling for all smokers seeking LDCT was superior to a mailed brochure containing contact information for cessation services at increasing cessation rates at 12 months.61 Brief cessation counseling delivered on the day of LDCT screening may be feasible but not adequately intensive for long-term smoking abstinence.59 Two small studies of intensive cessation support found favorable cessation rates.62,63 A recently published systematic review and meta-analysis64 of the effectiveness of smoking cessation in the context of lung cancer screening identified 83 relevant trials and categorized their tobacco treatments as electronic/ web- based intervention, in- person counseling, pharmacotherapy, and telephone counseling. At 6-month follow-up, electronic/web-based (OR = 1.14; 95% CI = 1.00–1.25), in-person counseling (OR = 1.46; 95% CI = 1.25–1.70), and pharmacotherapy (OR = 1.53; 95% CI = 1.33–1.77) interventions significantly increased the odds of smoking abstinence. Telephone counseling increased the odds of quitting but did not reach statistical significance. At 12-month follow-up, in-person counseling and pharmacotherapy remained efficacious. The potential benefits for lung cancer screening to accelerate smoking cessation may go unrealized in the absence of access to evidence-based tobacco treatments. In an ancillary NLST study, clinician-delivered 5As (the five major steps to tobacco cessation intervention: Ask, Advise, Assess, Assist, and Arrange), particularly offering assistance and arranging follow-up, was associated with smoking cessation after lung cancer screening; however, delivery of cessation assistance was relatively low and arranging follow-up cessation counseling was very low.65 Screening sites vary in their readiness and capacity for delivering high-quality tobacco treatment. In a national survey of smoking cessation practices at lung cancer screening sites, most sites reported assessing patients’ smoking status (99%) and advising to quit (91%), but fewer provided cessation coaching or referral (60%) or recommended cessation medications (33%).66 Patient-, clinician-, and system-level barriers exist and await further needed research on implementation processes and outcome. Engagement of smokers seeking lung cancer screening in tobacco treatment services can also be challenging. Individuals seeking lung cancer screening present with variable levels of cessation motivation67 and are likely different in many ways from volunteers who participated in the initial screening trials.7,68 Some screening-seeking smokers report low quitting readiness,46,69 whereas others are highly motivated to quit and may have actively sought out screening because of concerns about their lung health.54 Given the older age and heavy smoking history of screening eligible smokers, it is important to address smokers’ nihilism (“why bother”?) and acknowledge that there is compelling evidence that cessation among older, long-term smokers will prevent deaths from lung cancer and other tobacco- related diseases.70 It is apparent that simply understanding the elevated risk of lung cancer and other diseases associated with smoking does not necessarily lead to smoking cessation.71 Overestimation of the benefit of cancer screening alone and the minimization of harm reduction that can be achieved through smoking behavior change may contribute to low uptake of tobacco treatment services.72 A recent qualitative study involving a small sample (N = 45) of NLST participants showed that while most patients reported that screening led to increased reflection on the harms and long-term consequences of smoking, as many as half of the patients reported avoidance of thinking about perceived lung cancer risk.71 There is a misperception among some patients that “undergoing screening yields the same benefits as smoking cessation” and “everyone who undergoes screening will benefit.”73 In a larger subsample of smokers (N = 430) from the NLST assessed 1 year following their initial LDCT, patients showed no significant changes in risk perceptions from baseline as a function of screening results, nor was risk perception associated with quitting, which occurred in only 10% of the patients.74 Many critics of lung cancer screening have cautioned that negative screening results may mitigate current smokers’ concerns about the health consequences of smoking, thereby reducing motivation to quit. There remain concerns about whether those who receive a negative screening scan result will perceive a “license to smoke,” also referred to as a “health certificate” effect,52,73,75 and false reassurance resulting in reduced quitting motivation.73 To date, findings examining whether lung cancer screening results (normal, incidental, or suspicious) influence cessation outcomes have been mixed. It is encouraging that consistently negative scans have not been associated with greater relapse among long- term former smokers or among baseline smokers. In a substudy69 of smokers who participated in the NLST, a false-positive screen was associated with increased smoking cessation among current smokers and less relapse among recent quitters. Fortunately, consistently negative screens were not associated with greater relapse among long-term former smokers. Adherence to Annual Repeat Screening, Follow-Up, and Treatment Recommendations Promoting adherence to annual repeat screening guidelines as well as follow-up and treatment recommendations for screening abnormalities are all necessary for ensuring that lung cancer screening achieves its full benefit in reducing lung cancer morbidity and mortality. Although identifying and addressing barriers for adherence is a common behavioral issue for all cancer screening guidelines, the research literature focusing on understanding the extent and drivers of adherence in the context of lung cancer screening is nascent and is currently informed by findings from relevant studies examining adherence to older, well-established cancer screening guidelines such as breast and colorectal cancer. To date, older lung cancer screening studies examining chest radiography have found wide variation in repeat screening nonadherence with rates ranging from 10% to 45%.76,77 Within the Prostate, Lung, Colorectal, and Ovarian screening trial, 14% of screening enrollees were nonadherent to one or more annual, repeat screenings using chest radiography.78 Among individuals with a false-positive screening result, nonadherence was 17.2% compared to 10.3% among those with negative screening results. Montes and colleagues found that 45% of smokers did not return for annual repeat lung cancer screening with LDCT.79 Future Directions Given expertise in the psychological and behavioral aspects of cancer prevention and control, there are critical research needs and clinical roles for psycho-oncologists to play in realizing the full 83 84 SECTION II Cancer Screening in Normal and At-Risk public health benefit of lung cancer screening. There are also several high-priority research needs that will benefit from continued input from investigators with relevant expertise. As lung cancer screening programs are more widely implemented, it is critical to identify the most salient factors associated with lung cancer screening participation. Early research has supported that lung cancer screening participation is influenced by multilevel factors at the individual, provider, and healthcare system levels. There is value in understanding all variables at all levels that have the potential to influence clinical outcomes. However, for lung cancer screening to be effective, addressing the critical psychosocial factors and their key relationships associated with screening behavior in this population at the individual level is vital. The current state of the science has provided researchers with relevant psychosocial factors and informed a theoretical foundation on which to develop tailored interventions for those most at risk for lung cancer. To better understand the psychological harms of lung cancer screening, it is also recommended that lung cancer screening registries capture follow-up data on clinically meaningful psychological distress and lung cancer–specific anxiety. It is important to consider interventions such as the addition of nurse navigators to assist patients toward scheduling and completion of the initial lung scan as well as to promote adherence to follow-up. This may serve as a layer of psychosocial support for the patient as well as individuals newly diagnosed with lung cancer as they navigate the healthcare system. As the science moves forward, it is essential that multilevel interventions be developed to address the individual psychosocial variables that have the potential to negatively influence lung cancer screening participation, to support both patients and providers in the shared decision-making process specific to a complex cancer screening decision like lung cancer, and to increase awareness of the need for greater access to psychosocial support services for the individual.80 Specifically, deconstructing the components of patient-clinician discussions that lead to positive decision and behavioral outcomes is critical to inform the development of the next generation of decision support tools and alternative communication strategies, and to shift the paradigm further in health communication beyond increasing patient knowledge. Future research is also needed to better understand the role of lung cancer screening results and associated risk perceptions on smoking cessation. Future research is needed to identify effective ways of communicating lung cancer screening results so as to optimize comprehension, motivate health behavior change (i.e., quitting smoking), promote adherence to repeat annual screening/follow-up recommendations, and minimize transient anxiety. REFERENCES 1. American Cancer Society. Cancer Facts & Figures. 2019; https:// www.cancer.org/ r esearch/ c ancer- f acts- s tatistics/ a ll- c ancer- facts-figures/cancer-facts-figures-2019.html. Accessed February 28, 2019. 2. Latest global cancer data: Cancer burden rises to 18.1 million new cases and 9.6 million cancer deaths in 2018 [Press release]. September 12, 2018. 3. Aberle DR, Adams AM, Berg CD, et al. Reduced lung-cancer mortality with low-dose computed tomographic screening. N Engl J Med. 2011;365(5):395–409. 4. U.S. Preventive Services Task Force. United States Preventive Services Task Force Lung Cancer Screening Guidelines. 2013; https://www.uspreventiveservicestaskforce.org/ Accessed February 28, 2019. 5. van den Bergh KA, Essink-Bot ML, Bunge EM, et al. Impact of computed tomography screening for lung cancer on participants in a randomized controlled trial (NELSON trial). Cancer. 2008;113(2):396–404. 6. Ashraf H, Saghir Z, Dirksen A, et al. Smoking habits in the randomised Danish Lung Cancer Screening Trial with low-dose CT: Final results after a 5-year screening programme. Thorax. 2014;69(6):574–579. 7. Brain K, Carter B, Lifford KJ, et al. Impact of low-dose CT screening on smoking cessation among high-risk participants in the UK Lung Cancer Screening Trial. Thorax. 2017;72(10):912–918. 8. Jemal A, Fedewa SA. Lung cancer screening with low-dose computed tomography in the United States-2010 to 2015. JAMA Oncol. 2017;3(9):1278–1281. 9. Canadian Task Force on Preventive Health Care. Lung Cancer (2016). 2016; https://canadiantaskforce.ca/guidelines/published- guidelines/lung-cancer/. Accessed January 14, 2020. 10. Centers for Disease Control and Prevention. 2019; https://www. cdc.gov. 11. Mullen KA, Coyle D, Manuel D, et al. Economic evaluation of a hospital-initiated intervention for smokers with chronic disease, in Ontario, Canada. Tob Control. 2015;24(5):489–496. 12. Carter-Harris L, Davis LL, Rawl SM. Lung cancer screening participation: Developing a conceptual model to guide research. Res Theory Nurs Pract. 2016;30(4):333–352. 13. Carter-Harris L, Ceppa DP, Hanna N, Rawl SM. Lung cancer screening: What do long-term smokers know and believe? Health Expect. 2017;20(1):59–68. 14. Champion VL, Skinner CS, Menon U, et al. A breast cancer fear scale: Psychometric development. J Health Psychol. 2004;9(6):753–762. 15. Byrne MM, Weissfeld J, Roberts MS. Anxiety, fear of cancer, and perceived risk of cancer following lung cancer screening. Med Decis Making. 2008;28(6):917–925. 16. Wools A, Dapper EA, Leeuw JRJd. Colorectal cancer screening participation: A systematic review. Eur J Public Health. 2015;26(1):158–168. 17. Tanner NT, Egede LE, Shamblin C, Gebregziabher M, Silvestri GA. Attitudes and beliefs toward lung cancer screening among US Veterans. Chest. 2013;144(6):1783–1787. 18. Bergamo C, Lin JJ, Smith C, et al. Evaluating beliefs associated with late-stage lung cancer presentation in minorities. J Thorac Oncol. 2013;8(1):12–18. 19. Carter-Harris L, Slaven JE, Monohan P, Rawl SM. Development and psychometric evaluation of the lung cancer screening health belief scales. Cancer Nurs. 2017;40(3):237–244. 20. Carter-Harris L, Slaven JE, Monahan PO, Draucker CB, Vode E, Rawl SM. Understanding lung cancer screening behaviour using path analysis. J Med Screen. 2020;27(2):105–112. 21. Goldman RE, Diaz JA, Kim I. Perspectives of colorectal cancer risk and screening among Dominicans and Puerto Ricans: Stigma and misperceptions. Qual Health Res. 2009;19(11):1559–1568. 22. Carter-Harris L. Leveraging social media to change the public conversation on lung cancer stigma. World Conference on Lung Cancer. September 2018; Toronto, Canada. 23. Carter-Harris L, Brandzel S, Wernli KJ, Roth JA, Buist DSM. A qualitative study exploring why individuals opt out of lung cancer screening. Fam Pract. 2017;34(2):239–244. CHAPTER 11 Lung Cancer Screening 24. Smits SE, McCutchan GM, Hanson JA, Brain KE. Attitudes towards lung cancer screening in a population sample. Health Expect. 2018;21(6):1150–1158. 25. Miles A, Rainbow S, von Wagner C. Cancer fatalism and poor self-rated health mediate the association between socioeconomic status and uptake of colorectal cancer screening in England. Cancer Epidemiol Biomarkers Prev. 2011;20(10):2132–2140. 26. Bach PB, Mirkin JN, Oliver TK, et al. Benefits and harms of CT screening for lung cancer: A systematic review. JAMA. 2012;307(22):2418–2429. 27. Black WC, Gareen IF, Soneji SS, et al. Cost-effectiveness of CT screening in the National Lung Screening Trial. N Engl J Med. 2014;371(19):1793–1802. 28. Clark ME, Bedford LE, Young B, et al. Lung cancer CT screening: Psychological responses in the presence and absence of pulmonary nodules. Lung Cancer. 2018;124:160–167. 29. Taghizadeh N, Tremblay A, Cressman S, et al. Health-related quality of life and anxiety in the PAN-CAN lung cancer screening cohort. BMJ Open. 2019;9(1):e024719. 30. Freiman MR, Clark JA, Slatore CG, et al. Patients’ knowledge, beliefs, and distress associated with detection and evaluation of incidental pulmonary nodules for cancer: Results from a multicenter survey. J Thorac Oncol. 2016;11(5):700–708. 31. Wardle J, Pope R. The psychological costs of screening for cancer. J Psychosom Res. 1992;36(7):609–624. 32. Chad-Friedman E, Coleman S, Traeger LN, et al. Psychological distress associated with cancer screening: A systematic review. Cancer. 2017;123(20):3882–3894. 33. Taylor KL, Shelby RR, Gelmann EE, McGuire CC. Quality of life and trial adherence among participants in the prostate, lung, colorectal, and ovarian cancer screening trial. J Natl Cancer Inst. 2004;96(14):1083–1094. 34. Brewer NT, Salz T, Lillie SE. Systematic review: The long- term effects of false-positive mammograms. Ann Intern Med. 2007;146(7):502–510. 35. Salz T, Richman AR, Brewer NT. Meta-analyses of the effect of false-positive mammograms on generic and specific psychosocial outcomes. Psycho-oncology. 2010;19(10):1026–1034. 36. van der Velde JL, Blanker MH, Stegmann ME, de Bock GH, Berger MY, Berendsen AJ. A systematic review of the psychological impact of false-positive colorectal cancer screening: What is the role of the general practitioner? Eur J Cancer Care (Engl). 2017;26(3):10.1111/ecc.12709. doi:10.1111/ecc.12709. 37. Raz DJ, Nelson RA, Kim JY, Sun V. Pilot study of a video intervention to reduce anxiety and promote preparedness for lung cancer screening. Cancer Treat Res Comm. 2018;16:1–8. 38. Institute of Medicine (US) Committee on Quality of Health Care in America. Crossing the Quality Chasm: A New Health System for the 21st Century. Washington, DC: National Academies Press; 2001. 39. Epstein RM, Street RL. Patient- Centered Communication in Cancer Care: Promoting Healing and Reducing Suffering. Bethesda, MD: National Cancer Institute, NIH Publication No. 07-6225; 2007. 40. Advisory Commission’s Final Report “Quality First: Better Health Care for All Americans.” President’s Advisory Commission on Consumer Protection and Quality in the Health Care Industry. 1998. https://govinfo.library.unt.edu/hcquality/ 41. Hamilton JG, Lillie SE, Alden DL, et al. What is a good medical decision? A research agenda guided by perspectives from multiple stakeholders. J Behav Med. 2017;40(1):52–68. 42. Lillie SE, Partin M, Rice K, et al. The Effects of Shared Decision Making on Cancer Screening—A Systematic Review. Washington, DC: Department of Veterans Affairs (US); September; 2014. 43. Shay LA, Lafata JE. Where is the evidence? A systematic review of shared decision making and patient outcomes. Med Decis Making. 2015;35(1):114–131. 44. Peterson EB, Ostroff JS, DuHamel KN, et al. Impact of provider- patient communication on cancer screening adherence: A systematic review. Prev Med. 2016;93:96–105. 45. Roth JA, Carter-Harris L, Brandzel S, Buist DSM, Wernli KJ. A qualitative study exploring patient motivations for screening for lung cancer. PloS One. 2018;13(7):e0196758. 46. Taylor KL, Cox LS, Zincke N, Mehta L, McGuire C, Gelmann E. Lung cancer screening as a teachable moment for smoking cessation. Lung Cancer. 2007;56(1):125–134. 47. Slatore CG, Baumann C, Pappas M, Humphrey LL. Smoking behaviors among patients receiving computed tomography for lung cancer screening. Systematic review in support of the U.S. preventive services task force. Ann Am Thorac Soc. 2014;11(4):619–627. 48. van der Aalst CM, Bergh KAMvd, Willemsen MC, de Koning HJ, van Klaveren RJ. Lung cancer screening and smoking abstinence: 2 year follow-up data from the Dutch–Belgian randomised controlled lung cancer screening trial. Thorax. 2010;65(7):600–605. 49. Fu SS, Melzer AC, Fabbrini AE, et al. Association of invitation to lung cancer screening and tobacco use outcomes in a VA demonstration project. Prev Med Rep. 2019;16:101023. 50. Kaufman AR, Dwyer LA, Land SR, Klein WMP, Park ER. Smoking- related health beliefs and smoking behavior in the National Lung Screening Trial. Addictive Behav. 2018;84:27–32. 51. Fucito LM, Czabafy S, Hendricks PS, Kotsen C, Richardson D, Toll BA. Pairing smoking- cessation services with lung cancer screening: A clinical guideline from the Association for the Treatment of Tobacco Use and Dependence and the Society for Research on Nicotine and Tobacco. Cancer. 2016;122(8):1150–1159. 52. Pedersen JH, Tønnesen P, Ashraf H. Smoking cessation and lung cancer screening. Ann Transl Med. 2016;4(8):157. 53. Minnix JA, Karam-Hage M, Blalock JA, Cinciripini PM. The importance of incorporating smoking cessation into lung cancer screening. Transl Lung Cancer Res. 2018;7(3):272–280. 54. Joseph AM, Rothman AJ, Almirall D, et al. Lung cancer screening and smoking cessation clinical trials. SCALE (smoking cessation within the context of lung cancer screening) collaboration. Am J Respir Crit Care Med. 2018;197(2):172–182. 55. Meltzer LR, Unrod M, Simmons VN, et al. Capitalizing on a teachable moment: Development of a targeted self-help smoking cessation intervention for patients receiving lung cancer screening. Lung Cancer. 2019;130:121–127. 56. Taylor KL, Deros DE, Fallon S, et al. Study protocol for a telephone- based smoking cessation randomized controlled trial in the lung cancer screening setting: The lung screening, tobacco, and health trial. Contemp Clin Trials. 2019;82:25–35. 57. Pua BB, Dou E, O’Connor K, Crawford CB. Integrating smoking cessation into lung cancer screening programs. Clin Imaging. 2016;40(2):302–306. 58. Iaccarino JM, Duran C, Slatore CG, Wiener RS, Kathuria H. Combining smoking cessation interventions with LDCT lung cancer screening: A systematic review. Prev Med. 2019;121:24–32. 59. Marshall HM, Courtney DA, Passmore LH, et al. Brief tailored smoking cessation counseling in a lung cancer screening population is feasible: A pilot randomized controlled trial. Nicotine Tob Res. 2016;18(7):1665–1669. 60. Zeliadt SB, Greene PA, Krebs P, et al. A proactive telephone- delivered risk communication intervention for smokers 85 86 SECTION II Cancer Screening in Normal and At-Risk 61. 62. 63. 64. 65. 66. 67. 68. 69. 70. participating in lung cancer screening: A pilot feasibility trial. J Smok Cessat. 2018;13(3):137–144. Tremblay A, Taghizadeh N, Huang J, et al. A randomized controlled study of integrated smoking cessation in a lung cancer screening program. J Thorac Oncol. 2019;14(9):1528–1537. Ferketich AK, Otterson GA, King M, Hall N, Browning KK, Wewers ME. A pilot test of a combined tobacco dependence treatment and lung cancer screening program. Lung Cancer. 2012;76(2):211–215. Pozzi P, Munarini E, Bravi F, et al. A combined smoking cessation intervention within a lung cancer screening trial: A pilot observational study. Tumori. 2015;101(3):306–311. Cadham CJ, Jayasekera JC, Advani SM, et al. Smoking cessation interventions for potential use in the lung cancer screening setting: A systematic review and meta-analysis. Lung Cancer. 2019;135:205–216. Park ER, Gareen IF, Japuntich S, et al. Primary care provider- delivered smoking cessation interventions and smoking cessation among participants in the National Lung Screening Trial. JAMA Intern Med. 2015;175(9):1509–1516. Ostroff JS, Copeland A, Borderud SP, Li Y, Shelley DR, Henschke CI. Readiness of lung cancer screening sites to deliver smoking cessation treatment: Current practices, organizational priority, and perceived barriers. Nicotine Tob Res. 2016;18(5):1067–1075. Young B, Vedhara K, Kendrick D, et al. Determinants of motivation to quit in smokers screened for the early detection of lung cancer: A qualitative study. BMC Public Health. 2018;18(1):1276. Tammemagi MC, Berg CD, Riley TL, Cunningham CR, Taylor KL. Impact of lung cancer screening results on smoking cessation. J Natl Cancer Inst. 2014;106(6):dju084. Clark MA, Gorelick JJ, Sicks JD, et al. The relations between false positive and negative screens and smoking cessation and relapse in the National Lung Screening Trial: Implications for public health. Nicotine Tob Res. 2016;18(1):17–24. Gellert C, Schottker B, Brenner H. Smoking and all-cause mortality in older people: Systematic review and meta-analysis. Arch Intern Med. 2012;172(11):837–844. 71. Park ER, Streck JM, Gareen IF, et al. A qualitative study of lung cancer risk perceptions and smoking beliefs among national lung screening trial participants. Nicotine Tob Res. 2014;16(2):166–173. 72. Fagerlin A, Sepucha KR, Couper MP, Levin CA, Singer E, Zikmund-Fisher BJ. Patients’ knowledge about 9 common health conditions: The DECISIONS survey. Med Decis Making. 2010;30(5 Suppl):35S–52S. 73. Zeliadt SB, Heffner JL, Sayre G, et al. Attitudes and perceptions about smoking cessation in the context of lung cancer screening. JAMA Intern Med. 2015;175(9):1530–1537. 74. Park ER, Gareen IF, Jain A, et al. Examining whether lung screening changes risk perceptions: National Lung Screening Trial participants at 1-year follow-up. Cancer. 2013;119(7):1306–1313. 75. van der Aalst CM, van Klaveren RJ, van den Bergh KAM, Willemsen MC, de Koning HJ. The impact of a lung cancer computed tomography screening result on smoking abstinence. Eur Respir J. 2011;37(6):1466–1473. 76. Melamed MR, Flehinger BJ, Zaman MB, Heelan RT, Perchick WA, Martini N. Screening for early lung cancer. Results of the Memorial Sloan-Kettering study in New York. Chest. 1984;86(1):44–53. 77. Kubik A, Parkin DM, Khlat M, Erban J, Polak J, Adamec M. Lack of benefit from semi-annual screening for cancer of the lung: Follow- up report of a randomized controlled trial on a population of high- risk males in Czechoslovakia. Int J Cancer. 1990;45:26–33. 78. Ford ME, Havstad SL, Flickinger L, Johnson CC. Examining the effects of false positive lung cancer screening results on subsequent lung cancer screening adherence. Cancer Epidemiol Biomarkers Prev. 2003;12(1):28–33. 79. Montes U, Seijo LM, Campo A, Alcaide AB, Bastarrika G, Zulueta JJ. Factors determining early adherence to a lung cancer screening protocol. Eur Respir J. 2007;30(3):532. 80. Kathuria H, Detterbeck FC, Fathi JT, et al. Stakeholder research priorities for smoking cessation interventions within lung cancer screening programs. Am J Respir Crit Care Med. 2017;196(9):1202–1212. 81. Lung Cancer Screening Guidelines. NCCN Guidelines for Patients. 2020; https://www.nccn.org/patients/guidelines/lung_screening/ index.html. 12 Skin Cancer Screening Jennifer L. Hay and Stephanie N. Christian Introduction Skin cancers are the most commonly diagnosed cancer in the United States (US), and they can very often be efficiently detected on the skin surface, as they usually arise as a skin change or growth on the skin.1 Melanoma, the most deadly form of skin cancer, is the fifth most common cancer in the US, with about 96,500 persons expected to develop melanoma in 2019.1 Unfortunately, the incidence of melanoma has tripled in the last four decades, and the incidence and mortality are highest among adults over 55,2 yet there is also a rising prominence of this potentially fatal skin cancer in young adults. Over 4.3 million adults are treated for keratinocyte cancers, including squamous cell carcinoma (SCC) and basal cell carcinoma (BCC), annually in the US, and population-based studies show these cancers are increasing steadily. Although keratinocyte cancers, in particular BCC, impart low risk of death, they are common and expensive to treat. Most melanomas are caused by ultraviolet radiation (UVR), predominately from sun exposure and indoor tanning beds. Keratinocyte cancers share UVR exposure as a major risk factor. Outside of the US, skin cancer is most prevalent in regions with large fair-skinned populations such as Europe, North America, and Australia. Between the years of 1953 and 2008, numerous studies have reported that skin cancer rates stabilized or decreased for populations in many of these regions.3 Despite the overall decline, as of 2018, the highest rates of keratinocyte cancers worldwide have been found in New Zealand, Australia, and North America.4 Skin cancer screening involves the process of having either the whole or partial sections of the skin visually examined for any changes in appearance to detect suspicious lesions for potential biopsy. This can be conducted by a trained health care professional, such as a dermatologist or primary care physician, or conducted by the patient, as a skin self-examination. Skin self-examination is arguably an important strategy for screening, given that most melanomas are detected by patients and their family members.5 An easy-to- remember rubric has been developed to guide patients in remembering the factors that may prompt further skin self-examinations by the general public.6 Such visual inspection would seem to be an intuitive strategy to increase earlier detection of malignancies of the skin. Indeed, if caught early, all forms of skin cancer are curable. However, at the current time, there is no national recommendation for skin cancer screening in the US7 and thus physicians and patients are often confronted with the decision about whether to conduct or recommend such screenings in the absence of firm guidance. In this chapter, we will first review the current evidence and recommendations for skin cancer screening in the US. Second, we will review the current rates of screening, both overall and in higher-risk subgroups. Third, we will review the recent evidence for the use of interventions to enhance skin cancer screening. Fourth, we will present some innovative use of new technology to increase skin cancer screening. Finally, we will present future directions for this research. Current Evidence and Recommendations In 2016, the United States Preventive Services Task Force (USPSTF), which makes evidence-based recommendations about clinical preventive services, released a statement that the current evidence available did not warrant recommendation for the use of skin cancer screening.7 The report states, “At present, there is insufficient evidence for any population that regular visual skin examination by a clinician can reduce skin cancer–related morbidity and mortality.” The USPSTF was guided by the lack of evidence from a randomized controlled trial confirming the benefit of screening in reducing morbidity and mortality related to skin cancer, as well as the potential risks that could be associated with routine skin cancer screening in the general population. These risks were specified as the anticipated increase in detection and treatment of skin cancers, such as basal cell cancers, that do not impact life expectancy; the high number of biopsies and excisions that may be generated by screening to treat small numbers of skin cancers and melanoma; and the complications and potential poor cosmetic outcomes that can result from such excisions. For instance, Wernli and colleagues argued from the USPSTF perspective that, although rates of skin cancer have increased since 1986, mortality rates have remained stable, indicating the potential that increased screening may be leading to the finding of clinically less significant cases of skin cancer instead of more harmful invasive tumors.8 Numerous medical and public health organizations have followed suit with the USPSTF by not issuing official guidelines, yet have tried to provide some guidance for the general public with information and resources on how to advocate for themselves regarding follow-up of any noticed changes with their skin, particularly among 88 SECTION II Cancer Screening in Normal and At-Risk subpopulations with skin cancer risk factors. Skin cancer risk factors can include light skin, hair, and eyes; multiple moles; and a family or personal history of skin cancer. Current recommendations and resources for the general public from major medical and public health organizations can be found in Table 12.1. The general theme across organizations is that while there is no consensus on standard guidelines for the public, those who are at higher risk for skin cancer are encouraged to consult with their clinicians regarding personalized treatment plans for physician and self-screening. For instance, the American Academy of Dermatology recommends that those with a history of melanoma should have a full body exam at least once annually in addition to regular self-screenings,9 and the Skin Cancer Foundation recommends a full body exam more often than annually if at higher risk.10 Similar to the US, there is currently no formal population-based screening program in New Zealand, Australia, or the UK. However, Cancer Council Australia recommends that those at higher risk for developing skin cancer, such as those with fair skin, a tendency to burn or tan easily, light eyes and hair color, increased number of moles, or a family and/or personal history of skin cancer, undergo physician skin examinations every 6 months, and to conduct self-examinations regularly.11 Additionally, the Cancer Society of New Zealand has recommended that those at higher risk for the onset of skin cancer develop evidence-based management strategies for early detection with their clinicians.12 Guidelines for screening by the British Association of Dermatologists are that those at moderately increased risk, defined as patients with a previous history of melanoma or a large number of moles, some of which may be atypical, should be referred to a specialist and counseled on how to conduct a self-examination; those at a “greatly increased risk of melanoma,” including those with “giant congenital” nevi, should be monitored for life by a specialist; and those with a family history of melanoma or pancreatic cancer in three or more members should be referred to a specialist.13 A seminal observational study of systematic skin cancer screening was recently mounted in Germany, which highlights some of the challenges inherent in interpreting nonrandomized studies. This important example of the widespread implementation of population-based, one-time screening was conducted in the region of Schleswig-Holstein, Germany, in the north of Germany, starting in 2008, where a 1-year skin cancer screening program was integrated into routine primary care. This SCREEN program (Skin Cancer Research to Provide Evidence for Effectiveness of Screening in Northern Germany) was initiated by the Association of Dermatological Prevention in 2003 to establish the efficacy of population-based screening. Initial promising evidence found that offering community screening over 5 years saw an increase in incidence rates of skin cancer diagnoses and, importantly, a 48% decrease in melanoma mortality, yet subsequent analyses found that over time the initial decrease in mortality was transient and returned to levels observed before screening initiation within the next 5 years. Given that this study was an observational study and not a randomized controlled trial, there are many factors that may have accounted for these findings, the most important of which were differences in physician reporting of cause for death in this area.14 Since these results have come out, other groups around the world have continued to deliberate as to whether a well-designed randomized controlled trial could confirm whether the value of skin cancer screening is possible or worthwhile. For instance, a group in Norway concluded that given the risks to overdiagnosis and treatment, the rarity of melanoma detracts from mounting a trial. In contrast, Cust and colleagues15 responded that the number needed to invite for screening to prevent one melanoma death is inversely proportional to the mortality rate in the cohort, making it potentially worthwhile to conduct such a trial in a part of the world with very high melanoma incidence. Further, these authors note that the integration of new technologies such as total body photography, dermoscopy, and artificial intelligence would not make a trial obsolete, but could be incorporated into the trial design and lead to improvements in sensitivity and specificity, increasing cost-effectiveness. A multidisciplinary US group including expertise from dermatology; dermatopathology; cutaneous, surgical, and medical oncology; clinical research; epidemiology; and social psychology have recently responded to the USPSTF 2016 recommendations with a set of well-justified skin cancer screening recommendations. These risk-based, data-driven guidelines for skin cancer screening are modeled based on USPSTF guidelines for other cancers.16 These guidelines highlight the remarkable safety, ease, and low-cost nature of total body skin examination (TBSE), compared to screening for other cancers such as breast and colorectal cancer, which is defined as evaluation of the entire skin surface, including scalp, face, ears, neck, chest, abdomen, back, buttocks, genitals, upper and lower extremities, hands, feet, eyes (iris and sclera), oral mucosa, hair, and Table 12.1. Guidelines for Early Detection of Skin Cancer for General Public from Public Health Organizations in the United States Organization Official Guidelines for General Public Resources for General Public 1 Division of Cancer Prevention and Control, Centers for Disease Control and Prevention (CDC)37 No official guidelines for general public Provide information through their website and recommend the public report any noted changes in the skin to a clinician for further review 2 United States Preventive Services Task Force (USPSTF)38 No official guidelines for general public Provides facts about skin cancer and recommendations to speak with a clinician38 3 American Academy of Dermatology (AAD)9 No official guidelines for general public Encourages all people to conduct self-examinations regularly and provides services through their SPOTme program39 4 American Cancer Society (ACS)40 No official guidelines for general public Provides extensive details and information on how to conduct skin examinations through their website and prevention report 5 National Council on Skin Cancer Prevention (NCSCP)41 Self-examination once a month Provides prevention behavioral recommendations for public 6 Skin Cancer Foundation10 Yearly screening with physician; monthly self-exam Provides information regarding self-exam, tips for what to do for the yearly screening, and resources to find a physician CHAPTER 12 Skin Cancer Screening nails. Based on international recommendations from countries with similar levels of elevated risk, these authors recommend a target age range of 35 to 75 years for at least annual TBSE, and targeting risk groups such as those with a personal or family history of melanoma or a hereditary predisposition to melanoma based on a developing number of melanoma susceptibility genes; phenotypic factors indicating higher risk, such as blonde or red hair; or a history of overexposure to ultraviolet radiation, such as a history of blistering or peeling sunburns. They also note that detection of keratinocyte cancers cannot be considered only a harm of screening, as it adds to the costs and workload, but also a benefit of screening, given that important elements of quality of life are gained from their improved diagnosis and early excision. As such, these authors strongly argue that risk-based skin cancer screening is justifiable and warranted and could reduce morbidity and mortality associated with melanoma. Current Rates of Screening in General Population and High-Risk Subgroups Completion of skin cancer screening is low in the US, with about 1 in 5 adults reporting having ever received a TBSE from a clinician. Multiple national probability surveys have included assessment of skin cancer screening that have allowed for estimates of prevalence of skin cancer screening in large population subgroups. In general, results show an increased prevalence of screening over time, with higher rates among those with skin cancer risk factors. The National Health Interview Survey (NHIS), a nationally representative survey sponsored by the Centers for Disease Control and Prevention’s (CDC) National Center for Health Statistics, shows that as of 2015, about 21% of US adults reported ever having had a TBSE to check for cancer by a dermatologist or other physician (N = 33,672), and about 45% of those had one within the past year. Of these, two- thirds (66%) reported having a skin check as part of a routine exam, while 27% had one because of a skin problem.17 We are not aware of studies specifying population-level rates of skin self-examination, but reviews examining rates of skin self-examination across smaller studies range extremely widely.18 Fortunately, screening rates are higher among those with skin cancer risk factors. A study incorporating NHIS data from 201019 indicated that rates of screening among high-risk individuals differ modestly from those classified as medium-risk or low-risk groups. Among those in the high-risk group, including those who identify as non-Hispanic, White, or older than 65 years old and with a history of sunburns and a family history of skin cancer, it is estimated that 24% had ever received at least one TBSE, and about 11% had done so in the past year. These lower rates of recent screening in those at higher risk may indicate that they may have adopted screening over time, rather than more recently, as might be the case with those in the general population. Among those in the medium-risk group, rates of screening are comparable. This group is defined as the remainder of the White non-Hispanic population, where 24% are estimated to have ever had a TBSE, and 10% within the past year. Indeed, for those who engage in indoor tanning, screening may be more important than for those who do not due to their increased risk of developing skin cancer. In a study utilizing 2015 NHIS data, it was found that about 30% of those who have ever indoor tanned report ever having a TBSE by a physician; further, indoor tanners had been screened for skin cancer at younger ages than those who had never tanned.20 The highest rates of screening are likely concentrated in the melanoma survivorship population. As such, a small 2014 study analyzing health behaviors of melanoma survivors within a US cancer center from 1995 to 2011 found that 94% of melanoma survivors reported routinely receiving clinical skin examinations since their diagnoses.21 In addition to clinical examinations, studies have shown that about half of recurrent melanomas have been identified first by the patient or loved one before being officially diagnosed by a health care provider, indirectly indicating enhanced self-screening in the survivorship population.5 Internationally, a study examining the role of various factors in determining both skin cancer self-examinations and clinical examinations between 2007 and 2008 (N = 8,178) in Australia, the US, and Europe found that those with a history of melanoma had much higher rates of both types of screening compared to those without a history of melanoma; further, rates of both types of screening were higher among those living in Australia and the US compared to Europe.22 Interventions to Improve Skin Cancer Screening Interventions to improve skin cancer screening have targeted medical providers, including physicians and medical students, with the aim to improve rates of completion of screening among medical professionals such as primary care physicians who see patients regularly and have a focus on preventive care. Some of these efforts have taken an opportunistic approach whereby physicians are trained to distinguish benign versus suspicious lesions on areas of the body that are exposed during their typical primary care examination, such as the chest and back, that would reduce the need for a special visit for clinical skin examination. Interventions have also been developed to encourage patients to conduct regular skin self-examinations. In the following section, we present some recent examples of this research. Many of the efforts targeted to medical providers have focused on nondermatologically trained clinicians. Such efforts have included training for physicians to identify benign versus suspicious lesions, to conduct clinical skin examinations, and to use novel strategies to aid in detection such as dermatoscopes. One promising example includes the INFORMED (Internet Curriculum for Melanoma Early Detection) program, which was developed to provide a web-based early detection training program that includes a large image database for imaging. This program involves 1–2 hours of training that can be delivered via traditional textbook format or case-based format, which includes nine case vignettes illustrating teaching points with quizzes and feedback. The course improved primary care physicians’ ability to manage skin lesions without increasing dermatologist referrals, and improvement was still evident at 6 months.23 Of note, INFORMED was focused largely on improving physicians’ ability to diagnose and manage skin lesions correctly rather than on general knowledge about skin cancer risks per se, so that they may not necessarily be better prepared to offer skin cancer prevention advice to patients. Further work with INFORMED moved this training to a large, observational, year-long study in a large health care system at the University of Pittsburgh and a relatively higher-risk sample of older individuals than previous skin cancer screening programs have targeted. Of note, physicians did not receive compensation for 89 90 SECTION II Cancer Screening in Normal and At-Risk conducting the screenings, increasing the generalizability of the study findings. This study focused on increasing use of full-body skin examination (FBSE) by primary care physicians and included promotion of the program to physicians as well as patients, and finally adjustment of the electronic medical record to include FBSE as a recommended preventive service for individuals over age 35. This large study included over 300,000 patients seen in an office visit by a primary care physician in 2014; about 53,000 patients were included in the screened cohort. Findings indicated a higher likelihood of melanoma diagnoses in those who were screened compared to those who were unscreened; melanomas in screened patients were thinner than those in unscreened patients. The incidence of thicker melanomas (1 mm or thicker) was the same in screened and unscreened groups, however. Importantly, these findings are observational, not the result of a randomized trial, so there may be confounding factors that may have influenced screening as well as data quality. Medical students have also been a focus of training to encourage skin cancer examination, including, for example, the development of a film, The Integrated Skin Exam, to increase medical students’ knowledge, identification of higher-risk groups, the characteristics of suspicious nevi, and intentions to conduct skin examinations, which showed promising effects in second-year medical students recruited across the US.24 The film is available through the American Academy of Dermatology Medical Student Core Curriculum. Another important effort to enhance skin cancer screening in primary care providers has focused on opportunistic screening, or the ability of physicians to identify suspicious lesions when they are conducting standard primary care examinations. As such, the focus here is not on thorough or comprehensive examinations per se. These authors conducted a randomized trial (N = 89 primary care physicians) to examine a mastery learning course that included training in use of dermoscopy, which is a noninvasive in vivo technique commonly used by dermatologists to visualize lesions, as well as deliberate practice and feedback to reach a standard. In the intervention group, fewer benign lesions were referred for additional follow-up within 3 months of the training and physicians referred significantly more melanomas in that time period. Most of the melanomas were located on the head and neck, which is consistent with the opportunistic, rather than comprehensive, nature of these examinations.25 Importantly, a reduction in referrals for benign lesions is quite important given that this can potentially reduce health care costs, decrease patient anxiety, and reduce office visit burden. Additional efforts have been mounted to encourage skin self- examination, both in the general population and in community settings, and also among patients at higher risk of developing skin cancers. One important and successful effort to increase skin cancer screening rates in primary care patients included the Check-It- Out intervention that promoted thorough skin self-examination. Importantly, physicians did not participate in the intervention and in fact were blinded to the group assignment.26 The study randomized primary care patients to intervention or control condition (dietary improvement). Results found that thorough skin self-examination increased in the intervention group at 2, 6, and 12 months, and although skin surgeries increased initially, there were no differences in surgery rates at 6 and 12 months between intervention and control groups.26 Additional analyses looking at the most important components of the intervention arm found that the video that was developed for the study, the hand mirror, a brochure developed by the American Cancer Society, and sample photos led to higher rates of skin self-examination. Importantly, the clinicians who agreed to have their patients participate in the Check-It-Out study may be more prevention focused than the general community of primary care physicians. An important community-wide effort to improve skin self-examination, the SkinWatch study, included skin self-examination self-help guides, skin cancer seminars, skin cancer education to local medical practitioners, and free, open-access skin screenings in Queensland, Australia, and found promising effects on screening rates, which were attenuated after the open-access skin screenings were eliminated.27 There has been a focus, as well, on higher-risk patients. For example, patients who attend pigmented lesion clinics undergo enhanced dermatologist surveillance given their very high- risk phenotypes, but also can benefit from skin self-examinations conducted between office visits. Marek and colleagues conducted a trial examining the use of a smartphone mobile application (app) that included a set of total body photographs that were taken professionally in their dermatologists’ office to document skin lesions.28 A trial including 69 patients randomized them evenly across four arms: either use of the mobile app alone or use of the mobile app along with either skin exam reminders, sending monthly performance reports to an accountability partner identified by the patient such as a spouse, or use of both the reminders and accountability partner. Outcomes were evaluated at 6 months. In all arms, skin self-examination increased from 58% at baseline to 83% at 6-month follow-up; there were no differences by intervention group. The study was shown to be efficacious, and this seemed especially true for hard-to-examine areas such as the lower back and back of legs. Participants found the reminders to be especially useful. Other efforts have focused on encouraging skin self-examination in specific circumstances where patients may be especially receptive. For instance, Robinson and colleagues developed an interesting intervention targeting women undergoing mammography screening who reported melanoma risk factors such as a history of sunburn, indoor tanning, or personal or family history of melanoma. In the changing room while waiting for their mammogram, they were exposed to an informational poster and brochure promoting risk- targeted skin self-examination advice. The authors argue that mammography is a great way to target higher-risk patients, given that most melanomas occur in those over age 40, making this a feasible approach to getting women to focus on skin self-examination. Half the group also received a reminder to complete skin self-examination at 1 week. At 1 month, 80% of those who had completed the follow- up survey had performed skin self-examination,25 which was a great improvement over baseline skin self-examination assessment (30% reported ever having done a skin self-examination). Further, anxiety was not increased in the intervention as associated with skin self-examination. Other efforts have included combined interventions to encourage skin cancer prevention as well as early detection strategies, especially in higher-risk individuals. For example, Bowen and colleagues focused on increasing both physician screening and skin self-examination in family units as well as sun protection, where at least one family member had a history of melanoma, one was a first-degree family member of the patient, and one was a parent of a young child, with very promising indicators for both types of screening.29 The PennSCAPE trial recently examined the use of CHAPTER 12 Skin Cancer Screening mailed tailored print materials compared to generic print materials in primary care patients to encourage both sun protection and skin self-examination behaviors. Materials were tailored to patient risk level and found promising outcomes.30 In addition to promising increases in sun protection, the intervention also increased use of skin self-examination and skin examinations by a health care provider. Recent Use of Innovative Technology Innovation strategies to motivate screening, as well as ways to improve the accuracy of screening, have been evolving quickly. Strategies that serve as a cue to action, or enhance screening motivation, have been developed. For example, electronic reminders have been used to facilitate completion of skin cancer screening, as well as the use of digital interventions, using tablets and mobile phones to prompt and support total skin self-examinations. These online, interactive interventions can be targeted to family members who can aid patients in conducting skin examinations. Further, the provision of photographs of patients’ entire skin surface31 has also provided promising prompts to perform skin self-examination. More recently, the use of mobile teledermoscopy has been examined as a way for patients to conduct skin self-examination and then to use their mobile telephone to photograph and send in the photographs for dermatologist assessment; results have shown patient acceptance of this technology, yet with some important caveats regarding patient trust, and mixed findings on clinical accuracy of the photographs sent in by patients for examination,32 supporting the use of teledermoscopy as a complement rather than a replacement for clinical skin examination. Motivation to increase screening might be galvanized by personalized information, such as genetic information.33 Other strategies are focused more specifically on accuracy, or improving the sensitivity and specificity of the screening, which can reduce unnecessary biopsies and corresponding screening costs. Such emerging technology includes dermoscopy, confocal microscopy, and digital photographic imaging, including artificial intelligence,34 which have the potential to improve screening efforts among the wider population of health care providers as well as for patient- initiated examination. However, use of dermoscopy is relatively low among US nondermatologist physicians, but higher in those who graduated from medical school more recently and those who see more skin cancer patients.35 Barriers to use in primary care include the time and resources to adequately train nondermatologists. Of note, an innovative intervention has been developed for individuals who received radiation as children for their malignancies and who are at significantly increased risk for skin cancer. It uses patient- driven innovative health technology—teledermoscopy—whereby patients take photographs of concerning moles and then transfer them electronically for dermatological assessment; this was combined with web and print materials encouraging the patients to conduct skin self-examinations and request physician examinations.36 Future Directions There are a number of barriers that stand in the way of increasing skin cancer screening in the US and around the world that require work to surmount them. First, the lack of USPSTF recommendations limits physician reimbursement for screening and may detract from the devotion of time and effort for physicians to complete screening or encourage self-screening in their patient populations. As of 2019, Illinois has provided reimbursement for screening, which will present an important opportunity to examine effects of these changes in reimbursement over time. Important, well-justified efforts to justify more uniform and intensive screening for higher-risk groups may change physician practice over time. Second, preferences among patients for physicians and health care providers to conduct exams—rather than to learn and consistently practice skin self- examinations—may continue to limit skin self-examination uptake, even in higher-risk populations, and requires further study. Third, given limitations in the dermatology workforce in the US and other countries, there is a need for continued efforts to extend the capacity to conduct thorough screenings by primary care physicians, as well as physician assistants and nurses, which will be important as interest and demand for health care–provided screenings increases. Efforts to test the delivery of education to primary care providers over broad geographical areas via video technology may enhance knowledge acquisition and practice change in primary care. Finally, the use of new technology, such as artificial intelligence and other technology, may substantially improve diagnostic accuracy and reach across dermatologists and other physicians who may start to practice more regular screening in their higher-risk patients. Overall, while current rates of screening are low in the general population, risk-based screening seems justified enough to continue robust efforts to train a broad range of physicians and health care providers to conduct skin cancer screening and to encourage skin self-examination. Given the associated morbidity and dramatically increasing cost to treat advanced melanomas, such efforts are strongly warranted from both a patient well-being and a cost perspective. ACKNOWLEDGMENTS We acknowledge with thanks the technical assistance of Liliane Sar-Graycar and Teddy Smith in the completion of this chapter. REFERENCES 1. American Cancer Society. Cancer Facts & Figures 2019. Atlanta, Georgia: American Cancer Society; 2019. 2. National Cancer Institute. Cancer stat facts: melanoma of the skin. https://seer.cancer.gov/statfacts/html/melan.html. 3. Erdmann F, Lortet-Tieulent J, Schüz J, et al. International trends in the incidence of malignant melanoma 1953–2008—are recent generations at higher or lower risk? Int J Cancer. 2013;132(2):385–400. 4. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68(6):394–424. 5. Aviles- Izquierdo JA, Molina- Lopez I, Rodriguez- Lomba E, Marquez-Rodas I, Suarez-Fernandez R, Lazaro-Ochaita P. Who detects melanoma? Impact of detection patterns on characteristics and prognosis of patients with melanoma. J Am Acad Dermatol. 2016;75(5):967–974. 6. Tsao H, Olazagasti JM, Cordoro KM, et al. Early detection of melanoma: reviewing the ABCDEs. J Am Acad Dermatol. 2015;72(4):717–723. 91 92 SECTION II Cancer Screening in Normal and At-Risk 7. U.S. Preventive Services Task Force. Screening for skin cancer: US Preventive Services Task Force recommendation statement. JAMA. 2016;316(4):429–435. 8. Wernli KJ, Henrikson NB, Morrison CC, Nguyen M, Pocobelli G, Blasi PR. Screening for skin cancer in adults: Updated evidence report and systematic review for the US Preventive Services Task Force. JAMA. 2016;316(4):436–447. 9. American Academy of Dermatology. Stats in skin cancer. 2019; https://www.aad.org/media/stats-skin-cancer. Accessed November 1, 2019. 10. Skin Cancer Foundation. Early detection: overview. 2019; https:// skincancer.org/early-detection/. 11. Cancer Council Australia. Position statement-screening and early detection of skin cancer. 2019; https://wiki.cancer.org.au/policy/ Position_statement_-_Screening_and_early_detection_of_skin_ cancer. 12. Cancer Society of New Zealand. Position statement on screening and early detection of skin cancer. 2010; https:// c ancernz.org.nz/ a ssets/ P ositions- S tatements/ P S- ScreeningandEarlyDetectionofSkinCancer-Jul2013.pdf. 13. Marsden JR, Newton-Bishop JA, Burrows L, et al. Revised UK guidelines for the management of cutaneous melanoma 2010. J Plast Reconstr Aes Surg JPRAS. 2010;63(9):1401–1419. 14. Boniol M, Autier P, Gandini S. Melanoma mortality following skin cancer screening in Germany. BMJ Open. 2015;5(9):e008158. 15. Cust AE, Aitken JF, Baade PD, Whiteman DC, Soyer HP, Janda M. Why a randomized melanoma screening trial may be a good idea. Br J Dermatol. 2018;179(5):1227–1228. 16. Johnson MM, Leachman SA, Aspinwall LG, et al. Skin cancer screening: recommendations for data-driven screening guidelines and a review of the US Preventive Services Task Force controversy. Melanoma Manage. 2017;4(1):13–37. 17. National Center for Health Statistics. 2015 National Health Interview Survey. Atlanta, GA: Centers for Disease Control and Prevention; 2015. 18. Kasparian NA, McLoone JK, Meiser B. Skin cancer-related prevention and screening behaviors: a review of the literature. J Behav Med. 2009;32(5):406–428. 19. Lakhani NA, Saraiya M, Thompson TD, King SC, Guy GP Jr. Total body skin examination for skin cancer screening among U.S. adults from 2000 to 2010. Prevent Med. 2014;61:75–80. 20. Heckman CJ, Handorf E, Auerbach MV. Prevalence and correlates of skin cancer screening among indoor tanners and nontanners. JAMA Dermatol. 2018;154(5):554–560. 21. Palesh O, Aldridge- Gerry A, Bugos K, et al. Health behaviors and needs of melanoma survivors. Support Care Cancer. 2014;22(11):2973–2980. 22. Kasparian NA, Branstrom R, Chang YM, et al. Skin examination behavior: the role of melanoma history, skin type, psychosocial factors, and region of residence in determining clinical and self-conducted skin examination. Arch Dermatol. 2012;148(10):1142–1151. 23. Eide MJ, Asgari MM, Fletcher SW, et al. Effects on skills and practice from a web-based skin cancer course for primary care providers. J Am Board Family Med JABFM. 2013;26(6):648–657. 24. Garg A, Wang J, Reddy SB, et al. The Integrated Skin Exam film: an educational intervention to promote early detection of melanoma by medical students. J Am Acad Dermatol. 2014;70(1):115–119. 25. Robinson JK, Perez M, Abou-el-Seoud D, et al. Targeted melanoma screening: risk self-assessment and skin self-examination education delivered during mammography of women. JNCI Cancer Spectrum. 2019;3(3):pkz047, https://doi.org/10.1093/ jncics/pkz047 26. Weinstock MA, Risica PM, Martin RA, et al. Melanoma early detection with thorough skin self-examination: the “Check It Out” randomized trial. Am J Prevent Med. 2007;32(6):517–524. 27. Aitken JF, Youl PH, Janda M, Lowe JB, Ring IT, Elwood M. Increase in skin cancer screening during a community-based randomized intervention trial. Int J Cancer. 2006;118(4):1010–1016. 28. Marek AJ, Chu EY, Ming ME, Khan ZA, Kovarik CL. Piloting the use of smartphones, reminders, and accountability partners to promote skin self-examinations in patients with total body photography: a randomized controlled trial. Am J Clin Dermatol. 2018;19(5):779–785. 29. Bowen DJ, Hay J, Meischke H, Mayer JA, Harris-Wai J, Burke W. Randomized trial of a web-based survivor intervention on melanoma prevention behaviors of first-degree relatives. Cancer Causes Control: CCC. 2019;30(3):225–233. 30. Glanz K, Volpicelli K, Jepson C, Ming ME, Schuchter LM, Armstrong K. Effects of tailored risk communications for skin cancer prevention and detection: the PennSCAPE randomized trial. Cancer Epidemiol Biomarkers Prevent. 2015;24(2):415–421. 31. Oliveria SA, Chau D, Christos PJ, Charles CA, Mushlin AI, Halpern AC. Diagnostic accuracy of patients in performing skin self-examination and the impact of photography. Arch Dermatol. 2004;140(1):57–62. 32. Manahan MN, Soyer HP, Loescher LJ, et al. A pilot trial of mobile, patient- performed teledermoscopy. Br J Dermatol. 2015;172(4):1072–1080. 33. Kanetsky PA, Hay JL. Marshaling the translational potential of MC1R for precision risk assessment of melanoma. Cancer Prevent Res (Phila, Pa). 2018;11(3):121–124. 34. Esteva A, Kuprel B, Novoa RA, et al. Dermatologist-level classification of skin cancer with deep neural networks. Nature. 2017;542(7639):115–118. 35. Fee JA, McGrady FP, Rosendahl C, Hart ND. Dermoscopy use in primary care: a scoping review. Dermatol Pract Concept. 2019;9(2):98–104. 36. Daniel CL, Armstrong GT, Keske RR, et al. Advancing Survivors’ Knowledge (ASK) about skin cancer study: study protocol for a randomized controlled trial. Trials. 2015;16:109. 37. Division of Cancer Prevention and Control, Centers for Disease Control and Prevention. What screening tests are there? 2019; https://www.cdc.gov/cancer/skin/basic_info/screening.htm. 38. U.S. Preventive Services Task Force. Screening for Skin Cancer: Consumer Guide. 2016. 39. Okhovat JP, Beaulieu D, Tsao H, et al. The first 30 years of the American Academy of Dermatology skin cancer screening program: 1985-2014. J Am Acad Dermatol. 2018;79(5):884–891. e883. 40. American Cancer Society. Cancer Prevention & Early Detection Facts & Figures 2019–2020. 2019. 41. National Council on Skin Cancer Prevention. Skin cancer prevention tips. 2016; https://skincancerprevention.org/learning/. Accessed November 1, 2019. SECTION III Screening and Testing for Germ Line and Somatic Mutations Paul B. Jacobsen (Section Editor) 13 Psychosocial Issues in Genetic Testing for Breast/ Ovarian Cancer 95 Mary Jane Esplen, Jonathan Hunter, and Eveline M. A. Bleiker 14 Psychosocial Issues in Genetic Testing for Hereditary Colorectal Cancer 102 Sukh Makhnoon and Susan K. Peterson 15 Psychosocial Issues in Genomic Testing, Including Genomic Testing for Targeted Therapies 110 Megan Best 16 Psychosocial Issues Related to Liquid Biopsy for ctDNA in Individuals at Normal and Elevated Risk 116 Jada G. Hamilton, Amanda Watsula-Morley, and Alicia Latham 13 Psychosocial Issues in Genetic Testing for Breast/Ovarian Cancer Mary Jane Esplen, Jonathan Hunter, and Eveline M. A. Bleiker Background Approximately 10% of all breast cancers are due to hereditary factors, with the majority caused by mutations in two autosomal dominant breast cancer genes, BRCA1 and BRCA2. Therefore, identifying mutation carriers means that genetic counseling and testing of individuals, with or without cancer, can occur to inform health-related decision-making. For instance, mutations in these genes confer cumulative risks of breast cancer of 72% in BRCA1 mutation carriers and 69% in BRCA2 mutation carriers by age 80.1 Women with BRCA1/2 mutations who develop breast cancer also have a 20-year cumulative incidence of contralateral breast cancer estimated at 26% to 40%. In addition, BRCA1/2 mutations place women at risk for ovarian cancer with a cumulative risk of 44% by age 80 in BRCA1 carriers and 17% in BRCA2 carriers.1 An increased risk for prostate and pancreas cancer has also been reported.1 This knowledge of being at high risk of developing cancer offers opportunities for prevention. However, a number of psychological and social challenges have been identified with cancer genetic counseling; these will be discussed in this chapter. Outline of Genetic Counseling Individuals with characteristics suggestive of a hereditary cancer gene may be referred to specialty genetic clinics to see a genetic counselor and/or geneticist. These characteristics include having multiple cases of cancer in more than one generation, a young age of onset, bilateral breast cancer, breast and ovarian cancer, and triple- negative breast tumor at a young age. Genetic counseling aims to help counselees comprehend and adapt to the medical, psychological, and familial implications of having genetic contribution to disease. In BRCA counseling, individuals are educated about their breast and ovarian cancer risk, including risk of prostate cancer for men. This usually takes place across two consultations, at least one of which is performed before a genetic test, typically face to face with a trained genetics professional. More recently, to meet growing demand, alternative models of genetic service delivery using telephone or group counseling are underway.2 Outcomes of Genetic Testing for Breast Cancer The genetic test may result in a number of different outcomes: (1) a counselee is determined to have a pathogenic BRCA1 or BRCA2 mutation known in his or her family, defined as being a “carrier”; (2) a counselee is not a carrier of a pathogenic BRCA1 or BRCA2 mutation, a “noncarrier”; (3) no known BRCA1 or BRCA2 mutation is detected in the family or the counselee, despite a strong family history of breast/ ovarian cancer, an “inconclusive” result; or (4) an unclassified variant/ variant of uncertain clinical significance (UV) is found, so contribution of this BRCA1 or BRCA2 variant to cancer risk is “undefined.” As a result of this genetic screening, individuals can make individualized decisions. Proven carriers of a BRCA1/2 mutation have a substantially increased risk of breast or ovarian cancer, so periodic surveillance (mammography and/or breast magnetic resonance imaging) or preventive mastectomy and/or oophorectomy are recommended.3 Women who are proven to be noncarriers have no increased risk and are advised to participate in population-based breast cancer screening. For those individuals receiving an inconclusive test result, evidence- based guidelines are lacking; however, routine screening options are recommended and preventive surgery may also be considered. Treatment-Focused Genetic Testing Previously, breast cancer patients at increased genetic risk were offered genetic counseling and testing following completion of oncology treatment. However, a new model of “rapid” genetic testing in which testing happens before their primary surgery is now available.4 This process allows women to receive information on their carrier status close to their breast cancer diagnosis so that they can incorporate this knowledge into treatment decisions. These women may, for example, opt for mastectomy instead of lumpectomy, or for an immediate contralateral prophylactic mastectomy to prevent breast cancer in the unaffected breast. 96 Section III Screening and Testing for Germ Line and Somatic Mutations Initially, concerns were raised about the possibility that providing genetic testing at the time of diagnosis could result in both an informational and emotional overload at a stressful time. However, a recent study showed that, despite the additional distress burden, 73% percent of patients who received the results of genetic testing before surgery recommended genetic testing to other high-risk breast cancer patients, and 88% believed that the best timing of genetic testing was between breast cancer diagnosis and surgery.4 A recent randomized trial of rapid DNA testing before surgery among newly diagnosed breast cancer patients also showed no significant differences between study groups in psychosocial outcomes.4 The Possible Burden of Intense Surveillance and Prophylactic Surgery Following confirmation of a BRCA mutation, women are faced with difficult choices around how best to manage their risk of developing breast or ovarian cancer. Options to consider are enhanced surveillance with magnetic resonance imaging (MRI), mammography, transvaginal ultrasound scan of ovaries and fallopian tubes, risk-reducing surgery to their breast tissue, removal of their ovaries and/or fallopian tubes, chemoprevention, and lifestyle interventions. While prophylactic surgeries offer the best chance for reduced risk, such surgeries impact quality of life, body image, and sexuality. We will review each option with respect to its psychological consequences and benefits. Breast Cancer Surveillance Carriers of a BRCA1/2 mutation, as well as women who receive an inconclusive genetic test result, are all offered an intensive surveillance program, including mammography, MRI, and a clinical breast examination by a physician. In two reviews of women with a family history of breast cancer or with BRCA1/2 mutations undergoing intensive surveillance, normal levels of distress and better general health compared to the general population were found.5 However, if recall tests or false positives for a mammographic finding occur, anxiety inevitably elevates.5 Prophylactic Mastectomy By having a risk-reducing mastectomy, a woman with BRCA1/2 lowers her risk by about 90%. Furthermore, female breast cancer patients who carry a BRCA1/2 mutation have an increased risk of developing a second primary breast cancer.6 Contralateral prophylactic mastectomy (CPM) has been reported to lead to a reduction of up to 95% in the risk of contralateral breast cancer. However, there is still some debate on the effect on both overall and breast cancer– specific survival.7 Studies on the long-term satisfaction and psychosocial impact of risk-reducing surgery in breast cancer patients, or women at increased risk with a positive family history, report both positive and negative consequences. There is an overall high level of satisfaction, favorable effects on emotional stability and stress,8 and lower level of breast cancer concerns compared to women who have not had prophylactic mastectomy.9 Negative effects include a compromised body appearance and a reduced sense of femininity.8,9 In newly diagnosed breast cancer patients who have contralateral prophylactic mastectomy, no differences have been found in the first year after surgery in quality of life and distress, as compared to women who opt for breast conservation or unilateral mastectomy.10 Despite the potential negative impact of contralateral mastectomy, such as an adverse impact on body appearance, sense of femininity, sexual relationships, and unanticipated reoperations due to complications with reconstruction, the large majority of women report being satisfied with their decision to undergo prophylactic contralateral mastectomy.11 Recent research suggests that particular psychosocial factors are associated with pursuing prophylactic surgery. The age of the woman, her status as a parent, the gender of her children, and guilt about passing on a mutation to a child can all play a role.8,12 Psychological factors related to the BRCA experience, such as distress prior to surgery or uncertainty about familial support and medical options, also contribute to the decision to have prophylactic surgery. One study found that the more distress a woman experiences related to testing positive for BRCA, the less likely she is to have surgery and to continue with surveillance.12 Individuals who report practitioner-initiated discussions about undergoing bilateral risk-reducing mastectomy may be more prone to having regret. An approach focused exclusively on statistical risk reduction, without taking into account potential consequences of surgery and opportunity for reflection on other management options, may result in increased psychological issues postoperatively. Exploring motivational factors and assessing psychosocial functioning are critical. It has been recommended that women considering prophylactic mastectomy be referred to a psychosocial clinician to discuss options and to ensure that the woman is making an informed decision not driven by fear or based on perceived bias of one health professional—or other individuals, such as anecdotal advice from well-meaning friends or family.13 Studies of partners indicate that they are focused most on supporting the preservation of overall health of their partner with BRCA1/2;14 however, they too can experience difficulties related to the woman’s altered body image and sexual functioning, and should be offered psychosocial support.14 Ovarian Cancer Surveillance and Risk-Reducing Salpingo-Oophorectomy Until recently, ovarian cancer screening with a CA-125 test every 4 months and annual ultrasound was recommended as one of the “preventative” options for women at increased risk. Although final results of large trials are yet to be reported, initial studies suggest that gynecological screening has not led to decreased ovarian cancer mortality.15 Therefore, screening for ovarian cancer in asymptomatic women is no longer a standard recommendation. However, for women with a BRCA1/2 mutation, risk-reducing bilateral salpingo-oophorectomy is recommended, as it is believed to reduce the risk of ovarian cancer by 80%–96%. Furthermore, salpingo-oophorectomy contributes to a reduced breast cancer risk by 50% when performed prior to menopause. Madalinska et al.16 found favorable effects of prophylactic oophorectomy in terms of reduced cancer worries and low perceived cancer risk. However, as the surgery results in the onset of menopause, the observed benefits need to be weighed against impacts on body image and an increase in endocrine and sexual symptoms. CHAPTER 13 Psychosocial Issues in Genetic Testing for Breast/Ovarian Cancer Psychosocial Issues Encountered by Individuals Undergoing Genetic Counseling and Testing While it is generally recognized that many individuals will benefit from genetic testing and counseling for breast cancer, genetic testing may induce serious psychosocial problems. Testing Positive for a Genetic Mutation Reviews and a meta-analysis indicate that the majority of counselees informed of carrier status do not exhibit heightened or clinically relevant levels of depression, anxiety, and/or distress, at least beyond the short term, as assessed by standardized questionnaires with established score thresholds for clinical relevance.17,18 However, depending on the type and timing of the assessment, a subset of approximately 25% of counselees demonstrate high levels of psychological morbidity, sufficient to merit interventions.18 It has become increasingly clear that the historical context of the individual coming forward for testing, such as that related to the prior experience of cancer in the family, cancer-related deaths, the genetic test result of a sibling, and having children or not, play an increasing role in understanding who may require added psychological support. Risk Factors for Increased Psychosocial Distress A listing of risk factors for increased emotional distress appears in Box 13.1. A robust predictor of post–genetic testing distress is the pretest emotional state or distress level.17,18 Among the risk factors for high distress is the experience of parental cancer in childhood.19 Specifically, women who have lost their mothers to cancer report more cancer-specific distress.20,21 Similarly, individuals who have cared for and/or lost a parent during adolescence are more vulnerable to psychological distress and may request additional psychosocial support during the genetic counseling process.22,23 Related risk factors for distress are the recent loss of a close relative to cancer, the recent diagnosis of cancer in the counselee,18,22 and problems encountered communicating with family members.24 Recognition and support of the counselee who is the “family messenger,” typically the first utilizer of predictive testing and preventive surgery in the family, are important—especially when they feel that they must provide a “good example” for the rest of the family. Furthermore, special caution is needed in those families in which clear expectations exist about carrier status. If the test outcome differs from what is anticipated, families often have more difficulty adapting.22,23 Finally, psychosocial support is frequently needed by those who are confronted with the decision to undergo risk-reducing surgery. Pre-as well as postsurgery support can help individuals in making a well-balanced decision about undergoing surgery and facilitate the adaptation to the operation.25 Identification of Psychosocial Problems Specific brief screening instruments have been developed to identify increased psychosocial risk at the beginning of genetic counseling. The Genetic Psychosocial Risk Instrument (GPRI) identifies individuals at risk for psychological distress.22 Another brief instrument, the Psychosocial Aspects of Hereditary Cancer (PAHC) questionnaire, detects individuals experiencing current psychosocial problems.23 Additional instruments assess psychological response after genetic testing. Examples include the Psychological Adaptation to Genetic Information Scale (PAGIS)26 and the Multidimensional Impact of Cancer Risk Assessment (MICRA).27 Receiving a Negative Test Result Some individuals will receive the good news that they do not carry a genetic mutation. This is typically associated with relief and reductions in both general and breast cancer–specific anxiety. Surprisingly, however, testing negatively can cause struggles adjusting to a new sense of personal risk. Frequently, individuals with a strong family history of cancer have integrated a sense of being at increased risk to cancer and feel a strong identification with the relative(s) affected by the disease. A negative test is therefore unexpected, and individuals have the task of integrating this incongruent information into their sense of self. This task is complicated by the feeling that one is giving up being a member of a group, which can be felt as a distressing separation from loved ones. Other feelings that can emerge for those Box 13.1. Factors Associated with Psychological Risk or Adjustment Difficulties Sociodemographic ♦ Younger age ♦ Female sex ♦ Being unmarried ♦ Lower socioeconomic status ♦ Having children Cancer and symptom related ♦ Recent diagnosis ♦ Recent history of breast symptoms ♦ Past experience of cancer and its impact on the family Psychosocial ♦ Increased risk perception ♦ Appraisal of high relevance and threat ♦ Loss of close relative to cancer, especially loss of mother at young age ♦ Caregiving of family member with cancer ♦ Prior history of additional life losses/trauma ♦ Premorbid psychological condition ♦ Current level of psychological functioning (e.g., presence of depression, anxiety, disease-specific worry) ♦ Low self-esteem ♦ Feeling stigmatized by BRCA1/2 status ♦ Expectation of receiving a negative test result ♦ Coping style (e.g., avoidant coping, anxious preoccupied, health monitoring, using suppression as emotional regulation strategy, pessimistic) ♦ Low level of social support Family ♦ Being first in family tested ♦ Low level of family cohesion ♦ Knowledge of siblings’ positive result ♦ Anticipated or actual changes in relationships as a result of testing ♦ Anxious partner ♦ Frequent concern for children; guilt of passing on mutation 97 98 Section III Screening and Testing for Germ Line and Somatic Mutations testing negative include feelings of guilt about being unjustly spared the legacy of the familial disease, a form of “survivor guilt.”24 Inconclusive Test Results Even if an individual receives an “inconclusive” result, it is still possible for the individual to carry a gene mutation that is rare, or not yet determined to be significant. Such a result causes clinicians concern, as they worry that the individual could feel reassured but in fact be at some elevated risk. The individual’s comprehension of an inconclusive test result must be reviewed during follow-up, particularly in relation to adherence with surveillance and preventive options.28 There is some evidence that those who receive inconclusive results experience higher levels of distress compared to carriers and noncarriers,28 so follow-up of both comprehension of cancer risk and coping with uncertainty is necessary. Family Communication Issues Genetic testing results have implications for family members beyond the consultee. It can be difficult to notify family members who may have never been met or who do not live close by about a genetic test result, for fear of causing emotional upset.29 In addition, individuals with genetic knowledge may also feel pressure to encourage family members to be tested. When these individuals are not interested, conflict can follow. Women with a BRCA1/2 gene mutation and their partners can find it particularly challenging to inform their offspring of a mutation.30 Studies have shown that parents have a strong interest in notifying underage and adult offspring,31 although sex of the child can make a difference, with informing daughters of greater concern.32 Mothers can be conflicted in that they wish to ensure their adult daughters are enrolled in the appropriate surveillance programs and adopt healthy lifestyle behaviors, yet can simultaneously feel a need to not disclose the mutation to protect the child from psychological burden. Parents with younger children have an additional challenge of explaining absences or changes due to medical appointments or procedures and communicating complex genetic information, while striving to reinforce a current state of good health. Studies on offspring show that children demonstrate a range of responses to the initial disclosure and are more likely to recall favorable and/or neutral responses.33 Although some children have sufficient literacy around genetic information, others demonstrate misinformation about cancer risk, similar to that of adults.33 Adolescents and young adult children do demonstrate psychological hardiness around the potential threat and may focus on healthy ways to control cancer via lifestyle choices, seeing the positive side of the situation. A parent’s style and perception can play a pivotal role, as children often take behavioral cues from their parents.33 Overall, early studies show adjustment and resilience in the short term, but there can be adverse impacts, such as concern around cancer risk or challenges in coping with information. Less is known about the long-term impact of genetic knowledge on offspring. Family communication challenges can be helped by healthcare professionals. Decisional aids and guides offer a developmental framework to guide discussions in communication with offspring. They are being tested to help guide when and how to disclose, recognizing that most parents plan on having the discussion in the near future, regardless of the age of the child,31,34 Family-oriented interventions that guide the sharing of sensitive information are also helpful29 as how individuals perceive a relative’s opinion of genetic testing, or sense of control, can influence whether or not individuals share genetic information.29 As well, given that individuals with greater depression or inconclusive results may be less likely to disclose genetic test results,29 targeted psychosocial support to address these specific barriers are indicated. BRCA1 and BRCA2 Carriers and Assisted Reproduction Individuals carrying a BRCA1/2 mutation are frequently concerned about passing their predisposition to their children even to the point of foregoing childbearing.35 Healthcare professionals will be increasingly involved in discussions and decisional counseling regarding reproductive options in families with a known mutation. In recent decades, genetic testing for hereditary cancers before birth has become available through prenatal diagnosis (PND) and preimplantation genetic diagnosis (PGD). PND is performed during early pregnancy at 10–20 weeks. If the fetus is found to be a BRCA1/ 2 carrier, the pregnancy can be terminated. PGD, also called embryo selection, is a technique that involves in vitro fertilization with a biopsy at the six-to eight-cell stage of the embryo, 3 days after insemination. Only noncarrier embryos are transferred to the uterus. PGD has been used to avoid the potential risk of a miscarriage and decisions about pregnancy termination, both associated with PND. PND and PGD have first been applied to prevention of births of children with cancer predisposition syndromes that have early ages of onset, but PGD has been advocated for by members of families affected by hereditary breast/ovarian cancer. Potential reasons for not utilizing PGD include psychological, economic, and ethical considerations. A major factor is patient lack of awareness about applicable reproductive technologies. For example, in studies of acceptance of PGD among hereditary cancer family members, 50%–60% of those surveyed had not heard of PGD until the survey.36 Cost, restriction of services to major medical centers, lack of contact with reproductive specialists, and hesitancy among oncologists to bring up PGD as an option have been discussed as other limiting factors.36 Another potential factor is the negative judgment that such a step may imply about the value of a life lived with a cancer-predisposing hereditary syndrome. Personal and familial cancer experience may also influence such decisions. The use of decision aids and/or decisional counseling can be helpful in assisting individuals to make a balanced decision. Psychological Approaches That Support Genetic Testing Having well- designed genetic counseling to provide information and strategies for direct family communication reduces psychosocial distress.17 Studies have focused on psychoeducational approaches, added counseling sessions with an educator or psychologist, or using decisional aids (DAs) in counseling to facilitate decision-making concerning risk management options. CHAPTER 13 Psychosocial Issues in Genetic Testing for Breast/Ovarian Cancer DAs are available to support the decision to have genetic testing,37 or to undergo risk-reducing surgery versus surveillance,25 and are found to reduce decisional conflict, facilitate adjustment, and improve patient satisfaction.25 For example, a computerized decision aid (tailored to age, menopausal status, and breast cancer history) was found, in a randomized controlled trial, to be effective among carriers who were initially undecided about managing their breast cancer risk. It facilitated decision-making, decreased decisional conflict, and increased satisfaction.25 Similar decisional aids (both web and paper based) show reductions in decisional conflict, clarification of personal values and preferences, and improvements in satisfaction.25,27,37 Psychosocial support provided in one-on-one sessions or via telephone have also demonstrated benefit.38 For example, five weekly telephone sessions of psychosocial support following standardized genetic counseling for BRCA1/2 reduced cancer-related distress, anxiety, and depressive symptoms in the short term.38 While differences were not maintained at 1 year, the authors suggest that the alleviation of distress in the short term is especially important, given that it is the time when important risk management decisions are being made. For individuals with higher levels of cancer worry or with communication challenges that interrupt sleep or interfere with optimal coping and decision-making, additional follow-up is recommended. Enhanced genetic counseling guided by the Cognitive-Social Health Information Processing (C-SHIP) model39 adds a session with a health educator who uses the contemplation of possible genetic testing scenarios to elicit relevant cognitive and affective reactions. This helps women prepare and plan for using specific coping strategies to manage these reactions. The enhanced counseling group demonstrated greater knowledge compared to controls postintervention.39 Behavioral interventions, such as relaxation, mindfulness-based stress reduction, or distraction techniques, can assist in addressing anxiety and the lifelong stress associated with repeated medical and screening appointments. Cognitively oriented strategies typical of cognitive behavioral therapy used to manage anxiety or to facilitate medical decision-making are helpful and can be employed in individual or group formats to learn about the connections among thoughts, moods, and behaviors. Mental exercises such as thought records assist individuals in gaining insight on specific self-beliefs and catastrophic or rigid thinking habits. A counselor can help to identify inaccurate cognitions and to encourage more realistic interpretations of the circumstances. Psychodynamic therapies may be suitable but have yet to be tested. Because psychodynamic therapies focus on how previous relationships and bereavements relate to current distress, they may be well suited to helping people appreciate how previous losses related to cancer impact on current medical decision-making. Wellisch and Lindberg20 have highlighted the unique issues that are highly relevant in young women who lost a mother to cancer, particularly during their adolescence. These women can develop a sense that it is inevitable that they will suffer and die from cancer, just as their mothers did. In a psychodynamic framework this self-belief can be explored and eventually challenged within the safety of the therapeutic relationship. There is also evidence showing benefit through professionally led support groups that provide direct social support and incorporate therapeutic factors such as the vicarious learning that occurs among peers.40 Groups have shown benefit by decreasing cancer worry or general anxiety and may be particularly helpful to individuals who have endured prior loss to cancer.40 For example, principles of supportive-expressive group therapy (SEGT) have been applied to the genetic counseling context.40 The SEGT model focuses on the existential impact on one’s life of the genetic risk, and through the here and now of the group encourages individuals to live fully authentically.40 Sessions focus on explorations of past familial experiences of cancer and their influence on evaluation of risk, as well as their impact on current medical decision-making. The model also creates a forum for the processing of unresolved grief and the exploration of adjustment challenges. This occurs via an emphasis on the legitimacy of a full range of emotion and the centrality of relationships in giving value to life, a tenet taken from existential psychotherapy.40 There is further need for randomized controlled trials to address the psychosocial needs of the BRCA1/2 population. A number of interventions are currently being evaluated, including strategies to address the psychosexual and psychological impacts following risk- reducing surgery and lifestyle-behavioral interventions (e.g., physical fitness). Future Directions Information around BRCA1/2 testing has the potential to provide many benefits; however, genetic information can also result in emotional distress or decisional conflict around preventative options. Psychosocial, emotional, personal historical, cultural, and familial contextual factors play an important role in how an individual adapts to, and utilizes, genetic information. For those found to be at risk of maladaptive coping, standardized instruments can assist in identifying them and offering tailored interventions known to facilitate accurate knowledge and adjustment. During the coming years we will be faced with a number of additional challenges. These include the implementation of screening questionnaires for psychosocial issues as part of standard care, and the trend to offer fewer counseling sessions. The more recent availability of genetic knowledge of single nucleotide polymorphisms (SNPs) related to breast cancer risk and how best to convey the modest increased risk of these gene variants will require further study. There are also populations not yet included in research, such as various cultural minorities, who have less frequently taken up genetic counseling for cancer, whose specific issues need to be addressed. Further, direct-to-consumer testing, for example, through mail-order applications, requires further study to assess the impact of receiving genetic knowledge in the absence of genetic counseling. In this evolving field of genetics, it is of great importance that psychosocial researchers and clinical workers such as psychologists and social workers remain closely involved to ensure the best quality of care for this unique group of high-risk individuals and families. REFERENCES 1. Noone AM, Howlader N, Krapcho M, et al. SEER Cancer Statistics Review, 1975–2015. Bethesda, MD: National Cancer Institute; 2018. 2. Kinney AY, Steffen LE, Brumbach BH, et al. Randomized noninferiority trial of telephone delivery of BRCA1/ 2 genetic counseling compared with in-person counseling: 1-year follow-up. J Clin Oncol. 2016;34(24):2914–2924. 99 100 Section III Screening and Testing for Germ Line and Somatic Mutations 3. Daly MB, Pilarski R, Berry M, et al. NCCN guidelines insights: genetic/familial high-risk assessment: breast and ovarian, version 2.2017. J Natl Comprehens Cancer Netw JNCCN. 2017;15(1):9–20. 4. Wevers MR, Ausems MG, Verhoef S, et al. Does rapid genetic counseling and testing in newly diagnosed breast cancer patients cause additional psychosocial distress? Results from a randomized clinical trial. Genet Med. 2016;18(2):137–144. 5. Nelson HD, Pappas M, Cantor A, Haney E, Holmes R. Risk assessment, genetic counseling, and genetic testing for brca-related cancer in women: updated evidence report and systematic review for the US Preventive Services Task Force. JAMA. 2019;322(7):666–685. 6. Rhiem K, Engel C, Graeser M, et al. The risk of contralateral breast cancer in patients from BRCA1/2 negative high risk families as compared to patients from BRCA1 or BRCA2 positive families: a retrospective cohort study. Breast Cancer Res. 2012;14(6):R156. 7. Lostumbo L, Carbine NE, Wallace J. Prophylactic mastectomy for the prevention of breast cancer. Cochrane Database Syst Rev. 2010(11):CD002748. 8. Razdan SN, Patel V, Jewell S, McCarthy CM. Quality of life among patients after bilateral prophylactic mastectomy: a systematic review of patient- reported outcomes. Qual Life Res. 2016;25(6):1409–1421. 9. Gopie JP, Mureau MA, Seynaeve C, et al. Body image issues after bilateral prophylactic mastectomy with breast reconstruction in healthy women at risk for hereditary breast cancer. Fam Cancer. 2013;12(3):479–487. 10. Tercyak KP, Peshkin BN, Brogan BM, et al. Quality of life after contralateral prophylactic mastectomy in newly diagnosed high- risk breast cancer patients who underwent BRCA1/2 gene testing. J Clin Oncol. 2007;25(3):285–291. 11. Song CV, Teo SH, Taib NA, Yip CH. Surgery for BRCA, TP53 and PALB2: a literature review. Ecancermedicalscience. 2018;12:863. 12. Hesse-Biber S, An C. Genetic testing and post-testing decision making among BRCA-positive mutation women: a psychosocial approach. J Genet Couns. 2016;25(5):978–992. 13. Glassey R, Saunders C, Hardcastle SJ. Commentary: bilateral risk- reducing mastectomy is the safest strategy in BRCA1 carriers. Front Psychol. 2017;8:121. 14. Rowland E, Metcalfe A. A systematic review of men’s experiences of their partner’s mastectomy: coping with altered bodies. Psychooncology. 2014;23(9):963–974. 15. van der Velde NM, Mourits MJ, Arts HJ, et al. Time to stop ovarian cancer screening in BRCA1/2 mutation carriers? Int J Cancer. 2009;124(4):919–923. 16. Madalinska JB, Hollenstein J, Bleiker E, et al. Quality-of-life effects of prophylactic salpingo-oophorectomy versus gynecologic screening among women at increased risk of hereditary ovarian cancer. J Clin Oncol. 2005;23(28):6890–6898. 17. Lombardi L, Bramanti SM, Babore A, et al. Psychological aspects, risk and protective factors related to BRCA genetic testing: a review of the literature. Support Care Cancer. 2019;27(10):3647–3656. 18. Eijzenga W, Hahn DE, Aaronson NK, Kluijt I, Bleiker EM. Specific psychosocial issues of individuals undergoing genetic counseling for cancer—a literature review. J Genet Couns. 2014;23(2):133–146. 19. van Oostrom I, Meijers-Heijboer H, Duivenvoorden HJ, et al. Experience of parental cancer in childhood is a risk factor for psychological distress during genetic cancer susceptibility testing. Ann Oncol. 2006;17(7):1090–1095. 20. Wellisch DK, Lindberg NM. A psychological profile of depressed and nondepressed women at high risk for breast cancer. Psychosomatics. 2001;42(4):330–336. 21. Wenzel L, Osann K, Lester J, et al. Biopsychological stress factors in BRCA mutation carriers. Psychosomatics. 2012;53(6):582–590. 22. Esplen MJ, Cappelli M, Wong J, et al. Development and validation of a brief screening instrument for psychosocial risk associated with genetic testing: a pan-Canadian cohort study. BMJ Open. 2013;3(3):e002227. 23. Eijzenga W, Bleiker EM, Hahn DE, et al. Psychosocial aspects of hereditary cancer (PAHC) questionnaire: development and testing of a screening questionnaire for use in clinical cancer genetics. Psychooncology. 2014;23(8):862–869. 24. van Oostrom I, Meijers-Heijboer H, Duivenvoorden HJ, et al. A prospective study of the impact of genetic susceptibility testing for BRCA1/2 or HNPCC on family relationships. Psychooncology. 2007;16(4):320–328. 25. Schwartz MD, Valdimarsdottir HB, DeMarco TA, et al. Randomized trial of a decision aid for BRCA1/BRCA2 mutation carriers: impact on measures of decision making and satisfaction. Health Psychol. 2009;28(1):11–19. 26. Read CY, Perry DJ, Duffy ME. Design and psychometric evaluation of the Psychological Adaptation to Genetic Information Scale. J Nurs Scholarsh. 2005;37(3):203–208. 27. Cella D, Hughes C, Peterman A, et al. A brief assessment of concerns associated with genetic testing for cancer: the Multidimensional Impact of Cancer Risk Assessment (MICRA) questionnaire. Health Psychol. 2002;21(6):564–572. 28. Maheu C, Thorne S. Receiving inconclusive genetic test results: an interpretive description of the BRCA1/2 experience. Res Nurs Health. 2008;31(6):553–562. 29. Montgomery SV, Barsevick AM, Egleston BL, et al. Preparing individuals to communicate genetic test results to their relatives: report of a randomized control trial. Fam Cancer. 2013;12(3):537–546. 30. Tercyak KP, Mays D, DeMarco TA, et al. Decisional outcomes of maternal disclosure of BRCA1/2 genetic test results to children. Cancer Epidemiol Biomarkers Prev. 2013;22(7):1260–1266. 31. Santerre-Theil A, Bouchard K, St-Pierre D, Drolet AM, Chiquette J, Dorval M. Development of a tool to guide parents carrying a BRCA1/2 mutation share genetic results with underage children. J Cancer Educ. 2018;33(3):569–575. 32. Peshkin BN, Demarco TA, Tercyak KP. On the development of a decision support intervention for mothers undergoing BRCA1/ 2 cancer genetic testing regarding communicating test results to their children. Fam Cancer. 2010;9(1):89–97. 33. Tercyak KP, Bronheim SM, Kahn N, et al. Cancer genetic health communication in families tested for hereditary breast/ovarian cancer risk: a qualitative investigation of impact on children’s genetic health literacy and psychosocial adjustment. Transl Behav Med. 2019;9(3):493–503. 34. Werner- Lin A, Merrill SL, Brandt AC, Barnett RE, Matloff ET. Talking with children about adult-onset hereditary cancer risk: a developmental approach for parents. J Genet Couns. 2018;27(3):533–548. 35. Chan JL, Johnson LNC, Sammel MD, et al. Reproductive decision- making in women with BRCA1/2 mutations. J Genet Couns. 2017;26(3):594–603. 36. Vadaparampil ST, Quinn GP, Knapp C, Malo TL, Friedman S. Factors associated with preimplantation genetic diagnosis acceptance among women concerned about hereditary breast and ovarian cancer. Genet Med. 2009;11(10):757–765. 37. Grimmett C, Pickett K, Shepherd J, et al. Systematic review of the empirical investigation of resources to support decision-making regarding BRCA1 and BRCA2 genetic testing in women with breast cancer. Patient Educ Couns. 2018;101(5):779–788. CHAPTER 13 Psychosocial Issues in Genetic Testing for Breast/Ovarian Cancer 38. Graves KD, Wenzel L, Schwartz MD, et al. Randomized controlled trial of a psychosocial telephone counseling intervention in BRCA1 and BRCA2 mutation carriers. Cancer Epidemiol Biomarkers Prev. 2010;19(3):648–654. 39. Roussi P, Sherman KA, Miller S, et al. Enhanced counselling for women undergoing BRCA1/2 testing: impact on knowledge and psychological distress-results from a randomised clinical trial. Psychol Health. 2010;25(4):401–415. 40. Esplen MJ, Hunter J, Leszcz M, et al. A multicenter study of supportive-expressive group therapy for women with BRCA1/ BRCA2 mutations. Cancer. 2004;101(10):2327–2340. 101 14 Psychosocial Issues in Genetic Testing for Hereditary Colorectal Cancer Sukh Makhnoon and Susan K. Peterson Introduction Genetic testing for hereditary colorectal cancer (CRC) syndromes is increasingly used in clinical practice to ascertain inherited disease susceptibility or genetic etiology. A primary benefit of genetic testing is the resulting ability to provide targeted prevention strategies and surveillance for the proband as well as their family. Psychosocial research on hereditary CRC has focused on understanding individuals’ motivations and decisions regarding genetic testing, the psychological impact of genetic risk notification, the effects on family and interpersonal relationships, and factors influencing the uptake of risk reduction options (e.g., screening, risk-reducing surgery, or chemoprevention). We review the literature on the impact of genetic counseling (GC) and genetic testing on individuals at risk for two common hereditary CRC syndromes, Lynch syndrome and familial adenomatous polyposis. Findings from these studies can guide clinicians in understanding why people seek GC and genetic testing, how they cope with the results of testing, and how they subsequently integrate that information into cancer prevention and treatment decisions. Hereditary Colorectal Cancer An estimated 30% of all CRCs involve a hereditary component, approximately 5% of which are caused by highly penetrant inherited Mendelian pathogenic variants. The most common Mendelian risk of CRC (2–4% of all CRCs) is due to Lynch syndrome (LS), also known as hereditary nonpolyposis colon cancer (HNPCC). LS is an autosomal dominant condition conferred by inherited mutations in mismatch repair (MMR) genes including MLH1, MSH2, MSH6, and PMS2 and deletions in EPCAM.1 LS is characterized by a 70– 80% lifetime risk of CRC and 50–60% risk of endometrial cancer, with increased lifetime risks for ovarian, stomach, small bowel, hepatobiliary tract, pancreatic, urinary tract, brain, and skin cancers.2 Identification of these genetic mutations allows for targeted risk reduction in mutation carriers and for notification of risk to relatives who may also be carriers. Familial adenomatous polyposis (FAP) is the second most common inherited CRC syndrome with a prevalence of 1 in 10,000 individuals. FAP is an autosomal dominant condition that results from germline mutations in the APC gene with a penetrance of 100%, meaning that cancer develops universally in mutation carriers. FAP is characterized by the development of hundreds to thousands of colonic adenomas, beginning in early adolescence, and inevitable CRC in untreated individuals. Prophylactic colectomy (i.e., removal of part of the colon) is the ultimate treatment for FAP, and surgery is often completed in early adulthood to try to prevent the development of CRC. Other rarer forms of hereditary CRCs include autosomal dominant conditions such as attenuated FAP, Peutz-Jeghers syndrome (PJS), and juvenile polyposis syndrome (JPS), and autosomal recessive MUTYH-associated polyposis (MAP). The etiologies of the remaining 25% of inherited CRCs are not completely understood. They are likely caused by either alterations in single genes that are less penetrant but more common than those associated with the well-characterized syndromes or alterations in multiple susceptibility loci that have additive effects. Landscape of Genetic Testing Options Genetic testing for LS currently involves a time- consuming multistep process that begins with immunohistochemistry (IHC) and/or microsatellite instability (MSI) tumor testing for MMR deficiency associated with LS genetic variants. For assays suggestive of deficiency or loss of function on MMR genes, a conclusive germline sequencing test determines whether there is a pathogenic variant present. Current guidelines recommend universal tumor screening (UTS) of colorectal and endometrial tumors through MSI or IHC, regardless of patients’ family history to comprehensively identify persons at risk of LS. Patients whose tumors show abnormal MSI or IHC results through UTS should be referred for GC and genetic testing (gold standard). Around 20 candidate genes connected with hereditary CRC have been identified thus far, many of which are included in germline CHAPTER 14 Psychosocial Issues in Genetic Testing for Hereditary Colorectal Cancer gene sequencing panels. These include Mendelian genes with high penetrance as well as less well-understood genes with moderate penetrance. Targeted or panel genetic testing for hereditary CRC is important for prevention through screening and surveillance (e.g., colonoscopy), chemoprevention (e.g., Sulindac), and prophylactic surgery (colectomy), and illness management for index patients as well as their relatives. With increasing use of multigene panel tests, ever more secondary and inconclusive findings are being discovered, which presents unique clinical management and psychosocial challenges. Massively parallel sequencing of protein coding regions of genes (through exome sequencing [ES]) are being explored to replace traditional sequencing due to their increased ability to find pathogenic variants and decreasing cost. However, these next- generation sequencing tests are not yet a part of routine clinical testing for CRC as they present important challenges including interpretation of incidental and uncertain findings, counseling before and after testing, and informed consent of patients. Psychosocial Issues of Genetic Counseling Clinical genetic testing is best accompanied by formal pre-and posttest GC because of the significant medical and emotional impact genetic test results can have on patients and their family members. Counseling is important to inform patients about the possibility of probabilistic or uncertain information derived from genetic tests, the consequent healthcare choices one may face, and their potential psychosocial consequences, and to offer emotional and decision support. Yet, evidence suggests that rates of referral to GC, rates of attendance, and availability of counseling services are suboptimal. According to US national data from 2015, 4.8 million in the US have undergone GC, 1.3 million for CRC, of which 49% were male and 51% female.3 The majority of participants who reported receiving GC reported they did so because their doctor recommended it (66%), with smaller proportions describing self (12%), family (10%), or media (5%) influences as the primary reason. In the clinical setting, uptake of LS GC among patients ranged from 30% to 87%, with various logistical and psychosocial concerns reported as barriers to uptake. Patients who were more likely to undergo GC were more educated, older, and married and had a family history of LS-associated cancers (85% in those with family history vs. 35% overall).4 Cost and logistical barriers, emotional concerns, family concerns, and low perceived personal relevance were reported as important considerations for those declining GC. New models of GC are being tested to counteract some of these access challenges and to solve the lack of availability of genomics professionals to return large volumes of genetic test results. Innovative alternate modes of counseling including telegenetic delivery of counseling via the internet, web-based platforms, and phone counseling are noninferior to in-person counseling and can solve the issue of limited access to genetic services.5 Uptake Uptake of LS Testing UTS offered through routine care results in high uptake of LS genetic testing and has already been integrated into standard clinical practice at many healthcare systems.6 Patients who are offered germline LS testing as a part of UTS clinical workflow have more active follow-up, which facilitates referral to GC and improves completion of genetic testing. In fact, most patients who are offered LS genetic testing through UTS tend to accept testing, although there is some variation in uptake across studies. For example, in a secondary care setting in the UK, all 23 defective MMR CRC cases identified through UTS underwent GC and were offered and took up genetic testing.7 In other clinical oncology practice settings, more patients underwent germline testing when systematic tumor screening programs were implemented.8 On the other hand, nonsystematic approaches to screening that rely on clinicians or patients for referral perform less well than UTS. Reported uptake of genetic testing for LS varies from 14% to 75% across studies, reflecting possible cultural and sample selection biases. Patients may miss germline testing because they were not referred for counseling or because counseled patients did not meet the criteria for testing. Certain subgroups of patients may be less likely to follow through with genetic testing after receiving GC for abnormal tumor screening results, including younger patients and those with lower incomes. This is particularly concerning since CRC patients diagnosed at younger ages present acutely at more advanced stages than is typical for older patients9 and are more likely to carry a germline mutation.10 Uptake of APC Testing The question asked about APC testing is typically “when?” rather than “whether?” due to the medical and psychosocial benefits for children and adolescents of learning whether they are APC mutation carriers. The optimal timing of APC testing occurs when the individual being tested is of sufficient age, maturity, and psychological stability to understand the reasons that testing is being offered and the implications of the test result. There is general consensus not to test children before the age of 10–12 years (in the absence of clinical symptoms) since there are no medical benefits and there is a potential detrimental psychological impact. There are data suggesting that uptake of genetic testing for FAP is higher than that for LS and may be above 80% among asymptomatic at-risk adults11 and 96% for children ages 10–16.12 This may be because mutation is nearly 100% penetrant and FAP is preventable through surgery. Population Genetic Screening Population genetic screening, including screening for LS, is being discussed given the increased national focus on precision health and genomics-informed care. Although it is not yet offered, LS is suitable for population screening for two main reasons: (1) it has a highly penetrant and well-defined genetic cause, with existing clinical interventions, and has been designated by the Centers for Disease Control and Prevention (CDC) as “Tier 1 genomic application” and (2) the population prevalence is almost double than previously estimated. Discrete choice research shows that the general population is willing to participate in population genetic screening for CRC,13 but preferences varied depending on individual experiences, perceived anxiety, worry about colonoscopy frequency, and the probability of developing CRC. Access and Referral Referral to genetic evaluation of hereditary CRC syndromes is critical for genetic test uptake; however, referral rates remain suboptimal 103 104 Section III Screening and Testing for Germ Line and Somatic Mutations with important disparities in access. For example, among 165 eligible individuals who met Amsterdam II clinical criteria for HNPCC, only 31% reported being advised to undergo GC by their doctor, and only 7% had undergone genetic testing.14 Despite similar rates of colorectal tumor analysis, minority patients were less likely to be recommended for genetic evaluation or to undergo germline testing for LS.15 Surveys of healthcare provider knowledge have consistently shown major deficiencies in the understanding of genetics risk assessment and testing and ability to identify and differentiate risk status. Other factors that positively affect access are clinic location, ability to pay, insurance, discouragement by family members, patient attitudes, norms, and education level. One barrier that needs to be overcome to improve referral rates is the taking of complete cancer family histories. Taking a detailed history is pivotal in identifying patients with high-risk personal and family history of cancer who will benefit from referral for genetic evaluation and genetic testing. A 2014 study of 212 medical practices with 10,466 patients found that first-degree relative family histories were recorded for 77% of patients and second-degree family histories for 61.5% of patients, with significantly better documentation for breast cancer patients than for patients with CRC.16 However, complicating matters, in early-onset CRC (<50 years of age), although a significant proportion of patients carry germline cancer predisposition genes, nearly half did not have a family history and one-fifth did not meet the National Comprehensive Cancer Network (NCCN) clinical criteria for relevant syndrome,17 which is why UTS may be critical to capture all patients who may benefit from genetic testing. Motivation Motivation for LS Testing Once referred, factors that positively affect test uptake include having a personal history of cancer, having more relatives with LS-related cancers, higher perceived risk of CRC and related cancer, stronger beliefs that hereditary CRC would influence one’s life and that of one’s offspring, and more intrusive thoughts about CRC.18 Uptake of genetic testing did not differ by gender, but higher educational level, having a spouse or partner, and being employed correlated positively with uptake. Both men and women appear motivated to seek testing to determine whether offspring are at increased cancer risk. Women may be more likely than men to want testing to determine whether they require enhanced cancer screening and to undergo testing as a response to recommendations of a physician or genetic counselor.19 Decliners of genetic testing for LS were more likely to report depressive symptoms, to be nonadherent to colorectal screening recommendations, to have less confidence that they could cope with a positive test result, to be concerned about insurability, and to worry about the emotional impact of genetic testing on self and family.18 Psychosocial issues related to secondary and uncertain findings derived from ES, and acceptability of ES over panel testing need to be better understood before clinical implementation. In a discrete choice experiment conducted within a pragmatic trial of ES versus panel sequencing, participants preferred to undergo tests that detect a higher proportion of individuals with a definitive genetic etiology and involve a shorter wait time.20 Most adults accept and were satisfied with the results, and few reported distress or expected unsolicited findings.21 Patients who underwent ES preferred to filter secondary findings to avoid information overload and to avoid learning what the future holds. Motivation for FAP Testing Patient or parent attitudes seem to differ from professional guidelines of genetic testing for FAP. In an Australian study of young adults with FAP, 61% had a preference to test their children at birth or very early in life (before age 10).12 In a qualitative study of children tested for FAP below age 10, none of the parents regretted the timing of genetic testing and none observed changes in the mental or physical health in their child after testing. The age at which children with a parent with FAP had genetic testing varied in a recent study from 1 to 16 years, both overly early and late assessments.22 The major reasons for testing at the young age were (1) testing of all children in the family at the same moment, (2) certainty for the future, and (3) preparing the child for future surveillance. Also, young genetic testing did not lead to colon surveillance before it was indicated. Genetic testing for FAP at a young age is experienced as causing no harm by parents.23 This suggests the need for ongoing counseling of parents with FAP about the benefits and risks of genetic testing for their children. GC and follow-up along the life span are recommended, in recognition of the lifelong issues raised by FAP.12 Risk Perception Decision-making for genetic testing assumes an accurate understanding of the admittedly complex risks that are involved. Accurate risk perception for LS is complicated by the fact that there are multiple cancer risks to consider. Although GC increases accuracy of risk perception for LS-associated cancers, not all counseled individuals accurately report their cancer risks.24 Inaccurate cancer risk perceptions among individuals undergoing GC for LS encompass both over-and underestimation of risk, with only one-third to one- half of participants in one study accurately reporting the levels of risk conveyed in the GC they attended.25 For example, even though mutation carriers have higher cancer risk, there was no difference in risk perception of HNPCC between carriers and noncarriers by 12 months after testing,26 and perceived risk in carriers was lower 12 months posttest compared with before undergoing genetic testing.26 Even when no pathogenic variant or low-risk variants are identified, some patients remained in a heightened state of risk perception.27 Perceived risk is a strong moderator of emotion and an influential predictor of psychobehavioral outcomes,28 but it is shaped not only by genetic results but also by lived experiences such as previous diagnosis and family history of cancer, perceived controllability, existing therapies for managing illness, and information provided during GC. Among a nationally representative sample of 15,085 men and women, a higher proportion of individuals with CRC risk perceptions concordant with their objective risk undergo GC or testing for CRC risk.29 Emotional Impact Psychosocial outcomes of genetic testing in the context of single or multigene testing have been studied extensively and there is consensus that negative outcomes of genetic testing are few and far between. When observed, negative psychosocial effects are short-lived and participants return to baseline within a few months after testing. Negative Emotional Impact in LS Overall, psychosocial harms associated with communication of genomic information are mild and transient. Research suggests that individuals who have received test results for LS-related mutations CHAPTER 14 Psychosocial Issues in Genetic Testing for Hereditary Colorectal Cancer experienced immediate increased general distress,30 cancer-specific distress,31 and worry about cancer,30 but that the mean increases did not raise scores above normal levels. Generally, distress receded over the first year following genetic testing30 and was at pretest levels by 12 months.31 One study three years after disclosure of test results showed scores similar to those before genetic testing, except that noncarriers’ cancer-specific distress was significantly lower than baseline scores.32 In general, there was no difference in general distress between carriers and noncarriers due to genetic testing or disclosure of test result. In longitudinal studies, there was no significant change in cancer-specific distress or worry33 between pre-and posttest result disclosure. However, there are subgroups of tested individuals at greater- than- average risk of psychological distress following testing. Women, younger people, nonwhites, and individuals with less satisfactory social support and lower educational levels had higher levels of general and cancer-specific distress, regardless of mutation status in the 12 months following testing.30 Other studies have reported that individuals with a prior history of major or minor depression or those with more affected first-degree relatives or those reporting more intense grief reactions had greater distress one to six months after disclosure.34,35 A recent study showed that reductions in distress over the first six months following disclosure for LS genetic testing was moderated by the individual’s health information coping style. Generally, individuals testing negative experienced relief and decreased distress, which was long-lasting.30 Such individuals are advised that they no longer require enhanced screening and can return to following general population guidelines for colonoscopy screening. It has been found, however, that some at-risk individuals testing negative for LS-related mutations evidence distrust of the test result to the extent that they do not give up colorectal screening,36 although this has not been found in all studies.24 In fact, participants’ pretest emotional state predicts subsequent distress to a much greater degree. The potential psychological harms of next-generation sequencing, which generates vast amounts of complex data that can be highly uncertain and reveal information unrelated to clinical context for sequencing, is concerning, but current evidence shows no clinically significant psychological harms of returning ES results to patients.37 Among CRC/polyposis patients, there was no significant change in anxiety or depressive symptoms following return of ES results pre- and postdisclosure.37 In a qualitative study of LS patients with variants of uncertain significance (VUS), most perceived various types of uncertainty associated with their VUS. Half of the participants appraised their variant as a danger and implemented coping strategies to reduce the threat of developing cancer. Problematically, the majority of participants were unaware of the possibility of a VUS before receiving their result and expected reclassification over time.38 Individuals with VUS may also have persistent levels of genetic test– related distress as well as more difficulty in recalling the meaning of their results than those with more definitive test results.38 In a quantitative study of ES, test-related uncertainty among adults with colon polyps or cancer was lower than parents of pediatric patients but higher than healthy adults.37 Among patients with clinical suspicion of hereditary cancer, 12 months after result disclosure, carriers of moderate-penetrance variants had higher distress and uncertainty scores compared with carriers of high-penetrance variants.39 Research on psychosocial outcomes of secondary findings is in its infancy and qualitative studies show that participants found secondary findings unexpected but not shocking. It is difficult to conduct quantitative studies to measure the psychosocial impact of secondary findings as return of secondary findings without return of primary results is uncommon. Negative Emotional Impact in FAP Similar to LS, psychometric data indicate that FAP patients and at- risk relatives as a group do not exhibit clinical symptoms of mental health problems after clinical or genetic diagnosis. However, some subgroups are more vulnerable to distress. The psychological impact of testing children for FAP needs special attention. Several studies have investigated the distress of children tested for APC mutations. Mean scores for individuals in these studies remain in the normal range on measures of anxiety, depression, and behavioral functioning following disclosure of test results.11 There is evidence that the impact of genetic testing of individual family members is moderated by knowledge of the results of other family members.40 In families where some children tested positive and others negative, the parents who were not the FAP mutation carriers had significantly increased depression scores after disclosure of their children’s results. Similarly, in a Norwegian study of 22 adolescent offsprings of a parent with FAP, 30–70% fulfilled criteria for a psychiatric diagnosis.40 This finding was independent of the FAP status of the offspring, illustrating the psychological burden of having an affected parent. It was only in families with FAP that unaffected children sometimes described feeling a sense of guilt when their sibling was affected.41 Individuals testing negative for APC mutations, while relieved of much of the medical burden of the disease, often experience guilt about avoiding the lifelong worry and need for intervention that their parents and/or siblings often share.42 Anxiety may be high in these children as well, and their need for support should be considered in any plan to offer emotional counseling to FAP family members. In a cross-sectional study of adults who underwent APC genetic testing, mutation carriers had higher levels of state anxiety than noncarriers and were more likely to have clinically significant anxiety levels. Lower optimism and lower self-esteem were associated with higher anxiety in this study, and FAP-related distress, perceived seriousness of FAP, and belief in the accuracy of genetic testing were associated with more state anxiety among carriers. In a long- term follow-up study, 20% had moderate to severe distress,43 26% had received psychosocial support, and an additional 30% wanted more psychosocial care. Forty percent said that work and relationships were adversely affected by FAP.43 Similarly, moderate to severe psychological distress was reported in 30% of the partners of FAP patients. While they also reported problems with work, leisure activities, and relationships, most partners reported good overall quality of life.43 An Australian study of 18-to 35-year-old young adults with or at risk for FAP, which utilized FAP-specific measures of distress, suggested that unmarried (i.e., single) individuals and those who had had more extensive initial surgery had greater distress.11 Psychological functioning was highest among subjects who had not yet had any surgery, with more negative outcomes related to body image, sexual functioning, and affect reported by those who had had ileal pouch–anal anastomosis surgery. Having such intrusive surgery during the period of young adulthood when sexual 105 106 Section III Screening and Testing for Germ Line and Somatic Mutations identity and sexual relationships are being established is, the authors believe, highly problematic. They advise that when medically feasible, psychosocial factors should be incorporated into surgical decision-making. Positive Emotional Impact There are psychological benefits of genetic testing for people who do not inherit the high-risk mutations, such as short-and long-term decreases in cancer worry, general anxiety, and depression.44 They also have the removal of uncertainty, the assurance of knowing that their children are not at risk, avoidance of intensive cancer surveillance, and removal of concerns regarding genetic discrimination.44 ES for CRC/polyps showed no difference in positive feelings.37 However, these findings of positive and negative emotional impact should be considered in light of the fact that most studies conducted to date are biased toward Caucasian women with high education and socioeconomic status and do not generalize to subpopulations of people who are particularly vulnerable. Clinicians should still be vigilant about negative effects on their patients. Interventions to manage psychosocial problems have been evaluated and shown to be promising in reducing the problems. Decision Aids Given the complexity of the decision to undergo genetic testing, researchers have begun to test innovative strategies to facilitate education and decision-making about inherited cancer risk and genetic testing. These include development of disease-specific decision aids to enhance the counseling experience,45 to inform pretest counseling for incidental genomic results,46 and for reproductive decisions.47 Decision-making about genetic testing for LS is a multistep process, and decision aids may be particularly useful in this context. Cancer Screening An important determinant of the success of GC and testing is the degree to which at-risk individuals who are found to be mutation carriers alter their prevention behaviors. Among at-risk carriers, compliance with screening recommendations assessed from six months to seven years after genetic testing showed increases. Rates of appropriate surveillance was higher among individuals who had undergone genetic evaluation compared with those who had not and in those who had a first-degree relative with CRC.48 Screening adherence in carriers may be enhanced by increasing commitment and self-efficacy regarding colonoscopy, as well as greater perceived benefits and fewer perceived barriers of screening.49 Lack of consistent and clear advice about surveillance recommendations, especially for rarer cancer types, is a barrier to adequate risk management behavior among LS carriers. Providers surveyed across 21 countries reported that most (78%) provided recommendations or made referrals for gynecologic screening, but only 46% made recommendations for urologic screening, and most did not recommend screening/surveillance for other types of cancers such as neurologic, pancreatic, and hepatobiliary.50 Chemoprevention with aspirin as a risk management strategy was used least commonly (35%) compared to recommendations for colonoscopy.50 The burden and challenge of LS can be significantly reduced by finding knowledgeable providers who clearly and empathetically describe the complex, ongoing need for screening and the results of screening tests. System barriers include difficulty navigating visits of numerous specialists, who are often in different hospital systems and often have differing expectations for the type and frequency of screening tests needed. These problems highlight the need for coordinated, multidisciplinary care by specialists familiar with hereditary cancer. Family Communication and Cascade Testing Psychosocial predictors of familial communication of genetic information have largely focused on families with known mutation carriers and higher risk. Research shows that a family-mediated approach of familial communication of genomic risk is complex, and often selective and incomplete. Patients are motivated to share due to feelings of responsibility or obligation toward relatives, due to a desire to prevent disease in relatives,51 and because the family members are emotionally close and share information.52 First-degree relatives, especially female siblings, children, and parents, are told about genetic results more often than second-or third-degree relatives.51,53 On the other hand, inhibiting influences include difficulty conveying cancer risk information,53 and 57% of respondents being informed by a family member found it burdensome.5. Little is known about family communication in FAP families. Despite the importance of germline testing for family members, uptake is still considerably lower than 100%, with LS test uptake rates among at-risk family members reported between 41% and 90%.55 Predictors of being tested include age, gene, one or more tested siblings, no siblings, and parent under endoscopic surveillance.56 Nontested differed from tested respondents in that they were younger, they were less closely related to the index patient, and a lower proportion had children. The most important reasons for declining genetic testing were anticipating problems with life insurance and mortgage, being content with life as it is, and not experiencing any physical complaints.54 Unaffected members of mutation-carrying families who declined genetic testing reported the following barriers: a lack of knowledge of the availability of genetic testing, a lack of trust in genetic test information, a desire to see a stronger benefit from genetic testing before proceeding, and a sense that there may be more negative than positive outcomes from genetic testing.57 There is an unmet need for better/innovative access to counseling/ testing to identify at-risk individuals in extended families of mutation carriers. For instance, tailored in-person or telephone formats for providing CRC risk counseling, incorporating behavioral interventions, appear to improve knowledge and risk perceptions, with high client satisfaction.58 A family-mediated approach of communication, although generally acceptable to most patients,54 is less effective than provider-initiated approaches at increasing cascade testing rates.55 Although uptake following a proband-mediated contact was higher for LS than for hereditary breast and ovarian cancer (HBOC) families,55 there is both a need and a desire for more direct contact of at-risk family members to inform them of their risk and invite them to participate in GC and testing.59 CHAPTER 14 Psychosocial Issues in Genetic Testing for Hereditary Colorectal Cancer Influence of Genetic Testing on Childbearing Data is emerging about attitudes toward childbearing and prenatal genetic testing among individuals at risk for LS. Among 161 individuals assessed just prior to undergoing clinical genetic testing for LS, the majority already had at least one child, and only 9% said that their familial cancer risk had led them to decide not to have (more) children. When asked if it was ethical to offer prenatal testing (preimplantation genetic diagnosis [PGD] or prenatal genetic testing ), 66% agreed/strongly agreed, 25% were neutral, and only 9% strongly disagreed. Of the 48 subjects planning future pregnancies, 56% thought their test result would not influence their childbearing plans, 29% thought they would plan to have their children earlier in order to have prophylactic oophorectomy sooner, 27% would consider adoption, and only 10% said they would not have children if they were found to be a mutation carrier. At one-year posttest follow-up, however, considerably fewer mutation carriers evidenced interest in prenatal testing, with only 22% considering PGD for a future pregnancy.60 Of 17 MMR mutation carriers, 57% preferred spontaneous natural conception versus 28% and 35% who chose prenatal genetic testing and PGD, respectively.61 None of the main sociodemographical, psychological, or medical variables (including fear of transmitting mutations) were significantly associated with the reproductive preferences.61 Prenatal genetic screening seems higher among adults with FAP than among those undergoing testing for LS.62 Studies reported high interest in PGD among affected subjects but also indicated that many young adults with FAP are not routinely informed about PGD and prenatal genetic testing options.63 Most would not consider terminating the pregnancy if the fetus was affected. Due to the nearly 100% penetrance, early age of onset, and doubt over whether colectomy is a “real treatment,” PGD for FAP was approved by organizations like the Human Fertilisation and Embryology Authority despite their initial misgivings. Future Directions The literature on the psychosocial aspects of hereditary CRC is considerably more limited than that on the psychosocial aspects of GC and genetic testing for hereditary breast and ovarian cancer, but it has grown substantially in recent years. Generally, psychosocial factors influence the decision for genetic testing and risk management strategies; uptake of GC and genetic testing varies across studies; and distress following LS testing, particularly in the long term, is low for both carriers and noncarriers, although distress is slightly elevated following genetic testing for FAP. Distress may be related to the intensity of recommended screening, single marital status, test results of other family members, or the impact of the surgery. These factors, many tied to psychological rather than medical variables, suggest that counseling, both genetic and psychological, is important for individuals dealing with hereditary disease, not just at the time of testing, but also at many other points along the trajectory of patient care for these diseases. One of the differentiating features of hereditary disease is that, as patients have said, “It’s never over.” As such, support services must be geared to the ongoing needs of those who continue to face genetic risk for serious diseases. Future research will help to define the best ways to provide such services. The issues of how best to inform relatives about familial mutations predisposing to markedly elevated, early cancer risks and the definition of duty to warn in the context of genetic testing for diseases where potentially life-saving interventions exist raise interesting and challenging questions. Attitudes may be changing about the relative value of confidentiality and privacy and the need to inform at-risk relatives about their high levels of cancer risk and about options to detect or prevent cancer. Recognizing that there are some families where relatives do not communicate puts the onus on medical professionals to communicate with such members. To save lives, all family members of someone carrying a deleterious mutation must be informed by whomever is able to do so (family member, physician, or genetic counselor). Being notified of the possibility twice versus not being notified at all is a much better solution to this problem.34 Important but underexplored psychosocial issues exist regarding the increasing use of multigene panel testing, especially as genes of varying penetrance, associated cancer risks, and uncertainties are introduced. Research is needed to examine possible patterns of patient responses to various types of genetic information to develop approaches to help patients prepare for and adapt to these test results. In addition, research is needed to alleviate current methodological challenges including lack of sensitive instruments to detect subtle psychological responses expected in response to genetic testing, which may be limiting our abilities to accurately quantify psychosocial outcomes. Psychosocial factors play a large role in the impact of cancer genetic testing for CRC. Uptake of testing, adherence to screening recommendations, surgical choice, and family communication are likely to be influenced by the individual’s experience of hereditary illness in their family, the individual’s affective style and information preferences, and other little-studied social factors, such as socioeconomic status and access to genetic health services. Such factors will impact the ultimate integration of genetic testing into general medical practice. It is also important that routine taking of multigenerational cancer family histories for all CRC patients becomes standard of care. Hereditary CRC offers a model illustrating the potentially life-saving value of genetic testing where critical attention to psychological issues and support needs may optimize utilization. REFERENCES 1. Snyder C, Hampel H. Hereditary colorectal cancer syndromes. Seminars in oncology nursing. 2019;35(1):58–78. 2. Yurgelun MB, Hampel H. Recent advances in Lynch syndrome: Diagnosis, treatment, and cancer prevention. American Society of Clinical Oncology educational book American Society of Clinical Oncology Annual Meeting. 2018;38:101–109. 3. Stamp MH, Gordon OK, Childers CP, Childers KK. Painting a portrait: Analysis of national health survey data for cancer genetic counseling. Cancer medicine. 2019;8(3):1306–1314. 4. Irons RF, Contino KM, Horte JJ, et al. Success of referral to genetic counseling after positive lynch syndrome screening test. International journal of colorectal disease. 2017;32(9):1345–1348. 5. Buchanan AH, Rahm AK, Williams JL. Alternate service delivery models in cancer genetic counseling: A mini-review. Frontiers in oncology. 2016;6:120. 107 108 Section III Screening and Testing for Germ Line and Somatic Mutations 6. Beamer LC, Grant ML, Espenschied CR, et al. Reflex immunohistochemistry and microsatellite instability testing of colorectal tumors for Lynch syndrome among US cancer programs and follow-up of abnormal results. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 2012;30(10):1058–1063. 7. Cavazza A, Radia C, Harlow C, Monahan KJ. Experience of the implementation and outcomes of universal testing for Lynch syndrome in the United Kingdom. Colorectal disease: the official journal of the Association of Coloproctology of Great Britain and Ireland. 2019;21(7):760–766. 8. Cohen SA, Laurino M, Bowen DJ, et al. Initiation of universal tumor screening for Lynch syndrome in colorectal cancer patients as a model for the implementation of genetic information into clinical oncology practice. Cancer. 2016;122(3):393–401. 9. Plunkett M, Murray M, Frizelle F, Teague L, Hinder V, Findlay M. Colorectal adenocarcinoma cancer in New Zealand in those under 25 years of age (1997– 2007). ANZ journal of surgery. 2014;84(5):371–375. 10. Pearlman R, Frankel WL, Swanson B, et al. Prevalence and spectrum of germline cancer susceptibility gene mutations among patients with early- onset colorectal cancer. JAMA oncology. 2017;3(4):464–471. 11. Andrews L, Mireskandari S, Jessen J, et al. Impact of familial adenomatous polyposis on young adults: Quality of life outcomes. Diseases of the colon and rectum. 2007;50(9):1306–1315. 12. Andrews L, Mireskandari S, Jessen J, et al. Impact of familial adenomatous polyposis on young adults: Attitudes toward genetic testing, support, and information needs. Genetics in medicine: official journal of the American College of Medical Genetics. 2006;8(11):697–703. 13. Veldwijk J, Lambooij MS, Kallenberg FG, et al. Preferences for genetic testing for colorectal cancer within a population-based screening program: A discrete choice experiment. European journal of human genetics: EJHG. 2016;24(3):361–366. 14. Patel SG, Ahnen DJ, Kinney AY, et al. Knowledge and uptake of genetic counseling and colonoscopic screening among individuals at increased risk for lynch syndrome and their endoscopists from the Family Health Promotion Project. The American journal of gastroenterology. 2016;111(2):285–293. 15. Muller C, Lee SM, Barge W, et al. Low referral rate for genetic testing in racially and ethnically diverse patients despite universal colorectal cancer screening. Clinical gastroenterology and hepatology: the official clinical practice journal of the American Gastroenterological Association. 2018;16(12):1911–1918.e1912. 16. Wood ME, Kadlubek P, Pham TH, et al. Quality of cancer family history and referral for genetic counseling and testing among oncology practices: A pilot test of quality measures as part of the American Society of Clinical Oncology Quality Oncology Practice Initiative. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 2014;32(8):824–829. 17. Stoffel EM, Koeppe E, Everett J, et al. Germline genetic features of young individuals with colorectal cancer. Gastroenterology. 2018;154(4):897–905.e891. 18. Bleiker EM, Esplen MJ, Meiser B, Petersen HV, Patenaude AF. 100 years Lynch syndrome: What have we learned about psychosocial issues? Familial cancer. 2013;12(2):325–339. 19. Esplen MJ, Berk T, Butler K, Gallinger S, Cohen Z, Trinkhaus M. Quality of life in adults diagnosed with familial adenomatous polyposis and desmoid tumor. Diseases of the colon and rectum. 2004;47(5):687–695; discussion 695–686. 20. Weymann D, Veenstra DL, Jarvik GP, Regier DA. Patient preferences for massively parallel sequencing genetic testing of colorectal cancer risk: A discrete choice experiment. European journal of human genetics: EJHG. 2018;26(9):1257–1265. 21. Sie AS, Prins JB, van Zelst-Stams WA, Veltman JA, Feenstra I, Hoogerbrugge N. Patient experiences with gene panels based on exome sequencing in clinical diagnostics: High acceptance and low distress. Clinical genetics. 2015;87(4):319–326. 22. Levine FR, Coxworth JE, Stevenson DA, Tuohy T, Burt RW, Kinney AY. Parental attitudes, beliefs, and perceptions about genetic testing for FAP and colorectal cancer surveillance in minors. Journal of genetic counseling. 2010;19(3):269–279. 23. Kattentidt-Mouravieva AA, den Heijer M, van Kessel I, Wagner A. How harmful is genetic testing for familial adenomatous polyposis (FAP) in young children; the parents’ experience. Familial cancer. 2014;13(3):391–399. 24. Claes E, Denayer L, Evers-Kiebooms G, et al. Predictive testing for hereditary nonpolyposis colorectal cancer: Subjective perception regarding colorectal and endometrial cancer, distress, and health-related behavior at one year post-test. Genetic testing. 2005;9(1):54–65. 25. Domanska K, Nilbert M, Soller M, Silfverberg B, Carlsson C. Discrepancies between estimated and perceived risk of cancer among individuals with hereditary nonpolyposis colorectal cancer. Genetic testing. 2007;11(2):183–186. 26. Heshka JT, Palleschi C, Howley H, Wilson B, Wells PS. A systematic review of perceived risks, psychological and behavioral impacts of genetic testing. Genetics in medicine: official journal of the American College of Medical Genetics. 2008;10(1):19–32. 27. Nusbaum R, Leventhal KG, Hooker GW, et al. Translational genomic research: Protocol development and initial outcomes following SNP testing for colon cancer risk. Translational behavioral medicine. 2013;3(1):17–29. 28. Oliveri S, Ferrari F, Manfrinati A, Pravettoni G. A systematic review of the psychological implications of genetic testing: A comparative analysis among cardiovascular, neurodegenerative and cancer diseases. Frontiers in genetics. 2018;9:624. 29. Turbitt E, Roberts MC, Taber JM, et al. Genetic counseling, genetic testing, and risk perceptions for breast and colorectal cancer: Results from the 2015 National Health Interview Survey. Preventive medicine. 2019;123:12–19. 30. Gritz ER, Peterson SK, Vernon SW, et al. Psychological impact of genetic testing for hereditary nonpolyposis colorectal cancer. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 2005;23(9):1902–1910. 31. Meiser B, Collins V, Warren R, et al. Psychological impact of genetic testing for hereditary non- polyposis colorectal cancer. Clinical genetics. 2004;66(6):502–511. 32. Collins VR, Meiser B, Ukoumunne OC, Gaff C, St John DJ, Halliday JL. The impact of predictive genetic testing for hereditary nonpolyposis colorectal cancer: Three years after testing. Genetics in medicine: official journal of the American College of Medical Genetics. 2007;9(5):290–297. 33. Yanes T, Willis AM, Meiser B, Tucker KM, Best M. Psychosocial and behavioral outcomes of genomic testing in cancer: A systematic review. European journal of human genetics: EJHG. 2019;27(1):28–35. 34. van Oostrom I, Meijers-Heijboer H, Duivenvoorden HJ, et al. Experience of parental cancer in childhood is a risk factor for psychological distress during genetic cancer susceptibility testing. Annals of oncology: official journal of the European Society for Medical Oncology. 2006;17(7):1090–1095. CHAPTER 14 Psychosocial Issues in Genetic Testing for Hereditary Colorectal Cancer 35. Murakami Y, Okamura H, Sugano K, et al. Psychologic distress after disclosure of genetic test results regarding hereditary nonpolyposis colorectal carcinoma. Cancer. 2004;101(2):395–403. 36. Halbert CH, Lynch H, Lynch J, et al. Colon cancer screening practices following genetic testing for hereditary nonpolyposis colon cancer (HNPCC) mutations. Archives of internal medicine. 2004;164(17):1881–1887. 37. Robinson JO, Wynn J, Biesecker B, et al. Psychological outcomes related to exome and genome sequencing result disclosure: A meta-analysis of seven Clinical Sequencing Exploratory Research (CSER) Consortium studies. Genetics in medicine: official journal of the American College of Medical Genetics. 2019;21(12):2781–2790. 38. Solomon I, Harrington E, Hooker G, et al. Lynch syndrome limbo: Patient understanding of variants of uncertain significance. Journal of genetic counseling. 2017;26(4):866–877. 39. Esteban I, Vilaro M, Adrover E, et al. Psychological impact of multigene cancer panel testing in patients with a clinical suspicion of hereditary cancer across Spain. Psycho-oncology. 2018;27(6):1530–1537. 40. Gjone H, Diseth TH, Fausa O, Novik TS, Heiberg A. Familial adenomatous polyposis: Mental health, psychosocial functioning and reactions to genetic risk in adolescents. Clinical genetics. 2011;79(1):35–43. 41. Plumridge G, Metcalfe A, Coad J, Gill P. Parents’ communication with siblings of children affected by an inherited genetic condition. Journal of genetic counseling. 2011;20(4):374–383. 42. Codori AM, Zawacki KL, Petersen GM, et al. Genetic testing for hereditary colorectal cancer in children: long- term psychological effects. American journal of medical genetics Part A. 2003;116a(2):117–128. 43. Douma KF, Bleiker EM, Vasen HF, Gundy CM, Gerritsma MA, Aaronson NK. Psychological distress and quality of life of partners of individuals with familial adenomatous polyposis. Psycho- oncology. 2011;20(2):146–154. 44. Palomaki GE, McClain MR, Melillo S, Hampel HL, Thibodeau SN. EGAPP supplementary evidence review: DNA testing strategies aimed at reducing morbidity and mortality from Lynch syndrome. Genetics in medicine: official journal of the American College of Medical Genetics. 2009;11(1):42–65. 45. Wakefield CE, Meiser B, Homewood J, Ward R, O’Donnell S, Kirk J. Randomized trial of a decision aid for individuals considering genetic testing for hereditary nonpolyposis colorectal cancer risk. Cancer. 2008;113(5):956–965. 46. Mighton C, Carlsson L, Clausen M, et al. Development of patient “profiles” to tailor counseling for incidental genomic sequencing results. European journal of human genetics: EJHG. 2019;27(7):1008–1017. 47. Reumkens K, Tummers MHE, Gietel-Habets JJG, et al. Online decision support for persons having a genetic predisposition to cancer and their partners during reproductive decision-making. Journal of genetic counseling. 2019;28(3):533–542. 48. Stoffel EM, Mercado RC, Kohlmann W, et al. Prevalence and predictors of appropriate colorectal cancer surveillance in Lynch syndrome. American journal of gastroenterology. 2010;105(8):1851–1860. 49. Burton-Chase AM, Hovick SR, Peterson SK, et al. Changes in screening behaviors and attitudes toward screening from pre-test genetic counseling to post-disclosure in Lynch syndrome families. Clinical genetics. 2013;83(3):215–220. 50. Pan JY, Haile RW, Templeton A, et al. Worldwide practice patterns in Lynch syndrome diagnosis and management, based on data from the International Mismatch Repair Consortium. Clinical gastroenterology and hepatology: the official clinical practice journal of the American Gastroenterological Association. 2018;16(12):1901– 1910.e1911. 51. Gaff CL, Clarke AJ, Atkinson P, et al. Process and outcome in communication of genetic information within families: A systematic review. European journal of human genetics: EJHG. 2007;15(10):999–1011. 52. Peterson SK, Watts BG, Koehly LM, et al. How families communicate about HNPCC genetic testing: Findings from a qualitative study. American journal of medical genetics Part C, Seminars in medical genetics. 2003;119c(1):78–86. 53. Petersen J, Koptiuch C, Wu YP, et al. Patterns of family communication and preferred resources for sharing information among families with a Lynch syndrome diagnosis. Patient education and counseling. 2018;101(11):2011–2017. 54. Leenen CH, Heijer M, van der Meer C, Kuipers EJ, van Leerdam ME, Wagner A. Genetic testing for Lynch syndrome: Family communication and motivation. Familial cancer. 2016;15(1):63–73. 55. Menko FH, Ter Stege JA, van der Kolk LE, et al. The uptake of presymptomatic genetic testing in hereditary breast- ovarian cancer and Lynch syndrome: A systematic review of the literature and implications for clinical practice. Familial cancer. 2019;18(1):127–135. 56. Seppala TT, Pylvanainen K, Mecklin JP. Uptake of genetic testing by the children of Lynch syndrome variant carriers across three generations. European journal of human genetics: EJHG. 2017;25(11):1237–1245. 57. Keogh LA, Niven H, Rutstein A, Flander L, Gaff C, Jenkins M. Choosing not to undergo predictive genetic testing for hereditary colorectal cancer syndromes: Expanding our understanding of decliners and declining. Journal of behavioral medicine. 2017;40(4):583–594. 58. Esplen MJ, Harrington S, Leung YW, et al. Telephone versus in- person colorectal cancer risk and screening intervention for first- degree relatives: A randomized controlled trial. Cancer. 2019;125(13):2272–2282. 59. Lynch HT, Snyder C, Stacey M, et al. Communication and technology in genetic counseling for familial cancer. Clinical genetics. 2014;85(3):213–222. 60. Dewanwala A, Chittenden A, Rosenblatt M, et al. Attitudes toward childbearing and prenatal testing in individuals undergoing genetic testing for Lynch syndrome. Familial cancer. 2011;10(3):549–556. 61. Duffour J, Combes A, Crapez E, et al. Reproductive decision- making in MMR mutation carriers after results disclosure: Impact of psychological status in childbearing options. Journal of genetic counseling. 2016;25(3):432–442. 62. Rich TA, Liu M, Etzel CJ, et al. Comparison of attitudes regarding preimplantation genetic diagnosis among patients with hereditary cancer syndromes. Familial cancer. 2014;13(2):291–299. 63. Douma KF, Aaronson NK, Vasen HF, Verhoef S, Gundy CM, Bleiker EM. Attitudes toward genetic testing in childhood and reproductive decision- making for familial adenomatous polyposis. European journal of human genetics: EJHG. 2010;18(2):186–193. 109 15 Psychosocial Issues in Genomic Testing, Including Genomic Testing for Targeted Therapies Megan Best Introduction Cancer may be regarded as the product of cumulative somatic genetic changes, with some arising in individuals who have inherited a germline predisposition to cancer, modified by environmental exposures.1 Genomic testing has now entered clinical practice, driven by improved technology, patient demand, and the need for more effective cancer treatment. At a time when increased costs of clinical research have slowed the development of therapeutic options, advances in genomics offer great promise of improving outcomes in the prevention and treatment of cancer through personalization of medical care tailored to individual genetic status. Traditionally, tumors have been named after the organ in which they developed and categorized on the basis of histology (microscopic anatomy). However, these methods do not reflect the complicated underlying molecular events that drive the cancer process. By surveying thousands of genes at once, using DNA arrays (genomic tests), it is now possible to read the molecular signature of an individual patient’s tumor as well as their whole genome. Analysis of tumor molecular signatures has revealed new classes of cancer that go beyond distinctions based on histological appearance alone.2 This allows oncologists to offer personalized treatment for cancer, potentially identifying new cancer therapies for selected patients who are likely to respond. At the same time, it may be possible to reduce unnecessary side effects and toxicity by avoiding unnecessary treatments in patients who will not benefit. It also allows for identification of those who are, or are not, at increased risk of developing a particular cancer, allowing for personalized prevention strategies. However, for this promise to be fully realized, patients need to cope with the testing process and act on the results. The process of genomic testing can be highly complex. The human genome contains approximately 23,000 genes. Genomic testing always involves examination of multiple genes. This may be a “panel” of whole genes or single nucleotide polymorphisms (SNPs) (changes in nucleotides within genes), the whole exome (the part of the genome that codes for proteins), or the whole genome. Testing can assess genes in the germline or somatic changes in the individual cancer. The psychological impact of single gene testing has been well studied. However, there are important differences between both germline and somatic genomics and single gene testing that have implications for psychological outcomes, making it difficult to assume that response to single gene results will generalize to the genomic setting. Complexity Single gene testing involves examining a gene of known significance, so that the result will be either positive or negative for a particular variant. Genomic testing, in contrast, can deliver a number of different outcomes, and in some cases can represent an unprecedented volume of results to process. Germline testing is performed on a sample of “normal” cells and can reveal pathogenic variants (which may have also been inherited by blood relatives) that are (1) relevant to the target cancer and clinically actionable, guiding risk prevention; (2) relevant to the target cancer but nonclinically actionable (no proven treatments); (3) secondary (relevant to other cancers and diseases) and clinically actionable, guiding risk prevention; (4) secondary and nonclinically actionable (no proven treatments); or (5) of unknown or uncertain significance (variants of unknown significance [VUS]). Patients with pathogenic germline variants that signal high risk of a particular disease can be offered more intensive risk management strategies. Somatic testing involves genomic testing of tumor (either fresh or archived tissue or liquid biopsy) for patients who have solid tumors such as lung, colon, breast, kidney, and liver cancers. It is commonly used in two ways. The first is to assess patient prognosis to guide treatment choices. This is known as risk for recurrence (RFR) testing. The second is to identify biomarker genes that can be linked to specific treatments directed against the genetic mutations in the tumor. It is increasingly clear that there are molecularly distinct subtypes of various common cancers, with different therapeutic approaches required for each subtype. CHAPTER 15 Psychosocial Issues in Genomic Testing, Including Genomic Testing for Targeted Therapies A number of molecular targets have been identified, with ongoing enrollment of patients in clinical trials to assess predictive biomarkers of efficacy. This type of test is known as molecular tumor profiling (MTP). MTP can identify (1) somatic variants (relevant only to the patient) that are clinically actionable (guiding treatment) and treatment may or may not be accessible to the patient; (2) somatic variants that are not clinically actionable (no proven treatments); (3) no somatic variants; or (4) genes of unknown significance (GUS) (genes known to be a biomarker for a different cancer to the patient). There is a small chance that pathogenic variants identified in the tumor by MTP may have germline (inherited) origin. If suspected, further testing is required for confirmation. These findings have implications for blood relatives. Uncertainty Although uncertainty pervades medical information, its scope in genomics may be unprecedented. The taxonomy of medical uncertainty of Han et al.3 identifies three principal sources of uncertainty: indeterminate outcomes (probability), imprecise risk estimates, and complexity, all of which are relevant in genomics. Genomic testing is technically complex through all its steps, from sample acquisition, to analysis, to clinical report generation and interpretation, to communication of results to the patient—each of which introduces uncertainty regarding, for example, the accuracy and reliability of test results. The clinical uses of genomic testing introduce other uncertainties regarding the benefits and harms of genomic test information, the optimal strategies for communicating this information to patients, and the consequences of genomic testing for patients, family members, the health care system, and society.4 Practitioners who obtain consent to genomic testing from patients face the challenge of conveying these uncertainties to ensure informed choice and mitigate unrealistic expectations, while patients must absorb and cope with the information both before and after testing. Germline results, whether identified by somatic or germline testing, have implications for future health and reproductive choice, and communication within families may be required to ensure everyone potentially affected is aware of their risk. Findings of unknown/uncertain significance, whose meaning may or may not become clearer over time, can be confusing and worrying to patients and physicians. In addition, results may be both diagnostic (confirming the subtype of cancer) and predictive (indicating future cancer risk), which have traditionally had different ethical norms guiding practice. While patient autonomy and shared decision-making are established standards in health care, there are currently no guidelines on what information patients should be told, nor how uncertainty should be approached, in genomic testing. The current evidence base primarily reports on the general or noncancer population, where the potential benefits of a positive result (such as access to novel therapeutic options) will differ. Mainstreaming Oncologists were among the first clinicians to incorporate tumor genomics into routine management to guide treatment choice (e.g., the KRAS gene in colorectal cancer, the BRAF gene in melanoma).5 Given the rapidly falling costs of all types of genomic tests and the increasing number of targetable variants, it is likely that oncologists will exponentially increase the number of genomic tests ordered. These factors have paved the way to mainstreaming of genomic testing. Mainstreaming refers to genomic testing ordered by non- genetics-trained health professionals. This enables higher uptake of appropriate germline genetic testing in cancer patients at suspected risk for hereditary cancer as well as RFR and MTP to guide therapy. Although the use of MTP to discover novel therapy appears to be well understood and accepted by patients,6 there are significant challenges involved with moving genomic testing into mainstream medicine. First, patient knowledge of other types of genomic testing appears to be low.7 Second, genomics is a rapidly evolving area where it is also difficult for practitioners to stay up to date with interpretation of results and therapeutic options. Third, the potential for gene changes such as germline, secondary, and VUS/GUS means that both patients and families may be confronted with information they had not been seeking nor were prepared to face; oncologists are not used to dealing with healthy family members in relation to a cancer patient’s test results. Furthermore, genomic test results can be difficult for nongenetically trained staff to interpret. This situation requires a new skill set for oncologists. There is ample evidence that nongenetics health professionals have deficits in genetic literacy, which may limit their ability to explain genomic testing results to their patients (and their families) effectively. Nongenetics health professionals report difficulties in family history taking and risk communication and have been shown to misinterpret test results, possibly leading to incorrect management.8 The American Society of Clinical Oncology identified the need to assist oncologists in the task of adopting genomic medicine as a matter of priority.9 In the short term, there is a shortfall of knowledge and expertise in communication within the oncology community with regard to supporting cancer patients undergoing testing, which further increases the chance of psychological risk. Psychological Issues in Genomic Testing Research on the psychosocial impact of genomic testing so far has been biased toward breast cancer and Caucasian women with high socioeconomic status and education,7 and internationally, genomic datasets are dominated by Caucasian populations. As a result, VUS will be reported more frequently in ethnic minority patients, who may also have lower health literacy, and therefore may represent a group at higher risk of psychological sequelae to genomic testing. Germline Testing Knowledge and Understanding There is no relevant validated measure of genomic knowledge that allows comparison across studies. The Knowledge of Genome Sequencing (KOGS) is currently being validated in a cancer cohort for the first time.10 A recent review7 showed that knowledge of germline panel testing is sometimes poor but increases significantly after counseling, particularly knowledge of the limitations of testing. In one study included in the review, of patients with a family history of cancer, higher knowledge was associated with lower avoidance of receiving genomic results and higher decisional satisfaction. However, the study also reported that a minority of patients misunderstood test results, with VUS findings interpreted by 4% as 111 112 Section III Screening and Testing for Germ Line and Somatic Mutations equivalent to a negative result, and 20% of patients not realizing that a positive result indicated increased cancer risk. In two studies of participants undergoing SNP testing included in the review, recall of test results up to 5 months was accurate but had no impact on risk perception. Risk perception was often shaped by family or personal history of cancer and did not change after counseling. There were no changes from baseline to after receiving results in perceptions of heredity or perceived risk of finding a heritable cause for cancer, and expectations of heredity were not associated with distress, coping style, or illness perception.7 Another hypothetical study asking primary care patients about SNP testing for colon cancer risk found that most people preferred information about the test to be delivered in person by a health care professional rather than by written information.11 Psychological Impact Several studies have shown no impact of germline testing on generalized anxiety, depression, or cancer-specific anxiety between baseline and follow-up.7 However, another study that compared groups according to whether or not they were affected by cancer, and whether or not they were positive for a pathogenic variant, found that unaffected patients with a family history of breast or ovarian cancer who received positive results had significantly higher levels of intrusive thoughts, avoidance, and distress.7 Intermediate levels of some measures of distress were reported among unaffected patients with VUS results. In this study, distress was higher in those with lower knowledge scores and among African Americans compared with Caucasians. Qualitative studies of participants undergoing SNP testing reported emotional responses such as reassurance, surprise, or disappointment when results did not match family or personal history of cancer.7 Measurement of illness perception between baseline and follow- up indicated no impact from germline testing in one study.7 The psychological impact of receiving genomic test results can be measured by the Multidimensional Impact of Cancer Risk Assessment (MICRA) questionnaire and consists of three subscales: test- related distress, positive experience, and uncertainty. It was used in two studies.7 One showed no change and the other found higher distress scores in unaffected participants with a pathogenic variant or VUS finding, compared to others defined by cancer status and variant status. Distress was higher among African Americans and those of younger age. In the cohort characterized by cancer status and test results, test-related positive experience was lowest among the no cancer/VUS and the cancer/pathogenic variant–positive groups. Lower positive experience was associated with younger age. Receipt of results had no impact on uncertainty in two studies, and intolerance for ambiguity does not appear to impact on coping ability.7 In one study, no association was found between ambiguity tolerance and the MICRA, avoidance and distress, or satisfaction with decision-making.7 Decisional satisfaction and satisfaction with genetic services were high and not altered by counseling, test results, or cancer status in three studies.7 Satisfaction was reduced with lower knowledge, lower levels of education, younger age, and Asian and Hispanic race compared to Caucasian. Satisfaction with the decision to undergo testing decreased over time since testing. Regret was uncommon, and dissatisfaction was not related to the testing itself, but the long wait for results.7 One hypothetical study found that cancer patients valued genomic results over standard test results.7 In qualitative studies, patients have expressed concern about the psychological burden of risk information, and insurance and privacy concerns. Reported concerns about discrimination are uncommon.7 Behavioral Impact Investigation so far suggests that people are more likely to use their germline panel test results in decision-making if they have a positive result, in the form of taking up additional screening among people currently unaffected by cancer and making treatment choices among those diagnosed with cancer. However, in one study, some people with a VUS or negative result, which does not necessarily indicate no risk, also acted on their results.7 For those undergoing SNP testing, there is limited evidence that it can independently predict behavioral change. Suggested factors associated with behavioral change as a result of the testing process include education during counseling and patient anxiety.7 Family communication has been studied for those undergoing SNP germline testing. Two studies found that sharing of results with relatives was motivated by perceived “shared risk” or with the goal of motivating health behavior change.7 Patients with high-risk results were more likely to report the result to their health care professional compared to those with average or low risk.7 One study examining perceived utility of germline genomic testing showed no significant impact, although in that study, a change in management as a result of testing occurred for a small minority only.7 Somatic Testing By definition, all somatic testing is performed on patients with a cancer diagnosis. Early research demonstrated that cancer patients had positive attitudes and expectations of genomic testing to assist in guiding therapy, although those who have a strong interest are likely to overestimate its potential benefit.7,12 A recent review7 found that somatic RFR testing (used to guide chemotherapy uptake in early cancer) may be associated with negative psychosocial outcomes. Few studies have described cancer patients’ views of MTP. Much of the scant research in this area to date has been hypothetical or conducted on noncancer patients.13 Knowledge and Understanding From this review, it is seen that generally patient knowledge of genomics is low, although most participants in these studies who were undergoing RFR testing understood that the results could aid chemotherapy decisions. Fewer understood that the test result indicated the future chance of metastasis. In two North American studies, the majority of patients did not understand terms such as “precision medicine” or “personalized medicine.”13,14 Even among patients with higher health literacy, less than 40% reported familiarity with terms like “personalized medicine,” “genomics,” and “biomarkers.”14 Another study of advanced cancer patients found a median of 75% knowledge questions correctly answered.15 Qualitative studies suggested that patient misunderstanding about RFR somatic testing prompted increased anxiety. Participants with higher knowledge scores were more likely to prefer an active role in decision-making, have fewer concerns about testing, be highly educated, be younger, be diagnosed more recently, have less comorbidity, have higher CHAPTER 15 Psychosocial Issues in Genomic Testing, Including Genomic Testing for Targeted Therapies income, be more likely to be employed full time, have relatives who previously underwent chemotherapy, and be of Caucasian ancestry. Inadequacies in knowledge pose barriers to the adoption of testing. For example, one survey of patients with advanced cancer found that less than half (48%) felt they had sufficient knowledge of benefits and risks to make an informed decision about whether to pursue genomic testing, and 34% of patients wanted formal genetic counseling before participating in MTP.15 One study of early-stage breast cancer patients in the review found that 62% of their cohort sought information about their results, and those who sought information were significantly more knowledgeable. Perceived risk was significantly reduced after RFR somatic testing and correlated with actual risk scores of recurrence posttest. Previous studies have documented confusion among patients regarding whether the results refer to the identification of germline or somatic mutations (i.e., results that may be relevant to blood relatives versus those that pertain to the patient only).13 It is not known how communication about test results can be best tailored to improve understanding of this distinction. In view of the finding that self-perceived deficiencies in understanding represent a barrier to test utilization, it has been recommended that education and information tools be developed to support patients undergoing somatic tumor screening.7,13,15 Those undergoing somatic testing have also expressed a desire for professional support in the event of communicating unsolicited genetic information to family members.16 This underscores the importance of providing adequate information and counseling before results are returned. Written information has previously been described as reducing patient anxiety and assisting patients in returning genetic information to their family.17 Psychological Impact A study included in the review showed that state anxiety for RFR testing decreased 12 months after receiving results and was positively correlated with decisional conflict both before and after testing.7 Increased anxiety was also associated with receiving results, a delay in return of results, or misconceptions about the meaning of the results. In addition, increased anxiety was associated with more cancer-related distress, higher actual recurrence risk, inaccessible results, and discordance between the clinical recurrence score and the genomic result. One author found high levels of cancer-specific distress in over one-third (38.7%) of RFR participants. Higher perceived RFR was associated with lower satisfaction, higher levels of worry, and Caucasian ancestry. In several studies, women who prefer a passive role in decision-making and who received intermediate- recurrence risk results had higher cancer-related distress, but not worry about recurrence.7 Targeted treatment is an area of rapid development, and at present the number of patients matched to targeted treatment, and the number of targeted treatments available to patients, is small. This is a reflection of the challenge of accessing novel drugs for patients with cancer, as well as the need for more research to identify anticancer therapies that target identified tumor biomarkers. Care should be taken to manage patient expectations. Patients have also reported some concerns, including worries about psychological harm related to intrusive thoughts due to unwanted knowledge.7 It is possible that patients with a cancer diagnosis may hold high hopes for MTP to provide new treatments and feel disappointed if no actionable result is found, even if they had been advised that the chances of a useful outcome were low. Equally, if clinically actionable results are found and the relevant drug is not available to the patient, or they are deemed too unwell for treatment, they may feel angry and abandoned.12 Attitudes The recent review found that most patients undergoing RFR thought that the test results were accurate and useful. Reported benefits of undergoing somatic testing included the ability to reduce uncertainty and improve treatment options, thus empowering the participants. Overestimation of the validity of the tests increased its value.7 In hypothetical scenarios, patients are generally willing to undergo testing with the belief that genetic information about their cancer will improve their care.13,15,18 However, participants also attributed advantages to somatic testing that are normally attributed to germline testing, such as cancer prevention and motivation for behavior change.13 Most patients undergoing RFR said they would have it again and recommend it to others. One study found that decisional conflict lessened after results were received, and satisfaction with decision persisted to follow-up one year later.7 Reported drawbacks of somatic testing in hypothetical studies included learning things about the cancer that were better left unknown, as well as concerns over psychological harm and test accuracy.13 In the case of targeted treatment, drawbacks included the risk of disappointment from a negative result or a positive result but no access to the relevant treatment.7 Concerns were increased in participants with a higher perceived risk of recurrence.7 In deciding to undergo MTP, the primary motive for advanced cancer patients was personal benefit. Seen as cutting-edge technology, MTP was perceived as a source of hope, offering promises of targeted and therefore more effective therapy.12,15 Studies in cancer patients who have actually been offered MTP have primarily noted unequivocal patient acceptance, but also reports of information overload and misunderstanding, causing unrealistic expectations, anxiety, and uncertainty.6,7,16,19 Self-perceived lack of knowledge did not reduce willingness to participate. Patient hopes of benefit were enhanced by the promise of novel and targeted treatment but challenged by nonfindings or by limited access to relevant trials.12 In hypothetical studies, cancer patients have identified disadvantages of MTP such as disclosure of unwanted information or that reporting of results could counteract denial. Concerns over psychological harm and test accuracy were also noted.13 Studies of cancer patients have identified differences between racial groups. One study reported that, even if testing was financially covered, nonwhite participants were less willing to undergo MTP for the selection of approved drugs or experimental drugs.18 Another reported that concerns over psychological harm were higher among blacks as compared with whites.13 The most important factors that might discourage MTP were the potential for complications if a biopsy were required and a delay in treatment while waiting for results.15 While not a major concern, cancer patients have listed concerns about privacy and confidentiality of test results and potential for test results to lead to health, life, or disability insurance discrimination as reasons not to pursue somatic genomic testing for cancer.13,15 Insurance concerns appear greater in countries with private health care systems, and it is noted that several countries have passed 113 114 Section III Screening and Testing for Germ Line and Somatic Mutations legislation to prevent discrimination based on genetic testing. Cost has also been listed as a disadvantage.13,15 Preferences for Receipt of Results Most participants want to be informed of results that had implications for treatment with discussion focused on practical issues relevant to treatment.12,18 In one study 35% of patients only wanted to receive MTP results that would directly influence their own treatment decisions.15 For cancer patients undergoing MTP, family obligations informed a willingness to receive information about germline results (a rare outcome, but which can occur). This information tended to be perceived as burdensome given the advanced disease of the patient and the challenges of coping with illness. Cancer risk information could also be perceived as irrelevant for a patient with a short prognosis.7,12 Possibly reflecting this ambivalence was the finding that in a small sample of gastrointestinal cancer patients undergoing somatic genome testing, one of eight patients decided not to go ahead with tests to confirm a germline finding following genetic counseling, therefore leaving the implications for blood relatives unknown.20 Thus, although the result is rare, somatic genomic sequencing must be accompanied by a plan for return of germline results, in partnership with genetic counseling. This information will be important for any blood relatives who wish to undergo germline testing themselves, as the target variant (index case) for that particular family will then be known, and test utility will increase as a result.21 Behavioral Impact Little attention has been given to communication of germline results in somatic testing. Somatic RFR results and the level of risk both influenced chemotherapy decisions in the majority of cases, though the most influential factor for some patients remained the doctor’s opinion.7 In a qualitative hypothetical scenario, one study found that almost all cancer patients undergoing somatic genome testing had a positive attitude toward receiving unsolicited findings from MTP.16 However, after receiving information about the types of unsolicited findings and possible psychological, social, and financial impacts, most participants wanted to receive only subsets of genetic information. Their main concern was their own and others’ (including family members) ability to cope with the increased risk of having a genetic disorder and the anticipated emotional burden. Even when family members were happy for the participant to receive all possible information, the gatekeeper role persisted. Future Directions Genomic testing in cancer can identify germline and somatic genetic changes that have implications for the prevention, diagnosis, and treatment of cancer. The growing understanding of the molecular basis of disease is revolutionizing cancer care by allowing the personalization of patient management. However, challenges in the testing process include its innate complexity, the uncertainty it can generate, and the increased demands on oncology staff due to the mainstreaming of genetics. Current understanding of patient responses to single gene testing may not translate to the genomic sphere. Research is in its early stages, but early reports of poor patient understanding of genomic testing, mixed results regarding distress from testing, and variable impact of results on patient behavior indicate the need for further investigation. Future research directions should include a better understanding of what information should be included in the consent process, how patients can be best supported in communication of germline findings to relevant family members, and ways to motivate individuals to adopt preventative strategies to reduce their risk of developing cancer. Due to the current bias in research to female Caucasians, a broader range of cancers and populations should be included in future studies. In this way the full benefits of genomic oncology may be realized. REFERENCES 1. Lichtenstein P, Holm NV, Verkasalo PK, Iliadou A, Kaprio J, Koskenvuo M, et al. Environmental and heritable factors in the causation of cancer—analyses of cohorts of twins from Sweden, Denmark, and Finland. New England Journal of Medicine. 2000;343(2):78–85. 2. Liotta L, Petricoin E. Molecular profiling of human cancer. Nature Reviews Genetics. 2000;1(1):48–56. 3. Han PKJ, Klein WMP, Arora NK. Varieties of uncertainty in health care. Medical Decision Making. 2011;31(6):828–838. 4. Han P, Umstead K, Bernhardt B, Green R, Joffe S, Koenig BA, et al. A taxonomy of medical uncertainties in clinical genome sequencing. Genetics in Medicine. 2017;19(8):918. 5. Allegra CJ, Jessup JM, Somerfield MR, Hamilton SR, Hammond EH, Hayes DF, et al. American Society of Clinical Oncology provisional clinical opinion: testing for KRAS gene mutations in patients with metastatic colorectal carcinoma to predict response to anti–epidermal growth factor receptor monoclonal antibody therapy. Journal of Clinical Oncology. 2009;27(12):2091–2096. 6. Liang R, Meiser B, Smith S, Kasparian N, Lewis C, Chin M, et al. Advanced cancer patients’ attitudes towards, and experiences with, screening for somatic mutations in tumours: a qualitative study. European Journal of Cancer Care. 2017;26(6):10.1111/ ecc.12600. 7. Yanes T, Willis AM, Meiser B, Tucker KM, Best M. Psychosocial and behavioral outcomes of genomic testing in cancer: a systematic review. European Journal of Human Genetics. 2018;27(1):28–35. 8. Ha VTD, Frizzo-Barker J, Chow-White P. Adopting clinical genomics: a systematic review of genomic literacy among physicians in cancer care. BMC Medical Genomics. 2018;11(1):18. 9. Robson ME, Storm CD, Weitzel J, Wollins DS, Offit K. American Society of Clinical Oncology policy statement update: genetic and genomic testing for cancer susceptibility. Journal of Clinical Oncology. 2010;28(5):893–901. 10. Sanderson SC, Loe BS, Freeman M, Gabriel C, Stevenson DC, Gibbons C, et al. Development of the Knowledge of Genome Sequencing (KOGS) questionnaire. Patient Education and Counseling. 2018;101(11):1966–1972. 11. Leventhal K-G, Tuong W, Peshkin BN, Salehizadeh Y, Fishman MB, Eggly S, et al. “Is it really worth it to get tested?”: primary care patients’ impressions of predictive SNP testing for colon cancer. Journal of Genetic Counseling. 2013;22(1):138–151. 12. Best MC, Bartley N, Jacobs C, Juraskova I, Goldstein D, Newson AJ, et al. Patient perspectives on molecular tumor profiling: “why wouldn’t you?” BMC Cancer. 2019;19(1):753. CHAPTER 15 Psychosocial Issues in Genomic Testing, Including Genomic Testing for Targeted Therapies 13. Gray SW, Hicks-Courant K, Lathan CS, Garraway L, Park ER, Weeks JC. Attitudes of patients with cancer about personalized medicine and somatic genetic testing. Journal of Oncology Practice. 2012. doi:JOP.2012.000626. 14. Williams JR, Yeh VM, Bruce MA, Szetela C, Ukoli F, Wilkins CH, et al. Precision medicine: familiarity, perceived health drivers, and genetic testing considerations across health literacy levels in a diverse sample. Journal of Genetic Counseling. 2019;28(1):59–69. 15. Blanchette PS, Spreafico A, Miller FA, Chan K, Bytautas J, Kang S, et al. Genomic testing in cancer: patient knowledge, attitudes, and expectations. Cancer. 2014;120(19):3066–3073. 16. Bijlsma RM, Wessels H, Wouters RHP, May AM, Ausems MGEM, Voest EE, et al. Cancer patients’ intentions towards receiving unsolicited genetic information obtained using next- generation sequencing. Familial Cancer. 2018;17(2):309–316. 17. Hallowell N, Murton F. The value of written summaries of genetic consultations. Patient Education and Counseling. 1998;35(1):27–34. 18. Yusuf RA, Rogith D, Hovick SR, Peterson SK, Burton-Chase AM, Fellman BM, et al. Attitudes toward molecular testing for personalized cancer therapy. Cancer. 2015;121(2):243–250. 19. Schrader KA, Cheng DT, Joseph V, Prasad M, Walsh M, Zehir A, et al. Germline variants in targeted tumor sequencing using matched normal DNA. JAMA Oncology. 2016;2(1):104–111. 20. Catenacci DVT, Amico AL, Nielsen SM, Geynisman DM, Rambo B, Carey GB, et al. Tumor genome analysis includes germline genome: are we ready for surprises? International Journal of Cancer. 2015;136(7):1559–1567. 21. Morrow A, Jacobs C, Best M, Greening S, Tucker K. Genetics in palliative oncology: a missing agenda? A review of the literature and future directions. Supportive Care in Cancer. 2018;26(3):721–730. 115 16 Psychosocial Issues Related to Liquid Biopsy for ctDNA in Individuals at Normal and Elevated Risk Jada G. Hamilton, Amanda Watsula-Morley, and Alicia Latham Introduction With the advent of next-generation sequencing (NGS) and improved understanding of the genomic changes underlying cancer initiation and progression, there is increasing interest in the potential for liquid biopsy. Liquid biopsy involves the minimally invasive sampling of blood or plasma to detect biomarkers indicative of cancer. Liquid biopsy can detect cell-free DNA (cfDNA), defined as small fragments of nucleic acid found in the peripheral circulation. In cancer patients, a percentage (estimated at less than 1% in early-stage disease and up to 40% in advanced disease) of this unbound nucleic acid is specific to and derives from the patient’s dying tumor cells and is referred to as circulating tumor DNA (ctDNA).1,2 Analyzing genetic alterations (i.e., single base substitutions, translocations, insertions, and deletions) in ctDNA through NGS platforms has recently emerged as a potential method to monitor response to oncological treatment and assess for early disease recurrence. Higher levels of ctDNA are generally detected in metastatic disease and specific tumor types, as certain cancers are more prone to ctDNA shedding.2,3 Background Clinical Applications Analysis of ctDNA has the potential for application in multiple contexts and populations. One primary context is in the management of patients who are already diagnosed with cancer. For instance, ctDNA assays may be useful for informing treatment selection in patients with advanced cancers, consistent with the goals of precision medicine. A few Food and Drug Administration (FDA)-approved ctDNA assays exist, including the Cobas EGFR Mutation Test v2 that detects variants in the epidermal growth factor receptor (EGFR) gene among patients with metastatic non–small cell lung cancer, and the therascreen PIK3CA assay that detects variants in the PIK3CA gene among patients with metastatic breast cancer.4 In these cases, ctDNA is analyzed to identify known somatic (i.e., tumor) gene variants that can be targeted with specific therapeutics. There is hope that ctDNA can be used to inform decisions about adjuvant therapy for patients with cancer following primary surgery or radiation treatment, because it is not always clear how to best treat patients who lack evidence of residual or metastatic disease.5 For example, there is some retrospective evidence to suggest that analysis of ctDNA as a marker of residual disease could offer insight into whether such patients would benefit from adjuvant therapy.5–7 Similarly, ctDNA analysis could ultimately provide earlier warning about relapse or disease recurrence among cancer survivors than other existing surveillance methods.5,7 An additional promising context for ctDNA analysis is as a cancer screening tool among healthy individuals. The prospect of having a blood-based, minimally invasive test that could be widely deployed in the population to detect cancer at its earliest stages is extremely appealing. Such a test would be a powerful tool for reducing cancer morbidity and mortality in the general population. However, to achieve this goal, a ctDNA-based screening assay would need to both be cost-effective and have extremely high specificity—that is, the test would need to have a high probability of classifying those without cancer as negative, as well as return few false-positive results.5 Benefits Analysis of ctDNA has a number of important advantages when compared to traditional biopsy of solid tumor tissue. For one, given that ctDNA assays only require a blood sample, this approach is much less invasive and burdensome to the patient and has the potential to be used in cases where accessing an adequate tissue sample is difficult. Analysis of ctDNA may also provide information about the spectrum of genetic changes (i.e., genetic heterogeneity) that exist within a given tumor or among multiple tumor sites (e.g., metastases) within a patient, the entire scope of which may be missed with the biopsy of tumor tissue alone. In addition, ctDNA can be more easily and economically sampled serially over time, and could therefore provide improved longitudinal information about how a patient’s CHAPTER 16 Psychosocial Issues Related to Liquid Biopsy for ctDNA in Individuals at Normal and Elevated Risk disease responds to treatment. This stands in contrast to serial imaging studies that have multiple risks including radiation and heavy- metal exposure in many high-resolution contrasted studies.4,6,8 Limitations Despite the exciting promise of ctDNA assays for use among both cancer-affected and unaffected populations, these tests currently have crucial limitations. For most applications, evidence is lacking to support the clinical validity or clinical utility of these tests. Clinical validity is the ability of a test to accurately detect or predict the clinical disorder of interest. Clinical utility is the ability of a test to significantly improve measurable patient clinical outcomes including morbidity and mortality.9,10 A 2018 review conducted by the American Society of Clinical Oncology and the College of American Pathologists concluded that except for specific ctDNA assays that have received regulatory approval, such as those used in a few types of advanced cancer, currently most ctDNA tests have insufficient evidence to demonstrate clinical validity, and most have no evidence to support clinical utility.6 Therefore, presently there is insufficient evidence to demonstrate the clinical validity or clinical utility of ctDNA tests in most advanced cancer settings, as well as in early-stage cancer, treatment monitoring, and detecting residual disease, or for cancer screening in asymptomatic individuals.6 Consequently, there is a serious need for additional research including well-designed clinical trials to confirm the anticipated clinical benefits of ctDNA assays. Psychosocial Considerations As future research seeks to establish the clinical utility of ctDNA assays, psychosocial outcomes should be included in the balance of risks and benefits of these tests. Although virtually unexplored empirically at this time, there are notable potential psychosocial implications of ctDNA assays. Incidental Discovery of Germline Variants One relevant consideration arises from the fact that it is not currently possible to separate ctDNA at the time of testing from other circulating DNA within a patient.4 Thus, efforts to sequence ctDNA will invariably involve analyzing normal (i.e., germline or heritable) DNA as well. Distinguishing somatic variants from germline variants is not always easily achieved, and this process has the potential to lead to the incidental detection of disease-predisposing germline DNA variants.6 For example, a variant in the gene BRCA1, which is associated with hereditary breast and ovarian cancer, or in the gene TP53, which is associated with Li-Fraumeni syndrome and elevated risks for breast cancer, brain cancer, sarcomas, and other cancers, may be detected and require additional clinical testing to determine if it arises from the tumor or the germline. If such a variant is found to be germline in origin, this can have far-reaching medical and psychosocial implications for patients and their families because it reveals important information about their future cancer risks.11,12 Overtreatment and Overdiagnosis Additional psychosocial considerations relate to the prospect of false-positive results arising from ctDNA assays, particularly in the context of screening healthy, asymptomatic individuals.6 If a ctDNA assay provides false-positive results, then the potential exists for engendering substantial emotional distress among individuals, as well as leading to unnecessary overtreatment with physical, psychological, and financial costs of whichever medical interventions are subsequently adopted. A ctDNA test could also detect genetic changes that would never actually develop into cancer; such overdiagnosis, or the diagnosis of a medical problem that would have not ultimately caused harm, is a risk that has been recognized in other cancer screening settings such as with mammography in breast cancer and prostate-specific antigen (PSA) testing in prostate cancer.6,13,14 Nonetheless, many individuals do not fully understand the risks of overdiagnosis,15,16 which can complicate communication about the harms and benefits of screening tests and present challenges in promoting patients’ informed medical decision making about test adoption. Uncertainty and Anxiety Even if ctDNA assays are developed with acceptable levels of sensitivity and specificity, these tests will not be perfect in their classification of individuals as cancer-affected versus unaffected, and the possibility exists for medical uncertainty, confusion, and anxiety on behalf of patients as well as their healthcare providers. For example, ctDNA assays may accurately detect a relevant genetic change but will likely be unable to provide insight into where a tumor is located within the body;2 if a developing tumor cannot be detected with additional clinical work-up (e.g., imaging), how should a patient with an abnormal ctDNA result be treated and followed? How will this ambiguity be effectively communicated to a patient in a way that balances the potentially conflicting goals of ensuring adequate comprehension, minimizing anxiety, and sustaining motivation to participate in what may be a protracted diagnostic process? Uncertainty can be a substantial source of stress, and patients can experience a variety of negative emotional, cognitive, and behavioral responses to medical uncertainty;17 determining how to manage and minimize these negatives outcomes of the uncertainty that will arise from ctDNA assays represents an important area for future investigation. Quality of Life It is also unclear what the impact on overall quality of life will be for a patient who learns of their risk of cancer development, progression, or recurrence through a ctDNA assay that does not align with other clinical tests or symptom experiences; these patients may need to undergo long-term, repeated surveillance without clear guidance for their medical management. Larger questions remain about how ctDNA as an early marker of cancer may have implications for insurability or discrimination, or how equitable access to this potential innovation in cancer control will be achieved so that existing health disparities are minimized rather than exacerbated. These issues are similar to some of the ethical, legal, and social concerns that have been highlighted, and remain largely unresolved, in the setting of genetic testing for disease predisposition.18–20 Future Directions Using ctDNA as a biomarker detectable through liquid biopsy has great promise for improving disease outcomes among patients affected by cancer, as well as healthy individuals who could benefit from a novel cancer screening test. However, despite substantial 117 118 Section III Screening and Testing for Germ Line and Somatic Mutations enthusiasm for ctDNA assays, evidence is generally lacking for the clinical validity and clinical utility of these tests in many settings.6 Carefully designed clinical trials are required to provide empiric support for the highly anticipated benefits of ctDNA assays. Future research is needed to clarify the benefits and risks of these tests not only in terms of cancer morbidity and mortality but also to address the difficult questions regarding potential psychosocial effects of ctDNA assays that currently lack clear answers. REFERENCES 1. Pantel K, Alix-Panabieres C. Real-time liquid biopsy in cancer patients: Fact or fiction? Cancer Res. Nov 1 2013;73(21):6384–6388. 2. Bettegowda C, Sausen M, Leary RJ, et al. Detection of circulating tumor DNA in early-and late-stage human malignancies. Sci Transl Med. Feb 19 2014;6(224):224ra224. 3. Vogelstein B, Papadopoulos N, Velculescu VE, Zhou S, Diaz LA, Jr., Kinzler KW. Cancer genome landscapes. Science. Mar 29 2013;339(6127):1546–1558. 4. Snow A, Chen D, Lang JE. The current status of the clinical utility of liquid biopsies in cancer. Expert Rev Mol Diagn. Nov 2019;19(11):1031–1041. 5. Mattox AK, Bettegowda C, Zhou S, Papadopoulos N, Kinzler KW, Vogelstein B. Applications of liquid biopsies for cancer. Sci Transl Med. 2019;11(507):eaay1984. 6. Merker JD, Oxnard GR, Compton C, et al. Circulating tumor DNA analysis in patients with cancer: American Society of Clinical Oncology and College of American Pathologists joint review. J Clin Oncol. Jun 1 2018;36(16):1631–1641. 7. Tie J, Wang Y, Tomasetti C, et al. Circulating tumor DNA analysis detects minimal residual disease and predicts recurrence in patients with stage II colon cancer. Sci Transl Med. Jul 6 2016;8(346):346ra392. 8. Neumann MHD, Bender S, Krahn T, Schlange T. ctDNA and CTCs in liquid biopsy—Current status and where we need to progress. Comput Struct Biotechnol J. 2018;16:190–195. 9. Teutsch SM, Bradley LA, Palomaki GE, et al. The Evaluation of Genomic Applications in Practice and Prevention (EGAPP) SECTION IV Screening and Assessment in Psychosocial Oncology Wendy W. T. Lam (Section Editor) 17 Screening and Assessment for Distress 121 Alex J. Mitchell 18 Assessment, Screening, and Case Finding for Depression and Anxiety in People with Cancer 130 Kristine A. Donovan and Paul B. Jacobsen 19 Screening for Delirium and Dementia in the Cancer Patient 137 Christian Bjerre-Real, James C. Root, Yesne Alici, Julia A. Kearney, and William S. Breitbart 20 Screening and Assessment for Cognitive Problems 146 Alexandra M. Gaynor, James C. Root, Elizabeth Ryan, and Tim A. Ahles 17 Screening and Assessment for Distress Alex J. Mitchell Introduction Distress is a very common and clinically relevant complication of cancer that can occur at any time on the patient pathway.1 The United States National Comprehensive Cancer Network (NCCN) recommends screening patients at the initial visit soon after diagnosis and at each visit, although the screening schedule may be revised as clinically indicated (Box 17.1).2 The exact prevalence of distress is difficult to summarize precisely because it is influenced by numerous variables. However, large datasets show that distress is highest within the first 3 months of a cancer diagnosis and decreases within the first year in most patients (see Figure 17.1). Whereas 8% of the general population and 20% of primary care attendees experience distress at any given time, 35% to 80% of cancer patients report distress when they are asked.3–5 Scores from 10,000 patients’ scores on the Distress Thermometer (DT) are shown (see Figure 17.2). Rates of zero, mild, moderate, and severe distress are approximately 15%, 40%, 30%, and 10%, respectively (Figure 17.2). Predictors of distress are concurrent complications such as cancer pain, function limitations, and reduced quality of life, which are more powerful determinants of distress than cancer type or cancer stage. That said, patients with prostate cancer appear to have about 30% lower rates of distress across multiple studies. Distress is often overlooked in clinical practice, and clinicians’ opinions on distress often do not correspond with patients’ self- reported views.6 Mitchell and colleagues looked at identification of distress by chemotherapy cancer nurses across 400 nurse-patient consultations.7 Nurse practitioners had a detection sensitivity of 50% and specificity of 80%. Unresolved distress is associated with lower adherence to treatment, slower rehabilitation, lower satisfaction with care, higher costs of overall care, and poorer survival rates.8–11 Distress is not a formal psychiatric diagnosis alone but a symptom of emotional upset arising from any cause, which is an important “red flag” that indicates there are unresolved problems and should prompt further inquiry, follow-up, and appropriate psychosocial help. Even mild distress can be an indicator of underlying unmet needs as well as more severe psychiatric disorders such as depression and anxiety. Our group found twice the rate of unmet needs (defined using a custom problem list) in patients with mild distress versus no distress in 480 cancer patients in Leicester, UK (see Figure 17.3). Recognition and Screening for Distress Most organizations agree that recognizing distress (as well as the source of distress) should be a priority for all clinicians, not specifically psychologists, psychiatrists, and social workers. Many, but not all, also recommend that distress screening should be integrated into a comprehensive assessment of well-being and conducted routinely.12 Indeed, there was an attempt to unify national cancer plans to integrate a comprehensive psychosocial approach in cancer care.13 Yet, despite the potential benefits, uptake has been slow, particularly in low-income countries. Screening is designed to quickly ascertain which individuals in a large population need further assessment (Box 17.2; Figure 17.4), and therefore most instruments are not intended to be diagnostic but aim to open a dialogue and provide a simple severity rating. Early studies concentrated on tool development and validation. However, screening tool accuracy does not mean screening will necessarily bring patient benefits. To be successful, evidence-based treatment also has to be available, and also has to be offered (where needed) and accepted. This does not always happen (see Evidence base for Implementing Distress Screening in Clinical Settings). Many authors have pointed out that the evidence base for distress screening is complex and needs to be interpreted with care. Previous narrative reviews have come to different conclusions about the merits of distress screening. For example, some have failed to find any relevant randomized controlled trials, while Box 17.1. Phases of Cancer Risk When Screening Might Be Considered (from National Comprehensive Cancer Network) • • • • • • • • • • Finding a suspicious symptom Being informed about the diagnosis Awaiting treatment Change or end of treatment Discharge from hospital Surviving cancer Failure of treatment Recurrence or progression of disease Advanced phase of illness Approaching the end of life National Comprehensive Cancer Network (NCCN): Distress management. Version 2.2017. 122 SECTION IV Screening and Assessment in Psychosocial Oncology 0.4 0.4 0.37 0.35 0.3 0.18 0.17 0.235 0.24 0.24 0.23 0.2 0.2 0.16 0.18 0.23 0.25 0.18 0.1 0 <3 months 6 months 12 months 5 years 3 years BJGP Open 2019; 3 (3) Older Cancer Patients Middle Aged Cancer Patients Older Primary Care Controls Figure 17.1. Distribution of DT scores across 10,000 individual patient data returns. DT = 10 3.0% DT = 9 3.0% DT = 8 7.0% DT = 7 9.0% DT = 0 16.0% DT = 1 10.0% DT = 6 8.0% DT = 2 11.0% DT = 5 14.0% DT = 3 11.0% DT = 4 8.0% Figure 17.2. Distribution of DT scores across 10,000 individual patient data returns. 15 12.8 11 10 6.4 5 17 0 No distress (score –0) Mild distress (score 1–4) Moderate distress (score 5–7) Severe distress (score 8–10) Mitchell et al (2020) unpublished data Figure 17.3. Frequency of unmet needs on a problem list according to DT scores in 480 mixed cancer patients. CHAPTER 17 Screening and Assessment for Distress 80% 60% 63% 40% 34% 20% 0% Help from family and friends Does not recognize need 10% 8% 6% Don’t know who to ask Question about availability Other reasons Figure 17.4. Reasons for declining psychosocial help. Box 17.2. Phases of Distress Screening Screening Study The application of a diagnostic test or clinical assessment to optimally rule out those without the disorder with minimal false negatives (missed cases). Case Finding Study The application of a diagnostic test or clinical assessment to optimally identify those with the disorder with minimal false positives. Implementation Trial A comparison of one group of clinicians (or center) using screening compared with another group without screening, ideally randomized. others have omitted data from nonrandomized trials or observational screening studies.14,15 Distress Screening Tools Clinicians and researchers wishing to identify and measure distress have a choice between a growing number of tools that have good evidence of acceptability and accuracy in oncology and palliative settings (Table 17.1).16,17 It is generally accepted that brief tools taking around 5 minutes or less to complete are most practical for busy clinical settings. Longer tools may still find a use particularly in waiting rooms, in reception areas, and at home. In 1998 the NCCN released a one- item visual analog scale (VAS) known as the Distress Thermometer (DT). It is often used with the 39-item problem checklist of unmet needs, which takes around 2 minutes to complete. The cutoff for significant distress is still debated, but most commonly a threshold of ≥ 5 (sometimes ≥ 4) is recommended.18,19 Using the Hospital Anxiety and Depression Scale summary score (HADS-T)20 as the criterion reference and at a prevalence of distress of 23%, Ma et al.21 found that the DT has a pooled sensitivity of 82%, a specificity of 73%, a positive predictive value of 48%, and a negative predictive value of 93%. Using the clinical utility index (http://www.clinicalutility.co.uk), the DT appears to be a relatively “poor” case-finding tool but a “good” method of screening for distress (Table 17.1). A number of alternatives to the DT exist, including the Psychological Distress Inventory (PDI), Brief Symptom Inventory (BSI), General Health Questionnaire (GHQ), and Symptom Checklist 90-R (SCL-90), but they all have limited evidence in cancer and/or a narrow focus. Two broader multidomain tools, namely the Edmonton Symptom Assessment System (ESAS) and the Emotion Thermometers (ET), are promising as they cover a broader range of relevant conditions while retaining a brief format. Multidomain tools allow the clinician to screen for mental symptoms such as anxiety and depression and not simply distress. Both the ESAS and ET have undergone considerable independent validation as well as recent implementation studies. The ESAS includes six physical symptoms of cancer (pain, tiredness, nausea, drowsiness, appetite, and shortness of breath) and three psychosocial symptoms (well-being, depression, and anxiety). Provisional evidence suggests reasonable performance in diagnostic validity studies of distress based on the HADS-T. The ET (http://www.emotionthermometers. com) includes four core emotions—distress, depression, anxiety, and anger—and uniquely inquires about need for help. It can also be extended with quality of life, memory, and pain add-ons. The ET has the same 11-point VAS as the DT. Validation from 17 studies in early- and late-stage cancer settings suggest that the ET improves upon the accuracy of the DT.22,23 Evidence Base for Implementing Distress Screening in Clinical Settings Several groups have examined the key question of whether distress screening actually improves clinical care.24,25 Well-designed, large-scale studies comparing care before and after implementation of screening (sequential cohort) or in groups randomized to screening have been gradually forthcoming (Table 17.2).15 Of these implementation studies, only one reasonably consistent outcome measure is receipt of psychosocial referral. An early Cochrane review found only three qualifying studies and failed to demonstrate any effect of distress screening on meaningful outcomes. Carlson et al. found 13 randomized and 10 nonrandomized trials of screening for distress/quality of life that measured patient well-being.15 Of the RCTs, 5 of 13 studies reported additional 123 124 SECTION IV Screening and Assessment in Psychosocial Oncology Table 17.1. Updated Summary of Distress Tools Validated in Multiple Cancer Settings Compared with Weighted Any Mental Disorder Sensitivity (Incl. Adjustment Disorder) Weighted Specificity Weighted Positive Predictive Value HADS (N = 16)* (overall accuracy = 73.3%) 65.2% (95% CI = 62.3%–68.1% 77.2% (95% CI = 75.5%–79.0%) 58.2% 82.0% (95% CI = 55.4%–61.0%) (95% CI = 80.4%–83.7%) 0.379 (95% CI = 0.379–-0.380) Qualitative grade = “poor” 0.634 (95% CI = 0.633–0.634) Qualitative grade = “fair” Distress Thermometer (N = 9)** (overall accuracy = 69.9%) 84.0% (95% CI = 80.0%–88.0%) 63.0% 52.4% 89.0% (95% CI = 61.0%–66.0%) (95% CI = 49.0%–55.8%) (95% CI = 86.8%–91.2%) 0.449 (95% CI = 0.448–0.450) Qualitative grade = “poor” 0.558 (95% CI = 0.557–0.559) Qualitative grade = “fair” Compared with HADS-T Distress Weighted Sensitivity Weighted Specificity Weighted Positive Predictive Value Distress Thermometer (N = 27)** (overall accuracy = 75.1%) 82.0% (95% CI = 80.0%–84.0%) 47.6% 93.1% (95% CI = 45.7%–49.4%) (95% CI = 92.4%–93.9) 0.390 (95% CI = 0.389–0.391) Qualitative grade = “poor” 0.680 (95% CI = 0.679–0.680) Qualitative grade = “good” Single Verbal Depression Question (N = 3)*** (overall accuracy = 62.3%) 76.1% 59.8% (95% CI = 67.2.0%–85.0%) (95% CI = 55.4%–64.1%) 25.7% 93.2% (95% CI = 20.4%–31.0%) (95% CI = 90.4%–96.0%) 0.195 (95% CI = 0.191–199) Qualitative grade = “very poor” 0.557 (95% CI = 0.555–0.559) Qualitative grade = “fair” Two Verbal Depression Questions (e.g., PHQ2) (N = 3)*** (overall accuracy = 73.3%) 75.5% (95% CI = 67.3%–83.7%) 72.6% 37.6% 93.1% (95% CI = 68.63%–76.5%) (95% CI = 31.1%–44.1%) (95% CI = 90.6%–95.7%) 0.283 (95% CI = 0.279–0.288) Qualitative grade = “very poor” 0.676 (95% CI = 0.675–0.677) Qualitative grade = “good” Single-item Anxiety VAS (AnxT or ESAS-A ) (N = 3)*** (overall accuracy = 71.9%) 77.9% (95% CI = 73.0%–82.8%) 71.1 (95% CI = 68.1%–74.1%) 44.6% 91.5% (95% CI = 40.1%–49.1%) (95% CI = 89.4%–93.6%) 0.348 (95% CI = 0.335–0.350) Qualitative grade = “very poor” 0.651 (95% CI = 0.650–0.661) Qualitative grade = “good” Single-item Depression VAS (DepT or ESAS-D) (N = 3)*** (overall accuracy = 71.9%) 76.8% (95% CI = 64.0%–87.5%) 70.5% (95% CI = 55.7%–83.3%) 43.2% 91.0% (95% CI = 39.2%–48.2%) (95% CI = 88.9%–93.2%) 0.336 (95% CI = 0.334–0.338) Qualitative grade = “very poor” 0.642 (95% CI = 0.641–0.643) Qualitative grade = “good” 73.0% (95% CI = 72.0%–74.0%) Weighted Negative Clinical Utility for Predictive Value Case Finding (CUI+) Weighted Negative Clinical Utility for Predictive Value Case Finding (CUI+) Clinical Utility for Screening (CUI-) Clinical Utility for Screening (CUI-) All calculations from http://www.clinicalutility.co.uk * Data from Mitchell AJ, Meader N, Symonds P. Diagnostic validity of the Hospital Anxiety and Depression Scale (HADS) in cancer and palliative settings: a meta-analysis. J Affect Disord. 2010 Nov;126(3):335–48. ** Data adapted from Ma X, Zhang J, Zhong W, Shu C, Wang F, Wen J, Zhou M, Sang Y, Jiang Y, Liu L. The diagnostic role of a short screening tool-the distress thermometer: a meta- analysis. Support Care Cancer. 2014 Feb 8 [Epub ahead of print]. *** New calculation for this chapter. added benefits on patient well-being in screened compared with unscreened patients. Perhaps the most important message was that screening without mandatory follow-up care was consistently unsuccessful. Screening with mandatory follow-up is likely to be beneficial compared with no screening (and treatment as usual), but even in this case only if follow-up is consistent and local psychosocial resources adequate.24 We previously showed that combining implementation studies using mandatory follow-up in a meta- analysis reveals that “enhanced screening” increased the chances of receiving a psychosocial referral by twofold (risk ratio [RR] = 2.03; 95% confidence interval [CI] = 1.13 to 3.65; p < 0.01) compared to those with nonenhanced screening.26 A new study from Schuurhuizen et al. adds additional insights into the pros and cons of screening.27 They conducted a cluster randomized trial across 16 hospitals where patients in the screening arm were screened for psychological distress with the HADS and the DT (including the Problem List) on three occasions and then offered stepped care in comparison to usual care. Of 349 randomized patients, 184 received screening and 60% of these patients screened positive for psychological distress, but despite this, the study was halted. Only 46 patients (25.0%) entered stepped care after screening; instead, many used watchful waiting (15%) and 11% used other steps. The course of distress did not differ among patients assigned to the stepped care group versus the care as usual group most likely due to the low uptake of stepped care. The low uptake of care, despite a high prevalence rate of distress, is consistent with findings in other studies showing low acceptance rates of psychological treatment (see Uptake of Help as a Key Barrier to Screening Success). Some patients needing psychological support show no signs or symptoms of distress, whereas others with elevated levels of distress do not report unmet needs or a desire for professional help.28 These effects reduce the effectiveness of distress screening outcomes. Has Widespread Dissemination of Distress Screening Been Successful? Screening together with follow-up in settings where resources are adequate can improve clinical outcomes, but does screening offer more benefits than risks? Large-scale studies are beginning to accrue that inform us regarding the difficulties of implementing distress screening in clinical practice and help answer the question, when is screening successful and when is it unsuccessful? For example, CHAPTER 17 Screening and Assessment for Distress Table 17.2. Summary of Comparative Distress Implementation Screening Studies Author Country Screening Target Screening Beneficial? PROs Improved? Referrals Improved? Communication Improved? Acceptability of Screening? Distress | Depression No No NR No NR Rosenbloom et al. US (2007) Mood | Quality of life No No NR NR NR Mills et al. (2009) UK Quality of life No (deleterious) No NR Yes, but not significantly High Braeken et al. (2011) Germany Distress No No Yes, but not significantly NR Mixed Hollingworth et al. (2012) UK Distress | Quality of life No No NR NR High Ploos van Amstel et al (2019) Netherlands Distress No No (QoL) NR NR NR van der Meulen et al (2018) Netherlands Distress No No (QoL / NR Depression) NR Yes Geerse et al (2017) Netherlands Distress No No (QoL / NR Depression) NR NR Yes Randomized Unsuccessful Maunsell et al. (1996) Canada Randomized Partially Successful Sarna (1998) US Distress Yes NR Yes NR McLachlan et al. (2001) Australia Distress | Depression | Quality of life Partial (in depressed Yes (in patients) depressed only) NR Yes (in depressed only) NR Detmar et al. (2002) Netherlands Quality of life Partial No NR Yes (but only for social functioning, fatigue, and dyspnea) NR Velikova et al. (2004) UK Distress | Quality of life Yes Yes NR Yes, but not significantly Mixed Macvean et al. (2007) Australia Depression | Quality of life | Unmet needs Yes Yes NR (depression) NR Good Girgis et al. (2009) Australia Depression | Quality of life | Unmet needs Partial (in communication and action) No Yes, and significant (but full data not presented) Yes, and significant (but High full data not presented) Carlson et al. (2010) Canada Distress Yes (in breast and lung cancer) No Yes, significantly NR High Carlson et al. (2012) Canada Distress No Yes Yes (access to services), significantly NR High Klinkhammer- Schalke et al. (2012) Germany Quality of life Yes Yes NR NR Not reported Singer et al. (2016) Germany Depression/anxiety No No (Distress) Yes NR NR Schuurhuizen et al (2019) Netherlands Depression/anxiety Partial (patient satisfaction and cognitive functioning) No (Distress) Yes, but not significantly (16% vs. 12%) NR Yes (87%) Nonrandomized unsuccessful Boyes et al. (2006) Australia Depression | Anxiety | No Unmet needs No NR NR Yes Mitchell et al. (2012) Distress | Depression | Anxiety No NR NR Partial UK Not significantly (continued ) 125 126 SECTION IV Screening and Assessment in Psychosocial Oncology Table 17.2. Continued Author Country Screening Target Screening Beneficial? PROs Improved? Referrals Improved? Communication Improved? Acceptability of Screening? Quality of life Partial (in communication and action) Yes NR Yes, but not significantly NR Pruyn et al. (2004) Netherlands Distress Yes NR Yes, significantly Yes, but not significantly High Bramsen et al. (2008) Netherlands Distress | Quality of life Yes Partial Yes, significantly NR NR Hilarius et al. (2008) Netherlands Quality of life Partial (in recognition and action) No Yes, but not significantly Yes, significantly overall NR Thewes et al. (2009) Australia Distress | Unmet needs Partial (in referral delay) No Yes, but not significantly NR Yes Shimizu et al. (2010) Japan Distress Partial (in referral) No/ Unknown Yes, significantly NR NR Ito et al. (2011) Japan Distress Partial (in referral delay) No Yes, significantly NR NR Grassi et al. (2011) Italy Distress Partial (in referral) No Yes, significantly No NR Zemlin et al. (2011) Distress Partial (in communication) NR NR Yes, significantly Yes Nonrandomized Successful Taenzer et al. (2000) Canada Germany Gotz et al. recently examined results from a comprehensive cancer center in Zurich where 4,541 of 11,184 inpatients completed the DT (40.6%).29 Of these, 45.8% screened above the threshold (≥ 5), and 48.2% of these at-risk patients were screened a second time. Of those that screened positive, only 23.2% wanted a psycho-oncology referral compared with 4.8% of those that screened negative. An additional 30% wanted a referral for spiritual or social care. However, one in five of screened positive patients had already been recognized and referred prior to the screen taking place. In Canada, beginning in 2015, the System Performance Initiative at the Canadian Partnership Against Cancer (http://systemperformance.ca) implemented widescale ESAS-r screening to assess patient-reported outcomes in over 200,000 patients. Approximately 46.6% were distressed and 53.5% nondistressed.30 Outcomes have yet to be fully reported, but preliminary evidence suggests that screening prompted a significant increase in psychosocial social work referrals and palliative care referrals.31 The American College of Surgeons’ Commission on Cancer (ACS CoC) adopted routine distress screening as an accreditation requirement in 2015.23 In an early review of 8,409 electronic health records across 55 American cancer centers, 62.7% followed a prescribed distress screening protocol, although this rate was 43.3% in National Cancer Institute (NCI)-designated Cancer Centers.32,33 Screening rates may be increasing, however. In a new 2019 survey of 27 NCCN Member Institutions, Geske and Johnson found that 87% of centers were conducting routine screening for distress. However, follow-up screening remained inconsistent, as did documentation of psychosocial screening results and receipt of psycho-oncology care.34 In the same sample, only 65% of institutions routinely tracked clinical contacts and referrals, and 70% tracked rates of adherence to screening protocols.35 Uptake of Help as a Key Barrier to Screening Success A number of studies have addressed the important question of how many cancer patients are offered and then accept psychosocial help. The current literature suggests that of patients with unmet psychosocial needs, 40% are offered some kind of psychosocial help and 30% are offered a referral to a psycho-oncology specialist. Clearly this number is limited by the uneven international provision of dedicated psycho-oncology teams, particularly in low-and middle- income countries. Research suggests that 40% of patients who were offered specific psychosocial interventions are willing to immediately consider this psychosocial help (rather than defer or refuse), but ultimately only one in three of distressed patients actually attend for psychosocial help. Several larger studies on this topic offer valuable information. Thalén-Lindström and colleagues followed 495 patients’ screening using the HADS in Uppsala, Sweden. Fifty-seven percent of screen-positive patients refused further assessment, but ultimately enhanced screening led to support and treatments for 24% of the screen-positive patients vs. only 2% in those screened under usual care conditions.36 Riedl et al. retrospectively analyzed routine data from 944 cancer outpatients treated at the Department of Therapeutic Radiology, Innsbruck.37 Only 20% were identified as distressed, and 42.7% of these wished for psycho-oncology support. Six percent of patients scoring as nondistressed also wished for psycho-oncology support. Skaczkowski et al. conducted retrospective analysis of cancer care in Melbourne, Australia, covering 833 patients seen in 2015.38 Eighty-six percent consented to distress screening. Half scored 4 or higher on the DT. Seventy-seven percent of distressed patients CHAPTER 17 Screening and Assessment for Distress were offered a referral for help, and of the remainder, 83% had a discussion with their clinician about support care; thus, only 3% received nothing despite a high distress rate. Interestingly, a referral offer from a clinician was significantly predicted by a higher level of distress, but acceptance by the patients was significantly related to lower distress. Also, physical treatment was accepted by 87%, but emotional support was accepted by only 53%. Reasons for declining support (Figure 17.4) include male sex, not feeling depressed, perceived overload, and feeling well informed about psychological support.39,40 Future Directions Screening for distress in clinical practice remains a challenge. Despite the successful development and validation of numerous instruments and large-scale implementation in a number of cancer centers, several key barriers (Box 17.3) have prevented screening from being universally approved. Additionally, the evidence base for the effect of screening implementation on patient outcomes or referral rates has been disappointing, not necessarily due to inadequacy of distress screening itself, but rather due to a failure to unify essential components of high-quality care such as patient follow-up or the offer of appropriate treatment options. Without all components of quality of care in place, and without a demonstrable difference to patients seen locally, routine screening is almost always seen as an unnecessary burden to patients and staff. The NCCN was among the first to call for routine screening of distress in patients with cancer in 1999.1 In 2015 the ACS CoC mandated a systematic protocol for distress screening and referral that made screening a requirement for cancer center accreditation.5 The success of screening in routine care depends as much on Box 17.3. Barriers to Screening Success 1. Inaccuracy Barrier Screening is not 100% accurate and will yield false positives and false negatives. 2. Double-Counting Barrier A proportion of cases identified by screening will already be known to be distressed and in receipt of care. 3. No Suitable Treatment Barrier No suitable treatment is available or offered to screen- positive patients. 4. No Follow-Up Barrier No adequate follow-up is available or offered to screen-positive patients. 5. Worried Screen-Negative Persons/“Worried Well” Some patients (approximately 10%) who screen negative still want help regardless of the absence of distress. 6. Burden Barrier Clinicians will often see screening as too time consuming and results too difficult to interpret. 7. Interpretation Barrier Clinicians may have difficulty following the algorithm or interpreting the score on complex screening tools. 8. Declining Help Barrier A major proportion of patients who screen positive will decline help that is available. acceptability as it does on accuracy. Acceptability is the rate-limiting step in screening implementation. This is acceptability not just for the screening procedure, but also for its scoring interpretation and any linked follow-up and treatment. Several studies have reported that under optimal conditions, it is possible to screen large numbers of patients with few refusals. These large screening programs are often assisted by funded researchers or investment in computerized touch-screen terminals. Reception to screening programs run by frontline clinicians is more mixed. For example, physicians in Leeds, UK, found quality-of-life screening at least quite useful in 43% of encounters but of little use in 30%.41 Mitchell et al. reported that British cancer clinicians felt screening was useful in only 43% of assessments and not useful in 36%.42 Zebrack et al. found that between 15% and 35% of clinicians felt screening slowed down clinical procedures, and between 40% and 80% felt screening helps patients receive appropriate care.43 Given these limitations, how can screening processes be improved in the future? First, clinicians should not be given the task of screening without time to perform it, an understanding of how it might help, and regular feedback of screening success. Second, patients should not be expected to complete lengthy questionnaires outside of their first language. Third, screening should focus on unmet needs that can be addressed. Distress is a red flag, but it is identification of the underlying cause that is paramount. Not uncommonly, concerns unrelated to cancer contribute to cancer patients’ distress. Finally, having identified unmet needs, clinicians have a duty of care to attempt to resolve them using the best treatment option available, while keeping the patient informed. Despite strong recommendations of many professional societies and accreditation agencies, to date very few cancer centers have adopted routine screening for distress, depression, quality of life, or unmet needs (Box 17.3). Screening success thus depends on three key variables: accuracy, acceptability, and high-quality follow-up care. Psychosocial needs remain the most overlooked of all cancer complications. Psychological assessment and enhanced distress screening together with appropriate follow-up and tailored acceptable interventions will help reduce this gap in unmet psychosocial needs. REFERENCES 1. National Comprehensive Cancer Network (NCCN): Distress management. Version 2.2017. NCCN, 2017. 2. Pirl W, Fann J, Greer J, Braun I, Deshields T, et al. Recommendations for the implementation of distress screening programs in cancer centers: report from the American Psychosocial Oncology Society (APOS), Association of Oncology Social Work (AOSW), and Oncology Nursing Society (ONS) joint task force. Cancer. 2014 Oct 1;120(19):2946–2954. 3. Tomitaka S, Kawasaki Y, Ide K, Akutagawa M, Ono Y, Furukawa TA. Distribution of psychological distress is stable in recent decades and follows an exponential pattern in the US population. Sci Rep. 2019;9:11982. 4. Carlson L, Zelinski E, Toivonen K, et al. Prevalence of psychosocial distress in cancer patients across 55 North American cancer centers. JOURNAL OF Psychosocial Oncology Oncol. Volume: 37 Issue: (1) Pages: 5–21. Published: JAN 2 2019 5. Dauphin S, Jansen L, De Burghgraeve T, Deckx L, Buntinx F, van den Akker M. Long- term distress in older patients with 127 128 SECTION IV Screening and Assessment in Psychosocial Oncology cancer: a longitudinal cohort study. BJGP Open. 2019 Oct 29;3(3) bjgpopen19X101658. doi:10.3399/bjgpopen19X101658 6. Keller M, Sommerfeldt S, Fischer C, et al. Recognition of distress and psychiatric morbidity in cancer patients: a multi-method approach. Ann Oncol. 2004;15:1243–1249. 7. Mitchell A, Hussain N, Grainger L, Symonds P. Identification of patient-reported distress by clinical nurse specialists in routine oncology practice: a multicentre UK study. Psychooncology. 2011 Oct;20(10):1076–1083. 8. Von Essen L, Larsson G, Oberg K, Sjödén P. “Satisfaction with care”: associations with health-related quality of life and psychosocial function among Swedish patients with endocrine gastrointestinal tumours. Eur J Cancer Care. 2002;11:91–99. 9. Geerse O, Brandenbarg D, Kerstjens H, Berendsen A, Duijts S, Burger H, Holtman G, Hoekstra-Weebers J, Hiltermann T. The distress thermometer as a prognostic tool for one-year survival among patients with lung cancer. Lung Cancer. 2019 Apr;130:101–107. 10. Nipp R, El-Jawahri A, Moran S. The relationship between physical and psychological symptoms and health care utilization in hospitalized patients with advanced cancer. Cancer. 2017;123:4720–4727. 11. Batty G, Russ T, Stamatakis E. Psychological distress in relation to site specific cancer mortality: pooling of unpublished data from 16 prospective cohort studies. BMJ. 2017;356:j108. 12. IOM 2008 Institute of Medicine. Cancer Care for the Whole Patient: Meeting Psychosocial Health Needs. Washington, DC: National Academies Press, 2008. 13. Grassi L, Watson M. Psychosocial care in cancer: an overview of psychosocial programmes and national cancer plans of countries within the International Federation of Psycho-Oncology Societies. Psychooncology. 2012;21(10):1027–1033. 14. Luckett T, Butow P, King M. Improving patient outcomes through the routine use of patient-reported data in cancer clinics: future directions. Psychooncology. 2009;18:1129–1138. 15. Carlson L, Waller A, Mitchell A. Screening for distress and unmet needs in patients with cancer: review and recommendations. J Clin Oncol. 2012 Apr 10;30(11):1160–1177. Epub 2012 Mar 12. 16. Mitchell A. Short screening tools for cancer-related distress: a review and diagnostic validity meta-analysis. J Natl Compr Canc Netw. 2010 Apr;8(4):487–494. 17. Vodermaier A, Linden W, Siu C. Screening for emotional distress in cancer patients: a systematic review of assessment instruments. J Natl Cancer Inst. 2009 Nov 4;101(21):1464–1488. 18. Mehnert A, Hartung T, Friedrich M, et al. One in two cancer patients is significantly distressed: prevalence and indicators of distress. Psychooncology. 2018 Jan;27(1):75–82. 19. National Comprehensive Cancer Network. NCCN Clinical Practice Guidelines in Oncology: Distress Management. v2.2019. Referenced with permission from the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®) for Distress Management V.2.2019. © National Comprehensive Cancer Network, Inc. 2019. All rights reserved. To view the most recent and complete version of the guideline, go online to NCCN.org. 2019. Retrieved from http://www.nccn.org/professionals/physician_gls/distress.pdf. 20. Zigmond A, Snaith R. The Hospital Anxiety and Depression Scale. Acta Psychiatr Scand. 1983;67:361–370. 21. Ma X, Zhang J, Zhong W, et al. The diagnostic role of a short screening tool-the distress thermometer: a meta-analysis. Support Care Cancer. 2014 Feb 8. 22. Mitchell A, Baker- Glenn E, Symonds P. Can the Distress Thermometer be improved by additional mood domains? Part I. Initial validation of the Emotion Thermometers tool. Psychooncology. 2010 Feb;19(2):125–133. 23. Harju E, Michel G, Katharina, K. A systematic review on the use of the emotion thermometer in individuals diagnosed with cancer. Psycho-oncology. 2019;28(9):1803–1818. 24. Mitchell A. Screening for cancer-related distress: when is implementation successful and when is it unsuccessful? Acta Oncol. 2013;52:216–224. 25. McCarter K, Britton B, Baker A, et al. Interventions to improve screening and appropriate referral of patients with cancer for psychosocial distress: systematic review. BMJ Open. 2018 Jan 5;8(1):e017959. 26. Mitchell AJ. Psycho- Oncology. Chapter 48: Screening and Assessment for Distress. 3rd edition, edited by Jimmie C. Holland, William S. Breitbart, Paul B. Jacobsen, Matthew J. Loscalzo, Ruth McCorkle, Phyllis N. Butow. Oxford University Press, New York. 27. Schuurhuizen CSEW, Braamse AMJ, Beekman ATF, et al. Screening and stepped care targeting psychological distress in patients with metastatic colorectal cancer: the TES Cluster Randomized Trial. J Natl Compr Canc Netw. 2019 Aug 1;17(8):911–920. doi:10.6004/ jnccn.2019.7285. 28. Brebach R, Sharpe L, Costa S. Psychological intervention targeting distress for cancer patients: a meta-analytic study investigating uptake and adherence. Psychooncology. 2016;25:882–890. 29. Götz A, Kröner A, Jenewein J, Spirig R. Evaluation of the adherence of distress screening with the distress thermometer in cancer patients 4 years after implementation. Support Care Cancer. 2019 Aug;27(8):2799–2807. 30. Tran K, Zomer S, Green E, et al. Measuring patient-reported outcomes to improve cancer care in Canada: an analysis of provincial survey data. Curr Oncol. 2018 Apr;25(2):176–179. 31. Hui D, Titus A, Curtis T, et al. Implementation of the Edmonton Symptom Assessment System for symptom distress screening at a community cancer center: a pilot program. Oncologist. 2017 Aug;22(8):995–1001. 32. Zebrack B, Kayser K, Bybee D, et al. A practice-based evaluation of distress screening protocol adherence and medical service utilization. J Natl Compr Canc Netw. 2017;15(7):903–912. 33. Jacobsen P, Ransom S. Implementation of NCCN distress management guidelines by member institutions. J Natl Compr Canc Netw. 2007;5:99–103. 34. Geske S, Johnson R. Using the distress thermometer to guide electronic referrals to psychosocial services. J Psychosoc Oncol. 2020;38(1):20–35. 35. Donovan K, Deshields T, Corbett C, Riba M. Update on the implementation of NCCN Guidelines for Distress Management by NCCN member institutions. J Natl Compr Canc Netw. 2019 Oct 1;17(10):1251–1256. 36. Thalén- Lindström A, Larsson G, Glimelius B, Johansson B. Anxiety and depression in oncology patients: a longitudinal study of a screening, assessment and psychosocial support intervention. Acta Oncol. 2013 Jan;52(1):118–127. 37. Riedl D, Gastl R, Gamper E, et al. Cancer patients’ wish for psychological support during outpatient radiation therapy: findings from a psychooncological monitoring program in clinical routine. Strahlenther Onkol. 2018 Jul;194(7):655–663. 38. Skaczkowski G, Sanderson P, Shand M, Byrne A, Wilson C. Factors associated with referral offer and acceptance following supportive CHAPTER 17 Screening and Assessment for Distress care problem identification in a comprehensive cancer service. Eur J Cancer Care (Engl). 2018 Sep;27(5):e12869. 39. Pichler T, Dinkel A, Marten-Mittag B, et al. Factors associated with the decline of psychological support in hospitalized patients with cancer. Psychooncology. 2019 Oct;28(10):2049–2059. 40. Ernst J, Faller H, Koch U, et al. Doctor’s recommendations for psychosocial care: frequency and predictors of recommendations and referrals. PLoS One. 2018 Oct 4;13(10):e0205160. 41. Cull A, Gould A, House A, et al. Validating automated screening for psychological distress by means of computer touchscreens for use in routine oncology practice. Br J Cancer. 2001 Dec 14;85(12):1842–1849. 42. Mitchell A, Lord K, Slattery J, Grainger L, Symonds P. How feasible is implementation of distress screening by cancer clinicians in routine clinical care? Cancer. 2012 Dec 15;118(24):6260–6269. 43. Zebrack B, Kayser K, Sundstrom L, et al. Psychosocial distress screening implementation in cancer care: an analysis of adherence, responsiveness, and acceptability. J Clin Oncol. 2015 Apr 1;33(10):1165–1170. 129 18 Assessment, Screening, and Case Finding for Depression and Anxiety in People with Cancer Kristine A. Donovan and Paul B. Jacobsen Introduction The development of the field of psychosocial oncology has been spurred by research using assessment instruments capable of documenting the negative impact of cancer and its treatment on patients’ emotional well-being. The need to address these problems has led to many of the same instruments being used as outcome measures in studies evaluating the benefits of interventions designed to maintain or improve patients’ emotional well-being. In both instances, the field has been advanced by having instruments available that possess good psychometric properties and are suitable for use with people with cancer. In this chapter, we identify assessment instruments commonly used to assess depression and anxiety in psychosocial oncology research and summarize the evidence regarding the relative merits of these instruments. Over the past decade, there has been growing interest in extending the use of assessment instruments into everyday clinical practice. This interest has been driven, in part, by clinical practice guidelines and consensus statements, which recommend that patients’ emotional well-being be routinely assessed to identify those patients in need of psychosocial care. These recommendations have stimulated considerable research examining how well brief assessment instruments function as ways to screen for clinically significant problems in emotional well-being and to identify cases of mood and anxiety disorders. Given these developments, we also identify in this chapter brief assessment instruments that can be used for depression and anxiety screening and case finding and summarize the available evidence regarding how well they function when used for these purposes in adults with cancer. Judging the relative merits of assessment instruments is a complex task. Among the issues to be considered are (1) the criteria by which to select instruments to review, (2) the criteria by which to evaluate the instruments, (3) the evidence to be used as part of the evaluation process, and (4) the evaluation of the available evidence. Systematic reviews and meta-reviews are very useful in this regard given the requirements that these types of reviews clearly specify their selection and evaluation criteria, the evidence used for evaluation purposes, and the methods used to form evaluations. Accordingly, in this chapter, we draw as much as possible on published systematic and meta-reviews of assessment, screening, and case finding for depression and anxiety in adults with cancer. Assessment of Depression As stated previously, psychosocial oncology has been advanced through the use of assessment instruments to document cancer’s negative impact on emotional well-being as well as the potential benefits of interventions designed to maintain or improve emotional well-being. A 2010 publication by Luckett and colleagues1 remains, to the best of our knowledge, the most recent systematic review to evaluate the relative merits of patient-reported outcome measures for assessing depression in adults with cancer. The stated purpose of this review was to identify optimal patient-reported outcome measures (PROs) of depression (as well as anxiety and distress) for evaluating psychosocial interventions for English-speaking adults in active treatment for cancer. The authors searched publications appearing between 1999 and 2009 to identify all PROs used to assess depression, anxiety, and distress in randomized controlled trials (RCTs) enrolling English-speaking cancer patients. This process resulted in the identification of 30 instruments. These measures then underwent a review that excluded those (1) not suitable for people with any type or stage of cancer, (2) in which one-third or more of the items were considered to be confounded by cancer or its treatment, and (3) for which there were no published data on reliability and validity in an English-speaking cancer sample. The measures remaining, including 10 instruments assessing depression, were then evaluated on the following eight criteria based on data compiled from relevant articles, websites, and manuals: validity, reliability, ability to identify treatment effects in RCTs, criterion validity against a diagnostic interview, availability of comparison data, number of psychological constructs, length, and ease CHAPTER 18 Assessment, Screening, and Case Finding for Depression and Anxiety in People with Cancer of administration and cognitive burden. Scores (0, 5, or 10) were assigned for each criterion based on the extent to which the available evidence suggested the measure possessed favorable features, with higher scores indicative of more favorable features. Finally, scores for four of the criteria (validity, reliability, ability to identify treatment effects, and criterion validity against a diagnostic interview) were weighted (i.e., were multiplied by) 1.5, while the remainder were weighted 1.0. A summary of the weighted scores for the 10 measures assessing depression appears in Table 18.1. It should be noted that several of these measures feature multiple subscales, one of which measures depression (e.g., Hospital Anxiety and Depression Scale [HADS]), while others only measure depression (e.g., Center for Epidemiological Studies Depression Scale [CES-D]). As shown in Table 18.1, the HADS earned the highest overall score among measures assessing depression followed by the Profile of Mood States-37 (POMS-37) and the CES-D. The HADS’ highest overall score reflects the abundant evidence of its psychometric properties and its efficiency in assessing depression, anxiety, and distress using only 14 items. The HADS also scored highly for the availability of comparison data and its ability to identify treatment effects in RCTs. The HADS Depression Scale (HADS-D) consists of seven of the 14 HADS items. The review notes that all seven items assess emotional experiences consistent with major depressive disorder. The HADS is notable for its deliberate omission of somatic items designed to avoid possible confounding of psychological symptoms with disease-or treatment-related symptoms. The POMS-37 is an unofficial short form of the 65-item POMS that was developed specifically for use with cancer patients to reduce test burden. It scored consistently well on most measures of psychometric properties. In contrast, it scored poorly in terms of criterion validity due largely to limited evidence of its criterion validity relative to a diagnostic interview. The Depression-Dejection subscale makes up eight of the 37 items. The CES-D also scored consistently well on most measures of psychometric properties, but like the POMS-37 scored poorly in terms of criterion validity due to limited evidence. Another feature that counted against the CES-D is that it requires 20 items to assess a single construct (i.e., depression). Although short forms of the CES- D have been developed, they have not been used widely in studies of cancer patients. The authors’ decision to exclude measures in which one-third or more of the items could be confounded by symptoms or side effects (e.g., loss of appetite) resulted in the exclusion of at least two widely used measures of depression: the Beck Depression Inventory- II (BDI- II) and the Patient Health Questionnaire- 9 (PHQ- 9). Although concerns have been raised that a measure’s performance characteristics may be compromised by the presence of somatic content, results of two studies using the PHQ-9 challenge the view that items assessing somatic symptoms reduce a measure’s accuracy in identifying depression in the context of cancer.2,3 Accordingly, future reviews of instruments to assess depression in cancer should Table 18.1. Weighted Scores for Patient-Reported Outcome Measures Assessing Depression and Anxiety Criterion Validity Summary Ratings Reliability Ability to Identify Treatment Effects Criterion Validity Comparison Data No. of Constructs Length Ease and Burden Raw Score (/ 80) Weighted Score (/100) Weight 1.5 1.5 1.5 1.5 1.0 1.0 1.0 1.0 HADS (D, A) 7.5 7.5 15 7.5 10 10 10 10 65 77.5 POMS-37 (D, A) 15 7.5 7.5 0 5 5 10 10 65 77.5 CES-D (D) 7.5 7.5 15 15 5 0 0 5 40 55 POMS-65 (D, A) 7.5 7.5 15 0 5 5 5 10 45 55 BSI-18/53 (D, A) 7.5 0 7.5 0 10 5 10 10 45 50 SCL-90-R (D, A) 7.5 7.5 7.5 0 5 5 5 10 40 47.5 GHQ-28 (D, A) 0 0 0 7.5 5 5 10 10 35 37.5 CES-D-15 (D) 7.5 7.5 0 0 5 0 5 5 25 30 MHI-38 (D, A) 0 0 0 0 5 5 10 5 25 25 C-SOSI (D) 0 7.5 0 0 0 0 10 10 25 27.5 Adapted from Luckett et al.1 HADS = Hospital Anxiety and Depression Scale; POMS = Profile of Mood States; CES-D = Center for Epidemiologic Studies Depression Scale; BSI = Brief Symptom Inventory; SCL = Symptom Checklist; GHQ = General Health Questionnaire; MHI = Mental Health Inventory; C-SOSI = Calgary Symptoms of Stress Inventory; D = Depression measure; A = Anxiety measure. 131 132 SECTION IV Screening and Assessment in Psychosocial Oncology consider also evaluating the relative merits of measures that incorporate somatic content. Assessment of Anxiety Using the same search and exclusion procedures described earlier, the review by Luckett and colleagues1 yielded seven measures of anxiety (see Table 18.1). These measures were evaluated and rated based on the same criteria used with the depression measures. All seven of the anxiety measures feature multiple subscales, one of which measures anxiety. The list does not include any measures that only assess anxiety. As with the depression measures, the two top-scoring anxiety measures are the HADS and the POMS-37. The third highest scored measure is the full-length POMS-65. Strengths of the HADS have been described previously. The HADS Anxiety Scale makes up seven of the 14 HADS items. The review noted that three of these items assess emotional experiences as distinct from generalized anxiety disorder, thus raising concerns about criterion validity. The strengths and weaknesses of the POMS-37, which also apply to the POMS-65, have been described previously. The Tension-Anxiety subscale of the POM-37 consists of six items and the corresponding subscale of the POMS-65 consists of nine items. One additional anxiety measure merits discussion. The State-Trait Anxiety Inventory (STAI) consists of 20 items assessing trait anxiety and 20 items assessing state anxiety. It was excluded from full evaluation by Luckett and colleagues1 based on a lack of sufficient evidence of its validity and reliability despite the STAI having been used in 30 RCTs identified by the authors. Additional research conducted subsequent to this review may suggest a reconsideration of the usefulness of the STAI in assessing anxiety in people with cancer. Screening and Case Finding for Depression In 2015, Wakefield and colleagues4 published a meta-review (i.e., a review of reviews) evaluating the strengths and weaknesses of depression measures used for screening and case finding in adults diagnosed with cancer. Consistent with prior work, screening was defined as the ability to rule out patients without depression with minimal false negatives, and case finding was defined as the ability to rule in patients with depression with minimal false positives. Systematic and narrative reviews published in English between 1999 and 2014 were eligible for inclusion. Measures of general distress were excluded unless they were specifically evaluated as screening or case finding methods for depression. The search yielded 12 systematic and seven narrative reviews that encompassed 372 original studies and assessed more than 50 depression measures. The authors note that 11 of the 12 systematic reviews met at least 20 of the 27 Preferred Reporting Items for Systematic Review and Meta-analyses (PRISMA) criteria. Ten reviews assessed the suitability of depression measures for screening and three reviews assessed their suitability for case finding. As part of their evidence synthesis, the authors summarize the positive and negative features of the most commonly reviewed and recommended measures of depression. Similar to assessment, the HADS was the most widely evaluated measure. Positive features identified include its strength as a screening measure. Limitations include its weakness as a case finding measure and the variability in the recommended cut-points reported in the literature. Additionally, there was concern as to which HADS scale or subscale (HADS-Total, HADS-A, or HADS- D) is most suitable because different reviews have evaluated different combinations of HADS scores. Similar to assessment, the BDI or its variations (BDI-II) and the CES-D have also been widely evaluated. Strengths of BDI-based measures include their adequate screening and case finding performance. Weaknesses include the length of the full measure (21 items) and its extended recall period (two weeks) that may limit use in certain contexts. Strengths of the CES-D include its adequate performance as a screening measure and the ease of completion. Weaknesses include concerns about its suitability for geriatric patients given the scope of item content. Two additional measures mentioned in the meta-review merit discussion. The Distress Thermometer (DT) is a single-item measure that, although designed to screen for distress, has been evaluated as a depression screening measure. The authors note that one review included in their meta-review concluded that the DT performed adequately as a screening method for depression. However, another review concluded that, given the high rate of false negatives, the DT and other ultra-short instruments should not be used in isolation in screening for depression. The PHQ-9 is a nine-item measure developed originally for use in primary care and obstetrics-gynecology settings to screen for major depressive disorder. The item content corresponds with the diagnostic criteria for major depressive disorder. Although the PHQ-9 was noted to have strong psychometric properties in medical populations, it has been rated poorly in prior reviews based on concerns about the two-week recall period, the presence of somatic content, and reportedly low reliability and validity in patients with cancer. It should be noted that more recent research suggests that the PHQ-9 possesses desirable properties as a depression screening and case finding instrument in people with cancer.4 The picture that emerges from this meta-review is that there is no single measure for which unqualified support was evident and that no single measure is ideal for all types of patients and time points. Additionally, caution is urged regarding the omission of somatic symptoms since omission of such content does not appear to substantially improve the screening performance of common depression measures. Screening and Case Finding for Anxiety In 2009, Vodermaier and colleagues published a systematic review designed to identify and evaluate instruments used to screen for emotional distress in cancer patients.5 The authors systematically searched for published English-language studies using terms that were consistent with the definition of distress as “a state of negative affect that is suggestive of affective disorders (i.e., minor or major depressive disorder and dysthymia), anxiety disorders, and adjustment disorders (depressive, anxious, or mixed).” Studies were included if they attempted to validate an interviewer-administered or standardized self-administered instrument in a sample of cancer patients. The review was further restricted to studies evaluating measures consisting of 50 or fewer items. This process led to the identification of 33 instruments, including instruments designed to measure CHAPTER 18 Assessment, Screening, and Case Finding for Depression and Anxiety in People with Cancer anxiety alone or both depression and anxiety. As previously noted, in their systematic review, Luckett and colleagues1 also identified instruments used to evaluate anxiety alone or with depression in RCTs of psychosocial interventions in cancer patients. For purposes of this chapter, we have combined findings from both reviews, selecting among the identified instruments those that were evaluated in more than one study or RCT and are generally considered to be suitable for screening and case finding for anxiety, either alone or in combination with depression. The HADS was recommended by both Luckett et al.1 and Vodermaier et al.5 as a screener when anxiety or mixed affective disorders are of concern based on the strength of the large body of evidence for its psychometric properties and its length. Both reviews noted the evidence for its superiority in screening for anxiety over screening for depression but ultimately concluded that the HADS- D is superior to the HADS-A and HADS-T in ruling out, and similar to in ruling in, cases of mixed affective disorders (depression, anxiety, adjustment disorders combined). This conclusion was supported by findings of a 2011 systematic review and meta-analysis by Vodermaier et al.6 that examined the accuracy of the HADS as a screening tool for mental disorders (not specifically anxiety disorders) and depressive disorders alone compared to a structured or semistructured clinical interview. Based on the review and pooled validation of 28 studies, they concluded that the HADS-A was inferior to the HADS-T and HADS-D subscale in screening for depression and for any mental disorders. Both subscales had lower diagnostic accuracy than the total scale; this may reflect the fact that cancer patients often have symptoms indicative of adjustment disorders where symptoms include a mix of depression and anxiety. As noted in all three reviews, optimal cutoff scores for the HADS have varied across studies, making its screening and diagnostic accuracy equally variable. Nevertheless, studies that have compared the HADS with other candidate measures as well as structured clinical interviews suggest the HADS performs better or comparably with other measures. The POMS-37 was also recommended by Luckett et al.1 based on the evidence for its validity and responsiveness. Items from the tension-anxiety scale of the POMS-37 (and POMS-65) were noted to closely resemble clinical diagnostic criteria though with an emphasis on cardinal symptoms of anxiety rather than the full range of possible symptoms. Two additional measures warrant attention. The Post Traumatic Stress Disorder Checklist (PCLC) was the only anxiety-specific instrument evaluated by Vodermaier et al.5 The PCLC is a 17-item measure that corresponds to key symptoms of posttraumatic stress disorder (PTSD). The instrument can be modified to fit a specific timeframe and any (nonmilitary) traumatic event. Although data regarding its use are limited, its psychometric properties are considered adequate. The Generalized Anxiety Disorder scale (GAD-7) is a seven-item instrument that screens for typical symptoms of anxiety disorders in the previous two weeks. It includes two items from the PHQ-4A and five additional items that facilitate screening for GAD. While anxiety disorders include phobias, panic disorder, social anxiety, and generalized anxiety, it is recommended that cancer patients be screened for GAD specifically. This is because GAD is the most common anxiety disorder and frequently occurs with other mood or anxiety disorders. A recent study by Esser et al.7 used data from a multicenter study of more than 2,000 cancer patients to examine the diagnostic accuracy of the GAD-7 and HADS-A in detecting GAD compared to the Composite International Diagnostic Interview for Oncology, a standardized interview for cancer patients. Overall accuracy as measured by the area under the receiver operating characteristics curve for the GAD-7 and HADS-A were identical and adequate. Notably, the choice of optimal cutoff points for both measures differed from the recommended cutoffs. Sensitivity and specificity for identifying GAD was optimal at >7 for the GAD-7 rather than the recommended >10, and at >8 rather than the recommended >11 for HADS-A. The researchers suggest that high levels of anxiety in cancer patients may not be indicative of pathological GAD and that the diagnostic accuracy and optimal cutoffs of both the GAD-7 and the HADS-A may be different in cancer patients. Clinical Practice Guideline Recommendations for Depression and Anxiety Screening In this section, we review recommendations for depression and anxiety screening contained in three clinical practice guidelines: the Pan-Canadian practice guideline for screening, assessment, and management of psychosocial distress, depression, and anxiety in adults with cancer developed by the Canadian Partnership against Cancer and the Canadian Association of Psychosocial Oncology;8 an adaptation of the Pan-Canadian practice guideline developed by the American Society of Clinical Oncology9 (ASCO); and guidelines for the screening, assessment, and management of anxiety and depression in adults cancer patients developed for the Psycho-oncology Co-operative Research Group (PoCoG) in Australia.10 The Pan-Canadian guideline was first issued in 2010 and updated in 2015.8 It is based on a combination of expert panel recommendations, adaptation of existing guidelines, and a systematic review of evidence on management of distress, depression, and anxiety. A notable feature of this guideline is the identification of the source and strength of evidence for key recommendations. In general, recommendations related to screening were rated as being based on moderate-or low-quality evidence. Regarding depression-specific screening measures and processes, the guideline recommends an initial screening using any of three brief screeners: the depression item from the Edmonton Symptom Assessment System-revised (ESAS-r), the DT, or the PHQ-2. For patients who score at moderate or severe levels on the ESAS-r or DT (i.e., >3), a more focused assessment is recommended that may include administration of the full PHQ-9 and evaluation of diagnostic criteria for depressive disorders. Subsequent triage to different clinical care pathways involves a determination of whether depressive symptoms are minimal, moderate, or more severe based in part on whether PHQ-9 scores are 1 to 7, 8 to 14, and 15 or greater, respectively. Regarding anxiety-specific screening measures and processes, the guideline recommends an initial screening using the anxiety item from the ESAS-r. For patients who score at moderate or severe levels (i.e., >3), a more focused assessment is recommended and may include administration of the GAD-7. Subsequent triage to different clinical care pathways involves a determination of whether anxiety symptoms are minimal, moderate, or more severe based in part on whether GAD-7 scores are 0 to 9, 10 to 14, and 15 or greater, respectively. 133 134 SECTION IV Screening and Assessment in Psychosocial Oncology The ASCO adaptation of the Pan-Canadian guideline was published in 2014.9 The adaptation was conducted by an expert panel with multidisciplinary representation. Although very similar to the Pan-Canadian guideline, several differences are apparent. For example, instead of ESAS-r items, the ASCO adaptation recommends initial screening with the PHQ-2 for depression (followed by the full PHQ-9 for patients who score >1) and with the full GAD-7 for anxiety. PHQ-9 and GAD-7 cut-points identical to those described in the Pan-Canadian guideline are then recommended to triage patients into different care pathways based on whether they have mild, moderate, or more severe symptomatology. It should be noted that although the focus is on the PHQ-9 and the GAD-7, both the Pan- Canadian guideline and the ASCO adaption identify several other measures of depression (e.g., CES-D) and anxiety (e.g., STAI) that can be used to classify symptoms as mild, moderate, or severe. The clinical pathways described in the Australian guidelines were published in 2015.10 They were developed based on a review of existing guidelines, systematic reviews, and meta-analyses relevant to screening, assessment, and management of anxiety and depression in cancer patients and the general public; interviews with key multidisciplinary staff; an online Delphi process that engaged PoCoG members; and input from a multidisciplinary panel. Similar to the Pan-Canadian guideline, these guidelines recommend initial screening for depression and anxiety using the ESAS-r and the DT. If potential depression or anxiety is identified using either of these instruments based on cutoff scores (ESAS depression item >1; ESAS anxiety item >2; DT >3), the guidelines recommend administration of a more detailed screening tool such as the HADS. Should scores on either the HADS anxiety or depression subscales exceed 7, a formal assessment is then recommended to confirm caseness of depression or anxiety. Subsequent triage to care pathways is based on clinical assessment as to whether symptoms are minimal, mild, moderate, severe, or very severe. This brief review illustrates similarities and differences in depression and anxiety screening recommendations across these three clinical practice guidelines. Similarities include recommendations for a two-stage screening process in which administration of a one- or two-item measure is used to decide whether administration of a longer screening measure is warranted. Differences include the specific measures recommended for the initial and subsequent screening. While the Pan-Canadian and Australian guidelines include recommendations to use ESAS-r items or the DT as part of initial screening, the ASCO adaptation focuses on use of the PHQ-2 based on a view that the ESAS-r is not widely used in the United States. This decision raises questions about the extent to which practical considerations (i.e., regional preferences) should guide measure selection relative to other factors (i.e., psychometric considerations and screening objectives). Regarding additional screening, the major difference is that the Pan-Canadian guideline and the ASCO adaptation focus on use of the PHQ-9 and GAD-7, while the Australian guidelines focus on use of the HADS. Although all of these measures have demonstrated their utility as screeners, they differ notably in item content. The PHQ-9 and GAD-7 feature items consistent with diagnostic criteria for mood and anxiety disorders, whereas the HADS features items designed to assess nonsomatic symptoms of depression and anxiety. These two different approaches reflect a major difference in how to approach screening. That is, should screening be conducted with measures of emotional distress developed specifically for medical populations (i.e., the HADS) or with measures developed specifically to capture symptoms of mental disorders in a wide range of populations? The answer would seem to lie in research that directly compares these two approaches in routine clinical practice. For example, a randomized trial could be conducted to examine whether differences occur in referral for full assessment and assignment to pathways when clinics employ the HADS versus the PHQ-9/GAD-7 and, if so, whether these differences have implications for the overall management of depression and anxiety in the patients screened. Future Directions This chapter has provided detailed information about the relative merits of several commonly used English-language instruments for assessment, screening, and case finding for depression and anxiety in cancer patients. Based on the evidence presented, several conclusions can be drawn and future directions suggested. First, the CES-D and HADS are the instruments that consistently received positive ratings across existing reviews. Among their notable features are the favorable ratings they earned for reliability,1,5 validity,1 availability of comparison data,1 ability to identify treatment effects,1 screening performance,1,5 and overall rating.1,5 That said, these two instruments are not without their weaknesses, especially when used for screening purposes. As previously noted, while the depression subscale of the HADS possesses good accuracy as a screening tool for depression, the performance of the anxiety subscale has been found to be inferior.6 Similarly, the HADS has performed reasonably well in identifying individuals without mental disorders but is not recommended to be used alone to identify individuals with suspected mood, anxiety, or mixed mental disorders.11 Thus, while these instruments have demonstrated psychometric rigor and some degree of diagnostic accuracy, research is still needed to identify those instruments optimally suited for assessment, screening, and case finding for depression and anxiety. Second, it is clear that no single instrument can be recommended for all possible applications. Clinicians and researchers must make choices depending on the circumstances for which they intend to use an instrument. In certain instances, the time available to complete an instrument will dictate the use of a very brief instrument (e.g., ESAS-r). In other instances, such as an intervention study, the demonstrated ability of an instrument to identify treatment effects (e.g., CES-D) may be the paramount consideration. In yet other instances, the demonstrated ability of an instrument to screen accurately for mental disorders (e.g., PHQ-9) may drive instrument selection. Accordingly, careful consideration of an instrument’s capabilities in relation to the intended use(s) should be the prime factor guiding instrument selection. Thus, research is needed to fully elucidate the clinical-and research-related factors that may systematically guide the selection of instruments for assessment, screening, and case finding. Third, existing systematic reviews of depression and anxiety assessment and screening instruments are characterized by a number of features that limit their utility. These features include lack of consistency across reviews in the methods used to select instruments CHAPTER 18 Assessment, Screening, and Case Finding for Depression and Anxiety in People with Cancer for evaluation, the features of each instrument evaluated, and the criteria used to evaluate instruments on those features. With respect to future directions, ongoing, systematic efforts to introduce greater standardization in the reporting of psychometric properties of assessment instruments should facilitate more consistent approaches in future systematic reviews. One effort notable in this regard is the Consensus-based Standards for the Selection of Health Measurement Instruments (COSMIN) initiative, which has developed a standardized taxonomy of instrument properties, along with standardized definitions and ratings.12,13 Another important limitation of many existing systematic reviews is their exclusive focus on studies published in English, based on instruments written in English. With the ever-increasing international scope of psychosocial oncology, information about instruments available in languages other than English is increasingly needed. Beyond information about availability, clinicians and researchers need critical appraisals of the methods used to create versions in other languages. That is, information is needed about the extent to which versions in other languages have been created using recommended procedures for translation and cultural adaptation of patient-reported outcome measures.14 Once created in other languages, the new versions then also need to be revalidated because it cannot be assumed that an instrument’s properties in one language and cultural group carry over to its properties in another language and cultural group.15 A final conclusion is that clinicians and researchers need to stay abreast of ongoing work that is likely to influence future recommendations for depression and anxiety assessment and screening in cancer patients. Along these lines, additional research is needed to clarify the utility of existing assessment instruments, such as the State Trait Anxiety Inventory16 and the Beck Depression Inventory II,17 which have received limited attention to date in the psychosocial oncology literature. Future recommendations may also be influenced by research evaluating the utility of several newly developed assessment and screening instruments when administered to cancer patients. An important development in this regard is the Patient- Reported Outcomes Measurement Information System (PROMIS) initiative.18 As part of this effort, item banks were developed for a number of constructs, including anxiety and depression.19 These item banks have been calibrated so that they can be used to create scales of varying length, including scales in which items are selected and presented to individuals based on their responses to previous items. This approach, known as computer- adaptive testing, is likely to grow in importance since it has the potential to reduce the number of items that need to be administered while simultaneously reducing measurement error.20 Another notable feature of PROMIS measures is the availability of cross-walk tables that permit estimation of scores on the CES-D, the BDI-II, and the PHQ-9 based on PROMIS depression scores as well as estimation of scores on the GAD-7, Mood and Anxiety Symptom Questionnaire (MASQ), and Positive and Negative Affect Scale (PANAS) based on PROMIS anxiety scores.21,22 Many of these advances, which reflect the application of item response theory to development and administration of patient-reported outcomes measures, will spur future research and have the potential to dramatically change future approaches to assessment, screening, and case finding for depression and anxiety in people with cancer. REFERENCES 1. Luckett T, Butow PN, King MT, et al. A review and recommendations for optimal outcome measures of anxiety, depression, and general distress in studies evaluating psychosocial interventions for English-speaking adults with heterogeneous cancer diagnoses. Support Care Cancer. 2010;18:1241–1262. 2. Mitchell AJ, Lord K, Symonds P. Which symptoms are indicative of DSMIV depression in cancer settings? An analysis of the diagnostic significance of somatic and non-somatic symptoms. J Affective Dis. 2012;138:137–148. 3. Jones SM, Ludman EJ, McCorkle R, et al. A differential item function analysis of somatic symptoms of depression in people with cancer. J Affective Dis. 2015;170:131–137. 4. Wakefield CE, Butow PN, Aaronson NA, et al. Patient-reported depression measures in cancer: a meta-review. Lancet Psychiatry. 2015;2:635–647. 5. Vodermaier A, Linden W, Siu C. Screening for emotional distress in cancer patients: a systematic review of assessment instruments. J Natl Cancer Inst. 2009;101:1464–1488. 6. Vodermaier A, Millman RD. Accuracy of the Hospital Anxiety and Depression Scale as a screening tool in cancer patients: a systematic review and meta- analysis. Support Care Cancer. 2011;19:1899–1908. 7. Esser P, Hartung TJ, Friedrick M, et al. The Generalized Anxiety Disorder Screener (GAD-7) and the anxiety module of the Hospital and Depression Scale (HADS-A) as screening tools for generalized anxiety disorder among cancer patients. Psychooncology. 2018;27:1509–1516. 8. Howell D, Keshavarz H, Esplen MJ, et al. A Pan Canadian Practice Guideline: Screening, Assessment and Care of Psychosocial Distress, Depression, and Anxiety in Adults with Cancer. Toronto: Canadian Partnership Against Cancer and the Canadian Association of Psychosocial Oncology; 2015. 9. Andersen BL, DeRubeis RJ, Berman BS, et al. Screening, assessment, and care of anxiety and depressive symptoms in adults with cancer: an American Society of Clinical Oncology guideline adaptation. J Clin Onc. 2014;32:1605. 10. Butow P, Price MA, Shaw JM, et al. Clinical pathway for the screening, assessment and management of anxiety and depression in adult cancer patients: Australian guidelines. Psychooncology. 2015;24:987–1001. 11. Mitchell AJ, Meader N, Symonds P. Diagnostic validity of the Hospital Anxiety and Depression Scale (HADS) in cancer and palliative settings: a meta-analysis. J Affective Dis. 2010;126:335–348. 12. Mokkink LB, Terwee CB, Patrick DL, et al. The COSMIN study reached international consensus on taxonomy, terminology, and definitions of measurement properties for health-related patient- reported outcomes. J Clin Epidemiol. 2010;63:737–745. 13. Terwee CB, Mokkink LB, Knol DL, Ostelo RWJG, Bouter LM, de Vet HCW. Rating the methodological quality in systematic reviews of studies on measurement properties: a scoring system for the COSMIN checklist. Qual Life Res. 2012;21:651–657. 14. Wild D, Grove A, Martin M, et al. Principles of good practice for the translation and cultural adaptation process for patient-reported outcomes (PRO) measures: report of the ISPOR task force for translation and cultural adaptation. Value Health. 2005;8:94–104. 15. Eremenco SL, Cella D, Arnold BJ. A comprehensive method for the translation and cross-cultural validation of health status questionnaires. Eval Health Prof. 2005;28:212–232. 135 136 SECTION IV Screening and Assessment in Psychosocial Oncology 16. Spielberger CD, Gorsuch RL, Lushene R, Vagg PR, Jacobs GA. Manual for the State- Trait Anxiety Inventory. Palo Alto, CA: Consulting Psychologists; 1983. 17. Beck AT, Steer RA, Brown GK. BDI-II Manual. San Antonio, TX: Psychological Corporation; 1996. 18. Cella D, Riley W, Stone A, et al. The Patient-Reported Outcomes Measurement Information System (PROMIS) developed and tested its first wave of adult self-reported health outcome item banks: 2005–2008. J Clin Epidemiol. 2010;63:1179–1194. 19. Pilkonis PA, Choi SW, Reise SP, et al. Item banks for measuring emotional distress from the Patient-Reported Outcomes Measurement Information System (PROMIS): depression, anxiety, and anger. Assessment. 2011;8:263–283. 20. Weiss DJ. Computerized adaptive testing for effective and efficient measurement in counseling and education. Meas Eval Couns Dev. 2004;37:70–84. 21. Choi SW, Schalet B, Cook KF, Cella D. Establishing a common metric for depressive symptoms: linking the BDI-II, CES-D, and PHQ-9 to PROMIS Depression. Psychol Assess. 2014;26:513–527. 22. Schalet BD, Cook KF, Choi SW, Cella D. Establishing a common metric for self-reported anxiety: linking the MASQ, PANAS, and GAD-7 to PROMIS Anxiety. J Anxiety Disord. 2014;28:88–96. 19 Screening for Delirium and Dementia in the Cancer Patient Christian Bjerre-Real, James C. Root, Yesne Alici, Julia A. Kearney, and William S. Breitbart Introduction Cognitive syndromes are commonly encountered among cancer patients at all age groups. Although aging is an important risk factor for cognitive disorders, in oncology settings there are additional risk factors, including primary or metastatic brain tumors, medical comorbidities, cancer treatments, depression, and anxiety, that place all age groups at risk for varying degrees of cognitive impairment at different stages of illness trajectory. Delirium, an acute-onset and common cognitive syndrome among hospitalized cancer patients, has been shown to increase risk of long-term cognitive impairment. Delirium can be seen in acutely ill cancer patients across the lifespan. Patients with underlying cognitive deficits are at increased risk for delirium. It is strongly recommended to routinely screen for delirium in inpatient oncology settings to identify patients with delirium in a timely manner, to treat underlying etiologies, and to prevent adverse outcomes associated with prolonged delirium. Dementia syndromes, referred to as major neurocognitive disorders in the Diagnostic and Statistical Manual of Mental Disorders, 5th edition (DSM 5),1 are routinely screened for as part of the comprehensive geriatric assessment among older cancer patients. Patients with cognitive deficits may not be able to carry out activities of daily living, such as medication maintenance, food preparation, and transportation. Treatment adherence, vigilance in monitoring and reporting of cancer and cancer treatment– related symptoms, and the ability to make informed decisions on cancer treatment options necessitate intact cognitive functioning. Therefore, screening for cognitive syndromes among cancer patients is an important part of initial assessment, particularly in high-risk populations. This chapter will provide a comprehensive review of the screening and assessment measures used to identify and assess patients with delirium and dementia. A brief overview of common dementia syndromes is also included as relevant to psycho- oncologists caring for older adults with cancer. An overview of cancer and cancer treatment–related cognitive impairment is provided in Chapter 36. Delirium Delirium is the most common cognitive syndrome encountered among hospitalized cancer patients. The prevalence of delirium ranges between 25% and 40% among cancer patients, with higher rates (i.e., up to 85%) occurring among the terminally ill.2 A detailed overview of delirium in cancer patients is provided in Chapter 44. In the following sections, we review screening and assessment of delirium among cancer patients, along with a review of the screening and assessment tools available for use in cancer patients. Delirium: Screening and Assessment Delirium is an acute change in level of alertness, awareness, attention, cognition, and behavior secondary to a general medical condition or medications. Sleep-wake cycle changes, disordered thought process, incoherent speech, psychomotor retardation or agitation, and perceptual disturbances are associated features of delirium. Patients with psychomotor retardation are frequently mistaken for suffering from depression by clinicians. Emergent psychiatry consults for patients with suicidal ideation not uncommonly reveal cases with unrecognized delirium. Cognitive disturbances in delirium can consist of myriad deficits, including disorientation, executive dysfunction, memory impairment, visuospatial deficits, and language disturbances. History obtained from family, staff, and other caregivers is critical for delirium diagnosis. Acute onset and fluctuating course are the main distinguishing features of delirium from dementia and other cognitive syndromes. Physical exam should focus on signs of infection, sepsis, dehydration, hypoxia, withdrawal states, and intoxication. Examination for focal neurological findings, myoclonus, and frontal release signs are the essential components of neurological exam in patients with delirium. Laboratory testing including comprehensive metabolic panel, complete blood count, vitamin B12 and thiamine levels, thyroid function tests, urinalysis, blood cultures, and brain imaging should be considered, based on history and physical examination findings, to identify underlying medical etiologies. Review of the medication 138 SECTION IV Screening and Assessment in Psychosocial Oncology list and recent dose changes and comparison with home medication lists should be completed to rule out delirium due to medications. An electroencephalogram (EEG) may be helpful in diagnosing nonconvulsive status epilepticus that may solely present with acute- onset mental status changes. Generalized slowing on EEG can be helpful in differentiating delirium from depression, where such a finding would not be expected. Patients with dementia can also present with generalized slowing findings on EEG. The gold standard diagnosis of delirium is the clinician’s assessment based on the DSM 5 delirium diagnostic criteria.1 There are several assessment and rating scales developed specifically for delirium, each with its own strengths and weaknesses (see Table 19.1).3,4 The Confusion Assessment Method (CAM), developed by Inouye and colleagues in 1990, provides a screening algorithm with four items (acute onset and fluctuating course, inattention, and either disorganized thinking or altered level of consciousness) that can be used by nurses and others with less advanced psychiatric training.5 However, the sensitivity of the CAM can be significantly affected by the experience of the user. Estimated administration time is under five minutes. The CAM is not useful in assessing the severity of delirium, but the CAM-S, a recently developed version of the CAM, can be used to assess delirium severity among medically ill older adults.6 At our institution, the CAM is routinely administered by bedside nurses every shift to all adult patients to screen for delirium. Ely and colleagues developed the Confusion Assessment Method for ICU (CAM-ICU) for use in nonverbal patients, such Table 19.1. Overview of Commonly Used Delirium Screening and Assessment Scales Confusion Assessment Method (CAM)5 ♦ ♦ ♦ ♦ ♦ ♦ ♦ Delirium Rating Scale-R-989 ♦ Delirium Rating Scale-R-98 was developed by Trzepacz and colleagues to address shortcomings of the Delirium ♦ ♦ ♦ ♦ ♦ ♦ Memorial Delirium Assessment Scale (MDAS)11,35 ♦ ♦ ♦ ♦ ♦ ♦ ♦ ♦ ♦ ♦ Nursing Delirium Screening Scale (Nu-DESC)31 Rating Scale, including the inability to distinguish between subtypes of delirium, lack of a separate item for attention deficits, and lack of clarification for “clouding of consciousness” Includes 16 clinician-rated items, 13 for severity and 3 for diagnosis. The rating is applicable to the preceding 24 hours It successfully differentiates delirium from dementia, depression, and schizophrenia Administered by trained clinicians It has excellent psychometric properties It is mostly used for phenomenological and treatment research purposes It has been validated in different languages MDAS assesses for the subtype and severity of delirium MDAS has 10 items, rated from 0 (none) to 3 (severe), with a maximum possible score of 30 A score of 13 has been recommended as a cutoff for establishing the diagnosis of delirium in oncology settings A cutoff score of 7 has yielded the highest sensitivity and specificity rates for delirium diagnosis in palliative care settings25 It has excellent psychometric properties It distinguishes between patients with delirium, dementia, or no cognitive impairment It has been validated in a number of different languages It is the most widely used delirium assessment scale in oncology and palliative care settings Physician-rated Takes about 10–15 minutes to administer ♦ Nu-DESC has been adapted from a former delirium assessment tool, namely the Confusion Rating Scale ♦ Composed of 5 items including orientation, behavior, communication, perceptual disturbances, and psychomotor ♦ ♦ ♦ ♦ ♦ ♦ Delirium Observation Scale (DOS)32 Most commonly used delirium screening instrument in general medical settings, specifically among older adults Four-item algorithm can be used to quickly screen for delirium Requires well-trained raters Excellent psychometric properties when used by well-trained raters Cognitive testing is required to assess the cognitive items of the scale Validated for use in a number of languages For severity assessment, CAM-S has been developed retardation It allows for continuous symptom assessment It has been validated for use in oncology settings Administered by nursing Takes about 1–2 minutes It has good psychometric properties It has been validated in other languages ♦ Adapted from the Delirium Observational Screening Scale ♦ Composed of 13 items including orientation, attention, thought organization, perceptual disturbances, psychomotor ♦ ♦ ♦ ♦ retardation, and behavior It allows for multiple nursing shifts assessment and quantification Administered by nursing Takes about 1–2 minutes It has been used in the oncology setting Single Question in Delirium (SQID)16,17 ♦ Single question meant to be asked to the caregiver: ♦ “Do you feel that [patient’s name] has been more confused lately?” ♦ Tested against CAM and psychiatric interview Cornell Assessment of Pediatric Delirium (CAPD)12 ♦ ♦ ♦ ♦ 8-question rating scale performed by nursing staff in the PICU Takes less than 2 minutes to complete Excellent overall sensitivity and specificity Loses specificity with developmental delay, but sensitivity remains high CHAPTER 19 Screening for Delirium and Dementia in the Cancer Patient as those who are mechanically ventilated.7 It is important to limit its use to nonverbal, mechanically ventilated patients in intensive care unit settings due to its low sensitivity in detecting delirium in other settings.8 The Delirium Rating Scale-Revised 98 (DRS-R-98) is a 16-item scale (13 severity and 3 diagnostic items) that can be used to measure severity of delirium as well as to differentiate delirium from dementia, depression, or schizophrenia. It is designed to be administered by an experienced expert and has been found to be highly sensitive.9 The Delirium Observation Scale (DOS) is a nursing-rated scale based on 13 observations that can be useful in the screening of delirium and has shown promising results in its detection in the advanced cancer patient population.10 The Memorial Delirium Assessment Scale (MDAS) is a physician-rated scale that can be used to measure the severity of delirium in addition to diagnosis of delirium (see Box 19.1). It has 10 items and takes under 15 minutes to complete. This instrument was designed to be administered at several time points in the same day to assess changes in delirium severity.11 The MDAS and DRS-98 include items to assess cognitive impairment; therefore, additional cognitive assessment tools are not required when assessing delirium with them. A cognitive assessment tool is required to reliably assess a delirious patient’s attention disturbances and other cognitive impairment when using the CAM algorithm to diagnose delirium. The Cornell Assessment of Pediatric Delirium (CAPD)12 is a valid, rapid, observational nursing screen that is used for detection of delirium in the pediatric age group. The CAPD screen is designed to allow for behavioral and developmentally informed observations to be scaled and summarized in a total score, which indicates whether a child is likely to be delirious. Adapted from the CAM-ICU, two additional pediatric tools have been developed for the direct assessment of children under 5 years old (psCAM-ICU)13 and children over 5 years old (pCAM-ICU).14 The supportive educational videos on the icudelirium.org website are a key educational tool in teaching clinicians how to recognize delirium in infants and children.15 The Single Question in Delirium (SQiD) is a single directed question to the caregiver asking whether the patient has been more confused than usual. This screening method showed moderate sensitivity/specificity.16,17 Future Directions: Recommendations for Research on Screening and Assessment of Delirium in Oncology Settings Delirium is associated with increased morbidity and mortality, with increasing evidence for long-term cognitive impairment following an episode of delirium. A meta-analysis by Witlox and colleagues found that delirium in elderly patients is associated with increased risk of death (38% vs. 28% in controls) at an average follow-up of 23 months, increased risk of institutionalization (33% vs. 11%) at an average follow-up of 15 months, and increased risk of dementia (63% vs. 8%) at an average follow-up of 4 years.18 The growing data on short-term and long-term outcomes of delirium strongly support the need to screen for delirium among hospitalized patients. As more healthcare data becomes digitized in an accessible way through the electronic health record, opportunities for the development of delirium predictive risk models based on multivariable analysis through deep learning algorithms become a possibility. Developing such models for the development of delirium in the cancer population will help identify patients at highest risk for delirium who might benefit most from delirium prevention interventions. Current guidelines on the assessment of delirium in cancer patients, including the assessment of etiologies, risk factors, and phenomenology, are largely based on the assessment of older adults with delirium. Evidence-based screening and assessment guidelines in oncology and palliative care settings continue to emerge and will improve delirium care in this patient population. Dementia Dementia, referred to as major neurocognitive disorders in DSM 5,1 is a progressive cognitive decline characterized by impairment in cognitive functioning (e.g., memory, language, executive functioning, visuospatial skills) that interferes with the ability to perform activities of daily functioning independently. It is estimated that dementia affects between 2.4 million and 5.5 million people in the United States.19 Cancer patients with cognitive impairment are at increased risk of functional dependence and medication nonadherence and are at greater risk of death. Cognitive and functional assessment for mild cognitive impairment and dementia is recommended for all older adults with cancer. The National Comprehensive Cancer Network (NCCN) Older Adult Oncology Guidelines(2020)20 recommend all cognitively impaired patients to be cared for by a multidisciplinary geriatric oncology team throughout their treatment. For patients with suspected impaired cognitive function, or for patients with self-reported or family-reported concerns for cognitive impairment, a consultation with a cognitive disorders specialist is recommended. The NCCN guidelines recommend periodic reassessment of cognitive functioning for those with cognitive impairment and for all older adults when considering changes to the treatment plan. In this section, we will provide a review of screening and assessment for dementia syndromes and a brief overview of different cognitive syndromes. Dementia: Screening and Assessment The initial assessment of an individual suspected of having any cognitive impairment starts with a detailed history with input from a family member or caregiver who can provide information on the patient’s previous level of functioning, the temporal course and rate of cognitive decline, and any behavioral or functional changes noted along with cognitive impairment. The time course and associated symptoms represent the key information in distinguishing between different forms of cognitive disorders and ruling out depression or delirium. For neurocognitive disorders such as dementia, the onset of symptoms is gradual and progressive as opposed to the abrupt and rapid onset usually seen in delirium.4 As part of the initial assessment, there should be a review of the patient’s baseline and current ability to carry out his or her instrumental activities of daily living (IADLs). IADLs include use of the telephone, shopping, food preparation, housekeeping, laundry, grooming, mode of transportation, responsibility for medications, and handling of finances. It is important to find out to what degree the impairment is secondary to physical impairments, as opposed to a cognitive impairment. Asking a caregiver if he or she has 139 140 SECTION IV Screening and Assessment in Psychosocial Oncology Box 19.1 Memorial Delirium Assessment Scale (MDAS) Item 1. Reduced Level of Consciousness (Awareness): Rate the patient’s current awareness of and interaction with the environment (interviewer, other people/objects in the room; e.g., ask patients to describe their surroundings). 0: none (patient spontaneously fully aware of environment and interacts appropriately) 1: mild (patient is unaware of some elements in the environment, or not spontaneously interacting appropriately with the interviewer; becomes fully aware and appropriately interactive when prodded strongly; interview is prolonged but not seriously disrupted) 2: moderate (patient is unaware of some or all elements in the environment, or not spontaneously interacting with the interviewer; becomes incompletely aware and inappropriately interactive when prodded strongly; interview is prolonged but not seriously disrupted) 3: severe (patient is unaware of all elements in the environment with no spontaneous interaction or awareness of the interviewer, so that the interview is difficult to impossible, even with maximal probing). Item 2. Disorientation: Rate current state by asking the following 10 orientation items: date, month, day, year, season, floor, name of hospital, city, state, country. 0: none (patient knows 9–10 items) 1: mild (patient knows 7–8 items) 2: moderate (patient knows 5–6 items) 3: severe (patient knows no more than 4 items) Item 3. Short-Term Memory Impairment: Rate current state by using repetition and delayed recall of 3 words (patient must immediately repeat and recall words 5 minutes later after an intervening task. Use alternate sets of 3 words for successive evaluations [e.g., apple, table, tomorrow; sky, cigar, justice]). 0: none (all 3 words repeated and recalled) 1: mild (all 3 repeated, patient fails to recall 1) 2: moderate (all 3 repeated, patient fails to recall 2–3) 3: severe (patient fails to repeat 1 or more words) Item 4. Impaired Digit Span: Rate current performance by asking subjects to repeat first 3, 4, then 5 digits forward and then 3, then 4 backward; continue to the next step only if patient succeeds at the previous one. 0: none (patient can do at least 5 numbers forward and 4 backward) 1: mild (patient can do at least 5 numbers forward, 3 backward) 2: moderate (patient can do 4–5 numbers forward, cannot do 3 backward) 3: severe (patient can do no more than 3 numbers forward) Item 5. Reduced Ability to Maintain and Shift Attention: As indicated during the interview by questions needing to be rephrased and/ or repeated because patient’s attention wanders, patient loses track, or patient is distracted by outside stimuli or is overabsorbed in a task. 0: none (none of the above; patient maintains and shifts attention normally) 1: mild (above attentional problems occur once or twice without prolonging the interview) 2: moderate (above attentional problems occur often, prolonging the interview without seriously disrupting it) 3: severe (above attentional problems occur constantly, disrupting and making the interview difficult to impossible) Item 6. Disorganized Thinking: As indicated during the interview by rambling, irrelevant, or incoherent speech or by tangential, circumstantial, or faulty reasoning. Ask patient a somewhat complex question (e.g., Describe your current medical condition). 0: none (patient’s speech is coherent and goal directed) 1: mild (patient’s speech is slightly difficult to follow; responses to questions are slightly off target but not so much as to prolong the interview) 2: moderate (disorganized thoughts or speech are clearly present, such that interview is prolonged but not disrupted) 3: severe (examination is very difficult or impossible due to disorganized thinking or speech) Item 7. Perceptual Disturbance: Misperceptions, illusions, hallucinations inferred from inappropriate behavior during the interview or admitted by subject, as well as those elicited from nurse/family/chart accounts of the past several hours or of the time since last examination: 0: none (no misperceptions, illusions, or hallucinations) 1: mild (misperceptions or illusions related to sleep, fleeting hallucinations on 1–2 occasions without inappropriate behavior) 2: moderate (hallucinations or frequent illusions on several occasions with minimal inappropriate behavior that does not disrupt the interview) 3: severe (frequent or intense illusions or hallucinations with persistent inappropriate behavior that disrupts the interview or interferes with medical care) Item 8. Delusions: Rate delusions inferred from inappropriate behavior during the interview or admitted by the patient, as well as delusions elicited from nurse/family/chart accounts of the past several hours or of the time since the previous examination. 0: none (no evidence of misinterpretations or delusions) 1: mild (misinterpretations or suspiciousness without clear delusional ideas or inappropriate behavior) 2: moderate (delusions admitted by the patient or evidenced by his or her behavior that do not or only marginally disrupt the interview or interfere with medical care) 3: severe (persistent and/or intense delusions resulting in inappropriate behavior, disrupting the interview or seriously interfering with medical care) Item 9. Decreased or Increased Psychomotor Activity: Rate activity over past several hours, as well as activity during interview, by circling a: hypoactive b: hyperactive c: elements of both present 0: none (normal psychomotor activity) a b c 1: mild (Hypoactivity is barely noticeable, expressed as slightly slowing of movement. Hyperactivity is barely noticeable or appears as simple restlessness.) a b c 2: moderate (Hypoactivity is undeniable, with marked reduction in the number of movements or marked slowness of movement; subject rarely spontaneously moves or speaks. Hyperactivity is undeniable, subject moves almost constantly; in both cases, exam is prolonged as a consequence.) a b c 3: severe (Hypoactivity is severe; patient does not move or speak without prodding or is catatonic. Hyperactivity is severe; patient is constantly moving, overreacts to stimuli, requires surveillance and/ or restraint; getting through the exam is difficult or impossible.) Item 10. Sleep-Wake Cycle Disturbance (Disorder of Arousal): Rate patient’s ability to either sleep or stay awake at the appropriate times. Utilize direct observation during the interview, as well as reports from nurses, family, patient, or charts describing sleep-wake cycle disturbance over the past several hours or since last examination. Use observations of the previous night for morning evaluations only. 0: none (at night, sleeps well; during the day, has no trouble staying awake) 1: mild (mild deviation from appropriate sleepfulness and wakefulness states: at night, difficulty falling asleep or transient night awakenings, needs medication to sleep well; during the day, reports periods of drowsiness or, during the interview, is drowsy but can easily fully awaken him-or herself) 2: moderate (moderate deviations from appropriate sleepfulness and wakefulness states: at night, repeated and prolonged night awakening; during the day, reports of frequent and prolonged napping or, during the interview, can only be roused to complete wakefulness by strong stimuli) 3: severe (severe deviations from appropriate sleepfulness and wakefulness states: at night, sleeplessness; during the day, patient spends most of the time sleeping or, during the interview, cannot be roused to full wakefulness by any stimuli) Adapted from Breitbart W, Rosenfeld B, Roth A, Smith MJ, Cohen K, Passik S. The Memorial Delirium Assessment Scale. J Pain Symptom Manag. 1997 Mar;13(3):128–137. CHAPTER 19 Screening for Delirium and Dementia in the Cancer Patient concerns for the patient’s safety can also provide key information regarding the level of impairment. The history should additionally include review of exposures to risk factors associated with cognitive impairment, such as history of alcohol or illicit substance use, as well as exposure to toxins. A thorough review of a patient’s medications, including use of supplements and over-the-counter drugs, must be completed. Opioids, benzodiazepines, and anticholinergic medications are often associated with cognitive impairment in vulnerable patients. Cancer type and cancer-related treatments received by the patient should be reviewed as potential contributors to cognitive impairment. Chemotherapeutic agents, radiation, hormone therapies, immunotherapies, and surgical treatments have all been associated with cognitive changes (see Chapter 36).21 Clinicians should inquire about history of chronic medical conditions such as hypertension, vascular disease, hyperlipidemia, diabetes mellitus, and obesity, as they have all been shown to increase the risk of cognitive impairment. A history of psychiatric illnesses should also be reviewed. Patients with major depressive disorder can often present with mild forgetfulness, previously termed “pseudo-dementia.” Feelings of sadness, dysphoria, hopelessness, helplessness, anhedonia, lack of motivation, appetite disturbance, sleep disturbance, and suicidality are supportive of a diagnosis of depression, as opposed to dementia or other cognitive syndromes. In depressed patients, the cognitive impairments tend to be mild and would have not been present prior to the onset of depression. During cognitive testing the depressed patient often responds, “I don’t know.” Indifference to cognitive deficits is common among depressed patients, anosognosia is common in Alzheimer’s disease, and indifference or ignorance of impairment is typical in frontotemporal dementia. Minimization of even self-recognized deficits is common in most. Fatigue secondary to cancer and cancer-related treatments is commonly encountered among cancer patients and may contribute to cognitive dysfunction. Patients suffering from severe fatigue could appear inattentive and sedated and can present with short-term memory impairments. Once the fatigue is managed properly, cognitive deficits should resolve. A comprehensive physical examination with emphasis on neurological signs should be performed. The presence of asterixis and frontal release signs like a palmomental reflex or grasp reflex may be observed in cases of delirium or advanced dementia. Ataxia and focal motor or sensory findings may be present in dementia, especially in the presence of brain lesions or vitamin deficiencies. Laboratory testing, including a urinalysis; basic metabolic panel; lipid profile; liver function tests; thyroid function tests; syphilis screen; HIV test; paraneoplastic panel; vitamin levels including vitamin B12, thiamine, and folate; and cerebrospinal fluid are useful in identifying reversible causes of the cognitive changes if clinically indicated. Neuroimaging, such as a computed tomography (CT) scan or magnetic resonance imaging (MRI), is needed to evaluate for potential structural causes of the impairment. Information obtained through history and physical examination should guide clinicians as to the extent of laboratory testing and neuroimaging. An important component of the evaluation is the objective assessment of an individual’s cognitive abilities, either with a screening tool in the case of overt cognitive dysfunction or with comprehensive neuropsychological assessment in cases of lesser severity where higher sensitivity to milder cognitive dysfunction is required. This can be performed in the office or at the bedside with the use of a cognitive assessment tool. Cognitive screening tools (see Chapter 36, Box 36.1), such as the Montreal Cognitive Assessment (MoCA)22 and the Mini-Mental Status Exam (MMSE),23 are designed to test multiple domains of cognition, including short-term memory, language, generative naming, visuospatial skills, and executive functioning. Screening measures generally specify cutoffs for normal versus abnormal performance. The NCCN Older Adult Oncology Guidelines Version 2020 continue to indicate Mini- Cog as a screening tool for cognitive impairment in older adults with cancer.20,24 Cognitive tests can provide a sense of how severe the impairment is and can assist in making a diagnosis. Repeated administration of the same cognitive assessment tool at different time points is important in tracking improvements or the rate of further decline in cognition. Comprehensive neuropsychological testing may be required in most patients with cancer to differentiate between cancer and cancer-related cognitive changes, neurodegenerative diseases, and vascular and other forms of cognitive syndromes.25,26 Common Cognitive Syndromes In this section, we will review commonly encountered cognitive syndromes in oncology settings, particularly among older adults, the age group most commonly affected by cancer. Table 19.2 lists the commonly used dementia screening and assessment tools. Dementia Due to Alzheimer’s Disease Alzheimer’s disease is the most common form of dementia. The most significant risk factor for Alzheimer’s disease is age, with incidence of disease doubling every five years after age 65.27,28 In Alzheimer’s disease, the cognitive and behavioral symptoms typically have an insidious onset and gradual progression. Early in the course the predominant feature is memory loss. Patients typically preserve social cognition and procedural memory until very late. Deficits in language, visuospatial impairments, and executive function are typically seen as the disease progresses. Noncognitive neuropsychiatric disturbances, including apathy, irritability, agitation, restlessness, anxiety, and depression, are commonly seen in patients with Alzheimer’s disease. Patients with advanced Alzheimer’s dementia can present with aggression and psychosis.4 The brain pathology in Alzheimer’s is characterized by neuronal loss and an accumulation of misfolded proteins, known as neurofibrillary tangles and amyloid plaques.27 Genetic predisposition has been identified in early-onset cases, and commercial testing for mutations of the known Alzheimer’s disease genes, such apolipoprotein E (APOE), presenilin 1 (PSEN1), or presenilin 2 (PSEN2), is available. The use of positron emission tomography (PET) scans to detect amyloid plaque density and testing of cerebrospinal fluid for amyloid and tau levels may be useful in diagnosing cases of Alzheimer’s when the clinical picture is unclear.4 Vascular Dementia Vascular dementia is the second most common form of dementia seen in older adults. In patients with vascular dementia, the onset of cognitive symptoms typically coincides with cerebrovascular events. 141 142 SECTION IV Screening and Assessment in Psychosocial Oncology Table 19.2. Overview of Commonly Used Clinician-Rated Dementia Screening and Assessment Scales MMSE23 MINICOG24 MOCA22 SLUMS33 Freund Clock Drawing Test34 Domains tested Orientation, attention, comprehension, calculations, memory, language, visuospatial ability Memory, language comprehension, visual- motor skills, executive function Orientation, memory, clock, visuospatial ability, fluency, language, abstraction, calculations, executive function, attention Orientation, memory, animal fluency, attention, clock drawing Memory, executive function, visuospatial ability Advantages One of the most commonly used cognitive assessment tools Not strongly influenced by Better testing of education executive functioning, more sensitive to mild impairment, freely available online in a number of different languages Emphasizes memory tasks, good for distinguishing between mild cognitive impairment and dementia Not influenced by education. Seven-point scoring of the clock draw allows for improved interrater reliability and improved identification of cognitive deficits Disadvantages Can miss deficits in well educated. Limited executive function testing. Not freely available Scoring of clock drawing test is vulnerable to different interpretations. Cannot be used in visually impaired patients and patients with impaired manual dexterity Cannot distinguish Less evaluation of Cannot be used in visually between mild cognitive language, constructions, impaired patients and impairment and dementia and executive functioning patients with impaired manual dexterity Time to administer 10–15 minutes 5 minutes 10–15 minutes The cognitive decline is especially seen in areas of executive function and working memory. Neuroimaging is helpful in identifying the presence of cerebrovascular disease, which can range from large strokes to small vessel disease. The cognitive impairments usually occur in a stepwise fashion, with periods of decline followed by periods of stability and eventually further decline. Symptoms such as mood and personality changes, abulia, and emotional lability may be seen. Patients often initially present with depressive symptoms and on further screening are found to have deficits in executive functioning. The condition is thought to be secondary to atherosclerosis and arteriosclerosis in the cerebral vessels. The common risk factors include dyslipidemia, hypertension, diabetes mellitus, smoking, obesity, and atrial fibrillation, as well as other conditions that increase the risk of cerebral emboli. Frontotemporal Dementia Frontotemporal dementia is the most common cause of early- onset dementia in individuals younger than 65 years old, with the behavioral variant accounting for approximately 50% of cases, and nonfluent and semantic variants the rest. Frontotemporal dementia is characterized by the gradual and progressive development of personality changes, as well as behavioral and/or language impairment. The hallmark behavioral changes are apathy or disinhibition. Patients can lose interest in self-care and socialization and can often present with socially inappropriate behaviors. These patients are sometimes noted to be hyper-oral, and changes in their diet can be observed. Patients who develop personality changes can have changes in their social style and beliefs. Cognitive deficits are less prominent in frontotemporal dementia early in the course of the disease; as the disease progresses, executive dysfunction (i.e., changes in planning and organizing, abstraction, and cognitive flexibility) is more common. These patients can demonstrate poor judgment and are easily distracted. Cognitive testing frequently shows impairments in abstract 10 minutes 5 minutes reasoning, response inhibition, and mental flexibility. Learning, memory, and perceptual-motor abilities tend to be spared, especially in early stages. Neuroimaging will reveal a distinct pattern of atrophy of the frontal lobes. Lewy Body Dementia Lewy body dementia is the second most common type of neurodegenerative dementia after Alzheimer’s disease. Lewy body dementia is associated with an insidious onset and gradual progression of cognitive decline. The condition can present with fluctuating cognition with pronounced variations in attention and alertness, recurrent visual hallucinations that are often well formed and detailed, and features of parkinsonism that appear after the cognitive decline. Patients with Lewy body dementia are prone to frequent falls, syncope, and autonomic dysfunction. Since visual hallucinations may be common in presentation, Lewy body dementia cases may be misidentified as a primary psychotic disorder. This is an important distinction, since Lewy body cases are very sensitive to treatment with antipsychotics and their use may lead to neuroleptic malignant syndrome (NMS). The arousal disturbance in these patients can appear similar to delirium, but the course of the decline is gradual, as opposed to an acute onset. Further medical workup does not reveal any underlying medical etiologies in patients with Lewy body dementia unless they have superimposed delirium. The disease is caused by misfolded proteins, specifically alpha-synuclein, and is referred to as a synucleinopathy. Dementia Due to Vitamin Deficiencies Vitamins such as cyanocobalamin (B12), thiamine (B1), and folate are essential for normal functioning of the brain and nervous system. These vitamins are essential to cell metabolism, DNA synthesis, and energy production. When deficient in any of these vitamins, one can develop myriad neurologic impairments, including dementia. If given a history of a patient with CHAPTER 19 Screening for Delirium and Dementia in the Cancer Patient cognitive deficits and poor nutritional status, persistent nausea and vomiting, or altered gastrointestinal absorption, as seen in gastric cancer or after bariatric surgery, one should consider a dementia due to vitamin deficiency. Early recognition of such cases is important because the cognitive impairments may be reversed with repletion of the vitamin. Gait disturbances, neuropathy, anemia, weakness, cognitive impairment, depression, and a red beefy tongue are some of the characteristic findings of patients with dementia due to vitamin B12 deficiency. Patients with folate deficiency may also present with mood disturbance, cognitive impairment, and anemia. Thiamine-deficient patients can develop Wernicke’s encephalopathy, a condition characterized by mental status changes, gait disturbance, and ophthalmoplegia. Wernicke’s encephalopathy has long been thought of as a condition limited to patients with alcoholism; however, recent literature has shown that the syndrome is not infrequently seen in those with poor nutrition. Frail patients with a malignancy, who often suffer from low appetite and vomiting, are especially vulnerable.29 Screening for deficiencies can be easily achieved by ordering blood vitamin levels, and repletion can be done by vitamin supplementation. Dementia Due to HIV Infection HIV disease is an infection caused by exposure to the human immunodeficiency virus type I. The route of transmission is contact with infected bodily fluids. The virus, once contracted, impairs functioning of CD-4 lymphocytes, cells crucial to a functioning immune system. The virus can affect a variety of brain regions, resulting in the development of cognitive impairment. Typically, the affected individual can present with deficits in executive functioning, processing speed, and attention; in severe cases, language, emotional control, and affect may be affected. Other neurologic impairments may also be present in severe cases, including incoordination, ataxia, and motor slowing. Dementia due to HIV is more commonly seen in patients with high viral load measured in the cerebrospinal fluid or those who have other signs of advanced HIV. The course of cognitive impairment can vary from rapid progression to slow decline to improvement or even resolution. One would not expect to observe an abrupt onset of mental status changes, which would more likely indicate a delirium, requiring a comprehensive workup to identify potential medical causes, with special attention to ruling out the presence of an opportunistic infection. It is important to note that HIV-infected patients may suffer from cognitive impairments even in the absence of full-blown AIDS. Mild Cognitive Impairment Mild cognitive impairment, referred to as “mild neurocognitive impairment” in the DSM 5,1 is a condition defined as a modest cognitive decline from an individual’s previous level of functioning, greater than expected for age and education level. The impairment does not affect one’s ability to sustain independence in activities of daily living, but the individual likely experiences a need for greater effort, accommodation, or strategies for coping with the deficit when managing complex tasks. Mild cognitive impairment is typically seen in adults over the age 65, with prevalence ranging from 3% to 19% of the population in that age group.30 Many patients with mild cognitive impairment will progress to developing dementia, and therefore they should be monitored closely. Factors like prolonged exposure to anticholinergic medications, cerebrovascular disease, misfolded protein deposition, and genetic mutation have been identified as potentially playing a role in the pathophysiology of the syndrome.31 Patients presenting with mild cognitive impairment may develop an Alzheimer’s dementia, a Lewy body dementia, or other types of cognitive syndromes depending on the underlying etiology. Patients with mild cognitive impairment secondary to cancer and cancer treatment–related cognitive changes may not progress to a full-blown dementia. This continues to be an area in need of further study among cancer patients (see Chapter 36). The cognitive dysfunction in patients with mild cognitive impairment may not be apparent in a casual conversation. Typically, those close to the patient describe the individual as now being forgetful of things they once recalled easily or of recent events that they would normally be interested in. Cognitive assessment tools, identical to those used in screening for dementia, can be used for diagnosing mild cognitive impairment, but these often lack the sensitivity to identify such subtle impairments. Comprehensive neuropsychological testing is mostly required to uncover the problem. Screening and diagnosing mild cognitive impairment are important, as this will help identify individuals potentially at risk of developing more severe impairment in the coming years. This can allow individuals, families, and healthcare providers an opportunity to plan more effectively for future treatments. Future Directions: Recommendations for Research on Screening and Assessment of Dementia in Oncology Settings In 2019, a systematic review by the US Preventive Services Task Force (USPSTF) concluded that several brief screening instruments can adequately detect cognitive impairment, especially in populations with a higher prevalence of underlying dementia. The review also concluded that there is no empirical evidence, however, that screening for cognitive impairment or early diagnosis of cognitive impairment improves patient, caregiver, family, or clinician decision-making or other important outcomes.19 It is important to note that the USPSTF review findings are applicable to healthy, community-dwelling older adults, and these findings highlight the need to improve research on outcomes and interventions for the cognitively impaired. In oncology settings, medical comorbidities, cancer and cancer treatments, comorbid depression, and increased prevalence of delirium predispose patients to cognitive deficits. Therefore, current guidelines recommend screening for cognitive impairment among older adults with cancer. Although older adults are at higher risk for cognitive impairment, adults with cancer are more likely to experience cognitive impairment than those without a cancer diagnosis. Therefore, screening for cognitive impairment among all patients with a cancer diagnosis should be considered. Future research should focus on identifying risk factors for cognitive impairment, how to best screen for cognitive impairment, studying morbidity and mortality outcomes of cognitive impairment, and clarifying the impact of cancer and cancer-related treatments on cognition among cancer patients of all age groups. The development of predictive models in the future can help discern cancer patients at higher risk for cognitive impairment. 143 144 SECTION IV Screening and Assessment in Psychosocial Oncology REFERENCES 1. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders: Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition. Arlington, VA: American Psychiatric Association, 2013. 2. Breitbart W, Alici Y. Agitation and delirium at the end of life: “We couldn’t manage him.” JAMA. 2008 Dec 24;300(24):2898–2910, E1. doi:10.1001/jama.2008.885. 3. Smith MJ, Breitbart WS, Platt MM. A critique of instruments and methods to detect, diagnose and rate delirium. J Pain Symptom Manag. 1995 Jan;10(1):35–77. 4. Scharre DW, Trzepacz PT. Evaluation of cognitive impairment in older adults. Focus. 2013 Oct 1;11(4):482–500. 5. Inouye SK, VanDyck CH, Alessi CA, Balkin S, Siegal AP, Horwitz RI. Clarifying confusion: The Confusion Assessment Method. A new method for detecting delirium. Ann Intern Med. 1990;113:941–948. 6. Inouye SK, Kosar CM, Tommet D, et al. The CAM-S: Development and validation of a new scoring system for delirium severity in 2 cohorts: The CAM-S Score for Delirium Severity. Ann Intern Med. 2014 Apr;160(8):526–533. 7. Ely EW, Inouye SK, Bernard GR, et al. Delirium in mechanically ventilated patients: Validity and reliability of the confusion assessment method for the intensive care unit (CAM-ICU). JAMA. 2001 Dec 5;286(21):2703–2710. 8. Neufeld KJ, Hayat MJ, Coughlin JM, et al. Evaluation of two intensive care delirium screening tools for non-critically ill hospitalized patients. Psychosomatics. 2011 Mar–Apr;52(2):133–140. 9. Trzepacz PT, Mittal D, Torres R, Kanary K, Norton J, Jimerson N. Validation of the Delirium Rating Scale-revised-98: Comparison with the delirium rating scale and the cognitive test for delirium. J Neuropsych Clin N. 2001 Spring;13(2):229–242. 10. Neefjes ECW, van der Vorst MJDL, Boddaert MSA, et al. Accuracy of the Delirium Observational Screening Scale (DOS) as a screening tool for delirium in patients with advanced cancer. BMC Cancer. 2019;19(1):160. 11. Breitbart W, Rosenfeld B, Roth A, Smith MJ, Cohen K, Passik S. The Memorial Delirium Assessment Scale. J Pain Symptom Manag. 1997 Mar;13(3):128–137. 12. Traube C, Silver G, Kearney J, et al. Cornell Assessment of Pediatric Delirium: A valid, rapid, observational tool for screening delirium in the PICU. Crit Care Med. 2014 Mar;42(3):656–663. doi:10.1097/CCM.0b013e3182a66b76 13. Smith HAB, Gangopadhyay M, Goben CM, et al. The Preschool Confusion Assessment Method for the ICU (psCAM-ICU): Valid and reliable delirium monitoring for critically ill infants and children. Crit Care Med. 2016 Mar;44(3):592–600. doi:10.1097/ CCM.0000000000001428 14. Smith HAB, Boyd J, Fuchs DC, et al. Diagnosing delirium in critically ill children: Validity and reliability of the Pediatric Confusion Assessment Method for the Intensive Care Unit. Crit Care Med. 2011 Jan;39(1):150–157. doi:10.1097/CCM.0b013e3181feb489 15. Critical Illness, Brain Dysfunction, and Survivorship (CIBS) Center. Pediatric Care. https://www.icudelirium.org/medical- professionals/pediatric-care. Accessed on 2/16/2020. 16. Lin H-S, Eeles E, Pandy S, Pinsker D, Brasch C, Yerkovich S. Screening in delirium: A pilot study of two screening tools, the Simple Query for Easy Evaluation of Consciousness (SQeeC) and Single Question in Delirium (SQiD). Australas J Ageing. 2015. doi:10.1111/ajag.12216 17. Sands MB, Danotc BP, Hartshom A, Ryan CJ, Lujic S. Single Question in Delirium (SQiD): Testing its efficacy against psychiatrist interview, the Confusion Assessment Method and the Memorial Delirium Assessment Scale. Palliat Med. 2010;24:561– 565. doi:10.1177/0269216310371556 18. Witlox J, Eurelings LS, de Jonghe JF, Kalisvaart KJ, Eikelenboom P, van Gool WA. Delirium in elderly patients and the risk of postdischarge mortality, institutionalization, and dementia: a meta-analysis. JAMA. 2010 Jul 28;304(4):443–451. 19. Patnode CD, Perdue LA, Rossom RC, Rushkin MC, Redmond N, Thomas RG, Lin JS. Screening for Cognitive Impairment in Older Adults: An Evidence Update for the U.S. Preventive Services Task Force [Internet]. AHRQ Publication No. 19-05257-EF-1v. Rockville, MD: Agency for Healthcare Research and Quality (US); 2019. 20. NCCN Older Adult Oncology Guidelines. https://www.nccn.org/ professionals/physician_gls/pdf/senior.pdf. Accessed on 2/ 16/ 2020. 21. Janelsins MC, Kesler SR, Ahles TA, Morrow GR. Prevalence, mechanisms, and management of cancer-related cognitive impairment. Int Rev Psychiatry. 2014 Feb;26(1):102–113. doi:10.3109/ 09540261.2013.864260 22. Nasreddine ZS, Phillips NA, Bédirian V, et al. The Montreal Cognitive Assessment, MoCA: A brief screening tool for mild cognitive impairment. JAGS. 2005 Apr;53:695–699. 23. Folstein MF, Folstein SE, McHugh PR. “Mini-mental state”: A practical method for grading the cognitive state of patients for the clinician. J Psychiat Res. 1975 Nov;12(3):189–198. doi:10.1016/ 0022-3956(75)90026-6. PMID 1202204 24. Borson S, Scanlan JM, Chen P, Ganguli M. The Mini-Cog as a screen for dementia: Validation in a population-based sample. J Am Geriatr Soc. 2003;51(10):1451–1454. 25. Lange M, Rigal O, Clarisse B, et al. Cognitive dysfunctions in elderly cancer patients: A new challenge for oncologists. Cancer Treat Rev. 2014 Jul;40(6):810–817. pii:S0305-7372(14)00046-2. doi:10.1016/j.ctrv.2014.03.003 26. Ahles TA. Cognitive changes associated with cancer and cancer treatment. Semin Oncol Nurs. 2013 Nov;29(4):229–231. 27. Querfurth HW, La Ferla FM. Alzheimer’s disease. New Eng J Med. 2010;362(4):329–344. 28. Hirtz D, Thurman DJ, Gwinn-Hardy K, Mohammad M, Chaudhari AR, Zalutsky R. How common are the “common” neurologic disorders? Neurology. 2007 Jan 30;68(5):326–327. 29. Isenberg-Grzeda E, Shen MJ, Alici Y, Wills J, Nelson C, Breitbart W. High rate of thiamine deficiency among inpatients with cancer referred for psychiatric consultation: Results of a single site prevalence study. Psychooncology. 2017 Sep;26(9):1384–1389. 30. Gauthier S, Reisberg B, Zaudig M, et al. Mild cognitive impairment. Lancet. 2006 Apr 15;367(9518):1262–1270. 31. Gaudreau JD, Gagnon P, Harel F, Tremblay A, Roy MA. Fast, systematic, and continuous delirium assessment in hospitalized patients: The nursing delirium screening scale. J Pain Symptom Manag. 2005;29(4):368–375. 32. Schuurmans MJ, Shortridge- Baggett LM, Duursma SA. The Delirium Observation Screening Scale: A screening instrument for delirium. Res Theory Nurs Pract. 2003;17(1):31–50. 33. Tariq SH, Tumosa N, Chibnall JT, Perry MH 3rd, Morley JE. Comparison of the Saint Louis University mental status examination and the mini-mental state examination for detecting dementia and mild neurocognitive disorder: A pilot study. Am J Geriatr Psychiat. 2006 Nov;14(11):900–910. CHAPTER 19 Screening for Delirium and Dementia in the Cancer Patient 34. Lycke M, Ketelaars L, Boterberg T, et al. Validation of the Freund Clock Drawing Test as a screening tool to detect cognitive dysfunction in elderly cancer patients undergoing comprehensive geriatric assessment. Psychooncology. 2014 Apr 11. doi:10.1002/ pon.3540 35. Lawlor PG, Nekolaichuk C, Gagnon B, Mancini IL, Pereira JL, Bruera ED. Clinical utility, factor analysis, and further validation of the memorial delirium assessment scale in patients with advanced cancer: Assessing delirium in advanced cancer. Cancer. 2000 Jun 15;88(12):2859–2867. 145 20 Screening and Assessment for Cognitive Problems Alexandra M. Gaynor, James C. Root, Elizabeth Ryan, and Tim A. Ahles Introduction Cognitive dysfunction associated with cancer and cancer treatment has been increasingly recognized and reported by patients. In breast cancer survivors, the most widely investigated cancer cohort, cognitive dysfunction is experienced in 17%–75% of women, at time points between six months and 20 years after completion of treatment.1 A majority of longitudinal studies utilizing objective cognitive assessments have found significant changes from pre-to posttreatment.2 Interestingly, studies have found evidence for cognitive dysfunction prior to adjuvant therapy in 20%–30% of patients diagnosed with breast cancer.2 Due to this research and the increasing awareness of potential cognitive difficulties by patients, objective measures of cognitive functioning that can detect subtle declines in function are necessary. This chapter focuses on the relative utility of brief screening measures and more extensive neuropsychological batteries to detect cancer-associated cognitive dysfunction (CACD) in survivors. Overview of Screening Measures A wealth of screening instruments has been developed for the detection of cognitive difficulties and impairment in general, as well as in a wide range of specific neurological syndromes. Screening measures have the advantage generally of being relatively brief while at the same time sampling from multiple cognitive domains. As a result, these measures have been suggested as a first-line evaluation of potential cognitive impairment preceding or in parallel with comprehensive neuropsychological assessment. A significant drawback to the use of cognitive screening measures in cancer survivors is the limited sensitivity of these measures to mild cognitive dysfunction, which may be more typical in this cohort. Most screening measures were developed for the detection of moderate to severe cognitive impairment, such as that seen in traumatic brain injury and degenerative dementing conditions. In contrast, the mild cognitive difficulties that are sometimes exhibited following cancer treatment will often remain undetected when cognitive screening measures are used. A related drawback to the use of screening measures lies in the fact that results are generally interpreted categorically in reference to a threshold score (i.e., cognitively impaired or intact), rather than on a continuum of ability (i.e., in relation to an individual’s relative position on a normal distribution). As a result, even meaningful decline in a cancer survivor from the high average to average range of ability would be classified as intact when based solely on the results of a cognitive screening measure. A further complication in using cognitive screening measures in the detection of subtle cognitive dysfunction is the likely ceiling effect that will be exhibited across most screening instruments, particularly in previously high- functioning individuals. These issues notwithstanding, cognitive screening measures may be useful in the early stages of evaluation of cognitive difficulties in cancer survivors when interpreted cautiously and when the limitations and relative insensitivity to subtle cognitive decline are kept in mind. The choice of cognitive screening measure should be guided by the expected severity of cognitive decline, as well as the potential etiology of cognitive difficulties. They will be particularly useful in cases in which significant cognitive dysfunction is suspected, as in specific neurological syndromes and dementing disorders in later life. They will be less useful in cases of subtle cognitive dysfunction, as in CACD. To date, no screening measure has been developed and tested specifically for the detection of treatment-associated cognitive difficulties following cancer treatment. As a result, we focus on the sensitivity and specificity of these measures in detecting dementing conditions as well as mild cognitive impairment (MCI), with the caveat that CACD is far less severe than dementia and likely less severe than MCI in the majority of cases. For this review, six common cognitive screening measures are discussed. The Mini- Mental State Exam, the Mini-Cog, and the Blessed Orientation Memory Concentration Test were developed to detect relatively frank and severe dysfunction, while the Montreal Cognitive Assessment, the High Sensitivity Cognitive Screen, and the Repeated Battery for the Assessment of Neuropsychological Status were developed to detect relatively milder or subtler cognitive dysfunction. CHAPTER 20 Screening and Assessment for Cognitive Problems Mini-Mental State Exam Montreal Cognitive Assessment The Mini-Mental State Exam (2nd edition; MMSE-2) is a brief (10– 15 minutes) 30-item measure that assesses orientation to time and place, registration, language function, short-term memory, working memory, and construction.3 Significant improvements have been made with the second edition, including age and education corrections. Traditionally, a cut score of 24, but as high as 26, has been used on the MMSE to suggest a greater probability of cognitive impairment. Sensitivity and specificity vary, depending on age, ethnicity, and education, as well as disease. In a meta-analysis of 34 dementia studies, the MMSE exhibited sensitivity and specificity of 80% and 81%, respectively.4 Significantly, in a sample of highly educated, primarily Caucasian individuals using the original MMSE, a cut score of 26 or below yielded a balance of sensitivity and specificity of 89% and 91%, respectively, indicating that demographic factors can be expected to alter clinical interpretation of MMSE performance.5 In identifying cases of MCI, in contrast either to healthy subjects or to Alzheimer’s dementia (AD), the MMSE has been found to perform poorly.4 Given that treatment-related cognitive effects are expected to be far less severe than in either MCI or frank dementia, the MMSE-2 is unlikely to be of clinical utility in assessing for CACD. Studies that have administered the MMSE in research protocols evaluating the effects of cancer treatment on cognitive function do so to either screen for baseline cognitive impairment for exclusion purposes or as a primary outcome measure. Of these, two studies found a significant effect of group status on MMSE performance,6,7 although results were somewhat limited by small sample size and lack of control group, and further research is needed to determine the ability of the MMSE to detect subtle cognitive deficits in cancer survivors. The Montreal Cognitive Assessment (MoCA) was developed as a brief (10 minutes; 30 items) screening measure for MCI. In a validation study of the MoCA on MCI, AD, and healthy control subjects, the MoCA outperformed the MMSE in discriminating MCI,11 with sensitivity and specificity values for the MoCA of 90% and 87%, respectively, in contrast to MMSE sensitivity and specificity values of 81% and 100%, respectively. Greater sensitivity and specificity of the MoCA over the MMSE has been found in subsequent studies as well,12,13 in addition to better ability to identify domain-specific cognitive dysfunction missed by the MMSE. While the MoCA is sensitive to subtler cognitive dysfunction, it is still likely that treatment-related cognitive effects will be of lesser magnitude in cancer survivors. Arcuri et al.14 used Rasch analyses to examine the psychometric properties of MoCA in cancer survivors and found adequate item fit and measurement of a unidimensional construct but lacked items of sufficiently high difficulty, suggesting it may not be as useful in predicting impairment in survivors with higher cognitive ability. With regard to studies testing MoCA performance in cancer patients, several case studies and one study of 88 male cancer patients showed abnormal MoCA performance during or immediately after treatment, but deficits improved or resolved at one-year follow-up.15,16 The potential influence of practice effects that may better explain cognitive improvement was not addressed. In a large, sample-based study17 that included breast cancer survivors treated with or without chemotherapy and a healthy control group, no difference in MoCA performance was found. Mini-Cog The Mini-Cog8 combines a three-word recall task with the Clock Drawing Test (CDT) and has seen increasing use in preoperative and inpatient evaluations due to its brevity (3 minutes) and sensitivity to moderate and severe dysfunction. In a large, population- based study, the Mini-Cog exhibited sensitivity and specificity values of 76% and 89%, respectively, for detection of dementia, and performed as well as the MMSE (79% and 88%). Aside from the shorter administration time than the MMSE, the Mini-Cog has also been found to be less affected by the education, ethnicity, and language characteristics of the patient.9 In detection of MCI in a sample with dementia and a relatively greater number of individuals classified as “cognitive impairment–no dementia” (CIND), the Mini-Cog performed similarly to previous studies in identifying dementia (sensitivity: 76%; specificity: 73%) but poorly in detecting either dementia or CIND (sensitivity: 39%; specificity: 78%), indicating lesser accuracy in the identification of milder cognitive dysfunction. However, one study examining the CDT in particular found that it had promise in detecting cognitive dysfunction in cancer survivors. Using receiver operating characteristic (ROC) analyses for a cutoff score of ≤4, the CDT showed good diagnostic accuracy (0.88), sensitivity (80.7%), and specificity (81.1%),10 suggesting it might be better suited toward testing cognition in cancer survivors, while the Mini-Cog as a whole may be better suited to detect more severe cognitive dysfunction. Blessed Orientation Memory Concentration Test The Blessed Orientation Memory Concentration (BOMC) test18 is a brief screening measure originally developed to assess for dementia, which has been used in geriatric populations, including older adults with cancer. However, it has not been well validated for use in screening for more MCIs such as those typically seen in CACD. One recent study found that BOMC scores were associated with depressive symptoms, physical function, and limitations in social activities in older adults with cancer prior to treatment, but even statistically significant associations between BOMC and other measures of function were not necessarily clinically significant.19 Therefore, although the BOMC may be useful as a screening measure given its brief administration and association with self-reported function in older adults, further research is needed to establish an appropriate cutoff score for the BOMC that reflects clinically significant cognitive deficits specific to cancer survivors. High Sensitivity Cognitive Screen The High Sensitivity Cognitive Screen (HSCS) is a longer (20–30 minutes) cognitive screening measure that tests executive functioning, visuospatial ability, psychomotor speed, attention and concentration, language, and memory.20 Rather than a discrete cut score, the HSCS classifies individuals into one of five categories: normal, borderline, mild, moderate, and severe. The screen was designed to be sensitive to subtle cognitive dysfunction and has been used in previous studies investigating cognitive effects of cancer treatments. The HSCS has demonstrated strong diagnostic accuracy and agreement with comprehensive neuropsychological assessment with 93% accuracy 147 148 SECTION IV Screening and Assessment in Psychosocial Oncology in noncancer samples. In cancer samples using the HSCS, Brezden et al.21 found a significant difference between breast cancer patients on active chemotherapy treatment and healthy controls, but not when compared with breast cancer survivors with a past history of chemotherapy treatment. Tchen et al.22 administered the HSCS to women on active chemotherapy treatment for breast cancer and healthy controls and found a 16% rate of moderate to severe impairment in the active treatment group compared to only 4% moderate to severe impairment in the healthy control group. A second study by the same group,23 which followed the same patients and controls over one-and two-year time points, found 4.4% and 3.8% of patients with moderate to severe dysfunction as compared to 3.6% and 0% in the healthy control group. Downie et al.24 administered the HSCS to a single group of women diagnosed with breast cancer who were on active chemotherapy treatment and compared objective results to subjective, self-reported cognitive dysfunction. Difficulties with language abilities (61%) and memory (48%) were found using the HSCS, although these rates were lower than would be expected given patient self-report (78% and 95%, respectively); the most significant discrepancy between objective and subjective measures was found in attention abilities (10% objective versus 90% subjective). Vardy et al.25 administered the HSCS over three time points (mean interval = 17 days) to a group predominantly composed of breast cancer survivors (94%) within two years of completion of treatment. While 30% of patients (6/20) exhibited moderate to severe cognitive impairment at baseline, significant putative practice effects reduced this rate to 5% (1/19) at time 2, and this rate remained at time 3 (1/18). The Repeated Battery for the Assessment of Neuropsychological Status The Repeated Battery for the Assessment of Neuropsychological Status (RBANS) is a brief battery addressing immediate memory, visuospatial/ constructional ability, language, attention, and delayed memory. Although originally developed to assess dementia in older adults, the RBANS has been utilized in numerous other clinical populations with cognitive impairment and is appealing given its brevity of administration, high test-retest reliability, and minimal practice effects due to multiple equivalent alternate forms.26 A small number of studies has also suggested it may have promise in detecting CACD in cancer patients and survivors. For example, in a longitudinal study of breast cancer patients, Jansen et al.27 found that scores on multiple RBANS subtests significantly declined during treatment relative to baseline, and all but visuospatial skill deficits returned to baseline performance at 6-month follow-up. The few studies utilizing RBANS as a screening measure in cancer patients and survivors suggest it may be more sensitive than the MMSE and HSCS in detecting subtle cognitive impairment; however, there is evidence that, similar to the MoCA, it is limited in its ability to detect impairment in individuals with higher cognitive function.28 Review of Research: Screening Measures Does screening for cognitive impairment in cancer patients lead to better outcomes? Of the screening instruments used, the MMSE does not appear to provide additional information. Yamada et al.29 used the MMSE with older breast cancer survivors >10 years posttreatment to examine brief global cognitive functioning and found lower MMSE scores in the cancer survivors; however, their scores (mean = 27.6; standard deviation [SD] = 2.1) were still above the MMSE cutoff. Thus, a clinician would not typically refer them for further assessment. Complicating the interpretation of this result is the age of the sample (65 and older). The significant difference between the breast cancer survivors and the controls was 1.7 points on average, which is not likely to result in a clinically meaningful difference. As reviewed earlier, the MoCA has demonstrated diagnostic accuracy similar to the MMSE but exhibited a return to normal performance following treatment in single case studies, and, in a sample of breast cancer survivors, revealed no difference in performance between groups.17 The RBANS appears to be promising as a brief and sensitive screening measure of CACD, and is less influenced by practice effects resulting from repeated testing, but is limited in its ability to detect impairment in patients with higher intellectual functioning.28 The HSCS has been used in several studies22–25 assessing the cognitive effects of cancer treatments. Although the HSCS has demonstrated strong diagnostic accuracy and agreement with comprehensive neuropsychological assessment, studies in cancer survivors suggest inconsistent sensitivity; further limitations include longer administration times (20–30 minutes) and potentially significant practice effects in cases in which serial assessments may be indicated. Due to insensitivity to subtle cognitive dysfunction and the limitations of screening instruments, comprehensive neuropsychological assessment remains the primary means by which to assess CACD. We now turn to describing the comprehensive neuropsychological assessment process and specific recommendations for the assessment of CACD. Overview of Comprehensive Neuropsychological Assessment The neuropsychological assessment begins with a diagnostic interview that determines the patient’s main cognitive complaints, cancer treatments, and medical, neurological, psychiatric, psychosocial, substance use, and educational/vocational history, all of which will help to contextualize the results of the neuropsychological assessment. If a neuropsychological assessment is considered appropriate, formal testing is recommended, which will vary in terms of time and measures administered, depending on patient characteristics and referral question; time for an assessment may range from one to several hours depending on patient-specific factors. A flexible approach to test selection allows the clinician to tailor the assessment to the specific referral question; while most practitioners will employ a somewhat standard “core” battery of measures, additional measures may be administered for specific difficulties or symptoms (multitasking, attention/concentration issues over longer periods of time). Neuropsychological and psychological measures are generally in paper-and-pencil as well as interview and computer- administered format, and should have acceptable validity and reliability. Most batteries, regardless of syndrome, will attempt to sample cognitive ability in multiple domains, including attention and concentration, psychomotor speed, verbal functioning, visuospatial reasoning, praxis and construction, verbal and visual learning and recall, and executive functioning (abstraction, reasoning, cognitive flexibility, problem solving, planning and organization). Depending on the clinician, measures of personality and emotional CHAPTER 20 Screening and Assessment for Cognitive Problems and psychological functioning may also be administered as either a standard part of the assessment or when there is suspicion that emotional distress or a significant psychiatric issue may be affecting cognition. Specifically, for the assessment of CACD, test selection and interpretation of results should be guided by empirical literature and clinical experience. Four early meta-analyses reported significant effects in multiple domains, including in visual and verbal memory, but these analyses included either studie s with multiple cancer types and therapies30 or studies that measured cognitive function during active treatment.30–33 The most recent meta-analysis that included only breast cancer survivors not on active treatment found cross- sectional and longitudinal effects in verbal and visuospatial functioning.34 To develop a harmonized battery of measures for the assessment of CACD, the International Cognition and Cancer Task Force (ICCTF) brought together two working groups with experts in cognition and cancer.1 In selecting tests, the ICCTF required adequate psychometric properties, test- retest reliability, suitability for multinational application, and the availability of alternate test forms (to eliminate practice effects in serial testing). Other criteria considered were the frequent use of the test in a specific area of research being investigated and the use of the test by other cooperative groups. Since previous studies revealed a frontal subcortical pattern of impairment, recommended domains included learning and memory, processing speed, and executive functioning (especially the more complex aspects of attention). Specific measures included the Hopkins Verbal Learning Test-Revised (HVLT-R),35 the Trail Making Test (TMT),36 and the Controlled Oral Word Association (COWA)37 test of the Multilingual Aphasia Examination. A test of working memory (which involves the temporary storage and manipulation of information) was not included in the core battery of recommended tests because none of the current available measures met all of the selective criteria outlined by the ICCTF. However, it is recommended that the core battery be supplemented with a test of working memory based on the neuropsychologist’s preference. We have included a table of measures that we use as part of a flexible battery approach that is guided by the recommendations of the ICCTF, together with previously published research, as well as our own clinical experience within our neuropsychology service (Table 20.1). Following administration of the neuropsychological tests, patient performance on individual measures is compared to normative groups defined by age, or, increasingly, age, education, gender, and ethnicity, to ensure the most exact matching of patients to their respective cohorts. In contrast to a “deficit testing” model of assessment, in which performance is categorized as either normal or aberrant, comparison of patients’ performance to normative groups, and to their own premorbid functioning, allows for finer gradations of interpretation; test results can indicate how well, or how poorly, an individual patient performs on a given task, and also allows for detection not only of absolute deficits but also of deficits that are relative to the patient’s normative cohort or to their own premorbid functioning. Uses of Screening and Assessment What is the best approach to assessing the presence of CACD? Deciding whether to administer a screening instrument or a flexible Table 20.1. List of Commonly Used Neuropsychological Measures in the Assessment of Posttreatment Cognitive Dysfunction Measure Function Premorbid Intelligence Test of Premorbid Functioning (TOPF) A measure estimating premorbid cognitive abilities Verbal Ability Controlled Oral Word Association Test Animal Naming Test Boston Naming Test A timed measure of phonemic fluency A timed measure of semantic fluency A measure of confrontation naming (word finding) Learning and Memory Hopkins Verbal Learning Test-R Logical Memory I and II (WMS-IV) Brief Visuospatial Memory Test-R A measure of verbal list learning and recall A measure of verbal story learning and recall A measure of visual figure learning and memory Attention Digit Span (WAIS-IV) Arithmetic (WAIS-IV) Continuous Performance Test (CPT) A measure of brief span of attention A measure of brief span of attention and working memory A measure of sustained attention Processing Speed The Trail Making Test (Part A) The Trail Making Test (Part B) Digit Symbol—Coding (WAIS-IV) Symbol Search (WAIS-IV) A timed measure of visual scanning and motor speed A timed measure of visual scanning, motor speed, and set-shifting A timed measure of visual associative learning A timed measure of visual scanning and attention Visual Reasoning/Construction Rey-Osterreith Complex Figure Judgment of Line Orientation Block Design (WAIS-IV) A measure of visual construction A measure of visuospatial judgment and reasoning A timed measure of visual construction and reasoning Executive Functioning Wisconsin Card Sorting Task (WCST) A measure of abstract reasoning and Stroop Task problem-solving A measure of word reading, color naming, and inhibition Psychological/Emotional Personality Assessment Inventory (PAI) Beck Depression Inventory (BDI) Generalized Anxiety Disorder Scale (GAD-7) A self-report measure of psychological functioning A self-report measure of depressive symptomatology A self-report measure of anxiety symptomatology neuropsychological assessment depends on the time allotted, the patient’s condition (i.e., fatigue), and the nature of the patient’s symptoms (i.e., does his or her presentation or the caregiver’s report resemble an incipient dementia, or are the symptoms more subtle?). The limitations of screening instruments have been thoroughly reviewed. Given these limitations, it has been suggested that a combination of screening measures be used to ensure detection of subtle impairment in cancer survivors.22,28 However, a flexible neuropsychological assessment can be tailored to the patient’s individual reported difficulties and can be shortened or given over multiple 149 150 SECTION IV Screening and Assessment in Psychosocial Oncology appointments (in the case of a fatigued patient). Typically, this process is initiated once a patient reports concerns about their cognitive function to their oncologist, nurse practitioner, or other provider managing their care, at which point the provider should refer the patient to a neuropsychologist for a full assessment of neurocognitive function. Therefore, the use of patient-reported outcome measures, such as the Functional Assessment of Cancer Therapy-Cognitive Function (FACT-Cog),38 the PROMIS Cognitive Function,39 or the measurement of Everyday Cognition (ECog),40 may be useful in conjunction with screening measures, as high subjective complaints of functioning in combination with suspected CACD based on screening tests may warrant further testing using a thorough neuropsychological battery. We advocate a flexible, comprehensive neuropsychological assessment for patients with CACD because included measures exhibit higher sensitivity and specificity and typically have better normative data, which will ultimately yield superior information regarding the presence or absence of CACD. For a suspected dementia, depending on the complaints and the level of impairment (cognitive and functional), a screening instrument, sparing the patient’s time and reducing the distress associated with poor performance, is recommended over a more extensive battery. For instance, the brief Geriatric Assessment (GA),41 which includes a cognitive screening component and has been validated in clinical cancer trials and community oncology clinics, may be an appropriate screening instrument for suspected dementia in older adults with cancer. Future Directions CACD has been established as a lingering problem for some non- CNS cancer survivors over the past two decades through multiple research studies with increasing methodological scientific rigor. Strong interest remains in identifying screening instruments with sufficient sensitivity and specificity to identify patients who present with suspected CACD. We recommend that one focus of future research should be on the development of more accurate screening measures for CACD, and that patient-reported outcomes be used to determine whether further screening and assessment is warranted using objective measures of function. REFERENCES 1. Wefel JS, Vardy J, Ahles T, Schagen SB. International Cognition and Cancer Task Force recommendations to harmonise studies of cognitive function in patients with cancer. Lancet Oncol. 2011;12(7):703– 708. doi:10.1016/S1470-2045(10)70294-1 2. Ahles TA, Root JC. Cognitive effects of cancer and cancer treatments. Annu Rev Clin Psychol. 2018;14(1):425–451. doi:10.1146/ annurev-clinpsy-050817-084903 3. Folstein MF, Folstein SE, McHugh PR. Mini-Mental State. J Psychiatr Res. 1975;12(3):189–198. doi:10.1016/0022-3956(75)90026-6 4. Mitchell AJ. A meta-analysis of the accuracy of the Mini-Mental State Examination in the detection of dementia and mild cognitive impairment. J Psychiatr Res. 2009;43(4):411–431. doi:10.1016/ j.jpsychires.2008.04.014 5. O’Bryant SE, Humphreys JD, Smith GE, et al. Detecting dementia with the Mini-Mental State Examination in highly educated individuals. Arch Neurol. 2008;65(7):963–967. doi:10.1001/ archneur.65.7.963 6. Yamada TH, Denburg NL, Beglinger LJ, Schultz SK. Neuropsychological outcomes of older breast cancer survivors: cognitive features ten or more years after chemotherapy. J Neuropsychiatry Clin Neurosci. 2010;22(1):48–54. doi:10.1176/ jnp.2010.22.1.48 7. Hoppe S, Rainfray M, Fonck M, et al. Functional decline in older patients with cancer receiving first-line chemotherapy. J Clin Oncol. 2013;31(31):3877–3882. doi:10.1200/JCO.2012.47.7430 8. Borson S, Scanlan J, Brush M, Vitaliano P, Dokmak A. The Mini- Cog: a cognitive “vital signs” measure for dementia screening in multi- lingual elderly. Int J Geriatr Psychiatry. 2000;15(11):1021–1027. 9. Borson S, Scanlan JM, Watanabe J, Tu S-P, Lessig M. Simplifying detection of cognitive impairment: comparison of the Mini-Cog and Mini-Mental State Examination in a multiethnic sample: MINI- COG and MMSE. J Am Geriatr Soc. 2005;53(5):871–874. doi:10.1111/j.1532-5415.2005.53269.x 10. Ketelaars L, Pottel L, Lycke M, et al. Use of the Freund Clock Drawing Test within the Mini-Cog as a screening tool for cognitive impairment in elderly patients with or without cancer. J Geriatr Oncol. 2013;4(2):174–182. doi:10.1016/j.jgo.2012.10.175 11. Nasreddine ZS, Phillips NA, Bedirian V, et al. The Montreal Cognitive Assessment, MoCA: a brief screening tool for mild cognitive impairment. J Am Geriatr Soc. 2005;53(4):695–699. doi:10.1111/j.1532-5415.2005.53221.x 12. Dong Y, Lee WY, Basri NA, et al. The Montreal Cognitive Assessment is superior to the Mini–Mental State Examination in detecting patients at higher risk of dementia. Int Psychogeriatr. 2012;24(11):1749–1755. doi:10.1017/S1041610212001068 13. Markwick A, Zamboni G, de Jager CA. Profiles of cognitive subtest impairment in the Montreal Cognitive Assessment (MoCA) in a research cohort with normal Mini- Mental State Examination (MMSE) scores. J Clin Exp Neuropsychol. 2012;34(7):750–757. doi:10.1080/13803395.2012.672966 14. Arcuri GG, Palladini L, Dumas G, Lemoignan J, Gagnon B. Exploring the measurement properties of the Montreal Cognitive Assessment in a population of people with cancer. Support Care Cancer Off J Multinatl Assoc Support Care Cancer. 2015;23(9):2779–2787. doi:10.1007/s00520-015-2643-7 15. Mandilaras V, Wan-Chow-Wah D, Monette J, Gaba F, Monette M, Alfonso L. The impact of cancer therapy on cognition in the elderly. Front Pharmacol. 2013;4. doi:10.3389/fphar.2013.00048 16. Regier NG, Naik AD, Mulligan EA, et al. Cancer-related cognitive impairment and associated factors in a sample of older male oral-digestive cancer survivors. Psychooncology. 2019;28(7):1551– 1558. doi:10.1002/pon.5131 17. Chen X, Zhu C, Li J, et al. Dissociation of decision making under ambiguity and decision making under risk in breast cancer patients receiving adjuvant chemotherapy: a neuropsychological study. Brain Res. 2013;1533:63–72. doi:10.1016/j.brainres.2013.08.015 18. Katzman R, Brown T, Fuld P, Peck A, Schechter R, Schimmel H. Validation of a short Orientation-Memory-Concentration Test of cognitive impairment. Am J Psychiatry. 1983;140(6):734–739. doi:10.1176/ajp.140.6.734 19. Nakamura ZM, Deal AM, Nyrop KA, Choi SK, Wood WA, Muss HB. Associations of functional, psychosocial, medical, and socio- demographic factors with cognitive screening in chemotherapy naïve patients with breast cancer. Psychooncology. 2019;28(1):167– 173. doi:10.1002/pon.4928 20. Fogel BS. The high sensitivity cognitive screen. Int Psychogeriatr. 1991;3(2):273–288. CHAPTER 20 Screening and Assessment for Cognitive Problems 21. Brezden CB, Phillips K-A, Abdolell M, Bunston T, Tannock IF. Cognitive function in breast cancer patients receiving adjuvant chemotherapy. J Clin Oncol. 2000;18(14):2695–2701. doi:10.1200/ JCO.2000.18.14.2695 22. Tchen N, Juffs HG, Downie FP, et al. Cognitive function, fatigue, and menopausal symptoms in women receiving adjuvant chemotherapy for breast cancer. J Clin Oncol. 2003;21(22):4175–4183. doi:10.1200/JCO.2003.01.119 23. Fan HGM, Houédé-Tchen N, Yi Q-L, et al. Fatigue, menopausal symptoms, and cognitive function in women after adjuvant chemotherapy for breast cancer: 1-and 2-year follow-up of a prospective controlled study. J Clin Oncol. 2005;23(31):8025–8032. doi:10.1200/JCO.2005.01.6550 24. Downie FP, Mar Fan HG, Houédé-Tchen N, Yi Q, Tannock IF. Cognitive function, fatigue, and menopausal symptoms in breast cancer patients receiving adjuvant chemotherapy: evaluation with patient interview after formal assessment. Psychooncology. 2006;15(10):921–930. doi:10.1002/pon.1035 25. Vardy J, Wong K, Yi Q, et al. Assessing cognitive function in cancer patients. Support Care Cancer. 2006;14(11):1111–1118. doi:10.1007/s00520-006-0037-6 26. Loughan AR, Braun SE, Lanoye A. Repeatable Battery for the Assessment of Neuropsychological Status (RBANS): preliminary utility in adult neuro-oncology. Neuro-Oncol Pract. 2019;6(4):289– 296. doi:10.1093/nop/npy050 27. Jansen CE, Cooper BA, Dodd MJ, Miaskowski CA. A prospective longitudinal study of chemotherapy-induced cognitive changes in breast cancer patients. Support Care Cancer. 2011;19(10):1647– 1656. doi:10.1007/s00520-010-0997-4 28. Cheung YT, Tan EH-J, Chan A. An evaluation on the neuropsychological tests used in the assessment of postchemotherapy cognitive changes in breast cancer survivors. Support Care Cancer. 2012;20(7):1361–1375. doi:10.1007/s00520-012-1445-4 29. Yamada TH, Denburg NL, Beglinger LJ, Schultz SK. Neruopsychological outcomes of older breast cancer survivors: cognitive features ten or more years after chemotherapy. J Neuropsychiatry Clin Neurosci. 2010;22:48–54. 30. Anderson- Hanley C, Sherman ML, Riggs R, Agocha VB, Compas BE. Neuropsychological effects of treatments for adults with cancer: a meta-analysis and review of the literature. J Int Neuropsychol Soc JINS. 2003;9(7):967– 982. doi:10.1017/ S1355617703970019 31. Falleti MG, Sanfilippo A, Maruff P, Weih L, Phillips K-A. The nature and severity of cognitive impairment associated with adjuvant chemotherapy in women with breast cancer: a meta-analysis of the current literature. Brain Cogn. 2005;59(1):60–70. doi:10.1016/ j.bandc.2005.05.001 32. Jansen CE, Miaskowski C, Dodd M, Dowling G, Kramer J. A metaanalysis of studies of the effects of cancer chemotherapy on various domains of cognitive function. Cancer. 2005;104(10):2222–2233. doi:10.1002/cncr.21469 33. Stewart A, Bielajew C, Collins B, Parkinson M, Tomiak E. A meta-analysis of the neuropsychological effects of adjuvant chemotherapy treatment in women treated for breast cancer. Clin Neuropsychol. 2006;20(1):76–89. doi:10.1080/138540491005875 34. Jim SL, Phillips KM, Chait S. Meta-analysis of cognitive functioning in breast cancer survivors previously treated with standard-dose chemotherapy. J Clin Oncol. 2012;30:3578–3587. 35. Shapiro AM, Benedict RH, Schretlen D, Brandt J. Construct and concurrent validity of the Hopkins Verbal Learning Test-revised. Clin Neuropsychol. 1999;13(3):348–358. doi:10.1076/clin.13.3.348.1749 36. Reitan RM. Trail Making Test: Manual for Administration and Scoring. Tucson, AZ: Reitan Neuropsychological Laboratory; 1971. 37. Heaton RK, Miller SW, Taylor MJ, Grant I. Revised Comprehensive Norms for an Expanded Halstead-Reitan Battery: Demographically Adjusted Neuropsychological Norms for African American and Caucasian Adults. Lutz, FL: Psychological Assessment Resources; 2004. 38. Wagner LI, Sweet JJ, Butt ZA, Lai JS, Cella D. Measuring patient self-reported cognitive function: development of the functional assessment of cancer therapy-cognitive function instrument. J Support Oncol. 2009;7(6):32–39. 39. Cella D, Riley W, Stone A, et al. The Patient-Reported Outcomes Measurement Information System (PROMIS) developed and tested its first wave of adult self-reported health outcome item banks: 2005–2008. J Clin Epidemiol. 2010;63(11):1179–1194. doi:10.1016/j.jclinepi.2010.04.011 40. Farias ST, Mungas D, Reed BR, et al. The measurement of Everyday Cognition (ECog): scale development and psychometric properties. Neuropsychology. 2008;22(4):531– 544. doi:10.1037/ 0894-4105.22.4.531 41. Hurria A, Gupta S, Zauderer M, et al. Developing a cancer- specific geriatric assessment. Cancer. 2005;104(9):1998–2005. doi:10.1002/cncr.21422 151 SECTION V Psychological Issues Related to Site of Cancer Mark Lazenby (Section Editor) 21 Melanoma 155 Nadine A. Kasparian and Iris Bartula 27 Gynecologic Cancers 196 Heidi S. Donovan and Teresa H. Thomas 22 Lung Cancer 162 Marianne Davies 28 Hematopoietic Dyscrasias and Stem Cell Transplantation/CAR-T Cell Therapy 203 Jesse R. Fann and Nicole Bates 23 Breast Cancer 169 M. Tish Knobf and Youri Hwang 24 Colorectal Cancer 176 Anne Miles and Claudia Redeker 25 Prostate Cancer and Genitourinary Malignancies 182 Andrew J. Roth and Alejandro Gonzalez-Restrepo 26 Gastrointestinal Cancers 189 Daniel C. McFarland and William S. Breitbart 29 Head and Neck Cancer 215 Loreto Fernández González, Jonathan Irish, and Gary Rodin 30 Central Nervous System Tumors 221 Alan D. Valentine 31 HIV Infection and AIDS-Associated Neoplasms 226 Joanna S. Dognin and Peter A. Selwyn 21 Melanoma Nadine A. Kasparian and Iris Bartula Introduction Comprehensive psychological care of people with melanoma spans all phases of the disease trajectory, including risk assessment, diagnosis, treatment, recovery, and survivorship, as well as the transition from curative to palliative care, death, and bereavement. Each person will experience a range of physical, practical, and psychosocial challenges unique to their circumstances. A diagnosis of melanoma has the potential to change many aspects of an individual’s life from self-identity, body image, self-esteem, and perceived well- being to family roles and relationships, career opportunities, friendships, and finances. These changes are likely to manifest differently depending on a range of sociodemographic, disease, and psychological factors. In cancer and other chronic illnesses, high levels of psychological distress can be associated with reduced engagement in cancer screening, treatment and risk management recommendations, delays in seeking medical advice, lower satisfaction with clinical care, higher health care costs, poorer social functioning, greater morbidity and mortality, and poorer overall quality of life (QOL).1 Identifying patients most likely to experience psychological distress and impaired QOL and facilitating timely engagement with effective interventions to ameliorate these difficulties remain important clinical and research endeavors. The objective of this chapter is to provide all members of the diagnostic and treatment team (dermatologists, oncologists, surgeons, general practitioners, nurses, psychologists, social workers, psychiatrists, pathologists, clinical geneticists, genetic counselors, physiotherapists, occupational therapists), patients, and families, as well as those in training and research, with an evidence-based overview of the psychosocial aspects relevant to people affected by melanoma. Melanoma Incidence Melanoma is a cancer of melanin-producing cells that most frequently occurs in the skin, but occasionally may present in the mouth, intestines, or eyes. Worldwide, melanoma accounts for 3% of all skin cancers and represents 4.5% of all new cancer diagnoses.2 Incidence is increasing, especially in fair-skinned populations, with Australia and New Zealand reporting the highest rates of melanoma in the world.2,3 Increased incidence is commonly attributed to environmental factors, such as ultraviolet radiation (UVR) exposure, as well as increased screening and early detection practices.3 While presentations vary, a “typical” patient is most likely to be male, aged 55–64 years, and fair-skinned, and to present with a lesion on his torso.2 Melanoma Risk Factors and Prevention Strategies Awareness of melanoma risk factors can help target screening and population-based prevention strategies (Table 21.1).2,3 Primary prevention strategies aim to reduce UVR exposure, while secondary prevention strategies aim to improve early disease detection (Table 21.2). Level of implementation and success of prevention strategies vary across countries. Effects of primary prevention strategies have been limited in the United Kingdom, the United States, Norway, and Sweden, while Australia has achieved a demonstrably slowing incidence of melanoma following broad implementation.3 National screening policies also vary; Germany, for example, recommends routine skin cancer screening for all adults aged over 35 years, while the United States does not have a national screening program. Clinical guidelines are consistent in recommending regular surveillance for people at high risk of melanoma.4 Melanoma Staging Staging of melanoma includes consideration of tumor characteristics, presence of lymph node involvement, and distant metastases, factors associated with survival. Evidence-based guidelines outline five stages: • Stage 0 (melanoma in situ)—tumor is confined to the outer layers of the skin (epidermis), with no evidence of spreading. • Stages I and II—no evidence of lymph node involvement or distant metastases. Stages I and II are distinguished by lesion depth (Breslow thickness) and presence of ulceration (invasion of surrounding tissue). • Stage III—evidence of regional lymph node involvement. 156 SECTION V Psychological Issues Related to Site of Cancer Table 21.1. Melanoma Risk Factors * Risk Factor Features That Increase Risk Ultraviolet radiation (UVR) exposure • • • • Skin, hair, and eye pigmentation • Fairer pigmentation. • Tendency for skin to freckle or burn in sun. Nevi* number and characteristics • Presence of more than 100 nevi. • More than 2 dysplastic (atypical) nevi. Gene mutations (implicated in 5–12% of all melanomas) • Strong family history of melanoma (≥3 first-degree relatives with melanoma) • One or more first-degree relatives with a diagnosis of pancreatic cancer. • Personal history of multiple melanomas. • Mutations in tumor suppressor gene CDKN2A. Intermittent, short, intense UVR exposure increases cutaneous melanoma risk. Chronic, cumulative UVR exposure increases lentigo maligna melanoma risk. Childhood sun exposure. UVR exposure resulting from indoor tanning. Collection of benign melanin-producing cells. • Stage IV—evidence of distant metastases to skin, soft tissue, muscles, nonregional lymph nodes, lungs, abdomen, bones, or the brain. Melanoma Survival and Mortality Among skin cancers, melanoma has the highest mortality. Globally, mortality rates steadily increased in the 1980s, peaked in 1988– 1990, then maintained a slow increase.3 Females and younger patients tend to have better survival.3,4 Five-year survival for patients with early, localized melanoma is excellent, ranging from 82% (Stage II) to 99% (Stage I). Five-year survival rates for Stage III patients range from 32% to 93%, depending on tumor characteristics and number of lymph nodes involved. Survival for Stage IV patients is low (15–20%) but improving with rapid advances in new targeted and immunotherapies.4 Melanoma Treatments Stage I–III melanoma is typically treated with wide local excision, to prevent local recurrence at the margin or near the initial lesion. Some patients may require sentinel lymph node biopsy to investigate the presence of micro-metastases. Surgery may be offered to Stage IV patients with the goal of palliation (symptom management), with some evidence of improved survival.5 Adjuvant systemic therapy is used to prevent disease recurrence or progression (spread to other organs).4 Targeted and immunotherapies have been shown to improve survival of Stage IV patients.4,5 Chemotherapy may be offered to Stage IV patients for symptom control. Radiation therapy may be offered to patients at increased risk of recurrence to achieve better local and regional control, and to patients with metastatic melanoma to reduce pain and neurocognitive symptoms associated with brain metastases. It is usually used in combination with other systemic treatements.5 Quality of Life QOL is a key consideration, particularly as novel medical therapies increase the likelihood of longer-term survival for people with advanced disease. Early-stage melanoma patients appear to have QOL similar to that of the general population.6 QOL impairments are most common immediately postdiagnosis and gradually improve over time. Poorer physical health and greater comorbidities are the most consistent correlates of poorer QOL, while patient age, sex, and social support are inconsistently associated with QOL.6 Patients with advanced melanoma participating in clinical trials of targeted and immunotherapies may report improved QOL; however, evaluation of the relationship between adverse treatment effects (including new concomitant toxicities), difficulties with ongoing adherence to oral therapies, and QOL, without excluding patients who discontinue therapy, is needed.7 Routine QOL assessment is strongly recommended and measures can be broadly grouped into three categories: (1) melanoma specific, (2) cancer specific, and (3) generic measures, with some studies suggesting generic measures may not be sensitive to functional changes associated with melanoma,6 while others provide conflicting evidence.8 Table 21.2. Melanoma Prevention Strategies Primary Prevention Strategies Secondary Prevention Strategies • Sun protection policies (e.g., mandatory sun protection factor labeling • Screening using visual skin examination. This can be achieved by inspection of the on sunscreens). • Population-based education on, and encouragement of, sun-safe behaviors (e.g., use of broad-spectrum sunscreen; protective clothing, hats, and sunglasses; avoidance of outdoors when ultraviolet radiation (UVR) is above 3). entire skin surface, conducted by either - nonphysician, usually the patients themselves (skin self-examination [SSE]) or a partner (partner skin examination [PSE]), or - medical professional, usually a dermatologist (clinical skin examination [CSE],), which may include the use of total body photography, dermoscopy, and/or confocal microscopy. CHAPTER 21 Melanoma Physical Functioning Surgery may result in pain, numbness, and lymphedema (localized swelling of the body that may result from lymph node dissection). Physical concerns tend to be most prevalent immediately following surgery, with most symptoms improving over time or with physical therapy, while other difficulties (e.g., lymphedema) may require ongoing management.9 Fatigue is common following systemic and radiation therapy, or in the context of vitamin D deficiency that may arise due to restricted sun exposure10 with consequences for QOL, functional capacity, and well-being. Patient age, additional chronic health conditions, higher self-blame and fear of cancer recurrence, and lower social support are associated with greater fatigue. The Clinical Oncology Society of Australia has recommended exercise as a safe and effective strategy for managing cancer-related fatigue, as well as providing other physical and mental health benefits; however, implementation may be difficult, as patients often perceive fatigue as a barrier to regular exercise, despite perceiving exercise as helpful.10 Body Image Concerns Change in body image resulting from surgical scars or lymphedema can alter self- perceptions and impact intimate relationships.9,11 Body image concerns often arise when there is a discrepancy between anticipated and actual scar appearance. People with melanoma frequently report feeling inadequately prepared for the changes resulting from surgery and perceiving a mismatch between surgeons’ and their own perceptions of a healing scar. Women tend to be at greater risk of body image disturbance than men, and body image concerns are associated with greater anxiety and depression, as well as lower QOL.9,11 Use of clothing to cover scars and lymphedema, cognitive strategies (e.g., perspective-taking), and social support have been identified by melanoma patients as helpful in coping with body image concerns,9,11 but evidence-based psychological interventions to support patients with body image disturbance are lacking. Social Functioning While many patients report relying mostly on family and friends for support, some describe concerns sharing information about their diagnosis and treatment, for fear of the emotional impact on family members. This may leave some patients feeling isolated and preferring support from other melanoma patients with similar concerns and experiences.9 Overall, social support tends to predict adjustment to melanoma diagnosis and treatment, with advanced cancer patients reporting the greatest difficulties in social functioning.11 Melanoma diagnosis may also lead to altered social activities to reduce sun exposure, as well as attempts to educate family and friends about melanoma and its risk factors. Some patients report substantial financial stress, especially if they are unable to work due to treatment or illness. Patients have also expressed concerns the community may not perceive melanoma as a serious health condition, given most melanomas are detected early and are associated with good prognosis. This may minimize the validity of patients’ needs and concerns and impede timely access to support.9 Psychological Functioning Many patients experience the period of diagnostic uncertainty between detection of a suspicious lesion and receipt of skin biopsy results as highly stressful.12 Following diagnosis, patients often experience shock and intense fear, sadness, and/or anger. Anxiety is frequently reported by patients in the days and weeks before follow-up appointments and may manifest both physically and psychologically, including symptoms such as diarrhea, nausea, and sleeplessness. As some patients enter palliative care, anxiety about the end of life, symptom management, and existential issues are commonly reported. The distress evoked by these transitions may not only cause immediate suffering but also influence a range of patient behaviors;9,11 thus, understanding the range of emotional, behavioral, and physical responses to melanoma (Table 21.3), as well as effective and sustainable interventions to reduce distress, remains an important clinical and research endeavor. Mental Health At least 30% of people with melanoma report levels of psychological distress, particularly anxiety, indicative of a need for clinical intervention.12 While depression and anxiety commonly coexist, anxiety is uniquely characterized by symptoms of pervasive and uncontrollable worry, whereas depression is typified by prolonged low mood, increased irritability, social withdrawal, and diminished interest or pleasure in activities. Reported prevalence of psychological distress varies across studies, potentially reflecting sample differences in melanoma type and stage, time since diagnosis, treatment type, timing and method of data collection, clinical trial involvement, environment (e.g., areas of high versus low UVR exposure), and cultural differences. Anxiety is reported by 20–32% of patients, while 15–19% report symptoms indicative of depression.11,12 Melanoma Table 21.3. Common Emotional, Behavioral, and Physical Responses to the Diagnosis of Skin Cancer Emotional Responses Behavioral Responses Physical Responses • • • • • • • • • • • • Social withdrawal or an increased need to be with others • Hypervigilance with sun protection and early skin cancer detection, or • • • • • • • Shock Feeling “numb” Disbelief Fear and worry Anxiety Anger Confusion Uncertainty Sadness or distress Depression Grief disengagement from screening and health behavior recommendations • Delays in seeking medical care • Altered interest in and disengagement from pleasurable activities • Substance misuse Sleep disturbance Appetite changes Headaches Heart palpitations Nausea Changes in bowel movements Difficulty concentrating 157 158 SECTION V Psychological Issues Related to Site of Cancer patients report the third highest four-week prevalence of any psychological disorder (39%), behind only breast (42%) and head and neck (41%) cancer patients.13 Fear of Cancer Recurrence or Progression Extensive evidence shows people who have had melanoma experience marked and enduring fear of cancer recurrence (FCR). FCR is typically defined as fear, worry, or concern relating to the possibility that cancer will come back or progress. FCR is generally greatest in the days and weeks prior to medical consultations12 and may manifest as (1) preoccupation, worry, rumination, or intrusive thoughts about cancer; (2) maladaptive coping; (3) impairments in daily functioning; (4) intense distress; and (5) difficulties thinking about the future. While some degree of fear is a normal response to cancer, high levels are associated with anxiety and depression, reduced participation in cancer screening, delays in seeking medical care, lower satisfaction with clinical care, higher health care costs, poorer social functioning, greater morbidity and mortality, and poorer quality of life.1 An Australian study found 72% of early-stage melanoma patients reported levels of FCR indicative of a need for clinical intervention at least two years after initial diagnosis, irrespective of actual recurrence risk.14 The uncertainty that drives cancer-related fear and anxiety may be even greater for those affected by advanced disease, where the longer-term outcomes of fast-changing medical treatments are not fully known. People with melanoma want help with FCR; assistance with “fears about the cancer spreading” is the most frequently endorsed, and most distressing, unmet need reported by melanoma patients soon after diagnosis and years later. A population-based cohort study of 2,615 cancer survivors (n = 469 melanoma) found that satisfaction with information provision was associated with lower FCR,15 suggesting that clear information about recurrence risk and management strategies may at least partially alleviate FCR. Patients with high FCR are likely to benefit from interventions featuring both psychoeducation and psychotherapy.16,17 Familial Melanoma and Psychological Distress Kasparian et al.18 examined psychological responses reported by Australians who were informed of the identification of a family- specific mutation in the CDKN2A gene, which, in Australia, is associated with a lifetime melanoma risk of 91%. Following notification, levels of anxiety and depressive symptoms in this cohort were comparable to population norms.18 In a prospective cohort study examining outcomes of genetic testing for melanoma risk, Kasparian et al.19 found that compared to baseline, individuals identified as carriers of a pathogenic CDKN2A mutation reported significantly reduced anxiety at 2 weeks, and reduced depression at 2 weeks and 12 months, following receipt of genetic test results. Carriers also reported a greater frequency of clinical skin examination at 12- month follow-up, providing evidence of healthy psychological and behavioral adjustment following participation in genetic testing for melanoma risk. Factors That Increase Vulnerability to Psychological Distress A variety of factors may increase vulnerability to psychological distress in people with melanoma (Table 21.4). Beliefs about melanoma and its treatment and prognosis may play a greater role in determining stress responses than disease factors.20 Decline in physical functioning is also a key correlate; compared with patients experiencing low distress, those who report high distress also report worse evaluations of current and future health, greater pain, lower energy, and greater interference in daily functioning.21 Coping and Resilience Psychological outcomes associated with melanoma are influenced, at least in part, by a person’s coping responses, which can be defined as beliefs and behaviors with an adaptive purpose during times of threat or adversity.2 Patients who adopt active coping strategies demonstrate better adjustment to melanoma and longer relapse-free periods compared to those who adopt passive or avoidant strategies (Table 21.5).12 While some patients, particularly those with advanced melanoma, may feel overwhelmed by the physical and psychological consequences of their disease, the multidisciplinary health care team can support coping through open, patient-centered communication and empathic acknowledgment of patients’ needs and concerns, and by adopting a holistic approach to QOL.11 Supportive Care Needs A large, multicenter study carried out in Australia found that over 90% of patients with early-stage melanoma reported at least one unmet supportive care need following diagnosis.22 Of the 10 most highly ranked unmet needs, three were psychological (FCR, uncertainty about the future, concerns or worries about others) and seven were informational, including an unmet need for greater information on melanoma recurrence and progression, treatment options, and preventive behaviors.22 Unmet supportive care needs are also commonly reported by advanced melanoma patients, with about Table 21.4. Demographic, Clinical, and Psychosocial Factors Associated with Increased Vulnerability to Emotional Distress in People with Melanoma Demographic Factors Clinical Factors Psychosocial Factors • • • • • • • • • • • • • • • • • Female gender Younger age Lower educational attainment Unemployment Financial hardship Greater physical deterioration Lower physical quality of life Greater functional impairments Diagnosis of advanced melanoma Greater treatment toxicity effects Fatigue Limited social and instrumental (practical) support Absence of a spouse or a committed partner Negative beliefs about melanoma Greater fear of cancer recurrence or progression Passive or avoidant coping styles Concerns about the implications of melanoma for one’s family • History of mental illness CHAPTER 21 Melanoma Table 21.5. Common Coping Styles Coping Style Brief Description Examples Active-behavioral coping Overt behavioral attempts to deal directly with melanoma and its effects • • • • • Active- cognitive coping Attitudes, beliefs, thoughts, and perceptions about melanoma • • • • Avoidance Attempts to actively avoid the problem or indirectly reduce emotional tension through distraction • Denial of the diagnosis or the need for treatment • Disengagement from treatment or screening regimes • Avoidance of any reminders that are associated with melanoma (e.g., not driving Enlisting the help and support of others Seeking and accepting professional and/or peer support Adhering to treatment and screening protocols Seeking relevant information Adopting a healthier lifestyle (e.g., improving diet, exercise, sleep, work-life balance) • Use of complementary therapies in addition to recommended medical treatment Accepting the reality of one’s illness Acceptance of one’s emotional responses Forming realistic beliefs and expectations Creating time and space to think about and make sense of one’s experiences and the cancer-related information provided past the skin cancer clinic) • Overuse of distraction at the cost of quality of life • Compulsive internet searches about diagnosis and treatment, searching for examples of positive outcomes • Substance use or abuse half of patients with Stage III/IV melanoma reporting concerns regarding their understanding of treatment options and potential side effects, as well as unmet needs related to coping with fear of cancer progression, living with uncertainty, anxiety, depression, and concern about loved ones.11 Mental Health Care for People with Melanoma Clinical practice guidelines consistently recommend structured psychological interventions be made available to all melanoma patients. Numerous psychoeducational interventions comprising various combinations of education (e.g., on risk of recurrence), behavioral or skills training (e.g., skin self-examination), and/or psychotherapy (e.g., working through emotional responses to cancer) have been developed to meet this need.23 Interventions vary in intensity (e.g., single versus multiple sessions), mode of delivery (e.g., face to face, telehealth, digital or web based), delivery agents (e.g., psychologists, nurses, social workers, general practitioners, dermatologists, peers), and therapeutic modalities used (e.g., cognitive behavioral therapy, brief psychodynamic psychotherapy, structured psychoeducation)—but all share a common goal to assist people with melanoma to adjust physically and psychologically to the disease and its treatment. Educational interventions appear generally effective in improving melanoma knowledge and skin self-examination efficacy and engagement, while psychotherapeutic interventions have demonstrated efficacy in reducing fear of cancer recurrence, psychological distress, depression, and anxiety.23 Dieng et al., for example, examined the effects of a brief psychological intervention targeting FCR in patients at high risk of developing another primary melanoma. The intervention consisted of a psychoeducational resource25coupled with three telehealth-based psychotherapy sessions individualized to address each patient’s support needs and preferences.16,17 Participants were randomized to receive the intervention (n = 80) or usual care (n = 84), with psychotherapy sessions timed in relation to dermatological appointments. Significant improvements in FCR severity, psychological stress, and melanoma knowledge were found for intervention group participants immediately posttreatment and at six months postintervention,16 and improvements in FCR persisted to at least 12 months postintervention.17 Cost-Effectiveness of Psychological Interventions As evidence for the efficacy of psychological interventions grows, studies evaluating the fiscal costs of mental health care are also needed to guide implementation and allocation of limited health resources. In Australia, Dieng et al.25 prospectively evaluated the cost- effectiveness of a brief psychological intervention to reduce FCR in early-stage melanoma patients. The mean cost of the intervention was AU$1,614 per participant (including intervention development costs), and the cost-effectiveness acceptability curve demonstrated a 78% probability of the intervention being cost-effective relative to the control (usual care) at a threshold of AU$50,000 per extra person avoiding FCR. From the perspective of the Australian health system, this represents good value for money. Current Care and Future Directions A stepped care approach, where people with melanoma are offered increasingly comprehensive information and mental health care as their level of need increases, may ensure optimal health outcomes. As a first step, all patients presenting at a pigmented lesion, oncology, or dermatology clinic should be provided with detailed information regarding their condition and emotional support from their primary health care provider, to address patient needs at this highly stressful time. Box 21.1 provides strategies to use in consultations to facilitate patient understanding of information and psychological adjustment. 159 160 SECTION V Psychological Issues Related to Site of Cancer Box 21.1. Interactional Skills and Communication Strategies to Support Mental Health and Reduce Unmet Needs in People with Melanoma Interactional Skills and Communication Strategies • Open and empathic discussion of the diagnosis, treatment, prognosis, and life expectancy, as well as how the cancer might affect other aspects of the patient’s life • Face-to-face communication in a quiet and private environment • Use of verbal and nonverbal cues to show empathy and emotional support • Initiating communication about emotional well-being • Exploration of the patient’s thoughts, feelings, and concerns • Active listening • Providing time and space for reflection and questions • Encouraging the patient to be involved in treatment decisions • Full and clear explanation of all medical terms used • Checks of patient’s understanding and recall of information • Sensitivity to the person’s age, gender, ethnicity, literacy level, and cultural background • Having the people wanted by the patient present in the consultation • Provision of information about what a patient is likely to experience before, during, and after a procedure or clinical encounter • Use of simple diagrams and pictures to support communication, where appropriate • Provision of a tape of, or written information summarizing, the consultation • Use of a “question prompt sheet” to help the patient ask potentially relevant and important questions In addition to considering the vulnerability factors for psychological distress in Table 21.4, routine screening with a brief screening tool, such as the Distress Thermometer, may assist in identifying patients requiring additional support. Some patients who screen positively on the Distress Thermometer may not want psychological treatment but could benefit from educational interventions,23 while those who screen positively and want additional psychological care may benefit from interventions including psychotherapy alongside education.16 Recently there has been an explosion of research into online delivery of mental health care (i.e., eHealth or mHealth) to increase accessibility and affordability and bring effective interventions to scale. Meta-analytic evidence suggests small to moderate effect sizes, often equivalent to face-to-face care.26 While we know of no online or smartphone applications (“apps”) specifically designed to meet the psychological needs of melanoma patients, there are a growing number of apps for the monitoring or tracking of skin lesions, and this area is likely to expand exponentially in the future. Tailored education and skills-based training on how to recognize and address the needs of patients experiencing psychological difficulties, including timely referral to appropriate resources and services, is recommended for health professionals. While an empathic, patient-centered approach can greatly assist patients and their families, recognition of the emotional challenges, including burnout and compassion fatigue, experienced by health professionals working in this field is also vital. Strategies to address these concerns are likely to lead to improved therapeutic relationships and enhanced professional and patient satisfaction. REFERENCES 1. Simard S, Thewes B, Humphris G, et al. Fear of cancer recurrence in adult cancer survivors: A systematic review of quantitative studies. J Cancer Survivorship. 2013;7:300–322. 2. Yamamoto M, Sondak VK. Epidemiology, Risk Factors, and Clinical Presentation of Melanoma. New York: Oxford University Press; 2015. 3. Matthews NH, Li W, Qureshi AA, Weinstock MA, Cho E. Epidemiology of melanoma In: Ward WH, Farma JM, eds. Cutaneous Melanoma Etiology and Therapy. Brisbane, Australia: Codon Publications; 2017. 4. Schadendorf D, van Akkooi ACJ, Berking C, et al. Melanoma. Lancet. 2018;392(10151):971–984. 5. Ko JM, Geller AC, Swetter SM. Melanoma In: The American Cancer Society’s Oncology in Practice. Hoboken, NJ: John Wiley & Sons; 2018. 6. Hamel JF, Pe M, Coens C, et al. A systematic review examining factors influencing health related quality of life among melanoma cancer survivors. Eur J Cancer. 2016;69:189–198. 7. Malkhasyan KA, Zakharia Y, Milhem M. Quality-of-life outcomes in patients with advanced melanoma: A review of the literature. Pigment Cell Melanoma Res. 2017;30(6):511–520. 8. Dieng M, Kasparian NA, Cust AE, et al. Sensitivity of preference- based quality-of-life measures for economic evaluations in early- stage melanoma. JAMA Dermatol. 2018;154(1):52–59. 9. Vogel RI, Strayer LG, Ahmed RL, Blaes A, Lazovich D. A qualitative study of quality of life concerns following a melanoma diagnosis. J Skin Cancer. 2017;2017:2041872. 10. Hyatt A, Drosdowsky A, Williams N, et al. Exercise behaviors and fatigue in patients receiving immunotherapy for advanced melanoma: A cross-sectional survey via social media. Integr Cancer Ther. 2019;18:1534735419864431. 11. Dunn J, Watson M, Aitken JF, Hyde MK. Systematic review of psychosocial outcomes for patients with advanced melanoma. Psychooncology. 2017;26(11):1722–1731. 12. Kasparian NA, McLoone JK, Butow PN. Psychosocial responses and coping strategies among patients with malignant melanoma: A systematic review of the literature Arch Dermatol. 2009;145(12):1415–1427. 13. Mehnert A, Brähler E, Faller H, et al. Four-week prevalence of mental disorders in patients with cancer across major tumor entities. J Clin Oncol. 2014;32(31):3540–3546. 14. Costa DSJ, Dieng M, Cust AE, Butow PN, Kasparian NA. Psychometric properties of the Fear of Cancer Recurrence Inventory: An item response theory approach. Psycho-Oncol. 2016;25(7):832–838. 15. van de Wal M, van de Poll-Franse L, Prins J, Gielissen M. Does fear of cancer recurrence differ between cancer types? A study from the population- based PROFILES registry. Psycho-Oncol. 2016;25(7):772–778. 16. Dieng M, Butow PN, Costa D, et al. Psycho-educational intervention to reduce fear of cancer recurrence in people at high risk of developing another primary melanoma: Results of a randomised controlled trial. J Clin Oncol. 2016;34(36):4405–4414. 17. Dieng M, Morton RL, Costa DSJ, et al. Benefits of a brief psychological intervention targeting fear of cancer recurrence in people at high risk of developing another melanoma: 12-month follow-up results of a randomized controlled trial. Br J Dermatol. 2020;182(4):860–868. 18. Kasparian N, Meiser B, Butow P, Simpson J, Mann G. Predictors of psychological distress among individuals with a strong family history of malignant melanoma. Clin Genet. 2008;73:121–131. CHAPTER 21 Melanoma 19. Kasparian N, Meiser B, Butow P, Simpson J, Mann G. Genetic testing for melanoma risk: A prospective cohort study of uptake and outcomes among Australian families. Genet Med. 2009;11(4):265–278. 20. Hamama-Raz Y, Solomon Z, Schachter J, Azizi E. Objective and subjective stressors and the psychological adjustment of melanoma survivors. Psycho-Oncol. 2007;16:287–294. 21. Trask PC, Paterson AG, Hayasaka S, Dunn RL, Riba M, Johnson T. Psychosocial characteristics of individuals with non-stage IV melanoma. J Clin Oncol. 2001;19(11):2844–2850. 22. Beesley VL, Smithers BM, Khosrotehrani K, et al. Supportive care needs, anxiety, depression and quality of life amongst newly diagnosed patients with localised invasive cutaneous melanoma in Queensland, Australia. Psychooncology. 2015;24(7):763–770. 23. McLoone JK, Menzies SW, Meiser B, Mann GJ, Kasparian NA. Psycho-educational interventions for people affected by melanoma: A systematic review. Psycho-Oncol. 2013;22:1444–1456. 24. Kasparian NA, Mireskandari S, Butow PN, et al. “Melanoma: Questions and answers.” Development and evaluation of a psycho- educational resource for people with a history of melanoma. Support Care Cancer. 2016;24(12):4849–4859. 25. Dieng M, Khanna N, Kasparian NA, et al. Cost-effectiveness of a psycho-educational intervention targeting fear of cancer recurrence in people treated for early-stage melanoma. Appl Health Econ Health Policy. 2019;17(5):669–681. 26. Kim AR, Park HA. Web-based self-management support interventions for cancer survivors: A systematic review and meta- analyses. Stud Health Technol Inform. 2015;216:142–147. 161 22 Lung Cancer Marianne Davies Introduction In the past decade, there have been significant advances in the treatment of non–small cell lung cancer. Despite these advances, lung cancer remains the leading cause of cancer-related mortality worldwide. Lung cancer is usually diagnosed at advanced stages, associated with complex disease-related symptoms, leading to overall poor prognosis. As a result, patients, their families, and their caregivers experience significant levels of psychosocial distress, greater than those with other cancers. This chapter will provide an overview of the burden of lung cancer, associated preventative risk factors, screening recommendations, diagnostic considerations, treatment landscape, and psychological considerations through the disease trajectory. Epidemiology Lung cancer is the leading cause of cancer-related deaths in the United States (US) and worldwide. In 2018, over 2 million persons were diagnosed with lung cancer, with 1,761,007 associated death worldwide. This accounts for 18% of cancer-related deaths.1,2 In the US alone, it is estimated that there will be 228,820 new cases with 135,720 deaths. This accounts for 25% of all cancer deaths, more than breast, prostate, colorectal, and brain combined.3 Survival rates have slowly improved over the past years. Approximately 57% of patients are diagnosed with advanced-stage disease, with a 5-year relative survival of only 5%. For those diagnosed with regional disease, the 5-year relative survival rate is 33%, and for those with localized disease, it is 61%.3 With diagnostic and treatment advances, the number of people living longer with lung cancer is expected to grow, contributing to a larger population living with complex needs. Worldwide, lung cancer incidence is increasing for men and women, with rates higher in developed countries.1 In contrast, lung cancer death rates in the US have dropped by 51% among males since 1990 and by 26% in females since 2002.3 This is the most rapid decline in recent years. The decline in death rates in the US reflects tobacco cessation patterns, while the rise in incidence worldwide is associated with increased tobacco use. In contrast, there is an increasing incidence of lung cancer in never smokers (LCINS). LCINS is the seventh leading cause of death in the US and worldwide. It accounts for approximately 15% of cases in the US and 25% of new cases worldwide.1,3 LCINS is more common in women than men.4 There has also been an emerging trend of increased incidence of lung cancer in young women compared with young men, not correlated with smoking patterns, suggesting gender-specific histologic changes in the lung over time.5 Lung cancer patterns are influenced by smoking habits, age, gender, race/ethnicity, socioeconomic region, and education level. The median age of diagnosis among smokers is 70 years old. The risk of death from smoking-related lung cancer is correlated with age of initiation, with greatest risk in those who begin in their early teens or younger. It is also influenced by the number of packs smoked, total number of years, depth of inhalation, and type of product smoked. The incidence and mortality from lung cancer vary by race and ethnicity within each country. This may be attributed to access to care, later stage at diagnosis, and environmental influences. Mortality rates are higher in lower socioeconomic regions and among those with less than a high school education. Risk Factors The single greatest risk for development of lung cancer is smoking, attributed to 90% of cases, outweighing all other factors combined. People who smoke are 15% to 30% more likely to develop lung cancer than nonsmokers. Second-hand smoke contributes to 7,000 deaths from lung cancer in the US alone.3 Both primary and secondary smoke impact are dose dependent. Electronic cigarettes, sometimes called vapes, have sparked considerable controversy over risks from long-term use. The devices deliver nicotine, along with other chemicals, into the lungs. Initially marketed as a tool to support smoking cessation, there has been societal uptake in use, particularly among children and young adults. Use of these devices is associated with greater risk of subsequent cigarette smoking. There is also an increased risk of addiction, permanent lung damage, and cancer with use. The risk is increased for users of multiple tobacco products.6 The Centers for Disease Control and Prevention (CDC), US Department of Health and Human Services, and US Surgeon General have partnered to promote a national campaign, “Know the Risks: e-Cigarettes & Young People,”6 aimed at the reduction of use of e-cigarettes in the young population. CHAPTER 22 Lung Cancer In the US, the second leading cause of lung cancer is due to radon, contributing to 20,000 deaths per year.3 Radon, a naturally occurring radioactive gas released from soil as it decays, can seep into homes and get trapped. Persons’ exposure to asbestos, historically found in insulation, in the workplace (i.e., shipyards, building contractors, mines, and factories) are several times more likely to die from lung cancer. This risk is increased in people who smoke. Additional hazardous exposures include metals (i.e., chromium, cadmium, arsenic), radiation exposure (i.e., chest radiation), air pollution, diesel exhaust, aromatic hydrocarbons, indoor air pollution from use of unprocessed fossil fuels, and biomass (coal and wood). Several additional factors have been suggested to contribute to lung cancer, including history of chronic lung infections or injuries, age, and genetic predispositions. Diets rich in cruciferous vegetables and fruits may exert some protection, while high intake of red meats may increase the risk of developing lung cancer. Prevention and Early Detection Cigarette smoking is the number one preventable cause of disease and lung cancer. Prevention of smoking uptake and promotion of smoking cessation efforts is the most impactful strategy to reduce the lung cancer burden worldwide. Smoking cessation before the age of 40 reduces the risk of dying from smoking-related disease by about 90%. Former smokers remain at increased risk of developing lung cancer; however, by 10 years after quitting, the risk of dying from lung cancer is half of that of one who continues to smoke.6 Smoking cessation after lung cancer diagnosis offers many physical and mental benefits. This includes better outcomes from cancer- directed treatment, faster recovery from treatments, improved survival, and improved health-related quality of life (HRQOL).7 Continuing to smoke, after a lung cancer, is associated with an increase in treatment-related complications, decreased efficacy of treatments, decreased HRQOL, increased risk of recurrence, development of second cancers, and decreased survival.8 Counseling and motivational interviewing can be effective strategies to support smokers through the cessation process. Electronic cigarettes and other nicotine substitutes may be helpful when used as part of an evidence-based smoking cessation program with cognitive- behavioral intervention. Disappointingly, many patients continue to smoke beyond diagnosis. Stigma and nihilism, on the part of patients and health care providers, may influence referrals to and participation in smoking cessation efforts. It is imperative of all health care providers to encourage and provide access to smoking cessation programs. Early-stage lung cancer is usually asymptomatic. The survival rate of persons diagnosed at a localized stage approximates 61%. Unfortunately, a large percentage of patients present with advanced disease, when a patient is symptomatic. The National Lung Cancer Screening Trial (NLST), a large randomized controlled study of over 50,000 healthy individuals at high risk for lung cancer, demonstrated that screening with a low-dose computed tomography (LDCT) scan led to a 20% relative reduction in the mortality from lung cancer compared with standard yearly chest x-ray.9 Based on these results, the US Preventive Services Task Force (USPSTF), the American Cancer Society (ACS), and several other organizations recommend annual screening of high-risk individuals aged 55 to 80 years with a 30-pack-year smoking history who currently smoke or who have quit within the past 15 years.10 However, there is a high rate of benign nodules, and thus false positives, identified. Shared decision-making strategies should be incorporated to guide patients through the evaluation of the potential benefits, risks, and harm of screening. The screening process may infer psychosocial benefits (i.e., earlier diagnosis and potential for behavior change such as smoking cessation) as well as harms (i.e., worry, fear, distress with false-positive results, or unwarranted reassurance with false-negative results). The American Thoracic Society (ATS) and American College of Chest Physicians (ACCP) stress the importance of a structured, multidisciplinary screening program that includes patient counseling, support, and follow-up.11 Patients with more advanced lung cancer often have subtle symptoms for several months before seeking medical evaluation. The most common symptoms in patients are dry cough, dyspnea, fatigue, weight loss, chest pain, hemoptysis, and hoarseness. Weight loss and poor performance status at presentation are poor prognostic indicators of overall survival. All patients suspected of having lung cancer should begin prompt diagnostic evaluation. Diagnosis, Classification, and Staging The initial diagnostic imaging test obtained is typically a chest x- ray, which should be compared to prior chest imaging to determine changes. All suspicious findings on x-ray or exam should be evaluated further by chest computed tomography (CT) or positron emission tomography (PET) scan. Additional diagnostic imaging, including CT of the abdomen/pelvis and magnetic resonance imaging (MRI) of the brain, may be indicated to determine the extent of disease burden. The extent of disease identified will guide the next steps in diagnosis. Patients suspected of lung cancer on diagnostic imaging must undergo a tissue biopsy. Biopsy can be obtained through endobronchial ultrasound (EBUS), bronchoscopy, transthoracic CT-guided biopsy, or thoracentesis. If the patient is a candidate for surgical resection, a mediastinoscopy should be done for biopsy and staging of mediastinal node involvement. The least invasive biopsy with highest potential yield of tissues is recommended. Adequate tumor tissue is necessary for histologic classification, immunohistochemical (IHC) analysis, and focused molecular profiling. A liquid biopsy is an alternative strategy for diagnosis if insufficient tissue is obtained at biopsy, a tumor is not accessible by conventional methods, or the patient is too frail to undergo an invasive procedure. Liquid biopsy captures circulating tumor cells, and tumor DNA fragments from blood, serum, urine, and pleural fluid can be evaluated for histologic classification and molecular profiling. There are two major classifications of lung cancer: non–small cell lung cancer (NSCLC), making up 85%, and small cell lung cancer (SCLC), accounting for 15%. NSCLC is further classified into three histologic subtypes: adenocarcinoma (40%), squamous cell carcinoma (25%), and large cell carcinoma (10%).12 Adenocarcinoma is the most common variant identified in LCINS. There have been major advances in the identification of driver gene mutations in lung cancer through molecular profiling. All patients with confirmed advanced-stage lung cancer should undergo additional mutational testing at time of diagnosis, to direct potentially efficacious targeted 163 164 SECTION V Psychological Issues Related to Site of Cancer therapies and avoidance of therapies unlikely to be of benefit. There are several different testing platforms available and these will vary across institutions. In some cases, tissue specimens may have to be sent out to third-party testing sites. Programmed death ligand-1 (PD-L1) a coregulator molecule that can be expressed on tumor cells and inhibit T-cell-mediated cell death. T-cells express PD-1, a negative regulator. Binding of PD-1 to PD-L1/ PD-L2 leads to suppression of T-cell activity. CTLA-4 is a negative regulator present in the primary lymph nodes. Immune checkpoint inhibitors have been developed to block PD-1, PD-L1, and CTLA4. The presence of PD-L1 expression on tumors may be used to inform treatment selection. In general, PD-L1 expression is higher in patients with a higher tumor mutation burden, such as that seen in smokers. The most common single driver mutations in adenocarcinoma are EGFR mutation (25%), EMLA-4 ALK rearrangements (5%), NTRK (6%), KRAS (20%), and ROS1 (1%). Additional emerging mutations include AKT, BRAF, FGFR, ERBB2/HER2, MET, PTEN, and RET. Most of the driver mutations are found in never or light smokers. KRAS, however, is more common in smokers. Rarely are these driver mutations seen concurrently (1%–3%). It should be noted that the rates of mutations that have emerged vary by country, with Asian nonsmokers harboring a higher percentage of single driver mutations. The National Comprehensive Cancer Network (NCCN) recommends that all patients with NSCLC be tested for EGFR, ALK, ROS1, BRAF, NTRK, and PD-L1.13 In addition, they support broad profiling to help identify rarer mutations for which there might already be effective treatments available. Regardless of smoking history, all patients should be tested to identify patients that might benefit from targeted therapy. Genetic mutations are being investigated in other subtypes of lung cancer as well. Lung cancer is staged based on the internationally accepted TNM staging system last updated in 2018.14 Regular updates reflect advances in prognostic and survival data. TNM stage is based on the size of the primary tumor (T) at the greatest dimension and level of invasion into surrounding structure, extent of regional lymph node (N) involvement, and the presence or absence of metastatic spread outside of lung tissue or distant disease. The stages range from I to IV, with further detailed subclassifications to help guide treatment selection. Stage I and II are localized, Stage III is regional, and Stage IV is metastatic. Stage IV includes oligometastatic disease as well as widespread metastases. Historically, SCLC was classified using a two-stage system: limited and extensive disease. Limited disease is that which can be encompassed in a radiation therapy field. Cancer-Directed Treatment The treatment of lung cancer has been evolving rapidly over the past two decades. Advances in diagnostic approaches, molecular profiling, and development of targeted therapies have contributed to the improvement in survival and HRQOL. In addition, targeted therapies and immune checkpoints, initially only used for advanced disease, are being investigated in early-stage disease in an attempt to reduce recurrence of disease. Five-year survival rates for early-stage localized, regional, and advanced NSCLC are 61%, 35%, and 6%, respectively. However, the rates for SCLC remain much lower at 27%, 16%, and 3%, respectively.3 Early-stage NSCLC (Stage I) is treated with surgery as curative intent. In Stage II, adjuvant chemotherapy or chemoradiation following surgical resection is recommended. For patients that are not surgical candidates, definitive radiation is used. The foundation of treatment for Stage III NSCLC is combined- modality treatment. Treatment includes concurrent or sequential chemotherapy and radiation therapy prior to or following surgical resection for Stage IIIA. In Stage IIIB, with unresectable tumor, therapy is concurrent or sequential chemotherapy over four to six cycles. Patients then remain in close surveillance. In 2018, the paradigm shifted in the management of Stage III unresectable disease following combination therapy. The PACIFIC study demonstrated improved overall survival (66.3% vs. 55%) in patients receiving durvalumab, an anti- PD- L1 immune checkpoint inhibitor, for 1 year following definitive chemoradiotherapy treatment versus surveillance.15 It is not recommended in patients who have undergone a surgical resection. The goal of treatment for Stage IV NSCLC is to control disease, palliate symptoms, and improve survival. Treatment selection is based on histologic subtype, presence of molecular driver mutation, level of PD-L1 expression, performance status, and presence of comorbidities. EGFR, ALK, NTRK, BRAF V600E, and ROS 1 are driver mutations more common in nonsmokers, for which targeted tyrosine kinase inhibitors (TKIs) are indicated. Unfortunately, almost all patients develop progression of disease within 10 to 12 months on first-generation TKIs. This is due to a tumor’s acquired mechanisms of resistance. For EGFR, a common mechanism of resistance is the acquired T790 mutation at exon 20 of the EGFR gene. Osimertinib is a third-generation EGFR-TKI developed to overcome this mutation. It has subsequently been moved into the first-line setting, demonstrating improved efficacy, ability to cross the blood-brain barrier, and reduced toxicity profile. Similarly, first-generation ALK TKIs have been replaced by next-generation alectinib for patients with ALK translocations. The first-generation TKIs crizotinib and larotrectinib are currently approved for ROS1 and NTRK mutations, respectively. Combination dabrafenib and trametinib is approved for treatment of NSCLC with BRAF V600E mutations. KRAS mutations are found in approximately 25% of lung adenocarcinomas and are more common in smokers. KRAS G12C is a subset that accounts for approximately 13% of NSCLC. New TKIs targeting this subset are currently in development with promising results. It is likely that the accelerated rate of personalized targeted therapy will continue to evolve. The Lung-MAP study is a multicenter study sponsored by the National Cancer Institute’s National Clinical Trials Network (NCTN). Patients underdo a comprehensive genomic profiling that evaluates over 200 cancer-related genes for alterations. Based on genomic profiling results, patients are randomized to substudies and matched to investigational drugs or offered an immunotherapy combination. The benefits of a targeted TKI are convenience and overall improved toxicity profile than cytotoxic chemotherapy. However, treatment can be associated with anxiety related to the molecular tumor profiling (MTP) process, anticipation of treatment resistance, need for repeat biopsies, increased responsibility of self-management, and financial toxicity. Patients experience anxiety while awaiting the results of MTP, hopeful to have an actionable mutation and disappointed or depressed if no mutation is identified.16 Many of the oral oncolytics cost more than $100,000 per year.17 Copays alone can CHAPTER 22 Lung Cancer place a financial burden on patients and families. This can impact adherence to therapy and treatment efficacy. Cytotoxic chemotherapy and immune checkpoint therapy, alone or in combination, are recommended for patients that progress on TKI therapy or do not harbor any driver mutations. Treatment decisions are based on performance status, underlying comorbid medical conditions, burden of disease, and level of PD-L1 expression. Although PD-L1 expression can be elevated in patients with single driver mutations, targeted therapy is recommended first. Maintenance therapy may continue in patients who respond to initial treatment. Radiation therapy may be indicated to palliate symptoms or relieve obstructive disease. Stereotactic body radiation therapy (SBRT) may be useful to target small oligometastatic disease, and gamma knife radiosurgery is recommended for brain metastases. Limited or early-stage SCLC is treated with combination chemotherapy and radiation therapy. Extensive-stage SCLC is treated with systemic chemotherapy. Strong consideration is given to prophylactic whole brain radiation in patients with SCLC. To date, there are no molecular driver mutations in SCLC for which there are available drugs. Psychological Issues Commonly Faced in Lung Cancer Distress, Anxiety, and Depression. Patients with lung cancer have a high symptom burden from cancer and cancer treatments due in part to late disease stage at diagnosis.18,19 Symptom burden can result in impaired emotional status, social functioning, and quality of life (QOL). The prevalence of distress in newly diagnosed lung cancer patients has been reported to be the highest compared to other cancers, with up to 61% reporting distress that often persists through illness.20 Anxiety and depression rates are higher in lung cancer patients than in those with other cancers.21 Pain, fatigue, breathlessness, and sleep disturbances all contribute to higher levels of distress, anxiety, and depression.22 Smokers experience higher levels of anxiety over nonsmokers.23 Patients who are younger at diagnosis, female, employed, and with later-stage disease are more likely to report greater severity of emotional problems at diagnosis.24,25 Greater severity of emotional problems is associated with worse QOL indicators (i.e., mental, physical, social, spiritual, and emotional), an increase in physical symptoms burden, reduced adherence to treatment, and decreased survival.25–27 In addition, this contributes to a 4.4 times higher rate of suicide among lung cancer patients compared to other cancer diagnoses, which is most significant at time of diagnosis and in patients over age 50.28 Despite the high prevalence of unmet needs, psychological distress remains unrecognized and suboptimally managed, which can lead to poorer quality of life, lower satisfaction with care, poorer adherence to treatment, and decreased survival.25,29 Stigma. Lung cancer patients experience higher levels of internal and external stigma through the disease trajectory. Stigma develops due to a strong association with smoking history and perception that the disease is self-inflicted.30 Approximately 95% of lung cancer patients experience at least one element of stigma.31 Internalized Table 22.1. Lung Cancer–Specific Assessment Tools Instrument No. Items Dimensions Measured Coping Orientation to Problems Experienced (COPE) 60 Problem-focused coping and emotion-focused coping and measures of coping responses; a Brief COPE Inventory consists of only 28 statements European Organization for Research and Treatment of Cancer Quality of Life Questionnaire Core 30 (QLQ-C30) 30 Global health status, functional scales (physical, life role, cognition, emotional, and social), and symptom scales (fatigue, nausea/vomiting, pain, dyspnea, insomnia, appetite loss, constipation, diarrhea, and financial problems) Lung Cancer Specific Module (EORTC-QLQ-LC13) 13 Dyspnea, coughing, dysphagia, hemoptysis, sore mouth/tongue, peripheral neuropathy, hair loss, chest pain, pain in arm/shoulder, other pain sites, taking analgesics Cataldo Lung Cancer Stigma Scale (CLSS) 31 Self-perceived stigma; also available in short form; adapted from tools used in HIV Lung Cancer Stigma Inventory 25 Overall stigma, perceived stigma, internalized stigma, and constrained disclosure; developed specifically for lung cancer Edmonton Symptom Assessment Scale Functional Assessment of Cancer Therapy (FACT) FACT-Lung Cancer FACT- EGFRI-18 9 27 7 18 Lung Cancer Symptom Scale (LCSS) Psychosocial Screen for Cancer (PSSCAN-R) questionnaire General core instrument: physical, social/family, emotional, and functional well-being Lung cancer–specific disease symptoms Patient-reported outcomes to assess effect of EGFRI on health-related quality of life Measure of six major symptoms (cough, dyspnea, fatigue, pain, hemoptysis, and anorexia) and effect on overall symptom distress, functional activities, and global quality of life. Two scales: one completed by patient and one by health care provider 21 Schedule for the Evaluation of Individual Quality of Life (SEIQoL-Q) Sheffield Profile for Assessment and Referral to Care (SPARC®) Physical and psychological symptoms of anxiety and depression Social support, psychosocial needs, and symptoms of anxiety and depression Self-reporting tool; 12 life domains, weighted and individual quality of life index is calculated 45 Assess level of distress in 7 broad areas including physical and psychosocial, as well as need for services 165 166 SECTION V Psychological Issues Related to Site of Cancer stigma stems from feelings of shame and guilt, self-blame, and anticipated stigma. Anticipated stigma may lead to constrained disclosure, the avoidance of sharing one’s diagnosis with others. External stigma and biases lead to discrimination, blame, and social rejection. Evidence suggests pervasive societal stigma toward lung cancer patients and their caregivers over other cancers.32–34 Even patients who have never smoked experience societal stigma. Stigma is associated with reduced involvement in screening, delays in early detection for symptoms, reduced treatment adherence, and social isolation.30,35,36 Stigma leads to increased social isolation, loneliness, depression, and poor QOL.31,32,36,37 Patients with higher levels of stigma are more likely to have a higher symptom burden, decreased social support, and increased social constraints, leading to poorer health outcomes.38 In response to the impact of societal stigma, the Lung Cancer Alliance launched a campaign in 2012 to combat stigma with the theme “No One Deserves to Die.” All patients diagnosed with lung cancer should be screened for stigma so that interventions can be put into place to minimize the negative impact on health outcomes. Family-Centered Concerns. Family members may be at increased risk of developing lung cancer. First-degree relatives living in the same household are at double the risk.39 This leads to strain in the relationship due to blame. The caregiver dyad may be strained by feelings of self-guilt if the family/caregiver were the predominant smokers in the household. Caring for patients with long-term cancer can be a long journey, as cancer survival rates have been gradually increasing with advances in therapies and supportive care. Lung cancer patients have higher supportive care needs due to higher symptom burden compared to other cancers. This places strain on the caregiver dyad. Caregiver burden is associated with the patient’s physical and psychological functioning. Patient QOL is linked to higher level of burden and more severe emotional problems in caregivers.40 These issues reinforce the need to address the psychological needs of the dyad. Table 22.2. Education and Support Services for Patients with Lung Cancer Organization Website/Contact Description and Resources ALK Positive http://www.alkpositive.org Online support for patients, families, and friends of those diagnosed with ALK- positive lung cancer; advocacy and education American Cancer Society http://www.cancer.org Online patient and family educational materials; support for finding and paying for treatment; caregiver support; end-of-life support; support is across the lifespan American Lung Association https:/www.lung.org Research support; anti-tobacco campaigns; education and training; CT screening support; LUNG FORCE Initiative: women with lung cancer Cancer Support Community https://www.cancersupportcommunity.org Merger of the Wellness Community, Gilda’s Club, and MyLifeLine; research, advocacy, education; walk-in centers to provide emotional and social support; digital support network Caring Ambassador Lung Cancer Program https://lungcancercap.org/ Mission is to help patients with lung cancer become ambassadors for their own health; advocacy and educational resources The CHEST Foundation https://foundation.chetnet.org Supports research, community service, and patient education related to lung disease including cancer CURE® https://www.curetoday.com/tumor/lung Cancer updates, research, and education EGFResisters https://egfrcancer.org Advocacy for patients diagnosed with EGFR-positive gene lung cancer; supports research Global Resource for Advancing Cancer Education (GRACE) https://cancergrace.org Provides education online, in forums, and videos; online communities GO2 Foundation for lung cancer https://go2foundation.org Support@go2foundation.org Advocacy; merger of two previous organizations, the Bonnie J. Addario Lung Cancer Foundation (ALCF) and Lung Cancer Alliance (LCA); educational, support, and referral services International Association for the Study of Lung Cancer (IASLC) http://www.iaslc.org Research, collaboration, education, patient advocacy Lung Cancer Action Network (LungCAN®) http://lungcan.org A collaborative group of lung cancer advocacy organizations joined to raise awareness about lung cancer; educational resources; links to clinical trials and support services Lungevity https://lungevity.org Patient advocacy organization; funds scientific and patient-focused research; provides information and resources to patients and families; educational resources, online peer-to-peer support, and in-person survivorship programs Lung Cancer Research Foundation http://www.lungcancerresearchfoundation. org Supports clinical and bench research; patient and caregiver educational materials and workshops; lung cancer support line; Free to Breath walks Lung Cancer Foundation of America https://lcfamerica.org Supports research; patient education Mesothelioma Applied Research Foundation http://www.curemesoorg Advocacy, education, support groups, financial assist Stand Up to Cancer https://standuptocancer.org Advocacy; funds research; education CHAPTER 22 Lung Cancer Survivorship Concerns. Due to poor prognosis for advanced lung cancer, survivorship is described as beginning at 1 year postdiagnosis. Lung cancer survivors suffer high disease burden and endure symptoms long after therapy that negatively impact QOL indicators (i.e., global health and physical, role, emotional, cognitive, and social functioning).41 Symptoms of dyspnea, pulmonary restrictions, pain, and poor sleep quality are some of the many residual symptoms that have been reported up to 3 years after diagnosis, leading to poor QOL.42 Lung cancer survivors have higher levels of distress compared with survivors of other cancers.25,31 At least 40% of lung cancer survivors report distress that negatively impacts QOL.19 Patients with lung cancer should continually be assessed for the psychological impact of cancer throughout survivorship. Assessment of Psychological Distress Distress screening is recommended by the Commission on Cancer (COC) and endorsed by the NCCN, Oncology Nursing Society (ONS), and American Society of Clinical Oncology (ASCO) to be done at least once during pivotal points in the disease trajectory. The distress screening tool has demonstrated predictive value in estimating 1-year survival and may guide the integration of palliative care services.43 Several assessment tools have been developed to assess quality of life, symptoms, functional capacity, and stigma in cancer patients. Some of the tools are generic, while others have been developed to capture dimensions specific to lung cancer patients. All patients with lung cancer should undergo screening for psychological stressors at the time of diagnosis and throughout the disease trajectory. Early identification of needs provides opportunity to intervene and support patients. A summary of tools is listed in Table 22.1. Supportive Services Supportive care interventions that are aimed at reducing distress can lead to improved QOL. Palliative care support services can improve depression and QOL in lung patients. In the randomized study of patients with lung cancer, early integration of palliative care services demonstrated significant improvements in QOL with less aggressive care at end of life with longer survival benefit.44,45 Ferrell et al.46 assessed the impact of an interdisciplinary approach versus standard of care on lung cancer patients undergoing treatment. The intervention group patients were discussed at interdisciplinary meetings and received additional supportive care referral and four educational sessions. Patients in the intervention group had lower psychological distress, greater spiritual well-being, improved symptoms, and improved QOL. Shared decision making (SDM) is an important element of delivering comprehensive care to lung cancer patients, especially for those in whom there is no curative option. SDM is an approach in which providers present the best available evidence to patients when making a health care decision and integrates the patient’s goals of care in deciding on a plan of care. It is particularly helpful for preference-sensitive decisions. The use of SDM tools and decision aids can guide the direction of screening, testing, and treatment. Currently, there is a dearth of SDM tools developed specifically for lung cancer patients.47 Professional organizations and patient advocacy groups are available to provide additional supportive care services to lung cancer patients in the community. Table 22.2 provides a list of such organizations and descriptions of available services. REFERENCES 1. Ferlay J, Colombet M, Soerjomataram I, et al. Estimating the global cancer incidence and mortality in 2018. Int J Cancer. 2018. 144. 1941–1953. 2. Barta JA, Powell CA, Wisnivesky JP. Global epidemiology of lung cancer. Ann Glob Health. 2019. 85 (1). doi:10.5334/aogh.2419 3. Siegel RL, Miller KD, Jemal A. Cancer statistics 2020. CA Cancer J Clin. 2020. 70: 7–30. 4. Courade S, Zakman G, Millero B, et al. Lung cancer in never smokers-a review. Eur J Cancer. 2012. 48: 1299–1311. 5. Fidler-Benaoudia MM, Torre LA, Bray F, Ferley J, Jemal A. Lung cancer incidence in young women vs. young men: a systematic analysis in 40 countries. Int J Cancer. 2020 [Epub ahead of print]. doi:10.1002/ijc.32809 6. US Department of Health & Human Services. Smoking Cessation: A Report of the Surgeon General. Atlanta, GA: U.S. Department of Health and Human Services, Centers for Disease Control and Prevention, National Center for Chronic Disease Prevention and Health Promotion, Office on Smoking and Health, 2020. 7. Alberg AJ, Shopland DR, Cummings KM. The 2014 Surgeon General’s report: commemorating the 50th Anniversary of the 1964 Report of the Advisory Committee to the US Surgeon General and updating the evidence on the health consequences of cigarette smoking. Am J Epidemiol. 2014. 179: 403–412. 8. Jassem J. Tobacco smoking after diagnosis of cancer: clinical aspects. Transl Lung Cancer Res. 2019. 8 (Suppl 1): S50–S58. doi:10.21037/tlcr.2019.04.01 9. NLSTR Team. Reduced lung cancer mortality with low-dose computed tomographic screening. N Engl J Med. 2011. 365: 395–409. 10. Moyer VA, on behalf of the U.S. Preventive Services Task Force. Screening for lung cancer: U.S. Preventive Services Task Force Recommendation Statement. Ann Intern Med. 2014. 160 (5): 330–338. 11. Mazzone P, Powell CA, Arenberg D, et al. Components necessary for high-quality lung cancer screening: American College of Chest Physicians and American Thoracic Society Policy Statement. Chest. 2015. 147 (2): 295–303. 12. Travis WD, Brambilla E, Nicholson AG, et al., on behalf of the WHO Panel. The 2015 World Health Organization Classification of lung tumors. Impact of genetic, clinical and radiologic advances since 2004 classification. J Thorac Oncol. 2015. 10 (9): 1243–1260. 13. National Comprehensive Cancer Network Inc. The NCCN Biomarkers Compendia®. https://www.nccn.org/professionals/biomarker/. Accessed: February 20, 2020. 14. Detterbeck FC, Boffa DJ, Kim AW, Tanoue LT. The 8th edition lung cancer stage classification. Chest. 2017. 151 (1): 193–203. 15. Antonia SJ, Villegas A, Daniel D, et al., for the PACIFIC Investigators. Overall survival with Durvalumab after chemoradiotherapy in Stage III NSCLC. NEJM. 2018. 379: 2342–2350. 16. Best MC, Bartley N, Jacobs C, et al., and members of the PiGeOn Project. Patient perspectives on molecular profiling: “why wouldn’t you?” BMC Cancer. 2019. 19: 753. 167 168 SECTION V Psychological Issues Related to Site of Cancer 17. Doshi JA, Li P, Huo, H, Pettit AR, Armstrong KA. Association of patient out-of-pocket costs with prescription abandonment and delay in fills of novel oral anticancer agents. J Clin Oncol. 2018. 36: 476–482. 18. Carlson LE, Waller A, Groff SL, Bultz BD. Screening for distress, the sixth vital sign, in lung cancer patient: effects on pain, fatigue and common problems-secondary outcomes of a randomized controlled trial. Psycho-Oncology. 2013. 22. 1880–1888. 19. Zabora J, Brintzenhofeszoc K, Curbow B, Hooker G, Piantadosi S. The prevalence of psychological distress by cancer site. Psychooncology. 2001. 10 (1): 19–28. 20. Sung MR, Patel MV, Djalalov S, et al. Evolution of symptom burden of advanced lung cancer over a decade. Lung Cancer. 2017. 18 (3): 274–280.e6. 21. Linden W, Vodermaier A, Mackenzie R, Greig D. Anxiety and depression after cancer diagnosis: prevalence rates by cancer type, gender and age. Affect Disord. 2012. 141: 343–351. 22. Sherry V, Guerra C, Ranganathan A, Schneider SM. Metastatic lung cancer and distress. CJON. 2017. 21 (3): 379–383. 23. Choi SH, Chan RR, Lehto RH. Relationships between smoking status and psychological distress, optimism, and health environment perceptions at time of diagnosis of actual or suspected lung cancer. Cancer Nurs. 2019. 42 (2): 156–163. 24. Kuon J, Vogt J, Mehnert A, et al. Symptoms and needs of patients with advanced lung cancer: early prevalence assessment. Oncol Res Treat. 2019. Published online: October 21, 2019. doi:10.1159/ 000502751 25. Morrison EJ, Novotny PJ, Sloan JA, et al. Emotional problems, quality of life, and symptom burden in patients with lung cancer. Clin Lung Cancer. 2017. 18 (5): 497–503. 26. Leung B, Laskin J, Wu J, Bates A, Ho C. Assessing the psychosocial needs of newly diagnosed patients with lung cancer: identifying factors associated with distress. Psycho-Oncology. 2019. 28: 815–821. 27. Polanski J, Jankowska-Polanska B, Rosinczuk J, Chabowski M, Szymanska-Chabowska A. Quality of life of patients with lung cancer. OncoTargets Ther. 2016. 9: 1023–1028. 28. Zaorsky NG, Zhang Y, Tuanquin L, Bluethmann SM, Park HS, Chhinchilli VM. Suicide among cancer patients. Nat Commun. 2019. 10: 207. doi:10.1038/s41467-018-08170 29. Sullivan DR, Forsberg CW, Ganzini L, et al. Longitudinal changes in depression symptoms and survival among patients with lung cancer: a national cohort assessment. J Clin Onc. 2016. 34 (33): 3984–3991. 30. Weiss J, Yang H, Weiss S, et al. Stigma, self-blame and satisfaction with care among patients with lung cancer. J Psychosoc Oncol. 2016. 35 (2): 166–179. 31. Hamann HA, Ostroff JS, Marks EG, Gerber DE, Schiller JH, Lee SJC. Stigma among patients with lung cancer: a patient-reported measurement model. Psycho-Oncology. 2014. 23: 81–92. 32. Ernst J, Mehnert A, Dietz A, Hornemann B, Esser P. Perceived stigmatization and its impact on quality of life-results from a 33. 34. 35. 36. 37. 38. 39. 40. 41. 42. 43. 44. 45. 46. 47. large register-based study including breast, colon, prostate and lung cancer patients. BMC Cancer. 2017. 17: 741. doi:10.1186/ s12885-017-3742-2 Mazieres J, Pujol J-L, Kalampalikis N, et al. Perception of lung cancer among the general population and comparison with other cancers. J Thoraci Oncol. 2015. 10 (3): 420–425. Sriram N, Mills J, Lang E, et al. Attitudes and stereotypes in lung cancer versus breast cancer. PLoS ONE. 2015. 10 (12): e0145715. Hamann HA, Ver Hoeve ES, Carter-Harris L, Studts JL, Ostroff JS. Multilevel opportunities to address lung cancer stigma across the cancer control continuum. J Thorac Oncol. 2018. 13 (8): 1062–1075. Occhipinti W, Dunn J, O’Connell DL, et al. Lung cancer stigma across the social network: patient and caregiver perspectives. J Thorac Oncol. 2018. 13 (10): 1443–1453. Hyland KA, Small BJ, Gray JE, et al. Loneliness as a mediator of the relationship of social cognitive variables with depressive symptoms and quality of life in lung cancer patients beginning treatment. Psycho-Oncology. 2019. 28: 1234–1242. Johnson LA, Schreier AM, Swanson M, Moye JP, Ridner S. Stigma and quality of life in patients with advanced lung cancer. Oncol Nurs Forum. 2019. 46 (3): 318–328. Musolf AM, Simpson CL, de Andrade M. et al. Familial lung cancer: a brief history from the earliest work to the most recent studies. Genes. 2017. 8 (1): 36. Tan J-Y, Molassiotis A, Lloyd-Williams M, Yorke J. Burden, emotional distress and quality of life among informal caregivers of lung cancer patients: an exploratory study. Eur J Cancer Care. 2018. 27: e12691. doi:10.1111/ecc.12691 Vijayvergia N, Shah PC, Denlinger CS. Survivorship in non-small cell lung cancer: challenges faced and steps forward. J Natl Compr Canc Netw. 2015. 13 (9): 1151–1161. Hechtner M, Eichler M, Wehler B, et al. Quality of life in NSCLC survivors-a multicenter cross-sectional study. J Thorac Oncol. 2019. 14 (3): 420–435. Geerse OP, Bradenbarg D, Kerstjens HAM, et al. The distress thermometer as a prognostic tool for one-year survival among patients with lung cancer. Lung Cancer. 2019. 130: 101–107. Temel JS, Greer JA, El-Jawahri A, et al. Effects of early integrated palliative care in patients with lung cancer and GI cancer: a randomized clinical trial. J Clin Oncol. 2017. 35: 834–841. Temel JS, Greer JA, Muzikansky A, et al. Early palliative care for patients with metastatic non-small cell cancer. NEJM. 2010. 363 (8): 733–742. Ferrell B, Sun V, Hurria A, et al. Interdisciplinary care for patients with lung cancer. J Pain Symp Manage. 2015. 50: 758–767. Spronk I, Meijers MC, Heins MJ, et al. Availability and effectiveness of decision aids for supporting shared decision making in patients with advanced colorectal and lung cancer: results from a systematic review. Eur J Cancer Care. 2019. 28: e13079. 23 Breast Cancer M. Tish Knobf and Youri Hwang Introduction There are > 3 million breast cancer survivors (BCSs), and the estimates for new cancer cases in women in the United States in 2019 are 268,600 cases of invasive cancer and 48,100 of ductal carcinoma in situ (DCIS).1 Nearly 80% of breast cancers are diagnosed in women > 50 years, and this age group accounts for the majority of deaths. A significant disparity persists between white and black women, with a 40% higher death rate among black women, and this disparity is greatest in young black women (< 50 years of age) compared to white women.1 A leading contributor to the disparate mortality rate is the higher incidence of triple-negative breast cancer (TNBC), especially in younger black women. The lack of estrogen receptors and progesterone receptors and human epidermal growth factor 2 (HER2) overexpression define TNBC, which accounts for 15–20% of new cases but is associated with limited treatment options and more unfavorable tumor characteristics, resulting in shorter survival and higher mortality. Beyond TNBC, higher mortality in black women is associated with multiple factors, such as social determinants of health, obesity, poor lifestyle behaviors, comorbidities, and access to quality screening and care. Diagnosis and management of breast cancer are dynamic in response to advances in understanding the biology and the precision health approach to treatment based on genetics and unique biologic characteristics. Widespread genetic risk testing, routine use of treatment response predictor and prognostic tests, increased use of magnetic resonance imaging (MRI), the dramatic rise in prophylactic mastectomy, advances in reconstructive surgery, treatment options for TNBC, changes in the surgical management of the axilla, and a greater role for neoadjuvant therapy are salient examples of the dynamic changes that contribute to the complexity of the experience, the decision-making process, and psychosocial adjustment. The purpose of this chapter is to describe treatment choices for breast cancer and the psychological responses across the trajectory of breast cancer from the definitive diagnosis through disease-free survivorship or end of life. Psychological Responses across the Breast Cancer Trajectory The trajectory of breast cancer begins with diagnosis and progresses through treatment and into survivorship (long-term survival or a shortened survival due to a recurrence, second cancer, or comorbid illness). There have been four distinct types of psychological distress associated with the breast cancer trajectory: chronic, delayed, recovered, and resilient.2 Based on the literature and clinical experience, these four trajectories can easily be identified with patient exemplars. A chronic trajectory might represent a younger woman who experiences treatment-induced menopause, cognitive changes, vasomotor symptoms with associated sleep alterations, and vaginal atrophy leading to sexual problems while managing competing life demands of school-age children, a marital relationship, and midcareer employment. The recovery trajectory represents the majority of women who experience some level of physical and psychological symptoms of distress during treatment but by one year later have improved significantly and successfully integrated the experience into everyday life.3 The delayed trajectory is similar to the recovery one, except that some additional traumatic event or complication occurs that creates additional stress, delaying the recovery process. The resilient type could describe a midlife or older woman who experiences minimal physical symptoms of distress during treatment, has strong social support, reports being well informed of what to expect, has effective communication with her providers, and experiences minimal life disruption. A middle-range theory of “Carrying On” has been proposed to explain how women respond and behave across the trajectory (Figure 23.1).4 There is a linear progression from the first stage of Being Focused, but for many women, stages 2 to 4 may represent a recursive relationship rather than a linear process. Being Focused is the stage of entry into the oncology world, characterized by a learning curve for information, terminology, providers, and the scope of cancer treatment while trying to manage everyday life. Dealing with Uncertainty is strongly influenced by information adequacy, effective relational communication with providers, symptom assessment and management, and explanation of symptom attribution. The better prepared a woman is for the experience, the more the level of uncertainty and associated psychological distress will be minimized. Developing Awareness generally occurs after treatment and women begin to explore and process the meaning of the diagnosis and the experience of treatment and treatment sequalae, which may include persistent physical and psychological symptom distress. They are also faced with the challenge of integrating the experience into their lives and may be opposed to the common 170 SECTION V Psychological Issues Related to Site of Cancer Stage Being Focused Dealing with Uncertainty Developing Awareness Balancing Culture Experience Crisis Response Early Adaptation Experiencing Treatment Side Effects Beginning to Understand Scope of Experience Transition to Survivorship Behaviors Learning Adapting to Treatment Managing Everyday Life Getting Information Communicating with Providers Relying on Self Exploring Meaning Living-Managing Symptoms Integrating into One’s Life Being Wary Struggling with the System Keeping Healthy Finding Support Figure 23.1. “Carrying on”: a middle-range theory of patient responses across the cancer trajectory. message of returning to normal or conceptualizing life as a new normal in the context of their distress, whether it be physical or psychosocial. The final stage is Balancing, which represents the transition work into survivorship, a time when there are less frequent oncology provider visits, lower levels of social support, an unclear role of primary care provider and/or specialist (e.g., gynecologist), and inconsistent access to survivorship resources. Keeping healthy is an important component of Balancing,4 and lifestyle behaviors, specifically routine physical activity, have been identified as an intervention to reduce psychological distress.5 It is critical to identify the determinants of psychological distress and how these determinants or risk factors affect how a woman processes the experience in the context of moving through the trajectory.2–4 Risk factors and predictors of psychological distress for women newly diagnosed with breast cancer include younger age, pre-existing psychological problems, presence of comorbid illness, chemotherapy treatment, moderate to severe physical symptom distress, limited available social support, non-Caucasian ethnicity, lower socioeconomic status, poorer functional performance, and breast cancer recurrence.3,5 Factors such as interpersonal characteristics, preferences for information, status of family and partner relationships, sexual orientation, role of spirituality in one’s life, and effectiveness of patient- provider communication can positively or negatively influence emotional well-being and psychosocial adjustment. Factors Contributing to Psychological Distress in Younger Women. Young women (18 to 49 years of age) account for 20% to 30% of BCSs. While many BCSs experience persistent symptoms such as anxiety, depressive mood, impaired cognitive functioning, pain, fatigue, fear of recurrence, and sleep disturbance after completion of curative intent primary therapy (e.g., surgery, chemotherapy, radiotherapy), younger BCSs report greater psychological symptom distress and poorer quality of life compared to their older counterparts.6 Unique to young BCSs is the developmental stage in life, which may include being single, being married with young children, being in early or midcareer employment, having a heightened fear of recurrence, and experiencing alterations in sexual function due to premature induced menopause, all of which contribute to higher levels of psychological distress.7 Young BCSs are at increased risk of treatment-induced premature menopause, leading to loss of fertility. Regardless of their plan about future pregnancies, young BCSs consider loss of fertility as loss of options to have a child in the future.8 Women who have not completed their families and who are younger than 40 years of age tend to have concerns about potential infertility after treatment.9 Childless women are particularly vulnerable to intrusive thoughts and likely to use avoidance coping.10 The emotional distress from loss of fertility may remain throughout their lifetime.7 Impaired body image is also commonly reported, but young BCSs have increased difficulty adjusting to their physical changes compared to older BCSs.11 Poorer body image in young BCSs is associated with type of surgery and adjuvant therapy (chemotherapy, endocrine, radiation), resulting in physical and psychological distress and alterations in sexuality.12 Up to 50% of young BCSs report sexual dysfunction,8 which can negatively impact the partner relationship as well as overall emotional well-being. Breast cancer survivorship appears unique for young women. Concerns regarding fertility, body image, and sexual functioning adversely influence psychological well-being, and are not always sufficiently addressed by providers. It is critical to identify young BCSs who are at risk of persistent psychological distress and the underlying causes in order to appropriately intervene. CHAPTER 23 Breast Cancer Primary Management of the Breast For the majority of women, initial decision-making relates to the primary treatment of the breast, with either breast conservation surgery (local tumor removal) with radiation therapy, or mastectomy with or without reconstruction. For selected patients, this initial decision-making may also include consideration of contralateral prophylactic mastectomy.13 These surgical options are presented for women with invasive cancer as well as DCIS. For a subset of patients, such as those with TNBC, HER2-positive breast cancer, or high-risk luminal B subtype, initial decision-making will include neoadjuvant systemic therapy as an option.14 Breast Conservation Surgery with Radiotherapy There are three decades of clinical trials that have established the survival equivalency of breast conservation surgery/radiotherapy (BCS/RT) to mastectomy. A shared decision-making model between the patient and provider is strongly recommended to evaluate risks, benefits, cosmetic outcomes, and patient preferences. The majority of women are eligible for BCS/RT with few contraindications, specifically previous thoracic radiation, pregnancy, connective tissue disease, unfavorable tumor characteristics (e.g., large tumors), or diffuse disease. Conventional-fraction whole breast irradiation (WBI) delivers 40–50 Gy followed by a boost to the tumor bed.15 Higher-fraction WBI (or accelerated radiation) delivers 40–42.5 Gy and utilizes higher fractions over a period of 1–3 weeks compared to 5–6 weeks for conventional-fraction WBI. The decision should consider the individual patient factors, such as tumor characteristics, stage, grade, age, and patient preferences.15 Fatigue is the most common side effect of radiation, regardless of the delivery method. Skin changes, sensation changes, and breast swelling follow a similar trajectory to the symptom of fatigue, increasing in frequency and severity by midtreatment through the end of radiotherapy, with gradual improvement at 3 and 6 months after therapy is complete.16 Black women report more frequent and more severe skin reactions, which are associated with pain, psychological distress, alterations in body image, and interference with daily function.17 Providers need to increase their skills in assessment and monitoring of skin changes in patients of color. While cosmesis is generally rated as good to excellent for the majority of women who receive breast irradiation, skin reactions, asymmetry associated with incision-site reactions, long-term hypopigmentation, and fibrosis can influence a woman’s psychological response. Management of the Axilla The surgical approach to axillary lymph nodes has shifted over the past decade, with sentinel lymph node (SLN) biopsy recommended as the initial procedure.13 For patients with negative or one to two positive sentinel lymph nodes, there is no advantage in clinical outcomes to following with an axillary lymph node dissection. However, axillary lymph node dissection (ALND) is recommended for patients with two or more positive sentinel lymph nodes because of prognostic implications of the extent of tumor burden in the axilla and to inform treatment choices. The initial approach with SLN biopsy has significantly decreased the surgical morbidity (sensory changes, functional limitations, infection risk, and risk of lymphedema). Lymphedema occurs in approximately 25%–30% of women with ALND compared to 4%–5% with SLN biopsy. Lymphedema can be managed but not cured and is associated with psychological distress, functional limitations, body image concerns, alterations in clothing, pain, and higher risk of infection, and it significantly impacts all domains of quality of life (QOL).18 Mastectomy and Reconstructive Surgery The goal in the initial decision-making phase is to offer the option of reconstructive surgery if a woman is considering mastectomy as a treatment choice. There are many factors supporting this goal: influence of surgical procedure for breast removal on reconstruction outcomes, indication for postmastectomy radiotherapy, patient preference, patient characteristics that may influence feasibility and choice of reconstructive procedures, and patient expectations.19 Breast reconstructive choices include placement of an implant or use of autologous tissue to create the breast mound. Implant reconstruction is a common reconstructive choice among women as no donor site is needed for tissue, and there is a relatively quick recovery.19 For women desiring implant reconstruction, a common approach is placement of a tissue expander, followed by placement of a permanent implant. The advantage of using a tissue expander prior to permanent implant placement is to allow the muscle and skin to stretch over time, creating a better pocket for the permanent implant. The disadvantages of the saline tissue expander are multiple visits for injections for the expander over a protracted time period and discomfort or pain associated with expansion procedures. Advances in implant reconstruction include carbon dioxide–filled tissue expanders, which allow for gradual expansion, minimizing or eliminating the disadvantages of the saline protocols.20 Complications of implant reconstruction include extrusion, implant failure, and capsular contraction, the latter being most common and associated with additional procedures and distress for the patient. There have been major advances in the field of onco-plastic surgery for breast cancer with autologous tissue reconstruction. An increase in expertise and use of microvascular tissue transfer with a transverse rectus abdominus myocutaneous (TRAM) flap (free, or muscle sparing), deep inferior epigastric artery perfuse (DIEP) flap, superior inferior epigastric artery (SIEP) flap, and non-abdomen- based options have changed the reconstructive options and outcomes.20 Advantages of autologous reconstruction include a more natural-looking breast, a long-lasting procedure, and an option for women with larger breasts. Disadvantages include a longer hospital stay, risk of infection, wound dehiscence, flap necrosis (at donor and recipient site), prolonged recovery, and donor site morbidity.19 Women who are offered reconstruction and elect to undergo the procedure tend to be white, younger, higher socioeconomic status, and concerned about body image. There is a disparity in reconstructive surgery between black and white women, which is partially explained by educational level, complexity of decision-making process, and financial/insurance issues. Patient outcomes that have been investigated following breast surgery include body image, quality of life, cosmesis, and psychosocial functioning. For body image (see Chapter 39), there appear to be small differences among women’s evaluation of outcome, with slightly better body image for women who had breast conservation with radiotherapy or reconstruction compared to mastectomy. 171 172 SECTION V Psychological Issues Related to Site of Cancer Musculoskeletal Upper Extremity Problems after Breast Cancer Treatment. Musculoskeletal problems after primary therapy for breast cancer occur in 25%–30% of women, resulting in physical and psychological distress and interference with everyday activities. The common upper extremity problems include decreased range of motion (ROM), reduced joint mobility, flexion and abduction impairments, decreased muscle strength, weakness, stiffness, tightness, and pain and are associated with ALND (6%–31%) and SLND (3%– 24%), and radiation increases the risk of upper extremity dysfunction.21 Upper extremity problems may persist for years and impact activities of daily living, leisure-time and work activities, and quality of life. Assessment of the arm and shoulder and impact on everyday function is recommended over the first year after treatment. Upper extremity problems will not usually resolve without treatment, and patients should be referred to an orthopedic provider and/or physical therapist for treatment. Prophylactic Mastectomy Decision-making for women who are genetically predisposed to breast cancer is complex (see Chapter 13). Bilateral prophylactic mastectomy (BPM) is an option for women who are gene carriers and have a strong family history, high-risk histologic features, and/ or surveillance issues.22 While BPM significantly reduces the risk of future breast cancer (> 90%), it is extensive surgery with a prolonged recovery. Yet, women report being satisfied with the decision and having lower psychological distress (e.g., anxiety, fear), and many rate body image favorably despite sensory changes and mixed effects on sexuality and relationships.22 Contralateral prophylactic mastectomy (CPM) is an option for the noncancerous breast, often chosen by women who elect mastectomy rather than BCS/RT for the affected breast.13 There has been a dramatic increase in women with early-stage breast cancer choosing to have a CPM. A positive family history, younger age, worse tumor characteristics, fear of future breast cancer, the experience of multiple repeated imaging, heightened surveillance, and a desire for symmetry characterize those women who are more likely to choose contralateral prophylactic surgery.23 CPM is associated with a 90%–95% reduction in risk for a future breast cancer, but women need to be well informed about individual risk for optimal decision making and understand that CPM does not provide 100% protection and has no influence on risk of local recurrence in the affected breast.13,23 While many women report satisfaction with their decision, the type of informational preparation and actual experience following surgery have resulted in psychological distress related to body image, problems with implants, poor cosmetic outcomes, and alterations in sexuality. For women with implant reconstruction, some have not felt prepared for the feel of the implant, potential complications (i.e., failure, capsular contraction), pain and discomfort with tissue expander procedures, and the unnatural look of the implanted breast mound. Sexuality issues have focused on the loss of erotic nipple sensations and distress about not being informed of such changes. For women with autologous reconstructive surgery, many expressed feeling unprepared for the length of recovery, numbness, pain, scarring (two sites: recipient and donor), body image, and the need for emotional preparation and psychological support. Adjuvant and Neoadjuvant Therapy Indications and treatment for adjuvant and neoadjuvant therapy have dramatically evolved over the last two decades.14 There has been a trend of escalation and de-escalation in the field in an attempt to determine which subgroups of patients will benefit from additional or combined treatment and which patients will benefit from less therapy with potential for lower toxicity.24 Treatment decision making is highly complex and dependent on tumor pathology, tumor subtypes, axillary lymph node involvement, status of estrogen and progesterone receptors, HER2 status, gene profiling, and patient characteristics such as age, comorbid illnesses, perception of risks and benefits, and preferences. The subtype of TNBC limits treatment options, and most women with TNBC are offered neoadjuvant therapy with chemotherapy and/or targeted agents. As TNBC is considered the most immunogenic subtype of breast cancer, with higher programmed cell death-1 (PD-1) expression, this has led to exploration of therapy with immune checkpoint inhibitors, especially for women with residual disease following neoadjuvant treatment.25 Checkpoint inhibitors are associated with a broad spectrum of significant organ system immune-related adverse events,26 which adds to the complexity of the decision-making process for women with TNBC. The first known study exploring the psychological response to TNBC was conducted and women described TNBC as “an addendum” to breast cancer.27 The supporting themes addressed the impact of learning the prognostic implications of TNBC and uncertainty (due to limited treatment options), described as “flying without a net” and having a steep learning curve due to the volume of information and compressed decision-making process. In summary, the experience for women with TNBC was characterized by fear, uncertainty, insecurity, and emotional distress.27 Adjuvant and neoadjuvant therapies for non-TNBC include chemotherapy, endocrine therapy, and targeted therapies, which can be delivered as a single modality but more often are combined and/or delivered sequentially, dependent on the tumor pathology and characteristics.24,28 Chemotherapy agents in these regimens generally include anthracyclines, docetaxel, cyclophosphamide, carboplatin, and capecitabine. Anti-estrogen therapy with tamoxifen and aromatase inhibitors (AIs) are used for hormone-positive postmenopausal women and for hormone receptor–positive premenopausal women; tamoxifen or an aromatase inhibitor combined with ovarian suppression has become a therapy option,24 but the routine use of an aromatase inhibitor with ovarian suppression remains controversial based on current evidence.29 HER2-positive breast cancer occurs in approximately 20%–30% of patients. The initial anti-HER2 agent trastuzumab (Herceptin) has been shown to significantly improve disease-free and overall survival.30 The identification of novel anti-HER2 agents with different mechanisms of action has been a focus in the last decade and includes pertuzumab, trastuzumab-emtansine (TD-M1), lapatinib (reversible HER2 growth factor receptor kinase inhibitor), and neratinib (irreversible pan-HER2 inhibitor).24,30 Breast cancer survivors with HER2-positive disease at low risk for recurrence will be recommended nonanthracycline adjuvant therapy with trastuzumab, and those at higher risk will have adjuvant or neoadjuvant treatment recommended with an anthracycline or nonanthracycline multiple drug regimen combined with an anti-HER2 agent or agents.14,24,28,30 CHAPTER 23 Breast Cancer The duration of anti-HER2 agents continues to be studied for efficacy and research continues to identify the optimal therapy for subgroups of patients as there is considerable heterogeneity among HER2-positive tumors.30 Side Effects and Psychological Responses. Physical symptoms are frequently associated with psychological distress, especially if the patient did not feel prepared, the physical symptoms were rated as moderate to severe, and/or interventions were not timely or effective. Adjuvant chemotherapy regimens are associated with a varied physical symptom profile, including but not limited to fatigue, peripheral neuropathy, changes in memory and concentration, alopecia, premature menopause, weight gain, skin and nail changes, and alterations in sleep, most of which persist after therapy is completed. Both tamoxifen and AIs are associated with menopausal symptoms of hot flashes, night sweats, sleep alterations, vaginal discharge (tamoxifen), vaginal dryness, and dyspareunia (AIs). AI therapy is associated with moderate to severe musculoskeletal symptoms, resulting in one-half to two-thirds of women discontinuing therapy, compromising the survival benefit. The most common barrier to adherence is musculoskeletal symptoms, but there are several other contributing socioeconomic factors (e.g., younger age, ability to pay) and psychosocial variables, specifically poor patient-provider communication, psychological depressive symptoms, and lack of perceived efficacy.31 Two late side effects include bone loss related to premature menopause and AI therapy and cardiac toxicity related to anthracyclines, anti-HER2 agents, and emerging targeted agents.32,33 No evidence was found on the psychological responses to being informed of these potential long-term adverse effects, but women can be at risk for psychological distress when sequalae such as osteopenia, osteoporosis, fractures, and decreased cardiovascular function and heart failure occur. Patients need to be adequately prepared with understandable information for acute, persistent, and long-term effects of therapy,4 as feeling unprepared for what is going to happen leads to greater uncertainty, distress, and ineffective coping. Pharmacologic, psychologic, and biobehavioral interventions have been evaluated for symptom management. Several therapy side effects are reviewed in Section VI (Chapters 32–37). Physical activity, specifically moderate-level activity of 30 minutes most days of the week, is a behavioral intervention that has been shown to improve fatigue, depressive symptoms, anxiety, quality of life, cardiovascular fitness, joint pains, and sleep.34,35 As many breast cancer patients are medication averse, physical activity is an excellent evidence-based intervention that should be recommended during and after therapy to minimize physical and psychological symptom distress and improve function and well-being.5,34,35 Transition to Survivorship The end of treatment and the beginning of the transition into survivorship are associated with emotional distress; anxiety; uncertainty; diminished support from providers, the healthcare system, and family; and persistent physical symptoms.3 By the end of 1 year, QOL and psychological adjustment significantly improve for the majority of women. However, the psychological responses associated with ending therapy, the process of reintegrating into life, and addressing the expectation to “return to normal” have resulted in emotional highs and lows over the first year. The vulnerability associated with cancer and the side effects of cancer treatment make many women question the concept of normal, or even the suggested “new” normal, as they perceive themselves as forever changed by the experience. Many will embrace the pink ribbon breast cancer survivor image, but some women feel that the emotional concerns of uncertainty and fear of recurrence are contrary to a perception of being a survivor.3 How well a woman adjusts over the course of the first year can also depend on persistent physical symptoms, degree of symptom distress, informational adequacy to manage symptoms, and known risks for late effects, such as osteoporosis and cardiovascular disease. Younger age, limited social support, poor communication with providers, personal life stressors, prior or pre-existing psychological problems, and decreased ability to work or carry out desired activities can further challenge a woman’s psychological recovery. The American Society of Clinical Oncology provides a guideline for assessment of treatment effects, evidence-based recommendations for management (e.g., acupuncture, physical activity for musculoskeletal health), surveillance, health promotion, and care coordination.36 An important part of the guideline is health promotion to minimize risk of comorbid illness, breast cancer recurrence, or a new cancer. Healthy eating and physical activity are two key components to health promotion. As black women have a higher rate of obesity and sedentary behavior, interventions to address this population are critical and should be tailored to cultural and socioeconomic factors.37 Black breast cancer survivors are interested in staying healthy and avoiding comorbid illness (e.g., diabetes, cardiovascular disease), and culturally designed interventions are feasible and can improve healthy lifestyle behaviors.38 Metastatic Breast Cancer: Transition to Long-Term Therapy and Palliative Care The majority of women will have hormone-positive cancer (≥ 75%), and the rapidly evolving understanding of molecular mechanisms has led to expanded therapy options. Critical to management of metastatic breast cancer is to balance the potential treatment benefit with the toxicity profile to maximize QOL. The first line of treatment is endocrine therapy with sequential use of agents as patients progress.39 Following endocrine therapy, four nonendocrine drugs have been approved: the mTor inhibitor everolimus and three cyclin-dependent kinases (CDK 4/6), palbociclib, ribociclib, and abemaciclib.40 The side effect profile of endocrine therapy is mild to moderately distressing for the majority of patients with a reported overall good QOL. In contrast, the nonendocrine therapies have much worse toxicity profiles. Everolimus is associated with stomatitis, pneumonitis, and gastrointestinal toxicity, while the CDKs include dose-limiting neutropenia, and some women develop elevation in liver enzymes and QTc prolongation.40 While most toxicities can be managed with dose reduction or interruption, the effects of the adverse side effects on an individual’s quality of life are not yet fully known.40 While there is no question that these therapeutic advances have extended survival in women with metastatic disease, it is imperative that every patient participate in the ongoing assessment of the risk-benefit profile. Communication, support, and introducing palliative care early on are essential as women with metastatic breast cancer transition from sequential therapies and beyond (see Chapters 73 and 74). 173 174 SECTION V Psychological Issues Related to Site of Cancer Future Directions Heterogeneity characterizes not only the biology of breast cancer but also the women who are diagnosed. As we personalize treatment based on the biology of the cancer, we need to personalize our interventions to address determinants of psychological distress across the trajectory of the breast cancer continuum. Longer follow-up (> 1 year) for psychological responses to persistent and late effects of therapy is indicated for BCSs. Newer advances in the treatment of metastatic breast cancer have resulted in longer survival, and research is needed in this population related to psychological well- being, coping, goals of care, and decision making as one transitions across treatment options to end of life. REFERENCES 1. DeSantis CE, Ma J, Gaudet MM, Newman LA, Miller KD, Sauer AG, Jemal A, Siegel RL. Breast cancer statistics 2019. CA Cancer J Clin. 2019;69:438–451. 2. Lam WW, Shing YT, Bonanno GA, Mancini AD, Fielding R. Distress trajectories at the first year diagnosis of breast cancer in relation to 6 years survivorship. Psycho-Oncol. 2012;21:90–99. 3. Knobf MT. Clinical update: psychosocial responses in breast cancer survivors. Semin Oncol Nurs. 2011;27:e1–e14. 4. Knobf MT. Being prepared: essential to self-care and quality of life for the person with cancer. Clin J Oncol Nurs. 2013;17:255–261. 5. Syrowatka A, Motulsky A, Kurteva S, Hanley JA, Dixon WG, Meguerditchian AN, Tamblyn R. Predictors of distress in female breast cancer survivors: a systematic review. Breast Cancer Res Treat. 2017;165:229–245. 6. Champion VL, Wagner LI, Monahan PO, Daggy J, Smith L, Cohee A, Ziner KW, Haase JE, Miller KD, Pradhan K, Unverzagt FW, Cella D, Ansari B, Sledge GW. Comparison of younger and older breast cancer survivors and age-matched controls on specific and overall quality of life domains. Cancer. 2014;120(15):2237–2246. https://doi.org/10.1002/cncr.28737 7. Campbell-Enns H, Woodgate RL. The psychosocial experiences of women with breast cancer across the lifespan: a systematic review. Psycho-Oncol. 2017;26:1711–1721. 8. Ahmad S, Fergus K, McCarthy M. Psychosocial issues experienced by young women with breast cancer: the minority group with the majority of need. Curr Opin Support Palliat Care. 2015;9(3):271–278. 9. Peate M, Meiser B, Hickey M, Friedlander M. The fertility- related concerns, needs and preferences of younger women with breast cancer: a systematic review. Breast Cancer Res Treat. 2009;116(2): 215. https://doi.org/10.1007/s10549-009-0401-6 10. Gonçalves V, Quinn GP. Review of fertility preservation issues for young women with breast cancer. Human Fertil. 2016;19(3):152–165. 11. Male DA, Fergus KD, Cullen K. Sexual identity after breast cancer: sexuality, body image, and relationship repercussions. Curr Opin Support Palliat Care 2016;10(1):66–74. 12. Paterson C, Lengacher CA, Donovan KA, Kip KE, Tofthagen CS. Body image in younger breast cancer survivors: a systematic review. Cancer Nurs. 2016;39(1):E39–E58. 13. Newman LA. Decision-making in the surgical management of invasive breast cancer. Part1: lumpectomy, mastectomy and contralateral mastectomy. Oncology. 2017;92:359–368. 14. Caparica R, Brandao M, Piccart M. Systemic treatment of patients with early breast cancer: recent updates and state of the art. Breast. 2019;48S1:S7–S20. 15. Recht A, McArthur H, Solin LJ, Tendulkar R, Whitley A, Giuliano A. Contemporary guidelines in whole-breast irradiation: an alternative perspective. Int J Radiat Oncol Biol Phys. 2019;104(3):567–573. 16. Knobf MT, Sun, Y. A longitudinal study of symptoms and self-care activities in women treated with primary radiotherapy for breast cancer. Canc Nurs. 2005;28:210–218. 17. Schnur JB, Ouelette SC, DiLorenzo TA, Green S, Montgomery GH. A qualitative analysis of acute skin toxicity among breast cancer radiotherapy patients. Psycho-Oncology. 2010;20:260–268. 18. Paskett ED, Dean JA, Oliveri JM, Harrop JP. Cancer- related lymphedema risk factors, diagnosis, treatment and impact: a review. J Clin Oncol. 2012;30:3726–3733. 19. Dellacroce FJ, Wolfe ET. Breast reconstruction. Surg Clin N Am. 2013;93:445–454. 20. O’Halloran N, Potter S, Kerin M, Lowery A. Recent advances and future directions in postmastectomy breast reconstruction. Clin Breast Cancer. 2018;4:e571–e585. 21. Hidding JT, Beurskens CH, vander Wees PJ, Laarhover WM, Nijhuis-vander Sanden MW. Treatment related impairments in arm and shoulder in patients with breast cancer: a systematic review. PLos ONE 2014;9(5) e96748. 22. Razdan SN, Patel V, Jewell S, McCarthy CM. Quality of life among patients after bilateral prophylactic mastectomy: a systematic review of patient reported outcomes. Qual Life Res. 2016;25(6):1409–1421. 23. Ager B, Butow P, Jansen J, Phillips K, Porter D. Contralateral prophylactic mastectomy (CPM): a systematic review of patient reported factors and psychological predictors influencing choice and satisfaction. Breast. 2016:107–120. 24. Ponde NF, Zardavas D, Piccart M. Progress for adjuvant systemic therapy for breast cancer. Nat Rev Clin Oncol. 2019;16:27–44. 25. Kwa MJ, Adams S. Checkpoint inhibitors in triple-negative breast cancer (TNBC): where to go from here. Cancer. 2018;124:2086–2103. 26. Davies M. Acute and long-term adverse events associated with checkpoint blockade. Semin Oncol Nurs. 2019;35:150926. doi:org/ 10.106.soncn.2019.08.005 27. Turkman Y, Kennedy H, Harris L, Knobf MT. “An addendum to breast cancer”: the triple negative experience. Support Care Cancer. 2016;24:3715–3721. 28. Jasra S, Anampa J. Anthracycline use for early stage breast cancer in the modern era: a review. Curr Treat Options Oncol. 2018;19(6):30. doi.org/10.1007/s11864-018-0547-8 29. Chlebowski RT, Pan K, Col NF. Ovarian suppression in combination endocrine adjuvant therapy in pre-menopausal women in early breast cancer. Breast Cancer Res Treat 2017;161:185–190. 30. Pernas S, Barroso-Sousa R, Tolaney SM. Optimal treatment of early stage HER2-positive breast cancer. Cancer 2018;124:4455–4466. 31. Paranjpe R, John G, Trivedi M, Abughosh S. Identifying adherence barriers to oral endocrine therapy among breast cancer survivors. Breast Cancer Res Treat. 2019;174:297–305. 32. Aydiner A. Meta-analysis of breast cancer outcome and toxicity in adjuvant trials of aromatase inhibitors in postmenopausal women. Breast. 2013;22:121–129. 33. Rozner RN, Frishman WH. Cardiovascular effects of chemotherapy used in the treatment of breast cancer. Cardiol Rev. 2019;27(2):87–96. 34. Park S, Knobf MT, Jeon S. Endocrine therapy-related symptoms and quality of life in female cancer survivors in the Yale Fitness Intervention Trial. J Nurs Scholarship. 2019;513:317–325. 35. Palesh O, Scheiber C, Kelser S, Mustian K, Koopman C, Schapira L. Management of side effects during and post-treatment breast cancer survivors. Breast J. 2018;24:167–175. CHAPTER 23 Breast Cancer 36. Runowicz CD, Leach CR, Henry NL, Henry KS, Mackey HT, Cowens-Alvarado RL, et al. American Cancer Society/American Society Clinical Oncology breast cancer survivorship care guideline. J Clin Oncol. 2016;34(6):611–635. 37. Coughlin SS, Yoo W, Whitehead M, Smith SA. Advancing breast cancer survivorship among African American women. Breast Cancer Res Treat. 2015;153:252–261. 38. Knobf MT, Erdos D, Jeon S. Healthy sisters: A feasibility study of health behavior intervention for women of color breast cancer survivors. J Psychosocial Oncol. 2018. doi.org/10.1080/ 07347332.2018.14600004 39. Chalasani P. Optimizing quality of life in patients with hormone receptor-positive metastatic breast cancer: treatment options and considerations. Oncology. 2017;93:143–156. 40. Cazzaniga ME, Danesi R, Girmenia C, Invernizzi P, Elvevi A, Uguccioni M. Management of toxicities associated with targeted therapies for HR-positive metastatic breast cancer: a multidisciplinary approach is the key to success. Breast Cancer Res Treat. 2019;176:483–494. 175 24 Colorectal Cancer Anne Miles and Claudia Redeker Introduction Colorectal cancer (CRC) is the third most common cancer and second most common cause of cancer death worldwide, with 1.8 million cases diagnosed and 881,000 deaths in 2018. Survival rates are strongly linked to stage at diagnosis, with 90% surviving localized disease (Stage I) but only 10% surviving once the disease has spread to distal organs (Stage IV). Although global CRC incidence and mortality rates are falling, different patterns are observed between countries. Increases in incidence are seen in countries adopting “Westernized” lifestyles, due to the causal role of lifestyle factors such as obesity and lack of exercise in CRC onset, while declines in mortality are observed in high-resource countries able to offer more effective treatment and early detection initiatives, such as screening programs.1 resonance imaging (MRI), are noisy and involve full-body immersion into a tube, and can induce anxiety and claustrophobia in a substantial proportion of patients. Particular sectors of the population may find scans more difficult than others. For example, high distress and the presence of comorbidities have both been associated with finding whole-body MRI more challenging, independent of other demographic and clinical variables, among patients with suspected CRC.3 In a retrospective study asking CRC patients in Canada what their specific needs had been during the diagnostic phase, 31.6% reported informational and 20.3% reported emotional needs. While the majority felt their needs had been met at the time, 77.9% reported that they had not been offered help coping with their anxiety, highlighting the need for increased emotional support during the diagnostic phase.4 Diagnostic Pathways and Diagnostic Delay Obtaining a Diagnosis Psychological issues arise from the moment CRC is suspected, through treatment, to living with or beyond cancer. Patients with suspected cancer have to undergo medical investigations and attend medical appointments, while facing the threat of a serious illness. Research into distress during the diagnostic phase has shown that between 33% and 60% of patients report clinical levels of anxiety before getting a diagnosis (defined as anxiety high enough to be categorized as an anxiety disorder using psychiatric assessment tools). These levels are equivalent to those observed in patients with a confirmed diagnosis. The majority of studies report that anxiety levels remain the same or increase in patients ultimately diagnosed with cancer, while anxiety typically reduces in those with a noncancer diagnosis.2 Similar effects have been observed for quality of life (QoL), with patients with suspected cancer reporting the equivalent or poorer QoL as patients with a confirmed diagnosis.2 The process of undergoing investigations for suspected cancer presents numerous challenges. In addition to the anxiety associated with waiting for test results, patients with suspected CRC may need to undergo bowel preparations or have injections, some of which may involve a radioactive ligand (such as for positron emission tomography [PET]/computed tomography [CT]), promoting concerns about radiation risk. Scans, such as whole-body magnetic There are a number of different pathways to a diagnosis of CRC. The most common include screening, planned pathways for the investigation of symptoms of suspected cancer, routine referral for the investigation of symptoms, and emergency presentation (defined as a cancer diagnosis within 28 days of either attending the accident and emergency department of a hospital or an emergency hospital admission). Screening and planned-care pathways (e.g., urgent referral to a specialist following cancer “alarm” signals) are designed to diagnose CRC sooner and improve survival rates. The likelihood of experiencing these different pathways, though, varies internationally; for example, organized screening programs for CRC are seen in the majority of European countries but are largely absent in most countries in Central and South America, the Middle East, and Africa. In addition, where offered, uptake rates vary widely from 68.2% in the Netherlands, for instance, to 16% in parts of Canada.5 Little work has examined the psychological consequences of the different pathways patients may take to a diagnosis. People who have cancer diagnosed at screening are more likely to have earlier-stage disease, requiring simpler treatment, such as surgery alone. A quantitative study in Scotland found that CRC patients diagnosed via fecal occult blood test (FOBt) screening reported better QoL between 3.5 and 12 years postdiagnosis than people diagnosed following a negative FOBt, or than people who were diagnosed when living in an CHAPTER 24 Colorectal Cancer area that did not offer screening at the time,6 highlighting superior patient-reported outcomes in screen-detected patients. Perceived quality of care following treatment for CRC in England is also highest among patients diagnosed via screening and worse among patients diagnosed following emergency presentation.7 While patients diagnosed via emergency presentation have longer surgeries, longer admissions, and more readmissions, which may negatively influence their experience of care, they give more negative evaluations of issues such as staff explanations before and after surgery and of not being involved as much as they wanted to be in treatment decisions.7 Hence, patient-reported outcomes as well as mortality rates can be improved if more CRC patients are diagnosed via screening. The main reason for the introduction of diagnostic pathways, such as screening and standardized cancer patient pathways for symptoms of suspected cancer, is to improve cancer outcomes by diagnosing CRC at an earlier stage and reducing the time to treatment. Patients who wait longer for a diagnosis typically undergo a higher number of medical consultations and investigations. Patients who report a higher number of visits prior to a diagnosis, or a longer period of time between help seeking and confirmation of their diagnosis, report lower satisfaction with their care. For example, patients with anal cancer were more likely to be unsatisfied with their care if they believed there had been a delay in their diagnosis, but satisfaction levels were much lower if patients believed the cause of the delay was due to the medical profession rather than their own fault for failing to seek prompt medical attention.8 Some delay in diagnosis can be attributed to cancer site and the nonspecific nature of cancer symptoms, making some cancers harder to diagnose than others. However, delay can also arise from inefficiencies or errors in care. Increased diagnostic delay has been associated with misdiagnoses, such as attributing the symptoms to a disease other than CRC, failure to examine the patient, and negative or false- negative results.9 CRC false-negative rates of 8% were reported in a district general hospital in the UK across the three modalities of double-contrast barium enema, colonoscopy, and CT colonography, with the highest false-negative rates observed for barium enema and the lowest for colonoscopy,10 suggesting that missed cancers are not uncommon. Research into the psychological consequences of having a cancer missed, though, is scant. The study mentioned earlier, comparing psychological outcomes by method of CRC detection in Scotland, found no evidence of adverse psychological outcomes among people who had an “interval” cancer (i.e., a cancer diagnosis following a negative or clear screening outcome), as compared with people diagnosed with CRC in an area not offering screening at the time, on measures of perceived diagnostic delay, QoL, and depression.6 However, participants were surveyed between 3.5 and 12 years postdiagnosis. Thus, having a cancer missed had probably not proven fatal, potentially attenuating any adverse effects. The same data showed that CRC survivors who believed their cancer could have been diagnosed sooner reported greater cancer-related distress and were more likely to be classified as having posttraumatic stress disorder (PTSD). While part of the relationship between perceived delay and distress was explained by QoL, disease stage at diagnosis and treatment received did not explain the association.11 The reasons for higher distress among those who believed their cancer could have been diagnosed sooner remain unclear, but could relate to factors associated with delay or the way in which the cancer was diagnosed. Criteria for PTSD (in the Diagnostic and Statistical Manual of Mental Disorders, fifth edition [DSM-5]) specify that “Medical incidents that qualify as traumatic events involve sudden, catastrophic events.”12 In the context of cancer, trauma could arise from diagnostic pathways such as emergency presentation. Given the obvious distress associated with undergoing investigations for cancer, research has examined whether a more rapid diagnosis is associated with enhanced psychological well-being and patient satisfaction with care. However, a more rapid diagnosis means a faster transition from being healthy to being a patient; the diagnosis may be more shocking and hence harder to accept or adjust to. Again, there is little research specifically on patients with suspected CRC, but in a review of rapid diagnostic pathways (one-or two-stop shops), Brocken et al. found that anxiety tends to reduce among people with benign outcomes but is sustained or increased among those diagnosed with cancer.2 A more rapid diagnosis therefore reduces the duration of anxiety for those with benign outcomes but did not appear to be either beneficial or harmful for people ultimately diagnosed with cancer. Treatment Treatment for CRC is strongly linked to diagnostic stage. Surgery alone is typically recommended for patients with Stage I CRC. Patients with Stage III or high-risk Stage II colon cancer (e.g., those with large tumors) are offered adjuvant (postsurgical) chemotherapy, while patients with rectal cancer are typically offered neoadjuvant (presurgical) radiotherapy with or without chemotherapy to reduce tumor size prior to surgery, as well as to reduce the risk of local recurrence. Such treatment for rectal cancer may also be followed by further with adjuvant therapy to stop the emergence of distal disease. Treatment for Stage IV disease can still have curative intent with surgical resection of distant metastases, but palliative chemotherapy has also led to reductions in tumor size, making surgical resection possible at a later date. Shared Decision Making Shared decision making is central to patient-centered care, whereby patients take an active role in decisions about their treatment. For patients with either Stage II or III CRC, a key decision is whether to have adjuvant chemotherapy, and if so, which chemotherapy drugs to take. The chemotherapy offered is typically fluorouracil or capecitabine, given on its own or in combination with oxaliplatin. In patients with Stage II CRC, the risk of cancer recurrence is around 20–40%, but chemotherapy can only prevent a recurrence in 3–7% of patients.13 Because some of the side effects of chemotherapy can be permanent or even life-threatening, the balance of benefits and harms is marginal in this patient group and it is important that the decision be based on patient preferences. Eighty-five percent of Stage III CRCs relapse within 5 years. Chemotherapy can prevent relapse in up to 25% of patients and is therefore offered routinely. However, the addition of oxaliplatin carries the risk of permanent neuropathy and only offers small additional protection against recurrence, of around 5%.14 Recent research has shown that reducing the duration of adjuvant chemotherapy from 6 to 3 months can halve the risk of experiencing permanent neuropathy without adversely affecting survival rates,14 and 177 178 SECTION V Psychological Issues Related to Site of Cancer that both therapy duration and therapy composition should be discussed and agreed upon with patients. Decision Support Current levels of patient involvement in decision making are suboptimal. For example, in England, patient perceptions of their involvement in cancer treatment decisions are currently assessed via the Cancer Patient Experience Survey (CPES). The CPES is conducted by the National Health Service (NHS) England to monitor cancer care and drive forward improvements. Analysis of patient responses to the question “Were you as involved in decisions about which treatment you would have as you wanted?” showed that rectal and anal cancer patients were less likely to report positive experiences of involvement in treatment decisions than colon cancer patients. People who had had colon cancer reported more positive responses than people with most other cancer types.15 However, people who responded that there was only one type of treatment suitable for them were excluded from the analysis. People are often unaware that they have a choice of therapy, so the proportion of positive responses to this question may be inflated. Some rectal cancer patients may be given a choice between a permanent ostomy (a surgically created opening in the body for the discharge of bodily waste) and sphincter-sparing surgery, provided that the probabilities of survival and recurrence are similar. In patients who choose, or need, an ostomy, research has shown that the postoperative period is smoother and emotionally less distressing when it follows adequate preoperative preparation, and when the placement of the stoma (the opening in the abdomen) has taken into account skinfolds and patient preference (e.g., with regard to clothing).16 Even when appropriate discussions between patient and provider take place, involving patients in shared decision making presents a number of challenges. While patients with CRC want information about their cancer, particularly in relation to their prognosis and treatment options, there are difficulties in effectively communicating information about likelihoods and potential outcomes of treatment, and adequately supporting patients in making decisions about their treatment. Patient decision aids (PDAs) are specifically designed to assist patients, for example, by presenting likelihood information in multiple formats (e.g., as absolute risk, and in both numeric and graphic formats) and including a values clarification exercise to help patients work out what is more important to them—for example, maximizing their chances of avoiding a recurrence or accepting a small increased risk of recurrence to avoid long-term treatment side effects. Although evidence suggests that decision aids can be of benefit in increasing people’s knowledge, enhancing people’s accuracy about the likelihood of different outcomes and helping people feel both better about the information they receive and clearer about what is important to them, few decision aids have been developed to help people make decisions around CRC treatment.17 As people are more likely to survive CRC, the difficulties they face living with the long-term effects of the decisions they have made, and the treatments they have had, become more important. Living with and beyond Colorectal Cancer CRC patients and survivors often report emotional difficulties; problems with bowel, urinary, or sexual function; and issues with body image following major surgery, either for resection of disease, the placement of a stoma, or both. Such problems can impact both the patient’s social life and ability to work. The likelihood of experiencing such issues is higher among people diagnosed with later-stage disease and those receiving chemotherapy or radiotherapy. Emotional Difficulties and Quality of Life Anxiety, depression, and PTSD are frequently comorbid conditions among cancer survivors. A meta-analyses of the prevalence of anxiety and depression among people 2 or more years postdiagnosis found that while rates of depression were equivalent to people without cancer (11.6% vs. 10%), rates of anxiety were higher (17.9% vs. 13.9%).18 Rates of PTSD are also more common among cancer survivors than people with no history of the disease, although prevalence is typically lower than that of depression and anxiety, with a reported 6.4% point prevalence and lifetime risk of 12.6%.19 The proportion of patients reporting emotional difficulties is usually highest shortly after diagnosis and declines over time. However, emotional trajectories vary across patients. Dunn et al. conducted a longitudinal, prospective study of distress among CRC patients between 5 months and 5 years postdiagnosis using the Brief Symptom Inventory-18 (a measure combining anxiety, depression, and somatization). They observed four patterns: consistently low distress, observed in 19.4% of patients; medium-level distress (going from “case” to “noncase”) in 29.4% of patients; medium increase (going from noncase to case) in 38.5% of patients; and high distress (remaining at case level over time) in 12.5% of patients. The odds of being in one of the medium-or high-distress trajectories compared with the consistently low-distress group were higher for patients with Stage III or IV disease, after controlling for age, gender, educational level, and social support. While treatment type was a significant predictor in unadjusted analyses, it was not significant in the presence of other variables.20 Particular subgroups of the population show greater vulnerability to emotional problems postdiagnosis. A study conducted on CRC patients attending cancer clinics in Scotland found depression was more likely among women, younger people, and people with higher levels of deprivation.21 Higher prevalence of anxiety, depression, and symptoms of traumatic stress have also been reported among patients who had adjuvant radiotherapy or chemotherapy compared to those receiving surgery alone, although the impact of disease stage, independent of treatment received, was not examined.22 Among CRC survivors who had completed treatment for Stage II or Stage III disease, distress was higher among people who reported treatment-related side effects, such as peripheral neuropathy. QoL measures capture patients’ subjective assessment of physical, functional, psychological, and social well-being. Different QoL trajectories have also been observed in CRC survivors, followed up from 5 months to 5 years postdiagnosis.23 Using the Functional Assessment of Cancer Therapy-Colorectal (FACT-C), which has physical, functional, social/family, and emotional well-being and CRC-specific symptom subscales, Dunn et al. found four different QoL trajectories: constant high QoL, observed in 26.2% of patients; constant medium, observed in 47.1%; medium decrease, observed in 7.4% of patients, whereby patients reported a marked decrease in QoL 2 years postdiagnosis; and constant low QoL, reported by 19.2% of patients.20 Compared to the reference category of consistently CHAPTER 24 Colorectal Cancer high QoL, patients were more likely to be in the QoL trajectories of medium decrease or constant low if they had more advanced disease at diagnosis, although membership was not associated with treatment received. However, other studies show links between specific treatments or treatment outcomes on QoL. Adverse effects of radiotherapy on long-term bowel and sexual function have been reported in numerous studies, but perceived quality of care at the time of treatment (e.g., lack of treatment-related information, poor control of treatment-related side effects) also predicts subsequent QoL in CRC patients, showing that events experienced during treatment can have a lasting impact on both patients and their family caregivers.22 Treatment-Related Side Effects CRC patients undergoing radiotherapy or chemotherapy may experience a number of side effects, some of which persist once active treatment has finished. A population-based study in Ireland on symptom burden of CRC survivors 1 to 3 years postdiagnosis found the three most commonly reported symptoms were fatigue, insomnia, and flatulence, all of which were reported by over 20% of survivors.24 This study also found symptoms tended to co-occur, and certain symptoms were more common in patients who had rectal cancer or had a stoma. Clinically relevant fatigue (defined as worthy of further clinical attention) has been reported in a third of CRC patients attending a regional center in Scotland and is more likely among patients who have received radiotherapy or chemotherapy in the preceding 2 months.25 The chemotherapy drug oxaliplatin carries a risk of peripheral neuropathy (PN). Symptoms of PN include insensitivity to cold, numbness, pain, and trouble with balance. Such symptoms have been associated with higher rates of anxiety and depression and can affect people’s ability to sleep as well as conduct normal activities of daily living, including work. Estimates of long-term prevalence vary, but one study found that 29% of patients experienced significant neuropathy 3 years after treatment.14 Radiotherapy increases the risk of bowel and urinary problems. Bowel problems such as diarrhea can adversely affect QoL in CRC survivors both in the short and longer term,26 resulting in poorer QoL than among people who have never had cancer. However, incontinence following radiotherapy also depends on whether patients have had a stoma or not, which, while reducing fecal incontinence, confers problems of its own. Patients with Ostomies Rectal cancer patients’ concern about a permanent ostomy frequently supersedes all other considerations (see Box 24.1). Difficulties with self-care are common, with 63% of ostomates (people with an ostomy) reporting at least one self-care challenge.27 While there have been many appliance improvements (e.g., two-piece pouches, stoma plugs, flushable pouches) to help ostomates resume social and physical activities and enhance confidence, the continuing large number of ostomates who adjust poorly highlights the need for strategies to increase self-efficacy and utilizing appliance improvements to their full potential.27 There is some evidence that female ostomates fare worse than male ostomates. A systematic review found that in all studies included in the analysis, female ostomates showed significantly worse QoL in several domains (emotional, physical, and mental health) compared to male ostomates. Ostomies also have a greater impact on QoL in younger patients compared to older patients.28 Difficulties with sleep appear to be another concern for ostomy patients, especially for women. The main distress is related to pouch leaks and sleeping positions, with some ostomy patients reporting fatigue due to sleep deprivation. Furthermore, complications due to the ostomy such as parastomal hernia, parastomal abscess or fistula, retraction, stomal ischemia/ necrosis, stoma stenosis, prolapse, bleeding, small or large bowel obstruction, and dehydration from high ostomy output, with peristomal leakage and skin breakdown being the most common complications, further decrease ostomates’ QoL. Surprisingly, some ostomates delay treatment for complications for years, waiting too long, or do not contact a health professional at all. While ostomy complications occur most often within the first 5 years, the risk of developing a complication remains lifelong. Body Image and Sexual Functioning Sexuality and sexual dysfunction can play an essential role in the psychosocial health of CRC survivors, including those with an ostomy. Di Fabio et al. found that 76% of rectal cancer patients reported sexual dysfunction concerns.29 Most commonly for males, the presence of an ostomy is associated with higher erectile dysfunction and lower ejaculatory control. Genital pain and lack of lubrication are often reported by women. Unsurprisingly, reduced intercourse frequency is often the consequence. If unaddressed, these sexual difficulties persist or even worsen and correlate with reduced QoL, including body image, self-esteem, distress, and social and physical functioning. There are several known barriers that hinder the management of sexual dysfunction and satisfaction, such as embarrassment, lack of or inadequate information by the care provider before or after surgery, and the timing of interventions.29 Box 24.1. Common QoL Issues for Patients with Ostomies The creation of an ostomy is sometimes required in the surgical resection of low-lying rectal cancers. Permanent ostomies are associated with psychological, physical, and social difficulties. An individual’s adaptation and the ability to deal with an ostomy can affect quality of life (QoL). Common QoL issues for patients with permanent ostomies include: • Depression • Chronic anxiety • Social isolation • Sexuality • Body image and appearance • Self-management of ostomy — Leaks and spillage — Skin problems — Embarrassment due to odor, leakage, and noise • Dietary concerns • Constipation • Comorbidities • Disrupted sleep due to pouch leaks and sleeping position • Financial difficulties in paying for ostomy supplies • Interference with work and social activities • Physical activities • Travel difficulties • Clothing restrictions • Constant need to adjust to living with an ostomy 179 180 SECTION V Psychological Issues Related to Site of Cancer Most treatments for sexual dysfunction have focused on pharmacological options, with varying results. While some treatments are effective in reversing iatrogenic erectile dysfunction, adherence may be an issue. Treatments for women, such as topical testosterone application for low sexual desire, carry potential safety risks and thus are not an option. There is some evidence that psychological interventions can significantly improve sexual dysfunction in cancer survivors. Gender- specific approaches and timing of the intervention appear to be key components for CRC patients. Moreover, body image concerns may contribute to sexual dysfunction and recovery and ought to be addressed. Patients with ostomies are more likely to have negative feelings about their body appearance, which can lead to diminished intimacy and greater isolation. Intimacy concerns are not just related to patients; ostomies also affect spouses and other partners. Persson et al. found that spouses of CRC survivors with an ostomy had difficulty with the partner’s altered body, reported feeling distant from their partners because of the distress caused by the ostomy surgery, reported difficulty looking at the stoma, and struggled to hide feelings of disgust.30 Twenty-three percent of CRC survivors with an ostomy indicated that their partner responded negatively during the first sexual experience after ostomy surgery, and 30% stated that their partner reacted with much caution, fearing they would hurt the stoma. Overall, women described their husbands’ and partners’ support as central to their psychosocial adjustment to having an ostomy. frustrated at not being able to do what they used to do, and a quarter reported worrying about finances. A third of patients reported often feeling anxiety or worry, and fear of dying, with 71.4% finding uncertainty the most difficult thing to cope with. Poor coordination of care, lack of involvement of their general practitioner or primary care provider, and lack of access to support services were also reported. Overall experiences of having chronic cancer were worse among younger patients, patients not currently in a relationship, and those who had had cancer for longer. Despite the increase in studies investigating unmet needs among cancer patients, survivors (people who have had cancer but are currently cancer-free), and chronic cancer patients, interventions to address these needs have been more limited and the majority have failed to successfully address them.33 Future Directions People diagnosed with CRC face numerous challenges throughout the diagnostic, treatment, and posttreatment phases. Emotional difficulties, particularly anxiety and FCR, can persist for years. Treatments for CRC can result in problems with bowel, urinary, or sexual function and issues with body image following major surgery or the presence of a stoma. Such problems can impact both the patient’s social life and ability to work, and result in financial difficulties. Fatigue and difficulty sleeping remain common problems among patients with chronic cancer. Fear of Recurrence and Ability to Work Evidence, though limited, suggests that better QoL and greater Fear of cancer recurrence (FCR) is a common worry, with a third of levels of satisfaction with cancer care are reported among patients CRC survivors reporting concerns a median of 5 years postdiagnosis. with screen-detected disease. However, there are marked differences One Dutch study found no association between FCR in CRC sur- across countries in screening provision. A substantial proportion of patients living with and beyond vivors and disease stage or treatment received, but greater FCR was cancer report unmet needs around living with anxiety, uncertainty, associated with higher distress and poorer QoL.31 The impact of a cancer diagnosis on financial strain is and fear of dying, as well as the need for medical support and inforunderexplored, but research has shown that survivors of gastroin- mation. Interventions to reduce unmet needs in cancer patients have testinal cancers are more likely to be unemployed compared with had little success to date. Future research should aim to minimize the psychological and people with no history of cancer (48.8% vs. 33.4%).32 Reasons for unemployment given include physical limitations and cancer- physical consequences of a CRC diagnosis. Promoting earlier diagrelated symptoms. Inability to work can cause high levels of distress nosis, for example, via screening, is one of the best ways of achieving this. More effective interventions are required to help CRC patients in a substantial proportion of survivors. in the challenges they face before and after a cancer diagnosis, such Unmet Needs as the management of long-term side effects of cancer-related treatImprovements in cancer treatment mean more people live with ment such as fatigue, and emotional difficulties that may persist for cancer as a “chronic illness” (defined as people with active, ad- years. Greater patient involvement in decision making around treatvanced, or metastatic disease that cannot be cured but that can ments should be encouraged to ensure individuals decide how best be managed and is not considered end-of-life care). Recent re- to balance quantity with QoL, rather than maximizing survival at search into the symptoms and care needs of this group includes any cost. development of a questionnaire (the Chronic Cancer Experiences Questionnaire) and assessment of difficulties in relation to clinical REFERENCES services, self-management, and self-care; needs for independent 1. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. living, work, finances, and benefits; psychological experiences, supGlobal cancer statistics 2018: GLOBOCAN estimates of incidence port, and services; and patients’ experience of ongoing symptoms. and mortality worldwide for 36 cancers in 185 countries. CA Cancer This research found that the prevalence of cancer-specific symptoms J Clin. 2018;68(6):394–424. was high, with 80% reporting fatigue and 65.9% trouble sleeping. 2. Brocken P, Prins JB, Dekhuijzen PN, van der Heijden HF. The faster Approximately one-third of patients reported always or often feeling the better? A systematic review on distress in the diagnostic phase that there were limitations in their physical abilities, social activiof suspected cancer, and the influence of rapid diagnostic pathways. ties, and ability to engage in hobbies; a similar proportion reported Psychooncology. 2012;21(1):1–10. difficulties planning ahead, 45.2% reported always or often feeling CHAPTER 24 Colorectal Cancer 3. Evans REC, Taylor SA, Beare S, et al. Perceived patient burden and acceptability of whole body MRI for staging lung and colorectal cancer; comparison with standard staging investigations. Br J Radiol. 2018;91(1086):20170731. 4. Wiljer D, Walton T, Gilbert J, et al. Understanding the needs of colorectal cancer patients during the pre-diagnosis phase. J Cancer Educ. 2013;28(3):402–407. 5. Navarro M, Nicolas A, Ferrandez A, Lanas A. Colorectal cancer population screening programs worldwide in 2016: an update. World J Gastroenterol. 2017;23(20):3632–3642. 6. Miles A, McClements PL, Steele RJ, Redeker C, Sevdalis N, Wardle J. The psychological impact of a colorectal cancer diagnosis following a negative fecal occult blood test result. Cancer Epidemiol Biomarkers Prev. 2015;24(7):1032–1038. 7. Salika T, Abel GA, Mendonca SC, et al. Associations between diagnostic pathways and care experience in colorectal cancer: evidence from patient- reported data. Frontline Gastroenterol. 2018;9(3):241–248. 8. Chiu S, Joseph K, Ghosh S, Cornand RM, Schiller D. Reasons for delays in diagnosis of anal cancer and the effect on patient satisfaction. Can Fam Physician. 2015;61(11):e509–e516. 9. Mitchell E, Macdonald S, Campbell NC, Weller D, Macleod U. Influences on pre-hospital delay in the diagnosis of colorectal cancer: a systematic review. Br J Cancer. 2008;98(1):60–70. 10. Than M, Witherspoon J, Shami J, Patil P, Saklani A. Diagnostic miss rate for colorectal cancer: an audit. Ann Gastroenterol. 2015;28(1):94–98. 11. Miles A, McClements PL, Steele RJ, Redeker C, Sevdalis N, Wardle J. Perceived diagnostic delay and cancer-related distress: a cross- sectional study of patients with colorectal cancer. Psychooncology. 2017;26(1):29–36. 12. Kangas M. DSM-5 trauma and stress-related disorders: implications for screening for cancer-related stress. Front Psychiatry. 2013;4:122. 13. Gray R, Barnwell J, McConkey C, Hills RK, Williams NS, Kerr DJ. Adjuvant chemotherapy versus observation in patients with colorectal cancer: a randomised study. Lancet. 2007;370(9604):2020–2029. 14. Iveson TJ, Kerr RS, Saunders MP, et al. 3 versus 6 months of adjuvant oxaliplatin-fluoropyrimidine combination therapy for colorectal cancer (SCOT): an international, randomised, phase 3, non-inferiority trial. Lancet Oncol. 2018;19(4):562–578. 15. El Turabi A, Abel GA, Roland M, Lyratzopoulos G. Variation in reported experience of involvement in cancer treatment decision making: evidence from the National Cancer Patient Experience Survey. Br J Cancer. 2013;109(3):780–787. 16. Person B, Ifargan R, Lachter J, Duek SD, Kluger Y, Assalia A. The impact of preoperative stoma site marking on the incidence of complications, quality of life, and patient’s independence. Dis Colon Rectum. 2012;55(7):783–787. 17. Goldwag J, Marsicovetere P, Scalia P, et al. The impact of decision aids in patients with colorectal cancer: a systematic review. BMJ Open. 2019;9(9):e028379. 18. Mitchell AJ, Ferguson DW, Gill J, Paul J, Symonds P. Depression and anxiety in long-term cancer survivors compared with spouses 19. 20. 21. 22. 23. 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. and healthy controls: a systematic review and meta- analysis. Lancet Oncol. 2013;14(8):721–732. Abbey G, Thompson SB, Hickish T, Heathcote D. A meta- analysis of prevalence rates and moderating factors for cancer-related post-traumatic stress disorder. Psychooncology. 2015;24(4):371–381. Dunn J, Ng SK, Holland J, et al. Trajectories of psychological distress after colorectal cancer. Psychooncology. 2013;22(8):1759–1765. Walker J, Hansen CH, Martin P, et al. Prevalence, associations, and adequacy of treatment of major depression in patients with cancer: a cross-sectional analysis of routinely collected clinical data. Lancet Psychiatry. 2014;1(5):343–350. Graca PM, Figueiredo AP, Fincham FD. Anxiety, depression, traumatic stress and quality of life in colorectal cancer after different treatments: a study with Portuguese patients and their partners. Eur J Oncol Nurs. 2012;16(3):227–232. Dunn J, Ng SK, Breitbart W, et al. Health-related quality of life and life satisfaction in colorectal cancer survivors: trajectories of adjustment. Health Qual Life Outcomes. 2013;11:46. O’Gorman C, Stack J, O’Ceilleachair A, et al. Colorectal cancer survivors: an investigation of symptom burden and influencing factors. BMC Cancer. 2018;18(1):1022. Storey DJ, Waters RA, Hibberd CJ, et al. Clinically relevant fatigue in cancer outpatients: the Edinburgh Cancer Centre symptom study. Ann Oncol. 2007;18(11):1861–1869. Jansen L, Koch L, Brenner H, Arndt V. Quality of life among long- term (>/=5 years) colorectal cancer survivors—systematic review. Eur J Cancer. 2010;46(16):2879–2888. Bulkley JE, McMullen CK, Grant M, Wendel C, Hornbrook MC, Krouse RS. Ongoing ostomy self- care challenges of long- term rectal cancer survivors. Support Care Cancer. 2018;26(11):3933–3939. Kristensen HO, Thyo A, Christensen P. Systematic review of the impact of demographic and socioeconomic factors on quality of life in ostomized colorectal cancer survivors. Acta Oncol. 2019;58(5):566–572. Di Fabio F, Koller M, Nascimbeni R, Talarico C, Salerni B. Long-term outcome after colorectal cancer resection. Patients’ self-reported quality of life, sexual dysfunction and surgeons’ awareness of patients’ needs. Tumori. 2008;94(1):30–35. Persson E, Severinsson E, Hellstrom AL. Spouses’ perceptions of and reactions to living with a partner who has undergone surgery for rectal cancer resulting in a stoma. Cancer Nurs. 2004;27(1):85–90. Custers JA, Gielissen MF, Janssen SH, de Wilt JH, Prins JB. Fear of cancer recurrence in colorectal cancer survivors. Support Care Cancer. 2016;24(2):555–562. de Boer AG, Frings-Dresen MH. Employment and the common cancers: return to work of cancer survivors. Occup Med (Lond). 2009;59(6):378–380. Carey M, Lambert S, Smits R, Paul C, Sanson-Fisher R, Clinton- McHarg T. The unfulfilled promise: a systematic review of interventions to reduce the unmet supportive care needs of cancer patients. Support Care Cancer. 2012;20(2):207–219. 181 25 Prostate Cancer and Genitourinary Malignancies Andrew J. Roth and Alejandro Gonzalez-Restrepo Introduction Prostate and genitourinary (GU) cancers are common.1 With the exception of testicular cancer, the incidence of GU cancers (e.g., prostate, bladder, renal, and penile cancers) increases with advancing age. Thus, understanding coincident life phase characteristics is important in optimizing the ability of each patient to cope with his or her cancer. The effect of treatment on the quality of life (QOL) of patients has become more significant as survival has improved for many of these cancers. QOL areas of concern include coping with body image and integrity changes, varying degrees of sexual and physical intimacy dysfunction, and infertility. These issues compound the generic difficulties of coping with cancer, such as dealing with pain, fatigue, and other complications of treatment, including challenges of daily functioning, lifestyle changes, and career uncertainty. Primary treatment decisions are complicated by comparing the curability and longevity potential of different options with posttreatment QOL concerns, especially as newer treatments arise that do not yet have significant track records. Prostate Cancer Prostate cancer is the most common nonskin cancer in males in the United States, with almost 200,000